用户名: 密码: 验证码:
VEGF-C和Raf-1基因共沉默对人肺癌细胞株A549体外生长影响的研究
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:观察VEGF-C和RAF-1基因共沉默对人肺癌细胞A549增殖、侵袭、细胞周期等的影响,并初步探讨其机制。
     方法:构建靶向VEGF-C和RAF-1基因的shRNA,酶切鉴定、测序正确后,转染A549细胞,分别采用RT-PCR和Western blot方法检测mRNA和蛋白表达,选择有效干扰序列,并筛选出稳定表达shRNA的细胞系。将VEGF-C和RAF-1构建到同一个质粒载体,筛选出稳定表达双shRNA的细胞系,并观察其抑制效果。MTT法检测细胞增殖能力,Boyden小室检测细胞侵袭能力,流式细胞仪(FCM)检测蛋白表达、细胞周期及细胞凋亡水平。Western blot方法检测相关信号通路Caspase3及PDK1、AKT、RAF和ERK磷酸化水平情况。
     结果:成功构建靶向VEGF-C和RAF-1基因的有效shRNA,并筛选出稳定转染细胞系A549-VEGF-Ce和A549-RAF-1e,检测mRNA和蛋白表达明显受到抑制。成功构建联合表达VEGF-C和RAF-1基因的有效shRNA表达载体,并筛选出稳定转染细胞系A549-RAF-1-VEGF-Ce,检测mRNA和蛋白表达与单独VEGF-C和RAF-1shRNA作用一致。MTT法检测结果表明A549-Raf-1-VEGF-Ce与A549-Raf-le以及A549-VEGF-Ce的增殖能力与对照组细胞A549相比明显降低。A549-Raf-le增殖的平均值较A549-VEGF-Ce略低,但无统计学差异。A549-Raf-1-VEGF-Ce的增殖能力与A549-VEGF-Ce相比显著下降。体外趋化侵袭实验显示,A549-Raf-le组、A549-VEGF-Ce组以及A549-Raf-1-VEGF-Ce组的侵袭细胞数比A549组和各无效shRNA组侵袭细胞数减少,各实验组之间差异无显著性。检测细胞周期分布发现,A549-VEGF-Ce细胞其G1期细胞比例高于其他各组细胞,而A549-Raf-le组细胞出现了凋亡峰。A549-Raf-1-VEGF-Ce组细胞G1期的细胞与A549-VEGF-Ce组相比减少的,其凋亡明显增高。进一步检测各实验组细胞的凋亡水平,A549-Raf-1-VEGF-Ce细胞的凋亡率(24%)高于A549-Raf-le细胞的凋亡率(17%),其他各组细胞凋亡率无明显变化,与细胞周期分析结果相一致。Western blot结果显示在A549-Raf-1-VEGF-Ce和A549-Raf-le中能检测到活化的Caspase3大亚基片段,各组细胞中AKT、PDK1、RAF的磷酸化水平无明显变化,A549-Raf-1-VEGF-Ce细胞ERK的磷酸化水平有较明显的下降。
     结论:稳定转染靶向VEGF-C和RAF-1基因的shRNA能够抑制A549细胞的增殖与体外侵袭能力,并调控细胞周期分布及细胞凋亡。同时干扰RAF-1和VEGF-C有一定的协同效应,与Caspase3途径及ERK的磷酸化水平增高有关。
Objective To explore the effects and possible mechanisms of RNA interference targeting VEGF-C and RAF-1 mRNA on the proliferation, invasion and cell cycle of human lung cancer cell line A549.
     Methods According to the basic principle of RNA interference, plasmid which expressed short hairpin RNA (shRNA) corresponding to VEGF-C and RAF-1 were respectively constructed by DNA recombining technology. After restriction endonuclease digestion and sequence were identified, the plasmid was transfected into A549 cell. Then the expression of mRNA and protein was detected by RT-PCR and Western blot. According to the result, the effective sequence was selected and the stable cell line which expressed shRNA was screened out. Plasmid vector with both VEGF-C and RAF-1 were constructed, and the stable cell line transfected with that were built. MTT was used to test the cell growth, Boyden methods was used to dectect the the invasion ability of A549 cell, flowcytometry was used to observe the expression level of protein, cell cycle and apoptosis, and Western blot was using to test the the level of associated pathway such as Caspase3, PDK1, AKT, RAF and ERK phosphorylation.
     Results The expressions of mRNA and protein of A549-VEGF-Ce and A549-RAF-le cell line which was transfected with shRNA targeting VEGF-C or RAF-1 gene were significantly inhibited. The inhibited effect of A549-RAF-1-VEGF-Ce cell line which contained two kinds of shRNA was same as A549-VEGF-Ce or A549-RAF-le cell line. According to MTT results, the cell proliferation ability of A549-Raf-1-VEGF-Ce, A549-Raf-le and A549-VEGF-Ce cell lines were significantly lower than the control group.
     The average number of A549-RAF-le cell line was a little lower than that of A549-VEGF-Ce, but there was no significant difference between them. Nevertheless, the proliferation ability of A549-Raf-1-VEGF-Ce cell was dramaticly lower than that of A549-VEGF-Ce. In the invasion test in vitro, the invasion cell number of A549-Raf-le, A549-VEGF-Ce and A549-Raf-1-VEGF-Ce was lower than control cell group, but there were no significant difference among them. In cell cycle tested by flowcytometry, the proportion of cell number in G1 phase of A549-VEGF-Ce was higher than other cell groups and a apoptosis peak was emerged in A549-Raf-le group.
     The A549-Raf-1-VEGF-Ce cell line with lower proportion of G1 phase cell than A549-VEGF-Ce had obviously more apoptosis cell. Further examination about the apoptosis level of all cell lines showed that the apoptosis ratio of A549-Raf-1-VEGF-Ce (24%) was higher than that of A549-Raf-le (17%) and the ration of other groups which kept in line with cell cycle test had no significant changes. Western blot results showed that actived large subunit of Caspase3 was caught both in 549-Raf-1-VEGF-Ce and A549-Raf-le group, and the phosphorylation level of AKT, PDK1 and RAF in all cell groups had no significant difference exception of the obvious decrease of phosphorylation level of ERK in A549-Raf-1-VEGF-Ce cell line.
