Rift Valley Fever

裂谷热
  • 文章类型: Journal Article
    裂谷热病毒(RVFV)可能导致以发烧为特征的紧急疾病,肌肉疼痛,甚至人类或反刍动物的死亡。然而,目前还没有批准的预防或治疗RVFV感染的抗病毒药物.虽然治疗性抗体在几项研究中显示出了有希望的预防或治疗潜力,许多研究正在进行中,尤其是在传染病领域。在这些研究中,mRNA-LNP平台显示出巨大的应用潜力,在COVID-19大流行之后。以前,我们获得了抗RVFV的中和抗体,它被命名为A38蛋白,并被证实具有很高的结合和中和能力。在这项研究中,我们旨在鉴定有效优化的序列,并在体外表达优先的mRNA编码抗体.值得注意的是,我们有效地表达了mRNA编码的蛋白,并使用mRNA-LNP平台产生了A38-mRNA-LNP.进行体内药动学实验,并分别设置两组mRNA-A38组和A38蛋白组,它们来自mRNA-LNP和质粒DNA表达的蛋白质,分别。肌内注射A38-mRNA-LNPs,A38蛋白通过静脉给药,与注射游离A38蛋白相比,mRNA编码蛋白提供更长的循环半衰期,证明了它们通过mRNA编码蛋白维持持久蛋白浓度的独特能力。这些关于mRNA编码抗体的临床前数据强调了其在未来预防传染病的潜力。
    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.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:裂谷热(RVF)被世界卫生组织列为优先疾病之一。本研究旨在描述RVF病毒的全球分布,并了解其演变的动态变化,患病率,在打破地理障碍的过程中爆发。
    方法:采用随机效应模型,进行系统文献综述和meta分析以估计宿主的RVF患病率。使用NCBI数据库中的核苷酸序列进行基于分子钟的系统发育分析以估计RVF病毒核苷酸取代率。RVF病毒流行率,核苷酸取代率,并对两次打破地理障碍之前和之后的疫情进行了比较,分别。
    结果:从1930年到2022年,在48个国家的26种宿主中报告了RVF病毒。由于RVF打破了地理障碍,(1)核苷酸替代率在2000年首次传播到非洲之后显着增加;(2)人类的患病率从1.92%(95%CI:0.86-3.25%)显着增加到3.03%(95%CI:2.09-4.12%)在1977年打破了撒哈拉沙漠的地理障碍之后,在2000年之后达到5.24%(95%CI:3.81-6.82%),(3)在人类中蝙蝠和人类之间可能存在RVF病毒溢出,并加速人类的病毒替代率。在疫情爆发期间,在人类中,RVF病毒替代率加速。75.00%的裂谷热爆发发生在洪水和(或)强降雨后0-2个月。
    结论:RVF引起大流行的风险增加,需要在“一个健康”上进行全球合作,以防止潜在的流行病。
    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.
    CONCLUSIONS: RVF has the increasing risk to cause pandemics, and global collaboration on \"One Health\" is needed to prevent potential pandemics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    裂谷热病毒(RVFV)是一种蚊子传播的布尼亚病毒,可引起人类严重和潜在致命的出血热。自噬是一种自我降解过程,可以在多个感染步骤限制病毒复制。在这项研究中,我们评估了RVFV触发的自噬对病毒复制和免疫反应的影响.我们的结果表明,RVFV感染可触发自噬体形成并诱导完全自噬。通过消耗自噬相关基因5(ATG5)来损害自噬通量,ATG7或螯合体1(SQSTM1)或自噬抑制剂的治疗显着降低了病毒RNA合成和子代病毒的产生。机械上,我们的研究结果表明,RVFV核蛋白(NP)C端结构域与自噬受体SQSTM1相互作用,并促进SQSTM1-微管相关蛋白1轻链3B(LC3B)相互作用和自噬.NPC末端结构域的缺失损害了NP与SQSTM1之间的相互作用及其触发自噬的能力。值得注意的是,RVFV触发的自噬促进巨噬细胞的病毒感染,但不在其他测试细胞类型,包括Huh7肝细胞和人脐静脉内皮细胞,提示这种机制的细胞类型特异性。进一步揭示了RVFVNP触发的自噬抑制感染巨噬细胞中的抗病毒先天性免疫应答以促进病毒复制。这些结果提供了对RVFV触发的自噬机制的新见解,并表明靶向自噬途径以开发针对RVFV的抗病毒药物的潜力。重要性我们显示RVFV感染诱导完整的自噬过程。核心自噬基因ATG5、ATG7或SQSTM1的耗竭或巨噬细胞中自噬的药理学抑制强烈抑制RVFV复制。我们进一步揭示了RVFVNPC末端结构域与SQSTM1相互作用并增强SQSTM1/LC3B相互作用以促进自噬。RVFVNP触发的自噬强烈抑制了病毒诱导的干扰素刺激基因在感染的巨噬细胞中的表达,但在其他测试的细胞类型中没有。我们的研究为RVFV触发的自噬机制提供了新的见解,并强调了靶向自噬通量以开发针对该病毒的抗病毒药物的潜力。
    Rift Valley fever virus (RVFV) is a mosquito-borne bunyavirus that causes severe and potentially fatal hemorrhagic fever in humans. Autophagy is a self-degradative process that can restrict viral replication at multiple infection steps. In this study, we evaluated the effects of RVFV-triggered autophagy on viral replication and immune responses. Our results showed that RVFV infection triggered autophagosome formation and induced complete autophagy. Impairing autophagy flux by depleting autophagy-related gene 5 (ATG5), ATG7, or sequestosome 1 (SQSTM1) or treatment with autophagy inhibitors markedly reduced viral RNA synthesis and progeny virus production. Mechanistically, our findings demonstrated that the RVFV nucleoprotein (NP) C-terminal domain interacts with the autophagy receptor SQSTM1 and promotes the SQSTM1-microtubule-associated protein 1 light chain 3 B (LC3B) interaction and autophagy. Deletion of the NP C-terminal domain impaired the interaction between NP and SQSTM1 and its ability to trigger autophagy. Notably, RVFV-triggered autophagy promoted