甲型流感病毒(H1N1)感染介导的小鼠肺免疫损伤机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
流感是一种急性呼吸道传染病,患病率高,在老年和儿童中死亡率很高,世界范围内每年都有大量的流感死亡病例报道,流感对世界公共卫生已经构成了严重的威胁。特别是2009年世界范围内暴发的猪源性甲型流感病毒,传染性强,波及世界213个国家和地区,造成了17483例死亡病例,拉响了世界范围内防控流感的警钟,近期关于猪源性甲型流感病毒对达菲耐药的报道,使流感病毒的防控工作更为艰难。
     从临床学角度分析,甲型流感病毒是流感病毒三个亚型中最为重要的亚型,属正粘病毒,根据其表面血凝素(HA)及神经氨酸酶(NA)分子的抗原性不同,分别有16(HA)与9(NA)个亚型。病毒基因组的点突变或基因节段重组将会产生新的变异株,从而引起大范围的传染或暴发性流行。上个世纪共出现四次流感大流行,最为严重的是1918年的大流感,造成了世界范围内5千多万人死亡。到目前为止,每年都有一定范围内流感暴发流行的病例报道。因此,深入研究甲型流感病毒重症感染的致病机制,对于选择抗病毒治疗及控制潜在的流感暴发流行显得至关重要。流感病毒致死性感染的小鼠模型研究发现,失控的及异常激活的天然免疫应答,和感染过程中大量的巨噬细胞与中性粒细胞快速招募至肺局部是高致病禽流感致病的主要因素,此外巨噬细胞及中性粒细胞与肺部增加的细胞因子及趋化因子有关,而升高的细胞因子水平被认为是介导甲型流感病毒病症的主要原因,亦是决定高致病性禽流感病毒致死性感染症状的主要原因。然而,抑制细胞因子应答并不能保护机体免于致死性感染,感染早期清除巨噬细胞及中性粒细胞对疾病的结局亦无显著影响,这些研究提示巨噬细胞及中性粒细胞在甲型流感病毒重症感染病例中的复杂生物学效应。另外,巨噬细胞及中性粒细胞本身也是病毒感染的靶细胞,巨噬细胞及中性粒细胞被直接感染可能会导致包括抗体应答在内的获得性免疫应答不能够被有效激活。
     本研究主要探讨甲型流感病毒感染介导的小鼠肺免疫损伤机制,对流感病毒感染Balb/c小鼠后免疫损伤的发生、发展及转归进行动态观察,分别从细胞与分子层面揭示流感病毒感染介导的免疫损伤机制,探讨这些免疫细胞与分子在感染过程中发挥的双重作用,同时观察比较不同毒力病毒感染过程中介导免疫损伤的主要细胞与免疫分子差异。研究内容共分为三部分:1、甲型流感病毒A/PR/8/34(H1N1)感染介导的小鼠肺免疫损伤机制初步研究,分三节(1)甲型流感病毒A/PR/8/34(H1N1)感染Balb/C小鼠肺损伤动物模型的建立;(2)甲型流感病毒A/PR/8/34(H1N1)感染介导的小鼠肺免疫损伤机制初探;(3)甲型流感病毒A/PR/8/34(H1N1)感染介导的肺损伤过程中粘膜及二级淋巴器官的免疫应答分析;2、甲型流感病毒A/Beijing/501/2009(H1N1)感染介导的小鼠肺损伤机制初步研究,分三节(1)甲型流感病毒A/Beijing/501/2009(H1N1)感染Balb/C小鼠肺损伤动物模型的建立;(2)甲型流感病毒A/Beijing/501/2009(H1N1)感染介导的小鼠肺损伤机制初探;(3)甲型流感病毒A/Beijing/501/2009(H1N1)感染介导的肺损伤过程中粘膜及二级淋巴器官的免疫应答分析;3、甲型流感病毒A/Beijing/501/2009(H1N1)与A/PR/8/34(H1N1)感染介导的小鼠肺损伤机制差异分析。
     研究结果:1、两株病毒感染后小鼠体重变化,死亡率及肺病理变化说明,两株病毒感染介导的小鼠肺损伤模型建立成功;病毒感染后早期即可在鼻及肺中检测到病毒复制,至感染末期,病毒呈持续性复制;2、两株病毒感染造成了小鼠肺损伤的同时,介导了复杂的应答变化,无论是病毒感染早期还是晚期,均检测到了大量的巨噬细胞与中性粒细胞的累积,统计学分析结果显示,中性粒细胞与肺病毒载量、肺及血清中抗体应答密切相关,肺中抗体应答与肺中病毒载量显著相关;病毒感染后巨噬细胞及中性粒细胞的浸润,促进了肺细胞因子应答,细胞因子应答参与调节抗体应答;3、病毒感染过程中,肺部除检测到较强的抗体应答,也检测到了较强的细胞因子分泌细胞应答及抗体分泌细胞应答,这两种应答反应共同调节肺抗体应答的强度及维持的时间;4、两株病毒感染小鼠肺损伤的同时,检测到了粘膜局部及二级淋巴器官产生了T、B细胞应答反应,病变累及肺器官的同时,引起了全身免疫系统的应答反应,共同参与机体的免疫防御;5、两株病毒感染小鼠半数致死剂量,体重变化及死亡率等指标反应了两株病毒在小鼠上的毒力不同,其介导肺免疫损伤过程中诱发的各种应答反应规律不尽相同,总的来说,A/Beijing/501/2009(H1N1)病毒诱发的免疫应答反应早于A/PR/8/34(H1N1)病毒,强度亦强于后者; 6、A/Beijing/501/2009(H1N1),A/California/04/2009(H1N1)及A/PR/8/34三株病毒蛋白序列比对分析结果显示,在已知的重要功能性位点及包括受体结合区域在内的未知功能性发生了突变,这些突变与病毒的毒力直接相关。
     结论:甲型流感病毒感染介导的小鼠肺损伤过程是一个多因素参与的复杂过程,其中病毒持续复制是造成损伤的主要因素之一,肺中大量的巨噬细胞及中性粒细胞的累积在感染的早期是损伤的主要因素,随着疾病的进程,中性粒细胞与抗体协同作用,共同促进病毒的清除及疾病的恢复;病毒引发的肺局部损伤性应答与全身系统免疫应答的激活密不可分,肺局部的应答参与系统性应答。
Influenza is an epidemic, acute respiratory disease that has a high rate of mortality, especially among the elderly and children, and a large number of deaths due to influenza are reported every year all over the world. In 2009, a novel swine-origin influenza virus capable of rapid human transmission was reported. As of 4 April 2010, worldwide more than 213 countries and overseas territories or communities have reported laboratory confirmed cases of pandemic influenza H1N1 2009, including over 17700 deaths. Surveillance network has been build worldwide since March 2009. The recent report on the drug resistance to Oseltamivir phosphate capsules made the prevention and control of pandemic influenza more difficult.
