Next Article in Journal
From a Molecule to a Drug: Chemical Features Enhancing Pharmacological Potential
Next Article in Special Issue
Effects of Different Extraction Methods in Pharmacopoeia on the Content and Structure Transformation of Ginsenosides
Previous Article in Journal
Essential Oils in Respiratory Mycosis: A Review
Previous Article in Special Issue
Identification of a Hydrogen-Sulfide-Releasing Isochroman-4-One Hybrid as a Cardioprotective Candidate for the Treatment of Cardiac Hypertrophy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Therapeutic Potential of Kaemferol and Other Naturally Occurring Polyphenols Might Be Modulated by Nrf2-ARE Signaling Pathway: Current Status and Future Direction

1
College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
2
Department of Pharmacy, Bashir Institute of Health Sciences, Islamabad 45400, Pakistan
3
Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
4
Department of Clinical Laboratory, College of Applied Medical Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
5
Department of Allied Health Sciences, Bashir Institute of Health Sciences, Islamabad 45400, Pakistan
6
Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY 10463, USA
7
Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy
*
Authors to whom correspondence should be addressed.
Molecules 2022, 27(13), 4145; https://doi.org/10.3390/molecules27134145
Submission received: 21 April 2022 / Revised: 13 June 2022 / Accepted: 20 June 2022 / Published: 28 June 2022
(This article belongs to the Special Issue Natural Compounds: A Lead for Drug Discovery and Development)

Abstract

:
Kaempferol is a natural flavonoid, which has been widely investigated in the treatment of cancer, cardiovascular diseases, metabolic complications, and neurological disorders. Nrf2 (nuclear factor erythroid 2-related factor 2) is a transcription factor involved in mediating carcinogenesis and other ailments, playing an important role in regulating oxidative stress. The activation of Nrf2 results in the expression of proteins and cytoprotective enzymes, which provide cellular protection against reactive oxygen species. Phytochemicals, either alone or in combination, have been used to modulate Nrf2 in cancer and other ailments. Among them, kaempferol has been recently explored for its anti-cancer and other anti-disease therapeutic efficacy, targeting Nrf2 modulation. In combating cancer, diabetic complications, metabolic disorders, and neurological disorders, kaempferol has been shown to regulate Nrf2 and reduce redox homeostasis. In this context, this review article highlights the current status of the therapeutic potential of kaempferol by targeting Nrf2 modulation in cancer, diabetic complications, neurological disorders, and cardiovascular disorders. In addition, we provide future perspectives on kaempferol targeting Nrf2 modulation as a potential therapeutic approach.

1. Introduction

Nrf2 (nuclear factor erythroid 2-related factor 2) is a transcription factor involved in carcinogenesis and other diseases, playing an important role in modulating redox status and inflammation [1]. In the context of oncology, Nrf2 targeting has been considered an important chemotherapy and chemoprevention strategy [2]. It has been demonstrated that the modulation of Nrf2 leads to cell protection from cancer promotion and initiation [3]. The enhanced activation and overexpression of Nrf2 renders the cells resistant to radiotherapy and conventional chemotherapy [4]. In addition, the quenching and production of reactive oxygen species and free radicals-related intracellular/extracellular signaling needs to be strictly regulated upon cellular damage [5]. Nrf2 also plays a critical role in cardiovascular disease [6], and Nrf2 targeted activation might open new avenues in cardiovascular disease therapeutics. In addition to carcinogenesis and cardiovascular disease, studies have shown the role Nrf2 in diabetes complications and neurological disorders [7,8,9].
Natural products based phytochemicals increase cancer cell death primarily by regulating Nrf2 [10], leading to pro-autophagy, oxidative stress, pro-apoptosis and inhibition of expression of cytoprotective genes [11]. Several phytochemicals exert their anticancer or chemo preventive role by Nrf2 modulation [12]. These include quercetin, curcumin, and resveratrol [13]. Natural Nrf2 inhibitors, such as luteolin, malabaricone, wogonin, and ascorbic acid reduce the production of Nrf2 and thus offer chemo sensitizer and anti-carcinogenic activity in tumors [14].
Kaempferol is a natural flavonoid extracted from tea. Other sources include kale, grapes, citrus fruits, vegetables, beans, apple, broccoli, and tomatoes. Physically, it is crystalline in nature with pure yellow color. It is slightly soluble in water [15], and it has shown efficacy as an anticancer, anti-inflammatory, and anti-arthritic compound, given its antioxidant and antiviral activity [16,17,18].
This review article highlights the therapeutic role of kaempferol in cancer, cardiovascular diseases, diabetic complications, and neurological disorders by targeting Nrf2 modulation. The current status of kaempferol targeting Nrf2 modulation is underpinned along with the future directions that need further exploration of this versatile phytochemical.

2. Nrf2 Activation/Inhibition

Nrf2 (nuclear factor erythroid 2-related factor 2) activation results in the expression of several proteins and cytoprotective enzymes that provide cellular protection against reactive oxygen species (ROS), thus offering cytoprotection [19]. Nrf2 is thus believed to be the first defense line against agents that cause cancer initiation and promotion.
Nrf2 under normal conditions is sequestered in cytoplasm and attached to its repressor receptor known as Keap1 (Kelch-like ECH-associated protein 1) [20]. Beyond its function in cell protection, it is implicated in a wide network regulating anti-inflammatory response and metabolic reprogramming, i.e., a key regulator of cell fate and a strategic player in the control of cell transformation and response to viral infections [21]. Several phytochemicals, such as isothiocyanate, act as Nrf2 activators, binding to the SH group of keap-1 protein and, in turn, leading to inhibition of Nrf2 degradation. Under oxidant stress conditions, Nrf2 is translocated to the nucleus, where it induces a variety of genes involved in the antioxidant defense [22].
Several protein kinases, i.e., AMP-activated protein kinases, tyrosine protein kinases, and mitogen-activated protein kinases, lead to Nrf2 post-translational modifications that trigger the release of Nrf2. These protein kinases phosphorylate Nrf2, which, in turn, modulates its activity and stability. Such phosphorylation and consecutive activation of Nrf2 leads to therapeutic potential in inflammatory disorders [23]. The p62 protein is an autophagy receptor for the degradation of proteins and mitochondria. Keap 1 is degraded after interaction with p62 protein via autophagy, in turn leading to stabilization of Nrf2 [24,25]. On the other hand, increased phosphorylation of p62 and autophagy impairment lead to Nrf2 activation, promoting proliferation of cancer cells. Nrf2 protein levels are also regulated by epigenetic mechanisms, such as microRNA and methylation of Keap 1 promoters [26].
Nrf2 also contributes to chemoresistance, proliferation, and invasion [27]. Thus, Nrf2 pro-oncogenic activation in cancer is associated with mechanisms that involve both genetic and epigenetic alterations [28]. Nrf2 leads to cancer cell proliferation following certain metabolic re-programming [29]. In normal cells, Nrf2 signaling cascades afford defensive mechanisms, while attenuated Nrf2 levels lead to tumorigenesis [30,31]. For example, Nrf2-null mice were found more susceptible to exposure to carcinogens [32]. Nrf2 is the key transcription factor regulating antioxidant and xenobiotic exposure response. When oxidative stress increases, Nrf2 translocates to the cell nucleus and forms heterodimer with small Maf (sMaf) proteins. Nrf2/sMaf heterodimer binds specifically to a cis-acting enhancer called antioxidant response element (ARE) and initiates transcription of genes encoding antioxidant and detoxification proteins [33]. Nrf2 modulation in cancer cells is schematically illustrated in Figure 1.
In human lung cancer, persistent Nrf2 modulation has been shown to result in toxicity attenuation. Conversely, cellular response to drug therapy and ionizing radiations was enhanced upon Nrf2 knockdown [34]. In tumor metastasis, Nrf2 modulation has been shown to result in inhibition of pro-metastatic transcription factor Bach1 degradation, which resulted in promoting lung cancer [35]. Furthermore, Nrf2 modulation was found to be linked to RhoA gene activation, leading to metastasis and proliferation of breast cancer cells [36]. A reduction in antioxidant defense or excessive production of free radicals has been shown to lead to redox imbalance, in turn inducing cardiovascular diseases.
In diabetes mellitus, Nrf2, in addition to its protective response, exerts other significant functions that contribute to the management of diabetes mellitus [37].
Therefore, Nrf2 targeting may pave new ways in the therapy of life-threatening disorders including diabetes.

