Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Arch Virol ; 168(10): 263, 2023 Sep 29.
Article in English | MEDLINE | ID: mdl-37775596

ABSTRACT

Since 2006, highly pathogenic avian influenza (HPAI) subtypes H5Nx have adversely affected poultry production in Nigeria. Successive waves of infections in the last two decades have raised concerns about the ability to contain infections by biosecurity alone, and evidence of recurrent outbreaks suggests a need for adoption of additional control measures such as vaccination. Although vaccination can be used to control virus spread and reduce the morbidity and mortality caused by HPAI, no country using vaccination alone as a control measure against HPAI has been able to eliminate or prevent re-infection. To inform policy in Nigeria, we examined the intricacies of HPAI vaccination, government regulations, and scientific data regarding what kind of vaccines can be used based on subtype, whether inactivated or live attenuated should be used, when to deliver vaccine either proactively or reactively, where to apply vaccination either in disease control zones, regionally, or nationally, and how to vaccinate the targeted poultry population for optimum success. A resurgence of HPAI outbreaks in Nigeria since 2018, after the country was declared free of the epidemic following the first outbreak in 2006, has led to enhanced intervention. Controlled vaccination entails monitoring the application of vaccines, the capacity to differentiate vaccinated from infected (DIVA) flocks, and assessing seroconversion or other immune correlates of protection. Concurrent surveillance for circulating avian influenza virus (AIV) and analyzing AIV isolates obtained via surveillance efforts for genetic and/or antigenic mismatch with vaccine strains are also important. Countries with high investment in commercial poultry farms like Nigeria may identify and zone territories where vaccines can be applied. This may include ring vaccination to control HPAI in areas or production systems at risk of infection. Before adoption of vaccination as an additional control measure on commercial poultry farms, two outcomes must be considered. First, vaccination is an admission of endemicity. Secondly, vaccinated flocks may no longer be made accessible to international poultry markets in accordance with WOAH trade regulations. Vaccination must therefore be approached with utmost caution and be guided by science-based evidence throughout the implementation strategy after thorough risk assessment. Influenza vaccine research, development, and controlled application in addition to biosecurity may be a precautionary measure in the evolving HPAI scenario in Nigeria.


Subject(s)
Influenza A virus , Influenza Vaccines , Influenza in Birds , Animals , Humans , Influenza in Birds/epidemiology , Influenza in Birds/prevention & control , Nigeria/epidemiology , Disease Outbreaks/prevention & control , Disease Outbreaks/veterinary , Poultry , Vaccination/veterinary
2.
BMC Vet Res ; 18(1): 393, 2022 Nov 09.
Article in English | MEDLINE | ID: mdl-36348373

ABSTRACT

BACKGROUND: Influenza A virus (IAV) is an important respiratory pathogen of pigs that affects pig health, well-being and productivity, has zoonotic potential, and has significant economic impact for producers. The ultimate goal is to maintain herds free from IAV. Due to the probability of IAV introduction into the herds, it is also desirable for herds to have some immunity to the virus. In this study, we evaluated a protocol that combined sow vaccination with the implementation of internal biosecurity practices during the pre-weaning period with the goal to wean IAV negative pigs. Five IAV positive breeding herds were vaccinated twice, 3 weeks apart with a herd-specific autogenous vaccine. For the subsequent 8 weeks, a biosecurity protocol was maintained, consisting of no pig movements after 3 days of age, no use of nurse sows, workers changing disposable gloves between litters, workers not stepping into farrowing crates, and daily disinfection of tools and materials used to handle pigs. RESULTS: Following these interventions, four of the five treatment farms had significant reductions in IAV detection (p value < 0.05). Three of the farms tested negative at all sampling points post-intervention and one farm had a 21% reduction in IAV positivity. CONCLUSIONS: This study indicates that a protocol that combines sow vaccination and enhanced biosecurity practices may limit IAV transmission among piglets and enable the weaning of groups of pigs free from the virus.