     Influenza A virus is the most important among the three types of the virus from a clinical point of view. Influenza A viruses belong to the family Orthomyxoviridae. On the basis of the antigenicity of their haemagglutinin (HA) and neuraminidase (NA) molecules, they are classified into 16 HA subtypes (H1–H16) and 9 NA subtypes (N1–N9). The point mutations and reassortment events of the viral genomes contribute to the emergence of new variants or strains with epidemic or pandemic potential. Influenza A viruses have caused several pandemics during the last century, and continue to cause annual epidemics. The pandemic of 1918-1919 killed as many as 50 million people worldwide. Understanding of the mechanisms of increased pathogenicity of fatal influenza A viral infection is thus critical to optimize the antiviral treatment strategy and control the potential pandemic. The uncontrolled and aberrant activation of the innate immune system had been implicated in the mice model of fatal influenza A viral infection. A significantly rapid cell recruitment of macrophages and neutrophils into the lungs were assumed to play a role in the pathogenesis associated with H5N1 highly pathogenic avian influenza virus infection (HPAI). In addition, macrophages and neutrophils were associated with increased secretion and enrollment of some cytokine and chemokines, and the increased levels of cytokines are suggested to mediate influenza A infection signs and may play a role in the severe symptoms of fatal cases of HPAI H5N1 influenza virus infection. However, inhibition of the cytokine response was found not to protect against the lethal influenza A infection, and neutrophil or macrophage depletion initiated after infection in the early stage were also found not to have a significant effect on the disease outcome. These findings suggested complicated biological effect of macrophages and neutrophils in the fatal influenza A viral infection. In addtion, innate immune cells such as macrophages and neutrophils are the targets of influenza A viruses. The direct infection of macrophages and neutrophils may contribute to an inability to mount an adaptive immune response, such as antibody stimulation.
     In this study, the mechanism of immunopathology mediated by influenza A virus infection was carried out. To discover the role of these complicated factors during the infection, and evaluate the cellular and molecular discrepancy of different virulence virus, the initiation, development and outcome of the pulmonary immunopathology was investigated. Three parts were included in this study. On the first part, immunopatholgy on mice mediated by A/PR/8/34 (H1N1) influenza virus was studied. This part included: (1) The establishment and evaluation of mouse model for immunopathology on mice with influenza A/PR/8/34 (H1N1) virus infection. (2) Primary study on the mechanism of immunopathology on mice mediated by influenza A/PR/8/34 (H1N1) virus. (3) The immune response of mucosal and secondary lymphoid tissue during the pulmonary injury of mice with influenza A/PR/8/34 (H1N1) virus infection. On the second part, preliminary research on the mechanism of immunopothology on mice mediated by A/Beijing/501/2009 (H1N1) virus was studied. This part included: (1) The establishment and evaluation of mouse model for immunopathology on mice with influenza A/Beijing/501/2009 (H1N1) virus infection. (2) Primary study on the mechanism of immunopathology on mice mediated by influenza A/Beijing/501/2009 (H1N1) virus. (3) The immune response of mucosal and secondary lymphocyte tissue during the pulmonary injury of mice with influenza A/Beijing/501/2009 (H1N1) virus infection. In the third part, we compared the discrepancy pulmonary immune injury induced by A/Beijing/501/2009 (H1N1) and A/PR/8/34 (H1N1) virus infection.
     Results are as follows. First, we successfully established the mouse model for immunopathology on mice with two strains of influenza A virus by monitoring the weight lost, death ratio and histopathology of lung. Viral replication of two strains of influenza A virus in nasal and lung were detected in the early and late stage of infection. Second, complicated immune response was companied with the immunopathology during the infection. Large number of macrophage and neutrophils were accumulated both in the early stage and later stage of infection. Statics analysis shows that there is close correlation between the neutrophils with the viral load, serum and pulmonary antibody response. Close correlation also be found between the antibody responses with the viral load in lung. The infiltration of macrophage and neutrophils promote the cytokine response thereby implicated in the antibody response. Third, beside the robust antibody response, cytokine secreting cells (CSCs) and antibody secreting cells (ASCs) response which regulates the kinetics and magnitude of antibody response were also detected in lung of mice post infection. Fourth, cellular and humoral response in mucosal and second lymphoid organs cooperate with the immune response in lung contributes to the systemic protective response during the infection. Fifth, the different virulence of two influenza A virus strains is demonstrated by lethal dose fifty (LD50), weight lost and death ratio on mice, furthermore, the response initiated by two strains is not similar. Taken together, the response induced by A/Beijng/501/2009 (H1N1) is earlier and stronger than that induced by A/PR/8/34 (H1N1) virus. Sixth, the alignment of the protein sequency among A/Beijing/501/2009(H1N1), A/California/04/2009 and A/PR/8/34(H1N1) virus shows that there are mutations located either in known molecular determinants that confer pathogenicity and antiviral drug resistance and the glycan receptor-binding sites and antigenic loops which should be further characterized of their functional determinants.
