Vesicular stomatitis Indiana virus

囊性口炎印第安纳病毒
  • 文章类型: Journal Article
    已经研究了组蛋白去乙酰化家族蛋白在通过去乙酰化非组蛋白蛋白调节病毒复制中的功能。RIG-I(维甲酸诱导基因I)是RNA病毒诱导的先天抗病毒信号通路中的关键蛋白。我们之前的研究表明,HDAC8(组蛋白去乙酰化酶8)参与先天抗病毒免疫反应,但病毒感染过程中的潜在机制仍不清楚。在这项研究中,我们表明HDAC8参与了水泡性口炎病毒(VSV)复制的调节。HDAC8的过表达抑制,而敲低促进VSV复制。进一步的探索表明,HDAC8与RIG-I相互作用并脱乙酰,这最终导致增强先天抗病毒免疫反应。总的来说,我们的数据清楚地表明,HDAC8通过促进RIG-I介导的干扰素产生和下游信号通路抑制VSV复制.
    Histone deacetylates family proteins have been studied for their function in regulating viral replication by deacetylating non-histone proteins. RIG-I (Retinoic acid-inducible gene I) is a critical protein in RNA virus-induced innate antiviral signaling pathways. Our previous research showed that HDAC8 (histone deacetylase 8) involved in innate antiviral immune response, but the underlying mechanism during virus infection is still unclear. In this study, we showed that HDAC8 was involved in the regulation of vesicular stomatitis virus (VSV) replication. Over-expression of HDAC8 inhibited while knockdown promoted VSV replication. Further exploration demonstrated that HDAC8 interacted with and deacetylated RIG-I, which eventually lead to enhance innate antiviral immune response. Collectively, our data clearly demonstrated that HDAC8 inhibited VSV replication by promoting RIG-I mediated interferon production and downstream signaling pathway.
    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
    疫苗的热稳定性,特别是包膜病毒载体疫苗,在任何需要的地方对他们的交付仍然是一个挑战。病毒载体疫苗的冷冻干燥是一种有前途的方法,但由于从病毒的外部和内部去除水的过程仍然具有挑战性。在包膜病毒的情况下,冷冻干燥会导致信封上的应力增加,这通常会导致病毒的失活。在这项研究中,我们设计了一种冷冻干燥表达SARS-CoV-2刺突糖蛋白的重组水泡性口炎病毒(VSV)的方法。由于VSV的包膜由50%的脂质和50%的蛋白质组成,制剂研究集中在载体的蛋白质和脂质部分。制剂主要使用蔗糖制备,海藻糖,和山梨糖醇作为冷冻保护剂;甘露醇作为冻干保护剂;和组氨酸作为缓冲剂。最初,在不同的最终水分含量水平下,研究了rVSV-SARS-CoV-2的感染性和滤饼稳定性。在3-6%的水分含量下发现了感染性病毒滴度的高恢复(〜0.5至1log损失),冻干蛋糕没有变质。为了进一步减少感染性病毒滴度损失,研究了赋形剂的组成和浓度。冷冻保护剂和冻干保护剂从5%增加到10%,加上0.5%的明胶,导致提高的感染性病毒滴度的恢复和稳定的饼形成。此外,冷冻干燥过程的二次干燥温度对rVSV-SARS-CoV-2的感染性有显著影响。当温度升高到20°C以上时,载体的感染性急剧下降。在整个长期稳定性研究中,含有10%糖(蔗糖/海藻糖)的配方,10%甘露醇,0.5%明胶,和10mM组氨酸在2-8℃下显示令人满意的稳定性6个月。这种冷冻干燥工艺的开发和优化的配方最大限度地减少了对昂贵的冷链配送系统的需求。
    The thermostability of vaccines, particularly enveloped viral vectored vaccines, remains a challenge to their delivery wherever needed. The freeze-drying of viral vectored vaccines is a promising approach but remains challenging due to the water removal process from the outer and inner parts of the virus. In the case of enveloped viruses, freeze-drying induces increased stress on the envelope, which often leads to the inactivation of the virus. In this study, we designed a method to freeze-dry a recombinant vesicular stomatitis virus (VSV) expressing the SARS-CoV-2 spike glycoprotein. Since the envelope of VSV is composed of 50% lipids and 50% protein, the formulation study focused on both the protein and lipid portions of the vector. Formulations were prepared primarily using sucrose, trehalose, and sorbitol as cryoprotectants; mannitol as a lyoprotectant; and histidine as a buffer. Initially, the infectivity of rVSV-SARS-CoV-2 and the cake stability were investigated at different final moisture content levels. High recovery of the infectious viral titer (~0.5 to 1 log loss) was found at 3-6% moisture content, with no deterioration in the freeze-dried cakes. To further minimize infectious viral titer loss, the composition and concentration of the excipients were studied. An increase from 5 to 10% in both the cryoprotectants and lyoprotectant, together with the addition of 0.5% gelatin, resulted in the improved recovery of the infectious virus titer and stable cake formation. Moreover, the secondary drying temperature of the freeze-drying process showed a significant impact on the infectivity of rVSV-SARS-CoV-2. The infectivity of the vector declined drastically when the temperature was raised above 20 °C. Throughout a long-term stability study, formulations containing 10% sugar (sucrose/trehalose), 10% mannitol, 0.5% gelatin, and 10 mM histidine showed satisfactory stability for six months at 2-8 °C. The development of this freeze-drying process and the optimized formulation minimize the need for a costly cold chain distribution system.
    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
    溶瘤病毒(OV)治疗的快速发展领域需要开发新的和改进的分析方法,以在生产和开发过程中表征病毒。病毒蛋白的准确监测和绝对定量对于OV产品表征至关重要,可以促进对感染的理解,免疫原性,和病毒复制的发展阶段。像多反应监测(MRM)这样的靶向质谱方法提供了直接检测和定量由替代肽代表的特定靶向蛋白质的可靠方法。我们通过将超高效液相色谱(UPLC)与Sciex6500三级四极杆质谱仪相结合,利用了MRM的功能,开发了一种准确,绝对地定量假型水疱性口炎病毒(VSV)的结构蛋白的测定法。用作新的生物治疗剂(以下称为VSV-GP,以区别于天然VV)。新的UPLC-MRM方法使用重标记的参考标准替代肽提供绝对定量。以已知的精确量添加到标准和样品中时,参考标准对样品制备和/或仪器性能期间的任何小扰动进行标准化和说明,导致准确和精确的定量。由于MRM的多重性质,同时对所有靶向蛋白进行定量。优化的测定已被增强以量化经处理的GP1和GP2蛋白的比率,同时测量包膜蛋白GP复合物(GPC;全长GPC)的任何剩余或未加工形式。
    目的:近年来,溶瘤病毒治疗的发展取得了相当大的势头。水泡性口炎病毒糖蛋白(VSV-GP)是溶瘤病毒治疗平台中出现的一种新的生物治疗剂。可以准确和精确地定量病毒蛋白的新型分析测定法是成功开发作为生物治疗剂的病毒载体的必要条件。我们开发了一种基于超高效液相色谱多反应监测的测定法,以量化VSV-GP不同结构蛋白的绝对浓度。GP复合物(GPC)的完全处理是病毒感染性的先决条件。该测定法扩展了定量全长GPC的潜力,这提供了对GPC处理的理解(以及分别对GP1和GP2的定量)。我们在追踪用VSV-GP感染的HEK-293-F生产细胞系中的GPC处理中使用该测定。
    The rapidly developing field of oncolytic virus (OV) therapy necessitates the development of new and improved analytical approaches for the characterization of the virus during production and development. Accurate monitoring and absolute quantification of viral proteins are crucial for OV product characterization and can facilitate the understanding of infection, immunogenicity, and development stages of viral replication. Targeted mass spectrometry methods like multiple reaction monitoring (MRM) offer a robust way to directly detect and quantify specific targeted proteins represented by surrogate peptides. We have leveraged the power of MRM by combining ultra-high performance liquid chromatography (UPLC) with a Sciex 6500 triple-stage quadrupole mass spectrometer to develop an assay that accurately and absolutely quantifies the structural proteins of a pseudotyped vesicular stomatitis virus (VSV) intended for use as a new biotherapeutic (designated hereafter as VSV-GP to differentiate it from native VSV). The new UPLC-MRM method provides absolute quantification with the use of heavy-labeled reference standard surrogate peptides. When added in known exact amounts to standards and samples, the reference standards normalize and account for any small perturbations during sample preparation and/or instrument performance, resulting in accurate and precise quantification. Because of the multiplexed nature of MRM, all targeted proteins are quantified at the same time. The optimized assay has been enhanced to quantify the ratios of the processed GP1 and GP2 proteins while simultaneously measuring any remaining or unprocessed form of the envelope protein GP complex (GPC; full-length GPC).
    OBJECTIVE: The development of oncolytic viral therapy has gained considerable momentum in recent years. Vesicular stomatitis virus glycoprotein (VSV-GP) is a new biotherapeutic emerging in the oncolytic viral therapy platform. Novel analytical assays that can accurately and precisely quantify the viral proteins are a necessity for the successful development of viral vector as a biotherapeutic. We developed an ultra-high performance liquid chromatography multiple reaction monitoring-based assay to quantify the absolute concentrations of the different structural proteins of VSV-GP. The complete processing of GP complex (GPC) is a prerequisite for the infectivity of the virus. The assay extends the potential for quantifying full-length GPC, which provides an understanding of the processing of GPC (along with the quantification of GP1 and GP2 separately). We used this assay in tracking GPC processing in HEK-293-F production cell lines infected with VSV-GP.
    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
    干扰素(IFN)系统保护哺乳动物免受病毒感染引起的疾病。IFN合成是由病毒感染激活的模式识别受体信号通路诱导的。IFN从感染的细胞分泌,并通过结合细胞表面受体并触发数百个IFN刺激的基因和蛋白质的诱导作用于邻近细胞。其中许多阻止病毒复制的不同步骤。IFN诱导的四三肽重复蛋白(IFIT)是RNA结合蛋白家族。我们和其他人以前曾报道,IFIT2保护小鼠免受许多嗜神经RNA病毒的侵害;事实上,Ifit2-/-小鼠对水泡性口炎病毒(VSV)的鼻内或皮下感染非常敏感。这里,使用新生成的条件敲除鼠标,我们报道,Ifit2表达仅在神经元细胞中的消融足以使小鼠对由鼻内,但不是皮下的,VSV感染。另一种转基因小鼠品系,表达不能结合RNA的突变体IFIT2,与Ifit2-/-小鼠一样容易感染VSV。这些结果表明,IFIT2RNA结合活性对于保护小鼠免受VSV鼻内感染引起的神经系统疾病至关重要。IMPORTANCE干扰素(IFN)抗病毒作用由干扰素刺激基因编码的蛋白质介导。IFN刺激基因(IFIT2)是这样一种蛋白质,抑制许多RNA病毒在小鼠大脑中的复制和由此产生的神经病理学。我们的研究揭示了IFIT2的工作原理。通过仅在神经元细胞中消除Ifit2表达,使用新生成的条件敲除鼠标行,我们表明,感染小鼠的神经元中的Ifit2诱导是干扰素抗病毒功能所必需的。IFIT2没有已知的酶活性;相反,它通过与细胞或病毒蛋白或RNA结合而起作用。我们设计了一种新的小鼠品系,该品系表达了无法结合RNA的突变体IFIT2。这些小鼠对水泡性口炎病毒感染非常敏感,表明IFIT2的RNA结合特性对于其体内抗病毒功能至关重要。
    The interferon (IFN) system protects mammals from diseases caused by virus infections. IFN synthesis is induced by pattern recognition receptor signaling pathways activated by virus infection. IFN is secreted from the infected cells and acts upon neighboring cells by binding cell surface receptors and triggering induction of hundreds of IFN-stimulated genes and proteins, many of which block different steps of virus replication. The IFN-induced tetratricopeptide repeat proteins (IFIT) are a family of RNA-binding proteins. We and others have previously reported that IFIT2 protects mice from many neurotropic RNA viruses; indeed, Ifit2-/- mice are very susceptible to intranasal or subcutaneous infections with vesicular stomatitis virus (VSV). Here, using a newly generated conditional knockout mouse, we report that ablation of Ifit2 expression only in neuronal cells was sufficient to render mice susceptible to neuropathogenesis caused by intranasal, but not subcutaneous, infection of VSV. Another genetically modified mouse line, expressing a mutant IFIT2 that cannot bind RNA, was as susceptible to VSV infection as Ifit2-/- mice. These results demonstrated that IFIT2 RNA-binding activity is essential for protecting mice against neurological diseases caused by intranasal infection of VSV.IMPORTANCEInterferon\'s (IFN\'s) antiviral effects are mediated by the proteins encoded by the interferon-stimulated genes. IFN-stimulated genes (IFIT2) is one such protein, which inhibits replication of many RNA viruses in the mouse brain and the resultant neuropathology. Our study sheds light on how IFIT2 works. By ablating Ifit2 expression only in neuronal cells, using a newly generated conditional knockout mouse line, we showed that Ifit2 induction in the neurons of the infected mouse was necessary for antiviral function of interferon. IFIT2 has no known enzyme activity; instead, it functions by binding to cellular or viral proteins or RNAs. We engineered a new mouse line that expressed a mutant IFIT2 that cannot bind RNA. These mice were very susceptible to infection with vesicular stomatitis virus indicating that the RNA-binding property of IFIT2 was essential for its antiviral function in vivo.
    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
    疱疹病毒B(BV)是一种人畜共患病毒,属于单纯型,与人类单纯疱疹病毒(HSV)相同的属。BV通常在其自然宿主中建立无症状感染,猕猴.然而,在人类中,BV感染导致严重的神经系统疾病和死亡。因此,BV研究只能在高安全级别设施中进行(即,生物安全级别[BSL]4),BV进入的机制尚未完全阐明。在这项研究中,我们产生了假型水泡性口炎病毒(VSV)表达BV糖蛋白使用G-互补VSVΔG系统,我们命名为VSV/BVpv。我们发现四种BV糖蛋白(即,gB,gD,gH,和gL)是生产高滴度VSV/BVpv所需的。此外,VSV/BVpv细胞进入依赖于gD与其细胞受体nectin-1的结合。用内体酸化抑制剂预处理Vero细胞不影响VSV/BVpv感染。结果表明,VSV/BVpv通过与Vero细胞的质膜直接融合而进入,表明其进入途径与天然HSV相似。此外,我们开发了基于VSV/BVpv的化学发光还原中和测试(CRNT),该方法检测了猕猴血浆样品中抗BV的中和抗体,具有较高的灵敏度和特异性。至关重要的是,本研究中产生的VSV/BVpv可在BSL-2条件下通过gD-nectin-1相互作用以及BV与Vero细胞质膜的直接融合来研究初始进入过程。重要疱疹病毒B(BV)是一种针对人类的高致病性人畜共患病毒。BV属于Simplexvius属,与人类单纯疱疹病毒(HSV)相同的属。与HSV相比,BV的细胞进入机制尚不完全清楚。操纵感染性BV的研究程序应在生物安全级别(BSL)-4设施中进行。由于假病毒提供了一个安全的病毒进入模型,因为它们无法产生传染性子代病毒,我们试图通过修饰BV糖蛋白的表达结构来产生带有BV糖蛋白的假型水泡性口炎病毒(VSV/BVpv),并成功获得了高滴度的VSV/BVpv。这项研究为构建VSV/BVpv及其研究BV感染提供了新的信息。
    Herpes B virus (BV) is a zoonotic virus and belongs to the genus Simplexvius, the same genus as human herpes simplex virus (HSV). BV typically establishes asymptomatic infection in its natural hosts, macaque monkeys. However, in humans, BV infection causes serious neurological diseases and death. As such, BV research can only be conducted in a high containment level facility (i.e., biosafety level [BSL] 4), and the mechanisms of BV entry have not been fully elucidated. In this study, we generated a pseudotyped vesicular stomatitis virus (VSV) expressing BV glycoproteins using G-complemented VSV∆G system, which we named VSV/BVpv. We found that four BV glycoproteins (i.e., gB, gD, gH, and gL) were required for the production of a high-titer VSV/BVpv. Moreover, VSV/BVpv cell entry was dependent on the binding of gD to its cellular receptor nectin-1. Pretreatment of Vero cells with endosomal acidification inhibitors did not affect the VSV/BVpv infection. The result indicated that VSV/BVpv entry occurred by direct fusion with the plasma membrane of Vero cells and suggested that the entry pathway was similar to that of native HSV. Furthermore, we developed a VSV/BVpv-based chemiluminescence reduction neutralization test (CRNT), which detected the neutralization antibodies against BV in macaque plasma samples with high sensitivity and specificity. Crucially, the VSV/BVpv generated in this study can be used under BSL-2 condition to study the initial entry process through gD-nectin-1 interaction and the direct fusion of BV with the plasma membrane of Vero cells.IMPORTANCEHerpes B virus (BV) is a highly pathogenic zoonotic virus against humans. BV belongs to the genus Simplexvius, the same genus as human herpes simplex virus (HSV). By contrast to HSV, cell entry mechanisms of BV are not fully understood. The research procedures to manipulate infectious BV should be conducted in biosafety level (BSL)-4 facilities. As pseudotyped viruses provide a safe viral entry model because of their inability to produce infectious progeny virus, we tried to generate a pseudotyped vesicular stomatitis virus bearing BV glycoproteins (VSV/BVpv) by modification of expression constructs of BV glycoproteins, and successfully obtained VSV/BVpv with a high titer. This study has provided novel information for constructing VSV/BVpv and its usefulness to study BV 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
    新生儿Fc受体(FcRn)回收免疫球蛋白G,FcRn的抑制在临床上用于自身免疫性疾病的治疗。在这项工作中,使用水疱性口炎病毒(VSV)小鼠感染模型系统,我们确定了FcRn在病毒感染过程中的作用。虽然中和抗体的诱导和这些抗体的长期保护在FcRn缺陷小鼠中几乎没有受到影响,FcRn缺陷限制了天然IgG(VSV特异性)抗体的量。缺乏天然抗体(nAbs)限制了巨噬细胞中VSV的早期控制,病毒在几个器官中加速繁殖,导致VSV扩散到神经组织,导致致命的结果。将天然IgG过继转移到FcRn缺陷小鼠中限制了VSV在FcRn缺陷小鼠中的早期传播并提高了FcRn敲除小鼠的存活率。与此相符,在感染前用极低剂量的VSV接种FcRn小鼠类似地防止了感染后的死亡。总之,我们确定了nAb在VSV感染期间的重要性。缺乏FcRn限制了nAb,从而增强了对病毒感染的易感性。
    Neonatal Fc receptor (FcRn) recycles immunoglobulin G, and inhibition of FcRn is used clinically for treatment of autoimmune diseases. In this work, using the vesicular stomatitis virus (VSV) mouse infection model system, we determined the role of FcRn during virus infection. While induction of neutralizing antibodies and long-term protection of these antibodies was hardly affected in FcRn deficient mice, FcRn deficiency limited the amount of natural IgG (VSV-specific) antibodies. Lack of natural antibodies (nAbs) limited early control of VSV in macrophages, accelerated propagation of virus in several organs, led to the spread of VSV to the neural tissue resulting in fatal outcomes. Adoptive transfer of natural IgG into FcRn deficient mice limited early propagation of VSV in FcRn deficient mice and enhanced survival of FcRn knockout mice. In line with this, vaccination of FcRn mice with very low dose of VSV prior to infection similarly prevented death after infection. In conclusion we determined the importance of nAbs during VSV infection. Lack of FcRn limited nAbs and thereby enhanced the susceptibility to virus 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
    背景:转移性乳腺癌是女性癌症死亡的主要原因。目前的治疗选择通常与不良副作用和不良结局有关。证明需要有效的新疗法。免疫疗法可以在许多癌症中提供持久的结果;然而,转移性三阴性乳腺癌取得的成功有限.我们在临床前模型中测试了结合不同的免疫疗法是否可以针对转移性三阴性乳腺癌。
    方法:使用原发性和转移性4T1三阴性乳腺癌模型,我们检查了溶瘤性水泡性口炎病毒(VSVΔM51)的治疗作用,该病毒被设计为表达呼肠孤病毒衍生的融合相关小跨膜蛋白p14(VSV-p14)或p15(VSV-p15)。这些病毒单独递送或与通过过继转移负载α-半乳糖神经酰胺的树突状细胞介导的自然杀伤T(NKT)细胞活化疗法组合递送。
    结果:用VSV-p14或VSV-p15单独治疗原发性4T1肿瘤会增加免疫原性肿瘤细胞死亡,肿瘤生长减弱,与对照溶瘤病毒(VSV-GFP)处理和未处理的小鼠相比,以及增强的免疫细胞浸润和活化。当与NKT细胞活化疗法联合使用时,溶瘤VSV-p14和VSV-p15将所有小鼠的转移性肺负荷降低至无法检测的水平,并产生免疫记忆,如体外回忆反应增强(肿瘤杀伤和细胞因子产生)和再攻击后肿瘤生长受损所证明。
    结论:将NKT细胞免疫疗法与增强的溶瘤病毒疗法相结合,可提高肺转移的抗肿瘤免疫靶向性,并为转移性乳腺癌提供了有希望的治疗策略。
    BACKGROUND: Metastatic breast cancer is a leading cause of cancer death in woman. Current treatment options are often associated with adverse side effects and poor outcomes, demonstrating the need for effective new treatments. Immunotherapies can provide durable outcomes in many cancers; however, limited success has been achieved in metastatic triple negative breast cancer. We tested whether combining different immunotherapies can target metastatic triple negative breast cancer in pre-clinical models.
    METHODS: Using primary and metastatic 4T1 triple negative mammary carcinoma models, we examined the therapeutic effects of oncolytic vesicular stomatitis virus (VSVΔM51) engineered to express reovirus-derived fusion associated small transmembrane proteins p14 (VSV-p14) or p15 (VSV-p15). These viruses were delivered alone or in combination with natural killer T (NKT) cell activation therapy mediated by adoptive transfer of α-galactosylceramide-loaded dendritic cells.
    RESULTS: Treatment of primary 4T1 tumors with VSV-p14 or VSV-p15 alone increased immunogenic tumor cell death, attenuated tumor growth, and enhanced immune cell infiltration and activation compared to control oncolytic virus (VSV-GFP) treatments and untreated mice. When combined with NKT cell activation therapy, oncolytic VSV-p14 and VSV-p15 reduced metastatic lung burden to undetectable levels in all mice and generated immune memory as evidenced by enhanced in vitro recall responses (tumor killing and cytokine production) and impaired tumor growth upon rechallenge.
    CONCLUSIONS: Combining NKT cell immunotherapy with enhanced oncolytic virotherapy increased anti-tumor immune targeting of lung metastasis and presents a promising treatment strategy for metastatic breast cancer.
    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
    对溶瘤病毒疗法的反应中异质性的存在对临床有效性构成了障碍,因为对这种治疗的抗性可以通过抑制病毒在肿瘤内的传播而发生,可能导致治疗失败。这里我们显示4-辛基衣康酸(4-OI),克雷布斯循环代谢产物衣康酸酯的化学衍生物,在包括人和鼠抗性癌细胞系在内的各种模型中增强VSVΔ51的溶瘤病毒疗法,三维(3D)患者来源的结肠肿瘤和器官型脑肿瘤切片。此外,4-OI与VSVA51组合改善抗性鼠结肠肿瘤模型中的治疗结果。机械上,我们发现4-OI通过修饰MAVS和IKKβ中的半胱氨酸残基而独立于NRF2/KEAP1轴抑制癌细胞的抗病毒免疫。我们提出,代谢物衍生药物与溶瘤病毒试剂的组合可以通过直接干扰I型IFN和NF-κB介导的抗病毒反应来大大提高抗癌治疗效果。
    The presence of heterogeneity in responses to oncolytic virotherapy poses a barrier to clinical effectiveness, as resistance to this treatment can occur through the inhibition of viral spread within the tumor, potentially leading to treatment failures. Here we show that 4-octyl itaconate (4-OI), a chemical derivative of the Krebs cycle-derived metabolite itaconate, enhances oncolytic virotherapy with VSVΔ51 in various models including human and murine resistant cancer cell lines, three-dimensional (3D) patient-derived colon tumoroids and organotypic brain tumor slices. Furthermore, 4-OI in combination with VSVΔ51 improves therapeutic outcomes in a resistant murine colon tumor model. Mechanistically, we find that 4-OI suppresses antiviral immunity in cancer cells through the modification of cysteine residues in MAVS and IKKβ independently of the NRF2/KEAP1 axis. We propose that the combination of a metabolite-derived drug with an oncolytic virus agent can greatly improve anticancer therapeutic outcomes by direct interference with the type I IFN and NF-κB-mediated antiviral responses.
    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
    我们通过使用3种稳定制剂评估了冻干对2种基于水泡性口炎病毒的疫苗的体外作用,并证明了冻干/重组疫苗在豚鼠中的保护性免疫力。冻干增加了疫苗的稳定性,但特定的基于水疱性口炎病毒的疫苗都需要进行广泛的分析以优化稳定制剂.
    We evaluated the in vitro effects of lyophilization for 2 vesicular stomatitis virus-based vaccines by using 3 stabilizing formulations and demonstrated protective immunity of lyophilized/reconstituted vaccine in guinea pigs. Lyophilization increased stability of the vaccines, but specific vesicular stomatitis virus-based vaccines will each require extensive analysis to optimize stabilizing formulations.
    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
    人畜共患冠状病毒对人类健康构成持续威胁,新发现的蝙蝠传播病毒如猪急性腹泻综合征冠状病毒(SADS-CoV)导致仔猪高死亡率。体外研究表明,SADS-CoV可以感染来自不同物种的细胞系,包括人类,强调其对人类健康的潜在风险。然而,缺乏研究病毒进入的工具,随着疫苗或抗病毒治疗的缺乏,延续了这种威胁。为了解决这个问题,我们设计了水泡性口炎病毒(VSV)的感染性分子克隆,用SADS-CoV尖峰(S)替换其天然糖蛋白(G),并在3\'前导区插入Venus报告基因,以产生具有复制能力的rVSV-Venus-SADS病毒。rVSV-Venus-SADSS的连续传代导致在S蛋白的细胞质尾部中鉴定出11个氨基酸的截短,由于S蛋白的细胞膜锚定增加,这允许更有效的病毒繁殖。S蛋白整合到rVSV-Venus-SADSSΔ11颗粒中,对SADS-CoVS1蛋白免疫兔的血清中和敏感。此外,我们发现TMPRSS2促进SADS-CoV刺突介导的细胞进入。此外,我们使用初免-加强免疫策略评估了接种rVSV-Venus-SADSSΔ11的小鼠的血清中和能力,揭示针对SADS-CoV感染的有效中和抗体。总之,我们开发了一种安全实用的工具,用于研究SADS-CoV入室情况,并探索重组VSV载体SADS-CoV疫苗的潜力.重要的人畜共患冠状病毒,如猪急性腹泻综合征冠状病毒(SADS-CoV),对人类和动物健康构成持续威胁。为了解决这个问题,我们通过修改水泡性口炎病毒(VSV)设计了一个安全有效的工具,创造了一种具有复制能力的rVSV-金星-SADS病毒。通过连续通道,我们优化了病毒以增强膜锚定,病毒繁殖的关键因素。这种改良的病毒,rVSV-Venus-SADSSΔ11,被证明容易中和,为潜在的疫苗开辟道路。此外,我们的研究揭示了TMPRSS2在SADS-CoV进入中的作用。用rVSV-Venus-SADSSΔ11接种的小鼠产生了针对SADS-CoV的有效中和抗体。总之,我们的工作为研究SADS-CoV入门提供了一种安全实用的工具,并探索了重组VSV载体SADS-CoV疫苗的前景.
    Zoonotic coronaviruses pose a continuous threat to human health, with newly identified bat-borne viruses like swine acute diarrhea syndrome coronavirus (SADS-CoV) causing high mortality in piglets. In vitro studies indicate that SADS-CoV can infect cell lines from diverse species, including humans, highlighting its potential risk to human health. However, the lack of tools to study viral entry, along with the absence of vaccines or antiviral therapies, perpetuates this threat. To address this, we engineered an infectious molecular clone of Vesicular Stomatitis Virus (VSV), replacing its native glycoprotein (G) with SADS-CoV spike (S) and inserting a Venus reporter at the 3\' leader region to generate a replication-competent rVSV-Venus-SADS S virus. Serial passages of rVSV-Venus-SADS S led to the identification of an 11-amino-acid truncation in the cytoplasmic tail of the S protein, which allowed more efficient viral propagation due to increased cell membrane anchoring of the S protein. The S protein was integrated into rVSV-Venus-SADS SΔ11 particles, susceptible to neutralization by sera from SADS-CoV S1 protein-immunized rabbits. Additionally, we found that TMPRSS2 promotes SADS-CoV spike-mediated cell entry. Furthermore, we assessed the serum-neutralizing ability of mice vaccinated with rVSV-Venus-SADS SΔ11 using a prime-boost immunization strategy, revealing effective neutralizing antibodies against SADS-CoV infection. In conclusion, we have developed a safe and practical tool for studying SADS-CoV entry and exploring the potential of a recombinant VSV-vectored SADS-CoV vaccine.IMPORTANCEZoonotic coronaviruses, like swine acute diarrhea syndrome coronavirus (SADS-CoV), pose a continual threat to human and animal health. To combat this, we engineered a safe and efficient tool by modifying the Vesicular Stomatitis Virus (VSV), creating a replication-competent rVSV-Venus-SADS S virus. Through serial passages, we optimized the virus for enhanced membrane anchoring, a key factor in viral propagation. This modified virus, rVSV-Venus-SADS SΔ11, proved susceptible to neutralization, opening avenues for potential vaccines. Additionally, our study revealed the role of TMPRSS2 in SADS-CoV entry. Mice vaccinated with rVSV-Venus-SADS SΔ11 developed potent neutralizing antibodies against SADS-CoV. In conclusion, our work presents a secure and practical tool for studying SADS-CoV entry and explores the promise of a recombinant VSV-vectored SADS-CoV vaccine.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号