用户名: 密码: 验证码:
负载GST抗原树突状细胞疫苗抗日本血吸虫感染的保护效应研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
血吸虫病是一种严重危害人民健康,影响社会经济发展的重大传染病。主要分布于非洲、中东、南美和东南亚的76个国家和地区。在东南亚,目前血吸虫病主要流行于5个国家,即中国、菲律宾、印尼、柬埔寨和老挝。虽然经过几十年持续的防治,主要是大规模的人群化疗,各国流行区人群的感染率都比较低,但是流行的状况不稳定。我国为日本血吸虫病流行区。2008年全国调查资料显示,共有现症患者40余万,受威胁人口1亿。血吸虫病引起政府的高度重视,确定为重点防治的传染性疾病之一。
     我国血吸虫病防治工作面临的主要问题,包括重复感染严重存在、唯一有效化疗药吡喹酮的长期使用可能产生抗药株、大量哺乳动物保虫宿主的存在。因而寻找长效防治措施成为主要目标。疫苗预防作用被作为血吸虫病综合防治重要的措施之一而倍受关注。世界卫生组织(WHO)专家认为,化疗手段(短效)必须与长效的措施(疫苗)相结合。在疫苗研制的依据上,多年的探索表明,至少在啮齿类和灵长类的某些动物模型,获得性免疫应答可杀灭血吸虫。但是,尽管历经半个世纪的努力,血吸虫病疫苗目前仍然没有取得突破性进展,部分原因在于血吸虫在长期与宿主的共进化过程中,建立起复杂的保护体系,抵御和防御宿主免疫系统的攻击。在日本血吸虫病疫苗领域,近年来最大的进展在于克隆了数以百计的候选抗原,但单一候选抗原分子诱导的保护力鲜有超过40%。而世界卫生组织曾经公布的被认为最有希望的6种曼氏血吸虫病的候选抗原已全部完成测试,无一能稳定诱导40%以上的保护力。WHO认为可能是抗原供应前后制剂稳定性差,以及抗原和佐剂质控不严所致。多年的研究提示抗原递呈作用的重要性,佐剂的选择和免疫接种方案优化也需进一步探讨。
     树突状细胞是机体免疫系统岗哨工作的承担者,对自身或来自外界的抗原进行识别和加工。它们不仅识别加工抗原,还能活化初始T细胞,并且调节T细胞应答的性质,即诱导免疫耐受或是使免疫向Thl/Th2方向极化。DC这些重要的生物功能使通过对DC干涉调节免疫应答成为可能。目前已知,佐剂可通过激活DC而增强抗原应答,DC本身也被称为天然佐剂。经抗原体外刺激的DC免疫机体后可有效地诱导T细胞依赖的免疫力,还可诱导小鼠产生强的抗体应答。目前,基于DC的治疗和免疫预防广泛用于癌症和感染性疾病,取得显著进展。在血吸虫候选抗原中,谷胱甘肽S转移酶GST被认为是最有前途的候选抗原。有鉴于此,本文尝试用GST负载的树突状细胞联合CpG ODN免疫小鼠,研究其诱导的体液免疫和细胞免疫,并评价其免疫保护力。研究共分三部分:
     一日本血吸虫抗原负载树突状细胞的实验研究
     含日本血吸虫大陆株26kDa GST抗原基因的工程菌(Sj26GST/PBV220/DH5α)温控表达后,超声粉碎,应用亲和层析法纯化出重组GST抗原(简称GST),并经SDS-PAGE和Western blotting鉴定。可溶性虫卵抗原按常规制备。共采用两种树突状细胞进行血吸虫抗原负载的研究。小鼠骨髓来源树突状细胞(BMDC)制备培养方法参照文献,用IL-4和GM-CSF诱导培养。培养至第7天调整细胞浓度至2×105/ml接种于24孔培养板,每孔1ml。分别加入不同浓度的SEA、GST刺激细胞,并设LPS和PBS对照组。流式细胞仪检测树突状细胞表面分子CD40、 CD80、CD86的表达情况。结果表明,同本血吸虫抗原GST负载BMDC的最适浓度为1μg/ml.与LPS刺激不同,SEA、GST负载BMDC后,BMDC表面分子CD40、CD80、CD86表达并未上调,表现为不成熟表型。树突状细胞DC2.4按常规方法培养。分别加入不同浓度的GST进行抗原负载,比较树突状细胞表面分子CD40、CD80、CD86的表达水平。FITC标记的抗GST单克隆抗体检测GST抗原负载的情况。荧光显微镜下观察GST抗原负载的树突状细胞内可见特异的绿色荧光,表明抗原被细胞所摄取。流式细胞术分析结果表明,抗原刺激DC2.4的最适浓度为10μg/ml。日本血吸虫抗原GST、SEA负载DC2.4后,可检测到DC2.4表面CD40、CD86分子的平均荧光强度增强,而CD80增强不明显。
     二负载GST抗原的树突状细胞疫苗联合CpG-ODN免疫小鼠对日本血吸虫感染的保护性效应研究
     比较两种不同CpG ODN序列对树突状细胞DC2.4的刺激效果。流式分析结果表明,CpG ODN最佳刺激浓度为0.5μM, CpG0ODN1序列促DC成熟效果要优于CpG0DN2,且能进一步促进GST负载的树突状细胞的成熟。动物实验共分7组,每组小鼠10只,分别为:DC免疫组、BSA刺激DC免疫组、GST刺激DC免疫组、GST+CpG ODN刺激DC免疫组、CpG ODN刺激DC免疫组、GST蛋白免疫组、不免疫对照组(以下简称DC、BSA-DC、GST-DC、GST+CpG ODN-DC、CpG ODN-DC、GSTpr组)。1-5组每鼠皮下注射1×10。个细胞,共免疫三次,每次间隔两周。第6组第一次每鼠免疫50μg蛋白加完全弗氏佐剂,第二次免疫50μg蛋白加不完全弗氏佐剂,第三次免疫10μg蛋白加不完全弗氏佐剂。末次免疫两周后,每组半数小鼠留作细胞免疫相关检测分析。其余小鼠经皮肤感染30条尾蚴。每次免疫前及攻击感染前采血。攻击感染6周后门静脉灌注法收集成虫,计算减虫率。收集肝脏计数虫卵,计算每克肝组织的虫卵数(EPG)和减卵率。结果:与对照组比较,DC、BSA-DC、GST-DC、 GST+CpG ODN-DC、CpG ODN-DC和GSTpr组小鼠的减虫率分别为9.33%、2.7%、21.33%、53.33%、0和24%,减卵率分别为10.76%、19.37%、39.05%、64.24%、33.64%和34.90%,GST+CpG ODN-DC组小鼠有显著的减虫和减卵效果(P<0.05)。
     三GST抗原负载树突状细胞疫苗联合CpG ODN对日本血吸虫感染的免疫保护机制研究