     In conclusion, the process of immunopathology mediated by influenza A virus is multifaceted and highly complicated, involving different arms of the host response, all of which appear to be required for efficient virus clearance. The continuous replication of viral and accumulation of macrophage and neutophils in lung in the early stage of infection are the main reason for immune-mediated pathology. In the late stage of infection, neutrophils cooperate with the antibody response contributed to control the viral burden. Local pulmonary pathologic response is connected with the initiation of system immune response.
引文
[1] Neumann GT. Noda and Kawaoka Y. Emergence and pandemic potential of swine-origin H1N1 influenza virus. Nature. 2009, 459 (7249): 931-9.
    [2] Chan MC, Cheung CY, Chui WH et al. Proinflammatory cytokine responses induced by influenza A (H5N1) viruses in primary human alveolar and bronchial epithelial cells. Respir Res. 2005, 6 (1): 135.
    [3] Hsieh YC, Wu TZ, Liu DP et al. Influenza pandemics: past, present and future. J Formos Med Assoc. 2006, 105 (1): 1-6.
    [4] De Jong MD, Simmons CP, Thanh TT et al. Fatal outcome of human influenza A (H5N1) is associated with high viral load and hypercytokinemia. Nat Med. 2006, 12 (10): 1203-1207.
    [5] Szretter KJ, Gangappa S, Lu X et al. Role of host cytokine responses in the pathogenesis of avian H5N1 influenza viruses in mice. J Virol. 2007, 81 (6): 2736-2744.
    [6] Kobasa D, Jones SM, Shinya K et al. Aberrant innate immune response in lethal infection of macaques with the 1918 influenza virus. Nature. 2007, 445 (7125): 319-323.
    [7] Sun JR. Madan CL, Karp and Braciale TJ. Effector T cells control lung inflammation during acute influenza virus infection by producing IL-10. Nat Med. 2009, 15(3): 277-84.
    [8] Brown DM, Román E, Swain SL. CD4 T cell responses to influenza infection. Semin Immunol. 2004, 16 (3): 171-7.
    [9] McKimm-Breschkin JL. A simplified plaque assay for respiratory syncytial virus—direct visualization of plaques without immunostaining. Journal of Virological Methods 2004, 120(1): 113–117.
    [10] Payne AF, Binduga-Gajewska I, Kauffman EB et al.Quantitation of ?aviviruses by ?uorescent focus assay. Journal of Virological Methods.2006, 134(1-2): 183–189.
    [11] Wei Z, McEvoy M, Razinkov V et al. Schenerman. Biophysical characterization of in?uenza virus subpopulations using ?eld ?ow fractionation and multiangle light scattering: Correlation of particle counts, size distribution and infectivity. Journal of Virological Methods. 2007, 144(1-2): 122–132.
    [12] Jia, N, Li C, Liu YX et al. Lower cellular immune responses to influenza A (H3N2) in the elderly. J Med Virol. 2009, 81(8): 1471-1476.
    [13] Manz RA, Arce S, Cassese G et al. Humoral immunity and long-lived plasma cells. Curr Opin Immunol. 2002, 14(4): 517-21.
    [14] Kutzler MA, Robinson TM, Chattergoon MA et al. Coimmunization with an optimized IL-15 plasmid results in enhanced function and longevity of CD8 T cells that are partially independent of CD4 T cell help. J Immunol. 2005, 175(1): 112–123.
    [15] Jelley-Gibbs DM, Dibble JP, Brown DM et al. Persistent depots of influenza antigen fail to induce a cytotoxic CD8 T cell response. J Immunol. 2007, 178(12): 7563-7570.
    [16] Shin-ichi Tamura, Takeski Tanumoto and Takeshi Kurata. Mechanisms of broad cross-protection provided by influenza virus infection and their application to vaccines. Jpn JInfect Dis. 2005, 58(4): 195-207.
    [17] Mosmann TR, Cherwinski H, Bond MW et al. Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins, 1986. J Immunol. 2005, 175(1): 5—14.
    [18] Fazilleau N, McHeyzer-Williams LJ, McHeyzer-Williams MG. Local development of effector and memory T helper cells. Curr Opin Immunol. 2007, 19(3): 259—267
    [19] Kash JC, Tumpey TM, Proll SC et al. Genomic analysis of increased host immune and cell death responses induced by 1918 influenza virus. Nature. 2006, 443(7111): 578-581.
    [20] Suzuki K, Okada H, Itoh T et al. Association of increased pathogenicity of Asian H5N1 highly pathogenic avian influenza viruses in chickens with highly efficient viral replication accompanied by early destruction of innate immune responses. J Virol. 2009, 83(15): 7475-7486.
    [21] Perrone LA, Plowden JK, Garcia-Sastre A et al. H5N1 and 1918 pandemic influenza virus infection results in early and excessive infiltration of macrophages and neutrophils in the lungs of mice. PLoS Pathog. 2008, 4(8): e1000115
    [22] Fujisawa H. Neutrophils play an essential role in cooperation with antibody in both protection against and recovery from pulmonary infection with influenza virus in mice. J Virol. 2008, 82(6): 2772-2783.
