Intracellular Pathways and Mechanisms of Colored Secondary Metabolites in Cancer Therapy
Abstract
:1. Introduction
2. Plant Metabolites: General Overview
3. Plant Colored Secondary Metabolites of Interest for Oncology Field
4. Signaling Molecules and Biological Pathways Targeted by Plant Secondary Metabolites in Cancer
4.1. Apoptosis Pathway
4.1.1. Caspases-Regulated Phases
4.1.2. NF-kB Pathway
4.1.3. ROS
4.2. Cell Survival Pathways
4.2.1. PI3K/Akt Pathway
4.2.2. MAPK Pathways
4.2.3. JAK/STAT Pathway
4.3. Cell Cycle Mechanism
4.4. Angiogenesis
4.5. Metabolic Alterations
5. Combination Therapies Involving Secondary Metabolites
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Conflicts of Interest
References
- ReFaey, K.; Tripathi, S.; Grewal, S.S.; Bhargav, A.G.; Quinones, D.J.; Chaichana, K.L.; Antwi, S.O.; Cooper, L.T.; Meyer, F.B.; Dronca, R.S.; et al. Cancer Mortality Rates Increasing vs. Cardiovascular Disease Mortality Decreasing in the World: Future Implications. Mayo Clin. Proc. Innov. Qual. Outcomes 2021, 5, 645–653. [Google Scholar] [CrossRef] [PubMed]
- Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef]
- Bailon-Moscoso, N.; Cevallos-Solorzano, G.; Romero-Benavides, J.C.; Orellana, M.I. Natural Compounds as Modulators of Cell Cycle Arrest: Application for Anticancer Chemotherapies. Curr. Genom. 2017, 18, 106–131. [Google Scholar] [CrossRef] [PubMed]
- Datta, N.; Chakraborty, S.; Basu, M.; Ghosh, M.K. Tumor Suppressors Having Oncogenic Functions: The Double Agents. Cells 2020, 10, 46. [Google Scholar] [CrossRef] [PubMed]
- Denais, C.; Lammerding, J. Nuclear mechanics in cancer. Adv. Exp. Med. Biol. 2014, 773, 435–470. [Google Scholar] [CrossRef]
- Artene, S.A.; Turcu-Stiolica, A.; Ciurea, M.E.; Folcuti, C.; Tataranu, L.G.; Alexandru, O.; Purcaru, O.S.; Tache, D.E.; Boldeanu, M.V.; Silosi, C.; et al. Comparative effect of immunotherapy and standard therapy in patients with high grade glioma: A meta-analysis of published clinical trials. Sci. Rep. 2018, 8, 11800. [Google Scholar] [CrossRef]
- Dricu, A.; Sergiu-Bogdan, C.; Brismar, K.; Biberfeld, P.; Andersson, L.C. A synthetic peptide derived from the human eosinophil-derived neurotoxin induces apoptosis in Kaposi’s sarcoma cells. Anticancer Res. 2004, 24, 1427–1432. [Google Scholar]
- Sun, D.; Gao, W.; Hu, H.; Zhou, S. Why 90% of clinical drug development fails and how to improve it? Acta Pharm. Sin. B 2022, 27, R713–R715. [Google Scholar] [CrossRef]
- Carapancea, M.; Alexandru, O.; Fetea, A.S.; Dragutescu, L.; Castro, J.; Georgescu, A.; Popa-Wagner, A.; Bäcklund, M.L.; Lewensohn, R.; Dricu, A. Growth factor receptors signaling in glioblastoma cells: Therapeutic implications. J. Neurooncol. 2009, 92, 137–147. [Google Scholar] [CrossRef]
- Alexandru, O.; Dragutescu, L.; Tataranu, L.; Ciubotaru, V.; Sevastre, A.; Georgescu, A.M.; Purcaru, O.; Danoiu, S.; Bäcklund, L.M.; Dricu, A. Helianthin induces antiproliferative effect on human glioblastoma cells in vitro. J. Neurooncol. 2011, 102, 9–18. [Google Scholar] [CrossRef]
- Islam, M.S. Natural Products and Disease Prevention, Relief and Treatment. Nutrients 2022, 14, 2396. [Google Scholar] [CrossRef] [PubMed]
- Mihai, D.P.; Seremet, O.C.; Nitulescu, G.; Ivopol, M.; Sevastre, A.-S.; Negres, S.; Ivopol, G.; Nitulescu, G.M.; Olaru, O.T. Evaluation of Natural Extracts in Animal Models of Pain and Inflammation for a Potential Therapy of Hemorrhoidal Disease. Sci. Pharm. 2019, 87, 14. [Google Scholar] [CrossRef]
- Sevastre, A.S.; Costachi, A.; Tataranu, L.G.; Brandusa, C.; Artene, S.A.; Stovicek, O.; Alexandru, O.; Danoiu, S.; Sfredel, V.; Dricu, A. Glioblastoma pharmacotherapy: A multifaceted perspective of conventional and emerging treatments (Review). Exp. Med. 2021, 22, 1408. [Google Scholar] [CrossRef] [PubMed]
- Alexandru, O.; Georgescu, A.; Ene, L.; Tudorica, V.; Dricu, A. Natural and synthetic dye compounds: Applications in glioblastomas therapy. Rev. Romana Neurol. 2015, 14, 70–76. [Google Scholar] [CrossRef]
- Tataranu, L.; Georgescu, A.M.; Buteică, S.A.; Siloși, I.; Mogoșanu, G.D.; Purcaru, Ș.O.; Alexandru, O.; Stovicek, P.; Brîndușa, C.; Doşa, M.; et al. Ligustrum vulgare hydroalcoholic extract induces apoptotic cell death in human primary brain tumour cells. Farmacia 2017, 65, 766–771. [Google Scholar]
- Mogosanu, G.D.; Buteica, S.A.; Purcaru, S.O.; Croitoru, O.; Georgescu, A.M.; Serban, F.; Tataranu, L.; Alexandru, O.; Dricu, A. Rationale and in vitro efficacy of Ligustrum vulgare hydroalcoholic extract for the treatment of brain tumors. Int. J. Clin. Exp. Pathol. 2016, 9, 8286–8296. [Google Scholar]
- Lichota, A.; Gwozdzinski, K. Anticancer Activity of Natural Compounds from Plant and Marine Environment. Int. J. Mol. Sci. 2018, 19, 3533. [Google Scholar] [CrossRef]
- Wink, M.; Ashour, M.L.; El-Readi, M.Z. Secondary metabolites from plants inhibiting ABC transporters and reversing resistance of cancer cells and microbes to cytotoxic and antimicrobial agents. Front. Microbiol. 2012, 3, 130. [Google Scholar] [CrossRef]
- Chandran, H.; Meena, M.; Barupal, T.; Sharma, K. Plant tissue culture as a perpetual source for production of industrially important bioactive compounds. Biotechnol. Rep. 2020, 26, e00450. [Google Scholar] [CrossRef]
- Kramar, A.; Kostic, M.M. Bacterial Secondary Metabolites as Biopigments for Textile Dyeing. Textiles 2022, 2, 13. [Google Scholar] [CrossRef]
- Wang, S.; Alseekh, S.; Fernie, A.R.; Luo, J. The Structure and Function of Major Plant Metabolite Modifications. Mol. Plant 2019, 12, 899–919. [Google Scholar] [CrossRef] [PubMed]
- Isah, T. Stress and defense responses in plant secondary metabolites production. Biol. Res. 2019, 52, 39. [Google Scholar] [CrossRef] [PubMed]
- Seca, A.M.L.; Pinto, D.C.G.A. Biological Potential and Medical Use of Secondary Metabolites. Medicines 2019, 6, 66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guerriero, G.; Berni, R.; Muñoz-Sanchez, J.A.; Apone, F.; Abdel-Salam, E.M.; Qahtan, A.A.; Alatar, A.A.; Cantini, C.; Cai, G.; Hausman, J.F.; et al. Production of Plant Secondary Metabolites: Examples, Tips and Suggestions for Biotechnologists. Genes 2018, 9, 309. [Google Scholar] [CrossRef] [PubMed]
