DOI QR코드

DOI QR Code

Potential roles of reactive oxygen species derived from chemical substances involved in cancer development in the female reproductive system

  • Kim, Soo-Min (Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University) ;
  • Hwang, Kyung-A (Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University) ;
  • Choi, Kyung-Chul (Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University)
  • Received : 2018.03.18
  • Accepted : 2018.06.18
  • Published : 2018.11.30

Abstract

Reactive oxygen species (ROS) are major sources of cellular oxidative stress. Specifically, cancer cells harbor genetic alterations that promote a continuous and elevated production of ROS. While such oxidative stress conditions could be harmful to normal cells, they facilitate cancer cell growth in multiple ways by causing DNA damage and genomic instability, and ultimately by reprogramming cancer cell metabolism. This review provides up to date findings regarding the roles of ROS generation induced by diverse biological molecules and chemicals in representative women's cancer. Specifically, we describe the cellular signaling pathways that regulate direct or indirect interactions between ROS homeostasis and metabolism within female genital cancer cells.

Keywords

References

  1. Kim J, Kim J and Bae JS (2016) ROS homeostasis and metabolism: a critical liaison for cancer therapy. Exp Mol Med 48, e269 https://doi.org/10.1038/emm.2016.119
  2. Chrissobolis S and Faraci FM (2008) The role of oxidative stress and NADPH oxidase in cerebrovascular disease. Trends Mol Med 14, 495-502 https://doi.org/10.1016/j.molmed.2008.09.003
  3. Gius D and Spitz DR (2006) Redox signaling in cancer biology. Antioxid Redox Signal 8, 1249-1252 https://doi.org/10.1089/ars.2006.8.1249
  4. Galaris D, Mantzaris M and Amorgianiotis C (2008) Oxidative stress and aging: the potential role of iron. Hormones (Athens) 7, 114-122 https://doi.org/10.1007/BF03401502
  5. Van Wagoner DR (2008) Oxidative stress and inflammation in atrial fibrillation: role in pathogenesis and potential as a therapeutic target. J Cardiovasc Pharmacol 52, 306-313 https://doi.org/10.1097/FJC.0b013e31817f9398
  6. Rojas A, Silva R, Figueroa H and Morales MA (2008) Oxidative stress in tumor microenvironment--Its role in angiogenesis. Zhongguo Fei Ai Za Zhi 11, 297-305
  7. Giannoni E, Parri M and Chiarugi P (2012) EMT and oxidative stress: a bidirectional interplay affecting tumor malignancy. Antioxid Redox Signal 16, 1248-1263 https://doi.org/10.1089/ars.2011.4280
  8. Marengo B, Nitti M, Furfaro AL et al (2016) Redox Homeostasis and Cellular Antioxidant Systems: Crucial Players in Cancer Growth and Therapy. Oxid Med Cell Longev 2016, 6235641
  9. Ngo C, Chereau C, Nicco C, Weill B, Chapron C and Batteux F (2009) Reactive oxygen species controls endometriosis progression. Am J Pathol 175, 225-234 https://doi.org/10.2353/ajpath.2009.080804
  10. Jiang Z, Fletcher NM, Ali-Fehmi R et al (2011) Modulation of redox signaling promotes apoptosis in epithelial ovarian cancer cells. Gynecol Oncol 122, 418-423 https://doi.org/10.1016/j.ygyno.2011.04.051
  11. Bhat AV, Hora S, Pal A, Jha S and Taneja R (2017) Stressing the (epi)genome: dealing with ROS in cancer. Antioxid Redox Signal [Epub ahead of print]
  12. Strzelczyk JK and Wiczkowski A (2012) Oxidative damage and carcinogenesis. Contemp Oncol (Pozn) 16, 230-233
  13. Panieri E and Santoro MM (2016) ROS homeostasis and metabolism: a dangerous liason in cancer cells. Cell Death Dis 7, e2253 https://doi.org/10.1038/cddis.2016.105
  14. Moloney JN and Cotter TG (2018) ROS signalling in the biology of cancer. Semin Cell Dev Biol 80, 50-64 https://doi.org/10.1016/j.semcdb.2017.05.023
  15. Sieber OM, Heinimann K and Tomlinson IP (2003) Genomic instability--the engine of tumorigenesis? Nat Rev Cancer 3, 701-708 https://doi.org/10.1038/nrc1170
  16. Gao X and Schottker B (2017) Reduction-oxidation pathways involved in cancer development: a systematic review of literature reviews. Oncotarget 8, 51888-51906 https://doi.org/10.18632/oncotarget.17128
  17. Morry J, Ngamcherdtrakul W and Yantasee W (2017) Oxidative stress in cancer and fibrosis: Opportunity for therapeutic intervention with antioxidant compounds, enzymes, and nanoparticles. Redox Biol 11, 240-253 https://doi.org/10.1016/j.redox.2016.12.011
  18. Beckman RA and Loeb LA (2017) Evolutionary dynamics and significance of multiple subclonal mutations in cancer. DNA Repair (Amst) 56, 7-15 https://doi.org/10.1016/j.dnarep.2017.06.002
  19. Chan L, He L, Zhou B et al (2017) Cancer-targeted Selenium Nanoparticles Sensitize Cancer Cells to Continuous gamma Radiation to Achieve Synergetic Chemo-Radiotherapy. Chem Asian J 12, 3053-3060 https://doi.org/10.1002/asia.201701227
  20. He H, Chang R, Zhang T, Yang C and Kong Z (2017) ATM mediates DAB2IP-deficient bladder cancer cell resistance to ionizing radiation through the p38MAPK and NF-kappaB signaling pathway. Mol Med Rep 16, 1216-1222 https://doi.org/10.3892/mmr.2017.6689
  21. Xu Z, Zhang F, Bai C et al (2017) Sophoridine induces apoptosis and S phase arrest via ROS-dependent JNK and ERK activation in human pancreatic cancer cells. J Exp Clin Cancer Res 36, 124 https://doi.org/10.1186/s13046-017-0590-5
  22. Preya UH, Lee KT, Kim NJ, Lee JY, Jang DS and Choi JH (2017) The natural terthiophene alpha-terthienylmethanol induces S phase cell cycle arrest of human ovarian cancer cells via the generation of ROS stress. Chem Biol Interact 272, 72-79 https://doi.org/10.1016/j.cbi.2017.05.011
  23. Khan F, Khan I, Farooqui A and Ansari IA (2017) Carvacrol Induces Reactive Oxygen Species (ROS)-mediated Apoptosis Along with Cell Cycle Arrest at G0/G1 in Human Prostate Cancer Cells. Nutr Cancer 69, 1075-1087 https://doi.org/10.1080/01635581.2017.1359321
  24. McCubrey JA, Steelman LS, Chappell WH et al (2007) Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim Biophys Acta 1773, 1263-1284 https://doi.org/10.1016/j.bbamcr.2006.10.001
  25. Liu CT and Liu MY (2017) Daily sesame oil supplementation attenuates local renin-angiotensin system via inhibiting MAPK activation and oxidative stress in cardiac hypertrophy. J Nutr Biochem 42, 108-116 https://doi.org/10.1016/j.jnutbio.2016.05.006
  26. Huang H, Du W and Brekken RA (2017) Extracellular Matrix Induction of Intracellular Reactive Oxygen Species. Antioxid Redox Signal 27, 774-784 https://doi.org/10.1089/ars.2017.7305
  27. Ha H and Lee HB (2003) Reactive oxygen species and matrix remodeling in diabetic kidney. J Am Soc Nephrol 14, S246-249 https://doi.org/10.1097/01.ASN.0000077411.98742.54
  28. Seth D and Rudolph J (2006) Redox regulation of MAP kinase phosphatase 3. Biochemistry 45, 8476-8487 https://doi.org/10.1021/bi060157p
  29. Klaunig JE, Kamendulis LM and Hocevar BA (2010) Oxidative stress and oxidative damage in carcinogenesis. Toxicol Pathol 38, 96-109 https://doi.org/10.1177/0192623309356453
  30. Galadari S, Rahman A, Pallichankandy S and Thayyullathil F (2017) Reactive oxygen species and cancer paradox: To promote or to suppress? Free Radic Biol Med 104, 144-164 https://doi.org/10.1016/j.freeradbiomed.2017.01.004
  31. Red-Horse K, Zhou Y, Genbacev O et al (2004) Trophoblast differentiation during embryo implantation and formation of the maternal-fetal interface. J Clin Invest 114, 744-754 https://doi.org/10.1172/JCI200422991
  32. Hod T, Cerdeira AS and Karumanchi SA (2015) Molecular Mechanisms of Preeclampsia. Cold Spring Harb Perspect Med 5, a023473 https://doi.org/10.1101/cshperspect.a023473
  33. Kumar V, Abbas AK and Aster JC (2015) Robbins and Cotran pathologic basis of disease, Ninth edition. ed, Elsevier/Saunders, Philadelphia, PA
  34. Park SH, Park A, Kim JY, Kwon JH and Koh SB (2009) A case of non-gestational choriocarcinoma arising in the ovary of a postmenopausal woman. J Gynecol Oncol 20, 192-194 https://doi.org/10.3802/jgo.2009.20.3.192
  35. Lurain JR and Nejad B (2005) Secondary chemotherapy for high-risk gestational trophoblastic neoplasia. Gynecol Oncol 97, 618-623 https://doi.org/10.1016/j.ygyno.2005.02.004
  36. Renschler MF (2004) The emerging role of reactive oxygen species in cancer therapy. Eur J Cancer 40, 1934-1940 https://doi.org/10.1016/j.ejca.2004.02.031
  37. Yedjou CG and Tchounwou PB (2012) In vitro assessment of oxidative stress and apoptotic mechanisms of garlic extract in the treatment of acute promyelocytic leukemia. J Cancer Sci Ther 2012, 6
  38. Vallejo MJ, Salazar L and Grijalva M (2017) Oxidative Stress Modulation and ROS-Mediated Toxicity in Cancer: A Review on In Vitro Models for Plant-Derived Compounds. Oxid Med Cell Longev 2017, 4586068
  39. Huovinen M, Loikkanen J, Naarala J and Vahakangas K (2015) Toxicity of diuron in human cancer cells. Toxicol In Vitro 29, 1577-1586 https://doi.org/10.1016/j.tiv.2015.06.013
  40. Heazell AE, Taylor NN, Greenwood SL, Baker PN and Crocker IP (2009) Does altered oxygenation or reactive oxygen species alter cell turnover of BeWo choriocarcinoma cells? Reprod Biomed Online 18, 111-119 https://doi.org/10.1016/S1472-6483(10)60432-4
  41. Ham J, Lim W, Bazer FW and Song G (2018) Silibinin stimluates apoptosis by inducing generation of ROS and ER stress in human choriocarcinoma cells. J Cell Physiol 233, 1638-1649 https://doi.org/10.1002/jcp.26069
  42. Lim W, Yang C, Jeong M, Bazer FW and Song G (2017) Coumestrol induces mitochondrial dysfunction by stimulating ROS production and calcium ion influx into mitochondria in human placental choriocarcinoma cells. Mol Hum Reprod 23, 786-802 https://doi.org/10.1093/molehr/gax052
  43. Kim SM, Lee HM, Hwang KA and Choi KC (2017) Benzo(a)pyrene induced cell cycle arrest and apoptosis in human choriocarcinoma cancer cells through reactive oxygen species-induced endoplasmic reticulum-stress pathway. Food Chem Toxicol 107, 339-348 https://doi.org/10.1016/j.fct.2017.06.048
  44. Jeon SY, Hwang KA and Choi KC (2016) Effect of steroid hormones, estrogen and progesterone, on epithelial mesenchymal transition in ovarian cancer development. J Steroid Biochem Mol Biol 158, 1-8 https://doi.org/10.1016/j.jsbmb.2016.02.005
  45. Rojas V, Hirshfield KM, Ganesan S and Rodriguez- Rodriguez L (2016) Molecular Characterization of Epithelial Ovarian Cancer: Implications for Diagnosis and Treatment. Int J Mol Sci 17, E2113 https://doi.org/10.3390/ijms17122113
  46. Naora H and Montell DJ (2005) Ovarian cancer metastasis: integrating insights from disparate model organisms. Nat Rev Cancer 5, 355-366 https://doi.org/10.1038/nrc1611
  47. Auersperg N, Wong AS, Choi KC, Kang SK and Leung PC (2001) Ovarian surface epithelium: biology, endocrinology, and pathology. Endocr Rev 22, 255-288
  48. Bell DA (2005) Origins and molecular pathology of ovarian cancer. Mod Pathol 18 Suppl 2, S19-32 https://doi.org/10.1038/modpathol.3800306
  49. Fidler IJ (2002) The organ microenvironment and cancer metastasis. Differentiation 70, 498-505 https://doi.org/10.1046/j.1432-0436.2002.700904.x
  50. Holschneider CH and Berek JS (2000) Ovarian cancer: epidemiology, biology, and prognostic factors. Semin Surg Oncol 19, 3-10 https://doi.org/10.1002/1098-2388(200007/08)19:1<3::AID-SSU2>3.0.CO;2-S
  51. McCluggage WG (2011) Morphological subtypes of ovarian carcinoma: a review with emphasis on new developments and pathogenesis. Pathology 43, 420-432 https://doi.org/10.1097/PAT.0b013e328348a6e7
  52. Glorieux C, Sandoval JM, Fattaccioli A et al (2016) Chromatin remodeling regulates catalase expression during cancer cells adaptation to chronic oxidative stress. Free Radic Biol Med 99, 436-450 https://doi.org/10.1016/j.freeradbiomed.2016.08.031
  53. Tsikouras P, Bouchlariotou S, Vrachnis N et al (2013) Endometrial cancer: molecular and therapeutic aspects. Eur J Obstet Gynecol Reprod Biol 169, 1-9 https://doi.org/10.1016/j.ejogrb.2013.01.018
  54. Llaurado M, Ruiz A, Majem B et al (2012) Molecular bases of endometrial cancer: new roles for new actors in the diagnosis and the therapy of the disease. Mol Cell Endocrinol 358, 244-255 https://doi.org/10.1016/j.mce.2011.10.003
  55. Ali AT (2013) Risk factors for endometrial cancer. Ceska Gynekol 78, 448-459
  56. Kim JY, Lee SG, Chung JY et al (2011) Ellipticine induces apoptosis in human endometrial cancer cells: the potential involvement of reactive oxygen species and mitogen-activated protein kinases. Toxicology 289, 91-102 https://doi.org/10.1016/j.tox.2011.07.014
  57. Manohar M, Fatima I, Saxena R, Chandra V, Sankhwar PL and Dwivedi A (2013) (-)-Epigallocatechin-3-gallate induces apoptosis in human endometrial adenocarcinoma cells via ROS generation and p38 MAP kinase activation. J Nutr Biochem 24, 940-947 https://doi.org/10.1016/j.jnutbio.2012.06.013
  58. Yoshioka T, Yogosawa S, Yamada T, Kitawaki J and Sakai T (2013) Combination of a novel HDAC inhibitor OBP-801/YM753 and a PI3K inhibitor LY294002 synergistically induces apoptosis in human endometrial carcinoma cells due to increase of Bim with accumulation of ROS. Gynecol Oncol 129, 425-432 https://doi.org/10.1016/j.ygyno.2013.02.008
  59. Monge M, Colas E, Doll A et al (2009) Proteomic approach to ETV5 during endometrial carcinoma invasion reveals a link to oxidative stress. Carcinogenesis 30, 1288-1297 https://doi.org/10.1093/carcin/bgp119