リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Bioinformatics and Functional Analyses Implicate Potential Roles for EOGT and L-fringe in Pancreatic Cancers」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Bioinformatics and Functional Analyses Implicate Potential Roles for EOGT and L-fringe in Pancreatic Cancers

Rashu, Barua 名古屋大学

2022.12.22

概要

【Abstract】
Notch signaling receptors, ligands, and their downstream target genes are dysregulated in pancreatic ductal adenocarcinoma (PDAC), suggesting a role of Notch signaling in pancreatic tumor development and progression. However, dysregulation of Notch signaling by posttranslational modification of Notch receptors remains poorly understood. Here, we analyzed the Notch-modifying glycosyltransferase involved in the regulation of the ligand-dependent Notch signaling pathway. Bioinformatic analysis revealed that the expression of epidermal growth factor (EGF) domain-specific O-linked N-acetylglucosamine (EOGT) and Lunatic fringe (LFNG) positively correlates with a subset of Notch signaling genes in PDAC. The lack of EOGT or LFNG expression inhibited the proliferation and migration of Panc-1 cells, as observed by the inhibition of Notch activation. EOGT expression is significantly increased in the basal subtype, and low expression of both EOGT and LFNG predicts better overall survival in PDAC patients. These results imply potential roles for EOGT- and LFNG-dependent Notch signaling in PDAC.

【Methods and Materials】
EOGT and LFNG expression in pancreatic ductal adenocarcinoma (PDAC) were analyzed by using GEPIA2 database. The correlation between glycosyltransferase genes and Notch target genes was determined based on TCGA datasets. Endogenous expression levels of EOGT in human PDAC cell lines were compared to a normal pancreatic ductal cell line (H6C7) by immunoblotting. Further, EOGT expression was verified by Immunostaining. Cell growth inhibition by DAPT in Panc-1 cell line was determined by IncuCyte ZOOM Live-Cell Analysis System. CRISPR/Cas9- mediated lentiviral gene editing technique was used to knockout EOGT in Panc-1 cell line. Evaluation of CRISPR/Cas9-mediated genome editing was confirmed by T7 endonuclease assay and immunoblotting. Cell proliferation assay was performed in both wild type Panc-1 and EOGT lack Panc-1 cell line using IncuCyte ZOOM Live-Cell Analysis System. Cell migration (Wound healing) was analyzed by using IncuCyte ZOOM Live-Cell Analysis system in in both wild type Panc-1 and EOGT knockout Panc-1 cell lines. Knockout of LFNG was performed by CRISPR/Cas9-mediated lentivirus transduction. Genome editing was confirmed by T7 endonuclease assay and sequencing. Cell proliferation in Panc-1 cells lacking LFNG were analyzed by IncuCyte Zoom system. Wound healing migration assay to compare the motility of wild-type and LFNG-Ko Panc-1 cells was analyzed by IncuCyte Zoom system. Kaplan-Meier survival curve for overall survival in pancreatic ductal adenocarcinoma patients was analyzed by EZR software (version 1.52), which is a graphical user interface for R software (version 4.0.2).

【Results】
Among Notch target genes, HES1 and HEY1 expressions were higher in both basal and classical subtypes of PDAC. In contrast, the expression of EOGT expression is significantly increased in basal subtype of PDAC, which represent a more aggressive phenotype. LFNG expression was not significantly altered (Figure 1A). However, these data did not exclude the possibility of multiple, rather than a single, glycosyltransferases contributing to dysregulated Notch signaling in PDAC. Correlation analysis between glycosyltransferase and upregulated Notch target genes revealed a significant positive correlation between EOGT and HEY1 (r = 0.47, p = 4.2x 10-11). In contrast, a positive correlation between LFNG and HES1 (r = 0.48, p = 8.4 × 10−12) was found (Figure 1B).

We found that four PDAC cell lines (Panc-1, BxPC3, Panc03.27, and CAPAN-2) out of ten showed higher EOGT expression compared to H6C7 cells (Figure 2A). We also verified the expression of EOGT by Immunostaining in the four PDAC cell lines (Figure 2B). Based on the immunoblotting and Immunostaining assays, we selected Panc-1, which showed prominent EOGT expression for functional analysis.

