Thinking Outside the Therapeutic Box: The Potential of Polyphenols in Preventing Chemotherapy-Induced Endothelial Dysfunction
Abstract
:1. Introduction
2. Chemotoxic-Induced Endothelial Dysfunction
3. What Are Polyphenols?
3.1. Polyphenols: The Underlying Chemistry
3.1.1. Phenolic Acids
3.1.2. Flavonoids
3.1.3. Stilbenes
3.1.4. Lignans
4. Polyphenols’ Therapeutic Mechanisms
4.1. Modulation of Oxidative Stress and eNOS
4.2. Controlling the Peptide Environment
4.3. Attenuating Cellular Senescence
5. Future Perspectives
5.1. Overcoming Bioavailability Concerns
5.2. Looking Beyond Chemotherapy
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
- International Association of Cancer Registries. GLOBOCAN. Available online: https://gco.iarc.fr/today/online-analysis-table?v=2020&mode=cancer&mode_population=continents&population=900&populations=900&key=asr&sex=0&cancer=39&type=0&statistic=5&prevalence=0&population_group=0&ages_group%5B%5D=0&ages_group%5B%5D=17&group_cancer=1&include_nmsc=0&include_nmsc_other=1 (accessed on 23 January 2025).
- Miller, K.D.; Nogueira, L.; Devasia, T.; Mariotto, A.B.; Yabroff, K.R.; Jemal, A.; Kramer, J.; Siegel, R.L. Cancer treatment and survivorship statistics, 2022. CA Cancer J. Clin. 2022, 72, 409–436. [Google Scholar] [CrossRef]
- Krzyszczyk, P.; Acevedo, A.; Davidoff, E.J.; Timmins, L.M.; Marrero-Berrios, I.; Patel, M.; White, C.; Lowe, C.; Sherba, J.J.; Hartmanshenn, C.; et al. The growing role of precision and personalized medicine for cancer treatment. Technology 2018, 6, 79–100. [Google Scholar] [CrossRef]
- Siddiqui, M.; Rajkumar, S.V. The high cost of cancer drugs and what we can do about it. Mayo Clin. Proc. 2012, 87, 935–943. [Google Scholar] [CrossRef]
- Florescu, M.; Cinteza, M.; Vinereanu, D. Chemotherapy-induced Cardiotoxicity. Maedica 2013, 8, 59–67. [Google Scholar]
- Lustberg, M.B.; Kuderer, N.M.; Desai, A.; Bergerot, C.; Lyman, G.H. Mitigating long-term and delayed adverse events associated with cancer treatment: Implications for survivorship. Nat. Rev. Clin. Oncol. 2023, 20, 527–542. [Google Scholar] [CrossRef]
- Terwoord, J.D.; Beyer, A.M.; Gutterman, D.D. Endothelial dysfunction as a complication of anti-cancer therapy. Pharmacol. Ther. 2022, 237, 108116. [Google Scholar] [CrossRef]
- Strongman, H.; Gadd, S.; Matthews, A.A.; Mansfield, K.E.; Stanway, S.; Lyon, A.R.; Dos-Santos-Silva, I.; Smeeth, L.; Bhaskaran, K. Does Cardiovascular Mortality Overtake Cancer Mortality During Cancer Survivorship?: An English Retrospective Cohort Study. JACC CardioOncol. 2022, 4, 113–123. [Google Scholar] [CrossRef]
- Zaorsky, N.G.; Churilla, T.M.; Egleston, B.L.; Fisher, S.G.; Ridge, J.A.; Horwitz, E.M.; Meyer, J.E. Causes of death among cancer patients. Ann. Oncol. 2017, 28, 400–407. [Google Scholar] [CrossRef]
- Fidler, M.M.; Reulen, R.C.; Henson, K.; Kelly, J.; Cutter, D.; Levitt, G.A.; Frobisher, C.; Winter, D.L.; Hawkins, M.M. Population-based long-term cardiac-specific mortality among 34 489 five-year survivors of childhood cancer in Great Britain. Circulation 2017, 135, 951–963. [Google Scholar] [CrossRef]
- Sturgeon, K.M.; Deng, L.; Bluethmann, S.M.; Zhou, S.; Trifiletti, D.M.; Jiang, C.; Kelly, S.P.; Zaorsky, N.G. A population-based study of cardiovascular disease mortality risk in US cancer patients. Eur. Heart J. 2019, 40, 3889–3897. [Google Scholar] [CrossRef]
- Lai, S.W.; Lin, C.L.; Liao, K.F. Association between tamoxifen use and acute myocardial infarction in women with breast cancer. Medicine 2019, 98, e13925. [Google Scholar] [CrossRef]
- FDA. Doxorubicin Prescribing Information; FDA: Silver Spring, MD, USA, 2019.
- FDA. Epirubicin Prescribing Information; FDA: Silver Spring, MD, USA, 2020.
- FDA. Blenoxane (Bleomycin) Prescribing Information; FDA: Silver Spring, MD, USA, 2010.
- FDA. Cisplatin Prescribing Information; FDA: Silver Spring, MD, USA, 2022.
- FDA. Fluorouracil Prescribing Information; FDA: Silver Spring, MD, USA, 2024.
- FDA. Etoposide Prescribing Information; FDA: Silver Spring, MD, USA, 2017.
- FDA. Docetaxel Prescribing Information; FDA: Silver Spring, MD, USA, 2020.
- Dong, H.; Yao, L.; Wang, M.; Wang, M.; Li, X.; Sun, X.; Yu, X.; Guo, J.; Li, X.; Xu, Y. Can ACEI/ARB prevent the cardiotoxicity caused by chemotherapy in early-stage breast cancer?—A meta-analysis of randomized controlled trials. Transl. Cancer Res. 2020, 9, 7034. [Google Scholar] [CrossRef]
- Gao, Y.; Wang, R.; Jiang, J.; Hu, Y.; Li, H.; Wang, Y. ACEI/ARB and beta-blocker therapies for preventing cardiotoxicity of antineoplastic agents in breast cancer: A systematic review and meta-analysis. Heart Fail. Rev. 2023, 28, 1405–1415. [Google Scholar] [CrossRef]
- Di Pietro, N.; Baldassarre, M.P.A.; Cichelli, A.; Pandolfi, A.; Formoso, G.; Pipino, C. Role of Polyphenols and Carotenoids in Endothelial Dysfunction: An Overview from Classic to Innovative Biomarkers. Oxidative Med. Cell Longev. 2020, 2020, 6381380. [Google Scholar] [CrossRef]
- Goszcz, K.; Duthie, G.G.; Stewart, D.; Leslie, S.J.; Megson, I.L. Bioactive polyphenols and cardiovascular disease: Chemical antagonists, pharmacological agents or xenobiotics that drive an adaptive response? Br. J. Pharmacol. 2017, 174, 1209–1225. [Google Scholar] [CrossRef]
- Rathod, N.B.; Elabed, N.; Punia, S.; Ozogul, F.; Kim, S.K.; Rocha, J.M. Recent Developments in Polyphenol Applications on Human Health: A Review with Current Knowledge. Plants 2023, 12, 1217. [Google Scholar] [CrossRef]
- Pardo Sanz, A.; Zamorano, J.L. ‘Cardiotoxicity’: Time to define new targets? Eur. Heart J. 2020, 41, 1730–1732. [Google Scholar] [CrossRef]
- Martín, M.; Esteva, F.J.; Alba, E.; Khandheria, B.; Pérez-Isla, L.; García-Sáenz, J.A.; Márquez, A.; Sengupta, P.; Zamorano, J. Minimizing cardiotoxicity while optimizing treatment efficacy with trastuzumab: Review and expert recommendations. Oncologist 2009, 14, 1–11. [Google Scholar] [CrossRef]
- Herrmann, J. Vascular toxic effects of cancer therapies. Nat. Rev. Cardiol. 2020, 17, 503–522. [Google Scholar] [CrossRef]
- Brunner, H.; Cockcroft, J.R.; Deanfield, J.; Donald, A.; Ferrannini, E.; Halcox, J.; Kiowski, W.; Lüscher, T.F.; Mancia, G.; Natali, A.; et al. Endothelial function and dysfunction. Part II: Association with cardiovascular risk factors and diseases. A statement by the Working Group on Endothelins and Endothelial Factors of the European Society of Hypertension. J. Hypertens. 2005, 23, 233–246. [Google Scholar] [CrossRef]
- Leite, A.R.; Borges-Canha, M.; Cardoso, R.; Neves, J.S.; Castro-Ferreira, R.; Leite-Moreira, A. Novel Biomarkers for Evaluation of Endothelial Dysfunction. Angiology 2020, 71, 397–410. [Google Scholar] [CrossRef]
- Meinardi, M.T.; Gietema, J.A.; van Veldhuisen, D.J.; van der Graaf, W.T.; de Vries, E.G.; Sleijfer, D.T. Long-term chemotherapy-related cardiovascular morbidity. Cancer Treat. Rev. 2000, 26, 429–447. [Google Scholar] [CrossRef]
- Hamilos, M.; Petousis, S.; Parthenakis, F. Interaction between platelets and endothelium: From pathophysiology to new therapeutic options. Cardiovasc. Diagn. Ther. 2018, 8, 568–580. [Google Scholar] [CrossRef]
- Wu, K.K.; Thiagarajan, P. Role of endothelium in thrombosis and hemostasis. Annu. Rev. Med. 1996, 47, 315–331. [Google Scholar] [CrossRef]
- Kharbanda, R.K.; Walton, B.; Allen, M.; Klein, N.; Hingorani, A.D.; MacAllister, R.J.; Vallance, P. Prevention of inflammation-induced endothelial dysfunction: A novel vasculo-protective action of aspirin. Circulation 2002, 105, 2600–2604. [Google Scholar] [CrossRef]
- Antoniades, C.; Demosthenous, M.; Tousoulis, D.; Antonopoulos, A.S.; Vlachopoulos, C.; Toutouza, M.; Marinou, K.; Bakogiannis, C.; Mavragani, K.; Lazaros, G.; et al. Role of asymmetrical dimethylarginine in inflammation-induced endothelial dysfunction in human atherosclerosis. Hypertension 2011, 58, 93–98. [Google Scholar] [CrossRef]
- Boutin, G.; Yuzugulen, J.; Pranjol, M.Z.I. Endothelin-based markers for endothelial dysfunction in chemotherapy-induced cardiotoxicity. J. Mol. Cell. Cardiol. Plus 2023, 6, 100053. [Google Scholar] [CrossRef]
