ArticlePDF AvailableLiterature Review

Polyphenols in the Mediterranean Diet: From Dietary Sources to microRNA Modulation

MDPI
Antioxidants
Authors:
  • Dynamical Business & Science Society - DBSS International SAS
  • Universita' degli Studi "Magna Græcia" di Catanzaro (Italy)

Abstract and Figures

It is now well established that polyphenols are a class of natural substance that offers numerous health benefits; they are present in all plants in very different quantities and types. On the other hand, their bioavailability, and efficacy is are not always well proven. Therefore, this work aims to discuss some types of polyphenols belonging to Mediterranean foods. We chose six polyphenols—(1) Naringenin, (2) Apigenin, (3) Kaempferol, (4) Hesperidin, (5) Ellagic Acid and (6) Oleuropein—present in Mediterranean foods, describing dietary source and their chemistry, as well as their pharmacokinetic profile and their use as nutraceuticals/supplements, in addition to the relevant element of their capability in modulating microRNAs expression profile.
This content is subject to copyright.
antioxidants
Review
Polyphenols in the Mediterranean Diet: From Dietary Sources
to microRNA Modulation
Roberto Cannataro 1,2 , Alessia Fazio 1, Chiara La Torre 1,2, Maria Cristina Caroleo 1, 2, * and Erika Cione 1,2


Citation: Cannataro, R.; Fazio, A.; La
Torre, C.; Caroleo, M.C.; Cione, E.
Polyphenols in the Mediterranean
Diet: From Dietary Sources to microRNA
Modulation. Antioxidants 2021,10, 328.
https://doi.org/10.3390/antiox10020328
Academic Editor: Maria
Cristina Albertini
Received: 1 February 2021
Accepted: 16 February 2021
Published: 23 February 2021
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2021 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
1Department of Pharmacy, Health and Nutritional Sciences, Department of Excellence 2018-2022,
University of Calabria, Edificio Polifunzionale, 87036 Rende (CS), Italy; r.cannataro@gmail.com (R.C.);
alessia.fazio@unical.it (A.F.); latorre.chiara@libero.it (C.L.T.); erika.cione@unical.it (E.C.)
2GalaScreen Laboratories, Department of Pharmacy, Health and Nutrition Sciences, University of Calabria,
87036 Rende (CS), Italy
*Correspondence: mariacristina.caroleo@unical.it
Abstract:
It is now well established that polyphenols are a class of natural substance that offers
numerous health benefits; they are present in all plants in very different quantities and types. On
the other hand, their bioavailability, and efficacy is are not always well proven. Therefore, this
work aims to discuss some types of polyphenols belonging to Mediterranean foods. We chose
six polyphenols—(1) Naringenin, (2) Apigenin, (3) Kaempferol, (4) Hesperidin, (5) Ellagic Acid and
(6) Oleuropein—present in Mediterranean foods, describing dietary source and their chemistry, as
well as their pharmacokinetic profile and their use as nutraceuticals/supplements, in addition to the
relevant element of their capability in modulating microRNAs expression profile.
Keywords: polyphenols; nutraceutical; microRNA; epigenomic
1. Introduction
Angel Keys, a biologist and physiologist who based his conclusions on his studies
focusing on the dietary habits of people living in Southern Italy, was the first to present
the phrase “Mediterranean diet” to the popular imagination, investing it with a scientific
and cultural meaning. Now, following the joint candidacy of Italy, Spain, Greece and
Morocco, followed by Cyprus, Croatia and Portugal, it has the recognition of not only
UNESCO, but also of the WHO and FAO. The Mediterranean diet varies by country and
region, so it has a range of definitions. In general, however, it is high in vegetables, fruits,
legumes (such as beans), nuts, cereals, fish and unsaturated fats (such as olive oil). It
usually includes a low intake of meat and dairy foods. In particular, some plants are
characteristic of the Mediterranean vegetation; some have been present for thousands of
years, such as olive trees, walnuts, oregano, pomegranates, onions and others including
various citrus species [
1
]. The Mediterranean diet represents one of the first examples of
a positive correlation between diet and cardiovascular health; in fact, a diet that involves
the frequent eating of fruit and vegetables, possibly in season, in addition to seeds and
olive oil, has been shown to exhibit significant benefits in health terms, resulting not only
in preventing cardiovascular disease, but also diabetes, obesity and even various forms
of cancer [
2
5
], although the dietary polyphenol intake in Europe seems to be high in
the north [
6
]. This effect can be explained, at least in part, by the regular and varied
intake of food polyphenols that characterize the Mediterranean diet. Although there is
no set recommended daily dose, polyphenols have an important role in modulating and
preventing various diseases, such as cardiovascular [
6
], as well as inflammatory diseases
such as arthritis [
7
]. Probably the most important actions by polyphenols are carried out in
the regulation and management of reactive oxygen species (ROS) and immunomodulation.
The pathways that are influenced by much of them are those of nuclear factor kappa-light-
chain-enhancer of activated B cells (NF-
κ
B), mitogen-activated protein Kinase (MAPK) and
Antioxidants 2021,10, 328. https://doi.org/10.3390/antiox10020328 https://www.mdpi.com/journal/antioxidants
Antioxidants 2021,10, 328 2 of 24
arachidonic acids and phosphatidylinositide 3-kinases/protein kinase B (PI3K/AkT) as an
inhibitor. On the other hand, they upregulate superoxide dismutase (SOD), catalase and
glutathione peroxidase (GPx) expression: GPR40 [811].
Polyphenols are very often linked to the colors of the plants that contain them. They
are present in practically all plant species and in various parts of the plant itself, especially
in the leaves, fruits and roots [
12
]. On the other hand, while showing promising
in vitro
activity, they often present the obstacle of bioavailability, which does not always make
them so useful if tested directly on humans. Polyphenols have a typical molecular structure
with one or more aromatic rings, and one or more double bonds are present in the molecule.
This structure guarantees an antioxidant action for all classes, as there is delocalization
of the free radical itself, with consequent antioxidant activity. [
12
]. Together with this,
polyphenols have a genomic and epigenomic action, in fact there are numerous studies
that underline their regulatory action, among others, on NF-
κ
B, MAPK and nuclear factor
erythroid related factor 2 (Nrf2) [
11
,
13
,
14
]. In addition to that, they show epigenetic activity
in modulating microRNAs expression and from this point of view the microRNAs could
represent a useful evaluation tool to study polyphenols action in human. In this review, we
choose six polyphenols—(1) Naringenin, (2) Apigenin, (3) Kaempferol, (4) Hesperidin, (5)
Ellagic Acid and (6) Oleuropein—present in Mediterranean foods, as this dietetic lifestyle
is linked to better health status [25].
2. Dietary Sources
2.1. Naringenin
Naringenin is especially abundant in rosemary (55.1 mg/100 g) and present in grape-
fruit juice (37.76 mg/100 mL), red wine (0.75 mg/100 mL) and orange juice (
0.07 mg/100 mL
)
(Figure 1). Naringenin is a flavonoid belonging to the subclass of flavanones, also often
found in food in its glycosides form. [15].
Antioxidants2021,10,3282of24
chainenhancerofactivatedBcells(NF‐κB),mitogenactivatedproteinKinase(MAPK)
andarachidonicacidsandphosphatidylinositide3kinases/proteinkinaseB(PI3K/AkT)
asaninhibitor.Ontheotherhand,theyupregulatesuperoxidedismutase(SOD),catalase
andglutathioneperoxidase(GPx)expression:GPR40[8–11].
Polyphenolsareveryoftenlinkedtothecolorsoftheplantsthatcontainthem.They
arepresentinpracticallyallplantspeciesandinvariouspartsoftheplantitself,especially
intheleaves,fruitsandroots[12].Ontheotherhand,whileshowingpromisinginvitro
activity,theyoftenpresenttheobstacleofbioavailability,whichdoesnotalwaysmake
themsousefuliftesteddirectlyonhumans.Polyphenolshaveatypicalmolecularstruc
turewithoneormorearomaticrings,andoneormoredoublebondsarepresentinthe
molecule.Thisstructureguaranteesanantioxidantactionforallclasses,asthereisdelo
calizationofthefreeradicalitself,withconsequentantioxidantactivity.[12].Together
withthis,polyphenolshaveagenomicandepigenomicaction,infacttherearenumerous
studiesthatunderlinetheirregulatoryaction,amongothers,onNF‐κB,MAPKandnu
clearfactorerythroidrelatedfactor2(Nrf2)[11,13,14].Inadditiontothat,theyshowepi
geneticactivityinmodulatingmicroRNAsexpressionandfromthispointofviewthemi
croRNAscouldrepresentausefulevaluationtooltostudypolyphenolsactioninhuman.
Inthisreview,wechoosesixpolyphenols—(1)Naringenin,(2)Apigenin,(3)Kaempferol,
(4)Hesperidin,(5)EllagicAcidand(6)Oleuropein—presentinMediterraneanfoods,as
thisdieteticlifestyleislinkedtobetterhealthstatus[2–5].
2.DietarySources
2.1.Naringenin
Naringeninisespeciallyabundantinrosemary(55.1mg/100g)andpresentingrape
fruitjuice(37.76mg/100mL),redwine(0.75mg/100mL)andorangejuice(0.07mg/100
mL)(Figure1).Naringeninisaflavonoidbelongingtothesubclassofflavanones,also
oftenfoundinfoodinitsglycosidesform.[15].
Figure1.Mainfoodsandbeveragesthatcontainnaringenin,accordingtothedatabasePhenol
Explore(http://phenolexplorer.eu/)andUSDADatabasefortheflavonoidcontentofselected
foods(https://www.ars.usda.gov/).
2.2.Apigenin
ThenameapigeninderivesfromthegenusApiumintheApiaceaealsoknownas
Umbelliferaeandisfoundasauniqueingredientinchamomile(Matricariachamomilla),an
annualherbnativetowesternAsiaandEurope.Drinkspreparedfromchamomilecontain
from0.8%to1.2%ofapigenin.Apigeninisabundantinavarietyofotherdietarysources
[16],includingfruitsandvegetables(Figure2),suchasceleryseeds(78.65mg/100g),spin
ach(62.0mg/100g),parsley(45.04mg/100g),marjoram(4.40mg/100g),Italianoregano
(3.5mg/100g),sage(2.40mg/100g),chamomile(3to5mg/100g)andpistachio(0.03
mg/100g),buttherichestsourcesaretherespectivedriedsources.[17].
Figure 1.
Main foods and beverages that contain naringenin, according to the database Phenol-
Explore (http://phenol-explorer.eu/ accessed on 29 January 2021) and USDA Database for the
flavonoid content of selected foods (https://www.ars.usda.gov/ accessed on 13 February 2021).
2.2. Apigenin
The name apigenin derives from the genus Apium in the Apiaceae also known as
Umbelliferae and is found as a unique ingredient in chamomile (Matricaria chamomilla), an
annual herb native to western Asia and Europe. Drinks prepared from chamomile contain
from 0.8% to 1.2% of apigenin. Apigenin is abundant in a variety of other dietary sources [
16
],
including fruits and vegetables (Figure 2), such as celery seeds (7
8.65 mg/100 g)
, spinach
(62.0 mg/100 g), parsley (45.04 mg/100 g), marjoram (
4.40 mg/100 g)
, Italian oregano
(
3.5 mg/100 g
), sage (
2.40 mg/100 g)
, chamomile (3 to 5 mg/100 g) and pistachio
(0.03 mg/100 g)
,
but the richest sources are the respective dried sources. [17].
Antioxidants 2021,10, 328 3 of 24
Antioxidants2021,10,3283of24
Figure2.Mainfoodsthatcontainapigenin,accordingtothedatabasePhenolExplore(http://phenolexplorer.eu/)and
USDADatabasefortheflavonoidcontentofselectedfoods(https://www.ars.usda.gov/).
2.3.Kaempferol
Therichestplantsourcesofkaempferolaregreenleafyvegetables,suchasspinach
(55mg/100g),cabbage(47mg/100g)andbroccoli(7.2mg/100g),butalsoinonions(4.5
mg/100g)andblueberries(3.17mg/100g).Regardingdrinks,kaempferolismainlypre
sentinblacktea(1.7mg/100mL)andredwine(0.23mg/100mL).Agoodpercentageis
alsopresentinspicessuchascapers(104.29mg/100g),cumin(38.6mg/100g)andcloves
(23.8mg/100g)(Figure3).
Figure3.Mainfoodsandbeveragesthatcontainkaempferol,accordingtothedatabasePhenol
Explore(http://phenolexplorer.eu/)andUSDADatabasefortheflavonoidcontentofselected
foods(https://www.ars.usda.gov/).
Figure 2.
Main foods that contain apigenin, according to the database Phenol-Explore (http://phenol-
explorer.eu/ accessed on 29 January 2021) and USDA Database for the flavonoid content of selected
foods (https://www.ars.usda.gov/ accessed on 13 February 2021).
2.3. Kaempferol
The richest plant sources of kaempferol are green leafy vegetables, such as spinach
(55 mg/100 g), cabbage (47 mg/100 g) and broccoli (7.2 mg/100 g), but also in onions
(4.5 mg/100 g) and blueberries (3.17 mg/100 g). Regarding drinks, kaempferol is mainly
present in black tea (1.7 mg/100 mL) and red wine (0.23 mg/100 mL). A good percentage is
also present in spices such as capers (104.29 mg/100 g), cumin (38.6 mg/100 g) and cloves
(23.8 mg/100 g) (Figure 3).
Antioxidants2021,10,3283of24
Figure2.Mainfoodsthatcontainapigenin,accordingtothedatabasePhenolExplore(http://phenolexplorer.eu/)and
USDADatabasefortheflavonoidcontentofselectedfoods(https://www.ars.usda.gov/).
2.3.Kaempferol
Therichestplantsourcesofkaempferolaregreenleafyvegetables,suchasspinach
(55mg/100g),cabbage(47mg/100g)andbroccoli(7.2mg/100g),butalsoinonions(4.5
mg/100g)andblueberries(3.17mg/100g).Regardingdrinks,kaempferolismainlypre
sentinblacktea(1.7mg/100mL)andredwine(0.23mg/100mL).Agoodpercentageis
alsopresentinspicessuchascapers(104.29mg/100g),cumin(38.6mg/100g)andcloves
(23.8mg/100g)(Figure3).
Figure3.Mainfoodsandbeveragesthatcontainkaempferol,accordingtothedatabasePhenol
Explore(http://phenolexplorer.eu/)andUSDADatabasefortheflavonoidcontentofselected
foods(https://www.ars.usda.gov/).
Figure 3.
Main foods and beverages that contain kaempferol, according to the database Phenol-
Explore (http://phenol-explorer.eu/ accessed on 29 January 2021) and USDA Database for the
flavonoid content of selected foods (https://www.ars.usda.gov/ accessed on 13 February 2021).
Antioxidants 2021,10, 328 4 of 24
2.4. Hesperidin
Hesperidin and its aglycone, hesperetin, are two flavonoids, which together with rutin
and quercetin, are the main compounds of citrus fruits and, for this reason, this compound is
called “citroflavonoid”. It is present mainly in blood orange (
43.71 mg/100 mL
), mandarin
juice (36.11 mg/100 mL), blond orange juice (25.85 mg/100 mL), lemon (
17.81 mg/100 mL
)
and lime (13.41 mg/100 g) (Figure 4). The presence of this compound has also been detected
in plants; a high value is reported in peppermint (480.65 mg/100 g).
Antioxidants2021,10,3284of24
2.4.Hesperidin
Hesperidinanditsaglycone,hesperetin,aretwoflavonoids,whichtogetherwithru
tinandquercetin,arethemaincompoundsofcitrusfruitsand,forthisreason,thiscom
poundiscalled“citroflavonoid”.Itispresentmainlyinbloodorange(43.71mg/100mL),
mandarinjuice(36.11mg/100mL),blondorangejuice(25.85mg/100mL),lemon(17.81
mg/100mL)andlime(13.41mg/100g)(Figure4).Thepresenceofthiscompoundhasalso
beendetectedinplants;ahighvalueisreportedinpeppermint(480.65mg/100g).
Figure4.MainfoodsthatcontainhesperidinaccordingtothedatabasePhenolExplore(http://phe
nolexplorer.eu/)andUSDADatabasefortheflavonoidcontentofselectedfoods
(https://www.ars.usda.gov/).
2.5.EllagicAcid
Ellagicacid,adilactoneofthedimergallicacid,isapolyphenolfoundinfruitand
vegetables.Somefoodscontainamorecomplexversioncalledellagitannin,whichwillbe
convertedintoellagicacidbyorganism.Ellagicacidisprevalentinberries(Figure5).
Foodshighinellagicacidarechestnut(735.44mg/100g),blackberries(43.67mg/100g),
blackraspberries(38mg/100g),walnuts(28.50mg/100g),cloudberries(15.30mg/100g),
pomegranatejuice(2.06mg/100mL),strawberries(1.24mg/100g),redraspberries(1.14
mg/100g)andmuscadinegrape(0.90mg/100g)[18].Ellagicacidissynthetizedbyplants
asadefensemechanismagainstinfectionsandparasites.
Figure5.Structureandmainfoodsthatcontainellagicacid,accordingtothedatabasePhenol
Explore(http://phenolexplorer.eu/).ThereisnovaluereportedfortheUSDADatabaseforthe
flavonoidcontentofselectedfoods(https://www.ars.usda.gov/).
2.6.Oleuropein
Oleuropeinisthemoleculeresponsibleforthebittertasteofolivesandisthemost
commonphenoliccomponentintheleaves,seeds,pulpandskinofunripeolives.Alt
houghabundant,thiscompoundundergoeshydrolysisduringfruitripeningleadingto
Figure 4.
Main foods that contain hesperidin according to the database Phenol-Explore (http://
phenol-explorer.eu/ accessed on 29 January 2021) and USDA Database for the flavonoid content of
selected foods (https://www.ars.usda.gov/ accessed on 13 February 2021).
2.5. Ellagic Acid
Ellagic acid, a dilactone of the dimer gallic acid, is a polyphenol found in fruit and
vegetables. Some foods contain a more complex version called ellagitannin, which will
be converted into ellagic acid by organism. Ellagic acid is prevalent in berries
(Figure 5
).
Foods high in ellagic acid are chestnut (735.44 mg/100 g), blackberries (
43.67 mg/100 g
),
black raspberries (38 mg/100 g), walnuts (28.50 mg/100 g), cloudberries (1
5.30 mg/100 g
),
pomegranate juice (2.06 mg/100 mL), strawberries (1.24 mg/100 g), red raspberries
(
1.14 mg/100 g
) and muscadine grape (0.90 mg/100 g) [
18
]. Ellagic acid is synthetized by
plants as a defense mechanism against infections and parasites.