We performed gene editing with the CRISPR/Cas9-mediated lentivirus technique in Panc-1 cell. After lentiviral transduction and Blasticidin S selection, the lack of EOGT was confirmed by the anti-EOGT (AER61) antibody (Figure 3A & 3B). To investigate whether the decreased Notch activity inhibits Panc-1 cell proliferation, Panc1 cells were treated with DAPT, a γ-secretase inhibitor that blocks Notch signaling. The inhibitory effect of DAPT on the proliferation of Panc-1 cells was observed in a timedependent manner (Figure 4). This result suggested that decreased Notch activity can substantially reduce the cell proliferation of Panc-1.
To examine the effect of knockout of EOGT in the Panc-1 cells, we observed the proliferation between wild-type and EOGT-KO cells at an interval of 6h over 66h. EOGT-KO cells exhibited slow growth relative to wild-type parental control cells. This result showed that EOGT promotes cell proliferation in Panc-1 cells (Figure 5A).

Analysis of the relative wound healing density revealed that EOGT-KO Panc-1 showed significant decreases in re-capturing the wound portion compared to wild-type control cells (Figure 5B). Compared to the control cells, EOGT-KO clones showed a slower migration rate, presenting a depletion of wound density highest 20%, within 18h. The differences in the migration rate among the KO clones appear to be due to the cancer cells’ clonal variation.

To compare the effect of EOGT and LFNG in the growth and motility of Panc-1 cells, we generated LFNG-KO cells by CRISPR/Cas9-mediated lentivirus transduction. The cutting efficacy was analyzed by T7 endonuclease assay. Then, by limiting dilution, single-cell clones were isolated. GFP-positive clones were selected for sequencing to confirm LFNG knockout in Panc-1 cells (Figure 6).

To evaluate the roles of LFNG in cell proliferation, Panc-1 cells lacking LFNG were analyzed. The result showed that the cell proliferation of LFNG-KO cells was lower than that of parental control cells (Figure 7A). These data suggested that LFNG promotes cell proliferation of Panc-1 cells. (Figure 7B).

Kaplan-Meier survival curves revealed no association between EOGT expression and overall survival in PDAC (Figure 8A). Additionally, no association was observed between LFNG expression and overall survival (Figure 8B). In contrast, lower of both EOGT and LFNG is associated with a better prognosis of PDAC (p= 0.0195) (Figure 8C).

【Discussion】
Bioinformatic analysis data suggested that the contribution of EOGT and LFNG to Notch signaling is qualitatively different, possibly through the differential impact on multiple Notchligand pairs, which leads to the transactivation of distinct sets of Notch signaling target genes, including HES1 and HEY1. Further in-depth bioinformatics analyses included Biclustering methods, will help elucidate the pathological relevance of the observed correlations in tumor progression.

We showed the roles of EOGT in cell proliferation and migration assay in cancer cells. In our study, loss of EOGT or LFNG resulted in a similar cellular phenotype, suggesting that both genes cooperate in the tumor properties of PDAC cells. Furthermore, the combination of lower LFNG and EOGT expression in PDAC serves as an excellent prognostic marker, implying that both Notch modifiers modulates disease progression. Given that EOGT and LFNG modulate ligand-induced Notch signaling, these results suggested that changes in the expression of these glycosyltransferases result in altered Notch activity, as indicated by the correlation between LFNG and HES1, or EOGT and HEY1 expression in the PDAC database. Therefore, dysregulated Notch signaling mediated by changes in the expression of EOGT and LFNG would affect the prognosis of PDAC. Although this study implicates potential roles for EOGT- and LFNGdependent Notch signaling in PDAC, further studies will be necessary to clarify the roles of Notch-modifying enzymes in dysregulated Notch activity and the development and progression of PDAC at the molecular level.