     ELISA方法检测免疫血清特异性抗体及其亚类。结果表明,三次免疫后,GST蛋白免疫组产生的抗体滴度最高,并明显高于其他各组。GST-DC组和GST+CpG ODN-DC细胞免疫组的抗体滴度也明显提高,加强免疫后抗体亚类IgGl、IgG2a水平呈现上升趋势。末次免疫后每组小鼠各取5只,分别取小鼠脾脏和淋巴结,制备单细胞悬液。CCK-8法检测淋巴细胞增殖情况。结果表明,抗原刺激后,GST-DC组、GST+CpG ODN-DC组有较明显的增殖。淋巴细胞分离液分离脾脏淋巴细胞,同时以GST抗原(15μg/m1)体外刺激细胞,收集48h、72h和96h培养上清,ELISA法检测诱生的IL-4、IFN-γ和IL-10细胞因子水平。结果表明,与对照组相比,各组IL-4、IL-10水平并未明显提高,组间无明显差异;而相比对照组,GST+CpG ODN-DC组和GST-DC组IFN-γ水平均显著提高,与对照组相比上升了10倍(P<0.05)。上述结果表明,GST+CpG ODN-DC和GST-DC疫苗免疫C57BL/6小鼠后,诱导了体液免疫和细胞免疫应答,以Thl型细胞免疫为主导。
Schistosomiasis remains to be one of the major parasitic diseases of mankind, particularly in sub-Saharan Africa, Middle East, South America and Southeast Asia. And Schistosoma japonicum, one of the species of human schistosomiasis, is still endemic in China, Philippines, Indonesia, Cambodia and Laos. Owing to the availability of effective chemotherapeutic agents, large-scale population chemotheary, the prevalence in the endemic area is low. But epidemic status is unsteady. China is epidemic for Schistosoma japonicum, with400,000cases of schistosomiasis japonica and100million population at risk according to national survey in2008. And now it is regarded as one of the severe infectious diseases that should be focus on diseases control by China government.
     Schistosomiasis control in China has obstacles and confronted problems such as the reinfection in particular; the potential drug resistance in repeated chemotherapy, with the only effective drug praziquantel; a wide range of mammalian reservoir host. To develop of a long-term effective control measure has become a major task, of which vaccine development brought widely attention. Experts of WHO suggest that chemotherapy (short-term) must be combined with vaccine (long-term) in schistosomiasis control measures. In the recent years, efforts have accumulated a plenty of evidences in some animal models, including rodent and primate, that schistosome can be defeated by acquired immune response. But a recombinant antigen vaccine against schistosome remains elusive, in part because the parasite deploys complex defensive and offensive strategies to combat immune attack. The most significant progress made in recent years in vaccine against Schistosoma japonicum is gene cloning encoding vaccine candidates. Hundreds of antigens were cloned and expressed and their protective effects were tested in animal but only few induced protection over40%. The6candidate antigens regarded as the most promising ones by WHO have been retested, and unfortunately no one induced protection over40%steadily. The probable reasons may be the inconsistency of antigen preparation and quality control of antigen and adjuvant. A lot of research progress highlights to be recognized the importance of antigen presenting, adjuvance choosing and optimization of immunization routes.
