Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Virology ; 443(1): 99-105, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23725691

RESUMO

African swine fever virus (ASFV) causes an acute haemorrhagic disease of domestic pigs against which there is no effective vaccine. The attenuated ASFV strain OUR T88/3 has been shown previously to protect vaccinated pigs against challenge with some virulent strains including OUR T88/1. Two genes, DP71L and DP96R were deleted from the OUR T88/3 genome to create recombinant virus OUR T88/3ΔDP2. Deletion of these genes from virulent viruses has previously been shown to reduce ASFV virulence in domestic pigs. Groups of 6 pigs were immunised with deletion virus OUR T88/3ΔDP2 or parental virus OUR T88/3 and challenged with virulent OUR T88/1 virus. Four pigs (66%) were protected by inoculation with the deletion virus OUR T88/3ΔDP2 compared to 100% protection with the parental virus OUR T88/3. Thus the deletion of the two genes DP71L and DP96R from OUR T88/3 strain reduced its ability to protect pigs against challenge with virulent virus.


Assuntos
Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/imunologia , Febre Suína Africana/prevenção & controle , Deleção de Sequência , Vacinas Virais/imunologia , Fatores de Virulência/genética , Fatores de Virulência/imunologia , Febre Suína Africana/imunologia , Febre Suína Africana/patologia , Febre Suína Africana/virologia , Animais , Índice de Gravidade de Doença , Baço/virologia , Suínos , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Viremia/imunologia , Viremia/prevenção & controle , Virulência
2.
PLoS One ; 8(4): e60574, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23593251

RESUMO

The protective efficacy of recombinant vaccines expressing serotype 8 bluetongue virus (BTV-8) capsid proteins was tested in a mouse model. The recombinant vaccines comprised plasmid DNA or Modified Vaccinia Ankara viruses encoding BTV VP2, VP5 or VP7 proteins. These constructs were administered alone or in combination using either a homologous prime boost vaccination regime (rMVA/rMVA) or a heterologous vaccination regime (DNA/rMVA). The DNA/rMVA or rMVA/rMVA prime-boost were administered at a three week interval and all of the animals that received VP2 generated neutralising antibodies. The vaccinated and non-vaccinated-control mice were subsequently challenged with a lethal dose of BTV-8. Mice vaccinated with VP7 alone were not protected. However, mice vaccinated with DNA/rMVA or rMVA/rMVA expressing VP2, VP5 and VP7 or VP2 alone were all protected.


Assuntos
Vírus Bluetongue/imunologia , Bluetongue/imunologia , Bluetongue/prevenção & controle , Proteínas do Capsídeo/imunologia , Receptor de Interferon alfa e beta/deficiência , Vacinas de DNA/imunologia , Vacinas Sintéticas/imunologia , Animais , Anticorpos Neutralizantes/sangue , Bluetongue/sangue , Bluetongue/virologia , Galinhas , Chlorocebus aethiops , Camundongos , Plasmídeos/imunologia , RNA Viral/sangue , Receptor de Interferon alfa e beta/metabolismo , Vacinação , Células Vero , Viremia/imunologia , Viremia/prevenção & controle , Viremia/virologia
3.
Virus Res ; 173(1): 110-21, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23201582

RESUMO

African swine fever virus (ASFV) infection usually results in an acute haemorrhagic disease with a mortality rate approaching 100% in domestic pigs. However, pigs can survive infection with less-virulent isolates of ASFV and may become chronically infected. Surviving animals are resistant to challenge with homologous or, in some cases, closely related isolates of the virus indicating that pigs can develop protective immunity against ASFV. During asymptomatic, non-virulent ASFV infections natural killer cell activity increases in pigs, suggesting this cell type plays a role in ASFV immunity. Furthermore, depletion of CD8(+) lymphocytes from ASFV immune pigs demolishes protective immunity against related virulent viruses. This suggests that ASFV specific antibody alone is not sufficient for protection against ASFV infection and that there is an important role for the CD8(+) lymphocyte subset in ASFV protective immunity. These results were supported by DNA immunization studies, demonstrating a correlation between the protection afforded against lethal challenge and the detection of a large number of vaccine-induced antigen-specific CD8(+) T-cells. Peripheral blood mononuclear cells (PBMCs) from ASF immune pigs protected from clinical disease show higher proportions of ASFV specific CD4(+)CD8(high+) double positive cytotoxic T cells than PBMCs from ASF immune but clinically diseased pig. The frequency of ASFV specific IFNγ producing T cells induced by immunization correlates to the degree of protection from ASFV challenge, and this may prove to be a useful indicator of any potential cross-protection against heterologous ASFV isolates.


