TGF-β不敏感的树突状细胞疫苗对前列腺癌的免疫治疗作用研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的:树突状细胞(DC)是目前在肿瘤免疫中功能最强的一种抗原递呈细胞,以其为基础制成的肿瘤疫苗,可多途径诱导机体产生特异性抗肿瘤免疫。但是肿瘤细胞分泌大量的转化生长因子(TGF-β)抑制DC递呈抗原和促进DC凋亡。我们采用前列腺癌TRAMP-C2细胞裂解产物作为抗原加载C57BL/6小鼠骨髓DC,诱导出前列腺癌特异性的DC。修饰TβRII基因,构建逆转录病毒载体,体外基因转染前列腺癌特异性的DC,检测转染后前列腺癌特异性DC的表面抗原、细胞增殖性、分泌功能变化,评价TGF-β不敏感的DC疫苗对TRAMP-C2前列腺癌荷瘤C57BL/6小鼠的免疫治疗作用。
     方法:1)构建含有显性负相TGF-βII型受体(TβRIIDN)质粒的逆转录病毒:含有TβRIIDN质粒的逆转录病毒转染293包装细胞,32℃下共转染12小时,10% DMEM培养基37℃下孵化过夜,PBS漂洗后同样条件再次转染293细胞24小时,收集上清,获得含有TβRIIDN重组逆转录病毒。
     2)负载前列腺癌抗原的DC细胞分离、培养及鉴定:将C57BL/6小鼠新鲜骨髓用红细胞去除法获取DC细胞,加入含rhIL-4(1000U/ml)、GM-CSF(1000U/ml)的完全培养基,隔日半量换液,在第6d梯度离心收获非粘附细胞为非成熟的DC细胞。反复冻融获得TRAMP-C2细胞裂解物,裂解物反复刺激非成熟的DC细胞获得负载前列腺癌抗原的成熟DC细胞。于光镜下形态学鉴定,用流式细胞仪检测细胞表型MHC class II,CD40,CD11c,CD80和CD86。
     3)DC细胞转染,获取TβRIIDN-DC细胞:含有TβRIIDN重组逆转录病毒转染负载前列腺癌抗原的DC细胞5%CO2、37℃下孵化48小时,获得表达TβRIIDN的DC细胞,流式细胞仪检测DC细胞表型变化。
     4)TβRIIDN-DC细胞体外实验:不同的DC细胞与TGF-β共同培养,流式细胞仪测定表面共刺激分子(CD80/CD86)变化,脱氧胸腺嘧啶苷核素掺入法进行增殖抑制实验,Western blot检测SMAD-2和磷酸化SMAD-2的表达。
     5)DC疫苗体内抗肿瘤评价:建立前列腺癌TRAMP-C2细胞皮下荷瘤C57BL/6小鼠模型,皮下荷瘤小鼠40只,随机分为4组,每组10只,分别皮下注射TβRIIDN-DC细胞、负载前列腺癌抗原的DC细胞、GFP-DC细胞和磷酸盐缓冲液(PBS),15天后重复注射,酶联免疫吸附(ELISA)测定细胞因子IFN-γ和IL-12变化,观察各组荷瘤小鼠的生存期和肿瘤的体积改变。
     6)体内CTL活性检测:最后一次疫苗免疫后5 d取其脾脏,分离、活化T淋巴细胞,Cr51释放实验测定肿瘤特异性杀伤T细胞(CTL)对TRAMP-C2细胞的细胞毒作用。黑色素瘤细胞B16-F1作为对照靶细胞。
     结果:
     1)流式细胞仪分析CD11c表达,小鼠骨髓来源的DC细胞纯度为90.8%。含有TβRIIDN和GFP的逆转录病毒转染效率分别为92.7%和90.6%。
     2)流式细胞仪检测不成熟DC细胞、负载前列腺癌抗原的成熟DC细胞、TβRIIDN-DC细胞、GFP-DC细胞的CD86、CD80、CD40、CD11c及MHC-II分子表型改变,成熟DC细胞、TβRIIDN-DC细胞、GFP-DC细胞表型表达明显高于不成熟DC细胞(P<0.01),成熟DC细胞、TβRIIDN-DC细胞、GFP-DC细胞之间细胞表型无明显差别(P>0.05)。说明表达TβRIIDN不影响DC细胞的分子表型。
     3)负载前列腺癌抗原的成熟DC细胞、TβRIIDN-DC细胞、GFP-DC细胞与5 ng/ml的TGF-β孵育后,Western blot在各组中均可以检测到Smad-2, TβRIIDN-DC细胞中却没有检测到磷酸化的Smad-2。表明TβRII显性负相表达阻断了DC细胞的TGF-β信号通路。
     4)与TGF-β共同培养72小时后,非转染的成熟DC细胞、GFP-DC细胞和TβRIIDN-DC细胞的脱氧胸腺嘧啶苷核素摄入抑制率分别为62.5%、65.5%和15%,TβRIIDN-DC细胞的脱氧胸腺嘧啶苷核素摄入抑制率与其他各组比较差异有显著统计学意义(P<0.05)。在给予TGF-β的正常条件下,非转染的成熟DC细胞、GFP-DC细胞不能增殖并且15天之内死亡,然而,TβRIIDN-DC细胞继续正常增殖和生长,表明TβRIIDN-DC细胞对TGF-β的抗增殖作用不敏感。
     5)非转染的成熟DC细胞、GFP-DC细胞和TβRIIDN-DC细胞与TGF-β及肿瘤抗原共同培养7天,流式细胞仪测定表面共刺激分子(CD80/CD86),在TGF-β作用下CD80和CD86在TβRIIDN-DC细胞的表达明显高于其他各组(P<0.01)。
     6)成功地构建了前列腺癌TRAMP-C2细胞皮下荷瘤C57BL/6小鼠模型,接种非转染的DC细胞、GFP-DC细胞和TβRIIDN-DC细胞的荷瘤小鼠与对照组相比能明显抑制肿瘤生长(P<0.01,P<0.05 and P<0.05,vs.对照组),其中TβRIIDN-DC组抑制效果更明显,有2例肿瘤完全消失。这表明TβRIIDN-DC疫苗有更强的抗肿瘤效果。50天后接种PBS、非转染的DC细胞、GFP-DC细胞和TβRIIDN-DC细胞的荷瘤小鼠生存率分别为0%、20%、30%和80%。Mantel-Haenszel log-rank统计分析表明:TβRIIDN-DC细胞与其他2组相比差别有统计学意义(P<0.01)。这些结果说明TGF-β不敏感的DC细胞能有效提高TRAMP-C2皮下荷瘤小鼠的生存率。
     7)接种PBS的荷瘤小鼠体内测出IFN-γ和IL-12基础水平,接种非转染的DC细胞、GFP-DC细胞的荷瘤小鼠体内IFN-γ和IL-12水平明显升高,接种TβRIIDN-DC细胞的荷瘤小鼠体内该两种细胞因子水平升高更明显。
     8)标准的Cr51释放实验测定CTL杀伤活性,非转染的DC细胞、GFP-DC细胞和TβRIIDN-DC细胞处理的荷瘤小鼠CTL对TRAMP-C2细胞有较高的杀伤活性,其中,TGF-β不敏感的DC细胞诱导出最强的CTL杀伤活性(E/T比率为100:1时杀伤活性为85%),对无关的黑色素瘤B16-F1细胞没有杀伤作用。结果表明阻断的TGF-β信号通路能增加肿瘤特异性杀伤活性,提高DC疫苗的效能。
     结论:
     1)成功构建了含有TβRIIDN质粒的逆转录病毒;
     2)采用前列腺癌TRAMP-C2细胞裂解产物作为抗原加载C57BL/6小鼠骨髓DC,诱导出了前列腺癌特异性的DC;
     3)首次使用修饰后TβRII基因转染前列腺癌特异性的DC,使TβRII显性负相表达,阻断TGF-β信号通路;
     4)TβRIIDN并不影响转染DC细胞的表型,在TGF-β作用下TβRIIDN-DC表面共刺激分子表达增高;
