Targeting Cancer with Phytochemicals via Their Fine Tuning of the Cell Survival Signaling Pathways
Abstract
:1. Introduction
2. Insights on the Role of Flavonoids in Cancer
2.1. Flavonoids and Apoptosis
2.2. Autophagy and the Mitochondria–ER Stress Relatonships Leading to Cancer Cell Death
2.3. Epithelial Mesenchymal Transition (EMT) and Cell Cycle Arrest
2.4. Insights into the Capability of Raw Food and Plant Extracts Containing Phytochemicals to Prevent and Counteract Cancer
3. Conclusions and Future Remarks
Funding
Conflicts of Interest
Abbrevations
AAK | Akt-activated kinase |
Akt | protein kinase B |
AMPK | adenosine-monophosphate-activated proteinkinase |
ARE | antioxidant-responsive element |
ASK1 | apoptosis signal-regulating kinase 1 |
Atg12–Atg5 | autophagy factor12–autophagy factor5 |
Atg7 | autophagy factor 7 |
Bax | Bcl-2-associated X protein |
Bcl-2 | B-cell lymphoma 2 (an antiapoptotic protein) |
Bid | BH3-interacting domain |
BCRP | breast cancer resistance protein |
BNP3 | Bcl-2-adenovirusE1B 19 kDaprotein-interactingprotein3 |
6-CEPN | 6-C-(E-phenylethenyl)naringenin |
Cip1 | cyclin-inhibitor protein 1 (p21) |
CYP | cytochrome P450 |
DAF16 | Dauer-associated factor 16 |
DAPK2 | death-associated protein kinase 2 |
DRP1 | dynamin-related protein 1 |
ERK | extracellularsignal-regulated kinase |
FOXO | forkhead box O3 |
HO-1 | heme oxygenase 1 |
IKK | IκB kinase |
iNOS | inducible nitric oxide synthase |
IRE-JNK-CHOP | iron response element- Janus kinase-cyclophosphamide-hydroxydaunorubicin-oncovyn-prednisone) |
JAK2 | Janus kinase 2 |
JNK | Janus kinase |
p38MAPK | p38 mitogen-activated protein kinase |
MEK | a synonymof MAPK |
MDR1 | multidrug resistant 1 |
Mfn2 | mitofusin-2 |
mTOR | mammalian target of rapamycin |
NB4 | an acute promyelocytic leukemia cell line |
NF-κB | nuclear factor kappa-light-chain-enhancer of activated B cells |
Nrf2 | nuclear factor erythroid 2-related factor 2 |
PARP | poly-ADP-ribose polymerase |
PERK–ATF4 | protein kinase RNA-like endoplasmic reticulum kinase–activating transcription factor 4 |
PGC1α | peroxisome proliferator-activated receptor gamma coactivator 1-alpha |
PGE2 | prostaglandin E2 |
pEGFR | phosphorylated epithelial growth factor receptor |
PKA-PI3K | protein kinase A-phosphoinositide 3 kinase |
Raf1 | Ras-associated factor 1 |
SIR 2-1 | sirtuin 2-1 |
SIRT1 | sirtuin 1 |
Snail/Slug | transcription actors in EMT |
STAT3 | signal transducer and activator of transcription 3 |
TFAM | transcription factor A, mitochondrial |
TRAIL | TNF-related apoptosis-inducing ligand |
Wnt | wingless/integrated |
References
- Chen, C.Y.; Kao, C.L.; Liu, C.M. The Cancer Prevention, Anti-Inflammatory and Anti Oxidation of Bioactive Phytochemicals Targeting the TLR4 Signaling Pathway. Int. J. Mol. Sci. 2018, 19, 2729. [Google Scholar] [CrossRef] [PubMed]
- Chiou, Y.S.; Li, S.; Ho, C.T.; Pan, M.H. Prevention of Breast Cancer by Natural Phytochemicals: Focusing on Molecular Targets and Combinational Strategy. Mol. Nutr. Food Res. 2018, e1800392. [Google Scholar] [CrossRef] [PubMed]
- Bonam, S.R.; Wu, Y.S.; Tunki, L.; Chellian, R.; Halmuthur, M.S.K.; Muller, S.; Pandy, V. What Has come out from Phytomedicines and Herbal Edibles for the Treatment of Cancer? Chem. Med. Chem. 2018, 13, 1854–1872. [Google Scholar] [CrossRef] [PubMed]
- Galati, G.; O’Brien, P.J. Potential toxicity of flavonoids and other dietary phenolics: Significance for their chemopreventive and anticancer properties. Free Radic. Biol. Med. 2004, 37, 287–303. [Google Scholar] [CrossRef] [PubMed]
- Liu, R.H. Health benefits of fruit and vegetables are from additive and synergistic combinations of phytochemicals. Am. J. Clin. Nutr. 2003, 78 (Suppl. 3), S517–S520. [Google Scholar] [CrossRef] [PubMed]
- Sunan, W.; Meckling, K.; Marcone, M.F.; Tasao, R. Can phytochemical antioxidant rich foods act as anti-cancer agents? Food Res. Int. 2011, 44, 2545–2554. [Google Scholar]
- Yang, J.H.; Shin, B.Y.; Han, J.Y.; Kim, M.G.; Wi, J.E.; Kim, Y.W.; Cho, I.J.; Kim, S.C.; Shin, S.M.; Ki, S.H. Isorhamnetin protects against oxidative stress by activating Nrf2 and inducing the expression of its target genes. Toxicol. Appl. Pharmacol. 2014, 274, 293–301. [Google Scholar] [CrossRef] [PubMed]
- Aboonabi, A.; Singh, I. Chemopreventive role of anthocyanins in atherosclerosis via activation of Nrf2-ARE as an indicator and modulator of redox. Biomed. Pharmacother. 2015, 72, 30–36. [Google Scholar] [CrossRef] [PubMed]
- Costa, L.G.; Garrick, J.M.; Roquè, P.J.; Pellacani, C. Mechanisms of Neuroprotection by Quercetin: Counteracting Oxidative Stress and More. Oxid. Med. Cell. Longev. 2016, 2016, 2986796. [Google Scholar] [CrossRef] [PubMed]
- Tebay, L.E.; Robertson, H.; Durant, S.T.; Vitale, S.R.; Penning, T.M.; Dinkova-Kostova, A.T.; Hayes, J.D. Mechanisms of activation of the transcription factor Nrf2 by redox stressors, nutrient cues, and energy status and the pathways through which it attenuates degenerative disease. Free Radic. Biol. Med. 2015, 88, 108–146. [Google Scholar] [CrossRef] [Green Version]
- Ighodaro, Q.M.; Akinloye, Q.A. First line defence antioxidants-superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPX): Their fundamental role in the entire antioxidant defence grid Alexandria. J. Med. 2017, in press. [Google Scholar] [CrossRef]
- Clere, N.; Faure, S.; Martinez, M.C.; Andriantsitohaina, R. Anticancer properties of flavonoids: Roles in various stages of carcinogenesis. Cardiovasc. Hematol. Agents Med. Chem. 2011, 9, 62–77. [Google Scholar] [CrossRef] [PubMed]
- Qiu, T.; Wu, D.; Yang, L.; Ye, H.; Wang, Q.; Cao, Z.; Tang, K. Exploring the Mechanism of Flavonoids Through Systematic Bioinformatics Analysis. Front. Pharmacol. 2018, 9, 918. [Google Scholar] [CrossRef] [PubMed]
- Rauf, A.; Imran, M.; Khan, I.A.; Ur-Rehman, M.; Gilani, S.A.; Mehmood, Z.; Mubarak, M.S. Anticancer potential of quercetin: A comprehensive review. Phytother. Res. 2018. [Google Scholar] [CrossRef] [PubMed]
- Cook, M.T. Mechanism of metastasis suppression by luteolin in breast cancer. Breast Cancer (Dove Med. Press) 2018, 10, 89–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sak, K. Site-specific anticancer effects of dietary flavonoid quercetin. Nutr. Cancer 2014, 66, 177–193. [Google Scholar] [CrossRef] [PubMed]
- Chirumbolo, S.; Bjørklund, G. PERM Hypothesis: The Fundamental Machinery Able to Elucidate the Role of Xenobiotics and Hormesis in Cell Survival and Homeostasis. Int. J. Mol. Sci. 2017, 18, 165. [Google Scholar] [CrossRef] [PubMed]
- Sabharwal, S.S.; Schumacker, P.T. Mitochondrial ROS in cancer: Initiators, amplifiers or an Achilles’ heel? Nat. Rev. Cancer 2014, 14, 709–721. [Google Scholar] [CrossRef] [PubMed]
- Ralph, S.J.; Rodríguez-Enríquez, S.; Neuzil, J.; Saavedra, E.; Moreno-Sánchez, R. The causes of cancer revisited: “mitochondrial malignancy” and ROS-induced oncogenic transformation—Why mitochondria are targets for cancer therapy. Mol. Asp. Med. 2010, 31, 145–170. [Google Scholar] [CrossRef] [PubMed]
- Wallace, D.C. Mitochondria and cancer. Nat. Rev. Cancer 2012, 12, 685–698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bellot, G.L.; Liu, D.; Pervaiz, S. ROS, autophagy, mitochondria and cancer: Ras, the hidden master? Mitochondrion 2013, 13, 155–162. [Google Scholar] [CrossRef] [PubMed]
- Xiong, L.G.; Chen, Y.J.; Tong, J.W.; Gong, Y.S.; Huang, J.A.; Liu, Z.H. Epigallocatechin-3-gallate promotes healthy lifespan through mitohormesis during early-to-mid adulthood in Caenorhabditis elegans. Redox Biol. 2018, 14, 305–315. [Google Scholar] [CrossRef] [PubMed]
- Steib, K.; Schäffner, I.; Jagasia, R.; Ebert, B.; Lie, D.C. Mitochondria modify exercise-induced development of stem cell-derived neurons in the adult brain. J. Neurosci. 2014, 34, 6624–6633. [Google Scholar] [CrossRef] [PubMed]
- Naik, P.P.; Birbrair, A.; Bhutia, S.K. Mitophagy-driven metabolic switch reprograms stem cell fate. Cell Mol. Life Sci. 2018. [Google Scholar] [CrossRef] [PubMed]
- Mitra, K. Mitochondrial fission-fusion as an emerging key regulator of cell proliferation and differentiation. Bioessays 2013, 35, 955–964. [Google Scholar] [CrossRef] [PubMed]
- Dal Yontem, F.; Kim, S.H.; Ding, Z.; Grimm, E.; Ekmekcioglu, S.; Akcakaya, H. Mitochondrial dynamic alterations regulate melanoma cell progression. J. Cell Biochem. 2018. [Google Scholar] [CrossRef] [PubMed]
- Huang, Y.; Chen, K.; Ren, Q.; Yi, L.; Zhu, J.; Zhang, Q.; Mi, M. Dihydromyricetin Attenuates Dexamethasone-Induced Muscle Atrophy by Improving Mitochondrial Function via the PGC-1α Pathway. Cell Physiol. Biochem. 2018, 49, 758–779. [Google Scholar] [CrossRef] [PubMed]
- Lennon, F.E.; Salgia, R. Mitochondrial dynamics: Biology and therapy in lung cancer. Expert Opin. Investig. Drugs 2014, 23, 675–692. [Google Scholar] [CrossRef] [PubMed]
- Zhao, J.; Zhang, J.; Yu, M.; Xie, Y.; Huang, Y.; Wolff, D.W.; Abel, P.W.; Tu, Y. Mitochondrial dynamics regulates migration and invasion of breast cancer cells. Oncogene 2013, 32, 4814–4824. [Google Scholar] [CrossRef] [PubMed]
- Che, T.F.; Lin, C.W.; Wu, Y.Y.; Chen, Y.J.; Han, C.L.; Chang, Y.L.; Wu, C.T.; Hsiao, T.H.; Hong, T.M.; Yang, P.C. Mitochondrial translocation of EGFR regulates mitochondria dynamics and promotes metastasis in NSCLC. Oncotarget 2015, 6, 37349–37366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- White, E.; Mehnert, J.M.; Chan, C.S. Autophagy, Metabolism, and Cancer. Clin. Cancer Res. 2015, 21, 5037–5046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Goldsmith, J.; Levine, B.; Debnath, J. Autophagy and cancer metabolism. Methods Enzymol. 2014, 542, 25–57. [Google Scholar] [PubMed]
- Poillet-Perez, L.; Despouy, G.; Delage-Mourroux, R.; Boyer-Guittaut, M. Interplay between ROS and autophagy in cancer cells, from tumor initiation to cancer therapy. Redox Biol. 2015, 4, 184–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hasima, N.; Ozpolat, B. Regulation of autophagy by polyphenolic compounds as a potential therapeutic strategy for cancer. Cell Death Dis. 2014, 5, e1509. [Google Scholar] [CrossRef] [PubMed]
- Radogna, F.; Dicato, M.; Diederich, M. Cancer-type-specific crosstalk between autophagy, necroptosis and apoptosis as a pharmacological target. Biochem. Pharmacol. 2015, 94, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Khorsandi, L.; Orazizadeh, M.; Niazvand, F.; Abbaspour, M.R.; Mansouri, E.; Khodadadi, A. Quercetin induces apoptosis and necroptosis in MCF-7 breast cancer cells. Bratisl. Lek. Listy 2017, 118, 123–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lin, W.; Tongyi, S. Role of Bax/Bcl-2 family members in green tea polyphenol induced necroptosis of p53-deficient Hep3B cells. Tumour Biol. 2014, 35, 8065–8075. [Google Scholar] [CrossRef] [PubMed]
- White, E. The role for autophagy in cancer. J. Clin. Investig. 2015, 125, 42–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brunelli, E.; Pinton, G.; Bellini, P.; Minassi, A.; Appendino, G.; Moro, L. Flavonoid-induced autophagy in hormone sensitive breast cancer cells. Fitoterapia 2009, 80, 327–332. [Google Scholar] [CrossRef] [PubMed]
- Zheng, N.; Liu, L.; Liu, W.W.; Li, F.; Hayashi, T.; Tashiro, S.I.; Onodera, S.; Ikejima, T. Crosstalk of ROS/RNS and autophagy in silibinin-induced apoptosis of MCF-7 human breast cancer cells in vitro. Acta Pharmacol. Sin. 2017, 38, 277–289. [Google Scholar] [CrossRef] [PubMed]
- Han, X.; Liu, C.F.; Gao, N.; Zhao, J.; Xu, J. Kaempferol suppresses proliferation but increases apoptosis and autophagy by up-regulating microRNA-340 in human lung cancer cells. Biomed. Pharmacother. 2018, 108, 809–816. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.W.; Hu, J.J.; Fu, R.Q.; Liu, X.; Zhang, Y.H.; Li, J.; Liu, L.; Li, Y.N.; Deng, Q.; Luo, Q.S.; et al. Flavonoids inhibit cell proliferation and induce apoptosis and autophagy through downregulation of PI3Kγ mediated PI3K/AKT/mTOR/p70S6K/ULK signaling pathway in human breast cancer cells. Sci. Rep. 2018, 8, 11255. [Google Scholar] [CrossRef] [PubMed]
- Kania, E.; Pająk, B.; Orzechowski, A. Calcium homeostasis and ER stress in control of autophagy in cancer cells. Biomed. Res. Int. 2015, 2015, 352794. [Google Scholar] [CrossRef] [PubMed]
- Verfaillie, T.; Garg, A.D.; Agostinis, P. Targeting ER stress induced apoptosis and inflammation in cancer. Cancer Lett. 2013, 332, 249–264. [Google Scholar] [CrossRef] [PubMed]
- Duluc, L.; Soleti, R.; Clere, N.; Andriantsitohaina, R.; Simard, G. Mitochondria as potential targets of flavonoids: Focus on adipocytes and endothelial cells. Curr. Med. Chem. 2012, 19, 4462–4474. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Chen, W.; Zhang, B.; Tian, F.; Zhou, Z.; Liao, X.; Li, C.; Zhang, Y.; Han, Y.; Wang, Y.; et al. Lon in maintaining mitochondrial and endoplasmic reticulum homeostasis. Arch. Toxicol. 2018, 92, 1913–1923. [Google Scholar] [CrossRef] [PubMed]
- Polo, M.; Alegre, F.; Moragrega, A.B.; Gibellini, L.; Marti-Rodrigo, A.; Blas-Garcia, A.; Esplugues, J.V.; Apostolova, N. Lon protease: A novel mitochondrial matrix protein in the interconnection between drug-induced mitochondrial dysfunction and endoplasmic reticulum stress. Br. J. Pharmacol. 2017, 174, 4409–4429. [Google Scholar] [CrossRef] [PubMed]
- Hori, O.; Ichinoda, F.; Tamatani, T.; Yamaguchi, A.; Sato, N.; Ozawa, K.; Kitao, Y.; Miyazaki, M.; Harding, H.P.; Ron, D.; et al. Transmission of cell stress from endoplasmic reticulum to mitochondria: Enhanced expression of Lon protease. J. Cell Biol. 2002, 157, 1151–1160. [Google Scholar] [CrossRef] [PubMed]
- Rainbolt, T.K.; Saunders, J.M.; Wiseman, R.L. Stress-responsive regulation of mitochondria through the ER unfolded protein response. Trends Endocrinol. Metab. 2014, 25, 528–537. [Google Scholar] [CrossRef] [PubMed]
- Ehren, J.L.; Maher, P. Concurrent regulation of the transcription factors Nrf2 and ATF4 mediates the enhancement of glutathione levels by the flavonoid fisetin. Biochem. Pharmacol. 2013, 85, 1816–1826. [Google Scholar] [CrossRef] [PubMed]
- Li, X.M.; Liu, J.; Pan, F.F.; Shi, D.D.; Wen, Z.G.; Yang, P.L. Quercetin and aconitine synergistically induces the human cervical carcinoma HeLa cell apoptosis via endoplasmic reticulum (ER) stress pathway. PLoS ONE 2018, 13, e0191062. [Google Scholar] [CrossRef] [PubMed]
- Liu, C.M.; Ma, J.Q.; Sun, J.M.; Feng, Z.J.; Cheng, C.; Yang, W.; Jiang, H. Association of changes in ER stress-mediated signaling pathway with lead-induced insulin resistance and apoptosis in rats and their prevention by A-type dimeric epigallocatechin-3-gallate. Food Chem. Toxicol. 2017, 110, 325–332. [Google Scholar] [CrossRef] [PubMed]
- García-Aguilar, A.; Cuezva, J.M. A Review of the Inhibition of the Mitochondrial ATP Synthase by IF1 in vivo: Reprogramming Energy Metabolism Inducing Mitohormesis. Front. Physiol. 2018, 9, 1322. [Google Scholar] [CrossRef] [PubMed]
- Bravo-Sagua, R.; Parra, V.; Ortiz-Sandoval, C.; Navarro-Marquez, M.; Rodríguez, A.E.; Diaz-Valdivia, N.; Sanhueza, C.; Lopez-Crisosto, C.; Tahbaz, N.; Rothermel, B.A.; et al. Caveolin-1 impairs PKA-DRP1-mediated remodelling of ER-mitochondria communication during the early phase of ER stress. Cell Death Differ. 2018. [Google Scholar] [CrossRef] [PubMed]
- Chang, C.R.; Blackstone, C. Drp1 phosphorylation and mitochondrial regulation. EMBO Rep. 2007, 8, 1088–1089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cui, L.; Li, Z.; Chang, X.; Cong, G.; Hao, L. Quercetin attenuates vascular calcification by inhibiting oxidative stress and mitochondrial fission. Vascul. Pharmacol. 2017, 88, 21–29. [Google Scholar] [CrossRef] [PubMed]
- Ye, F.; Wang, H.; Zhang, L.; Zou, Y.; Han, H.; Huang, J. Baicalein induces human osteosarcoma cell line MG-63 apoptosis via ROS-induced BNIP3 expression. Tumour Biol. 2015, 36, 4731–4740. [Google Scholar] [CrossRef] [PubMed]
- Herr, I.; Debatin, K.M. Cellular stress response and apoptosis in cancer therapy. Blood 2001, 98, 2603–2614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chircop, M.; Speidel, D. Cellular stress responses in cancer and cancer therapy. Front. Oncol. 2014, 4, 304. [Google Scholar] [CrossRef] [PubMed]
- Ozbey, U.; Attar, R.; Romero, M.A.; Alhewairini, S.S.; Afshar, B.; Sabitaliyevich, U.Y.; Hanna-Wakim, L.; Ozcelik, B.; Farooqi, A.A. Apigenin as an effective anticancer natural product: Spotlight on TRAIL, WNT/β-catenin, JAK-STAT pathways, and microRNAs. J. Cell Biochem. 2018, in press. [Google Scholar] [CrossRef] [PubMed]
- Liao, Y.; Xu, Y.; Cao, M.; Huan, Y.; Zhu, L.; Jiang, Y.; Shen, W.; Zhu, G. Luteolin Induces Apoptosis and Autophagy in Mouse Macrophage ANA-1 Cells via the Bcl-2 Pathway. J. Immunol. Res. 2018, 2018, 4623919. [Google Scholar] [CrossRef] [PubMed]
- Dong, X.; Zhang, J.; Yang, F.; Wu, J.; Cai, R.; Wang, T.; Zhang, J. Effect of luteolin on the methylation status of the OPCML gene and cell growth in breast cancer cells. Exp. Ther. Med. 2018, 16, 3186–3194. [Google Scholar] [CrossRef] [PubMed]
- Yumnam, S.; Raha, S.; Kim, S.M.; Venkatarame Gowda Saralamma, V.; Lee, H.J.; Ha, S.E.; Heo, J.D.; Lee, S.J.; Kim, E.H.; Lee, W.S.; et al. Identification of a novel biomarker in tangeretin-induced cell death in AGS human gastric cancer cells. Oncol. Rep. 2018, in press. [Google Scholar] [CrossRef] [PubMed]
- Surichan, S.; Arroo, R.R.; Tsatsakis, A.M.; Androutsopoulos, V.P. Tangeretin inhibits the proliferation of human breast cancer cells via CYP1A1/CYP1B1 enzyme induction and CYP1A1/CYP1B1-mediated metabolism to the product 4’ hydroxy tangeretin. Toxicol. In Vitro 2018, 50, 274–284. [Google Scholar] [CrossRef] [PubMed]
- Hirpara, K.V.; Aggarwal, P.; Mukherjee, A.J.; Joshi, N.; Burman, A.C. Quercetin and its derivatives: synthesis, pharmacological uses with special emphasis on anti-tumor properties and prodrug with enhanced bio-availability. Anticancer Agents Med. Chem. 2009, 9, 138–161. [Google Scholar] [CrossRef] [PubMed]
- Kuo, W.T.; Tsai, Y.C.; Wu, H.C.; Ho, Y.J.; Chen, Y.S.; Yao, C.H.; Yao, C.H. Radiosensitization of non-small cell lung cancer by kaempferol. Oncol. Rep. 2015, 34, 2351–2356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Choi, J.B.; Kim, J.H.; Lee, H.; Pak, J.N.; Shim, B.S.; Kim, S.H. Reactive Oxygen Species and p53 Mediated Activation of p38 and Caspases is Critically Involved in Kaempferol Induced Apoptosis in Colorectal Cancer Cells. J. Agric. Food Chem. 2018, 66, 9960–9967. [Google Scholar] [CrossRef] [PubMed]
- Ye, C.; Zhang, C.; Huang, H.; Yang, B.; Xiao, G.; Kong, D.; Tian, Q.; Song, Q.; Song, Y.; Tan, H.; et al. The Natural Compound Myricetin Effectively Represses the Malignant Progression of Prostate Cancer by Inhibiting PIM1 and Disrupting the PIM1/CXCR4 Interaction. Cell Physiol. Biochem. 2018, 48, 1230–1244. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Gong, X.; Jiang, R.; Lin, D.; Zhou, T.; Zhang, A.; Li, H.; Zhang, X.; Wan, J.; Kuang, G.; et al. Fisetin Inhibited Growth and Metastasis of Triple-Negative Breast Cancer by Reversing Epithelial-to-Mesenchymal Transition via PTEN/Akt/GSK3β Signal Pathway. Front. Pharmacol. 2018, 9, 772. [Google Scholar] [CrossRef] [PubMed]
- Zhu, Y.; Rao, Q.; Zhang, X.; Zhou, X. Galangin induced antitumor effects in human kidney tumor cells mediated via mitochondrial mediated apoptosis, inhibition of cell migration and invasion and targeting PI3K/ AKT/mTOR signalling pathway. J. BUON 2018, 23, 795–799. [Google Scholar] [PubMed]
- Wu, Q.; Kroon, P.A.; Shao, H.; Needs, P.W.; Yang, X. Differential Effects of Quercetin and Two of Its Derivatives, Isorhamnetin and Isorhamnetin-3-glucuronide, in Inhibiting the Proliferation of Human Breast-Cancer MCF-7 Cells. J. Agric. Food Chem. 2018, 66, 7181–7189. [Google Scholar] [CrossRef] [PubMed]
- Elango, R.; Athinarayanan, J.; Subbarayan, V.P.; Lei, D.K.Y.; Alshatwi, A.A. Hesperetin induces an apoptosis-triggered extrinsic pathway and a p53- independent pathway in human lung cancer H522 cells. J. Asian Nat. Prod. Res. 2018, 20, 559–569. [Google Scholar] [CrossRef] [PubMed]
- GumushanAktas, H.; Akgun, T. Naringenin inhibits prostate cancer metastasis by blocking voltage-gated sodium channels. Biomed. Pharmacother. 2018, 106, 770–775. [Google Scholar] [CrossRef] [PubMed]
- Haque, M.W.; Bose, P.; Siddique, M.U.M.; Sunita, P.; Lapenna, A.; Pattanayak, S.P. Taxifolin binds with LXR (α & β) to attenuate DMBA-induced mammary carcinogenesis through mTOR/Maf-1/PTEN pathway. Biomed. Pharmacother. 2018, 105, 27–36. [Google Scholar] [PubMed]
- Haque, M.W.; Pattanayak, S.P. Taxifolin Inhibits 7,12-Dimethylbenz(a)anthracene-induced Breast Carcinogenesis by Regulating AhR/CYP1A1 Signaling Pathway. Pharmacogn. Mag. 2018, 13 (Suppl. 4), S749–S755. [Google Scholar] [PubMed]
- Grube, S.; Ewald, C.; Kögler, C.; Lawson McLean, A.; Kalff, R.; Walter, J. Achievable Central Nervous System Concentrations of the Green Tea Catechin EGCG Induce Stress in Glioblastoma Cells in Vitro. Nutr. Cancer 2018, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Xiao, X.; Jiang, K.; Xu, Y.; Peng, H.; Wang, Z.; Liu, S.; Zhang, G. (-)-Epigallocatechin-3-gallate induces cell apoptosis in chronic myeloid leukemia by regulating Bcr/Abl-mediated p38-MAPK/JNK and JAK2/STAT3/AKT signaling pathways. Clin. Exp. Pharmacol. Physiol. 2018. [Google Scholar] [CrossRef] [PubMed]
- Rawangkan, A.; Wongsirisin, P.; Namiki, K.; Iida, K.; Kobayashi, Y.; Shimizu, Y.; Fujiki, H.; Suganuma, M. Green Tea Catechin Is an Alternative Immune Checkpoint Inhibitor that Inhibits PD-L1 Expression and Lung Tumor Growth. Molecules 2018, 23, 2071. [Google Scholar] [CrossRef] [PubMed]
- Stadlbauer, S.; Steinborn, C.; Klemd, A.; Hattori, F.; Ohmori, K.; Suzuki, K.; Huber, R.; Wolf, P.; Gründemann, C. Impact of Green Tea Catechin ECG and Its Synthesized Fluorinated Analogue on Prostate Cancer Cells and Stimulated Immunocompetent Cells. Planta Med. 2018, 84, 813–819. [Google Scholar] [CrossRef] [PubMed]
- Mukherjee, S.; Hussaini, R.; White, R.; Atwi, D.; Fried, A.; Sampat, S.; Piao, L.; Pan, Q.; Banerjee, P. TriCurin, a synergistic formulation of curcumin, resveratrol, and epicatechin gallate, repolarizes tumor-associated macrophages and triggers an immune response to cause suppression of HPV+ tumors. Cancer Immunol. Immunother. 2018, 67, 761–774. [Google Scholar] [CrossRef] [PubMed]
- Ma, X.; Ning, S. Cyanidin-3-glucoside attenuates the angiogenesis of breast cancer via inhibiting STAT3/VEGF pathway. Phytother. Res. 2018. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Zhu, Y.; Zhang, W.; Peng, X.; Zhou, J.; Li, F.; Han, B.; Liu, X.; Ou, Y.; Yu, X. Delphinidin induced protective autophagy via mTOR pathway suppression and AMPK pathway activation in HER-2 positive breast cancer cells. BMC Cancer 2018, 18, 342. [Google Scholar] [CrossRef] [PubMed]
- Kang, H.M.; Park, B.S.; Kang, H.K.; Park, H.R.; Yu, S.B.; Kim, I.R. Delphinidin induces apoptosis and inhibits epithelial-to-mesenchymal transition via the ERK/p38 MAPK-signaling pathway in human osteosarcoma cell lines. Environ. Toxicol. 2018, 33, 640–649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, C.H.; Zhang, Y.X.; Tang, L.H.; Yang, X.J.; Cui, W.M.; Han, C.C.; Ji, W.Y. MicroRNA-1469, a p53-responsive microRNA promotes Genistein induced apoptosis by targeting Mcl1 in human laryngeal cancer cells. Biomed. Pharmacother. 2018, 106, 665–671. [Google Scholar] [CrossRef] [PubMed]
- Venza, I.; Visalli, M.; Oteri, R.; Beninati, C.; Teti, D.; Venza, M. Genistein reduces proliferation of EP3-expressing melanoma cells through inhibition of PGE2-induced IL-8 expression. Int. Immunopharmacol. 2018, 62, 86–95. [Google Scholar] [CrossRef] [PubMed]
- Dutta, B.; Park, J.E.; Qing, I.T.Y.; Kon, O.L.; Sze, S.K. Soy-Derived Phytochemical Genistein Modifies Chromatome Topology to Restrict Cancer Cell Proliferation. Proteomics 2018, 18, e1700474. [Google Scholar] [CrossRef] [PubMed]
- Liang, Y.S.; Qi, W.T.; Guo, W.; Wang, C.L.; Hu, Z.B.; Li, A.K. Genistein and daidzein induce apoptosis of colon cancer cells by inhibiting the accumulation of lipid droplets. Food Nutr. Res. 2018, 62. [Google Scholar] [CrossRef] [PubMed]
- Brusselmans, K.; Vrolix, R.; Verhoeven, G.; Swinnen, J.V. Induction of cancer cell apoptosis by flavonoids is associated with their ability to inhibit fatty acid synthase activity. J. Biol. Chem. 2005, 280, 5636–5645. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.S.; Lei, J.P.; Wei, G.Q.; Chen, H.; Ma, C.Y.; Jiang, H.Z. Natural fatty acid synthase inhibitors as potent therapeutic agents for cancers: A review. Pharm. Biol. 2016, 54, 1919–1925. [Google Scholar] [CrossRef] [PubMed]
- Bandyopadhyay, S.; Zhan, R.; Wang, Y.; Pai, S.K.; Hirota, S.; Hosobe, S.; Takano, Y.; Saito, K.; Furuta, E.; Iiizumi, M.; et al. Mechanism of apoptosis induced by the inhibition of fatty acid synthase in breast cancer cells. Cancer Res. 2006, 66, 5934–5940. [Google Scholar] [CrossRef] [PubMed]
- Li, B.; Espinoza, I.; Liu, H.; Lupu, R. Inhibition of fatty acid synthase induces reactive oxygen species (ROS) to inhibit HER2 overexpressing beast cancer cell growth. Cancer Research. In Proceedings of the 2007 98th AACR Annual Meeting, Los Angeles, CA, USA, 14–18 April 2007; Abst 4462. American Association for Cancer Research: Philadelphia, PA, USA, 2007. [Google Scholar]
- Niki, E. Oxidative stress and antioxidants: Distress or eustress? Arch Biochem. Biophys. 2016, 595, 19–24. [Google Scholar] [CrossRef] [PubMed]
- Murakami, A. Dose-dependent functionality and toxicity of green tea polyphenols in experimental rodents. Arch. Biochem. Biophys. 2014, 557, 3–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Desikan, R.; Hancock, J.; Neill, S. Reactive Oxygen Species as Signalling Molecules in “Antioxidants Reactive Oxygen Species in Plants”; Smirnoff, N., Ed.; Blackwell Publl: Oxford, UK, 2005; Chapter 7; pp. 169–195. [Google Scholar]
- Kumari, S.; Badana, A.K.; GMM, G.S.; Malla, R. Reactive Oxygen Species: A Key Constituent in Cancer Survival. Biomark. Insights 2018, 13. [Google Scholar] [CrossRef] [PubMed]
- Rubio, V.; García-Pérez, A.I.; Herráez, A.; Diez, J.C. Different roles of Nrf2 and NF-κB in the antioxidant imbalance produced by esculetin or quercetin on NB4 leukemia cells. Chem. Biol. Interact. 2018, 294, 158–166. [Google Scholar] [CrossRef] [PubMed]
- Khatami, M. Cancer; an induced disease of twentieth century! Induction of tolerance, increased entropy and ‘Dark Energy’: Loss of biorhythms (Anabolism v. Catabolism). Clin. Transl. Med. 2018, 7, 20. [Google Scholar] [CrossRef] [PubMed]
- Isono, M.; Sato, A.; Asano, T.; Okubo, K.; Asano, T. Evaluation of Therapeutic Potential of Phenoxodiol, a Novel Isoflavone Analog, in Renal Cancer Cells. Anticancer Res. 2018, 38, 5709–5716. [Google Scholar] [CrossRef] [PubMed]
- Chien, K.J.; Yang, M.L.; Tsai, P.K.; Su, C.H.; Chen, C.H.; Horng, C.T.; Yeh, C.H.; Chen, W.Y.; Lin, M.L.; Chen, C.J.; et al. Safrole induced cytotoxicity, DNA damage, and apoptosis in macrophages via reactive oxygen species generation and Akt phosphorylation. Environ. Toxicol. Pharmacol. 2018, 64, 94–100. [Google Scholar] [CrossRef] [PubMed]
- Im, E.; Yeo, C.; Lee, E.O. Luteolin induces caspase-dependent apoptosis via inhibiting the AKT/osteopontin pathway in human hepatocellular carcinoma SK-Hep-1 cells. Life Sci. 2018, 209, 259–266. [Google Scholar] [CrossRef] [PubMed]
- Gu, Z.F.; Zhang, Z.T.; Wang, J.Y.; Xu, B.B. Icariin exerts inhibitory effects on the growth and metastasis of KYSE70 human esophageal carcinoma cells via PI3K/AKT and STAT3 pathways. Environ. Toxicol. Pharmacol. 2017, 54, 7–13. [Google Scholar] [CrossRef] [PubMed]
- Yu, M.; Qi, B.; Xiaoxiang, W.; Xu, J.; Liu, X. Baicalein increases cisplatin sensitivity of A549 lung adenocarcinoma cells via PI3K/Akt/NF-κB pathway. Biomed. Pharmacother. 2017, 90, 677–685. [Google Scholar] [CrossRef] [PubMed]
- Zhou, H.; Li, X.M.; Meinkoth, J.; Pittman, R.N. Akt regulates cell survival and apoptosis at a postmitochondrial level. J. Cell Biol. 2000, 151, 483–494. [Google Scholar] [CrossRef] [PubMed]
- Ahn, J.; Won, M.; Choi, J.H.; Kim, Y.S.; Jung, C.R.; Im, D.S.; Kyun, M.L.; Lee, K.; Song, K.B.; Chung, K.S. Reactive oxygen species-mediated activation of the Akt/ASK1/p38 signaling cascade and p21(Cip1) downregulation are required for shikonin-induced apoptosis. Apoptosis 2013, 18, 870–881. [Google Scholar] [CrossRef] [PubMed]
- Pan, J.; Chang, Q.; Wang, X.; Son, Y.; Zhang, Z.; Chen, G.; Luo, J.; Bi, Y.; Chen, F.; Shi, X. Reactive oxygen species-activated Akt/ASK1/p38 signaling pathway in nickel compound-induced apoptosis in BEAS 2B cells. Chem. Res. Toxicol. 2010, 23, 568–577. [Google Scholar] [CrossRef] [PubMed]
- Puckett, M.C.; Goldman, E.H.; Cockrell, L.M.; Huang, B.; Kasinski, A.L.; Du, Y.; Wang, C.Y.; Lin, A.; Ichijo, H.; Khuri, F.; et al. Integration of apoptosis signal-regulating kinase 1-mediated stress signaling with the Akt/protein kinase B-IκB kinase cascade. Mol. Cell Biol. 2013, 33, 2252–2259. [Google Scholar] [CrossRef] [PubMed]
- Kaltschmidt, B.; Greiner, J.F.W.; Kadhim, H.M.; Kaltschmidt, C. Subunit-Specific Role of NF-κB in Cancer. Biomedicines 2018, 6, 44. [Google Scholar] [CrossRef] [PubMed]
- Ashraf, A.H.M.Z.; Afroze, S.H.; Yamauchi, K.; Zawieja, D.C.; Keuhl, T.J.; Erlandson, L.W.; Uddin, M.N. Differential Mechanism of Action of 3,4′,7-O-trimethylquercetin in Three Types of Ovarian Cancer Cells. Anticancer Res. 2018, 38, 5131–5137. [Google Scholar] [CrossRef] [PubMed]
- Valko, M.; Rhodes, C.J.; Moncol, J.; Izakovic, M.; Mazur, M. Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem. Biol. Interact. 2006, 160, 1–40. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.T.; Chen, Y.T.; Hsieh, Y.H.; Weng, C.J.; Yeh, J.C.; Yang, S.F.; Lin, C.W.; Yang, J.S. Glabridin induces apoptosis and cell cycle arrest in oral cancer cells through the JNK1/2 signaling pathway. Environ. Toxicol. 2018, 33, 679–685. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.; Zhang, Y.; Ding, K.; Zhang, H.; Zhao, Q.; Liu, Z.; Xu, Y. Involvement of JNK1/2-NF-κBp65 in the regulation of HMGB2 in myocardial ischemia/reperfusion-induced apoptosis in human AC16 cardiomyocytes. Biomed. Pharmacother. 2018, 106, 1063–1071. [Google Scholar] [CrossRef] [PubMed]
- Cincin, Z.B.; Kiran, B.; Baran, Y.; Cakmakoglu, B. Hesperidin promotes programmed cell death by downregulation of nongenomic estrogen receptor signalling pathway in endometrial cancer cells. Biomed. Pharmacother. 2018, 103, 336–345. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Pi, C.; Wang, G. Inhibition of PI3K/Akt/mTOR pathway by apigenin induces apoptosis and autophagy in hepatocellular carcinoma cells. Biomed. Pharmacother. 2018, 103, 699–707. [Google Scholar] [CrossRef] [PubMed]
- Celińska-Janowicz, K.; Zaręba, I.; Lazarek, U.; Teul, J.; Tomczyk, M.; Pałka, J.; Miltyk, W. Constituents of Propolis: Chrysin, Caffeic Acid, p-Coumaric Acid, and Ferulic Acid Induce PRODH/POX-Dependent Apoptosis in Human Tongue Squamous Cell Carcinoma Cell (CAL-27). Front. Pharmacol. 2018, 9, 336. [Google Scholar] [CrossRef] [PubMed]
- Gaman, L.; Dragos, D.; Vlad, A.; Robu, G.C.; Radoi, M.P.; Stroica, L.; Badea, M.; Gilca, M. Phytoceuticals in Acute Pancreatitis: Targeting the Balance between Apoptosis and Necrosis. Evid. Based Complement. Altern. Med. 2018, 2018, 5264592. [Google Scholar] [CrossRef] [PubMed]
- Yeung, A.W.K.; El-Demerdash, A.; Berindan-Neagoe, I.; Atanasov, A.G.; Ho, Y.S. Molecular Responses of Cancers by Natural Products: Modifications of Autophagy Revealed by Literature Analysis. Crit. Rev. Oncog. 2018, 23, 347–370. [Google Scholar] [CrossRef] [PubMed]
- Zhang, C.; Hao, Y.; Wu, L.; Dong, X.; Jiang, N.; Cong, B.; Liu, J.; Zhang, W.; Tang, D.; De Perrot, M.; et al. Curcumin induces apoptosis and inhibits angiogenesis in murine malignant mesothelioma. Int. J. Oncol. 2018, in press. [Google Scholar] [CrossRef] [PubMed]
- Chung, S.S.; Dutta, P.; Austin, D.; Wang, P.; Awad, A.; Vadgama, J.V. Combination of resveratrol and 5-flurouracil enhanced anti-telomerase activity and apoptosis by inhibiting STAT3 and Akt signaling pathways in human colorectal cancer cells. Oncotarget 2018, 9, 32943–32957. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Zhou, P.; Qin, S.; Xu, D.; Liu, Y.; Fu, W.; Ruan, B.; Zhang, L.; Zhang, Y.; Wang, X.; et al. The Curcumin Analogs 2-Pyridyl Cyclohexanone Induce Apoptosis via Inhibition of the JAK2-STAT3 Pathway in Human Esophageal Squamous Cell Carcinoma Cells. Front. Pharmacol. 2018, 9, 820. [Google Scholar] [CrossRef] [PubMed]
- Baek, S.H.; Ko, J.H.; Lee, H.; Jung, J.; Kong, M.; Lee, J.W.; Lee, J.; Chinnathambi, A.; Zayed, M.E.; Alharbi, S.A.; et al. Resveratrol inhibits STAT3 signaling pathway through the induction of SOCS-1: Role in apoptosis induction radiosensitization in head neck tumor cells. Phytomedicine 2016, 23, 566–577. [Google Scholar] [CrossRef] [PubMed]
- Masuelli, L.; Benvenuto, M.; Di Stefano, E.; Mattera, R.; Fantini, M.; De Feudis, G.; De Smaele, E.; Tresoldi, I.; Giganti, M.G.; Modesti, A.; et al. Curcumin blocks autophagy and activates apoptosis of malignant mesothelioma cell lines and increases the survival of mice intraperitoneally transplanted with a malignant mesothelioma cell line. Oncotarget 2017, 8, 34405–34422. [Google Scholar] [CrossRef] [PubMed]
- Wu, X.P.; Xiong, M.; Xu, C.S.; Duan, L.N.; Dong, Y.Q.; Luo, Y.; Niu, T.H.; Lu, C.R. Resveratrol induces apoptosis of human chronic myelogenous leukemia cells in vitro through p38 and JNK-regulated H2AX phosphorylation. Acta Pharmacol. Sin. 2015, 36, 353–361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zheng, R.; Deng, Q.; Liu, Y.; Zhao, P. Curcumin Inhibits Gastric Carcinoma Cell Growth and Induces Apoptosis by Suppressing the Wnt/β-Catenin Signaling Pathway. Med. Sci. Monit. 2017, 23, 163–171. [Google Scholar] [CrossRef] [PubMed]
- Zou, Y.; Yang, J.; Jiang, D. Resveratrol inhibits canonical Wnt signaling in human MG-63 osteosarcoma cells. Mol. Med. Rep. 2015, 12, 7221–7226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Divya, C.S.; Pillai, M.R. Antitumor action of curcumin in human papillomavirus associated cells involves downregulation of viral oncogenes, prevention of NFkB and AP-1 translocation, and modulation of apoptosis. Mol. Carcinog. 2006, 45, 320–332. [Google Scholar] [CrossRef] [PubMed]
- Elshaer, M.; Chen, Y.; Wang, X.J.; Tang, X. Resveratrol: An overview of its anti-cancer mechanisms. Life Sci. 2018, 207, 340–349. [Google Scholar] [CrossRef] [PubMed]
- Shirley, S.; Micheau, O. Targeting c-FLIP in cancer. Cancer Lett. 2013, 332, 141–150. [Google Scholar] [CrossRef] [PubMed]
- Wright, C.; Iyer, A.K.V.; Yakisich, J.S.; Azad, N. Anti-Tumorigenic Effects of Resveratrol in Lung Cancer Cells through Modulation of c-FLIP. Curr. Cancer Drug Targets 2017, 17, 669–680. [Google Scholar] [CrossRef] [PubMed]
- Chin, H.K.; Horng, C.T.; Liu, Y.S.; Lu, C.C.; Su, C.Y.; Chen, P.S.; Chiu, H.Y.; Tsai, F.J.; Shieh, P.C.; Yang, J.S. Kaempferol inhibits angiogenic ability by targeting VEGF receptor-2 and downregulating the PI3K/AKT, MEK and ERK pathways in VEGF-stimulated human umbilical vein endothelial cells. Oncol. Rep. 2018, 39, 2351–2357. [Google Scholar] [CrossRef] [PubMed]
- Semaan, J.; Pinon, A.; Rioux, B.; Hassan, L.; Limami, Y.; Pouget, C.; Fagnère, C.; Sol, V.; Diab-Assaf, M.; Simon, A.; et al. Resistance to 3-HTMC-Induced Apoptosis Through Activation of PI3K/Akt, MEK/ERK, and p38/COX-2/PGE(2) Pathways in Human HT-29 and HCT116 Colorectal Cancer Cells. J. Cell Biochem. 2016, 117, 2875–2885. [Google Scholar] [CrossRef] [PubMed]
- Wani, Z.A.; Guru, S.K.; Rao, A.V.; Sharma, S.; Mahajan, G.; Behl, A.; Kumar, A.; Sharma, P.R.; Kamal, A.; Bhushan, S.; et al. A novel quinazolinone chalcone derivative induces mitochondrial dependent apoptosis and inhibits PI3K/Akt/mTOR signaling pathway in human colon cancer HCT-116 cells. Food Chem. Toxicol. 2016, 87, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Ren, K.W.; Li, Y.H.; Wu, G.; Ren, J.Z.; Lu, H.B.; Li, Z.M.; Han, X.W. Quercetin nanoparticles display antitumor activity via proliferation inhibition and apoptosis induction in liver cancer cells. Int. J. Oncol. 2017, 50, 1299–1311. [Google Scholar] [CrossRef] [PubMed]
- Xu, Y.; Gao, C.C.; Pan, Z.G.; Zhou, C.W. Irigenin sensitizes TRAIL-induced apoptosis via enhancing pro-apoptotic molecules in gastric cancer cells. Biochem. Biophys. Res. Commun. 2018, 496, 998–1005. [Google Scholar] [CrossRef] [PubMed]
- Jaudan, A.; Sharma, S.; Malek, S.N.A.; Dixit, A. Induction of apoptosis by pinostrobin in human cervical cancer cells: Possible mechanism of action. PLoS ONE 2018, 13, e0191523. [Google Scholar] [CrossRef] [PubMed]
- Kang, C.H.; Molagoda, I.M.N.; Choi, Y.H.; Park, C.; Moon, D.O.; Kim, G.Y. Apigenin promotes TRAIL-mediated apoptosis regardless of ROS generation. Food Chem. Toxicol. 2018, 111, 623–630. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Tian, B.; Wang, Y.; Ding, H. Kaempferol Sensitizes Human Ovarian Cancer Cells-OVCAR-3 and SKOV-3 to Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL)-Induced Apoptosis via JNK/ERK-CHOP Pathway and Up-Regulation of Death Receptors 4 and 5. Med. Sci. Monit. 2017, 23, 5096–5105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Gump, J.M.; Thorburn, A. Autophagy and apoptosis: What is the connection? Trends Cell Biol. 2011, 21, 387–392. [Google Scholar] [CrossRef] [PubMed]
- Noguchi, M.; Hirata, N.; Suizu, F. The links between AKT and two intracellular proteolytic cascades: Ubiquitination and autophagy. Biochim. Biophys. Acta 2014, 1846, 342–352. [Google Scholar] [CrossRef] [PubMed]
- Xie, L.L.; Shi, F.; Tan, Z.; Li, Y.; Bode, A.M.; Cao, Y. Mitochondrial network structure homeostasis and cell death. Cancer Sci. 2018, in press. [Google Scholar] [CrossRef] [PubMed]
- Fan, Y.J.; Zong, W.X. The cellular decision between apoptosis and autophagy. Chin. J. Cancer 2013, 32, 121–129. [Google Scholar] [PubMed]
- Gordy, C.; He, Y.W. The crosstalk between autophagy and apoptosis: Where does this lead? Protein Cell 2012, 3, 17–27. [Google Scholar] [CrossRef] [PubMed]
- Thorburn, A. Apoptosis and autophagy: Regulatory connections between two supposedly different processes. Apoptosis 2008, 13, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Zhou, F.; Yang, Y.; Xing, D. Bcl-2 and Bcl-xL play important roles in the crosstalk between autophagy and apoptosis. FEBS J. 2011, 278, 403–413. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Yao, S. JNK-Bcl-2/Bcl-xL-Bax/Bak Pathway Mediates the Crosstalk between Matrine-Induced Autophagy and Apoptosis via Interplay with Beclin 1. Int. J. Mol. Sci. 2015, 16, 25744–25758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Webster, B.R.; Scott, I.; Traba, J.; Han, K.; Sack, M.N. Regulation of autophagy and mitophagy by nutrient availability and acetylation. Biochim. Biophys. Acta 2014, 1841, 525–534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hosp, F.; Lassowskat, I.; Santoro, V.; De Vleesschauwer, D.; Fliegner, D.; Redestig, H.; Mann, M.; Christian, S.; Hannah, M.A.; Finkemeier, I. Lysine acetylation in mitochondria: From inventory to function. Mitochondrion 2017, 33, 58–71. [Google Scholar] [CrossRef] [PubMed]
- Jiang, K.; Wang, W.; Jin, X.; Wang, Z.; Ji, Z.; Meng, G. Silibinin, a natural flavonoid, induces autophagy via ROS-dependent mitochondrial dysfunction and loss of ATP involving BNIP3 in human MCF7 breast cancer cells. Oncol. Rep. 2015, 33, 2711–2718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Levy, J.M.M.; Towers, C.G.; Thorburn, A. Targeting autophagy in cancer. Nat. Rev. Cancer 2017, 17, 528–542. [Google Scholar] [CrossRef] [PubMed]
- Kaczanowski, S. Apoptosis: its origin, history, maintenance and the medical implications for cancer and aging. Phys. Biol. 2016, 13, 031001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guo, J.Y.; Chen, H.Y.; Mathew, R.; Fan, J.; Strohecker, A.M.; Karsli-Uzunbas, G.; Kamphorst, J.J.; Chen, G.; Lemons, J.M.; Karantza, V.; et al. Activated Ras requires autophagy to maintain oxidative metabolism and tumorigenesis. Genes Dev. 2011, 25, 460–470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guo, J.Y.; Xia, B.; White, E. Autophagy-mediated tumor promotion. Cell 2013, 155, 1216–1219. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Fan, D.; Ru, B.; Cheng, K.W.; Hu, S.; Zhang, J.; Li, E.T.; Wang, M. 6-C-(E-phenylethenyl)naringenin induces cell growth inhibition and cytoprotective autophagy in colon cancer cells. Eur. J. Cancer 2016, 68, 38–50. [Google Scholar] [CrossRef] [PubMed]
- Yuan, L.; Wei, S.; Wang, J.; Liu, X. Isoorientin induces apoptosis and autophagy simultaneously by reactive oxygen species (ROS)-related p53, PI3K/Akt, JNK, and p38 signaling pathways in HepG2 cancer cells. J. Agric. Food Chem. 2014, 62, 5390–5400. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Wang, H.; Cong, Z.; Xu, J.; Zhu, J.; Ji, X.; Ding, K. Wogonoside induces autophagy-related apoptosis in human glioblastoma cells. Oncol. Rep. 2014, 32, 1179–1187. [Google Scholar] [CrossRef] [PubMed]
- Yi, H.S.; Chang, J.Y.; Shong, M. The mitochondrial unfolded protein response and mitohormesis: A perspective on metabolic diseases. J. Mol. Endocrinol. 2018, 61, R91–R105. [Google Scholar] [CrossRef] [PubMed]
- Xue, J.; Li, R.; Zhao, X.; Ma, C.; Lv, X.; Liu, L.; Liu, P. Morusin induces paraptosis-like cell death through mitochondrial calcium overload and dysfunction in epithelial ovarian cancer. Chem. Biol. Interact. 2018, 283, 59–74. [Google Scholar] [CrossRef] [PubMed]
- Lee, D.; Kim, I.Y.; Saha, S.; Choi, K.S. Paraptosis in the anti-cancer arsenal of natural products. Pharmacol. Ther. 2016, 162, 120–133. [Google Scholar] [CrossRef] [PubMed]
- Mi, X.; Wang, C.; Sun, C.; Chen, X.; Huo, X.; Zhang, Y.; Li, G.; Xu, B.; Zhang, J.; Xie, J.; et al. Xanthohumol induces paraptosis of leukemia cells through p38 mitogen activated protein kinase signaling pathway. Oncotarget 2017, 8, 31297–31304. [Google Scholar] [CrossRef] [PubMed]
- Yumnam, S.; Hong, G.E.; Raha, S.; Saralamma, V.V.; Lee, H.J.; Lee, W.S.; Kim, E.H.; Kim, G.S. Mitochondrial Dysfunction and Ca(2+) Overload Contributes to Hesperidin Induced Paraptosis in Hepatoblastoma Cells, HepG2. J. Cell Physiol. 2016, 231, 1261–1268. [Google Scholar] [CrossRef] [PubMed]
- Lalaoui, N.; Lindqvist, L.M.; Sandow, J.J.; Ekert, P.G. The molecular relationships between apoptosis, autophagy and necroptosis. Semin. Cell Dev. Biol. 2015, 39, 63–69. [Google Scholar] [CrossRef] [PubMed]
- Coleman, V.; Sa-Nguanmoo, P.; Koenig, J.; Schulz, T.J.; Grune, T.; Klaus, S.; Kipp, A.P.; Ost, M. Partial involvement of Nrf2 in skeletal muscle mitohormesis as an adaptive response to mitochondrial uncoupling. Sci. Rep. 2018, 8, 2446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cox, C.S.; McKay, S.E.; Holmbeck, M.A.; Christian, B.E.; Scortea, A.C.; Tsay, A.J.; Newman, L.E.; Shadel, G.S. Mitohormesis in Mice via Sustained Basal Activation of Mitochondrial and Antioxidant Signaling. Cell Metab. 2018, 28, 776–786. [Google Scholar] [CrossRef] [PubMed]
- Yong, C.Q.Y.; Tang, B.L. A Mitochondrial Encoded Messenger at the Nucleus. Cells 2018, 7, 105. [Google Scholar] [CrossRef] [PubMed]
- Dandawate, P.R.; Subramaniam, D.; Jensen, R.A.; Anant, S. Targeting cancer stem cells and signaling pathways by phytochemicals: Novel approach for breast cancer therapy. Semin. Cancer Biol. 2016, 40–41, 192–208. [Google Scholar] [CrossRef] [PubMed]
- Liu, F.; Zhang, S.; Yin, M.; Guo, L.; Xu, M.; Wang, Y. Nobiletin inhibits hypoxia-induced epithelial-mesenchymal transition in renal cell carcinoma cells. J. Cell Biochem. 2018, in press. [Google Scholar] [CrossRef] [PubMed]
- Wang, N.; Zhou, F.; Guo, J.; Zhu, H.; Luo, S.; Cao, J. Euxanthone suppresses tumor growth and metastasis in colorectal cancer via targeting CIP2A/PP2A pathway. Life Sci. 2018, 209, 498–506. [Google Scholar] [CrossRef] [PubMed]
- Chang, J.H.; Cheng, C.W.; Yang, Y.C.; Chen, W.S.; Hung, W.Y.; Chow, J.M.; Chen, P.S.; Hsiao, M.; Lee, W.J.; Chien, M.H. Downregulating CD26/DPPIV by apigenin modulates the interplay between Akt and Snail/Slug signaling to restrain metastasis of lung cancer with multiple EGFR statuses. J. Exp. Clin. Cancer Res. 2018, 37, 199. [Google Scholar] [CrossRef] [PubMed]
- Shrivastava, S.; Jeengar, M.K.; Thummuri, D.; Koval, A.; Katanaev, V.L.; Marepally, S.; Naidu, V.G.M. Cardamonin, a chalcone, inhibits human triple negative breast cancer cell invasiveness by downregulation of Wnt/β-catenin signaling cascades and reversal of epithelial-mesenchymal transition. Biofactors 2017, 43, 152–169. [Google Scholar] [CrossRef] [PubMed]
- Dogan Turacli, I.; Demirtas Korkmaz, F.; Candar, T.; Ekmekci, A. Flavopiridol’s effects on metastasis in KRAS mutant lung adenocarcinoma cells. J. Cell Biochem. 2018, in press. [Google Scholar] [CrossRef] [PubMed]
- Cichon, M.A.; Radisky, D.C. ROS-induced epithelial-mesenchymal transition in mammary epithelial cells is mediated by NF-kB-dependent activation of Snail. Oncotarget 2014, 5, 2827–2838. [Google Scholar] [CrossRef] [PubMed]
- Fukawa, T.; Kajiya, H.; Ozeki, S.; Ikebe, T.; Okabe, K. Reactive oxygen species stimulates epithelial mesenchymal transition in normal human epidermal keratinocytes via TGF-beta secretion. Exp. Cell Res. 2012, 318, 1926–1932. [Google Scholar] [CrossRef] [PubMed]
- Jiang, J.; Wang, K.; Chen, Y.; Chen, H.; Nice, E.C.; Huang, C. Redox regulation in tumor cell epithelial-mesenchymal transition: Molecular basis and therapeutic strategy. Signal. Transduct. Target Ther. 2017, 2, 17036. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Cao, L.; Han, L.; Xu, Q.; Ma, Q. Superoxide dismutase promotes the epithelial-mesenchymal transition of pancreatic cancer cells via activation of the H2O2/ERK/NF-κB axis. Int. J. Oncol. 2015, 46, 2613–2620. [Google Scholar] [CrossRef] [PubMed]
- Guerra, F.; Guaragnella, N.; Arbini, A.A.; Bucci, C.; Giannattasio, S.; Moro, L. Mitochondrial Dysfunction: A Novel Potential Driver of Epithelial-to-Mesenchymal Transition in Cancer. Front. Oncol. 2017, 7, 295. [Google Scholar] [CrossRef] [PubMed]
- Gugnoni, M.; Sancisi, V.; Manzotti, G.; Gandolfi, G.; Ciarrocchi, A. Autophagy and epithelial-mesenchymal transition: An intricate interplay in cancer. Cell Death Dis. 2016, 7, e2520. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Cheng, Q.; Yin, H.; Yang, G. Regulation of autophagy and EMT by the interplay between p53 and RAS during cancer progression (Review). Int. J. Oncol. 2017, 51, 18–24. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Khor, T.O.; Shu, L.; Su, Z.Y.; Fuentes, F.; Lee, J.H.; Kong, A.N. Plants vs. Cancer: A Review on Natural Phytochemicals in Preventing and Treating Cancers and Their Druggability. Anticancer Agents Med. Chem. 2012, 12, 1281–1305. [Google Scholar] [CrossRef] [PubMed]
- Block, G.; Patterson, B.; Subar, A. Fruit, vegetables, and cancer prevention: A review of the epidemiological evidence. Nutr. Cancer 1992, 18, 1–29. [Google Scholar] [CrossRef] [PubMed]
- Mollakhalili Meybodi, N.; Mohammad Mortazavian, A.; Bahadori Monfared, A.; Sohrabvandi, S.; Aghaei Meybodi, F. Phytochemicals in Cancer Prevention: A Review of the Evidence. Iran. J. Cancer Prev. 2017, 10, e7219. [Google Scholar] [CrossRef]
- Pasko, P.; Makowska-Was, J.; Chlopicka, J.; Szlosarczyk, M.; Tyszka-Czochara, M.; Dobrowolska-Iwanek, J.; Agnieszka Galanty, A. South Siberian fruits: Their selected chemical constituents, biological activity, and traditional use in folk medicine and daily nutrition. J. Med. Plants Res. 2012, 6, 4698–4706. [Google Scholar] [CrossRef]
- Mulabagal, V.; Lang, G.A.; DeWitt, D.L.; Dalavoy, S.S.; Nair, M.G. Anthocyanin content, lipid peroxidation and cyclooxygenase enzyme inhibitory activities of sweet and sour cherries. J. Agric. Food Chem. 2009, 57, 1239–1246. [Google Scholar] [CrossRef] [PubMed]
- Bobe, G.; Wang, B.; Seeram, N.P.; Nair, M.G.; Bourquin, L.D. Dietary anthocyanin-rich tart cherry extract inhibits intestinal tumorigenesis in APC(Min) mice fedsuboptimal levels of sulindac. J. Agric. Food Chem. 2006, 54, 9322–9328. [Google Scholar] [CrossRef] [PubMed]
- Voronina, L.M.; Kravchenko, G.B.; Zagaiko, A.L.; Upyr, L.V.; Popovich, V.P. The study of some plant extracts’ effect on the exudative phase of inflammation induced by introduction of different phlogogenes. News Pharm. 2010, 1, 66–68. [Google Scholar]
- Noratto, G.; Porter, W.; Byrne, D.; Cisneros-Zevallos, L. Identifying peach and plum polyphenols with chemopreventive potential against estrogen-independent breastcancer cells. J. Agric. Food Chem. 2009, 57, 5219–5226. [Google Scholar] [CrossRef] [PubMed]
- Lea, M.A.; Ibeh, C.; des Bordes, C.; Vizzotto, M.; Cisneros-Zevallos, L.; Byrne, D.H.; Okie, W.R.; Moyer, M.P. Inhibition of growth and induction of differentiation of colon cancer cells by peach and plum phenolic compounds. Anticancer Res. 2008, 28, 2067–2076. [Google Scholar] [PubMed]
- Zagayko, A.L.; Senyuk, I.V.; Lenchyk, L.V.; Galimullin, R.M. Study of the antioxidant activity of the extract from common plum leaves. Ukr. Biopharm. J. 2014, 1, 25–28. [Google Scholar]
- Kang, S.Y.; Seeram, N.P.; Nair, M.G.; Bourquin, L.D. Tart cherry anthocyanins inhibit tumor development in Apc(Min) mice and reduce proliferation of human colon cancer cells. Cancer Lett. 2003, 194, 13–19. [Google Scholar] [CrossRef]
- Lefranc, F.; Tabanca, N.; Kiss, R. Assessing the anticancer effects associated with food products and/or nutraceuticals using in vitro and in vivo preclinical development-related pharmacological tests. Semin. Cancer Biol. 2017, 46, 14–32. [Google Scholar] [CrossRef] [PubMed]
- Bag, A.; Bag, N. Tea Polyphenols and Prevention of Epigenetic Aberrations in Cancer. J. Nat. Sci. Biol. Med. 2018, 9, 2–5. [Google Scholar] [CrossRef] [PubMed]
- Pandey, K.B.; Rizvi, S.I. Plant polyphenols as dietary antioxidants in human health and disease. Oxid. Med. Cell Longev. 2009, 2, 270–278. [Google Scholar] [CrossRef] [PubMed]
- Busch, C.; Burkard, M.; Leischner, C.; Lauer, U.M.; Frank, J.; Venturelli, S. Epigenetic activities of flavonoids in the prevention and treatment of cancer. Clin. Epigenet. 2015, 7, 64. [Google Scholar] [CrossRef] [PubMed]
- Russo, M.; Spagnuolo, C.; Tedesco, I.; Bilotto, S.; Russo, G.L. The flavonoid quercetin in disease prevention and therapy: Facts and fancies. Biochem. Pharmacol. 2012, 83, 6–15. [Google Scholar] [CrossRef] [PubMed]
- Lim, T.G.; Kwon, J.Y.; Kim, J.; Song, N.R.; Lee, K.M.; Heo, Y.S.; Lee, H.J.; Lee, K.W. Cyanidin-3-glucoside suppresses B[a]PDE-induced cyclooxygenase-2 expression by directlyinhibiting Fyn kinase activity. Biochem. Pharmacol. 2011, 82, 167–174. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.E.; Kwon, J.Y.; Seo, S.K.; Son, J.E.; Jung, S.K.; Min, S.Y.; Hwang, M.K.; Heo, Y.S.; Lee, K.W.; Lee, H.J. Cyanidin suppresses ultraviolet B-induced COX-2 expression in epidermal cells by targeting MKK4, MEK1, and Raf-1. Biochem. Pharmacol. 2010, 79, 1473–1482. [Google Scholar] [CrossRef] [PubMed]
- Dreiseitel, A.; Oosterhuis, B.; Vukman, K.V.; Schreier, P.; Oehme, A.; Locher, S.; Hajak, G.; Sand, P.G. Berry anthocyanins and anthocyanidins exhibit distinct affinities for the efflux transporters BCRP and MDR1. Br. J. Pharmacol. 2009, 158, 1942–1950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ullah, N.; Ahmad, M.; Aslam, H.; Tahir, M.A.; Aftab, M.; Bibi, N.; Ahmad, S. Green tea phytocompounds as anticancer: A review. Asian Pac. J. Trop. Dis. 2016, 6, 330–336. [Google Scholar] [CrossRef]
- Moreno, F.S.; Heidor, R.; Pogribny, I.P. Nutritional Epigenetics and the Prevention of Hepatocellular Carcinoma with Bioactive Food Constituents. Nutr. Cancer 2016, 68, 719–733. [Google Scholar] [CrossRef] [PubMed]
- Park, J.S.; Rho, H.S.; Kim, D.H.; Chang, I.S. Enzymatic preparation of kaempferol from green tea seed and its antioxidant activity. J. Agric. Food Chem. 2006, 54, 2951–2956. [Google Scholar] [CrossRef] [PubMed]
- Nöthlings, U.; Murphy, S.P.; Wilkens, L.R.; Henderson, B.E.; Kolonel, L.N. Flavonols and pancreatic cancer risk: The multiethnic cohort study. Am. J. Epidemiol. 2007, 166, 924–931. [Google Scholar] [CrossRef] [PubMed]
- Cui, Y.; Morgenstern, H.; Greenland, S.; Tashkin, D.P.; Mao, J.T.; Cai, L.; Cozen, W.; Mack, T.M.; Lu, Q.Y.; Zhang, Z.F. Dietary flavonoid intake and lung cancer—A population-based case-control study. Cancer 2008, 112, 2241–2248. [Google Scholar] [CrossRef] [PubMed]
- Gacche, R.N.; Shegokar, H.D.; Gond, D.S.; Yang, Z.; Jadhav, A.D. Evaluation of selected flavonoids as antiangiogenic, anticancer, and radical scavenging agents: An experimental and in silico analysis. Cell Biochem. Biophys. 2011, 61, 651–663. [Google Scholar] [CrossRef] [PubMed]
- Calderón-Montaño, J.M.; Burgos-Morón, E.; Pérez-Guerrero, C.; López-Lázaro, M. A review on the dietary flavonoid kaempferol. Mini Rev. Med. Chem. 2011, 11, 298–344. [Google Scholar] [CrossRef] [PubMed]
- Gupta, S.C.; Kim, J.H.; Prasad, S.; Aggarwal, B.B. Regulation of survival, proliferation, invasion, angiogenesis, and metastasis of tumor cells through modulation of inflammatory pathways by nutraceuticals. Cancer Metastasis Rev. 2010, 29, 405–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hsu, C.P.; Shih, Y.T.; Lin, B.R.; Chiu, C.F.; Lin, C.C. Inhibitory effect and mechanisms of an anthocyanins- and anthocyanidins-rich extract from purple-shoot tea on colorectal carcinoma cell proliferation. J. Agric. Food Chem. 2012, 60, 3686–3692. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Adams, L.S.; Chen, S.; Killian, C.; Ahmed, A.; Seeram, N.P. Eugenia jambolana Lam.berry extract inhibits growth and induces apoptosis of human breast cancer but not non-tumorigenic breast cells. J. Agric. Food Chem. 2009, 57, 826–831. [Google Scholar] [CrossRef] [PubMed]
- Lu, J.N.; Lee, W.S.; Nagappan, A.; Chang, S.H.; Choi, Y.H.; Kim, H.J.; Kim, G.S.; Ryu, C.H.; Shin, S.C.; Jung, J.M.; et al. Anthocyanins From the Fruit of VitiscoignetiaePulliat Potentiate the Cisplatin Activity by Inhibiting PI3K/Akt Signaling Pathways in Human Gastric Cancer Cells. J. Cancer Prev. 2015, 20, 50–56. [Google Scholar] [CrossRef] [PubMed]
- Rani, N.; Bharti, S.; Krishnamurthy, B.; Bhatia, J.; Sharma, C.; Kamal, M.A.; Ojha, S.; Arya, D.S. Pharmacological Properties and Therapeutic Potential of Naringenin: A Citrus Flavonoid of Pharmaceutical Promise. Curr. Pharm. Des. 2016, 22, 4341–4359. [Google Scholar] [CrossRef] [PubMed]
- Mir, I.A.; Tiku, A.B. Chemopreventive and therapeutic potential of “naringenin,” a flavanone present in citrus fruits. Nutr. Cancer 2015, 67, 27–42. [Google Scholar] [CrossRef] [PubMed]
- Caselli, A.; Cirri, P.; Santi, A.; Paoli, P. Morin: A Promising Natural Drug. Curr. Med. Chem. 2016, 23, 774–791. [Google Scholar] [CrossRef] [PubMed]
- Yamamoto, T.; Sakamoto, C.; Tachiwana, H.; Kumabe, M.; Matsui, T.; Yamashita, T.; Shinagawa, M.; Ochiai, K.; Saitoh, N.; Nakao, M. Endocrine therapy-resistant breast cancer model cells are inhibited by soybean glyceollin I through Eleanor non-coding RNA. Sci. Rep. 2018, 8, 15202. [Google Scholar] [CrossRef] [PubMed]
Classification | Compound | Activity | References | |
---|---|---|---|---|
Flavones | Apigenin | ↑ | Anticancer activity via the Wnt/β-catenin pathway and JAK-STAT. Induction of apoptsis in TRAIL-resistant cancers | [60,61,62,63,64] |
Luteolin | ↑ | Induction of apoptosis and autophagy in ANA-1 cells via the p38, JNK and Akt signaling pathways, inhibiting Bcl-2 and beclin-1 and activating caspase-3 and caspase-8 | ||
↓ | Proliferation of BT474 and MCF-7 breast cancer cells | |||
↑ | Apoptosis in BT474 and MCF-7 breast cancer cells | |||
↑ | Apoptosis in ACS gastric cancer | |||
Tangeritin | ↓ | Cell cycle in MCF7 and MDA-MB-468 breast cancer cells via the CYP1A1/CYP1B1-mediated metabolism | ||
Flavonols | Quercetin | ↓ | Many types of cancer via apoptosis and inhibition of cell replication | [41,65,66,67,68,69,70,71] |
Kaempferol | ↑ | Apoptosis and autophagy in human lung cancer cells A549 via upregulation of miR-340 | ||
↑ | Apoptosis in HCT116, HCT15, and SW480 colorectal cancer cells | |||
Myricetin | ↓ | Prostate cancer cell metastasis by cytotoxic activity | ||
Fisetin | ↓ | Growth and metastasis and EMT in MDA-MB-231 and BT549 breast cancer cells | ||
Galangin | ↓ | Proliferation of human kidney A498 cancer cells by the induction of apoptosis-targeted PI3K/Akt/mTOR signaling | ||
Isorhamnetin | ↓ | Growth of MCF-7 breast cancer cells | ||
Flavanones (citrus fruit flavonoids) | Hesperetin | ↑ | Apoptosis in H522 lung cancer cells | [72,73] |
Naringenin | ↓ | Prostate cancer metastasis via voltage-gated sodium channel blockage | ||
Flavanonols | Taxifolin | ↓ | Mammary carcinogenesis via the LXR-mTOR/Maf1/PTEN axis and the CYP1A1- and CYP1B1-mediated cancer | [74,75] |
Flavans (Flavanols) Green tea catechins | Epigallocatechingallate (EGCG) | ↑ | Chemoprevention in glioblastoma | [76,77,78,79,80] |
↑ | Apoptosis in chronic myeloid leukemia by Bcr/Abl-mediated p38-MAPK/JNK and JAK2/STAT3/AKT signaling | |||
Catechin, EGCG | ↓ | Lung tumor growth via the inhibition of programmed cell death-ligand1 (PD-L1) | ||
Epicatechin-3-O-gallate (ECG) | ↓ | LNCaP and PC-3 prostate cancer cell growth | ||
Epigallocatechin (EGC) | ↓ | Suppression of HPV and tumors with curcumin and resveratrol | ||
Anthocyanidins | Cyanidin | ↓ | Angiogenesis in breast cancer via the STAT3/VEGF pathway and miR124 mediated STAT3 downregulation | [81,82,83] |
Delphinidin | ↑ | Apoptosis and autophagy in HER-2 positive breast cancer MDA-MB-453 and BT474 cells | ||
↑ | Apoptosis and EMT in human osteosarcoma cell lines via the ERK2/p38MAPK pathway | |||
Isoflavonoids | Genistein | ↑ | Apoptosis in Mcl1 human laryngeal cancer cells | [84,85,86,87] |
↓ | Proliferation of EP3-expressing melanoma | |||
Alters epigenetic in MDA-MB-231 breast cancer cells | ||||
Daidzein | ↑ | Apoptosis in colon cancer cells |
© 2018 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Chirumbolo, S.; Bjørklund, G.; Lysiuk, R.; Vella, A.; Lenchyk, L.; Upyr, T. Targeting Cancer with Phytochemicals via Their Fine Tuning of the Cell Survival Signaling Pathways. Int. J. Mol. Sci. 2018, 19, 3568. https://doi.org/10.3390/ijms19113568
Chirumbolo S, Bjørklund G, Lysiuk R, Vella A, Lenchyk L, Upyr T. Targeting Cancer with Phytochemicals via Their Fine Tuning of the Cell Survival Signaling Pathways. International Journal of Molecular Sciences. 2018; 19(11):3568. https://doi.org/10.3390/ijms19113568
Chicago/Turabian StyleChirumbolo, Salvatore, Geir Bjørklund, Roman Lysiuk, Antonio Vella, Larysa Lenchyk, and Taras Upyr. 2018. "Targeting Cancer with Phytochemicals via Their Fine Tuning of the Cell Survival Signaling Pathways" International Journal of Molecular Sciences 19, no. 11: 3568. https://doi.org/10.3390/ijms19113568
APA StyleChirumbolo, S., Bjørklund, G., Lysiuk, R., Vella, A., Lenchyk, L., & Upyr, T. (2018). Targeting Cancer with Phytochemicals via Their Fine Tuning of the Cell Survival Signaling Pathways. International Journal of Molecular Sciences, 19(11), 3568. https://doi.org/10.3390/ijms19113568