Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Article in German | MEDLINE | ID: mdl-33902142

ABSTRACT

The porcine respiratory disease complex describes a clinical condition that often manifests as treatment-resistant respiratory disease of growing to finishing pigs. Its multifactorial etiology includes infectious and non-infectious factors. Besides management and hygiene conditions, particularly viral and bacterial pathogens contribute to the development and course of PRDC. The porcine reproductive and respiratory syndrome virus (PRRSV), porcine circovirus type 2 (PCV2), influenza A virus (IAV) and Mycoplasma (M.) hyopneunoniae are considered as the major pathogens involved in PRDC. The clinical outcome and necropsy findings may differ depending on the involvement of the different pathogens. The complex nature of the PRDC impedes the diagnostic and preventive measures on affected farms. The present review provides insight into the pathomorphology, pathogenesis and inter-pathogen-interactions and aims to support practitioners in implementing purposeful diagnostic and preventive measures.


Subject(s)
Circoviridae Infections , Circovirus , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Respiratory Tract Diseases , Swine Diseases , Animals , Circoviridae Infections/diagnosis , Circoviridae Infections/veterinary , Farms , Porcine Reproductive and Respiratory Syndrome/diagnosis , Porcine Reproductive and Respiratory Syndrome/therapy , Respiratory Tract Diseases/veterinary , Swine , Swine Diseases/diagnosis
2.
Front Immunol ; 10: 572, 2019.
Article in English | MEDLINE | ID: mdl-30972067

ABSTRACT

The rapid evolution of porcine reproductive and respiratory syndrome viruses (PRRSV) poses a major challenge to effective disease control since available vaccines show variable efficacy against divergent strains. Knowledge of the antigenic targets of virus-neutralizing antibodies that confer protection against heterologous PRRSV strains would be a catalyst for the development of next-generation vaccines. Key to discovering these epitopes is the isolation of neutralizing monoclonal antibodies (mAbs) from immune pigs. To address this need, we sought to establish systems to enable the isolation of PRRSV neutralizing porcine mAbs. We experimentally produced a cohort of immune pigs by sequential challenge infection with four heterologous PRRSV strains spanning PRRSV-1 subtypes and PRRSV species. Whilst priming with PRRSV-1 subtype 1 did not confer full protection against a subsequent infection with a PRRSV-1 subtype 3 strain, animals were protected against a subsequent PRRSV-2 infection. The infection protocol resulted in high serum neutralizing antibody titers against PRRSV-1 Olot/91 and significant neutralization of heterologous PRRSV-1/-2 strains. Enriched memory B cells isolated at the termination of the study were genetically programmed by transduction with a retroviral vector expressing the Bcl-6 transcription factor and the anti-apoptotic Bcl-xL protein, a technology we demonstrated efficiently converts porcine memory B cells into proliferating antibody-secreting cells. Pools of transduced memory B cells were cultured and supernatants containing PRRSV-specific antibodies identified by flow cytometric staining of infected MARC-145 cells and in vitro neutralization of PRRSV-1. Collectively, these data suggest that this experimental system may be further exploited to produce a panel of PRRSV-specific mAbs, which will contribute both to our understanding of the antibody response to PRRSV and allow epitopes to be resolved that may ultimately guide the design of immunogens to induce cross-protective immunity.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Porcine respiratory and reproductive syndrome virus/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , B-Lymphocytes/immunology , Cell Line , Epitopes/genetics , Immunologic Memory/genetics , Immunologic Memory/immunology , Neutralization Tests , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/therapy , Proto-Oncogene Proteins c-bcl-6/genetics , Swine , bcl-X Protein/genetics
3.
Vet Immunol Immunopathol ; 200: 40-51, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29776611

