1
|
Arunsiripate TT, Groeltz-Thrush J, Saeng-Chuto K, Guo B, Michael A, Siepker C, Derscheid RJ, Rahe MC, Zhang J, Burrough E, Piñeyro PE. Diagnostic investigation of porcine hemagglutinating encephalomyelitis virus as potential pathogen associated with respiratory clinical signs and pulmonary lesions in pigs. Microb Pathog 2025; 203:107493. [PMID: 40120700 DOI: 10.1016/j.micpath.2025.107493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 02/27/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Porcine hemagglutinating encephalomyelitis virus (PHEV) is a member of the genus Betacoronavirus, known for its impact on the central and peripheral nervous systems in pigs. Traditionally associated with vomiting and wasting disease (VWD) and encephalomyelitis, PHEV was first reported in Canada in the late 1950s and has since been identified in numerous countries. Although serologic studies indicate global dissemination, the prevalence of PHEV remains unclear due to sporadic reporting and lack of active surveillance. Neonatal pigs are particularly vulnerable, with outbreaks resulting in high morbidity and mortality. Histopathological findings typically include non-suppurative encephalomyelitis and lymphoplasmacytic perivascular cuffs, gliosis, and neuronal degeneration. Recent observations have suggested a potential role for PHEV in respiratory disease, a hypothesis prompted by cases of influenza-like symptoms in pigs in Michigan in 2015 and corroborated by subsequent reports. This study aims to explore this possibility through a combination of clinical outbreak analysis and retrospective investigation. PHEV was confirmed via qPCR in 83.33 % of pigs examined for respiratory disease, with histological lesions such as necrotizing bronchitis and bronchiolitis. In-situ hybridization (ISH) confirmed the presence of PHEV mRNA in respiratory epithelium, and immunohistochemical analysis revealed significant macrophage infiltration in affected lung. Phylogenetic analysis indicated that PHEV strains from respiratory cases cluster closely with historical respiratory strains, though distinct from neurologic strains. This genetic differentiation suggests possible phenotypic variation contributing to respiratory tropism. The retrospective study identified PHEV in 7.62 % of cases with necrotizing bronchitis or bronchiolitis, reinforcing the virus's potential role in respiratory disease. Notably, PHEV co-infection with other respiratory pathogens such as PRRSV was observed, suggesting it may contribute to the porcine respiratory disease complex (PRDC). These findings suggest that PHEV is a significant respiratory pathogen in swine, warranting its inclusion in the differential diagnosis for respiratory disease in nursery pigs. Future research should focus on elucidating the pathogenesis of PHEV in respiratory disease, host-virus interactions, and the virus's impact on immune response and secondary infections. Understanding these factors will be crucial in developing effective preventive and therapeutic strategies against PHEV in swine.
Collapse
Affiliation(s)
- Trevor T Arunsiripate
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Jennifer Groeltz-Thrush
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Kepalee Saeng-Chuto
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Baoqing Guo
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Alyona Michael
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA; Department of Pathobiology, College of Veterinary Medicine, Auburn University, AL, USA
| | - Christopher Siepker
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Rachel J Derscheid
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Michael C Rahe
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA; Department of Population Health and Pathobiology, North Caroline State University, NC, USA
| | - Jianqiang Zhang
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Eric Burrough
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Pablo E Piñeyro
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA.
| |
Collapse
|
2
|
Nelsen A, Knudsen D, Hause BM. Identification of a Novel Astrovirus Associated with Bovine Respiratory Disease. Transbound Emerg Dis 2023; 2023:8512021. [PMID: 40303779 PMCID: PMC12017199 DOI: 10.1155/2023/8512021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 03/20/2023] [Accepted: 03/24/2023] [Indexed: 05/02/2025]
Abstract
Astroviruses (AstVs) cause gastrointestinal disease in mammals and avians. Emerging evidence suggests that some AstVs have extraintestinal tissue tropism, with AstVs detected in the liver, kidney, central nervous system, and the respiratory tract variably associated with disease. In cattle, AstV infection has been linked to gastroenteric or neurologic disease. Here, metagenomic sequencing of a lung from a bovine with respiratory disease identified a novel AstV with a predicted capsid-encoding ORF2 amino acid sequence with 66% identity to caprine astrovirus (CAstV G2.1). A quantitative reverse transcription PCR (qRT-PCR) targeting ORF2 found four out of 49 (8%) lungs and one out of 48 (2%) enteric samples obtained from bovine diagnostic submissions positive for the novel bovine astrovirus (BAstV). In two strongly qRT-PCR-positive lung samples, intense novel BAstV nucleic acid signals were mainly localized in the cytoplasm of alveolar macrophages and mononuclear cells using RNAscope® in situ hybridization (ISH). Genetic analysis of two novel BAstV genomes determined from qRT-PCR positive samples found high similarity for ORF1ab nucleotide sequence (92.1% and 93.9%) to BAstV strain BSRI-1, while ORF2 nucleotide sequence was most similar to CAstV G2.1 (74.6% and 77.6%). Phylogenetic analysis of the novel BAstV sequences found a close genetic relationship to the single BAstV (BSRI-1) previously identified from a bovine respiratory sample as well as bovine and caprine AstVs identified from various tissues. Further research is needed to determine the clinical significance of BAstV in respiratory diseases.
Collapse
Affiliation(s)
- April Nelsen
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, South Dakota, USA
| | - David Knudsen
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, South Dakota, USA
- Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, South Dakota, USA
| | - Ben M. Hause
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, South Dakota, USA
- Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, South Dakota, USA
| |
Collapse
|
3
|
Zuckerman NS, Shulman LM. Next-Generation Sequencing in the Study of Infectious Diseases. Infect Dis (Lond) 2023. [DOI: 10.1007/978-1-0716-2463-0_1090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
4
|
Nieuwenhuijse DF, van der Linden A, Kohl RHG, Sikkema RS, Koopmans MPG, Oude Munnink BB. Towards reliable whole genome sequencing for outbreak preparedness and response. BMC Genomics 2022; 23:569. [PMID: 35945497 PMCID: PMC9361258 DOI: 10.1186/s12864-022-08749-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 07/08/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To understand the dynamics of infectious diseases, genomic epidemiology is increasingly advocated, with a need for rapid generation of genetic sequences during outbreaks for public health decision making. Here, we explore the use of metagenomic sequencing compared to specific amplicon- and capture-based sequencing, both on the Nanopore and the Illumina platform for generation of whole genomes of Usutu virus, Zika virus, West Nile virus, and Yellow Fever virus. RESULTS We show that amplicon-based Nanopore sequencing can be used to rapidly obtain whole genome sequences in samples with a viral load up to Ct 33 and capture-based Illumina is the most sensitive method for initial virus determination. CONCLUSIONS The choice of sequencing approach and platform is important for laboratories wishing to start whole genome sequencing. Depending on the purpose of genome sequencing the best choice can differ. The insights presented in this work and the shown differences in data characteristics can guide labs to make a well informed choice.
Collapse
Affiliation(s)
| | | | - Robert H G Kohl
- Departement of Virology of the Vaccination Programme, RIVM, Bilthoven, the Netherlands
| | - Reina S Sikkema
- Viroscience Department, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | - Bas B Oude Munnink
- Viroscience Department, Erasmus Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
5
|
Bovine Respiratory Syncytial Virus Genome Sequences from Cattle with Clinical Respiratory Disease in Kansas, 2021. Microbiol Resour Announc 2022; 11:e0031322. [PMID: 35412352 PMCID: PMC9119100 DOI: 10.1128/mra.00313-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We report two near-complete bovine respiratory syncytial virus genome sequences collected from 10-month-old cattle with respiratory disease in Kansas in December 2021. No other respiratory pathogens were confirmed in the samples. These genome sequences update the currently circulating BRSV field strains in the United States.
Collapse
|
6
|
Doerksen T, Christensen T, Lu A, Noll L, Bai J, Henningson J, Palinski R. Assessment of Porcine Rotavirus-associated virome variations in pigs with enteric disease. Vet Microbiol 2022; 270:109447. [PMID: 35561657 DOI: 10.1016/j.vetmic.2022.109447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/14/2022] [Accepted: 04/23/2022] [Indexed: 11/29/2022]
|
7
|
Novel hepatoviruses in synanthropic bats in the upper Midwestern United States. Arch Virol 2022; 167:2749-2751. [PMID: 36136250 PMCID: PMC9510175 DOI: 10.1007/s00705-022-05610-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/14/2022] [Indexed: 12/14/2022]
Abstract
A nearly complete genome sequence of hepatovirus G was isolated from an Eptesicus fuscus bat submitted for rabies virus testing due to human exposure in South Dakota. The predicted polyprotein sequence was 78.2% and 74.4% identical to genotypes G1 and G2, respectively, recovered from bats in Ghana. Quantitative PCR on 90 E. fuscus bats showed that eight (8.9%) were positive for hepatovirus G. Targeted sequencing of the VP2 region of the genome for five positive samples showed >99% identity to hepatovirus G strain Ef15893, demonstrating that hepatovirus G commonly circulates in E. fuscus bats in the upper Midwest.
Collapse
|
8
|
Nelsen A, Lin CM, Hause BM. Porcine Parvovirus 2 Is Predominantly Associated With Macrophages in Porcine Respiratory Disease Complex. Front Vet Sci 2021; 8:726884. [PMID: 34485445 PMCID: PMC8414833 DOI: 10.3389/fvets.2021.726884] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/05/2021] [Indexed: 11/27/2022] Open
Abstract
Porcine respiratory disease complex (PRDC) is a significant source of morbidity and mortality, manifested by pneumonia of multiple etiologies, where a variety of pathogens and environment and management practices play a role in the disease. Porcine reproductive and respiratory syndrome virus (PRRSV), influenza A virus (IAV), and porcine circovirus 2 (PCV2) are well-established pathogens in PRDC. Porcine parvovirus 2 (PPV2) has been identified in both healthy and clinically diseased pigs at a high prevalence worldwide. Despite widespread circulation, the significance of PPV2 infection in PRDC and its association with other co-infections are unclear. Here, PPV2 was detected in the lung tissue in 39 of 100 (39%) PRDC-affected pigs by quantitative polymerase chain reaction (qPCR). Using in situ hybridization (ISH) in conjunction with tissue microarrays (TMA), PPV2 infection was localized in alveolar macrophages and other cells in the lungs with interstitial pneumonia in 28 of 99 (28.2%) samples. Viral load tended to correlate with the number of macrophages in the lungs. Assessment of the frequency, viral titers, and tissue distributions showed no association between infection of PPV2 and other major viral respiratory pathogens. In one-third of the PPV2-positive samples by qPCR, no other known viruses were identified by metagenomic sequencing. The genome sequences of PPV2 were 99.7% identical to the reference genomes. Although intensive intranuclear and intracytoplasmic signals of PPV2 were mainly detected in alveolar macrophages by ISH, no obvious virus replication was noted in in vitro cell culture. Together, these results suggest that PPV2 is associated, but may not be the sole causative agent, with PRDC, warranting the control and prevention of this underdiagnosed virus.
Collapse
Affiliation(s)
- April Nelsen
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, United States
| | - Chun-Ming Lin
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, United States.,Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD, United States
| | - Ben M Hause
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, United States.,Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD, United States
| |
Collapse
|
9
|
Near-Complete Sequence of a Highly Divergent Reovirus Genome Recovered from Callinectes sapidus. Microbiol Resour Announc 2021; 10:10/1/e01278-20. [PMID: 33414308 PMCID: PMC8407731 DOI: 10.1128/mra.01278-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
This report describes the sequence of a reovirus genome, discovered in Callinectes sapidus in Brazil. The genome sequence of Callinectes sapidus reovirus 2 (CsRV2) consists of 12 segments that encode 13 putative proteins. The predicted RNA-dependent RNA polymerase is highly similar to that of Eriocheir sinensis reovirus 905, suggesting that CsRV2 also belongs to the genus Cardoreovirus.
Collapse
|
10
|
Persistent infection of American bison (Bison bison) with bovine viral diarrhea virus and bosavirus. Vet Microbiol 2020; 252:108949. [PMID: 33338948 DOI: 10.1016/j.vetmic.2020.108949] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/29/2020] [Indexed: 01/01/2023]
Abstract
Bovine viral diarrhea viruses (BVDV) are significant pathogens of cattle, leading to losses associated with reproductive failure, respiratory disease and immune dysregulation. While cattle are the reservoir for BVDV, a wide range of domestic and wild ruminants are susceptible to infection and disease caused by BVDV. Samples from four American bison (Bison bison) from a captive herd were submitted for diagnostic testing due to their general unthriftiness. Metagenomic sequencing on pooled nasal swabs and serum identified co-infection with a BVDV and a bovine bosavirus. The BVDV genome was more similar to the vaccine strain Oregon C24 V than to other BVDV sequences in GenBank, with 92.7 % nucleotide identity in the open reading frame. The conserved 5'-untranslated region was 96.3 % identical to Oregon C24 V. Bosavirus has been previously identified in pooled fetal bovine serum but its clinical significance is unknown. Sequencing results were confirmed by virus isolation and PCR detection of both viruses in serum and nasal swab samples from two of the four bison. One animal was co-infected with both BVDV and bosavirus while separate individuals were positive solely for BVDV or bosavirus. Serum and nasal swabs from these same animals collected 51 days later remained positive for BVDV and bosavirus. These results suggest that both viruses can persistently infect bison. While the etiological significance of bosavirus infection is unknown, the ability of BVDV to persistently infect bison has implications for BVDV control and eradication programs. Possible synergy between BVDV and bosavirus persistent infection warrants further study.
