Mostrar el registro sencillo del ítem

dc.contributor.authorMaldonado Rojas, Wilson
dc.date.accessioned2017-06-01T19:57:35Z
dc.date.available2017-06-01T19:57:35Z
dc.date.issued2016
dc.identifier.citationTD542.1 / M293es
dc.identifier.urihttps://hdl.handle.net/11227/4567
dc.identifier.urihttp://dx.doi.org/10.57799/11227/151
dc.descriptionThesis (Doctor in Environmental Toxicology). -- University cartagena. Faculty of Pharmaceutical Sciences. Environmental Toxicology Program, 2016es
dc.description.abstractIn this thesis are presented the results of a multidisciplinary methodological strategy that included powerful computational tools such as molecular docking (AutoDock vina, GOLD and Surflex-Dock, SYBYL), pharmocophoremapping (Ligand Scout 3.0) combined with statistical methods (cluster analisis, lincar discriminant analysis, linear regression, among others) to the search for natural compounds with anticancer activity by inhibition of cancer targets such as, COX-2, iNos and DNMT1.es
dc.format.mediumapplication/pdf
dc.language.isoenges
dc.publisherUniversity cartagenaes
dc.rights.urihttps://creativecommons.org/licenses/by-nc-nd/4.0
dc.subjectPruebas de toxicidad in vitroes
dc.subjectCompuestos orgánicos - Investigacioneses
dc.subjectBlancos moleculareses
dc.subjectCáncer - Inhibidores de proteasaes
dc.titleNatural bioactive compounds as inhibitors of cáncer targetses
dc.typeTrabajo de grado - Doctoradospa
dcterms.referencesLiu, L., Li, J., Kundu, J.K., Surh, Y.-J. Piceatannol inhibits phorbol ester-induced expression of COX-2 and iNOS in HR-1 hairless mouse skin by blocking the activation of NFκB and AP-1. Inflammat Res. 2014, 63, 1013-21.
dcterms.referencesKatiyar, S.K., Singh, T., Prasad, R., Sun, Q., Vaid, M. Epigenetic Alterations in Ultraviolet Radiation‐Induced Skin Carcinogenesis: Interaction of Bioactive Dietary Components on Epigenetic Targets. Photochem Photobiol. 2012, 88, 1066-74.
dcterms.referencesKerr, J.F., Winterford, C.M., Harmon, B.V. Apoptosis. Its significance in cancer and cancer therapy. Cancer. 1994, 73, 2013-26.
dcterms.referencesNordling, C. A new theory on the cancer-inducing mechanism. British J Cancer. 1953, 7, 68.
dcterms.referencesBarreto, J.A., O’Malley, W., Kubeil, M., Graham, B., Stephan, H., Spiccia, L. Nanomaterials: applications in cancer imaging and therapy. Adv Mater. 2011, 23.
dcterms.referencesSiegel, R., Naishadham, D., Jemal, A. Cancer statistics, 2012. CA Cancer J Clin. 2012, 62, 10-29.
dcterms.referencesHarris, C.C. Chemical and physical carcinogenesis: advances and perspectives for the 1990s. Cancer Res. 1991, 51, 5023s-44s.
dcterms.referencesHumans, I.W.G.o.t.E.o.C.R.t. Biological agents. Volume 100 B. A review of human carcinogens. IARC monographs on the evaluation of carcinogenic risks to humans/World Health Organization, International Agency for Research on Cancer. 2012, 100, 1.
dcterms.referencesDoll, R., Cook, P. Summarizing indices for comparison of cancer incidence data. Int J Cancer. 1967, 2, 269-79.
dcterms.referencesThun, M.J., DeLancey, J.O., Center, M.M., Jemal, A., Ward, E.M. The global burden of cancer: priorities for prevention. Carcinogenesis. 2010, 31, 100-10.
dcterms.referencesClifford, G.M., Polesel, J., Rickenbach, M., Dal Maso, L., Keiser, O., Kofler, A., et al. Cancer risk in the Swiss HIV Cohort Study: associations with immunodeficiency, smoking, and highly active antiretroviral therapy. J Natl Cancer Inst. 2005, 97, 425-32
dcterms.referencesFerlay, J., Soerjomataram, I., Ervik, M., Dikshit, R., Eser, S., Mathers, C., et al. Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 11 [Internet]. Lyon, France: International Agency for Research on Cancer. GLOBOCAN. 2013; 2012 v1. 0, htt p. globocan. iarc. fr. Accessed. 2015, 30.
dcterms.referencesKnudson, A.G. Hereditary cancer, oncogenes, and antioncogenes. Cancer Res. 1985, 45, 1437-43.
dcterms.referencesMarnett, L.J. Oxyradicals and DNA damage. Carcinogenesis. 2000, 21, 361-70
dcterms.referencesOrganization, W.H. Cancer control: knowledge into action: WHO guide for effective programmes, World Health Organization, 2007.
dcterms.referencesMiller, B. Cancer: We Can Win The War Against Cancer By Aggresively pursuing Prevention, Oak Publication Sdn Bhd, 2005
dcterms.referencesSmith, R.A., Cokkinides, V., Eyre, H.J. Cancer screening in the United States, 2007: a review of current guidelines, practices, and prospects. CA Cancer J Clin. 2007, 57, 90-104
dcterms.referencesSabatino, S.A., Habarta, N., Baron, R.C., Coates, R.J., Rimer, B.K., Kerner, J., et al. Interventions to increase recommendation and delivery of screening for breast, cervical, and colorectal cancers by healthcare providers: systematic reviews of provider assessment and feedback and provider incentives. Am J Prevent Med. 2008, 35, S67-S74.
dcterms.referencesHanahan, D., Weinberg, R.A. Hallmarks of cancer: the next generation. cell. 2011, 144, 646-74.
dcterms.referencesNguyen, D.X., Bos, P.D., Massagué, J. Metastasis: from dissemination to organspecific colonization. Nat Rev Cancer. 2009, 9, 274-84.
dcterms.referencesJoyce, J.A., Pollard, J.W. Microenvironmental regulation of metastasis. Nat Rev Cancer. 2009, 9, 239-52
dcterms.referencesOvadje, P., Roma, A., Steckle, M., Nicoletti, L., Arnason, J.T., Pandey, S. Advances in the research and development of natural health products as main stream cancer therapeutics. Evid Based Compl Alternat Med 2015, 2015.
dcterms.referencesHarvey, A.L., Edrada-Ebel, R., Quinn, R.J. The re-emergence of natural products for drug discovery in the genomics era. Nat Rev Drug Dis. 2015, 14, 111-29.
dcterms.referencesDe Monte, C., Carradori, S., Secci, D., D'Ascenzio, M., Guglielmi, P., Mollica, A., et al. Synthesis and pharmacological screening of a large library of 1, 3, 4-thiadiazolines as innovative therapeutic tools for the treatment of prostate cancer and melanoma. Eur J Med Chem. 2015, 105, 245-62.
dcterms.references] Walls, B., Jordan, L., Diaz, L., Miller, R. Targeted therapy for cutaneous oncology: a review of novel treatment options for non-melanoma skin cancer: part I. J Drugs Dermatol. 2014, 13, 947-52
dcterms.referencesD'Orazio, J., Jarrett, S., Amaro-Ortiz, A., Scott, T. UV Radiation and the Skin. Int J Mol Sci. 2013, 14, 12222-48.
dcterms.referencesNandakumar, V., Vaid, M., Tollefsbol, T.O., Katiyar, S.K. Aberrant DNA hypermethylation patterns lead to transcriptional silencing of tumor suppressor genes in UVBexposed skin and UVB-induced skin tumors of mice. Carcinogenesis. 2011, 32, 597-604.
dcterms.referencesLiggett, W., Sidransky, D. Role of the p16 tumor suppressor gene in cancer. J Clin Oncol. 1998, 16, 1197-206.
dcterms.referencesZhai, Y., Dang, Y., Gao, W., Zhang, Y., Xu, P., Gu, J., et al. P38 and JNK signal pathways are involved in the regulation of phlorizin against UVB‐induced skin damage. Experimen Dermatol. 2015.
dcterms.referencesVicentini, F.T.M.C., He, T., Shao, Y., Fonseca, M.J.V., Verri Jr, W.A., Fisher, G.J., et al. Quercetin inhibits UV irradiation-induced inflammatory cytokine production in primary human keratinocytes by suppressing NF-κB pathway. J Dermatol Sci. 2011, 61, 162-8.
dcterms.referencesKoehn, F.E., Carter, G.T. The evolving role of natural products in drug discovery. Nat Rev Drug Discov. 2005, 4, 206-20.
dcterms.referencesSahu, S., Saraf, S., Kaur, C.D. Biocompatible nanoparticles for sustained topical delivery of anticancer phytoconstituent quercetin. Pakistan journal of biological sciences: PJBS. 2013, 16, 601-9.
dcterms.referencesTaveira, S.F., de Campos, A., Luciana, M., de Santana, D.C., Nomizo, A., de Freitas, L.A.P., et al. Development of cationic solid lipid nanoparticles with factorial design-based studies for topical administration of doxorubicin. J Biomed Nanotechnol. 2012, 8, 219-28
dcterms.referencesMangalathillam, S., Rejinold, N.S., Nair, A., Lakshmanan, V.-K., Nair, S.V., Jayakumar, R. Curcumin loaded chitin nanogels for skin cancer treatment via the transdermal route. Nanoscale. 2012, 4, 239-50.
dcterms.referencesBar-Sela, G., Epelbaum, R., Schaffer, M. Curcumin as an anti-cancer agent: review of the gap between basic and clinical applications. Curr Med Chem. 2010, 17, 190-7.
dcterms.referencesYao, K., Chen, H., Lee, M.-H., Langfald, A., Kim, M.O., Yu, D.H., et al. Kaempferol suppresses solar ultraviolet radiation-induced skin cancers by targeting RSK2 and MSK1. Cancer Res. 2014, 7, 958-67.
dcterms.referencesIbbotson, S. Topical 5‐aminolaevulinic acid photodynamic therapy for the treatment of skin conditions other than non‐melanoma skin cancer. British J Dermatol. 2002, 146, 178- 88.
dcterms.referencesAfaq, F., Katiyar, S. Dietary Phytochemicals and Chemoprevention of Solar Ultraviolet Radiation-Induced Skin Cancer. In: Nutraceuticals and Cancer, Sarkar, F.H. Ed., Springer Netherlands, 2012, pp. 295-321.
dcterms.referencesLitterman, N.K., Lipinski, C.A., Bunin, B.A., Ekins, S. Computational prediction and validation of an expert’s evaluation of chemical probes. J Chem Inf Model. 2014, 54, 2996- 3004.
dcterms.referencesBarelier, S., Eidam, O., Fish, I., Hollander, J., Figaroa, F., Nachane, R., et al. Increasing chemical space coverage by combining empirical and computational fragment screens. ACS Chem Biol. 2014, 9, 1528-35.
dcterms.referencesSliwoski, G., Kothiwale, S., Meiler, J., Lowe, E.W. Computational methods in drug discovery. Pharmacol Rev. 2014, 66, 334-95
dcterms.referencesPatil, S.P., Ballester, P.J., Kerezsi, C.R. Prospective virtual screening for novel p53– MDM2 inhibitors using ultrafast shape recognition. J Comput Aided Mol Des. 2014, 28, 89-97.
dcterms.referencesKumar, V., Krishna, S., Siddiqi, M.I. Virtual screening strategies: Recent advances in the identification and design of anti-cancer agents. Methods. 2015, 71, 64-70.
dcterms.referencesTuccinardi, T., Poli, G., Dell'Agnello, M., Granchi, C., Minutolo, F., Martinelli, A. Receptor-based virtual screening evaluation for the identification of estrogen receptor β ligands. J Enzyme Inhib Med Chem. 2014, 1-9.
dcterms.referencesSiegel, R.L., Miller, K.D., Jemal, A. Cancer statistics, 2016. CA Cancer J Clin. 2016, 66, 7-30.
dcterms.referencesde Vries, E., Arroyave, I., Pardo, C., Wiesner, C., Murillo, R., Forman, D., et al. Trends in inequalities in premature cancer mortality by educational level in Colombia, 1998– 2007. J Epidemiol Comm Health. 2015, 69, 408-15.
dcterms.referencesDe Vries, E., Meneses, M.X., Piñeros, M. Años de vida perdidos como una medida de carga de cáncer en Colombia, 1997-2012. Biomédica. 2016, 36.
dcterms.referencesAngel, J., Ibarra, J., Diaz, S., Lehmann, C., Garcia, M., Guzman, L., et al. Clinical behavior of breast cancer in men in a Latin American population. Rev Colomb Cancerol. 2015, 19, 150-5.
dcterms.referencesCombariza, J.F., Lombana, M., Torres, A.M., Castellanos, A.M., Arango, M. General features and epidemiology of lymphoma in Colombia. A multicentric study. Ann Hematol. 2015, 94, 975-80.
dcterms.referencesMuñoz, N., Bravo, L.E. Epidemiology of cervical cancer in Colombia. Salud Publica Mex. 2014, 56, 431-9.
dcterms.referencesSanchez, G., Nova, J., de la Hoz, F. Risk factors for basal cell carcinoma: a study from the national dermatology center of Colombia. Act Dermo-Sifiliograf 2012, 103, 294-300.
dcterms.referencesVallejo-Torres, L., Morris, S., Kinge, J., Poirier, V., Verne, J. Measuring current and future cost of skin cancer in England. J Pub Health. 2013, fdt032
dcterms.referencesPardo, C., Murillo, R., Piñeros, M., Castro, M.Á. Casos nuevos de cáncer en el Instituto Nacional de Cancerología, Colombia, 2002. Rev Colomb Cancerol. 2003, 7, 4-19
dcterms.referencesNova-Villanueva, J., Sánchez-Vanegas, G., de Quintana, L.P. Cáncer de Piel: Perfil Epidemiológico de un Centro de Referencia en Colombia. Rev Salud Pub. 2007, 9, 4.
dcterms.referencesColombia. Ministerio de Ambiente, Vivienda y Desarrollo Territorial. . Ozono. Bogotá: IDEAM. 2002
dcterms.referencesKala, R., Shah, H.N., Martin, S.L., Tollefsbol, T.O. Epigenetic-based combinatorial resveratrol and pterostilbene alters DNA damage response by affecting SIRT1 and DNMT enzyme expression, including SIRT1-dependent γ-H2AX and telomerase regulation in triplenegative breast cancer. BMC cancer. 2015, 15, 1.
dcterms.referencesKim, H., Ramirez, C.N., Su, Z.-Y., Kong, A.-N.T. Epigenetic modifications of triterpenoid ursolic acid in activating Nrf2 and blocking cellular transformation of mouse epidermal cells. J Nutr Biochem. 2015.
dcterms.referencesMaini, S., Fahlman, B.M., Krol, E.S. Flavonols Protect Against UV Radiation-Induced Thymine Dimer Formation in an Artificial Skin Mimic. J Pharm Pharmaceut Sci. 2015, 18.