Antioxidants2021,10,3284of24
2.4.Hesperidin
Hesperidinanditsaglycone,hesperetin,aretwoflavonoids,whichtogetherwithru
tinandquercetin,arethemaincompoundsofcitrusfruitsand,forthisreason,thiscom
poundiscalled“citroflavonoid”.Itispresentmainlyinbloodorange(43.71mg/100mL),
mandarinjuice(36.11mg/100mL),blondorangejuice(25.85mg/100mL),lemon(17.81
mg/100mL)andlime(13.41mg/100g)(Figure4).Thepresenceofthiscompoundhasalso
beendetectedinplants;ahighvalueisreportedinpeppermint(480.65mg/100g).
Figure4.MainfoodsthatcontainhesperidinaccordingtothedatabasePhenolExplore(http://phe
nolexplorer.eu/)andUSDADatabasefortheflavonoidcontentofselectedfoods
(https://www.ars.usda.gov/).
2.5.EllagicAcid
Ellagicacid,adilactoneofthedimergallicacid,isapolyphenolfoundinfruitand
vegetables.Somefoodscontainamorecomplexversioncalledellagitannin,whichwillbe
convertedintoellagicacidbyorganism.Ellagicacidisprevalentinberries(Figure5).
Foodshighinellagicacidarechestnut(735.44mg/100g),blackberries(43.67mg/100g),
blackraspberries(38mg/100g),walnuts(28.50mg/100g),cloudberries(15.30mg/100g),
pomegranatejuice(2.06mg/100mL),strawberries(1.24mg/100g),redraspberries(1.14
mg/100g)andmuscadinegrape(0.90mg/100g)[18].Ellagicacidissynthetizedbyplants
asadefensemechanismagainstinfectionsandparasites.
Figure5.Structureandmainfoodsthatcontainellagicacid,accordingtothedatabasePhenol
Explore(http://phenolexplorer.eu/).ThereisnovaluereportedfortheUSDADatabaseforthe
flavonoidcontentofselectedfoods(https://www.ars.usda.gov/).
2.6.Oleuropein
Oleuropeinisthemoleculeresponsibleforthebittertasteofolivesandisthemost
commonphenoliccomponentintheleaves,seeds,pulpandskinofunripeolives.Alt
houghabundant,thiscompoundundergoeshydrolysisduringfruitripeningleadingto
Figure 5.
Structure and main foods that contain ellagic acid, according to the database Phenol-
Explore (http://phenol-explorer.eu/ accessed on 29 January 2021). There is no value reported for the
USDA Database for the flavonoid content of selected foods (https://www.ars.usda.gov/ accessed on
13 February 2021).
2.6. Oleuropein
Oleuropein is the molecule responsible for the bitter taste of olives and is the most
common phenolic component in the leaves, seeds, pulp and skin of unripe olives. Although
Antioxidants 2021,10, 328 5 of 24
abundant, this compound undergoes hydrolysis during fruit ripening leading to the pro-
duction of other important compounds such as hydroxytyrosol and ester derivatives [
19
].
It is important to underline that the oleuropein content may also depend on the variety of
the olive (in fact black and green olives contain 72.02 mg/100 mL and
55.58 mg/100 mL
,
respectively (Figure 6)), but also, and above all, on the processing techniques used to obtain
the oil [20].
Antioxidants2021,10,3285of24
theproductionofotherimportantcompoundssuchashydroxytyrosolandesterderiva
tives[19].Itisimportanttounderlinethattheoleuropeincontentmayalsodependonthe
varietyoftheolive(infactblackandgreenolivescontain72.02mg/100mLand55.58
mg/100mL,respectively(Figure6)),butalso,andaboveall,ontheprocessingtechniques
usedtoobtaintheoil[20].
Figure6.Oliveisthemainfoodthatcontainsoleuropien,accordingtothedatabasePhenolEx
plore(http://phenolexplorer.eu/).ThereisnovaluereportedfortheUSDADatabasefortheflavo
noidcontentofselectedfoods(https://www.ars.usda.gov/).
3.Chemistry
Polyphenolsarenaturalcompoundssynthesizedexclusivelybyplantsandcharac
terizedbytwophenylringsatleastandoneormorehydroxylsubstituents.Thisdescrip
tioncomprehendsalargenumberofheterogeneouscompoundswithreferencetotheir
complexity.Allphenoliccompoundsaresynthesizedthroughthephenylpropanoidpath
way(Figure7),startingfromtheaminoacidphenylalanine[21],whichis,inturn,aprod
uctoftheshikimatepathway;thelatterlinkscarbohydratemetabolismtothebiosynthesis
ofaromaticaminoacidsandsecondarymetabolitesshikimatepathway[22].
Figure 6.
Olive is the main food that contains oleuropien, according to the database Phenol-Explore
(http://phenol-explorer.eu/ accessed on 29 January 2021). There is no value reported for the USDA
Database for the flavonoid content of selected foods (https://www.ars.usda.gov/ accessed on
13 February 2021).
3. Chemistry
Polyphenols are natural compounds synthesized exclusively by plants and character-
ized by two phenyl rings at least and one or more hydroxyl substituents. This description
comprehends a large number of heterogeneous compounds with reference to their com-
plexity. All phenolic compounds are synthesized through the phenylpropanoid pathway
(Figure 7), starting from the amino acid phenylalanine [
21
], which is, in turn, a product
of the shikimate pathway; the latter links carbohydrate metabolism to the biosynthesis of
aromatic amino acids and secondary metabolites shikimate pathway [22].
The phenylpropanoid pathway leads to different classes of compounds which can
be can be simply classified into flavonoids and nonflavonoids, or be subdivided in many
subclasses depending on the structural diversity. This arises from the number of phenol
units within the structure, substituent groups, variations in hydroxylation pattern and/or
the linkage type between phenol units.
The flavonoid pathway (calchone synthase) leads to the synthesis of six major classes
of metabolites, such as flavonols, flavan-3-ols, anthocyanidins, proanthocyanidins and
anthocyanins. All these compounds share the basic structure of diphenyl propanes (C6-C3-
C6), in which phenolic rings (ring A and ring B) are usually linked by a heterocyclic ring
(ring C), which usually is a closed pyran, as shown in Figure 8[23].
Most flavonoids occur in edible plants and foods as
β
-glycosides, bound to one or
more sugar units with the exception of flavan-3-ols (catechins and proanthocyanidins) and
glycosylated isoflavones that are exposed to microbial
β
-glucosidases, which catalyze the
hydrolysis of the glycosidic bond.
3.1. Naringenin
Naringenin is a flavorless, colorless flavanone with three hydroxy groups at the 7, 5
and 4
0
carbons (Figure 9) [
24
]. It may be found both in the aglycol form, naringenin (a), or in
its glycosidic form, naringin (b), which has the addition of the disaccharide neohesperidose
attached via a glycosidic linkage at carbon.
Antioxidants 2021,10, 328 6 of 24
Entering the flavone synthesis pathway, enzyme chalcone synthase (CHS) catalyzes
consecutive condensations of three equivalents of malonyl CoA followed by aromatization
to convert starting p-coumaroyl-CoA to chalcone [
25
]. Chalcone isomerase (CHI) performs
stereospecific isomerization of tetrahydroxychalcone to (2S)-flavanone, which is the branch
point precursor of many important downstream flavonoids, including flavones (Figure 10).
Figure 7.
Phenylpropanoid Pathway. The metabolites of the shikimate pathway and p-coumaroyl
CoA are shaded in grey.
3.2. Kaempferol
Kaempferol (3,5,7-trihydroxy-2-(4-hydroxyphenyl)-4H-chromen-4-one) is a natural
flavonol found in common foods derived from plants and fruits. It is biosynthesized from
naringenin via a 2-hydroxy intermediate (Figure 12).
Antioxidants 2021,10, 328 7 of 24
Antioxidants2021,10,3287of24
Figure8.Basicstructuresofflavonoidsubclasses.
Mostflavonoidsoccurinedibleplantsandfoodsasβglycosides,boundtooneor
moresugarunitswiththeexceptionofflavan3ols(catechinsandproanthocyanidins)
andglycosylatedisoflavonesthatareexposedtomicrobialβglucosidases,whichcatalyze
thehydrolysisoftheglycosidicbond.
3.1.Naringenin
Naringeninisaflavorless,colorlessflavanonewiththreehydroxygroupsatthe7,5
and4′carbons(Figure9)[24].Itmaybefoundbothintheaglycolform,naringenin(a),or
initsglycosidicform,naringin(b),whichhastheadditionofthedisaccharideneohesper
idoseattachedviaaglycosidiclinkageatcarbon.
Figure9.Structuresofnaringenin(a)andnaringin(b).
Enteringtheflavonesynthesispathway,enzymechalconesynthase(CHS)catalyzes
consecutivecondensationsofthreeequivalentsofmalonylCoAfollowedbyaromatiza
tiontoconvertstartingpcoumaroylCoAtochalcone[25].Chalconeisomerase(CHI)per
formsstereospecificisomerizationoftetrahydroxychalconeto(2S)flavanone,whichisthe
branchpointprecursorofmanyimportantdownstreamflavonoids,includingflavones
(Figure10).
Figure 8. Basic structures of flavonoid subclasses.
Antioxidants2021,10,3287of24
Figure8.Basicstructuresofflavonoidsubclasses.
Mostflavonoidsoccurinedibleplantsandfoodsasβglycosides,boundtooneor
moresugarunitswiththeexceptionofflavan3ols(catechinsandproanthocyanidins)
andglycosylatedisoflavonesthatareexposedtomicrobialβglucosidases,whichcatalyze
thehydrolysisoftheglycosidicbond.
3.1.Naringenin
Naringeninisaflavorless,colorlessflavanonewiththreehydroxygroupsatthe7,5
and4′carbons(Figure9)[24].Itmaybefoundbothintheaglycolform,naringenin(a),or
initsglycosidicform,naringin(b),whichhastheadditionofthedisaccharideneohesper
idoseattachedviaaglycosidiclinkageatcarbon.
Figure9.Structuresofnaringenin(a)andnaringin(b).
Enteringtheflavonesynthesispathway,enzymechalconesynthase(CHS)catalyzes
consecutivecondensationsofthreeequivalentsofmalonylCoAfollowedbyaromatiza
tiontoconvertstartingpcoumaroylCoAtochalcone[25].Chalconeisomerase(CHI)per
formsstereospecificisomerizationoftetrahydroxychalconeto(2S)flavanone,whichisthe
branchpointprecursorofmanyimportantdownstreamflavonoids,includingflavones
(Figure10).
Figure 9. Structures of naringenin (a) and naringin (b).
Antioxidants2021,10,3288of24
OH
O
S
CoA p-coumaroyl-CoA
OO
OHS
CoA
malonyl-CoA 2x
CHS
OOH
HO O
OH
naringenin
OOH
HO OH
OH
chalcone
CHI
Figure10.Biosyntheticpathwayofnaringenin.
3.2.Apigenin
Apigenin(4,5,7trihydroxyflavone),isanaturalproductbelongingtotheflavone
classthatistheaglyconeofseveralnaturallyoccurringglycosides.Itisayellowcrystalline
solidthathasbeenusedtodyewool.Theapproximately650knownflavonesarisefrom
flavanonesbyoxidativeprocessescatalyzedbyaflavanonesynthase(FNS)enzyme(Fig
ure11)[26].TwotypesofFNShavepreviouslybeendescribed;FNSI,asolubleenzyme
thatuses2oxogluturate,Fe2+andascorbateascofactorsandFNSII,amembranebound,
NADPHdependentcytochromep450monooxygenase[27].
Figure11.Biosyntheticpathwayofapigenin.
3.3.Kaempferol
Kaempferol(3,5,7trihydroxy2(4hydroxyphenyl)4Hchromen4one)isanatural
flavonolfoundincommonfoodsderivedfromplantsandfruits.Itisbiosynthesizedfrom
naringeninviaa2hydroxyintermediate(Figure12).
Figure12.Biosynthesisofkaempferol.
Figure 10. Biosynthetic pathway of naringenin.
Antioxidants 2021,10, 328 8 of 24
3.3. Apigenin
Apigenin (4
0
,5,7-trihydroxyflavone), is a natural product belonging to the flavone
class that is the aglycone of several naturally occurring glycosides. It is a yellow crystalline
solid that has been used to dye wool. The approximately 650 known flavones arise
from flavanones by oxidative processes catalyzed by a flavanone synthase (FNS) enzyme
(Figure 11)
[
26
]. Two types of FNS have previously been described; FNS I, a soluble enzyme
that uses 2-oxogluturate, Fe2+ and ascorbate as cofactors and FNS II, a membrane bound,
NADPH dependent cytochrome p450 monooxygenase [27].
Antioxidants2021,10,3288of24
OH
O
S
CoA p-coumaroyl-CoA
OO
OHS
CoA
malonyl-CoA 2x
CHS
OOH
HO O
OH
naringenin
OOH
HO OH
OH
chalcone
CHI
Figure10.Biosyntheticpathwayofnaringenin.
3.2.Apigenin
Apigenin(4,5,7trihydroxyflavone),isanaturalproductbelongingtotheflavone
classthatistheaglyconeofseveralnaturallyoccurringglycosides.Itisayellowcrystalline
solidthathasbeenusedtodyewool.Theapproximately650knownflavonesarisefrom
flavanonesbyoxidativeprocessescatalyzedbyaflavanonesynthase(FNS)enzyme(Fig
ure11)[26].TwotypesofFNShavepreviouslybeendescribed;FNSI,asolubleenzyme
thatuses2oxogluturate,Fe2+andascorbateascofactorsandFNSII,amembranebound,
NADPHdependentcytochromep450monooxygenase[27].
Figure11.Biosyntheticpathwayofapigenin.
3.3.Kaempferol
Kaempferol(3,5,7trihydroxy2(4hydroxyphenyl)4Hchromen4one)isanatural
flavonolfoundincommonfoodsderivedfromplantsandfruits.Itisbiosynthesizedfrom
naringeninviaa2hydroxyintermediate(Figure12).
Figure12.Biosynthesisofkaempferol.
Figure 11. Biosynthetic pathway of apigenin.
Antioxidants2021,10,3288of24
OH
O
S
CoA p-coumaroyl-CoA
OO
OHS
CoA
malonyl-CoA 2x
CHS
OOH
HO O
OH
naringenin
OOH
HO OH
OH
chalcone
CHI
Figure10.Biosyntheticpathwayofnaringenin.
3.2.Apigenin
Apigenin(4,5,7trihydroxyflavone),isanaturalproductbelongingtotheflavone
classthatistheaglyconeofseveralnaturallyoccurringglycosides.Itisayellowcrystalline
solidthathasbeenusedtodyewool.Theapproximately650knownflavonesarisefrom
flavanonesbyoxidativeprocessescatalyzedbyaflavanonesynthase(FNS)enzyme(Fig
ure11)[26].TwotypesofFNShavepreviouslybeendescribed;FNSI,asolubleenzyme
thatuses2oxogluturate,Fe2+andascorbateascofactorsandFNSII,amembranebound,
NADPHdependentcytochromep450monooxygenase[27].
Figure11.Biosyntheticpathwayofapigenin.
3.3.Kaempferol
Kaempferol(3,5,7trihydroxy2(4hydroxyphenyl)4Hchromen4one)isanatural
flavonolfoundincommonfoodsderivedfromplantsandfruits.Itisbiosynthesizedfrom
naringeninviaa2hydroxyintermediate(Figure12).
Figure12.Biosynthesisofkaempferol.
Figure 12. Biosynthesis of kaempferol.
3.4. Hesperidin
Hesperidin is a flavanone glycoside found in citrus isolated in 1828 by French chemist
Lebreton from the white inner layer of citrus peels (mesocarp, albedo) [
28
]. The structure
consists of a flavanone aglycone, hesperetin, similar to naringenin, which differs from it for
the different pattern of substitution on the B ring, which is functionalized with hydroxy
group on 3
0
carbon and methoxy group on 4
0
carbon, whereas the naringenin lacks the
methoxy group (Figure 13) [29].
Antioxidants2021,10,3289of24
3.4.Hesperidin
Hesperidinisaflavanoneglycosidefoundincitrusisolatedin1828byFrenchchem
istLebretonfromthewhiteinnerlayerofcitruspeels(mesocarp,albedo)[28].Thestruc
tureconsistsofaflavanoneaglycone,hesperetin,similartonaringenin,whichdiffersfrom
itforthedifferentpatternofsubstitutionontheBring,whichisfunctionalizedwithhy
droxygroupon3′carbonandmethoxygroupon4′carbon,whereasthenaringeninlacks
themethoxygroup(Figure13)[29].
Figure13.Structureofhesperidin.
3.5.EllagicAcid
Ellagicacidis2,3,7,8tetrahydroxy[l]benzopyrano[5,4,3cde][1]benzopyran5,10
dione[30].Itisformedbydimerizationofgallicacidbyoxidativecouplingwithintramo
lecularlactonizationofbothcarboxylicacidgroups;thus,itisadilactoneofthedimerof
gallicacid(Figure14).
OH
HO
HO
O
OH
gallic acid
O
OH
O
HO
OH
HO
HO
OH
OH
OH
hexahydroxydiphenic acid
O
O
OH
OH
O
O
HO
HO
ellagic acid
Figure14.Gallicandellagicacids.
3.6.Oleuropein
Oleuropeinisaglycosylatedsecoiridoidproducedbysecondarymetabolismof
plantsandispresentinallolivetissues.Thetermoleuropeinisderivedfromthebotanical
nameoftheolivetree,Oleaeuropaea.Oleuropeinisanesterofelenolicacidwith3,4dihy
droxyphenylethanol(hydroxytyrosol),whichislinkedtoaunitofglucosebyaβglyco
sidicbond(Figure15)[31].
O
O
O
O
HO
O
elenolic acid
OH
HO
HO
3,4-dihydroxyphenylet hanol
HO
HO O
O
O
H
OO
O
oleuropein
O
HO OH
OH
CH
2
OH
Figure15.Structureofoleuropein.
Figure 13. Structure of hesperidin.
Antioxidants 2021,10, 328 9 of 24
3.5. Ellagic Acid
Ellagic acid is 2,3,7,8-tetrahydroxy[l]benzo-pyrano-[5,4,3-cde] [
1
] benzopyran-5,10-
dione [
30
]. It is formed by dimerization of gallic acid by oxidative coupling with intramolec-
ular lactonization of both carboxylic acid groups; thus, it is a dilactone of the dimer of gallic
acid (Figure 14).
Antioxidants2021,10,3289of24
3.4.Hesperidin
Hesperidinisaflavanoneglycosidefoundincitrusisolatedin1828byFrenchchem
istLebretonfromthewhiteinnerlayerofcitruspeels(mesocarp,albedo)[28].Thestruc
tureconsistsofaflavanoneaglycone,hesperetin,similartonaringenin,whichdiffersfrom
itforthedifferentpatternofsubstitutionontheBring,whichisfunctionalizedwithhy
droxygroupon3′carbonandmethoxygroupon4′carbon,whereasthenaringeninlacks
themethoxygroup(Figure13)[29].
Figure13.Structureofhesperidin.
3.5.EllagicAcid
Ellagicacidis2,3,7,8tetrahydroxy[l]benzopyrano[5,4,3cde][1]benzopyran5,10
dione[30].Itisformedbydimerizationofgallicacidbyoxidativecouplingwithintramo
lecularlactonizationofbothcarboxylicacidgroups;thus,itisadilactoneofthedimerof
gallicacid(Figure14).
OH
HO
HO
O
OH
galli c acid
O
OH
O
HO
OH
HO
HO
OH
OH
OH
hexahydroxydipheni c acid
O
O
OH
OH
O
O
HO
HO
ellagi c acid
Figure14.Gallicandellagicacids.
3.6.Oleuropein
Oleuropeinisaglycosylatedsecoiridoidproducedbysecondarymetabolismof
plantsandispresentinallolivetissues.Thetermoleuropeinisderivedfromthebotanical
nameoftheolivetree,Oleaeuropaea.Oleuropeinisanesterofelenolicacidwith3,4dihy
droxyphenylethanol(hydroxytyrosol),whichislinkedtoaunitofglucosebyaβglyco
sidicbond(Figure15)[31].
O
O
O
O
HO
O
elenoli c acid
OH
HO
HO
3,4-dihydroxyphenylet hanol
HO
HO O
O
O
H
OO
O
oleuropein
O
HO OH
OH
CH
2
OH
Figure15.Structureofoleuropein.
Figure 14. Gallic and ellagic acids.
3.6. Oleuropein
Oleuropein is a glycosylated secoiridoid produced by secondary metabolism of plants
and is present in all olive tissues. The term oleuropein is derived from the botanical
name of the olive tree, Olea europaea. Oleuropein is an ester of elenolic acid with 3,4-
dihydroxyphenylethanol (hydroxytyrosol), which is linked to a unit of glucose by a
β
-
glycosidic bond (Figure 15) [31].
Antioxidants2021,10,3289of24
3.4.Hesperidin
Hesperidinisaflavanoneglycosidefoundincitrusisolatedin1828byFrenchchem
istLebretonfromthewhiteinnerlayerofcitruspeels(mesocarp,albedo)[28].Thestruc
tureconsistsofaflavanoneaglycone,hesperetin,similartonaringenin,whichdiffersfrom
itforthedifferentpatternofsubstitutionontheBring,whichisfunctionalizedwithhy
droxygroupon3′carbonandmethoxygroupon4′carbon,whereasthenaringeninlacks
themethoxygroup(Figure13)[29].
Figure13.Structureofhesperidin.
3.5.EllagicAcid
Ellagicacidis2,3,7,8tetrahydroxy[l]benzopyrano[5,4,3cde][1]benzopyran5,10
dione[30].Itisformedbydimerizationofgallicacidbyoxidativecouplingwithintramo
lecularlactonizationofbothcarboxylicacidgroups;thus,itisadilactoneofthedimerof
gallicacid(Figure14).
OH
HO
HO
O
OH
galli c acid
O
OH
O
HO
OH
HO
HO
OH
OH
OH
hexahydroxydipheni c acid
O
O
OH
OH
O
O
HO
HO
ellagi c acid
Figure14.Gallicandellagicacids.
3.6.Oleuropein
Oleuropeinisaglycosylatedsecoiridoidproducedbysecondarymetabolismof
plantsandispresentinallolivetissues.Thetermoleuropeinisderivedfromthebotanical
nameoftheolivetree,Oleaeuropaea.Oleuropeinisanesterofelenolicacidwith3,4dihy
droxyphenylethanol(hydroxytyrosol),whichislinkedtoaunitofglucosebyaβglyco
sidicbond(Figure15)[31].
O
O
O
O
HO
O
elenoli c acid
OH
HO
HO
3,4-dihydroxyphenylet hanol
HO
HO O
O
O
H
OO
O
oleuropei n
O
HO OH
OH
CH
2
OH
Figure15.Structureofoleuropein.
Figure 15. Structure of oleuropein.
Secoiridoid conjugates that contain an esterified phenolic moiety, such as oleuropein,
result from a branching in the mevalonic acid pathway in which terpene synthesis (oleoside
moiety) and phenylpropanoid metabolism (phenolic moiety) merge [
32
]. This is illustrated
schematically in Figure 16.
Antioxidants2021,10,32810of24
Secoiridoidconjugatesthatcontainanesterifiedphenolicmoiety,suchasoleuropein,
resultfromabranchinginthemevalonicacidpathwayinwhichterpenesynthesis(oleo
sidemoiety)andphenylpropanoidmetabolism(phenolicmoiety)merge[32].Thisisil
lustratedschematicallyinFigure16.
Figure16.Asimplifiedschemeoftheoleuropeinbiosynthesis.
4.microRNAs
MicroRNAs(miRs)areauniqueclassofshortendogenousRNAs.Theyaresingle
strandednoncodingRNAsabletomodulategeneexpressionbybindingtothecomple
mentaryregionsof3UTRsequenceofspecificmRNAtargets.Withthisbiochemistry,
miRsallowsmRNAdegradationorinhibitstranslation.Thepeculiarregulatorycapability
makesthemcrucialfornormaldevelopmentinplantsandanimals[33].Intheareaof
molecularbiology,itisgenerallyacceptedthatmiRNAshaveevolvedindependentlyin
distinctlineages.However,recentstudiesonmiRsinnonbilaterianmetazoans,plants
andseveralalgaeraisethepossibilitythatthelastcommonancestorofbothlineagesmight
haveemployedamiRspathwayforposttranscriptionalregulation[33].Antioxidantand
microRNAsareanemergingfieldofresearch,especiallyinregardtopolyphenolsepige
neticability[34].Todate,searchingPubMedwiththewords“polyphenolsandmi
croRNAs”showsthat209papersarecurrentlypresent(accessedon25January2021at
7:00p.m.).Focusingthesearchto“NaringeninandmicroRNAs”,“Apigeninandmi
croRNAs”,“KaempferolandmicroRNAs”,“HesperidinandmicroRNAs”,“EllagicAcid
andmicroRNAs,and“OleuropeinandmicroRNA”producedsixpapersfornaringenin
andmicroRNAs,19onapigeninandmicroRNAs,22onkaempferolandmicroRNAs,six
onhesperidinandmicroRNAs,14onellagicacidandmicroRNAsandeightonoleuropein
andmicroRNAs.
Figure 16. A simplified scheme of the oleuropein biosynthesis.
Antioxidants 2021,10, 328 10 of 24
4. microRNAs
MicroRNAs (miRs) are a unique class of short endogenous RNAs. They are single-
stranded non-coding RNAs able to modulate gene expression by binding to the comple-
mentary regions of 3
0
UTR sequence of specific mRNA targets. With this biochemistry,
miRs allows mRNA degradation or inhibits translation. The peculiar regulatory capability
makes them crucial for normal development in plants and animals [
33
]. In the area of
molecular biology, it is generally accepted that miRNAs have evolved independently in
distinct lineages. However, recent studies on miRs in non-bilaterian metazoans, plants
and several algae raise the possibility that the last common ancestor of both lineages
might have employed a miRs pathway for post-transcriptional regulation [
33
]. Antioxidant
and microRNAs are an emerging field of research, especially in regard to polyphenols
epigenetic ability [
34
]. To date, searching PubMed with the words “polyphenols and
microRNAs” shows that 209 papers are currently present (accessed on 25 January 2021 at
7:00 p.m.). Focusing the search to “Naringenin and microRNAs”, “Apigenin and microR-
NAs”, “Kaempferol and microRNAs”, “Hesperidin and microRNAs”, “Ellagic Acid and
microRNAs”, and “Oleuropein and microRNA” produced six papers for naringenin and
microRNAs, 19 on apigenin and microRNAs, 22 on kaempferol and microRNAs, six on
hesperidin and microRNAs, 14 on ellagic acid and microRNAs and eight on oleuropein
and microRNAs.
4.1. Naringenin
The polyphenols naringenin was tested in diverse cell systems displaying epigenetic
property by regulating miRs, which in turn regulates the gene expression profile.
Naringenin treatment protects trophoblasts and endothelial cells from the harmful
high glucose environment by downregulating the miR-140-3p. In the interim, insulin
receptor alpha and insulin-like growth factor 1 receptor expression were upregulated
and the glucose uptake increased in naringenin treated trophoblasts and endothelial cells.
Therefore, naringenin was proposed by Zhao et al. as a treatment candidate, for gestational
diabetes [
35
]. Another evidence into the diabetes field by naringenin comes from its ca-
pability to ameliorated kidney injury through the let-7a. Yan et al. using mesangial cells
(MMCs) and diabetic rats as experimental models demonstrate that naringenin led to an
upregulation of let-7a under high glucose conditions affecting the expressions of fibronectin
and collagen VI in MMCs. In addition, let-7a upregulation in renal tissue diminished the
expression of transforming growth factor-
β
1 receptor 1 (TGFBR1), required for the regula-
tion of Let-7a/TGFBR1 signaling pathway in diabetic nephropathy [
36
]. Naringenin has
neuroprotective. In fact, in PC12 cells naringenin decreased the expression of miR-224-3p in
a dose-dependent manner and increased the expressions of SOD1 mRNA and protein [
37
].
Furthermore, in rats, after spinal cord injury, the protective effect of naringenin was exerted
through the repression of miR-223. This miR is a fine-tuner granulocyte production playing
a fundamental role on neutrophils activation of the inflammatory response [38].
In lung cancer, naringenin biochemical activity inhibits migration and invasion, as
well as tumor growth, through the regulation of the microRNA-3619-5p, which belong to
a regulating axis with circular RNA FOXM1 and sperm-associated antigen 5 [
39
]. Lastly,
Curti et al. recently demonstrate that naringenin is able to affect the miR-17-3p involved
in the control of antioxidant endogenous system [
40
]. Using a Caco cell, a well charac-
terized
in vitro
model which mimics the intestinal barrier, it is demonstrated that single
enantiomers of naringenin R and S have similar activity on this miR, while their equimolar
racemic mixture does not [41].
4.2. Apigenin
Apigenin in hepatocellular carcinoma cell line was noted to upregulate miR-520b
and miR-101 to inhibit tumor growth. The miRs miR-520b and miR-101 are involved
in Autophagy Related 7 protein (ATG7) and Nfr2 pathways, respectively, sensitize to
doxorubicin treatment [
42
,
43
]. In neuron derivative tumor, miR 16, miR-138 and miR-423
Antioxidants 2021,10, 328 11 of 24
were identified to be modulated by apigenin. In particular, miR-16 in glioma cells was
upregulated negatively influencing the cell B cell CLL/lymphoma 2 (BCL2)/NFkB/Matrix
Metallopeptidase 9 (MMP9) axis [
44
]. Similarly, in neuroblastoma cell, apigenin increased
the expression of miR-138, while in glioma, stem cells knockdown miR-423 enhances
sensitivity to apigenin involving the mitochondrial cell death pathways [45,46].
Apigenin could inhibit differentiation in TGF-
β
1-stimulated cardiac fibroblast, as well
the synthesis of extra cellular matrix (ECM) component. This mechanism might partly
be attributable to the reduction of miR-155-5p which control the c-Ski expression and the
axis in which is involved, which might result in the inhibition of small mother against
decapentaplegic (Smad)2/3 and p-Smad2/3 expressions [47].
Apigenin alleviates myocardial reperfusion injury (RI) in rats by downregulating
miR-15b. This miR was found to be increased during myocardial RI, determining a down-
regulation of JAK2 that promotes myocardial apoptosis and ROS production, aggravating
the myocardial injury. Apigenin treatment can downregulate miR-15b expression and
increase the expression of Janus kinase 2 (JAK2) and the activity of Signal transducer and
activator of transcription 3 (STAT3) pathway, reducing myocardial apoptosis and ROS
production and alleviating myocardial RI [
48
]. Lastly, in mice, apigenin improved glucose
intolerance by suppression of matured miR103 expression levels [
49
]. Apigenin normalized
let-7f expression in epididymal fat tissues preventing colonic inflammation, associated with
high fat diet-induced obesity [
50
]. Linked to lipids metabolism a liver-specific microRNA,
miR-122, is also an important factor for hepatitis C virus (HCV), replication. Apigenin,
in vitro
, significantly reduced mature miR122 expression levels in a dose-dependent man-
ner. Therefore, its intake, either via regular diet or supplements, may decrease HCV
replication in chronically infected patients [51].
4.3. Kaempferol
In human lung cancer cells, kaempferol was able to up-regulate miR-340, along
with the inactivation of the phosphatidylinositol-3-kinase (PI3K)/AKT pathways [
52
].
Likewise, in hepatocellular carcinoma cell line it remarkably reduces the expression of
miR-21 leading to the inactivation of the same PI3K/AKT pathway [
53
]. Kaempferol
is known to induce cardioprotective effects. In human aortic endothelial cells (HAECs),
kaempferol induced the upregulation of miR-26a-5p, which, targeting the toll-like receptor 4
(TLR4), was able to inactivate the TLR4/nuclear factor kappa B (NF-
κ
B) signaling pathway.
This biochemical mechanism improves the oxidized low-density lipoprotein-stimulated
HAECs [
54
]. In the same cell system, kaempferol increased miR-203 reversing the results
led by lipopolysaccharides (LPS)-induced inflammatory injury [
55
]. In vascular smooth
muscle cell (VSMC), kaempferol induces miR-21 expression inhibiting of cell migration [
56
].
The myocardial protective property of kaempferol was confirmed in ischemic heart disease
(IHD), using primary cardiomyocytes and myoblast cell line H9c2. Under oxygen-glucose
deprivation kaempferol exposure promoted the down-regulation of miR-15b, which target
Bcl-2 and TLR4 [
57
]. Furthermore, kaempferol enhanced miR-21 level in H9c2 cells exposed
to hypoxia/reoxygenation (H/R) and inhibition of miR-21 induced by transfection with
miR-21 inhibitor significantly blocked the protection of kaempferol against H/R-induced
H9c2 cell injury. Furthermore, kaempferol eliminated H/R-induced oxidative stress and
inflammatory response by the decreases in ROS generation and malondialdehyde content,
as well as the increase in antioxidant enzyme superoxide dismutase and glutathione
peroxidase activities [
58
]. Kaempferol exerts anti-inflammatory activities and has been
recognized as an effective agent for alleviating the clinical symptoms of osteoarthritis (OA)
by decreasing miR-146a in the chondrogenic cell line ATDC5 activated PI3K/AKT/mTOR
signaling pathway. In a rat model of OA, the expression of miR-146a in cartilage tissues
was repressed by kaempferol [59].
In osteoblast precursor cell line MC3T3-E1, kaempferol enhanced the expression level
of miR-101 promoting osteoblast proliferation, migration and differentiation [60].
Antioxidants 2021,10, 328 12 of 24
4.4. Hesperidin
Aberrant oxidative stress was implicated in the environmental contaminant Di-(2-
ethylhexyl) phthalate (DEHP)-induced testicular toxicity in which the miR-126-3p and the
miR-181a are overexpressed. Hesperidin administration normalized their levels beside to
other biochemical markers [
61
]. To the best of our knowledge, the present study demon-
strated for the first time that the administration of hesperidin decreased the expression
of ZEB2 by upregulating the expression of miR 132, which in turn promoted apopto-
sis and inhibited the proliferation of NSCLC cells [
62
]. Pre-treatment with hesperidin
(2
5, 50, 100 mg/kg
) for 7 days prevented these abnormalities induced by LPS injection.
In contrast, this effect of hesperidin was abolished by a miRNA-132 antagomir. Taken
together, these results suggest that the antidepressant-like mechanisms of hesperidin are at
least partially related to decreased pro-inflammatory cytokine levels via the miRNA-132
pathway in the brain [63].
4.5. Ellagic Acid
In high glucose-induced T2DM HepG2 cells, ellagic Acid (EA) was able to elevate
the miR-223 expression level, downregulating both, mRNA and protein levels of keap1.
This led to the upregulation of Nrf2, SOD1 and SOD2 protein levels. Therefore, EA
ameliorates oxidative stress and insulin resistance in the cell system used as model [
64
].
Another beneficial biochemical activity of EA was evident in ventricular remodeling after
myocardial infarction. EA improved ventricular remodeling by up-regulating miR-140-3p
expression and inhibiting MKK6 expression [65].
4.6. Oleuropein
In ovarian cancer was induced in xenograft mice model. Mice exposed to radiation
with the simultaneous administration of oleuropein. Oleuropein sensitized ovarian cells to
radiation altering the expression of miR-299. This miR was suppressed by a hypoxia in-
ducible factor and relieved in response to oleuropein, which in turn suppressed heparanase
1 expression and consequently led to increased sensitivity to radiation due to synergistic
effect of oleuropein with radiation [
66
]. Similarly, in nasopharyngeal carcinoma, oleuropein
strongly enhanced radio-sensitivity through the downregulation of miR-519d [67].
5. Pharmacokinetic Profile
Results from a growing number of studies unveiled polyphenols as promising thera-
peutic agents due to their broad spectrum of biological activities; however, the effectiveness
of these compounds in disease prevention and human health improvement is tightly related
and limited to their bioavailability [
68
]. The concept of bioavailability encompasses several
variables such as intestinal absorption, metabolism by gut microbiota, intestinal and liver
metabolism, biological properties of metabolites, distribution at tissues level and excretion
which in turn depend upon the chemical structure of xenobiotics. In addition, the various
chemical forms of polyphenols lead to high variability in their rate and extent of intestinal
absorption, as well as in the nature of circulating metabolites. Currently there is an increas-
ing interest in biological properties of apigenin owing to it proved relatively low toxicity
and effectiveness on cells with impaired biochemistry, such as cancer cells [
69
]. According
to biopharmaceutics classification system (BCS) that correlates
in vitro
dissolution with
in vivo
bioavailability, Apigenin is categorized as BCS class II drug due to its low solubility
and high permeability. Absorption of this compound occurs in the small intestine by both
passive and carrier-mediated saturable mechanism [
70
]; furthermore, the gastrointestinal
tract plays a crucial role in the metabolism and conjugation of apigenin before the entry
of the compound into the systemic circulation and the liver [
71
]. It is worth mentioning
that apigenin is naturally present in plants as glycosides. It has been hypothesized that
apigenin glucosides can be hydrolyzed into apigenin by cytosolic
β
-glucosidase (CBG) and
lactase-phlorizin hydrolase (LPH), which are enzymes produced by the liver, intestinal cells
or the gut microbiota [
70
,
72
,
73
]. LPH has been shown to hydrolyze flavonoid glycosides
Antioxidants 2021,10, 328 13 of 24
and the resulting aglycone may then enter epithelial cells by passive diffusion [
74
]. The
absorbed apigenin undergoes extensive Phase I and Phase II metabolism [75], accounting
for the low bioavailability of the compound.
5.1. Naringenin
The gut microbiota also plays a crucial role in the naringenin low availability as it
determines extensive pre-systemic metabolism of the compound leading to the formation
of degradation products such as phenolic acids [
76
]. As naringenin, hesperidin has limited
bioavailability due to the presence of the rutosin moiety. The removal of either rutinoside or
rhamnose from the molecule improves bioavailability and promotes a faster achievement
of the maximum plasma concentration [77].
5.2. Apigenin
In both rats and humans, apigenin has been documented to produce glucuronide,
sulfate conjugates or luteolin as major metabolites [
78
,
79
]. In addition, glucuronidation
reactions also occur in the intestine and intestinal disposition may be more important than
hepatic one in the first-pass metabolism of apigenin [
78
]. Of note, this natural flavone
modulates efflux proteins, especially P-glycoprotein (P-gp) and metabolic enzyme CYP3A4,
thus inducing clinically relevant drug-drug interactions by alteration of bioavailability
and distribution profiles of targeted drug such as TK inhibitors or paclitaxel [
72
]. The
elimination of Apigenin takes place through urine and feces and it is a slow process,
therefore an accumulation of the flavone in tissues seems possible [72].
5.3. Kaempeferol
Kaempferol shows up a more favorable bioavailability profile. Absorption of the
compound occurs in the small intestine through passive and facilitated diffusion or active
transport [
80
]. Following absorption phase kaempferol undergoes metabolic transforma-
tion in the glucuronides and sulfoconjugates forms at both liver [
81
] and small intestine
by enteric conjugation enzymes [
80
]. Kaempferol and its glycosides are also metabolized
in the colon by the bacterial microflora that releases the aglycones and broke aglycone C3
ring to form compounds (i.e., 4-methylphenol, phloroglucinol and 4-hydroxyphenylacetic
acid) that are either be absorbed and distributed to tissues by systemic circulation or be
excreted in feces and urine [
82
85
]. Of note, combination of kaempferol with quercetin
increase its bioavailability, thus improving its biological efficacy [86].
5.4. Hesperidin
Hesperidin bioavailability is also affected by various conditions including the health
status [
87
] and the concomitant administration of this compound with other flavonoids
such as quercetin, rutin, daidzein and chrysin [
88
]. Hesperidin requires deglycosylation
into hesperitin by gut microflora to be absorbed. The absorption phase occurs at the
level of colonocytes by proton-coupled active transport and transcellular passive diffu-
sion [
89
,
90
]. Hesperitin is then selectively metabolized into the liver to eriodictyol by both
the cytochrome P450 isoforms CYP1A and CYP1B1. Afterward, eriodictyol undergoes
methylation and it is transformed to homoeriodictyol (3
0
-O-methylated) or hesperitin
4
0
-O-methylated. The other hesperitin metabolites comprise hesperitin glucuronides (7-O-
glucuronide and 3
0
-O-glucuronide, hesperitin sulfates, 7-O- and 3
0
-O-sulfate, hesperitin
sulfoglucuronides and homoeriodictyol glucuronides. It is worth mentioning that the
first-pass metabolism of hesperitin occurs in intestinal cells leading to the formation of
hesperitin 7-O-glucuronide and 3
0
-O-glucuronide, which represents the major hesperitin
metabolites
in vivo
[
91
,
92
]. Hesperitin is also a selective inhibitor of cytochrome P450
CYP1B1 [
93
]. This observation provides a possible explanation of the compound anti-
tumor activity being the enzyme involved in facilitating carcinogenesis process. Moreover,
hesperitin enhances bioavailability of co-administered drugs such as diltiazem, verapamil
and vincristine through inhibition of CYP3A4 or P-gp efflux [
94
96
]. The metabolites of
Antioxidants 2021,10, 328 14 of 24
hesperidin/hesperitin are detected in urine but not in feces, thus suggesting a further
bacterial degradation to ring fission products and phenolic acids in the colon [97].
5.5. Ellagic Acid
The poor systemic bioavailability also affects the mechanism of action across condi-
tions and doses of ellagic acid (EA) as demonstrated by several
in vivo
studies [
98
]. In
fruits and nuts, EA exists in either its free form, as EA-glycosides, or bound as ellagitannin
(ET) [
99
,
100
]. However, only a small portion of free EA is absorbed in the stomach, since ET
are resistant to acidic pH. ET hydrolysis occurs in the small intestine, yielding to the release
of EA. This latter is absorbed mainly by passive diffusion, although the involvement of
a protein-mediated transport cannot be ruled out as suggested by
in vitro
experiment on
Caco-2 cells line model [
101
]. In the systemic circulation, EA undergoes a massive first pass
effect, being transformed in methyl esters, dimethyl esters or glucuronides, measurable
in human plasma from 1 to 5 h after ET ingestion [
102
]. In the meantime, unabsorbed ET
and EA fractions are mostly converted to a family of metabolites called urolithins by gut
microbiota. Urolithins contain a common lipophilic moiety, thus resulting in a net improve-
ment of bioavailability compared to EA [
103
]. However, the difference in gut microbiota
composition leads to a wide variability in microbial metabolism of EA among individuals.
Indeed, humans may produce no urolithins, highly active urolithins or less active urolithin,
hence EA consumption may not exert equal health benefits in all subjects [
104
106
]. The
low EA oral bioavailability was also confirmed in human pharmacokinetic studies demon-
strating the rapid metabolism of the compound and the existence in the absorption phase
of saturable mechanism [107].
5.6. Oleuropein
Poor data and often conflicting results exist on the oleuropein pharmacokinetic from
EVOO or olive leaves in humans [
108
111
]. This discrepancy may be due to several factors.
Indeed, the route of administration, genotype, age, sex, interaction with food and the
different extraction processes deeply affect oleuropein bioavailability [
112
]. It has been
reported that oral oleuropein ingestion is resistant to the stomach acidic pH and it is quickly
absorbed in the small intestine, reaching a maximum plasma concentration earlier than
conjugated metabolites of hydroxytyrosol in humans. Of note these latter represented the
major fraction of the oleuropein phenolic metabolites in plasma and urine after intake [
113
],
suggesting potential complete metabolization of oleuropein to hydroxytyrosol and other
catabolic products. Attention has also been payed to the gut microbiota.
In vitro
and
in vivo
approaches demonstrated that oleuropein was rapidly deglycosylated to oleuropeinA
by human fecal microbiota and then metabolized into elenolic acid and hydroxytyrosol
by microbial esterase activity [
114
]. Further studies have shown that the conversion of
oleuropein into hydroxytyrosol was performed by acid by Lactobacillus plantarum [
115
]
and based on this observation oral granules for the co-delivery of Lactobacillus plantarum
and a standardized olive leaf extract were developed in order to promote oleuropein
metabolism and ensure high levels of hydroxytyrosol [116].
6. Health Effects
6.1. Naringenin
Being present in foods known to be beneficial for health, such as citrus fruits and
tomatoes, naringenin boasts numerous studies, even if, as happens for all molecules of
plant origin, mainly
in vivo
and
in vitro
. In this sense, various studies show a favorable
action in oncology, as it acts by limiting the progression of the cell cycle and angiogenesis;
favoring apoptosis and acting directly on some carcinogens [
117
119
]. It also shows
interesting qualities in the management of type 2 diabetes, obesity and metabolic syndrome;
improving insulin sensitivity probably by regulating the action of AMPK, regulating
the action of amylases and modulating inflammation via inhibition of NF-
κ
B; beneficial
actions are reported on the prevention of liver diseases in particular with an interesting
Antioxidants 2021,10, 328 15 of 24
mechanism, according to which naringenin should be incorporated in cell membranes thus
providing a greater protective action [
120
,
121
]. Preventive action on neurodegenerative
diseases can also be explained with the mechanisms proposed above [
121
]. Possessing
immunomodulatory and antiviral activity, naringenin has also been proposed to support
therapies against COVID-19 [
122
]. As for almost all polyphenols, the main problem is
bioavailability, although many studies have used orange juice and not extracts or the
molecule as it is; for this reason, different strategies are evaluated to make the molecule
more bioavailable, as the doses that should be drawn from
in vitro
or
in vivo
studies
should be between 25 and 50 mg per kg of body weight [
123
,
124
], even if some studies
seem to confirm an absorption or at least a retention by the microbiota, with consequent
beneficial action on it, with doses ranging from 200 to 500 mg [
125
127
]. In a case report
by Murugesan et al., an improvement in insulin sensitivity is reported with an orange
juice dosage of 150 mg naringenin for eight weeks [
128
]. In one of the few human studies
48 postmenopausal women took 210 mg of naringenin from grapefruit juice for 6 months,
showing a clear benefit on arterial stiffness [129].
6.2. Apigenin
Like many substances of plant origin, apigenin has also been used for a long time
through sources that are part of traditional medicines or common uses, remembering, in
this regard, honey and chamomile [
130
,
131
]. There are many studies in which apigenin
shows a very promising potential as an antioxidant [
132
] and as an adjuvant for numerous
pathological states, such as diabetes, cancer, depression, amnesia and Alzheimer’s [
131
];
on the other hand, the studies on humans are few and the compound is extracted mainly
from chamomile. Zick et al. tested a standardized chamomile extract that provided
15 mg of apigenin on 34 patients in double blind versus placebo, evaluating the quality of
sleep and therefore the impact on insomnia, showing moderate positive effects: different
quantities should probably be tested, as there are no adverse effects [
133
]. In two other
papers
[134,135]
, the effect of apigenin, still supplied as chamomile extract, on anxiety dis-
orders was evaluated: the first followed a protocol with a variable dose from
8 to 13 mg
of
apigenin; in the second, a constant dose of 18mg of active ingredients, in both cases, showed
positive effects, justifying their use in support of any drug therapy. A very interesting
hypothesis comes from the work of Vollmer et al., from which it can be deduced that part
of the apigenin is excreted intact with the feces; therefore, it is able to reach the microbiota
present in the large intestine and thus positively influence the peculiar function [136].
6.3. Kaempeferol
Kaempferol is a polyphenol widespread in food, present in crucifers, but also in other
foods used quite commonly in various food cultures. For this reason, there are also evalua-
tions of epidemiological studies which positively correlate its intake with the prevention
and treatment of various diseases [
137
]. A review Kashyap et al. [
138
] shows conflicting
results from epidemiological studies: multiple positive results are highlighted, especially
in the chemopreventive action, but not all studies report the same effects [
139
]. Human
studies are not numerous, despite the promising results
in vitro
and
in vivo [140143]
;
epidemiological studies show a protective effect from cardiovascular diseases and a de-
crease in IL6 (with consequent modulation of inflammation) from a dosage of kaempferol
included in a range of 2–12 mg per day, combined; however, with other polyphenols [
139
];
moreover, from an interesting study a protective effect on osteoporysis would result, ac-
cording to various mechanisms that would lead to a decrease in adipogenesis, an increase
in chondrogenesis and osteoblastogenesis [144].
6.4. Hesperidin
Hesperidin is the polyphenol most present in oranges and other citrus fruits, whose
beneficial effect is well known, but often related to vitamin C and not to polyphenols. The
beneficial effects demonstrated
in vivo
and
in vitro
are multiple, as for all polyphenols,
Antioxidants 2021,10, 328 16 of 24
thanks to their chemical structure, the antioxidant action is well present. In addition to
this, a neuroprotective action was highlighted, beneficial on the cardiovascular system,
bone health, on glycemic regulation and not least on the microbiota [
145
147
]; the dosages
of hesperidin used in human studies vary from a minimum of 200 mg to a maximum
of 500 mg, remaining, although largely in non-toxic or harmful dosages; some show
improved cardiovascular health (64 subjects for 6 weeks 500 mg/day); however, positive
effects are also shown in the management of hemorrhoids (70 subjects for 6 months of use),
in neuroprotection (more than one study with dosages ranging from 200 to 500 mg/day),
in the prevention and treatment of osteoporosis [
148
152
] and even in sports: 500 mg
of hesperidin seems to improve aerobic performance in cyclists, probably thanks to the
antioxidant action.
6.5. Ellagic Acid
This compound is very often linked to pomegranate; in fact, many papers refer to the
use of the juice of the fruit, which, however, contains various polyphenols. In fact, in some
cases the juice has proved more effective than the extract itself [
153
,
154
]. Common with
all polyphenols, ellagic acid also has an antioxidant activity, directly and as a regulator of
NF-
κ
B [
155
,
156
]. In addition,
in vitro
and
in vivo
, it has also shown a regulatory action on
inflammatory prostaglandins and on the onset of atherogenesis; many studies, however,
are performed
in vitro
or
in vivo
. As for humans, a point to be clarified is the bioavailability
and conversion of ellagic acid into urolithin, with relative efficacy of the latter [
157
,
158
]. It
is interesting to note how the suboptimal absorption at the gastrointestinal level makes
it available for the microbiota; from some papers, it seems that this favors some bacterial
strains favorable for health, such as the Firmicutes: Bacteroidetes ratio [
159
]. The last study
involved 20 subjects by administering 1000 mg of ellagic acid for four weeks. Another area
where ellagic acid seems to have a positive effect is that relating to the central nervous
system [
160
]; Ying et al. [
161
], administered to a group of 150 obese subjects, 50 mg of
ellagic acid for 12 weeks, a positive effect was recorded not only on the lipid profile, but
also on the BNDF, with improvement of the cognitive capacity of the subjects. Amma
et al. [
162
], evaluated the action of ellagic acid in promoting recovery after an intense
weightlifting workout; the study, carried out on nine subjects, showed a modulating effect
on all the parameters relating to the antioxidant state, although it should be emphasized,
however, that pomegranate juice was provided, and not ellagic acid alone. In the review of
Huang et al., no positive effects of glycemic control are evident.
6.6. Oleuropein
Oleuropein and its derivatives should represent the center of the Mediterranean diet,
as a characteristic of the olive tree and consequently of the olive oil; the benefits, proven
in vivo
and
in vitro
are multiple and attributable to various pathways, to mention the
antiatherogenic, cardioprotective, anticancer, neuroprotective, antidiabetic, anti-obesity,
regulating lipids, antimicrobial and antiviral effects [
163
], but also others less frequently
combined with polyphenols, such as the positive action on osteoarthritis [
164
] or in-
flammatory bowel diseases [
165
] and immunomodulation in general [
166
]; Somerville
et al. [
167
], tested a supplement containing 100 mg of oleuropein on 32 high school athletes
of good level, assessing their tendency to get sick and reported a positive correlation;
De Bock et al. [168]
demonstrated an improvement in insulin sensitivity by administer-
ing a supplement containing about 52 mg of oleuropein and 10mg of hydroxytyrosol to
46 obese subjects, in crossover versus placebo; with the same dosage, Lockyer et al. [
169
],
on 16 subjects, analyzed the action of inflammatory cytokines and cardiovascular health,
a modulating action was found in particular on IL8; overall, dosages of oleuropein from
5
0 to 150 mg
per day and/or hydroxytorosol between 10 and 50 mg seem to be safe and
effective to ensure an anti-inflammatory and antioxidant effect, which is beneficial in im-
proving various conditions, such as atherosclerosis. Similar to other polyphenols [
170
,
171
].
Antioxidants 2021,10, 328 17 of 24
7. Conclusions
Polyphenols certainly show great potential in assisting nutritionist or physician to co-
treat various pathologies with a marked inflammatory component. Indeed, the numerous
studies on the Mediterranean diet show how this eating style disfavors the onset of diseases
that are the major cause of death worldwide, such as cardiovascular diseases, diabetes and
cancer. The contemporary diet, in particular in the West, with the exception of north Europe,
is often lacking of fruit and vegetables, the main sources of polyphenols; therefore, the
consumption of these food categories should be strongly encouraged. On the other hand,
the use of polyphenols as supplements could also have a strong impact on health. Although
there are many
in vitro
and
in vivo
studies, those performed in humans are few and often
not organized in a systematic way. Often the vegetable extract in toto or the vegetable is
used, so it is difficult to understand which of the compounds had the predominant effect,
also because they often act in synergy. In the current state of knowledge, we strongly
recommend the use of fruit and vegetables in the diet and to consider the intake ranging
from 50 to 200 mg/day of polyphenols, an amount that can also be doubled in pathological
conditions, to ensure a beneficial action, at least from an anti-inflammatory and antioxidant
viewpoint. As shown, polyphenols have an epigenetic action in particular on miRNAs,
and this could also be a fascinating field of study to evaluate, in a clear and systematic way,
even in clinical trials, the action of polyphenols.
Author Contributions:
Conceptualization, R.C. and E.C., C.L.T. wrote the dietary sources section; A.F.
wrote the chemistry section; E.C. wrote the microRNAs section; M.C.C. wrote the pharmacokinetic
profile section; R.C wrote the health effects section; R.C. and E.C wrote the conclusion; M.C.C.
coordinated the study. All authors have read and agreed to the published version of the manuscript.
Funding: This research received no external funding.
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Martínez-Huélamo, M.; Rodríguez-Morató, J.; Boronat, A.; de la Torre, R. Modulation of Nrf2 by Olive Oil and Wine Polyphenols
and Neuroprotection. Antioxidants 2017,26, 73. [CrossRef]
2.
Georgoulis, M.; Kontogianni, M.D.; Yiannakouris, N. Mediterranean diet and diabetes: Prevention and treatment. Nutrients
2014
,
6, 1406–1423. [CrossRef]
3.
Widmer, R.J.; Flammer, A.J.; Lerman, L.O.; Lerman, A. The Mediterranean diet, its components, and cardiovascular disease. Am.
J. Med. Sci. 2015,128, 229–238. [CrossRef]
4.
Mentella, M.C.; Scaldaferri, F.; Ricci, C.; Gasbarrini, A.; Miggiano, G.A.D. Cancer and Mediterranean Diet: A Review. Nutrients
2019,11, 2059. [CrossRef]
5.
Zamora-Ros, R.; Knaze, V.; Rothwell, J.A.; Hémon, B.; Moskal, A.; Overvad, K.; Tjønneland, A.; Kyrø, C.; Fagherazzi, G.;
Boutron-Ruault, M.C.; et al. Dietary polyphenol intake in Europe: The European Prospective Investigation into Cancer and
Nutrition (EPIC) study. Eur J. Nutr. 2016,55, 1359. [CrossRef]
6.
Gollucke, A.P.; Peres, R.C.; Odair, A., Jr.; Ribeiro, D.A. Polyphenols: A nutraceutical approach against diseases. Recent Pat. Food
Nutr. Agric. 2013,5, 214–219.
7.
Oliviero, F.; Scanu, A.; Zamudio-Cuevas, Y.; Punzi, L.; Spinella, P. Anti-inflammatory effects of polyphenols in arthritis. J. Sci.
Food Agric. 2018,98, 1653–1659. [CrossRef] [PubMed]
8.
Leiherer, A.; Mündlein, A.; Drexel, H. Phytochemicals and Their Impact on Adipose Tissue Inflammation and Diabetes. Vasc.
Pharmac. 2013,58, 3–20. [CrossRef]
9.
Domitrovic, R. The Molecular Basis for the Pharmacological Activity of Anthocyans. Curr. Med. Chem.
2011
,18, 4454–4469.
[CrossRef] [PubMed]
10.
González, R.; Ballester, I.; López-Posadas, R.; Suárez, M.D.; Zarzuelo, A.; Martínez-Augustin, O.; Sánchez de Medina, F. Effects of
Flavonoids and Other Polyphenols on Inflammation. Crit. Rev. Food Sci. Nutr. 2011,51, 331–362. [CrossRef] [PubMed]
11.
Carullo, G.; Perri, M.; Manetti, F.; Aiello, F.; Caroleo, M.C.; Cione, E. Quercetin-3-oleoyl derivatives as new gpr40 agonists:
Molecular docking studies and functional evaluation. Bioorganic Med. Chem. Lett. 2019,29, 1761–1764. [CrossRef]
12.
Dinu, M.; Pagliai, G.; Casini, A.; Sofi, F. Mediterranean diet and multiple health outcomes: An umbrella review of meta-analyses
of observational studies and randomised trials. Eur. J. Clin. Nutr. 2018,72, 30–43. [CrossRef]
13.
Alfa, H.H.; Arroo, R.R.J. Over 3 decades of research on dietary flavonoid antioxidants and cancer prevention: What have we
achieved? Phytochem. Rev. 2019,18, 989–1004. [CrossRef]
Antioxidants 2021,10, 328 18 of 24
14.
Zhou, Y.; Jiang, Z.; Lu, H.; Xu, Z.; Tong, R.; Shi, J.; Jia, G. Recent Advances of Natural Polyphenols Activators for Keap1-Nrf2
Signaling Pathway. Chem. Biodivers. 2019,16, e1900400. [CrossRef]
15.
Salehi, B.; Valere, P.; Fokou, T.; Sharifi-Rad, M.; Zucca, P.; Pezzani, R.; Martins, N.; Sharifi-Rad, J. The Therapeutic Potential of
Naringenin: A Review of Clinical Trials. Pharmaceuticals 2019,12, 11. [CrossRef]
16. Sibel, K.; Sedef, N.E.L. Quercetin, luteolin, apigenin and kaempferol contents of some foods. Food Chem. 1999,66, 289–292.
17.
McKay, D.L.; Blumberg, J.B. A review of the bioactivity and potential health benefits of chamomile tea (Matricaria recutita L.).
Phytother. Res. 2006,20, 519–530. [CrossRef]
18. Shukla, S.; Gupta, S. Apigenin: A promising molecule for cancer prevention. Pharm. Res. 2010,27, 962–978. [CrossRef]
19.
Benincasa, C.; La Torre, C.; Plastina, P.; Fazio, A.; Perri, E.; Caroleo, M.C.; Gallelli, L.; Cannataro, R.; Cione, E. Hydroxytyrosyl
Oleate: Improved Extraction Procedure from Olive Oil and By-Products, and In Vitro Antioxidant and Skin Regenerative
Properties. Antioxidants 2019,8, 233. [CrossRef]
20.
Chen, H.; Zuo, Y.; Deng, Y. Separation and determination of flavonoids and other phenolic compounds in cranberry juice by
high-performance liquid chromatography. J. Chromatogr. A 2001,13, 387–395. [CrossRef]
21.
Barbaro, B.; Toietta, G.; Maggio, R.; Arciello, M.; Tarocchi, M.; Galli, A.; Balsano, C. Effects of the Olive-Derived Polyphenol
Oleuropein on Human Health. Int. J. Mol. Sci. 2014,15, 18508–18524. [CrossRef]
22. Vogt, T. Phenylpropanoid Biosynthesis. Mol. Plant. 2010,3, 2–20. [CrossRef]
23.
Santos-Sánchez, N.C.; Salas-Coronado, R.; Hernández-Carlos, B.; Villanueva-Cañongo, C. Shikimic Acid Pathway in Biosynthesis
of Phenolic Compounds. In Plant Physiological Aspects of Phenolic Compounds, 1st ed.; Soto-Hernández, M., García-Mateos, R.,
Palma-Tenango, M., Eds.; Huajuapan de León: Oaxaca, Mexico, 2019; p. 1.15.
24.
Khoddami, A.; Wilkes, M.A.; Roberts, T.H. Techniques for Analysis of Plant Phenolic Compounds. Molecules
2013
,18, 2328–2375.
[CrossRef]
25.
Austin, M.B.; Noel, J.P. The chalcone synthase superfamily of type III polyketide synthases. Nat. Prod. Rep.
2003
,20, 79–110.
[CrossRef] [PubMed]
26.
Martens, S.; Forkmann, G.; Matern, U.; Lukacin, R. Cloning of parsley flavone synthase, I. Phytochemistry
2001
,58, 43–46.
[CrossRef]
27.
Leonard, E.; Yan, Y.; Lim, K.H.; Koffas, M.A. Investigation of two distinct flavone synthases for plant-specific flavone biosynthesis
in Saccharomyces cerevisiae. Appl. Environ. Microbiol. 2005,71, 8241–8248. [CrossRef]
28.
Garg, A.; Garg, S.; Zaneveld, L.J.D.; Singla, A.K. Chemistry and pharmacology of the citrus bioflavonoid hesperidin. Phytother.
Res. 2001,15, 655–669. [CrossRef] [PubMed]
29. Binkowska, I. Hesperidin: Synthesis and characterization of bioflavonoid complex. Sn. Appl. Sci. 2020,2, 445. [CrossRef]
30.
Girish, C.; Pradhan, S.C. Herbal Drugs on the Liver. In Liver Pathophysiology, 1st ed.; Muriel, P., Ed.; Academic Press: Cambridge,
MA, USA, 2017; pp. 605–620.
31.
El Riachy, M.; Priego-Capote, F.; Rallo, L.L.L.; Luque de Castro, M.D. Hydrophilic antioxidants of virgin olive oil. Part 2:
Biosynthesis and biotransformation of phenolic compounds in virgin olive oil as affected by agronomic and processing factors.
Eur. J. Lipid Sci. Technol. 2011,113, 692–707. [CrossRef]
32.
Koudounas, K.; Banilas, G.; Michaelidis, C.; Demoliou, C. Stamatis Rigas and Polydefkis Hatzopoulos. A defence-related Olea
europaea
β
-glucosidase hydrolyses and activates oleuropein into a potent protein cross-linking agent. J. Exp. Bot.
2015
,66,
2093–2106. [CrossRef]
33.
Moran, Y.; Agron, M.; Praher, D.; Technau, U. The evolutionary origin of plant and animal microRNAs. Nat. Ecol. Evol.
2017
,1, 27.
[CrossRef]
34.
Corrêa, T.A.; Rogero, M.M. Polyphenols regulating microRNAs and inflammation biomarkers in obesity. Nutrition
2019
,59,
150–157. [CrossRef]
35. Zhao, C.; Zhao, C.; Zhao, H. Defective insulin receptor signaling in patients with gestational diabetes is related to dysregulated
miR-140 which can be improved by naringenin. Int. J. Biochem. Cell Biol. 2020,128, 105824. [CrossRef]
36.
Yan, N.; Wen, L.; Peng, R.; Li, H.; Liu, H.; Peng, H.; Sun, Y.; Wu, T.; Chen, L.; Duan, Q.; et al. Naringenin Ameliorated Kidney
Injury through Let-7a/TGFBR1 Signaling in Diabetic Nephropathy. J. Diabetes Res. 2016,2016, 8738760. [CrossRef]
37.
Li, H.; Liu, M.W.; Yang, W.; Wan, L.J.; Yan, H.L.; Li, J.C.; Tang, S.Y.; Wang, Y.Q. Naringenin induces neuroprotection against
homocysteine-induced PC12 cells via the upregulation of superoxide dismutase 1 expression by decreasing miR-224-3p expression.
J. Biol. Regul. Homeost. Agents 2020,34, 421–433.
38.
Shi, L.B.; Tang, P.F.; Zhang, W.; Zhao, Y.P.; Zhang, L.C.; Zhang, H. Naringenin inhibits spinal cord injury-induced activation of
neutrophils through miR-223. Gene 2016,592, 128–133. [CrossRef]
39.
Tan, Z.; Sun, Y.; Liu, M.; Xia, L.; Cao, F.; Qi, Y.; Song, Y. Naringenin Inhibits Cell Migration, Invasion, and Tumor Growth by
Regulating circFOXM1/miR-3619-5p/SPAG5 Axis in Lung Cancer. Cancer Biother. Radiopharm. 2020,00, 1–13. [CrossRef]
40.
Cannataro, R.; Caroleo, M.C.; Fazio, A.; La Torre, C.; Plastina, P.; Gallelli, L.; Lauria, G.; Cione, E. Ketogenic Diet and microRNAs
Linked to Antioxidant Biochemical Homeostasis. Antioxidants 2019,8, 269. [CrossRef]
41.
Curti, V.; Di Lorenzo, A.; Rossi, D.; Martino, E.; Capelli, E.; Collina, S.; Daglia, M. Enantioselective Modulatory Effects of
Naringenin Enantiomers on the Expression Levels of miR-17-3p Involved in Endogenous Antioxidant Defenses. Nutrients
2017
,
9, 215. [CrossRef] [PubMed]
Antioxidants 2021,10, 328 19 of 24
42.
Gao, A.M.; Zhang, X.Y.; Hu, J.N.; Ke, Z.P. Apigenin sensitizes hepatocellular carcinoma cells to doxorubic through regulating
miR-520b/ATG7 axis. Chem. Biol. Interact. 2018,280, 45–50. [CrossRef] [PubMed]
43.
Gao, A.M.; Zhang, X.Y.; Ke, Z.P. Apigenin sensitizes BEL-7402/ADM cells to doxorubicin through inhibiting miR-101/Nrf2
pathway. Oncotarget 2017,8, 82085–82091. [CrossRef]
44.
Chen, X.J.; Wu, M.Y.; Li, D.H.; You, J. Apigenin inhibits glioma cell growth through promoting microRNA-16 and suppression of
BCL-2 and nuclear factor-κB/MMP 9. Mol. Med. Rep. 2016,14, 2352–2358. [CrossRef] [PubMed]
45.
Chakrabarti, M.; Banik, N.L.; Ray, S.K. miR-138 overexpression is more powerful than hTERT knockdown to potentiate apigenin
for apoptosis in neuroblastoma in vitro and in vivo. Exp. Cell Res. 2013,319, 1575–1585. [CrossRef]
46.
Wan, Y.; Fei, X.; Wang, Z.; Jiang, D.; Chen, H.; Wang, M.; Zhou, S. miR-423-5p knockdown enhances the sensitivity of glioma stem
cells to apigenin through the mitochondrial pathway. Tumour. Biol. 2017,39, 1010428317695526. [CrossRef]
47.
Wang, F.; Fan, K.; Zhao, Y.; Xie, M.L. Apigenin attenuates TGF-
β
1-stimulated cardiac fibroblast differentiation and extracellular
matrix production by targeting miR-155-5p/c-Ski/Smad pathway. J. Ethnopharmacol. 2021,265, 113195. [CrossRef]
48.
Wang, P.; Sun, J.; Lv, S.; Xie, T.; Wang, X. Apigenin Alleviates Myocardial Reperfusion Injury in Rats by Downregulating miR-15b.
Med. Sci. Monit. 2019,25, 2764–2776. [CrossRef]
49.
Ohno, M.; Shibata, C.; Kishikawa, T.; Yoshikawa, T.; Takata, A.; Kojima, K.; Akanuma, M.; Kang, Y.J.; Yoshida, H.; Otsuka, M.;
et al. The flavonoid apigenin improves glucose tolerance through inhibition of microRNA maturation in miRNA103 transgenic
mice. Sci. Rep. 2013,3, 2553. [CrossRef] [PubMed]
50.
Gentile, D.; Fornai, M.; Colucci, R.; Pellegrini, C.; Tirotta, E.; Benvenuti, L.; Segnani, C.; Ippolito, C.; Duranti, E.; Virdis, A.; et al.
The flavonoid compound apigenin prevents colonic inflammation and motor dysfunctions associated with high fat diet-induced
obesity. PLoS ONE 2018,13, e0195502. [CrossRef] [PubMed]
51.
Shibata, C.; Ohno, M.; Otsuka, M.; Kishikawa, T.; Goto, K.; Muroyama, R.; Kato, N.; Yoshikawa, T.; Takata, A.; Koike, K. The
flavonoid apigenin inhibits hepatitis C virus replication by decreasing mature microRNA122 levels. Virology
2014
,462–463, 42–48.
[CrossRef] [PubMed]
52.
Han, X.; Liu, C.F.; Gao, N.; Zhao, J.; Xu, J. Kaempferol suppresses proliferation but increases apoptosis and autophagy by
up-regulating microRNA-340 in human lung cancer cells. Biomed. Pharm. 2018,108, 809–816. [CrossRef]
53.
Zhu, G.; Liu, X.; Li, H.; Yan, Y.; Hong, X.; Lin, Z. Kaempferol inhibits proliferation, migration, and invasion of liver cancer HepG2
cells by down-regulation of microRNA-21. Int. J. Immunopathol. Pharm. 2018,32, 2058738418814341. [CrossRef]
54.
Zhong, X.; Zhang, L.; Li, Y.; Li, P.; Li, J.; Cheng, G. Kaempferol alleviates ox-LDL-induced apoptosis by up-regulation of
miR-26a-5p via inhibiting TLR4/NF-
κ
B pathway in human endothelial cells. Biomed. Pharm.
2018
,108, 1783–1789. [CrossRef]
[PubMed]
55.
Cui, S.; Tang, J.; Wang, S.; Li, L. Kaempferol protects lipopolysaccharide-induced inflammatory injury in human aortic endothelial
cells (HAECs) by regulation of miR-203. Biomed. Pharm. 2019,115, 108888. [CrossRef] [PubMed]
56.
Kim, K.; Kim, S.; Moh, S.H.; Kang, H. Kaempferol inhibits vascular smooth muscle cell migration by modulating BMP-mediated
miR-21 expression. Mol. Cell Biochem. 2015,407, 143–149. [CrossRef]
57.
Li, L.; Shao, Y.; Zheng, H.; Niu, H. Kaempferol Regulates miR-15b/Bcl-2/TLR4 to Alleviate OGD-Induced Injury in H9c2 Cells.
Int. Heart J. 2020,61, 585–594. [CrossRef]
58.
Huang, J.; Qi, Z. miR-21 mediates the protection of kaempferol against hypoxia/reoxygenation-induced cardiomyocyte injury via
promoting Notch1/PTEN/AKT signaling pathway. PLoS ONE 2020,15, e0241007. [CrossRef]
59.
Jiang, R.; Hao, P.; Yu, G.; Liu, C.; Yu, C.; Huang, Y.; Wang, Y. Kaempferol protects chondrogenic ATDC5 cells against inflammatory
injury triggered by lipopolysaccharide through down-regulating miR-146a. Int. Immunopharmacol.
2019
,69, 373–381. [CrossRef]
60.
Wang, Y.; Chen, H.; Zhang, H. Kaempferol promotes proliferation, migration and differentiation of MC3T3-E1 cells via up-
regulation of microRNA-101. Artif. Cells Nanomed. Biotechnol. 2019,47, 1050–1056. [CrossRef]
61.
Helmy, H.S.; Senousy, M.A.; El-Sahar, A.E.; Sayed, R.H.; Saad, M.A.; Elbaz, E.M. Aberrations of miR-126-3p, miR-181a and sirtuin1
network mediate Di-(2-ethylhexyl) phthalate-induced testicular damage in rats: The protective role of hesperidin. Toxicology
2020
,
433-434, 152406. [CrossRef]
62.
Tan, S.; Dai, L.; Tan, P.; Liu, W.; Mu, Y.; Wang, J.; Huang, X.; Hou, A. Hesperidin administration suppresses the proliferation
of lung cancer cells by promoting apoptosis via targeting the miR-132/ZEB2 signalling pathway. Int. J. Mol. Med.
2020
,46,
2069–2077. [CrossRef] [PubMed]
63.
Li, M.; Shao, H.; Zhang, X.; Qin, B. Hesperidin Alleviates Lipopolysaccharide-Induced Neuroinflammation in Mice by Promoting
the miRNA-132 Pathway. Inflammation 2016,39, 1681–1689. [CrossRef] [PubMed]
64.
Ding, X.; Jian, T.; Wu, Y.; Zuo, Y.; Li, J.; Lv, H.; Ma, L.; Ren, B.; Zhao, L.; Li, W.; et al. Ellagic acid ameliorates oxidative stress
and insulin resistance in high glucose-treated HepG2 cells via miR-223/keap1-Nrf2 pathway. Biomed. Pharm.
2019
,110, 85–94.
[CrossRef]
65.
Wei, D.Z.; Lin, C.; Huang, Y.Q.; Wu, L.P.; Huang, M.Y. Ellagic acid promotes ventricular remodeling after acute myocardial
infarction by up-regulating miR-140-3p. Biomed. Pharm. 2017,95, 983–989. [CrossRef] [PubMed]
66.
Xing, Y.; Cui, D.; Wang, S.; Wang, P.; Xing, X.; Li, H. Oleuropein represses the radiation resistance of ovarian cancer by inhibiting
hypoxia and microRNA-299-targetted heparanase expression. Food Funct. 2017,8, 2857–2864. [CrossRef] [PubMed]
67.
Xu, T.; Xiao, D. Oleuropein enhances radiation sensitivity of nasopharyngeal carcinoma by downregulating PDRG1 through
HIF1α-repressed microRNA-519d. J. Exp. Clin. Cancer Res. 2017,36, 3. [CrossRef]
Antioxidants 2021,10, 328 20 of 24
68.
Manach, C.; Scalbert, A.; Morand, C.; Remesy, C.; Jimenez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr.
2004,79, 727–747. [CrossRef]
69.
Gupta, S.; Afaq, F.; Mukhtar, H. Selective growth inhibitory, cell-cycle deregulatory and apoptotic response of apigenin in normal
versus human prostate carcinoma cells. Biochem. Biophys. Res. Commun. 2001,87, 914–920. [CrossRef]
70.
Zhang, D.; Liu, Y.; Huang, Y.; Gao, S.; Qian, S. Biopharmaceutics classification and intestinal absorption study of apigenin. Int. J.
Pharm. 2012,436, 311–317. [CrossRef] [PubMed]
71.
Liu, Y.; Hu, M. Absorption and Metabolism of Flavonoids in the Caco-2 Cell Culture Model and a Perused Rat Intestinal Model.
Drug Metab. Dispos. 2002,30, 370–377. [CrossRef]
72.
Tang, D.; Chen, K.; Huang, L.; Li, J. Pharmacokinetic properties and drug interactions of apigenin, a natural flavone. Expert Opin.
Drug Metab. Toxicol. 2017,13, 323–330. [CrossRef]
73.
Németh, K.; Plumb, G.W.; Berrin, J.G.; Juge, N.; Jacob, R.; Naim, H.Y.; Williamson, G.; Swallow, D.M.; Kroon, P.A. Deglycosylation
by small intestinal epithelial cell
β
-glucosidases is a critical step in the absorption and metabolism of dietary flavonoid glycosides
in humans. Eur. J. Nutr. 2003,42, 29–42. [CrossRef]
74.
Manach, C.; Donovan, J.L. Pharmacokinetics and metabolism of dietary flavonoids in humans. Free Rad. Res.
2004
,38, 771–785.
[CrossRef]
75.
Cardona, F.; Andrés-Lacueva, C.; Tulipani, S.; Tinahones, F.J.; Queipo-Ortuño, M.I. Benefits of polyphenols on gut microbiota and
implications in human health. J. Nutr. Biochem. 2013,24, 1415–1422. [CrossRef]
76.
Kanaze, F.; Bounartzi, M.; Georgarakis, M.; Niopas, I. Pharmacokinetics of the citrus flavanone aglycones hesperetin and
naringenin after single oral administration in human subjects. Eur. J. Clin. Nutr. 2007,61, 472–477. [CrossRef]
77.
Londono-Londono, J.; De Lima, V.R.; Jaramillo, C.; Creczynski-Pasa, T. Hesperidin and hesperetin membrane interaction:
Understanding the role of 7-O-glycoside moiety in flavonoids. Arch. Biochem. Biophys. 2010,499, 6–16. [CrossRef]
78.
Chen, J.; Lin, H.; Hu, M. Metabolism of flavonoids via enteric recycling: Role of intestinal disposition. J. Pharm. Exp. Ther.
2003
,
304, 1228–1235. [CrossRef]
79.
Gradolatto, A. Pharmacokinetics and metabolism of apigenin in female and male rats after a single oral administration. Drug Met.
Disp. 2004,33, 49–54. [CrossRef]
80.
Crespy, V.; Morand, C.; Besson, C.; Cotelle, N.; Vezin, H.; Demigne, C.; Remesy, C. The splanchnic metabolism of flavonoids
highly differed according to the nature of the compound. Am. J. Physiol. 2003,284, G980–G988. [CrossRef]
81.
Yodogawa, S.; Arakawa, T.; Sugihara, N.; Furuno, K. Glucurono- and sulfo-conjugation of kaempferol in rat liver subcellular
preparations and cultured hepatocytes. Biol. Pharm. Bull. 2003,26, 1120–1124. [CrossRef]
82.
Barve, A.; Chen, C.; Hebbar, V.; Desiderio, J.; Saw, C.L.; Kong, A.N. Metabolism, oral bioavailability and pharmacokinetics of
chemopreventive kaempferol in rats. Biopharm. Drug Dispos. 2009,30, 356–365. [CrossRef]
83.
Bonetti, A.; Marotti, I.; Dinelli, G. Urinary excretion of kaempferol from common beans (Phaseolus vulgaris L.) in humans. Int. J.
Food Sci. Nutr. 2007,58, 261–269. [CrossRef] [PubMed]
84.
DuPont, M.S.; Day, A.J.; Bennett, R.N.; Mellon, F.A.; Kroon, P.A. Absorption of kaempferol from endive, a source of kaempferol-3-
glucuronide, in humans. Eur. J. Clin. Nutr. 2004,58, 947–954. [CrossRef]
85.
Wang, F.M.; Yao, T.W.; Zeng, S. Disposition of quercetin and kaempferol in human following an oral administration of Ginkgo
biloba extract tablets. Eur. J. Drug Metab. Pharm. 2003,28, 173–177. [CrossRef]
86.
López-Lázaro, M. A new view of carcinogenesis and an alternative approach to cancer therapy. Mol. Med.
2010
,16, 144–153.
[CrossRef]
87.
Silberberg, M.; Gil-Izquierdo, A.; Combaret, L.; Remesy, C.; Scalbert, A.; Morand, C. Flavanone metabolism in healthy and
tumor-bearing rats. Biomed. Pharm. 2006,60, 529–535. [CrossRef] [PubMed]
88.
Brand, W.; Padilla, B.; Van Bladeren, P.J.; Williamson, G.; Rietjens, I.M. The effect of coadministered flavonoids on the metabolism
of hesperetin and the disposition of its metabolites in Caco-2 cell monolayers. Mol. Nutr. Food Res.
2010
,54, 851–860. [CrossRef]
89.
Manach, C.; Morand, C.; Gil-Izquierdo, A.; Bouteloup-Demange, C.; Remesy, C. Bioavailability in humans of the flavanone’s
hesperidin and narirutin after the ingestion of two doses of orange juice. Eur. J. Clin. Nutr. 2003,57, 235–242. [CrossRef]
90.
Nielsen, I.L.F.; Chee, W.S.S.; Poulsen, L.; Offord-Cavin, E.; Rasmussen, S.E.; Frederiksen, H.; Enslen, M.; Barron, D.; Horcajada,
M.N.; Williamson, G. Bioavailability is improved by enzymatic modification of the citrus flavonoid hesperidin in humans: A
randomized, double-blind, crossover trial. J. Nutr. 2006,136, 404–408. [CrossRef]
91.
Brand, W.; Boersma, M.G.; Bik, H.; Hoek-van den Hil, E.F.; Vervoort, J.; Barron, D.; Meinl, W.; Glatt, H.; Williamson, G.; van
Bladeren, P.J.; et al. Phase II metabolism of hesperetin by individual UDP-glucuronosyltransferases and sulfotransferases and rat
and human tissue samples. Drug Metab. Dispos. 2010,38, 617–625. [CrossRef]
92.
Matsumoto, H.; Ikoma, Y.; Sugiura, M.; Yano, M.; Hasegawa, Y. Identification and quantification of the conjugated metabolites
derived from orally administered hesperidin in rat plasma. J. Agric. Food Chem. 2004,52, 6653–6659. [CrossRef]
93.
Doostdar, H.; Burke, M.D.; Mayer, R.T. Bioflavonoids: Selective substrates and inhibitors for cytochrome P450 CYP1A and
CYP1B1. Toxicology 2000,144, 31–38. [CrossRef]
94.
Cho, Y.A.; Choi, D.H.; Choi, J.S. Effect of hesperidin on the oral pharmacokinetics of diltiazem and its main metabolite,
desacetyldiltiazem, in rats. J. Pharm. Pharm. 2009,61, 825–829. [CrossRef] [PubMed]
95.
Mitsunaga, Y.; Takanaga, H.; Matsuo, H.; Naito, M.; Tsuruo, T.; Ohtani, H.; Sawada, Y. Effect of bioflavonoids on vincristine
transport across blood–brain barrier. Eur. J. Pharm. 2000,395, 193–201. [CrossRef]
Antioxidants 2021,10, 328 21 of 24
96.
Piao, Y.J.; Choi, J.S. Enhanced bioavailability of verapamil after oral administration with hesperidin in rats. Arch. Pharm Res.
2008
,
31, 518–522. [CrossRef] [PubMed]
97.
Honohan, T.; Hale, R.L.; Brown, J.P.; Wingard, R.E., Jr. Synthesis and metabolic fate of hesperetin3-14C. J. Agric. Food Chem.
1976
,
24, 906–911. [CrossRef]
98.
Zuccari, G.; Baldassari, S.; Ailuno, G.; Turrini, F.; Alfei, S.; Caviglioli, G. Formulation Strategies to Improve Oral Bioavailability of
Ellagic Acid. Appl. Sci. 2020,10, 3353. [CrossRef]
99.
Amakura, Y.; Okada, M.; Tsuji, A.; Tonogai, Y. High-performance liquid chromatography determination with photodiode array
detection of ellagic acid in fresh and processed fruits. J. Chromatogr. B 2000,896, 87–93.
100.
Clifford, M.N.; Scalbert, A. Ellagitannins nature, occurrence and dietary burden. J. Sci. Food Agric.
2000
,80, 1118–1125. [CrossRef]
101.
Mao, X.; Wu, L.-F.; Zhao, H.; Liang, W.-Y.; Chen, W.J.; Han, S.-X.; Yang-Pi Cui, Q.Q.; Li, S.; Yang, G.-H.; Shao, Y.-Y.; et al. Transport
of Corilagin, Gallic Acid, and Ellagic Acid from Fructus Phyllanthi Tannin Fraction in Caco-2 Cell Monolayers. Evid. Based
Complement. Altern Med. 2016,2016, 9205379. [CrossRef]
102.
Mertens-talcot, S.U.; Jilma-Stohlawetz, P.; Rios, J.; Hingorani, L.; Derendorf, H. Absorption metabolism and antioxidant effects of
pomegranate. J. Agric. Food Chem. 2006,54, 8956–8961.
103.
Tomás-Barberán, F.A.; García-Villalba, R.; González-Sarrías, A.; Selma, M.V.; Espín, J.C. Ellagic acid metabolism by human gut
microbiota: Consistent observation of three urolithin phenotypes in intervention trials, independent of food source, age, and
health status. J. Agric. Food Chem. 2014,62, 6535–6538. [CrossRef]
104. Tomás-Barberán, F.A.; González-Sarrias, A.; Garcıa-Villalba, R.; Nunez-Sanchez, M.A.; Selma, M.V.; Garcıa-Conesa, M.T.; Espın,
J.C. Urolithins, the rescue of “old” metabolites to understand a “new” concept: Metabotypes as a nexus among phenolic
metabolism, microbiota dysbiosis, and host health status. Mol. Nutr. Food Res. 2017,61, 1500901. [CrossRef] [PubMed]
105.
Cortés-Martín, A.; García-Villalba, R.; González-Sarrías, A.; Romo-Vaquero, M.; Loria-Kohen, V.; Ramírez-de-Molina, A.; Tomás-
Barberán, F.A.; Selma, M.V.; Espín, J.C. The gut microbiota urolithin metabotypes revisited: The human metabolism of ellagicacid
is mainly determined by aging. Food Funct. 2018,9, 4100–4106. [CrossRef]
106.
Seeram, N.P.; Henning, S.M.; Zhang, Y.; Suchard, M.; Li, Z.; Heber, D. Pomegranate juice ellagitannin metabolites are present in
human plasma and some persist in urine for up to 48 h. J. Nutr. 2006,136, 2481–2485. [CrossRef]
107.
González-Sarrías, A.; García-Villalba, R.; Núñez-Sánchez, M.Á.; Tomé-Carneiro, J.; Zafrilla, P.; Mulero, J.; Tomás-Barberán, F.A.;
Espín, J.C. Identifying the limits for ellagic acid bioavailability: A crossover pharmacokinetic study in healthy volunteers after
consumption of pomegranate extracts. J. Funct. Foods 2015,19, 225–235. [CrossRef]
108.
Visioli, F.; Galli, C.; Bornet, F.; Mattei, A.; Patelli, R.; Galli, G.; Caruso, D. Olive oil phenolics are dose-dependently absorbed in
humans. Febs. Lett. 2000,468, 159–160. [CrossRef]
109.
Miró-Casas, E.; FarréAlbaladejo, M.; Covas, M.I.; Rodriguez, J.O.; Menoyo Colomer, E.; Lamuela Raventós, R.M.; de la Torre, R.
Capillary gas chromatography-mass spectrometry quantitative determination of hydroxytyrosol and tyrosol in human urine after
olive oil intake. Anal. Biochem. 2001,294, 63–72. [CrossRef] [PubMed]
110.
Vissers, M.N.; Zock, P.L.; Roodenburg, A.J.C.; Leenen, R.; Katan, M.B. Olive Oil Phenols Are Absorbed in Humans. J. Nutr.
2002
,
132, 409–417. [CrossRef]
111.
Lemonakis, N.; Mougios, V.; Halabalaki, M.; Skaltsounis, A.-L.; Gikas, E. A novel bioanalytical method based on UHPLC-
HRMS/MS for the quantification of oleuropein in human serum. Application to a pharmacokinetic study: Quantification of
oleuropein in human serum. Biomed. Chromatogr. 2016,30, 2016–2023. [CrossRef] [PubMed]
112.
de Bock, M.; Thorstensen, E.B.; Derraik, J.G.B.; Henderson, H.V.; Hofman, P.L.; Cutfield, W.S. Human absorption and metabolism
of oleuropein and hydroxytyrosol ingested as olive (Olea europaea L.) leaf extract. Mol. Nutr. Food Res.
2013
,57, 2079–2085.
[CrossRef]
113.
Mosele, J.I.; Martín-Peláez, S.; Macià, A.; Farràs, M.; Valls, R.-M.; Catalán, Ú.; Motilva, M.-J. Faecal microbial metabolism of olive
oil phenolic compounds: In vitro and in vivo approaches. Mol. Nutr. Food Res. 2014,58, 1809–1819. [CrossRef] [PubMed]
114.
Santos, M.M.; Piccirillo, C.; Castro, P.M.L.; Kalogerakis, N.; Pintado, M.E. Bioconversion of oleuropein to hydroxytyrosol by lactic
acid bacteria. World J. Microbiol. Biotechnol. 2012,28, 2435–2440. [CrossRef]
115.
Aponte, M.; Ungaro, F.; d’Angelo, I.; De Caro, C.; Russo, R.; Blaiotta, G.; Dal Piaz, F.; Calignano, A.; Miro, A. Improving
in vivo
conversion of oleuropein into hydroxytyrosol by oral granules containing probiotic Lactobacillus plantarum 299v and an Olea
europaea standardized extract. Int. J. Pharm. 2018,543, 73–82. [CrossRef] [PubMed]
116.
Arafah, A.; Rehman, M.U.; Mir, T.M.; Wali, A.F.; Ali, R.; Qamar, W.; Khan, R.; Ahmad, A.; Aga, S.S.; Alqahtani, S.; et al.
Multi-Therapeutic Potential of Naringenin (4
0
,5,7-Trihydroxyflavonone): Experimental Evidence and Mechanisms. Plants
2020
,
9, 1784. [CrossRef]
117.
Lim, W.; Park, S.; Bazer, F.W.; Song, G. Naringenin-Induced Apoptotic Cell Death in Prostate Cancer Cells Is Mediated via the
PI3K/AKT and MAPK Signaling Pathways. J. Cell Biochem. 2017,118, 1118–1131. [CrossRef] [PubMed]
118.
Casey, S.C.; Amedei, A.; Aquilano, K.; Azmi, A.S.; Benencia, F.; Bhakta, D.; Bilsland, A.E.; Boosani, C.S.; Chen, S.; Ciriolo, M.R.;
et al. Cancer prevention and therapy through the modulation of the tumor microenvironment. Semin. Cancer Biol.
2015
,35,
S199–S223. [CrossRef]
119.
Alam, M.A.; Subhan, N.; Rahman, M.M.; Uddin, S.J.; Reza, H.M.; Sarker, S.D. Effect of citrus flavonoids, naringin and naringenin,
on metabolic syndrome and their mechanisms of action. Adv. Nutr. 2014,5, 404–417. [CrossRef] [PubMed]
Antioxidants 2021,10, 328 22 of 24
120.
Zaidun, N.H.; Thent, Z.C.; Latiff, A.A. Combating oxidative stress disorders with citrus flavonoid: Naringenin. Life Sci.
2018
,208,
111–122. [CrossRef] [PubMed]
121.
Nouri, Z.; Fakhri, S.; El-Senduny, F.F.; Sanadgol, N.; Abd-ElGhani, G.E.; Farzaei, M.H.; Chen, J.T. On the Neuroprotective Effects
of Naringenin: Pharmacological Targets, Signaling Pathways, Molecular Mechanisms, and Clinical Perspective. Biomolecules
2019
,
9, 690. [CrossRef] [PubMed]
122.
Alberca, R.W.; Teixeira, F.M.E.; Beserra, D.R.; de Oliveira, E.A.; Andrade, M.M.S.; Pietrobon, A.J.; Sato, M.N. Perspective: The
Potential Effects of Naringenin in COVID-19. Front. Immunol. 2020,11, 570919. [CrossRef]
123.
Joshi, R.; Kulkarni, Y.A.; Wairkar, S. Pharmacokinetic, pharmacodynamic and formulations aspects of Naringenin: An update.
Life Sci. 2018,215, 43–56. [CrossRef] [PubMed]
124.
Chen, T.; Su, W.; Yan, Z.; Wu, H.; Zeng, X.; Peng, W.; Gan, L.; Zhang, Y.; Yao, H. Identification of naringin metabolites mediated
by human intestinal microbes with stable isotope-labeling method and UFLC-Q-TOF-MS/MS. J. Pharm. Biomed. Anal.
2018
,161,
262–272. [CrossRef] [PubMed]
125.
Kay, C.D.; Pereira-Caro, G.; Ludwig, I.A.; Clifford, M.N.; Crozier, A. Anthocyanins and Flavanones Are More Bioavailable than
Previously Perceived: A Review of Recent Evidence. Annu. Rev. Food Sci. Technol. 2017,8, 155–180. [CrossRef] [PubMed]
126.
Murugesan, N.; Woodard, K.; Ramaraju, R.; Greenway, F.L.; Coulter, A.A.; Rebello, C.J. Naringenin Increases Insulin Sensitivity
and Metabolic Rate: A Case Study. J. Med. Food 2020,23, 343–348. [CrossRef] [PubMed]
127.
Habauzit, V.; Verny, M.A.; Milenkovic, D.; Barber-Chamoux, N.; Mazur, A.; Dubray, C.; Morand, C. Flavanones protect from
arterial stiffness in postmenopausal women consuming grapefruit juice for 6 mo: A randomized, controlled, crossover trial. Am.
J. Clin. Nutr. 2015,102, 66–74. [CrossRef] [PubMed]
128.
Olas, B. Honey and Its Phenolic Compounds as an Effective Natural Medicine for Cardiovascular Diseases in Humans? Nutrients
2020,12, 283. [CrossRef] [PubMed]
129.
Salehi, B.; Venditti, A.; Sharifi-Rad, M.; Kr˛egiel, D.; Sharifi-Rad, J.; Durazzo, A.; Lucarini, M.; Santini, A.; Souto, E.B.; Novellino,
E.; et al. The Therapeutic Potential of Apigenin. Int. J. Mol. Sci. 2019,20, 1305. [CrossRef] [PubMed]
130.
Li, F.; Lang, F.; Zhang, H.; Xu, L.; Wang, Y.; Zhai, C.; Hao, E. Apigenin Alleviates Endotoxin-Induced Myocardial Toxicity by
Modulating Inflammation, Oxidative Stress, and Autophagy. Oxid. Med. Cell Longev. 2017,2017, 2302896. [CrossRef] [PubMed]
131.
Zick, S.M.; Wright, B.D.; Sen, A.; Arnedt, J.T. Preliminary examination of the efficacy and safety of a standardized chamomile
extract for chronic primary insomnia: A randomized placebo-controlled pilot study. BMC Complement. Altern. Med.
2011
,11, 78.
[CrossRef]
132.
Amsterdam, J.D.; Shults, J.; Soeller, I.; Mao, J.J.; Rockwell, K.; Newberg, A.B. Chamomile (Matricaria recutita) may provide
antidepressant activity in anxious, depressed humans: An exploratory study. Altern. Health Med. 2012,18, 44–49.
133.
Mao, J.J.; Xie, S.X.; Keefe, J.R.; Soeller, I.; Li, Q.S.; Amsterdam, J.D. Long-term chamomile (Matricaria chamomilla L.) treatment for
generalized anxiety disorder: A randomized clinical trial. Phytomedicine 2016,23, 1735–1742. [CrossRef]
134.
Vollmer, M.; Esders, S.; Farquharson, F.M.; Neugart, S.; Duncan, S.H.; Schreiner, M.; Louis, P.; Maul, R.; Rohn, S. Mutual Interaction
of Phenolic Compounds and Microbiota: Metabolism of Complex Phenolic Apigenin-C- and Kaempferol-O-Derivatives by
Human Fecal Samples. J. Agric. Food Chem. 2018,66, 485–497. [CrossRef] [PubMed]
135.
Ren, J.; Lu, Y.; Qian, Y.; Chen, B.; Wu, T.; Ji, G. Recent progress regarding kaempferol for the treatment of various diseases. Exp.
Med. 2019,18, 2759–2776. [CrossRef] [PubMed]
136.
Devi, K.P.; Malar, D.S.; Nabavi, S.F.; Sureda, A.; Xiao, J.; Nabavi, S.M.; Daglia, M. Kaempferol and inflammation: From chemistry
to medicine. Pharm. Res. 2015,99, 1–10. [CrossRef] [PubMed]
137.
Imran, M.; Rauf, A.; Shah, Z.A.; Saeed, F.; Imran, A.; Arshad, M.U.; Ahmad, B.; Bawazeer, S.; Atif, M.; Peters, D.G.; et al.
Chemo-preventive and therapeutic effect of the dietary flavonoid kaempferol: A comprehensive review. Phytother. Res.
2019
,33,
263–275. [CrossRef] [PubMed]
138.
Kashyap, D.; Sharma, A.; Tuli, H.S.; Sak, K.; Punia, S.; Mukherjee, T.K. Kaempferol—A dietary anticancer molecule with multiple
mechanisms of action: Recent trends and advancements. J. Funct. Foods 2017,30, 203–219. [CrossRef]
139.
Navarro, S.L.; Schwarz, Y.; Song, X.; Wang, C.-Y.; Chen, C.; Trudo, S.P.; Kristal, A.R.; Kratz, M.; Eaton, D.L.; Lampe, J.W.
Cruciferous Vegetables Have Variable E_ects on Biomarkers of Systemic Inflammation in a Randomized Controlled Trial in
Healthy Young Adults. J. Nutr. 2014,144, 1850–1857. [CrossRef] [PubMed]
140.
Alam, W.; Khan, H.; Shah, M.A.; Cauli, O.; Saso, L. Kaempferol as a Dietary Anti-Inflammatory Agent: Current Therapeutic
Standing. Molecules 2020,25, 4073. [CrossRef] [PubMed]
141.
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. Devel. 2019,13, 3497–3514. [CrossRef]
142.
Kim, J.; Wie, M.B.; Ahn, M.; Tanaka, A.; Matsuda, H.; Shin, T. Benefits of hesperidin in central nervous system disorders: A review.
Anat. Cell Biol. 2019,52, 369–377. [CrossRef]
143.
Jawien, A.; Bouskela, E.; Allaert, F.A.; Nicolaïdes, A.N. The place of Ruscus extract, hesperidin methyl chalcone, and vitamin C in
the management of chronic venous disease. Int. Angiol. 2017,36, 31–41.
144.
Stevens, Y.; Rymenant, E.V.; Grootaert, C.; Camp, J.V.; Possemiers, S.; Masclee, A.; Jonkers, D. The Intestinal Fate of Citrus
Flavanones and Their Effects on Gastrointestinal Health. Nutrients 2019,11, 1464. [CrossRef]
145.
Homayouni, F.; Haidari, F.; Hedayati, M.; Zakerkish, M.; Ahmadi, K. Blood pressure lowering and anti-inflammatory effects of
hesperidin in type 2 diabetes; a randomized double-blind controlled clinical trial. Phytother. Res.
2018
,32, 1073–1079. [CrossRef]
Antioxidants 2021,10, 328 23 of 24
146.
Salden, B.N.; Troost, F.J.; de Groot, E.; Stevens, Y.R.; Garcés-Rimón, M.; Possemiers, S.; Winkens, B.; Masclee, A.A. Randomized
clinical trial on the efficacy of hesperidin 2S on validated cardiovascular biomarkers in healthy overweight individuals. Am. J.
Clin. Nutr. 2016,104, 1523–1533. [CrossRef]
147.
Hajialyani, M.; Hosein Farzaei, M.; Echeverría, J.; Nabavi, S.M.; Uriarte, E.; Sobarzo-Sánchez, E. Hesperidin as a Neuroprotective
Agent: A Review of Animal and Clinical Evidence. Molecules 2019,24, 648. [CrossRef]
148.
Corsale, I.; Carrieri, P.; Martellucci, J.; Piccolomini, A.; Verre, L.; Rigutini, M.; Panicucci, S. Flavonoid mixture (diosmin, troxerutin,
rutin, hesperidin, quercetin) in the treatment of I-III degree hemorroidal disease: A double-blind multicenter prospective
comparative study. Int. J. Colorectal. Dis. 2018,33, 1595–1600. [CrossRef]
149.
Martin, B.R.; McCabe, G.P.; McCabe, L.; Jackson, G.S.; Horcajada, M.N.; Offord-Cavin, E.; Peacock, M.; Weaver, C.M. Effect of
Hesperidin with and Without a Calcium (Calcilock) Supplement on Bone Health in Postmenopausal Women. J. Clin. Endocrinol.
Metab. 2016,101, 923–927. [CrossRef]
150.
Kerimi, A.; Nyambe-Silavwe, H.; Gauer, J.S.; Tomás-Barberán, F.A.; Williamson, G. Pomegranate juice, but not an extract, confers
a lower glycemic response on a high-glycemic index food: Randomized, crossover, controlled trials in healthy subjects. Am. J.
Clin. Nutr. 2017,106, 1384–1393. [CrossRef]
151.
Long, J.; Guo, Y.; Yang, J.; Henning, S.M.; Lee, R.P.; Rasmussen, A.; Zhang, L.; Lu, Q.Y.; Heber, D.; Li, Z. Bioavailability and
bioactivity of free ellagic acid compared to pomegranate juice. Food Funct. 2019,10, 6582–6588. [CrossRef]
152.
Danesi, F.; Ferguson, L.R. Could Pomegranate Juice Help in the Control of Inflammatory Diseases? Nutrients
2017
,9, 958.
[CrossRef]
153.
Ríos, J.L.; Giner, R.M.; Marín, M.; Recio, M.C. A Pharmacological Update of Ellagic Acid. Planta Med.
2018
,84, 1068–1093.
[CrossRef] [PubMed]
154.
Alfei, S.; Marengo, B.; Zuccari, G. Oxidative Stress, Antioxidant Capabilities, and Bioavailability: Ellagic Acid or Urolithins?
Antioxidants 2020,9, 707. [CrossRef] [PubMed]
155.
Kang, I.; Buckner, T.; Shay, N.F.; Gu, L.; Chung, S. Improvements in Metabolic Health with Consumption of Ellagic Acid and
Subsequent Conversion into Urolithins: Evidence and Mechanisms. Adv. Nutr. 2016,7, 961–972. [CrossRef]
156.
Li, Z.; Henning, S.M.; Lee, R.P.; Lu, Q.Y.; Summanen, P.H.; Thames, G.; Corbett, K.; Downes, J.; Tseng, C.H.; Finegold, S.M.; et al.
Pomegranate extract induces ellagitannin metabolite formation and changes stool microbiota in healthy volunteers. Food Funct.
2015,6, 2487–2495. [CrossRef] [PubMed]
157.
Ahmed, T.; Setzer, W.N.; Nabavi, S.F.; Orhan, I.E.; Braidy, N.; Sobarzo-Sanchez, E.; Nabavi, S.M. Insights Into Effects of Ellagic
Acid on the Nervous System: A Mini Review. Curr. Pharm. Des. 2016,22, 1350–1360. [CrossRef] [PubMed]
158.
Liu, Y.; Yu, S.; Wang, F.; Yu, H.; Li, X.; Dong, W.; Lin, R.; Liu, Q. Chronic administration of ellagic acid improved the cognition in
middle-aged overweight men. Appl. Physiol. Nutr. Metab. 2018,43, 266–273. [CrossRef]
159.
Ammar, A.; Turki, M.; Hammouda, O.; Chtourou, H.; Trabelsi, K.; Bouaziz, M.; Abdelkarim, O.; Hoekelmann, A.; Ayadi, F.;
Souissi, N.; et al. Effects of Pomegranate Juice Supplementation on Oxidative Stress Biomarkers Following Weightlifting Exercise.
Nutrients 2017,9, 819. [CrossRef]
160.
Karkovi´c Markovi´c, A.; Tori´c, J.; Barbari´c, M.; Jakobuši´c Brala, C. Hydroxytyrosol, Tyrosol and Derivatives and Their Potential
Effects on Human Health. Molecules 2019,24, 2001. [CrossRef]
161.
D’Adamo, S.; Cetrullo, S.; Panichi, V.; Mariani, E.; Flamigni, F.; Borzì, R.M. Nutraceutical Activity in Osteoarthritis Biology: A
Focus on the Nutrigenomic Role. Cells 2020,9, 1232. [CrossRef]
162.
Larussa, T.; Imeneo, M.; Luzza, F. Olive Tree Biophenols in Inflammatory Bowel Disease: When Bitter is Better. Int. J. Mol. Sci.
2019,20, 1390. [CrossRef]
163.
Santangelo, C.; Vari, R.; Scazzocchio, B.; De Sanctis, P.; Giovannini, C.; D’Archivio, M.; Masella, R. Anti-inflammatory Activity of
Extra Virgin Olive Oil Polyphenols: Which Role in the Prevention and Treatment of Immune-Mediated Inflammatory Diseases?
Endocr. Metab. Immune Disord. Drug Targets 2018,18, 36–50. [CrossRef]
164.
Somerville, V.; Moore, R.; Braakhuis, A. The Effect of Olive Leaf Extract on Upper Respiratory Illness in High School Athletes: A
Randomised Control Trial. Nutrients 2019,11, 358. [CrossRef] [PubMed]
165.
de Bock, M.; Derraik, J.G.; Brennan, C.M.; Biggs, J.B.; Morgan, P.E.; Hodgkinson, S.C.; Hofman, P.L.; Cutfield, W.S. Olive (Olea
europaea L.) leaf polyphenols improve insulin sensitivity in middle-aged overweight men: A randomized, placebo-controlled,
crossover trial. PLoS ONE 2013,8, e57622. [CrossRef]
166.
Lockyer, S.; Corona, G.; Yaqoob, P.; Spencer, J.P.; Rowland, I. Secoiridoids delivered as olive leaf extract induce acute improvements
in human vascular function and reduction of an inflammatory cytokine: A randomised, double-blind, placebo-controlled, cross-
over trial. Br. J. Nutr. 2015,114, 75–83. [CrossRef] [PubMed]
167.
Lopez-Huertas, E.; Fonolla, J. Hydroxytyrosol supplementation increases vitamin C levels
in vivo
. A human volunteer trial.
Redox. Biol. 2017,11, 384–389. [CrossRef]
168.
Quirós-Fernández, R.; López-Plaza, B.; Bermejo, L.M.; Palma-Milla, S.; Gómez-Candela, C. Supplementation with Hydroxytyrosol
and Punicalagin Improves Early Atherosclerosis Markers Involved in the Asymptomatic Phase of Atherosclerosis in the Adult
Population: A Randomized, Placebo-Controlled, Crossover Trial. Nutrients 2019,11, 640. [CrossRef]
169.
Bellavite, P.; Donzelli, A. Hesperidin and SARS-CoV-2: New Light on the Healthy Function of Citrus Fruits. Antioxidants
2020
,
9, 742. [CrossRef]
Antioxidants 2021,10, 328 24 of 24
170.
Dabeek, W.M.; Ventura Marra, M. Dietary Quercetin and Kaempferol: Bioavailability and Potential Cardiovascular-Related
Bioactivity in Humans. Nutrients 2019,11, 2288. [CrossRef] [PubMed]
171.
Cione, E.; La Torre, C.; Cannataro, R.; Caroleo, M.C.; Plastina, P.; Gallelli, L. Quercetin, Epigallocatechin Gallate, Curcumin, and
Resveratrol: From Dietary Sources to Human MicroRNA Modulation. Molecules 2019,25, 63. [CrossRef]
... It is a flavonoid that is widely found as a glycoside in vegetables, fruits, herbs, and plant beverages, such as chamomile infusions [48,49]. According to the literature [50], the highest concentration of AP (78.65 mg) is found in 100 g of celery seeds ( Figure 3). ...
... It is a flavonoid that is widely found as a glycoside in vegetables, fruits, herbs, and plant beverages, such as chamomile infusions [48,49]. According to the literature [50], the highest concentration of AP (78.65 mg) is found in 100 g of celery seeds (Figure 3). The flavonoid was classified as Class II according to the Biopharmaceutical Classification System (BCS). ...
... Apigenin content in individual plant products based on[50]. Prepared in Canva ® (Sydney, Australia). ...
Article
Full-text available
Antibiotic resistance in microorganisms is an escalating global concern, exacerbated by their formation of biofilms, which provide protection through an extracellular matrix and communication via quorum sensing, enhancing their resistance to treatment. This situation has driven the search for alternative approaches, particularly those using natural compounds. This study explores the potential of phytochemicals, such as quercetin, apigenin, arbutin, gallic acid, proanthocyanidins, and rutin, known for their antibacterial properties and ability to inhibit biofilm formation and disrupt mature biofilms. The methods used in this study included a comprehensive review of current literature assessing the bioavailability, distribution, and effective concentrations of these compounds in treating biofilm-associated infections. The results indicate that these phytochemicals exhibit significant antibacterial effects, reduce biofilm’s structural integrity, and inhibit bacterial communication pathways. Moreover, their potential use in combination with existing antibiotics may enhance therapeutic outcomes. The findings support the conclusion that phytochemicals offer promising additions to anti-biofilm strategies and are capable of complementing or replacing conventional treatments, with appropriate therapeutic levels and delivery mechanisms being key to their effectiveness. This insight underscores the need for further research into their clinical applications for treating infections complicated by biofilms.
... The Mediterranean diet, characterized by a high intake of fruits, vegetables, polyphenol-rich foods, fiber, and vitamins, exemplifies a model of healthy eating [28,29]. The antioxidant and anti-inflammatory properties inherent in this diet can alleviate psoriasis symptoms and have been found to have a negative correlation with the severity of the condition [30]. ...
Article
Full-text available
Psoriasis is a chronic autoimmune and autoinflammatory disorder defined by abnormal skin cell turnover and inflammation, resulting in the formation of plaques on the skin. Although biologic therapies targeting interleukin (IL)-17 and IL-23 have significantly improved the treatment landscape for moderateto-severe psoriasis, they are not effective for all patients. This highlights the need for additional therapeutic strategies. In recent years, exploring novel treatment avenues such as targeting IL-21, small nucleolar RNA (snoRNA) Snora73, the gut microbiome, and natural remedies have shown increasing promise in managing psoriasis. Interleukin-21 is a cytokine that plays a critical role in the differentiation and function of Th17 cells, which are central to the pathogenesis of psoriasis. Recent studies have demonstrated that neutralizing IL-21 with specific antibodies can help restore immune homeostasis, reducing disease severity and improving patient outcomes. Targeting IL-21 may be particularly beneficial for patients resistant to conventional therapies like IL-17 and IL-23 inhibitors. In addition to IL-21, snoRNA Snora73 has emerged as a novel target for psoriasis treatment. Snora73 regulates cell proliferation by interacting with miR-30745p and pre-B-cell leukemia homeobox 1 (PBX1), promoting abnormal cell turnover in psoriasis. The gut microbiome is increasingly recognized for its role in autoimmune diseases, including psoriasis. Imbalances in the microbiome have been linked to disease exacerbation, triggering systemic inflammation and altering immune responses. Moreover, various natural treatments have gained attention for their anti-inflammatory properties. These natural therapies could serve as adjuncts to existing treatments, offering a complementary approach that minimizes side effects while improving patient outcomes. Targeting IL-21, Snora73, and the gut microbiome, as well as utilizing natural treatments, may provide new opportunities for more effective, personalized management of psoriasis.
... 33 It is also known that many dietary supplement agents listed in Table 4 can alter the activities of many enzymes such as GSK-3β and CDK5 involved in Aβ and tau modifications but also affect the levels of miRNA transcripts for these genes. 152 Additionally, because of the changes in the levels of various miRNAs associated with AD development, some of them can be used as a potential diagnosis tool as well as therapeutics for AD. Since many different miRNAs can be contained in circulating extracellular vesicles (EVs) or exosomes, use of EVs and exosomes for diagnostic and therapeutic (theragnostic) purposes for AD patients or animal models need to be further characterized. ...
Article
Full-text available
Alzheimer’s disease (AD) is an aging-related irreversible neurodegenerative disease affecting mostly the elderly population. The main pathological features of AD are the extracellular Aβ plaques generated by APP cleavage through the amyloidogenic pathway, the intracellular neurofibrillary tangles (NFT) resulting from the hyperphosphorylated tau proteins, and cholinergic neurodegeneration. However, the actual causes of AD are unknown, but several studies suggest hereditary mutations in PSEN1 and -2, APOE4, APP, and the TAU genes are the major perpetrators. In order to understand the etiology and pathogenesis of AD, various hypotheses are proposed. These include the following hypotheses: amyloid accumulation, tauopathy, inflammation, oxidative stress, mitochondrial dysfunction, glutamate/excitotoxicity, cholinergic deficiency, and gut dysbiosis. Currently approved therapeutic interventions are donepezil, galantamine, and rivastigmine, which are cholinesterase inhibitors (ChEIs), and memantine, which is an N-methyl-d-aspartate (NMDA) antagonist. These treatment strategies focus on only symptomatic management of AD by attenuating symptoms but not regeneration of neurons or clearance of Aβ plaques and hyperphosphorylated Tau. This review focuses on the pathophysiology, novel therapeutic targets, and disease-altering treatments such as α-secretase modulators, active immunotherapy, passive immunotherapy, natural antioxidant products, nanomaterials, antiamyloid therapy, tau aggregation inhibitors, transplantation of fecal microbiota or stem cells, and microtubule stabilizers that are in clinical trials or still under investigation.
Chapter
There is no consensus yet on the dietary treatment of migraine, which causes headaches and recurrent attacks and is caused by vascular, biochemical, neurogenic or platelet-based causes. However, various dietary patterns, such as the Mediterranean diet and DASH, have come to the fore. Dietary components may affect migraine clinical outcomes by improving vasodilation, systemic inflammation, and cerebral glucose metabolism pathways. The Mediterranean diet has a neuroprotective role by preventing excessive release of inflammatory mediators and reducing neuroinflammation, pain stimulation, oxidative stress, and lipid peroxidation. The Mediterranean diet is associated with a lower risk of severe headache or migraine and may be a strategy for reducing migraine duration, frequency, and severity. The DASH diet model, which includes plant-based foods with low sodium content, has gained popularity because of its therapeutic effect on systemic inflammatory processes and its antimigraine properties. DASH may help alleviate migraine symptoms by preventing oxidative stress, endothelial dysfunction, and neuroinflammation. In addition, DASH has been associated with reducing the severity and frequency of migraine attacks because of its low sodium-induced antivasodilator effect.
Article
Sarcopenia and osteoporosis are age-related musculoskeletal pathologies that often develop in parallel, and numerous studies support the concept of a bone–muscle unit, where deep interaction between the two tissues takes place. In Mediterranean areas, the lowest incidence of osteoporosis within Europe is observed, so the Mediterranean diet was suggested to play an important role. Consequently, in this study, oleuropein, a phenolic compound found in olive oil, and polydatin, another natural polyphenol found in the Mediterranean diet, were evaluated to determine their beneficial effects on bone and muscle metabolism. In human osteoblasts and skeletal muscle myoblasts, the effects were examined, and, after analyzing the cytotoxic effect to find non-toxic doses, the modulation of bone and muscle differentiation markers was evaluated at the gene and protein levels using PCR, Western blot, and immunohistochemistry. Interestingly, the compounds increased markers involved in osteoblast differentiation, such as osteocalcin, type I collagen, and dentin-sialo-phosphoprotein, as well as markers involved in myoblast differentiation, such as myogenic regulatory factors and creatine kinase. These effects were most noticeable when the compounds were administered together. These results suggest a beneficial role for oleuropein–polydatin association on bone and muscle tissue pathologies simultaneously.
Article
Cancer is a significant cause of death worldwide. It has been suggested that the consumption of flavonoids decreases the risk for cancer by increasing phase II enzymes, such as Nicotinamide Adenine Dinucleotide Phosphate Hydrogen (NAD(P)H) quinone oxidoreductase 1 (NQO1), glutathione S-transferases, and Uridine 5'-diphospho- (UDP)-glucuronosyltransferases that assist in removing carcinogens from the human body. Flavonoids are bioactive compounds found in a variety of dietary sources, including fruits, vegetables, legumes, nuts, and teas. As such, it is important to investigate which flavonoids are involved in the metabolism of carcinogens to help reduce the risk of cancer. Therefore, the objective of this narrative review was to investigate the effects of commonly consumed flavonoids on NQO1 mRNA expression, protein, and activity in human cell and murine models. PubMed was used to search for peer-reviewed journal articles, which demonstrated that selected flavonoids (e.g., quercetin, apigenin, luteolin, genistein, and daidzein) increase NQO1, and therefore, increase the excretion of carcinogens. However, more research is needed regarding the mechanisms by which flavonoids induce NQO1. Furthermore, it is suggested that future efforts focus on providing precise flavonoid recommendations to decrease the risk factors for chronic diseases.
Article
Full-text available
Extensive research has been carried out during the last few decades, providing a detailed account of thousands of discovered phytochemicals and their biological activities that have the potential to be exploited for a wide variety of medicinal purposes. These phytochemicals, which are pharmacologically important for clinical use, primarily consist of polyphenols, followed by terpenoids and alkaloids. There are numerous published reports indicating the primary role of phytochemicals proven to possess therapeutic potential against several diseases. However, not all phytochemicals possess significant medicinal properties, and only some of them exhibit viable biological effects. Naringenin, a flavanone found in citrus fruits, is known to improve immunity, repair DNA damage, and scavenge free radicals. Despite the very low bioavailability of naringenin, it is known to exhibit various promising biological properties of medicinal importance, including anti-inflammatory and antioxidant activities. This review focuses on the various aspects related to naringenin, particularly its physicochemical, pharmacokinetic, and pharmacodynamic properties. Furthermore, various pharmacological activities of naringenin, such as anticancer, antidiabetic, hepatoprotective, neuroprotective, cardioprotective, nephroprotective, and gastroprotective effects, have been discussed along with their mechanisms of action.
Article
Full-text available
Kaempferol, a natural flavonoid compound, possesses potent myocardial protective property in ischemia/reperfusion (I/R), but the underlying mechanism is not well understood. The present study was aimed to explore whether miR-21 contributes to the cardioprotective effect of kaempferol on hypoxia/reoxygenation (H/R)-induced H9c2 cell injury via regulating Notch/phosphatase and tensin homologue (PTEN)/Akt signaling pathway. Results revealed that kaempferol obviously attenuates H/R-induced the damages of H9c2 cells as evidence by the up-regulation of cell viability, the down-regulation of lactate dehydrogenase (LDH) activity, the reduction of apoptosis rate and pro-apoptotic protein (Bax) expression, and the increases of anti-apoptotic protein (Bcl-2) expression. In addition, kaempferol enhanced miR-21 level in H9c2 cells exposed to H/R, and inhibition of miR-21 induced by transfection with miR-21 inhibitor significantly blocked the protection of kaempferol against H/R-induced H9c2 cell injury. Furthermore, kaempferol eliminated H/R-induced oxidative stress and inflammatory response as illustrated by the decreases in reactive oxygen species generation and malondialdehyde content, the increases in antioxidant enzyme superoxide dismutase and glutathione peroxidase activities, the decreases in pro-inflammatory cytokines interleukin (IL)-1β, IL-8 and tumor necrosis factor-alpha levels, and an increase in anti-inflammatory cytokine IL-10 level, while these effects of kaempferol were all reversed by miR-21 inhibitor. Moreover, results elicited that kaempferol remarkably blocks H/R-induced the down-regulation of Notch1 expression, the up-regulation of PTEN expression, and the reduction of P-Akt/Akt, indicating that kaempferol promotes Notch1/PTEN/AKT signaling pathway, and knockdown of Notch1/PTEN/AKT signaling pathway induced by Notch1 siRNA also abolished the protection of kaempferol against H/R-induced the damage of H9c2 cells. Notably, miR-21 inhibitor alleviated the promotion of kaempferol on Notch/PTEN/Akt signaling pathways in H9c2 cells exposed to H/R. Taken together, these above findings suggested thatmiR-21 mediates the protection of kaempferol against H/R-induced H9c2 cell injuryvia promoting Notch/PTEN/Akt signaling pathway.
Article
Full-text available
This aim of the present study was to identify the relationship between hesperidin and microRNA (miR)‑132, and to study the role of hesperidin and miR‑132 in the pathogenesis of non‑small cell lung cancer (NSCLC). Computational analysis and luciferase assays were performed to identify the target of miR‑132. Subsequently, reverse transcription‑quantitative PCR and western blot assays were used to detect the effect of miR‑132 and hesperidin on the expression of haematological and neurological expressed 1 (HN1) and zinc finger E‑box binding homeobox 2 (ZEB2). Finally, MTT assays and flow cytometry analysis were used to investigate the effect of hesperidin on cell proliferation and apoptosis. ZEB2 was identified as a target gene of miR‑132, and transfection with miR‑132 mimic reduced the luciferase activity of the wild‑type ZEB2 3'‑untranslated region (3'‑UTR) but not that of the mutant ZEB2 3'‑UTR. By contrast, neither transfection with miR‑132 mimic nor hesperidin treatment affected HN1 expression. Furthermore, hesperidin evidently inhibited cell proliferation and promoted apoptosis in a dose‑dependent manner. Furthermore, the tumour volume in rats transplanted with NSCLC cells and treated with hesperidin was notably smaller compared with that in rats transplanted with NSCLC cells alone, while treatment with hesperidin significantly reduced the colony formation efficiency of NSCLC cells by increasing miR‑132 expression and decreasing ZEB2 expression. To the best of our knowledge, the present study demonstrated for the first time that the administration of hesperidin decreased the expression of ZEB2 by upregulating the expression of miR‑132, which in turn promoted apoptosis and inhibited the proliferation of NSCLC cells.
Article
Full-text available
Coronavirus disease 2019 (COVID-19), caused by Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), was declared a pandemic by the World Health Organization in March 2020. Severe COVID-19 cases develop severe acute respiratory syndrome, which can result in multiple organ failure, sepsis, and death. The higher risk group includes the elderly and subjects with pre-existing chronic illnesses such as obesity, hypertension, and diabetes. To date, no specific treatment or vaccine is available for COVID-19. Among many compounds, naringenin (NAR) a flavonoid present in citrus fruits has been investigated for antiviral and anti-inflammatory properties like reducing viral replication and cytokine production. In this perspective, we summarize NAR potential anti-inflammatory role in COVID-19 associated risk factors and SARS-CoV-2 infection.
Article
Full-text available
Inflammation is a physiological response to different pathological, cellular or vascular damages due to physical, chemical or mechanical trauma. It is characterized by pain, redness, heat and swelling. Current natural drugs are carefully chosen as a novel therapeutic strategy for the management of inflammatory diseases. Different phytochemical constituents are present in natural products. These phytochemicals have high efficacy both in vivo and in vitro. Among them, flavonoids occur in many foods, vegetables and herbal medicines and are considered as the most active constituent, having the ability to attenuate inflammation. Kaempferol is a polyphenol that is richly found in fruits, vegetables and herbal medicines. It is also found in plant-derived beverages. Kaempferol is used in the management of various ailments but there is no available review article that can summarize all the natural sources and biological activities specifically focusing on the anti-inflammatory effect of kaempferol. Therefore, this article is aimed at providing a brief updated review of the literature regarding the anti-inflammatory effect of kaempferol and its possible molecular mechanisms of action. Furthermore, the review provides the available updated literature regarding the natural sources, chemistry, biosynthesis, oral absorption, metabolism, bioavailability and therapeutic effect of kaempferol.
Article
Full-text available
Among the many approaches to Coronavirus disease 2019 (COVID-19) prevention, the possible role of nutrition has so far been rather underestimated. Foods are very rich in substances, with a potential beneficial effect on health, and some of these could have an antiviral action or be important in modulating the immune system and in defending cells from the oxidative stress associated with infection. This short review draws the attention on some components of citrus fruits, and especially of the orange (Citrus sinensis), well known for its vitamin and flavonoid content. Among the flavonoids, hesperidin has recently attracted the attention of researchers, because it binds to the key proteins of the Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Several computational methods, independently applied by different researchers, showed that hesperidin has a low binding energy, both with the coronavirus “spike” protein, and with the main protease that transforms the early proteins of the virus (pp1a and ppa1b) into the complex responsible for viral replication. The binding energy of hesperidin to these important components is lower than that of lopinavir, ritonavir, and indinavir, suggesting that it could perform an effective antiviral action. Furthermore, both hesperidin and ascorbic acid counteract the cell damaging effects of the oxygen free radicals triggered by virus infection and inflammation. There is discussion about the preventive efficacy of vitamin C, at the dose achievable by the diet, but recent reviews suggest that this substance can be useful in the case of strong immune system burden caused by viral disease. Computational methods and laboratory studies support the need to undertake apposite preclinical, epidemiological, and experimental studies on the potential benefits of citrus fruit components for the prevention of infectious diseases, including COVID-19.
Article
Full-text available
Oxidative stress (OS), triggered by overproduction of reactive oxygen and nitrogen species, is the main mechanism responsible for several human diseases. The available one-target drugs often face such illnesses, by softening symptoms without eradicating the cause. Differently, natural polyphenols from fruits and vegetables possess multi-target abilities for counteracting OS, thus representing promising therapeutic alternatives and adjuvants. Although in several in vitro experiments, ellagitannins (ETs), ellagic acid (EA), and its metabolites urolithins (UROs) have shown similar great potential for the treatment of OS-mediated human diseases, only UROs have demonstrated in vivo the ability to reach tissues to a greater extent, thus appearing as the main molecules responsible for beneficial activities. Unfortunately, UROs production depends on individual metabotypes, and the consequent extreme variability limits their potentiality as novel therapeutics, as well as dietary assumption of EA, EA-enriched functional foods, and food supplements. This review focuses on the pathophysiology of OS; on EA and UROs chemical features and on the mechanisms of their antioxidant activity. A discussion on the clinical applicability of the debated UROs in place of EA and on the effectiveness of EA-enriched products is also included.
Article
Gestational diabetes (GDM) affects about 20 % of pregnancies globally. Defective insulin receptor (IR) signaling has been found in the placenta from patients with GDM, but the underly mechanism is still unclear. In the present study, the mRNA and protein levels of IR-α, insulin receptor substrate 1(IRS-1) and inulin like growth factor 1 receptor (IGF1R) were detected in the placenta tissue samples from 33 GDM patients and 20 healthy controls. Reduced IR-α protein level was observed in both obese and non-obese GDM patients, and decreased IGF1R protein level was found in obese GDM patients. However, the IR-α and IGF1R mRNAs level was not significantly altered in GDM patients. Subsequently, the expression of 10 miRNAs that have the potential targeting IR-α and IGF1R was examined by qRT-PCR in the placenta, and miR-140-3p was found overexpressed. Through dual-luciferase assay and immunoblotting, miR-140-3p was confirmed to suppress IR-α and IGF1R expression via targeting the 3’UTRs. As a treatment candidate, naringenin downregulated miR-140-3p level in trophoblasts and endothelial cells. Meanwhile, IR-α and IGF1R expression was upregulated by naringenin, and the glucose uptake was increased in naringenin treated trophoblasts and endothelial cells. Finally, naringenin upregulated cell viability, migration capacity of HTR-8/SVneo and HUVEC cells, and increased HUVEC cells angiogenesis in high glucose condition. In conclusion, miR-140-3p overexpression contributes to the defective placental IR signaling in patients with GDM. Naringenin treatment protects trophoblasts and endothelial cells from the harmful high glucose environment which have the potential for GDM treatment.
Article
Ethnopharmacological relevance Apigenin is a natural flavonoid compound present in the chamomile (Matricaia chamomilla L.) from the Asteraceae family, which is used in the treatment of cardiovascular diseases by traditional healers, but its effects on differentiation and extracellular matrix (ECM) production of cardiac fibroblasts (CFs) induced by transforming growth factor beta 1 (TGF-β1) are poorly understood. Aim of the study : This study aimed to examine these effects and potential molecular mechanisms and to provide a new application of apigenin in the prevention and trement of cardiac fibrosis. Materials and methods The TGF-β1-stimulated CFs or the combination of TGF-β1-stimulated and microRNA-155-5p (miR-155-5p) inhibitor- or mimic -transfected CFs were treated with or without apigenin. The expression levels of intracellular related mRNA and proteins were detected by real-time polymerase chain reaction and western blot methods, respectively. The luciferase reporter gene containing cellular Sloan-Kettering Institute (c-Ski) wild or mutant type 3’-UTR was used and the luciferase activity was examined to verify the direct link of miR-155-5p and c-Ski. Results After treatment of TGF-β1-stimulated CFs with 6-24 μM apigenin, the expression of c-Ski was increased, while those of miR-155-5p, α-smooth muscle actin, collagen Ⅰ/Ⅲ, Smad2/3, and p-Smad2/3 were decreased. After transfection of CFs with the miR-155-5p inhibitor or mimic, the similar or inverse results were respectively observed as well. The combination of TGF-β1 and miR-155-5p inhibitor or mimic might cause an antagonistical or synergic effect, respectively, and apigenin addition could enhance the effects of the inhibitor and antagonize the effects of the mimic. Luciferase reporter gene assay demonstrated that c-Ski was a direct target of miR-155-5p. Conclusion These findings suggested that apigenin could inhibit the differentiation and ECM production in TGF-β1-stimulated CFs, and its mechanisms might partly be attributable to the reduction of miR-155-5p expression and subsequent increment of c-Ski expression, which might result in the inhibition of Smad2/3 and p-Smad2/3 expressions.
Article
Background: Naringenin has been reported to play an anticancer role by inhibiting the metastasis of tumor cells, but the roles of naringenin and the molecular mechanisms mediated by it in lung cancer remain to be elucidated. Materials and Methods: Cell migration and invasion were determined by transwell assay. The expression levels of circular RNA FOXM1 (circFOXM1), microRNA-3619-5p (miR-3619-5p), and sperm-associated antigen 5 (SPAG5) in lung cancer were measured by quantitative real-time polymerase chain reaction. Western blot assay was used to detect the levels of all proteins. The interaction between microRNA and circular RNA or mRNA was validated using dual-luciferase reporter assay. The models of xenograft mice were established to evaluate the effect of naringenin on tumor growth in vivo. Results: Naringenin restrained migration and invasion of lung cancer cells in a dose-dependent manner. In addition, naringenin could decrease the expression levels of circFOXM1 and SPAG5, which were highly expressed in lung cancer and increase the level of miR-3619-5p with low expression in lung cancer in a dose-dependent manner. miR-3619-5p has an interactive relationship with circFOXM1 or SPAG5, and circFOXM1 could enhance SPAG5 expression by sponging miR-3619-5p in naringenin-treated lung cancer cells. Moreover, naringenin inhibited cell migration, invasion, and tumor growth by regulating circFOXM1/miR-3619-5p/SPAG5 axis in lung cancer. Conclusion: Naringenin hindered cell metastasis in vitro and tumor growth in vivo by reducing circFOXM1 and SPAG5 expression and inducing the expression of miR-3619-5p in lung cancer.