Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.360
Filtrar
1.
Virol J ; 21(1): 104, 2024 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-38702807

RESUMO

BACKGROUND: Rift Valley fever (RVF) is a zoonotic viral disease of increasing intensity among humans in Africa and the Arabian Peninsula. In Uganda, cases reported prior to 2016 were mild or not fully documented. We report in this paper on the severe morbidity and hospital-based mortality of human cases in Uganda. METHODS: Between November 2017 and March 2020 human cases reported to the Uganda Virus Research Institute (UVRI) were confirmed by polymerase chain reaction (PCR). Ethical and regulatory approvals were obtained to enrol survivors into a one-year follow-up study. Data were collected on socio-demographics, medical history, laboratory tests, potential risk factors, and analysed using Stata software. RESULTS: Overall, 40 cases were confirmed with acute RVF during this period. Cases were not geographically clustered and nearly all were male (39/40; 98%), median age 32 (range 11-63). The median definitive diagnosis time was 7 days and a delay of three days between presumptive and definitive diagnosis. Most patients (31/40; 78%) presented with fever and bleeding at case detection. Twenty-eight (70%) cases were hospitalised, out of whom 18 (64%) died. Mortality was highest among admissions in regional referral (11/16; 69%) and district (4/5; 80%) hospitals, hospitalized patients with bleeding at case detection (17/27; 63%), and patients older than 44 years (9/9; 100%). Survivors mostly manifested a mild gastro-intestinal syndrome with nausea (83%), anorexia (75%), vomiting (75%), abdominal pain (50%), and diarrhoea (42%), and prolonged symptoms of severe disease including jaundice (67%), visual difficulties (67%), epistaxis (50%), haemoptysis (42%), and dysentery (25%). Symptom duration varied between two to 120 days. CONCLUSION: RVF is associated with high hospital-based mortality, severe and prolonged morbidity among humans that present to the health care system and are confirmed by PCR. One-health composite interventions should be developed to improve environmental and livestock surveillance, prevent infections, promptly detect outbreaks, and improve patient outcomes.


Assuntos
Febre do Vale de Rift , Humanos , Uganda/epidemiologia , Febre do Vale de Rift/mortalidade , Febre do Vale de Rift/epidemiologia , Masculino , Adulto , Pessoa de Meia-Idade , Adolescente , Feminino , Adulto Jovem , Criança , Vírus da Febre do Vale do Rift/genética , Mortalidade Hospitalar , Morbidade , Fatores de Risco
2.
Virulence ; 15(1): 2329447, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38548679

RESUMO

MicroRNAs (miRNAs) are small non-coding RNAs that regulate the post-transcriptional expression of target genes. Virus-encoded miRNAs play an important role in the replication of viruses, modulate gene expression in both the virus and host, and affect their persistence and immune evasion in hosts. This renders viral miRNAs as potential targets for therapeutic applications, especially against pathogenic viruses that infect humans and animals. Rift Valley fever virus (RVFV) is a mosquito-borne zoonotic RNA virus that causes severe disease in both humans and livestock. High mortality among newborn lambs and abortion storms are key characteristics of an RVF outbreak. To date, limited information is available on RVFV-derived miRNAs. In this study, computational methods were used to analyse the RVFV genome for putative pre-miRNA genes, which were then analysed for the presence of mature miRNAs. We detected 19 RVFV-encoded miRNAs and identified their potential mRNAs targets in sheep (Ovis aries), the most susceptible host. The identification of significantly enriched O. aries genes in association with RVFV miRNAs will help elucidate the molecular mechanisms underlying RVFV pathogenesis and potentially uncover novel drug targets for RVFV.


