. , 00 1.1. Propolis, an ancient product that has become fashionable

.. .. Biological,

, Abbreviation: DHT, 5a-dihydrotestosterone

, ABCA1, ATP-binding cassette A1

A. R. , cIAP-2, baculoviral IAP repeat-containing protein3; Bax, Bcl-2associated X protein; CAPE, caffeic acid phenethyl ester

, ERK, extracellular signal-regulated kinases; IL1-b, interleukin 1-b

. Lxrs,

, matrix metalloproteinases; NFkB, nuclear factor kappa-light-chain-enhancer of activated B cells; TLR, toll-like receptor; p70S6K, ribosomal protein S6 kinase beta-1; cTAP-1, MMP

. Tnfa, tumor necrosis factor a; TRAIL, tumor necrosis factor related apoptosis inducing ligand; u-PA, urokinase-type plasminogen activator

X. , X. Inhibitor-of-apoptosis-protein.-references-alencar, S. M. Oldoni, T. L. Castro, M. L. Cabral et al., Chemical composition and biological activity of a new type of Brazilian propolis: red propolis, J. Ethnopharmacol, vol.113, pp.278-283, 2007.

A. M. Alizadeh, H. Afrouzan, N. Dinparast-djadid, A. C. Sawaya, S. Azizian et al., Chemoprotection of MNNGinitiated gastric cancer in rats using Iranian propolis, Arch. Iran. Med, vol.18, issue.1, pp.18-23, 2015.

N. Amini-sarteshnizi, M. Mobini-dehkordi, S. Khosravi-farsani, and H. Teimori, Anticancer activity of ethanolic extract of propolis on AGS cell line, J. HerbMed Pharmacol, pp.29-34, 2015.

V. S. Bankova, S. L. De-castro, and C. Marcucci-maria, Propolis: recent advances in chemistry and plant origin, pp.3-15, 2000.
URL : https://hal.archives-ouvertes.fr/hal-00891696

V. Bankova, M. Popova, S. Bogdanov, and A. Sabatini, Chemical composition of European propolis: expected and unexpected results, Z. Naturforschung C J. Biosci, vol.57, pp.530-533, 2002.

V. Bankova, Chemical diversity of propolis and the problem of standardization, J. Ethnopharmacol, vol.100, issue.1, pp.114-117, 2005.

V. Bankova, Recent trends and important developments in propolis research. Evid.-Based Complement, Altern. Med. ECAM, vol.2, pp.29-32, 2005.

Y. Barlak, O. De-ger, M. Colak, S. C. Karatayl?, A. M. Bozday? et al., Caffeic acid phenethyl ester protects against the dopaminergic neuronal loss induced by 6hydroxydopamine in rats, Proteome Sci, vol.9, pp.86-94, 2011.

B. Bayala, I. H. Bassole, R. Scifo, C. Gnoula, L. Morel et al., Anticancer activity of essential oils and their chemical components-a review, Am. J. Cancer Res, vol.4, pp.591-607, 2014.

A. P. Bazo, M. A. Rodrigues, J. M. Sforcin, J. L. De-camargo, L. R. Ribeiro et al., Protective action of propolis on the rat colon carcinogenesis, Teratog. Carcinog. Mutagen, vol.22, pp.183-194, 2002.

L. Benguedouar, M. Lahouel, S. C. Gangloff, A. Durlach, F. Grange et al., Ethanolic extract of algerian propolis and galangin decreased murine melanoma T, Anticancer Agents Med. Chem, vol.16, pp.1172-1183, 2016.

T. Boam, K. Boutabet, W. Kebsa, M. Alyane, and M. Lahouel, Polyphenolic fraction of Algerian propolis protects rat kidney against acute oxidative stress induced by doxorubicin, Indian J. Nephrol, vol.9, pp.101-106, 2011.

G. A. Burdock, Review of the biological properties and toxicity of bee propolis (propolis), Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc, vol.36, pp.347-363, 1998.

O. G. Celemli, F. Hatjina, L. Charistos, A. Schiesser, and A. Ozkirim, More insight into the chemical composition of Greek propolis; differences and similarities with Turkish propolis, Z. Naturforschung C J. Biosci, vol.68, pp.429-438, 2013.

J. Chen, Y. Long, M. Han, T. Wang, Q. Chen et al., Water-soluble derivative of propolis mitigates scopolamine-induced learning and memory impairment in mice, Pharmacol. Biochem. Behav, vol.90, issue.1, pp.441-446, 2008.

Z. Chen, A. Rasul, C. Zhao, F. M. Millimouno, I. Tsuji et al., Antiproliferative and apoptotic effects of pinocembrin in human prostate cancer cells, Bangladesh J. Pharmacol, pp.255-263, 2013.

R. Christov, B. Trusheva, M. Popova, V. Bankova, and M. Bertrand, Chemical composition of propolis from Canada: its antiradical activity and plant origin, Nat. Prod. Res, vol.20, pp.531-536, 2006.

C. Chuu, H. Lin, M. F. Ciaccio, J. M. Kokontis, R. J. Hause et al., Caffeic acid phenethyl ester suppresses the proliferation of human prostate cancer cells through inhibition of p70S6K and Akt signaling networks, Cancer Prev. Res. Phila. Pa, vol.5, pp.788-797, 2012.

R. Claus, R. Kinscherf, C. Gehrke, G. Bonaterra, P. Basnet et al., Antiapoptotic effects of propolis extract and propol on human macrophages exposed to minimally modified low density lipoprotein, Arzneimittelforschung, vol.50, pp.373-379, 2000.

C. O. Da-silva-frozza, E. Da-silva-brum, A. Alving, S. Moura, J. A. Henriques et al., LC-MS analysis of Hep-2 and Hek-293 cell lines treated with Brazilian red propolis reveals differences in protein expression, J. Pharm. Pharmacol, vol.68, pp.1073-1084, 2016.

N. Zabaiou, Brazilian propolis antileishmanial and immunomodulatory effects. Evid.-Based Complement, Altern. Med. ECAM, vol.2013, p.673058, 2013.

C. C. Da-silveira, M. De, L. M. Fernandes, M. L. Silva, D. A. Luz et al., Brazilian red propolis: phytochemical screening, antioxidant activity and effect against cancer cells, Cellular and clinical pharmacology of the taxanes docetaxel and paclitaxel-a review, vol.15, pp.488-494, 2014.

V. Dimov, N. Ivanovska, V. Bankova, and S. Popov, Immunomodulatory action of propolis: IV. Prophylactic activity against gram-negative infections and adjuvant effect of the water-soluble derivative, Vaccine, vol.10, pp.817-823, 1992.

K. F. Dota, M. E. Consolaro, T. I. Svidzinski, and M. L. Bruschi, Antifungal activity of Brazilian propolis microparticles against yeasts isolated from vulvovaginal candidiasis. Evid.-Based Complement, Altern. Med. ECAM, 2011.

N. Duran, M. Muz, G. Culha, G. Duran, and B. Ozer, GC-MS analysis and antileishmanial activities of two Turkish propolis types, Parasitol. Res, vol.108, pp.95-105, 2011.

S. I. Falcão, M. Vilas-boas, L. M. Estevinho, C. Barros, M. R. Domingues et al., Phenolic characterization of Northeast Portuguese propolis: usual and unusual compounds, Anal. Bioanal. Chem, vol.396, pp.887-897, 2010.

C. V. Fontanilla, Z. Ma, X. Wei, J. Klotsche, L. Zhao et al., Caffeic acid phenethyl ester prevents 1methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurodegeneration, Neuroscience, vol.188, pp.135-141, 2011.

C. V. Fontanilla, X. Wei, L. Zhao, B. Johnstone, R. M. Pascuzzi et al., Caffeic acid phenethyl ester extends survival of a mouse model of amyotrophic lateral sclerosis, Neuroscience, vol.205, pp.185-193, 2012.

C. O. Frozza, C. S. Garcia, G. Gambato, M. D. De-souza, M. Salvador et al., Chemical characterization: antioxidant and cytotoxic activities of Brazilian red propolis, Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc, vol.52, pp.137-142, 2013.

B. Havsteen, Flavonoids, a class of natural products of high pharmacological potency, Biochem. Pharmacol, vol.32, issue.1, pp.1141-1148, 1983.

S. Huang, C. Zhang, K. Wang, G. Q. Li, and F. Hu, Recent advances in the chemical composition of propolis, Mol. Basel Switz, vol.19, pp.19610-19632, 2014.

A. Iio, K. Ohguchi, H. Maruyama, S. Tazawa, Y. Araki et al., Ethanolic extracts of Brazilian red propolis increase ABCA1 expression and promote cholesterol efflux from THP-1 macrophages, Phytomed. Int. J. Phytother. Phytopharm, vol.19, pp.383-388, 2012.

A. Jemal, R. Siegel, E. Ward, Y. Hao, J. Xu et al., Cancer statistics, CA Cancer J. Clin, vol.58, pp.71-96, 2008.

S. Khacha-ananda, K. Tragoolpua, P. Chantawannakul, and Y. Tragoolpua, Antioxidant and anti-cancer cell proliferation activity of propolis extracts from two extraction methods. Asian Pac, J. Cancer Prev. APJCP, vol.14, pp.6991-6995, 2013.

T. Kobayashi, T. Nakata, and T. Kuzumaki, Effect of flavonoids on cell cycle progression in prostate cancer cells, Cancer Lett, vol.176, pp.17-23, 2002.

Y. Komoda, Isolation of flavonoids from Populus nigra as delta 4-3-ketosteroid (5 alpha) reductase inhibitors, Chem. Pharm. Bull. (Tokyo), vol.37, pp.3128-3130, 1989.

R. Kubina, A. Kaba?a-dzik, A. Dziedzic, B. Bielec, R. D. Wojtyczka et al., The ethanol extract of polish propolis exhibits anti-proliferative and/or pro-apoptotic effect on HCT 116 colon cancer and me45 malignant melanoma cells in vitro conditions, Adv. Clin. Exp. Med, vol.24, pp.203-212, 2015.

K. Kunimasa, M. Ahn, T. Kobayashi, R. Eguchi, S. Kumazawa et al., Brazilian propolis suppresses angiogenesis by inducing apoptosis in tube-forming endothelial cells through inactivation of survival signal ERK1/2. Evid.-Based Complement, Altern. Med. ECAM, p.870753, 2011.

M. Lee, Y. H. Kim, W. Park, W. G. Ahn, O. K. Park et al., Novel antidepressant-like activity of propolis extract mediated by enhanced glucocorticoid receptor function in the hippocampus. Evid.-Based Complement, Altern. Med. ECAM, p.217853, 2013.

H. Li, A. Kapur, J. X. Yang, S. Srivastava, D. G. Mcleod et al., Antiproliferation of human prostate cancer cells by ethanolic extracts of Brazilian propolis and its botanical origin, Int. J. Oncol, vol.31, pp.601-606, 2007.

Y. Li, H. Xuan, Q. Shou, Z. Zhan, X. Lu et al., Therapeutic effects of propolis essential oil on anxiety of restraint-stressed mice, Hum. Exp. Toxicol, vol.31, pp.157-165, 2012.

H. Lin, S. S. Jiang, and C. Chuu, Caffeic acid phenethyl ester causes p21 induction Akt signaling reduction, and growth inhibition in PC-3 human prostate cancer cells, PLoS One, vol.7, 2012.

R. Liu, J. Li, J. Song, D. Zhou, C. Huang et al., Pinocembrin improves cognition and protects the neurovascular unit in Alzheimer related deficits, Neurobiol. Aging, vol.35, pp.1275-1285, 2014.

A. A. Lopes, T. S. Ferreira, R. T. Nesi, M. Lanzetti, K. M. Pires et al., Antioxidant action of propolis on mouse lungs exposed to short-term cigarette smoke, Bioorg. Med. Chem, vol.1, pp.7570-7577, 2013.

B. A. Machado, R. P. Silva, A. De, G. Barreto, S. S. Costa et al., Chemical composition and biological activity of extracts obtained by supercritical extraction and ethanolic extraction of brown, green and red propolis derived from different geographic regions in Brazil, PLoS One, vol.11, 2016.

K. Mceleny, R. Coffey, C. Morrissey, J. M. Fitzpatrick, and R. W. Watson, Caffeic acid phenethyl ester-induced PC-3 cell apoptosis is caspase-dependent and mediated through the loss of inhibitors of apoptosis proteins, BJU Int, vol.94, pp.402-406, 2004.

M. G. Miguel, S. Nunes, S. A. Dandlen, A. M. Cavaco, and M. D. Antunes, Phenols and antioxidant activity of hydro-alcoholic extracts of propolis from Algarve: south of Portugal, Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc, vol.48, pp.3418-3423, 2010.

E. M. Muli and J. M. Maingi, Antibacterial activity of Apis mellifera L. propolis collected in three regions of Kenya, J. Venom. Anim. Toxins Trop. Dis, pp.655-663, 2007.

H. K. Nair, K. V. Rao, R. Aalinkeel, S. Mahajan, R. Chawda et al., Inhibition of prostate cancer cell colony formation by the flavonoid quercetin correlates with modulation of specific regulatory genes, Clin. Diagn. Lab. Immunol, vol.11, pp.63-69, 2004.

J. Nam, A. R. Sharma, L. T. Nguyen, C. Chakraborty, G. Sharma et al., The neuroprotective effects of brazilian green propolis on neurodegenerative damage in human neuronal SH-SY5Y cells, Oxid. Med. Cell. Longev, vol.7984327, pp.493-497, 2006.

C. L. Orsatti, F. Missima, A. C. Pagliarone, T. F. Bachiega, M. C. Búfalo et al., Propolis immunomodulatory action in vivo on Toll-like receptors 2 and 4 expression and on pro-inflammatory cytokines production in mice, Phytother. Res. PTR, vol.24, pp.1141-1146, 2010.

N. Orsoli-c, A. H. Knezevi-c, L. Sver, S. Terzi-c, and I. Basi-c, Immunomodulatory and antimetastatic action of propolis and related polyphenolic compounds, J. Ethnopharmacol, vol.94, pp.307-315, 2004.

D. Papachroni, K. Graikou, I. Kosalec, H. Damianakos, V. Ingram et al., Phytochemical analysis and biological evaluation of selected African propolis samples from Cameroon and Congo, Nat. Prod. Commun, vol.10, issue.1, pp.67-70, 2015.

Y. K. Park, S. M. Alencar, and C. L. Aguiar, Botanical origin and chemical composition of Brazilian propolis, J. Agric. Food Chem, vol.50, pp.2502-2506, 2002.

