RVFV

RVFV
  • 文章类型: Journal Article
    类病毒是一类分段的负义单链RNA病毒,通常由编码四种不同蛋白质的三个RNA片段组成。致病性斑马病毒株的出现,例如撒哈拉以南非洲的裂谷热静脉病毒(RVFV),东亚和东南亚的血小板减少综合征病毒(SFTSV)近年来,美国的心脏地带病毒(HRTV)对全球公共卫生提出了相当大的挑战。先天免疫系统作为宿主抵抗病原体入侵的初始防御机制起着至关重要的作用。除了继续进行旨在阐明斑病毒流行病学特征的研究外,在研究其病毒毒力因子(糖蛋白,非结构蛋白,和核蛋白)和潜在的宿主-病原体相互作用。具体来说,努力集中在理解病毒免疫逃避的机制上,病毒组装和出口,以及涉及免疫细胞的宿主免疫网络,程序性细胞死亡,炎症,核酸受体,等。此外,大量的技术进步,包括宏基因组学,代谢组学,单细胞转录组学,蛋白质组学,基因编辑,单克隆抗体,和疫苗,已被用于进一步了解斑状病毒的发病机制和宿主免疫反应。因此,这项审查旨在全面概述当前对东道国认可机制的理解,病毒免疫逃避,以及在人类致病性斑病毒感染期间的潜在治疗方法,特别关注RVFV和SFTSV。
    Phenuiviruses are a class of segmented negative-sense single-stranded RNA viruses, typically consisting of three RNA segments that encode four distinct proteins. The emergence of pathogenic phenuivirus strains, such as Rift Valley fever phlebovirus (RVFV) in sub-Saharan Africa, Severe Fever with Thrombocytopenia Syndrome Virus (SFTSV) in East and Southeast Asia, and Heartland Virus (HRTV) in the United States has presented considerable challenges to global public health in recent years. The innate immune system plays a crucial role as the initial defense mechanism of the host against invading pathogens. In addition to continued research aimed at elucidating the epidemiological characteristics of phenuivirus, significant advancements have been made in investigating its viral virulence factors (glycoprotein, non-structural protein, and nucleoprotein) and potential host-pathogen interactions. Specifically, efforts have focused on understanding mechanisms of viral immune evasion, viral assembly and egress, and host immune networks involving immune cells, programmed cell death, inflammation, nucleic acid receptors, etc. Furthermore, a plethora of technological advancements, including metagenomics, metabolomics, single-cell transcriptomics, proteomics, gene editing, monoclonal antibodies, and vaccines, have been utilized to further our understanding of phenuivirus pathogenesis and host immune responses. Hence, this review aims to provide a comprehensive overview of the current understanding of the mechanisms of host recognition, viral immune evasion, and potential therapeutic approaches during human pathogenic phenuivirus infections focusing particularly on RVFV and SFTSV.
    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(RVF),蚊媒跨界人畜共患病,2012年在卢旺达的牲畜中首次被证实,此后几乎每年都有零星病例报告。2018年,我国经历了第一次大疫情,紧随其后的是2022年的第二次。为了确定循环病毒谱系及其祖先起源,2018年爆发的两个基因组序列,三十六,41,和38个小序列(S),中等(M),和大(L)基因组片段,分别,从2022年爆发产生。2022年爆发的所有样本都是从屠宰场收集的。进行了最大似然和基于贝叶斯的系统发育分析。研究结果表明,RVF病毒属于单一谱系,C,在两次爆发期间循环,并与2016年至2019年在乌干达分离的裂谷热病毒共享一个共同祖先,也与2006/2007年在肯尼亚报告的最大的东非裂谷热疫情有关,坦桑尼亚,索马里。除了野生型病毒,在屠宰场动物中发现了RVFV克隆13疫苗株的遗传证据,证明在肉类相关行业工作的人可能存在职业暴露风险,结果未知。这些结果为RVFV谱系C在非洲的持续广泛传播提供了更多证据,并强调了在应对RVF紧急情况方面需要有效的国家和国际基于OneHealth的合作方法。
    Rift Valley fever (RVF), a mosquito-borne transboundary zoonosis, was first confirmed in Rwanda\'s livestock in 2012 and since then sporadic cases have been reported almost every year. In 2018, the country experienced its first large outbreak, which was followed by a second one in 2022. To determine the circulating virus lineages and their ancestral origin, two genome sequences from the 2018 outbreak, and thirty-six, forty-one, and thirty-eight sequences of small (S), medium (M), and large (L) genome segments, respectively, from the 2022 outbreak were generated. All of the samples from the 2022 outbreak were collected from slaughterhouses. Both maximum likelihood and Bayesian-based phylogenetic analyses were performed. The findings showed that RVF viruses belonging to a single lineage, C, were circulating during the two outbreaks, and shared a recent common ancestor with RVF viruses isolated in Uganda between 2016 and 2019, and were also linked to the 2006/2007 largest East Africa RVF outbreak reported in Kenya, Tanzania, and Somalia. Alongside the wild-type viruses, genetic evidence of the RVFV Clone 13 vaccine strain was found in slaughterhouse animals, demonstrating a possible occupational risk of exposure with unknown outcome for people working in meat-related industry. These results provide additional evidence of the ongoing wide spread of RVFV lineage C in Africa and emphasize the need for an effective national and international One Health-based collaborative approach in responding to RVF emergencies.
