博碩士論文 992404002 詳細資訊




以作者查詢圖書館館藏 以作者查詢臺灣博碩士 以作者查詢全國書目 勘誤回報 、線上人數:6 、訪客IP:3.138.102.178
姓名 吳俊誼(Jun-I Wu)  查詢紙本館藏   畢業系所 生命科學系
論文名稱 Crabp2 透過 HuR 以及 Integrin β1/FAK/ERK 訊 息傳導促進肺癌細胞轉移
(Crabp2 Promotes Metastasis of Lung Cancer Cells via HuR and Integrin β1/FAK/ERK Signaling)
檔案 [Endnote RIS 格式]    [Bibtex 格式]    [相關文章]   [文章引用]   [完整記錄]   [館藏目錄]   [檢視]  [下載]
  1. 本電子論文使用權限為同意立即開放。
  2. 已達開放權限電子全文僅授權使用者為學術研究之目的,進行個人非營利性質之檢索、閱讀、列印。
  3. 請遵守中華民國著作權法之相關規定,切勿任意重製、散佈、改作、轉貼、播送,以免觸法。

摘要(中) 許多類型的癌症腫瘤已經發現會高表現 CRABP2 並且關聯到病人的較差存活率。肺癌腫瘤也已經發現會高表現 CRABP2 ,但是 CRABP2 在肺癌轉移中的角色仍不清楚。本研究發現 Crabp2 在高轉移能力的 C10F4 肺癌細胞的表現會高過低轉移能力的肺癌細胞。我們發現 CRABP2 在臨床檢體的高表現與淋巴轉移,較差的存活率以及較高的復發率有關。抑制 Crabp2 的表現量會降低肺癌細胞的移動,侵襲,懸浮狀態下的存活率,以及老鼠模式中的轉移能力。 在免疫沉澱法中, Crabp2 能夠與 HuR 共同沉澱。 高表現 Crabp2 可以增加 HuR 的表現量,並且HuR可以促進 integrin β1 表現以及 FAK 和 ERK 的磷酸化。 抑制 HuR 或是 integrin β1 的表現,亦或是對細胞處理 FAK 或 ERK 的抑制劑都能夠降低 Crabp2 促進肺癌細胞移動,侵襲,以及懸浮狀態下存活率的能力。 另外,與單獨處理 gemcitbine 或是 irinotecan 相比,抑制 Crabp2 能夠進一步抑制肺癌細胞的生長。 CRABP2 在人類腫瘤檢體中的表現量與細胞壓力 (stress) 標記分子 CHOP 的表現量呈現正相關。 因此,我們的研究顯示出 Crabp2 在肺癌細胞轉移中扮演促進的角色。 CRABP2 可以作為一個潛在的預後標記分子,並且標靶 CRABP2 可以作為一個潛在的抑制肺癌轉移策略。 此外,我們也發現了間隙蛋白 Connexin 30.3 (Cx30.3, GJB4) 會促進肺癌細胞的轉移能力,因此我們也回顧了連接蛋白 (gap junction proteins, connexins)在癌症轉移中的角色。 間隙蛋白是一種四次穿膜的蛋白,能夠在相鄰的兩個細胞中間形成間隙連接 (gap junction) 並且因此促進細胞間的溝通 (gap junctional intercellular communication; GJIC) 。 傳統上認為間隙蛋白的角色在於形成半通道 (hemichannels) 後進一步組裝成間隙連接,並以此促進離子以及小分子的傳遞。 許多研究中均觀察到原位腫瘤細胞中的間隙蛋白表現量下降或是移位到細胞質內,因而造成 GJIC 的缺失。 因此,間隙蛋白一般被認為會抑制腫瘤。 然而,近年的研究發現了間隙蛋白在癌症轉移中可能還扮演不同的角色: 在病人的轉移組織切片中發現了 Connexin 43 (Cx43, GJA1) 以及 Connexin 26 (Cx26, GJB2) 的表現量以及它們出現在細胞膜的比率相較於原位腫瘤有所上升。 Cx43 以及 Cx26 所調控的 GJIC 能夠促進癌細胞的移動能力以及對於胸腔內皮細胞的貼附。 許多研究也報導了間隙蛋白在不同癌症類型中的表現以及功能。 在這裡我們聚焦回顧並且討論 1) 間隙蛋白在臨床檢體上的表現與病人預後的關聯性,2) 間隙蛋白在癌症轉移以及抗藥性中的角色,以及3) 將標靶間隙蛋白的分子作為抑制轉移藥物的應用以及考量。 總結來說,間隙蛋白可以做為潛在的癌症預後標記分子,以及開發干擾癌症轉移以及抗藥性的標的。
摘要(英) Increased CRABP2 levels have been found in various types of cancer, and are associated with poor patients’ survival. Although CRABP2 is found to be overexpressed in lung cancer, its role in metastasis of lung cancer is unclear. In this study, Crabp2 was overexpressed in high-metastatic C10F4 than low-metastatic lung cancer cells. Analysis of clinical samples revealed that high CRABP2 levels were correlated with lymph node metastases, poor overall survival, and increased recurrence. Knockdown of Crabp2 decreased migration, invasion, anoikis resistance, and in vivo metastasis. Crabp2 was co-immunoprecipitated with HuR, and overexpression of Crabp2 increased HuR levels, which promoted integrin β1/FAK/ERK signaling. Inhibition of HuR or integrin β1/FAK/ERK signaling reversed the promoting effect of Crabp2 in migration, invasion, and anoikis resistance. Knockdown of Crabp2 further inhibited the growth of cancer cells as compared with that by gemcitabine or irinotecan alone. The expression of Crabp2 in human lung tumors was correlated with stress marker CHOP. In conclusion, our findings have identified the promoting role of Crabp2 in anoikis resistance and metastasis. CRABP2 may serve as a prognostic marker and targeting CRABP2 may be exploited as a modality to reduce metastasis. In parallel, our laboratory has found the promoting role of Connexin 30.3 (Cx30.3, GJB4) in metastasis of lung cancer cells, and thus we reviewed the role of gap junction protein connexins in cancer metastasis. Connexin, a four-pass transmembrane protein, contributes to the assembly of gap junctions among neighboring cells and thus facilitates gap junctional intercellular communication (GJIC). Traditionally, the roles of connexins were thought to mediate formation of hemichannels and GJIC assembly for transportation of ions and small molecules. Many studies have observed loss of GJIC, due to reduced expression or altered cytoplasmic localization of connexins, in primary tumor cells. Connexins are generally considered tumor-suppressive. However, recent studies of clinical samples suggested a different role of connexins in that expression levels and membrane localization of connexins, including Connexin 43 (Cx43, GJA1) and Connexin 26 (Cx26, GJB2), were found to be enhanced in metastatic lesions of cancer patients. Cx43- and Cx26-mediated GJIC was found to promote cancer cell migration and adhesion to the pulmonary endothelium. Regulatory circuits involved in the induction of connexins and their functional effects have also been reported in various types of cancer. Here we focus on the recent findings in the correlation between the expression of connexins and patients’ prognosis, their roles in metastasis and chemoresistance, as well as the implications and concerns of using connexin-targeting drugs as anti-metastatic therapeutics. Overall, connexins may serve as biomarkers for cancer prognosis and as therapeutic targets for intervening metastasis and chemoresistance.
關鍵字(中) ★ 肺癌
★ 轉移
★ Crabp2
★ HuR
關鍵字(英) ★ Lung cancer
★ Metastasis
★ Crabp2
★ HuR
論文目次 中文摘要 I
Abstract III
Declaration V
Acknowledgments VI
Publications arising from this thesis VII
Table of contents VIII
Abbreviation XII
Chapter I: Introduction 1
Epidemiology of lung cancer 1
Cancer metastasis 1
The role of Crabp2 in cancer progression 2
The role of HuR in cancer progression 2
The role of gap junction proteins connexins in cancer progression 3
Figures 6
Figure I-1. The assembly of connexins into gap junctions. 6
Chapter II: Materials and Methods 7
Cell culture 7
Establishment of high-metastatic subline 7
RNA extraction and real-time RT-PCR 7
Tissue Samples 8
Exon array 8
Antibodies and reagents 8
Western blot 9
Migration and invasion assay 9
Generation of stable cell lines and plasmid construction 9
Anoikis assay 10
Tail vein metastasis assay 10
IHC staining and analysis 11
Cell viability assays 11
Immunoprecipitation 11
Statistical analysis 11
Table 13
Table II-1. The list of primers and oligomers used in this study. 13
Table II-2. Information on antibodies and reagents used in this study. 16
Chapter III: Crabp2 Promotes Metastasis of Lung Cancer Cells via HuR and Integrin β1/FAK/ERK Signaling 18
Rationale 18
Results 18
Establishment of high-metastatic C10F4 lung cancer cells 18
Crabp2 is overexpressed in high-metastatic C10F4 lung cancer cells 19
CRABP2 is associated with tumor progression, poor overall survival, and recurrence of lung cancer patients 20
Knockdown of Crabp2/CRABP2 suppresses migration, invasion, anoikis resistance, and in vivo metastasis 21
Crabp2 promotes integrin β1/FAK/ERK signaling via HuR 22
Crabp2 promotes migration, invasion, and anoikis resistance via HuR and integrin β1/FAK/ERK signaling 23
CRABP2/Crabp2 knockdown has an additive but not synergistic effect on the inhibitory effects of gemcitabine and irinotecan on cell viability 24
Identification of the upstream regulating factor(s) of Crabp2 25
Discussion 26
Figures 29
Figure III-1. C10F4 cells have higher migration and invasion ability than C9F6 cells. 29
Figure III-2. C10F4 cells have higher metastasis ability than C9F6 cells. 30
Figure III-3. Time course of lung metastasis of C10F4 and C9F6 cells. 31
Figure III-4. Crabp2 is overexpressed in C10F4 than C9F6 cells and CRABP2 is a lung tumor-overexpressing gene. 32
Figure III-5. CRABP2 is a lung tumor-overexpressing gene. 34
Figure III-6. Crabp2 is overexpressed in C10F4 cells. 35
Figure III-7. CRABP2 is overexpressed in blood buffy coat RNA samples of N2-N3 patients. 36
Figure III-8. CRABP2 is associated with advanced tumor stage and lymph node status of lung cancer patients. 37
Figure III-9. CRABP2 is associated with poor overall survival of lung cancer patients. 38
Figure III-10. CRABP2 is associated with poor survival of lung cancer patients. 39
Figure III-11. CRABP2 is associated with first progression after surgery of lung cancer patients. 40
Figure III-12. CRABP2 is associated with recurrence of lung cancer patients. 41
Figure III-13. Knockdown of Crabp2/CRABP2. 42
Figure III-14. Knockdown of Crabp2 inhibited proliferation of C10F4 cells. 43
Figure III-15. Knockdown of Crabp2 inhibited migration and invasion of C10F4 cells. 44
Figure III-16. Knockdown of CRABP2 inhibited migration and invasion of H1650 cells. 45
Figure III-17. Knockdown of Crabp2 inhibited anoikis resistance of C10F4 cells. 46
Figure III-18. Knockdown of CRABP2 inhibited anoikis resistance of H1650 cells. 47
Figure III-19. Knockdown of Crabp2 inhibited metastasis of C10F4 cells. 48
Figure III-20. The inhibitory effect of retinoic acid on anoikis resistance of C10F4 cells is irrelative to Crabp2. 49
Figure III-21. Crabp2 is co-immunoprecipitated with HuR. 50
Figure III-22. HuR and ITGB1 are inhibited by knockdown of Crabp2 in C10F4 cells. 51
Figure III-23. HuR and ITGB1 are enhanced by overexpression of Crabp2 in C10F4 cells. 52
Figure III-24. Knockdown of Crabp2/CRABP2 inhibits HuR-ITGB1/FAK/ERK signaling. 53
Figure III-25. Overexpression of Crabp2/CRABP2 promotes HuR-ITGB1/FAK/ERK signaling. 54
Figure III-26. Knockdown of HuR and ITGB1 by siRNAs. 55
Figure III-27. HuR and ITGB1/FAK/ERK signaling are needed by Crabp2 to promote the migration and invasion of C10F4 cells. 56
Figure III-28. HuR is needed by Crabp2 to promote the anoikis resistance of C10F4 cells. 57
Figure III-29. ITGB1/FAK/ERK signaling is needed by Crabp2 to promote the anoikis resistance of C10F4 cells. 58
Figure III-30. C10F4 cells are not sensitive to erlotinib. 59
Figure III-31. H1650 cells are not sensitive to erlotinib. 60
Figure III-32. CRABP2 knockdown has an additive but not synergistic effect on the inhibitory effects of gemcitabine and irinotecan on H1650 cell viability. 61
Figure III-33. Crabp2 knockdown further inhibited the growth of C10F4 cells as compared with that by gemcitabine or irinotecan alone. 62
Figure III-34. Crabp2 is inducible by tunicamycin. 63
Figure III-35. CRABP2, CHOP, E2F1, E2F7 are overexpressed in lung tumors. 64
Figure III-36. High CHOP levels are associated with poor survival and first progression after surgery of lung cancer patients. 65
Figure III-37. CRABP2 is correlated with CHOP in human lung tumors. 66
Figure III-38. CRABP2 is correlated with cell stress marker CHOP, E2F1, and E2F7. 67
Figure III-39. Working hypothesis of this study. 68
Tables 69
Table III-1. Correlation of CRABP2 expression levels with clinicopathologic features of lung tumors. 69
Table III-2. Multivariate survival analysis of CRABP2 in lung cancer. 71
Table III-3. Multivariate survival analysis of ERCC1 in lung cancer. 72
Table III-4. Multivariate survival analysis of p27 in lung cancer. 73
Table III-5. Multivariate survival analysis of RRM1 in lung cancer. 74
Table III-6. CRABP2 mRNA levels in sensitive and resistant lines of seven chemotherapeutics using data from Barrentina. 75
Chapter IV: (Review) Emerging Roles of Gap Junction Proteins Connexins in Cancer Metastasis, Chemoresistance and Clinical Application 77
Rationale 77
Connexin 43: 77
The suppressing roles Connexin 43 in tumorigenesis 77
Increased expression and membrane localization of Connexin 43 in metastatic lesions 79
Connexin 43-mediated GJIC enhances cell-cell adhesion and extravasation 79
Carboxyl-terminal tail of Connexin 43 promotes cell migration via p38 81
MicroRNAs that regulate metastatic behaviors of cancer cells via targeting Connexin 43 82
The transcriptional regulation for Connexin 43 expression 83
Connexin 26: 84
Cytoplasmic Connexin 26 is correlated with lymph node metastasis and poor prognosis 84
Cytoplasmic Connexin 26 promotes tumor growth, EMT, and invasion 84
Increased expression and membrane localization of Connexin 26 in metastatic lesions 85
Connexin 26-mediated GJIC promotes migration as single cells via reducing cell-cell adhesion 85
Connexin 32: 87
Cytoplasmic Connexin 32 promotes proliferation, migration and metastasis 87
Connexin 32-mediated GJIC inhibits migration via enhancing cell-cell aggregation 87
Connexin 31, 31.1, 46, and 30.3: 88
Connexins in tumor stroma: 89
Increased Connexin 43, 26, and 30 levels in tumor stroma 89
Stromal Cx43 promotes migration and invasion via GJIC 90
Effects of connexins on chemotherapy 90
Bystander effects mediated by GJIC 90
The GJIC-independent role of connexins in chemotherapy 91
Factors contributing to the variety of connexin-mediated functions 92
Therapeutic applications of connexins as targets 94
Compounds against connexins as potential anti-metastatic drugs 94
Connexins involved in the anti-metastatic effects of therapeutics 95
Figures 97
Figure IV-1. Functional roles of Cx43 in tumor progression. 97
Figure IV-2. Regulatory circuits of Cx43 in tumor progression. 98
Figure IV-3. Functional roles of Cx26 and Cx32 in tumor progression. 99
Tables 100
Table IV-1. Cx43 expression in clinical samples and its correlation with patients’ clinical outcomes. 100
Table IV-2. Cx26/Cx32 expression in clinical samples and their correlation with patients’ clinical outcomes. 102
Table IV-3. Novel compounds that target connexins to inhibit metastasis. 104
Chapter V: General discussion and future works 106
CRABP2 is a potential blood marker assessing lymph node status of lung cancer patients 106
Genes promoting metastasis could be different from those for primary tumor growth 107
The potential link between the Crabp2-HuR axis and connexins/other proteins regulating metastatic behaviors of lung cancer cells 107
Concerns of targeting connexins and potential strategies 108
To develop the peptide inhibitor for the Crabp2-HuR interaction 109
To validate if Crabp2 regulates Cx32 levels via HuR 110
To validate if the Crabp2 levels of white blood cells can be affected by lung tumor cells 110
To validate if Crabp2 regulates stem cell-like features of lung cancer cells 111
To validate if blood CRABP2 levels are enhanced in early stage lung cancer patients versus healthy people and can serve as an early detection marker for lung cancer 111
參考文獻
參考文獻 [1] M.A. Duggan, W.F. Anderson, S. Altekruse, L. Penberthy, M.E. Sherman, The Surveillance, Epidemiology, and End Results (SEER) Program and Pathology: Toward Strengthening the Critical Relationship, Am. J. Surg. Pathol., 40 (2016) e94–e102.
[2] P.M. de Groot, C.C. Wu, B.W. Carter, R.F. Munden, The epidemiology of lung cancer, Transl Lung Cancer Res, 7 (2018) 220–233.
[3] J.S. Chang, L.-T. Chen, Y.-S. Shan, S.-F. Lin, S.-Y. Hsiao, C.-R. Tsai, S.-J. Yu, H.-J. Tsai, Comprehensive Analysis of the Incidence and Survival Patterns of Lung Cancer by Histologies, Including Rare Subtypes, in the Era of Molecular Medicine and Targeted Therapy: A Nation-Wide Cancer Registry-Based Study From Taiwan, Medicine , 94 (2015) e969.
[4] B.-Y. Wang, J.-Y. Huang, C.-Y. Cheng, C.-H. Lin, J.-L. Ko, Y.-P. Liaw, Lung cancer and prognosis in taiwan: a population-based cancer registry, J. Thorac. Oncol., 8 (2013) 1128–1135.
[5] N. Riggi, M. Aguet, I. Stamenkovic, Cancer Metastasis: A Reappraisal of Its Underlying Mechanisms and Their Relevance to Treatment, Annu. Rev. Pathol., 13 (2018) 117–140.
[6] S. Valastyan, R.A. Weinberg, Tumor metastasis: molecular insights and evolving paradigms, Cell, 147 (2011) 275–292.
