Biological effects of combined resveratrol and vitamin D3 on ovarian tissue

Francesca Uberti, Vera Morsanuto, Silvio Aprile, Sabrina Ghirlanda, Ian Stoppa, Andrea Cochis, Giorgio Grosa, Lia Rimondini, Claudio Molinari, Francesca Uberti, Vera Morsanuto, Silvio Aprile, Sabrina Ghirlanda, Ian Stoppa, Andrea Cochis, Giorgio Grosa, Lia Rimondini, Claudio Molinari

Abstract

Background: Resveratrol (3,5,4'-trihydroxy-trans-stilbene) is a natural antioxidant polyphenol able to exert a wide range of biological effect on several tissues. Despite its important beneficial properties, it has a low water solubility, which limits its therapeutic applications in humans. Resveratrol also acts as a phytoestrogen that modulates estrogen receptor (ER)-mediated transcription. In addition, it has been shown that ovarian tissues benefit greatly from vitamin D3, which exerts its beneficial effects through VDR receptors. The aim was to evaluate the cooperative effects of resveratrol combined with vitamin D3 on ovarian cells and tissues and some other organs as well. Moreover, the modulation of specific intracellular pathways involving ER and VDR receptors has been studied.

Methods: The experiments were performed both in vitro and in vivo, to analyze cell viability, radical oxygen species production, signal transductions through Western Blot, and resveratrol quantification by HPLC.

Results: Cell viability, radical oxygen species production, and intracellular pathways have been studied on CHO-K1 cells. Also, the relative mechanism activated following oral intake in female Wistar rats as animal model was investigated, evaluating bioavailability, biodistribution and signal transduction in heart, kidney, liver and ovarian tissues. Both in in vitro and in vivo experiments, resveratrol exerts more evident effects when administered in combination with vitD in ovarian cells, showing a common biphasic cooperative effect: The role of vitamin D3 in maintaining and supporting the biological activity of resveratrol has been clearly observed. Moreover, resveratrol plus vitamin D3 blood concentrations showed a biphasic absorption rate.

Conclusions: Such results could be used as a fundamental data for the development of new therapies for gynecological conditions, such as hot-flashes.

Keywords: Bioavailability; Hot flashes; Phytoalexins; Resveratrol; Vitamin D3.

Conflict of interest statement

Authors’ information

Not applicable.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Time-course and dose-response study of CHO-K1 viability measured by MTT test. a time-course of RES 10 μM; b time-course of RES 25 μM; c time-course of RES 50 μM; d time-course of RES 100 μM. Reported data are means ± SD of five independent experiments. * not significant vs control; point without symbol p < 0.05 vs control. RES = resveratrol. The effect of solvent alone is reported as well
Fig. 2
Fig. 2
The cooperative effect of RES and vitD during time on cell viability and ROS production in CHO-K1 cells. a cell viability and b ROS production measured during time-course study in presence of RES and vitD alone and combined. Reported data are means ± SD in (a) and they are expressed as means ± SD(%) in (b) of five independent experiments. * not significant vs control; points without symbol, p < 0.05 vs control. RES = resveratrol 50 μM; vitD = vitamin D3 100 nM; RES + vitD = co-stimulation of RES 50 μM with vitD 100 nM
Fig. 3
Fig. 3
Measure of intracellular concentration of RES alone and combined with vitD in CHO-K1 cells in a time-course study. Reported data are means ± SD of five independent experiments. * not significant vs control; points without symbol, p < 0.05 vs control. RES = resveratrol 50 μM; RES + vitD = co-stimulation of RES 50 μM with vitamin D 100 nM
Fig. 4
Fig. 4
Western Blot, densitometric analysis and protein activation of CHO-K1 cells stimulated with RES and vitD alone and together. In a (ERK/MAPK), b (Akt), c (SOD) activations by ELISA are reported as means ± SD(%) of five independent experiments. In d (ERβ receptor) and e (VDR receptor) Western blot (on the left) and densitometric analysis (on the right) are reported. In the right column the specific densitometric analysis is reported and expressed as means ± SD(%) of five independent experiments. * p < 0.05 vs control; ** p < 0.05 vs RES alone; φp < 0.05 vs vitD; the bars, p < 0.05 between RES + vitD at different times. RES = resveratrol 50 μM; vitD = vitamin D3 100 nM; RES + vitD = co-stimulation of RES 50 μM with vitD 100 nM
Fig. 5
Fig. 5
Bioavailability, vitamin D quantification and ROS production in in vivo experiments. Female rats (n = 48) were treated with RES 0.5 mg alone (n = 24), combined with vitD 0.4 μg (n = 24) and with vitD 0.4 μg alone (n = 18) by gavage. The animals were sacrificed at specific time-points (ranging 2–270 min) and plasma samples were collected. In a RES plasma concentration (μg/L), in b vitD plasma quantification and in c the ROS production are reported. The results are expressed as means ± SD in panel (a) and as means ± SD(%) in panel (b) of 4 independent experiments. All data p < 0.05 vs control. RES = resveratrol 0.5 mg; RES + vitD = preparation composed of RES 0.5 mg and vitD 0.4 μg
Fig. 6
Fig. 6
Western blot, densitometric analysis and protein activity of ovarian tissue obtained from female rats (n = 32) treated with RES alone (n = 16) and combined with vitD (n = 16). In the upper (a) an example of Western Blot taken at different time (ranging 2–30 min) of Cyclin D1, ERβ receptor and VDR receptor is reported. In the downstream the specific densitometric analysis of Cyclin D1 (b), ERβ receptor (c), and VDR receptor (d) is reported and expressed as means ± SD(%) of 4 independent experiments. In e SOD activity by ELISA was reported as means ± SD(%) of 4 independent experiments. * p < 0.05 vs control; ** p < 0.05 vs RES alone; the bars, p < 0.05 between RES + vitD at different times. RES = resveratrol 0.5 mg; RES + vitD = preparation composed of RES 0.5 mg and vitD 0.4 μg
Fig. 7
Fig. 7
Biodistribution of RES (μg/L) in in vivo experiments. Female rats (n = 40) were treated with RES 0.5 mg alone (n = 20) and combined with vitD 0.4 μg (n = 20) by gavage. The animals were sacrificed at specific time-points (ranging 30–270 min) and heart (a), kidney (b) and liver (c) were collected. The results are expressed as means ± SD (μg/L) of 4 independent experiments. * not significant vs control; point without symbol, p < 0.05 vs control. RES = resveratrol 0.5 mg; RES + vitD = preparation composed of RES 0.5 mg and vitD 0.4 μg

