Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Front Immunol ; 15: 1350267, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38545109

RESUMO

Introduction: African swine fever virus (ASFV) is a nucleocytoplasmic large DNA virus (NCLDV) that encodes its own host-like RNA polymerase (RNAP) and factors required to produce mature mRNA. The formation of accurate mRNA 3' ends by ASFV RNAP depends on transcription termination, likely enabled by a combination of sequence motifs and transcription factors, although these are poorly understood. The termination of any RNAP is rarely 100% efficient, and the transcriptional "readthrough" at terminators can generate long mRNAs which may interfere with the expression of downstream genes. ASFV transcriptome analyses reveal a landscape of heterogeneous mRNA 3' termini, likely a combination of bona fide termination sites and the result of mRNA degradation and processing. While short-read sequencing (SRS) like 3' RNA-seq indicates an accumulation of mRNA 3' ends at specific sites, it cannot inform about which promoters and transcription start sites (TSSs) directed their synthesis, i.e., information about the complete and unprocessed mRNAs at nucleotide resolution. Methods: Here, we report a rigorous analysis of full-length ASFV transcripts using long-read sequencing (LRS). We systematically compared transcription termination sites predicted from SRS 3' RNA-seq with 3' ends mapped by LRS during early and late infection. Results: Using in-vitro transcription assays, we show that recombinant ASFV RNAP terminates transcription at polyT stretches in the non-template strand, similar to the archaeal RNAP or eukaryotic RNAPIII, unaided by secondary RNA structures or predicted viral termination factors. Our results cement this T-rich motif (U-rich in the RNA) as a universal transcription termination signal in ASFV. Many genes share the usage of the same terminators, while genes can also use a range of terminators to generate transcript isoforms varying enormously in length. A key factor in the latter phenomenon is the highly abundant terminator readthrough we observed, which is more prevalent during late compared with early infection. Discussion: This indicates that ASFV mRNAs under the control of late gene promoters utilize different termination mechanisms and factors to early promoters and/or that cellular factors influence the viral transcriptome landscape differently during the late stages of infection.


Assuntos
Vírus da Febre Suína Africana , Suínos , Animais , Vírus da Febre Suína Africana/genética , Transcrição Gênica , RNA Polimerases Dirigidas por DNA , RNA Mensageiro/genética , RNA
2.
PLoS One ; 19(3): e0293049, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38512923

RESUMO

African swine fever (ASF) is a devastating disease of domestic pigs that has spread across the globe since its introduction into Georgia in 2007. The etiological agent is a large double-stranded DNA virus with a genome of 170 to 180 kb in length depending on the isolate. Much of the differences in genome length between isolates are due to variations in the copy number of five different multigene families that are encoded in repetitive regions that are towards the termini of the covalently closed ends of the genome. Molecular epidemiology of African swine fever virus (ASFV) is primarily based on Sanger sequencing of a few conserved and variable regions, but due to the stability of the dsDNA genome changes in the variable regions occur relatively slowly. Observations in Europe and Asia have shown that changes in other genetic loci can occur and that this could be useful in molecular tracking. ASFV has been circulating in Western Africa for at least forty years. It is therefore reasonable to assume that changes may have accumulated in regions of the genome other than the standard targets over the years. At present only one full genome sequence is available for an isolate from Western Africa, that of a highly virulent isolate collected from Benin during an outbreak in 1997. In Cameroon, ASFV was first reported in 1981 and outbreaks have been reported to the present day and is considered endemic. Here we report three full genome sequences from Cameroon isolates of 1982, 1994 and 2018 outbreaks and identify novel single nucleotide polymorphisms and insertion-deletions that may prove useful for molecular epidemiology studies in Western Africa and beyond.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Suínos , Animais , Febre Suína Africana/epidemiologia , Camarões/epidemiologia , Sus scrofa/genética , Análise de Sequência , Análise de Sequência de DNA
3.
Emerg Microbes Infect ; 12(2): 2265661, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37781934

