Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
PLoS Pathog ; 20(3): e1012128, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38547254

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) is known to suppress the type I interferon (IFNs-α/ß) response during infection. PRRSV also activates the NF-κB signaling pathway, leading to the production of proinflammatory cytokines during infection. In swine farms, co-infections of PRRSV and other secondary bacterial pathogens are common and exacerbate the production of proinflammatory cytokines, contributing to the porcine respiratory disease complex (PRDC) which is clinically a severe disease. Previous studies identified the non-structural protein 1ß (nsp1ß) of PRRSV-2 as an IFN antagonist and the nucleocapsid (N) protein as the NF-κB activator. Further studies showed the leucine at position 126 (L126) of nsp1ß as the essential residue for IFN suppression and the region spanning the nuclear localization signal (NLS) of N as the NF-κB activation domain. In the present study, we generated a double-mutant PRRSV-2 that contained the L126A mutation in the nsp1ß gene and the NLS mutation (ΔNLS) in the N gene using reverse genetics. The immunological phenotype of this mutant PRRSV-2 was examined in porcine alveolar macrophages (PAMs) in vitro and in young pigs in vivo. In PAMs, the double-mutant virus did not suppress IFN-ß expression but decreased the NF-κB-dependent inflammatory cytokine productions compared to those for wild-type PRRSV-2. Co-infection of PAMs with the mutant PRRSV-2 and Streptococcus suis (S. suis) also reduced the production of NF-κB-directed inflammatory cytokines. To further examine the cytokine profiles and the disease severity by the mutant virus in natural host animals, 6 groups of pigs, 7 animals per group, were used for co-infection with the mutant PRRSV-2 and S. suis. The double-mutant PRRSV-2 was clinically attenuated, and the expressions of proinflammatory cytokines and chemokines were significantly reduced in pigs after bacterial co-infection. Compared to the wild-type PRRSV-2 and S. suis co-infection control, pigs coinfected with the double-mutant PRRSV-2 exhibited milder clinical signs, lower titers and shorter duration of viremia, and lower expression of proinflammatory cytokines. In conclusion, our study demonstrates that genetic modification of the type I IFN suppression and NF-κB activation functions of PRRSV-2 may allow us to design a novel vaccine candidate to alleviate the clinical severity of PRRS-2 and PRDC during bacterial co-infection.


Assuntos
Coinfecção , Interferon Tipo I , Síndrome Respiratória e Reprodutiva Suína , Vírus da Síndrome Respiratória e Reprodutiva Suína , Suínos , Animais , Vírus da Síndrome Respiratória e Reprodutiva Suína/metabolismo , Citocinas/genética , Citocinas/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Macrófagos Alveolares/metabolismo , Interferon Tipo I/metabolismo , Síndrome Respiratória e Reprodutiva Suína/genética , Síndrome Respiratória e Reprodutiva Suína/metabolismo
2.
Infect Immun ; 90(4): e0057421, 2022 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-35254092

RESUMO

Viral respiratory infections predispose lungs to bacterial coinfections causing a worse outcome than either infection alone. Porcine reproductive and respiratory syndrome virus (PRRSV) causes pneumonia in pigs and is often associated with bacterial coinfections. We examined the impact of providing weanling pigs a Bacillus-based direct-fed microbial (DFM) on the syndrome resulting from infection with either Salmonella enterica serotype Choleraesuis alone, or in combination with PRRSV. Nine days after the bacterial challenge, Salmonella was isolated from ileocecal lymph nodes of all challenged pigs regardless of DFM treatment. Compared to the single bacterial challenge, the dual challenge with Salmonella and PRRSV resulted in a pathogenic synergy exhibited by a higher rate of Salmonella colonization in the lung and a more extensive and severe interstitial pneumonia. Provision of DFM to dually challenged pigs reduced the rate of lung colonization by Salmonella, eliminated or reduced the presence of PRRSV in the lung, and reduced the extent and severity of gross lung pathology. Dually challenged pigs that received DFM had increased concentrations of interleukin 1 (IL-1) and IL-8 in lung lavage fluids, accompanied by increased expression in their blood cells of nucleotide-binding oligomerization domain receptor 2 (NOD2) and triggering receptor expressed in myeloid cells 1 (TREM-1) molecules. These changes in pulmonary inflammatory cytokine production and increased expression of NOD2 and TREM-1 suggest that the DFM exerted a systemic modulating effect on innate immunity. These observations are consistent with the notion that tonic stimulation by gut-derived microbial products can poise innate immunity to fight infections in the respiratory tract.