Assuntos
Culicidae , MicroRNAs , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Humanos , Gravidez , Feminino , Animais , Ovinos/genética , Vírus da Febre do Vale do Rift/genética , Febre do Vale de Rift/genética , Febre do Vale de Rift/epidemiologia , Culicidae/genética , Surtos de Doenças , MicroRNAs/genética
3.
J Gen Virol ; 105(3)2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38546100

RESUMO

Rift Valley fever virus (RVFV) is an emerging arboviral disease with pandemic potential. While infection is often self-limiting, a subset of individuals may develop late-onset encephalitis, accounting for up to 20 % of severe cases. Importantly, individuals displaying neurologic disease have up to a 53 % case fatality rate, yet the neuropathogenesis of RVFV infection remains understudied. In this study, we evaluated whether ex vivo postnatal rat brain slice cultures (BSCs) could be used to evaluate RVFV infection in the central nervous system. BSCs mounted an inflammatory response after slicing, which resolved over time, and they were viable in culture for at least 12 days. Infection of rat BSCs with pathogenic RVFV strain ZH501 induced tissue damage and apoptosis over 48 h. Viral replication in BSCs reached up to 1×107 p.f.u. equivalents/ml, depending on inoculation dose. Confocal immunofluorescent microscopy of cleared slices confirmed direct infection of neurons as well as activation of microglia and astrocytes. Further, RVFV-infected rat BSCs produced antiviral cytokines and chemokines, including MCP-1 and GRO/KC. This study demonstrates that rat BSCs support replication of RVFV for ex vivo studies of neuropathogenesis. This allows for continued and complementary investigation into RVFV infection in an ex vivo postnatal brain slice culture format.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Ratos , Animais , Vírus da Febre do Vale do Rift/fisiologia , Citocinas , Encéfalo , Morte Celular
4.
PLoS Pathog ; 20(3): e1012093, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38512999

RESUMO

Rift Valley fever virus (RVFV) is a viral zoonosis that causes severe disease in ruminants and humans. The nonstructural small (NSs) protein is the primary virulence factor of RVFV that suppresses the host's antiviral innate immune response. Bioinformatic analysis and AlphaFold structural modeling identified four putative LC3-interacting regions (LIR) motifs (NSs 1-4) in the RVFV NSs protein, which suggest that NSs interacts with the host LC3-family proteins. Using, isothermal titration calorimetry, X-ray crystallography, co-immunoprecipitation, and co-localization experiments, the C-terminal LIR motif (NSs4) was confirmed to interact with all six human LC3 proteins. Phenylalanine at position 261 (F261) within NSs4 was found to be critical for the interaction of NSs with LC3, retention of LC3 in the nucleus, as well as the inhibition of autophagy in RVFV infected cells. These results provide mechanistic insights into the ability of RVFV to overcome antiviral autophagy through the interaction of NSs with LC3 proteins.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Humanos , Vírus da Febre do Vale do Rift/metabolismo , Proteínas não Estruturais Virais/metabolismo , Autofagia , Antivirais/metabolismo
5.
PLoS Negl Trop Dis ; 18(3): e0011756, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38427694

RESUMO

Rift Valley fever (RVF) is a mosquito-borne viral zoonosis caused by the Rift Valley fever virus (RVFV) that can infect domestic and wild animals. Although the RVFV transmission cycle has been well documented across Africa in savanna ecosystems, little is known about its transmission in tropical rainforest settings, particularly in Central Africa. We therefore conducted a survey in northeastern Gabon to assess RVFV circulation among wild and domestic animals. Among 163 wildlife samples tested using RVFV-specific RT-qPCR, four ruminants belonging to subfamily Cephalophinae were detected positive. The phylogenetic analysis revealed that the four RVFV sequences clustered together with a virus isolated in Namibia within the well-structured Egyptian clade. A cross-sectional survey conducted on sheep, goats and dogs living in villages within the same area determined the IgG RVFV-specific antibody prevalence using cELISA. Out of the 306 small ruminants tested (214 goats, 92 sheep), an overall antibody prevalence of 15.4% (95% CI [11.5-19.9]) was observed with a higher rate in goats than in sheep (20.1% versus 3.3%). RVFV-specific antibodies were detected in a single dog out of the 26 tested. Neither age, sex of domestic animals nor season was found to be significant risk factors of RVFV occurrence. Our findings highlight sylvatic circulation of RVFV for the first time in Gabon. These results stress the need to develop adequate surveillance plan measures to better control the public health threat of RVFV.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Ovinos , Cães , Animais Domésticos , Animais Selvagens , Gabão/epidemiologia , Estudos Transversais , Ecossistema , Filogenia , Ruminantes , Cabras , Anticorpos Antivirais , Florestas , Estudos Soroepidemiológicos
6.
Sci Rep ; 14(1): 3904, 2024 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-38365824

