Resveratrol Targets a Variety of Oncogenic and Oncosuppressive Signaling for Ovarian Cancer Prevention and Treatment
Abstract
:1. Introduction
2. Introduction of Resveratrol
2.1. Sources
2.2. Chemistry of Resveratrol
2.3. Absorption and Metabolism of Resveratrol
2.4. Bioavailability of Resveratrol
3. Molecular Mechanisms of Resveratrol Related to Ovarian Cancer
3.1. Inhibition of Carcinogen Activation
3.2. Estrogen Effect
3.3. Antioxidant and Pro-Oxidant Effects
3.4. Inhibition of Angiogenesis
3.5. Anti-Inflammatory Effects of Resveratrol
4. Effect of Resveratrol on Prevention and Treatment of Ovarian Cancer
4.1. In Vitro
4.1.1. Anti-Proliferative and Apoptosis Inducing Activity
4.1.2. Modulate Autophagy
4.1.3. Attenuation of Immune-Suppressive Microenvironment via Inhibition of IDO
4.1.4. Sensitization Chemo-Sensitizing Effects
4.1.5. Inhibition of Epithelial Mesenchymal Transition and Metastasis
4.1.6. Impaired Ovarian CSCs
4.2. In Xenograft Models
5. Effects of Resveratrol in Combination with Other Compounds on Ovarian Cancer
6. Analogs of Resveratrol against Ovarian Cancer
7. Conclusions and Future Directions
Author Contributions
Funding
Conflicts of Interest
References
- Duska, L.R.; Kohn, E.C. The new classifications of ovarian, fallopian tube, and primary peritoneal cancer and their clinical implications. Ann. Oncol. 2017, 28, 8–12. [Google Scholar] [CrossRef]
- Fathalla, M.F. Incessant Ovulation-a factor in Ovarian Neoplasia. Lancet 1971, 2, 163. [Google Scholar] [CrossRef]
- Whittemore, A.S.; Harris, R.; Itnyre, J. Characteristics Relating to Ovarian-Cancer Risk-Collaborative Analysis of 12 United-States Case-Control Studies 2. Invasive Epithelial Ovarian Cancers in White Women. Am. J. Epidemiol. 1992, 136, 1184–1203. [Google Scholar] [CrossRef] [PubMed]
- Bookman, M.A.; Brady, M.F.; McGuire, W.P.; Harper, P.G.; Alberts, D.S.; Friedlander, M.; Colombo, N.; Fowler, J.M.; Argenta, P.A.; De Geest, K.; et al. Evaluation of New Platinum-Based Treatment Regimens in Advanced-Stage Ovarian Cancer: A Phase III Trial of the Gynecologic Cancer InterGroup. J. Clin. Oncol. 2009, 27, 1419–1425. [Google Scholar] [CrossRef]
- Ahmed, N.; Stenvers, K.L. Getting to know ovarian cancer ascites: Opportunities for targeted therapy-based translational research. Front. Oncol. 2013, 3, 256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cooke, S.L.; Brenton, J.D. Evolution of platinum resistance in high-grade serous ovarian cancer. Lancet Oncol. 2011, 12, 1169–1174. [Google Scholar] [CrossRef]
- Shapiro, C.L. Highlights of Recent Findings on Quality-of-Life Management for Patients with Cancer and Their Survivors. JAMA Oncol. 2016, 2, 1401–1402. [Google Scholar] [CrossRef]
- Zong, X.Y.; Nephew, K.P. Ovarian Cancer Stem Cells: Role in Metastasis and Opportunity for Therapeutic Targeting. Cancers 2019, 11, 934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thomasset, S.C.; Berry, D.P.; Garcea, G.; Marczylo, T.; Steward, W.P.; Gescher, A.J. Dietary polyphenolic phytochemicals-Promising cancer chemopreventive agents in humans? A review of their clinical properties. Int. J. Cancer 2007, 120, 451–458. [Google Scholar] [CrossRef]
- Rauf, A.; Imran, M.; Butt, M.S.; Nadeem, M.; Peters, D.G.; Mubarak, M.S. Resveratrol as an anti-cancer agent: A review. Crit. Rev. Food Sci. 2018, 58, 1428–1447. [Google Scholar] [CrossRef]
- Zhang, Y.A.-O.; Li, Y.; Sun, C.; Chen, X.; Han, L.; Wang, T.; Liu, J.; Chen, X.; Zhao, D. Effect of Pterostilbene, a Natural Derivative of Resveratrol, in the Treatment of Colorectal Cancer through Top1/Tdp1-Mediated DNA Repair Pathway. Cancers 2021, 13, 4002. [Google Scholar] [CrossRef]
- Yang, L.; Yin, J.; Wu, J.; Qiao, L.A.-O.; Zhao, E.M.; Cai, F.; Ye, H.A.-O. Engineering genetic devices for in vivo control of therapeutic T cell activity triggered by the dietary molecule resveratrol. Proc. Natl. Acad. Sci. USA 2021, 118, e2106612118. [Google Scholar] [CrossRef] [PubMed]
- Ren, B.; Kwah, M.X.; Liu, C.; Ma, Z.; Shanmugam, M.K.; Ding, L.; Xiang, X.; Ho, P.C.; Wang, L.; Ong, P.S.; et al. Resveratrol for cancer therapy: Challenges and future perspectives. Cancer Lett. 2021, 515, 63–72. [Google Scholar] [CrossRef]
- Malaguarnera, L. Influence of Resveratrol on the Immune Response. Nutrients 2019, 11, 946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jeandet, P.; Douillet-Breuil, A.-C.; Bessis, R.; Debord, S.; Sbaghi, M.; Adrian, M. Phytoalexins from the Vitaceae: Biosynthesis, phytoalexin gene expression in transgenic plants, antifungal activity, and metabolism. J. Agric. Food Chem. 2002, 50, 2731–2741. [Google Scholar] [CrossRef] [PubMed]
- Baur, J.A.; Sinclair, D.A. Therapeutic potential of resveratrol: The in vivo evidence. Nat. Rev. Drug Discov. 2006, 5, 493–506. [Google Scholar] [CrossRef]
- Jang, M.S.; Cai, E.N.; Udeani, G.O.; Slowing, K.V.; Thomas, C.F.; Beecher, C.W.W.; Fong, H.H.S.; Farnsworth, N.R.; Kinghorn, A.D.; Mehta, R.G.; et al. Cancer chemopreventive activity of resveratrol, a natural product derived from grapes. Science 1997, 275, 218–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Athar, M.; Back, J.H.; Kopelovich, L.; Bickers, D.R.; Kim, A.L. Multiple molecular targets of resveratrol: Anti-carcinogenic mechanisms. Arch. Biochem. Biophys. 2009, 486, 95–102. [Google Scholar] [CrossRef] [Green Version]
