超嗜热古菌Pyrococcus horikoshii OT3组蛋白介导野生型p53基因治疗鼻咽癌的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
基因导入系统是基因治疗的核心技术,基因治疗面临的首要问题是载体的选择,安全无毒、生物相容性好的非病毒载体已成为基因治疗研究的热点。本文利用来源于超嗜热古菌Pyrococcus horikoshii重组古菌组蛋白(HPhA),构建了HPhA/pCMV-p53(wt)基因导入系统,探讨HPhA携带野生型p53基因对恶性肿瘤基因治疗的可行性。我们考察外源基因的转录水平和蛋白质表达水平及基因表达产物对癌细胞周期及细胞凋亡的影响;并建立裸鼠肿瘤模型,进行荷瘤裸鼠的体内肿瘤生长情况及肿瘤细胞的凋亡的考察,探索其在体内基因治疗方面的应用价值;同时研究不同给药途径及剂量的HPhA在小鼠中血药浓度随时间变化的趋势,并采用房室模型法,利用Topfit软件计算分析相应的药动学参数。最后考察重组古菌组蛋白的安全性。结果表明,HPhA能携带外源基因pCMV-p53高效转染肿瘤细胞,外源基因表达产物可有效启动细胞凋亡信号转导通路,抑制肿瘤细胞生长,促使肿瘤细胞调亡;在小鼠体内,HPhA肌肉注射给药和静脉注射给药在小鼠体内的药代动力学行为符合开放性二室模型;静脉注射给药由中央向周边室分布速度较快,消除为剂量依赖性非线性动力学过程;同时进行重组古菌组蛋白在鼠模型中体液免疫和细胞免疫应答的研究,具有较低的免疫原性,是理想的新型基因药物载体。
Gene therapy is becoming an important strategy in the treatment of various diseases such as cancer、AIDS、cystic fibrosis and vaccination. Effective gene transfer is an important prerequisite for gene therapy. The development of novel widely application DNA transfection system is an important goal of current molecular biological research. Generally, there are two main types of gene delivery vector: viral and non-viral. Viral vectors, the current favorites for intracellular DNA delivery, suffer from immunogenicity、nonspecificity and inherent risks of complications such as interference with the activity of tissue-specific promoters. Non-viral delivery systems are expected to have little or no harmful side-effects. The development of such systems with high efficiency、avoiding toxic and immunogenic problems, is essential and urgent.
     One approach focuses on the use of DNA-binding proteins as carriers of foreign genes into the host cell. The potential advantages of protein gene transfer include ease of use, production, purity, homogeneity, ability to target nucleic acids to specific cell types, the potential for cost-effective large-scale manufacture and the lack of limitation on production costs.
     We have previously described an archaeal histone-like protein-based (HPhA) gene delivery system and showed that HPhA formed stable non-covalent complexes with nucleic acids and improved their delivery by usingβ-galactosidase as a reporter gene. HPhA, an archaeal histone-like protein shares the properties of eukaryal histones: small, basic, toroidally wrap DNA, protect DNA from nuclease digestion and/or form nucleosome-like structures. These features make it potential carrier of gene deliveries.
     The p53 gene has been extensively studied and is known to play a critical role in cell regulation and control of apoptosis. Gene therapy using the p53 gene has been proposed for and performed with inactivation of p53 function. However, efficient and safe gene delivery remains a key issue for p53 based gene therapy. Because the functional loss of p53 gene was now well recognized as the most common event in carcinognesis, it was not surprising that wild-type p53 gene introduction had recently been used as a cancer gene therapy.
     In the present study, we attempted to use HPhA with naked plasmid DNA, which contains a wt-p53 tumor suppressor gene, and we evaluated the transfection efficiency and antitumor activity in human p53-deleted Nasopharyngeal Carcinoma (CNE) cells in vitro and in vivo. We observed that p53 delivery resulted in a more differential growth inhibition pattern in cancer cells in vitro and in vivo. The results showed a significant efficiency of gene transfer and gene expression. Higher transfection of HPhA/p53 complexes in CNE cells seemed to be responsible for higher expression of p53 mRNA. Efficient cell growth inhibition by HPhA/p53 complexes could be due to the increase of the expression of p53 proteins as well as due to the effect of histones on the proliferation of CNE cells. When using histones as DNA vectors, excess of the added histones could potentially interfere with cellular histones, and play regulatory function in transcription and slowed the cell cycle progression. Flow cytometric analysis suggested that the cytotoxic effect of HPhA/p53 was the result of apoptosis and cell cycle arrest. Encouraging results were also obtained in studies using tumor xenografted nude mice. Local HPhA/p53 treatment by intra-tumoral injection led to a significant inhibition of tumor growth consistent with the in vitro effect of HPhA-p53 on human cancer cells.
     Thus, it was supposed that functional exogenous p53 could be efficiently delivered in cancer cell lines using HPhA as a nonviral vector. Serum did not exert any inhibition of the transfection mediated by HPhA, the HPhA/p53 complexes could improve the stability of plasmid DNA in the presence of serum. In contrast, many cationic lipid-based transfection systems were inactivated by medium containing as low as 5-10% serum. Thus, the higher transfetion efficiency of plasmid DNA may be attributed to the enhanced stability of plasmid HPhA/DNA complexes. Moreover, the enhanced mRNA expression of p53 may increase the protein levels of p53 in the cells transfected with HPhA/p53 complexes compared to the cells treated with Lipofectamine complexes or the plasmid alone. And the penetration of histone into tissue-cultured cells occurring by direct translocation through the cell plasma membrane and not by a typical endocytosis, may facilitate the escape of DNA from the endosome. HPhA may be highly efficient gene delivery.
