ArticlePDF Available

Mangiferin and Hesperidin Transdermal Distribution and Permeability through the Skin from Solutions and Honeybush Extracts (Cyclopia sp.)—A Comparison Ex Vivo Study

Authors:

Abstract and Figures

Polyphenolic compounds—mangiferin and hesperidin—are, among others, the most important secondary metabolites of African shrub Cyclopia sp. (honeybush). The aim of this study was to compare the percutaneous absorption of mangiferin and hesperidin from solutions (water, ethanol 50%, (v/v)) and extracts obtained from green and fermented honeybush (water, ethanol 50%, (v/v)). Research was performed with the Bronaugh cells, on human dorsal skin. The mangiferin and hesperidin distributions in skin layers (stratum corneum, epidermis, and dermis) and in acceptor fluid (in every 2, 4, 6, and 24 h) were evaluated by HPLC–Photodiode Array Coulometric and Coulometric Electrochemical Array Detection. The transdermal distribution of hesperidin was also demonstrated by fluorescence microscopy. Results indicated that mangiferin and hesperidin were able to cross the stratum corneum and penetrate into the epidermis and dermis. An advantage of hesperidin penetration into the skin from the water over ethanol solution was observed (451.02 ± 14.50 vs. 357.39 ± 4.51 ng/cm2), as well as in the mangiferin study (127.56 ± 9.49 vs. 97.23 ± 2.92 ng/cm2). Furthermore, mangiferin penetration was more evident from nonfermented honeybush ethanol extract (189.85 ± 4.11 ng/cm2) than from solutions. The permeation of mangiferin and hesperidin through the skin to the acceptor fluid was observed regardless of whether the solution or the honeybush extract was applied. The highest ability to permeate the skin was demonstrated for the water solution of hesperidin (250.92 ± 16.01 ng/cm2), while the hesperidin occurring in the extracts permeated in a very low capacity. Mangiferin from nonfermented honeybush ethanol extract had the highest ability to permeate to the acceptor fluid within 24 h (152.36 ± 8.57 ng/cm2).
Content may be subject to copyright.
molecules
Article
Mangiferin and Hesperidin Transdermal Distribution
and Permeability through the Skin from Solutions
and Honeybush Extracts (Cyclopia sp.)—A Comparison
Ex Vivo Study
Anna Hering 1, * , Jadwiga Renata Ochocka 1, *, Helena Baranska 2, Krzysztof Cal 2
and Justyna Stefanowicz-Hajduk 1, *


Citation: Hering, A.; Ochocka, J.R.;
Baranska, H.; Cal, K.;
Stefanowicz-Hajduk, J. Mangiferin
and Hesperidin Transdermal
Distribution and Permeability
through the Skin from Solutions and
Honeybush Extracts (Cyclopia sp.)—A
Comparison Ex Vivo Study. Molecules
2021,26, 6547. https://doi.org/
10.3390/molecules26216547
Academic Editor: Francesco Cacciola
Received: 12 October 2021
Accepted: 26 October 2021
Published: 29 October 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 Biology and Pharmaceutical Botany, Medical University of Gdansk, 80-416 Gdansk, Poland
2Department of Pharmaceutical Technology, Medical University of Gdansk, 80-416 Gdansk, Poland;
helena.baranska@gmail.com (H.B.); krzysztof.cal@gumed.edu.pl (K.C.)
*Correspondence: anna.hering@gumed.edu.pl (A.H.); jadwiga.ochocka@gumed.edu.pl (J.R.O.);
justynastef@gumed.edu.pl (J.S.-H.)
Abstract:
Polyphenolic compounds—mangiferin and hesperidin—are, among others, the most
important secondary metabolites of African shrub Cyclopia sp. (honeybush). The aim of this study
was to compare the percutaneous absorption of mangiferin and hesperidin from solutions (water,
ethanol 50%, (v/v)) and extracts obtained from green and fermented honeybush (water, ethanol 50%,
(v/v)). Research was performed with the Bronaugh cells, on human dorsal skin. The mangiferin and
hesperidin distributions in skin layers (stratum corneum, epidermis, and dermis) and in acceptor fluid
(in every 2, 4, 6, and 24 h) were evaluated by HPLC–Photodiode Array Coulometric and Coulometric
Electrochemical Array Detection. The transdermal distribution of hesperidin was also demonstrated
by fluorescence microscopy. Results indicated that mangiferin and hesperidin were able to cross
the stratum corneum and penetrate into the epidermis and dermis. An advantage of hesperidin
penetration into the skin from the water over ethanol solution was observed
(451.02 ±14.50
vs.
357.39
±
4.51 ng/cm
2
), as well as in the mangiferin study (127.56
±
9.49 vs. 97.23
±
2.92 ng/cm
2
).
Furthermore, mangiferin penetration was more evident from nonfermented honeybush ethanol
extract (189.85
±
4.11 ng/cm
2
) than from solutions. The permeation of mangiferin and hesperidin
through the skin to the acceptor fluid was observed regardless of whether the solution or the
honeybush extract was applied. The highest ability to permeate the skin was demonstrated for the
water solution of hesperidin (250.92
±
16.01 ng/cm
2
), while the hesperidin occurring in the extracts
permeated in a very low capacity. Mangiferin from nonfermented honeybush ethanol extract had the
highest ability to permeate to the acceptor fluid within 24 h (152.36 ±8.57 ng/cm2).
Keywords:
skin penetration; skin permeation; HPLC; Fabaceae; honeybush; fluorescent microscopy
1. Introduction
Natural products used in the form of cosmetics or nutraceuticals nowadays are gaining
importance. They are usually well known in ethnomedicine as a relatively cheap and safe
source of active compounds [
1
]. There are many synthetic products on the market that are
dermatologically effective, though they cause unwanted side-effects. The most commonly
observed are redness, irritation, rashes, and itching. In addition, such products cannot
be often used in the area of injured skin, on mucosa, or in high amounts, due to the toxic
systemic effects [
2
]. Exposure to biotic and abiotic stress generates oxidative stress among
skin macromolecules and, in consequence, their dysfunction or degradation [
3
5
]. Due
to the fact that skin is considered the most external and first barrier of the human body,
its arrangement is essential to maintain internal homeostasis. The first layer—the stratum
corneum, is composed of dead, keratin-filled cells. However, they contain substances
Molecules 2021,26, 6547. https://doi.org/10.3390/molecules26216547 https://www.mdpi.com/journal/molecules
Molecules 2021,26, 6547 2 of 16
with hygroscopic properties, responsible for binding water, and enzymes participating in
metabolic processes, the activity of which is responsible for the acidic pH of the skin surface.
The specific arrangement of the stratum corneum and the hydrophobic properties create
this impenetrable barrier layer to pathogens and foreign substances [
3
,
6
,
7
]. Penetration
through the stratum corneum barrier is the basic parameter in studies of the transdermal
distribution of xenobiotics [
8
,
9
]. With the growing interest in the percutaneous route of
administration of medicinal substances as an alternative to oral intake, intensive research
has been started on increasing the permeability of the stratum corneum layer [
7
]. The main
problems of those studies are: determining whether the introduced substance should target
the dermis or reach the bloodstream; physical and biological properties of a xenobiotic;
the drug partition between cumulation in the skin and permeation through the skin;
individual differences and skin diseases [
9
12
]. Considering polyphenols as almost perfect
antioxidants, which can be used to protect the skin and its macromolecules against oxidative
stress [
13
,
14
], their ability to penetrate into and permeate through skin layers should be
under consideration. The permeability depends, among others, on the structure of the
molecules and their chemical properties; however, the transdermal distribution of these
compounds could be modified by the composition of the delivery system [15,16].
Polyphenols are a group of pharmacologically active, secondary metabolites, widely
spread in nature. It is confirmed that polyphenols have ability to protect skin macro-
molecules from internal and external degradation. Natural sources of specific polyphenolic
compositions combined with high quantitative content are in demand [1].
Cyclopia spp. (Fabaceae) is an endemic shrub growing wild and cultivated on planta-
tions only within Cape Province in the Republic of South Africa. Leaves and branches from
honeybush are collected and dried (green honeybush) or fermented and dried (fermented
honeybush), to prepare the tea beverage with many health properties [
17
]. The richness in
polyphenolic compounds, low tannins, and lack of caffeine have made this sweet-tasting
tea beverage a highly attractive alternative for Camellia sinensis tea [
18
,
19
]. Additionally,
extracts from honeybush are traditionally used by the local people in different skin dis-
eases [
17
]. The tisane is becoming increasingly more popular in commercial use among
consumers, and researchers.
