Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 250
Filtrar
1.
Methods Mol Biol ; 2824: 81-89, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39039407

RESUMO

The Rift Valley fever virus (RVFV) is an arthropod-borne, zoonotic, hemorrhagic fever virus that can cause severe diseases both in livestock and humans. The spread of RVFV in areas previously considered as non-endemic together with the absence of licensed vaccines for use in humans and animals poses a major health and economic threat worldwide. It is therefore crucial to make major progresses in our understanding and management of this virus and its zoonosis. RVFV is considered a bioterrorism pathogen, and, thus, only a few institutes, facilities, and personnel are legally authorized to detain it and handle it. Moreover, this virus must be manipulated in a biosafety level 3 (BSL3) laboratory following strict biosafety protocols to ensure that biosecurity's highest standards are met. Only certain attenuated strains such as the MP12 strain can be handled in BSL2 laboratories, depending on the country considered. To assist researchers in working with RVFV in the safest possible conditions, this chapter presents validated methods for effective RVFV decontamination and inactivation.


Assuntos
Descontaminação , Vírus da Febre do Vale do Rift , Inativação de Vírus , Animais , Descontaminação/métodos , Humanos , Febre do Vale de Rift/prevenção & controle , Febre do Vale de Rift/transmissão , Febre do Vale de Rift/virologia , Contenção de Riscos Biológicos/métodos , Células Vero , Chlorocebus aethiops
2.
Methods Mol Biol ; 2824: 385-395, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39039425

RESUMO

Rift Valley fever (RVF) caused by Rift Valley fever virus (RVFV) is a major health concern for both domesticated animals and humans in certain endemic areas of Africa. With changing environmental conditions and identification of vectors capable of transmitting the virus, there is high risk of RVFV spreading into other parts of the world. Furthermore, unavailability of effective vaccines in the event of an outbreak can be a major challenge as witnessed recently in case of SARS-CoV2 pandemic. Hence, identifying potential vaccines and testing their protective efficacy in preclinical models before clinical testing is the absolute need of the hour. Here, we describe methods used to quantify virus-specific T cell responses in mice that were immunized with RVFV strains or antigens.


Assuntos
Vírus da Febre do Vale do Rift , Linfócitos T , Vacinas Virais , Animais , Camundongos , Linfócitos T/imunologia , Vírus da Febre do Vale do Rift/imunologia , Vacinas Virais/imunologia , Febre do Vale de Rift/imunologia , Febre do Vale de Rift/prevenção & controle , Imunização/métodos , Vacinação/métodos , Antígenos Virais/imunologia
3.
J Infect Dev Ctries ; 18(7): 1090-1099, 2024 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-39078795

RESUMO

INTRODUCTION: This immunoinformatic study identified potential epitopes from the envelopment polyprotein (Gn/Gc) of Rift Valley fever virus (RVFV), a pathogenic virus causing severe fever in humans and livestock. Effective vaccination is crucial for controlling RVFV outbreaks. The identification of suitable epitopes is crucial for the development of safe and effective vaccines. METHODOLOGY: Protein sequences were obtained from the UniProt database, and evaluated through VaxiJen v2.0 to predict the B and T-cell epitopes within the RVFV glycoprotein. Gn/Gc protein sequences were analyzed with bioinformatics tools and algorithms. The predicted T-cell and B-cell epitopes were evaluated for antigenicity, allergenicity, and toxicity by the VaxiJen v2.0 system, AllerTop v2.0, and ToxinPred server, respectively. RESULTS: We employed computational methods to screen the RVFV envelopment polyprotein encompassing N-terminal and C-terminal glycoprotein segments, to discover antigenic T- and B-cell epitopes. Our analysis unveiled multiple potential epitopes within the RVFV glycoprotein, specifically within the Gn/Gc protein sequences. Subsequently, we selected eleven cytotoxic T-lymphocytes (CTL) and four helper T-lymphocytes (HTL) for population coverage analysis, which collectively extended to cover 97.04% of the world's population, representing diverse ethnicities and regions. Notably, the CTL epitope VQADLTLMF exhibited binding affinity to numerous human leukocyte antigen (HLA) alleles. The identification of glycoprotein (Gn/Gc) epitopes through this immunoinformatic study bears significant implications for advancing the development of an effective RVFV vaccine. CONCLUSIONS: These findings provide valuable insights into the immunological aspects of the disease and may contribute towards the development of broad-spectrum antiviral therapies targeting other RNA viruses with similar polymerase enzymes.