     5)TGF-β不敏感的DC细胞能明显提高TRAMP-C2皮下荷瘤小鼠的生存率,明显抑制肿瘤生长,而且有2例肿瘤完全消失。TGF-β不敏感的DC细胞处理的荷瘤小鼠体内IFN-γ和IL-12水平明显升高;
     6)TGF-β不敏感的DC细胞诱导出最强的CTL杀伤活性,E/T比率为100:1时杀伤活性为85%,CTL具有肿瘤特异性,对无关的黑色素瘤B16-F1细胞没有杀伤作用。
Objective
     DCs are highly potent initiators of the immune response, characterized by their ability to engulf, process, and present antigens to T lymphocytes. In recent years, DC-based anti-tumor vaccines have emerged as promising strategies for cancer immunotherapy. But high levels of TGF-βproduced by cancer cells inhibit the ability of DCs to present antigen, stimulate tumor-sensitized T lymphocytes. C57BL/6 murine bone marrow DCs were pulsed with freeze–thawed TRAMP-C2 tumor lysate and induced prostate cancer specific DCs. We successfully constructed a retrovirus containing dominant-negative TGF-βtype II receptor (TβRIIDN). The tumor lysate-pulsed DCs were rendered TGF-βinsensitive by infecting with a retrovirus containing TβRIIDN, leading to the blockade of TGF-βsignals to members of the Smad family. After transfection we detected surface antigen, cell proliferation, secretion and evaluated the immunotherapy of TGF-β insensitive DC vaccines to TRAMP-C2 tumor-bearing C57BL/6 mice. Materials and methods
     1. To construct a retrovirus containing dominant-negative TGF-βtype II receptor (TβRIIDN): Pantropic GP293 retroviral packaging cells were seeded at a density of 2.5×106 cells in T-25 collagen I-coated flasks 24 h before plasmid transfection in antibiotic-free 10% Dulbecco’s Modified Eagle Medium. A mixture of 2μg retroviral plasmid and 2μg vesicular stomatitis virus envelope G protein (VSV-G) envelope plasmid was cotransfected in serum-free DMEM using LipofectAMINE-Plus, according to the manufacturer’s protocols. Briefly, cells were transfected for 12 h followed by the addition of an equivalent volume of 10%DMEM and incubation for an additional 12 h. Afterward, the supernatant was aspirated, the cells were rinsed gently in PBS, and 3 ml of fresh 10%DMEM was added each flask. 24 h later, virus-containing supernatant was collected and used to infect target cells.
     2. Isolation, cultivation and identification of tumor lysate-pulsed DC: Erythrocyte-depleted murine bone marrow cells were obtained from the femurs and tibiae of C57BL/6 mice under aseptic conditions and cultured at 1×106 cells/ml in CM supplemented with 1000 U/ml recombinant murine granulocyte-macrophage-colony-stimulating factor (rmGM-CSF) and 1000 U/ml recombinant murine interleukin-4 (rmIL-4). The medium was replaced on day 2 with additional recombinant cytokines. On day 6, nonadherent DCs were harvested by gradient centrifugation and were further purified with MACS CD11c beads. Immature DCs were pulsed with freeze–thawed tumor lysate to obtain tumor lysate-pulsed DC. Tumor lysate-pulsed DCs were identified under light microscope. Immatured DCs, tumor lysate-pulsed DCs, GFP-transduced DCs and tumor lysate-pulsed TGF-β-insensitive DCs, which were cultured for an additional 18 h, were analyzed by flow cytometry, using a panel of Abs specific for MHC class II, CD40, CD11c, CD80, and CD86.
