Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.571
Filter
1.
Emerg Microbes Infect ; 13(1): 2353292, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38712345

ABSTRACT

ABSTRACTRapid evolution of highly pathogenic avian influenza viruses (HPAIVs) is driven by antigenic drift but also by reassortment, which might result in robust replication in and transmission to mammals. Recently, spillover of clade 2.3.4.4b HPAIV to mammals including humans, and their transmission between mammalian species has been reported. This study aimed to evaluate the pathogenicity and transmissibility of a mink-derived clade 2.3.4.4b H5N1 HPAIV isolate from Spain in pigs. Experimental infection caused interstitial pneumonia with necrotizing bronchiolitis with high titers of virus present in the lower respiratory tract and 100% seroconversion. Infected pigs shed limited amount of virus, and importantly, there was no transmission to contact pigs. Notably, critical mammalian-like adaptations such as PB2-E627 K and HA-Q222L emerged at low frequencies in principal-infected pigs. It is concluded that pigs are highly susceptible to infection with the mink-derived clade 2.3.4.4b H5N1 HPAIV and provide a favorable environment for HPAIV to acquire mammalian-like adaptations.


Subject(s)
Influenza A Virus, H5N1 Subtype , Mink , Orthomyxoviridae Infections , Swine Diseases , Animals , Mink/virology , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/veterinary , Swine , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/physiology , Influenza A Virus, H5N1 Subtype/isolation & purification , Swine Diseases/virology , Swine Diseases/transmission , Spain , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Shedding
2.
Emerg Infect Dis ; 30(6): 1228-1231, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38782033
3.
Nat Food ; 5(5): 413-422, 2024 May.
Article in English | MEDLINE | ID: mdl-38724686

ABSTRACT

Salmonella enterica causes severe food-borne infections through contamination of the food supply chain. Its evolution has been associated with human activities, especially animal husbandry. Advances in intensive farming and global transportation have substantially reshaped the pig industry, but their impact on the evolution of associated zoonotic pathogens such as S. enterica remains unresolved. Here we investigated the population fluctuation, accumulation of antimicrobial resistance genes and international serovar Choleraesuis transmission of nine pig-enriched S. enterica populations comprising more than 9,000 genomes. Most changes were found to be attributable to the developments of the modern pig industry. All pig-enriched salmonellae experienced host transfers in pigs and/or population expansions over the past century, with pigs and pork having become the main sources of S. enterica transmissions to other hosts. Overall, our analysis revealed strong associations between the transmission of pig-enriched salmonellae and the global pork trade.


Subject(s)
Salmonella enterica , Animals , Salmonella enterica/genetics , Salmonella enterica/isolation & purification , Swine , Europe/epidemiology , Humans , Salmonella Infections, Animal/epidemiology , Salmonella Infections, Animal/transmission , Salmonella Infections, Animal/microbiology , Swine Diseases/microbiology , Swine Diseases/transmission , Swine Diseases/epidemiology , Animal Husbandry/methods , Pork Meat/microbiology , Americas/epidemiology , Food Microbiology
5.
Vet Microbiol ; 293: 110101, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718529

ABSTRACT

Cross-species transmission of coronaviruses has been continuously posing a major challenge to public health. Pigs, as the major animal reservoirs for many zoonotic viruses, frequently mediate viral transmission to humans. This study comprehensively mapped the relationship between human and porcine coronaviruses through in-depth bioinformatics analysis. We found that human coronavirus OC43 and porcine coronavirus PHEV share a close phylogenetic relationship, evidenced by high genomic homology, similar codon usage patterns and comparable tertiary structure in spike proteins. Inoculation of infectious OC43 viruses in organoids derived from porcine small and large intestine demonstrated that porcine intestinal organoids (pIOs) are highly susceptible to human coronavirus OC43 infection and support infectious virus production. Using transmission electron microscopy, we visualized OC43 viral particles in both intracellular and extracellular compartments, and observed abnormalities of multiple organelles in infected organoid cells. Robust OC43 infections in pIOs result in a significant reduction of organoids viability and widespread cell death. This study bears essential implications for better understanding the evolutionary origin of human coronavirus OC43, and provides a proof-of-concept for using pIOs as a model to investigate cross-species transmission of human coronavirus.