RESUMO

Rift Valley Fever (RVF) is a zoonosis transmitted by Aedes and Culex mosquitoes, and is considered a priority pathogen by the WHO. RVF epidemics mostly occur in Africa and can decimate livestock herds, causing significant economic losses and posing health risks for humans. RVF transmission is associated with the occurrence of El Niño events that cause floods in eastern Africa and favour the emergence of mosquitoes in wetlands. Different risk models have been developed to forecast RVF transmission risk but very few studies have validated models at pan-African scale. This study aims to validate the skill of the Liverpool Rift Valley Fever model (LRVF) in reproducing RVF epidemics over Africa and to explore the relationship between simulated climatic suitability for RVF transmission and large-scale climate modes of variability such as the El Niño Southern Oscillation (ENSO) and the Dipole Mode Index (DMI). Our results show that the LRVF model correctly simulates RVF transmission hotspots and reproduces large epidemics that affected African countries. LRVF was able to correctly reproduce major RVF epidemics in Somalia, Kenya, Zambia and to a lesser extent for Mauritania and Senegal. The positive phases of ENSO and DMI are associated with an increased risk of RVF over the Horn of Africa, with important time lags. Following research activities should focus on the development of predictive modelling systems at different time scales.


Assuntos
Aedes , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Humanos , Febre do Vale de Rift/epidemiologia , Surtos de Doenças , Zoonoses/epidemiologia , Quênia/epidemiologia
8.
Mol Pharm ; 21(3): 1342-1352, 2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-38295278

RESUMO

Rift Valley fever virus (RVFV) could cause an emergency illness characterized by fever, muscle pain, and even death in humans or ruminants. However, there are no approved antiviral drugs that prevent or treat RVFV infection. While therapeutic antibodies have shown promising potential for prevention or treatment in several studies, many studies are ongoing, especially in the field of infectious diseases. Among these studies, the mRNA-LNP platform shows great potential for application, following the COVID-19 pandemic. Previously, we have obtained a neutralizing antibody against RVFV, which was named A38 protein and verified to have a high binding and neutralization ability. In this study, we aimed to identify an effectively optimized sequence and expressed the prioritized mRNA-encoded antibody in vitro. Notably, we effectively expressed mRNA-encoded protein and used the mRNA-LNP platform to generate A38-mRNA-LNP. Pharmacokinetic experiments were conducted in vivo and set up in two groups of mRNA-A38 group and A38 protein group, which were derived from mRNA-LNP and plasmid DNA-expressed proteins, respectively. A38-mRNA-LNPs were administrated by intramuscular injection, A38 proteins were administrated by intravenous administration, and their unique ability to maintain long-lasting protein concentrations by mRNA-encoded protein was demonstrated with the mRNA-encoded protein providing a longer circulating half-life compared to injection of the free A38 protein. These preclinical data on the mRNA-encoded antibody highlighted its potential to prevent infectious diseases in the future.


Assuntos
Doenças Transmissíveis , Lipossomos , Nanopartículas , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Humanos , Vírus da Febre do Vale do Rift/genética , Febre do Vale de Rift/prevenção & controle , Pandemias , Anticorpos Antivirais
9.
Sci Total Environ ; 917: 170302, 2024 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-38272089

