Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
1.
Viruses ; 13(12)2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34960798

RESUMO

Small ruminant lentiviruses (SRLVs) are a group of highly divergent viruses responsible for global infection in sheep and goats. In a previous study we showed that SRLV strains found in mixed flocks in Poland belonged to subtype A13 and A18, but this study was restricted only to the few flocks from Malopolska region. The present work aimed at extending earlier findings with the analysis of SRLVs in mixed flocks including larger numbers of animals and flocks from different part of Poland. On the basis of gag and env sequences, Polish SRLVs were assigned to the subtypes B2, A5, A12, and A17. Furthermore, the existence of a new subtypes, tentatively designed as A23 and A24, were described for the first time. Subtypes A5 and A17 were only found in goats, subtype A24 has been detected only in sheep while subtypes A12, A23, and B2 have been found in both sheep and goats. Co-infection with strains belonging to different subtypes was evidenced in three sheep and two goats originating from two flocks. Furthermore, three putative recombination events were identified within gag and env SRLVs sequences derived from three sheep. Amino acid (aa) sequences of immunodominant epitopes in CA protein were well conserved while Major Homology Region (MHR) had more alteration showing unique mutations in sequences of subtypes A5 and A17. In contrast, aa sequences of surface glycoprotein exhibited higher variability confirming type-specific variation in the SU5 epitope. The number of potential N-linked glycosylation sites (PNGS) ranged from 3 to 6 in respective sequences and were located in different positions. The analysis of LTR sequences revealed that sequences corresponding to the TATA box, AP-4, AML-vis, and polyadenylation signal (poly A) were quite conserved, while considerable alteration was observed in AP-1 sites. Interestingly, our results revealed that all sequences belonging to subtype A17 had unique substitution T to A in the fifth position of TATA box and did not have a 11 nt deletion in the R region which was noted in other sequences from Poland. These data revealed a complex picture of SRLVs population with ovine and caprine strains belonging to group A and B. We present strong and multiple evidence of dually infected sheep and goats in mixed flocks and present evidence that these viruses can recombine in vivo.


Assuntos
Cabras/virologia , Infecções por Lentivirus/transmissão , Lentivirus/genética , Recombinação Genética , Ovinos/virologia , Animais , Lentivirus/classificação , Infecções por Lentivirus/virologia , Filogenia , Sequências Repetidas Terminais
2.
Viruses ; 13(6)2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34200141

RESUMO

The transmission of viruses from animal hosts into humans have led to the emergence of several diseases. Usually these cross-species transmissions are blocked by host restriction factors, which are proteins that can block virus replication at a specific step. In the natural virus host, the restriction factor activity is usually suppressed by a viral antagonist protein, but this is not the case for restriction factors from an unnatural host. However, due to ongoing viral evolution, sometimes the viral antagonist can evolve to suppress restriction factors in a new host, enabling cross-species transmission. Here we examine the classical case of this paradigm by reviewing research on APOBEC3 restriction factors and how they can suppress human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV). APOBEC3 enzymes are single-stranded DNA cytidine deaminases that can induce mutagenesis of proviral DNA by catalyzing the conversion of cytidine to promutagenic uridine on single-stranded viral (-)DNA if they escape the HIV/SIV antagonist protein, Vif. APOBEC3 degradation is induced by Vif through the proteasome pathway. SIV has been transmitted between Old World Monkeys and to hominids. Here we examine the adaptations that enabled such events and the ongoing impact of the APOBEC3-Vif interface on HIV in humans.


Assuntos
Desaminases APOBEC/genética , Interações Hospedeiro-Patógeno/genética , Infecções por Lentivirus/genética , Infecções por Lentivirus/transmissão , Lentivirus de Primatas/fisiologia , Zoonoses Virais/transmissão , Animais , Produtos do Gene vif/química , Produtos do Gene vif/metabolismo , Infecções por HIV/genética , Infecções por HIV/transmissão , Infecções por HIV/virologia , HIV-1/fisiologia , Humanos , Infecções por Lentivirus/virologia , Ligação Proteica , Isoformas de Proteínas , Relação Estrutura-Atividade , Produtos do Gene vif do Vírus da Imunodeficiência Humana/química , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo
3.
J Virol ; 95(13): e0017821, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-33762419