RESUMO

African swine fever virus is a complex DNA virus that causes high fatality in pigs and wild boar and has a great socio-economic impact. An attenuated genotype II strain was constructed by replacing the gene for wildtype CD2v protein with versions in which single or double amino acid substitutions were introduced to reduce or abrogate the binding to red blood cells and reduce virus persistence in blood. The mutant CD2v proteins were expressed at similar levels to the wildtype protein on the surface of infected cells. Three recombinant viruses also had K145R, EP153R, and in one virus DP148R genes deleted. Following immunization of pigs, the virus with a single amino acid substitution in CD2v, Q96R, induced moderate levels of replication, and 100% protection against virulent ASFV. Two additional recombinant viruses had two amino acid substitutions in CD2v, Q96R, and K108D, and induced no binding to red blood cells in vitro. In immunized pigs, reduced levels of virus in blood and strong early ASFV-specific antibody and cellular responses were detected. After challenge low to moderate replication of challenge virus was observed. Reduced clinical signs post-challenge were observed in pigs immunized with the virus from which DP148R gene was deleted. Protection levels of 83-100% were maintained across a range of doses. Further experiments with virus GeorgiaΔDP148RΔK145RΔEP153R-CD2v_mutantQ96R/K108D showed low levels of virus dissemination in tissue and transient clinical signs at high doses. The results support further evaluation of GeorgiaΔDP148RΔK145RΔEP153R-CD2v_mutantQ96R/K108D as a vaccine candidate.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Vacinas Virais , Suínos , Animais , Vírus da Febre Suína Africana/fisiologia , Febre Suína Africana/prevenção & controle , Proteínas Virais/genética , Genótipo , Anticorpos Antivirais
4.
J Virol ; 97(10): e0110623, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37796125

RESUMO

IMPORTANCE: African swine fever virus (ASFV) causes a lethal disease of pigs with high economic impact in affected countries in Africa, Europe, and Asia. The virus encodes proteins that inhibit host antiviral defenses, including the type I interferon response. Host cells also activate cell death through a process called apoptosis to limit virus replication. We showed that the ASFV A179L protein, a BCL-2 family apoptosis inhibitor, is important in reducing apoptosis in infected cells since deletion of this gene increased cell death and reduced virus replication in cells infected with the A179L gene-deleted virus. Pigs immunized with the BeninΔA179L virus showed no clinical signs and a weak immune response but were not protected from infection with the deadly parental virus. The results show an important role for the A179L protein in virus replication in macrophages and virulence in pigs and suggest manipulation of apoptosis as a possible route to control infection.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Apoptose , Deleção de Genes , Macrófagos , Proteínas Proto-Oncogênicas c-bcl-2 , Suínos , Proteínas Virais , Virulência , Animais , Febre Suína Africana/virologia , Vírus da Febre Suína Africana/genética , Macrófagos/virologia , Proteínas Proto-Oncogênicas c-bcl-2/deficiência , Proteínas Proto-Oncogênicas c-bcl-2/genética , Suínos/virologia , Virulência/genética , Replicação Viral , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Proteínas Virais/genética
5.
Pathogens ; 12(8)2023 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-37623982

RESUMO

African swine fever virus (ASFV) is known to be very stable and can remain infectious over long periods of time especially at low temperatures and within different matrices, particularly those containing animal-derived organic material. However, there are some gaps in our knowledge pertaining to the survivability and infectivity of ASFV in groundwater. This study aims to determine the stability and infectivity of the cell culture-adapted ASFV strain BA71V by plaque assay after incubation of the virus within river water samples at three different environmentally relevant temperatures (4 °C, 15 °C, and 21 °C) over the course of 42 days. The results from this study indicate that ASFV can remain stable and infectious when maintained at 4 °C in river water for more than 42 days, but as incubation temperatures are increased, the stability is reduced, and the virus is no longer able to form plaques after 28 days and 14 days, respectively, when stored at 15 °C and 21 °C. Characterizing the survivability of ASFV in groundwater can allow us to develop more appropriate inactivation and disinfection methods to support disease control and mitigate ASFV outbreaks.

6.
Pathogens ; 12(7)2023 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-37513703

RESUMO

African swine fever (ASF) is an economically important disease due to high morbidity and mortality rates and the ability to affect all ages and breeds of pigs. Biosecurity measures to prevent the spread of the causative agent, African swine fever virus (ASFV), include prescriptive cleaning and disinfection procedures. The aim of this study was to establish the biocidal effects of twenty-four commercially available disinfectants including oxidizing agents, acids, aldehydes, formic acids, phenol, and mixed-class chemistries against ASFV. The products were prepared according to the manufacturer's instructions and a suspension assay was performed with ASFV strain, BA71V using Vero cells (African green monkey cells) to test efficacy in reducing ASFV infection of cells. Generally, disinfectants containing formic acid and phenolic compounds, as well as oxidizing agents reduced viral titers of ASFV by over 4 log10 at temperatures ranging from 4 °C to 20 °C. Hydrogen peroxide, aldehyde, and quaternary ammonium compounds containing disinfectants were cytotoxic, limiting the detection of viral infectivity reductions to less than 4 log10. These preliminary results can be used to target research on disinfectants which contain active ingredients with known efficacy against ASFV under conditions recommended for the country where their use will be applied.