Subject(s)
Computational Biology , Coronavirus Infections , Coronavirus OC43, Human , Intestines , Organoids , Phylogeny , Animals , Organoids/virology , Swine , Humans , Coronavirus Infections/virology , Coronavirus Infections/transmission , Coronavirus Infections/veterinary , Coronavirus OC43, Human/physiology , Coronavirus OC43, Human/genetics , Intestines/virology , Swine Diseases/virology , Swine Diseases/transmission , Genome, Viral
6.
J Antimicrob Chemother ; 79(5): 962-967, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38442335

ABSTRACT

BACKGROUND: Livestock-associated MRSA (LA-MRSA) transmission/cross-contamination can occur at abattoir through colonized pigs, increasing occupational hazards and health concerns for workers. To assess this risk we used genomics to identify LA-MRSA lineages present in batches of pigs sent to slaughter and distribution of clones. METHODS: WGS was performed on 85 LA-MRSA previously isolated from six abattoirs from 105 batches of pigs sent from 100 UK farms. spa typing and MLST were performed on all isolates. A mashtree tree was constructed to compare genomes of the LA-MRSA with 1281 global isolates from livestock and humans. A phylogenetic tree and pairwise SNP distance matrices were built from whole genomes of 109 isolates closest to those from abattoirs to compare evolutionary relationships and identify clones. RESULTS: All abattoir isolates belonged to CC398 and were mainly of spa type t011, although other spa types were present. Phylogenetic analysis confirmed the abattoir isolates were most closely related to each other and to pig LA-MRSA from across Europe, indicating a common evolutionary origin with related lineages colonizing UK pigs.Comparison of genomes using SNPs suggested between one and four clones were transferring between pigs from different batches. Transmission likely occurred on farm premises, during transportation, and/or within abattoirs through contact with contaminated surfaces in lairage or post-stunning. CONCLUSIONS: Genomics forensically identified related isolates/clones circulating in pigs at slaughter, showing contamination occurs often. Results suggest that further genomic tracking will identify hotspots, and improvements in measures such as biosecurity and disinfection will help reduce risk for workers.


Subject(s)
Abattoirs , Livestock , Methicillin-Resistant Staphylococcus aureus , Phylogeny , Staphylococcal Infections , Whole Genome Sequencing , Animals , Swine , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Methicillin-Resistant Staphylococcus aureus/classification , Staphylococcal Infections/transmission , Staphylococcal Infections/microbiology , Staphylococcal Infections/veterinary , Staphylococcal Infections/epidemiology , United Kingdom/epidemiology , Livestock/microbiology , Multilocus Sequence Typing , Swine Diseases/transmission , Swine Diseases/microbiology , Genomics , Genome, Bacterial , Polymorphism, Single Nucleotide , Humans , Genotype
7.
Virol Sin ; 39(2): 205-217, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38346538

ABSTRACT

Swine are regarded as "intermediate hosts" or "mixing vessels" of influenza viruses, capable of generating strains with pandemic potential. From 2020 to 2021, we conducted surveillance on swine H1N2 influenza (swH1N2) viruses in swine farms located in Guangdong, Yunnan, and Guizhou provinces in southern China, as well as Henan and Shandong provinces in northern China. We systematically analyzed the evolution and pathogenicity of swH1N2 isolates, and characterized their replication and transmission abilities. The isolated viruses are quadruple reassortant H1N2 viruses containing genes from pdm/09 H1N1 (PB2, PB1, PA and NP genes), triple-reassortant swine (NS gene), Eurasian Avian-like (HA and M genes), and recent human H3N2 (NA gene) lineages. The NA, PB2, and NP of SW/188/20 and SW/198/20 show high gene similarities to A/Guangdong/Yue Fang277/2017 (H3N2). The HA gene of swH1N2 exhibits a high evolutionary rate. The five swH1N2 isolates replicate efficiently in human, canine, and swine cells, as well as in the turbinate, trachea, and lungs of mice. A/swine/Shandong/198/2020 strain efficiently replicates in the respiratory tract of pigs and effectively transmitted among them. Collectively, these current swH1N2 viruses possess zoonotic potential, highlighting the need for strengthened surveillance of swH1N2 viruses.