S. Patel, Emerging adjuvant therapy for cancer: propolis and its constituents, J. Diet. Suppl, vol.13, pp.245-268, 2016.

A. Petrova, M. Popova, C. Kuzmanova, I. Tsvetkova, H. Naydenski et al., New biologically active compounds from Kenyan propolis, Fitoterapia, vol.81, pp.509-514, 2010.

A. L. Piccinelli, C. Lotti, L. Campone, O. Cuesta-rubio, M. Campo-fernandez et al., Cuban and Brazilian red propolis: botanical origin and comparative analysis by high-performance liquid chromatography-photodiode array detection/electrospray ionization tandem mass spectrometry, J. Agric. Food Chem, vol.59, pp.6484-6491, 2011.

A. L. Piccinelli, T. Mencherini, R. Celano, Z. Mouhoubi, A. Tamendjari et al., Chemical composition and antioxidant activity of Algerian propolis, J. Agric. Food Chem, vol.61, pp.5080-5088, 2013.

A. J. Pommier, G. Alves, E. Viennois, S. Bernard, Y. Communal et al., Liver X Receptor activation downregulates AKT survival signaling in lipid rafts and induces apoptosis of prostate cancer cells, Oncogene, vol.29, pp.2712-2723, 2010.

A. J. Pommier, J. Dufour, G. Alves, E. Viennois, H. De-boussac et al., Validated methods for the quantification of biologically active constituents of poplar-type propolis, Phytochem. Anal. PCA, vol.9, pp.235-240, 2004.

M. P. Popova, I. B. Chinou, I. N. Marekov, and . Bankova, Terpenes with antimicrobial activity from Cretan propolis, Phytochemistry, vol.1, pp.1262-1271, 2009.

M. P. Popova, K. Graikou, I. Chinou, and V. S. Bankova, GC-MS profiling of diterpene compounds in Mediterranean propolis from Greece, J. Agric. Food Chem, vol.58, pp.3167-3176, 2010.

M. P. Popova, B. Trusheva, D. Antonova, S. Cutajar, D. Mifsud et al., The specific chemical profile of Mediterranean propolis from Malta, pp.1431-1435, 2011.

M. Popova, R. Dimitrova, H. T. Al-lawati, I. Tsvetkova, H. Najdenski et al., Omani propolis: chemical profiling, antibacterial activity and new propolis plant sources, Chem. Cent. J, vol.7, p.158, 2013.

E. N. Quiroga, D. A. Sampietro, J. R. Soberón, M. A. Sgariglia, and M. A. Vattuone, Propolis from the northwest of Argentina as a source of antifungal principles, J. Appl. Microbiol, vol.101, pp.103-110, 2006.

M. M. Rahman, A. Richardson, and M. Sofian-azirun, Antibacterial activity of propolis and honey against Staphylococcus aureus and Escherichia coli, Afr. J. Microbiol. Res, pp.1872-1878, 2010.

J. S. Reis, G. B. Oliveira, M. C. Monteiro, C. S. Machado, Y. R. Torres et al., Antidepressant-and anxiolytic-like activities of an oil extract of propolis in rats, Phytomed. Int. J. Phytother. Phytopharm, vol.21, pp.1466-1472, 2014.

A. A. Righi, T. R. Alves, G. Negri, L. M. Marques, H. Breyer et al., Brazilian red propolis: unreported substances, antioxidant and antimicrobial activities, J. Sci. Food Agric, vol.91, pp.2363-2370, 2011.

S. M. Rizk, H. F. Zaki, and M. A. Mina, Propolis attenuates doxorubicin-induced testicular toxicity in rats, Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc, vol.67, pp.176-186, 2014.

R. S. Rosenberg-zand, D. J. Jenkins, and E. P. Diamandis, Flavonoids and steroid hormone-dependent cancers, J. Chromatogr. B Analyt. Technol. Biomed. Life. Sci, vol.777, pp.219-232, 2002.

A. I. Rushdi, N. Adgaba, N. I. Bayaqoob, A. Al-khazim, B. I. Simoneit et al., Characteristics and chemical compositions of propolis from Ethiopia, vol.3, p.253, 2014.

S. Ryu, W. Lim, F. W. Bazer, and G. Song, Chrysin induces death of prostate cancer cells by inducing ROS and ER stress, J. Cell. Physiol doi, 2017.

E. I. Salim, A. El-magid, A. D. Farara, K. M. Maria, and D. S. , Antitumoral and antioxidant potential of egyptian propolis against the PC3 prostate cancer cell line. Asian Pac, J. Cancer Prev. APJCP, vol.16, pp.7641-7651, 2015.

S. Samarghandian, J. T. Afshari, and S. Davoodi, Chrysin reduces proliferation and induces apoptosis in the human prostate cancer cell line pc-3. Clin. Sao Paulo Braz, vol.66, pp.1073-1079, 2011.

D. A. Sampietro, M. M. Sampietro-vattuone, and M. A. Vattuone, Immunomodulatory activity of Apis mellifera propolis from the North of Argentina, LWT-Food Sci. Technol, pp.9-15, 2016.

J. T. Sanderson, J. Hordijk, M. S. Denison, M. F. Springsteel, M. H. Nantz et al., Induction and inhibition of aromatase (CYP19) activity by natural and synthetic flavonoid compounds in H295R human adrenocortical carcinoma cells, Toxicol. Sci, vol.82, pp.70-79, 2004.

A. C. Sawaya, I. B. Cunha, M. Marcucci, R. F. De-oliveira-rodrigues, and M. N. Eberlin, Brazilian propolis of tetragonisca angustula and apis mellifera, Apidologie, pp.398-407, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00892193

N. Segueni, A. A. Magid, M. Decarme, S. Rhouati, M. Lahouel et al., Inhibition of stromelysin-1 by caffeic acid derivatives from a propolis sample from Algeria, Planta Med, vol.77, pp.999-1004, 2011.

J. M. Sforcin, Propolis and the immune system: a review, J. Ethnopharmacol, vol.113, pp.1-14, 2007.

J. M. Sforcin, Biological properties and therapeutic applications of propolis, Phytother. Res. PTR, vol.30, pp.894-905, 2016.

K. Shen, S. Hung, L. Yin, C. Huang, C. Chao et al., Acacetin a flavonoid, inhibits the invasion and migration of human prostate cancer DU145 cells via inactivation of the p38 MAPK signaling pathway, Mol. Cell. Biochem, vol.333, pp.279-291, 2010.

S. Silici, M. Ünlü, and G. Vardar-Ünlü, Antibacterial activity and phytochemical evidence for the plant origin of Turkish propolis from different regions, World J. Microbiol. Biotechnol, vol.23, pp.1797-1803, 2007.

B. B. Silva, P. L. Rosalen, J. A. Cury, M. Ikegaki, V. C. Souza et al., Chemical composition and botanical origin of red propolis: a new type of brazilian propolis. Evid.-Based Complement, Altern. Med. ECAM, vol.5, pp.313-316, 2008.

R. Silva-carvalho, . Miranda-gonc, V. Salves, A. M. Ferreira, S. M. Cardoso et al., Antitumoural and antiangiogenic activity of Portuguese propolis in in vitro and in vivo models, J. Funct Foods, pp.160-171, 2014.

M. Soda, D. Hu, S. Endo, M. Takemura, J. Li et al., Design: synthesis and evaluation of caffeic acid phenethyl ester-based inhibitors targeting a selectivity pocket in the active site of human aldo-keto reductase 1B10, Eur. J. Med. Chem, vol.48, pp.321-329, 2012.

L. Sun, A. Chen, H. Hung, Y. Chien, J. Huang et al., Chrysin: a histone deacetylase 8 inhibitor with anticancer activity and a suitable candidate for the standardization of Chinese propolis, J. Agric. Food Chem, vol.60, pp.11748-11758, 2012.

E. Szliszka, Z. P. Czuba, J. Bronikowska, A. Mertas, A. Paradysz et al., Ethanolic extract of propolis augments TRAIL-induced apoptotic death in prostate cancer cells. Evid.-Based Complement, Altern. Med. ECAM, vol.38, pp.941-953, 2011.

R. Trivedi and D. P. Mishra, Trailing TRAIL resistance: novel targets for TRAIL sensitization in cancer cells, Front. Oncol, vol.5, 2015.

J. Tseng, C. Lin, L. Su, H. Fu, S. Yang et al., CAPE suppresses migration and invasion of prostate cancer cells via activation of noncanonical Wnt signaling, Oncotarget, vol.7, pp.38010-38024, 2016.

I. Turan, S. Demir, S. Misir, K. Kilinc, A. Mentese et al., Cytotoxic effect of turkish propolis on liver, colon, breast, cervix and prostate cancer cell lines, Trop. J. Pharm. Res, pp.777-782, 2015.

M. J. Valente, A. F. Baltazar, R. Henrique, L. Estevinho, and M. Carvalho, Biological activities of Portuguese propolis: protection against free radical-induced erythrocyte damage and inhibition of human renal cancer cell growth in vitro, Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc, vol.49, pp.86-92, 2011.

V. D. Wagh, Propolis: a wonder bees product and its pharmacological potentials, Adv. Pharmacol. Sci, p.308249, 2013.

Y. Wang, Y. Miao, A. Z. Mir, L. Cheng, L. Wang et al., Inhibition of beta-amyloid-induced neurotoxicity by pinocembrin through Nrf2/HO-1 pathway in SH-SY5Y cells, J. Neurol. Sci, vol.368, pp.223-230, 2016.

M. A. Watanabe, M. K. Amarante, B. J. Conti, and J. M. Sforcin, Cytotoxic constituents of propolis inducing anticancer effects: a review, J. Pharm. Pharmacol, vol.63, pp.1378-1386, 2011.

E. Zhang, R. Wang, S. Guo, and B. Liu, An update on antitumor activity of naturally occurring chalcones. Evid.-Based Complement, Altern. Med. ECAM, p.815621, 2013.

W. Zhang, Y. Lan, Q. Huang, and Z. Hua, Galangin induces B16F10 melanoma cell apoptosis via mitochondrial pathway and sustained activation of p38 MAPK, Cytotechnology, vol.65, pp.447-455, 2013.

W. Zhang, B. Tang, Q. Huang, and Z. Hua, Galangin inhibits tumor growth and metastasis of B16F10 melanoma, J. Cell. Biochem, vol.114, pp.152-161, 2013.

R. Almushatat, A. Talwar, D. Mcardle, P. A. Williamson, C. Sattar et al., Vitamin antioxidants, lipid peroxidation and the systemic inflammatory response in patients with prostate cancer, Int J Cancer, vol.118, pp.1051-1053, 2006.

C. O. Beauchamp and I. Fridovich, Superoxide dismutase: improved assays and an assay applicable to acrylamide gels, Anal Biochem, vol.44, pp.276-287, 1971.

D. Bonnefont-rousselot, Stress oxydant et vieillissement. Spectra Biol, vol.157, pp.23-26, 2007.

A. Claiborne, Catalase activity. In: CRC Handbook of Methods for Oxygen Radical Research, pp.283-284, 1985.

V. Ducros, M. Ferry, P. Faure, N. Belin, J. C. Renversez et al., Distribution of selenium in plasma of French women: relation to age and selenium status, Clin Chem, vol.46, pp.731-733, 2000.

G. Ellman, Tissue sulfhydryl groups, Arch Biochem Biophys, vol.32, pp.70-77, 1959.

A. Favier, Le stress oxydant. Int er^ et conceptuel et exp erimental dans la compr ehension des m ecanismes des maladies et potentiel th erapeutique, pp.108-115, 2003.

M. Gard-es-albert, D. Bonnefont-rousselot, Z. Abedinzadeh, and D. Jore, Esp eces r eactives de l'oxyg ene. Comment l'oxyg ene peut-il devenir toxique?, pp.91-96, 2003.

W. H. Habig, M. J. Pabst, and W. B. Jakoby, Glutathione Stransferase, the first enzymatic step in mercapturic acid formation, J Biol Chem, vol.249, pp.7130-7139, 1974.

K. Jung, B. Seidel, B. Rudolph, M. Lein, M. V. Cronauer et al., Antioxidant enzymes in malignant prostate cell lines and in primary cultured prostatic cells, Free Radic Biol Med, vol.23, pp.127-133, 1997.

L. Khandrika, B. Kumar, S. Koul, P. Maroni, and H. K. Koul, Role of oxidative stress in prostate cancer, Cancer Lett, vol.282, pp.125-136, 2009.

W. M. Kwiatek, G. Bana-s-a,-bana-s-k,-cinque, G. Dyduch, G. Falkenberg, A. Kisiel et al., Podg orczyk M (2007) Micro and bulk analysis of prostate tissues classified as hyperplasia, Spectrochim Acta, Part B, vol.62, pp.707-710

O. H. Lowry, N. J. Rosebrough, A. L. Farr, and R. J. Randall, Protein measurement with the Folin phenol reagent, J Biol Chem, vol.193, pp.265-275, 1951.

A. Lukaszewicz-hussain, Organophosphate insecticide chlorfenvinphos affects superoxide dismutase, catalase and malondialdehyde in rat liver, Polish J Environ Stud, vol.10, pp.279-282, 2001.

A. ?ukaszewicz-hussain and J. Moniuszko-jakoniuk, Chlorfenvinphos, an organophosphate insecticide, affects liver mitochondria antioxidative enzymes, glutathione and hydrogen peroxide concentration, Polish J Environ Stud, vol.13, pp.397-401, 2004.

F. Mehraein-ghomi, L. E. Church, D. R. Thompson, T. A. Basu, H. S. Wilding et al., JunD mediates androgen-induced oxidative stress in androgen dependent LNCaP human prostate cancer cells, Prostate, vol.68, pp.924-934, 2008.

H. Okhawa, N. Ohishi, and K. Yagi, Assay of lipid peroxides in animal tissue by thiobarbituric reaction, Anal Biochem, vol.95, pp.351-358, 1979.

R. Olinski, T. H. Zastawny, M. Foksinski, A. Barecki, and M. Dizdaroglu, DNA base modifications and antioxidant enzyme activities in human benign prostatic hyperplasia, Free Radic Biol Med, vol.18, pp.807-813, 1995.

D. E. Paglia and W. N. Valentine, Studies on the quantitative and qualitative characterization of erythrocyte glutathione peroxidase, J Lab Clin Med, vol.70, pp.158-169, 1967.

P. Pasupathi, G. Saravanan, and J. Farook, Oxidative stress bio markers and antioxidant status in cigarette smokers compared to nonsmokers, J Pharm Sci Res, vol.1, pp.55-62, 2009.

