重组沙门菌表达结核分枝杆菌保护性抗原ESAT-6、CFP-10及其黏膜免疫机理研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
结核病(Tuberculosis, TB)由来已久,自有人类历史记录开始,就有了TB的记录。迄今为止,约2亿人被TB夺去生命,而这个数字仍在上升,世界卫生组织宣布TB是人类目前面临的最大威胁之一。1882年,Robert Koch证实了结核分枝杆菌(Mycobacterium tuberculosis,MTB)是TB的病原。全世界已有1/3人口约20亿人感染了MTB,现有结核病人2千万,每年新发病人数约880万,每年死亡人数高达160万。目前广泛用于预防TB的唯一疫苗是卡介苗(Bacille Calmette Guérin, BCG),但其免疫保护效果对于肺结核及弥散性结核分别约为50%和80%,存在显著差异。TB野毒株的高度耐药以及与人免疫缺陷病毒( Human immunodeficiency virus,HIV)共感染的不断上升,使TB警戒再次升高,并强调要利用免疫手段来控制TB的感染和传播。
     TB特异性免疫应答在清除MTB感染中发挥重要作用。BCG对青少年TB的预防具有重要的作用,但对于成人肺结核的保护作用有限,因此在设计新型抗结核疫苗时要考虑到如何预防肺结核。基因组学比较分析显示BCG中要比MTB缺失约100多个编码序列,其中的一些序列编码重要的抗原,当这些抗原被重新重组入BCG中都能够增强抗结核的能力。BCG中缺失RD1(Regions of difference 1),而RD1中的编码基因esxB和esxA基因分别编码培养滤液蛋白10(Culture filter protein 10, CFP-10)和早期分泌抗原靶6(Earlier secreted antigen target 6, ESAT-6),它们仅存在于临床分离的致病性MTB及牛分枝杆菌野毒株(M. bovis),能够诱导有效的Th1应答,并在动物实验中显示其对TB的保护效力,因而成为TB免疫诊断以及疫苗研究中最有力的候选抗原。为此,本研究以沙门菌及其鞭毛蛋白为载体,原核表达并运送ESAT-6、CFP-10蛋白,经滴鼻免疫,探讨了MTB保护性抗原ESAT-6和CFP-10蛋白诱导的黏膜免疫机理。
     1.结核分枝杆菌ESAT-6、CFP-10蛋白的克隆、表达及免疫原性分析
     为分析结核分枝杆菌分泌性抗原ESAT-6和CFP-10蛋白的免疫原性,利用PCR扩增了ESAT-6、CFP-10编码基因,经克隆筛选和测序鉴定后,构建了原核表达质粒pBCX-esat及pET-cfp。将两质粒分别转化宿主菌BL21(DE3),进行诱导表达。SDS-PAGE分析结果表明,重组的ESAT-6和CFP-10蛋白均获表达,融合蛋白的大小分别约为39kD和19kD。将CFP-10与本室研制的针对CFP-10蛋白的单克隆抗体6E8进行Western-blotting分析。结果显示,CFP-10融合蛋白能与特异性单克隆抗体6E8反应。同时,在ELISA实验中,重组ESAT-6蛋白与医院结核病人血清呈阳性反应。将ESAT-6及CFP-10融合蛋白分别以腹腔注射免疫C57BL/6小鼠,并于第二次免疫后7d,取小鼠脾脏细胞,用胞内细胞因子染色法检测ESAT-6及CFP-10蛋白特异性CD4+ T细胞分泌的IFN-γ及IL-4。结果两组蛋白均能诱导较高水平的IFN-γ,而IL-4水平低于IFN-γ水平。此外,经检测脾脏中CD3+ T细胞的CD69分子表达发现,ESAT-6和CFP-10蛋白免疫组的CD69水平显著上调。从而说明重组ESAT-6和CFP-10蛋白具有良好的免疫原性;免疫小鼠后,能够激活CD3+ T细胞,并诱导以细胞免疫为主的Th1应答,即重组蛋白ESAT-6及CFP-10具有Th1免疫原性。
     2.重组沙门菌表达的ESAT-6、CFP-10及其特异性免疫应答分析
     为了分析重组沙门菌诱导ESAT-6及CFP-10蛋白特异性免疫应答,将ESAT-6及CFP-10蛋白编码基因分别插入原核载体pYA3333中,将构建正确的质粒分别命名为pYA33-esat和pYA33-cfp。通过电穿孔法转化减毒鼠伤寒沙门菌X4550,获得重组菌X4550(33-esat)和X4550(33-cfp)。以每只1×108 cfu剂量的重组菌滴鼻免疫C57BL/6小鼠,间隔18d,在进行第2次免疫后10d取免疫小鼠脾脏、肺脏、肠系膜淋巴结(Mesenteric lymph node, MLN)及派伊尔氏结(Peyer’s patches, PP)细胞,以ESAT-6和CFP-10蛋白作为刺激原,检测抗原特异性的IFN-γ分泌细胞和IL-4分泌细胞。结果表明,经沙门菌表达运送的结核分枝杆菌抗原ESAT-6和CFP-10均能诱导特异性的免疫应答。在肺脏及PP细胞中,检测到较高水平的IFN-γ分泌细胞,与IL-4分泌细胞比较,差异显著。在脾脏和MLN中,免疫应答呈现平衡状态,IFN-γ分泌细胞和IL-4分泌细胞数量相当。此外,对脾脏、MLN、PP及支气管肺泡灌洗液(Bronchoalveolar lavage fluid, BAF)细胞中的共刺激分子检测结果表明,两组重组菌均能上调共刺激分子的表达。利用ELISA分别在免疫鼠的血清和BAF /肠黏膜中检测到ESAT-6及CFP-10蛋白特异性的IgG和IgA抗体。从而表明,以滴鼻方式接种重组减毒沙门菌,能够诱导ESAT-6及CFP-10蛋白特异性的细胞免疫应答,在黏膜邻近器官组织中,免疫应答趋于细胞免疫,而在黏膜远端免疫器官和组织中,免疫应答则处于平衡状态。以滴鼻免疫重组菌,不仅能诱导有效的体液免疫还可以激发局部的黏膜免疫,这为呼吸道疾病的防控提供了新的依据。
     3.沙门菌鞭毛蛋白嵌合表达的ESAT-6及其免疫学特性分析
     为研究以沙门菌鞭毛系统作为运送载体诱导的ESAT-6特异性免疫应答,利用overlap PCR将ESAT-6蛋白编码基因插入沙门菌鞭毛蛋白基因fliC的高变区域,构建成嵌合鞭毛片段fliC/esat。将fliC/esat片段插入原核表达载体pET,构建的质粒命名为pET-fliC/esat。将质粒pET-fliC/esat通过电穿孔法转化沙门菌LB5000,重组菌命名为LB5000(fliC/esat)。用P22噬菌体介导LB5000(fliC/esat)向都柏林沙门菌SL5928的转导,利用PCR、动力学检测及凝集试验鉴定并获得阳性转导菌SL5928(fliC/esat)。将转导菌SL5928(fliC/esat)感染结肠癌细胞HT-29,证实转导菌具有良好的感染能力;黏附力试验证实,转导菌具有良好的黏附特性。利用RT-PCR技术检测转导菌SL5928(fliC/esat)感染BMDC、F4/80+ MФ和肠上皮细胞(Enterocyte, EC)后的TLR-5 mRNA表达,结果在3h时,三种细胞就能表达TLR-5 mRNA;至6h,TLR-5 mRNA表达量有所升高。在体内实验中,将转导菌以每只1×107cfu滴鼻免疫C57BL/6小鼠,并于第二次免疫后10d,检测脾脏、肺脏、MLN、PP及鼻咽相关淋巴结(Nasopharynx associated lymph node,NALT)细胞中的IFN-γ和IL-4分泌细胞数。结果在肺脏、PP及NALT细胞中,IFN-γ分泌细胞与IL-4分泌细胞水平相比较显著升高,免疫应答趋向于Th1型;而在脾脏及MLN中,IFN-γ与IL-4分泌细胞数无显著性差异,免疫应答呈现Th1/Th2混合应答,处于平衡状态。而在体内的CTL实验中发现,与重组菌X4550(33-esat)相比较,转导菌SL5928(fliC/esat)诱导的CTL具有较强的特异性杀伤力。在血清及BAF的抗体检测中显示,转导菌能够诱导一定水平的血清IgG和黏膜IgA抗体。综上所述,鞭毛中嵌合MTB抗原ESAT-6后,不影响鞭毛自身特性,经沙门菌鞭毛系统运送,能够诱导ESAT-6抗原特异的细胞及黏膜免疫应答,这为TB疫苗研究开辟了新的研究方向。
     4.嵌合鞭毛蛋白fliC/esat与BCG黏膜共免疫诱导的免疫应答分析
     为研究嵌合鞭毛蛋白在TB免疫的中作用,将原核表达质粒pET-fliC/esat及pET-fliC转化大肠杆菌BL21(DE3),诱导表达嵌合蛋白fliC/esat及鞭毛蛋白fliC。SDS-PAGE分析结果表明,嵌合蛋白及fliC蛋白均以可溶性表达,大小分别约为64kD和57kD。将嵌合蛋白和fliC蛋白分别进行Western-blotting分析,以鼠伤寒沙门菌全菌血清作为一抗,结果显示嵌合蛋白及fliC蛋白均能与多抗血清反应。与ESAT-6蛋白相比较,嵌合蛋白及鞭毛蛋白fliC在体外均能诱导BMDC成熟。体外刺激F4/80+ MФ细胞实验表明,ESAT-6蛋白、fliC蛋白及嵌合蛋白均能上调腹腔MФ共刺激分子CD40、CD80、CD86和CD54。三种蛋白刺激BMDC及分选的F4/80+ MФ细胞分泌IL-12p70的检测结果显示,在刺激BMDC时,与ESAT-6蛋白组相比较,嵌合蛋白能够诱导分泌较高水平的IL-12p70;而在刺激F4/80+ MФ细胞时,与对照相比较,三种蛋白刺激后,F4/80+ MФ细胞均不能分泌有效的IL-12p70。在体内实验中,用嵌合蛋白静脉注射免疫C57BL/6小鼠,结果嵌合蛋白免疫组能够显著增强ESAT-6特异性免疫应答,而且免疫应答以Th1为主。此外,将BCG与嵌合蛋白共滴鼻免疫发现,在肺脏、PP、NALT及脾脏中均能诱导显著的Th1应答,与嵌合蛋白单一免疫组相比较,BCG能够增强ESAT-6特异性的免疫水平。检测CD8+CD69+细胞结果表明,BCG与嵌合蛋白共免疫组的CD8+CD69+细胞百分数均高于单独BCG及嵌合蛋白免疫组。从而说明,鞭毛嵌合表达ESAT-6抗原,有利于增强ESAT-6的特异性应答,鞭毛蛋白对ESAT-6抗原具有佐剂作用。BCG与嵌合蛋白共免疫结果提示,BCG与嵌合ESAT-6抗原的鞭毛蛋白之间具有协同作用,能够提高ESAT-6特异的免疫应答水平。
Tuberculosis (TB) has a long history. It was presented before the beginning of recorded history. TB, together with acquired immune deficiency syndrome(AIDS) and malaria, remains today one of the leading infectious diseases. TB causes an estimated of at least 200 millions deaths now, and it was declared a global emergency by the World Health Organization. In 1882, Robert Koch identified Mycobacterium tuberculosis (MTB) as the causative agent of TB. More than one third of people over the world have infected MTB, and there has more than 20 million patients. There were an estimated 8.8 million new TB cases per year. A total of 1.6 million people died of TB, including 195000 patients infected with HIV. The reported efficacy of the currently used Bacillus Calmette-Guérin (BCG) vaccine against TB is highly variable, ranging from 50% against pulmonary tuberculosis to 80% against disseminated tuberculosis. The alarming increase in the incidence of TB , due to emergence of TB strains resistant to all major chemotherapeutic drugs and to co-infection with human immunodeficiency virus (HIV), has emphasized the need to develop immunological tools for TB control.
     Adaptive immunity against TB plays an immportant role in clearing infectious MTB. BCG vaccine protected efficiently against leprosy as well as childhood manifestations of TB. However, the protective efficacy against adult pulmonary TB was limited. Therefore, the new vaccine against TB should include protective efficacy against pulmonary TB. Comparative genomics identified up to 100 coding sequences absent from BCG but present in MTB. Some of these coding sequences encode potential antigens that could improve immunogenicity if reintroduced into BCG. RD1(Regions of Difference 1) is one of the regions absent from BCG. Among the missing genes are the coding sequences esxB and esxA, which respectively encode CFP-10 (Culture filter protein 10, CFP-10) and ESAT-6 (Earlier secreted antigen target 6, ESAT-6), low-molecular-weight proteins that induce potent Th1 responses. Both M.bovis and some clinical pathogenic MTB share the two proteins. ESAT-6 and CFP-10 protein elicit protection against TB in animal models and become two of the most potential candidates in diagnosis and vaccine research. The purpose of this study were to: (1) express ESAT-6 and CFP-10 antigens using attenuated Salmonella and/or chimeric flagellin vector; (2) analyze the mucosal immune responses and their mechanisms induced by recombinant ESAT-6 and CFP-10 proteins through intranasally immunization; (3) study the immune responses initiated by chimeric flagellin fliC/esat co-immunized with BCG.
     1. Cloning, expression and immunogenic analysis of Mycobacterium tuberculosis ESAT-6 and CFP-10 protein
     To analyze immunogenic characteristics of ESAT-6 and CFP-10 proteins of Mycobacterium tuberculosis. ESAT-6 and CFP-10 coding genes were cloned and identified by PCR. Prokaryotic exprssion plasmids pBCX-esat and pET-cfp were constructed harboring the coding gene respectively and transformed into E.coli BL21(DE3). Recombinant bacteria were identified and induced by IPTG. SDS-PAGE results showed that both ESAT-6 and CFP-10 proteins were expressed and the sizes of them were 34kD and 19kD, respectively. Western-blotting results showed that fusion protein CFP-10 could react with the specific mAb 6E8. And, fusion protein ESAT-6 can bind to the serum of TB patients in ELISA assay. C57BL/6 mice were immunized intraperitoneally with ESAT-6 and CFP-10 fusion proteins respectively, which were emulsified by Freund's adjuvant. At the 7th day of second immunization in three-week intervels, splenocytes were collected from the immunized mice. IFN-γ-producing and IL-4-producing CD4~+ T cells were detected by intracellular cytokine staining assay. The results showed that both ESAT-6 and CFP-10 proteins can induce higher level of IFN-γcompared with that of IL-4. Furthermore, both ESAT-6 and CFP-10 proteins can significantly up-regulate CD69 molecule expression on CD3~+ T cells. Overall, both ESAT-6 and CFP-10 fusion proteins are capable of up-regulating CD69 molecule of CD3~+ T cells and inducing Th1 immune responses.