Assuntos
Bacillus , Coinfecção , Pneumonia , Vírus da Síndrome Respiratória e Reprodutiva Suína , Salmonella enterica , Animais , Salmonella , Sorogrupo , Suínos , Receptor Gatilho 1 Expresso em Células Mieloides
3.
J Virol ; 92(2)2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29070690

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) infects alveolar macrophages (AMϕ), causing dysregulated alpha interferon (IFN-α) and tumor necrosis factor alpha (TNF-α) production through a mechanism(s) yet to be resolved. Here, we show that AMϕ infected with PRRSV secreted a reduced quantity of IFN-α following exposure of the cell to synthetic double-stranded RNA (dsRNA). This reduction did not correlate with reduced IFNA1 gene transcription. Rather, it coincided with two events that occurred late during infection and that were indicative of translational attenuation, specifically, the activation of eukaryotic translation initiation factor 2α (eIF2α) and the appearance of stress granules. Notably, the typical rapid production of TNF-α by AMϕ exposed to lipopolysaccharide (LPS) was suppressed or enhanced by PRRSV, depending on when the LPS exposure occurred after virus infection. If exposure was delayed until 6 h postinfection (hpi) so that the development of the cytokine response coincided with the time in which phosphorylation of eIF2α by the stress sensor PERK (protein kinase RNA [PKR]-like ER kinase) occurred, inhibition of TNF-α production was observed. However, if LPS exposure occurred at 2 hpi, prior to a detectable onset of eIF2α phosphorylation, a synergistic response was observed due to the earlier NF-κB activation via the stress sensor IRE1α (inositol-requiring kinase 1α). These results suggest that the asynchronous actions of two branches of the unfolded protein response (UPR), namely, IRE1α, and PERK, activated by ER stress resulting from the virus infection, are associated with enhancement or suppression of TNF-α production, respectively.IMPORTANCE The activation of AMϕ is controlled by the microenvironment to deter excessive proinflammatory cytokine responses to microbes that could impair lung function. However, viral pneumonias frequently become complicated by secondary bacterial infections, triggering severe inflammation, lung dysfunction, and death. Although dysregulated cytokine production is considered an integral component of the exacerbated inflammatory response in viral-bacterial coinfections, the mechanism responsible for this event is unknown. Here, we show that PRRSV replication in porcine AMϕ triggers activation of the IRE1α branch of the UPR, which causes a synergistic TNF-α response to LPS exposure. Thus, the severe pneumonias typically observed in pigs afflicted with PRRSV-bacterial coinfections could result from dysregulated, overly robust TNF-α production in response to opportunistic pathogens that is not commensurate with the typical restrained reaction by uninfected AMϕ. This notion could help in the design of therapies to mitigate the severity of viral and bacterial coinfections.


Assuntos
Interferon-alfa/genética , Macrófagos Alveolares/virologia , Síndrome Respiratória e Reprodutiva Suína/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/fisiologia , Resposta a Proteínas não Dobradas , Animais , Linhagem Celular , Fator de Iniciação 2 em Eucariotos/metabolismo , Genótipo , Interferon-alfa/metabolismo , Lipopolissacarídeos/efeitos adversos , Macrófagos Alveolares/imunologia , Síndrome Respiratória e Reprodutiva Suína/genética , Síndrome Respiratória e Reprodutiva Suína/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Suínos , Fator de Necrose Tumoral alfa/metabolismo , Replicação Viral , eIF-2 Quinase/metabolismo
4.
J Vet Med Educ ; 44(3): 471-479, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28876996

RESUMO

The organization and delivery of a curriculum is the responsibility of the faculty in educational institutions. Curricular revision is often a hotly debated topic in any college faculty. At the University of Illinois, a 2006 mandate for curriculum modernization from the American Veterinary Medical Association Council on Education provided impetus for a long-discussed curricular revision. After two iterations and a lengthy development process, a new curriculum was gradually implemented at Illinois with the August 2009 matriculation of the Class of 2013. The goals of the revision included earlier clinical exposure for veterinary students through introductions to clinical rotations in years 1 to 3 and an integrated body systems approach in lecture/laboratory courses. A new Clinical Skills Learning Center facilitates development of clinical skills earlier in the curriculum and promotes the development of those skills throughout all 4 years of the curriculum. New outcomes assessments include comprehensive written examinations and Objective Structured Clinical Examinations (OSCEs) in years 2 and 3. Curriculum management, including grading of clinical rotations in all 4 years, is achieved through a commercially available software package. For the past 5 years, when candidates were asked why they chose to apply to Illinois, the new curriculum (27.4%) was the most common answer given during interviews. The Illinois revision has resulted in measurably increased veterinary student self-confidence (p<.001) at graduation.