     Dendritic cells (DC) are the crucial sentinel cells of the adaptive immune system, continuously sampling the antigenic environment. They not only process and present antigens to activate naive T lymphocytes but also regulate the nature of the T cell response obtained, determining whether it leads to tolerance or to Th1or Th2effector T cell responses. These central roles make manipulation of the DC system an attractive strategy for modulating immune responses. It is now clear that one of the roles of adjuvants which enhance response to antigens is to activate the DC system, while DC themselves have been termed "Nature's Adjuvants". Injection of in vitro pulsed DC may provide an efficient way for inducing T-dependent immunity without the use of external adjuvant and induce strong specific humoral responses in vivo. DC has been used in a very direct way in immunotherapy or prevention of cancer and infectious diseases and has made great success. Of all the vaccine candidate antigens for schistosomiasis, GST is regarded as the most promising one. On that account, we try to use GST pulsed DC to vaccinate mice and evaluate their humoral and cell-mediated immune response. The study included following three parts.
     1. Loading antigen of Schistosoma japonicum to dendritic cell
     The recombinant plasmid Sj26GST-PBV220containing GST gene was expressed in E.coli DH5a by temperature control. And then26kDa GST was purified by affinity chromatography and ananlyzed by SDS-PAGE and Western blotting. Soluble egg antigen was prepared as routine. We used two kinds of dendritic cells, bone marrow derived dendritic cell and DC2.4, to study the cell phenotype after antigen loaing. Bone marrow derived dendritic cells of mice were induced and cultured in the presence of GM-CSF and IL-4, as was described in reference. After culture for7days, cells were used to seed a flat-bottomed24-well culture microtitre plate, with2×105cells per well dispensed into1ml. Antigens, both GST and SEA were added in different concentration to load BMDC. LPS or PBS were also used to pulse BMDC and served as controls. The expression level of CD40, CD80, CD86were examined and analyzed by fluorescence-activated cell sorter (FACS). Results showed that the optimal concentration for antigen loading to BMDC was1μg/ml. The expression level of CD40, CD80, CD86were not raised in the surface of BMDC loaded with SEA or GST, which was significantly different from that loaded with LPS. So the phenotype of BMDC loaded with SEA or GST was an immature one. Dendritic cell2.4, a cell line of bone marrow derived dendritic cell of mouse, was cultured as routine. Antigen of GST in different concentration was added and the cells were pulsed for16h and then analyzed by FACS. Anti-GST monoclonal antibody conjugated with FITC was used to analyze the antigen loading. Specific green fluorescence was observed in GST pulsed DC under a fluorescence microscope, which indicated a successful antigen loading. According to the results of FACS analysis, the optimal loading concentration for GST antigen was10μg/ml. After loading, the mean fluorescence intensity of CD40and CD86, but not CD80on the surface of DC2.4, were significantly enhanced.
     2. Research on protective efficacy induced by dendritic cells pulsed with GST against Schistosoma japonicum infection
     The difference of immunostimulatory activities between two kinds of CpG ODN was compared. According to the results of FACS analysis, CpG ODN1was superior to the other in that it induced more mature dendritic cell. Seventy mice were divided into seven groups, vaccinated with GST protein, DC2.4alone or DC2.4pulsed with BSA, GST, GST+CpG ODN, CpG ODN respectively. For the cells vaccinated groups, each mouse was immunized with106cells subcutaneously for three times, at2weeks interval. and then challenged with30±1cercaria two weeks post immunization. For the GST protein vaccinated group, each mouse was vaccinated subcutaneously for50μg GST emulsified with complete Freund's adjuvant and then with incomplete Freund's adjuvant, and10μg GST with incomplete Freund's adjuvant for the last immunization. Two week post-immunization, half of mice in each group were sacrificed for cell-mediated immune response analysis and the rest were challenged with30±1cercaria. Six weeks later, all mice were sacrificed and worms were recovered. The average of adult worm and eggs per gram (EPG) of liver were calculated. Sera were collected before immunization and2week after each immunization. The protective efficacy was evaluated with worm reduction rate and egg reduction rate of the liver. Results showed that worm and egg burden in test groups were significantly lower than unvaccinated control group. The worm reduction rates of groups of DC, BSA-DC, GST-DC, GST+CpG ODN-DC, CpG ODN-DC and GST protein, were9.33%,2.67%,21.33%,53.33%,0,24%, respectively. And the EPG reduction rates were10.76%、19.37%、39.05%、64.24%、33.64%and34.90%respectively. GST+CpG ODN pulsed DC vaccine induced significant worm reduction rate and EPG reduction rate (P<0.05)