     2. Immune responses specific for ESAT-6 and CFP-10 antigen expressed by recombinant Salmonella typhimurium
     To determine mucosal immune responses induced by recombinant Salmonella harboring ESAT-6 and CFP-10 antigens, prokaryotic exprssion plasmids pYA33-esat and pYA33-cfp carrying the ESAT-6 and CFP-10 coding genes were constructed firstly and eletro-transformed into an attenuated strain X4550 of Salmonella typhimurium, the recombinant bacteria were named as X4550(33-esat) and X4550(33-cfp), respectively. C57BL/6 mice were immunized intranasally with 1×108 cfu recombinant bacteria in 18d intervals. Cells from spleen, lung, mesenteric lymph node (MLN) and Peyer’s patches (PP) were collected from mice after second immunization, and the specific IFN-γ-secreting cells and IL-4-secreting cells were detected using ESAT-6 and CFP-10 proteins as stimuli. The results showed that the immune responses specific for ESAT-6 and CFP-10 proteins could be detected by the ELISPOT assay. In lung and PP cells, immune responses against ESAT-6 and CFP-10 proteins both were biased toward Th1 response, the frequency of IFN-γ-secreting cells was much higher than that of IL-4 -secreting cells. However, in splenocytes and MLN cells, the antigen specific immune responses acted as Th1 and Th2 balance, the frequency of IFN-γ-secreting cells was close to that of IL-4-secreting cells. Also, costimulatory molecules CD80 and CD86 from splenocytes, MLN, PP and bronchoalveolar lavage fluid (BAF) cells were up-regulated after the second immunization. Furthermore, IgG and IgA antibodies from serum, BAF and fecal extracts were detected by ELISA respectively, and the levels of antibodies in immunized group had significantly increased compared with those of the negative control. These results suggested that intranasal immunization of recombinant Salmonella not only can induce cellular immune responses, but also can elicit humoral and mucosal immune responses. It provided the bases for the development of mocusal vaccine against infectious disease of respiratory tract.
     3. Immunologic characteristics of ESAT-6 antigen expessed by Salmonella chimeric flagellin
     To understand the mucosal immune responses against ESAT-6 in the Salmonella chimeric flagellin system, which act as a vector to deliver ESAT-6 antigen of Mycobacterium tuberculosis. Chimeric flagellin gene fliC/esat were constructed by overlap PCR technique. The ESAT-6 coding gene was inserted to the hypervariable region of Salmonella flagellin gene fliC. Prokaryotic exprssion plasmid pET-fliC/esat carrying the chimeric fragment was constructed firstly and eletro-transformed into the strain LB5000 of Salmonella typhimurium, the recombinant bacteria were named as LB5000(fliC/esat). And then, the transduction was conducted between LB5000(fliC/esat) and Salmonella dublin strain SL5928 using bacteriophage P22HT int. The positive transductants were further identified by PCR, motolity observation and serological agglutination assay. In order to determine the invasion and adhesion capacities of SL5928(f/e). The colon carcinoma cells HT-29 were infected with SL5928(f/e). The results showed that SL5928(f/e) had favourable invasion and adhesion. It was detected TLR-5 mRNA from BMDC, F4/80+ MФand enterocytes infected with SL5928(f/e) by RT-PCR. All three type cells expressed TLR-5 mRNA at 3h post infection and the quantity of TLR-5 mRNA up-regulated at 6h. Furthermore, C57BL/6 mice were immunized intranasally with 1×107 cfu SL5928(f/e) in 18d intervals. Cells from spleen, lung, MLN, PP and nasopharynx associated lymph node (NALT) were harvested from mice after second immunization, and the specific IFN-γ-secreting cells and IL-4 secreting cells were detected using ESAT-6 protein or peptide of ESAT-6(1-20aa) as stimuli. The results showed that in lung, PP cells and NALT cells, immune responses against ESAT-6 protein or ESAT-6 peptide (1-20aa) both were biased toward Th1 response, the frequency of IFN-γ-secreting cells was markedly higher than that of IL-4 -secreting cells. However, in spleen and MLN cells, the frequency of IFN-γ-secreting cells was close to that of IL-4-secreting cells. Then, the percentage of specific lysis of SL5928(f/e) was higher than that of X4550(33-esat). Finally, the levels of IgG and IgA antibodies from serum and BAF in immunized group were significantly higher compared with those of the control group. These results suggested that Salmonella chimeric flagellin delivery system was effective after intranasal immunization. The cellular and mucosal immune responses specific for ESAT-6 can be induced by tranductant SL5928(f/e). This system could develop a new pathway for TB vaccine.
     4. Immune responses initiated by intranasal co-immunization of chimeric flagellin fliC/esat and BCG
     To define the role of ESAT-6 chimeric flagellin in TB immunology, prokaryotic expression plasmids pET-fliC/esat and pET-fliC were transformed into E.coli and induced by IPTG. SDS-PAGE results showed the ESAT-6 chimeric flagellin and fliC protein both are soluble. The sizes of the two proteins were 64kD and 57kD respectively. Western-blotting analysis showed that chimeric flagellin and fliC protein both could be bind to serum of Salmonella typhymurium. BMDC maturation was triggered by both ESAT-6 chimeric flagellin and fliC protein. In contrast, ESAT-6 protein alone didn’t activate BMDC. There were up-regulated the expression level of CD40, CD80, CD86 and CD54 costimulatory molecules on F4/80+ MФafter the stimulation of ESAT-6, ESAT-6 chimeric flagellin and fliC protein, respectively. IL-12p70 was detected in the superantants of BMDC and F4/80+ MФ. The results showed that chimeric flagellin was able to induce higher level of IL-12p70 than that of ESAT-6 protein. However, there weren’t any IL-12p70 from F4/80+ MФafter the three proteins stimulating. In order to further demonstrate the adjuvant activity of flagellin, C57BL/6 mice were immunized intravenously with ESAT-6 chimeric flagellin. The results showed that ESAT-6 chimeric flagellin could enhance the immunogenic activity of ESAT-6 protein. Furthermore, mice were co-immunized with BCG and ESAT-6 chimeric flagellin. This immune strategy increased the level of immune responses and biased toward Th1 response in lung, PP, NALT and spleen. Percentage of CD8+CD69+ cells of co-immunization group were higher than that of BCG or ESAT-6 chimeric protein group. Overall, flagellin can present adjuvant activity for ESAT-6 protein, and the novel co-immunization of Chimeric flagellin and BCG could enhance the specific Th1 immune responses.
引文
[1] Rastogi D, Ratner AJ, Prince A. Host-bacterial interactions in the initiation of inflammation. Paediatr Respir Rev. 2001, 2: 245-252.
    [2] Boyton RJ and Openshaw PJ. Pulmonary defences to acute respiratory infection. Br Med Bull. 2002, 61:1-12.
    [3] Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature.1998, 392: 245-252.
    [4] Dieu M-C, Vanbervliet B, Vicari A, et al. Selective recruitment of immature and mature dendritic cells by distinct chemokines expressed in different anatomic sites. J Exp Med. 1998, 188:373-386.
    [5]Iwasaki A, Kelsall BL. Unique functions of CD11b+, CD8α+, and double-negative Peyer’s patch dendritic cells. J Immunol. 2001, 166:4884–4890.
    [6] Hochrein H, Shortman K, Vremec D, et al. Differential production of IL-12, IFN-alpha, and IFN-gamma by mouse dendritic cell subsets. J Immunol. 2001, 166:5448-5455.
    [7] Aliberti J, Reis e Sousa C, Schito M, et al. CCR5 provides a signal for microbial induced production of IL-12 by CD8α+ dendritic cells. Nat. Immunol. 2000, 1:83-87.
    [8] Maldonado-Lopez R, Maliszewski C, Urbain J, Moser M. Cytokines regulate the capacity of CD8α+ and CD8ααdendritic cells to prime Th1/Th2 cells in vivo. J Immunol. 2001, 167: 4345–4350.
    [9]Wick MJ. The role of dendritic cells in the immune response to Salmonella. Immunol Lett. 2003, 85(2): 99-102.
    [10] Pulendran B, Lingappa J, Kennedy M.K, J. Smith, M. Teepe, A. Rudensky, C.R. Maliszewski, E. Maraskovsky, J. Immunol. 1997, 159 :2222-2231.
    [11] Iwasaki A, Kelsall BL. Localization of distinct Peyer’s patch dendritic cell subsets and their recruitment by chemokines macrophage inflammatory protein (MIP)-3α, MIP-3β, and secondary lymphoid organ chemokine. J Exp Med. 2000, 191:1381–1394.
    [12] Vremec D, Shortman K. Dendritic cell subtypes in mouse lymphoid organs: cross -correlation of surface markers, changes with incubation, and differences among thymus, spleen, and lymph nodes. J Immunol. 1997, 159:565–573.
    [13] Vremec D, Pooley J, Hochrein H, et al. CD4 and CD8 expression by dendritic cell subtypes in mouse thymus and spleen. J Immunol. 2000, 164:2978–2986.
    [14] Shortman K and Liu YJ. Mouse and human dendriticcell subtypes. Nat Rev Immunol. 2002, 2: 151– 161.
    [15] Daro E, Pulendran B, Brasel K, et al. Polyethylene glycol-modified GM-CSF expands CD11bhigh CD11chigh but not CD11blowCD11chigh murine dendritic cells in vivo: a comparative analysis with Flt3 ligand. J Immunol. 2000, 165: 49-58.
    [16] Kamath AT, Pooley J, O’Keeffe MA, et al. The development, maturation, and turnover rate of mouse spleen dendritic cells. J Immunol. 2000, 165:6762-6770.
    [17] Yrlid U, Wick MJ. Antigen presentation capacity and cytokine production by murine splenic dendritic cell subsets upon Salmonella encounter. J Immunol. 2002, 169:108–116.
    [18] Valdez Y, Mah W, Winslow MM, et al. Major histocompatibility complex class II presentation of cell-associated antigen is mediated by CD8α+ dendritic cells in vivo. J Exp Med 2002, 195:683–694.
    [19] Moron G, Rueda P, Casal I, et al. CD8α+CD11β+ dendritic cells present exogenous virus like particles to CD8+ T cells and subsequentlyexpress CD8αand CD205 molecules. J Exp Med. 2002, 195:1233–1245.
    [20] Kelsall BL, Biron CA, Sharma O, et al. Dendritic cells at the host-pathogen interface.Nat Immunol. 2002, 3: 699–702.
    [21] Pulendran B, Kumar P, Cutler CW, et al. Lipopolysaccharides from distinct pathogens induce different classes of immune responses in vivo. J Immunol. 2001, 167:5067–5076.
    [22] Den Haan JM, Bevan MJ. Constitutive versus activation-dependent cross-presentation of immune complexes by CD8α+ and CD8α- dendritic cells in vivo. J Exp Med. 2002, 196:817–827.
    [23] Schlecht G, Leclerc C, Dadaglio G. Induction of CTL and nonpolarized Th cell responses by CD8α+ and CD8ααdendritic cells. J Immunol. 2001,167:4215–4221.
    [24] Cella M, Engering A, Pinet V, et al. Inflammatory stimuli induce accumulation of MHC class II complexes on dendritic cells. Nature. 1997,388:782–787.
    [25] Rescigno M, Citterio S, Thery C, et al. Bacteria-induced neo-biosynthesis, stabilization, and surface expression of functional class I molecules in mouse dendritic cells. Proc Natl Acad Sci USA. 1998, 95: 5229–5234.
    [26] Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998, 392: 245–252.
    [27] Montoya M, Schiavoni G, Mattei F, et al. Type I interferons produced by dendritic cells promote their phenotypic and functional activation. Blood. 2002, 99:3263–3271.
    [28] Geissmann F, Launay P, Pasquier B,et al. A subset of human dendritic cells expresses IgA Fc receptor (CD89), which mediates internalization and activation upon cross-linking by IgA complexes. J Immunol. 2001, 166:346–352.
    [29] Sauter B, Albert ML, Francisco L, et al. Consequences of cell death: exposure to necrotic tumor cells, but not primary tissue cells or apoptotic cells, induces the maturation of immunostimulatory dendritic cells. J Exp Med. 2000, 191: 423–434.
    [30] Muraille E, De Trez C, Pajak B, et al. T cell-dependent maturation of dendritic cells in response to bacterial superantigens. J Immunol. 2002, 168:4352–4360.
    [31] Theill LE, Boyle WJ, Penninger JM. et al.T cells, bone loss, and mammalian evolution. Annu Rev Immunol. 2002, 20:795–823.
    [32] Van Kooten C, Banchereau J. CD40-CD40 ligand. J Leukoc Biol. 2000, 67:2–17.
    [33] Janeway CA Jr., Medzhitov R. Innate immune recognition. Annu Rev Immunol. 2002, 20:197–216.
    [34] Akira S, Takeda K, Kaisho T. Toll-like receptors: critical proteins linking innate and acquired immunity. Nat Immunol. 2001, 2:675–680.
    [35] Medzhitov R, Preston-Hurlburt P, Janeway CA, Jr. A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature. 1997, 388:394–397.
    [36] Kaisho T, Takeuchi O, Kawai T, et al. Endotoxin-induced maturation of MyD88-deficient dendritic cells. J Immunol. 2001, 166: 5688–5694.
    [37] Scanga CA, Aliberti J, Jankovic D, et al. Cutting edge: MyD88 is required for resistance to Toxoplasma gondii infection and regulates parasite-induced IL-12 production by dendritic cells. JImmunol. 2002, 168:5997–6001.
    [38]Schnare M, Barton GM, Holt AC, et al. Toll-like receptors control activation of adaptive immune responses. Nat Immunol. 2001, 2:947–950.
    [39]Crowley MT, Reilly CR, Lo D. Influence of lymphocytes on the presence and organization of dendritic cell subsets in the spleen. J Immunol. 1999, 163: 4894–4900.
    [40]Ingulli E, Ulman DR, Lucido MM,et al. In situ analysis reveals physical interactions between CD11b+ dendritic cells and antigen-specific CD4 T cells after subcutaneous injection of antigen. J Immunol. 2002, 169:2247–2252.