Collapse
|
11
|
Kaplan BS, Falkenberg S, Dassanayake R, Neill J, Velayudhan B, Li F, Vincent AL. Virus strain influenced the interspecies transmission of influenza D virus between calves and pigs. Transbound Emerg Dis 2020; 68:3396-3404. [PMID: 33259672 DOI: 10.1111/tbed.13943] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 11/19/2020] [Accepted: 11/26/2020] [Indexed: 02/01/2023]
Abstract
Influenza D viruses (IDV) belong to a new genus in the family Orthomyxoviridae. IDV is the aetiologic agent of acute, mild respiratory disease in ungulate species with agricultural importance (cattle, pigs, sheep, goats, camels, etc.). Despite the initial isolate being of porcine origin, serological data suggest cattle to be the primary host of IDV. The study aims were twofold: elucidating species-specific replication kinetics of IDV in bovine and porcine hosts and defining the interspecies potential with two different IDV strains. Three calves and three pigs were intranasally inoculated with the prototypic strain D/swine/Oklahoma/1334/2017 or a genetically distinct cattle isolate, D/bovine/Texas/72/2017. Two days following infection, three naïve pigs and three naïve calves were co-housed with inoculated calves and pigs, respectively. The species of IDV origin had no effect on virus replication kinetics in the upper respiratory tract of inoculated calves and pigs; similar shedding profiles were observed for each species and virus. However, interspecies transmission was found to be associated with virus origin species; D/bovine/Texas/72/2017 and D/swine/Oklahoma/1334/2017 were directly transmitted only to contact calves or pigs, respectively. Even so, transmission efficiency was higher for calves compared to pigs. Together, these data show that cattle and pigs are permissive for IDV replication, but IDV transmission may be species dependent. Host-specific mutations likely influenced transmission efficiencies between agriculturally important mammalian species.
Collapse
Affiliation(s)
- Bryan S Kaplan
- Virus and Prion Research Unit, National Animal Disease Center, USDA-ARS, Ames, IA, USA
| | - Shollie Falkenberg
- Ruminant Diseases and Immunology Unit, National Animal Disease Center, USDA-ARS, Ames, IA, USA
| | - Rohana Dassanayake
- Ruminant Diseases and Immunology Unit, National Animal Disease Center, USDA-ARS, Ames, IA, USA
| | - John Neill
- Ruminant Diseases and Immunology Unit, National Animal Disease Center, USDA-ARS, Ames, IA, USA
| | - Binu Velayudhan
- North Carolina Veterinary Diagnostic Laboratory System, Raleigh, NC, USA
| | - Feng Li
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, USA
| | - Amy L Vincent
- Virus and Prion Research Unit, National Animal Disease Center, USDA-ARS, Ames, IA, USA
| |
Collapse
|
12
|
Multivariate analysis as a method to evaluate antigenic relationships between BVDV vaccine and field strains. Vaccine 2020; 38:5764-5772. [PMID: 32690424 DOI: 10.1016/j.vaccine.2020.07.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/29/2020] [Accepted: 07/06/2020] [Indexed: 02/08/2023]
Abstract
Bovine viral diarrhea virus (BVDV) is comprised of two species, BVDV-1 and BVDV-2, but given the genetic diversity among pestiviruses, at least 21 subgenotypes are described for BVDV-1 and 4 for BVDV-2. Genetic characterization can be achieved through complete or partial sequencing and phylogeny, but antigenic characterization can be difficult to determine due to the antigenic diversity and cross-neutralization that exists among isolates. The traditional method for evaluating antigenic relationships between pestivirus isolates is the virus neutralization (VN) assay, but interpretation of the data to determine antigenic difference can be unclear. Data from this study utilized a multivariate analysis for visualization of VN results to analyze the antigenic relationships between vaccine strains and multiple field isolates. Polyclonal sera were generated against 6 BVDV strains currently contained in vaccine formulations, and each serum was used in VN's to measure the neutralizing antibody titers against 15 BVDV field isolates characterized as prevalent and divergent subgenotypes in the USA. Principal component analysis (PCA) were performed on the VN assay datasets, and results were interpreted from PCA clustering within the PCA dendrogram and scatter plot. The results demonstrated clustering patterns among isolates suggestive of antigenic differences. While expected, the BVDV-1 and BVDV-2 isolates did not cluster together and had the greatest spatial distribution. In addition, other BVDV isolates had distinct spatial patterns suggesting antigenically divergent isolates. This analysis provides an alternative and more efficient means to analyze large VN datasets to visualize antigenic relationships between pestivirus isolates. This analysis could be beneficial for vaccine development and evaluation of efficacy, since most vaccines cannot fully protect animals from the broad range diversity of BVDV viruses.
Collapse
|
13
|
Shivanna V, Cino-Ozuna AG, Heskett C, Marthaler DG, Ganta C. Pseudocowpox virus infection in an American bison (Bison bison). BMC Vet Res 2020; 16:241. [PMID: 32660468 PMCID: PMC7359452 DOI: 10.1186/s12917-020-02464-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 07/07/2020] [Indexed: 12/04/2022] Open
Abstract
Background The present report describes a case of pseudocowpox virus (PCPV) infection in a seven-year-old female bison euthanized due to a history of declining condition and sores on the vulva and udder. Case presentation External examination revealed multifocal, raised, keratinized plaques (0.5–2 cm) covering the skin of the ventral surface of the tail, perineum, caudoventral abdomen, udder, both inguinal recesses, and the medial aspects of both thighs. No significant gross lesions were present in the reminder of the tissues examined. Histopathological examination of the affected skin showed moderate epidermal hyperplasia with rete pegs, marked parakeratotic hyperkeratosis with crusts of degenerate neutrophils and cell debris, and few epithelial cells undergoing ballooning degeneration with occasional eosinophilic intracytoplasmic inclusion bodies (3–5 μm Bollinger body). Negative staining electron microscopy from skin revealed typical Parapoxvirus (PPV) particles, which were also confirmed by real-time PCR (Ct =18.6). Metagenomic analysis of the skin samples revealed only poxviruses. The bison parapox B2L envelope gene clustered with other parapox sequences identified from ruminants. Conclusions This is the first report of PCPV virus infection in an American bison. Identification of novel susceptible hosts of parapox viruses sheds light on the viral evolution and highlights the importance of potential economic impact of this disease to the bison industry.
Collapse
Affiliation(s)
- Vinay Shivanna
- Kansas State University Veterinary Diagnostic Laboratory, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, 66506, USA
| | - A Giselle Cino-Ozuna
- Kansas State University Veterinary Diagnostic Laboratory, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, 66506, USA
| | - Cody Heskett
- Kansas State University Veterinary Diagnostic Laboratory, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, 66506, USA
| | - Douglas G Marthaler
- Kansas State University Veterinary Diagnostic Laboratory, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, 66506, USA
| | - Charan Ganta
- Kansas State University Veterinary Diagnostic Laboratory, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, 66506, USA.
| |
Collapse
|
14
|
Walz PH, Chamorro MF, M Falkenberg S, Passler T, van der Meer F, R Woolums A. Bovine viral diarrhea virus: An updated American College of Veterinary Internal Medicine consensus statement with focus on virus biology, hosts, immunosuppression, and vaccination. J Vet Intern Med 2020; 34:1690-1706. [PMID: 32633084 PMCID: PMC7517858 DOI: 10.1111/jvim.15816] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 12/01/2022] Open
Abstract
Control of bovine viral diarrhea virus (BVDV) in cattle populations across most of the world has remained elusive in spite of advances in knowledge about this viral pathogen. A central feature of virus perseverance in cattle herds is the unique mechanism of persistent infection. Managing BVDV infection in herds involves controlling persistently infected carrier animals using a multidimensional approach of vaccination, biosecurity, and identification of BVDV reservoirs. A decade has passed since the original American College of Veterinary Internal Medicine consensus statement on BVDV. While much has remained the same with respect to clinical signs of disease, pathogenesis of infection including persistent infection, and diagnosis, scientific articles published since 2010 have led to a greater understanding of difficulties associated with control of BVDV. This consensus statement update on BVDV presents greater focus on topics currently relevant to the biology and control of this viral pathogen of cattle, including changes in virus subpopulations, infection in heterologous hosts, immunosuppression, and vaccination.
Collapse
Affiliation(s)
- Paul H Walz
- College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Manuel F Chamorro
- College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Shollie M Falkenberg
- USDA Agricultural Research Service, National Animal Disease Center, Ames, Iowa, USA
| | - Thomas Passler
- College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Frank van der Meer
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Amelia R Woolums
- College of Veterinary Medicine, Mississippi State University, Starkville, Mississippi, USA
| |
Collapse
|
15
|
Marine RL, Magaña LC, Castro CJ, Zhao K, Montmayeur AM, Schmidt A, Diez-Valcarce M, Ng TFF, Vinjé J, Burns CC, Nix WA, Rota PA, Oberste MS. Comparison of Illumina MiSeq and the Ion Torrent PGM and S5 platforms for whole-genome sequencing of picornaviruses and caliciviruses. J Virol Methods 2020; 280:113865. [PMID: 32302601 PMCID: PMC9119587 DOI: 10.1016/j.jviromet.2020.113865] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 02/04/2020] [Accepted: 04/06/2020] [Indexed: 02/06/2023]
Abstract
Next-generation sequencing is a powerful tool for virological surveillance. While Illumina® and Ion Torrent® sequencing platforms are used extensively for generating viral RNA genome sequences, there is limited data comparing different platforms. The Illumina MiSeq, Ion Torrent PGM and Ion Torrent S5 platforms were evaluated using a panel of sixteen specimens containing picornaviruses and human caliciviruses (noroviruses and sapoviruses). The specimens were processed, using combinations of three library preparation and five sequencing kits, to assess the quality and completeness of assembled viral genomes, and an estimation of cost per sample to generate the data was calculated. The choice of library preparation kit and sequencing platform was found to impact the breadth of genome coverage and accuracy of consensus viral genomes. The Ion Torrent S5 510 chip runs produced more reads at a lower cost per sample than the highest output Ion Torrent PGM 318 chip run, and generated the highest proportion of reads for enterovirus D68 samples. However, indels at homopolymer regions impacted the accuracy of consensus genome sequences. For lower throughput sequencing runs (i.e., Ion Torrent 510 and Illumina MiSeq Nano V2), the cost per sample was lower on the MiSeq platform, whereas with higher throughput runs (Ion Torrent 530 and Illumina MiSeq V2) there is less of a difference in the cost per sample between the two sequencing platforms ($5.47-$10.25 more per sample for an Ion Torrent 530 chip run when multiplexing 24 samples). These findings suggest that the Ion Torrent S5 and Illumina MiSeq platforms are both viable options for genomic sequencing of RNA viruses, each with specific advantages and tradeoffs.
Collapse
Affiliation(s)
- Rachel L Marine
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Laura C Magaña
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA; Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, USA
| | - Christina J Castro
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA; Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, USA
| | - Kun Zhao
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | | - Marta Diez-Valcarce
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA; Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, USA
| | - Terry Fei Fan Ng
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jan Vinjé
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Cara C Burns
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - W Allan Nix
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Paul A Rota
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - M Steven Oberste
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
16
|
Russell GC, Zadoks RN, Willoughby K, Bachofen C. Bovine viral diarrhoea virus loses quasispecies diversity rapidly in culture. Microb Genom 2020; 6:e000343. [PMID: 32160141 PMCID: PMC7276709 DOI: 10.1099/mgen.0.000343] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/07/2020] [Indexed: 12/25/2022] Open
Abstract
Bovine viral diarrhoea (BVD) is an important disease of cattle, with significant impacts on animal health and welfare. The wide host range of the causative pestiviruses may lead to formation of virus reservoirs in other ruminant or wildlife species, presenting a concern for the long-term success of BVD eradication campaigns. It is likely that the quasispecies nature of these RNA viruses contributes to their interspecies transmission by providing genetic plasticity. Understanding the spectrum of sequence variants present in persistently infected (PI) animals is, therefore, essential for studies of virus transmission. To analyse quasispecies diversity without amplification bias, we extracted viral RNA from the serum of a PI cow, and from cell culture fluid after three passages of the same virus in culture, to produce cDNA without amplification. Sequencing of this material using Illumina 250 bp paired-read technology produced full-length virus consensus sequences from both sources and demonstrated the quasispecies diversity of this pestivirus A genotype 1a field strain within serum and after culture. We report the distribution and diversity of over 800 SNPs and provide evidence for a loss of diversity after only three passages in cell culture, implying that cultured viruses cannot be used to understand quasispecies diversity and may not provide reliable molecular markers for source tracing or transmission studies. Additionally, both serum and cultured viruses could be sequenced as a set of 25 overlapping PCR amplicons that demonstrated the same consensus sequences and the presence of many of the same quasispecies variants. The observation that aspects of the quasispecies structure revealed by massively parallel sequencing are also detected after PCR and Sanger sequencing suggests that this approach may be useful for small or difficult to analyse samples.
