521
Views
1
CrossRef citations to date
0
Altmetric
REVIEW

Review of the Potential Therapeutic Effects and Molecular Mechanisms of Resveratrol on Endometriosis

, , , , , , , & show all
Pages 741-763 | Received 13 Jan 2023, Accepted 08 Apr 2023, Published online: 12 May 2023

References

  • Andres MP, Arcoverde FVL, Souza CCC, Fernandes LFC, Abrão MS, Kho RM. Extrapelvic endometriosis: a systematic review. J Minim Invasive Gynecol. 2020;27(2):373–389. doi:10.1016/j.jmig.2019.10.004
  • Zondervan KT, Becker CM, Missmer SA, Longo DL. Endometriosis. N Engl J Med. 2020;382(13):1244–1256. doi:10.1056/NEJMra1810764
  • Garcia-Fernandez J, García-Velasco JA. Endometriosis and reproduction: what we have learned. Yale J Biol Med. 2020;93(4):571–577.
  • Prosperi Porta R, Sangiuliano C, Cavalli A, et al. Effects of breastfeeding on endometriosis-related pain: a prospective observational study. Int J Environ Res Public Health. 2021;18(20):10602. doi:10.3390/ijerph182010602
  • Armour M, Lawson K, Wood A, Smith CA, Abbott J, Thumbikat P. The cost of illness and economic burden of endometriosis and chronic pelvic pain in Australia: a national online survey. PLoS One. 2019;14(10):e0223316. doi:10.1371/journal.pone.0223316
  • Halici BNA, Aktoz F, Kabakci M, Kiran G, Ozcan P. Analysis of preoperative and postoperative quality of life, sexual function, and sleep in patients with endometriosis: a prospective cohort study. Arch Gynecol Obstet. 2022;307(1):113–120. doi:10.1007/s00404-022-06562-9
  • Kim HS, Kim TH, Chung HH, Song YS. Risk and prognosis of ovarian cancer in women with endometriosis: a meta-analysis. Br J Cancer. 2014;110(7):1878–1890. doi:10.1038/bjc.2014.29
  • Kajiyama H, Suzuki S, Yoshihara M, et al. Endometriosis and cancer. Free Radic Biol Med. 2019;133:186–192. doi:10.1016/j.freeradbiomed.2018.12.015
  • Kalaitzopoulos DR, Samartzis N, Kolovos GN, et al. Treatment of endometriosis: a review with comparison of 8 guidelines. BMC Womens Health. 2021;21(1):397. doi:10.1186/s12905-021-01545-5
  • Qin Z, Dong Z, Liu J, et al. A preliminary study on the effects of black cohosh preparations on bone metabolism of rat models with GnRH-a-induced peri-menopausal symptoms. Front Endocrinol. 2022;13:854345. doi:10.3389/fendo.2022.854345
  • Chen J, Wang H, Dong Z, et al. GnRH-a-induced perimenopausal rat modeling and black cohosh preparations’ effect on rat’s reproductive endocrine. Front Endocrinol. 2021;12:683552. doi:10.3389/fendo.2021.683552
  • Corte LD, Noventa M, Ciebiera M, et al. Phytotherapy in endometriosis: an up-to-date review. J Complement Integr Med. 2020;17(3). doi:10.1515/jcim-2019-0084
  • Zakhari A, Delpero E, McKeown S, Tomlinson G, Bougie O, Murji A. Endometriosis recurrence following post-operative hormonal suppression: a systematic review and meta-analysis. Hum Reprod Update. 2021;27(1):96–107. doi:10.1093/humupd/dmaa033
  • Guo SW. Recurrence of endometriosis and its control. Hum Reprod Update. 2009;15(4):441–461. doi:10.1093/humupd/dmp007
  • Mikus M, Vitale SG, Coric M, et al. State of the art, new treatment strategies, and emerging drugs for non-hormonal treatment of endometriosis: a systematic review of randomized control trials. Gynecol Endocrinol. 2022;38(11):911–917. doi:10.1080/09513590.2022.2133105
  • Malaguarnera L. Influence of resveratrol on the immune response. Nutrients. 2019;11(5):946. doi:10.3390/nu11050946
  • Yuan L, Zhou M, Huang D, et al. Resveratrol inhibits the invasion and metastasis of colon cancer through reversal of epithelial‑ mesenchymal transition via the AKT/GSK‑3β/Snail signaling pathway. Mol Med Rep. 2019;20(3):2783–2795. doi:10.3892/mmr.2019.10528
  • Galiniak S, Aebisher D, Bartusik-Aebisher D. Health benefits of resveratrol administration. Acta Biochim Pol. 2019;66(1):13–21. doi:10.18388/abp.2018_2749
  • Kasiotis KM, Pratsinis H, Kletsas D, Haroutounian SA. Resveratrol and related stilbenes: their anti-aging and anti-angiogenic properties. Food Chem Toxicol. 2013;61:112–120. doi:10.1016/j.fct.2013.03.038
  • Rodríguez-Enríquez S, Pacheco-Velázquez SC, Marín-Hernández Á, et al. Resveratrol inhibits cancer cell proliferation by impairing oxidative phosphorylation and inducing oxidative stress. Toxicol Appl Pharmacol. 2019;370:65–77. doi:10.1016/j.taap.2019.03.008
  • Fu X, Li M, Tang C, Huang Z, Najafi M. Targeting of cancer cell death mechanisms by resveratrol: a review. Apoptosis. 2021;26(11–12):561–573. doi:10.1007/s10495-021-01689-7
  • Burns J, Yokota T, Ashihara H, Lean ME, Crozier A. Plant foods and herbal sources of resveratrol. J Agric Food Chem. 2002;50(11):3337–3340. doi:10.1021/jf0112973