RESUMO

As the hosts of lentiviruses, almost 40 species of felids (family Felidae) are distributed around the world, and more than 20 feline species test positive for feline immunodeficiency virus (FIV), a lineage of lentiviruses. These observations suggest that FIVs globally infected a variety of feline species through multiple cross-species transmission events during a million-year history. Cellular restriction factors potentially inhibit lentiviral replication and limit cross-species lentiviral transmission, and cellular APOBEC3 deaminases are known as a potent restriction factor. In contrast, lentiviruses have evolutionary-acquired viral infectivity factor (Vif) to neutralize the APOBEC3-mediated antiviral effect. Because the APOBEC3-Vif interaction is strictly specific for viruses and their hosts, a comprehensive investigation focusing on Vif-APOBEC3 interplay can provide clues that will elucidate the roles of this virus-host interplay on cross-species transmission of lentiviruses. Here, we performed a comprehensive investigation with 144 patterns of a round robin test using 18 feline APOBEC3Z3 genes, an antiviral APOBEC3 gene in felid, and 8 FIV Vifs and derived a matrix showing the interplay between feline APOBEC3Z3 and FIV Vif. We particularly focused on the interplay between the APOBEC3Z3 of three felids (domestic cat, ocelot, and Asian golden cat) and an FIV Vif (strain Petaluma), and revealed that residues 65 and 66 of the APOBEC3Z3 protein of multiple felids are responsible for the counteraction triggered by FIV Petaluma Vif. Altogether, our findings can be a clue to elucidate not only the scenarios of the cross-species transmissions of FIVs in felids but also the evolutionary interaction between mammals and lentiviruses. IMPORTANCE Most of the emergences of new virus infections originate from the cross-species transmission of viruses. The fact that some virus infections are strictly specific for the host species indicates that certain "species barriers" in the hosts restrict cross-species jump of viruses, while viruses have evolutionary acquired their own "arms" to overcome/antagonize/neutralize these hurdles. Therefore, understanding of the molecular mechanism leading to successful cross-species viral transmission is crucial for considering the menus of the emergence of novel pathogenic viruses. In the field of retrovirology, APOBEC3-Vif interaction is a well-studied example of the battles between hosts and viruses. Here, we determined the sequences of 11 novel feline APOBEC3Z3 genes and demonstrated that all 18 different feline APOBEC3Z3 proteins tested exhibit anti-feline immunodeficiency virus (FIV) activity. Our comprehensive investigation focusing on the interplay between feline APOBEC3 and FIV Vif can be a clue to elucidate the scenarios of the cross-species transmissions of FIVs in felids.


Assuntos
Desaminase APOBEC-1/metabolismo , Produtos do Gene vif/metabolismo , Vírus da Imunodeficiência Felina/metabolismo , Infecções por Lentivirus/transmissão , Animais , Gatos , Linhagem Celular , Células HEK293 , Especificidade de Hospedeiro/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Infecções por Lentivirus/patologia , Panthera , Replicação Viral/fisiologia
4.
Commun Biol ; 4(1): 12, 2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33398025

RESUMO

Urban expansion can fundamentally alter wildlife movement and gene flow, but how urbanization alters pathogen spread is poorly understood. Here, we combine high resolution host and viral genomic data with landscape variables to examine the context of viral spread in puma (Puma concolor) from two contrasting regions: one bounded by the wildland urban interface (WUI) and one unbounded with minimal anthropogenic development (UB). We found landscape variables and host gene flow explained significant amounts of variation of feline immunodeficiency virus (FIV) spread in the WUI, but not in the unbounded region. The most important predictors of viral spread also differed; host spatial proximity, host relatedness, and mountain ranges played a role in FIV spread in the WUI, whereas roads might have facilitated viral spread in the unbounded region. Our research demonstrates how anthropogenic landscapes can alter pathogen spread, providing a more nuanced understanding of host-pathogen relationships to inform disease ecology in free-ranging species.


Assuntos
Vírus da Imunodeficiência Felina/genética , Infecções por Lentivirus/veterinária , Puma/microbiologia , Urbanização , Animais , Colorado/epidemiologia , Feminino , Fluxo Gênico , Infecções por Lentivirus/epidemiologia , Infecções por Lentivirus/transmissão , Masculino , Filogeografia , Puma/genética
5.
PLoS One ; 15(11): e0239916, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33206648

RESUMO

This study aimed to evaluate by means of Nested Polymerase Chain Reaction (nPCR), co-cultivation and sequencing, with genetic comparison between strains (mother/newborn), the occurrence of vertical transmission of Small Ruminant Lentiviruses (SRLV) from naturally occurring nannies infected for their offspring. For the detection of SRLV seropositive progenitors, blood was collected from 42 nannies in the final third of gestation in tubes with and without anticoagulant. The diagnostic tests used were Western Blot (WB) and nPCR. During the period of birth, the same blood collection procedure was performed on 73 newborns at zero hours of birth, with the same diagnostic tests. Seventeen blood samples from seven-day-old kids, proven positive for SRLV by nPCR, chosen at random, were subjected to coculture in goat synovial membrane (GSM) cells for 105 days. The pro-viral DNA extracted from the cell supernatant from the coculture was subjected to nPCR. For DNA sequencing from the nPCR products, nine positive samples were chosen at random, four nannies with their respective offspring, also positive. Each sample was performed in triplicate, thus generating 27 nPCR products of which only 19 were suitable for analysis. Among the 42 pregnant goats, in 50% (21/42) pro-viral DNA was detected by nPCR, while in the WB, only 7.14% (3/42) presented antibodies against SRLV. Regarding neonates, of the 73 kids, 34 (46.57%) were positive for the virus, using the nPCR technique, while in the serological test (WB), three positive animals (4.10%) were observed. The coculture of the 17 samples with a positive result in the nPCR was confirmed in viral isolation by amplification of the SRLV pro-viral DNA. When aligned, the pro-viral DNA sequences (nannies and their respective offspring) presented homology in relation to the standard strain CAEV Co. It was concluded that the transmission of SRLV through intrauterine route was potentially the source of infection in the newborn goats.