Collapse
Affiliation(s)
- George C. Russell
- Moredun Research Institute, Pentlands Science Park, Midlothian EH26 0PZ, UK
| | - Ruth N. Zadoks
- Moredun Research Institute, Pentlands Science Park, Midlothian EH26 0PZ, UK
- Present address: Sydney School of Veterinary Science, University of Sydney, Camden, NSW, Australia
| | - Kim Willoughby
- Moredun Research Institute, Pentlands Science Park, Midlothian EH26 0PZ, UK
| | - Claudia Bachofen
- Moredun Research Institute, Pentlands Science Park, Midlothian EH26 0PZ, UK
- Present address: Institute of Virology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, CH-8057 Zürich, Switzerland
| |
Collapse
|
17
|
Gao X, Li Q, Fang L, Song H, Cui Z, Meng F, Zhang Z. The follicle promotes the evolution of variants of avian leukosis virus subgroup J in vertical transmission. Biochem Biophys Res Commun 2019; 521:1089-1094. [PMID: 31733830 DOI: 10.1016/j.bbrc.2019.11.042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 11/17/2022]
Abstract
Avian leukosis virus (ALV) is one of the main causative agent of tumor development, which brings enormous economic losses to the poultry industry worldwide. ALV can be transmitted horizontally and vertically, and the latter often give rise to more adverse pathogenicity. However, the propagation and evolution of ALV underlying vertical transmission remain not-well understood. Herein, an animal model for the evolution of variants of ALV subgroup J (ALV-J) in the vertical transmission was built and different organs from infected hens and plasma from their ALV-positive progenies were collected, and then three segments in the hypervariable regions of ALV (gp85-A, gp85-B, LTR-C) were amplified and sequenced using conventional Sanger sequencing and MiSeq high-throughput sequencing, respectively. The results showed that the genomic diversity of ALV-J occurred in different organs from ALV-J infected hen, and that the dominant variants in different organs of parental hens, especially in follicle, changed significantly compared with original inoculum strain. Notably, the dominant variants in progenies exhibited higher homologies with variants in parental hens' follicle (88.9%-98.9%) than other organs (85.6%-91.1%), and most consistent mutations in the variants were observed between the progenies and parental hen's follicle. Furthermore, HyPhy analysis indicated that the global selection pressure value (ω) in the follicle is significantly higher than those in other organs. In summary, an animal model for vertical transmission was built and our findings revealed the evolution of variants of ALV in the process of vertical transmission, moreover, the variants were most likely to be taken to the next generation via follicle, which may be related to the higher selection pressure follicle underwent.
Collapse
Affiliation(s)
- Xintao Gao
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100081, China; Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China
| | - Qiuchen Li
- College of Veterinary Medicine, China Agricultural University, Beijing, 100094, China
| | - Lichun Fang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China
| | - Haozhi Song
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100081, China
| | - Zhizhong Cui
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong, 271018, China
| | - Fanfeng Meng
- College of Veterinary Medicine, China Agricultural University, Beijing, 100094, China.
| | - Zhifang Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100081, China.
| |
Collapse
|
18
|
Arruda B, Piñeyro P, Derscheid R, Hause B, Byers E, Dion K, Long D, Sievers C, Tangen J, Williams T, Schwartz K. PCV3-associated disease in the United States swine herd. Emerg Microbes Infect 2019; 8:684-698. [PMID: 31096848 PMCID: PMC6534263 DOI: 10.1080/22221751.2019.1613176] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Porcine circovirus-associated disease encompasses multiple disease syndromes including porcine circovirus 2 systemic diseases, reproductive failure, and porcine dermatitis and nephropathy syndrome. Until recently, porcine circovirus 2 was the only species associated with the porcine circovirus-associated disease. In this report, diagnostic investigations of thirty-six field cases submitted from multiple production systems, numerous sites and varied geographic locations demonstrated porcine circovirus 3 within lesions by in situ hybridization including fetuses with myocarditis, weak-born neonatal piglets with encephalitis and myocarditis, from cases of porcine dermatitis and nephropathy syndrome, and in weaned pigs with systemic periarteritis. Porcine circovirus 3 was detected by PCR in numerous fetuses and perinatal piglets at high viral loads (trillions of genome copies per mL of tissue homogenate). Samples from all cases in this study were assayed and found negative for porcine circovirus 2 by PCR. Metagenomic sequencing was performed on a subset of reproductive cases, consisting of sixteen fetuses/fetal sample pools. PCV3 was identified in all pools and the only virus identified in fourteen pools. Based on these data, porcine circovirus 3 is considered a putative cause of reproductive failure, encephalitis and myocarditis in perinatal piglets, porcine dermatitis and nephropathy syndrome, and periarteritis in swine in the United States.
Collapse
Affiliation(s)
- Bailey Arruda
- a Department of Veterinary Diagnostic and Production Animal Medicine , Iowa State University , Ames , IA , USA
| | - Pablo Piñeyro
- a Department of Veterinary Diagnostic and Production Animal Medicine , Iowa State University , Ames , IA , USA
| | - Rachel Derscheid
- a Department of Veterinary Diagnostic and Production Animal Medicine , Iowa State University , Ames , IA , USA
| | - Ben Hause
- b Cambridge Technologies , Worthington , MN , USA
| | | | - Kate Dion
- d The Hanor Company of Wisconsin, LLC , Enid , OK , USA
| | | | | | - Jon Tangen
- d The Hanor Company of Wisconsin, LLC , Enid , OK , USA
| | | | - Kent Schwartz
- a Department of Veterinary Diagnostic and Production Animal Medicine , Iowa State University , Ames , IA , USA
| |
Collapse
|
19
|
Silveira S, Falkenberg SM, Dassanayake RP, Walz PH, Ridpath JF, Canal CW, Neill JD. In vitro method to evaluate virus competition between BVDV-1 and BVDV-2 strains using the PrimeFlow RNA assay. Virology 2019; 536:101-109. [PMID: 31415943 DOI: 10.1016/j.virol.2019.07.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/26/2019] [Accepted: 07/31/2019] [Indexed: 12/29/2022]
Abstract
Bovine viral diarrhea viruses (BVDV), segregated in BVDV-1 and BVDV-2 species, lead to substantial economic losses to the cattle industry worldwide. It has been hypothesized that there could be differences in level of replication, pathogenesis and tissue tropism between BVDV-1 and BVDV-2 strains. Thus, this study developed an in vitro method to evaluate virus competition between BVDV-1 and BVDV-2 strains. To this end the competitive dynamics of BVDV-1a, BVDV-1b, and BVDV-2a strains in cell cultures was evaluated by a PrimeFlow RNA assay. Similar results were observed in this study, as was observed in an earlier in vivo transmission study. Competitive exclusion was observed as the BVDV-2a strains dominated and excluded the BVDV-1a and BVDV-1b strains. The in vitro model developed can be used to identify viral variations that result in differences in frequency of subgenotypes detected in the field, vaccine failure, pathogenesis, and strain dependent variation in immune responses.
Collapse
Affiliation(s)
- S Silveira
- Laboratório de Virologia, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - S M Falkenberg
- Ruminant Diseases and Immunology Unit, National Animal Disease Center/ARS/USDA, Ames, IA, USA.
| | - R P Dassanayake
- Ruminant Diseases and Immunology Unit, National Animal Disease Center/ARS/USDA, Ames, IA, USA
| | - P H Walz
- Department of Pathobiology, College of Veterinary Medicine, 129 Sugg Laboratory, Auburn University, AL, 36849, USA
| | - J F Ridpath
- Ruminant Diseases and Immunology Unit, National Animal Disease Center/ARS/USDA, Ames, IA, USA
| | - C W Canal
- Laboratório de Virologia, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - J D Neill
- Ruminant Diseases and Immunology Unit, National Animal Disease Center/ARS/USDA, Ames, IA, USA
| |
Collapse
|
20
|
Kumar D, Chaudhary S, Lu N, Duff M, Heffel M, McKinney CA, Bedenice D, Marthaler D. Metagenomic Next-Generation Sequencing Reveal Presence of a Novel Ungulate Bocaparvovirus in Alpacas. Viruses 2019; 11:E701. [PMID: 31370351 PMCID: PMC6724020 DOI: 10.3390/v11080701] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/26/2019] [Accepted: 07/27/2019] [Indexed: 12/15/2022] Open
Abstract
Viruses belonging to the genus Bocaparvovirus(BoV) are a genetically diverse group of DNA viruses known to cause respiratory, enteric, and neurological diseases in animals, including humans. An intestinal sample from an alpaca (Vicugnapacos) herd with reoccurring diarrhea and respiratory disease was submitted for next-generation sequencing, revealing the presence of a BoV strain. The alpaca BoV strain (AlBoV) had a 58.58% whole genome nucleotide percent identity to a camel BoV from Dubai, belonging to a tentative ungulate BoV 8 species (UBoV8). Recombination events were lacking with other UBoV strains. The AlBoV genome was comprised of the NS1, NP1, and VP1 proteins. The NS1 protein had the highest amino acid percent identity range (57.89-67.85%) to the members of UBoV8, which was below the 85% cut-off set by the International Committee on Taxonomy of Viruses. The low NS1 amino acid identity suggests that AlBoV is a tentative new species. The whole genome, NS1, NP1, and VP1 phylogenetic trees illustrated distinct branching of AlBoV, sharing a common ancestor with UBoV8. Walker loop and Phospholipase A2 (PLA2) motifs that are vital for virus infectivity were identified in NS1 and VP1 proteins, respectively. Our study reports a novel BoV strain in an alpaca intestinal sample and highlights the need for additional BoV research.
Collapse
Affiliation(s)
- Deepak Kumar
- Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Suman Chaudhary
- Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Nanyan Lu
- Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Michael Duff
- Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Mathew Heffel
- Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Caroline A McKinney
- Department of Clinical Sciences, Cummings School of Veterinary Medicine at Tufts University, 200 Westboro Road, North Grafton, MA 01536, USA
| | - Daniela Bedenice
- Department of Clinical Sciences, Cummings School of Veterinary Medicine at Tufts University, 200 Westboro Road, North Grafton, MA 01536, USA
| | - Douglas Marthaler
- Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
21
|
Neill JD, Crossley BM, Mosena AC, Ridpath JF, Bayles DO, Hietala SK, Killian ML, Falkenberg SM. Genomic and antigenic characterization of a cytopathic bovine viral diarrhea virus 1i isolated in the United States. Virology 2019; 535:279-282. [PMID: 31357167 DOI: 10.1016/j.virol.2019.07.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/23/2019] [Accepted: 07/23/2019] [Indexed: 11/18/2022]
Abstract
Bovine viral diarrhea viruses (BVDV) are a common global viral pathogen of ruminants. Considerable genetic variability is found amongst BVDV1 isolates, with at least 21 subgenotypes being described. In the United States, BVDV1a and 1b are the only subgenotypes described to date. Here, the genomic sequence of CA2005, a cytopathic BVDV1, was determined. This virus, isolated in California, did not segregate into either BVDV1a or 1b subgenotypes. BLAST analysis showed CA2005 was most closely related to BVDV1i isolates. CA2005 was also the first cytopathic BVDV1i and one of few non-1a, non-1b cytopathic viruses reported. The genomic sequence was 15,752 nucleotides in length. Cytopathogenicity was conferred by duplication of the NS3 protein with a small ubiquitin B insertion at the border of the NS2/NS3 proteins. Virus neutralization assays using antisera against BVDV1a vaccine viruses revealed variable neutralization, suggesting modified live vaccines may not be totally protective against CA2005 and similar viruses.
Collapse
Affiliation(s)
- John D Neill
- USDA, Agricultural Research Service, National Animal Disease Center, Ruminant Diseases and Immunology Research Unit, 1920 Dayton Ave., Ames, IA, 50010, USA.
| | - Beate M Crossley
- California Animal Health and Food Safety Laboratory System, Davis Branch, University of California, Davis, CA, 95616, USA
| | - Ana C Mosena
- Laboratorio de Virologia, Faculdade de Veterinaria, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, Rio Grande Do Sul, Brazil
| | - Julia F Ridpath
- USDA, Agricultural Research Service, National Animal Disease Center, Ruminant Diseases and Immunology Research Unit, 1920 Dayton Ave., Ames, IA, 50010, USA
| | - Darrell O Bayles
- USDA, Agricultural Research Service, National Animal Disease Center, Bacterial Diseases Research Unit, 1920 Dayton Ave., Ames, IA, 50010, USA
| | - Sharon K Hietala
- California Animal Health and Food Safety Laboratory System, Davis Branch, University of California, Davis, CA, 95616, USA
| | - Mary Lea Killian
- USDA, Animal and Plant Inspection Service, National Veterinary Services Laboratory, 1920 Dayton Ave., Ames, IA, 50010, USA
| | - Shollie M Falkenberg
- USDA, Agricultural Research Service, National Animal Disease Center, Ruminant Diseases and Immunology Research Unit, 1920 Dayton Ave., Ames, IA, 50010, USA
| |
Collapse
|
22
|
Falkenberg SM, Dassanayake RP, Walz P, Casas E, Neill JD, Ridpath JF. Frequency of bovine viral diarrhea virus detected in subpopulations of peripheral blood mononuclear cells in persistently infected animals and health outcome. Vet Immunol Immunopathol 2019; 207:46-52. [DOI: 10.1016/j.vetimm.2018.11.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/15/2018] [Accepted: 11/23/2018] [Indexed: 10/27/2022]
|
23
|
Neill JD, Workman AM, Hesse R, Bai J, Porter EP, Meadors B, Anderson J, Bayles DO, Falkenberg SM. Identification of BVDV2b and 2c subgenotypes in the United States: Genetic and antigenic characterization. Virology 2018; 528:19-29. [PMID: 30553108 DOI: 10.1016/j.virol.2018.12.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/03/2018] [Accepted: 12/03/2018] [Indexed: 01/22/2023]
Abstract
Bovine viral diarrhea virus (BVDV), a ubiquitous pathogen of cattle, causes subclinical to severe acute disease. Two species of BVDV are recognized, BVDV1 and BVDV2 with BVDV1 divided into at least 21 subgenotypes and BVDV2 into 3-4 subgenotypes, most commonly using sequences from the 5' untranslated region (5' UTR). We report genomic sequencing of 8 BVDV2 isolates that did not segregate into the 2a subgenotype; but represented two additional BVDV2 subgenotypes. One BVDV2 subgenotype was previously recognized only in Asia. The other seven viruses fell into a second subgenotype that was first reported in Brazil and the U.S. in 2002. Neutralization assays using antiserum raised against vaccine strain BVDV2a 296c revealed varying degrees of neutralization of genetically diverse BVDV2 isolates. Neutralization titers decreased from 1.8 to more than a four log(2) decrease. This study illustrated the considerable genetic and antigenic diversity in BVDV2 circulating in the U.S.