The pharmacological activity of Cyclopia spp. is caused by the synergic effect of
polyphenolic fraction—xanthones, flavanones, flavones, isoflavones, coumestans, pheno-
lic acids, and others [
20
]. Therefore, extracts from honeybush have strong antioxidant
properties with a large amount of mangiferin and hesperidin, compounds well known as
oxidative stress fighters [
17
,
20
26
]. These compounds, among others, are also responsible
for the skin UV protection [
20
,
27
] and inhibition of wrinkle formation [
28
]. In addition,
Bartoszewski et al. indicated that mangiferin and hesperidin are safe for keratinocytes [
29
].
Mangiferin, a C-glucosyl xanthone, is widely distributed in nature. It is presented
abundantly in the families Anacardiaceae [
30
], Liliaceae [
31
], or Fabaceae [
18
]. The many
important healing pharmacological activities of mangiferin include analgesic, immunomod-
ulatory, antibacterial, and antidiabetic [
32
34
]. One of the major properties of mangiferin is
the lowering level of oxidative stress, which promotes in the long-term many degradation
diseases [
35
37
]. Despite numerous
in vitro
analyses confirming mangiferin activity, its
bioavailability is limited by both low solubility and absorption from the intestine [
38
,
39
].
Among the new routes of improving mangiferin bioavailability is topical application. It was
confirmed that mangiferin has the ability to pass through the stratum corneum, penetrate
the epidermis and dermis, and inhibit the degradation of collagen and elastin, as well as
wrinkle formation [
40
,
41
]. Additional results have confirmed mangiferin’s ability to inter-
act with macromolecules of the skin and increase the regenerative capacity of wounded
skin [
42
]. Mangiferin has a high potential in dermatology; however, many aspects of the
compound action in the skin have not yet been investigated.
Hesperidin, a flavanone (hesperetin-7-O-rutinoside), is abundantly presented in cit-
rus fruits [
43
,
44
]. Mostly known from cardiovascular protection and anti-inflammatory
functions, hesperidin has proved significant systemic effects, which are limited by the low
Molecules 2021,26, 6547 3 of 16
water solubility and low absorption from the intestine [
45
51
]. In recent years, derma-
tological studies have focused on hesperidin’s ability to improve the skin functions and
condition [
52
]. The transdermal activity of hesperidin is mainly related to its antioxidant
properties. It has been confirmed that hesperidin catalyses the scarring process through
the reduction of oxidative stress around the wounded area [
52
,
53
], and enhances the ability
to protect keratinocytes against UV-induced damage [5456]. It has also been proven that
hesperidin inhibits the inflammatory reaction generated by keratinocytes when exposed to
oxidative stress [
57
]. In addition, hesperidin, by reducing free radicals generated in the
course of the melanogenesis process, inhibits melanogenesis and melanosome transport,
resulting in skin lightening [
58
60
]. The topical application of hesperidin improved skin
barrier permeability [
61
]. What is important is that the compound has no toxic effects after
neither oral nor topical application, whereas many ingredients present in cosmetic products
may cause allergic reactions and be responsible for adverse systemic effects [
52
,
62
65
].
Until now, animal models or tissue cultures have been used in studies of hesperidin, while
the transdermal distribution of this polyphenol after application on human skin has never
been analysed.
In this study, we estimated the permeation and distribution of mangiferin and hes-
peridin among skin layers ex vivo after the application of two types of solutions (water and
ethanol) and Cyclopia sp. extracts (water and ethanol). The results of Cyclopia sp. extracts,
as well as hesperidin permeation and transdermal distribution, were shown in this work
for the first time.
2. Results
Extracts A, B, C, and D (Table 1), as well as water or ethanol solutions of hesperidin or
mangiferin, were applied on the skin surface. After 2, 4, 6, and 24 h, samples of acceptor
fluid were taken to quantitatively analyse both compounds. The estimation of hesperidin
and mangiferin distribution among skin layers was determined after 24 h of experiment,
followed by skin layers’ separations and extractions. For the quantitative determination of
mangiferin and hesperidin in water and ethanol extracts applied on the skin (summarised
in Table 1), and in solutions extracted from individual layers of the skin, as well as in
acceptor fluids, HPLC analysis with electrochemical detection was utilised.
Table 1.
Summary of content of mangiferin and hesperidin [ng/mL] in Cyclopia sp. extracts analysed
by HPLC-UV.
Cyclopia
sp.
Extract
Type of Plant Material Solvent for the Extracts
Preparation Mangiferin Hesperidin
Anonfermented (“green”) H2O 1593.1 ±31.08 483.6 ±16.74
Bfermented H2O 967.2 ±22.17 376.13 ±7.07
Cnonfermented (“green”) 50% EtOH (v/v) 4695.68 ±65.33 423.98 ±13.01
Dfermented 50% EtOH (v/v) 2962.05 ±62.14 304.5 ±9
The ex vivo experiments revealed that mangiferin [
40
] and hesperidin from solutions
(water or ethanol) or extracts (water or ethanol) are capable of passing through the stratum
corneum and penetrating into the deeper layers of the skin, as well as permeating through
the skin.
2.1. Hesperidin Permeation into the Skin from Solutions and Honeybush Extracts
The ability of hesperidin to cross the skin barrier—the stratum corneum—has been
estimated and demonstrated in Figure 1and Table 2.
Molecules 2021,26, 6547 4 of 16
Table 2.
Accumulation of hesperidin analysed by HPLC-UV and HPLC-EC among skin layers: stratum corneum, epidermis,
and dermis, and hesperidin permeation through the skin after 2, 4, 6, and 24 h (ng/cm
2
) from the application of hesperidin
solutions (HeH2O, HeEtOH) or honeybush extracts (A, B, C, D).
HeH2O HeEtOH Extract A Extract B Extract C Extract D
S.c. 77.74 ±5.44 56.77 ±12.59 59.03 ±0.84 40.21 ±9.9 64.82 ±17.39 48.87 ±8.68
Epidermis 147.9 ±8.35 115.03±0.5 66.56 ±0.9 69.54 ±8.51 68.92 ±11 88.85 ±16.68
Dermis 225.38 ±29.72 185.59 ±0.45 121.85 ±0.56 107.33 ±12.56 80.21 ±12.05 87.44 ±5.78
2 h 83.18 ±12.79 71.33 ±12.38 5.08 ±1.22 6.72 ±1.84 8.43 ±0.83 10.31 ±3.06
4 h 117.18 ±12.79 88.13 ±20.04 8.87 ±2.35 8 ±1.35 11.18 ±0.7 12 ±2.91
6 h 151.08 ±22.35 108.77 ±14.38 10.97 ±0.38 9.13 ±0.38 12.51 ±3.91 14.31 ±2.91
24 h 250.92 ±16.01 132 ±14.93 16.72 ±4.22 13.23 ±4.24 17.79 ±3.84 22.41 ±1.56
S.c. stratum corneum; each average value was obtained from three independent repetitions.
Molecules2021,26,xFORPEERREVIEW4of16
Table2.AccumulationofhesperidinanalysedbyHPLCUVandHPLCECamongskinlayers:stratumcorneum,epider
mis,anddermis,andhesperidinpermeationthroughtheskinafter2,4,6,and24h(ng/cm2)fromtheapplicationofhes
peridinsolutions(HeH2O,HeEtOH)orhoneybushextracts(A,B,C,D).
HeH2OHeEtOHExtractAExtractBExtractCExtractD
S.c.77.74±5.4456.77±12.5959.03±0.8440.21±9.964.82±17.3948.87±8.68
Epidermis147.9±8.35115.03±0.566.56±0.969.54±8.5168.92±1188.85±16.68
Dermis225.38±29.72185.59±0.45121.85±0.56107.33±12.5680.21±12.0587.44±5.78
2h83.18±12.7971.33±12.385.08±1.226.72±1.848.43±0.8310.31±3.06
4h117.18±12.7988.13±20.048.87±2.358±1.3511.18±0.712±2.91
6h151.08±22.35108.77±14.3810.97±0.389.13±0.3812.51±3.9114.31±2.91
24h250.92±16.01132±14.9316.72±4.2213.23±4.2417.79±3.8422.41±1.56
S.c.stratumcorneum;eachaveragevaluewasobtainedfromthreeindependentrepetitions.
Figure1.Hesperidindistributionamongskinlayers:stratumcorneum,epidermis,anddermis(ng/cm2)afterapplication
and24hofincubationwithhesperidinsolutions:water(HeH2O,4μg/mL),ethanol(HeEtOH,4μg/mL,50%(v/v)),or
honeybushextracts:A,B,C,D.Eachaveragevaluewasobtainedfromthreeindependentrepetitions.Errorbarsrepresent
standarddeviations.Significantdifferencesamongsamplesaremarkedwithanasterisk(p<0.05).