  • Sales JM, Resurreccion AV. Resveratrol in peanuts. Crit Rev Food Sci Nutr. 2014;54(6):734–770. doi:10.1080/10408398.2011.606928
  • Lyons MM, Yu C, Toma RB, et al. Resveratrol in raw and baked blueberries and bilberries. J Agric Food Chem. 2003;51(20):5867–5870. doi:10.1021/jf034150f
  • Sanders TH, McMichael RW Jr, Hendrix KW. Occurrence of resveratrol in edible peanuts. J Agric Food Chem. 2000;48(4):1243–1246. doi:10.1021/jf990737b
  • Shi J, Zeng Q, Liu Y, Pan Z. Alternaria sp. MG1, a resveratrol-producing fungus: isolation, identification, and optimal cultivation conditions for resveratrol production. Appl Microbiol Biotechnol. 2012;95(2):369–379. doi:10.1007/s00253-012-4045-9
  • Chen X, He H, Wang G, et al. Stereospecific determination of cis- and trans-resveratrol in rat plasma by HPLC: application to pharmacokinetic studies. Biomed Chromatogr. 2007;21(3):257–265. doi:10.1002/bmc.747
  • Camont L, Cottart CH, Rhayem Y, et al. Simple spectrophotometric assessment of the trans-/cis-resveratrol ratio in aqueous solutions. Anal Chim Acta. 2009;634(1):121–128. doi:10.1016/j.aca.2008.12.003
  • Salehi B, Mishra AP, Nigam M, et al. Resveratrol: a double-edged sword in health benefits. Biomedicines. 2018;6(3). doi:10.3390/biomedicines6030091
  • Caruso F, Tanski J, Villegas-Estrada A, Rossi M. Structural basis for antioxidant activity of trans-resveratrol: ab initio calculations and crystal and molecular structure. J Agric Food Chem. 2004;52(24):7279–7285. doi:10.1021/jf048794e
  • Ovesná Z, Horváthová-Kozics K. Structure-activity relationship of trans-resveratrol and its analogues. Neoplasma. 2005;52(6):450–455.
  • Carter LG, D’Orazio JA, Pearson KJ. Resveratrol and cancer: focus on in vivo evidence. Endocr Relat Cancer. 2014;21(3):R209–R225. doi:10.1530/erc-13-0171
  • Zaffaroni N, Beretta GL. Resveratrol and prostate cancer: the power of phytochemicals. Curr Med Chem. 2021;28(24):4845–4862. doi:10.2174/0929867328666201228124038
  • Vernousfaderani EK, Akhtari N, Rezaei S, et al. Resveratrol and colorectal cancer: a molecular approach to clinical researches. Curr Top Med Chem. 2021;21(29):2634–2646. doi:10.2174/1568026621666211105093658
  • Sun X, Fu P, Xie L, et al. Resveratrol inhibits the progression of cervical cancer by suppressing the transcription and expression of HPV E6 and E7 genes. Int J Mol Med. 2021;47(1):335–345. doi:10.3892/ijmm.2020.4789
  • Wu JM, Hsieh TC. Resveratrol: a cardioprotective substance. Ann N Y Acad Sci. 2011;1215:16–21. doi:10.1111/j.1749-6632.2010.05854.x
  • Tan CS, Loh YC, Tew WY, Yam MF. Vasorelaxant effect of 3,5,4’-trihydroxy-trans-stilbene (resveratrol) and its underlying mechanism. Inflammopharmacology. 2020;28(4):869–875. doi:10.1007/s10787-019-00682-6
  • Rocha-González HI, Ambriz-Tututi M, Granados-Soto V. Resveratrol: a natural compound with pharmacological potential in neurodegenerative diseases. CNS Neurosci Ther. 2008;14(3):234–247. doi:10.1111/j.1755-5949.2008.00045.x
  • Amaya SC, Savaris RF, Filipovic CJ, et al. Resveratrol and endometrium: a closer look at an active ingredient of red wine using in vivo and in vitro models. Reprod Sci. 2014;21(11):1362–1369. doi:10.1177/1933719114525271
  • Saliba C, Jaafoury H, El Hajj M, Nicolas G, Haidar Ahmad H. Abdominal wall endometriosis: a case report. Cureus. 2019;11(2):e4061. doi:10.7759/cureus.4061
  • Janša V, Klančič T, Pušić M, et al. Proteomic analysis of peritoneal fluid identified COMP and TGFBI as new candidate biomarkers for endometriosis. Sci Rep. 2021;11(1):20870. doi:10.1038/s41598-021-00299-2
  • Jiang L, Wan Y, Feng Z, et al. Long noncoding RNA UCA1 is related to autophagy and apoptosis in endometrial stromal cells. Front Oncol. 2020;10:618472. doi:10.3389/fonc.2020.618472
  • Azam INA, Wahab NA, Mokhtar MH, Shafiee MN, Mokhtar NM. Roles of microRNAs in regulating apoptosis in the pathogenesis of endometriosis. Life. 2022;12(9):1321. doi:10.3390/life12091321
  • Yerlikaya G, Balendran S, Pröstling K, et al. Comprehensive study of angiogenic factors in women with endometriosis compared to women without endometriosis. Eur J Obstet Gynecol Reprod Biol. 2016;204:88–98. doi:10.1016/j.ejogrb.2016.07.500
  • Ahn SH, Monsanto SP, Miller C, Singh SS, Thomas R, Tayade C. Pathophysiology and immune dysfunction in endometriosis. Biomed Res Int. 2015;2015:795976. doi:10.1155/2015/795976
  • Filip L, Duică F, Prădatu A, et al. Endometriosis associated infertility: a critical review and analysis on etiopathogenesis and therapeutic approaches. Medicina. 2020;56(9):460. doi:10.3390/medicina56090460