viral infection in macrophages but not in other tested cell types, including Huh7 hepatocytes and human umbilical vein endothelial cells, suggesting cell type specificity of this mechanism. It was further revealed that RVFV NP-triggered autophagy dampens antiviral innate immune responses in infected macrophages to promote viral replication. These results provide novel insights into the mechanisms of RVFV-triggered autophagy and indicate the potential of targeting the autophagy pathway to develop antivirals against RVFV. IMPORTANCE We showed that RVFV infection induced the complete autophagy process. Depletion of the core autophagy genes ATG5, ATG7, or SQSTM1 or pharmacologic inhibition of autophagy in macrophages strongly suppressed RVFV replication. We further revealed that the RVFV NP C-terminal domain interacted with SQSTM1 and enhanced the SQSTM1/LC3B interaction to promote autophagy. RVFV NP-triggered autophagy strongly inhibited virus-induced expression of interferon-stimulated genes in infected macrophages but not in other tested cell types. Our study provides novel insights into the mechanisms of RVFV-triggered autophagy and highlights the potential of targeting autophagy flux to develop antivirals against this virus.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    裂谷热病毒(RVFV)是尼伯病毒属的成员,Phenuiviridae家族的20个属之一。RVFV在动物和人类中引起疾病,并通过沙蝇或蜱传播。然而,对RVFV的研究受到生物安全3级(BSL-3)遏制要求的限制。假型病毒克服了这种限制,因为它可以在BSL-2环境中处理。假型RVFV具有与正宗病毒相同的包膜蛋白结构,模拟受体结合和膜融合到宿主细胞的相同过程。因此,假型的静脉病毒是研究这些病毒的感染机制以及筛选抑制药物和开发治疗性单克隆抗体的有用工具。
    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.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    裂谷热(RVF)是由裂谷热病毒(RVFV)引起的人畜共患疾病,Bunyavirales的Phenuiviridae家族中一种新兴的虫媒病毒,有可能在人类和牲畜中引起严重疾病。它增加了反刍动物流产或胎儿畸形的发生率,并导致人类出现脑炎或出血热等临床表现。一旦病毒入侵,细胞或生物体的先天免疫系统被激活以产生干扰素(IFN)并阻止病毒增殖。同时,RVFV启动对策,以限制转录和蛋白质水平的抗病毒反应。RVFV非结构蛋白(NSs)是关键的毒力因子,不仅进行免疫逃避,而且影响细胞复制周期并具有细胞病变效应。在这次审查中,我们总结了宿主细胞根据IFN信号转导途径采用的先天免疫,以及RVFV主要具有NSs蛋白抑制活性的免疫逃避机制。阐明宿主先天免疫和RVFV免疫逃避之间的军备竞赛为药物靶标筛选提供了新的途径,并为当前和未来的流行病提供了可能的解决方案。
    Rift Valley fever (RVF) is a zoonotic disease caused by Rift Valley fever virus (RVFV), an emerging arbovirus within the Phenuiviridae family of Bunyavirales that has potential to cause severe diseases in both humans and livestock. It increases the incidence of abortion or foetal malformation in ruminants and leads to clinical manifestations like encephalitis or haemorrhagic fever in humans. Upon virus invasion, the innate immune system from the cell or the organism is activated to produce interferon (IFN) and prevent virus proliferation. Meanwhile, RVFV initiates countermeasures to limit antiviral responses at transcriptional and protein levels. RVFV nonstructural proteins (NSs) are the key virulent factors that not only perform immune evasion but also impact the cell replication cycle and has cytopathic effects. In this review, we summarize the innate immunity host cells employ depending on IFN signal transduction pathways, as well as the immune evasion mechanisms developed by RVFV primarily with the inhibitory activity of NSs protein. Clarifying the arms race between host innate immunity and RVFV immune evasion provides new avenues for drug target screening and offers possible solutions to current and future epidemics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    裂谷热病毒(RVFV)是引起动物和人类严重疾病的最重要的毒力病原体之一。然而,目前没有批准的疫苗来预防人的RVFV感染。尚未报道人腺病毒血清型4(Ad4)作为RVFV疫苗的载体的用途。这里,我们报道了表达密码子优化形式的RVFV糖蛋白Gn和Gc(称为Ad4-GnGc)的可复制重组Ad4载体的产生。用Ad4-GnGc进行的肌内免疫在小鼠中引起针对RVFV和细胞免疫应答的强效中和抗体。单次低剂量接种Ad4-GnGc完全保护干扰素-α/β受体缺陷型A129小鼠免受致死性RVFV感染。更重要的是,Ad4-GnGc功效不受预先存在的对腺病毒血清型5的免疫的影响,其目前广泛存在于人群中。这些结果表明Ad4-GnGc是针对RVFV的有希望的疫苗候选物。