    [41] Jiao X, Lo-Man R, Guermonprez P, et al. Dendritic cells are host cells for mycobacteria in vivo that trigger innate and acquired immunity. J Immunol. 2002, 168:1294–1301.
    [42] Aggarwal BB and Higuchi M. Role of ceramide in tumour necrosis factor-mediated apoptosis and nuclear factor- kappa B activation. Biochem Sco Trans. 1998, 25(4):1166-1171.
    [43] Balkwill F and Burke F. The Cytokine Network. Immunol Today. 1989, 10(9):299-304.
    [44] Roitt I, Brostoff J, Male D. Immunology. 5th ed. Philadelphia, PA: Mosby; 1998.
    [45]Moser M, Murphy KM. Dendritic cell regulation of Th1/Th2 development. Nat Immunol. 2000, 1: 199-205.
    [46] Romagnani G, Annunziato F, Piccinni MP, et al. Th1/Th2 cells, their associated molecules and role in pathophysiology. Eur Cytokine Netw. 2000, 11(3):510-511.
    [47] Dent LA. For better or worse: common determinants influencing health and disease in parasitic infections, asthma and reproductive biology. J Reprod Immunol. 2002, 57:255-272.
    [48] Lafaille JJ. The role of helper T cell subsets in autoimmune diseases. Cytokine Growth Factor Rev. 1998, 9: 139- 151.
    [49]Mantovani A, Sozzani S. Chemokines (Chapter 5). In: Balkwill F, ed. The Cytokine Network. Oxford: Oxford University Press; 2000.
    [50] Matzinger R Tolerance, danger, and the extended family. Annu Rev Immunol. 1994,12: 991-1045.
    [51] McGuirk P, Mills KH. Pathogen-specific regulatory T cells provoke a shift in the Th1/ Th2 paradigm in immunity to infectious diseases. Trends Immunol. 2002, 23:450-455.
    [52] Groux H, O'Garra A, Bigler M, et al. A CD4+ T-cell subset inhibits antigen specific T-cell responses and prevents colitis. Nature. 1997, 389:737-742.
    [53]Fukaura H, Kent SC, Pietrusewicz MJ, et al. Induction of circulating myelin basic protein and proteolipid protein-specific transforming growth factor-beta1-secreting Th3 T cells by oral administration of myelin in multiple sclerosis patients. J Clin Invest. 1996, 98:70-77.
    [54] Gorelik L, Flavell RA. Transforming growth factor-beta in T-cell biology. Nat Rev Immunol. 2002, 2:46-53.
    [55] Lienhardt C, Azzurri A, Amedei A, el al. Active tuberculosis in Africa is associated with reduced Th1 and increased Th2 activity in vivo. Eur J Immunol. 2002, 32:1605-1613.
    [56] Newport MJ, Huxley CM, Huston S, et al. A mutation in the interferon-gamma-receptor gene and susceptibility to mycobacterial infection. N Engl J Med. 1996, 335:1941-1949.
    [57] Davis SS. Nasal vaccines. Adv Drug Deliv Rev. 2001, 51: 21-42.
    [58] Brandtzaeg P and Pabst R. Let's go mucosal: communication on slippery ground. Trends Immunol. 2004, 25: 570-577.
    [59] Kiyono H and Fukuyama S. The mucosal immune system: features of inductive and effector sites to consider in mucosal immunization and vaccine development. Nat Rev Immunol. 2004, 4: 699-710.
    [60] Nicod LP, Cochand L, Dreher D. Antigen presentation in the lung: dendritic cells and macrophages. Sarcoidosis. Vasc Diffuse Lung Dis. 2000, 17:246-255.
    [61] Ogra PL. Mucosal immunity: some historical perspective on host-pathogen interactions and implications for mucosal vaccines. Immunol Cell Biol. 2003, 81: 23-33.
    [62] Banchereau J and Steinman RM. Dendritic cells and the control of immunity. Nature. 1998, 392: 245 -252.
    [63] Banchereau J, Briere F, Caux C, et al. Immunobiology of dendritic cells. Annu Rev Immunol. 2000, 18: 767-811.
    [64] Kaul D and Ogra PL. Mucosal responses to parenteral and mucosal vaccines. Dev Biol. Stand 1998, 95:141-146.
    [65] Aittoniemi J, Koskinen S, Laippala P, et al. The significance of IgG subclasses and mannan-binding lectin (MBL) for susceptibility to infection in apparently healthy adults with IgA deficiency. 1999, 116(3):505-508.
    [66] Johansen FE, Braathen R, Brandtzaeg P. Role of J chain in secretory immunoglobulin formation. Scand. J. Immunol. 2000, 52:240-248.
    [67] Mostov KE. Transepithelial transport of immunoglobulins. Annu Rev Immunol. 1994, 12: 63-84.
    [68] Norderhaug IN, Johansen FE., Schjerven H, et al. Regulation of the formation and external transport of secretory immunoglobulins. Crit Rev Immunol. 1999, 19: 481-508.
    [69] Mestecky J, Russell MW, Elson CO. Intestinal IgA: novel views on its function in the defence of the largest mucosal surface. Gut. 1999, 44: 2-5.
    [70] Kaetzel CS, Robinson JK, Chintalacharuvu KR, et al. The polymeric immunoglobulin receptor (secretory component) mediates transport of immune complexes across epithelial cells: a local defense function for IgA. Proc Natl Acad Sci USA.1991, 88: 8796-8800.
    [71]王晓东,黄志华. sIgA在肠道免疫中的作用.国际消化病杂志. 2006, 26(5): 339-341.
    [72]Burns JW, Siadat-Pajouh M, Krishnaney AA, et al. Protective effect of rotavirus VP6-specific IgA monoclonal antibodies that lack neutralizing activity. Science.1996, 272: 104-107.
    [73] Mazanec MB, Coudret CL, Fletcher DR. Intracellular neutralization of influenza virus by immunoglobulin A anti-hemagglutinin monoclonal antibodies. J Virol. 1995, 69:1339-1343.
    [74] Morton HC, van Egmond M, van de Winkel JG. Structure and function of human IgA Fc receptors (Fc alpha R). Crit Rev Immunol. 1996,16: 423-440.
    [75]Cunningham-Rundles C. Physiology of IgA and IgA deficiency. J Clin Immunol. 2001, 21: 303-309.
    [76] Ammann AJ and Hong R. Selective IgA deficiency: presentation of 30 cases and a review of the literature. Medicine (Baltimore). 1971, 50: 223-236.
    [77] Natvig IB, Johansen FE, Nordeng TW, et al. Mechanism for enhanced external transfer of dimeric IgA over pentameric IgM: studies of diffusion, binding to the human polymeric Ig receptor, and epithelial transcytosis. J Immunol. 1997,159: 4330-4340.
    [78]Savilahti E, Klemola T, Carlsson B,et al. Inadequacy of mucosal IgM antibodies in selective IgA deficiency: excretion of attenuated polio viruses is prolonged. J Clin Immunol. 1988, 8: 89-94.
    [79]Burks AW Jr and Steele RW. Selective IgA deficiency. Ann Allergy. 1986, 57: 3-13.
    [80]Hammarstr?m L, Vorechovsky I, Webster D. Selective IgA deficiency (SIgAD) and common variable immunodeficiency (CVID). Clin Exp Immunol. 2000, 120: 225-231.
    [81]Schaffer FM, Monteiro RC, Volanakis JE, et al. IgA deficiency. Immunodefic Rev. 1991,3: 15-44.
    [82] Michael S, Sarah M and Zhou X. Adenoviral Vectors for Mucosal Vaccination Against Infectious Diseases Viral Immuno. 2005, 18(2):283–291.
    [83]Hogan RJ, Usherwood EJ, Zhong W, et al. Activated antigen-specific CD8+ T cells persist in the lungs following recovery from respiratory virus infections. J Immunol. 2001, 166: 1813–1822.
    [84]Hogan RJ, Cauley LS, Ely KH, et al. Long-term maintenance of virus-specific effector memory CD8+ T cells in the lung airways depends on proliferation. J Immunol. 2002, 169: 4976– 4981.
    [85]Ely KH, Roberts AD and Woodland DL. Cutting edge: effector memory CD8+ T cells in the lung airways retain the potential to mediate recall responses. J. Immunol. 2003, 171: 3338–3342.
    [86] Woodland DL. Cell-mediated immunity to respiratory virus infections. Curr Opin Immunol. 2003, 15: 430–435.
    [87]Gallichan WS and Rosenthal KL. Long-lived cytotoxic T lymphocyte memory in mucosal tissues after mucosal but not systemic immunization. J Exp Med. 1996,184: 1879–1890.
    [88] Michael S, Xizhong Z, Sarah M, et al. Mechanisms of Mucosal and Parenteral TuberculosisVaccinations: Adenoviral-Based Mucosal Immunization Preferentially Elicits Sustained Accumulation of Immune Protective CD4 and CD8 T Cells within the Airway Lumen. J Immunol. 2005, 174: 7986–7994.
    [89]Cripps AW, Kyd JM, Foxwell AR. Vaccines and mucosal immunisation. Vaccine. 2001, 19: 2513 -2515.
    [90] Sedgmen BJ, Meeusen EN, Lofthouse SA. Alternative routes of mucosal immunization in large animals. Immunol Cell Biol. 2004, 82:10-16.
    [91]孟晓丽,殷国荣,杨亚波,等.弓形虫复合黏膜疫苗滴鼻免疫小鼠诱导的特异性抗体动态观察.中国生物制品学杂志. 2007, 20(1):51-53.
    [92] Nugent J, Po AL, Scott EM. Design and delivery of non-parenteral vaccines. J Clin Pharm Ther. 1998,23: 257-285.
    [93] Renauld-Mongenie G, Mielcarek N, Cornette J, et al. Induction of mucosal immune responses against a heterologous antigen fused to filamentous hemagglutinin after intranasal immunization with recombinant Bordetella pertussis. Proc Natl Acad Sci USA.1996, 93:7944 -7949.
    [94] Partidos CD. Intranasal vaccines: forthcoming challenges. 2000, 3: 273-281.
    [95]Brandtzaeg P and Pabst R. Let’s go mucosal: communication on slippery ground. Trends Immunol. 2004, 25: 570–577.
    [96] Rudin A, Johansson EL, Bergquist C, et al. Differential kinetics and distribution of antibodies in serum and nasal and vaginal secretions after nasal and oral vaccination of humans. Infect Immun. 1998, 66: 3390–3396.
    [97] Brandtzaeg P, Johansen FE. Mucosal B cells: phenotypic characteristics, transcriptional regulation, and homing properties. Immunol Rev. 2005, 206:32–63.
    [98] Brandtzaeg P. History of oral tolerance and mucosal immunity. Ann NY Acad Sci. 1996, 778:1–27.
    [99] Hagiwara Y, McGhee JR, Fujihashi K, et al. Protective mucosal immunity in aging is associated with functional CD4+ T cells in nasopharyngealassociated lymphoreticular tissue. J Immunol. 2003, 170: 1754–1762.
    [100] Kaufmann SH. Recent findings in immunology give tuberculosis vaccines a new boost. Trends Immunol. 2005, 26:660–667.
    [101] Flynn JL, Chan J, Triebold KJ, et al. An essential role for interferon gamma in resistance to Mycobacterium tuberculosis infection. J Exp Med. 1993, 178:2249–2254.
    [102] Flynn JL, Goldstein MM, Triebold KJ,et al. Major histocompatibility complex class I-restricted T cells are required for resistance to Mycobacterium tuberculosis infection. Proc Natl Acad Sci USA. 1992, 89:12013–12017.
    [103] Feng CG, Bean AG, Hooi H, et al. Increase in gamma interferon-secreting CD8(+), as well as CD4(+), T cells in lungs following aerosol infection with Mycobacterium tuberculosis. Infect Immun. 1999, 67:3242–3247.
    [104] Ladel CH, Blum C, Dreher A, et al. Protective role of gamma/delta T cells and alpha/beta T cells in tuberculosis. Eur J Immunol. 1995, 25:2877–2881.
    [105] Hamasur B, Haile M, Pawlowski A, et al. A mycobacterial lipoarabinomannan specific monoclonal antibody and its F(ab’) fragment prolong survival of mice infected with Mycobacterium tuberculosis. Clin Exp Immunol. 2004, 138:30–38.
    [106] Glatman-Freedman A. The role of antibody-mediated immunity in defense against Mycobacterium tuberculosis: advances toward a novel vaccine strategy. Tuberculosis (Edinburgh). 2006, 86:191–197.
    [107] Brown RM, Cruz O, Brennan M, et al. Lipoarabinomannan-reactive human secretory immunoglobulin A responses induced by mucosal bacille Calmette-Guerin vaccination. J Infect Dis. 2003, 187:513–517.
    [108] Tjarnlund A, Rodriguez A, Cardona PJ, et al. Polymeric IgR knockout mice are more susceptible to mycobacterial infections in the respiratory tract than wild-type mice. Int Immunol. 2006, 18:807–816.
    [109] Doherty TM, Olsen AW, van Pinxteren L, et al. Oral vaccination with subunit vaccines protects animals against aerosol infection with Mycobacterium tuberculosis. Infect Immun. 2002, 70:3111–3121.
    [110] Lagranderie M, Chavarot P, Balazuc AM, et al. Immunogenicity and protective capacity of Mycobacterium bovis BCG after oral or intragastric administration in mice. Vaccine. 2000, 18: 1186–1195.
    [111] Lagranderie M, Nahori MA, Balazuc AM, et al. Dendritic cells recruited to the lung shortly after intranasal delivery of Mycobacterium bovis BCG drive the primary immune response towards a type 1 cytokine production. Immunol. 2003, 108:352–364.
    [112] Goonetilleke NP, McShane H, Hannan CM, et al. Enhanced immunogenicity and protective efficacy against Mycobacterium tuberculosis of bacille Calmette-Guerin vaccine using mucosal administration and boosting with a recombinant modified vaccinia virus Ankara. J Immunol. 2003, 171:1602–1609.
    [113] Aldwell FE, Keen DL, Parlane NA, et al. Oral vaccination with Mycobacterium bovis BCG in a lipid formulation induces resistance to pulmonary tuberculosis in brushtail possums. Vaccine. 2003, 22: 70–76.
    [114] Aldwell FE, Tucker IG, de Lisle GW, et al. Oral delivery of Mycobacterium bovis BCG in a lipid formulation induces resistance to pulmonary tuberculosis in mice. Infect Immun. 2003, 71:101–108.