7.
Viruses ; 16(1)2023 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-38275939

RESUMO

The 2023 International African Swine Fever Workshop (IASFW) took place in Beijing, China, on 18-20 September 2023. It was jointly organized by the U.S.-China Center for Animal Health (USCCAH) at Kansas State University (KSU) and the Chinese Veterinary Drug Association (CVDA) and sponsored by the United States Department of Agriculture Foreign Agricultural Service (USDA-FAS), Harbin Veterinary Research Institute, and Zoetis Inc. The objective of this workshop was to provide a platform for ASF researchers around the world to unite and share their knowledge and expertise on ASF control and prevention. A total of 24 outstanding ASF research scientists and experts from 10 countries attended this meeting. The workshop included presentations on current ASF research, opportunities for scientific collaboration, and discussions of lessons and experiences learned from China/Asia, Africa, and Europe. This article summarizes the meeting highlights and presents some critical issues that need to be addressed for ASF control and prevention in the future.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Suínos , Animais , Humanos , Febre Suína Africana/prevenção & controle , Febre Suína Africana/epidemiologia , Ásia , China/epidemiologia , África/epidemiologia , Sus scrofa , Surtos de Doenças/veterinária
8.
mSphere ; 7(6): e0037822, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36317894

RESUMO

African swine fever virus (ASFV) causes a highly contagious hemorrhagic disease with case fatality rates approaching 100% in domestic pigs. ASFV is responsible for substantial economic losses, but despite ongoing efforts, no vaccine or antiviral agent is currently available. Attempts to control the spread of ASFV are dependent on early detection, adherence to biosecurity measures, and culling of infected herds. However, an effective antiviral agent may be used in lieu of or in conjunction with a vaccine to effectively curb ASFV outbreaks. The dose-dependent antiviral activities of two amidate prodrugs (compounds 1a and 1b) of O-2-alkylated 3-fluoro-2-(phosphonomethoxy)propyl cytosine [(R)-O-2-alkylated FPMPC] against ASFV isolates of four different genotypes were determined. Both compounds were found to inhibit ASFV progeny virus output by >90% at noncytotoxic concentrations (<25 µM) in primary porcine macrophages. Analysis of viral transcription and viral protein synthesis indicated that these acyclic nucleotide analogues inhibited late gene expression. Interestingly, time-of-addition studies suggest different viral targets of the compounds, which may be attributed to their differing amino acid prodrug moieties. In view of their promising antiviral activity, these nucleotide analogues merit further evaluation as potential prophylactic and/or therapeutic agents against ASFV infection and their antiviral efficacy in vivo should be considered. IMPORTANCE African swine fever virus is a highly contagious hemorrhagic viral disease. Since its transcontinental spread to Georgia in 2007, ASFV has continued to spread across the globe into countries previously without infection. It is responsible for substantial losses in the domestic pig population and presents a significant threat to the global swine industry. Despite ongoing efforts, there are no vaccines currently available; in their absence, antiviral agents may be a viable alternative. The significance of our research is in identifying the pan-genotype antiviral activity of prodrugs of O-2-alkylated 3-fluoro-2-(phosphonomethoxy)propyl cytosine, which will drive further research on the development of these compounds as antivirals against ASFV.


Assuntos
Vírus da Febre Suína Africana , Pró-Fármacos , Suínos , Animais , Vírus da Febre Suína Africana/genética , Pró-Fármacos/farmacologia , Nucleosídeos/farmacologia , Antivirais/farmacologia , Sus scrofa , Genótipo , Nucleotídeos
9.
Antiviral Res ; 208: 105433, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36252822