- Mushtaq, S.; Abbasi, B.H.; Uzairm, B.; Abbasi, R. Natural products as reservoirs of novel therapeutic agents. EXCLI J. 2018, 17, 420–451. [Google Scholar] [CrossRef]
- Sevastre, A.-S.; Hodorog, A.D. Recent investigations on anticancer properties of Azo-dyes. Med. Oncol. 2021, 2, 11–25. [Google Scholar] [CrossRef]
- Jouda, J.B.; Tamokou, J.D.; Mbazoa, C.D.; Sarkar, P.; Bag, P.K.; Wandji, J. Anticancer and antibacterial secondary metabolites from the endophytic fungus Penicillium sp. CAM64 against multi-drug resistant Gram-negative bacteria. Afr. Health Sci. 2016, 16, 734–743. [Google Scholar] [CrossRef]
- Noman, E.; Al-Shaibani, M.M.; Bakhrebah, M.A.; Almoheer, R.; Al-Sahari, M.; Al-Gheethi, A.; Radin Mohamed, R.M.S.; Almulaiky, Y.Q.; Abdulaal, W.H. Potential of Anti-Cancer Activity of Secondary Metabolic Products from Marine Fungi. J. Fungi 2021, 7, 436. [Google Scholar] [CrossRef]
- Lin, H.Y.; Lin, Y.S.; Shih, S.P.; Lee, S.B.; El-Shazly, M.; Chang, K.M.; Yang, Y.S.H.; Lee, Y.L.; Lu, M.C. The Anti-Proliferative Activity of Secondary Metabolite from the Marine Streptomyces sp. against Prostate Cancer Cells. Life 2021, 11, 1414. [Google Scholar] [CrossRef]
- Majchrzak-Celińska, A.; Kleszcz, R.; Studzińska-Sroka, E.; Łukaszyk, A.; Szoszkiewicz, A.; Stelcer, E.; Jopek, K.; Rucinski, M.; Cielecka-Piontek, J.; Krajka-Kuźniak, V. Lichen Secondary Metabolites Inhibit the Wnt/β-Catenin Pathway in Glioblastoma Cells and Improve the Anticancer Effects of Temozolomide. Cells 2022, 11, 1084. [Google Scholar] [CrossRef]
- Abotaleb, M.; Samuel, S.M.; Varghese, E.; Varghese, S.; Kubatka, P.; Liskova, A.; Büsselberg, D. Flavonoids in Cancer and Apoptosis. Cancers 2018, 11, 28. [Google Scholar] [CrossRef] [PubMed]
- Wei, J.; Gou, Z.; Wen, Y.; Luo, Q.; Huang, Z. Marine compounds targeting the PI3K/Akt signaling pathway in cancer therapy. Biomed. Pharmacother. 2020, 129, 110484. [Google Scholar] [CrossRef] [PubMed]
- Dillon, M.; Lopez, A.; Lin, E.; Sales, D.; Perets, R.; Jain, P. Progress on Ras/MAPK Signaling Research and Targeting in Blood and Solid Cancers. Cancers 2021, 13, 5059. [Google Scholar] [CrossRef] [PubMed]
- Abotaleb, M.; Liskova, A.; Kubatka, P.; Büsselberg, D. Therapeutic Potential of Plant Phenolic Acids in the Treatment of Cancer. Biomolecules 2020, 10, 221. [Google Scholar] [CrossRef] [PubMed]
- Sitarek, P.; Skala, E.; Toma, M.; Wielanek, M.; Szemraj, J.; Skorski, T.; Bialas, A.J.; Sakowicz, T.; Kowalczyk, T.; Radek, M.; et al. Transformed Root Extract of Leonurus sibiricus Induces Apoptosis through Intrinsic and Extrinsic Pathways in Various Grades of Human Glioma Cells. Pathol. Oncol. Res. 2017, 23, 679–687. [Google Scholar] [CrossRef]
- Kubatka, P.; Mazurakova, A.; Samec, M.; Koklesova, L.; Zhai, K.; Al-Ishaq, R.; Kajo, K.; Biringer, K.; Vybohova, D.; Brockmueller, A.; et al. Flavonoids against non-physiologic inflammation attributed to cancer initiation, development, and progression—3PM pathways. EPMA J. 2021, 12, 559–587. [Google Scholar] [CrossRef]
- Khoo, H.E.; Azlan, A.; Tang, S.T.; Lim, S.M. Anthocyanidins and anthocyanins: Colored pigments as food, pharmaceutical ingredients, and the potential health benefits. Food Nutr. Res. 2017, 61, 1361779. [Google Scholar] [CrossRef]
- Bonesi, M.; Leporini, M.; Tenuta, M.C.; Tundis, R. The Role of Anthocyanins in Drug Discovery: Recent Developments. Curr. Drug Discov. Technol. 2020, 17, 286–298. [Google Scholar] [CrossRef]
- Diaconeasa, Z.M.; Frond, A.D.; Ştirbu, I.; Dumitrița Rugina, D.; Socaciu, C. Anthocyanins-Smart Molecules for Cancer Prevention. In Phytochemicals—Source of Antioxidants and Role in Disease Prevention; Asao, T., Asaduzzaman, M., Eds.; IntechOpen: London, UK, 2018. [Google Scholar] [CrossRef]
- De Arruda Nascimento, E.; de Lima Coutinho, L.; da Silva, C.J.; de Lima, V.L.A.G.; Dos Santos Aguiar, J. In vitro anticancer properties of anthocyanins: A systematic review. Biochim. Biophys. Acta Rev. Cancer 2022, 1877, 188748. [Google Scholar] [CrossRef]
- Tong, X.; Pelling, J.C. Targeting the PI3K/Akt/mTOR axis by apigenin for cancer prevention. Anticancer Agents Med. Chem. 2013, 13, 971–978. [Google Scholar] [CrossRef]
- Zhao, G.; Han, X.; Cheng, W.; Ni, J.; Zhang, Y.; Lin, J.; Song, Z. Apigenin inhibits proliferation and invasion, and induces apoptosis and cell cycle arrest in human melanoma cells. Oncol. Rep. 2017, 37, 2277–2285. [Google Scholar] [CrossRef] [PubMed]
- Shukla, S.; Shankar, E.; Fu, P.; MacLennan, G.T.; Gupta, S. Suppression of NF-κB and NF-κB-Regulated gene expression by apigenin through IκBα and IKK pathway in TRAMP mice. PLoS ONE 2015, 10, e0138710. [Google Scholar] [CrossRef] [Green Version]
- Pan, F.F.; Zheng, Y.B.; Shi, C.J.; Zhang, F.W.; Zhang, J.F.; Fu, W.M. H19-Wnt/β-catenin regulatory axis mediates the suppressive effects of apigenin on tumor growth in hepatocellular carcinoma. Eur J. Pharmacol. 2021, 893, 173810. [Google Scholar] [CrossRef] [PubMed]
- Memariani, Z.; Abbas, S.Q.; Ul Hassan, S.S.; Ahmadi, A.; Chabra, A. Naringin and naringenin as anticancer agents and adjuvants in cancer combination therapy: Efficacy and molecular mechanisms of action, a comprehensive narrative review. Pharmacol. Res. 2021, 171, 105264. [Google Scholar] [CrossRef]
- Alexandru, O.; Georgescu, A.M.; Ene, L.; Purcaru, S.O.; Serban, F.; Popescu, A.; Brindusa, C.; Tataranu, L.G.; Ciubotaru, V.; Dricu, A. The effect of curcumin on low-passage glioblastoma cells in vitro. J. Cancer Res. Ther. 2016, 12, 1025–1032. [Google Scholar] [CrossRef]
- Alexandru, O.; Georgescu, A.M.; Dragoi, A.; Ciurea, M.E.; Taisescu, C.I.; Tataranu, L.G.; Brindusa, C.; Boldeanu, M.V.; Purcaru, S.O.; Silosi, C.A.; et al. In vitro Antineoplastic Activity of Dye Compounds on Human Glioblastoma Cells. Rev. Chim. 2019, 70, 112–117. [Google Scholar] [CrossRef]
- Mohammadi Kian, M.; Salemi, M.; Bahadoran, M.; Haghi, A.; Dashti, N.; Mohammadi, S.; Rostami, S.; Chahardouli, B.; Babakhani, D.; Nikbakht, M. Curcumin Combined with Thalidomide Reduces Expression of STAT3 and Bcl-xL, Leading to Apoptosis in Acute Myeloid Leukemia Cell Lines. Drug Des. Dev. Ther. 2020, 14, 185–194. [Google Scholar] [CrossRef]
- Jung, E.M.; Lim, J.H.; Lee, T.J.; Park, J.W.; Choi, K.S.; Kwon, T.K. Curcumin sensitizes tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis through reactive oxygen species-mediated upregulation of death receptor 5 (DR5). Carcinogenesis 2005, 26, 1905–1913. [Google Scholar] [CrossRef]
- Siddiqui, F.A.; Prakasam, G.; Chattopadhyay, S.; Rehman, A.U.; Padder, R.A.; Ansari, M.A.; Irshad, R.; Mangalhara, K.; Bamezai, R.N.K.; Husain, M.; et al. Curcumin decreases Warburg effect in cancer cells by down-regulating pyruvate kinase M2 via mTOR-HIF1α inhibition. Sci. Rep. 2018, 8, 8323. [Google Scholar] [CrossRef]
- Mazidi, M.; Karimi, E.; Meydani, M.; Ghayour-Mobarhan, M.; Ferns, G.A. Potential effects of curcumin on peroxisome proliferator-activated receptor-γ in vitro and in vivo. World J. Methodol. 2016, 6, 112–117. [Google Scholar] [CrossRef]
- Chen, D.; Xi, Y.; Zhang, S.; Weng, L.; Dong, Z.; Chen, C.; Wu, T.; Xiao, J. Curcumin attenuates inflammation of Macrophage-derived foam cells treated with Poly-L-lactic acid degradation via PPARγ signaling pathway. J. Mater. Sci. Mater. Med. 2022, 33, 33. [Google Scholar] [CrossRef] [PubMed]
- Bernardo, A.; Plumitallo, C.; De Nuccio, C.; Visentin, S.; Minghetti, L. Curcumin promotes oligodendrocyte differentiation and their protection against TNF-α through the activation of the nuclear receptor PPAR-γ. Sci. Rep. 2021, 11, 4952. [Google Scholar] [CrossRef]
- Wang, E.; Braun, M.S.; Wink, M. Chlorophyll and Chlorophyll Derivatives Interfere with Multi-Drug Resistant Cancer Cells and Bacteria. Molecules 2019, 24, 2968. [Google Scholar] [CrossRef] [PubMed]
- Siddavaram, N.; Ramamurthi, V.P.; Veeran, V.; Mishra, R. Chlorophyllin abrogates canonical Wnt/β-catenin signaling and angiogenesis to inhibit the development of DMBA-induced hamster cheek pouch carcinomas. Cell Oncol. 2012, 35, 385–395. [Google Scholar] [CrossRef] [PubMed]
- Maoka, T. Carotenoids as natural functional pigments. J. Nat. Med. 2020, 74, 1–16. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.L.; Zhao, Y.N.; Shi, Z.Z.; Cong, D.; Bai, Y.S. Lutein Inhibits Cell Growth and Activates Apoptosis via the PI3K/AKT/mTOR Signaling Pathway in A549 Human Non-Small-Cell Lung Cancer Cells. J. Environ. Pathol. Toxicol Oncol. 2018, 37, 341–350. [Google Scholar] [CrossRef]
- Kavalappa, Y.P.; Gopal, S.S.; Ponesakki, G. Lutein inhibits breast cancer cell growth by suppressing antioxidant and cell survival signals and induces apoptosis. J. Cell Physiol. 2021, 236, 1798–1809. [Google Scholar] [CrossRef]
- Sheng, Y.N.; Luo, Y.H.; Liu, S.B.; Xu, W.T.; Zhang, Y.; Zhang, T.; Xue, H.; Zuo, W.B.; Li, Y.N.; Wang, C.Y.; et al. Zeaxanthin Induces Apoptosis via ROS-Regulated MAPK and AKT Signaling Pathway in Human Gastric Cancer Cells. OncoTargets Ther. 2020, 13, 10995–11006. [Google Scholar] [CrossRef]
- Merecz-Sadowska, A.; Sitarek, P.; Śliwiński, T.; Zajdel, R. Anti-Inflammatory Activity of Extracts and Pure Compounds Derived from Plants viavia Modulation of Signaling Pathways, Especially PI3K/AKT in Macrophages. Int. J. Mol. Sci. 2020, 21, 9605. [Google Scholar] [CrossRef]
- Sadowska-Bartosz, I.; Bartosz, G. Biological Properties and Applications of Betalains. Molecules 2021, 26, 2520. [Google Scholar] [CrossRef]
- Yi, E.Y.; Kim, Y.J. Betaine Inhibits in vitro and in vivo Angiogenesis through Suppression of the NF-kB and Akt Signaling Pathways. Int. J. Oncol. 2012, 41, 1879–1885. [Google Scholar] [CrossRef] [PubMed]
- Kar, F.; Hacioglu, C.; Kacar, S.; Sahinturk, V.; Kanbak, G. Betaine suppresses cell proliferation by increasing oxidative stress-mediated apoptosis and inflammation in DU-145 human prostate cancer cell line. Cell Stress Chaperones 2019, 24, 871–881. [Google Scholar] [CrossRef] [PubMed]
- Saber, A.; Abedimanesh, N.; Somi, M.; Khosroushahi, A.Y. Antiproliferative and Apoptotic Effects of Red Beetroot and Betanin on Human Colorectal Cancer Cell Lines. Res. Sq. 2021. [Google Scholar] [CrossRef]
- Zhao, H.; Wu, L.; Yan, G.; Chen, Y.; Zhou, M.; Wu, Y.; Li, Y. Inflammation and tumor progression: Signaling pathways and targeted intervention. Signal Transduct. Target. Ther. 2021, 6, 263. [Google Scholar] [CrossRef] [PubMed]
- Brentnall, M.; Rodriguez-Menocal, L.; De Guevara, R.L.; Cepero, E.; Boise, L.H. Caspase-9, caspase-3 and caspase-7 have distinct roles during intrinsic apoptosis. BMC Cell Biol. 2013, 14, 32. [Google Scholar] [CrossRef]
- Shawgo, M.E.; Shelton, S.N.; Robertson, J.D. Caspase-mediated Bak activation and cytochrome c release during intrinsic apoptotic cell death in Jurkat cells. J. Biol. Chem. 2008, 283, 35532–35538. [Google Scholar] [CrossRef]
- Guicciardi, M.E.; Gores, G.J. Life and death by death receptors. FASEB J. 2009, 23, 1625–1637. [Google Scholar] [CrossRef]
- Zhu, B.S.; Xing, C.G.; Lin, F.; Fan, X.Q.; Zhao, K.; Qin, Z.H. Blocking NF-κB nuclear translocation leads to p53-related autophagy activation and cell apoptosis. World J. Gastroenterol. 2011, 17, 478–487. [Google Scholar] [CrossRef]
- Gousias, K.; Theocharous, T.; Simon, M. Mechanisms of Cell Cycle Arrest and Apoptosis in Glioblastoma. Biomedicines 2022, 10, 564. [Google Scholar] [CrossRef]
- Wusirika, R.; Kumari, A.; Rahman, N.; Mandave, P. Anticancer Activities of Plant Secondary Metabolites: Rice Callus Suspension Culture as a New Paradigm. Rice Sci. 2021, 28, 13–30. [Google Scholar] [CrossRef]
- Ye, C.; Zheng, S.; Jiang, D.; Lu, J.; Huang, Z.; Liu, Z.; Zhou, H.; Zhuang, C.; Li, J. Initiation and Execution of Programmed Cell Death and Regulation of Reactive Oxygen Species in Plants. Int. J. Mol. Sci. 2021, 22, 12942. [Google Scholar] [CrossRef] [PubMed]
- Nazim, U.M.; Park, S. Luteolin sensitizes human liver cancer cells to TRAIL-induced apoptosis viavia autophagy and JNK-mediated death receptor 5 upregulation. Int. J. Oncol. 2019, 54, 665–672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ashkenazi, A.; Holland, P.; Gail Eckhardt, S. Ligand-Based Targeting of Apoptosis in Cancer: The Potential of Recombinant Human Apoptosis Ligand 2/Tumor Necrosis Factor–Related Apoptosis-Inducing Ligand (rhApo2L/TRAIL). J. Clin. Oncol. 2008, 26, 3621–3630. [Google Scholar] [CrossRef]
- Ramos, S. Effects of dietary flavonoids on apoptotic pathways related to cancer chemoprevention. J. Nutr. Biochem. 2007, 18, 427–442. [Google Scholar] [CrossRef]