Assuntos
Biologia Computacional , Epitopos de Linfócito B , Epitopos de Linfócito T , Vírus da Febre do Vale do Rift , Vírus da Febre do Vale do Rift/imunologia , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito B/imunologia , Humanos , Vacinas Virais/imunologia , Febre do Vale de Rift/prevenção & controle , Febre do Vale de Rift/imunologia , Animais
4.
Viruses ; 16(7)2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-39066162

RESUMO

Rift Valley fever (RVF) is a mosquito-borne zoonotic viral disease endemic to Africa and the Middle East. Live-attenuated RVF vaccines have been studied for both veterinary and human use due to their strong immunogenicity and cost-effective manufacturing. The live-attenuated MP-12 vaccine has been conditionally approved for veterinary use in the U.S.A., and next-generation live-attenuated RVF vaccine candidates are being actively researched. Assessing the virulence phenotype of vaccine seeds or lots is crucial for managing vaccine safety. Previously, preweaning 19-day-old outbred CD1 mice have been used to evaluate the MP-12 strain. This study aimed to characterize the relative virulence of three live-attenuated RVF vaccine strains in 19-day-old inbred C57BL/6 mice: the recombinant MP-12 (rMP-12), the RVax-1, and the ∆NSs-∆NSm-rZH501 strains. Although this mouse model did not show dose-dependent pathogenesis, mice that succumbed to the infection exhibited distinct brain pathology. Mice infected with ∆NSs-∆NSm-rZH501 showed an infiltration of inflammatory cells associated with infected neurons, and focal lesions formed around virus-infected cells. In contrast, mice infected with rMP-12 or RVax-1 showed a minimal association of inflammatory cells in the brain, yet the virus spread diffusely. The preweaning model is likely useful for evaluating host responses to attenuated RVFV strains, although further refinement may be necessary to quantitate the virulence among different RVFV strains or vaccine lots.


Assuntos
Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Vacinas Atenuadas , Vacinas Virais , Animais , Vírus da Febre do Vale do Rift/patogenicidade , Vírus da Febre do Vale do Rift/imunologia , Vírus da Febre do Vale do Rift/genética , Febre do Vale de Rift/virologia , Febre do Vale de Rift/patologia , Febre do Vale de Rift/prevenção & controle , Febre do Vale de Rift/imunologia , Camundongos , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Virulência , Feminino
5.
Emerg Microbes Infect ; 13(1): 2373313, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38946528

RESUMO

Rift Valley fever (RVF) is a mosquito-borne zoonotic disease caused by RVF virus (RVFV). RVFV infections in humans are usually asymptomatic or associated with mild febrile illness, although more severe cases of haemorrhagic disease and encephalitis with high mortality also occur. Currently, there are no licensed human vaccines available. The safety and efficacy of a genetically engineered four-segmented RVFV variant (hRVFV-4s) as a potential live-attenuated human vaccine has been tested successfully in mice, ruminants, and marmosets though the correlates of protection of this vaccine are still largely unknown. In the present study, we have assessed hRVFV-4s-induced humoral and cellular immunity in a mouse model of RVFV infection. Our results confirm that a single dose of hRVFV-4s is highly efficient in protecting naïve mice from developing severe disease following intraperitoneal challenge with a highly virulent RVFV strain and data show that virus neutralizing (VN) serum antibody titres in a prime-boost regimen are significantly higher compared to the single dose. Subsequently, VN antibodies from prime-boost-vaccinated recipients were shown to be protective when transferred to naïve mice. In addition, hRVFV-4s vaccination induced a significant virus-specific T cell response as shown by IFN-γ ELISpot assay, though these T cells did not provide significant protection upon passive transfer to naïve recipient mice. Collectively, this study highlights hRVFV-4s-induced VN antibodies as a major correlate of protection against lethal RVFV infection.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Vacinas Atenuadas , Vacinas Virais , Animais , Vírus da Febre do Vale do Rift/imunologia , Vírus da Febre do Vale do Rift/genética , Febre do Vale de Rift/prevenção & controle , Febre do Vale de Rift/imunologia , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Camundongos , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Feminino , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/administração & dosagem , Modelos Animais de Doenças , Imunidade Celular , Linfócitos T/imunologia , Imunidade Humoral , Camundongos Endogâmicos BALB C , Interferon gama/imunologia , Vacinação
6.
PLoS Pathog ; 20(6): e1012343, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38935789

RESUMO

Rift Valley fever virus (RVFV) is an encephalitic bunyavirus that can infect neurons in the brain. There are no approved therapeutics that can protect from RVFV encephalitis. Innate immunity, the first line of defense against infection, canonically antagonizes viruses through interferon signaling. We found that interferons did not efficiently protect primary cortical neurons from RVFV, unlike other cell types. To identify alternative neuronal antiviral pathways, we screened innate immune ligands and discovered that the TLR2 ligand Pam3CSK4 inhibited RVFV infection, and other bunyaviruses. Mechanistically, we found that Pam3CSK4 blocks viral fusion, independent of TLR2. In a mouse model of RVFV encephalitis, Pam3CSK4 treatment protected animals from infection and mortality. Overall, Pam3CSK4 is a bunyavirus fusion inhibitor active in primary neurons and the brain, representing a new approach toward the development of treatments for encephalitic bunyavirus infections.


Assuntos
Lipopeptídeos , Neurônios , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Vírus da Febre do Vale do Rift/efeitos dos fármacos , Camundongos , Lipopeptídeos/farmacologia , Febre do Vale de Rift/virologia , Febre do Vale de Rift/prevenção & controle , Neurônios/metabolismo , Neurônios/virologia , Camundongos Endogâmicos C57BL , Humanos , Imunidade Inata/efeitos dos fármacos , Encefalite Viral/virologia , Encefalite Viral/imunologia , Encefalite Viral/prevenção & controle , Encefalite Viral/tratamento farmacológico , Antivirais/farmacologia
7.
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
8.
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
9.
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
11.
J Med Entomol ; 60(6): 1197-1213, 2023 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-37862067

RESUMO

Rift Valley fever virus (RVFV) (Bunyavirales: Phlebovirus) is a prominent vector-borne zoonotic disease threat to global agriculture and public health. Risks of introduction into nonendemic regions are tied to changing climate regimes and other dynamic environmental factors that are becoming more prevalent, as well as virus evolutionary factors and human/animal movement. Endemic to the African continent, RVFV has caused large epizootics at the decadal scale since the early 20th century but has spread to the Arabian Peninsula and shows increasing patterns of interepizootic transmission on the annual scale. This virus can be transmitted by mosquitoes as well as through direct contact with infected tissues and can cause sporadic to widespread morbidity and mortality in domestic ungulate livestock as well as humans. High viremias in infected livestock moved for legal and illegal trade as well as in infected mosquitoes or human travelers can spread this virus worldwide. With increasing global commerce, it is likely RVFV will be introduced to new areas with suitable hosts, mosquito vector species, and environments. However, the strong mosquito component of RVFV epidemiology combined with advancements in vaccines, diagnostics, and virus evolutionary factors create opportunities for strategies to leverage models of connectivity among potential source and emerging regions to target surveillance and mitigation activities to reduce the risk of RVFV introduction, or contain the virus should it be introduced, into new regions.


Assuntos
Culicidae , Phlebovirus , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Humanos , Febre do Vale de Rift/epidemiologia , Febre do Vale de Rift/prevenção & controle , Zoonoses/prevenção & controle
12.
PLoS Negl Trop Dis ; 17(8): e0011560, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37611063

RESUMO

Epidemics of Rift Valley fever (RVF), a mosquito-borne zoonotic disease caused by RVF virus, have been linked to exceptionally heavy rainfall and widespread flooding. The disease is endemic in most African countries and pose a major global health risk. Given that the disease was reported in various districts of Tanzania, we hypothesized a lack of knowledge about RVF epidemiology among agropastoral and pastoral communities. The research took place in a period of 7 months, from July, 2021 to January, 2022. The aim of this study was to assess the knowledge, attitudes, and practices (KAP) among the agropastoral and pastoral communities of Ngorongoro district towards RVF. The survey employed a mixed method system, which included 3 focus groups (each comprised 12 individuals), 20 key informant interviews and administration of questionnaire (N = 352) in agropastoral and pastoral community members of Ngorongoro district. The relationship between demographic characteristics and communities' knowledge, attitudes, and practices regarding RVF was observed using a multiple logistic regression model. A total of 352 participants were interviewed, with the majority (67.61%) being male and 32.39% being female, majority (39.5%) attending primary school, and majority (58.2%) being pastoralists. The findings showed that only 36.1%, 38.64% and 16.19% of participants had good knowledge, positive attitude and good practices regarding RVF respectively. Significant demographic factors related with knowledge included: gender (OR = 1.9, CI = 1.03-3.56, P = 0.041), education levels (primary: OR = 3.97, CI = 2-8.16, P = 0.000; secondary: OR = 15.27, CI = 5.5-46.23, P = 0.000 and college: OR = 34. 23, CI = 5.4-67.22, P = 0.000), and locality (Pinyinyi: OR = 0.14, CI = 0.05-0.38, P = 0.000 and Sale: OR = 0.14, CI = 0.04-0.44, P = 0.001). Male participants showed significant positive attitude towards RVF compared to female (OR = 2.37, CI = 1.35-4.17, P = 0.003). Individuals with formal education showed a significant positive attitude toward RVF compared to informal (OR>1, P<0.05). Agropastoral members showed a significant negative attitude toward RVF compared to pastoralists (OR = 0.51, CI = 0.26-0.99, P = 0.048). The calculated RVF prevention practices values were insignificantly (P = 0.853) correlated with knowledge values. The significant correlation between knowledge and attitude, as well as attitude and practice were found (P<0.05). In general, the study revealed poor knowledge, negative attitude and poor practices of communities towards RVF. The lack of regular education programs to make the communities aware of the disease was implicated for these findings. This recommends that provision of health education should be a long-term practice among agropastoral and pastoral communities in order to prevent further RVF outbreaks in Tanzania.


Assuntos
Ecossistema , Febre do Vale de Rift , Animais , Feminino , Masculino , Humanos , Febre do Vale de Rift/epidemiologia , Febre do Vale de Rift/prevenção & controle , Tanzânia/epidemiologia , Conhecimentos, Atitudes e Prática em Saúde , Meio Ambiente
14.
Lancet Infect Dis ; 23(8): 956-964, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37060917

RESUMO

BACKGROUND: Rift Valley fever is a viral epidemic illness prevalent in Africa that can be fatal or result in debilitating sequelae in humans. No vaccines are available for human use. We aimed to evaluate the safety and immunogenicity of a non-replicating simian adenovirus-vectored Rift Valley fever (ChAdOx1 RVF) vaccine in humans. METHODS: We conducted a phase 1, first-in-human, open-label, dose-escalation trial in healthy adults aged 18-50 years at the Centre for Clinical Vaccinology and Tropical Medicine, Oxford, UK. Participants were required to have no serious comorbidities or previous history of receiving an adenovirus-based vaccine before enrolment. Participants were non-randomly allocated to receive a single ChAdOx1 RVF dose of either 5 × 109 virus particles (vp), 2·5 × 1010 vp, or 5 × 1010 vp administered intramuscularly into the deltoid of their non-dominant arm; enrolment was sequential and administration was staggered to allow for safety to be assessed before progression to the next dose. Primary outcome measures were assessment of adverse events and secondary outcome measures were Rift Valley fever neutralising antibody titres, Rift Valley fever GnGc-binding antibody titres (ELISA), and cellular response (ELISpot), analysed in all participants who received a vaccine. This trial is registered with ClinicalTrials.gov (NCT04754776). FINDINGS: Between June 11, 2021, and Jan 13, 2022, 15 volunteers received a single dose of either 5 × 109 vp (n=3), 2·5 × 1010 vp (n=6), or 5 × 1010 vp (n=6) ChAdOx1 RVF. Nine participants were female and six were male. 14 (93%) of 15 participants reported solicited local adverse reactions; injection-site pain was the most frequent (13 [87%] of 15). Ten (67%) of 15 participants (from the 2·5 × 1010 vp and 5 × 1010 vp groups only) reported systemic symptoms, which were mostly mild in intensity, the most common being headache (nine [60%] of 15) and fatigue (seven [47%]). All unsolicited adverse events reported within 28 days were either mild or moderate in severity; gastrointestinal symptoms were the most common reaction (at least possibly related to vaccination), occurring in four (27%) of 15 participants. Transient decreases in total white cell, lymphocyte, or neutrophil counts occurred at day 2 in some participants in the intermediate-dose and high-dose groups. Lymphopenia graded as severe occurred in two participants in the 5 × 1010 vp group at a single timepoint, but resolved at the subsequent follow-up visit. No serious adverse events occurred. Rift Valley fever neutralising antibodies were detectable across all dose groups, with all participants in the 5 × 1010 vp dose group having high neutralising antibody titres that peaked at day 28 after vaccination and persisted through the 3-month follow-up. High titres of binding IgG targeting Gc glycoprotein were detected whereas those targeting Gn were comparatively low. IFNγ cellular responses against Rift Valley fever Gn and Gc glycoproteins were observed in all participants except one in the 5 × 1010 vp dose group. These IFNγ responses peaked at 2 weeks after vaccination, were highest in the 5 × 1010 vp dose group, and tended to be more frequent against the Gn glycoprotein. INTERPRETATION: ChAdOx1 RVF was safe, well tolerated, and immunogenic when administered as a single dose in this study population. The data support further clinical development of ChAdOx1 RVF for human use. FUNDING: UK Department of Health and Social Care through the UK Vaccines Network, Oak Foundation, and the Wellcome Trust. TRANSLATION: For the Swahili translation of the abstract see Supplementary Materials section.


Assuntos
Febre do Vale de Rift , Vacinas Virais , Humanos , Adulto , Masculino , Feminino , Animais , Febre do Vale de Rift/prevenção & controle , Anticorpos Neutralizantes , Glicoproteínas , Reino Unido , Imunogenicidade da Vacina , Anticorpos Antivirais , Método Duplo-Cego
15.
Adv Exp Med Biol ; 1407: 253-264, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36920701

RESUMO

Rift Valley fever virus (RVFV) is a member of the Phlebovirus genus, one of the 20 genera in the Phenuiviridae family. RVFV causes disease in animals and humans and is transmitted by sandflies or ticks. However, research into RVFV is limited by the requirement for biosafety level 3 (BSL-3) containment. Pseudotyped virus overcomes this limitation as it can be handled in a BSL-2 environment. Pseudotyped RVFV possesses an identical envelope protein structure to that of the authentic virus, simulating the same process of receptor binding and membrane fusion to host cells. Pseudotyped phleboviruses are therefore useful tools to study the infection mechanism of these viruses and for the screening of inhibitory drugs and the development of therapeutic monoclonal antibodies.


Assuntos
Phlebovirus , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Humanos , Phlebovirus/genética , Febre do Vale de Rift/prevenção & controle , Pseudotipagem Viral , Vírus da Febre do Vale do Rift/genética
18.
PLoS Negl Trop Dis ; 17(1): e0010460, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36634153

RESUMO

Rift Valley fever virus (RVFV) is a zoonotic arbovirus that has profound impact on domestic ruminants and can also be transmitted to humans via infected animal secretions. Urban areas in endemic regions across Africa have susceptible animal and human hosts, dense vector distributions, and source livestock (often from high risk locations to meet the demand for animal protein). Yet, there has never been a documented urban outbreak of RVF. To understand the likely risk of RVFV introduction to urban communities from their perspective and guide future initiatives, we conducted focus group discussions with slaughterhouse workers, slaughterhouse animal product traders, and livestock owners in Kisumu City and Ukunda Town in Kenya. For added perspective and data triangulation, in-depth interviews were conducted one-on-one with meat inspector veterinarians from selected slaughterhouses. A theoretical framework relevant to introduction, transmission, and potential persistence of RVF in urban areas is presented here. Urban livestock were primarily mentioned as business opportunities, but also had personal sentiment. In addition to slaughtering risks, perceived risk factors included consumption of fresh milk. High risk groups' knowledge and experience with RVFV and other zoonotic diseases impacted their consideration of personal risk, with consensus towards lower risk in the urban setting compared to rural areas as determination of health risk was said to primarily rely on hygiene practices rather than the slaughtering process. Groups relied heavily on veterinarians to confirm animal health and meat safety, yet veterinarians reported difficulty in accessing RVFV diagnostics. We also identified vulnerable public health regulations including corruption in meat certification outside of the slaughterhouse system, and blood collected during slaughter being used for food and medicine, which could provide a means for direct RVFV community transmission. These factors, when compounded by diverse urban vector breeding habitats and dense human and animal populations, could create suitable conditions for RVFV to arrive an urban center via a viremic imported animal, transmit to locally owned animals and humans, and potentially adapt to secondary vectors and persist in the urban setting. This explorative qualitative study proposes risk pathways and provides initial insight towards determining how urban areas could adapt control measures and plan future initiatives to better understand urban RVF potential.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Animais , Humanos , Quênia/epidemiologia , Gado/virologia , Carne , Febre do Vale de Rift/prevenção & controle , Febre do Vale de Rift/transmissão , Ruminantes/virologia , Zoonoses/prevenção & controle , Zoonoses/transmissão , Fatores de Risco , População Urbana , Matadouros/legislação & jurisprudência , Matadouros/normas , Inocuidade dos Alimentos
19.
Viral Immunol ; 36(1): 33-40, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36399689

RESUMO

Rift Valley fever virus (RVFV) is an emerging arbovirus that affects both ruminants and humans. RVFV causes severe and recurrent outbreaks in Africa and the Arabian Peninsula with a significant risk for emergence into new locations. Although there are a variety of RVFV veterinary vaccines for use in endemic areas, there is currently no licensed vaccine for human use; therefore, there is a need to develop and assess new vaccines. Herein, we report a live-attenuated recombinant vaccine candidate for RVFV, based on the previously described genomic reconfiguration of the conditionally licensed MP12 vaccine. There are two general strategies used to develop live-attenuated RVFV vaccines, one being serial passage of wild-type RVFV strains to select attenuated mutants such as Smithburn, Clone 13, and MP12 vaccine strains. The second strategy has utilized reverse genetics to attenuate RVFV strains by introducing deletions or insertions within the viral genome. The novel candidate vaccine characterized in this report contains a two-segmented genome that lacks the medium viral segment (M) and two virulence genes (nonstructural small and nonstructural medium). The vaccine candidate, named r2segMP12, was evaluated for the production of neutralizing antibodies to RVFV in outbred CD-1 mice. The immune response induced by the r2segMP12 vaccine candidate was directly compared to the immune response induced by the rMP12 parental strain vaccine. Our study demonstrated that a single immunization with the r2segMP12 vaccine candidate at 105 plaque-forming units elicited a higher neutralizing antibody response than the rMP12 vaccine at the same vaccination titer without the need for a booster.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Vacinas Virais , Humanos , Animais , Camundongos , Vírus da Febre do Vale do Rift/genética , Febre do Vale de Rift/prevenção & controle , Febre do Vale de Rift/epidemiologia , Vacinas Atenuadas/genética , Vacinas Virais/genética , Anticorpos Neutralizantes
20.
Front Immunol ; 13: 907675, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36439179

RESUMO

Rift Valley fever virus (RVFV) is one of the most important virulent pathogens causing severe disease in animals and humans. However, there is currently no approved vaccine to prevent RVFV infection in humans. The use of human adenovirus serotype 4 (Ad4) as a vector for an RVFV vaccine has not been reported. Here, we report the generation of a replication-competent recombinant Ad4 vector expressing codon-optimized forms of the RVFV glycoproteins Gn and Gc (named Ad4-GnGc). Intramuscular immunization with Ad4-GnGc elicited robust neutralizing antibodies against RVFV and cellular immune responses in mice. A single low-dose vaccination with Ad4-GnGc completely protected interferon-α/ß receptor-deficient A129 mice from lethal RVFV infection. More importantly, Ad4-GnGc efficacy was not affected by pre-existing immunity to adenovirus serotype 5, which currently exists widely in populations. These results suggest that Ad4-GnGc is a promising vaccine candidate against RVFV.


Assuntos
Infecções por Adenoviridae , Vacinas contra Adenovirus , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Vacinas Virais , Camundongos , Humanos , Animais , Vírus da Febre do Vale do Rift/genética , Febre do Vale de Rift/prevenção & controle , Adenoviridae/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA