Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
2.
Viruses ; 9(11)2017 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-29149056

RESUMO

Intrinsic factors of the innate immune system include the apolipoprotein B editing enzyme catalytic polypeptide-like 3 (APOBEC3) protein family. APOBEC3 inhibits replication of different virus families by cytosine deamination of viral DNA and a not fully characterized cytosine deamination-independent mechanism. Sheep are susceptible to small ruminant lentivirus (SRLVs) infection and contain three APOBEC3 genes encoding four proteins (A3Z1, Z2, Z3 and Z2-Z3) with yet not deeply described antiviral properties. Using sheep blood monocytes and in vitro-derived macrophages, we found that A3Z1 expression is associated with lower viral replication in this cellular type. A3Z1 transcripts may also contain spliced variants (A3Z1Tr) lacking the cytidine deaminase motif. A3Z1 exogenous expression in fully permissive fibroblast-like cells restricted SRLVs infection while A3Z1Tr allowed infection. A3Z1Tr was induced after SRLVs infection or stimulation of blood-derived macrophages with interferon gamma (IFN-γ). Interaction between truncated isoform and native A3Z1 protein was detected as well as incorporation of both proteins into virions. A3Z1 and A3Z1Tr interacted with SRLVs Vif, but this interaction was not associated with degradative properties. Similar A3Z1 truncated isoforms were also present in human and monkey cells suggesting a conserved alternative splicing regulation in primates. A3Z1-mediated retroviral restriction could be constrained by different means, including gene expression and specific alternative splicing regulation, leading to truncated protein isoforms lacking a cytidine-deaminase motif.


Assuntos
Citosina Desaminase/genética , Lentivirus/fisiologia , Replicação Viral , Processamento Alternativo/genética , Animais , Citosina Desaminase/química , Citosina Desaminase/metabolismo , Regulação da Expressão Gênica , Haplorrinos , Humanos , Interferon gama/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Motivos de Nucleotídeos/genética , Isoformas de Proteínas/genética , Ovinos
3.
Vet Res ; 47: 1, 2016 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-26738942

RESUMO

Small ruminant lentivirus (SRLV) infection causes losses in the small ruminant industry due to reduced animal production and increased replacement rates. Infection of wild ruminants in close contact with infected domestic animals has been proposed to play a role in SRLV epidemiology, but studies are limited and mostly involve hybrids between wild and domestic animals. In this study, SRLV seropositive red deer, roe deer and mouflon were detected through modified ELISA tests, but virus was not successfully amplified using a set of different PCRs. Apparent restriction of SRLV infection in cervids was not related to the presence of neutralizing antibodies. In vitro cultured skin fibroblastic cells from red deer and fallow deer were permissive to the SRLV entry and integration, but produced low quantities of virus. SRLV got rapidly adapted in vitro to blood-derived macrophages and skin fibroblastic cells from red deer but not from fallow deer. Thus, although direct detection of virus was not successfully achieved in vivo, these findings show the potential susceptibility of wild ruminants to SRLV infection in the case of red deer and, on the other hand, an in vivo SRLV restriction in fallow deer. Altogether these results may highlight the importance of surveilling and controlling SRLV infection in domestic as well as in wild ruminants sharing pasture areas, and may provide new natural tools to control SRLV spread in sheep and goats.


Assuntos
Cervos , Fibroblastos/virologia , Infecções por Lentivirus/veterinária , Lentivirus/fisiologia , Doenças dos Ovinos/virologia , Carneiro Doméstico , Replicação Viral/fisiologia , Animais , Animais Selvagens , Anticorpos Antivirais/sangue , Especificidade de Anticorpos , Ensaio de Imunoadsorção Enzimática , Infecções por Lentivirus/sangue , Infecções por Lentivirus/transmissão , Ovinos , Doenças dos Ovinos/sangue , Doenças dos Ovinos/transmissão , Internalização do Vírus
4.
Vet J ; 204(1): 88-93, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25766510

RESUMO

The major challenges in diagnosing small ruminant lentivirus (SRLV) infection include early detection and genotyping of strains of epidemiological interest. A longitudinal study was carried out in Rasa Aragonesa sheep experimentally infected with viral strains of genotypes A or B from Spanish neurological and arthritic SRLV outbreaks, respectively. Sera were tested with two commercial ELISAs, three based on specific peptides and a novel combined peptide ELISA. Three different PCR assays were used to further assess infection status. The kinetics of anti-viral antibody responses were variable, with early diagnosis dependent on the type of ELISA used. Peptide epitopes of SRLV genotypes A and B combined in the same ELISA well enhanced the overall detection rate, whereas single peptides were useful for genotyping the infecting strain (A vs. B). The results of the study suggest that a combined peptide ELISA can be used for serological diagnosis of SRLV infection, with single peptide ELISAs useful for subsequent serotyping.