- Zhang, J.Y.; Yi, T.; Liu, J.; Zhao, Z.Z.; Chen, H.B. Quercetin induces apoptosis via the mitochondrial pathway in KB and KBv200 cells. J. Agric. Food Chem. 2013, 61, 2188–2195. [Google Scholar] [CrossRef]
- Hashemzaei, M.; Delarami Far, A.; Yari, A.; Heravi, R.E.; Tabrizian, K.; Taghdisi, S.M.; Sadegh, S.E.; Tsarouhas, K.; Kouretas, D.; Tzanakakis, G.; et al. Anticancer and apoptosis-inducing effects of quercetin in vitro and in vivo. Oncol. Rep. 2017, 38, 819–828. [Google Scholar] [CrossRef] [PubMed]
- Maurya, A.K.; Vinayak, M. PI-103 and Quercetin Attenuate PI3K-AKT Signaling Pathway in T-Cell Lymphoma Exposed to Hydrogen Peroxide. PLoS ONE 2016, 11, e0160686. [Google Scholar] [CrossRef] [PubMed]
- Ullah, A.; Munir, S.; Badshah, S.L.; Khan, N.; Ghani, L.; Poulson, B.G.; Emwas, A.H.; Jaremko, M. Important Flavonoids and Their Role as a Therapeutic Agent. Molecules 2020, 25, 5243. [Google Scholar] [CrossRef] [PubMed]
- Al-Fatlawi, A.A.; Al-Fatlawi, A.A.; Irshad, M.; Zafaryab, M.; Alam Rizvi, M.M.; Ahmad, A. Rice Bran Phytic Acid Induced Apoptosis Through Regulation of Bcl-2/Bax and p53 Genes in HepG2 Human Hepatocellular Carcinoma Cells. Asian Pacific Journal of Cancer Prevention. Asian Pac. Organ. Cancer Prev. 2014, 15, 3731–3736. [Google Scholar] [CrossRef]
- Okda, T.M.; Katry, M.A.; Ragab, N.M.; Shalkami, A.S. Phytic acid potentiates oxaliplatin effects in colorectal cancer induced by 1,2-DMH: The role of miR-224 and miR-200a. Contemp. Oncol. 2021, 25, 118–124. [Google Scholar] [CrossRef]
- Singh, V.; Khurana, A.; Navik, U.; Allawadhi, P.; Bharani, K.K.; Weiskirchen, R. Apoptosis and Pharmacological Therapies for Targeting Thereof for Cancer Therapeutics. Science 2022, 4, 15. [Google Scholar] [CrossRef]
- Sikora, E.; Bielak-Żmijewska, A.; Magalska, A.; Piwocka, K.; Mosieniak, G.; Kalinowska, M.; Widlak, P.; Cymerman, I.A.; Bujnicki, J.M. Curcumin induces caspase-3-dependent apoptotic pathway but inhibits DNA fragmentation factor 40/caspase-activated DNase endonuclease in human Jurkat cells. Mol. Cancer Ther. 2006, 5, 927–934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Song, W.; Ren, Y.J.; Liu, L.L.; Zhao, Y.Y.; Li, Q.F.; Yang, H.B. Curcumin induced the cell death of immortalized human keratinocytes (HaCaT) through caspase-independent and caspase-dependent pathways. Food Funct. 2021, 12, 8669–8680. [Google Scholar] [CrossRef]
- Westman, J.; Grinstein, S.; Marques, P.E. Phagocytosis of Necrotic Debris at Sites of Injury and Inflammation. Front. Immunol. 2020, 10, 3030. [Google Scholar] [CrossRef]
- Chauhan, A.; Islam, A.U.; Prakash, H.; Singh, S. Phytochemicals targeting NF-κB signaling: Potential anti-cancer interventions. J. Pharm. Anal. 2022, 12, 394–405. [Google Scholar] [CrossRef] [PubMed]
- Verzella, D.; Pescatore, A.; Capece, D.; Vecchiotti, D.; Ursini, M.V.; Franzoso, G.; Alesse, E.; Zazzeroni, F. Life, death, and autophagy in cancer: NF-κB turns up everywhere. Cell Death Dis. 2020, 11, 210. [Google Scholar] [CrossRef]
- Yeo, H.; Lee, Y.H.; Koh, D.; Lim, Y.; Shin, S.Y. Chrysin Inhibits NF-κB-Dependent CCL5 Transcription by Targeting IκB Kinase in the Atopic Dermatitis-Like Inflammatory Microenvironment. Int. J. Mol. Sci. 2020, 21, 7348. [Google Scholar] [CrossRef]
- Kim, J.H.; Na, H.J.; Kim, C.K.; Kim, J.Y.; Ha, K.S.; Lee, H.; Chung, H.T.; Kwon, H.J.; Kwon, Y.G.; Kim, Y.M. The non-provitamin A carotenoid, lutein, inhibits NF-kappaB-dependent gene expression through redox-based regulation of the phosphatidylinositol 3-kinase/PTEN/Akt and NF-kappaB-inducing kinase pathways: Role of H2O2 in NF-kappaB activation. Free Radic. Biol. Med. 2008, 45, 885–896. [Google Scholar] [CrossRef]
- Assar, E.A.; Vidalle, M.C.; Chopra, M.; Hafizi, S. Lycopene acts through inhibition of IκB kinase to suppress NF-κB signaling in human prostate and breast cancer cells. Tumour Biol. 2016, 37, 9375–9385. [Google Scholar] [CrossRef] [PubMed]
- Palozza, P.; Serini, S.; Torsello, A.; Di Nicuolo, F.; Piccioni, E.; Ubaldi, V.; Pioli, C.; Wolf, F.I.; Calviello, G. β-Carotene Regulates NF-κB DNA-Binding Activity by a Redox Mechanism in Human Leukemia and Colon Adenocarcinoma Cells. J. Nutr. 2003, 133, 381–388. [Google Scholar] [CrossRef]
- Arcaro, A.; Guerreiro, A.S. The phosphoinositide 3-kinase pathway in human cancer: Genetic alterations and therapeutic implications. Curr. Genom. 2007, 8, 271–306. [Google Scholar] [CrossRef]
- D’Ignazio, L.; Batie, M.; Rocha, S. Hypoxia and Inflammation in Cancer, Focus on HIF and NF-κB. Biomedicines 2017, 5, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, J.; Fu, M.; Wu, J.; Fan, F.; Zhang, X.; Li, C.; Yang, H.; Wu, Y.; Yin, Y.; Hua, W. The Anti-Glioma Effect of Juglone Derivatives through ROS Generation. Front. Pharmacol. 2022, 13, 911760. [Google Scholar] [CrossRef] [PubMed]
- Tibodeau, J.D.; Isham, C.R.; Bible, K.C. Annatto constituent cis-bixin has selective antimyeloma effects mediated by oxidative stress and associated with inhibition of thioredoxin and thioredoxin reductase. Antioxid. Redox. Signal 2010, 13, 987–997. [Google Scholar] [CrossRef]
- Cai, W.; Zhang, B.; Duan, D.; Wu, J.; Fang, J. Curcumin targeting the thioredoxin system elevates oxidative stress in HeLa cells. Toxicol. Appl. Pharmacol. 2012, 262, 341–348. [Google Scholar] [CrossRef]
- Zhang, Y.; Zhang, M.; Hu, G.; Zhang, Z.; Song, R. Elevated system exposures of baicalin after combinatory oral administration of rhein and baicalin: Mainly related to breast cancer resistance protein (ABCG2), not UDP-glucuronosyltransferases. J. Ethnopharmacol. 2020, 250, 112528. [Google Scholar] [CrossRef]
- Wang, Q.; Wang, H.; Jia, Y.; Pan, H.; Ding, H. Luteolin induces apoptosis by ROS/ER stress and mitochondrial dysfunction in gliomablastoma. Cancer Chemother. Pharmacol. 2017, 79, 1031–1041. [Google Scholar] [CrossRef]
- Baird, L.; Yamamoto, M. The Molecular Mechanisms Regulating the KEAP1-NRF2 Pathway. Mol. Cell Biol. 2020, 40, e00099-20. [Google Scholar] [CrossRef]
- Ben-Dor, A.; Steiner, M.; Gheber, L.; Danilenko, M.; Dubi, N.; Linnewiel, K.; Zick, A.; Sharoni, Y.; Levy, J. Carotenoids activate the antioxidant response element transcription system. Mol. Cancer Ther. 2005, 4, 177–186. [Google Scholar] [CrossRef]
- Maioli, E.; Greci, L.; Soucek, K.; Hyzdalova, M.; Pecorelli, A.; Fortino, V.; Valacchi, G. Rottlerin Inhibits ROS Formation and Prevents NFkB Activation in MCF-7 and HT-29 Cells. J. Biomed. Biotechnol. 2009, 2009, 742936. [Google Scholar] [CrossRef]
- Drețcanu, G.; Știrbu, I.; Leoplold, N.; Cruceriu, D.; Danciu, C.; Stănilă, A.; Fărcaș, A.; Borda, I.M.; Iuhas, C.; Diaconeasa, Z. Chemical Structure, Sources and Role of Bioactive Flavonoids in Cancer Prevention: A Review. Plants 2022, 11, 1117. [Google Scholar] [CrossRef] [PubMed]
- Zhang, B.; Shi, Z.-L.; Liu, B.; Yan, X.-B.; Feng, J.; Tao, H.-M. Enhanced anticancer effect of gemcitabine by genistein in osteosarcoma: The role of Akt and nuclear factor-κB. Anti-Cancer Drugs 2010, 21, 288–296. [Google Scholar] [CrossRef] [PubMed]
- Aoki, H.; Takada, Y.; Kondo, S.; Sawaya, R.; Aggarwal, B.B.; Kondo, Y. Evidence that curcumin suppresses the growth of malignant gliomas in vitro and in vivo through induction of autophagy: Role of akt and extracellular signal-regulated kinase signaling pathways. Mol. Pharmacol. 2007, 72, 29–39. [Google Scholar] [CrossRef] [PubMed]
- Ji, Y.; Li, L.; Ma, Y.-X.; Li, W.-T.; Li, L.; Zhu, H.-Z.; Wu, M.-H.; Zhou, J.-R. Quercetin inhibits growth of hepatocellular carcinoma by apoptosis induction in part viavia autophagy stimulation in mice. J. Nutr. Biochem. 2019, 69, 108–119. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Zhang, Z.-S.; Zhang, X.-H.; Yang, S.-N.; Liu, D.; Diao, C.-R.; Wang, H.; Zheng, F.-P. Cyanidin inhibits EMT induced by oxaliplatin viavia targeting the PDK1-PI3K/Akt signaling pathway. Food Funct. 2019, 10, 592–601. [Google Scholar] [CrossRef]
- Khan, M.A.; Siddiqui, S.; Ahmad, I.; Singh, R.; Mishra, D.P.; Srivastava, A.N.; Ahmad, R. Phytochemicals from Ajwa dates pulp extract induce apoptosis in human triple-negative breast cancer by inhibiting AKT/mTOR pathway and modulating Bcl-2 family proteins. Sci. Rep. 2021, 11, 10322. [Google Scholar] [CrossRef]
- Qiang, Z.; Meng, L.; Yi, C.; Yu, L.; Chen, W.; Sha, W. Curcumin regulates the miR-21/PTEN/Akt pathway and acts in synergy with PD98059 to induce apoptosis of human gastric cancer MGC-803 cells. J. Int. Med. Res. 2019, 47, 1288–1297. [Google Scholar] [CrossRef]
- Wei, M.; Ma, R.; Huang, S.; Liao, Y.; Ding, Y.; Li, Z.; Guo, Q.; Tan, R.; Zhang, L.; Zhao, L. Oroxylin A increases the sensitivity of temozolomide on glioma cells by hypoxia-inducible factor 1α/hedgehog pathway under hypoxia. J. Cell. Physiol. 2019, 234. [Google Scholar] [CrossRef]
- Golinska, M.; Troy, H.; Chung, Y.L.; McSheehy, P.M.; Mayr, M.; Yin, X.; Ly, L.; Williams, K.J.; Airley, R.E.; Harris, A.L.; et al. Adaptation to HIF-1 deficiency by upregulation of the AMP/ATP ratio and phosphofructokinase activation in hepatomas. BMC Cancer 2011, 11, 198. [Google Scholar] [CrossRef]
- Hu, K.; Babapoor-Farrokhran, S.; Rodrigues, M.; Deshpande, M.; Puchner, B.; Kashiwabuchi, F.; Hassan, S.J.; Asnaghi, L.; Handa, J.T.; Merbs, S.; et al. Hypoxia-inducible factor 1 upregulation of both VEGF and ANGPTL4 is required to promote the angiogenic phenotype in uveal melanoma. Oncotarget 2016, 7, 7816–7828. [Google Scholar] [CrossRef]
- Kim, H.-S.; Wannatung, T.; Lee, S.; Yang, W.K.; Chung, S.H.; Lim, J.-S.; Choe, W.; Kang, I.; Kim, S.-S.; Ha, J. Quercetin Enhances Hypoxia-Mediated Apoptosis viavia Direct Inhibition of AMPK Activity in HCT116 Colon Cancer. Apoptosis 2012, 17, 938–949. [Google Scholar] [CrossRef] [PubMed]
- Huang, L.; Peng, B.; Nayak, Y.; Wang, C.; Si, F.; Liu, X.; Dou, J.; Xu, H.; Peng, G. Baicalein and Baicalin Promote Melanoma Apoptosis and Senescence via Metabolic Inhibition. Front. Cell Dev. Biol. 2020, 8, 836. [Google Scholar] [CrossRef] [PubMed]
- Wang, F.-R.; Jiang, Y.-S. Effect of Treatment with Baicalein on the Intracerebral Tumor Growth and Survival of Orthotopic Glioma Models. J. Neurooncol. 2015, 124, 5–11. [Google Scholar] [CrossRef]
- Hussain, I.; Waheed, S.; Ahmad, K.A.; Pirog, J.E.; Syed, V. Scutellaria baicalensis targets the hypoxia-inducible factor-1α and enhances cisplatin efficacy in ovarian cancer. J. Cell. Biochem. 2018, 119, 7515–7524. [Google Scholar] [CrossRef] [PubMed]
- Fang, B.; Chen, X.; Wu, M.; Kong, H.; Chu, G.; Zhou, Z.; Zhang, C.; Chen, B. Luteolin Inhibits Angiogenesis of the M2-like TAMs viavia the Downregulation of Hypoxia Inducible Factor-1α and the STAT3 Signalling Pathway under Hypoxia. Mol. Med. Rep. 2018, 18, 2914–2922. [Google Scholar] [CrossRef]
- Triantafyllou, A.; Mylonis, I.; Simos, G.; Bonanou, S.; Tsakalof, A. Flavonoids Induce HIF-1α but Impair Its Nuclear Accumulation and Activity. Free Radic. Biol. Med. 2008, 44, 657–670. [Google Scholar] [CrossRef]
- Rojas, E.A.; Corchete, L.A.; De Ramón, C.; Krzeminski, P.; Quwaider, D.; García-Sanz, R.; Martínez-López, J.; Oriol, A.; Rosiñol, L.; Bladé, J.; et al. Expression of p53 protein isoforms predicts survival in patients with multiple myeloma. Am. J. Hematol. 2022, 97, 700–710. [Google Scholar] [CrossRef]
- Cargnello, M.; Roux, P.P. Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. MicroBiol. Mol. Biol. Rev. 2011, 75, 50–83. [Google Scholar] [CrossRef]
- Zhang, Q.; Wang, J.; Duan, M.T.; Han, S.P.; Zeng, X.Y.; Wang, J.Y. NF-κB, ERK, p38 MAPK and JNK contribute to the initiation and/or maintenance of mechanical allodynia induced by tumor necrosis factor-alpha in the red nucleus. Brain Res. Bull. 2013, 99, 132–139. [Google Scholar] [CrossRef]
- Zhang, Y.; Xu, M.; Zhang, X.; Chu, F.; Zhou, T. MAPK/c-Jun signaling pathway contributes to the upregulation of the anti-apoptotic proteins Bcl-2 and Bcl-xL induced by Epstein-Barr virus-encoded BARF1 in gastric carcinoma cells. Oncol. Lett. 2018, 15, 7537–7544. [Google Scholar] [CrossRef]
- Goel, A.; Prasad, A.K.; Parmar, V.S.; Ghosh, B.; Saini, N. Apoptogenic effect of 7,8-diacetoxy-4-methylcoumarin and 7,8-diacetoxy-4-methylthiocoumarin in human lung adenocarcinoma cell line: Role of NF-kappaB, Akt, ROS and MAP kinase pathway. Chem. Biol. Interact. 2009, 179, 363–374. [Google Scholar] [CrossRef] [PubMed]
- Granado-Serrano, A.B.; Martín, M.A.; Bravo, L.; Goya, L.; Ramos, S. Quercetin induces apoptosis via caspase activation, regulation of Bcl-2, and inhibition of PI-3-kinase/Akt and ERK pathways in a human hepatoma cell line (HepG2). J. Nutr. 2006, 136, 2715–2721. [Google Scholar] [CrossRef] [Green Version]
- Lu, X.; Li, Y.; Li, X.; Aisa, H.A. Luteolin induces apoptosis in vitro through suppressing the MAPK and PI3K signaling pathways in gastric cancer. Oncol. Lett. 2017, 14, 1993–2000. [Google Scholar] [CrossRef] [PubMed]
- Hormozi, M.; Ghoreishi, S.; Baharvand, P. Astaxanthin induces apoptosis and increases activity of antioxidant enzymes in LS-180 cells. Artif. Cells Nanomed. Biotechnol. 2019, 47, 891–895. [Google Scholar] [CrossRef] [PubMed]
- Feng, X.Q.; Rong, L.W.; Wang, R.X.; Zheng, X.L.; Zhang, L.; Zhang, L.; Lin, Y.; Wang, X.; Li, Z.P. Luteolin and sorafenib combination kills human hepatocellular carcinoma cells through apoptosis potentiation and JNK activation. Oncol. Lett. 2018, 16, 648–653. [Google Scholar] [CrossRef]
- Jang, C.H.; Moon, N.; Lee, J.; Kwon, M.J.; Oh, J.; Kim, J.S. Luteolin Synergistically Enhances Antitumor Activity of Oxaliplatin in Colorectal Carcinoma via AMPK Inhibition. Antioxidants 2022, 11, 626. [Google Scholar] [CrossRef]
- Li, W.; Yang, W.; Liu, Y.; Chen, S.; Chin, S.; Qi, X.; Zhao, Y.; Liu, H.; Wang, J.; Mei, X.; et al. MicroRNA-378 enhances inhibitory effect of curcumin on glioblastoma. Oncotarget 2017, 8, 73938–73946. [Google Scholar] [CrossRef]
- Kang, N.; Wang, M.M.; Wang, Y.H.; Zhang, Z.N.; Cao, H.R.; Lv, Y.H.; Yang, Y.; Fan, P.H.; Qiu, F.; Gao, X.M. Tetrahydrocurcumin induces G2/M cell cycle arrest and apoptosis involving p38 MAPK activation in human breast cancer cells. Food Chem. Toxicol. 2014, 67, 193–200. [Google Scholar] [CrossRef]
- Nocito, M.C.; De Luca, A.; Prestia, F.; Avena, P.; La Padula, D.; Zavaglia, L.; Sirianni, R.; Casaburi, I.; Puoci, F.; Chimento, A.; et al. Antitumoral Activities of Curcumin and Recent Advances to Improve Its Oral Bioavailability. Biomedicines 2021, 9, 1476. [Google Scholar] [CrossRef]
- Hsiao, P.-C.; Chang, J.-H.; Lee, W.-J.; Ku, C.-C.; Tsai, M.-Y.; Yang, S.-F.; Chien, M.-H. The Curcumin Analogue, EF-24, Triggers p38 MAPK-Mediated Apoptotic Cell Death via Inducing PP2A-Modulated ERK Deactivation in Human Acute Myeloid Leukemia Cells. Cancers 2020, 12, 2163. [Google Scholar] [CrossRef]
- Hu, X.; Li, J.; Fu, M.; Zhao, X.; Wang, W. The JAK/STAT signaling pathway: From bench to clinic. Signal Transduct. Target. Ther. 2021, 6, 402. [Google Scholar] [CrossRef] [PubMed]
- Gupta, S.C.; Phromnoi, K.; Aggarwal, B.B. Morin inhibits STAT3 tyrosine 705 phosphorylation in tumor cells through activation of protein tyrosine phosphatase SHP1. Biochem. Pharmacol. 2013, 85, 898–912. [Google Scholar] [CrossRef] [PubMed]
- Nam, S.; Buettner, R.; Turkson, J.; Kim, D.; Cheng, J.Q.; Muehlbeyer, S.; Hippe, F.; Vatter, S.; Merz, K.H.; Eisenbrand, G.; et al. Indirubin derivatives inhibit Stat3 signaling and induce apoptosis in human cancer cells. Proc. Natl. Acad. Sci. USA 2005, 17, 5998–6003. [Google Scholar] [CrossRef] [PubMed]
- Gong, L.; Li, Y.; Nedeljkovic-Kurepa, A.; Sarkar, F.H. Inactivation of NF-κ B by genistein is mediated via Akt signaling pathway in breast cancer cells. Oncogene 2003, 22, 4702–4709. [Google Scholar] [CrossRef] [PubMed]
- Pan, H.; Zhou, W.; He, W.; Liu, X.; Ding, Q.; Ling, L.; Zha, X.; Wang, S. Genistein inhibits MDA-MB-231 triple-negative breast cancer cell growth by inhibiting NF-κB activity via the Notch-1 pathway. Int. J. Mol. Med. 2012, 30, 337–343. [Google Scholar] [CrossRef]
- Zhang, M.; Ikeda, K.; Xu, J.W.; Yamori, Y.; Gao, X.M.; Zhang, B.L. Genistein suppresses adipogenesis of 3T3-L1 cells via multiple signal pathways. Phytother. Res. 2009, 23, 713–718. [Google Scholar] [CrossRef]
- Martín-Cordero, C.; López-Lázaro, M.; Gálvez, M.; Ayuso, M.J. Curcumin as a DNA topoisomerase II poison. J. Enzym. Inhib. Med. Chem. 2003, 18, 505–509. [Google Scholar] [CrossRef]
- Zhou, N.; Yan, Y.; Li, W.; Wang, Y.; Zheng, L.; Han, S.; Yan, Y.; Li, Y. Genistein inhibition of topoisomerase IIα expression participated by Sp1 and Sp3 in HeLa cell. Int. J. Mol. Sci. 2009, 10, 3255–3268. [Google Scholar] [CrossRef]
- Gordon, O.N.; Luis, P.B.; Ashley, R.E.; Osheroff, N.; Schneider, C. Oxidative transformation of demethoxy- and bisdemethoxycurcumin: Products, mechanism of formation, and poisoning of human topoisomerase IIα. Chem. Res. Toxicol. 2015, 28, 989–996. [Google Scholar] [CrossRef]
- Ziyad, S.; Iruela-Arispe, M.L. Molecular mechanisms of tumor angiogenesis. Genes Cancer 2011, 2, 1085–1096. [Google Scholar] [CrossRef]
- Sevastre, A.S.; Buzatu, I.M.; Baloi, C.; Oprita, A.; Dragoi, A.; Tataranu, L.G.; Alexandru, O.; Tudorache, S.; Dricu, A. ELTD1-An Emerging Silent Actor in Cancer Drama Play. Int. J. Mol. Sci. 2021, 22, 5151. [Google Scholar] [CrossRef] [PubMed]
- Oprita, A.; Baloi, S.C.; Staicu, G.A.; Alexandru, O.; Tache, D.E.; Danoiu, S.; Micu, E.S.; Sevastre, A.S. Updated Insights on EGFR Signaling Pathways in Glioma. Int. J. Mol. Sci. 2021, 22, 587. [Google Scholar] [CrossRef] [PubMed]
- Popescu, A.M.; Alexandru, O.; Brindusa, C.; Purcaru, S.O.; Tache, D.E.; Tataranu, L.G.; Taisescu, C.; Dricu, A. Targeting the VEGF and PDGF signaling pathway in glioblastoma treatment. Int. J. Clin. Exp. Pathol. 2015, 8, 7825–7837. [Google Scholar] [PubMed]
- Salehi, B.; Sharifi-Rad, J.; Cappellini, F.; Reiner, Ž.; Zorzan, D.; Imran, M.; Sener, B.; Kilic, M.; El-Shazly, M.; Fahmy, N.M.; et al. The Therapeutic Potential of Anthocyanins: Current Approaches Based on Their Molecular Mechanism of Action. Front. Pharmacol. 2020, 11, 1300. [Google Scholar] [CrossRef] [PubMed]
- Medda, R.; Lyros, O.; Schmidt, J.L.; Jovanovic, N.; Nie, L.; Link, B.J.; Otterson, M.F.; Stoner, G.D.; Shaker, R.; Rafiee, P. Anti inflammatory and anti angiogenic effect of black raspberry extract on human esophageal and intestinal microvascular endothelial cells. Microvasc. Res. 2015, 97, 167–180. [Google Scholar] [CrossRef] [PubMed]
- Bagchi, D.; Sen, C.K.; Bagchi, M.; Atalay, M. Anti-angiogenic, antioxidant, and anti-carcinogenic properties of a novel anthocyanin-rich berry extract formula. Biochemistry 2004, 69, 75–80. [Google Scholar] [CrossRef]
- Choi, J.; Lee, D.H.; Jang, H.; Park, S.Y.; Seol, J.W. Naringenin exerts anticancer effects by inducing tumor cell death and inhibiting angiogenesis in malignant melanoma. Int. J. Med. Sci. 2020, 17, 3049–3057. [Google Scholar] [CrossRef]
- Kim, M.H.; Jeong, Y.J.; Cho, H.J.; Hoe, H.S.; Park, K.K.; Park, Y.Y.; Choi, Y.H.; Kim, C.H.; Chang, H.W.; Park, Y.J.; et al. inhibits angiogenesis through the suppression of HIF-1α and VEGF expression in A549 lung cancer cells. Oncol. Rep. 2017, 37, 777–784. [Google Scholar] [CrossRef]
- Saddiq, A.A.; El-Far, A.H.; Mohamed Abdullah, S.A.; Godugu, K.; Almaghrabi, O.A.; Mousa, S.A. Curcumin, thymoquinone, and 3, 3′-diindolylmethane combinations attenuate lung and liver cancers progression. Front. Pharmacol. 2022, 13, 936996. [Google Scholar] [CrossRef]
- Wang, T.Y.; Chen, J.X. Effects of Curcumin on Vessel Formation Insight into the Pro- and Antiangiogenesis of Curcumin. Evid.-Based Complement. Alternat. Med. 2019, 2019, 1390795. [Google Scholar] [CrossRef]
- Liu, J.; Zhu, P.; Song, P.; Xiong, W.; Chen, H.; Peng, W.; Wang, S.; Li, S.; Fu, Z.; Wang, Y.; et al. Pretreatment of Adipose Derived Stem Cells with Curcumin Facilitates Myocardial Recovery via Antiapoptosis and Angiogenesis. Stem Cells Int. 2015, 2015, 638153. [Google Scholar] [CrossRef]
- Zang, M.; Hu, L.; Zhang, B.; Zhu, Z.; Li, J.; Zhu, Z.; Yan, M.; Liu, B. Luteolin suppresses angiogenesis and vasculogenic mimicry formation through inhibiting Notch1-VEGF signaling in gastric cancer. Biochem. Biophys. Res. Commun. 2017, 490, 913–919. [Google Scholar] [CrossRef]
- Chiu, W.T.; Shen, S.C.; Chow, J.M.; Lin, C.W.; Shia, L.T.; Chen, Y.C. Contribution of reactive oxygen species to migration/invasion of human glioblastoma cells U87 via ERK-dependent COX-2/PGE(2) activation. NeuroBiol. Dis. 2010, 37, 118–129. [Google Scholar] [CrossRef] [PubMed]
- Song, W.; Zhao, X.; Xu, J.; Zhang, H. Quercetin inhibits angiogenesis-mediated human retinoblastoma growth by targeting vascular endothelial growth factor receptor. Oncol. Lett. 2017, 14, 3343–3348. [Google Scholar] [CrossRef]
- Lupo, G.; Cambria, M.T.; Olivieri, M.; Rocco, C.; Caporarello, N.; Longo, A.; Zanghì, G.; Salmeri, M.; Foti, M.C.; Anfuso, C.D. Anti-angiogenic effect of quercetin and its 8-methyl pentamethyl ether derivative in human microvascular endothelial cells. J. Cell. Mol. Med. 2019, 23, 6565–6577. [Google Scholar] [CrossRef]
- Lirdprapamongkol, K.; Sakurai, H.; Abdelhamed, S.; Yokoyama, S.; Maruyama, T.; Athikomkulchai, S.; Viriyaroj, A.; Awale, S.; Yagita, H.; Ruchirawat, S.; et al. A flavonoid chrysin suppresses hypoxic survival and metastatic growth of mouse breast cancer cells. Oncol. Rep. 2013, 30, 2357–2364. [Google Scholar] [CrossRef]
- Liao, Z.Y.; Liang, I.C.; Li, H.J.; Wu, C.C.; Lo, H.M.; Chang, D.C.; Hung, C.F. Chrysin Inhibits High Glucose-Induced Migration on Chorioretinal Endothelial Cells via VEGF and VEGFR Down-Regulation. Int. J. Mol. Sci. 2020, 21, 5541. [Google Scholar] [CrossRef]
- Jangid, A.K.; Solanki, R.; Patel, S.; Medicherla, K.; Pooja, D.; Kulhari, H. Improving Anticancer Activity of Chrysin using Tumor Microenvironment pH-Responsive and Self-Assembled Nanoparticles. ACS Omega 2022, 7, 15919–15928. [Google Scholar] [CrossRef] [PubMed]
- Warburg, O. On the origin of cancer cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef]
- Chen, J.Q.; Russo, J. Dysregulation of glucose transport, glycolysis, TCA cycle and glutaminolysis by oncogenes and tumor suppressors in cancer cells. Biochim. Biophys. Acta 2012, 1826, 370–384. [Google Scholar] [CrossRef]
- Krzeslak, A.; Wojcik-Krowiranda, K.; Forma, E.; Jozwiak, P.; Romanowicz, H.; Bienkiewicz, A.; Brys, M. Expression of GLUT1 and GLUT3 glucose transporters in endometrial and breast cancers. Pathol. Oncol. Res. 2012, 18, 721–728. [Google Scholar] [CrossRef] [PubMed]
- Harmon, A.W.; Patel, Y.M. Naringenin inhibits glucose uptake in MCF-7 breast cancer cells: A mechanism for impaired cellular proliferation. Breast Cancer Res. Treat. 2004, 85, 103–110. [Google Scholar] [CrossRef] [PubMed]
- Noori, S.; Rezaei Tavirani, M.; Deravi, N.; Mahboobi Rabbani, M.I.; Zarghi, A. Naringenin Enhances the Anti-Cancer Effect of Cyclophosphamide against MDA-MB-231 Breast Cancer Cells Via Targeting the STAT3 Signaling Pathway. Iran J. Pharm. Res. 2020, 19, 122–133. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Z.; Jin, G.; Ge, Y.; Guo, Z. Naringenin inhibits migration of breast cancer cells via inflammatory and apoptosis cell signaling pathways. Inflammopharmacology 2019, 27, 1021–1036. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Zhang, J.; Zhang, M.; Song, Y.; Zhang, Y.; Fan, S.; Ren, S.; Fu, L.; Zhang, N.; Hui, H.; et al. Baicalein resensitizes tamoxifen-resistant breast cancer cells by reducing aerobic glycolysis and reversing mitochondrial dysfunction via inhibition of hypoxia-inducible factor-1α. Clin. Transl. Med. 2021, 11, e577. [Google Scholar] [CrossRef] [PubMed]
- Cheng, C.S.; Tan, H.Y.; Wang, N.; Chen, L.; Meng, Z.; Chen, Z.; Feng, Y. Functional inhibition of lactate dehydrogenase suppresses pancreatic adenocarcinoma progression. Clin. Transl. Med. 2021, 11, e467. [Google Scholar] [CrossRef]
- Deiab, S.; Mazzio, E.; Messeha, S.; Mack, N.; Soliman, K.F. High-Throughput Screening to Identify Plant Derived Human LDH-A Inhibitors. Eur. J. Med. Plants 2013, 3, 603–615. [Google Scholar] [CrossRef]
- Rahnasto-Rilla, M.; Järvenpää, J.; Huovinen, M.; Schroderus, A.M.; Ihantola, E.L.; Küblbeck, J.; Khadeer, M.; Moaddel, R.; Lahtela-Kakkonen, M. Effects of galloflavin and ellagic acid on sirtuin 6 and its anti-tumorigenic activities. Biomed. Pharmacother. 2020, 131, 110701. [Google Scholar] [CrossRef]
- Tao, L.; Wei, L.; Liu, Y.; Ding, Y.; Liu, X.; Zhang, X.; Wang, X.; Yao, Y.; Lu, J.; Wang, Q.; et al. Gen-27, a newly synthesized flavonoid, inhibits glycolysis and induces cell apoptosis via suppression of hexokinase II in human breast cancer cells. Biochem. Pharmacol. 2017, 125, 12–25. [Google Scholar] [CrossRef]
- Clinicaltrials. Available online: https://clinicaltrials.gov clinical trial NCT02446795 (accessed on 10 August 2022).
- Howells, L.M.; Iwuji, C.O.O.; Irving, G.R.B.; Barber, S.; Walter, H.; Sidat, Z.; Griffin-Teall, N.; Singh, R.; Foreman, N.; Patel, S.R. Curcumin Combined with FOLFOX Chemotherapy Is Safe and Tolerable in Patients with Metastatic Colorectal Cancer in a Randomized Phase IIa Trial. J. Nutr. 2019, 149, 1133–1139. [Google Scholar] [CrossRef]
- Zhuang, E.; Uchio, E.; Lilly, M.; Zi, X.; Fruehauf, J.P. A phase II study of docetaxel plus lycopene in metastatic castrate resistant prostate cancer. Biomed. Pharmacother. 2021, 143, 112226. [Google Scholar] [CrossRef] [PubMed]
- Zhang, P.; Lai, Z.L.; Chen, H.F.; Zhang, M.; Wang, A.; Jia, T.; Sun, W.Q.; Zhu, X.M.; Chen, X.F.; Zhao, Z.; et al. Curcumin synergizes with 5-fluorouracil by impairing AMPK/ULK1-dependent autophagy, AKT activity and enhancing apoptosis in colon cancer cells with tumor growth inhibition in xenograft mice. J. Exp. Clin. Cancer Res. 2017, 36, 190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Banerjee, S.; Singh, S.K.; Chowdhury, I.; Lillard, J.W., Jr.; Singh, R. Combinatorial effect of curcumin with docetaxel modulates apoptotic and cell survival molecules in prostate cancer. Front. Biosci. 2017, 9, 235–245. [Google Scholar] [CrossRef]
- Wang, H.; Luo, Y.; Qiao, T.; Wu, Z.; Huang, Z. Luteolin sensitizes the antitumor effect of cisplatin in drug-resistant ovarian cancer via induction of apoptosis and inhibition of cell migration and invasion. J. Ovarian Res. 2018, 11, 93. [Google Scholar] [CrossRef]
- Zhang, L.; Liu, Q.; Huang, L.; Yang, F.; Liu, A.; Zhang, J. Combination of lapatinib and luteolin enhances the therapeutic efficacy of lapatinib on human breast cancer through the FOXO3a/NQO1 pathway. Biochem Biophys. Res. Commun. 2020, 531, 364–371. [Google Scholar] [CrossRef]
- Lim, W.; Song, G. Inhibitory effects of delphinidin on the proliferation of ovarian cancer cells via PI3K/AKT and ERK 1/2 MAPK signal transduction. Oncol. Lett. 2017, 14, 1810–1818. [Google Scholar] [CrossRef]
- Zhang, Y.; Zhu, X.; Huang, T.; Chen, L.; Liu, Y.; Li, Q.; Song, J.; Ma, S.; Zhang, K.; Yang, B.; et al. β-Carotene synergistically enhances the anti-tumor effect of 5-fluorouracil on esophageal squamous cell carcinoma in vivo and in vitro. Toxicol. Lett. 2016, 261, 49–58. [Google Scholar] [CrossRef]
- Kordiak, J.; Bielec, F.; Jabłonski, S.; Pastuszak-Lewandoska, D. Role of Beta-Carotene in Lung Cancer Primary Chemoprevention: A Systematic Review with Meta-Analysis and Meta-Regression. Nutrients 2022, 14, 1361. [Google Scholar] [CrossRef]
- Shehatta, N.H.; Okda, T.M.; Omran, G.A.; Abd-Alhaseeb, M.M. Baicalin; a promising chemopreventive agent, enhances the antitumor effect of 5-FU against breast cancer and inhibits tumor growth and angiogenesis in Ehrlich solid tumor. Biomed. Pharmacother. 2022, 146, 112599. [Google Scholar] [CrossRef]
- Tsvetkova, Y.; Beztsinna, N.; Baues, M.; Klein, D.; Rix, A.; Golombek, S.K.; Al Rawashdeh, W.; Gremse, F.; Barz, M.; Koynov, K.; et al. Balancing Passive and Active Targeting to Different Tumor Compartments Using Riboflavin-Functionalized Polymeric Nanocarriers. Nano Lett. 2017, 17, 4665–4674. [Google Scholar] [CrossRef]
Class | Subclass | Examples | Targets | Type of Study | References |
---|---|---|---|---|---|
Flavonoids | Flavones | Luteolin | JNK, Akt, Caspase 9, 8, 3 NF-kB, Bcl-2, Bax, Bad, SOD, Catalases, MAPK, Bak, Cyclin D1, Cdk 4, 6, Rb, Fas, FasL | in vitro tests | [73,80,81,98,116,125,126,153] |
Caspase 9, 8, 3, Bax, ROS | in vivo tests | [81,127] | |||
Chrysin | NF-kB, p21, p38, Akt, Caspase-3, TNFα, Cdk 2, 4 | in vitro tests | [88,157,158] | ||
NF-kB, p65, MAPK | in vivo tests | [157] | |||
Baicalein | Akt | in vitro tests | [97,115,166] | ||
Ras, Raf, MAPK, Bcl-2, p53, Bax, Caspase 3, 9 | in vivo tests | [97,113,114] | |||
Apigenin | Akt, PI3K, p53, | in vitro tests | [41,42] | ||
p21, NF-kB, Caspase-3 | in vivo tests | [41,43,44] | |||
Anthocyanins | Delphinidin | PI3K, Akt, mTOR, p38, NF-kB, Bax/Bcl-2 | in vitro tests | [149] | |
Cyanidin | PI3K, Akt, mTOR | in vitro tests | [106] | ||
Malvidin | p53, p38, Akt, MAPK | in vitro tests | [39,118,146] | ||
Flavonols | Quercetin | Caspase-3,9, ROS, PI3K, Akt, Bad/Bax, NF-kB, p53, Bcl-2/Bax | in vitro tests | [76,77,78,107,112,154,155,156] | |
Bcl-2, Caspase-3, Akt, MAPK | in vivo tests | [81,105] | |||
Morin | STAT3, Src, Bcl-2, Bcl-xL, | in vitro tests | [133] | ||
Myricetin | Akt/mTOR, Bcl-2 | in vitro tests | [107] | ||
Flavonones | Naringenin | ERK, MAPK, STAT3, NF-kB, p65, ROS, PI3K/AKT/mTOR | in vitro tests | [148,163,164,165] | |
NF-kB, ROS | in vivo tests | [165] | |||
Naringin | ERK, PI3K/AKT/mTOR, ROS | in vitro tests | [45] | ||
Isoflavonoids | Genistein | Akt, NF-kB, PI3K, ROS | in vitro tests | [103,135,136,137,170] | |
Chlorophylls | Chlorophyll | PI3K, Akt | in vitro tests | [54] | |
Wnt/β-catenin | in vivo tests | [55] | |||
Carotenoids | Carotene and Beta-carotene | PPARγ, p21, Nrf2, Cyclin D1, Bax/Bcl-2, NF-kB, p21, ROS | in vitro tests | [86,90,91,107] | |
Lutein | PI3K, Akt, NF-kB | in vivo tests | [89] | ||
Xanthins | Asta-, Neo-, Vola-, Anthero-, Zea-Xantin | ROS, MAPK, Akt | in vitro tests | [125] | |
Lycopene | NF-kB, Cyclin D1, p27, Caspase 8, 9, Bcl-2 | in vitro tests | [86,90,91] | ||
Betalains | Beta-cyanins | Bcl-2, BAD, NF-kB, Akt Caspase 3, 8, 9, | in vitro tests | [61,62,63,64] | |
NF-kB, Akt | in vivo tests | [62] |
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
Sevastre, A.-S.; Manea, E.V.; Popescu, O.S.; Tache, D.E.; Danoiu, S.; Sfredel, V.; Tataranu, L.G.; Dricu, A. Intracellular Pathways and Mechanisms of Colored Secondary Metabolites in Cancer Therapy. Int. J. Mol. Sci. 2022, 23, 9943. https://doi.org/10.3390/ijms23179943
Sevastre A-S, Manea EV, Popescu OS, Tache DE, Danoiu S, Sfredel V, Tataranu LG, Dricu A. Intracellular Pathways and Mechanisms of Colored Secondary Metabolites in Cancer Therapy. International Journal of Molecular Sciences. 2022; 23(17):9943. https://doi.org/10.3390/ijms23179943
Chicago/Turabian StyleSevastre, Ani-Simona, Elena Victoria Manea, Oana Stefana Popescu, Daniela Elise Tache, Suzana Danoiu, Veronica Sfredel, Ligia Gabriela Tataranu, and Anica Dricu. 2022. "Intracellular Pathways and Mechanisms of Colored Secondary Metabolites in Cancer Therapy" International Journal of Molecular Sciences 23, no. 17: 9943. https://doi.org/10.3390/ijms23179943
APA StyleSevastre, A. -S., Manea, E. V., Popescu, O. S., Tache, D. E., Danoiu, S., Sfredel, V., Tataranu, L. G., & Dricu, A. (2022). Intracellular Pathways and Mechanisms of Colored Secondary Metabolites in Cancer Therapy. International Journal of Molecular Sciences, 23(17), 9943. https://doi.org/10.3390/ijms23179943