    [115] Buddle BM, Aldwell FE, Skinner MA, et al. Effect of oral vaccination of cattle with lipid-formulated BCG on immune responses and protection against bovine tuberculosis. Vaccine. 2005, 23: 3581–3589.
    [116] Santosuosso M, Zhang X, McCormick S, et al. Mechanisms of mucosal and parenteral tuberculosis vaccinations:adenoviral-based mucosal immunization preferentially elicits sustained accumulation of immune protective CD4 and CD8 T cells within the airway lumen. J Immunol. 2005, 174:7986–7994.
    [117] Wang J, Thorson L, Stokes RW, et al. Single mucosal, but not parenteral, immunization with recombinant adenoviral-based vaccine provides potent protection from pulmonary tuberculosis. J Immunol. 2004, 173:6357–6365.
    [118] Chen L, Wang J, Zganiacz A,et al. Single intranasal mucosal Mycobacterium bovis BCG vaccination confers improved protection compared to subcutaneous vaccination against pulmonary tuberculosis. Infect Immun. 2004, 72: 238–246.
    [119] Xing Z, Lichty BD. Use of recombinant virus-vectored tuberculosis vaccines for respiratory mucosal immunization. Tuberculosis (Edinburgh). 2006, 86:211–217.
    [120] Kawahara M, Matsuo K, Honda M. Intradermal and oral immunization with recombinant Mycobacterium bovis BCG expressing the simian immunodeficiency virus Gag protein induces long-lasting, antigen-specific immune responses in guinea pigs. Clin Immunol. 2006, 119:67–78.
    [121] Gheorghiu M. BCG-induced mucosal immune responses. Int J Immunopharmacol. 1994, 16:435–444.
    [122] Dietrich J, Andersen C, Rappuoli R, et al. Mucosal administration of Ag85B-ESAT-6 protects against infection with Mycobacterium tuberculosis and boosts prior bacillus Calmette-Gue′rin immunity. J Immunol. 2006, 177: 6353–6360.
    [123] Lindblad EB, Elhay MJ, Silva R, et al. Adjuvant modulation of immune responses to tuberculosis subunit vaccines. Infect Immun. 1997, 65:623–629.
    [124] Hogarth PJ, Jahans KJ, Hecker R, et al. Evaluation of adjuvants for protein vaccines against tuberculosis in guinea pigs. Vaccine. 2003, 21:977–982.
    [125] Giri PK, Sable SB, Verma I, et al. Comparative evaluation of intranasal and subcutaneous route of immunization for development of mucosal vaccine against experimental tuberculosis. FEMS Immunol Med Microbiol. 2005, 45:87–93.
    [126] Haile M, Hamasur B, Jaxmar T, et al. Nasal boost with adjuvanted heat-killed BCG or arabinomannan–protein conjugate improves primary BCGinduced protection in C57BL/6 mice. Tuberculosis (Edinburgh). 2005, 85:107–114.
    [127] Haile M, Schroder U, Hamasur B, et al. Immunization with heat-killed Mycobacterium bovis Bacille Calmette- Guerin (BCG) in Eurocine L3 adjuvant protects against tuberculosis. Vaccine. 2004, 22:1498–1508.
    [128] Hamasur B, Haile M, Pawlowski A, et al. Mycobacterium tuberculosis arabinomannan–protein conjugates protect against tuberculosis. Vaccine. 2003, 21:4081–4093.
    [129] Takahashi H, Sasaki K, Takahashi M, et al. Mutant Escherichia coli enterotoxin as a mucosal adjuvant induces specific Th1 responses of CD4+ and CD8+ T cells to nasal killed-Bacillus Calmette -Guérin in mice.Vaccine. 2006, 24:3591–3598.
    [130] D’Souza S, Rosseels V, Denis O, et al. Improved tuberculosis DNA vaccines by formulation in cationic lipids. Infect Immun. 2002, 70:3681–3688.
    [131] Singh M and O'Hagan D. Advances in vaccine adjuvants. Nat Biotechnol. 1999,17:1075- 1081.
    [132] Freytag LC and Clements JD. Bacterial toxins as mucosal adjuvants. Curr Top Microbiol. Immunol. 1999, 236:215-236.
    [133] Boyaka PN, Marinaro M, Jackson RJ, et al. IL-12 is an effective adjuvant for induction of mucosal immunity. J Immunol. 1999, 162: 122-128.
    [134] Rincon M, Anguita J, Nakamura T, et al. Interleukin (IL)-6 directs the differentiation of IL-4-producing CD4+ T cells. J Exp Med. 1997, 185: 461-469.
    [135] Baca-Estrada ME, Foldvari MM, Snider MM, et al. Intranasal immunization with liposome -formulated Yersinia pestis vaccine enhances mucosal immune responses. Vaccine. 2000, 18: 2203-2211.
    [136]焦凤超,焦新安,潘志明,等.减毒鼠伤寒沙门氏菌介导的H5亚型禽流感病毒DNA疫苗的实验免疫效果研究.病毒学报. 2006, 22(1):6-10.
    [137] Haile M, Schroder U, Hamasur B, et al. Immunization with heat-killed Mycobacterium bovis bacille Calmette-Guerin (BCG) in Eurocine L3 adjuvant protects against tuberculosis. Vaccine. 2004, 22:1498 -1508.
    [138] Hiroi T, Goto H, Someya K, et al. HIV mucosal vaccine: nasal immunization with rBCG-V3J1 induces a long term V3J1 peptide-specific neutralizing immunity in Th1- and Th2-deficient conditions. J Immunol. 2001, 167: 5862-5867.
    [139] Xing Z. The hunt for new tuberculosis vaccines: anti-TB immunity and rational design of vaccines. Curr Pharm Des. 2001, 7: 1015–1037.
    [140] McMichael A.J. HIV vaccines. Annu Rev Immunol. 2006, 24: 227–255.
    [141] Glyn Hewinson R. TB vaccines for the World. Tuberculosis. 2005, 85:1–6.
    [142] Hiroi T, Iwatani K, Iijima H, et al. Nasal immune system: distinctive Th0 and Th1/Th2 type environments in murine nasal-associated lymphoid tissues and nasal passage, respectively. Eur J Immunol. 1998, 28:3346–3353.
    [143] Jones HP, Hodge LM, Fujihashi K, et al. The pulmonary environment promotes Th2 cell responses after nasal-pulmonary immunization with antigen alone, but Th1 responses are induced during instances of intense immune stimulation. J Immunol. 2001, 167:4518–4526.
    [144] Carcaboso AM, Hernandez RM, Igartua M, et al. Potent, long lasting systemic antibody levels and mixed Th1/Th2 immune response after nasal immunization with malaria antigen loaded PLGA microparticles. Vaccine. 2004, 22:1423–1432.
    [145] Akbari O, Stock P, DeKruyff RH, et al. Mucosal tolerance and immunity: regulating the development of allergic disease and asthma. Int Arch Allergy Immunol 2003, 130:108–118.
    [146] Helgeby A, Robson NC, Donachie AM, et al. The combined CTA1-DD/ISCOM adjuvant vector promotes priming of mucosal and systemic immunity to incorporated antigens by specific targeting of B cells. J Immunol. 2006, 176:3697–3706.
    [147] Andersen CS, Dietrich J, Agger EM, et al. The combined CTA1-DD/ISCOMs vector is an effective intranasal adjuvant for boosting prior BCG immunity to Mycobacterium tuberculosis. Infect Immun. 2007, 75(1): 408-416.
    [148] Roman M, Martin-Orozco E, Goodman JS, et al. Immunostimulatory DNA sequences function as T helper-1-promoting adjuvants. Nat Med. 1997, 3:849–854.
    [149]Bafica A, Scanga CA, Feng CG, et al. TLR9 regulates Th1 responses and cooperates with TLR2 in mediating optimal resistance to Mycobacterium tuberculosis. J Exp Med. 2005, 202: 1715–1724.
    [150] Bloom BR, Widdus R. Vaccine visions and their global impact.Nat Med. 1998, 4:480–484.
    [151] Brandtzaeg P. Role of secretory antibodies in the defence against infections. Int J Med Microbiol 2003, 293:3–15.
    [152] Berzofsky JA, Ahlers JD, Belyakov IM. Strategies for designing and optimizing new generation vaccines. Nat Rev Immunol. 2001, 1:209–219.
    [153]崔萍,于三科.黏膜免疫佐剂及免疫途径研究进展.动物医学进展. 2007, 28(2): 81-84.
    [1] World Health Organization. Tuberculosis. WHO fact sheet No 104. 2002 Geneva: WHO.
    [2]Corbett EL, Watt CJ, Walker N, et al. The growing burden of tuberculosis: global trends and interactions with the HIV epidemic. Arch Intern Med. 2003, 163: 1009-1021.
    [3] De Cock KM. and Chaisson RE. Will DOTS do it? A reappraisal of tuberculosis control in countries with high rates of HIV infection. Int J Tuberc Lung Dis. 1999, 3: 457-465.
    [4]Morris RS, Pfeiffer DU, Jackson R. The epidemiology of Mycobacterium bovis infections. Vet Microbiol. 1994, 40:153-177.
    [5] Van Soolingen D, Hoogenboezem T, De Haas PE, et al. A novel pathogenic taxon of the Mycobacterium tuberculosis complex, Canetti: characterization of an exceptional isolate from Africa. Int J Syst Bacteriol. 1997, 47: 1236-1245.
    [6] Van Soolingen D, Van der Zanden AG, De Haas PE, et al. Diagnosis of Mycobacterium microti infections among humans by using novel genetic markers. J Clin Microbiol. 1998, 36: 1840-1845.
    [7] Kremer K, Van Soolingen D, Van Embden J, et al. Mycobacterium microti: more widespread than previously thought. J Clin Microbiol. 1998, 36: 2793-2794.
    [8] Brosch R, Gordon SV, Marmiesse M, et al. A new evolutionary scenario for the Mycobacterium tuberculosis complex. Proc Natl Acad Sci USA. 2002, 99: 3684-3689.
    [9]Riley RL, Mills CC, Nyka W, et al. Aerial dissemination of pulmonary tuberculosis. A two-year study of contagion in a tuberculosis ward. Am J Epidemiol. 1995, 142: 3-14.
    [10]Armstrong JA. and Hart PD. Phagosome-lysosome interactions in cultured macrophages infected with virulent tubercle bacilli. Reversal of the usual nonfusion pattern and observations on bacterial survival. J Exp Med. 1975, 142: 1-16.
    [11]Frieden TR, Sterling TR, Munsiff SS, et al. Tuberculosis. Lancet.2003,362: 887-899.
    [12] Hickman SP, Chan J, Salgame P. Mycobacterium tuberculosis induces differential cytokine production from dendritic cells and macrophages with divergent effects on naive T cell polarization. J Immunol. 2002, 168: 4636-4642.
    [13]Nau GJ, Richmond JF, Schlesinger A, et al. Human macrophage activation programs induced by bacterial pathogens. Proc Natl Acad Sci USA. 2002, 99: 1503-1508.
    [14]Noss EH, Harding CV, Boom WH. Mycobacterium tuberculosis inhibits MHC class II antigen processing in murine bone marrow macrophages. Cell Immunol. 2000, 201: 63-74.
    [15]Ting LM, Kim AC, Cattamanchi A, et al. Mycobacterium tuberculosis inhibits IFN-gamma transcriptional responses without inhibiting activation of STAT1. J Immunol. 1999, 163:3898 -3906.
    [16] Schluger NW and Rom WN. The host immune response to tuberculosis. Am J Respir Crit Care Med. 1998, 157: 679-691.
    [17]Gebbardt B, Hammarskj?ld ML, Kadner R. Mycobacterium. In: Volk, W. (Ed.) Essentials of Medical Microbiology. Fifth Edition. Philadelphia: Lippincott-Raven Publishers. 1996, 429-439
    [18]Shafer RW and Edlin BR. Tuberculosis in patients infected with human immunodeficiency virus: perspective on the past decade. Clin Infect Dis. 1996, 22: 683-704.
    [19] Okuyama Y, Nakaoka Y, Kimoto K, et al. Tuberculous spondylitis (Pott's disease) with bilateral pleural effusion. Intern. Med. 1996, 35: 883-885.
    [20] Gorse GJ and Belshe RB. Male genital tuberculosis: a review of the literature with instructive case reports. Rev Infect Dis. 1985, 7: 511-524.
    [21] Thwaites G, Chau TT., Mai NT, et al. Tuberculous meningitis. J Neurol Neurosurg Psychiatry. 2000, 68: 289-299.
    [22] Hill AR, Premkumar S, Brustein S, et al. Disseminated tuberculosis in the acquired immunodeficiency syndrome era. Am Rev Respir Dis. 1991, 144: 1164-1170.
    [23] Downing JF, Pasula R, Wright JR, et al. Surfactant protein A promotes attachment of Mycobacterium tuberculosis to alveolar macrophages during infection with human immunodeficiency virus. Proc Natl Acad Sci USA. 1995, 92: 4848-4852.
    [24] Gaynor CD, McCormack FX, Voelker DR, et al. Pulmonary surfactant protein A mediates enhanced phagocytosis of Mycobacterium tuberculosis by a direct interaction with human macrophages. J Immunol. 1995, 155: 5343-5351.
    [25] Quesniaux V, Fremond C, Jacobs M, et al. Toll-like receptor pathways in the immune responses to mycobacteria. Microbes Infect. 2004, 6: 946-959.
    [26] Brightbill HD, Libraty DH, Krutzik SR, et al. Host defense mechanisms triggered by microbial lipoproteins through toll-like receptors. Science. 1999, 285: 732-736.
    [27] Schluger NW. Recent advances in our understanding of human host responses to tuberculosis. Respir Res. 2001, 2: 157-163.
    [28] Means TK, Wang S, Lien E, et al. Human toll-like receptors mediate cellular activation by Mycobacterium tuberculosis. J Immunol. 1999, 163: 3920-3927.
    [29] Bodnar KA, Serbina NV, Flynn JL. Fate of Mycobacterium tuberculosis within murine dendritic cells. Infect. Immun. 2001, 69: 800-809.
    [30] Hertz CJ, Kiertscher SM., Godowski PJ, et al. Microbial lipopeptides stimulate dendritic cell maturation via Toll-like receptor 2. J Immunol. 2001, 166: 2444-2450.