Assuntos
Estágio Clínico , Currículo/tendências , Educação em Veterinária/organização & administração , Faculdades de Medicina Veterinária/organização & administração , Acreditação , Educação em Veterinária/normas , Humanos , Illinois , Inovação Organizacional , Faculdades de Medicina Veterinária/normas
5.
Arch Virol ; 161(6): 1579-89, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27008569

RESUMO

Porcine reproductive and respiratory syndrome (PRRS) is a leading cause of economic burden to the pork industry worldwide. The routinely used modified live PRRS virus vaccine (PRRS-MLV) induces clinical protection, but it has safety concerns. Therefore, in an attempt to develop a safe and protective inactivated PRRSV vaccine, we generated PRRS-virus-like-particles (PRRS-VLPs) containing the viral surface proteins GP5-GP4-GP3-GP2a-M or GP5-M using a novel baculovirus expression system. Our in vitro results indicated that the desired PRRSV proteins were incorporated in both the VLPs preparations based on their reactivity in immunogold electron microscopy and ELISA. To boost their immunogenicity in pigs, we entrapped the PRRS-VLPs in PLGA nanoparticles and coadministered them intranasally with a potent adjuvant. We then evaluated their efficacy in pigs against a viral challenge using a virulent heterologous field isolate. Our results indicated that PRRS-VLPs induced an anamnestic immune response, since we observed boosted IgG and IFN-γ production in vaccinated and virus-challenged animals, but not during the pre-challenge period. Importantly, a two-log reduction in the lung viral load was detected in PRRS-VLP-vaccinated animals. In conclusion, we generated PRRS-VLPs containing up to five viral surface proteins and demonstrated their immunogenicity in pigs, but further studies are required to improve its immunogenicity and efficacy as a vaccine candidate.


Assuntos
Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Linhagem Celular , Citocinas/metabolismo , Genes Virais , Pulmão/imunologia , Pulmão/virologia , Síndrome Respiratória e Reprodutiva Suína/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Sus scrofa , Suínos , Vacinas de Produtos Inativados/genética , Vacinas de Produtos Inativados/imunologia , Vacinas de Partículas Semelhantes a Vírus/genética , Vacinas de Partículas Semelhantes a Vírus/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Carga Viral , Vacinas Virais/genética
6.
Vet Sci ; 10(3)2023 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-36977259

RESUMO

Point-of-care diagnostic technologies are becoming more widely available for production species. Here, we describe the application of reverse transcription loop-mediated isothermal amplification (RT-LAMP) to detect the matrix (M) gene of influenza A virus in swine (IAV-S). M-specific LAMP primers were designed based on M gene sequences from IAV-S isolated in the USA between 2017 and 2020. The LAMP assay was incubated at 65 °C for 30 min, with the fluorescent signal read every 20 s. The assay's limit of detection (LOD) was 20 M gene copies for direct LAMP of the matrix gene standard, and 100 M gene copies when using spiked extraction kits. The LOD was 1000 M genes when using cell culture samples. Detection in clinical samples showed a sensitivity of 94.3% and a specificity of 94.9%. These results show that the influenza M gene RT-LAMP assay can detect the presence of IAV in research laboratory conditions. With the appropriate fluorescent reader and heat block, the assay could be quickly validated as a low-cost, rapid, IAV-S screening tool for use on farms or in clinical diagnostic labs.

7.
J Virol ; 85(6): 2703-13, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21191013

RESUMO

Although enveloped viruses typically trigger the prodigious secretion of alpha interferon (IFN-α) by plasmacytoid dendritic cells (pDC), porcine pDC remain quiescent when exposed to porcine reproductive and respiratory syndrome virus (PRRSV). This inactivity is likely due to virus-mediated interference since the typical IFN-α response by either purified or nonsorted porcine pDC to transmissible gastroenteritis virus (TGEV) or the Toll-like receptor 9 agonist, oligodeoxynucleotide (ODN) D19, was markedly reduced in the presence of PRRSV. Suppression occurred independently of virus viability and acidification of pDC early endosomes but correlated with diminished levels of IFN-α mRNA. This change was attributed to an abrogation of transcription resulting from a decrease in the otherwise enhanced amounts of the requisite interferon regulatory factor 7 (IRF-7), whose gene expression in turn was limited as a consequence of a lessened availability of nuclear-localized signal transducer and activator of transcription 1 (STAT1). While PRRSV also inhibited tumor necrosis factor alpha (TNF-α) synthesis by pDC responding to either agent, only the interleukin-2 (IL-2) and IL-6 production instigated by ODN D19 exposure was blocked. Likewise, PRRSV did not impact a specific TGEV-associated enhancement of IL-8 expression. Moreover, an augmented phosphorylation of NF-κB seen in activated pDC was not only unaffected by PRRSV but actually occurred in its presence. Thus, as supported by a demonstrated resilience of pDC to PRRSV infection, this pathogen may interact with a cell surface protein(s) to selectively impede the completion of cascades involved in cytokine production by stimulated pDC.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/virologia , Interferon Tipo I/antagonistas & inibidores , Síndrome Respiratória e Reprodutiva Suína/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/patogenicidade , Animais , Regulação para Baixo , Expressão Gênica , Fator Regulador 7 de Interferon/biossíntese , América do Norte , Oligodesoxirribonucleotídeos/imunologia , RNA Mensageiro/biossíntese , Suínos , Vírus da Gastroenterite Transmissível/imunologia
8.
Emerg Microbes Infect ; 9(1): 1245-1253, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32515659

RESUMO

The main target cells for African swine fever virus (ASFV) replication in pigs are of monocyte macrophage lineage and express markers typical of the intermediate to late stages of differentiation. The lack of a porcine cell line, which accurately represents these target cells, limits research on virus host interactions and the development of live-attenuated vaccine strains. We show here that the continuously growing, growth factor dependent ZMAC-4 porcine macrophage cell line is susceptible to infection with eight different field isolates of ASFV. Replication in ZMAC-4 cells occurred with similar kinetics and to similar high titres as in primary porcine bone marrow cells. In addition we showed that twelve passages of an attenuated strain of ASFV, OURT88/3, in ZMAC-4 cells did not reduce the ability of this virus to induce protection against challenge with virulent virus. Thus, the ZMAC-4 cells provide an alternative to primary cells for ASFV replication.