Assuntos
Vírus da Febre Suína Africana/imunologia , Febre Suína Africana/imunologia , Imunidade Celular , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , DNA Viral/química , DNA Viral/genética , Interferon gama/metabolismo , Células Matadoras Naturais/imunologia , Dados de Sequência Molecular , Análise de Sequência de DNA , Suínos , Linfócitos T Citotóxicos/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia
4.
Vet Res ; 43: 40, 2012 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-22546071

RESUMO

Bluetongue virus (BTV) is a double stranded (ds) RNA virus (genus Orbivirus; family Reoviridae), which is considered capable of infecting all species of domestic and wild ruminants, although clinical signs are seen mostly in sheep. BTV is arthropod-borne ("arbovirus") and able to productively infect and replicate in many different cell types of both insects and mammalian hosts. Although the organ and cellular tropism of BTV in ruminants has been the subject of several studies, many aspects of its pathogenesis are still poorly understood, partly because of inherent problems in distinguishing between "virus replication" and "virus presence".BTV replication and organ tropism were studied in a wide range of infected sheep tissues, by immuno-fluorescence-labeling of non-structural or structural proteins (NS2 or VP7 and core proteins, respectively) using confocal microscopy to distinguish between virus presence and replication. These results are compared to gross and microscopic pathological findings in selected organs from infected sheep. Replication was demonstrated in two major cell types: vascular endothelial cells, and agranular leukocytes which morphologically resemble lymphocytes, monocytes/macrophages and/or dendritic cells. Two organs (the skin and tonsils) were shown to support relatively high levels of BTV replication, although they have not previously been proposed as important replication sites during BTV infection. The high level of BTV replication in the skin is thought to be of major significance for the pathogenesis and transmission of BTV (via biting insects) and a refinement of our current model of BTV pathogenesis is discussed.


Assuntos
Vírus Bluetongue/fisiologia , Bluetongue/virologia , Ceratopogonidae/fisiologia , Pele/virologia , Animais , Vírus Bluetongue/genética , Vírus Bluetongue/isolamento & purificação , Comportamento Alimentar , Cadeia Alimentar , Imuno-Histoquímica/veterinária , Inflamação/veterinária , Inflamação/virologia , Microscopia Confocal/veterinária , Especificidade de Órgãos , Ovinos , Proteínas do Core Viral/metabolismo , Proteínas não Estruturais Virais/metabolismo
5.
PLoS One ; 6(3): e17545, 2011 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-21423801

RESUMO

Bluetongue virus (BTV) and epizootic haemorrhagic disease virus (EHDV) are related orbiviruses, transmitted between their ruminant hosts primarily by certain haematophagous midge vectors (Culicoides spp.). The larger of the BTV outer-capsid proteins, 'VP2', can be cleaved by proteases (including trypsin or chymotrypsin), forming infectious subviral particles (ISVP) which have enhanced infectivity for adult Culicoides, or KC cells (a cell-line derived from C. sonorensis). We demonstrate that VP2 present on purified virus particles from 3 different BTV strains can also be cleaved by treatment with saliva from adult Culicoides. The saliva proteins from C. sonorensis (a competent BTV vector), cleaved BTV-VP2 more efficiently than those from C. nubeculosus (a less competent/non-vector species). Electrophoresis and mass spectrometry identified a trypsin-like protease in C. sonorensis saliva, which was significantly reduced or absent from C. nubeculosus saliva. Incubating purified BTV-1 with C. sonorensis saliva proteins also increased their infectivity for KC cells ∼10 fold, while infectivity for BHK cells was reduced by 2-6 fold. Treatment of an 'eastern' strain of EHDV-2 with saliva proteins of either C. sonorensis or C. nubeculosus cleaved VP2, but a 'western' strain of EHDV-2 remained unmodified. These results indicate that temperature, strain of virus and protein composition of Culicoides saliva (particularly its protease content which is dependent upon vector species), can all play a significant role in the efficiency of VP2 cleavage, influencing virus infectivity. Saliva of several other arthropod species has previously been shown to increase transmission, infectivity and virulence of certain arboviruses, by modulating and/or suppressing the mammalian immune response. The findings presented here, however, demonstrate a novel mechanism by which proteases in Culicoides saliva can also directly modify the orbivirus particle structure, leading to increased infectivity specifically for Culicoides cells and, in turn, efficiency of transmission to the insect vector.