  • Clower L, Fleshman T, Geldenhuys WJ, Santanam N. Targeting oxidative stress involved in endometriosis and its pain. Biomolecules. 2022;12(8):1055. doi:10.3390/biom12081055
  • Till SR, Nakamura R, Schrepf A, As-Sanie S. Approach to diagnosis and management of chronic pelvic pain in women: incorporating chronic overlapping pain conditions in assessment and management. Obstet Gynecol Clin North Am. 2022;49(2):219–239. doi:10.1016/j.ogc.2022.02.006
  • Crain DA, Janssen SJ, Edwards TM, et al. Female reproductive disorders: the roles of endocrine-disrupting compounds and developmental timing. Fertil Steril. 2008;90(4):911–940. doi:10.1016/j.fertnstert.2008.08.067
  • Sophonsritsuk A, Tantanavipas S, Tingthanatikul Y, Lertvikool S, Dittharot K, Waiyaput W. Effects of ethinyl estradiol in combined oral contraceptives on cell proliferation and apoptosis in ectopic endometrial tissue: a randomized controlled study. J Fam Reprod Health. 2021;15(1):45–52. doi:10.18502/jfrh.v15i1.6077
  • Bruner-Tran KL, Osteen KG, Taylor HS, Sokalska A, Haines K, Duleba AJ. Resveratrol inhibits development of experimental endometriosis in vivo and reduces endometrial stromal cell invasiveness in vitro. Biol Reprod. 2011;84(1):106–112. doi:10.1095/biolreprod.110.086744
  • Rudzitis-Auth J, Menger MD, Laschke MW. Resveratrol is a potent inhibitor of vascularization and cell proliferation in experimental endometriosis. Hum Reprod. 2013;28(5):1339–1347. doi:10.1093/humrep/det031
  • Delbandi AA, Mahmoudi M, Shervin A, Heidari S, Kolahdouz-Mohammadi R, Zarnani AH. Evaluation of apoptosis and angiogenesis in ectopic and eutopic stromal cells of patients with endometriosis compared to non-endometriotic controls. BMC Womens Health. 2020;20(1):3. doi:10.1186/s12905-019-0865-4
  • Wei WD, Ruan F, Tu FX, Zhou CY, Lin J. Expression of suppressor of cytokine signaling-3 and caspase-3 in endometriosis and their correlation. Zhonghua Bing Li Xue Za Zhi. 2013;42(8):515–518. doi:10.3760/cma.j.issn.0529-5807.2013.08.003
  • Madanes D, Meresman G, Valla SA, et al. Resveratrol impairs cellular mechanisms associated with the pathogenesis of endometriosis. Reprod Biomed Online. 2022;44(6):976–990. doi:10.1016/j.rbmo.2022.02.008
  • Fan YZ, Zhang JT, Yang HC, Yang YQ. Expression of MMP-2, TIMP-2 protein and the ratio of MMP-2/TIMP-2 in gallbladder carcinoma and their significance. World J Gastroenterol. 2002;8(6):1138–1143. doi:10.3748/wjg.v8.i6.1138
  • Khazaei MR, Rashidi Z, Chobsaz F, Niromand E, Khazaei M. Inhibitory effect of resveratrol on the growth and angiogenesis of human endometrial tissue in an in vitro three-dimensional model of endometriosis. Reprod Biol. 2020;20(4):484–490. doi:10.1016/j.repbio.2020.07.012
  • Kolahdouz-Mohammadi R, Delbandi AA, Khodaverdi S, Arefi S, Arablou T, Shidfar F. The effects of resveratrol treatment on Bcl-2 and bax gene expression in endometriotic compared with non-endometriotic stromal cells. Iran J Public Health. 2020;49(8):1546–1554. doi:10.18502/ijph.v49i8.3900
  • Taguchi A, Koga K, Kawana K, et al. Resveratrol enhances apoptosis in endometriotic stromal cells. Am J Reprod Immunol. 2016;75(4):486–492. doi:10.1111/aji.12489
  • Dull AM, Moga MA, Dimienescu OG, Sechel G, Burtea V, Anastasiu CV. Therapeutic approaches of resveratrol on endometriosis via anti-inflammatory and anti-angiogenic pathways. Molecules. 2019;24(4):667. doi:10.3390/molecules24040667
  • Burney RO, Giudice LC. Pathogenesis and pathophysiology of endometriosis. Fertil Steril. 2012;98(3):511–519. doi:10.1016/j.fertnstert.2012.06.029
  • Thanatsis N, Filindris T, Siampalis A, et al. The effect of novel medical nonhormonal treatments on the angiogenesis of endometriotic lesions. Obstet Gynecol Surv. 2021;76(5):281–291. doi:10.1097/ogx.0000000000000888
  • Rocha ALL, Reis FM, Taylor RN. Angiogenesis and endometriosis. Obstet Gynecol Int. 2013;2013:859619. doi:10.1155/2013/859619
  • Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med. 1995;1(1):27–31. doi:10.1038/nm0195-27
  • Chung AS, Ferrara N. Developmental and pathological angiogenesis. Annu Rev Cell Dev Biol. 2011;27(1):563–584. doi:10.1146/annurev-cellbio-092910-154002
  • McMahon G. VEGF receptor signaling in tumor angiogenesis. oncologist. 2000;5(Suppl 1):3–10. doi:10.1634/theoncologist.5-suppl_1-3
  • Apte RS, Chen DS, Ferrara N. VEGF in signaling and disease: beyond discovery and development. Cell. 2019;176(6):1248–1264. doi:10.1016/j.cell.2019.01.021