ABSTRACT

In this study, we administered specially developed chitosan/alginate nanoparticle encapsulated BV (CH/AL-BV) which has slow-releasing properties and mucosal adhesiveness to pig via nasal route and evaluate whether it can facilitate systemic immune response and improve clearance of porcine reproductive and respiratory syndrome virus (PRRSV). The CH/AL-BV-administered group with PRRSV vaccination showed significantly enhanced Th1-related responses including a high population of CD4+ T lymphocyte and cytokine mRNA levels including interferon-gamma (IFN-γ) and interleukin (IL)-12 and increased PRRSV-specific IgG levels. In the PRRSV challenge experiment, the CH/AL-BV group showed a significant decrease of viral burden in the sera and tissues (lung and bronchial lymph node) and mild interstitial pneumonia signs on both lung gross examination and microscopic evaluation with high levels of PRRSV-specific IgG and viral neutralizing antibody. CH/AL-BV also effectively induced not only Th1-related immune responses including increase in portion of CD4+ T lymphocyte, cytokines (IFN-γ and IL-12), and transcriptional factors (STAT4 and T-bet), but also stimulated IFN-γ-secreting cell families such as CD4+ T lymphocytes and Th/memory cells. Interestingly, the CH/AL-BV group showed decrease in PRRSV-specific immune-suppressive actions, including the T regulatory cell population and its related cytokines (IL-10 and TGF-ß) and transcriptional factors (STAT5 and Foxp3). Therefore, nasal-delivered CH/AL-BV may effectively induce non-specific immune stimulating actions, particularly those related to Th1 responses and viral clearance activities against PRRSV infection. Based on these results, CH/AL-BV could be a promising strategy for overcoming the disadvantages of classical PRRSV vaccination and can be applied as a preventive agent against PRRSV and other viral diseases, particularly those with immune-suppressive characteristics.


Subject(s)
Antibody Formation/drug effects , Bee Venoms/pharmacology , Porcine Reproductive and Respiratory Syndrome/therapy , Porcine respiratory and reproductive syndrome virus/immunology , T-Lymphocytes/drug effects , Administration, Intranasal/veterinary , Alginates/administration & dosage , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibody Formation/immunology , Bee Venoms/administration & dosage , Chitosan/administration & dosage , Drug Delivery Systems/veterinary , Glucuronic Acid/administration & dosage , Hexuronic Acids/administration & dosage , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Immunity, Humoral/drug effects , Immunity, Humoral/immunology , Nanoparticles/administration & dosage , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/pathology , Swine , T-Lymphocytes/immunology
4.
Gene ; 649: 93-98, 2018 Apr 05.
Article in English | MEDLINE | ID: mdl-29366756

ABSTRACT

Interferon-gamma-inducible protein 30 (IFI30) is an IFN-γ-inducible protein that is involved in MHC class II-restricted antigen processing and MHC class I-restricted cross-presentation pathways of adaptive immunity. The present study aimed to investigate the effects of porcine IFI30 expression on PRRSV proliferation in host cells. MARC-145 cells and pig Sertoli (ST) cells were infected with PRRSV after transfection with porcine IFI30 expression vectors and an empty vector. PRRSV copy numbers were analyzed by absolute real-time quantitative PCR, and the results showed that porcine IFI30 expression could significantly inhibit PRRSV transcription. Western blot analysis also determined that IFI30 expression could reduce the production of PRRSV M protein. Flow cytometric analysis indicated that the apoptosis of MARC-145 cells, which are non-porcine but highly permissive to PRRSV cells, was significantly decreased in the IFI30 expression group. In porcine ST cells, apoptosis was significantly increased in IFI30 knockdown cells but not in IFI30-overexpressing cells (**p < 0.01). In conclusion, porcine IFI30 expression may inhibit PRRSV proliferation and host cell apoptosis in vitro.


Subject(s)
Interferon-gamma/pharmacology , Porcine Reproductive and Respiratory Syndrome/therapy , Porcine respiratory and reproductive syndrome virus/drug effects , Animals , Apoptosis , Cell Line , Cell Proliferation , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Porcine Reproductive and Respiratory Syndrome/metabolism , Porcine respiratory and reproductive syndrome virus/metabolism , Swine/genetics , Swine/metabolism , Virus Replication/drug effects
5.
Trends Microbiol ; 25(12): 968-979, 2017 12.
Article in English | MEDLINE | ID: mdl-28652073

ABSTRACT

PRRSV (porcine reproductive and respiratory syndrome virus) is a major economically significant pathogen that has adversely impacted the global swine industry for almost 30 years. Currently PRRSV is estimated to cause losses of almost US$600 million per year in the USA. Except for new mutants that continually emerge during PRRSV outbreaks, our understanding of the virology, origin, and evolution of PRRSV and the host's immune response are largely inadequate. Such limited knowledge impedes development of effective methods to eradicate this virus. In this review, we systematically describe recent advances in anti-PRRSV research, especially focusing on those techniques with the potential to transform current anti-PRRSV strategies. Furthermore, a combination of these new techniques may provide creative insights to guide future PRRSV control and prevention.


