白花丹醌通过VEGFR2-RAS信号通路抑制肿瘤血管新生以及LGR4通过调节肿瘤干细胞促进乳腺癌发生发展的机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
本人在攻读博士学位期间的研究分为两部分,分别从药物开发及基因治疗两个不同方向阐述了本人在肿瘤治疗方面的研究成果。第一部分讨论了中草药单体白花丹醌通过抗血管新生抑制肿瘤的机制研究,第二部分讨论了LGR4/GPR48通过影响肿瘤干细胞从而调节乳腺癌发生发展的研究。
     1.血管新生是在已生成血管的基础上,通过内皮细胞的增殖、迁移以及与周围环境的交流与协调,最终形成新的血管的多步骤过程.由于血管新生是几乎所有肿瘤发生发展的共性,针对其开发的药物尤其具有临床价值。
     白花丹醌是从中草药白花丹中提取出来的黄色针状结晶,先前的研究表明其具有抗病毒、抗生育、抗凝血以及抗肿瘤的功效,最近也有研究表明其在抗动脉粥样硬化以及抗炎中的作用,但其在肿瘤血管新生中的相关研究尚未有人报道。因此在本研究的第一部分,我们发现白花丹醌能抑制由VEGF诱导的内皮细胞的生长及迁移等生物学功能,并且在鸡胚尿囊膜和小鼠角膜的血管新生经典实验中有着明显的抗血管新生功效。在结肠癌和前列腺癌的裸鼠荷瘤实验中,白花丹醌进一步的抑制了体内肿瘤血管新生。在白花丹醌影响血管新生的分子机理探索的过程中,我们发现白花丹醌显著地抑制了内皮细胞在VEGF诱导下F-actin应力纤维的形成,而调节这种应力纤维的一条重要信号通路就是Rac-PAK1-LIMK-Cofilin通路,另外我们也发现白花丹醌对内皮细胞的生长也有强烈的抑制作用,其中紧密相关的信号通路是MEK-ERK/JNK通路。这两条通路的共同上游是Ras蛋白,在内皮细胞中Ras蛋白又由VEGFR2所调控。因此我们检验了白花丹醌在这个信号级联反应中的作用,并证明了白花丹醌能够通过抑制VEGFR2的磷酸化从而抑制由VEGF活化的Ras下游的信号,并进一步影响内皮细胞迁移及生长,最终抑制肿瘤血管新生。因此,白花丹醌很可能成为抗肿瘤的候选药物。
     2.肿瘤干细胞是肿瘤研究领域的新理论,该理论认为肿瘤的发生与转移都是有赖于存在于肿瘤细胞群体中极少部分的获得了干细胞特性的肿瘤细胞。该理论对于临床治疗药物耐受性,抗辐射性以及许多肿瘤的休眠行为提供了很好的解释。这个由干细胞驱动的肿瘤形成的理论引起了很多讨论与问题,包括是否在靶向快速分裂细胞的抗肿瘤治疗同时靶向到那些缓慢自我更新的肿瘤干细胞。因此最近对于肿瘤干细胞的特征的研究更多的集中在鉴定出那些使得肿瘤干细胞对于治疗干预更敏感更易受到攻击的信号通路。
     G蛋白偶联受体是一大类膜结合蛋白家族,其中包括的蛋白都具有七次跨膜的结构,并承担将胞外刺激转导到胞内的功能。这类蛋白广泛地参与了对于包括免疫系统、神经系统发育等在内的生理病理过程的调控,因此GPCR已经成为现今50-60%的疾病治疗药物的直接或间接靶点。越来越多的实验及临床数据发现肿瘤细胞能够将GPCR的正常生理功能劫为己用,从而促进自己的生长,逃避免疫识别,增加自己的营养和氧气供应,侵入周围组织并最终散播到另外的器官。因此,调控GPCR以及其下游靶点可能为新的抗肿瘤策略以及肿瘤诊断与防治提供了新的机会。
     LGR4/GPR48是G蛋白偶联受体大家族里的一员,在我们实验室研制的LGR4基因敲除小鼠中,发现了在骨发育,眼睛发育,肾功能以及雄性生殖系统中的缺陷,最近发表的文章提示LGR4在乳腺及前列腺发育中也起到调节作用,但是至今鲜有对LGR4在肿瘤中的研究。基于我们实验室初步的研究证明,LGR4在乳腺癌的发生发展中起到促进作用,但是否LGR4的这种对乳腺癌的促进作用是通过对肿瘤于细胞的调节而达到的尚未可知。所以我们通过将LGR4基因敲出老鼠与乳腺癌肿瘤干细胞小鼠模型进行杂交,发现将LGR4敲除之后,由WNT1驱动的肿瘤形成以及转移受到了明显的抑制,且存在于肿瘤中的以CD90+CD24+作为表面标志的乳腺癌肿瘤干细胞的功能被强烈抑制了。而存在于LGR4表达缺失的WNT1肿瘤中的肿瘤干细胞的发生频率也受到大幅度下调,其自我更新、分化及肿瘤再造功能都降低了。并且LGR4的下调能促进肿瘤细胞对化疗药物的敏感性。
     因此,LGR4很有可能通过调节肿瘤干细胞而促进乳腺癌的发生发展,并很可能成为乳腺癌治疗的很好的靶点。
To better elaborate my research achievements from the aspects of drug development and gene therapy, my research work was divided into two distinct parts. In the first part,by which mechanism, plumbagin, the Chinese herb monomer, inhibits tumor growth through anti-angiogenesis function was discussed; in the second part, how LGR4/GPR48regulate breast cancer initiation and progression through cancer stem cell was lucubrated.