  • Mahnke JL, Dawood MY, Huang JC. Vascular endothelial growth factor and interleukin-6 in peritoneal fluid of women with endometriosis. Fertil Steril. 2000;73(1):166–170. doi:10.1016/s0015-0282(99)00466-5
  • Machado DE, Abrao MS, Berardo PT, Takiya CM, Nasciutti LE. Vascular density and distribution of vascular endothelial growth factor (VEGF) and its receptor VEGFR-2 (Flk-1) are significantly higher in patients with deeply infiltrating endometriosis affecting the rectum. Fertil Steril. 2008;90(1):148–155. doi:10.1016/j.fertnstert.2007.05.076
  • Bourlev V, Volkov N, Pavlovitch S, Lets N, Larsson A, Olovsson M. The relationship between microvessel density, proliferative activity and expression of vascular endothelial growth factor-A and its receptors in eutopic endometrium and endometriotic lesions. Reproduction. 2006;132(3):501–509. doi:10.1530/rep.1.01110
  • Potlog-Nahari C, Stratton P, Winkel C, et al. Urine vascular endothelial growth factor-A is not a useful marker for endometriosis. Fertil Steril. 2004;81(6):1507–1512. doi:10.1016/j.fertnstert.2003.10.040
  • Young VJ, Ahmad SF, Duncan WC, Horne AW. The role of TGF-β in the pathophysiology of peritoneal endometriosis. Hum Reprod Update. 2017;23(5):548–559. doi:10.1093/humupd/dmx016
  • Young VJ, Brown JK, Maybin J, Saunders PT, Duncan WC, Horne AW. Transforming growth factor-β induced Warburg-like metabolic reprogramming may underpin the development of peritoneal endometriosis. J Clin Endocrinol Metab. 2014;99(9):3450–3459. doi:10.1210/jc.2014-1026
  • Young VJ, Brown JK, Saunders PT, Duncan WC, Horne AW, Dettman R. The peritoneum is both a source and target of TGF-β in women with endometriosis. PLoS One. 2014;9(9):e106773. doi:10.1371/journal.pone.0106773
  • Ricci AG, Olivares CN, Bilotas MA, et al. Natural therapies assessment for the treatment of endometriosis. Hum Reprod. 2013;28(1):178–188. doi:10.1093/humrep/des369
  • Cenksoy PO, Oktem M, Erdem O, et al. A potential novel treatment strategy: inhibition of angiogenesis and inflammation by resveratrol for regression of endometriosis in an experimental rat model. Gynecol Endocrinol. 2015;31(3):219–224. doi:10.3109/09513590.2014.976197
  • Bayoglu Tekin Y, Guven S, Kirbas A, Kalkan Y, Tumkaya L, Guvendag Guven ES. Is resveratrol a potential substitute for leuprolide acetate in experimental endometriosis? Eur J Obstet Gynecol Reprod Biol. 2015;184:1–6. doi:10.1016/j.ejogrb.2014.10.041
  • Ergenoğlu AM, Yeniel A, Erbaş O, et al. Regression of endometrial implants by resveratrol in an experimentally induced endometriosis model in rats. Reprod Sci. 2013;20(10):1230–1236. doi:10.1177/1933719113483014
  • Bahrami A, Ayen E, Razi M, Behfar M. Effects of atorvastatin and resveratrol against the experimental endometriosis; evidence for glucose and monocarboxylate transporters, neoangiogenesis. Life Sci. 2021;272:119230. doi:10.1016/j.lfs.2021.119230
  • Arablou T, Aryaeian N, Khodaverdi S, et al. The effects of resveratrol on the expression of VEGF, TGF-β, and MMP-9 in endometrial stromal cells of women with endometriosis. Sci Rep. 2021;11(1):6054. doi:10.1038/s41598-021-85512-y
  • Arablou T, Delbandi AA, Khodaverdi S, et al. Resveratrol reduces the expression of insulin-like growth factor-1 and hepatocyte growth factor in stromal cells of women with endometriosis compared with nonendometriotic women. Phytother Res. 2019;33(4):1044–1054. doi:10.1002/ptr.6298
  • Khodarahmian M, Amidi F, Moini A, et al. A randomized exploratory trial to assess the effects of resveratrol on VEGF and TNF-α 2 expression in endometriosis women. J Reprod Immunol. 2021;143(2021):103248. doi:10.1016/j.jri.2020.103248
  • Lukács L, Kovács AR, Pál L, Szűcs S, Lampé R. Evaluating the phagocytic index of peripheral leukocytes in endometriosis by plasma experiments. Medicina. 2022;58(7):925. doi:10.3390/medicina58070925
  • Yuan DP, Gu L, Long J, et al. Shikonin reduces endometriosis by inhibiting RANTES secretion and mononuclear macrophage chemotaxis. Exp Ther Med. 2014;7(3):685–690. doi:10.3892/etm.2013.1458
  • Tamai H, Kinugasa M, Nishio M, Miyake M. Peritoneal inclusion cysts treated with a levonorgestrel-releasing intrauterine system: a case report. Case Rep Womens Health. 2019;22:e00113. doi:10.1016/j.fertnstert.2008.08.067
  • Kolahdouz-Mohammadi R, Shidfar F, Khodaverdi S, et al. Resveratrol treatment reduces expression of MCP-1, IL-6, IL-8 and RANTES in endometriotic stromal cells. J Cell Mol Med. 2021;25(2):1116–1127. doi:10.1111/jcmm.16178
  • Chopyak VV, Koval HD, Havrylyuk AM, Lishchuk-Yakymovych KA, Potomkina HA, Kurpisz MK. Immunopathogenesis of endometriosis - a novel look at an old problem. Cent Eur J Immunol. 2022;47(1):109–116. doi:10.5114/ceji.2022.113830