     Efficient gene delivery is one of the most important tasks in current non-viral gene therapy research. An ideal vector may find its application for in vivo and in vitro transfection of cells or grafts for transplantation and for various protocols of a localized gene therapy. Nuclear proteins, such as histones mediated gene transfer system might be an ideal system for gene transfer. Histones, which were natural DNA binding and condensing proteins, offered good advantages to serve as a safe and efficient gene delivery tool. Histones had been demonstrated to have higher efficiency of gene transfer compared with other non-viral transfer systems. Therefore, it was thought to be one of the most attractive alternative vectors.
     This study was designed to evaluate the efficiency and potential applicability of HPhA carrier system mediated p53 gene therapy. The results showed the HPhA-mediated transfection of tumor suppressor gene p53 was effective in inducing apoptosis and inhibiting tumor growth. Our results suggested the HPhA-mediated p53 delivery might have the potential for clinical application of nonviral vector mediated cancer therapy.
     The fate of gene delivery vectors in the body, i.e., their pharmacokinetics, is also another important factor in in vivo gene therapy. The pharmacokinetics determines the availability of the vector following in vivo administration. To our knowledge, no literature information is available on the pharmacokinetics of HPhA or other related histone.
     A competitive direct enzyme-linked immunosorbent assay (ELISA) has been developed to determine concentrations of an archaeal histone-like protein (HPhA) in mice plasma. Direct ELISA can be regarded as the simplest form of ELISA, of which there are two formats. One is antibody-coated ELISA, and the other is antigen-coated ELISA. In this study, the first format was developed to determine HPhA concentrations in rat plasma used for the investigation of the pharmacokinetics of HPhA. The most widely used enzymes for labeling are horseradish peroxidase (HRP), alkaline phosphatase (AP), andβ-galactosidase. In this study, horseradish peroxidase was selected to label gossypol because HRP had been proved with high turnover number and stability.
     Firstly, Anti-HPhA monoclonal antibody was purified by ammonium sulfate precipitation and ProteinA AffinityPak. The antigen (HPhA) was labeled with horseradish peroxidase by periodate oxidation method. Both the purified antibody and the enzyme-labeled HPhA were employed to develop cdELISA to facilitate further analysis of HPhA pharmacokinetics. The purified antibody was diluted in a coating buffer, carbonate/bicarbonate buffer (pH 9.6). After coating, any excess free antibody was removed by a washing step with a neutral buffered solution (PBST). The blocking solution (1% BSA in PBS, containing 0.1% sodium azide) was then added. After another washing step, a known amount of HRP labeled HPhA with HPhA standards or samples was added. After washing away the unbound materials, the TMB substrate was added and the color developed was reversely proportional to the amount of antigen from standards or samples. The resultant CV and RE in ELISA were within 15% at all concentrations determined for all species, so the ELISA had satisfactory accurary and precision and was an appropriate assay method to examine the pharmacokinetics of HPhA. The ELISA was suitable for measuring the compound over long periods of time and may provide an alternative technique for monitoring drug levels in clinical samples. The assay was used to determine the pharmacokinetics of HPhA in mice.
     The pharmacokinetic results of the HPhA after i.m. injection in rats showed that the plasma concentration-time course fitted a two-compartment open model. The pharmacokinetic results of the HPhA after i.v. injection in rats showed that the plasma concentration-time course fitted a two-compartment open model. The short half-life of theα?phase indicated that the HPhA was rapidly distributed from the central to the peripheral compartment after injection.
     We investigated the immunogenicity of the recombinant archaeal histone (HphA) as a non-viral gene delivery in BALB/c mice, which provided a good base for safe and effective application of HphA. And mice were divided randomly, and then immunized by HphA、HphA combining with Freuds adjuvant and bovine serum albumin combining with Freuds adjuvant. Humoral immunoresponse was evaluated with the serum IgG level tested by ELISA. And cellullar immunologic response was evaluated with stimulation indexes and levels of secreted IFN-γof splenocytes. Results showed Mice immunized with HphA produced no specific antibody, and the stimulation indexes and levels of IFN-γhad no difference from the control group. The HphA had no immunogenicity in mouse,and could not produce humoral and cellular immune response.
     In this article, HPhA proved to enhance the in vitro and in vivo efficiency of p53 gene transfer and is a promising new strategy for p53 gene therapy. Pharmacokinetic and safety analysis of HPhA will give us information about the events occurring in the body following administration, and it is very powerful tool to make significant contribution in the rational design of HPhA mediated gene therapy. All demonstrates that HPhA may serve as a promising, wildly applicable and highly efficient tool for gene delivery and gene therapy.
引文
[1] Jacobs AH, Voges J, et al. Imaging in gene therapy of patients with glioma. J Neurooncol. 2003, 65: 291~305.
    [2] Naker DR. Public acceptance of human gene therapy and perceptions of human genetic manipulation. Hum Gene Ther. 1992, 3: 511~518
    [3] Anderson WF. Human gene therapy. Nature. 1998, 392: 25~30.
    [4] 翁粱. 超嗜热古菌 pyrococcus horikoshii OT3 组蛋白的克隆、表达、性质表征及其在基因治疗中的应用. 博士论文. 2004
    [5] Niculescu-Duvaz I, Springer CJ. Introduction to the background, principles, and state of the art in suicide gene therapy. Mol Biotechnol. 2005, 30:71-88.