Assuntos
Ensaio de Imunoadsorção Enzimática/veterinária , Infecções por Lentivirus/veterinária , Lentivirus/genética , Peptídeos/química , Doenças dos Ovinos/virologia , Animais , Anticorpos Antivirais/sangue , Genótipo , Lentivirus/classificação , Infecções por Lentivirus/diagnóstico , Infecções por Lentivirus/virologia , Masculino , Reação em Cadeia da Polimerase/veterinária , Testes Sorológicos , Ovinos , Doenças dos Ovinos/diagnóstico
5.
Antimicrob Agents Chemother ; 58(11): 6660-7, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25155589

RESUMO

A mouse model was developed for in vivo monitoring of infection and the effect of antimicrobial treatment against Staphylococcus aureus biofilms, using the [(18)F]fluoro-deoxyglucose-MicroPET ([(18)F]FDG-MicroPET) image technique. In the model, sealed Vialon catheters were briefly precolonized with S. aureus strains ATCC 15981 or V329, which differ in cytotoxic properties and biofilm matrix composition. After subcutaneous implantation of catheters in mice, the S. aureus strain differences found in bacterial counts and the inflammatory reaction triggered were detected by the regular bacteriological and histological procedures and also by [(18)F]FDG-MicroPET image signal intensity determinations in the infection area and regional lymph node. Moreover, [(18)F]FDG-MicroPET imaging allowed the monitoring of the rifampin treatment effect, identifying the periods of controlled infection and those of reactivated infection due to the appearance of bacteria naturally resistant to rifampin. Overall, the mouse model developed may be useful for noninvasive in vivo determinations in studies on S. aureus biofilm infections and assessment of new therapeutic approaches.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Rifampina/farmacologia , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/patogenicidade , Animais , Aderência Bacteriana , Cateteres de Demora , Modelos Animais de Doenças , Feminino , Fluordesoxiglucose F18/metabolismo , Camundongos , Testes de Sensibilidade Microbiana , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/efeitos dos fármacos
6.
Vet J ; 202(2): 323-8, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25168719

RESUMO

Production and excretion of small ruminant lentiviruses (SRLVs) varies with the stage of the host reproductive cycle, suggesting hormonal involvement in this variation. Stress may also affect viral expression. To determine if hormones affect SRLV transcriptional activity, the expression of green fluorescent protein (GFP) driven by the promoters in the U3-cap region of the long terminal repeats (LTRs) of different strains of SRLV was assessed in cell culture. High concentrations of steroids (progesterone, cortisol and dehydroepiandrosterone) inhibited expression of GFP driven by SRLV promoters. This effect decreased in a dose-dependent manner with decreasing concentrations of steroids. In some strains, physiological concentrations of cortisol or dehydroepiandrosterone (DHEA) induced the expression of GFP above the baseline. There was strain variation in sensitivity to hormones, but this differed for different hormones. The presence of deletions and a 43 base repeat in the U3 region upstream of the TATA box of the LTR made strain EV1 less sensitive to DHEA. However, no clear tendencies or patterns were observed when comparing strains of different genotypes and/or subtypes, or those triggering different forms of disease.