    [31] Gonzalez-Juarrero M, Turner OC, Turner J, et al. Temporal and spatial arrangement of lymphocytes within lung granulomas induced by aerosol infection with Mycobacterium tuberculosis. Infect. Immun. 2001, 69:1722-1728.
    [32] Saunders BM and Cooper AM. Restraining mycobacteria: role of granulomas in mycobacterial infections. Immunol Cell Biol. 2000, 78: 334-341.
    [33] Flesch IE and Kaufmann SH. Activation of tuberculostatic macrophage functions by gamma interferon, interleukin-4, and tumor necrosis factor. Infect Immun. 1990, 58: 2675-2677.
    [34] Cooper AM, Magram J, Ferrante J, et al. Interleukin 12 (IL-12) is crucial to the development of protective immunity in mice intravenously infected with Mycobacterium tuberculosis. J Exp Med. 1997, 186: 39-45.
    [35] Casanova JL and Abel L. Genetic dissection of immunity to mycobacteria: the human model. Annu Rev Immunol. 2002, 20: 581-620.
    [36] Scanga CA, Mohan VP, Yu K, et al. Depletion of CD4(+) T cells causes reactivation of murine persistent tuberculosis despite continued expression of interferon gamma and nitric oxide synthase 2. J Exp Med. 2000.192: 347-358.
    [37] Tan JS, Canaday DH, Boom WH, et al. Human alveolar T lymphocyte responses to Mycobacterium tuberculosis antigens: role for CD4+ and CD8+ cytotoxic T cells and relative resistance of alveolar macrophages to lysis. J Immunol. 1997, 159: 290-297.
    [38] Porcelli SA and Modlin RL. The CD1 system: antigen-presenting molecules for T cell recognition of lipids and glycolipids. Annu Rev Immunol. 1999, 17: 297-329.
    [39] Sousa AO, Mazzaccaro RJ, Russell RG, et al. Relative contributions of distinct MHC class I-dependent cell populations in protection to tuberculosis infection in mice. Proc Natl Acad Sci USA. 2000, 97: 4204-4208.
    [40]师长宏,王晓武,朱德生,等.表达Ag85B与ESAT-6融合蛋白的基因疫苗的免疫学特性.中国结核和呼吸杂志. 2005, 28(11):777-780.
    [41] Serbina NV and Flynn JL. Early emergence of CD8(+) T cells primed for production of type 1 cytokines in the lungs of Mycobacterium tuberculosis-infected mice. Infect Immun. 1999, 67: 3980-3988.
    [42]Boom WH. Gammadelta T cells and Mycobacterium tuberculosis. Microbes Infect. 1999, 1: 187-195.
    [43] Dieli F, Ivanyi J, Marsh P, et al. Characterization of lung gamma delta T cells following intranasal infection with Mycobacterium bovis bacillus Calmette-Guerin. J Immunol. 2003, 170: 463-469.
    [44] Boom WH, Chervenak KA, Mincek MA,et al. Role of the mononuclear phagocyte as an antigen-presenting cell for human gamma delta T cells activated by live Mycobacterium tuberculosis. Infect Immun. 1992, 60: 3480-3488.
    [45] Cooper AM, Dalton DK, Stewart TA, et al. Disseminated tuberculosis in interferon gamma gene-disrupted mice. J Exp Med. 1993, 178: 2243-2247.
    [46]North RJ and Izzo AA. Granuloma formation in severe combined immunodeficient (SCID) mice in response to progressive BCG infection. Tendency not to form granulomas in the lung is associated with faster bacterial growth in this organ. Am J Pathol. 1993, 142: 1959-1966.
    [47]Fenton MJ and Vermeulen MW. Immunopathology of tuberculosis: roles of macrophages and monocytes. Infect Immun. 1996, 64: 683-690.
    [48]Saunders BM and Cooper AM. Restraining mycobacteria: role of granulomas in mycobacterial infections. Immunol Cell Biol. 2000, 78: 334-341.
    [49]Chan J, Xing Y, Magliozzo RS, et al. Killing of virulent Mycobacterium tuberculosis by reactive nitrogen intermediates produced by activated murine macrophages. J Exp Med. 1992, 175:1111-1122.
    [50]Pedroza-Gonzalez A, Garcia-Romo GS, Aguilar-Leon D, et al. In situ analysis of lung antigen- presenting cells during murine pulmonary infection with virulent Mycobacterium tuberculosis. Int J Exp Pathol. 2004, 85: 135-145.
    [51]Garcia-Romo GS, Pedroza-Gonzalez A, Aguilar-Leon D, et al. Airways infection with virulent Mycobacterium tuberculosis delays the influx of dendritic cells and the expression of costimulatory molecules in mediastinal lymph nodes. Immunol. 2004, 112:661-668.
    [52]Gumperz JE, Brenner MB. CD1-specific T cells in microbial immunity. Curr Opin Immunol. 2001, 13: 471-478.
    [53]Engering AJ, Cella M, Fluitsma D, et al. The mannose receptor functions as a high capacity and broad specificity antigen receptor in human dendritic cells. Eur J Immunol. 1997, 27: 2417-2425.
    [54]Fanger NA, Wardwell K, Shen L, et al. Type I (CD64) and type II (CD32) Fc gamma receptor-mediated phagocytosis by human blood dendritic cells. J Immunol. 1996, 157: 541-548.
    [55]Geijtenbeek TB, Van Vliet SJ, Koppel EA, et al. Mycobacteria target DC-SIGN to suppress dendritic cell function. J Exp Med. 2003, 197: 7-17.
    [56]Tailleux L, Schwartz O, Herrmann JL, et al. DC-SIGN is the major Mycobacterium tuberculosis receptor on human dendritic cells. J Exp Med. 2003, 197: 121-127.
    [57]Figdor CG, van Kooyk Y, Adema GJ. C-type lectin receptors on dendritic cells and Langerhans cells. Nat Rev Immunol. 2002, 2: 77-84.
    [58]Jarrossay D, Napolitani G, Colonna M,et al. Specialization and complementarity in microbial molecule recognition by human myeloid and plasmacytoid dendritic cells. Eur J Immunol. 2001, 31: 3388-3393.
    [59]Kadowaki N, Ho S, Antonenko S, et al. Subsets of human dendritic cell precursors express different toll-like receptors and respond to different microbial antigens. J Exp Med. 2001, 194: 863-869.
    [60]Means TK, Jones BW, Schromm AB, et al. Differential effects of a Toll-like receptor antagonist on Mycobacterium tuberculosis-induced macrophage responses. J Immunol. 2001, 166: 4074-4082.
    [61] Means TK, Wang S, Lien E, et al. Human toll-likereceptors mediate cellular activation by Mycobacterium tuberculosis. J Immunol. 1999, 163: 3920-3927.
    [62] Dieu MC, Vanbervliet B, Vicari A, et al. Selective recruitment of immature and mature dendritic cells by distinct chemokines expressed in different anatomic sites. J Exp Med. 1998, 188:373-386.
    [63] Kriehuber E, Breiteneder-Geleff S, Groeger M, et al. Isolation and characterization of dermal lymphatic and blood endothelial cells reveal stable and functionally specialized cell lineages. J Exp Med. 2001, 194: 797-808.
    [64] Bhatt K, Hickman SP, Salgame P. Cutting edge: a new approach to modeling early lung immunity in murine tuberculosis. J Immunol. 2004, 172: 2748-2751.
    [65] Turley SJ, Inaba K, Garrett WS, et al. Transport of peptide-MHC class II complexes in developing dendritic cells. Science. 2000, 288: 522-527.
    [66] Steinman RM. Dendritic cells and the control of immunity: enhancing the efficiency of antigen presentation. Mt Sinai J Med. 2001, 68: 160-166.
    [67] Bodnar KA, Serbina NV, Flynn JL. Fate of Mycobacterium tuberculosis within murine dendritic cells. Infect Immun. 2001, 69: 800-809.
    [68]Fortsch D, Rollinghoff M, Stenger S. IL-10 converts human dendritic cells into macrophage-like cells with increased antibacterial activity against virulent Mycobacterium tuberculosis.J Immunol. 2000, 165: 978-987.
    [69]Giacomini E, Iona E, Ferroni L, et al. Infection of human macrophages and dendritic cells with Mycobacterium tuberculosis induces a differential cytokine gene expression that modulates T cell response. J Immunol. 2001, 166: 7033-7041.
    [70]Hanekom WA, Mendillo M, Manca C, et al. Mycobacterium tuberculosis inhibits maturation of human monocyte-derived dendritic cells in vitro. J Infect Dis. 2003, 188: 257-266.
    [71]Jiao X, Lo-Man R, Guermonprez P, et al. Dendritic cells are host cells for mycobacteria in vivo that trigger innate and acquired immunity. J Immunol. 2002, 168: 1294-1301.
    [72]Stenger S, Hanson DA, Teitelbaum R, et al. An antimicrobial activity of cytolytic T cells mediated by granulysin. Science. 1998, 282: 121-125.
    [73]Lopez-Marin LM, Segura E, Hermida-Escobedo C, et al. 6,6′-Dimycoloyl trehalose from a rapidly growing Mycobacterium: an alternative antigen for tuberculosis serodiagnosis. FEMS Immunol Med Microbiol. 2003, 36: 47-54.
    [74]Ebner S, Ratzinger G, Krosbacher B, et al. Production of IL-12 by human monocyte derived dendritic cells is optimal when the stimulus is given at the onset of maturation, and is further enhanced by IL-4. J Immunol. 2001, 166: 633-641.
    [75]Wozniak TM, Ryan AA, Triccas JA, et al. Plasmid interleukin-23 (IL-23), but not plasmid IL-27, enhances the protective efficacy of a DNA vaccine against Mycobacterium tuberculosis infection. Infect Immun. 2006, 74:557-565.
    [76] Kadowaki N, Ho S, Antonenko S, et al. Subsets of human dendritic cell precursors express different toll-like receptors and respond to different microbial antigens. J Exp Med. 2001, 194: 863-869.
    [77] Humphreys IR, Stewart GR, Turner DJ, et al. A role for dendritic cells in the dissemination of mycobacterial infection. Microbes Infect. 2006, 8: 1339-1346.
    [78] Sakula A. BCG: who were Calmette and Guerin? Thorax. 1983, 38: 806-812.
    [79] Fine PE and Rodrigues LC. Modern vaccines. Mycobacterial diseases. Lancet.1990,335: 1016 -1020.
    [80] Chennai. Fifteen year follow up of trial of BCG vaccines in south India for tuberculosis prevention. Indian J Med Res. 1999, 110: 56-69.
    [81] Hess J and Kaufmann SH. Live antigen carriers as tools for improved anti-tuberculosis vaccines. FEMS Immunol Med Microbiol. 1999, 23: 165-173.
    [82] Falero-Diaz G, Challacombe S, Banerjee D, et al. Intranasal vaccination of mice against infection with Mycobacterium tuberculosis. Vaccine. 2000, 18: 3223-3229.
    [83] Lyadova IV, Vordermeier HM, Eruslanov EB, et al. Intranasal BCG vaccination protects BALB/c mice against virulent Mycobacterium bovis and accelerates production of IFN-gamma in their lungs. Clin Exp Immunol. 2001, 126: 274-279.
    [84] Lewis KN, Liao R, Guinn KM, et al. Deletion of RD1 from Mycobacterium tuberculosis mimics bacille Calmette- Guerin attenuation. J Infect Dis. 2003, 187:117-123.
    [85] Coler RN, Campos-Neto A, Ovendale P, et al. Vaccination with the T cell antigen MTB 8.4 protects against challenge with Mycobacterium tuberculosis. J Immunol. 2001, 166:6227-6235.
    [86] Brandt L, Elhay M, Rosenkrands I, et al. ESAT-6 subunit vaccination against Mycobacterium tuberculosis. Infect Immun. 2000, 68:791-795.
    [87]王利娴,卢贤瑜.结核杆菌分泌性蛋白及相关疫苗研究进展.国际生物制品学杂志. 2006, 29(4): 157-159.
    [88] Gey Van Pittius NC, Gamiedien J, Hide W, et al. The ESAT-6 gene cluster of Mycobacterium tuberculosis and other high G+C Gram-positive bacteria. Genome Bio. 2001, 2(10):1-18.
    [89] Horwitz MA, Harth G, Dillon BJ, et al. Recombinant Bacillus Galmette-Guerin (BCG) vaccines expressing the Mycobacterium tuberculosis 30-kDa major secretory protein induce greater protective immunity against tuberculosis than conventional BCG vaccines in a highly susceptible animan model. Proc Natl Acad Sci USA. 2000, 97:13853-13858.
    [90] Neuwald AF, Aravind L, Spouge JL, et al. AAA+: a class of chaperone-like ATPases associated with the assembly, operation, and disassembly of protein complexes. Genome Res. 1999, 9:27-43.
    [91] Pallen MJ. The ESAT-6/WXG100 superfamily-and new Gram-positive secretion system? Trends Microbiol. 2002, 10:209-212.
    [92] Renshaw PS, Panagiotidou P, Whelan A, et al. Conclusive evidence that the major T-cell antigen of the M. tuberculosis complex ESAT-6 and CFP-10 from a tight, 1:1 complex and characterisation of the structural properties of ESAT-6, CFP-10 and the ESAT-6-CFP-10 complex: implications for pathogenesis and virulence. J Biol Chem. 2002, 277(24):21598-21603.
    [93] Jones BE, Young SM, Antoniskis D, et al. Relationship of the manifestations of tuberculosis to CD4 cell counts in patients with human immunodeficiency virus infections. Am Rev Respir Dis. 1993, 148: 1292-1297.
    [94] Goletti D, Weissman D, Jackson RW, et al. Effect of Mycobacterium tuberculosis on HIV replication. Role of immune activation. J Immunol. 1996, 157: 1271-1278.
    [95] Mariani F, Goletti D, Ciaramella A, et al. Macrophage response to Mycobacterium tuberculosis during HIV infection: relationships between macrophage activation and apoptosis. Curr Mol Med. 2001, 1: 209-216.
    [96] Nakata K, Rom WN, Honda Y, et al. Mycobacterium tuberculosis enhances human immunodeficiency virus-1 replication in the lung. Am J Respir Crit Care Med. 1997, 155: 996–1003.
    [97] Sutherland R, Yang H, Scriba TJ, et al. Impaired IFN-gamma-secreting capacity in mycobacterial antigen -specific CD4 T cells during chronic HIV-1 infection despite longterm HAART. AIDS. 2006, 20:821-829.