dcterms.referencesParedes‐Gonzalez, X., Fuentes, F., Jeffery, S., Saw, C.L.L., Shu, L., Su, Z.Y., et al. Induction of NRF2‐mediated gene expression by dietary phytochemical flavones apigenin and luteolin. Biopharmaceut Drug Disp. 2015, 36, 440-51
dcterms.referencesAtanasov, A.G., Waltenberger, B., Pferschy-Wenzig, E.-M., Linder, T., Wawrosch, C., Uhrin, P., et al. Discovery and resupply of pharmacologically active plant-derived natural products: A review. Biotechnology Adv. 2015, 33, 1582-614.
dcterms.referencesHowat, S., Park, B., Oh, I.S., Jin, Y.-W., Lee, E.-K., Loake, G.J. Paclitaxel: biosynthesis, production and future prospects. N Biotechnol. 2014, 31, 242-5
dcterms.referencesReddy, L., Odhav, B., Bhoola, K. Natural products for cancer prevention: a global perspective. Pharmacol Therap. 2003, 99, 1-13.
dcterms.referencesMann, J. Natural products in cancer chemotherapy: past, present and future. Nat Rev Cancer. 2002, 2, 143-8.
dcterms.referencesRamos, A.A., Castro-Carvalho, B., Prata-Sena, M., Dethoup, T., Buttachon, S., Kijjoa, A., et al. Crude extracts of marine-derived and soil fungi of the genus Neosartorya exhibit selective anticancer activity by inducing cell death in colon, breast and skin cancer cell lines. Pharmacogn Res. 2016, 8, 8.
dcterms.referencesShrivastava, N., Kamal, S., Varma, A. Cancer and Its Biology. A Textbook of Molecular Biotechnology. 2009, 401.
dcterms.referencesMontesano, R., Hall, J. Environmental causes of human cancers. Eur J Cancer. 2001, 37, 67-87.
dcterms.referencesLichtenstein, P., Holm, N.V., Verkasalo, P.K., Iliadou, A., Kaprio, J., Koskenvuo, M., et al. Environmental and heritable factors in the causation of cancer—analyses of cohorts of twins from Sweden, Denmark, and Finland. N Engl J Med. 2000, 343, 78-85.
dcterms.referencesNCI, N.C.I., 2016
dcterms.referencesZhou, B.-B.S., Zhang, H., Damelin, M., Geles, K.G., Grindley, J.C., Dirks, P.B. Tumour-initiating cells: challenges and opportunities for anticancer drug discovery. Nat Rev Drug Dis. 2009, 8, 806-23
dcterms.referencesHermann, P.C., Huber, S.L., Herrler, T., Aicher, A., Ellwart, J.W., Guba, M., et al. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell. 2007, 1, 313-23.
dcterms.referencesEliceiri, B.P., Cheresh, D.A. The role of αv integrins during angiogenesis: insights into potential mechanisms of action and clinical development. J Clin Invest. 1999, 103, 1227-30.
dcterms.referencesClevers, H. The cancer stem cell: premises, promises and challenges. Nat Med. 2011, 313-9.
dcterms.referencesYasuhara, T., Suzuki, T., Katsura, M., Miyagawa, K. Rad54B serves as a scaffold in the DNA damage response that limits checkpoint strength. Nat Commun. 2014, 5.
dcterms.referencesHanahan, D., Weinberg, R.A. Hallmarks of cancer: the next generation. cell. 2011, 144, 646-74.
dcterms.referencesNguyen, D.X., Bos, P.D., Massagué, J. Metastasis: from dissemination to organspecific colonization. Nat Rev Cancer. 2009, 9, 274-84.
dcterms.referencesSaha, K., Hornyak, T.J., Eckert, R.L. Epigenetic cancer prevention mechanisms in skin cancer. The AAPS journal. 2013, 15, 1064-71.
dcterms.referencesFraga, M.F., Herranz, M., Espada, J., Ballestar, E., Paz, M.F., Ropero, S., et al. A mouse skin multistage carcinogenesis model reflects the aberrant DNA methylation patterns of human tumors. Cancer Res. 2004, 64, 5527-34.
dcterms.referencesTanemura, A., Terando, A.M., Sim, M.-S., van Hoesel, A.Q., de Maat, M.F., Morton, D.L., et al. CpG island methylator phenotype predicts progression of malignant melanoma. Clin Cancer Res. 2009, 15, 1801-7.
dcterms.referencesvan den Hurk, K., Niessen, H.E., Veeck, J., van den Oord, J.J., van Steensel, M.A., Zur Hausen, A., et al. Genetics and epigenetics of cutaneous malignant melanoma: a concert out of tune. BBA Rev Cancer. 2012, 1826, 89-102.
dcterms.referencesLiu, L., Li, J., Kundu, J.K., Surh, Y.-J. Piceatannol inhibits phorbol ester-induced expression of COX-2 and iNOS in HR-1 hairless mouse skin by blocking the activation of NFκB and AP-1. Inflammat Res. 2014, 63, 1013-21.
dcterms.referencesCavasotto, C.N., Orry, W., Andrew, J. Ligand docking and structure-based virtual screening in drug discovery. Curr Top Med Chem. 2007, 7, 1006-14.
dcterms.referencesVerdonk, M.L., Cole, J.C., Hartshorn, M.J., Murray, C.W., Taylor, R.D. Improved protein–ligand docking using GOLD. Proteins: Struct Funct Bioinf. 2003, 52, 609-23.
dcterms.referencesJain, A.N. Surflex-Dock 2.1: robust performance from ligand energetic modeling, ring flexibility, and knowledge-based search. J Comp Aided Mol Des. 2007, 21, 281-306
dcterms.referencesTrott, O., Olson, A.J. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem. 2010, 31, 455-61.
dcterms.referencesZhou, X., Li, Y., Chen, X. Computational identification of bioactive natural products by structure activity relationship. J Mol Graph Modell. 2010, 29, 38-45.
dcterms.referencesThoppil, R.J., Bishayee, A. Terpenoids as potential chemopreventive and therapeutic agents in liver cancer. World J Hepatol. 2011, 3, 228-49.
dcterms.referencesLucas, L., Russell, A., Keast, R. Molecular mechanisms of inflammation. Antiinflammatory benefits of virgin olive oil and the phenolic compound oleocanthal. Curr Pharm Des. 2011, 17, 754-68.
dcterms.referencesLila, M.A., Raskin, I. Health‐related Interactions of Phytochemicals. J Food Sci. 2005, 70, R20-R7
dcterms.referencesMishra, B.B., Tiwari, V.K. Natural products: an evolving role in future drug discovery. Eur J Med Chem. 2011, 46, 4769-807.
dcterms.referencesMirhendi, H., Makimura, K., Khoramizadeh, M., Yamaguchi, H. A one-enzyme PCRRFLP assay for identification of six medically important Candida species. Nihon Ishinkin Gakkai Zasshi. 2006, 47, 225-9
dcterms.referencesGirish, P., Anjaneyulu, A., Viswas, K., Shivakumar, B., Anand, M., Patel, M., et al. Meat species identification by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) of mitochondrial 12S rRNA gene. Meat Sci. 2005, 70, 107-12.
dcterms.referencesAlSuhaibani, E., Voudouris, C., Al-Atiyat, R., Kotzamumin, A., Vontas, J., Margaritopoulos, J. Identification of a point mutation in the ace1 gene of Therioaphis trifolli maculata and detection of insecticide resistance by a diagnostic PCR–RFLP assay. Bull Entomol Res. 2015, 105, 712-6.
dcterms.referencesDowgier, G., Mari, V., Losurdo, M., Larocca, V., Colaianni, M.L., Cirone, F., et al. A duplex real-time PCR assay based on TaqMan technology for simultaneous detection and differentiation of canine adenovirus types 1 and 2. J Virol Methods. 2016, 234, 1-6.
dcterms.referencesSousa, D.R.T.d., Santos, C.S.d.S., Wanke, B., Júnior, S., Santos, M.C.d., Cruz, K.S., et al. PCR-RFLP as a useful tool for diagnosis of invasive mycoses in a healthcare facility in the North of Brazil. Electron J Biotech. 2015, 18, 231-5.
dcterms.referencesXu, W., Morris, U., Aydin-Schmidt, B., Msellem, M.I., Shakely, D., Petzold, M., et al. SYBR green real-time PCR-RFLP assay targeting the Plasmodium cytochrome b gene–a highly sensitive molecular tool for malaria parasite detection and species determination. PloS one. 2015, 10, e0120210.
dcterms.referencesRampersad, S.N. Multiple applications of Alamar Blue as an indicator of metabolic function and cellular health in cell viability bioassays. Sensors. 2012, 12, 12347-60.
dcterms.referencesStockert, J.C., Blázquez-Castro, A., Cañete, M., Horobin, R.W., Villanueva, Á. MTT assay for cell viability: Intracellular localization of the formazan product is in lipid droplets. Acta Histochem. 2012, 114, 785-96.
dcterms.referencesLim, S.-W., Loh, H.-S., Ting, K.-N., Bradshaw, T.D., Allaudin, Z.N. Reduction of MTT to Purple Formazan by Vitamin E Isomers in the Absence of Cells. Trop Life Sci Res. 2015, 26, 111.
dcterms.referencesLiu, C., Hou, L., Liu, L., Qu, Z., Wang, X., Sun, X., et al. In Vitro Cytotoxicity Evaluation of Highly Absorbent Foam Dressings Based on Silver Zirconium Phosphate via IC50 Value. J Biomat Nanobiotech. 2016, 7, 37.
dcterms.referencesHoldgate, G., Geschwindner, S., Breeze, A., Davies, G., Colclough, N., Temesi, D., et al. Biophysical methods in drug discovery from small molecule to pharmaceutical. Methods Mol Biol. 2013,1008, 327-55
dcterms.referencesEnyedy, É.A., Dömötör, O., Bali, K., Hetényi, A., Tuccinardi, T., Keppler, B.K. Interaction of the anticancer gallium (III) complexes of 8-hydroxyquinoline and maltol with human serum proteins. J Biol Inorg Chem. 2015, 20, 77-88.
dcterms.referencesDomínguez-García, M., Ortega-Zúñiga, C., Meléndez, E. New tungstenocenes containing 3-hydroxy-4-pyrone ligands: antiproliferative activity on HT-29 and MCF-7 cell lines and binding to human serum albumin studied by fluorescence spectroscopy and molecular modeling methods. J Biol Inorg Chem. 2013, 18, 195-209.
dcterms.referencesNarváez-Pita, X., Ortega-Zuniga, C., Acevedo-Morantes, C.Y., Pastrana, B., OliveroVerbel, J., Maldonado-Rojas, W., et al. Water soluble molybdenocene complexes: Synthesis, cytotoxic activity and binding studies to ubiquitin by fluorescence spectroscopy, circular dichroism and molecular modeling. J Inorg Biochem. 2014, 132, 77-91.
dcterms.referencesLopes, J.L., Miles, A.J., Whitmore, L., Wallace, B.A. Distinct circular dichroism spectroscopic signatures of polyproline II and unordered secondary structures: Applications in secondary structure analyses. Protein Sci. 2014, 23, 1765-72
dcterms.referencesKitts, D.D. Bioactive substances in food: identification and potential uses. Can J Physiol Pharmacol. 1994, 72, 423-424.
dcterms.referencesPan, M.H., Lai, C.S., Dushenkov, S., Ho, C.T. Modulation of inflammatory genes by natural dietary bioactive compounds. J Agric Food Chem. 2009, 57, 4467-447.
dcterms.referencesBohlin, L., Göransson, U., Alsmark, C., Wedén, C., Backlund, A. Natural products in modern life science. Phytochem Rev. 2010, 9, 279-301.
dcterms.referencesShen, C.L., Yeh, J.K., Cao, J., Wang, J.S. Green tea and bone metabolism. Nutr Res. 2009, 29, 437-456.
dcterms.referencesChakraborti, A.K., Garg, S.K., Kumar, R., Motiwala, H.F., Jadhavar, P.S. Progress in COX-2 inhibitors: a journey so far. Curr Med Chem. 2010, 17, 1563-1593.
dcterms.referencesKiefer, J.R., Pawlitz, J.L., Moreland, K.T., Stegeman, R.A., Hood, W.F., Gierse, J.K., Stevens, A.M., Goodwin, D.C., Rowlinson, S.W., Marnett, L.J., Stallings, W.C., Kurumbail, R.G. Structural insights into the stereochemistry of the cyclooxygenase reaction. Nature 2000, 405, 97-101.
dcterms.referencesWilken, R., Veena, M.S., Wang, M.B., Srivatsan, E.S. Curcumin: a review of anticancer properties and therapeutic activity in head & neck squamous cell carcinoma. Mol Cancer 2011, 10, 12
dcterms.referencesFrasca, G., Panico, A.M., Bonina, F., Messina, R., Rizza, L., Musumeci, G., Rapisarda, P., Cardile, V. Involvement of inducible nitric oxide synthase and cyclooxygenase2 in the anti-inflammatory effects of a red orange extract in human chondrocytes. Nat Prod Res. 2010, 24, 1469-1480.
dcterms.referencesJin, M., Suh, S.J., Yang, J.H., Lu, Y., Kim, S.J., Kwon, S., Jo, T.H., Kim, J.W., Park, Y.I., Ahn, G.W., Lee, C.K., Kim, C.H., Son, J.K., Son, K.H., Chang, H.W. Anti-inflammatory activity of bark of Dioscorea batatas DECNE through the inhibition of iNOS and COX-2 expressions in RAW264.7 cells via NF-κB and ERK1/2 inactivation. Food Chem Toxicol. 2010, 48, 3073-3079.
dcterms.referencesMay, A., Zacharias, M. Accounting for global protein deformability during protein– protein and protein–ligand docking. BBA- Proteins Proteom 2005, 1754, 225-231.
dcterms.referencesTrott, O., Olson, A.J. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization and multithreading. J Comput Chem. 2009, 31, 455-461.
dcterms.referencesVerdonk, M.L., Cole, J.C., Hartshorn, M.J., Murray, C.W., Taylor, R.D. Improved protein–ligand docking using GOLD. Proteins: Struct Funct Gen. 2003, 52, 609-623.
dcterms.referencesJain, A.N., Surflex-Dock 2.1: Robust performance from ligand energetic modeling, ring flexibility, and knowledge-based search. J Comput Aided Mol Des. 2007, 21, 281-306.
dcterms.referencesTang, Y., Zhu, W., Chen, K., Jiang, H. New technologies in computer-aided drug design: Toward target identification and new chemical entity discovery. Drug Discov Today Tech. 2006, 3, 307-313.
dcterms.referencesUtsintong, M., Talley, T.T., Taylor, P.W., Olson, A.J., Vajragupta, O. Virtual screening against alpha-cobratoxin. J Biomol Screen. 2009, 14, 1109-1118.
dcterms.referencesRCSB PDB Protein Data Bank | Home ww.pdb.org/pdb/home/home.do.
dcterms.referencesSYBYL molecular modeling software, Version 8.1. 2007,. Tripos, St. Louis, MO, U.S.A
dcterms.referencesFrisch, M., Trucks, G., Schlegel, H., Scuseria, G., Robb, M., Cheeseman, J., et al. Gaussian 03, revision B. 05; Gaussian. Inc., Pittsburgh, PA. 2003, 12478
dcterms.referencesGuha, R., Howard, M.T., Hutchison, G.R., Murray-Rust, P., Rzepa, H., Steinbeck, C., Wegner, J.K., Willighagen, E. The blue obelisk -- interoperability in chemical informatics. J Chem Inf Model. 2006, 46, 991-998.