RESUMO

BACKGROUND: Rift valley fever (RVF) is listed as one of prioritized diseases by WHO. This study aims to describe RVF virus' landscape distribution globally, and to insight dynamics change of its evolution, prevalence, and outbreaks in the process of breaking geographical barriers. METHODS: A systematic literature review and meta-analyses was conducted to estimate RVF prevalence by hosts using a random-effect model. Molecular clock-based phylogenetic analyses were performed to estimate RVF virus nucleotide substitution rates using nucleotide sequences in NCBI database. RVF virus prevalence, nucleotide substitution rates, and outbreaks were compared before and after breaking geographical barriers twice, respectively. RESULTS: RVF virus was reported from 26 kinds of hosts covering 48 countries from 1930 to 2022. Since RVF broke geographical barriers, (1) nucleotide substitution rates significantly increased after firstly spreading out of Africa in 2000, (2) prevalence in humans significantly increased from 1.92 % (95 % CI: 0.86-3.25 %) to 3.03 % (95 % CI: 2.09-4.12 %) after it broke Sahara Desert geographical barriers in 1977, and to 5.24 % (95 % CI: 3.81-6.82 %) after 2000, (3) RVF outbreaks in humans and the number of wildlife hosts presented increasing trends. RVF virus spillover may exist between bats and humans, and accelerate viral substitution rates in humans. During outbreaks, the RVF virus substitution rates accelerated in humans. 60.00 % RVF outbreaks occurred 0-2 months after floods and (or) heavy rainfall. CONCLUSION: RVF has the increasing risk to cause pandemics, and global collaboration on "One Health" is needed to prevent potential pandemics.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Humanos , Prevalência , Filogenia , Febre do Vale de Rift/epidemiologia , Febre do Vale de Rift/prevenção & controle , Surtos de Doenças , Nucleotídeos
10.
J Med Primatol ; 53(1): e12687, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38083993

RESUMO

We tested for Rift Valley fever virus (RVFV) from at least 15 species of non-human primates. RVFV IgG/IgM antibodies were detected in 3.7% (2 out of 53) of chimpanzees (Pan troglodytes) and in 1.4% (1 out of 72) of unidentified non-human primate species. This study was the first investigation of RVFV in monkeys in Cameroon.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Febre do Vale de Rift/diagnóstico , Camarões , Anticorpos Antivirais , Primatas , Estudos Soroepidemiológicos
11.
Methods Mol Biol ; 2733: 101-113, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38064029

RESUMO

Rift Valley fever virus (RVFV) is an important mosquito-borne virus that can cause severe disease manifestations in humans including ocular damage, vision loss, late-onset encephalitis, and hemorrhagic fever. In ruminants, RVFV can cause high mortality rates in young animals and high rates of abortion in pregnant animals resulting in an enormous negative impact on the economy of affected regions. To date, no licensed vaccines in humans or anti-RVFV therapeutics for animal or human use are available. The development of reverse genetics has facilitated the generation of recombinant infectious viruses that serve as powerful tools for investigating the molecular biology and pathogenesis of RVFV. Infectious recombinant RVFV can be rescued entirely from cDNAs containing predetermined mutations in their genomes to investigate virus-host interactions and mechanisms of pathogenesis and generate live-attenuated vaccines. In this chapter, we will describe the experimental procedures for the implementation of RVFV reverse genetics.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Humanos , Vírus da Febre do Vale do Rift/genética , Febre do Vale de Rift/genética , Febre do Vale de Rift/prevenção & controle , Genética Reversa , Vacinas Atenuadas/genética , Mutação
12.
Viruses ; 15(12)2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-38140610

RESUMO

Rift Valley fever virus (RVFV) is a mosquito-borne zoonotic pathogen causing disease in livestock and humans. Whilst initially restricted to the African continent, recent spread to the Arabian Peninsula has highlighted the likelihood of entry into new regions. Due to the absence of a regulatory-approved human vaccine, work is ongoing to develop and assess countermeasures. As such, small animal models play a pivotal role in providing information on disease pathogenesis and elucidating which intervention strategies confer protection. To develop and establish the BALB/c mouse model, we challenged mice with RVFV grown from two separate cell lines: one derived from mosquitoes (C6/36) and the other mammalian derived (Vero E6). Following infection, we assessed the clinical course of disease progression at days 1 and 3 post-challenge and evaluated viral tropism and immune analytes. The results demonstrated that RVFV infection was affected by the cell line used to propagate the challenge virus, with those grown in insect cells resulting in a more rapid disease progression. The lowest dose that caused uniform severe disease remained the same across both virus preparations. In addition, to demonstrate reproducibility, the lowest dose was used for a subsequent infection study using male and female animals. The results further demonstrated that male mice succumbed to infection more rapidly than their female counterparts. Our results establish an RVFV mouse model and key parameters that affect the course of disease progression in BALB/c mice.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Masculino , Feminino , Humanos , Animais , Camundongos , Camundongos Endogâmicos BALB C , Reprodutibilidade dos Testes , Progressão da Doença , Mamíferos
13.
Sci Rep ; 13(1): 18688, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37907670