J. Pincemail, K. Bonjean, K. Cayeux, and J. O. Defraigne, Prooxidant-antioxidant shift induced by androgen treatment of human prostate carcinoma cells, J Natl Cancer Inst, vol.16, pp.40-48, 1997.

M. Samir and N. M. Kholy, Thiobarbituric acid reactive substances in patients with laryngeal cancer, Clin Otolaryngol, vol.24, pp.232-234, 1999.

M. Savas, Oxidative stress in benign prostate hyperplasia, Studies on Men's Health and Fertility, 2012.

M. Valko, C. J. Rhodes, J. Moncol, M. Izakovic, and M. Mazur, Free radicals, metals and antioxidants in oxidative stressinduced cancer, Chem Biol Interact, vol.160, pp.1-40, 2006.

, Références bibliographiques

C. Abate-shen and M. M. Shen, Mouse models of prostate carcinogenesis, Trends Genet, vol.18, issue.5, pp.1-5, 2002.

C. Abate-shen and M. M. Shen, Molecular genetics of prostate cancer, Genes Dev, vol.14, pp.2410-2434, 2010.

M. Abdelrahim, R. Smith, and S. Safe, Aryl hydrocarbon receptor gene silencing with small inhibitory RNA differentially modulates Ah-responsiveness in MCF-7 and HepG2 cancer cells, 2003.

, Mol Pharmacol, vol.63, pp.1373-1381

P. A. Abrahamsson, Neuroendocrine differentiation and hormone-refractory prostate cancer, Prostate Suppl, vol.6, pp.3-8, 1996.

I. U. Agoulnik and N. L. Weigel, Androgen receptor action in hormonedependent and recurrent prostate cancer, J Cell Biochem, vol.99, pp.362-372, 2006.

S. M. Alencar, T. Oldoni, M. L. Castro, I. Cabral, C. M. Costa-neto et al., Chemical composition and biological activity of a new type of Brazilian propolis: red propolis, J Ethnopharmacol, vol.113, pp.278-283, 2007.

R. T. Allen, M. W. Cluck, and D. K. Agrawal, Mechanisms controlling cellular suicide: role of Bcl2 and caspases, Cell Mol Life Sci, vol.54, pp.427-445, 1998.

Y. Amakura, T. Tsutsumi, M. Nakamura, H. Handa, M. Yoshimura et al., Tectochrysin in propolis is a potent natural aryl hydrocarbon receptor ligand, Planta Med, vol.76, p.676, 2010.

Y. Amakura, T. Tsutsumi, K. Sasaki, M. Nakamura, T. Yoshida et al., Influence of food polyphenols on aryl hydrocarbon receptor-signaling pathway estimated by in vitro bioassay, Phytochemistry, vol.69, issue.18, pp.3117-3130, 2008.

N. Amini-sarteshnizi, M. Mobini-dehkordi, S. Khosravi-farsani, and H. Teimori, Anticancer activity of ethanolic extract of propolis on AGS cell line, J HerbMed Pharmacol, vol.4, issue.1, pp.29-34, 2015.

G. L. Andriole, E. D. Crawford, R. L. Grubb, S. S. Buys, D. Chia et al., Mortality results from a randomized prostate-cancer screening trial, N Engl J Med, vol.360, pp.1310-1319, 2009.

S. Anttila, J. Hakkola, P. Tuominen, E. Elovaara, K. Husgafvel-pursiainen et al., Methylation of cytochrome P4501A1 promoter in the lung is associated with tobacco smoking, Cancer Res, vol.63, pp.8623-8628, 2003.

H. M. Arafa, H. A. Aly, M. F. Abd-ellah, and H. M. El-refaey, Hesperidin attenuates benzo[?]pyrene-induced testicular toxicity in rats via regulation of oxidant/antioxidant balance, 2009.
DOI : 10.1177/0748233709106624

, Toxicol Ind Health, vol.25, issue.6, pp.417-427

J. Arey and R. Atkinson, Photochemical reactions of PAH in the atmosphere P.E.T. Douben (Ed.)In: PAHs: an ecotoxicological perspective, pp.47-63, 2003.

B. G. Armstrong, E. Hutchinson, J. Unwin, and T. Fletcher, Lung Cancer Risk after Exposure to Polycyclic Aromatic Hydrocarbons: A Review and Meta-Analysis, Environ Health Perspect, vol.112, issue.9, pp.970-978, 2004.

Z. Arsova-sarafinovska, A. Eken, N. Matevska, O. Erdem, A. Sayal et al., Increased oxidative/nitrosative stress and decreased antioxidant enzyme activities in prostate cancer, Clin Biochem, vol.42, pp.1228-1235, 2009.

K. Aso, S. Kanno, T. Tadano, S. Satoh, and M. Ishikawa, Inhibitory effect of propolis on the growth of human leukemia U937, Biol Pharm Bull, vol.27, issue.5, pp.727-730, 2004.

A. , Toxicological Profile For Polycyclic Aromatic Hydrocarbons, p.458, 1995.

S. Awale, F. Li, H. Onozuka, H. Esumi, Y. Tezuka et al., Constituents of Brazilian red propolis and their preferential cytotoxic activity against human pancreatic PANC-1 cancer cell line in nutrient-deprived condition, Bioorg Med Chem, vol.16, pp.181-189, 2008.

M. Banasiewicz, G. Nelson, A. Swank, N. Grubor, J. Ross et al., Identification and quantitation of benzo[a]pyrenederived DNA adducts formed at low adduction level in mice lung tissue, Anal Biochem, vol.334, issue.2, pp.390-400, 2004.

V. Bankova, Chemical diversity of propolis and the problem of standardization, J Ethnopharmacol, vol.100, pp.114-117, 2005.

V. Bankova, Recent trends and important developments in propolis research. Evid.-Based Complement, Altern Med ECAM, vol.2, pp.29-32, 2005.

V. S. Bankova, S. L. De-castro, M. Maria, and C. , Propolis: recent advances in chemistry and plant origin, Apidologie, pp.3-15, 2000.
URL : https://hal.archives-ouvertes.fr/hal-00891696

B. Barlaam, S. Cosulich, S. Degorce, M. Fitzek, S. Green et al., Discovery of (R)-8-(1-(3,5difluorophenylamino)ethyl)-N,N-dimethyl-2-morpholino-4-oxo-4H-chromene-6-carboxamide (AZD8186): a potent and selective inhibitor of PI3K? and PI3K? for the treatment of PTENdeficient cancers, J Med Chem, vol.58, pp.943-962, 2015.

Y. Barlak, O. De?er, M. Colak, S. C. Karatayl, A. M. Bozday et al., Effect of Turkish propolis extracts on proteome of prostate cancer cell line, Proteome Sci, vol.9, p.74, 2011.

S. Baron, M. Manin, C. Beaudoin, L. Leotoing, Y. Communal et al., Androgen receptor mediates non-genomic activation of phosphatidylinositol 3-OH kinase in androgen-sensitive epithelial cells, J Biol Chem, vol.279, issue.15, pp.14579-14586, 2004.

R. Barouki, X. Coumoul, and P. M. Fernandez-salguero, The arylhydrocarbon receptor, more than a xenobiotic-interacting protein, FEBS Lett, vol.581, pp.3608-3615, 2007.

V. Basset, V. Flamandb, S. Crouzetc, and G. Ploussard, Choix des nouveaux traitements médicaux dans le cancer de la prostate résistant à la castration : marqueurs prédictifs et évaluation de l'efficacité, Prog Urol, vol.23, issue.1, pp.44-48, 2013.

C. Bastide, C. Bagnis, P. Mannoni, J. Hassoun, and F. Bladou, A Nod Scid mouse model to study human prostate cancer, Prostate Cancer Prostatic Dis, vol.5, pp.311-315, 2002.

C. O. Beauchamp and I. Fridovich, Superoxide dismutase: improved assays and an assay applicable to acrylamide gels, Anal Biochem, vol.44, pp.276-287, 1971.

E. Benedettini, P. Nguyen, and M. Loda, The pathogenesis of prostate cancer: from molecular to metabolic alterations, Diagn Histopathol (Oxf), vol.14, issue.5, pp.195-201, 2008.

L. Benguedouar, M. Lahouel, S. C. Gangloff, A. Durlach, F. Grange et al., Ethanolic Extract of Algerian Propolis and Galangin Decreased Murine Melanoma T, Anticancer Agents Med Chem, vol.16, issue.9, pp.1172-1183, 2016.

G. J. Berchem, M. Bosseler, L. Y. Sugars, H. J. Voeller, S. Zeitlin et al., Androgens induce resistance to Bcl-2-mediated apoptosis in LNCaP prostate cancer cells, Cancer Res, vol.55, pp.735-738, 1995.

D. M. Berney, A. Gopalan, S. Kudahetti, G. Fisher, L. Ambroisine et al., Ki-67 and outcome in clinically localised prostate cancer: analysis of conservatively treated prostate cancer patients from the Trans-Atlantic Prostate Group study, Br J Cancer, vol.100, issue.6, pp.888-893, 2009.

I. M. Berquin, Y. Min, R. Wu, H. Wu, and Y. Q. Chen, Expression signature of the mouse prostate, J Biol Chem, vol.280, issue.43, pp.36442-36451, 2005.

D. R. Berthold, G. R. Pond, F. Soban, R. De-wit, M. Eisenberger et al., Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer: updated survival in the TAX 327 study, J Clin Oncol, vol.26, pp.242-245, 2008.

E. Birben, U. M. Sahiner, C. Sackesen, S. Erzurum, and O. Kalayci, Oxidative Stress and Antioxidant Defense, World Allergy Organ J, vol.5, issue.1, pp.9-19, 2012.

T. Boam, Anti-androgenic effects of flavonols in prostate cancer. Ecancermedicalscience, vol.9, p.585, 2015.

C. Bonne and J. P. Raynnaud, Assay of androgen binding sites by exchange with methyltrienolone (R 1881), Steroids, vol.27, issue.4, pp.497-507, 1975.

J. L. Bosch, K. Tilling, A. M. Bohnen, C. H. Bangma, and J. L. Donovan, Establishing normal reference ranges for prostate volume change with age in the population-based Krimpen-study: Prediction of future prostate volume in individual men, Prostate, vol.67, pp.1816-1824, 2007.

M. C. Bosland, M. K. Prinsen, T. J. Dirksen, and B. J. Spit, (a)anthracene, and 3,2'-dimethyl-4-aminobiphenyl, following sequential treatment with cyproterone acetate and testosterone propionate, Cancer Res, vol.7, issue.3, pp.700-709, 1990.

P. J. Bostrom, A. S. Bjartell, J. W. Catto, S. E. Eggener, H. Lilja et al., Genomic predictors of outcome in prostate cancer, Eur Urol, vol.68, pp.1033-1044, 2015.

D. P. Bottaro, J. S. Rubin, D. L. Faletto, A. M. Chan, T. E. Kmiecik et al., Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product, Science, vol.251, issue.4995, pp.802-804, 1991.

M. Bouchard and C. Viau, Urinary excretion of benzo[a]pyrene metabolites following intravenous, oral, and cutaneous benzo[a]pyrene administration, Can J Physiol Pharmacol, vol.75, pp.185-192, 1997.

K. Boutabet, W. Kebsa, M. Alyane, and M. Lahouel, Polyphenolic fraction of Algerian propolis protects rat kidney against acute oxidative stress induced by doxorubicin, Indian J Nephrol, vol.21, issue.2, pp.101-106, 2011.

P. C. Boutros, M. Fraser, N. J. Harding, R. De-borja, D. Trudel et al., Spatial genomic heterogeneity within localized, vol.47, pp.736-745, 2015.

M. K. Brawer, D. M. Peehl, T. A. Stamey, and D. G. Bostwick, Keratin immunoreactivity in the benign and neoplastic human prostate, Cancer Res, vol.45, pp.3663-3667, 1985.

A. Briganti, U. Capitanio, N. Suardi, A. Gallina, A. Salonia et al., Benign Prostatic Hyperplasia and Its Aetiologies, Eur Urol, vol.8, pp.865-871, 2009.

G. N. Brooke and C. L. Bevan, The Role of Androgen Receptor Mutations in Prostate Cancer Progression, Curr Genomics, vol.9, pp.18-25, 2009.

L. Bubendorf, J. Kononen, P. Koivisto, P. Schraml, H. Moch et al., Survey of gene amplifications during prostate cancer progression by high-throughput fluorescence in situ hybridization on tissuemicroarrays, Cancer Res, vol.59, pp.803-806, 1999.

M. Bui and R. E. Reiter, Stem cell genes in androgenindependent prostate cancer, Cancer Metastasis Rev, vol.17, pp.391-399, 1998.

G. A. Burdock, Review of the biological properties and toxicity of bee propolis (propolis), Food Chem Toxicol. Int J Publ Br Ind Biol Res Assoc, vol.36, pp.347-363, 1998.

W. F. Busby, H. Smith, C. L. Crespi, and B. W. Penman, Mutagenicity of benzo[a]pyrene and dibenzopyrenes in the Salmonella typhimurium TM677 and the MCL-5 human cell forward mutation assays, Mutat Res, vol.342, issue.1-2, pp.9-16, 1995.

C. Cai and S. P. Balk, Intratumoral androgen biosynthesis in prostate cancer pathogenesis and response to therapy, Endocr-Relat Cancer, vol.18, pp.175-182, 2011.

M. H. Cardone, N. Roy, H. R. Stennicke, G. S. Salvesen, T. F. Franke et al., Regulation of cell death protease caspase-9 by phosphorylation, Science, vol.282, pp.1318-1321, 1998.

A. G. Carroll, H. J. Voeller, L. Sugars, and E. P. Gelmann, p53 oncogene mutations in three human prostate cancer cell lines, Prostate, vol.23, pp.123-134, 1993.

J. P. Carson, G. Kulik, and M. J. Weber, Antiapoptotic signaling in LNCaP prostate cancer cells: a survival signaling pathway independent of phosphatidylinositol 3'-kinase and Akt/protein kinase B, Cancer Res, vol.59, pp.1449-1453, 1999.

M. V. Chad, L. Tien-min, and E. P. Richard, AHR signaling in prostate growth, morphogenesis, and disease, Biochem Pharmacol, vol.77, issue.4, pp.566-576, 2009.

D. Chakravarti, J. C. Pelling, E. L. Cavalieri, and E. G. Rogan, Relating aromatic hydrocarboninduced DNA adducts and c-H-ras mutations in mouse skin papillomas: the role of apurinic sites, Proc Natl Acad Sci, vol.92, issue.22, pp.10422-10426, 1995.