Assuntos
Vírus da Febre Suína Africana/fisiologia , Técnicas de Cultura de Células/métodos , Macrófagos/citologia , Vacinas Atenuadas/farmacologia , Febre Suína Africana/virologia , Vírus da Febre Suína Africana/imunologia , Vírus da Febre Suína Africana/isolamento & purificação , Animais , Células da Medula Óssea/virologia , Linhagem Celular , Proliferação de Células , Macrófagos/virologia , Inoculações Seriadas , Suínos , Vacinas Atenuadas/imunologia , Replicação Viral
9.
Virus Res ; 280: 197898, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32061619

RESUMO

The NC229 research consortium was created in 1999 in response to the emergence of porcine reproductive and respiratory syndrome virus (PRRSV), a viral agent responsible for devastating economic losses to the swine industry. The project follows the traditional "consortium" approach for Multistate Agricultural Research driven through the US State Agricultural Experiment Stations (SAES), wherein stakeholder-driven needs to combat swine infectious diseases are identified and scientific solutions pursued by combining funds from federal, state, commodity groups, and the animal health industry. The NC229 consortium was the main driving force in successfully competing for a USDA multi-station Coordinated Agricultural Project (PRRS CAP-I) in 2004-2008, immediately followed by a renewal for 2010-2014 (PRRS CAP-II)-, resulting in an overall record achievement of almost $10 million dollars. The CAP funding was not only useful for quality research, extension, and education in PRRS and related diseases, but also instrumental in enabling the group to leverage swine industry funding of more than $34 million dollars, distributed between creative research and extension on PRRS during the last 20 years. The North American/International PRRS Symposium, now recognized by the community as a highly effective platform for the exchange of basic research findings and fundamental translational technology, is directly derived from the NC229 consortium. Other significant offshoots from NC229 include the PHGC (PRRS Host Genomic Consortium), a platform for discoveries on the role of host genetics during PRRSV infection, since 2007. Since 2009, the NC229 consortium has expanded its collective research interests beyond PRRSV to include nine other emerging viral diseases of swine. In the current project (2019-2024), African Swine Fever Virus (ASFV) retains a central focus, with the goal of harnessing the group's expertise in promoting preparedness for the global control of ASFV.


Assuntos
Doenças Transmissíveis Emergentes/veterinária , Doenças Transmissíveis Emergentes/virologia , Pesquisa/organização & administração , Viroses/veterinária , Animais , Congressos como Assunto , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Pesquisa/economia , Participação dos Interessados , Suínos , Estados Unidos , Viroses/prevenção & controle
10.
J Am Vet Med Assoc ; 232(10): 1530-5, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18479244

RESUMO

OBJECTIVE: To determine whether 6.5-week-old gilts that have not previously been exposed to porcine reproductive and respiratory syndrome (PRRS) virus can be acclimatized to an endemic strain of the virus by commingling with age-matched gilts inoculated with the endemic PRRS virus strain and whether 10.5-week-old gilts can be acclimatized by commingling with age-matched inoculated or contact-exposed animals. DESIGN: Randomized controlled longitudinal study. ANIMALS: 80 gilts seronegative for PRRS on a farm in the Midwestern United States with a history of PRRS. PROCEDURES: 20 gilts were inoculated with the endemic PRRS virus strain at 6.5 weeks of age (group 1) and were commingled with 20 gilts that were not inoculated (group 2). Four weeks later, the remaining 40 gilts (group 3) were commingled with gilts in groups 1 and 2. Presence of viral RNA in the tonsils, seroconversion rate, serum neutralizing antibody titers, interferon-gamma-mediated cellular immunity, and reproductive outcomes were analyzed. RESULTS: Acclimatization of PRRS virus-naïve pigs was achieved by means of contact exposure at both 6.5 and 10.5 weeks of age. No differences were observed among the 3 groups with respect to development of anti-PRRS virus-specific immune responses or reproductive outcomes. CONCLUSIONS AND CLINICAL RELEVANCE: Results suggested that contact exposure of 6.5- to 10.5-week-old pigs that had not previously been exposed to PRRS virus to pigs inoculated with endemic PRRS virus may be an efficient acclimatization strategy for controlling outbreaks on commercial farms on which PRRS is endemic.