Assuntos
Vírus Bluetongue/patogenicidade , Bluetongue/virologia , Ceratopogonidae/metabolismo , Insetos Vetores/metabolismo , Proteínas e Peptídeos Salivares/metabolismo , Vírion/química , Vírion/patogenicidade , Animais , Vírus Bluetongue/efeitos dos fármacos , Linhagem Celular , Ceratopogonidae/efeitos dos fármacos , Quimotripsina/metabolismo , Eletroforese em Gel de Poliacrilamida , Peso Molecular , Inibidores de Proteases/farmacologia , Saliva/efeitos dos fármacos , Saliva/metabolismo , Ovinos , Temperatura , Tripsina/metabolismo , Proteínas Virais/metabolismo , Vírion/efeitos dos fármacos
6.
Vaccine ; 28(5): 1397-403, 2010 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-19895921

RESUMO

The spread of bluetongue virus (BTV) is most successfully controlled by vaccination of susceptible ruminant populations. Currently two different types of BTV vaccines are used for this purpose; inactivated, mostly monovalent vaccine formulations and modified live virus vaccines (MLVs). Clinical signs and viraemia in Dorset Poll sheep vaccinated with BTV-4 and BTV-16 MLVs or inoculated with homogenates of midges (C. sonorensis and C. nubeculosus) previously infected with BTV-4 MLV are presented. All sheep vaccinated with the two MLVs mounted an infectious viraemia lasting for a minimum of 9 up to 23 days post vaccination and developed a range of clinical signs associated with BTV infection. Peak viraemia titres recorded in individual sheep ranged from 3.5 to 6.83 log(10)TCID(50)/ml indicating a high potential for infection of vector insects and onward transmission. The implications of these results are discussed with reference to the current outbreaks of BTV occurring in northern Europe and in relation to the future development of vaccines for this virus.


Assuntos
Vírus Bluetongue/imunologia , Bluetongue/imunologia , Vacinação , Vacinas Virais/farmacologia , Viremia/imunologia , Animais , Bluetongue/prevenção & controle , Bluetongue/virologia , Ceratopogonidae/virologia , Feminino , Insetos Vetores/virologia , Masculino , Ovinos , Fatores de Tempo , Vacinas de Produtos Inativados/imunologia , Vacinas de Produtos Inativados/farmacologia , Vacinas Virais/imunologia
7.
J Virol ; 83(17): 8789-99, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19553336

RESUMO

Bluetongue virus (BTV) is the etiological agent of bluetongue, a hemorrhagic disease of ruminants (particularly sheep), which causes important economic losses around the world. BTV is transmitted primarily via the bites of infected midges, which inject the virus into the ruminant's skin during blood feeding. The virus initially replicates in the draining lymph node and then disseminates to secondary organs where it induces edema, hemorrhages, and necrosis. In this study, we show that ovine conventional dendritic cells (cDCs) are the primary targets of BTV that contribute to the primary dissemination of BTV from the skin to draining lymph nodes. Lymph cDCs support BTV RNA and protein synthesis, as well as the production of infectious virus belonging to several different BTV serotypes, regardless of their level of attenuation. Afferent lymph cell subsets, other than cDCs, showed only marginal levels of BTV protein expression. BTV infection provoked a massive recruitment of cDCs to the sheep skin and afferent lymph, providing cellular targets for infection. Although BTV productively infects cDCs, no negative impact on their physiology was detected. Indeed, BTV infection and protein expression in cDCs enhanced their survival rate. Several serotypes of BTV stimulated the surface expression of the CD80 and CD86 costimulatory molecules on cDCs as well as the mRNA synthesis of cytokines involved in inflammation and immunity, i.e., interleukin-12 (IL-12), IL-1beta, and IL-6. BTV-infected cDCs stimulated antigen-specific CD4 and CD8 proliferation as well as gamma interferon production. BTV initially targets cDCs while preserving their functional properties, reflecting the optimal adaptation of the virus to its host cells for its first spread.