     3. Infection of DCs with Retrovirus: Tumor lysate-pulsed DCs were infected with the retrovirus containing TβRIIDN or GFP vector. The cells were rinsed gently in 5%CO2, 37℃for 48 hours to get TβRIIDN DCs. The immunophenotype of TβRIIDN DCs were analyzed by flow cytometry.
     4. Experiment of TβRIIDN DCs in vitro: Different DCs were cultured with TGF-β. The surface co-stimulatory molecules (CD80/CD86) were analyzed by flow cytometry. The antiproliferative effects of TGF-βwere observed by thymidine incorporation assay. Smad-2 and phosphorylated Smad-2 were detected by Western blot.
     5. Antitumor Analyses in Vivo: TRAMP-C2 tumors were established in mice. 40 C57BL/6 tumor-bearing mice were divided into four groups randomly and inoculated s.c. with nontransduced DCs, tumor lysate-pulsed TGF-β-insensitive DCs, GFP-transduced DCs, or PBS. The vaccination was repeated on day 15. Serum levels of IFN-γand IL-12 were determined by enzyme-linked immunoabsorbant assay (ELISA). Tumor growth and mouse survival were monitored daily post-inoculation.
     6. Cytotoxicity T Lymphocyte Assays: Splenic cells were obtained from the tumor-bearing mice 5 days after the final vaccination and cocultured with X-ray (40 Gy)-irradiated TRAMP-C2 cells (2×105) in 24-well plates for 4 days. The activated T-cells were harvested and used as effector cells against 51Cr-loaded TRAMP-C2 target cells. An irrelevant cancer cell line, mouse melanoma cell line, B16-F1 was used as a nonspecific control.
     Results:
     1. Erythrocyte-depleted murine bone marrow cells were freshly isolated from the femurs and tibiae of C57BL/6 mice. The purity of bone marrow–derived DC, determined through analysis on CD11c staining by flow cytometry, was 90.8%. Tumor lysate-pulsed DCs were infected with the TβRIIDN-containing or GFP control retrovirus. The infection efficiency was determined by flow cytometry analysis. They were 92.7% and 90.6%, respectively, for the T?RIIDN- containing and the GFP control retroviruses.
     2. DC surface molecules MHC class II, CD40, CD11c, CD80 and CD86, were analyzed and compared among immatured DCs, tumor lysate-pulsed DCs, GFP-transduced DCs and TβRIIDN-transduced DCs by flow cytometry. The expression of CD86, CD80, MHC class II and CD40 on tumor lysate-pulsed DCs, GFP-transduced DCs and TβRIIDN-transduced DCs was higher than those on immatured DCs (P<0.01). No obvious differences were observed among tumor lysate-pulsed DCs, GFP-transduced DCs and TβRIIDN-transduced DCs (P>0.05), indicating that the transduction of TβRIIDN did not affect the immunophenotype of DCs.
     3. Smad-2 and phosphorylated Smad-2 were detected by Western blot analysis after the TβRIIDN-transduced or GFP-transduced DCs were treated with 5 ng/ml TGF-β1. The presence of Smad-2 was detected in all DC groups. But, phosphorylated Smad-2 was only detected in nontransduced and GFP-transduced DCs in response to TGF-β1; absence of phosphorylated Smad-2 in TβRIIDN- transduced DCs confirmed that TGF-βsignal transduction was blocked by the presence of the TβRIIDN.
     4. The inhibitory rate of TGF-βon thymidine uptake was compared among the TβRIIDN-transduced DCs, GFP-transduced, and nontransduced DCs after the addition of TGF-β1 for 72 h. TGF-β1 showed a dramatic antiproliferative effect on the established nontransduced and GFP-transduced DCs, inhibiting uptake by a mean of 62.5% and 65.5% respectively. Whereas the mean inhibitory rate of thymidine uptake by TβRIIDN-transduced DCs was 15%, the resistance to the antiproliferative effects of TβRIIDN-transduced DCs was statistically significant when compared with the other groups (P<0.05). Importantly, when cells were maintained under normal growth conditions in the presence of TGF-β1, the nontransduced and GFP-transduced DCs failed to proliferate and died within 15 days. TβRIIDN-transduced DCs, however, continued to proliferate and grow normally, showing significant resistance to the antiproliferative effects of TGF-β1.
     5. Nontransduced DCs, GFP-transduced DCs and TβRIIDN- transduced DCs were cultured with freeze–thawed tumor lysate and 10 ng/mL TGF-β1 for 7 days. The surface co-stimulatory molecules CD80 and CD86, were analyzed by flow cytometry. As expected, expression of CD86 and CD80 was higher on TβRIIDN-transduced DCs than on nontransduced DCs and GFP-transduced DCs (P<0.01) in the presence of TGF-β1.
     6. To assess the antitumor effect of the TβRIIDN-transduced DC vaccine in vivo, TRAMP-C2 tumors were established in C57BL/6 mice. A suspension of nontransduced DCs, GFP-transduced DCs, or TβRIIDN-transduced DCs was injected into tumor-bearing mice (n = 10/group). PBS was used as a negative control. These experimental groups were designed to evaluate whether blocking TGF-βsignaling alters the efficacy of DC vaccine in inducing anti-tumor immune responses and mortality of the tumor-bearing mice. Immunization with TβRIIDN-transduced DCs, GFP-transduced DCs or nontransduced DCs significantly suppressed the growth of the tumor (P<0.01, P<0.05 and P<0.05, vs. control, respectively), with the TβRIIDN-transduced DCs showing the more significant inhibitory effect. Complete tumor regression occurred in 20% of TRAMP-C2-tumor-bearing mice that were treated with TβRIIDN-transduced DCs. These results indicated that the tumor lysate-pulsed TGF-β-insensitive DC vaccine had the strongest anti-tumor effect in all the vaccination groups. Another four groups of ten mice were used to evaluate the survival rate after 50 days. Results showed that survival rate of the untreated, GFP-vector control and nontransduced DCs treated mice was 0%, 20%and 30% respectively, while the survival of the TβRIIDN-transduced DCs treated cohort was 80%. Statistical analysis by using the Mantel-Haenszel log-rank test indicated a significant difference between the TGF-β-insensitive-DC and the other two control groups (P<0.01). The result demonstrate that the TGF-β-insensitive DC vaccine was effective in improving the survival rate in mice bearing TGF-β-secreting tumors.