Assuntos
Doenças das Cabras/transmissão , Transmissão Vertical de Doenças Infecciosas/veterinária , Infecções por Lentivirus/transmissão , Provírus/isolamento & purificação , Doenças dos Ovinos/transmissão , Animais , Animais Recém-Nascidos/virologia , Linhagem Celular , DNA Viral/sangue , Feminino , Doenças das Cabras/virologia , Cabras/virologia , Lentivirus/isolamento & purificação , Infecções por Lentivirus/veterinária , Reação em Cadeia da Polimerase , Gravidez , Provírus/genética , Análise de Sequência de DNA , Ovinos/virologia , Doenças dos Ovinos/virologia
6.
Sci Rep ; 10(1): 2243, 2020 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-32042070

RESUMO

Small ruminant lentiviruses (SRLVs) are found in sheep in Germany and Iran. SRLVs have been classified into four genotypes: A-C and E. Genotype A has been subdivided into 20 subtypes. Previous studies suggested that, first, the ancestors of genotype A are those SRLVs found in Turkey, second, the evolution of SRLVs is related to the domestication process, and, third, SRLV infection was first observed in sheep in Iceland and the source of that infection was a flock imported from Germany. This study generated, for the first time, partial SRLV sequence data from German and Iranian sheep, enhancing our knowledge of the genetic and evolutionary relationships of SRLVs, and their associations with the domestication process. Based on 54 SRLV sequences from German and Iranian sheep, our results reveal: (1) SRLV subtypes A4, A5, A11, A16 and A21 (new) are found in German sheep and A22 (new) in Iranian sheep. (2) Genotype A has potentially an additional ancestor (A22), found in Iran, Lebanon and Jordan. (3) Subtype A22 is likely an old version of SRLVs. (4) The transmission routes of some SRLVs are compatible with domestication pathways. (5) This study found no evidence of Icelandic subtype A1 in German sheep.


Assuntos
Infecções por Lentivirus , Lentivirus/classificação , Lentivirus/isolamento & purificação , Filogenia , Doenças dos Ovinos , Carneiro Doméstico/virologia , Animais , Ásia , Domesticação , Europa (Continente) , Infecções por Lentivirus/transmissão , Infecções por Lentivirus/veterinária , Infecções por Lentivirus/virologia , Ovinos , Doenças dos Ovinos/transmissão , Doenças dos Ovinos/virologia
7.
Cell Host Microbe ; 26(3): 359-368.e8, 2019 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-31447307

RESUMO

Tetherin is a host defense factor that physically prevents virion release from the plasma membrane. The Nef accessory protein of simian immunodeficiency virus (SIV) engages the clathrin adaptor AP-2 to downregulate tetherin via its DIWK motif. As human tetherin lacks DIWK, antagonism of tetherin by Nef is a barrier to simian-human transmission of non-human primate lentiviruses. To determine the molecular basis for tetherin counteraction, we reconstituted the AP-2 complex with a simian tetherin and SIV Nef and determined its structure by cryoelectron microscopy (cryo-EM). Nef refolds the first α-helix of the ß2 subunit of AP-2 to a ß hairpin, creating a binding site for the DIWK sequence. The tetherin binding site in Nef is distinct from those of most other Nef substrates, including MHC class I, CD3, and CD4 but overlaps with the site for the restriction factor SERINC5. This structure explains the dependence of SIVs on tetherin DIWK and consequent barrier to human transmission.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Antígeno 2 do Estroma da Médula Óssea/química , Antígeno 2 do Estroma da Médula Óssea/farmacologia , Infecções por Lentivirus/prevenção & controle , Infecções por Lentivirus/transmissão , Zoonoses/virologia , Complexo 2 de Proteínas Adaptadoras/química , Complexo 2 de Proteínas Adaptadoras/metabolismo , Subunidades beta do Complexo de Proteínas Adaptadoras/química , Animais , Sítios de Ligação , Complexo CD3/metabolismo , Antígenos CD4/metabolismo , Membrana Celular/efeitos dos fármacos , Microscopia Crioeletrônica , Regulação para Baixo , Produtos do Gene nef/química , Produtos do Gene nef/metabolismo , Células HEK293 , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Infecções por Lentivirus/virologia , Proteínas de Membrana/metabolismo , Modelos Moleculares , Cultura Primária de Células , Conformação Proteica , Conformação Proteica em alfa-Hélice , Dobramento de Proteína , Domínios e Motivos de Interação entre Proteínas , Alinhamento de Sequência , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Síndrome de Imunodeficiência Adquirida dos Símios/transmissão , Vírus da Imunodeficiência Símia/metabolismo , Vírion/efeitos dos fármacos
8.
Arq. Inst. Biol ; 86: e0752018, 2019. ilus, tab
Artigo em Inglês | LILACS, VETINDEX | ID: biblio-1046025

RESUMO

Infections by small ruminant lentiviruses (SRLVs) affect goats and sheep causing chronic multisystemic diseases that generate great economic losses. The caprine lentivirus (CLV) and the ovine lentivirus (OLV) present tropism for cells of the monocyte/macrophage lineage, which are directly associated with the main route of transmission through the ingestion of milk and colostrum from infected animals. In this manner, controlling this route is of paramount importance. Currently, researches have investigated the use of chemical additives in milk that can preserve colostrum or milk and inactivate microbiological agents. Among the compounds, sodium dodecyl sulfate (SDS) has been shown to be satisfactory in the chemical inactivation of HIV and CLV in milk, and also as a biocide in goat colostrum.(AU)


As lentiviroses de pequenos ruminantes (LVPRs) são infecções que afetam caprinos e ovinos, causando doenças multissistêmicas crônicas, ocasionando grandes perdas econômicas. Os agentes causadores, lentivírus caprino (LVC) e o lentivírus ovino (LVO), apresentam tropismo por células da linhagem monocítico--fagocitária, as quais estão diretamente associadas à principal via de transmissão, por meio da ingestão de leite e colostro provindos de animais infectados. Desse modo, o controle por esta via é de suma importância. Atualmente, pesquisas vêm sendo desenvolvidas para o uso de aditivos químicos no leite, que possam conservar o colostro ou leite, e inativar agentes microbiológicos presentes. Dentre estes, o dodecil sulfato de sódio (SDS) vem apresentando resultados satisfatórios na inativação química do HIV e LVC em leite, e ainda como biocida em colostro caprino.(AU)


Assuntos
Animais , Dodecilsulfato de Sódio/farmacologia , Ruminantes/virologia , Infecções por Lentivirus/tratamento farmacológico , Lentivirus Ovinos-Caprinos/efeitos dos fármacos , Ovinos/virologia , Infecções por Lentivirus/transmissão , Colostro/virologia , Leite/virologia
9.
Retrovirology ; 15(1): 31, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29636069

RESUMO

BACKGROUND: The apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3 (APOBEC3; A3) gene family appears only in mammalian genomes. Some A3 proteins can be incorporated into progeny virions and inhibit lentiviral replication. In turn, the lentiviral viral infectivity factor (Vif) counteracts the A3-mediated antiviral effect by degrading A3 proteins. Recent investigations have suggested that lentiviral vif genes evolved to combat mammalian APOBEC3 proteins, and have further proposed that the Vif-A3 interaction may help determine the co-evolutionary history of cross-species lentiviral transmission in mammals. RESULTS: Here we address the co-evolutionary relationship between two New World felids, the puma (Puma concolor) and the bobcat (Lynx rufus), and their lentiviruses, which are designated puma lentiviruses (PLVs). We demonstrate that PLV-A Vif counteracts the antiviral action of APOBEC3Z3 (A3Z3) of both puma and bobcat, whereas PLV-B Vif counteracts only puma A3Z3. The species specificity of PLV-B Vif is irrespective of the phylogenic relationships of feline species in the genera Puma, Lynx and Acinonyx. We reveal that the amino acid at position 178 in the puma and bobcat A3Z3 is exposed on the protein surface and determines the sensitivity to PLV-B Vif-mediated degradation. Moreover, although both the puma and bobcat A3Z3 genes are polymorphic, their sensitivity/resistance to PLV Vif-mediated degradation is conserved. CONCLUSIONS: To the best of our knowledge, this is the first study suggesting that the host A3 protein potently controls inter-genus lentiviral transmission. Our findings provide the first evidence suggesting that the co-evolutionary arms race between lentiviruses and mammals has occurred in the New World.


Assuntos
Citosina Desaminase/genética , Interações Hospedeiro-Patógeno/genética , Infecções por Lentivirus/transmissão , Infecções por Lentivirus/virologia , Lentivirus/fisiologia , Animais , Gatos , Citosina Desaminase/química , Citosina Desaminase/metabolismo , Resistência à Doença , Evolução Molecular , Produtos do Gene vif , Vírus da Imunodeficiência Felina/classificação , Vírus da Imunodeficiência Felina/genética , Lentivirus/classificação , Mutação com Perda de Função , Modelos Moleculares , Filogenia , Polimorfismo Genético , Conformação Proteica , Proteólise , Relação Estrutura-Atividade , Treonina/química , Treonina/genética
10.
Viruses ; 10(4)2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29642583

RESUMO

The replication of lentiviruses highly depends on host cellular factors, which defines their species-specific tropism. Cellular restriction factors that can inhibit lentiviral replication were recently identified. Feline immunodeficiency virus (FIV) was found to be sensitive to several feline cellular restriction factors, such as apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3 (APOBEC3) and tetherin, but FIV evolved to counteract them. Here, we describe the molecular mechanisms by which feline APOBEC3 restriction factors inhibit FIV replication and discuss the molecular interaction of APOBEC3 proteins with the viral antagonizing protein Vif. We speculate that feline APOBEC3 proteins could explain some of the observed FIV cross-species transmissions described in wild Felids.


