首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到19条相似文献,搜索用时 187 毫秒
1.
冠状病毒是有包膜的单股正链RNA病毒。作为人和动物的重要致病原,冠状病毒感染主要导致宿主呼吸系统、肝脏、胃肠道以及神经系统出现急性或慢性症状。2000年以来,传染性非典型肺炎和中东呼吸综合征的暴发,以及猪流行性腹泻病毒在全球猪群中的暴发流行,引起大家对动物冠状病毒的极大重视。S蛋白具有受体结合活性和膜融合活性,是冠状病毒感染细胞的关键蛋白;S蛋白在病毒的组织或宿主嗜性和毒力等方面发挥重要作用。本文重点对近年来冠状病毒S蛋白的结构、功能以及S蛋白与受体相互作用的研究进行综述,以期为冠状病毒的入侵机制和反向遗传学研究以及受体阻断药物的开发提供参考。  相似文献   

2.
猪δ冠状病毒(porcine deltacoronavirus,PDCoV)是目前新发现的唯一一种感染哺乳动物的δ冠状病毒。PDCoV主要感染猪的小肠,特别是空肠和回肠,造成小肠绒毛上皮细胞萎缩,引起严重的萎缩性肠炎,临床症状主要表现为新生仔猪水样腹泻、呕吐和脱水死亡,给养猪业造成很大的经济损失。2014年以来全球暴发的仔猪腹泻中,PDCoV单一感染检出率占有一定的比例,还与其他猪冠状病毒存在较高比例的共感染现象。随着PDCoV毒株的基因组测序完成和病毒的分离成功,以及病毒与宿主互作研究的推进,对该病毒有了更多的认知。本文根据现有的文献报道,结合本课题组的研究进展,对猪δ冠状病毒的流行、基因组结构的遗传多样性、病毒感染受体和对宿主先天免疫应答调控机制的研究进展进行了综述,以帮助相关人员对PDCoV有全面和深入的了解。  相似文献   

3.
猪肠道冠状病毒是引起仔猪腹泻的重要致病因素,主要感染小肠绒毛上皮细胞,给养猪业造成了巨大的经济损失。由于缺乏能模拟胃肠道高度复杂生理特性的体外研究模型,猪肠道冠状病毒感染与宿主肠上皮之间相互作用的研究也受到了极大的限制。随着干细胞技术的快速发展,一种能模拟肠道复杂的细胞类型及空间结构的体外模型——肠小体引起了人们的广泛关注。与传统的细胞系相比,肠小体不仅能模拟肠的结构和功能,同时还保留宿主的遗传特性,有望成为研究宿主-肠道病原相互作用的一种理想模型。本文就猪肠道冠状病毒以及肠小体在肠道病原研究中的应用进行综述,以期为猪肠道冠状病毒的基础研究提供新的思路与见解。  相似文献   

4.
猪丁型冠状病毒HB-BD株的分离与鉴定   总被引:1,自引:0,他引:1  
猪丁型冠状病毒(Porcine deltacoronavirus,PDCoV)是一种新的引起仔猪腹泻的冠状病毒,目前国内对PDCoV分离的研究较少。为从腹泻仔猪粪便及肠道内容物中分离鉴定PDCoV,本研究将RT-PCR检测为PDCoV病原阳性的腹泻样本,接种到ST细胞,进行病毒的分离传代。通过观察其细胞病变,RT-PCR和间接免疫荧光方法检测鉴定,并对分离株的S、M、N基因进行测序鉴定及序列分析。结果成功分离得到了PDCoV HB-BD株。经序列同源性分析发现,HB-BD分离株S、M和N基因与近年来国内外的流行毒株同源性很高,核苷酸同源性分别为95.8%~99.1%、98.6%~99.4%和97.8%~99.4%。进化树分析表明HB-BD分离株与中国毒株亲缘关系比其他国家近,处于同一分支。结果证实从腹泻仔猪的肠道内容物中分离并鉴定得到了一株猪丁型冠状病毒,本研究为后续分离株的致病性及生物学特性研究奠定了基础。  相似文献   

5.
冠状病毒感染有相对严格的宿主和组织特异性,其中部分病毒演化中会发生细胞嗜性改变。冠状病毒的跨宿主感染能力主要取决于病毒表面棘突蛋白的变异及其与受体相互作用的特异性改变。棘突蛋白的变异主要集中在受体结合域(RBD),其他区域也与病毒感染的宿主细胞特异性有关。另外,较大的RNA基因组、独特的套氏亚基因组转录、复制过程中模板转换引起的高频率基因重组等使冠状病毒不断出现毒力或宿主变异,而共感染和持续性感染则为病毒重组及跨宿主感染提供了机会。  相似文献   

6.
猪传染性胃肠炎(transmissible gastroenteritis,TGE)是由猪传染性胃肠炎病毒(transmissible gas-troenteritis virus,TGEV)引起的一种急性、高度接触性传染病,以呕吐、水样腹泻、脱水和对2周龄以内仔猪高度致死率为特征[1]。猪传染性胃肠炎病毒隶属于冠状病毒科冠状病毒属,是引起仔猪病毒性腹泻的重要病原,其基因组为单股正链的有感染性不分节段的RNA,TGEV结构蛋白主要由S、N、Ms、M蛋白组成[2]。其中n基因指导合成病毒的核衣壳蛋白(N),它是一种磷酸化的蛋白,存在于病毒粒子的内部,其分子质量为47kD[3],与病毒基因组组成核衣壳;N…  相似文献   

7.
8.
为研究猪氨基肽酶(Porcine Aminopeptidase N,pAPN)是否作为猪流行性腹泻病毒(Porcine epidemic diarrhea virus,PEDV)的细胞感染受体,通过转染技术,使PEDV非容许性细胞MDCK表达pAPN,并用PEDV感染转染细胞。结果发现转染的MDCK细胞可以感染PEDV,并且该病毒可以在转染细胞中连续传代。免疫荧光法鉴定存在病毒抗原。进一步实验证实,抗pAPN血清可以抑制PEDV感染转染的MDCK细胞。这些结果展示转染的MDCK细胞、pAPN表达及PEDV病毒复制之间存在直接联系,证明pAPN是PEDV的细胞感染受体之一。  相似文献   

9.
冠状病毒是一大类能够引起呼吸系统疾病,从而威胁人类健康的病毒.目前,对冠状病毒诱导细胞凋亡及其机制研究甚少.本研究以动物冠状病毒 猪流行性腹泻病毒(PEDV) 为模型探讨冠状病毒诱导细胞凋亡效应及其可能作用机制. 通过流式细胞术检测发现感染PEDV病毒后细胞凋亡率明显升高,且PEDV诱导细胞凋亡呈时间和剂量依赖性(P<0.05或P<0.01);进一步研究发现,冠状病毒木瓜样蛋白酶(PLP)在病毒引起凋亡过程中起重要作用.实验发现,转染PEDV-PLP质粒后,caspase-3活化体表达水平明显升高. 提示冠状病毒PLP蛋白酶通过激活caspase-3在病毒诱导细胞凋亡过程中起着关键作用. 以上结果为研究人类冠状病毒PLP蛋白功能及其通过细胞凋亡调节宿主抗病毒天然免疫机制提供重要基础.  相似文献   

10.
冠状病毒(Coronavirus)是具有包膜的正单链RNA病毒,基因组大小介于26 000与32 000 nt之间,编码刺突蛋白(S)、包膜蛋白(E)、膜蛋白(M)和核壳蛋白(N)等四种结构蛋白、复制酶(ORF1a/b)与若干辅助蛋白,部分病毒还具有血细胞凝集素酯酶(HE),这些蛋白除维持病毒结构,还有促进感染与抵抗宿主免疫反应等功能,其中刺突蛋白可与宿主细胞表面的受体结合,使病毒包膜和宿主细胞的膜融合以感染细胞.冠状病毒的感染会影响细胞的许多信号转导途径,引发免疫反应,是一类可感染哺乳动物与鸟类的病毒.  相似文献   

11.
The surface glycoprotein S of transmissible gastroenteritis virus (TGEV) has two binding activities. (i) Binding to porcine aminopeptidase N (pAPN) is essential for the initiation of infection. (ii) Binding to sialic acid residues on glycoproteins is dispensable for the infection of cultured cells but is required for enteropathogenicity. By comparing parental TGEV with mutant viruses deficient in the sialic acid binding activity, we determined the contributions of both binding activities to the attachment of TGEV to cultured cells. In the presence of a functional sialic acid binding activity, the amount of virus bound to two different porcine cell lines was increased sixfold compared to the binding of the mutant viruses. The attachment of parental virus was reduced to levels observed with the mutants when sialic acid containing inhibitors was present or when the cells were pretreated with neuraminidase. In virus overlay binding assays with immobilized cell surface proteins, the mutant virus only recognized pAPN. In addition, the parental virus bound to a high-molecular-mass sialoglycoprotein. The recognition of pAPN was sensitive to reducing conditions and was not dependent on sialic acid residues. On the other hand, binding to the sialic acid residues of the high-molecular-mass glycoprotein was observed regardless of whether the cellular proteins had been separated under reducing or nonreducing conditions. We propose that binding to a surface sialoglycoprotein is required for TGEV as a primary attachment site to initiate infection of intestinal cells. This concept is discussed in the context of other viruses that use two different receptors to infect cells.  相似文献   

12.
Transmissible gastroenteritis coronavirus (TGEV) is a porcine pathogen causing enteric infections that are lethal for suckling piglets. The enterotropism of TGEV is connected with the sialic acid binding activity of the viral surface protein S. Here we show that, among porcine intestinal brush border membrane proteins, TGEV recognizes a mucin-type glycoprotein designated MGP in a sialic acid-dependent fashion. Virus binding assays with cryosections of the small intestine from a suckling piglet revealed the binding of TGEV to mucin-producing goblet cells. A nonenteropathogenic mutant virus that lacked a sialic acid binding activity was unable to bind to MGP and to attach to goblet cells. Our results suggest a role of MGP in the enteropathogenicity of TGEV.  相似文献   

13.
D B Tresnan  R Levis    K V Holmes 《Journal of virology》1996,70(12):8669-8674
Two members of coronavirus serogroup I, human respiratory coronavirus HCV-229E and porcine transmissible gastroenteritis virus (TGEV), use aminopeptidase N (APN) as their cellular receptors. These viruses show marked species specificity in receptor utilization, as HCV-229E can utilize human but not porcine APN, while TGEV can utilize porcine but not human APN. To determine whether feline APN could serve as a receptor for two feline coronaviruses in serogroup I, feline infectious peritonitis virus (FIPV) and feline enteric coronavirus (FeCV), we cloned the cDNA encoding feline APN (fAPN) by PCR from cDNA isolated from a feline cell line and stably expressed it in FIPV- and FeCV-resistant mouse and hamster cells. The predicted amino acid sequence of fAPN shows 78 and 77% identity with human and porcine APN, respectively. When inoculated with either of two biologically different strains of FIPV or with FeCV, fAPN-transfected mouse and hamster cells became infected and viral antigens developed in the cytoplasm. Infectious FIPV was released from hamster cells stably transfected with fAPN. The fAPN-transfected mouse and hamster cells were challenged with other coronaviruses in serogroup I including canine coronavirus, porcine coronavirus TGEV, and human coronavirus HCV-229E. In addition to serving as a receptor for the feline coronaviruses, fAPN also served as a functional receptor for each of these serogroup I coronaviruses as shown by development of viral antigens in the cytoplasm of infected mouse or hamster cells stably transfected with fAPN. In contrast, fAPN did not serve as a functional receptor for mouse hepatitis virus (MHV-A59), which is in serogroup II and utilizes mouse biliary glycoprotein receptors unrelated to APN. Thus, fAPN serves as a receptor for a much broader range of group I coronaviruses than human and porcine APNs. The human, porcine, and canine coronaviruses in serogroup I that are able to use fAPN as a receptor have previously been shown to infect cats without causing disease. Therefore, host factors in addition to receptor specificity apparently affect the virulence and transmissibility of nonfeline serogroup I coronaviruses in the cat.  相似文献   

14.
Porcine deltacoronavirus (PDCoV), also known as porcine coronavirus HKU15, was first detected in North America in early 2014 and associated with enteric disease in pigs, resulting in an urgent need to further investigate the ecology of this virus. While assays detecting nucleic acids were implemented quickly, assays to detect anti-PDCoV antibodies have not been available. In this study, an indirect anti-PDCoV IgG enzyme-linked immunosorbent assay (ELISA) based on the putative S1 portion of the spike protein was developed and utilized to determine the prevalence of anti-PDCoV IgG in U.S. pigs. The diagnostic sensitivity of the PDCoV ELISA was 91% with a diagnostic specificity of 95%. A total of 968 serum samples were tested including samples with confirmed infection with PDCoV, porcine epidemic diarrhea virus (PEDV), transmissible gastroenteritis virus or porcine respiratory coronavirus. There was no cross-reactivity with any of the other coronaviruses. Among 355 arbitrarily selected serum samples collected in 2014 and originating from 51 farms across 18 U.S. states, anti-PDCoV IgG antibodies were detected in 8.7% of the samples and in 25.5% of the farms whereas anti-PEDV IgG was detected in 22.8% of the samples and in 54.9% of the farms. In addition, anti-PDCoV IgG antibodies were detected in archived samples collected in 2010, perhaps indicating an earlier undetected introduction into the U.S. pig population. Overall, the obtained data suggest that PDCoV seroprevalence in U.S. pigs is lower compared to PEDV and PDCoV may have been introduced to the U.S. prior to PEDV.  相似文献   

15.
The hemagglutinating activity of transmissible gastroenteritis virus (TGEV), an enteric porcine coronavirus, was analyzed and found to be dependent on the presence of alpha-2,3-linked sialic acid on the erythrocyte surface. N-Glycolylneuraminic acid was recognized more efficiently by TGEV than was N-acetylneuraminic acid. For an efficient hemagglutination reaction the virions had to be treated with sialidase. This result suggests that the sialic acid binding site is blocked by virus-associated competitive inhibitors. Porcine respiratory coronavirus (PRCV), which is serologically related to TGEV but not enteropathogenic, was found to be unable to agglutinate erythrocytes. Incubation with sialidase did not induce a hemagglutinating activity of PRCV, indicating that the lack of this activity is an intrinsic property of the virus and not due to the presence of competitive inhibitors. Only monoclonal antibodies to an antigenic site that is absent from the S protein of PRCV were able to prevent TGEV from agglutinating erythrocytes. The epitope recognized by these antibodies is located within a stretch of 224 amino acids that is missing in the S protein of PRCV. Our results indicate that the sialic acid binding activity is also located in that portion of the S protein. The presence of a hemagglutinating activity in TGEV and its absence in PRCV open the possibility that the sialic acid binding activity contributes to the enterotropism of TGEV.  相似文献   

16.
B Delmas  J Gelfi  E Kut  H Sjstrm  O Noren    H Laude 《Journal of virology》1994,68(8):5216-5224
The swine-specific coronavirus transmissible gastroenteritis virus (TGEV) uses pig aminopeptidase-N (pAPN) as a cellular receptor. We showed that the human aminopeptidase-N (hAPN) cannot substitute for pAPN in this respect, although the two enzymes have 80% amino acid sequence identity. In order to map the TGEV binding site on pAPN, we constructed a series of APN cDNA chimeras between pAPN and hAPN and analyzed them for their capacity to confer infectivity. The region between residues 717 and 813 was found to be essential for infectivity. This region also contains the epitopes for three TGEV-blocking monoclonal antibodies directed against pAPN. These data support the view that the catalytic site and the TGEV receptor site are located in different domains. Moreover, APN inhibitors and mutations in the catalytic site had no obvious effect on permissiveness for virus, thus providing evidence that the APN enzymatic activity is not involved in the process of infection.  相似文献   

17.
The coronaviruses (CoVs) are enveloped viruses of animals and humans associated mostly with enteric and respiratory diseases, such as the severe acute respiratory syndrome and 10–20% of all common colds. A subset of CoVs uses the cell surface aminopeptidase N (APN), a membrane-bound metalloprotease, as a cell entry receptor. In these viruses, the envelope spike glycoprotein (S) mediates the attachment of the virus particles to APN and subsequent cell entry, which can be blocked by neutralizing antibodies. Here we describe the crystal structures of the receptor-binding domains (RBDs) of two closely related CoV strains, transmissible gastroenteritis virus (TGEV) and porcine respiratory CoV (PRCV), in complex with their receptor, porcine APN (pAPN), or with a neutralizing antibody. The data provide detailed information on the architecture of the dimeric pAPN ectodomain and its interaction with the CoV S. We show that a protruding receptor-binding edge in the S determines virus-binding specificity for recessed glycan-containing surfaces in the membrane-distal region of the pAPN ectodomain. Comparison of the RBDs of TGEV and PRCV to those of other related CoVs, suggests that the conformation of the S receptor-binding region determines cell entry receptor specificity. Moreover, the receptor-binding edge is a major antigenic determinant in the TGEV envelope S that is targeted by neutralizing antibodies. Our results provide a compelling view on CoV cell entry and immune neutralization, and may aid the design of antivirals or CoV vaccines. APN is also considered a target for cancer therapy and its structure, reported here, could facilitate the development of anti-cancer drugs.  相似文献   

18.
Aminopeptidase N (APN), a 150-kDa metalloprotease also called CD13, serves as a receptor for serologically related coronaviruses of humans (human coronavirus 229E [HCoV-229E]), pigs, and cats. These virus-receptor interactions can be highly species specific; for example, the human coronavirus can use human APN (hAPN) but not porcine APN (pAPN) as its cellular receptor, and porcine coronaviruses can use pAPN but not hAPN. Substitution of pAPN amino acids 283 to 290 into hAPN for the corresponding amino acids 288 to 295 introduced an N-glycosylation sequon at amino acids 291 to 293 that blocked HCoV-229E receptor activity of hAPN. Substitution of two amino acids that inserted an N-glycosylation site at amino acid 291 also resulted in a mutant hAPN that lacked receptor activity because it failed to bind HCoV-229E. Single amino acid revertants that removed this sequon at amino acids 291 to 293 but had one or five pAPN amino acid substitution(s) in this region all regained HCoV-229E binding and receptor activities. To determine if other N-linked glycosylation differences between hAPN, feline APN (fAPN), and pAPN account for receptor specificity of pig and cat coronaviruses, a mutant hAPN protein that, like fAPN and pAPN, lacked a glycosylation sequon at 818 to 820 was studied. This sequon is within the region that determines receptor activity for porcine and feline coronaviruses. Mutant hAPN lacking the sequon at amino acids 818 to 820 maintained HCoV-229E receptor activity but did not gain receptor activity for porcine or feline coronaviruses. Thus, certain differences in glycosylation between coronavirus receptors from different species are critical determinants in the species specificity of infection.  相似文献   

19.
Sialic acids as receptor determinants for coronaviruses   总被引:3,自引:0,他引:3  
Among coronaviruses, several members are able to interact with sialic acids. For bovine coronavirus (BCoV) and related viruses, binding to cell surface components containing N-acetyl-9- O-acetylneuraminic acid is essential for initiation of an infection. These viruses resemble influenza C viruses because they share not only the receptor determinant, but also the presence of an acetylesterase that releases the 9- O-acetyl group from sialic acid and thus abolishes the ability of the respective sialoglycoconjugate to function as a receptor for BCoV. As in the case of influenza viruses, the receptor-destroying enzyme of BCoV is believed to facilitate the spread of virus infection by removing receptor determinants from the surface of infected cells and by preventing the formation of virus aggregates. Another coronavirus, porcine transmissible gastroenteritis virus (TGEV) preferentially recognizes N-glycolylneuraminic acid. TGEV does not contain a receptor-destroying enzyme and does not depend on the sialic acid binding activity for infection of cultured cells. However, binding to sialic acids is required for the enteropathogenicity of TGEV. Interaction with sialoglycoconjugates may help the virus to pass through the sialic acid-rich mucus layer that covers the viral target cells in the epithelium of the small intestine. We discuss that the BCoV group of viruses may have evolved from a TGEV-like ancestor by acquiring an acetylesterase gene through heterologous recombination.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号