Thequantitativeanalysisofhesperidindistributedamongskinlayersafteritsappli
cationfromsolutionsindicatedtheadvantageofhesperidinpermeationbothfromwater
andethanolsolutions.Thehighestamountofhesperidinwasdetectedinthedermisin
thecaseofbothsolutions,anditwasalmostfourtimeshigherthaninthestratum
corneum.Theresultswere225.38±29.72and185.59±0.45ng/cm2forthewaterandetha
nolsolutions,respectively.TheHPLCanalysisalsoshowedthatthehesperidinconcen
trationwasalmosttwotimeshigherintheepidermisthaninthestratumcorneumforboth
thesolutions(Table2).
Theadvantageofhesperidincumulationamongskinlayersaftertheapplicationof
solutionscomparedtohoneybushextracts(A,B,C,andD)wasevidenced.Theamount
ofhesperidininthestratumcorneum,epidermis,anddermiswashigherforwaterthan
forethanolsolution.
Intheexperimentswiththeextracts,thelowestamountofhesperidinwasforethanol
extractBinthestratumcorneum.Theconcentrationofhesperidinintheepidermisafter
honeybushextractswereappliedontheskinsurfacewascomparablewithaslightpre
dominanceofhesperidincumulationfromextractD.Amongthedermis,thedifferences
inhesperidincumulationweremorevisible:thelowesthesperidinconcentrationwas
Figure 1.
Hesperidin distribution among skin layers: stratum corneum, epidermis, and dermis (ng/cm
2
) after application
and 24 h of incubation with hesperidin solutions: water (HeH
2
O, 4
µ
g/mL), ethanol (HeEtOH, 4
µ
g/mL, 50% (v/v)), or
honeybush extracts: A, B, C, D. Each average value was obtained from three independent repetitions. Error bars represent
standard deviations. Significant differences among samples are marked with an asterisk (p< 0.05).
The quantitative analysis of hesperidin distributed among skin layers after its applica-
tion from solutions indicated the advantage of hesperidin permeation both from water and
ethanol solutions. The highest amount of hesperidin was detected in the dermis in the case
of both solutions, and it was almost four times higher than in the stratum corneum. The
results were 225.38
±
29.72 and 185.59
±
0.45 ng/cm
2
for the water and ethanol solutions,
respectively. The HPLC analysis also showed that the hesperidin concentration was almost
two times higher in the epidermis than in the stratum corneum for both the solutions
(Table 2).
The advantage of hesperidin cumulation among skin layers after the application of
solutions compared to honeybush extracts (A, B, C, and D) was evidenced. The amount of
hesperidin in the stratum corneum, epidermis, and dermis was higher for water than for
ethanol solution.
In the experiments with the extracts, the lowest amount of hesperidin was for ethanol
extract B in the stratum corneum. The concentration of hesperidin in the epidermis
after honeybush extracts were applied on the skin surface was comparable with a slight
predominance of hesperidin cumulation from extract D. Among the dermis, the differences
in hesperidin cumulation were more visible: the lowest hesperidin concentration was
detected for extracts C and D, whereas the hesperidin cumulation after the application of
Molecules 2021,26, 6547 5 of 16
extracts A and B exceeded 100 ng/cm
2
. Both for the solutions and extracts, the highest
concentration of hesperidin was detected in the dermis (Figure 1and Table 2).
2.2. Hesperidin Permeability through the Skin from Solutions and Honeybush Extracts
HPLC analysis revealed hesperidin’s ability to permeate through the skin to the
acceptor fluid (Figure 2).
Molecules2021,26,xFORPEERREVIEW5of16
detectedforextractsCandD,whereasthehesperidincumulationaftertheapplicationof
extractsAandBexceeded100ng/cm2.Bothforthesolutionsandextracts,thehighest
concentrationofhesperidinwasdetectedinthedermis(Figure1andTable2).
2.2.HesperidinPermeabilitythroughtheSkinfromSolutionsandHoneybushExtracts
HPLCanalysisrevealedhesperidin’sabilitytopermeatethroughtheskintotheac
ceptorfluid(Figure2).
Figure2.Hesperidinpermeationthroughtheskinafter2,4,6,and24h(ng/cm2)fromtheapplication
ofhesperidinsolutions:water(HeH2O,4μg/mL),ethanol(HeEtOH,4μg/mL,50%(v/v)),orhoney
bushextracts:A,B,C,D.
Thepermeationofhesperidinafterapplicationontheskinsurfaceofhesperidinso
lutions—(HeH2O,HeEtOH)—wassimilarandwithinthelimitsofthestatisticalerrorin
thefirst2h.Thequantifiedanalysisshowedahighamountofhesperidinpresentedin
acceptorfluidafter2hfromapplication,whichindicatedtherelativelyquickpenetration
ofthecompoundthroughtheskin.Afurtherincreaseintheamountofhesperidinwas
observedafter4and6h.After24h,theHPLCanalysisshowedanamountofhesperidin
inthefluidatleasttwicehighercomparedtotheamountdeterminedafter2h(Table2).
Hesperidinpermeationthroughtheskin,aftertheapplicationofhoneybushextracts
ontheskinsurface,wasalsoobserved,anditincreasedduringthetimeofincubation.
After24hofhesperidin,theamountintheacceptorfluid(calculatedoncm2oftheskin)
washighestforextractD(22.41±1.56ng/cm2).Fortheremainingextracts,theobtained
amountsofthecompoundinthefluiddecreasedasfollows:C>A>B.
2.3.TransdermalDistributionofHesperidinwiththeUseofFluorescenceMicroscopy
Fluorescencemicroscopywasutilisedforconfirmationofthetransdermaldistribu
tionofhesperidinafter24hofincubationoftheethanolsolutiononthehumanskinsur
faceexvivo.Followedbycompletionofthepermeationexperiment,transverseskincuts
weremade.Formicroscopicanalysis,onlythepiecesofskinwithallvisiblelayersofthe
skinwereused.Freshlytakencrosscuttingsofskinwereanalysedwithafluorescence
microscope(NikonEclipse50,filterUV2A,Ex330–380nm).Theresultsintheformof
photographsarepresentedinFigure3.Comparativeobservationoftheskinwithpure
Figure 2.
Hesperidin permeation through the skin after 2, 4, 6, and 24 h (ng/cm
2
) from the application
of hesperidin solutions: water (HeH
2
O, 4
µ
g/mL), ethanol (HeEtOH, 4
µ
g/mL, 50% (v/v)), or
honeybush extracts: A, B, C, D.
The permeation of hesperidin after application on the skin surface of hesperidin
solutions—(HeH
2
O, HeEtOH)—was similar and within the limits of the statistical error
in the first 2 h. The quantified analysis showed a high amount of hesperidin presented in
acceptor fluid after 2 h from application, which indicated the relatively quick penetration
of the compound through the skin. A further increase in the amount of hesperidin was
observed after 4 and 6 h. After 24 h, the HPLC analysis showed an amount of hesperidin
in the fluid at least twice higher compared to the amount determined after 2 h (Table 2).
Hesperidin permeation through the skin, after the application of honeybush extracts
on the skin surface, was also observed, and it increased during the time of incubation.
After 24 h of hesperidin, the amount in the acceptor fluid (calculated on cm
2
of the skin)
was highest for extract D (22.41
±
1.56 ng/cm
2
). For the remaining extracts, the obtained
amounts of the compound in the fluid decreased as follows: C > A > B.
2.3. Transdermal Distribution of Hesperidin with the Use of Fluorescence Microscopy
Fluorescence microscopy was utilised for confirmation of the transdermal distribution
of hesperidin after 24 h of incubation of the ethanol solution on the human skin surface
ex vivo. Followed by completion of the permeation experiment, transverse skin cuts
were made. For microscopic analysis, only the pieces of skin with all visible layers of the
skin were used. Freshly taken cross-cuttings of skin were analysed with a fluorescence
microscope (Nikon Eclipse 50, filter UV 2A, Ex 330–380 nm). The results in the form of
photographs are presented in Figure 3. Comparative observation of the skin with pure
ethanol (control, Figure 3A) and ethanolic hesperidin solution applied on the skin showed
a strong, bright yellow-green fluorescence in the analysed sample (Figure 3B). According
Molecules 2021,26, 6547 6 of 16
to the literature data, the observed fluorescence colour is characteristic for hesperidin [
66
].
The intensity of fluorescence was low in the area of the stratum corneum and epidermis,
while the dermis layer showed clearly visible yellow-green fluorescence. In this image
(Figure 3B), the strong fluorescence presented on the external surface of the skin indicated
that some part of hesperidin did not penetrate into the stratum corneum, and it was not
completely washed away after the end of the experiment. This hesperidin was probably
deposited on the surface of the skin.
Molecules2021,26,xFORPEERREVIEW6of16
ethanol(control,Figure3A)andethanolichesperidinsolutionappliedontheskinshowed
astrong,brightyellowgreenfluorescenceintheanalysedsample(Figure3B).According
totheliteraturedata,theobservedfluorescencecolourischaracteristicforhesperidin[66].