    [98] Van Rie A, Warren R, Richardson M, et al. Exogenous reinfection as a cause of recurrent tuberculosis after curative treatment. N Engl J Med. 1999, 341: 1174-1179.
    [99] Van Rie A, Victor TC, Richardson M, et al. Reinfection and mixed infection cause changing Mycobacterium tuberculosis drug-resistance patterns. Am J Respir Crit Care Med. 2005, 172:636 -42.
    [100] Whalen CC, Johnson JL, Okwera A, et al. A trial of three regimens to prevent tuberculosis in Ugandan adults infected with the human immunodeficiency virus. Uganda-Case Western Reserve University Research Collaboration. N Engl J Med. 1997, 337: 801-808.
    [101] Nor NM. and Musa M. Approaches towards the development of a vaccine against tuberculosis: recombinant BCG and DNA vaccine. Tuberculosis (Edinb). 2004, 84:102-109.
    [102] Horwitz MA, Harth G, Dillon BJ, et al. Recombinant bacillus calmette-guerin (BCG) vaccines expressing the Mycobacterium tuberculosis 30-kDa major secretory protein induce greater protective immunity against tuberculosis than conventional BCG vaccines in a highly susceptible animal model. Proc Natl Acad Sci USA. 2000, 97:13853-13858.
    [103] Smith DA, ParishT, Stoker NG, et al. Characterization of auxotrophic mutants of Mycobacterium tuberculosis and their potential as vaccine candidates. Infect. Immun. 2001, 69: 1142-1150.
    [104] Coler RN, Campos-Neto A, Ovendale P,et al. Vaccination with the T cell antigen Mtb 8.4 protects against challenge with Mycobacterium tuberculosis. J Immunol. 2001, 166:6227 -6235.
    [105] Skeiky YA, Ovendale PJ, Jen S,et al. T cell expression cloning of a Mycobacterium tuberculosis gene encoding a protective antigen associated with the early control of infection. J Immunol. 2000, 165: 7140-7149.
    [106] Mustafa AS, Amoudy HA, Wiker HG, et al. Comparison of antigen-specific T-cell responses of tuberculosis patients using complex or single antigens of Mycobacterium tuberculosis. Scand J Immunol. 1998, 48:535-543.
    [107] Brodin P, Rosenkrands I, Andersen P, et al. ESAT-6 proteins: protective antigens and virulence factors? Trends Microbiol. 2004, 12: 500-508.
    [108] Silva CL. The potential use of heat-shock proteins to vaccinate against mycobacterial infections. Microbes Infect. 1999, 1:429-435.
    [109] Lefevre P, Braibant M, De Wit L, et al. Three different putative phosphate transport receptors are encoded by the Mycobacterium tuberculosis genome and are present at the surface of Mycobacterium bovis BCG. J Bacteriol. 1997, 179:2900-2906.
    [110] Thoma-Uszynski S, Kiertscher SM, Ochoa MT, et al. Activation of toll-like receptor 2 on human dendritic cells triggers induction of IL-12, but not IL-10. J Immunol. 2000, 165: 3804-3810.
    [111] Neufert C, Pai RK, Noss EH, et al. Mycobacterium tuberculosis 19-kDa lipoprotein promotes neutrophil activation. J Immunol. 2001, 167:1542-1549.
    [112] Bonato VL, Lima VM, Tascon RE, et al. Identification and characterization of protective T cells in hsp65 DNA-vaccinated and Mycobacterium tuberculosis-infected mice. Infect Immun. 1998, 66: 169-175.
    [113] Fonseca DP, Benaissa-Trouw B, Van Engelen M, et al. Induction of cell-mediated immunity against Mycobacterium tuberculosis using DNA vaccines encoding cytotoxic and helper T-cell epitopes of the 38-kilodalton protein. Infect Immun. 2001, 69: 4839-4845.
    [114] Huygen K, Content J, Denis O, et al. Immunogenicity and protective efficacy of a tuberculosis DNA vaccine. Nat Med. 1996, 2:893-898.
    [115] Manam S, Ledwith BJ, Barnum AB, et al. Plasmid DNA vaccines: tissue distribution and effects of DNA sequence, adjuvants and delivery method on integration into host DNA. Intervirology. 2000, 43:273-281.
    [116]李晖,李榕,钟森,等.结核病Mtb814基因疫苗构建、表达及诱导的细胞免疫应答.解放军医学杂志. 2006, 31(6):556-558.
    [117]Jones BD, Ghori N, Falkow S. Salmonella typhimurium initiates murine infection by penetrating anddestroying the specialized epithelial M cells of the Peyer’s patches. J Exp Med. 1994, 180:15-23.
    [118] Pehheiter KL, Mathur N, Giles D, et al. Non-invasive Salmonella typhimurium mutants are avirulent because of an inability to enter and destroy M cells of ileal Peyer’s patches. Mol Microbiol. 1997, 24:697-709.
    [119] Vasquez-Torres A, Jones-Carson J, B?umler AJ, et al. Extraintestinal dissemination of Salmonella by CD18 -expressing phagocytes. Nature. 1999, 401:804-808.
    [120] Rescigno M, Urbano M, Valzasina B, et al. Dendritic cells express tight junction proteins and penetrate gut epithelia monolayers to sample bacteria. Nat Immunol. 2001, 2:361-367.
    [121] Iwasaki A, Kelsall BL. Unique functions of CD11b– CD8α+ and double-negative Peyer’s patch dendritic cells. J Immunol. 2001, 166:4884-4890.
    [122] Buchmeier, N.A and Heffron F. Induction of Salmonella stress proteins upon infection of macrophages. Infect. Immun. 1989, 57 (1):1-7.
    [123]Yrlid U, Svensson M, H?kansson A, et al. In vivo activation of dendritic cells and T cells during Salmonella enterica serovar Typhimurium infection. Infect Immun. 2001, 69(9):5726-5735.
    [124]Yrlid U, Wick MJ. Antigen presentation capacity and cytokine production by murine splenic dendritic cell subsets upon Salmonella encounter. J Immunol. 2002, 169:108-116.
    [125]Svensson M, Johansson C, Wick MJ. Salmonella enterica serovar typhimurium-induced maturation of bone marrow-derived dendritic cells. Infect Immun. 2000, 68:6311-6320.
    [126] Wick MJ, Ljunggren HG. Processing of bacterial antigens for peptide presentation on MHC class I molecules.Immunol Rev. 1999, 172 :153-162.
    [127]Svensson M, Wick MJ. Classical MHC-I peptide presentation of a bacterial fusion protein by bone marrow -derived dendritic cells. Eur J Immunol. 1999, 29 :180-188.
    [128] Yrlid U, Svensson M, Kirby AC, Wick M.J. Antigen-presenting cells and anti-Salmonella immunity. Microb Infect. 2001, (14)3:1239-1248.
    [129] Macnab RM. Genetics biogenesis of bacterial flagella. Annu Rev Genet. 1992, 26:131–158.
    [130] Yonekura K, Maki-Yonekura S, Namba K. Complete atomic model of the bacterial flagellar filament by electron cryomicroscopy. Nature. 2003, 424:643-650
    [131] Joys TM. The covalent structure of the phase-1 flagellar filament protein of Salmonella typhimurium and its comparison with other flagellins. J Biol Chem. 1985, 260(29):15758–15761.
    [132] Wei LN and Joys TM. Covalent structure of three phase-1 flagellar filament proteins of Salmonella. J Mol Biol. 1985, 186:791–803.
    [133] Smith NH, Selander RK. Sequence invariance of the antigen-coding central region of the phase 1 flagellar. J Bacteriol. 1990, 172:603–609.
    [134] Samatey FA, Imada K, Nagashima S, et al. Structure of the bacterial flagellar protofilament and implications for a switch for supercoiling. Nature. 2001, 410:331–337.
    [135] Yonekura K, Maki-Yonekura S, Namba K. Complete atomic model of the bacterial flagellar filament by electron cryomicroscopy. Nature.2003,424:643–650.
    [136] Joys TM and Schodel F. Epitope mapping of the d flagellar antigen of Salmonella muenchen. Infect Immun. 1991, 59:3330–3332.
    [137] McDermott PF, Ciacci-Woolwine F, Snipes JA, et al. High-affinity interaction between gram-negtive flagellin and a cell surface polypeptide results in human monocyte activation. Infect Immun. 2000, 68:5525-5529.
    [138] Steiner TS, Nataro JP, Poteet-Smith CE, et al. Enteroaggregative Escherichia coli expresses a novel flagellin that causes IL-8 release from intestinal epithelial cells. J Clin Invest. 2000, 105:1769-1777.
    [139] Eaves-Pyles TD, Wong HR, Odoms K, et al. Salmonella flagellin-dependent proinflammatory responses are localized to the conserved amino and carboxyl regions of the protein. J Immunol. 2001, 167:7009-7016.
    [140] Donnelly MA and Steiner T S. Two nonadjacent regions in enteroaggregative Escherichia coli flagellin are required for activation of toll-like receptor 5. J Biol Chem. 2002, 277:40456- 40461.
    [141] Cario E and Podolsky DK. Differential alteration in intestinal epithelial cell expression of toll-like receptor 3 (TLR3) and TLR4 in inflammatory bowel disease. Infect Immun. 2000, 68:7010–7017.
    [142] Muzio M, Bosisio D, Polentarutti N, et al. Differential expression and regulation of toll-like receptors (TLR) in human leukocytes: selective expression of TLR3 in dendritic cells. J Immunol. 2000, 164:5998–6004.
    [143] Hayashi F, Smith KD, Ozinsky A, et al. The innate immune response to bacterial flagellin is mediated by Toll-like receptor 5. Nature. 2001, 410:1099–1103.
    [144] Gewirtz AT, Nava TA, Lyons S, et al. Cutting edge: bacterial flagellin activates basolaterally expressed TLR-5 to induce epithelial proinflammatory gene expression. J Immunol. 2001, 167: 1882–1885.
    [145] Homma M, Fujita H, Yamaguch S, et al. Regions of Salmonella typhimurium flagellin essential for its polymerization and excretion. J Bacteriol. 1987, 169:291-296.
    [146] Kuwajima G. Construction of a minimum-size functional flagellin of Escherichia coli. J Bacteriol. 1988, 170: 3305-3309.
    [147]Okazaki N, Matsuo S, Saito K, et al. Conversion of the Salmonella phase 1flagellin gene fliC to the phase 2 gene fljB on the Escherichia coli K-12 Chromosome. J Bacteriol. 1993, 175(3):758 -766.
    [148]曹军平,焦新安,杜元钊,等.沙门氏菌鞭毛蛋白的提纯与鉴定. 1996, 1:14-16.
    [149]焦新安,倪振亚,张如宽,等.肠炎沙门氏菌鞭毛蛋白基因快速克隆和鉴定. 1998, 19(3):43-47.
    [150]Salete M, Newton C, Chaim O, et al. Immune response to cholera toxin epitope inserted in Salmonella Flagellin. Science. 1989, 244:70-72.
    [151]Naresh K, Verma H, Kirk Z, et al. Induction of a cellular immune response to a defined T-cell epitope as an insert in the flagellin of a live vaccine strain of Salmonella. Vaccine. 1995, 13(3):235-244.
    [152]Raphael L and Ruth A. Synthetic recombinant influenza vaccine induces efficient long-term immunity and cross-strain protection. Vaccine. 1996, 14:85-92.
    [153] Ben-Yedidia T, Tarrab-Hazdai R, Schechtman D, et al. Intranasal administration of synthetic recombinant peptide-based vaccine protects mice from infection by Schistosoma mansoni. Infect Immun. 1999, 67: 4360–4366.
    [154] Carsiotis M, Weinstein DL, Karch H, et al. Flagella of Salmonella typhimurium are a virulence factor in infected C57BL/6J mice. Infect. Immun. 1984, 46:814–818.
    [155] Wolfgang MC, Jyot J, Goodman AL, et al. Pseudomonas aeruginosa regulates flagellin expression as part of a global response to airway fluid from cystic fibrosis patients. Proc Natl Acad Sci USA. 2004, 101:6664–6668.
    [156] Zeng H, Carlson AQ, Guo Y, et al. Flagellin is the major proinflammatory determinant of enteropathogenic Salmonella. J. Immunol. 2003, 171:3668–3674.
    [157] Jeon SH, Ben-Yedidia T, and Arnon R. Intranasal immunization with synthetic recombinant vaccine containing multiple epitopes of influenza virus. Vaccine. 2002, 20:2772–2780.
    [158] McSorley SJ, Ehst BD, Yu Y, et al. Bacterial flagellin is an effective adjuvant for CD4+ T cells in vivo. J Immunol. 2002, 169:3914–3919.
    [159]Anna NH and Steven BM. Mucosal administration of flagellin induces innate immunity in the mouse lung. Infect Immun. 2004, 72(11):6676-6679.
    [160] Oscar P, Michael M and Suzanne M.M. Cellular mechanisms of the adjuvant activity of the flagellin component FljB of Salmonella enterica Serovar Typhimurium to potentiate mucosal and systemic responses. Infect Immun. 2005, 73(10): 6763–6770.
    [161] Matam VK, Huixia W, Rheinallt J, et al. Flagellin suppresses epithelial apoptosis and limits disease during enteric infection. J America Patho. 2006, 169(5):1686-1700.
    [162] Hui Z, Huixia W, Valerie S, et al. Flagellin/TLR-5 responses in epithelia reveal intertwined activation of inflammatory and apoptotic pathways. Am J Physiol Gastrointest Liver Physiol . 2005, 290:96-108
    [163] McSorley SJ, Ehst BD, Yu Y, et al. Bacterial flagellin is an effective adjuvant for CD4+ T cells invivo. J Immunol. 2002, 169:3914–3919.
    [164] Means TK, Hayashi F, Smith KD, et al. The Toll-like receptor 5 stimulus bacterial flagellin induces maturation and chemokine production in human dendritic cells. J Immunol . 2003, 170: 5165–5175.
    [165] Didierlaurent A, Ferrero I, Otten LA, et al. Flagellin promotes myeloid differentiation factor 88-dependent development of Th2-type response. J Immunol. 2004, 172:6922–6930.
    [166] Agrawal S, Agrawal A, Doughty B, et al. Cutting edge: different toll-like receptor agonists instruct dendritic cells to induce distinct Th responses via differential modulation of extracellular signal-regulated kinase–mitogen -activated protein kinase and c-Fos. J Immunol. 2003, 171: 4984–4989.
    [167] Strindelius L, Filler M, Sjoholm I. Mucosal immunization with purified flagellin from Salmonella induces systemic and mucosal immune responses in C3H/HeJ mice. Vaccine. 2004, 22:3797–3808.