     7. In animals injected with PBS, there was a basal level of IFN-γand IL-12. In animals received nontransduced DCs or GFP-transduced DCs, there was a significant increase in levels of both cytokines. A further increase in serum IL-12 and IFN-γwas observed when these cells were rendered insensitive to TGF-? (the T?RIIDN-transduced DCs group), suggesting the increase of activated immune cells in these hosts.
     8. The ability of CTLs to lyse TRAMP-C2 cells was assayed in vitro using a standard 51Cr release assay. Compared with the PBS-vaccinated group, mice treated with TβRIIDN-transduced DCs, GFP-transduced DCs, or non-transduced DCs all showed a higher cytotoxicity against the TRAMP-C2 cells. The most potent TRAMP-C2-specific splenic CTL response was induced by the TGF-?-insensitive DCs in the tumor bearer (85% killing activity at an effector:target cell ratio of 100:1). No apparent lysis was observed against irrelevant B16-F1 cells. This result indicates that tumor-specific cytolysis is generated by blocking TGF-βsignaling, which enhances the efficacy of DC vaccines.
     Conclusion
     1. 1. We successfully constructed a retrovirus containing dominant-negative TGF-βtype II receptor (TβRIIDN).
     2. C57BL/6 murine bone marrow DCs were pulsed with freeze–thawed TRAMP-C2 tumor lysate and induced prostate cancer specific DCs.
     3. The tumor lysate-pulsed DCs were rendered TGF-βinsensitive by infecting with a retrovirus containing dominant-negative TGF-βtype II receptor (TβRIIDN), leading to the blockade of TGF-βsignals to members of the Smad family.
     4. Expression of TβRIIDN did not affect the phenotype of transduced DCs. Expression of CD86 and CD80 was higher on TβRIIDN-transduced DCs than on nontransduced DCs and GFP-transduced DCs (P<0.01) in the presence of TGF-β1.
     5. TβRIIDN-transduced DCs suppressed tumor growth and increased survival rate of TRAMP-C2 tumor-bearing mice. Furthermore, complete tumor regression occurred in 2 vaccinated mice. TβRIIDN-transduced DCs induced higher IFN-γand IL-12 level in vivo.
     6. The most potent TRAMP-C2-specific splenic CTL response was induced by the TGF-?-insensitive DCs in the tumor bearer (85% killing activity at an effector:target cell ratio of 100:1). No apparent lysis was observed against irrelevant B16-F1 cells.
引文
1. Mellman I, Steinman RM. Dendritic cells: specialized and regulated antigen processing machines. Cell 2001;106(3):255-8.
    2. Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, et al. Immunobiology of dendritic cells. Annu Rev Immunol 2000;18:767-811.
    3. Sato K, Kawasaki H, Nagayama H, Enomoto M, Morimoto C, Tadokoro K, et al. TGF-beta 1 reciprocally controls chemotaxis of human peripheral blood monocyte-derived dendritic cells via chemokine receptors. J Immunol 2000;164(5):2285-95.
    4. Takayama T, Morelli AE, Onai N, Hirao M, Matsushima K, Tahara H, et al. Mammalian and viral IL-10 enhance C-C chemokine receptor 5 but down-regulate C-C chemokine receptor 7 expression by myeloid dendritic cells: impact on chemotactic responses and in vivo homing ability. J Immunol 2001;166(12):7136-43.
    5. Chatzaki E, Kouimtzoglou E, Margioris AN, Gravanis A. Transforming growth factor beta1 exerts an autocrine regulatory effect on human endometrial stromal cell apoptosis, involving the FasL and Bcl-2 apoptotic pathways. Mol Hum Reprod 2003;9(2):91-5.
    6. Song K, Cornelius SC, Danielpour D. Development and characterization of DP-153, a nontumorigenic prostatic cell line that undergoes malignant transformation by expression of dominant-negative transforming growth factor beta receptor type II. Cancer Res 2003;63(15):4358-67.
    7. Heiser A, Coleman D, Dannull J, Yancey D, Maurice MA, Lallas CD, et al. Autologous dendritic cells transfected with prostate-specific antigen RNA stimulate CTL responses against metastatic prostate tumors. J Clin Invest 2002;109(3):409-17.
    8. Small EJ, Fratesi P, Reese DM, Strang G, Laus R, Peshwa MV, et al. Immunotherapy of hormone-refractory prostate cancer with antigen-loaded dendritic cells. J Clin Oncol 2000;18(23):3894-903.
    9. 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):1142-62.
    10. Shortman K, Caux C. Dendritic cell development: multiple pathways to nature's adjuvants. Stem Cells 1997;15(6):409-19.
    11. Koopmann JO, Hammerling GJ, Momburg F. Generation, intracellular transport and loading of peptides associated with MHC class I molecules. Curr Opin Immunol 1997;9(1):80-8.
    12. Chen Z, Dehm S, Bonham K, Kamencic H, Juurlink B, Zhang X, et al. DNA array and biological characterization of the impact of the maturation status of mouse dendritic cells on their phenotype and antitumor vaccination efficacy. Cell Immunol 2001;214(1):60-71.
    13. Hirao M, Onai N, Hiroishi K, Watkins SC, Matsushima K, Robbins PD, et al. CC chemokine receptor-7 on dendritic cells is induced after interaction with apoptotic tumor cells: critical role in migration from the tumor site to draining lymph nodes. Cancer Res 2000;60(8):2209-17.
