用户名: 密码: 验证码:
Toll样受体3、9在皮肤中的作用机制和信号转导通路研究及药物的干预
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的:研究TLR3、TLR9在皮肤天然免疫系统中的作用及调控机制,深入了解皮肤在天然免疫和获得性免疫中的桥梁作用,为预防和治疗皮肤免疫性疾病提供新的作用靶点。
     方法:通过体外培养人角质形成细胞株HaCaT细胞和真皮成纤维细胞,应用免疫组化、real-time PCR、流式细胞术和Western blotting方法考察TLR3、TLR9在两种细胞上的调控特征,探讨TLRs在天然免疫和获得性免疫中的桥梁作用。在此基础上,研究地塞米松、霉酚酸和他克莫司对TLR3、TLR9及其信号转导通路的干预作用。探讨CpG-ODN的乙肝质粒DNA柔性脂质体经皮肤以TLR9为靶标进行免疫的可行性,并从离体角度研究经皮免疫的机制,证明TLR9及其信号通路在经皮免疫接种中的作用机制。
     结果:确定了TLR3、TLR9在HaCaT和真皮成纤维细胞有表达,且可特异性的识别其相应的配体poly(I:C)和ODN2216,其信号转导通路是MyD88依赖性的,通过调控MyD88信号通路,可激活获得性免疫,使得Th1和Th2型细胞因子的分泌均有不同程度的增加,但是以诱导Th1型为主。Th1型细胞因子也可对TLR3、TLR9及其信号转导通路进行调控,形成天然免疫-获得性免疫调控网络。EGF可显著增强HaCaT细胞TLR3、TLR9的表达,调控其信号转导通路。免疫抑制剂霉酚酸和地塞米松对正常的HaCaT细胞的TLR3、TLR9受体及其信号通路无显著性影响,但对配体激活状态下HaCaT细胞表现出免疫抑制作用。他克莫司并未显示出对TLR3、TLR9及其信号转导通路的影响作用。构建了新的可表达CpG-ODN的乙肝质粒DNA柔性脂质体,证明了其经皮肤以TLR9为靶标进行免疫的可行性,并从离体角度研究经皮免疫的机制,研究了TLR9及其信号通路的作用机制,有助于更好的产生抗体和细胞免疫反应。
     结论:研究了TLR3、TLR9在皮肤天然免疫系统中的作用及调控机制,探讨了免疫抑制剂和新型疫苗对该靶点作用的可行性,为该领域新的TLRs激动剂和拮抗剂的研制开发展现了广阔的前景,同时为经皮免疫提供了良好的技术支持和理论依据。
The immune system is complex defense system that evolving through ages. Itsbasic function is to recognize the foreign antigen and the upcoming signals of danger,and start the down-stream activation events to eliminate the foreign pathogen andmaintain the homeostasis in the body. The immune system of mammary animalsconsists of two parts—natural immune and acquired immune. Natural immune is alsocalled inherent immune, which is very old and possessed by all kinds of multi-cellularanimals and includes many kinds of different host defense mechanism. The inherentimmune has a quick action on the antigens, but with no memories. Like the skin, itacts as the first defense to our body. And the acquired immune only exists in vertebralanimals, which is an advanced defense system mediated by the antigen-specific Tcells and B cells. It has a slow action, but also with a memory to the antigens. Thenatural immune is the first step in an immune response. The pathogens are swallowedby the antigen presenting cells (APC). And then the antigens are degenerated andpresented in form of MHCⅡantigen-peptides to the T-cell to activate the immuneresponse. Meanwhile the immune response is also activated through the recognition ofpathogens and activation of the immune and inflammatory cells to eliminate theinvading microbes.
     Toll like receptors (TLRs) family is a new kind of mammary pattern recognitionreceptors (PRRs), which mediate the recognition of the pathogen-associatedmolecular pattern (PAMPs), and play an important role in the natural immune system.The TLR3 is involved in the recognition of virus dsRNA, and TLR9 mainly presentedin the surface of APC, with a specific recognition of the CpG gene sequence in thebacterial genome. The recognition of microbes, APC, activation of T-cell and cellsthat trigger the acquired immune response were all linked together by the TLRs. Andthe TLRs are activated by binding with their ligands and then transmit the signalsdown-stream through the Toll/IL-1R pathway, which finally activates transcription factors such as NF-κB、JNK、P38、ERK. Then the expression of MHCⅠand MHCⅡmolecules and their costimulating factors are up-regulated, the ability of APCs aresimulated, and various kinds of acute inflammatory factors such as IL-1、IL-6、IL-12and TNF-αare induced. So the interaction between TLRs and their ligands is animportant bridge in the process of natural and acquired immune responses.
     The skin is the first defense in the natural immune system, and plays animportant role in the prevention of foreign microbe infection. First, it acts as aphysical barrier to the pathogens. Second, the skin can secrete many kinds ofcytokines, chemotactic factors and antibiotic peptides to mediate the natural andacquired immune process, and facilitate the elimination of pathogens. But the specificunderlying mechanisms of these defense systems are still unclear. Virus dsRNA andbacterial CpG are the main pathogenic factors in the skin-related diseases, and theyare the exactly the ligands of TLR3、TLR9. So is there an expression of TLR3、TLR9in the skin under normal circumstances? If there's an expression of these factors,where do they come from, and how do they function? If there's no expression of thefactors by the skin, are they induced from foreign cells? And how do these factorsactivate the signaling pathway to trigger a immune response? There questions willelucidate the roles of TLR3、TLR9 in the skin natural immune system.
     This study is to observe the expression patterns of TLR3、TLR9 in two kinds ofcells—epidermis keratinocyte HaCaT and dermis fibroblast, by means ofimmunohistochemistry, real-time PCR, flowcytometry, and western blotting. After thedetermination of the existence of the TLR3、TLR9, we investigate the regulationeffects of TLR3、TLR9 ligands—poly(I:C), ODN2216 and LPS on the expression ofTLR3、TLR9, and discuss their influences to the signaling pathway. Meanwhile, aninvestigation of the time-dose relation in the secretion of Th1 and Th2 cytokines iscarried out to serve as a theoretical basis for the role of TLR3、TLR9 in the skinnatural immune system. Then discuss the regulation role of cytokines to the naturalimmune system. After that, we investigate the effect of immunosuppressants such asFK506, MPA and Dex on the expression of TLR3、TLR9 receptors and discover newtargets for the drugs, providing a theoretical basis for the development of new immunosuppressants. Finally, investigate the possibility of transdermal immune inboth in vivo and in vitro studies, after the construction of Hepatitis B plasmid DNAliposome with the expression of CpG-ODN and discuss the expression of HBsAG indifferent stages in the immune response process and elucidate the effect of vacantliposome and vaccine liposome on the skin natural immune system.
     The study consists of the following five parts:
     Part One: The expression of TLR3、TLR9 in human HaCaT and fibroblasts.
     1. Immunohistochemistry results showed that there are positive expressions of TLR3、TLR9 in the normal HaCaT cells and dermis fibroblasts.
     2. Real-time PCR showed that there are expressions of TLR3、TLR9 in normal HaCaTcells and dermis fibroblasts and the expressions can be significantly enhanced byLPS、poly(I:C)、ODN2216, and acts in a ligand specific fashion.
     3. FC results showed that there are significant positive expressions of TLR3、TLR9 innormal HaCaT cells and dermis fibroblasts (P<0.05). Various simulating factors allexerts influence over the expression of the TLR3 in HaCaT cells, with poly (I: C) cansignificantly upregulate the coexpression of TLR3 in the surface and inside of thecells. ODN2216 has no significant effect on the expression of TLR3 in two cell types,but can significantly upreguiate the coexpression of TLR9. poly (I:C) has nosignificant effect on the TLR9 expression in two cell types. And LPS can significantlyupregulate the coexpression of TLR3、TLR9 in both cell types.