Subject(s)
Influenza A virus , Orthomyxoviridae Infections , Swine Diseases , Animals , Swine , Female , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/veterinary , Farms , Weaning , Biosecurity , Vaccination/veterinary
3.
BMC Vet Res ; 18(1): 84, 2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35236347

ABSTRACT

BACKGROUND: African swine fever (ASF) is a highly contagious and devastating pig disease that has caused extensive global economic losses. Understanding ASF virus (ASFV) transmission dynamics within a herd is necessary in order to prepare for and respond to an outbreak in the United States. Although the transmission parameters for the highly virulent ASF strains have been estimated in several articles, there are relatively few studies focused on moderately virulent strains. Using an approximate Bayesian computation algorithm in conjunction with Monte Carlo simulation, we have estimated the adequate contact rate for moderately virulent ASFV strains and determined the statistical distributions for the durations of mild and severe clinical signs using individual, pig-level data. A discrete individual based disease transmission model was then used to estimate the time to detect ASF infection based on increased mild clinical signs, severe clinical signs, or daily mortality. RESULTS: Our results indicate that it may take two weeks or longer to detect ASF in a finisher swine herd via mild clinical signs or increased mortality beyond levels expected in routine production. A key factor contributing to the extended time to detect ASF in a herd is the fairly long latently infected period for an individual pig (mean 4.5, 95% P.I., 2.4 - 7.2 days). CONCLUSION: These transmission model parameter estimates and estimated time to detection via clinical signs provide valuable information that can be used not only to support emergency preparedness but also to inform other simulation models of evaluating regional disease spread.


Subject(s)
African Swine Fever Virus , African Swine Fever , Swine Diseases , African Swine Fever/diagnosis , African Swine Fever/epidemiology , Animals , Bayes Theorem , Disease Outbreaks/veterinary , Swine , Swine Diseases/diagnosis , Swine Diseases/epidemiology
4.
Vet Res ; 51(1): 42, 2020 Mar 14.
Article in English | MEDLINE | ID: mdl-32169091

ABSTRACT

The mechanisms of transmission of influenza A virus (IAV) and porcine reproductive and respiratory syndrome virus (PRRSV) in pigs during the pre-weaning period are not fully elucidated. Since viable IAV and PRRSV can be found on the udder skin of lactating sows and the use of nurse sows is a common management practice, we developed a novel nurse sow model to evaluate the transmission of IAV and PRRSV from lactating sows to their adopted piglets. In two studies, we infected pigs with either IAV or PRRSV who then contaminated the udder skin of lactating dams with their nasal and oral secretions while suckling. Once the skin was confirmed virus positive for IAV and PRRSV, the sows were moved to separate empty clean rooms to adopt IAV and PRRSV negative suckling piglets. After adoption, 1 out of eight (12.5%) piglets tested IAV positive 1-day post-adoption (dpa) and the entire litter (8 out of 8) became positive by 4 dpa. In the case of PRRSV, 3 out of 11 (27.3%) pigs tested rRT-PCR positive 2 dpa and there were 7 out of 11 (63.6%) pigs positive at the termination of the study at 7 dpa. This study documented the transmission of IAV and PRRSV between litters of piglets by nurse sows and highlights the importance of the nurse sow-piglet as a unit that contributes to the maintenance of endemic infections in breeding herds. The use of nurse sows in pig farms, though beneficial for minimizing pre-weaning mortality and maximizing farm productivity, is seemingly detrimental as this practice may facilitate the transmission of IAV and PRRSV to piglets prior to weaning.


Subject(s)
Influenza A virus/physiology , Orthomyxoviridae Infections/veterinary , Porcine Reproductive and Respiratory Syndrome/transmission , Porcine respiratory and reproductive syndrome virus/physiology , Swine Diseases/transmission , Animals , Disease Models, Animal , Female , Orthomyxoviridae Infections/transmission , Proof of Concept Study , Swine
5.
Vet Res ; 51(1): 89, 2020 Jul 09.
Article in English | MEDLINE | ID: mdl-32646490

ABSTRACT

Influenza A viruses evolve rapidly to escape host immunity. In swine, this viral evolution has resulted in the emergence of multiple H1 and H3 influenza A virus (IAV) lineages in the United States (US) pig populations. The heterologous prime-boost vaccination strategy is a promising way to deal with diverse IAV infection in multiple animal models. However, whether or not this vaccination strategy is applicable to US swine to impart immunity against infection from North American strains of IAV is still unknown. We performed a vaccination-challenge study to evaluate the protective efficacy of using multivalent inactivated vaccine and/or a live attenuated IAV vaccine (LAIV) in pigs following multiple prime-boost vaccination protocols against a simultaneous H1N1 and H3N2 IAV infection. Our data show that pigs in the heterologous prime-boost vaccination group had more favorable outcomes consistent with a better response against virus challenge than non-vaccinated pigs. Additionally, delivering a multivalent heterologous inactivated vaccine boost to pigs following a single LAIV administration was also beneficial. We concluded the heterologous prime boost vaccination strategy may potentiate responses to suboptimal immunogens and holds the potential applicability to control IAV in the North American swine industry. However, more studies are needed to validate the application of this vaccination approach under field conditions.