RESUMO

Rift Valley fever (RVF) is a mosquito-borne zoonotic disease causing acute hemorrhagic fever. Accurate identification of mutations and phylogenetic characterization of RVF virus (RVFV) require whole-genome analysis. Universal primers to amplify the entire RVFV genome from clinical samples with low copy numbers are currently unavailable. Thus, we aimed to develop universal primers applicable for all known RVFV strains. Based on the genome sequences available from public databases, we designed eight pairs of universal PCR primers covering the entire RVFV genome. To evaluate primer universality, four RVFV strains (ZH548, Kenya 56 (IB8), BIME-01, and Lunyo), encompassing viral phylogenetic diversity, were chosen. The nucleic acids of the test strains were chemically synthesized or extracted via cell culture. These RNAs were evaluated using the PCR primers, resulting in successful amplification with expected sizes (0.8-1.7 kb). Sequencing confirmed that the products covered the entire genome of the RVFV strains tested. Primer specificity was confirmed via in silico comparison against all non-redundant nucleotide sequences using the BLASTn alignment tool in the NCBI database. To assess the clinical applicability of the primers, mock clinical specimens containing human and RVFV RNAs were prepared. The entire RVFV genome was successfully amplified and sequenced at a viral concentration of 108 copies/mL. Given the universality, specificity, and clinical applicability of the primers, we anticipate that the RVFV universal primer pairs and the developed method will aid in RVFV phylogenomics and mutation detection.


Assuntos
Febres Hemorrágicas Virais , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Humanos , Vírus da Febre do Vale do Rift/genética , Filogenia , Sequenciamento Completo do Genoma , RNA
14.
PLoS One ; 18(11): e0292085, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38019849

RESUMO

BACKGROUND: In Africa, little is known about the role of telephone hotlines in outbreak response. We systematically reviewed the role and effectiveness of hotlines on outbreak response in Africa. METHOD: We used the Cochrane handbook and searched five databases. The protocol was registered on PROSPERO (CRD42021247141). Medline, Embase, PsycINFO, Global Health and Web of Science were searched from 30 June 2020 to August 2020 for studies on the use of telephone hotlines in outbreak response in Africa published between January 1995 and August 2020. The search was also repeated on 16 September 2022. Data on effectiveness (alerts generated, cases confirmed) were extracted from peer-reviewed studies. Meta-analysis of alerts generated, and proportion of cases confirmed was done using the random effects model. The quality of studies was assessed using the Joanna Briggs Institute (JBI) tools. The heterogeneity and publication bias were assessed using the Galbraith and funnel plots, respectively. RESULTS: Our search yielded 1251 non-duplicate citations that were assessed. 41 full texts were identified, and 21 studies were included in the narrative synthesis, while 12 were included in the meta-analysis. The hotlines were local (seven studies) or national (three studies). A combination of a local and national hotline was used in one study. The hotlines were set up for unusual respiratory events (one study), polio (one study), Ebola (10 studies), COVID-19 (two studies), malaria (one study), influenza-like illnesses (ILI) (one study) and rift valley fever in livestock (one study). Hotlines were mainly used for outbreak surveillance at the local level. A total of 332,323 alerts were generated, and 67,658 met the case definition, corresponding to an overall pooled proportion of alerts generated(sensitivity) of 38% (95%CI: 24-52%). The sensitivity was 41% (95% CI: 24-59%) for local hotlines and 26%(95%CI:5-47%) for national hotlines. Hotlines were also used for surveillance of rift valley fever in livestock (one study) vaccination promotion (one study), death reporting (five studies), rumour tracking and fighting misinformation (two studies) and community engagement (five studies). The studies were of low to moderate quality with high publication bias and heterogeneity(I2 = 99%). The heterogeneity was not explained by the sample size. CONCLUSION: These data suggest that telephone hotlines can be effective in outbreak disease surveillance in Africa. Further implementation research is needed to scale up telephone hotlines in rural areas.