[7] Y.-N. Kim, K.H. Koo, J.Y. Sung, U.-J. Yun, H. Kim, Anoikis resistance: an essential prerequisite for tumor metastasis, Int. J. Cell Biol., 2012 (2012) 306879.
[8] M.L. Taddei, E. Giannoni, T. Fiaschi, P. Chiarugi, Anoikis: an emerging hallmark in health and diseases, J. Pathol., 226 (2012) 380–393.
[9] N.M. Fofaria, S.K. Srivastava, STAT3 induces anoikis resistance, promotes cell invasion and metastatic potential in pancreatic cancer cells, Carcinogenesis, 36 (2015) 142–150.
[10] P. Zhang, Y. Song, Y. Sun, X. Li, L. Chen, L. Yang, Y. Xing, AMPK/GSK3β/β-catenin cascade-triggered overexpression of CEMIP promotes migration and invasion in anoikis-resistant prostate cancer cells by enhancing metabolic reprogramming, FASEB J., (2018) fj201701078R.
[11] Y.-L. Tai, L.-C. Chen, T.-L. Shen, Emerging roles of focal adhesion kinase in cancer, Biomed Res. Int., 2015 (2015) 690690.
[12] S.M. Frisch, K. Vuori, E. Ruoslahti, P.Y. Chan-Hui, Control of adhesion-dependent cell survival by focal adhesion kinase, J. Cell Biol., 134 (1996) 793–799.
[13] E. Pachmayr, C. Treese, U. Stein, Underlying Mechanisms for Distant Metastasis - Molecular Biology, Visc Med, 33 (2017) 11–20.
[14] N.L. Collins, M.J. Reginato, J.K. Paulus, D.C. Sgroi, J. Labaer, J.S. Brugge, G1/S cell cycle arrest provides anoikis resistance through Erk-mediated Bim suppression, Mol. Cell. Biol., 25 (2005) 5282–5291.
[15] V.M. Golubovskaya, Focal adhesion kinase as a cancer therapy target, Anticancer Agents Med. Chem., 10 (2010) 735–741.
[16] M. Kohno, J. Pouyssegur, Targeting the ERK signaling pathway in cancer therapy, Ann. Med., 38 (2006) 200–211.
[17] M.E. Roh, M. Cosgrove, K. Gorski, I.S. Hitchcock, Off-targets effects underlie the inhibitory effect of FAK inhibitors on platelet activation: studies using Fak-deficient mice, J. Thromb. Haemost., 11 (2013) 1776–1778.
[18] M. Donovan, B. Olofsson, A.L. Gustafson, L. Dencker, U. Eriksson, The cellular retinoic acid binding proteins, J. Steroid Biochem. Mol. Biol., 53 (1995) 459–465.
[19] Y. Kainov, I. Favorskaya, V. Delektorskaya, G. Chemeris, A. Komelkov, A. Zhuravskaya, L. Trukhanova, E. Zueva, B. Tavitian, N. Dyakova, I. Zborovskaya, E. Tchevkina, CRABP1 provides high malignancy of transformed mesenchymal cells and contributes to the pathogenesis of mesenchymal and neuroendocrine tumors, Cell Cycle, 13 (2014) 1530–1539.
[20] R.-Z. Liu, E. Garcia, D.D. Glubrecht, H.Y. Poon, J.R. Mackey, R. Godbout, CRABP1 is associated with a poor prognosis in breast cancer: adding to the complexity of breast cancer cell response to retinoic acid, Mol. Cancer, 14 (2015) 129.
[21] S.-S. Han, W.J. Kim, Y. Hong, S.-H. Hong, S.-J. Lee, D.R. Ryu, W. Lee, Y.H. Cho, S. Lee, Y.-J. Ryu, J.Y. Won, H. Rhee, J.H. Park, S.J. Jang, J.S. Lee, C.-M. Choi, J.C. Lee, S.D. Lee, Y.-M. Oh, RNA sequencing identifies novel markers of non-small cell lung cancer, Lung Cancer, 84 (2014) 229–235.
[22] S.G. Codreanu, M.D. Hoeksema, R.J.C. Slebos, L.J. Zimmerman, S.M.J. Rahman, M. Li, S.-C. Chen, H. Chen, R. Eisenberg, D.C. Liebler, P.P. Massion, Identification of Proteomic Features To Distinguish Benign Pulmonary Nodules from Lung Adenocarcinoma, J. Proteome Res., 16 (2017) 3266–3276.
[23] Y. Zhang, H. Wang, J. Wang, L. Bao, L. Wang, J. Huo, X. Wang, Global analysis of chromosome 1 genes among patients with lung adenocarcinoma, squamous carcinoma, large-cell carcinoma, small-cell carcinoma, or non-cancer, Cancer Metastasis Rev., 34 (2015) 249–264.
[24] F. Bolognani, A.-I. Gallani, L. Sokol, D.S. Baskin, N. Meisner-Kober, mRNA stability alterations mediated by HuR are necessary to sustain the fast growth of glioma cells, J. Neurooncol., 106 (2012) 531–542.
[25] N.-C. Meisner, W. Filipowicz, Properties of the Regulatory RNA-Binding Protein HuR and its Role in Controlling miRNA Repression, Adv. Exp. Med. Biol., 700 (2011) 106–123.
[26] W. Yan, Y. Zhang, J. Zhang, S.-J. Cho, X. Chen, HuR is necessary for mammary epithelial cell proliferation and polarity at least in part via ΔNp63, PLoS One, 7 (2012) e45336.
[27] J. Wang, Y. Guo, H. Chu, Y. Guan, J. Bi, B. Wang, Multiple functions of the RNA-binding protein HuR in cancer progression, treatment responses and prognosis, Int. J. Mol. Sci., 14 (2013) 10015–10041.
[28] J. Wang, W. Zhao, Y. Guo, B. Zhang, Q. Xie, D. Xiang, J. Gao, B. Wang, Z. Chen, The expression of RNA-binding protein HuR in non-small cell lung cancer correlates with vascular endothelial growth factor-C expression and lymph node metastasis, Oncology, 76 (2009) 420–429.
[29] R. Muralidharan, J. Panneerselvam, A. Chen, Y.D. Zhao, A. Munshi, R. Ramesh, HuR-targeted nanotherapy in combination with AMD3100 suppresses CXCR4 expression, cell growth, migration and invasion in lung cancer, Cancer Gene Ther., 22 (2015) 581–590.
[30] M. Heinonen, A. Hemmes, K. Salmenkivi, K. Abdelmohsen, S.-T. Vilén, M. Laakso, M. Leidenius, T. Salo, S. Hautaniemi, M. Gorospe, P. Heikkilä, C. Haglund, A. Ristimäki, Role of RNA binding protein HuR in ductal carcinoma in situ of the breast, J. Pathol., 224 (2011) 529–539.
[31] A.C. Vreeland, L. Levi, W. Zhang, D.C. Berry, N. Noy, Cellular retinoic acid-binding protein 2 inhibits tumor growth by two distinct mechanisms, J. Biol. Chem., 289 (2014) 34065–34073.
[32] A.C. Vreeland, S. Yu, L. Levi, D. de Barros Rossetto, N. Noy, Transcript stabilization by the RNA-binding protein HuR is regulated by cellular retinoic acid-binding protein 2, Mol. Cell. Biol., 34 (2014) 2135–2146.
[33] D.W. Laird, Life cycle of connexins in health and disease, Biochem. J, 394 (2006) 527–543.
[34] J.L. Esseltine, D.W. Laird, Next-Generation Connexin and Pannexin Cell Biology, Trends Cell Biol., (2016).
[35] S. Boitano, E.R. Dirksen, W.H. Evans, Sequence-specific antibodies to connexins block intercellular calcium signaling through gap junctions, Cell Calcium, 23 (1998) 1–9.
[36] M. Sanson, V. Marcaud, E. Robin, C. Valéry, F. Sturtz, B. Zalc, Connexin 43-mediated bystander effect in two rat glioma cell models, Cancer Gene Ther., 9 (2002) 149–155.
[37] A. Gupta, H. Anderson, A.M. Buo, M.C. Moorer, M. Ren, J.P. Stains, Communication of cAMP by connexin43 gap junctions regulates osteoblast signaling and gene expression, Cell. Signal., 28 (2016) 1048–1057.
[38] L. Garcia-Rodríguez, S. Pérez-Torras, M. Carrió, A. Cascante, I. García-Ribas, A. Mazo, C. Fillat, Connexin-26 is a key factor mediating gemcitabine bystander effect, Mol. Cancer Ther., 10 (2011) 505–517.
[39] H. Goodall, B. Maro, Major loss of junctional coupling during mitosis in early mouse embryos, J. Cell Biol., 102 (1986) 568–575.
[40] L.S. Stein, J. Boonstra, R.C. Burghardt, Reduced cell-cell communication between mitotic and nonmitotic coupled cells, Exp. Cell Res., 198 (1992) 1–7.
[41] M. Vinken, T. Vanhaecke, P. Papeleu, S. Snykers, T. Henkens, V. Rogiers, Connexins and their channels in cell growth and cell death, Cell. Signal., 18 (2006) 592–600.
[42] H.A. Coller, Is cancer a metabolic disease?, Am. J. Pathol., 184 (2014) 4–17.
[43] T. Aasen, M. Mesnil, C.C. Naus, P.D. Lampe, D.W. Laird, Gap junctions and cancer: communicating for 50 years, Nat. Rev. Cancer, 16 (2016) 775–788.
[44] D. Banerjee, Connexin’s Connection in Breast Cancer Growth and Progression, Int. J. Cell Biol., 2016 (2016) 9025905.
[45] S.L. Phillips, C.B. Williams, J.N. Zambrano, C.J. Williams, E.S. Yeh, Connexin 43 in the development and progression of breast cancer: What’s the connection? (Review), Int. J. Oncol., 51 (2017) 1005–1013.
[46] L. Kanczuga-Koda, S. Sulkowski, A. Lenczewski, M. Koda, A. Wincewicz, M. Baltaziak, M. Sulkowska, Increased expression of connexins 26 and 43 in lymph node metastases of breast cancer, J. Clin. Pathol., 59 (2006) 429–433.
[47] B. Tang, Z.-H. Peng, P.-W. Yu, G. Yu, F. Qian, Expression and significance of Cx43 and E-cadherin in gastric cancer and metastatic lymph nodes, Med. Oncol., 28 (2011) 502–508.
[48] Y.W. Zhang, I. Morita, M. Ikeda, K.W. Ma, S. Murota, Connexin43 suppresses proliferation of osteosarcoma U2OS cells through post-transcriptional regulation of p27, Oncogene, 20 (2001) 4138–4149.
[49] S. Sirnes, J. Bruun, M. Kolberg, A. Kjenseth, G.E. Lind, A. Svindland, A. Brech, A. Nesbakken, R.A. Lothe, E. Leithe, E. Rivedal, Connexin43 acts as a colorectal cancer tumor suppressor and predicts disease outcome, Int. J. Cancer, 131 (2012) 570–581.
[50] S.R. Polusani, E.A. Kalmykov, A. Chandrasekhar, S.N. Zucker, B.J. Nicholson, Cell coupling mediated by connexin 26 selectively contributes to reduced adhesivity and increased migration, J. Cell Sci., 129 (2016) 4399–4410.
[51] M.K. Elzarrad, A. Haroon, K. Willecke, R. Dobrowolski, M.N. Gillespie, A.-B. Al-Mehdi, Connexin-43 upregulation in micrometastases and tumor vasculature and its role in tumor cell attachment to pulmonary endothelium, BMC Med., 6 (2008) 20.
[52] K. Stoletov, J. Strnadel, E. Zardouzian, M. Momiyama, F.D. Park, J.A. Kelber, D.P. Pizzo, R. Hoffman, S.R. VandenBerg, R.L. Klemke, Role of connexins in metastatic breast cancer and melanoma brain colonization, J. Cell Sci., 126 (2013) 904–913.
[53] C. Lamiche, J. Clarhaut, P.-O. Strale, S. Crespin, N. Pedretti, F.-X. Bernard, C.C. Naus, V.C. Chen, L.J. Foster, N. Defamie, M. Mesnil, F. Debiais, L. Cronier, The gap junction protein Cx43 is involved in the bone-targeted metastatic behaviour of human prostate cancer cells, Clin. Exp. Metastasis, 29 (2012) 111–122.
[54] J.H.-C. Lin, T. Takano, M.L. Cotrina, G. Arcuino, J. Kang, S. Liu, Q. Gao, L. Jiang, F. Li, H. Lichtenberg-Frate, S. Haubrich, K. Willecke, S.A. Goldman, M. Nedergaard, Connexin 43 Enhances the Adhesivity and Mediates the Invasion of Malignant Glioma Cells, J. Neurosci., 22 (2002) 4302–4311.
[55] W. Zhang, C. Nwagwu, D.M. Le, V.W. Yong, H. Song, W.T. Couldwell, Increased invasive capacity of connexin43-overexpressing malignant glioma cells, J. Neurosurg., 99 (2003) 1039–1046.
[56] R. Oliveira, C. Christov, J.S. Guillamo, S. de Boüard, S. Palfi, L. Venance, M. Tardy, M. Peschanski, Contribution of gap junctional communication between tumor cells and astroglia to the invasion of the brain parenchyma by human glioblastomas, BMC Cell Biol., 6 (2005) 7.
[57] B. Tang, Z.-H. Peng, P.-W. Yu, G. Yu, F. Qian, D.-Z. Zeng, Y.-L. Zhao, Y. Shi, Y.-X. Hao, H.-X. Luo, Aberrant expression of Cx43 is associated with the peritoneal metastasis of gastric cancer and Cx43-mediated gap junction enhances gastric cancer cell diapedesis from peritoneal mesothelium, PLoS One, 8 (2013) e74527.
[58] A.S. Sun, H.C. Yeh, L.H. Wang, Y.P. Huang, H. Maeda, A. Pivazyan, C. Hsu, E.R. Lewis, H.W. Bruckner, T.M. Fasy, Pilot study of a specific dietary supplement in tumor-bearing mice and in stage IIIB and IV non-small cell lung cancer patients, Nutr. Cancer, 39 (2001) 85–95.
[59] D.W. Bahler, J.G. Frelinger, L.W. Harwell, E.M. Lord, Reduced tumorigenicity of a spontaneous mouse lung carcinoma following H-2 gene transfection, Proc. Natl. Acad. Sci. U. S. A., 84 (1987) 4562–4566.
[60] K.-T. Lin, J. Gong, C.-F. Li, T.-H. Jang, W.-L. Chen, H.-J. Chen, L.-H. Wang, Vav3-rac1 signaling regulates prostate cancer metastasis with elevated Vav3 expression correlating with prostate cancer progression and posttreatment recurrence, Cancer Res., 72 (2012) 3000–3009.
[61] Y.-M. Yeh, C.-M. Chuang, K.-C. Chao, L.-H. Wang, MicroRNA-138 suppresses ovarian cancer cell invasion and metastasis by targeting SOX4 and HIF-1α, Int. J. Cancer, 133 (2013) 867–878.
[62] M. Labelle, R.O. Hynes, The initial hours of metastasis: the importance of cooperative host-tumor cell interactions during hematogenous dissemination, Cancer Discov., 2 (2012) 1091–1099.
[63] D.R. Rhodes, J. Yu, K. Shanker, N. Deshpande, R. Varambally, D. Ghosh, T. Barrette, A. Pandey, A.M. Chinnaiyan, ONCOMINE: a cancer microarray database and integrated data-mining platform, Neoplasia, 6 (2004) 1–6.
[64] I.J. Fidler, M.L. Kripke, The challenge of targeting metastasis, Cancer Metastasis Rev., 34 (2015) 635–641.
[65] H. Uramoto, F. Tanaka, Recurrence after surgery in patients with NSCLC, Transl Lung Cancer Res, 3 (2014) 242–249.
[66] B. Győrffy, P. Surowiak, J. Budczies, A. Lánczky, Online survival analysis software to assess the prognostic value of biomarkers using transcriptomic data in non-small-cell lung cancer, PLoS One, 8 (2013) e82241.
[67] J. Li, J. Lin, Y. Luo, M. Kuang, Y. Liu, Multivariate Analysis of Prognostic Biomarkers in Surgically Treated Endometrial Cancer, PLoS One, 10 (2015) e0130640.
[68] M. Paesmans, Prognostic and predictive factors for lung cancer, Breathe, 9 (2012) 112–121.
[69] L.E. Coate, T. John, M.-S. Tsao, F.A. Shepherd, Molecular predictive and prognostic markers in non-small-cell lung cancer, Lancet Oncol., 10 (2009) 1001–1010.
[70] R.-Z. Liu, S. Li, E. Garcia, D.D. Glubrecht, H.Y. Poon, J.C. Easaw, R. Godbout, Association between cytoplasmic CRABP2, altered retinoic acid signaling, and poor prognosis in glioblastoma, Glia, 64 (2016) 963–976.
[71] S. Fischer-Huchzermeyer, A. Dombrowski, C. Hagel, V.F. Mautner, J. Schittenhelm, A. Harder, The Cellular Retinoic Acid Binding Protein 2 Promotes Survival of Malignant Peripheral Nerve Sheath Tumor Cells, Am. J. Pathol., 187 (2017) 1623–1632.
[72] K.M.C. Mohammad Sultan, Retinoid Signaling in Cancer and Its Promise for Therapy, J Carcinog Mutagen, (2013).
[73] N. Mukherjee, P.J. Lager, M.B. Friedersdorf, M.A. Thompson, J.D. Keene, Coordinated posttranscriptional mRNA population dynamics during T-cell activation, Mol. Syst. Biol., 5 (2009) 288.
[74] Y.-C. Teng, C.-F. Lee, Y.-S. Li, Y.-R. Chen, P.-W. Hsiao, M.-Y. Chan, F.-M. Lin, H.-D. Huang, Y.-T. Chen, Y.-M. Jeng, C.-H. Hsu, Q. Yan, M.-D. Tsai, L.-J. Juan, Histone demethylase RBP2 promotes lung tumorigenesis and cancer metastasis, Cancer Res., 73 (2013) 4711–4721.
[75] K.K. Ganguly, S. Pal, S. Moulik, A. Chatterjee, Integrins and metastasis, Cell Adh. Migr., 7 (2013) 251–261.
[76] F.J. Sulzmaier, C. Jean, D.D. Schlaepfer, FAK in cancer: mechanistic findings and clinical applications, Nat. Rev. Cancer, 14 (2014) 598–610.
[77] T. Miyazaki, H. Kato, M. Nakajima, M. Sohda, Y. Fukai, N. Masuda, R. Manda, M. Fukuchi, K. Tsukada, H. Kuwano, FAK overexpression is correlated with tumour invasiveness and lymph node metastasis in oesophageal squamous cell carcinoma, Br. J. Cancer, 89 (2003) 140–145.
[78] W. Guo, F.G. Giancotti, Integrin signalling during tumour progression, Nat. Rev. Mol. Cell Biol., 5 (2004) 816–826.
[79] R. Rosell, L. Crino, K. Danenberg, G. Scagliotti, G. Bepler, M. Taron, V. Alberola, M. Provencio, C. Camps, F. De Marinis, J.J. Sanchez, R. Peñas, Targeted therapy in combination with gemcitabine in non-small cell lung cancer, Semin. Oncol., 30 (2003) 19–25.
[80] Y. Lin, X. Wang, H. Jin, EGFR-TKI resistance in NSCLC patients: mechanisms and strategies, Am. J. Cancer Res., 4 (2014) 411–435.
[81] B. Wills, A.F. Cardona, L. Rojas, A. Ruiz-Patiño, O. Arrieta, N. Reguart, H. Carranza, C. Vargas, J. Otero, L. Corrales, C. Martín, M. Cuello, L.E. Pino, C. Rolfo, R. Rosell, Z.L. Zatarain-Barrón, Latin-American Consortium for the Investigation of Lung Cancer (CLICaP), Survival Outcomes According to TIMP1 and EGFR Expression in Heavily Treated Patients with Advanced Non-small Cell Lung Cancer who Received Biweekly Irinotecan Plus Bevacizumab, Anticancer Res., 37 (2017) 6429–6436.
[82] Y. Xu, M.A. Villalona-Calero, Irinotecan: mechanisms of tumor resistance and novel strategies for modulating its activity, Ann. Oncol., 13 (2002) 1841–1851.
[83] W.-A. Wang, J. Groenendyk, M. Michalak, Endoplasmic reticulum stress associated responses in cancer, Biochim. Biophys. Acta, 1843 (2014) 2143–2149.
[84] A.S. Lee, The glucose-regulated proteins: stress induction and clinical applications, Trends Biochem. Sci., 26 (2001) 504–510.
[85] Y. Xu, Z. Chen, G. Zhang, Y. Xi, R. Sun, X. Wang, W. Wang, F. Chai, X. Li, HSP90B1 overexpression predicts poor prognosis in NSCLC patients, Tumour Biol., 37 (2016) 14321–14328.
[86] B.-Y. Jin, G.-H. Fu, X. Jiang, H. Pan, D.-K. Zhou, X.-Y. Wei, L. Zhou, L. Chung, S.-S. Zheng, CRABP2 and FABP5 identified by 2D DIGE profiling are upregulated in human bladder cancer, Chin. Med. J. , 126 (2013) 3787–3789.
[87] A. Gupta, P. Kessler, J. Rawwas, B.R.G. Williams, Regulation of CRABP-II expression by MycN in Wilms tumor, Exp. Cell Res., 314 (2008) 3663–3668.
[88] A. Gupta, B.R.G. Williams, S.M. Hanash, J. Rawwas, Cellular retinoic acid-binding protein II is a direct transcriptional target of MycN in neuroblastoma, Cancer Res., 66 (2006) 8100–8108.
[89] W. Xiao, H. Hong, A. Awadallah, S. Yu, L. Zhou, W. Xin, CRABP-II is a highly sensitive and specific diagnostic molecular marker for pancreatic ductal adenocarcinoma in distinguishing from benign pancreatic conditions, Hum. Pathol., 45 (2014) 1177–1183.
[90] A. Toyama, A. Suzuki, T. Shimada, C. Aoki, Y. Aoki, Y. Umino, Y. Nakamura, D. Aoki, T.-A. Sato, Proteomic characterization of ovarian cancers identifying annexin-A4, phosphoserine aminotransferase, cellular retinoic acid-binding protein 2, and serpin B5 as histology-specific biomarkers, Cancer Sci., 103 (2012) 747–755.
[91] A.P. Percicote, G.L. Mardegan, E.S. Gugelmim, S.O. Ioshii, A.P. Kuczynski, S. Nagashima, L. de Noronha, Tissue expression of retinoic acid receptor alpha and CRABP2 in metastatic nephroblastomas, Diagn. Pathol., 13 (2018) 9.
[92] A.H. Cory, T.C. Owen, J.A. Barltrop, J.G. Cory, Use of an aqueous soluble tetrazolium/formazan assay for cell growth assays in culture, Cancer Commun., 3 (1991) 207–212.
[93] D.L. Crowe, R. Kim, R.A.S. Chandraratna, Retinoic acid differentially regulates cancer cell proliferation via dose-dependent modulation of the mitogen-activated protein kinase pathway, Mol. Cancer Res., 1 (2003) 532–540.
[94] T.T. Schug, D.C. Berry, N.S. Shaw, S.N. Travis, N. Noy, Opposing effects of retinoic acid on cell growth result from alternate activation of two different nuclear receptors, Cell, 129 (2007) 723–733.
[95] R.M. Connolly, N.K. Nguyen, S. Sukumar, Molecular pathways: current role and future directions of the retinoic acid pathway in cancer prevention and treatment, Clin. Cancer Res., 19 (2013) 1651–1659.
[96] J.L. Napoli, Cellular retinoid binding-proteins, CRBP, CRABP, FABP5: Effects on retinoid metabolism, function and related diseases, Pharmacol. Ther., 173 (2017) 19–33.
[97] B. Weber, H. Hager, B.S. Sorensen, T. McCulloch, A. Mellemgaard, A.A. Khalil, E. Nexo, P. Meldgaard, EGFR mutation frequency and effectiveness of erlotinib: a prospective observational study in Danish patients with non-small cell lung cancer, Lung Cancer, 83 (2014) 224–230.
[98] H.A. Yu, M.E. Arcila, M.D. Hellmann, M.G. Kris, M. Ladanyi, G.J. Riely, Poor response to erlotinib in patients with tumors containing baseline EGFR T790M mutations found by routine clinical molecular testing, Ann. Oncol., 25 (2014) 423–428.
[99] C. Willmore-Payne, J.A. Holden, C.T. Wittwer, L.J. Layfield, The use of EGFR exon 19 and 21 unlabeled DNA probes to screen for activating mutations in non-small cell lung cancer, J. Biomol. Tech., 19 (2008) 217–224.
[100] M.L. Sos, M. Koker, B.A. Weir, S. Heynck, R. Rabinovsky, T. Zander, J.M. Seeger, J. Weiss, F. Fischer, P. Frommolt, K. Michel, M. Peifer, C. Mermel, L. Girard, M. Peyton, A.F. Gazdar, J.D. Minna, L.A. Garraway, H. Kashkar, W. Pao, et al., PTEN loss contributes to erlotinib resistance in EGFR-mutant lung cancer by activation of Akt and EGFR, Cancer Res., 69 (2009) 3256–3261.
[101] M. Prieto-Vila, R.-U. Takahashi, W. Usuba, I. Kohama, T. Ochiya, Drug Resistance Driven by Cancer Stem Cells and Their Niche, Int. J. Mol. Sci., 18 (2017).
[102] M. Ghosh, H.L. Aguila, J. Michaud, Y. Ai, M.-T. Wu, A. Hemmes, A. Ristimaki, C. Guo, H. Furneaux, T. Hla, Essential role of the RNA-binding protein HuR in progenitor cell survival in mice, J. Clin. Invest., 119 (2009) 3530–3543.
[103] M. Singh, A.R. Martinez, S. Govindaraju, B.S. Lee, HuR inhibits apoptosis by amplifying Akt signaling through a positive feedback loop, J. Cell. Physiol., 228 (2013) 182–189.
[104] X. Guan, Cancer metastases: challenges and opportunities, Acta Pharm Sin B, 5 (2015) 402–418.
[105] G. Spagnol, A.J. Trease, L. Zheng, M. Gutierrez, I. Basu, C. Sarmiento, G. Moore, M. Cervantes, P.L. Sorgen, Connexin43 Carboxyl-Terminal Domain Directly Interacts with β-Catenin, Int. J. Mol. Sci., 19 (2018).
[106] Z. Ai, A. Fischer, D.C. Spray, A.M. Brown, G.I. Fishman, Wnt-1 regulation of connexin43 in cardiac myocytes, J. Clin. Invest., 105 (2000) 161–171.
[107] R.P. Huang, M.Z. Hossain, R. Huang, J. Gano, Y. Fan, A.L. Boynton, Connexin 43 (cx43) enhances chemotherapy-induced apoptosis in human glioblastoma cells, Int. J. Cancer, 92 (2001) 130–138.
[108] H.-T. Xu, Q.-C. Li, Y.-X. Zhang, Y. Zhao, Y. Liu, Z.-Q. Yang, E.-H. Wang, Connexin 43 recruits E-cadherin expression and inhibits the malignant behaviour of lung cancer cells, Folia Histochem. Cytobiol., 46 (2008) 315–321.
[109] K.C. Alaga, M. Crawford, L. Dagnino, D.W. Laird, Aberrant Cx43 Expression and Mislocalization in Metastatic Human Melanomas, J. Cancer, 8 (2017) 1123–1128.
[110] L. Kańczuga-Koda, M. Sulkowska, M. Koda, J. Reszeć, W. Famulski, M. Baltaziak, S. Sulkowski, Expression of connexin 43 in breast cancer in comparison with mammary dysplasia and the normal mammary gland, Folia Morphol. , 62 (2003) 439–442.
[111] Q.-L. Liang, B.-R. Wang, G.-Q. Chen, G.-H. Li, Y.-Y. Xu, Clinical significance of vascular endothelial growth factor and connexin43 for predicting pancreatic cancer clinicopathologic parameters, Med. Oncol., 27 (2010) 1164–1170.
[112] Zhang Y., Xu H., Wang E., [Expressions of connexin 43 and E-cadherin and their correlation in non-small cell lung cancer], Zhongguo Fei Ai Za Zhi, 8 (2005) 103–106.
[113] Sun W.-H., Liu H.-M., Li Y.-J., Ji X.-R., Liang D.-P., [A study of the relationship between the expression of connexin43, E-cadherin and biological behaviors of human laryngeal cancer], Zhonghua Er Bi Yan Hou Ke Za Zhi, 39 (2004) 293–297.
[114] L. Puzzo, R. Caltabiano, R. Parenti, S. Trapasso, E. Allegra, Connexin 43 (Cx43) Expression in Laryngeal Squamous Cell Carcinomas: Preliminary Data on Its Possible Prognostic Role, Head Neck Pathol., 10 (2016) 292–297.
[115] J.W. Smyth, T.-T. Hong, D. Gao, J.M. Vogan, B.C. Jensen, T.S. Fong, P.C. Simpson, D.Y.R. Stainier, N.C. Chi, R.M. Shaw, Limited forward trafficking of connexin 43 reduces cell-cell coupling in stressed human and mouse myocardium, J. Clin. Invest., 120 (2010) 266–279.
[116] J.E. Klaunig, L.M. Kamendulis, The role of oxidative stress in carcinogenesis, Annu. Rev. Pharmacol. Toxicol., 44 (2004) 239–267.
[117] V. Sosa, T. Moliné, R. Somoza, R. Paciucci, H. Kondoh, M.E. LLeonart, Oxidative stress and cancer: an overview, Ageing Res. Rev., 12 (2013) 376–390.
[118] J. Ming, Y. Zhou, J. Du, S. Fan, B. Pan, Y. Wang, L. Fan, J. Jiang, miR-381 suppresses C/EBPα-dependent Cx43 expression in breast cancer cells, Biosci. Rep., 35 (2015).
[119] Z.-J. Lin, J. Ming, L. Yang, J.-Z. Du, N. Wang, H.-J. Luo, Mechanism of Regulatory Effect of MicroRNA-206 on Connexin 43 in Distant Metastasis of Breast Cancer, Chin. Med. J. , 129 (2016) 424–434.
[120] J. Ming, Y. Zhou, J. Du, S. Fan, B. Pan, Y. Wang, L. Fan, J. Jiang, Identification of miR-200a as a novel suppressor of connexin 43 in breast cancer cells, Biosci. Rep., 35 (2015).
[121] F. Gava, L. Rigal, O. Mondesert, E. Pesce, B. Ducommun, V. Lobjois, Gap junctions contribute to anchorage-independent clustering of breast cancer cells, BMC Cancer, 18 (2018) 221.
[122] L.A.B. Elias, D.D. Wang, A.R. Kriegstein, Gap junction adhesion is necessary for radial migration in the neocortex, Nature, 448 (2007) 901–907.
[123] M.E. el-Sabban, B.U. Pauli, Cytoplasmic dye transfer between metastatic tumor cells and vascular endothelium, J. Cell Biol., 115 (1991) 1375–1382.
[124] A. Zhang, M. Hitomi, N. Bar-Shain, Z. Dalimov, L. Ellis, K.K. Velpula, G.C. Fraizer, R.G. Gourdie, J.D. Lathia, Connexin 43 expression is associated with increased malignancy in prostate cancer cell lines and functions to promote migration, Oncotarget, 6 (2015) 11640–11651.
[125] J. Behrens, P. Kameritsch, S. Wallner, U. Pohl, K. Pogoda, The carboxyl tail of Cx43 augments p38 mediated cell migration in a gap junction-independent manner, Eur. J. Cell Biol., 89 (2010) 828–838.
[126] S. Ghosh, A. Kumar, R.P. Tripathi, S. Chandna, Connexin-43 regulates p38-mediated cell migration and invasion induced selectively in tumour cells by low doses of γ-radiation in an ERK-1/2-independent manner, Carcinogenesis, 35 (2014) 383–395.
[127] A.J. Siller-Jackson, S. Burra, S. Gu, X. Xia, L.F. Bonewald, E. Sprague, J.X. Jiang, Adaptation of connexin 43-hemichannel prostaglandin release to mechanical loading, J. Biol. Chem., 283 (2008) 26374–26382.
[128] G.M. Yusubalieva, V.P. Baklaushev, O.I. Gurina, Y.A. Zorkina, I.L. Gubskii, G.L. Kobyakov, A.V. Golanov, S.A. Goryainov, G.E. Gorlachev, A.N. Konovalov, A.A. Potapov, V.P. Chekhonin, Treatment of poorly differentiated glioma using a combination of monoclonal antibodies to extracellular connexin-43 fragment, temozolomide, and radiotherapy, Bull. Exp. Biol. Med., 157 (2014) 510–515.
[129] L. Franco, E. Zocchi, C. Usai, L. Guida, S. Bruzzone, A. Costa, A. De Flora, Paracrine roles of NAD+ and cyclic ADP-ribose in increasing intracellular calcium and enhancing cell proliferation of 3T3 fibroblasts, J. Biol. Chem., 276 (2001) 21642–21648.
[130] J.M. Rhett, E.S. Yeh, The Potential for Connexin Hemichannels to Drive Breast Cancer Progression through Regulation of the Inflammatory Response, Int. J. Mol. Sci., 19 (2018).
[131] K.A. Schalper, D. Carvajal-Hausdorf, M.P. Oyarzo, Possible role of hemichannels in cancer, Front. Physiol., 5 (2014) 237.
[132] Y. Fu, Z.-M. Shao, Q.-Z. He, B.-Q. Jiang, Y. Wu, Z.-G. Zhuang, Hsa-miR-206 represses the proliferation and invasion of breast cancer cells by targeting Cx43, Eur. Rev. Med. Pharmacol. Sci., 19 (2015) 2091–2104.
[133] Z. Wang, D.-Z. Wang, G.C.T. Pipes, E.N. Olson, Myocardin is a master regulator of smooth muscle gene expression, Proc. Natl. Acad. Sci. U. S. A., 100 (2003) 7129–7134.
[134] H. Li, Y. Xiang, L.-J. Fan, X.-Y. Zhang, J.-P. Li, C.-X. Yu, L.-Y. Bao, D.-S. Cao, W.-B. Xing, X.-H. Liao, T.-C. Zhang, Myocardin inhibited the gap protein connexin 43 via promoted miR-206 to regulate vascular smooth muscle cell phenotypic switch, Gene, 616 (2017) 22–30.
[135] B. Davidson, V.M. Abeler, M. Førsund, A. Holth, Y. Yang, Y. Kobayashi, L. Chen, G.B. Kristensen, I.-M. Shih, T.-L. Wang, Gene expression signatures of primary and metastatic uterine leiomyosarcoma, Hum. Pathol., 45 (2014) 691–700.
[136] D. Ryszawy, M. Sarna, M. Rak, K. Szpak, S. Kędracka-Krok, M. Michalik, M. Siedlar, E. Zuba-Surma, K. Burda, W. Korohoda, Z. Madeja, J. Czyż, Functional links between Snail-1 and Cx43 account for the recruitment of Cx43-positive cells into the invasive front of prostate cancer, Carcinogenesis, 35 (2014) 1920–1930.
[137] Y. Wang, J. Liu, X. Ying, P.C. Lin, B.P. Zhou, Twist-mediated Epithelial-mesenchymal Transition Promotes Breast Tumor Cell Invasion via Inhibition of Hippo Pathway, Sci. Rep., 6 (2016) 24606.
[138] Y. Naoi, Y. Miyoshi, T. Taguchi, S.J. Kim, T. Arai, Y. Tamaki, S. Noguchi, Connexin26 expression is associated with lymphatic vessel invasion and poor prognosis in human breast cancer, Breast Cancer Res. Treat., 106 (2007) 11–17.
[139] K. Ezumi, H. Yamamoto, K. Murata, M. Higashiyama, B. Damdinsuren, Y. Nakamura, N. Kyo, J. Okami, C.Y. Ngan, I. Takemasa, M. Ikeda, M. Sekimoto, N. Matsuura, H. Nojima, M. Monden, Aberrant Expression of Connexin 26 Is Associated with Lung Metastasis of Colorectal Cancer, Clin. Cancer Res., 14 (2008) 677–684.
[140] Y. Naoi, Y. Miyoshi, T. Taguchi, S.J. Kim, T. Arai, N. Maruyama, Y. Tamaki, S. Noguchi, Connexin26 expression is associated with aggressive phenotype in human papillary and follicular thyroid cancers, Cancer Lett., 262 (2008) 248–256.
[141] T. Inose, H. Kato, H. Kimura, A. Faried, N. Tanaka, M. Sakai, A. Sano, M. Sohda, M. Nakajima, Y. Fukai, T. Miyazaki, N. Masuda, M. Fukuchi, H. Kuwano, Correlation between connexin 26 expression and poor prognosis of esophageal squamous cell carcinoma, Ann. Surg. Oncol., 16 (2009) 1704–1710.
[142] J. Yang, G. Qin, M. Luo, J. Chen, Q. Zhang, L. Li, L. Pan, S. Qin, Reciprocal positive regulation between Cx26 and PI3K/Akt pathway confers acquired gefitinib resistance in NSCLC cells via GJIC-independent induction of EMT, Cell Death Dis., 6 (2015) e1829.
[143] A. Ito, F. Katoh, T.R. Kataoka, M. Okada, N. Tsubota, H. Asada, K. Yoshikawa, S. Maeda, Y. Kitamura, H. Yamasaki, H. Nojima, A role for heterologous gap junctions between melanoma and endothelial cells in metastasis, J. Clin. Invest., 105 (2000) 1189–1197.
[144] A. Duflot-Dancer, M. Mesnil, H. Yamasaki, Dominant-negative abrogation of connexin-mediated cell growth control by mutant connexin genes, Oncogene, 15 (1997) 2151–2158.
[145] L. Kanczuga-Koda, M. Sulkowska, M. Koda, R. Rutkowski, S. Sulkowski, Increased expression of gap junction protein--connexin 32 in lymph node metastases of human ductal breast cancer, Folia Histochem. Cytobiol., 45 Suppl 1 (2007) S175–80.
[146] V. Krutovskikh, G. Mazzoleni, N. Mironov, Y. Omori, A.M. Aguelon, M. Mesnil, F. Berger, C. Partensky, H. Yamasaki, Altered homologous and heterologous gap-junctional intercellular communication in primary human liver tumors associated with aberrant protein localization but not gene mutation of connexin 32, Int. J. Cancer, 56 (1994) 87–94.
[147] Q. Li, Y. Omori, Y. Nishikawa, T. Yoshioka, Y. Yamamoto, K. Enomoto, Cytoplasmic accumulation of connexin32 protein enhances motility and metastatic ability of human hepatoma cells in vitro and in vivo, Int. J. Cancer, 121 (2007) 536–546.
[148] J. Yang, B. Liu, Q. Wang, D. Yuan, X. Hong, Y. Yang, L. Tao, Connexin 32 and its derived homotypic gap junctional intercellular communication inhibit the migration and invasion of transfected HeLa cells via enhancement of intercellular adhesion, Mol. Med. Rep., 4 (2011) 971–979.
[149] Y. Yang, S.-K. Qin, Q. Wu, Z.-S. Wang, R.-S. Zheng, X.-H. Tong, H. Liu, L. Tao, X.-D. He, Connexin-dependent gap junction enhancement is involved in the synergistic effect of sorafenib and all-trans retinoic acid on HCC growth inhibition, Oncol. Rep., 31 (2014) 540–550.
[150] E. Fujimoto, H. Sato, S. Shirai, Y. Nagashima, K. Fukumoto, H. Hagiwara, E. Negishi, K. Ueno, Y. Omori, H. Yamasaki, K. Hagiwara, T. Yano, Connexin32 as a tumor suppressor gene in a metastatic renal cell carcinoma cell line, Oncogene, 24 (2005) 3684–3690.
[151] H. Sato, H. Hagiwara, H. Senba, K. Fukumoto, Y. Nagashima, H. Yamasaki, K. Ueno, T. Yano, The inhibitory effect of connexin 32 gene on metastasis in renal cell carcinoma, Mol. Carcinog., 47 (2008) 403–409.
[152] H. Hagiwara, H. Sato, S. Shirai, S. Kobayashi, K. Fukumoto, T. Ishida, T. Seki, T. Ariga, T. Yano, Connexin 32 down-regulates the fibrinolytic factors in metastatic renal cell carcinoma cells, Life Sci., 78 (2006) 2249–2254.
[153] Y. Yang, N. Zhang, J. Zhu, X.-T. Hong, H. Liu, Y.-R. Ou, F. Su, R. Wang, Y.-M. Li, Q. Wu, Downregulated connexin32 promotes EMT through the Wnt/β-catenin pathway by targeting Snail expression in hepatocellular carcinoma, Int. J. Oncol., 50 (2017) 1977–1988.
[154] B. Zhao, W. Zhao, Y. Wang, Y. Xu, J. Xu, K. Tang, S. Zhang, Z. Yin, Q. Wu, X. Wang, Connexin32 regulates hepatoma cell metastasis and proliferation via the p53 and Akt pathways, Oncotarget, 6 (2015) 10116–10133.
[155] D. Zhang, C. Chen, Y. Li, X. Fu, Y. Xie, Y. Li, Y. Huang, Cx311 acts as a tumour suppressor in non-small cell lung cancer (NSCLC) cell lines through inhibition of cell proliferation and metastasis, J. Cell. Mol. Med., 16 (2012) 1047–1059.
[156] L. Xu, S.-W. Chen, X.-Y. Qi, X.-X. Li, Y.-B. Sun, Ginsenoside improves papillary thyroid cancer cell malignancies partially through upregulating connexin 31, Kaohsiung J. Med. Sci., 34 (2018) 313–320.
[157] Jin J., Li C., You J., Zhang B., [miR-610 suppresses lung cancer cell proliferation and invasion by targeting GJA3], Zhonghua Zhong Liu Za Zhi, 36 (2014) 405–411.
[158] Y.-P. Lin, J.-I. Wu, C.-W. Tseng, H.-J. Chen, L.-H. Wang, Gjb4 serves as a novel biomarker for lung cancer and promotes metastasis and chemoresistance via Src activation, Oncogene, (2018).
[159] J. Radić, B. Krušlin, M. Šamija, M. Ulamec, M. Milošević, M. Jazvić, I. Šamija, J.J. Grah, A. Bolanča, Z. Kusić, Connexin 43 Expression in Primary Colorectal Carcinomas in Patients with Stage III and IV Disease, Anticancer Res., 36 (2016) 2189–2196.
[160] X. Zhang, Y. Sun, Z. Wang, Z. Huang, B. Li, J. Fu, Up-regulation of connexin-43 expression in bone marrow mesenchymal stem cells plays a crucial role in adhesion and migration of multiple myeloma cells, Leuk. Lymphoma, 56 (2015) 211–218.
[161] N.K. Haass, D. Ripperger, E. Wladykowski, P. Dawson, P.A. Gimotty, C. Blome, F. Fischer, P. Schmage, I. Moll, J.M. Brandner, Melanoma progression exhibits a significant impact on connexin expression patterns in the epidermal tumor microenvironment, Histochem. Cell Biol., 133 (2010) 113–124.
[162] W.C. Sin, Q. Aftab, J.F. Bechberger, J.H. Leung, H. Chen, C.C. Naus, Astrocytes promote glioma invasion via the gap junction protein connexin43, Oncogene, 35 (2016) 1504–1516.
[163] C. Beck, S. Cayeux, S.D. Lupton, B. Dörken, T. Blankenstein, The thymidine kinase/ganciclovir-mediated “suicide” effect is variable in different tumor cells, Hum. Gene Ther., 6 (1995) 1525–1530.
[164] T. Jimenez, W.P. Fox, C.C.G. Naus, J. Galipeau, D.J. Belliveau, Connexin over-expression differentially suppresses glioma growth and contributes to the bystander effect following HSV-thymidine kinase gene therapy, Cell Commun. Adhes., 13 (2006) 79–92.
[165] S. Matono, T. Tanaka, S. Sueyoshi, H. Yamana, H. Fujita, K. Shirouzu, Bystander effect in suicide gene therapy is directly proportional to the degree of gap junctional intercellular communication in esophageal cancer, Int. J. Oncol., 23 (2003) 1309–1315.
[166] K. Rtibi, D. Grami, S. Selmi, M. Amri, H. Sebai, L. Marzouki, Vinblastine, an anticancer drug, causes constipation and oxidative stress as well as others disruptions in intestinal tract in rat, Toxicol Rep, 4 (2017) 221–225.
[167] P.H.M. De Mulder, C.M.L. van Herpen, P.A.F. Mulders, Current treatment of renal cell carcinoma, Ann. Oncol., 15 Suppl 4 (2004) iv319–28.
[168] H. Sato, H. Senba, N. Virgona, K. Fukumoto, T. Ishida, H. Hagiwara, E. Negishi, K. Ueno, H. Yamasaki, T. Yano, Connexin 32 potentiates vinblastine-induced cytotoxicity in renal cell carcinoma cells, Mol. Carcinog., 46 (2007) 215–224.
[169] H. Sato, K. Fukumoto, S. Hada, H. Hagiwara, E. Fujimoto, E. Negishi, K. Ueno, T. Yano, Enhancing effect of connexin 32 gene on vinorelbine-induced cytotoxicity in A549 lung adenocarcinoma cells, Cancer Chemother. Pharmacol., 60 (2007) 449–457.
[170] M.C. Piccirillo, G. Daniele, M. Di Maio, J. Bryce, G. De Feo, A. Del Giudice, F. Perrone, A. Morabito, Vinorelbine for non-small cell lung cancer, Expert Opin. Drug Saf., 9 (2010) 493–510.
[171] R.P. Huang, Y. Fan, M.Z. Hossain, A. Peng, Z.L. Zeng, A.L. Boynton, Reversion of the neoplastic phenotype of human glioblastoma cells by connexin 43 (cx43), Cancer Res., 58 (1998) 5089–5096.
[172] M. Uzu, H. Sato, R. Yamada, T. Kashiba, Y. Shibata, K. Yamaura, K. Ueno, Effect of enhanced expression of connexin 43 on sunitinib-induced cytotoxicity in mesothelioma cells, J. Pharmacol. Sci., 128 (2015) 17–26.
[173] D.W. Laird, Connexin phosphorylation as a regulatory event linked to gap junction internalization and degradation, Biochim. Biophys. Acta, 1711 (2005) 172–182.
[174] J.L. Solan, P.D. Lampe, Connexin phosphorylation as a regulatory event linked to gap junction channel assembly, Biochim. Biophys. Acta, 1711 (2005) 154–163.
[175] Y. Guo, C. Martinez-Williams, D.E. Rannels, Gap junction-microtubule associations in rat alveolar epithelial cells, Am. J. Physiol. Lung Cell. Mol. Physiol., 285 (2003) L1213–21.
[176] R.G. Johnson, R.A. Meyer, X.-R. Li, D.M. Preus, L. Tan, H. Grunenwald, A.F. Paulson, D.W. Laird, J.D. Sheridan, Gap junctions assemble in the presence of cytoskeletal inhibitors, but enhanced assembly requires microtubules, Exp. Cell Res., 275 (2002) 67–80.
[177] P.E. Martin, G. Blundell, S. Ahmad, R.J. Errington, W.H. Evans, Multiple pathways in the trafficking and assembly of connexin 26, 32 and 43 into gap junction intercellular communication channels, J. Cell Sci., 114 (2001) 3845–3855.
[178] B.N.G. Giepmans, Gap junctions and connexin-interacting proteins, Cardiovasc. Res., 62 (2004) 233–245.
[179] T. Kojima, N. Sawada, H. Chiba, Y. Kokai, M. Yamamoto, M. Urban, G.H. Lee, E.L. Hertzberg, Y. Mochizuki, D.C. Spray, Induction of tight junctions in human connexin 32 (hCx32)-transfected mouse hepatocytes: connexin 32 interacts with occludin, Biochem. Biophys. Res. Commun., 266 (1999) 222–229.
[180] T. Kojima, Y. Kokai, H. Chiba, M. Yamamoto, Y. Mochizuki, N. Sawada, Cx32 but not Cx26 is associated with tight junctions in primary cultures of rat hepatocytes, Exp. Cell Res., 263 (2001) 193–201.
[181] T.A. Martin, W.G. Jiang, Loss of tight junction barrier function and its role in cancer metastasis, Biochim. Biophys. Acta, 1788 (2009) 872–891.
[182] A. Ito, N. Morita, D. Miura, Y.-I. Koma, T.R. Kataoka, H. Yamasaki, Y. Kitamura, Y. Kita, H. Nojima, A derivative of oleamide potently inhibits the spontaneous metastasis of mouse melanoma BL6 cells, Carcinogenesis, 25 (2004) 2015–2022.
[183] D. Miura, Y. Kida, H. Nojima, Camellia oil and its distillate fractions effectively inhibit the spontaneous metastasis of mouse melanoma BL6 cells, FEBS Lett., 581 (2007) 2541–2548.
[184] S. Ferrati, A.K. Gadok, A.D. Brunaugh, C. Zhao, L.A. Heersema, H.D.C. Smyth, J.C. Stachowiak, Connexin membrane materials as potent inhibitors of breast cancer cell migration, J. R. Soc. Interface, 14 (2017).
[185] G. Charras, E. Paluch, Blebs lead the way: how to migrate without lamellipodia, Nat. Rev. Mol. Cell Biol., 9 (2008) 730.
[186] P.E.M. Martin, W.H. Evans, Incorporation of connexins into plasma membranes and gap junctions, Cardiovasc. Res., 62 (2004) 378–387.
[187] A.W. Hunter, R.J. Barker, C. Zhu, R.G. Gourdie, Zonula occludens-1 alters connexin43 gap junction size and organization by influencing channel accretion, Mol. Biol. Cell, 16 (2005) 5686–5698.
[188] J.M. Rhett, J. Jourdan, R.G. Gourdie, Connexin 43 connexon to gap junction transition is regulated by zonula occludens-1, Mol. Biol. Cell, 22 (2011) 1516–1528.
[189] G.S. Ghatnekar, C.L. Grek, D.G. Armstrong, S.C. Desai, R.G. Gourdie, The effect of a connexin43-based Peptide on the healing of chronic venous leg ulcers: a multicenter, randomized trial, J. Invest. Dermatol., 135 (2015) 289–298.
[190] C.L. Grek, J.M. Rhett, J.S. Bruce, M.A. Abt, G.S. Ghatnekar, E.S. Yeh, Targeting connexin 43 with α-connexin carboxyl-terminal (ACT1) peptide enhances the activity of the targeted inhibitors, tamoxifen and lapatinib, in breast cancer: clinical implication for ACT1, BMC Cancer, 15 (2015) 296.
[191] V.C. Jordan, The role of tamoxifen in the treatment and prevention of breast cancer, Curr. Probl. Cancer, 16 (1992) 129–176.
[192] D. Bilancia, G. Rosati, A. Dinota, D. Germano, R. Romano, L. Manzione, Lapatinib in breast cancer, Ann. Oncol., 18 Suppl 6 (2007) vi26–30.
[193] M. Jaraíz-Rodríguez, M.D. Tabernero, M. González-Tablas, A. Otero, A. Orfao, J.M. Medina, A. Tabernero, A Short Region of Connexin43 Reduces Human Glioma Stem Cell Migration, Invasion, and Survival through Src, PTEN, and FAK, Stem Cell Reports, 9 (2017) 451–463.
[194] B.W. Calder, J. Matthew Rhett, H. Bainbridge, S.A. Fann, R.G. Gourdie, M.J. Yost, Inhibition of connexin 43 hemichannel-mediated ATP release attenuates early inflammation during the foreign body response, Tissue Eng. Part A, 21 (2015) 1752–1762.
[195] J.M. Rhett, B.W. Calder, S.A. Fann, H. Bainbridge, R.G. Gourdie, M.J. Yost, Mechanism of action of the anti-inflammatory connexin43 mimetic peptide JM2, Am. J. Physiol. Cell Physiol., 313 (2017) C314–C326.
[196] S.N. Shishido, A. Delahaye, A. Beck, T.A. Nguyen, The anticancer effect of PQ1 in the MMTV-PyVT mouse model, Int. J. Cancer, 134 (2014) 1474–1483.
[197] C.-F. Tsai, Y.-K. Cheng, D.-Y. Lu, S.-L. Wang, C.-N. Chang, P.-C. Chang, W.-L. Yeh, Inhibition of estrogen receptor reduces connexin 43 expression in breast cancers, Toxicol. Appl. Pharmacol., 338 (2018) 182–190.
[198] T.N. Zamay, G.S. Zamay, O.S. Kolovskaya, R.A. Zukov, M.M. Petrova, A. Gargaun, M.V. Berezovski, A.S. Kichkailo, Current and Prospective Protein Biomarkers of Lung Cancer, Cancers , 9 (2017).
[199] N. Okabe, J. Ezaki, T. Yamaura, S. Muto, J. Osugi, H. Tamura, J.-I. Imai, E. Ito, Y. Yanagisawa, R. Honma, M. Gotoh, S. Watanabe, S. Waguri, H. Suzuki, FAM83B is a novel biomarker for diagnosis and prognosis of lung squamous cell carcinoma, Int. J. Oncol., 46 (2015) 999–1006.
[200] M. Cristofanilli, G.T. Budd, M.J. Ellis, A. Stopeck, J. Matera, M.C. Miller, J.M. Reuben, G.V. Doyle, W.J. Allard, L.W.M.M. Terstappen, D.F. Hayes, Circulating tumor cells, disease progression, and survival in metastatic breast cancer, N. Engl. J. Med., 351 (2004) 781–791.
[201] M. Uhlén, L. Fagerberg, B.M. Hallström, C. Lindskog, P. Oksvold, A. Mardinoglu, Å. Sivertsson, C. Kampf, E. Sjöstedt, A. Asplund, I. Olsson, K. Edlund, E. Lundberg, S. Navani, C.A.-K. Szigyarto, J. Odeberg, D. Djureinovic, J.O. Takanen, S. Hober, T. Alm, et al., Proteomics Tissue-based map of the human proteome, Science, 347 (2015) 1260419.
[202] D.J. Kim, W.J. Kim, M. Lim, Y. Hong, S.-J. Lee, S.-H. Hong, J. Heo, H.-Y. Lee, S.-S. Han, Plasma CRABP2 as a Novel Biomarker in Patients with Non-Small Cell Lung Cancer, J. Korean Med. Sci., 33 (2018) e178.
[203] I. López de Silanes, M. Zhan, A. Lal, X. Yang, M. Gorospe, Identification of a target RNA motif for RNA-binding protein HuR, Proc. Natl. Acad. Sci. U. S. A., 101 (2004) 2987–2992.
[204] W. Zhang, A.C. Vreeland, N. Noy, RNA-binding protein HuR regulates nuclear import of protein, J. Cell Sci., 129 (2016) 4025–4033.
[205] F. Milletti, Cell-penetrating peptides: classes, origin, and current landscape, Drug Discov. Today, 17 (2012) 850–860.
[206] G. Hardavella, R. George, T. Sethi, Lung cancer stem cells-characteristics, phenotype, Transl Lung Cancer Res, 5 (2016) 272–279.
指導教授 王陸海 陳盛良(Lu-Hai Wang Shen-Liang Chen) 審核日期 2019-3-29
推文 facebook   plurk   twitter   funp   google   live   udn   HD   myshare   reddit   netvibes   friend   youpush   delicious   baidu   
網路書籤 Google bookmarks   del.icio.us   hemidemi   myshare   

若有論文相關問題,請聯絡國立中央大學圖書館推廣服務組 TEL:(03)422-7151轉57407,或E-mail聯絡  - 隱私權政策聲明