     4. Western blotting results showed that there are expressions of TLR3、TLR9 in bothnormal HaCaT cells and dermis fibroblasts and they can be regulated by variousfactors. Poly (I: C) and LPS can significantly increase the expression of TLR3 in bothcells, while ODN2216 has no effect. And ODN2216 and LPS can significantlyincrease the expression of TLR9 in both cells, while poly (I: C) has no effect.
     All the studies showed that there are TLRs expressions in HaCaT cells and dermisfibroblasts which will play key role in the natural immune system. After binding withthe ligands, the same kind of biological characteristics as previous studies showed up,which is indicative an important role of both cells in the natural immune response.
     Part Two: The effect of activation of TLR3、TLR9 on the secretion of Th1/Th2cytokines in HaCaT cells and fibroblasts and the MyD88 signaling pathway.
     1. 1. Real-time PCR results showed that there are expressions of IFN-γ, IL-4 andMyD88 mRNA in normal HaCaT cells and dermis fibroblasts. And after poly(I:C),ODN2216 and LPS treatments with the HaCaT cells, the impacts on the connectinMyD88 are the same. And after treatments with FB cells, poly (I:C) has moresignificant effect on MyD88, while LPS has more significant effect on the secretion ofIFN-γand IL-4.
     2. 2. ELISA assays have elucidated the time-effect relation in the secretion of IFN-γand IL-4 in both cells after the treatment of poly(I:C), LPS and ODN2216. the resultsshowed that both cells can secret some IFN-γand IL-4 in normal status. And aftertreatment with poly(I:C), ODN2216 and LPS for 0、6、12、24、48h, the concentrationof IFN-γand IL-4 in the supernatant fluids are elevated, and there's a more significantincreasing trend for IFN-γ.
     3. The western blotting assays have semi-quantitatively detected the changes inthe expression of MyD88 after different ligands treatments in both cells. The resultsshowed expression in both cells and the expression could be upregulated by theTLR3、TLR9 ligands.
     The studies in this part confirmed that the interaction between ligands and TLR3、TLR9 are not only regulated by the expression of receptors but also through theactivation of acquired immune response and influence over the expression of MyD88which will regulate the secretion of cytokines, induce the TH1 immune reaction.Therefore, the expression of TLR3、TLR9 with specific drugs and both the secretionof cytokines and signaling pathway would be affected, which has important roles inthe skin immune system. These results also confirmed that TLR3、TLR9 play animportant role in the natural and acquired immune response. And we can make furtherresearches in an integrated fashion, and a new age of the immunological research isopened to us.
     Part Three: The effects of IFN-γ、TNF-αand EGF on the TLR3、TLR9 and theirsignaling pathway in HaCaT cells and FB cells
     1. Detected the expression level of TLR3、TLR9 and MyD88 mRNA by means ofreal-time PCR in HaCaT cells and FB cells after treatment with IFN-γ(50ng·mL~(-1))、TNF-α(100ng·mL~(-1))and EGF(10ng·mL~(-1))for 24h. The results showed an elevationin the TLR3、TLR9 and MyD88 mRNA levels in both two cells (EGF has. nosignificant effect on the expression of TLR3、TLR9 and the signaling pathway in FBcells), from 2~2-2~8 folds.
     2. Detected the amount of the receptors on the surface and inside the HaCaT cells andFB cells with flow cytometry after treatment with IFN-γ(50ng·mL.(-1))、TNF-α(100ng·mL~(-1))and EGF(10ng·mL~(-1))for 24h. the results showed an elevation in theexpression of TLR3、TLR9 after treatment with IFN-γ、TNF-αand EGF. And afterremoving the effects of membrane-expression, there's no significant differences foundin the expression level of receptors inside the cells for TLR3 and significant elevationfor TLRg. In normal FB cells, except the EGF, IFN-γ、TNF-αboth can increase thelevel of receptors. And after removing the effects of membrane-expression, there'ssignificant increase found in the expression level of both TLR3、TLR9 inside thecells.
     3. Detected the expression level of the TLR3、TLR9 with western blotting in HaCaTcells and FB cells after treatment with IFN-γ(50ng·mL~(-1))、TNF-α(100ng·mL~(-1))andEGF (10ng·mL~(-1))for 24h. The results showed that IFN-γ、TNF-αand EGF all cansignificantly increase the expression of MyD88 in normal HaCaT cells. And TNF-αcan significantly increase the expression of MyD88 in normal FB cells, while EGBhas no effect.
     Studies in this part IFN-γ, TNF-αand EGF can affect TLRs and their signalingpathway in different fashions. These results provide us a new drug target fortransdermal immune therapy and give us a new way to the development of newvaccines to treat and prevent the immunological and inflammatory diseases.
     Part Four : The regulation of immunosuppressants on the TLR3、TLR9 and theirsignaling pathway in HaCaT cells.
     1. Real-time PCR showed that given single dose of DEX、FK506 and MPA didn'taffect the expression of TLR3、TLR9 and MyD88. But in the presence of poly (I:C),the expression of TLR3 and MyD88 increased significantly. After co-culture withpoly (I:C), DEX can significantly increase the expression of TLR3 and MyD88, whileFK506 has no significant effect. The expression of TLR9 will be significantlyelevated after co-culture of DEX and poly (I: C). There's significant elevation in theexpression of TLR9 and MyD88 in the presence of only ODN2216, while there'ssignificant elevation in the induced expression of TLR9 and MyD88 after Co-cultureof ODN2216 and DEX. MPA can also increase the expression level of TLR9, whileFK506 has no obvious effect. MPA can increase the expression of MyD88 afterco-culture with poly(I:C)、ODN2216.
     2. FC results showed that given single dose of DEX、FK506 and MPA can notchange the expression pattern of TLR3、TLR9 on the surface and inside the HaCaTcells. The expression of TLR3 can be elevated in the presence of DEX and poly (I: C),while there's no effect of FK506 and MPA on the expression of TLR3. Single dose ofpoly (I: C)、ODN221, FK506 and MPA exerts no effect on the expression of TLR3inside the cells, but it will be significantly elevated in the presence of DEX and poly(I: C). ODN2216 has no effect on the expression of TLR3, but it will increase theexpression of TLR3 inside the cell in the presence of DEX. When given together withODN2216, DEX can significantly increase the expression of TLR9, and so does MPA.But FK506 exerts no effect over the expression of TLR9. The expression level insideand outside the cell will be elevated in the presence of DEX and poly (I: C).
     3. Western blotting results showed that DEX、MPA can significantly increase theexpression of MyD88. And FK506 has no effect on the expression of TLR3、TLR9and their signaling pathway.
     4. ELISA results showed that the secretion of IL-4 and IFN-γhas no difference aftersingle treatment with DEX、FK506 and MPA only (the MPA group has a significantdepression in 12h.). But in the presence of poly (I: C), DEX and MPA can significantly reduce the secretion of IL-4. And the secretion of IL-4 can also besignificantly suppressed in the presence of ODN2216 and DEX. MPA only exerts itssuppression at 24h while FK506 has significant suppression at both 12h and 24h.DEX、FK506 and MPA cannot suppress the secretion of IFN-γin the presence ofpoly(I: C). And only MPA has significant suppression both at 12h and 24h.
     The results of the studies suggest that the above three kinds of drugs cansuppress the expression of TLR3、TLR9 in skin cells, and increase the secretion ofcytokines, reduce the expression of cytokines, therefore exert a anti-inflammatoryeffect through their effect over the MyD88 singling pathway.
     Part Five: The design of TLR9-targeted HBsAg vaccine and study of themechanism of its transdermal immune response.
     1. Preparation of the flexible liposome. Construct the eukaryon vector pVAX/HBS-CpG which can express the Pre-S2/S protein and CpG sequence at the sametime. The Hepatitis B DNA vaccine cation flexible liposome was successfullyconstructed with the material of biodegradable and skin compatible DPPC andsoftener of octadecylamine. Investigate the size, distribution, shape, deformation rateand release rate, and determine the in vivo immune effect. The liposome was preparedby retro-evaporation and high-pressure homogen. Then investigate various parameters.Immunohistochemical assays were carried out to identify the distribution of HBsAgand titration of the antibody in the skin after local dermal administration of thevaccine. The results showed that the flexible liposome are transparent suspension witha encapsulating rate of (64.5±7.84)%, the diameter is (110.5±17.7) nm. EM showeda round shape. And no significant difference observed after storage under 4℃for 6months. Immunohistochemistry results showed strong positive expression of HBsAgat day 2, 3, 5, 7, with a main distribution in the hair follicles and tubular gland.
     2. The in vivo immune study showed that the IgG titration has no significantdifference with the use of flexible liposome. Transdermal administration of theflexible liposome can induce a titration of antibody 3-4 fold higher than conventionalliposome. And the flexible liposome has a higher penetrativity and immunity, with more effect afterlocal administration to prevent the hepatitis B.(p>0.05).
     3. Detect the effect of vacant liposome and HBsAg plasmid DNA liposome on theexpression of TLR3、TLR9 in HaCaT cells and FB cells by means of real-time PCR.The results showed that both cells has an increased expression of TLR3、TLR9, butthe expression levels are significantly different (p<0.05). And there is no significantdifference in the receptor expression after treatment of vacant liposome and HBsAgplasmid DNA liposome (p>0.05).
     4. Detect the effect of vacant liposome and HBsAg plasmid DNA liposome on theexpression levels of TLR3、TLR9 inside and on the surface of HaCaT cells and FBcells by means of FC. MFI of the TLR3、TLR9 on the membrane of HaCaT cells hasincreased 32.1%and 34.7%respectively, but the expression levels are significantlydifferent (p<0.05). And there is no significant difference in the receptor expressionafter treatment of vacant liposome and HBsAg plasmid DNA liposome (p>0.05).
     5. Investigate the effect of vacant liposome and HBsAg plasmid DNA liposome on theexpression levels of MyD88 in HaCaT cells and FB cells by means of westernblotting. The results showed that the expression levels of MyD88 increased at 4h aftervacant liposome and HBsAg plasmid DNA liposome treatment, without adose-dependant fashion, which indicates an alternative pathway other than MyD88 inthe signaling of TLR in the two kinds of cells.
     6. Investigate the effect of vacant liposome and HBsAg plasmid DNA liposome on thetime-effect relation of the secretion of Th1 and Th2 cytokines (IFN-γ, and IL-4) inHaCaT cells and FB cells by means of ELISA. The results showed that there's a dose-and time-dependant increase after vacant liposome and HBsAg plasmid DNAliposome treatment, with a maximum effect at 48h and the expression level in HBsAgplasmid DNA liposome group has a significant elevation (p<0.05).
     The study in this part is to design HBsAg vaccine targeted in TLR9 and maketransdermal immune possible. It will enhance our knowledge about the underlyingmechanism, and provide a helpful reference to the designation and enhancement ofthe immune response. The results showed that the vaccine can increase the immunitythrough the increase of TLRs which provide a theoretical foundation for thedevelopment of transdermal immunological drugs in the future.
     In conclusion, this study elucidates the expression pattern of TLR3、TLR9receptors in the skin in various ways, discusses the role and regulation mechanism intransdermal and immunosuppressant action, and provides a new target for thetreatment of inflammatory diseases. The discussion about the possibility of effectsexerted by different immunosuppressants on the target provides a new insight into thedevelopment of new TLR promoter and antagonist, and provides a technical supportand theoretical foundation for the development of transdermal immunological drugsin the future.
引文
1. Ulevitch RJ. Therapeutics targeting the innate immune system. Nat Rev Immunol. 2004; 4(7):512-20.
    2. Ulevitch RJ, Mathison JC, Correia J. Innate immune responses during infection. Vaccine. 2004; 22 Suppl 1:S25-30.
    3. Kawai T, Akira S. Innate immune recognition of viral infection. Nat Immunol. 2006; 7(2):131-7.
    4. Zhong B, Tien P, Shu HB. Innate immune responses: crosstalk of signaling and regulation of gene transcription. Virology. 2006; 352(1): 14-21.
    5. Trinchieri G, Sher A. Cooperation of Toll-like receptor signals in innate immune defence. Nat Rev Immunol. 2007; 7(3): 179-90.
    6. Uematsu S, Akira S. Toll-like receptors and innate immunity. J Mol Med. 2006; 84(9):712-25.
    7. Pasare C, Medzhitov R. Toll-like receptors: linking innate and adaptive immunity. Microbes Infect. 2004; 6(15):1382-7.
    8. Sandor F, Buc M. Toll-like receptors. I. Structure, function and their ligands. Folia Biol (Praha). 2005; 51(5):148-57.
    9. West AP, Koblansky AA, Ghosh S. Recognition and signaling by toll-like receptors. Annu Rev Cell Dev Biol. 2006; 22:409-37.
    10. Sandor F, Buc M. Toll-iike receptors. Ⅱ. Distribution and pathways involved in TLR signalling. Folia Biol (Praha). 2005; 51 (6): 188-97.
    11. Yarovinsky F, Zhang D, Andersen JF, et al. TLR11 activation of dendritic cells by a protozoan profilin-like protein. Science. 2005, 308(5728): 1626-9.
    12. Kaisho T, Akira S. Toll-like receptor function and signaling. J Allergy CIin Immunol. 2006; 117(5):979-87.
    13. Martin MU, Wesche H. Summary and comparison of the signaling mechanisms of the Toll/interleukin-1 receptor family. Biochim Biophys Acta. 2002; 1592(3):265-80.
    14. Oda K, Kitano H. A comprehensive map of the toll-like receptor signaling network. Mol Syst Biol. 2006; 2:1-20.
    
    15. Doyle SL, O'Neill LA. Toll-like receptors: from the discovery of NF-kappaB to new insights into transcriptional regulations in innate immunity. Biochem Pharmacol. 2006; 72(9):1102-13.
    
    16. Parker LC, Prince LR, Sabroe I. Translational mini-review series on Toll-like receptors: networks regulated by Toll-like receptors mediate innate and adaptive immunity. Clin Exp Immunol. 2007; 147(2): 199-207.
    
    17. Dunne A, O'Neill LA. The interleukin-1 receptor/Toll-like receptor superfamily:signal transduction during inflammation and host defense. Sci STKE. 2003 25;2003(171):re3
    
    18. Pasare C, Medzhitov R. Toll-like receptors: linking innate and adaptive immunity.Adv Exp Med Biol. 2005; 560:11-8.
    
    19. Hoebe K, Janssen E, Beutler B. The interface between innate and adaptive immunity. Nat Immunol. 2004; 5(10):971-4.
    
    20. Heine H, Lien E. Toll-like receptors and their function in innate and adaptive immunity. Int Arch Allergy Immunol. 2003; 130(3):180-92.
    
    21. Netea MG, Meer JW, Sutmuller RP, et al. From the Thl/Th2 paradigm towards a Toll-like receptor/T-helper bias. Antimicrob Agents Chemother. 2005;49(10):3991-6.
    
    22. Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat Rev Immunol. 2003; 3(2):133-46.
    
    23. Amati L, Pepe M, Passeri ME, et al. Toll-like receptor signaling mechanisms involved in dendritic cell activation: potential therapeutic control of T cell polarization. Curr Pharm Des. 2006; 12(32):4247-54.
    
    24. Liu X, Mosoian A, Li-Yun Chang T, et al. Gonococcal lipooligosaccharide suppresses HIV infection in human primary macrophages through induction of innate immunity. J Infect Dis. 2006; 194(6):751-9.
    
    25. Mahfouz M, Hashimoto W, Das Gupta TK, et al. Bacterial proteins and CpG-rich extrachromosomal DNA in potential cancer therapy. Plasmid. 2007; 57(1):4-17.
    26. Romagne F. Current and future drugs targeting one class of innate immunity receptors: the Toll-like receptors. Drug Discov Today. 2007; 12(1-2):80-7.
    