Theintensityoffluorescencewaslowintheareaofthestratumcorneumandepidermis,
whilethedermislayershowedclearlyvisibleyellowgreenfluorescence.Inthisimage
(Figure3B),thestrongfluorescencepresentedontheexternalsurfaceoftheskinindicated
thatsomepartofhesperidindidnotpenetrateintothestratumcorneum,anditwasnot
completelywashedawayaftertheendoftheexperiment.Thishesperidinwasprobably
depositedonthesurfaceoftheskin.
Figure3.Hesperidindistributioninthehumanskin(A—thecontrol,ethanol96%alone,B—ethanolhesperidinsolution
96%,0.01mg/mL).Ethanolandhesperidinsolutionswereappliedonthehumanskinexvivo.After24hofincubation,
skinwaswashedinwaterandanalysedunderafluorescencemicroscope,NikonEclipse50,filterUV2A,Ex330–380nm.
Thecomplexstructureoftheskinanditsmacromoleculesdetermineitsautolumi
nescence,whichisvisibleintheleftimage(Figure3A,skinsampleonwhichonlyethanol
wasapplied,controlsample)[67].Theobtainedpicturesindicatethathesperidinenhances
skinfluorescence.
Thefluorescencemicroscopyresultsconfirmthequantitativeanalysisofthetrans
dermaldistributionofhesperidinafteritsapplicationandincubationontheskinsurface
(Table2).Thefluorescencewasnoticeableespeciallyintheareaofthedermiswhere,ac
cordingtotheresultspresentedinFigure1,hesperidinunderwentthehighestabsorption.
However,accordingtoXieetal.,hesperetin,theaglyconofhesperidin,interactswithpro
teins,affectingwithslightfluorescencethequenchingofmacromolecules[68].Aninter
actioncouldbecausedbybothhydrophobicandelectrostaticbondsbetweenproteinsand
hesperetin.Itwasindicatedthathydroxylgroupsarethemostimportantintheproton
transversion[69].Hesperetin,whichinteractswithproteins,canbestoredandfloated
withthem[68].Ourcomparisondataofthecontrolandintenseyellowgreenfluorescence
presentedintherightpicture(Figure3B)suggestthatnonbondedhesperidinwasrespon
sibleforthebrightfluorescence.Aprobablyunknownamountofhesperidininteracted
withmacromoleculesoftheepidermisordermis,resultinginquenching.However,the
amountofnoninteractedhesperidinwasstillenoughtogivevisiblefluorescenceinthe
dermis.ThefreehesperidinpresentedinthedermislayerisneededtoprotectECMmac
romoleculesfrominternalandexternaloxidativestress[14].

Figure 3.
Hesperidin distribution in the human skin (
A
—the control, ethanol 96% alone,
B
—ethanol hesperidin solution
96%, 0.01 mg/mL). Ethanol and hesperidin solutions were applied on the human skin ex vivo. After 24 h of incubation, skin
was washed in water and analysed under a fluorescence microscope, Nikon Eclipse 50, filter UV 2A, Ex 330–380 nm.
The complex structure of the skin and its macromolecules determine its auto-luminescence,
which is visible in the left image (Figure 3A, skin sample on which only ethanol was applied,
control sample) [67]. The obtained pictures indicate that hesperidin enhances skin fluorescence.
The fluorescence microscopy results confirm the quantitative analysis of the trans-
dermal distribution of hesperidin after its application and incubation on the skin surface
(Table 2)
. The fluorescence was noticeable especially in the area of the dermis where, ac-
cording to the results presented in Figure 1, hesperidin underwent the highest absorption.
However, according to Xie et al., hesperetin, the aglycon of hesperidin, interacts with
proteins, affecting with slight fluorescence the quenching of macromolecules [
68
]. An
interaction could be caused by both hydrophobic and electrostatic bonds between proteins
and hesperetin. It was indicated that hydroxyl groups are the most important in the proton
transversion [
69
]. Hesperetin, which interacts with proteins, can be stored and floated with
them [
68
]. Our comparison data of the control and intense yellow-green fluorescence pre-
sented in the right picture (Figure 3B) suggest that nonbonded hesperidin was responsible
for the bright fluorescence. A probably unknown amount of hesperidin interacted with
macromolecules of the epidermis or dermis, resulting in quenching. However, the amount
of noninteracted hesperidin was still enough to give visible fluorescence in the dermis. The
free hesperidin presented in the dermis layer is needed to protect ECM macromolecules
from internal and external oxidative stress [14].
2.4. Comparison of Mangiferin Permeation into the Skin and Permeability through the Skin from
Solutions and Honeybush Extracts
The ability of mangiferin to cross the skin stratum corneum barrier was estimated
and demonstrated in our previous paper [
40
] where we analysed both the mangiferin
distribution among skin layers and the permeation through the skin to the acceptor fluid,
after application of mangiferin solutions (MH
2
OH, MEtOH) on the skin surface. In this
Molecules 2021,26, 6547 7 of 16
study, we additionally estimated the distribution and permeation of the honeybush extracts
(A, B, C, D) through the skin.
HPLC analysis with electrochemical detection indicated that mangiferin is capable
of entering into and crossing through the stratum corneum, after the application of both
mangiferin solutions and honeybush extracts (Table 3and Figure 4).
Table 3.
Accumulation of mangiferin analysed by HPLC-UV among skin layers: stratum corneum, epidermis, and dermis,
and mangiferin permeation through the skin after 2, 4, 6, and 24 h (ng/cm
2
) from the application of mangiferin solutions
(MH2O, MEtOH) or honeybush extracts (A, B, C, D).
MH2O MEtOH Extract A Extract B Extract C Extract D
S.c. 32.62 ±4.61 29.92 ±2.07 24.46 ±2.36 29.44 ±7.51 56.92 ±3.72 26.92 ±1.53
Epidermis 41.62 ±7.45 32.08 ±1.97 32.74 ±2.45 48.51 ±7.08 74.26 ±5.95 32.72 ±2.74
Dermis 53.32 ±16.41 35.23 ±4.73 23.95 ±2.15 29.13 ±1,84 58.67 ±2.67 24.1 ±1.97
2 h 30.23 ±5.22 48.85 ±7.37 30.97 ±1.54 33.85 ±5.31 61.03 ±5.05 34.1 ±7.44
4 h 35.38 ±5.9 59.08 ±6.76 39.95 ±1.72 42.31 ±8.18 82.36 ±7.27 43.28 ±6.74
6 h 40.54 ±7.77 69.15 ±7 49.23 ±2.66 50.92 ±7.52 111.54 ±9.39 56.56 ±4.66
24 h 66.15 ±11.92 77.78 ±8.38 59.33 ±3.76 62.82 ±9.46 152.36 ±8.57 75.69 ±2.14
S.c. stratum corneum; each average value was obtained from three independent repetitions.
Molecules2021,26,xFORPEERREVIEW7of16
2.4.ComparisonofMangiferinPermeationintotheSkinandPermeabilitythroughtheSkinfrom
SolutionsandHoneybushExtracts
Theabilityofmangiferintocrosstheskinstratumcorneumbarrierwasestimated
anddemonstratedinourpreviouspaper[40]whereweanalysedboththemangiferindis
tributionamongskinlayersandthepermeationthroughtheskintotheacceptorfluid,
afterapplicationofmangiferinsolutions(MH2OH,MEtOH)ontheskinsurface.Inthis
study,weadditionallyestimatedthedistributionandpermeationofthehoneybushex
tracts(A,B,C,D)throughtheskin.
HPLCanalysiswithelectrochemicaldetectionindicatedthatmangiferiniscapableof
enteringintoandcrossingthroughthestratumcorneum,aftertheapplicationofboth
mangiferinsolutionsandhoneybushextracts(Table3andFigure4).
Table3.AccumulationofmangiferinanalysedbyHPLCUVamongskinlayers:stratumcorneum,epidermis,anddermis,
andmangiferinpermeationthroughtheskinafter2,4,6,and24h(ng/cm2)fromtheapplicationofmangiferinsolutions
(MH2O,MEtOH)orhoneybushextracts(A,B,C,D).
MH2OMEtOHExtractAExtractBExtractCExtractD
S.c.32.62±4.6129.92±2.0724.46±2.3629.44±7.5156.92±3.7226.92±1.53
Epidermis41.62±7.4532.08±1.9732.74±2.4548.51±7.0874.26±5.9532.72±2.74
Dermis53.32±16.4135.23±4.7323.95±2.1529.13±1,8458.67±2.6724.1±1.97
2h30.23±5.2248.85±7.3730.97±1.5433.85±5.3161.03±5.0534.1±7.44
4h35.38±5.959.08±6.7639.95±1.7242.31±8.1882.36±7.2743.28±6.74
6h40.54±7.7769.15±749.23±2.6650.92±7.52111.54±9.3956.56±4.66
24h66.15±11.9277.78±8.3859.33±3.7662.82±9.46152.36±8.5775.69±2.14
S.c.stratumcorneum;eachaveragevaluewasobtainedfromthreeindependentrepetitions.