    [23] Hashimoto Y, Moki T, Takizawa T et al. Evidence for phagocytosis of influenza virus-infected, apoptotic cells by neutrophils and macrophages in mice. J Immunol. 2007, 178(4): 2448-2457.
    [24] Kobasa D, Takada A, Shinya K,et al. Enhanced virulence of influenza A viruses with the haemagglutinin of the 1918 pandemic virus. Nature. 2004, 431(7009): 703-707.
    [25] Korteweg C, Gu J. Pathology, molecular biology, and pathogenesis of avian influenza A (H5N1) infection in humans. Am J Pathol. 2008, 172(5): 1155-1170.
    [26] Jones SA. Directing transition from innate to acquired immunity: defining a role for IL-6. J Immunol. 2005, 175(6): 3463-3468.
    [27] Hayden FG, Fritz R, Lobo MC et al. Local and systemic cytokine responses during experimental human influenza A virus infection. Relation to symptom formation and host defense. J Clin Invest. 1998, 101(3): 643-649.
    [28] Skoner DP, Gentile DA, Patel A et al. Evidence for cytokine mediation of disease expression in adults experimentally infected with influenza A virus. J Infect Dis. 1999, 180(1): 10-14.
    [29] Cheung CY, Poon LL, Lau AS et al. Induction of proinflammatory cytokines in human macrophages by influenza A (H5N1) viruses: a mechanism for the unusual severity of human disease? Lancet. 2002, 360(9348): 1831-1837.
    [30] de Jong MD, Bach VC, Phan TQ et al. Fatal avian influenza A (H5N1) in a child presenting with diarrhea followed by coma. N Engl J Med. 2005, 352(7): 686-691.
    [31] Lee DC, Cheung CY, Law AH et al. p38 mitogen-activated protein kinase-dependent hyperinduction of tumor necrosis factor alpha expression in response to avian influenza virus H5N1. J Virol. 2005, 79(16): 10147-10154.
    [32] Xu T, Qiao J, Zhao L et al. Acute respiratory distress syndrome induced by avian influenzaA (H5N1) virus in mice. Am J Respir Crit Care Med.2006, 174(9): 1011-1017.
    [33] Salomon R, Hoffmann E and Webster RG. Inhibition of the cytokine response does not protect against lethal H5N1 influenza infection. Proc Natl Acad Sci U S A. 2007, 104(30): 12479-12481.
    [34] Couper KN, Blount DG and Riley EM. IL-10: the master regulator of immunity to infection. J Immunol. 2008, 180(9): 5771-5777.
    [35] Pestka S, Krause CD, Sarkar D et al. Interleukin-10 and related cytokines and receptors. Annu Rev Immunol. 2004, 22: 929-979.
    [36] Seo SH, Webby R, Webster RG. No apoptotic deaths and different levels of inductions of inflammatory cytokines in alveolar macrophages infected with influenza viruses. Virology. 2004, 329(2): 270-279.
    [37] Tumpey TM, García-Sastre A, Taubenberger JK,et al. Pathogenicity of influenza viruses with genes from the 1918 pandemic virus: functional roles of alveolar macrophages and neutrophils in limiting virus replication and mortality in mice. J Virol. 2005, 79(23): 14933-14944.
    [38] Luke TC, Kilbane EM, Jackson JL et al. Meta-analysis: convalescent blood products for Spanish influenza pneumonia: a future H5N1 treatment? Ann Intern Med. 2006, 145(8): 599-609.
    [39] Khurana S, Suguitan AL Jr, Rivera Y et al. Antigenic fingerprinting of H5N1 avian influenza using convalescent sera and monoclonal antibodies reveals potential vaccine and diagnostic targets. PLoS Med. 2009, 6(4): e1000049.
    [40] Simmons CP, Bernasconi NL, Suguitan AL et al. Prophylactic and therapeutic efficacy of human monoclonal antibodies against H5N1 influenza. PLoS Med.2007, 4(5): e178.
    [41] Throsby M, van den Brink E, Jongeneelen M et al. Heterosubtypic neutralizing monoclonal antibodies cross-protective against H5N1 and H1N1 recovered from human IgM+ memory B cells. PLoS One. 2008, 3(12): e3942.
    [42] Zhou B, Zhong N and Guan Y. Treatment with convalescent plasma for influenza A (H5N1) infection. N Engl J Med.2007, 357(14): 1450-1451.
    [43] Beigel J and Bray M. Current and future antiviral therapy of severe seasonal and avian influenza. Antiviral Res. 2008, 78(1): 91-102.
    [44] WHO, 2009a Pandemic (H1N1) 2009—Update http://www.who.int/csr/don/ 208 2009 07 27/en/index.html.
    [45] Peiris JS, Poon LL, Guan Y. Emergence of a novel swine-origin influenza (S-OIV) H1N1 virus in humans. J Clin Virol. 2009, 45(3): 169-173.
    [46] WHO, 2009. CDC Protocol of Realtime RTPCR for Influenza A(H1N1), 28 April 2009., 212 http://www.who.int/csr/resources/publications/swineflu/ SwineH1Assay-2009 20090430.pdf.
    [47] Lamb RA, Krug RM. Orthomyxoviridae: the viruses and their replication. Fields Virology, 2001, 4(1): 1487–1531.
    [48] Potter CW. A history of influenza. J Appl Microbiol 2001, 91: 572–579.
    [49] Shinya K, Ebina M, Yamada S et al. Avian flu: influenza virus receptors in the human airway. Nature. 2006, 440(7083): 435–436.
    [50] Doherty PC, Turner SJ, Webby RG et al. Influenza and the challenge for immunology. Nat Immunol.2006, 7(5): 449–455.
