用户名: 密码: 验证码:
DcR3在食管鳞状细胞癌组织中的表达及相关机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
背景
     食管癌是人类八种最常见的恶性肿瘤之一。在全世界每年新增的30万病例中,约70 %发生在我国,并且其发病率仍呈上升趋势。食管癌总体预后不佳,总的2年和5年生存率分别为35%~42%和15%~24%。食管癌主要分为食管鳞状细胞癌(ESCC)和食管腺癌,而前者占了90%以上。因此,研究ESCC的发病机制,寻找其新的诊断和治疗靶点,已经成为提高食管癌预防和治疗效果的当务之急。在肿瘤的发生、发展中有一对矛盾贯穿始终,那就是机体的免疫监视和肿瘤的免疫逃逸。而诱导肿瘤特异性的细胞毒性T淋巴细胞(CTL)反应和瘤细胞自身生长停滞或凋亡是机体用以清除肿瘤细胞的主要手段。LIGHT及FasL介导的凋亡是体内最常见的凋亡模式,在免疫调节和肿瘤的免疫监视中起着重要作用。DcR3是TNFRSF的一个新成员。研究发现,DcR3通过竞争性结合可以阻断经由其配体FasL、LIGHT以及TL1A所介导的肿瘤细胞的凋亡;同时DcR3还可抑制免疫应答中T细胞的增殖并影响DCs的分化和成熟,并抑制CD4+T细胞在混合淋巴细胞反应(MLR)中的增殖;此外,DcR3还可抑制T细胞的趋化效应,降低T细胞与抗原提呈细胞间的相互作用。研究显示,DcR3 mRNA低表达于正常人体组织中的胃、脊髓、淋巴结、肺、脾、结肠组织及多数造血细胞中,而高表达于胃肠道恶性肿瘤、乳腺癌、结肠癌、肺癌、胶质细胞瘤和淋巴瘤等多种恶性肿瘤中。研究证实,高表达DcR3是肿瘤细胞逃避机体免疫监视和杀伤的一个重要机制,其表达水平同多种肿瘤的进展程度、转移情况、对化疗的抵抗性以及不良预后有明显的相关性。因此,作为一种候选的肿瘤标志物和基因治疗标靶,DcR3目前已经成为了肿瘤诊断和治疗相关研究的热点。研究DcR3 mRNA和蛋白在人ESCC细胞系和ESCC病人组织标本中的表达情况并分析DcR3的表达与疾病的侵袭、转移、复发乃至预后间的关联性,将有助于评价DcR3作为ESCC的临床及预后的分子标志的可能性;而探讨其表达调控的可能机制则将有助于ESCC肿瘤基因治疗靶点的筛选。因此,关于DcR3在ESCC病人中的高表达及其相关机制的研究对于该疾病的防治有相当重要的意义。
     目的
     研究DcR3基因在ESCC病人肿瘤组织和相应的远癌组织中的表达情况,分析DcR3基因的表达与病人临床病理特征之间的关系,为评价DcR3作为ESCC的临床及预后的分子标志提供实验依据;检测DcR3基因启动子和编码区的多态性位点在重庆地区汉族ESCC人群中的基因型和等位基因频率,探讨DcR3启动子多态性与ESCC患病风险之间的关联,筛查与ESCC肿瘤组织中DcR3基因高表达相关联的SNP位点,为进一步研究和分析影响其表达调控的分子机制奠定基础;检测DcR3基因启动子区的甲基化状态差异,探讨ESCC肿瘤组织DcR3启动子甲基化状态是否参与其表达调控;考察DcR3 RNAi对ESCC肿瘤细胞的生长能力、侵袭能力和凋亡等方面的影响,初步探讨DcR3在人ESCC细胞发生和发展过程中可能作用和机制。
     方法
     1.收集109例ESCC肿瘤组织和对应的远癌组织标本,通过半定量逆转录聚合酶链反应(Semi-quantitative RT-PCR)检测其中DcR3 mRNA表达情况;
     2.运用免疫组织化学检测52例ESCC病人肿瘤组织中DcR3蛋白表达情况并行半定量分析;
     3.选取DcR3基因启动子和编码区四个多态性位点(-369G/T、-323A/C、-321C/T、147C/T),通过PCR扩增产物测序的方法了解这些多态位点在重庆地区正常人群中的频率分布,检测上述多态位点在ESCC人群中的基因型和等位基因频率;
     4.选择4对DcR3 mRNA表达状态有明显差异的癌及相对应的远癌组织标本,利用重亚硫酸盐修饰后测序法(BSP)检测DcR3基因启动子区的甲基化状态;
     5.构建基于miR-30的、靶向DcR3的慢病毒干扰载体;
     6.用干扰载体、包装质粒pMD2G及包膜质粒psAX2共同转染293FT细胞进行病毒包装;
    
     7.通过流式细胞仪(FACS)感染后的KYSE150细胞,获得DcR3 RNAi的ESCC细胞模型;
     8.利用Real-time PCR和Western Blotting检测DcR3 RNAi后瘤细胞上DcR3的表达情况;
     9.绘制体外培养的感染细胞、对照细胞和亲本细胞的生长曲线;
     10.通过Transwell侵袭小室实验检测DcR3 RNAi与LIGHT-Fc协同作用对瘤细胞体外侵袭能力的影响;
     11.运用Tunel试剂盒检测DcR3 RNAi与LIGHT-Fc协同作用对瘤细胞凋亡的影响。
     结果
     1. ESCC肿瘤标本中DcR3 mRNA表达率为73.4%,而远癌组织为47.7%(p<0.01);分析发现DcR3 mRNA的表达与病人的性别、年龄、部位和组织分化程度没有明显的相关性,而与肿瘤组织的外侵程度(p<0.05)、区域淋巴结转移情况(p<0.05)和临床TNM分期(p<0.05)有较强的相关性;
     2. ESCC肿瘤组织中DcR3蛋白的表达强弱与mRNA表达基本一致,DcR3的过表达率为28.8%;DcR3的过表达与ESCC病人区域淋巴结转移(p<0.05)和TNM分期(p<0.05)有明显的相关性;
     3.重庆地区汉族人群中-369G/T、-323A/C、-321C/T三个位点不存在多态性;而147C/T位点等位基因C的频率为43.7%,略高于中国汉族人(39.0%)和日本人(40.0%)。147C/T多态位点是与重庆地区汉族人群ESCC易感性相关联的一个风险位点,而且该多态位点的等位基因型C具有显性遗传效应,携带等位基因C的基因型CC和CT的ESCC发病风险显著增加(p<0.05)。但该多态位点与ESCC肿瘤组织中DcR3蛋白的异常高表达无明显的相关性;
     4. DcR3阳性的肿瘤组织和DcR3阴性的远癌组织,基因的启动子区的甲基化状态相同,即基因启动子区所有的CpG位点都呈非甲基化状态;
     5.成功设计并构建了基于miR-30的、靶向DcR3的慢病毒干扰载体;包装获得的病毒上清在体外能有效地感染人KYSE150细胞;
     6.经FACS分选,Western Blotting检测证实载体pPRIME-DcR3.3i能有效地抑制瘤细胞DcR3的蛋白表达,抑制率为82.6%;
     7. DcR3 RNAi对KYSE150细胞的体外生长无明显影响;
     8. DcR3 RNAi与LIGHT-Fc协同能有效地抑制瘤细胞的体外侵袭能力;
     9. DcR3 RNAi明显促进了LIGHT-Fc对KYSE150细胞的凋亡诱导。
     结论
     ESCC肿瘤组织中DcR3在mRNA和蛋白质水平较之对应的远癌组织存在明显的高表达,其高表达与肿瘤的区域淋巴结转移情况和临床TNM分期有较强的相关性。重庆地区汉族人群中147C/T位点是与ESCC易感性相关联的一个风险位点,该位点的等位基因型C具有显性遗传效应;ESCC病人肿瘤组织中DcR3的异常高表达与该基因CpG岛的甲基化状态异常无关; DcR3 RNAi对肿瘤细胞的体外生长没有直接的抑制效应,但能与LIGHT-Fc协同,有效地抑制瘤细胞的体外侵袭能力并促进对瘤细胞的凋亡诱导;DcR3对于ESCC是一个潜在的肿瘤生物标记。
Background: Esophageal cancer is one of the eight most popular malignant tumors in human. Among 300,000 new diagnosed cases every year in the world, seventy percent were in China and the disease incidence still increases recently. One of the main problems of esophageal cancer is that long-term survival is very low, the totally 2-year survival rate is 35%-42% and only 15%-24% for 5-year survival. Esophageal cancer can be characterized into ESCC (esophageal squamous cell carcinoma) and esophageal adenocarcinoma, while ninty percent disease cases are ESCC. So it is very urgent to study the pathogenesis of ESCC and develop new target for diagnosis and treatment.
     As we known, the balance between the immunological surveillance of host and the immune escape of tumor is tightly regulated during the tumorigenesis and tumor development. Inducing the tumor-specific CTL response and/or the apotosis of tumor cells are two important methods to eliminate tumor cells. LIGHT and FasL are two members of TNF super family (TNFRSF). They are the most common pathway for inducing the apotosis in vivo, and play essential role in immune regulation and the tumor immune monitoring. DcR3 is a new member of TNFRSF, which can competitively bind to Fas, LIGHT and TL1A and then inhibite the apotosis of tumor cells induced by these three molecular. In the other hand, during the immune response, DcR3 can inhibit the proliferation of T cells and the differentiation and maturation of dendritic cells (DCs), especially the proliferation of CD4+ T cells in MLR. Besides, DcR3 can also inhibit the chemotaxis of T cells and reduce the interact between T cells and antigen processing cells.
     Previous studies showed that DcR3 mRNA expression level was very low in many normal human tissues, such as stomach, spinal cord, lymph node, lung, spleen, colon tissue and most hematopoietic cells, but contrarily, in many kinds of malignant tumor, including gastrointestinal cancer, breast cancer, carcinoma of colon, lung cancer, spongiocytoma and lymphoma, DcR3 mRNA expression level was very high. It was reported that overexpression of DcR3 was an important mechanism for tumor cells escaping from immune surveillance and destruction. In many different kinds of cancer, the expression level of DcR3 is clearly related with the progress of carcinogenesis, cancer metastasis, the resistance to chemotherapy and the prognosis of tumor. So, DcR3, as a candidate cancer biomarker and target for cancer gene therapy, becomes a very hot point in cancer research. Studying the expression of DcR3 on the mRNA and protein level in ESCC cell strains and patient biopsies, exploring the relationship between DcR3 expression and the invasion, metastasis, recurrence and prognosis of tumor are very helpful to evaluate the possibility of using DcR3 as a molecular marker in ESCC’s clinical diagnose and prognosis prediction. In the meantime, investigating the regulation mechanism behind DcR3 expression can help us develop a new gene therapy approach to treat ESCC. These facts suggested that studying the expression of DcR3 and how it is regulated are very important in the prevention and treatment of ESCC.
     Aims: To identify the expression of DcR3 gene in ESCC patient’s samples and analyze its possible correlation between DcR3 expression and their clinicopathologic features. The polymorphic sites frequencies of DcR3 promoter and coding region in Chongqing normal population and ESCC patients would to be detected, in order to address the possible correlation between polymorphism of DcR3 promoter and ESCC risk, and screen the SNP sites associated with DcR3 high expression. For exploring whether the the methylation was involved in the regulation of DcR3 expression, the methylation difference in promoter region of DcR3 was to be tested. In addition, the effects of DcR3 RNAi on ESCC tumor metastasis, invasion and immune escape were to be detected, and the biological behavior and the mechanism of DcR3 in the progression of human ESCC cells were also to be investigated.
     Methods: 1. Semi-quantitative RT-PCR assay was utilized to detect the expression of DcR3 mRNA in 109 ESCC tumor tissues and corresponding normal tissues far away from tumor.
     2. Immunohistochemistry and semi-quantitative analysis were performed to test DcR3 protein expressions from 52 ESCC patients.
     3. DcR3 promotor and four polymorphic sites (-369G/T、-323A/C、-321C/T、147C/T) in coding region were selected, these polymorphic sites frequencies in Chongqing normal population and their frequencies in ESCC patients were studied with PCR sequencing to address the possible correlation between polymorphism of DcR3 promoter and ESCC risk.
     4. Based on their significant difference of DcR3 expression with each other, four pairs of sample from the tumor tissues and corresponding normal tissues far away from tumor were choosed, and their methylation status of the CpG in the promoter region of DcR3 were detected by bisulfite sequencing assay(BSA).
     5. A DcR3 targeted RNA interfering lentivirus vector based on a new miR30 like was constructed to inhibit the expression of DcR3 in esophageal squamous cell line KYSE150.
     6. The verified recombined vector was co-transfect with pMD2g and psAX2 into 293FT packing cell line to produce the virus.
     7. ESCC cell model of DcR3 RNAi was obtained by FACS.
     8. The DcR3 expression level of infected cells were evaluated by Real-time PCR and Western blotting.
     9. The growth curves of infected cells, controlled cells and wild cells were drawn.
     10. The effects of DcR3 RNAi collaborated with hLIGHT-Fc on invasion activity of infected ESCC cells were studied by Transwell loculus assay.