    [168] Hess J, Ladel C, Miko D, et al. Salmonella typhimurium aroA-infection in gene-targeted immunodeficient mice: major role of CD4+ TCR-alpha beta cells and INF-gamma in bacterial clearance independent of intracellular location. J Immunol. 1996, 156:3321–3326.
    [169] Srinivasan A, Foley J, McSorley SJ. Massive number of antigenspecific CD4+ T cells during vaccination with live attenuated Salmonella causes interclonal competition. J Immunol. 2004, 172: 6884–6893.
    [170] Cunningham AF, Khan M, Ball J, et al. Responses to the soluble flagellar protein FliC are Th2, while those to FliC on Salmonella are Th1. Eur J Immunol. 2004, 34: 2986–2995.
    [171] Kubo T, Hatton RD, Oliver J, et al. Regulatory T cell suppression and anergy are differentially regulated by proinflammatory cytokines produced by TLR-activated dendritic cells. J Immunol. 2004, 173: 7249–7258.
    [172] Crellin NK, Garcia RV, Hadisfar O, et al. Human CD4+ T cells express TLR5 and its ligand flagellin enhances the suppressive capacity and expression of Foxp3 in CD4+CD25+T regulatory cells. J Immunol. 2005,175: 8051–8059.
    [173] McKee AS and Pearce EJ. CD25+CD4+ T cells contribute to Th2 polarization during helminth infection by suppressing Th1 response development. J Immunol. 2004, 173: 1224–1231.
    [174] Aseffa AA, Gumy PL, MacDonald HR, et al. The early IL-4 response to Leishmania major and theresulting Th2 cell maturation steering progressive disease in BALB/c mice are subject to the control of regulatory CD4+CD25+T cells. J Immunol. 2002, 169:3232–3241.
    [175] Lucia S, Anna R, Dario B, et al. Antitumor activity of the TLR-5 ligand flagellin in mouse models of cancer. J Immunol. 2006, 176:6624-6630.
    [1] World Health Organization. Tuberculosis. WHO fact sheet no 104. Geneva: WHO. 2002.
    [2]Corbett EL, Watt CJ, Walker N, et al. The growing burden of tuberculosis: global trends and interactions with the HIV epidemic. Arch Intern Med. 2003,163:1009-1021.
    [3]De Cock KM and Chaisson RE. Will DOTS do it? A reappraisal of tuberculosis control in countries with high rates of HIV infection. Int J Tuberc Lung Dis. 1999,3: 457-465.
    [4] Brodin P, Rosenkrands I, Andersen P,et al. ESAT-6 proteins: protective antigens and virulence factors? Trends Microbiol. 2004,12(11):500-508.
    [5] Young D.B, Kaufmann SHE, Hermans PW, et al. Mycobacterial protein antigens: a compilation, Mol. Microbiol. 1992,6:133–145.
    [6] Alexander SP, Priscille B, Laleh M, et al. Recombinant BCG exporting ESAT-6 confers enhanced protection against tuberculosis. Nature Medicine. 2003,9(5):533-539.
    [7] Lewis KN, Liao R, Guinn KM, et al. Deletion of RD1 from Mycobacterium tuberculosis mimics bacille Calmette-Guérin attenuation. J Infect Dis. 2003,187:117 -123.
    [8] Skj?t RL, Oettinger T, Rosenkrands I, et al. Comparative evaluation of low -molecular-mass proteins from Mycobacterium tuberculosis identifies members of the ESAT-6 family as immunodominant T-cell antigens. Infect Immun.2000, 68(1):214-220.
    [9] Renshaw PS, Panagiotidou P, Whelan A, et al. Conclusive evidence that the major T-cell antigen of the M. tuberculosis complex ESAT-6 and CFP-10 from a tight, 1:1 complex and characterisationof the structural properties of ESAT-6, CFP-10 and the ESAT-6-CFP-10 complex: implications for pathogenesis and virulence. J Biol Chem. 2002,277(24):21598-21603.
    [9]王国治.结核菌素的制造与质量控制.中国防痨杂志。1991,13(3) :140– 142.
    [11] Behr MA, Wilson MA, Gill WP, et al . Comparative genomics of BCG vaccines by whole-genome DNA microarray. Science. 1999, 284 :1520– 1522
    [12] Silva CL. The potential use of heat-shock proteins to vaccinate against mycobacterial infections. Microbes Infect. 1999,1: 429-435.
    [13] Lefevre P, Braibant M, De Wit L, et al. Three different putative phosphate transport receptors are encoded by the Mycobacterium tuberculosis genome and are present at the surface of Mycobacterium bovis BCG. J Bacteriol. 1997, 179: 2900-2906.
    [14] Thoma-Uszynski S, Kiertscher SM, Ochoa MT, et al. Activation of toll-like receptor 2 on human dendritic cells triggers induction of IL-12, but not IL-10. J Immunol. 2000,165: 3804-3810.
    [15] Neufert C, Pai RK, Noss EH,et al. Mycobacterium tuberculosis 19-kDa lipoprotein promotes neutrophil activation. J Immunol. 2001,167: 1542-1549.
    [16].Chapman AL , Munkanta M, Wilkinson KA , et al . Rapid detection of active and latent tuberculosis infection in HIV-positive individuals by enumeration of Mycobacterium tuberculosis specific T cells. AIDS. 2002, 16(17) : 2285– 2293
    [17]Ravn P , Munk ME , Andersen AB , et al . Prospective evaluation of a whole-blood test using Mycobacterium tuberculosis specific antigens ESAT-6 and CFP-10 for diagnosis of tuberculosis. Clin Diag Lab Immunol. 2005, 12 (4) :491– 496
    [18]Gey Van Pittius NC, Gamieldien J, Hide W, et al. The ESAT-6 gene cluster of Mycobacterium tuberculosis and other high G+C Gram-positive bacteria. Genome Biol. 2001, 2(10):1-18.
    [19] Daugelat S, Kowall J, Mattow J, et al. The RD1 proteins of Mycobacterium tuberculosis: expression in Mycobacterium smegmatis and biochemical characterization. Microbes Infect. 2003, 5(12):1082-1095.
    [20] Berthet FX, Rasmussen PB, Rosenkrands I, et al. A Mycobacterium tuberculosis operon encoding ESAT-6 and a novel low-molecular-mass culture filtrate protein (CFP-10). Microbiology. 1998, 144:3195–3203.
    [21] Munk ME, Arend SM, Broc I k, et al. Use of ESAT-6 and CFP-10 antigens for diagnosis of extrapulmonary tuberculosis. J Infect Dis. 2001,183:175–176.
    [22] Brusasca PN, Colangeli R, Lyashchenko KP, et al. Immunological characterization of antigens encoded by the RD1 region of the Mycobacterium tuberculosis genome. Scand J Immunol. 2001, 54:448–452.
    [23] Laleh M, Marcela S, Zdenka J, et al. An increse in Antimycobacterial Th1-cell responses by prime-boost protocols of immunization does not enhance protection against Tuberculosis. Infect Immun. 2006,74(4):2128-2137.
    [24] Philip C.H, Dolly J.S, Annette F, et al. ESAT-6/CFP-10 Fusion protein and peptides for optimal diagnosis of Mycobacterium tuberculosis infection by ex vivo Enzyme-linked immunospot assay in the Gambia. J Clin Micro. 2005, 43(5):2070-2074.
    [25]Van Pinxteren LAH , Ravn P, Agger EM, et al. Diagnosis of tuberculosis based on the two specific antigens ESAT-6 and CFP-10. Clin Diag Lab Immun. 2000, 7:155-160.
    [26] Fabrizio V, Stefano R, Enrico P, et al. ESAT-6 peptide recognition by Bovine CD8+ lymphocytes of naturally infected cows in herds from southern Italy.Clin Vaccine Immuno. 2006, 13(4): 530-533.
    [27] Brock I, Weldeingh K, Leyten EM, et al. Specific T-cell epitopes for immunoassay-based diagnosis of Mycobacterium tuberculosis infection. J Clin Microbiol. 2004,42(6):2379-2387.
    [1] RL Riley, Mills CC, Nyka W, et al. Aerial dissemination of pulmonary tuberculosis. A two-year study of contagion in a tuberculosis ward. Am J Epidemiol. 1995, 142: 3-14.
    [2] Frieden TR, Sterling TR, Munsiff SS,et al. Tuberculosis. Lancet. 2003, 362: 887-899.
    [3] Dietrich J, Andersen C, Rappuoli R, et al. Mucosal administration of Ag85B-ESAT-6 protects against infection with Mycobacterium tuberculosis and boosts prior bacillus Calmette-Gue′rin immunity. J Immunol. 2006, 177:6353–6360.
    [4] Santosuosso M, Zhang X, McCormick S, et al. Mechanisms of mucosal and parenteral tuberculosis vaccinations: adenoviral-based mucosal immunization preferentially elicits sustainedaccumulation of immune protective CD4 and CD8T cells within the airway lumen. J Immunol. 2005, 174:7986–7994.
    [5] Wu HY, Nikolova EB, Beagley KW, et al. Development of antibody-secreting cells and antigen -specific T cells in cervical lymph nodes after intranasal immunization. Infect Immun. 1997, 65:227–235.
    [6] Santosuosso M, McCormick S, Roediger E, et al. Mucosal luminal manipulation of T cell geography switches on protective efficacy by otherwise ineffective parenteral genetic immunization. J Immunol. 2007, 178(4):2387-2395.
    [7] Wang J, Thorson L, Stokes RW, et al. Single mucosal, but not parenteral, immunization with recombinant adenoviral-based vaccine provides potent protection from pulmonary tuberculosis. J Immunol. 2004, 173:6357–6365.
    [8] Sims K, Kevin P, Una S, et al. Advances in the deveplpment of bacterial vector technology. Exp Rev Vaccines. 2003, 2(1): 31-43.
    [9] Brandtzaeg P. History of oral tolerance and mucosal immunity. Ann NY Acad Sci 1996, 778:1–27.
    [10]Hagiwara Y, McGhee JR, Fujihashi K, et al. Protective mucosal immunity in aging is associated with functional CD4+ T cells in nasopharyngeal associated lymphoreticular tissue. J Immunol. 2003, 170:1754–1762.
    [11] Carpenter ZK, Williamson ED, Eyles JE. Mucosal delivery of microparticle encapsulated ESAT-6 induces robust cellmediated responses in the lung milieu. J Control Release. 2005, 104: 67–77.
    [12]Santosuosso M, McCormick S, Zhang X, et al.Intranasal boosting with an Adenovirus- Vectored vaccine markedly enhances protection by parenteral Mycobacterium bovis BCG immunization against pulmonary tuberculosis. Infect Immun. 2006, 74:4634–4643.
    [13]Sambrook J, Russell DW.分子克隆实验指南.黄培堂等译.第三版.北京:科学出版社,2002.
    [14]Xing Z. The hunt for new tuberculosis vaccines: anti-TB immunity and rational design of vaccines. Curr Pharm Des. 2001, 7:1015–1037.
    [15]McMichael AJ. HIV vaccines. Annu. Rev. Immunol. 2006, 24:227–255.
    [16]Glyn Hewinson R. TB vaccines for the World. Tuberculosis 2005, 85:1–6.
    [17]Brandtzaeg P and Pabst R. Let’s go mucosal: communication on slippery ground. Trends Immunol. 2004, 25:570–577.
    [18]Brandtzaeg P, Johansen FE. Mucosal B cells: phenotypic characteristics, transcriptional regulation, and homing properties. Immunol Rev. 2005, 206:32–63.
    [19]Brandtzaeg P. History of oral tolerance and mucosal immunity. Ann NY Acad Sci. 1996, 778: 1–27.
    [20]王芳,焦新安,顾健,等.表达GFP的重组减毒鼠伤寒沙门氏菌感染抗原呈递细胞的动态变化. 2003, 19(5):425-427.
    [21]Coler RN, Vaccination with the T cell antigen MTB 8.4 protects against challenge with Mycobacterium tuberculosis. 2001, 166:6227-6235.
    [22]张辉,焦新安,潘志明,等.减毒鼠伤寒沙门氏菌运送CD8+ T细胞表位的细胞免疫应答.细胞与分子免疫学杂志. 2006, 22(2):137-140
    [23]Lafaille JJ. The role of helper T cell subsets in autoimmune diseases. Cytokine Growth Factor Rev. 1998, 9:139-151.
    [24]Brightbill HD, Libraty DH, Krutzik SR, et al. Host defense mechanisms triggered by microbial lipoproteins through toll-like receptors. Science.1999, 285:732-736.
    [25]Laleh M, Priscille B, Roland B, et al. Influence of ESAT-6 secretion system 1(RD1) of Mycobaterium tuberculosis on the interaction between Mycobacteria and the host immune system. Immuno. 2005, 174:3570-3579.
    [26] K?lleniusa G, Pawlowskia A, Brandtzaegc P,et al. Should a new tuberculosis vaccine be administered intranasally? Tuberculosis. 2007, 87(4):257–266.
    [27]Appleman LJ and Boussiotis VA. T cell anergy and costimulation. Immunol Rev. 2003, 192: 161–180.
    [28]Lenschow DJ, Walunas TL and Bluestone JA. CD28/B7 system of T cell costimulation. Annu Rev Immunol. 1996, 14:233–258.
    [29]Borriello F, Sethna MP, Boyd SD, et al. B7-1 and B7-2 have overlapping, critical roles in immunoglobulin class switching and germinal center formation. Immunity. 1997, 6:303–313.
    [30] Kuchroo VK, Das MP, Brown JA, et al. B7-1 and B7-2 costimulatory molecules activate differentially the Th1/Th2 development pathways: application to autoimmune disease therapy. Cell. 1995, 80:707–718.
    [31]Lanier LL, Fallon SO, Somoza C, et al. CD80 (B7) and CD86 (B70) provide similar costimulatory signals for T cell proliferation, cytokine production, and generation of CTL. J Immunol. 1995, 154:97–105.
    [32] Lespagnard L, Mettens P, De Smedt T, et al. The immune response induced in vivo by dendritic cells is dependent on B7-1 or B7-2, but the inhibition of both signals does not lead to tolerance. Int Immunol. 1998, 10:295–304.
    [33] Kaufmann SH. Recent findings in immunology give tuberculosis vaccines a new boost. Trends Immunol. 2005, 26:660–667.