     3. The study on immune mechanism induced by GST+CpG ODN pulsed DC vaccine
     Titres of antigen-specific total IgG,1gG1,1gG2a,1gG2b antibodies in serum samples obtained from vaccinated mice were determined by a GST-based ELISA. Results showed that the antibody level was the highest in the group vaccinated with GST protein. Of the five groups vaccinated with DC, both GST-DC group and GST+CpG ODN-DC group exhibited higher antibody level, comparing to the unvaccinated control groups. And levels of1gG1and lgG2a were both raised as the times of vaccination increased. Two weeks after the last immnuzation, mice were sacrified and spleen and mesenteric lymph nodes were harvested under sterile conditions. These spleens and lymph nodes were pressed through a nylon mesh respectively to remove tissue debris and generate a single-cell suspension. Lymphocyte proliferation assay was performed by the method of CCK8. Results showed that lymphocytes from GST-DC group and GST+CpG ODN group challenged with antigen in vitro proliferated significantly. Spleen lymphocytes were separated using lymphocyte separating reagents. Cytokines released from spleen lymphocyte stimulated in vitro with15μg of GST per ml were determined in the culture supernatants collected at48h,72h and96h. The concentrations of IL-4, IL-10and IFN-y were determined by ELISA. Splenocytes from mice vaccinated with GST-DC and GST+CpG ODN-DC secreted larger amounts of IFN-y than that in other groups (P<0.05). And IL-4and IL-10level among varied groups were not significantly different. These results suggest a potential Th1/Th2immune response but Th1dominating.
引文
1. Steinman RM, Cohn ZA. Identification of a novel cell type in peripheral lymphoid organs of mice. Ⅰ. Morphology, quantitation, tissue distribution. J Exp Med,1973,137(5):1142-1162.
    2. Caux C, Masacri C, Vanbervlier B, et al. CD34+hematopoietic progenitors from human cord blood differentiate along two independent dendritic cell pathways in response to granulocyte-macrophage-colony-stimulating factor plus tumor necrosis factor:Ⅱ functional analysis[J]. J Blood,1997,90(4):1458-1470.
    3. Banchereau J, Briere F, Caux C, et al. Immunobiology of dendritic cells[J]. Annu Rev Immunol,2000,18:767-811.
    4. Traver D, Akashi K, Manz M, et al. Development of CDα~positive dendritic cells from a common myeloid progenitor[J]. Science,2000,290(5499):2152-2154.
    5. Manz MG, Traver D, Miyamoto T, et al. Dendritic cell potentials of early lymphoid and myeloid progenitors [J]. Blood,2001,97(11):3333-3341.
    6. del Hoyo GM, Martin P, Vargas HH, et al. Characterization of a common precursor population for dendritic cells[J]. Nature,2002,415(6875):1043-1047.
    7.陈慰峰.医学免疫学[M].北京:人民卫生出版社,2001.136-137.
    8. Sallusto F, Cella M, Danieli C, et al. Dendritic cells use macropinocytosis and the mannose receptor to concentrate macromole-cules in the major histocompatibility complex class Ⅱ compartment:downregulation by cytokines and bacterial products[J]. J Exp Med,1995,182: 389-400.
    9. Winzler C, Rovere P, Rescigno M, et al. Maturation stages of mouse dendritic cells in growth factor-dependent long-term cultures[J]. J Exp Med,1997,185:317-328.
    10. Cella M, Engering A, Pinet V, et al. Inflammatory stimuli induce accumulation of MHC class II complexes on dendritic cells [J]. Nature,1997,388:782-787.
    11.林慰慈,薛彬,魏雪涛.免疫学[M].北京:科学出版社,2001.20.
    12. Lutz MB, Schuler G. Immature, semi-mature and fully mature dendritic cells:which signals induce tolerance or immunity [J].Trends Immunol,2002,23:445-449.
    13. Manickasingham SP, Edwards AD, Schulz O, et al. The ability of murine dendritic cell subsets to direct T helper cell differentiation is dependent on microbial signals[J]. Eur J Immunol,2003,33:101-107.
    14. Kapsenberg ML. Dendriticcell control of pathogen driven T cell pol arization[J]. Nat Rev Immunol,2003,3:984-993.
    15. Perona-wright G, Jenkins SJ, MacDonald AS. Dendritic cell activation and function in response to Schistosoma mansoni [J]. Int J Parasitol,2006,36:711-721.
    16. van Riet E, Everts B, Retra K. Combined TLR2 and TLR4 ligation in the context of bacterial or helminth extracts in human monocyte derived dendritic cells:molecular correlates for Thl/Th2 polarization., BMC Immunol,2009,9:10
    17. Liu SX, Song GC, Xu YX, et al. Immunization of mice w ith recombinant Sjc26GST induces a pronounced antifecundity effect after experimental infection with Chinese Schistosoma ja onicum. Vaccine,1995; 13(6):603-607
    18. Son Y, Egawa S, Tatsumi T, et al. A novel bulk-culture method for generating mature dendritic cells from mouse bone marrow cells. J Immunol Methods,2002,262(1-2):145-157
    19. MacDonald AS, Straw AD, Bauman B. CD8-dendritic cell activation status plays an integral role in influencing Th2 response development[J]. J Immunol,2001,167(4):1982-1988.
