ZNAČAJ OKSIDATIVNOG STRESA U PATOGENEZI OVARIJALNOG KARCINOMA

  • Petar Simić Medicinski fakultet Univerzitet u Beogradu
  • Marija Plješa Ercegovac
  • Vesna Ćorić
  • Vladimir Pažin
Ključne reči: ovarijalni karcinom, oksidativni stres, patogeneza ovarijalnog karcinoma

Sažetak


Ovarijalni karcinom predstavlja najsmrtonosniji ginekološki malignitet. Nespecifične tegobe, uz otkrivanje bolesti u uznapredovalom stadijumu, predstavljaju osnovne faktore loše prognoze sa ukupnim petogodišnjim preživljavanjem od 45%. Za sada, terapiju prvog izbora u tretmanu ovarijalnog karcinoma predstavlja hirurško lečenje sa ciljem potpune eliminacije makroskopske bolesti, uz adjuvantnu primenu hemioterapije. Važnost rane dijagnoze se ogleda u brojnim sprovedenim prospektivnim istraživanjima, koja nisu identifikovala efikasan način rane detekcije ove bolesti. Oksidativni stres, kao disbalans u produkciji i eliminaciji reaktivnih jedinjenja kiseonika, dokazani je faktor karcinogeneze kod mnogih karcinoma. Reaktivne kiseonične vrste imaju svoj uticaj, kako na patološke, tako i na fiziološke procese. Kada je u pitanju oksidativni stres, literaturni podaci ukazuju na njegov značaj u  patogenezi ovarijalnog karcinoma kroz sledeća četiri aspekta ćelijskog funkcionisanja: (i) genetske alteracije, (ii) signalni putevi, (iii) uticaj na transkripcione faktore, kao i na (iv) tumorsko mikro-okruženje. Reaktivne kiseonične vrste mogu da dovedu do nastanka mutacija, što prouzrokuje inicijalnu genezu, rast i progresiju tumora tkiva. Takođe, dugoročno povećana koncentracija slobodnih radikala može imati citotoksični efekat, koji nastaje usled modulacije redoks-zavisnih puteva apoptoze. Pod uticajem slobodnih radikala, dolazi do oštećenja biomolekula i formiranja produkata ove interakcije, što za posledicu ima pojavu ovih jedinjenja u tkivu, plazmi, urinu kao i u mnogim drugim odeljcima u kojima se mogu identifikovati. S obzirom na izrazito nepoznatu etiologiju, kao i heterogenost ovarijalnog karcinoma, a uz do sada sprovedena istraživanja koja su najvećim delom bila fokusirana na operativno ili medikamentozno zbrinjavanje, cilj ovog preglednog rada je da se čitaocima približi značajna uloga oksidativnog stresa u procesu patogeneze ovarijalnog karcinoma.

Reference

Doherty JA, Peres LC, Wang C, Way GP, Greene CS, Schildkraut JM. Challenges and Opportunities in Studying the Epidemiology of Ovarian Cancer Subtypes. Curr Epidemiol Rep. 2017 Sep;4(3):211-220. doi: 10.1007/s40471-017-0115-y.

Reid BM, Permuth JB, Sellers TA. Epidemiology of ovarian cancer: a review. Cancer Biol Med. 2017 Feb;14(1):9-32. doi: 10.20892/j.issn.2095-3941.2016.0084.

Harter P, Hilpert F, Mahner S, Kommoss S, Heitz F, du Bois A. Role of cytoreductive surgery in recurrent ovarian cancer. Expert Rev Anticancer Ther. 2009 Jul;9(7):917-22. doi: 10.1586/era.09.49.

Howlader N, Morton LM, Feuer EJ, Besson C, Engels EA. Contributions of Subtypes of Non-Hodgkin Lymphoma to Mortality Trends. Cancer Epidemiol Biomarkers Prev. 2016 Jan;25(1):174-9. doi: 10.1158/1055-9965.EPI-15-0921.

Lheureux S, Gourley C, Vergote I, Oza AM. Epithelial ovarian cancer. Lancet. 2019 Mar 23;393(10177):1240-1253. doi: 10.1016/S0140-6736(18)32552-2.

Bewtra C, Watson P, Conway T, Read-Hippee C, Lynch HT. Hereditary ovarian cancer: a clinicopathological study. Int J Gynecol Pathol. 1992 Jul;11(3):180-7. doi: 10.1097/00004347-199207000-00003.

Jacobs IJ, Menon U, Ryan A, Gentry-Maharaj A, Burnell M, Kalsi JK, et al. Ovarian cancer screening and mortality in the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS): a randomised controlled trial. Lancet Lond Engl. 2016 Mar 5;387(10022):945–56. doi: 10.1016/S0140-6736(15)01224-6.

Skates SJ, Greene MH, Buys SS, Mai PL, Brown P, Piedmonte M, et al. Early Detection of Ovarian Cancer using the Risk of Ovarian Cancer Algorithm with Frequent CA125 Testing in Women at Increased Familial Risk – Combined Results from Two Screening Trials. Clin Cancer Res. 2017 Jul 15;23(14):3628–37. doi: 10.1158/1078-0432.CCR-15-2750.

Shih IeM, Kurman RJ. Ovarian tumorigenesis: a proposed model based on morphological and molecular genetic analysis. Am J Pathol. 2004 May;164(5):1511-8. doi: 10.1016/s0002-9440(10)63708-x.

Kurman RJ, Shih IeM. The Dualistic Model of Ovarian Carcinogenesis: Revisited, Revised, and Expanded. Am J Pathol. 2016 Apr;186(4):733-47. doi: 10.1016/j.ajpath.2015.11.011.

Singer CA, Baker KJ, McCaffrey A, AuCoin DP, Dechert MA, Gerthoffer WT. p38 MAPK and NF-kappaB mediate COX-2 expression in human airway myocytes. Am J Physiol Lung Cell Mol Physiol. 2003 Nov;285(5):L1087-98. doi: 10.1152/ajplung.00409.2002.

Sheu JJC, Lee CC, Hua CH, Li CI, Lai MT, Lee SC, et al. LRIG1 modulates aggressiveness of head and neck cancers by regulating EGFR-MAPK-SPHK1 signaling and extracellular matrix remodeling. Oncogene. 2014 Mar 13;33(11):1375–84. doi: 10.1038/onc.2013.98.

Vang R, Levine DA, Soslow RA, Zaloudek C, Shih IM, Kurman RJ. Molecular Alterations of TP53 are a Defining Feature of Ovarian High-Grade Serous Carcinoma: A Rereview of Cases Lacking TP53 Mutations in The Cancer Genome Atlas Ovarian Study. Int J Gynecol Pathol Off J Int Soc Gynecol Pathol. 2016 Jan;35(1):48–55. doi: 10.1097/PGP.0000000000000207.

Boon WC, Chow JD, Simpson ER. The multiple roles of estrogens and the enzyme aromatase. Prog Brain Res. 2010;181:209-32. doi: 10.1016/S0079-6123(08)81012-6.

Chen P, Li B, Ou-Yang L. Role of estrogen receptors in health and disease. Front Endocrinol (Lausanne). 2022 Aug 18;13:839005. doi: 10.3389/fendo.2022.839005.

Revankar CM, Cimino DF, Sklar LA, Arterburn JB, Prossnitz ER. A transmembrane intracellular estrogen receptor mediates rapid cell signaling. Science. 2005 Mar 11;307(5715):1625-30. doi: 10.1126/science.1106943.

Yan Y, Liu H, Wen H, Jiang X, Cao X, Zhang G, et al. The novel estrogen receptor GPER regulates the migration and invasion of ovarian cancer cells. Mol Cell Biochem. 2013 Jun;378(1-2):1-7. doi: 10.1007/s11010-013-1579-9.

Goh YX, Jalil J, Lam KW, Husain K, Premakumar CM. Genistein: A Review on its Anti-Inflammatory Properties. Front Pharmacol. 2022 Jan 24;13:820969. doi: 10.3389/fphar.2022.820969.

Chan N, Toppmeyer DL. The Final Verdict: Chemotherapy Benefits Estrogen Receptor-Negative Isolated Local Recurrence. J Clin Oncol. 2018 Apr 10;36(11):1058-1059. doi: 10.1200/JCO.2017.77.4877.

Sang C, Song Y, Jin TW, Zhang S, Fu L, Zhao Y, et al. Bisphenol A induces ovarian cancer cell proliferation and metastasis through estrogen receptor-α pathways. Environ Sci Pollut Res Int. 2021 Jul;28(27):36060–8. doi: 10.1007/s11356-021-13267-0.

Di Meo S, Venditti P. Evolution of the Knowledge of Free Radicals and Other Oxidants. Oxid Med Cell Longev. 2020;2020:9829176. doi: 10.1155/2020/9829176.

Kamata H, Hirata H. Redox regulation of cellular signalling. Cell Signal. 1999 Jan;11(1):1-14. doi: 10.1016/s0898-6568(98)00037-0.

Jabs T. Reactive oxygen intermediates as mediators of programmed cell death in plants and animals. Biochem Pharmacol. 1999 Feb 1;57(3):231–45. doi: 10.1016/s0006-2952(98)00227-5.

Fridovich I. Superoxide radicals, superoxide dismutases and the aerobic lifestyle. Photochem Photobiol. 1978 Oct-Nov;28(4-5):733-41. doi: 10.1111/j.1751-1097.1978.tb07009.x.

Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med. 2010 Dec 1;49(11):1603–16. doi: 10.1016/j.freeradbiomed.2010.09.006.

Redza-Dutordoir M, Averill-Bates DA. Activation of apoptosis signalling pathways by reactive oxygen species. Biochim Biophys Acta. 2016 Dec;1863(12):2977–92. doi: 10.1016/j.bbamcr.2016.09.012.

Ding DN, Xie LZ, Shen Y, Li J, Guo Y, Fu Y, et al. Insights into the Role of Oxidative Stress in Ovarian Cancer. Oxid Med Cell Longev. 2021;2021:8388258. doi: 10.1155/2021/8388258.

Ye M, Dewi L, Liao YC, Nicholls A, Huang CY, Kuo CH. DNA oxidation after exercise: a systematic review and meta-analysis. Front Physiol. 2023;14:1275867. doi: 10.3389/fphys.2023.1275867.

Djukic T, Simic T, Radic T, Matic M, Pljesa-Ercegovac M, Suvakov S, et al. GSTO1*C/GSTO2*G haplotype is associated with risk of transitional cell carcinoma of urinary bladder. Int Urol Nephrol. 2015 Apr;47(4):625–30. doi: 10.1007/s11255-015-0933-0.

Bumbasirevic U, Bojanic N, Pljesa-Ercegovac M, Zivkovic M, Djukic T, Zekovic M, et al. The Polymorphisms of Genes Encoding Catalytic Antioxidant Proteins Modulate the Susceptibility and Progression of Testicular Germ Cell Tumor. Cancers. 2022 Feb 20;14(4):1068. doi: 10.3390/cancers14041068.

Valavanidis A, Vlachogianni T, Fiotakis C. 8-hydroxy-2’ -deoxyguanosine (8-OHdG): A critical biomarker of oxidative stress and carcinogenesis. J Environ Sci Health Part C Environ Carcinog Ecotoxicol Rev. 2009 Apr;27(2):120–39. doi: 10.1080/10590500902885684.

Ni J, Wang J. [Mitochondrial genome variants and neurodegenerative diseases]. Zhonghua Yi Xue Yi Chuan Xue Za Zhi Zhonghua Yixue Yichuanxue Zazhi Chin J Med Genet. 2020 Aug 10;37(8):898–902. doi: 10.3760/cma.j.issn.1003-9406.2020.08.023.

Mijatović S, Savić-Radojević A, Plješa-Ercegovac M, Simić T, Nicoletti F, Maksimović-Ivanić D. The Double-Faced Role of Nitric Oxide and Reactive Oxygen Species in Solid Tumors. Antioxid Basel Switz. 2020 Apr 30;9(5):374. doi: 10.3390/antiox9050374.

Pakuła M, Mikuła-Pietrasik J, Stryczyński Ł, Uruski P, Szubert S, Moszyński R, et al. Mitochondria-related oxidative stress contributes to ovarian cancer-promoting activity of mesothelial cells subjected to malignant ascites. Int J Biochem Cell Biol. 2018 May;98:82–8. doi: 10.1016/j.biocel.2018.03.011.

Yang X, Yu Y, Huang X, Chen Q, Wu H, Wang R, et al. Delivery of platinum (II) drugs with bulky ligands in trans-geometry for overcoming cisplatin drug resistance. Mater Sci Eng C Mater Biol Appl. 2019 Mar;96:96–104. doi: 10.1016/j.msec.2018.10.092.

Hou L, Wang K, Zhang C, Sun F, Che Y, Zhao X, et al. Complement receptor 3 mediates NADPH oxidase activation and dopaminergic neurodegeneration through a Src-Erk-dependent pathway. Redox Biol. 2018 Apr;14:250–60. doi: 10.1016/j.redox.2017.09.017.

Bai X, Liu F, Liu Y, Li C, Wang S, Zhou H, et al. Toward a systematic exploration of nano-bio interactions. Toxicol Appl Pharmacol. 2017 May 15;323:66–73. doi: 10.1016/j.taap.2017.03.011.

Frijhoff J, Winyard PG, Zarkovic N, Davies SS, Stocker R, Cheng D, et al. Clinical Relevance of Biomarkers of Oxidative Stress. Antioxid Redox Signal. 2015 Nov 10;23(14):1144–70. doi: 10.1089/ars.2015.6317.

Zheng M, Hu Y, Liu O, Li S, Wang Y, Li X, et al. Oxidative Stress Response Biomarkers of Ovarian Cancer Based on Single-Cell and Bulk RNA Sequencing. Oxid Med Cell Longev. 2023;2023:1261039. doi: 10.1155/2023/1261039.

Liu Q, Yang X, Yin Y, Zhang H, Yin F, Guo P, et al. Identifying the Role of Oxidative Stress-Related Genes as Prognostic Biomarkers and Predicting the Response of Immunotherapy and Chemotherapy in Ovarian Cancer. Oxid Med Cell Longev. 2022;2022:6575534. doi: 10.1155/2022/6575534.

Stocker R, Keaney JF. Role of oxidative modifications in atherosclerosis. Physiol Rev. 2004 Oct;84(4):1381–478. doi: 10.1152/physrev.00047.2003.

Wu D, Zhai Q, Shi X. Alcohol-induced oxidative stress and cell responses. J Gastroenterol Hepatol. 2006 Oct;21 Suppl 3:S26-29. doi: 10.1111/j.1440-1746.2006.04589.x.

Kryston TB, Georgiev AB, Pissis P, Georgakilas AG. Role of oxidative stress and DNA damage in human carcinogenesis. Mutat Res. 2011 Jun 3;711(1–2):193–201. doi: 10.1016/j.mrfmmm.2010.12.016.

Jelić M, Mandić A, Kladar N, Sudji J, Božin B, Srdjenović B. Lipid Peroxidation, Antioxidative Defense and Level of 8-Hydroxy-2-Deoxyguanosine in Cervical Cancer Patients. J Med Biochem. 2018 Jul 1;37(3):336–45. doi: 10.1515/jomb-2017-0053.

Pylväs-Eerola M, Karihtala P, Puistola U. Preoperative serum 8-hydroxydeoxyguanosine is associated with chemoresistance and is a powerful prognostic factor in endometrioid-type epithelial ovarian cancer. BMC Cancer. 2015 Jul 2;15:493. doi: 10.1186/s12885-015-1504-6.

Fujimoto Y, Imanaka S, Yamada Y, Ogawa K, Ito F, Kawahara N, et al. Comparison of redox parameters in ovarian endometrioma and its malignant transformation. Oncol Lett [Internet]. 2018 Jul 31 [cited 2024 Mar 31]; Available from: http://www.spandidos-publications.com/10.3892/ol.2018.9242.

Dean RT, Fu S, Stocker R, Davies MJ. Biochemistry and pathology of radical-mediated protein oxidation. Biochem J. 1997 May 15;324 ( Pt 1)(Pt 1):1–18. doi: 10.1042/bj3240001.

Gil L, Siems W, Mazurek B, Gross J, Schroeder P, Voss P, et al. Age-associated analysis of oxidative stress parameters in human plasma and erythrocytes. Free Radic Res. 2006 May;40(5):495–505. doi: 10.1080/10715760600592962.

Davies SS, Bodine C, Matafonova E, Pantazides BG, Bernoud-Hubac N, Harrison FE, et al. Treatment with a γ-ketoaldehyde scavenger prevents working memory deficits in hApoE4 mice. J Alzheimers Dis JAD. 2011;27(1):49–59. doi: 10.3233/JAD-2011-102118.

Mažibrada I, Djukić T, Perović S, Plješa-Ercegovac M, Plavšić L, Bojanin D, et al. The association of hs-CRP and fibrinogen with anthropometric and lipid parameters in non-obese adolescent girls with polycystic ovary syndrome. J Pediatr Endocrinol Metab [Internet]. 2018 Oct 18 [cited 2024 Apr 9];0(0). Available from: https://www.degruyter.com/document/doi/10.1515/jpem-2017-0511/html.

Objavljeno
2024/07/09
Rubrika
Pregledni članci