     11. The effects of DcR3 RNAi collaborated with hLIGHT-Fc on apoptosis sensitivity of infected ESCC cells were investigated by Tunel assay.
     Results: 1. The expression of DcR3 mRNA in 109 ESCC tumor tissues was 73.4% and 47.7% in the corresponding normal tissues far away from tumor. The expression of DcR3 mRNA was no close relation with gender, age, localization and tissue differential degree of patients, but strong related with tumor infiltration (p<0.05), regional lymph node metastasis (p<0.05) and clinical TNM stage (p<0.05).
     2. DcR3 protein expression was consistent with DcR3 mRNA expression. The overexpression rate of DcR3 was 28.8%, and there was a correlation between DcR3 overexpression and regional lymph node metastasis (p<0.05) and clinical TNM stage (p<0.05).
     3. Polymorphism was found only existe at 147C/T, instead of -369G/T、-323A/C or -321 C/T, on Chongqing Han population. The minor allel frequence (MAF) was 43.7%, which was a little bit higher than the whole Chinese Han population(39.0%) and Janpense population(40.0%). 147C/T polymorphism was a risk site of susceptibility to ESCC in Chongqing Han population, and the 147C had the dominant inheritance effect comparing with 147T. The risk of susceptibility to ESCC was significantly increased in CC or CT, comparing with TT (p<0.05). But no significant correlation between this site and the upregulation of DcR3 protein was found.
     4. There was no difference of the methylation status between the cancer tissues and matched normal tissues. All the CpG sites in the promoter region were unmethylated.
     5. The new miR30 like sequence based lentivirus interfering vector was successfully constructed and the virus-containing cell culture supernatants could infect KYSE150 cells efficiently.
     6. FACS sorting and Western Blotting showed that the pPRIME-DcR3.3i vector could inhibit the expression of DcR3 protein with the 82.6% inhibition rate.
     7. There was no significant suppressing effect of DcR3 RNAi on infected KYSE150 cells.
     8. The invasion activity could be inhibited by the joint action of DcR3 RNAi and hLIGHT-Fc.
     9. DcR3 RNAi obviously promoted the apoptosis of KYSE150 cell induced by hLIGHT-Fc.
     Conclusion: The mRNA and protein expression level of DcR3 in ESCC tumor tissues are higher than that in matched normal tissues. Increased expression of DcR3 is significant related with regional lymph node metastasis and clinical TNM stage. 147C/T polymorphism is a risk site of susceptibility to ESCC in Chongqing Han population, and the 147C has the dominant inheritance effect. The abnormal high expression of DcR3 in ESCC patients is no relationship with methylation status of the CpG in the promoter region of DcR3. DcR3 RNAi has no direct inhibition on growth of ESCC, but it can inhibit invasion effectly and induce apoptosis with the collaboration of hLIGHT-Fc. Above all, DcR3 is a potential ESCC clinical biomarker.
引文
1 Kamangar F, Dores GM, Anderson WF. Patterns of cancer incidence, mortality, and prevalence across five continents: defining priorities to reduce cancer disparities in different geographic regions of the world. J Clin Oncol, 2006. 2137-50.
    2 Enzinger PC, Mayer RJ. Esophageal cancer. N Engl J Med, 2003. 2241-52.
    3 Brown LM, Hoover R, Silverman D, et al. Excess incidence of squamous cell esophageal cancer among US Black men: role of social class and other risk factors. Am J Epidemiol, 2001. 114-22.
    4 Pitti RM, Marsters SA, Lawrence DA, et al. Genomic amplification of a decoy receptor for Fas ligand in lung and colon cancer. Nature, 1998,396:699-703.
    5 Yu KY, Kwon B, Ni J, et al. A newly identified member of tumor necrosis factor receptor superfamily (TR6) suppresses LIGHT-mediated apoptosis. J Biol Chem, 1999,274:13733-6.
    6 Bai C, Connolly B, Metzker ML, et al. Overexpression of M68/DcR3 in human gastrointestinal tract tumors independent of gene amplification and its location in a four-gene cluster. PNAS, 2000,97:1230-5.
    7 Otsuki T, Tomokuni A, Sakaguchi H, et al. Over-expression of the decoy receptor 3 (DcR3) gene in peripheral blood mononuclear cells (PBMC) derived from silicosis patients. Clin Exp Immunol, 2000,119:323-7.
    8 Roth W, Isenmann S, Nakamura M, et al. Soluble decoy receptor 3 is expressed by malignant gliomas and suppresses CD95 ligand-induced apoptosis and chemotaxis. Cancer Res, 2001,61:2759-65.
    9 Arakawa Y, Tachibana O, Hasegawa M, et al. Frequent gene amplification and overexpression of decoy receptor 3 in glioblastoma. Acta Neuropathol (Berl), 2005,109:294-8.
    10 Ohshima K, Haraoka S, Sμgihara M, et al. Amplification and expression of a decoy receptor for fas ligand (DcR3) in virus (EBV or HTLV-I) associated lymphomas. Cancer Lett, 2000,160:89-97.
    11 Connolly K, Cho YH, Duan R, et al. In vivo inhibition of Fas ligand-mediated killingby TR6, a Fas ligand decoy receptor. J Pharmacol Exp Ther, 2001,298:25-33.
    12 Migone TS, Zhang J, Luo X, et al. TL1A is a TNF-like ligand for DR3 and TR6/DcR3 and functions as a T cell costimμLator. Immunity, 2002,16:479-92.
    13 Hsu TL, Chang YC, Chen SJ, et al. ModμLation of dendritic cell differentiation and maturation by decoy receptor 3. J Immunol (Baltimore, Md: 1950), 2002,168:4846-53.
    14 Wan X, Shi G, Semenuk M, et al. DcR3/TR6 modμLates immune cell interactions. J Cell Biochem, 2003,89:603-12.
    15 Hsu TL, Wu YY, Chang YC, et al. Attenuation of Th1 response in decoy receptor 3 transgenic mice. J Immunol (Baltimore, Md: 1950), 2005,175:5135-45.
    16 Wu Y, Han B, Luo H, et al. DcR3/TR6 effectively prevents islet primary nonfunction after transplantation. Diabetes, 2003,52:2279-86.
    17 Macher-Goeppinger S, AμLmann S, Wagener N, et al. Decoy receptor 3 is a prognostic factor in renal cell cancer. Neoplasia, 2008,10:1049-56.
    18 Takahama Y, Yamada Y, Emoto K, et al. The prognostic significance of overexpression of the decoy receptor for Fas ligand (DcR3) in patients with gastric carcinomas. Gastric Cancer, 2002,5:61-8.
    19 Simon I, Liu Y, Krall KL, et al. Evaluation of the novel serum markers B7-H4, Spondin 2, and DcR3 for diagnosis and early detection of ovarian cancer. Gynecol Oncol, 2007,106:112-8.
    20 Connor JP, Felder M. Ascites from epithelial ovarian cancer contain high levels of functional decoy receptor 3 (DcR3) and is associated with platinum resistance. Gynecol Oncol, 2008,111:330-5.
    21 Chang PM, Chen PM, Hsieh SL, et al. Expression of a soluble decoy receptor 3 in patients with diffuse large B-cell lymphoma predicts clinical outcome. Int J Oncol, 2008,33:549-54.
    22 Landegren U, Nilsson M, Kwok PY. Reading bits of genetic information: methods for single-nucleotide polymorphism analysis. Genome Res, 1998,8:769-76.
    23 Bird AP. CpG-rich islands and the function of DNA methylation. Nature, 1986,321:209-13.
    24 Esteller M, Corn PG, Baylin SB, et al. A gene hypermethylation profile of human cancer. Cancer Res, 2001,61:3225-9.
    25 Esteller M. CpG island hypermethylation and tumor suppressor genes: a booming present, a brighter future. Oncogene, 2002,21:5427-40.
    26 Hannon GJ, Rossi JJ. Unlocking the potential of the human genome with RNA interference. Nature, 2004,431:371-8.
    27 Hutvagner G, Zamore PD. A microRNA in a mμLtiple-turnover RNAi enzyme complex. Science, 2002,297:2056-60.
    28 Sobin LH, Fleming ID. TNM Classification of Malignant Tumors, fifth edition (1997). Union Internationale Contre le Cancer and the American Joint Committee on Cancer. Cancer, 1997,80:1803-4.
    29 Ibrahim SM, Ringel J, Schmidt C, et al. Pancreatic adenocarcinoma cell lines show variable susceptibility to TRAIL-mediated cell death. Pancreas, 2001,23:72-9.
    30 Tsuji S, Hosotani R, Yonehara S, et al. Endogenous decoy receptor 3 blocks the growth inhibition signals mediated by Fas ligand in human pancreatic adenocarcinoma. Int J Cancer, 2003,106:17-25.
    31 Shen HW, Gao SL, Wu YL, et al. Overexpression of decoy receptor 3 in hepatocellμLar carcinoma and its association with resistance to Fas ligand-mediated apoptosis. World J Gastroenterol JT - World journal of gastroenterology : WJG, 2005,11:5926-30.
    32 Li H, Zhang L, Lou H, et al. Overexpression of decoy receptor 3 in precancerous lesions and adenocarcinoma of the esophagus. Am J Clin Pathol, 2005,124:282-7.
    33 Wu Y, Guo E, Yu J, et al. High DcR3 expression predicts stage pN2-3 in gastric cancer. Am J Clin Oncol, 2008,31:79-83.
    34 Schwendel A, Richard F, Langreck H, et al. Chromosome alterations in breast carcinomas: frequent involvement of DNA losses including chromosomes 4q and 21q. Br J Cancer, 1998,78:806-11.
    35 Sakakura C, Mori T, Sakabe T, et al. Gains, losses, and amplifications of genomic materials in primary gastric cancers analyzed by comparative genomic hybridization. Genes Chromosomes Cancer, 1999,24:299-305.
    36 MμLeris M, Almeida A, GerbaμLt-Seureau M, et al. Identification of amplified DNA sequences in breast cancer and their organization within homogeneously staining regions. Genes Chromosomes Cancer, 1995,14:155-63.
    37 Altura RA, Maris JM, Li H, et al. Novel regions of chromosomal loss in familial neuroblastoma by comparative genomic hybridization. Genes Chromosomes Cancer, 1997,19:176-84.
    38 Green DR. Apoptotic pathways: the roads to ruin. Cell, 1998,94:695-8.
    39 Rooney IA, Butrovich KD, Glass AA, et al. The lymphotoxin-beta receptor is necessary and sufficient for LIGHT-mediated apoptosis of tumor cells. J Biol Chem, 2000,275:14307-15.
    40 Shi G, Wu Y, Zhang J, et al. Death decoy receptor TR6/DcR3 inhibits T cell chemotaxis in vitro and in vivo. J Immunol (Baltimore, Md: 1950), 2003,171:3407-14.
    41 Wu Y, Han B, Sheng H, et al. Clinical significance of detecting elevated serum DcR3/TR6/M68 in malignant tumor patients. Int J Cancer, 2003,105:724-32.
    42蒋知俭.统计分析在医学课题中的应用.北京:人民卫生出版社,2008.
    43 Wang DG, Fan JB, Siao CJ, et al. Large-scale identification, mapping, and genotyping of single-nucleotide polymorphisms in the human genome. Science, 1998,280: 1077-82.
    44 Collins FS, Brooks LD, Chakravarti A. A DNA polymorphism discovery resource for research on human genetic variation. Genome Res, 1998,8:1229-31.
    45 Brookes AJ. The essence of SNPs. Gene, 1999,234:177-86.
    46 Lai E. Application of SNP technologies in medicine: lessons learned and future challenges. Genome Res, 2001,11:927-9.
    47 Vogelauer M, Wu J, Suka N, et al. Global histone acetylation and deacetylation in yeast. Nature, 2000,408:495-8.
    48房静远.表型遗传修饰与肿瘤.上海:上海科学技术出版社,2003.
    49 Baylin SB, Herman JG. DNA hypermethylation in tumorigenesis: epigenetics joins genetics. Trends Genet, 2000,16:168-74.
    50 Li Q, Ahuja N, Burger PC, et al. Methylation and silencing of the Thrombospondin-1 promoter in human cancer. Oncogene, 1999,18:3284-9.
    51 Herman JG, Graff JR, Myohanen S, et al. Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. PNAS, 1996,93:9821-6.
    52 Frommer M, McDonald LE, Millar DS, et al. A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands.PNAS, 1992,89:1827-31.
    53 Salmon P, Trono D. Production and titration of lentiviral vectors. Curr Protoc Hum Genet, 2007,Chapter 12:Unit 12.10.
    54 Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods, 2001,25:402-8.
    55薛庆善.体外培养的原理与技术.北京:科学出版社,2001.
    56 Fire A, Xu S, Montgomery MK, et al. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature, 1998,391:806-11.
    57 Elbashir SM, Harborth J, Lendeckel W, et al. Duplexes of 21-nucleotide RNAs mediate RNA interference in cμLtured mammalian cells. Nature, 2001,411:494-8.
    58 Silva J, Chang K, Hannon GJ, et al. RNA-interference-based functional genomics in mammalian cells: reverse genetics coming of age. Oncogene, 2004,23:8401-9.