    14. Arrighi JF, Hauser C, Chapuis B, Zubler RH, Kindler V. Long-term culture of human CD34(+) progenitors with FLT3-ligand, thrombopoietin, and stemcell factor induces extensive amplification of a CD34(-)CD14(-) and a CD34(-)CD14(+) dendritic cell precursor. Blood 1999;93(7):2244-52.
    15. Fanger NA, Voigtlaender D, Liu C, Swink S, Wardwell K, Fisher J, et al. Characterization of expression, cytokine regulation, and effector function of the high affinity IgG receptor Fc gamma RI (CD64) expressed on human blood dendritic cells. J Immunol 1997;158(7):3090-8.
    16. Olweus J, BitMansour A, Warnke R, Thompson PA, Carballido J, Picker LJ, et al. Dendritic cell ontogeny: a human dendritic cell lineage of myeloid origin. Proc Natl Acad Sci U S A 1997;94(23):12551-6.
    17. Lipscomb MF, Masten BJ. Dendritic cells: immune regulators in health and disease. Physiol Rev 2002;82(1):97-130.
    18. Steinman RM, Dhodapkar M. Active immunization against cancer with dendritic cells: the near future. Int J Cancer 2001;94(4):459-73.
    19. Yoneyama H, Matsuno K, Zhang Y, Nishiwaki T, Kitabatake M, Ueha S, et al. Evidence for recruitment of plasmacytoid dendritic cell precursors to inflamed lymph nodes through high endothelial venules. Int Immunol 2004;16(7):915-28.
    20. Turley SJ, Inaba K, Garrett WS, Ebersold M, Unternaehrer J, Steinman RM, et al. Transport of peptide-MHC class II complexes in developing dendritic cells. Science 2000;288(5465):522-7.
    21. Ito T, Inaba M, Inaba K, Toki J, Sogo S, Iguchi T, et al. A CD1a+/CD11c+ subset of human blood dendritic cells is a direct precursor of Langerhans cells. J Immunol 1999;163(3):1409-19.
    22. Bottomly K. T cells and dendritic cells get intimate. Science 1999;283(5405):1124-5.
    23. Kuwana M. Induction of anergic and regulatory T cells by plasmacytoid dendritic cells and other dendritic cell subsets. Hum Immunol 2002;63(12):1156-63.
    24. Morikawa Y, Tohya K, Ishida H, Matsuura N, Kakudo K. Different migration patterns of antigen-presenting cells correlate with Th1/Th2-type responses in mice. Immunology 1995;85(4):575-81.
    25. Markowicz S, Engleman EG. Granulocyte-macrophage colony-stimulating factor promotes differentiation and survival of human peripheral blood dendritic cells in vitro. J Clin Invest 1990;85(3):955-61.
    26. Yao V, Platell C, Hall JC. Dendritic cells. ANZ J Surg 2002;72(7):501-6.
    27. Mackensen A, Herbst B, Chen JL, Kohler G, Noppen C, Herr W, et al. Phase I study in melanoma patients of a vaccine with peptide-pulsed dendritic cells generated in vitro from CD34(+) hematopoietic progenitor cells. Int J Cancer 2000;86(3):385-92.
    28. Liu A, Takahashi M, Narita M, Zheng Z, Kanazawa N, Abe T, et al. Generation of functional and mature dendritic cells from cord blood and bone marrow CD34+ cells by two-step culture combined with calcium ionophore treatment. J Immunol Methods 2002;261(1-2):49-63.
    29. Berthier R, Martinon-Ego C, Laharie AM, Marche PN. A two-step culture method starting with early growth factors permits enhanced production of functional dendritic cells from murine splenocytes. J Immunol Methods 2000;239(1-2):95-107.
    30. Seager Danciger J, Lutz M, Hama S, Cruz D, Castrillo A, Lazaro J, et al. Method for large scale isolation, culture and cryopreservation of human monocytes suitable for chemotaxis, cellular adhesion assays, macrophageand dendritic cell differentiation. J Immunol Methods 2004;288(1-2):123-34.
    31. Goxe B, Latour N, Chokri M, Abastado JP, Salcedo M. Simplified method to generate large quantities of dendritic cells suitable for clinical applications. Immunol Invest 2000;29(3):319-36.
    32. Son YI, Egawa S, Tatsumi T, Redlinger RE Jr, Kalinski P, Kanto T. A novel bulk-culture method for generating mature dendritic cells from mouse bone marrow cells. J Immunol Methods 2002;262(1-2):145-57.
    33. Buchler T, Kovarova L, Musilova R, Bourkova L, Ocadlikova D, Bulikova A, et al. Generation of dendritic cells using cell culture bags--description of a method and review of literature. Hematology 2004;9(3):199-205.
    34. Strobl H, Knapp W. TGF-beta1 regulation of dendritic cells. Microbes Infect 1999;1(15):1283-90.
    35. Xia CQ, Kao KJ. Suppression of interleukin-12 production through endogenously secreted interleukin-10 in activated dendritic cells: involvement of activation of extracellular signal-regulated protein kinase. Scand J Immunol 2003;58(1):23-32.
    36. Morelli AE, Zahorchak AF, Larregina AT, Colvin BL, Logar AJ, Takayama T, et al. Cytokine production by mouse myeloid dendritic cells in relation to differentiation and terminal maturation induced by lipopolysaccharide or CD40 ligation. Blood 2001;98(5):1512-23.
    37. Rescigno M, Martino M, Sutherland CL, Gold MR, Ricciardi-Castagnoli P. Dendritic cell survival and maturation are regulated by different signaling pathways. J Exp Med 1998;188(11):2175-80.
    38. Holt PG. The role of airway dendritic cell populations in regulation of T-cell responses to inhaled antigens: atopic asthma as a paradigm. J Aerosol Med2002;15(2):161-8.