Collapse
Affiliation(s)
- John D Neill
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, USDA, ARS, Ames, IA 50010, USA.
| | - Aspen M Workman
- US Meat Animal Research Center, USDA, ARS, Clay Center, NE 68933, USA
| | - Richard Hesse
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS, 66502 USA
| | - Jianfa Bai
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS, 66502 USA
| | - Elizabeth Poulsen Porter
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS, 66502 USA
| | - Barbara Meadors
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS, 66502 USA
| | - Joe Anderson
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS, 66502 USA
| | - Darrell O Bayles
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, USDA, ARS, Ames, IA 50010, USA
| | - Shollie M Falkenberg
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, USDA, ARS, Ames, IA 50010, USA
| |
Collapse
|
24
|
Parreira R. Laboratory Methods in Molecular Epidemiology: Viral Infections. Microbiol Spectr 2018; 6:10.1128/microbiolspec.ame-0003-2018. [PMID: 30387412 PMCID: PMC11633636 DOI: 10.1128/microbiolspec.ame-0003-2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Indexed: 01/05/2023] Open
Abstract
Viruses, which are the most abundant biological entities on the planet, have been regarded as the "dark matter" of biology in the sense that despite their ubiquity and frequent presence in large numbers, their detection and analysis are not always straightforward. The majority of them are very small (falling under the limit of 0.5 μm), and collectively, they are extraordinarily diverse. In fact, the majority of the genetic diversity on the planet is found in the so-called virosphere, or the world of viruses. Furthermore, the most frequent viral agents of disease in humans display an RNA genome, and frequently evolve very fast, due to the fact that most of their polymerases are devoid of proofreading activity. Therefore, their detection, genetic characterization, and epidemiological surveillance are rather challenging. This review (part of the Curated Collection on Advances in Molecular Epidemiology of Infectious Diseases) describes many of the methods that, throughout the last few decades, have been used for viral detection and analysis. Despite the challenge of having to deal with high genetic diversity, the majority of these methods still depend on the amplification of viral genomic sequences, using sequence-specific or sequence-independent approaches, exploring thermal profiles or a single nucleic acid amplification temperature. Furthermore, viral populations, and especially those with RNA genomes, are not usually genetically uniform but encompass swarms of genetically related, though distinct, viral genomes known as viral quasispecies. Therefore, sequence analysis of viral amplicons needs to take this fact into consideration, as it constitutes a potential analytic problem. Possible technical approaches to deal with it are also described here. *This article is part of a curated collection.
Collapse
Affiliation(s)
- Ricardo Parreira
- Unidade de Microbiologia Médica/Global Health and Tropical Medicine (GHTM) Research Centre, Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), 1349-008 Lisboa, Portugal
| |
Collapse
|
25
|
Walper SA, Lasarte Aragonés G, Sapsford KE, Brown CW, Rowland CE, Breger JC, Medintz IL. Detecting Biothreat Agents: From Current Diagnostics to Developing Sensor Technologies. ACS Sens 2018; 3:1894-2024. [PMID: 30080029 DOI: 10.1021/acssensors.8b00420] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Although a fundamental understanding of the pathogenicity of most biothreat agents has been elucidated and available treatments have increased substantially over the past decades, they still represent a significant public health threat in this age of (bio)terrorism, indiscriminate warfare, pollution, climate change, unchecked population growth, and globalization. The key step to almost all prevention, protection, prophylaxis, post-exposure treatment, and mitigation of any bioagent is early detection. Here, we review available methods for detecting bioagents including pathogenic bacteria and viruses along with their toxins. An introduction placing this subject in the historical context of previous naturally occurring outbreaks and efforts to weaponize selected agents is first provided along with definitions and relevant considerations. An overview of the detection technologies that find use in this endeavor along with how they provide data or transduce signal within a sensing configuration follows. Current "gold" standards for biothreat detection/diagnostics along with a listing of relevant FDA approved in vitro diagnostic devices is then discussed to provide an overview of the current state of the art. Given the 2014 outbreak of Ebola virus in Western Africa and the recent 2016 spread of Zika virus in the Americas, discussion of what constitutes a public health emergency and how new in vitro diagnostic devices are authorized for emergency use in the U.S. are also included. The majority of the Review is then subdivided around the sensing of bacterial, viral, and toxin biothreats with each including an overview of the major agents in that class, a detailed cross-section of different sensing methods in development based on assay format or analytical technique, and some discussion of related microfluidic lab-on-a-chip/point-of-care devices. Finally, an outlook is given on how this field will develop from the perspective of the biosensing technology itself and the new emerging threats they may face.
Collapse
Affiliation(s)
- Scott A. Walper
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, D.C. 20375, United States
| | - Guillermo Lasarte Aragonés
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, D.C. 20375, United States
- College of Science, George Mason University Fairfax, Virginia 22030, United States
| | - Kim E. Sapsford
- OMPT/CDRH/OIR/DMD Bacterial Respiratory and Medical Countermeasures Branch, U.S. Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Carl W. Brown
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, D.C. 20375, United States
- College of Science, George Mason University Fairfax, Virginia 22030, United States
| | - Clare E. Rowland
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, D.C. 20375, United States
- National Research Council, Washington, D.C. 20036, United States
| | - Joyce C. Breger
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, D.C. 20375, United States
| | - Igor L. Medintz
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, D.C. 20375, United States
| |
Collapse
|
26
|
Overview of Trends in the Application of Metagenomic Techniques in the Analysis of Human Enteric Viral Diversity in Africa's Environmental Regimes. Viruses 2018; 10:v10080429. [PMID: 30110939 PMCID: PMC6115975 DOI: 10.3390/v10080429] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/03/2018] [Accepted: 08/10/2018] [Indexed: 12/19/2022] Open
Abstract
There has been an increase in the quest for metagenomics as an approach for the identification and study of the diversity of human viruses found in aquatic systems, both for their role as waterborne pathogens and as water quality indicators. In the last few years, environmental viral metagenomics has grown significantly and has enabled the identification, diversity and entire genome sequencing of viruses in environmental and clinical samples extensively. Prior to the arrival of metagenomics, traditional molecular procedures such as the polymerase chain reaction (PCR) and sequencing, were mostly used to identify and classify enteric viral species in different environmental milieu. After the advent of metagenomics, more detailed reports have emerged about the important waterborne viruses identified in wastewater treatment plant effluents and surface water. This paper provides a review of methods that have been used for the concentration, detection and identification of viral species from different environmental matrices. The review also takes into consideration where metagenomics has been explored in different African countries, as well as the limitations and challenges facing the approach. Procedures including sample processing, experimental design, sequencing technology, and bioinformatics analysis are discussed. The review concludes by summarising the current thinking and practices in the field and lays bare key issues that those venturing into this field need to consider and address.
Collapse
|
27
|
Kuca T, Passler T, Newcomer BW, Neill JD, Galik PK, Riddell KP, Zhang Y, Walz PH. Identification of Conserved Amino Acid Substitutions During Serial Infection of Pregnant Cattle and Sheep With Bovine Viral Diarrhea Virus. Front Microbiol 2018; 9:1109. [PMID: 29928264 PMCID: PMC5998738 DOI: 10.3389/fmicb.2018.01109] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 05/09/2018] [Indexed: 11/13/2022] Open
Abstract
Bovine viral diarrhea virus (BVDV) is an economically important pathogen of cattle that can also infect a wide range of domestic and wild species including sheep, goats, deer, camelids, and pigs. BVDV isolates are genetically highly diverse and previous work demonstrated that many substitutions were introduced in the viral genome during acute infections in cattle. In contrast, only limited information exists regarding changes occurring during BVDV infections in species other than cattle. The purpose of this study was to determine the changes introduced in the open reading frame (ORF) of the BVDV genome during serial infection of pregnant cattle and sheep with an isolate of bovine origin. Serial experimental inoculations were performed in six pregnant heifers and six pregnant ewes using BVDV-1b isolate AU526 in the first heifer and ewe, and serum from the preceding acutely infected dam thereafter. Complete ORF sequences were determined for 23 BVDV-1b isolates including AU526, one isolate from each pregnant dam, and one isolate from each BVDV-positive offspring born to these dams. Sequence comparison revealed that greater numbers of substitutions occurred during serial infection of pregnant sheep than of pregnant cattle. Furthermore, multiple host-specific amino acid changes were gradually introduced and conserved. These changes were more abundant in ovine isolates and occurred primarily in the E2 coding region. These results suggest that BVDV infections in heterologous species may serve as a significant source of viral genetic diversity and may be associated with adaptive changes.
Collapse
Affiliation(s)
- Thibaud Kuca
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Thomas Passler
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Benjamin W. Newcomer
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - John D. Neill
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, United States
| | - Patricia K. Galik
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Kay P. Riddell
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Yijing Zhang
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Paul H. Walz
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| |
Collapse
|
28
|
Zhang Y, Zhang C, Li B, Li Y, He XZ, Li A, Wu W, Duan SX, Qiu FZ, Wang J, Shen XX, Yang MJ, Li DX, Ma XJ. VSITA, an Improved Approach of Target Amplification in the Identification of Viral Pathogens. BIOMEDICAL AND ENVIRONMENTAL SCIENCES : BES 2018; 31:272-279. [PMID: 29773090 PMCID: PMC7135048 DOI: 10.3967/bes2018.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/03/2018] [Indexed: 06/08/2023]
Abstract
OBJECTIVE Unbiased next generation sequencing (NGS) is susceptible to interference from host or environmental sequences. Consequently, background depletion and virome enrichment techniques are usually needed for clinical samples where viral load is much lower than background sequences. METHODS A viral Sequence Independent Targeted Amplification (VSITA) approach using a set of non-ribosomal and virus-enriched octamers (V8) was developed and compared with traditionally used random hexamers (N6). Forty-five archived clinical samples of different types were used in parallel to compare the V8 and N6 enrichment performance of viral sequences and removal performance of ribosomal sequences in the step of reverse transcription followed by quantitative PCR (qPCR). Ten sera samples from patients with fever of unknown origin and 10 feces samples from patients with diarrhea of unknown origin were used in comparison of V8 and N6 enrichment performance following NGS analysis. RESULTS A minimum 30 hexamers matching to viral reference sequences (sense and antisense) were selected from a dataset of random 4,096 (46) hexamers (N6). Two random nucleotides were added to the 5' end of the selected hexamers, and 480 (30 × 42) octamers (V8) were obtained. In general, VSITA approach showed higher enrichment of virus-targeted cDNA and enhanced ability to remove unwanted ribosomal sequences in the majorities of 45 predefined clinical samples. Moreover, VSITA combined with NGS enabled to detect not only more viruses but also achieve more viral reads hit and higher viral genome coverage in 20 clinical samples with diarrhea or fever of unknown origin. CONCLUSION The VSITA approach designed in this study is demonstrated to possess higher sensitivity and broader genome coverage than traditionally used random hexamers in the NGS-based identification of viral pathogens directly from clinical samples.
