Yıl: 2023 Cilt: 8 Sayı: 1 Sayfa Aralığı: 20 - 26 Metin Dili: İngilizce DOI: 10.4274/cjms.2022.2022-29 İndeks Tarihi: 30-05-2023

A New Gene Therapy Approach by Tenascin-C Genome Editing Induces Apoptosis and Cell Cycle Arrest in Triple Negative Breast Cancer Cells

Öz:
BACKGROUND/AIMS: There is a pressing need for new therapies for the most aggressive subtype of breast cancer, triple-negative breast cancer (TNBC). Tenascin-C (TN-C) codes for a tumor microenvironment-specific protein, which promotes apoptosis evasion and cell proliferation. The aim of this study was to knock down TN-C by using the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system to induce cancer cell apoptosis and stunt cell proliferation, laying the grounds for a new gene therapy approach in TNBC. MATERIALS and METHODS: The human TNBC cell line, MDA-MB-231 cells were transfected by TN-C-specific CRISPR/Cas9 plasmids. TN-C messenger RNA levels were assessed by real-time polymerase chain reaction to determine the knock-down efficiency. Two days after the transfection, the percentage of apoptotic cells and the proportion of cells in cell cycle phases were compared between the treatment and the control groups using flow cytometry. The resultant change in cell proliferation due to the knock-down was determined by MTT assay. RESULTS: Transfection with the TN-C CRISPR/Cas9 plasmid reduced TN-C levels in the cells by approximately 49% relative to the scrambled control CRISPR/Cas9 transfected cells. This TN-C downregulation increased the percentage of cells in apoptosis and induced G1-phase arrest. The combined effect of apoptosis and cell cycle arrest led to a significant decrease in the number of cancer cells in the treatment group. CONCLUSION: Our successful preliminary study of a potential TNBC gene therapy based on TN-C genome editing by the CRISPR/Cas9 system led to significant decrease in TNBC cell numbers and it justifies the testing of this system in more advanced preclinical studies.
Anahtar Kelime:

Belge Türü: Makale Makale Türü: Araştırma Makalesi Erişim Türü: Erişime Açık
  • 1. Thun MJ, DeLancey JO, Center MM, Jemal A, Ward EM. The global burden of cancer: priorities for prevention. Carcinogenesis. 2010; 31(1): 100-10.
  • 2. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021; 71(3): 209-49.
  • 3. Hanahan D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022; 12(1): 31-46.
  • 4. Singh R, Letai A, Sarosiek K. Regulation of apoptosis in health and disease: the balancing act of BCL-2 family proteins. Nat Rev Mol Cell Biol. 2019; 20(3): 175-93.
  • 5. Mohammad RM, Muqbil I, Lowe L, Yedjou C, Hsu HY, Lin LT, et al. Broad targeting of resistance to apoptosis in cancer. Semin Cancer Biol. 2015; 35: S78-103.
  • 6. Bożyk A, Wojas-Krawczyk K, Krawczyk P, Milanowski J. Tumor Microenvironment-A short Review of Cellular and Interaction Diversity. Biology. 2022; 11(6): 929.
  • 7. Nallanthighal S, Heiserman JP, Cheon DJ. The Role of The Extracellular Matrix in Cancer Stemness. Front Cell Dev Biol. 2019; 7: 86.
  • 8. Hui L, Chen Y. Tumor microenvironment: Sanctuary of the devil. Cancer Lett. 2015; 368(1): 7-13.
  • 9. Chong HC, Tan CK, Huang RL, Tan NS. Matricellular proteins: a sticky affair with cancers. J Oncol. 2012; 2012: 351089.
  • 10. Cai J, Lu W, Du S, Guo Z, Wang H, Wei W, et al. Tenascin-C Modulates Cell Cycle Progression to Enhance Tumour Cell Proliferation through AKT/FOXO1 Signalling in Pancreatic Cancer. J Cancer. 2018; 9(23): 4449-62.
  • 11. Guttery DS, Shaw JA, Lloyd K, Pringle JH, Walker RA. Expression of tenascin-C and its isoforms in the breast. Cancer Metastasis Rev. 2010; 29(4): 595-606.
  • 12. Midwood KS, Orend G. The role of tenascin-C in tissue injury and tumorigenesis. J Cell Commun Signal. 2009; 3(3-4): 287-310.
  • 13. Ming X, Qiu S, Liu X, Li S, Wang Y, Zhu M, et al. Prognostic Role of Tenascin-C for Cancer Outcome: A Meta-Analysis. Technol Cancer Res Treat. 2019; 18: 1533033818821106.
  • 14. Hirakawa MP, Krishnakumar R, Timlin JA, Carney JP, Butler KS. Gene editing and CRISPR in the clinic: current and future perspectives. Bioscience Rep. 2020; 40(4): BSR20200127.