Assuntos
Vírus Bluetongue/crescimento & desenvolvimento , Células Dendríticas/virologia , Linfa/virologia , Pele/virologia , Animais , Antígeno B7-1/biossíntese , Antígeno B7-2/biossíntese , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Feminino , Interferon gama/metabolismo , Interleucinas/biossíntese , RNA Viral/biossíntese , Ovinos , Proteínas Virais/biossíntese
8.
J Virol ; 83(5): 2310-20, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19109387

RESUMO

Increasing evidence points to the importance of the interferon (IFN) response in determining the host range and virulence of African swine fever virus (ASFV). Infection with attenuated strains of ASFV leads to the upregulation of genes controlled by IFN pathways, including myxovirus resistance (Mx) genes that are potent effectors of the antiviral state. Mx gene products are known to inhibit the replication of many negative-sense single-stranded RNA viruses, as well as double-stranded RNA viruses, positive-sense single-stranded RNA viruses, and the reverse-transcribing DNA virus hepatitis B virus. Here, we provide data that extend the known range of viruses inhibited by Mx to include the large double-stranded DNA viruses. Stably transfected Vero cells expressing human MxA protein did not support ASFV plaque formation, and virus replication in these cells was reduced 100-fold compared with that in control cells. In contrast, ASFV replication in cells expressing MxB protein or a mutant MxA protein was similar to that in control Vero cells. There was a drastic reduction in ASFV late protein synthesis in MxA-expressing cells, correlating with the results of previous work on the effect of IFN on viral replication. Strikingly, the inhibition of ASFV replication was linked to the recruitment of MxA protein to perinuclear viral assembly sites, where the protein surrounded the virus factories. Interactions between ASFV and MxA were similar to those seen between MxA and different RNA viruses, suggesting a common inhibitory mechanism.


Assuntos
Vírus da Febre Suína Africana/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Replicação Viral , Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/fisiologia , Animais , Chlorocebus aethiops , DNA Viral/genética , Humanos , Proteínas de Resistência a Myxovirus , Biossíntese de Proteínas , Suínos , Transcrição Gênica , Células Vero , Montagem de Vírus
9.
J Virol ; 80(22): 11385-92, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16956944

RESUMO

Viral interference with secretory cargo is a common mechanism for pathogen immune evasion. Selective down regulation of critical immune system molecules such as major histocompatibility complex (MHC) proteins enables pathogens to mask themselves from their host. African swine fever virus (ASFV) disrupts the trans-Golgi network (TGN) by altering the localization of TGN46, an organelle marker for the distal secretory pathway. Reorganization of membrane transport components may provide a mechanism whereby ASFV can disrupt the correct secretion and/or cell surface expression of host proteins. In the study reported here, we used the tsO45 temperature-sensitive mutant of the G protein of vesicular stomatitis virus to show that ASFV significantly reduces the rate at which the protein is delivered to the plasma membrane. This is linked to a general reorganization of the secretory pathway during infection and a specific, microtubule-dependent disruption of structural components of the TGN. Golgin p230 and TGN46 are separated into distinct vesicles, whereupon TGN46 is depleted. These data suggest that disruption of the TGN by ASFV can slow membrane traffic during viral infection. This may be functionally important because infection of macrophages with virulent isolates of ASFV increased the expression of MHC class I genes, but there was no parallel increase in MHC class I molecule delivery to the plasma membrane.


Assuntos
Vírus da Febre Suína Africana/fisiologia , Membrana Celular/metabolismo , Proteínas de Membrana/metabolismo , Microtúbulos/virologia , Rede trans-Golgi/virologia , Animais , Chlorocebus aethiops , Microtúbulos/fisiologia , Células Vero , Rede trans-Golgi/ultraestrutura
10.
Virus Res ; 121(2): 223-8, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16934904

RESUMO

To identify foot-and-mouth disease virus (FMDV) specific T-cell epitopes within the non-structural protein 3D in swine, pentadecapeptides were tested in proliferation and Interferon-gamma ELISPOT assays using lymphocytes from two strains of inbred miniature pigs (c/c and d/d haplotype) experimentally infected with FMDV. Lymphocytes of c/c pigs recognized peptides from three different regions in 3D, d/d lymphocytes recognized peptides from two regions, one of them being adjacent to an epitope of c/c pigs and comprising amino acid residues 346-370. Analyses of the response of d/d lymphocytes against peptides representing the structural protein 1A revealed another novel T-cell epitope. Investigation of the phenotype of responding lymphocytes showed a response of CD4(+)CD8(+)MHC-class-II(+) cells, identifying them as activated T-helper cells. This is the first report on FMDV specific T-cell epitopes recognized by swine leukocyte antigen (SLA) inbred swine and provides information useful for the design of novel vaccines against FMDV.