D. Chakravarti, D. Venugopal, P. C. Mailander, J. L. Meza, S. Higginbotham et al., The role of polycyclic aromatic hydrocarbon-DNA adducts in inducing mutations in mouse skin, Mutat Res, vol.649, issue.1-2, pp.161-178, 2008.

C. D. Chen, D. S. Welsbie, C. Tran, S. H. Baek, R. Chen et al., Molecular determinants of resistance to antiandrogen therapy, Nat Med, vol.10, issue.1, pp.33-39, 2004.

J. Chen, L. Xiao, J. N. Rao, T. Zou, L. Liu et al., JunD Represses Transcription and Translation of the Tight Junction Protein Zona Occludens-1, 2008.

, Modulating Intestinal Epithelial Barrier Function, Mol Biol Cell, vol.19, pp.3701-3712

J. Z. Chen, N. Gokden, G. F. Greene, B. Green, and F. F. Kadlubar, Simultaneous generation of multiple mitochondrial DNA mutations in human prostate tumors suggests mitochondrial hypermutagenesis, Carcinogenesis, vol.24, pp.1481-1487, 2003.

L. Chen, M. Stacewicz-sapuntzakis, C. Duncan, R. Sharifi, L. Ghosh et al., , 2001.

, Oxidative DNA damage in prostate cancer patients consuming tomato sauce-based entrees as a whole-food intervention, J Natl Cancer Inst, vol.93, pp.1872-1879

S. Chen, S. Gulla, C. Cai, and S. P. Balk, Androgen receptor serine 81 phosphorylation mediates chromatin binding and transcriptional activation, J Biol Chem, vol.287, pp.8571-8583, 2012.

X. Chen, H. An, L. Ao, L. Sun, W. Liu et al., The combined toxicity of dibutyl phthalate and benzo[a]pyrene on the reproductive system of male Sprague Dawley rats in vivo, J Hazard Mater, vol.186, issue.1, pp.835-841, 2011.

Z. Chen, A. Rasul, C. Zhao, F. M. Millimouno, I. Tsuji et al., Antiproliferative and apoptotic effects of pinocembrin in human prostate cancer cells, Bangladesh J Pharmacol, vol.8, issue.3, pp.255-262, 2013.

K. Z. Ching, E. Ramsey, N. Pettigrew, R. Cunha, M. Jason et al., Expression of mRNA for epidermal growth factor, transforming growth factor-alpha and their receptor in human prostate tissue and cell lines, Mol Cell Biochem, vol.126, issue.2, pp.151-158, 1993.

J. K. Chipman, P. C. Hirom, G. S. Frost, and P. Millburn, The biliary excretion and enterohepatic circulation of benzo(a)pyrene and its metabolites in the rat, Biochem Pharmacol, vol.30, pp.937-944, 1981.

S. C. Chiu, M. J. Wang, H. H. Yang, S. P. Chen, S. Y. Huang et al., Activation of NAG-1 via JNK signaling revealed an isochaihulactone-triggered cell death in human LNCaP prostate cancer cells, BMC Cancer, issue.11, p.146, 2011.

D. R. Church, E. Lee, T. A. Thompson, H. S. Basu, M. O. Ripple et al., Induction of AP-1 activity by androgen activation of the androgen receptor in LNCaP human prostate carcinoma cells, The Prostate, vol.63, pp.155-168, 2005.

C. Chuu, H. Lin, M. F. Ciaccio, J. M. Kokonti, R. J. Hause et al., Caffeic acid phenethyl ester suppresses the proliferation of human prostate cancer cells through inhibition of p70S6K and Akt signaling networks, Cancer Prev Res Phila, vol.5, pp.788-797, 2012.

F. H. Claas and G. J. Van-steenbrugge, Expression of HLA-like structures on a permanent human tumor line PC-93, Tissue Antigens, vol.21, pp.227-232, 1983.

R. Claus, R. Kinscherf, C. Gehrke, G. Bonaterra, P. Basnet et al., Antiapoptotic effects of propolis extract and propol on human macrophages exposed to minimally modified low density lipoprotein, Drug Res, vol.50, pp.373-379, 2000.

K. Coffey and C. N. Robson, Regulation of the androgen receptor by post-translational modifications, J Endocrinol, vol.215, pp.221-237, 2012.

R. N. Coffey, R. W. Watson, and J. M. Fitzpatrick, Signaling for the caspases: their role in prostate cell apoptosis, J Urol, vol.165, pp.5-14, 2001.

D. Cunningham and Z. You, In vitro and in vivo model systems used in prostate cancer research, J Biol Methods, vol.2, issue.1, pp.1-14, 2015.

U. Czy?ewska, K. Siemionow, I. Zar?ba, and W. Miltyk, Proapoptotic Activity of Propolis and Their Components on Human Tongue Squamous Cell Carcinoma Cell Line (CAL-27), PLoS One, vol.11, issue.6, p.157091, 2016.

S. S. Da-silva, G. Thomé, S. Da, A. Cataneo, M. M. Miranda et al., Brazilian propolis antileishmanial and immunomodulatory effects, Evid-Based Complement Altern Med, p.673058, 2013.

C. Da-silveira, M. De, L. Fernandes, M. L. Silva, D. A. Luz et al., Neurobehavioral and Antioxidant Effects of Ethanolic Extract of Yellow Propolis, Oxid Med Cell Longev, p.2906953, 2016.

B. D. Darimont, R. L. Wagner, J. W. Apriletti, M. R. Stallcup, P. J. Kushner et al., Structure and specificity of nuclear receptor-coactivator interactions, Genes Dev, vol.12, pp.3343-3356, 1998.

M. S. Darshan, M. S. Loftus, M. Thadani-mulero, B. P. Levy, D. Esciun et al., Taxaneinduced blockade to nuclear accumulation of the androgen receptor predicts clinical responses in metastatic prostate cancer, Cancer Res, vol.71, pp.6019-6029, 2011.

S. R. Datta, H. Dudek, X. Tao, S. Masters, H. Fu et al., Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery, Cell, vol.91, pp.231-241, 1997.

D. Jong, W. H. Kroese, E. D. Vos, J. G. Van-loveren, and H. , Detection of immunotoxicity of benzo[a]pyrene in a subacute toxicity study after oral exposure in rats, Toxicol Sci, vol.50, pp.214-220, 1999.

I. De-mendonça, I. Porto, M. De, T. G. Do-nascimento, N. S. Souza et al., Brazilian red propolis: phytochemical screening, antioxidant activity and effect against cancer cells, BMC Complement Altern Med, vol.15, p.357, 2015.

D. Nunzio, C. Kramer, G. Marberger, M. Montironi, R. Nelson et al., The Controversial Relationship Between Benign Prostatic Hyperplasia and Prostate Cancer: The Role of Inflammation, Eur Urol, vol.60, issue.1, pp.106-117, 2011.

V. A. De-weger, J. H. Beijnen, and J. Schellens, Cellular and clinical pharmacology of the taxanes docetaxel and paclitaxel-a review, Anticancer Drugs, vol.25, pp.488-494, 2014.

J. A. Decaprio, J. W. Ludlow, J. Figge, J. Y. Shew, C. M. Huang et al., SV40 large tumor antigen forms a specific complex with the product of the retinoblastoma susceptibility gene, Cell, vol.54, issue.2, pp.275-283, 1988.

S. M. Dehm and D. J. Tindall, Molecular regulation of androgen action in prostate cancer, J Cell Biochem, vol.99, pp.333-344, 2006.

S. M. Dehm and D. J. Tindall, Androgen receptor structural and functional elements: role and regulation in prostate cancer, Mol Endocrinol, vol.21, pp.2855-2863, 2007.
DOI : 10.1210/me.2007-0223

URL : https://academic.oup.com/mend/article-pdf/21/12/2855/8958094/mend2855.pdf

A. D. Delbridge, S. Grabow, A. Strasser, and D. L. Vaux, Thirty years of BCL-2: translating cell death discoveries into novel cancer therapies, Nat Rev Cancer, vol.16, pp.99-109, 2016.
DOI : 10.1038/nrc.2015.17

H. L. Devlin and M. Mudryj, Progression of prostate cancer: multiple pathways to androgen independence, Cancer Lett, vol.274, issue.2, pp.177-186, 2009.
DOI : 10.1016/j.canlet.2008.06.007

J. Digiovanni, L. Beltran, A. Rupp, R. G. Harvey, and R. D. Gill, Further analysis of c-Haras mutations in papillomas initiated by several polycyclic aromatic hydrocarbons and papillomas from uninitiated, promoter-treated skin in SENCAR mice, Mol Carcinog, vol.8, issue.4, pp.272-279, 1993.

V. Dimov, N. Ivanovska, V. Bankova, and S. Popov, Immunomodulatory action of propolis: IV. Prophylactic activity against gram-negative infections and adjuvant effect of the water-soluble derivative, Vaccine, vol.10, pp.817-823, 1992.

D. M. Di-toro, J. A. Mcgrath, and D. J. Hansen, Technical basis for narcotic chemicals and polycyclic aromatic hydrocarbon criteria. I. Water and tissue, Environ Toxicol Chem, vol.19, pp.1951-1970, 2000.

D. Djakiew, Dysregulated expression of growth factors and their receptors in the development of prostate cancer, Prostate, vol.42, issue.2, pp.150-160, 2010.

K. Dota, M. Consolaro, T. Svidzinski, and M. L. Bruschi, Antifungal Activity of Brazilian Propolis Microparticles against Yeasts Isolated from Vulvovaginal Candidiasis. Evid.Based Complement, Evid Based Complement Alternat Med, 2011.
DOI : 10.1093/ecam/neq029

URL : http://downloads.hindawi.com/journals/ecam/2011/201953.pdf

H. Dotzlaw, U. Moehren, S. Mink, A. C. Cato, J. A. Iniguez-lluhi et al., The amino terminus of the human AR is target for corepressor action and antihormone agonism, Mol Endocrinol, vol.16, pp.661-673, 2002.

A. P. Drabovich, P. Saraon, K. Jarvi, and E. P. Diamandis, Seminal plasma as a diagnostic fluid for male reproductive system disorders, Nat Rev Urol, vol.11, pp.278-288, 2014.
DOI : 10.1038/nrurol.2014.74

R. Dutour and D. Poirier, Inhibitors of cytochrome P450 (CYP) 1B1, Eur J Med Chem, 2017.
DOI : 10.1016/j.ejmech.2017.04.042

B. Ebert, A. Seidel, and A. Lampen, Identification of BCRP as transporter of benzo[a]pyrene conjugates metabolically formed in Caco-2 cells and its induction by Ah-receptor agonists, Carcinogenesis, vol.26, issue.10, pp.1754-1763, 2005.

, Modulating Role of Panax Ginseng in Experimentally Induced Benign Prostatic Hyperplasia in Adult Male Albino Rats, Austin J Anat, vol.2, issue.1, pp.1-8, 2015.

G. Espinosa, R. Esposito, A. Kazzazi, and B. Djavan, Vitamin D and benign prostatic hyperplasia-a review, Can J Urol, vol.20, issue.4, pp.6820-6825, 2013.

S. P. Flanagan, Nude', a new hairless gene with pleiotropic effects in the mouse, Genet Res, vol.8, issue.3, pp.295-309, 1966.

K. Förster, R. Preuss, B. Rossbach, T. Brüning, J. Angerer et al., 3Hydroxybenzo[a]pyrene in the urine of workers with occupational exposure to polycyclic aromatic hydrocarbons in different industries, Occup Environ Med, vol.65, pp.224-229, 2008.

B. A. Foster, J. R. Gingrich, E. D. Kwon, C. Madias, and N. M. Greenberg, Characterization of prostatic epithelial cell lines derived from transgenic adenocarcinoma of the mouse prostate (TRAMP) model, Cancer Res, vol.57, issue.16, pp.3325-3330, 1997.

H. Foth, R. Kahl, and G. F. Kahl, Pharmacokinetics of low doses of benzo[a]pyrene in the rat, Food Chem Toxicol, vol.26, pp.45-51, 1988.

S. B. Frank and C. K. Miranti, Disruption of prostate epithelial differentiation pathways and prostate cancer development, Front Oncol, vol.3, issue.273, pp.1-23, 2013.

M. Freitas, V. Alves, A. Sarmento-ribeiro, and A. Mota-pinto, Polycyclic Aromatic Hydrocarbons May Contribute for Prostate Cancer Progression, J Cancer Ther, vol.4, pp.37-46, 2013.

D. A. Frohlich, M. T. Mccabe, R. A. Arnold, and M. L. Day, The role of Nrf2 in increased reactive oxygen species and DNA damage in prostate tumorigenesis, Oncogene, vol.27, pp.4353-4362, 2008.

C. Füllhase and M. P. Schneider, 5-Alpha-Reductase Inhibitors and Combination Therapy, Urol Clin North Am, vol.43, issue.3, pp.325-336, 2016.

P. Galani, Diagnosis & prognosis of prostate cancer, J Adv Med Dent Scie Res, vol.3, issue.5, pp.49-53, 2015.

W. Gao, C. E. Bohl, and D. J. , Chemistry and Structural Biology of Androgen Receptor, Chem Rev, vol.105, issue.9, pp.3352-3370, 2005.

J. Gasmi and J. T. Sanderson, Growth inhibitory, antiandrogenic, and pro-apoptotic effects of punicic acid in LNCaP human prostate cancer cells, J Agric Food Chem, vol.58, issue.23, pp.12149-12156, 2010.
URL : https://hal.archives-ouvertes.fr/pasteur-00819575

I. Geramoutsos, K. Gyftopoulos, P. Perimenis, V. Thanou, D. Liagka et al., Clinical correlation of prostatic lithiasis with chronic pelvic pain syndromes in young adults, Eur Urol, vol.45, pp.333-338, 2004.

U. Gluschnaider, G. Hidas, G. Cojocaru, V. Yutkin, Y. Ben-neriah et al., Beta-TrCP inhibition reduces prostate cancer cell growth via upregulation of the aryl hydrocarbon receptor, PLoS One, vol.5, issue.2, pp.1-12, 2010.

P. Grant and S. Ramasamy, An Update on Plant Derived Anti-Androgens, Int J Endocrinol Metab, vol.10, issue.2, pp.497-502, 2012.

G. Gundem, P. Van-loo, B. Kremeyer, L. B. Alexandrov, J. M. Tubio et al., The evolutionary history of lethal metastatic prostate cancer, Nature, vol.520, pp.353-357, 2015.

G. Gupta-elera, A. R. Garrett, R. A. Robison, and K. L. Neill, The role of oxidative stress in prostate cancer, Eur J Cancer Prev, vol.21, issue.2, pp.155-162, 2012.

W. H. Habig, M. J. Pabst, and W. B. Jakoby, Glutathione Stransferase, the first enzymatic step in mercapturic acid formation, J Biol Chem, vol.249, pp.7130-7139, 1974.

A. R. Hamid, R. Umbas, and C. A. Mochtar, Recent role of inflammation in prostate diseases: chemoprevention development opportunity, Acta Med Indones, vol.43, pp.59-65, 2011.

H. Y. Han, S. Shan, X. Zhang, N. L. Wang, X. P. Lu et al., Down-regulation of prostate specific antigen in LNCaP cells by flavonoids from the pollen of Brassica napus L, Phytomedicine, vol.14, issue.5, pp.338-343, 2007.

D. M. Heery, E. Kalkhoven, S. Hoare, and M. G. Parker, A signature motif in transcriptional coactivators mediates binding to nuclear receptors, Nature, vol.387, pp.733-736, 1997.

C. Helsen, T. Van-den-broeck, A. Voet, S. Prekovic, H. Van-poppel et al., Androgen receptor antagonists for prostate cancer therapy, Endocr Relat Cancer, vol.21, issue.4, pp.105-118, 2014.

S. L. Hockley, V. M. Arlt, D. Brewer, R. Te-poele, P. Workman et al., , 2007.

A. , DNA-damage-mediated gene expression responses induced by benzo(a)pyrene in human cell lines, Chem Res Toxicol, vol.20, pp.1797-1810

M. C. Hodgson, H. C. Shen, A. N. Hollenberg, and S. P. Balk, Structural basis for nuclear receptor corepressor recruitment by antagonist-liganded androgen receptor, Mol Cancer Ther, vol.7, pp.3187-3194, 2008.

M. K. Hong, G. Macintyre, D. C. Wedge, P. Van-loo, K. Patel et al., Tracking the origins and drivers of subclonal metastatic expansion in prostate cancer, Nat Commun, vol.6, p.6605, 2015.

J. S. Horoszewicz, S. S. Leong, T. M. Chu, Z. L. Wajsman, M. Friedman et al., The LNCaP cell line-a new model for studies on human prostatic carcinoma, Prog Clin Biol Res, vol.37, pp.115-132, 1980.

J. S. Horoszewicz, S. S. Leong, E. Kawinski, J. P. Karr, H. Rosenthal et al., LNCaP model of human prostatic carcinoma, Cancer Res, vol.43, issue.4, pp.1809-1818, 1983.

Y. Hu, T. Wang, G. D. Stormo, and J. I. Gordon, RNA interference of achaete-scute homolog 1 in mouse prostate neuroendocrine cells reveals its gene targets and DNA binding sites, Proc Nat Acad Sci, vol.101, issue.15, pp.5559-5564, 2004.

S. Huang, C. Zhang, K. Wang, G. Q. Li, and F. Hu, Recent advances in the chemical composition of propolis, Mol Basel Switz, vol.19, pp.19610-19632, 2014.

C. Huggins and C. V. Hodges, Studies on Prostatic Cancer. I. The Effect of Castration, of Estrogen and of Androgen Injection on Serum Phosphatases in Metastatic Carcinoma of the Prostate, Cancer Res, vol.1, issue.4, pp.293-297, 1941.

M. Huncharek, K. S. Haddock, R. Reid, and B. Kupelnick, Smoking as a risk factor for prostate cancer: a meta-analysis of 24 prospective cohort studies, Am J Public Health, vol.100, issue.4, pp.693-701, 2010.

I. , Some Non-heterocyclic Polycyclic Aromatic Hydrocarbons and Some Related Exposures, IARC Monogr Eval Carcinog Risks Hum, vol.92, p.856, 2010.

I. , IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, IARC Monographs, vol.100, 2012.

J. T. Isaacs and D. S. Coffey, Etiology and disease process of benign prostatic hyperplasia, Prostate Suppl, vol.2, pp.33-50, 1989.

M. Ittmann, J. Huang, E. Radaelli, P. Martin, S. Signoretti et al., Animal Models of Human Prostate Cancer: The Consensus Report of the New York Meeting of the Mouse Models of Human Cancers Consortium Prostate Pathology Committee, Cancer Res, vol.73, issue.9, pp.2718-2736, 2013.

A. Jemal, F. Bray, M. M. Center, J. Ferlay, E. Ward et al., Global cancer statistics, CA Cancer J Clin, vol.61, pp.69-90, 2011.

A. Jemal, R. Siegel, E. Ward, Y. Hao, J. Xu et al., Cancer statistics, vol.58, pp.71-96, 2008.

B. C. Jessie, C. Q. Sun, H. R. Irons, F. F. Marshall, D. C. Wallace et al., Accumulation of mitochondrial DNA deletions in the malignant prostate of patients of different ages, Exp Gerontol, vol.37, pp.169-174, 2001.

J. Jiao, S. Wang, R. Qiao, I. Vivanco, P. A. Watson et al., Murine cell lines derived from Pten null prostate cancer show the critical role of PTEN in hormone refractory prostate cancer development, Cancer Res, vol.67, issue.13, pp.6083-6091, 2007.

J. B. Turner, L. Zhou, Z. Zhou, E. L. Handelsman, and D. J. , Ethnicity and migration as determinants of human prostate size, J Clin Endocrinol Metab, vol.84, pp.3613-3619, 1999.

D. J. Jollow, J. R. Mitchell, N. Zampaglione, and J. R. Gillette, Bromobenzene-induced liver necrosis. Protective role of glutathione and evidence for 3,4-bromobenzene oxide as the hepatotoxic metabolite, Pharmacology, vol.11, issue.3, pp.151-169, 1974.

L. Kai and A. S. Levenson, Combination of resveratrol and antiandrogen flutamide has synergistic effect on androgen receptor inhibition in prostate cancer cells, Anticancer Res, vol.31, issue.10, pp.3323-3330, 2011.

A. Kapoor, Benign prostatic hyperplasia (BPH) management in the primary care setting, Can J Urol, vol.19, issue.1, pp.10-17, 2012.

E. R. Kasala, L. N. Bodduluru, C. C. Barua, C. S. Sriram, and R. Gogoi, Benzo(a)pyrene induced lung cancer: Role of dietary phytochemicals in chemoprevention, Pharmacol Rep, vol.67, issue.5, pp.996-1009, 2015.

M. Kashani, G. Steiner, A. Haitel, K. Schaufler, T. Thalhammer et al., Expression of the aryl hydrocarbon receptor (AhR) and the aryl hydrocarbon receptor nuclear translocator (ARNT) in fetal, benign hyperplastic, and malignant prostate, Prostate, vol.37, issue.2, pp.98-108, 1998.

Y. Kawamura, E. Kamata, Y. Ogawa, and Y. Saito, The Effect of Various Foods on the Intestinal-Absorption of Benzo(a)Pyrene in Rats, Shokuhin Eiseigaku Zasshi, vol.29, pp.21-25, 1988.

P. L. Kellokumpu-lehtinen, M. Hjalm-eriksson, C. Thellenberg-karlsson, L. Åström, L. Franzen et al., Toxicity in patients receiving adjuvant docetaxel + hormonal treatment after radical radiotherapy for intermediate or high-risk prostate cancer: a preplanned safety report of the SPCG-13 trial, Prostate Cancer Prostatic Dis, vol.15, pp.303-307, 2012.

C. J. Kemp, Animal Models of Chemical Carcinogenesis: Driving Breakthroughs in Cancer Research for 100 Years, Cold Spring Harb Protoc, vol.2015, issue.10, pp.865-874, 2015.

S. A. Kenfield, M. J. Stampfer, J. M. Chan, and E. Giovannucci, Smoking and prostate cancer survival and recurrence, JAMA, vol.305, issue.24, pp.2548-2555, 2011.

S. Khacha-ananda, K. Tragoolpua, P. Chantawannakul, and Y. Tragoolpua, Antioxidant and anti-cancer cell proliferation activity of propolis extracts from two extraction methods, Asian Pac J Cancer Prev, vol.14, pp.6991-6995, 2013.

L. Khandrika, B. Kumar, S. Koul, P. Maroni, and H. K. Koul, Role of Oxidative Stress in Prostate Cancer, Cancer Lett, vol.282, issue.2, pp.125-136, 2009.

J. Kim, S. I. Koo, and S. K. Noh, Green tea extract markedly lowers the lymphatic absorption and increases the biliary secretion of 14C-benzo[a]pyrene in rats, J Nutr Biochem, vol.23, issue.8, pp.1007-1011, 2012.

K. B. Kim and B. M. Lee, Oxidative stress to DNA, protein, and antioxidant enzymes (superoxide dismutase and catalase) in rats treated with benzo(a)pyrene, Cancer Lett, vol.113, pp.205-212, 1997.

T. Kobayashi, T. Nakata, and T. Kuzumaki, Effect of flavonoids on cell cycle progression in prostate cancer cells, Cancer Lett, vol.176, pp.17-23, 2002.

S. Kojima, M. Inahara, H. Suzuki, T. Ichikawa, and Y. Furuya, Implications of insulin-like growth factor-I for prostate cancer therapies, Int J Urol, vol.16, issue.2, pp.161-167, 2009.

A. Kollara and T. J. Brown, Modulation of aryl hydrocarbon receptor activity by four and a half LIM domain 2, Int J Biochem Cell Biol, vol.41, issue.5, pp.1182-1188, 2009.

A. Kollara and T. J. Brown, Four and a half LIM domain 2 alters the impact of aryl hydrocarbon receptor on androgen receptor transcriptional activity, J Steroid Biochem Mol Biol, vol.118, issue.1-2, pp.51-58, 2010.

Y. Komoda, Isolation of flavonoids from Populus nigra as delta 4-3-ketosteroid (5 alpha) reductase inhibitors, Chem Pharm Bull, vol.37, pp.3128-3130, 1989.

M. Krajewska, S. Krajewski, J. I. Epstein, A. Shabaik, J. Sauvageot et al., Immunohistochemical analysis of bcl-2, bax, bcl-xL, and mcl-1 expression on prostate cancers, Am J Pathol, vol.148, pp.1567-1576, 1996.

G. Kroemer, N. Zamzami, and S. A. Susin, Mitochondrial control of apoptosis, Immunol Today, vol.18, issue.1, pp.44-51, 1997.

O. N. Kryvenko and J. Epstein, Prostate Cancer Grading A Decade After the 2005 Modified Gleason Grading System, Arch Pathol Lab Med, vol.140, pp.1140-1152, 2016.

R. Kubina, A. Kaba?a-dzik, A. Dziedzic, B. Bielec, R. D. Wojtyczka et al., The Ethanol Extract of Polish Propolis Exhibits Anti-Proliferative and/or Pro-Apoptotic Effect on HCT 116 Colon Cancer and Me45 Malignant Melanoma Cells In Vitro Conditions, Adv Clin Exp Med, vol.24, pp.203-212, 2015.

N. Kyprianou, H. Tu, and S. C. Jacobs, Apoptotic versus proliferative activities in human benign prostatic hyperplasia, Hum Pathol, vol.27, pp.668-675, 1996.

R. M. Kypta and J. Waxman, Wnt/?-catenin signalling in prostate cancer, Nat Rev Urol, vol.9, pp.418-428, 2012.

L. M. Lacorte, F. K. Delella, E. Amorim, J. R. Justulin, A. F. Godinho et al., Early Changes Induced by Short-Term Low-Dose Cadmium Exposure in Rat Ventral and Dorsolateral Prostates, Microsc Res Tech, vol.74, pp.988-997, 2011.

N. Lallous, K. Dalal, A. Cherkasov, and P. S. Rennie, Targeting Alternative Sites on the Androgen Receptor to Treat Castration-Resistant Prostate Cancer, Int J Mol Sci, vol.14, pp.12496-12519, 2013.

D. J. Lamb and L. Zhang, Challenges in Prostate Cancer Research: Animal Models for Nutritional Studies of Chemoprevention and Disease Progression, J Nutr, vol.135, issue.12, pp.3009-3015, 2005.

D. A. Leach, S. M. Powell, and C. L. Bevan, Women in cancer tematic review: New roles for nuclear receptors in prostate cancer, Endocr Relat Cancer, vol.23, issue.11, pp.85-108, 2016.

B. K. Lee, M. Y. Chung, and K. W. Lee, Benzo[a]pyrene-7,8-diol-9,10-epoxide inhibits gap junction intercellular communication via phosphorylation of tumor progression locus 2 in WBF344 rat liver epithelial cells, Mol Carcinog, vol.54, issue.5, pp.351-358, 2013.

S. H. Lee, D. Johnson, R. Luong, and Z. Sun, Crosstalking between androgen and PI3K/AKT signaling pathways in prostate cancer cells, J Biol Chem, vol.290, issue.5, pp.2759-2768, 2015.

C. Legraverend, T. M. Guenthner, and D. W. Nebert, Importance of the route of administration for genetic differences in benzo[a]pyrene-induced in utero toxicity and teratogenicity, Teratology, vol.29, pp.35-47, 1984.

C. L. Lemieux, G. R. Douglas, J. Gingerich, S. Phonethepswath, D. K. Torous et al., Simultaneous measurement of benzo[a]pyreneinduced Pig-a and lacZ mutations, micronuclei and DNA adducts in Muta Mouse, Environ Mol Mutagen, vol.52, issue.9, pp.756-765, 2011.

H. Lepor, A. Kazzazi, and B. Djavan, ?-Blockers for benign prostatic hyperplasia: the new era, Curr Opin Urol, vol.22, issue.1, pp.7-15, 2012.

S. M. Lesko, L. Rosenberg, and S. Shapiro, Family history and prostate cancer risk, Am J Epidemiol, vol.144, pp.1041-1047, 1996.

F. Li, S. Awale, Y. Tezuka, and S. Kadota, Cytotoxicity of constituents from Mexican propolis against a panel of six different cancer cell lines, Nat Prod Commun, vol.5, issue.10, pp.1601-1606, 2010.

H. Li, A. Kapur, J. X. Yang, S. Srivastava, D. G. Mcleod et al., Antiproliferation of human prostate cancer cells by ethanolic extracts of Brazilian propolis and its botanical origin, Int J Oncol, vol.31, pp.601-606, 2007.

H. Lin, S. S. Jiang, and C. Chuu, Caffeic acid phenethyl ester causes p21 induction, Akt signaling reduction, and growth inhibition in PC-3 human prostate cancer cells, PloS One, vol.7, p.31286, 2012.

H. Lin, C. Lin, C. Huo, P. Hsiao, L. Su et al., Caffeic acid phenethyl ester induced cell cycle arrest and growth inhibition in androgen-independent prostate cancer cells via regulation of Skp2, p53, p21Cip1 and p27Kip1, Oncotarget, vol.6, issue.9, pp.6684-6707, 2015.