    39. Bilsborough J, Viney JL. Gastrointestinal dendritic cells play a role in immunity, tolerance, and disease. Gastroenterology 2004;127(1):300-9.
    40. Christensen HR, Frokiaer H, Pestka JJ. Lactobacilli differentially modulate expression of cytokines and maturation surface markers in murine dendritic cells. J Immunol 2002;168(1):171-8.
    41. Hope JC, Thom ML, McCormick PA, Howard CJ. Interaction of antigen presenting cells with mycobacteria. Vet Immunol Immunopathol 2004;100(3-4):187-95.
    42. Geijtenbeek TB, Van Vliet SJ, Koppel EA, Sanchez-Hernandez M, Vandenbroucke-Grauls CM, Appelmelk B, et al. Mycobacteria target DC-SIGN to suppress dendritic cell function. J Exp Med 2003;197(1):7-17.
    43. Murabayashi N, Kurita-Taniguchi M, Ayata M, Matsumoto M, Ogura H, Seya T. Susceptibility of human dendritic cells (DCs) to measles virus (MV) depends on their activation stages in conjunction with the level of CDw150: role of Toll stimulators in DC maturation and MV amplification. Microbes Infect 2002;4(8):785-94.
    44. Servet-Delprat C, Vidalain PO, Bausinger H, Manie S, Le Deist F, Azocar O, et al. Measles virus induces abnormal differentiation of CD40 ligand-activated human dendritic cells. J Immunol 2000;164(4):1753-60.
    45. Li L, Liu D, Hutt-Fletcher L, Morgan A, Masucci MG, Levitsky V. Epstein-Barr virus inhibits the development of dendritic cells by promoting apoptosis of their monocyte precursors in the presence of granulocyte macrophage-colony-stimulating factor and interleukin-4. Blood 2002;99(10):3725-34.
    46. Oki M, Ando K, Hagihara M, Miyatake H, Shimizu T, Miyoshi H, et al. Efficient lentiviral transduction of human cord blood CD34(+) cells followed by their expansion and differentiation into dendritic cells. Exp Hematol 2001;29(10):1210-7.
    47. Norbury CC, Chambers BJ, Prescott AR, Ljunggren HG, Watts C. Constitutive macropinocytosis allows TAP-dependent major histocompatibility complex class I presentation of exogenous soluble antigen by bone marrow-derived dendritic cells. Eur J Immunol 1997;27(1):280-8.
    48. Adema GJ, Hartgers F, Verstraten R, de Vries E, Marland G, Menon S, et al. A dendritic-cell-derived C-C chemokine that preferentially attracts naive T cells. Nature 1997;387(6634):713-7.
    49. Lu L, Qian S, Hershberger PA, Rudert WA, Lynch DH, Thomson AW. Fas ligand (CD95L) and B7 expression on dendritic cells provide counter-regulatory signals for T cell survival and proliferation. J Immunol 1997;158(12):5676-84.
    50. Albert ML, Sauter B, Bhardwaj N. Dendritic cells acquire antigen from apoptotic cells and induce class I-restricted CTLs. Nature 1998; 392(6671):86-9.
    51. Thery C, Regnault A, Garin J, Wolfers J, Zitvogel L, Ricciardi-Castagnoli P, et al. Molecular characterization of dendritic cell-derived exosomes. Selective accumulation of the heat shock protein hsc73. J Cell Biol 1999;147(3):599-610.
    52. Almand B, Resser JR, Lindman B, Nadaf S, Clark JI, Kwon ED, et al. Clinical significance of defective dendritic cell differentiation in cancer. Clin Cancer Res 2000;6(5):1755-66.
    53. Gabrilovich DI, Ciernik IF, Carbone DP. Dendritic cells in antitumor immune responses. I. Defective antigen presentation in tumor-bearing hosts. Cell Immunol 1996;170(1):101-10.
    54. Lespagnard L, Gancberg D, Rouas G, Leclercq G, de Saint-Aubain Somerhausen N, Di Leo A, et al. Tumor-infiltrating dendritic cells in adenocarcinomas of the breast: a study of 143 neoplasms with a correlation to usual prognostic factors and to clinical outcome. Int J Cancer 1999;84(3):309-14.
    55. Steinbrink K, Wolfl M, Jonuleit H, Knop J, Enk AH. Induction of tolerance by IL-10-treated dendritic cells. J Immunol 1997;159(10):4772-80.
    56. Kiertscher SM, Luo J, Dubinett SM, Roth MD. Tumors promote altered maturation and early apoptosis of monocyte-derived dendritic cells. J Immunol 2000;164(3):1269-76.
    57. Grabbe S, Beissert S, Schwarz T, Granstein RD. Dendritic cells as initiators of tumor immune responses: a possible strategy for tumor immunotherapy?. Immunol Today 1995;16(3):117-21.
    58. D'Orazio TJ, Niederkorn JY. A novel role for TGF-beta and IL-10 in the induction of immune privilege. J Immunol 1998;160(5):2089-98.
    59. Gabrilovich DI, Chen HL, Girgis KR, Cunningham HT, Meny GM, Nadaf S, et al. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med 1996;2(10):1096-103.
    60. Allavena P, Piemonti L, Longoni D, Bernasconi S, Stoppacciaro A, Ruco L, et al. IL-10 prevents the differentiation of monocytes to dendritic cells but promotes their maturation to macrophages. Eur J Immunol1998;28(1):359-69.
    61. Takahashi A, Kono K, Ichihara F, Sugai H, Fujii H, Matsumoto Y. Vascular endothelial growth factor inhibits maturation of dendritic cells induced by lipopolysaccharide, but not by proinflammatory cytokines. Cancer Immunol Immunother 2004;53(6):543-50.
    62. Chaux P, Moutet M, Faivre J, Martin F, Martin M. Inflammatory cells infiltrating human colorectal carcinomas express HLA class II but not B7-1 and B7-2 costimulatory molecules of the T-cell activation. Lab Invest 1996;74(5):975-83.