  • Liu Y, Wang J, Zhang X. An update on the multifaceted role of NF-kappaB in endometriosis. Int J Biol Sci. 2022;18(11):4400–4413. doi:10.7150/ijbs.72707
  • Li MZ, Wu YH, Ali M, Wu XQ, Nie MF. Endometrial stromal cells treated by tumor necrosis factor-α stimulate macrophages polarized toward M2 via interleukin-6 and monocyte chemoattractant protein-1. J Obstet Gynaecol Res. 2020;46(2):293–301. doi:10.1111/jog.14135
  • Bacci M, Capobianco A, Monno A, et al. Macrophages are alternatively activated in patients with endometriosis and required for growth and vascularization of lesions in a mouse model of disease. Am J Pathol. 2009;175(2):547–556. doi:10.2353/ajpath.2009.081011
  • Wang C, Chen Z, Zhao X, et al. Transcriptome-based analysis reveals therapeutic effects of resveratrol on endometriosis in arat model. Drug Des Devel Ther. 2021;15:4141–4155. doi:10.2147/DDDT.S323790
  • Singh S, Anshita D, Ravichandiran V. MCP-1: function, regulation, and involvement in disease. Int Immunopharmacol. 2021;101(PtB):107598. doi:10.1016/j.intimp.2021.107598
  • Borrelli GM, Abrão MS, Mechsner S. Can chemokines be used as biomarkers for endometriosis? A systematic review. Hum Reprod. 2014;29(2):253–266. doi:10.1093/humrep/det401
  • Heidari S, Kolahdouz-Mohammadi R, Khodaverdi S, Tajik N, Delbandi AA. Expression levels of MCP-1, HGF, and IGF-1 in endometriotic patients compared with non-endometriotic controls. BMC Womens Health. 2021;21(1):422. doi:10.1186/s12905-021-01560-6
  • Kang YJ, Jeung IC, Park A, et al. An increased level of IL-6 suppresses NK cell activity in peritoneal fluid of patients with endometriosis via regulation of SHP-2 expression. Hum Reprod. 2014;29(10):2176–2189. doi:10.1093/humrep/deu172
  • Song Y, Su RW, Joshi NR, et al. Interleukin-6 (IL-6) activates the NOTCH1 signaling pathway through E-proteins in endometriotic lesions. J Clin Endocrinol Metab. 2020;105(5):1316–1326. doi:10.1210/clinem/dgaa096
  • Piva M, Horowitz GM, Sharpe-Timms KL. Interleukin-6 differentially stimulates haptoglobin production by peritoneal and endometriotic cells in vitro: a model for endometrial-peritoneal interaction in endometriosis. J Clin Endocrinol Metab. 2001;86(6):2553–2561. doi:10.1210/jcem.86.6.7613
  • Carmona F, Chapron C, Martínez-Zamora M, et al. Ovarian endometrioma but not deep infiltrating endometriosis is associated with increased serum levels of interleukin-8 and interleukin-6. J Reprod Immunol. 2012;95(1–2):80–86. doi:10.1016/j.jri.2012.06.001
  • Li C, Zhao HL, Li YJ, et al. The expression and significance of leukemia inhibitory factor, interleukin-6 and vascular endothelial growth factor in Chinese patients with endometriosis. Arch Gynecol Obstet. 2021;304(1):163–170. doi:10.1007/s00404-021-05980-5
  • Delbandi AA, Mahmoudi M, Shervin A, et al. Eutopic and ectopic stromal cells from patients with endometriosis exhibit differential invasive, adhesive, and proliferative behavior. Fertil Steril. 2013;100(3):761–769. doi:10.1016/j.fertnstert.2013.04.041
  • Arici A, Tazuke SI, Attar E, Kliman HJ, Olive DL. Interleukin-8 concentration in peritoneal fluid of patients with endometriosis and modulation of interleukin-8 expression in human mesothelial cells. Mol Hum Reprod. 1996;2(1):40–45. doi:10.1093/molehr/2.1.40
  • Santegoets KC, Wenink MH, van den Berg WB, Radstake TR, Bayry J. Fc gamma receptor IIb on GM-CSF macrophages controls immune complex mediated inhibition of inflammatory signals. PLoS One. 2014;9(10):e110966. doi:10.1371/journal.pone.0110966
  • Zhong YC, Zhou XF, Hou CM, Li WP. Effect of danefukang on symptoms and biomarkers in women with endometriosis. Taiwan J Obstet Gynecol. 2019;58(2):218–222. doi:10.1016/j.tjog.2019.01.009
  • Riccio LDGC, Santulli P, Marcellin L, Abrão MS, Batteux F, Chapron C. Immunology of endometriosis. Best Pract Res Clin Obstet Gynaecol. 2018;50:39–49. doi:10.1016/j.bpobgyn.2018.01.010
  • Kodarahmian M, Amidi F, Moini A, et al. The modulating effects of resveratrol on the expression of MMP-2 and MMP-9 in endometriosis women: a randomized exploratory trial. Gynecol Endocrinol. 2019;35(8):719–726. doi:10.1080/09513590.2019.1576612
  • Chu H, Li H, Guan X, et al. Resveratrol protects late endothelial progenitor cells from TNF-α-induced inflammatory damage by upregulating Krüppel-like factor-2. Mol Med Rep. 2018;17(4):5708–5715. doi:10.3892/mmr.2018.8621