Assuntos
Epitopos de Linfócito T/imunologia , Vírus da Febre Aftosa/imunologia , Febre Aftosa/imunologia , Linfócitos T/imunologia , Sequência de Aminoácidos , Animais , Animais Endogâmicos , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Haplótipos , Antígenos de Histocompatibilidade/imunologia , Memória Imunológica , Interferon gama/análise , Leucócitos Mononucleares/imunologia , Ativação Linfocitária , Dados de Sequência Molecular , Peptídeos/genética , Peptídeos/imunologia , Suínos , Porco Miniatura , Especificidade do Receptor de Antígeno de Linfócitos T , Linfócitos T Auxiliares-Indutores/imunologia , Proteínas não Estruturais Virais/síntese química , Proteínas não Estruturais Virais/imunologia
11.
Immunogenetics ; 58(5-6): 481-6, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16738944

RESUMO

Human killer immunoglobulin-like receptors (KIR) are expressed on natural killer (NK) cells and are involved in their immunoreactivity. While KIR with a long cytoplasmic tail deliver an inhibitory signal when bound to their respective major histocompatibility complex class I ligands, KIR with a short cytoplasmic tail can activate NK responses. The expansion of the KIR gene family originally appeared to be a phenomenon restricted to primates (human, apes, and monkeys) in comparison to rodents, which via convergent evolution have numerous C-type lectin-like Ly49 molecules that function analogously. Further studies have shown that multiple KIR are also present in cow and horse. In this study, we have identified by comparative genomics the first and possibly only KIR gene, named KIR2DL1, in the domesticated pig (Sus scrofa) allowing further evolutionary comparisons to be made. It encodes a protein with two extracellular immunoglobulin domains (D0 + D2), and a long cytoplasmic tail containing two inhibitory motifs. We have mapped the pig KIR2DL1 gene to chromosome 6q. Flanked by LILRa, LILRb, and LILRc, members of the leukocyte immunoglobulin-like receptor (LILR) family, on the centromeric end, and FCAR, NCR1, NALP7, NALP2, and GP6 on the telomeric end, pig demonstrates conservation of synteny with the human leukocyte receptor complex (LRC). Both the porcine KIR and LILR genes have diverged sufficiently to no longer be clearly orthologous with known human LRC family members.


Assuntos
Leucócitos/imunologia , Receptores Imunológicos/classificação , Receptores Imunológicos/genética , Sus scrofa/genética , Sequência de Aminoácidos , Animais , Cromossomos/genética , Dados de Sequência Molecular , Filogenia , Receptores KIR , Receptores KIR2DL1 , Sus scrofa/imunologia
12.
Vet Immunol Immunopathol ; 110(3-4): 279-92, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16325923

RESUMO

In this study we have used the expression of perforin to characterize subsets of porcine cytotoxic lymphocytes. Perforin positive lymphocytes expressed both CD2 and CD8alpha, most were small dense lymphocytes (SDL) and up to 90% were CD3 negative. However, the numbers of perforin positive T-cells increased with the age of the animal and their populations increased after specific antigen stimulation in vitro. The remaining perforin positive lymphocytes were large and granular and contained more CD3+CD5+CD6+ T-cells (-40%) of which a substantial proportion also co-expressed CD4. Perforin was expressed in subpopulations of both CD8alphaalpha and CD8alphabeta lymphocytes, but was not expressed in gammadelta T-cells or monocyte/macrophages. The perforin positive CD3- subset was phenotypically homogeneous and defined as CD5-CD6-CD8beta-CD16+CD11b+. This population had NK activity and expressed mRNA for the NK receptor NKG2D, and adaptors DAP10 and DAP12. Perforin positive T-cells (CD3+) could be divided into at least three subsets. The first subset was CD4-CD5+CD6+CD11b-CD16- most were small dense lymphocytes with cytotoxic T-cell activity but not all expressed CD8beta. The second subset was mainly observed in the large granular lymphocytes. Their phenotype was CD4+CD5+CD6+CD8beta+CD16-CD11b- and also showed functional CTL activity. Thus not all of double positive T-cells are memory helper T-cells. The third subset did not express the T-cell co-receptor CD6, but up to half of them expressed another T-cell co-receptor CD5. The majority of this subset expressed CD11b and CD16, thus the third perforin positive T-cell subset was CD3+CD4-CD5+CD6-CD8beta+/-CD11b+CD16+, and possessed MHC-unrestricted cytotoxicity and LAK activity.


Assuntos
Células Matadoras Naturais/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Glicoproteínas de Membrana/metabolismo , Suínos/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Animais , Antígenos CD/metabolismo , Regulação da Expressão Gênica , Células Matadoras Naturais/metabolismo , Tecido Linfoide/metabolismo , Glicoproteínas de Membrana/genética , Perforina , Fenótipo , Proteínas Citotóxicas Formadoras de Poros , Subpopulações de Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...