Figure4.Mangiferindistributionamongskinlayers:stratumcorneum,epidermis,anddermis(ng/cm2)afterapplication
and24hofincubationofmangiferinsolutions:water(MH2O,25μg/mL),ethanol(25μg/mL,MEtOH,50%(v/v)),orhon
eybushextracts:A,B,C,D.Eachaveragevaluewasobtainedfromthreeindependentrepetitions.Errorbarsrepresent
standarddeviations.Significantdifferencesamongsamplesaremarkedwithanasterisk(p<0.05).
Thehighestamountofmangiferinthatenteredintoandpermeatedthroughthestra
tumcorneumispresentedforextractC,whichisthemostpromisingsourceofmangiferin
forskinpermeation.Thecompoundaccumulationwashighestintheepidermis,while,in
thestratumcorneumanddermis,theamountofdeterminedmangiferinwascomparable
(Table3).Thesecondsignificantamountofmangiferinintransdermaldistributionwas
Figure 4. Mangiferin distribution among skin layers: stratum corneum, epidermis, and dermis (ng/cm2) after application
and 24 h of incubation of mangiferin solutions: water (MH
2
O, 25
µ
g/mL), ethanol (25
µ
g/mL, MEtOH, 50% (v/v)), or
honeybush extracts: A, B, C, D. Each average value was obtained from three independent repetitions. Error bars represent
standard deviations. Significant differences among samples are marked with an asterisk (p< 0.05).
The highest amount of mangiferin that entered into and permeated through the stra-
tum corneum is presented for extract C, which is the most promising source of mangiferin
for skin permeation. The compound accumulation was highest in the epidermis, while, in
the stratum corneum and dermis, the amount of determined mangiferin was comparable
(Table 3). The second significant amount of mangiferin in transdermal distribution was
observed after the application of MH
2
O (Table 3and Figure 4). In this case, the mangiferin
cumulation was highest in the dermis.
Molecules 2021,26, 6547 8 of 16
The concentration of mangiferin detected in the stratum corneum was similar to each
other with only slight differences: MH
2
O > MEtOH > B > D > A (Table 3). However,
the highest amount was detected for extract C. In the epidermis, the lowest concentra-
tion of mangiferin was estimated for extracts A and D and ethanol solution MEtOH,
while mangiferin from extract B and MH
2
O indicated a more significant accumulation of
the compound. In the dermis, differences in mangiferin concentration were as follows:
C > MH
2
O > MEtOH > B > D = A. The lowest accumulation of mangiferin was detected
among skin layers after the application of extracts A and D.
The mangiferin permeation through the skin from all analysed solutions and honey-
bush extracts was constant and occurred relatively quickly (Figure 5). The most effective
permeation of mangiferin into the acceptor fluid, during the 24 h of analysis, was observed
for extract C, while the least was for MH
2
O, extracts A and B. The mangiferin permeation
process for extracts A and B was similar, and a slight increase was observed for the MEtOH
solution and extract D.
Molecules2021,26,xFORPEERREVIEW8of16
observedaftertheapplicationofMH2O(Table3andFigure4).Inthiscase,themangiferin
cumulationwashighestinthedermis.
Theconcentrationofmangiferindetectedinthestratumcorneumwassimilartoeach
otherwithonlyslightdifferences:MH2O>MEtOH>B>D>A(Table3).However,the
highestamountwasdetectedforextractC.Intheepidermis,thelowestconcentrationof
mangiferinwasestimatedforextractsAandDandethanolsolutionMEtOH,whileman
giferinfromextractBandMH2Oindicatedamoresignificantaccumulationofthecom
pound.Inthedermis,differencesinmangiferinconcentrationwereasfollows:C>MH2O
>MEtOH>B>D=A.Thelowestaccumulationofmangiferinwasdetectedamongskin
layersaftertheapplicationofextractsAandD.
Themangiferinpermeationthroughtheskinfromallanalysedsolutionsandhoney
bushextractswasconstantandoccurredrelativelyquickly(Figure5).Themosteffective
permeationofmangiferinintotheacceptorfluid,duringthe24hofanalysis,wasobserved
forextractC,whiletheleastwasforMH2O,extractsAandB.Themangiferinpermeation
processforextractsAandBwassimilar,andaslightincreasewasobservedfortheME
tOHsolutionandextractD.
Figure5.Mangiferinpermeationthroughtheskinafter2,4,6,and24h(ng/cm2)fromtheapplicationofmangiferinsolu
tions:water(MH2O,25μg/mL),ethanol(MEtOH,25μg/mL,50%(v/v)),orhoneybushextracts:A,B,C,D.
3.Discussion
Thefirststageintheprocessofxenobioticpenetrationintotheskinandpermeation
throughtheskinisovercomingthestratumcorneumbarrier,whichistreatedasasemi
permeablemembrane.Bothanalysedcompoundsinourstudy—mangiferinandhesperi
din—havelogPbetween1and3(hesperidin:1.78,mangiferin:2.73);themangiferinmolar
massdoesnotexceed500Da(422.34g/mol),thoughthehesperidinmolarmasishigher
withthevalueof610.19g/mol.Thissuggeststhatmangiferinandhesperidinshouldbe
abletopassthestratumcorneum[12,70,71].Thelimitingfactorforbothpolyphenolscould
betheirlowwatersolubilityandabranchedstructure[72,73].Ouranalysisrevealedthe
abilityofmangiferinandhesperidintopermeatethestratumcorneumlayerfromboth
honeybushextractsandsolutions,withapredominanceofhesperidinfromthewaterso
lutionandmangiferinfromtheethanolextractofgreenhoneybush(extractC).Bothcom
poundspresentedarelativelyfastabilitytopermeatethroughtheskin,especiallyfrom
appliedsolutionsforhesperidinandextractCformangiferin.Thesesolutionsandthe
extractappliedontheskinexhibitedthehighestcumulationofanalysedcompounds
Figure 5.
Mangiferin permeation through the skin after 2, 4, 6, and 24 h (ng/cm
2
) from the application of mangiferin
solutions: water (MH2O, 25 µg/mL), ethanol (MEtOH, 25 µg/mL, 50% (v/v)), or honeybush extracts: A, B, C, D.
3. Discussion
The first stage in the process of xenobiotic penetration into the skin and perme-
ation through the skin is overcoming the stratum corneum barrier, which is treated as
a semi-permeable membrane. Both analysed compounds in our study—mangiferin and
hesperidin—have logP between 1 and 3 (hesperidin: 1.78, mangiferin: 2.73); the mangiferin
molar mass does not exceed 500 Da (422.34 g/mol), though the hesperidin molar mas is
higher with the value of 610.19 g/mol. This suggests that mangiferin and hesperidin should
be able to pass the stratum corneum [
12
,
70
,
71
]. The limiting factor for both polyphenols
could be their low water solubility and a branched structure [
72
,
73
]. Our analysis revealed
the ability of mangiferin and hesperidin to permeate the stratum corneum layer from
both honeybush extracts and solutions, with a predominance of hesperidin from the water
solution and mangiferin from the ethanol extract of green honeybush (extract C). Both
compounds presented a relatively fast ability to permeate through the skin, especially from
applied solutions for hesperidin and extract C for mangiferin. These solutions and the ex-
tract applied on the skin exhibited the highest cumulation of analysed compounds among
skin layers, which make them the most promising sources of mangiferin and hesperidin
release into the skin and through the skin.
Molecules 2021,26, 6547 9 of 16
Surprisingly, the lowest hesperidin amount from both solutions was accumulated in
the stratum corneum, and the highest amount was detected in the dermis. Our analysis
showed that hesperidin sourced from the water solution penetrated into the skin and
permeated through the skin with an advantage over hesperidin from the ethanol solution.
These data could indicate that for hesperidin, ethanol is not a sorption promoter [
74
].
Despite the lower hesperidin solubility in water [
72
], a higher amount of this polyphenol
can permeate to the dermis after the application of a water solution on the skin. The dermis,
as the largest layer of the skin, is composed of sensitive to oxidative stress proteins, and
the presence of hesperidin, a compound with strong antioxidant properties, can work as
effective protection against macromolecules [52].
