Phenolic Compounds Exerting Lipid-Regulatory, Anti-Inflammatory and Epigenetic Effects as Complementary Treatments in Cardiovascular Diseases
Abstract
:1. Introduction
2. Biologically Active Phenolic Compounds and Their Mechanisms of Action
2.1. Hydroxycinnamic Acids Group
2.2. Stilbenes
2.3. Flavonoids Group
2.3.1. Flavonols
2.3.2. Flavones
2.3.3. Flavanones
2.3.4. Flavanols
2.3.5. Isoflavones
2.3.6. Anthocyanidins
2.4. Guaiacols Group
3. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- European Society of Cardiology. 2019 ESC/EAS guidelines for the management of dyslipidaemias: Lipid modification to reduce cardiovascular risk. Atherosclerosis 2019, 290, 140–205. [Google Scholar] [CrossRef] [Green Version]
- Bloom, D.E.; Cafiero, E.T.; Jané-Llopis, E.; Abrahams-Gessel, S.; Bloom, L.R.; Fathima, S.; Feigl, A.B.; Gaziano, T.; Mowafi, M.; Pandya, A.; et al. The Global Economic Burden of Non-Communicable Diseases; World Economic Forum: Geneva, Switzerland, 2011. [Google Scholar]
- Cicero, A.F.G.; Colletti, A.; Bajraktari, G.; Descamps, O.; Djuric, D.M.; Ezhov, M.; Fras, Z.; Katsiki, N.; Langlois, M.; Latkovskis, G.; et al. Lipid-lowering nutraceuticals in clinical practice: Position paper from an International Lipid Expert Panel. Nutr. Rev. 2017, 75, 731–767. [Google Scholar] [CrossRef] [PubMed]
- Sando, K.R.; Knight, M. Nonstatin therapies for management of dyslipidemia: A review. Clin. Ther. 2015, 37, 2153–2179. [Google Scholar] [CrossRef] [PubMed]
- Uehara, Y.; Chiesa, G.; Saku, K. High-Density Lipoprotein-Targeted Therapy and Apolipoprotein A-I Mimetic Peptides. Circ. J. 2015, 79, 2523–2528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Musunuru, K. Atherogenic dyslipidemia: Cardiovascular risk and dietary intervention. Lipids 2010, 45, 907–914. [Google Scholar] [CrossRef] [Green Version]
- Sima, A.V.; Stancu, C.S.; Simionescu, M. Vascular endothelium in atherosclerosis. Cell Tissue Res. 2009, 335, 191–203. [Google Scholar] [CrossRef]
- Pavlovic, J.; Kavousi, M.; Ikram, M.A.; Leening, M.J.G. Updated treatment thresholds in the 2019 ESC/EAS dyslipidaemia guidelines substantially expand indications for statin use for primary prevention at population level: Results from the Rotterdam Study. Atherosclerosis 2020. [Google Scholar] [CrossRef]
- Wolin, M.S. Interactions of oxidants with vascular signaling systems. Arterioscler. Thromb. Vasc. Biol. 2000, 20, 1430–1442. [Google Scholar] [CrossRef] [Green Version]
- Puddu, P.; Puddu, G.M.; Cravero, E.; De Pascalis, S.; Muscari, A. The emerging role of cardiovascular risk factor-induced mitochondrial dysfunction in atherogenesis. J. Biomed. Sci. 2009, 16, 112. [Google Scholar] [CrossRef] [Green Version]
- Wattanapitayakul, S.K.; Bauer, J.A. Oxidative pathways in cardiovascular disease: Roles, mechanisms, and therapeutic implications. Pharmacol. Ther. 2001, 89, 187–206. [Google Scholar] [CrossRef]
- Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.; Mazur, M.; Telser, J. Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 2007, 39, 44–84. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Duan, W.; Lin, Y.; Yi, W.; Liang, Z.; Yan, J.; Wang, N.; Deng, C.; Zhang, S.; Li, Y.; et al. SIRT1 activation by curcumin pretreatment attenuates mitochondrial oxidative damage induced by myocardial ischemia reperfusion injury. Free Radic. Biol. Med. 2013, 65, 667–679. [Google Scholar] [CrossRef] [PubMed]
- Martindale, J.L.; Holbrook, N.J. Cellular response to oxidative stress: Signaling for suicide and survival. J. Cell. Physiol. 2002, 192, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.H.; Khor, T.O.; Shu, L.; Su, Z.Y.; Fuentes, F.; Kong, A.N. Dietary phytochemicals and cancer prevention: Nrf2 signaling, epigenetics, and cell death mechanisms in blocking cancer initiation and progression. Pharmacol. Ther. 2013, 137, 153–171. [Google Scholar] [CrossRef] [Green Version]
- Ortiz-Munoz, G.; Couret, D.; Lapergue, B.; Bruckert, E.; Meseguer, E.; Amarenco, P.; Meilhac, O. Dysfunctional HDL in acute stroke. Atherosclerosis 2016, 253, 75–80. [Google Scholar] [CrossRef]
- Carnuta, M.G.; Stancu, C.S.; Toma, L.; Sanda, G.M.; Niculescu, L.S.; Deleanu, M.; Popescu, A.C.; Popescu, M.R.; Vlad, A.; Dimulescu, D.R.; et al. Dysfunctional high-density lipoproteins have distinct composition, diminished anti-inflammatory potential and discriminate acute coronary syndrome from stable coronary artery disease patients. Sci. Rep. 2017, 7, 7295. [Google Scholar] [CrossRef]
- Parhofer, K.G. Increasing HDL-cholesterol and prevention of atherosclerosis: A critical perspective. Atheroscler. Suppl. 2015, 18, 109–111. [Google Scholar] [CrossRef]
- Variji, A.; Shokri, Y.; Fallahpour, S.; Zargari, M.; Bagheri, B.; Abediankenari, S.; Alizadeh, A.; Mahrooz, A. The combined utility of myeloperoxidase (MPO) and paraoxonase 1 (PON1) as two important HDL-associated enzymes in coronary artery disease: Which has a stronger predictive role? Atherosclerosis 2019, 280, 7–13. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, M.T.; Fernando, S.; Schwarz, N.; Tan, J.T.; Bursill, C.A.; Psaltis, P.J. Inflammation as a Therapeutic Target in Atherosclerosis. J. Clin. Med. 2019, 8, 1109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, Y.; Xian, X.; Wang, Z.; Bi, Y.; Chen, Q.; Han, X.; Tang, D.; Chen, R. Research Progress on the Relationship between Atherosclerosis and Inflammation. Biomolecules 2018, 8, 80. [Google Scholar] [CrossRef] [Green Version]
- Bi, Y.; Chen, J.; Hu, F.; Liu, J.; Li, M.; Zhao, L. M2 Macrophages as a Potential Target for Antiatherosclerosis Treatment. Neural Plast. 2019, 2019, 6724903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pamukcu, B.; Lip, G.Y.; Shantsila, E. The nuclear factor—kappa B pathway in atherosclerosis: A potential therapeutic target for atherothrombotic vascular disease. Thromb. Res. 2011, 128, 117–123. [Google Scholar] [CrossRef] [PubMed]
- Sosnowska, B.; Mazidi, M.; Penson, P.; Gluba-Brzozka, A.; Rysz, J.; Banach, M. The sirtuin family members SIRT1, SIRT3 and SIRT6: Their role in vascular biology and atherogenesis. Atherosclerosis 2017, 265, 275–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grebe, A.; Hoss, F.; Latz, E. NLRP3 Inflammasome and the IL-1 Pathway in Atherosclerosis. Circ. Res. 2018, 122, 1722–1740. [Google Scholar] [CrossRef]
- Botker, H.E.; Hausenloy, D.; Andreadou, I.; Antonucci, S.; Boengler, K.; Davidson, S.M.; Deshwal, S.; Devaux, Y.; Di Lisa, F.; Di Sante, M.; et al. Practical guidelines for rigor and reproducibility in preclinical and clinical studies on cardioprotection. Basic Res. Cardiol. 2018, 113, 39. [Google Scholar] [CrossRef] [Green Version]
- Jacquier, A. The complex eukaryotic transcriptome: Unexpected pervasive transcription and novel small RNAs. Nat. Rev. Genet. 2009, 10, 833–844. [Google Scholar] [CrossRef]
- Papageorgiou, N.; Tslamandris, S.; Giolis, A.; Tousoulis, D. MicroRNAs in Cardiovascular Disease: Perspectives and Reality. Cardiol. Rev. 2016, 24, 110–118. [Google Scholar] [CrossRef]
- Gomes, C.P.C.; Schroen, B.; Kuster, G.M.; Robinson, E.L.; Ford, K.; Squire, I.B.; Heymans, S.; Martelli, F.; Emanueli, C.; Devaux, Y.; et al. Regulatory RNAs in Heart Failure. Circulation 2020, 141, 313–328. [Google Scholar] [CrossRef]
- Moore, K.J.; Rayner, K.J.; Suarez, Y.; Fernandez-Hernando, C. microRNAs and cholesterol metabolism. Trends Endocrinol. Metab. TEM 2010, 21, 699–706. [Google Scholar] [CrossRef] [Green Version]
- Bartel, D.P. MicroRNAs: Target recognition and regulatory functions. Cell 2009, 136, 215–233. [Google Scholar] [CrossRef] [Green Version]
- Forman, J.J.; Coller, H.A. The code within the code: MicroRNAs target coding regions. Cell Cycle 2010, 9, 1533–1541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vickers, K.C.; Palmisano, B.T.; Shoucri, B.M.; Shamburek, R.D.; Remaley, A.T. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat. Cell Biol. 2011, 13, 423–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arroyo, J.D.; Chevillet, J.R.; Kroh, E.M.; Ruf, I.K.; Pritchard, C.C.; Gibson, D.F.; Mitchell, P.S.; Bennett, C.F.; Pogosova-Agadjanyan, E.L.; Stirewalt, D.L.; et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc. Natl. Acad. Sci. USA 2011, 108, 5003–5008. [Google Scholar] [CrossRef] [Green Version]
- Weber, J.A.; Baxter, D.H.; Zhang, S.; Huang, D.Y.; Huang, K.H.; Lee, M.J.; Galas, D.J.; Wang, K. The microRNA spectrum in 12 body fluids. Clin. Chem. 2010, 56, 1733–1741. [Google Scholar] [CrossRef] [PubMed]
- Devaux, Y. Transcriptome of blood cells as a reservoir of cardiovascular biomarkers. Biochim. Biophys. Acta. Mol. Cell Res. 2017, 1864, 209–216. [Google Scholar] [CrossRef] [PubMed]
- Cortez, M.A.; Bueso-Ramos, C.; Ferdin, J.; Lopez-Berestein, G.; Sood, A.K.; Calin, G.A. MicroRNAs in body fluids--the mix of hormones and biomarkers. Nat. Rev. Clin. Oncol. 2011, 8, 467–477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Devaux, Y.; Vausort, M.; Goretti, E.; Nazarov, P.V.; Azuaje, F.; Gilson, G.; Corsten, M.F.; Schroen, B.; Lair, M.L.; Heymans, S.; et al. Use of circulating microRNAs to diagnose acute myocardial infarction. Clin. Chem. 2012, 58, 559–567. [Google Scholar] [CrossRef] [Green Version]
- Jansen, F.; Yang, X.; Proebsting, S.; Hoelscher, M.; Przybilla, D.; Baumann, K.; Schmitz, T.; Dolf, A.; Endl, E.; Franklin, B.S.; et al. MicroRNA expression in circulating microvesicles predicts cardiovascular events in patients with coronary artery disease. J. Am. Heart Assoc. 2014, 3, e001249. [Google Scholar] [CrossRef] [Green Version]
- Niculescu, L.S.; Simionescu, N.; Sanda, G.M.; Carnuta, M.G.; Stancu, C.S.; Popescu, A.C.; Popescu, M.R.; Vlad, A.; Dimulescu, D.R.; Simionescu, M.; et al. MiR-486 and miR-92a Identified in Circulating HDL Discriminate between Stable and Vulnerable Coronary Artery Disease Patients. PLoS ONE 2015, 10, e0140958. [Google Scholar] [CrossRef]
- Simionescu, N.; Niculescu, L.S.; Carnuta, M.G.; Sanda, G.M.; Stancu, C.S.; Popescu, A.C.; Popescu, M.R.; Vlad, A.; Dimulescu, D.R.; Simionescu, M.; et al. Hyperglycemia Determines Increased Specific MicroRNAs Levels in Sera and HDL of Acute Coronary Syndrome Patients and Stimulates MicroRNAs Production in Human Macrophages. PLoS ONE 2016, 11, e0161201. [Google Scholar] [CrossRef]
- Niculescu, L.S.; Dulceanu, M.D.; Stancu, C.S.; Carnuta, M.G.; Barbalata, T.; Sima, A.V. Probiotics administration or the high-fat diet arrest modulates microRNAs levels in hyperlipidemic hamsters. J. Funct. Foods 2019, 56, 295–302. [Google Scholar] [CrossRef]
- Afman, L.; Milenkovic, D.; Roche, H.M. Nutritional aspects of metabolic inflammation in relation to health--insights from transcriptomic biomarkers in PBMC of fatty acids and polyphenols. Mol. Nutr. Food Res. 2014, 58, 1708–1720. [Google Scholar] [CrossRef] [PubMed]
- Sobhani, M.; Farzaei, M.H.; Kiani, S.; Khodarahmi, R. Immunomodulatory; Anti-inflammatory/antioxidant Effects of Polyphenols: A Comparative Review on the Parental Compounds and Their Metabolites. Food Rev. Int. 2020, 1–53. [Google Scholar] [CrossRef]
- Subramaniam, S.; Selvaduray, K.R.; Radhakrishnan, A.K. Bioactive Compounds: Natural Defense Against Cancer? Biomolecules 2019, 9, 758. [Google Scholar] [CrossRef] [Green Version]
- Jamwal, R. Bioavailable curcumin formulations: A review of pharmacokinetic studies in healthy volunteers. J. Integr. Med. 2018, 16, 367–374. [Google Scholar] [CrossRef]
- DiSilvestro, R.A.; Joseph, E.; Zhao, S.; Bomser, J. Diverse effects of a low dose supplement of lipidated curcumin in healthy middle aged people. Nutr. J. 2012, 11, 79. [Google Scholar] [CrossRef] [Green Version]
- Mollazadeh, H.; Mahdian, D.; Hosseinzadeh, H. Medicinal plants in treatment of hypertriglyceridemia: A review based on their mechanisms and effectiveness. Phytomed. Int. J. Phytother. Phytopharmacol. 2019, 53, 43–52. [Google Scholar] [CrossRef]
- Asai, A.; Nakagawa, K.; Miyazawa, T. Antioxidative effects of turmeric, rosemary and capsicum extracts on membrane phospholipid peroxidation and liver lipid metabolism in mice. Biosci. Biotechnol. Biochem. 1999, 63, 2118–2122. [Google Scholar] [CrossRef]
- Shin, S.K.; Ha, T.Y.; McGregor, R.A.; Choi, M.S. Long-term curcumin administration protects against atherosclerosis via hepatic regulation of lipoprotein cholesterol metabolism. Mol. Nutr. Food Res. 2011, 55, 1829–1840. [Google Scholar] [CrossRef]
- Hamer, M.; O’Donovan, G.; Stamatakis, E. High-Density Lipoprotein Cholesterol and Mortality: Too Much of a Good Thing? Arterioscler. Thromb. Vasc. Biol. 2018, 38, 669–672. [Google Scholar] [CrossRef] [Green Version]
- Soetikno, V.; Sari, F.R.; Sukumaran, V.; Lakshmanan, A.P.; Mito, S.; Harima, M.; Thandavarayan, R.A.; Suzuki, K.; Nagata, M.; Takagi, R.; et al. Curcumin prevents diabetic cardiomyopathy in streptozotocin-induced diabetic rats: Possible involvement of PKC-MAPK signaling pathway. Eur. J. Pharm. Sci. 2012, 47, 604–614. [Google Scholar] [CrossRef] [PubMed]
- Yu, W.; Wu, J.; Cai, F.; Xiang, J.; Zha, W.; Fan, D.; Guo, S.; Ming, Z.; Liu, C. Curcumin alleviates diabetic cardiomyopathy in experimental diabetic rats. PLoS ONE 2012, 7, e52013. [Google Scholar] [CrossRef] [Green Version]
- Tanwar, V.; Sachdeva, J.; Kishore, K.; Mittal, R.; Nag, T.C.; Ray, R.; Kumari, S.; Arya, D.S. Dose-dependent actions of curcumin in experimentally induced myocardial necrosis: A biochemical, histopathological, and electron microscopic evidence. Cell Biochem. Funct. 2010, 28, 74–82. [Google Scholar] [CrossRef] [PubMed]
- Nazam Ansari, M.; Bhandari, U.; Pillai, K.K. Protective role of curcumin in myocardial oxidative damage induced by isoproterenol in rats. Hum. Exp. Toxicol. 2007, 26, 933–938. [Google Scholar] [CrossRef] [PubMed]
- Soares, M.P.; Seldon, M.P.; Gregoire, I.P.; Vassilevskaia, T.; Berberat, P.O.; Yu, J.; Tsui, T.Y.; Bach, F.H. Heme oxygenase-1 modulates the expression of adhesion molecules associated with endothelial cell activation. J. Immunol. 2004, 172, 3553–3563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Olszanecki, R.; Gebska, A.; Korbut, R. The role of haem oxygenase-1 in the decrease of endothelial intercellular adhesion molecule-1 expression by curcumin. Basic Clin. Pharmacol. Toxicol. 2007, 101, 411–415. [Google Scholar] [CrossRef]
- Nabavi, S.F.; Barber, A.J.; Spagnuolo, C.; Russo, G.L.; Daglia, M.; Nabavi, S.M.; Sobarzo-Sanchez, E. Nrf2 as molecular target for polyphenols: A novel therapeutic strategy in diabetic retinopathy. Crit. Rev. Clin. Lab. Sci. 2016, 53, 293–312. [Google Scholar] [CrossRef]
- Kang, E.S.; Woo, I.S.; Kim, H.J.; Eun, S.Y.; Paek, K.S.; Kim, H.J.; Chang, K.C.; Lee, J.H.; Lee, H.T.; Kim, J.H.; et al. Up-regulation of aldose reductase expression mediated by phosphatidylinositol 3-kinase/Akt and Nrf2 is involved in the protective effect of curcumin against oxidative damage. Free Radic. Biol. Med. 2007, 43, 535–545. [Google Scholar] [CrossRef]
- Yang, H.; Xu, W.; Zhou, Z.; Liu, J.; Li, X.; Chen, L.; Weng, J.; Yu, Z. Curcumin attenuates urinary excretion of albumin in type II diabetic patients with enhancing nuclear factor erythroid-derived 2-like 2 (Nrf2) system and repressing inflammatory signaling efficacies. Exp. Clin. Endocrinol. Diabetes 2015, 123, 360–367. [Google Scholar] [CrossRef] [Green Version]
- Wongeakin, N.; Bhattarakosol, P.; Patumraj, S. Molecular mechanisms of curcumin on diabetes-induced endothelial dysfunctions: Txnip, ICAM-1, and NOX2 expressions. BioMed Res. Int. 2014, 2014, 161346. [Google Scholar] [CrossRef]
- Zeng, C.; Zhong, P.; Zhao, Y.; Kanchana, K.; Zhang, Y.; Khan, Z.A.; Chakrabarti, S.; Wu, L.; Wang, J.; Liang, G. Curcumin protects hearts from FFA-induced injury by activating Nrf2 and inactivating NF-kappaB both in vitro and in vivo. J. Mol. Cell. Cardiol. 2015, 79, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Coban, D.; Milenkovic, D.; Chanet, A.; Khallou-Laschet, J.; Sabbe, L.; Palagani, A.; Vanden Berghe, W.; Mazur, A.; Morand, C. Dietary curcumin inhibits atherosclerosis by affecting the expression of genes involved in leukocyte adhesion and transendothelial migration. Mol. Nutr. Food Res. 2012, 56, 1270–1281. [Google Scholar] [CrossRef] [PubMed]
- Pirvulescu, M.M.; Gan, A.M.; Stan, D.; Simion, V.; Calin, M.; Butoi, E.; Tirgoviste, C.I.; Manduteanu, I. Curcumin and a Morus alba extract reduce pro-inflammatory effects of resistin in human endothelial cells. Phytother. Res. PTR 2011, 25, 1737–1742. [Google Scholar] [CrossRef] [PubMed]
- Monfoulet, L.E.; Mercier, S.; Bayle, D.; Tamaian, R.; Barber-Chamoux, N.; Morand, C.; Milenkovic, D. Curcumin modulates endothelial permeability and monocyte transendothelial migration by affecting endothelial cell dynamics. Free Radic. Biol. Med. 2017, 112, 109–120. [Google Scholar] [CrossRef] [PubMed]
- Karimian, M.S.; Pirro, M.; Majeed, M.; Sahebkar, A. Curcumin as a natural regulator of monocyte chemoattractant protein-1. Cytokine Growth Factor Rev. 2017, 33, 55–63. [Google Scholar] [CrossRef]
- Hao, Q.; Chen, X.; Wang, X.; Dong, B.; Yang, C. Curcumin Attenuates Angiotensin II-Induced Abdominal Aortic Aneurysm by Inhibition of Inflammatory Response and ERK Signaling Pathways. Evid.-Based Complement. Altern. Med. eCAM 2014, 2014, 270930. [Google Scholar] [CrossRef]
- Wang, J.; Dong, S. ICAM-1 and IL-8 are expressed by DEHP and suppressed by curcumin through ERK and p38 MAPK in human umbilical vein endothelial cells. Inflammation 2012, 35, 859–870. [Google Scholar] [CrossRef]
- Tsai, I.J.; Chen, C.W.; Tsai, S.Y.; Wang, P.Y.; Owaga, E.; Hsieh, R.H. Curcumin supplementation ameliorated vascular dysfunction and improved antioxidant status in rats fed a high-sucrose, high-fat diet. Appl. Physiol. Nutr. Metab. 2018, 43, 669–676. [Google Scholar] [CrossRef]
- Han, Y.; Sun, H.J.; Tong, Y.; Chen, Y.Z.; Ye, C.; Qiu, Y.; Zhang, F.; Chen, A.D.; Qi, X.H.; Chen, Q.; et al. Curcumin attenuates migration of vascular smooth muscle cells via inhibiting NFkappaB-mediated NLRP3 expression in spontaneously hypertensive rats. J. Nutr. Biochem. 2019, 72, 108212. [Google Scholar] [CrossRef]
- Sun, H.J.; Ren, X.S.; Xiong, X.Q.; Chen, Y.Z.; Zhao, M.X.; Wang, J.J.; Zhou, Y.B.; Han, Y.; Chen, Q.; Li, Y.H.; et al. NLRP3 inflammasome activation contributes to VSMC phenotypic transformation and proliferation in hypertension. Cell Death Dis. 2017, 8, e3074. [Google Scholar] [CrossRef] [Green Version]
- Yin, H.; Guo, Q.; Li, X.; Tang, T.; Li, C.; Wang, H.; Sun, Y.; Feng, Q.; Ma, C.; Gao, C.; et al. Curcumin Suppresses IL-1beta Secretion and Prevents Inflammation through Inhibition of the NLRP3 Inflammasome. J. Immunol. 2018, 200, 2835–2846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, F.; Guo, N.; Cao, G.; Zhou, J.; Yuan, Z. Molecular analysis of curcumin-induced polarization of murine RAW264.7 macrophages. J. Cardiovasc. Pharmacol. 2014, 63, 544–552. [Google Scholar] [CrossRef] [PubMed]
- Chen, F.Y.; Zhou, J.; Guo, N.; Ma, W.G.; Huang, X.; Wang, H.; Yuan, Z.Y. Curcumin retunes cholesterol transport homeostasis and inflammation response in M1 macrophage to prevent atherosclerosis. Biochem. Biophys. Res. Commun. 2015, 467, 872–878. [Google Scholar] [CrossRef] [PubMed]
- Miao, Y.; Zhao, S.; Gao, Y.; Wang, R.; Wu, Q.; Wu, H.; Luo, T. Curcumin pretreatment attenuates inflammation and mitochondrial dysfunction in experimental stroke: The possible role of Sirt1 signaling. Brain Res. Bull. 2016, 121, 9–15. [Google Scholar] [CrossRef] [PubMed]
- Wu, Y.; Xu, J.; Xu, J.; Zheng, W.; Chen, Q.; Jiao, D. Study on the mechanism of JAK2/STAT3 signaling pathway-mediated inflammatory reaction after cerebral ischemia. Mol. Med. Rep. 2018, 17, 5007–5012. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Suwanwela, N.C.; Patumraj, S. Curcumin by down-regulating NF-kB and elevating Nrf2, reduces brain edema and neurological dysfunction after cerebral I/R. Microvasc. Res. 2016, 106, 117–127. [Google Scholar] [CrossRef]
- Tu, X.K.; Yang, W.Z.; Chen, J.P.; Chen, Y.; Ouyang, L.Q.; Xu, Y.C.; Shi, S.S. Curcumin inhibits TLR2/4-NF-kappaB signaling pathway and attenuates brain damage in permanent focal cerebral ischemia in rats. Inflammation 2014, 37, 1544–1551. [Google Scholar] [CrossRef] [PubMed]
- Hatcher, H.; Planalp, R.; Cho, J.; Torti, F.M.; Torti, S.V. Curcumin: From ancient medicine to current clinical trials. Cell. Mol. Life Sci. CMLS 2008, 65, 1631–1652. [Google Scholar] [CrossRef]
- Lin, K.; Chen, H.; Chen, X.; Qian, J.; Huang, S.; Huang, W. Efficacy of Curcumin on Aortic Atherosclerosis: A Systematic Review and Meta-Analysis in Mouse Studies and Insights into Possible Mechanisms. Oxid. Med. Cell. Longev. 2020, 2020, 1520747. [Google Scholar] [CrossRef]
- Mirzabeigi, P.; Mohammadpour, A.H.; Salarifar, M.; Gholami, K.; Mojtahedzadeh, M.; Javadi, M.R. The Effect of Curcumin on some of Traditional and Non-traditional Cardiovascular Risk Factors: A Pilot Randomized, Double-blind, Placebo-controlled Trial. Iran. J. Pharm. Res. IJPR 2015, 14, 479–486. [Google Scholar]
- Baum, L.; Cheung, S.K.; Mok, V.C.; Lam, L.C.; Leung, V.P.; Hui, E.; Ng, C.C.; Chow, M.; Ho, P.C.; Lam, S.; et al. Curcumin effects on blood lipid profile in a 6-month human study. Pharmacol. Res. 2007, 56, 509–514. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.S.; Su, Y.F.; Yang, H.W.; Lee, Y.H.; Chou, J.I.; Ueng, K.C. Lipid-lowering effects of curcumin in patients with metabolic syndrome: A randomized, double-blind, placebo-controlled trial. Phytother. Res. PTR 2014, 28, 1770–1777. [Google Scholar] [CrossRef]
- Panahi, Y.; Ghanei, M.; Bashiri, S.; Hajihashemi, A.; Sahebkar, A. Short-term Curcuminoid Supplementation for Chronic Pulmonary Complications due to Sulfur Mustard Intoxication: Positive Results of a Randomized Double-blind Placebo-controlled Trial. Drug Res. 2015, 65, 567–573. [Google Scholar] [CrossRef]
- Simion, V.; Stan, D.; Constantinescu, C.A.; Deleanu, M.; Dragan, E.; Tucureanu, M.M.; Gan, A.M.; Butoi, E.; Constantin, A.; Manduteanu, I.; et al. Conjugation of curcumin-loaded lipid nanoemulsions with cell-penetrating peptides increases their cellular uptake and enhances the anti-inflammatory effects in endothelial cells. J. Pharm. Pharmacol. 2016, 68, 195–207. [Google Scholar] [CrossRef]
- Ma, F.; Liu, F.; Ding, L.; You, M.; Yue, H.; Zhou, Y.; Hou, Y. Anti-inflammatory effects of curcumin are associated with down regulating microRNA-155 in LPS-treated macrophages and mice. Pharm. Biol. 2017, 55, 1263–1273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, J.; Wang, Q.; Rao, G.; Qiu, J.; He, R. Curcumin improves perfusion recovery in experimental peripheral arterial disease by upregulating microRNA-93 expression. Exp. Ther. Med. 2019, 17, 798–802. [Google Scholar] [CrossRef] [PubMed]
- Geng, H.H.; Li, R.; Su, Y.M.; Xiao, J.; Pan, M.; Cai, X.X.; Ji, X.P. Curcumin protects cardiac myocyte against hypoxia-induced apoptosis through upregulating miR-7a/b expression. Biomed. Pharmacother. 2016, 81, 258–264. [Google Scholar] [CrossRef]
- Tian, L.; Song, Z.; Shao, W.; Du, W.W.; Zhao, L.R.; Zeng, K.; Yang, B.B.; Jin, T. Curcumin represses mouse 3T3-L1 cell adipogenic differentiation via inhibiting miR-17-5p and stimulating the Wnt signalling pathway effector Tcf7l2. Cell Death Dis. 2017, 8, e2559. [Google Scholar] [CrossRef] [Green Version]
- El-Seedi, H.R.; El-Said, A.M.; Khalifa, S.A.; Goransson, U.; Bohlin, L.; Borg-Karlson, A.K.; Verpoorte, R. Biosynthesis, natural sources, dietary intake, pharmacokinetic properties, and biological activities of hydroxycinnamic acids. J. Agric. Food Chem. 2012, 60, 10877–10895. [Google Scholar] [CrossRef]
- David, I.G.; Bizgan, A.M.; Popa, D.E.; Buleandra, M.; Moldovan, Z.; Badea, I.A.; Tekiner, T.A.; Basaga, H.; Ciucu, A.A. Rapid determination of total polyphenolic content in tea samples based on caffeic acid voltammetric behaviour on a disposable graphite electrode. Food Chem. 2015, 173, 1059–1065. [Google Scholar] [CrossRef]
- Rebelo, M.J.; Rego, R.; Ferreira, M.; Oliveira, M.C. Comparative study of the antioxidant capacity and polyphenol content of Douro wines by chemical and electrochemical methods. Food Chem. 2013, 141, 566–573. [Google Scholar] [CrossRef] [PubMed]
- Garrido, E.; Cerqueira, A.S.; Chavarria, D.; Silva, T.; Borges, F.; Garrido, J. Microencapsulation of caffeic acid phenethyl ester and caffeic acid phenethyl amide by inclusion in hydroxypropyl-beta-cyclodextrin. Food Chem. 2018, 254, 260–265. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.J.; Zeng, J.; Yang, B.K.; Zhong, Y.M. Bioavailability of caffeic acid in rats and its absorption properties in the Caco-2 cell model. Pharm. Biol. 2014, 52, 1150–1157. [Google Scholar] [CrossRef] [PubMed]
- Moya, C.; Manez, S. Paraoxonases: Metabolic role and pharmacological projection. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2018, 391, 349–359. [Google Scholar] [CrossRef]
- Wang, X.; Stavchansky, S.; Zhao, B.; Bynum, J.A.; Kerwin, S.M.; Bowman, P.D. Cytoprotection of human endothelial cells from menadione cytotoxicity by caffeic acid phenethyl ester: The role of heme oxygenase-1. Eur. J. Pharmacol. 2008, 591, 28–35. [Google Scholar] [CrossRef]
- Kim, J.K.; Jang, H.D. Nrf2-mediated HO-1 induction coupled with the ERK signaling pathway contributes to indirect antioxidant capacity of caffeic acid phenethyl ester in HepG2 cells. Int. J. Mol. Sci. 2014, 15, 12149–12165. [Google Scholar] [CrossRef] [Green Version]
- Dinkova-Kostova, A.T.; Holtzclaw, W.D.; Cole, R.N.; Itoh, K.; Wakabayashi, N.; Katoh, Y.; Yamamoto, M.; Talalay, P. Direct evidence that sulfhydryl groups of Keap1 are the sensors regulating induction of phase 2 enzymes that protect against carcinogens and oxidants. Proc. Natl. Acad. Sci. USA 2002, 99, 11908–11913. [Google Scholar] [CrossRef] [Green Version]
- Taylan, M.; Kaya, H.; Demir, M.; Evliyaoglu, O.; Sen, H.S.; Firat, U.; Keles, A.; Yilmaz, S.; Sezgi, C. The Protective Effects of Caffeic Acid Phenethyl Ester on Acetylsalicylic Acid-induced Lung Injury in Rats. J. Investig. Surg. 2016, 29, 328–334. [Google Scholar] [CrossRef]
- Moon, M.K.; Lee, Y.J.; Kim, J.S.; Kang, D.G.; Lee, H.S. Effect of caffeic acid on tumor necrosis factor-alpha-induced vascular inflammation in human umbilical vein endothelial cells. Biol. Pharm. Bull. 2009, 32, 1371–1377. [Google Scholar] [CrossRef] [Green Version]
- Toma, L.; Sanda, G.M.; Niculescu, L.S.; Deleanu, M.; Stancu, C.S.; Sima, A.V. Caffeic acid attenuates the inflammatory stress induced by glycated LDL in human endothelial cells by mechanisms involving inhibition of AGE-receptor, oxidative, and endoplasmic reticulum stress. BioFactors 2017, 43, 685–697. [Google Scholar] [CrossRef]
- Ibitoye, O.B.; Ajiboye, T.O. Dietary phenolic acids reverse insulin resistance, hyperglycaemia, dyslipidaemia, inflammation and oxidative stress in high-fructose diet-induced metabolic syndrome rats. Arch. Physiol. Biochem. 2018, 124, 410–417. [Google Scholar] [CrossRef] [PubMed]
- Shahbazi, S.; Sahrawat, T.R.; Ray, M.; Dash, S.; Kar, D.; Singh, S. Drug Targets for Cardiovascular-Safe Anti-Inflammatory: In Silico Rational Drug Studies. PLoS ONE 2016, 11, e0156156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Murase, T.; Misawa, K.; Minegishi, Y.; Aoki, M.; Ominami, H.; Suzuki, Y.; Shibuya, Y.; Hase, T. Coffee polyphenols suppress diet-induced body fat accumulation by downregulating SREBP-1c and related molecules in C57BL/6J mice. Am. J. Physiol. Endocrinol. Metab. 2011, 300, E122–E133. [Google Scholar] [CrossRef] [Green Version]
- Matboli, M.; Eissa, S.; Ibrahim, D.; Hegazy, M.G.A.; Imam, S.S.; Habib, E.K. Caffeic Acid Attenuates Diabetic Kidney Disease via Modulation of Autophagy in a High-Fat Diet/Streptozotocin- Induced Diabetic Rat. Sci. Rep. 2017, 7, 2263. [Google Scholar] [CrossRef]
- Hou, C.Y.; Tain, Y.L.; Yu, H.R.; Huang, L.T. The Effects of Resveratrol in the Treatment of Metabolic Syndrome. Int. J. Mol. Sci. 2019, 20, 535. [Google Scholar] [CrossRef] [Green Version]
- Amri, A.; Chaumeil, J.C.; Sfar, S.; Charrueau, C. Administration of resveratrol: What formulation solutions to bioavailability limitations? J. Controll. Release 2012, 158, 182–193. [Google Scholar] [CrossRef]
- Chimento, A.; De Amicis, F.; Sirianni, R.; Sinicropi, M.S.; Puoci, F.; Casaburi, I.; Saturnino, C.; Pezzi, V. Progress to Improve Oral Bioavailability and Beneficial Effects of Resveratrol. Int. J. Mol. Sci. 2019, 20, 1381. [Google Scholar] [CrossRef] [Green Version]
- Cho, I.J.; Ahn, J.Y.; Kim, S.; Choi, M.S.; Ha, T.Y. Resveratrol attenuates the expression of HMG-CoA reductase mRNA in hamsters. Biochem. Biophys. Res. Commun. 2008, 367, 190–194. [Google Scholar] [CrossRef]
- Kumar, B.J.; Joghee, N. Resveratrol supplementation in patients with type 2 diabetes mellitus: A prospective, open label, randomized controlled trial. Int. Res. J. Pharm. 2013, 4, 245–249. [Google Scholar] [CrossRef]
- Movahed, A.; Nabipour, I.; Lieben Louis, X.; Thandapilly, S.J.; Yu, L.; Kalantarhormozi, M.; Rekabpour, S.J.; Netticadan, T. Antihyperglycemic effects of short term resveratrol supplementation in type 2 diabetic patients. Evid.-Based Complement. Altern. Med. eCAM 2013, 2013, 851267. [Google Scholar] [CrossRef] [Green Version]
- Sahebkar, A. Effects of resveratrol supplementation on plasma lipids: A systematic review and meta-analysis of randomized controlled trials. Nutr. Rev. 2013, 71, 822–835. [Google Scholar] [CrossRef] [PubMed]
- Luca, S.V.; Macovei, I.; Bujor, A.; Miron, A.; Skalicka-Wozniak, K.; Aprotosoaie, A.C.; Trifan, A. Bioactivity of dietary polyphenols: The role of metabolites. Crit. Rev. Food Sci. Nutr. 2020, 60, 626–659. [Google Scholar] [CrossRef] [PubMed]
- Gong, M.; Garige, M.; Varatharajalu, R.; Marmillot, P.; Gottipatti, C.; Leckey, L.C.; Lakshman, R.M. Quercetin up-regulates paraoxonase 1 gene expression with concomitant protection against LDL oxidation. Biochem. Biophys. Res. Commun. 2009, 379, 1001–1004. [Google Scholar] [CrossRef] [PubMed]
- Noll, C.; Hamelet, J.; Matulewicz, E.; Paul, J.L.; Delabar, J.M.; Janel, N. Effects of red wine polyphenolic compounds on paraoxonase-1 and lectin-like oxidized low-density lipoprotein receptor-1 in hyperhomocysteinemic mice. J. Nutr. Biochem. 2009, 20, 586–596. [Google Scholar] [CrossRef] [PubMed]
- Garige, M.; Gong, M.; Varatharajalu, R.; Lakshman, M.R. Quercetin up-regulates paraoxonase 1 gene expression via sterol regulatory element binding protein 2 that translocates from the endoplasmic reticulum to the nucleus where it specifically interacts with sterol responsive element-like sequence in paraoxonase 1 promoter in HuH7 liver cells. Metab. Clin. Exp. 2010, 59, 1372–1378. [Google Scholar] [CrossRef] [Green Version]
- Barbalata, T.; Deleanu, M.; Carnuta, M.G.; Niculescu, L.S.; Raileanu, M.; Sima, A.V.; Stancu, C.S. Hyperlipidemia Determines Dysfunctional HDL Production and Impedes Cholesterol Efflux in the Small Intestine: Alleviation by Ginger Extract. Mol. Nutr. Food Res. 2019, 63, e1900029. [Google Scholar] [CrossRef]
- Guo, R.; Su, Y.; Liu, B.; Li, S.; Zhou, S.; Xu, Y. Resveratrol suppresses oxidised low-density lipoprotein-induced macrophage apoptosis through inhibition of intracellular reactive oxygen species generation, LOX-1, and the p38 MAPK pathway. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2014, 34, 603–616. [Google Scholar] [CrossRef]
- Bonnefont-Rousselot, D. Resveratrol and Cardiovascular Diseases. Nutrients 2016, 8, 250. [Google Scholar] [CrossRef]
- Wicinski, M.; Socha, M.; Walczak, M.; Wodkiewicz, E.; Malinowski, B.; Rewerski, S.; Gorski, K.; Pawlak-Osinska, K. Beneficial Effects of Resveratrol Administration-Focus on Potential Biochemical Mechanisms in Cardiovascular Conditions. Nutrients 2018, 10, 1813. [Google Scholar] [CrossRef] [Green Version]
- Borra, M.T.; Smith, B.C.; Denu, J.M. Mechanism of human SIRT1 activation by resveratrol. J. Biol. Chem. 2005, 280, 17187–17195. [Google Scholar] [CrossRef] [Green Version]
- Yeung, F.; Hoberg, J.E.; Ramsey, C.S.; Keller, M.D.; Jones, D.R.; Frye, R.A.; Mayo, M.W. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J. 2004, 23, 2369–2380. [Google Scholar] [CrossRef] [Green Version]
- Bonizzi, G.; Karin, M. The two NF-kappaB activation pathways and their role in innate and adaptive immunity. Trends Immunol. 2004, 25, 280–288. [Google Scholar] [CrossRef]
- Malaguarnera, L. Influence of Resveratrol on the Immune Response. Nutrients 2019, 11, 946. [Google Scholar] [CrossRef] [Green Version]
- Shakibaei, M.; Buhrmann, C.; Mobasheri, A. Resveratrol-mediated SIRT-1 interactions with p300 modulate receptor activator of NF-kappaB ligand (RANKL) activation of NF-kappaB signaling and inhibit osteoclastogenesis in bone-derived cells. J. Biol. Chem. 2011, 286, 11492–11505. [Google Scholar] [CrossRef] [Green Version]
- Price, N.L.; Gomes, A.P.; Ling, A.J.; Duarte, F.V.; Martin-Montalvo, A.; North, B.J.; Agarwal, B.; Ye, L.; Ramadori, G.; Teodoro, J.S.; et al. SIRT1 is required for AMPK activation and the beneficial effects of resveratrol on mitochondrial function. Cell Metab. 2012, 15, 675–690. [Google Scholar] [CrossRef] [Green Version]
- Theodotou, M.; Fokianos, K.; Mouzouridou, A.; Konstantinou, C.; Aristotelous, A.; Prodromou, D.; Chrysikou, A. The effect of resveratrol on hypertension: A clinical trial. Exp. Ther. Med. 2017, 13, 295–301. [Google Scholar] [CrossRef] [Green Version]
- Chen, J.; Cao, X.; Cui, Y.; Zeng, G.; Chen, J.; Zhang, G. Resveratrol alleviates lysophosphatidylcholine-induced damage and inflammation in vascular endothelial cells. Mol. Med. Rep. 2018, 17, 4011–4018. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Z.; Chen, N.; Liu, J.B.; Wu, J.B.; Zhang, J.; Zhang, Y.; Jiang, X. Protective effect of resveratrol against acute lung injury induced by lipopolysaccharide via inhibiting the myd88-dependent Toll-like receptor 4 signaling pathway. Mol. Med. Rep. 2014, 10, 101–106. [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-alpha-treated endothelial cells: Evidence for an anti-inflammatory cascade mediated by the miR-221/222/AMPK/p38/NF-kappaB pathway. Sci. Rep. 2017, 7, 44689. [Google Scholar] [CrossRef]
- Lin, J.W.; Yang, L.H.; Ren, Z.C.; Mu, D.G.; Li, Y.Q.; Yan, J.P.; Wang, L.X.; Chen, C. Resveratrol downregulates TNF-alpha-induced monocyte chemoattractant protein-1 in primary rat pulmonary artery endothelial cells by P38 mitogen-activated protein kinase signaling. Drug Des. Dev. Ther. 2019, 13, 1843–1853. [Google Scholar] [CrossRef] [Green Version]
- Pan, W.; Yu, H.; Huang, S.; Zhu, P. Resveratrol Protects against TNF-alpha-Induced Injury in Human Umbilical Endothelial Cells through Promoting Sirtuin-1-Induced Repression of NF-KB and p38 MAPK. PLoS ONE 2016, 11, e0147034. [Google Scholar] [CrossRef] [Green Version]
- Chen, M.L.; Yi, L.; Jin, X.; Liang, X.Y.; Zhou, Y.; Zhang, T.; Xie, Q.; Zhou, X.; Chang, H.; Fu, Y.J.; et al. Resveratrol attenuates vascular endothelial inflammation by inducing autophagy through the cAMP signaling pathway. Autophagy 2013, 9, 2033–2045. [Google Scholar] [CrossRef]
- Park, D.W.; Baek, K.; Kim, J.R.; Lee, J.J.; Ryu, S.H.; Chin, B.R.; Baek, S.H. Resveratrol inhibits foam cell formation via NADPH oxidase 1- mediated reactive oxygen species and monocyte chemotactic protein-1. Exp. Mol. Med. 2009, 41, 171–179. [Google Scholar] [CrossRef]
- Yang, Y.; Li, S.; Yang, Q.; Shi, Y.; Zheng, M.; Liu, Y.; Chen, F.; Song, G.; Xu, H.; Wan, T.; et al. Resveratrol reduces the proinflammatory effects and lipopolysaccharide- induced expression of HMGB1 and TLR4 in RAW264.7 cells. Cell. Phys. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2014, 33, 1283–1292. [Google Scholar] [CrossRef]
- Jakus, P.B.; Kalman, N.; Antus, C.; Radnai, B.; Tucsek, Z.; Gallyas, F., Jr.; Sumegi, B.; Veres, B. TRAF6 is functional in inhibition of TLR4-mediated NF-kappaB activation by resveratrol. J. Nutr. Biochem. 2013, 24, 819–823. [Google Scholar] [CrossRef]
- Inanaga, K.; Ichiki, T.; Matsuura, H.; Miyazaki, R.; Hashimoto, T.; Takeda, K.; Sunagawa, K. Resveratrol attenuates angiotensin II-induced interleukin-6 expression and perivascular fibrosis. Hypertens. Res. 2009, 32, 466–471. [Google Scholar] [CrossRef]
- Zhang, J.; Chen, J.; Yang, J.; Xu, C.W.; Pu, P.; Ding, J.W.; Jiang, H. Resveratrol attenuates oxidative stress induced by balloon injury in the rat carotid artery through actions on the ERK1/2 and NF-kappa B pathway. Cell. Phys. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2013, 31, 230–241. [Google Scholar] [CrossRef]
- Deng, Z.Y.; Hu, M.M.; Xin, Y.F.; Gang, C. Resveratrol alleviates vascular inflammatory injury by inhibiting inflammasome activation in rats with hypercholesterolemia and vitamin D2 treatment. Inflamm. Res. 2015, 64, 321–332. [Google Scholar] [CrossRef]
- Chang, G.R.; Chen, P.L.; Hou, P.H.; Mao, F.C. Resveratrol protects against diet-induced atherosclerosis by reducing low-density lipoprotein cholesterol and inhibiting inflammation in apolipoprotein E-deficient mice. Iran. J. Basic Med. Sci. 2015, 18, 1063–1071. [Google Scholar]
- Cong, X.; Li, Y.; Lu, N.; Dai, Y.; Zhang, H.; Zhao, X.; Liu, Y. Resveratrol attenuates the inflammatory reaction induced by ischemia/reperfusion in the rat heart. Mol. Med. Rep. 2014, 9, 2528–2532. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Xie, C.; Zhuang, J.; Li, H.; Yao, Y.; Shao, C.; Wang, H. Resveratrol attenuates inflammation in the rat heart subjected to ischemia-reperfusion: Role of the TLR4/NF-kappaB signaling pathway. Mol. Med. Rep. 2015, 11, 1120–1126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Espinoza, J.L.; Trung, L.Q.; Inaoka, P.T.; Yamada, K.; An, D.T.; Mizuno, S.; Nakao, S.; Takami, A. The Repeated Administration of Resveratrol Has Measurable Effects on Circulating T-Cell Subsets in Humans. Oxid. Med. Cell. Longev. 2017, 2017, 6781872. [Google Scholar] [CrossRef] [PubMed]
- Yoshino, J.; Conte, C.; Fontana, L.; Mittendorfer, B.; Imai, S.; Schechtman, K.B.; Gu, C.; Kunz, I.; Rossi Fanelli, F.; Patterson, B.W.; et al. Resveratrol supplementation does not improve metabolic function in nonobese women with normal glucose tolerance. Cell Metab. 2012, 16, 658–664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ramirez-Garza, S.L.; Laveriano-Santos, E.P.; Marhuenda-Munoz, M.; Storniolo, C.E.; Tresserra-Rimbau, A.; Vallverdu-Queralt, A.; Lamuela-Raventos, R.M. Health Effects of Resveratrol: Results from Human Intervention Trials. Nutrients 2018, 10, 1892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tili, E.; Michaille, J.J.; Adair, B.; Alder, H.; Limagne, E.; Taccioli, C.; Ferracin, M.; Delmas, D.; Latruffe, N.; Croce, C.M. Resveratrol decreases the levels of miR-155 by upregulating miR-663, a microRNA targeting JunB and JunD. Carcinogenesis 2010, 31, 1561–1566. [Google Scholar] [CrossRef] [PubMed]
- Michaille, J.J.; Piurowski, V.; Rigot, B.; Kelani, H.; Fortman, E.C.; Tili, E. MiR-663, a MicroRNA Linked with Inflammation and Cancer That Is under the Influence of Resveratrol. Medicines 2018, 5, 74. [Google Scholar] [CrossRef] [Green Version]
- Bigagli, E.; Cinci, L.; Paccosi, S.; Parenti, A.; D’Ambrosio, M.; Luceri, C. Nutritionally relevant concentrations of resveratrol and hydroxytyrosol mitigate oxidative burst of human granulocytes and monocytes and the production of pro-inflammatory mediators in LPS-stimulated RAW 264.7 macrophages. Int. Immunopharmacol. 2017, 43, 147–155. [Google Scholar] [CrossRef]
- Lancon, A.; Kaminski, J.; Tili, E.; Michaille, J.J.; Latruffe, N. Control of MicroRNA expression as a new way for resveratrol to deliver its beneficial effects. J. Agric. Food Chem. 2012, 60, 8783–8789. [Google Scholar] [CrossRef]
- Chen, J.F.; Mandel, E.M.; Thomson, J.M.; Wu, Q.; Callis, T.E.; Hammond, S.M.; Conlon, F.L.; Wang, D.Z. The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation. Nat. Genet. 2006, 38, 228–233. [Google Scholar] [CrossRef]
- Zhang, Y.; Lu, Y.; Ong’achwa, M.J.; Ge, L.; Qian, Y.; Chen, L.; Hu, X.; Li, F.; Wei, H.; Zhang, C.; et al. Resveratrol Inhibits the TGF-beta1-Induced Proliferation of Cardiac Fibroblasts and Collagen Secretion by Downregulating miR-17 in Rat. BioMed Res. Int. 2018, 2018, 8730593. [Google Scholar] [CrossRef] [Green Version]
- Mukhopadhyay, P.; Mukherjee, S.; Ahsan, K.; Bagchi, A.; Pacher, P.; Das, D.K. Restoration of altered microRNA expression in the ischemic heart with resveratrol. PLoS ONE 2010, 5, e15705. [Google Scholar] [CrossRef] [Green Version]
- Campagnolo, P.; Hong, X.; di Bernardini, E.; Smyrnias, I.; Hu, Y.; Xu, Q. Resveratrol-Induced Vascular Progenitor Differentiation towards Endothelial Lineage via MiR-21/Akt/beta-Catenin Is Protective in Vessel Graft Models. PLoS ONE 2015, 10, e0125122. [Google Scholar] [CrossRef] [Green Version]
- Tome-Carneiro, J.; Larrosa, M.; Yanez-Gascon, M.J.; Davalos, A.; Gil-Zamorano, J.; Gonzalvez, M.; Garcia-Almagro, F.J.; Ruiz Ros, J.A.; Tomas-Barberan, F.A.; Espin, J.C.; et al. One-year supplementation with a grape extract containing resveratrol modulates inflammatory-related microRNAs and cytokines expression in peripheral blood mononuclear cells of type 2 diabetes and hypertensive patients with coronary artery disease. Pharmacol. Res. 2013, 72, 69–82. [Google Scholar] [CrossRef]
- Cione, E.; La Torre, C.; Cannataro, R.; Caroleo, M.C.; Plastina, P.; Gallelli, L. Quercetin, Epigallocatechin Gallate, Curcumin, and Resveratrol: From Dietary Sources to Human MicroRNA Modulation. Molecules 2019, 25, 63. [Google Scholar] [CrossRef] [Green Version]
- Nie, J.; Zhang, L.; Zhao, G.; Du, X. Quercetin reduces atherosclerotic lesions by altering the gut microbiota and reducing atherogenic lipid metabolites. J. Appl. Microbiol. 2019, 127, 1824–1834. [Google Scholar] [CrossRef]
- Rasouli, H.; Farzaei, M.H.; Khodarahmi, R. Polyphenols and their benefits: A review. Int. J. Food Prop. 2017, 20, 1700–1741. [Google Scholar] [CrossRef] [Green Version]
- Basu, A.; Das, A.S.; Majumder, M.; Mukhopadhyay, R. Antiatherogenic Roles of Dietary Flavonoids Chrysin, Quercetin, and Luteolin. J. Cardiovasc. Pharmacol. 2016, 68, 89–96. [Google Scholar] [CrossRef]
- Kawai, Y.; Nishikawa, T.; Shiba, Y.; Saito, S.; Murota, K.; Shibata, N.; Kobayashi, M.; Kanayama, M.; Uchida, K.; Terao, J. Macrophage as a target of quercetin glucuronides in human atherosclerotic arteries: Implication in the anti-atherosclerotic mechanism of dietary flavonoids. J. Biol. Chem. 2008, 283, 9424–9434. [Google Scholar] [CrossRef] [Green Version]
- Sanchez, M.; Galisteo, M.; Vera, R.; Villar, I.C.; Zarzuelo, A.; Tamargo, J.; Perez-Vizcaino, F.; Duarte, J. Quercetin downregulates NADPH oxidase, increases eNOS activity and prevents endothelial dysfunction in spontaneously hypertensive rats. J. Hypertens. 2006, 24, 75–84. [Google Scholar] [CrossRef] [Green Version]
- Carlstrom, J.; Symons, J.D.; Wu, T.C.; Bruno, R.S.; Litwin, S.E.; Jalili, T. A quercetin supplemented diet does not prevent cardiovascular complications in spontaneously hypertensive rats. J. Nutr. 2007, 137, 628–633. [Google Scholar] [CrossRef] [PubMed]
- Duarte, J.; Galisteo, M.; Ocete, M.A.; Perez-Vizcaino, F.; Zarzuelo, A.; Tamargo, J. Effects of chronic quercetin treatment on hepatic oxidative status of spontaneously hypertensive rats. Mol. Cell. Biochem. 2001, 221, 155–160. [Google Scholar] [CrossRef]
- Min, Z.; Yangchun, L.; Yuquan, W.; Changying, Z. Quercetin inhibition of myocardial fibrosis through regulating MAPK signaling pathway via ROS. Pak. J. Pharm. Sci. 2019, 32, 1355–1359. [Google Scholar]
- Li, P.G.; Sun, L.; Han, X.; Ling, S.; Gan, W.T.; Xu, J.W. Quercetin induces rapid eNOS phosphorylation and vasodilation by an Akt-independent and PKA-dependent mechanism. Pharmacology 2012, 89, 220–228. [Google Scholar] [CrossRef] [PubMed]
- Bhaskar, S.; Sudhakaran, P.R.; Helen, A. Quercetin attenuates atherosclerotic inflammation and adhesion molecule expression by modulating TLR-NF-kappaB signaling pathway. Cell. Immunol. 2016, 310, 131–140. [Google Scholar] [CrossRef] [PubMed]
- Hung, C.H.; Chan, S.H.; Chu, P.M.; Tsai, K.L. Quercetin is a potent anti-atherosclerotic compound by activation of SIRT1 signaling under oxLDL stimulation. Mol. Nutr. Food Res. 2015, 59, 1905–1917. [Google Scholar] [CrossRef] [PubMed]
- Xiao, H.B.; Lu, X.Y.; Sun, Z.L.; Zhang, H.B. Kaempferol regulates OPN-CD44 pathway to inhibit the atherogenesis of apolipoprotein E deficient mice. Toxicol. Appl. Pharmacol. 2011, 257, 405–411. [Google Scholar] [CrossRef]
- Che, J.; Liang, B.; Zhang, Y.; Wang, Y.; Tang, J.; Shi, G. Kaempferol alleviates ox-LDL-induced apoptosis by up-regulation of autophagy via inhibiting PI3K/Akt/mTOR pathway in human endothelial cells. Cardiovasc. Pathol. 2017, 31, 57–62. [Google Scholar] [CrossRef]
- Kleemann, R.; Verschuren, L.; Morrison, M.; Zadelaar, S.; van Erk, M.J.; Wielinga, P.Y.; Kooistra, T. Anti-inflammatory, anti-proliferative and anti-atherosclerotic effects of quercetin in human in vitro and in vivo models. Atherosclerosis 2011, 218, 44–52. [Google Scholar] [CrossRef]
- Bhaskar, S.; Helen, A. Quercetin modulates toll-like receptor-mediated protein kinase signaling pathways in oxLDL-challenged human PBMCs and regulates TLR-activated atherosclerotic inflammation in hypercholesterolemic rats. Mol. Cell. Biochem. 2016, 423, 53–65. [Google Scholar] [CrossRef]
- Calabriso, N.; Scoditti, E.; Massaro, M.; Pellegrino, M.; Storelli, C.; Ingrosso, I.; Giovinazzo, G.; Carluccio, M.A. Multiple anti-inflammatory and anti-atherosclerotic properties of red wine polyphenolic extracts: Differential role of hydroxycinnamic acids, flavonols and stilbenes on endothelial inflammatory gene expression. Eur. J. Nutr. 2016, 55, 477–489. [Google Scholar] [CrossRef]
- Vera, M.; Torramade-Moix, S.; Martin-Rodriguez, S.; Cases, A.; Cruzado, J.M.; Rivera, J.; Escolar, G.; Palomo, M.; Diaz-Ricart, M. Antioxidant and Anti-Inflammatory Strategies Based on the Potentiation of Glutathione Peroxidase Activity Prevent Endothelial Dysfunction in Chronic Kidney Disease. Cell. Phys. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2018, 51, 1287–1300. [Google Scholar] [CrossRef]
- Qureshi, A.A.; Tan, X.; Reis, J.C.; Badr, M.Z.; Papasian, C.J.; Morrison, D.C.; Qureshi, N. Suppression of nitric oxide induction and pro-inflammatory cytokines by novel proteasome inhibitors in various experimental models. Lipids Health Dis. 2011, 10, 177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lu, X.L.; Zhao, C.H.; Yao, X.L.; Zhang, H. Quercetin attenuates high fructose feeding-induced atherosclerosis by suppressing inflammation and apoptosis via ROS-regulated PI3K/AKT signaling pathway. Biomed. Pharmacother. 2017, 85, 658–671. [Google Scholar] [CrossRef] [PubMed]
- Li, B.; Yang, M.; Liu, J.W.; Yin, G.T. Protective mechanism of quercetin on acute myocardial infarction in rats. Genet. Mol. Res. GMR 2016, 15, 15017117. [Google Scholar] [CrossRef]
- Zahedi, M.; Ghiasvand, R.; Feizi, A.; Asgari, G.; Darvish, L. Does Quercetin Improve Cardiovascular Risk factors and Inflammatory Biomarkers in Women with Type 2 Diabetes: A Double-blind Randomized Controlled Clinical Trial. Int. J. Prev. Med. 2013, 4, 777–785. [Google Scholar] [PubMed]
- Chekalina, N.; Burmak, Y.; Petrov, Y.; Borisova, Z.; Manusha, Y.; Kazakov, Y.; Kaidashev, I. Quercetin reduces the transcriptional activity of NF-kB in stable coronary artery disease. Indian Heart J. 2018, 70, 593–597. [Google Scholar] [CrossRef] [PubMed]
- Dabeek, W.M.; Marra, M.V. Dietary Quercetin and Kaempferol: Bioavailability and Potential Cardiovascular-Related Bioactivity in Humans. Nutrients 2019, 11, 2288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boesch-Saadatmandi, C.; Loboda, A.; Wagner, A.E.; Stachurska, A.; Jozkowicz, A.; Dulak, J.; Doring, F.; Wolffram, S.; Rimbach, G. Effect of quercetin and its metabolites isorhamnetin and quercetin-3-glucuronide on inflammatory gene expression: Role of miR-155. J. Nutr. Biochem. 2011, 22, 293–299. [Google Scholar] [CrossRef] [PubMed]
- Boesch-Saadatmandi, C.; Wagner, A.E.; Wolffram, S.; Rimbach, G. Effect of quercetin on inflammatory gene expression in mice liver in vivo—Role of redox factor 1, miRNA-122 and miRNA-125b. Pharmacol. Res. 2012, 65, 523–530. [Google Scholar] [CrossRef]
- Wang, M.; Firrman, J.; Liu, L.; Yam, K. A Review on Flavonoid Apigenin: Dietary Intake, ADME, Antimicrobial Effects, and Interactions with Human Gut Microbiota. BioMed Res. Int. 2019, 2019, 7010467. [Google Scholar] [CrossRef]
- Tang, D.; Chen, K.; Huang, L.; Li, J. Pharmacokinetic properties and drug interactions of apigenin, a natural flavone. Expert Opin. Drug Metab. Toxicol. 2017, 13, 323–330. [Google Scholar] [CrossRef] [PubMed]
- Zhang, K.; Song, W.; Li, D.; Jin, X. Apigenin in the regulation of cholesterol metabolism and protection of blood vessels. Exp. Ther. Med. 2017, 13, 1719–1724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jung, U.J.; Cho, Y.Y.; Choi, M.S. Apigenin Ameliorates Dyslipidemia, Hepatic Steatosis and Insulin Resistance by Modulating Metabolic and Transcriptional Profiles in the Liver of High-Fat Diet-Induced Obese Mice. Nutrients 2016, 8, 305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qin, W.; Ren, B.; Wang, S.; Liang, S.; He, B.; Shi, X.; Wang, L.; Liang, J.; Wu, F. Apigenin and naringenin ameliorate PKCbetaII-associated endothelial dysfunction via regulating ROS/caspase-3 and NO pathway in endothelial cells exposed to high glucose. Vasc. Pharmacol. 2016, 85, 39–49. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.C.; Ke, W.H.; Ceng, L.H.; Hsieh, C.W.; Wung, B.S. Calcium- and phosphatidylinositol 3-kinase/Akt-dependent activation of endothelial nitric oxide synthase by apigenin. Life Scie. 2010, 87, 743–749. [Google Scholar] [CrossRef]
- Pothineni, N.V.K.; Karathanasis, S.K.; Ding, Z.; Arulandu, A.; Varughese, K.I.; Mehta, J.L. LOX-1 in Atherosclerosis and Myocardial Ischemia: Biology, Genetics, and Modulation. J. Am. Coll. Cardiol. 2017, 69, 2759–2768. [Google Scholar] [CrossRef]
- Wang, J.; Liao, Y.; Fan, J.; Ye, T.; Sun, X.; Dong, S. Apigenin inhibits the expression of IL-6, IL-8, and ICAM-1 in DEHP-stimulated human umbilical vein endothelial cells and in vivo. Inflammation 2012, 35, 1466–1476. [Google Scholar] [CrossRef]
- Ren, B.; Qin, W.; Wu, F.; Wang, S.; Pan, C.; Wang, L.; Zeng, B.; Ma, S.; Liang, J. Apigenin and naringenin regulate glucose and lipid metabolism, and ameliorate vascular dysfunction in type 2 diabetic rats. Eur. J. Pharmacol. 2016, 773, 13–23. [Google Scholar] [CrossRef]
- Mueller, M.; Hobiger, S.; Jungbauer, A. Anti-inflammatory activity of extracts from fruits, herbs and spices. Food Chem. 2010, 122, 987–996. [Google Scholar] [CrossRef]
- Comalada, M.; Ballester, I.; Bailon, E.; Sierra, S.; Xaus, J.; Galvez, J.; de Medina, F.S.; Zarzuelo, A. Inhibition of pro-inflammatory markers in primary bone marrow-derived mouse macrophages by naturally occurring flavonoids: Analysis of the structure-activity relationship. Biochem. Pharmacol. 2006, 72, 1010–1021. [Google Scholar] [CrossRef]
- Ren, K.; Jiang, T.; Zhou, H.F.; Liang, Y.; Zhao, G.J. Apigenin Retards Atherogenesis by Promoting ABCA1-Mediated Cholesterol Efflux and Suppressing Inflammation. Cell. Phys. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2018, 47, 2170–2184. [Google Scholar] [CrossRef] [PubMed]
- Yamagata, K.; Miyashita, A.; Matsufuji, H.; Chino, M. Dietary flavonoid apigenin inhibits high glucose and tumor necrosis factor alpha-induced adhesion molecule expression in human endothelial cells. J. Nutr. Biochem. 2010, 21, 116–124. [Google Scholar] [CrossRef] [PubMed]
- Luo, Y.; Shang, P.; Li, D. Luteolin: A Flavonoid that Has Multiple Cardio-Protective Effects and Its Molecular Mechanisms. Front. Pharmacol. 2017, 8, 692. [Google Scholar] [CrossRef] [Green Version]
- Hostetler, G.L.; Ralston, R.A.; Schwartz, S.J. Flavones: Food Sources, Bioavailability, Metabolism, and Bioactivity. Adv. Nutr. 2017, 8, 423–435. [Google Scholar] [CrossRef] [Green Version]
- Rangboo, V.; Noroozi, M.; Zavoshy, R.; Rezadoost, S.A.; Mohammadpoorasl, A. The Effect of Artichoke Leaf Extract on Alanine Aminotransferase and Aspartate Aminotransferase in the Patients with Nonalcoholic Steatohepatitis. Int. J. Hepatol. 2016, 2016, 4030476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sahebkar, A.; Pirro, M.; Banach, M.; Mikhailidis, D.P.; Atkin, S.L.; Cicero, A.F.G. Lipid-lowering activity of artichoke extracts: A systematic review and meta-analysis. Crit. Rev. Food Sci. Nutr. 2018, 58, 2549–2556. [Google Scholar] [CrossRef]
- Xia, F.; Wang, C.; Jin, Y.; Liu, Q.; Meng, Q.; Liu, K.; Sun, H. Luteolin protects HUVECs from TNF-alpha-induced oxidative stress and inflammation via its effects on the Nox4/ROS-NF-kappaB and MAPK pathways. J. Atheroscler. Thromb. 2014, 21, 768–783. [Google Scholar] [CrossRef] [Green Version]
- Xiao, C.; Xia, M.L.; Wang, J.; Zhou, X.R.; Lou, Y.Y.; Tang, L.H.; Zhang, F.J.; Yang, J.T.; Qian, L.B. Luteolin Attenuates Cardiac Ischemia/Reperfusion Injury in Diabetic Rats by Modulating Nrf2 Antioxidative Function. Oxid. Med. Cell. Longev. 2019, 2019, 2719252. [Google Scholar] [CrossRef]
- Jia, Z.; Nallasamy, P.; Liu, D.; Shah, H.; Li, J.Z.; Chitrakar, R.; Si, H.; McCormick, J.; Zhu, H.; Zhen, W.; et al. Luteolin protects against vascular inflammation in mice and TNF-alpha-induced monocyte adhesion to endothelial cells via suppressing IKappaBalpha/NF-kappaB signaling pathway. J. Nutr. Biochem. 2015, 26, 293–302. [Google Scholar] [CrossRef] [Green Version]
- Zhang, L.; Wang, X.; Zhang, L.; Virgous, C.; Si, H. Combination of curcumin and luteolin synergistically inhibits TNF-alpha-induced vascular inflammation in human vascular cells and mice. J. Nutr. Biochem. 2019, 73, 108222. [Google Scholar] [CrossRef]
- Kim, H.J.; Lee, W.; Yun, J.M. Luteolin inhibits hyperglycemia-induced proinflammatory cytokine production and its epigenetic mechanism in human monocytes. Phytother. Res. PTR 2014, 28, 1383–1391. [Google Scholar] [CrossRef] [PubMed]
- Zaragoza, C.; Villaescusa, L.; Monserrat, J.; Zaragoza, F.; Alvarez-Mon, M. Potential Therapeutic Anti-Inflammatory and Immunomodulatory Effects of Dihydroflavones, Flavones, and Flavonols. Molecules 2020, 25, 1017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hong, X.; Zhao, X.; Wang, G.; Zhang, Z.; Pei, H.; Liu, Z. Luteolin Treatment Protects against Renal Ischemia-Reperfusion Injury in Rats. Mediat. Inflamm. 2017, 2017, 9783893. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ding, X.; Zheng, L.; Yang, B.; Wang, X.; Ying, Y. Luteolin Attenuates Atherosclerosis Via Modulating Signal Transducer And Activator Of Transcription 3-Mediated Inflammatory Response. Drug Des. Dev. Ther. 2019, 13, 3899–3911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, P.; Sun, J.; Lv, S.; Xie, T.; Wang, X. Apigenin Alleviates Myocardial Reperfusion Injury in Rats by Downregulating miR-15b. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2019, 25, 2764–2776. [Google Scholar] [CrossRef] [PubMed]
- Bian, C.; Xu, T.; Zhu, H.; Pan, D.; Liu, Y.; Luo, Y.; Wu, P.; Li, D. Luteolin Inhibits Ischemia/Reperfusion-Induced Myocardial Injury in Rats via Downregulation of microRNA-208b-3p. PLoS ONE 2015, 10, e0144877. [Google Scholar] [CrossRef]
- Arango, D.; Diosa-Toro, M.; Rojas-Hernandez, L.S.; Cooperstone, J.L.; Schwartz, S.J.; Mo, X.; Jiang, J.; Schmittgen, T.D.; Doseff, A.I. Dietary apigenin reduces LPS-induced expression of miR-155 restoring immune balance during inflammation. Mol. Nutr. Food Res. 2015, 59, 763–772. [Google Scholar] [CrossRef]
- Zhang, Z.; Xu, P.; Yu, H.; Shi, L. Luteolin protects PC-12 cells from H2O2-induced injury by up-regulation of microRNA-21. Biomed. Pharmacother. 2019, 112, 108698. [Google Scholar] [CrossRef]
- Ning, B.B.; Zhang, Y.; Wu, D.D.; Cui, J.G.; Liu, L.; Wang, P.W.; Wang, W.J.; Zhu, W.L.; Chen, Y.; Zhang, T. Luteolin-7-diglucuronide attenuates isoproterenol-induced myocardial injury and fibrosis in mice. Acta Pharmacol. Sin. 2017, 38, 331–341. [Google Scholar] [CrossRef] [Green Version]
- Kara, S.; Gencer, B.; Karaca, T.; Tufan, H.A.; Arikan, S.; Ersan, I.; Karaboga, I.; Hanci, V. Protective effect of hesperetin and naringenin against apoptosis in ischemia/reperfusion-induced retinal injury in rats. Sci. World J. 2014, 2014, 797824. [Google Scholar] [CrossRef] [Green Version]
- Kanaze, F.I.; Bounartzi, M.I.; Georgarakis, M.; Niopas, I. Pharmacokinetics of the citrus flavanone aglycones hesperetin and naringenin after single oral administration in human subjects. Eur. J. Clin. Nutr. 2007, 61, 472–477. [Google Scholar] [CrossRef] [Green Version]
- Joshi, R.; Kulkarni, Y.A.; Wairkar, S. Pharmacokinetic, pharmacodynamic and formulations aspects of Naringenin: An update. Life Sci. 2018, 215, 43–56. [Google Scholar] [CrossRef] [PubMed]
- Miceli, N.; Mondello, M.R.; Monforte, M.T.; Sdrafkakis, V.; Dugo, P.; Crupi, M.L.; Taviano, M.F.; De Pasquale, R.; Trovato, A. Hypolipidemic effects of Citrus bergamia Risso et Poiteau juice in rats fed a hypercholesterolemic diet. J. Agric. Food Chem. 2007, 55, 10671–10677. [Google Scholar] [CrossRef] [PubMed]
- Mulvihill, E.E.; Assini, J.M.; Sutherland, B.G.; DiMattia, A.S.; Khami, M.; Koppes, J.B.; Sawyez, C.G.; Whitman, S.C.; Huff, M.W. Naringenin decreases progression of atherosclerosis by improving dyslipidemia in high-fat-fed low-density lipoprotein receptor-null mice. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 742–748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Borradaile, N.M.; de Dreu, L.E.; Barrett, P.H.; Behrsin, C.D.; Huff, M.W. Hepatocyte apoB-containing lipoprotein secretion is decreased by the grapefruit flavonoid, naringenin, via inhibition of MTP-mediated microsomal triglyceride accumulation. Biochemistry 2003, 42, 1283–1291. [Google Scholar] [CrossRef] [PubMed]
- Lee, C.H.; Jeong, T.S.; Choi, Y.K.; Hyun, B.H.; Oh, G.T.; Kim, E.H.; Kim, J.R.; Han, J.I.; Bok, S.H. Anti-atherogenic effect of citrus flavonoids, naringin and naringenin, associated with hepatic ACAT and aortic VCAM-1 and MCP-1 in high cholesterol-fed rabbits. Biochem. Biophys. Res. Commun. 2001, 284, 681–688. [Google Scholar] [CrossRef]
- Jung, U.J.; Kim, H.F.; Lee, J.S.; Lee, J.F.; Lee, M.K.; Lee, M.F.; Kim, H.O.; Kim, H.F.; Park, E.J.; Park, E.F.; et al. Naringin supplementation lowers plasma lipids and enhances erythrocyte antioxidant enzyme activities in hypercholesterolemic subjects. Clin. Nutr. 2003, 22, 561–568. [Google Scholar] [CrossRef]
- Perna, S.; Spadaccini, D.; Botteri, L.; Girometta, C.; Riva, A.; Allegrini, P.; Petrangolini, G.; Infantino, V.; Rondanelli, M. Efficacy of bergamot: From anti-inflammatory and anti-oxidative mechanisms to clinical applications as preventive agent for cardiovascular morbidity, skin diseases, and mood alterations. Food Sci. Nutr. 2019, 7, 369–384. [Google Scholar] [CrossRef] [Green Version]
- Rizza, S.; Muniyappa, R.; Iantorno, M.; Kim, J.A.; Chen, H.; Pullikotil, P.; Senese, N.; Tesauro, M.; Lauro, D.; Cardillo, C.; et al. Citrus polyphenol hesperidin stimulates production of nitric oxide in endothelial cells while improving endothelial function and reducing inflammatory markers in patients with metabolic syndrome. J. Clin. Endocrinol. Metab. 2011, 96, E782–E792. [Google Scholar] [CrossRef]
- Yamamoto, M.; Jokura, H.; Hashizume, K.; Ominami, H.; Shibuya, Y.; Suzuki, A.; Hase, T.; Shimotoyodome, A. Hesperidin metabolite hesperetin-7-O-glucuronide, but not hesperetin-3′-O-glucuronide, exerts hypotensive, vasodilatory, and anti-inflammatory activities. Food Funct. 2013, 4, 1346–1351. [Google Scholar] [CrossRef]
- Rendeiro, C.; Dong, H.; Saunders, C.; Harkness, L.; Blaze, M.; Hou, Y.; Belanger, R.L.; Altieri, V.; Nunez, M.A.; Jackson, K.G.; et al. Flavanone-rich citrus beverages counteract the transient decline in postprandial endothelial function in humans: A randomised, controlled, double-masked, cross-over intervention study. Br. J. Nutr. 2016, 116, 1999–2010. [Google Scholar] [CrossRef] [PubMed]
- Borgatti, M.; Mancini, I.; Bianchi, N.; Guerrini, A.; Lampronti, I.; Rossi, D.; Sacchetti, G.; Gambari, R. Bergamot (Citrus bergamia Risso) fruit extracts and identified components alter expression of interleukin 8 gene in cystic fibrosis bronchial epithelial cell lines. BMC Biochem. 2011, 12, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xie, J.; Zhang, X.; Zhang, L. Negative regulation of inflammation by SIRT1. Pharmacol. Res. 2013, 67, 60–67. [Google Scholar] [CrossRef]
- Testai, L.; Piragine, E.; Piano, I.; Flori, L.; Da Pozzo, E.; Miragliotta, V.; Pirone, A.; Citi, V.; Di Cesare Mannelli, L.; Brogi, S.; et al. The Citrus Flavonoid Naringenin Protects the Myocardium from Ageing-Dependent Dysfunction: Potential Role of SIRT1. Oxid. Med. Cell. Longev. 2020, 2020, 4650207. [Google Scholar] [CrossRef] [Green Version]
- Miwa, Y.; Mitsuzumi, H.; Sunayama, T.; Yamada, M.; Okada, K.; Kubota, M.; Chaen, H.; Mishima, Y.; Kibata, M. Glucosyl hesperidin lowers serum triglyceride level in hypertriglyceridemic subjects through the improvement of very low-density lipoprotein metabolic abnormality. J. Nutr. Sci. Vitaminol. 2005, 51, 460–470. [Google Scholar] [CrossRef] [Green Version]
- Demonty, I.; Lin, Y.; Zebregs, Y.E.; Vermeer, M.A.; van der Knaap, H.C.; Jakel, M.; Trautwein, E.A. The citrus flavonoids hesperidin and naringin do not affect serum cholesterol in moderately hypercholesterolemic men and women. J. Nutr. 2010, 140, 1615–1620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Assini, J.M.; Mulvihill, E.E.; Huff, M.W. Citrus flavonoids and lipid metabolism. Curr. Opin. Lipidol. 2013, 24, 34–40. [Google Scholar] [CrossRef] [PubMed]
- Hsu, C.L.; Fang, S.C.; Yen, G.C. Anti-inflammatory effects of phenolic compounds isolated from the flowers of Nymphaea mexicana Zucc. Food Funct. 2013, 4, 1216–1222. [Google Scholar] [CrossRef]
- Raza, S.S.; Khan, M.M.; Ahmad, A.; Ashafaq, M.; Islam, F.; Wagner, A.P.; Safhi, M.M.; Islam, F. Neuroprotective effect of naringenin is mediated through suppression of NF-kappaB signaling pathway in experimental stroke. Neuroscience 2013, 230, 157–171. [Google Scholar] [CrossRef]
- Fuior, E.V.; Deleanu, M.; Constantinescu, C.A.; Rebleanu, D.; Voicu, G.; Simionescu, M.; Calin, M. Functional Role of VCAM-1 Targeted Flavonoid-Loaded Lipid Nanoemulsions in Reducing Endothelium Inflammation. Pharmaceutics 2019, 11, 391. [Google Scholar] [CrossRef] [Green Version]
- Liang, J.N.; Zou, X.; Fang, X.H.; Xu, J.D.; Xiao, Z.; Zhu, J.N.; Li, H.; Yang, J.; Zeng, N.; Yuan, S.J.; et al. The Smad3-miR-29b/miR-29c axis mediates the protective effect of macrophage migration inhibitory factor against cardiac fibrosis. (BBA)-Mol. Basis Dis. 2019, 1865, 2441–2450. [Google Scholar] [CrossRef] [PubMed]
- Murakami, A. Dose-dependent functionality and toxicity of green tea polyphenols in experimental rodents. Arch. Biochem. Biophys. 2014, 557, 3–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cai, Z.Y.; Li, X.M.; Liang, J.P.; Xiang, L.P.; Wang, K.R.; Shi, Y.L.; Yang, R.; Shi, M.; Ye, J.H.; Lu, J.L.; et al. Bioavailability of Tea Catechins and Its Improvement. Molecules 2018, 23, 2346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Onakpoya, I.; Spencer, E.; Heneghan, C.; Thompson, M. The effect of green tea on blood pressure and lipid profile: A systematic review and meta-analysis of randomized clinical trials. Nutr. Metab. Cardiovasc. Dis. NMCD 2014, 24, 823–836. [Google Scholar] [CrossRef] [PubMed]
- Kim, A.; Chiu, A.; Barone, M.K.; Avino, D.; Wang, F.; Coleman, C.I.; Phung, O.J. Green tea catechins decrease total and low-density lipoprotein cholesterol: A systematic review and meta-analysis. J. Am. Diet. Assoc. 2011, 111, 1720–1729. [Google Scholar] [CrossRef] [PubMed]
- Zheng, X.X.; Xu, Y.L.; Li, S.H.; Liu, X.X.; Hui, R.; Huang, X.H. Green tea intake lowers fasting serum total and LDL cholesterol in adults: A meta-analysis of 14 randomized controlled trials. Am. J. Clin. Nutr. 2011, 94, 601–610. [Google Scholar] [CrossRef] [Green Version]
- Koo, S.I.; Noh, S.K. Green tea as inhibitor of the intestinal absorption of lipids: Potential mechanism for its lipid-lowering effect. J. Nutr. Biochem. 2007, 18, 179–183. [Google Scholar] [CrossRef] [Green Version]
- Abe, I.; Seki, T.; Umehara, K.; Miyase, T.; Noguchi, H.; Sakakibara, J.; Ono, T. Green tea polyphenols: Novel and potent inhibitors of squalene epoxidase. Biochem. Biophys. Res. Commun. 2000, 268, 767–771. [Google Scholar] [CrossRef]
- Naveed, M.; BiBi, J.; Kamboh, A.A.; Suheryani, I.; Kakar, I.; Fazlani, S.A.; FangFang, X.; Kalhoro, S.A.; Yunjuan, L.; Kakar, M.U.; et al. Pharmacological values and therapeutic properties of black tea (Camellia sinensis): A comprehensive overview. Biomed. Pharmacother. 2018, 100, 521–531. [Google Scholar] [CrossRef]
- Santos, H.O.; Macedo, R.C.O. Cocoa-induced (Theobroma cacao) effects on cardiovascular system: HDL modulation pathways. Clin. Nutr. ESPEN 2018, 27, 10–15. [Google Scholar] [CrossRef]
- Snoussi, C.; Ducroc, R.; Hamdaoui, M.H.; Dhaouadi, K.; Abaidi, H.; Cluzeaud, F.; Nazaret, C.; Le Gall, M.; Bado, A. Green tea decoction improves glucose tolerance and reduces weight gain of rats fed normal and high-fat diet. J. Nutr. Biochem. 2014, 25, 557–564. [Google Scholar] [CrossRef] [PubMed]
- Shrestha, S.; Ehlers, S.J.; Lee, J.Y.; Fernandez, M.L.; Koo, S.I. Dietary green tea extract lowers plasma and hepatic triglycerides and decreases the expression of sterol regulatory element-binding protein-1c mRNA and its responsive genes in fructose-fed, ovariectomized rats. J. Nutr. 2009, 139, 640–645. [Google Scholar] [CrossRef] [PubMed]
- Pang, J.; Zhang, Z.; Zheng, T.Z.; Bassig, B.A.; Mao, C.; Liu, X.; Zhu, Y.; Shi, K.; Ge, J.; Yang, Y.J.; et al. Green tea consumption and risk of cardiovascular and ischemic related diseases: A meta-analysis. Int. J. Cardiol. 2016, 202, 967–974. [Google Scholar] [CrossRef] [PubMed]
- Mangels, D.R.; Mohler, E.R., 3rd. Catechins as Potential Mediators of Cardiovascular Health. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 757–763. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.; Sun, Z.; Chu, P.; Li, H.; Ahsan, A.; Zhou, Z.; Zhang, Z.; Sun, B.; Wu, J.; Xi, Y.; et al. EGCG protects against homocysteine-induced human umbilical vein endothelial cells apoptosis by modulating mitochondrial-dependent apoptotic signaling and PI3K/Akt/eNOS signaling pathways. Apoptosis 2017, 22, 672–680. [Google Scholar] [CrossRef]
- Wu, C.C.; Hsu, M.C.; Hsieh, C.W.; Lin, J.B.; Lai, P.H.; Wung, B.S. Upregulation of heme oxygenase-1 by Epigallocatechin-3-gallate via the phosphatidylinositol 3-kinase/Akt and ERK pathways. Life Sci. 2006, 78, 2889–2897. [Google Scholar] [CrossRef]
- Suzuki-Sugihara, N.; Kishimoto, Y.; Saita, E.; Taguchi, C.; Kobayashi, M.; Ichitani, M.; Ukawa, Y.; Sagesaka, Y.M.; Suzuki, E.; Kondo, K. Green tea catechins prevent low-density lipoprotein oxidation via their accumulation in low-density lipoprotein particles in humans. Nutr. Res. 2016, 36, 16–23. [Google Scholar] [CrossRef]
- Tas, S.; Sarandol, E.; Ziyanok, S.; Aslan, K.; Dirican, M. Effects of green tea on serum paraoxonase/arylesterase activities in streptozotocin-induced diabetic rats. Nutr. Res. 2005, 25, 1061–1074. [Google Scholar] [CrossRef]
- Ludwig, A.; Lorenz, M.; Grimbo, N.; Steinle, F.; Meiners, S.; Bartsch, C.; Stangl, K.; Baumann, G.; Stangl, V. The tea flavonoid epigallocatechin-3-gallate reduces cytokine-induced VCAM-1 expression and monocyte adhesion to endothelial cells. Biochem. Biophys. Res. Commun. 2004, 316, 659–665. [Google Scholar] [CrossRef]
- Arola-Arnal, A.; Blade, C. Proanthocyanidins modulate microRNA expression in human HepG2 cells. PLoS ONE 2011, 6, e25982. [Google Scholar] [CrossRef] [Green Version]
- Baselga-Escudero, L.; Blade, C.; Ribas-Latre, A.; Casanova, E.; Suarez, M.; Torres, J.L.; Salvado, M.J.; Arola, L.; Arola-Arnal, A. Resveratrol and EGCG bind directly and distinctively to miR-33a and miR-122 and modulate divergently their levels in hepatic cells. Nucleic Acids Res. 2014, 42, 882–892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, H.; Bian, S.; Yang, C.S. Green tea polyphenol EGCG suppresses lung cancer cell growth through upregulating miR-210 expression caused by stabilizing HIF-1alpha. Carcinogenesis 2011, 32, 1881–1889. [Google Scholar] [CrossRef] [Green Version]
- Kaelin, W.G., Jr.; Ratcliffe, P.J. Oxygen sensing by metazoans: The central role of the HIF hydroxylase pathway. Mol. Cell 2008, 30, 393–402. [Google Scholar] [CrossRef] [PubMed]
- Jaiswal, N.; Akhtar, J.; Singh, S.P.; Badruddeen; Ahsan, F. An Overview on Genistein and its Various Formulations. Drug Res. 2019, 69, 305–313. [Google Scholar] [CrossRef] [PubMed]
- Beavers, D.P.; Beavers, K.M.; Miller, M.; Stamey, J.; Messina, M.J. Exposure to isoflavone-containing soy products and endothelial function: A Bayesian meta-analysis of randomized controlled trials. Nutr. Metab. Cardiovasc. Dis. NMCD 2012, 22, 182–191. [Google Scholar] [CrossRef]
- Yu, D.; Shu, X.O.; Li, H.; Yang, G.; Cai, Q.; Xiang, Y.B.; Ji, B.T.; Franke, A.A.; Gao, Y.T.; Zheng, W.; et al. Dietary isoflavones, urinary isoflavonoids, and risk of ischemic stroke in women. Am. J. Clin. Nutr. 2015, 102, 680–686. [Google Scholar] [CrossRef] [Green Version]
- Si, H.; Liu, D. Genistein, a soy phytoestrogen, upregulates the expression of human endothelial nitric oxide synthase and lowers blood pressure in spontaneously hypertensive rats. J. Nutr. 2008, 138, 297–304. [Google Scholar] [CrossRef] [Green Version]
- Zhang, H.P.; Zhao, J.H.; Yu, H.X.; Guo, D.X. Genistein ameliorated endothelial nitric oxidase synthase uncoupling by stimulating sirtuin-1 pathway in ox-LDL-injured HUVECs. Environ. Toxicol. Pharmacol. 2016, 42, 118–124. [Google Scholar] [CrossRef]
- Ras, R.T.; Streppel, M.T.; Draijer, R.; Zock, P.L. Flow-mediated dilation and cardiovascular risk prediction: A systematic review with meta-analysis. Int. J. Cardiol. 2013, 168, 344–351. [Google Scholar] [CrossRef]
- Hazim, S.; Curtis, P.J.; Schar, M.Y.; Ostertag, L.M.; Kay, C.D.; Minihane, A.M.; Cassidy, A. Acute benefits of the microbial-derived isoflavone metabolite equol on arterial stiffness in men prospectively recruited according to equol producer phenotype: A double-blind randomized controlled trial. Am. J. Clin. Nutr. 2016, 103, 694–702. [Google Scholar] [CrossRef] [Green Version]
- Park, M.H.; Ju, J.W.; Kim, M.; Han, J.S. The protective effect of daidzein on high glucose-induced oxidative stress in human umbilical vein endothelial cells. Z. für Naturforschung C J. Biosci. 2016, 71, 21–28. [Google Scholar] [CrossRef] [PubMed]
- Setchell, K.D.; Clerici, C.; Lephart, E.D.; Cole, S.J.; Heenan, C.; Castellani, D.; Wolfe, B.E.; Nechemias-Zimmer, L.; Brown, N.M.; Lund, T.D.; et al. S-equol, a potent ligand for estrogen receptor beta, is the exclusive enantiomeric form of the soy isoflavone metabolite produced by human intestinal bacterial flora. Am. J. Clin. Nutr. 2005, 81, 1072–1079. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rowlands, D.J.; Chapple, S.; Siow, R.C.; Mann, G.E. Equol-stimulated mitochondrial reactive oxygen species activate endothelial nitric oxide synthase and redox signaling in endothelial cells: Roles for F-actin and GPR30. Hypertension 2011, 57, 833–840. [Google Scholar] [CrossRef] [PubMed]
- Joy, S.; Siow, R.C.; Rowlands, D.J.; Becker, M.; Wyatt, A.W.; Aaronson, P.I.; Coen, C.W.; Kallo, I.; Jacob, R.; Mann, G.E. The isoflavone Equol mediates rapid vascular relaxation: Ca2+-independent activation of endothelial nitric-oxide synthase/Hsp90 involving ERK1/2 and Akt phosphorylation in human endothelial cells. J. Biol. Chem. 2006, 281, 27335–27345. [Google Scholar] [CrossRef] [Green Version]
- Babu, P.V.; Si, H.; Fu, Z.; Zhen, W.; Liu, D. Genistein prevents hyperglycemia-induced monocyte adhesion to human aortic endothelial cells through preservation of the cAMP signaling pathway and ameliorates vascular inflammation in obese diabetic mice. J. Nutr. 2012, 142, 724–730. [Google Scholar] [CrossRef]
- Han, S.; Wu, H.; Li, W.; Gao, P. Protective effects of genistein in homocysteine-induced endothelial cell inflammatory injury. Mol. Cell. Biochem. 2015, 403, 43–49. [Google Scholar] [CrossRef]
- Xu, L.; Liu, J.T.; Li, K.; Wang, S.Y.; Xu, S. Genistein inhibits Ang II-induced CRP and MMP-9 generations via the ER-p38/ERK1/2-PPARgamma-NF-kappaB signaling pathway in rat vascular smooth muscle cells. Life Sci. 2019, 216, 140–146. [Google Scholar] [CrossRef]
- Zhang, H.; Zhao, Z.; Pang, X.; Yang, J.; Yu, H.; Zhang, Y.; Zhou, H.; Zhao, J. Genistein Protects Against Ox-LDL-Induced Inflammation Through MicroRNA-155/SOCS1-Mediated Repression of NF-kB Signaling Pathway in HUVECs. Inflammation 2017, 40, 1450–1459. [Google Scholar] [CrossRef]
- Gan, M.; Zheng, T.; Shen, L.; Tan, Y.; Fan, Y.; Shuai, S.; Bai, L.; Li, X.; Wang, J.; Zhang, S.; et al. Genistein reverses isoproterenol-induced cardiac hypertrophy by regulating miR-451/TIMP2. Biomed. Pharmacother. 2019, 112, 108618. [Google Scholar] [CrossRef]
- Krga, I.; Milenkovic, D. Anthocyanins: From Sources and Bioavailability to Cardiovascular-Health Benefits and Molecular Mechanisms of Action. J. Agric. Food Chem. 2019, 67, 1771–1783. [Google Scholar] [CrossRef]
- Eker, M.E.; Aaby, K.; Budic-Leto, I.; Brncic, S.R.; El, S.N.; Karakaya, S.; Simsek, S.; Manach, C.; Wiczkowski, W.; Pascual-Teresa, S. A Review of Factors Affecting Anthocyanin Bioavailability: Possible Implications for the Inter-Individual Variability. Foods 2019, 9, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hollands, W.J.; Armah, C.N.; Doleman, J.F.; Perez-Moral, N.; Winterbone, M.S.; Kroon, P.A. 4-Week consumption of anthocyanin-rich blood orange juice does not affect LDL-cholesterol or other biomarkers of CVD risk and glycaemia compared with standard orange juice: A randomised controlled trial. Br. J. Nutr. 2018, 119, 415–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Niculescu, L.S.; Sanda, G.M.; Simionescu, N.; Sima, A.V. Bilberries exert an anti-atherosclerotic effect in lipid-loaded macrophages. Cent. Eur. J. Biol. 2014, 9, 268–276. [Google Scholar] [CrossRef] [Green Version]
- Farrell, N.; Norris, G.; Lee, S.G.; Chun, O.K.; Blesso, C.N. Anthocyanin-rich black elderberry extract improves markers of HDL function and reduces aortic cholesterol in hyperlipidemic mice. Food Funct. 2015, 6, 1278–1287. [Google Scholar] [CrossRef] [PubMed]
- Wallace, T.C.; Slavin, M.; Frankenfeld, C.L. Systematic Review of Anthocyanins and Markers of Cardiovascular Disease. Nutrients 2016, 8, 32. [Google Scholar] [CrossRef] [PubMed]
- Sorrenti, V.; Mazza, F.; Campisi, A.; Di Giacomo, C.; Acquaviva, R.; Vanella, L.; Galvano, F. Heme oxygenase induction by cyanidin-3-O-beta-glucoside in cultured human endothelial cells. Mol. Nutr. Food Res. 2007, 51, 580–586. [Google Scholar] [CrossRef]
- Xu, J.W.; Ikeda, K.; Yamori, Y. Upregulation of endothelial nitric oxide synthase by cyanidin-3-glucoside, a typical anthocyanin pigment. Hypertension 2004, 44, 217–222. [Google Scholar] [CrossRef] [Green Version]
- Kennedy, M.A.; Barrera, G.C.; Nakamura, K.; Baldan, A.; Tarr, P.; Fishbein, M.C.; Frank, J.; Francone, O.L.; Edwards, P.A. ABCG1 has a critical role in mediating cholesterol efflux to HDL and preventing cellular lipid accumulation. Cell Metab. 2005, 1, 121–131. [Google Scholar] [CrossRef] [Green Version]
- Terasaka, N.; Yu, S.; Yvan-Charvet, L.; Wang, N.; Mzhavia, N.; Langlois, R.; Pagler, T.; Li, R.; Welch, C.L.; Goldberg, I.J.; et al. ABCG1 and HDL protect against endothelial dysfunction in mice fed a high-cholesterol diet. J. Clin. Investig. 2008, 118, 3701–3713. [Google Scholar] [CrossRef] [Green Version]
- Huang, W.; Zhu, Y.; Li, C.; Sui, Z.; Min, W. Effect of Blueberry Anthocyanins Malvidin and Glycosides on the Antioxidant Properties in Endothelial Cells. Oxid. Med. Cell. Longev. 2016, 2016, 1591803. [Google Scholar] [CrossRef] [Green Version]
- Paixao, J.; Dinis, T.C.; Almeida, L.M. Malvidin-3-glucoside protects endothelial cells up-regulating endothelial NO synthase and inhibiting peroxynitrite-induced NF-kB activation. Chem.-Biol. Interact. 2012, 199, 192–200. [Google Scholar] [CrossRef] [PubMed]
- Huang, T.W.; Chang, C.L.; Kao, E.S.; Lin, J.H. Effect of Hibiscus sabdariffa extract on high fat diet-induced obesity and liver damage in hamsters. Food Nutr. Res. 2015, 59, 29018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davinelli, S.; Bertoglio, J.C.; Zarrelli, A.; Pina, R.; Scapagnini, G. A Randomized Clinical Trial Evaluating the Efficacy of an Anthocyanin-Maqui Berry Extract (Delphinol(R)) on Oxidative Stress Biomarkers. J. Am. Coll. Nutr. 2015, 34 (Suppl. S1), 28–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jennings, A.; Welch, A.A.; Fairweather-Tait, S.J.; Kay, C.; Minihane, A.M.; Chowienczyk, P.; Jiang, B.; Cecelja, M.; Spector, T.; Macgregor, A.; et al. Higher anthocyanin intake is associated with lower arterial stiffness and central blood pressure in women. Am. J. Clin. Nutr. 2012, 96, 781–788. [Google Scholar] [CrossRef] [Green Version]
- Chen, C.Y.; Yi, L.; Jin, X.; Zhang, T.; Fu, Y.J.; Zhu, J.D.; Mi, M.T.; Zhang, Q.Y.; Ling, W.H.; Yu, B. Inhibitory effect of delphinidin on monocyte-endothelial cell adhesion induced by oxidized low-density lipoprotein via ROS/p38MAPK/NF-kappaB pathway. Cell Biochem. Biophys. 2011, 61, 337–348. [Google Scholar] [CrossRef]
- Lee, Y.; Lee, J.Y. Blackcurrant (Ribes nigrum) Extract Exerts an Anti-Inflammatory Action by Modulating Macrophage Phenotypes. Nutrients 2019, 11, 975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rodriguez-Mateos, A.; Istas, G.; Boschek, L.; Feliciano, R.P.; Mills, C.E.; Boby, C.; Gomez-Alonso, S.; Milenkovic, D.; Heiss, C. Circulating Anthocyanin Metabolites Mediate Vascular Benefits of Blueberries: Insights From Randomized Controlled Trials, Metabolomics, and Nutrigenomics. J. Gerontol. Ser. A 2019, 74, 967–976. [Google Scholar] [CrossRef] [PubMed]
- Marques, C.; Fernandes, I.; Norberto, S.; Sa, C.; Teixeira, D.; de Freitas, V.; Mateus, N.; Calhau, C.; Faria, A. Pharmacokinetics of blackberry anthocyanins consumed with or without ethanol: A randomized and crossover trial. Mol. Nutr. Food Res. 2016, 60, 2319–2330. [Google Scholar] [CrossRef] [PubMed]
- Singletary, K. Ginger: An Overview of Health Benefits. Nutr. Today 2010, 45, 171–183. [Google Scholar] [CrossRef]
- Arablou, T.; Aryaeian, N.; Valizadeh, M.; Sharifi, F.; Hosseini, A.; Djalali, M. The effect of ginger consumption on glycemic status, lipid profile and some inflammatory markers in patients with type 2 diabetes mellitus. Int. J. Food Sci. Nutr. 2014, 65, 515–520. [Google Scholar] [CrossRef]
- Santos Braga, S. Ginger: Panacea or Consumer’s Hype? Appl. Sci. 2019, 9, 1570. [Google Scholar] [CrossRef] [Green Version]
- Gao, H.; Guan, T.; Li, C.; Zuo, G.; Yamahara, J.; Wang, J.; Li, Y. Treatment with ginger ameliorates fructose-induced Fatty liver and hypertriglyceridemia in rats: Modulation of the hepatic carbohydrate response element-binding protein-mediated pathway. Evid.-Based Complement. Altern. Med. eCAM 2012, 2012, 570948. [Google Scholar] [CrossRef] [PubMed]
- Carnuta, M.G.; Deleanu, M.; Barbalata, T.; Toma, L.; Raileanu, M.; Sima, A.V.; Stancu, C.S. Zingiber officinale extract administration diminishes steroyl-CoA desaturase gene expression and activity in hyperlipidemic hamster liver by reducing the oxidative and endoplasmic reticulum stress. Phytomedicine 2018, 48, 62–69. [Google Scholar] [CrossRef] [PubMed]
- Lei, L.; Liu, Y.; Wang, X.; Jiao, R.; Ma, K.Y.; Li, Y.M.; Wang, L.; Man, S.W.; Sang, S.; Huang, Y.; et al. Plasma cholesterol-lowering activity of gingerol- and shogaol-enriched extract is mediated by increasing sterol excretion. J. Agric. Food Chem. 2014, 62, 10515–10521. [Google Scholar] [CrossRef] [PubMed]
- Misawa, K.; Hashizume, K.; Yamamoto, M.; Minegishi, Y.; Hase, T.; Shimotoyodome, A. Ginger extract prevents high-fat diet-induced obesity in mice via activation of the peroxisome proliferator-activated receptor delta pathway. J. Nutr. Biochem. 2015, 26, 1058–1067. [Google Scholar] [CrossRef]
- Semwal, R.B.; Semwal, D.K.; Combrinck, S.; Viljoen, A.M. Gingerols and shogaols: Important nutraceutical principles from ginger. Phytochemistry 2015, 117, 554–568. [Google Scholar] [CrossRef]
- Toma, L.; Raileanu, M.; Deleanu, M.; Stancu, C.S.; Sima, A.V. Novel molecular mechanisms by which ginger extract reduces the inflammatory stress in TNFα—Activated human endothelial cells; decrease of Ninjurin-1, TNFR1 and NADPH oxidase subunits expression. J. Funct. Foods 2018, 48, 654–664. [Google Scholar] [CrossRef]
- Wang, Y.K.; Hong, Y.J.; Yao, Y.H.; Huang, X.M.; Liu, X.B.; Zhang, C.Y.; Zhang, L.; Xu, X.L. 6-Shogaol Protects against Oxidized LDL-Induced Endothelial Injruries by Inhibiting Oxidized LDL-Evoked LOX-1 Signaling. Evid.-Based Complement. Altern. Med. eCAM 2013, 2013, 503521. [Google Scholar] [CrossRef]
- Lee, T.Y.; Lee, K.C.; Chen, S.Y.; Chang, H.H. 6-Gingerol inhibits ROS and iNOS through the suppression of PKC-alpha and NF-kappaB pathways in lipopolysaccharide-stimulated mouse macrophages. Biochem. Biophys. Res. Commun. 2009, 382, 134–139. [Google Scholar] [CrossRef]
- Ho, S.C.; Chang, Y.H. Comparison of Inhibitory Capacities of 6-, 8- and 10-Gingerols/Shogaols on the Canonical NLRP3 Inflammasome-Mediated IL-1beta Secretion. Molecules 2018, 23, 466. [Google Scholar] [CrossRef] [Green Version]
- Wang, S.; Tian, M.; Yang, R.; Jing, Y.; Chen, W.; Wang, J.; Zheng, X.; Wang, F. 6-Gingerol Ameliorates Behavioral Changes and Atherosclerotic Lesions in ApoE(-/-) Mice Exposed to Chronic Mild Stress. Cardiovasc. Toxicol. 2018, 18, 420–430. [Google Scholar] [CrossRef]
- Li, J.; Wang, S.; Yao, L.; Ma, P.; Chen, Z.; Han, T.L.; Yuan, C.; Zhang, J.; Jiang, L.; Liu, L.; et al. 6-gingerol ameliorates age-related hepatic steatosis: Association with regulating lipogenesis, fatty acid oxidation, oxidative stress and mitochondrial dysfunction. Toxicol. Appl. Pharmacol. 2019, 362, 125–135. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.; Lee, M.S.; Jung, S.; Son, H.Y.; Park, S.; Kang, B.; Kim, S.Y.; Kim, I.H.; Kim, C.T.; Kim, Y. Ginger Extract Ameliorates Obesity and Inflammation via Regulating MicroRNA-21/132 Expression and AMPK Activation in White Adipose Tissue. Nutrients 2018, 10, 1567. [Google Scholar] [CrossRef] [Green Version]
- Nelson, K.M.; Dahlin, J.L.; Bisson, J.; Graham, J.; Pauli, G.F.; Walters, M.A. The Essential Medicinal Chemistry of Curcumin. J. Med. Chem. 2017, 60, 1620–1637. [Google Scholar] [CrossRef] [PubMed]
- Baell, J.; Walters, M.A. Chemistry: Chemical con artists foil drug discovery. Nature 2014, 513, 481–483. [Google Scholar] [CrossRef] [PubMed]
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Toma, L.; Sanda, G.M.; Niculescu, L.S.; Deleanu, M.; Sima, A.V.; Stancu, C.S. Phenolic Compounds Exerting Lipid-Regulatory, Anti-Inflammatory and Epigenetic Effects as Complementary Treatments in Cardiovascular Diseases. Biomolecules 2020, 10, 641. https://doi.org/10.3390/biom10040641
Toma L, Sanda GM, Niculescu LS, Deleanu M, Sima AV, Stancu CS. Phenolic Compounds Exerting Lipid-Regulatory, Anti-Inflammatory and Epigenetic Effects as Complementary Treatments in Cardiovascular Diseases. Biomolecules. 2020; 10(4):641. https://doi.org/10.3390/biom10040641
Chicago/Turabian StyleToma, Laura, Gabriela Maria Sanda, Loredan Stefan Niculescu, Mariana Deleanu, Anca Volumnia Sima, and Camelia Sorina Stancu. 2020. "Phenolic Compounds Exerting Lipid-Regulatory, Anti-Inflammatory and Epigenetic Effects as Complementary Treatments in Cardiovascular Diseases" Biomolecules 10, no. 4: 641. https://doi.org/10.3390/biom10040641
APA StyleToma, L., Sanda, G. M., Niculescu, L. S., Deleanu, M., Sima, A. V., & Stancu, C. S. (2020). Phenolic Compounds Exerting Lipid-Regulatory, Anti-Inflammatory and Epigenetic Effects as Complementary Treatments in Cardiovascular Diseases. Biomolecules, 10(4), 641. https://doi.org/10.3390/biom10040641