G. Cragg and D. Newman, Plants as a source of anti-cancer agents, Journal of Ethnopharmacology, vol.100, issue.1-2, pp.72-79, 2005.
DOI : 10.1016/j.jep.2005.05.011

V. Srivastava, A. Negi, J. Kumar, M. Gupta, and S. Khanuja, Plant-based anticancer molecules: A chemical and biological profile of some important leads, Bioorganic & Medicinal Chemistry, vol.13, issue.21, pp.5892-5908, 2005.
DOI : 10.1016/j.bmc.2005.05.066

T. Sharif, M. Alhosin, C. Auger, C. Minker, J. Kim et al., Aronia melanocarpa Juice Induces a Redox-Sensitive p73-Related Caspase 3-Dependent Apoptosis in Human Leukemia Cells, PLoS ONE, vol.45, issue.3, p.32526
DOI : 10.1371/journal.pone.0032526.t001

URL : https://hal.archives-ouvertes.fr/hal-00684449

R. Ali, Z. Mirza, G. Ashraf, M. Kamal, S. Ansari et al., New anticancer agents: recent developments in tumor therapy, pp.2999-3005, 2012.

P. Russo, D. Bufalo, A. Cesario, and A. , Flavonoids Acting on DNA Topoisomerases: Recent Advances and Future Perspectives in Cancer Therapy, Current Medicinal Chemistry, vol.19, issue.31, pp.5287-5293
DOI : 10.2174/092986712803833272

J. Lee, E. Ko, J. Cho, H. Park, J. Lee et al., Methylation and immunoexpression of p16(INK4a) tumor suppressor gene in primary Breast Cancer tissue and their quantitative p16(INK4a) Hypermethylation in plasma by eeal-time PCR, pp.554-561

J. Herman, A. Merlo, L. Mao, R. Lapidus, J. Issa et al., Inactivation of the CDKN2/p16/MTS1 gene is frequently associated with aberrant DNA methylation in all common human cancers, Cancer Res, vol.55, pp.4525-4530, 1995.

M. Serrano, The Tumor Suppressor Protein p16INK4a, Experimental Cell Research, vol.237, issue.1, pp.7-13, 1997.
DOI : 10.1006/excr.1997.3824

G. Avvakumov, J. Walker, S. Xue, Y. Li, S. Duan et al., Structural basis for recognition of hemi-methylated DNA by the SRA domain of human UHRF1, Nature, vol.39, issue.7214, pp.822-825, 2008.
DOI : 10.1038/nature07273

URL : https://hal.archives-ouvertes.fr/hal-00339497

C. Bronner, M. Achour, Y. Arima, T. Chataigneau, H. Saya et al., The UHRF family: Oncogenes that are drugable targets for cancer therapy in the near future?, Pharmacology & Therapeutics, vol.115, issue.3, pp.419-434, 2007.
DOI : 10.1016/j.pharmthera.2007.06.003

URL : https://hal.archives-ouvertes.fr/hal-00176128

M. Bostick, J. Kim, P. Esteve, A. Clark, S. Pradhan et al., UHRF1 Plays a Role in Maintaining DNA Methylation in Mammalian Cells, Science, vol.317, issue.5845, pp.1760-1764, 2007.
DOI : 10.1126/science.1147939

H. Chen, H. Ma, H. Inuzuka, J. Diao, F. Lan et al., DNA Damage regulates UHRF1 Stability via the SCFbeta-TrCP E3 Ligase, Mol Cell Biol, vol.2013, issue.336, pp.1139-1148

M. Muto, Y. Kanari, E. Kubo, T. Takabe, T. Kurihara et al., Targeted Disruption of Np95 Gene Renders Murine Embryonic Stem Cells Hypersensitive to DNA Damaging Agents and DNA Replication Blocks, Journal of Biological Chemistry, vol.277, issue.37
DOI : 10.1074/jbc.M205189200

X. Li, Q. Meng, and S. Fan, Adenovirus-mediated expression of UHRF1 reduces the radiosensitivity of cervical cancer HeLa cells to ??-irradiation, Acta Pharmacologica Sinica, vol.153, issue.4, pp.458-466, 2009.
DOI : 10.1158/1535-7163.MCT-05-0193