Subject(s)
Communicable Disease Control/methods , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/veterinary , Swine Diseases/prevention & control , Vaccination/veterinary , Animals , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Sus scrofa , Swine , Swine Diseases/virology , Vaccines, Attenuated/immunology
6.
Emerg Infect Dis ; 25(4): 691-700, 2019 04.
Article in English | MEDLINE | ID: mdl-30730827

ABSTRACT

The genetic diversity of influenza A viruses circulating in swine in Mexico complicates control efforts in animals and presents a threat to humans, as shown by influenza A(H1N1)pdm09 virus. To describe evolution of swine influenza A viruses in Mexico and evaluate strains for vaccine development, we sequenced the genomes of 59 viruses and performed antigenic cartography on strains from 5 regions. We found that genetic and antigenic diversity were particularly high in southeast Mexico because of repeated introductions of viruses from humans and swine in other regions in Mexico. We identified novel reassortant H3N2 viruses with genome segments derived from 2 different viruses that were independently introduced from humans into swine: pandemic H1N1 viruses and seasonal H3N2 viruses. The Mexico swine viruses are antigenically distinct from US swine lineages. Protection against these viruses is unlikely to be afforded by US virus vaccines and would require development of new vaccines specifically targeting these diverse strains.


Subject(s)
Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza, Human/virology , Orthomyxoviridae Infections/virology , Reassortant Viruses/genetics , Animals , Antigens, Viral/immunology , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Mexico , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/prevention & control , Swine
7.
BMC Vet Res ; 15(1): 61, 2019 Feb 14.
Article in English | MEDLINE | ID: mdl-30764815

ABSTRACT

BACKGROUND: Influenza A virus (IAV) is an important pathogen in pigs that affects productivity and has important public health implications because of its zoonotic nature. Surveillance is central to the control of influenza, however, detection of IAV infections can be challenging in endemically infected herds with low prevalence of infection. METHODS: In groups of suckling (18-21 days of age) and growing (35-45 days of age) pigs, we compared various sampling approaches to detect, isolate and sequence IAV using individual (nasal swabs, nasal wipes and oropharyngeal swabs), group (oral fluids, surface wipes and sow udder skin wipes) and environmental (airborne particles deposited on surfaces and air samples) sampling approaches. All samples were tested by IAV rRT-PCR and a subset was used for virus isolation and direct sequencing. RESULTS: In general, environmental and group samples resulted in higher odd ratios (range = 3.87-16.5, p-value < 0.05) of detecting a positive sample by rRT-PCR compared to individual pooled samples, except for oropharyngeal swabs (OR = 8.07, p-value < 0.05). In contrast, individual samples were most likely to yield a viral isolate by cell culture. Oropharyngeal swabs in suckling pigs (78.4%), and nasal swabs (47.6%) or nasal wipes (45%) in growing pigs, and udder wipes in lactating sows (75%) were the preferred samples to obtain an isolate. CONCLUSIONS: Our findings indicate that group and environmental sampling strategies should be considered in influenza surveillance programs in particular if the goal is just to detect infection. This study provides new information on sampling approaches to conduct effective influenza surveillance in pigs and identifies udder wipes from lactating sows as a novel sample type that offers a convenient, cheap and sensitive manner to monitor IAV in litters prior to weaning.


Subject(s)
Orthomyxoviridae Infections/veterinary , Swine Diseases/epidemiology , Animals , Animals, Suckling/virology , Environmental Microbiology , Influenza A virus/genetics , Orthomyxoviridae Infections/epidemiology , Population Surveillance/methods , Real-Time Polymerase Chain Reaction/veterinary , Sampling Studies , Sequence Analysis, DNA/veterinary , Swine , Swine Diseases/virology
8.
BMC Vet Res ; 15(1): 147, 2019 May 14.
Article in English | MEDLINE | ID: mdl-31088548