Assuntos
Anticorpos Antivirais/sangue , Síndrome Respiratória e Reprodutiva Suína/transmissão , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Vacinação/veterinária , Adaptação Fisiológica/imunologia , Fatores Etários , Animais , Animais Recém-Nascidos , Exposição Ambiental , Ensaio de Imunoadsorção Enzimática/métodos , Ensaio de Imunoadsorção Enzimática/veterinária , Feminino , Estudos Longitudinais , Testes de Neutralização/métodos , Testes de Neutralização/veterinária , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Distribuição Aleatória , Suínos/crescimento & desenvolvimento , Aumento de Peso
11.
Vet Microbiol ; 123(1-3): 69-85, 2007 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-17376612

RESUMO

The efficacy of two different types of commercial vaccines against PRRSV (Euro-type) was evaluated based on clinical parameters upon challenge as well as post-challenge virological profiles (viremia and viral load in tissues upon necropsy, measured in both cases by quantitative real time PCR). In an attempt to establish correlates of protective immunity, two commonly proposed parameters predictive of immunity were measured: (1) serologic responses (ELISA and neutralizing antibodies), (2) frequency of gamma interferon-producing cells in peripheral blood mononuclear cell fraction. The vaccines compared consisted of two commercially available products that are regularly marketed in Spain: one modified live virus and one killed vaccine. The efficacy assay was carried out by vaccinating twice 3 weeks apart groups of 5 and-a-half month-old female swine and then challenging them with a European type 1 PRRSV strain (Lelystad). The results obtained indicate that the modified live virus vaccine was the only type of vaccine capable of establishing protective immunity, as measured by viral load in blood and tissues. The killed vaccine, in spite of this product evoking a spontaneous interferon-gamma response and post-challenge titers of virus-neutralizing antibody, evoked no measurable protective immunity. In the case of the modified live vaccine, the protection exhibited did not appear to be based on humoral but rather on cell-mediated immunity.


Assuntos
Interferon gama/biossíntese , Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Química Farmacêutica , Feminino , Interferon gama/metabolismo , Interleucina-10/metabolismo , Síndrome Respiratória e Reprodutiva Suína/metabolismo , Testes Sorológicos , Organismos Livres de Patógenos Específicos , Suínos
12.
J Am Vet Med Assoc ; 228(7): 1082-8, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16579789

RESUMO

OBJECTIVE: To compare immunologic responses and reproductive outcomes in sows housed under field conditions following controlled exposure to a wild-type strain of porcine reproductive and respiratory syndrome virus (PRRSV strain WTV) or vaccination with a modified-live virus (MLV) vaccine. DESIGN: Randomized controlled trial. ANIMALS: 30 PRRSV-naïve 10-week-old female pigs. PROCEDURE: Humoral and cell-mediated immune responses were monitored while pigs were held in isolation for 84 days after inoculation with the WTV strain (n = 10), inoculation with the WTV strain and 42 days later vaccination with a killed-virus vaccine (10), or vaccination with an MLV vaccine (10). Reproductive outcomes were measured after pigs were released into the farm herd. RESULTS: Inoculation with the WTV strain, regardless of whether a killed-virus vaccine was subsequently administered, elicited faster and more substantial production of strain-specific neutralizing antibodies, as well as a more rapid generation of interferon-gamma secreting cells, than did vaccination with the MLV vaccine. Despite the enhanced immune responses in pigs inoculated with the WTV strain, animals vaccinated with the MLV vaccine produced a mean of 2.45 more pigs than did sows exposed to the WTV strain, mainly because of a lower rate for failure to conceive. CONCLUSIONS AND CLINICAL RELEVANCE: Results suggest that current assays of immunity to PRRSV correlate only imperfectly with degree of clinical protection and that the practice of controlled exposure of sows to a circulating PRRSV strain should be reconsidered in light of negative clinical outcomes.


Assuntos
Anticorpos Antivirais/sangue , Síndrome Respiratória e Reprodutiva Suína/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Reprodução , Vacinas Virais/imunologia , Animais , Feminino , Tamanho da Ninhada de Vivíparos , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Síndrome Respiratória e Reprodutiva Suína/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/patogenicidade , Gravidez , Taxa de Gravidez , Suínos , Vacinas Atenuadas , Vacinas de Produtos Inativados , Vacinas Virais/administração & dosagem
13.
J Am Vet Med Assoc ; 226(10): 1707-11, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15906573