    27. Sandor F, Buc M. Toll-like receptors. III. Biological significance and impact for human medicine. Folia Biol (Praha). 2005; 51(6): 198-203.
    
    28. Homer AA. Update on toll-like receptor ligands and allergy: implications for immunotherapy. Curr Allergy Asthma Rep. 2006; 6(5):395-401.
    
    29. Cristofaro P, Opal SM. Role of Toll-like receptors in infection and immunity: clinical implications. Drugs. 2006; 66(1): 15-29.
    
    30. Bowie AG. Translational mini-review series on Toll-like receptors: recent advances in understanding the role of Toll-like receptors in anti-viral immunity.Clin Exp Immunol. 2007; 147(2):217-26.
    
    31. Bos JD, Kapsenberg ML. The skin immune system: progress in cutaneous biology. Immunol Today. 1993; 14(2):75-8.
    
    32. Sorensen OE, Thapa DR, Rosenthal A, et al. Differential regulation of beta defensin expression in human skin by microbial stimuli. J Immunol. 2005;174(8):4870-9.
    
    33. Greene CM, Mc Elvaney NG. Toll-like receptor expression and function in airway epithelial cells. Arch Immunol Ther Exp. 2005; 53(5):418-27.
    
    34. Cario E, Podolsky DK. Toll-like receptor signaling and its relevance to intestinal inflammation. Ann N Y Acad Sci. 2006; 1072:332-8.
    
    35. Song PI, Park YM, Abraham T, Harten B, et al. Human keratinocytes express functional CD14 and toll-like receptor 4 J Invest Dermatol. 2002; 119(2):424-32.
    
    36. Kawai K, Shimura H, Minagawa M, et al. Expression of functional Toll-like receptor 2 on human epidermal keratinocytes. J Dermatol Sci. 2002;30(3):185-94.
    
    37. Takeuchi J, Watari E, Shinya E, et al. Down-regulation of Toll-like receptor expression in monocyte-derived Langerhans cell-like cells: implications of low-responsiveness to bacterial components in the epidermal Langerhans cells.Biochem Biophys Res Commun. 2003; 306(3):674-9.
    38. Mildner M, Eckhart L, Lengauer B, et al. Hepatocyte growth factor/scatter factor inhibits UVB induced apoptosis of human keratinocytes via the PI-3-kinase pathway. J Invest Dermatol 1999; 113(6):1136-7.
    
    39. Chaturvedi V, Qin JZ, Denning MF, et al. Apoptosis in proliferating, senescent, and immortalized keratinocytes. J Biol Chem. 1999; 274(33):23358-67.
    
    40. Dobrovolskaia MA, Medvedev AE, Thomas KE, et al. Induction of in vitro reprogramming by Toll-like receptor (TLR) 2 and TLR4 agonists in murine macrophages: effects of TLR "homotolerance" versus "heterotolerance" on NF-kappa B signaling pathway components. J Immunol. 2003; 170(1):508-19.
    
    41. Poynter ME, Cloots R, Woerkom T, et al. NF-kappa B activation in airways modulates allergic inflammation but not hyperresponsiveness. J Immunol. 2004;173(11):7003-9.
    
    42. O'neill LA, Bowie AG. The family of five: TIR-domain-containing adaptors in Toll-like receptor signalling. Nat Rev Immunol. 2007; 7(5):353-64.
    
    43. Chi H, Barry SP, Roth RJ, et al. Dynamic regulation of pro- and anti-inflammatory cytokines by MAPK phosphatase 1 (MKP-1) in innate immune responses. Proc Natl Acad Sci USA. 2006; 103(7):2274-9.
    
    44. Rao N, Nguyen S, Ngo K, et al. A novel splice variant of interleukin-1 receptor (IL-lR)-associated kinase 1 plays a negative regulatory role in Toll/IL-1R-induced inflammatory signaling. Mol Cell Biol. 2005;25(15):6521-32.
    
    45. Mohamadzadeh M, Chen L, Olinger GG, et al. Filoviruses and the balance of innate, adaptive, and inflammatory responses. Viral Immunol. 2006;19(4):602-12.
    
    46. Shimura H, Nitahara A, Ito A, et al. Up-regulation of cell surface Toll-like receptor 4-MD2 expression on dendritic epidermal T cells after the emigration from epidermis during cutaneous inflammation. J Dermatol Sci. 2005;37(2):101-10.
    47. Su X, Li S, Meng M, Qian W, et al. TNF receptor-associated factor-1 (TRAF1) negatively regulates Toll/IL-1 receptor domain-containing adaptor inducing IFN-beta (TRIF)-mediated signaling. Eur J Immunol. 2006; 36(1): 199-206.
    
    48. Tokumaru S, Sayama K, Shirakata Y, et al. Induction of keratinocyte migration via transactivation of the epidermal growth factor receptor by the antimicrobial peptide LL-37.J Immunol. 2005; 175(7):4662-8.
    
    49. Aarbiou J, Ertmann M, Wetering S, et al. Human neutrophil defensins induce lung epithelial cell proliferation in vitro. J Leukoc Biol. 2002; 72(1): 167-74.
    
    50. Akashi-Takamura S, Miyake K. Toll-like receptors (TLRs) and immune disorders.J Infect Chemother. 2006; 12(5):233-40.
    
    51. Barrera S, Aleman M, Sasiain Mdel C. Toll-like receptors in human infectious diseases. Curr Pharm Des. 2006; 12(32):4173-84.
    
    52. Mullick AE, Tobias PS, Curtiss LK. Toll-like receptors and atherosclerosis: key contributors in disease and health? Immunol Res. 2006; 34(3): 193-209.
    
    53. Mori Y, Yoshimura A, Ukai T, et al. Immunohistochemical localization of Toll-like receptors 2 and 4 in gingival tissue from patients with periodontitis.Oral Microbiol Immunol. 2003; 18(1):54-8.
    
    54. Cario E, Podolsky DK. Toll-like receptor signaling and its relevance to intestinal inflammation. Ann N Y Acad Sci. 2006; 1072:332-8.
    