Assuntos
Desidroepiandrosterona/metabolismo , Regulação Viral da Expressão Gênica , Hidrocortisona/metabolismo , Progesterona/metabolismo , Regiões Promotoras Genéticas , Sequências Repetidas Terminais , Vírus Visna-Maedi/genética , Animais , Sequência de Bases , Proteínas de Fluorescência Verde/genética , Dados de Sequência Molecular , Plasmídeos/genética , Pneumonia Intersticial Progressiva dos Ovinos/virologia , Ovinos , Doenças dos Ovinos/virologia , Visna/virologia , Vírus Visna-Maedi/metabolismo
7.
Vet Res ; 44: 83, 2013 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-24070317

RESUMO

Small ruminant lentiviruses (SRLV) infect the monocyte/macrophage lineage inducing a long-lasting infection affecting body condition, production and welfare of sheep and goats all over the world. Macrophages play a pivotal role on the host's innate and adaptative immune responses against parasites by becoming differentially activated. Macrophage heterogeneity can tentatively be classified into classically differentiated macrophages (M1) through stimulation with IFN-γ displaying an inflammatory profile, or can be alternatively differentiated by stimulation with IL-4/IL-13 into M2 macrophages with homeostatic functions. Since infection by SRLV can modulate macrophage functions we explored here whether ovine and caprine macrophages can be segregated into M1 and M2 populations and whether this differential polarization represents differential susceptibility to SRLV infection. We found that like in human and mouse systems, ovine and caprine macrophages can be differentiated with particular stimuli into M1/M2 subpopulations displaying specific markers. In addition, small ruminant macrophages are plastic since M1 differentiated macrophages can express M2 markers when the stimulus changes from IFN-γ to IL-4. SRLV replication was restricted in M1 macrophages and increased in M2 differentiated macrophages respectively according to viral production. Identification of the infection pathways in macrophage populations may provide new targets for eliciting appropriate immune responses against SRLV infection.


Assuntos
Citocinas/genética , Regulação da Expressão Gênica , Doenças das Cabras/imunologia , Infecções por Lentivirus/veterinária , Lentivirus/fisiologia , Macrófagos/imunologia , Doenças dos Ovinos/imunologia , Animais , Células CHO , Cricetulus , Citocinas/metabolismo , Marcadores Genéticos , Doenças das Cabras/virologia , Cabras , Células HEK293 , Humanos , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/virologia , Lipopolissacarídeos , Macrófagos/citologia , Macrófagos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Ovinos , Doenças dos Ovinos/virologia
8.
Viruses ; 5(8): 1948-63, 2013 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-23917352

RESUMO

Multisystemic disease caused by Small Ruminant Lentiviruses (SRLV) in sheep and goats leads to production losses, to the detriment of animal health and welfare. This, together with the lack of treatments, has triggered interest in exploring different strategies of immunization to control the widely spread SRLV infection and, also, to provide a useful model for HIV vaccines. These strategies involve inactivated whole virus, subunit vaccines, DNA encoding viral proteins in the presence or absence of plasmids encoding immunological adjuvants and naturally or artificially attenuated viruses. In this review, we revisit, comprehensively, the immunization strategies against SRLV and analyze this double edged tool individually, as it may contribute to either controlling or enhancing virus replication and/or disease.


Assuntos
Doenças das Cabras/prevenção & controle , Infecções por Lentivirus/veterinária , Lentivirus/imunologia , Doenças dos Ovinos/prevenção & controle , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia , Animais , Doenças das Cabras/imunologia , Doenças das Cabras/patologia , Cabras , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/patologia , Infecções por Lentivirus/prevenção & controle , Ovinos , Doenças dos Ovinos/imunologia , Doenças dos Ovinos/patologia , Vacinação/efeitos adversos , Vacinação/métodos , Vacinas Virais/efeitos adversos
9.
Viruses ; 5(4): 1175-207, 2013 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-23611847

RESUMO

Small ruminant lentiviruses (SRLV) cause a multisystemic chronic disease affecting animal production and welfare. SRLV infections are spread across the world with the exception of Iceland. Success in controlling SRLV spread depends largely on the use of appropriate diagnostic tools, but the existence of a high genetic/antigenic variability among these viruses, the fluctuant levels of antibody against them and the low viral loads found in infected individuals hamper the diagnostic efficacy. SRLV have a marked in vivo tropism towards the monocyte/macrophage lineage and attempts have been made to identify the genome regions involved in tropism, with two main candidates, the LTR and env gene, since LTR contains primer binding sites for viral replication and the env-encoded protein (SU ENV), which mediates the binding of the virus to the host's cell and has hypervariable regions to escape the humoral immune response. Once inside the host cell, innate immunity may interfere with SRLV replication, but the virus develops counteraction mechanisms to escape, multiply and survive, creating a quasi-species and undergoing compartmentalization events. So far, the mechanisms of organ tropism involved in the development of different disease forms (neurological, arthritic, pulmonary and mammary) are unknown, but different alternatives are proposed. This is an overview of the current state of knowledge on SRLV genetic variability and its implications in tropism as well as in the development of alternative diagnostic assays.