Collapse
Affiliation(s)
- Yi Zhang
- Key Laboratory for Medical Virology, National Health and Family Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Chen Zhang
- Key Laboratory for Medical Virology, National Health and Family Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Bo Li
- State Key Laboratory of Plant Genomics and National Center for Plant Gene Research, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100730, China
| | - Yang Li
- Key Laboratory for Medical Virology, National Health and Family Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Xiao Zhou He
- Key Laboratory for Medical Virology, National Health and Family Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Acher Li
- Key Laboratory for Medical Virology, National Health and Family Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Wei Wu
- Key Laboratory for Medical Virology, National Health and Family Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Su Xia Duan
- Key Laboratory for Medical Virology, National Health and Family Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; Pediatric Research Institute, Children's Hospital of Hebei Province, Shijiazhuang 050031, Hebei, China
| | - Fang Zhou Qiu
- Key Laboratory for Medical Virology, National Health and Family Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; Pediatric Research Institute, Children's Hospital of Hebei Province, Shijiazhuang 050031, Hebei, China
| | - Ji Wang
- Key Laboratory for Medical Virology, National Health and Family Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Xin Xin Shen
- Key Laboratory for Medical Virology, National Health and Family Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Meng Jie Yang
- Key Laboratory for Medical Virology, National Health and Family Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - De Xin Li
- Key Laboratory for Medical Virology, National Health and Family Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Xue Jun Ma
- Key Laboratory for Medical Virology, National Health and Family Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| |
Collapse
|
29
|
He Y, Taylor TL, Dimitrov KM, Butt SL, Stanton JB, Goraichuk IV, Fenton H, Poulson R, Zhang J, Brown CC, Ip HS, Isidoro-Ayza M, Afonso CL. Whole-genome sequencing of genotype VI Newcastle disease viruses from formalin-fixed paraffin-embedded tissues from wild pigeons reveals continuous evolution and previously unrecognized genetic diversity in the U.S. Virol J 2018; 15:9. [PMID: 29329546 PMCID: PMC5767055 DOI: 10.1186/s12985-017-0914-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/18/2017] [Indexed: 11/26/2022] Open
Abstract
Background Newcastle disease viruses (NDV) are highly contagious and cause disease in both wild birds and poultry. A pigeon-adapted variant of genotype VI NDV, often termed pigeon paramyxovirus 1, is commonly isolated from columbids in the United States and worldwide. Complete genomic characterization of these genotype VI viruses circulating in wild columbids in the United States is limited, and due to the genetic variability of the virus, failure of rapid diagnostic detection has been reported. Therefore, in this study, formalin-fixed paraffin-embedded (FFPE) samples were subjected to next-generation sequencing (NGS) to identify and characterize these circulating viruses, providing valuable genetic information. NGS enables multiple samples to be deep-sequenced in parallel. When used on FFPE samples, this methodology allows for retrospective studies of infectious organisms. Methods FFPE wild pigeon tissue samples (kidney, liver and spleen) from 10 mortality events in the U.S. between 2010 and 2016 were analyzed using NGS to detect and sequence NDV genomes from randomly amplified total RNA. Results were compared to the previously published immunohistochemistry (IHC) results conducted on the same samples. Additionally, phylogenetic analyses were conducted on the complete and partial fusion gene and complete genome coding sequences. Results Twenty-three out of 29 IHC-positive FFPE pigeon samples were identified as positive for NDV by NGS. Positive samples produced an average genome coverage of 99.6% and an average median depth of 199. A previously described sub-genotype (VIa) and a novel sub-genotype (VIn) of NDV were identified as the causative agent of 10 pigeon mortality events in the U.S. from 2010 to 2016. The distribution of these viruses from the North American lineages match the distribution of the Eurasian collared-doves and rock pigeons in the U.S. Conclusions This work reports the first successful evolutionary study using deep sequencing of complete NDV genomes from FFPE samples of wild bird origin. There are at least two distinct U.S. lineages of genotype VI NDV maintained in wild pigeons that are continuously evolving independently from each other and have no evident epidemiological connections to viruses circulating abroad. These findings support the hypothesis that columbids are serving as reservoirs of virulent NDV in the U.S. Electronic supplementary material The online version of this article (10.1186/s12985-017-0914-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ying He
- Southeast Poultry Research Laboratory, Agricultural Research Service, USDA, 934 College Station Rd, Athens, GA, 30605, USA.,College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
| | - Tonya L Taylor
- Southeast Poultry Research Laboratory, Agricultural Research Service, USDA, 934 College Station Rd, Athens, GA, 30605, USA
| | - Kiril M Dimitrov
- Southeast Poultry Research Laboratory, Agricultural Research Service, USDA, 934 College Station Rd, Athens, GA, 30605, USA
| | - Salman L Butt
- Southeast Poultry Research Laboratory, Agricultural Research Service, USDA, 934 College Station Rd, Athens, GA, 30605, USA.,Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - James B Stanton
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Iryna V Goraichuk
- Southeast Poultry Research Laboratory, Agricultural Research Service, USDA, 934 College Station Rd, Athens, GA, 30605, USA.,National Scientific Center Institute of Experimental and Clinical Veterinary Medicine, Kharkiv, Ukraine
| | - Heather Fenton
- Southeastern Cooperative Wildlife Disease Study, Athens, GA, USA
| | - Rebecca Poulson
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Jian Zhang
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Corrie C Brown
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Hon S Ip
- National Wildlife Health Center-US Geological Survey, Madison, WI, USA
| | - Marcos Isidoro-Ayza
- National Wildlife Health Center-US Geological Survey, Madison, WI, USA.,Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA
| | - Claudio L Afonso
- Southeast Poultry Research Laboratory, Agricultural Research Service, USDA, 934 College Station Rd, Athens, GA, 30605, USA.
| |
Collapse
|
30
|
Spear A, Wang FX, Kappes MA, Das PB, Faaberg KS. Progress toward an enhanced vaccine: Eight marked attenuated viruses to porcine reproductive and respiratory disease virus. Virology 2018; 516:30-37. [PMID: 29324359 DOI: 10.1016/j.virol.2017.12.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/21/2017] [Accepted: 12/25/2017] [Indexed: 01/31/2023]
Abstract
Recombinant viruses of strain Ingelvac® PRRS porcine reproductive and respiratory syndrome virus (PRRSV) modified live virus vaccine were produced with two individual small in-frame deletions in nonstructural protein 2 (nsp2; Δ23 and Δ87) and also the same deletions supplanted with foreign tags (Δ23-V5, Δ23-FLAG, Δ23-S, Δ87-V5, Δ87-FLAG, Δ87-S). The viruses, but one (Δ87-FLAG), were stable for 10 passages and showed minimal effects on in vitro growth. Northern hybridization showed that the Δ23-tagged probe detected intracellular viral genome RNA as well as shorter RNAs that may represent heteroclite species, while the Δ87-tagged probe detected predominantly only genome length RNAs. When the tagged viruses were used to probe nsp2 protein in infected cells, perinuclear localization similar to native nsp2 was seen. Dual infection of Δ23-S and Δ87-S viruses allowed some discrimination of individual tagged nsp2 protein, facilitating future research. The mutants could potentially also be used to differentiate infected from vaccinated animals.
Collapse
Affiliation(s)
- Allyn Spear
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, USA.
| | - Feng-Xue Wang
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, USA.
| | - Matthew A Kappes
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, USA.
| | - Phani B Das
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, USA.
| | - Kay S Faaberg
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, USA.
| |
Collapse
|
31
|
Wright TL, Eshar D, Carpenter JW, Lin D, Padmanabhan A, Peddireddi L, Cino G. Suspected Hepadnavirus Association with a Hepatocellular Carcinoma in a Black-Tailed Prairie Dog (Cynomys ludovicianus). J Comp Pathol 2017; 157:284-290. [PMID: 29169624 DOI: 10.1016/j.jcpa.2017.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 09/12/2017] [Accepted: 09/21/2017] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinomas are the most commonly reported neoplasm of black-tailed prairie dogs (Cynomys ludovicianus). In several other closely related Sciuridae species, infection with species-specific hepadnaviruses is associated with the development of these tumours, but such a hepadnavirus has not yet been identified in any prairie dog species, although its presence has been hypothesized previously. An adult prairie dog was humanely destroyed due to progressive illness and the identification of a cranial abdominal mass that was determined on histopathology to be a hepatocellular carcinoma. Deep sequencing of the tumour tissue identified the presence of a hepadnavirus, similar in its genetic structure to woodchuck hepatitis virus. Electron microscopy showed the presence of viral particles similar in structure to other hepadnaviral particles. This report suggests that a hepadnavirus may be associated with the development of hepatocellular carcinomas in the prairie dog.
Collapse
Affiliation(s)
- T L Wright
- Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - D Eshar
- Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA.
| | - J W Carpenter
- Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - D Lin
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - A Padmanabhan
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - L Peddireddi
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - G Cino
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
32
|
van Geelen AGM, Anderson TK, Lager KM, Das PB, Otis NJ, Montiel NA, Miller LC, Kulshreshtha V, Buckley AC, Brockmeier SL, Zhang J, Gauger PC, Harmon KM, Faaberg KS. Porcine reproductive and respiratory disease virus: Evolution and recombination yields distinct ORF5 RFLP 1-7-4 viruses with individual pathogenicity. Virology 2017; 513:168-179. [PMID: 29096159 DOI: 10.1016/j.virol.2017.10.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/29/2017] [Accepted: 10/02/2017] [Indexed: 01/14/2023]
Abstract
Recent cases of porcine reproductive and respiratory syndrome virus (PRRSV) infection in United States swine-herds have been associated with high mortality in piglets and severe morbidity in sows. Analysis of the ORF5 gene from such clinical cases revealed a unique restriction fragment polymorphism (RFLP) of 1-7-4. The genome diversity of seventeen of these viruses (81.4% to 99.8% identical; collected 2013-2015) and the pathogenicity of 4 representative viruses were compared to that of SDSU73, a known moderately virulent strain. Recombination analyses revealed genomic breakpoints in structural and nonstructural regions of the genomes with evidence for recombination events between lineages. Pathogenicity varied between the isolates and the patterns were not consistent. IA/2014/NADC34, IA/2013/ISU-1 and IN/2014/ISU-5 caused more severe disease, and IA/2014/ISU-2 did not cause pyrexia and had little effect on pig growth. ORF5 RFLP genotyping was ineffectual in providing insight into isolate pathogenicity and that other parameters of virulence remain to be identified.
Collapse
Affiliation(s)
- Albert G M van Geelen
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, USA
| | - Tavis K Anderson
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, USA
| | - Kelly M Lager
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, USA
| | - Phani B Das
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, USA
| | - Nicholas J Otis
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, USA
| | - Nestor A Montiel
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, USA
| | - Laura C Miller
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, USA
| | - Vikas Kulshreshtha
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, USA
| | - Alexandra C Buckley
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, USA
| | - Susan L Brockmeier
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, USA
| | - Jianqiang Zhang
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Phillip C Gauger
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Karen M Harmon
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Kay S Faaberg
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, USA.
| |
Collapse
|
33
|
Falkenberg SM, Dassanayake RP, Neill JD, Ridpath JF. Improved detection of bovine viral diarrhea virus in bovine lymphoid cell lines using PrimeFlow RNA assay. Virology 2017; 509:260-265. [PMID: 28688346 DOI: 10.1016/j.virol.2017.06.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/23/2017] [Accepted: 06/27/2017] [Indexed: 01/29/2023]
Abstract
Bovine viral diarrhea virus (BVDV) infections, whether as acute, persistent or contributing to co-infections, result in significant losses for cattle producers. Although, BVDV can be identified readily by real-time PCR and ELISA, detection and quantification of viral infection at the single cell level is extremely difficult. Detection at the single lymphoid cell level is important due to the immunomodulation that accompanies BVDV infection. A novel PrimeFlow RNA assay using in-situ detection of BVDV was evaluated. The model used to develop this technique included three BL-3 cell lines with different infection statuses, one not infected with BVDV, one infected with BVDV and one dual infected with BVDV and bovine leukosis virus. Using RNA probes specific for the BVDV-2a Npro-Erns coding region, BVDV RNA was detected from both contaminated BL-3 cell lines by flow cytometry and fluorescent microscopy. This is the first report on in-situ detection of BVDV at the single-cell level.
Collapse
Affiliation(s)
- Shollie M Falkenberg
- Ruminant Disease and Immunology Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA 50010, United States.
| | - Rohana P Dassanayake
- Ruminant Disease and Immunology Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA 50010, United States
| | - John D Neill
- Ruminant Disease and Immunology Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA 50010, United States
| | - Julia F Ridpath
- Ruminant Disease and Immunology Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA 50010, United States
| |
Collapse
|
34
|
Lin HH, Liao YC. drVM: a new tool for efficient genome assembly of known eukaryotic viruses from metagenomes. Gigascience 2017; 6:1-10. [PMID: 28369462 PMCID: PMC5466706 DOI: 10.1093/gigascience/gix003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 01/15/2017] [Indexed: 11/29/2022] Open
Abstract
Background: Virus discovery using high-throughput next-generation sequencing has become more commonplace. However, although analysis of deep next-generation sequencing data allows us to identity potential pathogens, the entire analytical procedure requires competency in the bioinformatics domain, which includes implementing proper software packages and preparing prerequisite databases. Simple and user-friendly bioinformatics pipelines are urgently required to obtain complete viral genome sequences from metagenomic data. Results: This manuscript presents a pipeline, drVM (detect and reconstruct known viral genomes from metagenomes), for rapid viral read identification, genus-level read partition, read normalization, de novo assembly, sequence annotation, and coverage profiling. The first two procedures and sequence annotation rely on known viral genomes as a reference database. drVM was validated via the analysis of over 300 sequencing runs generated by Illumina and Ion Torrent platforms to provide complete viral genome assemblies for a variety of virus types including DNA viruses, RNA viruses, and retroviruses. drVM is available for free download at: https://sourceforge.net/projects/sb2nhri/files/drVM/ and is also assembled as a Docker container, an Amazon machine image, and a virtual machine to facilitate seamless deployment. Conclusions: drVM was compared with other viral detection tools to demonstrate its merits in terms of viral genome completeness and reduced computation time. This substantiates the platform's potential to produce prompt and accurate viral genome sequences from clinical samples.
Collapse
|
35
|
Ridpath JF, Neill JD, Palmer MV, Bauermann FV, Falkenberg SM, Wolff PL. Isolation and characterization of a novel cervid adenovirus from white-tailed deer (Odocoileus virginianus) fawns in a captive herd. Virus Res 2017; 238:198-203. [PMID: 28662929 DOI: 10.1016/j.virusres.2017.06.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/23/2017] [Accepted: 06/24/2017] [Indexed: 10/19/2022]
Abstract
A novel adenovirus, CeAdV1, was isolated from buffy coat and nasal swab samples collected from two captive white-tailed deer (Odocoileus virginianus) fawns. The isolation was an incidental finding in the course of screening animals for use in a research study on an unrelated pathogen. In the screening process, virus isolation was performed on both nasal swabs and buffy coat samples and cytopathic effect was observed. Electron microscopy revealed viral particles with the shape and morphology of an adenovirus. Next generation sequencing followed by phylogenetic analysis classified this virus to the Mastadenovirus genus. Its sequence was genetically distinct from all other recognized species in this genus, with only 76% sequence identity to its closest genetic match, bovine adenovirus 3 (BAdV3). The virus could be propagated in bovine derived cells but grew to a higher titer in cervid derived cells. Inoculation of white-tailed deer fawns with the isolated virus resulted in pyrexia, depletion of thymus tissue and mild respiratory disease. Comparative serology performed using convalescent sera revealed distinct antigenic differences between the novel cervid adenovirus and BAdV3. A retrospective serological survey of the captive deer herd indicated that this virus had been circulating in the herd for at least 14 years with no report of clinical disease. A survey of serum collected from free ranging mule deer residing in Nevada revealed high serum titers against this novel adenovirus.