RESUMO

OBJECTIVE: To determine whether cell-mediated immunity against porcine reproductive and respiratory syndrome (PRRS) virus is correlated with protection against reproductive failure in sows during clinical outbreaks of PRRS in commercial herds. DESIGN: Outbreak investigation in 4 swine breeding herds. ANIMALS: 97 sows. PROCEDURES: On each farm, blood samples were collected from sows with clinical signs (abortion or increased fetal death; case sows) and from clinically normal sows (control sows). The intensity of the cell-mediated immune (CMI) response was determined by use of an interferon-gamma enzyme-linked immunospot (ELISPOT) assay. Multiple logistic regression analyses and t tests were used to compare ELISPOT assay values between case and control sows. Multiple linear regression was used to investigate associations between cell-mediated immunity and the magnitude of clinical signs. RESULTS: In 2 farms, case sows had lower ELISPOT assay values than control sows. A negative association between the intensity of the CMI response and the number of pigs born dead per litter was detected on 1 farm. In 1 farm, no association was detected between the intensity of the CMI response and protection against reproductive failure. CONCLUSIONS AND CLINICAL RELEVANCE: Evidence that a strong CMI response was correlated with protection against clinical PRRS was detected in 3 of 4 farms. However, farms and sows within farms varied considerably in their immune responsiveness and in the degree to which they were protected clinically. Increasing cell-mediated immunity within infected herds has the potential to decrease clinical reproductive disease, but only if the sources of intra- and interfarm variation in the intensity of cell-mediated immunity to PRRS virus can be identified.


Assuntos
Aborto Animal/virologia , Surtos de Doenças/veterinária , Morte Fetal/veterinária , Síndrome Respiratória e Reprodutiva Suína/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Animais , Feminino , Morte Fetal/virologia , Imunidade Celular , Modelos Logísticos , Síndrome Respiratória e Reprodutiva Suína/epidemiologia , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Gravidez , Fatores de Risco , Suínos
14.
Vet Immunol Immunopathol ; 167(1-2): 1-14, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26209116

RESUMO

Many highly effective vaccines have been produced against viruses whose virulent infection elicits strong and durable protective immunity. In these cases, characterization of immune effector mechanisms and identification of protective epitopes/immunogens has been informative for the development of successful vaccine programs. Diseases in which the immune system does not rapidly clear the acute infection and/or convalescent immunity does not provide highly effective protection against secondary challenge pose a major hurdle for clinicians and scientists. Porcine reproductive and respiratory syndrome virus (PRRSV) falls primarily into this category, though not entirely. PRRSV causes a prolonged infection, though the host eventually clears the virus. Neutralizing antibodies can provide passive protection when present prior to challenge, though infection can be controlled in the absence of detectable neutralizing antibodies. In addition, primed pigs (through natural exposure or vaccination with a modified-live vaccine) show some protection against secondary challenge. While peripheral PRRSV-specific T cell responses have been examined, their direct contribution to antibody-mediated immunity and viral clearance have not been fully elucidated. The innate immune response following PRRSV infection, particularly the antiviral type I interferon response, is meager, but when provided exogenously, IFN-α enhances PRRSV immunity and viral control. Overall, the quality of immunity induced by natural PRRSV infection is not ideal for informing vaccine development programs. The epitopes necessary for protection may be identified through natural exposure or modified-live vaccines and subsequently applied to vaccine delivery platforms to accelerate induction of protective immunity following vaccination. Collectively, further work to identify protective B and T cell epitopes and mechanisms by which PRRSV eludes innate immunity will enhance our ability to develop more effective methods to control and eliminate PRRS disease.


Assuntos
Síndrome Respiratória e Reprodutiva Suína/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Imunidade Adaptativa , Animais , Anticorpos Neutralizantes/biossíntese , Anticorpos Antivirais/biossíntese , Antígenos Virais , Reações Cruzadas , Epitopos , Interações Hospedeiro-Patógeno/imunologia , Evasão da Resposta Imune , Imunidade Celular , Imunidade Inata , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Síndrome Respiratória e Reprodutiva Suína/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/patogenicidade , Sus scrofa , Suínos , Linfócitos T/imunologia , Vacinas Virais/imunologia
15.
Viral Immunol ; 15(4): 533-47, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12513925

RESUMO

The immunology of porcine reproductive and respiratory syndrome virus (PRRS) begins with an initial encounter of PRRSV with the pig. Regardless of the route of entry of PRRSV--via inhalation, intramuscular vaccination, insemination, or other routes--productive infection occurs predominately in alveolar macrophages of the lung. Thus, innate responses of the lung and the alveolar macrophage comprise the initial defense against PRRSV. The virus appears not to elicit innate interferon and cytokine responses characteristic of other strongly immunogenic viral pathogens, and its effects are consistent with induction of a weak adaptive immune response. Humoral and cell-mediated immunity is induced in due course, and results in clearance of virus from the circulation but not from lymphoid tissues, where the infection becomes persistent. Subsequent reexposure to PRRSV elicits an anamnestic response that is partially to completely protective. Within this unconventional picture of anti-PRRSV immunity lie a variety of unresolved issues, including the nature of protective immunity within individual pigs and among pigs in commercial populations, the efficacy of protective immunity against genetically different PRRSV isolates, the effects of developmental age, sex, genetics, and other host factors on the immune response to PRRSV, and the possible suppression of host immunity to other pathogens.