Assuntos
Desaminases APOBEC/metabolismo , Doenças do Gato/transmissão , Vírus da Imunodeficiência Felina/fisiologia , Infecções por Lentivirus/veterinária , Desaminases APOBEC/genética , Animais , Doenças do Gato/virologia , Gatos , Produtos do Gene vif/deficiência , Produtos do Gene vif/metabolismo , Infecções por Lentivirus/transmissão , Infecções por Lentivirus/virologia , Modelos Biológicos , Especificidade da Espécie , Replicação Viral
11.
Mol Ecol ; 26(22): 6487-6498, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28987024

RESUMO

Urban expansion has widespread impacts on wildlife species globally, including the transmission and emergence of infectious diseases. However, there is almost no information about how urban landscapes shape transmission dynamics in wildlife. Using an innovative phylodynamic approach combining host and pathogen molecular data with landscape characteristics and host traits, we untangle the complex factors that drive transmission networks of feline immunodeficiency virus (FIV) in bobcats (Lynx rufus). We found that the urban landscape played a significant role in shaping FIV transmission. Even though bobcats were often trapped within the urban matrix, FIV transmission events were more likely to occur in areas with more natural habitat elements. Urban fragmentation also resulted in lower rates of pathogen evolution, possibly owing to a narrower range of host genotypes in the fragmented area. Combined, our findings show that urban landscapes can have impacts on a pathogen and its evolution in a carnivore living in one of the most fragmented and urban systems in North America. The analytical approach used here can be broadly applied to other host-pathogen systems, including humans.


Assuntos
Evolução Molecular , Fluxo Gênico , Vírus da Imunodeficiência Felina/genética , Infecções por Lentivirus/transmissão , Lynx/virologia , Urbanização , Animais , Animais Selvagens/virologia , Teorema de Bayes , Ecossistema , Los Angeles , Modelos Genéticos , Filogenia , Análise Espacial
12.
J Anim Ecol ; 86(6): 1469-1482, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28884827

RESUMO

Heterogeneity within pathogen species can have important consequences for how pathogens transmit across landscapes; however, discerning different transmission routes is challenging. Here, we apply both phylodynamic and phylogenetic community ecology techniques to examine the consequences of pathogen heterogeneity on transmission by assessing subtype-specific transmission pathways in a social carnivore. We use comprehensive social and spatial network data to examine transmission pathways for three subtypes of feline immunodeficiency virus (FIVPle ) in African lions (Panthera leo) at multiple scales in the Serengeti National Park, Tanzania. We used FIVPle molecular data to examine the role of social organization and lion density in shaping transmission pathways and tested to what extent vertical (i.e., father- and/or mother-offspring relationships) or horizontal (between unrelated individuals) transmission underpinned these patterns for each subtype. Using the same data, we constructed subtype-specific FIVPle co-occurrence networks and assessed what combination of social networks, spatial networks or co-infection best structured the FIVPle network. While social organization (i.e., pride) was an important component of FIVPle transmission pathways at all scales, we find that FIVPle subtypes exhibited different transmission pathways at within- and between-pride scales. A combination of social and spatial networks, coupled with consideration of subtype co-infection, was likely to be important for FIVPle transmission for the two major subtypes, but the relative contribution of each factor was strongly subtype-specific. Our study provides evidence that pathogen heterogeneity is important in understanding pathogen transmission, which could have consequences for how endemic pathogens are managed. Furthermore, we demonstrate that community phylogenetic ecology coupled with phylodynamic techniques can reveal insights into the differential evolutionary pressures acting on virus subtypes, which can manifest into landscape-level effects.


Assuntos
Coinfecção/veterinária , Vírus da Imunodeficiência Felina/fisiologia , Infecções por Lentivirus/veterinária , Leões , Animais , Coinfecção/transmissão , Coinfecção/virologia , Vírus da Imunodeficiência Felina/classificação , Infecções por Lentivirus/transmissão , Infecções por Lentivirus/virologia , Leões/fisiologia , Filogenia , Comportamento Social , Tanzânia
13.
Pesqui. vet. bras ; 37(8): 805-812, Aug. 2017. tab, graf
Artigo em Português | LILACS, VETINDEX | ID: biblio-895497