    [34]Flynn JL, Chan J, Triebold KJ, et al. An essential role for interferon gamma in resistance to Mycobacterium tuberculosis infection. J Exp Med. 1993, 178:2249–2254.
    [35]Flynn JL, Goldstein MM, Triebold KJ,et al. Major histocompatibility complex class I- restricted T cells are required for resistance to Mycobacterium tuberculosis infection. Proc Natl Acad Sci USA. 1992, 89:12013–12017.
    [36]Feng CG, Bean AG, Hooi H, et al. Increase in gamma interferon-secreting CD8(+), as well as CD4(+), T cells in lungs following aerosol infection with Mycobacterium tuberculosis. Infect Immun. 1999, 67:3242–3247.
    [37]Ladel CH, Blum C, Dreher A, et al. Protective role of gamma/delta T cells and alpha/beta T cells in tuberculosis. Eur J Immunol. 1995, 25:2877–2881.
    [1] Kavermann H, Burns BP, Angermüller K, et al. Identification and Characterization of Helicobacter pylori Genes Essential for Gastric Colonization J Exp Med. 2003, 197(7): 813-822.
    [2] Chua KL, Chan YY and Gan YH. Flagella are virulence determinants of Burkholderia pseudomaller. Infect Immun. 2000, 71:1622-1629.
    [3] Dietrich C, Heuner HG, Brand BC, et al. Flagellum of Legionella pneumophila positively affects the early phase of infection of eukaryotic host cells. Infect Immun. 2001, 69:2116-2122.
    [4] Van Asten FJ, Hendriks HG, Koninkx JF, et al. Inactivation of the flagellin gene of Salmonella enterica serotype enteritidis strongly reduces invasion into differentiated Caco-2 cells. FEMS Microbiol Lett. 2000, 185:175-179.
    [5] Cookson BT and Bevan MJ. Identification of a natural T cell epitope presented by Salmonella-infected macrophages and recognized by T cells from orally immunized mice. J Immunol. 1997, 158:4310-4319.
    [6] Hayashi F, Means TK and Luster AD. Toll-like receptors stimulate human neutrophil function. Blood. 2003, 102:2660-2669.
    [7] Means TK, Hayashi F, Smith KD, et al. The Toll-like receptor 5 stimulus bacterial flagellin induces maturation and chemokine production in human dendritic cells. J Immunol. 2003, 170: 5165 -5175.
    [8] Smith NH, Selander RK. Sequence invariance of the antigen-coding central region of the phase 1flagellar. J Bacteriol. 1990, 172:603–609.
    [9] Yonekura K, Maki-Yonekura S, Namba K. Complete atomic model of the bacterial flagellar filament by electron cryomicroscopy. Nature. 2003,424:643-650
    [10] Salete M, Newton C, Chaim O, et al. Immune response to cholera toxin epitope inserted in Salmonella Flagellin. Science. 1989, 244:70-72.
    [11] Naresh K, Verma H, Kirk Z. et al. Induction of a cellular immune response to a defined T-cell epitope as an insert in the flagellin of a live vaccine strain of Salmonella. Vaccine. 1995, 13(3):235 -244.
    [12] Cunningham AF, Khan M, Ball J, et al. Responses to the soluble flagellar protein FliC are Th2, while those to FliC on Salmonella are Th1. Eur J Immunol. 2004, 34:2986–2995.
    [13] New ton SM C, Jacob C O , Stocker B A D. Immune responseto cho lera toxin epitope inserted in Salmonella flagellin. Science. 1989, 244:70-72.
    [14] Bullas L R, Ryu J2I. Salmonella typh imurium LT2 strainswhich are r - m+ for all thee chomosomally located system s of DNA restriction and modification. J Bacteriol. 1983, 156(1): 471 -474.
    [15] Mustafa AS, Shaban FA, Al-Attiyah R, et al. Human Th1 cell lines recognize the Mycobacterium tuberculosis ESAT-6 antigen and its peptides in association with frequently expressed HLA classⅡmolecules. Scand J Immunol. 2003,57(2):125-134.
    [16] Gey van Pittius NC, Warren RM, van Helden PD. ESAT-6 and CFP-10: what is the diagnosis? Infect Immun. 2002, 70(11):6509-6510.
    [17] Munk ME, Arend SM, Brock I, et al. Use of ESAT-6 and CFP-10 antigens for diagnosis of extrapulmonary tuberculosis.J Infect Dis. 2001, 183:175–176.
    [18] Pollock JM, Buddle BM, and Andersen P. Towards more accurate diagnosis of bovine tuberculosis using defined antigens. Tuberculosis. 2001, 81:65–69.
    [19] Maloy S R , Stewart VJ , Taylor R K. Genetic analysis of pathogenic bacteria : A laboratory manual . New York : Cold Spring Harbor Laboratory Press.1996.
    [20] Richard MC, Scott NM, William RH, et al. Progression of Armed CTL from Draining Lymph Node to Spleen Shortly After Localized Infection with Herpes Simplex Virus. J Immunol. 2002, 168: 834–838.
    [21] Inaba K, Inaba M, Romani N, et al. Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor. J Exp Med. 1992, 176(6):1693–1702.
    [22] Testi R, Phillips JH and Lanier LL. T cell activation via Leu-23(CD69). J Immunol. 1989, 143: 11-23.
    [23] Lihao C, Jun W, Anna Z,et al. Single intranasal mucosal Mycobacterium bovis BCG vaccination confers improved protection compared to subcutaneous vaccination against pulmonary Tuberculosis. Infect Immun. 2004, 72(1):238–246
    [24] Tree JA, Williams A, Clark S, et al. Intranasal bacille Calmette-Guérin (BCG) vaccine dosage needs balancing between protection and lung pathology. Clin Exp Immunol. 2004, 138:405-409.
    [25] Gunilla K, Andrzej P, Per B,et al. Should a new tuberculosis vaccine be administered intranasally? Tuberculosis. 2007, 87:257–266.
    [26] Power CA, Wei G, Bretscher PA. Mycobacterial dose de?nes the Th1/Th2 nature of the immune response independently of whether immunization is administered by the intravenous, subcutaneous, or intradermal route. Infect Immun. 1998, 66:5743-5750
    [27] Marshall BG, Wangoo A, O’OGaora P, et al. Enhanced antimycobacterial response to recombinant Mycobacterium bovis BCG expressing latency-associated peptide. Infect Immun. 2001, 69:6676-6682
    [28]申蓉,李丽珍. CD69与免疫功能的调节.国外医学免疫学分册. 2004, 27(1):24-27.
    [1] Eriksson AM, Schon KM, Lycke NY. The cholera toxin-derived CTA1-DD vaccine adjuvant administered intranasally does not cause inflammation or accumulate in the nervous tissues. J Immunol. 2004, 173:3310–3319.
    [2] Takahashi H, Sasaki K, Takahashi M, et al. Mutant Escherichia coli enterotoxin as a mucosal adjuvant induces specific Th1 responses of CD4+ and CD8+ T cells to nasal killed-Bacillus Calmette-Gue′rin in mice. Vaccine. 2006, 24:3591–3598.
    [3] Jakobsen H, Bjarnarson S, Del Giudice G, et al. Intranasal immunization with pneumococcal conjugate vaccines with LT-K63, a nontoxic mutant of heat-Labile enterotoxin, as adjuvant rapidly induces protective immunity against lethal pneumococcal infections in neonatal mice. Infect Immun. 2002, 70:1443–1452.
    [4] Haile M, Schroder U, Hamasur B, et al. Immunization with heat-killed Mycobacterium bovis Bacille Calmette-Guerin (BCG) in Eurocine L3 adjuvant protects against tuberculosis. Vaccine. 2004, 22: 1498–1508.
    [5] Oscar Pino, Michael Martin and Suzanne M. Michalek cellular mechanisms of the adjuvant activity of the flagellin component FljB of Salmonella enterica serovar Typhimurium to potentiate mucosal and systemic responses. Infect Immun. 2005, 73(10):6763–6770.
    [6] Cunningham AF, Khan M, Ball J, et al. Responses to the soluble flagellar protein FliC are Th2, while those to FliC on Salmonella are Th1. Eur J Immunol. 2004, 34:2986–2995.
    [7]Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol. 2004, 4(7):499–511.
    [8] Akira S, Takeda K, Kaisho T. Toll-like receptors: critical proteins linking innate and acquired immunity. Nat Immunol. 2001, 2(8):675–680.
    [9] Iwasaki A, Medzhitov R. Toll-like receptor control of the adaptive immune responses. Nat Immunol. 2004, 5(10):987–995.
    [10] Medzhitov R, Janeway Jr CA. Innate immune induction of the adaptive immune response. Cold Spring Harb Symp Quant Biol. 1999, 64:429–35.
    [11] Liew FY, Parish CR. Regulation of the immune response by antibody. II. The effects of different classes of passive guinea pig antibody on humoral and cell-mediated immunity in rats. Cell Immunol. 1972, 5(4):499–519.
    [12] Parish CR. The relationship between humoral and cell-mediated immunity. Transplant Rev. 1972, 13:35–66.
    [13] Felix G, Duran JD, Volko S, et al. Plants have a sensitive perception system for the most conserved domain of bacterial flagellin. Plant J. 1999, 18(3):265–76.
    [14]Naresh K, Verma H, Kirk Z. et al. Induction of a cellular immune response to a defined T-cellepitope as an insert in the flagellin of a live vaccine strain of Salmonella. Vaccine. 1995, 13(3): 235-244.
    [15]Raphael L and Ruth A. Synthetic recombinant influenza vaccine induces efficient long-term immunity and cross-strain protection. Vaccine. 1996, 14:85-92.
    [16]Sambrook J, Russell DW.分子克隆实验指南.黄培堂等译.第三版.北京:科学出版社,2002.
    [17] Mollenkopf HJ, Hahnke K, Kaufmann SH. Transcriptional responses in mouse lungs induced by vaccination with Mycobacterium bovis BCG and infection with Mycobacterium tuberculosis. Microbes Infect. 2006, 8(1):136-144.
    [18]Honko AN, Mizel SB. Mucosal administration of flagellin induces innate immunity in the mouse lung. Infect Immun. 2004, 72:6676–6679.
    [19]Terry KM, Fumitaka H, Kelly DS, et al. The toll-like receptor 5 stimulus bacterial flagellin induces maturation and chemokine production in human dendritic cell. J Immuno. 2003, 170: 5165 -5175.
    [20]Steven EA, Erik R, Mark DW, et al. Activation of innate immunity, inflammation, and potentiation of DNA vaccination through mammalian expression of the TLR5 agonist Flagellin. J Immuno. 2005, 175: 3882–3891.
    [21]Brown MG, Scalzo AA , Matsumoto K, et al . The natural killer gene complex: a genetic basis for understanding natural killer cell function and innate immunity. Immunol Review. 1997, 155(1) : 53-65.
    [22]Marmiesse M, Brodin P, Buchrieser C, et al. Macro-array and bioinformatic analyses reveal mycobacterial‘core’genes, variation in the ESAT-6 gene family and new phylogenetic markers for the Mycobacterium tuberculosis complex. Microbiology. 2004, 150:483-496.
    [23]Tekaia F, Gordon SV, Garnier T, et al. Analysis of the proteome of Mycobacterium tuberculosis in silico. Tuber Lung Dis. 1999, 79(6):329-342.
    [24]James WH, Andrea RJ, Jie T, et al. Vaccination with recombinant fusion proteins incorporating Toll-like receptor ligands induces rapid cellular and humoral immunity. Vaccine. 2007, 25:763-775.
    [25]Cuadros C, Lopez-Hernandez FJ, Dominguez AL, et al. Flagellin fusion proteins as adjuvants or vaccines induce specific immune responses. Infect Immun. 2004, 72(5):2810-2816.
    [26]Xu Y, Wang B, Chen J, et al. Chimaeric protein improved immunogenicity compared with fusion protein of Ag85B and ESAT-6 antigens of Mycobaterium tuberculosis. Scand J Immunol. 2006, 64(5):476-481.
    [27] Hess J, Ladel C, Miko D, et al. Salmonella typhimurium aroA-infection in gene-targeted immunodeficient mice: major role of CD4+ TCR-alpha beta cells and INF-gamma in bacterial clearance independent of intracellular location. J Immunol. 1996, 156:3321–3326.
    [28] Srinivasan A, Foley J, McSorley SJ. Massive number of antigenspecific CD4+ T cells during vaccination with live attenuated Salmonella causes interclonal competition. J Immunol. 2004, 172: 6884–6893.
    [29] Means TK, Hayashi F, Smith KD, et al. The Toll-like receptor 5 stimulus bacterial flagellin induces maturation and chemokine production in human dendritic cells. J Immunol . 2003, 170: 5165–5175.
    [30] Cunningham AF, Khan M, Ball J, et al. Responses to the soluble flagellar protein FliC are Th2,while those to FliC on Salmonella are Th1. Eur J Immunol. 2004, 34: 2986–2995.
    [31] McSorley SJ, Ehst BD, Yu Y, et al. Bacterial flagellin is an effective adjuvant for CD4+ T cells in vivo. J Immunol. 2002, 169:3914–3919.
    [32] Agrawal S, Agrawal A, Doughty B, et al. Cutting edge: different toll-like receptor agonists instruct dendritic cells to induce distinct Th responses via differential modulation of extracellular signal-regulated kinase–mitogen-activated protein kinase and c-Fos. J Immunol. 2003, 171: 4984–4989.
    [33] Strindelius L, Filler M, Sjoholm I. Mucosal immunization with purified flagellin from Salmonella induces systemic and mucosal immune responses in C3H/HeJ mice. Vaccine. 2004, 22:3797–3808.
    [34]Chen L, Wang J, Zganiacz A, et al. Single Intranasal Mucosal Mycobacterium bovis BCG Vaccination Confers Improved Protection Compared to Subcutaneous Vaccination against pulmonary Tuberculosis. Infect Immun. 2004, 72(1):238-46.
    [35] Tree JA, Williams A, Clark S, et al.Intranasal Bacille Calmette-Guérin(BCG) vaccine dosage needs balancing between protection and lung pathology. Clin Exp Immunol. 2004, 138:405-409.
    [36] Brandtzaeg P. History of oral tolerance and mucosal immunity. Ann NY Acad Sci. 1996, 778: 1–27.
    [37] Rudin A, Johansson EL, Bergquist C, et al. Differential kinetics and distribution of antibodies in serum and nasal and vaginal secretions after nasal and oral vaccination of humans. Infect Immun. 1998, 66:3390–3396.

© 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