3. Phytochemical as Nrf2 Modulators

Among the phytochemicals, polyphenols are the most extensively studied and diversified group of phytochemicals [38]. Flavonoids and phenolic acids are classified as two major classes of polyphenols. Hydroxycinammic and hydroxybenzoic acid are classified as main classes of phenolic acid. Further, flavonoids are classified as flavonols, flavones, flavanol, flavonones, and isoflavone [39,40]. Various molecules and signaling pathways implicated in cell death, proliferation, and differentiation are targeted by polyphenols [41,42,43]. The relationship between phytochemicals and Nrf2 signaling is shown in Figure 2. In the prophylaxis of cancer, the cytoprotective role of Nrf2 was elaborated based on expression of various genes through activation of Nrf2 signaling. Nrf2 induction by phytochemicals has been investigated recently, showing Nrf2 activation [44].
Tea components have shown efficacy in in vitro and in vivo disease models, including cancer [45,46,47]. The derivatives of betalamic acid, xanthohumol, betalains, and tannins have been explored to a lesser degree; nonetheless, they do exhibit efficacy in modulating Nrf2 [48]. Tannic acid has been shown in vitro to increase the expression of phase II enzymes downstream of Nrf2 [49]. Betanin was explored in hepatoma-derived HepG2 cell lines in hepatic cancer, showing anticarcinogenic and hepatoprotective effects by Nrf2 activation [50], consistent with findings with xanthohumol. In addition, xanthohumol via activation of Nrf2 signaling has shown anti-inflammatory efficacy in microglial BV2 cells [51]. Induction of the AMPK/GSK3β-Nrf2 pathway was shown to afford protection in acute lung injury [52]. Urosolic acid was investigated in epidermal JB6P+ cells of a mouse skin cancer model, showing Nrf2 activation and increased antioxidant efficacy [53].
Other phytochemicals, such as apigenin and luteolin, were evaluated in NSCLC/A549 cells of non-small cell lung cancer and Bel-740ADM cells of hepatocellular carcinoma. Both reduced protein and Nrf2-mRNA levels, resulting in anti-cancer activity [54,55]. Nrf2 inhibitors were also investigated for cancer treatment. Quassinoid brusatol, a potent inhibitor of Nrf2 that acts via stimulation of Nrf2 poly-ubiquitination, led to reduction in Nrf2 protein levels [28]. Studies in human xenograft myeloid leukemia models showed that quercetin induced apoptosis via reduction in nuclear translocation of Nrf2 and induction of proteasomal degradation via Nrf2 [56]. Details on selected phytochemicals modulating Nrf2 are shown in Table 1.
Combination delivery of phytochemicals has also shown significant effect through modulation of Nrf2. Resveratrol, xanthohumol, and phenethyl isothiocyanate when co-delivered in PANC-1 cells of pancreatic cancer have shown increased DNA and Nrf2 binding along with increased Nrf2 expression [69].

4. Pharmacological Actions of Kaempferol

Kaempferol belongs to a class of natural flavonol. Kaempferol (Figure 3) has been shown to reduce the risk of chronic diseases, such as cancer, diabetes, obesity, and liver injury [70,71]. Kaempferol has also been used for its anti-inflammatory potential in various chronic and acute inflammatory conditions, such as acute lung injury, disc degeneration, and colitis [15]. In acute lung injury, kaempferol, has shown efficacy in mitigating pulmonary inflammatory responses and suppress MAPKs and NF-kB signaling pathway [72]. In disc degeneration, kaempferol has been shown to enhance the viability of bone marrow-derived mesenchymal stem cells through increased cell proliferation and LPS (lipopolysaccharides)-induced cell apoptosis [73]. In colitis, kaempferol has been shown to suppress inflammatory activity [74].
In the hippocampus, kaempferols attenuate hippocampus apoptosis and memory defects induced by Cd-Cl2, targeting Akt/mTOR signaling pathway [75]. It also elevates the level of butyrate receptors, tight junction proteins, and transporters in intestinal mucosa, leading to prophylactic treatment of liver injury induced by alcohol [76]. Kaempferol has been also used for prostate cancer treatment. It promotes apoptosis and inhibits cell proliferation [77]. By regulating vasohibin-1 and ERK signaling, kaempferol prevents high glucose-induced injury in retinal ganglion cells [78]. In addition, kaempferol has a bone-sparing effect via mTOR/PI3K/Akt signaling pathway [79,80]. Co-delivery of kaempferol, quercetin, and pterostilbene has been shown to activate Nrf2 signaling pathways synergistically in hepatic cells, resulted and attenuate ROS generation. Such combination leads to increased binding of Nrf2 to ARE [81].
Promoting mitochondrial function by reduction of oxidative stress is a suitable approach for coping with oxidative stress. D-ribose accumulation in mesangial cells leads to the production of ROS and induces advanced glycation end-products, which, in turn, leads to apoptosis. Kaempferol via autophagy repair mechanism has shown to reverse such effects [82]. In acute lung injury, kaempferol has been shown to reduce damage by altering the ubiquitination of TNF receptor-associated factor-6 (TRAF6). In addition, kaempferol via miR-181a upregulation and MAPK/ERK inactivation leads to proliferation suppression of human gastric cancer cells [83].
The neuroprotective effect of kaempferol was evaluated in striatal injury models. Kaempferol provided a neuroprotective effect by maintaining the integrity of the blood-brain barrier, abating neuroinflammation, and downregulation of the toll-like receptor 4 (TLR4) signaling pathway [84]. Such effects were attributed to protein kinase B/β-catenin cascade (AKT) upregulation and was explored in a mouse model [85], showing reduction in DNA fragmentation and increased cell proliferation targeting PI3K/AKT pathway [86]. Kaempferol has also been shown to inhibit uterine fibroid cells [87]. The inflammation and oxidative stress induced by AGE-RAGE/MAPK was significantly reduced by kaempferol in diabetic rats [88]. Finally, kaempferol inhibits the NF-κB pathway activation in osteoarthritis chondrocytes decreasing the level of interleukin-1β-stimulated pro-inflammatory mediators [89].

5. Kaemperol Affects Nrf2 Activation in Different Pathological Conditions/Diseases

5.1. Cancer

In order to reduce oxidative stress in cancers, many potential signaling and molecular pathways are involved, and kaempferol has been considered an effective agent targeting Nrf2 signaling pathway [90]. In combating cancer, kaempferol regulates transcriptional pathways and restores redox homeostasis, as shown in Figure 4.
The regulation of the Nrf2 signaling pathway affects various factors and other signaling, which, in turn, plays a significant role in cancer [91]. Nrf2 is involved in drug resistance during lung cancer therapy and cancer cell survival [92]. Kaempferol was evaluated for its anti-Nrf2 inhibition potential. Lung cancer A549 and NCIH460 cell lines were used, followed by Nrf2 reporter assay, showing reduction in protein and Nrf2 mRNA levels along with down regulation of Nrf2 target gene transcription. Kaempferol resulted in no change in NFκBp65 and phospho NFκBp65 levels. In addition, kaempferol-induced Nrf2 inhibition resulted in accumulation of reactive oxygen species, sensitizing non-small cell lung cancer cells to apoptosis [93]. Kaempferol-loaded nanoparticles were fabricated and evaluated in a hepatocellular carcinoma model, offering a suitable delivery system for kaempferol during hepatic cancer and Nrf2 signal modulation [94].
Modified Xiaoyao powder—Chinese traditional medicine has been used in the treatment and prevention of breast cancer. Nrf2 was investigated in MCF7 breast cancer cell line followed by determination of chemo preventive action Nrf2 knockdown and Nrf2 wild-type MCF-10A cells, showing upregulation of Nrf2 expression with reduction in oxidative stress. The active compounds were kaempferol and quercetin [95].

5.2. Cardiovascular Diseases

Inflammatory responses and oxidative stress are mediators of vascular pathology [96,97]. Kaempferol has been evaluated for its protective action in vascular endothelium in a mouse model. Kaempferol administration in cardiovascular injury significantly increased the level of Nrf2 along with attenuation of antioxidant levels [98]. The proposed mechanism of kaempferol on atherosclerosis is illustrated in Figure 5.
Kaempferol led to reduction in heart Nrf2 level with elevation in the level of Keap-1 mRNA [99]. Several drugs and targets were investigated for the management and treatment of cardiac hypertrophy [100,101,102]. In this context, kaempferol’s effect on collagen accumulation induced by angiotensin II was investigated in C57BL/6 mice, showing that cardiac dysfunction and fibrosis induced by angiotensin II was significantly reduced by kaempferol administration. In addition, the angiotensin II-related oxidative stress and inflammation was prevented by kaempferol through regulation of AMPK/NRF2 signaling pathway [103]. In addition, kaempferol normalized lipid level and blood vessel morphology with suppression of apoptosis and inflammation, secondary to activation of Nrf2 and estrogen receptor coupled with G-proteins [104].

5.3. Diabetes Complications

Nrf2 activators are effective in mitigating diabetic complications. Nrf2 play a significant role in insulin sensitivity and beta cell function [105]. Kaempferol has been shown to be effective against diabetic nephropathy secondary to its antioxidant potential [106]. Nrf2 levels have been shown to increase in response to kaempferol and to improve heart function [107]. ARE driven genes transcription plays an important role in the regulation of oxidative stress induced by transient hyperglycemia in normal beta cells [105]. Kaempferol modulates of Nrf2 diabetic complications and delays its progression [108].

5.4. Neurological Disorders

Kaempferol has been evaluated for its therapeutic potential in neuroinflammation. Results of the study revealed that Nrf2 binding in microglia to DNA was enhanced by kaempferol, inhibiting neuroinflammation [109]. The neuroprotective effect of kaempferol via Nrf2 regulation was evaluated in a rat model upon chlorpyrifos treatment, a food contaminant and an agricultural pesticide. Results showed a significant protection through kaempferol against neuronal damage, mediated by GSK3β and inhibition of Nrf2 induction [110]. Kaempferol also significantly (p ≤ 0.001) increased the total antioxidant capacity and improved the memory via elevation of Nrf2 [111].

6. Kaempferol Modification during Digestion and Colonic Fermentation

Kaempferol is a poorly absorbed flavonoid. It is also modified during digestion and colonic fermentation. In a recent study, kaempferol-3-glucoside and rutinoside were identified by HPLC. The contents posed modifications at gastric level as compared to intestinal and oral digestion. The rutinosides formed during transport from mouth to intestine showed half reduction in its value, but was stable in the gastrointestinal tract [112]. Under oral and gastrointestinal conditions, the kaempferol glycosides were also found unstable. The conversion of kaempferol glycosides into aglycone kaempferol was also reported following hydrolysis by glycosidases during oral and intestinal digestion. The resultant contents then bind to starch in the digestive tract [113]. Furthermore, another study showed kaempferol glycosides degradation and absorption during gastrointestinal digestion [114]. The Nrf2 interactome is functionally linked to cytoprotection in low-grade stress, chronic inflammation, metabolic alterations, mechanical stress, and ROS formation [115]. Analysis of these molecular profiles suggests alterations of Nrf2 expression and activity as a common mechanism in a subnetwork of diseases referred to as the Nrf2 diseasome. In addition to the role of Nrf2 in various diseases, acute and regular exercise can also induce a state of “low stress” through the production of ROS and stimulation of other mechanoreceptors that activates Nrf2 to modulate endogenous antioxidant systems.

7. Conclusions and Future Directions

Kaempferol is a natural flavonoid that offers therapeutic potential. The main sources of kaempferol include tea, kale, spinach, grapes, and gingko biloba leaves. It exhibits the potential of reducing the risk of certain chronic diseases such as cancer, diabetes, obesity, and liver injury. In addition, it has been used in many acute and chronic inflammatory conditions.
The clinical applications of kaempferol are hindered by its low bioavailability. Thus, to improve the clinical efficacy, there is need for structural modifications and novel formulation development. Specific site modification via glycosylation or methylation might be effective because these compounds endogenously occur in plants. Furthermore, the low bioavailability of kaempferol has limited its use in cancer therapy, therefore combination delivery of kaempferol with other anti-cancer drugs. The delivery of kaempferol as nanotechnology scaffolds also offers a versatile choice for cancer and other pathologic treatments.
In modulating Nrf2, several phytochemicals have shown synergistic effects after co-delivery. Kaempferol in the same may be used with other phytochemicals, resembling the combination of phytochemicals in food matrix.

Author Contributions

Y.H., K.F.A. and A.H.K. written the different chapters of the article as an initial draft. M.A., L.S. and H.K. revised, supervised and approved the final draft. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tu, W.; Wang, H.; Li, S.; Liu, Q.; Sha, H. The anti-inflammatory and anti-oxidant mechanisms of the Keap1/Nrf2/ARE signaling pathway in chronic diseases. Aging Dis. 2019, 10, 637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Schmidlin, C.J.; Shakya, A.; Dodson, M.; Chapman, E.; Zhang, D.D. The intricacies of NRF2 regulation in cancer. In Seminars in Cancer Biology; Elsevier: Amsterdam, The Netherlands, 2021; pp. 110–119. [Google Scholar]
  3. Panieri, E.; Buha, A.; Telkoparan-Akillilar, P.; Cevik, D.; Kouretas, D.; Veskoukis, A.; Skaperda, Z.; Tsatsakis, A.; Wallace, D.; Suzen, S. Potential applications of NRF2 modulators in cancer therapy. Antioxidants 2020, 9, 193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Noman, A.S.M.; Parag, R.R.; Rashid, M.I.; Islam, S.; Rahman, M.Z.; Chowdhury, A.A.; Sultana, A.; Jerin, C.; Siddiqua, A.; Rahman, L. Chemotherapeutic resistance of head and neck squamous cell carcinoma is mediated by EpCAM induction driven by IL-6/p62 associated Nrf2-antioxidant pathway activation. Cell Death Dis. 2020, 11, 1–15. [Google Scholar] [CrossRef] [PubMed]
  5. Cortese-Krott, M.M.; Koning, A.; Kuhnle, G.G.; Nagy, P.; Bianco, C.L.; Pasch, A.; Wink, D.A.; Fukuto, J.M.; Jackson, A.A.; van Goor, H. The reactive species interactome: Evolutionary emergence, biological significance, and opportunities for redox metabolomics and personalized medicine. Antioxid. Redox Signal. 2017, 27, 684–712. [Google Scholar] [CrossRef] [Green Version]
  6. Satta, S.; Mahmoud, A.M.; Wilkinson, F.L.; Yvonne Alexander, M.; White, S.J. The role of Nrf2 in cardiovascular function and disease. Oxidative Med. Cell. Longev. 2017, 2017, 9237263. [Google Scholar] [CrossRef]
  7. Negi, G.; Kumar, A.; Joshi, R.P.; Sharma, S.S. Oxidative stress and Nrf2 in the pathophysiology of diabetic neuropathy: Old perspective with a new angle. Biochem. Biophys. Res. Commun. 2011, 408, 1–5. [Google Scholar] [CrossRef]
  8. Kim, M.-J.; Jeon, J.-H. Recent Advances in Understanding Nrf2 Agonism and Its Potential Clinical Application to Metabolic and Inflammatory Diseases. Int. J. Mol. Sci. 2022, 23, 2846. [Google Scholar] [CrossRef]
  9. Abdelkader, N.F.; Ibrahim, S.M.; Moustafa, P.E.; Elbaset, M.A. Inosine mitigated diabetic peripheral neuropathy via modulating GLO1/AGEs/RAGE/NF-κB/Nrf2 and TGF-β/PKC/TRPV1 signaling pathways. Biomed. Pharmacother. 2022, 145, 112395. [Google Scholar] [CrossRef]
  10. Thiruvengadam, M.; Venkidasamy, B.; Subramanian, U.; Samynathan, R.; Ali Shariati, M.; Rebezov, M.; Girish, S.; Thangavel, S.; Dhanapal, A.R.; Fedoseeva, N. Bioactive Compounds in Oxidative Stress-Mediated Diseases: Targeting the NRF2/ARE Signaling Pathway and Epigenetic Regulation. Antioxidants 2021, 10, 1859. [Google Scholar] [CrossRef]
  11. Robledinos-Antón, N.; Fernández-Ginés, R.; Manda, G.; Cuadrado, A. Activators and inhibitors of NRF2: A review of their potential for clinical development. Oxidative Med. Cell. Longev. 2019, 2019, 9372182. [Google Scholar] [CrossRef]
  12. Wang, P.; Long, F.; Lin, H.; Wang, S.; Wang, T. Dietary Phytochemicals Targeting Nrf2 to Enhance the Radiosensitivity of Cancer. Oxidative Med. Cell. Longev. 2022, 2022, 7848811. [Google Scholar] [CrossRef] [PubMed]
  13. Magesh, S.; Chen, Y.; Hu, L. Small molecule modulators of K eap1-N rf2-ARE pathway as potential preventive and therapeutic agents. Med. Res. Rev. 2012, 32, 687–726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. He, F.; Antonucci, L.; Karin, M. NRF2 as a regulator of cell metabolism and inflammation in cancer. Carcinogenesis 2020, 41, 405–416. [Google Scholar] [CrossRef] [PubMed]
  15. Ren, J.; Lu, Y.; Qian, Y.; Chen, B.; Wu, T.; Ji, G. Recent progress regarding kaempferol for the treatment of various diseases. Exp. Ther. Med. 2019, 18, 2759–2776. [Google Scholar] [CrossRef] [Green Version]
  16. Al-Nour, M.Y.; Ibrahim, M.M.; Elsaman, T. Ellagic acid, Kaempferol, and Quercetin from Acacia nilotica: Promising combined drug with multiple mechanisms of action. Curr. Pharmacol. Rep. 2019, 5, 255–280. [Google Scholar] [CrossRef] [Green Version]
  17. Yang, L.; Li, H.; Yang, M.; Zhang, W.; Li, M.; Xu, Y.; Li, J.; Kang, J.; Zhang, J.; Guo, S. Exploration in the mechanism of kaempferol for the treatment of gastric cancer based on network pharmacology. BioMed Res. Int. 2020, 2020, 5891016. [Google Scholar] [CrossRef] [PubMed]
  18. Xiao, X.; Hu, Q.; Deng, X.; Shi, K.; Zhang, W.; Jiang, Y.; Ma, X.; Zeng, J.; Wang, X. Old wine in new bottles: Kaempferol is a promising agent for treating the trilogy of liver diseases. Pharmacol. Res. 2022, 175, 106005. [Google Scholar] [CrossRef]
  19. Jung, B.-J.; Yoo, H.-S.; Shin, S.; Park, Y.-J.; Jeon, S.-M. Dysregulation of NRF2 in cancer: From molecular mechanisms to therapeutic opportunities. Biomol. Ther. 2018, 26, 57. [Google Scholar] [CrossRef] [Green Version]
  20. Saha, S.; Buttari, B.; Panieri, E.; Profumo, E.; Saso, L. An overview of Nrf2 signaling pathway and its role in inflammation. Molecules 2020, 25, 5474. [Google Scholar] [CrossRef]
  21. Emanuele, S.; Celesia, A.; D’Anneo, A.; Lauricella, M.; Carlisi, D.; De Blasio, A.; Giuliano, M. The Good and Bad of Nrf2: An Update in Cancer and New Perspectives in COVID-19. Int. J. Mol. Sci. 2021, 22, 7963. [Google Scholar] [CrossRef]
  22. Silva-Islas, C.A.; Maldonado, P.D. Canonical and non-canonical mechanisms of Nrf2 activation. Pharmacol. Res. 2018, 134, 92–99. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, T.; Lv, Y.-F.; Zhao, J.-L.; You, Q.-D.; Jiang, Z.-Y. Regulation of Nrf2 by phosphorylation: Consequences for biological function and therapeutic implications. Free Radic. Biol. Med. 2021, 168, 129–141. [Google Scholar] [CrossRef] [PubMed]
  24. Krajka-Kuźniak, V.; Paluszczak, J.; Baer-Dubowska, W. The Nrf2-ARE signaling pathway: An update on its regulation and possible role in cancer prevention and treatment. Pharmacol. Rep. 2017, 69, 393–402. [Google Scholar] [CrossRef] [PubMed]
  25. Shah, S.Z.A.; Zhao, D.; Hussain, T.; Sabir, N.; Mangi, M.H.; Yang, L. p62-Keap1-NRF2-ARE pathway: A contentious player for selective targeting of autophagy, oxidative stress and mitochondrial dysfunction in prion diseases. Front. Mol. Neurosci. 2018, 11, 310. [Google Scholar] [CrossRef] [Green Version]
  26. Sun, X.; Ou, Z.; Chen, R.; Niu, X.; Chen, D.; Kang, R.; Tang, D. Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology 2016, 63, 173–184. [Google Scholar] [CrossRef]
  27. Raghunath, A.; Sundarraj, K.; Arfuso, F.; Sethi, G.; Perumal, E. Dysregulation of Nrf2 in hepatocellular carcinoma: Role in cancer progression and chemoresistance. Cancers 2018, 10, 481. [Google Scholar] [CrossRef] [Green Version]
  28. Panieri, E.; Saso, L. Potential applications of NRF2 inhibitors in cancer therapy. Oxidative Med. Cell. Longev. 2019, 2019, 8592348. [Google Scholar] [CrossRef] [Green Version]
  29. Song, M.-Y.; Lee, D.-Y.; Chun, K.-S.; Kim, E.-H. The Role of NRF2/KEAP1 signaling pathway in cancer metabolism. Int. J. Mol. Sci. 2021, 22, 4376. [Google Scholar] [CrossRef]
  30. Truong, V.L.; Jun, M.; Jeong, W.S. Role of resveratrol in regulation of cellular defense systems against oxidative stress. Biofactors 2018, 44, 36–49. [Google Scholar] [CrossRef]
  31. Chartoumpekis, D.V.; Yagishita, Y.; Fazzari, M.; Palliyaguru, D.L.; Rao, U.N.; Zaravinos, A.; Khoo, N.K.; Schopfer, F.J.; Weiss, K.R.; Michalopoulos, G.K. Nrf2 prevents Notch-induced insulin resistance and tumorigenesis in mice. JCI Insight 2018, 3, e97735. [Google Scholar] [CrossRef]
  32. Shelton, P.; Jaiswal, A.K. The transcription factor NF-E2-related factor 2 (Nrf2): A protooncogene? FASEB J. 2013, 27, 414–423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Li, W.; Yu, S.; Liu, T.; Kim, J.-H.; Blank, V.; Li, H.; Kong, A.-N.T. Heterodimerization with small Maf proteins enhances nuclear retention of Nrf2 via masking the NESzip motif. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2008, 1783, 1847–1856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Yoshino, H.; Murakami, K.; Nawamaki, M.; Kashiwakura, I. Effects of Nrf2 knockdown on the properties of irradiated cell conditioned medium from A549 human lung cancer cells. Biomed. Rep. 2018, 8, 461–465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Lignitto, L.; LeBoeuf, S.E.; Homer, H.; Jiang, S.; Askenazi, M.; Karakousi, T.R.; Pass, H.I.; Bhutkar, A.J.; Tsirigos, A.; Ueberheide, B. Nrf2 activation promotes lung cancer metastasis by inhibiting the degradation of Bach1. Cell 2019, 178, 316–329.e318. [Google Scholar] [CrossRef] [PubMed]
  36. Zhang, C.; Wang, H.-J.; Bao, Q.-C.; Wang, L.; Guo, T.-K.; Chen, W.-L.; Xu, L.-L.; Zhou, H.-S.; Bian, J.-L.; Yang, Y.-R. NRF2 promotes breast cancer cell proliferation and metastasis by increasing RhoA/ROCK pathway signal transduction. Oncotarget 2016, 7, 73593. [Google Scholar] [CrossRef] [Green Version]
  37. Uruno, A.; Yagishita, Y.; Yamamoto, M. The Keap1–Nrf2 system and diabetes mellitus. Arch. Biochem. Biophys. 2015, 566, 76–84. [Google Scholar] [CrossRef] [Green Version]
  38. Mitra, T.; Bhattacharya, R. Phytochemicals modulate cancer aggressiveness: A review depicting the anticancer efficacy of dietary polyphenols and their combinations. J. Cell. Physiol. 2020, 235, 7696–7708. [Google Scholar] [CrossRef]
  39. Cutrim, C.S.; Cortez, M.A.S. A review on polyphenols: Classification, beneficial effects and their application in dairy products. Int. J. Dairy Technol. 2018, 71, 564–578. [Google Scholar] [CrossRef]
  40. Singla, R.K.; Dubey, A.K.; Garg, A.; Sharma, R.K.; Fiorino, M.; Ameen, S.M.; Haddad, M.A.; Al-Hiary, M. Natural Polyphenols: Chemical Classification, Definition of Classes, Subcategories, and Structures; Oxford University Press: Oxford, UK, 2019; pp. 1397–1400. [Google Scholar]
  41. Mirza-Aghazadeh-Attari, M.; Ekrami, E.M.; Aghdas, S.A.M.; Mihanfar, A.; Hallaj, S.; Yousefi, B.; Safa, A.; Majidinia, M. Targeting PI3K/Akt/mTOR signaling pathway by polyphenols: Implication for cancer therapy. Life Sci. 2020, 255, 117481. [Google Scholar] [CrossRef]
  42. Jantan, I.; Haque, M.A.; Arshad, L.; Harikrishnan, H.; Septama, A.W.; Mohamed-Hussein, Z.-A. Dietary polyphenols suppress chronic inflammation by modulation of multiple inflammation-associated cell signaling pathways. J. Nutr. Biochem. 2021, 93, 108634. [Google Scholar] [CrossRef]
  43. Almatroodi, S.A.; Alsahli, M.A.; Almatroudi, A.; Verma, A.K.; Aloliqi, A.; Allemailem, K.S.; Khan, A.A.; Rahmani, A.H. Potential therapeutic targets of quercetin, a plant flavonol, and its role in the therapy of various types of cancer through the modulation of various cell signaling pathways. Molecules 2021, 26, 1315. [Google Scholar] [CrossRef] [PubMed]
  44. Clifford, T.; Acton, J.P.; Cocksedge, S.P.; Davies, K.A.B.; Bailey, S.J. The effect of dietary phytochemicals on nuclear factor erythroid 2-related factor 2 (Nrf2) activation: A systematic review of human intervention trials. Mol. Biol. Rep. 2021, 48, 1745–1761. [Google Scholar] [CrossRef] [PubMed]
  45. Rudra, A.; Arvind, I.; Mehra, R. Polyphenols: Types, sources and therapeutic applications. Int. J. Home Sci. 2021, 7, 69–75. [Google Scholar] [CrossRef]
  46. 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. 2021, 37, 759–811. [Google Scholar] [CrossRef]
  47. 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]
  48. Krajka-Kuźniak, V.; Baer-Dubowska, W. Modulation of Nrf2 and NF-κB Signaling Pathways by Naturally Occurring Compounds in Relation to Cancer Prevention and Therapy. Are Combinations Better Than Single Compounds? Int. J. Mol. Sci. 2021, 22, 8223. [Google Scholar] [CrossRef]
  49. Baer-Dubowska, W.; Szaefer, H.; Majchrzak-Celińska, A.; Krajka-Kuźniak, V. Tannic acid: Specific form of tannins in cancer chemoprevention and therapy-old and new applications. Curr. Pharmacol. Rep. 2020, 6, 28–37. [Google Scholar] [CrossRef] [Green Version]
  50. Krajka-Kuźniak, V.; Paluszczak, J.; Szaefer, H.; Baer-Dubowska, W. Betanin, a beetroot component, induces nuclear factor erythroid-2-related factor 2-mediated expression of detoxifying/antioxidant enzymes in human liver cell lines. Br. J. Nutr. 2013, 110, 2138–2149. [Google Scholar] [CrossRef] [Green Version]
  51. Lee, I.-S.; Lim, J.; Gal, J.; Kang, J.C.; Kim, H.J.; Kang, B.Y.; Choi, H.J. Anti-inflammatory activity of xanthohumol involves heme oxygenase-1 induction via NRF2-ARE signaling in microglial BV2 cells. Neurochem. Int. 2011, 58, 153–160. [Google Scholar] [CrossRef]
  52. Lv, H.; Liu, Q.; Wen, Z.; Feng, H.; Deng, X.; Ci, X. Xanthohumol ameliorates lipopolysaccharide (LPS)-induced acute lung injury via induction of AMPK/GSK3β-Nrf2 signal axis. Redox Biol. 2017, 12, 311–324. [Google Scholar] [CrossRef]
  53. Kim, H.; Ramirez, C.N.; Su, Z.-Y.; Kong, A.-N.T. Epigenetic modifications of triterpenoid ursolic acid in activating Nrf2 and blocking cellular transformation of mouse epidermal cells. J. Nutr. Biochem. 2016, 33, 54–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Hussein, M.C.; Bektas, N.; Ozturk, Y.; Arslan, R. Antinociception Induced by Moringa Stenopetela (Baker f.) Cufod. Leaves Extract and Possible Mechanisms of Action. Braz. J. Pharm. Sci. 2022, 58, 118578. [Google Scholar] [CrossRef]
  55. Arlt, A.; Sebens, S.; Krebs, S.; Geismann, C.; Grossmann, M.; Kruse, M.; Schreiber, S.; Schäfer, H. Inhibition of the Nrf2 transcription factor by the alkaloid trigonelline renders pancreatic cancer cells more susceptible to apoptosis through decreased proteasomal gene expression and proteasome activity. Oncogene 2013, 32, 4825–4835. [Google Scholar] [CrossRef] [PubMed]
  56. De Prax, M.C.A.; Ferro, K.P.; Santos, I.; Torello, C.O.; Salazar-Terreros, M.; Saad, S.T.O. NRF2 Is Targeted By the Polyphenol Quercetin and Induces Apoptosis, in Part, through up Regulation of Pro Apoptotic Mirs. Blood 2019, 134, 2529. [Google Scholar] [CrossRef]
  57. Paredes-Gonzalez, X.; Fuentes, F.; Jeffery, S.; Saw, C.L.L.; Shu, L.; Su, Z.Y.; Kong, A.N.T. Induction of NRF2-mediated gene expression by dietary phytochemical flavones apigenin and luteolin. Biopharm. Drug Dispos. 2015, 36, 440–451. [Google Scholar] [CrossRef]
  58. Lee, S.; Choi, S.-Y.; Choo, Y.-Y.; Kim, O.; Tran, P.T.; Dao, C.T.; Min, B.-S.; Lee, J.-H. Sappanone A exhibits anti-inflammatory effects via modulation of Nrf2 and NF-κB. Int. Immunopharmacol. 2015, 28, 328–336. [Google Scholar] [CrossRef]
  59. Krajka-Kuźniak, V.; Cykowiak, M.; Szaefer, H.; Kleszcz, R.; Baer-Dubowska, W. Combination of xanthohumol and phenethyl isothiocyanate inhibits NF-κB and activates Nrf2 in pancreatic cancer cells. Toxicol. Vitr. 2020, 65, 104799. [Google Scholar] [CrossRef]
  60. Licznerska, B.; Szaefer, H.; Krajka-Kuźniak, V. R-sulforaphane modulates the expression profile of AhR, ERα, Nrf2, NQO1, and GSTP in human breast cell lines. Mol. Cell. Biochem. 2021, 476, 525–533. [Google Scholar] [CrossRef]
  61. Lu, Y.; Sun, Y.; Zhu, J.; Yu, L.; Jiang, X.; Zhang, J.; Dong, X.; Ma, B.; Zhang, Q. Oridonin exerts anticancer effect on osteosarcoma by activating PPAR-γ and inhibiting Nrf2 pathway. Cell Death Dis. 2018, 9, 1–16. [Google Scholar] [CrossRef] [Green Version]
  62. Zhong, Y.; Zhang, F.; Sun, Z.; Zhou, W.; Li, Z.Y.; You, Q.D.; Guo, Q.L.; Hu, R. Drug resistance associates with activation of Nrf2 in MCF-7/DOX cells, and wogonin reverses it by down-regulating Nrf2-mediated cellular defense response. Mol. Carcinog. 2013, 52, 824–834. [Google Scholar] [CrossRef] [PubMed]
  63. Sahin, K.; Yenice, E.; Bilir, B.; Orhan, C.; Tuzcu, M.; Sahin, N.; Ozercan, I.H.; Kabil, N.; Ozpolat, B.; Kucuk, O. Genistein prevents development of spontaneous ovarian cancer and inhibits tumor growth in hen model. Cancer Prev. Res. 2019, 12, 135–146. [Google Scholar] [CrossRef] [Green Version]
  64. Krajka-Kuźniak, V.; Szaefer, H.; Stefański, T.; Sobiak, S.; Cichocki, M.; Baer-Dubowska, W. The effect of resveratrol and its methylthio-derivatives on the Nrf2-ARE pathway in mouse epidermis and HaCaT keratinocytes. Cell. Mol. Biol. Lett. 2014, 19, 500–516. [Google Scholar] [CrossRef]
  65. Liu, C.-M.; Ma, J.-Q.; Xie, W.-R.; Liu, S.-S.; Feng, Z.-J.; Zheng, G.-H.; Wang, A.-M. Quercetin protects mouse liver against nickel-induced DNA methylation and inflammation associated with the Nrf2/HO-1 and p38/STAT1/NF-κB pathway. Food Chem. Toxicol. 2015, 82, 19–26. [Google Scholar] [CrossRef]
  66. Chen, B.; Zhang, Y.; Wang, Y.; Rao, J.; Jiang, X.; Xu, Z. Curcumin inhibits proliferation of breast cancer cells through Nrf2-mediated down-regulation of Fen1 expression. J. Steroid Biochem. Mol. Biol. 2014, 143, 11–18. [Google Scholar] [CrossRef]
  67. Rehman, M.U.; Rashid, S.; Arafah, A.; Qamar, W.; Alsaffar, R.M.; Ahmad, A.; Almatroudi, N.M.; Alqahtani, S.; Rashid, S.M.; Ahmad, S.B. Piperine regulates Nrf-2/Keap-1 signalling and exhibits anticancer effect in experimental colon carcinogenesis in wistar rats. Biology 2020, 9, 302. [Google Scholar] [CrossRef]
  68. Zhang, N.; Dou, D.; Ran, X.; Kang, T. Neuroprotective effect of arctigenin against neuroinflammation and oxidative stress induced by rotenone. Rsc Adv. 2018, 8, 2280–2292. [Google Scholar] [CrossRef] [Green Version]
  69. Cykowiak, M.; Krajka-Kuźniak, V.; Baer-Dubowska, W. Combinations of phytochemicals more efficiently than single components activate Nrf2 and induce the expression of antioxidant enzymes in pancreatic cancer cells. Nutr. Cancer 2021, 74, 1–16. [Google Scholar] [CrossRef]
  70. Imran, M.; Rauf, A.; Shah, Z.A.; Saeed, F.; Imran, A.; Arshad, M.U.; Ahmad, B.; Bawazeer, S.; Atif, M.; Peters, D.G. Chemo-preventive and therapeutic effect of the dietary flavonoid kaempferol: A comprehensive review. Phytother. Res. 2019, 33, 263–275. [Google Scholar] [CrossRef]
  71. Wong, S.K.; Chin, K.-Y.; Ima-Nirwana, S. The osteoprotective effects of kaempferol: The evidence from in vivo and in vitro studies. Drug Des. Dev. Ther. 2019, 13, 3497. [Google Scholar] [CrossRef] [Green Version]
  72. Chen, X.; Yang, X.; Liu, T.; Guan, M.; Feng, X.; Dong, W.; Chu, X.; Liu, J.; Tian, X.; Ci, X. Kaempferol regulates MAPKs and NF-κB signaling pathways to attenuate LPS-induced acute lung injury in mice. Int. Immunopharmacol. 2012, 14, 209–216. [Google Scholar] [CrossRef]
  73. Zhu, J.; Tang, H.; Zhang, Z.; Zhang, Y.; Qiu, C.; Zhang, L.; Huang, P.; Li, F. Kaempferol slows intervertebral disc degeneration by modifying LPS-induced osteogenesis/adipogenesis imbalance and inflammation response in BMSCs. Int. Immunopharmacol. 2017, 43, 236–242. [Google Scholar] [CrossRef]
  74. Park, M.-Y.; Ji, G.E.; Sung, M.-K. Dietary kaempferol suppresses inflammation of dextran sulfate sodium-induced colitis in mice. Dig. Dis. Sci. 2012, 57, 355–363. [Google Scholar] [CrossRef]
  75. El-Kott, A.F.; Bin-Meferij, M.M.; Eleawa, S.M.; Alshehri, M.M. Kaempferol protects against cadmium chloride-induced memory loss and hippocampal apoptosis by increased intracellular glutathione stores and activation of PTEN/AMPK induced inhibition of Akt/mTOR signaling. Neurochem. Res. 2020, 45, 295–309. [Google Scholar] [CrossRef]
  76. Chen, J.; Xuan, Y.-H.; Luo, M.-X.; Ni, X.-G.; Ling, L.-Q.; Hu, S.-J.; Chen, J.-Q.; Xu, J.-Y.; Jiang, L.-Y.; Si, W.-Z. Kaempferol alleviates acute alcoholic liver injury in mice by regulating intestinal tight junction proteins and butyrate receptors and transporters. Toxicology 2020, 429, 152338. [Google Scholar] [CrossRef]
  77. Da, J.; Xu, M.; Wang, Y.; Li, W.; Lu, M.; Wang, Z. Kaempferol promotes apoptosis while inhibiting cell proliferation via androgen-dependent pathway and suppressing vasculogenic mimicry and invasion in prostate cancer. Anal. Cell. Pathol. 2019, 2019, 1907698. [Google Scholar] [CrossRef] [Green Version]
  78. Zhao, L.; Sun, J.; Shi, S.; Qin, X.; Zhang, K.; Xu, J. Kaempferol protects retinal ganglion ceils from high-glucose-induced injury by regulating vasohibin-1. Neurosci. Lett. 2020, 716, 134633. [Google Scholar] [CrossRef]
  79. Sharma, A.R.; Nam, J.-S. Kaempferol stimulates WNT/β-catenin signaling pathway to induce differentiation of osteoblasts. J. Nutr. Biochem. 2019, 74, 108228. [Google Scholar] [CrossRef]
  80. Lei, X.; Guo, J.; Wang, Y.; Cui, J.; Feng, B.; Su, Y.; Zhao, H.; Yang, W.; Hu, Y. Inhibition of endometrial carcinoma by Kaempferol is interceded through apoptosis induction, G2/M phase cell cycle arrest, suppression of cell invasion and upregulation of m-TOR/PI3K signalling pathway. J. Balk. Union Oncol. 2019, 24, 1555–1561. [Google Scholar]
  81. Saw, C.L.L.; Guo, Y.; Yang, A.Y.; Paredes-Gonzalez, X.; Ramirez, C.; Pung, D.; Kong, A.-N.T. The berry constituents quercetin, kaempferol, and pterostilbene synergistically attenuate reactive oxygen species: Involvement of the Nrf2-ARE signaling pathway. Food Chem. Toxicol. 2014, 72, 303–311. [Google Scholar] [CrossRef]
  82. Zhang, F.; Ma, C. Kaempferol suppresses human gastric cancer SNU-216 cell proliferation, promotes cell autophagy, but has no influence on cell apoptosis. Braz. J. Med. Biol. Res. 2019, 52, e7843. [Google Scholar] [CrossRef] [Green Version]
  83. Qian, J.; Chen, X.; Chen, X.; Sun, C.; Jiang, Y.; Qian, Y.; Zhang, Y.; Khan, Z.; Zhou, J.; Liang, G. Kaempferol reduces K63-linked polyubiquitination to inhibit nuclear factor-κB and inflammatory responses in acute lung injury in mice. Toxicol. Lett. 2019, 306, 53–60. [Google Scholar] [CrossRef]
  84. Yang, Y.-L.; Cheng, X.; Li, W.-H.; Liu, M.; Wang, Y.-H.; Du, G.-H. Kaempferol attenuates LPS-induced striatum injury in mice involving anti-neuroinflammation, maintaining BBB integrity, and down-regulating the HMGB1/TLR4 pathway. Int. J. Mol. Sci. 2019, 20, 491. [Google Scholar] [CrossRef] [Green Version]
  85. Gao, W.; Wang, W.; Peng, Y.; Deng, Z. Antidepressive effects of kaempferol mediated by reduction of oxidative stress, proinflammatory cytokines and up-regulation of AKT/β-catenin cascade. Metab. Brain Dis. 2019, 34, 485–494. [Google Scholar] [CrossRef]
  86. Santos-Buelga, C.; González-Paramás, A.M.; Oludemi, T.; Ayuda-Durán, B.; González-Manzano, S. Plant phenolics as functional food ingredients. Adv. Food Nutr. Res. 2019, 90, 183–257. [Google Scholar]
  87. Li, Y.; Ding, Z.; Wu, C. Mechanistic study of the inhibitory effect of kaempferol on uterine fibroids in vitro. Med. Sci. Monit.: Int. Med. J. Exp. Clin. Res. 2016, 22, 4803. [Google Scholar] [CrossRef] [Green Version]
  88. Suchal, K.; Malik, S.; Khan, S.I.; Malhotra, R.K.; Goyal, S.N.; Bhatia, J.; Ojha, S.; Arya, D.S. Molecular pathways involved in the amelioration of myocardial injury in diabetic rats by kaempferol. Int. J. Mol. Sci. 2017, 18, 1001. [Google Scholar] [CrossRef] [Green Version]
  89. Zhuang, Z.; Ye, G.; Huang, B. Kaempferol alleviates the interleukin-1β-induced inflammation in rat osteoarthritis chondrocytes via suppression of NF-κB. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2017, 23, 3925. [Google Scholar] [CrossRef] [Green Version]
  90. Sharma, N.; Biswas, S.; Al-Dayan, N.; Alhegaili, A.S.; Sarwat, M. Antioxidant role of kaempferol in prevention of hepatocellular carcinoma. Antioxidants 2021, 10, 1419. [Google Scholar] [CrossRef]
  91. Zimta, A.-A.; Cenariu, D.; Irimie, A.; Magdo, L.; Nabavi, S.M.; Atanasov, A.G.; Berindan-Neagoe, I. The role of Nrf2 activity in cancer development and progression. Cancers 2019, 11, 1755. [Google Scholar] [CrossRef] [Green Version]
  92. Okazaki, K.; Anzawa, H.; Katsuoka, F.; Kinoshita, K.; Sekine, H.; Motohashi, H. CEBPB is Required for NRF2-Mediated Drug Resistance in NRF2-Activated Non-Small Cell Lung Cancer Cells. J. Biochem. 2022, 171, 567–578. [Google Scholar] [CrossRef]
  93. Fouzder, C.; Mukhuty, A.; Kundu, R. Kaempferol inhibits Nrf2 signalling pathway via downregulation of Nrf2 mRNA and induces apoptosis in NSCLC cells. Arch. Biochem. Biophys. 2021, 697, 108700. [Google Scholar] [CrossRef]
  94. Kazmi, I.; Al-Abbasi, F.A.; Afzal, M.; Altayb, H.N.; Nadeem, M.S.; Gupta, G. Formulation and Evaluation of Kaempferol Loaded Nanoparticles against Experimentally Induced Hepatocellular Carcinoma: In Vitro and In Vivo Studies. Pharmaceutics 2021, 13, 2086. [Google Scholar] [CrossRef]
  95. Yang, Y.; Wang, Y.; Wang, T.; Jiang, X.; Wang, L. Screening active components of modified Xiaoyao powder as NRF2 agonists. Cell Biochem. Funct. 2017, 35, 518–526. [Google Scholar] [CrossRef]
  96. Yu, Y.; Yan, Y.; Niu, F.; Wang, Y.; Chen, X.; Su, G.; Liu, Y.; Zhao, X.; Qian, L.; Liu, P. Ferroptosis: A cell death connecting oxidative stress, inflammation and cardiovascular diseases. Cell Death Discov. 2021, 7, 1–10. [Google Scholar] [CrossRef]
  97. Matyas, C.; Haskó, G.; Liaudet, L.; Trojnar, E.; Pacher, P. Interplay of cardiovascular mediators, oxidative stress and inflammation in liver disease and its complications. Nat. Rev. Cardiol. 2021, 18, 117–135. [Google Scholar] [CrossRef]
  98. Yao, H.; Sun, J.; Wei, J.; Zhang, X.; Chen, B.; Lin, Y. Kaempferol protects blood vessels from damage induced by oxidative stress and inflammation in association with the Nrf2/HO-1 signaling pathway. Front. Pharmacol. 2020, 11, 1118. [Google Scholar] [CrossRef]
  99. Zhang, L.; Guo, Z.; Wang, Y.; Geng, J.; Han, S. The protective effect of kaempferol on heart via the regulation of Nrf2, NF-κβ, and PI3K/Akt/GSK-3β signaling pathways in isoproterenol-induced heart failure in diabetic rats. Drug Dev. Res. 2019, 80, 294–309. [Google Scholar] [CrossRef]
  100. Raghunathan, S.; Goyal, R.K.; Patel, B.M. Selective inhibition of HDAC2 by magnesium valproate attenuates cardiac hypertrophy. Can. J. Physiol. Pharmacol. 2017, 95, 260–267. [Google Scholar] [CrossRef] [Green Version]
  101. Patel, B.M. Sodium butyrate controls cardiac hypertrophy in experimental models of rats. Cardiovasc. Toxicol. 2018, 18, 1–8. [Google Scholar] [CrossRef]
  102. Sharma, B.; Chaube, U.; Patel, B.M. Beneficial effect of silymarin in pressure overload induced experimental cardiac hypertrophy. Cardiovasc. Toxicol. 2019, 19, 23–35. [Google Scholar] [CrossRef]
  103. Du, Y.; Han, J.; Zhang, H.; Xu, J.; Jiang, L.; Ge, W. Kaempferol prevents against Ang II-induced cardiac remodeling through attenuating Ang II-induced inflammation and oxidative stress. J. Cardiovasc. Pharmacol. 2019, 74, 326. [Google Scholar] [CrossRef] [PubMed]
  104. Feng, Z.; Wang, C.; Yue; Jin; Meng, Q.; Wu, J.; Sun, H. Kaempferol-induced GPER upregulation attenuates atherosclerosis via the PI3K/AKT/Nrf2 pathway. Pharm. Biol. 2021, 59, 1106–1116. [Google Scholar] [CrossRef]
  105. Baumel-Alterzon, S.; Katz, L.S.; Brill, G.; Garcia-Ocaña, A.; Scott, D.K. Nrf2: The master and captain of beta cell fate. Trends Endocrinol. Metab. 2021, 32, 7–19. [Google Scholar] [CrossRef] [PubMed]
  106. Alshehri, A.S. Kaempferol attenuates diabetic nephropathy in streptozotocin-induced diabetic rats by a hypoglycaemic effect and concomitant activation of the Nrf-2/Ho-1/antioxidants axis. Arch. Physiol. Biochem. 2021, 15, 1–14. [Google Scholar] [CrossRef] [PubMed]
  107. Alshehri, A.; El-Kott, A.; Eleawa, S.; El-Gerbed, M.; Khalifa, H.; El-Kenawy, A.; Albadrani, G.; Abdel-Daim, M. Kaempferol protects against streptozotocin-induced diabetic cardiomyopathy in rats by a hypoglycemic effect and upregulating SIRT1. J. Physiol. Pharm. 2021, 72, 339–355. [Google Scholar]
  108. Negi, C.K.; Jena, G. Nrf2, a novel molecular target to reduce type 1 diabetes associated secondary complications: The basic considerations. Eur. J. Pharmacol. 2019, 843, 12–26. [Google Scholar] [CrossRef]
  109. Velagapudi, R.; Jamshaid, F.; Lepiarz, I.; Katola, F.O.; Hemming, K.; Olajide, O.A. The tiliroside derivative, 3-O-[(E)-(2-oxo-4-(p-tolyl) but–3–en–1–yl] kaempferol produced inhibition of neuroinflammation and activation of AMPK and Nrf2/HO-1 pathways in BV-2 microglia. Int. Immunopharmacol. 2019, 77, 105951. [Google Scholar] [CrossRef]
  110. Hussein, R.M.; Mohamed, W.R.; Omar, H.A. A neuroprotective role of kaempferol against chlorpyrifos-induced oxidative stress and memory deficits in rats via GSK3β-Nrf2 signaling pathway. Pestic. Biochem. Physiol. 2018, 152, 29–37. [Google Scholar] [CrossRef]
  111. Jafari, A.; Babaei, P.; Rohampour, K.; Rashtiani, S. The Effect of Kaempferol on Autophagy and Nrf-2 Signaling in a Rat Model of Aβ1-42-induced Alzheimer’s Disease. Casp. J. Neurol. Sci. 2022, 8, 7–16. [Google Scholar] [CrossRef]
  112. Gayoso, L.; Claerbout, A.-S.; Calvo, M.I.; Cavero, R.Y.; Astiasarán, I.; Ansorena, D. Bioaccessibility of rutin, caffeic acid and rosmarinic acid: Influence of the in vitro gastrointestinal digestion models. J. Funct. Foods 2016, 26, 428–438. [Google Scholar] [CrossRef]
  113. Takahama, U.; Hirota, S. Effects of starch on nitrous acid-induced oxidation of kaempferol and inhibition of α-amylase-catalysed digestion of starch by kaempferol under conditions simulating the stomach and the intestine. Food Chem. 2013, 141, 313–319. [Google Scholar] [CrossRef] [PubMed]
  114. Sun, Y.; Guo, F.; Peng, X.; Cheng, K.; Xiao, L.; Zhang, H.; Li, H.; Jiang, L.; Deng, Z. Metabolism of Phenolics of Tetrastigma hemsleyanum Roots under In Vitro Digestion and Colonic Fermentation as Well as Their In Vivo Antioxidant Activity in Rats. Foods 2021, 10, 2123. [Google Scholar] [CrossRef] [PubMed]
  115. Marchev, A.S.; Dimitrova, P.A.; Burns, A.J.; Kostov, R.V.; Dinkova-Kostova, A.T.; Georgiev, M.I. Oxidative stress and chronic inflammation in osteoarthritis: Can NRF2 counteract these partners in crime? Ann. N. Y. Acad. Sci. 2017, 1401, 114–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Nrf2 activation/inhibition in cancer cells. Various molecular mechanisms in cancer cells constitute activation of Nrf2, leading to gene expression associated with progression of tumors. The resultant effect is in the form of activation of metabolic reprogramming, enhanced cell proliferation, drug resistance, and adaptation to stress. Nrf2 antioxidant activation also induces an imbalance in the carbon metabolism in cancer. Keap-1; kelch-like ECH-associated proteins, Nrf2; nuclear factor erythroid 2-related factor 2, MAF; musculoaponeurotic fibrosarcoma protein.
Figure 1. Nrf2 activation/inhibition in cancer cells. Various molecular mechanisms in cancer cells constitute activation of Nrf2, leading to gene expression associated with progression of tumors. The resultant effect is in the form of activation of metabolic reprogramming, enhanced cell proliferation, drug resistance, and adaptation to stress. Nrf2 antioxidant activation also induces an imbalance in the carbon metabolism in cancer. Keap-1; kelch-like ECH-associated proteins, Nrf2; nuclear factor erythroid 2-related factor 2, MAF; musculoaponeurotic fibrosarcoma protein.
Molecules 27 04145 g001
Figure 2. Nrf2 activation by phytochemicals leading to various events. MAF (musculoaponeurotic fibrosarcoma protein) activates antioxidant response genes by interacting with Nrf2 through ARE signaling, indicating that in response to electrophilic and oxidative stresses. In addition, in cancer cells, the imbalance in Nrf2/ARE signaling leads to drug resistance, poor prognosis, and cell proliferation. So, the consecutive Nrf2 activation might cope with the consequences in a positive manner.
Figure 2. Nrf2 activation by phytochemicals leading to various events. MAF (musculoaponeurotic fibrosarcoma protein) activates antioxidant response genes by interacting with Nrf2 through ARE signaling, indicating that in response to electrophilic and oxidative stresses. In addition, in cancer cells, the imbalance in Nrf2/ARE signaling leads to drug resistance, poor prognosis, and cell proliferation. So, the consecutive Nrf2 activation might cope with the consequences in a positive manner.
Molecules 27 04145 g002
Figure 3. Chemical structure of kaempferol.
Figure 3. Chemical structure of kaempferol.
Molecules 27 04145 g003
Figure 4. Kaempferol antioxidant potential via modulation of Nrf2. Reactive oxygen species metabolism is inhibited by kaempferol via acting on Nrf2 complex. After disintegration of the Nrf2 complex, it is translocated to the nucleus where it binds to Maf; as a result, the expression of target genes takes place, eventually causing the inhibition of inflammation, oxidation, and induction of autophagy.
Figure 4. Kaempferol antioxidant potential via modulation of Nrf2. Reactive oxygen species metabolism is inhibited by kaempferol via acting on Nrf2 complex. After disintegration of the Nrf2 complex, it is translocated to the nucleus where it binds to Maf; as a result, the expression of target genes takes place, eventually causing the inhibition of inflammation, oxidation, and induction of autophagy.
Molecules 27 04145 g004
Figure 5. Kaempferol proposed mechanism on atherosclerosis. Kaempferol via inhibition of increased inflammation and reactive oxygen species through modulation of endothelial dysfunction that results in inhibition of atherosclerosis.
Figure 5. Kaempferol proposed mechanism on atherosclerosis. Kaempferol via inhibition of increased inflammation and reactive oxygen species through modulation of endothelial dysfunction that results in inhibition of atherosclerosis.
Molecules 27 04145 g005
Table 1. Phytochemicals (non-nutrient) modulating Nrf2.
Table 1. Phytochemicals (non-nutrient) modulating Nrf2.
PhytochemicalsModelConcentrationEffects on Nrf2References
ApigeninHepG2 cells6.2 µMDecreased proteins and mRNA levels of Nrf2[57]
SappanoneRAW264.7 cells30 µMNrf2 increased nuclear translocation[58]
XanthohumolPANC-1 cells5–10 µMNrf2 and DNA binding along with Nrf2
increased expression
[59]
SulforaphaneMCF-7 cells5, 10 and 20 µMIncreased Nrf2
expression
[60]
OridininMG-63 cells, c nude mice30 mg/kgNrf2 decreased nuclear translocation[61]
WogoninMCF-7 cells60 µMDecreased Nrf2
expression
[62]
GenisteinLaying Hen model52.48 mg/henIncreased Nrf2
expression
[63]
ResveratrolHaCaT cells60 µMIncreased nuclear level of Nrf2[64]
QuercetinICR mice (male)40–80 mg/kgMimicked nuclear translocation of Nrf2[65]
Curcumin MCF-7 cells20–40 µMNrf2 increased
expression
[66]
PiperineWistar rats (male)30–60 mg/kgIncreased Nrf2
expression
[67]
ArctigeninSD rats (male)20 mg/kgIncreased SOD
expression
[68]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hussain, Y.; Khan, H.; Alsharif, K.F.; Hayat Khan, A.; Aschner, M.; Saso, L. The Therapeutic Potential of Kaemferol and Other Naturally Occurring Polyphenols Might Be Modulated by Nrf2-ARE Signaling Pathway: Current Status and Future Direction. Molecules 2022, 27, 4145. https://doi.org/10.3390/molecules27134145

AMA Style

Hussain Y, Khan H, Alsharif KF, Hayat Khan A, Aschner M, Saso L. The Therapeutic Potential of Kaemferol and Other Naturally Occurring Polyphenols Might Be Modulated by Nrf2-ARE Signaling Pathway: Current Status and Future Direction. Molecules. 2022; 27(13):4145. https://doi.org/10.3390/molecules27134145

Chicago/Turabian Style

Hussain, Yaseen, Haroon Khan, Khalaf F. Alsharif, Amjad Hayat Khan, Michael Aschner, and Luciano Saso. 2022. "The Therapeutic Potential of Kaemferol and Other Naturally Occurring Polyphenols Might Be Modulated by Nrf2-ARE Signaling Pathway: Current Status and Future Direction" Molecules 27, no. 13: 4145. https://doi.org/10.3390/molecules27134145

Article Metrics

Back to TopTop