Subject(s)
Evolution, Molecular , Influenza A Virus, H1N2 Subtype , Orthomyxoviridae Infections , Reassortant Viruses , Swine Diseases , Animals , Swine , Reassortant Viruses/genetics , Reassortant Viruses/pathogenicity , Reassortant Viruses/isolation & purification , China/epidemiology , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/veterinary , Swine Diseases/virology , Swine Diseases/transmission , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H1N2 Subtype/pathogenicity , Influenza A Virus, H1N2 Subtype/isolation & purification , Humans , Mice , Dogs , Phylogeny , Virus Replication , Public Health , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza, Human/virology , Influenza, Human/transmission , Mice, Inbred BALB C , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/isolation & purification , Virulence , Female
8.
Vet Res ; 54(1): 4, 2023 Jan 24.
Article in English | MEDLINE | ID: mdl-36694192

ABSTRACT

In 2019 a low pathogenic H3N1 avian influenza virus (AIV) caused an outbreak in Belgian poultry farms, characterized by an unusually high mortality in chickens. Influenza A viruses of the H1 and H3 subtype can infect pigs and become established in swine populations. Therefore, the H3N1 epizootic raised concern about AIV transmission to pigs and from pigs to humans. Here, we assessed the replication efficiency of this virus in explants of the porcine respiratory tract and in pigs, using virus titration and/or RT-qPCR. We also examined transmission from directly, intranasally inoculated pigs to contact pigs. The H3N1 AIV replicated to moderate titers in explants of the bronchioles and lungs, but not in the nasal mucosa or trachea. In the pig infection study, infectious virus was only detected in a few lung samples collected between 1 and 3 days post-inoculation. Virus titers were between 1.7 and 4.8 log10 TCID50. In line with the ex vivo experiment, no virus was isolated from the upper respiratory tract of pigs. In the transmission experiment, we could not detect virus transmission from directly inoculated to contact pigs. An increase in serum antibody titers was observed only in the inoculated pigs. We conclude that the porcine respiratory tract tissue explants can be a useful tool to assess the replication efficiency of AIVs in pigs. The H3N1 AIV examined here is unlikely to pose a risk to swine populations. However, continuous risk assessment studies of emerging AIVs in pigs are necessary, since different virus strains will have different genotypic and phenotypic traits.


Subject(s)
Influenza A virus , Influenza in Birds , Poultry Diseases , Animals , Humans , Antibodies, Viral/blood , Chickens , Influenza in Birds/transmission , Influenza in Birds/virology , Lung , Poultry Diseases/transmission , Poultry Diseases/virology , Swine , Swine Diseases/transmission , Swine Diseases/virology
9.
Vet Res ; 53(1): 50, 2022 Jul 07.
Article in English | MEDLINE | ID: mdl-35799280

ABSTRACT

Humans can become infected with hepatitis E virus (HEV) by consumption of undercooked pork. To reduce the burden of HEV in humans, mitigation on pig farms is needed. HEV is found on most pig farms globally, yet within-farm seroprevalence estimates vary considerably. Understanding of the underlying variation in infection dynamics within and between farms currently lacks. Therefore, we investigated HEV infection dynamics by sampling 1711 batches of slaughter pigs from 208 Dutch farms over an 8-month period. Four farm types, conventional, organic, and two types with strict focus on biosecurity, were included. Sera were tested individually with an anti-HEV antibody ELISA and pooled per batch with PCR. All farms delivered seropositive pigs to slaughter, yet batches (resembling farm compartments) had varying results. By combining PCR and ELISA results, infection moment and extent per batch could be classified as low transmission, early, intermediate or late. Cluster analysis of batch infection moments per farm resulted in four clusters with distinct infection patterns. Cluster 1 farms delivered almost exclusively PCR negative, ELISA positive batches to slaughter (PCR-ELISA+), indicating relatively early age of HEV infection. Cluster 2 and 3 farms delivered 0.3 and 0.7 of batches with intermediate infection moment (PCR+ELISA+) respectively and only few batches with early infection. Cluster 4 farms delivered low transmission (PCR-ELISA-) and late infection (PCR+ELISA-) batches, demonstrating that those farms can prevent or delay HEV transmission to farm compartments. Farm type partly coincided with cluster assignment, indicating that biosecurity and management are related to age of HEV infection.


Subject(s)
Abattoirs , Aging , Farms , Hepatitis E , Swine Diseases , Swine , Age Factors , Animals , Cluster Analysis , Cross-Sectional Studies , Enzyme-Linked Immunosorbent Assay , Farms/standards , Farms/statistics & numerical data , Hepatitis E/epidemiology , Hepatitis E/transmission , Hepatitis E/veterinary , Hepatitis E/virology , Hepatitis E virus/genetics , Hepatitis E virus/isolation & purification , Polymerase Chain Reaction , Seroepidemiologic Studies , Swine/virology , Swine Diseases/epidemiology , Swine Diseases/transmission , Swine Diseases/virology
10.
J Virol ; 96(14): e0047722, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35758666

ABSTRACT

The mechanisms of colostrum-mediated virus transmission are difficult to elucidate because of the absence of experimental animal models and the difficulties in tissue sample collection from mothers in the peripartum period. Porcine epidemic diarrhea virus (PEDV) is a reemerging enteropathogenic coronavirus that has catastrophic impacts on the global pig industry. PEDV primarily infects neonatal piglets by multiple routes, especially 1- to 2-day-old neonatal piglets. Here, our epidemiological investigation and animal challenge experiments revealed that PEDV could be vertically transmitted from sows to neonatal piglets via colostrum, and CD3+ T cells in the colostrum play an important role in this process. The results showed that PEDV colonizing the intestinal epithelial cells (IECs) of orally immunized infected sows could be transferred to CD3+ T cells located just beneath the IECs. Next, PEDV-carrying CD3+ T cells, with the expression of integrin α4ß7 and CCR10, migrate from the intestine to the mammary gland through blood circulation. Arriving in the mammary gland, PEDV-carrying CD3+ T cells could be transported across mammary epithelial cells (MECs) into the lumen (colostrum), as illustrated by an autotransfusion assay and an MECs/T coculture system. The PEDV-carrying CD3+ T cells in colostrum could be interspersed between IECs of neonatal piglets, causing intestinal infection via cell-to-cell contact. Our study demonstrates for the first time that colostrum-derived CD3+ T cells comprise a potential route for the vertical transmission of PEDV. IMPORTANCE The colostrum represents an important infection route for many viruses. Here, we demonstrate the vertical transmission of porcine epidemic diarrhea virus (PEDV) from sows to neonatal piglets via colostrum. PEDV colonizing the intestinal epithelial cells could transfer the virus to CD3+ T cells located in the sow intestine. The PEDV-carrying CD3+ T cells in the sow intestine, with the expression of integrin α4ß7 and CCR10, arrive at the mammary gland through blood circulation and are transported across mammary epithelial cells into the lumen, finally leading to intestinal infection via cell-to-cell contact in neonatal piglets. Our study not only demonstrates an alternative route of PEDV infection but also provides an animal model of vertical transmission of human infectious disease.


Subject(s)
Colostrum , Coronavirus Infections , Infectious Disease Transmission, Vertical , Porcine epidemic diarrhea virus , Swine Diseases , Animals , Animals, Newborn , Colostrum/virology , Coronavirus Infections/transmission , Coronavirus Infections/veterinary , Female , Infectious Disease Transmission, Vertical/veterinary , Porcine epidemic diarrhea virus/physiology , Swine , Swine Diseases/transmission , Swine Diseases/virology , T-Lymphocytes/virology
11.
J Virol ; 96(9): e0038022, 2022 05 11.
Article in English | MEDLINE | ID: mdl-35435723

ABSTRACT

Crossing the endothelium from the entry site and spreading in the bloodstream are crucial but obscure steps in the pathogenesis of many emerging viruses. Previous studies confirmed that porcine epidemic diarrhea virus (PEDV) caused intestinal infection by intranasal inoculation. However, the role of the nasal endothelial barrier in PEDV translocation remains unclear. Here, we demonstrated that PEDV infection causes nasal endothelial dysfunction to favor viral dissemination. Intranasal inoculation with PEDV compromised the integrity of endothelial cells (ECs) in nasal microvessels. The matrix metalloproteinase 7 (MMP-7) released from the PEDV-infected nasal epithelial cells (NECs) contributed to the destruction of endothelial integrity by degrading the tight junctions, rather than direct PEDV infection. Moreover, the proinflammatory cytokines released from PEDV-infected NECs activated ECs to upregulate ICAM-1 expression, which favored peripheral blood mononuclear cells (PBMCs) migration. PEDV could further exploit migrated cells to favor viral dissemination. Together, our results reveal the mechanism by which PEDV manipulates the endothelial dysfunction to favor viral dissemination and provide novel insights into how coronavirus interacts with the endothelium. IMPORTANCE The endothelial barrier is the last but vital defense against systemic viral transmission. Porcine epidemic diarrhea virus (PEDV) can cause severe atrophic enteritis and acute viremia. However, the mechanisms by which the virus crosses the endothelial barrier and causes viremia are poorly understood. In this study, we revealed the mechanisms of endothelial dysfunction in PEDV infection. The viral infection activates NECs and causes the upregulation of MMP-7 and proinflammatory cytokines. Using NECs, ECs, and PBMCs as in vitro models, we determined that the released MMP-7 contributed to the destruction of endothelial barrier, and the released proinflammatory cytokines activated ECs to facilitate PBMCs migration. Moreover, the virus further exploited the migrated cells to promote viral dissemination. Thus, our results provide new insights into the mechanisms underlying endothelial dysfunction induced by coronavirus infection.


Subject(s)
Coronavirus Infections , Endothelium , Porcine epidemic diarrhea virus , Swine Diseases , Virus Shedding , Animals , Coronavirus Infections/transmission , Coronavirus Infections/virology , Cytokines , Endothelium/virology , Intercellular Adhesion Molecule-1/genetics , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Matrix Metalloproteinase 7/metabolism , Porcine epidemic diarrhea virus/physiology , Swine , Swine Diseases/immunology , Swine Diseases/transmission , Swine Diseases/virology , Viremia
12.
Transbound Emerg Dis ; 69(1): 72-87, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34237198

ABSTRACT

Transboundary movement of animal feed and feed ingredients has been identified as a route for pathogen incursions. While imports of animals and animal-derived products are highly regulated for the purpose of infectious disease prevention, there has been less consideration of the viability of infectious agents in inanimate products, such as feed. This study investigated the ability of foot-and-mouth disease virus (FMDV) to remain infectious as a contaminant of commercial whole pig feed and select pig feed ingredients, and to establish the minimum infectious dose (MIDF ) required to cause foot-and-mouth disease (FMD) in pigs that consumed contaminated feed. FMDV viability in vitro varied depending on virus strain, feed product, and storage temperature, with increased duration of infectivity in soybean meal compared to pelleted whole feed. Specifically, both strains of FMDV evaluated remained viable through to the end of the 37 day observation period in experimentally contaminated soybean meal stored at 4 or 20°C . The MIDF for pigs consuming contaminated feed varied across virus strains and exposure duration in the range of 106.2 to 107 TCID50 . The ability of FMDV to cause infection in exposed pigs was mitigated by pre-treatment of feed with two commercially available feed additives, based on either formaldehyde (SalCURB®) or lactic acid (Guardian™). Our findings demonstrate that FMDV may remain infectious in pig feed ingredients for durations compatible with transoceanic transport. Although the observed MIDF was relatively high, variations in feeding conditions and biophysical characteristics of different virus strains may alter the probability of infection. These findings may be used to parameterize modelling of the risk of FMDV incursions and to regulate feed importation to minimize the risk of inadvertent importation.


Subject(s)
Animal Feed/virology , Food Contamination , Foot-and-Mouth Disease , Swine Diseases , Animals , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease/transmission , Foot-and-Mouth Disease Virus , Swine , Swine Diseases/prevention & control , Swine Diseases/transmission
13.
Emerg Microbes Infect ; 11(1): 91-94, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34839786

ABSTRACT

In order to assess the risk of SARS-CoV-2 infection, transmission and reservoir development in swine, we combined results of an experimental and two observational studies. First, intranasal and intratracheal challenge of eight pigs did not result in infection, based on clinical signs and PCR on swab and lung tissue samples. Two serum samples returned a low positive result in virus neutralization, in line with findings in other infection experiments in pigs. Next, a retrospective observational study was performed in the Netherlands in the spring of 2020. Serum samples (N =417) obtained at slaughter from 17 farms located in a region with a high human case incidence in the first wave of the pandemic. Samples were tested with protein micro array, plaque reduction neutralization test and receptor-binding-domain ELISA. None of the serum samples was positive in all three assays, although six samples from one farm returned a low positive result in PRNT (titers 40-80). Therefore we conclude that serological evidence for large scale transmission was not observed. Finally, an outbreak of respiratory disease in pigs on one farm, coinciding with recent exposure to SARS-CoV-2 infected animal caretakers, was investigated. Tonsil swabs and paired serum samples were tested. No evidence for infection with SARS-CoV-2 was found. In conclusion, Although in both the experimental and the observational study few samples returned low antibody titer results in PRNT infection with SARS-CoV-2 was not confirmed. It was concluded that sporadic infections in the field cannot be excluded, but large-scale SARS-CoV-2 transmission among pigs is unlikely.


Subject(s)
COVID-19/veterinary , SARS-CoV-2/physiology , Swine Diseases/epidemiology , Swine Diseases/transmission , Swine Diseases/virology , Animals , Environmental Exposure , Netherlands/epidemiology , Public Health Surveillance , Retrospective Studies , Swine
14.
Transbound Emerg Dis ; 69(1): 66-71, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34694061

ABSTRACT

Porcine epidemic diarrhoea virus (PEDV) infects pigs of all ages causing vomiting and diarrhoea. PEDV is transmitted via the oral-faecal route, and a very low dose is enough to infect susceptible pigs, resulting in significant production losses. This short communication aims to describe the introduction of PEDV into a 10,000-sow farrow-to-wean farm located in northwest Mexico. Following the onset of clinical signs, an outbreak investigation was conducted to determine the most probable route of introduction. Based on data collected from interviews, construction of a timeline of events, and the detection of PEDV RNA in feed samples and samples collected from various surfaces of feed transport vehicles, it was concluded that the most probable route for PEDV incursion into this breeding herd was contaminated feed or a contaminated feed transport vehicle. This paper describes how feed or feed transport could serve as potential routes of PEDV infection to a farm and highlights the importance of establishing biosecurity programs to mitigate these risks.


Subject(s)
Animal Feed/virology , Coronavirus Infections , Food Contamination , Porcine epidemic diarrhea virus , Swine Diseases , Animals , Biosecurity , Coronavirus Infections/epidemiology , Coronavirus Infections/transmission , Coronavirus Infections/veterinary , Diarrhea/epidemiology , Diarrhea/veterinary , Disease Outbreaks/veterinary , Female , Mexico/epidemiology , Porcine epidemic diarrhea virus/genetics , Swine , Swine Diseases/epidemiology , Swine Diseases/transmission , Swine Diseases/virology
15.
Comput Math Methods Med ; 2021: 6985008, 2021.
Article in English | MEDLINE | ID: mdl-34671417

ABSTRACT

Swine influenza viruses (SIVs) can unforeseeably cross the species barriers and directly infect humans, which pose huge challenges for public health and trigger pandemic risk at irregular intervals. Computational tools are needed to predict infection phenotype and early pandemic risk of SIVs. For this purpose, we propose a feature representation algorithm to predict cross-species infection of SIVs. We built a high-quality dataset of 1902 viruses. A feature representation learning scheme was applied to learn feature representations from 64 well-trained random forest models with multiple feature descriptors of mutant amino acid in the viral proteins, including compositional information, position-specific information, and physicochemical properties. Class and probabilistic information were integrated into the feature representations, and redundant features were removed by feature space optimization. High performance was achieved using 20 informative features and 22 probabilistic information. The proposed method will facilitate SIV characterization of transmission phenotype.


Subject(s)
Influenza A virus/genetics , Influenza A virus/pathogenicity , Orthomyxoviridae Infections/veterinary , Swine Diseases/virology , Algorithms , Amino Acid Sequence , Amino Acids/analysis , Amino Acids/genetics , Animals , Computational Biology , Host Specificity , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza A virus/classification , Influenza, Human/epidemiology , Influenza, Human/transmission , Influenza, Human/virology , Machine Learning , Models, Statistical , Mutation , Orthomyxoviridae Infections/virology , Pandemics , Risk Factors , Swine , Swine Diseases/transmission , Viral Proteins/chemistry , Viral Proteins/genetics
16.
Nat Microbiol ; 6(11): 1455-1465, 2021 11.
Article in English | MEDLINE | ID: mdl-34702977

ABSTRACT

Understanding the evolutionary adaptations that enable avian influenza viruses to transmit in mammalian hosts could allow better detection of zoonotic viruses with pandemic potential. We applied ancestral sequence reconstruction to gain viruses representing different adaptive stages of the European avian-like (EA) H1N1 swine influenza virus as it transitioned from avian to swine hosts since 1979. Ancestral viruses representing the avian-like precursor virus and EA swine influenza viruses from 1979-1983, 1984-1987 and 1988-1992 were reconstructed and characterized. Glycan-binding analyses showed stepwise changes in the haemagglutinin receptor-binding specificity of the EA swine influenza viruses-that is, from recognition of both α2,3- and α2,6-linked sialosides to recognition of α2,6-linked sialosides only; however, efficient transmission in piglets was enabled by adaptive changes in the viral polymerase protein and nucleoprotein, which have been fixed since 1983. PB1-Q621R and NP-R351K increased viral replication and transmission in piglets when introduced into the 1979-1983 ancestral virus that lacked efficient transmissibility. The stepwise adaptation of an avian influenza virus to a mammalian host suggests that there may be opportunities to intervene and prevent interspecies jumps through strategic coordination of surveillance and risk assessment activities.


Subject(s)
Adaptation, Physiological , Influenza A Virus, H1N1 Subtype/physiology , Influenza in Birds/virology , Orthomyxoviridae Infections/veterinary , Swine Diseases/virology , Animals , Birds , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A Virus, H1N1 Subtype/classification , Influenza A Virus, H1N1 Subtype/genetics , Influenza in Birds/transmission , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/virology , Phylogeny , Polysaccharides/chemistry , Polysaccharides/metabolism , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Swine , Swine Diseases/metabolism , Swine Diseases/transmission , Virus Replication
17.
PLoS Negl Trop Dis ; 15(10): e0009885, 2021 10.
Article in English | MEDLINE | ID: mdl-34705827

ABSTRACT

BACKGROUND: The pork tapeworm (Taenia solium) is a parasitic helminth that imposes a major health and economic burden on poor rural populations around the world. As recognized by the World Health Organization, a key barrier for achieving control of T. solium is the lack of an accurate and validated simulation model with which to study transmission and evaluate available control and elimination strategies. CystiAgent is a spatially-explicit agent based model for T. solium that is unique among T. solium models in its ability to represent key spatial and environmental features of transmission and simulate spatially targeted interventions, such as ring strategy. METHODS/PRINCIPAL FINDINGS: We validated CystiAgent against results from the Ring Strategy Trial (RST)-a large cluster-randomized trial conducted in northern Peru that evaluated six unique interventions for T. solium control in 23 villages. For the validation, each intervention strategy was replicated in CystiAgent, and the simulated prevalences of human taeniasis, porcine cysticercosis, and porcine seroincidence were compared against prevalence estimates from the trial. Results showed that CystiAgent produced declines in transmission in response to each of the six intervention strategies, but overestimated the effect of interventions in the majority of villages; simulated prevalences for human taenasis and porcine cysticercosis at the end of the trial were a median of 0.53 and 5.0 percentages points less than prevalence observed at the end of the trial, respectively. CONCLUSIONS/SIGNIFICANCE: The validation of CystiAgent represented an important step towards developing an accurate and reliable T. solium transmission model that can be deployed to fill critical gaps in our understanding of T. solium transmission and control. To improve model accuracy, future versions would benefit from improved data on pig immunity and resistance, field effectiveness of anti-helminthic treatment, and factors driving spatial clustering of T. solium infections including dispersion and contact with T. solium eggs in the environment.


Subject(s)
Cysticercosis/transmission , Cysticercosis/veterinary , Swine Diseases/transmission , Taenia solium/physiology , Zoonoses/transmission , Animals , Cysticercosis/epidemiology , Cysticercosis/parasitology , Epidemiological Models , Female , Humans , Peru/epidemiology , Prospective Studies , Rural Population/statistics & numerical data , Spatial Analysis , Swine , Swine Diseases/epidemiology , Swine Diseases/parasitology , Taenia solium/genetics , Taenia solium/isolation & purification , Zoonoses/epidemiology , Zoonoses/parasitology
18.
Am J Trop Med Hyg ; 106(1): 182-186, 2021 10 25.
Article in English | MEDLINE | ID: mdl-34695799

ABSTRACT

Rift Valley fever phlebovirus (RVFV) is a mosquito-transmitted phlebovirus (Family: Phenuiviridae, Order: Bunyavirales) causing severe neonatal mortality and abortion primarily in domestic ruminants. The susceptibility of young domestic swine to RVFV and this species' role in geographic expansion and establishment of viral endemicity is unclear. Six commercially bred Landrace-cross piglets were inoculated subcutaneously with 105 plaque-forming units of RVFV ZH501 strain and two piglets received a sham inoculum. All animals were monitored for clinical signs, viremia, viral shedding, and antibody response for 14 days. Piglets did not develop evidence of clinical disease, become febrile, or experience decreased weight gain during the study period. A brief lymphopenia followed by progressive lymphocytosis was observed following inoculation in all piglets. Four piglets developed a brief viremia for 2 days post-inoculation and three of these had detectable virus in oronasal secretions three days post-inoculation. Primary inoculated piglets seroconverted and those that developed detectable viremias had the highest titers assessed by serum neutralization (1:64-1:256). Two viremic piglets had a lymphoplasmacytic encephalitis with glial nodules; RVFV was not detected by immunohistochemistry in these sections. While young piglets do not appear to readily develop clinical disease following RVFV infection, results suggest swine could be subclinically infected with RVFV.


Subject(s)
Rift Valley Fever/immunology , Rift Valley fever virus/immunology , Swine Diseases/virology , Animals , Brain/pathology , Brain/virology , Disease Susceptibility , Female , Immunohistochemistry , Liver/pathology , Liver/virology , Lymph Nodes/pathology , Lymph Nodes/virology , Male , RNA, Viral/blood , RNA, Viral/genetics , RNA, Viral/isolation & purification , Rift Valley Fever/blood , Rift Valley Fever/transmission , Rift Valley Fever/virology , Rift Valley fever virus/isolation & purification , Rift Valley fever virus/pathogenicity , Spleen/pathology , Spleen/virology , Sus scrofa , Swine , Swine Diseases/blood , Swine Diseases/immunology , Swine Diseases/transmission , Viremia/blood , Viremia/immunology , Viremia/virology
19.
Viruses ; 13(10)2021 09 28.
Article in English | MEDLINE | ID: mdl-34696373

ABSTRACT

The recently discovered porcine circovirus 3 (PCV3) belongs to the Circovirus genus of the Circoviridae family together with the other three PCVs, PCV1, PCV2, and PCV4. As reported, PCV3 can infect pig, wild boar, and several other intermediate hosts, resulting in single or multiple infections in the affected animal. The PCV3 infection can lead to respiratory diseases, digestive disorders, reproductive disorders, multisystemic inflammation, and immune responses. Up to now, PCV3 infection, as well as the disease caused by PCV3, has been reported in many swine farms worldwide with high positive rates, which indicates that the virus may be another important pathogen in the swine industry. Therefore, we reviewed the current progress on epidemiology and pathobiology of PCV3, which may provide the latest knowledge of the virus and PCV3-related diseases.


Subject(s)
Circoviridae Infections/pathology , Circoviridae Infections/veterinary , Circovirus/pathogenicity , Swine Diseases/pathology , Animals , Circoviridae Infections/transmission , Circovirus/classification , Phylogeny , Swine , Swine Diseases/transmission , Swine Diseases/virology
20.
Virology ; 563: 88-97, 2021 11.
Article in English | MEDLINE | ID: mdl-34500147

ABSTRACT

Two experimental challenge studies were conducted to evaluate the pathogenesis of a porcine parainfluenza virus type 1 (PPIV-1) isolate. Four-week-old conventional (CON) pigs were challenged in Study 1 and six-week-old caesarean derived/colostrum deprived (CDCD) pigs were challenged in Study 2. Results indicate that PPIV-1 shedding and replication occur in the upper and lower respiratory tracts of CON and CDCD pigs as detected by RT-qPCR and immunohistochemistry. Mild macroscopic lung lesions were observed in CON pigs but not in CDCD pigs. Microscopic lung lesions were mild and consisted of peribronchiolar lymphocytic cuffing and epithelial proliferation in CON and CDCD pigs. Serum neutralizing antibodies were detected in the CON and CDCD pigs by 14 and 7 days post inoculation, respectively. This study provides evidence that in spite of PPIV-1 infection and replication in challenged swine, significant clinical respiratory disease was not observed.


Subject(s)
Cesarean Section , Colostrum/immunology , Paramyxoviridae Infections/veterinary , Paramyxoviridae/classification , Swine Diseases/virology , Animals , Antibodies, Neutralizing , Antibodies, Viral , Lung Diseases/veterinary , Lung Diseases/virology , Paramyxoviridae Infections/transmission , Paramyxoviridae Infections/virology , Swine , Swine Diseases/immunology , Swine Diseases/transmission , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...