用户名: 密码: 验证码:
重配甲型H3N2人流感疫苗的制备及免疫原性研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
流感病毒属于正粘病毒科(Orthomyxoviridae),是一种含有8个基因片段的分节段RNA病毒。病毒分为甲(A)、乙(B)、丙(C)三型,其中A型对人类危害最严重,A型引起的并发症危害大,易造成神经-内分泌-免疫网络的紊乱。
     A型流感病毒的基因组是负链RNA,因而直接不具有感染性,各个RNA片段需要与聚合酶蛋白(PB2、PBI、PA,统称为P蛋白)及核蛋白(NP)结合在一起形成核糖核蛋白复合物即RNPs才有活性。流感病毒感染时,病毒首先与宿主细胞表面的特异性HA受体唾液酸(SA,N-乙酞神经氨酸)残基结合,通过融膜进入细胞后释放出RNPS,RNPS进入细胞核才开始病毒基因组的复制和转录,每个RNA片段单独组成一个转录单位,转录出mRNA和互补RNA(cRNA),mRNA翻译合成病毒蛋白,cRNA复制生成子代RNA病毒,然后在细胞浆中组装成完整的病毒粒子。
     流感病毒的反向遗传操作技术建立的难度较大,原因在于要同时在细胞内形成8个功能性核糖核蛋白复合物(RNPs)。流感病毒拯救技术的发展滞后于其它负义RNA病毒,流感病毒与大多数其它负义RNA病毒不同在于流感病毒基因组是在细胞核内复制。经过近10年的发展,在1999年Neumann等和Fodor等分别报道了改造后完全以质粒为基础的反向遗传技术,这是流感病毒拯救技术发展史上的转折点。其优点是不再像早期的方法那样感染细胞不再需要为RNA合成提供辅助病毒,从而避免了大量的筛选工作。
     本研究采用RG技术,通过8质粒流感病毒拯救系统,将冷适应、减毒病毒株A/AA/6/60(H2N2)的6个内部基因全片段克隆入pAD3000,其中的PB2、PB1、NP基因人工引入共5个氨基酸的突变。同时把2006~2007年流行株A/Wisconsin/67/2005(H3N2)的2个表面基因片段克隆入pAD3000,从而构建了8个转录/表达载体,用于拯救重配H3N2亚型人流感病毒减毒株,为流感减毒活疫苗的制备提供了保证。
     研究采用由美国St.Jude儿童研究医院的Robert Webster博士提供的上述PR8 8质粒系统,一一替换成构建好的用于重配病毒拯救的8个转录/表达质粒,共有8种7+1组合,验证其中每一个基因片段是否能够正常工作;同时用PR8的HA和NA表面基因以及H1N1的表面基因与验证好的冷适应株的6个内部骨架组合(6+2组合,重配病毒分别简写为rgAA-PR8和rgAA-H1N1),共转染细胞,验证冷适应株A/AA/6/60的6个内部基因是否能够协同发挥作用,同时对重配病毒的生物学特性进行了鉴定;在此基础上,接着把2006~2007年流行株A/Wisconsin/67/2005(H3N2)的表面基因与A/AA/6/60的6个骨架基因进行重排组合,对产生的重配病毒(6+2组合,简写为rgAA-H3N2)进行了初步鉴定,深入的研究还在进行之中。这些研究内容为我们弄清楚流感病毒基因组及其蛋白质在病毒复制及致病机制中的作用建立了平台,也为冷适应流感减毒活疫苗的研制以及粘膜免疫保护机制的研究提供了理论依据。
     本部分实验针对弱毒疫苗的特点,建立了rgAA-H3N2病毒株的三级毒种库并进行系统、全面鉴定,同时免疫BALB/c小鼠利用HI和ELISA等方法对其免疫原性进行了初步研究,为H3N2亚型流感减毒活疫苗的研发奠定了理论基础。
     这些研究为研制高效、安全的冷适应流感减毒活疫苗以及阐明流感病毒呼吸道粘膜免疫保护机制奠定了基础,为其它病毒减毒活疫苗研制奠定了基础。
Influenza virus is the pathogen of influenza, which is a kind of highly contagious disease with the first incidence rate among all of the infectious diseases and hazardous infection which WHO declares to be strictly monitored. The three flu pandemics of the 20~(th) century were devastating. Especially in the 1918/1919 "Spanish flu", more than 20 million people died which exceeded the total number of people killed during the World War I and most of them were young people. Till now people could not control the disease due to the high variability of influenza virus in spite of the rapid progress of the modern scientific technology.
     Influenza virus, a member of the family Orthomyxoviridae, is a negative sense RNA virus with eight genomic segments. Influenza viruses are divided into types A, B and C. Influenza A viruses are the principal causes of influenza in humans which cause serious complications and disorder of immune nerve-endocrine-network. Moreover, it imposes a huge burden on the human health and the national economy.
     The antigens of the encoding influenza virus include HA, NA, NP and M polypeptides. HA and NA are two major antigens on the surface of influenza virus. HA is the major protective antigen of influenza virus to stimulate the production of neutralizing antibody. Comparison with HA, NA is conserved in a degree,which stimulates the production of antibody as same. As a result of seasonal antigenic variation of hemagglutinin(HA)and neuraminidase(NA)which decide the specificity of the pathogen, new virus strain appears annually, which pose a major obstacle to control influenza.