  • 15. Yang Z, Zhang C, Feng Y, Quan M, Cui Y, Xuan Y. Tenascin-C predicts poor outcomes for patients with colorectal cancer and drives cancer stemness via Hedgehog signaling pathway. Cancer Cell Int. 2020; 20: 122.
  • 16. Hongu T, Pein M, Insua-Rodríguez J, Gutjahr E, Mattavelli G, Meier J, et al. Perivascular tenascin C triggers sequential activation of macrophages and endothelial cells to generate a pro-metastatic vascular niche in the lungs. Nat Cancer. 2022; 3(4): 486-504.
  • 17. Insua-Rodríguez J, Pein M, Hongu T, Meier J, Descot A, Lowy CM, et al. Stress signaling in breast cancer cells induces matrix components that promote chemoresistant metastasis. EMBO Mol Med. 2018; 10(10): e9003.
  • 18. Tucker RP, Degen M. Revisiting the Tenascins: Exploitable as cancer targets? Front Oncol. 2022; 12: 908247.
  • 19. Li ZL, Zhang HL, Huang Y, Huang JH, Sun P, Zhou NN, et al. Autophagy deficiency promotes triple-negative breast cancer resistance to T cell mediated cytotoxicity by blocking tenascin-C degradation. Nat Commun. 2020; 11(1): 3806.
  • 20. Bergin ART, Loi S. Triple-negative breast cancer: recent treatment advances. F1000Res. 2019; 8: 1342.
  • 21. Deepak KGK, Vempati R, Nagaraju GP, Dasari VR, SN, Rao DN, Malla RR. Tumor microenvironment: Challenges and opportunities in targeting metastasis of triple negative breast cancer. Pharmacol Res. 2020; 153: 104683.
  • 22. Wawrzyniak D, Grabowska M, Głodowicz P, Kuczyński K, Kuczyńska B, Fedoruk-Wyszomirska A, et al. Down-regulation of tenascin-C inhibits breast cancer cells development by cell growth, migration, and adhesion impairment. Languino LR, editor. PLoS One. 2020; 15(8): e0237889.
  • 23. Wang B, Liu K, Lin HY, Bellam N, Ling S, Lin WC. 14-3-3Tau regulates ubiquitin independent proteasomal degradation of p21, a novel mechanism of p21 downregulation in breast cancer. Mol Cell Biol. 2010; 30(6): 1508-27.
  • 24. Shi M, He X, Wei W, Wang J, Zhang T, Shen X. Tenascin-C induces resistance to apoptosis in pancreatic cancer cell through activation of ERK/NF-κB pathway. Apoptosis. 2015; 20(6): 843-57.
  • 25. Seo Y, Shin TH, Ahn JS, Oh SJ, Shin YY, Yang JW, et al. Human Tonsil-Derived Mesenchymal Stromal Cells Maintain Proliferating and ROS-Regulatory Properties via Stanniocalcin-1. Cells. 2020; 9(3): 636.
  • 26. Onarheim KH, Iversen JH, Bloom DE. Economic Benefits of Investing in Women’s Health: A Systematic Review. PLoS One. 2016; 11(3): e0150120.
  • 27. Oskarsson T, Acharyya S, Zhang XH, Vanharanta S, Tavazoie SF, Morris PG, et al. Breast cancer cells produce tenascin C as a metastatic niche component to colonize the lungs. Nat Med. 2011; 17(7): 867-74.
  • 28. Sun Z, Velázquez-Quesada I, Murdamoothoo D, Ahowesso C, Yilmaz A, Spenlé C, et al. Tenascin-C increases lung metastasis by impacting blood vessel invasions. Matrix Biol. 2019; 83: 26-47.
  • 29. Teschendorf C, Warrington KH, Siemann DW, Muzyczka N. Comparison of the EF-1 alpha and the CMV promoter for engineering stable tumor cell lines using recombinant adeno-associated virus. Anticancer Res. 2002; 22(6A): 3325-30.
  • 30. Radzisheuskaya A, Shlyueva D, Müller I, Helin K. Optimizing sgRNA position markedly improves the efficiency of CRISPR/dCas9-mediated transcriptional repression. Nucleic Acids Res. 2016; 44(18): e141.
  • 31. Patel AG, Sarkaria JN, Kaufmann SH. Nonhomologous end joining drives poly(ADP-ribose) polymerase (PARP) inhibitor lethality in homologous recombination-deficient cells. Proc Natl Acad Sci USA. 2011; 108(8): 3406-11.
  • 32. Iyoda T, Fujita M, Fukai F. Biologically Active TNIIIA2 Region in Tenascin-C Molecule: A Major Contributor to Elicit Aggressive Malignant Phenotypes From Tumors/Tumor Stroma. Front Immunol. 2020; 11: 610096.
  • 33. Qian S, Tan X, Liu X, Liu P, Wu Y. Exosomal Tenascin-c induces proliferation and invasion of pancreatic cancer cells by WNT signaling. Onco Targets Ther. 2019; 12: 3197-205.
APA Bareke H, Salva E, OZBAS TURAN S (2023). A New Gene Therapy Approach by Tenascin-C Genome Editing Induces Apoptosis and Cell Cycle Arrest in Triple Negative Breast Cancer Cells. , 20 - 26. 10.4274/cjms.2022.2022-29
Chicago Bareke Halin,Salva Emine,OZBAS TURAN SUNA A New Gene Therapy Approach by Tenascin-C Genome Editing Induces Apoptosis and Cell Cycle Arrest in Triple Negative Breast Cancer Cells. (2023): 20 - 26. 10.4274/cjms.2022.2022-29
MLA Bareke Halin,Salva Emine,OZBAS TURAN SUNA A New Gene Therapy Approach by Tenascin-C Genome Editing Induces Apoptosis and Cell Cycle Arrest in Triple Negative Breast Cancer Cells. , 2023, ss.20 - 26. 10.4274/cjms.2022.2022-29
AMA Bareke H,Salva E,OZBAS TURAN S A New Gene Therapy Approach by Tenascin-C Genome Editing Induces Apoptosis and Cell Cycle Arrest in Triple Negative Breast Cancer Cells. . 2023; 20 - 26. 10.4274/cjms.2022.2022-29
Vancouver Bareke H,Salva E,OZBAS TURAN S A New Gene Therapy Approach by Tenascin-C Genome Editing Induces Apoptosis and Cell Cycle Arrest in Triple Negative Breast Cancer Cells. . 2023; 20 - 26. 10.4274/cjms.2022.2022-29
IEEE Bareke H,Salva E,OZBAS TURAN S "A New Gene Therapy Approach by Tenascin-C Genome Editing Induces Apoptosis and Cell Cycle Arrest in Triple Negative Breast Cancer Cells." , ss.20 - 26, 2023. 10.4274/cjms.2022.2022-29
ISNAD Bareke, Halin vd. "A New Gene Therapy Approach by Tenascin-C Genome Editing Induces Apoptosis and Cell Cycle Arrest in Triple Negative Breast Cancer Cells". (2023), 20-26. https://doi.org/10.4274/cjms.2022.2022-29
APA Bareke H, Salva E, OZBAS TURAN S (2023). A New Gene Therapy Approach by Tenascin-C Genome Editing Induces Apoptosis and Cell Cycle Arrest in Triple Negative Breast Cancer Cells. Cyprus Journal of Medical Sciences, 8(1), 20 - 26. 10.4274/cjms.2022.2022-29
Chicago Bareke Halin,Salva Emine,OZBAS TURAN SUNA A New Gene Therapy Approach by Tenascin-C Genome Editing Induces Apoptosis and Cell Cycle Arrest in Triple Negative Breast Cancer Cells. Cyprus Journal of Medical Sciences 8, no.1 (2023): 20 - 26. 10.4274/cjms.2022.2022-29
MLA Bareke Halin,Salva Emine,OZBAS TURAN SUNA A New Gene Therapy Approach by Tenascin-C Genome Editing Induces Apoptosis and Cell Cycle Arrest in Triple Negative Breast Cancer Cells. Cyprus Journal of Medical Sciences, vol.8, no.1, 2023, ss.20 - 26. 10.4274/cjms.2022.2022-29
AMA Bareke H,Salva E,OZBAS TURAN S A New Gene Therapy Approach by Tenascin-C Genome Editing Induces Apoptosis and Cell Cycle Arrest in Triple Negative Breast Cancer Cells. Cyprus Journal of Medical Sciences. 2023; 8(1): 20 - 26. 10.4274/cjms.2022.2022-29
Vancouver Bareke H,Salva E,OZBAS TURAN S A New Gene Therapy Approach by Tenascin-C Genome Editing Induces Apoptosis and Cell Cycle Arrest in Triple Negative Breast Cancer Cells. Cyprus Journal of Medical Sciences. 2023; 8(1): 20 - 26. 10.4274/cjms.2022.2022-29
IEEE Bareke H,Salva E,OZBAS TURAN S "A New Gene Therapy Approach by Tenascin-C Genome Editing Induces Apoptosis and Cell Cycle Arrest in Triple Negative Breast Cancer Cells." Cyprus Journal of Medical Sciences, 8, ss.20 - 26, 2023. 10.4274/cjms.2022.2022-29
ISNAD Bareke, Halin vd. "A New Gene Therapy Approach by Tenascin-C Genome Editing Induces Apoptosis and Cell Cycle Arrest in Triple Negative Breast Cancer Cells". Cyprus Journal of Medical Sciences 8/1 (2023), 20-26. https://doi.org/10.4274/cjms.2022.2022-29