Inhibition of Enterovirus 71 by Adenosine Analog NITD008
逆转录病毒介导增强型绿色荧光蛋白基因在大鼠骨髓间充质干细胞中的表达1
逆转录病毒介导增强型绿色荧光蛋白基因在大鼠骨髓间充质干细胞中的表达1曾智凤,易著文*,黄丹琳,曹艳,何庆南,吴小川,党西强,何小解,莫双红中南大学湘雅二医院小儿肾脏病研究室,湖南长沙 (410011)E-mail:zengzhifeng198129@摘要:本文采用密度梯度离心法分离培养BMSCs,诱导培养液诱导其向成脂肪方向分化,并行细胞表面抗原鉴定,及油红O染色评价细胞成脂肪情况。
在此基础上,采用逆转录病毒pLEGFP-N1对细胞进行绿色荧光蛋白标记,观察细胞形态学改变及荧光表达的时间与强度,计算转染率,以期探讨绿色荧光蛋白标记SD大鼠BMSCs的可行性,为细胞移植寻找一种理想的示踪标记方法。
实验结果显示细胞扩增迅速,形态良好,纯度较高,经诱导后细胞内可见脂滴。
逆转录病毒载体pLEGFP-N1成功标记SD大鼠骨髓间充质干细胞,并对4周体外培养进行了良好的标记。
因此认为逆转录病毒pLEGF-N1转染效率高,转染成功的BMSCs可以长期稳定表达目的基因,是一种理想的病毒载体。
荧光蛋白标记技术可以作为种子细胞良好的示踪标记方法。
关键词:骨髓基质干细胞;成脂肪诱导;增强型绿色荧光蛋白1. 引言骨髓间充质干细胞(BMSCs)具有自我更新、多向分化潜能,且容易获取及体外扩增培养,成为近年来成体干细胞研究的热点[1..2]。
深入认识BMSCs植入受体后的生物学行为,对于探索其作用机制至关重要,这就需要有效标记BMSCs。
随着人们对增强型绿色荧光蛋白(EGFP)研究的逐渐深入,为解决这一难题提供了可能。
本实验旨在探讨逆转录病毒pLEGFP-N1转染BMSCs的可行性,进而寻找一种较为理想的细胞示踪标记方法。
2. 材料与方法2.1材料清洁级SD大鼠(150g),由中南大学湘雅二医院实验动物中心提供。
F12/DMEM,DMEM 培养基,胰蛋白酶,胎牛血清为Hyclone公司产品。
Ficoll-Paque淋巴细胞分离液为Pharmacia 公司产品。
肠道病毒71VP1基因的原核表达及其抗血清的制备_谢维欣
基金项目:国家自然科学基金(31272586);国家转基因重大专项(2009ZX08007-006B,2011ZX08007-002,2011ZX08008-004);山东省青年自然科学基金项目(ZR2010ZR029);山东省自然科学基金(ZR2010CM012);济南市高校院所主创新计划(201004027)作者单位:150030哈尔滨,东北农业大学生命科学学院(谢维欣,李杰,何洪彬);250100济南,山东省农业科学院奶牛研究中心(谢维欣,武建明,王洪梅,何洪彬);250100济南,济南军区军犬训练大队(方永志)*通信作者:何洪彬,E-mail :hongbinh@肠道病毒71型(Enterovirus 71,EV71)是导致手足口病(hand,foot and mouth disease,HFMD)以及心肌炎、肺水肿、无菌性脑膜炎等多种并发症的主要病原体之一,多发于5岁以下儿童,以1~3岁最为常见[1]。
EV71属于小RNA 病毒科肠道病毒属,病毒颗粒为二十面立体对称的球型结构,直径为24~30nm [2-3],常引起多种与神经系统相关的严重疾病[4]。
1969年EV71被首次报道,1974年从美国加利福尼亚暴发的神经系统疾病患者中分离得到[5],近年来在世界范围内多次爆发和流行,在我国流行该病毒类型多为C4亚型[6-7]。
EV71病毒衣壳由60个亚单位构成,每个亚单位由4种衣壳蛋白(VP1~VP4)组成,除了VP4包埋在病毒粒子外壳的内侧,VP1~VP3形成病毒的外表面,均可与患者的免疫系统直接接触[8-9],其中VP1具有较多主要的中和抗原决定簇且遗传多样性与病毒血清型也完全对应,其编码的衣壳蛋白在感染宿主细胞时起重要作用,因此VP1成为最重要的病毒诊断·论著·[文章编号]1000-8861(2013)01-0038-05肠道病毒71VP1基因的原核表达及其抗血清的制备谢维欣,武建明,李杰,方永志,王洪梅,何洪彬*[摘要]目的克隆并表达重组肠道病毒71(EV71)VP1基因,进行抗血清的制备并检测抗血清效价。
SYBR Green Ⅰ实时荧光定量RT-PCR测定肠道病毒71型(EV71)RNA拷贝数方法的建立
SYBR Green Ⅰ实时荧光定量RT-PCR测定肠道病毒71型(EV71)RNA拷贝数方法的建立朵建英;王卫;丛喆;刘强;魏强【摘要】目的建立SYBR Green Ⅰ荧光染料实时定量RT-PCR方法,测定实验动物等来源的EV71病毒RNA.方法运用EV71 VP1保守区引物,优化real time RT-PCR条件,运用NASBA方法扩增EV71病毒RNA,计算拷贝数,经10倍系列稀释做出标准曲线,作为EV71病毒RNA定量检测的外标准品.结果应用Qiagen公司QuantiTect SYBR Green RT-PCR Kit,该标准品可精确定量到100copies/μL,PCR扩增效率达到99.5%.结论SYBR GreenⅠ荧光染料实时定量PCR 法测定EV71病毒RNA拷贝数的方法敏感性高、稳定性好,可用于EV71病毒RNA载量的定量测定.【期刊名称】《中国比较医学杂志》【年(卷),期】2010(020)007【总页数】5页(P27-31)【关键词】肠道病毒71型(EV71);模型,动物;病毒载量;实时定量RT-PCR【作者】朵建英;王卫;丛喆;刘强;魏强【作者单位】中国医学科学院实验动物研究所,北京协和医学院比较医学中心,卫生部人类疾病比较医学重点实验室,北京,100021;中国医学科学院实验动物研究所,北京协和医学院比较医学中心,卫生部人类疾病比较医学重点实验室,北京,100021;中国医学科学院实验动物研究所,北京协和医学院比较医学中心,卫生部人类疾病比较医学重点实验室,北京,100021;中国医学科学院实验动物研究所,北京协和医学院比较医学中心,卫生部人类疾病比较医学重点实验室,北京,100021;中国医学科学院实验动物研究所,北京协和医学院比较医学中心,卫生部人类疾病比较医学重点实验室,北京,100021【正文语种】中文【中图分类】R373.33肠道病毒71型(Enterovirus 71,EV71)为手足口病(hand-foot-mouth disease,HFMD)主要病原体[1,2],属于小RNA病毒科,肠道病毒属成员,其基因组为约7408个核苷酸的单股正链RNA[3,4]。
瑞德西韦对感染肠道病毒71型的人横纹肌瘤细胞和ICR乳鼠的抗病毒活性
第47卷第6期2021年11月吉林大学学报(医学版)Journal of Jilin University(Medicine Edition)Vol.47No.6Nov.2021DOI:10.13481/j.1671‑587X.20210614瑞德西韦对感染肠道病毒71型的人横纹肌瘤细胞和ICR乳鼠的抗病毒活性任晓风1,闫赟政2,李微2,李月香2,肖军海2,曹瑞源2,李永刚1(1.锦州医科大学基础医学院病原生物学教研室,辽宁锦州121000;2.军事科学院军事医学研究院毒物药物研究所,北京100850)[摘要]目的目的:探讨瑞德西韦(RDV)在细胞与动物水平对肠道病毒71型(EV71)的抗病毒活性,并阐明其抗病毒作用机制。
方法方法:基于人横纹肌瘤(RD)细胞进行RDV抗肠道病毒活性评价,检测RDV对EV71、柯萨奇病毒A6型(CA6)、肠道病毒D68型(EVD68)和柯萨奇病毒16型(CA16)的半数毒性浓度(CC50)和半数有效浓度(EC50),计算选择指数(SI)。
将RD细胞分为细胞对照组(不处理)、病毒对照组和给药组。
病毒对照组RD细胞给予EV71病毒液,给药组RD细胞再给予不同浓度(0.005、0.015、0.046、0.137、0.410、1.230、3.700、11.110、33.330和100.000µmol·L-1)RDV。
72h后,采用CellTiter-Glo®Luminescent检测试剂盒测定各组RD细胞活性。
在抗EV71细胞活性评价实验中,将RD细胞分为给药组和病毒对照组,给药组RD细胞给予不同浓度(0.03、0.10、0.30、0.80和2.50µmol·L-1)RDV。
30h后,采用实时荧光定量PCR(RT-qPCR)法检测各组RD细胞中EV71RNA表达水平,采用Western blotting法和免疫荧光法检测各组RD细胞中EV71结构蛋白VP1表达量。
干扰性核糖核酸可防丙肝
干扰性核糖核酸可防丙肝
美国斯坦福大学最近在《自然》杂志上发表研究成果指出,通过植入特定基因孕育的新生代实验鼠,其体内自然产生的干扰性核糖核酸(RNAi),可有效防止细胞受到丙型肝炎病毒的感染。
相关研究还发现,类似的干扰性核糖核酸在动物实验中被证实可有效抑制艾滋病病毒、脊髓灰质炎病毒感染。
