Rift Valley fever virus

裂谷热病毒
  • 文章类型: Journal Article
    裂谷热病毒(RVFV)感染,一种高致病性蚊子传播的人畜共患病毒,引发严重的炎症发病机制,但炎症激活的潜在机制目前尚不清楚。这里,我们报告说,RVFV的非结构蛋白NSs触发线粒体损伤,激活NLRP3炎性体,导致体内病毒发病机制。发现NSs的宿主转录抑制作用导致髓系细胞白血病-1(MCL-1)的快速下调,Bcl-2(B细胞淋巴瘤蛋白2)蛋白家族的促存活成员。MCL-1下调导致线粒体中的BAK激活,这触发了mtROS的产生和氧化的线粒体DNA(ox-mtDNA)释放到细胞质中。胞浆ox-mtDNA结合并激活触发RVFV感染细胞中NLRP3-GSDMD细胞凋亡的NLRP3炎性体。在诱导转录抑制中受损的NSs突变病毒(RVFV-NSsRM)既不触发MCL-1下调,也不触发NLRP3-GSDMD焦亡。Nlrp3-/-小鼠模型的RVFV感染表明,RVFV触发的NLRP3细胞凋亡有助于体内RVFV炎性发病机理和致命感染。用RVFV-NSsRM突变病毒感染类似地显示减轻的炎症发病机制和降低的死亡率。一起来看,这些结果揭示了毒力因子通过诱导宿主转录抑制来激活线粒体MCL-1-BAK轴从而触发NLRP3依赖性炎症发病机制的机制.
    Infection of Rift Valley fever virus (RVFV), a highly pathogenic mosquito-borne zoonotic virus, triggers severe inflammatory pathogenesis but the underlying mechanism of inflammation activation is currently unclear. Here, we report that the non-structural protein NSs of RVFV triggers mitochondrial damage to activate the NLRP3 inflammasome leading to viral pathogenesis in vivo. It is found that the host transcription inhibition effect of NSs causes rapid down-regulation of myeloid cell leukemia-1(MCL-1), a pro-survival member of the Bcl-2 (B-cell lymphoma protein 2) protein family. MCL-1 down-regulation led to BAK activation in the mitochondria, which triggered mtROS production and release of oxidized mitochondrial DNA (ox-mtDNA) into the cytosol. Cytosolic ox-mtDNA binds and activates the NLRP3 inflammasome triggering NLRP3-GSDMD pyroptosis in RVFV infected cells. A NSs mutant virus (RVFV-NSsRM) that is compromised in inducing transcription inhibition did not trigger MCL-1 down-regulation nor NLRP3-GSDMD pyroptosis. RVFV infection of the Nlrp3-/- mouse model demonstrated that the RVFV-triggered NLRP3 pyroptosis contributed to RVFV inflammatory pathogenesis and fatal infection in vivo. Infection with the RVFV-NSsRM mutant virus similarly showed alleviated inflammatory pathogenesis and reduced fatality rate. Taken together, these results revealed a mechanism by which a virulence factor activates the mitochondrial MCL-1-BAK axis through inducing host transcription inhibition to trigger NLRP3-dependent inflammatory pathogenesis.
    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
    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
    背景:属于Phenuiviridae家族的裂谷热病毒(RVFV)是一种称为裂谷热(RVF)的人畜共患疾病的原因。目前,RVFV已经从非洲传播到亚洲,并且由于其导致高死亡率的能力,它严重影响了许多社会的人类健康和经济发展。临床需要用于RVFV感染的血清诊断的高度特异性和敏感性系统。
    方法:用重组RVFV核衣壳(rRVFV-N)蛋白免疫BALB/c小鼠,脾细胞与SP2/0骨髓瘤细胞融合,建立杂交瘤细胞系。纯化并表征分泌的单克隆抗体(MAb)。建立并评估了使用新MAb检测IgG和IgM的酶联免疫吸附测定(ELISA)系统。与基于灭活病毒和兔多克隆抗体的ELISA系统相比,测试了来自肯尼亚的96名志愿者和93名疑似RVF患者的血清样品。
    结果:建立了三种抗rRVFV-N蛋白的单克隆抗体。基于MAb的夹心IgGELISA和IgM捕获ELISA的性能与使用灭活病毒或多克隆抗体的ELISA系统完全匹配。
    结论:开发了重组RVFV-N蛋白特异性单克隆抗体,它们为RVFV研究提供了有用的工具。用于检测IgG和IgM的基于MAb的ELISA系统为诊断人的RVFV感染提供了安全和有用的选择。
    BACKGROUND: Rift Valley fever virus (RVFV) belonging to the Phenuiviridae family is responsible for a zoonotic disease called Rift Valley fever (RVF). Currently, RVFV has spread from Africa to Asia, and due to its ability to cause high mortality rates, it has significantly impacted human health and economic development in many societies. Highly specific and sensitive systems for sero-diagnosis of RVFV infection are needed for clinical use.
    METHODS: BALB/c mice were immunized with recombinant RVFV nucleocapsid (rRVFV-N) protein and the spleen cells fused with SP2/0 myeloma cells to create hybridoma cell lines. The secreted monoclonal antibodies (MAbs) were purified and characterized. Enzyme-linked immunosorbent assay (ELISA) systems for the detection of IgG and IgM using the new MAbs were established and evaluated. Serum samples from 96 volunteers and 93 patients of suspected RVF from Kenya were tested compared with the ELISA systems based on inactivated viruses and the rabbit polyclonal antibody.
    RESULTS: Three monoclonal antibodies against rRVFV-N protein were established. The performance of the MAb-based sandwich IgG ELISA and the IgM capture ELISA perfectly matched the ELISA systems using the inactivated virus or the polyclonal antibody.
    CONCLUSIONS: Recombinant RVFV-N protein-specific MAbs were developed and they offer useful tools for RVFV studies. The MAb-based ELISA systems for detecting IgG and IgM offer safe and useful options for diagnosing RVFV infections in humans.
    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)病毒在世界范围内普遍存在,对人类生命和财产构成严重威胁。RVF病毒聚合酶在病毒的复制和转录中起着至关重要的作用。这里,我们描述了如何表达和纯化这种聚合酶,并对其体外活性测定进行测试。
    Rift Valley fever (RVF) virus is widespread worldwide and poses a severe threat to human life and property. RVF viral polymerase plays a vital role in the replication and transcription of the virus. Here, we describe how to express and purify this polymerase and perform tests for its in vitro activity assays.
    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)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
    全球人类活动的增加和农村地区的快速发展增加了人类与野生动物接触的可能性。大多数布尼亚病毒是人畜共患的,爆发可能会导致大量的生命损失,经济收缩,和社会不稳定。许多布尼亚病毒需要在最高水平的生物防护中进行操作,如生物安全4级(BSL-4)实验室,这种资源的匮乏限制了这些病原体疫苗的开发速度。同时,新技术已经创造出来,用来创新疫苗,如mRNA疫苗平台和基于生物信息学的抗原设计。这里,我们总结了目前需要在最高生物防护水平下工作的三种不同布尼亚病毒的疫苗开发:克里米亚-刚果出血热病毒(CCHFV),裂谷热病毒(RVFV),和汉坦病毒(HTNV),并提供了可以进一步探索的观点和潜在的未来方向,以开发针对人类和牲畜的特定疫苗。
    Increased human activities around the globe and the rapid development of once rural regions have increased the probability of contact between humans and wild animals. A majority of bunyaviruses are of zoonotic origin, and outbreaks may result in the substantial loss of lives, economy contraction, and social instability. Many bunyaviruses require manipulation in the highest levels of biocontainment, such as Biosafety Level 4 (BSL-4) laboratories, and the scarcity of this resource has limited the development speed of vaccines for these pathogens. Meanwhile, new technologies have been created, and used to innovate vaccines, like the mRNA vaccine platform and bioinformatics-based antigen design. Here, we summarize current vaccine developments for three different bunyaviruses requiring work in the highest levels of biocontainment: Crimean-Congo Hemorrhagic Fever Virus (CCHFV), Rift Valley Fever Virus (RVFV), and Hantaan virus (HTNV), and provide perspectives and potential future directions that can be further explored to advance specific vaccines for humans and livestock.
    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)

公众号