The confirmation of hesperidin cumulation among skin layers and the interaction
of hesperidin molecules was observed with fluorescence microscopy. The experiment
confirmed that a lower yellow-green fluorescence assigned to hesperidin could be seen
in the stratum corneum and epidermis, while the distribution of the xenobiotic in the
dermis was brighter. The intensity of fluorescence in the epidermis and dermis after
hesperidin application proved the presence of free hesperidin, which did not interact with
skin macromolecules [
68
]. The permanent hesperidin interaction with molecules of the
dermis extracellular matrix (ECM) was generally slight, though the mechanism of the
protection can differ from the conventional antioxidant trail [
75
77
]. Nevertheless, the
experiment was carried out ex vivo, and it should be emphasised that in a living organism,
the partition of free hesperidin and that associated with macromolecules could be different.
The ECM is the largest element of healthy skin and is responsible for homeostasis and the
preservation of the mechanical and biological properties of the skin. It is highly complex
system, the protection of which by antioxidants is thus essential [21,68].
It should be emphasised that fluorescence techniques are used to determine transder-
mal drug uptake, though they could be utilised only in the case of molecules with fluores-
cence properties that have different wavelengths than skin macromolecules [
40
,
67
,
78
,
79
].
As it was indicated in our research, hesperidin with its fluorescence properties can be
successfully analysed with these techniques [
66
]. However, the visualisation of the trans-
dermal distribution of hesperidin from honeybush extracts was impossible due to a large
number of different polyphenols with fluorescence abilities presented in the extracts [17].
The present study also included the analysis of hesperidin permeation from the skin
after the application of hesperidin solutions during the 24 h. The research indicated
that hesperidin occurred in the acceptor fluid relatively quickly, especially after the first
two hours. During the following hours, hesperidin permeation was constantly occurring
despite the solution used, with the advantage of water solution. The differences between the
permeability of hesperidin derived from water and ethanol solution were seen especially
after 24 h of the analysis. The experimental data obtained in this part confirmed that
hesperidin from water solution has a better ability to penetrate into the skin and permeate
through the skin than hesperidin can from ethanol solution.
We also analysed hesperidin permeation to the skin and through the skin after appli-
cation of the honeybush extracts on the skin surface. The experiments showed the best
accumulation of hesperidin in the dermis, also from the extracts obtained with water. These
results indicate that, regardless of the solution or extracts applied on the skin, hesperidin
permeates in a higher amount to the dermis from the water carrier. There is also a slight
advantage in the penetration into the skin of hesperidin from water-unfermented extracts
compared to fermented honeybush (A > B).
A major disadvantage of using plant material is the high variability in chemical
composition and antioxidant properties, which depends not only on the changing envi-
ronmental conditions, but also on the time of harvest, as well as storage and transport
conditions [
20
,
80
82
]. If we consider polyphenolic fractions as a source of antioxidants
in the skin layers, it is important to estimate “competition” and “enhancer” properties of
chemicals [15,16]. Honeybush extracts contain many biologically active molecules, which
could potentially improve or inhibit the penetration of the most quantitative compounds
Molecules 2021,26, 6547 10 of 16
into the skin: mangiferin and hesperidin [
83
]. In the case of both compounds, no compari-
son analysis of the cumulation into the skin or permeation through the skin from solutions
and plant extracts has been performed so far.
Despite the applied solutions or honeybush extracts on the skin surface, mangiferin
presented a lower ability than hesperidin to penetrate into the skin. Ethanol extract from
the green honeybush (extract C) applied on the human skin surface showed the highest
mangiferin penetration into the skin and the fastest ability to permeate to the acceptor
fluid during the 24 h of conducted analysis. There was also no evidence of an advantage
in accumulation and permeation studies of mangiferin from solutions than mangiferin
from honeybush extracts. The water solution of mangiferin, similar to hesperidin solution,
indicated a higher ability for transdermal distribution than ethanol solution. Our study
showed that mangiferin has a limited ability to pass the stratum corneum barrier and to
obtain higher concentrations among skin layers.
However, the above results of mangiferin permeation from honeybush extracts (espe-
cially from nonfermented ethanol—C) are generally promising, especially considering the
fact that more mangiferin was absorbed from the ethanol extract than from solutions with
higher concentrations of the compound. The obtained data of mangiferin permeation do
not give an unambiguous answer that ethanol could be a sorption promoter for mangiferin
delivered into the skin from honeybush extracts. The synergism of biological activity of
polyphenolic compounds in this plant species may be responsible for this effect [
84
,
85
].
In addition, the strengthening action of hesperidin on mangiferin has been previously
observed [29,83].
In conclusion, mangiferin and hesperidin were detected in all skin layers and in the
acceptor fluid in the form of both solutions and extracts applied on the human skin surface.
The analysis revealed the predominance of the transdermal cumulation of hesperidin over
mangiferin. On the other hand, permeation through the skin was better for mangiferin
than for hesperidin. Our ex vivo study on mangiferin showed that its permeation into
and through the skin is significantly higher for the ethanol-nonfermented extract than
for solutions. The hesperidin transdermal distribution and permeation study on Cyclopia
sp. extracts are described in this work for the first time. The results clearly indicated
that hesperidin solutions and mangiferin, originating from the ethanol extract of “green”
honeybush, can be a source of transdermal release of the analysed molecules, which have
strong protective and regenerative properties in the skin. The compounds may be used as
antioxidants in dermatology and in topical ways of drug administration, especially due to
the limited oral absorption of both polyphenols.
4. Materials and Methods
4.1. Materials
Mangiferin, hesperidin, and sodium azide were purchased from Sigma Chemical Co.
(St. Louis, MO, USA). HPLC-grade methanol and ethanol were purchased from P.O.Ch.
(Gliwice, Poland).
4.2. Plant Material and Extract Preparation
Plant material was composed of dried leaves and branches declared as the mixtures
of different honeybush species (Cyclopia sp.). Nonfermented (“green”) Cyclopia sp. material
was purchased from Ukajali Marcin Majka Co., (Krakow, Poland) for preparation of extracts
A and C. Fermented Cyclopia sp. plant material was purchased from KAWON-HURT Co.,
(Gostyn, Poland) for preparation of extracts B and D. Plant material and used solvents
are summarised in Table 1. Representative samples of each plant material were deposited
at the Department of Biology and Pharmaceutical Botany, Medical University of Gdansk,
Poland. Extract preparation: 15 g of each plant material was mechanically homogenised
to obtain a particle size < 2.0 mm. Extraction with 100 mL of water or ethanol (50%, v/v)
was performed three times with the use of an ultrasonic water bath (50 Hz) for 30 min at
90
C. After filtration through Whattman filter paper (390
µ
m pore size), obtained extracts
Molecules 2021,26, 6547 11 of 16
were evaporated under vacuum at 60
C. The dry residue was redissolved in either water
or 50% (v/v) ethanol to the final concentration of 30 mg/mL. For the quantitative analysis
of mangiferin and hesperidin in the resulting extracts, HPLC with Photodiode Array
Coulometric and Coulometric Electrochemical Array Detection was used.
4.3. Penetration and Permeation Studies
The studies were performed with the use of human cadaver full skin. Before the
experiment, the skin was collected from the region of the thorax of six 40–60-years-old
donors, and stored frozen at
20
C. The solutions and extracts were applied as an
infinite dose: mangiferin and hesperidin, and ethanol (50%, v/v) and water solutions at
concentrations of 25
µ
g/mL and 4
µ
g/mL, respectively; and extracts A–D at a concentration
of 30 mg/mL. The ethanol at a concentration of 50% (v/v) was applied on the skin as a
blank control. The skin (thickness—0.75 mm; diffusion area—0.65 cm
2
) was placed in
the flow-through Bronaugh diffusion cell apparatus (Figure 6). The static diffusion cell is
composed of two chambers: donor and acceptor. Between these chambers, a part of skin
is placed. The analysed sample is applied on the upper skin surface and the dermis is in
constant contact with homogenised and circulated acceptor fluid. The permeant from the
skin can be taken to the analysis at a specific time period.
Molecules2021,26,xFORPEERREVIEW11of16
ofGdansk,Poland.Extractpreparation:15gofeachplantmaterialwasmechanicallyho
mogenisedtoobtainaparticlesize<2.0mm.Extractionwith100mLofwaterorethanol
(50%,v/v)wasperformedthreetimeswiththeuseofanultrasonicwaterbath(50Hz)for
30minat90°C.AfterfiltrationthroughWhattmanfilterpaper(390μmporesize),ob
tainedextractswereevaporatedundervacuumat60°C.Thedryresiduewasredissolved
ineitherwateror50%(v/v)ethanoltothefinalconcentrationof30mg/mL.Forthequan
titativeanalysisofmangiferinandhesperidinintheresultingextracts,HPLCwithPhoto
diodeArrayCoulometricandCoulometricElectrochemicalArrayDetectionwasused.
4.3.PenetrationandPermeationStudies
Thestudieswereperformedwiththeuseofhumancadaverfullskin.Beforetheex
periment,theskinwascollectedfromtheregionofthethoraxofsix40–60yearsolddo
nors,andstoredfrozenat−20°C.Thesolutionsandextractswereappliedasaninfinite
dose:mangiferinandhesperidin,andethanol(50%,v/v)andwatersolutionsatconcentra
tionsof25μg/mLand4μg/mL,respectively;andextractsA–Dataconcentrationof30
mg/mL.Theethanolataconcentrationof50%(v/v)wasappliedontheskinasablank
control.Theskin(thickness—0.75mm;diffusionarea—0.65cm
2
)wasplacedintheflow
throughBronaughdiffusioncellapparatus(Figure6).Thestaticdiffusioncelliscomposed
oftwochambers:donorandacceptor.Betweenthesechambers,apartofskinisplaced.
Theanalysedsampleisappliedontheupperskinsurfaceandthedermisisinconstant
contactwithhomogenisedandcirculatedacceptorfluid.Thepermeantfromtheskincan
betakentotheanalysisataspecifictimeperiod.
Figure6.Diffusioncellapparatus.
Salinesolution,20mL(with0.005%sodiumazide),wasrecirculatedbeneaththeskin
withaconstantrateof10mL/h.Acceptorfluidensuredthesinkcondition.Solutionsor
extractswereappliedonthesurfaceoftheskinandleftfor24h.Thechambersystemwas
incubatedatthetemperatureof37°C±0.5°C.After2,4,6,and24hfromtheapplication
ofpenetrants,samplesofacceptorfluidwerecollected.Beforeskinlayers’disconnection,
thepenetrantswereremovedfromtheskin.Thestratumcorneumwasseparatedbythe
tapestrippingmethod,using30fragmentsofa3madhesivetapewiththefollowingpa
rameters:pressure~1kg/cm
2
(appliedbystamp),2sdurationofpressure,andarapid
removalrateatanangleof45°.Theepidermisanddermiswereisolatedbytheheatsep
arationtechnique[86].Thewholeskinwasimmersedinwaterat60°Cfor45s;afterward,
theskinlayersweredividedbytweezers.Allskinlayerswereextractedwithmethanol
andtheobtainedsolutionswereanalysedbyHPLC.
TheanalysiswasundertherevisionandapprovaloftheIndependentBioethicsCom
missionforResearchoftheMedicalUniversityofGdansk(numberNKBBN/12041/2014).
Thepermeabilityandpenetrationdatawereexpressedasmeanvalues±standard
deviation(SD).Statisticalcomparisonsoftheresultswereanalysedwithatwoway
ANOVAwithposthocTukeytest(p<0.05).
Figure 6. Diffusion cell apparatus.
Saline solution, 20 mL (with 0.005% sodium azide), was recirculated beneath the skin
with a constant rate of 10 mL/h. Acceptor fluid ensured the sink condition. Solutions or
extracts were applied on the surface of the skin and left for 24 h. The chamber system
was incubated at the temperature of 37
C
±
0.5
C. After 2, 4, 6, and 24 h from the
application of penetrants, samples of acceptor fluid were collected. Before skin layers’
disconnection, the penetrants were removed from the skin. The stratum corneum was
separated by the tape-stripping method, using 30 fragments of a 3 m adhesive tape with
the following parameters: pressure ~1 kg/cm
2
(applied by stamp), 2 s duration of pressure,
and a rapid removal rate at an angle of 45
. The epidermis and dermis were isolated by the
heat separation technique [
86
]. The whole skin was immersed in water at 60
C for 45 s;
afterward, the skin layers were divided by tweezers. All skin layers were extracted with
methanol and the obtained solutions were analysed by HPLC.
The analysis was under the revision and approval of the Independent Bioethics Commis-
sion for Research of the Medical University of Gdansk (number NKBBN/120-41/2014).
The permeability and penetration data were expressed as mean values
±
standard
deviation (SD). Statistical comparisons of the results were analysed with a two-way ANOVA
with post hoc Tukey test (p< 0.05).
4.4. Chromatography
The HPLC system and analysis condition were described previously [
29
]. Briefly,
the HPLC system was equipped with a spectrophotometric diode array 340S detector
pump P 580, column thermostat STH 585, and automated sample injector ASI-100 (Dionex
Corporation, Sunnyvale, CA, USA). In addition, for the results confirmation, an additional
Molecules 2021,26, 6547 12 of 16
detector was utilised: Coulochem II electrochemical detector with a 5020 model guard and
a 5010 model analytical cell (ESA, Chelmsford, MA, USA) operated by the Chromeleon
chromatography-management system v. 6.8 (Dionex). The separation of mangiferin and
hesperidin was performed on a Hypersil Gold C
18
column (150 mm
×
4.6 mm I.D., 5
µ
m
particle) equipped with a Hypersil Gold guard column (10 mm
×
4.6 mm I.D., 5
µ
m
particle) (Thermo Electron Corporation, Dreieich, Germany).
Before use, the isocratic mobile phase (15 mM of sodium phosphate, pH 4.0 with
85% orthophosphoric acid and acetonitrile (65/35 (v/v)) was filtered (0.22
µ
m membrane
filter) and degassed by vacuum. The flow rate was maintained at 1.0 mL/min and the
injection volume was 20
µ
L. The temperature in the column was 20
C and that in the
automated sample injector was set at 8
C. To establish the electrochemical behaviour of
mangiferin and hesperidin, repeated injections of working standard solutions (0.1 mg/mL)
and detection at potentials from
0.5 to +1.2 V were studied [
87
]. Both mangiferin and
hesperidin exhibited good responses of hydrodynamic voltammograms in the ranges from
+0.35 to +0.95 V. The applied potentials on the guard cell, electrode E1, and electrode E2
were +1.1 V, +0.35 V, and +0.95 V, respectively. The photodiode array detector was set at
225, 254, 280, and 360 nm wavelengths, respectively [83].
Mangiferin and hesperidin exhibited a good linearity of calibration curves in the in-
vestigated ranges (10.0–100.0 ng, R
2
> 0.9995). Limits of detection (LOD) and quantification
(LOQ) for mangiferin and hesperidin were 0.35 and 9.7, and 0.24 and 8.6 ng/mL, respec-
tively. Precision and reproducibility were estimated from six consecutive replicates, and the
R.S.D. values for mangiferin and hesperidin were lower than 0.9% and 3.7%, respectively.
4.5. Fluorescence Microscopy
The utilisation of fluorescence microscopy to confirm the distribution of mangiferin
in the different skin layers was described by Ochocka et al., 2017 [
40
]. Both mangiferin
and hesperidin are substances having fused heterocyclic rings in their structure, which
determine their ability to fluoresce [
88
,
89
]. The excitation and emission maxima of hes-
peridin are 350 nm and 420 nm, respectively [
66
]. Biomolecules such as NADPH, elastin,
and collagen also exhibit fluorescence [
67
], which makes observation much more difficult
because interactions between chemical compounds having chromophores can enhance or
quench fluorescence [90].
To confirm the transdermal distribution of hesperidin with the use of fluorescence
microscopy, an additional experiment was performed. The hesperidin solution applied on
the skin was 10
µ
g/mL (96% EtOH (v/v)); in the control study, only ethanol was used (96%
EtOH (v/v)). The course of the research was analogous to that in the permeation study, but
only without collecting samples from the outlet and separation of the skin layers after the
experiment. Skin removed from the Bronaught cells was frozen at
60
C in a Cryotome
E (Thermo Electron Corporation, Rugby, UK). Just before fluorescence microscopy, several
cross-cuttings of the frozen skin were performed in order to obtain the transverse profile of the
skin. Sections with all visible layers of the skin profile were placed on a laboratory glass slide.
Freshly thawed skin cuttings were analysed with a fluorescence microscope (Nikon Eclipse
50 with filter UV 2A: 330 nm excitation, 380 nm emission, equipped with the lamp, Nikon
super-high-pressure mercury, and the camera, Nikon 05–5MC). The analysis of the obtained
images (magnification 100 times) was performed in the program NIS Elements AR3.2.
Author Contributions:
Conceptualization, A.H.; Data curation, J.R.O., K.C. and J.S.-H.; Formal anal-
ysis, H.B. and K.C.; Funding acquisition, J.R.O.; Investigation, A.H.; Methodology, A.H., J.R.O. and
H.B.; Project administration, K.C. and J.S.-H.; Supervision, J.R.O.; Validation, J.S.-H.; Visualization,
K.C.; Writing—original draft, A.H. and J.S.-H. All authors have read and agreed to the published
version of the manuscript.
Funding: This research recived no external funding.
Molecules 2021,26, 6547 13 of 16
Institutional Review Board Statement:
The analysis was under the revision and approval of the
Independent Bioethics Commission for Research of the Medical University of Gdansk (number
NKBBN/120-41/2014).