Assuntos
Lentivirus/fisiologia , Ruminantes/virologia , Animais , Variação Genética , Lentivirus/classificação , Infecções por Lentivirus/diagnóstico , Filogenia , Tropismo Viral
10.
Comp Immunol Microbiol Infect Dis ; 36(4): 405-13, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23582860

RESUMO

Visna/Maedi virus (VMV) is a lentivirus that infects cells of the monocyte/macrophage lineage in sheep. Infection with VMV may lead to Visna/Maedi (VM) disease, which causes a multisystemic inflammatory disorder causing pneumonia, encephalitis, mastitis and arthritis. The role of ovine immune response genes in the development of VM disease is not fully understood. In this work, sheep of the Rasa Aragonesa breed were divided into two groups depending on the presence/absence of VM-characteristic clinical lesions in the aforementioned organs and the relative levels of candidate gene expression, including cytokines and innate immunity loci were measured by qPCR in the lung and udder. Sheep with lung lesions showed differential expression in five target genes: CCR5, TLR7, and TLR8 were up regulated and IL2 and TNFα down regulated. TNFα up regulation was detected in the udder.


Assuntos
Regulação Viral da Expressão Gênica/imunologia , Pulmão/virologia , Glândulas Mamárias Animais/virologia , Pneumonia Intersticial Progressiva dos Ovinos/imunologia , Vírus Visna-Maedi/imunologia , Animais , Feminino , Perfilação da Expressão Gênica/veterinária , Modelos Lineares , Pulmão/imunologia , Glândulas Mamárias Animais/imunologia , Pneumonia Intersticial Progressiva dos Ovinos/virologia , RNA Viral/química , RNA Viral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Ovinos , Vírus Visna-Maedi/genética
11.
Vet Res ; 43: 43, 2012 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-22591485

RESUMO

Thirty-one sheep naturally infected with small ruminant lentiviruses (SRLV) of known genotype (A or B), and clinically affected with neurological disease, pneumonia or arthritis were used to analyse mannose receptor (MR) expression (transcript levels) and proviral load in virus target tissues (lung, mammary gland, CNS and carpal joints). Control sheep were SRLV-seropositive asymptomatic (n = 3), seronegative (n = 3) or with chronic listeriosis, pseudotuberculosis or parasitic cysts (n = 1 in each case). MR expression and proviral load increased with the severity of lesions in most analyzed organs of the SRLV infected sheep and was detected in the affected tissue involved in the corresponding clinical disease (CNS, lung and carpal joint in neurological disease, pneumonia and arthritis animal groups, respectively). The increased MR expression appeared to be SRLV specific and may have a role in lentiviral pathogenesis.


Assuntos
Regulação da Expressão Gênica , Lectinas Tipo C/genética , Infecções por Lentivirus/veterinária , Lentivirus Ovinos-Caprinos/isolamento & purificação , Lectinas de Ligação a Manose/genética , Provírus/isolamento & purificação , Receptores de Superfície Celular/genética , Doenças dos Ovinos/genética , Carga Viral/veterinária , Animais , Artrite/genética , Artrite/veterinária , Artrite/virologia , Encefalite/genética , Encefalite/veterinária , Encefalite/virologia , Feminino , Lectinas Tipo C/metabolismo , Infecções por Lentivirus/genética , Infecções por Lentivirus/virologia , Masculino , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Especificidade de Órgãos , Pneumonia/genética , Pneumonia/veterinária , Pneumonia/virologia , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Receptores de Superfície Celular/metabolismo , Ovinos , Doenças dos Ovinos/virologia , Espanha
12.
Vet Res ; 43: 31, 2012 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-22515195