     1. Angiogenesis is the multi-process composing of endothelial cell proliferation, migration and interaction as well as corporation with the environments based on the existing vessels. Because angiogenesis is the commonality for all the tumor development, drugs designed to target angiogenesis are of high clinical value.
     Plumbagin is a crystal compound isolated from some Chinese herbs such as plumbaginaceae, Ebenaceae and Droseraceae. Previous studies showed its antibacterial, antifertility, anticoagulant and anti-cancer properties. And, recent studies reported its antiatherosclerotic and anti-inflammation effects as well. However its function in tumor angiogenesis has not yet been reported. So in the first part, I discovered that plumbagin can specifically inhibit endothelial cell proliferation and VEGF-induced migration. Furthermore, in chick embryo chorioallantoic membrane and mouse corneal assay, plumbagin exhibits strong anti-angiogenesis effect. In the human colon cancer and prostate cancer xenograft assays, plumbagin inhibits angiogenesis in vivo. To explore the underlined molecular mechanism, we found that plumbagin significantly decreased the ability of endothelial cell to from F-actin stress fiber under the VEGF induction. One of the most important signal pathways that cause this phenomenon is Rac-PAK1-LIMK-Cofilin pathway. Besides, plumbagin also regulates the proliferation of endothelial cells, which is controlled by MEK-ERK/JNK pathway. Both of these two pathways have the common upper stream regulator Ras, which is controlled by VEGFR2. Hence, we tested the role of plumbagin in this cascade, and proved that plmbagin can inhibit the phosphorylation of VEGFR2in the endothelial cells and down-regulate the VEGF activation for downstream signals of Ras pathway, then affects endothelial cell migration and proliferation, and ultimately inhibits tumor angiogenesis. Based on our study, plumbagin could be an anti-cancer drug candidate.
     2. Cancer stem study is a new field for cancer research, and it is believed that the carcinomas originate from so called cancer stem cell population upon the oncogenic hit. This theory also provides new explanation on drug resistance, anti-radioactivity properties of cancer cells. Inevitably, some discussions and questions were raised up coming with the new theory, including whether the cancer treatment should switch from the traditional to the newly merging one. As a result, more research has been done to target on signaling pathways that are unique for cancer stem cell.
     G coupled protein family consists of proteins that are characterized by a7transmembarane domain, and it function as a signal transducer. The loss of function of these proteins will lead to multiple human diseases, so50%to60%drugs approved by FDA are designed to specifically target this particular family. Numerous studies have found that tumor cell can take advantage GPCR signaling to overcome the obstacles preventing them from grow wild, and invade other organs through circulation by getting the nutrients and oxygen they need to survive. Therefore, it is important to unravel the mechanism under the regulation of GPCR family, and the success to do so will definitely help us get new strategy to diagnose and eventually win the battle against cancer.
     LGR4/GPR48belongs to GPCR family, and we have found that the deletion of LGR4in mouse model led to severe development defect in bone, eye, renal and reproduction system. Our most recent paper also points out the importance of LGR4mammary gland development. However, the function of LGR4in cancer including breast cancer is barely known. Base on the data we have so far, we can conclude that LGR4must play a role in breast cancer, but the mechanism still not clear. To address this question, we crossed LGR4null/ko mouse with WNT1driven mouse tumor model. Interestingly, the progression of tumor, and the function of cells labeled by CD90+CD24+, the marker for breast cancer stem cell, are significantly suppressed in WNT+LGR4-/-LGR4mice. We also proved that the cancer stem cells population sorted from Wntl+LGR4-/-mice have poorer ability in self-renewal, in ivtro differentiation and in vivo tumor regeneration. Altogether, LGR4could be an essential pathway for cancer stem cell, therefore it has potential to become a promising drug target.
引文
1. Jemal A, et al. (2011) Global cancer statistics. (Translated from eng) CA Cancer J Clin 61 (2):69-90 (in eng).
    2. Miller AB, Hoogstraten B, Staquet M, & Winkler A (1981) Reporting results of cancer treatment. (Translated from eng) Cancer 47(1):207-214 (in eng).
    3. Siegel R, Naishadham D, & Jemal A (2013) Cancer statistics,2013. (Translated from eng) CA Cancer J Clin 63(1):11-30 (in eng).
    4. Hung A & Mullins CD (2013) Relative effectiveness and safety of chemotherapy in elderly and nonelderly patients with stage III colon cancer:a systematic review. (Translated from eng) Oncologist 18(1):54-63 (in eng).
    5. Dalibon N, Moutafis M, & Fischler M (2004) Laparoscopically assisted versus open colectomy for colon cancer. (Translated from eng) N Engl JMed 351(9):933-934; author reply 933-934 (in eng).
    6. Laurent-Puig P, et al. (2009) Analysis of PTEN, BRAF, and EGFR status in determining benefit from cetuximab therapy in wild-type KRAS metastatic colon cancer. (Translated from eng) J Clin Oncol 27(35):5924-5930 (in eng).
    7. Westra JL, et al. (2005) Determination of TP53 mutation is more relevant than microsatellite instability status for the prediction of disease-free survival in adjuvant-treated stage III colon cancer patients. (Translated from eng) J Clin Oncol 23(24):5635-5643 (in eng).
    8. Ames BN (2001) DNA damage from micronutrient deficiencies is likely to be a major cause of cancer. (Translated from eng) Mutat Res 475(1-2):7-20 (in eng).
    9. Mettlin C (1997) The American Cancer Society National Prostate Cancer Detection Project and National patterns of prostate cancer detection and treatment. (Translated from eng) CA Cancer J Clin 47(5):265-272 (in eng).