【Conclusion】
EOGT- and LFNG- dependent Notch signaling plays critical roles in PDAC. Both EOGT and LFNG impact cell proliferation and migration in cancer cells. The dysregulated Notch signaling due to higher expression of EOGT or LFNG leads to poor prognosis.

この論文で使われている画像

参考文献

1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [CrossRef] [PubMed]

2. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [CrossRef] [PubMed]

3. Vincent, A.; Herman, J.; Schulick, R.; Hruban, R.H.; Goggins, M. Pancreatic cancer. Lancet 2011, 378, 607–620. [CrossRef]

4. Kleeff, J.; Korc, M.; Apte, M.; La Vecchia, C.; Johnson, C.D.; Biankin, A.V.; E Neale, R.; Tempero, M.; Tuveson, D.A.; Hruban, R.H.; et al. Pancreatic cancer. Nat. Rev. Dis. Prim. 2016, 2, 16022. [CrossRef]

5. Bliss, L.A.; Witkowski, E.R.; Yang, C.J.; Tseng, J.F. Outcomes in operative management of pancreatic cancer. J. Surg. Oncol. 2014, 110, 592–598. [CrossRef]

6. Ntziachristos, P.; Lim, J.S.; Sage, J.; Aifantis, I. From Fly Wings to Targeted Cancer Therapies: A Centennial for Notch Signaling. Cancer Cell 2014, 25, 318–334. [CrossRef]

7. Tashima, Y.; Okajima, T. Congenital diseases caused by defective O-glycosylation of Notch receptors. Nagoya J. Med Sci. 2018, 80, 299–307.

8. Espinoza, I.; Miele, L. Notch inhibitors for cancer treatment. Pharmacol. Ther. 2013, 139, 95–110. [CrossRef]

9. Miele, L.; Espinoza, I.; Pochampally, R.; Watabe, K.; Xing, F. Notch signaling: Targeting cancer stem cells and epithelial-tomesenchymal transition. OncoTargets Ther. 2013, 6, 1249–1259. [CrossRef] [PubMed]

10. Avila, J.L.; Kissil, J.L. Notch signaling in pancreatic cancer: Oncogene or tumor suppressor? Trends Mol. Med. 2013, 19, 320–327. [CrossRef]

11. Nakhai, H.; Siveke, J.T.; Klein, B.; Mendoza-Torres, L.; Mazur, P.K.; Algül, H.; Radtke, F.; Strobl, L.; Zimber-Strobl, U.; Schmidt, G. Conditional ablation of Notch signaling in pancreatic development. Development 2008, 135, 2757–2765. [CrossRef]

12. Hanlon, L.; Avila, J.L.; Demarest, R.M.; Troutman, S.; Allen, M.; Ratti, F.; Rustgi, A.K.; Stanger, B.Z.; Radtke, F.; Adsay, V.; et al. Notch1 Functions as a Tumor Suppressor in a Model of K-ras–Induced Pancreatic Ductal Adenocarcinoma. Cancer Res. 2010, 70, 4280–4286. [CrossRef] [PubMed]

13. Agrawal, N.; Frederick, M.J.; Pickering, C.R.; Bettegowda, C.; Chang, K.; Li, R.J.; Fakhry, C.; Xie, T.-X.; Zhang, J.; Wang, J.; et al. Exome Sequencing of Head and Neck Squamous Cell Carcinoma Reveals Inactivating Mutations in NOTCH1. Science 2011, 333, 1154–1157. [CrossRef]

14. Kopan, R.; Ilagan, M.X.G. The Canonical Notch Signaling Pathway: Unfolding the Activation Mechanism. Cell 2009, 137, 216–233. [CrossRef]

15. Plentz, R.; Park, J.; Rhim, A.D.; Abravanel, D.; Hezel, A.F.; Sharma, S.V.; Gurumurthy, S.; Deshpande, V.; Kenific, C.; Settleman, J.; et al. Inhibition of γ-Secretase Activity Inhibits Tumor Progression in a Mouse Model of Pancreatic Ductal Adenocarcinoma. Gastroenterology 2009, 136, 1741–1749.e6. [CrossRef] [PubMed]

16. Miyamoto, Y.; Maitra, A.; Ghosh, B.; Zechner, U.; Argani, P.; A Iacobuzio-Donahue, C.; Sriuranpong, V.; Iso, T.; Meszoely, I.M.; Wolfe, M.S.; et al. Notch mediates TGFα-induced changes in epithelial differentiation during pancreatic tumorigenesis. Cancer Cell 2003, 3, 565–576. [CrossRef]

17. Ye, J.; Wen, J.; Ning, Y.; Li, Y. Higher notch expression implies poor survival in pancreatic ductal adenocarcinoma: A systematic review and meta-analysis. Pancreatology 2018, 18, 954–961. [CrossRef]

18. Thomas, M.M.; Zhang, Y.; Mathew, E.; Kane, K.T.; Maillard, I.; Di Magliano, M.P. Epithelial Notch signaling is a limiting step for pancreatic carcinogenesis. BMC Cancer 2014, 14, 862. [CrossRef] [PubMed]

19. Song, H.; Wang, Y.; Lan, H.; Zhang, Y. Expression of Notch receptors and their ligands in pancreatic ductal adenocarcinoma. Exp. Ther. Med. 2018, 16, 53–60. [CrossRef] [PubMed]

20. Ma, J.; Xia, J.; Miele, L.; Sarkar, F.H.; Wang, Z. Notch Signaling Pathway in Pancreatic Cancer Progression. Pancreat. Disord. Ther. 2013, 3, 3. [CrossRef]

21. Matsuura, A.; Ito, M.; Sakaidani, Y.; Kondo, T.; Murakami, K.; Furukawa, K.; Nadano, D.; Matsuda, T.; Okajima, T. O-LinkedNAcetylglucosamine Is Present on the Extracellular Domain of Notch Receptors. J. Biol. Chem. 2008, 283, 35486–35495. [CrossRef]

22. Tashima, Y.; Stanley, P. Antibodies That Detect O-Linked β-d-N-Acetylglucosamine on the Extracellular Domain of Cell Surface Glycoproteins. J. Biol. Chem. 2014, 289, 11132–11142. [CrossRef] [PubMed]

23. Cohen, I.; Silberstein, E.; Perez, Y.; Landau, D.; Elbedour, K.; Langer, Y.; Kadir, R.; Volodarsky, M.; Sivan, S.; Narkis, G.; et al. Autosomal recessive Adams–Oliver syndrome caused by homozygous mutation in EOGT, encoding an EGF domain-specific O-GlcNAc transferase. Eur. J. Hum. Genet. 2013, 22, 374–378. [CrossRef]

24. Ogawa, M.; Okajima, T. Structure and function of extracellular O-GlcNAc. Curr. Opin. Struct. Biol. 2019, 56, 72–77. [CrossRef]

25. Hassed, S.; Li, S.; Mulvihill, J.; Aston, C.; Palmer, S. Adams-Oliver syndrome review of the literature: Refining the diagnostic phenotype. Am. J. Med Genet. Part A 2017, 173, 790–800. [CrossRef]

26. Sawaguchi, S.; Varshney, S.; Ogawa, M.; Sakaidani, Y.; Yagi, H.; Takeshita, K.; Murohara, T.; Kato, K.; Sundaram, S.; Stanley, P.; et al. O-GlcNAc on NOTCH1 EGF repeats regulates ligand-induced Notch signaling and vascular development in mammals. eLife 2017, 6, 7280. [CrossRef] [PubMed]

27. Dunwoodie, S.L. Mutation of the fucose-specific β1,3 N-acetylglucosaminyltransferase LFNG results in abnormal formation of the spine. Biochim. Biophys. Acta BBA Mol. Basis Dis. 2009, 1792, 100–111. [CrossRef] [PubMed]

28. Kakuda, S.; Lopilato, R.K.; Ito, A.; Haltiwanger, R.S. Canonical Notch ligands and Fringes have distinct effects on NOTCH1 and NOTCH2. J. Biol. Chem. 2020, 295, 14710–14722. [CrossRef]

29. Zhang, S.; Chung, W.-C.; Xu, K. Lunatic Fringe is a potent tumor suppressor in Kras-initiated pancreatic cancer. Oncogene 2016, 35, 2485–2495. [CrossRef] [PubMed]

30. Ogawa, M.; Senoo, Y.; Ikeda, K.; Takeuchi, H.; Okajima, T. Structural Divergence in O-GlcNAc Glycans Displayed on Epidermal Growth Factor-like Repeats of Mammalian Notch1. Molecules 2018, 23, 1745. [CrossRef]

31. Varshney, S.; Stanley, P. Multiple roles for O-glycans in Notch signalling. FEBS Lett. 2018, 592, 3819–3834. [CrossRef]

32. Kakuda, S.; Haltiwanger, R.S. Deciphering the Fringe-Mediated Notch Code: Identification of Activating and Inhibiting Sites Allowing Discrimination between Ligands. Dev. Cell 2017, 40, 193–201. [CrossRef] [PubMed]

33. Haltom, A.R.; Jafar-Nejad, H. The multiple roles of epidermal growth factor repeatO-glycans in animal development. Glycobiology 2015, 25, 1027–1042. [CrossRef] [PubMed]

34. Juiz, N.; Elkaoutari, A.; Bigonnet, M.; Gayet, O.; Roques, J.; Nicolle, R.; Iovanna, J.L.; Dusetti, N. Basal-like and classical cells coexist in pancreatic cancer revealed by single-cell analysis on biopsy-derived pancreatic cancer organoids from the classical subtype. FASEB J. 2020, 34, 12214–12228. [CrossRef] [PubMed]

35. Nandagopal, N.; Santat, L.A.; Lebon, L.; Sprinzak, D.; Bronner, M.E.; Elowitz, M.B. Dynamic Ligand Discrimination in the Notch Signaling Pathway. Cell 2018, 172, 869–880.e19. [CrossRef]

36. Lopez-Fernandez, A.; Rodriguez-Baena, D.; Gomez-Vela, F.; Divina, F.; Garcia-Torres, M. A multi-GPU biclustering algorithm for binary datasets. J. Parallel Distrib. Comput. 2021, 147, 209–219. [CrossRef]

37. Orzechowski, P.; Sipper, M.; Huang, X.; Moore, J.H. EBIC: An evolutionary-based parallel biclustering algorithm for pattern discovery. Bioinformatics 2018, 34, 3719–3726. [CrossRef]

38. Xie, J.; Ma, A.; Zhang, Y.; Liu, B.; Cao, S.; Wang, C.; Xu, J.; Zhang, C.; Ma, Q. QUBIC2: A novel and robust biclustering algorithm for analyses and interpretation of large-scale RNA-Seq data. Bioinformatics 2020, 36, 1143–1149. [CrossRef]

39. Bhattacharya, A.; Cui, Y. A GPU-accelerated algorithm for biclustering analysis and detection of condition-dependent coexpression network modules. Sci. Rep. 2017, 7, 1–9. [CrossRef]

40. Du, X.; Cheng, Z.; Li, Y.; Zhou, Z.-G.; Yang, L.; Zhang, M.-M. Suppressive effects of gamma-secretase inhibitor DAPT on the proliferation of pancreatic cancer cells. Sichuan da xue xue bao. Yi xue ban J. Sichuan Univ. Med Sci. Ed. 2013, 44, 699–702.

41. Harbuzariu, A.; Rampoldi, A.; Daley-Brown, D.S.; Candelaria, P.; Harmon, T.L.; Lipsey, C.C.; Beech, D.J.; Quarshie, A.; Ilies, G.O.; Gonzalez, R.R. Leptin-Notch signaling axis is involved in pancreatic cancer progression. Oncotarget 2016, 8, 7740–7752. [CrossRef]

42. Hu, Y.; Su, H.; Ling, C.; Guoliang, Q.; Cheng, L.; Qin, R.; Qing, G.; Liu, H. The NOTCH Ligand JAGGED2 Promotes Pancreatic Cancer Metastasis Independent of NOTCH Signaling Activation. Mol. Cancer Ther. 2015, 14, 289–297. [CrossRef]

43. Chen, D.; Wang, H.; Bao, Y.; Xie, K. Notch signaling molecule is involved in the invasion of MiaPaCa2 cells induced by CoCl2 via regulating epithelial-mesenchymal transition. Mol. Med. Rep. 2018, 17, 4965–4972. [CrossRef] [PubMed]

44. Lieber, M.; Mazzetta, J.; Nelson-Rees, W.; Kaplan, M.; Todaro, G. Establishment of a continuous tumor-cell line (PANC-1) from a human carcinoma of the exocrine pancreas. Int. J. Cancer 1975, 15, 741–747. [CrossRef]

45. Watanabe, M. Metabolic Profiling Comparison of Human Pancreatic Ductal Epithelial Cells and Three Pancreatic Cancer Cell Lines using NMR Based Metabonomics. J. Mol. Biomarkers Diagn. 2012, 3. [CrossRef]

46. Single, A.; Beetham, H.; Telford, B.J.; Guilford, P.; Chen, A. A Comparison of Real-Time and Endpoint Cell Viability Assays for Improved Synthetic Lethal Drug Validation. J. Biomol. Screen. 2015, 20, 1286–1293. [CrossRef] [PubMed]

47. Naito, Y.; Hino, K.; Bono, H.; Ui-Tei, K. CRISPRdirect: Software for designing CRISPR/Cas guide RNA with reduced off-target sites. Bioinformatics 2015, 31, 1120–1123. [CrossRef]

48. Wang, T.; Wei, J.J.; Sabatini, D.M.; Lander, E.S. Genetic Screens in Human Cells Using the CRISPR-Cas9 System. Science 2014, 343, 80–84. [CrossRef] [PubMed]

49. Narimatsu, Y.; Joshi, H.J.; Yang, Z.; Gomes, C.; Chen, Y.-H.; Lorenzetti, F.C.; Furukawa, S.; Schjoldager, K.T.; Hansen, L.; Clausen, H.; et al. A validated gRNA library for CRISPR/Cas9 targeting of the human glycosyltransferase genome. Glycobiology 2018, 28, 295–305. [CrossRef]

50. Giuliano, C.J.; Lin, A.; Girish, V.; Sheltzer, J.M. Generating Single Cell–Derived Knockout Clones in Mammalian Cells with CRISPR/Cas9. Curr. Protoc. Mol. Biol. 2019, 128, e100. [CrossRef]

51. Abrahimi, P.; Chang, W.G.; Kluger, M.S.; Qyang, Y.; Tellides, G.; Saltzman, W.M.; Pober, J.S. Efficient Gene Disruption in Cultured Primary Human Endothelial Cells by CRISPR/Cas9. Circ. Res. 2015, 117, 121–128. [CrossRef] [PubMed]

52. Alam, S.M.D.; Tsukamoto, Y.; Ogawa, M.; Senoo, Y.; Ikeda, K.; Tashima, Y.; Takeuchi, H.; Okajima, T. N-Glycans on EGF domain-specific O-GlcNAc transferase (EOGT) facilitate EOGT maturation and peripheral endoplasmic reticulum localization. J. Biol. Chem. 2020, 295, 8560–8574. [CrossRef] [PubMed]

53. Libério, M.S.; Sadowski, M.C.; Soekmadji, C.; Davis, R.A.; Nelson, C.C. Differential Effects of Tissue Culture Coating Substrates on Prostate Cancer Cell Adherence, Morphology and Behavior. PLOS ONE 2014, 9, e112122. [CrossRef] [PubMed]

54. Tang, Z.; Kang, B.; Li, C.; Chen, T.; Zhang, Z. GEPIA2: An enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res. 2019, 47, 556–560. [CrossRef] [PubMed]

参考文献をもっと見る