RESUMO

African swine fever virus (ASFV) causes a haemorrhagic disease affecting wild boar and domestic pigs which can result in morbidity and fatality rates of up to 100%. ASFV is a large double-stranded DNA virus which replicates predominantly in the cell cytoplasm and codes for its replication and transcription machinery. No vaccine is widely available and control depends on early detection, culling of infected herds and adherence to biosecurity measures. In this study the small molecule nucleoside analogue, cyclic cidofovir (cHPMPC), was evaluated for its ability to inhibit replication of four different ASFV genotypes in primary porcine macrophages. Time of addition studies demonstrated that cHPMPC effectively inhibits ASFV replication and late gene expression when added pre-infection or early post-infection but not when added at late times, suggesting the drug target may be the virus DNA polymerase, or the RNA polymerase involved in late transcription. Oral administration of cHPMPC delayed onset of clinical signs and significantly reduced viral titres in blood and tissues of treated pigs. These results indicate that cHPMPC is a promising compound for further development to control ASFV outbreaks.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Suínos , Animais , Vírus da Febre Suína Africana/genética , Febre Suína Africana/tratamento farmacológico , Febre Suína Africana/prevenção & controle , Nucleosídeos/farmacologia , Nucleosídeos/metabolismo , Antivirais/farmacologia , Antivirais/metabolismo , Replicação Viral , Sus scrofa
10.
Eur J Wildl Res ; 68(6): 69, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36213142

RESUMO

Contact between wild animals and farmed livestock may result in disease transmission with huge financial, welfare and ethical consequences. Conflicts between people and wildlife can also arise when species such as wild boar (Sus scrofa) consume crops or dig up pasture. This is a relatively recent problem in England where wild boar populations have become re-established in the last 20 years following a 500-year absence. The aim of this pilot study was to determine if and how often free-living wild boar visited two commercial pig farms near the Forest of Dean in southwest England. We placed 20 motion-sensitive camera traps at potential entry points to, and trails surrounding, the perimeter of two farmyards housing domestic pigs between August 2019 and February 2021, covering a total of 6030 trap nights. Forty wild boar detections were recorded on one farm spread across 27 nights, with a median (range) of 1 (0 to 7) night of wild boar activity per calendar month. Most of these wild boar detections occurred between ten and twenty metres of housed domestic pigs. No wild boar was detected at the other farm. These results confirm wild boar do visit commercial pig farms, and therefore, there is potential for contact and pathogen exchange between wild boar and domestic pigs. The visitation rates derived from this study could be used to parameterise disease transmission models of pathogens common to domestic pigs and wild boars, such as the African swine fever virus, and subsequently to develop mitigation strategies to reduce unwanted contacts.

11.
Ecol Evol ; 12(6): e9031, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35784084

RESUMO

Predicting the likelihood of wildlife presence at potential wildlife-livestock interfaces is challenging. These interfaces are usually relatively small geographical areas where landscapes show large variation over small distances. Models of wildlife distribution based on coarse data over wide geographical ranges may not be representative of these interfaces. High-resolution data can help identify fine-scale predictors of wildlife habitat use at a local scale and provide more accurate predictions of species habitat use. These data may be used to inform knowledge of interface risks, such as disease transmission between wildlife and livestock, or human-wildlife conflict.This study uses fine-scale habitat use data from wild boar (Sus scrofa) based on activity signs and direct field observations in and around the Forest of Dean in Gloucestershire, England. Spatial logistic regression models fitted using a variant of penalized quasi-likelihood were used to identify habitat-based and anthropogenic predictors of wild boar signs.Our models showed that within the Forest of Dean, wild boar signs were more likely to be seen in spring, in forest-type habitats, closer to the center of the forest and near litter bins. In the area surrounding the Forest of Dean, wild boar signs were more likely to be seen in forest-type habitats and near recreational parks and less likely to be seen near livestock.This approach shows that wild boar habitat use can be predicted using fine-scale data over comparatively small areas and in human-dominated landscapes, while taking account of the spatial correlation from other nearby fine-scale data-points. The methods we use could be applied to map habitat use of other wildlife species in similar landscapes, or of movement-restricted, isolated, or fragmented wildlife populations.

12.
Methods Mol Biol ; 2503: 73-94, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35575887

RESUMO

Genetic manipulation of ASFV has been increasingly used not only for the development of live attenuated vaccines but also as an indispensable tool to further our understanding of the virus-host interactions. Here we present methods for isolation of porcine bone marrow cells and purification of recombinant ASFV using both chromogenic and fluorescent reporters. We also describe in detail a newly developed method to purify genetically modified ASFV using fluorescence-activated cell sorting (FACS).


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Febre Suína Africana/prevenção & controle , Vírus da Febre Suína Africana/genética , Animais , Células da Medula Óssea , Suínos , Vacinas Atenuadas , Proteínas Virais/genética
13.
J Virol ; 96(6): e0189921, 2022 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-35044212

RESUMO

African swine fever virus multigene family (MGF) 360 and 505 genes have roles in suppressing the type I interferon response and in virulence in pigs. The role of the individual genes is poorly understood. Different combinations of these genes were deleted from the virulent genotype II Georgia 2007/1 isolate. Deletion of five copies of MGF 360 genes, MGF360-10L, -11L, -12L, -13L, and -14L, and three copies of MGF505-1R, -2R, and -3R reduced virus replication in macrophages and attenuated virus in pigs. However, only 25% of the immunized pigs were protected against challenge. Deletion of MGF360-12L, -13L, and -14L and MGF505-1R in combination with a negative serology marker, K145R (GeorgiaΔK145RΔMGF(A)), reduced virus replication in macrophages and virulence in pigs, since no clinical signs or virus genome in blood were observed following immunization. Four of six pigs were protected after challenge. In contrast, deletion of MGF360-13L and -14L, MGF505-2R and -3R, and K145R (GeorgiaΔK145RΔMGF(B)) did not reduce virus replication in macrophages. Following immunization of pigs, clinical signs were delayed, but all pigs reached the humane endpoint. Deletion of genes MGF360-12L, MGF505-1R, and K145R reduced replication in macrophages and attenuated virulence in pigs since no clinical signs or virus genome in blood were observed following immunization. Thus, the deletion of MGF360-12L and MGF505-1R, in combination with K145R, was sufficient to dramatically attenuate virus infection in pigs. However, only two of six pigs were protected, suggesting that deletion of additional MGF genes is required to induce a protective immune response. Deletion of MGF360-12L, but not MGF505-1R, from the GeorgiaΔK145R virus reduced virus replication in macrophages, indicating that MGF360-12L was most critical for maintaining high levels of virus replication in macrophages. IMPORTANCE African swine fever has a high socioeconomic impact and no vaccines to aid control. The African swine fever virus (ASFV) has many genes that inhibit the host's interferon response. These include related genes that are grouped into multigene families, including MGF360 and 505. Here, we investigated which MGF360 and 505 genes were most important for viral attenuation and protection against genotype II strains circulating in Europe and Asia. We compared viruses with deletions of MGF genes. Deletion of just two MGF genes in combination with a third gene, K145R, a possible marker for vaccination, is sufficient for virus attenuation in pigs. Deletion of additional MGF360 genes was required to induce higher levels of protection. Furthermore, we showed that the deletion of MGF360-12L, combined with K145R, impairs virus replication in macrophages in culture. Our results have important implications for understanding the roles of the ASFV MGF genes and for vaccine development.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Proteínas Virais , Vacinas Virais , Virulência , Replicação Viral , Febre Suína Africana/prevenção & controle , Febre Suína Africana/virologia , Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/imunologia , Animais , Deleção de Genes , Genótipo , Macrófagos/virologia , Família Multigênica/genética , Suínos , Proteínas Virais/genética , Proteínas Virais/imunologia , Vacinas Virais/genética , Vacinas Virais/imunologia , Virulência/genética , Replicação Viral/genética
14.
J Virol ; 96(5): e0193921, 2022 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-35019713