    20. Jankovic D, Kullberg MC, Caspar P, et al. Parasite-induced Th2 polarization is associated with down-regulated dendritic cell responsiveness to Thl stimuli and a transient delay in T lymphocyte cycling[J]. J Immunol,2004,173(4):2419-2427.
    21. Zaccone P, Fehervari Z, Jones FM, et al. Schistosoma mansoni antigens modulate the activity of the innate immune response and prevent onset of type 1 diabete. Eur J Immunol,2003, 33(5):1439.
    22. Faveeuw C, Angeli V, Fontaine J, et al. Antigen presentation by CD1d contributes to the amplification of Th2 responses to schistosoma mansoni glycoconjugates in mice[J]. J Immunol,2002,169(2):906-912.
    23. Balic A, Harcus Y, Holland MJ, et al. Selective maturation of dendritic cells by Nippostrongylus brasiliensis-secreted proteins drives Th2 immune responses. Eur J Immunol, 2004,34(11):3047-3059.
    24. Shen, Z., G. Reznikoff, G. Dranoff, et al. Cloned dendritic cells can present exogenous antigens on both MHC class Ⅰ and class Ⅱ molecules. J. Immunol,1997,158:2723-2730.
    25.罗建飞,陈必成,陈忠华.小鼠骨髓来源树突状细胞和DC2.4形态与功能比较.中国康复,2005,20(3):145-146
    26.郑磊,包杰,王前,等.小鼠骨髓源性树突状细胞和DC2.4中Re1B基因表达水平的比较研究.热带医学杂志,2006,6(7):745-748
    27. Aline F, Bout D, Amigorena S, et al. Toxoplasma gondii antigen-pulsed-dendritic cell-derived exosomes induce a protective immune response against T. gondii infection. Infect Immun. 2004 Jul;72(7):4127-37.
    28. Cheadle EJ, Donnell DO, Selby PJ, et al. Closely Related mycobacterial strains demonstrate contrasting levels of efficacy as antitumor vaccines and are processed for major histocompatibility complex class Ⅰ presentation by multiple routes in dendritic cells. Folia Biologica(Praha) 2004,50:184-193
    29. Mendoza L, Bubenik J, Simova J. Prophylactic, therapeutic and anti-metastatic effects of BMDC and DC lines in mice carrying HPV 16-associated tumours. Int J Oncol,2003, 23(1):243-247
    30. Indrova M, Reinis M, Bubenik J, et al. Immunogenicity of Dendritic Cell-Based HPV 16 E6/E7 Peptide Vaccines:CTL Activation and Protective Effects. Folia Biologica (Praha), 2004,50,184-193.
    31. Rhulea A, Raseb B, Smith JR, et al. Toll-like receptor ligand-induced activation of murine DC2.4 cells is attenuated by Panax notoginseng. J Ethnopharmacol.2008,116(1):179-186.
    32. Fang F, Wang Y, Li R, et al. Transcription Factor E2F1 Suppresses Dendritic Cell Maturation. J Immunol.2010
    1. Wilson RA, Coulson PS. Immune effector mechanisms against schistosomiasis:looking for a chink in the parasite's armour.Trend in Parasitology,2008,25(9):423-431.
    2. Coulson PS. The radiation-atteduated vaccine against schistosomes in animal models: paradigm for a human vaccine? Adv Parasitol,1997,39:271-336
    3. Kariuki TM. Parameters of the atteduated schistosome vaccine evaluated in the olive baboon. Infect Immun,2004,72:5526-5529
    4.李小红,曹建平,刘述先.我国血吸虫病疫苗候选抗原研究新进展.中国人兽共患病杂志,2005,21(10):901-905
    5.刘述先,曹建平.寄生虫病疫苗研究的现状及展望。中国寄生虫学与寄生虫病杂志,2005,23(5):362-368
    6. Tokunaga T; Yamamoto H; Shimada S; et al. Antitumor activity of deoxyribonucleic acid fraction from Mycobacterium bovis BCG. I. Isolation, physicochemical characterization, and antitumor activity. Journal of the National Cancer Institute,1984,72 (4):955-62.