     To date, there is no effective therapeutic tool for influenza. Though influenza vaccines have been produced for over 50 years, vaccination is still the principal method of prophylaxis. Vaccines can be characterized as whole virus vaccines, split virus vaccines, surface antigen vaccines, DNA vaccines and live attenuated vaccines. Due to frequent antigenic shift, vaccine strain used in production needs to be changed annually. Commercially available trivalent inactivated vaccines suffer from limited efficacy in cross-protective immunity and routes of administration, given by intramuscular injection. Surface antigen vaccines can be used safely, but targeting mainly HA and NA antigen. They are not sufficiently cross-reactive to protect against antigenic variants, vaccine production is time-consuming and they are not fit for seasonal influenza. DNA vaccine developed late and aimed at protective antigen formulation. There are still some problems such as large antigen dose and gene integration with chromosomal which are difficult to solve. However, live attenuated vaccine is able to induce strong systemic immunity and humoral responses and local mucosal immune responses, but remains invariably virulence and potential rebound of toxicity. In view of the status quo of influenza vaccines, much attention has been given to the development of effective and neotype influenza vaccines. Recently, the reverse genetics provided a favourable opportunity for live attenuated vaccines. Efforts are currently under way to develop a live attenuated influenza vaccine which based on reverse genetics. Since 1999, Neumann and Hoffmann et al have developed an eight-plasmid DNA transfection system for the rescue of infectious influenza A virus entirely from cloned cDNA. The development opens the way for the study of live attenuated influenza vaccine. Rescuing recombinant influenza virus vaccine on the MDV-A background is a focus of influenza vaccine. In this dissertation, we used the eight-plasmid rescue system and selected 6 internal gene of influenza strain A/Ann Arbor/6/60(H2N2) as the genetic backbone for generation of recombinant H1N1 subtype influenza virus and 6+2 reassortants expressing the HA and NA from the 2006~2007 circulating strain A/New Caledonia/20/99(H1N1). Finally, we successfully established a cold-adapted H1N1 subtype influenza virus rescue system, this reverse genetics platform is strategically important for vaccine development and influenza prevention.
     The whole dissertation consists of three parts:
     1. Construction of eight-plasmid system for influenza virus
     Based on functional bi-directional transcription/expression vector pAD3000, influenza virus strain A/AnnArbor/6/60 (H2N2), a cold-adapted (ca), temperature sensitive (ts), live attenuated was used as the master donor virus (MDV) for virus rescue, in which six internal gene fragments were fully synthesized. Meanwhile, five amino acid substitutions have been artificially altered by human intervention and six pairs specific primers were designed according to references. The 2006~2007 circulating influenza virus strain A/Wisconsin/67/2005(H3N2) was propagated in 10-day-old embryonated chicken eggs and concentrated by density gradient centrifugation on sucrose. Total RNA was extracted from infected allantoic fluid. The HA and NA gene were amplified using the universal primer pairs according to Hoffmann. Expand High Fidelity PCR System was used and 8~10 clones were sequenced in order to confirm the authenticity of 5' and 3' ends. Eight transcription /expression plasmids were obtained and named as pMDV-A-PB2, pMDV-A-PB1, pMDV-A-PA, pMDV-A-NP, pMDV-A-M, pMDV-A-NS, pHW214-H and pHW214-NA, respectively. Finally, transcription and expression of vRNA and mRNA were realized by using one vector and pol I /pol II RNA polymerase of cells after transfection, which contribute to the construction of cold-adapted influenza virus rescue system.
     2. Preparation of reassortant H1N1 subtype influenza virus
     2.1 Validation of cold-adapted influenza virus rescue system
     Some desired infectious H1N1 subtype recombinant viruses were produced by co-transfection a single gene segment from MDV-A together with the complementary seven segments from control A/PR/8/34 strain. In addition, HA and NA derived from strain A/New Caledonia/20/99 or A/PR/8/34 and other internal gene segments from A/Ann Arbor/6/60, in other words, there are eight kinds of 7+1 and a kind of 6+2 combinations. The results revealed that each of the 8 genomic segments cloned in pAD3000 was showed to be functionally expressed in a reassortant experiment and 6 internal gene backbone work synergialy. Meanwhile, it prepared for further screening and construction cold-adapted reassortant virus. The transfection reaction using the 8 A/PR/8/34 plasmids was used as a positive control. During these courses, progeny virus for eggs propagation is produced in 35mm dishes by optimizing rescue systems.
     Then, we identified and analyzed the partial biological properties of the recoveried viruses PB2/PR8, PR8 and PR8/rMDV-A. The rescued viruses had similar morphology to wild-type virus strain by electronic microscope. Amino acid substitutions were present in progeny virus by RT-PCR. The reassortant virus was identified by HA, HI and IFA experiment. It showed that reassortant virus propagation prefer to lower temperature such as 33 degree by PFU of PR8/rMDV-A. PR8/rMDV-A grew to high titer and weak virulence in embryonic eggs during passage of the virus and the cytoplasmic effect on infected MDCK cells were correlated with the strain A/New Caledonia/20/99, virulence and propagation in lower temperature were correlated with the master donor virus strain A/Ann Arbor/6/60. These results showed that the cold-adapted rescue system worked well.
     2.2 Generation and identification of H3N2 subtype attenuated influenza virus vaccine
     Based on the functional cold-adapted virus rescue system, we reassorted HA and NA gene of the 2006~2007 circulating influenza virus strain A/Wisconsin/67/2005(H3N2). A positive H3N2 subtype recombinant virus was obtained successfully by co-transfecting COS-1 cells. A HA titer of approximately 1:512~1:1024 were detected in the first passage of the rescued viruses. Virus titer was stable and the homology of HA and NA gene were above 99.9% following four passages. The virion morphology of the rescued viruses was verified to be identical to that of the wild-type PR8-strain. Preliminary identification was carried out and further study is in progress. The establishment of cold-adapted H1N1 subtype rescue system by RG will contribute to development of cold-adapted human influenza virus vaccine candidates and mucosal immune mechanisms against influenza.
     3. Construction of Three-tier Virus Seed Library and immunogenicity test of rgAA-H3N2
     3.1 Construction of Three-tier Virus Seed Library of rgAA-H3N2
     The three-tier virus seed library of rgAA-H3N2 was constructed which contained the primary seed lots, the master seed lots and working seed lots. The titers of seeds from working seed lots were above 7.2 LogTCID_(50)/ml. The seeds were qualified in sterility and mycoplasma tests, and negative in haemadsorption and non-haemadsorption tests. In test for adventitious agents, both the survival rates of mice and suckling mice were more than 86 %. All the quality indexes of rgAA-H3N2 strain complied with the Chinese Requirements for Biologics 2005. It may be used as the seeds for production of cold-adapted attenuated influenza vaccine.