    59 Lagos-Quintana M, Rauhut R, Yalcin A, et al. Identification of tissue-specific microRNAs from mouse. Curr Biol, 2002,12:735-9.
    60 Sempere LF, Freemantle S, Pitha-Rowe I, et al. Expression profiling of mammalian microRNAs uncovers a subset of brain-expressed microRNAs with possible roles in murine and human neuronal differentiation. Genome Biol, 2004,5:R13.
    61 Pasquinelli AE, Reinhart BJ, Slack F, et al. Conservation of the sequence and temporal expression of let-7 heterochronic regμLatory RNA. Nature, 2000,408:86-9.
    62 Lee Y, Ahn C, Han J, et al. The nuclear RNase III Drosha initiates microRNA processing. Nature, 2003,425:415-9.
    63 Sui G, Soohoo C, Affar el B, et al. A DNA vector-based RNAi technology to suppress gene expression in mammalian cells. PNAS, 2002,99:5515-20.
    64 Stegmeier F, Hu G, Rickles RJ, et al. A lentiviral microRNA-based system for single-copy polymerase II-regμLated RNA interference in mammalian cells. PNAS, 2005,102:13212-7.
    65 Zeng Y, Wagner EJ, CμLlen BR. Both natural and designed micro RNAs can inhibit the expression of cognate mRNAs when expressed in human cells. Mol Cell, 2002,9:1327-33.
    66 Naldini L, Blomer U, Gage FH, et al. Efficient transfer, integration, and sustained long-term expression of the transgene in adμLt rat brains injected with a lentiviralvector. PNAS, 1996,93:11382-8.
    67 Naldini L, Blomer U, Gallay P, et al. In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science, 1996,272:263-7.
    68 Miyoshi H, Takahashi M, Gage FH, et al. Stable and efficient gene transfer into the retina using an HIV-based lentiviral vector. PNAS, 1997,94:10319-23.
    69 Goldman MJ, Lee PS, Yang JS, et al. Lentiviral vectors for gene therapy of cystic fibrosis. Hum Gene Ther, 1997,8:2261-8.
    70熊刚,杨康,白云. LIGHT-Fc基因转染对食管癌细胞株Eca109抑制作用的初步研究.第三军医大学学报, 2004,26:686-689.
    71黄钢,白云,姜曼,等.人重组LIGHT-Fc分子的真核表达及其抗肿瘤活性研究.细胞与分子免疫学杂志, 2003,19:499-502.
    72 Yang CR, Hsieh SL, Teng CM, et al. Soluble decoy receptor 3 induces angiogenesis by neutralization of TL1A, a cytokine belonging to tumor necrosis factor superfamily and exhibiting angiostatic action. Cancer research, 2004,64:1122-9.
    1 Pitti RM, Marsters SA, Lawrence DA, et al. Genomic amplification of a decoy receptor for Fas ligand in lung and colon cancer. Nature, 1998,396:699-703.
    2 Yu KY, Kwon B, Ni J, et al. A newly identified member of tumor necrosis factor receptor superfamily (TR6) suppresses LIGHT-mediated apoptosis. J Biol Chem, 1999,274:13733-6.
    3 Bai C, Connolly B, Metzker ML, et al. Overexpression of M68/DcR3 in human gastrointestinal tract tumors independent of gene amplification and its location in a four-gene cluster. PNAS, 2000,97:1230-5.
    4 Schwendel A, Richard F, Langreck H, et al. Chromosome alterations in breast carcinomas: frequent involvement of DNA losses including chromosomes 4q and 21q. Br J Cancer, 1998,78:806-11.
    5 Sakakura C, Mori T, Sakabe T, et al. Gains, losses, and amplifications of genomic materials in primary gastric cancers analyzed by comparative genomic hybridization. Genes Chromosomes Cancer, 1999,24:299-305.
    6 Muleris M, Almeida A, Gerbault-Seureau M, et al. Identification of amplified DNA sequences in breast cancer and their organization within homogeneously staining regions. Genes Chromosomes Cancer, 1995,14:155-63.
    7 Altura RA, Maris JM, Li H, et al. Novel regions of chromosomal loss in familial neuroblastoma by comparative genomic hybridization. Genes Chromosomes Cancer, 1997,19:176-84.
    8 Otsuki T, Tomokuni A, Sakaguchi H, et al. Over-expression of the decoy receptor 3 (DcR3) gene in peripheral blood mononuclear cells (PBMC) derived from silicosis patients. Clin Exp Immunol, 2000,119:323-7.
    9 Ohshima K, Haraoka S, Sugihara M, et al. Amplification and expression of a decoy receptor for fas ligand (DcR3) in virus (EBV or HTLV-I) associated lymphomas. Cancer Lett, 2000,160:89-97.
    10 Tsuji S, Hosotani R, Yonehara S, et al. Endogenous decoy receptor 3 blocks the growth inhibition signals mediated by Fas ligand in human pancreatic adenocarcinoma.Int J Cancer, 2003,106:17-25.
    11 Ibrahim SM, Ringel J, Schmidt C, et al. Pancreatic adenocarcinoma cell lines show variable susceptibility to TRAIL-mediated cell death. Pancreas, 2001,23:72-9.
    12 Shen HW, Gao SL, Wu YL, et al. Overexpression of decoy receptor 3 in hepatocellular carcinoma and its association with resistance to Fas ligand-mediated apoptosis. World J Gastroenterol, 2005,11:5926-30.
    13 Roth W, Isenmann S, Nakamura M, et al. Soluble decoy receptor 3 is expressed by malignant gliomas and suppresses CD95 ligand-induced apoptosis and chemotaxis. Cancer Res, 2001,61:2759-65.
    14 Arakawa Y, Tachibana O, Hasegawa M, et al. Frequent gene amplification and overexpression of decoy receptor 3 in glioblastoma. Acta Neuropathol (Berl), 2005,109:294-8.
    15 Li H, Zhang L, Lou H, et al. Overexpression of decoy receptor 3 in precancerous lesions and adenocarcinoma of the esophagus. Am J Clin Pathol, 2005,124:282-7.
    16 Macher-Goeppinger S, Aulmann S, Wagener N, et al. Decoy receptor 3 is a prognostic factor in renal cell cancer. Neoplasia, 2008,10:1049-56.
    17 Wu Y, Guo E, Yu J, et al. High DcR3 expression predicts stage pN2-3 in gastric cancer. Am J Clin Oncol, 2008,31:79-83.
    18 Hayashi S, Miura Y, Nishiyama T, et al. Decoy receptor 3 expressed in rheumatoid synovial fibroblasts protects the cells against Fas-induced apoptosis. Arthritis Rheum, 2007,56:1067-75.