Collapse
Affiliation(s)
- Julia F Ridpath
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, PO Box 70, 1920 Dayton Avenue, Ames, IA, 50010, USA.
| | - John D Neill
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, PO Box 70, 1920 Dayton Avenue, Ames, IA, 50010, USA
| | - Mitchel V Palmer
- Bacterial Diseases of Livestock Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, PO Box 70, 1920 Dayton Avenue, Ames, IA, 50010, USA
| | - Fernando V Bauermann
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, PO Box 70, 1920 Dayton Avenue, Ames, IA, 50010, USA
| | - Shollie M Falkenberg
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, PO Box 70, 1920 Dayton Avenue, Ames, IA, 50010, USA
| | - Peregrine L Wolff
- Nevada Department of Wildlife, 6980 Sierra Center Pkwy, Suite 120, Reno, NV, 89511, USA
| |
Collapse
|
36
|
Fulton RW, Neill JD, Saliki JT, Landis C, Burge LJ, Payton ME. Genomic and antigenic characterization of bovine parainfluenza-3 viruses in the United States including modified live virus vaccine (MLV) strains and field strains from cattle. Virus Res 2017; 235:77-81. [PMID: 28416404 PMCID: PMC7172726 DOI: 10.1016/j.virusres.2017.04.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/13/2017] [Accepted: 04/11/2017] [Indexed: 10/29/2022]
Abstract
This study investigated the genetic and antigenic characterization of parainfluenza-3 virus (PI3V) of cattle. Using molecular tests including real time PCR and viral genome sequencing, PI3V strains could be separated into PI3V types, including PI3V A, PI3V B, and PI3V C. Isolates from cattle with bovine respiratory disease clinical signs and commercial vaccines in the U.S. with MLV PI3V were typed using these molecular tests. All the MLV vaccine strains tested were PI3V A. In most cases PI3V field strains from calves receiving MLV vaccines were types heterologous to the vaccine type A. Also antigenic differences were noted as PI3V C strains had lower antibody levels than PI3V A in serums from cattle receiving MLV PI3V A vaccines. This study further demonstrates there is genetic variability of U.S. PI3V strains and also antigenic variability. In addition, isolates from cattle with BRD signs and receiving MLV vaccines may have heterologous types to the vaccines, and molecular tests should be performed to differentiate field from vaccine strains. Potentially the efficacy of current PI3V A vaccines should be evaluated with other types such a PI3V B and PI3V C.
Collapse
Affiliation(s)
- R W Fulton
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74078 USA.
| | - J D Neill
- U.S. Department of Agriculture, Agricultural Research Service, National Animal Diseases Center, Ames, IA 50010 USA
| | - J T Saliki
- Athens Veterinary Diagnostic Laboratory, University of Georgia, Athens, GA 30602 USA
| | - C Landis
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74078 USA
| | - L J Burge
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74078 USA
| | - M E Payton
- Department of Statistics, Oklahoma State University, Stillwater, OK 74078 USA
| |
Collapse
|
37
|
Vitosh-Sillman S, Loy JD, Brodersen B, Kelling C, Doster A, Topliff C, Nelson E, Bai J, Schirtzinger E, Poulsen E, Meadors B, Anderson J, Hause B, Anderson G, Hesse R. Experimental infection of conventional nursing pigs and their dams with Porcine deltacoronavirus. J Vet Diagn Invest 2017; 28:486-97. [PMID: 27578872 DOI: 10.1177/1040638716654200] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Porcine deltacoronavirus (PDCoV) is a newly identified virus that has been detected in swine herds of North America associated with enteric disease. The aim of this study was to demonstrate the pathogenicity, course of infection, virus kinetics, and aerosol transmission of PDCoV using 87 conventional piglets and their 9 dams, including aerosol and contact controls to emulate field conditions. Piglets 2-4 days of age and their dams were administered an oronasal PDCoV inoculum with a quantitative real-time reverse transcription (qRT)-PCR quantification cycle (Cq) value of 22 that was generated from a field sample having 100% nucleotide identity to USA/Illinois121/2014 determined by metagenomic sequencing and testing negative for other enteric disease agents using standard assays. Serial samples of blood, serum, oral fluids, nasal and fecal swabs, and tissues from sequential autopsy, conducted daily on days 1-8 and regular intervals thereafter, were collected throughout the 42-day study for qRT-PCR, histopathology, and immunohistochemistry. Diarrhea developed in all inoculated and contact control pigs, including dams, by 2 days post-inoculation (dpi) and in aerosol control pigs and dams by 3-4 dpi, with resolution occurring by 12 dpi. Mild to severe atrophic enteritis with PDCoV antigen staining was observed in the small intestine of affected piglets from 2 to 8 dpi. Mesenteric lymph node and small intestine were the primary sites of antigen detection by immunohistochemistry, and virus RNA was detected in these tissues to the end of the study. Virus RNA was detectable in piglet fecal swabs to 21 dpi, and dams to 14-35 dpi.
Collapse
Affiliation(s)
- Sarah Vitosh-Sillman
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE (Vitosh-Sillman, Loy, Brodersen, Kelling, Doster, Topliff)Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD (Nelson)Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Manhattan, KS (Bai, Schirtzinger, Poulsen, Meadors, J Anderson, Hause, G Anderson, Hesse)
| | - John Dustin Loy
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE (Vitosh-Sillman, Loy, Brodersen, Kelling, Doster, Topliff)Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD (Nelson)Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Manhattan, KS (Bai, Schirtzinger, Poulsen, Meadors, J Anderson, Hause, G Anderson, Hesse)
| | - Bruce Brodersen
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE (Vitosh-Sillman, Loy, Brodersen, Kelling, Doster, Topliff)Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD (Nelson)Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Manhattan, KS (Bai, Schirtzinger, Poulsen, Meadors, J Anderson, Hause, G Anderson, Hesse)
| | - Clayton Kelling
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE (Vitosh-Sillman, Loy, Brodersen, Kelling, Doster, Topliff)Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD (Nelson)Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Manhattan, KS (Bai, Schirtzinger, Poulsen, Meadors, J Anderson, Hause, G Anderson, Hesse)
| | - Alan Doster
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE (Vitosh-Sillman, Loy, Brodersen, Kelling, Doster, Topliff)Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD (Nelson)Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Manhattan, KS (Bai, Schirtzinger, Poulsen, Meadors, J Anderson, Hause, G Anderson, Hesse)
| | - Christina Topliff
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE (Vitosh-Sillman, Loy, Brodersen, Kelling, Doster, Topliff)Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD (Nelson)Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Manhattan, KS (Bai, Schirtzinger, Poulsen, Meadors, J Anderson, Hause, G Anderson, Hesse)
| | - Eric Nelson
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE (Vitosh-Sillman, Loy, Brodersen, Kelling, Doster, Topliff)Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD (Nelson)Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Manhattan, KS (Bai, Schirtzinger, Poulsen, Meadors, J Anderson, Hause, G Anderson, Hesse)
| | - Jianfa Bai
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE (Vitosh-Sillman, Loy, Brodersen, Kelling, Doster, Topliff)Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD (Nelson)Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Manhattan, KS (Bai, Schirtzinger, Poulsen, Meadors, J Anderson, Hause, G Anderson, Hesse)
| | - Erin Schirtzinger
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE (Vitosh-Sillman, Loy, Brodersen, Kelling, Doster, Topliff)Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD (Nelson)Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Manhattan, KS (Bai, Schirtzinger, Poulsen, Meadors, J Anderson, Hause, G Anderson, Hesse)
| | - Elizabeth Poulsen
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE (Vitosh-Sillman, Loy, Brodersen, Kelling, Doster, Topliff)Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD (Nelson)Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Manhattan, KS (Bai, Schirtzinger, Poulsen, Meadors, J Anderson, Hause, G Anderson, Hesse)
| | - Barbara Meadors
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE (Vitosh-Sillman, Loy, Brodersen, Kelling, Doster, Topliff)Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD (Nelson)Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Manhattan, KS (Bai, Schirtzinger, Poulsen, Meadors, J Anderson, Hause, G Anderson, Hesse)
| | - Joseph Anderson
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE (Vitosh-Sillman, Loy, Brodersen, Kelling, Doster, Topliff)Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD (Nelson)Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Manhattan, KS (Bai, Schirtzinger, Poulsen, Meadors, J Anderson, Hause, G Anderson, Hesse)
| | - Benjamin Hause
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE (Vitosh-Sillman, Loy, Brodersen, Kelling, Doster, Topliff)Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD (Nelson)Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Manhattan, KS (Bai, Schirtzinger, Poulsen, Meadors, J Anderson, Hause, G Anderson, Hesse)
| | - Gary Anderson
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE (Vitosh-Sillman, Loy, Brodersen, Kelling, Doster, Topliff)Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD (Nelson)Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Manhattan, KS (Bai, Schirtzinger, Poulsen, Meadors, J Anderson, Hause, G Anderson, Hesse)
| | - Richard Hesse
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE (Vitosh-Sillman, Loy, Brodersen, Kelling, Doster, Topliff)Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD (Nelson)Kansas State Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Manhattan, KS (Bai, Schirtzinger, Poulsen, Meadors, J Anderson, Hause, G Anderson, Hesse)
| |
Collapse
|
38
|
Dimitrov KM, Sharma P, Volkening JD, Goraichuk IV, Wajid A, Rehmani SF, Basharat A, Shittu I, Joannis TM, Miller PJ, Afonso CL. A robust and cost-effective approach to sequence and analyze complete genomes of small RNA viruses. Virol J 2017; 14:72. [PMID: 28388925 PMCID: PMC5384157 DOI: 10.1186/s12985-017-0741-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 03/29/2017] [Indexed: 01/26/2023] Open
Abstract
Background Next-generation sequencing (NGS) allows ultra-deep sequencing of nucleic acids. The use of sequence-independent amplification of viral nucleic acids without utilization of target-specific primers provides advantages over traditional sequencing methods and allows detection of unsuspected variants and co-infecting agents. However, NGS is not widely used for small RNA viruses because of incorrectly perceived cost estimates and inefficient utilization of freely available bioinformatics tools. Methods In this study, we have utilized NGS-based random sequencing of total RNA combined with barcode multiplexing of libraries to quickly, effectively and simultaneously characterize the genomic sequences of multiple avian paramyxoviruses. Thirty libraries were prepared from diagnostic samples amplified in allantoic fluids and their total RNAs were sequenced in a single flow cell on an Illumina MiSeq instrument. After digital normalization, data were assembled using the MIRA assembler within a customized workflow on the Galaxy platform. Results Twenty-eight avian paramyxovirus 1 (APMV-1), one APMV-13, four avian influenza and two infectious bronchitis virus complete or nearly complete genome sequences were obtained from the single run. The 29 avian paramyxovirus genomes displayed 99.6% mean coverage based on bases with Phred quality scores of 30 or more. The lower and upper quartiles of sample median depth per position for those 29 samples were 2984 and 6894, respectively, indicating coverage across samples sufficient for deep variant analysis. Sample processing and library preparation took approximately 25–30 h, the sequencing run took 39 h, and processing through the Galaxy workflow took approximately 2–3 h. The cost of all steps, excluding labor, was estimated to be 106 USD per sample. Conclusions This work describes an efficient multiplexing NGS approach, a detailed analysis workflow, and customized tools for the characterization of the genomes of RNA viruses. The combination of multiplexing NGS technology with the Galaxy workflow platform resulted in a fast, user-friendly, and cost-efficient protocol for the simultaneous characterization of multiple full-length viral genomes. Twenty-nine full-length or near-full-length APMV genomes with a high median depth were successfully sequenced out of 30 samples. The applied de novo assembly approach also allowed identification of mixed viral populations in some of the samples. Electronic supplementary material The online version of this article (doi:10.1186/s12985-017-0741-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kiril M Dimitrov
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Road, Athens, GA, 30605, USA
| | - Poonam Sharma
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Road, Athens, GA, 30605, USA
| | | | - Iryna V Goraichuk
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Road, Athens, GA, 30605, USA.,National Scientific Center Institute of Experimental and Clinical Veterinary Medicine, 83 Pushkinskaya Street, Kharkiv, 61023, Ukraine
| | - Abdul Wajid
- Quality Operations Laboratory (QOL), University of Veterinary and Animal Sciences, Syed Abdul Qadir Jilani Road, Lahore, 54000, Pakistan.,Institute of Biochemistry and Biotechnology, University of Veterinary and Animal Sciences, Syed Abdul Qadir Jilani Road, Lahore, 54000, Pakistan
| | - Shafqat Fatima Rehmani
- Quality Operations Laboratory (QOL), University of Veterinary and Animal Sciences, Syed Abdul Qadir Jilani Road, Lahore, 54000, Pakistan
| | - Asma Basharat
- Quality Operations Laboratory (QOL), University of Veterinary and Animal Sciences, Syed Abdul Qadir Jilani Road, Lahore, 54000, Pakistan
| | - Ismaila Shittu
- Regional Laboratory for Animal Influenza and other Transboundary Animal Diseases, National Veterinary Research Institute, PMB01, Vom, 930010, Plateau State, Nigeria
| | - Tony M Joannis
- Regional Laboratory for Animal Influenza and other Transboundary Animal Diseases, National Veterinary Research Institute, PMB01, Vom, 930010, Plateau State, Nigeria
| | - Patti J Miller
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Road, Athens, GA, 30605, USA
| | - Claudio L Afonso
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Road, Athens, GA, 30605, USA.
| |
Collapse
|
39
|
Wang CH, Nie K, Zhang Y, Wang J, Zhou SF, Li XN, Zhou HY, Qi SX, Ma XJ. An Improved Barcoded Oligonucleotide Primers-based Next-generation Sequencing Approach for Direct Identification of Viral Pathogens in Clinical Specimens. BIOMEDICAL AND ENVIRONMENTAL SCIENCES : BES 2017; 30:22-34. [PMID: 28245896 PMCID: PMC7136949 DOI: 10.3967/bes2017.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 12/20/2016] [Indexed: 06/06/2023]
Abstract
OBJECTIVE To provide a feasible and cost-effective next-generation sequencing (NGS) method for accurate identification of viral pathogens in clinical specimens, because enormous limitations impede the clinical use of common NGS, such as high cost, complicated procedures, tremendous data analysis, and high background noise in clinical samples. METHODS Viruses from cell culture materials or clinical specimens were identified following an improved NGS procedure: reduction of background noise by sample preprocessing, viral enrichment by barcoded oligonucleotide (random hexamer or non-ribosomal hexanucleotide) primer-based amplification, fragmentation-free library construction and sequencing of one-tube mixtures, as well as rapid data analysis using an in-house pipeline. RESULTS NGS data demonstrated that both barcoded primer sets were useful to simultaneously capture multiple viral pathogens in cell culture materials or clinical specimens and verified that hexanucleotide primers captured as many viral sequences as hexamers did. Moreover, direct testing of clinical specimens using this improved hexanucleotide primer-based NGS approach provided further detailed genotypes of enteroviruses causing hand, foot, and mouth disease (HFMD) and identified other potential viruses or differentiated misdiagnosis events. CONCLUSION The improved barcoded oligonucleotide primer-based NGS approach is simplified, time saving, cost effective, and appropriate for direct identification of viral pathogens in clinical practice.