     3.2 Immunogenicity of reassortment virus rgAA-H3N2
     The working seed lots of rgAA-H3N2 were injected into allantoic cavity of 10 days embryonated chick eggs. The allantoic fluid was collected which HA titre≥2~8 after incubation. The purified antigen was got by ultracentrifugation. Six-eight week old BALB/c mice were immunized with rgAA-H3N2 intranasally and subcutaneous injection. The immune dose was 10~5TCID50 and the internal time was 28d between two immune. The serum were collected after 28d of primary immune, and 14d of second immune, neutral antibodies were detected by HI test. Specificity sera IgG were detected by ELISA. At the same time ,the local sIgA in lung wash and nasal wash were detected by ELISA. These results opens the way for farther research on gene function and novel vaccine candidate of cold-adapted, live attenuated human influenza virus.
引文
[1]Liang SH, Mozdzanowska K, Palladino G, et al. Heterosubtypic immunity to influenza type A virus in mice-effector mechanisms and their longevity. J Immunol, 1994,152:1653-61.
    [2]Yewdell JW, Bennink JR, Smith GL, et al. Influenza virus nucleoprotein is a major target antigen for cross-reactive anti-influenza A virus cytotoxic T lymphocytes. Proc Natl Acad Sci USA, 1985,82:1785-9.
    [3]Zambon MC. Epidemiology and pathogenesis of influenza. J Antimicrobial Chemotherapy, 1999, 44:3-9.
    [4]Goto H, Kawaoka Y. A novel mechanism for the acquisition of virulence by a human influenza A virus. Proc Natl Acad Sci USA, 1998,95:10224-8.
    [5]Stuart-Harris CH. A neurotropic strain of human influenza virus. Lancet, 1939,236: 497-9.
    [6]Lazarowitz SG, Goldberg AR, Choppin PW. Proteolytic cleavage by plasmin of the HA polypeptide of influenza virus: host cell activation of serum plasminogen. Virology, 1973, 56: 172-80.
    [7]Reid AH, Fanning TG, Janczewski TA, et al. Characterization of the 1918 "Spanish" influenza virus neuraminidase gene. Proc Natl Acad Sci U S A , 2000,97: 6785-90.
    [8]Almond JW. A single gene determines the host range of influenza virus. Nature, 1977, 270:617-8.
    [9]Subbarao EK, London W, Mupphy BR. A single amino acid in the PB2 gene of influenza A virus is a determinant of host range. J Virol, 1993,67(4):1761~4.
    
    [10]Hoffmann E, Stech J, Leneva I, et al. Characterization of the influenza A virus gene pool in avian species in southern China: was H6N1 a derivative or a precursor of H5N1 ? J Virol, 2000, 74(14):6309~15.
    [11]Guan Y, Shortridge KF, Krauss S, et al. Molecular characterization of H9N2 influenza viruses: Were they the donors of the " internal" genes of H5N1 viruses in Hong Kong? Proc Natl Acad Sci USA, 1999,96:9363-7.
    [12]Perales B, Ortin J. The influenza A virus PB2 polymerase subunit is required for the replication of viral RNA. J Virol, 1997,71(2):1381~5.
    [13]Guan Y, Shortridge KF, Krauss S. H9N2 influenza viruses possessing H5N1-like internal genomes continue to circulate in poultry in southeastern China. J Virol, 2000, 74(20):9372~80.
    [14]Massin P, Werf SV, Naffakh N. Residue 627 of PB2 is a determinant of cold sensitivity in RNA replication of avian influenza viruses. J Virol, 2001,5398~404.
    [15]McCauley JW, Penn CR. The crucial cut-off temperature of avian influenza viruses. Virus Res, 1990,17(3):191~8.
    [16]Luytjes W, Krystal M, Enami M et al. Amplification, expression, and packaging of foreign gene by influenza virus. Cell, 1989, 59:1107-13.
    [17]Naffakh N, Massin P, van der Werf S. The transcription/replication activity of the polymerase of influenza A viruses is not correlated with the level of proteolysis induced by the PA subunit. Virology, 2001,285(2):244~52.
    [18]Pinto LH, Holsinger, LJ, Lamb, RA. Influenza virus M2 protein has ion channel activity. Cell, 1992,69:517-28.
    [19]Bui M. Wills EG, Helenius A, et al. Role of the influenza virus Ml protein in nuclear export of viral ribonucleoproteins. J Virol, 2000, 74: 1781-6.
    [20]O'Neill RE, Talon J, Palase P. The influenza virus NEP(NS2protein) mediates the nuclear export of viral ribonucleoproteins. EMBO J, 1998,17:288-96.
    [21]Higgins DA. Comparative immunology of avian species. In: Davison TF, Morris TR, Payne LN, editors. Poultry immunology, 24th ed. Abingdon: Carfax Publishing Co, 1996. p. 149-205.
    [22]Wiley DC, Wilson IA, Skehel JJ. Structural identification of the antibody-binding sites of Hong Kong influenza haemagglutinin and their involvement in antigenic variation. Nature, 1981,289:373-8.
    [23]Philpott M, Easterday BC, Hinshaw VS. Neutralizing epitopes of the H5 hemagglutinin from a virulent avian influenza virus and their relationship to pathogenicity. J Virol, 1989,63:3453-8.
    [24]Johansson BE, Bucher DJ, Kilbourne ED. Purified influenza virus hemagglutinin and neuraminidase are equivalent in stimulation of antibody response but induce contrasting types of immunity to infection. J Virol, 1989,63:1239-46.
    [25]Rott R, Becht H, Orlich M. The significance of influenza virus neuraminidase in immunity. J Gen Virol, 1974,22:35-41.
    [26]McNulty MS, Allan GM, Adair BM. Efficacy of avian influenza neuraminidase-specific vaccines in chickens. Avian Pathol,1986,5:107~15.
    [27]Chen Z, Matsuo K, Asanuma H, et al. Enhanced protection against a lethal influenza virus challenge by immunization with both hemagglutinin-and neuraminidase-expressing DNAs. Vaccine, 1999,17:653-9.
    [28]Kodihalli S, Haynes JR, Robinson HL, et al. Cross-protection among lethal H5N2 influenza viruses induced by DNA vaccine to the hemagglutinin. J Virol, 1997,71(5):3391-6.
