帳號:guest(3.138.34.177)          離開系統
字體大小: 字級放大   字級縮小   預設字形  

詳目顯示

以作者查詢圖書館館藏以作者查詢臺灣博碩士論文系統以作者查詢全國書目
作者(中文):葉家宇
作者(外文):Ip, Ka-U
論文名稱(中文):探討Foxm1基因對EGFR突變所誘發腫瘤形成之角色
論文名稱(外文):Role of Foxm1 in mutant EGFR induced mouse lung tumorigenesis
指導教授(中文):王翊青
指導教授(外文):Wang, I-Ching
口試委員(中文):陳仁焜
梁美智
口試委員(外文):Chen, Jen-Kun
Liang, Mei-Chih
學位類別:碩士
校院名稱:國立清華大學
系所名稱:生物科技研究所
學號:102080404
出版年(民國):105
畢業學年度:104
語文別:英文
論文頁數:97
中文關鍵詞:肺腺癌表皮生長因子受體Forkhead box M1基因工程小鼠微米級電腦斷層掃描造影系統
外文關鍵詞:adenocarcinomaEpidermal growth factor receptor, EGFRForkhead box M1, Foxm1Genetical Engineered Mouse Modelmicro computed tomography, microCT
相關次數:
  • 推薦推薦:0
  • 點閱點閱:65
  • 評分評分:*****
  • 下載下載:0
  • 收藏收藏:0
肺癌是全世界癌症死亡的首要原因。分子流行病學研究顯示,肺癌致癌基因存在差異。例如表皮生長因子受體(epidermal growth factor receptor,EGFR)發生突變的非小細胞肺癌(NSCLCs, non-small cell lung cancer)在亞洲人中的比例,比白人患者顯著較高。在台灣,特別是有關非吸煙女性患者的肺腺癌,EGFRL858R點突變發生頻率為最高。
之前研究顯示Forkhead box protein M1(Foxm1)在細胞增殖、分化,以及多種細胞類型的腫瘤發生中參與重要角色,是細胞中重要的轉錄因子。最近透過基因轉殖小鼠模式的研究發現,Foxm1在致癌物及致癌基因誘發肺癌癌化過程上扮演重要角色。在此,我們透過基因工程小鼠(Genetical Engineered Mouse Model)肺癌模式,條件性基因剔除老鼠肺部細支氣管上Club 細胞中的Foxm1基因(CCSP/EGFRL858R/Foxm1-/-),發現剔除Foxm1 會使EGFR誘發的腫瘤及增生病變顯著減少;同時,以微米級電腦斷層掃描造影系統(micro computed tomography, microCT)分析肺內部結構,以及肺臟石蠟切片作H&E染色病理學檢測,發現剔除Foxm1組中明顯減少點突變EGFRL858R誘發之腫瘤數目。此外,我們也發現,於氣管內注射表現CRE的腺病毒(Ad-Cre)以誘發Foxm1基因剔除,可使致癌EGFR所誘發的肺腫瘤萎縮(CCSP/EGFRL858R/Foxm1fl/fl)。我們的實驗結果證明,Foxm1在EGFR致癌突變所引起之肺部腫瘤發生中至關重要。
Lung cancer is the leading cause of cancer death worldwide. Many driver mutations for lung cancer have been identified and recent research showed that the spectrum of these oncogenic mutations is variable among racial groups. Molecular epidemiology of mutations in lung cancers showed that prevalence of EGFR oncogenic mutations in Asian patients with non-small cell lung cancers (NSCLCs) is remarkably higher than Caucasian patients. In Taiwan, EGFRL858R missense mutation is predominant found in lung adenocarcinoma (ADC), especially for non-smoker women patients.
Transcription factor Forkhead Box m1 (Foxm1) plays important roles in cellular proliferation, differentiation and tumorigenesis of multiple cell types. Previous research showed that Foxm1 is highly expressed in human lung cancers and we have demonstrated that Foxm1 is required for the initiation of lung tumorigenesis induced by carcinogens or genetically induced lung cancer models. Herein, we are investigating whether Foxm1 is involved in oncogenic EGFRL858R mutation induced lung ADC. We used a genetically engineered mouse model that renders conditional deletion of Foxm1 gene in lung Club cells. The result showed that knockout Foxm1 in airway epithelial cells caused a striking reduction in the size of lesions in EGFRL858R expressing lungs as detected by the pre-clinical micro-computed tomography (microCT), which is consistent with the results from H&E-stained lung pathological examination of cancer. We demonstrated that deletion of Foxm1 prevented the initiation of lung tumors by oncogenic EGFRL858R in lung ADC. Additionally, intratracheal injections of Cre expressing adenovirus (Ad-Cre) to mice with pre-existed EGFR mutated lung tumors caused lung tumor regression. Our results demonstrated that Foxm1 is critical for EGFRL858R -mediated lung tumorigenesis.
中文摘要 - 1 -
ABSTRACT - 2 -
ABBREVIATIONS - 3 -
1. INTRODUCTION - 11 -
1.1 Lung cancer and gene mutation-related lung adenocarcinoma - 11 -
1.2 Roles of Foxm1 related with carcinogenesis - 13 -
1.3 Signal pathway related with EGFR - 15 -
1.4 Mouse models for lung cancer research - 16 -
1.5 MicroCT technique for the study of mouse model - 18 -
1.6 Adenovirus expressing Cre-recombinase (Ad-Cre) and intratracheal injection for gene knockout - 20 -
2. OBJECTIVE OF THIS STUDY - 22 -
3. MATERIAL AND METHODS: - 23 -
3.1 Mouse preparation - 23 -
3.1.1 List of mouse source - 23 -
3.1.2 Quadruple tangencies mice model - 23 -
3.1.3 Keeping and breeding mice - 23 -
3.1.4 Breeding strategy of transgenic mice for conditional gene activation using inducible Cre mice - 24 -
3.1.5 Mouse genomic DNA extraction and genotyping - 24 -
3.2 MicroCT procedures - 27 -
3.2.1 Preparation of micro-CT imaging - 27 -
3.2.2 MicroCT image acquisition - 27 -
3.2.3 MicroCT image analysis - 28 -
3.3 processing of mouse tissue samples - 28 -
3.3.1 Harvesting mouse lungs - 28 -
3.3.1 Total lung RNA extraction - 29 -
3.3.2 Paraffin blocks embedding - 29 -
3.3.3 Hematoxylin and Eosin (H&E) staining - 29 -
3.3.4 Quantitative analysis mRNA expression by Real-time PCR - 29 -
3.4 Adenovirus infection - 30 -
3.4.1 Construction of Cre expression adenovirus - 30 -
3.4.2 Cell culture - 31 -
3.4.3 preparation of Recombinant Adenovirus - 31 -
3.4.4 Intratracheal injection for C/EGFR/Foxm1fl/fl transgenic mice - 32 -
3.5 Statistical analysis - 32 -
4. RESULT - 33 -
4.1 Generation of Doxycycline inducible Foxm1 expression in quadruple tangencies mice was switched on by doxycycline treatment. - 33 -
4.2 Inducible knock out Foxm1 prolong the surviving of the oncogenic EGFRL858R mutant lung cancer model mice - 34 -
4.3 Inducible knock out Foxm1 could significant induce the lung tissue tumor in the oncogenic EGFRL858R mutant lung cancer model by microCT image analysis - 35 -
4.4 Comparison of microCT image with histopathology shows the induction of tumorigenesis through the Foxm1 knock out by the Cre expression - 36 -
4.5 Lung adenocarcinoma induction of intratracheal injection adenovirus expressing Cre-recombinase by knock out Foxm1 expression - 36 -
5. DISCUSSION - 38 -
6. ACKNOWLEDGEMENTS - 43 -
7. REFERENCES - 44 -
8. FIGURE - 52 -
Figure 1. Schematic diagram of EGFRL858R transgenic mice conditional activation by Tet-On system - 52 -
Figure 2. The breeding strategy of conditional activation of EGFR and knockout Foxm1 through Tet-on system - 53 -
Figure 3. The Schematic of expression of Foxm1 knock out in EGFR mutant mice model - 54 -
Figure 4. Increase of weight in EGFR-mutated mouse lungs - 55 -
Figure 5. Kaplan–Meier survival curve for mouse expressing oncogenic EGFR w/ and w/o Foxm1 - 56 -
Figure 6. micro-computed tomography (microCT) shows a decrease of lung tumor loading in C/EGFR/Foxm1 KO mice with 3.5 month Dox diet - 57 -
Figure 7. micro-computed tomography (microCT) shows a decrease of lung tumor loading in C/EGFR/Foxm1 KO mice with 5 month Dox diet - 58 -
Figure 8. Quantitative analysis for microCT images of lung tumor loading in mice - 59 -
Figure 9. Percentage of soft tissue volume/ total lung volume in mice after Dox treatment for 3.5 months - 60 -
Figure 10. Percentage of soft tissue volume/ total lung volume in mice after Dox treatment for 5 months - 61 -
Figure 11. H&E staining of lungs for histopathological analysis in 3.5months Dox diet mice - 62 -
Figure 12. H&E staining of lungs for histopathological analysis in 5months Dox diet mice - 63 -
Figure 13. The RNA levels of C/EGFR/Foxm1fl/fl ,C/EGFR/Foxm1-/- in mouse lungs - 64 -
Figure 14. Schematic diagram of using Cre-recombinase expressing adenovirus (ad-Cre) to induce Foxm1 deletion in EGFR expressing lung tumor - 65 -
Figure 15. The procedure for Ad-Cre intratracheal injection (IT injection) and lung tumor microCT image analysis - 66 -
Figure 16. Decreased tumor loading in EGFRL858R mouse lungs after Ad-Cre IT treatment - 67 -
Figure 17. Model for EGFRL858R missense mutation related with Foxm1 expression - 68 -
Figure s1. The lung appearance of C/EGFR/Foxm1 fl/fl and C/EGFR/Foxm1-/- transgenic mice - 69 -
Figure s2. Increase of weight in EGFR-mutated mouse lungs with different gender - 70 -
Figure s3. Kaplan–Meier survival curve for EGFR mutant male-mouse - 71 -
Figure s4. Kaplan–Meier survival curve for EGFR mutant female-mouse - 72 -
Figure s5. Three-dimension of littermate control mouse treat with 3.5 months Dox diet - 73 -
Figure s6. Three-dimension of C/EGFR/Foxm1fl/fl mouse treat with 3.5 months Dox diet - 74 -
Figure s7. Three-dimension of C/EGFR/Foxm1-/- mouse treat with 3.5 months Dox diet - 75 -
Figure s8. Three-dimension of C/EGFR/Foxm1fl/fl mouse treat with 5 months Dox diet - 76 -
Figure s9. Three-dimension of C/EGFR/Foxm1-/- mouse treat with 5 months Dox diet - 77 -
Figure s10. Image captured by the microCT of littermate control with Dox 3.5 months. - 79 -
Figure s11. Image captured by the microCT of C/EGFR/Foxm1 fl/fl with Dox 3.5 months - 81 -
Figure s12. Image captured by the microCT of C/EGFR/Foxm1 -/- with Dox 3.5 months - 83 -
Figure s13. Image captured by the microCT of C/EGFR/Foxm1 fl/fl and C/EGFR/Foxm1 -/- with Dox 5 months - 85 -
Figure s14. Distribution of three EGFR mutant mice groups in Dox diet treatment 3.5 months - 86 -
Figure s15. Distribution of three groups EGFR mutant mice groups in Dox diet treatment 5 months - 87 -
Figure s16. The method of comparison of before/after Ad-Cre IT injection of mice - 88 -
9. TABLES - 89 -
Table 1. Full list of EGFR mutant mice model - 91 -
Table 2. Full list of analysis in mouse weight and lung weight - 92 -
Table 3. Full list of quantitative analysis in total lung volume and soft tissue volume - 94 -
10. APPENDIX - 95 -
Figure. a1: MicroCT Image Reconstruction and Voxels - 95 -
Figure. a2: Calculation of CT number - 96 -
Figure. a3: Hounsfield Unit Scale - 97 -
1. Siegel, R.L., K.D. Miller, and A. Jemal, Cancer statistics, 2016. CA: A Cancer Journal for Clinicians, 2016. 66(1): p. 7-30.
2. Miller, Y.E., Pathogenesis of Lung Cancer: 100 Year Report. American Journal of Respiratory Cell and Molecular Biology, 2005. 33(3): p. 216-223.
3. Wakamatsu, N., et al., Overview of the Molecular Carcinogenesis of Mouse Lung Tumor Models of Human Lung Cancer. Toxicologic pathology, 2007. 35(1): p. 75-80.
4. Husain AN, Kumar V. The lung. In: Kumar V, Abbas AK, Fausto N, editors. Pathologic Basis of Disease. Elsevier; Philadelphia, PA: 2005. pp. 711–72.
5. Chen, Z., et al., Non-small-cell lung cancers: a heterogeneous set of diseases. Nat Rev Cancer, 2014. 14(8): p. 535-546.
6. Davidson, M.R., A.F. Gazdar, and B.E. Clarke, The pivotal role of pathology in the management of lung cancer. Journal of Thoracic Disease, 2013. 5(Suppl 5): p. S463-S478.
7. Bronte, G., et al., Driver mutations and differential sensitivity to targeted therapies: a new approach to the treatment of lung adenocarcinoma. Cancer Treatment Reviews, 2010. 36, Supplement 3: p. S21-S29.
8. Midha, A., S. Dearden, and R. McCormack, EGFR mutation incidence in non-small-cell lung cancer of adenocarcinoma histology: a systematic review and global map by ethnicity (mutMapII). American Journal of Cancer Research, 2015. 5(9): p. 2892-2911.
9. B. Han, S.T., K. Hagiwara, N. Normanno, L. Wulandari, L. Konstantin Konstantinovich, A. Hudoyo, M. Ratcliffe, R. McCormack, M. Reck, Determining the prevalence of EGFR mutations in Asian and Russian patients (pts) with advanced non-small-cell lung cancer (aNSCLC) of adenocarcinoma (ADC) and non-ADC histology: IGNITE study. Annals of Oncology (2015) 26 (suppl_1): 29-44. 10.1093/annonc/mdv050.
10. Hsu, K.H., et al., Identification of five driver gene mutations in patients with treatment-naive lung adenocarcinoma in Taiwan. PLoS One, 2015. 10(3): p. e0120852.
11. Ren, S., et al., Analysis of driver mutations in female non-smoker Asian patients with pulmonary adenocarcinoma. Cell Biochem Biophys, 2012. 64(2): p. 155-60.
12. Herbst, R.S., Review of epidermal growth factor receptor biology. Int J Radiat Oncol Biol Phys, 2004. 59(2 Suppl): p. 21-6.
13. Burgess, A.W., et al., An Open-and-Shut Case? Recent Insights into the Activation of EGF/ErbB Receptors. Molecular Cell, 2003. 12(3): p. 541-552.
14. Yarden, Y. and J. Schlessinger, Epidermal growth factor induces rapid, reversible aggregation of the purified epidermal growth factor receptor. Biochemistry, 1987. 26(5): p. 1443-1451.
15. Ladanyi, M. and W. Pao, Lung adenocarcinoma: guiding EGFR-targeted therapy and beyond. Mod Pathol, 0000. 21(S2): p. S16-S22.
16. Inukai, M., et al., Presence of Epidermal Growth Factor Receptor Gene T790M Mutation as a Minor Clone in Non–Small Cell Lung Cancer. Cancer Research, 2006. 66(16): p. 7854-7858.
17. Korver, W., et al., The HumanTRIDENT/HFH-11/FKHL16Gene: Structure, Localization, and Promoter Characterization. Genomics, 1997. 46(3): p. 435-442.
18. Clark, K.L., et al., Co-crystal structure of the HNF-3/fork head DNA-recognition motif resembles histone H5. Nature, 1993. 364(6436): p. 412-20.
19. Ye, H., et al., Hepatocyte nuclear factor 3/fork head homolog 11 is expressed in proliferating epithelial and mesenchymal cells of embryonic and adult tissues. Molecular and Cellular Biology, 1997. 17(3): p. 1626-1641.
20. Kim, I.-M., et al., The Forkhead Box M1 Transcription Factor Is Essential for Embryonic Development of Pulmonary Vasculature. Journal of Biological Chemistry, 2005. 280(23): p. 22278-22286.
21. Xue, Y.J., et al., Overexpression of FoxM1 is associated with tumor progression in patients with clear cell renal cell carcinoma. J Transl Med, 2012. 10: p. 200.
22. Ustiyan, V., et al., Forkhead Box M1 Transcriptional Factor is Required for Smooth Muscle Cells during Embryonic Development of Blood Vessels and Esophagus. Developmental biology, 2009. 336(2): p. 266-279.
23. Wierstra, I. and J. Alves, FOXM1, a typical proliferation-associated transcription factor. Biol Chem, 2007. 388(12): p. 1257-74.
24. Wang, I.C., et al., Forkhead Box M1 Regulates the Transcriptional Network of Genes Essential for Mitotic Progression and Genes Encoding the SCF (Skp2-Cks1) Ubiquitin Ligase. Molecular and Cellular Biology, 2005. 25(24): p. 10875-10894.
25. Teh, M.-T., et al., FOXM1 Is a Downstream Target of Gli1 in Basal Cell Carcinomas. Cancer Research, 2002. 62(16): p. 4773-4780.
26. Penzo, M., et al., Sustained NF-κB activation produces a short-term cell proliferation block in conjunction with repressing effectors of cell cycle progression controlled by E2F or FoxM1. Journal of cellular physiology, 2009. 218(1): p. 215-227.
27. Pandit, B., M. Halasi, and A.L. Gartel, p53 negatively regulates expression of FoxM1. Cell Cycle, 2014. 8(20): p. 3425-3427.
28. Pilarsky, C., et al., Identification and Validation of Commonly Overexpressed Genes in Solid Tumors by Comparison of Microarray Data. Neoplasia (New York, N.Y.), 2004. 6(6): p. 744-750.
29. Costa, R.H., et al., New and unexpected: forkhead meets ARF. Current Opinion in Genetics & Development, 2005. 15(1): p. 42-48.
30. Kalin, T.V., et al., Increased Levels of the FoxM1 Transcription Factor Accelerate Development and Progression of Prostate Carcinomas in both TRAMP and LADY Transgenic Mice. Cancer research, 2006. 66(3): p. 1712-1720.
31. Gemenetzidis, E., et al., FOXM1 Upregulation Is an Early Event in Human Squamous Cell Carcinoma and it Is Enhanced by Nicotine during Malignant Transformation. PLoS ONE, 2009. 4(3): p. e4849.
32. Myatt, S.S. and E.W.F. Lam, The emerging roles of forkhead box (Fox) proteins in cancer. Nat Rev Cancer, 2007. 7(11): p. 847-859.
33. Kim, I.M., et al., The Forkhead Box m1 transcription factor stimulates the proliferation of tumor cells during development of lung cancer. Cancer Res, 2006. 66(4): p. 2153-61.
34. Y. LIU, X.C., Y. GU, L. ZHU, Y. QIAN, D. PEI, W. Zhang, Y. SHU, FOXM1 overexpression is associated with cisplatin resistance in non-small cell lung cancer and mediates sensitivity to cisplatin in A549 cells via the JNK/mitochondrial pathway. NEOPLASMA, 2015. 62: p. No. 1: 61-71.
35. Xu, N., et al., FoxM1 Is Associated with Poor Prognosis of Non-Small Cell Lung Cancer Patients through Promoting Tumor Metastasis. PLoS ONE, 2013. 8(3): p. e59412.
36. Wang, I.C., et al., Foxm1 transcription factor is required for the initiation of lung tumorigenesis by oncogenic KrasG12D. Oncogene, 2014. 33(46): p. 5391-5396.
37. Cui, J., et al., HGF/Met and FOXM1 form a positive feedback loop and render pancreatic cancer cells resistance to Met inhibition and aggressive phenotypes. Oncogene, 2016.
38. Ke, Y., et al., miR-149 Inhibits Non-Small-Cell Lung Cancer Cells EMT by Targeting FOXM1. Biochemistry Research International, 2013. 2013: p. 506731.
39. Hynes, N.E. and H.A. Lane, ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer, 2005. 5(5): p. 341-354.
40. Seshacharyulu, P., et al., Targeting the EGFR signaling pathway in cancer therapy. Expert Opinion on Therapeutic Targets, 2012. 16(1): p. 15-31.
41. Biscardi, J.S., et al., c-Src-mediated Phosphorylation of the Epidermal Growth Factor Receptor on Tyr845 and Tyr1101 Is Associated with Modulation of Receptor Function. Journal of Biological Chemistry, 1999. 274(12): p. 8335-8343.
42. Buday, L. and J. Downward, Epidermal growth factor regulates p21ras through the formation of a complex of receptor, Grb2 adapter protein, and Sos nucleotide exchange factor. Cell, 1993. 73(3): p. 611-620.
43. Diaz-Flores, E., et al., Phospholipase C γ and Phosphoinositide 3-Kinase Link Cytokines to ERK Activation in Hematopoietic Cells With Normal and Oncogenic Kras. Science signaling, 2013. 6(304): p. ra105-ra105.
44. Iida, M., et al., Yes and Lyn play a role in nuclear translocation of the epidermal growth factor receptor. Oncogene, 2013. 32(6): p. 759-767.
45. Zimmer, S., et al., Epidermal growth factor receptor mutations in non-small cell lung cancer influence downstream Akt, MAPK and Stat3 signaling. Journal of Cancer Research and Clinical Oncology, 2008. 135(5): p. 723-730.
46. Mencalha, A.L., et al., Forkhead Box M1 (FoxM1) Gene Is a New STAT3 Transcriptional Factor Target and Is Essential for Proliferation, Survival and DNA Repair of K562 Cell Line. PLoS ONE, 2012. 7(10): p. e48160.
47. Yang, C., et al., FOXM1 promotes the epithelial to mesenchymal transition by stimulating the transcription of Slug in human breast cancer. Cancer Lett, 2013. 340(1): p. 104-12.
48. Cheon, D.-J. and S. Orsulic, Mouse Models of Cancer. Annual Review of Pathology: Mechanisms of Disease, 2011. 6(1): p. 95-119.
49. Stewart, T.A., P.K. Pattengale, and P. Leder, Spontaneous mammary adenocarcinomas in transgenic mice that carry and express MTV/myc fusion genes. Cell, 1984. 38(3): p. 627-637.
50. Politi, K., et al., Lung adenocarcinomas induced in mice by mutant EGF receptors foundin human lung cancers respondto a tyrosine kinase inhibitor orto down-regulation of the receptors. Genes & Development, 2006. 20(11): p. 1496-1510.
51. Li, D., et al., Bronchial and Peripheral Murine Lung Carcinomas Induced by T790M-L858R Mutant EGFR Respond to HKI-272 and Rapamycin Combination Therapy. Cancer Cell, 2007. 12(1): p. 81-93.
52. Kim, I.-M., et al., The Forkhead Box m1 Transcription Factor Stimulates the Proliferation of Tumor Cells during Development of Lung Cancer. Cancer Research, 2006. 66(4): p. 2153-2161.
53. Badea, C.T., et al., In vivo small-animal imaging using micro-CT and digital subtraction angiography. Phys Med Biol, 2008. 53(19): p. R319-50.
54. 陳仁焜, 簡介電腦斷層掃描/正子斷層掃描/單光子放射斷層掃描在臨床前動物實驗的應用. 中華實驗動物協會, 2012. 第25期會訊.
55. Almajdub, M., et al., In-vivo high-resolution X-ray microtomography for liver and spleen tumor assessment in mice. Contrast Media & Molecular Imaging, 2007. 2(2): p. 88-93.
56. Graham, K.C., et al., Noninvasive Quantification of Tumor Volume in Preclinical Liver Metastasis Models Using Contrast-Enhanced X-Ray Computed Tomography. Investigative Radiology, 2008. 43(2): p. 92-99.
57. Simon, B.A., et al., Computed Tomography Studies of Lung Mechanics. Proceedings of the American Thoracic Society, 2005. 2(6): p. 517-521.
58. Leow, C.C., X.-D. Wang, and W.-Q. Gao, Novel method of generating prostate-specific Cre–LoxP gene switching via intraductal delivery of adenovirus. The Prostate, 2005. 65(1): p. 1-9.
59. Hasegawa-Baba, Y., et al., Intratracheal Instillation Methods and the Distribution of Administered Material in the Lung of the Rat. Journal of Toxicologic Pathology, 2014. 27(3-4): p. 197-204.
60. Park, K.-S., et al., Characterization of the cell of origin for small cell lung cancer. Cell Cycle, 2011. 10(16): p. 2806-2815.
61. DuPage, M., A.L. Dooley, and T. Jacks, Conditional mouse lung cancer models using adenoviral or lentiviral delivery of Cre recombinase. Nature protocols, 2009. 4(7): p. 1064-1072.
62. Wang, I.C., et al., Deletion of Forkhead Box M1 Transcription Factor from Respiratory Epithelial Cells Inhibits Pulmonary Tumorigenesis. PLoS ONE, 2009. 4(8): p. e6609.
63. Pallier, K., et al., TWIST1 a New Determinant of Epithelial to Mesenchymal Transition in EGFR Mutated Lung Adenocarcinoma. PLoS ONE, 2012. 7(1): p. e29954.
64. Krupczak-Hollis, K., et al., The mouse Forkhead Box m1 transcription factor is essential for hepatoblast mitosis and development of intrahepatic bile ducts and vessels during liver morphogenesis. Developmental Biology, 2004. 276(1): p. 74-88.
65. Shofer, S., et al., A MicroCT-Based Method for the Measurement of Pulmonary Compliance in Healthy and Bleomycin-Exposed Mice. Experimental lung research, 2007. 33(3-4): p. 169-183.
66. Badea, C.T., et al., Computed Tomography Imaging of Primary Lung Cancer in Mice Using a Liposomal-Iodinated Contrast Agent. PLoS ONE, 2012. 7(4): p. e34496.
67. Ustiyan, V., et al., Foxm1 Transcription Factor Is Critical for Proliferation and Differentiation of Clara Cells during Development of Conducting Airways. Developmental biology, 2012. 370(2): p. 198-212.
68. Prost, S., et al., Adenovirus-mediated Cre deletion of floxed sequences in primary mouse cells is an efficient alternative for studies of gene deletion. Nucleic Acids Research, 2001. 29(16): p. e80-e80.
69. Major, M.L., R. Lepe, and R.H. Costa, Forkhead Box M1B Transcriptional Activity Requires Binding of Cdk-Cyclin Complexes for Phosphorylation-Dependent Recruitment of p300/CBP Coactivators. Molecular and Cellular Biology, 2004. 24(7): p. 2649-2661.
(此全文未開放授權)
電子全文
摘要
 
 
 
 
第一頁 上一頁 下一頁 最後一頁 top
* *