Collapse
Affiliation(s)
- Chun Hua Wang
- Key Laboratory for Medical Virology, National Health and Fam-ily Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Kai Nie
- Key Laboratory for Medical Virology, National Health and Fam-ily Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Yi Zhang
- Key Laboratory for Medical Virology, National Health and Fam-ily Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ji Wang
- Key Laboratory for Medical Virology, National Health and Fam-ily Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Shuai Feng Zhou
- Key Laboratory for Medical Virology, National Health and Fam-ily Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; Center for Disease Prevention and Control of Hunan Province, Changsha 410005, Hunan, China
| | - Xin Na Li
- Key Laboratory for Medical Virology, National Health and Fam-ily Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Hang Yu Zhou
- Key Laboratory for Medical Virology, National Health and Fam-ily Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Shun Xiang Qi
- Institute for Viral Disease Control and Prevention, Center for Disease Control and Prevention of Hebei, Shijiazhuang 050000, Hebei, China
| | - Xue Jun Ma
- Key Laboratory for Medical Virology, National Health and Fam-ily Planning Commission, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| |
Collapse
|
40
|
A Novel Porcine Circovirus Distantly Related to Known Circoviruses Is Associated with Porcine Dermatitis and Nephropathy Syndrome and Reproductive Failure. J Virol 2016; 91:JVI.01879-16. [PMID: 27795441 DOI: 10.1128/jvi.01879-16] [Citation(s) in RCA: 422] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 10/20/2016] [Indexed: 01/21/2023] Open
Abstract
Porcine circovirus-associated disease (PCVAD) is clinically manifested by postweaning multisystemic wasting syndrome (PMWS), respiratory and enteric disease, reproductive failure, and porcine dermatitis and nephropathy syndrome (PDNS). Porcine circovirus 2 (PCV2) is an essential component of PCVAD, although an etiologic role in PDNS is not well established. Here, a novel circovirus, designated porcine circovirus 3 (PCV3), was identified in sows that died acutely with PDNS-like clinical signs. The capsid and replicase proteins of PCV3 are only 37% and 55% identical to PCV2 and bat circoviruses, respectively. Aborted fetuses from sows with PDNS contained high levels of PCV3 (7.57 × 107 genome copies/ml), and no other viruses were detected by PCR and metagenomic sequencing. Immunohistochemistry (IHC) analysis of sow tissue samples identified PCV3 antigen in skin, kidney, lung, and lymph node samples localized in typical PDNS lesions, including necrotizing vasculitis, glomerulonephritis, granulomatous lymphadenitis, and bronchointerstitial pneumonia. Further study of archived PDNS tissue samples that were negative for PCV2 by IHC analysis identified 45 of 48 that were PCV3 positive by quantitative PCR (qPCR), with 60% of a subset also testing positive for PCV3 by IHC analysis. Analysis by qPCR of 271 porcine respiratory disease diagnostic submission samples identified 34 PCV3-positive cases (12.5%), and enzyme-linked immunosorbent assay detection of anti-PCV3 capsid antibodies in serum samples found that 46 (55%) of 83 samples tested were positive. These results suggest that PCV3 commonly circulates within U.S. swine and may play an etiologic role in reproductive failure and PDNS. Because of the high economic impact of PCV2, this novel circovirus warrants further studies to elucidate its significance and role in PCVAD. IMPORTANCE While porcine circovirus 2 (PCV2) was first identified in sporadic cases of postweaning multisystemic wasting syndrome in Canada in the early 1990s, an epidemic of severe systemic disease due to PCV2 spread worldwide in the ensuing decade. Despite being effectively controlled by commercial vaccines, PCV2 remains one of the most economically significant viruses of swine. Here, a novel porcine circovirus (PCV3) that is distantly related to known circoviruses was identified in sows with porcine dermatitis and nephropathy syndrome (PDNS) and reproductive failure. PCV2, which has previously been associated with these clinical presentations, was not identified. High levels of PCV3 nucleic acid were observed in aborted fetuses by quantitative PCR, and PCV3 antigen was localized in histologic lesions typical of PDNS in sows by immunohistochemistry (IHC) analysis. PCV3 was also identified in archival PDNS diagnostic samples that previously tested negative for PCV2 by IHC analysis. The emergence of PCV3 warrants further investigation.
Collapse
|
41
|
Mitra N, Cernicchiaro N, Torres S, Li F, Hause BM. Metagenomic characterization of the virome associated with bovine respiratory disease in feedlot cattle identified novel viruses and suggests an etiologic role for influenza D virus. J Gen Virol 2016; 97:1771-1784. [PMID: 27154756 DOI: 10.1099/jgv.0.000492] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Bovine respiratory disease (BRD) is the most costly disease affecting the cattle industry. The pathogenesis of BRD is complex and includes contributions from microbial pathogens as well as host, environmental and animal management factors. In this study, we utilized viral metagenomic sequencing to explore the virome of nasal swab samples obtained from feedlot cattle with acute BRD and asymptomatic pen-mates at six and four feedlots in Mexico and the USA, respectively, in April-October 2015. Twenty-one viruses were detected, with bovine rhinitis A (52.7 %) and B (23.7 %) virus, and bovine coronavirus (24.7 %) being the most commonly identified. The emerging influenza D virus (IDV) tended to be significantly associated (P=0.134; odds ratio=2.94) with disease, whereas viruses commonly associated with BRD such as bovine viral diarrhea virus, bovine herpesvirus 1, bovine respiratory syncytial virus and bovine parainfluenza 3 virus were detected less frequently. The detection of IDV was further confirmed using a real-time PCR assay. Nasal swabs from symptomatic animals had significantly more IDV RNA than those collected from healthy animals (P=0.04). In addition to known viruses, new genotypes of bovine rhinitis B virus and enterovirus E were identified and a newly proposed species of bocaparvovirus, Ungulate bocaparvovirus 6, was characterized. Ungulate tetraparvovirus 1 was also detected for the first time in North America to our knowledge. These results illustrate the complexity of the virome associated with BRD and highlight the need for further research into the contribution of other viruses to BRD pathogenesis.
Collapse
Affiliation(s)
- Namita Mitra
- Veterinary Diagnostic Laboratory, 1800 Denison Avenue, Kansas State University, Manhattan, KS 66506, USA
| | - Natalia Cernicchiaro
- Department of Diagnostic Medicine and Pathobiology, 1800 Denison Avenue, Kansas State University, Manhattan, KS 66506, USA
| | - Siddartha Torres
- Merck Animal Health, 2 Giralda Farms, Madison City, NJ 07940, USA
| | - Feng Li
- Departments of Biology and Microbiology and Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Ben M Hause
- Department of Diagnostic Medicine and Pathobiology, 1800 Denison Avenue, Kansas State University, Manhattan, KS 66506, USA.,Veterinary Diagnostic Laboratory, 1800 Denison Avenue, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
42
|
Hause BM, Palinski R, Hesse R, Anderson G. Highly diverse posaviruses in swine faeces are aquatic in origin. J Gen Virol 2016; 97:1362-1367. [PMID: 27002315 DOI: 10.1099/jgv.0.000461] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Posaviruses are a group of highly divergent viruses identified in swine faeces that are distantly related to other members of the order Picornavirales. Eighteen posavirus genomes were assembled from 10 out of 25 (40 %) faecal-swab pools collected from healthy adult swine. Phylogenetic analysis of the conserved RNA-dependent RNA polymerase (Pol) domain found that posaviruses form a large, highly diverse, monophyletic clade, which includes similar viruses identified in human (husavirus) and fish (fisavirus) faeces or intestinal contents, respectively. Quantitative reverse transcription PCR analysis of water samples collected from commercial swine barns identified four out of 19 (21 %) samples were positive using a 5'-nuclease assay targeting the Pol region of posavirus 1. ICPD (immunoprecipitation coupled to PCR detection) assays to explore serological evidence of posavirus infection found only a single positive sample, suggesting posaviruses do not commonly infect swine, and together these results suggests a likely aquatic host.
Collapse
Affiliation(s)
- Ben M Hause
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS 66506, USA
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, USA
| | - Rachel Palinski
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, USA
| | - Richard Hesse
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, USA
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS 66506, USA
| | - Gary Anderson
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, USA
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
43
|
Discovery of a novel Parvovirinae virus, porcine parvovirus 7, by metagenomic sequencing of porcine rectal swabs. Virus Genes 2016; 52:564-7. [PMID: 26995221 DOI: 10.1007/s11262-016-1322-1] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/12/2016] [Indexed: 12/23/2022]
Abstract
Parvoviruses are a diverse group of viruses containing some of the smallest known species that are capable of infecting a wide range of animals. Metagenomic sequencing of pooled rectal swabs from adult pigs identified a 4103-bp contig consisting of two major open reading frames encoding proteins of 672 and 469 amino acids (aa) in length. BLASTP analysis of the 672-aa protein found 42.4 % identity to fruit bat (Eidolon helvum) parvovirus 2 (EhPV2) and 37.9 % to turkey parvovirus (TuPV) TP1-2012/HUN NS1 proteins. The 469-aa protein had no significant similarity to known proteins. Genetic and phylogenetic analyses suggest that PPV7, EhPV2, and TuPV represent a novel genus in the family Parvoviridae. Quantitative PCR screening of 182 porcine diagnostic samples found a total of 16 positives (8.6 %). Together, these data suggest that PPV7 is a highly divergent novel parvovirus prevalent within the US swine.
Collapse
|
44
|
Flowers EM, Bachvaroff TR, Warg JV, Neill JD, Killian ML, Vinagre AS, Brown S, Almeida ASE, Schott EJ. Genome Sequence Analysis of CsRV1: A Pathogenic Reovirus that Infects the Blue Crab Callinectes sapidus Across Its Trans-Hemispheric Range. Front Microbiol 2016; 7:126. [PMID: 26904003 PMCID: PMC4748042 DOI: 10.3389/fmicb.2016.00126] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 01/25/2015] [Indexed: 01/15/2023] Open
Abstract
The blue crab, Callinectes sapidus Rathbun, 1896, which is a commercially important trophic link in coastal ecosystems of the western Atlantic, is infected in both North and South America by C. sapidus Reovirus 1 (CsRV1), a double stranded RNA virus. The 12 genome segments of a North American strain of CsRV1 were sequenced using Ion Torrent technology. Putative functions could be assigned for 3 of the 13 proteins encoded in the genome, based on their similarity to proteins encoded in other reovirus genomes. Comparison of the CsRV1 RNA-dependent RNA polymerase (RdRP) sequence to genomes of other crab-infecting reoviruses shows that it is similar to the mud crab reovirus found in Scylla serrata and WX-2012 in Eriocheir sinensis, Chinese mitten crab, and supports the idea that there is a distinct “Crabreo” genus, different from Seadornavirus and Cardoreovirus, the two closest genera in the Reoviridae. A region of 98% nucleotide sequence identity between CsRV1 and the only available sequence of the P virus of Macropipus depurator suggests that these two viruses may be closely related. An 860 nucleotide region of the CsRV1 RdRP gene was amplified and sequenced from 15 infected crabs collected from across the geographic range of C. sapidus. Pairwise analysis of predicted protein sequences shows that CsRV1 strains in Brazil can be distinguished from those in North America based on conserved residues in this gene. The sequencing, annotation, and preliminary population metrics of the genome of CsRV1 should facilitate additional studies in diverse disciplines, including structure-function relationships of reovirus proteins, investigations into the evolution of the Reoviridae, and biogeographic research on the connectivity of C. sapidus populations across the Northern and Southern hemispheres.