    19 Kim S, Fotiadu A, Kotoula V. Increased expression of soluble decoy receptor 3 in acutely inflamed intestinal epithelia. Clin Immunol, 2005,115:286-94.
    20 Bamias G, Siakavellas SI, Stamatelopoulos KS, et al. Circulating levels of TNF-like cytokine 1A (TL1A) and its decoy receptor 3 (DcR3) in rheumatoid arthritis. Clin Immunol, 2008,129:249-55.
    21 Suda T, Takahashi T, Golstein P, et al. Molecular cloning and expression of the Fas ligand, a novel member of the tumor necrosis factor family. Cell, 1993,75:1169-78.
    22 Takahashi T, Tanaka M, Inazawa J, et al. Human Fas ligand: gene structure, chromosomal location and species specificity. Int Immunol, 1994,6:1567-74.
    23 Tanaka M, Suda T, Haze K, et al. Fas ligand in human serum. Nat Med,1996,2:317-22.
    24 Mita E, Hayashi N, Iio S, et al. Role of Fas ligand in apoptosis induced by hepatitis C virus infection. Biochem Biophys Res Commun, 1994,204:468-74.
    25 Oshimi Y, Oda S, Honda Y, et al. Involvement of Fas ligand and Fas-mediated pathway in the cytotoxicity of human natural killer cells. J Immunol, 1996,157: 2909-15.
    26 D'Alessio A, Riccioli A, Lauretti P, et al. Testicular FasL is expressed by sperm cells. PNAS, 2001,98:3316-21.
    27 Hunt JS, Vassmer D, Ferguson TA, et al. Fas ligand is positioned in mouse uterus and placenta to prevent trafficking of activated leukocytes between the mother and the conceptus. J Immunol, 1997,158:4122-8.
    28 Griffith TS, Brunner T, Fletcher SM, et al. Fas ligand-induced apoptosis as a mechanism of immune privilege. Science, 1995,270:1189-92.
    29 Matsui K, Fine A, Zhu B, et al. Identification of two NF-kappa B sites in mouse CD95 ligand (Fas ligand) promoter: functional analysis in T cell hybridoma. J Immunol, 1998,161:3469-73.
    30 Green DR. Apoptotic pathways: the roads to ruin. Cell, 1998,94:695-8.
    31 Badley AD, McElhinny JA, Leibson PJ, et al. Upregulation of Fas ligand expression by human immunodeficiency virus in human macrophages mediates apoptosis of uninfected T lymphocytes. J Virol, 1996,70:199-206.
    32 Hahne M, Rimoldi D, Schroter M, et al. Melanoma cell expression of Fas(Apo-1/CD95) ligand: implications for tumor immune escape. Science, 1996,274: 1363-6.
    33 Lynch DH, Ramsdell F, Alderson MR. Fas and FasL in the homeostatic regulation of immune responses. Immunol Today, 1995,16:569-74.
    34 Arai H, Gordon D, Nabel EG, et al. Gene transfer of Fas ligand induces tumor regression in vivo. PNAS, 1997,94:13862-7.
    35 Seino K, Kayagaki N, Okumura K, et al. Antitumor effect of locally produced CD95 ligand. Nat Med, 1997,3:165-70.
    36 Chen JJ, Sun Y, Nabel GJ. Regulation of the proinflammatory effects of Fas ligand (CD95L). Science, 1998,282:1714-7.
    37 Schaub FJ, Han DK, Liles WC, et al. Fas/FADD-mediated activation of a specific program of inflammatory gene expression in vascular smooth muscle cells. Nat Med, 2000,6:790-6.
    38 Mauri DN, Ebner R, Montgomery RI, et al. LIGHT, a new member of the TNF superfamily, and lymphotoxin alpha are ligands for herpesvirus entry mediator. Immunity, 1998,8:21-30.
    39 Harrop JA, McDonnell PC, Brigham-Burke M, et al. Herpesvirus entry mediator ligand (HVEM-L), a novel ligand for HVEM/TR2, stimulates proliferation of T cells and inhibits HT29 cell growth. J Biol Chem, 1998,273:27548-56.
    40 Misawa K, Nosaka T, Kojima T, et al. Molecular cloning and characterization of a mouse homolog of human TNFSF14, a member of the TNF superfamily. Cytogenet Cell Genet, 2000,89:89-91.
    41 Rooney IA, Butrovich KD, Glass AA, et al. The lymphotoxin-beta receptor is necessary and sufficient for LIGHT-mediated apoptosis of tumor cells. J Biol Chem, 2000,275:14307-15.
    42 Granger SW, Butrovich KD, Houshmand P, et al. Genomic characterization of LIGHT reveals linkage to an immune response locus on chromosome 19p13.3 and distinct isoforms generated by alternate splicing or proteolysis. J Immunol, 2001,167:5122-8.
    43 Morel Y, Schiano de Colella JM, Harrop J, et al. Reciprocal expression of the TNF family receptor herpes virus entry mediator and its ligand LIGHT on activated T cells: LIGHT down-regulates its own receptor. J Immunol, 2000,165:4397-404.
    44 Castellano R, Van Lint C, Peri V, et al. Mechanisms regulating expression of the tumor necrosis factor-related light gene. Role of calcium-signaling pathway in the transcriptional control. J Biol Chem, 2002,277:42841-51.
    45 Tamada K, Shimozaki K, Chapoval AI, et al. LIGHT, a TNF-like molecule, costimulates T cell proliferation and is required for dendritic cell-mediated allogeneic T cell response. J Immunol, 2000,164:4105-10.
    46 Lu G, Janjic BM, Janjic J, et al. Innate direct anticancer effector function of human immature dendritic cells. II. Role of TNF, lymphotoxin-alpha(1)beta(2), Fas ligand, and TNF-related apoptosis-inducing ligand. J Immunol, 2002,168:1831-9.
    47 Janjic BM, Lu G, Pimenov A, et al. Innate direct anticancer effector function of humanimmature dendritic cells. I. Involvement of an apoptosis-inducing pathway. J Immunol, 2002,168:1823-30.
    48 Tamada K, Shimozaki K, Chapoval AI, et al. Modulation of T-cell-mediated immunity in tumor and graft-versus-host disease models through the LIGHT co-stimulatory pathway. Nat Med, 2000,6:283-9.
    49 Chen MC, Hsu TL, Luh TY, et al. Overexpression of bcl-2 enhances LIGHT- and interferon-gamma -mediated apoptosis in Hep3BT2 cells. J Biol Chem, 2000,275: 38794-801.
    50 Wang J, Chun T, Lo JC, et al. The critical role of LIGHT, a TNF family member, in T cell development. J Immunol, 2001,167:5099-105.