H. Mistry, L. Tamblyn, H. Butt, D. Sisgoreo, A. Gracias et al., UHRF1 is a genome caretaker that facilitates the DNA damage response to ??-irradiation, Genome Integrity, vol.1, issue.1, p.7, 2010.
DOI : 10.1186/2041-9414-1-7

F. Wang, Y. Yang, C. Shi, P. Zhang, M. Moyer et al., UHRF1 promotes cell growth and metastasis through repression of p16(ink(4)a) in colorectal cancer, Ann Surg Oncol, vol.2012, issue.8, pp.192753-2762

M. Vaid, R. Prasad, T. Singh, V. Jones, and S. Katiyar, Grape seed proanthocyanidins reactivate silenced tumor suppressor genes in human skin cancer cells by targeting epigenetic regulators, Toxicology and Applied Pharmacology, vol.263, issue.1, pp.122-130
DOI : 10.1016/j.taap.2012.06.013

M. Achour, M. Mousli, M. Alhosin, I. A. Peluso, J. Muller et al., Epigallocatechin-3-gallate upregulates tumor suppressor gene expression via a reactive oxygen species-dependent down-regulation of UHRF1, Biochem Biophys Res Commun, vol.2012, issue.4301, pp.208-212
URL : https://hal.archives-ouvertes.fr/hal-00789025

L. Floch and E. , Contribution à une etude ethnobotanique de la flore tunisienne Tunisie: Tunis: ministere de l'enseignement superieur et de la recherche scientifique, p.192, 1983.

N. Belboukhari and A. Cheriti, Analysis and isolation of saponins from Limoniastrum feei by LC-UV, Chemistry of Natural Compounds, vol.48, issue.2, pp.756-758, 2009.
DOI : 10.1007/s10600-009-9455-2

N. Belboukhari and A. Cheriti, Anti-microbial activity of aerial part crude extracts from Limoniastrum feei, Asian J of Plant Sci, vol.4, pp.496-498, 2005.

M. Krifa, I. Bouhlel, L. Ghedira-chekir, and K. Ghedira, Immunomodulatory and cellular anti-oxidant activities of an aqueous extract of Limoniastrum guyonianum gall, Journal of Ethnopharmacology, vol.146, issue.1, pp.243-249, 2013.
DOI : 10.1016/j.jep.2012.12.038

M. Chaabi, N. Beghidja, S. Benayache, and A. Lobstein, Abstract, Zeitschrift f??r Naturforschung C, vol.63, issue.11-12, pp.801-807, 2008.
DOI : 10.1515/znc-2008-11-1204

N. Trabelsi, S. Oueslati, H. Falleh, P. Waffo-teguo, Y. Papastamoulis et al., Isolation of powerful antioxidants from the medicinal halophyte Limoniastrum guyonianum, Food Chemistry, vol.135, issue.3, pp.1419-1424
DOI : 10.1016/j.foodchem.2012.05.120

F. Lemarie, C. Chang, D. Blatchford, R. Amor, G. Norris et al., Antitumor activity of the tea polyphenol epigallocatechin-3-gallate encapsulated in targeted vesicles after intravenous administration, Nanomedicine, vol.8, issue.2, pp.181-192, 2013.
DOI : 10.2217/nnm.12.83

G. Li, Y. Chen, Z. Hou, H. Xiao, J. H. Lu et al., Pro-oxidative activities and dose-response relationship of (-)-epigallocatechin-3-gallate in the inhibition of lung cancer cell growth: a comparative study in vivo and in vitro, Carcinogenesis, vol.31, issue.5, pp.31902-910, 2010.
DOI : 10.1093/carcin/bgq039

F. Sun, X. Zheng, J. Ye, T. Wu, J. Wang et al., Potential Anticancer Activity of Myricetin in Human T24 Bladder Cancer Cells Both In Vitro and In Vivo, Nutrition and Cancer, vol.4, issue.4, pp.599-606
DOI : 10.1016/S0014-5793(96)01442-1

C. Liang, X. Zhang, H. Li, Y. Tao, L. Tao et al., Gallic acid induces the apoptosis of human osteosarcoma cells in vitro and in vivo via the regulation of mitogen-activated protein kinase pathways, Cancer Biother Radiopharm, vol.2012, issue.10, pp.27701-710

G. Pottier-alapetite, Flore de laTunisie: angiospermes, dicotyledones, apetales, dialypetales, tunisie: ministère de l'enseignement supérieur et de la recherche scientifique et ministère de l'agriculture. Tunisia, p.210, 1979.