- Chen, X.; He, H.; Wang, G.; Yang, B.; Ren, W.; Ma, L.; Yu, Q. Stereospecific determination of cis- and trans-resveratrol in rat plasma by HPLC: Application to pharmacokinetic studies. Biomed. Chromatogr. 2007, 21, 257–265. [Google Scholar] [CrossRef] [PubMed]
- Camont, L.; Cottart, C.-H.; Rhayem, Y.; Nivet-Antoine, V.; Djelidi, R.; Collin, F.; Beaudeux, J.-L.; Bonnefont-Rousselot, D. Simple spectrophotometric assessment of the trans-/cis-resveratrol ratio in aqueous solutions. Anal. Chim. Acta 2009, 634, 121–128. [Google Scholar] [CrossRef]
- Keylor, M.H.; Matsuura, B.S.; Stephenson, C.R. Chemistry and Biology of Resveratrol-Derived Natural Products. Chem. Rev. 2015, 115, 8976–9027. [Google Scholar] [CrossRef]
- Walle, T. Bioavailability of resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 9–15. [Google Scholar] [CrossRef]
- Fan, P.; Marston, A.; Hay, A.-E.; Hostettmann, K. Rapid separation of three glucosylated resveratrol analogues from the invasive plant Polygonum cuspidatum by high-speed countercurrent chromatography. J. Sep. Sci. 2009, 32, 2979–2984. [Google Scholar] [CrossRef] [PubMed]
- Walle, T.; Hsieh, F.; DeLegge, M.H.; Oatis, J.E.; Walle, U.K. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab. Dispos. 2004, 32, 1377–1382. [Google Scholar] [CrossRef] [Green Version]
- Kaldas, M.I.; Walle, U.K.; Walle, T. Resveratrol transport and metabolism by human intestinal Caco-2 cells. J. Pharm. Pharmacol. 2003, 55, 307–312. [Google Scholar] [CrossRef] [PubMed]
- Patel, K.R.; Brown, V.A.; Jones, D.J.L.; Britton, R.G.; Hemingway, D.; Miller, A.S.; West, K.P.; Booth, T.D.; Perloff, M.; Crowell, J.A.; et al. Clinical pharmacology of resveratrol and its metabolites in colorectal cancer patients. Cancer Res. 2010, 70, 7392–7399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Goldberg, D.M.; Yan, J.; Fau-Soleas, G.J.; Soleas, G.J. Absorption of three wine-related polyphenols in three different matrices by healthy subjects. Clin. Biochem. 2003, 36, 79–87. [Google Scholar] [CrossRef]
- Vitaglione, P.; Sforza, S.; Galaverna, G.; Ghidini, C.; Caporaso, N.; Vescovi, P.P.; Fogliano, V.; Marchelli, R. Bioavailability of trans-resveratrol from red wine in humans. Mol. Nutr. Food Res. 2005, 49, 495–504. [Google Scholar] [CrossRef] [PubMed]
- Bode, L.M.; Bunzel, D.; Huch, M.; Cho, G.-S.; Ruhland, D.; Bunzel, M.; Bub, A.; Franz, C.M.A.P.; Kulling, S.E. In vivo and in vitro metabolism of trans-resveratrol by human gut microbiota. Am. J. Clin. Nutr. 2013, 97, 295–309. [Google Scholar] [CrossRef]
- Timmers, S.; de Ligt, M.; Phielix, E.; van de Weijer, T.; Hansen, J.; Moonen-Kornips, E.; Schaart, G.; Kunz, I.; Hesselink, M.K.; Schrauwen-Hinderling, V.B.; et al. Resveratrol as Add-on Therapy in Subjects with Well-Controlled Type 2 Diabetes: A Randomized Controlled Trial. Diabetes Care 2016, 39, 2211–2217. [Google Scholar] [CrossRef] [Green Version]
- Patel, K.R.; Scott, E.; Brown, V.A.; Gescher, A.J.; Steward, W.P.; Brown, K. Clinical trials of resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 161–169. [Google Scholar] [CrossRef]
- Jannin, B.; Menzel, M.; Berlot, J.-P.; Delmas, D.; Lançon, A.; Latruffe, N. Transport of resveratrol, a cancer chemopreventive agent, to cellular targets: Plasmatic protein binding and cell uptake. Biochem. Pharmacol. 2004, 68, 1113–1118. [Google Scholar] [CrossRef]
- Belguendouz, L.; Frémont, L.; Gozzelino, M.-T. Interaction of transresveratrol with plasma lipoproteins. Biochem. Pharmacol. 1998, 55, 811–816. [Google Scholar] [CrossRef]
- Lamuela-Raventós, R.M.; Covas, I.; Fitó, M.; Marrugat, J.; de La Torre-Boronat, M.C. Detection of dietary antioxidant phenolic compounds in human LDL. Clin. Chem. 1999, 45, 1870–1872. [Google Scholar] [CrossRef] [PubMed]
- Urpí-Sardà, M.; Jáuregui, O.; Lamuela-Raventós, R.M.; Jaeger, W.; Miksits, M.; Covas, M.-I.; Andres-Lacueva, C. Uptake of diet resveratrol into the human low-density lipoprotein. Identification and quantification of resveratrol metabolites by liquid chromatography coupled with tandem mass spectrometry. Anal. Chem. 2005, 77, 3149–3155. [Google Scholar] [CrossRef]
- Urpi-Sarda, M.; Zamora-Ros, R.; Lamuela-Raventos, R.; Cherubini, A.; Jauregui, O.; de la Torre, R.; Covas, M.I.; Estruch, R.; Jaeger, W.; Andres-Lacueva, C.; et al. HPLC-tandem mass spectrometric method to characterize resveratrol metabolism in humans. Clin. Chem. 2007, 53, 292–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maier-Salamon, A.; Böhmdorfer, M.; Riha, J.; Thalhammer, T.; Szekeres, T.; Jaeger, W. Interplay between metabolism and transport of resveratrol. Ann. N. Y. Acad. Sci. 2013, 1290, 98–106. [Google Scholar] [CrossRef]
- Kool, M.; van der Linden, M.; de Haas, M.; Scheffer, G.L.; de Vree, J.M.; Smith, A.J.; Jansen, G.; Peters, G.J.; Ponne, N.; Scheper, R.J.; et al. MRP3, an organic anion transporter able to transport anti-cancer drugs. Proc. Natl. Acad. Sci. USA 1999, 96, 6914–6919. [Google Scholar] [CrossRef] [Green Version]
- Sedláková, I.; Laco, J.; Caltová, K.; Cervinka, M.; Tošner, J.; Rezác, A.; Špacek, J. Clinical significance of the resistance proteins LRP, Pgp, MRP1, MRP3, and MRP5 in epithelial ovarian cancer. Int. J. Gynecol. Cancer 2015, 25, 236–243. [Google Scholar] [CrossRef]