    [6] Niculescu-Duvaz D, Niculescu-Duvaz I, Springer CJ. Design of prodrugs for suicide gene therapy. Methods Mol Med. 2004, 90:161-202.
    [7] 刘军,赵斌. 肝癌自杀基因治疗研究进展. 国外医学肿瘤学分册,2000, 27: 248-250.
    [8] Kausch I, Ardelt P, Bohle A, Ratliff TL. Immune gene therapy in urology. Curr Urol Rep. 2002, 3: 82-89.
    [9] 字应伟. 肿瘤基因治疗研究进展.思茅师范高等专科学校学报, 2005, 21: 6-9.
    [10] Kim HL, Belldegrun AS, Figlin RA. Immune gene therapy for kidney cancer: the search for a magic trigger. Mol Ther. 2003, 7:153-4.
    [11] Kausch I, Ardelt P, Bohle A, et al. Immune gene therapy in urology. Curr Urol Rep. 2002, 3: 82-89.
    [12] Woo SL, Jia F, Zou L, Gabriel MT. Functional tissue engineering for ligament healing: potential of antisense gene therapy. Ann Biomed Eng. 2004, 32: 342-351.
    [13] Raizada MK, Katovich MJ, Wang H, et al. Is antisense gene therapy a step in the right direction in the control of hypertension? Am J Physiol. 1999, 277: H423-32.
    [14] 田长海,李祺福,欧阳高亮. 肿瘤反基因治疗研究进展. 国外医学肿瘤学分册. 2001, 28: 274-276.
    [15] Davis BM, Roth JC, Liu L. Characterization of the P140K, PVP(138-140)MLK, and G156A O6-methylguanine-DNA methyltransferase mutants: implications for drug resistance gene therapy. Hum Gene Ther. 1999, 10: 2769-2778.
    [16] 吴瑞琼. 肿瘤耐药基因治疗研究新进展. 国外医学肿瘤学分册.1999, 26:68-72.
    [17] Neff T, Beard BC, Peterson LJ, et al. Polyclonal chemoprotection against temozolomide in a large-animal model of drug resistance gene therapy. Blood. 2005, 105: 997-1002.
    [18] Hofseth L J, Hussain S P, Curtis C. Harris. p53: 25 years after its discovery. Trence in Pharmacological Sciences. 2004, 25: 177-181.
    [19] Yonish-Rouach E, Resnitzky D, Lotem J, et al. Wild-type p53 induces apoptosis of myeloid leukaemic cells that is inhibited by interleukin-6, Nature. 1991, 352: 345–347.
    [20] Shaw P, Bovey R, Tardy S, et al. Induction of apoptosis by wild-type p53 in a human colon tumor-derived cell line. Proc Nat. Acad Sci. 1992, 89: 4495–4499.
    [21] Sigal A. Rotter V. Oncogenic mutations of the p53 tumor suppressor: the demons of the guardian of the genome. Cancer Res. 2000, 60: 6788–6793.
    [22] Greenblatt MS, Bennett WP, Hollstein M, et al. Mutations in the p53 tumorsuppressorgene: clues to cancer etiology and molecular pathogenesis. Cancer Res.1994, 54: 4855–4878.
    [23] Alain GZ, Karin R, Jennifer B, New Insights into p53 Regulation and Gene Therapy for Cancer. Biochemical Pharmacology. 2000, 60: 1153–1163.
    [24] Harris CC. p53 Tumor suppressor gene: from the basic research laboratory to the clinic – an abridged historical perspective. Carcinogenesis. 1996, 17: 1187–1198.
    [25] Xin L. P53: a heavily dictated dictator of life and death. Current Opinion in Genetics & Development. 2005, 15: 27-33.
    [26] Vousden KH, Lu X. Live or let die: the cells response to p53. Nat. Rev. Cancer. 2002, 2: 594–604.
    [27] Vogelstein B, Lane D, Levine A.J. Surfing the p53 network, Nature. 2000, 408 : 307–310.
    [28] Michalak E, Villunger A, Erlacher M, et al. Death squads enlisted by the tumour suppressor p53. Biochemical and Biophysical Research Communications. 2005, 331: 786–798.
    [29] Hofseth LJ, Hussain SP, Curtis CH. p53: 25 years after its discovery. Trence in Pharmacological Sciences. 2004, 25: 177-181.
    [30] Yonish-Rouach E, Resnitzky D, Lotem J, et al. Wild-type p53 induces apoptosis of myeloid leukaemic cells that is inhibited by interleukin-6. Nature. 1991, 352: 345–347.
    [31] Shaw P, Bovey R, Tardy S, et al. Induction of apoptosis by wild-type p53 in a human colon tumor-derived cell line. Proc Nat. Acad Sci. 1992, 89: 4495–4499.
    [32] Ramqvist T, Magnusson KP, Wang Y, et al. Wild-type p53 induces apoptosis in aBurkitt lymphoma (BL) line that carries mutant p53. Oncogene. 1993, 8: 1495–1500.
    [33] Johnson P, Chung S, Benchimol S. Growth suppression of Friend virus-transformed erythroleukemia cells by p53 protein is accompanied by hemoglobin production and is sensitive toerythropoietin. Mol Cell. Bio. 1993, 13:1456–1462.
    [34] 马 煜,钱德芳,曹利平. p53 蛋白研究进展 国外医学临床生物化学与检验学分册. 2002, 23: 41-42.
    [35] Wang X. The expanding role of mitochondria in apoptosis. Genes Dev. 2001, 15: 2922–2933.
    [36] Danial NN, Korsmeyer SJ. Cell death: critical control points. Cell. 2004, 116: 205–219.