    [29]Swayne DE, Garcia M, Beck JR, et al. Protection against diverse highly pathogenic H5 avian influenza viruses in chickens immunized with a recombinant fowlpox vaccine containing an H5 avian influenza hemagglutinin gene insert. Vaccine,2000,18:1088~95.
    [30]Swayne DE, Beck JR, Garcia M, et al. Influence of virus strain and antigen mass on efficacy of H5 avian influenza inactivated vaccines. Avian Pathol, 1999,28:245-56.
    [31]Slepushkin VA, Katz JM, Black RA, et al. Protection of mice against influenza A virus challenge by vaccination with baculovirus-expressed M2 protein. Vaccine, 1995, 13:1399-402.
    [32]Zebedee SL, Lamb RA. Influenza A virus M2 protein: monoclonal antibody restriction of virus growth and detection of M2 in virions. J Virol,1988,62:2762~72.
    [33]Frace AM, Klimov AI, Rowe T, et al. Modified M2 proteins produce heterotypic immunity against influenza A virus. Vaccine,1999,17:2237~44.
    [35]Gelb JJ, Nix WA, Gellman SD. Infectious bronchitis virus antibodies in tears and their relationship to immunity. Avian Dis, 1998,42:364-74.
    [36]Jayawardane GWL, Spradbrow PB. Mucosal immunity in chickens vaccinated with the V4 strain of Newcastle Disease virus. Vet Microbiol, 1995,46:69-77.
    [37]Takada A, Kida H. Protective immune response of chickens against Newcastle Disease, induced by the intranasal vaccination with inactivated virus. Vet Microbiol, 1996,50:17-25.
    [38]Russell PH, Ezeifeka GO. The Hitchner B1 strain of Newcastle Disease virus induces high levels of IgA, IgG and IgM in newly hatched chicks. Vaccine,1995,13:61-6.
    [39]Higgins DA, Shortridge KF, Ng PLK.Bile immunoglobulin of the duck (Anas platyrhynchos ). II. Antibody response in influenza A virus infections. Immunology, 1987, 62:499-504.
    [40]Magor KE, Warr GW, Bando Y,et al. Secretory immune system of the duck(Anas platyrhynchos ). Identification and expression of the genes encoding IgA and IgM heavy chains. Eur J Immunol, 1998,28:1063-8.
    [41]Hodge LM, Marinaro M, Jones HP, et al. Immunoglobulin A (IgA) responses and IgE-associated inflammation along the respiratory tract after mucosal but not systemic immunization. Infect Immun.,2001,69(4):2328~38.
    [42]Guo L, Lu X, Kang SM, et al. Enhancement of mucosal immune responses by chimeric influenza HA/SHIV virus-like particles. Virology,2003,313(2):502~13.
    [43]Shapira-Nahor O, Zakay-Rones Z. Local and peripheral cell-mediated immune response to influenza virus in mice. J Med Virol, 1985,15(1):81 ~91.
    [44]Reeth KV, Gregory V, Hay A, et al. Protection against a European H1N2 swine influenza virus in pigs previously infected with H1N1 and/or H3N2 subtypes. Vaccine , 2003, 21:1375-81.
    [45]Liang SH, Mozdzanowska K, Palladino G, et al. Heterosubtypic immunity to influenza type A virus in mice-effector mechanisms and their longevity. J Immunol, 1994,152:1653-61.
    [46]Yewdell JW, Bennink JR, Smith GL, et al. Influenza virus nucleoprotein is a major target antigen for cross-reactive anti-influenza A virus cytotoxic T lymphocytes. Proc Natl Acad Sci USA, 1985,82:1785-9.
    [47]Doherty PC, Christensen JP. Assessing complexity: the dynamics of virus-specific T cell responses. Annu Rev Immunol, 2000,18:561-92.
    [48]Heinen PP, de Boer-Luijtze EA, Bianchi ATJ. Respiratory and systemic humoral and cellular immune responses of pigs to a heterosubtypic influenza A virus infection. J Gen Virol, 2001. 82:2697-707.
    [49]Huan H. Nguyen, Frederik W. van Ginkel,et al. Mechanism of heterosubtypic immunity to influenza A virus infection. International Congress Series,2001,1219:333-340.
    [50]Mak NK, Zhang YH, Ada GL, et al. Humoral and cellular responses of mice to infection with a cold-adapted influenza A virus variant. Infect Immun,1982,38(1):218-25.
    [51]Zinkernagel RM. Some general aspects of immunity to viruses. Vaccine, 1994,12:1493-4.
    [52]Campen H, Easterday BC, Hinshaw VS, et al. Virulent avian influenza A viruses: their effect in avian lymphocytes and macrophages in vivo and in vitro. J Gen Virol, 1989,70:2887-95.
    [53]Campen H, Easterday BC, Hinshaw VS, et al. Destruction of lymphocytes by a virulent avian influenza A virus. J Gen Virol, 1989,70:467-72.
    
    [54]Hinshaw VS, Olsen CW, Dybdahlsissoko N, et al. Apoptosis: a mechanism of cell killing by influenza A and B viruses. J Virol, 1994,68:3667-73.
    [55]Laudert E, Sivanandan V, Halvorson D. Effect of an H5N1 avian influenza virus infection on the immune system of mallard ducks. Avian Dis, 1993,37:845-53.
    [56]Yewdell JW, Hackett CJ. The specificity and function of T lymphocytes induced by influenza A virus,In The Influenza Viruses(Krug RM,ed.) 1989, Plenum Press,New York, p61.
    [57]McMichael AJ. Cytotoxic T lymphocytes specific for influenza virus. Curr Top Microbiol and Immunol,1994,189:75~91.
    [58]Lin YL, Askonas BA. Biological properties of an influenza A virus specific killer T cell clone—inhibition of virus replication in vivo and induction of delayed type hypersensitivity reactions. J Exp Med, 1981,154:225-34.
    [59]Bender BS, Croghan T, Small PA. Transgenic mice lacking class-I(CD8+) cytotoxic lymphocytes T(CTL) have delayed recovery and increased mortality following viral challenge. Clin Res, 1992, 40:247A.