- Al-Qaisi, M.; Kharbanda, R.K.; Mittal, T.K.; Donald, A.E. Measurement of endothelial function and its clinical utility for cardiovascular risk. Vasc. Health Risk Manag. 2008, 4, 647–652. [Google Scholar] [CrossRef]
- Celermajer, D.S.; Sorensen, K.E.; Gooch, V.M.; Spiegelhalter, D.J.; Miller, O.I.; Sullivan, I.D.; Lloyd, J.K.; Deanfield, J.E. Non-invasive detection of endothelial dysfunction in children and adults at risk of atherosclerosis. Lancet 1992, 340, 1111–1115. [Google Scholar] [CrossRef]
- Thijssen, D.H.; Black, M.A.; Pyke, K.E.; Padilla, J.; Atkinson, G.; Harris, R.A.; Parker, B.; Widlansky, M.E.; Tschakovsky, M.E.; Green, D.J. Assessment of flow-mediated dilation in humans: A methodological and physiological guideline. Am. J. Physiol. Heart Circ. Physiol. 2011, 300, H2–H12. [Google Scholar] [CrossRef]
- Clayton, Z.S.; Hutton, D.A.; Mahoney, S.A.; Seals, D.R. Anthracycline chemotherapy-mediated vascular dysfunction as a model of accelerated vascular aging. Aging Cancer 2021, 2, 45–69. [Google Scholar] [CrossRef]
- Anastasiou, M.; Oikonomou, E.; Theofilis, P.; Papamikroulis, G.A.; Gazouli, M.; Kalogeras, K.; Lygkoni, S.; Pesiridis, T.; Goliopoulou, A.; Papatheodoridi, A.; et al. Prolonged impact of anti-cancer therapy on endothelial function and arterial stiffness in breast cancer patients. Vasc. Pharmacol. 2023, 152, 107195. [Google Scholar] [CrossRef]
- Thijssen, D.H.J.; Bruno, R.M.; van Mil, A.; Holder, S.M.; Faita, F.; Greyling, A.; Zock, P.L.; Taddei, S.; Deanfield, J.E.; Luscher, T.; et al. Expert consensus and evidence-based recommendations for the assessment of flow-mediated dilation in humans. Eur. Heart J. 2019, 40, 2534–2547. [Google Scholar] [CrossRef]
- Libby, P.; Ridker, P.M.; Hansson, G.K. Inflammation in atherosclerosis: From pathophysiology to practice. J. Am. Coll. Cardiol. 2009, 54, 2129–2138. [Google Scholar] [CrossRef]
- Zhang, Q.; Malik, P.; Pandey, D.; Gupta, S.; Jagnandan, D.; Belin de Chantemele, E.; Banfi, B.; Marrero, M.B.; Rudic, R.D.; Stepp, D.W.; et al. Paradoxical activation of endothelial nitric oxide synthase by NADPH oxidase. Arter. Thromb. Vasc. Biol. 2008, 28, 1627–1633. [Google Scholar] [CrossRef]
- Räsänen, M.; Degerman, J.; Nissinen, T.A.; Miinalainen, I.; Kerkelä, R.; Siltanen, A.; Backman, J.T.; Mervaala, E.; Hulmi, J.J.; Kivelä, R.; et al. VEGF-B gene therapy inhibits doxorubicin-induced cardiotoxicity by endothelial protection. Proc. Natl. Acad. Sci. USA 2016, 113, 13144–13149. [Google Scholar] [CrossRef]
- Alp, N.J.; McAteer, M.A.; Khoo, J.; Choudhury, R.P.; Channon, K.M. Increased endothelial tetrahydrobiopterin synthesis by targeted transgenic GTP-cyclohydrolase I overexpression reduces endothelial dysfunction and atherosclerosis in ApoE-knockout mice. Arter. Thromb. Vasc. Biol. 2004, 24, 445–450. [Google Scholar] [CrossRef]
- Mao, A.; Zhang, P.; Zhang, K.; Kan, H.; He, D.; Han, X.; Wang, Z.; Tang, C.; Ma, X. Endothelial TRPV4-eNOS coupling as a vital therapy target for treatment of hypertension. Br. J. Pharmacol. 2022, 179, 2297–2312. [Google Scholar] [CrossRef]
- Gao, M.; Han, J.; Zhu, Y.; Tang, C.; Liu, L.; Xiao, W.; Ma, X. Blocking endothelial TRPV4-Nox2 interaction helps reduce ROS production and inflammation, and improves vascular function in obese mice. J. Mol. Cell Cardiol. 2021, 157, 66–76. [Google Scholar] [CrossRef]
- McQuillan, L.P.; Leung, G.K.; Marsden, P.A.; Kostyk, S.K.; Kourembanas, S. Hypoxia inhibits expression of eNOS via transcriptional and posttranscriptional mechanisms. Am. J. Physiol. 1994, 267, H1921–H1927. [Google Scholar] [CrossRef]
- Fish, J.E.; Matouk, C.C.; Yeboah, E.; Bevan, S.C.; Khan, M.; Patil, K.; Ohh, M.; Marsden, P.A. Hypoxia-inducible expression of a natural cis-antisense transcript inhibits endothelial nitric-oxide synthase. J. Biol. Chem. 2007, 282, 15652–15666. [Google Scholar] [CrossRef]
- Kanjanapan, Y.; Gilbourd, D.; Pranavan, G. Acute ischaemic stroke following cisplatin-based chemotherapy for testicular cancer. BMJ Case Rep. 2020, 13, e235005. [Google Scholar] [CrossRef]
- Dyhl-Polk, A.; Schou, M.; Vistisen, K.K.; Sillesen, A.S.; Serup-Hansen, E.; Faber, J.; Klausen, T.W.; Bojesen, S.E.; Vaage-Nilsen, M.; Nielsen, D.L. Myocardial Ischemia Induced by 5-Fluorouracil: A Prospective Electrocardiographic and Cardiac Biomarker Study. Oncologist 2021, 26, e403–e413. [Google Scholar] [CrossRef]
- Csapo, M.; Lazar, L. Chemotherapy-Induced Cardiotoxicity: Pathophysiology and Prevention. Clujul Med. 2014, 87, 135–142. [Google Scholar] [CrossRef]
- Hay, K.A.; Hanafi, L.A.; Li, D.; Gust, J.; Liles, W.C.; Wurfel, M.M.; López, J.A.; Chen, J.; Chung, D.; Harju-Baker, S.; et al. Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy. Blood 2017, 130, 2295–2306. [Google Scholar] [CrossRef]
- Sara, J.D.; Kaur, J.; Khodadadi, R.; Rehman, M.; Lobo, R.; Chakrabarti, S.; Herrmann, J.; Lerman, A.; Grothey, A. 5-fluorouracil and cardiotoxicity: A review. Ther. Adv. Med. Oncol. 2018, 10, 1758835918780140. [Google Scholar] [CrossRef]
- Altieri, P.; Murialdo, R.; Barisione, C.; Lazzarini, E.; Garibaldi, S.; Fabbi, P.; Ruggeri, C.; Borile, S.; Carbone, F.; Armirotti, A.; et al. 5-fluorouracil causes endothelial cell senescence: Potential protective role of glucagon-like peptide 1. Br. J. Pharmacol. 2017, 174, 3713–3726. [Google Scholar] [CrossRef]
- Hammond, S.T.; Baumfalk, D.R.; Parr, S.K.; Butenas, A.L.E.; Scheuermann, B.C.; Turpin, V.G.; Behnke, B.J.; Hashmi, M.H.; Ade, C.J. Impaired microvascular reactivity in patients treated with 5-fluorouracil chemotherapy regimens: Potential role of endothelial dysfunction. Int. J. Cardiol. Heart Vasc. 2023, 49, 101300. [Google Scholar] [CrossRef]
- Shanmuganathan, J.W.D.; Kragholm, K.; Tayal, B.; Polcwiartek, C.; Poulsen, L.; El-Galaly, T.C.; Fosbøl, E.L.; D’Souza, M.; Gislason, G.; Køber, L.; et al. Risk for Myocardial Infarction Following 5-Fluorouracil Treatment in Patients With Gastrointestinal Cancer: A Nationwide Registry-Based Study. JACC CardioOncol. 2021, 3, 725–733. [Google Scholar] [CrossRef]
- Fung, C.; Fossa, S.D.; Milano, M.T.; Sahasrabudhe, D.M.; Peterson, D.R.; Travis, L.B. Cardiovascular Disease Mortality After Chemotherapy or Surgery for Testicular Nonseminoma: A Population-Based Study. J. Clin. Oncol. 2015, 33, 3105–3115. [Google Scholar] [CrossRef]
- Wolf, M.B.; Baynes, J.W. The anti-cancer drug, doxorubicin, causes oxidant stress-induced endothelial dysfunction. Biochim. Biophys. Acta 2006, 1760, 267–271. [Google Scholar] [CrossRef]
- Nadar, S.; Blann, A.D.; Lip, G.Y. Antihypertensive therapy and endothelial function. Curr. Pharm. Des. 2004, 10, 3607–3614. [Google Scholar] [CrossRef]
- Nadar, S.; Blann, A.D.; Lip, G.Y. Endothelial dysfunction: Methods of assessment and application to hypertension. Curr. Pharm. Des. 2004, 10, 3591–3605. [Google Scholar] [CrossRef]
- El-Gizawy, M.M.; Hosny, E.N.; Mourad, H.H.; Abd-El Razik, A.N. Curcumin nanoparticles ameliorate hepatotoxicity and nephrotoxicity induced by cisplatin in rats. Naunyn Schmiedebergs Arch. Pharmacol. 2020, 393, 1941–1953. [Google Scholar] [CrossRef]
- Wang, S.; Prizment, A.; Thyagarajan, B.; Blaes, A. Cancer Treatment-Induced Accelerated Aging in Cancer Survivors: Biology and Assessment. Cancers 2021, 13, 427. [Google Scholar] [CrossRef]
- Ioffe, D.; Bhatia-Patel, S.C.; Gandhi, S.; Hamad, E.A.; Dotan, E. Cardiovascular Concerns, Cancer Treatment, and Biological and Chronological Aging in Cancer: JACC Family Series. JACC CardioOncol. 2024, 6, 143–158. [Google Scholar] [CrossRef]
- Ohtani, N. The roles and mechanisms of senescence-associated secretory phenotype (SASP): Can it be controlled by senolysis? Inflamm. Regen. 2022, 42, 11. [Google Scholar] [CrossRef]
- Clayton, Z.S.; Brunt, V.E.; Hutton, D.A.; Casso, A.G.; Ziemba, B.P.; Melov, S.; Campisi, J.; Seals, D.R. Tumor Necrosis Factor Alpha-Mediated Inflammation and Remodeling of the Extracellular Matrix Underlies Aortic Stiffening Induced by the Common Chemotherapeutic Agent Doxorubicin. Hypertension 2021, 77, 1581–1590. [Google Scholar] [CrossRef]
- Schafer, M.J.; Zhang, X.; Kumar, A.; Atkinson, E.J.; Zhu, Y.; Jachim, S.; Mazula, D.L.; Brown, A.K.; Berning, M.; Aversa, Z.; et al. The senescence-associated secretome as an indicator of age and medical risk. JCI Insight 2020, 5, e133668. [Google Scholar] [CrossRef]
- Shi, Y.; Inoue, S.; Shinozaki, R.; Fukue, K.; Kougo, T. Release of cytokines from human umbilical vein endothelial cells treated with platinum compounds in vitro. Jpn. J. Cancer Res. 1998, 89, 757–767. [Google Scholar] [CrossRef]
- Mantovani, A.; Bussolino, F.; Dejana, E. Cytokine regulation of endothelial cell function. FASEB J. 1992, 6, 2591–2599. [Google Scholar] [CrossRef]
- Alexander, M.R.; Moehle, C.W.; Johnson, J.L.; Yang, Z.; Lee, J.K.; Jackson, C.L.; Owens, G.K. Genetic inactivation of IL-1 signaling enhances atherosclerotic plaque instability and reduces outward vessel remodeling in advanced atherosclerosis in mice. J. Clin. Investig. 2012, 122, 70–79. [Google Scholar] [CrossRef]
- Libby, P. Interleukin-1 Beta as a Target for Atherosclerosis Therapy: Biological Basis of CANTOS and Beyond. J. Am. Coll. Cardiol. 2017, 70, 2278–2289. [Google Scholar] [CrossRef]
- Mulholland, M.; Depuydt, M.A.C.; Jakobsson, G.; Ljungcrantz, I.; Grentzmann, A.; To, F.; Bengtsson, E.; Jaensson Gyllenbäck, E.; Grönberg, C.; Rattik, S.; et al. Interleukin-1 receptor accessory protein blockade limits the development of atherosclerosis and reduces plaque inflammation. Cardiovasc. Res. 2024, 120, 581–595. [Google Scholar] [CrossRef]
- Pan, H.; Song, X.; Rajewski, A.; Wickline, S.A. Single cell sequencing unveils endothelial alterations after cisplatin treatment. Eur. Heart J. 2022, 43, ehac544.3046. [Google Scholar] [CrossRef]
- Farha, A.K.; Gan, R.Y.; Li, H.B.; Wu, D.T.; Atanasov, A.G.; Gul, K.; Zhang, J.R.; Yang, Q.Q.; Corke, H. The anticancer potential of the dietary polyphenol rutin: Current status, challenges, and perspectives. Crit. Rev. Food Sci. Nutr. 2022, 62, 832–859. [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]
- Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef]
- Cheynier, V.; Comte, G.; Davies, K.M.; Lattanzio, V.; Martens, S. Plant phenolics: Recent advances on their biosynthesis, genetics, and ecophysiology. Plant Physiol. Biochem. 2013, 72, 1–20. [Google Scholar] [CrossRef]
- Santana-Méridas, O.; González-Coloma, A.; Sánchez-Vioque, R. Agricultural residues as a source of bioactive natural products. Phytochem. Rev. 2012, 11, 447–466. [Google Scholar] [CrossRef]
- Barbaro, B.; Toietta, G.; Maggio, R.; Arciello, M.; Tarocchi, M.; Galli, A.; Balsano, C. Effects of the olive-derived polyphenol oleuropein on human health. Int. J. Mol. Sci. 2014, 15, 18508–18524. [Google Scholar] [CrossRef]
- Yang, L.; Zhang, Y.; Zhu, M.; Zhang, Q.; Wang, X.; Wang, Y.; Zhang, J.; Li, J.; Yang, L.; Liu, J.; et al. Resveratrol attenuates myocardial ischemia/reperfusion injury through up-regulation of vascular endothelial growth factor B. Free Radic. Biol. Med. 2016, 101, 1–9. [Google Scholar] [CrossRef]
- Khurana, S.; Hollingsworth, A.; Piche, M.; Venkataraman, K.; Kumar, A.; Ross, G.M.; Tai, T.C. Antiapoptotic actions of methyl gallate on neonatal rat cardiac myocytes exposed to H2O2. Oxidative Med. Cell Longev. 2014, 2014, 657512. [Google Scholar] [CrossRef]
- Aguilar-Alonso, P.; Vera-López, O.; Brambila-Colombres, E.; Segura-Badilla, O.; Avalos-López, R.; Lazcano-Hernández, M.; Navarro-Cruz, A.R. Evaluation of Oxidative Stress in Cardiomyocytes during the Aging Process in Rats Treated with Resveratrol. Oxidative Med. Cell Longev. 2018, 2018, 1390483. [Google Scholar] [CrossRef]
- Hernández, M.; Wicz, S.; Santamaría, M.H.; Corral, R.S. Curcumin exerts anti-inflammatory and vasoprotective effects through amelioration of NFAT-dependent endothelin-1 production in mice with acute Chagas cardiomyopathy. Mem. Inst. Oswaldo Cruz 2018, 113, e180171. [Google Scholar] [CrossRef]
- Palomer, X.; Capdevila-Busquets, E.; Alvarez-Guardia, D.; Barroso, E.; Pallàs, M.; Camins, A.; Davidson, M.M.; Planavila, A.; Villarroya, F.; Vázquez-Carrera, M. Resveratrol induces nuclear factor-κB activity in human cardiac cells. Int. J. Cardiol. 2013, 167, 2507–2516. [Google Scholar] [CrossRef]
- Chen, Y.W.; Chou, H.C.; Lin, S.T.; Chen, Y.H.; Chang, Y.J.; Chen, L.; Chan, H.L. Cardioprotective Effects of Quercetin in Cardiomyocyte under Ischemia/Reperfusion Injury. Evid. Based Complement. Altern. Med. 2013, 2013, 364519. [Google Scholar] [CrossRef]
- Tong, Z.; Xie, Y.; He, M.; Ma, W.; Zhou, Y.; Lai, S.; Meng, Y.; Liao, Z. VDAC1 deacetylation is involved in the protective effects of resveratrol against mitochondria-mediated apoptosis in cardiomyocytes subjected to anoxia/reoxygenation injury. Biomed. Pharmacother. 2017, 95, 77–83. [Google Scholar] [CrossRef]
- Carresi, C.; Musolino, V.; Gliozzi, M.; Maiuolo, J.; Mollace, R.; Nucera, S.; Maretta, A.; Sergi, D.; Muscoli, S.; Gratteri, S.; et al. Anti-oxidant effect of bergamot polyphenolic fraction counteracts doxorubicin-induced cardiomyopathy: Role of autophagy and c-kit(pos)CD45(neg)CD31(neg) cardiac stem cell activation. J. Mol. Cell Cardiol. 2018, 119, 10–18. [Google Scholar] [CrossRef]
- Li, T.; Chen, L.; Yu, Y.; Yang, B.; Li, P.; Tan, X.Q. Resveratrol alleviates hypoxia/reoxygenation injury-induced mitochondrial oxidative stress in cardiomyocytes. Mol. Med. Rep. 2019, 19, 2774–2780. [Google Scholar] [CrossRef] [PubMed]
- Sharma, A.; Parikh, M.; Shah, H.; Gandhi, T. Modulation of Nrf2 by quercetin in doxorubicin-treated rats. Heliyon 2020, 6, e03803. [Google Scholar] [CrossRef]
- Lin, M.Y.; Cheng, W.T.; Cheng, H.C.; Chou, W.C.; Chen, H.I.; Ou, H.C.; Tsai, K.L. Baicalin Enhances Chemosensitivity to Doxorubicin in Breast Cancer Cells via Upregulation of Oxidative Stress-Mediated Mitochondria-Dependent Apoptosis. Antioxidants 2021, 10, 1506. [Google Scholar] [CrossRef]
- Xiao, J.; Sun, G.B.; Sun, B.; Wu, Y.; He, L.; Wang, X.; Chen, R.C.; Cao, L.; Ren, X.Y.; Sun, X.B. Kaempferol protects against doxorubicin-induced cardiotoxicity in vivo and in vitro. Toxicology 2012, 292, 53–62. [Google Scholar] [CrossRef]
- Zhu, H.; Luo, P.; Fu, Y.; Wang, J.; Dai, J.; Shao, J.; Yang, X.; Chang, L.; Weng, Q.; Yang, B.; et al. Dihydromyricetin prevents cardiotoxicity and enhances anticancer activity induced by adriamycin. Oncotarget 2015, 6, 3254–3267. [Google Scholar] [CrossRef]
- Wang, S.; Dusting, G.J.; May, C.N.; Woodman, O.L. 3′,4′-Dihydroxyflavonol reduces infarct size and injury associated with myocardial ischaemia and reperfusion in sheep. Br. J. Pharmacol. 2004, 142, 443–452. [Google Scholar] [CrossRef]
- Tresserra-Rimbau, A.; Lamuela-Raventos, R.M.; Moreno, J.J. Polyphenols, food and pharma. Current knowledge and directions for future research. Biochem. Pharmacol. 2018, 156, 186–195. [Google Scholar] [CrossRef]
- Ros, E.; Martínez-González, M.A.; Estruch, R.; Salas-Salvadó, J.; Fitó, M.; Martínez, J.A.; Corella, D. Mediterranean diet and cardiovascular health: Teachings of the PREDIMED study. Adv. Nutr. 2014, 5, 330s–336s. [Google Scholar] [CrossRef]
- Khan, J.; Deb, P.K.; Priya, S.; Medina, K.D.; Devi, R.; Walode, S.G.; Rudrapal, M. Dietary Flavonoids: Cardioprotective Potential with Antioxidant Effects and Their Pharmacokinetic, Toxicological and Therapeutic Concerns. Molecules 2021, 26, 4021. [Google Scholar] [CrossRef]
- Pandey, K.B.; Rizvi, S.I. Protective effect of resveratrol on formation of membrane protein carbonyls and lipid peroxidation in erythrocytes subjected to oxidative stress. Appl. Physiol. Nutr. Metab. 2009, 34, 1093–1097. [Google Scholar] [CrossRef]
- Iglesias-Carres, L.; Mas-Capdevila, A.; Bravo, F.I.; Aragonès, G.; Arola-Arnal, A.; Muguerza, B. A comparative study on the bioavailability of phenolic compounds from organic and nonorganic red grapes. Food Chem. 2019, 299, 125092. [Google Scholar] [CrossRef] [PubMed]
- Bento-Silva, A.; Koistinen, V.M.; Mena, P.; Bronze, M.R.; Hanhineva, K.; Sahlstrøm, S.; Kitrytė, V.; Moco, S.; Aura, A.M. Factors affecting intake, metabolism and health benefits of phenolic acids: Do we understand individual variability? Eur. J. Nutr. 2020, 59, 1275–1293. [Google Scholar] [CrossRef] [PubMed]
- Mills, C.E.; Flury, A.; Marmet, C.; Poquet, L.; Rimoldi, S.F.; Sartori, C.; Rexhaj, E.; Brenner, R.; Allemann, Y.; Zimmermann, D.; et al. Mediation of coffee-induced improvements in human vascular function by chlorogenic acids and its metabolites: Two randomized, controlled, crossover intervention trials. Clin. Nutr. 2017, 36, 1520–1529. [Google Scholar] [CrossRef]
- Mubarak, A.; Bondonno, C.P.; Liu, A.H.; Considine, M.J.; Rich, L.; Mas, E.; Croft, K.D.; Hodgson, J.M. Acute effects of chlorogenic acid on nitric oxide status, endothelial function, and blood pressure in healthy volunteers: A randomized trial. J. Agric. Food Chem. 2012, 60, 9130–9136. [Google Scholar] [CrossRef] [PubMed]
- Hole, A.S.; Rud, I.; Grimmer, S.; Sigl, S.; Narvhus, J.; Sahlstrøm, S. Improved bioavailability of dietary phenolic acids in whole grain barley and oat groat following fermentation with probiotic Lactobacillus acidophilus, Lactobacillus johnsonii, and Lactobacillus reuteri. J. Agric. Food Chem. 2012, 60, 6369–6375. [Google Scholar] [CrossRef]
- Saura-Calixto, F. Dietary fiber as a carrier of dietary antioxidants: An essential physiological function. J. Agric. Food Chem. 2011, 59, 43–49. [Google Scholar] [CrossRef]
- Manach, C.; Williamson, G.; Morand, C.; Scalbert, A.; Rémésy, C. Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am. J. Clin. Nutr. 2005, 81, 230s–242s. [Google Scholar] [CrossRef]
- Tsao, R.; Deng, Z. Separation procedures for naturally occurring antioxidant phytochemicals. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2004, 812, 85–99. [Google Scholar] [CrossRef]
- Grajales Hernández, D.; Ruiz, M.; Contreras-Jácquez, V.; Mateos-Díaz, J. Biotransformation of phenolic acids from by-products using heterogeneous biocatalysts: One more step towards a circular economy. Curr. Opin. Green. Sustain. Chem. 2021, 32, 100550. [Google Scholar] [CrossRef]
- Nardini, M.; Forte, M.; Vrhovsek, U.; Mattivi, F.; Viola, R.; Scaccini, C. White wine phenolics are absorbed and extensively metabolized in humans. J. Agric. Food Chem. 2009, 57, 2711–2718. [Google Scholar] [CrossRef]
- Pérez-Jiménez, J.; Hubert, J.; Hooper, L.; Cassidy, A.; Manach, C.; Williamson, G.; Scalbert, A. Urinary metabolites as biomarkers of polyphenol intake in humans: A systematic review. Am. J. Clin. Nutr. 2010, 92, 801–809. [Google Scholar] [CrossRef] [PubMed]
- Stevenson, D.E.; Hurst, R.D. Polyphenolic phytochemicals--just antioxidants or much more? Cell Mol. Life Sci. 2007, 64, 2900–2916. [Google Scholar] [CrossRef] [PubMed]
- Shen, N.; Wang, T.; Gan, Q.; Liu, S.; Wang, L.; Jin, B. Plant flavonoids: Classification, distribution, biosynthesis, and antioxidant activity. Food Chem. 2022, 383, 132531. [Google Scholar] [CrossRef]
- Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef]
- Karak, P. Biological activities of flavonoids: An overview. Int. J. Pharm. Sci. Res. 2019, 10, 1567–1574. [Google Scholar] [CrossRef]
- Wang, R.; Li, W.; Fang, C.; Zheng, X.; Liu, C.; Huang, Q. Extraction and identification of new flavonoid compounds in dandelion Taraxacum mongolicum Hand.-Mazz. with evaluation of antioxidant activities. Sci. Rep. 2023, 13, 2166. [Google Scholar] [CrossRef]
- Shu, Z.; Yang, Y.; Yang, L.; Jiang, H.; Yu, X.; Wang, Y. Cardioprotective effects of dihydroquercetin against ischemia reperfusion injury by inhibiting oxidative stress and endoplasmic reticulum stress-induced apoptosis via the PI3K/Akt pathway. Food Funct. 2019, 10, 203–215. [Google Scholar] [CrossRef]
- Andres, A.; Zeier, M.; Eckert, C. Acute Kidney Injury Following High-Dose Vitamin C Treatment. Dtsch. Arztebl. Int. 2019, 116, 756. [Google Scholar] [CrossRef]
- Nielsen, I.L.F.; Chee, W.S.S.; Poulsen, L.; Offord-Cavin, E.; Rasmussen, S.E.; Frederiksen, H.; Enslen, M.; Barron, D.; Horcajada, M.-N.; Williamson, G. Bioavailability Is Improved by Enzymatic Modification of the Citrus Flavonoid Hesperidin in Humans: A Randomized, Double-Blind, Crossover Trial. J. Nutr. 2006, 136, 404–408. [Google Scholar] [CrossRef]
- Shulman, M.; Cohen, M.; Soto-Gutierrez, A.; Yagi, H.; Wang, H.; Goldwasser, J.; Lee-Parsons, C.W.; Benny-Ratsaby, O.; Yarmush, M.L.; Nahmias, Y. Enhancement of naringenin bioavailability by complexation with hydroxypropyl-β-cyclodextrin. PLoS ONE 2011, 6, e18033, Erratum in PLoS ONE 2012, 7. [Google Scholar]
- Manach, C.; Donovan, J.L. Pharmacokinetics and metabolism of dietary flavonoids in humans. Free Radic. Res. 2004, 38, 771–785. [Google Scholar] [CrossRef] [PubMed]
- Hanasaki, Y.; Ogawa, S.; Fukui, S. The correlation between active oxygens scavenging and antioxidative effects of flavonoids. Free Radic. Biol. Med. 1994, 16, 845–850. [Google Scholar] [CrossRef] [PubMed]
- Ursini, F.; Maiorino, M.; Morazzoni, P.; Roveri, A.; Pifferi, G. A novel antioxidant flavonoid (IdB 1031) affecting molecular mechanisms of cellular activation. Free Radic. Biol. Med. 1994, 16, 547–553. [Google Scholar] [CrossRef] [PubMed]
- van Acker, S.A.; van den Berg, D.J.; Tromp, M.N.; Griffioen, D.H.; van Bennekom, W.P.; van der Vijgh, W.J.; Bast, A. Structural aspects of antioxidant activity of flavonoids. Free Radic. Biol. Med. 1996, 20, 331–342. [Google Scholar] [CrossRef]
- Jovanovic, S.V.; Steenken, S.; Tosic, M.; Marjanovic, B.; Simic, M.G. Flavonoids as Antioxidants. J. Am. Chem. Soc. 1994, 116, 4846–4851. [Google Scholar] [CrossRef]
- Jovanovic, S.V.; Steenken, S.; Hara, Y.; Simic, M.G. Reduction potentials of flavonoid and model phenoxyl radicals. Which ring in flavonoids is responsible for antioxidant activity? J. Chem. Soc. Perkin Trans. 1996, 2, 2497–2504. [Google Scholar] [CrossRef]
- Tyuryaeva, I.; Lyublinskaya, O. Expected and Unexpected Effects of Pharmacological Antioxidants. Int. J. Mol. Sci. 2023, 24, 9303. [Google Scholar] [CrossRef]
- Cortés-Jofré, M.; Rueda, J.R.; Asenjo-Lobos, C.; Madrid, E.; Bonfill Cosp, X. Drugs for preventing lung cancer in healthy people. Cochrane Database Syst. Rev. 2020, 3, Cd002141. [Google Scholar] [CrossRef]
- Cui, Q.; Du, R.; Liu, M.; Rong, L. Lignans and Their Derivatives from Plants as Antivirals. Molecules 2020, 25, 183. [Google Scholar] [CrossRef]
- Singla, R.K.; Dubey, A.K.; Garg, A.; Sharma, R.K.; Fiorino, M.; Ameen, S.M.; Haddad, M.A.; Al-Hiary, M. Natural Polyphenols: Chemical Classification, Definition of Classes, Subcategories, and Structures. J. AOAC Int. 2019, 102, 1397–1400. [Google Scholar] [CrossRef]
- Aatif, M. Current Understanding of Polyphenols to Enhance Bioavailability for Better Therapies. Biomedicines 2023, 11, 2078. [Google Scholar] [CrossRef] [PubMed]
- Parage, C.; Tavares, R.; Réty, S.; Baltenweck-Guyot, R.; Poutaraud, A.; Renault, L.; Heintz, D.; Lugan, R.; Marais, G.A.; Aubourg, S.; et al. Structural, functional, and evolutionary analysis of the unusually large stilbene synthase gene family in grapevine. Plant Physiol. 2012, 160, 1407–1419. [Google Scholar] [CrossRef] [PubMed]
- Hapeshi, A.; Benarroch, J.M.; Clarke, D.J.; Waterfield, N.R. Iso-propyl stilbene: A life cycle signal? Microbiology 2019, 165, 516–526. [Google Scholar] [CrossRef]
- Câmara, J.S.; Albuquerque, B.R.; Aguiar, J.; Corrêa, R.C.G.; Gonçalves, J.L.; Granato, D.; Pereira, J.A.M.; Barros, L.; Ferreira, I. Food Bioactive Compounds and Emerging Techniques for Their Extraction: Polyphenols as a Case Study. Foods 2020, 10, 37. [Google Scholar] [CrossRef]
- Basholli-Salihu, M.; Schuster, R.; Mulla, D.; Praznik, W.; Viernstein, H.; Mueller, M. Bioconversion of piceid to resveratrol by selected probiotic cell extracts. Bioprocess. Biosyst. Eng. 2016, 39, 1879–1885. [Google Scholar] [CrossRef]