Assuntos
Febre do Vale de Rift , Viroses , Humanos , Animais , Linhas Diretas , Surtos de Doenças/prevenção & controle , África/epidemiologia
15.
Viruses ; 15(11)2023 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-38005818

RESUMO

Mosquitoes in the genera Aedes and Culex are vectors of Rift Valley fever virus (RVFV), which emerges in periodic epidemics in Africa and Saudi Arabia. Factors that influence the transmission dynamics of RVFV are not well characterized. To address this, we interrogated mosquito host-signaling responses through analysis of differentially expressed genes (DEGs) in two mosquito species with marked differences in RVFV vector competence: Aedes aegypti (Aae, low competence) and Culex tarsalis (Cxt, high competence). Mosquito-host transcripts related to three different signaling pathways were investigated. Selected genes from the Wingless (Wg, WNT-beta-catenin) pathway, which is a conserved regulator of cell proliferation and differentiation, were assessed. One of these, dishevelled (DSH), differentially regulates progression/inhibition of the WNT and JNK (c-Jun N-terminal Kinase) pathways. A negative regulator of the JNK-signaling pathway, puckered, was also assessed. Lastly, Janus kinase/signal transducers and activators of transcription (JAK-STAT) are important for innate immunity; in this context, we tested domeless levels. Here, individual Aae and Cxt were exposed to RVFV MP-12 via oral bloodmeals and held for 14 days. Robust decreases in DEGs in both Aae and Cxt were observed. In particular, Aae DSH expression, but not Cxt DSH, was correlated to the presence/absence of viral RNA at 14 days post-challenge (dpc). Moreover, there was an inverse relationship between the viral copy number and aaeDSH expression. DSH silencing resulted in increased viral copy numbers compared to controls at 3 dpc, consistent with a role for aaeDSH in antiviral immunity. Analysis of cis-regulatory regions for the genes of interest revealed clues to upstream regulation of these pathways.


Assuntos
Aedes , Culex , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Vírus da Febre do Vale do Rift/genética , Mosquitos Vetores
16.
Viruses ; 15(11)2023 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-38005861

RESUMO

Rift Valley fever virus (RVFV) is considered to be a high biodefense priority based on its threat to livestock and its ability to cause human hemorrhagic fever. RVFV-infected livestock are also a significant risk factor for human infection by direct contact with contaminated blood, tissues, and aborted fetal materials. Therefore, livestock vaccination in the affected regions has the direct dual benefit and one-health approach of protecting the lives of millions of animals and eliminating the risk of severe and sometimes lethal human Rift Valley fever (RVF) disease. Recently, we have developed a bovine herpesvirus type 1 (BoHV-1) quadruple gene mutant virus (BoHV-1qmv) vector that lacks virulence and immunosuppressive properties due to the deletion of envelope proteins UL49.5, glycoprotein G (gG), gE cytoplasmic tail, and US9 coding sequences. In the current study, we engineered the BoHV-1qmv further by incorporating a chimeric gene sequence to express a proteolytically cleavable polyprotein: RVFV envelope proteins Gn ectodomain sequence fused with bovine granulocyte-macrophage colony-stimulating factor (GMCSF) and Gc, resulting in a live BoHV-1qmv-vectored subunit vaccine against RVFV for livestock. In vitro, the resulting recombinant virus, BoHV-1qmv Sub-RVFV, was replicated in cell culture with high titers. The chimeric Gn-GMCSF and Gc proteins expressed by the vaccine virus formed the Gn-Gc complex. In calves, the BoHV-1qmv Sub-RVFV vaccination was safe and induced moderate levels of the RVFV vaccine strain, MP12-specific neutralizing antibody titers. Additionally, the peripheral blood mononuclear cells from the vaccinated calves had six-fold increased levels of interferon-gamma transcription compared with that of the BoHV-1qmv (vector)-vaccinated calves when stimulated with heat-inactivated MP12 antigen in vitro. Based on these findings, we believe that a single dose of BoHV-1qmv Sub-RVFV vaccine generated a protective RVFV-MP12-specific humoral and cellular immune response. Therefore, the BoHV-1qmv sub-RVFV can potentially be a protective subunit vaccine for cattle against RVFV.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Vacinas Virais , Animais , Bovinos , Humanos , Vírus da Febre do Vale do Rift/genética , Anticorpos Neutralizantes , Anticorpos Antivirais , Leucócitos Mononucleares , Imunidade Celular , Vacinas Atenuadas/genética , Vacinas de Subunidades Antigênicas
17.
Viruses ; 15(11)2023 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-38005928