Assuntos
Anticorpos Antivirais/sangue , Imunidade Celular , Síndrome Respiratória e Reprodutiva Suína/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Animais , Síndrome Respiratória e Reprodutiva Suína/virologia , Suínos
16.
Viral Immunol ; 15(4): 557-66, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12513927

RESUMO

The immune response of swine to vaccination with a live, attenuated PRRSV was assessed in the presence and absence of cytokine adjuvants or cholera toxin (CT) to address the hypothesis that adjuvant danger signals, that is, inflammatory cytokines and bacterial extoxin, stimulate a more robust immune response. Animals received four injections of recombinant porcine IL-1 and IL-6, IL-12 alone, or CT alone within 1 week of intramuscular administration of a vaccine strain of PRRSV, Ingelvac MLV. Serological and cell-mediated responses were monitored for 42 days after vaccination and for a further 10 days after challenge with the virulent VR2332 strain of PRRSV. First, the principal observation was an enhancing effect of IL-12 on the interferon gamma response to PRRSV, with a more rapid and heightened PRRSV-specific interferon gamma ELISPOT response in peripheral blood mononuclear cells. The more rapid and robust development of a cell-mediated immune response, as determined by this assay, suggests that IL-12 may influence the induction of antigen-specific T cell responses. Second, animals that received CT adjuvant displayed a more robust antibody response to GP5, the major envelope glycoprotein. Anti-GP5 titers peaked at 21 days after vaccination at more than twice the level of any other treatment, for which the peak response was at 28 days. Third, there was no evidence of PRRSV immunosuppression of immunity to unrelated antigens, including circovirus. CT is a potent mucosal adjuvant, particularly for antibody responses. It acts in part through the production of IL-1 in macrophages, but its effect was not replaced by combination treatment with IL-1 and IL-6. In sum, the results suggest that cytokine adjuvants, particularly IL-12, and CT have the potential to enhance immune responses to live viral vaccines.


Assuntos
Adjuvantes Imunológicos/efeitos adversos , Toxina da Cólera/efeitos adversos , Citocinas/efeitos adversos , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Vacinação/veterinária , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Toxina da Cólera/imunologia , Citocinas/imunologia , Feminino , Interleucina-12/efeitos adversos , Interleucina-12/imunologia , Masculino , Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Suínos , Linfócitos T/imunologia , Vacinas Atenuadas/imunologia
17.
Vet Immunol Immunopathol ; 102(3): 199-216, 2004 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-15507306

RESUMO

The natural response of pigs to porcine reproductive and respiratory syndrome virus (PRRSV) infections and vaccinations needs to be altered so that better protection is afforded against both homologous and heterologous challenges by this pathogen. To address this problem, real-time gene expression assays were coupled with cytokine Elispot and protein analyses to assess the nature of the anti-PRRSV response of pigs immunized with modified live virus (MLV) vaccine. Although T helper 1 (Th1) immunity was elicited in all vaccinated animals, as evidenced by the genesis of PRRSV-specific interferon-gamma secreting cells (IFNG SC), the overall extent of the memory response was variable and generally weak. Peripheral blood mononuclear cells (PBMC) isolated from these pigs responded to PRRSV exposure with a limited increase in their expression of the Th1 immune markers, IFNG, tumor necrosis factor-alpha and interleukin-15 (IL15), and a reduction in the quantity of mRNAs encoding the innate and inflammatory proteins, IL1B, IL8 and IFNA. Efforts to enhance Th1 immunity, by utilizing an expression plasmid encoding porcine IFNA (pINA) as an adjuvant, resulted in a temporary increase in the frequency of PRRSV-specific IFNG SC but only minor changes overall in the expression of Th1 associated cytokine or innate immune marker mRNA by virus-stimulated PBMC. Administration of pINA, however, did correlate with decreased IL1B secretion by cultured, unstimulated PBMC but had no effect on their ability to release IFNG. Thus, while exogenous addition of IFNA during PRRSV vaccination has an impact on the development of a Th1 immune response, other alterations will be required for substantial boosting of virus-specific protection.


Assuntos
Regulação da Expressão Gênica/imunologia , Interferon-alfa/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Suínos/imunologia , Vacinas Virais/imunologia , Adjuvantes Imunológicos , Animais , Citocinas/imunologia , Imunidade Celular , Interferon-alfa/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Plasmídeos/genética , Plasmídeos/imunologia , Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Análise de Regressão , Fatores de Tempo , Regulação para Cima/imunologia , Vacinas Atenuadas/imunologia
18.
Vet Immunol Immunopathol ; 102(3): 299-314, 2004 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-15507313