- Dwibedi, V.; Kalia, S.; Saxena, S. Isolation and enhancement of resveratrol production in Xylaria psidii by exploring the phenomenon of epigenetics: Using DNA methyltransferases and histone deacetylase as epigenetic modifiers. Mol. Biol. Rep. 2019, 46, 4123–4137. [Google Scholar] [CrossRef]
- El Khawand, T.; Courtois, A.; Valls, J.; Richard, T.; Krisa, S. A review of dietary stilbenes: Sources and bioavailability. Phytochem. Rev. 2018, 17, 1007–1029. [Google Scholar] [CrossRef]
- Lara-Ochoa, F.; Sandoval-Minero, L.C.; Espinosa-Pérez, G. A new synthesis of resveratrol. Tetrahedron Lett. 2015, 56, 5977–5979. [Google Scholar] [CrossRef]
- Feng, C.; Chen, J.; Ye, W.; Liao, K.; Wang, Z.; Song, X.; Qiao, M. Synthetic Biology-Driven Microbial Production of Resveratrol: Advances and Perspectives. Front. Bioeng. Biotechnol. 2022, 10, 833920. [Google Scholar] [CrossRef]
- Licznerska, B.; Szaefer, H.; Wierzchowski, M.; Mikstacka, R.; Papierska, K.; Baer-Dubowska, W. Evaluation of the effect of the new methoxy-stilbenes on expression of receptors and enzymes involved in estrogen synthesis in cancer breast cells. Mol. Cell Biochem. 2018, 444, 53–62. [Google Scholar] [CrossRef]
- Ahmed, T.; Javed, S.; Javed, S.; Tariq, A.; Šamec, D.; Tejada, S.; Nabavi, S.F.; Braidy, N.; Nabavi, S.M. Resveratrol and Alzheimer’s Disease: Mechanistic Insights. Mol. Neurobiol. 2017, 54, 2622–2635. [Google Scholar] [CrossRef] [PubMed]
- Walle, T.; Hsieh, F.; DeLegge, M.H.; Oatis, J.E., Jr.; Walle, U.K. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab. Dispos. 2004, 32, 1377–1382. [Google Scholar] [CrossRef] [PubMed]
- Amri, A.; Chaumeil, J.C.; Sfar, S.; Charrueau, C. Administration of resveratrol: What formulation solutions to bioavailability limitations? J. Control. Release Off. J. Control. Release Soc. 2012, 158, 182–193. [Google Scholar] [CrossRef] [PubMed]
- Patel, K.R.; Andreadi, C.; Britton, R.G.; Horner-Glister, E.; Karmokar, A.; Sale, S.; Brown, V.A.; Brenner, D.E.; Singh, R.; Steward, W.P.; et al. Sulfate metabolites provide an intracellular pool for resveratrol generation and induce autophagy with senescence. Sci. Transl. Med. 2013, 5, 205ra133. [Google Scholar] [CrossRef]
- Knobloch, J.; Sibbing, B.; Jungck, D.; Lin, Y.; Urban, K.; Stoelben, E.; Strauch, J.; Koch, A. Resveratrol impairs the release of steroid-resistant inflammatory cytokines from human airway smooth muscle cells in chronic obstructive pulmonary disease. J. Pharmacol. Exp. Ther. 2010, 335, 788–798. [Google Scholar] [CrossRef]
- Bagniewska-Zadworna, A.; Barakat, A.; Łakomy, P.; Smoliński, D.J.; Zadworny, M. Lignin and lignans in plant defence: Insight from expression profiling of cinnamyl alcohol dehydrogenase genes during development and following fungal infection in Populus. Plant Sci. Int. J. Exp. Plant Biol. 2014, 229, 111–121. [Google Scholar] [CrossRef]
- Garros, L.; Drouet, S.; Corbin, C.; Decourtil, C.; Fidel, T.; Lebas de Lacour, J.; Leclerc, E.A.; Renouard, S.; Tungmunnithum, D.; Doussot, J.; et al. Insight into the Influence of Cultivar Type, Cultivation Year, and Site on the Lignans and Related Phenolic Profiles, and the Health-Promoting Antioxidant Potential of Flax (Linum usitatissimum L.) Seeds. Molecules 2018, 23, 2636. [Google Scholar] [CrossRef]
- Yang, C.S.; Landau, J.M.; Huang, M.T.; Newmark, H.L. Inhibition of carcinogenesis by dietary polyphenolic compounds. Annu. Rev. Nutr. 2001, 21, 381–406. [Google Scholar] [CrossRef]
- Adlercreutz, H. Lignans and human health. Crit. Rev. Clin. Lab. Sci. 2007, 44, 483–525. [Google Scholar] [CrossRef]
- Smeds, A.I.; Eklund, P.C.; Sjöholm, R.E.; Willför, S.M.; Nishibe, S.; Deyama, T.; Holmbom, B.R. Quantification of a broad spectrum of lignans in cereals, oilseeds, and nuts. J. Agric. Food Chem. 2007, 55, 1337–1346. [Google Scholar] [CrossRef]
- Caisheng, Q.; Wang, H.; Guo, Y.; Long, S.; Wang, Y.F.; Abbasi, A.; Guo, X.; Jarvis, D. Comparison of Fatty Acid Composition, Phytochemical Profiles and Antioxidant Activities in Four Flax (Linum usitatissimum L.) Varieties. Oil Crop Sci. 2020, 5, 136–141. [Google Scholar] [CrossRef]
- Hosseinian, F.; Muir, A.; Westcott, N.; Krol, E. Antioxidant capacity of flaxseed lignans in two model systems. J. Oil Fat. Ind. 2006, 83, 835–840. [Google Scholar] [CrossRef]
- Pietrofesa, R.A.; Chatterjee, S.; Kadariya, Y.; Testa, J.R.; Albelda, S.M.; Christofidou-Solomidou, M. Synthetic Secoisolariciresinol Diglucoside (LGM2605) Prevents Asbestos-Induced Inflammation and Genotoxic Cell Damage in Human Mesothelial Cells. Int. J. Mol. Sci. 2022, 23, 85. [Google Scholar] [CrossRef]
- Touré, A.; Xueming, X. Flaxseed Lignans: Source, Biosynthesis, Metabolism, Antioxidant Activity, Bio-Active Components, and Health Benefits. Compr. Rev. Food Sci. Food Saf. 2010, 9, 261–269. [Google Scholar] [CrossRef]
- Guo, J.; Huang, X.; Dou, L.; Yan, M.; Shen, T.; Tang, W. Aging and aging-related diseases: From molecular mechanisms to interventions and treatments. Signal Transduct. Target. Ther. 2022, 7, 1. [Google Scholar] [CrossRef]
- Varricchi, G.; Ameri, P.; Cadeddu, C.; Ghigo, A.; Madonna, R.; Marone, G.; Mercurio, V.; Monte, I.; Novo, G.; Parrella, P.; et al. Antineoplastic Drug-Induced Cardiotoxicity: A Redox Perspective. Front. Physiol. 2018, 9, 167. [Google Scholar] [CrossRef]
- Loffredo, L.; Carnevale, R.; Perri, L.; Catasca, E.; Augelletti, T.; Cangemi, R.; Albanese, F.; Piccheri, C.; Nocella, C.; Pignatelli, P.; et al. NOX2-mediated arterial dysfunction in smokers: Acute effect of dark chocolate. Heart 2011, 97, 1776–1781. [Google Scholar] [CrossRef]
- Polycarpou, E.; Meira, L.B.; Carrington, S.; Tyrrell, E.; Modjtahedi, H.; Carew, M.A. Resveratrol 3-O-D-glucuronide and resveratrol 4′-O-D-glucuronide inhibit colon cancer cell growth: Evidence for a role of A3 adenosine receptors, cyclin D1 depletion, and G1 cell cycle arrest. Mol. Nutr. Food Res. 2013, 57, 1708–1717. [Google Scholar] [CrossRef]
- Mazzone, G.; Malaj, N.; Russo, N.; Toscano, M. Density functional study of the antioxidant activity of some recently synthesized resveratrol analogues. Food Chem. 2013, 141, 2017–2024. [Google Scholar] [CrossRef]
- Nam, K.Y.; Damodar, K.; Lee, Y.; Park, L.S.; Gim, J.G.; Park, J.P.; Jeon, S.H.; Lee, J.T. Design and Synthesis of π-Extended Resveratrol Analogues and In Vitro Antioxidant and Anti-Inflammatory Activity Evaluation. Molecules 2021, 26, 646. [Google Scholar] [CrossRef]
- Hung, L.M.; Su, M.J.; Chu, W.K.; Chiao, C.W.; Chan, W.F.; Chen, J.K. The protective effect of resveratrols on ischaemia-reperfusion injuries of rat hearts is correlated with antioxidant efficacy. Br. J. Pharmacol. 2002, 135, 1627–1633. [Google Scholar] [CrossRef] [PubMed]
- Sieg, G.; Müller, I.; Weißer, K.; Werncke, C.G. Taming the stilbene radical anion. Chem. Sci. 2022, 13, 13872–13878. [Google Scholar] [CrossRef] [PubMed]
- Sato, A.; Nishioka, S.; Kiuchi, M.; Imada, Y.; Makino, K.; Nakagawa, K.; Tanaka, R.; Matsumura, Y.; Ohkita, M. Grape Extract from Chardonnay Seeds Restores Deoxycorticosterone Acetate-Salt-Induced Endothelial Dysfunction and Hypertension in Rats. Biol. Pharm. Bull. 2020, 43, 59–67. [Google Scholar] [CrossRef]
- Edirisinghe, I.; Burton-Freeman, B.; Varelis, P.; Kappagoda, T. Strawberry extract caused endothelium-dependent relaxation through the activation of PI3 kinase/Akt. J. Agric. Food Chem. 2008, 56, 9383–9390. [Google Scholar] [CrossRef]
- Ndiaye, M.; Chataigneau, M.; Lobysheva, I.; Chataigneau, T.; Schini-Kerth, V.B. Red wine polyphenol-induced, endothelium-dependent NO-mediated relaxation is due to the redox-sensitive PI3-kinase/Akt-dependent phosphorylation of endothelial NO-synthase in the isolated porcine coronary artery. FASEB J. 2005, 19, 455–457. [Google Scholar] [CrossRef]
- Bhatt, S.R.; Lokhandwala, M.F.; Banday, A.A. Resveratrol prevents endothelial nitric oxide synthase uncoupling and attenuates development of hypertension in spontaneously hypertensive rats. Eur. J. Pharmacol. 2011, 667, 258–264. [Google Scholar] [CrossRef]
- Chantarudee, A.; Phuwapraisirisan, P.; Kimura, K.; Okuyama, M.; Mori, H.; Kimura, A.; Chanchao, C. Chemical constituents and free radical scavenging activity of corn pollen collected from Apis mellifera hives compared to floral corn pollen at Nan, Thailand. BMC Complement. Altern. Med. 2012, 12, 45. [Google Scholar] [CrossRef]
- Bansal, P.; Paul, P.; Nayak, P.G.; Pannakal, S.T.; Zou, J.-h.; Laatsch, H.; Priyadarsini, K.I.; Unnikrishnan, M.K. Phenolic compounds isolated from Pilea microphylla prevent radiation-induced cellular DNA damage. Acta Pharm. Sin. B 2011, 1, 226–235. [Google Scholar] [CrossRef]
- Kumar, S.; Pandey, A.K. Chemistry and biological activities of flavonoids: An overview. Sci. World J. 2013, 2013, 162750. [Google Scholar] [CrossRef]
- Flanagan, E.; Müller, M.; Hornberger, M.; Vauzour, D. Impact of Flavonoids on Cellular and Molecular Mechanisms Underlying Age-Related Cognitive Decline and Neurodegeneration. Curr. Nutr. Rep. 2018, 7, 49–57. [Google Scholar] [CrossRef]
- Li, L.; Lyall, G.K.; Martinez-Blazquez, J.A.; Vallejo, F.; Tomas-Barberan, F.A.; Birch, K.M.; Boesch, C. Blood Orange Juice Consumption Increases Flow-Mediated Dilation in Adults with Overweight and Obesity: A Randomized Controlled Trial. J. Nutr. 2020, 150, 2287–2294. [Google Scholar] [CrossRef] [PubMed]
- Heiss, C.; Finis, D.; Kleinbongard, P.; Hoffmann, A.; Rassaf, T.; Kelm, M.; Sies, H. Sustained increase in flow-mediated dilation after daily intake of high-flavanol cocoa drink over 1 week. J. Cardiovasc. Pharmacol. 2007, 49, 74–80. [Google Scholar] [CrossRef] [PubMed]
- Sanguigni, V.; Manco, M.; Sorge, R.; Gnessi, L.; Francomano, D. Natural antioxidant ice cream acutely reduces oxidative stress and improves vascular function and physical performance in healthy individuals. Nutrition 2017, 33, 225–233. [Google Scholar] [CrossRef]
- Li, H.; Liu, L.; Cao, Z.; Li, W.; Liu, R.; Chen, Y.; Li, C.; Song, Y.; Liu, G.; Hu, J.; et al. Naringenin ameliorates homocysteine induced endothelial damage via the AMPKα/Sirt1 pathway. J. Adv. Res. 2021, 34, 137–147. [Google Scholar] [CrossRef]
- Xu, Q.; Si, L.Y. Protective effects of AMP-activated protein kinase in the cardiovascular system. J. Cell Mol. Med. 2010, 14, 2604–2613. [Google Scholar] [CrossRef]
- Daubney, J.; Bonner, P.L.; Hargreaves, A.J.; Dickenson, J.M. Cardioprotective and cardiotoxic effects of quercetin and two of its in vivo metabolites on differentiated h9c2 cardiomyocytes. Basic. Clin. Pharmacol. Toxicol. 2015, 116, 96–109. [Google Scholar] [CrossRef]
- Lee, G.H.; Kim, C.Y.; Zheng, C.; Jin, S.W.; Kim, J.Y.; Lee, S.Y.; Kim, M.Y.; Han, E.H.; Hwang, Y.P.; Jeong, H.G. Rutaecarpine Increases Nitric Oxide Synthesis via eNOS Phosphorylation by TRPV1-Dependent CaMKII and CaMKKβ/AMPK Signaling Pathway in Human Endothelial Cells. Int. J. Mol. Sci. 2021, 22, 9407. [Google Scholar] [CrossRef]
- Serreli, G.; Deiana, M. Role of Dietary Polyphenols in the Activity and Expression of Nitric Oxide Synthases: A Review. Antioxidants 2023, 12, 147. [Google Scholar] [CrossRef]
- Park, E.-S.; Kang, J.; Jang, Y.; Park, J.-S.; Jang, S.; Kim, D.-E.; Kim, B.; Shin, H.-S. Cardioprotective effects of rhamnetin in H9c2 cardiomyoblast cells under H2O2-induced apoptosis. J. Ethnopharmacol. 2014, 153, 552–560. [Google Scholar] [CrossRef]
- Boonla, O.; Kukongviriyapan, U.; Pakdeechote, P.; Kukongviriyapan, V.; Pannangpetch, P.; Prachaney, P.; Greenwald, S.E. Curcumin improves endothelial dysfunction and vascular remodeling in 2K-1C hypertensive rats by raising nitric oxide availability and reducing oxidative stress. Nitric Oxide 2014, 42, 44–53. [Google Scholar] [CrossRef]
- Zhang, Q.; Liu, J.; Duan, H.; Li, R.; Peng, W.; Wu, C. Activation of Nrf2/HO-1 signaling: An important molecular mechanism of herbal medicine in the treatment of atherosclerosis via the protection of vascular endothelial cells from oxidative stress. J. Adv. Res. 2021, 34, 43–63. [Google Scholar] [CrossRef] [PubMed]
- Dinkova-Kostova, A.T.; Liby, K.T.; Stephenson, K.K.; Holtzclaw, W.D.; Gao, X.; Suh, N.; Williams, C.; Risingsong, R.; Honda, T.; Gribble, G.W.; et al. Extremely potent triterpenoid inducers of the phase 2 response: Correlations of protection against oxidant and inflammatory stress. Proc. Natl. Acad. Sci. USA 2005, 102, 4584–4589. [Google Scholar] [CrossRef] [PubMed]
- Kulkarni, S.R.; Donepudi, A.C.; Xu, J.; Wei, W.; Cheng, Q.C.; Driscoll, M.V.; Johnson, D.A.; Johnson, J.A.; Li, X.; Slitt, A.L. Fasting induces nuclear factor E2-related factor 2 and ATP-binding Cassette transporters via protein kinase A and Sirtuin-1 in mouse and human. Antioxid. Redox Signal. 2014, 20, 15–30. [Google Scholar] [CrossRef]
- Giovinazzo, G.; Grieco, F. Functional Properties of Grape and Wine Polyphenols. Plant Foods Hum. Nutr. 2015, 70, 454–462. [Google Scholar] [CrossRef]
- Kao, C.L.; Chen, L.K.; Chang, Y.L.; Yung, M.C.; Hsu, C.C.; Chen, Y.C.; Lo, W.L.; Chen, S.J.; Ku, H.H.; Hwang, S.J. Resveratrol protects human endothelium from H(2)O(2)-induced oxidative stress and senescence via SirT1 activation. J. Atheroscler. Thromb. 2010, 17, 970–979. [Google Scholar] [CrossRef]
- Gimblet, C.J.; Kruse, N.T.; Geasland, K.; Michelson, J.; Sun, M.; Mandukhail, S.R.; Wendt, L.H.; Eyck, P.T.; Pierce, G.L.; Jalal, D.I. Effect of Resveratrol on Endothelial Function in Patients with CKD and Diabetes: A Randomized Controlled Trial. Clin. J. Am. Soc. Nephrol. 2024, 19, 161–168. [Google Scholar] [CrossRef]
- Xia, N.; Strand, S.; Schlufter, F.; Siuda, D.; Reifenberg, G.; Kleinert, H.; Förstermann, U.; Li, H. Role of SIRT1 and FOXO factors in eNOS transcriptional activation by resveratrol. Nitric Oxide 2013, 32, 29–35. [Google Scholar] [CrossRef]
- Kim, J.A.; Formoso, G.; Li, Y.; Potenza, M.A.; Marasciulo, F.L.; Montagnani, M.; Quon, M.J. Epigallocatechin gallate, a green tea polyphenol, mediates NO-dependent vasodilation using signaling pathways in vascular endothelium requiring reactive oxygen species and Fyn. J. Biol. Chem. 2007, 282, 13736–13745. [Google Scholar] [CrossRef]
- Zhang, Q.J.; Wang, Z.; Chen, H.Z.; Zhou, S.; Zheng, W.; Liu, G.; Wei, Y.S.; Cai, H.; Liu, D.P.; Liang, C.C. Endothelium-specific overexpression of class III deacetylase SIRT1 decreases atherosclerosis in apolipoprotein E-deficient mice. Cardiovasc. Res. 2008, 80, 191–199. [Google Scholar] [CrossRef]
- Mattagajasingh, I.; Kim, C.S.; Naqvi, A.; Yamamori, T.; Hoffman, T.A.; Jung, S.B.; DeRicco, J.; Kasuno, K.; Irani, K. SIRT1 promotes endothelium-dependent vascular relaxation by activating endothelial nitric oxide synthase. Proc. Natl. Acad. Sci. USA 2007, 104, 14855–14860. [Google Scholar] [CrossRef]
- Thomas, J.; Patel, A.; Syam, D.; Sreevallabhan, S.; Madhavamenon, K.; Mohanan, R. Clove bud (Syzygium aromaticum L.) polyphenol helps to mitigate metabolic syndrome by establishing intracellular redox homeostasis and glucose metabolism: A randomized, double-blinded, active-controlled comparative study. J. Funct. Foods 2022, 98, 105273. [Google Scholar] [CrossRef]
- Liu, C.-W.; Sung, H.-C.; Lin, S.-R.; Wu, C.-W.; Lee, C.-W.; Lee, I.T.; Yang, Y.-F.; Yu, I.S.; Lin, S.-W.; Chiang, M.-H.; et al. Resveratrol attenuates ICAM-1 expression and monocyte adhesiveness to TNF-α-treated endothelial cells: Evidence for an anti-inflammatory cascade mediated by the miR-221/222/AMPK/p38/NF-κB pathway. Sci. Rep. 2017, 7, 44689. [Google Scholar] [CrossRef] [PubMed]
- Zhao, W.; Feng, H.; Guo, S.; Han, Y.; Chen, X. Danshenol A inhibits TNF-α-induced expression of intercellular adhesion molecule-1 (ICAM-1) mediated by NOX4 in endothelial cells. Sci. Rep. 2017, 7, 12953. [Google Scholar] [CrossRef] [PubMed]
- Malekmohammad, K.; Bezsonov, E.E.; Rafieian-Kopaei, M. Role of Lipid Accumulation and Inflammation in Atherosclerosis: Focus on Molecular and Cellular Mechanisms. Front. Cardiovasc. Med. 2021, 8, 707529. [Google Scholar] [CrossRef]
- Kong, P.; Cui, Z.-Y.; Huang, X.-F.; Zhang, D.-D.; Guo, R.-J.; Han, M. Inflammation and atherosclerosis: Signaling pathways and therapeutic intervention. Signal. Transduct. Target. Ther. 2022, 7, 131. [Google Scholar] [CrossRef]
- Zhang, Y.; Liu, H.; Tang, W.; Qiu, Q.; Peng, J. Resveratrol prevents TNF-α-induced VCAM-1 and ICAM-1 upregulation in endothelial progenitor cells via reduction of NF-κB activation. J. Int. Med. Res. 2020, 48, 300060520945131. [Google Scholar] [CrossRef]
- Masaki, T.; Sawamura, T. Endothelin and endothelial dysfunction. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2006, 82, 17–24. [Google Scholar] [CrossRef]
- Marasciulo, F.L.; Montagnani, M.; Potenza, M.A. Endothelin-1: The yin and yang on vascular function. Curr. Med. Chem. 2006, 13, 1655–1665. [Google Scholar] [CrossRef]
- Böhm, F.; Pernow, J. The importance of endothelin-1 for vascular dysfunction in cardiovascular disease. Cardiovasc. Res. 2007, 76, 8–18. [Google Scholar] [CrossRef]
- Dong, F.; Zhang, X.; Wold, L.E.; Ren, Q.; Zhang, Z.; Ren, J. Endothelin-1 enhances oxidative stress, cell proliferation and reduces apoptosis in human umbilical vein endothelial cells: Role of ETB receptor, NADPH oxidase and caveolin-1. Br. J. Pharmacol. 2005, 145, 323–333. [Google Scholar] [CrossRef]
- Kowalczyk, A.; Kleniewska, P.; Kolodziejczyk, M.; Skibska, B.; Goraca, A. The role of endothelin-1 and endothelin receptor antagonists in inflammatory response and sepsis. Arch. Immunol. Ther. Exp. 2015, 63, 41–52. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.M.; Kim, Y. Is Curcumin Intake Really Effective for Chronic Inflammatory Metabolic Disease? A Review of Meta-Analyses of Randomized Controlled Trials. Nutrients 2024, 16, 1728. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.C.; Chen, J.J.; Chan, P.; Cheng, C.F.; Cheng, T.H. Inhibition of cyclic strain-induced endothelin-1 gene expression by resveratrol. Hypertension 2003, 42, 1198–1205. [Google Scholar] [CrossRef] [PubMed]
- Storniolo, C.E.; Roselló-Catafau, J.; Pintó, X.; Mitjavila, M.T.; Moreno, J.J. Polyphenol fraction of extra virgin olive oil protects against endothelial dysfunction induced by high glucose and free fatty acids through modulation of nitric oxide and endothelin-1. Redox Biol. 2014, 2, 971–977. [Google Scholar] [CrossRef]
- Khan, N.Q.; Lees, D.M.; Douthwaite, J.A.; Carrier, M.J.; Corder, R. Comparison of red wine extract and polyphenol constituents on endothelin-1 synthesis by cultured endothelial cells. Clin. Sci. 2002, 103 (Suppl. S48), 72s–75s. [Google Scholar] [CrossRef]
- Dzau, V.J. Theodore Cooper Lecture: Tissue angiotensin and pathobiology of vascular disease: A unifying hypothesis. Hypertension 2001, 37, 1047–1052. [Google Scholar] [CrossRef]
- Griendling, K.K.; Lassègue, B.; Murphy, T.J.; Alexander, R.W. Angiotensin II receptor pharmacology. Adv. Pharmacol. 1994, 28, 269–306. [Google Scholar] [CrossRef]
- Kossmann, S.; Hu, H.; Steven, S.; Schönfelder, T.; Fraccarollo, D.; Mikhed, Y.; Brähler, M.; Knorr, M.; Brandt, M.; Karbach, S.H.; et al. Inflammatory monocytes determine endothelial nitric-oxide synthase uncoupling and nitro-oxidative stress induced by angiotensin II. J. Biol. Chem. 2014, 289, 27540–27550. [Google Scholar] [CrossRef]
- Rincón, J.; Correia, D.; Arcaya, J.L.; Finol, E.; Fernández, A.; Pérez, M.; Yaguas, K.; Talavera, E.; Chávez, M.; Summer, R.; et al. Role of Angiotensin II type 1 receptor on renal NAD(P)H oxidase, oxidative stress and inflammation in nitric oxide inhibition induced-hypertension. Life Sci. 2015, 124, 81–90. [Google Scholar] [CrossRef]
- Ra, J.-E.; Woo, S.-Y.; Jin, H.; Lee, M.J.; Kim, H.Y.; Ham, H.; Chung, I.-M.; Seo, W.D. Evaluation of antihypertensive polyphenols of barley (Hordeum vulgare L.) seedlings via their effects on angiotensin-converting enzyme (ACE) inhibition. Appl. Biol. Chem. 2020, 63, 38. [Google Scholar] [CrossRef]
- Kim, E.N.; Kim, M.Y.; Lim, J.H.; Kim, Y.; Shin, S.J.; Park, C.W.; Kim, Y.S.; Chang, Y.S.; Yoon, H.E.; Choi, B.S. The protective effect of resveratrol on vascular aging by modulation of the renin-angiotensin system. Atherosclerosis 2018, 270, 123–131. [Google Scholar] [CrossRef] [PubMed]
- Hickson, L.J.; Langhi Prata, L.G.P.; Bobart, S.A.; Evans, T.K.; Giorgadze, N.; Hashmi, S.K.; Herrmann, S.M.; Jensen, M.D.; Jia, Q.; Jordan, K.L.; et al. Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease. EBioMedicine 2019, 47, 446–456. [Google Scholar] [CrossRef] [PubMed]
- Nambiar, A.; Kellogg, D., 3rd; Justice, J.; Goros, M.; Gelfond, J.; Pascual, R.; Hashmi, S.; Masternak, M.; Prata, L.; LeBrasseur, N.; et al. Senolytics dasatinib and quercetin in idiopathic pulmonary fibrosis: Results of a phase I, single-blind, single-center, randomized, placebo-controlled pilot trial on feasibility and tolerability. EBioMedicine 2023, 90, 104481. [Google Scholar] [CrossRef] [PubMed]
- Zumerle, S.; Sarill, M.; Saponaro, M.; Colucci, M.; Contu, L.; Lazzarini, E.; Sartori, R.; Pezzini, C.; Rinaldi, A.; Scanu, A.; et al. Targeting senescence induced by age or chemotherapy with a polyphenol-rich natural extract improves longevity and healthspan in mice. Nat. Aging 2024, 4, 1231–1248. [Google Scholar] [CrossRef]
- Khemais-Benkhiat, S.; Idris-Khodja, N.; Ribeiro, T.P.; Silva, G.C.; Abbas, M.; Kheloufi, M.; Lee, J.-O.; Toti, F.; Auger, C.; Schini-Kerth, V.B. The Redox-sensitive Induction of the Local Angiotensin System Promotes Both Premature and Replicative Endothelial Senescence: Preventive Effect of a Standardized Crataegus Extract. J. Gerontol. Ser. A 2016, 71, 1581–1590. [Google Scholar] [CrossRef]
- Jamal, J.; Mustafa, M.R.; Wong, P.F. Paeonol protects against premature senescence in endothelial cells by modulating Sirtuin 1 pathway. J. Ethnopharmacol. 2014, 154, 428–436. [Google Scholar] [CrossRef]
- Zhang, E.; Guo, Q.; Gao, H.; Xu, R.; Teng, S.; Wu, Y. Metformin and Resveratrol Inhibited High Glucose-Induced Metabolic Memory of Endothelial Senescence through SIRT1/p300/p53/p21 Pathway. PLoS ONE 2015, 10, e0143814. [Google Scholar] [CrossRef]
- D’Archivio, M.; Filesi, C.; Varì, R.; Scazzocchio, B.; Masella, R. Bioavailability of the polyphenols: Status and controversies. Int. J. Mol. Sci. 2010, 11, 1321–1342. [Google Scholar] [CrossRef]
- Moriwaki, M.; Kito, K.; Nakagawa, R.; Kapoor, M.P.; Matsumiya, Y.; Fukuhara, T.; Kamiya, U. Bioavailability comparison between a compound comprising hesperetin-7-glucoside with β-cyclodextrin and a mixture of hesperidin and dextrin in healthy adult human males. Biosci. Biotechnol. Biochem. 2021, 85, 2195–2199. [Google Scholar] [CrossRef]
- Nectoux, A.M.; Abe, C.; Huang, S.-W.; Ohno, N.; Tabata, J.; Miyata, Y.; Tanaka, K.; Tanaka, T.; Yamamura, H.; Matsui, T. Absorption and Metabolic Behavior of Hesperidin (Rutinosylated Hesperetin) after Single Oral Administration to Sprague-Dawley Rats. J. Agric. Food Chem. 2019, 67, 9812–9819. [Google Scholar] [CrossRef]
- Sinkar, S.R.; Kombe, S.V.; Samarth, V.D.; Satpute, S.V.; Deshattiwar, N.J.; Burade, N.V.; Dhuldhaj, U.P. Chemistry and Biological Activities of Quercetin: A Bioactive Flavonoid. Res. Journey—Int. E-Res. J. 2022, 297, 1–16. [Google Scholar]
- Sun, L.-L.; Wang, M.; Zhang, H.-J.; You, G.-j.; Liu, Y.-N.; Ren, X.-L.; Deng, Y.-R. The influence of polysaccharides from Ophiopogon japonicus on 2,3,5,4′-tetrahydroxy-stilbene-2-O-β-d-glucoside about biopharmaceutical properties in vitro and pharmacokinetics in vivo. Int. J. Biol. Macromol. 2018, 119, 677–682. [Google Scholar] [CrossRef] [PubMed]
- Sahraeian, S.; Rashidinejad, A.; Golmakani, M.-T. Recent advances in the conjugation approaches for enhancing the bioavailability of polyphenols. Food Hydrocoll. 2024, 146, 109221. [Google Scholar] [CrossRef]
- Guo, Y.; Sun, Q.; Wu, F.G.; Dai, Y.; Chen, X. Polyphenol-Containing Nanoparticles: Synthesis, Properties, and Therapeutic Delivery. Adv. Mater. 2021, 33, e2007356. [Google Scholar] [CrossRef]
- Alexander, A.; Ajazuddin; Patel, R.J.; Saraf, S.; Saraf, S. Recent expansion of pharmaceutical nanotechnologies and targeting strategies in the field of phytopharmaceuticals for the delivery of herbal extracts and bioactives. J. Control Release 2016, 241, 110–124. [Google Scholar] [CrossRef]
- Prasad, S.; Tyagi, A.K.; Aggarwal, B.B. Recent developments in delivery, bioavailability, absorption and metabolism of curcumin: The golden pigment from golden spice. Cancer Res. Treat. 2014, 46, 2–18. [Google Scholar] [CrossRef]
- Chen, L.; Alrbyawi, H.; Poudel, I.; Arnold, R.D.; Babu, R.J. Co-delivery of Doxorubicin and Ceramide in a Liposomal Formulation Enhances Cytotoxicity in Murine B16BL6 Melanoma Cell Lines. AAPS PharmSciTech 2019, 20, 99. [Google Scholar] [CrossRef]
- Zhou, H.; Qian, Q.; Chen, Q.; Chen, T.; Wu, C.; Chen, L.; Zhang, Z.; Wu, O.; Jin, Y.; Wang, X.; et al. Enhanced Mitochondrial Targeting and Inhibition of Pyroptosis with Multifunctional Metallopolyphenol Nanoparticles in Intervertebral Disc Degeneration. Small 2024, 20, e2308167. [Google Scholar] [CrossRef]
- Safinya, C.R.; Ewert, K.K. Materials chemistry: Liposomes derived from molecular vases. Nature 2012, 489, 372–374. [Google Scholar] [CrossRef]
- Negi, P.; Aggarwal, M.; Sharma, G.; Rathore, C.; Sharma, G.; Singh, B.; Katare, O.P. Niosome-based hydrogel of resveratrol for topical applications: An effective therapy for pain related disorder(s). Biomed. Pharmacother. 2017, 88, 480–487. [Google Scholar] [CrossRef]
- Moghassemi, S.; Hadjizadeh, A. Nano-niosomes as nanoscale drug delivery systems: An illustrated review. J. Control Release 2014, 185, 22–36. [Google Scholar] [CrossRef] [PubMed]
- Michalicha, A.; Belcarz, A.; Giannakoudakis, D.A.; Staniszewska, M.; Barczak, M. Designing Composite Stimuli-Responsive Hydrogels for Wound Healing Applications: The State-of-the-Art and Recent Discoveries. Materials 2024, 17, 278. [Google Scholar] [CrossRef] [PubMed]
- Bagher, Z.; Ehterami, A.; Safdel, M.H.; Khastar, H.; Semiari, H.; Asefnejad, A.; Davachi, S.M.; Mirzaii, M.; Salehi, M. Wound healing with alginate/chitosan hydrogel containing hesperidin in rat model. J. Drug Deliv. Sci. Technol. 2020, 55, 101379. [Google Scholar] [CrossRef]
- Tyagi, N.; De, R.; Begun, J.; Popat, A. Cancer therapeutics with epigallocatechin-3-gallate encapsulated in biopolymeric nanoparticles. Int. J. Pharm. 2017, 518, 220–227. [Google Scholar] [CrossRef]
- Lambert, J.D.; Hong, J.; Kim, D.H.; Mishin, V.M.; Yang, C.S. Piperine enhances the bioavailability of the tea polyphenol (-)-epigallocatechin-3-gallate in mice. J. Nutr. 2004, 134, 1948–1952. [Google Scholar] [CrossRef]
- Agulló, V.; García-Viguera, C.; Domínguez-Perles, R. The use of alternative sweeteners (sucralose and stevia) in healthy soft-drink beverages, enhances the bioavailability of polyphenols relative to the classical caloric sucrose. Food Chem. 2022, 370, 131051. [Google Scholar] [CrossRef]
- Saik, A.Y.; Lim, Y.Y.; Stanslas, J.; Choo, W.S. Enzymatic synthesis of quercetin oleate esters using Candida antarctica lipase B. Biotechnol. Lett. 2017, 39, 297–304. [Google Scholar] [CrossRef]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
- Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef]
- Kruger, S.; Ilmer, M.; Kobold, S.; Cadilha, B.L.; Endres, S.; Ormanns, S.; Schuebbe, G.; Renz, B.W.; D’Haese, J.G.; Schloesser, H.; et al. Advances in cancer immunotherapy 2019—Latest trends. J. Exp. Clin. Cancer Res. 2019, 38, 268. [Google Scholar] [CrossRef]
- Sordo-Bahamonde, C.; Lorenzo-Herrero, S.; Gonzalez-Rodriguez, A.P.; Martínez-Pérez, A.; Rodrigo, J.P.; García-Pedrero, J.M.; Gonzalez, S. Chemo-Immunotherapy: A New Trend in Cancer Treatment. Cancers 2023, 15, 2912. [Google Scholar] [CrossRef] [PubMed]
- Lim, S.O.; Li, C.W.; Xia, W.; Cha, J.H.; Chan, L.C.; Wu, Y.; Chang, S.S.; Lin, W.C.; Hsu, J.M.; Hsu, Y.H.; et al. Deubiquitination and Stabilization of PD-L1 by CSN5. Cancer Cell 2016, 30, 925–939. [Google Scholar] [CrossRef] [PubMed]
- Verdura, S.; Cuyàs, E.; Cortada, E.; Brunet, J.; Lopez-Bonet, E.; Martin-Castillo, B.; Bosch-Barrera, J.; Encinar, J.A.; Menendez, J.A. Resveratrol targets PD-L1 glycosylation and dimerization to enhance antitumor T-cell immunity. Aging 2020, 12, 8–34. [Google Scholar] [CrossRef]
- Liu, K.; Sun, Q.; Liu, Q.; Li, H.; Zhang, W.; Sun, C. Focus on immune checkpoint PD-1/PD-L1 pathway: New advances of polyphenol phytochemicals in tumor immunotherapy. Biomed. Pharmacother. 2022, 154, 113618. [Google Scholar] [CrossRef]
- Mileo, A.M.; Nisticò, P.; Miccadei, S. Polyphenols: Immunomodulatory and Therapeutic Implication in Colorectal Cancer. Front. Immunol. 2019, 10, 729. [Google Scholar] [CrossRef]
- Focaccetti, C.; Izzi, V.; Benvenuto, M.; Fazi, S.; Ciuffa, S.; Giganti, M.G.; Potenza, V.; Manzari, V.; Modesti, A.; Bei, R. Polyphenols as Immunomodulatory Compounds in the Tumor Microenvironment: Friends or Foes? Int. J. Mol. Sci. 2019, 20, 1714. [Google Scholar] [CrossRef]
- 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.; et al. 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]
- Ostwal, V.; Ramaswamy, A.; Bhargava, P.; Srinivas, S.; Mandavkar, S.; Chaugule, D.; Peelay, Z.; Baheti, A.; Tandel, H.; Jadhav, V.K.; et al. A pro-oxidant combination of resveratrol and copper reduces chemotherapy-related non-haematological toxicities in advanced gastric cancer: Results of a prospective open label phase II single-arm study (RESCU III study). Med. Oncol. 2022, 40, 17. [Google Scholar] [CrossRef]
Chemotherapy Class | Drug | Indications | Source |
---|---|---|---|
Anthracyclines | Doxorubicin | Acute leukaemias; Hodgkin’s lymphoma; non-Hodgkin’s lymphoma; solid tumours (including breast cancer); soft tissue sarcoma; bladder cancer; AIDS-related Kaposi’s sarcoma; ovarian cancer; multiple myeloma | [14] |
Epirubicin | Breast cancer; gastric cancer; small cell lung cancer; ovarian cancer; colorectal cancer; lymphoma; leukaemia; multiple myeloma; bladder cancer | [15] | |
Non-anthracycline antitumour antibiotics | Bleomycin | Squamous cell carcinoma of the head and neck, cervix, and external genitalia; testicular cancer; non-Hodgkin’s lymphoma; Hodgkin’s lymphoma | [16] |
Alkylating agents | Cisplatin | Testicular cancer; ovarian cancer; lung cancer; bladder cancer; squamous cell carcinoma of the head and neck; cervical carcinoma | [17] |
Antimetabolites | Fluorouracil | Solid tumours (including gastrointestinal tract and breast cancers); colorectal cancer; superficial malignant and premalignant skin lesions | [18] |
Topoisomerase inhibitors | Etoposide | Testicular cancer; small cell lung cancer; Hodgkin’s lymphoma; non-Hodgkin’s lymphoma; acute myeloid leukaemia; ovarian cancer; gestational trophoblastic neoplasia | [19] |
Mitotic inhibitors | Docetaxel | Breast cancer; non-small cell lung cancer; prostate cancer; gastric adenocarcinoma; squamous cell carcinoma of the head and neck | [20] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 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
Tillman, L.; Margalef Rieres, J.; Ahjem, E.; Bishop-Guest, F.; McGrath, M.; Hatrick, H.; Pranjol, M.Z.I. Thinking Outside the Therapeutic Box: The Potential of Polyphenols in Preventing Chemotherapy-Induced Endothelial Dysfunction. Cells 2025, 14, 566. https://doi.org/10.3390/cells14080566
Tillman L, Margalef Rieres J, Ahjem E, Bishop-Guest F, McGrath M, Hatrick H, Pranjol MZI. Thinking Outside the Therapeutic Box: The Potential of Polyphenols in Preventing Chemotherapy-Induced Endothelial Dysfunction. Cells. 2025; 14(8):566. https://doi.org/10.3390/cells14080566
Chicago/Turabian StyleTillman, Luke, Jaume Margalef Rieres, Elena Ahjem, Fynn Bishop-Guest, Meghan McGrath, Helena Hatrick, and Md Zahidul Islam Pranjol. 2025. "Thinking Outside the Therapeutic Box: The Potential of Polyphenols in Preventing Chemotherapy-Induced Endothelial Dysfunction" Cells 14, no. 8: 566. https://doi.org/10.3390/cells14080566
APA StyleTillman, L., Margalef Rieres, J., Ahjem, E., Bishop-Guest, F., McGrath, M., Hatrick, H., & Pranjol, M. Z. I. (2025). Thinking Outside the Therapeutic Box: The Potential of Polyphenols in Preventing Chemotherapy-Induced Endothelial Dysfunction. Cells, 14(8), 566. https://doi.org/10.3390/cells14080566