RESUMO

Rift Valley fever phlebovirus (RVFV) is a zoonotic pathogen that causes Rift Valley fever (RVF) in livestock and humans. Currently, there is no licensed human vaccine or antiviral drug to control RVF. Although multiple species of animals and humans are vulnerable to RVFV infection, host factors affecting susceptibility are not well understood. To identify the host factors or genes essential for RVFV replication, we conducted CRISPR-Cas9 knockout screening in human A549 cells. We then validated the putative genes using siRNA-mediated knock-downs and CRISPR-Cas9-mediated knock-out studies. The role of a candidate gene in the virus replication cycle was assessed by measuring intracellular viral RNA accumulation, and the virus titers were analyzed using plaque assay or TCID50 assay. We identified approximately 900 genes with potential involvement in RVFV infection and replication. Further evaluation of the effect of six genes on viral replication using siRNA-mediated knock-downs revealed that silencing two genes (WDR7 and LRP1) significantly impaired RVFV replication. For further analysis, we focused on the WDR7 gene since the role of the LRP1 gene in RVFV replication was previously described in detail. WDR7 knockout A549 cell lines were generated and used to dissect the effect of WRD7 on a bunyavirus, RVFV, and an orthobunyavirus, La Crosse encephalitis virus (LACV). We observed significant effects of WDR7 knockout cells on both intracellular RVFV RNA levels and viral titers. At the intracellular RNA level, WRD7 affected RVFV replication at a later phase of its replication cycle (24 h) when compared with the LACV replication, which was affected in an earlier replication phase (12 h). In summary, we identified WDR7 as an essential host factor for the replication of two different viruses, RVFV and LACV, both of which belong to the Bunyavirales order. Future studies will investigate the mechanistic role through which WDR7 facilitates phlebovirus replication.


Assuntos
Phlebovirus , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Humanos , Vírus da Febre do Vale do Rift/genética , Phlebovirus/genética , Replicação Viral , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Proteínas Adaptadoras de Transdução de Sinal
18.
Prev Vet Med ; 221: 106071, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37984160

RESUMO

To assess pastoralists' and agropastoralists' knowledge on Rift Valley fever (RVF), participatory epidemiological studies were conducted with 215 livestock keepers and 27 key informants in Napak, Butebo, Isingiro and Lyantonde districts, Uganda, between January and February 2022. Livestock keepers in all four districts had knowledge of RVF and even had local names or descriptions for it. Pastoralists and agropastoralists possessed valuable knowledge of RVF clinical descriptions and epidemiological risk factors such as the presence of infected mosquitoes, living in flood-prone areas, and excessive rainfall. RVF was ranked among the top ten most important cattle diseases. Pastoralists called RVF Lonyang, symbolizing a disease associated with jaundice, high fever, abortions in pregnant cows, and sudden death in calves. Key informants identified infected domestic animals, the presence of infected mosquitoes, livestock movement and trade, and infected wild animals as risk pathways for the introduction of RVF into an area. Drinking raw blood and milk was perceived as the most likely pathway for human exposure to RVF virus; while the highest consequence was high treatment costs. The results indicate that pastoralists provided key epidemiological information that could be essential for designing an effective national RVF surveillance and early warning system.