ABSTRACT

BACKGROUND: Avian influenza (AI) is an infectious viral disease that affects several species and has zoonotic potential. Due to its associated health and economic repercussions, minimizing AI outbreaks is important. However, most control measures are generic and mostly target pathways important for the conventional poultry farms producing chickens, turkeys, and eggs and may not target other pathways that may be specific to the upland game bird sector. The goal of this study is to provide evidence to support the development of novel strategies for sector-specific AI control by comparing and contrasting practices and potential pathways for spread in upland game bird farms with those for conventional poultry farms in the United States. Farm practices and processes, seasonality of activities, geographic location and inter-farm distance were analyzed across the sectors. All the identified differences were framed and discussed in the context of their associated pathways for virus introduction into the farm and subsequent between-farm spread. RESULTS: Differences stemming from production systems and seasonality, inter-farm distance and farm densities were evident and these could influence both fomite-mediated and local-area spread risks. Upland game bird farms operate under a single, independent owner rather than being contracted with or owned by a company with other farms as is the case with conventional poultry. The seasonal marketing of upland game birds, largely driven by hunting seasons, implies that movements are seasonal and customer-vendor dynamics vary between industry groups. Farm location analysis revealed that, on average, an upland game bird premises was 15.42 km away from the nearest neighboring premises with birds compared to 3.74 km for turkey premises. Compared to turkey premises, the average poultry farm density in a radius of 10 km of an upland game bird premises was less than a half, and turkey premises were 3.8 times (43.5% compared with 11.5%) more likely to fall within a control area during the 2015 Minnesota outbreak. CONCLUSIONS: We conclude that the existing differences in the seasonality of production, isolated geographic location and epidemiological seclusion of farms influence AI spread dynamics and therefore disease control measures should be informed by these and other factors to achieve success.


Subject(s)
Animal Husbandry/methods , Galliformes , Influenza A virus , Influenza in Birds/epidemiology , Animals , Disease Outbreaks , Geography , Influenza in Birds/prevention & control , Influenza in Birds/transmission , Seasons , United States
9.
J Virol ; 91(18)2017 09 15.
Article in English | MEDLINE | ID: mdl-28659482

ABSTRACT

Influenza A viruses (IAVs) are endemic in swine and represent a public health risk. However, there is limited information on the genetic diversity of swine IAVs within farrow-to-wean farms, which is where most pigs are born. In this longitudinal study, we sampled 5 farrow-to-wean farms for a year and collected 4,190 individual nasal swabs from three distinct pig subpopulations. Of these, 207 (4.9%) samples tested PCR positive for IAV, and 124 IAVs were isolated. We sequenced the complete genomes of 123 IAV isolates and found 31 H1N1, 26 H1N2, 63 H3N2, and 3 mixed IAVs. Based on the IAV hemagglutinin, seven different influenza A viral groups (VGs) were identified. Most of the remaining IAV gene segments allowed us to differentiate the same VGs, although an additional viral group was identified for gene segment 3 (PA). Moreover, the codetection of more than one IAV VG was documented at different levels (farm, subpopulation, and individual pigs), highlighting the environment for potential IAV reassortment. Additionally, 3 out of 5 farms contained IAV isolates (n = 5) with gene segments from more than one VG, and 79% of all the IAVs sequenced contained a signature mutation (S31N) in the matrix gene that has been associated with resistance to the antiviral amantadine. Within farms, some IAVs were detected only once, while others were detected for 283 days. Our results illustrate the maintenance and subsidence of different IAVs within swine farrow-to-wean farms over time, demonstrating that pig subpopulation dynamics are important to better understand the diversity and epidemiology of swine IAVs.IMPORTANCE On a global scale, swine are one of the main reservoir species for influenza A viruses (IAVs) and play a key role in the transmission of IAVs between species. Additionally, the 2009 IAV pandemics highlighted the role of pigs in the emergence of IAVs with pandemic potential. However, limited information is available regarding the diversity and distribution of swine IAVs on farrow-to-wean farms, where novel IAVs can emerge. In this study, we studied 5 swine farrow-to-wean farms for a year and characterized the genetic diversity of IAVs among three different pig subpopulations commonly housed on this type of farm. Using next-generation-sequencing technologies, we demonstrated the complex distribution and diversity of IAVs among the pig subpopulations studied. Our results demonstrated the dynamic evolution of IAVs within farrow-to-wean farms, which is crucial to improve health interventions to reduce the risk of transmission between pigs and from pigs to people.