    [60]Nguyen HH, Moldoveanu Z, Novak MW, et al. Heterosubtypic immunity to lethal influenza A virus infection is associated with virus-specific CD8+ cytotoxic T lymphocyte responses induced in mucosa-associated tissues. Virology, 1999;254:50~60.
    [61]Vignuzzi M, Gerbaud S, van der Werf S, et al. Naked RNA immunization with replicons derived from poliovirus and Semliki Forest virus genomes for the generation of a cytotoxic T cell response against the influenza A virus nucleoprotein. J Gen Virol, 2001,82:1737-47.
    [62]Pertmer TM, Oran AE, Moser JM,et al. DNA vaccines for influenza virus: differential effects of maternal antibody on immune responses to hemagglutinin and nucleoprotein. J Virol, 2000, 74(17):7787~93.
    [63]Graham MB, Braciale TJ. Resistance to and recovery from lethal influenza virus infection in B lymphocyte-deficient mice. J Exp Med, 1997,186(12):2063~8.
    [64]Nathen CF, Murray HW, Conn ZA. The macrophage as an effector cell. N Engl J Med, 1980, 303:622-6.
    [65]Powell PC. Macrophages and other nonlymphoid cells contributing to immunity. In: Toivanen A, Toivanen P,editors. Avian immunology. Boca Raton, 1987. p. 195-212.
    [66]Peschke T, Bender A, Nain M, et al. Role of macrophage cytokines in influenza a virus infections. Immunobiology, 1993,189:340-55.
    [67]Rodgers B, Mims CA. Interaction of influenza virus with mouse macrophages. Infect Immun, 1981,31:751-7.
    [68]Lowy RJ, Dimitrov DS. Characterization of influenza virus-induced death of J774.1 macrophages. Exp Cell Res, 1997,234:249-58.
    [69]Lyon JA, Hinshaw VS. Replication of influenza A viruses in an avian macrophage cell line. J Gen Virol, 1991,72:2011-3.
    [70]Dieter RR, Golemboski KA, Bloom SE, et al. The avian macrophage in cellular immunity. In:Sharma JM, editor. Avian cellular immunology,Boca Raton, 1991, p. 71-97.
    [71]Larson HE, Parry RP, Tyrrell DA. Impaired polymorphonuclear leucocyte chemotaxis after influenza virus infection. Br J Dis Chest, 1980,74:56-62.
    [72]Kodihalli S, Sivanandan V, Nagaraja KV, et al. Effect of avian influenza virus infection on the phagocytic function of systemic phagocytes and pulmonary macrophages of turkeys. Avian Dis, 1994,38:93-102.
    [73]Ronni T, Sareneva T, Pirhonen J, et al. Activation of IFN-alpha, IFN-gamma, MxA, and IFN regulatory factor 1 genes in influenza A virus-infected human peripheral blood mononuclear cells. J Immunol, 1995,154:2764-74.
    [74]Monteiro JM, Harvey C, Trinchieri G. Role of interleukin-12 in primary influenza virus infection. J Virol, 1998,72:4825-31.
    [75]Hayden FG, Fritz RS, Lobo MC, et al. Local and systematic cytokine responses during experimental human influenza A virus infection. J Clin Invest, 1998,101:643~9.
    [76]Schat KA. T-cell immunity: mechanisms and soluble mediators. In: Sharma JM, editor. Avian cellular immunology. Boca Raton, 1991, p. 35-51.
    
    [77]Klasing KC. Avian leukocytic cytokines. Poultry Sci, 1994,73:1035-43.
    [78]Kristien Van Reeth. Cytokines in the pathogenesis of influenza. Veterinary Microbiology, 2000,74: 109-16.
    [79]Schultz-Cherry S, Hinshaw VS. Influenza virus neuraminidase activates latent transforming growth factor beta. J Virol, 1996,70(12):8624~9.
    [80]WahI SM. Transforming growth factor beta (TGF-beta) in inflammation: a cause and a cure. J Clin Immunol, 1992,12:61-74.
    [81]Staheli P. Interferon-induced proteins and the antiviral state. Adv Virus Res, 1990, 38:147-200.
    
    [82]Pavlovic J, Zurcher T, Haller O, et al. Resistance to influenza virus and vesicular stomatitis virus conferred by expression of human MxA protein. J Virol, 1990,64:3370~5.
    [83]Staeheli P, Haller O. Interferon-induced Mx protein: a mediator of cellular resistance to influenza virus. Interferon ,1987,8:1-23.
    [84]Haller O, Arnheiter H, Lindenmann J. Genetically determined resistance to infection by hepatotropic influenza A virus in mice: effect of immunosuppression.Infect Immun, 1976, 13:844-54.
    [85]Lindenmann J, Deuel E, Fanconi S, et al. Inborn resistance of mice to myxoviruses: macrophages express phenotype in vitro. J Exp Med, 147;531~40.
    [86]Haller O, Arnheiter H, Lindenmann J. Natural, genetically determined resistance toward influenza virus in hemopoietic mouse chimeras. Role of mononuclear phagocytes. J Exp Med, 1979,150:117-26.
    [87]Ru. M. Interferon-mediated development of influenza virus resistance in hybrids between Mx gene-bearing and control mouse embryo fibroblasts. J Gen Virol,1983,64:1291~300.
    [88]Horisberger MA, Haller O, Amheiter H. Interferon-dependent genetic resistance to influenza virus in mice: virus replication in macrophages is inhibited at an early step. J Gen Virol, 1980,50:205-10.
    [89]Isaacs A, Lindenmann J. Virus interference. 1. The interferon. Proc R Soc Lond [Biol], 1957, 147:258-67.
    [90]Sekellick MJ, Ferrandino AE, Hopkins DA, et al. Chicken interferon gene: cloning, expression, and analysis. J Interferon Res, 1994,14:71-9.
    [91]Schultz U, Kaspers B, Rinderle C, et al. Recombinant chicken interferon from Escherichia coli and transfected COS cells is biologically active. Eur J Biochem, 1995,229:73-6.
    [92]Bernasconi D, Schultz U, Staeheli P. The interferon-induced Mx protein of chickens lacks antiviral activity. J Interferon Cytokine Res, 1995,15:47-53.