    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/β 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.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    裂谷热病毒(RVFV)是一种人畜共患的虫媒病毒,对人类和动物都有致病性。目前,没有已证明有效的RVFV药物或许可的疫苗可用于人类或动物。因此,迫切需要开发有效的治疗方案来控制这种病毒感染。RVFV糖蛋白N(GN),糖蛋白C(GC),和核衣壳(N)蛋白是有吸引力的抗病毒药物靶标,因为它们在RVFV复制中具有关键作用。在目前的研究中,对6000多种具有已知抗病毒活性的植物化学物质进行了基于对接的虚拟筛选这些保守的RVFV蛋白.前五名热门化合物,calyxinC,calyxinD,calyxinJ,gericudraninsA,和眼脂溶液素C表现出对所有三种靶蛋白的最佳结合。此外,来自分子动力学(MD)模拟和MM/GBSA分析的多个参数证实了蛋白质-配体复合物的稳定性,并揭示了这些化合物可能充当RVFV复制的潜在泛抑制剂。我们的计算分析可能有助于开发有希望的针对RVFV感染的有效药物。
    The Rift Valley fever virus (RVFV) is a zoonotic arbovirus and pathogenic to both humans and animals. Currently, no proven effective RVFV drugs or licensed vaccine are available for human or animal use. Hence, there is an urgent need to develop effective treatment options to control this viral infection. RVFV glycoprotein N (GN), glycoprotein C (GC), and nucleocapsid (N) proteins are attractive antiviral drug targets due to their critical roles in RVFV replication. In present study, an integrated docking-based virtual screening of more than 6000 phytochemicals with known antiviral activities against these conserved RVFV proteins was conducted. The top five hit compounds, calyxin C, calyxin D, calyxin J, gericudranins A, and blepharocalyxin C displayed optimal binding against all three target proteins. Moreover, multiple parameters from the molecular dynamics (MD) simulations and MM/GBSA analysis confirmed the stability of protein-ligand complexes and revealed that these compounds may act as potential pan-inhibitors of RVFV replication. Our computational analyses may contribute toward the development of promising effective drugs against RVFV infection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Rift Valley fever (RVF) is a vector-borne viral disease of major animal and public health importance. In 2018-19, it caused an epidemic in both livestock and human populations of the island of Mayotte. Using Bayesian modelling approaches, we assessed the spatio-temporal pattern of RVF virus (RVFV) infection in livestock and human populations across the island, and factors shaping it. First, we assessed if (i) livestock movements, (ii) spatial proximity from communes with infected animals, and (iii) livestock density were associated with the temporal sequence of RVFV introduction into Mayotte communes\' livestock populations. Second, we assessed whether the rate of human infection was associated with (a) spatial proximity from and (b) livestock density of communes with infected animals. Our analyses showed that the temporal sequence of RVFV introduction into communes\' livestock populations was associated with livestock movements and spatial proximity from communes with infected animals, with livestock movements being associated with the best model fit. Moreover, the pattern of human cases was associated with their spatial proximity from communes with infected animals, with the risk of human infection sharply increasing if livestock in the same or close communes were infected. This study highlights the importance of understanding livestock movement networks in informing the design of risk-based RVF surveillance programs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    Rift Valley fever (RVF) is a mosquito-borne zoonotic disease. Since its first outbreak in 1930, RVF epidemics have caused huge economic losses and public health impacts in Africa. In 2000, RVF became a disease of global concern as it spread to the Arabian Peninsula. In our study, a Geographic Information System-based risk assessment for the occurrence of Rift Valley fever in China was established by means of ecological niche modelling. Based on occurrence records (RVF records from FAO EMPRES-i, vector records from literatures and GBIF) and high-resolution environmental layers, the prediction maps of RVF occurrence probability and distribution of five potential RVF vectors in China were modelled using Maxent. An internal validation was adopted for model verification, and high AUC values were obtained (0.918 for RVF and 0.837-0.992 for vectors). By overlaying the RVF prediction map with the combined RVF vector prediction map using Fuzzy overlay tool (\'AND\' operator) of ArcMap 10.2, we got the first risk map of possible RVF vector transmission. This map was further overlaid with the latest livestock distribution map (\'AND\' operator) to generate the second risk map of possible RVF threat to domestic livestock. The south-west border provinces in China, Yunnan, Guangxi and Tibet were predicted to have a high possibility of RVF occurrence. Conditions conducive to the local amplification of RVF also exist in these areas. Temperature seasonality, mean temperature of dry season and precipitation of the driest month were considered as key environmental variables for RVF, and common environmental conditions were found conductive for vectors. It is suggested to establish proper surveillance systems in south-west border areas to minimize the possibility of RVF invasion. Our findings can serve as a valuable reference for prevention measures to be implemented.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    The Rift Valley fever virus (RVFV) is an arthropod-borne virus that can not only cause severe disease in domestic animals but also in humans. However, the licensed vaccines or available therapeutics for humans do not exist. Here, we report two Gn-specific neutralizing antibodies (NAbs), isolated from a rhesus monkey immunized with recombinant human adenoviruses type 4 expressing Rift Valley fever virus Gn and Gc protein (rHAdV4-GnGcopt). The two NAbs were both able to protect host cells from RVFV infection. The interactions between NAbs and Gn were then characterized to demonstrate that these two NAbs might preclude RVFV glycoprotein rearrangement, hindering the exposure of fusion loops in Gc to endosomal membranes after the virus invades the host cell. The target region for the two NAbs is located in the Gn domain III, implying that Gn is a desired target for developing vaccines and neutralizing antibodies against RVFV.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号