RESUMO

A Artrite Encefalite Caprina se caracteriza por ser multissistêmica e infecciosa, causada por um lentivírus. O estudo teve como objetivo avaliar a transmissibilidade do Lentivírus Caprino, para fêmeas e sua prole, por meio de sêmen infectado experimentalmente. Para tanto, onze fêmeas livres de CAEV foram inseminadas artificialmente com sêmen de bode livre de CAEV ao qual foi adicionado CAEV-Cork para obter título infectante com carga viral em 105 TCID50/ml. (grupo experimental 1). Destas, seis obtiverem prenhez confirmada, e a sua prole (n=6) constituiu o grupo experimental 2. Duas cabras livres de CAEV foram inseminadas artificialmente com sêmen do mesmo bode, sem o inócuo viral, constituindo-se o grupo controle. O diagnóstico da infecção pelo Lentivírus Caprino, foi realizado por IDGA, cELISA e nested-PCR. As fêmeas foram monitoradas durante 210 dias pós inseminação artificial. Já as proles foram imediatamente separadas das mães após o nascimento, e monitoradas nos momentos hora zero, aos quinze dias de idade e mensalmente, até doze meses de idade. Em relação às cabras, 56,96%(9/158) apresentaram positividade para cELISA, 24,05% (38/158) foram positivas a IDGA e nenhuma para nested-PCR. Em relação aos cabritos, 11,28% (15/133) amostras positivas para nested-PCR, 5,26% (7/133) amostras positivas para IDGA e nenhum para cELISA. As proles do grupo controle apresentaram resultados negativos para as três técnicas. A positividade encontrada em nested-PCR pode indicar grande importância para identificação de animais infectados, porém soronegativos, em situações de soroconversão tardia. De acordo com os resultados, concluiu-se que há a transmissão do Lentivírus caprino para a prole e para as mães pelo sêmen infectado.(AU)


Caprine Arthritis Encephalitis is a multisystemic infectious disease, caused by a lentivirus. The objective of this study was to evaluate the transmissibility of caprine lentivirus to goats and their offspring, through experimentally infected semen. Therefore, eleven free-CAEV goats were artificially inseminated using semen from a free-CAEV buck experimentally infected with CAEV-Cork strain (experimental group one). Pregnancy was confirmed in only six goats and their offspring (n=6) constituted the experimental group two. Two free-CAEV females were artificially inseminated with semen from the same seronegative buck, without viral inoculum to constitute the control group. The diagnosis of caprine lentivirus infection was performed using AGID, cELISA and nested-PCR. All females were monitored for 210 days after artificial insemination. Kids were immediately separated from their mothers after birth, and monitored at zero time, 15 days old and monthly until 12 months old. Regarding goat samples, 56.96% (9/159) were positive in cELISA, 24.05% (38/158) were positive in IDGA and none was positive in nested-PCR. Regarding to the offspring samples, 11.28% (15/133) and 5.26% (7/133) were positive in nested-PCR and IDGA, respectively, while no sample was positive in cELISA. The control group showed no positives in the three techniques. The positivity observed to nested-PCR may show its importance to identify infected, but seronegative animals, in late seroconversion situations. According to results, the transmission of caprine lentivirus to offspring and their mothers through infected semen is possible.(AU)


Assuntos
Animais , Sêmen/virologia , Cabras , Infecções por Lentivirus/transmissão , Infecções por Lentivirus/veterinária , Lentivirus Ovinos-Caprinos , Vírus da Artrite-Encefalite Caprina , Animais Recém-Nascidos , Ensaio de Imunoadsorção Enzimática/veterinária , Reação em Cadeia da Polimerase/veterinária , Imunodifusão/veterinária
14.
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
15.
Braz J Microbiol ; 46(3): 867-74, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26413072

RESUMO

This study was conducted in order to evaluate the transmission of caprine lentivirus to sheep using different experimental groups. The first one (colostrum group) was formed by nine lambs receiving colostrum from goats positive for small ruminant lentiviruses (SRLV). The second group (milk group) was established by nine lambs that received milk of these goats. Third was a control group, consisting of lambs that suckled colostrum and milk of negative mothers. Another experimental group (contact group) was formed by eight adult sheep, confined with two naturally infected goats. The groups were monitored by immunoblotting (IB), enzyme-linked immunosorbent assay (ELISA), agar gel immunodiffusion (AGID) and nested polymerase chain reaction (nPCR). All lambs that suckled colostrum and milk of infected goats and six sheep of the contact group had positive results in the nPCR, although seroconversion was detected only in three of the exposed animals, with no clinical lentiviruses manifestation, in 720 days of observation. There was a close relationship between viral sequences obtained from infected animals and the prototype CAEV-Cork. Thus, it was concluded that SRLV can be transmitted from goats to sheep, however, the degree of adaptation of the virus strain to the host species probably interferes with the infection persistence and seroconversion rate.


Assuntos
Vírus da Artrite-Encefalite Caprina/patogenicidade , Colostro/virologia , Doenças das Cabras/transmissão , Infecções por Lentivirus/transmissão , Doenças dos Ovinos/transmissão , Vírus Visna-Maedi/patogenicidade , Animais , Anticorpos Antivirais/sangue , Doenças das Cabras/virologia , Cabras/virologia , Interações Hospedeiro-Patógeno/fisiologia , Infecções por Lentivirus/virologia , Ruminantes/virologia , Soroconversão/fisiologia , Ovinos/virologia , Doenças dos Ovinos/virologia
16.
Braz. j. microbiol ; 46(3): 867-874, July-Sept. 2015. tab, ilus
Artigo em Inglês | LILACS | ID: lil-755808