    51 Wang J, Foster A, Chin R, et al. The complementation of lymphotoxin deficiency with LIGHT, a newly discovered TNF family member, for the restoration of secondary lymphoid structure and function. Eur J Immunol, 2002,32:1969-79.
    52 Tan KB, Harrop J, Reddy M, et al. Characterization of a novel TNF-like ligand and recently described TNF ligand and TNF receptor superfamily genes and their constitutive and inducible expression in hematopoietic and non-hematopoietic cells. Gene, 1997,204:35-46.
    53 Chew LJ, Pan H, Yu J, et al. A novel secreted splice variant of vascular endothelial cell growth inhibitor. FASEB J, 2002,16:742-4.
    54 Migone TS, Zhang J, Luo X, et al. TL1A is a TNF-like ligand for DR3 and TR6/DcR3 and functions as a T cell costimulator. Immunity, 2002,16:479-92.
    55 Bamias G, Martin C 3rd, Marini M, et al. Expression, localization, and functional activity of TL1A, a novel Th1-polarizing cytokine in inflammatory bowel disease. J Immunol, 2003,171:4868-74.
    56 Cassatella MA, Pereira-da-Silva G, Tinazzi I, et al. Soluble TNF-like cytokine (TL1A) production by immune complexes stimulated monocytes in rheumatoid arthritis. J Immunol, 2007,178:7325-33.
    57 Papadakis KA, Zhu D, Prehn JL, et al. Dominant role for TL1A/DR3 pathway in IL-12 plus IL-18-induced IFN-gamma production by peripheral blood and mucosal CCR9+ T lymphocytes. J Immunol, 2005,174:4985-90.
    58 Prehn JL, Mehdizadeh S, Landers CJ, et al. Potential role for TL1A, the newTNF-family member and potent costimulator of IFN-gamma, in mucosal inflammation. Clin Immunol, 2004,112:66-77.
    59 Papadakis KA, Prehn JL, Landers C, et al. TL1A synergizes with IL-12 and IL-18 to enhance IFN-gamma production in human T cells and NK cells. J Immunol, 2004,172:7002-7.
    60 Wen L, Zhuang L, Luo X, et al. TL1A-induced NF-kappaB activation and c-IAP2 production prevent DR3-mediated apoptosis in TF-1 cells. J Biol Chem, 2003,278:39251-8.
    61 Bamias G, Mishina M, Nyce M, et al. Role of TL1A and its receptor DR3 in two models of chronic murine ileitis. PNAS, 2006,103:8441-6.
    62 Takedatsu H, Michelsen KS, Wei B, et al. TL1A (TNFSF15) regulates the development of chronic colitis by modulating both T-helper 1 and T-helper 17 activation. Gastroenterology, 2008,135:552-67.
    63 Kang YJ, Kim WJ, Bae HU, et al. Involvement of TL1A and DR3 in induction of pro-inflammatory cytokines and matrix metalloproteinase-9 in atherogenesis. Cytokine, 2005,29:229-35.
    64 Pinti M, Troiano L, Nasi M, et al. Genetic polymorphisms of Fas (CD95) and FasL (CD178) in human longevity: studies on centenarians. Cell Death Differ, 2002,9:431-8.
    65 Connolly K, Cho YH, Duan R, et al. In vivo inhibition of Fas ligand-mediated killing by TR6, a Fas ligand decoy receptor. J Pharmacol Exp Ther, 2001,298:25-33.
    66 Ju ST, Panka DJ, Cui H, et al. Fas(CD95)/FasL interactions required for programmed cell death after T-cell activation. Nature, 1995,373:444-8.
    67 Scheu S, Alferink J, Potzel T, et al. Targeted disruption of LIGHT causes defects in costimulatory T cell activation and reveals cooperation with lymphotoxin beta in mesenteric lymph node genesis. J Exp Med, 2002,195:1613-24.
    68 Wang J, Lo JC, Foster A, et al. The regulation of T cell homeostasis and autoimmunity by T cell-derived LIGHT. J Clin Invest, 2001,108:1771-80.
    69 Yu KY, Kwon B, Ni J, et al. A newly identified member of tumor necrosis factor receptor superfamily (TR6) suppresses LIGHT-mediated apoptosis. J Biol Chem, 1999,274:13733-6.
    70 Gill RM, Coleman NM, Hunt JS. Differential cellular expression of LIGHT and its receptors in early gestation human placentas. J Reprod Immunol, 2007,74:1-6.
    71 Gill RM, Ni J, Hunt JS. Differential expression of LIGHT and its receptors in human placental villi and amniochorion membranes. Am J Pathol, 2002,161:2011-7.
    72 Gill RM, Hunt JS. Soluble receptor (DcR3) and cellular inhibitor of apoptosis-2 (cIAP-2) protect human cytotrophoblast cells against LIGHT-mediated apoptosis. Am J Pathol, 2004,165:309-17.
    73 Zhang J, Salcedo TW, Wan X, et al. Modulation of T-cell responses to alloantigens by TR6/DcR3. J Clin Invest, 2001,107:1459-68.
    74 Wan X, Zhang J, Luo H, et al. A TNF family member LIGHT transduces costimulatory signals into human T cells. J Immunol, 2002,169:6813-21.
    75 Shi G, Luo H, Wan X, et al. Mouse T cells receive costimulatory signals from LIGHT, a TNF family member. Blood, 2002,100:3279-86.
    76 Yang CR, Hsieh SL, Teng CM, et al. Soluble decoy receptor 3 induces angiogenesis by neutralization of TL1A, a cytokine belonging to tumor necrosis factor superfamily and exhibiting angiostatic action. Cancer Res, 2004,64:1122-9.
    77 Hsu TL, Chang YC, Chen SJ, et al. Modulation of dendritic cell differentiation and maturation by decoy receptor 3. J Immunol, 2002,168:4846-53.
    78 Hsu TL, Wu YY, Chang YC, et al. Attenuation of Th1 response in decoy receptor 3 transgenic mice. J Immunol, 2005,175:5135-45.
    79 Wan X, Shi G, Semenuk M, et al. DcR3/TR6 modulates immune cell interactions. J Cell Biochem, 2003,89:603-12.
    80 Shi G, Wu Y, Zhang J, et al. Death decoy receptor TR6/DcR3 inhibits T cell chemotaxis in vitro and in vivo. J Immunol, 2003,171:3407-14.
    81 Chang YC, Chan YH, Jackson DG, et al. The glycosaminoglycan-binding domain of decoy receptor 3 is essential for induction of monocyte adhesion. J Immunol, 2006,176:173-80.
    82 Yang CR, Hsieh SL, Ho FM, et al. Decoy receptor 3 increases monocyte adhesion to endothelial cells via NF-kappa B-dependent up-regulation of intercellular adhesion molecule-1, VCAM-1, and IL-8 expression. J Immunol, 2005,174:1647-56.
    83 Hsu MJ, Lin WW, Tsao WC, et al. Enhanced adhesion of monocytes via reversesignaling triggered by decoy receptor 3. Exp Cell Res, 2004,292:241-51.
    84 Chang YC, Hsu TL, Lin HH, et al. Modulation of macrophage differentiation and activation by decoy receptor 3. J Leukoc Biol, 2004,75:486-94.
    85 Robinson LJ, Borysenko CW, Blair HC. Tumor necrosis factor family receptors regulating bone turnover: new observations in osteoblastic and osteoclastic cell lines. Ann N Y Acad Sci, 2007,1116:432-43.
    86 Yang CR, Wang JH, Hsieh SL, et al. Decoy receptor 3 (DcR3) induces osteoclast formation from monocyte/macrophage lineage precursor cells. Cell Death Differ, 2004,11 Suppl 1:S97-107.
    87 Tang CH, Hsu TL, Lin WW, et al. Attenuation of bone mass and increase of osteoclast formation in decoy receptor 3 transgenic mice. J Biol Chem, 2007,282:2346-54.
    88 Edwards JR, Sun SG, Locklin R, et al. LIGHT (TNFSF14), a novel mediator of bone resorption, is elevated in rheumatoid arthritis. Arthritis Rheum, 2006,54:1451-62.
    89 Wu YY, Chang YC, Hsu TL, et al. Sensitization of cells to TRAIL-induced apoptosis by decoy receptor 3. J Biol Chem, 2004,279:44211-8.
    90 Wu Y, Han B, Sheng H, et al. Clinical significance of detecting elevated serum DcR3/TR6/M68 in malignant tumor patients. Int J Cancer, 2003,105:724-32.
    91 Takahama Y, Yamada Y, Emoto K, et al. The prognostic significance of overexpression of the decoy receptor for Fas ligand (DcR3) in patients with gastric carcinomas. Gastric Cancer, 2002,5:61-8.
    92 Simon I, Liu Y, Krall KL, et al. Evaluation of the novel serum markers B7-H4, Spondin 2, and DcR3 for diagnosis and early detection of ovarian cancer. Gynecol Oncol, 2007,106:112-8.
    93 Connor JP, Felder M. Ascites from epithelial ovarian cancer contain high levels of functional decoy receptor 3 (DcR3) and is associated with platinum resistance. Gynecol Oncol, 2008,111:330-5.
    94 Chang PM, Chen PM, Hsieh SL, et al. Expression of a soluble decoy receptor 3 in patients with diffuse large B-cell lymphoma predicts clinical outcome. Int J Oncol, 2008,33:549-54.
    95 Mild G, Bachmann F, Boulay JL, et al. DCR3 locus is a predictive marker for
    5-fluorouracil-based adjuvant chemotherapy in colorectal cancer. Int J Cancer,2002,102:254-7.
    96 Chen J, Zhang L, Kim S. Quantification and detection of DcR3, a decoy receptor in TNFR family. J Immunol Methods, 2004,285:63-70.
    97段伟宏,苏忠学,吴泰璜. DcR3反义RNA与顺铂对肝癌细胞凋亡影响的比较.中国现代普通外科进展, 2005,8:90-92.
    98段伟宏,苏忠学,吴泰璜.转染反义DcR3真核表达载体协同氟尿嘧啶诱导肝癌细胞凋亡.中华肝胆外科杂志, 2006,12:98-100.
    99吴泰璜,段伟宏,苏忠学,等.反义DcR3 RNA转染诱导肝癌细胞凋亡的作用.中华微生物学和免疫学杂志, 2005,25:799-802.
    100 Shi G, Mao J, Yu G, et al. Tumor vaccine based on cell surface expression of DcR3/TR6. J Immunol, 2005,174:4727-35.
    101 Wu SF, Liu TM, Lin YC, et al. Immunomodulatory effect of decoy receptor 3 on the differentiation and function of bone marrow-derived dendritic cells in nonobese diabetic mice: from regulatory mechanism to clinical implication. J Leukoc Biol, 2004,75:293-306.
    102 Sung HH, Juang JH, Lin YC, et al. Transgenic expression of decoy receptor 3 protects islets from spontaneous and chemical-induced autoimmune destruction in nonobese diabetic mice. J Exp Med, 2004,199:1143-51.
    103 Ka SM, Sytwu HK, Chang DM, et al. Decoy receptor 3 ameliorates an autoimmune crescentic glomerulonephritis model in mice. J Am Soc Nephrol, 2007,18:2473-85.
    104 Lee CS, Hu CY, Tsai HF, et al. Elevated serum decoy receptor 3 with enhanced T cell activation in systemic lupus erythematosus. Clin Exp Immunol, 2008,151:383-90.
    105 Han B, Bojalil R, Amezcua-Guerra LM, et al. DcR3 as a diagnostic parameter and risk factor for systemic lupus erythematosus. Int Immunol, 2008,20:1067-75.
    106 Wroblewski VJ, Witcher DR, Becker GW, et al. Decoy receptor 3 (DcR3) is proteolytically processed to a metabolic fragment having differential activities against Fas ligand and LIGHT. Biochem Pharmacol, 2003,65:657-67.
    107 Kitamura Y, Hashimoto S, Mizuta N, et al. Fas/FasL-dependent apoptosis of alveolar cells after lipopolysaccharide-induced lung injury in mice. Am J Respir Crit Care Med, 2001,163:762-9.
    108 Mizuta M, Nakajima H, Mizuta N, et al. Fas ligand released by activated monocytescauses apoptosis of lung epithelial cells in human acute lung injury model in vitro. Biol Pharm Bull, 2008,31:386-90.
    109 Wortinger MA, Foley JW, Larocque P, et al. Fas ligand-induced murine pulmonary inflammation is reduced by a stable decoy receptor 3 analogue. Immunology, 2003,110:225-33.
    110 Martin TR, Hagimoto N, Nakamura M, Matute-Bello G. Apoptosis and Epithelial Injury in the Lungs. Proc Am Thorac Soc, 2005,2:214-220.
    111 Matute-Bello G, Liles WC, Frevert CW, et al. Blockade of the Fas/FasL system improves pneumococcal clearance from the lungs without preventing dissemination of bacteria to the spleen. J Infect Dis, 2005,191:596-606.
    112 Wu Y, Han B, Luo H, et al. DcR3/TR6 effectively prevents islet primary nonfunction after transplantation. Diabetes, 2003,52:2279-86.
    113王槐志,黎万玲,张玉君,等.诱骗受体3对大鼠移植肝脏的保护作用.中华消化外科杂志, 2007,6:356-359.

© 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