- Cottart, C.H.; Nivet-Antoine, V.; Laguillier-Morizot, C.; Beaudeux, J.-L. Resveratrol bioavailability and toxicity in humans. Mol. Nutr. Food Res. 2010, 54, 7–16. [Google Scholar] [CrossRef] [PubMed]
- Bertelli, A.A.; Giovannini, L.; Stradi, R.; Urien, S.; Tillement, J.P.; Bertelli, A. Kinetics of trans- and cis-resveratrol (3,4′,5-trihydroxystilbene) after red wine oral administration in rats. Int. J. Clin. Pharmacol. Res. 1996, 16, 77–81. [Google Scholar]
- Bertelli, A.A.; Giovannini, L.; Stradi, R.; Bertelli, A.; Tillement, J.P. Plasma, urine and tissue levels of trans- and cis-resveratrol (3,4′,5-trihydroxystilbene) after short-term or prolonged administration of red wine to rats. Int. J. Tissue React. 1996, 18, 67–71. [Google Scholar]
- Guo, L.; Peng, Y.; Yao, J.; Sui, L.; Gu, A.; Wang, J. Anticancer activity and molecular mechanism of resveratrol-bovine serum albumin nanoparticles on subcutaneously implanted human primary ovarian carcinoma cells in nude mice. Cancer Biother. Radiopharm. 2010, 25, 471–477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guo, L.Y.; Peng, Y.; Li, Y.L.; Yao, J.P.; Zhang, G.M.; Chen, J.; Wang, J.; Sui, L.H. Cell death pathway induced by resveratrol-bovine serum albumin nanoparticles in a human ovarian cell line. Oncol. Lett. 2015, 9, 1359–1363. [Google Scholar] [CrossRef] [Green Version]
- Simpson, E.R.; Kilgore, M.W.; Mahendroo, M.S.; Means, G.D.; Corbin, C.J.; Mendelson, C.R. Regulation of human aromatase cytochrome P450 gene expression. J. Steroid Biochem. Mol. Biol. 1992, 43, 923–930. [Google Scholar] [CrossRef]
- Chen, Z.-H.; Hurh, Y.-J.; Na, H.-K.; Kim, J.H.; Chun, Y.-J.; Kim, D.H.; Kang, K.-S.; Cho, M.-H.; Surh, Y.-J. Resveratrol inhibits TCDD-induced expression of CYP1A1 and CYP1B1 and catechol estrogen-mediated oxidative DNA damage in cultured human mammary epithelial cells. Carcinogenesis 2004, 25, 2005–2013. [Google Scholar] [CrossRef]
- Wang, Y.; Lee, K.W.; Chan, F.L.; Chen, S.; Leung, L.K. The red wine polyphenol resveratrol displays bilevel inhibition on aromatase in breast cancer cells. Toxicol. Sci. 2006, 92, 71–77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Issa, R.M.; Lebeau, A.; Grob, T.; Holst, F.; Moch, H.; Terracciano, L.; Choschzick, M.; Sauter, G.; Simon, R. Estrogen receptor gene amplification occurs rarely in ovarian cancer. Mod. Pathol. 2009, 22, 191–196. [Google Scholar] [CrossRef] [PubMed]
- Gehm, B.D.; McAndrews, J.M.; Chien, P.-Y.; Jameson, J.L. Resveratrol, a polyphenolic compound found in grapes and wine, is an agonist for the estrogen receptor. Proc. Natl. Acad. Sci. USA 1997, 94, 14138–14143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bowers, J.L.; Tyulmenkov, V.V.; Jernigan, S.C.; Klinge, C.M. Resveratrol acts as a mixed agonist/antagonist for estrogen receptors alpha and beta. Endocrinology 2000, 141, 3657–3667. [Google Scholar] [CrossRef]
- Abou-Zeid, L.A.; El-Mowafy, A.M. Differential recognition of resveratrol isomers by the human estrogen receptor-alpha: Molecular dynamics evidence for stereoselective ligand binding. Chirality 2004, 16, 190–195. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.; Cao, H.J.; Davis, F.B.; Tang, H.-Y.; Davis, P.J.; Lin, H.-Y. Oestrogen inhibits resveratrol-induced post-translational modification of p53 and apoptosis in breast cancer cells. Br. J. Cancer 2004, 91, 178–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gambini, J.; Ingles, M.; Olaso, G.; Lopez-Grueso, R.; Bonet-Costa, V.; Gimeno-Mallench, L.; Mas-Bargues, C.; Abdelaziz, K.M.; Gomez-Cabrera, M.C.; Vina, J.; et al. Properties of Resveratrol: In Vitro and In Vivo Studies about Metabolism, Bioavailability, and Biological Effects in Animal Models and Humans. Oxid. Med. Cell Longev. 2015, 2015, 837042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kluth, D.; Banning, A.; Paur, I.; Blomhoff, R.; Brigelius-Flohé, R. Modulation of pregnane X receptor- and electrophile responsive element-mediated gene expression by dietary polyphenolic compounds. Free Radic. Biol. Med. 2007, 42, 315–325. [Google Scholar] [CrossRef] [PubMed]
- Belguendouz, L.; Fremont, L.; Linard, A. Resveratrol inhibits metal ion-dependent and independent peroxidation of porcine low-density lipoproteins. Biochem. Pharmacol. 1997, 53, 1347–1355. [Google Scholar] [CrossRef]
- Frankel, E.; Waterhouse, A.; Kinsella, J. Inhibition of human LDL oxidation by resveratrol. Lancet 1993, 341, 1103–1104. [Google Scholar] [CrossRef]
- Han, J.; Suga, K.; Hayashi, K.; Okamoto, Y.; Umakoshi, H. Multi-Level Characterization of the Membrane Properties of Resveratrol-Incorporated Liposomes. J. Phys. Chem. B 2017, 121, 4091–4098. [Google Scholar] [CrossRef] [PubMed]
- Liu, G.-S.; Zhang, Z.-S.; Yang, B.; He, W. Resveratrol attenuates oxidative damage and ameliorates cognitive impairment in the brain of senescence-accelerated mice. Life Sci. 2012, 91, 872–877. [Google Scholar] [CrossRef]
- Yoshida, Y.; Furuta, S.; Niki, E. Effects of metal chelating agents on the oxidation of lipids induced by copper and iron. Biochim. Biophys. Acta 1993, 1210, 81–88. [Google Scholar] [CrossRef]