  • Yu H, Pan C, Zhao S, Wang Z, Zhang H, Wu W. Resveratrol inhibits tumor necrosis factor-alpha-mediated matrix metalloproteinase-9 expression and invasion of human hepatocellular carcinoma cells. Biomed Pharmacother. 2008;62(6):366–372. doi:10.1016/j.biopha.2007.09.006
  • Morita Y, Wada-Hiraike O, Yano T, et al. Resveratrol promotes expression of SIRT1 and StAR in rat ovarian granulosa cells: an implicative role of SIRT1 in the ovary. Reprod Biol Endocrinol. 2012;10(1):14. doi:10.1186/1477-7827-10-14
  • Singh UP, Singh NP, Singh B, et al. Resveratrol (trans-3,5,4’-trihydroxystilbene) induces silent mating type information regulation-1 and down-regulates nuclear transcription factor-kappaB activation to abrogate dextran sulfate sodium-induced colitis. J Pharmacol Exp Ther. 2010;332(3):829–839. doi:10.1124/jpet.109.160838
  • Kiernan R, Brès V, Ng RW, et al. Post-activation turn-off of NF-kappa B-dependent transcription is regulated by acetylation of p65. J Biol Chem. 2003;278(4):2758–2766. doi:10.1074/jbc.M209572200
  • Mehri F, Ranjbar A, Shirafkan N, Asl SS, Esfahani M. The protective effect of resveratrol on diazinon-induced oxidative stress and glucose hemostasis disorder in rats’ liver. J Biochem Mol Toxicol. 2022;37(7):e23063. doi:10.1002/jbt.23063
  • Taguchi A, Wada-Hiraike O, Kawana K, et al. Resveratrol suppresses inflammatory responses in endometrial stromal cells derived from endometriosis: a possible role of the sirtuin 1 pathway. J Obstet Gynaecol Res. 2014;40(3):770–778. doi:10.1111/jog.12252
  • Lai ZZ, Yang HL, Ha SY, et al. Cyclooxygenase-2 in endometriosis. Int J Biol Sci. 2019;15(13):2783–2797. doi:10.7150/ijbs.35128
  • Cai X, Liu M, Zhang B, Zhao SJ, Jiang SW. Phytoestrogens for the management of endometriosis: findings and issues. Pharmaceuticals. 2021;14(6). doi:10.3390/ph14060569
  • Nanda A, Thangapandi K, Banerjee P, et al. Cytokines, angiogenesis, and extracellular matrix degradation are augmented by oxidative stress in endometriosis. Ann Lab Med. 2020;40(5):390–397. doi:10.3343/alm.2020.40.5.390
  • Maia JH, Haddad C, Pinheiro N, Casoy J. Advantages of the association of resveratrol with oral contraceptives for management of endometriosis-related pain. Int J Womens Health. 2012;4(1):543–549. doi:10.2147/IJWH.S36825
  • da Silva DM, Gross LA, Neto EPG, Lessey BA, Savaris RF. The use of resveratrol as an adjuvant treatment of pain in endometriosis: a randomized clinical trial. J Endocrinol Soc. 2017;1(4):359–369. doi:10.1210/js.2017-00053
  • Cacciottola L, Donnez J, Dolmans MM. Can endometriosis-related oxidative stress pave the way for new treatment targets? Int J Mol Sci. 2021;22(13):7138. doi:10.3390/ijms22137138
  • Lingappan K. NF-κB in oxidative stress. Curr Opin Toxicol. 2018;7:81–86. doi:10.1016/j.cotox.2017.11.002
  • Van Langendonckt A, Casanas-Roux F, Donnez J. Oxidative stress and peritoneal endometriosis. Fertil Steril. 2002;77(5):861–870. doi:10.1016/S0015-0282(02)02959-X
  • Gupta S, Agarwal A, Krajcir N, Alvarez JG. Role of oxidative stress in endometriosis. Reprod Biomed Online. 2006;13(1):126–134. 2291. doi:10.1016/S1472-6483(10)62026-3
  • Bach Knudsen KE, Lærke HN, Hedemann MS, et al. Impact of diet-modulated butyrate production on intestinal barrier function and inflammation. Nutrients. 2018;10(10):1499. doi:10.3390/nu10101499
  • González-Ramos R, Donnez J, Defrère S, et al. Nuclear factor-kappa B is constitutively activated in peritoneal endometriosis. Mol Hum Reprod. 2007;13(7):503–509. doi:10.1093/molehr/gam033
  • Vallée A, Lecarpentier Y. Curcumin and endometriosis. Int J Mol Sci. 2020;21(7):2440. doi:10.3390/ijms21072440
  • Qiu XM, Lai ZZ, Ha SY, et al. IL-2 and IL-27 synergistically promote growth and invasion of endometriotic stromal cells by maintaining the balance of IFN-γ and IL-10 in endometriosis. Reproduction. 2020;159(3):251–260. doi:10.1530/rep-19-0411
  • Liu P, Pan Q. Butein inhibits oxidative stress injury in rats with chronic heart failure via ERK/Nrf2 signaling. Cardiovasc Ther. 2022;2022:8684014. doi:10.1155/2022/8684014
  • Yavuz S, Aydin N, Celik O, Yilmaz E, Ozerol E, Tanbek K. Resveratrol successfully treats experimental endometriosis through modulation of oxidative stress and lipid peroxidation. J Cancer Res. 2014;10(2):324–329. doi:10.4103/0973-1482.136619
  • Sampson JA. Peritoneal endometriosis due to the menstrual dissemination of endometrial tissue into the peritoneal cavity. Am J Obstet Gynecol. 1927;14:422–469. doi:10.1016/S0002-9378(15)30003-X
  • Jana S, Rudra DS, Paul S, Swarnakar S. Curcumin delays endometriosis development by inhibiting MMP-2 activity. Indian J Biochem Biophys. 2012;49(5):342–348.