    10. Schroder FH, et al. (1992) The TNM classification of prostate cancer. (Translated from eng) Prostate Suppl 4:129-138 (in eng).
    11. Soloway MS, et al. (1988) Stratification of patients with metastatic prostate cancer based on extent of disease on initial bone scan. (Translated from eng) Cancer 61(1):195-202 (in eng).
    12. Logothetis CJ & Lin SH (2005) Osteoblasts in prostate cancer metastasis to bone. (Translated from eng) Nat Rev Cancer 5(1):21-28 (in eng).
    13. Ganesh B, Saoba SL, Sarade MN, & Pinjari SV (2011) Risk factors for prostate cancer:An hospital-based case-control study from Mumbai, India. (Translated from eng) Indian J Urol 27(3):345-350 (in eng).
    14. Rowlands MA, et al. (2012) Circulating insulin-like growth factors and IGF-binding proteins in PSA-detected prostate cancer:the large case-control study ProtecT. (Translated from eng) Cancer Res72(2):503-515(in eng).
    15. Catto JW, et al. (2011) Suitability of PSA-detected localised prostate cancers for focal therapy: experience from the ProtecT study. (Translated from eng) Br J Cancer 105(7):931-937 (in eng).
    16. Schroder FH (2001) Diagnosis, characterization and potential clinical relevance of prostate cancer detected at low PSA ranges. (Translated from eng) Eur Urol 39 Suppl 4:49-53 (in eng).
    17. Sharifi N, Gulley JL, & Dahut WL (2005) Androgen deprivation therapy for prostate cancer. (Translated from eng) JAMA 294(2):238-244 (in eng).
    18. Kelsey JL (1979) A review of the epidemiology of human breast cancer. (Translated from eng) Epidemiol Rev 1:74-109 (in eng).
    19. Parkin DM & Fernandez LM (2006) Use of statistics to assess the global burden of breast cancer. (Translated from eng) Breast J 12 Suppl 1:S70-80 (in eng).
    20. Dupont WD & Page DL (1985) Risk factors for breast cancer in women with proliferative breast disease. (Translated from eng) NEngl J Med 312(3):146-151 (in eng).
    21. Stanton AL & Snider PR (1993) Coping with a breast cancer diagnosis:a prospective study. (Translated from eng) Health Psychol 12(1):16-23 (in eng).
    22. Harbeck N (2013) [Breast cancer:tumor biology-based concepts for surgical and drug treatment]. (Translated from ger) Dtsch Med Wochenschr 138(5):180-182 (in ger).
    23. Dickler MN & Norton L (2001) The MORE trial:multiple outcomes for raloxifene evaluation--breast cancer as a secondary end point:implications for prevention. (Translated from eng) Ann N YAcad Sci 949:134-142 (in eng).
    24. Cauley JA, et al. (2001) Continued breast cancer risk reduction in postmenopausal women treated with raloxifene:4-year results from the MORE trial. Multiple outcomes of raloxifene evaluation. (Translated from eng) Breast Cancer Res Treat 65(2):125-134 (in eng).
    25. Jin S & Ye K (2013) Targeted drug delivery for breast cancer treatment. (Translated from eng) Recent Pat Anticancer Drug Discov 8(2):143-153 (in eng).
    26. Burstein HJ, et al. (2003) Trastuzumab and vinorelbine as first-line therapy for HER2-overexpressing metastatic breast cancer:multicenter phaseⅡtrial with clinical outcomes, analysis of serum tumor markers as predictive factors, and cardiac surveillance algorithm. (Translated from eng) JClin Oncol 21(15):2889-2895 (in eng).
    27. Eiermann W, et al. (2013) The 21-gene recurrence score assay impacts adjuvant therapy recommendations for ER-positive, node-negative and node-positive early breast cancer resulting in a risk-adapted change in chemotherapy use. (Translated from eng) Ann Oncol 24(3):618-624 (in eng).
    28. Hainaut P & Plymoth A (2013) Targeting the hallmarks of cancer:towards a rational approach to next-generation cancer therapy. (Translated from eng) Curr Opin Oncol 25(1):50-51 (in eng).
    29. Hanahan D & Weinberg RA (2011) Hallmarks of cancer:the next generation. (Translated from eng) Cell 144(5):646-674 (in eng).
    30. Friedl P & Alexander S (2011) Cancer invasion and the microenvironment:plasticity and reciprocity. (Translated from eng) Cell 147(5):992-1009 (in eng).
    31. Atala A (2012) Re:Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. (Translated from eng) J Urol 187(4):1506-1507(in eng).
    32. Lonardo E, Frias-Aldeguer J, Hermann PC, & Heeschen C (2012) Pancreatic stellate cells form a niche for cancer stem cells and promote their self-renewal and invasiveness. (Translated from eng) Cell Cycle 11(7):1282-1290 (in eng).
    33. Ribatti D & Vacca A (2008) The role of microenvironment in tumor angiogenesis. (Translated from eng) Genes Nutr 3(1):29-34 (in eng).
    34. Malanchi I, et al. (2012) Interactions between cancer stem cells and their niche govern metastatic colonization. (Translated from eng) Nature 481(7379):85-89 (in eng).
    35. de Almeida Sassi F, Lunardi Brunetto A, Schwartsmann G, Roesler R, & Abujamra AL (2012) Glioma revisited:from neurogenesis and cancer stem cells to the epigenetic regulation of the niche. (Translated from eng) J Oncol 2012:537861 (in eng).
    36. Wang Z & Ouyang G (2012) Periostin:a bridge between cancer stem cells and their metastatic niche. (Translated from eng) Cell Stem Cell 10(2):111-112 (in eng).