Subject(s)
Genetic Variation , Influenza A virus/classification , Influenza A virus/isolation & purification , Orthomyxoviridae Infections/veterinary , Sequence Analysis, DNA , Swine Diseases/epidemiology , Swine Diseases/virology , Animals , Farms , Genotype , Influenza A virus/genetics , Longitudinal Studies , Molecular Epidemiology , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/virology , Swine
10.
J Gen Virol ; 98(11): 2663-2675, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29058649

ABSTRACT

Swine are a key reservoir host for influenza A viruses (IAVs), with the potential to cause global pandemics in humans. Gaps in surveillance in many of the world's largest swine populations impede our understanding of how novel viruses emerge and expand their spatial range in pigs. Although US swine are intensively sampled, little is known about IAV diversity in Canada's population of ~12 million pigs. By sequencing 168 viruses from multiple regions of Canada, our study reveals that IAV diversity has been underestimated in Canadian pigs for many years. Critically, a new H1 clade has emerged in Canada (H1α-3), with a two-amino acid deletion at H1 positions 146-147, that experienced rapid growth in Manitoba's swine herds during 2014-2015. H1α-3 viruses also exhibit a higher capacity to invade US swine herds, resulting in multiple recent introductions of the virus into the US Heartland following large-scale movements of pigs in this direction. From the Heartland, H1α-3 viruses have disseminated onward to both the east and west coasts of the United States, and may become established in Appalachia. These findings demonstrate how long-distance trading of live pigs facilitates the spread of IAVs, increasing viral genetic diversity and complicating pathogen control. The proliferation of novel H1α-3 viruses also highlights the need for expanded surveillance in a Canadian swine population that has long been overlooked, and may have implications for vaccine design.


Subject(s)
Evolution, Molecular , Influenza A virus/classification , Influenza A virus/isolation & purification , Orthomyxoviridae Infections/veterinary , Swine Diseases/epidemiology , Swine Diseases/virology , Animals , Canada/epidemiology , Influenza A virus/genetics , Molecular Epidemiology , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/virology , Swine , United States/epidemiology
11.
Proc Natl Acad Sci U S A ; 111(22): E2241-50, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24843157

ABSTRACT

Influenza viruses bind to host cell surface glycans containing terminal sialic acids, but as studies on influenza binding become more sophisticated, it is becoming evident that although sialic acid may be necessary, it is not sufficient for productive binding. To better define endogenous glycans that serve as viral receptors, we have explored glycan recognition in the pig lung, because influenza is broadly disseminated in swine, and swine have been postulated as an intermediary host for the emergence of pandemic strains. For these studies, we used the technology of "shotgun glycomics" to identify natural receptor glycans. The total released N- and O-glycans from pig lung glycoproteins and glycolipid-derived glycans were fluorescently tagged and separated by multidimensional HPLC, and individual glycans were covalently printed to generate pig lung shotgun glycan microarrays. All viruses tested interacted with one or more sialylated N-glycans but not O-glycans or glycolipid-derived glycans, and each virus demonstrated novel and unexpected differences in endogenous N-glycan recognition. The results illustrate the repertoire of specific, endogenous N-glycans of pig lung glycoproteins for virus recognition and offer a new direction for studying endogenous glycan functions in viral pathogenesis.


Subject(s)
Glycomics/methods , Influenza in Birds/metabolism , Influenza, Human/metabolism , Lung/virology , Orthomyxoviridae/metabolism , Receptors, Virus/metabolism , Agglutination Tests , Animals , Birds , Chickens , Erythrocytes/virology , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H1N2 Subtype/isolation & purification , Influenza A Virus, H1N2 Subtype/metabolism , Influenza A Virus, H1N2 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza A Virus, H3N2 Subtype/metabolism , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza in Birds/virology , Influenza, Human/virology , Lectins/metabolism , Lung/metabolism , Orthomyxoviridae/isolation & purification , Orthomyxoviridae/pathogenicity , Polysaccharides/metabolism , Species Specificity , Swine , Virulence
12.
J Gen Virol ; 96(10): 2982-2993, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26251306

ABSTRACT

To gain insight into the evolution of influenza A viruses (IAVs) during infection of vaccinated pigs, we experimentally infected a 3-week-old naive pig with a triple-reassortant H1N1 IAV and placed the seeder pig in direct contact with a group of age-matched vaccinated pigs (n = 10). We indexed the genetic diversity and evolution of the virus at an intra-host level by deep sequencing the entire genome directly from nasal swabs collected at two separate samplings during infection. We obtained 13 IAV metagenomes from 13 samples, which included the virus inoculum and two samples from each of the six pigs that tested positive for IAV during the study. The infection produced a population of heterogeneous alleles (sequence variants) that was dynamic over time. Overall, 794 polymorphisms were identified amongst all samples, which yielded 327 alleles, 214 of which were unique sequences. A total of 43 distinct haemagglutinin proteins were translated, two of which were observed in multiple pigs, whereas the neuraminidase (NA) was conserved and only one dominant NA was found throughout the study. The genetic diversity of IAVs changed dynamically within and between pigs. However, most of the substitutions observed in the internal gene segments were synonymous. Our results demonstrated remarkable IAV diversity, and the complex, rapid and dynamic evolution of IAV during infection of vaccinated pigs that can only be appreciated with repeated sampling of individual animals and deep sequence analysis.