dcterms.referencesYao, Y., Sang, W., Zhou, M., Ren, G. Phenolic composition and antioxidant activities of 11 celery cultivars. J Food Sci. 2010, 75, 9-13.
dcterms.referencesStuetz, W., Prapamontol, T., Hongsibsong, S., Biesalski, H.K. Polymethoxylated flavones, flavanone glycosides, carotenoids, and antioxidants in different cultivation types of tangerines (Citrus reticulata Blanco cv. Sainampueng) from Northern Thailand. J Agric Food Chem. 2010, 58, 6069-6074.
dcterms.referencesAbenavoli, L., Capasso, R., Milic, N., Capasso, F. Milk thistle in liver diseases: past, present, future. Phytother Res. 2010, 24, 1423-1432.
dcterms.referencesSarić, A., Sobocanec, S., Balog, T., Kusić, B., Sverko, V., Dragović-Uzelac, V., Levaj, B., Cosić, Z., Safranko, Z.M., Marotti, T. Improved antioxidant and anti-inflammatory potential in mice consuming sour cherry juice (Prunus cerasus cv. Maraska). Plant Foods Hum Nutr. 2009, 64, 231-237.
dcterms.referencesCastellarin, S.D., Di Gaspero, G. Transcriptional control of anthocyanin biosynthetic genes in extreme phenotypes for berry pigmentation of naturally occurring grapevines. BMC Plant Biol. 2007, 30, 7-46.
dcterms.referencesBasini, G., Bussolati, S., Santini, S.E., Grasselli, F. The impact of the phytooestrogen genistein on swine granulosa cell function. J Anim Physiol Anim Nutr (Berl). 2010, 94, 374-382.
dcterms.referencesWang, P., Aronson, W.J., Huang, M., Zhang, Y., Lee, R.P., Heber, D., Henning, S.M. Green tea polyphenols and metabolites in prostatectomy tissue: Implications for Cancer prevention. Cancer Prev Res. 2010, 3, 985-993.
dcterms.referencesLien, L.T., Yeh, H.S., Su, W.T. Effect of adding extracted hesperetin, naringenin and pectin on egg cholesterol, serum traits and antioxidant activity in laying hens. Arch Anim Nutr. 2008, 62, 33-43.
dcterms.referencesVasanthi, H.R., Mukherjee, S., Das, D.K. Potential health benefits of broccoli- a chemico-biological overview. Mini Rev Med Chem. 2009, 9, 749-759
dcterms.referencesLai, C.S., Li, S., Chai, C.Y., Lo, C.Y., Ho, C.T., Wang, Y.J., Pan, M.H. Inhibitory effect of citrus 5-hydroxy-3,6,7,8,3',4'- hexamethoxyflavone on 12-O-tetradecanoylphorbol 13- acetate-induced skin inflammation and tumor promotion in mice. Carcinogenesis 2007, 28, 2581-2588.
dcterms.referencesKaura, S., Modib, N.H., Pandac, D., Roya, N. Probing the binding site of curcumin in Escherichia coli and Bacillus subtilis FtsZ - A structural insight to unveil antibacterial activity of curcumin. Eur J Med Chem. 2010, 45, 4209-4214.
dcterms.referencesRasyid, A., Rahman, A.R., Jaalam, K., Lelo, A. Effect of different curcumin dosages on human gall bladder. Asia Pac J Clin Nutr. 2002, 11, 314-318
dcterms.referencesKalantari, H., Das, D.K. Physiological effects of resveratrol. Biofactors. 2010, 36, 401-406.
dcterms.referencesWeng, C.J., Wu, C.F., Huang, H.W., Ho, C.T., Yen, G.C. Anti-invasion effects of 6- shogaol and 6-gingerol, two active components in ginger, on human hepatocarcinoma cells. Mol Nutr Food Res. 2010, 54, 1618-1627.
dcterms.referencesBernardes, W.A., Lucarini, R., Tozatti, M.G., Souza, M.G., Andrade Silva, M.L., da Silva Filho, A.A., Martins, C.H., Miller Crotti, A.E., Pauletti, P.M., Groppo, M., Cunha, W.R. Antimicrobial activity of Rosmarinus officinalis against oral pathogens: relevance of carnosica acid and carnosol. Chem Biodivers. 2010, 7, 1835-1840.
dcterms.referencesAlosi, J.A., McDonald, D.E., Schneider, J.S., Privette, A.R., McFadden, D.W. Pterostilbene inhibits breast cancer in vitro through mitochondrial depolarization and induction of caspase-dependent apoptosis. J Surg Res. 2010, 161, 195-201.
dcterms.referencesVisanji, J.M., Duthie, S.J., Pirie, L., Thompson, D.G., Padfield, P.J. Dietary isothiocyanates inhibit Caco-2 cell proliferation and induce G2/M phase cell cycle arrest, DNA damage, and G2/M checkpoint activation. J Nutr. 2004, 134, 3121-3126.
dcterms.referencesSkupinska, K., Misiewicz-Krzeminska, I., Lubelska, K., Kasprzycka-Guttman, T. The effect of isothiocyanates on CYP1A1 and CYP1A2 activities induced by polycyclic aromatic hydrocarbons in Mcf7 cells. Toxicol In Vitro. 2009, 23, 763-771
dcterms.referencesDéziel, B.A., Patel, K., Neto, C., Gottschall-Pass, K., Hurta, R.A. Proanthocyanidins from the american cranberry (Vaccinium macrocarpon), inhibit matrix metalloproteinase-2 and matrix metalloproteinase-9 activity in human prostate cancer cells via alterations in multiple cellular signaling pathways. J Cell Biochem. 2010, 111, 742-754.
dcterms.referencesTheodosiou, M., Laudet, V., Schubert, M. From carrot to clinic: an overview of the retinoic acid signaling pathway, Cell. Mol. Life Sci. 67, 2010, 1423-1445
dcterms.referencesG.L. Ambrosini, N.H. de Klerk, L. Fritschi, D. Mackerras, B. Musk, Fruit, vegetable, vitamin A intakes, and prostate cancer risk, Prostate Cancer Prostatic Dis. 2008, 11, 61-66.
dcterms.referencesMuhammad, S.A., Muhammad, S., Waqar, A., Masood, P., Raghav, Y. Chlorinated monoterpene ketone, acylated β-sitosterol glycosides and a flavanone glycoside from Mentha longifolia (Lamiaceae). Phytochemistry 2002, 59, 889-895.
dcterms.referencesVogel, J.T., Tieman, D.M., Sims, C.A., Odabasi, A.Z., Clark, D.G., Klee, H.J. Carotenoid content impacts flavor acceptability in tomato (Solanum lycopersicum). J Sci Food Agric. 2010, 90, 2233-2340
dcterms.referencesThürmann, P.A., Steffen, J., Zwernemann, C., Aebischer, C.P., Cohn, W., Wendt, G., Schalch, W. Plasma concentration response to drinks containing beta-carotene as carrot juice or formulated as a water dispersible powder. Eur J Nutr. 2002, 41, 228-235.
dcterms.referencesBurns-Whitmore, B.L., Haddad, E.H., Sabaté, J., Jaceldo-Siegl, K., Tanzman, J., Rajaram, S. Effect of n-3 fatty acid enriched eggs and organic eggs on serum lutein in freeliving lacto-ovo vegetarians. Eur J Clin Nutr. 2010, 64, 1332-1337.
dcterms.referencesMills, J.D., Bailes, J.E., Sedney, C.L., Hutchins, H., Sears, B. Omega-3 fatty acid supplementation and reduction of traumatic axonal injury in a rodent head injury model. J Neurosurg. 2010, 114, 77-84.
dcterms.referencesMann, N.J., O'Connell, S.L., Baldwin, K.M., Singh, I., Meyer, B.J. Effects of seal oil and tuna-fish oil on platelet parameters and plasma lipid levels in healthy subjects. Lipids 2010, 45, 669-681.
dcterms.referencesWolber, G., Langer, T. LigandScout: 3-D pharmacophores derived from proteinbound ligands and their use as virtual screening filters. J Chem Inf Model. 2005, 45, 160-169.
dcterms.referencesPubChem. http://pubchem.ncbi.nlm.nih.gov.
dcterms.referencesFilizola, M., Perez, J.J., Palomer, A., Mauleón, D. Comparative molecular modeling study of the three-dimensional structures of prostaglandin endoperoxide H2 synthase 1 and 2 (COX-1 and COX-2). J Mol Graph Model. 1997, 15, 290-300.
dcterms.referencesDewitt, D.L. Cox-2-selective inhibitors: The new super aspirins. Mol Pharm 1999, 55, 625-631.
dcterms.referencesZhou, Z., Wang, Y., Bryant. S.H. Computational analysis of the cathepsin B inhibitors activities through LR-MMPBSA binding affinity calculation based on docked complex. J Comput Chem. 2009, 30, 2165-2175.
dcterms.referencesLev-Ari, S., Strier, L., Kazanov, D., Madar-Shapiro, L., Dvory-Sobol, H., Pinchuk, I., Marian, B., Lichtenberg, D., Arber, N. Celecoxib and curcumin synergistically inhibit the growth of colorectal cancer cells. Clin Cancer Res. 2005, 11, 6738-6744.
dcterms.referencesLev-Ari, S., Strier, L., Kazanov, D., Elkayam, O., Lichtenberg, D., Caspi D., Arber, N. Curcumin synergistically potentiates the growth-inhibitory and pro-apoptotic effects of celecoxib in osteoarthritis synovial adherent cells. Rheumatology 2006, 45, 171-177.
dcterms.referencesSelvam, C., Jachak, S.M., Thilagavathi, R., Chakraborti, A.K. Design, synthesis, biological evaluation and molecular docking of curcumin analogues as antioxidant, cyclooxygenase inhibitory and anti-inflammatory agents. Bioorg Med Chem Lett. 2005, 15, 1793-1797.
dcterms.referencesFurse, K.E., Pratt, D.A., Porter, N.A., Lybrand, T.P. Molecular dynamics simulations of arachidonic acid complexes with COX-1 and COX-2: insights into equilibrium behavior. Biochemistry 2006, 45, 3189-3205.
dcterms.referencesWu, S.J., Tam, K.W., Tsai, Y.H., Chang, C.C., Chao, J.C. Curcumin and saikosaponin A inhibit chemical-induced liver inflammation and fibrosis in rats. Am J Chin Med. 2010, 38, 99-111.
dcterms.referencesFurse, K.E., Pratt, D.A., Porter, N.A., Lybrand, T.P. Molecular dynamics simulations of arachidonic acid complexes with COX-1 and COX-2: insights into equilibrium behavior. Biochemistry 2006, 45, 3189-3205.
dcterms.referencesWu, S.J., Tam, K.W., Tsai, Y.H., Chang, C.C., Chao, J.C. Curcumin and saikosaponin A inhibit chemical-induced liver inflammation and fibrosis in rats. Am J Chin Med. 2010, 38, 99-111.
dcterms.referencesSikora, E., Scapagnini, G., Barbagallo, M. Curcumin, inflammation, ageing and agerelated diseases. Immun Ageing. 2010, 7, 1.
dcterms.referencesMoghaddam, S.J., Barta, P., Mirabolfathinejad, S.G., Ammar-Aouchiche, Z., Garza, N.T., Vo, T.T., Newman, R.A., Aggarwal, B.B., Evans, C.M., Tuvim, M.J., Lotan, R., Dickey, B.F. Curcumin inhibits COPD-like airway inflammation and lung cancer progression in mice. Carcinogenesis 2009, 30, 1949-1956
dcterms.referencesJurenka, J.S. Anti-inflammatory properties of curcumin, a major constituent of Curcuma longa: a review of preclinical and clinical research. Altern Med Rev. 2009, 14, 141- 153.
dcterms.referencesGalati, G., Sabzevari, O., Wilson, J.X., O’Brien, P.J. Prooxidant activity and cellular effects of the phenoxyl radicals of dietary flavonoids and other polyphenolics. Toxicology 2002, 177, 91-104
dcterms.referencesAkihisa, T., Yasukawa, K., Yamaura, M., Ukiya, M., Kimura, Y., Shimizu, N., Koichi, A. Triterpene alcohol and sterol ferulates from rice bran and their anti-inflammatory effects. J Agric Food Chem. 2000, 48, 2313-2319.
dcterms.referencesSikora, E., Scapagnini, G., Barbagallo, M. Curcumin, inflammation, ageing and agerelated diseases. Immun Ageing. 2010, 7, 1.
dcterms.referencesMoghaddam, S.J., Barta, P., Mirabolfathinejad, S.G., Ammar-Aouchiche, Z., Garza, N.T., Vo, T.T., Newman, R.A., Aggarwal, B.B., Evans, C.M., Tuvim, M.J., Lotan, R., Dickey, B.F. Curcumin inhibits COPD-like airway inflammation and lung cancer progression in mice. Carcinogenesis 2009, 30, 1949-1956.
dcterms.referencesJurenka, J.S. Anti-inflammatory properties of curcumin, a major constituent of Curcuma longa: a review of preclinical and clinical research. Altern Med Rev. 2009, 14, 141- 153.
dcterms.referencesGalati, G., Sabzevari, O., Wilson, J.X., O’Brien, P.J. Prooxidant activity and cellular effects of the phenoxyl radicals of dietary flavonoids and other polyphenolics. Toxicology 2002, 177, 91-104.
dcterms.referencesAkihisa, T., Yasukawa, K., Yamaura, M., Ukiya, M., Kimura, Y., Shimizu, N., Koichi, A. Triterpene alcohol and sterol ferulates from rice bran and their anti-inflammatory effects. J Agric Food Chem. 2000, 48, 2313-2319
dcterms.referencesLeelawat, K., Ohuchida, K., Mizumoto, K., Mahidol, C., Tanaka, M. All-trans retinoic acid inhibits the cell proliferation but enhances the cell invasion through up-regulation of cmet in pancreatic cancer cells. Cancer Lett. 2005, 224, 303-310
dcterms.referencesLi, R.R., Pang, L.L., Du, Q., Shi, Y., Dai, W.J., Yin, K.S. Apigenin inhibits allergeninduced airway inflammation and switches immune response in a murine model of asthma. Immunopharmacol Immunotoxicol. 2010, 32, 364-370.
dcterms.referencesVela, E.M., Knostman, K.A., Mott, J.M., Warren, R.L., Garver, J.N., Vela, L.J., Stammen, R.L. Genistein, a general kinase inhibitor, as a potential antiviral for arenaviral hemorrhagic fever as described in the Pirital virus-Syrian golden hamster model. Antiviral Res. 2010, 87, 318-328.
dcterms.referencesWatkins, B.A., Hannon, K., Ferruzzi, M., Li, Y. Dietary PUFA and flavonoids as deterrents for environmental pollutants. J Nutr Biochem. 2007, 18, 196-205.
dcterms.referencesCalderone, V., Chericoni, S., Martinelli, C., Testai, L., Nardi, A., Morelli, I., Breschi, M.C., Martinotti, E. Vasorelaxing effects of flavonoids: investigation on the possible involvement of potassium channels, Naunyn Schmiedebergs. Arch Pharmacol. 2004, 370, 290-298.
dcterms.referencesLee, K.M., Lee, K.W., Jung, S.K., Lee, E.J., Heo, Y.S., Bode, A.M., Lubet, R.A., Lee, H.J., Dong, Z. Kaempferol inhibits UVB-induced COX-2 expression by suppressing Src kinase activity. Biochem Pharmacol. 2010, 80, 2042-2049
dcterms.referencesMassaro, M., Habib, A., Lubrano, L., Del Turco, S., Lazzerini, G., Bourcier, T., Weksler, B.B., De Caterina, R. The omega-3 fatty acid docosahexaenoate attenuates endothelial cyclooxygenase-2 induction through both NADP[H] oxidase and PKCε inhibition. Proc Natl Acad Sci. 2006,103, 15184-15189.
dcterms.referencesHare, A.A., Leng, L., Gandavadi, S., Du, X., Cournia, Z., Bucala, R., Jorgensen, W.L. Optimization of N-benzyl-benzoxazol-2-ones as receptor antagonists of macrophage migration inhibitory factor (MIF). Bioorg Med Chem Lett. 2010, 20, 5811-5814.
dcterms.referencesGonzales, A.M., Orlando, R.A. Curcumin and resveratrol inhibit nuclear factorkappaB-mediated cytokine expression in adipocytes. Nutr Metab. 2008, 5, 17.
dcterms.referencesB. Du, L. Jiang, Q. Xia, L. Zhong, Synergistic Inhibitory effects of curcumin and 5- fluorouracil on the growth of the human colon cancer cell Line HT-29. Chemotherapy 2006, 52, 23-28
dcterms.referencesLev-Ari, S., Starr, A., Vexler, A., Karaush, V., Loew, V., Greif, J., Fenig, E., Aderka, D., Ben-Yosef, R. Inhibition of pancreatic and lung adenocarcinoma cell survival by curcumin is associated with increased apoptosis, down-regulation of COX-2 and EGFR and inhibition of Erk1/2 activity. Anticancer Res. 2006, 26, 4423-4430.
dcterms.referencesGafner, S., Lee, S.K., Cuendet, M., Barthélémy, S., Vergnes, L., Labidalle, S., Mehta, R.G., Boone, C.W., Pezzuto, J.M. Biologic evaluation of curcumin and structural derivatives in cancer chemoprevention model systems. Phytochemistry. 2004, 65 2849-2859.
dcterms.referencesGafner, S., Lee, S.K., Cuendet, M., Barthélémy, S., Vergnes, L., Labidalle, S., Mehta, R.G., Boone, C.W., Pezzuto, J.M. Biologic evaluation of curcumin and structural derivatives in cancer chemoprevention model systems. Phytochemistry. 2004, 65 2849-2859.
dcterms.referencesJ. Hong, M. Bose, J. Ju, J.H. Ryu, X. Chen, S. Sang, M.J. Lee, C.S. Yang, Modulation of arachidonic acid metabolism by curcumin and related β-diketone derivatives: effects on cytosolic phospholipase A2, cyclooxygenases and 5-lipoxygenase. Carcinogenesis 2004, 25, 1671-1679.
dcterms.referencesKim, B.H., Kang, K.S., Lee, Y.S. Effect of retinoids on LPS-induced COX-2 expression and COX-2 associated PGE2 release from mouse peritoneal macrophages and TNF-α release from rat peripheral blood mononuclear cells. Toxicol Let. 2004, 150, 191-201
dcterms.referencesLiang, Y.C., Huang, Y.T., Tsai, S.H., Lin-Shiau, S.Y., Chen, C.F., Lin, J.K. Suppression of inducible cyclooxygenase and inducible nitric oxide synthase by apigenin and related flavonoids in mouse macrophages. Carcinogenesis 1999, 20 1945-1952.
dcterms.referencesDia, V.P., Berhow, M.A., Gonzalez de mejia, E. Bowman-birk inhibitor and genistein among soy compounds that synergistically inhibit nitric oxide and prostaglandin E2 pathways in lipopolysaccharide-induced macrophages. J Agric Food Chem. 2008, 56, 11707-117
dcterms.referencesF. Ye, J. Wu, T. Dunn, J. Yi, X. Tong, D. Zhang. Inhibition of cyclooxygenase-2 activity in head and neck cancer cells by genistein. Cancer Lett. 2004, 211, 39-46.
dcterms.referencesLiang, Y.C., Tsai, S.H., Tsai, D.C., Lin-Shiau, S.Y., Lin, J.K. Suppression of inducible cyclooxygenase and nitric oxide synthase through activation of peroxisome proliferator-activated receptor-Q by flavonoids in mouse macrophages. FEBS Lett. 2001, 496, 12-18.
dcterms.referencesWang, H., Nair, M.G., Strasburg, G.M., Chang, Y.C., Booren, A.M., Gray, J.I., DeWitt, D.L. Antioxidant and antiinflammatory activities of anthocyanins and their aglycon, cyanidin, from tart cherries. J. Nat Prod. 1999, 62, 294-296.
dcterms.referencesSeeram, N.P., Zhang, Y., Nair, M.G. Inhibition of proliferation of human cancer cells and cyclooxygenase enzymes by anthocyanidins and catechins. Nutr. Cancer. 2003, 46 101- 106.
dcterms.referencesRingbom, T., Huss, U. Å. Stenholm, S. Flock, L. Skattebøl, P. Perera, L. Bohlin. COX-2 inhibitory effects of naturally occurring and modified fatty acids. J Nat Prod 2001, 64, 745-749.
dcterms.referencesCalviello, G., Di Nicuolo, F., Gragnoli, S., Piccioni, E., Serini, S., Maggiano, N., Tringali, G., Navarra, P., Ranelletti, F.O., Palozza, P. n-3 PUFAs reduce VEGF expression in human colon cancer cells modulating the COX-2/PGE2 induced ERK-1 and -2 and HIF-1a induction pathway. Carcinogenesis. 2004, 25, 2303-2310.
dcterms.referencesTakano-Ishikawaa, Y., Gotoa, M., Yamakia, K. Structure–activity relations of inhibitory effects of various flavonoids on lipopolysaccharide-induced prostaglandin E2 production in rat peritoneal macrophages: comparison between subclasses of flavonoids. Phytomedicine 2006, 13, 310-317.
dcterms.referencesTjendraputra, E., Tran, V.H., Biu-Brennan, D., Roufogalis B.D., Duke, C.C. Effect of ginger constituents and synthetic analogues on cyclooxygenase-2 enzyme in intact cells. Bioorg Chem. 2001, 29, 156-163.
dcterms.referencesCao, H., Yu, R., Tao, Y., Nikolic, D., van Breemen, R.B. Measurement of cyclooxygenase inhibition using liquid chromatography–tandem mass spectrometry, J Pharm Biomed Anal. 2011, 54, 230-235.
dcterms.referencesY.J. Surh, K.S. Chun, H.H. Cha, S.S. Han, Y.S. Keum, K.K. Park, S.S. Lee, Molecular mechanisms underlying chemopreventive activities of anti-inflammatory phytochemicals: down-regulation of COX-2 and iNOS through suppression of NF-κβ activation. Mutat Res. 2001, 480, 243-268.
dcterms.referencesKole, L., Giri, B., Manna, S.K., Pal, B., Ghosh, S. Biochanin-A, an isoflavon, showed anti-proliferative and anti-inflammatory activities through the inhibition of iNOS expression, p38-MAPK and ATF-2 phosphorylation and blocking NF-κβ nuclear translocation. Eur J Pharmacol. 2011, 653, 8-15.
dcterms.referencesChung, E.Y., Kim, B.H., Hong, J.T., Lee, C.K., Ahn, B., Nam, S.Y., Han, S.B., Kim, Y. Resveratrol down-regulates interferon-γ-inducible inflammatory genes in macrophages: molecular mechanism via decreased STAT-1 activation, J Nutr Biochem. 2011, 22, 902-909.
dcterms.referencesWang, H.M., Zhao, Y.X., Zhang, S., Liu, G.D., Kang, W.Y., Tang, H.D.. Ding, J.Q., Chen, S.D. PPARgamma agonist curcumin reduces the amyloid-beta-stimulated inflammatory responses in primary astrocytes, J Alzheimers Dis. 2010, 20 1189-1199.
dcterms.referencesGarg, R., Gupta, S., Maru, G.B. Dietary curcumin modulates transcriptional regulators of phase I and phase II enzymes in benzo[a]pyrene-treated mice: mechanism of its anti-initiating action, Carcinogenesis 2008, 29, 1022-1032.
dcterms.referencesAhn, J., Lee, H., Kim, S., Ha, T. Curcumin-induced suppression of adipogenic differentiation is accompanied by activation of Wnt/beta-catenin signaling, Am. J. Physiol. Cell Physiol. 2010, 298, 1510-1516.
dcterms.referencesDay, B.J. Antioxidants as potential therapeutics for lung fibrosis. Antiox Red Sign. 2008, 10, 355-70.
dcterms.referencesSong, F.-L., Gan, R.-Y., Zhang, Y., Xiao, Q., Kuang, L., Li, H.-B. Total phenolic contents and antioxidant capacities of selected Chinese medicinal plants. Int J Mol Sci. 2010, 11, 2362-72
dcterms.referencesLü, J.M., Lin, P.H., Yao, Q., Chen, C. Chemical and molecular mechanisms of antioxidants: experimental approaches and model systems. J Cell Mol Med. 2010, 14, 840- 60.
dcterms.referencesKamat, C.D., Gadal, S., Mhatre, M., Williamson, K.S., Pye, Q.N., Hensley, K. Antioxidants in central nervous system diseases: preclinical promise and translational challenges. J Alzheimers Dis. 2008, 15, 473-93
dcterms.referencesKris-Etherton, P.M., Hecker, K.D., Bonanome, A., Coval, S.M., Binkoski, A.E., Hilpert, K.F., et al. Bioactive compounds in foods: their role in the prevention of cardiovascular disease and cancer. Am J Med. 2002, 113, 71-88.
dcterms.referencesDelcourt, C., Korobelnik, J.-F., Barberger-Gateau, P., Delyfer, M.-N., Rougier, M.-B., Le Goff, M., et al. Nutrition and age-related eye diseases: the Alienor (Antioxydants, Lipides Essentiels, Nutrition et maladies OculaiRes) Study. J Nutr Health Aging. 2010, 14, 854-61.
dcterms.referencesWang, S.Y., Chen, C.-T., Sciarappa, W., Wang, C.Y., Camp, M.J. Fruit quality, antioxidant capacity, and flavonoid content of organically and conventionally grown blueberries. J Agric Food Chem. 2008, 56, 5788-94.
dcterms.referencesXia, E.-Q., Deng, G.-F., Guo, Y.-J., Li, H.-B. Biological activities of polyphenols from grapes. Int J Mol Sci. 2010, 11, 622-46.
dcterms.referencesHeim, K.E., Tagliaferro, A.R., Bobilya, D.J. Flavonoid antioxidants: chemistry, metabolism and structure-activity relationships. J Nutr Biochem. 2002, 13, 572-84.
dcterms.referencesHammerstone, J.F., Lazarus, S.A., Schmitz, H.H. Procyanidin content and variation in some commonly consumed foods. J Nutr. 2000, 130, 2086S-92S
dcterms.referencesNichols, J.A., Katiyar, S.K. Skin photoprotection by natural polyphenols: antiinflammatory, antioxidant and DNA repair mechanisms. Arch Dermatol Res. 2010, 302, 71- 83.
dcterms.referencesRahman, M.A., Amin, A.R., Shin, D.M. Chemopreventive potential of natural compounds in head and neck cancer. Nutr Cancer. 2010, 62, 973-87
dcterms.referencesRahman, M.A., Amin, A.R., Shin, D.M. Chemopreventive potential of natural compounds in head and neck cancer. Nutr Cancer. 2010, 62, 973-87
dcterms.referencesHooper, L., Kroon, P.A., Rimm, E.B., Cohn, J.S., Harvey, I., Le Cornu, K.A., et al. Flavonoids, flavonoid-rich foods, and cardiovascular risk: a meta-analysis of randomized controlled trials. Am J Clin Nutr. 2008, 88, 38-50.
dcterms.referencesHooper, L., Kroon, P.A., Rimm, E.B., Cohn, J.S., Harvey, I., Le Cornu, K.A., et al. Flavonoids, flavonoid-rich foods, and cardiovascular risk: a meta-analysis of randomized controlled trials. Am J Clin Nutr. 2008, 88, 38-50.
dcterms.referencesZaslaver, M., Offer, S., Kerem, Z., Stark, A.H., Weller, J.I., Eliraz, A., et al. Natural compounds derived from foods modulate nitric oxide production and oxidative status in epithelial lung cells. J Agric Food Chem. 2005, 53, 9934-9.
dcterms.referencesBian, K., Murad, F. Nitric oxide (NO)--biogeneration, regulation, and relevance to human diseases. Front Biosci. 2003, 8, d264-78.
dcterms.referencesGarcin, E.D., Arvai, A.S., Rosenfeld, R.J., Kroeger, M.D., Crane, B.R., Andersson, G., et al. Anchored plasticity opens doors for selective inhibitor design in nitric oxide synthase. Nat Chem Biol. 2008, 4, 700-7.
dcterms.referencesRosenfeld, R.J., Garcin, E.D., Panda, K., Andersson, G., Åberg, A., Wallace, A.V., et al. Conformational changes in nitric oxide synthases induced by chlorzoxazone and nitroindazoles: crystallographic and computational analyses of inhibitor potency. Biochemistry. 2002, 41, 13915-25.
dcterms.referencesLi, D., Stuehr, D.J., Yeh, S.-R., Rousseau, D.L. Heme distortion modulated by ligandprotein interactions in inducible nitric-oxide synthase. J Biol Chem. 2004, 279, 26489-99.
dcterms.referencesPan, M.-H., Lai, C.-S., Dushenkov, S., Ho, C.-T. Modulation of inflammatory genes by natural dietary bioactive compounds. J Agric Food Chem. 2009, 57, 4467-77.
dcterms.referencesMay, A., Zacharias, M. Accounting for global protein deformability during protein– protein and protein–ligand docking. BBA- Protein Proteom. 2005, 1754, 225-31.
dcterms.referencesMaldonado-Rojas, W., Olivero-Verbel, J. Potential interaction of natural dietary bioactive compounds with COX-2. J Mol Graph Model. 2011, 30, 157-66.
dcterms.referencesMaldonado-Rojas, W., Olivero-Verbel, J. Potential interaction of natural dietary bioactive compounds with COX-2. J Mol Graph Model. 2011, 30, 157-66.
dcterms.referencesTrott, O., Olson, A.J. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem. 2010, 31, 455-61.
dcterms.referenceswww.pdb.org/pdb/home/home.do. Protein Data Bank (RCSB PDB). 2011.
dcterms.referencesTripos:St. Louis, M. SYBYL 8.1. molecular modeling software. Tripos: St. Louis, MO, USA, 2007., USA, 2007., Vol. 8.1.
dcterms.referencesHwang, Y.P., Choi, J.H., Yun, H.J., Han, E.H., Kim, H.G., Kim, J.Y., et al. Anthocyanins from purple sweet potato attenuate dimethylnitrosamine-induced liver injury in rats by inducing Nrf2-mediated antioxidant enzymes and reducing COX-2 and iNOS expression. Food Chem Toxicol. 2011, 49, 93-9.
dcterms.referencesLi, J., Shi, R., Yang, C., Zhu, X. Exploration of the binding of benzimidazole-biphenyl derivatives to hemoglobin using docking and molecular dynamics simulation. Int J Biol Macromol. 2011, 48, 20-6.
dcterms.referencesFrisch, M., Trucks, G., Schlegel, H., Scuseria, G., Robb, M., Cheeseman, J., et al. Gaussian 03, revision B. 05; Gaussian. Inc., Pittsburgh, PA. 2003, 12478. [28] Guha, R., Howard, M.T., Hutchison, G.R., Murray-Rust, P., Rzepa, H., Steinbeck, C., et al. The Blue Obelisk-interoperability in chemical informatics. J Chem Inf Mod. 2006, 46, 991-8.
dcterms.referencesSanner, M.F. Python: a programming language for software integration and development. J Mol Graph Model. 1999, 17, 57-61.
dcterms.referencesWolber, G., Langer, T. LigandScout: 3-D pharmacophores derived from proteinbound ligands and their use as virtual screening filters. J Chem Inf Model. 2005, 45, 160-9.
dcterms.referencesDeLano, W.L. The PyMOL Molecular Graphics System; . DeLano Scientific LLC: San Carlos, C., USA, Ed., 1998–2003.
dcterms.referenceshttp://pubchem.ncbi.nlm.nih.gov. PubChem 2011.
dcterms.referencesNakane, M., Klinghofer, V., Kuk, J.E., Donnelly, J.L., Budzik, G.P., Pollock, J.S., et al. Novel potent and selective inhibitors of inducible nitric oxide synthase. Mol Pharmacol. 1995, 47, 831-4
dcterms.referencesDavey, D.D., Adler, M., Arnaiz, D., Eagen, K., Erickson, S., Guilford, W., et al. Design, synthesis, and activity of 2-imidazol-1-ylpyrimidine derived inducible nitric oxide synthase dimerization inhibitors. J Med Chem. 2007, 50, 1146-57.
dcterms.referencesConnolly, S., Aberg, A., Arvai, A., Beaton, H.G., Cheshire, D.R., Cook, A.R., et al. 2- Aminopyridines as highly selective inducible nitric oxide synthase inhibitors. Differential binding modes dependent on nitrogen substitution. J Med Chem. 2004, 47, 3320-3.
dcterms.referencesZhou, Z., Wang, Y., Bryant, S.H. Computational analysis of the cathepsin B inhibitors activities through LR‐MMPBSA binding affinity calculation based on docked complex. J Comput Chem. 2009, 30, 2165-75
dcterms.referencesFeng, R., Ni, H.-M., Wang, S.Y., Tourkova, I.L., Shurin, M.R., Harada, H., et al. Cyanidin-3-rutinoside, a natural polyphenol antioxidant, selectively kills leukemic cells by induction of oxidative stress. J Biol Chem. 2007, 282, 13468-76.
dcterms.referencesŠarić, A., Sobočanec, S., Balog, T., Kušić, B., Šverko, V., Dragović-Uzelac, V., et al. Improved antioxidant and anti-inflammatory potential in mice consuming sour cherry juice (Prunus Cerasus cv. Maraska). Plant Foods Hum Nutr. 2009, 64, 231-7.
dcterms.referencesYin, F., Liu, J., Ji, X., Wang, Y., Zidichouski, J., Zhang, J. Silibinin: A novel inhibitor of Aβ aggregation. Neurochem Int. 2011, 58, 399-403.
dcterms.referencesAbenavoli, L., Capasso, R., Milic, N., Capasso, F. Milk thistle in liver diseases: past, present, future. Phytother Res. 2010, 24, 1423-32.
dcterms.referencesCheng, J.-C., Kan, L.-S., Chen, J.-T., Chen, L.-G., Lu, H.-C., Lin, S.-M., et al. Detection of cyanidin in different-colored peanut testae and identification of peanut cyanidin 3-sambubioside. J Agric Food Chem. 2009, 57, 8805-11.
dcterms.referencesWang, L.-S., Stoner, G.D. Anthocyanins and their role in cancer prevention. Cancer lett. 2008, 269, 281-90.
dcterms.referencesHosseinian, F.S., Beta, T. Saskatoon and wild blueberries have higher anthocyanin contents than other Manitoba berries. J Agric Food Chem. 2007, 55, 10832-8.
dcterms.referencesIchiyanagi, T., Shida, Y., Rahman, M.M., Hatano, Y., Konishi, T. Bioavailability and tissue distribution of anthocyanins in bilberry (Vaccinium myrtillus L.) extract in rats. J Agric Food Chem. 2006, 54, 6578-87
dcterms.referencesKalantari, H., Das, D.K. Physiological effects of resveratrol. Biofactors. 2010, 36, 401- 6.
dcterms.referencesReber, J.D., Eggett, D.L., Parker, T.L. Antioxidant capacity interactions and a chemical/structural model of phenolic compounds found in strawberries. Int J Food Sci Nutr. 2011, 62, 445-52.
dcterms.referencesChen, P.-N., Kuo, W.-H., Chiang, C.-L., Chiou, H.-L., Hsieh, Y.-S., Chu, S.-C. Black rice anthocyanins inhibit cancer cells invasion via repressions of MMPs and u-PA expression. Chem Biol Interact. 2006, 163, 218-29.
dcterms.referencesYao, Y., Sang, W., Zhou, M., Ren, G. Phenolic composition and antioxidant activities of 11 celery cultivars. J Food Sci. 2010, 75, C9-C13.
dcterms.referencesBernardes, W.A., Lucarini, R., Tozatti, M.G., Souza, M.G., Andrade Silva, M.L., da Silva Filho, A.A., et al. Antimicrobial activity of Rosmarinus officinalis against oral pathogens: relevance of carnosic acid and carnosol. Chem Biodivers. 2010, 7, 1835-40.
dcterms.references] Castellarin, S.D., Di Gaspero, G. Transcriptional control of anthocyanin biosynthetic genes in extreme phenotypes for berry pigmentation of naturally occurring grapevines. BMC Plant Biol. 2007, 7, 1.
dcterms.referencesDeziel, B.A., Patel, K., Neto, C., Gottschall‐Pass, K., Hurta, R.A. Proanthocyanidins from the American Cranberry (Vaccinium macrocarpon) inhibit matrix metalloproteinase‐2 and matrix metalloproteinase‐9 activity in human prostate cancer cells via alterations in multiple cellular signalling pathways. J Cell Biochem. 2010, 111, 742-54.
dcterms.referencesWang, P., Aronson, W.J., Huang, M., Zhang, Y., Lee, R.-P., Heber, D., et al. Green tea polyphenols and metabolites in prostatectomy tissue: implications for cancer prevention. Cancer Prev Res. 2010, 3, 985-93.
dcterms.referencesLehtonen, H.-M., Rantala, M., Suomela, J.-P., Viitanen, M., Kallio, H. Urinary excretion of the main anthocyanin in lingonberry (Vaccinium vitis-idaea), cyanidin 3-Ogalactoside, and its metabolites. J Agric Food Chem. 2009, 57, 4447-51
dcterms.referencesVasanthi, H.R., Mukherjee, S., Das, D.K. Potential health benefits of broccoli-a chemico-biological overview. Mini Rev Med Chem. 2009, 9, 749-59.
dcterms.referencesMullen, W., Edwards, C.A., Serafini, M., Crozier, A. Bioavailability of pelargonidin-3- O-glucoside and its metabolites in humans following the ingestion of strawberries with and without cream. J Agric Food Chem. 2008, 56, 713-9.
dcterms.referencesRasyid, A., Rahman, A.R.A., Jaalam, K., Lelo, A. Effect of different curcumin dosages on human gall bladder. Asia Pac J Clin Nutr. 2002, 11, 314-8.
dcterms.referencesLai, C.-S., Li, S., Chai, C.-Y., Lo, C.-Y., Ho, C.-T., Wang, Y.-J., et al. Inhibitory effect of citrus 5-hydroxy-3, 6, 7, 8, 3′, 4′-hexamethoxyflavone on 12-O-tetradecanoylphorbol 13- acetate-induced skin inflammation and tumor promotion in mice. Carcinogenesis. 2007, 28, 2581-8.
dcterms.referencesTheodosiou, M., Laudet, V., Schubert, M. From carrot to clinic: an overview of the retinoic acid signaling pathway. Cell Mol Life Sci. 2010, 67, 1423-45
dcterms.referencesLien, T.F., Yeh, H.S., Su, W.T. Effect of adding extracted hesperetin, naringenin and pectin on egg cholesterol, serum traits and antioxidant activity in laying hens. Arch Anim Nutr. 2008, 62, 33-43.
dcterms.referencesAlosi, J.A., McDonald, D.E., Schneider, J.S., Privette, A.R., McFadden, D.W. Pterostilbene inhibits breast cancer in vitro through mitochondrial depolarization and induction of caspase-dependent apoptosis. J Surgical Res. 2010, 161, 195-201.
dcterms.referencesStuetz, W., Prapamontol, T., Hongsibsong, S., Biesalski, H.-K. Polymethoxylated flavones, flavanone glycosides, carotenoids, and antioxidants in different cultivation types of tangerines (Citrus reticulata Blanco cv. Sainampueng) from Northern Thailand. J Agric Food Chem. 2010, 58, 6069-74
dcterms.referencesBasini, G., Bussolati, S., Santini, S., Grasselli, F. The impact of the phyto‐oestrogen genistein on swine granulosa cell function. J Anim Physiol Anim Nutr. 2010, 94, e374-e82.
dcterms.referencesMann, N.J., O’Connell, S.L., Baldwin, K.M., Singh, I., Meyer, B.J. Effects of seal oil and tuna-fish oil on platelet parameters and plasma lipid levels in healthy subjects. Lipids. 2010, 45, 669-81.
dcterms.referencesWeng, C.J., Wu, C.F., Huang, H.W., Ho, C.T., Yen, G.C. Anti‐invasion effects of 6‐ shogaol and 6‐gingerol, two active components in ginger, on human hepatocarcinoma cells. Mol Nutr Food Res. 2010, 54, 1618-27.
dcterms.referencesVisanji, J.M., Duthie, S.J., Pirie, L., Thompson, D.G., Padfield, P.J. Dietary isothiocyanates inhibit Caco-2 cell proliferation and induce G2/M phase cell cycle arrest, DNA damage, and G2/M checkpoint activation. Journal Nutr. 2004, 134, 3121-6.
dcterms.referencesVogel, J.T., Tieman, D.M., Sims, C.A., Odabasi, A.Z., Clark, D.G., Klee, H.J. Carotenoid content impacts flavor acceptability in tomato (Solanum lycopersicum). J Sci Food Agric. 2010, 90, 2233-40.
dcterms.referencesAli, M.S., Saleem, M., Ahmad, W., Parvez, M., Yamdagni, R. A chlorinated monoterpene ketone, acylated β-sitosterol glycosides and a flavanone glycoside from Mentha longifolia (Lamiaceae). Phytochemistry. 2002, 59, 889-95.
dcterms.referencesSkupinska, K., Misiewicz-Krzeminska, I., Lubelska, K., Kasprzycka-Guttman, T. The effect of isothiocyanates on CYP1A1 and CYP1A2 activities induced by polycyclic aromatic hydrocarbons in Mcf7 cells. Toxicol In Vitro. 2009, 23, 763-71.
dcterms.referencesThürmann, P.A., Steffen, J., Zwernemann, C., Aebischer, C.-P., Cohn, W., Wendt, G., et al. Plasma concentration response to drinks containing β-carotene as carrot juice or formulated as a water dispersible powder. Eur J Nutr. 2002, 41, 228-35.
dcterms.referencesBurns-Whitmore, B., Haddad, E., Sabaté, J., Jaceldo-Siegl, K., Tanzman, J., Rajaram, S. Effect of n-3 fatty acid enriched eggs and organic eggs on serum lutein in freeliving lacto-ovo vegetarians. Eur J Clin Nutr. 2010, 64, 1332-7.
dcterms.referencesSalado, C., Olaso, E., Gallot, N., Valcarcel, M., Egilegor, E., Mendoza, L., et al. Resveratrol prevents inflammation-dependent hepatic melanoma metastasis by inhibiting the secretion and effects of interleukin-18. J Transl Med. 2011, 9, 59.
dcterms.referencesAu, A., Hasenwinkel, J., Frondoza, C. Silybin inhibits interleukin‐1β‐induced production of pro‐inflammatory mediators in canine hepatocyte cultures. J Vet Pharmacol Ther. 2011, 34, 120-9.
dcterms.referencesMauray, A., Felgines, C., Morand, C., Mazur, A., Scalbert, A., Milenkovic, D. Bilberry anthocyanin-rich extract alters expression of genes related to atherosclerosis development in aorta of apo E-deficient mice. Nutr Metab Card Dis. 2012, 22, 72-80.
dcterms.referencesSeymour, E., Lewis, S.K., Urcuyo-Llanes, D.E., Tanone, I.I., Kirakosyan, A., Kaufman, P.B., et al. Regular tart cherry intake alters abdominal adiposity, adipose gene transcription, and inflammation in obesity-prone rats fed a high fat diet. J Med Food. 2009, 12, 935-42.
dcterms.referencesKarlsen, A., Retterstøl, L., Laake, P., Paur, I., Kjølsrud-Bøhn, S., Sandvik, L., et al. Anthocyanins inhibit nuclear factor-κB activation in monocytes and reduce plasma concentrations of pro-inflammatory mediators in healthy adults. J Nutr. 2007, 137, 1951-4.
dcterms.referencesRamasamy, K., Dwyer-Nield, L.D., Serkova, N.J., Hasebroock, K.M., Tyagi, A., Raina, K., et al. Silibinin prevents lung tumorigenesis in wild-type but not in iNOS−/− mice: potential of real-time micro-CT in lung cancer chemoprevention studies. Clin Cancer Res. 2011, 17, 753-61.
dcterms.referencesChittezhath, M., Deep, G., Singh, R.P., Agarwal, C., Agarwal, R. Silibinin inhibits cytokine-induced signaling cascades and down-regulates inducible nitric oxide synthase in human lung carcinoma A549 cells. Mol Cancer Ther. 2008, 7, 1817-26.
dcterms.referencesZikri, N.N., Riedl, K.M., Wang, L.-S., Lechner, J., Schwartz, S.J., Stoner, G.D. Black raspberry components inhibit proliferation, induce apoptosis, and modulate gene expression in rat esophageal epithelial cells. Nutr Cancer. 2009, 61, 816-26.
dcterms.referencesJin, X.-H., Ohgami, K., Shiratori, K., Suzuki, Y., Koyama, Y., Yoshida, K., et al. Effects of blue honeysuckle (Lonicera caerulea L.) extract on lipopolysaccharide-induced inflammation in vitro and in vivo. Exp Eye Res. 2006, 82, 860-7.