    [51] Dawood FS, Jain S, Finelli L et al. Emergence of a novel swine-origin influenza A (H1N1) virus in humans. N Engl J Med. 2009, 360(25): 2605-2615.
    [52] Lange E, Kalthoff D, Blohm U et al. Pathogenesis and transmission of the novel swine origin influenza virus A/H1N1 after experimental infection of pigs. J Gen Virol. 2009, 90(9), 2119–2123.
    [53] Swine Influenza A (H1N1) Infection in Two Children--Southern California. CDC. Surveill Summ. 2009, 58:400-402.
    [54] Swine-Origin Influenza A (H1N1) Virus Infections in a School--New York City. Surveill Summ. 2009, 58(Dispatch):1-3.
    [55] Update: Infections With a Swine-Origin Influenza A (H1N1) Virus --- United States and Other Countries. Surveill Summ. 2009, 58:431-433.
    [56] Kumar S, Chusid MJ, Willoughby RE et al. Introduction of a novel swine-origin influenza A (H1N1) virus into Milwaukee, Wisconsin in 2009. Viruses.2009, 1(1): 72-83.
    [57] Tate MD, Brooks AG, Reading PC. The role of neutrophils in the upper and lower respiratory tract during influenza virus infection of mice. Respir Res. 2008, 9: 57.
    [58] Baumgarth N, Kelso A. Functionally distinct T cells in three compartments of the respiratory tract after influenza virus infection. Eur J Immunol 1996, 26(9): 2189– 2197.
    [59] Doherty PC, Topham DJ, Tripp RA et al. Effector CD4+ and CD8+ T-cell mechanisms in the control of respiratory virus infections. Immunol Rev.1997, 159: 105–117.
    [60] Flynn KJ, Belz GT, Altman JD et al. Virus-specific CD8+ T cells in primary and secondary influenza pneumonia. Immunity. 1998, 8(6): 683–691.
    [61] Shimomura E, Suzuki F, Ishida N. Characterization of cells infiltrating the lungs of x-irradiated and nude mice after influenza virus infection. Microbiol Immunol. 1982, 26(2): 129–138.
    [62] Wells MA, Albrecht P, Ennis FA. Recovery from a viral respiratory infection. I. Influenza pneumonia in normal and T-deficient mice. J Immunol. 1981, 126(3): 1036–1041.
    [63] Bruder D, Srikiatkhachorn A, Enelow RI. Cellular immunity and lung injury in respiratory virus infection. Viral Immunol 2006, 19(2): 147–155.
    [64] Peper RL, Van Campen H. Tumor necrosis factor as a mediator of inflammation in influenza A viral pneumonia. Microb Pathog 1995, 19(3): 175–183.
    [65] Hussell T, Pennycook A, Openshaw PJ. Inhibition of tumor necrosis factor reduces the severity of virus-specific lung immunopathology. Eur J Immunol 2001, 31(9): 2566–2573.
    [66] Coleclough C, Sealy R, Surman S,et al. Respiratory vaccination of mice against influenza virus: dissection of T- and B-cell priming functions. Scand J Immunol. 2005, 62 (1): 73-83.
    [67] Daniels MJ, Selgrade MK, Doerfler D, et al. Kinetic profile of influenza virus infection in three rat strains. Comp Med. 2003, 53(3):293-298.
    [68] van der Laan JW, Herberts C, Lambkin-Williams R, et al. Animal models in influenza vaccine testing. Expert Rev Vaccines.2008, 7(6):783-793.
    [69] Vahlenkamp TW, Harder TC, Giese M,et al. Protection of cats against lethal influenza H5N1 challenge infection. J Gen Virol. 2008, 89(Pt 4):968-974.
    [70] Giese M, Harder TC, Teifke JP, et al. Experimental infection and natural contact exposure of dogs with avian influenza virus (H5N1). Emerg Infect Dis.2008, 14(2):308-310.
    [71] Baskin CR, Garcia-Sastre A, Tumpey TM, et al. Integration of clinical data, pathology, and cDNA microarrays in influenza virus-infected pigtailed macaques (Macaca nemestrina). J Virol.2004, 78(19):10420-10432.
    [72] Svitek N, Rudd PA, Obojes K, et al. Severe seasonal influenza in ferrets correlates with reduced interferon and increased IL-6 induction.Virology. 2008, 376(1):53-59.
    [73] Davies A, Jones D, Bailey M, et al. Extracorporeal Membrane Oxygenation for 2009 Influenza A(H1N1) Acute Respiratory Distress Syndrome.2009 Jama 302(17):1888-1895.
    [74] Belser JA, Wadford DA, Pappas C, et al. Pathogenesis of pandemic influenza A (H1N1) and triple-reassortant swine influenza A (H1) viruses in mice. J Virol.2009, 84(9):4194-4203.
    [75] Itoh Y, Shinya K, Kiso M, et al. In vitro and in vivo characterization of new swine-origin H1N1 influenza viruses. Nature.2009, 460(7258):1021-1025.
    [76] Maines TR, Jayaraman A, Belser JA, et al. Transmission and pathogenesis of swine-origin 2009 A(H1N1) influenza viruses in ferrets and mice. Science.2009, 325(5939):484-487.
    [77] Munster VJ, de Wit E, van den Brand JM, et al. Pathogenesis and transmission of swine-origin 2009 A(H1N1) influenza virus in ferrets. Science. 2009, 325(5939):481-483.
    [78] Osterlund P, Pirhonen J, Ikonen N, et al. Pandemic H1N1 2009 influenza A virus induces weak cytokine responses in human macrophages and dendritic cells and is highly sensitive to the antiviral actions of interferons. J Virol.2010, 84(3):1414-1422.