干扰性核糖核酸可望成为病毒“克星”的这一重大发现,在美国生物不及医学界引起轰动。
专家解释,干扰性核糖核酸普遍存在于人体细胞内,只是细胞内98%的核糖核酸都没有作用。
核糖核酸对特定基因序列的病毒的抑制作用是一项空前的重大发现。
干扰性核糖核酸的长度仅21~25个碱基对,且专一性强,只要目标对象所含其中一个小单位“不对”,干扰性核糖核酸就不会产生作用,这项“专一的特性”,对未来干扰性核糖核酸被用来设计成为新药,通过基因治疗送至人体内对抗病毒的研究相当有利。
过去研究基因的功能,包括是否为致病关键基因,多是通过基因技术去除该研究基因,再观察下一代实验鼠是否出现病症,动辄需要一、两年。
未来运用RNAi,则可以通过体外实验,在短短一、两周内,研究该RNAi是否可以阻断信息RNA的生成,避免产生致病蛋白质。
最新一期的《自然》杂志中的一篇研究论文指出,特定的小型干扰性核糖核酸在进入人体细胞后,可以有效防止脊髓灰质炎病毒
感染人体细胞。
另外一项研究则发现,小型干扰笥核糖核酸可以防止人体细胞感染艾滋病病毒。
2019第二届中国药品安全技术大会暨中国(日照)生物医药产业合作论坛
中国医药生物技术2019年10月第14卷第5期Chin Med Biotechnol, October 2019, V ol. 14, No. 5 463[26]Jheng JR, Wang SC, Jheng CR, et al. Enterovirus 71 inducesdsRNA/PKR-dependent cytoplasmic redistribution of GRP78/BiP to promote viral replication. Emerg Microbes Infect, 2016, 5:e23. [27]Jheng JR, Lin CY, Horng JT, et al. Inhibition of enterovirus 71 entryby transcription factor XBP1. Biochem Biophys Res Commun, 2012, 420(4):882-887.[28]V oskoboinik I, Whisstock JC, Trapani JA. Perforin and granzymes:function, dysfunction and human pathology. Nat Rev Immunol, 2015, 15(6):388-400.[29]Wang LC, Kao CM, Ling P, et al. CD4 T-cell-independent antibodyresponse reduces enterovirus 71 lethality in mice by decreasing tissue viral loads. Clin Dev Immunol, 2012, 2012:580696.[30]David P, Megger DA, Kaiser T, et al. The PD-1/PD-L1 pathwayaffects the expansion and function of cytotoxic CD8+ T cells during an acute retroviral infection. Front Immunol, 2019, 10:54.[31]Belizário JE, Neyra JM, Setúbal Destro Rodrigues MF. When and howNK cell-induced programmed cell death benefits immunological protection against intracellular pathogen infection. Innate Immun, 2018, 24(8):452-465.[32]Zhu K, Yang J, Luo K, et al. TLR3 Signaling in macrophages isindispensable for the protective immunity of invariant natural killer T cells against enterovirus 71 infection. PLoS Pathog, 2015, 11(1): e1004613. [33]Xie J, Jiao Y, Qiu Z, et al. Significant elevation of B cells at the acutestage in enterovirus 71-infected children with central nervous system involvement. Scand J Infect Dis, 2010, 42(11-12):931-935.[34]Yao C, Xi C, Hu K, et al. Inhibition of enterovirus 71 replication andviral 3C protease by quercetin. Virol J, 2018, 15(1):116.[35]Lin L, Qin Y, Wu H, et al. Pyrrolidine dithiocarbamate inhibitsenterovirus 71 replication by down-regulating ubiquitin-proteasome system. Virus Res, 2015, 195:207-216.[36]Yen MH, Huang CI, Lee MS, et al. Artemisia capillaris inhibitedenterovirus 71-induced cell injury by preventing viral internalization.Kaohsiung J Med Sci, 2018, 34(3):150-159.[37]Yue Y, Li Z, Li P, et al. Antiviral activity of a polysaccharide fromLaminaria japonica against enterovirus 71. Biomed Pharmacother, 2017, 96: 256-262.[38]Hsieh YJ, Yen MH, Chiang LC, et al. Gan-Lu-Siao-Du-yin, aprescription of traditional Chinese medicine, inhibited enterovirus 71 replication, translation, and virus-induced cell apoptosis. J Ethnopharmacol, 2016, 185:132-139.[39]Qin Y, Lin L, Chen Y, et al. Curcumin inhibits the replication ofenterovirus 71 in vitro. Acta Pharm Sin B, 2014, 4(4):284-294. [40]Lv X, Qiu M, Chen D, et al. Apigenin inhibits enterovirus 71replication through suppressing viral IRES activity and modulating cellular JNK pathway. Antiviral Res, 2014, 109:30-41.·协会之窗·2019第二届中国药品安全技术大会暨中国(日照)生物医药产业合作论坛成功举办2019 年8 月7 日,2019 第二届中国药品安全技术大会暨中国(日照)生物医药产业合作论坛开幕。
肠道病毒71型灭活疫苗
灭活疫苗仍然存在问题
1
2
3
EV71 免疫保护机制复杂,除体液免疫外, 细胞免疫也发挥着重要的作用。然而,灭活疫苗主要诱导的是体液免疫, 细胞免疫不够充分。
灭活疫苗生产和运输成本高昂。疫苗效力较高而疫苗价格较低的常规免疫才符合我国的成本效益 , 这可能会制约灭活疫苗的应用。
EV71 本身存在抗体依赖性增强感染现象,不恰当的疫苗接种有可能会增加重症病例的风险。
Enterovirustype71inactivatedvaccine
课程:生物技术制药
班级:2013生技一班
肠道病毒71型灭活疫苗
EV71图片
肠道病毒 71 型( enterovirus 71, EV71) 为单股正链 RNA病毒,是小 RNA 病 毒 科 肠 道 病 毒 属 中 的 重 要 成 员 之 一。EV71 基因组由 7400 多个核苷酸构成, 包含一个开放读码框,编码大约 2200 个氨基酸的多聚蛋白。该多聚蛋白可被进一步水解成 P1、 P2 和 P3 三个前体蛋白,P1 前体蛋白编码VP1、 VP2、 VP3 和 VP4 四种结构蛋白,P2、 P3 前体蛋白编码 7种非结构蛋白( 2A ~ 2C 和 3A ~ 3D) EV71 包括 A、 B 和C 3 种基因型,11 个基因亚型, 每种亚型感染均可引起手足口病( hand, foot and mouth disease, HFMD
EV71 疫苗除能诱导中和抗体外,还能诱导细胞免疫应答,其也具有免疫保护作用。
EV71疫苗可行性
01
02
03
04
全病毒灭活疫苗
减毒活疫苗
基因工程疫苗
病毒样颗粒( VLPs)疫苗
肠道病毒 71 型疫苗分类
柯萨奇病毒A6型致手足口病研究进展
.340 .国际儿科学杂志2021年5月第48卷第5期Int J Pediatr,May 2021,Vol. 48,No. 5•综述•柯萨奇病毒A6型致手足口病研究进展夏晓\综述)王翠玲2(审校)1山西医科大学第一医院儿科,太原0300012山西医科大学第一医院预防保健科,太原030001通信作者:王翠玲,Email:139****9081@163. com【摘要】手足口病(hand,foot an d m outh disease,HFM D)系指由肠道病毒所致的传染病,中柯萨奇病毒A16(coxsackievirus A16,CA16)和肠道病毒71 (enterovirus 71,E V71)最常见。
近年来,有研究发现HFMD的肠道病毒病原谱发生了改变,柯萨奇病毒A6(c〇X sackievirus A6,C oxA6)新谱系逐渐成为优势株,在欧洲、亚太地区及北美爆发流行。
C〇X A6相关_D的爆发流行对HFMD的防控提出了新的挑战。
因此,该文对CoxA6的病原学、流行病学、临床特征、实验室诊断、预防措施及疫苗研究等作一综述,分析了HFMD相关CoxA6菌株的基因型与亚型,以期为HFMD的防控提供新的线索。
【关键词】肠道病毒;科萨奇病毒A6;手足口病D O I:10.3760/cma.j.issn.1673-4408. 2021.05. 013Hand,foot and mouth disease caused by Coxsackievirus A6Xia Xiao1, Wang Cuiling21 Department o f Pediatrics, the First Hospital of Shanxi Medical University, Taiyuan 030001, China2Department of Preventive Health Care, the First Hospital of Shanxi Medical University,Taiyuan030001, ChinaCorresponding author:Wang Cuiling}Email:139****************【Abstract】Hand,foot and mouth disease (HFMD) refers to infectious diseases caused by enteroviruses, of which coxsackievirus A16 (CA16) and enterovirus 71 (EV71) are the most common. In recentyears,studies have found that the enterovirus pathogen spectrum of HFMD has changed. The new lineage ofcoxsackievirus A6 (CoxA6) has gradually become a dominant strain, and it has spread in Europe, Asia Pacific and North America. The outbreak of CoxA6 related HFMD poses new challenges to the prevention and controlof HFMD. Therefore, this article reviews the etiology, epidemiology, clinical features, laboratory diagnosis,preventive measures and vaccine research of CoxA6, and analyzes the genotypes and subtypes of HFMD-relatedCoxA6 strains, in order to provide new clues for the prevention and control of HFMD.【Key words】Enterovirus; Coxsackievirus A6;Hand, foot and mouth diseaseDOI:10. 3760/cma. j. issn. 1673-4408.2021.05.013手足口病(hand,foot an d m ou th disease, HFMD)是由多种病毒引起的丙类传染病,主要感染 低龄儿童,呈自限性,一般持续7 ~ 10 d。
不同酶消化法提取猪原代肝细胞的效果比较
532024.4·试验研究0 引言猪圆环病毒(PCV )是Circoviridae 科Circovirus 属的一种无囊膜的单链环状DNA 病毒。
在已知的4个血清型中,PCV2为猪易感的致病性病毒[1]。
PCV2感染会诱导宿主免疫抑制引起猪圆环病毒病(PCVD ),包括断奶仔猪多系统衰竭综合征、新生仔猪先天性脑震颤、皮炎与肾病综合征、猪呼吸道病综合征、母猪繁殖障碍等,给全世界养猪业带来较大的经济损失,是世界各国的兽医与养猪业者公认的造成重大影响的猪传染病[2]。
PCV2的感染在猪生长发育的不同阶段有不同的组织嗜性。
但无论是胎儿阶段还是出生后,肝细胞都是PCV2感染和复制的靶细胞。
因此,PCV2也被视为一种能够诱导猪肝炎的病毒[3]。
且PCV2诱导的肝细胞凋亡在PCV2引发的相关病变和疾病的发病机制中具有关键性作用[4]。
因此,方便、快捷地获取大量有活性的猪肝细胞对于研究PCVD 的致病机制具有重大意义。
目前获取肝细胞常用的方法主要包括机械分离细胞法、非酶分离细胞法、离体酶消化法和酶灌流法等[5]。
因此,本试验采用简便、经济、无需特殊设备、仅需部分肝组织的离体酶消化法,比较不同酶消化分离猪原代肝细胞的效果,为一般实验室提取分离大量有活性的猪肝细胞提供参考。
1 材料与方法1.1 材料1.1.1 主要试剂新鲜猪肝组织,Hank's 平衡盐溶液(HBSS ),磷酸盐缓冲液(无菌PBS ),4%多聚甲醛(PFA ),收稿日期:2024-01-27基金项目:国家自然科学基金项目:复杂器官与组织在脾脏内的功能性再生(32230056)作者简介:周徐倩(1999-),女,汉族,浙江温州人,硕士在读,研究方向:组织工程与再生医学。
*通信作者简介:董磊(1978-),男,汉族,安徽阜阳人,博士,教授,研究方向:组织工程与再生医学、生物材料。
周徐倩,董磊.不同酶消化法提取猪原代肝细胞的效果比较[J].现代畜牧科技,2024,107(4):53-55. doi :10.19369/ki.2095-9737.2024.04.014. ZHOU Xuqian ,DONG Lei .Comparison of the Effect of Different Enzyme Digestion Methods on Extraction of Porcine Primary Hepatocytes[J].Modern Animal Husbandry Science & Technology ,2024,107(4):53-55.不同酶消化法提取猪原代肝细胞的效果比较周徐倩,董磊*(南京大学,江苏 南京 210023)摘要:猪肝细胞是猪圆环病毒的靶细胞,简单快速地提取猪原代肝细胞对于研究猪圆环病毒病的致病机制具有重要意义。
端粒酶反义寡聚脱氧核苷酸对人胃癌细胞的影响
端粒酶反义寡聚脱氧核苷酸对人胃癌细胞的影响
许兰涛;唐承薇
【期刊名称】《实用医学杂志》
【年(卷),期】2004(020)007
【摘要】目的:观察端粒酶反义寡聚脱氧核苷酸对人胃癌细胞株的作用.方法:用端粒酶反义寡聚脱氧核苷酸与人胃癌细胞株共同温育一定时间后,观察细胞形态变化,检测细胞DNA含量的分布,测定端粒酶活性.结果:端粒酶反义寡聚脱氧核苷酸能诱导人胃癌细胞凋亡,抑制端粒酶活性,在形态学上表现为细胞膜起泡、染色质固缩、核碎裂、凋亡小体形成;电泳呈凋亡特征性Ladder带;流式细胞仪分析显示,在G1期前出现亚2倍体凋亡峰:端粒酶活性抑制.结论:端粒酶反义寡聚脱氧核苷酸能诱导胃癌细胞凋亡,抑制端粒酶活性,抑制端合成,从而抑制胃癌细胞的增殖.
【总页数】3页(P725-727)
【作者】许兰涛;唐承薇
【作者单位】610041,成都市,四川大学华西医院消化内科;610041,成都市,四川大学华西医院消化内科
【正文语种】中文
【中图分类】R735.2
【相关文献】
1.端粒酶反义寡聚脱氧核苷酸诱导人胃癌细胞凋亡模型构建的研究 [J], 苏金龙;许兰涛
2.端粒酶反义寡聚脱氧核苷酸对人肝癌细胞株端粒酶活性作用的实验研究 [J], 许兰涛
3.端粒酶反义寡聚脱氧核苷酸对人胃癌细胞株的影响 [J], 王玉花;许兰涛
4.端粒酶反义寡聚脱氧核苷酸对MGC-803胃癌细胞的影响 [J], 孙大鹏;许德顺;韩冠英;罗喜刚
5.端粒酶反义寡聚脱氧核苷酸对子宫内膜癌细胞株的影响 [J], 杨蓉娟;许艳蕾;刘雪琴;魏丽莉
因版权原因,仅展示原文概要,查看原文内容请购买。
肠道病毒71型复制周期研究进展_陈生林 (1)
EV71的感染入侵,提 示 这 几 个 因 子 主 要 是 参 与 了 EV71对宿主细胞表 面 的 黏 附 过 程。 而 是 否 还 存 在 其他未知的功能性受体,以及现有的几种受体和因 子在 EV71入侵时是否及如何协同配合, 还有待 进 一步研究。
另外,EV71的衣壳蛋白 VP1可与宿主细胞表 面 的 波 形 蛋 白 结 合。Du 等 预 [10] 先 将 波 形 蛋 白 与 EV71共同孵育或用特 异 性 单 抗 封 闭 宿 主 细 胞 表 面
波形蛋白后,均可导致 EV71对宿主细胞的感染 性 下降;进一步定量实验结果显示,当细胞表面波形 蛋白减少时,吸附在宿主细胞表面的 EV71病毒颗 粒也显著减少。这些结果提示,波形蛋白极有可能 也是 EV71的吸附因子。
CHEN Sheng-lin,ZHU Yong-zhe,QI Zhong-tgy,Shanghai Key Laboratory of Medical Biodefense,
Second Military Medical University,Shanghai 200433,China Corresponding author:QI Zhong-tian,E-mail:qizt@smmu.edu.cn Abstract:Enterovirus 71 (EV71)is one of the primary pathogenic agents that cause hand,foot,and mouth disease (HFMD)mainly among young children.As the disease progresses,the virus may lead to severe neu- rological disorders with high mortality and disability rates.There are currently no effective prevention and treatment strategies to combat EV71infection.Investigating molecular mechanisms involved in EV71replica- tion cycle and cell infection could facilitate the progress for the design of new drugs and vaccines against this vi- rus.This article reviews the recent advances in the replication cycle of EV71. Key words:Enterovirus 71;Entry receptor;Translation;Replication;Assembly;Release
RNA干扰技术在肝星状细胞活化信号通路的应用
RNA干扰技术在肝星状细胞活化信号通路的应用燕晟;阳学风【摘要】肝纤维化是各种致病因子致肝细胞损伤后的修复过度.肝星状细胞(hepatic stellate cell,HSC)的激活被认为是肝纤维化的核心环节,实验证实多条信号传导通路参与HSC的激活.RNA干扰(RNA interference,RNAi)能有效沉默目的基因,以HSC活化信号通路为切入点,设计小干扰RNA(small interferingRNA,siRNA)抑制相关信号通路的活化,从而达到抑制HSC,激活是目前研究肝纤维化机制的主要手段.该研究综述了RNAi技术在研究HSC活化信号途径中的作用,以求为治疗肝纤维化提供一种新思路.【期刊名称】《中外医疗》【年(卷),期】2015(034)001【总页数】2页(P74-75)【关键词】肝纤维化;肝星状细胞;信号通路;RNA干扰技术【作者】燕晟;阳学风【作者单位】南华大学附属南华医院消化内科,湖南衡阳421001;南华大学附属南华医院消化内科,湖南衡阳421001【正文语种】中文【中图分类】R363肝纤维化是各种致病因子致肝细胞损伤后的修复过度,表现为肝结缔组织异常增生,可进一步发展为肝硬化甚至肝细胞癌,降低生活质量,出现并发症,严重危及生命。
HSC是形成肝纤维化的主要细胞,它的激活被认为是肝纤维化的核心环节。
多种信号传导通路与该激活过程的调控有关。
RNA干扰技术(RNA interference,RNAi)能特异性沉默目的基因,针对HSC活化信号途径中的分子设计RNAi,能逆转肝纤维化。
该研究着重阐述了肝星状细胞活化主要信号通路以及RNAi在这些信号通路的应用。
RNAi是通过沉默目标基因以研究基因和蛋白质功能的研究手段,被广泛的用于临床疾病研究。
作用的起始阶段正义链和反义链混合的双链(double-stranded RNA,dsRNA)在细胞内与核酸内切酶Ⅲ结合后,其被切割成siRNA。
益生菌对阿尔茨海默病作用的研究进展
益生菌对阿尔茨海默病作用的研究进展发布时间:2021-12-14T06:08:15.523Z 来源:《中国结合医学杂志》2021年12期作者:宋鑫萍1,2,李盛钰2,金清1[导读] 阿尔茨海默病已成为威胁全球老年人生命健康的主要疾病之一,患者数量逐年攀升,其护理的经济成本高,给全球经济造成重大挑战。
近年来研究显示,益生菌在适量使用时作为有益于宿主健康的微生物,在防治阿尔茨海默病方面具有积极影响,其作用机制可能通过调节肠道菌群,影响神经免疫系统,调控神经活性物质以及代谢产物,通过肠-脑轴影响该病发生和发展。
宋鑫萍1,2,李盛钰2,金清11.延边大学农学院,吉林延吉 1330022.吉林省农业科学院农产品加工研究所,吉林长春 130033摘要:阿尔茨海默病已成为威胁全球老年人生命健康的主要疾病之一,患者数量逐年攀升,其护理的经济成本高,给全球经济造成重大挑战。
近年来研究显示,益生菌在适量使用时作为有益于宿主健康的微生物,在防治阿尔茨海默病方面具有积极影响,其作用机制可能通过调节肠道菌群,影响神经免疫系统,调控神经活性物质以及代谢产物,通过肠-脑轴影响该病发生和发展。
本文综述了近几年来国内外益生菌对阿尔茨海默病的作用进展,以及其预防和治疗阿尔茨海默病的潜在作用机制。
关键词:益生菌;阿尔茨海默病;肠道菌群;机制Recent Progress in Research on Probiotics Effect on Alzheimer’s DiseaseSONG Xinping1,2,LI Shengyu2,JI Qing1*(1.College of Agricultural, Yanbian University, Yanji 133002,China)(2.Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences, Chanchun 130033, China)Abstract:Alzheimer’s disease has become one of the major diseases threatening the life and health of the global elderly. The number of patients is increasing year by year, and the economic cost of nursing is high, which poses a major challenge to the global economy. In recent years, studies have shown that probiotics, as microorganisms beneficial to the health of the host, have a positive impact on the prevention and treatment of Alzheimer’s disease. Its mechanism may be through regulating intestinal flora, affecting the nervous immune system, regulating the neuroactive substances and metabolites, and affecting the occurrence and development of the disease through thegut- brain axis. This paper reviews the progress of probiotics on Alzheimer’s disease at home and abroad in recent years, as well as its potential mechanism of prevention and treatment.Key words:probiotics; Alzheimer’s disease; gut microbiota; mechanism阿尔茨海默病(Alzheimer’s disease, AD),系中枢神经系统退行性疾病,属于老年期痴呆常见类型,临床特征主要包括:记忆力减退、认知功能障碍、行为改变、焦虑和抑郁等。
PPT Enterovirus 71研究进展
疫苗的开发
灭活病毒疫苗的研制
据报道以氢氧化铝为佐剂的甲醛灭活的EV71病毒疫苗,已于 1975年用于预防在保加利亚发生的HFMD流行。
减毒活疫苗
具有免疫途径简单,成本低,肠黏膜免疫原性高等优点,在发 展中国家得到了广泛的应用。具有免疫学效果,但其所引起的轻微 神经系统症状和在脊髓中可以分离出病毒表明其减毒并不完全,需 要进一步降低病毒的毒力。
EV71的致病机制
1 受体 病毒感染首先要通过细胞受体来完成黏附到易感细胞表面这一初始过程。病毒 的特异性受体不仅决定病毒的组织嗜性,而且在诱导细胞发生内吞的过程中扮 演着重要角色。EV71通过一组特异的受体进入宿主细胞。目前发现的EV71受 体主要有清道夫受体B2(SCARB2)、P选择素糖蛋白受体一1(PSGL一1/CDl62)、 唾液酸聚糖和DC—SIGN(CD209)。 2 天然免疫 证据表明EV71病毒感染诱发中枢神经系统炎症反应与补体激活有关。有研究发 现Ewl病毒感染人神经细胞SF268后,C3aRl、Cl qR、DAF、C4a等补体分子 表达明显升高。EV71感染也可以通过旁路活化系统产生C5a而刺激肺巨噬细胞 释放小分子趋化因子,诱导大量的多形核中性粒细胞聚集活化,导致组织损伤。 补体不仅广泛参与机体抗微生物防御反应以及免疫调节,在免疫病理学过程中 又是引起炎症等组织损伤的因子。 3 获得免疫 EV71感染后,胸腺细胞会产生IL-4和IL-2,这表明r11}11和Th2被激活。因 此,抗体和细胞介导的免疫反应在EV71感染后会被激活。 4 细胞因子 IL-6在EV71感染过程中起到保护机体的作用。IFN一1可直接激活其他免疫细胞, 在天然免疫和获得免疫中起重要作用。IFN一1可通过调节其他细胞因子的分泌 来抑制病毒复制,加速IL-6在单核细胞中的转录和复制,因此,病毒感染介导的 IFN。1分泌可加强IL-6的产生。
抗EV71多克隆抗体偶联的靶向磁性纳米颗粒对病毒的特异性富集
抗EV71多克隆抗体偶联的靶向磁性纳米颗粒对病毒的特异性富集霍美俊1,2*,张长清2,3*,闫 妍1,2,赵凌云1,貌盼勇4,唐劲天1(1. 清华大学工程物理系;粒子技术与辐射成像教育部重点实验室,北京100084 2北京中医药大学中药学院生物制药系,北京100102 3上海医药工业研究院,上海 2000401 4解放军302医院传染病研究所病毒研究室, 北京 100039)摘要 手足口病是幼儿和儿童常见的一种传染病,严重时会引起患者死亡。
EV71病毒(人肠道病毒71型)是引起手足口病的一种主要病毒。
为寻求一条快速诊断EV71病毒感染的新方法,将兔抗EV71多克隆抗体,通过戊二醛交联法偶联在氨基硅烷修饰的磁性纳米颗粒表面,获得抗EV71多克隆抗体偶联的靶向磁性纳米颗粒用于EV71病毒的检测。
通过酶联免疫、免疫荧光方法证实了抗体耦合在磁性纳米颗粒表面,并通过BCA法测得其耦合的效率为94.1%。
采用抗体偶联的磁性纳米颗粒对EV71病毒液中的病毒抗原进行吸附,通过ELISA法检测上清液中病毒含量的变化,并对磁性纳米颗粒表面吸附的病毒进行免疫荧光和核酸PCR检测,证明了抗体偶联的磁性纳米颗粒可以与病毒抗原特异性结合。
由于该纳米颗粒同时具有抗体的靶向性和磁性颗粒的磁响应性,对病毒抗原有较好的特异性吸附能力,可以用于低浓度大样本的EV71病毒抗原的富集检测。
利用抗体偶联的靶向磁性纳米颗粒同时具有可在病毒感染的细胞周围特异性富集及磁颗粒可在交变磁场下感应升温的双重功能,将其作为磁感应热疗的靶向介质,有望研制出病毒感染性疾病磁感应热疗的靶向介质,为靶向热疗病毒感染性疾病提供新的尝试。
关键词 EV71;抗体偶联;磁性纳米颗粒;病毒检测; 磁感应热疗Specific Enrichment of Viral Antigen with EV71 Polyclonal Antibody-coupledMagnetic Particles(HUO Meijun1, 2*, ZHANG Changqing2, 3*,YAN Yan1, 2, ZHAO Lingyun1, MAO Panyong4, SHENG Jun5, TANG Jintian1)(1. Department of Engineering Physics, Tsinghua University; Key Laboratory of Particle & Radiation Imaging, Ministry of Education, Beijing 100084, China 2. Department of Biopharmaceuticals, Beijing University of Chinese Medicine, Beijing 100102, 3. Shanghai Institute of Pharmaceutical Industry, Shanghai 200040, China 4. Department of Virology, Institute of infectious Diseases, 302 Hospital of PLA , Beijing 100039, China 5. Changchun Institute of Biological Products, Changchun 130062,)Abstract Hand, foot, and mouth disease (HFMD) is a common viral illness of infants and children. The disease causes fever and blister-like eruptions in the mouth and/or a skin rash. Enterovirus 71 (EV71) is notable as one of the major causative agents for HFMD, and is sometimes associated with severe central nervous system diseases. In this study, a novel method for specific作者简介:霍美俊,硕士研究生,研究方向为生物医学工程,电子信箱:huomeijun86@;张长清,硕士研究生,研究方向为生物医学工程,电子信箱:zhchq2933@;唐劲天(通信作者),教授,研究方向为肿瘤学,电子信箱:tangjt@;*为共同第一作者enrichment of viral antigen by EV-71 polyclonal antibody-coupled magnetic particles was developed. Targeted magnetic nanoparticles for specific recognition and enrichment of EV71 antigen were prepared by modification the polyclonal antibody of EV71 onto the surface of amino-silane coated magnetic nanoparticles (MNPs) through glutaraldehyde cross-linking. Antiserum of EV71 was generated by immunization of rabbits with purified EV71 antigen. Next, the serum was precipitated by saturated ammonium sulfate and then was ligated on the surface of Amino-silane-modified magnetic particles though glutaraldehyde cross-linking. Thus, targeted magnetic medium was prepared for specific enrichment of EV71. Both ELISA and immuno-fluorescence technique were applied to confirm the successful coupling of EV71 polyclonal antibody onto the surface of MNPs, and the coupling efficacy was as high as 94.1%. Specific enrichment of the EV71 antigen by the above-mentioned tareted MNPs was studied by immunofluorescence and PCR. Except for the specific enrichment of the virus antigen, possessing the inductive heating property under Alternative Magnetic Field (AMF), the antibody coupled MNPs can be further applied as agent of the Magnetic Mediated Hyperthermia (MMH) for targeted treatment of infective disease.Keywords EV71; magnetic particles; virus detection1 引 言病毒是许多疾病的病原微生物,严重威胁着人类健康。
- 1、下载文档前请自行甄别文档内容的完整性,平台不提供额外的编辑、内容补充、找答案等附加服务。
- 2、"仅部分预览"的文档,不可在线预览部分如存在完整性等问题,可反馈申请退款(可完整预览的文档不适用该条件!)。
- 3、如文档侵犯您的权益,请联系客服反馈,我们会尽快为您处理(人工客服工作时间:9:00-18:30)。
Published Ahead of Print 6 August 2014. 2014, 88(20):11915. DOI: 10.1128/JVI.01207-14. J. Virol. Bo Zhangand Bao-Di Shang, Peng Gong, Sylvie Alonso, Pei-Yong Shi Cheng-Lin Deng, Huimin Yeo, Han-Qing Ye, Si-Qing Liu,Analog NITD008Inhibition of Enterovirus 71 by Adenosine /content/88/20/11915Updated information and services can be found at: These include:REFERENCES/content/88/20/11915#ref-list-1at: This article cites 44 articles, 28 of which can be accessed free CONTENT ALERTSmore»articles cite this article), Receive: RSS Feeds, eTOCs, free email alerts (when new /site/misc/reprints.xhtml Information about commercial reprint orders: /site/subscriptions/To subscribe to to another ASM Journal go to: on October 31, 2014 by Tsinghua University/Downloaded fromInhibition of Enterovirus71by Adenosine Analog NITD008Cheng-Lin Deng,a,b Huimin Yeo,c Han-Qing Ye,a Si-Qing Liu,a,b Bao-Di Shang,a Peng Gong,a Sylvie Alonso,c Pei-Yong Shi,dBo Zhang a,bCAS Key Laboratory of Special Pathogens and Biosafety,Center for Emerging Infectious Diseases,Wuhan Institute of Virology,Chinese Academy of Sciences,Wuhan, China a;Key Laboratory of Agricultural and Environmental Microbiology,Wuhan Institute of Virology,Chinese Academy of Sciences,Wuhan,China b;Department of Microbiology,Yong Loo Lin School of Medicine,Immunology Programme,Life Sciences Institute,National University of Singapore,Singapore c;Novartis Institute for Tropical Diseases,Singapore dABSTRACTEnterovirus71(EV71)is a major viral pathogen in China and Southeast Asia.There is no clinically approved vaccine or antiviral therapy for EV71infection.NITD008,an adenosine analog,is an inhibitor offlavivirus that blocks viral RNA synthesis.Here we report that NITD008has potent antiviral activity against EV71.In cell culture,the compound inhibits EV71at a50%effective concentration of0.67M and a50%cytotoxic concentration of119.97M.When administered at5mg/kg in an EV71mouse model,the compound reduced viral loads in various organs and completely prevented clinical symptoms and death.To study the antiviral mechanism and drug resistance,we selected escape mutant viruses by culturing EV71with increasing concentrations of NITD008.Resistance mutations were reproducibly mapped to the viral3A and3D polymerase regions.Resistance analysis with recombinant viruses demonstrated that either a3A or a3D mutation alone could lead to resistance to NITD008.A combination of both3A and3D mutations conferred higher resistance,suggesting a collaborative interplay between the3A and3D proteins during viral replication.The resistance results underline the importance of combination therapy required for EV71treatment. IMPORTANCEHuman enterovirus71(EV71)has emerged as a major cause of viral encephalitis in children worldwide,especially in the Asia-Pacific region.Vaccines and antivirals are urgently needed to prevent and treat EV71infections.In this study,we report the in vitro and in vivo efficacy of NITD008(an adenosine analog)as an inhibitor of EV71.The efficacy results validated the potential of nucleoside analogs as antiviral drugs for EV71infections.Mechanistically,we showed that mutations in the viral3A and3D polymerases alone or in combination could confer resistance to NITD008.The resistance results suggest an intrinsic interaction between viral proteins3A and3D during replication,as well as the importance of combination therapy for the treatment of EV71 infections.E nterovirus71(EV71),a member of genus Enterovirus in thefamily Picornaviridae,is one of the major causative agents of diarrhea,rashes,and hand,foot,and mouth disease(HFMD)in children.Besides EV71,HFMD could also be caused by coxsacki-evirus A16(CV-A16)infection(1).Infection with EV71is some-times associated with severe central nervous system diseases(2). Since itsfirst isolation in1969(3),EV71outbreaks have occurred frequently in western Pacific region countries,including China, Japan,Malaysia,and Singapore.In China alone,EV71caused over 1million infections and hundreds of deaths each year from2009 to2011(/tjsj/fdcrbbg/).No effective anti-EV71drug is available,although several clinical trials of inacti-vated EV71vaccine have recently been completed(4,5).In a phase III trial,an inactivated alum adjuvant EV71vaccine showed effi-cacies of90%against EV71-associated HFMD and80.4%against EV71-associated diseases(including HFMD,herpangina,menin-gitis or encephalitis,febrile illness,viral exanthema,and respira-tory infection)(4).Despite the promising progress in vaccine de-velopment,antiviral therapy should still be pursued as a complementary means of intervention against EV71infection.EV71is a nonenveloped,single-positive-strand RNA virus. The genomic RNA is approximately7,000nucleotides in length and contains a single,long open reading frame(ORF)that is flanked by the5=untranslated region(UTR)and the3=UTR with a poly(A)tail.VPg(3B)is covalently linked to the5=end of the viral genome and involved in the initiation of viral RNA replica-tion(6).The5=UTR consists of two functional domains,a clover-leaf and an internal ribosome entry site,which contribute to viral replication and translation,respectively.The ORF encodes a single polyprotein that is initially processed by viral proteases into three precursor proteins,P1,P2,and P3(7).Precursor P1is further cleaved to yield structural proteins VP0,VP3,and VP1;and VP0is finally cleaved into VP2and VP4during virion formation.P2and P3are proteolytically processed into various nonstructural pro-teins associated with viral replication,such as2A pro,2BC,2B,2C, 3AB,3A,3B(VPg),3CD,3C,and3D(8,9).Following viral pro-tein synthesis,the genomic RNA serves as a template for negative-strand RNA synthesis,generating the intermediate double-stranded replicative-form(RF)RNA.The double-stranded RF RNA is then used as a template to synthesize more positive-strand Received29April2014Accepted30July2014Published ahead of print6August2014Editor:S.PerlmanAddress correspondence to Bo Zhang,zhangbo@,or Pei-Yong Shi,pei_yong.shi@.S.A.,P.-Y.S.,and B.Z.are co-senior authors of this article.Copyright©2014,American Society for Microbiology.All Rights Reserved.doi:10.1128/JVI.01207-14October2014Volume88Number20Journal of Virology p.11915–11915 on October 31, 2014 by Tsinghua University / Downloaded fromRNA.The nascent positive-strand RNA is encapsidated.Viral par-ticles are released from infected cells(10).Inhibitors have been reported for distinct targets of EV71and its closely related rhinovirus,as summarized in a recent review (11).Pleconaril,picodavir,and BPROZ-194were shown to target viral capsid protein VP1(12–14).Rupintrivir was found to inhibit the3C proteases of both human rhinovirus and EV71(15,16). Nucleoside analogs(such as2=-C-methylcystidine,N-6-modified purine,and ribavirin)have been reported to inhibit the viral poly-merase of EV71(17–20).None of these inhibitors has been suc-cessfully developed into a clinically approved drug.Therefore, new chemical entities are urgently needed for EV71antiviral de-velopment.NITD008is an adenosine nucleoside analog that has been re-ported to selectively inhibit viruses in the family Flaviviridae(21). Although NITD008showed efficacy in a dengue virus mouse model,it was not advanced to clinics because of the adversefind-ings of a preclinical toxicity study(21).Here we report that NITD008potently inhibits EV71in cell culture and in a mouse model,demonstrating that this compound could potentially be developed for EV71therapy.Mechanistically,we characterized the profile of EV71resistance to the compound.The resistance results showed that mutations in the viral3A and3D polymerase regions could confer resistance to NITD008,suggesting intimate cross talk between3A and3D during viral replication. MATERIALS AND METHODSViruses,cells,and compound.Vero(African green monkey kidney)cells were cultured in Dulbecco modified Eagle medium(DMEM;Invitrogen) with10%fetal bovine serum(FBS),100U/ml of penicillin,and100g/ml of streptomycin.The wild-type(WT)recombinant viruses were prepared from the infectious cDNA clone of EV71(22),stored as aliquots atϪ80°C, and used for antiviral activity assays and resistant virus selection.EV71 strain5865/SIN/000009(S41)(23)was used for an animal study. NITD008was synthesized as reported previously(21).Antiviral activity assay.Vero cells were seeded into12-well plates (1ϫ105/well),incubated for24h,and then incubated with viruses at a multiplicity of infection(MOI)of0.1PFU/cell for1h in DMEM with10% FBS.After the cells were washed once with culture medium,a different concentration of NITD008was added to each well.Cell cultures were collected at48h postinfection(p.i.),and viral titers in supernatants were quantified by plaque assay.The incubations for each concentration treat-ment were performed in triplicate.Cytotoxicity assay.A3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide(MTT)assay kit(a tetrazolium colorimetric assay; American Type Culture Collection)was used to determine the cytotoxic-ity of NITD008for Vero cells.We seeded Vero cells at1ϫ104per well(in 100l DMEM plus10%FBS)of a96-well plate,incubated them for6h, and then treated them with different concentrations of NITD008.After48 h of incubation,10l of MTT reagent was added and the plate was incu-bated for3.5h.Then100l of detergent reagents was added,and the plate was swirled gently and left in darkness at room temperature for4h.The absorbance of each well was measured with a microplate reader(Varios-kan Flash;Thermo Fisher)with a550-nmfilter.The cytotoxicity of NITD008was estimated by comparing the absorbance of the compound-treated cells with that of mock-treated cells.Generation of NITD008-resistant viruses.NITD008-resistant vi-ruses were selected by passaging WT EV71in Vero cells with increasing concentrations of NITD008.Two independent selections were carried out.For thefirst round of selection,Vero cells were infected with WT EV71at an MOI of0.1PFU/cell for1h,and then the infected cells were washed once and incubated with1M NITD008for another48h.The supernatants(designated P1)were collected and stored atϪ80°C.Viral titers from each selection were determined by plaque assay.The following selections were performed as described above.Different concentrations of NITD008are indicated for different passages(see Fig.2A).Sequencing of drug-resistant viruses.To identify mutations respon-sible for drug resistance,RNAs of selected viruses were extracted from virions in culture medium with RNeasy kits(Qiagen)according to the manufacturer’s instructions and amplified by reverse transcription(RT)-PCR with the SuperScript III one-step RT-PCR kit(Invitrogen).The com-plete viral genome was covered by four RT-PCRs with four pairs of prim-ers(22).The RT-PCR products were reclaimed by gel purification and sequenced.Preparation of recombinant EV71containing resistance mutations. EV71full-length cDNA clones with different mutations were constructed with the pACYC-EV71-FL infectious clone as the template(22).All mu-tations were introduced into the infectious clone by fusion PCR.Specifi-cally,fusion PCR products containing mutations within the2B/3A and 3D regions were introduced into pACYC-EV71-FL at the AatII-BsrGI and BsrGI-HindIII sites,respectively.All constructs were confirmed by DNA sequencing.To make recombinant viruses,WT and mutant genomic RNAs were in vitro transcribed from the HindIII-linearized cDNA plasmids with the MEGAscript T7kit(Ambion)according to the manufacturer’s protocols.The RNAs were transfected into Vero cells with DMRIE-C(Invitrogen).After transfection,supernatants were collected at different time points.The culture medium containing viruses was ali-quoted and stored atϪ80°C.Ethics statement.The animal experiments described here were ap-proved by the National University of Singapore Institutional Animal Care and Use Committee(protocol070/10).Nonterminal procedures were performed while animal were under anesthesia,and all efforts were made to minimize suffering.Mouse infection and drug treatment.Two-week-old AG129mice (B&K Universal)were bred and housed under specific-pathogen-free conditions in individual ventilated cages.Mice(nϭ8to10)were inocu-lated intraperitoneally(i.p.)with5865/SIN/000009(S41)(23)at a con-centration of107PFU/mouse as reported previously(24).Mice were treated orally with NITD008(6N HCl[1.5eq],1N NaOH[pH adjust-ment to3.5],and100mM citrate buffer[pH3.5]at a ratio of0.9:2.5:96.6) at a concentration of5mg/kg twice a day(b.i.d.)from the day of infection and for3consecutive days.Two control groups were included in the study design;one group was left untreated after infection,while the other was given the vehicle alone.Mice were monitored daily for20days for clinical illness and death.Clinical disease was scored as follows:0,healthy;1, ruffled hair and hunchbacked appearance;2,limb weakness;3,paralysis in one limb;4,paralysis in two limbs;5,death.To minimize animal suf-fering,mice were euthanized if they were paralyzed in two limbs.Determination of viral titers in organs from infected mice.Follow-ing systemic perfusion with50ml of sterile phosphate-buffered saline (PBS),organs and tissues were harvested from untreated infected mice and infected mice treated with NITD008or the vehicle at three different time points(days4,5,and6)p.i.and weighed.Samples were homoge-nized in DMEM,and the resulting homogenates were centrifuged at 14,000rpm for10min.Clarified supernatants werefiltered through a 0.22-m syringefilter(Millipore)before determination of the viral titer by plaque assay.Viral plaque assay.RD cells(ATCC CCL-136)were seeded into the wells of24-well plates(Nunc)at a density of105/well.The growth me-dium was removed,and the cells were infected with200-l volumes of suspensions containing10-fold serial virus dilutions ranging from10Ϫ1to 10Ϫ8.Infection was carried out for2h at37°C and5%CO2with rocking at half-hour intervals.One milliliter of1.2%Avicel in minimum essential medium(FMC BioPolymer)was added,and the cells were further incu-bated for3days.After the overlay medium was decanted,the cells were washed with PBS andfixed with4%paraformaldehyde.Crystal violet was used tofix and stain the cells.Plaques were then quantified via visual scoring.Deng et al. Journal of Virology on October 31, 2014 by Tsinghua University / Downloaded fromStatistical analyses.Differences between data on antiviral activity in cell culture were statistically evaluated with the open-source R-project software(Foundation for Statistical Computing,Vienna,Austria;http: ///).Antiviral activity was assessed with the Kruskal-Wallis rank sum test.Statistical analysis of the animal data was performed with GraphPad Prism,version5(GraphPad4Software).Data are ex-pressed as meansϮthe standard error of the means(SEM).Kaplan-Meier survival curves and clinical-score curves were analyzed with a log rank test and the Wilcoxon test,respectively.A two-tailed P value ofϽ0.05was considered statistically significant.RESULTSAntiviral activity of NITD008against EV71replication in cellculture.To facilitate antiviral activity discovery,we previouslydeveloped a stable EV71reporter virus containing an enhancedgreenfluorescent protein(eGFP)-encoding gene(eGFP-EV71)that could be used for high-throughput screening of inhibitors ofEV71(22).We tested NITD008in Vero cells infected with eGFP-EV71.The compound exhibited dose-dependent inhibition ofeGFP expression in infected cells(Fig.1A);under a phase-contrastmicroscope,equal numbers of cells from wells treated with differ-ent concentrations of the compound were observed(data notshown).To confirm the results from the reporter virus,we exam-ined the compound’s efficacy with WT EV71in a viral titer reduc-tion assay.As shown in Fig.1B,NITD008inhibited the WT virusat an EC50(50%effective concentration)of0.67M.At a concen-tration of4M,NITD008reduced viral titers by up to1,000-fold.Since infection with both EV71and CV-A16can cause HFMD,we tested NITD008for activity against CV-A16.The compoundshowed an EC50of0.64M(Fig.1C).To exclude the possibilitythat the inhibition of viral replication was due to NITD008-medi-ated cytotoxicity,we performed a cell proliferation-based cytotox-icity assay.As shown in Fig.1D,the compound showed a CC50(50%cytotoxic concentration)of119.97M in Vero cells.Even at16M NITD008,the cells retained around90%viability.Theresults allowed us to conclude that NITD008is a potent inhibitorof EV71in cell culture,with an estimated therapeutic index(TI[CC50/EC50])of179.It has been previously shown that rupintrivir(AG7088)effi-ciently blocked EV71replication as a potent3C proteinase inhib-itor(16,25,26).Therefore,we evaluated the antiviral effects ofdifferent concentrations of NITD008with or without rupintrivirby the Gluc-EV71infection assay(Fig.1E).The Gluc-EV71infec-tion assay was performed by infecting cells with complete EV71containing the gene for Gaussia luciferase(Gluc),and Gluc activ-ity was quantified to monitor antiviral activity.Our results indi-cated that the luciferase activities that resulted from dual treat-ments(NITD008plus0.25M rupintrivir and NITD008plus0.5M rupintrivir)were lower than when NITD008was used alone (P values were0.02and0.004,respectively),which was also thecase for treatment with rupintrivir alone(P values were both0.03).It is suggested that the combination of rupintrivir andNITD008had a more inhibitory effect on EV71replication than either NITD008or rupintrivir alone.Selection of resistant viruses.NITD008-resistant viruses were selected by continuously culturing EV71for18rounds in the pres-ence of gradually increasing concentrations of NITD008(Fig.2A), i.e.,1M for eight passages,1.5M for one passage,2M for four passages,and2.5M forfive passages.Two independent selec-tions(SelA and SelB)were performed to test the reproducibility of the resistance results.For each passage,viral titers in the mock-treated infection and the NITD008-treated infection were deter-mined;the viruses were subjected to resistance analysis with a viral titer reduction assay.Figure2B and C show two representative data from such resistance assays for passage8(P8)and P18,re-spectively.At P8,the selected viruses exhibited marginal resistance to1M NITD008compared with the WT virus.In contrast,at P18,both the SelA and SelB viruses exhibited significant resistance to NITD008.Specifically,at a2.5M inhibitor concentration, SelA and SelB viral titers were reduced by approximately50-and FIG1Antiviral and cytotoxic analyses of NITD008.(A)Antiviral activities of NITD008were analyzed by the eGFP-EV71infection assay.Different concen-trations of NITD008were added to Vero cells infected with eGFP-EV71at an MOI of0.1PFU/cell.At48h p.i.,eGFP expression was observed under a fluorescence microscope.Antiviral activity was indicated by the reduction of the number of eGFP-positive cells upon NITD008treatment.(B)Antiviral activity of NITD008analyzed by WT EV71viral titer reduction assay.Different concentrations of NITD008were added to Vero cells after1h of incubation with EV71at an MOI of0.1PFU/cell.At48h p.i.,culture medium was col-lected and viral titers were measured with plaque assays.Antiviral activity was indicated by the reduction of viral titers upon NITD008treatment.(C)Anti-viral activities of NITD008against CV-A16.The antiviral activity assay was performed the same way as that with EV71.(D)Cytotoxicity was evaluated by incubation of Vero cells with the indicated concentration of NITD008.At48h posttreatment,the viability of treated cells was measured by an MTT assay and presented as a percentage of that of untreated cells(see details in Materials and Methods).(E)Inhibition of EV71replication by a combination of rupintrivir and NITD008.The antiviral activities of different concentrations of NITD008, alone or in combination with rupintrivir,against EV71were tested.Vero cells were infected with Gluc-EV71at an MOI of0.1PFU/cell.The construction of Gluc-EV71will be reported elsewhere.At48h p.i.,infected cells were collected for the luciferase assay.Antiviral activity was indicated by a reduction of lucif-erase activity.Statistical significance(PϽ0.01)was detected for the antiviral activity of four treatments.NITD008as an Inhibitor of Enterovirus7111917 on October 31, 2014 by Tsinghua University / Downloaded from10-fold,respectively,whereas WT viral titers were suppressed by almost1,000-fold.These results demonstrate that resistant EV71 virus can be reproducibly obtained in cell culture and that the SelB virus is more resistant than the SelA virus to NITD008inhibition.As shown in Fig.2B and C,in the absence of NITD008,the WT and selected resistant viruses grew to comparable titers.In agree-ment with this result,similar plaque morphologies of the WT, SelA,and SelB viruses were observed(Fig.2D).The results indi-cate that the mutations that occurred in the resistant viruses did not reduce the magnitudes of replication in virus-infected cell cultures.Identification of resistance mutations.To identify mutations responsible for drug resistance,we sequenced the complete ge-nomes of P18SelA and SelB viruses,as well as the P18WT virus. No mutation was found in the P18WT virus.In contrast,several sense and nonsense mutations were found in the P18SelA and SelB viruses(Fig.3A).The nonsense mutations occurred at VP2, VP3,3B,and3C.The sense mutations in the SelA and SelB viruses were located in three regions.(i)An Ala¡Val substitution at amino acid position38of the2B protein(A38V2B)was found in SelB;(ii)two mutations in the3A protein,M62T3A and V75A3A, were recovered from SelA and SelB,respectively;and(iii)a com-mon mutation,V63A3D,in the3D polymerase was recovered from both selection viruses,while an additional M393L3D mutation in the3D protein was recovered from SelB.A close examination of the sequence chromatograms showed that the two mutations from SelA—M62T3A and V63A3D—contained pure adaptive se-quences(Fig.3B),whereas the two mutations from SelB—V63A3D and M393L3D—had adaptive sequences in a mixture with the WT sequence(Fig.3C).In summary,all of the amino acid changes were mapped to the3A and3D regions of both resistant viruses,except that an additional A38V2B mutation was found in SelB.Mutations in both3A and3D confer resistance to NITD008. Using an infectious cDNA clone of EV71,we examined the roles of the mutations recovered from the selection isolates in drug resis-tance.Individual or combined mutations were engineered into the infectious cDNA clone to generate recombinant viruses.The resistance phenotype of each recombinant virus was analyzed by comparing the viral titers after the virus was cultured in the pres-ence or absence of NITD008.The WT and selected(SelA and SelB) P18viruses were included as controls.Figure4A shows the resis-tance analysis of mutations recovered from a SelA isolate.Muta-tion M62T3A or V63A3D alone was sufficient to confer resistance to a level close to that of the SelA isolate.The M62T3A V63A3D double mutation slightly improved the resistance to a level equiv-alent to that of the original SelA isolate.The results demonstrate that both M62T3A and V63A3D play a role in resistance.Figure4B summarizes the resistance analysis of mutations re-covered from a SelB isolate.A mutant virus containing A38V2B alone did not show any resistance,whereas viruses containing the single mutation V75A3A,V63A3D,or M393L3D showed partial re-sistance.The V75A3A V63A3D,V75A3A M393L3D,and V63A3D M393L3D double mutations slightly increased resistance in com-parison with that of the single mutant viruses.The V75A3A V63A3D M393L3D triple mutation further increased resistance to a level similar to that of the original SelB isolate.Addition of A38V2B to the double mutation V75A3A V63A3D or to the triple mutation V75A3A V63A3D M393L3D did not change the level of resistance. These results demonstrate that(i)the mutation in2B(A38V2B) does not contribute to resistance,(ii)the mutations in3A (V75A3A)and3D(V63A3D and M393L3D)confer resistance,and (iii)combining two and three mutations in3A and3D additively increases resistance.Furthermore,Fig.4A and B clearly indicate that three mutations in SelB(V75A3A V63A3D M393L3D)account for the higher resistance than the two mutations in SelA(M62T3A V63A3D).NITD008treatment completely protected EV71-infected mice from lethal disease.The in vivo efficacy of NITD008was assessed in an AG129mouse model of EV71infection(24).Two-week-old AG129mice were infected i.p.with107PFU of EV71 strain41(S41).On the day of infection and for3consecutive days, the animals were administered NITD008at5mg/kg orally twice daily.Control groups consisted of untreated infected animals and infected animals given the vehicle alone.The animals were ob-served daily for20days p.i.,and clinical manifestations were scored.When two-limb paralysis was observed,the animals were euthanized.Untreated infected mice and infected animals treated with the vehicle displayed a100%mortality rate,with some deaths observed as early as6days p.i.(Fig.5A).These mice displayed a characteristic progression of severity of clinical symptoms,includ-ing ruffled hair,a hunched back,limb weakness,and one-limb paralysis,followed by two-limb paralysis,at which point the ani-mals were euthanized(Fig.5B).Although there was a delay in the manifestation of clinical symptoms in the vehicle-treated infected mice compared to that of the untreated infected group,treatment with the vehicle alone did not result in significant protection,as evidenced by the100%mortality rate observed by day11p.i.(Fig. 5A).In contrast,mice treated with NITD008remained healthyFIG2Selection and characterization of NITD008-resistant EV71.(A)Scheme used for selection of NITD008-resistant EV71.The concentrations of NITD008corresponding to different passages during selection are indicated. (B,C)Characterization of resistance phenotypes of P8(B)and P18(C)EV71. The indicated concentrations of NITD008were added to Vero cells after1h of incubation with EV71at an MOI of0.1PFU/cell.The viral titers in culture fluids in the presence or absence of NITD008were determined at48h p.i.byplaque assay.Two independent selections were performed(SelA and SelB).(D)Plaque morphologies of WT and selected resistant viruses.Deng et al. Journal of Virology on October 31, 2014 by Tsinghua University / Downloaded fromthroughout the course of the experiment and did not manifest any clinical signs of illness associated with severe EV71infection(Fig. 5A and B).These data thus demonstrate the protective efficacy of NITD008in a mouse model of severe EV71infection.Viral titers in organs from drug-treated infected mice were significantly reduced compared to those of the control groups. To further assess the in vivo protective efficacy of NITD008,at4,5, and6days p.i.,the viral titers in the front limbs,hind limbs,spines, and brains harvested from mice infected with EV71and treated with NITD008were quantified.These viral titers were compared to those measured in the control groups(untreated infected mice and infected mice treated with the vehicle).Infectious viruses were detected in the untreated infected mice and infected mice treated with the vehicle in all of the organs assessed(Fig.6A to D)accord-ing to a typical kinetic whereby the viruses accumulatedfirst in the limb muscles before reaching the spine and eventually accumu-lated in the brain,as reported previously(24).Interestingly,the viral titers measured in the infected mice treated with the vehicle were significantly lower than those measured in the untreated in-fected mice,suggesting that oral administration of the vehicle alone affects the infectivity of the virus.These lower viral titers correlate with the delayed appearance of the clinical manifesta-tions observed in this mouse group(Fig.5A and B).However, since eventually all of the mice in the vehicle-treated group dis-played the full range of clinical manifestations with a100%mor-tality rate,we conclude that the vehicle-mediated reduction in viral titers is not sufficient to protect the animals from EV71in-fection effectively.In contrast,significantly lower virus titers in all of the organs assessed,including the central nervous system,were measured in infected mice treated with NITD008(Fig.6A to D).Compared to those of vehicle-treated mice,reductions of average virus titers in the organs of NITD008-treated mice ranged from2-fold to a com-plete absence of viral detection(order of magnitude of5)by plaque assay.The results thus strongly support the in vivo protec-tive efficacy of NITD008against EV71.DISCUSSIONThe increase in EV71-associated morbidity and mortality in hu-mans has underlined the urgent need to develop an effective therapy for EV71infection.In this study,we demonstrated the in vitro and in vivo efficacy of NITD008,an adenosine analog,as anFIG3Mutations identified in NITD008-resistant EV71.(A)Mutations recovered from resistant viruses were tabulated according to the amino acid positions and the corresponding plete-genome sequencing of P18viruses from SelA and SelB was performed.Sense and nonsense mutations are in red and black,respectively.(B,C)Sequence chromatograms of mutations from resistant viruses.Representative sequencing chromatograms are presented for SelA(B) and SelB(C)viruses for P18selection.Original mutations are indicated at the top of each panel.Amino acid changes and positions in the corresponding proteins are indicated.NITD008as an Inhibitor of Enterovirus71 11919 on October 31, 2014 by Tsinghua University / Downloaded from。