Assuntos
Culicidae , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Gravidez , Feminino , Animais , Bovinos , Humanos , Febre do Vale de Rift/epidemiologia , Uganda/epidemiologia , Animais Domésticos , Fatores de Risco , Gado
19.
Trop Anim Health Prod ; 55(5): 345, 2023 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-37789189

RESUMO

Rift Valley fever (RVF) is a mosquito-borne viral disease that affects a variety of domestic animals, including cattle, sheep, goats, and camels, and has zoonotic potential. Although the rift valley fever virus (RVFV) is usually asymptomatic in camels, it can induce abortion in some pregnant animals. In the current study, a serosurvey was carried out to investigate the prevalence of RVFV antibodies and related risk factors in camels from four Egyptian governorates. A total of 400 serum samples were examined for anti-RVFV antibodies using a competitive enzyme-linked immunosorbent assay (c-ELISA). The results revealed that the overall prevalence of RVF among examined camels was 21.5% and the disease was more prevalent in Kafr ElSheikh governorate in Nile Delta of Egypt. In addition, the age group of camels with more than 5 years (OR=4.49, 95%CI: 1.39-14.49), the female sex (OR=3.38, 95%CI: 1.51-7.58), the emaciated animals (OR=1.52, 95%CI: 0.86-2.66), the summer season's infection (OR=5.98, 95%CI: 1.79-19.93), the presence of mosquitoes (OR= 2.88, 95%CI: 1.39-5.95), and the absence of mosquitoes control (OR=3.97, 95%CI: 2.09-7.57) were identified as risk factors for RVFV infection. The results of this study support knowledge on the risk factors for RVFV infection and demonstrate that camels raising in Egypt have RVFV antibodies. Quarantine measures or vaccination program should be implemented to reduce the likelihood of RVFV introduction, dissemination among susceptible animals, and ultimately transmission to humans.


Assuntos
Doenças dos Bovinos , Doenças das Cabras , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Doenças dos Ovinos , Humanos , Gravidez , Bovinos , Animais , Feminino , Ovinos , Camelus , Egito/epidemiologia , Estudos Soroepidemiológicos , Febre do Vale de Rift/epidemiologia , Cabras , Anticorpos Antivirais , Doenças dos Bovinos/epidemiologia , Doenças das Cabras/epidemiologia , Doenças dos Ovinos/epidemiologia
20.
Virol J ; 20(1): 221, 2023 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-37789354

RESUMO

BACKGROUND: The live-attenuated Rift Valley Fever Smithburn (SB) vaccine is one of the oldest products widely used in ruminants for control of RVF infections. Vaccinations with RVF Smithburn result in residual pathogenic effect and is limited for use in non-pregnant animals. Commercially available RVFV inactivated vaccines are considered safer options to control the disease. These products are prepared from virulent RVFV isolates and present occupational safety concerns. This research study evaluates the ability of an inactivated SB vaccine strain to elicit neutralising antibody response in sheep. METHODS: The RVF Smithburn vaccine was inactivated with binary ethylenimine at 37 °C. Inactivated RVFV cultures were adjuvanted with Montande™ Gel-01 and aluminium hydroxide (Al (OH)3) gel for immunogenicity and safety determination in sheep. The commercial RVF inactivated vaccine and a placebo were included as positive and negative control groups, respectively. RESULTS: Inactivated RVFV vaccine formulations were safe with all animals showing no clinical signs of RVFV infection and temperature reactions following prime-boost injections. The aluminium hydroxide formulated vaccine induced an immune response as early as 14 days post primary vaccination with neutralising antibody titre of 1:20 and a peak antibody titre of 1:83 was reached on day 56. A similar trend was observed in the animal group vaccinated with the commercial inactivated RVF vaccine obtaining the highest antibody titre of 1:128 on day 56. The neutralizing antibody levels remained within a threshold for the duration of the study. Merino sheep vaccinated with Montanide™ Gel-01-Smithburn were characterised with overall lower immune response when compared to aluminium hydroxide vaccine emulsions. CONCLUSIONS: These finding suggests that the inactivated RVF Smithburn vaccine strain adjuvanted with aluminium-hydroxide can be used an alternative to the products prepared from virulent RVFV isolates for protection of ruminants against the disease. The vaccine can further be evaluated for safety in pregnant ewes.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Doenças dos Ovinos , Vacinas Virais , Animais , Feminino , Hidróxido de Alumínio , Anticorpos Neutralizantes , Anticorpos Antivirais , Febre do Vale de Rift/prevenção & controle , Ruminantes , Ovinos , Doenças dos Ovinos/prevenção & controle , Vacinas Atenuadas , Vacinas de Produtos Inativados/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...