    [79] Woo PC, Tung ET, Chan KH, et al. Cytokine profiles induced by the novel swine-origin influenza A/H1N1 virus: implications for treatment strategies. J Infect Dis.2010, 201(3):346-353.
    [80] Kishimoto T. Interleukin-6: from basic science to medicine--40 years in immunology. Annu Rev Immunol.2005, 23:1-21.
    [81] Cash H, Relle M, Menke J, et al. Interleukin 6 (IL-6) deficiency delays lupus nephritis in MRL-Faslpr mice: the IL-6 pathway as a new therapeutic target in treatment of autoimmune kidney disease in systemic lupus erythematosus. J Rheumatol.2010, 37(1):60-70.
    [82] Yue C, You X, Zhao L, et al. The effects of adalimumab and methotrexate treatment on peripheral Th17 cells and IL-17/IL-6 secretion in rheumatoid arthritis patients. Rheumatol Int.2009.
    [83] Bermejo-Martin JF, Ortiz de Lejarazu R, et al. Th1 and Th17 hypercytokinemia as early host response signature in severe pandemic influenza. Crit Care.2009, 13(6):R201.
    [84] Giamarellos-Bourboulis EJ, Raftogiannis M, Antonopoulou A, et al. Effect of the novel influenza A (H1N1) virus in the human immune system. PLoS One.2009, 4(12):e8393.
    [85] Chen Z, Laurence A, O'Shea JJ. Signal transduction pathways and transcriptional regulation in the control of Th17 differentiation. Semin Immunol.2007, 19(6):400-408.
    [86] Crowe CR, Chen K, Pociask DA, Alcorn JF, et al. Critical role of IL-17RA in immunopathology of influenza infection. J Immunol.2009, 183(8):5301-5310.
    [87] Kawashima H, Go S, Kashiwagi Y, et al. Cytokine profiles of suction pulmonary secretions from children infected with pandemic influenza A(H1N1) 2009. Crit Care. 2010, 14(2):411.
    [88] To KK, Hung IF, Li IW, et al. Delayed clearance of viral load and marked cytokine activation in severe cases of pandemic H1N1 2009 influenza virus infection. Clin Infect Dis.2010, 50(6):850-859.
    [89] Sutton C, Brereton C, Keogh B, et al. A crucial role for interleukin (IL)-1 in the induction of IL-17-producing T cells that mediate autoimmune encephalomyelitis. J Exp Med.2006, 203(7):1685-1691.
    [90] Julkunen I, Sareneva T, Pirhonen J, et al. Molecular pathogenesis of influenza A virus infection and virus-induced regulation of cytokine gene expression. Cytokine Growth Factor Rev. 2001, 12(2-3):171-180.
    [91] Fedson DS. Confronting an influenza pandemic with inexpensive generic agents: can it be done? Lancet Infect Dis.2008, 8(9):571-576.
    [92] Dawson TC, Beck MA, Kuziel WA et al. Contrasting effects of CCR5 and CCR2 deficiency in the pulmonary inflammatory response to influenza A virus. Am J Pathol 2000, 156(6): 1951–1959.
    [93] Shaw MW, Arden NH, Maassab HF. New aspects of influenza viruses. Clin Microbiol Rev 1992, 5(1): 74–92.
    [94] Moskophidis D, Kioussis D. Contribution of virus-specific CD8+ cytotoxic T cells to virus clearance or pathologic manifestations of influenza virus infection in a T cell receptor transgenic mouse model. J Exp Med.1998, 188(2): 223–232
    [95] Bender BS, Taylor SF, Zander DS et al. Pulmonary immune response of young and aged mice after influenza challenge. J Lab Clin Med 1995, 126(2): 169–177.
    [96] Enelow RI, Mohammed AZ, Stoler MH et al. Structural and functional consequences of alveolar cell recognition by CD8(+) T lymphocytes in experimental lung disease. J Clin Invest 1998, 102(9): 1653–1661.
    [97] Hsieh SM, Chang SC. Insufficient perforin expression in CD8+ T cells in response to hemagglutinin from avian influenza (H5N1) virus. J Immunol. 2006, 176(8):4530-4533.
    [98] Wiley JA, Cerwenka A, Harkema JR,et al. Production of interferon-gamma by influenza hemagglutinin-specific CD8 effector T cells influences the development of pulmonary immunopathology. Am J Pathol. 2001, 158(1):119-130.
    [99] Lee HY, Topham DJ, Park SY,et al. Simulation and prediction of the adaptive immune response to influenza A virus infection. J Virol.2009, 83(14):7151-7165.
    [100] Wolf AI, Buehler D, Hensley SE,et al. Plasmacytoid dendritic cells are dispensable during primary influenza virus infection. J Immunol.2009, 182(2):871-879.
    [101] Miriam Shapiro-Shelef, Kathryn Calame. Regulation of plasma—cell development. [J] Nature Publishing Group. 2005, 3(5): 230-242.
    [102] Garten RJ, Davis CT, Russell CA, et al. Antigenic and genetic characteristics ofswine-origin 2009 A(H1N1) influenza viruses circulating in humans. Science.2009, 325(5937):197-201.
    [103] Massin P, van der Werf S, Naffakh N. Residue 627 of PB2 is a determinant of cold sensitivity in RNA replication of avian influenza viruses. J Virol. 2001, 75(11):5398-5404.
    [104] Soundararajan V, Tharakaraman K, Raman R, et al. Extrapolating from sequence--the 2009 H1N1 'swine' influenza virus. Nat Biotechnol.2009, 27(6):510-513.
    [1] Chan MC, Cheung CY, Chui WH, et al. Proinflammatory cytokine responses induced by influenza A (H5N1) viruses in primary human alveolar and bronchial epithelial cells. Respir Res. 2005,6(1):135.
    [2] Hsieh YC, Wu TZ, Liu DP,et al. Influenza pandemics: past, present and future. J Formos Med Assoc. 2006;105(1):1-6.
    [3] de Jong MD, Simmons CP, Thanh TT,et al. Fatal outcome of human influenza A (H5N1) is associated with high viral load and hypercytokinemia. Nat Med. 2006;12(10):1203-1207.
    [4] Peiris JS, Yu WC, Leung CW,et al. Re-emergence of fatal human influenza A subtype H5N1 disease. Lancet. 2004;363(9409):617-9.
    [5] To KF, Chan PK, Chan KF,et al. Pathology of fatal human infection associated with avian influenza A H5N1 virus. J Med Virol. 2001;63(3):242-6.
    [6] Mou SS, Nakagawa TA, Riemer EC,et al. Hemophagocytic lymphohistiocytosis complicating influenza A infection. Pediatrics. 2006;118(1):216-219.
    [7] Kobasa D, Takada A, Shinya K,et al. Enhanced virulence of influenza A viruses with the haemagglutinin of the 1918 pandemic virus. Nature. 2004;431(7009):703-7.
    [8] Korteweg C, Gu J. Pathology, molecular biology, and pathogenesis of avian influenza A (H5N1) infection in humans. Am J Pathol. 2008;172(5):1155-70.
    [9] Ostler T, Davidson W, Ehl S. Virus clearance and immunopathology by CD8(+) T cells during infection with respiratory syncytial virus are mediated by IFN-gamma. Eur J Immunol. 2002;32(8):2117-2123.
    [10] Bruder D, Srikiatkhachorn A, Enelow RI. Cellular immunity and lung injury in respiratory virus infection. Viral Immunol. 2006;19(2):147-55.
    [11] Zhang WJ, Sarawar S, Nguyen P, Lethal synergism between influenza infection and staphylococcal enterotoxin B in mice. J Immunol. 1996;157(11):5049-5060
    [12] Schmitz N, Kurrer M, Bachmann MF,et al. Interleukin-1 is responsible for acute lung immunopathology but increases survival of respiratory influenza virus infection. J Virol. 2005;79(10):6441-6448.
    [13] Neumann G, Kawaoka Y. Host range restriction and pathogenicity in the context of influenza pandemic. Emerg Infect Dis. 2006;12(6):881-6.
    [14] Lipatov AS, Andreansky S, Webby RJ,et al. Pathogenesis of Hong Kong H5N1 influenzavirus NS gene reassortants in mice: the role of cytokines and B- and T-cell responses. J Gen Virol. 2005;86(Pt 4):1121-30.
    [15] Snelgrove RJ, Edwards L, Rae AJ,et al. An absence of reactive oxygen species improves the resolution of lung influenza infection. Eur J Immunol. 2006;36(6):1364-73.
    [16] Rosenblum MD, Olasz E, Woodliff JE,et al. CD200 is a novel p53-target gene involved in apoptosis-associated immune tolerance. Blood. 2004;103(7):2691-8.
    [17] Kaufmann A, Salentin R, Meyer RG,et al. Defense against influenza A virus infection: essential role of the chemokine system. Immunobiology. 2001;204(5):603-13.
    [18] Seo SH, Webby R, Webster RG. No apoptotic deaths and different levels of inductions of inflammatory cytokines in alveolar macrophages infected with influenza viruses. Virology. 2004;329(2):270-9.
    [19] Tumpey TM, García-Sastre A, Taubenberger JK,et al. Pathogenicity of influenza viruses with genes from the 1918 pandemic virus: functional roles of alveolar macrophages and neutrophils in limiting virus replication and mortality in mice. J Virol. 2005;79(23):14933-44.
    [20] Wiley JA, Cerwenka A, Harkema JR,et al. Production of interferon-gamma by influenza hemagglutinin-specific CD8 effector T cells influences the development of pulmonary immunopathology. Am J Pathol. 2001;158(1):119-30.
    [21] Hsieh SM, Chang SC. Insufficient perforin expression in CD8+ T cells in response to hemagglutinin from avian influenza (H5N1) virus. J Immunol. 2006;176(8):4530-3.
    [22] Small BA, Dressel SA, Lawrence CW,et al. CD8(+) T cell-mediated injury in vivo progresses in the absence of effector T cells. J Exp Med. 2001;194(12):1835-46.
    [23] Humphreys IR, Walzl G, Edwards L,et al. A critical role for OX40 in T cell-mediated immunopathology during lung viral infection. J Exp Med. 2003;198(8):1237-42.
    [24] Brown DM, Román E, Swain SL. CD4 T cell responses to influenza infection. Semin Immunol. 2004;16(3):171-7.
    [1] Heil F, Hemmi H, Hochrein H,et al. Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science. 2004; 303(5663):1526-9.
    [2] Yoshikawa T, Matsuo K, Matsuo K,et al. Total viral genome copies and virus-Ig complexes after infection with influenza virus in the nasal secretions of immunized mice. J Gen Virol. 2004;85(8):2339-46.
    [3] Engelhardt OG, Sirma H, Pandolfi PP,et al. Mx1 GTPase accumulates in distinct nuclear domains and inhibits influenza A virus in cells that lack promyelocytic leukaemia protein nuclear bodies. J Gen Virol. 2004;85(Pt 8):2315-26.
    [4] Lipatov AS, Andreansky S, Webby RJ,et al. Pathogenesis of Hong Kong H5N1 influenza virus NS gene reassortants in mice: the role of cytokines and B- and T-cell responses. J Gen Virol. 2005;86(Pt 4):1121-30.