Subject(s)
Evolution, Molecular , Genetic Variation , Genome, Viral , Influenza A Virus, H1N1 Subtype/genetics , Orthomyxoviridae Infections/virology , Reassortant Viruses/genetics , Alleles , Animals , High-Throughput Nucleotide Sequencing , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/isolation & purification , Metagenome , Molecular Sequence Data , Nasal Mucosa/immunology , Nasal Mucosa/virology , Orthomyxoviridae Infections/immunology , RNA, Viral/genetics , Reassortant Viruses/growth & development , Reassortant Viruses/immunology , Reassortant Viruses/isolation & purification , Sequence Analysis, DNA , Swine , Viral Proteins/genetics
13.
J Virol ; 88(17): 10110-9, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24965467

ABSTRACT

UNLABELLED: The capacity of influenza A viruses to cross species barriers presents a continual threat to human and animal health. Knowledge of the human-swine interface is particularly important for understanding how viruses with pandemic potential evolve in swine hosts. We sequenced the genomes of 141 influenza viruses collected from North American swine during 2002 to 2011 and identified a swine virus that possessed all eight genome segments of human seasonal A/H3N2 virus origin. A molecular clock analysis indicates that this virus--A/sw/Saskatchewan/02903/2009(H3N2)--has likely circulated undetected in swine for at least 7 years. For historical context, we performed a comprehensive phylogenetic analysis of an additional 1,404 whole-genome sequences from swine influenza A viruses collected globally during 1931 to 2013. Human-to-swine transmission occurred frequently over this time period, with 20 discrete introductions of human seasonal influenza A viruses showing sustained onward transmission in swine for at least 1 year since 1965. Notably, human-origin hemagglutinin (H1 and H3) and neuraminidase (particularly N2) segments were detected in swine at a much higher rate than the six internal gene segments, suggesting an association between the acquisition of swine-origin internal genes via reassortment and the adaptation of human influenza viruses to new swine hosts. Further understanding of the fitness constraints on the adaptation of human viruses to swine, and vice versa, at a genomic level is central to understanding the complex multihost ecology of influenza and the disease threats that swine and humans pose to each other. IMPORTANCE: The swine origin of the 2009 A/H1N1 pandemic virus underscored the importance of understanding how influenza A virus evolves in these animals hosts. While the importance of reassortment in generating genetically diverse influenza viruses in swine is well documented, the role of human-to-swine transmission has not been as intensively studied. Through a large-scale sequencing effort, we identified a novel influenza virus of wholly human origin that has been circulating undetected in swine for at least 7 years. In addition, we demonstrate that human-to-swine transmission has occurred frequently on a global scale over the past decades but that there is little persistence of human virus internal gene segments in swine.


Subject(s)
Evolution, Molecular , Influenza A Virus, H3N2 Subtype/classification , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza, Human/transmission , Influenza, Human/virology , Orthomyxoviridae Infections/veterinary , Swine Diseases/virology , Animals , Genome, Viral , Humans , Influenza A Virus, H3N2 Subtype/genetics , Molecular Epidemiology , Orthomyxoviridae Infections/virology , RNA, Viral/genetics , Sequence Analysis, DNA , Swine
14.
Emerg Infect Dis ; 20(7): 1195-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24960190

ABSTRACT

We investigated the presence in US pigs of rotavirus H (RVH), identified in pigs in Japan and Brazil. From 204 samples collected during 2006-2009, we identified RVH in 15% of fecal samples from 10 US states, suggesting that RVH has circulated in the United States since 2002, but probably longer.


Subject(s)
Rotavirus Infections/virology , Rotavirus/genetics , Swine Diseases/virology , Swine/virology , Animals , Feces/virology , Japan , Phylogeny , Sequence Analysis, DNA/methods , United States
15.
Emerg Infect Dis ; 20(10): 1620-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25279722

ABSTRACT

Porcine epidemic diarrhea virus (PEDV), which emerged in the United States in 2013, has spread throughout North America. Limited availability of PEDV complete genomes worldwide has impeded our understanding of PEDV introduction into the United States. To determine the relationship between the North American strains and global emerging and historic PEDV strains, we sequenced and analyzed complete genomes of 74 strains from North America; the strains clustered into 2 distinct clades. Compared with the initially reported virulent US PEDV strains, 7 (9.7%) strains from 4 states contained insertions and deletions in the spike gene (S INDELs). These S INDEL strains share 99.8%-100% nt identity with each other and 96.2%-96.7% nt identity with the initial US strains. Furthermore, the S INDEL strains form a distinct cluster within North American clade II, sharing 98.6%-100% nt identity overall. In the United States, the S INDEL and original PEDV strains are co-circulating and could have been introduced simultaneously.


Subject(s)
Biological Evolution , Coronavirus Infections/veterinary , Porcine epidemic diarrhea virus/classification , Porcine epidemic diarrhea virus/isolation & purification , Swine Diseases/virology , Animals , Coronavirus Infections/epidemiology , Coronavirus Infections/virology , Genetic Variation , Genome, Viral , North America/epidemiology , Open Reading Frames/genetics , Porcine epidemic diarrhea virus/genetics , Reassortant Viruses , Swine , Swine Diseases/epidemiology , Time Factors , Viral Proteins/genetics , Viral Proteins/metabolism
16.
Avian Dis ; 67(4): 402-409, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38300659

ABSTRACT

The complexity of influenza A virus (IAV) infections in avian hosts leads to equally complex scenarios for the vaccination of poultry. Vaccination against avian influenza strains can be used to prevent infections from sources with a single strain of IAV. It has been used as a part of outbreak control strategies as well as a way to maintain production for both low and high pathogenicity outbreaks. Unlike other viral pathogens of birds, avian influenza vaccination when used against highly pathogenic avian influenza virus, is tied to international trade and thus is not freely available for use without specific permission.


Vacunación de aves comerciales contra la influenza aviar. La complejidad de las infecciones por el virus de la influenza A en las aves hospedadoras conduce a escenarios igualmente complejos para la vacunación en la avicultura. La vacunación contra cepas de influenza aviar se puede utilizar para prevenir infecciones provenientes de fuentes con una sola cepa del virus de influenza. Se ha utilizado como parte de las estrategias de control de brotes, así como como una forma de mantener la producción tanto en brotes de baja como de alta patogenicidad. A diferencia de otros patógenos virales de las aves, la vacunación contra la influenza aviar, cuando se usa contra el virus de la influenza aviar altamente patógeno, está vinculada al comercio internacional y por lo tanto, no está disponible para su uso sin un permiso específico.


Subject(s)
Influenza A virus , Influenza in Birds , Influenza, Human , Poultry Diseases , Animals , Humans , Poultry , Influenza in Birds/prevention & control , Commerce , Internationality , Poultry Diseases/prevention & control , Vaccination/veterinary
17.
Emerg Infect Dis ; 19(6): 954-60, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23735740

ABSTRACT

Veterinary diagnostic laboratories identify and characterize influenza A viruses primarily through passive surveillance. However, additional surveillance programs are needed. To meet this need, an active surveillance program was conducted at pig farms throughout the midwestern United States. From June 2009 through December 2011, nasal swab samples were collected monthly from among 540 groups of growing pigs and tested for influenza A virus by real-time reverse transcription PCR. Of 16,170 samples, 746 were positive for influenza A virus; of these, 18.0% were subtype H1N1, 16.0% H1N2, 7.6% H3N2, and 14.5% (H1N1)pdm09. An influenza (H3N2) and (H1N1)pdm09 virus were identified simultaneously in 8 groups. This active influenza A virus surveillance program provided quality data and increased the understanding of the current situation of circulating viruses in the midwestern US pig population.


Subject(s)
Influenza A virus , Orthomyxoviridae Infections/veterinary , Swine Diseases/epidemiology , Agriculture , Animals , History, 21st Century , Influenza A virus/classification , Influenza A virus/genetics , Midwestern United States/epidemiology , Public Health Surveillance , Seasons , Swine , Swine Diseases/history
18.
J Gen Virol ; 94(Pt 6): 1236-41, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23695819

ABSTRACT

To understand the evolution of swine-origin H3N2v influenza viruses that have infected 320 humans in the USA since August 2011, we performed a phylogenetic analysis at a whole genome scale of North American swine influenza viruses (n = 200). All viral isolates evolved from the prototypical North American H3 cluster 4 (c4), with evidence for further diversification into subclusters. At least ten distinct reassorted H3N2/pandemic H1N1 (rH3N2p) genotypes were identified in swine. Genotype 1 (G1) was most frequently detected in swine and all human H3N2v viruses clustered within a single G1 clade. These data suggest that the genetic requirements for transmission to humans may be restricted to a specific subset of swine viruses. Mutations at putative antigenic sites as well as reduced serological cross-reactivity among the H3 subclusters suggest antigenic drift of these contemporary viruses.