RESUMO

African swine fever virus (ASFV) has a major global economic impact. With a case fatality in domestic pigs approaching 100%, it currently presents the largest threat to animal farming. Although genomic differences between attenuated and highly virulent ASFV strains have been identified, the molecular determinants for virulence at the level of gene expression have remained opaque. Here, we characterize the transcriptome of ASFV genotype II Georgia 2007/1 (GRG) during infection of the physiologically relevant host cells, porcine macrophages. In this study, we applied cap analysis gene expression sequencing (CAGE-seq) to map th0e 5' ends of viral mRNAs at 5 and 16 h postinfection. A bioinformatics analysis of the sequence context surrounding the transcription start sites (TSSs) enabled us to characterize the global early and late promoter landscape of GRG. We compared transcriptome maps of the GRG isolate and the lab-attenuated BA71V strain that highlighted GRG virulence-specific transcripts belonging to multigene families, including two predicted MGF 100 genes, I7L and I8L. In parallel, we monitored transcriptome changes in the infected host macrophage cells. Of the 9,384 macrophage genes studied, transcripts for 652 host genes were differentially regulated between 5 and 16 h postinfection compared with only 25 between uninfected cells and 5 h postinfection. NF-κB activated genes and lysosome components such as S100 were upregulated, and chemokines such as CCL24, CXCL2, CXCL5, and CXCL8 were downregulated. IMPORTANCE African swine fever virus (ASFV) causes hemorrhagic fever in domestic pigs, with case fatality rates approaching 100% and no approved vaccines or antivirals. The highly virulent ASFV Georgia 2007/1 strain (GRG) was the first isolated when ASFV spread from Africa to the Caucasus region in 2007, then spreading through Eastern Europe and, more recently, across Asia. We used an RNA-based next-generation sequencing technique called CAGE-seq to map the starts of viral genes across the GRG DNA genome. This has allowed us to investigate which viral genes are expressed during early or late stages of infection and how this is controlled, comparing their expression to the nonvirulent ASFV-BA71V strain to identify key genes that play a role in virulence. In parallel, we investigated how host cells respond to infection, which revealed how the ASFV suppresses components of the host immune response to ultimately win the arms race against its porcine host.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Interações entre Hospedeiro e Microrganismos , Macrófagos , Proteínas Virais , Febre Suína Africana/imunologia , Febre Suína Africana/virologia , Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/imunologia , Animais , Perfilação da Expressão Gênica , República da Geórgia , Interações entre Hospedeiro e Microrganismos/imunologia , Macrófagos/imunologia , Macrófagos/virologia , Sus scrofa , Suínos , Transcriptoma , Proteínas Virais/genética , Proteínas Virais/imunologia
15.
J Virol ; 96(1): e0134021, 2022 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-34643433

RESUMO

The limited knowledge on the role of many of the approximately 170 proteins encoded by African swine fever virus restricts progress toward vaccine development. Previously, the DP148R gene was deleted from the genome of genotype I virulent Benin 97/1 isolate. This virus, BeninΔDP148R, induced transient moderate clinical signs after immunization and high levels of protection against challenge. However, the BeninΔDP148R virus and genome persisted in blood over a prolonged period. In the current study, deletion of either EP402R or EP153R genes individually or in combination from BeninΔDP148R genome was shown not to reduce virus replication in macrophages in vitro. However, deletion of EP402R dramatically reduced the period of infectious virus persistence in blood in immunized pigs from 28 to 14 days and virus genome from 59 to 14 days while maintaining high levels of protection against challenge. The additional deletion of EP153R (BeninΔDP148RΔEP153RΔEP402R) further attenuated the virus, and no viremia or clinical signs were observed postimmunization. This was associated with decreased protection and detection of moderate levels of challenge virus in blood. Interestingly, the deletion of EP153R alone from BeninΔDP148R did not result in further virus attenuation and did not reduce the period of virus persistence in blood. These results show that EP402R and EP153R have a synergistic role in reducing clinical signs and levels of virus in blood. IMPORTANCE African swine fever virus (ASFV) causes a disease of domestic pigs and wild boar which results in death of almost all infected animals. The disease has a high economic impact, and no vaccine is available. We investigated the role of two ASFV proteins, called EP402R and EP153R, in determining the levels and length of time virus persists in blood from infected pigs. EP402R causes ASFV particles and infected cells to bind to red blood cells. Deletion of the EP402R gene dramatically reduced virus persistence in blood but did not reduce the level of virus. Deletion of the EP153R gene alone did not reduce the period or level of virus persistence in blood. However, deleting both EP153R and EP402R resulted in undetectable levels of virus in blood and no clinical signs showing that the proteins act synergistically. Importantly, the infected pigs were protected following infection with the wild-type virus that kills pigs.


Assuntos
Vírus da Febre Suína Africana/fisiologia , Febre Suína Africana/virologia , Proteínas Virais/metabolismo , Viremia/virologia , Febre Suína Africana/imunologia , Febre Suína Africana/metabolismo , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Antígenos Virais/genética , Antígenos Virais/imunologia , Antígenos Virais/metabolismo , Biomarcadores , Células Cultivadas , Engenharia Genética , Genótipo , Interações Hospedeiro-Patógeno , Imunização , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Deleção de Sequência , Suínos , Proteínas Virais/genética , Proteínas Virais/imunologia , Vacinas Virais/imunologia , Virulência , Replicação Viral
16.
Viruses ; 13(8)2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34452339