    55. Gustot T, Lemmers A, Moreno C, et al. Differential liver sensitization to toll-like receptor pathways in mice with alcoholic fatty liver. Hepatology. 2006;43(5):989-1000.
    
    56. Candia L, Marquez J, Hernandez C, et al. Toll-like receptor-2 expression is upregulated in antigen-presenting cells from patients with psoriatic arthritis: a pathogenic role for innate immunity? J Rheumatol. 2007;34(2):374-9.
    
    57. Farina C, Theil D, Semlinger B, et al. Distinct responses of monocytes to Toll-like receptor ligands and inflammatory cytokines. Int Immunol. 2004;16(6):799-809.
    
    58. Hayashi F, Means TK, Luster AD. Toll-like receptors stimulate human neutrophil function. Blood. 2003; 102(7):2660-9.
    59. Kurt-Jones EA, Mandell L, Whitney C, et al. Role of toll-like receptor 2 (TLR2) in neutrophil activation: GM-CSF enhances TLR2 expression and TLR2-mediated interleukin 8 responses in neutrophils. Blood. 2002;100(5):1860-8.
    
    60. Faure E, Thomas L, Xu H, et al. Bacterial lipopolysaccharide and IFN-gamma induce Toll-like receptor 2 and oil-like receptor 4 expression in human endothelial cells: role of NF-kappa B activation. J Immunol. 2001;166(3):2018-24.
    
    61. Sing A, Tvardovskaia N, Rost D, et al. Contribution of toll-like receptors 2 and 4 in an oral Yersinia enterocolitica mouse infection model. Int J Med Microbiol.2003;293(5):341-8.
    
    62. Moqattash S, Lutton JD. Leukemia cells and the cytokine network. Proc Soc Exp Biol Med. 1998; 219(l):8-27.
    
    63. Negishi H, Ohba Y, Yanai H, et al. Negative regulation of Toll-like-receptor signaling by IRF-4.Proc Natl Acad Sci. 2005;102(44): 15989-94.
    
    64. Liew FY, Xu D, Brint EK, et al. Negative regulation of toll-like receptor-mediated immune responses. Nat Rev Immunol. 2005; 5(6):446-58.
    
    65. Camateros P, Moisan J, Henault J, et al. Toll-like receptors, cytokines and the immunotherapeutics of asthma. Curr Pharm Des. 2006; 12(19):2365-74.
    
    66. Kang SS, Kauls LS, Gaspari AA. Toll-like receptors: applications to dermatologic disease. J Am Acad Dermatol. 2006; 54(6):951-83.
    
    67. Boehm I. Apoptosis in physiological and pathological skin: implications for therapy. Curr Mol Med. 2006; 6(4):375-94.
    
    68. Esche C, Benedetto A, Beck LA. Keratinocytes in atopic dermatitis: inflammatory signals. Curr Allergy Asthma Rep. 2004; 4(4):276-84.
    
    69. Kim YM, Romero R, Chaiworapongsa T, et al. Dermatitis as a component of the fetal inflammatory response syndrome is associated with activation of Toll-like receptors in epidermal keratinocytes. Histopathology. 2006; 49(5):506-14.
    
    70. Kang SS, Kauls LS, Gaspari AA. Toll-like receptors: applications to dermatologic disease. J Am Acad Dermatol. 2006; 54(6):951-83.
    71. Rogatsky I, Ivashkiv LB. Glucocorticoid modulation of cytokine signaling.Tissue Antigens. 2006; 68(1):1-12.
    
    72. Necela BM, Cidlowski JA. Mechanisms of glucocorticoid receptor action in noninflammatory and inflammatory cells. Proc Am Thorac Soc. 2004;1(3):239-46.
    
    73. Smoak KA, Cidlowski JA. Mechanisms of glucocorticoid receptor signaling during inflammation. Mech Ageing Dev. 2004; 125(10-11):697-706.
    
    74. Bekersky I, Lilja H, Lawrence I. Tacrolimus pharmacology and nonclinical studies: from FK506 to protopic. Semin Cutan Med Surg. 2001; 20(4):226-32.
    
    75. Dumont FJ. FK506, an immunosuppressant targeting calcineurin function. Curr Med Chem. 2000; 7(7):731-48.
    
    76. Allison AC, Eugui EM. Mycophenolate mofetil and its mechanisms of action.Immunopharmacology. 2000; 47(2-3):85-118.
    
    77. Akashi-Takamura S, Miyake K. Toll-like receptors (TLRs) and immune disorders.J Infect Chemother. 2006; 12(5):233-40.
    
    78. Barrera S, Aleman M, Sasiain Mdel C. Toll-like receptors in human infectious diseases. Curr Pharm Des. 2006; 12(32):4173-84.
    
    79. Romagne F. Current and future drugs targeting one class of innate immunity receptors: the Toll-like receptors. Drug Discov Today. 2007; 12(l-2):80-7.
    
    80. Sandor F, Buc M. Toll-like receptors. III. Biological significance and impact for human medicine. Folia Biol (Praha). 2005; 51(6): 198-203.
    
    81. Horner AA. Update on toll-like receptor ligands and allergy: implications for immunotherapy. Curr Allergy Asthma Rep. 2006; 6(5):395-401.
    
    82. Camateros P, Moisan J, Henault J, et al. Toll-like receptors, cytokines and the immunotherapeutics of asthma. Curr PharmDes. 2006; 12(19):2365-74.
    
    83. Kang SS, Kauls LS, Gaspari AA. Toll-like receptors: applications to dermatologic disease. J Am Acad Dermatol. 2006; 54(6):951-83.
    
    84. Cristofaro P, Opal SM. Role of Toll-like receptors in infection and immunity:clinical implications. Drugs. 2006; 66(1):15-29.
    85. Hermoso MA, Matsuguchi T, Smoak K, et al. Glucocorticoids and tumor necrosis factor alpha cooperatively regulate toll-like receptor 2 gene expression. Mol Cell Biol 2004; 24(11):4743-56.
    