    [37] Woods YL. Lane DP. Exploiting the p53 pathway for cancer diagnosis and therapy. Hematol. 2003, 4: 233–247.
    [38] Borresen-Dale AL. TP53 and breast cancer. Hum. Mutat. 2003, 21: 292–300.
    [39] Sigal, A. Rotter V. Oncogenic mutations of the p53 tumor suppressor: the demons of the guardian of the genome. Cancer Res. 2000, 60: 6788–6793.
    [40] Greenblatt MS, Bennett WP, Hollstein M, et al. Mutations in the p53 tumor suppressorgene: clues to cancer etiology and molecular pathogenesis. Cancer Res.1994, 54: 4855–4878.
    [41] Szymanska K, Hainaut P. TP53 and mutations in human cancer. Acta Biochim Pol. 2003, 50: 231-238.
    [42] Hussain SP, Raja K, Amstad PA, et al. Increased p53 mutation load in nontumoroushuman liver of Wilson disease and hemochromatosis:oxyradicaloverload diseases. Proc. Natl. Acad. Sci. 2000, 97: 12770–12775.
    [43] Kirk GD, Camus-Randon AM, Mendy M, et al. Ser-249 p53 mutations in plasma DNA of patients with hepatocellular carcinoma from The Gambia. J. Natl.Cancer Inst. 2000, 92: 148–153.
    [44] Lepik D, Jaks V, Kadaja L, et al. Electroporation and carreier DNA cause p53 activation, cell cycle arrest, and apoptosis. Analysis Biochemistry. 2003, 318: 52-59.
    [45] Wolf JK, Mills GB, Bazzet L, Bast RC Jr, Roth JA, Gershenson DM. Adenovirus-mediated p53 growth inhibition of ovarian cancer cells is independent of endogenous p53 status. Gynecol Oncol. 1999, 75: 261-266.
    [46] Shono T, Tofilon PJ, Schaefer TS, Parikh D, Liu TJ, Lang FF. Apoptosis induced by adenovirus-mediated p53 gene transfer in human glioma correlates with site-specific phosphorylation. Cancer Res. 2002, 62: 1069-1076.
    [47] Kojima Y, Honda K, Kotegawa H, Kushihata F, Kobayashi N, Liu B, et al. Adenovirus-mediated p53 gene transfer to the bile duct by direct administration into the bile in a rat cholangitis model. J Surg Res. 2005, 128: 126-131.
    [48] Takahashi T, Carbone D, Nau MM, et al. Wild type but not mutant p53 suppressess the growth of human lung cancer cells learing multiple genetic lesions. Cancer Res. 1992, 52: 2340-2343.
    [49] Su S, Watanabe A, Yamamoto M. Mutations in p53 cDNA sequence introduced by retroviral vector. Biochem Biophys Res Commun. 2006, 340: 567-572.
    [50] Lu X, Lozano G, Donehower LA. Activities of wildtype and mutant p53 in suppression of homologous recombination as measured by a retroviral vector system. Mutat Res. 2003, 522: 69-83.
    [51] Kim CK, Choi EJ, Choi SH, Park JS, Haider KH, Ahn WS. Enhanced p53 gene transfer to human ovarian cancer cells using the cationic nonviral vector, DDC. Gynecol Oncol 2003, 90: 265-272.
    [52] Nakase M, Inui M, Okumura K, Kamei T, Nakamura S, Tagawa T. p53 gene therapy of human osteosarcoma using a transferrin-modified cationic liposome. Mol Cancer Ther. 2005, 4: 625-631.
    [53] Susan CM, Pedro R, Zheife Z, et al. In vitro and in vivo adnovirus-mediated p53 and p16 tumor suppressor therapy in ovarian cancer. Clin Cancer Res. 2001, 7: 1765-1772.
    [54] Tchernev G, Orfanos CE. Downregulation of cell cycle modulators p21, p27, p53, Rb and proapoptotic Bcl-2-related proteins Bax and Bak in cutaneous melanoma is associated with worse patient prognosis: preliminary findings. J Cutan Pathol. 2007, 34: 247-256.
    [55] Garcia-Tunon I, Ricote M, Ruiz A, et al. Cell cycle control related proteins (p53, p21, and Rb) and transforming growth factor beta (TGFbeta) in benign and carcinomatous (in situ and infiltrating) human breast: implications in malignant transformations. Cancer Invest. 2006, 24: 119-125.
    [56] Chen B, Timiryasova TM, Andres ML, et al. Evaluation of combined vaccinia virus-mediated antitumor gene therapy with p53, IL-2, and IL-12 in a glioma model. Cancer Gene Ther. 2000, 7:1437-47.
    [57] Lai KC, Chen WC, Jeng LB, et al. Association of genetic polymorphisms of MK, IL-4, p16, p21, p53 genes and human gastric cancer in Taiwan. Eur J Surg Oncol. 2005, 31: 1135-1140.
    [58] Russo P, Malacarne D, Falugi C, et al. RPR-115135, a farnesyltransferase inhibitor,increases 5-FU- cytotoxicity in ten human colon cancer cell lines: role of p53. Int J Cancer. 2002, 100: 266-275.
    [59] Osaki M, Tatebe S, Goto A, et al. 5-Fluorouracil (5-FU) induced apoptosis in gastric cancer cell lines: role of the p53. gene.Apoptosis. 1997, 2: 221-226.
    [60] Matsuhashi N, Saio M, Matsuo A, et al. The evaluation of gastric cancer sensitivity to 5-FU/CDDP in terms of induction of apoptosis: time- and p53 expression-dependency of anti-cancer drugs. Oncol Rep. 2005, 14: 609-615.
    [61] Masunaga S, Uto Y, Nagasawa H, et al. Evaluation of hypoxic cell radio-sensitizers in terms of radio-sensitizing and repair-inhibiting potential. Dependency on p53 status of tumor cells and the effects on intratumor quiescent cells. Anticancer Res. 2006, 26: 1261-270.
    [62] Mroz RM, Holownia A, Chyczewska E, et al. p53 N-terminal Ser-15 approximately P and Ser-20 approximately P levels in squamous cell lung cancer after radio/chemotherapy. Am J Respir Cell Mol Biol. 2004, 30: 564-568.
    [63] Verma IM, and Somia N. Gene therapy--promises, problems and prospects. Nature. 1997; 389: 239-242.
    [64] Deanna C and James K B. Gene Therapy for Cancer Treatment: Past, Present and Future. Clinical Medicine & Research. 2006, 3: 218-227
    [65] Balicki D, and Beutler E, Gene therapy of human disease. Medicine. 2002, 81: 69-86.
    [66] Kovesdi I, Brough DE, Bruder TJ, et al. Adenoviral vectorsfor gene transfer. Curr Opin. Biotechnol.. 1997, 85: 583-589.
    [67] Mahato RI, Smith LC and Rolland A. Pharmaceutical perspectives of nonviral gene therapy. Adv Genet. 1999, 41: 95-156.
    [68] Tousignan JD, Gates AL, Ingram LA, et al. Comprehensive analysis of theacute toxicities induced by systemic administration of cationic lipid:plasmid DNA complexes in mice. Hum Gene Ther. 2000, 11: 2493-2513.
    [69] Filion MC and Phillips NC. Toxicity and immunomodulatory activity ofliposomal vectors formulated with cationic lipids toward immune effectorcells. Biochim Biophys Acta. 1997, 1329: 345-356.
    [70] Kaouass M, Raymond B and Danuta Balicki. Histonefection: Novel and potent non-viral gene delivery. J Controlled Release. 2006, 113 : 245-254.
    [71] Reddy TR, Suryanarayana T. Archaebacterial histone-like proteins. Purification and characterization of helix stabilizing DNA binding proteins from the acidothermophile Sulfolobus acidocaldarius. J Biol Chem. 1989, 264: 17298-17308.
    [72] Green GR, Searcy DG, DeLange RJ.Histone-like protein in the Archaebacterium Sulfolobus acidocaldarius. Biochim Biophys Acta. 1983, 741: 251-257.
    [73] Chen X, Tang Y, Huang L.Overproduction and partial characterization of the Ssh7a and Ssh7b proteins from Sulfolobus shibatae.Wei Sheng Wu Xue Bao. 2000, 40: 359-364.
    [74] Zaitsev SV, Haberland A, Otto A, et al. H1 and HMG17 extracted from calf thymus nuclei are efficient DNA carriers in gene transfer. Gene Ther.1997, 4: 586–592.
    [75] Tanaka Y, Tawaramoto-Sasanuma M, Kawaguchi S, et al. Expression and purification of recombinant human histones. Methods. 2004, 33: 3-11.
    [76] Bharath MM, Khadake JR, Rao MR. Expression of rat histone H1d in Escherichia coli and its purification. Protein Expr Purif. 1998, 12: 38-44.
    [77] Chen J, Stickles RJ, Daichendt KA. Galactosylated histone-mediated gene transferand expression. Hum Gene The. 1994, 5: 429-35.
    [78] 戴菲寒, 任常春, 陈燕, 等. 基因工程技术制备以组蛋白为核心的新型基因导入系统. 肿瘤, 2002, 22: 349-354.
    [79] 戴菲寒, 陈燕, 龚毅, 等. 在大肠杆菌体内有效地表达以组蛋白为骨架的融合蛋白 HNHG. 生物化学与生物物理学报, 2002, 34: 800-803.
    [80] Esser D, Amanuma H, Yoshiki A, et al. A hyperthermostable bacterial histone-like protein as an efficient mediator for transfection of eukaryotic cells. Nat Biotechnol. 2000, 18: 1211-1213.
    [81] Zaitsev S, Buchwalow I, Haberland A,et al., Histone H1-mediated transfection: role of calcium inthe cellular uptake and intracellular fate of H1-DNA complexes. Acta Histochem. 2002, 104: 85-92.
    [82] Budker V, Hagstrom JE, Lapina O et al. Protein/amphipathic polyamine complexes enable highly efficient transfection with minimal toxicity. Biotechniques. 1997, 139: 142-147.
    [83] Fritz JD, Herweijer H, Zhang G, et al. Gene transfer into mammalian cells using histone-condensed plasmid DNA. Hum. Gene Ther. 1996, 7: 1395-1404.
    [84] Haberland A, Knaus T, Zaitsev SV, et al. Histone H1-mediated transfection: serum inhibition can be overcome by Ca2+ ions. Pharm. Res. 2000, 17: 229-235.
    [85] Hagstrom JE, Sebestyen MG, Budker V, et al. Complexes of non-cationic liposomes and histone H1 mediate efficient transfection of DNA without encapsulation. Biochim Biophys Acta. 1996, 1284: 47-55.
    [86] Kott M, Haberland A, Zaitsev S, et al. A new efficient method for transfection of neonatal cardiomyocytes using histone H1 in combination with DOSPER liposomaltransfection reagent. Somat. Cell Mol. Genet. 1998, 24: 257-261.
    [87] Lucius H, Haberland A, Zaitsev S, et al. Structure of transfection-active histone H1/DNA complexes. Mol Biol Rep. 2001, 28: 157-165.
    [88] Zaitsev SV, Haberland A, Otto A, et al. H1 and HMG17 extracted from calf thymus nuclei are efficient DNA carriers in gene transfer. Gene Ther. 1997, 4: 586-592.
    [89] Demirhan I, Hasselmayer O, Chandra A, et al. Histone-mediated transfer and expression of the HIV-1 tat gene in Jurkat cells. J Hum Virol. 1998, 1: 430-440.
    [90] Bottger M, Zaitsev SV, Otto A, et al. Acid nuclear extracts as mediators of gene transfer and expression. Biochim Biophys Acta. 1998, 1395: 78-87.
    [91] Haberland A, Knaus T, Zaitsev SV, , et al. Calcium ions as efficient cofactor of polycationmediated gene transfer. Biochim. Biophys. Acta.1999, 1445: 21-30.
    [92] Puebla I, Esseghir S, Mortlock A,, et al. recombinant H1 histone-based system for efficient delivery of nucleic acids. J Biotechnol. 2003, 105: 215-226.
    [93] Moran F, Montero, Azorin F. Condensation of DNA by the Cterminal domain of histone H-1. A circular-dichroism study. Biophys.Chem. 1985, 22: 125-129.
    [94] Mai VQ, Chen X, Hong R, Huang L.Small abundant DNA binding proteins from the thermoacidophilic archaeon Sulfolobus shibatae constrain negative DNA supercoils. J Bacteriol. 1998, 180: 2560-2563.
    [95] Grayling RA, Bailey KA, Reeve JN.DNA binding and nuclease protection by the HMf histones from the hyperthermophilic archaeon Methanothermus fervidus. Extremophiles. 1997, 1: 79-88.
    [96] Weng L, Liu D, Li Y, et al. An archaeal histone-like protein as an efficient DNA carrier in gene transfer. Biochim Biophys Acta. 2004, 1702: 209-216.
    [97] Dirk E, Hiroshi A, Atsushi Y, et al. A hyperthermostable bacterial histone-like protein as an efficient mediator for transfection of eukaryotic cells. Natura Biotechnology. 2000, 18: 1211-1213.
    [98] Hariton-Gazal E, Rosenbluh J, Graessmann A, et al. Direct translocation of histone molecules across cell membranes . J Cell Sci. 2003, 116: 4577-4586.
    [99] Rosenbluh J, Hariton-Gazal E, Dagan A et al. Translocation of histone proteins across lipid bilayers and Mycoplasma membranes. J Mol Biol. 2005, 345: 387-400.
    [100] Fritz JD, Herweijer H, Zhang G.F, et al. Gene transfer into mammalian cells using histone-condensed plasmid DNA. Hum Gene Ther. 1996, 7: 1395-1404.
    [101] Zaitsev S, Buchwalow I, Haberland A, et al. Histone H1-mediated transfection: role of calcium in the cellular uptake and intracellular fate of H1-DNA complexes. Acta Histochem. 2002, 104: 85-92.
    [102] Puebla I, Esseghir S, Mortlock A, et al. A recombinant H1 histone-based system for efficient delivery of nucleic acids. J Biotechnol. 2003, 105: 215-226.
    [103] Kott M, Haberland A, Zaitsev S, et al. A new efficient method for transfection of neonatal cardiomyocytes using histone H1 in combination with DOSPER liposomal transfection reagent. Somat. Cell Mol. Genet. 1998, 24: 257-261.
    [104] Balicki D, Putnam CD, Scaria PV, et al. Structure and function correlation in histone H2A peptide-mediated gene transfer. Proc Natl Acad Sci. 2002, 99: 7467-7471.
    [105] Demirhan I, Hasselmayer O, Chandra A, et al. Histone-mediated transfer and expression of the HIV-1 tat gene in Jurkat cells. J Hum Virol. 1998, 1: 430-440.
    [106] Balicki D, Beutler E. Histone H2A significantly enhances in vitro DNA transfection. Mol Med. 1997, 3: 782-787.
    [107] Filion MC, Phillips NC. Toxicity and immunomodulatory activity of liposomal vectors formulated with cationic lipids toward immune effector cells. Biochim. Biophys Acta, 1997, 1329: 345-356.
    [108] Lappalainen K, Jaaskelainen I, Syrjanen K, et al. Comparison of cell proliferation and toxicity assays using two cationic liposomes. Pharm Res, 1994, 11: 1127-1131.
    [109] Iborra S, Soto M, Carrion J, et al. Vaccination with a plasmid DNA cocktail encoding the nucleosomal histones of Leishmania confers protection against murine cutaneous leishmaniosis .Vaccine, 2004, 22: 3865-3876.
    [110] Singh M. Transferrin. A targeting ligand for liposomes and anticancer drugs. Curr Pharm Des. 1999, 5: 443-51.
    [111] Wagner E, Zenke M, Cotten M, et al. Transferrin-polycation conjugates as carriers for DNA uptake into cells. Proc Natl Acad Sci. 1990, 87: 3410-3414.
    [112] Prata MI, Sa ntos AC, Torres S, et al. Targeting of lanthanide (III) chelates of DOTA-type glycoconjugates to the hepatic asyaloglycoprotein receptor: cell internalization and animal imaging studies. Contrast Media Mol Imaging. 2006, 1:246-258.
    [113] Oishi M, Kataoka K, Nagasaki Y. pH-responsive three-layered PEGylated polyplex micelle based on a lactosylated ABC triblock copolymer as a targetable and endosome-disruptive nonviral gene vector. Bioconjug Chem. 2006, 17: 677-688.
    [114] Kwon AH, Inoue T, Ha-Kawa SK. Characterization of the asialoglycoprotein receptor under hypoxic conditions in primary cultured rat hepatocytes. J Nucl Med. 2005, 46: 321-325.
    [115] Huckett B, Aeiatti M, Hawtrey AO. Evidence for targeted gene transfer by receptor-mediated endocytosis: stable expression following insulin-directed entry ofneo into HepG2 cells. Biochem Pharmacol. 1990, 40: 253-263.
    [116] Rosenkranz AA, Yachmenev SV, Jans DA, et al. Receptor-mediated endocytosis and nulear transport of a transfecting DNA construct. Exp Cell Res. 1992, 199: 323-329.
    [117] Decreased Human Milk Concentration of Epidermal Growth Factor After Preterm Delivery of Intrauterine Growth-restricted Newborns. J Pediatr Gastroenterol Nutr. 2007, 44: 464-467.
    [118] Cristiano RJ, Roth JA. Epidermal growth factor mediated DNA delivery into lung cancer cells via the epidermal growth factor receptor. Cancer Gene Ther 1996, 3: 4-10.
    [119] Haberland A, Dalluge R, Zaitsev S, et al. Ligand-histone H1 conjugates: increased solubility of DNA complexes, but no enhanced transfection activity. Somat Cell Mol Genet, 1999, 25: 237–245.
    [120] Dai FH, Chen Y, Ren CC, et al. Construction of an EGF receptor-mediated histone H1 (0)-based gene delivery system. J Cancer Res Clin Oncol, 2003, 129: 456–462.
    [121] Li S and Huang L. Nonviral gene therapy: promises and challenges. Gene Therapy. 2000, 7: 31-34.
    [122] Junbo H, Li Q, Zaide W, et al. Receptor-mediated interleukin-2 gene transfer into human hepatoma cells. Int J Mol Med, 1999, 3: 601-608.
    [123] 王秋岩 蛋白质半合理设计改变古菌 Aeropyrum pernix K1 酯酶\酰基氨酰肽酶的底物特异性研究. 2006.
    [124] C. R. Woese., O. Kandler., M.L. Wheelis. Towards a natural system of organisms- proposal for the domains Archaea, Bacteria and Eukarya. Proc Natl Acad Sci USA 1990, 87: 4576-4579.
    [125] P. Klenk., R.A. Clayton., J.F. Tomb. et al. The complete genome sequence of the hyperthermophilic, sulphate-reducing archaeon Archaeoglobus fulgidus. Nature. 1997, 390:364-370.
    [126] P. Klenk., R.A. Clayton., J.F. Tomb. et al. The complete genome sequence of the hyperthermophilic, sulphate-reducing archaeon Archaeoglobus fulgidus. Nature. 1997, 390: 364-370.
    [127] Kawarabayasi., M. Sawada., H. Horikawa. et al.: Complete sequence and gene organization of the genome of a hyper- thermophilic Archaebacterium, Pyrococcus horikoshii OT3. DNA Res. 1998, 5: 55-76.
    [128] FitzGibbon., A.J. Choi., J.H. Miller, et al. A fosmid-based genomic map and identification of 474 genes of the hyperthermo -philic archaeon Pyrobaculum aerophilum. Extremophiles .1997, 1: 36-51.
    [129] Kawarabayasi Y, Sawada M, Horikawa H, et al. Complete sequence and gene organization of the genome of a hyper-thermophilic archaebacterium, Pyrococcus horikoshii OT3. DNA Res. 1998, 5: 55-76.
    [130] Weng L, Feng Y, Ji X, et al. Recombinant expression and characterization of an extremely hyperthermophilic archaeal histone from Pyrococcus horikoshii OT3 . Protein Expr Purif. 2004, 33: 145-152.
    [131] Decanniere K, Babu AM, K Sandman, et al. Crystal structures of recombinant histones HMfA and HMfB from the hyperthermophilic archaeon Methanothermus fervidus. J Mol Biol. 2000, 303: 35-47.
    [132] Starich MR, Sandman K, Reeve JN, et al. NMR structure of HMfB from the hyperthermophile, Methanothermus fervidus, confirms that this archaeal protein is a histone. J Mol Biol. 1996, 255: 187-203.
    [133] Li T, Sun F, Ji X, et al. Structure based hyperthermostability of archaeal histone HphA from Pyrococcus horikoshii. J.Mol.Biol. 2003, 325: 1031-1037.
    [134] Weng L, Liu D, Li Y, et al. An archaeal histone-like protein as an efficient DNA carrier in gene transfer . Biochim Biophys Acta. 2004, 1702: 209-216.
    [135] Wang, F., et al. Radiation-induced apoptosis of two Nasopharyngeal carcinoma cell lines. Chinese J. oncol. 1998, 20: 119-121.
    [136] Nagayama Y, Yokoi H, Takeda K, et al. Adenovirus-mediated tumor suppressor p53 gene therapy for anaplastic thyroid carcinoma in vitro and in vivo. J Clin Endocrinol Metab. 2000, 85: 4081-4086.
    [137] Lappalainen K, Jaaskelainen I, Syrjanen K, et al. Comparison of cell proliferation and toxicity assays using two cationic liposomes. Pharm Res. 1994, 11: 1127-1131.
    [138] Baake M, Bauerle M, Doenecke D, et al. Core histones and linker histones are imported into the nucleus by different pathways. Eur J Cell Biol. 2001, 80:669-677.
    [139] Baake M, Doenecke D, Albig W. Baake M, Characterisation of nuclear localizations signals of the four human core histones. J Cell Biochem. 2001, 81: 333-346.
    [140] Johnson-Saliba M, Siddon NA, Clarkson MJ, et al.Distinct importin recognition properties of histones and chromatin assembly factors. FEBS Lett. 2000, 467: 169-174.
    [141] Takakura Y, Nishikawa M, Yamashita F, et al. Development of gene drug delivery systems based on pharmacokinetic studies. Eur J Pharm Sci. 2001,13: 71-76.
    [142] Mahato RI, Takakura Y, Hashida M. Nonviral vectors for In vivo gene delivery: Physicochemical and pharmacokinetic considera-tions. Crit. Rev. Ther. Drug Carrier Syst. 1997, 14: 133–172.
    [143] 金伯泉. 细胞和分子免疫学实验技术.第四军医大学出版社 2003.
    [144] 朱立平,陈学清.免疫学常用实验方法.人民军医出版社 2000.
    [145] Carol AG and Leslie Z B. Pharmacokinetics and protein therapeutics. Advanced Drug Delivery Reviews .1990, 4: 359-386.
    [146] Matsukawa M, Takeda K, Shima H, et al. Enzyme-linked immunosorbent assay for TA-2005-glucuronide in human plasma. Journal of Pharmaceutical and Biomedical Analysis.1998, 17: 245–254.
    [147] Takakura Y, Fujita T, Hashida M, et al. Disposition characteristics of macromolecules in tumor-bearing mice. Pharm Res. 1990, 7: 339-346.
    [148] Nishida K, Mihara K, Takino T, et al.Hepatic disposition characteristics of electrically charged macromolecules in rat in vivo and in the perfused liver. Pharm Res. 1991, 8: 437-444.
    [149] Yamasaki Y, Sumimoto K, Nishikawa M, et al. Pharmacokinetic analysis of in vivo disposition of succinylated proteins targeted to liver nonparenchymal cells via scavenger receptors: importance of molecular size and negative charge density for in vivo recognition by receptors.J Pharmacol Exp Ther. 2002, 301: 467-477.
    [150] Hahida M, Mahato R I, Miyao T, et al. Pharmacokinetics and targeted delivery of proteins and genes. J control release. 1996, 41: 91-97.
    [151] Hiroyuki K, Hidetaka A, Hideyoshi A. Pharmacokinetic and pharmacodynamic considerations in gene therapy. Drug Discovery Today. 2003, 8: 990-996.
    [152] Takakura Y, Nishikawa M, Yamashita F, Hashida M. Development of gene drug delivery systems based on pharmacokinetic studies. European Journal of Pharmaceutical scieces. 2001, 13: 71-76.
    [153] Torchilin VP, Lukyanov AN, Peptide and protein drug delivery to and into tumors: challenges and solutions, Drug Discovery Today. 2003, 8: 259-266.
    [154] Christopher MW, Middaugh CR. Barriers to nonviral gene delivery. J Pharm Sci. 2003, 92: 203-217.
    [155] Zhou Hs, Liu DP, Liang CC. Chanllenges and Strategies: the immune responses in gene therapy. Med Res Rev, 2004, 24: 748-761.
    [156] BenMohamed L, Bertrand G, McNamara CD, et al. Identification of novel immunodominant CD4+ Th1-type T-cell peptide epitopes from herpes simplex virus glycoprotein D that confer protective immunity. J Virol. 2003, 77: 9463-9473.
    [157] Lowenstein PR. Virology and immunology of gene therapy, or virology and immunology of high MOI infection with defective viruses. Gene Ther. 2003, 10: 933-934.
    [158] McCormack JE, Martineau D, DePolo N, Maifert S, Akbarian L, Townsend K, LeeW, Irwin M, Sajjadi N, Jolly DJ, Warner J. Anti-vector immunoglobulin induced by retroviral vectors. Hum Gene Ther. 1997, 8: 1263-1273.
    [159] Brockstedt DG, Podsakoff GM, Fong L, Kurtzman G, Mueller-RuchholtzW, Engleman EG. Induction of immunity to antigens expressed by recombinant adeno-associated virus depends on the route of administration. Clin Immunol. 1999, 92: 67-75.
    [160] Krieg AM, Yi AK, Matson S, et al. CpG motifs in bacterial DNA trigger direct B-cell activation. Nature. 1995, 374: 546-549.
    [161] Yasuda K, Ogawa Y, Kishimoto M, Takagi T, Hashida M, Takakura Y. Plasmid DNA activates murine macrophages to induce inflammatory cytokines in a CpGmotif-independent manner by complex formation with cationic liposomes. Biochem Biophys Res Commun. 2002, 293: 344-348.
    [162] Shoda LK, Kegerreis KA, Suarez CE, Mwangi W, Knowles DP, Brown WC. Immunostimulatory CpGmodified plasmidDNAenhances IL-12, TNF-alpha, and NOproduction by bovine macrophages. J Leukoc Biol. 2001, 70: 103-112.
    [163] Krieg AM. The role of CpG motifs in innate immunity. Curr Opin Immunol. 2000; 12: 35-43.
    [164] Tan Y, Li S, Pitt BR, Huang L. The inhibitory role of CpG immunostimulatory motifs in cationic lipid vector-mediated transgene expression in vivo. Hum Gene Ther. 1999, 10: 2153-2161.

© 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