References

    1. Bastin J, Djouadi F. Resveratrol and Myopathy. Nutrients. 2016; doi:10.3390/nu8050254.
    1. Waterhouse AL. Wine phenolics. Ann N Y Acad Sci. 2002;957:21–36. doi: 10.1111/j.1749-6632.2002.tb02903.x.
    1. Athar M, Back JH, Tang X, et al. Resveratrol: a review of preclinical studies for human cancer prevention. Toxicol Appl Pharmacol. 2007;224:274–283. doi: 10.1016/j.taap.2006.12.025.
    1. Yu M, Liu H, Shi A, et al. Preparation of resveratrol-enriched and poor allergic protein peanut sprout from ultrasound treated peanut seeds. Ultrason Sonochem. 2016; doi:10.1016/j.ultsonch.2015.08.008.
    1. Harikumar KB, Aggarwal BB. Resveratrol: a multitargeted agent for age-associated chronic diseases. Cell Cycle. 2008;7:1020–1035. doi: 10.4161/cc.7.8.5740.
    1. Wenzel E, Somoza V. Metabolism and bioavailability of trans-resveratrol. Mol Nutr Food Res. 2005;49:472–481. doi: 10.1002/mnfr.200500010.
    1. Sessa M, Balestrieri ML, Ferrari G, et al. Bioavailability of encapsulated resveratrol into nanoemulsion-based delivery systems. Food Chem. 2014; doi:10.1016/j.foodchem.2013.09.088.
    1. Gambini J, Inglés M, Olaso G, et al. Properties of resveratrol: in vitro and in vivo studies about metabolism, bioavailability, and biological effects in animal models and humans. Oxidative Med Cell Longev. 2015; doi:10.1155/2015/837042.
    1. Soleas GJ, Angelini M, Grass L, et al. Absorption of trans-resveratrol in rats. Methods Enzymol. 2001;335:145–154. doi: 10.1016/S0076-6879(01)35239-4.
    1. Walle T, Hsieh F, DeLegge MH, et al. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab Dispos. 2004; doi:10.1124/dmd.104.000885.
    1. Goldberg DM, Yan J, Soleas GJ. Absorption of three wine-related polyphenols in three different matrices by healthy subjects. Clin Biochem. 2003; doi:10.1016/S0009-9120(02)00397-1.
    1. Ramalingam P, Ko YT. Validated LC-MS/MS method for simultaneous quantification of resveratrol levels in mouse plasma and brain and its application to pharmacokinetic and brain distribution studies. J Pharm Biomed Anal. 2016; doi:10.1016/j.jpba.2015.11.026.
    1. Singh G, Pai RS. Recent advances of resveratrol in nanostructured based delivery systems and in the management of HIV/AIDS. J Control Release. 2014; doi:10.1016/j.jconrel.2014.09.002.
    1. Summerlin N, Soo E, Thakur S, et al. Resveratrol nanoformulations: challenges and opportunities. Int J Pharm. 2015; doi:10.1016/j.ijpharm.2015.01.003.
    1. Pandey KB, Rizvi SI. Plant polyphenols as dietary antioxidants in human health and disease. Oxidative Med Cell Longev. 2009;2(5):270–278. doi: 10.4161/oxim.2.5.9498.
    1. Tessitore L, Davit A, Sarotto I, et al. Resveratrol depresses the growth of colorectal aberrant crypt foci by affecting bax and p21CIP expression. Carcinogenesis. 2000;21:1619–1622. doi: 10.1093/carcin/21.8.1619.
    1. Li ZG, Hong T, Shimada Y, et al. Suppression of N-nitrosomethylbenzylamine (NMBA)-induced esophageal tumorigenesis in F344 rats by resveratrol. Carcinogenesis. 2002;23:1531–1536. doi: 10.1093/carcin/23.9.1531.
    1. Hambrock A, de Oliveira Franz CB, Hiller S, et al. Resveratrol binds to the sulfonylurea receptor (SUR) and induces apoptosis in a SUR subtype-specific manner. J Biol Chem. 2007;282:3347–3356. doi: 10.1074/jbc.M608216200.