RESUMO

This study was conducted in order to evaluate the transmission of caprine lentivirus to sheep using different experimental groups. The first one (colostrum group) was formed by nine lambs receiving colostrum from goats positive for small ruminant lentiviruses (SRLV). The second group (milk group) was established by nine lambs that received milk of these goats. Third was a control group, consisting of lambs that suckled colostrum and milk of negative mothers. Another experimental group (contact group) was formed by eight adult sheep, confined with two naturally infected goats. The groups were monitored by immunoblotting (IB), enzyme-linked immunosorbent assay (ELISA), agar gel immunodiffusion (AGID) and nested polymerase chain reaction (nPCR). All lambs that suckled colostrum and milk of infected goats and six sheep of the contact group had positive results in the nPCR, although seroconversion was detected only in three of the exposed animals, with no clinical lentiviruses manifestation, in 720 days of observation. There was a close relationship between viral sequences obtained from infected animals and the prototype CAEV-Cork. Thus, it was concluded that SRLV can be transmitted from goats to sheep, however, the degree of adaptation of the virus strain to the host species probably interferes with the infection persistence and seroconversion rate.

.


Assuntos
Animais , Vírus da Artrite-Encefalite Caprina/patogenicidade , Colostro/virologia , Doenças das Cabras/transmissão , Infecções por Lentivirus/transmissão , Doenças dos Ovinos/transmissão , Vírus Visna-Maedi/patogenicidade , Anticorpos Antivirais/sangue , Doenças das Cabras/virologia , Cabras/virologia , Interações Hospedeiro-Patógeno/fisiologia , Infecções por Lentivirus/virologia , Ruminantes/virologia , Soroconversão/fisiologia , Doenças dos Ovinos/virologia , Ovinos/virologia
17.
J Infect Dis ; 210 Suppl 3: S654-9, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25414420

RESUMO

Small-animal models of lentivirus transmission have repeatedly demonstrated transmission by cell-associated virus via vaginal, rectal, and oral routes. The earliest experiments were in the cat/feline immunodeficiency virus model, followed a decade later by successful vaginal transmission of cell-associated human immunodeficiency virus (HIV) in mice bearing transplanted human immune cells. After early unsuccessful attempts at cell-associated transmission in nonhuman primates, renewed investigation in diverse primate models has now confirmed the findings from the cat and humanized mouse models. Improvements in humanized mouse models have made them the preferred small-animal models to study HIV mucosal transmission. They provide complementary systems to nonhuman primate models to aid in the elucidation of the many remaining questions on the mechanism of and means to prevent both cell-associated and cell-free HIV transmission across mucosal barriers.


Assuntos
Infecções por HIV/transmissão , HIV-1/fisiologia , Infecções por Lentivirus/transmissão , Lentivirus/fisiologia , Animais , Gatos/virologia , Modelos Animais de Doenças , Infecções por HIV/prevenção & controle , Humanos , Leucócitos/virologia , Macaca mulatta/virologia , Camundongos/virologia
18.
Vet J ; 201(2): 184-8, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24698667

RESUMO

Conflicting accounts have been published in the veterinary literature regarding transmission of feline immunodeficiency virus (FIV) between cohabiting cats in mixed households, and the mechanics of possible casual transmission, if it occurs, are poorly understood. Similarly, there are conflicting reports of vertical transmission of FIV. The aim of the present study was to document the FIV serological status of cats taken into two rescue shelters. At rescue shelter 1 (Rescue 1), cats cohabited in a multi-cat household of FIV-negative and naturally-infected, FIV-positive cats. A study was performed that combined a retrospective review of records of FIV serological status at intake (Test 1) and prospective FIV serological testing (Tests 2 and 3). Retrospective records were analyzed at rescue shelter 2 (Rescue 2), where FIV-positive queens with litters of nursing kittens were taken into the shelter, before being rehomed. FIV serology was performed on all kittens after weaning. Initial test results (Test 1) for 138 cohabiting cats from Rescue 1 showed that there were 130 FIV-negative cats and eight FIV-positive cats (six male neutered and two female spayed). A second test (Test 2), performed in 45 of the FIV-negative and five of the FIV-positive cats at median 28 months after Test 1 (range, 1 month to 8.8 years) showed that results were unchanged. Similarly, a third test (Test 3), performed in four of the original FeLV-negative cats and one remaining FIV-positive cat at median 38 months after Test 1 (range, 4 months to 4 years), also showed that results were unchanged. These results show a lack of evidence of FIV transmission, despite years of exposure to naturally-infected, FIV-positive cats in a mixed household. At Rescue 2, records were available from five FIV-positive queens with 19 kittens. All 19 kittens tested FIV-negative, suggesting that vertical transmission had not occurred.