    37. Das Thakur M, et al. (2013) Modelling vemurafenib resistance in melanoma reveals a strategy to forestall drug resistance. (Translated from eng) Nature 494(7436):251-255 (in eng).
    38. Hambardzumyan D, et al. (2008) PI3K pathway regulates survival of cancer stem cells residing in the perivascular niche following radiation in medulloblastoma in vivo. (Translated from eng) Genes Dev 22(4):436-448 (in eng).
    39. Alsford S, et al. (2012) High-throughput decoding of antitrypanosomal drug efficacy and resistance. (Translated from eng) Nature 482(7384):232-236 (in eng).
    40. McCormick F (2001) New-age drug meets resistance. (Translated from eng) Nature 412(6844):281-282 (in eng).
    41. Field HJ & Darby GK (1980) Strategies of drug resistance in herpes simplex. (Translated from eng) Nature 286(5776):842 (in eng).
    42. Takahashi T, Ueno H, & Shibuya M (1999) VEGF activates protein kinase C-dependent, but Ras-independent Raf-MEK-MAP kinase pathway for DNA synthesis in primary endothelial cells. (Translated from eng) Oncogene 18(13):2221-2230 (in eng).
    43. Cheng Y, et al. (2009) Oridonin induces G2/M arrest and apoptosis via activating ERK-p53 apoptotic pathway and inhibiting PTK-Ras-Raf-JNK survival pathway in murine fibrosarcoma L929 cells. (Translated from eng) Arch Biochem Biophys 490(1):70-75 (in eng).
    44. Maruta H, Tikoo A, Shakri R, & Shishido T (1999) The anti-RAS cancer drug MKT-077 is an F-actin cross-linker. (Translated from eng)Ann N YAcad Sci 886:283-284 (in eng).
    45. Kirstein MN, Moore MM, & Dudek AZ (2006) Review of selected patents for cancer therapy targeting tumor angiogenesis. (Translated from eng) Recent Pat Anticancer Drug Discov 1(2):153-161 (in eng).
    46. Berezin DP & Filatov VN (1989) [Trends in the survival of cancer patients:is there progress in the anticancer fight]. (Translated from rus) Vopr Onkol 35(5):545-559 (in rus).
    47. Zhang XC, et al. (2013) [Consensus on dignosis for ALK positive non-small cell lung cancer in China, the 2013 version]. (Translated from chi) Zhonghua Bing Li Xue Za Zhi 42(6):402-406 (in chi).
    48. Guan L, Dai Y, & Luo R (2013) Translational research in oncology research & development and its impact on early development in China:report of the 5th Annual Meeting of the US Chinese Anti-Cancer Association (USCACA) at 2013 AACR Annual Meeting. (Translated from eng) Chin J Cancer 32(7):357-362 (in eng).
    49. Wu Q, et al. (2009) Extracellular calcium increases CXCR4 expression on bone marrow-derived cells and enhances pro-angiogenesis therapy. (Translated from eng) J Cell Mol Med 13(9B):3764-3773 (in eng).
    50. Taniyama Y, Azuma J, Iwabayashi M, Morishita R, & Rakugi H (2011) [Therapeutic angiogenesis for coronary artery]. (Translated from jpn) Nihon Rinsho 69 Suppl 7:74-79 (in jpn).
    51. Elbaz M & Malavaud B (1999) [VEGF in therapeutic coronary and peripheral artery angiogenesis]. (Translated from fre) Pathol Biol (Paris) 47(4):380-384 (in fre).
    52. Cochain C, Channon KM, & Silvestre JS (2013) Angiogenesis in the infarcted myocardium. (Translated from eng) Antioxid Redox Signal 18(9):1100-1113 (in eng).
    53. Matsuno H, et al. (2002) Treatment with the angiogenesis inhibitor endostatin:a novel therapy in rheumatoid arthritis. (Translated from eng) JRheumatol 29(5):890-895 (in eng).
    54. Folkman J (1996) Tumor angiogenesis and tissue factor. (Translated from eng) Nat Med 2(2):167-168(in eng).
    55. Mund JA & Case J (2011) The role of circulating endothelial progenitor cells in tumor angiogenesis. (Translated from eng) Curr Stem Cell Res Ther 6(2):115-121 (in eng).
    56. Ribatti D (2004) The involvement of endothelial progenitor cells in tumor angiogenesis. (Translated from eng) J Cell Mol Med 8(3):294-300 (in eng).
    57. He LL, Zhang WJ, Su H, & Xu DG (2007) [Synergism between Ang-2 and VEGF and its application of anti-angiogenesis in tumor therapy-review]. (Translated from chi) Zhongguo Shi Yan Xue Ye Xue Za Zhi 15(2):445-448 (in chi).
    58. Young PP, Hofling AA, & Sands MS (2002) VEGF increases engraftment of bone marrow-derived endothelial progenitor cells (EPCs) into vasculature of newborn murine recipients. (Translated from eng) Proc Natl Acad Sci US A 99(18):11951-11956 (in eng).
    59. Asahara T, et al. (1999) VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. (Translated from eng) EMBO J 18(14):3964-3972 (in eng).
    60. Chen X, et al. (2013) KH902 suppresses high glucose-induced migration and sprouting of human retinal endothelial cells by blocking VEGF and PIGF. (Translated from eng) Diabetes Obes Metab 15(3):224-233 (in eng).
    61. Semczuk-Sikora A, et al. (2007) [Maternal serum concentration of angiogenic factors:PIGF, VEGF and VEGFR-1 and placental volume in pregnancies complicated by intrauterine growth restriction]. (Translated from pol) Ginekol Pol 78(10):783-786 (in pol).
    62. Wang YQ, et al. (2004) Regulatory role of vHL/HIF-lalpha in hypoxia-induced VEGF production in hepatic stellate cells. (Translated from eng) Biochem Biophys Res Commun 317(2):358-362(in eng).
    63. Folkman J (2007) Angiogenesis:an organizing principle for drug discovery? (Translated from eng) Nat Rev Drug Discov 6(4):273-286 (in eng).
    64. Vincent L & Rafii S (2004) Vascular frontiers without borders:multifaceted roles of platelet-derived growth factor (PDGF) in supporting postnatal angiogenesis and lymphangiogenesis. (Translated from eng) Cancer Cell 6(4):307-309 (in eng).
    65. Xue Y, et al. (2012) PDGF-BB modulates hematopoiesis and tumor angiogenesis by inducing erythropoietin production in stromal cells. (Translated from eng) Nat Med 18(1):100-110 (in eng).
    66. Lin Z, Liu Y, Sun Y, & He X (2011) Expression of Ets-1, Ang-2 and maspin in ovarian cancer and their role in tumor angiogenesis. (Translated from eng) J Exp Clin Cancer Res 30:31 (in eng).
    67. Appelmann I, Liersch R, Kessler T, Mesters RM, & Berdel WE (2010) Angiogenesis inhibition in cancer therapy:platelet-derived growth factor (PDGF) and vascular endothelial growth factor (VEGF) and their receptors:biological functions and role in malignancy. (Translated from eng) Recent Results Cancer Res 180:51-81 (in eng).
    68. Klagsbrun M & Eichmann A (2005) A role for axon guidance receptors and ligands in blood vessel development and tumor angiogenesis. (Translated from eng) Cytokine Growth Factor Rev 16(4-5):535-548(in eng).
    69. Ergun S, et al. (2001) Endostatin inhibits angiogenesis by stabilization of newly formed endothelial tubes. (Translated from eng) Angiogenesis 4(3):193-206 (in eng).
    70. O'Reilly MS, et al. (1997) Endostatin:an endogenous inhibitor of angiogenesis and tumor growth. (Translated from eng) Cell 88(2):277-285 (in eng).
    71. Shintani S, et al. (2006) Anti-tumor effect of radiation response by combined treatment with angiogenesis inhibitor, TNP-470, in oral squamous cell carcinoma. (Translated from eng) Oral Oncol 42(1):66-72(in eng).
    72. Kim HR, et al. (2013) Multicentre phase II trial of bevacizumab combined with docetaxel-carboplatin for the neoadjuvant treatment of triple-negative breast cancer (KCSG BR-0905). (Translated from Eng) Ann Oncol (in Eng).
    73. Miller KD, Sweeney CJ, & Sledge GW, Jr. (2005) Can tumor angiogenesis be inhibited without resistance? (Translated from eng) EXS (94):95-112 (in eng).
    74. Portlock C (2006) Anti-angiogenesis and diffuse large B-cell lymphoma:a new treatment opportunity? (Translated from eng) Leuk Lymphoma 47(6):961-962 (in eng).
    75. De Craene B & Berx G (2013) Regulatory networks defining EMT during cancer initiation and progression. (Translated from eng) Nat Rev Cancer 13(2):97-110 (in eng).
    76. Nakaya Y & Sheng G (2013) EMT in developmental morphogenesis. (Translated from Eng) Cancer Lett (in Eng).
    77. Kerosuo L & Bronner-Fraser M (2012) What is bad in cancer is good in the embryo:importance of EMT in neural crest development. (Translated from eng) Semin Cell Dev Biol 23(3):320-332 (in eng).
    78. Biddle A & Mackenzie IC (2012) Cancer stem cells and EMT in carcinoma. (Translated from Eng) Cancer Metastasis Rev (in Eng).
    79. Brabletz T (2012) EMT and MET in metastasis:where are the cancer stem cells? (Translated from eng) Cancer Cell 22(6):699-701 (in eng).
    80. Ansieau S (2013) EMT in breast cancer stem cell generation. (Translated from eng) Cancer Lett 338(1):63-68(in eng).
    81. Javle MM, et al. (2007) Epithelial-mesenchymal transition (EMT) and activated extracellular signal-regulated kinase (p-Erk) in surgically resected pancreatic cancer. (Translated from eng) Ann Surg Oncol 14(12):3527-3533 (in eng).
    82. Xu X, et al. (2012) Genome-wide screening reveals an EMT molecular network mediated by Sonic hedgehog-Glil signaling in pancreatic cancer cells. (Translated from eng) PLoS One 7(8):e43119(in eng).
    83. Liu K & Ding S (2012) Target practice:modeling tumors with stem cells. (Translated from eng) Cell 149(6):1185-1187 (in eng).
    84. Schatton T, Frank NY, & Frank MH (2009) Identification and targeting of cancer stem cells. (Translated from eng) Bioessays 31(10):1038-1049 (in eng).
    85. Visvader JE & Lindeman GJ (2012) Cancer stem cells:current status and evolving complexities. (Translated from eng) Cell Stem Cell 10(6):717-728 (in eng).
    86. Valent P, et al. (2012) Cancer stem cell definitions and terminology:the devil is in the details. (Translated from eng) Nat Rev Cancer 12(11):767-775 (in eng).
    87. Brabletz T, Jung A, Spaderna S, Hlubek F, & Kirchner T (2005) Opinion:migrating cancer stem cells-an integrated concept of malignant tumour progression. (Translated from eng) Nat Rev Cancer 5(9):744-749 (in eng).
    88. Dean M, Fojo T, & Bates S (2005) Tumour stem cells and drug resistance. (Translated from eng) Nat Rev Cancer 5(4):275-284 (in eng).
    89. Cheng L, Ramesh AV, Flesken-Nikitin A, Choi J, & Nikitin AY (2010) Mouse models for cancer stem cell research. (Translated from eng) Toxicol Pathol 38(1):62-71 (in eng).
    90. Deshpande AJ & Buske C (2007) Lymphoid progenitors as candidate cancer stem cells in AML: new perspectives. (Translated from eng) Cell Cycle 6(5):543-545 (in eng).
    91. Regenbrecht CR, Lehrach H, & Adjaye J (2008) Stemming cancer:functional genomics of cancer stem cells in solid tumors. (Translated from eng) Stem Cell Rev 4(4):319-328 (in eng).
    92. Weinberg R, Fisher DE, & Rich J (2010) Dynamic and transient cancer stem cells nurture melanoma. (Translated from eng) Nat Med 16(7):758 (in eng).
    93. Bozzi F, Tamborini E, & Pilotti S (2012) The CD133 expression levels and its role as potential cancer stem cells marker in gastrointestinal stromal tumor. (Translated from eng) Int J Cancer 131(5):E849-850(in eng).
    94. Joshua B, et al. (2012) Frequency of cells expressing CD44, a head and neck cancer stem cell marker:correlation with tumor aggressiveness. (Translated from eng) Head Neck 34(1):42-49 (in eng).
    95. Engh JA (2010) Identification of a novel marker of brain tumor stem cells. (Translated from eng) Neurosurgery 67(4):N16 (in eng).
    96. Mao XG, et al. (2009) Brain Tumor Stem-Like Cells Identified by Neural Stem Cell Marker CD15. (Translated from eng) Transl Oncol 2(4):247-257 (in eng).
    97. Palapattu GS, et al. (2009) Selective expression of CD44, a putative prostate cancer stem cell marker, in neuroendocrine tumor cells of human prostate cancer. (Translated from eng) Prostate 69(7):787-798 (in eng).
    98. Wang Z, et al. (2010) Targeting miRNAs involved in cancer stem cell and EMT regulation:An emerging concept in overcoming drug resistance. (Translated from eng) Drug Resist Updat 13(4-5):109-118(in eng).
    99. Butler JM, Kobayashi H, & Rafii S (2010) Instructive role of the vascular niche in promoting tumour growth and tissue repair by angiocrine factors. (Translated from eng) Nat Rev Cancer 10(2):138-146(in eng).
    100. Villanueva T (2010) Cancer stem cells:Wnt--looking outside in. (Translated from eng) Nat Rev Cancer 10(6):386 (in eng).
    101. Zhu TS, et al. (2011) Endothelial cells create a stem cell niche in glioblastoma by providing NOTCH ligands that nurture self-renewal of cancer stem-like cells. (Translated from eng) Cancer Res 71(18):6061-6072 (in eng).
    102. Takebe N, Harris PJ, Warren RQ, & Ivy SP (2011) Targeting cancer stem cells by inhibiting Wnt, Notch, and Hedgehog pathways. (Translated from eng) Nat Rev Clin Oncol 8(2):97-106 (in eng).
    103. Medina V, Calvo MB, Diaz-Prado S, & Espada J (2009) Hedgehog signalling as a target in cancer stem cells. (Translated from eng) Clin Transl Oncol 11(4):199-207 (in eng).
    104. Tong DL, et al. (2011) A simpler method of preprocessing MALDI-TOF MS data for differential biomarker analysis:stem cell and melanoma cancer studies. (Translated from eng) Clin Proteomics 8:14 (in eng).
    105. Zhang M, et al. (2008) Identification of tumor-initiating cells in a p53-null mouse model of breast cancer. (Translated from eng) Cancer Res 68(12):4674-4682 (in eng).
    106. Cho RW, et al. (2008) Isolation and molecular characterization of cancer stem cells in MMTV-Wnt-1 murine breast tumors. (Translated from eng) Stem Cells 26(2):364-371 (in eng).
    107. Li Y, Hively WP, & Varmus HE (2000) Use of MMTV-Wnt-1 transgenic mice for studying the genetic basis of breast cancer. (Translated from eng) Oncogene 19(8):1002-1009 (in eng).
    108. Konigshoff M, et al. (2009) WNT1-inducible signaling protein-1 mediates pulmonary fibrosis in mice and is upregulated in humans with idiopathic pulmonary fibrosis. (Translated from eng) J Clin Invest 119(4):772-787 (in eng).
    109. Vaillant F, et al. (2008) The mammary progenitor marker CD61/beta3 integrin identifies cancer stem cells in mouse models of mammary tumorigenesis. (Translated from eng) Cancer Res 68(19):7711-7717 (in eng).
    110. Visvader JE & Lindeman GJ (2008) Cancer stem cells in solid tumours:accumulating evidence and unresolved questions. (Translated from eng) Nat Rev Cancer 8(10):755-768 (in eng).
    111. Sachlos E, et al. (2012) Identification of drugs including a dopamine receptor antagonist that selectively target cancer stem cells. (Translated from eng) Cell 149(6):1284-1297 (in eng).
    112. Baumann M, Krause M, & Hill R (2008) Exploring the role of cancer stem cells in radioresistance. (Translated from eng) Nat Rev Cancer 8(7):545-554 (in eng).
    113. Gilbertson RJ & Graham TA (2012) Cancer:Resolving the stem-cell debate. (Translated from eng) Nature 488(7412):462-463 (in eng).
    114. Cairns RA, Harris IS, & Mak TW (2011) Regulation of cancer cell metabolism. (Translated from eng) Nat Rev Cancer 11(2):85-95 (in eng).
    115. Hsu PP & Sabatini DM (2008) Cancer cell metabolism:Warburg and beyond. (Translated from eng) Cell 134(5):703-707 (in eng).
    116. Hausch F & Holsboer F (2012) The seven pillars of molecular pharmacology:GPCR research honored with Nobel Prize for chemistry. (Translated from eng) Angew Chem Int Ed Engl 51(49):12172-12175(in eng).
    117. Lappano R & Maggiolini M (2011) G protein-coupled receptors:novel targets for drug discovery in cancer. (Translated from eng) Nat Rev Drug Discov 10(1):47-60 (in eng).
    118. Rosenbaum DM, Rasmussen SG, & Kobilka BK (2009) The structure and function of G-protein-coupled receptors. (Translated from eng) Nature 459(7245):356-363 (in eng).
    119. Benovic JL (2012) G-protein-coupled receptors signal victory. (Translated from eng) Cell 151(6):1148-1150 (in eng).
    120. Cervantes D, Crosby C, & Xiang Y (2010) Arrestin orchestrates crosstalk between G protein-coupled receptors to modulate the spatiotemporal activation of ERK MAPK. (Translated from eng) Circ Res 106(1):79-88 (in eng).
    121. Dror RO, et al. (2011) Pathway and mechanism of drug binding to G-protein-coupled receptors. (Translated from eng) Proc Natl Acad Sci USA 108(32):13118-13123 (in eng).
    122. Gao Y, et al. (2006) Generation of a constitutively active mutant of human GPR48/LGR4, a G-protein-coupled receptor. (Translated from eng) Hokkaido Igaku Zasshi 81(2):101-105,107,109 (in eng).
    123. Mustata RC, et al. (2011) Lgr4 is required for Paneth cell differentiation and maintenance of intestinal stem cells ex vivo. (Translated from eng) EMBO Rep 12(6):558-564 (in eng).
    124. Mohri Y, Kato S, Umezawa A, Okuyama R, & Nishimori K (2008) Impaired hair placode formation with reduced expression of hair follicle-related genes in mice lacking Lgr4. (Translated from eng) Dev Dyn 237(8):2235-2242 (in eng).
    125. Ruffner H, et al. (2012) R-Spondin potentiates Wnt/beta-catenin signaling through orphan receptors LGR4 and LGR5. (Translated from eng) PLoS One 7(7):e40976 (in eng).
    126. Song H, et al. (2008) Inactivation of G-protein-coupled receptor 48 (Gpr48/Lgr4) impairs definitive erythropoiesis at midgestation through down-regulation of the ATF4 signaling pathway. (Translated from eng) JBiol Chem 283(52):36687-36697 (in eng).
    127. Kato S, et al. (2007) Eye-open at birth phenotype with reduced keratinocyte motility in LGR4 null mice. (Translated from eng) FEBS Lett 581(24):4685-4690 (in eng).
    128. Qian Y, et al. (2013) Lgr4-mediated Wnt/beta-catenin signaling in peritubular myoid cells is essential for spermatogenesis. (Translated from eng) Development 140(8):1751-1761 (in eng).
    129. Hoshii T, et al. (2007) LGR4 regulates the postnatal development and integrity of male reproductive tracts in mice. (Translated from eng) Biol Reprod 76(2):303-313 (in eng).
    130. Kato S, et al. (2006) Leucine-rich repeat-containing G protein-coupled receptor-4 (LGR4, Gpr48) is essential for renal development in mice. (Translated from eng) Nephron Exp Nephrol 104(2):e63-75(in eng).
    131. Oyama K, Mohri Y, Sone M, Nawa A, & Nishimori K (2011) Conditional knockout of Lgr4 leads to impaired ductal elongation and branching morphogenesis in mouse mammary glands. (Translated from eng) Sex Dev 5(4):205-212 (in eng).
    132. Breier G, et al. (2002) Transforming growth factor-beta and Ras regulate the VEGF/VEGF-receptor system during tumor angiogenesis. (Translated from eng) Int J Cancer 97(2):142-148 (in eng).
    133. Komatsu M & Ruoslahti E (2005) R-Ras is a global regulator of vascular regeneration that suppresses intimal hyperplasia and tumor angiogenesis. (Translated from eng) Nat Med 11(12):1346-1350(in eng).
    134. Oh YT, et al. (2012) Oncogenic Ras and B-Raf proteins positively regulate death receptor 5 expression through co-activation of ERK and JNK signaling. (Translated from eng) J Biol Chem 287(1):257-267(in eng).
    135. Neel NF, et al. (2011) The RalGEF-Ral Effector Signaling Network:The Road Less Traveled for Anti-Ras Drug Discovery. (Translated from eng) Genes Cancer 2(3):275-287 (in eng).
    136. Chen CA, Chang HH, Kao CY, Tsai TH, & Chen YJ (2009) Plumbagin, isolated from Plumbago zeylanica, induces cell death through apoptosis in human pancreatic cancer cells. (Translated from eng) Pancreatology 9(6):797-809 (in eng).
    137. Ahmad A, Banerjee S, Wang Z, Kong D, & Sarkar FH (2008) Plumbagin-induced apoptosis of human breast cancer cells is mediated by inactivation of NF-kappaB and Bcl-2. (Translated from eng) J Cell Biochem 105(6):1461-1471 (in eng).
    138. Srinivas P, Gopinath G, Banerji A, Dinakar A, & Srinivas G (2004) Plumbagin induces reactive oxygen species, which mediate apoptosis in human cervical cancer cells. (Translated from eng) Mol Carcinog 40(4):201-211 (in eng).
    139. Linseman DA & Loucks FA (2008) Diverse roles of Rho family GTPases in neuronal development, survival, and death. (Translated from eng) Front Biosci 13:657-676 (in eng).
    140. Olbrich L, Foehring D, Happel P, Brand-Saberi B, & Theiss C (2013) Fast rearrangement of the neuronal growth cone's actin cytoskeleton following VEGF stimulation. (Translated from eng) Histochem Cell Biol 139(3):431-445 (in eng).
    141. Jopling HM, et al. (2009) Rab GTPase regulation of VEGFR2 trafficking and signaling in endothelial cells. (Translated from eng) Arterioscler Thromb Vasc Biol 29(7):1119-1124 (in eng).
    142. Swendeman S, et al. (2008) VEGF-A stimulates ADAM17-dependent shedding of VEGFR2 and crosstalk between VEGFR2 and ERK signaling. (Translated from eng) Circ Res 103(9):916-918 (in eng).
    143. Glinka A, et al. (2011) LGR4 and LGR5 are R-spondin receptors mediating Wnt/beta-catenin and Wnt/PCP signalling. (Translated from eng) EMBO Rep 12(10):1055-1061 (in eng).
    144. de Lau W, et al. (2011) Lgr5 homologues associate with Wnt receptors and mediate R-spondin signalling. (Translated from eng) Nature 476(7360):293-297 (in eng).
    145. Jin YR & Yoon JK (2012) The R-spondin family of proteins:emerging regulators of WNT signaling. (Translated from eng) Int J Biochem Cell Biol 44(12):2278-2287 (in eng).
    146. Deng C, et al. (2013) Multi-functional norrin is a ligand for the LGR4 receptor. (Translated from eng) J Cell Sci 126(Pt 9):2060-2068 (in eng).
    147. Liu S, et al. (2013) Lgr4 gene deficiency increases susceptibility and severity of dextran sodium sulfate-induced inflammatory bowel disease in mice. (Translated from eng) J Biol Chem 288(13):8794-8803; discussion 8804 (in eng).
    148. Wang Y, et al. (2013) Lgr4 Regulates Mammary Gland Development and Stem Cell Activity through the Pluripotency Transcription Factor Sox2. (Translated from Eng) Stem Cells (in Eng).
    149. Luo W, et al. (2013) Lgr4 is a key regulator of prostate development and prostate stem cell differentiation. (Translated from Eng) Stem Cells (in Eng).
    150. Many AM & Brown AM (2010) Mammary stem cells and cancer:roles of Wnt signaling in plain view. (Translated from eng) Breast Cancer Res 12(5):313 (in eng).
    151. Vermeulen L, et al. (2008) Single-cell cloning of colon cancer stem cells reveals a multi-lineage differentiation capacity. (Translated from eng) Proc Natl Acad Sci U S A 105(36):13427-13432 (in eng).
    152. Hu Y & Smyth GK (2009) ELDA:extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. (Translated from eng) J Immunol Methods 347(1-2):70-78(in eng).
    153. Livingston DM & Silver DP (2008) Cancer:crossing over to drug resistance. (Translated from eng) Nature 451 (7182):1066-1067 (in eng).
    154. Johnston SR, et al. (1993) Acquired tamoxifen resistance in human breast cancer and reduced intra-tumoral drug concentration. (Translated from eng) Lancet 342(8886-8887):1521-1522 (in eng).
    155. Yeh KH, Lu YS, Hsiao CH, & Cheng AL (2003) High-dose tamoxifen modulates drug resistance to doxorubicin, dacarbazine and ifosfamide in metastatic uterine leiomyosarcoma. (Translated from eng) Anticancer Res 23(6D):5133-5137 (in eng).
    156. Drewa T, Styczynski J, & Szczepanek J (2008) Is the cancer stem cell population "a player" in multi-drug resistance? (Translated from eng) Acta Pol Pharm 65(4):493-500 (in eng).
    157. Hong W & Guan KL (2012) The YAP and TAZ transcription co-activators:key downstream effectors of the mammalian Hippo pathway. (Translated from eng) Semin Cell Dev Biol 23(7):785-793 (in eng).
    158. Zhao B, Tumaneng K, & Guan KL (2011) The Hippo pathway in organ size control, tissue regeneration and stem cell self-renewal. (Translated from eng) Nat Cell Biol 13(8):877-883 (in eng).
    159. Cordenonsi M, et al. (2011) The Hippo transducer TAZ confers cancer stem cell-related traits on breast cancer cells. (Translated from eng) Cell 147(4):759-772 (in eng).
    160. Tsai BP, Hoverter NP, & Waterman ML (2012) Blending hippo and WNT:sharing messengers and regulation. (Translated from eng) Cell 151 (7):1401-1403 (in eng).
    161. Yu FX, et al. (2012) Regulation of the Hippo-YAP pathway by G-protein-coupled receptor signaling. (Translated from eng) Cell 150(4):780-791 (in eng).
    162. de Sousa EMF, et al. (2011) Methylation of cancer-stem-cell-associated Wnt target genes predicts poor prognosis in colorectal cancer patients. (Translated from eng) Cell Stem Cell 9(5):476-485 (in eng).
    163. Dupont S, et al. (2011) Role of YAP/TAZ in mechanotransduction. (Translated from eng) Nature 474(7350):179-183 (in eng).
    164. Konsavage WM, Jr. & Yochum GS (2013) Intersection of Hippo/YAP and Wnt/beta-catenin signaling pathways. (Translated from eng) Acta Biochim Biophys Sin (Shanghai) 45(2):71-79 (in eng).

© 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