    [93]Bazzigher L, Schwarz A, Staeheli P. No enhanced influenza virus resistance of murine and avian cells expressing cloned duck Mx protein. Virology, 1993,195:100-12.
    [94]Hinshaw VS, Webster RG, Turner B. The perpetuation of orthomyxoviruses and paramyxoviruses in Canadian waterfowl. Can J Microbiol, 1980,26:622-9.
    [95]Song KD, Lillehoj HS, Choi KD, et al. Expression and functional characterization of recombinant chicken interferon-gamma. Vet Immuno Immunopathol, 1997,58:321-33.
    [96]Lowenthal JW, O'Neil TE, Broadway M, et al. Coadministration of IFN-g enhances antibody responses in chickens. J Interferon Cytokine Res, 1998,18:617-22.
    [97]Sekellick MJ, Lowenthal JW, O'Neil T, et al. Chicken interferon types I and II enhance synergistically the antiviral state and nitric oxide secretion. J Interferon Cytokine Res, 1998, 18:407-14.
    [98]Naikhin AN, Rekstin AR, Barantseva IB, et al. Immune response to live influenza vaccine. Vestn Ross Akad Med Nauk, 2002,(12):24~8.
    [99]Soboll G, Horohov DW, Aldridge BM, et al. Regional antibody and cellular immune responses to equine influenza virus infection, and particle mediated DNA vaccination. Vet Immunol Immunopathol, 2003,94(1-2):47~62.
    [100]Calain P, Roux L. The rule of six, a basic feature for efficient replication of Sendai virus defective interfering RNA. J Virol, 1993,67: 4822-30.
    [101]Radecke F, Spielhofer P, SchneiderH, et al. Rescue of Measles virus from cloned DNA. EMBO J, 1995,14.5773-84.
    
    [102] 薛强,仇华吉,童光志.RNA病毒的反向遗传学.生物技术通报,2001,4:1~4.
    [103]He B, Paterson RG, Ward CD, et al. Recovery of infectious SV5 from cloned DNA and expression of a foreign gene. Virology, 1997,237(2): 249-60.
    [104]Gassen U, Collins FM, Duprex W P, et al. Establishment of a rescue system for canine distemper virus. J Virol, 2000,74:10737-44.
    [105]Peeters BP, de Leeuw OS, Koch G, et al. Rescue of Newcastle disease virus from cloned cDNA : Evidence that cleavability of the fusion protein is a major determinant for virulence. J Virol, 1999,73:5001-9.
    [106]Hoffmann E, Stech J, Guan Y, et al. Universal primer set for the full-length amplification of all influenza A viruses. Arch Virol, 2001,146(12):2275~89.
    [107]Hirata T, Iida A, Shiraki-Iida T, et al. An improved method for recovery of F-defective Sendai virus expressing foreign genes from cloned cDNA. J Virol Methods, 2000, 104: 125-33.
    [108]Parks CL, Wang H, Kovacs GR, et al. Expression of a foreign gene by recombinant canine distemper virus recovered from cloned DNAs. Virus Res, 2002,83:131-47.
    [109]Subash C, Baron MD, Skinner MA, et al. Improved technique for transient expression and negative strand virus rescue using fowlpox T7 recombinant virus in mammalian cells. J Virological Methods, 2000, 89:119-27.
    [110] Poon LL, Pritlove DC, Sharps J, et al. The RNA polymerase of influenza virus, bound to the 5' end of virion RNA,acts in cis to polyadenylate mRNA. J Virol,1998,72(10):8214~19
    [111]Pritlove DC, Poon LL, Devenish LJ, et al. A hairpin loop at the 5'end of influenza A virus virion RNA is required for synthesis of poly(A)+ mRNA in vitro. J Virol, 1999,73(3):2109-14.
    [112]Collins PL, Mink MA, Stec DS. Rescue of synthetic analogs of respiratory syncytial virus genomic RNA and effect of truncations and mutations on the expression of a foreign reporter gene. Proc Natl Acad Sci USA, 1991,88: 9663-7.
    [113]Park KH, Huang T, Correia FF, et al. Rescue of a foreign gene by Sendai virus. Proc Natl Acad Sci USA, 1991, 88:5537-41.
    [114]Pattnaik AK, Wertz GW. Cells that express all five proteins of vesicular stomatitis virus from cloned cDNAs support replication, assembly, and budding of defective interfering particles. Proc Natl Acad Sci USA, 1991, 88, 1379-83.
    [115]Pattnaik AK, Wertz GW. Replication and amplification of defective interfering particle RNAs of vesicular stomatitis virus in cells expressing viral proteins from vectors containing cloned cDNAs. J Virol, 1990,64:2948-57.
    [116]Conzelmann KK, Cox JH, Thiel HJ. An L (polymerase)-deficient rabies virus defective interfering particle RNA is replicated and transcribed by heterologous helper virus L proteins. Virology, 1991, 184,655-63.
    [117]Calain P, Curran J, Kolakofsky D, et al. Molecular cloning of natural paramyxovirus copy-back defective interfering RNAs and their expression from DNA. Virology, 1992,191: 62-71.
    [118]Dimock K, Collins PL. Rescue of synthetic analogs of genomic RNA and replicative-intermediate RNA of human parainfluenza virus type 3. J Virol, 1993,67:2772-8.
    [119]Sidhu MS, Chan J, Kaelin K, et al. Rescue of synthetic measles virus minireplicons: measles genomic termini direct efficient expression and propagation of a reporter gene. Virology, 1995,208:800-7.
    [120]Muhlberger E, Lotfering B, Klenk HD, et al. Three of the four nucleocapsid proteins of Marburg virus, NP, VP35, and L, are sufficient to mediate replication and transcription of Marburg virus specific monocistronic minigenomes. J Virol, 1998,72:8756-64.
    [121]Muhlberger E, Weik M, Volchkov VE, et al. Comparison of the transcription and replication strategies of Marburg virus and Ebola virus by using artificial replication systems. J Virol, 1999,73:2333-42.