RESUMO

The Extradomain A from fibronectin (EDA) has an immunomodulatory role as fusion protein with viral and tumor antigens, but its effect when administered with bacteria has not been assessed. Here, we investigated the adjuvant effect of EDA in mice immunizations against Salmonella enterica subspecies enterica serovar Enteritidis (Salmonella Enteritidis). Since lipopolysaccharide (LPS) is a major virulence factor and the LPS O-polysaccharide (O-PS) is the immunodominant antigen in serological diagnostic tests, Salmonella mutants lacking O-PS (rough mutants) represent an interesting approach for developing new vaccines and diagnostic tests to differentiate infected and vaccinated animals (DIVA tests). Here, antigenic preparations (hot-saline extracts and formalin-inactivated bacterins) from two Salmonella Enteritidis rough mutants, carrying either intact (SEΔwaaL) or deep-defective (SEΔgal) LPS-Core, were used in combination with EDA. Biotinylated bacterins, in particular SEΔwaaL bacterin, decorated with EDAvidin (EDA and streptavidin fusion protein) improved the protection conferred by hot-saline or bacterins alone and prevented significantly the virulent infection at least to the levels of live attenuated rough mutants. These findings demonstrate the adjuvant effect of EDAvidin when administered with biotinylated bacterins from Salmonella Enteritidis lacking O-PS and the usefulness of BEDA-SEΔwaaL as non-live vaccine in the mouse model.


Assuntos
Vacinas Bacterianas/imunologia , Fibronectinas/imunologia , Lipopolissacarídeos/imunologia , Salmonella enteritidis/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/genética , Feminino , Fibronectinas/química , Lipopolissacarídeos/genética , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Terciária de Proteína , Salmonella enteritidis/genética , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia
13.
Vaccine ; 30(15): 2564-9, 2012 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-22326778

RESUMO

The development of effective vaccines against HIV-1 infection constitutes one of the major challenges in viral immunology. One of the protein candidates in vaccination against this virus is p24, since it is a conserved HIV antigen that has cytotoxic and helper T cell epitopes as well as B cell epitopes that may jointly confer enhanced protection against infection when used in immunization-challenge approaches. In this context, the adjuvant effect of EDA (used as EDAp24 fusion protein) and poly(I:C), as agonists of TLR4 and TLR3, respectively, was assessed in p24 immunizations using a recombinant Listeria monocytogenes HIV-1 Gag proteins (Lm-Gag, where p24 is the major antigen) for challenge in mice. Immunization with EDAp24 fusion protein together with poly(I:C) adjuvant induced a specific p24 IFN-γ production (Th1 profile) as well as protection against a Lm-Gag challenge, suggesting an additive or synergistic effect between both adjuvants. The combination of EDA (as a fusion protein with the antigen, which may favor antigen targeting to dendritic cells through TLR4) and poly(I:C) could thus be a good adjuvant candidate to enhance the immune response against HIV-1 proteins and its use may open new ways in vaccine investigations on this virus.


Assuntos
Adjuvantes Imunológicos/farmacologia , Fibronectinas/farmacologia , Produtos do Gene gag/imunologia , Proteína do Núcleo p24 do HIV/imunologia , Poli I-C/farmacologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/química , Animais , Antígenos Virais/imunologia , Feminino , Fibronectinas/administração & dosagem , Fibronectinas/química , Proteína do Núcleo p24 do HIV/administração & dosagem , Proteína do Núcleo p24 do HIV/química , Imunidade Celular/imunologia , Listeria monocytogenes/imunologia , Listeriose/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Poli I-C/administração & dosagem , Poli I-C/química , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/imunologia
14.
BMC Vet Res ; 8: 8, 2012 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-22281181