    86. Bornstein SR, Ziegler CG, Krug AW, et al. The role of toll-like receptors in the immune-adrenal crosstalk. Ann N Y Acad Sci. 2006; 1088:307-18.
    
    87. Lan CC, Kao YH, Huang SM, et al. FK506 independently upregulates transforming growth factor beta and downregulates inducible nitric oxide synthase in cultured human keratinocytes: possible mechanisms of how tacrolimus ointment interacts with atopic skin.Br J Dermatol. 2004;151(3):679-84.
    
    88. Michel G, Kemeny L, Homey B, et al. FK506 in the treatment of inflammatory skin disease: promises and perspectives. Immunol Today. 1996; 17(3): 106-8.
    
    89. Mydlarski PR. Mycophenolate mofetil: a dermatologic perspective. Skin Therapy Lett. 2005; 10(3):1-6.
    
    90. Rezaei N. Therapeutic targeting of pattern-recognition receptors. Int Immunopharmacol. 2006; 6(6):863-9.
    
    91. Duin D, Medzhitov R, Shaw AC. Triggering TLR signaling in vaccination.Trends Immunol. 2006; 27(l):49-55.
    
    92. Krieg AM. CpG motifs in bacterial DNA and their immune effects. Annu Rev Immunol. 2002; 20:709-60.
    
    93. McCluskie MJ, Krieg AM. Enhancement of infectious disease vaccines through TLR9-dependent recognition of CpG DNA. Curr Top Microbiol Immunol. 2006;311:155-78.
    
    94. Jong S, Chikh G, Sekirov L, et al. Encapsulation in liposomal nanoparticIes enhances the immunostimulatory, adjuvant and anti-tumor activity of subcutaneously administered CpG ODN. Cancer Immunol Immunother. 2007 Jan 23.[Epub ahead of print]
    
    95. Jiang W, Reich CF, Pisetsky DS. In vitro assay of immunostimulatory activities of plasmid vectors. Methods Mol Med. 2006; 127:55-70.
    96. Weeratna RD, Makinen SR, McCluskie MJ, et al. TLR agonists as vaccine adjuvants: comparison of CpG ODN and Resiquimod (R-848).Vaccine. 2005;23(45):5263-70.
    
    97. Tudor D, Dubuquoy C, Gaboriau V, et al. TLR9 pathway is involved in adjuvant effects of plasmid DNA-based vaccines. Vaccine. 2005; 23(10):1258-64.
    
    98. Klinman DM. CpG DNA as a vaccine adjuvant. Expert Rev Vaccines. 2003;2(2):305-15.
    
    99. Krieg AM. Therapeutic potential of Toll-like receptor 9 activation. Nat Rev Drug Discov.2006; 5(6):471-84.
    
    100.Schneeberger A, Wagner C, Zemann A, et al. CpG motifs are efficient adjuvants for DNA cancer vaccines. J Invest Dermatol. 2004; 123(2):371-9.
    101.Hammond, S.A., M. Guebre-Xabier, J. Yu, et al, Transcutaneous immunization:an emerging route of immunization and potent immunostimulation strategy. Crit Rev Ther Drug Carrier Syst. 2001,18(5): 503-26.
    102.Glenn G.M., R.T. Kenney, S.A. Hammond, et al., Transcutaneous immunization and immunostimulant strategies. Immunol Allergy Clin North Am. 2003, 23(4):787-813.
    103.Suzuki Y, Wakita D, Chamoto K, et al. Liposome-encapsulated CpG oligodeoxynucleotides as a potent adjuvant for inducing type 1 innate immunity.Cancer Res. 2004;64(23):8754-60.
    104.Yerly D, Di Giammarino L, Bih1 F, et al. Targets of emerging therapies for viral hepatitis B and C. Expert Opin Ther Targets. 2006; 10(6):833-50.
    105.Visvanathan K, Lewin SR. Immunopathogenesis: role of innate and adaptive immune responses. Semin Liver Dis. 2006; 26(2):104-15
    1. Katz, S. L., Future vaccines and a global perspective [J]. Lancet. 1997, 350(9093): 1767-70.
    2. Liu, M. A., Vaccine developments [J]. Nat Med. 1998, 4(5 Suppl): 515-9.
    3. Hammond, S. A., M. Guebre-Xabier, J. Yu, et al., Transcutaneous immunization: an emerging route of immunization and potent immunostimulation strategy [J]. Crit Rev Ther Drug Carrier Syst. 2001, 18(5): 503-26.
    4. Glenn, G. M., R. T. Kenney, S. A. Hammond, et al., Transcutaneous immunization and immunostimulant strategies [J]. Immunol Allergy Clin North Am. 2003, 23(4): 787-813.
    5. Bos, J. D., The skin as an organ of immunity [J]. Clin Exp Immunol. 1997, 107 Suppl 1: 3-5.
    6. Lappin, M. B., I. Kimber, and M. Norval, The role of dendritic cells in cutaneous immunity [J]. Arch Dermatol Res. 1996, 288(3): 109-21.
    7. Berger, C. L. and R. Edelson, The life cycle of cutaneous T cell lymphoma reveals opportunities for targeted drug therapy [J]. Curr Cancer Drug Targets. 2004, 4(7): 609-19.
    8. Chen, D. and L. G. Payne, Targeting epidermal Langerhans cells by epidermal powder immunization [J]. Cell Res. 2002, 12(2): 97-104.
    9. Kaiserlian, D. and N. Etchart, Epicutaneous and transcutaneous immunization using DNA or proteins [J]. Eur J Dermatol. 1999, 9(3): 169-76.
    10. Peachman, K. K., M. Rao, and C. R. Alving, Immunization with DNA through the skin [J]. Methods. 2003, 31(3): 232-42.
    
    11. Glenn, G.M., M. Rao, G.R. Matyas, et al., Skin immunization made possible by cholera toxin [J]. Nature. 1998, 391(6670): 851.
    
    12. Scharton-Kersten, T., J. Yu, R. Vassell, et al., Transcutaneous immunization with bacterial ADP-ribosylating exotoxins, subunits, and unrelated adjuvants[J]. Infect Immun. 2000, 68(9): 5306-13.
    
