You are on page 1of 7

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/293544034

Quercetin

Chapter · January 2016


DOI: 10.1016/B978-0-12-802147-7.00032-2

CITATIONS READS
23 1,139

6 authors, including:

Adhithiya Charli Huajun Jin


Charles River Laboratories International, Inc Iowa State University
27 PUBLICATIONS   589 CITATIONS    109 PUBLICATIONS   3,037 CITATIONS   

SEE PROFILE SEE PROFILE

Vellareddy Anantharam Arthi Kanthasamy


Iowa State University Iowa State University
167 PUBLICATIONS   9,266 CITATIONS    159 PUBLICATIONS   8,436 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Alterations in mitochondrial dynamics induced by tebufenpyrad and pyridaben in a dopaminergic neuronal cell culture model View project

p73 gene in Dopaminergic Neurons is Highly Susceptible to Manganese Neurotoxicity View project

All content following this page was uploaded by Adhithiya Charli on 08 January 2019.

The user has requested enhancement of the downloaded file.


C H A P T E R

32
Quercetin
Muhammet Ay, Adhithiya Charli, Huajun Jin, Vellareddy
Anantharam, Arthi Kanthasamy and Anumantha G. Kanthasamy

INTRODUCTION of oncogenes, including Mcl-1, Bcl-2, Ras, MEK, and


PI3K, or the upregulation of tumor suppressor genes,
Dietary flavonoids were first discovered by Nobel including p53 and p21 (Iwao and Tsukamoto, 1999;
Prize laureate Albert Szent Gyorgyi in 1936, and they are Sharma et  al., 2005; Lim et  al., 2007; Spagnuolo et  al.,
widely recognized for their potential beneficial effects 2011). In MDA-MB-231 human breast cancer cells, quer-
on human health. Quercetin is the major flavonoid in cetin was reported to suppress cell proliferation through
our daily diet (Table 32.1) (see further details at http:// the upregulation of Connexin 43 (Cx43) (Conklin et al.,
www.ars.usda.gov/SP2UserFiles/Place/80400525/ 2007), whose expression correlates inversely with the
Data/Flav/Flav_R03-1.pdf), and its estimated daily aggressiveness of the tumor (Laird et al., 1999). Similarly,
intake is between 5 and 40 mg (Hertog et al., 1995). After another study demonstrated that quercetin inhibited the
absorption, quercetin is mainly metabolized in the intes- growth of MCF-7 breast cancer cells and also down-
tine and liver. The plasma concentration of quercetin regulated the expression of survivin, which is highly
is normally in the nanomolar range, but it can reach expressed in most cancers (Deng et al., 2013).
the micromolar range after consumption of quercetin- Quercetin has been reported to induce apoptosis in a
rich foods (Russo et  al., 2012). Table 32.1 summarizes human prostate cell line (LNCaP) by inducing the dis-
foods with high quercetin levels. Quercetin contains sociation of Bax from Bcl-xL, activating caspases, and
three rings and five hydroxyl groups (Figure 32.1). inhibiting phosphorylation of Akt (Kim and Lee, 2007;
Quercetin mainly occurs as glycosides, ethers, and, to a Lee et al., 2008). Urokinase-type plasminogen activator
lesser extent, sulfates (Williams and Grayer, 2004), and (uPA) has been associated with cancer invasion, includ-
glycosylation of quercetin increases its hydrophilicity, ing prostate cancer. Quercetin significantly decreased the
stability, and bioavailability (He et al., 2006). So far, quer- expressions of uPA, MMP-2, MMP-9, EGF, β-catenin, and
cetin is the most extensively studied flavonoid that has NF-κB, and thus suppressed the proliferation, migration,
been shown to exhibit antioxidant, antiviral, antibacte- and invasion of prostate cancer cells (Vijayababu et al.,
rial, anti-inflammatory, and anticarcinogenic properties 2005; Senthilkumar et  al., 2011). Recently, the chemo-
(Duthie and Dobson, 1999; Ramos et  al., 2006; Boots preventive effects of quercetin have been studied in an
et al., 2008; Zandi et al., 2011). in vivo model of prostate cancer (Sharmila et al., 2014).
The results showed that quercetin significantly reversed
the reduction in superoxide dismutase (SOD), catalase
QUERCETIN AND CANCER (CAT), and GPx activities and the increase in Akt phos-
phorylation in prostate cancer-induced rats.
Quercetin has been considered a potential anticancer Activation of β-catenin/Tcf signaling plays a cen-
agent because it can inhibit cell proliferation and angio- tral role in colorectal carcinogenesis (Park et  al., 2005).
genesis and induce apoptosis and cellular senescence Interestingly, quercetin has been shown to inhibit cell
(Russo et  al., 1999; Lamson and Brignall, 2000; Zamin proliferation through suppression of β-catenin/Tcf sig-
et  al., 2009). Quercetin can induce cell death and cell naling and survivin expression in SW480 colon can-
cycle arrest in cancer cells through the downregulation cer cells (Shan et  al., 2009). Quercetin has also been

Nutraceuticals.
DOI: http://dx.doi.org/10.1016/B978-0-12-802147-7.00032-2 447 © 2012
2016 Elsevier Inc. All rights reserved.
448 32. QUERCETIN

TABLE 32.1 Dietary Sources and Concentrations of Quercetin

Quercetin Quercetin
content content
Food source (mg/100 g) Food source (mg/100 g)

Capers, raw 234 Red chard leaves, raw 7


Dill weed, fresh 55 Chicory greens, raw 6
Oregano, fresh 7 Chives, raw 5
FIGURE 32.1 Chemical structure of quercetin.
Apples with skin 4 Cilantro leaves, raw 53
Bayberries, raw 4 Corn poppy, leaves 26
Blackberries, raw 4 Dock, raw 86
Blueberries, raw 4 Drumstick leaves, raw 17 wherein pretreatment with quercetin protected against
Cherries, sweet, raw 2 Fennel leaves, raw 49 B(a)P-induced cytotoxicity by increasing the activity of
antioxidants (SOD, CAT, GPx, GST) (Kamaraj et al., 2007).
Chokeberry, raw 19 Hartwort leaves 29
More importantly, epidemiological studies reported an
Cranberries, raw 15 Kale, raw 23 inverse association of quercetin and lung cancer (Knekt
Crowberries, raw 5 Lettuce, red leaf, raw 8 et al., 2002; Cui et al., 2008).
Quercetin has also been shown to reduce chromo-
Currants, black, raw 4 Lovage, leaves, raw 170
somal aberration during 7,12-dimethylbenz(a)anthra-
Elderberries, raw 27 Mizuna 9 cene (DMBA)-induced mouse skin carcinogenesis
Figs, raw 5 Mustard greens, raw 9 (Sengupta et  al., 2001). Similarly, quercetin treatment
Goji berry, dried 14 Nalta jute, raw 24 inhibited the DMBA-TPA-induced tumor formation in
the BK5.IGF-1 transgenic mouse model for skin carcino-
Grapes, black 2 Okra, raw 21
genesis and it suppressed activation of IGF-1 signaling
Juniper berries, ripe 47 Onions, red, raw 39 (Jung et al., 2013).
Lingonberries, raw 13 Peppers, yellow wax, 51 In addition to the aforementioned studies, anticancer
raw effects of quercetin have also been investigated in other
Pitanga, raw 6 Radicchio, raw 32 types of cancer, including pancreatic, ovarian, and gas-
tric cancer (Angst et al., 2013; Maciejczyk and Surowiak,
Plums, purple, raw 2 Radish leaves, raw 70
2013; Ramachandran et al., 2013). Taken together, quer-
Rowanberries, raw 7 Rocket, wild, raw 66 cetin can act as an anticancer agent by virtue of its abil-
Saskatoon berries 17 Sweet potato leaves, 17 ity to modulate proapoptotic and antiapoptotic proteins,
raw cell cycle progression, angiogenesis, autophagy, inflam-
Arugula, raw 8 Watercress, raw 30 mation, protein kinases involved in cell proliferation,
Asparagus, raw 14 Chia seeds, raw 18
and telomerase activity (Lee et al., 2004; Psahoulia et al.,
2007; Boly et  al., 2011; Cosan et  al., 2011; Russo et  al.,
Broccoli, raw 3 Carob fiber 58 2012).

QUERCETIN AND CARDIOVASCULAR


demonstrated to suppress cell proliferation in HT-29 DISEASES
colon cancer cells by activating AMPK and decreasing
the expression of COX-2, which is known to be overex- Quercetin is a well-known inhibitor of cellular prolif-
pressed in colon cancer (Lee et al., 2009). Moreover, quer- eration and also an effective minimizer of DNA damage
cetin inhibited colorectal carcinogenesis and reduced the (Boyle et al., 2000; Choi et al., 2008). A natural source of
size of colorectal tumors in azoxymethane-treated rats quercetin from plants has been shown to increase the
(Dihal et al., 2006). bioavailability of this flavonoid (Miean and Mohamed,
Quercetin has been reported to induce apoptosis in 2001). Several studies have demonstrated a protective
A549 lung cancer cells via caspase-3 activation, PARP response by quercetin against cardiovascular disorders
cleavage, ERK activation, and c-Jun phosphorylation (Larson et al., 2012).
(Nguyen et  al., 2004). The chemopreventive efficacy of Quercetin exhibits a number of properties that lead
quercetin was investigated using benzo(a)pyrene (B(a) to cardiovascular protection: it is antioxidative (da Silva
P)-treated mice as an in vivo model of lung carcinogenesis, et al., 2000; Chaillou and Nazareno, 2006) and antiplatelet,