S. Chattopadhyay and S. Kumar, Identification and quantification of two biologically active polyisoprenylated benzophenones xanthochymol and isoxanthochymol in Garcinia species using liquid chromatography???tandem mass spectrometry, Journal of Chromatography B, vol.844, issue.1, pp.67-83, 2006.
DOI : 10.1016/j.jchromb.2006.07.045

Y. Yuan, D. Bone, and M. Carrington, Antioxidant activity of dulse (Palmaria palmata) extract evaluated in vitro, Food Chemistry, vol.91, issue.3, pp.485-494, 2005.
DOI : 10.1016/j.foodchem.2004.04.039

J. Zhishen, T. Mengcheng, and W. Jianming, The determination of flavonoid contents in mulberry and their scavenging effects on superoxide radicals, Food Chemistry, vol.64, issue.4, pp.555-559, 1999.
DOI : 10.1016/S0308-8146(98)00102-2

D. Pearson, The chemical analysis of foods, 1976.

T. Nwabueze, Effect of process variables on trypsin inhibitor activity (TIA) phytic acid and tannin content of extruded African bread fruitcorn-soy mixtures: a response surface analysis, pp.21-29, 2007.

M. Achour, X. Jacq, P. Ronde, M. Alhosin, C. Charlot et al., The interaction of the SRA domain of ICBP90 with a novel domain of DNMT1 is involved in the regulation of VEGF gene expression, Oncogene, vol.8, issue.15, pp.2187-2197, 2008.
DOI : 10.1006/excr.1999.4743

URL : https://hal.archives-ouvertes.fr/hal-00284782

A. Tien, S. Senbanerjee, A. Kulkarni, R. Mudbhary, B. Goudreau et al., /M arrest, activation of DNA damage response and apoptosis, Biochemical Journal, vol.37, issue.1, pp.175-185, 2011.
DOI : 10.1016/j.bbrc.2008.07.146

M. Alhosin, T. Sharif, M. Mousli, N. Etienne-selloum, G. Fuhrmann et al., Down-regulation of UHRF1, associated with re-expression of tumor suppressor genes, is a common feature of natural compounds exhibiting anti-cancer properties, Journal of Experimental & Clinical Cancer Research, vol.30, issue.1, p.3041, 2011.
DOI : 10.1158/1078-0432.CCR-10-1260

URL : https://hal.archives-ouvertes.fr/hal-00666930

A. Abusnina, T. Keravis, I. Yougbare, C. Bronner, and C. Lugnier, Anti-proliferative effect of curcumin on melanoma cells is mediated by PDE1A inhibition that regulates the epigenetic integrator UHRF1, Molecular Nutrition & Food Research, vol.19, issue.11, pp.551677-1689, 2011.
DOI : 10.1002/mnfr.201100307

URL : https://hal.archives-ouvertes.fr/hal-00667387

J. Choi, J. Kim, J. Lee, C. Kang, H. Kwon et al., Induction of cell cycle arrest and apoptosis in human breast cancer cells by quercetin, International Journal of Oncology, vol.19, pp.837-844, 2001.
DOI : 10.3892/ijo.19.4.837

C. Mu, P. Jia, Z. Yan, X. Liu, X. Li et al., Quercetin induces cell-cycle G1 arrest through elevating Cdk inhibitors p21 and p27 in human hepatoma cell line (HepG2), Methods and Findings in Experimental and Clinical Pharmacology, vol.29, issue.3, pp.179-183, 2007.
DOI : 10.1358/mf.2007.29.3.1092095

M. Ujiki, X. Ding, M. Salabat, D. Bentrem, L. Golkar et al., APIGENIN INHIBITS HUMAN PANCREATIC CANCER CELL PROLIFERATION THROUGH G2/M CELL CYCLE ARREST., Pancreas, vol.29, issue.4, p.76, 2006.
DOI : 10.1097/00006676-200411000-00046