    1. Bowers JL, Tyulmenkov VV, Jernigan SC, et al. Resveratrol acts as a mixed agonist/antagonist for estrogen receptors alpha and beta. Endocrinology. 2000;141:3657–3667. doi: 10.1210/endo.141.10.7721.
    1. Nwachukwu JC, Srinivasan S, Bruno NE, et al. Resveratrol modulates the inflammatory response via an estrogen receptor-signal integration network. elife. 2014; doi:10.7554/eLife.02057.
    1. Schmitt E, Lehmann L, Metzler M, et al. Hormonal and genotoxic activity of resveratrol. Toxicol Lett. 2002;136:133–142. doi: 10.1016/S0378-4274(02)00290-4.
    1. Klinge CM, Blankenship KA, Risinger KE, et al. Resveratrol and estradiol rapidly activate MAPK signaling through estrogen receptors alpha and beta in endothelial cells. J Biol Chem. 2005;280:7460–7468. doi: 10.1074/jbc.M411565200.
    1. Saleh MC, Connell BJ, Saleh TM. Resveratrol induced neuroprotection is mediated via both estrogen receptor subtypes, ER(α) and ER(β). Neurosci Lett. 2013; doi:10.1016/j.neulet.2013.05.057.
    1. Leo L, Surico D, Deambrogio F, et al. Dati preliminari sull’efficacia del resveratrolo in una nuova formulazione nel trattamento delle hot flushes. Minerva Ginecol. 2015;67:475–483.
    1. Nwachukwu JC, Li W, Pineda-Torra I, et al. Transcriptional regulation of the androgen receptor cofactor androgen receptor trapped clone-27. Mol Endocrinol. 2007; doi:10.1210/me.2007-0094.
    1. Nwachukwu JC, Southern MR, Kiefer JR, et al. Improved crystallographic structures using extensive combinatorial refinement. Structure. 2013; doi:10.1016/j.str.2013.07.025.
    1. Thill M, Fischer D, Kelling K, et al. Expression of vitamin D receptor (VDR), cyclooxygenase-2 (COX-2) and 15-hydroxyprostaglandin dehydrogenase (15-PGDH) in benign and malignant ovarian tissue and 25-hydroxycholecalciferol (25(OH2)D3) and prostaglandin E2 (PGE2) serum level in ovarian cancer patients. J Steroid Biochem Mol Biol. 2010; doi:10.1016/j.jsbmb.2010.03.049.
    1. Uberti F, Morsanuto V, Lattuada D, et al. Protective effects of vitamin D3 on fimbrial cells exposed to catalytic iron damage. J Ovarian Res. 2016; doi:10.1186/s13048-016-0243-x.
    1. Dampf Stone A, Batie SF, Sabir MS, et al. Resveratrol potentiates vitamin D and nuclear receptor signaling. J Cell Biochem. 2015; doi:10.1002/jcb.25070.
    1. Bartik L, Whitfield GK, Kaczmarska M, et al. Curcumin: a novel nutritionally derived ligand of the vitamin D receptor with implications for colon cancer chemoprevention. J Nutr Biochem. 2010;21:1153–1161. doi: 10.1016/j.jnutbio.2009.09.012.
    1. Davis CD, Milner JA. Nutrigenomics, vitamin D and cancer prevention. J Nutrigenet Nutrigenomics. 2011; doi:10.1159/000324175.
    1. Jurutka PW, Whitfield GK, Forster R, et al. Vitamin D: a fountain of youth in gene regulation. In: Gombart AF, et al., editors. Vitamin D:oxidative stress, immunity, and aging. Boca Raton: CRC Press; 2013. pp. 3–35.
    1. Guo C, Sinnott B, Niu B, et al. Synergistic induction of human cathelicidin antimicrobial peptide gene expression by vitamin D and stilbenoids. Mol Nutr Food Res. 2014;58:528–536. doi: 10.1002/mnfr.201300266.
    1. Li H, Xia N, Forstermann U. Cardiovascular effects and molecular targets of resveratrol. Nitric Oxide. 2012;26:102–110. doi: 10.1016/j.niox.2011.12.006.
    1. Hii CS, Ferrante A. The non-genomic actions of vitamin D. Nutrients. 2016; doi:10.3390/nu8030135.