    13. Anjuere, E, A. George-Chandy, F. Audant, et al., Transcutaneous immunization with cholera toxin B subunit adjuvant suppresses IgE antibody responses via selective induction of Th1 immune responses [J]. J Immunol. 2003,170(3): 1586-92.
    
    14. Henseleit, U., T. Rosenbach, and G. Kolde, Induction of apoptosis in human HaCaT keratinocytes[J]. Arch Dermatol Res. 1996,288(11): 676-83.
    
    15. Tierney, R., A.S. Beignon, R. Rappuoli, et al., Transcutaneous immunization with tetanus toxoid and mutants of Escherichia coli heat-labile enterotoxin as adjuvants elicits strong protective antibody responses [J]. J Infect Dis. 2003,188(5): 753-8.
    
    16. Beignon, A.S., J.R Briand, R. Rappuoli et al., The LTR72 mutant of heat-labile enterotoxin of Escherichia coli enhances the ability of peptide antigens to elicit CD4(+) T cells and secrete gamma interferon after coapplication onto bare skin[J]. Infect Immun. 2002,70(6): 3012-9.
    
    17. Krieg, A.M., CpG motifs in bacterial DNA and their immune effects [J]. Annu Rev Immunol. 2002, 20: 709-60.
    
    18. Beignon, A.S., J.R Briand, S. Muller, et al., Immunization onto bare skin with synthetic peptides: immunomodulation with a CpG-containing oligodeoxynucleotide and effective priming of influenza virus-specific CD4+ T cells [J]. Immunology. 2002,105(2): 204-12.
    
    19. Partidos, CD., A.S. Beignon, J.P. Briand, et al., Modulation of immune responses with transcutaneously deliverable adjuvants[J]. Vaccine. 2004,22(19): 2385-90.
    
    20. Watabe, S., K.Q. Xin, A. Ihata, et al., Protection against influenza virus challenge by topical application of influenza DNA vaccine [J]. Vaccine. 2001,19(31): 4434-44.
    
    21. Frech, S.A., R.T. Kenney, C.A. Spyr, et al., Improved immune responses to influenza vaccination in the elderly using an immunostimulant patch [J]. Vaccine. 2005, 23(7): 946-50.
    
    22. Yu, J., F. Cassels, T. Scharton-Kersten, et al, Transcutaneous immunization using colonization factor and heat-labile enterotoxin induces correlates of protective immunity for enterotoxigenic Escherichia coli [J]. Infect Immun. 2002, 70(3):1056-68.
    
    23. Guerena-Burgueno, F., E.R. Hall, D.N. Taylor, et al, Safety and immunogenicity of a prototype enterotoxigenic Escherichia coli vaccine administered transcutaneously[J].Infect Immun. 2002, 70(4): 1874-80.
    
    24. Partidos, CD., E. Moreau, O. Chaloin, et al, A synthetic HIV-1 Tat protein breaches the skin barrier and elicits Tat-neutralizing antibodies and cellular immunity [J]. Eur J Immunol. 2004, 34(12): 3723-31.
    
    25. Belyakov, I.M., S.A. Hammond, J.D. Ahlers, et al, Transcutaneous immunization induces mucosal CTLs and protective immunity by migration of primed skin dendritic cells [J].JClin Invest. 2004,113(7): 998-1007.
    
    26. Matyas, G.R., A.M. Friedlander, G.M. Glenn, et al., Needle-free skin patch vaccination method for anthrax[J]. Infect Immun. 2004, 72(2): 1181-3.
    
    27. Kenney, R.T., J. Yu, M. Guebre-Xabier, et al, Induction of protective immunity against lethal anthrax challenge with a patch [J]. J Infect Dis. 2004,190(4): 774-82.
    
    28. Hashimoto, M., J.L. Boyer, N.R. Hackett, et al., Induction of protective immunity to anthrax lethal toxin with a nonhuman primate adenovirus-based vaccine in the presence of preexisting anti-human adenovirus immunity[J]. Infect Immun. 2005,73(10): 6885-91.
    
    29. Eyles, J.E., S.J. Elvin, A. Westwood, et al, Immunisation against plague by transcutaneous and intradermal application of subunit antigens [J]. Vaccine. 2004,22(31-32): 4365-73.
    
    30. El-Ghorr, A.A., R.M. Williams, C. Heap, et al, Transcutaneous immunisation with herpes simplex virus stimulates immunity in mice [J]. FEMS Immunol Med Microbiol.2000, 29(4): 255-61.
    
    31. Berry, L.J., D.K. Hickey, K.A. Skelding, et al., Transcutaneous immunization with combined cholera toxin and CpG adjuvant protects against Chlamydia muridarum genital tract infection[J]. Infect Immun. 2004, 72(2): 1019-28.
    
    32. Beignon, A.S., F. Brown, P. Eftekhari, et al., A peptide vaccine administered transcutaneously together with cholera toxin elicits potent neutralising anti-FMDV antibody responses[J]. Vet Immunol Immunopathol. 2005,104(3-4): 273-80.
    
    33. Godefroy, S., L. Goestch, H. Plotnicky-Gilquin, et al, Immunization onto shaved skin with a bacterial enterotoxin adjuvant protects mice against respiratory syncytial virus (RSV) [J]. Vaccine. 2003,21(15): 1665-71.
    
    34. Ghoi, A.H., K. Smiley, and M. Basu, Induction of immune responses and partial protection in mice after skin immunization with rotavirus VP6 protein and the adjuvant LT(R192G)[J]. Vaccine. 2005,23(17-18): 2290-3.
    
    35. Mawas, R, M. Peyre, A.S. Beignon, et al, Successful induction of protective antibody responses against Haemophilus influenzae type b and diphtheria after transcutaneous immunization with the glycoconjugate polyribosyl ribitol phosphate-cross-reacting material 197 vaccine[J]. J Infect Dis. 2004,190(6): 1177-82.

© 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