    [122]Dunn EF, Pritlove DC, Jin H, et al. Transcription of a recombinant bunyavirus RNA template by transiently expressed bunyavirus proteins. Virology, 1995, 211:133-43.
    [123]Lopez N, Muller R, Prehaud C, et al. The L protein of Rift Valley fever virus can rescue viral ribonucleoproteins and transcribe synthetic genome-like RNA molecules. J Virol, 1995, 69:3972-9.
    [124]Flick R, Pettersson RF. Reverse genetics system for Uukuniemi virus (Bunyaviridae): RNA polymerase I-catalyzed expression of chimeric viral RNAs. J Virol, 2001, 75, 1643-55.
    [125]Lee KJ, Novella IS, Teng MN, et al. NP and L proteins of lymphocytic choriomeningitis virus (LCMV) are sufficient for efficient transcription and replication of LCMV genomic RNA analogs. J Virol, 2000, 74:3470-77.
    [126]Honda A, Ueda K, Nagata K, et al. RNA polymerase of influenza virus : role of NP in RNA chain elongation.J Biochemistry ,1988, 104:1021-26.
    [127]Honda A, Ueda K, Nagata, K, et al. Identification of the RNA polymerase-binding site on genome RNA of influenza virus. J Biochem(Tokyo),1987, 102: 1241-49.
    [128]Schnell MJ, MebatsionT, Conzelman KK. Infectious rabies virus from cloned cDNA. EMBO J, 1994,13:4195-203.
    [129]Garcin D, Pelet T, Calain P,et al. A highly recombinogenic system for the recovery of infectious Sendai paramyxovirus from cDNA: generation of a novel copy-back nondefective interfering virus. EMBO Journal, 1995,14: 6087-94.
    [130]Lawson ND, Stillman EA, Whitt, M A, et al. Recombinant vesicular stomatitis viruses from DNA. Proc Natl Acad Sci USA, 1995,92: 4477-81.
    [131]Radecke F, Spielhofer P, Schneider H, et al. Rescue of Measles viruses from cloned DNA. EMBO J, 1995, 14:5773-84.
    [132]Schneider H, Spielhofer P, Kaelin K, et al. Rescue of measles virus using a replication-deficient vaccinia-T7 vector. J Virological Methods, 1997, 64:57-64.
    [133]Takeda M, Takeuchi K, Miyajima N, et al. Recovery of pathogenic measles virus from cloned cDNA. J Virol, 2000, 74:6643-47.
    [134]Whelan SP, Ball LA, Barr JN, et al. Efficient recovery of infectious vesicular stomatitis virus entirely from cDNA clones. Proc Natl Acad Sci USA, 1995, 92(18): 8388-92.
    [135]Kato A, Sakai Y, Shioda T, et al. Initiation of Sendai virus multiplication from transfected cDNA or RNA with negative or positive sense. Genes to Cells, 1996,1: 569-579.
    [136]Baron, M D, Barrett T. Rescue of rinderpest virus from cloned cDNA. J Virol, 1997, 71: 1265-71.
    [137]Durbin AP, Hall SL, Siew JW. et al. Recovery of infectious human parainfluenza virus type 3 from cDNA. Virology, 1997, 235(2): 323-32.
    [138]Hoffman MA, Banerjee AK. An infectious clone of human parainfluenza virus type 3. J Virol, 1997, 71:4272-7.
    [139]Haller AA, Miller T, Mitiku M, et al. Expression of the surface glycoproteins of human parainfluenza virus type 3 by bovine parainfluenza virus type 3, a novel attenuated virus vaccine vector. J Virol, 2000,74:11626-35.
    [140]Jin H, Clarke D, Zhou HZ, et al. Recombinant human respiratory syncytial virus(RSV) from cNDA and construction of subgroup A and B chimeric RSV. Virology, 1998, 251: 206-214.
    [141]Buchholz UJ, Finke S, Conzelmann KK. Generation of bovine respiratory syncytial virus (BRSV) from cDNA: BRSV NS2 is not essential for virus replication in tissue culture, and the human RSV leader region acts as a functional BRSV genome promoter. J Virol, 1999,73:251-259.
    [142]Clarke DK, Sidhu MS,Johnson JE,et al. Rescue of mumps virus from cDNA. J Virol, 2000,74:4831-4838.
    [143]Volchkov VE, Volchkova VA, Muhlberger E, et al. Recovery of infectious Ebola virus from complementary DNA: RNA editing of the GP gene and viral cytotoxicity. Science,2001, 291: 1965-1969.
    [144]He B, Paterson RG, Ward CD, et al. Recovery of infectious SV5 from cloned DNA and expression of a foreign gene. Virology, 1997, 237:249-60.
    [145]Kawano M, Kaito M, Kozuka Y, et al. Recovery of infectious human parainfluenza type 2 virus from cDNA clones and properties of the defective virus without V-specific cysteine-rich domain. Virology, 2001,284, 99-112.
    [146]Romer-Oberdorfer A, Mundt E, Mebatsion T, et al. Generation of recombinant lentogenic Newcastle disease virus from cDNA. J Virol, 1999, 80:2987-95.
    [147]Krishnamurthy S, Huang Z, Samal SK. Recovery of a virulent strain of Newcastle disease virus from cloned cDNA: expression of a foreign gene results in growth retardation and attenuation. Virology, 2000, 278:168-82.
    [148]Collins PL, Hill MG, Camargo E, et al. Production of infectious human respiratory syncytial virus from cloned cDNA confirms an essential role for the transcription elongation factor from the 5' proximal open reading frame of the M2 mRNA in gene expression and provides a capability for vaccine development. Proc Natl Acad Sci USA, 1995,92:11563-7.
    [149]Neumann G, Feldmann H, Watanabe S, et al. Reverse genetics demonstrates that proteolytic processing of the Ebola virus glycoprotein is not essential for replication in cell culture. Journal of Virology, 2002, 76:406-10.
    [150]Bridgen A, Elliott RM. Rescue of a segment negative strand RNA virus entirely from cloned complimentary DNAs. Proc Natl Acad Sci USA, 1996, 92(1): 15400-4.