Collapse
Affiliation(s)
- Emily M Flowers
- Institute of Marine and Environmental Technology, University of Maryland Center for Environmental ScienceBaltimore, MD USA; University of Maryland School of MedicineBaltimore, MD USA
| | - Tsvetan R Bachvaroff
- Institute of Marine and Environmental Technology, University of Maryland Center for Environmental Science Baltimore, MD USA
| | - Janet V Warg
- National Veterinary Services Laboratories, Animal and Plant Health Inspection Service, United States Department of Agriculture Ames, IA USA
| | - John D Neill
- National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture Ames, IA USA
| | - Mary L Killian
- National Veterinary Services Laboratories, Animal and Plant Health Inspection Service, United States Department of Agriculture Ames, IA USA
| | - Anapaula S Vinagre
- Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| | - Shanai Brown
- Department of Biology, Morgan State University Baltimore, MD USA
| | - Andréa Santos E Almeida
- Institute of Marine and Environmental Technology, University of Maryland Center for Environmental Science Baltimore, MD USA
| | - Eric J Schott
- Institute of Marine and Environmental Technology, University of Maryland Center for Environmental Science Baltimore, MD USA
| |
Collapse
|
45
|
MULTICENTRIC T-CELL LYMPHOMA AND CUTANEOUS HEMANGIOSARCOMA IN A CAPTIVE CHEETAH (ACINONYX JUBATUS). J Zoo Wildl Med 2015; 46:961-6. [PMID: 26667562 DOI: 10.1638/2015-0118.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A 13-yr-old intact male cheetah (Acinonyx jubatus) presented for evaluation after a 4-mo history of intermittent lethargy and increased expiratory effort. The clinical signs were initially noted after the diagnosis and death of its 13-yr-old male sibling with solitary hepatic T-cell lymphoma. Physical examination findings included thin body condition, harsh lung sounds, peripheral lymphadenopathy, and a cutaneous mass on the right medial tarsus and scrotum. Excisional biopsies diagnosed well-differentiated cutaneous hemangiosarcomas. Thoracic radiographs revealed a cranial mediastinal mass. Complete blood count and serum biochemical analyses showed a leukocytosis with persistent lymphocytosis, progressive azotemia, and markedly elevated alkaline phosphatase. Because of the cheetah's declining quality of life, euthanasia was elected. Postmortem examination, histopathology, and immunohistochemical staining revealed multicentric T-cell lymphoma. Feline leukemia virus (FeLV) enzyme-linked immunosorbent assay, FeLV polymerase chain reaction (whole blood), and viral metagenomic analysis were negative. This is the first case of cutaneous hemangiosarcoma and multicentric T-cell lymphoma reported in a FeLV-negative cheetah.
Collapse
|
46
|
Palinski RM, Chen Z, Henningson JN, Lang Y, Rowland RRR, Fang Y, Prickett J, Gauger PC, Hause BM. Widespread detection and characterization of porcine parainfluenza virus 1 in pigs in the USA. J Gen Virol 2015; 97:281-286. [PMID: 26581410 DOI: 10.1099/jgv.0.000343] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Porcine parainfluenza virus 1 (PPIV1) was first identified in 2013 in slaughterhouse pigs in Hong Kong, China. Here, two near-complete genomes were assembled from swine exhibiting acute respiratory disease that were 90.0-95.3% identical to Chinese PPIV1. Analysis of the HN gene from ten additional PPIV1-positive samples found 85.0-95.5% identity, suggesting genetic diversity between strains. Molecular analysis identified 17 out of 279 (6.1%) positive samples from pigs with respiratory disease. Eleven nursery pigs from a naturally infected herd were asymptomatic; however, nasal swabs from six pigs and the lungs of a single pig were quantitative reverse transcriptase (qRT)-PCR positive. Histopathology identified PPIV1 RNA in the nasal respiratory epithelium and trachea. Two serological assays demonstrated seroconversion of infected pigs and further analysis of 59 swine serum samples found 52.5% and 66.1% seropositivity, respectively. Taken together, the results confirm the widespread presence of PPIV1 in the US swine herd.
Collapse
Affiliation(s)
- Rachel M Palinski
- Kansas State Department of Diagnostic Medicine and Pathobiology, Manhattan, Kansas, USA
| | - Zhenhai Chen
- Kansas State Department of Diagnostic Medicine and Pathobiology, Manhattan, Kansas, USA
| | - Jamie N Henningson
- Kansas State Department of Diagnostic Medicine and Pathobiology, Manhattan, Kansas, USA.,Kansas State Veterinary Diagnostic Laboratory, Manhattan, Kansas, USA
| | - Yuekun Lang
- Kansas State Department of Diagnostic Medicine and Pathobiology, Manhattan, Kansas, USA
| | - Raymond R R Rowland
- Kansas State Department of Diagnostic Medicine and Pathobiology, Manhattan, Kansas, USA
| | - Ying Fang
- Kansas State Department of Diagnostic Medicine and Pathobiology, Manhattan, Kansas, USA
| | - John Prickett
- Carthage Veterinary Service, Carthage, Illinois, USA
| | - Phillip C Gauger
- Department of Veterinary Diagnostic and Population Animal Medicine, Iowa State University, Ames, Iowa, USA
| | - Ben M Hause
- Kansas State Veterinary Diagnostic Laboratory, Manhattan, Kansas, USA.,Kansas State Department of Diagnostic Medicine and Pathobiology, Manhattan, Kansas, USA
| |
Collapse
|
47
|
Datta S, Budhauliya R, Das B, Chatterjee S, Vanlalhmuaka, Veer V. Next-generation sequencing in clinical virology: Discovery of new viruses. World J Virol 2015; 4:265-276. [PMID: 26279987 PMCID: PMC4534817 DOI: 10.5501/wjv.v4.i3.265] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 03/23/2015] [Accepted: 05/08/2015] [Indexed: 02/05/2023] Open
Abstract
Viruses are a cause of significant health problem worldwide, especially in the developing nations. Due to different anthropological activities, human populations are exposed to different viral pathogens, many of which emerge as outbreaks. In such situations, discovery of novel viruses is utmost important for deciding prevention and treatment strategies. Since last century, a number of different virus discovery methods, based on cell culture inoculation, sequence-independent PCR have been used for identification of a variety of viruses. However, the recent emergence and commercial availability of next-generation sequencers (NGS) has entirely changed the field of virus discovery. These massively parallel sequencing platforms can sequence a mixture of genetic materials from a very heterogeneous mix, with high sensitivity. Moreover, these platforms work in a sequence-independent manner, making them ideal tools for virus discovery. However, for their application in clinics, sample preparation or enrichment is necessary to detect low abundance virus populations. A number of techniques have also been developed for enrichment or viral nucleic acids. In this manuscript, we review the evolution of sequencing; NGS technologies available today as well as widely used virus enrichment technologies. We also discuss the challenges associated with their applications in the clinical virus discovery.
Collapse
|
48
|
Hause BM, Collin EA, Peddireddi L, Yuan F, Chen Z, Hesse RA, Gauger PC, Clement T, Fang Y, Anderson G. Discovery of a novel putative atypical porcine pestivirus in pigs in the USA. J Gen Virol 2015. [PMID: 26219947 DOI: 10.1099/jgv.0.000251] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Pestiviruses are some of the most significant pathogens affecting ruminants and swine. Here, we assembled a 11 276 bp contig encoding a predicted 3635 aa polyprotein from porcine serum with 68 % pairwise identity to that of a recently partially characterized Rhinolophus affinis pestivirus (RaPV) and approximately 25-28 % pairwise identity to those of other pestiviruses. The virus was provisionally named atypical porcine pestivirus (APPV). Metagenomic sequencing of 182 serum samples identified four additional APPV-positive samples. Positive samples originated from five states and ELISAs using recombinant APPV Erns found cross-reactive antibodies in 94 % of a collection of porcine serum samples, suggesting widespread distribution of APPV in the US swine herd. The molecular and serological results suggest that APPV is a novel, highly divergent porcine pestivirus widely distributed in US pigs.
Collapse
Affiliation(s)
- Ben M Hause
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, Kansas, USA.,Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas, USA
| | - Emily A Collin
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, Kansas, USA.,Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas, USA
| | - Lalitha Peddireddi
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, Kansas, USA.,Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas, USA
| | - Fangfeng Yuan
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas, USA
| | - Zhenhai Chen
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas, USA
| | - Richard A Hesse
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, Kansas, USA.,Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas, USA
| | - Phillip C Gauger
- Department of Veterinary Diagnostic and Population Animal Medicine, Iowa State University, Ames, Iowa, USA
| | - Travis Clement
- Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD, USA
| | - Ying Fang
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas, USA
| | - Gary Anderson
- Kansas State Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, Kansas, USA.,Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
49
|
Eberle KC, Neill JD, Venn-Watson SK, McGill JL, Sacco RE. Novel Atlantic bottlenose dolphin parainfluenza virus TtPIV-1 clusters with bovine PIV-3 genotype B strains. Virus Genes 2015; 51:198-208. [PMID: 26174699 DOI: 10.1007/s11262-015-1224-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 06/30/2015] [Indexed: 01/02/2023]
Abstract
Parainfluenza virus 3 (PIV-3) is a common viral infection not only in humans, but also in many other species. Serological evidence suggests that nearly 100 % of children in the United States have been infected with PIV-3 by 5 years of age. Similarly, in cattle, PIV-3 is commonly associated with bovine respiratory disease complex. A novel dolphin PIV-3 (TtPIV-1) was described by Nollens et al. in 2008 from a dolphin that was diagnosed with an unknown respiratory illness. At that time, TtPIV-1 was found to be most similar to, but distinct from, bovine PIV-3 (BPIV-3). In the present study, similar viral growth kinetics and pro-inflammatory cytokine (IL-1β, IL-6, and CXCL8) production were seen between BPIV-3 and TtPIV-1 in BEAS-2B, MDBK, and Vero cell lines. Initial nomenclature of TtPIV-1 was based on partial sequence of the fusion and RNA polymerase genes. Based on the similarities we saw with the in vitro work, it was important to examine the TtPIV-1 genome in more detail. Full genome sequencing and subsequent phylogenetic analysis revealed that all six viral genes of TtPIV-1 clustered within the recently described BPIV-3 genotype B strains, and it is proposed that TtPIV-1 be re-classified with BPIV-3 genotype B strains.
Collapse
Affiliation(s)
- Kirsten C Eberle
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA.,Molecular Cellular and Developmental Biology Graduate Program, Iowa State University, Ames, IA, USA.,Immunobiology Graduate Program, Iowa State University, Ames, IA, USA
| | - John D Neill
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| | - Stephanie K Venn-Watson
- Translational Medicine & Research Program, National Marine Mammal Foundation, San Diego, CA, USA
| | - Jodi L McGill
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA.,Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Randy E Sacco
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA. .,Molecular Cellular and Developmental Biology Graduate Program, Iowa State University, Ames, IA, USA. .,Immunobiology Graduate Program, Iowa State University, Ames, IA, USA.
| |
Collapse
|
50
|
Neill JD, Ridpath JF, Valayudhan BT. Identification and genome characterization of genotype B and genotype C bovine parainfluenza type 3 viruses isolated in the United States. BMC Vet Res 2015; 11:112. [PMID: 25976921 PMCID: PMC4438627 DOI: 10.1186/s12917-015-0431-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/08/2015] [Indexed: 11/30/2022] Open
Abstract
Background Bovine parainfluenza 3 viruses (BPI3V) are respiratory pathogens of cattle that cause disease singly but are often associated with bovine respiratory disease complex (BRDC) in conjunction with other viral and bacterial agents. Bovine vaccines currently contain BPI3V to provide protection against the virus, but there is no current information regarding the BPI3V strains that are circulating in the U.S. Results A project was initiated to sequence archival BPI3V isolates to study viral evolution over time. This was done with a deep sequencing protocol that generated sequences of multiple RNA virus genomes simultaneously. Analysis of the BPI3V sequences revealed that, in addition to the genotype A (BPI3Va) viruses previously described in the United States, there were two additional genotypes of BPI3V circulating that had been described only in Australia (BPI3Vb) and Asia (BPI3Vc). The U.S. BPI3Vb and BPI3Vc isolates showed some divergence from the Australian and Asian strains; the BPI3Vb were 93 % similar to the Australian Q5592 strain and the BPI3Vc viruses were 98 % similar to the 12Q061 strain that was described in South Korea. Overall, the three genotypes were 82 to 84 % identical to each other and 80 % identical to the most similar human PI3V. Cross-neutralization studies using an APHIS/NVSL BPI3V reference serum showed that neutralization titers against the genotype B and C viruses were 4- to ≥16-fold less then the titer against the APHIS BPI3Va reference strain, SF-4. Conclusions This study clearly demonstrated that BPI3Vb and BPI3Vc strains, previously thought to be foreign to the U.S., are indeed circulating in domestic livestock herds. Based on virus neutralization using polyclonal antisera, there were antigenic differences between viruses from these genotypes and the BPI3Va viruses that are included in currently marketed bovine vaccines. Further study of these viruses is warranted to determine pathogenic potential and cross-protection afforded by vaccination.
Collapse
Affiliation(s)
- John D Neill
- USDA, Agricultural Research Service, National Animal Disease Center, Ruminant Diseases and Immunology Research Unit, 1920 Dayton Avenue, Ames, IA, 50010, USA.
| | - Julia F Ridpath
- USDA, Agricultural Research Service, National Animal Disease Center, Ruminant Diseases and Immunology Research Unit, 1920 Dayton Avenue, Ames, IA, 50010, USA.
| | - Binu T Valayudhan
- Texas A&M Veterinary Medical Diagnostic Laboratory, Amarillo, TX, USA.
| |
Collapse
|