- Agarwal, K.; Sharma, A.; Talukder, G. Effects of copper on mammalian cell components. Chem. Biol. Interact. 1989, 69, 1–16. [Google Scholar] [CrossRef]
- Alarcón-De-La-Lastra, C.; Villegas, I. Resveratrol as an antioxidant and pro-oxidant agent: Mechanisms and clinical implications. Biochem. Soc. Trans. 2007, 35, 1156–1160. [Google Scholar] [CrossRef] [Green Version]
- Ahmad, A.; Syed, F.A.; Singh, S.; Hadi, S.M. Prooxidant activity of resveratrol in the presence of copper ions: Mutagenicity in plasmid DNA. Toxicol. Lett. 2005, 159, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Liao, D.; Johnson, R.S. Hypoxia: A key regulator of angiogenesis in cancer. Cancer Metastasis Rev. 2007, 26, 281–290. [Google Scholar] [CrossRef] [PubMed]
- Hickey, M.M.; Simon, M.C. Regulation of angiogenesis by hypoxia and hypoxia-inducible factors. Curr. Top. Dev. Biol. 2006, 76, 217–257. [Google Scholar] [CrossRef]
- Cao, Z.; Fang, J.; Xia, C.; Shi, X.; Jiang, B.-H. trans-3,4,5′-trihydroxystibene inhibits hypoxia-inducible factor 1 alpha and vascular endothelial growth factor expression in human ovarian cancer cells. Clin. Cancer Res. 2004, 10, 5253–5263. [Google Scholar] [CrossRef] [Green Version]
- Garvin, S.; Öllinger, K.; Dabrosin, C. Resveratrol induces apoptosis and inhibits angiogenesis in human breast cancer xenografts in vivo. Cancer Lett. 2006, 231, 113–122. [Google Scholar] [CrossRef]
- Grivennikov, S.I.; Greten, F.; Karin, M. Immunity, Inflammation, and Cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef] [Green Version]
- Fernandes, J.V.; Cobucci, R.; Jatobá, C.A.N.; Fernandes, T.; De Azevedo, J.W.V.; De Araújo, J.M.G. The role of the mediators of inflammation in cancer development. Pathol. Oncol. Res. 2015, 21, 527–534. [Google Scholar] [CrossRef]
- Seo, S.S.; Song, Y.S.; Kang, D.-H.; Park, I.A.; Bang, Y.J.; Kang, S.B.; Lee, H.P. Expression of cyclooxygenase-2 in association with clinicopathological prognostic factors and molecular markers in epithelial ovarian cancer. Gynecol. Oncol. 2004, 92, 927–935. [Google Scholar] [CrossRef]
- Ness, R.B.; Cottreau, C. Possible role of ovarian epithelial inflammation in ovarian cancer. J. Natl. Cancer Inst. 1999, 91, 1459–1467. [Google Scholar] [CrossRef] [Green Version]
- Risch, H.A.; Weiss, N.S.; Lyon, J.L.; Daling, J.R.; Liff, J.M. Events of Reproductive Life and the Incidence of Epithelial Ovarian-Cancer. Am. J. Epidemiol. 1983, 117, 128–139. [Google Scholar] [CrossRef]
- Kim, M.K.; Kim, K.; Han, J.Y.; Lim, J.M.; Song, Y.S. Modulation of inflammatory signaling pathways by phytochemicals in ovarian cancer. Genes Nutr. 2011, 6, 109–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schildkraut, J.M.; Moorman, P.G.; Halabi, S.; Calingaert, B.; Marks, J.R.; Berchuck, A. Analgesic drug use and risk of ovarian cancer. Epidemiology 2006, 17, 104–107. [Google Scholar] [CrossRef] [PubMed]
- Dvorakova, M.; Landa, P. Anti-inflammatory activity of natural stilbenoids: A review. Pharmacol. Res. 2017, 124, 126–145. [Google Scholar] [CrossRef] [PubMed]
- Manna, S.K.; Mukhopadhyay, A.; Aggarwal, B.B. Resveratrol suppresses TNF-induced activation of nuclear transcription factors NF-kappa B, activator protein-1, and apoptosis: Potential role of reactive oxygen intermediates and lipid peroxidation. J Immunol. 2000, 164, 6509–6519. [Google Scholar] [CrossRef] [Green Version]
- Tsai, S.H.; Lin-Shiau, S.Y.; Lin, J.K. Suppression of nitric oxide synthase and the down-regulation of the activation of NF kappa B in macrophages by resveratrol. Brit. J. Pharmacol. 1999, 126, 673–680. [Google Scholar] [CrossRef] [Green Version]
- Bast, R.C.; Hennessy, B.; Mills, G.B. The biology of ovarian cancer: New opportunities for translation. Nat. Rev. Cancer 2009, 9, 415–428. [Google Scholar] [CrossRef]
- Chou, C.H.; Wei, L.H.; Huang, Y.J.; Lai, K.P.; Chen, C.A.; Hsieh, C.Y. Up-regulation of interleukin-6 in human ovarian cancer cell via a Gi/PI3K-Akt/NF-kappa B pathway by lysophosphatidic acid, an ovarian cancer-activating factor. Carcinogenesis 2005, 26, 45–52. [Google Scholar] [CrossRef] [Green Version]
- Park, S.Y.; Jeong, K.J.; Lee, J.; Yoon, D.S.; Choi, W.S.; Kim, Y.K.; Han, J.W.; Kim, Y.M.; Kim, B.K.; Lee, H.Y. Hypoxia enhances LPA-induced HIF-1 alpha and VEGF expression: Their inhibition by resveratrol. Cancer Lett. 2007, 258, 63–69. [Google Scholar] [CrossRef]
- Zhang, Y.K.; Yang, S.F.; Yang, Y.; Liu, T. Resveratrol induces immunogenic cell death of human and murine ovarian carcinoma cells. Infect. Agents Cancer 2019, 14, 27. [Google Scholar] [CrossRef] [Green Version]
- Tan, L.J.; Wang, W.M.; He, G.; Kuick, R.D.; Gossner, G.; Kueck, A.S.; Wahl, H.; Opipari, A.W.; Liu, J.R. Resveratrol inhibits ovarian tumor growth in an in vivo mouse model. Cancer-Am. Cancer Soc. 2016, 122, 722–729. [Google Scholar] [CrossRef] [PubMed]
- Nessa, M.U.; Beale, P.; Chan, C.; Yu, J.Q.; Huq, F. Combinations of Resveratrol, Cisplatin and Oxaliplatin Applied to Human Ovarian Cancer Cells. Anticancer Res. 2012, 32, 53–59. [Google Scholar]
- Gwak, H.; Kim, S.; Dhanasekaran, D.N.; Song, Y.S. Resveratrol triggers ER stress-mediated apoptosis by disrupting N-linked glycosylation of proteins in ovarian cancer cells. Cancer Lett. 2016, 371, 347–353. [Google Scholar] [CrossRef] [PubMed]
- Vergara, D.; Simeone, P.; Toraldo, D.; Del Boccio, P.; Vergaro, V.; Leporatti, S.; Pieragostino, D.; Tinelli, A.; De Domenico, S.; Alberti, S.; et al. Resveratrol downregulates Akt/GSK and ERK signalling pathways in OVCAR-3 ovarian cancer cells. Mol. Biosyst. 2012, 8, 1078–1087. [Google Scholar] [CrossRef]
- Zhong, L.X.; Li, H.; Wu, M.L.; Liu, X.Y.; Zhong, M.J.; Chen, X.Y.; Liu, J.; Zhang, Y. Inhibition of STAT3 signaling as critical molecular event in resveratrol-suppressed ovarian cancer cells. J. Ovarian Res. 2015, 8, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Opipari, A.W.; Tan, L.; Boitano, A.E.; Sorenson, D.R.; Aurora, A.; Liu, J.R. Resveratrol-induced autophagocytosis in ovarian cancer cells. Cancer Res. 2004, 64, 696–703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lang, F.F.; Qin, Z.Y.; Li, F.; Zhang, H.L.; Fang, Z.H.; Hao, E.K. Apoptotic Cell Death Induced by Resveratrol Is Partially Mediated by the Autophagy Pathway in Human Ovarian Cancer Cells. PLoS ONE 2015, 10, e0129196. [Google Scholar] [CrossRef] [PubMed]
- Zhong, L.X.; Zhang, Y.; Wu, M.L.; Liu, Y.N.; Zhang, P.; Chen, X.Y.; Kong, Q.Y.; Liu, J.; Li, H. Resveratrol and STAT inhibitor enhance autophagy in ovarian cancer cells. Cell Death Discov. 2016, 2, 15071. [Google Scholar] [CrossRef]
- Zhong, L.X.; Nie, J.H.; Liu, J.; Lin, L.Z. Correlation of ARHI upregulation with growth suppression and STAT3 inactivation in resveratrol-treated ovarian cancer cells. Cancer Biomark. 2018, 21, 787–795. [Google Scholar] [CrossRef]
- Ferraresi, A.; Phadngam, S.; Morani, F.; Galetto, A.; Alabiso, O.; Chiorino, G.; Isidoro, C. Resveratrol inhibits IL-6-induced ovarian cancer cell migration through epigenetic up-regulation of autophagy. Mol. Carcinogen 2017, 56, 1164–1181. [Google Scholar] [CrossRef]
- Ferraresi, A.; Titone, R.; Follo, C.; Castiglioni, A.; Chiorino, G.; Dhanasekaran, D.N.; Isidoro, C. The protein restriction mimetic Resveratrol is an autophagy inducer stronger than amino acid starvation in ovarian cancer cells. Mol. Carcinogen 2017, 56, 2681–2691. [Google Scholar] [CrossRef] [PubMed]
- Engelke, L.H.; Hamacher, A.; Proksch, P.; Kassack, M.U. Ellagic Acid and Resveratrol Prevent the Development of Cisplatin Resistance in the Epithelial Ovarian Cancer Cell Line A2780. J. Cancer 2016, 7, 353–363. [Google Scholar] [CrossRef] [Green Version]
- Mikula-Pietrasik, J.; Sosinska, P.; Ksiazek, K. Resveratrol inhibits ovarian cancer cell adhesion to peritoneal mesothelium in vitro by modulating the production of alpha 5 beta 1 integrins and hyaluronic acid. Gynecol. Oncol. 2014, 134, 624–630. [Google Scholar] [CrossRef] [PubMed]
- Baribeau, S.; Chaudhry, P.; Parent, S.; Asselin, E. Resveratrol Inhibits Cisplatin-Induced Epithelial-to-Mesenchymal Transition in Ovarian Cancer Cell Lines. PLoS ONE 2014, 9, e86987. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seino, M.; Okada, M.; Shibuya, K.; Seino, S.; Suzuki, S.; Takeda, H.; Ohta, T.; Kurachi, H.; Kitanaka, C. Differential Contribution of ROS to Resveratrol-induced Cell Death and Loss of Self-renewal Capacity of Ovarian Cancer Stem Cells. Anticancer Res. 2015, 35, 85–96. [Google Scholar]
- Heiden, M.G.V.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.S.; Kim, M.S. The role of GSK3 in glucose homeostasis and the development of insulin resistance. Diabetes Res. Clin. Pract. 2007, 77, S49–S57. [Google Scholar] [CrossRef]
- Majewska, E.; Szeliga, M. AKT/GSK3 beta Signaling in Glioblastoma. Neurochem. Res. 2017, 42, 918–924. [Google Scholar] [CrossRef] [Green Version]
- AbuJawdeh, G.M.; Faix, J.D.; Niloff, J.; Tognazzi, K.; Manseau, E.; Dvorak, H.F.; Brown, L.F. Strong expression of vascular permeability factor (vascular endothelial growth factor) and its receptors in ovarian borderline and malignant neoplasms. Lab. Invest 1996, 74, 1105–1115. [Google Scholar]
- Darnell, J.E. Transcription factors as targets for cancer therapy. Nat. Rev. Cancer 2002, 2, 740–749. [Google Scholar] [CrossRef] [PubMed]
- Siddiquee, K.; Zhang, S.; Guida, W.C.; Blaskovich, M.A.; Greedy, B.; Lawrence, H.R.; Yip, M.L.R.; Jove, R.; McLaughlin, M.M.; Lawrence, N.J.; et al. Selective chemical probe inhibitor of Stat3, identified through structure-based virtual screening, induces antitumor activity. Proc. Natl. Acad. Sci. USA 2007, 104, 7391–7396. [Google Scholar] [CrossRef] [Green Version]
- Kim, B.I.; Kim, J.H.; Sim, D.Y.; Nam, M.; Jung, J.H.; Shim, B.; Lee, J.; Kim, S.H. Inhibition of JAK2/STAT3 and activation of caspase-9/3 are involved in KYS05090S-induced apoptosis in ovarian cancer cells. Int. J. Oncol. 2019, 55, 203–210. [Google Scholar] [CrossRef] [PubMed]
- Kang, R.; Zeh, H.J.; Lotze, M.T.; Tang, D. The Beclin 1 network regulates autophagy and apoptosis. Cell Death Differ. 2011, 18, 571–580. [Google Scholar] [CrossRef]
- Lu, Z.; Bast, R.C. The tumor suppressor gene ARHI (DIRAS3) inhibits ovarian cancer cell migration through multiple mechanisms. Cell Adhes. Migr. 2013, 7, 232–236. [Google Scholar] [CrossRef] [Green Version]
- Muller, A.J.; DuHadaway, J.B.; Donover, P.S.; Sutanto-Ward, E.; Prendergast, G.C. Inhibition of indoleamine 2,3-dioxygenase, an immunoregulatory target of the cancer suppression gene Bin1, potentiates cancer chemotherapy. Nat. Med. 2005, 11, 312–319. [Google Scholar] [CrossRef] [PubMed]
- Robinson, C.M.; Hale, P.T.; Carlin, J.M. NF-kappa B activation contributes to indoleamine dioxygenase transcriptional synergy induced by IFN-gamma and tumor necrosis factor-alpha. Cytokine 2006, 35, 53–61. [Google Scholar] [CrossRef]
- Sharma, M.D.; Hou, D.Y.; Liu, Y.J.; Koni, P.A.; Metz, R.; Chandler, P.; Mellor, A.L.; He, Y.; Munn, D.H. Indoleamine 2,3-dioxygenase controls conversion of Foxp3(+) Tregs to TH17-like cells in tumor-draining lymph nodes. Blood 2009, 113, 6102–6111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Noh, K.T.; Chae, S.H.; Chun, S.H.; Jung, I.D.; Kang, H.K.; Park, Y.M. Resveratrol suppresses tumor progression via the regulation of indoleamine 2,3-dioxygenase. Biochem. Bioph. Res. Commun. 2013, 431, 348–353. [Google Scholar] [CrossRef] [PubMed]
- Cornelison, R.; Llaneza, D.C.; Landen, C.N. Emerging Therapeutics to Overcome Chemoresistance in Epithelial Ovarian Cancer: A Mini-Review. Int. J. Mol. Sci. 2017, 18, 2171. [Google Scholar] [CrossRef] [Green Version]
- Banerjee, S.; Padhye, S.; Azmi, A.; Wang, Z.W.; Philip, P.A.; Kucuk, O.; Sarkar, F.H.; Mohammad, R.M. Review on Molecular and Therapeutic Potential of Thymoquinone in Cancer. Nutr. Cancer 2010, 62, 938–946. [Google Scholar] [CrossRef] [PubMed]
- Tan, D.S.P.; Agarwal, R.; Kaye, S.B. Mechanisms of transcoelomic metastasis in ovarian cancer. Lancet Oncol. 2006, 7, 925–934. [Google Scholar] [CrossRef]
- Van Leeuwen, F.N.; Giepmans, B.N.G.; Van Meeteren, L.A.; Moolenaar, W.H. Lysophosphatidic acid: Mitogen and motility factor. Biochem. Soc. Trans. 2003, 31, 1209–1212. [Google Scholar] [CrossRef] [PubMed]
- Fang, X.J.; Gaudette, D.; Furui, T.; Mao, M.L.; Estrella, V.; Eder, A.; Pustilnik, T.; Sasagawa, T.; Lapushin, R.; Yu, S.X.; et al. Lysophospholipid growth factors in the initiation, progression, metastases, and management of ovarian cancer. Lysophospholipids Eicosanoids Biol. Pathophysiol. 2000, 905, 188–208. [Google Scholar] [CrossRef]
- Nishida, N.; Yano, H.; Komai, K.; Nishida, T.; Kamura, T.; Kojiro, M. Vascular endothelial growth factor C and vascular endothelial growth factor receptor 2 are related closely to the prognosis of patients with ovarian carcinoma. Cancer-Am. Cancer Soc. 2004, 101, 1364–1374. [Google Scholar] [CrossRef]
- Al-Alem, L.; Curry, T.E. Ovarian cancer: Involvement of the matrix metalloproteinases. Reproduction 2015, 150, R55–R64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davidson, B.; Reich, R.; Berner, A.; Givant-Horwitz, V.; Goldberg, I.; Risberg, B.; Kristensen, G.B.; Trope, C.G.; Bryne, M.; Kopolovic, J.; et al. Ovarian carcinoma cells in serous effusions show altered MMP-2 and TIMP-2 mRNA levels. Eur. J. Cancer 2001, 37, 2040–2049. [Google Scholar] [CrossRef]
- Wang, L.; Madigan, M.C.; Chen, H.M.; Liu, F.H.; Patterson, K.I.; Beretov, J.; O’Brien, P.M.; Li, Y. Expression of urokinase plasminogen activator and its receptor in advanced epithelial ovarian cancer patients. Gynecol. Oncol. 2009, 114, 265–272. [Google Scholar] [CrossRef]
- Davidson, B.; Givant-Horwitz, V.; Lazarovici, P.; Risberg, B.; Nesland, J.M.; Trope, C.G.; Schaefer, E.; Reich, R. Matrix metalloproteinases (MMP), EMMPRIN (extracellular matrix metalloproteinase inducer) and mitogen-activated protein kinases (MAPK): Co-expression in metastatic serous ovarian carcinoma. Clin. Exp. Metastas 2003, 20, 621–631. [Google Scholar] [CrossRef]
- Wang, F.Q.; Fisher, J.; Fishman, D.A. MMP-1-PAR1 axis mediates LPA-induced epithelial ovarian cancer (EOC) invasion. Gynecol. Oncol. 2011, 120, 247–255. [Google Scholar] [CrossRef]
- Kanamori, Y.; Matsushima, M.; Minaguchi, T.; Kobayashi, K.; Sagae, S.; Kudo, R.; Terakawa, N.; Nakamura, Y. Correlation between expression of the Matrix metalloproteinase-1 gene in ovarian cancers and an insertion/deletion polymorphism in its promoter region. Cancer Res. 1999, 59, 4225–4227. [Google Scholar]
- Six, L.; Grimm, C.; Leodolter, S.; Tempfer, C.; Zeillinger, R.; Sliutz, G.; Speiser, P.; Reinthaller, A.; Hefler, L.A. A polymorphism in the matrix metalloproteinase-1 gene promoter is associated with the prognosis of patients with ovarian cancer. Gynecol. Oncol. 2006, 100, 506–510. [Google Scholar] [CrossRef]
- Singh, A.K.; Sharma, N.; Ghosh, M.; Park, Y.H.; Jeong, D.K. Emerging importance of dietary phytochemicals in fight against cancer: Role in targeting cancer stem cells. Crit. Rev. Food Sci. Nutr. 2017, 57, 3449–3463. [Google Scholar] [CrossRef]
- Hu, L.; McArthur, C.; Jaffe, R.B. Ovarian cancer stem-like side-population cells are tumourigenic and chemoresistant. Br. J. Cancer 2010, 102, 1276–1283. [Google Scholar] [CrossRef] [Green Version]
- Lee, M.-H.; Choi, B.Y.; Kundu, J.K.; Shin, Y.K.; Na, H.-K.; Surh, Y.-J. Resveratrol suppresses growth of human ovarian cancer cells in culture and in a murine xenograft model: Eukaryotic elongation factor 1A2 as a potential target. Cancer Res. 2009, 69, 7449–7458. [Google Scholar] [CrossRef] [Green Version]
- Zhong, L.X.; Wu, M.L.; Li, H.; Liu, J.; Lin, L.Z. Efficacy and safety of intraperitoneally administered resveratrol against rat orthotopic ovarian cancers. Cancer Manag. Res. 2019, 11, 6113–6124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tino, A.B.; Chitcholtan, K.; Sykes, P.H.; Garrill, A. Resveratrol and acetyl-resveratrol modulate activity of VEGF and IL-8 in ovarian cancer cell aggregates via attenuation of the NF-kappa B protein. J. Ovarian Res. 2016, 9, 84. [Google Scholar] [CrossRef] [Green Version]
- Raj, M.H.; Elmageed, Z.Y.A.; Zhou, J.; Gaur, R.; Nguyen, L.; A Azam, G.; Braley, P.; Rao, P.N.; Fathi, I.M.; Ouhtit, A. Synergistic action of dietary phyto-antioxidants on survival and proliferation of ovarian cancer cells. Gynecol. Oncol. 2008, 110, 432–438. [Google Scholar] [CrossRef] [Green Version]
- Marimuthu, P.; Kaur, K.; Kandalam, U.; Jasani, V.; Bukhari, N.; Nguyen, M.; Abdul, A.; Pervez, F.F.; Rathinavelu, A. Treatment of ovarian cancer cells with nutlin-3 and resveratrol combination leads to apoptosis via caspase activation. J. Med. Food 2011, 14, 46–52. [Google Scholar] [CrossRef]
- Arzuman, L.; Beale, P.; Yu, J.Q.; Proschogo, N.; Huq, F. Synthesis of a monofunctional platinum compound and its activity alone and in combination with phytochemicals in ovarian tumor models. Anticancer Res. 2014, 34, 7077–7090. [Google Scholar] [PubMed]
- Fatease, A.A.; Shah, V.; Nguyen, D.X.; Cote, B.; LeBlanc, N.; Rao, D.A.; Alani, A.W.G. Chemosensitization and mitigation of Adriamycin-induced cardiotoxicity using combinational polymeric micelles for co-delivery of quercetin/resveratrol and resveratrol/curcumin in ovarian cancer. Nanomedicine 2019, 19, 39–48. [Google Scholar] [CrossRef] [PubMed]
- Wen, W.; Lowe, G.; Roberts, C.M.; Finlay, J.A.-O.; Han, E.S.; Glackin, C.A.; Dellinger, T.H. Pterostilbene Suppresses Ovarian Cancer Growth via Induction of Apoptosis and Blockade of Cell Cycle Progression Involving Inhibition of the STAT3 Pathway. Int. J. Mol. Sci. 2018, 19, 1983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dong, J.; Guo, H.; Chen, Y. Pterostilbene induces apoptosis through caspase activation in ovarian cancer cells. Eur. J. Gynaecol. Oncol. 2016, 37, 342–347. [Google Scholar]
- Pei, H.L.; Mu, D.M.; Zhang, B. Anticancer Activity of Pterostilbene in Human Ovarian Cancer Cell Lines. Med. Sci. Monit. 2017, 23, 3192–3199. [Google Scholar] [CrossRef] [Green Version]
- Chen, C.W.; Li, Y.; Hu, S.; Zhou, W.; Meng, Y.; Li, Z.; Zhang, Y.; Sun, J.; Bo, Z.; DePamphilis, M.L.; et al. DHS (trans-4,4′-dihydroxystilbene) suppresses DNA replication and tumor growth by inhibiting RRM2 (ribonucleotide reductase regulatory subunit M2). Oncogene 2019, 38, 2364–2379. [Google Scholar] [CrossRef]
- Piotrowska, H.; Myszkowski, K.; Ziółkowska, A.; Kulcenty, K.; Wierzchowski, M.; Kaczmarek, M.; Murias, M.; Kwiatkowska-Borowczyk, E.; Jodynis-Liebert, J. Resveratrol analogue 3,4,4′,5-tetramethoxystilbene inhibits growth, arrests cell cycle and induces apoptosis in ovarian SKOV-3 and A-2780 cancer cells. Toxicol. Appl. Pharmacol. 2012, 263, 53–60. [Google Scholar] [CrossRef]
- Piotrowska-Kempisty, H.; Ruciński, M.; Borys, S.; Kucińska, M.; Kaczmarek, M.; Zawierucha, P.; Wierzchowski, M.; Łażewski, D.; Murias, M.; Jodynis-Liebert, J. 3′-hydroxy-3,4,5,4′-tetramethoxystilbene, the metabolite of resveratrol analogue DMU-212, inhibits ovarian cancer cell growth in vitro and in a mice xenograft model. Sci. Rep. 2016, 6, 32627. [Google Scholar] [CrossRef] [Green Version]
- Nowicki, A.; Skupin-Mrugalska, P.A.-O.; Jozkowiak, M.; Wierzchowski, M.; Rucinski, M.A.-O.; Ramlau, P.; Krajka-Kuzniak, V.A.-O.; Jodynis-Liebert, J.; Piotrowska-Kempisty, H. The Effect of 3′-Hydroxy-3,4,5,4′-Tetramethoxy -stilbene, the Metabolite of the Resveratrol Analogue DMU-212, on the Motility and Proliferation of Ovarian Cancer Cells. Int. J. Mol. Sci. 2020, 21, 1100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vergara, D.; De Domenico, S.; Tinelli, A.; Stanca, E.; Del Mercato, L.L.; Giudetti, A.M.; Simeone, P.; Guazzelli, N.; Lessi, M.; Manzini, C.; et al. Anticancer effects of novel resveratrol analogues on human ovarian cancer cells. Mol. Biosyst. 2017, 13, 1131–1141. [Google Scholar] [CrossRef] [Green Version]
- Yahfoufi, N.; Alsadi, N.; Jambi, M.; Matar, C. The Immunomodulatory and Anti-Inflammatory Role of Polyphenols. Nutrients 2018, 10, 1618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ko, J.H.; Sethi, G.; Um, J.Y.; Shanmugam, M.K.; Arfuso, F.; Kumar, A.P.; Bishayee, A.; Ahn, K.S. The Role of Resveratrol in Cancer Therapy. Int. J. Mol. Sci. 2017, 18, 2589. [Google Scholar] [CrossRef] [Green Version]
Structure | Metabolite | Location | Reference |
---|---|---|---|
trans-resveratrol | serum, plasma, urine | [24,28,29] | |
3,4′-O-dihydroxy-trans-stilbenes | urine | [29] | |
dihydroresveratrol | urine, plasma | [29,30] | |
lunularin | urine | [29] | |
trans-resveratrol-3-O-sulfate | plasma, urine | [24,31] | |
trans-resveratrol-4′-O-sulfate | plasma, urine | [24,31] | |
trans-resveratrol-3-O-glucuronide | serum, plasma, urine | [24,28,31] | |
trans-resveratrol-4′-O-glucuronide | serum, plasma, urine | [28,31] | |
trans-resveratrol-3,4′-O-disulfate | plasma | [31] |
Cell Lines | Dose & Time | Anticancer Effects | Mechanisms | Reference |
---|---|---|---|---|
A2780, SKOV3 | 25, 50 µM; 72 h | Inhibited the regrowth of A2780 cells but had no effect on SKOV3 cells | Inhibited glycolytic response | [81] |
A2780, A2780cisR and A2780ZD0473R | Variable; 24 h, 72 h | Decreased the resistance to the platinum drugs | ↓NF-κB | [82] |
PA-1, MDAH2774 and SKOV3 | 50 µM; 24 h, 48 h | Inhibited cell growth | ↑Apoptosis; ↑ER stress; ↓protein glycosylation;↓p-AKT; ↓GSK3β (phosphorylated at S9) | [83] |
OVCAR-3, SKOV3 | >50 µM; 30 min, 24 h, 48 h and 72 h | Inhibited proliferation, decreased drug resistance | ↓cyclin D1; ↓p-AKT; ↓p-GSK3β; ↓p-ERK | [84] |
A2780/CP70, OVCAR-3 | 12.5, 25, 37.5, 50, 75, 100, 150 μm; 6 h, 12 h | Inhibited ovarian cancer progression and angiogenesis | ↓HIF-1α protein;↓VEGF;↓AKT and MAPK phosphorylation | [65] |
OVCAR-3 | 120 μM; 48 h | Caused growth arrest and apoptosis | ↓HES1; ↓ Notch2; ↑E-cadherin; ↓p-STAT3; ↓SVV; ↓c-Myc; ↓Bcl-2 | [85] |
CAOV-3 | 120 μM; 48 h | Increased apoptosis fraction; G1 phase accumulation | ↓HES1; ↓Notch2;↓Wnt2; ↓p-STAT3;↓SVV; ↓c-Myc; ↓Bcl-2 | [85] |
A2780, CaOV3, ES-2, TOV112D, A1947 | 50–200 μM; Variable | Inhibits the proliferation and survival of ovarian carcinoma cells | ↑Cytochrome c;↑Caspase 9 | [86] |
OVCAR-3 and Caov-3 | 100 µM; 48 h | Inhibited cell growth, triggered autophagy | ↑Apoptosis; ↑ROS generation; ↑ATG5; ↑caspase 3; ↑cleavage from LC3-I to LC3-II | [87] |
OVCAR-3 and Caov-3 | 100 µM; 48 h | Distinct growth arrest, increased autophagy | ↑Apoptosis; ↓STAT3; ↓p-STAT3; ↑Beclin-1;↑cleavage from LC3-I to LC3-II | [88] |
OC-CAOV-3 and OVCAR-3 | 100 µM; 24 h, 48 h, 72 h | Reduced proliferation activities, increased autophagy | ↑Apoptosis; ↑ARHI;↓p-STAT3; ↓the metastasis induced by IL-6 | [89] |
NIH-OVCAR3 | 100 µM; 24 h, 48 h, 72 h | Inhibited cell migration | ↑ARHI; ↓p-STAT3;↑Beclin-1; ↓p-AKT; | [90] |
NIH-OVCAR3 | 100 µM; 1 h, 4 h, 24 h | Inhibited protein synthesis and cell growth, induced autophagy. | ↓mTORC1;↓p-AKT; ↑p-AMPK;↓p-4EBP1; ↑p-eIF2α | [91] |
A2780 | 10 µM; 48 h, 72 h, 26 weeks | Decreased proliferation, inhibited cell migration, prevented the development of cisplatin resistance | ↑ROS; ↓p-EGFR; ↓p-ErbB2; ↓p-ErbB3 | [92] |
A2780 and SKOV3 | Variable; 72 h | Inhibited cell adhesion, decreased metastasis. | ↓α5β1 integrin; ↑HA | [93] |
OVCAR-3 and CAOV-3 | Variable; 2 h | Inhibited LPA and hypoxia-induced cell migration | ↓HIF-1α;↓VEGF; ↓p-ERK; ↓p70S6Kinase | [79] |
A2780 and A2780CP | 0–60 µM; 24 h, 48 h, 72 h | Increased cell death; inhibited cell migration | ↓p-ERK; ↓Snail; ↑LC3B-II | [94] |
A2780 CSCs | Variable; 24 h, 48 h | Inhibits the survival of cells; increases the intracellular ROS level | ↑ROS; ↓Sox2; ↓Nanog | [95] |
Animal Models | Dose and Route | Anticancer Effects | Mechanisms | Frequency and Duration | Reference |
---|---|---|---|---|---|
C57BL/C mice were subcutaneously inoculated with ID8 cells | Amount of 50 or 100 mg/kg body weight; IP | Dramatically decreased tumor weight | ↓TGF-β; ↑IFN-γ | Daily; 21 days | [80] |
A fluorescent xenograft mouse model of ovarian cancer | Amount of 160 mg/kg; IP | Decreased tumor volume | Decreased the uptake of glucose | Daily; 14 days | [81] |
Female BALB/c mice injected with PA-1 cells | Amount of 50 or 100 mg/kg body weight; IP | Inhibited the tumor cell proliferation | ↓PCNA; ↓CD31; ↓eEF1A2; ↑caspase-3 | Daily; 4 weeks | [124] |
A rat orthotopic OC model was established using the rat NUTU-19 OC cell line | Amount of 20 mg/kg/day; IP | Effectively inhibited rat orthotopic ovarian cancer growth without affecting normal tissues | ↓STAT3; ↓p-STAT3; ↓serum CA125 levels; ↑ARHI; ↑PIAS3 | Daily; 2 weeks | [125] |
Combination | Cell Lines | Effect | Mechanisms | Reference |
---|---|---|---|---|
Resveratrol + acetyl-resveratrol | SKOV-3 | Inhibited cell growth | ↓VEGF; ↓NF-κB; ↑IL-8 | [126] |
Resveratrol + I3C | SKOV-3 | Resulted in cell cycle arrest; increased apoptosis | ↑tumor suppressor protein p21; ↓Rb; ↓SVV | [127] |
Resveratrol + nutlin-3 +TGF-β | A2780/CP70 | Induced apoptosis | ↑caspase-3; ↑caspase-9; ↑cytochrome c | [128] |
Resveratrol + LH3 | A2780, A2780 cisR and A2780 ZD0473R | Reduced cell death | ↑NF-κB | [129] |
Micellar resveratrol + quercetin + ADR | A2780ADR xenograft model | Reduced tumor growth | Chemosensitization | [130] |
Micellar resveratrol + quercetin + ADR; micellar resveratrol + curcumin + ADR | ES2-Luc xenograft model | Reduced tumor volume | Chemosensitization | [130] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Xu, X.-L.; Deng, S.-L.; Lian, Z.-X.; Yu, K. Resveratrol Targets a Variety of Oncogenic and Oncosuppressive Signaling for Ovarian Cancer Prevention and Treatment. Antioxidants 2021, 10, 1718. https://doi.org/10.3390/antiox10111718
Xu X-L, Deng S-L, Lian Z-X, Yu K. Resveratrol Targets a Variety of Oncogenic and Oncosuppressive Signaling for Ovarian Cancer Prevention and Treatment. Antioxidants. 2021; 10(11):1718. https://doi.org/10.3390/antiox10111718
Chicago/Turabian StyleXu, Xue-Ling, Shou-Long Deng, Zheng-Xing Lian, and Kun Yu. 2021. "Resveratrol Targets a Variety of Oncogenic and Oncosuppressive Signaling for Ovarian Cancer Prevention and Treatment" Antioxidants 10, no. 11: 1718. https://doi.org/10.3390/antiox10111718
APA StyleXu, X. -L., Deng, S. -L., Lian, Z. -X., & Yu, K. (2021). Resveratrol Targets a Variety of Oncogenic and Oncosuppressive Signaling for Ovarian Cancer Prevention and Treatment. Antioxidants, 10(11), 1718. https://doi.org/10.3390/antiox10111718