    7. Weiner GJ. CpG DNA in cancer immunotherapy. Curr Top Microbiol Immunol,2000, 247:157-170
    8. Krieg AM, Yi AK, Matson S, et al CpG motifs in bacterial DNA trigger direct B-cell activation. Nature,1995,374:546-549
    9. Bauer M,Heeg K, Wagner H, et al. DNA activates human immune cells through a CpG sequence-dependent manner. Immunology,1999,97(4):699-705
    10. Kline JN. Effect of CpG DNA on Thl/Th2 balance in asthma. Curr Top Microbiol Immunol, 2000,247:211-225
    11. Latz E, Schoenemeyer A, Visintin A, et al. TLR9 signals after translocating from the ER to CpGDNA in the lysosome [J]. Nat Immunol,2004,5 (2):190-198
    12. Ashkar AA, Bauer S, Mitchell WJ, et al. Local delivery of CpG oligodeoxynucleotides induces rapid changes in the genital mucosa and inhibits replication,but not entry,of herpes simplex virus type 2.J Virol.2003 Aug;77(16):8948-56.
    13. Becker Y.A point of view:HIV-1/AIEDS is an allergy but CpG ODN treatments may inhibit virus replication and reactivate the adaptive immunity---hypothesis and implications.Virus Genes.2005;30(1):127-31
    14. Isogawa M,Robek MD,Furuichi Y.Chisari FV.Toll-like receptor signaling inhibits hepatitis B virus replication in vivo.J Virol.2005 Jun;79(11):7269-72.
    15. Ito S,Ishii KJ,Jhata A,Klinman DM.Contribution of nitric oxide to CpG-mediated protection against Listeria monocytogenes.Infect Immun.2005a Jun;73(6):3803-5.
    16. Bourguin I, Moser M, Buzoni-Gatel D, et al. Murine dendritic cells pulsed in vitro with Toxoplasma gondii antigens induce protective immunity in vivo [J]. Infect Immun, 1998,66(10):4867-4874.
    17. Dimier-Poisson I, Aline F, Mevelec MN, et al. Protective mucosal Th2 immune response against Toxoplasma gondii bymurine mesenteric lymph node dendritic cells[J]. Infect Immun, 2003,71(9):5254-5265.
    18. Ahuja SS, Ressick RL, Sato N, et al. Dendritic cell-based anti-infective strategies:DCs engineered to secrete IL-12 are a potent vaccine in a murine model of an intracellular infection[J]. J Immunol,1999,163(7):3890
    19. Ghosh M, Pal C, Ray M, et al. Dendritic cell-based immunotherapy combined with antimony-based chemotherapy cures established murine visceral leishmaniasis[J]. J Immunol, 2003,170(11):5625
    20. Bruna-Romero O, Rodriguez A. Dendritic cells can initiate protective immune responses against malaria[J]. Infect Immun,2001,69(8):5173
    21. Liu SX, Song GC, Ding LY, et al. Comparative study on antigenicity and immunogenicity of 26-28kDa antigen and recombinant Sj26(rSj26) of'Schistosoma japonicum[J]. Southeast Asian J Trop Med Public Health,1993,24:65-69.
    22. Liu SX. An overview of molecular parasitology in China[J]. Chin Med J,1992,10:591-596
    23.刘述先,宋光承,丁丽韵,等.日本血吸虫中国大陆株GST抗原[J].中国寄生虫学与寄生虫病杂志,1992,10:312-13.
    24. Liu SX, Ding LY, Song GC, et al. The antigenicity of GST antigen extracted from Chinese strain of Schistosoma japonicum [J]. Southeast Asian J Trop Md Public Health,1993, 24:61-64.
    25. Liu SX, Song GC, Xu YX, et al. Immunization of mice with recombinant Sjc26 GST induces a pronounced anti-fecundity effect after experimental infection with Chinese Schistosoma japonicum[J]. Vaccine,1995,13:603-606
    26. Liu SX, Song GC, Xu YX, et al. Anti-fecundity immunity induced in pigs vaccinated with recombinant Schistosoma japonicum 26kDa glutathione-S-transferase [J]. Parasite Immunol, 1995,17:335-340.
    27. Liu SX, He YK, Song GC, Luo XS, Xu YX, McManus DP. Anti-fecundity immunity to Schistosoma japonicum induced in Chinese water buffaloes (Bos buffelus) after vaccination with recombinant 26 kDa glutathione-S-transferase (reSjc26GST) [J]. Vet Parasitol,1997, 69:39-47.
    28. Wu Z, Liu S, Zhang S, et al. Persistence of the protective immunity to Schistosoma japonicum in Chinese yellow cattle induced by recombinant 26kDa glutathione-S-transferase (reSjc26GST) [J]. Vet Parasitol,2004,123:167-177.
    29.蒋健敏,屠乐鸣,张素娥,等.日本血吸虫26kDa基因重组蛋白免疫小鼠抗体内血吸虫升值的作用[J].中国寄生虫学与寄生虫病杂志,1999,17:9-11.
    30.沈定文,罗金萍,李雍龙.Sj26基因转染DC抗血吸虫感染作用[J].中国公共卫生,2005,21(11):1393-1394
    31.罗金萍,陈喜圭,沈定文,等.树突状细胞抗日本血吸虫感染保护性免疫力的研究[J].咸宁学院学报(医学版),2004,18(2):91-94
    32.沈定文,罗金萍,李雍龙.基因转染树突状细胞联合免疫抗血吸虫感染作用[J].中国公共卫生,2008,24(10):1216-1217.
    33. Leonard JP, Link BK, Emmanouilides C, et al. Phase I trial of toll2 Like recep tor 9 agonist PF23512676 with and following rituximab in patientswith recurrent indolent and aggressive non hodgkin's lympho2 ma[J]. Clin Cancer Res,2007,13 (20):6168-6174.
    34. Cooper CL, Davis HL, MorrisML, et al. CPG 7909, an immunostimulatory TLR9 agonist oligodeoxynucleotide, as adjuvant to Engerix2BHBV vaccine in healthy adults:a double 2 blind phase Ⅰ/Ⅱ study[J]. J Clin Immunol,2004,24 (6):693-701.
    35. McHutchison JG, Bacon BR, Gordon SC, et al. Phase 1B, random- ized, double-blind, dose-escalation trial of CPG 10101 in patientswith chronic hepatitis C virus[J]. Hepatology, 2007,46 (5):1341-1349.
    36. Sparwasser T, Koch ES, Vabulas RM, et al. Bacterial DNA and immunostimulatory CpG oligonucleotides trigger maturation and activation of murine dendritic cells[J]. Eur J Immunol,1998,28 (6):2045-2054.
    37. Lipford GB, Bauer M, Blank C, et al. CpG containing synthetic digonucleotides promote B and cytotic T cell responses to protein antigen:a new class of vaccine adjuvants[J]. Eur J Immunol,1997,27(9):2340-2344.
    38. Ballas ZK, Rasmussen WL, Krieg AM. Induction of NK activity in murine and human cells by CpG motifs in oligodeoxynucleotides and bacterial DNA[J]. J Immunol,1996,157(5): 1840-1845.
    39. NiH T, Spellman SR, JeanW Cetal. Immunizationwith dendritic cells pulsed with tumor extract increases survival ofmice bearing in-tracranial gliomas [J]. J Neuro oncol,2001; 51: 1-9.
    40. InoueN, YamasakiS, KondoKetal. Dendritic cells coinjectedwith tumor cells treated with an anticancer drug to induce tumor rejection [J]. Surg Today,2003; 33:269-276.
    41. SmithA L, FazekasDe StGroth B. Antigen-pulsed CD8 alpha+ den-dritic cells generate an immune response after subcutaneous injection withouthoming to the draining lymph node [J]. J ExpMed,1999; 189:593-598.
    42. KonoK, TakahashiA, SugaiHet al. Dendritic cells pulsed with HER-2/neu-derived peptides can induce specificT-cell responses in patientswith gastric cancer [J]. Clin Cancer Res,2002; 8:3394-3400.
    43. PorgadorA, GilboaE. Bonemarrow-generated dendritic cellspulsed with a class Ⅰ-restricted peptide are potent inducers of cytotoxic T lymphocytes [J]. J Exp Med,1995; 182:255-260.
    44. Mayordomo J I, Zorina T, StorkusW Jet al. Bonemarrow-derived dendritic cells pulsedwith synthetic tumourpeptides elicitprotective and therapeutic antitumour immunity [J]. Nat Med, 1995; 1:1297-1302.
    45. Zhang S, ZengG, W ilkesD Setal. Dendritic cells transfectedwith interleukin-12 and pulsedwith tumorextract inhibitgrowth ofmurine prostatic carcinoma in vivo [J]. Prostate, 2003; 55:292-298.
    46. Wang TL, Ling M, Shi IM, etal. Intramuscular administration of E7-transfected dendritic cells generates the most potent E7-specific anti-tumor immunity [J]. Gene Ther,2000; 7:726-733.
    47. Saika T, Satoh T, Kusaka Net al. Route of administration influ-ences the antitumor effects of bone marrow-derived dendritic cell engineered to produce interleukin-12 in ametastaticmouse prostate cancermodel [J]. Cancer Gene Ther,2004; 11:317-324.
    48. Eggert AA, Schreurs MW, BoermanO Cetal. Biodistribution and vaccine efficiency of murine dendritic cells are dependent on the route of administration [J]. Cancer Res,1999; 59: 3340-3345.
    49. Okada N, TsujinoM, Hagiwara Yet al. Administration route-dependent vaccine efficiency ofmurine dendritic cellspulsedwith antigens [J]. Br JCancer,2001; 84:1564-1570.