P. Lin, H. Chang, W. L. Ho, M. H. Wu, and J. M. Su, Association of aryl hydrocarbon receptor and cytochrome P4501B1 expressions in human non-small cell lung cancers, Lung Cancer, vol.42, pp.255-261, 2003.

T. M. Lin, K. Ko, R. W. Moore, U. Simanainen, T. D. Oberley et al., Effects of aryl hydrocarbon receptor null mutation and in utero and lactational 2,3,7,8-tetrachlorodibenzo-pdioxin exposure on prostate and seminal vesicle development in C57BL/6 mice, Toxicol Sci, vol.68, pp.479-487, 2002.

J. Lindberg, D. Klevebring, W. Liu, M. Neiman, J. Xu et al., Exome sequencing of prostate cancer supports the hypothesis of independent tumour origins, Eur Urol, vol.63, pp.347-353, 2013.

J. M. Lobaccaro and C. Sultan, Normal sexual differentiation: molecular genetic and endocrinology, C R Seances Soc Biol Fil, vol.186, issue.4, pp.314-331, 1992.

S. Loeb, A. Kettermann, H. B. Carter, L. Ferrucci, E. J. Metter et al., Prostate volume changes over time: Results from the Baltimore Longitudinal Study of Aging, J Urol, vol.182, pp.1458-1462, 2009.

D. Lohnes, P. Kastner, A. Dierich, M. Mark, M. Lemeur et al., Function of retinoic acid receptor ? in the mouse, Cell, vol.73, issue.4, pp.643-658, 1993.

P. E. Lonergan and D. J. Tindall, Androgen receptor signaling in prostate cancer development and progression, J Carcinog, vol.10, p.20, 2011.

E. Lonn, J. Bosch, S. Yusuf, P. Sheridan, J. Pogue et al., Effects of long-term vitamin E supplementation on cardiovascular events and cancer: a randomized controlled trial, JAMA, vol.293, pp.1338-1347, 2005.

B. P. Lucey, W. A. Nelson-rees, and G. M. Hutchins, Henrietta Lacks, HeLa Cells, and Cell Culture Contamination, Arch Pathol Lab Med, vol.133, pp.1463-1467, 2009.

G. Luzzi, Prostatitis and male chronic pelvic pain. Medicine, vol.38, pp.314-317, 2010.

B. Machado, R. Silva, A. Barreto-g-de, S. S. Costa, D. F. Silva et al., Chemical Composition and Biological Activity of Extracts Obtained by Supercritical Extraction and Ethanolic Extraction of Brown, Green and Red Propolis Derived from Different Geographic Regions in Brazil, PloS One, vol.11, p.145954, 2016.

N. D. Madhavan and K. A. Naidu, Purification and partial characterization of peroxidase from human term placenta of non-smokers: Metabolism of benzo(a)pyrene-7,8-dihydrodiol, Placenta, vol.21, pp.501-509, 2000.

D. Mangal, D. Vudathala, J. H. Park, S. H. Lee, T. M. Penning et al., Analysis of 7,8dihydro-8-oxo-2?-deoxyguanosine in cellular DNA during oxidative stress, Chem Res Toxicol, vol.22, issue.5, pp.788-797, 2009.

G. E. Mao, G. Morris, Q. Y. Lu, W. Cao, V. E. Reuter et al., Glutathione S-transferase P1 Ile105Val polymorphism, cigarette smoking and prostate cancer, Cancer Detect Prev, vol.28, pp.368-374, 2004.

M. J. Mass, A. J. Jeffers, J. A. Ross, G. Nelson, A. J. Galati et al., Kiras oncogene mutations in tumors and DNA adducts formed by benz[j]aceanthrylene and benzo[a]pyrene in the lungs of strain A/J mice, Mol Carcinog, vol.8, issue.3, pp.186-192, 1993.

K. Mceleny, R. Coffey, C. Morrissey, J. M. Fitzpatrick, and R. Watson, Caffeic acid phenethyl ester-induced PC-3 cell apoptosis is caspase-dependent and mediated through the loss of inhibitors of apoptosis proteins, BJU Int, vol.94, pp.402-406, 2004.

R. A. Mckinnon, M. J. Sorich, and M. B. Ward, Cytochrome P450 Part 1: Multiplicity and Function, J Pharm Pract Res, vol.38, issue.1, pp.55-57, 2008.

J. E. Mcneal, The zonal anatomy of the prostate. The prostate, vol.2, pp.35-49, 1981.

J. E. Mcneal, Anatomy of the prostate and morphogenesis of BPH, Prog Clin Biol Res, vol.145, pp.27-53, 1984.

J. E. Mcneal, Normal histology of the prostate, Am J Surg Pathol, vol.12, issue.8, pp.619-633, 1988.

F. Mehraein-ghomi, E. Lee, D. R. Church, T. A. Thompson, H. S. Basu et al., JunD mediates androgen-induced oxidative stress in androgen dependent LNCaP human prostate cancer cells, Prostate, vol.68, pp.924-934, 2008.

V. J. Melendez-colon, A. Luch, A. Seidel, and W. M. Baird, Cancer initiation by polycyclic aromatic hydrocarbons results from formation of stable DNA adducts rather than apurinic sites, Carcinogenesis, vol.20, pp.1885-1891, 1999.

S. Memarzadeh, H. Cai, D. M. Janzen, L. Xin, R. Lukacs et al., Role of autonomous androgen receptor signaling in prostate cancer initiation is dichotomous and depends on the oncogenic signal, PNAS, vol.108, pp.7962-7967, 2011.

M. G. Miguel, S. Nunes, S. A. Dandlen, A. M. Cavaco, and M. D. Antunes, Phenols and antioxidant activity of hydro-alcoholic extracts of propolis from Algarve, South of Portugal, Food Chem Toxicol, vol.48, pp.3418-3423, 2010.

D. C. Miller, K. S. Hafez, A. Stewart, J. E. Montie, and J. T. Wei, Prostate carcinoma presentation.diagnosis and staging: an update from the National Cancer Data Base, Cancer, vol.98, issue.6, pp.1169-1178, 2003.

P. L. Minciullo, A. Inferrera, M. Navarra, G. Calapai, C. Magno et al., Oxidative stress in benign prostatic hyperplasia: a systematic review, Urol Int, vol.94, issue.3, pp.249-54, 2015.

Y. Mitsui, I. Chang, T. Kato, Y. Hashimoto, S. Yamamura et al., Functional role and tobacco smoking effects on methylation of CYP1A1 gene in prostate cancer, Oncotarget, vol.7, issue.31, pp.49107-49121, 2016.

R. D. Mittal, P. Kesarwani, R. Singh, D. Ahirwar, and A. Mandhani, GSTM1, GSTM3 and GSTT1 gene variants and risk of benign prostate hyperplasia in North India, Dis Markers, vol.26, pp.85-91, 2009.

R. Modica, M. Fiume, A. Guaitani, and I. Bartosek, Comparative kinetics of benz(a)anthracene, chrysene and triphenylene in rats after oral administration. I. Study with single compounds, Toxicol Lett, vol.18, pp.103-109, 1983.

D. Moir, A. Viau, I. Chu, J. Withey, and E. Mcmullen, Pharmacokinetics of benzo[a]pyrene in the rat, J Toxicol Environ Health A, vol.53, issue.7, pp.507-530, 1998.

C. S. Moraes, A. Daugsch, H. Li, J. S. Rhim, and Y. K. Park, Comparative antiproliferation of human prostate cancer cells by ethanolic extracts of two groups of Brazilian propolis, Ciênc Tecnol Aliment, vol.30, issue.2, pp.539-543, 2010.

E. M. Muli and J. M. Maingi, Antibacterial activity of Apis mellifera L. propolis collected in three regions of Kenya, J Venom Anim Toxins Trop Dis, pp.655-663, 2007.

S. D. Mundle, S. A. Gregory, H. D. Preislert, and A. Raza, Enzymatic programming of apoptotic cell death, Pathiobiol, vol.64, pp.161-170, 1996.

I. A. Murray, A. D. Patterson, and G. H. Perdew, Aryl hydrocarbon receptor ligands in cancer: friend and foe, Nat Rev Cancer, vol.14, pp.801-814, 2014.

M. Mustafa, A. F. Salih, E. M. Illzam, A. M. Sharifa, M. Suleiman et al., Prostate Cancer: Pathophysiology, Diagnosis, and Prognosis, IOSR-JDMS, vol.15, issue.6, pp.4-11, 2016.

R. B. Nagle, F. R. Ahmann, K. M. Mcdaniel, M. L. Paquin, V. A. Clark et al., Cytokeratin characterization of human prostatic carcinoma and its derived cell lines, Cancer Res, vol.47, pp.281-286, 1987.

H. K. Nair, K. Rao, R. Aalinkeel, S. Mahajan, R. Chawda et al., Inhibition of prostate cancer cell colony formation by the flavonoid quercetin correlates with modulation of specific regulatory genes, Clin Diagn Lab Immunol, vol.11, pp.63-69, 2004.

J. Nam, A. R. Sharma, L. T. Nguyen, C. Chakraborty, G. Sharma et al., Application of Bioactive Quercetin in Oncotherapy: From Nutrition to Nanomedicine, Mol Basel Switz, vol.21, p.108, 2016.

W. G. Nelson, A. M. De-marzo, T. L. Deweese, and W. B. Isaacs, The role of inflammation in the pathogenesis of prostate cancer, J Urol, vol.172, pp.6-11, 2004.

P. V. Nerurkar, H. A. Schut, L. M. Anderson, C. W. Riggs, L. W. Fornwald et al., Ahr locus phenotype in congenic mice influences hepatic and pulmonary DNA adduct levels of 2amino-3-methylimidazo[4,5-f]quinoline in the absence of cytochrome P450 induction, Mol Pharmacol, vol.49, pp.874-881, 1996.

A. S. Neto, M. Tobias-machado, M. L. Wroclawski, F. L. Fonseca, A. C. Pompeo et al., Molecular oncogenesis of prostate adenocarcinoma: role of the human epidermal growth factor receptor 2 (HER-2/neu), Tumori, vol.96, issue.5, pp.645-649, 2010.

D. J. Newman and G. M. Cragg, Natural products as sources of new drugs over the last 25 years, J Nat Prod, vol.70, issue.3, pp.461-477, 2007.

J. C. Nickel, Prostatitis, Can Urol Assoc J, vol.5, issue.5, pp.306-315, 2011.

S. Noori, An Overview of Oxidative Stress and Antioxidant Defensive System, Sci Rep, vol.1, issue.8, pp.1-9, 2012.

B. Oh, G. Figtree, D. Costa, T. Eade, G. Hruby et al., Oxidative stress in prostate cancer patients: A systematic review of case control studies, Prostate Int, vol.4, issue.3, pp.71-87, 2016.

A. B. Okey, An aryl hydrocarbon receptor odyssey to the shores of toxicology: the Deichmann Lecture, International Congress of Toxicology-XI, Toxicol Sci, vol.98, pp.5-38, 2007.

H. Okhawa, N. Ohishi, and K. Yagi, Assay of lipid peroxides in animal tissue by thiobarbituric reaction, Anal Biochem, vol.95, pp.351-358, 1979.

R. Olinski, T. H. Zastawny, M. Foksinski, A. Barecki, and M. Dizdaroglu, DNA base modifications and antioxidant enzyme activities in human benign prostatic hyperplasia, Free Radic Biol Med, vol.18, pp.807-813, 1995.

A. Oliveira, C. S. Shinobu, R. Longhini, S. L. Franco, and T. I. Svidzinski, Antifungal activity of propolis extract against yeasts isolated from onychomycosis lesions, Mem Inst Oswaldo Cruz, vol.101, pp.493-497, 2006.

C. L. Orsatti, F. Missima, A. C. Pagliarone, T. F. Bachiega, M. C. Búfalo et al., Propolis immunomodulatory action in vivo on Toll-like receptors 2 and 4 expression and on pro-inflammatory cytokines production in mice, Phytother Res, vol.24, pp.1141-1146, 2010.

N. Orsoli?, A. H. Knezevi?, L. Sver, S. Terzi?, and I. Basi?, Immunomodulatory and antimetastatic action of propolis and related polyphenolic compounds, J Ethnopharmacol, vol.94, pp.307-315, 2004.

M. R. Osborne and N. T. Crosby, Benzopyrenes. Cambridge Monographs on Cancer Research, p.352, 1987.

B. Ouyang, C. S. Baxter, H. Lam, S. Yeramaneni, L. Levin et al., Hypomethylation of dual specificity phosphatase 22 promoter correlates with duration of service in firefighters and is inducible by low-dose benzo[a]pyrene, J Occup Environ Med, vol.54, issue.7, pp.774-780, 2012.

X. Ouyang, T. L. Deweese, W. G. Nelson, and C. Abate-shen, Loss-of-function of Nkx3.1 promotes increased oxidative damage in prostate carcinogenesis, Cancer Res, vol.65, pp.6773-6779, 2005.

M. É. Parent, M. Désy, and J. Siemiatycki, Does Exposure to Agricultural Chemicals Increase the Risk of Prostate Cancer among Farmers?, Mcgill J Med, vol.12, issue.1, pp.70-77, 2009.

Y. K. Park, I. Fukuda, H. Ashida, S. Nishiumi, J. P. Guzman et al., Supression of dioxine mediated aryl hydrocarbon receptor transformation by ethanolic extracts of propolis, Biosci Biotechnol Biochem, vol.68, issue.4, pp.935-938, 2003.

F. Pellati, G. Orlandini, D. Pinetti, and S. Benvenuti, HPLC-DAD and HPLC-ESI-MS/MS methods for metabolite profiling of propolis extracts, J Pharm Biomed Anal, vol.55, issue.5, pp.934-948, 2011.

N. D. Patel and J. K. Parsons, Epidemiology and etiology of benign prostatic hyperplasia and bladder outlet obstruction, Indian J Urol, vol.30, issue.2, pp.170-176, 2014.