Subject(s)
Antiviral Agents/pharmacology , Porcine Reproductive and Respiratory Syndrome/therapy , Porcine respiratory and reproductive syndrome virus/drug effects , Animals , Antiviral Agents/therapeutic use , Disease Models, Animal , Disease Resistance/genetics , Immunity, Innate/immunology , Mice , MicroRNAs/genetics , Porcine Reproductive and Respiratory Syndrome/prevention & control , Porcine respiratory and reproductive syndrome virus/immunology , Swine , Viral Vaccines/immunology , Viral Vaccines/therapeutic use , Virus Replication/drug effects
6.
Transbound Emerg Dis ; 64(1): 194-203, 2017 Feb.
Article in English | MEDLINE | ID: mdl-25879825

ABSTRACT

The objective of this study was to compare the effects of two commercial type 1 porcine reproductive and respiratory syndrome virus (PRRSV)-modified live vaccines on type 1 and type 2 PRRSV shedding in the semen of experimentally infected boars. Upon challenge with PRRSV, unvaccinated boars exhibited an increase in daily rectal temperature (39.4-39.7°C). Vaccination of boars with type 1 PRRSV significantly reduced the amount of type 1 PRRSV load in blood and semen after challenge with type 1 PRRSV, but barely reduced the amount of type 2 PRRSV load in blood and semen after the type 2 PRRSV challenge. There were no significant differences in the reduction of viremia and seminal shedding of type 1 and type 2 PRRSV between the two commercial vaccines. The seminal shedding of PRRSV is independent of viremia. The reduction of type 1 PRRSV seminal shedding coincided with the appearance of type 1 PRRSV-specific interferon-γ secreting cells (IFN-γ-SC) in vaccinated type 1 PRRSV-challenged boars. The frequencies of type 1 PRRSV-specific IFN-γ-SC induced by type 1 PRRSV vaccine are relatively high compared to type 2 PRRSV-specific IFN-γ-SC induced by the same vaccine which may explain why type 1 PRRSV vaccine is more effective in reducing seminal shedding of type 1 PRRSV when compared to type 2 PRRSV in vaccinated challenged boars. These results provide clinical information on how to reduce seminal shedding of type 1 PRRSV in boars using type 1 PRRSV-modified live vaccine.


Subject(s)
Porcine Reproductive and Respiratory Syndrome/therapy , Porcine respiratory and reproductive syndrome virus/immunology , Viral Vaccines/immunology , Virus Shedding , Animals , Porcine Reproductive and Respiratory Syndrome/virology , Semen/virology , Swine , Vaccines, Attenuated/immunology
7.
BMC Vet Res ; 12: 90, 2016 Jun 06.
Article in English | MEDLINE | ID: mdl-27268206

ABSTRACT

BACKGROUND: Porcine reproductive and respiratory syndrome virus (PRRSV) induces one of most important devastating disease of swine worldwide, and the current methods poorly control it. Previous studies have indicated that the nonstructural protein 11 (nsp11) of PRRSV may be an important protein for the immune escape of PRRSV. RESULTS: Here, we firstly explored the effect of over-expression of nsp11 on PRRSV infection and found that over-expression of nsp11 enhanced the PRRSV titers while the small interfering RNA (siRNAs) specifically targeting nsp11 could reduce the PRRSV titers in MARC-145 cells. CONCLUSION: In conclusion, PRRSV nsp11 promotes PRRSV infection in MARC-145 cells and siRNAs targeting nsp11 may be a potential therapeutic strategy to control PRRSV in future.


