Polyphenols as Potent Epigenetics Agents for Cancer
Abstract
:1. Introduction
2. Oxidative DNA Damage and Polyphenols
3. Human Cancer and DNA Methylation
4. Cancer and Histones
5. Inhibitors of DNA Methylation
6. Inhibitors of Histone Modification
7. Polyphenol and Cancer
7.1. Epigenetics Mechanism of Kaempferol
7.2. Epigenetic Mechanism of Gallic Acid
7.3. Epigenetic Modulation of Curcumin in Cancer
7.4. Epigenetic Modulation of Resveratrol in Cancer
7.5. Epigenetic Modulation of EGCG in Cancer
7.6. Epigenetic Modulation of Quercetin in Cancer
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Vandegehuchte, M.B.; Janssen, C.R. Epigenetics and its implications for ecotoxicology. Ecotoxicology 2011, 20, 607–624. [Google Scholar] [CrossRef]
- Foolchand, A. Methyl Picolinic Acid Plays a Role in Epigenetic Modifications in Human HepG2 Liver Cells. Ph.D. Dissertation, University of Kwazulu-Natal, Berea, South Africa, 2019. [Google Scholar]
- Jablonka, E.; Lamb, M.J. The expanded evolutionary synthesis—A response to Godfrey-Smith, Haig, and West-Eberhard. Biol. Philos. 2007, 22, 453–472. [Google Scholar] [CrossRef]
- Deans, C.; Maggert, K.A. What do you mean, “epigenetic”? Genetics 2015, 199, 887–896. [Google Scholar] [CrossRef] [Green Version]
- Maccani, M.A.; Marsit, C.J. Epigenetics in the placenta. Am. J. Reprod. Immunol. 2009, 62, 78–89. [Google Scholar] [CrossRef] [Green Version]
- Wiesel-Motiuk, N.; Assaraf, Y.G. The key roles of the lysine acetyltransferases KAT6A and KAT6B in physiology and pathology. Drug Resist. Updat. 2020, 53, 100729. [Google Scholar] [CrossRef]
- Tost, J. DNA methylation: An introduction to the biology and the disease-associated changes of a promising biomarker. Mol. Biotechnol. 2010, 44, 71–81. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.; Kaang, B.-K. Epigenetic regulation and chromatin remodeling in learning and memory. Exp. Mol. Med. 2017, 49, e281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pastar, I.; Marjanovic, J.; Stone, R.C.; Chen, V.; Burgess, J.L.; Mervis, J.S.; Tomic-Canic, M. Epigenetic regulation of cellular functions in wound healing. Exp. Dermatol. 2021, 30, 1073–1089. [Google Scholar] [CrossRef]
- Kumar, H.; Chaudhary, A.; Singh, A.; Sukhija, N.; Panwar, A.; Saravanan, K.; Bhaladhare, A.; Kaisa, K.; Panigrahi, M. A review on epigenetics: Manifestations, modifications, methods & challenges. J. Entomol. Zool. Stud. 2020, 8, 1–6. [Google Scholar]
- Andersen, G.B.; Tost, J. A summary of the biological processes, disease-associated changes, and clinical applications of DNA methylation. DNA Methylation Protoc. 2018, 1708, 3–30. [Google Scholar]
- Castilho, R.M.; Squarize, C.H.; Almeida, L.O. Epigenetic modifications and head and neck cancer: Implications for tumor progression and resistance to therapy. Int. J. Mol. Sci. 2017, 18, 1506. [Google Scholar] [CrossRef] [Green Version]
- Biswas, S.; Rao, C.M. Epigenetics in cancer: Fundamentals and beyond. Pharmacol. Ther. 2017, 173, 118–134. [Google Scholar] [CrossRef] [PubMed]
- Johnson, C.; Warmoes, M.O.; Shen, X.; Locasale, J.W. Epigenetics and cancer metabolism. Cancer Lett. 2015, 356, 309–314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kinnaird, A.; Zhao, S.; Wellen, K.E.; Michelakis, E.D. Metabolic control of epigenetics in cancer. Nat. Rev. Cancer 2016, 16, 694–707. [Google Scholar] [CrossRef] [PubMed]
- Shankar, E.; Kanwal, R.; Candamo, M.; Gupta, S. Dietary phytochemicals as epigenetic modifiers in cancer: Promise and challenges. In Proceedings of the Seminars in Cancer Biology; Elsevier: Amsterdam, The Netherlands, 2016; pp. 82–99. [Google Scholar]
- Sapienza, C.; Issa, J.-P. Diet, nutrition, and cancer epigenetics. Annu. Rev. Nutr. 2016, 36, 665–681. [Google Scholar] [CrossRef]
- Carlos-Reyes, Á.; López-González, J.S.; Meneses-Flores, M.; Gallardo-Rincón, D.; Ruíz-García, E.; Marchat, L.A.; Astudillo-De La Vega, H.; Hernández de la Cruz, O.N.; López-Camarillo, C. Dietary compounds as epigenetic modulating agents in cancer. Front. Genet. 2019, 10, 79. [Google Scholar] [CrossRef] [Green Version]
- Lévesque, S.; Pol, J.G.; Ferrere, G.; Galluzzi, L.; Zitvogel, L.; Kroemer, G. Trial watch: Dietary interventions for cancer therapy. Oncoimmunology 2019, 8, e1591878. [Google Scholar] [CrossRef]
- Salisbury, D.; Bronas, U. Reactive Oxygen and Nitrogen Species: Impact on Endothelial Dysfunction. Nurs. Res. 2015, 64, 53–66. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Tsao, R. Dietary Polyphenols, Oxidative Stress and Antioxidant and Anti-inflammatory Effects. Curr. Opin. Food Sci. 2016, 8, 33–42. [Google Scholar] [CrossRef]
- Reis, J.F.; Monteiro, V.V.S.; De Souza Gomes, R.; Do Carmo, M.M.; Da Costa, G.V.; Ribera, P.C.; Monteiro, M.C. Action Mechanism and Cardiovascular Effect of Anthocyanins: A Systematic Review of Animal and Human Studies. J. Transl. Med. 2016, 14, 315. [Google Scholar] [CrossRef] [Green Version]
- Rudrapal, M.; Khairnar, S.J.; Khan, J.; Dukhyil, A.B.; Ansari, M.A.; Alomary, M.N.; Alshabrmi, F.M.; Palai, S.; Deb, P.K.; Devi, R. Dietary Polyphenols and Their Role in Oxidative Stress-Induced Human Diseases: Insights into Protective Effects, Antioxidant Potentials and Mechanism (s) of Action. Front. Pharmacol. 2022, 13, 806470. [Google Scholar] [CrossRef] [PubMed]
- Federico, A.; Morgillo, F.; Tuccillo, C.; Ciardiello, F.; Loguercio, C. Chronic inflammation and oxidative stress in human carcinogenesis. Int. J. Cancer 2007, 121, 2381–2386. [Google Scholar] [CrossRef]
- Khansari, N.; Shakiba, Y.; Mahmoudi, M. Chronic inflammation and oxidative stress as a major cause of age-related diseases and cancer. Recent Pat. Inflamm. Allergy Drug Discov. 2009, 3, 73–80. [Google Scholar] [CrossRef]
- Azqueta, A.; Collins, A. Polyphenols and DNA Damage: A Mixed Blessing. Nutrients 2016, 8, 785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Papiez, M.A. The influence of curcumin and (−)-epicatechin on the genotoxicity and myelosuppression induced by etoposide in bone marrow cells of male rats. Drug Chem. Toxicol. 2013, 36, 93–101. [Google Scholar] [CrossRef]
- García-Lafuente, A.; Guillamón, E.; Villares, A.; Rostagno, M.A.; Martínez, J.A. Flavonoids as anti-inflammatory agents: Implications in cancer and cardiovascular disease. Inflamm. Res. 2009, 58, 537–552. [Google Scholar] [CrossRef] [PubMed]
- Hazel, T.G.; Nathans, D.; Lau, L.F. A gene inducible by serum growth factors encodes a member of the steroid and thyroid hormone receptor superfamily. Proc. Natl. Acad. Sci. USA 1988, 85, 8444–8448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kamaraj, S.; Vinodhkumar, R.; Anandakumar, P.; Jagan, S.; Ramakrishnan, G.; Devaki, T. The effects of quercetin on antioxidant status and tumor markers in the lung and serum of mice treated with benzo (a) pyrene. Biol. Pharm. Bull. 2007, 30, 2268–2273. [Google Scholar] [CrossRef] [Green Version]
- Shankar, S.; Ganapathy, S.; Hingorani, S.R.; Srivastava, R.K. EGCG inhibits growth, invasion, angiogenesis and metastasis of pancreatic cancer. Front.Biosci. J. Virtual Libr. 2008, 13, 440–452. [Google Scholar] [CrossRef] [Green Version]
- Sharma, V.; Rao, L.J.M. A thought on the biological activities of black tea. Crit. Rev. Food Sci. Nutr. 2009, 49, 379–404. [Google Scholar] [CrossRef]
- Kim, S.Y.; Park, C.; Jang, H.-J.; Kim, B.-O.; Bae, H.-W.; Chung, I.-Y.; Kim, E.S.; Cho, Y.-H. Antibacterial strategies inspired by the oxidative stress and response networks. J. Microbiol. 2019, 57, 203–212. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Moustaid-Moussa, N.; Chen, L.; Mo, H.; Shastri, A.; Su, R.; Bapat, P.; Kwun, I.; Shen, C.-L. Novel insights of dietary polyphenols and obesity. J. Nutr. Biochem. 2014, 25, 1–18. [Google Scholar] [CrossRef] [PubMed]
- Cory, H.; Passarelli, S.; Szeto, J.; Tamez, M.; Mattei, J. The role of polyphenols in human health and food systems: A mini-review. Front. Nutr. 2018, 5, 87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Holásková, I.; Elliott, M.; Hanson, M.L.; Schafer, R.; Barnett, J.B. Prenatal cadmium exposure produces persistent changes to thymus and spleen cell phenotypic repertoire as well as the acquired immune response. Toxicol. Appl. Pharmacol. 2012, 265, 181–189. [Google Scholar] [CrossRef] [Green Version]
- Lewinska, A.; Wnuk, M.; Grabowska, W.; Zabek, T.; Semik, E.; Sikora, E.; Bielak-Zmijewska, A. Curcumin induces oxidation-dependent cell cycle arrest mediated by sirt7 inhibition of rdna transcription in human aortic smooth muscle cells. Toxicol. Lett. 2015, 233, 227–238. [Google Scholar] [CrossRef]
- Majidinia, M.; Bishayee, A.; Yousefi, B. Polyphenols: Major regulators of key components of DNA damage response in cancer. DNA Repair 2019, 82, 102679. [Google Scholar] [CrossRef]
- Rajendran, P.; Alzahrani, A.M.; Rengarajan, T.; Kaushik, R.; Arulselvan, P.; Umamaheswari, A. Polyphenols and Cancer. In Frontiers in Anti-Cancer Drug Discovery; Bentham Science Publishers: Singapore, 2019; Volume 10, p. 62. [Google Scholar]
- Locke, W.J.; Guanzon, D.; Ma, C.; Liew, Y.J.; Duesing, K.R.; Fung, K.Y.; Ross, J.P. DNA methylation cancer biomarkers: Translation to the clinic. Front. Genet. 2019, 10, 1150. [Google Scholar] [CrossRef]
- Koch, A.; Joosten, S.C.; Feng, Z.; de Ruijter, T.C.; Draht, M.X.; Melotte, V.; Smits, K.M.; Veeck, J.; Herman, J.G.; Van Neste, L. Analysis of DNA methylation in cancer: Location revisited. Nat. Rev. Clin. Oncol. 2018, 15, 459–466. [Google Scholar] [CrossRef]
- Paska, A.V.; Hudler, P. Aberrant methylation patterns in cancer: A clinical view. Biochem. Med. 2015, 25, 161–176. [Google Scholar] [CrossRef]
- Udali, S.; Guarini, P.; Ruzzenente, A.; Ferrarini, A.; Guglielmi, A.; Lotto, V.; Tononi, P.; Pattini, P.; Moruzzi, S.; Campagnaro, T. DNA methylation and gene expression profiles show novel regulatory pathways in hepatocellular carcinoma. Clin. Epigenetics 2015, 7, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Xu, R.-h.; Wei, W.; Krawczyk, M.; Wang, W.; Luo, H.; Flagg, K.; Yi, S.; Shi, W.; Quan, Q.; Li, K. Circulating tumour DNA methylation markers for diagnosis and prognosis of hepatocellular carcinoma. Nat. Mater. 2017, 16, 1155–1161. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Petropoulos, S.; Liu, J.; Cheishvili, D.; Zhou, R.; Dymov, S.; Li, K.; Li, N.; Szyf, M. The signature of liver cancer in immune cells DNA methylation. Clin. Epigenetics 2018, 10, 1–17. [Google Scholar] [CrossRef]
- Hao, X.; Luo, H.; Krawczyk, M.; Wei, W.; Wang, W.; Wang, J.; Flagg, K.; Hou, J.; Zhang, H.; Yi, S. DNA methylation markers for diagnosis and prognosis of common cancers. Proc. Natl. Acad. Sci. USA 2017, 114, 7414–7419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Villanueva, A.; Portela, A.; Sayols, S.; Battiston, C.; Hoshida, Y.; Méndez-González, J.; Imbeaud, S.; Letouzé, E.; Hernandez-Gea, V.; Cornella, H. DNA methylation-based prognosis and epidrivers in hepatocellular carcinoma. Hepatology 2015, 61, 1945–1956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, L.; Li, W. Epithelial–mesenchymal transition in human cancer: Comprehensive reprogramming of metabolism, epigenetics, and differentiation. Pharmacol. Ther. 2015, 150, 33–46. [Google Scholar] [CrossRef] [PubMed]
- Feinberg, A.P.; Koldobskiy, M.A.; Göndör, A. Epigenetic modulators, modifiers and mediators in cancer aetiology and progression. Nat. Rev. Genet. 2016, 17, 284–299. [Google Scholar] [CrossRef]
- Qian, Z.; Shen, Q.; Yang, X.; Qiu, Y.; Zhang, W. The role of extracellular vesicles: An epigenetic view of the cancer microenvironment. BioMed Res. Int. 2015, 2015, 649161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ilyas, M. Next-generation sequencing in diagnostic pathology. Pathobiology 2017, 84, 292–305. [Google Scholar] [CrossRef] [Green Version]
- Kurumizaka, H.; Kujirai, T.; Takizawa, Y. Contributions of histone variants in nucleosome structure and function. J. Mol. Biol. 2021, 433, 166678. [Google Scholar] [CrossRef]
- Bilokapic, S.; Strauss, M.; Halic, M. Histone octamer rearranges to adapt to DNA unwrapping. Nat. Struct. Mol. Biol. 2018, 25, 101–108. [Google Scholar] [CrossRef]
- Zou, T.; Hashiya, F.; Wei, Y.; Yu, Z.; Pandian, G.N.; Sugiyama, H. Direct observation of H3–H4 octasome by high-speed AFM. Chem. A Eur. J. 2018, 24, 15998–16002. [Google Scholar] [CrossRef] [PubMed]
- Ramazi, S.; Allahverdi, A.; Zahiri, J. Evaluation of post-translational modifications in histone proteins: A review on histone modification defects in developmental and neurological disorders. J. Biosci. 2020, 45, 1–29. [Google Scholar] [CrossRef]
- Millán-Zambrano, G.; Burton, A.; Bannister, A.J.; Schneider, R. Histone post-translational modifications—Cause and consequence of genome function. Nat. Rev. Genet. 2022, 23, 1–18. [Google Scholar] [CrossRef] [PubMed]
- van Wijnen, A.J.; Westendorf, J.J. Epigenetics as a new frontier in orthopedic regenerative medicine and oncology. J. Orthop. Res. 2019, 37, 1465–1474. [Google Scholar] [CrossRef] [PubMed]
- Borkiewicz, L. Histone 3 Lysine 27 Trimethylation Signature in Breast Cancer. Int. J. Mol. Sci. 2021, 22, 12853. [Google Scholar] [CrossRef]
- Shimizu, J.; Kawano, F. Exercise-induced histone H3 trimethylation at lysine 27 facilitates the adaptation of skeletal muscle to exercise in mice. J. Physiol. 2022, 600, 3331–3353. [Google Scholar] [CrossRef]
- Lu, K.; Tao, H.; Si, X.; Chen, Q. The histone H3 lysine 4 presenter WDR5 as an oncogenic protein and novel epigenetic target in cancer. Front. Oncol. 2018, 8, 502. [Google Scholar] [CrossRef]
- Tsai, C.-C.; Chien, M.-N.; Chang, Y.-C.; Lee, J.-J.; Dai, S.-H.; Cheng, S.-P. Overexpression of histone H3 lysine 27 trimethylation is associated with aggressiveness and dedifferentiation of thyroid cancer. Endocr. Pathol. 2019, 30, 305–311. [Google Scholar] [CrossRef]
- Blecua, P.; Martinez-Verbo, L.; Esteller, M. The DNA methylation landscape of hematological malignancies: An update. Mol. Oncol. 2020, 14, 1616–1639. [Google Scholar] [CrossRef]
- Zhou, M.; Yuan, M.; Zhang, M.; Lei, C.; Aras, O.; Zhang, X.; An, F. Combining histone deacetylase inhibitors (HDACis) with other therapies for cancer therapy. Eur. J. Med. Chem. 2021, 226, 113825. [Google Scholar] [CrossRef]
- Banik, D.; Moufarrij, S.; Villagra, A. Immunoepigenetics combination therapies: An overview of the role of HDACs in cancer immunotherapy. Int. J. Mol. Sci. 2019, 20, 2241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Roca, M.S.; Di Gennaro, E.; Budillon, A. Implication for cancer stem cells in solid cancer chemo-resistance: Promising therapeutic strategies based on the use of HDAC inhibitors. J. Clin. Med. 2019, 8, 912. [Google Scholar] [CrossRef] [PubMed]
- Movafagh, S.; Munson, A. Histone deacetylase inhibitors in cancer prevention and therapy. In Epigenetics of Cancer Prevention; Elsevier: Amsterdam, The Netherlands, 2019; pp. 75–105. [Google Scholar]
- Rojas-Espinosa, O.; Moreno-García, S.; Arce-Paredes, P.; Becerril-Villanueva, E.; Juárez-Ortega, M. Effect of dialyzable leukocyte extract, sodium butyrate, and valproic acid in the development of anergy in murine leprosy. Int. J. Mycobacteriology 2020, 9, 268. [Google Scholar] [CrossRef] [PubMed]
- Perona, M.; Thomasz, L.; Rossich, L.; Rodriguez, C.; Pisarev, M.A.; Rosemblit, C.; Cremaschi, G.A.; Dagrosa, M.A.; Juvenal, G.J. Radiosensitivity enhancement of human thyroid carcinoma cells by the inhibitors of histone deacetylase sodium butyrate and valproic acid. Mol. Cell. Endocrinol. 2018, 478, 141–150. [Google Scholar] [CrossRef]
- Sanaei, M.; Kavoosi, F. Effect of Valproic Acid on the Class I Histone Deacetylase 1, 2 and 3, Tumor Suppressor Genes p21WAF1/CIP1 and p53, and Intrinsic Mitochondrial Apoptotic Pathway, Pro-(Bax, Bak, and Bim) and anti-(Bcl-2, Bcl-xL, and Mcl-1) Apoptotic Genes Expression, Cell Viability, and Apoptosis Induction in Hepatocellular Carcinoma HepG2 Cell Line. Asian Pac. J. Cancer Prev. 2021, 22, 89–95. [Google Scholar]
- Elnozahi, N.A.; Abd ELAziz, E.A.; Helmy, M.W.; Bistawroos, A.E. Modulatory Effect of Sodium Butyrate on Anticancer Activity of Abemaciclib in MDA-MB-231 Human Breast Cancer Cells; Research Square: Durham, NC, USA, 2022. [Google Scholar] [CrossRef]
- Sanaei, M.; Kavoosi, F.; Moezzi, M.A. Effect of 5′-fluoro-2′-deoxycytidine and sodium butyrate on the genes of the intrinsic apoptotic pathway, p21, p53, cell viability, and apoptosis in human hepatocellular carcinoma cell lines. Iran. J. Pediatr. Hematol. Oncol. 2021, 11, 216–230. [Google Scholar] [CrossRef]
- Psilopatis, I.; Pergaris, A.; Giaginis, C.; Theocharis, S. Histone Deacetylase Inhibitors: A Promising Therapeutic Alternative for Endometrial Carcinoma. Dis. Markers 2021, 2021, 1–9. [Google Scholar] [CrossRef]
- Garmpis, N.; Damaskos, C.; Garmpi, A.; Georgakopoulou, V.E.; Sarantis, P.; Antoniou, E.A.; Karamouzis, M.V.; Nonni, A.; Schizas, D.; Diamantis, E. Histone deacetylase inhibitors in the treatment of hepatocellular carcinoma: Current evidence and future opportunities. J. Pers. Med. 2021, 11, 223. [Google Scholar] [CrossRef]
- Tsilimigras, D.I.; Ntanasis-Stathopoulos, I.; Moris, D.; Spartalis, E.; Pawlik, T.M. Histone deacetylase inhibitors in hepatocellular carcinoma: A therapeutic perspective. Surg. Oncol. 2018, 27, 611–618. [Google Scholar] [CrossRef]
- Zhang, H.; Zhao, X.; Liu, H.; Jin, H.; Ji, Y. Trichostatin A inhibits proliferation of PC3 prostate cancer cells by disrupting the EGFR pathway. Oncol. Lett. 2019, 18, 687–693. [Google Scholar] [CrossRef] [Green Version]
- Jang, Y.G.; Hwang, K.A.; Choi, K.C. Rosmarinic Acid, a Component of Rosemary Tea, Induced the Cell Cycle Arrest and Apoptosis through Modulation of HDAC2 Expression in Prostate Cancer Cell Lines. Nutrients 2018, 10, 1784. [Google Scholar] [CrossRef] [Green Version]
- Hontecillas-Prieto, L.; Flores-Campos, R.; Silver, A.; de Álava, E.; Hajji, N.; García-Domínguez, D.J. Synergistic Enhancement of Cancer Therapy Using HDAC Inhibitors: Opportunity for Clinical Trials. Front. Genet. 2020, 11, 578011. [Google Scholar] [CrossRef]
- Vaidya, G.N.; Rana, P.; Venkatesh, A.; Chatterjee, D.R.; Contractor, D.; Satpute, D.P.; Nagpure, M.; Jain, A.; Kumar, D. Paradigm shift of “classical” HDAC inhibitors to “hybrid” HDAC inhibitors in therapeutic interventions. Eur. J. Med. Chem. 2021, 209, 112844. [Google Scholar] [CrossRef]
- Hazafa, A.; Rehman, K.U.; Jahan, N.; Jabeen, Z. The Role of Polyphenol (Flavonoids) Compounds in the Treatment of Cancer Cells. Nutr. Cancer 2020, 72, 386–397. [Google Scholar] [CrossRef]
- Davatgaran-Taghipour, Y.; Masoomzadeh, S.; Farzaei, M.H.; Bahramsoltani, R.; Karimi-Soureh, Z.; Rahimi, R.; Abdollahi, M. Polyphenol nanoformulations for cancer therapy: Experimental evidence and clinical perspective. Int. J. Nanomed. 2017, 12, 2689–2702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Williamson, G. The role of polyphenols in modern nutrition. Nutr. Bull. 2017, 42, 226–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tsao, R. Chemistry and biochemistry of dietary polyphenols. Nutrients 2010, 2, 1231–1246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rana, A.; Samtiya, M.; Dhewa, T.; Mishra, V.; Aluko, R.E. Health benefits of polyphenols: A concise review. J. Food Biochem. 2022, 13, e14264. [Google Scholar] [CrossRef] [PubMed]
- Del Rio, D.; Costa, L.G.; Lean, M.E.; Crozier, A. Polyphenols and health: What compounds are involved? Nutr. Metab. Cardiovasc. Dis. 2010, 20, 1–6. [Google Scholar] [CrossRef] [PubMed]
- Rajendran, P.; Rengarajan, T.; Nandakumar, N.; Divya, H.; Nishigaki, I. Mangiferin in cancer chemoprevention and treatment: Pharmacokinetics and molecular targets. J. Recept. Signal Transduct. Res. 2015, 35, 76–84. [Google Scholar] [CrossRef]
- Rajendran, P.; Rengarajan, T.; Nishigaki, I.; Ekambaram, G.; Sakthisekaran, D. Potent chemopreventive effect of mangiferin on lung carcinogenesis in experimental Swiss albino mice. J. Cancer Res. Ther. 2014, 10, 1033–1039. [Google Scholar] [CrossRef] [PubMed]
- Drețcanu, G.; Iuhas, C.I.; Diaconeasa, Z. The Involvement of Natural Polyphenols in the Chemoprevention of Cervical Cancer. Int. J. Mol. Sci. 2021, 22, 8812. [Google Scholar] [CrossRef] [PubMed]
- Shah, D.; Gandhi, M.; Kumar, A.; Cruz-Martins, N.; Sharma, R.; Nair, S. Current insights into epigenetics, noncoding RNA interactome and clinical pharmacokinetics of dietary polyphenols in cancer chemoprevention. Crit. Rev. Food Sci. Nutr. 2021, 1–37. [Google Scholar] [CrossRef] [PubMed]
- Rajendran, P.; Rengarajan, T.; Nandakumar, N.; Palaniswami, R.; Nishigaki, Y.; Nishigaki, I. Kaempferol, a potential cytostatic and cure for inflammatory disorders. Eur. J. Med. Chem. 2014, 86, 103–112. [Google Scholar] [CrossRef] [PubMed]
- Amjad, E.; Sokouti, B.; Asnaashari, S. A systematic review of anti-cancer roles and mechanisms of kaempferol as a natural compound. Cancer Cell Int. 2022, 22, 260. [Google Scholar] [CrossRef] [PubMed]
- Mohan Shankar, G.; Swetha, M.; Keerthana, C.K.; Rayginia, T.P.; Anto, R.J. Cancer Chemoprevention: A Strategic Approach Using Phytochemicals. Front. Pharmacol. 2021, 12, 809308. [Google Scholar] [CrossRef]
- Sharma, N.; Biswas, S.; Al-Dayan, N.; Alhegaili, A.S.; Sarwat, M. Antioxidant Role of Kaempferol in Prevention of Hepatocellular Carcinoma. Antioxidants 2021, 10, 1419. [Google Scholar] [CrossRef]
- Qiu, W.; Lin, J.; Zhu, Y.; Zhang, J.; Zeng, L.; Su, M.; Tian, Y. Kaempferol Modulates DNA Methylation and Downregulates DNMT3B in Bladder Cancer. Cell. Physiol. Biochem. 2017, 41, 1325–1335. [Google Scholar] [CrossRef]
- Lu, L.; Wang, Y.; Ou, R.; Feng, Q.; Ji, L.; Zheng, H.; Guo, Y.; Qi, X.; Kong, A.N.; Liu, Z. DACT2 Epigenetic Stimulator Exerts Dual Efficacy for Colorectal Cancer Prevention and Treatment. Pharmacol. Res. 2018, 129, 318–328. [Google Scholar] [CrossRef]
- Kim, T.H.; Ku, S.-K.; Lee, I.-C.; Bae, J.-S. Anti-inflammatory effects of kaempferol-3-O-sophoroside in human endothelial cells. Inflamm. Res. 2012, 3, 217–224. [Google Scholar] [CrossRef]
- Kim, T.W.; Lee, S.Y.; Kim, M.; Cheon, C.; Ko, S.G. Kaempferol induces autophagic cell death via IRE1-JNK-CHOP pathway and inhibition of G9a in gastric cancer cells. Cell Death Dis. 2018, 9, 875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Imran, M.; Salehi, B.; Sharifi-Rad, J.; Aslam Gondal, T.; Saeed, F.; Imran, A.; Shahbaz, M.; Tsouh Fokou, P.V.; Umair Arshad, M.; Khan, H.; et al. Kaempferol: A Key Emphasis to Its Anticancer Potential. Molecules 2019, 24, 2277. [Google Scholar] [CrossRef] [PubMed]
- Bhosale, P.B.; Ha, S.E.; Vetrivel, P.; Kim, H.H.; Kim, S.M.; Kim, G.S. Functions of polyphenols and its anticancer properties in biomedical research: A narrative review. Transl. Cancer Res. 2020, 9, 7619–7631. [Google Scholar] [CrossRef] [PubMed]
- Jiang, Y.; Pei, J.; Zheng, Y.; Miao, Y.J.; Duan, B.Z.; Huang, L.F. Gallic Acid: A Potential Anti-Cancer Agent. Chin. J. Integr. Med. 2022, 28, 661–671. [Google Scholar] [CrossRef]
- Shi, C.J.; Zheng, Y.B.; Pan, F.F.; Zhang, F.W.; Zhuang, P.; Fu, W.M. Gallic Acid Suppressed Tumorigenesis by an LncRNA MALAT1-Wnt/β-Catenin Axis in Hepatocellular Carcinoma. Front. Pharmacol. 2021, 12, 708967. [Google Scholar] [CrossRef]
- Ko, E.B.; Jang, Y.G.; Kim, C.W.; Go, R.E.; Lee, H.K.; Choi, K.C. Gallic Acid Hindered Lung Cancer Progression by Inducing Cell Cycle Arrest and Apoptosis in A549 Lung Cancer Cells via PI3K/Akt Pathway. Biomol. Ther. 2022, 30, 151–161. [Google Scholar] [CrossRef]
- Aglan, H.A.; Ahmed, H.H.; El-Toumy, S.A.; Mahmoud, N.S. Gallic acid against hepatocellular carcinoma: An integrated scheme of the potential mechanisms of action from in vivo study. Tumour Biol. 2017, 39, 1010428317699127. [Google Scholar] [CrossRef] [Green Version]
- Lima, K.G.; Krause, G.C.; Schuster, A.D.; Catarina, A.V.; Basso, B.S.; De Mesquita, F.C.; Pedrazza, L.; Marczak, E.S.; Martha, B.A.; Nunes, F.B.; et al. Gallic acid reduces cell growth by induction of apoptosis and reduction of IL-8 in HepG2 cells. Biomed. Pharmacother. 2016, 84, 1282–1290. [Google Scholar] [CrossRef]
- Dorniani, D.; Saifullah, B.; Barahuie, F.; Arulselvan, P.; Hussein, M.Z.; Fakurazi, S.; Twyman, L.J. Graphene Oxide-Gallic Acid Nanodelivery System for Cancer Therapy. Nanoscale Res. Lett. 2016, 11, 491. [Google Scholar] [CrossRef] [Green Version]
- Silva, I.C.; Polaquini, C.R.; Regasini, L.O.; Ferreira, H.; Pavan, F.R. Evaluation of cytotoxic, apoptotic, mutagenic, and chemopreventive activities of semi-synthetic esters of gallic acid. Food Chem. Toxicol. 2017, 105, 300–307. [Google Scholar] [CrossRef] [Green Version]
- Weng, Y.P.; Hung, P.F.; Ku, W.Y.; Chang, C.Y.; Wu, B.H.; Wu, M.H.; Yao, J.Y.; Yang, J.R.; Lee, C.H. The inhibitory activity of gallic acid against DNA methylation: Application of gallic acid on epigenetic therapy of human cancers. Oncotarget 2018, 9, 361–374. [Google Scholar] [CrossRef] [Green Version]
- Bhattacharya, R.; Chatterjee, R.; Mandal, A.K.A.; Mukhopadhyay, A.; Basu, S.; Giri, A.K.; Chatterji, U.; Bhattacharjee, P. Theaflavin-Containing Black Tea Extract: A Potential DNA Methyltransferase Inhibitor in Human Colon Cancer Cells and Ehrlich Ascites Carcinoma-Induced Solid Tumors in Mice. Nutr. Cancer 2021, 73, 2447–2459. [Google Scholar] [CrossRef] [PubMed]
- Jang, Y.G.; Ko, E.B.; Choi, K.C. Gallic acid, a phenolic acid, hinders the progression of prostate cancer by inhibition of histone deacetylase 1 and 2 expression. J. Nutr. Biochem. 2020, 84, 108444. [Google Scholar] [CrossRef] [PubMed]
- Lee, W.; Lee, S.Y.; Son, Y.J.; Yun, J.M. Gallic Acid Decreases Inflammatory Cytokine Secretion Through Histone Acetyltransferase/Histone Deacetylase Regulation in High Glucose-Induced Human Monocytes. J. Med. Food 2015, 18, 793–801. [Google Scholar] [CrossRef]
- Soflaei, S.S.; Momtazi-Borojeni, A.A.; Majeed, M.; Derosa, G.; Maffioli, P.; Sahebkar, A. Curcumin: A Natural Pan-HDAC Inhibitor in Cancer. Curr. Pharm. Des. 2018, 24, 123–129. [Google Scholar] [CrossRef]
- Lee, S.J.; Krauthauser, C.; Maduskuie, V.; Fawcett, P.T.; Olson, J.M.; Rajasekaran, S.A. Curcumin-induced HDAC inhibition and attenuation of medulloblastoma growth in vitro and in vivo. BMC Cancer 2011, 11, 144. [Google Scholar] [CrossRef] [Green Version]
- Yu, J.; Peng, Y.; Wu, L.C.; Xie, Z.; Deng, Y.; Hughes, T.; He, S.; Mo, X.; Chiu, M.; Wang, Q.E.; et al. Curcumin down-regulates DNA methyltransferase 1 and plays an anti-leukemic role in acute myeloid leukemia. PLoS ONE 2013, 8, e55934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Teiten, M.H.; Dicato, M.; Diederich, M. Curcumin as a regulator of epigenetic events. Mol. Nutr. Food Res. 2013, 57, 1619–1629. [Google Scholar] [CrossRef]
- Soltani, B.; Ghaemi, N.; Sadeghizadeh, M.; Najafi, F. Curcumin confers protection to irradiated THP-1 cells while its nanoformulation sensitizes these cells via apoptosis induction. Cell Biol. Toxicol. 2016, 32, 543–561. [Google Scholar] [CrossRef]
- Kang, S.K.; Cha, S.H.; Jeon, H.G. Curcumin-induced histone hypoacetylation enhances caspase-3-dependent glioma cell death and neurogenesis of neural progenitor cells. Stem Cells Dev. 2006, 15, 165–174. [Google Scholar] [CrossRef]
- Yuan, Z.; Syed, M.A.; Panchal, D.; Rogers, D.; Joo, M.; Sadikot, R.T. Curcumin mediated epigenetic modulation inhibits TREM-1 expression in response to lipopolysaccharide. Int. J. Biochem. Cell Biol. 2012, 44, 2032–2043. [Google Scholar] [CrossRef] [PubMed]
- Parashar, G.; Parashar, N.C.; Capalash, N. Curcumin causes promoter hypomethylation and increased expression of FANCF gene in SiHa cell line. Mol. Cell. Biochem. 2012, 365, 29–35. [Google Scholar] [CrossRef] [PubMed]
- Shu, L.; Khor, T.O.; Lee, J.H.; Boyanapalli, S.S.; Huang, Y.; Wu, T.Y.; Saw, C.L.; Cheung, K.L.; Kong, A.N. Epigenetic CpG demethylation of the promoter and reactivation of the expression of Neurog1 by curcumin in prostate LNCaP cells. AAPS J. 2011, 13, 606–614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, C.; Su, Z.Y.; Khor, T.O.; Shu, L.; Kong, A.N. Sulforaphane enhances Nrf2 expression in prostate cancer TRAMP C1 cells through epigenetic regulation. Biochem. Pharmacol. 2013, 85, 1398–1404. [Google Scholar] [CrossRef] [Green Version]
- Kutluay, S.B.; Doroghazi, J.; Roemer, M.E.; Triezenberg, S.J. Curcumin inhibits herpes simplex virus immediate-early gene expression by a mechanism independent of p300/CBP histone acetyltransferase activity. Virology 2008, 373, 239–247. [Google Scholar] [CrossRef] [Green Version]
- Cui, L.; Miao, J.; Cui, L. Cytotoxic effect of curcumin on malaria parasite Plasmodium falciparum: Inhibition of histone acetylation and generation of reactive oxygen species. Antimicrob. Agents Chemother. 2007, 51, 488–494. [Google Scholar] [CrossRef] [Green Version]
- Zhao, W.; Zhou, X.; Qi, G.; Guo, Y. Curcumin suppressed the prostate cancer by inhibiting JNK pathways via epigenetic regulation. J. Biochem. Mol. Toxicol. 2018, 32, e22049. [Google Scholar] [CrossRef]
- Fernandes, G.F.S.; Silva, G.D.B.; Pavan, A.R.; Chiba, D.E.; Chin, C.M.; Dos Santos, J.L. Epigenetic Regulatory Mechanisms Induced by Resveratrol. Nutrients 2017, 9, 1201. [Google Scholar] [CrossRef] [Green Version]
- Imperador, C.H.L.; Scarim, C.B.; Bosquesi, P.L.; Lopes, J.R.; Cardinalli Neto, A.; Giarolla, J.; Ferreira, E.I.; Dos Santos, J.L.; Chin, C.M. Resveratrol and Curcumin for Chagas Disease Treatment-A Systematic Review. Pharmaceuticals 2022, 15, 609. [Google Scholar] [CrossRef]
- Albani, D.; Polito, L.; Signorini, A.; Forloni, G. Neuroprotective properties of resveratrol in different neurodegenerative disorders. BioFactors 2010, 36, 370–376. [Google Scholar] [CrossRef]
- Wahab, A.; Gao, K.; Jia, C.; Zhang, F.; Tian, G.; Murtaza, G.; Chen, J. Significance of Resveratrol in Clinical Management of Chronic Diseases. Molecules 2017, 22, 1329. [Google Scholar] [CrossRef] [Green Version]
- Carrizzo, A.; Forte, M.; Damato, A.; Trimarco, V.; Salzano, F.; Bartolo, M.; Maciag, A.; Puca, A.A.; Vecchione, C. Antioxidant effects of resveratrol in cardiovascular, cerebral and metabolic diseases. Food Chem. Toxicol. 2013, 61, 215–226. [Google Scholar] [CrossRef] [PubMed]
- Gong, W.H.; Zhao, N.; Zhang, Z.M.; Zhang, Y.X.; Yan, L.; Li, J.B. The inhibitory effect of resveratrol on COX-2 expression in human colorectal cancer: A promising therapeutic strategy. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 1136–1143. [Google Scholar]
- Afrin, S.; Giampieri, F.; Gasparrini, M.; Forbes-Hernández, T.Y.; Cianciosi, D.; Reboredo-Rodriguez, P.; Zhang, J.; Manna, P.P.; Daglia, M.; Atanasov, A.G.; et al. Dietary phytochemicals in colorectal cancer prevention and treatment: A focus on the molecular mechanisms involved. Biotechnol. Adv. 2020, 38, 107322. [Google Scholar] [CrossRef]
- Vernousfaderani, E.K.; Akhtari, N.; Rezaei, S.; Rezaee, Y.; Shiranirad, S.; Mashhadi, M.; Hashemi, A.; Khankandi, H.P.; Behzad, S. Resveratrol and Colorectal Cancer: A Molecular Approach to Clinical Researches. Curr. Top. Med. Chem. 2021, 21, 2634–2646. [Google Scholar] [CrossRef] [PubMed]
- Moreira, H.; Szyjka, A.; Grzesik, J.; Pelc, K.; Żuk, M.; Kulma, A.; Emhemmed, F.; Muller, C.D.; Gąsiorowski, K.; Barg, E. Celastrol and Resveratrol Modulate SIRT Genes Expression and Exert Anticancer Activity in Colon Cancer Cells and Cancer Stem-like Cells. Cancers 2022, 14, 1372. [Google Scholar] [CrossRef] [PubMed]
- Venturelli, S.; Berger, A.; Böcker, A.; Busch, C.; Weiland, T.; Noor, S.; Leischner, C.; Schleicher, S.; Mayer, M.; Weiss, T.S.; et al. Resveratrol as a pan-HDAC inhibitor alters the acetylation status of histone [corrected] proteins in human-derived hepatoblastoma cells. PLoS ONE 2013, 8, e73097. [Google Scholar] [CrossRef]
- Dhar, S.; Kumar, A.; Li, K.; Tzivion, G.; Levenson, A.S. Resveratrol regulates PTEN/Akt pathway through inhibition of MTA1/HDAC unit of the NuRD complex in prostate cancer. Biochim. Biophys. Acta 2015, 1853, 265–275. [Google Scholar] [CrossRef] [Green Version]
- Bankole, O.; Scambi, I.; Parrella, E.; Muccilli, M.; Bonafede, R.; Turano, E.; Pizzi, M.; Mariotti, R. Beneficial and Sexually Dimorphic Response to Combined HDAC Inhibitor Valproate and AMPK/SIRT1 Pathway Activator Resveratrol in the Treatment of ALS Mice. Int. J. Mol. Sci. 2022, 23, 1047. [Google Scholar] [CrossRef]
- Han, G.; Xia, J.; Gao, J.; Inagaki, Y.; Tang, W.; Kokudo, N. Anti-tumor effects and cellular mechanisms of resveratrol. Drug Discov. Ther. 2015, 9, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Kai, L.; Samuel, S.K.; Levenson, A.S. Resveratrol enhances p53 acetylation and apoptosis in prostate cancer by inhibiting MTA1/NuRD complex. Int. J. Cancer 2010, 126, 1538–1548. [Google Scholar] [CrossRef] [PubMed]
- Frazzi, R.; Valli, R.; Tamagnini, I.; Casali, B.; Latruffe, N.; Merli, F. Resveratrol-mediated apoptosis of hodgkin lymphoma cells involves SIRT1 inhibition and FOXO3a hyperacetylation. Int. J. Cancer 2013, 132, 1013–1021. [Google Scholar] [CrossRef] [PubMed]
- Adibi, P.; Faghihzadeh, F.; Hekmatdoost, A. Resveratrol and liver: A systematic review. J. Res. Med. Sci. 2015, 20, 797–810. [Google Scholar] [CrossRef]
- Li, Y.; Bäckesjö, C.M.; Haldosén, L.A.; Lindgren, U. Resveratrol inhibits proliferation and promotes apoptosis of osteosarcoma cells. Eur. J. Pharmacol. 2009, 609, 13–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Negri, A.; Naponelli, V.; Rizzi, F.; Bettuzzi, S. Molecular Targets of Epigallocatechin-Gallate (EGCG): A Special Focus on Signal Transduction and Cancer. Nutrients 2018, 10, 1936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saffari, Y.; Sadrzadeh, S.M. Green tea metabolite EGCG protects membranes against oxidative damage in vitro. Life Sci. 2004, 74, 1513–1518. [Google Scholar] [CrossRef] [PubMed]
- Wei, Y.; Chen, P.; Ling, T.; Wang, Y.; Dong, R.; Zhang, C.; Zhang, L.; Han, M.; Wang, D.; Wan, X.; et al. Certain (-)-epigallocatechin-3-gallate (EGCG) auto-oxidation products (EAOPs) retain the cytotoxic activities of EGCG. Food Chem. 2016, 204, 218–226. [Google Scholar] [CrossRef]
- Henning, S.M.; Wang, P.; Carpenter, C.L.; Heber, D. Epigenetic effects of green tea polyphenols in cancer. Epigenomics 2013, 5, 729–741. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Yuan, Y.Y.; Meeran, S.M.; Tollefsbol, T.O. Synergistic epigenetic reactivation of estrogen receptor-α (ERα) by combined green tea polyphenol and histone deacetylase inhibitor in ERα-negative breast cancer cells. Mol. Cancer 2010, 9, 274. [Google Scholar] [CrossRef] [Green Version]
- Chen, D.; Wan, S.B.; Yang, H.; Yuan, J.; Chan, T.H.; Dou, Q.P. EGCG, green tea polyphenols and their synthetic analogs and prodrugs for human cancer prevention and treatment. Adv. Clin. Chem. 2011, 53, 155–177. [Google Scholar] [CrossRef] [Green Version]
- Ferrari, E.; Bettuzzi, S.; Naponelli, V. The Potential of Epigallocatechin Gallate (EGCG) in Targeting Autophagy for Cancer Treatment: A Narrative Review. Int. J. Mol. Sci. 2022, 23, 6075. [Google Scholar] [CrossRef] [PubMed]
- Bansal, S.; Vyas, S.; Bhattacharya, S.; Sharma, M. Catechin prodrugs and analogs: A new array of chemical entities with improved pharmacological and pharmacokinetic properties. Nat. Prod. Rep. 2013, 30, 1438–1454. [Google Scholar] [CrossRef] [PubMed]
- Meeran, S.M.; Patel, S.N.; Chan, T.H.; Tollefsbol, T.O. A novel prodrug of epigallocatechin-3-gallate: Differential epigenetic hTERT repression in human breast cancer cells. Cancer Prev. Res. 2011, 4, 1243–1254. [Google Scholar] [CrossRef] [PubMed]
- Zeng, L.; Holly, J.M.; Perks, C.M. Effects of physiological levels of the green tea extract epigallocatechin-3-gallate on breast cancer cells. Front. Endocrinol. 2014, 5, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sheng, J.; Shi, W.; Guo, H.; Long, W.; Wang, Y.; Qi, J.; Liu, J.; Xu, Y. The Inhibitory Effect of (-)-Epigallocatechin-3-Gallate on Breast Cancer Progression via Reducing SCUBE2 Methylation and DNMT Activity. Molecules 2019, 24, 2899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kato, K.; Long, N.K.; Makita, H.; Toida, M.; Yamashita, T.; Hatakeyama, D.; Hara, A.; Mori, H.; Shibata, T. Effects of green tea polyphenol on methylation status of RECK gene and cancer cell invasion in oral squamous cell carcinoma cells. Br. J. Cancer 2008, 99, 647–654. [Google Scholar] [CrossRef] [Green Version]
- Rodríguez-García, C.; Sánchez-Quesada, C.; Gaforio, J.J. Dietary Flavonoids as Cancer Chemopreventive Agents: An Updated Review of Human Studies. Antioxidants 2019, 8, 137. [Google Scholar] [CrossRef] [Green Version]
- Dabeek, W.M.; Marra, M.V. Dietary Quercetin and Kaempferol: Bioavailability and Potential Cardiovascular-Related Bioactivity in Humans. Nutrients 2019, 11, 2288. [Google Scholar] [CrossRef] [Green Version]
- Vafadar, A.; Shabaninejad, Z.; Movahedpour, A.; Fallahi, F.; Taghavipour, M.; Ghasemi, Y.; Akbari, M.; Shafiee, A.; Hajighadimi, S.; Moradizarmehri, S.; et al. Quercetin and cancer: New insights into its therapeutic effects on ovarian cancer cells. Cell Biosci. 2020, 10, 32. [Google Scholar] [CrossRef] [Green Version]
- Kedhari Sundaram, M.; Hussain, A.; Haque, S.; Raina, R.; Afroze, N. Quercetin modifies 5′CpG promoter methylation and reactivates various tumor suppressor genes by modulating epigenetic marks in human cervical cancer cells. J. Cell. Biochem. 2019, 120, 18357–18369. [Google Scholar] [CrossRef]
- Izzo, S.; Naponelli, V.; Bettuzzi, S. Flavonoids as Epigenetic Modulators for Prostate Cancer Prevention. Nutrients 2020, 12, 1010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Septembre-Malaterre, A.; Boumendjel, A.; Seteyen, A.S.; Boina, C.; Gasque, P.; Guiraud, P.; Sélambarom, J. Focus on the high therapeutic potentials of quercetin and its derivatives. Phytomedicine Plus 2022, 2, 100220. [Google Scholar] [CrossRef] [PubMed]
- Alvarez, M.C.; Maso, V.; Torello, C.O.; Ferro, K.P.; Saad, S.T.O. The polyphenol quercetin induces cell death in leukemia by targeting epigenetic regulators of pro-apoptotic genes. Clin. Epigenetics 2018, 10, 139. [Google Scholar] [CrossRef] [PubMed]
- Bouyahya, A.; El Hachlafi, N.; Aanniz, T.; Bourais, I.; Mechchate, H.; Benali, T.; Shariati, M.A.; Burkov, P.; Lorenzo, J.M.; Wilairatana, P.; et al. Natural Bioactive Compounds Targeting Histone Deacetylases in Human Cancers: Recent Updates. Molecules 2022, 27, 2568. [Google Scholar] [CrossRef]
- Sharma, V.; Kumar, L.; Mohanty, S.K.; Maikhuri, J.P.; Rajender, S.; Gupta, G. Sensitization of androgen refractory prostate cancer cells to anti-androgens through re-expression of epigenetically repressed androgen receptor—Synergistic action of quercetin and curcumin. Mol. Cell. Endocrinol. 2016, 431, 12–23. [Google Scholar] [CrossRef]
- Priyadarsini, R.V.; Vinothini, G.; Murugan, R.S.; Manikandan, P.; Nagini, S. The flavonoid quercetin modulates the hallmark capabilities of hamster buccal pouch tumors. Nutr. Cancer 2011, 63, 218–226. [Google Scholar] [CrossRef]
- Xiao, X.; Shi, D.; Liu, L.; Wang, J.; Xie, X.; Kang, T.; Deng, W. Quercetin suppresses cyclooxygenase-2 expression and angiogenesis through inactivation of P300 signaling. PLoS ONE 2011, 6, e22934. [Google Scholar] [CrossRef]
- Gibellini, L.; Pinti, M.; Nasi, M.; Montagna, J.P.; De Biasi, S.; Roat, E.; Bertoncelli, L.; Cooper, E.L.; Cossarizza, A. Quercetin and cancer chemoprevention. J. Evid. Based Integr. Med. 2011, 2011, 591356. [Google Scholar] [CrossRef] [Green Version]
- Bidian, C.; Mitrea, D.R.; Vasile, O.G.; Filip, A.; Cătoi, A.F.; Moldovan, R.; Decea, N.; Albu, A. Quercetin and curcumin effects in experimental pleural inflammation. Med. Pharm. Rep. 2020, 93, 260–266. [Google Scholar] [CrossRef]
Polyphenols | Molecular Mechanism | Pre Clinical Model | Target Gene |
---|---|---|---|
DNA methylation | |||
Curcumin | DNMT inhibitor | Leukemia, esophageal | NA |
Epicatechin, epicatechin-gallate, epigalocatechin-3-gallate | DNMT inhibitor | Lung, colon cancer cells, esophageal, oral, breast cancers | RAŘ, MGMT, MLH1, CDKN2A, RECK, TERT, RXⱤ, CDX2, GSTP1, W1F1 |
Quercetin | DNMT inhibitor | Breast, colon, esophageal cancers | CDKN2A |
Resveratrol | DNMT inhibitor | Breast, Lungs cancers | NA |
Histone modifications | |||
Curcumin | HAT and HDAC inhibitor | anti-inflammatory anticancer, antioxidant, antiproliferative | GATA4, EOMES, GZMB, PRF1,H3/H4 deacetylation |
Epicatechin, epicatechin-gallate, epigalocatechin-3-gallate | HAT inhibitor | antioxidant, anticancer, anti-inflammatory | NF-kB, IL-6, BMI-1, EZH2, SUZ12, H3K27 trimethylation, H3/H4 acetylation |
Quercetin | SIRTI activator HAT inhibitor | anti-migration, anticancer, antiproliferative, antidiabetic, antioxidant | Inflammatory diseases |
Resveratrol | SIRTi activator | anti-migration, anticancer, antiproliferative, antidiabetic, antioxidant | TNF-˛, IL-8, RBP |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Rajendran, P.; Abdelsalam, S.A.; Renu, K.; Veeraraghavan, V.; Ben Ammar, R.; Ahmed, E.A. Polyphenols as Potent Epigenetics Agents for Cancer. Int. J. Mol. Sci. 2022, 23, 11712. https://doi.org/10.3390/ijms231911712
Rajendran P, Abdelsalam SA, Renu K, Veeraraghavan V, Ben Ammar R, Ahmed EA. Polyphenols as Potent Epigenetics Agents for Cancer. International Journal of Molecular Sciences. 2022; 23(19):11712. https://doi.org/10.3390/ijms231911712
Chicago/Turabian StyleRajendran, Peramaiyan, Salaheldin Abdelraouf Abdelsalam, Kaviyarasi Renu, Vishnupriya Veeraraghavan, Rebai Ben Ammar, and Emad A. Ahmed. 2022. "Polyphenols as Potent Epigenetics Agents for Cancer" International Journal of Molecular Sciences 23, no. 19: 11712. https://doi.org/10.3390/ijms231911712