    [151]Neumann G, Watanabe T, Ito H, et al. Generation of influenza A viruses entirely from cloned cDNAs. Proc Natl Acad Sci USA, 1999, 96, 9345-50.
    [152]Fodor E, Devenish L, Engelhardt OG, et al. Rescue of influenza A virus from recombinant DNA. J Virol, 1999,73:9679-82.
    [153]Wagner E, Engelhardt OG, Gruber S, et al. Rescue of recombinant Thogoto virus from cloned cDNA. 俄Introduction of foreign sequences into the genome of influenza A virus. Developments in Biological Standardization, 1994, 82:237-46.
    [155]Barclay WS, Palese P. Influenza B viruses with site-specific mutations introduced into the HA gene. J Virol, 1995, 69:1275-9.
    [156]Bergmann M, Muster T. Mutations in the nonconserved noncoding sequences of the influenza A virus segments affect viral vRNA formation. Virus Res, 1996, 44:23-31.
    [157]Zheng H, Palese P, Garcia-Sastre A. Nonconserved nucleotides at the 3' and 5' ends of an influenza A virus RNA play an important role in viral RNA replication. Virology, 1996, 217:242-51.
    [158]Murphy SK, Parks GD. Genome nucleotide lengths that are divisible by six are not essential but enhance replication of defective interfering RNAs of the paramyxovirus simian virus 5. Virology, 1997, 232:145-57.
    [159]He B, Lamb RA. Effect of inserting paramyxovirus simian virus 5 gene junctions at the HN/L gene junction : analysis of accumulation of mRNAs transcribed from rescued viable viruses. J Virol, 1999, 73:6228~34.
    
    [160]Peeters BP, Gruijthuijsen YK, de Leeuw OS, et al. Genome replication of Newcastle disease virus: involvement of the rule-of-six. Arch Virol, 2000, 145:1829-45.
    [161]Harty RN, Palese P. Mutations within noncoding terminal sequences of model RNAs of Sendai virus : influence on reporter gene expression. J Virol, 1995, 69:5128~31.
    [162]Hausmann S, Jacques JP, Kolakofsky D. Paramyxovirus RNA editing and the requirement for hexamer genome length. RNA, 1996, 2:1033-45.
    [163]Durbin AP, Siew JW, Murphy BR, et al. Minimum protein requirements for transcription and RNA replication of a minigenome of human parainfluenza virus type 3 and evaluation of the rule of six. Virology, 1997, 234:74-83.
    [164]Samal SK, Collins PL. RNA replication by a respiratory syncytial virus RNA analog does not obey the rule of six and retains a nonviral trinucleotide extension at the leader end. J Virol, 1996,70:5075-82.
    [165]Pattnaik AK, Ball LA, LeGrone A, et al. The termini of VSV DI particle RNAs are sufficient to signal RNA encapsidation, replication, and budding to generate infectious particles. Virology, 1995,206:760-4.
    [166]Martin J, Albo C, Ortin J, et al. In vitro reconstitution of active influenza virus ribonucleoprotein complexes using viral proteins purified from infected cells. J Gen Virol, 1992,73:1855-9.
    [167]Li S, Xu M, Coelingh K. Electroporation of influenza virus ribonucleoprotein complexes for rescue of the nucleoprotein and matrix genes. Virus Res, 1995,37:153-61.
    [168]Neumann G, Zobel A, Hobom G. RNA polymerase I-mediated expression of influenza viral RNA molecules. Virology, 1994,202: 477-9.
    [169]Li X, Palese P. Mutational analysis of the promoter required for influenza virus virion RNA ynthesis. J Virol, 1992, 66:4331-8.
    [170]Piccone ME, Fernandez-Sesma A, Palese P. Mutational analysis of the influenza virus vRNA romoter. Virus Res, 1993, 28: 99-112.
    [171]Fodor E, Pritlove DC, Brownlee GG. Characterization of the RNA-fork model of virion RNA in the initiation of transcription in influenza A virus. J Virol, 1995, 69: 4012-9.
    [172]Grassauer A, Egorov AY, Ferko B, et al. A host restriction-based selection system for influenza haemagglutinin transfectant viruses. J Gen Virol, 1998,79:1405-9.
    [173] Jin H, Subbarao K, Bagai S, et al. Palmitylation of the influenza virus hemagglutinin (H3) is not essential for virus assembly or infectivity. J Virol, 1996,70:1406-14.
    [174]Lin YP, Wharton SA, Martin J, et al. Adaptation of egg-grown and transfectant influenza viruses for growth in mammalian cells: selection of hemagglutinin mutants with elevated pH of membrane fusion. Virology, 1997,233: 402-10.
    [175]Castrucci MR, Kawaoka Y. Reverse genetics system for generation of an influenza A virus mutant containing a deletion of the carboxyl-terminal residue of M2 protein. J Virol, 1995,69: 2725-8.
    [176]Yasuda J, Bucher DJ, Ishihama A. Growth control of influenza A virus by M1 protein: analysis of transfectant viruses carrying the chimeric M gene. J Virol, 1994,68:8141-6.
    [177]Enami M. Improved technique to genetically manipulate influenza virus. International Conference on Negative Strand Viruses 1997, Dublin, Ireland, Abstr. 93:96.
    [178]Enami M, Enami K. Characterization of influenza virus NS1 protein by using a novel helper-virus-free reverse genetic system: J Virol, 2000, 74:5556-61.
    [179]Mena I, Vivo A, Perez E, et al. Rescue of a synthetic chloramphenicol acetyltransferase RNA into influenza virus-like particles obtained from recombinant plasmids. J Virol, 1996,70(8):5016-24.
    [180]Gomez-Puertas P, Mena I, Castillo M, et al. Efficient formation of influenza virus-like particles: dependence on the expression levels of viral proteins. J Gen Virol, 1999,80:1635-45.
    [181]Neumann G, Watanabe T, Kawaoka Y. Plasmid-driven formation of influenza virus-like particles. J Virol, 2000,74(1):547~51.
    [182]Gomez-Puertas P, Mena 1, Castillo M, et al. Efficient formation of influenza virus-like particles: dependence on the expression levels of viral proteins. J Gen Virol, 1999, 80:1635-45.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700