Subject(s)
Porcine respiratory and reproductive syndrome virus/physiology , Viral Nonstructural Proteins/physiology , Animals , Cell Line , Endoribonucleases/metabolism , Gene Expression , Gene Targeting , HEK293 Cells , Humans , Porcine Reproductive and Respiratory Syndrome/therapy , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/genetics , RNA, Small Interfering , Swine , Transfection , Viral Nonstructural Proteins/genetics , Virus Replication
8.
Brain Behav Immun ; 54: 243-251, 2016 May.
Article in English | MEDLINE | ID: mdl-26872419

ABSTRACT

Although poorly understood, early-life infection is predicted to affect brain microglial cells, making them hypersensitive to subsequent stimuli. To investigate this, we assessed gene expression in hippocampal tissue obtained from a previously published study reporting increased microglial cell activity and reduced hippocampal-dependent learning in neonatal piglets infected with porcine reproductive and respiratory syndrome virus (PRRSV), a virus that induces interstitial pneumonia. Infection altered expression of 455 genes, of which 334 were up-regulated and 121 were down-regulated. Functional annotation revealed that immune function genes were enriched among the up-regulated differentially expressed genes (DEGs), whereas calcium binding and synaptic vesicle genes were enriched among the down-regulated DEGs. Twenty-six genes encoding part of the microglia sensory apparatus (i.e., the sensome) were up-regulated (e.g., IL1R1, TLR2, and TLR4), whereas 15 genes associated with the synaptosome and synaptic receptors (e.g., NPTX2, GABRA2, and SLC5A7) were down-regulated. As the sensome may foretell microglia reactivity, we next inoculated piglets with culture medium or PRRSV at PD 7 and assessed hippocampal microglia morphology and function at PD 28 when signs of infection were waning. Consistent with amplification of the sensome, microglia from PRRSV piglets had enhanced responsiveness to chemoattractants, increased phagocytic activity, and secreted more TNFα in response to lipopolysaccharide and Poly I:C. Immunohistochemical staining indicated PRRSV infection increased microglia soma length and length-to-width ratio. Bipolar rod-like microglia not evident in hippocampus of control piglets, were present in infected piglets. Collectively, this study suggests early-life infection alters the microglia sensome as well as microglial cell morphology and function.


Subject(s)
Hippocampus/pathology , Microglia/physiology , Microglia/virology , Porcine Reproductive and Respiratory Syndrome/pathology , Animals , Brain/immunology , Cytokines/immunology , Disease Models, Animal , Gene Expression , Gene Expression Profiling , Hippocampus/metabolism , Hippocampus/virology , Microglia/metabolism , Porcine Reproductive and Respiratory Syndrome/genetics , Porcine Reproductive and Respiratory Syndrome/metabolism , Porcine Reproductive and Respiratory Syndrome/therapy , Porcine respiratory and reproductive syndrome virus/isolation & purification , Swine , Transcriptome/genetics
9.
Annu Rev Anim Biosci ; 4: 129-54, 2016.
Article in English | MEDLINE | ID: mdl-26646630

ABSTRACT

This review addresses important issues of porcine reproductive and respiratory syndrome virus (PRRSV) infection, immunity, pathogenesis, and control. Worldwide, PRRS is the most economically important infectious disease of pigs. We highlight the latest information on viral genome structure, pathogenic mechanisms, and host immunity, with a special focus on immune factors that modulate PRRSV infections during the acute and chronic/persistent disease phases. We address genetic control of host resistance and probe effects of PRRSV infection on reproductive traits. A major goal is to identify cellular/viral targets and pathways for designing more effective vaccines and therapeutics. Based on progress in viral reverse genetics, host transcriptomics and genomics, and vaccinology and adjuvant technologies, we have identified new areas for PRRS control and prevention. Finally, we highlight the gaps in our knowledge base and the need for advanced molecular and immune tools to stimulate PRRS research and field applications.


Subject(s)
Genome, Viral/genetics , Genomics , Host-Parasite Interactions , Porcine Reproductive and Respiratory Syndrome/prevention & control , Porcine respiratory and reproductive syndrome virus/immunology , Viral Vaccines/immunology , Animals , Gene Expression Profiling , Gene Expression Regulation , Immune System , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/therapy , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/pathogenicity , Reverse Genetics , Swine
10.
Antiviral Res ; 110: 60-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25086213

ABSTRACT

Virus replication depends upon host-cell processes in infected cells, and this is true for porcine reproductive and respiratory syndrome virus (PRRSV), the causative agent of PRRS that is a worldwide threat to the swine industry. Heme oxygenase-1 (HO-1) is a ubiquitously expressed inducible isoform of the first and rate-limiting enzyme for heme degradation. Our previous research suggested that HO-1 may play an important role in PRRSV infection. However, the function of HO-1 in PRRSV infection is unclear. In the present study, Marc-145, PK-15(CD163) cell lines and porcine alveolar macrophages (PAMs) were used to evaluate the effects of HO-1 induction and over-expression on the replication of two different PRRSV strains. Induction of HO-1 markedly decreased the replication of PRRSV strains in the different cells. Similarly, adenoviral-mediated over-expression of HO-1 also greatly decreased the replication of PRRSV. In contrast, ablation of HO-1 using small interfering RNA concomitantly increased PRRSV replication. Therefore, the data were consistent with HO-1 acting as an antiviral factor and these findings suggested that over-expression or induction of HO-1 may provide a potential therapeutic strategy against PRRSV infection.


Subject(s)
Heme Oxygenase-1/genetics , Macrophages, Alveolar/enzymology , Porcine Reproductive and Respiratory Syndrome/genetics , Porcine respiratory and reproductive syndrome virus/growth & development , Virus Replication/genetics , Animals , Cell Line , Chlorocebus aethiops , Heme Oxygenase-1/biosynthesis , Immunity, Innate/genetics , Porcine Reproductive and Respiratory Syndrome/therapy , RNA Interference , RNA, Small Interfering , Swine
11.
Viral Immunol ; 24(6): 475-82, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22111597

ABSTRACT

Reactive oxygen species (ROS) are produced predominantly by phagocytic cells in response to microbial infections. When produced at optimal levels ROS have potent antimicrobial properties. However, excessive production of ROS induces apoptosis/necrosis of infected as well as bystander cells, resulting in inflammatory pathology. Previously we showed that vaccination of pigs with a modified live porcine reproductive and respiratory syndrome virus vaccine (PRRS-MLV) administered intranasally with a potent mucosal adjuvant M. tuberculosis whole-cell lysate (Mtb WCL) induces protective immunity against PRRSV challenge. In this study, using bronchoalveolar lavage fluid cells and peripheral blood mononuclear cells harvested from that study were quantified for the levels of ROS produced. Our results indicated that in vaccinated pigs, levels of ROS were lower compared to unvaccinated PRRSV-challenged pigs. In unvaccinated but PRRSV-challenged pigs, the higher ROS production was associated with increased inflammatory lung pathology. In conclusion, our results suggest that intranasal immunization using PRRS-MLV along with a potent mucosal adjuvant protects pigs against both homologous and virulent heterologous PRRSV challenge, which was associated with reduced ROS production and reduced lung pathology compared to control virus-challenged pigs.


Subject(s)
Porcine Reproductive and Respiratory Syndrome/therapy , Porcine respiratory and reproductive syndrome virus/immunology , Reactive Oxygen Species/metabolism , Viral Vaccines/immunology , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Animals , Flow Cytometry , Immunity , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lung/immunology , Lung/metabolism , Lung/pathology , Lung/virology , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/metabolism , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/virology , Reactive Oxygen Species/immunology , Swine/immunology , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Viral Vaccines/administration & dosage
12.
Vaccine ; 29(46): 8192-204, 2011 Oct 26.
Article in English | MEDLINE | ID: mdl-21925560