RESUMO

Immunization of pigs with a modified live porcine reproductive and respiratory syndrome virus (PRRSV) vaccine initially elicits a weak interferon (IFN)-gamma response. To improve the immune response, an adjuvant consisting of plasmid encoding either porcine interleukin (IL)-12 or IFN-alpha was co-administered during vaccination. In the presence of either adjuvant, at least a three-fold increase in the primary virus-specific IFN-gamma response was observed. While this enhancement was only transient (1 week) when the IL-12 expressing plasmid was used, the effect was not only still apparent at 6 weeks after vaccination in the presence of the IFN-alpha expressing plasmid but even after challenge with a virulent genetically divergent PRRSV. In contrast, no effect of either adjuvant on the production of anti-virus antibodies was noticed throughout the study. Despite the apparent augmentation of a T helper (Th) 1 type response by the inclusion of IFN-alpha or IL-12 during vaccination, this modulation did not necessarily correlate with a reduction in viremia. Since a similar increase in the degree of the IFN-gamma response to the PRRSV vaccine could be achieved by substituting polyinosinic-polycytidylic acid in lieu of either cytokine, exposure to PRRSV in the presence of a variety of Th 1 polarizing molecules can positively influence the development of the cell-mediated immune response of swine to this pathogen. Conceivably, such intervention could be applied to improve the formulation of anti-PRRSV vaccines.


Assuntos
Interferon-alfa/imunologia , Interleucina-12/imunologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , RNA de Cadeia Dupla/imunologia , Suínos/imunologia , Células Th1/imunologia , Animais , Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/virologia , Fatores de Tempo , Vacinas Virais/imunologia
19.
Vet Immunol Immunopathol ; 100(1-2): 105-11, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15183000

RESUMO

This publication describes the cloning of full or partial length sequences for pig TBX21 (T-bet), MYD88, ICSBP1, CD8A (CD8alpha), CD8B (CD8beta), and CD28 cDNAs. Real-time PCR assays have been developed for the relative quantitation of these products as well as previously characterized transcripts that encode exon A-containing CD45, HLX1, IRF1, STAT1 and RPL32. When used for examining temporal immune gene expression in the liver of Toxoplasma gondii infected pigs, the positive regulators of Th1 responses, IRF1, MYD88, and STAT1, were found to be expressed prior to the simultaneous upregulation of interferon gamma (IFNG), HLX1 and TBX21 gene expression. In contrast, in the mesenteric lymph node (MLN), only expression of IRF1 and IFNG was significantly upregulated. Based on their demonstrated utility in establishing an immune response pathway, these PCR assays should be valuable additions to our swine immune toolkit.


Assuntos
Doenças dos Suínos/parasitologia , Células Th1/imunologia , Toxoplasma/imunologia , Toxoplasmose Animal/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/imunologia , Sequência de Bases , Antígenos CD28/genética , Antígenos CD28/imunologia , Antígenos CD8/genética , Antígenos CD8/imunologia , Dados de Sequência Molecular , Fator 88 de Diferenciação Mieloide , RNA de Protozoário/química , RNA de Protozoário/genética , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Proteínas Repressoras/genética , Proteínas Repressoras/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Análise de Sequência de DNA , Suínos , Doenças dos Suínos/imunologia , Proteínas com Domínio T/genética , Proteínas com Domínio T/imunologia , Toxoplasma/genética , Toxoplasmose Animal/parasitologia
20.
Tuberculosis (Edinb) ; 94(4): 374-82, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24836563

RESUMO

Tuberculosis is an important health concern for Asian elephant (Elephas maximus) populations worldwide, however, mechanisms underlying susceptibility to Mycobacterium tuberculosis are unknown. Proliferative responses assessed via brominated uridine incorporation and cytokine expression measured by real-time RT-PCR were evaluated in peripheral blood mononuclear cell (PBMC) cultures from 8 tuberculosis negative and 8 positive Asian elephants. Cultures were stimulated with Mycobacterium bovis purified protein derivative (PPD-B), M. tuberculosis culture filtrate protein (CFP)-10, and Mycobacterium avium PPD (PPD-A). Following stimulation with PPD-B, proliferation was higher (α = 0.005) in positive samples; no significant differences were detected following CFP-10 or PPD-A stimulation. Tumor necrosis factor (TNF)-α, interleukin (IL)-12, and interferon (IFN)-γ expression was greater in samples from positive elephants following stimulation with PPD-B (α = 0.025) and CFP-10 (α = 0.025 TNF-α and IL-12; α = 0.005 IFN-γ). Stimulation with PPD-A also produced enhanced IL-12 expression in positive samples (α = 0.025). Findings suggested that differences in immune cell function exist between tuberculosis positive and negative elephants. Proliferative responses and expression of TNF-α, IL-12, and IFN-γ in response to stimulation with PPD-B and CFP-10 differ between tuberculosis positive and negative elephants, suggesting these parameters may be important to tuberculosis immunopathogenesis in this species.


Assuntos
Elefantes/imunologia , Tuberculose/imunologia , Tuberculose/veterinária , Animais , Proteínas de Bactérias/imunologia , Proliferação de Células , Células Cultivadas , Citocinas/biossíntese , Citocinas/genética , Suscetibilidade a Doenças , Elefantes/microbiologia , Feminino , Regulação da Expressão Gênica/imunologia , Imunidade Celular , Leucócitos Mononucleares/imunologia , Masculino , RNA Mensageiro/genética , Tuberculina/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA