首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
Due to the high mortality associated with recent, widely circulating strains of H5N1 influenza virus in poultry, the recurring introduction of H5N1 viruses from birds to humans, and the difficulties in H5N1 eradication by elimination of affected flocks, an effective vaccine against HPAI (highly pathogenic avian influenza) is highly desirable. Using reverse genetics, a set of experimental live attenuated vaccine strains based on recombinant H5N1 influenza virus A/Viet Nam/1203/04 was generated. Each virus was attenuated through expression of a hemagglutinin protein in which the polybasic cleavage site had been removed. Viruses were generated which possessed a full-length NS1 or a C-terminally truncated NS1 protein of 73, 99, or 126 amino acids. Viruses with each NS genotype were combined with a PB2 polymerase gene which carried either a lysine or a glutamic acid at position 627. We predicted that glutamic acid at position 627 of PB2 would attenuate the virus in mammalian hosts, thus increasing the safety of the vaccine. All recombinant viruses grew to high titers in 10-day-old embryonated chicken eggs but were attenuated in mammalian cell culture. Induction of high levels of beta interferon by all viruses possessing truncations in the NS1 protein was demonstrated by interferon bioassay. The viruses were each found to be highly attenuated in a mouse model. Vaccination with a single dose of any virus conferred complete protection from death upon challenge with a mouse lethal virus expressing H5N1 hemagglutinin and neuraminidase proteins. In a chicken model, vaccination with a single dose of a selected virus encoding the NS1 1-99 protein completely protected chickens from lethal challenge with homologous HPAI virus A/Viet Nam/1203/04 (H5N1) and provided a high level of protection from a heterologous virus, A/egret/Egypt/01/06 (H5N1). Thus, recombinant influenza A/Viet Nam/1203/04 viruses attenuated through the introduction of mutations in the hemagglutinin, NS1, and PB2 coding regions display characteristics desirable for live attenuated vaccines and hold potential as vaccine candidates in poultry as well as in mammalian hosts.  相似文献   

2.
Sporadic human infections by a novel H7N9 virus occurred over a large geographic region in China. In this study, we show that Newcastle disease virus (NDV)-vectored H7 (NDV-H7) and NDV-H5 vaccines are able to induce antibodies with high hemagglutination inhibition (HI) titers and completely protect chickens from challenge with the novel H7N9 or highly pathogenic H5N1 viruses, respectively. Notably, a baculovirus-expressed H7 protein failed to protect chickens from H7N9 virus infection.  相似文献   

3.
4.
Human infections with highly pathogenic avian influenza viruses of the H5N1 subtype, frequently reported since 2003, result in high morbidity and mortality. It is feared that these viruses become pandemic, therefore the development of safe and effective vaccines is desirable. MVA-based H5N1 vaccines already proved to be effective when two immunizations with high doses were used. Dose-sparing strategies would increase the number of people that can be vaccinated when the amount of vaccine preparations that can be produced is limited. Furthermore, protective immunity is induced ideally after a single immunization. Therefore the minimal requirements for induction of protective immunity with a MVA-based H5N1 vaccine were assessed in mice. To this end, mice were vaccinated once or twice with descending doses of a recombinant MVA expressing the HA gene of influenza virus A/Vietnam/1194/04. The protective efficacy was determined after challenge infection with the homologous clade 1 virus and a heterologous virus derived from clade 2.1, A/Indonesia/5/05 by assessing weight loss, virus replication and histopathological changes. It was concluded that MVA-based vaccines allowed significant dose-sparing and afford cross-clade protection, also after a single immunization, which are favorable properties for an H5N1 vaccine candidate.  相似文献   

5.
There is a constant threat of zoonotic influenza viruses causing a pandemic outbreak in humans. It is virtually impossible to predict which virus strain will cause the next pandemic and it takes a considerable amount of time before a safe and effective vaccine will be available once a pandemic occurs. In addition, development of pandemic vaccines is hampered by the generally poor immunogenicity of avian influenza viruses in humans. An effective pre-pandemic vaccine is therefore required as a first line of defense. Broadening of the protective efficacy of current seasonal vaccines by adding an adjuvant may be a way to provide such first line of defense. Here we evaluate whether a seasonal trivalent virosomal vaccine (TVV) adjuvated with the saponin-based adjuvant Matrix-M (MM) can confer protection against avian influenza H5 and H7 virus strains in mice and ferrets. We demonstrate that mice were protected from death against challenges with H5N1 and H7N7, but that the protection was not complete as evidenced by severe clinical signs. In ferrets, protection against H7N9 was not observed. In contrast, reduced upper and lower respiratory tract viral loads and reduced lung pathology, was achieved in H5N1 challenged ferrets. Together these results suggest that, at least to some extent, Matrix-M adjuvated seasonal virosomal influenza vaccine can serve as an interim measure to decrease morbidity and mortality associated with a pandemic outbreak.  相似文献   

6.

Background

Highly-pathogenic avian influenza virus (HPAIV) and Newcastle disease virus (NDV) are the two most important poultry viruses in the world. Natural low-virulence NDV strains have been used as vaccines over the past 70 years with proven track records. We have previously developed a reverse genetics system to produce low-virulent NDV vaccine strain LaSota from cloned cDNA. This system allows us to use NDV as a vaccine vector for other avian pathogens.

Methodology/Principal Finding

Here, we constructed two recombinant NDVs (rNDVs) each of which expresses the hemagglutinin (HA) gene of HPAIV H5N1strain A/Vietnam/1203/2004 from an added gene. In one, rNDV (rNDV-HA), the open reading frame (ORF) of HA gene was expressed without modification. In the second, rNDV (rNDV-HAF), the ORF was modified so that the transmembrane and cytoplasmic domains of the encoded HA gene were replaced with those of the NDV F protein. The insertion of either version of the HA ORF did not increase the virulence of the rNDV vector. The HA protein was found to be incorporated into the envelopes of both rNDV-HA and rNDV-HAF. However, there was an enhanced incorporation of the HA protein in rNDV-HAF. Chickens immunized with a single dose of either rNDV-HA or rNDV-HAF induced a high titer of HPAIV H5-specific antibodies and were completely protected against challenge with NDV as well as lethal challenges of both homologous and heterologous HPAIV H5N1.

Conclusion and Significance

Our results suggest that these chimeric viruses have potential as safe and effective bivalent vaccines against NDV and. HPAIV. These vaccines will be convenient and affordable, which will be highly beneficial to the poultry industry. Furthermore, immunization with these vaccines will permit serological differentiation of vaccinated and avian influenza field virus infected animals.  相似文献   

7.
Current influenza vaccines are believed to confer protection against a narrow range of virus strains. The identification of broadly influenza neutralizing antibodies (bnAbs) has triggered efforts to develop vaccines providing ‘universal’ protection against influenza. Several bnAbs were isolated from humans recently vaccinated with conventional influenza vaccines, suggesting that such vaccines could, in principle, be broadly protective. Assessing the breadth-of-protection conferred to humans by influenza vaccines is hampered by the lack of in vitro correlates for broad protection. We designed and employed a novel human-to-mouse serum transfer and challenge model to analyze protective responses in serum samples from clinical trial subjects. One dose of seasonal vaccine induces humoral protection not only against vaccine-homologous H1N1 challenge, but also against H5N1 challenge. This heterosubtypic protection is neither detected, nor accurately predicted by in vitro immunogenicity assays. Moreover, heterosubtypic protection is transient and not boosted by repeated inoculations. Strategies to increase the breadth and duration of the protective response against influenza are required to obtain ‘universal’ protection against influenza by vaccination. In the absence of known correlates of protection for broadly protective vaccines, the human-to-mouse serum transfer and challenge model described here may aid the development of such vaccines.  相似文献   

8.
A safe and effective vaccine is the best way to prevent large-scale highly pathogenic avian influenza virus (HPAI) H5N1 outbreaks in the human population. The current FDA-approved H5N1 vaccine has serious limitations. A more efficacious H5N1 vaccine is urgently needed. Parainfluenza virus 5 (PIV5), a paramyxovirus, is not known to cause any illness in humans. PIV5 is an attractive vaccine vector. In our studies, a single dose of a live recombinant PIV5 expressing a hemagglutinin (HA) gene of H5N1 (rPIV5-H5) from the H5N1 subtype provided sterilizing immunity against lethal doses of HPAI H5N1 infection in mice. Furthermore, we have examined the effect of insertion of H5N1 HA at different locations within the PIV5 genome on the efficacy of a PIV5-based vaccine. Interestingly, insertion of H5N1 HA between the leader sequence, the de facto promoter of PIV5, and the first viral gene, nucleoprotein (NP), did not lead to a viable virus. Insertion of H5N1 HA between NP and the next gene, V/phosphorprotein (V/P), led to a virus that was defective in growth. We have found that insertion of H5N1 HA at the junction between the small hydrophobic (SH) gene and the hemagglutinin-neuraminidase (HN) gene gave the best immunity against HPAI H5N1 challenge: a dose as low as 1,000 PFU was sufficient to protect against lethal HPAI H5N1 challenge in mice. The work suggests that recombinant PIV5 expressing H5N1 HA has great potential as an HPAI H5N1 vaccine.  相似文献   

9.
There is a critical need to have vaccines that can protect against emerging pandemic influenza viruses. Commonly used influenza vaccines are killed whole virus that protect against homologous and not heterologous virus. Using chickens we have explored the possibility of using live low pathogenic avian influenza (LPAI) A/goose/AB/223/2005 H1N1 or A/WBS/MB/325/2006 H1N2 to induce immunity against heterologous highly pathogenic avian influenza (HPAI) A/chicken/Vietnam/14/2005 H5N1. H1N1 and H1N2 replicated in chickens but did not cause clinical disease. Following infection, chickens developed nucleoprotein and H1 specific antibodies, and reduced H5N1 plaque size in vitro in the absence of H5 neutralizing antibodies at 21 days post infection (DPI). In addition, heterologous cell mediated immunity (CMI) was demonstrated by antigen-specific proliferation and IFN-γ secretion in PBMCs re-stimulated with H5N1 antigen. Following H5N1 challenge of both pre-infected and naïve controls chickens housed together, all naïve chickens developed acute disease and died while H1N1 or H1N2 pre-infected chickens had reduced clinical disease and 70–80% survived. H1N1 or H1N2 pre-infected chickens were also challenged with H5N1 and naïve chickens placed in the same room one day later. All pre-infected birds were protected from H5N1 challenge but shed infectious virus to naïve contact chickens. However, disease onset, severity and mortality was reduced and delayed in the naïve contacts compared to directly inoculated naïve controls. These results indicate that prior infection with LPAI virus can generate heterologous protection against HPAI H5N1 in the absence of specific H5 antibody.  相似文献   

10.
[目的]建立禽流感H5N1病毒感染恒河猴的动物模型,探讨禽流感在哺乳类动物的发病机制。[方法]通过“环甲膜穿刺术”经气管注射鸡胚培养的禽流感H5N1病毒(AF148678;ACGoose/Guangdong/11961H5N1)感染恒河猴,观察恒河猴染毒后出现的临床体征,用显微计数法检测外周血白细胞的动态变化,用ELISA检测禽流感病毒特异性抗体变化规律,用流式细胞仪检测外周血T淋巴细胞及其亚群的动态变化。在染毒后第1天、第3天、第10天和第14天分别剖杀染毒组恒河猴1只,HE染色观察主要组织器官的病理变化,用病毒分离、免疫组化和RT-PCR三种方法分析禽流感病毒侵袭机体的特点。[结果]临床症状和体征:急性起病,表现为发热,呼吸困难,精神状态下降,活动度明显减少,食欲下降,咳嗽,紫绀等,肺部听诊双肺可闻及干、湿音。1、病理特点:以肺部损伤为主,伴多器官病变。肺部的病变主要表现为弥漫性肺泡损伤,先后经历渗出期、增生期和纤维化期;在肝脏、肾脏和中枢神经系统中也观察到变性、坏死等病理变化。2、病毒侵袭机体的特点:病毒只在呼吸系统中复制,不在呼吸道以外的组织器官中复制;肺内支气管上皮细胞、肺泡上皮细胞和肺巨噬细胞是禽流感病毒侵犯的主要细胞类型。3、外周血象特点:外周血白细胞总数、淋巴细胞数出现短暂的下降,中性粒细胞数先升后降,但均于感染第7天后逐渐恢复到正常水平。4、抗体变化特点:感染后第7~11天,抗体水平持续快速升高;感染第11天后,抗体水平呈逐渐缓慢升高趋势(观察到染毒后50天为止)。5、细胞免疫特点:细胞免疫功能受损,表现为CD3+T淋巴细、CD3+CD4+T淋巴细胞和CD3+CD8+T淋巴细胞均出现短暂的下降,但这种细胞免疫功能受损是可逆的,到感染第7天后逐渐恢复回升至正常。[结论]1、恒河猴感染后的临床特点、病理变化、外周血象、免疫反应等均与人禽流感严重病例相类似,表明该模型是成功的,可为禽流感病毒在人体内致病机理的研究以及抗禽流感病毒的药物和疫苗评价提供最近似于人类的动物模型。2、综合本研究的实验结果,我们认为,H5N1禽流感毒主要攻击的对象为呼吸系统,不在呼吸道以外的组织器官中复制。禽流感病毒感染引起的急性弥漫性肺损伤是发病的中心环节,其发病可能经过病毒侵入、复制阶段,免疫损伤阶段和多器官功能损伤阶段。  相似文献   

11.
CY Wu  YC Yeh  JT Chan  YC Yang  JR Yang  MT Liu  HS Wu  PW Hsiao 《PloS one》2012,7(8):e42363
The recent threats of influenza epidemics and pandemics have prioritized the development of a universal vaccine that offers protection against a wider variety of influenza infections. Here, we demonstrate a genetically modified virus-like particle (VLP) vaccine, referred to as H5M2eN1-VLP, that increased the antigenic content of NA and induced rapid recall of antibody against HA(2) after viral infection. As a result, H5M2eN1-VLP vaccination elicited a broad humoral immune response against multiple viral proteins and caused significant protection against homologous RG-14 (H5N1) and heterologous A/California/07/2009 H1N1 (CA/07) and A/PR/8/34 H1N1 (PR8) viral lethal challenges. Moreover, the N1-VLP (lacking HA) induced production of a strong NA antibody that also conferred significant cross protection against H5N1 and heterologous CA/07 but not PR8, suggesting the protection against N1-serotyped viruses can be extended from avian-origin to CA/07 strain isolated in humans, but not to evolutionally distant strains of human-derived. By comparative vaccine study of an HA-based VLP (H5N1-VLP) and NA-based VLPs, we found that H5N1-VLP vaccination induced specific and strong protective antibodies against the HA(1) subunit of H5, thus restricting the breadth of cross-protection. In summary, we present a feasible example of direction of VLP vaccine immunity toward NA and HA(2), which resulted in cross protection against both seasonal and pandemic influenza strains, that could form the basis for future design of a better universal vaccine.  相似文献   

12.
禽流感H5N1亚型病毒感染ICR小鼠的动物模型   总被引:3,自引:1,他引:2  
目的 建立H5N1禽流感病毒感染ICR小鼠的疾病动物模型.方法 将100 μL H5N1 禽流感病毒原液(EID50为105.37/0.2 mL) 鼻腔接种ICR小鼠,设生理盐水组、正常尿囊液组对照,接毒后14 d内每隔12 h观察一次,主要观测指标有临床体征、体重和体温变化、死亡率、病理变化、病毒分离和血清抗体检测 (ELISA方法).结果 被感染的ICR小鼠的病程可以划分为潜伏期 (第0~1天)、急性感染期 (第2~7天)、恢复期 (第8~14天),急性感染期表现出活动明显减少,弓背,反应性差,扎堆;接毒后第1天开始体温和体重下降,第6天体温和体重停止下降;接毒组ICR小鼠累计的死亡率为60%;急性感染期ICR小鼠的肺部病变最严重,表现为间质性肺炎,肺间质充血、水肿和淋巴细胞浸润,毛细血管扩张,上皮细胞变性、坏死、脱落,并有充血和单核细胞浸润;接毒后第1天至第8天可在小鼠的肺、脑、气管和心、肝、脾、肾分离到病毒;接毒后第6天从ICR小鼠血清中检测到抗体.结论 本实验室建立的H5N1禽流感病毒感染ICR小鼠的模型在临床表现、体重变化、死亡率、病理变化、病毒复制指标能达到禽流感病毒疾病模型的造模要求,符合人类禽流感感染疾病的基本特征.  相似文献   

13.
Avian influenza continues to circulate and remains a global health threat not least because of the associated high mortality. In this study antibody persistence, booster vaccine response and cross-clade immune response between two influenza A(H5N1) vaccines were compared. Participants aged over 18-years who had previously been immunized with a clade 1, A/Vietnam vaccine were re-immunized at 6-months with 7.5 μg of the homologous strain or at 22-months with a clade 2, alum-adjuvanted, A/Indonesia vaccine. Blood sampled at 6, 15 and 22-months after the primary course was used to assess antibody persistence. Antibody concentrations 6-months after primary immunisation with either A/Vietnam vaccine 30 μg alum-adjuvanted vaccine or 7.5 μg dose vaccine were lower than 21-days after the primary course and waned further with time. Re-immunization with the clade 2, 30 μg alum-adjuvanted vaccine confirmed cross-clade reactogenicity. Antibody cross-reactivity between A(H5N1) clades suggests that in principle a prime-boost vaccination strategy may provide both early protection at the start of a pandemic and improved antibody responses to specific vaccination once available.Trial Registration: ClinicalTrials.gov NCT00415129  相似文献   

14.
In preparing for the threat of a pandemic of avian H5N1 influenza virus, we need to consider the significant delay (4 to 6 months) necessary to produce a strain-matched vaccine. As some degree of cross-reactivity between seasonal influenza vaccines and H5N1 virus has been reported, this was further explored in the ferret model to determine the targets of protective immunity. Ferrets were vaccinated with two intramuscular inoculations of trivalent inactivated split influenza vaccine or subcomponent vaccines, with and without adjuvant, and later challenged with a lethal dose of A/Vietnam/1203/2004 (H5N1) influenza virus. We confirmed that vaccination with seasonal influenza vaccine afforded partial protection against lethal H5N1 challenge and showed that use of either AlPO4 or Iscomatrix adjuvant with the vaccine resulted in complete protection against disease and death. The protection was due exclusively to the H1N1 vaccine component, and although the hemagglutinin contributed to protection, the dominant protective response was targeted toward the neuraminidase (NA) and correlated with sialic acid cleavage-inhibiting antibody titers. Purified heterologous NA formulated with Iscomatrix adjuvant was also protective. These results suggest that adjuvanted seasonal trivalent vaccine could be used as an interim measure to decrease morbidity and mortality from H5N1 prior to the availability of a specific vaccine. The data also highlight that an inducer of cross-protective immunity is the NA, a protein whose levels are not normally monitored in vaccines and whose capacity to induce immunity in recipients is not normally assessed.  相似文献   

15.
16.
为了研究 H5N1 DNA 疫苗对小鼠和鸡的保护效率,用 H5N1 禽流感病毒 HA DNA 疫苗免疫 BALB/c 小鼠和 SPF 鸡 . 小鼠和鸡分别经电穿孔和肌肉注射免疫两次,间隔为 3 周 . 二次免疫后,用致死量的同源病毒进行攻毒实验 . 空白对照组在攻毒后全部死亡,而经电穿孔免疫的小鼠和鸡均获得了完全的保护,并能有效地抑制病毒在小鼠肺脏和鸡泄殖腔的繁殖 . 同时,电穿孔免疫的小鼠和鸡均产生了高水平的特异性抗体 . 经电穿孔免疫的小鼠攻毒后 CTL 反应明显加强 . 这些结果表明, HA DNA 疫苗能有效地保护小鼠和鸡对禽流感病毒的感染,同时也表明电穿孔免疫是 DNA 疫苗免疫的有效途径之一 .  相似文献   

17.
18.
Despite great efforts to control the infection of poultry with H5N1 viruses, these pathogens continue to evolve and spread in nature, threatening public health. Elucidating the characteristics of H5N1 avian influenza virus will benefit disease control and pandemic preparation. Here, we sequenced the genomes of 15 H5N1 avian influenza viruses isolated in Vietnam in 2006 and 2007 and performed phylogenetic analyses to compare these sequences with those of other viruses available in the public databases. Molecular characterization of the H5N1 viruses revealed that seven genetically distinct clades of H5N1 viruses have appeared in Vietnam. Clade 2.3.4 viruses existed in Vietnam as early as 2005. Fifteen viruses isolated during 2006 and 2007 belonged to clade 1 and clade 2.3.4, and were divided into five genotypes. Reassortants between the clade 1 and clade 2.3.4 viruses were detected in both North and South Vietnam. We also assessed the replication and pathogenicity of these viruses in mice and found that these isolates replicated efficiently and exhibited distinct virulence in mice. Our results provide important information regarding the diversity of H5N1 viruses in nature.  相似文献   

19.
The spread of the recently emerged, highly pathogenic H5N1 avian influenza virus has raised concern. Preclinical studies suggest that passive immunotherapy could be a new form of treatment for H5N1 virus infection. Here, a neutralizing monoclonal antibody (MAb) against the hemagglutinin (HA) of the influenza A/chicken/Hatay/2004 H5N1 virus, MAb 9F4, was generated and characterized. MAb 9F4 binds both the denatured and native forms of HA. It was shown to recognize the HA proteins of three heterologous strains of H5N1 viruses belonging to clades 1, 2.1, and 2.2, respectively. By use of lentiviral pseudotyped particles carrying HA on the surface, MAb 9F4 was shown to effectively neutralize the homologous strain, Hatay04, and another clade 1 strain, VN04, at a neutralization titer of 8 ng/ml. Furthermore, MAb 9F4 also neutralized two clade 2 viruses at a neutralizing titer of 40 ng/ml. The broad cross-neutralizing activity of MAb 9F4 was confirmed by its ability to neutralize live H5N1 viruses of clade 2.2.2. Epitope-mapping analysis revealed that MAb 9F4 binds a previously uncharacterized epitope below the globular head of the HA1 subunit. Consistently, this epitope is well conserved among the different clades of H5N1 viruses. MAb 9F4 does not block the interaction between HA and its receptor but prevents the pH-mediated conformational change of HA. MAb 9F4 was also found to be protective, both prophylactically and therapeutically, against a lethal viral challenge of mice. Taken together, our results showed that MAb 9F4 is a neutralizing MAb that binds a novel and well-conserved epitope in the HA1 subunit of H5N1 viruses.The highly pathogenic avian influenza A subtype H5N1 virus was first isolated from geese in Guangdong province, China, in 1996 (44). Since 2003, the H5N1 strains have caused major morbidity and mortality in poultry populations across Asia, Europe, and Africa (3, 25). In 1997, the virus was transmitted from chickens to humans in Hong Kong, causing 18 reported cases of illness, including 6 deaths (6, 7, 37). As of September 2009, there were 442 confirmed human infections in 15 countries, with an alarming fatality rate of 59% (42). Although occurrences of human H5N1 infection are sporadic and rare, its rapid dissemination, the ongoing evolution of the avian H5N1 virus, and the absence of anti-H5N1 herd immunity in humans raise concerns regarding a possible H5N1 influenza pandemic (2, 4, 13). Since human infections are associated with severe disease and high mortality, the consequences of a pandemic could be catastrophic.Current strategies against influenza include vaccination and antiviral drug treatment (1). Due to the existence of multiple antigenic clades and subclades of the H5N1 virus, the difficulty of predicting the major strain that may cause the next pandemic is the main obstacle to current vaccine development. Moreover, resistance to M2 ion channel inhibitors (rimantidine and amantidine) has been reported in H5N1 isolates (1, 5), and the neuraminidase inhibitors (oseltamivir and zanamivir) require higher doses and prolonged treatment (45), and resistance has been reported in children (21). Passive immunotherapy is now increasingly used to treat numerous human infectious diseases (28, 33). Convalescent-phase blood and serum products were used to improve clinical outcomes for severely ill influenza patients during the 1918 influenza pandemic (27). Promising results with mouse models using a neutralizing monoclonal antibody (MAb) for H5N1 influenza treatment (17, 26) and a report of the recovery of an H5N1 virus-infected patient after treatment with convalescent-phase plasma (47) indicate that MAbs could be a potential treatment against H5N1 viruses.The hemagglutinin (HA) protein is one of the two major surface glycoproteins on the envelope of influenza A virus, with 16 distinct types identified in the avian species. The HA protein is responsible for receptor binding to host cells and for viral entry and is therefore the primary target of neutralizing antibodies (Abs) (35). It is a homotrimer, with each subunit made up of two disulfide-linked polypeptides, HA1 and HA2. Structurally, each subunit consists of a membrane-proximal helix-rich stem structure and a membrane-distal receptor binding globular domain (35).In this study, we describe a MAb, named MAb 9F4, raised against the recombinant baculovirus-expressed HA protein of A/chicken/Hatay/2004 H5N1 virus. Its neutralizing property was investigated, and epitope mapping was performed. The MAb 9F4 binding site was found to lie outside previously characterized antigenic sites in the HA protein. This epitope is well conserved among the different clades of H5N1 viruses, consistent with the cross-neutralizing activity of MAb 9F4. The mode of inhibition was also investigated, and MAb 9F4 was found to mediate postattachment neutralization in a dose-dependent manner. Finally, the protective ability of MAb 9F4 was also evaluated in a mouse model, and it was shown to protect against lethal H5N1 challenge both prophylactically and therapeutically. Taken together, the data could provide new information for the design of an H5N1 vaccine, and MAb 9F4 may be a possible candidate for use in passive immunotherapy.  相似文献   

20.
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