    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
    摘要裂谷热(RVF)是由RVF病毒(RVFV)引起的蚊子传播的人畜共患疾病。人类的RVFV感染通常无症状或与轻度发热疾病有关,尽管也会发生更严重的出血性疾病和脑炎,死亡率很高。目前,没有获得许可的人类疫苗。已在小鼠中成功测试了基因工程四段RVFV变体(hRVFV-4s)作为潜在的减毒活人疫苗的安全性和有效性。反刍动物,尽管这种疫苗的保护作用的相关性在很大程度上仍然未知。在本研究中,我们评估了hRVFV-4s在RVFV感染小鼠模型中诱导的体液和细胞免疫。我们的结果证实,单剂量的hRVFV-4s在用高毒力的RVFV毒株腹膜内攻击后保护幼稚小鼠免于发展严重疾病方面非常有效,并且数据显示,初免-加强方案中的病毒中和(VN)血清抗体滴度明显高于单剂量。随后,来自初免-加强免疫接种的受体的VN抗体在转移到未处理的小鼠时显示出保护性。此外,hRVFV-4s疫苗接种诱导了显着的病毒特异性T细胞应答,如IFN-γELISpot测定所示,尽管这些T细胞在被动转移到原始受体小鼠后没有提供显著的保护。总的来说,这项研究强调了hRVFV-4s诱导的VN抗体是针对致死性RVFV感染的保护作用的主要相关因素。
    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.
    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(RVF)是由RVFV在人类和牲畜中引起的蚊子传播的病毒性人畜共患病。目前,目前尚无批准的疫苗或抗病毒疗法.此外,在沙特阿拉伯,缺乏常规筛查系统来监测人和动物的RVFV,这阻碍了预防措施的设计和开发以及对未来疫情的预测和RVFV的潜在复发。因此,我们进行了克隆,测序,测序和系统发育分析,核衣壳(N)蛋白基因。序列分析显示与从人和动物报告的RVFV分离株高度相似性。来自沙特阿拉伯的分离株(KU978775-Human)观察到最高的相似性(99.5%),其次是99.1%,来自其他位置的四个RVFV分离株(人和牛)。在整个N蛋白基因序列中观察到总共51个核苷酸和31个氨基酸的变异。系统发育关系与从沙特阿拉伯收集的其他分离株形成了封闭簇。因此,我们报告克隆,测序,测序和来自沙特阿拉伯的RVFV-N蛋白基因的系统发育分析。
    Rift Valley Fever (RVF) is a mosquito-borne viral zoonosis caused by RVFV in humans and livestock. Currently, there are no approved vaccines or antiviral therapies available. Additionally, in Saudi Arabia, there is a lack of a routine screening system to monitor RVFV in humans and animals which hinders to design and develop the preventive measures as well as the prediction of future outbreaks and the potential re-emergence of RVFV. Hence, we have performed the cloning, sequencing, and phylogenetic analysis, of nucleocapsid (N) protein gene. The sequence analysis showed high similarities with RVFV isolates reported from humans and animals. The highest similarity (99.5%) was observed with an isolate from Saudi Arabia (KU978775-Human) followed by 99.1% with four RVFV isolates (Human and Bovine) from other locations. A total of 51 nucleotides and 31 amino acid variations were observed throughout the N protein gene sequences. The phylogenetic relationship formed closed clusters with other isolates collected from Saudi Arabia. Thus, we report of the cloning, sequencing, and phylogenetic analysis of the RVFV-N protein gene from Saudi Arabia.
    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
    裂谷热静脉病毒(RVFV)是一种人畜共患病原体,可引起牲畜和人类的裂谷热(RVF)。目前,没有许可的人类疫苗或抗病毒药物来控制RVF。尽管多种动物和人类都容易感染RVFV,影响易感性的宿主因素尚不清楚。为了确定RVFV复制所必需的宿主因子或基因,我们在人A549细胞中进行了CRISPR-Cas9基因敲除筛选。然后,我们使用siRNA介导的敲除和CRISPR-Cas9介导的敲除研究验证了推定的基因。通过测量细胞内病毒RNA积累来评估候选基因在病毒复制周期中的作用。并使用噬斑测定或TCID50测定分析病毒滴度。我们鉴定了大约900个可能参与RVFV感染和复制的基因。使用siRNA介导的敲减对六个基因对病毒复制的影响的进一步评估表明,沉默两个基因(WDR7和LRP1)显着损害了RVFV复制。为了进一步分析,我们专注于WDR7基因,因为LRP1基因在RVFV复制中的作用之前已经有详细描述.产生WDR7敲除A549细胞系,并用于剖析WRD7对布尼亚病毒的影响,RVFV,和一种直鼻病毒,LaCrosse脑炎病毒(LACV)。我们观察到WDR7敲除细胞对胞内RVFVRNA水平和病毒滴度的显著影响。在细胞内RNA水平,与LACV复制相比,WRD7在其复制周期的后期(24小时)影响了RVFV复制,在较早的复制阶段(12小时)受到影响。总之,我们确定WDR7是两种不同病毒复制的必需宿主因子,RVFV和LACV,两者都属于Bunyavirales命令。未来的研究将研究WDR7促进静脉病毒复制的机制作用。
    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.
    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)基于其对牲畜的威胁和引起人类出血热的能力,被认为是高度生物防御的优先事项。感染RVFV的家畜也是人类直接接触污染血液感染的重要危险因素,组织,和流产的胎儿材料。因此,受影响地区的牲畜疫苗接种具有直接的双重利益和一种健康方法,可以保护数百万只动物的生命,并消除严重的,有时致命的人类裂谷热(RVF)疾病的风险。最近,我们开发了一种牛疱疹病毒1型(BoHV-1)四重基因突变病毒(BoHV-1qmv)载体,该载体由于包膜蛋白UL49.5,糖蛋白G(gG)的缺失而缺乏毒力和免疫抑制特性,gE细胞质尾,和US9编码序列。在目前的研究中,我们通过整合嵌合基因序列来进一步设计BoHV-1qmv,以表达可蛋白水解切割的多蛋白:RVFV包膜蛋白Gn胞外域序列与牛粒细胞-巨噬细胞集落刺激因子(GMCSF)和Gc融合,导致针对牲畜的RVFV的活BoHV-1qmv载体亚单位疫苗。体外,产生的重组病毒,BoHV-1qmvSub-RVFV,以高滴度在细胞培养物中复制。由疫苗病毒表达的嵌合Gn-GMCSF和Gc蛋白形成Gn-Gc复合物。在小牛中,BoHV-1qmv亚RVFV疫苗接种是安全的,诱导中等水平的RVFV疫苗株,MP12特异性中和抗体滴度。此外,在体外用热灭活的MP12抗原刺激时,与BoHV-1qmv(载体)疫苗接种的小牛相比,疫苗接种小牛的外周血单核细胞的干扰素-γ转录水平提高了6倍.基于这些发现,我们认为,单剂量BoHV-1qmvSub-RVFV疫苗可产生保护性RVFV-MP12特异性体液和细胞免疫应答.因此,BoHV-1qmv亚RVFV可能是牛针对RVFV的保护性亚单位疫苗。
    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.
    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
    裂谷热静脉病毒(RVFV)是一种人畜共患的蚊子传播的虫媒病毒,对人类和动物构成严重威胁。易感宿主对预防RVFV具有重要意义。适当的动物模型有助于更好地了解疾病的发生和发展,以及控制措施和疫苗研究。本文就动物宿主在病毒维持中的作用作一综述,并总结了RVFV的宿主范围。我们列出了在RVFV研究过程中一些常见的动物模型,这将为RVFV的预防和治疗提供一些重要的见解,以及研究裂谷热(RVF)的发病机制和疫苗。
    Rift Valley fever phlebovirus (RVFV) is a zoonotic mosquito-transmitted arbovirus, presenting a serious threat to humans and animals. Susceptible hosts are of great significance for the prevention of RVFV. Appropriate animal models are helpful to better understand the onset and development of diseases, as well as the control measures and vaccine research. This review focuses on the role of animal hosts in the maintenance of the virus, and summarizes the host range of RVFV. We list some common animal models in the process of RVFV research, which would provide some important insights into the prevention and treatment of RVFV, as well as the study of Rift Valley fever (RVF) pathogenesis and vaccines.
    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)是一种负义分段RNA病毒,可引起严重的出血热。三节段病毒基因组编码六(6)个多功能蛋白质,其在复制周期中的各种不同阶段与宿主因子接合。S片段编码核蛋白(N)和非结构蛋白S(NSs),M区段编码病毒糖蛋白Gn和Gc以及非结构蛋白M(NSm),L区段编码病毒聚合酶(L)。病毒糖蛋白Gn和Gc通过与许多宿主因子结合而负责进入。我们最近的研究发现了一种清道夫受体,LDL受体相关蛋白1(Lrp1),作为RVFV和相关病毒的潜在病毒宿主因子,包括Oropouche病毒(OROV)感染。巧合的是,最近的几项研究确定了其他LDL家族蛋白作为其他病毒家族的病毒进入因子和受体。总的来说,这些观察结果表明,高度保守的LDL家族蛋白可能在促进来自多个不同家族的病毒进入方面发挥重要作用.鉴于病毒和宿主因素在感染过程中的重要作用,这些相互作用的表征对于中和抗体和疫苗的治疗靶向至关重要。
    Rift Valley Fever Virus (RVFV) is a negative sense segmented RNA virus that can cause severe hemorrhagic fever. The tri-segmented virus genome encodes for six (6) multifunctional proteins that engage host factors at a variety of different stages in the replication cycle. The S segment encodes nucleoprotein (N) and nonstructural protein S (NSs), the M segment encodes viral glycoproteins Gn and Gc as well as nonstructural protein M (NSm) and the L segment encodes the viral polymerase (L). Viral glycoproteins Gn and Gc are responsible for entry by binding to a number of host factors. Our recent studies identified a scavenger receptor, LDL receptor related protein 1 (Lrp1), as a potential pro-viral host factor for RVFV and related viruses, including Oropouche virus (OROV) infection. Coincidentally, several recent studies identified other LDL family proteins as viral entry factors and receptors for other viral families. Collectively, these observations suggest that highly conserved LDL family proteins may play a significant role in facilitating entry of viruses from several distinct families. Given the significant roles of viral and host factors during infection, characterization of these interactions is critical for therapeutic targeting with neutralizing antibodies and vaccines.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    裂谷热静脉病毒(RVFV)是一种人畜共患病原体,可引起牲畜和人类的裂谷热(RVF)。目前,没有许可的人类疫苗或抗病毒药物来控制RVF。尽管多种动物和人类都容易感染RVFV,影响易感性的宿主因素尚不清楚。
    为了确定RVFV复制所必需的宿主因子或基因,我们在人A549细胞中进行了CRISPR-Cas9敲除筛选。然后,我们使用siRNA介导的基因敲除和CRISPR-Cas9介导的基因敲除研究来验证推定的基因。分别。通过测量细胞内病毒RNA积累来评估候选基因在病毒复制周期中的作用。和通过噬斑测定或TCID50测定的病毒滴度。
    我们鉴定了大约900个可能参与RVFV感染和复制的基因。使用siRNA介导的敲减对六个基因对病毒复制的影响的进一步评估发现,沉默两个基因(WDR7和LRP1)显着损害了RVFV复制。为了进一步分析,我们专注于WDR7基因,因为LRP1在RVFV复制中的作用之前已经有详细描述.产生敲除A549细胞系,并用于剖析WRD7对RVFV和另一种布尼亚病毒的影响,LaCrosse脑炎病毒(LACV)。我们观察到WDR7敲除细胞对胞内RVFVRNA水平和病毒滴度的显著影响。在细胞内RNA水平,当与受较早复制阶段(12h)影响的LACV相比时,WRD7在其复制周期的较晚阶段(24h)影响RVFV复制。
    总之,我们已经确定WDR7是两个相关布尼亚病毒复制的必需宿主因子,RVFV和LACV。未来的研究将研究WDR7促进尼伯病毒复制的机制作用。
    裂谷热静脉病毒是一种高后果病原体,感染多种动物物种和人类。目前,目前尚无可用于治疗人类裂谷热和防止裂谷热病毒侵入非流行国家的控制措施.RVFV在流行国家对动物和人类健康构成重大威胁。RVFV复制取决于主机的机器来完成其复制周期。因此,控制病毒复制的一种方法是破坏病毒和对复制重要的宿主蛋白之间的相互作用。在这项研究中,我们确定了一个宿主因素,WDR7基因,这对于RVFV复制至关重要。这种宿主因子的鉴定很重要,因为它可能导致抗病毒策略的发展,以控制人和动物的裂谷热。
    UNASSIGNED: 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.
    UNASSIGNED: To identify the host factors or genes essential for RVFV replication, we conducted a CRISPR-Cas9 knock-out screen in human A549 cells. We then validated the putative genes using siRNA-mediated knockdowns and CRISPR-Cas9-mediated knockout studies, respectively. The role of a candidate gene in the virus replication cycle was assessed by measuring intracellular viral RNA accumulation, and the virus titers by plaque assay or TCID50 assay.
    UNASSIGNED: 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 knockdowns found that silencing two genes (WDR7 and LRP1) significantly impaired RVFV replication. For further analysis, we focused on the WDR7 gene since the role of LRP1 in RVFV replication was previously described in detail. Knock-out A549 cell lines were generated and used to dissect the effect of WRD7 on RVFV and another bunyavirus, La Crosse encephalitis virus (LACV). We observed significant effects of WDR7 knock-out 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 (24h) when compared to LACV which was affected an earlier replication phase (12h).
    UNASSIGNED: In summary, we have identified WDR7 as an essential host factor for the replication of two relevant bunyaviruses, RVFV and LACV. Future studies will investigate the mechanistic role by which WDR7 facilitates Phlebovirus replication.
    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(RVF)是一种蚊子传播的病毒性人畜共患病,可导致反刍动物的胎儿和新生儿高死亡率,并导致人类轻度至致命的出血热。没有许可的RVF疫苗用于人类,而用于牲畜,商业上可获得的疫苗都是活的减毒的或灭活的,并且具有不希望的特征。减毒RVF活疫苗与反刍动物的致畸性和残留毒力有关,而灭活的活疫苗需要多次免疫来诱导和维持保护性免疫。此外,几乎所有许可的裂谷热疫苗都缺乏区分感染与接种疫苗的动物(DIVA)的特性,这使得它们不适合在裂谷热非流行国家使用.为了解决这些限制,正在开发与DIVA兼容的新型RVF疫苗,其安全性和有效性优于许可的疫苗,通过更好地了解RVF病毒的分子生物学和重组DNA技术的进步,从根本上得到了帮助。对于其中一些候选RVF疫苗,已在发现/可行性阶段证明了灭菌免疫,副作用最小。这篇综述强调了迄今为止在RVF疫苗研究和开发方面取得的进展,并讨论了突出的研究差距。
    Rift Valley fever (RVF) is a mosquito-borne viral zoonosis that causes high fetal and neonatal mortality in ruminants and a mild to fatal hemorrhagic fever in humans. There are no licensed RVF vaccines for human use while for livestock, commercially available vaccines are all either live attenuated or inactivated and have undesirable characteristics. The live attenuated RVF vaccines are associated with teratogenicity and residual virulence in ruminants while the inactivated ones require multiple immunisations to induce and maintain protective immunity. Additionally, nearly all licensed RVF vaccines lack the differentiating infected from vaccinated animals (DIVA) property making them inappropriate for use in RVF nonendemic countries. To address these limitations, novel DIVA-compatible RVF vaccines with better safety and efficacy than the licensed ones are being developed, aided fundamentally by a better understanding of the molecular biology of the RVF virus and advancements in recombinant DNA technology. For some of these candidate RVF vaccines, sterilizing immunity has been demonstrated in the discovery/feasibility phase with minimal adverse effects. This review highlights the progress made to date in RVF vaccine research and development and discusses the outstanding research gaps.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号