  • Vihinen P, Ala-Aho R, Kähäri VM. Matrix metalloproteinases as therapeutic targets in cancer. Curr Cancer Drug Targets. 2005;5(3):203–220. doi:10.2174/1568009053765799
  • Zakiyanov O, Kalousová M, Zima T, Tesař V. Matrix metalloproteinases in renal diseases: a critical appraisal. Kidney Blood Press Res. 2019;44(3):298–330. doi:10.1159/000499876
  • Ke J, Ye J, Li M, Zhu Z. The role of matrix metalloproteinases in endometriosis: a potential target. Biomolecules. 2021;11(11):1739. doi:10.3390/biom11111739
  • Simões G, Pereira T, Caseiro A. Matrix metaloproteinases in vascular pathology. Microvasc Res. 2022;143:104398. doi:10.1016/j.mvr.2022.104398
  • Chen Z, Wang C, Lin C, et al. Lipidomic alterations and PPARα activation induced by resveratrol lead to reduction in lesion size in endometriosis models. Oxid Med Cell Longev. 2021;2021:9979953. doi:10.1155/2021/9979953
  • Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 2014;15(3):178–196. doi:10.1038/nrm3758
  • Li L, Liu J, Xue H, et al. A TGF-β-MTA1-SOX4-EZH2 signaling axis drives epithelial-mesenchymal transition in tumor metastasis. Oncogene. 2020;39(10):2125–2139. doi:10.1038/s41388-019-1132-8
  • Xu X, Kong X, Liu T, et al. Metastasis-associated protein 1, modulated by miR-30c, promotes endometrial cancer progression through AKT/mTOR/4E-BP1 pathway. Gynecol Oncol. 2019;154(1):207–217. doi:10.1016/j.ygyno.2019.04.005
  • Kong X, Xu X, Zhou L, et al. MTA1, a target of resveratrol, promotes epithelial-mesenchymal transition of endometriosis via ZEB2. Mol Ther Methods Clin Dev. 2020;19:295–306. doi:10.1016/j.omtm.2020.09.013
  • Renda G, Gökkaya İ, Şöhretoğlu D. Immunomodulatory properties of triterpenes. Phytochem Rev. 2022;21(2):537–563. doi:10.1007/s11101-021-09785-x
  • Correa FJS, Andres MP, Rocha TP, et al. Invariant natural killer T-cells and their subtypes may play a role in the pathogenesis of endometriosis. Clinics. 2022;77:100032. doi:10.1016/j.clinsp.2022.100032
  • Huhtinen K, Desai R, Ståhle M, et al. Endometrial and endometriotic concentrations of estrone and estradiol are determined by local metabolism rather than circulating levels. J Clin Endocrinol Metab. 2012;97(11):4228–4235. doi:10.1210/jc.2012-1154
  • Chantalat E, Valera MC, Vaysse C, et al. Estrogen receptors and endometriosis. Int J Mol Sci. 2020;21(8):2815. doi:10.3390/ijms21082815
  • Gołąbek A, Kowalska K, Olejnik A. Polyphenols as a diet therapy concept for endometriosis— current opinion and future perspectives. Nutrients. 2021;13(4). doi:10.3390/nu13041347
  • Houshdaran S, Oke AB, Fung JC, Vo KC, Nezhat C, Giudice LC. Steroid hormones regulate genome-wide epigenetic programming and gene transcription in human endometrial cells with marked aberrancies in endometriosis. PLoS Genet. 2020;16(6):e1008601. doi:10.1371/journal.pgen.1008601
  • Chen P, Li B, Ou-Yang L. Role of estrogen receptors in health and disease. Front Endocrinol. 2022;13:839005. doi:10.3389/fendo.2022.839005
  • Xue Q, Lin Z, Cheng YH, et al. Promoter methylation regulates estrogen receptor 2 in human endometrium and endometriosis. Biol Reprod. 2007;77(4):681–687. doi:10.1095/biolreprod.107.061804
  • Han SJ, Jung SY, Wu SP, et al. Estrogen receptor β modulates apoptosis complexes and the inflammasome to drive the pathogenesis of endometriosis. Cell. 2015;163(4):960–974. doi:10.1016/j.cell.2015.10.034
  • Yang KR, Yu HC, Huang CY, et al. Bioprocessed production of resveratrol-enriched rice wine: simultaneous rice wine fermentation, extraction, and transformation of piceid to resveratrol from Polygonum cuspidatum roots. Foods. 2019;8(7):258. doi:10.3390/foods8070258
  • Walle T. Bioavailability of resveratrol. Ann N Y Acad Sci. 2011;1215:9–15. doi:10.1111/j.1749-6632.2010.05842.x
  • Marier JF, Vachon P, Gritsas A, Zhang J, Moreau JP, Ducharme MP. Metabolism and disposition of resveratrol in rats: extent of absorption, glucuronidation, and enterohepatic recirculation evidenced by a linked-rat model. J Pharmacol Exp Ther. 2002;302(1):369–373. doi:10.1124/jpet.102.033340
  • Henry-Vitrac C, Desmoulière A, Girard D, Mérillon JM, Krisa S. Transport, deglycosylation, and metabolism of trans-piceid by small intestinal epithelial cells. Eur J Nutr. 2006;45(7):376–382. doi:10.1007/s00394-006-0609-8
  • Wenzel E, Somoza V. Metabolism and bioavailability of trans-resveratrol. Mol Nutr Food Res. 2005;49(5):472–481. doi:10.1002/mnfr.200500010