RESUMO

African swine fever virus causes a frequently fatal disease of domestic pigs and wild boar that has a high economic impact across 3 continents. The large double-stranded DNA genome codes for approximately 160 proteins. Many of these have unknown functions and this hinders our understanding of the virus and host interactions. The purpose of the study was to evaluate the role of two virus proteins, K145R and DP148R, in virus replication in macrophages and virulence in pigs. To do this, the DP148R gene, alone or in combination with the K145R gene, was deleted from the virulent genotype II Georgia 2007/1 isolate. Neither of these deletions reduced the ability of the viruses to replicate in porcine macrophages compared to the parental wild-type virus. Pigs infected with GeorgiaΔDP148R developed clinical and post-mortem signs and high viremia, typical of acute African swine fever, and were culled on day 6 post-infection. The additional deletion of the K145R gene delayed the onset of clinical signs and viremia in pigs by 3 days, but pigs showed signs of acute African swine fever and were culled on days 10 or 13 post-infection. The results show that the deletion of DP148R did not attenuate the genotype II Georgia 2007/1 isolate, contrary to the results obtained with the genotype I Benin97/1 isolate. Additional deletion of the K145R gene delayed clinical signs, but infected pigs reached the humane endpoint. The deletion of additional genes would be required to attenuate the virus.


Assuntos
Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/patogenicidade , Febre Suína Africana/virologia , Proteínas Virais/genética , Vírus da Febre Suína Africana/fisiologia , Animais , Deleção de Genes , Macrófagos/virologia , Suínos , Proteínas Virais/metabolismo , Virulência , Replicação Viral
17.
Pathogens ; 10(6)2021 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-34207265

RESUMO

The understanding of the pathogenic mechanisms and the clinicopathological forms caused by currently circulating African swine fever virus (ASFV) isolates is incomplete. So far, most of the studies have been focused on isolates classified within genotypes I and II, the only genotypes that have circulated outside of Africa. However, less is known about the clinical presentations and lesions induced by isolates belonging to the other twenty-two genotypes. Therefore, the early clinicopathological identification of disease outbreaks caused by isolates belonging to, as yet, not well-characterised ASFV genotypes may be compromised, which might cause a delay in the implementation of control measures to halt the virus spread. To improve the pathological characterisation of disease caused by diverse isolates, we have refined the macroscopic and histopathological evaluation protocols to standardise the scoring of lesions. Domestic pigs were inoculated intranasally with different doses (high, medium and low) of ASFV isolate Ken05/Tk1 (genotype X). To complement previous studies, the distribution and severity of macroscopic and histopathological lesions, along with the amount and distribution of viral antigen in tissues, were characterised by applying the new scoring protocols. The intranasal inoculation of domestic pigs with high doses of the Ken05/Tk1 isolate induced acute forms of ASF in most of the animals. Inoculation with medium doses mainly induced acute forms of disease. A less severe but longer clinical course, typical of subacute forms, characterised by the presence of more widespread and severe haemorrhages and oedema, was observed in one pig inoculated with the medium dose. The severity of vascular lesions (haemorrhages and oedema) induced by high and medium doses was not associated with the amount of virus antigen detected in tissues, therefore these might be attributed to indirect mechanisms not evaluated in the present study. The absence of clinical signs, lesions and detectable levels of virus genome or antigen in blood from the animals inoculated with the lowest dose ruled out the existence of possible asymptomatic carriers or persistently infected pigs, at least for the 21 days period of the study. The results corroborate the moderate virulence of the Ken05/Tk1 isolate, as well as its capacity to induce both the acute and, occasionally, subacute forms of ASF when high and medium doses were administered intranasally.