ABSTRACT

Vaccination is the principal means used to control and treat porcine reproductive and respiratory syndrome virus (PRRSV) infection. An array of PRRS vaccine products is available in various regions of the world. However, despite extensive efforts, little progress has been made to improve efficacy since the first introduction of a live, attenuated vaccine in 1994 in the USA. Key limitations include: (a) uncertainty about the viral targets of protective immunity that prevents a research focus on individual viral structures and proteins, and frustrates efforts to design novel vaccines; (b) inability to establish clear immunological correlates of protection that requires laborious in vivo challenge models for evaluation of protection against challenge; and (c) the great genetic diversity of PRRSV which requires that challenge experiments be interpreted cautiously since it is not possible to predict how immunological protection against one isolate will translate to broadly cross-protective immunity. Economically significant levels of cross-protection that are provided to a variety of field isolates still cannot assure that effective protection will be conferred to isolates that might emerge in the future. In addition to these substantial barriers to new PRRSV vaccine development, there are enormous gaps in our understanding of porcine immunological mechanisms and processes that provide immunity to PRRSV infection and memory responses for long-term protection. Despite these impediments, we should be confident that progress will be made. Sequencing of the swine genome is providing a rich source of primary knowledge of gene structure and transcriptional regulation that is certain to reveal important insights about the mechanisms of anti-PRRSV immunity, and continued efforts to unravel the details of the interaction of PRRSV with pigs will lead to new insights that overcome the current limitations in the field.


Subject(s)
Porcine Reproductive and Respiratory Syndrome/prevention & control , Porcine Reproductive and Respiratory Syndrome/therapy , Porcine respiratory and reproductive syndrome virus/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Animals , Genetic Variation , Porcine respiratory and reproductive syndrome virus/classification , Porcine respiratory and reproductive syndrome virus/genetics , Swine , United States
13.
Viral Immunol ; 22(3): 173-80, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19435413

ABSTRACT

In this study, pigs were injected with a nonreplicating human adenovirus type 5 vector expressing porcine interferon-alpha (Ad5-pIFN-alpha) and then challenged with porcine reproductive and respiratory syndrome virus (PRRSV) to determine whether the presence of increased levels of IFN-alpha would decrease viral replication and/or disease. Groups of 10 pigs each were inoculated with Ad5-pIFN-alpha and not challenged, Ad5-pIFN-alpha and challenged with PRRSV 1 d later, or inoculated with a control adenovirus that does not express IFN-alpha (Ad5-null) and challenged 1 d later with PRRSV. IFN-alpha levels in all pigs inoculated with the Ad5-pIFN-alpha were elevated the day of challenge (1 d after inoculation), but were undetectable by 3 d after inoculation in the pigs that were not challenged with PRRSV. Pigs inoculated with Ad5-pIFN-alpha and challenged with PRRSV had lower febrile responses, a decreased percentage of lung involvement at 10 d post-infection, delayed viremia and antibody response, and higher serum IFN-alpha levels as a result of PRRSV infection, compared to pigs inoculated with Ad5-null and challenged with PRRSV. These results indicate that IFN-alpha can have protective effects if present during the time of infection with PRRSV.


Subject(s)
Adenoviridae/genetics , Interferon-alpha/biosynthesis , Interferon-alpha/immunology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine respiratory and reproductive syndrome virus/physiology , Virus Replication , Animals , Genetic Therapy/methods , Genetic Vectors , Interferon-alpha/blood , Interferon-gamma/blood , Lung/pathology , Porcine Reproductive and Respiratory Syndrome/pathology , Porcine Reproductive and Respiratory Syndrome/therapy , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Swine , Viremia
14.
Virus Res ; 135(1): 107-14, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18403041

ABSTRACT

PRRSV (porcine reproductive and respiratory syndrome virus) nucleocapsid (N) protein is the most abundant structural protein of the virus. During infection, the N protein is specifically localized to the nucleus and nucleolus in addition to its normal cytoplasmic distribution. Previously, a nuclear localization signal (NLS, 41-PGKK(N/S)KKKN)-null mutant virus (41-PGGGNKKKN) showed reduced viremia and increased production of neutralizing antibodies in infected pigs. However, the mutagenized NLS underwent strong selection pressure in the pig that resulted in partial or complete reversion and reacquisition of NLS function, and thus the biological effect of the NLS-null mutation needed further investigation. In the present study, a total of 9 "reversion resistant" mutants were generated by amino acid deletions and substitutions using an infectious cDNA clone. Two mutant clones (PG--SKKKS and PG--S-KKS) that produced progeny viruses were genetically stable for at least 20 passages in cell culture. Infection of pigs with those mutants induced neutralizing antibodies to higher titers than with wild-type virus. Both mutant viruses induced viremia of lower titer and of shorter duration than wild-type virus. RT-PCR from tonsils showed that both mutants persisted at a reduced level. Virus transmission to contact pigs was also lower in the mutant virus infected groups. No reversion to functional NLS was detected in either mutant from any pig. These data demonstrate that N protein nuclear localization is indeed associated with viral pathogenesis and host response to PRRS.


Subject(s)
Capsid Proteins/metabolism , Cell Nucleus/metabolism , Nuclear Localization Signals/genetics , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/pathogenicity , Amino Acid Sequence , Animals , Antibodies, Viral/blood , Base Sequence , Capsid Proteins/genetics , Cell Line , Cell Nucleus/virology , Mutation , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/therapy , Porcine Reproductive and Respiratory Syndrome/transmission , Porcine respiratory and reproductive syndrome virus/growth & development , Porcine respiratory and reproductive syndrome virus/metabolism , Protein Transport , Random Allocation , Swine , Viremia/immunology , Virulence
15.
Vet J ; 172(3): 506-14, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16169756

ABSTRACT

The purpose of this study was to assess the effects of two currently available commercial European-type modified-live virus (MLV) vaccines against porcine reproductive and respiratory syndrome in a reproductive pig model. Sixteen 90-day pregnant gilts were divided into four groups and allocated to one of the following intranasal treatments: group A gilts served as negative controls; group B gilts were exposed to a virulent European field strain; group C gilts were exposed to vaccine strain VP046 Bis and group D gilts to vaccine strain All-183. The results indicated that MLV strains can replicate in breeding animals and have the ability to cross the placenta. In particular, viraemia was detected in all gilts in group C and 2/4 gilts in group D, at least at one time point. In addition, transplacental infection was demonstrated in 3/4 gilts in group C and 2/4 gilts in group D. However, congenital and early postnatal infection did not have a marked detrimental effect on piglet performance when compared to negative controls, and no statistically significant differences were observed in most cases. Conversely, the reproductive performance of gilts in group B was significantly worse than that of the other groups. Specifically, the number of born-alive piglets, the survival rate of piglets during lactation and the mean weight of weaned pigs were significantly lower. It was concluded that the two commercial European-type MLV vaccines tested had no marked detrimental effects in pregnant gilts, although the MLV strains can cross the placenta leading to the birth of congenitally infected piglets.


Subject(s)
Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/therapy , Porcine respiratory and reproductive syndrome virus/immunology , Pregnancy Complications, Infectious/therapy , Viral Vaccines/therapeutic use , Animals , Animals, Newborn , Antibodies, Viral/blood , Female , Porcine Reproductive and Respiratory Syndrome/congenital , Porcine Reproductive and Respiratory Syndrome/prevention & control , Pregnancy , Pregnancy Complications, Infectious/immunology , Pregnancy Outcome/veterinary , Swine , Vaccines, Attenuated/immunology , Vaccines, Attenuated/therapeutic use , Viral Vaccines/immunology , Viremia/immunology , Viremia/veterinary
16.
Vet Microbiol ; 102(1-2): 11-8, 2004 Aug 19.
Article in English | MEDLINE | ID: mdl-15288922

ABSTRACT

Two experiments were conducted to investigate if virus shedding could be reduced following a killed porcine reproductive and respiratory syndrome virus (PRRSV) vaccination (KV) of PRRSV infected pigs. In experiment 1, PRRSV infected pigs were vaccinated with KV on days 14 and 28 following infection. Viremia and serum neutralizing (SN) antibody were compared to infected pigs with no KV. The second experiment was conducted in an identical manner. In addition to viremia and SN antibody, virus in oropharyngeal scrapings and interferon gamma (IFN-gamma) producing cells were monitored. Magnitude and duration of viremia were not different between KV vaccinated and non-vaccinated groups. No virus was detected in oropharyngeal scraping from any pig, nor was there a difference in the detection of viral RNA. In both experiments, however, increases in SN titer and number of IFN-gamma producing cells were observed. The SN titer was significantly higher in KV vaccinated groups than in non-vaccinated group on days 42 and 42-56 following infection in experiments 1 and 2, respectively. The number of IFN-gamma producing cells was slightly higher in KV vaccinated groups than in non-vaccinated group on days 42 and 63. These observations suggest that KV had no effect on virus shedding. However, previously infected pigs responded immunologically to KV, as demonstrated by increases in SN antibody titers and IFN-gamma producing cells.


Subject(s)
Porcine Reproductive and Respiratory Syndrome/therapy , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/immunology , Vaccination/veterinary , Viral Vaccines/pharmacology , Animals , Antibodies, Viral/blood , Enzyme-Linked Immunosorbent Assay/veterinary , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Neutralization Tests/veterinary , Oropharynx/virology , Porcine Reproductive and Respiratory Syndrome/blood , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine respiratory and reproductive syndrome virus/genetics , RNA, Viral/chemistry , RNA, Viral/genetics , Random Allocation , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Swine , Vaccines, Inactivated/immunology , Vaccines, Inactivated/pharmacology , Viral Vaccines/immunology , Viremia/veterinary , Virus Shedding
17.
Vet Microbiol ; 78(1): 29-37, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11118739

ABSTRACT

The objective of this research was to evaluate the efficacy of two antimicrobials (ampicillin and ceftiofur), a modified-live porcine reproductive and respiratory syndrome virus (PRRSV) vaccine, and low dose exposure to Streptococcus suis on disease associated with PRRSV/S. suis coinfection. Fifty-six, crossbred, PRRSV-free pigs were weaned at 10-12 days of age and randomly assigned to five treatment groups. All pigs were inoculated with 2ml of 10(6.4) TCID50/ml of high virulence PRRSV isolate VR-2385 intranasally at 29-31 days of age (day 0 of the study) followed 7 days later by intranasal inoculation with 2ml of 10(8.9)colony forming units(CFU)/ml S. suis type 2 isolate ISU VDL #40634/94. Pigs in group 1 (n=10) served as untreated infected positive controls. Pigs in group 2 (n=12) were treated with 5.0 mg/kg ceftiofur hydrochloride intramuscularly (IM) on days 8, 11, and 14. Pigs in group 3 (n=11) were treated with 11 mg/kg ampicillin IM on days 8-10. Pigs in group 4 (n=12) were vaccinated 14 days prior to PRRSV challenge with a commercial modified-live PRRSV vaccine. Pigs in group 5 (n=11) were exposed to a 1:100 dilution of the S. suis challenge inoculum 19 days prior to S. suis challenge. Mortality was 80, 25, 82, 83, and 36% in groups 1-5, respectively. The reduced dose S. suis exposure had some residual virulence, evidenced by S. suis induced meningitis in two pigs after exposure. Treatment with ceftiofur hydrochloride and reduced dose exposure to S. suis were the only treatments which significantly (P<0.05) reduced mortality associated with PRRSV/S. suis coinfection, significantly (P<0.05) reduced recovery of S. suis from tissues at necropsy, and significantly (P<0.05) reduced the severity of gross lung lesions.


Subject(s)
Ampicillin/therapeutic use , Cephalosporins/therapeutic use , Penicillins/therapeutic use , Porcine Reproductive and Respiratory Syndrome/microbiology , Porcine respiratory and reproductive syndrome virus/immunology , Streptococcal Infections/veterinary , Streptococcus suis/immunology , Swine Diseases/microbiology , Viral Vaccines/pharmacology , Animals , Combined Modality Therapy/veterinary , Lung/pathology , Porcine Reproductive and Respiratory Syndrome/pathology , Porcine Reproductive and Respiratory Syndrome/therapy , Streptococcal Infections/complications , Streptococcal Infections/pathology , Swine , Swine Diseases/pathology , Swine Diseases/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...