Modulation of NOS Isoforms by Dietary Polyphenols: Comparison
Please note this is a comparison between Version 3 by Jessie Wu and Version 2 by Jessie Wu.

Nitric oxide (NO) plays several key roles in the functionality of an organism, and it is usually released in numerous organs and tissues. There are mainly three isoforms of the enzyme that produce NO starting from the metabolism of arginine, namely endothelial nitric oxide synthase (eNOS), inducible nitric oxide synthase (iNOS), and neuronal nitric oxide synthase (nNOS). The expression and activity of these isoforms depends on the activation/deactivation of different signaling pathways at an intracellular level following different physiological and pathological stimuli. Compounds of natural origin such as polyphenols, which are obtainable through diet, have been widely studied in in vivo and in vitro investigations for their ability to induce or inhibit NO release, depending on the tissue. 

  • nitric oxide
  • polyphenols
  • signaling pathways

1. Activity and Expression of Inducible Nitric Oxide Synthase

The inducible nitric oxide synthase (iNOS) isoform is probably the most studied with regard to its modulation by compounds of natural origin. Regarding polyphenols, its expression has been measured in countless cell types and in different experimental systems, even with contrasting results depending on the cell type and the compound tested. In most cases, the modulation of iNOS expression has been studied as a function of some anti-inflammatory activity of the tested compounds, but the related countless studies are not reported in this review. Instead, weresearchers focused on the research reporting the mechanisms involved in the biological action of polyphenols on iNOS expression and activity. Almost all the signaling pathways studied in this context converge on the modulation of the activity of NF-ĸB, a transcription factor which, once translocated into the nucleus, activates the processes that lead to the expression of iNOS in manifold cell types [1] (Figure 1). The translocation occurs through an important step, which is the degradation of the inhibitor of NF-ĸB, i.e., IĸB (mainly its α isoform), which occurs via phosphorylation induced by kinases such as IKK, which are in turn activated by other upstream kinases [2]. Numerous studies have, therefore, focused on the regulation of these kinases by polyphenols of various origins and chemical structures. The regulation of iNOS activity by dietary phenols has been studied mainly at the intestinal level, where this enzyme plays a key role in acute and chronic inflammatory diseases and where polyphenols may accumulate in physiologically relevant concentrations [3], thus representing an important tool to prevent the onset and progression of intestinal and systemic diseases [4].
Figure 1. Modulation of iNOS expression by polyphenols involving the downregulation of upstream kinases and NF-κB. Abbreviations: IKK—IĸB kinase; iNOS—inducible nitric oxide synthase; IκB—inhibitor of nuclear factor kappa B; LPS—lipopolysaccharide; MAPK—mitogen-activated protein kinase; NF-κB—nuclear factor kappa-light-chain-enhancer of activated B cells; NO—nitric oxide; O2●−—superoxide anion; ONOO—peroxynitrite; PI3K—inositol 1,4,5-triphosphate kinase; and TNF-α—tumor necrosis factor-α.
Ellagic acid was tested in an experimental murine model of Crohn’s disease by intra-colonic administration of 2,4,6-trinitrobenzenesulfonic acid (TNBS) in rats. This polyphenol could reduce the activation of p38, JNK, and ERK1/2 MAPKs, thus preventing the degradation of inhibitory protein IĸB and inducing an inhibition of the p65-NF-κB level in colonic mucosa [5]. In an identical experimental model, the upstream inhibition of cytosolic IKK and the consequent preservation of IĸBα in colon tissue was instead observed regarding treatments with theaflavin-3,3′-digallate (TFDG) [6]. In the case of curcumin, the inhibitory effects occurred on p38 but not on JNK, however, leading to the downregulation of iNOS and COX-2 expression [7]. In an acute injury model obtained by intra-colonic administration of acetic acid in rats, curcumin showed instead an inhibitory action on JNK as well as on p38 [8]. Gallic acid, a low-molecular-weight trihydroxybenzoic acid, was tested in an experimental murine model of ulcerative colitis. It was observed that gallic acid reduced the activation and nuclear accumulation of hosphor-STAT3Y705, preventing the degradation of IκB and inhibiting the nuclear translocation of p65-NF-κB in colonic mucosa, thus decreasing the expression of iNOS and COX-2 [9]. Furthermore, in BALB/c mice subjected to ischemia/reperfusion, resveratrol significantly ameliorated subacute intestinal injury through the decrease of NO production as well as iNOS expression, upregulating the expression of the deacetylase Sirtuin 1 (Sirt1) and, consequently, inhibiting the activity of NF-κB [10]. In vitro, in an intestinal model of HT-29 cells, cyanidin-3-glucoside reduced cytokine-induced inflammation in terms of NO, PGE2, and IL-8 production and of iNOS and COX-2 expressions. The phenolic compound did not act to prevent IκB-α degradation but significantly reduced levels of activated STAT1 accumulated in the cell nucleus. Moreover, it was established that the phosphorylation of p38 was not involved in this protective effect [11]. In the same cell model stimulated with cytokines, luteolin significantly inhibited IL-8 production, COX-2 and iNOS expression, and NO overproduction. Mechanistically, the inhibition of the Janus kinase (JAK)/signal transducers and activators of transcription (STAT) pathway was identified as a major event in the observed anti-inflammatory effects [12]. iNOS activation induced by a combination of cytokines (IL-1β, TNF-α, IFN-γ) in HT-29 cells was also counteracted by resveratrol, following the downregulation of the JAK-STAT pathway, decreasing the levels of activated STAT1 in the nucleus and the cytokine-stimulated activation of SAPK/JNK pathway, whereas no effects were exerted on p38 [13]. It has been observed that in Caco-2 cells stimulated with LPS, resveratrol was able to inhibit the phosphorylation of IĸB and prevent the expression of iNOS downstream through the downregulation of the TRL-4 receptors, which are implicated in the proinflammatory effects of LPS [14]. In the same experimental model, ourthe research group has recently described how the main polyphenols of extra virgin olive oil (EVOO) and ferulic acid derivatives, as well as their phase II metabolites formed in vivo following their ingestion, have been able to inhibit the degradation of IĸBα by acting on the p38, ERK1/2, and JNK MAPKs, consequently preventing the activation of iNOS and the release of NO [15][16][17]. In the same investigations, it was also shown that EVOO polyphenols such as hydroxytyrosol, tyrosol, and their metabolites did not act through the downregulation of the Akt kinase, whereas ferulic acid and its metabolites and derivatives were able to inhibit both MAPK and Akt kinases phosphorylation and to activate the Nrf-2 pathway, which is known to be involved in the inhibition of NF-ĸB activity.
The mechanisms to control iNOS expression have also been studied in other tissues, although less thoroughly than in the gastrointestinal system and macrophages. In chondrocytes, for example, the release of NO has been studied in the context of chronic inflammatory diseases such as osteoarthritis, and it has been shown that compounds such as epigallocatechin gallate (EGCG), fisetin, and resveratrol are able to modulate the inflammatory response switching off the expression of iNOS through different signaling pathways: activation of the Sirt1 pathway and subsequent inactivation of NF-ĸB translocation and inhibition of p65 acetylation [18][19][20]. Some studies have been conducted on microglia because activated microglial cells play an important role in the proinflammatory status that characterizes neurodegenerative diseases in the central nervous system. Compounds belonging to the lignans family, such as sauquinone, have been shown in LPS-treated BV2 microglial cells to limit iNOS expression by acting on the downregulation of the Akt pathway but not on MAPKs [21]. Resveratrol, on the other hand, activated Akt via phosphorylation together with PTEN and mTOR, with the latter appearing to be the key to the anti-inflammatory activity ascertained in this restudyearch against MAPKs and NF-ĸB activation [22]. A quinolyl-substituted analogue of resveratrol, in addition to acting on MAPKs, has been shown to be able to act upstream by limiting the expression of TLR4 in microglia cells incubated with LPS [23]. Quercetin has been shown to suppress LPS-induced IKK, NF-κB, and AP-1 activation as well as the IFN-γ- induced NF-κB, STAT1, and interferon regulatory factor-1 (IRF-1) activation in BV2 cells. Almost all these factors are upstream of the NF-κB and JAK/STAT signaling pathways, and they downregulate iNOS expression [24]. Still in microglia but in EOC13.31 cells, EGCG acted by promoting Nrf2 expression through the inhibition of the NF-ĸB pathway activated by amyloid β (Aβ) [25]. At the hepatic level, a study in vitro by Kimbrough et al. [26] demonstrated that resveratrol was able to activate JNK phosphorylation and NF-ĸB translocation in isolated rat hepatocytes, but at the same time, iNOS mRNA expression was inhibited as well as the activation of Akt. In male Sprague-Dawley rats administrated with CCl4, chlorogenic acid proved to be able of efficiently inhibit liver fibrosis, and the protective effect was associated with the inhibition of the TLR4/MyD88/NF-ĸB/iNOS signaling pathway [27]. In the same experimental model, oligonol suppressed p65 activation, phosphorylation of MAPKs ERK1/2, JNK, and p38 as well as Akt, thus inhibiting iNOS expression and NO release [28]. In other tissues and organs, resveratrol was the most studied, showing an effect on several signaling pathways. In hypoxia-induced cardiomyocytes, resveratrol showed inhibitory effects on iNOS proteins and mRNA expression, and the suggested mechanism might be associated with HIF-1α inhibition [29]. In a spinal cord ischemia–reperfusion injury (IRI) rat model, it significantly decreased the levels of plasma nitrite/nitrate, iNOS mRNA and protein expressions, and phosphorylation of p38 [30]. Furthermore, resveratrol has been demonstrated to be able to specifically inhibit iNOS induction in muscle through a mechanism involving 5′ adenosine monophosphate-activated protein kinase (AMPK) but not Sirt1 activation in mice myocites and adipocites challenged with LPS [31]. In the context of vascular smooth muscle cell (VSMC) proliferation, which is linked to progression of hypertension and atherosclerosis, resveratrol did not alter iNOS protein level, but it dose-dependently increased the level of iNOS activity, of iNOS cofactor BH4, and of guanosine triphosphate cyclohydrolase I protein, the rate-limiting enzyme in BH4 biosynthesis [32].

2. Activity and Expression of Endothelial Nitric Oxide Synthase

As can be easily understood, most of the studies on the modulation of endothelial nitric oxide synthase (eNOS) by polyphenols have been assessed on endothelial cell cultures and ex vivo cardiovascular tissues. There are several mechanisms that have been investigated in the last twenty years, all of which are related to the maintenance of the basic functionality of eNOS or its recovery following pathological events (Figure 2). The most studied polyphenol is undoubtedly resveratrol, to which dozens of investigations on the subject have been dedicated. The activation of eNOS following phosphorylation is the major event that causes the release of moderate doses of NO useful for maintaining vascular tone and for its antiplatelet properties. As already described above, phosphorylation can occur in different sites and is caused upstream by the activation of signals such as PI3K/Akt, widely studied because it is modulated by polyphenols from different subclasses. In mice with artificial diabetes, the flavonoid morin increased NO levels and endothelial-dependent relaxation responses via Akt signaling, upregulating hosphor-Akt (at Ser473 and Thr308) and consequently hosphor-eNOS (at Ser1177) in aortas [33], as was also seen for protocatechuic acid in the aorta of male hypertensive rats [34]. The increase in the phosphorylation of Akt and eNOS was also observed in rats treated with tyrosol in an in vivo model of myocardial infarction [35]. The activation of Akt/eNOS was also evaluated as a measure of the restoration of endothelial function in diabetic rabbits with carotid damage, where EGCG was able to reactivate this molecular signaling pathway and improve cardiovascular functions [36]. The xanthonoid mangiferin markedly decreased plasma lipids and inflammatory levels in HFD-induced vascular injury in mice, probably via the PTEN/Akt/eNOS pathway, which was activated by mangiferin itself in HUVEC cells that had been previously treated with oxLDL [37]. Other polyphenols such as EGCG, ellagic acid, procyanidins, and some stilbenes have been tested in cell lines of different origins (HAEC, BAEC, HUVEC, and EA.hy926) obtaining similar results in terms of activation of the PI3K/Akt /eNOS pathway [38][39][40][41][42][43]. In addition to the free forms of polyphenols, their phase-II metabolites have also been recently studied by ourthe research group, which has demonstrated how some of the modulatory properties remain intact on the Akt/eNOS phosphorylation pathway, as shown by their precursors, such as hydroxytyrosol, tyrosol, ferulic acid, and some derivatives in HUVEC and HAEC cells [17][44][45]. Phosphorylation of eNOS can also occur at Ser633 site other than at Ser1177, in addition to dephosphorylation at Thr495, which displays the same physiological functions. The activation of these mechanisms has also been observed in human coronary endothelial cells by epicatechin, where the proposed upstream mechanism is the one related to the modulation of the CaMKII pathway, which induces eNOS uncoupling from caveolin-1 [46]. Another signaling pathway involved in eNOS activation and regulated by polyphenols is that of AMPK, which has been shown to be modulated in particular by resveratrol. It led to a significant increase in the phosphorylation of eNOS in vivo in mesenteric arteries from SHRs and Angiotensin-II infused mice [47], whereas in vitro in human endothelial cells, no change regarding its activity was observed [48]. In HAEC cells, the activation of the AMPK/eNOS pathway was instead verified after treatment with rosmarinic acid, which reverted the effects caused by H2O2 incubation [49].
Figure 2. Polyphenols elicit eNOS phosphorylation in endothelial cells through the activation of different upstream kinases. Abbreviations: AMPK—5′ AMP-activated protein kinase; CaMKII—Ca2+/calmodulin dependent kinase II; cGMP—guanosine 3′,5′-cyclic monophosphate; eNOS—endothelial nitric oxide synthase; MAPK—mitogen activated protein kinase; NO—nitric oxide; and PI3K, inositol 1,4,5-triphosphate kinase.
In HAEC and HUVEC, the efficacy of different polyphenols was also evaluated in modulation of the activation of eNOS by regulating the p38 and ERK1/2 MAPKs pathways. This was especially seen for resveratrol, which activated both p38 and ERK1/2 MAPKs in HUVEC in two different experiments [50][51], causing the phosphorylation of Ser1177; whereas in HUVEC and in platelets in vitro, the exact opposite effect was still seen in the context of contrasting the action of VEGF-mediated angiogenesis and oxidative stress [52][53]. The same outcome was observed for the resveratrol derivative piceatannol, which was proposed to bind with VEGF in HUVEC cells, thus attenuating VEGF receptor response and blocking VEGF-mediated downstream signaling pathways, including expressions of ERK1/2 and, consequently, of phosphorylated eNOS [54]. Malvidin was also tested in HUVEC, and it was proven to be effective in the promotion of the release of cGMP starting from the activation of PI3K/Akt/eNOS/NO/cGMP and that of ERK1/2/eNOS/NO/cGMP [55]. The enzymatic activity of eNOS is also regulated by acetylation/deacetylation processes. Short-term treatments of endothelial cells with resveratrol are known to lead to eNOS deacetylation at Lys496 and Lys506 in the CaM-binding domain, leading to an increase in eNOS activity. The mechanisms by which resveratrol is able to induce eNOS deacetylation are through the upregulation of Sirt1, which is usually downregulated by oxidative stress [56]. For this reason, the Sirt1 pathway has been studied in the context of deacetylation and, therefore, activation of eNOS by polyphenols. Frombaum et al. [51] reported, for example, that resveratrol acts on the release of NO also by modulating Sirt1 in HUVEC, as has been observed for salvianolic acid (in HUVEC as well) and for a nitrated derivative of hydroxytyrosol in vivo in diabetic mice [57][58]. In addition to the already mentioned eNOS phosphorylation, consequent to the activation of different signaling pathways, the increase of its expression induced by treatment with different phenolic compounds has been studied. For example, resveratrol was found to be active in increasing eNOS gene expression in a dose-dependent manner in 2T3 and MC3T3 osteoblasts, whereas it did not promote any significant enzyme activation via phosphorylation in the same cells [59]. Similarly, the same compound has improved coronary flow during reperfusion associated with increased eNOS, Sirt1, and hosphor-Akt protein expression in rat hearts [60]. Conversely, resveratrol in HAEC and HPAEC endothelial cells showed instead a suppression of eNOS, measured by RT-PCR [61]. Again, in HAEC, different results were observed after treatment with chlorogenic acid and with the oat derivative avenanthramide-2c, which promoted the expression of eNOS and, in the first case, also its dimerization necessary for NO release [62][63]. In a model of 2K-1C (two kidneys, one clip) rat renovascular hypertension, eNOS expression was restored by curcumin treatment, which simultaneously reduced oxidative stress and hypertension [64].
In addition to its expression at physiologically relevant levels, it is essential that eNOS works correctly by releasing NO. Under certain circumstances, indeed, eNOS itself can release O2●− instead of NO. This phenomenon, eNOS uncoupling, is closely linked to the availability of the crucial eNOS cofactor BH4. ONOO mediated oxidation of BH4 compromises eNOS function and leads to a vicious circle of BH4 destruction, further eNOS uncoupling, and increasing vascular ROS production [65]. In this regard, it was observed that resveratrol, well known as a powerful antioxidant, was able to prevent the uncoupling of eNOS induced by O2●− in hypertensive rats [66].

3. Activity and Expression of Neuronal Nitric Oxide Synthase

The neuronal isoform of NOS and its modulation by dietary polyphenols is still a little-explored field, despite its pharmacological relevance. There are very few investigations, and most of them are in vivo studies that show different pathological aspects related to the activity of nNOS. NO at the neuronal level is known to be implicated in many processes related to memory and learning, but above all, it is implicated in the etiopathogenesis of various neurological diseases. For instance, the implication of NO in Parkinson disease (PD) has been firstly proposed when high levels of nNOS and iNOS were found in the nigrostriatal region and basal ganglia of postmortem PD brains. In PD, the increase of NO levels is caused either by the overexpression of nNOS or by other mechanisms, including glutamate excitotoxicity. NO rapidly reacts with O2●− Anions formed during dopamine metabolism, thus generating ONOO, which is considered one of the main damaging substances in dopaminergic neuronal cells [67]. Therefore, given the growing incidence and mortality of these diseases, the main goal would be to find phenolic compounds which act selectively on this isoform. This would limit its functionality, especially in patients with full-blown pathology, without affecting the activity of other isoforms such as eNOS in the other tissues. In this regard, a polyphenolic extract of green tea that includes EGCG downregulated nNOS expression induced by 6-OHDA exposure to simulate PD conditions in SH-SY5Y cells [68]. An interesting in vivo study in mice, although not focused on the mechanism involved, showed that intervention with EGCG was able to limit the expression of nNOS in brain tissues in the context of memory deficits, characterized by high levels of enzyme expression [69]. The EGCG was also tested in rats, where it showed a dose-dependent suppressive effect on the nNOS expression in the nodose neurons of adult rats after severe hypoxic insult [70]. The same treatment was also carried out in rats prior to crushing their hypoglossal and vagus nerves, which showed elevated levels of nNOS compared to untreated rats. Even in this case, EGCG treatment was effective in restoring the expression levels of nNOS to values similar to those of the control [71]. A significant downregulation of nNOS protein expression along with reduced lipofuscin content was observed in middle-aged and aged rats treated with curcuminoids, suggesting that these compounds may act as a drug candidate for the prevention of deleterious effects of aging and age-associated neurodegenerative disorders [72]. Regarding seizure, it has been noted to some extent how the activity of nNOS can somehow diminish its deleterious effects. Oleuropein, a polyphenol from olives and EVOO, has been studied in this regard for its anticonvulsant properties in mice, and it has been seen that its effectiveness is derived in part from its ability to positively modulate the expression of nNOS. Its anticonvulsant effect was in fact completely reversed by acute pretreatment with L-NAME (a nonselective NOS inhibitor) and 7-NI (a selective inhibitor of nNOS) [73]. Being also found in other tissues, nNOS has been shown to be implicated in various physiological mechanisms outside the central and/or peripheral nervous system. For example, it has been observed how the localization of nNOS in the sarcolemma is essential for maintaining muscle activity and mass, and delocalization of nNOS from sarcolemma represents one major initiating event leading to disuse muscle atrophy. Delocalization of nNOS active molecules from sarcolemma to sarcoplasm has been recognized as a major event favoring increased NO availability at this site and improving activation by nitrosylation of atrophy regulators [74]. In this regard, Vitadello et al. [75] showed that curcumin administration to female Wistar rats counteracted the loss of mass and force by involving the Grp94 chaperone and mechanistically preserving the sarcolemmal localization of nNOS. In rats with fructose-induced hypertension, resveratrol activated nNOS to release NO to dilate large caliber vessels. Resveratrol effect was exerted through the modulation of ERK1/2-RSK-nNOS pathway by activating AMPK to negatively regulate Rac1-induced NADPH oxidase levels [76].

4. Simultaneous Activity of Dietary Polyphenols on Different Nitric Oxide Synthase Isoforms

The modulatory action on the mechanisms leading to the expression and/or activation/deactivation of the three NOS isoforms indicates that polyphenols can simultaneously perform different functions based on the tissue and the NOS isoform involved. Several studies have shown interesting implications from the perspective of the selectivity of polyphenols towards a specific NOS isoform, thus leading to relevant biological effects [Table 1]. As already mentioned above, resveratrol has been the main object of studies regarding the interaction between polyphenols and the biological activity of NOS isoforms. It has been shown to be able to inhibit cell growth and to promote apoptosis by the elevation of NO in human melanoma A375 cells, and Western blot analysis showed the expression of all the three NOS isoforms, all of which were increased [77]. Resveratrol showed concentration-related increases in eNOS and a simultaneous reduction in iNOS expression in human glaucomatous trabecular meshwork (TM) cells [78]. At the physiological level, it was also tested over the long term to evaluate its impact on endothelial function and on the activity of eNOS and iNOS. Overall, it was seen that it was able to increase the expression of eNOS in endothelial HUVEC but not its activation by phosphorylation, and at the same time, no significant effect occurred with regard to the expression of iNOS [79]. At the endothelial level, a hydroxylated analogue of resveratrol, i.e., piceatannol, was tested against the deleterious action of H2O2 in H9c2 cardiomyocytes. Piceatannol pretreatment was able to regulate PI3K-Akt-eNOS signaling pathway to alleviate peroxidative injury, and in parallel, it decreased iNOS expression [80].

References

  1. Hoesel, B.; Schmid, J.A. The complexity of NF-κB signaling in inflammation and cancer. Mol. Cancer 2013, 12, 86.
  2. Solt, L.A.; May, M.J. The IκB kinase complex: Master regulator of NF-κB signaling. Immunol. Res. 2008, 42, 3–18.
  3. Boronat, A.; Rodriguez-Morato, J.; Serreli, G.; Fito, M.; Tyndale, R.F.; Deiana, M.; de la Torre, R. Contribution of Biotransformations Carried Out by the Microbiota, Drug-Metabolizing Enzymes, and Transport Proteins to the Biological Activities of Phytochemicals Found in the Diet. Adv. Nutr. 2021, 12, 2172–2189.
  4. Serreli, G.; Melis, M.P.; Zodio, S.; Naitza, M.R.; Casula, E.; Penalver, P.; Lucas, R.; Loi, R.; Morales, J.C.; Deiana, M. Altered paracellular permeability in intestinal cell monolayer challenged with lipopolysaccharide: Modulatory effects of pterostilbene metabolites. Food Chem. Toxicol. 2020, 145, 111729.
  5. Rosillo, M.A.; Sanchez-Hidalgo, M.; Cardeno, A.; de la Lastra, C.A. Protective effect of ellagic acid, a natural polyphenolic compound, in a murine model of Crohn’s disease. Biochem. Pharmacol. 2011, 82, 737–745.
  6. Ukil, A.; Maity, S.; Das, P.K. Protection from experimental colitis by theaflavin-3,3′-digallate correlates with inhibition of IKK and NF-κB activation. Br. J. Pharmacol. 2006, 149, 121–131.
  7. Camacho-Barquero, L.; Villegas, I.; Sanchez-Calvo, J.M.; Talero, E.; Sanchez-Fidalgo, S.; Motilva, V.; Alarcon de la Lastra, C. Curcumin, a Curcuma longa constituent, acts on MAPK p38 pathway modulating COX-2 and iNOS expression in chronic experimental colitis. Int. Immunopharmacol. 2007, 7, 333–342.
  8. Sánchez-Calvo, J.M.; Villegas, I.; Sánchez-Fidalgo, S.; Camacho-Barquero, L.; Talero, E.; Motilva, V.; Alarcón de la Lastra, C. Protective effect of curcumin, aCurcuma longaconstituent, in early colonic inflammation in rats. Drug Dev. Res. 2009, 70, 425–437.
  9. Pandurangan, A.K.; Mohebali, N.; Esa, N.M.; Looi, C.Y.; Ismail, S.; Saadatdoust, Z. Gallic acid suppresses inflammation in dextran sodium sulfate-induced colitis in mice: Possible mechanisms. Int. Immunopharmacol. 2015, 28, 1034–1043.
  10. Dong, W.; Li, F.; Pan, Z.; Liu, S.; Yu, H.; Wang, X.; Bi, S.; Zhang, W. Resveratrol ameliorates subacute intestinal ischemia-reperfusion injury. J. Surg. Res. 2013, 185, 182–189.
  11. Serra, D.; Paixao, J.; Nunes, C.; Dinis, T.C.; Almeida, L.M. Cyanidin-3-glucoside suppresses cytokine-induced inflammatory response in human intestinal cells: Comparison with 5-aminosalicylic acid. PLoS ONE 2013, 8, e73001.
  12. Nunes, C.; Almeida, L.; Barbosa, R.M.; Laranjinha, J. Luteolin suppresses the JAK/STAT pathway in a cellular model of intestinal inflammation. Food Funct. 2017, 8, 387–396.
  13. Serra, D.; Rufino, A.T.; Mendes, A.F.; Almeida, L.M.; Dinis, T.C. Resveratrol modulates cytokine-induced Jak/STAT activation more efficiently than 5-aminosalicylic acid: An in vitro approach. PLoS ONE 2014, 9, e109048.
  14. Panaro, M.A.; Carofiglio, V.; Acquafredda, A.; Cavallo, P.; Cianciulli, A. Anti-inflammatory effects of resveratrol occur via inhibition of lipopolysaccharide-induced NF-κB activation in Caco-2 and SW480 human colon cancer cells. Br. J. Nutr. 2012, 108, 1623–1632.
  15. Serreli, G.; Melis, M.P.; Corona, G.; Deiana, M. Modulation of LPS-induced nitric oxide production in intestinal cells by hydroxytyrosol and tyrosol metabolites: Insight into the mechanism of action. Food Chem. Toxicol. 2019, 125, 520–527.
  16. Serra, G.; Incani, A.; Serreli, G.; Porru, L.; Melis, M.P.; Tuberoso, C.I.G.; Rossin, D.; Biasi, F.; Deiana, M. Olive oil polyphenols reduce oxysterols -induced redox imbalance and pro-inflammatory response in intestinal cells. Redox Biol. 2018, 17, 348–354.
  17. Serreli, G.; Naitza, M.R.; Zodio, S.; Leoni, V.P.; Spada, M.; Melis, M.P.; Boronat, A.; Deiana, M. Ferulic Acid Metabolites Attenuate LPS-Induced Inflammatory Response in Enterocyte-like Cells. Nutrients 2021, 13, 3152.
  18. Zheng, W.; Feng, Z.; You, S.; Zhang, H.; Tao, Z.; Wang, Q.; Chen, H.; Wu, Y. Fisetin inhibits IL-1β-induced inflammatory response in human osteoarthritis chondrocytes through activating SIRT1 and attenuates the progression of osteoarthritis in mice. Int. Immunopharmacol. 2017, 45, 135–147.
  19. Singh, R.; Ahmed, S.; Islam, N.; Goldberg, V.M.; Haqqi, T.M. Epigallocatechin-3-gallate inhibits interleukin-1β-induced expression of nitric oxide synthase and production of nitric oxide in human chondrocytes: Suppression of nuclear factor κB activation by degradation of the inhibitor of nuclear factor κB. Arthritis Rheum. 2002, 46, 2079–2086.
  20. Lei, M.; Wang, J.G.; Xiao, D.M.; Fan, M.; Wang, D.P.; Xiong, J.Y.; Chen, Y.; Ding, Y.; Liu, S.L. Resveratrol inhibits interleukin 1β-mediated inducible nitric oxide synthase expression in articular chondrocytes by activating SIRT1 and thereby suppressing nuclear factor-κB activity. Eur. J. Pharmacol. 2012, 674, 73–79.
  21. Jang, E.Y.; Yang, C.H.; Han, M.H.; Choi, Y.H.; Hwang, M. Sauchinone suppresses lipopolysaccharide-induced inflammatory responses through Akt signaling in BV2 cells. Int. Immunopharmacol. 2012, 14, 188–194.
  22. Zhong, L.M.; Zong, Y.; Sun, L.; Guo, J.Z.; Zhang, W.; He, Y.; Song, R.; Wang, W.M.; Xiao, C.J.; Lu, D. Resveratrol inhibits inflammatory responses via the mammalian target of rapamycin signaling pathway in cultured LPS-stimulated microglial cells. PLoS ONE 2012, 7, e32195.
  23. Hou, Y.; Zhang, Y.; Mi, Y.; Wang, J.; Zhang, H.; Xu, J.; Yang, Y.; Liu, J.; Ding, L.; Yang, J.; et al. A Novel Quinolyl-Substituted Analogue of Resveratrol Inhibits LPS-Induced Inflammatory Responses in Microglial Cells by Blocking the NF-κB/MAPK Signaling Pathways. Mol. Nutr. Food Res. 2019, 63, e1801380.
  24. Chen, J.C.; Ho, F.M.; Pei-Dawn Lee, C.; Chen, C.P.; Jeng, K.C.; Hsu, H.B.; Lee, S.T.; Wen Tung, W.; Lin, W.W. Inhibition of iNOS gene expression by quercetin is mediated by the inhibition of IκB kinase, nuclear factor-kappa B and STAT1, and depends on heme oxygenase-1 induction in mouse BV-2 microglia. Eur. J. Pharmacol. 2005, 521, 9–20.
  25. Cheng-Chung Wei, J.; Huang, H.C.; Chen, W.J.; Huang, C.N.; Peng, C.H.; Lin, C.L. Epigallocatechin gallate attenuates amyloid β-induced inflammation and neurotoxicity in EOC 13.31 microglia. Eur. J. Pharmacol. 2016, 770, 16–24.
  26. Kimbrough, C.W.; Lakshmanan, J.; Matheson, P.J.; Woeste, M.; Gentile, A.; Benns, M.V.; Zhang, B.; Smith, J.W.; Harbrecht, B.G. Resveratrol decreases nitric oxide production by hepatocytes during inflammation. Surgery 2015, 158, 1095–1101.
  27. Shi, H.; Dong, L.; Jiang, J.; Zhao, J.; Zhao, G.; Dang, X.; Lu, X.; Jia, M. Chlorogenic acid reduces liver inflammation and fibrosis through inhibition of toll-like receptor 4 signaling pathway. Toxicology 2013, 303, 107–114.
  28. Bak, J.; Je, N.K.; Chung, H.Y.; Yokozawa, T.; Yoon, S.; Moon, J.O. Oligonol Ameliorates CCl(4)-Induced Liver Injury in Rats via the NF-Kappa B and MAPK Signaling Pathways. Oxidative Med. Cell. Longev. 2016, 2016, 3935841.
  29. Wang, S.; Qian, Y.; Gong, D.; Zhang, Y.; Fan, Y. Resveratrol attenuates acute hypoxic injury in cardiomyocytes: Correlation with inhibition of iNOS-NO signaling pathway. Eur. J. Pharm. Sci. 2011, 44, 416–421.
  30. Fu, S.; Lv, R.; Wang, L.; Hou, H.; Liu, H.; Shao, S. Resveratrol, an antioxidant, protects spinal cord injury in rats by suppressing MAPK pathway. Saudi J. Biol. Sci. 2018, 25, 259–266.
  31. Centeno-Baez, C.; Dallaire, P.; Marette, A. Resveratrol inhibition of inducible nitric oxide synthase in skeletal muscle involves AMPK but not SIRT1. Am. J. Physiol. Endocrinol. Metab. 2011, 301, 922–930.
  32. Ekshyyan, V.P.; Hebert, V.Y.; Khandelwal, A.; Dugas, T.R. Resveratrol inhibits rat aortic vascular smooth muscle cell proliferation via estrogen receptor dependent nitric oxide production. J. Cardiovasc. Pharmacol. 2007, 50, 83–93.
  33. Taguchi, K.; Hida, M.; Hasegawa, M.; Matsumoto, T.; Kobayashi, T. Dietary polyphenol morin rescues endothelial dysfunction in a diabetic mouse model by activating the Akt/eNOS pathway. Mol. Nutr. Food Res. 2016, 60, 580–588.
  34. Masodsai, K.; Lin, Y.Y.; Chaunchaiyakul, R.; Su, C.T.; Lee, S.D.; Yang, A.L. Twelve-Week Protocatechuic Acid Administration Improves Insulin-Induced and Insulin-Like Growth Factor-1-Induced Vasorelaxation and Antioxidant Activities in Aging Spontaneously Hypertensive Rats. Nutrients 2019, 11, 699.
  35. Samuel, S.M.; Thirunavukkarasu, M.; Penumathsa, S.V.; Paul, D.; Maulik, N. Akt/FOXO3a/SIRT1-mediated cardioprotection by n-tyrosol against ischemic stress in rat in vivo model of myocardial infarction: Switching gears toward survival and longevity. J. Agric. Food Chem. 2008, 56, 9692–9698.
  36. Huang, H.; Jin, C.Y.; Bi, X.K.; Zhao, Y.B.; Xu, S.J.; Wang, M.H.; Yu, L.; Sun, Y.X.; Hu, D. Green Tea Polyphenol Epigallocatechin-3-Gallate Promotes Reendothelialization in Carotid Artery of Diabetic Rabbits by Reactivating Akt/eNOS Pathway. Front. Pharmacol. 2018, 9, 1305.
  37. Jiang, F.; Zhang, D.L.; Jia, M.; Hao, W.H.; Li, Y.J. Mangiferin inhibits high-fat diet induced vascular injury via regulation of PTEN/AKT/eNOS pathway. J. Pharmacol. Sci. 2018, 137, 265–273.
  38. Ou, H.C.; Lee, W.J.; Lee, S.D.; Huang, C.Y.; Chiu, T.H.; Tsai, K.L.; Hsu, W.C.; Sheu, W.H. Ellagic acid protects endothelial cells from oxidized low-density lipoprotein-induced apoptosis by modulating the PI3K/Akt/eNOS pathway. Toxicol. Appl. Pharmacol. 2010, 248, 134–143.
  39. Lu, Y.A.; Jiang, Y.; Yang, H.W.; Hwang, J.; Jeon, Y.J.; Ryu, B. Diphlorethohydroxycarmalol Isolated from Ishige okamurae Exerts Vasodilatory Effects via Calcium Signaling and PI3K/Akt/eNOS Pathway. Int. J. Mol. Sci. 2021, 22, 1610.
  40. 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.
  41. Fernandez-Cruz, E.; Cerezo, A.B.; Cantos-Villar, E.; Richard, T.; Troncoso, A.M.; Garcia-Parrilla, M.C. Inhibition of VEGFR-2 Phosphorylation and Effects on Downstream Signaling Pathways in Cultivated Human Endothelial Cells by Stilbenes from Vitis spp. J. Agric. Food Chem. 2019, 67, 3909–3918.
  42. Byun, E.B.; Ishikawa, T.; Suyama, A.; Kono, M.; Nakashima, S.; Kanda, T.; Miyamoto, T.; Matsui, T. A procyanidin trimer, C1, promotes NO production in rat aortic endothelial cells via both hyperpolarization and PI3K/Akt pathways. Eur. J. Pharmacol. 2012, 692, 52–60.
  43. Auger, C.; Kim, J.H.; Chabert, P.; Chaabi, M.; Anselm, E.; Lanciaux, X.; Lobstein, A.; Schini-Kerth, V.B. The EGCg-induced redox-sensitive activation of endothelial nitric oxide synthase and relaxation are critically dependent on hydroxyl moieties. Biochem. Biophys. Res. Commun. 2010, 393, 162–167.
  44. Serreli, G.; Le Sayec, M.; Diotallevi, C.; Teissier, A.; Deiana, M.; Corona, G. Conjugated Metabolites of Hydroxytyrosol and Tyrosol Contribute to the Maintenance of Nitric Oxide Balance in Human Aortic Endothelial Cells at Physiologically Relevant Concentrations. Molecules 2021, 26, 7480.
  45. Serreli, G.; Le Sayec, M.; Thou, E.; Lacour, C.; Diotallevi, C.; Dhunna, M.A.; Deiana, M.; Spencer, J.P.E.; Corona, G. Ferulic Acid Derivatives and Avenanthramides Modulate Endothelial Function through Maintenance of Nitric Oxide Balance in HUVEC Cells. Nutrients 2021, 13, 2026.
  46. Ramirez-Sanchez, I.; Maya, L.; Ceballos, G.; Villarreal, F. (-)-epicatechin activation of endothelial cell endothelial nitric oxide synthase, nitric oxide, and related signaling pathways. Hypertension 2010, 55, 1398–1405.
  47. Dolinsky, V.W.; Chakrabarti, S.; Pereira, T.J.; Oka, T.; Levasseur, J.; Beker, D.; Zordoky, B.N.; Morton, J.S.; Nagendran, J.; Lopaschuk, G.D.; et al. Resveratrol prevents hypertension and cardiac hypertrophy in hypertensive rats and mice. Biochim. Biophys. Acta 2013, 1832, 1723–1733.
  48. Elies, J.; Cuinas, A.; Garcia-Morales, V.; Orallo, F.; Campos-Toimil, M. Trans-resveratrol simultaneously increases cytoplasmic Ca(2+) levels and nitric oxide release in human endothelial cells. Mol. Nutr. Food Res. 2011, 55, 1237–1248.
  49. Zhou, H.; Fu, B.; Xu, B.; Mi, X.; Li, G.; Ma, C.; Xie, J.; Li, J.; Wang, Z. Rosmarinic Acid Alleviates the Endothelial Dysfunction Induced by Hydrogen Peroxide in Rat Aortic Rings via Activation of AMPK. Oxidative Med. Cell. Longev. 2017, 2017, 7091904.
  50. Klinge, C.M.; Blankenship, K.A.; Risinger, K.E.; Bhatnagar, S.; Noisin, E.L.; Sumanasekera, W.K.; Zhao, L.; Brey, D.M.; Keynton, R.S. Resveratrol and estradiol rapidly activate MAPK signaling through estrogen receptors α and β in endothelial cells. J. Biol. Chem. 2005, 280, 7460–7468.
  51. Frombaum, M.; Le Clanche, S.; Bonnefont-Rousselot, D.; Borderie, D. Antioxidant effects of resveratrol and other stilbene derivatives on oxidative stress and *NO bioavailability: Potential benefits to cardiovascular diseases. Biochimie 2012, 94, 269–276.
  52. Hu, W.H.; Chan, G.K.; Duan, R.; Wang, H.Y.; Kong, X.P.; Dong, T.T.; Tsim, K.W. Synergy of Ginkgetin and Resveratrol in Suppressing VEGF-Induced Angiogenesis: A Therapy in Treating Colorectal Cancer. Cancers 2019, 11, 1828.
  53. Gresele, P.; Pignatelli, P.; Guglielmini, G.; Carnevale, R.; Mezzasoma, A.M.; Ghiselli, A.; Momi, S.; Violi, F. Resveratrol, at concentrations attainable with moderate wine consumption, stimulates human platelet nitric oxide production. J. Nutr. 2008, 138, 1602–1608.
  54. Hu, W.H.; Dai, D.K.; Zheng, B.Z.; Duan, R.; Dong, T.T.; Qin, Q.W.; Tsim, K.W. Piceatannol, a Natural Analog of Resveratrol, Exerts Anti-angiogenic Efficiencies by Blockage of Vascular Endothelial Growth Factor Binding to Its Receptor. Molecules 2020, 25, 3769.
  55. Quintieri, A.M.; Baldino, N.; Filice, E.; Seta, L.; Vitetti, A.; Tota, B.; De Cindio, B.; Cerra, M.C.; Angelone, T. Malvidin, a red wine polyphenol, modulates mammalian myocardial and coronary performance and protects the heart against ischemia/reperfusion injury. J. Nutr. Biochem. 2013, 24, 1221–1231.
  56. Xia, N.; Forstermann, U.; Li, H. Resveratrol and endothelial nitric oxide. Molecules 2014, 19, 16102–16121.
  57. Zhai, J.; Qu, X.; Zhang, Y.; Gao, H.; Tao, L.; Song, Y.; Zhang, S. Salvianolic acid inhibits the effects of high glucose on vascular endothelial dysfunction by modulating the Sirt1-eNOS pathway. J. Biochem. Mol. Toxicol. 2019, 33, e22245.
  58. Wang, W.; Shang, C.; Zhang, W.; Jin, Z.; Yao, F.; He, Y.; Wang, B.; Li, Y.; Zhang, J.; Lin, R. Hydroxytyrosol NO regulates oxidative stress and NO production through SIRT1 in diabetic mice and vascular endothelial cells. Phytomedicine 2019, 52, 206–215.
  59. Zhao, M.; Ko, S.Y.; Garrett, I.R.; Mundy, G.R.; Gutierrez, G.E.; Edwards, J.R. The polyphenol resveratrol promotes skeletal growth in mice through a sirtuin 1-bone morphogenic protein 2 longevity axis. Br. J. Pharmacol. 2018, 175, 4183–4192.
  60. Fourny, N.; Lan, C.; Seree, E.; Bernard, M.; Desrois, M. Protective Effect of Resveratrol against Ischemia-Reperfusion Injury via Enhanced High Energy Compounds and eNOS-SIRT1 Expression in Type 2 Diabetic Female Rat Heart. Nutrients 2019, 11, 105.
  61. Hsieh, T.C.; Lu, X.; Guo, J.; Wu, J.M. Differential regulation of proliferation, cell cycle control and gene expression in cultured human aortic and pulmonary artery endothelial cells by resveratrol. Int. J. Mol. Med. 2010, 26, 743–749.
  62. Nie, L.; Wise, M.L.; Peterson, D.M.; Meydani, M. Avenanthramide, a polyphenol from oats, inhibits vascular smooth muscle cell proliferation and enhances nitric oxide production. Atherosclerosis 2006, 186, 260–266.
  63. Jiang, R.; Hodgson, J.M.; Mas, E.; Croft, K.D.; Ward, N.C. Chlorogenic acid improves ex vivo vessel function and protects endothelial cells against HOCl-induced oxidative damage, via increased production of nitric oxide and induction of Hmox-1. J. Nutr. Biochem. 2016, 27, 53–60.
  64. 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.
  65. Schmitt, C.A.; Dirsch, V.M. Modulation of endothelial nitric oxide by plant-derived products. Nitric Oxide 2009, 21, 77–91.
  66. 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.
  67. Aquilano, K.; Baldelli, S.; Rotilio, G.; Ciriolo, M.R. Role of Nitric Oxide Synthases in Parkinson’s Disease: A Review on the Antioxidant and Anti-inflammatory Activity of Polyphenols. Neurochem. Res. 2008, 33, 2416–2426.
  68. Guo, S.; Bezard, E.; Zhao, B. Protective effect of green tea polyphenols on the SH-SY5Y cells against 6-OHDA induced apoptosis through ROS–NO pathway. Free Radic. Biol. Med. 2005, 39, 682–695.
  69. Ding, L.; Gao, X.; Hu, J.; Yu, S. (-)Epigallocatechin-3-gallate attenuates anesthesia-induced memory deficit in young mice via modulation of nitric oxide expression. Mol. Med. Rep. 2018, 18, 4813–4820.
  70. Wei, I.H.; Wu, Y.C.; Wen, C.Y.; Shieh, J.Y. Green tea polyphenol (-)-epigallocatechin gallate attenuates the neuronal NADPH-d/nNOS expression in the nodose ganglion of acute hypoxic rats. Brain Res. 2004, 999, 73–80.
  71. Wei, I.H.; Tu, H.C.; Huang, C.C.; Tsai, M.H.; Tseng, C.Y.; Shieh, J.Y. (-)-Epigallocatechin gallate attenuates NADPH-d/nNOS expression in motor neurons of rats following peripheral nerve injury. BMC Neurosci. 2011, 12, 52.
  72. Rastogi, M.; Ojha, R.P.; Sagar, C.; Agrawal, A.; Dubey, G.P. Protective effect of curcuminoids on age-related mitochondrial impairment in female Wistar rat brain. Biogerontology 2014, 15, 21–31.
  73. Rahimi, N.; Delfan, B.; Motamed-Gorji, N.; Dehpour, A.R. Effects of oleuropein on pentylenetetrazol-induced seizures in mice: Involvement of opioidergic and nitrergic systems. J. Nat. Med. 2017, 71, 389–396.
  74. Vitadello, M.; Gherardini, J.; Gorza, L. The stress protein/chaperone Grp94 counteracts muscle disuse atrophy by stabilizing subsarcolemmal neuronal nitric oxide synthase. Antioxid. Redox Signal. 2014, 20, 2479–2496.
  75. Vitadello, M.; Germinario, E.; Ravara, B.; Libera, L.D.; Danieli-Betto, D.; Gorza, L. Curcumin counteracts loss of force and atrophy of hindlimb unloaded rat soleus by hampering neuronal nitric oxide synthase untethering from sarcolemma. J. Physiol. 2014, 592, 2637–2652.
  76. Yeh, T.C.; Shin, C.S.; Chen, H.H.; Lai, C.C.; Sun, G.C.; Tseng, C.J.; Cheng, P.W. Resveratrol regulates blood pressure by enhancing AMPK signaling to downregulate a Rac1-derived NADPH oxidase in the central nervous system. J. Appl. Physiol. 2018, 125, 40–48.
  77. Kim, M.Y. Nitric oxide triggers apoptosis in A375 human melanoma cells treated with capsaicin and resveratrol. Mol. Med. Rep. 2012, 5, 585–591.
  78. Avotri, S.; Eatman, D.; Russell-Randall, K. Effects of Resveratrol on Inflammatory Biomarkers in Glaucomatous Human Trabecular Meshwork Cells. Nutrients 2019, 11, 984.
  79. Takahashi, S.; Nakashima, Y. Repeated and long-term treatment with physiological concentrations of resveratrol promotes NO production in vascular endothelial cells. Br. J. Nutr. 2012, 107, 774–780.
  80. Wang, D.; Zhang, Y.; Zhang, C.; Gao, L.; Li, J. Piceatannol pretreatment alleviates acute cardiac injury via regulating PI3K-Akt-eNOS signaling in H9c2 cells. Biomed. Pharmacother. 2019, 109, 886–891.
  81. Babacanoglu, C.; Yildirim, N.; Sadi, G.; Pektas, M.B.; Akar, F. Resveratrol prevents high-fructose corn syrup-induced vascular insulin resistance and dysfunction in rats. Food Chem. Toxicol. 2013, 60, 160–167.
  82. Zhang, H.; Morgan, B.; Potter, B.J.; Ma, L.; Dellsperger, K.C.; Ungvari, Z.; Zhang, C. Resveratrol improves left ventricular diastolic relaxation in type 2 diabetes by inhibiting oxidative/nitrative stress: In vivo demonstration with magnetic resonance imaging. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, 985–994.
  83. Fukuda, S.; Kaga, S.; Zhan, L.; Bagchi, D.; Das, D.K.; Bertelli, A.; Maulik, N. Resveratrol ameliorates myocardial damage by inducing vascular endothelial growth factor-angiogenesis and tyrosine kinase receptor Flk-1. Cell Biochem. Biophys. 2006, 44, 43–49.
  84. Oktem, G.; Uysal, A.; Oral, O.; Sezer, E.D.; Olukman, M.; Erol, A.; Akgur, S.A.; Bilir, A. Resveratrol attenuates doxorubicin-induced cellular damage by modulating nitric oxide and apoptosis. Exp. Toxicol. Pathol. 2012, 64, 471–479.
  85. Jia, E.; Yan, Y.; Zhou, M.; Li, X.; Jiang, G.; Liu, W.; Zhang, D. Combined effects of dietary quercetin and resveratrol on growth performance, antioxidant capability and innate immunity of blunt snout bream (Megalobrama amblycephala). Anim. Feed. Sci. Technol. 2019, 256, 114268.
  86. Acquaviva, R.; Lanteri, R.; Li Destri, G.; Caltabiano, R.; Vanella, L.; Lanzafame, S.; Di Cataldo, A.; Li Volti, G.; Di Giacomo, C. Beneficial effects of rutin and L-arginine coadministration in a rat model of liver ischemia-reperfusion injury. Am. J. Physiol. Gastrointest. Liver Physiol. 2009, 296, 664–670.
  87. Adhikary, B.; Yadav, S.K.; Bandyopadhyay, S.K.; Chattopadhyay, S. Role of the COX-independent pathways in the ulcer-healing action of epigallocatechin gallate. Food Funct. 2011, 2, 338–347.
  88. Steffen, Y.; Jung, T.; Klotz, L.O.; Schewe, T.; Grune, T.; Sies, H. Protein modification elicited by oxidized low-density lipoprotein (LDL) in endothelial cells: Protection by (-)-epicatechin. Free Radic. Biol. Med. 2007, 42, 955–970.
  89. Franceschelli, S.; Pesce, M.; Ferrone, A.; Gatta, D.M.; Patruno, A.; Lutiis, M.A.; Quiles, J.L.; Grilli, A.; Felaco, M.; Speranza, L. Biological Effect of Licochalcone C on the Regulation of PI3K/Akt/eNOS and NF-κB/iNOS/NO Signaling Pathways in H9c2 Cells in Response to LPS Stimulation. Int. J. Mol. Sci. 2017, 18, 690.
  90. Abdel Aziz, M.T.; Motawi, T.; Rezq, A.; Mostafa, T.; Fouad, H.H.; Ahmed, H.H.; Rashed, L.; Sabry, D.; Senbel, A.; Al-Malki, A.; et al. Effects of a water-soluble curcumin protein conjugate vs. pure curcumin in a diabetic model of erectile dysfunction. J. Sex. Med. 2012, 9, 1815–1833.
More