18.
Trials ; 22(1): 240, 2021 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-33794962

RESUMO

BACKGROUND: Restricted and repetitive behaviours vary greatly across the autism spectrum, and although not all are problematic some can cause distress and interfere with learning and social opportunities. We have, alongside parents, developed a parent group based intervention for families of young children with autism, which aims to offer support to parents and carers; helping them to recognise, understand and learn how to respond to their child's challenging restricted repetitive behaviours. METHODS: The study is a clinical and cost-effectiveness, multi-site randomised controlled trial of the Managing Repetitive Behaviours (MRB) parent group intervention versus a psychoeducation parent group Learning About Autism (LAA) (n = 250; 125 intervention/125 psychoeducation; ~ 83/site) for parents of young children aged 3-9 years 11 months with a diagnosis of autism. All analyses will be done under intention-to-treat principle. The primary outcome at 24 weeks will use generalised estimating equation (GEE) to compare proportion of children with improved RRB between the MRB group and the LAA group. The GEE model will account for the clustering of children by parent groups using exchangeable working correlation. All secondary outcomes will be analysed in a similar way using appropriate distribution and link function. The economic evaluation will be conducted from the perspective of both NHS costs and family access to local community services. A 'within trial' cost-effectiveness analysis with results reported as the incremental cost per additional child achieving at least the target improvement in CGI-I scale at 24 weeks. DISCUSSION: This is an efficacy trial to investigate the clinical and cost-effectiveness of a parent group based intervention designed to help parents understand and manage their child's challenging RRB. If found to be effective, this intervention has the potential to improve the well-being of children and their families, reduce parental stress, greatly enhance community participation and potential for learning, and improve longer-term outcomes. TRIAL REGISTRATION: Trial ID: ISRCTN15550611 Date registered: 07/08/2018. Sponsor and Monitor: Cumbria, Northumberland, Tyne and Wear NHS Foundation Trust R&D Manager Lyndsey Dixon, Address: St Nicholas Hospital, Jubliee Road, Gosforth, Newcastle upon Tyne NE3 3XT, lyndsey.dixon@cntw.nhs.uk , Tel: 0191 246 7222.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Transtorno do Espectro Autista/diagnóstico , Transtorno do Espectro Autista/terapia , Criança , Pré-Escolar , Análise Custo-Benefício , Humanos , Relações Pais-Filho , Pais , Ensaios Clínicos Controlados Aleatórios como Assunto
19.
PeerJ ; 8: e10221, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33173619

RESUMO

Wild animals are the source of many pathogens of livestock and humans. Concerns about the potential transmission of economically important and zoonotic diseases from wildlife have led to increased surveillance at the livestock-wildlife interface. Knowledge of the types, frequency and duration of contacts between livestock and wildlife is necessary to identify risk factors for disease transmission and to design possible mitigation strategies. Observing the behaviour of many wildlife species is challenging due to their cryptic nature and avoidance of humans, meaning there are relatively few studies in this area. Further, a consensus on the definition of what constitutes a 'contact' between wildlife and livestock is lacking. A systematic review was conducted to investigate which livestock-wildlife contacts have been studied and why, as well as the methods used to observe each species. Over 30,000 publications were screened, of which 122 fulfilled specific criteria for inclusion in the analysis. The majority of studies examined cattle contacts with badgers or with deer; studies involving wild pig contacts with cattle or with domestic pigs were the next most frequent. There was a range of observational methods including motion-activated cameras and global positioning system collars. As a result of the wide variation and lack of consensus in the definitions of direct and indirect contacts, we developed a unified framework to define livestock-wildlife contacts that is sufficiently flexible to be applied to most wildlife and livestock species for non-vector-borne diseases. We hope this framework will help standardise the collection and reporting of contact data; a valuable step towards being able to compare the efficacy of wildlife-livestock observation methods. In doing so, it may aid the development of better disease transmission models and improve the design and effectiveness of interventions to reduce or prevent disease transmission.

20.
Viruses ; 12(6)2020 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-32516890

RESUMO

African swine fever (ASF) is a devastating disease in pigs, with no vaccines for control. The genetic manipulation of African swine fever virus (ASFV) is often tedious and time consuming. Here, we describe a method to manipulate the virus genome to produce gene deletion viruses in a much-reduced time. This method combines the conventional homologous recombination with fluorescent-activated cells sorting (FACS), to isolate and purify viruses expressing fluorescent reporter genes. With three rounds of single cell isolation via FACS and two rounds of limiting dilution, we deleted two additional genes, EP153R and EP402R, from Benin 97/1 ASFV lacking the DP148R gene. By combining different fluorescent markers, this method has the potential to greatly facilitate studies on understanding ASFV gene functions and develop candidate live-attenuated vaccines.


Assuntos
Vírus da Febre Suína Africana/genética , Febre Suína Africana/virologia , Vírus da Febre Suína Africana/imunologia , Animais , Engenharia Genética , Genoma Viral , Recombinação Genética , Suínos , Proteínas Virais/genética , Proteínas Virais/imunologia , Vacinas Virais/genética , Vacinas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA