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 「Genome-scale CRISPR/Cas9 screening for gemcitabine modulators in pancreatic cancer」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

Genome-scale CRISPR/Cas9 screening for gemcitabine modulators in pancreatic cancer

Mohammad, Masoudi 東京大学 DOI:10.15083/0002004557

2022.06.22

概要

Pancreatic cancer ranks eleventh among cancers in terms of the diagnosis all over the world, however concerning the cancer-related death it grades seventh. This statistics implies high mortality rate and poor prognosis of the pancreatic cancer. In point of fact, five year overall survival rate of pancreatic cancer is less than 5%, more than 90% of the patients diagnosed with this cancer die and median overall survival for the patients is 8 to 12 months for locally advanced disease.

Currently chemotherapy is the first-line standard treatment for pancreatic cancer and gemcitabine plays a central role in this treatment. Gemcitabine is an analog of deoxycytidine which carries two flour atoms (2’,2’–difluoro-2’-deoxycytidine, dFdC). Upon entrance to the cells via human nucleoside transporters, gemcitabine becomes phosphorylated by deoxycytidine kinase, pyrimidine nucleoside monophosphate kinase and nucleoside diphosphate kinase to form gemcitabine mono-, di- and triphosphate, respectively. Consequently, gemcitabine triphosphate is incorporated into the DNA chains by DNA polymerases and causes a masked chain termination of DNA replication, where DNA replication is halted following incorporation of one base after gemcitabine. Also, gemcitabine diphosphate affects DNA synthesis by reducing dNTP pool of the cells via covalent binding to the active site of ribonucleotide reductase and thereby its inactivation.

Although gemcitabine has been used as the standard first-line treatment for pancreatic cancer its efficacy is still modest (5.6 months overall survival) which warrants exploration to discover candidates for combinatorial therapy along with gemcitabine. In the search for potential gemcitabine sensitizers, a wide range of studies including large-scale screenings have been performed. Large-scale screenings to discover gemcitabine cytotoxity regulators in pancreatic cancer cell lines have been performed utilizing RNA interference (RNAi) libraries. While RNAi screening provides a very powerful tool for detecting the genes involved in our process of interest, lack of the full ablation of the gene function may decrease sensitivity of RNAi approach for large-scale screenings. Since RNAi interferes with the function of a gene at the level of RNA a complete removal of the gene function is not achievable, as remnant of the gene product after RNAi application may be sufficient for performing the gene function. This flaw can be coped by the application of the techniques like CRISPR/Cas9 that provide gene knockout and in turn full cessation of the gene function. In fact, CRISPR/Cas9 system has shown to produce more positive hits, 2 to 5 folds, compared to shRNA system, in loss-of-function screening.

In this study, a CRISPR/Cas9 library (GeCKO-v2) consisting of 123,411 unique sgRNAs targeting 19,050 human genes, was utilized to identify modulators of gemcitabine action in Panc1 pancreatic carcinoma cell line. In short, Lentiviral library was prepared using GeCKO-v2 plasmid library, cultured Panc1 cells were transfected with prepared lentiviral library, infected cells were selected utilizing puromycin, selected cells were split to three parts, one part was used as the baseline control and two other parts were treated either with gemcitabine or its vehicle. Following the gemcitabine screening, the cells were collected and genomic DNA was extracted, next generation sequencing libraries were prepared and subjected to massively parallel sequencing. Raw sequencing data were analyzed and copy numbers of the sgRNAs were acquired.

Efficacy of the genome-scale CRISPR/Cas9 knockout experiment was assessed by GSEA employing Gene Ontology All gene sets from Molecular Signature Database (MSigDB). The result showed that the essential gene sets including Multi Organism Metabolic Process, Ribosomal Subunit and Translational Initiation were depleted in the knockout cells library after 22 days. Also, to obtain essential gene sets for Panc1 cells survival, GSEA employing Hallmark gene sets was performed and MYC-targets, DNA-repair, G2M-checkpoint and E2F-targets gene sets appeared as essential.

Inventories of the genes whose sgRNAs were depleted or enriched in the presence of gemcitabine were obtained employing RIGER algorithm, which ranks the gene based on the differential effects of sgRNAs targeting a gene. Genes involved in gemcitabine metabolism can be classified to two categories. Positive gemcitabine modulators, genes that act in accordance with gemcitabine action and negative gemcitabine modulators, genes that act apposed to gemcitabine action and reduce its effectiveness. As an in-silico validation of the gemcitabine screening, GSEA was performed utilizing the lists of gemcitabine positive and negative modulators. As expected, the positive modulators
were enriched in gemcitabine-treated cells, while negative modulators did not show any enrichment.

As a result of the screening, SH3D21 appeared as the top gemcitabine sensitizer and sensitizing activity of SH3D21 was validated in knockout and knockdown experiments. Panc1 cells carrying an sgRNA targeting SH3D21 showed more sensitivity to gemcitabine as compared to the control sgRNAs carrying cells. The EC50 of gemcitabine was reduced to 41.1 nM in SH3D21-knockout cells compared to that of the control cells (56.8 nM). Silencing SH3D21 gene with an siRNA targeting SH3D21 mRNA resulted in increased sensitivity of Panc1 cells to gemcitabine as well.

PANTHER analysis revealed that the proportion of the proteins with transporter activity was decreased among top 1000 depleted genes compared to those of the entire library. SLC28A3 gene, which codes for one of the known transporters of gemcitabine, was absent among top 1000 depleted genes and present among top 1000 enriched genes while three other known gemcitabine transporters did not show this pattern. The decrease in the proportion of the transporters in top 1000 depleted genes was not limited to the known gemcitabine transporters. On the other hand, the genes involved in endocytosis, like KIAA0196, were among the top enriched genes in gemcitabine-treated cells and one of the three viral proteins that were among top 1000 enriched genes was ERVFRD-1 that is involved in endosomal transport. These data suggested the possibility of involvement of other transporting molecules/mechanisms (e.g. endocytosis) in gemcitabine uptake by Panc1 cells. To verify this hypothesis, an endocytosis inhibitor, chlorpromazine, was used in cell viability assay in the presence of gemcitabine. The result showed that chlorpromazine decreased the efficacy of gemcitabine, confirming that endocytosis has a part in gemcitabine cellular uptake.

In brief, we found SH3D21 as a novel gemcitabine sensitizer that may act as a therapeutic target to improve gemcitabine efficacy. Also, MYC pathway appeared as an essential pathway for the survival of Panc1 cells in our study and might serve as a target for treatment of pancreatic cancer. Furthermore, we found that endocytosis is involved in gemcitabine cellular uptake, a finding that may help planning new strategies for gemcitabine delivery.

参考文献

1. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86.

2. Güngör C, Hofmann BT, Wolters-Eisfeld G, Bockhorn M. Pancreatic cancer. Br J Pharmacol. 2014;171:849–58.

3. Ryan DP, Hong TS, Bardeesy N. Pancreatic Adenocarcinoma. N Engl J Med. 2014;35:353–4.

4. Lee HS, Park SW. Systemic chemotherapy in advanced pancreatic cancer. Gut Liver. 2016. page 340–7.

5. Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817–25.

6. Huang P, Chubb S, Hertel LW, Grindey GB, Plunkett W. Action of 2 , 2 - difluorodeoxycytidine on DNA synthesis. Cancer Res. 1991;51:6110.

7. Heinemann V, Xu YZ, Chubb S, Sen a, Hertel LW, Grindey GB, et al. Inhibition of ribonucleotide reduction in CCRF-CEM cells by 2’,2’-difluorodeoxycytidine. Mol Pharmacol. 1990;38:567–72.

8. Alvarellosa ML, Lamba J, Sangkuhla K, Thorna CF, Wange L, Klein DJ, et al. PharmGKB summary: gemcitabine pathway. Pharmacogenet Genomics. 2012;24:1.

9. Mackey JR, Mani RS, Selner M, Mowles D, Young JD, Belt JA, et al. Functional nucleoside transporters are required for gemcitabine influx and manifestation of toxicity in cancer cell lines. Cancer Res. 1998;58:4349–57.

10. Ritzel MWL, Ng AML, Yao SYM, Graham K, Loewen SK, Smith KM, et al. Molecular Identification and Characterization of Novel Human and Mouse Concentrative Na+-Nucleoside Cotransporter Proteins (hCNT3 and mCNT3) Broadly Selective for Purine and Pyrimidine Nucleosides (System cib). J Biol Chem. 2001;276:2914–27.

11. Heinemann V, Hertel L, Grindey G, Plunkett W. Comparison of the cellular pharmacokinetics and toxicity of 2′, 2′-difluorodeoxycytidine and 1-β-d- arabinofuranosylcytosine. Cancer Res. 1988;4024–31.

12. Bouffard DY, Laliberté J, Momparler RL. Kinetic studies on 2’,2’- difluorodeoxycytidine (gemcitabine) with purified human deoxycytidine kinase and cytidine deaminase. Biochem Pharmacol. 1993;45:1857–61.

13. Hatzis P, Al-Madhoon a S, Jüllig M, Petrakis TG, Eriksson S, Talianidis I. The intracellular localization of deoxycytidine kinase. J Biol Chem. 1998;273:30239– 43.

14. Van Rompay a R, Johansson M, Karlsson a. Phosphorylation of deoxycytidine analog monophosphates by UMP-CMP kinase: molecular characterization of the human enzyme. Mol Pharmacol. 1999;56:562–9.

15. Wong A, Soo RA, Yong WP, Innocenti F. Clinical pharmacology and pharmacogenetics of gemcitabine. Drug Metab. Rev. 2009. page 77–88.

16. Bepler G, Kusmartseva I, Sharma S, Gautam A, Cantor A, Sharma A, et al. RRM1 Modulated In Vitro and In Vivo Efficacy of Gemcitabine and Platinum in Non–Small-Cell Lung Cancer. J Clin Oncol. 2006;24:4731–7.

17. Wang C, Zhang W, Fu M, Yang A, Huang H, Xie J. Establishment of human pancreatic cancer gemcitabine‑resistant cell line with ribonucleotide reductase overexpression. Oncol Rep. 2015;33:383.

18. Duxbury MS, Ito H, Zinner MJ, Ashley SW, Whang EE. RNA interference targeting the M2 subunit of ribonucleotide reductase enhances pancreatic adenocarcinoma chemosensitivity to gemcitabine. Oncogene. 2004;23:1539–48.

19. Heinemann V, Xu YZ, Chubb S, Sen A, Plunkett W, Hertel LW, et al. Cellular Elimination of 2′,2′-Difluorodeoxycytidine 5′-Triphosphate: A Mechanism of Self-Potentiation. Cancer Res. 1992;52:533–9.

20. Plunkett W, Huang P, Gandhi V. Preclinical characteristics of gemcitabine. Anticancer Drugs. 1995;6 Suppl 6:7–13.

21. Fryer RA, Barlett B, Galustian C, Dalgleish AG. Mechanisms underlying gemcitabine resistance in pancreatic cancer and sensitisation by the iMiDTM lenalidomide. Anticancer Res. 2011;31:3747–56.

22. Li J, Wang R, Schweickert PG, Karki A, Yang Y, Kong Y, et al. Plk1 inhibition enhances the efficacy of gemcitabine in human pancreatic cancer. Cell Cycle. 2016;15:711–9.

23. Bhattacharjee V, Zhou Y, Yen TJ. A synthetic lethal screen identifies the vitamin D receptor as a novel gemcitabine sensitizer in pancreatic cancer cells. Cell Cycle. 2014;13:3839–56.

24. Azorsa DO, Gonzales IM, Basu GD, Choudhary A, Arora S, Bisanz KM, et al. Synthetic lethal RNAi screening identifies sensitizing targets for gemcitabine therapy in pancreatic cancer. J Transl Med. 2009;7:43.

25. Fredebohm J, Wolf J, Hoheisel JD, Boettcher M. Depletion of RAD17 sensitizes pancreatic cancer cells to gemcitabine. J Cell Sci. 2013;126:3380–9.

26. Hartenian E, Doench JG. Genetic screens and functional genomics using CRISPR/Cas9 technology. FEBS J. 2015;282:1383–93.

27. Ran FA, Hsu PPD, Wright J, Agarwala V, Scott D a, Zhang F. Genome engineering using the CRISPR-Cas9 system. Nat Protoc. 2013;8:2281–308.

28. Makarova KS, Haft DH, Barrangou R, Brouns SJJ, Charpentier E, Horvath P, et al. Evolution and Classification of the CRISPR–Cas Systems. Nat Rev Microbiol. 2011;9:467–77.

29. Sander JD, Joung JK. CRISPR-Cas systems for editing, regulating and targeting genomes. Nat Biotechnol. Nature Publishing Group; 2014;32:347–55.

30. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A Programmable Dual-RNA – Guided. 2012;337:816–22.

31. Keskin H, Shen Y, Huang F, Patel M, Yang T, Ashley K, et al. Transcript-RNA- templated DNA recombination and repair. Nature. Nature Publishing Group; 2014;515:436–9.

32. Wang T, Wei JJ, Sabatini DM, Lander ES. Genetic screens in human cells using the CRISPR-Cas9 system. Science. 2014;343:80–4.

33. Shalem O, Sanjana EN, Hartenian E, Zhang F. Genome-Scale CRISPR-Cas9 Knockout. Science. 2014;343:84–8.

34. Sanjana NE, Shalem O, Zhang F. Improved vectors and genome-wide libraries for CRISPR screening. Nat Methods. 2014;11:783–4.

35. Munoz DM, Cassiani PJ, Li L, Billy E, Korn JM, Jones MD, et al. CRISPR screens provide a comprehensive assessment of cancer vulnerabilities but generate false-positive hits for highly amplified genomic regions. Cancer Discov. 2016;6:900–13.

36. Lieber . Mazzetta, J,. Nelson-Rees, W,. Kaplan, M,. Todaro, G. M. Establishment of a continuous tumor-cell line (panc-1) from a human carcinoma of the exocrin pancreas. Int J Cancer. 1975;15:741–7.

37. Subramanian A, Tamayo P, Mootha V. GSEA : Gene set enrichment analysis Gene set enrichment analysis : A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005;102:15545-50.

38. Luo B, Cheung H. Highly parallel identification of essential genes in cancer cells. Proc Natl Acad Sci USA. 2008;105:20380–5.

39. Thomas PD, Campbell MJ, Kejariwal A, Mi H, Karlak B, Daverman R, et al. PANTHER: A library of protein families and subfamilies indexed by function. Genome Res. 2003;13:2129–41.

40. Lokossou AG, Toudic C, Barbeau B. Implication of human endogenous retrovirus envelope proteins in placental functions. Viruses. 2014;6:4609–27.

41. Vercauteren D, Vandenbroucke RE, Jones AT, Rejman J, Demeester J, De Smedt SC, et al. The use of inhibitors to study endocytic pathways of gene carriers: Optimization and pitfalls. Mol Ther. 2010;18:561–9.

42. Lewis CS, Voelkel-Johnson C, Smith CD. Suppression of c-Myc and RRM2 expression in pancreatic cancer cells by the sphingosine kinase-2 inhibitor ABC294640. Oncotarget. 2016;7:60181–92.

43. Stellas D, Szabolcs M, Koul S, Li Z, Polyzos A, Anagnostopoulos C, et al. Therapeutic effects of an anti-Myc drug on mouse pancreatic cancer. J Natl Cancer Inst. 2014;106.

44. Leung JY, Ehmann GL, Giangrande PH, Nevins JR. A role for Myc in facilitating transcription activation by E2F1. Oncogene. 2008;27:4172–9.

45. Fernandez PC, Frank SR, Wang L, Schroeder M, Liu S, Greene J, et al. Genomic targets of the human c-Myc protein. Genes Dev. 2003;17:1115–29.

46. Hellwege JN, Velez Edwards DR, Acra S, Chen K, Buchowski MS, Edwards TL. Association of gene coding variation and resting metabolic rate in a multi- ethnic sample of children and adults. BMC Obes. BMC Obesity; 2017;4:12.

47. Wong AK, Krishnan A, Yao V, Tadych A, Troyanskaya OG. IMP 2.0: A multi- species functional genomics portal for integration, visualization and prediction of protein functions and networks. Nucleic Acids Res. 2015;43:W128–33.

48. De Sousa Cavalcante L, Monteiro G. Gemcitabine: Metabolism and molecular mechanisms of action, sensitivity and chemoresistance in pancreatic cancer. Eur J Pharmacol. Elsevier; 2014;741:8–16.

49. Chrestian N, Dupré N, Gan-Or Z, Szuto A, Chen S, Venkitachalam A, et al. Clinical and genetic study of hereditary spastic paraplegia in Canada. Neurol Genet. 2017;3:e122.

50. Ishiura H, Takahashi Y, Hayashi T, Saito K, Furuya H, Watanabe M, et al. Molecular epidemiology and clinical spectrum of hereditary spastic paraplegia in the Japanese population based on comprehensive mutational analyses. J Hum Genet. Nature Publishing Group; 2014;59:163–72.

51. Derivery E, Gautreau A. Evolutionary conservation of the WASH complex, an actin polymerization machine involved in endosomal fission. Commun Integr Biol. 2010;3:227–30.

52. Fletcher SJ, Johnson B, Lowe GC, Bem D, Drake S, Lordkipanidz M, et al. SLFN14 mutations underlie thrombocytopenia with excessive bleeding and platelet secretion defects. J Clin Invest. 2015;125:3600–5.

53. Wong HY, Wang GM, Croessmann S, Zabransky DJ, Chu D, Garay JP, et al. TMSB4Y is a candidate tumor suppressor on the Y chromosome and is deleted in male breast cancer. Oncotarget. 2015;6.

54. Katoh K, Shibata H, Suzuki H, Nara A, Ishidoh K, Kominami E, et al. The ALG- 2-interacting protein Alix associates with CHMP4b, a human homologue of yeast Snf7 that is involved in multivesicular body sorting. J Biol Chem. 2003;278:39104–13.

55. Abad MA, Zou J, Medina-Pritchard B, Nigg EA, Rappsilber J, Santamaria A, et al. Ska3 Ensures Timely Mitotic Progression by Interacting Directly With Microtubules and Ska1 Microtubule Binding Domain. Sci Rep. 2016;6:34042.

56. Trazzi S, Bernardoni R, Diolaiti D, Politi V, Earnshaw WC, Perini G, et al. In vivo functional dissection of human inner kinetochore protein CENP-C. J Struct Biol. 2002;140:39–48.

57. Kim K, Lee S, Chang J, Rhee K. A novel function of CEP135 as a platform protein of C-NAP1 for its centriolar localization. Exp Cell Res. Elsevier Inc.2008;314:3692–700.

58. Hamed SS, Straubinger RM, Jusko WJ. Pharmacodynamic modeling of cell cycle and apoptotic effects of gemcitabine on pancreatic adenocarcinoma cells. Cancer Chemother Pharmacol. 2013;72:553–63.

59. Hesler RA, Huang JJ, Starr MD, Treboschi VM, Bernanke AG, Nixon AB, et al. TGF-β-induced stromal CYR61 promotes resistance to gemcitabine in pancreatic ductal adenocarcinoma through downregulation of the nucleoside transporters hENT1 and hCNT3. Carcinogenesis. 2016;37:1041–51.

60. Paproski RJ, Yao SYM, Favis N, Evans D, Young JD, Cass CE, et al. Human Concentrative Nucleoside Transporter 3 Transfection with Ultrasound and Microbubbles in Nucleoside Transport Deficient HEK293 Cells Greatly Increases Gemcitabine Uptake. PLoS One. 2013;8.

参考文献をもっと見る