S. Patel, Emerging Adjuvant Therapy for Cancer: Propolis and its Constituents, J Diet Suppl, vol.13, pp.245-268, 2016.

T. M. Penning, M. E. Burczynski, C. F. Hung, K. D. Mccoull, N. T. Palackal et al., , 1999.

, Dihydrodiol dehydrogenases and polycyclic aromatic hydrocarbon activation: generation of reactive and redox active o-quinones, Chem Res Toxicol, vol.12, issue.1, pp.1-18

J. A. Petros, A. K. Baumann, E. Ruiz-pesini, M. B. Amin, C. Q. Sun et al., mtDNA mutations increase tumorigenicity in prostate cancer, Proc Natl Acad Sci, vol.102, pp.719-724, 2005.
DOI : 10.1073/pnas.0408894102

URL : http://www.pnas.org/content/102/3/719.full.pdf

J. R. Petrulis, A. Kusnadi, P. Ramadoss, B. Hollingshead, and G. H. Perdew, The hsp90 Cochaperone XAP2 alters importin beta recognition of the bipartite nuclear localization signal of the Ah receptor and represses transcriptional activity, J Biol Chem, vol.278, pp.2677-2685, 2003.

D. H. Phillips and P. L. Grover, Polycyclic hydrocarbon activation: bay regions and beyond, Drug Metab Rev, vol.26, pp.443-467, 1994.
DOI : 10.3109/03602539409029808

W. B. Pratt and D. O. Toft, Steroid receptor interactions with heat shock protein and immunophilin chaperones, Endocr Rev, vol.18, pp.306-360, 1997.

D. Price, Comparative Aspects of Development and Structure in the Prostate, Natl Cancer Inst Monogr, vol.12, pp.1-27, 1963.

S. X. Qu and N. H. Stacey, Formation and persistence of DNA adducts in different target tissues of rats after multiple administration of benzo[a]pyrene, Carcinogenesis, vol.17, issue.1, pp.53-59, 1996.

M. M. Rahman, A. Richardson, and M. Sofian-azirun, Antibacterial activity of propolis and honey against Staphylococcus aureus and Escherichia coli, Afr J Microbiol Res, vol.4, issue.18, pp.1872-1878, 2010.

A. Ramesh, F. Inyang, D. B. Hood, A. E. Archibong, M. E. Knuckles et al., , 2001.

B. Metabolism and . Benzo, a)pyrene in F-344 rats following oral administration, Exp Toxic Pathol, vol.53, pp.275-290

N. Rampino, H. Yamamoto, Y. Ionov, Y. Li, H. Sawai et al., Somatic frameshift mutations in the BAX gene in colon cancers of the microsatellite mutator phenotype, Science, vol.275, pp.967-969, 1997.

D. Rea, V. Del-vecchio, G. Palma, A. Barbieri, M. Falco et al., Mouse Models in Prostate Cancer Translational Research: From Xenograft to PDX, Biomed Res Int, p.9750795, 2016.

T. Rengarajan, P. Rajendran, N. Nandakumar, B. Lokeshkumar, P. Rajendran et al., Exposure to polycyclic aromatic hydrocarbons with special focus on cancer, Asian Pac J Trop Biomed, vol.5, issue.3, pp.182-189, 2015.

A. Revel, H. Raanani, E. Younglai, J. Xu, I. Rogers et al., , 2003.

, Resveratrol, a natural aryl hydrocarbon receptor antagonist, protects lung from DNA damage and apoptosiscaused by benzo[a]pyrene, J Appl Toxicol, vol.23, pp.255-261

M. A. Reynolds, Molecular alterations in prostate cancer, Cancer Lett, vol.271, issue.1, pp.13-24, 2008.

M. Rieken, S. F. Shariat, L. A. Kluth, H. Fajkovic, M. Rink et al., Association of Cigarette Smoking and Smoking Cessation with Biochemical Recurrence of Prostate Cancer in Patients Treated with Radical Prostatectomy, Eur Urol, vol.68, pp.949-956, 2015.

S. M. Rizk, H. F. Zaki, and M. Mina, Propolis attenuates doxorubicin-induced testicular toxicity in rats, Food Chem Toxicol, vol.67, pp.176-186, 2014.

D. Robinson, E. M. Van-allen, Y. M. Wu, N. Schultz, R. J. Lonigro et al., Integrative clinical genomics of advanced prostate cancer. Cell, vol.161, pp.1215-1228, 2015.

K. F. Roby, Endocrine disruptors, Ovarian Toxicology, pp.153-169, 2014.

S. Rohrmann, J. Linseisen, N. Allen, H. B. Bueno-de-mesquita, N. F. Johnsen et al., Smoking and the risk of prostate cancer in the European Prospective Investigation into Cancer and Nutrition, Br J Cancer, vol.108, pp.708-714, 2013.

R. Zand, R. S. Jenkins, D. Diamandis, and E. P. , Flavonoids and steroid hormonedependent cancers, J Chromatogr B Analyt Technol Biomed Life Sci, vol.777, pp.219-232, 2002.

J. Ross, G. Nelson, A. Kligerman, G. Erexson, M. Bryant et al., Formation and persistence of novel benzo(a)pyrene adducts in rat lung, liver, and peripheral blood lymphocyte DNA, Cancer Res, vol.50, issue.16, pp.5088-5094, 1990.

E. T. Ruijter, C. A. Van-de-kaa, J. A. Schalken, F. M. Debruyne, and D. J. Ruiter, Histological grade heterogeneity in multifocal prostate cancer. Biological and clinical implications, J Pathol, vol.180, pp.295-299, 1996.

A. Russo, V. Cardile, F. Sanchez, N. Troncoso, A. Vanella et al., Chilean propolis: antioxidant activity and antiproliferative action in human tumor cell lines, Life Sci, vol.76, issue.5, pp.545-558, 2004.

B. A. Rybicki, C. Neslund-dudas, N. L. Nock, L. R. Schultz, L. Eklund et al., Prostate cancer risk from occupational exposure to polycyclic aromatic hydrocarbons interacting with the GSTP1 Ile105Val polymorphism, Cancer Detect Prev, vol.30, issue.5, pp.412-422, 2006.

B. Sadikovic and D. I. Rodenhiser, Benzopyrene exposure disrupts DNA methylation and growth dynamics in breast cancer cells, Toxicol Appl Pharmacol, vol.216, pp.458-468, 2006.

S. Safe, S. O. Lee, and U. H. Jin, Role of the Aryl Hydrocarbon Receptor in Carcinogenesis and Potential as a Drug Target, Toxicol Sci, vol.135, issue.1, pp.1-16, 2013.

M. Salemi, R. A. Condorelli, L. Vignera, S. Barone, N. Ridolfo et al., PARP-1 and CASP3 genes are up-regulated in LNCaP and PC-3 prostate cancer cell lines, Human Cell, vol.27, pp.172-175, 2014.

E. I. Salim, A. El-magid, A. D. Farara, K. M. Maria, and D. Sm, Antitumoral and Antioxidant Potential of Egyptian Propolis Against the PC3 Prostate Cancer Cell Line, Asian Pac J Cancer Prev, vol.16, issue.17, pp.7641-7651, 2015.

S. Samarghandian, J. T. Afshari, and S. Davoodi, Chrysin reduces proliferation and induces apoptosis in the human prostate cancer cell line pc-3, Clin Sao Paulo Braz, vol.66, pp.1073-1079, 2011.

D. A. Sampietro, S. Vattuone, M. M. Vattuone, and M. A. , Immunomodulatory activity of Apis mellifera propolis from the North of Argentina, LWT-Food Sci Technol, vol.70, pp.9-15, 2016.

M. G. Sanda, T. H. Beaty, R. E. Stutzman, B. Childs, and P. C. Walsh, Genetic susceptibility of benign prostatic hyperplasia, J Urol, vol.152, pp.115-119, 1994.

J. T. Sanderson, J. Hordijk, M. S. Denison, M. F. Springsteel, M. H. Nantz et al., Induction and inhibition of aromatase (CYP19) activity by natural and synthetic flavonoid compounds in H295R human adrenocortical carcinoma cells, Toxicol Sci, vol.82, pp.70-79, 2004.

M. Savas, Oxidative stress in benign prostate hyperplasia, Studies on Men's Health and Fertility, p.672, 2012.

N. Sawada, M. Inoue, M. Iwasaki, S. Sasazuki, T. Yamaji et al., Alcohol and smoking and subsequent risk of prostate cancer in Japanese men: The Japan Public Health Center-based prospective study, Int J Cancer, vol.134, pp.971-978, 2013.

M. Schnekenburger, L. Peng, and A. Puga, HDAC1 bound to the Cyp1a1 promoter blocks histone acetylation associated with Ah receptor-mediated trans-activation, Biochim Biophys Acta, vol.1769, pp.569-578, 2007.

F. H. Schröder, J. Hugosson, M. J. Roobol, T. Tammela, S. Ciatto et al., Screening and prostate-cancer mortality in a randomized European study, N Engl J Med, vol.360, pp.1320-1328, 2009.

A. Sciarra, D. Silverio, F. Salciccia, S. , A. Gomez et al., Inflammation and chronic prostatic diseases: evidence for a link?, Eur Urol, vol.52, pp.964-972, 2007.

N. Segueni, A. A. Magid, M. Decarme, S. Rhouati, M. Lahouel et al., Inhibition of stromelysin-1 by caffeic acid derivatives from a propolis sample from Algeria, Planta Med, vol.77, pp.999-1004, 2011.

S. Sen, P. Bhojnagarwala, L. Francey, D. Lu, J. Field et al., Chem Res Toxicol, vol.25, issue.10, pp.2117-2126, 2012.

E. C. Serefoglu, S. Altinova, N. S. Ugras, E. Akincioglu, E. Asil et al., How reliable is 12-core prostate biopsy procedure in the detection of prostate cancer?, Can Urol Assoc J, vol.7, issue.5-6, pp.293-298, 2013.

J. M. Sforcin and V. Bankova, Propolis: is there a potential for the development of new drugs?, J Ethnopharmacol, vol.133, issue.2, pp.253-260, 2011.

J. M. Sforcin, Propolis and the immune system: a review, J Ethnopharmacol, vol.113, pp.1-14, 2007.

J. M. Sforcin, Biological Properties and Therapeutic Applications of Propolis, Phytother Res, vol.30, pp.894-905, 2016.

A. A. Shafi, A. E. Yen, and N. L. Weigel, Androgen receptors in hormone-dependent and castration-resistant prostate cancer, Pharmacol Therap, vol.140, pp.223-238, 2013.

S. F. Shariat, M. Shalev, A. Menesses-diaz, I. Y. Kim, M. W. Kattan et al., Preoperative plasma levels of transforming growth factor beta (1) (TGF-beta(1)) stronglypredict progression in patients undergoing radical prostatectomy, J Clin Oncol, vol.19, issue.11, pp.2856-2864, 2001.

M. Sharma, W. W. Chuang, and Z. Sun, Phosphatidylinositol 3-kinase/Akt stimulates androgen pathway through GSK3beta inhibition and nuclear beta-catenin accumulation, J Biol Chem, vol.277, issue.34, pp.30935-30941, 2002.

D. Sheehan, G. Meade, V. M. Foley, and C. A. Dowd, Structure, function and evolution of glutathione transferases: implications for classification of non-mammalian members of an ancient enzyme superfamily, Biochem J, vol.360, issue.1, pp.1-16, 2001.

K. Shen, S. Hung, L. Yin, C. Huang, C. Chao et al., Acacetin, a flavonoid, inhibits the invasion and migration of human prostate cancer DU145 cells via inactivation of the p38 MAPK signaling pathway, Mol Cell Biochem, vol.333, pp.279-291, 2010.

M. M. Shen and C. Abate-shen, Molecular genetics of prostate cancer: new prospects for old challenges, Genes Dev, vol.24, pp.1967-2000, 2010.

T. Shimada, Xenobiotic-metabolizing enzymes involved in activation and detoxification of carcinogenic polycyclic aromatic hydrocarbons, Drug Metab Pharmacokinet, vol.21, issue.4, pp.257-276, 2006.

Y. Shimizu, Y. Nakatsuru, M. Ichinose, Y. Takahashi, H. Kume et al., Benzo[a] pyrene carcinogenicity is lost in mice lacking the aryl hydrocarbon receptor, PNAS, vol.97, issue.2, pp.779-782, 2000.

S. Shum, N. M. Jensen, and D. W. Nebert, The murine Ah locus: in utero toxicity and teratogenesis associated with genetic differences in benzo[a]pyrene metabolism, Teratology, vol.20, pp.365-376, 1979.

B. B. Silva, P. L. Rosalen, J. A. Cury, M. Ikegaki, V. C. Souza et al., Chemical composition and botanical origin of red propolis, a new type of brazilian propolis, Evid-Based Complement Altern Med, vol.5, pp.313-316, 2008.

T. Simoncini and A. R. Genazzani, Non-genomic actions of sex steroid hormones, Eur J Endocrinol, vol.148, issue.3, pp.281-292, 2003.

M. Simone-finstrom and M. Spivak, Propolis and bee health: the natural history and significance of resin use by honey bees, Apidologie, vol.41, issue.3, pp.295-311, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00892097

R. J. Simpson, Benign prostate hyperplasia, Br J Gen Pract, vol.47, pp.235-240, 1997.

S. M. Singh, S. Gauthier, and F. Labrie, Androgen receptor antagonists (antiandrogens): structure-activity relationships, Curr Med Chem, vol.7, pp.211-247, 2000.

R. Sinha, Y. Park, B. I. Graubard, M. F. Leitzmann, A. Hollenbeck et al., Meat and Meat-related Compounds and Risk of Prostate Cancer in a Large Prospective Cohort Study in the United States, Am J Epidemiol, vol.170, issue.9, pp.1165-1177, 2009.

R. E. Sobel and M. D. Sadar, Cell lines used in prostate cancer research: a compendium of old and new lines-Part 2, J Urol, vol.173, pp.360-372, 2005.

M. Soda, D. Hu, S. Endo, M. Takemura, J. Li et al., Design, synthesis and evaluation of caffeic acid phenethyl ester-based inhibitors targeting a selectivity pocket in the active site of human aldo-keto reductase 1B10, Eur J Med Chem, vol.48, pp.321-329, 2012.

V. L. Sparnins, A. W. Mott, G. Barany, and L. W. Wattenberg, Effects of allyl methyl trisulfide on glutathione S-transferase activity and BP-induced neoplasia in the mouse, Nutr Cancer, vol.8, pp.211-215, 1986.

V. Stambolic, A. Suzuki, J. L. De-la-pompa, G. M. Brothers, C. Mirtsos et al., Negative regulation of PKB/ Akt-dependent cell survival by the tumor suppressor PTEN, Cell, vol.95, pp.29-39, 1998.

K. M. Sterling and K. R. Cutroneo, Constitutive and inducible expression of cytochromes P4501A (CYP1A1 and CYP1A2) in normal prostate and prostate cancer cells, J Cell Biochem, vol.91, issue.2, pp.423-429, 2004.

D. W. Strand and A. S. Goldstein, The many ways to make a luminal cell and a prostate cancer cell, Endocr Relat Cancer, vol.22, issue.6, pp.187-197, 2015.

T. Su and D. J. Waxman, Impact of dimethyl sulfoxide on expression of nuclear receptors and drug-inducible cytochromes P450 in primary rat hepatocytes, Arch Biochem Biophys, vol.424, issue.2, pp.226-234, 2004.

Y. Sugimura, G. R. Cunha, and A. A. Donjacour, Morphogenesis of ductal networks in the mouse prostate, Biol Reprod, vol.34, issue.5, pp.961-971, 1986.

J. Suh, F. Payvandi, L. C. Edelstein, P. S. Amenta, W. X. Zong et al., Mechanisms of constitutive NF-kappaB activation in human prostate cancer cells, Prostate, vol.52, issue.3, pp.183-200, 2002.

E. Szliszka, Z. P. Czuba, J. Bronikowska, A. Mertas, A. Paradysz et al., Ethanolic Extract of Propolis Augments TRAIL-Induced Apoptotic Death in Prostate Cancer Cells. EvidBased, Complement Altern Med, p.535172, 2011.

E. Szliszka, Z. P. Czuba, J. Bronikowska, A. Mertas, A. Paradysz et al., Ethanolic Extract of Propolis Augments TRAIL-Induced Apoptotic Death in Prostate Cancer Cells. EvidBased, Complement Altern Med, vol.535172, p.11, 2009.

E. Szliszka, G. Zydowicz, B. Janoszka, C. Dobosz, G. Kowalczyk-ziomek et al., Ethanolic extract of Brazilian green propolis sensitizes prostate cancer cells to TRAIL-induced apoptosis, Int J Oncol, vol.38, pp.941-953, 2011.

N. N. Tam, Y. Gao, Y. K. Leung, and S. M. Ho, Androgenic regulation of oxidative stress in the rat prostate: involvement of NAD(P)H oxidases and antioxidant defense machinery during prostatic involution and regrowth, Am J Pathol, vol.163, pp.2513-2522, 2003.

M. H. Tan, J. Li, H. E. Xu, K. Melcher, and E. L. Yong, Androgen receptor: structure, role in prostate cancer and drug discovery, Acta Pharmacol Sin, vol.36, issue.1, pp.3-23, 2015.

D. G. Tang and A. T. Porter, Target to apoptosis: a hopeful weapon for prostate cancer, Prostate, vol.32, issue.4, pp.284-293, 1997.

X. Tekpli, E. Rivedal, M. Gorria, N. E. Landvik, M. Rissel et al., The B[a]P-increased intercellular communication via translocation of connexin-43 into gap junctions reduces apoptosis, Toxicol Appl Pharmacol, vol.242, pp.231-240, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00660130

X. Tekpli, S. Zienolddiny, V. Skaug, L. Stangeland, A. Haugen et al., DNA methylation of the CYP1A1 enhancer is associated with smoking-induced genetic alterations in human lung, Int J Cancer, vol.131, pp.1509-1516, 2012.

P. Tenke, J. Horti, P. Balint, and B. Kovacs, Prostate cancer screening, In: Prostate cancer, pp.65-81, 2007.

D. Thapa and R. Ghosh, Antioxidants for prostate cancer chemoprevention: Challenges and opportunities, Biochem Pharmacol, vol.83, pp.1319-1330, 2012.

J. L. Thielen, K. G. Volzing, L. S. Collier, L. E. Green, D. A. Largaespada et al., Markers of prostate region-specific epithelial identity define anatomical locations in the mouse prostate that are molecularly similar to human prostate cancers, Differentiation, vol.75, pp.49-61, 2007.

T. C. Thompson and G. Yang, Regulation of apoptosis in prostatic disease, Prostate Suppl, vol.9, pp.25-28, 2000.

J. Thyssen, J. Althoff, G. Kimmerle, and U. Mohr, Inhalation studies with benzo[a]pyrene in Syrian golden hamsters, J Natl Cancer Inst, vol.66, issue.3, pp.575-577, 1981.

C. Tran, O. Richmond, L. Aaron, and J. B. Powell, Inhibition of constitutive aryl hydrocarbon receptor (AhR) signaling attenuates androgen independent signaling and growth in (C4-2) prostate cancer cells, Biochem Pharmacol, vol.85, pp.753-762, 2013.

R. Trivedi and D. P. Mishra, Trailing TRAIL Resistance: Novel Targets for TRAIL Sensitization in Cancer Cells, Front Oncol, vol.5, p.69, 2015.

I. Turan, S. Demir, S. Misir, K. Kilinc, A. Mentese et al., Cytotoxic Effect of Turkish Propolis on Liver, Colon, Breast, Cervix and Prostate Cancer Cell Lines, Trop J Pharm Res, vol.14, issue.5, pp.777-782, 2015.
DOI : 10.4314/tjpr.v14i5.5

URL : https://www.ajol.info/index.php/tjpr/article/download/122908/112450

U. K. Udensi and P. B. Tchounwou, Oxidative stress in prostate hyperplasia and carcinogenesis, J Exp Clin Cancer Res, vol.35, issue.1, p.139, 2016.
DOI : 10.1186/s13046-016-0418-8

URL : https://jeccr.biomedcentral.com/track/pdf/10.1186/s13046-016-0418-8

M. J. Valente, A. F. Baltazar, R. Henrique, L. Estevinho, and M. Carvalho, Biological activities of Portuguese propolis: protection against free radical-induced erythrocyte damage and inhibition of human renal cancer cell growth in vitro, Food Chem Toxicol, vol.49, pp.86-92, 2011.

M. Valko, C. J. Rhodes, J. Moncol, M. Izakovic, and M. Mazur, Free radicals, metals and antioxidants in oxidative stress-induced cancer, Chem Biol Interact, vol.160, pp.1-40, 2006.
DOI : 10.1016/j.cbi.2005.12.009

I. W. Van-der-cruijsen-koeter, A. N. Vis, M. J. Roobol, M. F. Wildhagen, D. Koning et al., Comparison of screen detected and clinically diagnosed prostate cancer in the European randomized study of screening for prostate cancer, J Urol, vol.174, issue.1, pp.121-125, 2005.

M. E. Van-royen, W. A. Van-cappellen, C. De-vos, A. B. Houtsmuller, and J. Trapman, Stepwise androgen receptor dimerization, J Cell Sci, vol.125, pp.1970-1979, 2012.

J. Veldscholte, C. Ris-stalpers, G. G. Kuiper, G. Jenster, C. Berrevoets et al., A mutation in the ligand binding domain of the androgen receptor of human LNCaP cells affects steroid binding characteristics and response to anti-androgens, Biochem Biophys Res Commun, vol.173, issue.2, pp.534-540, 1990.

P. Verze, T. Cai, and S. Lorenzetti, The role of the prostate in male fertility, health and disease, Nat Rev Urol, vol.13, issue.7, pp.379-386, 2016.

C. A. Vogel and T. Haarmann-stemmann, The aryl hydrocarbon receptor repressor-More than a simple feedback inhibitor of AhR signaling: Clues for its role in inflammation and cancer, Curr Opin Toxicol, vol.2, pp.109-119, 2017.

F. E. Wagenlehner and K. G. Naber, Current challenges in the treatment of complicated urinary tract infections and prostatitis, Clin Microbiol Infect, vol.12, issue.3, pp.67-80, 2006.

Y. Wang, R. Cui, Y. Xiao, J. Fang, and Q. Xu, Effect of carotene and lycopene on the risk of prostate cancer: a systematic review and dose-response meta-analysis of observational studies, PLoS One, vol.10, issue.9, p.137427, 2015.

M. Watanabe, M. K. Amarante, B. J. Conti, and J. M. Sforcin, Cytotoxic constituents of propolis inducing anticancer effects: a review, J Pharm Pharmacol, vol.63, pp.1378-1386, 2011.
DOI : 10.1111/j.2042-7158.2011.01331.x

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1111/j.2042-7158.2011.01331.x

P. W. Wester, J. Muller, W. Slob, G. R. Mohn, P. M. Dortant et al., Carcinogenic activity of benzo[a]pyrene in a 2 year oral study in Wistar rats, Food Chem Toxicol, vol.50, issue.3-4, pp.927-935, 2012.

E. H. Weyand and D. R. Bevan, Benzo(a)pyrene disposition and metabolism in rats following intratracheal instillation, Cancer Res, vol.46, pp.5655-5661, 1986.
DOI : 10.1016/0304-3835(87)90086-3

A. M. Williams, I. Simon, P. K. Landis, C. Moser, W. Christens-barry et al., Prostatic growth rate determined from MRI data: Age-related longitudinal changes, J Androl, vol.20, pp.474-480, 1999.
DOI : 10.1097/00005392-199904020-00155

C. P. Wong, T. M. Bray, and E. Ho, Induction of proinflammatory response in prostate cancer epithelial cells by activated macrophages, Cancer Lett, vol.276, pp.38-46, 2009.

D. Wu and A. I. Cederbaum, Alcohol, oxidative stress, and free radical damage, Alcohol Res Health, vol.27, issue.4, pp.277-284, 2003.

W. Xiao, D. R. Hodge, L. Wang, X. Yang, X. Zhang et al., Co-operative functions between nuclear factors NFkappaB and CCAT/enhancer-binding protein-beta (C/EBP-beta) regulate the IL-6 promoter in autocrine human prostate cancer cells, Prostate, vol.61, issue.4, pp.354-370, 2004.
DOI : 10.1002/pros.20113

N. Xing, Y. Chen, S. H. Mitchell, and C. Y. Young, Quercetin inhibits the expression and function of the androgen receptor in LNCaP prostate cancer cells, Carcinogenesis, vol.22, issue.3, pp.409-414, 2001.

C. Xu, J. Chen, Z. Qiu, Q. Zhao, J. Luo et al., Ovotoxicity and PPAR-mediated aromatase downregulation in female Sprague-Dawley rats following combined oral exposure to benzo, 2010.
DOI : 10.1016/j.toxlet.2010.09.015

, Toxicol Lett, vol.199, pp.323-332

J. Xu, R. C. Wu, and B. W. Malley, Normal and cancer-related functions of the p160 steroid receptor co-activator (SRC) family, Nat Rev Cancer, vol.9, pp.615-630, 2009.

W. Xue and D. Warshawsky, Metabolic activation of polycyclic and heterocyclic aromatic hydrocarbons and DNA damage: a review, Toxicol Appl Pharmacol, vol.206, pp.73-93, 2005.

M. Yamashita, X. Zhang, T. Shiraishi, H. Uetsuki, and Y. Kakehi, Determination of percent area density of epithelial and stromal components in development of prostatic hyperplasia in spontaneously hypertensive rats, Urology, vol.61, pp.484-489, 2003.

C. C. Yang, S. S. Sun, C. Y. Lin, F. J. Chuang, and C. H. Kao, Differentiation of prostate cancer and benign prostatic hyperplasia: the clinical value of 201Tl SPECT-a pilot study, Ann Nucl Med, vol.17, issue.7, pp.521-524, 2003.

Y. Yang, J. Rafter, J. A. Gustafsson, J. Sjövall, and W. J. Griffiths, Analysis of the major mercapturic acid pathway metabolites of benzo[a]pyrene found in rat urine by nano-electrospray mass spectrometry, Rapid Commun Mass Spectrom, vol.12, issue.8, pp.465-471, 1998.

G. W. Yardy and S. F. Brewster, Wnt signalling and prostate cancer, Prostate Cancer Prostatic Dis, vol.8, issue.2, pp.119-126, 2005.

H. Yuan, A. Gong, and C. Y. Young, Involvement of transcription factor Sp1 in quercetinmediated inhibitory effect on the androgen receptor in human prostate cancer cells, Carcinogenesis, vol.26, issue.4, pp.793-801, 2005.

N. Zabaiou, A. Fouache, A. Trousson, S. Baron, A. Zellagui et al., Biological properties of propolis extracts: Something new from an ancient product, Chemistry and Physics of Lipids, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01939753

N. Zabaiou, D. Mabed, J. M. Lobaccaro, and M. Lahouel, Oxidative stress in benign prostate hyperplasia, Andrologia, vol.48, issue.1, pp.69-73, 2015.
URL : https://hal.archives-ouvertes.fr/tel-01930319

D. F. Zapata, L. E. Howard, W. J. Aronson, C. J. Kane, M. K. Terris et al., Smoking is a predictor of adverse pathological features at radical prostatectomy: Results from the Shared Equal Access Regional Cancer Hospital database, Int J Urol, vol.22, pp.658-662, 2015.

E. Zhang, R. Wang, S. Guo, and B. Liu, An update on antitumor activity of naturally occurring chalcones. Evid-Based, Complement Altern Med, p.815621, 2013.

L. Zhang, M. Huang, I. A. Blair, and T. M. Penning, Detoxication of benzo[a]pyrene-7,8-dione by sulfotransferases (SULTs) in human lung cells, J Biol Chem, vol.287, issue.35, pp.29909-29920, 2012.

L. Zhang, Y. Jin, M. Huang, and T. M. Penning, The role of human aldo-keto reductases in the metabolic activation and detoxication of polycyclic aromatic hydrocarbons: interconversion of PAH catechols and PAH o-quinones, Front Pharmacol, vol.3, issue.193, pp.1-12, 2012.

W. Zhang, Y. Lan, Q. Huang, and Z. Hua, Galangin induces B16F10 melanoma cell apoptosis via mitochondrial pathway and sustained activation of p38 MAPK, Cytotechnology, vol.65, pp.447-455, 2013.

W. Zhang, B. Tang, Q. Huang, and Z. Hua, Galangin inhibits tumor growth and metastasis of B16F10 melanoma, J Cell Biochem, vol.114, pp.152-161, 2013.

W. D. Zhong, J. Peng, H. C. He, D. Wu, Z. D. Han et al., Ki-67 and PCNA expression in prostate cancer and benign prostatic hyperplasia, Clin Invest Med, vol.31, issue.1, pp.8-15, 2008.

Y. Zhou, E. C. Bolton, and J. O. Jones, Androgens and androgen receptor signaling in prostate tumorigenesis, J Mol Endocrinol, vol.54, issue.1, pp.15-29, 2015.

M. Zhu and W. E. Fahl, Functional characterization of transcription regulators that interact with the electrophile response element, Biochem Biophys Res Commun, vol.289, pp.212-219, 2001.

M. L. Zhu, C. M. Horbinski, M. Garzotto, D. Z. Qian, T. M. Beer et al., Tubulintargeting chemotherapy impairs androgen receptor activity in prostate cancer, Cancer Res, vol.70, pp.7992-8002, 2010.