dcterms.referencesKhan, N., Afaq, F., Kweon, M.-H., Kim, K., Mukhtar, H. Oral consumption of pomegranate fruit extract inhibits growth and progression of primary lung tumors in mice. Cancer Res. 2007, 67, 3475-82.
dcterms.referencesLaua, F., Joseph, J., Mcdonald, J., Kalt, W. Attenuation of iNOS and COX-2 by blueberry polyphenols. J Funct Foods. 2009, 1, 274-83.
dcterms.referencesPergola, C., Rossi, A., Dugo, P., Cuzzocrea, S., Sautebin, L. Inhibition of nitric oxide biosynthesis by anthocyanin fraction of blackberry extract. Nitric oxide. 2006, 15, 30-9
dcterms.referencesLeiro, J.M., Alvarez, E., Arranz, J.A., Siso, I.G., Orallo, F. In vitro effects of mangiferin on superoxide concentrations and expression of the inducible nitric oxide synthase, tumour necrosis factor-α and transforming growth factor-β genes. Biochem Pharmacol. 2003, 65, 1361-71.
dcterms.referencesChin, Y.-W., Park, E.Y., Seo, S.-Y., Yoon, K.-D., Ahn, M.-J., Suh, Y.-G., et al. A novel iNOS and COX-2 inhibitor from the aerial parts of Rodgersia podophylla. Bioorg Med Chem Lett. 2006, 16, 4600-2.
dcterms.referencesOh, J.H., Lee, T.-J., Park, J.-W., Kwon, T.K. Withaferin A inhibits iNOS expression and nitric oxide production by Akt inactivation and down-regulating LPS-induced activity of NF-κB in RAW 264.7 cells. Eur J Pharmacol. 2008, 599, 11-7
dcterms.referencesCheshire, D.R., Åberg, A., Andersson, G.M., Andrews, G., Beaton, H.G., Birkinshaw, T.N., et al. The discovery of novel, potent and highly selective inhibitors of inducible nitric oxide synthase (iNOS). Bioorg Med Chem Lett. 2011, 21, 2468-71.
dcterms.referencesLe Bourdonnec, B., Leister, L.K., Ajello, C.A., Cassel, J.A., Seida, P.R., O’Hare, H., et al. Discovery of a series of aminopiperidines as novel iNOS inhibitors. Bioorg Med Chem Lett. 2008, 18, 336-43.
dcterms.referencesHare, A.A., Leng, L., Gandavadi, S., Du, X., Cournia, Z., Bucala, R., et al. Optimization of N-benzyl-benzoxazol-2-ones as receptor antagonists of macrophage migration inhibitory factor (MIF). Bioorg Med Chem Lett. 2010, 20, 5811-4.
dcterms.referencesRen, J., Singh, B.N., Huang, Q., Li, Z., Gao, Y., Mishra, P., et al. DNA hypermethylation as a chemotherapy target. Cell Signal. 2011, 23, 1082-93
dcterms.referencesVogt, A., Tamewitz, A., J Skoko, RP Sikorski, Giuliano, K., Lazo, J. The Benzo[c]phenanthridine Alkaloid, Sanguinarine, Is a Selective, Cell-active Inhibitor of Mitogen-activated Protein Kinase Phosphatase-1. J Biol Chem 2005, 280, 19078
dcterms.referencesVenza, M., Visalli, M., Catalano, T., Fortunato, C., Oteri, R., Teti, D., et al. Impact of DNA methyltransferases on the epigenetic regulation of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor expression in malignant melanoma. Biochem Biophys Res Commun. 2013, 441, 743-50.
dcterms.referencesFernandez, A.F., Huidobro, C., Fraga, M.F. De novo DNA methyltransferases: oncogenes, tumor suppressors, or both? Trends Genet. 2012, 28, 474-9
dcterms.referencesSun, X., He, Y., Huang, C., Ma, T.-T., Li, J. The epigenetic feedback loop between DNA methylation and microRNAs in fibrotic disease with an emphasis on DNA methyltransferases. Cell Signal. 2013, 25, 1870-6.
dcterms.referencesSanti, D.V., Garrett, C.E., Barr, P.J. On the mechanism of inhibition of DNA-cytosine methyltransferases by cytosine analogs. Cell. 1983, 33, 9-10
dcterms.referencesTaylor, S.M., Jones, P.A. Mechanism of action of eukaryotic DNA methyltransferase: Use of 5-azacytosine-containing DNA. J Mol Biol. 1982, 162, 679-92.
dcterms.referencesYoder, J.A., Soman, N.S., Verdine, G.L., Bestor, T.H. DNA (cytosine-5)- methyltransferases in mouse cells and tissues. studies with a mechanism-based probe. J Mol Biol. 1997, 270, 385-95.
dcterms.referencesYoo, J., Kim, J.H., Robertson, K.D., Medina-Franco, J.L. Molecular Modeling of Inhibitors of Human DNA Methyltransferase with a Crystal Structure: Discovery of a Novel DNMT1 Inhibitor. In: Advances in Protein Chemistry and Structural Biology, Christo, C., Tatyana, K.-C. Eds., Academic Press, 2012, Vol. Volume 87, pp. 219-47.
dcterms.referencesOkano, M., Bell, D.W., Haber, D.A., Li, E. DNA Methyltransferases Dnmt3a and Dnmt3b Are Essential for De Novo Methylation and Mammalian Development. Cell. 1999, 99, 247-57.
dcterms.referencesLi, J.-Y., Pu, M.-T., Hirasawa, R., Li, B.-Z., Huang, Y.-N., Zeng, R., et al. Synergistic Function of DNA Methyltransferases Dnmt3a and Dnmt3b in the Methylation of Oct4 and Nanog. Mol Cell Biol. 2007, 27, 8748-59.
dcterms.referencesDhawan, D., Ramos-Vara, J.A., Hahn, N.M., Waddell, J., Olbricht, G.R., Zheng, R., et al. DNMT1: An emerging target in the treatment of invasive urinary bladder cancer. Urol Oncol. 2013, 31, 1761-9.
dcterms.referencesCheng, P., Chen, H., Zhang, R.-P., Liu, S.-r., Zhou-Cun, A. Polymorphism in DNMT1 may modify the susceptibility to oligospermia. Reprod Biomed Online. 2014, 28, 644-9.
dcterms.referencesBian, E.-B., Huang, C., Wang, H., Chen, X.-X., Zhang, L., Lv, X.-W., et al. Repression of Smad7 mediated by DNMT1 determines hepatic stellate cell activation and liver fibrosis in rats. Toxicol Lett. 2014, 224, 175-85.
dcterms.referencesCastellano, S., Kuck, D., Sala, M., Novellino, E., Lyko, F., Sbardella, G. Constrained Analogues of Procaine as Novel Small Molecule Inhibitors of DNA Methyltransferase-1. J Med Chem. 2008, 51, 2321-5.
dcterms.referencesAsgatay, S., Champion, C., Marloie, G., Drujon, T., Senamaud-Beaufort, C., Ceccaldi, A., et al. Synthesis and Evaluation of Analogues of N-Phthaloyl-l-tryptophan (RG108) as Inhibitors of DNA Methyltransferase 1. J Med Chem. 2013, 57, 421-34.
dcterms.referencesSuzuki, T., Tanaka, R., Hamada, S., Nakagawa, H., Miyata, N. Design, synthesis, inhibitory activity, and binding mode study of novel DNA methyltransferase 1 inhibitors. Bioorg Med Chem Lett. 2010, 20, 1124-7.
dcterms.referencesValente, S., Liu, Y., Schnekenburger, M., Zwergel, C., Cosconati, S., Gros, C., et al. Selective Non-nucleoside Inhibitors of Human DNA Methyltransferases Active in Cancer Including in Cancer Stem Cells. J Med Chem. 2014, 57, 701-13
dcterms.referencesVan De Voorde, L., Speeckaert, R., Van Gestel, D., Bracke, M., De Neve, W., Delanghe, J., et al. DNA methylation-based biomarkers in serum of patients with breast cancer. Mutat Res Rev Mutat Res. 2012, 751, 304-25.
dcterms.referencesMai, A. Targeting Epigenetics in Drug Discovery. ChemMedChem. 2014, 9, 415-7
dcterms.referencesLyko, F., Brown, R. DNA Methyltransferase Inhibitors and the Development of Epigenetic Cancer Therapies. J Natl Cancer Inst. 2005, 97, 1498-506.
dcterms.referencesBillam, M., Sobolewski, M., Davidson, N. Effects of a novel DNA methyltransferase inhibitor zebularine on human breast cancer cells. Breast Cancer Res Treat. 2010, 120, 581- 92.
dcterms.referencesGray, S., Baird, A., O'Kelly, F., Nikolaidis, G., Almgren, M., Meunier, A., et al. Gemcitabine reactivates epigenetically silenced genes and functions as a DNA methyltransferase inhibitor. Int J Mol Med. 2012, 30 1505-11.
dcterms.referencesFandy, T.E., Jiemjit, A., Thakar, M., Rhoden, P., Suarez, L., Gore, S.D. Decitabine Induces Delayed Reactive Oxygen Species (ROS) Accumulation in Leukemia Cells and Induces the Expression of ROS Generating Enzymes. Clin Cancer Res. 2014, 20, 1249-58.
dcterms.referencesQuintas-Cardama, A., Santos, F.P.S., Garcia-Manero, G. Therapy with azanucleosides for myelodysplastic syndromes. Nat Rev Clin Oncol. 2010, 7, 433-44.
dcterms.referencesMéndez-Lucio, O., Tran, J., Medina-Franco, J.L., Meurice, N., Muller, M. Toward Drug Repurposing in Epigenetics: Olsalazine as a Hypomethylating Compound Active in a Cellular Context. ChemMedChem. 2014, 9, 560-5.
dcterms.referencesBrueckner, B., Garcia Boy, R., Siedlecki, P., Musch, T., Kliem, H.C., Zielenkiewicz, P., et al. Epigenetic Reactivation of Tumor Suppressor Genes by a Novel Small-Molecule Inhibitor of Human DNA Methyltransferases. Cancer Res. 2005, 65, 6305-11.
dcterms.referencesDatta, J., Ghoshal, K., Denny, W.A., Gamage, S.A., Brooke, D.G., Phiasivongsa, P., et al. A New Class of Quinoline-Based DNA Hypomethylating Agents Reactivates Tumor Suppressor Genes by Blocking DNA Methyltransferase 1 Activity and Inducing Its Degradation. Cancer Res. 2009, 69, 4277-85.
dcterms.references] Rilova, E., Erdmann, A., Gros, C., Masson, V., Aussagues, Y., Poughon-Cassabois, V., et al. Design, Synthesis and Biological Evaluation of 4-Amino-N-(4- aminophenyl)benzamide Analogues of Quinoline-Based SGI-1027 as Inhibitors of DNA Methylation. ChemMedChem. 2014, 9, 590-601.
dcterms.referencesMedina-Franco, J.L., López-Vallejo, F., Kuck, D., Lyko, F. Natural products as DNA methyltransferase inhibitors: a computer-aided discovery approach. Mol Divers. 2011, 15, 293–304.
dcterms.referencesCragg, G.M., Newman, D.J., Snader, K.M. Natural Products in Drug Discovery and Development. J Nat Prod. 1997, 60, 52-60.
dcterms.referencesMedina-Franco, J.L. ADVANCES IN COMPUTATIONAL APPROACHES FOR DRUG DISCOVERY BASED ON NATURAL PRODUCTS. Rev. Latinoamer. Quím. 2013, 41, 95-110.
dcterms.referencesLiu, Z., Xie, Z., Jones, W., Pavlovicz, R.E., Liu, S., Yu, J., et al. Curcumin is a potent DNA hypomethylation agent. Bioorg Med Chem Lett. 2009, 19, 706-9
dcterms.referencesVaid, M., Prasad, R., Singh, T., Jones, V., Katiyar, S.K. Grape seed proanthocyanidins reactivate silenced tumor suppressor genes in human skin cancer cells by targeting epigenetic regulators. Toxicol Appl Pharmacol. 2012, 263, 122-30.
dcterms.referencesFang, M.Z., Wang, Y., Ai, N., Hou, Z., Sun, Y., Lu, H., et al. Tea Polyphenol (−)- Epigallocatechin-3-Gallate Inhibits DNA Methyltransferase and Reactivates MethylationSilenced Genes in Cancer Cell Lines. Cancer Res. 2003, 63, 7563-70.
dcterms.referencesXie, Q., Bai, Q., Zou, L.-Y., Zhang, Q.-Y., Zhou, Y., Chang, H., et al. Genistein inhibits DNA methylation and increases expression of tumor suppressor genes in human breast cancer cells. Genes Chromosomes Cancer. 2014, 53, 422-31.
dcterms.referencesGarcía, J., Franci, G., Pereira, R., Benedetti, R., Nebbioso, A., Rodríguez-Barrios, F., et al. Epigenetic profiling of the antitumor natural product psammaplin A and its analogues. Bioorg Med Chem. 2011, 19, 3637-49.
dcterms.referencesAgarwal, S., Amin, K., Jagadeesh, S., Baishay, G., Rao, P., Barua, N., et al. Mahanine restores RASSF1A expression by down-regulating DNMT1 and DNMT3B in prostate cancer cells. Mol Cancer. 2013, 12, 99.
dcterms.referencesLee, W.J., Zhu, B.T. Inhibition of DNA methylation by caffeic acid and chlorogenic acid, two common catechol-containing coffee polyphenols. Carcinogenesis. 2006, 27, 269-77
dcterms.referencesFagan, R.L., Cryderman, D.E., Kopelovich, L., Wallrath, L.L., Brenner, C. Laccaic Acid A Is a Direct, DNA-competitive Inhibitor of DNA Methyltransferase 1. J Biol Chem. 2013, 288, 23858-67.
dcterms.referencesSiedlecki, P., Boy, R.G., Musch, T., Brueckner, B., Suhai, S., Lyko, F., et al. Discovery of Two Novel, Small-Molecule Inhibitors of DNA Methylation. J Med Chem. 2005, 49, 678-83.
dcterms.referencesMedina-Franco, J., Yoo, J. Docking of a novel DNA methyltransferase inhibitor identified from high-throughput screening: insights to unveil inhibitors in chemical databases. Mol Divers. 2013, 17, 337-44.
dcterms.referencesSingh, N., Dueñas-González, A., Lyko, F., Medina-Franco, J.L. Molecular Modeling and Molecular Dynamics Studies of Hydralazine with Human DNA Methyltransferase 1. ChemMedChem. 2009, 4, 792-9.
dcterms.referencesMedina-Franco, J., Méndez-Lucio, O., Yoo, J. Rationalization of Activity Cliffs of a Sulfonamide Inhibitor of DNA Methyltransferases with Induced-Fit Docking. Int J Mol Sci. 2014, 15, 3253-61.
dcterms.referencesTripos. Sybyl X. In: 1699S Hanley Rd, St. Louis, MO, 2005
dcterms.referencesMauri, A., Consonni, V., Pavan, M., Todeschini, R. DRAGON software: An easy approach to molecular descriptor calculations. Match-Commun Math Co. 2006, 56, 237-48
dcterms.referencesStatSoft. Statistica 8.0.360 In: Tulsa, OK, 2007.
dcterms.referencesMeneses-Marcel, A., Marrero-Ponce, Y., Machado-Tugores, Y., Montero-Torres, A., Pereira, D.M., Escario, J.A., et al. A linear discrimination analysis based virtual screening of trichomonacidal lead-like compounds: Outcomes of in silico studies supported by experimental results. Bioorg Med Chem Lett. 2005, 15, 3838-43.
dcterms.referencesJ W Godden, F L Stahura, Bajorath, J. Variability of Molecular Descriptors in Compound Databases Revealed by Shannon Entropy Calculations. J Chem Inf Comput Sci. 2000, 40, 796-800
dcterms.referencesBarigye, S.J., Marrero-Ponce, Y., Martínez López, Y., Artiles Martínez, L.M., PinoUrias, R.W., Martínez Santiago, O., et al. Relations Frequency Hypermatrices in Mutual, Conditional and Joint Entropy-Based Information Indices. J. Comput. Chem. 2013, 34, 259- 74
dcterms.referencesTrott, O., Olson, A.J. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem. 2010, 31, 455-61.
dcterms.referencesTuccinardi, T., Poli, G., Romboli, V., Giordano, A., Martinelli, A. Extensive consensus docking evaluation for ligand pose prediction and virtual screening studies. J. Chem. Inf. Model . 2014, 54, 2980-6.
dcterms.referencesCarrasco, M.P., Gut, J., Rodrigues, T., Ribeiro, M.H.L., Lopes, F., Rosenthal, P.J., et al. Exploring the Molecular Basis of Qobc1 Complex Inhibitors Activity to Find Novel Antimalarials Hits. Mol Inform. 2013, 32, 659-70.
dcterms.referencesJain, A. Surflex-Dock 2.1: Robust performance from ligand energetic modeling, ring flexibility, and knowledge-based search. J Comput Aided Mol Des. 2007, 21, 281-306.
dcterms.referencesArnatt, C.K., Zhang, Y. G Protein-Coupled Estrogen Receptor (GPER) Agonist Dual Binding Mode Analyses Toward Understanding of Its Activation Mechanism: A Comparative Homology Modeling Approach. Mol Inform. 2013, 32, 647-58.
dcterms.referencesMaldonado-Rojas, W., Olivero-Verbel, J. Food-Related Compounds That Modulate Expression of Inducible Nitric Oxide Synthase May Act as Its Inhibitors. Molecules. 2012, 17, 8118-35.
dcterms.referencesMaldonado-Rojas, W., Olivero-Verbel, J. Potential interaction of natural dietary bioactive compounds with COX-2. J Mol Graph Model. 2011, 30, 157-66.
dcterms.referencesDeLano, W.L. The PyMOL Molecular Graphics System. LLC, D.S. Ed., San Carlos, CA, USA, 1998–2003.
dcterms.referencesNguyen, E.D., Norn, C., Frimurer, T.M., Meiler, J. Assessment and challenges of ligand docking into comparative models of G-protein coupled receptors. PloS one. 2013, 8, e67302.
dcterms.referencesWolber, G., Langer, T. LigandScout: 3-D Pharmacophores Derived from ProteinBound Ligands and Their Use as Virtual Screening Filters. J Chem Inf Model. 2004, 45, 160- 9.
dcterms.referencesVuorinen, A., Nashev, L.G., Odermatt, A., Rollinger, J.M., Schuster, D. Pharmacophore Model Refinement for 11β-Hydroxysteroid Dehydrogenase Inhibitors: Search for Modulators of Intracellular Glucocorticoid Concentrations. Mol Inform. 2014, 33, 15-25.
dcterms.referencesBarigye, S.J., Pino Urias, R.W., Marrero-Ponce, Y., et al. IMMAN (Information Theory based Chemometric Analysis). CAMD-BIR Unit, Santa Clara 2013.
dcterms.referencesPortugal, J. Evaluation of molecular descriptors for antitumor drugs with respect to noncovalent binding to DNA and antiproliferative activity. BMC Pharmacol. 2009, 9, 11.
dcterms.referencesShi, L.M., Fan, Y., Myers, T.G., O'Connor, P.M., Paull, K.D., Friend, S.H., et al. Mining the NCI Anticancer Drug Discovery Databases: Genetic Function Approximation for the QSAR Study of Anticancer Ellipticine Analogues. J Chem Inf Comput Sci. 1998, 38, 189- 99.
dcterms.referencesZhang, S., Wei, L., Bastow, K., Zheng, W., Brossi, A., Lee, K.-H., et al. Antitumor Agents 252. Application of validated QSAR models to database mining: discovery of novel tylophorine derivatives as potential anticancer agents. J Comput Aided Mol Des. 2007, 21, 97-112.
dcterms.referencesMontero-Torres, A., García-Sánchez, R.N., Marrero-Ponce, Y., Machado-Tugores, Y., Nogal-Ruiz, J.J., Martínez-Fernández, A.R., et al. Non-stochastic quadratic fingerprints and LDA-based QSAR models in hit and lead generation through virtual screening: theoretical and experimental assessment of a promising method for the discovery of new antimalarial compounds. Eur J Med Chem. 2006, 41, 483-93.
dcterms.referencesCasañola-Martin, G.M., Marrero-Ponce, Y., Khan, M.T.H., Khan, S.B., Torrens, F., Pérez-Jiménez, F., et al. Bond-Based 2D Quadratic Fingerprints in QSAR Studies: Virtual and In vitro Tyrosinase Inhibitory Activity Elucidation. Chem Biol Drug Des. 2010, 76, 538-45.
dcterms.referencesGalvez-Llompart, M., Recio Iglesias, M., Gálvez, J., García-Domenech, R., DOI. Novel potential agents for ulcerative colitis by molecular topology: suppression of IL-6 production in Caco-2 and RAW 264.7 cell lines. Mol Divers. 2013, 17, 573–93.
dcterms.referencesYoo, J., Choi, S., Medina-Franco, J.L. Molecular modeling studies of the novel inhibitors of DNA methyltransferases SGI-1027 and CBC12: Implications for the mechanism of inhibition of DNMTs. PloS one. 2013, 8, e62152.
dcterms.referencesHare, A.A., Leng, L., Gandavadi, S., Du, X., Cournia, Z., Bucala, R., et al. Optimization of i N-benzyl-benzoxazol-2-ones as receptor antagonists of macrophage migration inhibitory factor (MIF). Bioorg Med Chem Lett. 2010, 20, 5811-4.
dcterms.referencesJia, D., Jurkowska, R.Z., Zhang, X., Jeltsch, A., Cheng, X. Structure of Dnmt3a bound to Dnmt3L suggests a model for de novo DNA methylation. Nature. 2007, 449, 248-51
dcterms.referencesNair, S., Ziaullah, Z., Rupasinghe, H.P.V. Phloridzin fatty acid esters induce apoptosis and alters gene expression in human liver cancer cells (261.2). FASEB J. 2014, 28.
dcterms.referencesEhrenkranz, J.R., Lewis, N.G., Ronald Kahn, C., Roth, J. Phlorizin: a review. Diabetes Metab Res Rev. 2005, 21, 31-8.
dcterms.referencesZhao, H., Yakar, S., Gavrilova, O., Sun, H., Zhang, Y., Kim, H., et al. Phloridzin improves hyperglycemia but not hepatic insulin resistance in a transgenic mouse model of type 2 diabetes. Diabetes. 2004, 53, 2901-9.
dcterms.referencesNajafian, M., Jahromi, M.Z., Nowroznejhad, M.J., Khajeaian, P., Kargar, M.M., Sadeghi, M., et al. Phloridzin reduces blood glucose levels and improves lipids metabolism in streptozotocin-induced diabetic rats. Mol Biol Rep. 2012, 39, 5299-306.
dcterms.referencesWang, G.-E., Li, Y.-F., Wu, Y.-P., Tsoi, B., Zhang, S.-J., Cao, L.-F., et al. Phloridzin improves lipoprotein lipase activity in stress-loaded mice via AMPK phosphorylation. Int J Food Sci Nutr. 2014, 0, 1-7.
dcterms.referencesHuang, W.C., Chang, W.T., Wu, S.J., Xu, P.Y., Ting, N.C., Liou, C.J. Phloretin and phlorizin promote lipolysis and inhibit inflammation in mouse 3T3-L1 cells and in macrophageadipocyte co-cultures. Mol Nutr Food Res. 2013, 57, 1803-13.
dcterms.referencesWang, X., Zhang, S., Liu, Y., Spichtig, D., Kapoor, S., Koepsell, H., et al. Targeting of sodium–glucose cotransporters with phlorizin inhibits polycystic kidney disease progression in Han: SPRD rats. Kidney Int. 2013.
dcterms.referencesLeveen, H.H., Leveen, R.F., LeVeen, E.G. Inhibiting glucose transport. Google Patents, 1989.
dcterms.referencesBhartiya, H.P., Gupta, P.C. A chalcone glycoside from the flowers of Adhatoda vasica. Phytochemistry. 1982, 21, 247.
dcterms.referencesAlvarez, M., Debattista, N., Pappano, N. Antimicrobial activity and synergism of some substituted flavonoids. Folia Microbiol (Praha). 2008, 53, 23-8.
dcterms.referencesGuan, L.-P., Zhao, D.-H., Chang, Y., Wen, Z.-S., Tang, L.-M., Huang, F.-F. Synthesis of 2, 4-dihydroxychalcone derivatives as potential antidepressant effect. Drug Res (Stuttg). 2013, 63, 46-51.
dcterms.referencesXie, C., Sun, Y., Pan, C.-Y., Tang, L.-M., Guan, L.-P. 2, 4-Dihydroxychalcone derivatives as novel potent cell division cycle 25B phosphatase inhibitors and protein tyrosine phosphatase 1B inhibitors. Pharmazie. 2014, 69, 257-62.
dcterms.referencesYang, G.-M., Yan, R., Wang, Z.-X., Zhang, F.-F., Pan, Y., Cai, B.-C. Antitumor effects of two extracts from Oxytropis falcata on hepatocellular carcinoma in vitro and in vivo. Chin J Nat Med. 2013, 11, 519-24.
dcterms.referencesSoidrou, S.H., Bousta, D., Lachkar, M., Hassane, S.O., El Youbi-Hamsas, A., El Mansouri, L., et al. Immunomodulatory Activity of Phenolic Fraction from Piper Borbonense and Cassytha Filiformis Growing in Comoros Islands. In: Chemistry: The Key to our Sustainable Future, Springer, 2014, pp. 105-12.
dcterms.referencesPolicegoudra, R., Chattopadhyay, P., Aradhya, S., Shivaswamy, R., Singh, L., Veer, V. Inhibitory effect of Tridax procumbens against human skin pathogens. J Herb Med. 2014, 4, 83-8.
dcterms.referencesBartolome, A.P., Villaseñor, I.M., Yang, W.-C. Bidens pilosa L.(Asteraceae): Botanical properties, traditional uses, phytochemistry, and pharmacology. Evid Based Complement Alternat Med. 2013, 2013.
dcterms.referencesChang, S.-L., Chiang, Y.-M., Chang, C.L.-T., Yeh, H.-H., Shyur, L.-F., Kuo, Y.-H., et al. Flavonoids, centaurein and centaureidin, from Bidens pilosa, stimulate IFN-γ expression. J Ethnopharmacol. 2007, 112, 232-6.
dcterms.referencesStutzman-Engwall, K.J., Otten, S., Hutchinson, C.R. Regulation of secondary metabolism in Streptomyces spp. and overproduction of daunorubicin in Streptomyces peucetius. J Bacteriol. 1992, 174, 144-54.
dcterms.referencesAdès, L., Chevret, S., Raffoux, E., Guerci-Bresler, A., Pigneux, A., Vey, N., et al. Long-term follow-up of European APL 2000 trial, evaluating the role of cytarabine combined with ATRA and Daunorubicin in the treatment of nonelderly APL patients. Am J Hematol. 2013, 88, 556-9.
dcterms.referencesChim, C.S., Wong, A.S.Y., Kwong, Y.L. Epigenetic inactivation of INK4/CDK/RB cell cycle pathway in acute leukemias. Annals of Hematology. 2003, 82, 738-42.
dcterms.referencesSchreier, V.N., Pethő, L., Orbán, E., Marquardt, A., Petre, B.A., Mező, G., et al. Protein Expression Profile of HT-29 Human Colon Cancer Cells after Treatment with a Cytotoxic Daunorubicin-GnRH-III Derivative Bioconjugate. PloS one. 2014, 9, e94041.
dcterms.referencesZanette, R.A., Kontoyiannis, D.P. Pre-exposure of Candida species to cytarabine and daunorubicin does not affect their in vitro antifungal susceptibility and virulence in flies. Virulence. 2013, 4, 344.
dcterms.referencesChhablani, J., Nieto, A., Hou, H., Wu, E.C., Freeman, W.R., Sailor, M.J., et al. Oxidized porous silicon particles covalently grafted with daunorubicin as a sustained intraocular drug delivery system. Invest Ophthalmol Vis Sci. 2013, 54, 1268-79.
dcterms.referencesHasinoff, B.B., Yalowich, J.C., Ling, Y., Buss, J.L. The effect of dexrazoxane (ICRF187) on doxorubicin- and daunorubicin-mediated growth inhibition of Chinese hamster ovary cells. Anticancer Drugs. 1996, 7, 558-67.
dcterms.referencesIgina, E., Tulemisova, K., Golubchikov, V., Nikitina, E. Comparison of antibiotic complexes formed by cultures of Streptomyces griseoruber. Antibiot Khimioter. 1989, 34, 13- 6.
dcterms.referencesOki, T. New anthracycline antibiotics. Jpn J Antibiot. 1977, 30, 70-84.
dcterms.referencesNettleton Jr, D.E., Balitz, D.M., Doyle, T.W., Bradner, W.T., Johnson, D.L., O'Herron, F.A., et al. Antitumor agents from bohemic acid complex, III. The isolation of marcellomycin, musettamycin, rudolphomycin, mimimycin, collinemycin, alcindoromycin, and bohemamine. J Nat Prod. 1980, 43, 242-58.
dcterms.referencesGao, J., Xu, Y., Yang, Y., Yang, Y., Zheng, Z., Jiang, W., et al. Identification of upregulators of human ATP-binding cassette transporter A1 via high-throughput screening of a synthetic and natural compound library. J Biomol Screen. 2008.
dcterms.referencesZhang, H.-Z., Kasibhatla, S., Wang, Y., Herich, J., Guastella, J., Tseng, B., et al. Discovery, characterization and SAR of gambogic acid as a potent apoptosis inducer by a HTS assay. Bioorg Med Chem. 2004, 12, 309-17.
dcterms.referencesCai, S.X., Drewe, J.A., Kasibhatla, S., Tseng, B., Wang, Y., Zhang, H.Z. Gambogic acid, analogs and derivatives as activators of caspases and inducers of apoptosis. Google Patents, 2002
dcterms.referencesLi, R., Chen, Y., Zeng, L.-l., Shu, W.-x., Zhao, F., Wen, L., et al. Gambogic acid induces G0/G1 arrest and apoptosis involving inhibition of SRC-3 and inactivation of Akt pathway in K562 leukemia cells. Toxicology. 2009, 262, 98-105
dcterms.referencesFurdas, S.D., Kannan, S., Sippl, W., Jung, M. Small Molecule Inhibitors of Histone Acetyltransferases as Epigenetic Tools and Drug Candidates. Arch Pharm (Weinheim). 2012, 345, 7-21.
dcterms.referencesLi, Y., Tollefsbol, T.O. Impact on DNA methylation in cancer prevention and therapy by bioactive dietary components. Curr Med Chem. 2010, 17, 2141.
dcterms.referencesYe, Y., Stivers, J.T. Fluorescence-based high-throughput assay for human DNA (cytosine-5)-methyltransferase 1. Anal Biochem. 2010, 401, 168-72.
dcterms.referencesDi, L., Kerns, E.H. Biological assay challenges from compound solubility: strategies for bioassay optimization. Drug Discov Today. 2006, 11, 446-51.
dcterms.referencesMaldonado-Rojas, W., Olivero-Verbel, J., Marrero-Ponce, Y. Computational fishing of new DNA methyltransferase inhibitors from natural products. J Mol Graph Model. 2015, 60, 43-54.
dcterms.referencesRazin, A., Riggs, A.D. DNA methylation and gene function. Science. 1980, 210, 604- 10.
dcterms.referencesLi, E., Beard, C., Jaenisch, R. Role for DNA methylation in genomic imprinting. Nature. 1993, 366, 362-5.
dcterms.referencesFeil, R. Environmental and nutritional effects on the epigenetic regulation of genes. Mutat Res Fundam Mol Mech Mut. 2006, 600, 46-57.
dcterms.referencesSimmons, D. Epigenetic influence and disease. Nat Educ. 2008, 1, 6
dcterms.referencesEgger, G., Liang, G., Aparicio, A., Jones, P.A. Epigenetics in human disease and prospects for epigenetic therapy. Nature. 2004, 429, 457-63.
dcterms.referencesJones, P.A., Takai, D. The role of DNA methylation in mammalian epigenetics. Science. 2001, 293, 1068-70.
dcterms.referencesEsteller, M., Corn, P.G., Baylin, S.B., Herman, J.G. A gene hypermethylation profile of human cancer. Cancer Res. 2001, 61, 3225-9.
dcterms.referencesBaylin, S.B. DNA methylation and gene silencing in cancer. Nat Clin Pract Oncol. 2005, 2, S4-S11.
dcterms.referencesRobertson, K.D., Uzvolgyi, E., Liang, G., Talmadge, C., Sumegi, J., Gonzales, F.A., et al. The human DNA methyltransferases (DNMTs) 1, 3a and 3b: coordinate mRNA expression in normal tissues and overexpression in tumors. Nucleic Acids Res. 1999, 27, 2291-8.
dcterms.referencesGonzalo, S., Jaco, I., Fraga, M.F., Chen, T., Li, E., Esteller, M., et al. DNA methyltransferases control telomere length and telomere recombination in mammalian cells. Nat Cell Biol. 2006, 8, 416-24
dcterms.referencesSanti, D.V., Garrett, C.E., Barr, P.J. On the mechanism of inhibition of DNA-cytosine methyltransferases by cytosine analogs. Cell. 1983, 33, 9-10.
dcterms.referencesYoo, J., Kim, J.H., Robertson, K.D., Medina-Franco, J.L. Molecular modeling of inhibitors of human DNA methyltransferase with a crystal structure: discovery of a novel DNMT1 inhibitor. Adv Prot Chem Struct Biol. 2012, 87.
dcterms.referencesOkano, M., Bell, D.W., Haber, D.A., Li, E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 1999, 99, 247-57.
dcterms.referencesMeilinger, D., Fellinger, K., Bultmann, S., Rothbauer, U., Bonapace, I.M., Klinkert, W.E., et al. Np95 interacts with de novo DNA methyltransferases, Dnmt3a and Dnmt3b, and mediates epigenetic silencing of the viral CMV promoter in embryonic stem cells. EMBO reports. 2009, 10, 1259-64.
dcterms.referencesBird, A. DNA methylation patterns and epigenetic memory. Gen Develop. 2002, 16, 6-21
dcterms.referencesLaw, J.A., Jacobsen, S.E. Establishing, maintaining and modifying DNA methylation patterns in plants and animals. Nat Rev Genet. 2010, 11, 204-20.
dcterms.referencesPeng, D.-F., Kanai, Y., Sawada, M., Ushijima, S., Hiraoka, N., Kitazawa, S., et al. DNA methylation of multiple tumor-related genes in association with overexpression of DNA methyltransferase 1 (DNMT1) during multistage carcinogenesis of the pancreas. Carcinogenesis. 2006, 27, 1160-8.
dcterms.referencesJones, P.A., Baylin, S.B. The fundamental role of epigenetic events in cancer. Nat Rev Genet. 2002, 3, 415-28.
dcterms.referencesZhang, B., Hu, L., Zang, M., Wang, H., Zhao, W., Li, J., et al. Helicobacter pylori CagA induces tumor suppressor gene hypermethylation by upregulating DNMT1 via AKTNFκB pathway in gastric cancer development. Oncotarget. 2016, 7, 9788-800.
dcterms.referencesSi, X., Liu, Y., Lv, J., Ding, H., Zhang, X., Shao, L., et al. ERα propelled aberrant global DNA hypermethylation by activating the DNMT1 gene to enhance anticancer drug resistance in human breast cancer cells. Oncotarget. 2016.
dcterms.referencesZhong, B., Vatolin, S., Idippily, N.D., Lama, R., Alhadad, L.A., Reu, F.J., et al. Structural optimization of non-nucleoside DNA methyltransferase inhibitor as anti-cancer agent. Bioorg Med Chem Lett. 2016
dcterms.referencesFlotho, C., Claus, R., Batz, C., Schneider, M., Sandrock, I., Ihde, S., et al. The DNA methyltransferase inhibitors azacitidine, decitabine and zebularine exert differential effects on cancer gene expression in acute myeloid leukemia cells. Leukemia. 2009, 23, 1019-28.
dcterms.referencesYoo, C.B., Jones, P.A. Epigenetic therapy of cancer: past, present and future. Nat Rev Drug Discov. 2006, 5, 37-50.
dcterms.referencesGnyszka, A., JASTRZĘBSKI, Z., Flis, S. DNA methyltransferase inhibitors and their emerging role in epigenetic therapy of cancer. Anticancer Res. 2013, 33, 2989-96.
dcterms.referencesStresemann, C., Brueckner, B., Musch, T., Stopper, H., Lyko, F. Functional diversity of DNA methyltransferase inhibitors in human cancer cell lines. Cancer Res. 2006, 66, 2794- 800.
dcterms.referencesSvedruzic, Z.e.M., Reich, N.O. DNA cytosine C5 methyltransferase Dnmt1: catalysisdependent release of allosteric inhibition. Biochemistry. 2005, 44, 9472-85.
dcterms.referencesMedina-Franco, J.L., Méndez-Lucio, O., Yoo, J. Rationalization of activity cliffs of a sulfonamide inhibitor of DNA methyltransferases with induced-fit docking. Int J Mol Sci. 2014, 15, 3253-61.
dcterms.referencesMedina-Franco, J.L., Yoo, J. Docking of a novel DNA methyltransferase inhibitor identified from high-throughput screening: insights to unveil inhibitors in chemical databases. Molec Divers. 2013, 17, 337-44
dcterms.referencesYang, X., Lay, F., Han, H., Jones, P.A. Targeting DNA methylation for epigenetic therapy. Trends Pharmacol Sci. 2010, 31, 536-46.
dcterms.referencesIssa, J.-P.J., Garcia-Manero, G., Giles, F.J., Mannari, R., Thomas, D., Faderl, S., et al. Phase 1 study of low-dose prolonged exposure schedules of the hypomethylating agent 5- aza-2′-deoxycytidine (decitabine) in hematopoietic malignancies. Blood. 2004, 103, 1635-40.
dcterms.referencesSilverman, L., Holland, J., Weinberg, R., Alter, B., Davis, R., Ellison, R., et al. Effects of treatment with 5-azacytidine on the in vivo and in vitro hematopoiesis in patients with myelodysplastic syndromes. Leukemia. 1993, 7, 21-9
dcterms.referencesAsgatay, S., Champion, C., Marloie, G., Drujon, T., Senamaud-Beaufort, C., Ceccaldi, A., et al. Synthesis and evaluation of analogues of N-phthaloyl-l-tryptophan (RG108) as inhibitors of DNA methyltransferase 1. J Med Chem. 2014, 57, 421-34.
dcterms.referencesMedina-Franco, J.L., López-Vallejo, F., Kuck, D., Lyko, F. Natural products as DNA methyltransferase inhibitors: a computer-aided discovery approach. Molec Divers. 2011, 15, 293-304.
dcterms.referencesBrueckner, B., Boy, R.G., Siedlecki, P., Musch, T., Kliem, H.C., Zielenkiewicz, P., et al. Epigenetic reactivation of tumor suppressor genes by a novel small-molecule inhibitor of human DNA methyltransferases. Cancer Res. 2005, 65, 6305-11.
dcterms.referencesMaldonado-Rojas, W., Olivero-Verbel, J. Potential interaction of natural dietary bioactive compounds with COX-2. J Mol Graph Model. 2011, 30, 157-66.
dcterms.referencesWolber, G., Langer, T. LigandScout: 3-D pharmacophores derived from proteinbound ligands and their use as virtual screening filters. J Chem Inf Model. 2005, 45, 160-9.
dcterms.referencesNarváez-Pita, X., Ortega-Zuniga, C., Acevedo-Morantes, C.Y., Pastrana, B., OliveroVerbel, J., Maldonado-Rojas, W., et al. Water soluble molybdenocene complexes: Synthesis, cytotoxic activity and binding studies to ubiquitin by fluorescence spectroscopy, circular dichroism and molecular modeling. J Inorg Biochem. 2014, 132, 77-91.
dcterms.referencesVistica, D.T., Skehan, P., Scudiero, D., Monks, A., Pittman, A., Boyd, M.R. Tetrazolium-based assays for cellular viability: a critical examination of selected parameters affecting formazan production. Cancer Res. 1991, 51, 2515-20.
dcterms.referencesDomínguez-García, M., Ortega-Zúñiga, C., Meléndez, E. New tungstenocenes containing 3-hydroxy-4-pyrone ligands: antiproliferative activity on HT-29 and MCF-7 cell lines and binding to human serum albumin studied by fluorescence spectroscopy and molecular modeling methods. J Biol Inorg Chem. 2013, 18, 195-209.
dcterms.referencesChen, G.C., Yang, J.T. Two-point calibration of circular dichrometer with d-10- camphorsulfonic acid. Anal Lett. 1977, 10, 1195-207.
dcterms.referencesChikan, N.A., Bhavaniprasad, V., Anbarasu, K., Shabir, N., Patel, T.N. From natural products to drugs for epimutation computer-aided drug design. Appl Biochem Biotech. 2013, 170, 164-75.
dcterms.referencesQureshi, W., Alirhayim, Z., Ahmed, A., Al-Mallah, M., AbdulAziz, K. Digoxin Use in Atrial Fibrillation Is Associated with Increased Risk of Mortality: A Systematic Review and Meta-Analysis of More than 200,000 Patients. J Am Coll Cardiol. 2015, 65.
dcterms.referencesKapoor, S. Digoxin and its Antineoplastic Properties: An Evolving Role in Oncology. J Pediatr hematol Oncol. 2014, 36, 666-7.
dcterms.referencesShi, X., Chen, X., Li, X., Lan, X., Zhao, C., Liu, S., et al. Gambogic acid induces apoptosis in imatinib-resistant chronic myeloid leukemia cells via inducing proteasome inhibition and caspase-dependent Bcr-Abl downregulation. Clin Cancer Res. 2014, 20, 151- 63.
dcterms.referencesPlatz, E.A., Yegnasubramanian, S., Liu, J.O., Chong, C.R., Shim, J.S., Kenfield, S.A., et al. A novel two-stage, transdisciplinary study identifies digoxin as a possible drug for prostate cancer treatment. Cancer Discov. 2011, 1, 68-77.
dcterms.referencesYeh, J.-Y., Huang, W.J., Kan, S.-F., Wang, P.S. Inhibitory effects of digitalis on the proliferation of androgen dependent and independent prostate cancer cells. J Urol. 2001, 166, 1937-42.
dcterms.referencesLiu, W., Guo, Q.-L., You, Q.-D., Zhao, L., Gu, H.-Y., Yuan, S.-T. Anticancer effect and apoptosis induction of gambogic acid in human gastric cancer line BGC-823. World J Gastroenterol. 2005, 11, 3655-9.
dcterms.referencesQiang, L., Yang, Y., You, Q.-D., Ma, Y.-J., Yang, L., Nie, F.-F., et al. Inhibition of glioblastoma growth and angiogenesis by gambogic acid: an in vitro and in vivo study. Biochem Pharmacol. 2008, 75, 1083-92.
dcterms.referencesGirault, I., Tozlu, S., Lidereau, R., Bièche, I. Expression analysis of DNA methyltransferases 1, 3A, and 3B in sporadic breast carcinomas. Clin Cancer Res. 2003, 9, 4415-22.
dcterms.referencesJin, B., Yao, B., Li, J.-L., Fields, C.R., Delmas, A.L., Liu, C., et al. DNMT1 and DNMT3B modulate distinct polycomb-mediated histone modifications in colon cancer. Cancer Res. 2009, 69, 7412-21.
dcterms.referencesSułkowska, A. Interaction of drugs with bovine and human serum albumin. J Mol Struct. 2002, 614, 227-32.
dcterms.referencesTayeh, N., Rungassamy, T., Albani, J.R. Fluorescence spectral resolution of tryptophan residues in bovine and human serum albumins. J Pharm Biomed Anal. 2009, 50, 107-16.
dcterms.referencesGao, Y., Shao, C., Ji, W., Xiao, M., Yi, F., Zhou, T., et al. Studies on the Binding Mechanism of VB 1 and VB 9 with Trypsin. Am J Anal Chem. 2013, 2013.
dcterms.referencesRoss, P.D., Subramanian, S. Thermodynamics of protein association reactions: forces contributing to stability. Biochemistry. 1981, 20, 3096-102
dcterms.referencesZsila, F. Circular dichroism spectroscopic detection of ligand binding induced subdomain IB specific structural adjustment of human serum albumin. J Phys Chem B. 2013, 117, 10798-806.
dcterms.referencesVarshney, A., Ansari, Y., Zaidi, N., Ahmad, E., Badr, G., Alam, P., et al. Analysis of binding interaction between antibacterial ciprofloxacin and human serum albumin by spectroscopic techniques. Cell Biochem Biophys. 2014, 70, 93-101.
dc.rights.accessopenAccess


Ficheros en el ítem

Thumbnail

Este ítem aparece en la(s) siguiente(s) colección(ones)

Mostrar el registro sencillo del ítem

https://creativecommons.org/licenses/by-nc-nd/4.0
Excepto si se señala otra cosa, la licencia del ítem se describe como https://creativecommons.org/licenses/by-nc-nd/4.0