Assuntos
Doenças do Gato/transmissão , Vírus da Imunodeficiência Felina/isolamento & purificação , Transmissão Vertical de Doenças Infecciosas/veterinária , Infecções por Lentivirus/veterinária , Animais , Anticorpos Antivirais/sangue , Doenças do Gato/virologia , Gatos , Feminino , Vírus da Imunodeficiência Felina/genética , Infecções por Lentivirus/transmissão , Infecções por Lentivirus/virologia , Vírus da Leucemia Felina/genética , Vírus da Leucemia Felina/isolamento & purificação , Masculino , Estudos Prospectivos , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Estudos Retrospectivos
19.
J Virol ; 88(10): 5687-705, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24623416

RESUMO

UNLABELLED: African green monkeys (AGMs) are naturally infected with simian immunodeficiency virus (SIV) at high prevalence levels and do not progress to AIDS. Sexual transmission is the main transmission route in AGM, while mother-to-infant transmission (MTIT) is negligible. We investigated SIV transmission in wild AGMs to assess whether or not high SIV prevalence is due to differences in mucosal permissivity to SIV (i.e., whether the genetic bottleneck of viral transmission reported in humans and macaques is also observed in AGMs in the wild). We tested 121 sabaeus AGMs (Chlorocebus sabaeus) from the Gambia and found that 53 were SIV infected (44%). By combining serology and viral load quantitation, we identified 4 acutely infected AGMs, in which we assessed the diversity of the quasispecies by single-genome amplification (SGA) and documented that a single virus variant established the infections. We thus show that natural SIV transmission in the wild is associated with a genetic bottleneck similar to that described for mucosal human immunodeficiency virus (HIV) transmission in humans. Flow cytometry assessment of the immune cell populations did not identify major differences between infected and uninfected AGM. The expression of the SIV coreceptor CCR5 on CD4+ T cells dramatically increased in adults, being higher in infected than in uninfected infant and juvenile AGMs. Thus, the limited SIV MTIT in natural hosts appears to be due to low target cell availability in newborns and infants, which supports HIV MTIT prevention strategies aimed at limiting the target cells at mucosal sites. Combined, (i) the extremely high prevalence in sexually active AGMs, (ii) the very efficient SIV transmission in the wild, and (iii) the existence of a fraction of multiparous females that remain uninfected in spite of massive exposure to SIV identify wild AGMs as an acceptable model of exposed, uninfected individuals. IMPORTANCE: We report an extensive analysis of the natural history of SIVagm infection in its sabaeus monkey host, the African green monkey species endemic to West Africa. Virtually no study has investigated the natural history of SIV infection in the wild. The novelty of our approach is that we report for the first time that SIV infection has no discernible impact on the major immune cell populations in natural hosts, thus confirming the nonpathogenic nature of SIV infection in the wild. We also focused on the correlates of SIV transmission, and we report, also for the first time, that SIV transmission in the wild is characterized by a major genetic bottleneck, similar to that described for HIV-1 transmission in humans. Finally, we report here that the restriction of target cell availability is a major correlate of the lack of SIV transmission to the offspring in natural hosts of SIVs.


Assuntos
Infecções por Lentivirus/veterinária , Doenças dos Macacos/transmissão , Doenças dos Macacos/virologia , Vírus da Imunodeficiência Símia/isolamento & purificação , Animais , Chlorocebus aethiops , Análise por Conglomerados , Feminino , Citometria de Fluxo , Gâmbia , Genótipo , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/transmissão , Infecções por Lentivirus/virologia , Subpopulações de Linfócitos/imunologia , Masculino , Dados de Sequência Molecular , Filogenia , Análise de Sequência de DNA , Vírus da Imunodeficiência Símia/classificação , Vírus da Imunodeficiência Símia/genética
20.
Viruses ; 5(8): 2043-61, 2013 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-23965529

RESUMO

Small ruminant lentiviruses (SRLV) are members of the Retrovirus family comprising the closely related Visna/Maedi Virus (VMV) and the Caprine Arthritis-Encephalitis Virus (CAEV), which infect sheep and goats. Both infect cells of the monocyte/macrophage lineage and cause lifelong infections. Infection by VMV and CAEV can lead to Visna/Maedi (VM) and Caprine Arthritis-Encephalitis (CAE) respectively, slow progressive inflammatory diseases primarily affecting the lungs, nervous system, joints and mammary glands. VM and CAE are distributed worldwide and develop over a period of months or years, always leading to the death of the host, with the consequent economic and welfare implications. Currently, the control of VM and CAE relies on the control of transmission and culling of infected animals. However, there is evidence that host genetics play an important role in determining Susceptibility/Resistance to SRLV infection and disease progression, but little work has been performed in small ruminants. More research is necessary to understand the host-SRLV interaction.


Assuntos
Doenças das Cabras/virologia , Interações Hospedeiro-Patógeno , Infecções por Lentivirus/veterinária , Lentivirus/patogenicidade , Doenças dos Ovinos/virologia , Animais , Doenças das Cabras/patologia , Doenças das Cabras/prevenção & controle , Doenças das Cabras/transmissão , Cabras , Infecções por Lentivirus/patologia , Infecções por Lentivirus/transmissão , Infecções por Lentivirus/virologia , Ovinos , Doenças dos Ovinos/patologia , Doenças dos Ovinos/prevenção & controle , Doenças dos Ovinos/transmissão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...