RESUMO

BACKGROUND: A central nervous system (CNS) disease outbreak caused by small ruminant lentiviruses (SRLV) has triggered interest in Spain due to the rapid onset of clinical signs and relevant production losses. In a previous study on this outbreak, the role of LTR in tropism was unclear and env encoded sequences, likely involved in tropism, were not investigated. This study aimed to analyze heterogeneity of SRLV Env regions--TM amino terminal and SU V4, C4 and V5 segments--in order to assess virus compartmentalization in CNS. RESULTS: Eight Visna (neurologically) affected sheep of the outbreak were used. Of the 350 clones obtained after PCR amplification, 142 corresponded to CNS samples (spinal cord and choroid plexus) and the remaining to mammary gland, blood cells, bronchoalveolar lavage cells and/or lung. The diversity of the env sequences from CNS was 11.1-16.1% between animals and 0.35-11.6% within each animal, except in one animal presenting two sequence types (30% diversity) in the CNS (one grouping with those of the outbreak), indicative of CNS virus sequence heterogeneity. Outbreak sequences were of genotype A, clustering per animal and compartmentalizing in the animal tissues. No CNS specific signature patterns were found. CONCLUSIONS: Bayesian approach inferences suggested that proviruses from broncoalveolar lavage cells and peripheral blood mononuclear cells represented the common ancestors (infecting viruses) in the animal and that neuroinvasion in the outbreak involved microevolution after initial infection with an A-type strain. This study demonstrates virus compartmentalization in the CNS and other body tissues in sheep presenting the neurological form of SRLV infection.


Assuntos
Vírus Visna-Maedi/genética , Visna/virologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Sistema Nervoso Central/virologia , Surtos de Doenças/veterinária , Genótipo , Glândulas Mamárias Animais/virologia , Dados de Sequência Molecular , Filogenia , Ovinos , Espanha/epidemiologia , Visna/epidemiologia
15.
Vet Res ; 42: 28, 2011 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-21314911

RESUMO

This study aims to characterize the mannose receptor (MR) gene in sheep and its role in ovine visna/maedi virus (VMV) infection. The deduced amino acid sequence of ovine MR was compatible with a transmembrane protein having a cysteine-rich ricin-type amino-terminal region, a fibronectin type II repeat, eight tandem C-type lectin carbohydrate-recognition domains (CRD), a transmembrane region, and a cytoplasmic carboxy-terminal tail. The ovine and bovine MR sequences were closer to each other compared to human or swine MR. Concanavalin A (ConA) inhibited VMV productive infection, which was restored by mannan totally in ovine skin fibroblasts (OSF) and partially in blood monocyte-derived macrophages (BMDM), suggesting the involvement of mannosylated residues of the VMV ENV protein in the process. ConA impaired also syncytium formation in OSF transfected with an ENV-encoding pN3-plasmid. MR transcripts were found in two common SRLV targets, BMDM and synovial membrane (GSM) cells, but not in OSF. Viral infection of BMDM and especially GSM cells was inhibited by mannan, strongly suggesting that in these cells the MR is an important route of infection involving VMV Env mannosylated residues. Thus, at least three patterns of viral entry into SRLV-target cells can be proposed, involving mainly MR in GSM cells (target in SRLV-induced arthritis), MR in addition to an alternative route in BMDM (target in SRLV infections), and an alternative route excluding MR in OSF (target in cell culture). Different routes of SRLV infection may thus coexist related to the involvement of MR differential expression.


Assuntos
Concanavalina A/farmacologia , Células Gigantes/virologia , Lectinas Tipo C/genética , Lectinas de Ligação a Manose/genética , Pneumonia Intersticial Progressiva dos Ovinos/imunologia , Receptores de Superfície Celular/genética , Vírus Visna-Maedi/fisiologia , Animais , Western Blotting/veterinária , Imuno-Histoquímica/veterinária , Lectinas Tipo C/química , Lectinas Tipo C/metabolismo , Macrófagos/imunologia , Receptor de Manose , Lectinas de Ligação a Manose/química , Lectinas de Ligação a Manose/metabolismo , Dados de Sequência Molecular , Pneumonia Intersticial Progressiva dos Ovinos/metabolismo , Pneumonia Intersticial Progressiva dos Ovinos/virologia , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Receptores Virais/química , Receptores Virais/genética , Receptores Virais/metabolismo , Análise de Sequência de Proteína/veterinária , Ovinos
16.
Vet J ; 189(1): 106-7, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20692857

RESUMO

The presence of proviral DNA, mRNA transcripts and/or viral proteins in small ruminant lentiviral infections may be intermittent. The aim of this study was to identify methods of avoiding small ruminant lentivirus (SRLV) transmission to ewes when using infected rams in artificial insemination (AI). Semen from rams, seropositive and PCR-positive in blood but consistently negative for both proviral DNA and viral protein expression in semen, was used to artificially inseminate 19 ewes. Follow-up investigation of these ewes and of two of their offspring indicated that under the study conditions virus transmission through insemination did not occur. These preliminary findings suggest that semen from SRLV-infected rams could be used for AI without the risk of transmitting virus to susceptible ewes or their lambs. Further larger studies will be required to confirm this finding.