    1. Mallebrera B, Brandolini V, Font G, et al. Cytoprotective effect of resveratrol diastereomers in CHO-K1 cells exposed to beauvericin. Food Chem Toxicol. 2015; doi:10.1016/j.fct.2015.03.028.
    1. Ortega I, Wong DH, Villanueva JA, et al. Effects of resveratrol on growth and function of rat ovarian granulosa cells. Fertil Steril. 2012; doi:10.1016/j.fertnstert.2012.08.004.
    1. Tomé-Carneiro J, Larrosa M, González-Sarrías A, et al. Resveratrol and clinical trials: the crossroad from in vitro studies to human evidence. Curr Pharm Des. 2013;19:6064–6093. doi: 10.2174/13816128113199990407.
    1. Miyashita M, Koga K, Izumi G, et al. Effects of 1,25-Dihydroxy vitamin D3 on endometriosis. J Clin Endocrinol Metab. 2016; doi:10.1210/jc.2016-1515.
    1. Uberti F, Bardelli C, Morsanuto V, et al. Role of vitamin D3 combined to alginates in preventing acid and oxidative injury in cultured gastric epithelial cells. BMC Gastroenterol. 2016;16:127. doi: 10.1186/s12876-016-0543-z.
    1. Uberti F, Lattuada D, Morsanuto V, et al. Vitamin D protects human endothelial cells from oxidative stress through the autophagic and survival pathways. J Clin Endocrinol Metab. 2014; doi:10.1210/jc.2013-2103.
    1. Uberti F, Morsanuto V, Bardelli C, et al. Protective effects of 1α,25-Dihydroxyvitamin D3 on cultured neural cells exposed to catalytic iron. Physiol Rep. 2016; 10.14814/phy2.12769.
    1. Bonnichsen M, Dragsted N, Hansen AK. The welfare impact of gavaging laboratory rats. Anim Welf. 2005;14:223–227.
    1. Turner PV, Vaughn E, Sunohara-Neilson J, et al. Oral gavage in rats: animal welfare evaluation. J Am Assoc Lab Anim Sci. 2012;51:25–30.
    1. Reagan-Shaw S, Nihal M, Ahmad N. Dose translation from animal to human studies revisited. FASEB J. 2008;22:659–661. doi: 10.1096/fj.07-9574LSF.
    1. Kampa M, Nistikaki A, Tsaousis V, et al. A new automated method for the determination of the total antioxidant capacity (TAC) of human plasma, based on the crocin bleaching assay. BMC Clin Pathol. 2002;2:3–18. doi: 10.1186/1472-6890-2-3.
    1. Conte A, Pellegrini S, Tagliazucchi D. Effect of resveratrol and catechin on PC12 tyrosine kinase activities and their synergistic protection from beta-amyloid toxicity. Drugs Exp Clin Res. 2003;29(5–6):243–255.
    1. Li Q, Huyan T, Ye LJ, et al. Concentration-dependent biphasic effects of resveratrol on human natural killer cells in vitro. J Agric Food Chem. 2014; doi:10.1021/jf502950u.
    1. Ortega I, Villanueva JA, Wong DH, et al. Resveratrol potentiates effects of simvastatin on inhibition of rat ovarian theca-interstitial cells steroidogenesis. J Ovarian Res. 2014; doi:10.1186/1757-2215-7-21.
    1. Ortega I, Duleba AJ. Ovarian actions of resveratrol. Ann N Y Acad Sci. 2015; doi:10.1111/nyas.12875.
    1. 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; doi:10.1186/1477-7827-10-14.
    1. Henry LA, Witt DM. Resveratrol: phytoestrogen effects on reproductive physiology and behavior in female rats. Horm Behav. 2002;41:220–228. doi: 10.1006/hbeh.2001.1754.
    1. Fulda S, Debatin KM. Resveratrol modulation of signal transduction in apoptosis and cell survival: a mini-review. Cancer Detect Prev. 2006;30:217–223. doi: 10.1016/j.cdp.2006.03.007.
    1. Brito PM, Mariano A, Almeida LM, et al. Resveratrol affords protection against peroxynitrite-mediated endothelial cell death: a role for intracellular glutathione. Chem Biol Interact. 2006;164:157–166. doi: 10.1016/j.cbi.2006.09.007.
    1. Gong QH, Wang Q, Shi JS, et al. Inhibition of caspases and intracellular free Ca2+ concentrations are involved in resveratrol protection against apoptosis in rat primary neuron cultures. Acta Pharmacol Sin. 2007;28:1724–1730. doi: 10.1111/j.1745-7254.2007.00666.x.
    1. Anekonda TS, Adamus G. Resveratrol prevents antibody-induced apoptotic death of retinal cells through upregulation of Sirt1 and Ku70. BMC Res Notes. 2008;1:122. doi: 10.1186/1756-0500-1-122.
    1. Cao C, Lu S, Kivlin R, et al. SIRT1 confers protection against UVB- and H(2)O(2)-induced cell death via modulation of p53 and JNK in cultured skin keratinocytes. J Cell Mol Med. 2009;13(9B):3632–3643. doi: 10.1111/j.1582-4934.2008.00453.x.
    1. Rubiolo JA, Mithieux G, Vega FV. Resveratrol protects primary rat hepatocytes against oxidative stress damage: activation of the Nrf2 transcription factor and augmented activities of antioxidant enzymes. Eur J Pharmacol. 2008;591:66–72. doi: 10.1016/j.ejphar.2008.06.067.
    1. Shin SM, Cho IJ, Kim SG. Resveratrol protects mitochondria against oxidative stress through AMP-activated protein kinase-mediated glycogen synthase kinase-3beta inhibition downstream of poly(ADP-ribose)polymerase-LKB1 pathway. Mol Pharmacol. 2009;76:884–895. doi: 10.1124/mol.109.058479.
    1. Wong DH, Villanueva JA, Cress AB, et al. Effects of resveratrol on proliferation and apoptosis in rat ovarian theca-interstitial cells. Mol Hum Reprod. 2010;16:251–259. doi: 10.1093/molehr/gaq002.
    1. Donesky BW, Adashi EY. Surgically induced ovulation in the polycystic ovary syndrome: wedge resection revisited in the age of laparoscopy. Fertil Steril. 1995;63:439–463. doi: 10.1016/S0015-0282(16)57408-1.
    1. Duleba AJ, Banaszewska B, Spaczynski RZ, et al. Success of laparoscopic ovarian wedge resection is related to obesity, lipid profile, and insulin levels. Fertil Steril. 2003;79:1008–1014. doi: 10.1016/S0015-0282(02)04848-3.
    1. Meng X, Maliakal P, Lu H, et al. Urinary and plasma levels of resveratrol and quercetin in humans, mice, and rats after ingestion of pure compounds and grape juice. J Agric Food Chem. 2004;52:935–942. doi: 10.1021/jf030582e.
    1. Baber R. The hot flush: symptom of menopause or sign of disease? Climacteric. 2017;20(4):291–292. doi: 10.1080/13697137.2017.1342346.
    1. Lethaby AE, Brown J, Marjoribanks J, et al. Phytoestrogens for vasomotor menopausal symptoms. Cochrane Database Syst Rev. 2007;4(4):CD001395.
    1. Davinelli S, Scapagnini G, Marzatico F, et al. Influence of equol and resveratrol supplementation on health-related quality of life in menopausal women: a randomized, placebo-controlled study. Maturitas. 2017;96:77–83. doi: 10.1016/j.maturitas.2016.11.016.
    1. Baur JA, Pearson KJ, Price NL, et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006; doi:10.1038/nature05354.
    1. Delmas D, Aires V, Limagne E, et al. Transport, stability, and biological activity of resveratrol. Ann N Y Acad Sci. 2011;1215:48–59. doi: 10.1111/j.1749-6632.2010.05871.x.
    1. Cottart CH, Nivet-Antoine V, Laguillier-Morizot C, et al. Resveratrol bioavailability and toxicity in humans. Mol Nutr Food Res. 2010; doi:10.1002/mnfr.200900437.

Source: PubMed

3
Abonneren