    50. FongL, BrockstedtD, Benike Cet al. Dendritic cells injected via different routes induce immunity in cancerpatients [J]. J Immuno,2001; 166:4254-4259
    51.谭晓华,刘端祺.不同免疫途径对抗原肽修饰DC疫苗免疫效应的影响,中国免疫学杂志,2006(22):1036-1040
    52. Kuribayashi, K., M. Tsukiyama, and T. Takenaka. Secretion patterns of Thl-and Th2-type cytokines in immune deviation caused by dendritic cells. Int. Arch. Allergy Immunol,1997, 114:30
    53. Dudda JC, Simon JC, Martin S. Dendritic cell immunization route determinesCD8+T cell trafficking to inflamed skin:role for tissue microenvironment and dendritic cells in establishmentof T cell-homing subsets [J]. J Immunol,2004,172:857-863
    54. Maayordomo JI, Zorina T, Storkus WJ, et al. Bone Marrow-Derived Dendritic Cells Serve as Potent Adjuvants for Peptide-Based Antitumor Vaccines. Stem Cells,1997,15:94-103
    55. Aichele P, Brduscha-Riem K, Zinkernagel RM et al. T cell priming versus T cell tolerance induced by synthetic peptides. J Exp Med 1995,182:261-266
    1. Wilson RA, Coulson PS. Immune effector mechanisms against schistosomiasis:looking for a chink in the parasite's armour.Trend in Parasitology,2008,25(9):423-431.
    2. Sher A, Hieny S, James SL, et al. Mechanisms of protective immunity against Schistosoma mansoni infection in mice vaccinated with irradiated cercariae. Ⅱ. Analysis of immunity in hosts deficient in T lymphocytes, B lymphocytes, or complement. J Immunol,1982; 128 (4):1880-1884
    3. Dillon, G.P. et al. (2008) Altered patterns of gene expression underlying the enhanced immunogenicity of radiation-attenuated schistosomes. PLoS Negl Trop Dis 2, e240
    4. Kelly EA, Colley DG. In vivo effects of monoclonal anti-L3T4 antibody on immune responsiveness of mice infected with Schistosoma mansoni. Reduction of irradiated cercariae-induced resistance. J Immunol,1988,140(8):2737-2745
    5. Smythies LE, Coulson PS, Wilson RA. Monoclonal antibody to IFN-gamma modifies pulmonary inflammatory responses and abrogates immunity to Schistosoma mansoni in mice vaccinated with attenuated cercariae. J Immunol,1992; 148 (5):1512-1518
    6. Osada Y, Janecharut T, Hata H, et al. Protective immunity to Schistosoma japonicum infection depends on the balance of T helper cytokine responses in mice vaccinated with gamma-irradiated cercariae. Parasite Immunol,2001,23(5):251-258
    7. Street, M. et al. TNF is essential for the cell-mediated protective immunity induced by the radiation-attenuated schistosome vaccine. J Immunol,1999,163,4489-4494
    8. McLaren DJ, Smithers SR. Serum from CBA/Ca mice vaccinated with irradiated cercariae of Schistosoma mansoni protects naive recipients through the recruitment of cutaneous effector cells. Parasitology,1988,97(Suppl):287-302
    9. Bickle QD, Andrews BJ, Doenhoff MJ, et al. Resistance against Schistosoma mansoni induced by highly irradiated infections:studies on species specificity of immunization and attempts to transfer resistance. Parasitology,1985,90(Suppl):301-312
    10. Jankovic D, Wynn TA, Kullberg MC, et al. Optimal vaccination against Schistosoma mansoni requires the induction of both B cell- and IFN-gamma dependent effector mechanisms. J Immunol,1999,162,345-351
    11. Kariuki, T.M. et al. Parameters of the attenuated schistosome vaccine evaluated in the olive baboon. Infect Immun,2004,72,5526-5529
    12. Cutts, L. and Wilson, R.A. Elimination of a primary schistosome infection from rats coincides with elevated IgE titres and mast cell degranulation. Parasite Immunol,1997,19,91-102
    13. Miller, H.R. et al. Hepatic recruitment of mast cells occurs in rats but not mice infected with Schistosoma mansoni. Parasite Immunol,1994,16,145-155
    14. Wilson, R.A. et al. Elimination of Schistosoma mansoni adult worms by rhesus macaques: basis for a therapeutic vaccine? PLoS Negl Trop Dis,2008,2, e290
    15. MosmannTR, Cherwinski H, MW B, et al. Two types of murine helper T cell clone. Ⅰ.Definition according to profiles of lymphokine activities and secreted proteins. J Immunol, 1986,136:2348-2357
    16.沈定文,罗金萍,李雍龙,,等.Sj26基因转染的树突状细胞对日本血吸虫感染的免疫保护机制研究.中国寄生虫学与寄生虫病杂志,2007,25(1):17-21
    17.曾庆仁,林雪迟,龚燕飞,等.一种新型日本血吸虫疫苗”童虫活细胞诱导小鼠产生有效保护性免疫力的研究.中国现代医学杂志,2004,14(14):84-87
    18. Smythies LE, Coulson PS, Wilson RA. Monoclonal antibody to IFN-gamma modifies pulmonary inflammatory responses and abrogates immunity to Schistosoma mansoni in mice vaccinated with attenuated cercariae. J. Immunol,1992,149:3654-3658.

© 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