    [5] Ko SY, Ko HJ, Chang WS,et al. alpha-Galactosylceramide can act as a nasal vaccine adjuvant inducing protective immune responses against viral infection and tumor. J Immunol. 2005;175(5):3309-17.
    [6] Kopf M, Abel B, Gallimore A,et al. Complement component C3 promotes T-cell priming and lung migration to control acute influenza virus infection. Nat Med. 2002;8(4):373-8.
    [7] Cavanagh LL, Bonasio R, Mazo IB,et al. Activation of bone marrow-resident memory T cells by circulating, antigen-bearing dendritic cells. Nat Immunol. 2005;6(10):1029-37.
    [8] Belz GT, Wilson NS, Smith CM,et al. Bone marrow-derived cells expand memory CD8+ T cells in response to viral infections of the lung and skin. Eur J Immunol. 2006;36(2):327-35.
    [9] Jelley-Gibbs DM, Brown DM, Dibble JP,et al. Unexpected prolonged presentation of influenza antigens promotes CD4 T cell memory generation. J Exp Med. 2005;202(5):697-706.
    [10] Wang JP, Kurt-Jones EA, Finberg RW. Innate immunity to respiratory viruses. Cell Microbiol. 2007;9(7):1641-6.
    [11] Fazilleau N, Eisenbraun MD, Malherbe L,et al. Lymphoid reservoirs of antigen-specific memory T helper cells. Nat Immunol. 2007;8(7):753-61.
    [12] Kohlmeier JE, Miller SC, Woodland DL. Cutting edge: Antigen is not required for the activation and maintenance of virus-specific memory CD8+ T cells in the lung airways. J Immunol. 2007;178(8):4721-5.
    [13] Day EB, Zeng W, Doherty PC,et al. The context of epitope presentation can influence functional quality of recalled influenza A virus-specific memory CD8+ T cells. J Immunol. 2007;179(4):2187-94.
    [14] Lawrence CW, Ream RM, Braciale TJ. Frequency, specificity, and sites of expansion ofCD8+ T cells during primary pulmonary influenza virus infection. J Immunol.2005; 174(9): 5332-40.
    [15] Marshall DR, Olivas E, Andreansky S,et al. Effector CD8+ T cells recovered from an influenza pneumonia differentiate to a state of focused gene expression. Proc Natl Acad Sci U S A. 2005;102(17):6074-9.
    [16] Legge KL, Braciale TJ. Lymph node dendritic cells control CD8+ T cell responses through regulated FasL expression. Immunity. 2005;23(6):649-59.
    [17] Turner SJ, Kedzierska K, Komodromou H,et al. Lack of prominent peptide-major histocompatibility complex features limits repertoire diversity in virus-specific CD8+ T cell populations. Nat Immunol. 2005;6(4):382-9.
    [18] Kedzierska K, La Gruta NL, Turner SJ,et al. Establishment and recall of CD8+ T-cell memory in a model of localized transient infection. Immunol Rev. 2006;211:133-45.
    [19] Crowe SR, Miller SC, Brown DM,et al. Uneven distribution of MHC class II epitopes within the influenza virus. Vaccine. 2006 ;24(4):457-67.
    [20] McKinstry KK, Golech S, Lee WH,et al. Rapid default transition of CD4 T cell effectors to functional memory cells. J Exp Med. 2007;204(9):2199-211.
    [21] Agrewala JN, Brown DM, Lepak NM,et al. Unique ability of activated CD4+ T cells but not rested effectors to migrate to non-lymphoid sites in the absence of inflammation. J Biol Chem. 2007;282(9):6106-15.
    [22] Shakhar G, Lindquist RL, Skokos D,et al. Stable T cell-dendritic cell interactions precede the development of both tolerance and immunity in vivo. Nat Immunol. 2005;6(7):707-14.
    [23] Henrickson SE, Mempel TR, Mazo IB,et al. T cell sensing of antigen dose governs interactive behavior with dendritic cells and sets a threshold for T cell activation. Nat Immunol. 2008;9(3):282-91.
    [24] Bradley LM, Haynes L, Swain SL. IL-7: maintaining T-cell memory and achieving homeostasis. Trends Immunol. 2005;26(3):172-6.
    [25] Belz GT, Bedoui S, Kupresanin F,et al. Minimal activation of memory CD8+ T cell by tissue-derived dendritic cells favors the stimulation of naive CD8+ T cells. Nat Immunol. 2007;8(10):1060-6.
    [26] Zammit DJ, Turner DL, Klonowski KD,et al. Residual antigen presentation after influenza virus infection affects CD8 T cell activation and migration. Immunity. 2006;24(4):439-49.
    [27] Jelley-Gibbs DM, Dibble JP, Brown DM,et al. Persistent depots of influenza antigen fail to induce a cytotoxic CD8 T cell response. J Immunol. 2007;178(12):7563-70.
    [28] McKinstry KK, Strutt TM, Swain SL. The effector to memory transition of CD4 T cells. Immunol Res. 2008;40(2):114-27.
    [29] Swain SL, Agrewala JN, Brown DM,et al. CD4+ T-cell memory: generation and multi-faceted roles for CD4+ T cells in protective immunity to influenza. Immunol Rev. 2006;211:8-22.
    [30] Odendahl M, Mei H, Hoyer BF,et al. Generation of migratory antigen-specific plasma blasts and mobilization of resident plasma cells in a secondary immune response. Blood. 2005;105(4):1614-21.
    [31] Coleclough C, Sealy R, Surman S,et al. Respiratory vaccination of mice against influenza virus: dissection of T- and B-cell priming functions. Scand J Immunol. 2005;62 Suppl 1:73-83.