  • Singh CK, Ndiaye MA, Ahmad N. Resveratrol and cancer: challenges for clinical translation. Biochim Biophys Acta. 2015;1852(6):1178–1185. doi:10.1016/j.bbadis.2014.11.004
  • Chimento A, De Amicis F, Sirianni R, et al. Progress to improve oral bioavailability and beneficial effects of resveratrol. Int J Mol Sci. 2019;20(6):1381. doi:10.3390/ijms20061381
  • Tsai MJ, Lu IJ, Fu YS, Fang YP, Huang YB, Wu PC. Nanocarriers enhance the transdermal bioavailability of resveratrol: in-vitro and in-vivo study. Colloids Surf B Biointerfaces. 2016;148:650–656. doi:10.1016/j.colsurfb.2016.09.045
  • Lin YC, Hu SC, Huang PH, Lin TC, Yen FL. Electrospun resveratrol-loaded polyvinylpyrrolidone/cyclodextrin nanofibers and their biomedical applications. Pharmaceutics. 2020;12(6):552. doi:10.3390/pharmaceutics12060552
  • Summerlin N, Soo E, Thakur S, Qu Z, Jambhrunkar S, Popat A. Resveratrol nanoformulations: challenges and opportunities. Int J Pharm. 2015;479(2):282–290. doi:10.1016/j.ijpharm.2015.01.003
  • Guo W, Li A, Jia Z, Yuan Y, Dai H, Li H. Transferrin modified PEG-PLA-resveratrol conjugates: in vitro and in vivo studies for glioma. Eur J Pharmacol. 2013;718(1–3):41–47. doi:10.1016/j.ejphar.2013.09.034
  • Edwards JA, Beck M, Riegger C, Bausch J. Safety of resveratrol with examples for high purity, trans-resveratrol, resVida(®). Ann N Y Acad Sci. 2011;1215:131–137. doi:10.1111/j.1749-6632.2010.05855.x
  • Zhong LX, Wu ML, Li H, Liu J, Lin LZ. Efficacy and safety of intraperitoneally administered resveratrol against rat orthotopic ovarian cancers. Cancer Manag Res. 2019;11:6113–6124. doi:10.2147/cmar.S206301
  • Szende B, Tyihák E, Király-Véghely Z. Dose-dependent effect of resveratrol on proliferation and apoptosis in endothelial and tumor cell cultures. Exp Mol Med. 2000;32(2):88–92. doi:10.1038/emm.2000.16
  • Wu H, Chen L, Zhu F, Han X, Sun L, Chen K. The cytotoxicity effect of resveratrol: cell cycle arrest and induced apoptosis of breast cancer 4T1 cells. Toxins. 2019;11(12):731. doi:10.3390/toxins11120731
  • Johnson WD, Morrissey RL, Usborne AL, et al. Subchronic oral toxicity and cardiovascular safety pharmacology studies of resveratrol, a naturally occurring polyphenol with cancer preventive activity. Food Chem Toxicol. 2011;49(12):3319–3327. doi:10.1016/j.fct.2011.08.023
  • Crowell JA, Korytko PJ, Morrissey RL, Booth TD, Levine BS. Resveratrol-associated renal toxicity. Toxicol Sci. 2004;82(2):614–619. doi:10.1093/toxsci/kfh263
  • Liu S, Zhao M, Zhou Y, et al. Resveratrol exerts dose-dependent anti-fibrotic or pro-fibrotic effects in kidneys: a potential risk to individuals with impaired kidney function. Phytomedicine. 2019;57:223–235. doi:10.1016/j.phymed.2018.12.024
  • Shaito A, Posadino AM, Younes N, et al. Potential adverse effects of resveratrol: a literature review. Int J Mol Sci. 2020;21(6):2084. doi:10.3390/ijms21062084
  • Amiot MJ, Romier B, Dao TM, et al. Optimization of trans-resveratrol bioavailability for human therapy. Biochimie. 2013;95(6):1233–1238. doi:10.1016/j.biochi.2013.01.008
  • Howells LM, Berry DP, Elliott PJ, et al. Phase I randomized, double-blind pilot study of micronized resveratrol (SRT501) in patients with hepatic metastases--safety, pharmacokinetics, and pharmacodynamics. Cancer Prev Res. 2011;4(9):1419–1425. doi:10.1158/1940-6207.Capr-11-0148
  • la Porte C, Voduc N, Zhang G, et al. Steady-state pharmacokinetics and tolerability of trans-resveratrol 2000 mg twice daily with food, quercetin and alcohol (ethanol) in healthy human subjects. Clin Pharmacokinet. 2010;49(7):449–454. doi:10.2165/11531820-000000000-00000
  • Almeida L, Vaz-da-Silva M, Falcão A, et al. Pharmacokinetic and safety profile of trans-resveratrol in a rising multiple-dose study in healthy volunteers. Mol Nutr Food Res. 2009;53(Suppl 1):S7–S15. doi:10.1002/mnfr.200800177
  • Boocock DJ, Faust GE, Patel KR, et al. Phase I dose escalation pharmacokinetic study in healthy volunteers of resveratrol, a potential cancer chemopreventive agent. Cancer Epidemiol Biomarkers Prev. 2007;16(6):1246–1252. doi:10.1158/1055-9965.Epi-07-0022
  • Sergides C, Chirilă M, Silvestro L, Pitta D, Pittas A. Bioavailability and safety study of resveratrol 500 mg tablets in healthy male and female volunteers. Exp Ther Med. 2016;11(1):164–170. doi:10.3892/etm.2015.2895
  • Singh AP, Singh R, Verma SS, et al. Health benefits of resveratrol: evidence from clinical studies. Med Res Rev. 2019;39(5):1851–1891. doi:10.1002/med.21565