Subject(s)
Influenza A Virus, H3N2 Subtype/genetics , Influenza, Human/virology , Orthomyxoviridae Infections/veterinary , Reassortant Viruses/genetics , Swine Diseases/virology , Animals , Cross Reactions , Genotype , Humans , Influenza A Virus, H3N2 Subtype/classification , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza, Human/immunology , Molecular Sequence Data , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Phylogeny , Reassortant Viruses/classification , Reassortant Viruses/immunology , Reassortant Viruses/isolation & purification , Swine , Swine Diseases/immunology , United States
19.
Porcine Health Manag ; 9(1): 13, 2023 May 15.
Article in English | MEDLINE | ID: mdl-37183258

ABSTRACT

Indirect transmission of influenza A virus (IAV) contributes to virus spread in pigs. To identify farm management activities with the ability to contaminate farmworkers' hands and clothing that then could be a source of virus spread to other pigs, we conducted a within-farm, prospective IAV surveillance study. Hands and clothes from farmworkers performing the activities of piglet processing, vaccination, or weaning were sampled before and after the activities were performed. Samples were tested by IAV rRT-PCR and virus viability was assessed by cell culture. A multivariate generalized linear model was used to detect associations of the activities with IAV contamination. Of the samples collected for IAV rRT-PCR testing, there were 16% (12/76) collected immediately after processing, 96% (45/48) collected after vaccination, and 94% (29/31) collected after weaning that tested positive. Samples collected immediately after vaccination and weaning, i.e., activities that took place during the peri-weaning period when pigs were about 3 weeks of age, had almost 6 times higher risk of IAV detection and had more samples IAV positive (p-value < 0.0001) than samples collected after processing, i.e., an activity that took place in the first few days of life. Both, hands and clothes had similar contamination rates (46% and 55% respectively, p-value = 0.42) and viable virus was isolated from both. Our results indicate that activities that involve the handling of infected piglets close to weaning age represent a significant risk for IAV dissemination due to the high level of IAV contamination found in farmworkers' hands and coveralls involved in the activities. Biosecurity protocols that include hand sanitation and changing clothing after performing activities with a high-risk of influenza contamination should be recommended to farmworkers to control and limit the mechanical spread of IAV between pigs.

20.
Front Vet Sci ; 10: 1245278, 2023.
Article in English | MEDLINE | ID: mdl-37799404

ABSTRACT

Influenza A virus poses a significant threat to public health and the swine industry. Vaccination is the primary measure for controlling the disease, but the effectiveness of vaccines can vary depending on the antigenic match between vaccine strains and circulating strains. In Chile, H1N1pdm09 and other lineages H1N2 and H3N2 have been detected in pigs, which are genetically distinct from the strains included in commercial vaccines. This study aimed to evaluate the cross-protection by commercial vaccines against strains circulating in Chile using the guinea pig model. For this study, four circulating strains [A/swine/Chile/H1A-7/2014(H1N2), A/swine/Chile/H1B-2/2014(H1N2), A/swine/Chile/H1P-12/2015(H1N1), and A/swine/Chile/H3-2/2015(H3N2)] were selected. Guinea pigs were divided into vaccinated and control groups. The vaccinated animals received either a multivalent antigenically heterologous or monovalent homologous vaccine, while the control animals remained unvaccinated. Following vaccination, all animals were intranasally challenged, and nasal wash samples were collected at different time points post-infection. The results showed that the homologous monovalent vaccine-induced hemagglutinin-specific antibodies against the Chilean pandemic H1N1pdm09 strain. However, the commercial heterologous multivalent vaccine failed to induce hemagglutinin-specific antibody titers against the H1N2 and H3N2 challenge strains. Furthermore, the homologous monovalent vaccine significantly reduced the duration of viral shedding and viral titers specifically against the Chilean pandemic H1N1pdm09 strain and heterologous multivalent vaccine only partial. These findings highlight the importance of regularly updating vaccine strains to match the circulating field strains for effective control of swine influenza. Further research is needed to develop vaccines that confer broader protection against diverse strains of swine influenza A virus.

SELECTION OF CITATIONS
SEARCH DETAIL