     Conclusion shRNA targeting VEGF-C and RAF-1 gene could inhibit the proliferation and invasion abilities of A549 cell and regulate the cell cycle and apoptosis of tumor cell. And RNA interference targeting on VEGF-C and RAF-1 gene simultaneously had a cooperative effect mainly due to the increased level of Caspase3 and phosphorylation form of ERK.
引文
1. Marcel FA, David A, M. JG, et al. The use of gene array technology and proteomics in the search of new targets of diseases for therapeutics. Toxicology Letters, 2009,186(1):45-51.
    2. Moitreyee CK, Christopher PM. Exploring the sounds of silence: RNAi-mediated gene silencing for target identification and validation. Drug Discovery Today, 2005, 10(22):1559-1565.
    3. Ji L, Nicole LS, Stephen JE. Principles of Cancer Therapy: Oncogene and Non-oncogene Addiction. Cell, 2009,136(5):823-837.
    4. Simon WJ, Patricia MS, Mark AL. siRNA for gene silencing: a route to drug target discovery. Current Opinion in Pharmacology, 2004,4(5):522-527.
    5. Martimprey H, Vauthier C, Malvy C, et al. Polymer nanocarriers for the delivery of small fragments of nucleic acids: Oligonucleotides and siRNA. European Journal of Pharmaceutics and Biopharmaceutics, 2009,71(3): 490-504.
    6. Jerry CC, Theodore BM, Kathleen MS. RNA interference and human disease. Molecular Genetics and Metabolism, 2003, 80(1-2):121-128.
    7. Wang XL, Xu Rz, Lu ZR. A peptide-targeted delivery system with pH-sensitive amphiphilic cell membrane disruption for efficient receptor-mediated siRNA delivery. Journal of Controlled Release, 2009,134(3): 207-213.
    8. Ray KML, Paul AW. RNA interference: From gene silencing to gene-specific therapeutics. Pharmacology & Therapeutics, 2005,107(2):222-239.
    9. Younghoon K, Manorama T, David JP, et al. Polymersome delivery of siRNA and antisense oligonucleotides. Journal of Controlled Release, 2009,134(2):132-140.
    10. Markus H, Vittorio Z, Mark H, et al. Preparation of small amounts of sterile siRNA-liposomes with high entrapping efficiency by dual asymmetric centrifugation (DAC). Journal of Controlled Release, 2009,135(1) :80-88.
    11. Daniela R, Andreas Z. Drug delivery of siRNA therapeutics: potentials and limits of nanosystems. Nanomedicine: Nanotechnology, Biology and Medicine, 2009, 5(1):8-20.
    
    12. Anthony JC, Danielle SWB, Craig LD, et al. Development of a novel endosomolytic diblock copolymer for siRNA delivery. Journal of Controlled Release, 2009,133(3):221-229.
    13. Yogesh P, Jayanth P. Polymeric nanoparticles for siRNA delivery and gene silencing. International Journal of Pharmaceutics, 2009, 367(l-2):195-203.
    14. Kentaro O, Kenichi Y, Shunsuke O, et al. Nonviral delivery of siRNA into mesenchymal stem cells by a combination of ultrasound and microbubbles. Journal of Controlled Release, 2009,133(2)146-153.
    15. Juliane N, Terry WJS, Olivia M, et al. Fast degrading polyesters as siRNA nano-carriers for pulmonary gene therapy. Journal of Controlled Release, 2008, 132(3):243-251.
    16. Elias F, Amelie B. State of the art and perspectives for the delivery of antisense oligonucleotides and siRNA by polymeric nanocarriers. International Journal of Pharmaceutics, 2008, 364(2):237-248.
    17. Jonathan KW, Glen FD, Masad JD. Chemically modified siRNA: tools and applications.Drug Discovery Today, 2008,13(19-20):842-855.
    18. David AC, Cai S, Diana MB, et al. Polymersome carriers: From self-assembly to siRNA and protein therapeutics. European Journal of Pharmaceutics and Biopharmaceutics, 2009,71(3):463-474.
    19. Suzanne S, Summer S, Vasant J, et al. Examination of real-time polymerase chain reaction methods for the detection and quantification of modified siRNA. Analytical Biochemistry, 2008, 379(1) :96-104.
    1. Ji L, Nicole LS, Stephen JE. Principles of Cancer Therapy: Oncogene and Non-oncogene Addiction. Cell, 2009,136(5):823-837.
    2. Marcel FA, David A, M. JG, et al.The use of gene array technology and proteomics in the search of new targets of diseases for therapeutics. Toxicology Letters, 2009,186(1):45-51.
    3. Martimprey H, Vauthier C, Malvy C, et al. Polymer nanocarriers for the delivery of small fragments of nucleic acids: Oligonucleotides and siRNA. European Journal of Pharmaceutics and Biopharmaceutics, 2009, 71(3): 490-504.
    4. Moitreyee CK, Christopher PM. Exploring the sounds of silence: RNAi-mediated gene silencing for target identification and validation. Drug Discovery Today, 2005,10(22):1559-1565.
    5. Ray KML, Paul AW. RNA interference: From gene silencing to gene-specific therapeutics. Pharmacology & Therapeutics, 2005,107(2):222-239.
    6. Su J, Chen X, Takuro K. A CD147-targeting siRNA inhibits the proliferation,invasiveness, and VEGF production of human malignant melanoma cells by down-regulating glycolysis. Cancer Letters, 2009, 273(1):140-147.
    7. Troy AM, Paul RC, Nathan MN, et al. Functional differences between adult and neonatal stem cells are abolished by VEGF siRNA. Journal of the American College of Surgeons, 2008, 207(3):S71.
    8. Sun HK, Ji HJ, Soo HL, et al. Local and systemic delivery of VEGF siRNA using polyelectrolyte complex micelles for effective treatment of cancer. Journal of Controlled Release, 2008,129(2):107-116.
    9. Naoyuki M, Yuuki T, Kosei T, et al. Anti-tumor effects of anti-VEGF siRNA encapsulated with PLGA microspheres in mice.Journal of Controlled Release, 2008,126(3):246-254.
    10. Soo HL, Sun HK, Tae GP. Anticancer effect by intratumoral and intravenous administration of VEGF siRNA polyelectrolyte complex micelles.Journal of Biotechnology, 2007,131(2):S48-S49.
    11. Sun HK, Ji HJ, Soo HL, et al.PEG conjugated VEGF siRNA for anti-angiogenic gene therapy. Journal of Controlled Release, 2006,116(2):123-129.
    12. Esther R, Annabelle V, Tilman S, et al. siRNA targeting VEGF inhibits hepatocellular carcinoma growth and tumor angiogenesis in vivo. Journal of Hepatology, 2008, 49(6): 977-984.
    13. Nithya R, Alex A. Inhibitors of Raf kinase and MEK signaling. Update on Cancer Therapeutics, 2007, 2(3):111-118.
    14. Eijiro O, Unnur PT. Different regulation of vascular endothelial growth Factor expression by the ERK and p38 kinase pathways in v-ras, v-raf, and v-myc transformed cells. Biochemical and Biophysical Research Communications, 2000,270(1):108-111.
    15. Pan MR, Chang HC, Hung WC. Non-steroidal anti-inflammatory drugs suppress the ERK signaling pathway via block of Ras/c-Raf interaction and activation of MAP kinase phosphatases. Cellular Signalling, 2008, 20(6):1134-1141.
    16. Wang XL, Xu Rz, Lu ZR. A peptide-targeted delivery system with pH-sensitive amphiphilic cell membrane disruption for efficient receptor-mediated siRNA delivery. Journal of Controlled Release, 2009,134(3): 207-213.
    17. Younghoon K,Manorama T, David JP, et al. Polymersome delivery of siRNA and antisense oligonucleotides. Journal of Controlled Release, 2009,134(2):132-140.
    18. Fatih C, Semra C, Christiaan K,et al. Disruption of Tumor Cell Adhesion Promotes Angiogenic Switch and Progression to Micrometastasis in RAF-Driven Murine Lung Cancer. Cancer Cell, 2007,12(2):145-159.
    19. Evan TK, Zheng F, Kam Y, et al. Raf kinase inhibitor protein: a prostate cancer metastasis suppressor gene. Cancer Letters, 2004, 207(2):131-137.
    20. Kathleen MWI,Navdeep KG, Sonja MM,et al. Loss of Raf Kinase Inhibitory Protein Induces Radioresistance in Prostate Cancer. International Journal of Radiation Oncology Biology Physics, 2008, 72(1):153-160.
    21. Kathryn EM, Catrin AP. Raf proteins and cancer: B-Raf is identified as a mutational target. Biochimica et Biophysica Acta (BBA) - Reviews on Cancer,2003,1653(1): 25-40.
    22. Samar A, Ulf RR, Petra H. Validation of a liquid chromatography assay for the quantification of the Raf kinase inhibitor BAY 43-9006 in small volumes of mouse serum. Journal of Chromatography B, 2004, 809(1):99-103.
    23. William MO, John BS, Stephane H, et al. Functional Proteomics Identifies Targets of Phosphorylation by B-Raf Signaling in Melanoma. Molecular Cell, 2009,
    24. Mathew JG, Richard M. Guilty as charged: B-RAF is a human oncogene. Cancer Cell, 2004, 6(4):313-319.
    25. David HB, Lori A.J, Hong C, et al. Activation of Raf-1 in Human Pancreatic Adenocarcinoma.Journal of Surgical Research, 1997, 69(1):199-204.
    26. Gillis HC, Pinchot SN, Chen H. The Synergistic Effects of Octreotide and RAF-1 Actvation in Carcinoid Cancer Cells. Journal of Surgical Research, 2009,151(2):249.
    27. Joshua DH, Jonas G, Brad N, et al. Potent and selective pyrazole-based inhibitors of B-Raf kinase. Bioorganic & Medicinal Chemistry letters, 2008, 18(16):4692-4695.
    28. Wang B, Feng Y, Song X, et al. Involvement of ERK, Bcl-2 family and caspase 3 in recombinant human activin A-induced apoptosis in A549. Toxicology, 2009,258(2-3):176-183.
    29. Eun YS, Navneet K, Mi YP, et al. Synthesis of amide and urea derivatives of benzothiazole as Raf-1 inhibitor. European Journal of Medicinal Chemistry, 2008,43(7):1519-1524.
    30. Caroline RWO, William LB, Linda SS, et al. The Raf signal transduction cascade as a target for chemotherapeutic intervention in growth factor-responsive tumors.Pharmacology & Therapeutics, 2000, 88(3):229-279.
    31. Pan MR, Chang HC, Hung WC. Non-steroidal anti-inflammatory drugs suppress the ERK signaling pathway via block of Ras/c-Raf interaction and activation of MAP kinase phosphatases. Cellular Signalling, 2008, 20(6):1134-1141.
    32. Nithya R, Alex A. Inhibitors of Raf kinase and MEK signaling. Update on Cancer Therapeutics, 2007, 2(3):111-118.
    33. Jared AG, Scott W, Chris C, et al.Role of Raf Kinase in Cancer: Therapeutic Potential of Targeting the Raf/MEK/ERK Signal Transduction Pathway. Seminars in Oncology, 2006, 33(4): 392-406.
    34. James AM, Linda SS, William HC, et al. Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, 2007, 1773(8):1263-1284.
    1. Simon WJ, Patricia MS, Mark AL. siRNA for gene silencing: a route to drug target discovery. Current Opinion in Pharmacology, 2004, 4(5):522-527.
    2. Ji L, Nicole LS, Stephen JE. Principles of Cancer Therapy: Oncogene and Non-oncogene Addiction. Cell, 2009,136(5):823-837.
    3. Marcel FA, David A, M. JG, et al. The use of gene array technology and proteomics in the search of new targets of diseases for therapeutics. Toxicology Letters, 2009,186(1):45-51.
    4. Martimprey H, Vauthier C, Malvy C, et al. Polymer nanocarriers for the delivery of small fragments of nucleic acids: Oligonucleotides and siRNA. European Journal of Pharmaceutics and Biopharmaceutics, 2009, 71(3): 490-504.
    5. Moitreyee CK, Christopher PM. Exploring the sounds of silence: RNAi-mediated gene silencing for target identification and validation. Drug Discovery Today, 2005,10(22): 1559-1565.
    6. Jerry CC, Theodore BM, Kathleen MS. RNA interference and human disease.Molecular Genetics and Metabolism, 2003, 80(1-2): 121-128.
    7. Ray KML, Paul AW. RNA interference: From gene silencing to gene-specific therapeutics. Pharmacology & Therapeutics, 2005,107(2):222-239.
    8. Patrick YL, Frank X, Martin CW. In Vivo Application of RNA Interference: From Functional Genomics to Therapeutics. Advances in Genetics, 2005, 54:115-142.
    9. Stephen JT, Patricia AK. The genesis of RNA interference, its potential clinical applications, and implications in gynecologic cancer. Gynecologic Oncology, 2005,99(3):736-741.
    10. Susan LU.The therapeutic potential of RNA interference. FEBS Letters, 2005,579(26):5996-6007.
    11. Helmuth HG, Arts GJ. Biology calls the targets: combining RNAi and disease biology.Drug Discovery Today, 2005,10(20):1385-1391
    12. Takeshi H, Taketoshi M, Nobutake Y,et al. SiRNAs do not induce RNA-dependent transcriptional silencing of retrovirus in human cells.FEBS Letters, 2007,581(25):4949-4954.
    13. Reda HS, Yves L, Nicolas L,et al. Short interfering RNA (siRNA), a novel therapeutic tool acting on angiogenesis. Biochimie, 2007, 89(10):1234-1244.
    14. Zhang BH, Pan XP, George PC,et al. microRNAs as oncogenes and tumor suppressors.Developmental Biology, 2007,302(1):l-12.
    15. Steven PO, Justin LM, Patrick LS, et al. MicroRNAs: Key Modulators of Posttranscriptional Gene Expression. Gastroenterology, 2009,136(1):17-25.
    16. Mohammed A, Heike A. MicroRNAs as regulatory molecules in cancer: a focus on models defining miRNA functions. Drug Discovery Today: Disease Models,2009,3
    17. Eric AH, Ann MG, Sapna KD. Direct detection and quantification of microRNAs.Analytical Biochemistry, 2009, 387(1):1-12.
    18. Mirnezami AHF, Pickard K, Zhang L, et al. MicroRNAs: Key players in carcinogenesis and novel therapeutic targets. European Journal of Surgical Oncology (EJSO), 2009, 35(4):339-347.
    19. Wolfgang MS, Alexander OS, Bernd J, et al. In vivo profile of the human leukocyte microRNA response to endotoxemia. Biochemical and Biophysical Research Communications, 2009, 380(3):437-441.
    20. Zhu G, Gilchrist R, Borley N, et al. Reduction of TSG101 protein has a negative impact on tumor cell growth. Int J Cancer, 2004,109(4):541-7.
    21. Hyunsuk S, Shinya O, Nobutaka F. Chemokine receptor CXCR4 as a therapeutic target for neuroectodermal tumors. Seminars in Cancer Biology, 2009, 19(2):123-134.
    22. Hart S, Fischer OM, Ullrich A. Cannabinoids induce cancer cell proliferation via tumor necrosis factor alpha converting enzyme (TACE/ADAM17)-mediated transactivation of the epidermal growth factor receptor. Cancer Res, 2004, 64(6):1943-50.
    23. Anastasia M, Olivia M, Ludger F, et al. In vivo pharmacokinetics, tissue distribution and underlying mechanisms of various PEI(-PEG)/siRNA complexes.Toxicology and Applied Pharmacology, 2009, 236(1): 97-108.
    24. Yan K, Ruan Lf, Ma LL,et al. RNA interference as a novel and powerful tool in immunopharmacological research. International Immunopharmacology, 2007,7(4):417-426.
    25. Yague E, Higgins CF, Raguz S.Complete reversal of multidrug resistance by stable expression of small interfering RNAs targeting MDR1. Gene Ther, 2004,11(14):1170-4.
    26. Clara M, Jeff S. Targeting the RAF-MEK-ERK pathway in cancer therapy. Cancer Letters, Available online 12 February 2009.
    27. Lekha DK, Alan RC. Gene manipulation through the use of small interfering RNA (siRNA): From in vitro to in vivo applications. Advanced Drug Delivery Reviews,2007, 59(2-3):87-100.
    28. Michael F, Guo F. MicroRNA biogenesis: there's more than one way to skin a cat. Biochimica et Biophysica Acta (BBA) - Gene Regulatory Mechanisms, 2008,1779(11):663-667.
    29. Christine CE, Brett PM. Therapeutic potential for microRNAs. Advanced Drug Delivery Reviews, 2007, 59(2-3):101-114.
    30. Eva S, Radek M, Jan S,et al. RNA regulation and cancer development. Cancer Letters, 2007, 246(1-2):12-23.
    31. Wu H,Hait W.N.,and Yang JM..Small interfering RNA-induced suppression of MDR1(P-glocoprotein)restores sensitivity to multidrug-resistant cancer cells.Cancer Res, 2003, 63(7):1515-9.
    32. Massimo M, Giorgia N, Stefano Let al. RNA interference: Implications for cancer treatment. Molecular Aspects of Medicine, 2007, 28(1): 143-166.
    33. Isabella F, Francesca RA, John C,et al. miR-155 gene: A typical multifunctional microRNA. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease,Available online 5 March 2009.
    34. Mathijs PV, Carlos S, Mariette S,et al. A Genetic Screen Implicates miRNA-372 and miRNA-373 As Oncogenes in Testicular Germ Cell Tumors. Cell, 2006,124(6): 1169-1181.
    35. Lu Y, Thomson JM, Ho YFW, et al. Transgenic over-expression of the microRNA miR-17-92 cluster promotes proliferation and inhibits differentiation of lung epithelial progenitor cells. Developmental Biology, 2007, 310(2):442-453.
    36. Lima RT, Martins LM, Guimaraes JE, et al. Specific downregulation of bcl-2 and xIAP by RNAi enhances the effects of chemotherapeutic agents in MCF-7 human breast cancer cells. Cancer Gene Ther, 2004,11(5):309-16.
    37. Suzanne MJ, Laura SS, Ester D, et al. ErbB2 and ErbB4 Cbl binding sites can functionally replace the ErbBl Cbl binding site. Cellular Signalling, 2009, 21(5):810-818.
    38. Zhang L, Yang N, Mohamed-Hadley A, et al. Vector-based RNAi, a novel tool for isoform-specific knock-down of VEGF and anti-angiogenesis gene therapy of cancer. Biochem Biophys Res Commun, 2003, 303(4): 1169-78.
    39. Jennifer LW, Thomas BT. Dicer is regulated by cellular stresses and interferons. Molecular Immunology, 2009, 46(6):1222-1228.
    40. Kathryn ZG, Amy DK, Jiri A,et al. Improving prediction of chemical carcinogenicity by considering multiple mechanisms and applying toxicogenomic approaches. Mutation Research/Reviews in Mutation Research, 2009,681(2-3) :230-240.
    41. Wilda M, Fuchs U, Wossmann W, et al. Killing of leukemia cells with a BCR/ABL fusion gene by RNA interference (RNAi). Oncogene, 2002, 21(37): 5716-24.
    42. Daniela R, Andreas Z. Drug delivery of siRNA therapeutics: potentials and limits of nanosystems. Nanomedicine: Nanotechnology, Biology and Medicine, 2009,5(1):8-20.
    43. Laura B. RNA-Based Therapeutics: Ready for Delivery? Cell, 2009,136(4): 581-584.
    44. Lin SL, Chuong CM, Ying SY. A Novel mRNA-cDNA interference phenomenon for silencing bcl-2 expression in human LNCaP cells. Biochem Biophys Res Commun, 2001, 28(3): 639-644.
    45. Ramiro G, Muller F, Amelia C, et al. MicroRNA expression and function in cancer. Trends in Molecular Medicine, 2006,12(12) :580-587.
    46. Piotr GR, Lindsey NJ, Buckminster JF,et al. RNA interference: Mechanisms of action and therapeutic consideration. Surgery, 2006,140(5):719-725.
    47. Dhananjay J, Kim JE, Rohidas A,et al. Aktl silencing efficiencies in lung cancer cells by sh/si/ssiRNA transfection using a reductable polyspermine carrier.Biomaterials, 2009, 30(8):1635-1647
    48. Rodrigo L, Anna SS, Sergio VA. Long intronic noncoding RNA transcription: Expression noise or expression choice? Genomics, 2009, 93(4):291-298.
    49. Zachary CH, Daniel MA, Andrea A. Adenovirus vector induced innate immune responses: Impact upon efficacy and toxicity in gene therapy and vaccine applications. Virus Research, 2008,132(1-2):1-14.
    50. Tetsuji H, Hiroyuki M, Kazufumi K,et al. RNA interference of PPARγ using fiber-modified adenovirus vector efficiently suppresses preadipocyte-to-adipocyte differentiation in 3T3-L1 cells. Gene, 2005, 348:157-165.
    51. Dirk G, Kusum P, Mark AK. Adeno-Associated Virus Vectors for Short Hairpin RNA Expression. Methods in Enzymology, 2005, 392:381-405.
    52. Laurence D, Martine M, Thiery JC, et al. Development of an in vivo adeno-associated virus-mediated siRNA approach to knockdown tyrosine hydroxylase in the lateral retrochiasmatic area of the ovine brain. Journal of Neuroscience Methods, 2008, 170(1):56-66.
    53. Liu CM, Liu DP, Dong WJ, et al. Retrovirus vector-mediated stable gene silencing in human cell. Biochemical and Biophysical Research Communications, 2004,313(3):716-720.
    54. Tanner M, Dyana S, Anne M, et al. Intensive RNAi with lentiviral vectors in mammalian cells. Methods, 2009, 47(4):298-303.
    55. Santhosh CV, Mayur CT, Rohan HK, et al. A lentiviral vector with novel multiple cloning sites: Stable transgene expression in vitro and in vivo. Biochemical and Biophysical Research Communications, 2008, 371(3):546-550.
    56. Nagy P, Arndt-Jovin DJ, Jovin TM. Small interfering RNAs suppress the expression of endogenous and GFP-fused epidermal growth factor receptor (erbBl) and induce apoptosis in erbBl-overexpressing cells. Exp Cell Res, 2003,15;285(l):39-49.
    57. Martinkova J, Gadher SJ, Hajduch M,et al. Challenges in cancer research and multifaceted approaches in cancer biomarker quest. FEBS Letters, Available online 25 March 2009
    58. Wang XL, Xu Rz, Lu ZR. A peptide-targeted delivery system with pH-sensitive amphiphilic cell membrane disruption for efficient receptor-mediated siRNA delivery. Journal of Controlled Release, 2009,134(3): 207-213.
    59. Younghoon K, Manorama T, David JP, et al. Polymersome delivery of siRNA and antisense oligonucleotides. Journal of Controlled Release, 2009,134(2): 132-140.
    60. Filleur S, Courtin A, Ait-Si-Ali S, et al. SiRNA-mediated inhibition of vascular endothelial growth factor severely limits tumor resistence to antiangiogenic thrombospondin-1 and slows tumor vascularization and growth. Cancer Res, 2003,63(14):3919-22.
    61. Chie KS, Hiromi S, Tadashi T, et al. Cancer Metastasis Is Accelerated through Immunosuppression during Snail-Induced EMT of Cancer Cells. Cancer Cell, 2009,15(3):195-206.
    62. Crnkovic-Mertens I, Hoppe-Seyler F, Butz K. Induction of apoptosis in tumor cells by siRNA-mediated silencing of the livin/ML-IAP/KIAP gene. Oncogene, 2003,22(51): 8330-6.
    63. Janusz R, Chloe M, Nathalie M,et al. Tissue factor in tumour progression. Best Practice & Research Clinical Haematology, 2009, 22 (1):71-83.
    64. Christiane N, Axel P, Alexandra S,et al. Modulation of the classical multidrug resistance (MDR) phenotype by RNA interference (RNAi). FEBS Letters, 2003,545(2-3): 144-150.
    65. Song E, Zhu P, Lee SK, et al. Antibody mediated in vivo delivery of small interfering RNAs via cell-surface receptors. Nat Biotechnol. 2005, 23(6):709-17.
    66. Cookson JC, Laughton CA. The levels of telomere-binding proteins in human tumours and therapeutic implications. European Journal of Cancer, 2009,45(4):536-550.
    67. Marja L, Maija B, Tuomas T, et al. VEGFs and receptors involved in angiogenesis versus lymphangiogenesis. Current Opinion in Cell Biology, Available online 21February 2009.
    68. Heidi AL, Madlaina B. Optimal targeting of the mTORCl kinase in human cancer.Current Opinion in Cell Biology, Available online 21 February 2009.
    69. Tali F, Revital A, Keren M, et al. Suppression of experimental autoimmune myasthenia gravis by inhibiting the signaling between IFN-γ inducible protein 10 (IP-10) and its receptor CXCR3. Journal of Neuroimmunology, Available online 20 February 2009.
    70. Sreenath VS, Jeffrey S. ErbBs in lung cancer. Experimental Cell Research, 2009,315(4): 557-571.
    71. Mariana L D, Emanuela M, Elizangela P, et al. Role of p53 in the induction of cyclooxygenase-2 by cisplatin or paclitaxel in non-small cell lung cancer cell lines.Cancer Letters , Available online 13 February 2009.
    72. Koen R, Roosmarijn EV, Joseph D,et al. Maintaining the silence: reflections on long-term RNAi. Drug Discovery Today, 2008,13(21-22):917-931.
    73. Candace KS, Li JL, Serguei PG. Adverse effects induced by short hairpin RNA expression in porcine fetal fibroblasts. Biochemical and Biophysical Research Communications, 2008, 370(1):113-117.
    74. Antonin DF, Tatiana N. siRNA and the lung: research tool or therapeutic drug? Current Opinion in Pharmacology, 2008, 8(3):280-285.
    75. Tara ML, Howell FM, Carl DN. Not miR-ly small RNAs: Big potential for microRNAs in therapy. Journal of Allergy and Clinical Immunology, 2008,121(2):309-319.
    76. John WP, Janice LP, Lee YS,et al. A Dicer-2-Dependent 80S Complex Cleaves Targeted mRNAs during RNAi in Drosophila. Cell, 2004,117(l):83-94.
    77. Yang M, Joerg M. Discovery, biology and therapeutic potential of RNA interference, microRNA and antagomirs. Pharmacology & Therapeutics, 2008,117(l):94-104.
    78. Andrei LG, Eugene SK. RNA interference in cancer. Biomolecular Engineering,2006, 23(1):17-34.
    79. Jonathan KW, Glen FD Masad JD. Chemically modified siRNA: tools and applications. Drug Discovery Today, 2008,13(19-20):842-855.
    80. Uta F, Arndt B. The Application of siRNA Technology to Cancer Biology Discovery. Advances in Cancer Research, 2006, 96:75-102.
    81. David AC, Cai SS, Diana MB, et al. Polymersome carriers: From self-assembly to siRNA and protein therapeutics. European Journal of Pharmaceutics and Biopharmaceutics, 2009, 71(3):463-474.
    82. Coral VAW. The dialectics of cancer: A theory of the initiation and development of cancer through errors in RNAi. Medical Hypotheses, 2006, 66(3):612-635.
    83. Aimee LJ, Peter SL. Noise amidst the silence: off-target effects of siRNAs? Trends in Genetics, 2004, 20(11):521-524.
    84. Katalin K, Michael B, Ni HP, et al. Suppression of RNA Recognition by Toll-like Receptors: The Impact of Nucleoside Modification and the Evolutionary Origin of RNA. Immunity, 2005, 23(2):165-175.
    85. Richard WC, Erik JS. Origins and Mechanisms of miRNAs and siRNAs. Cell,2009,136(4):642-655.
    86. Tseng YC, Subho M, Huang L. Lipid-based systemic delivery of siRNA. Advanced Drug Delivery Reviews, Available online 26 March 2009.
    87. Elizabeth RR, Zhang RW. Antisense, RNAi, and gene silencing strategies for therapy: Mission possible or impossible? Drug Discovery Today, 2008, 13(11-12):513-521.
    88. Xiong XB, Hasan U, Afsaneh L. Biodegradable amphiphilic poly(ethylene oxide)-block-polyesters with grafted polyamines as supramolecular nanocarriers for efficient siRNA delivery. Biomaterials, 2009, 30(2):242-253.
    1. Jared AG, Scott W, Chris C, et al. Role of Raf Kinase in Cancer: Therapeutic Potential of Targeting the Raf/MEK/ERK Signal Transduction Pathway. Seminars in Oncology, 2006, 33(4): 392-406.
    2. Fatih C, Semra C, Christiaan K, et al. Disruption of Tumor Cell Adhesion Promotes Angiogenic Switch and Progression to Micrometastasis in RAF-Driven Murine Lung Cancer. Cancer Cell, 2007,12(2): 145-159.
    3. Evan TK, Zheng F, Kam Y, et al. Raf kinase inhibitor protein: a prostate cancer metastasis suppressor gene. Cancer Letters, 2004, 207(2): 131-137.
    4. Kathleen MWI, Navdeep KG, Sonja MM,et al. Loss of Raf Kinase Inhibitory Protein Induces Radioresistance in Prostate Cancer. International Journal of Radiation Oncology Biology Physics, 2008, 72(1):153-160.
    5. Herbert C, Eleanor BCW, Stephen BB, et al. Differentiation of medullary thyroid cancer by C-Raf-1 silences expression of the neural transcription factor human achaete-scute homolog-1.Surgery, 1996,120(2):168-173.
    6. Kathryn EM, Catrin AP. Raf proteins and cancer: B-Raf is identified as a mutational target. Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, 2003,1653(1): 25-40.
    7. Caroline RWO, William LB, Linda SS, et al. The Raf signal transduction cascade as a target for chemotherapeutic intervention in growth factor-responsive tumors.Pharmacology & Therapeutics, 2000, 88(3):229-279.
    8. Nithya R, Alex A. Inhibitors of Raf kinase and MEK signaling. Update on Cancer Therapeutics, 2007, 2(3):111-118.
    9. William MO, John BS, Stephane H, et al. Functional Proteomics Identifies Targets of Phosphorylation by B-Raf Signaling in Melanoma. Molecular Cell, 2009,34(1):115-131.
    10. David HB, Lori A.J, Hong C, et al. Activation of Raf-1 in Human Pancreatic Adenocarcinoma. Journal of Surgical Research, 1997, 69(1):199-204.
    11. Gillis HC, Pinchot SN, Chen H. The Synergistic Effects of Octreotide and RAF-1 Actvation in Carcinoid Cancer Cells. Journal of Surgical Research, 2009,151(2):249.
    12. Joshua DH, Jonas G, Brad N, et al. Potent and selective pyrazole-based inhibitors of B-Raf kinase. Bioorganic & Medicinal Chemistry letters, 2008, 18(16):4692-4695.
    13. Eijiro O, Unnur PT. Different regulation of vascular endothelial growth Factor expression by the ERK and p38 kinase pathways in v-ras, v-raf, and v-myc transformed cells. Biochemical and Biophysical Research Communications, 2000,270(1):108-111.
    14. Eun YS, Navneet K, Mi YP, et al. Synthesis of amide and urea derivatives of benzothiazole as Raf-1 inhibitor. European Journal of Medicinal Chemistry, 2008,43(7):1519-1524.
    15. Pan MR, Chang HC, Hung WC. Non-steroidal anti-inflammatory drugs suppress the ERK signaling pathway via block of Ras/c-Raf interaction and activation of MAP kinase phosphatases. Cellular Signalling, 2008, 20(6):1134-1141.
    16. Mathew JG, Richard M. Guilty as charged: B-RAF is a human oncogene. Cancer Cell, 2004, 6(4):313-319.
    17. James AM, Linda SS, William HC, et al. Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, 2007,1773(8):1263-1284.
    18. Samar A, Ulf RR, Petra H. Validation of a liquid chromatography assay for the quantification of the Raf kinase inhibitor BAY 43-9006 in small volumes of mouse serum. Journal of Chromatography B, 2004, 809(1):99-103.
    19. Florian AK, David AT. Modelling oncogenic Ras/Raf signalling in the mouse.Current Opinion in Genetics & Development, 2009,19(1):4-11.
    20. Manuela B. Second nature: Biological functions of the Raf-1 "kinase". FEBS Letters, 2005, 579(15):3271-3277.
    21. Stacey J, Maria A, Peter JF. Expression status and mutational analysis of the Ras and B-raf genes in ovarian granulosa cell and epithelial tumors. Gynecologic Oncology, 2004, 95(3):603-609.
    22. Antoine G, Jochen F. RAF kinases and mitochondria. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, 2007,1773(8):1256-1262.
    23. Ivo RH, Alfred W, Adriaan PB, et al.Selection of phage-displayed Fab antibodies on the active conformation of Ras yields a high affinity conformation-specific antibody preventing the binding of c-Raf kinase to Ras. FEBS Letters, 1999,463(l-2):115-120.
    24. Carsten C, Edith G, Sabine H, et al. Effects of Raf-1 siRNA on human cerebral microvascular endothelial cells: A potential therapeutic strategy for inhibition of tumor angiogenesis. Brain Research, 2006,1125(1):147-154.
    25. Gr######nwald V, Morgan M, Ganser A, et al. Inhibitors of the Ras/Raf/MAPK signaling pathway sensitize pancreatic cancer cells resistant to EGFR inhibitors.European Journal of Cancer Supplements, 2004, 2(8): 108.
    26. Richard AB, Sherry P, Jennifer O, et al. Paclitaxel is preferentially cytotoxic to human cervical tumor cells with low Raf-1 kinase activity: implications for paclitaxel-based chemoradiation regimens. Radiotherapy and Oncology, 1998,48(3):329-334.
    27. Huira C, Haris GV, Guan KL. Mechanisms of regulating the Raf kinase family.Cellular Signalling, 2003,15(5):463-469.
    28. Maximilian B, Stefan D, Christine H, et al. Mitogen-Activated Protein Kinase Signaling is Activated in Prostate Tumors but not Mediated by B-RAF Mutations. European Urology, 2006, 50(5):1102-1110.
    29. Han CL, Bo T, John MS, et al. Loss of Raf Kinase Inhibitor Protein Promotes Cell Proliferation and Migration of Human Hepatoma Cells. Gastroenterology, 2006,131(4):1208-1217.
    30. Mano MS, Awada A, Minisini A, et al. Exquisite antitumour response to trastuzumab in a patient with no evidence of Ras-Raf-MAPK and PI3K-Akt pathways activation. The Breast, 2004, 13(4):347-349.
    31. Li N, David YG, Renata JS, et al. Focal Adhesion Kinase (FAK) Is A Downstream Effector of the Raf-1 Signal Transduction Pathway in Gastrointestinal Carcinoid Cancer Cells. Journal of Surgical Research, 2008,144(2):239.
    32. Vitor T, Paula S, Manuel S. B-RAF mutations in the etiopathogenesis, diagnosis, and prognosis of thyroid carcinomas. Human Pathology, 2006, 37(7):781-786.
    33. Deborah TL, Vitaly B, Alexander K, et al. Raf kinases: Function, regulation and role in human cancer. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, 2007,1773(8): 1196-1212.
    34. David GM, Matthew AP, Mary AM, et al. RAS/RAF mutation and defective DNA mismatch repair in endometrial cancers. American Journal of Obstetrics and Gynecology, 2004,190(4):935-939.
    35. Ashley K, Abram V, Muthusamy K,et al. ZM336372, A Raf-1 Activator, Inhibits Growth of Pheochromocytoma Cells. Journal of Surgical Research, 2006,133(1):42-45.
    36. Paul TCW, Mathew JG, Roe SM, et al. Mechanism of Activation of the RAF-ERK Signaling Pathway by Oncogenic Mutations of B-RAF. Cell, 2004,116(6):855-867.
    37. Jayaprakasha GK, Mandadi KK, Shibu MP, et al. Inhibition of colon cancer cell growth and antioxidant activity of bioactive compounds from Poncirus trifoliata Raf. Bioorganic & Medicinal Chemistry, 2007,15(14):4923-4932.
    38. Eva ME, Paul S, Karuna N, et al. Raf Kinase Inhibitory Protein Regulates Aurora B Kinase and the Spindle Checkpoint. Molecular Cell, 2006, 23(4):561-574.
    39. Neil T, John L. Recent progress in targeting the Raf/MEK/ERK pathway with inhibitors in cancer drug discovery. Current Opinion in Pharmacology, 2005,5(4):350-356.
    40. Gretchen AR, Emily JC, Channing JD. Renewing the conspiracy theory debate:does Raf function alone to mediate Ras oncogenesis? Trends in Cell Biology, 2004,14(11):639-647.
    41. Robert M, Larry RF, Steven CK, et al. An Enzyme-Linked Immunosorbent Assay for the Raf/MEK1/MAPK Signaling Cascade. Analytical Biochemistry, 2001,294(1):48-54.
    42. Golaun O, Devasis C, Ali RJ, et al. Raf-1 Kinase Inhibitor Protein: Structure,Function, Regulation of Cell Signaling, and Pivotal Role in Apoptosis. Advances in Cancer Research, 2004, 91:169-200.
    43. A Phosphatase Holoenzyme Comprised of Shoc2/Sur8 and the Catalytic Subunit of PP1 Pablo RV, Juan OP, Alma B, et al. Functions as an M-Ras Effector to Modulate Raf Activity.Molecular Cell, 2006, 22(2):217-230.
    44. James AM, Linda SS, Steven LA, et al. Roles of the RAF/MEK/ERK and PI3K/PTEN/AKT pathways in malignant transformation and drug resistance. Advances in Enzyme Regulation, 2006, 46(1):249-279.
    45. Mathew JG, Sareena R, Hugh P, et al. Wild-Type and Mutant B-RAF Activate C-RAF through Distinct Mechanisms Involving HeteTodimerization. Molecular Cell, 2005, 20(6):963-969.
    46. Evan TK, Zheng F, Meghan B. The role of Raf kinase inhibitor protein (RKIP) in health and disease. Biochemical Pharmacology, 2004, 68(6): 1049-1053.
    47. Marais R. The roles of B-RAF and C-RAF in human cancer.European Journal of Cancer Supplements, 2005, 3(4):10.
    48. Fu Z, Smith PC, Zhang L, et al. Effects of raf kinase inhibitor protein expression on suppression of prostate cancer metastasis. Urologic Oncology: Seminars and Original Investigations, 2004, 22(1):78-79.
    49. Mukherjee R, Edwards J, Underwood MA, et al. Raf-1 expression is related to early hormone-relapse in prostate cancer. European Urology Supplements, 2003,2(6):83.
    50. Michael L, Woo SK, Sang SH. Down-regulation of Raf-1 kinase is associated with paclitaxel resistance in human breast cancer MCF-7/Adr cells. Cancer Letters, 2003,193(1):57-64.
    51. Steven T, Mandy P, Kimberly SC, et al. Raf kinase inhibitory protein knockout mice: Expression in the brain and olfaction deficit. Brain Research Bulletin,2007,71(6):559-567.
    52. Roman H, Judith SSL. Unraveling the complexities of the Raf/MAP kinase pathway for pharmacological intervention. Trends in Molecular Medicine, 2002,8(4):S27-S31.
    53. Park SJ, Kim YY, Ju JW, et al. Alternative Splicing Variants of c-FLIP Transduce the Differential Signal through the Raf or TRAF2 in TNF-Induced Cell Proliferation. Biochemical and Biophysical Research Communications, 2001,289(5):1205-1210.
    54. Jan K, Steven JT, John MS, et al. Signaling crossroads: The function of Raf kinase inhibitory protein in cancer, the central nervous system and reproduction. Cellular Signalling, 2008, 20(1):1-9.
    55. Scott NP, Joel TA, Yinggang L, et al. Tautomycin suppresses growth and neuroendocrine hormone markers in carcinoid cells through activation of the Raf-1 pathway. The American Journal of Surgery, 2009, 197(3):313-319.

© 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