Assuntos
Inseminação Artificial/veterinária , Infecções por Lentivirus/veterinária , Lentivirus/isolamento & purificação , Sêmen/virologia , Doenças dos Ovinos/virologia , Animais , DNA Viral/análise , Feminino , Seguimentos , Inseminação Artificial/métodos , Infecções por Lentivirus/prevenção & controle , Infecções por Lentivirus/transmissão , Masculino , Reação em Cadeia da Polimerase/veterinária , Risco , Ovinos , Doenças dos Ovinos/prevenção & controle , Doenças dos Ovinos/transmissão , Proteínas Virais/análise
17.
Vet Immunol Immunopathol ; 139(2-4): 237-44, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21122927

RESUMO

Genotype E of small ruminant lentivirus has been recently described in goats from different breeds in Italy. Genotype E infection may differ from known genotypes since deletions of dUTPase and VPR proteins have been confirmed in different independent areas and goat breed, and play a key role on virus replication and pathogenesis. In particular, genotype E Roccaverano strain has been described as low pathogenic since does not lead to clinical symptoms in goats. In contrast, classical CAEV infected goats of the same area and breed presented arthritis. In this study, we have used intratracheal and intra-bone marrow routes to establish genotype E persistent infections. Humoral and cellular immune responses elicited in the host against genotype E and genotype B derived antigens were evaluated until 200 days post-inoculation. Compared to genotype B antigen, seroconversion against genotype E GAG P16-25 antigen was detected at 2-3 weeks after inoculation, significantly earlier and at higher titres. Interestingly, antibody avidity did not increase in the course of the experiment neither against P16-25 nor against SU5, both derived from genotype E. T cell proliferation against P25-GST fusion protein antigens derived from genotype E was firstly detected at 15 days post-inoculation and was maintained throughout time until week 20 post-infection, while T cell proliferation against the genotype B P25 was not produced by the end of the experiment at 20 weeks post-inoculation. The strength of reaction was also higher when using P25 E as stimulator antigen. In contrast with antibody and T cell proliferation, cytotoxic-T-lymphocyte (CTL) activity in the circulating lymphocytes (effector cells) using blood-derived macrophages (BDM) as target cells, was not strain specific being surprisingly higher against genotype B infected antigen presenting cells (APCs). This is the first study reporting experimentally induced immunological changes in SRLV genotype E infection and indicates that CTL activity may be the adaptive immune response able to induce protection against heterologous infection.


Assuntos
Doenças das Cabras/virologia , Infecções por Lentivirus/veterinária , Lentivirus/classificação , Lentivirus/genética , Animais , Anticorpos Antivirais/sangue , Afinidade de Anticorpos , Antígenos Virais , Proliferação de Células , Genótipo , Doenças das Cabras/imunologia , Cabras , Infecções por Lentivirus/imunologia , Linfócitos T Citotóxicos
18.
Vet Res ; 41(5): 58, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20423698

RESUMO

There are few reports on the pathogenesis of scrapie (Sc) and Visna/maedi virus (VMV) coinfections. The aim of this work was to study in vivo as well as post mortem both diseases in 91 sheep. Diagnosis of Sc and VMV infections allowed the distribution of animals into five groups according to the presence (+) or absence (-) of infection by Sc and VMV: Sc-/VMV-, Sc-/VMV+, Sc+/VMV- and Sc+/VMV+. The latter was divided into two subgroups, with and without VMV-induced lymphoid follicle hyperplasia (LFH), respectively. In both the lung and mammary gland, PrPSc deposits were found in the germinal center of hyperplasic lymphoid follicles in the subgroup of Sc+/VMV+ having VMV-induced LFH. This detection was always associated with (and likely preceded by) PrPSc observation in the corresponding lymph nodes. No PrPSc was found in other VMV-associated lesions. Animals suffering from scrapie had a statistically significantly lower mean age than the scrapie free animals at the time of death, with no apparent VMV influence. ARQ/ARQ genotype was the most abundant among the 91 ewes and the most frequent in scrapie-affected sheep. VMV infection does not seem to influence the scrapie risk group distribution among animals from the five groups established in this work. Altogether, these data indicate that certain VMV-induced lesions can favor PrPSc deposits in Sc non-target organs such as the lung and the mammary gland, making this coinfection an interesting field that warrants further research for a better comprehension of the pathogenesis of both diseases.


Assuntos
Pulmão/patologia , Glândulas Mamárias Animais/virologia , Pneumonia Intersticial Progressiva dos Ovinos/complicações , Proteínas PrPSc/isolamento & purificação , Scrapie/complicações , Vírus Visna-Maedi/isolamento & purificação , Animais , Feminino , Pneumonia Intersticial Progressiva dos Ovinos/virologia , Ovinos
19.
J Virol Methods ; 165(2): 161-7, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20116400

RESUMO

The aim of this study was the development of gag and pol dual labelled probe real-time PCR and RT PCR assays to quantify the proviral load and the transcripts of the British Visna/maedi virus EV1 strain. Primers and probes were chosen based on the consensus sequences of gag and pol clones representative of EV1 genetic variants. Both PCRs had a detection limit of 3 copies of target gene, with a linearity over 6 orders of magnitude. The performances of the two PCRs in vivo were evaluated and compared on a panel of DNAs extracted from blood of sheep infected experimentally with EV1. The pol assay detected in most cases lower numbers of viral molecules than gag assay, yielding some false negative results. The gag real-time RT PCR had a detection limit of 100 RNA molecules with a linearity over 5 orders of magnitude. This did not result in a lower performance of the RT PCR compared to the PCR in cells permissive for virus replication, which contain higher numbers of viral transcripts than proviral genomes. The real-time assays developed in this study, particularly the gag assay, provide a sensitive tool which can be used to quantify the viral load in experimental infections.


Assuntos
Genes gag , Genes pol , Pneumonia Intersticial Progressiva dos Ovinos/diagnóstico , Reação em Cadeia da Polimerase/métodos , Provírus/isolamento & purificação , Carga Viral/métodos , Vírus Visna-Maedi/isolamento & purificação , Visna/diagnóstico , Animais , Sequência de Bases , Dados de Sequência Molecular , Provírus/genética , Sensibilidade e Especificidade , Alinhamento de Sequência , Ovinos , Vírus Visna-Maedi/genética
20.
Vet Microbiol ; 138(3-4): 251-7, 2009 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-19406590

RESUMO

Small ruminant lentivirus (SRLV) belonging to the highly divergent genotype E has recently been identified in the Italian goat breed Roccaverano. In this report we have developed a specific serological test based on recombinant matrix/capsid antigen fusion protein. Performance has been evaluated and compared with a similar test based on genotype B antigen. Herds under study were selected according to the infectious status characterized by blood PCR and sequencing. Results clearly showed that B and E based recombinant ELISA only detected homologous infection and an apparent cross-reactivity was recorded in a herd in which co-infection was present. Three commercially available ELISAs showed different abilities in detecting genotype E infection, being the whole virus-based immunoassay the best choice. Genotype E-recombinant antigen was not detected in ELISA by three commercially available Mabs known to be cross-reactive among CAEV and MVV capsid antigens, further supporting the high divergence of the E genotype from others. Finally, a SRLV-free herd according to commercial ELISA testing, was analysed in the same area where genotype E was identified and few animals belonging to Roccaverano breed were found slightly reactive with the E antigens. Our results suggest that the prevalence of genotype E in other small ruminant populations may be conveniently estimated using a comparative assay based on a combination of genotype specific recombinant antigens and may highlight a wider space in which SRLVs evolve.


Assuntos
Ensaio de Imunoadsorção Enzimática/veterinária , Doenças das Cabras/diagnóstico , Infecções por Lentivirus/veterinária , Lentivirus Ovinos-Caprinos/isolamento & purificação , Aminoácido N-Acetiltransferase , Animais , Ensaio de Imunoadsorção Enzimática/métodos , Regulação Viral da Expressão Gênica , Genótipo , Doenças das Cabras/virologia , Cabras , Infecções por Lentivirus/diagnóstico , Lentivirus Ovinos-Caprinos/genética , Filogenia , Estudos Soroepidemiológicos , Proteínas Virais/química , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...