    63. Chaux P, Favre N, Martin M, Martin F. Tumor-infiltrating dendritic cells are defective in their antigen-presenting function and inducible B7 expression in rats. Int J Cancer 1997;72(4):619-24.
    64. Troy A, Davidson P, Atkinson C, Hart D. Phenotypic characterisation of the dendritic cell infiltrate in prostate cancer. J Urol 1998;160(1):214-9.
    65. Troy AJ, Summers KL, Davidson PJ, Atkinson CH, Hart DN. Minimal recruitment and activation of dendritic cells within renal cell carcinoma. Clin Cancer Res 1998;4(3):585-93.
    66. Riker A, Cormier J, Panelli M, Kammula U, Wang E, Abati A, et al. Immune selection after antigen-specific immunotherapy of melanoma. Surgery 1999;126(2):112-20.
    67. Matsui S, Ahlers JD, Vortmeyer AO, Terabe M, Tsukui T, Carbone DP, et al. A model for CD8+ CTL tumor immunosurveillance and regulation of tumor escape by CD4 T cells through an effect on quality of CTL. J Immunol 1999;163(1):184-93.
    68. Koido S, Ohana M, Liu C, Nikrui N, Durfee J, Lerner A, et al. Dendritic cellsfused with human cancer cells: morphology, antigen expression, and T cell stimulation. Clin Immunol 2004;113(3):261-9.
    69. Kao JY, Zhang M, Chen CM, Chen JJ. Superior efficacy of dendritic cell-tumor fusion vaccine compared with tumor lysate-pulsed dendritic cell vaccine in colon cancer. Immunol Lett 2005;101(2):154-9.
    70. 马俊芬, 黄幼田, 赵明耀, 杨洪艳, 郑智敏, 董子明. 转染pcDNA3-hCEA的人树突状细胞抑制 MGC803 裸鼠移植瘤的生长. 第四军医大学学报 2005;26(19):1802-1804.
    71. Gilboa E, Vieweg J. Cancer immunotherapy with mRNA-transfected dendritic cells. Immunol Rev 2004;199:251-63.
    72. Gong J, Nikrui N, Chen D, Koido S, Wu Z, Tanaka Y, et al. Fusions of human ovarian carcinoma cells with autologous or allogeneic dendritic cells induce antitumor immunity. J Immunol 2000;165(3):1705-11.
    73. Schadendorf D, Paschen A, Sun Y. Autologous, allogeneic tumor cells or genetically engineered cells as cancer vaccine against melanoma. Immunol Lett 2000;74(1):67-74.
    74. Siders WM, Vergilis KL, Johnson C, Shields J, Kaplan JM. Induction of specific antitumor immunity in the mouse with the electrofusion product of tumor cells and dendritic cells. Mol Ther 2003;7(4):498-505.
    75. Orentas RJ, Schauer D, Bin Q, Johnson BD. Electrofusion of a weakly immunogenic neuroblastoma with dendritic cells produces a tumor vaccine. Cell Immunol 2001;213(1):4-13.
    76. Tanaka H, Shimizu K, Hayashi T, Shu S. Therapeutic immune response induced by electrofusion of dendritic and tumor cells. Cell Immunol 2002;220(1):1-12.
    77. Cao X, Zhang W, Wang J, Zhang M, Huang X, Hamada H, et al. Therapy of established tumour with a hybrid cellular vaccine generated by using granulocyte-macrophage colony-stimulating factor genetically modified dendritic cells. Immunology 1999;97(4):616-25.
    78. Lamparski HG, Metha-Damani A, Yao JY, Patel S, Hsu DH, Ruegg C, et al. Production and characterization of clinical grade exosomes derived from dendritic cells. J Immunol Methods 2002;270(2):211-26.
    79. Fong L, Brockstedt D, Benike C, Breen JK, Strang G, Ruegg CL, et al. Dendritic cell-based xenoantigen vaccination for prostate cancer immunotherapy. J Immunol 2001;167(12):7150-6.
    80. Murphy GP, Tjoa BA, Simmons SJ, Jarisch J, Bowes VA, Ragde H, et al. Infusion of dendritic cells pulsed with HLA-A2-specific prostate-specific membrane antigen peptides: a phase II prostate cancer vaccine trial involving patients with hormone-refractory metastatic disease. Prostate 1999;38(1):73-8.
    81. Xia D, Zheng S, Zhang W, He L, Wang Q, Pan J, et al. Effective induction of therapeutic antitumor immunity by dendritic cells coexpressing interleukin-18 and tumor antigen. J Mol Med 2003;81(9):585-96.
    82. Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor beta in human disease. N Engl J Med 2000;342(18):1350-8.
    83. Zhu S, Goldschmidt-Clermont PJ, Dong C. Transforming growth factor-beta-induced inhibition of myogenesis is mediated through Smad pathway and is modulated by microtubule dynamic stability. Circ Res 2004;94(5):617-25.
    84. Suzuki H, Yagi K, Kondo M, Kato M, Miyazono K, Miyazawa K. c-Skiinhibits the TGF-beta signaling pathway through stabilization of inactive Smad complexes on Smad-binding elements. Oncogene 2004;23(29):5068-76.
    85. Shi Y, Massague J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell 2003;113(6):685-700.
    86. Liu C, Gaca MD, Swenson ES, Vellucci VF, Reiss M, Wells RG. Smads 2 and 3 are differentially activated by transforming growth factor-beta (TGF-beta ) in quiescent and activated hepatic stellate cells. Constitutive nuclear localization of Smads in activated cells is TGF-beta-independent. J Biol Chem 2003;278(13):11721-8.
    87. Chen HB, Rud JG, Lin K, Xu L. Nuclear targeting of transforming growth factor-beta-activated Smad complexes. J Biol Chem 2005;280(22):21329-36.