F. Xie, Q. Lang, M. Zhou, H. Zhang, Z. Zhang et al., The dietary flavonoid luteolin inhibits Aurora B kinase activity and blocks proliferation of cancer cells, European Journal of Pharmaceutical Sciences, vol.46, issue.5, pp.46388-396, 2012.
DOI : 10.1016/j.ejps.2012.03.002

Q. Zhang, X. Zhao, and Z. Wang, Cytotoxicity of flavones and flavonols to a human esophageal squamous cell carcinoma cell line (KYSE-510) by induction of G2/M arrest and apoptosis, Toxicology in Vitro, vol.23, issue.5, pp.797-807, 2009.
DOI : 10.1016/j.tiv.2009.04.007

M. Alhosin, A. Abusnina, M. Achour, T. Sharif, C. Muller et al., Induction of apoptosis by thymoquinone in lymphoblastic leukemia Jurkat cells is mediated by a p73-dependent pathway which targets the epigenetic integrator UHRF1, Biochemical Pharmacology, vol.79, issue.9, pp.791251-1260, 2010.
DOI : 10.1016/j.bcp.2009.12.015

URL : https://hal.archives-ouvertes.fr/hal-00507983

L. Boominathan, Some facts and thoughts: p73 as a tumor suppressor gene in the network of tumor suppressors, Molecular Cancer, vol.6, issue.1, p.27, 2007.
DOI : 10.1186/1476-4598-6-27

V. Nandakumar, M. Vaid, and S. Katiyar, (-)-Epigallocatechin-3-gallate reactivates silenced tumor suppressor genes, Cip1/p21 and p16INK4a, by reducing DNA methylation and increasing histones acetylation in human skin cancer cells, Carcinogenesis, vol.32, issue.4, pp.537-544, 2011.
DOI : 10.1093/carcin/bgq285

J. Medina-franco, F. Lopez-vallejo, D. Kuck, and F. Lyko, Natural products as DNA methyltransferase inhibitors: a computer-aided discovery approach, Molecular Diversity, vol.16, issue.2, pp.293-304, 2011.
DOI : 10.1007/s11030-010-9262-5

C. Chiang, T. Way, and J. Lin, Sensitizing HER2-overexpressing cancer cells to luteolin-induced apoptosis through suppressing p21WAF1/CIP1 expression with rapamycin, Molecular Cancer Therapeutics, vol.6, issue.7, pp.2127-2138, 2007.
DOI : 10.1158/1535-7163.MCT-07-0107

C. Davis and E. Uthus, DNA methylation, cancer susceptibility, and nutrient interactions, Exp Biol Med, vol.229, pp.988-995, 2004.

J. Huang, C. Plass, and C. Gerhauser, Cancer Chemoprevention by Targeting the Epigenome, Current Drug Targets, vol.12, issue.13, pp.1925-1956, 2011.
DOI : 10.2174/138945011798184155

N. Trabelsi, S. Oueslati, H. Falleh, P. Waffo-taguo, Y. Papastamoulis et al., Isolation of powerful antioxidants from the medicinal halophyte Limoniastrum guyonianum, Food Chemistry, vol.135, issue.3, pp.1419-1424, 2012.
DOI : 10.1016/j.foodchem.2012.05.120

L. Fini, M. Selgrad, V. Fogliano, G. Graziani, M. Romano et al., Annurca apple polyphenols have potent demethylating activity and can reactivate silenced tumor suppressor genes in colorectal cancer cells, J Nutr, vol.137, pp.2622-2628, 2007.

M. Unoki, T. Nishidate, and Y. Nakamura, ICBP90, an E2F-1 target, recruits HDAC1 and binds to methyl-CpG through its SRA domain, Oncogene, vol.23, issue.46, pp.7601-7610, 2004.
DOI : 10.1038/sj.onc.1208053

. Krifa, Limoniastrum guyonianum aqueous gall extract induces apoptosis in human cervical cancer cells involving p16 INK4A re-expression related to UHRF1 and DNMT1 down-regulation
URL : https://hal.archives-ouvertes.fr/inserm-00839325