NUTRACEUTICALS
QUERCETIN AND NEURODEGENERATIVE DISEASES 449
induces antismooth muscle cell proliferation and migra- QUERCETIN AND
tion, is an enhancer of cardiac cell mitochondrial func- NEURODEGENERATIVE DISEASES
tion, and is an inhibitor of NF-κB (Gryglewski et  al.,
1987; Duarte et  al., 1993a,b; Hajra et  al., 2000; Alcocer Oxidative stress is recognized as an important factor in
et al., 2002; Moon et al., 2003). the pathogenesis of several neurodegenerative diseases
Many animal studies involving diet supplementation such as Alzheimer’s disease (AD), Parkinson’s disease
with quercetin have shown to be protective against car- (PD), and Huntington’s disease (HD). The brain is highly
diac problems. Studies have demonstrated that a quer- susceptible to oxidative stress, given its high content of
cetin-supplemented diet attenuated the development of polyunsaturated fatty acids, high oxygen consumption,
cardiac hypertrophy in a rat model of pressure overload and lower antioxidant capacity (Sies, 1993; Bellissimo
(Han et  al., 2009; Gao et  al., 2014). Moreover, a recent et  al., 2001; Freitas et  al., 2004). Therefore, increasing
study conducted by Jimenez et al. (2015) also showed a the brain’s antioxidant capacity may provide protection
promising effect of quercetin. Their study revealed that against oxidative stress–induced neurodegeneration.
quercetin and its plasma metabolites effectively lowered Recently, different groups of polyphenols have been
levels of vascular superoxide by inhibiting NADPH tested for their disease-modifying potential for neu-
activity in the vascular smooth muscle cells. On the rodegenerative diseases. For quercetin, there is now
contrary, a few studies have shown that the risk and compelling evidence of its neuroprotective role in vari-
severity of cardiovascular problems were not lessened ous neurodegenerative diseases (Ansari et  al., 2009;
in a spontaneously hypertensive rat model receiving a Haleagrahara et al., 2013; Karuppagounder et al., 2013;
quercetin-supplemented diet (Carlstrom et al., 2007). Sabogal-Guaqueta et al., 2015).
Epidemiological evidence also indicates that querce- Quercetin has been explored as a promising disease-
tin is protective against cardiac diseases. One of the most modifying neuroprotective agent for AD in various
famous studies was the Zutphen Elderly Study, which experimental models. A glucoside form of quercetin,
demonstrated that increasing flavonoid (high quercetin quercetin-3′-glucoside, reduced H2O2-induced ROS gen-
content) intake by dietary means decreased the risk of eration and also protected against Aβ-induced cell death
heart disease mortality (Hertog et  al., 1993). One other in PC12 cells (Zhu et al., 2007). Similarly, another study
study showing a promising protective effect of querce- investigated whether quercetin protects against oxidative
tin against cardiovascular disease risk was performed stress and Aβ-induced cell death in primary neurons. It
by Egert et  al. (2009). The study involved overweight was found that lower concentrations of quercetin (5 and
subjects between ages 25 and 65 years with a high car- 10 µM) reduced protein carbonyl, 3-NT, and 4-hydroxy-
diovascular disease risk phenotype. The subjects were 2-nonenal levels, as wells as cell death in Aβ-treated
randomized in a double-blind, placebo-controlled cross- primary cortical neuron cultures, whereas higher con-
over trial, with the quercetin treatment group receiving centrations of quercetin (20 and 40 µM) showed oppo-
approximately 150 mg/day for a period of 6 weeks fol- site effects (Ansari et  al., 2009). Moreover, the role of
lowed by a 5-week washout period. The results of this ginkgo flavonols (mainly composed of quercetin, kaemp-
study stated that quercetin lowered the systolic blood ferol, and isorhamnetin) in a depression-related signal-
pressure and plasma levels of oxidized LDL in subjects ing pathway mediated by BDNF/pCREB was tested in
with a high cardiovascular disease risk phenotype (Egert double-transgenic (Tg APP/PS1) mice (Hou et al., 2010).
et al., 2009). During the past few decades, a number of They found that gingko flavonols stimulated the CREB-
consolidated studies have highlighted the efficiency of BDNF signaling pathway, reduced Aβ production in the
quercetin in preventing coronary heart disease (Formica hippocampus, and also reversed the spatial learning
and Regelson, 1995; Pace-Asciak et al., 1995; Saleem and deficit in transgenic mice, as evidenced by improved
Basha, 2010). Moreover, these studies also provided us performance in the Morris water maze. The potential
novel insights into understanding the effects of dietary neuroprotective effects of quercetin have also been
quercetin on body metabolism. Apart from the field of investigated in a triple-transgenic mouse model of AD
cardiovascular diseases, quercetin has also proved to (3xTg-AD) (Sabogal-Guaqueta et al., 2015). Their results
be a useful supplement for athletes. Many studies have revealed that quercetin treatment significantly reversed
shown that intake of quercetin supplements by athletes β-amyloidosis and tauopathy and reduced astroglio-
improved their performance and generally facilitated sis and microgliosis in 3xTg-AD mice. Furthermore, in
healthy maintenance of the body (Nieman et  al., 2010; comparison with vehicle-treated 3xTg-AD mice, quer-
Askari et al., 2013). In line with these data, several recent cetin-treated 3xTg-AD mice performed better in learn-
reports demonstrated that quercetin supplements also ing and memory tasks. Because acetylcholinesterase
strengthened the development of immunity in athletes (AChE) inhibitors that are widely used in the treatment
(Walsh et al., 2011; Gleeson, 2013). of AD may cause notable side effects, the possible AChE

NUTRACEUTICALS
450 32. QUERCETIN

inhibitory effect of quercetin was tested and compared rats. Finally, it has recently been shown that quercetin
with conventional drugs by an in silico analysis (Islam acts as an anti-inflammatory agent that restricts MPP+-
et al., 2013). Their analysis revealed that quercetin’s bind- induced microglial activation (Bournival et  al., 2012).
ing strength to the active site of the enzyme was better They found that pretreatment with quercetin reduced
than conventional drugs and that a 4-OH methylated MPP+-induced IL-6, IL-1β, iNOS, and TNFα expression
form of quercetin (azaleatin) had an even better binding in N9 microglial cells. They further demonstrated that
affinity than quercetin, suggesting the natural compound pretreatment with quercetin protected against cell death
quercetin has significant AChE inhibitory effect as com- induced by microglial activation when PC12 cells were
pared to widely used conventional drugs. cocultured with N9 cells treated with MPP+.
Because of its antioxidant and anti-inflammatory The protective effects of quercetin have also been
properties, quercetin has been gaining interest as a observed in other neurodegenerative diseases, includ-
neuroprotective agent for PD. Isoquercetin, a glycoside ing HD and Amyotrophic lateral sclerosis (Said Ahmed
form of quercetin, has been shown to play a neuropro- et  al., 2000; Sandhir and Mehrotra, 2013; Chakraborty
tective role against 6-OHDA-induced neurotoxicity in et  al., 2014). Collectively, all these studies suggest that
PC12 cells (Magalingam et al., 2014). In this study, they quercetin is a promising neuroprotective agent for the
showed that pretreatment with isoquercetin increased treatment of neurodegenerative diseases.
levels of ROS-scavenging enzymes (SOD, CAT, and
GPx) and reduced lipid peroxidation in 6-OHDA-treated
PC12 cells. Similarly, quercetin was shown to reduce CONCLUDING REMARKS AND
protein carbonyl content and lipid hydroperoxide levels FUTURE DIRECTIONS
in the striatum of 6-OHDA-treated rats (Haleagrahara
et  al., 2011). They also found that quercetin treatment This chapter describes the potential use of querce-
significantly reduced striatal dopamine depletion and tin in the prevention of cancer, cardiovascular diseases,
total glutathione (GSH) depletion and improved neuro- and neurodegenerative diseases. As described, there
nal survival in 6-OHDA-treated rats. Interestingly, quer- is strong evidence that quercetin can modulate sev-
cetin has also been shown to enhance cognitive function eral cellular signaling pathways involved in regulating
in 6-OHDA-treated rats as evidenced by improved per- the antioxidant response, cell survival, apoptosis, and
formance in the Morris water maze (Sriraksa et al., 2012). inflammation. More large-scale epidemiological studies
Additionally, their results demonstrated that quercetin and well-designed clinical trials are needed to further
treatment decreased hippocampal AChE activity and clarify the potential benefits and to substantiate the effi-
protected against neurodegeneration in all sub-regions of cacy of quercetin for the prevention and treatment of
the hippocampus. Likewise, quercetin was also reported human diseases.
to protect against 1-methyl-4-phenylpyridinium ion
(MPP+)-induced neurotoxicity in PC12 cells (Bournival
et  al., 2009). These authors found that pretreatment Acknowledgments
with quercetin significantly reduced MPP+-induced cell The writing of this chapter was supported by the National Institutes of
death, as evidenced by decreases in both caspase-3 acti- Health RO1 grants NS039958 and NS074443. The W. Eugene and Linda
Lloyd Endowed Chair to A.G.K. and the Dean Professorship to A.K.
vation and DNA fragmentation. Furthermore, decreased
are also acknowledged. The authors also acknowledge Gary Zenitsky
nuclear translocation of apoptosis-inducing factor and for his assistance in the preparation of this manuscript.
decreased cytochrome c release into cytosol were also
observed in this study. Another study explored the
potential protective effect of quercetin in a 1-methyl-4- References
phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) mouse Alcocer, F., Whitley, D., Salazar-Gonzalez, J.F., et  al., 2002. Quercetin
model of PD (Lv et al., 2012). Their results revealed that inhibits human vascular smooth muscle cell proliferation and
quercetin treatment reduced motor deficits and striatal migration. Surgery 131, 198–204.
dopamine depletion and also increased SOD and GPx Angst, E., Park, J.L., Moro, A., et  al., 2013. The flavonoid quercetin
inhibits pancreatic cancer growth in vitro and in vivo. Pancreas
activities in MPTP-treated mice. Furthermore, quercetin
42, 223–229.
treatment reduced lipid peroxidation in MPTP-treated Ansari, M.A., Abdul, H.M., Joshi, G., et  al., 2009. Protective effect of
mice, as evidenced by reduced immunoreactivity of quercetin in primary neurons against Abeta(1-42): relevance to
4-HNE in striatal neurons. Quercetin has also been Alzheimer’s disease. J. Nutr. Biochem. 20, 269–275.
shown to protect against rotenone-induced nigral neu- Askari, G., Ghiasvand, R., Paknahad, Z., et  al., 2013. The effects of
quercetin supplementation on body composition, exercise perfor-
rodegeneration (Karuppagounder et  al., 2013). Their
mance and muscle damage indices in athletes. Int. J. Prev. Med.
results showed that quercetin treatment improved mito- 4, 21–26.
chondrial complex I activity and reduced striatal dopa- Bellissimo, M.I., Amado, D., Abdalla, D.S., et al., 2001. Superoxide dis-
mine and nigral GSH depletion in rotenone-lesioned mutase, glutathione peroxidase activities and the hydroperoxide

NUTRACEUTICALS
REFERENCES 451
concentration are modified in the hippocampus of epileptic rats. Freitas, R.M., Nascimento, V.S., Vasconcelos, S.M., et al., 2004. Catalase
Epilepsy Res. 46, 121–128. activity in cerebellum, hippocampus, frontal cortex and striatum
Boly, R., Gras, T., Lamkami, T., et al., 2011. Quercetin inhibits a large after status epilepticus induced by pilocarpine in Wistar rats.
panel of kinases implicated in cancer cell biology. Int. J. Oncol. Neurosci. Lett. 365, 102–105.
38, 833–842. Gao, C., Chen, X., Li, J., et al., 2014. Myocardial mitochondrial oxida-
Boots, A.W., Wilms, L.C., Swennen, E.L., et  al., 2008. In vitro and ex tive stress and dysfunction in intense exercise: regulatory effects
vivo anti-inflammatory activity of quercetin in healthy volunteers. of quercetin. Eur. J. Appl. Physiol. 114, 695–705.
Nutrition 24, 703–710. Gleeson, M., 2013. Nutritional support to maintain proper immune
Bournival, J., Quessy, P., Martinoli, M.G., 2009. Protective effects of status during intense training. Nestle Nutr. Inst. Workshop Ser.
resveratrol and quercetin against MPP+ -induced oxidative stress 75, 85–97.
act by modulating markers of apoptotic death in dopaminergic Gryglewski, R.J., Korbut, R., Robak, J., et al., 1987. On the mechanism
neurons. Cell. Mol. Neurobiol. 29, 1169–1180. of antithrombotic action of flavonoids. Biochem. Pharmacol. 36,
Bournival, J., Plouffe, M., Renaud, J., et al., 2012. Quercetin and sesa- 317–322.
min protect dopaminergic cells from MPP+-induced neuroinflam- Hajra, L., Evans, A.I., Chen, M., et  al., 2000. The NF-kappa B signal
mation in a microglial (N9)-neuronal (PC12) coculture system. transduction pathway in aortic endothelial cells is primed for
Oxid. Med. Cell. Longev. 2012, 921941. activation in regions predisposed to atherosclerotic lesion forma-
Boyle, S.P., Dobson, V.L., Duthie, S.J., et al., 2000. Absorption and DNA tion. Proc. Natl. Acad. Sci. U.S.A. 97, 9052–9057.
protective effects of flavonoid glycosides from an onion meal. Eur. Haleagrahara, N., Siew, C.J., Mitra, N.K., et al., 2011. Neuroprotective
J. Nutr. 39, 213–223. effect of bioflavonoid quercetin in 6-hydroxydopamine-induced
Carlstrom, J., Symons, J.D., Wu, T.C., et al., 2007. A quercetin supple- oxidative stress biomarkers in the rat striatum. Neurosci. Lett.
mented diet does not prevent cardiovascular complications in 500, 139–143.
spontaneously hypertensive rats. J. Nutr. 137, 628–633. Haleagrahara, N., Siew, C.J., Ponnusamy, K., 2013. Effect of quercetin
Chaillou, L.L., Nazareno, M.A., 2006. New method to determine and desferrioxamine on 6-hydroxydopamine (6-OHDA) induced
antioxidant activity of polyphenols. J. Agric. Food Chem. 54, neurotoxicity in striatum of rats. J. Toxicol. Sci. 38, 25–33.
8397–8402. Han, J.J., Hao, J., Kim, C.H., et  al., 2009. Quercetin prevents cardiac
Chakraborty, J., Singh, R., Dutta, D., et  al., 2014. Quercetin improves hypertrophy induced by pressure overload in rats. J. Vet. Med.
behavioral deficiencies, restores astrocytes and microglia, and Sci. 71, 737–743.
reduces serotonin metabolism in 3-nitropropionic acid-induced He, X.Z., Wang, X., Dixon, R.A., 2006. Mutational analysis of the
rat model of Huntington’s disease. CNS Neurosci. Ther. 20, 10–19. Medicago glycosyltransferase UGT71G1 reveals residues that
Choi, E.J., Bae, S.M., Ahn, W.S., 2008. Antiproliferative effects of quer- control regioselectivity for (iso)flavonoid glycosylation. J. Biol.
cetin through cell cycle arrest and apoptosis in human breast Chem. 281, 34441–34447.
cancer MDA-MB-453 cells. Arch. Pharm. Res. 31, 1281–1285. Hertog, M.G., Feskens, E.J., Hollman, P.C., et al., 1993. Dietary antioxi-
Conklin, C.M., Bechberger, J.F., MacFabe, D., et  al., 2007. Genistein dant flavonoids and risk of coronary heart disease: the Zutphen
and quercetin increase connexin43 and suppress growth of breast Elderly Study. Lancet 342, 1007–1011.
cancer cells. Carcinogenesis 28, 93–100. Hertog, M.G., Kromhout, D., Aravanis, C., et al., 1995. Flavonoid intake
Cosan, D.T., Soyocak, A., Basaran, A., et  al., 2011. Effects of various and long-term risk of coronary heart disease and cancer in the
agents on DNA fragmentation and telomerase enzyme activities seven countries study. Arch. Intern. Med. 155, 381–386.
in adenocarcinoma cell lines. Mol. Biol. Rep. 38, 2463–2469. Hou, Y., Aboukhatwa, M.A., Lei, D.L., et  al., 2010. Anti-depressant
Cui, Y., Morgenstern, H., Greenland, S., et al., 2008. Dietary flavonoid natural flavonols modulate BDNF and beta amyloid in neurons
intake and lung cancer—a population-based case-control study. and hippocampus of double TgAD mice. Neuropharmacology
Cancer 112, 2241–2248. 58, 911–920.
da Silva, E.L., Abdalla, D.S., Terao, J., 2000. Inhibitory effect of flavo- Islam, M.R., Zaman, A., Jahan, I., et al., 2013. In silico QSAR analysis of
noids on low-density lipoprotein peroxidation catalyzed by mam- quercetin reveals its potential as therapeutic drug for Alzheimer’s
malian 15-lipoxygenase. IUBMB Life 49, 289–295. disease. J. Young Pharm. 5, 173–179.
Deng, X.H., Song, H.Y., Zhou, Y.F., et al., 2013. Effects of quercetin on Iwao, K., Tsukamoto, I., 1999. Quercetin inhibited DNA synthesis
the proliferation of breast cancer cells and expression of survivin. and induced apoptosis associated with increase in c-fos mRNA
Exp. Ther. Med. 6, 1155–1158. level and the upregulation of p21WAF1CIP1 mRNA and protein
Dihal, A.A., de Boer, V.C., van der Woude, H., et al., 2006. Quercetin, expression during liver regeneration after partial hepatectomy.
but not its glycosidated conjugate rutin, inhibits azoxymeth- Biochim. Biophys. Acta 1427, 112–120.
ane-induced colorectal carcinogenesis in F344 rats. J. Nutr. 136, Jimenez, R., Lopez-Sepulveda, R., Romero, M., et al., 2015. Quercetin
2862–2867. and its metabolites inhibit the membrane NADPH oxidase activity
Duarte, J., Perez-Vizcaino, F., Zarzuelo, A., et  al., 1993a. Vasodilator in vascular smooth muscle cells from normotensive and spontane-
effects of quercetin in isolated rat vascular smooth muscle. Eur. J. ously hypertensive rats. Food Funct. 6, 409–414.
Pharmacol. 239, 1–7. Jung, M., Bu, S.Y., Tak, K.H., et  al., 2013. Anticarcinogenic effect of
Duarte, J., Perez Vizcaino, F., Utrilla, P., et  al., 1993b. Vasodilatory quercetin by inhibition of insulin-like growth factor (IGF)-1 signal-
effects of flavonoids in rat aortic smooth muscle. Structure-activity ing in mouse skin cancer. Nutr. Res. Pract. 7, 439–445.
relationships. Gen. Pharmacol. 24, 857–862. Kamaraj, S., Vinodhkumar, R., Anandakumar, P., et  al., 2007. The
Duthie, S.J., Dobson, V.L., 1999. Dietary flavonoids protect human colo- effects of quercetin on antioxidant status and tumor markers in
nocyte DNA from oxidative attack in vitro. Eur. J. Nutr. 38, 28–34. the lung and serum of mice treated with benzo(a)pyrene. Biol.
Egert, S., Bosy-Westphal, A., Seiberl, J., et al., 2009. Quercetin reduces Pharm. Bull. 30, 2268–2273.
systolic blood pressure and plasma oxidised low-density lipopro- Karuppagounder, S.S., Madathil, S.K., Pandey, M., et al., 2013. Quercetin
tein concentrations in overweight subjects with a high-cardiovas- up-regulates mitochondrial complex-I activity to protect against
cular disease risk phenotype: a double-blinded, placebo-controlled programmed cell death in rotenone model of Parkinson’s disease
cross-over study. Br. J. Nutr. 102, 1065–1074. in rats. Neuroscience 236, 136–148.
Formica, J.V., Regelson, W., 1995. Review of the biology of quercetin Kim, Y.H., Lee, Y.J., 2007. TRAIL apoptosis is enhanced by quercetin
and related bioflavonoids. Food Chem. Toxicol. 33, 1061–1080. through Akt dephosphorylation. J. Cell. Biochem. 100, 998–1009.

NUTRACEUTICALS
452 32. QUERCETIN

Knekt, P., Kumpulainen, J., Jarvinen, R., et al., 2002. Flavonoid intake Russo, M., Palumbo, R., Tedesco, I., et  al., 1999. Quercetin and anti-
and risk of chronic diseases. Am. J. Clin. Nutr. 76, 560–568. CD95(Fas/Apo1) enhance apoptosis in HPB-ALL cell line. FEBS
Laird, D.W., Fistouris, P., Batist, G., et  al., 1999. Deficiency of con- Lett. 462, 322–328.
nexin43 gap junctions is an independent marker for breast tumors. Russo, M., Spagnuolo, C., Tedesco, I., et al., 2012. The flavonoid querce-
Cancer Res. 59, 4104–4110. tin in disease prevention and therapy: facts and fancies. Biochem.
Lamson, D.W., Brignall, M.S., 2000. Antioxidants and cancer, part 3: Pharmacol. 83, 6–15.
quercetin. Altern. Med. Rev. 5, 196–208. Sabogal-Guaqueta, A.M., Munoz-Manco, J.I., Ramirez-Pineda, J.R.,
Larson, A.J., Symons, J.D., Jalili, T., 2012. Therapeutic potential of quer- et  al., 2015. The flavonoid quercetin ameliorates Alzheimer’s
cetin to decrease blood pressure: review of efficacy and mecha- disease pathology and protects cognitive and emotional func-
nisms. Adv. Nutr. 3, 39–46. tion in aged triple transgenic Alzheimer’s disease model mice.
Lee, D.H., Szczepanski, M., Lee, Y.J., 2008. Role of Bax in querce- Neuropharmacology 93C, 134–145.
tin-induced apoptosis in human prostate cancer cells. Biochem. Said Ahmed, M., Hung, W.Y., Zu, J.S., et  al., 2000. Increased reac-
Pharmacol. 75, 2345–2355. tive oxygen species in familial amyotrophic lateral sclerosis with
Lee, L.T., Huang, Y.T., Hwang, J.J., et  al., 2004. Transinactivation of mutations in SOD1. J. Neurol. Sci. 176, 88–94.
the epidermal growth factor receptor tyrosine kinase and focal Saleem, T.S., Basha, S.D., 2010. Red wine: a drink to your heart. J
adhesion kinase phosphorylation by dietary flavonoids: effect on Cardiovasc. Dis. Res. 1, 171–176.
invasive potential of human carcinoma cells. Biochem. Pharmacol. Sandhir, R., Mehrotra, A., 2013. Quercetin supplementation is effec-
67, 2103–2114. tive in improving mitochondrial dysfunctions induced by 3-nitro-
Lee, Y.K., Park, S.Y., Kim, Y.M., et al., 2009. AMP kinase/cyclooxygen- propionic acid: implications in Huntington’s disease. Biochim.
ase-2 pathway regulates proliferation and apoptosis of cancer cells Biophys. Acta 1832, 421–430.
treated with quercetin. Exp. Mol. Med. 41, 201–207. Sengupta, A., Ghosh, S., Das, S., 2001. Modulation of DMBA induced
Lim, J.H., Park, J.W., Min, D.S., et al., 2007. NAG-1 up-regulation medi- genotoxicity in bone marrow by quercetin during skin carcinogen-
ated by EGR-1 and p53 is critical for quercetin-induced apoptosis esis. J. Exp. Clin. Cancer Res. 20, 131–134.
in HCT116 colon carcinoma cells. Apoptosis 12, 411–421. Senthilkumar, K., Arunkumar, R., Elumalai, P., et al., 2011. Quercetin
Lv, C., Hong, T., Yang, Z., et  al., 2012. Effect of quercetin in the inhibits invasion, migration and signalling molecules involved in
1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-induced mouse cell survival and proliferation of prostate cancer cell line (PC-3).
model of Parkinson’s disease. Evid. Based Complement. Alternat. Cell Biochem. Funct. 29, 87–95.
Med. 2012, 928643. Shan, B.E., Wang, M.X., Li, R.Q., 2009. Quercetin inhibit human SW480
Maciejczyk, A., Surowiak, P., 2013. Quercetin inhibits proliferation and colon cancer growth in association with inhibition of cyclin D1
increases sensitivity of ovarian cancer cells to cisplatin and pacli- and survivin expression through Wnt/beta-catenin signaling
taxel. Ginekol. Pol. 84, 590–595. pathway. Cancer Invest. 27, 604–612.
Magalingam, K.B., Radhakrishnan, A., Haleagrahara, N., 2014. Protective Sharma, H., Sen, S., Singh, N., 2005. Molecular pathways in the che-
effects of flavonol isoquercitrin, against 6-hydroxy dopamine mosensitization of cisplatin by quercetin in human head and neck
(6-OHDA)-induced toxicity in PC12 cells. BMC Res. Notes 7, 49. cancer. Cancer Biol. Ther. 4, 949–955.
Miean, K.H., Mohamed, S., 2001. Flavonoid (myricetin, quercetin, Sharmila, G., Bhat, F.A., Arunkumar, R., et al., 2014. Chemopreventive
kaempferol, luteolin, and apigenin) content of edible tropical effect of quercetin, a natural dietary flavonoid on prostate cancer
plants. J. Agric. Food Chem. 49, 3106–3112. in in vivo model. Clin. Nutr. 33, 718–726.
Moon, S.K., Cho, G.O., Jung, S.Y., et al., 2003. Quercetin exerts multiple Sies, H., 1993. Strategies of antioxidant defense. Eur. J. Biochem. 215,
inhibitory effects on vascular smooth muscle cells: role of ERK1/2, 213–219.
cell-cycle regulation, and matrix metalloproteinase-9. Biochem. Spagnuolo, C., Cerella, C., Russo, M., et al., 2011. Quercetin downregu-
Biophys. Res. Commun. 301, 1069–1078. lates Mcl-1 by acting on mRNA stability and protein degradation.
Nguyen, T.T., Tran, E., Nguyen, T.H., et al., 2004. The role of activated Br. J. Cancer 105, 221–230.
MEK-ERK pathway in quercetin-induced growth inhibition and Sriraksa, N., Wattanathorn, J., Muchimapura, S., et al., 2012. Cognitive-
apoptosis in A549 lung cancer cells. Carcinogenesis 25, 647–659. enhancing effect of quercetin in a rat model of Parkinson’s dis-
Nieman, D.C., Williams, A.S., Shanely, R.A., et  al., 2010. Quercetin’s ease induced by 6-hydroxydopamine. Evid. Based Complement.
influence on exercise performance and muscle mitochondrial bio- Alternat. Med. 2012, 823206.
genesis. Med. Sci. Sports Exerc. 42, 338–345. Vijayababu, M.R., Kanagaraj, P., Arunkumar, A., et al., 2005. Quercetin-
Pace-Asciak, C.R., Hahn, S., Diamandis, E.P., et al., 1995. The red wine induced growth inhibition and cell death in prostatic carcinoma
phenolics trans-resveratrol and quercetin block human platelet cells (PC-3) are associated with increase in p21 and hypophos-
aggregation and eicosanoid synthesis: implications for protection phorylated retinoblastoma proteins expression. J. Cancer Res.
against coronary heart disease. Clin. Chim. Acta 235, 207–219. Clin. Oncol. 131, 765–771.
Park, C.H., Chang, J.Y., Hahm, E.R., et  al., 2005. Quercetin, a potent Walsh, N.P., Gleeson, M., Pyne, D.B., et  al., 2011. Position statement.
inhibitor against beta-catenin/Tcf signaling in SW480 colon cancer Part two: maintaining immune health. Exerc. Immunol. Rev. 17,
cells. Biochem. Biophys. Res. Commun. 328, 227–234. 64–103.
Psahoulia, F.H., Moumtzi, S., Roberts, M.L., et al., 2007. Quercetin medi- Williams, C.A., Grayer, R.J., 2004. Anthocyanins and other flavonoids.
ates preferential degradation of oncogenic Ras and causes autoph- Nat. Prod. Rep. 21, 539–573.
agy in Ha-RAS-transformed human colon cells. Carcinogenesis Zamin, L.L., Filippi-Chiela, E.C., Dillenburg-Pilla, P., et  al., 2009.
28, 1021–1031. Resveratrol and quercetin cooperate to induce senescence-like
Ramachandran, L., Manu, K.A., Shanmugam, M.K., et  al., 2013. growth arrest in C6 rat glioma cells. Cancer Sci. 100, 1655–1662.
Isorhamnetin inhibits proliferation and invasion and induces Zandi, K., Teoh, B.T., Sam, S.S., et  al., 2011. Antiviral activity of four
apoptosis through the modulation of peroxisome proliferator- types of bioflavonoid against dengue virus type-2. Virol. J. 8, 560.
activated receptor gamma activation pathway in gastric cancer. J. Zhu, J.T., Choi, R.C., Chu, G.K., et al., 2007. Flavonoids possess neu-
Biol. Chem. 287, 38028–38040. roprotective effects on cultured pheochromocytoma PC12 cells: a
Ramos, F.A., Takaishi, Y., Shirotori, M., et al., 2006. Antibacterial and comparison of different flavonoids in activating estrogenic effect
antioxidant activities of quercetin oxidation products from yellow and in preventing beta-amyloid-induced cell death. J. Agric. Food
onion (Allium cepa) skin. J. Agric. Food Chem. 54, 3551–3557. Chem. 55, 2438–2445.

NUTRACEUTICALS

View publication stats

You might also like