    88. Kurisaki A, Kose S, Yoneda Y, Heldin CH, Moustakas A. Transforming growth factor-beta induces nuclear import of Smad3 in an importin-beta1 and Ran-dependent manner. Mol Biol Cell 2001;12(4):1079-91.
    89. Cordenonsi M, Dupont S, Maretto S, Insinga A, Imbriano C, Piccolo S. Links between tumor suppressors: p53 is required for TGF-beta gene responses by cooperating with Smads. Cell 2003;113(3):301-14.
    90. Qing J, Liu C, Choy L, Wu RY, Pagano JS, Derynck R. Transforming growth factor beta/Smad3 signaling regulates IRF-7 function and transcriptional activation of the beta interferon promoter. Mol Cell Biol 2004;24(3):1411-25.
    91. Moustakas A, Heldin CH. Non-Smad TGF-beta signals. J Cell Sci 2005;118(Pt 16):3573-84.
    92. Edlund S, Landstrom M, Heldin CH, Aspenstrom P. Transforming growthfactor-beta-induced mobilization of actin cytoskeleton requires signaling by small GTPases Cdc42 and RhoA. Mol Biol Cell 2002;13(3):902-14.
    93. Bhowmick NA, Ghiassi M, Bakin A, Aakre M, Lundquist CA, Engel ME, et al. Transforming growth factor-beta1 mediates epithelial to mesenchymal transdifferentiation through a RhoA-dependent mechanism. Mol Biol Cell 2001;12(1):27-36.
    94. Turco A, Scarpa S, Coppa A, Baccheschi G, Palumbo C, Leonetti C, et al. Increased TGFbeta type II receptor expression suppresses the malignant phenotype and induces differentiation of human neuroblastoma cells. Exp Cell Res 2000;255(1):77-85.
    95. Bandyopadhyay A, Zhu Y, Cibull ML, Bao L, Chen C, Sun L. A soluble transforming growth factor beta type III receptor suppresses tumorigenicity and metastasis of human breast cancer MDA-MB-231 cells. Cancer Res 1999;59(19):5041-6.
    96. Teicher BA. Malignant cells, directors of the malignant process: role of transforming growth factor-beta. Cancer Metastasis Rev 2001;20(1-2):133-43.
    97. Shariat SF, Menesses-Diaz A, Kim IY, Muramoto M, Wheeler TM, Slawin KM. Tissue expression of transforming growth factor-beta1 and its receptors: correlation with pathologic features and biochemical progression in patients undergoing radical prostatectomy. Urology 2004;63(6):1191-7.
    98. Bello-DeOcampo D, Tindall DJ. TGF-betal/Smad signaling in prostate cancer. Curr Drug Targets 2003;4(3):197-207.
    99. Tuxhorn JA, McAlhany SJ, Yang F, Dang TD, Rowley DR. Inhibition of transforming growth factor-beta activity decreases angiogenesis in a humanprostate cancer-reactive stroma xenograft model. Cancer Res 2002;62(21):6021-5.
    100. Shah AH, Tabayoyong WB, Kundu SD, Kim SJ, Van Parijs L, Liu VC, et al. Suppression of tumor metastasis by blockade of transforming growth factor beta signaling in bone marrow cells through a retroviral-mediated gene therapy in mice. Cancer Res 2002;62(24):7135-8.
    101. Bachman KE, Park BH. Duel nature of TGF-beta signaling: tumor suppressor vs. tumor promoter. Curr Opin Oncol 2005;17(1):49-54.
    102. Gatti E, Pierre P. Understanding the cell biology of antigen presentation: the dendritic cell contribution. Curr Opin Cell Biol 2003;15(4):468-73.
    103. Ardavin C. Origin, precursors and differentiation of mouse dendritic cells. Nat Rev Immunol 2003;3(7):582-90.
    104. Vegh Z, Mazumder A. Generation of tumor cell lysate-loaded dendritic cells preprogrammed for IL-12 production and augmented T cell response. Cancer Immunol Immunother 2003;52(2):67-79.
    105. Tobias CA, Shumsky JS, Shibata M, Tuszynski MH, Fischer I, Tessler A, et al. Delayed grafting of BDNF and NT-3 producing fibroblasts into the injured spinal cord stimulates sprouting, partially rescues axotomized red nucleus neurons from loss and atrophy, and provides limited regeneration. Exp Neurol 2003;184(1):97-113.
    106. Ferrer I, Krupinski J, Goutan E, Marti E, Ambrosio S, Arenas E. Brain-derived neurotrophic factor reduces cortical cell death by ischemia after middle cerebral artery occlusion in the rat. Acta Neuropathol (Berl) 2001;101(3):229-38.
    107. Zhang Q, Jang TL, Yang X, Park I, Meyer RE, Kundu S, et al. Infiltration oftumor-reactive transforming growth factor-beta insensitive CD8+ T cells into the tumor parenchyma is associated with apoptosis and rejection of tumor cells. Prostate 2006;66(3):235-47.
    108. Kaklamani VG, Pasche B. Role of TGF-beta in cancer and the potential for therapy and prevention. Expert Rev Anticancer Ther 2004;4(4):649-61.
    109. Yingling JM, Blanchard KL, Sawyer JS. Development of TGF-beta signalling inhibitors for cancer therapy. Nat Rev Drug Discov 2004;3(12):1011-22.
    110. Gorelik L, Flavell RA. Immune-mediated eradication of tumors through the blockade of transforming growth factor-beta signaling in T cells. Nat Med 2001;7(10):1118-22.
    111. Shah AH, Tabayoyong WB, Kimm SY, Kim SJ, Van Parijs L, Lee C. Reconstitution of lethally irradiated adult mice with dominant negative TGF-beta type II receptor-transduced bone marrow leads to myeloid expansion and inflammatory disease. J Immunol 2002;169(7):3485-91.

© 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