Informed Consent Statement:
The studies were conducted post mortem (ex vivo). For this type of
research in Poland, the only document required is the consent of the Bioethics Commission. Researchers
responsible for this stage of the work have an indefinite consent (number NKBBN/120-41/2014).
Data Availability Statement: All data are included in the manuscript.
Acknowledgments:
The authors would like to thank the Medical University of Gda´nsk for the
financial support of the study (statutory funds).
Conflicts of Interest: The authors declare no conflict of interest.
Sample Availability: Samples of the compounds are available from the authors.
References
1.
Meccariello, R.; D’Angelo, S. Impact of polyphenolic-food on longevity: An elixir of life. An overview. Antioxidants
2021
,10, 507.
[CrossRef]
2.
Khan, A.D.; Alam, M.N. Cosmetics and their associated adverse effects: A review. J. Appl. Pharm. Sci. Res.
2019
,2, 1–6. [CrossRef]
3.
Naylora, E.C.; Watsona, R.E.B.; Sherratt, M.J. Molecular aspects of skin ageing. Maturitas
2011
,69, 249–256. [CrossRef] [PubMed]
4.
Rahman, T.; Hosen, I.; Islam, M.; Shekhar, H. Oxidative stress and human health. Adv. Biosci. Biotechnol.
2012
,3, 997–1019.
[CrossRef]
5.
Tsuchida, K.; Kobayashi, M. Oxidative stress in human facial skin observed by ultraweak photon emission imaging and its
correlation with biophysical properties of skin. Sci. Rep. 2020,10, 9626–9632. [CrossRef] [PubMed]
6. Yagi, M.; Yonei, Y. Glycative stress and anti-aging: Glycative stress and skin aging. Glycative Stress Res. 2018,5, 50–54.
7. Bird, D.; Ravindra, N.M. Transdermal drug delivery and patches—An overview. Med. Devices Sens. 2020,3, e10069. [CrossRef]
8.
Pastore, M.N.; Kalia, Y.N.; Horstmann, M.; Roberts, M.S. Transdermal patches: History, development and pharmacology. Br. J.
Pharmacol. 2015,172, 2179–2209. [CrossRef]
9.
Kathe, K.; Kathpalia, H. Film forming systems for topical and transdermal drug delivery. Asian J. Pharm. Sci.
2017
,12, 487–497.
[CrossRef]
10.
Economidou, S.N.; Lamprou, D.A.; Douroumis, D. 3D printing applications for transdermal drug delivery. Int. J. Pharm.
2018
,
544, 415–424. [CrossRef] [PubMed]
11.
Margetts, L.; Sawyer, R. Transdermal drug delivery: Principles and opioid therapy. Contin. Educ. Anaesth. Crit. Care Pain
2007
,7,
171–176. [CrossRef]
12.
Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P. Experimental and computational approaches to estimate solubility and
permeability in drug discovery and envelopment setting. Adv. Drug Deliv. Rev. 1997,23, 3–25. [CrossRef]
13.
Silva, S.; Michniak-Kohn, B.; Leonardi, G.R. An overview about oxidation in clinical practice of skin aging. An. Bras. Dermatol.
2017,92, 367–374. [CrossRef] [PubMed]
14. Addor, F.A.S. Antioxidants in dermatology. An. Bras. Dermatol. 2017,92, 356–362. [CrossRef]
15.
Marwah, H.; Garg, T.; Goyal, A.K.; Rath, G. Permeation enhancer strategies in transdermal drug delivery. Drug Deliv.
2016
,23,
564–578. [CrossRef]
16.
Rehman, K.; Zulfakar, M.H. Recent advances in gel technologies for topical and t, ransdermal drug delivery. Drug Dev. Ind.
Pharm. 2013,40, 433–440. [CrossRef]
17.
Joubert, E.; Gelderblom, W.C.A.; Louw, A.; de Beer, D. South african herbal teas: Aspalathus linearis,Cyclopia spp. and Athrixia
phylicoides—A review. J. Ethnopharmacol. 2008,119, 376–412. [CrossRef]
18.
De Nysschen, A.M.; Van Wyk, B.E.; Van Heerden, F.; Schutte, A.L. The major phenolic compounds in the leaves of Cyclopia species
(Honeybush tea). Biochem. Syst. Ecol. 1996,24, 243–246. [CrossRef]
19.
Joubert, E.; Joubert, M.E.; Bester, C.; de Beer, D.; de Lange, J.H. Honeybush (Cyclopia spp.): From local cottage industry to global
markets-the catalytic and supporting role of research. S. Afr. J. Bot. 2011,77, 887–907. [CrossRef]
20.
Joubert, E.; de Beer, D.; Malherbe, C.J.; Muller, M.; Louw, A.; Gelderblom, W.C.A. Formal honeybush tea industry reaches 20-year
milestone-progress of product research targeting phenolic composition, quality and bioactivity. S. Afr. J. Bot.
2019
,127, 58–79.
[CrossRef]
21. Ganeshpurkar, A.; Saluja, A. The pharmacological potential of hesperidin. Indian J. Biochem. Biophys. 2019,56, 287–300.
22.
Jangra, A.; Arora, M.K.; Kisku, A.; Sharma, S. The multifaceted role of mangiferin in health and diseases: A review. Adv. Tradit.
Med. 2020, 1–25. [CrossRef]
23.
Agapouda, A.; Butterweck, V.; Hamburger, M.; de Beer, D.; Joubert, E.; Eckert, A. Honeybush extracts (Cyclopia spp.) rescue
mitochondrial functions and bioenergetics against oxidative injury. Oxid. Med. Cell Longev.
2020
,7, 1948602. [CrossRef] [PubMed]
24.
Dube, P.; Meyer, S.; Marnewick, J.L. Antimicrobial and antioxidant activities of different solvent extracts from fermented and
green honeybush (Cyclopia intermedia) plant material. S. Afr. J. Bot. 2017,110, 184–193. [CrossRef]
Molecules 2021,26, 6547 14 of 16
25.
De Beer, D.; Schulze, A.; Joubert, E.; de Villiers, A.; Malherbe, C.; Stander, M. Food ingredient extracts of Cyclopia subternata
(Honeybush): Variation in phenolic composition and antioxidant capacity. Molecules 2012,17, 14602–14624. [CrossRef]
26.
Van der Merwe, J.D.; de Beer, D.; Swanevelder, S.; Joubert, E.; Gelderblom, W.C.A. Dietary exposure to honeybush (Cyclopia)
polyphenol-enriched extracts altered redox status and expression of oxidative stress and antioxidant defenserelated genes in rat
liver. S. Afr. J. Bot. 2017,110, 230–239. [CrossRef]
27.
Antoinette, P.; Lester, M.D.; Fanie, R.; Jeanine, L.M. Photoprotection by honeybush extracts, hesperidin and mangiferin against
UVB-induced skin damage in SKH-1 mice. J. Photochem. Photobiol. B. 2011,103, 126–139.
28.
Im, A.R.; Song, J.H.; Lee, M.Y.; Yeon, S.H.; Um, K.A.; Chae, S. Anti-wrinkle effects of fermented and non-fermented Cyclopia
intermedia in hairless mice. BMC Complement. Altern. Med. 2014,14, 424–429. [CrossRef] [PubMed]
29.
Bartoszewski, R.; Hering, A.; Marszałł, M.; Stefanowicz-Hajduk, J.; Bartoszewska, S.; Kapoor, N.; Kochan, K.; Ochocka, J.R.
Mangiferin has an additive effect on the apoptotic properties of hesperidin in Cyclopia sp. tea extracts. PLoS ONE
2014
,9, e92128.
[CrossRef]
30.
Masibo, M.; He, Q. Major mango polyphenols and their potential significance to human health. Compr. Rev. Food Sci. Food Saf.
2008,7, 309–319. [CrossRef] [PubMed]
31.
Wang, J.; Lou, Z.; Zhu, Z.; Chai, Y.; Wu, Y. A rapid high-performance liquid chromatographic method for quantitative analysis of
antidiabetic-active components in Anemarrhena asphodeloides rhizomes. Chromatographia 2005,61, 633–636. [CrossRef]
32.
Dar, A.; Faizi, S.; Naqvi, S.; Roome, T.; Zikr-ur-Rehman, S.; Ali, M.; Firdous, S.; Moin, S.T. Analgesic and antioxidant activity of
mangiferin and its derivatives: The structure activity relationship. Biol. Pharm. Bull. 2005,28, 596–600. [CrossRef]
33.
Guha, S.; Chattopadhyav, V. Antitumor, immunomodulatory and anti-HIV effect of mangiferin, a naturally occurring glucosylx-
anthone. Chemotherapy 1996,42, 443–451. [CrossRef] [PubMed]
34.
Imran, M.; Arshad, M.S.; Butt, M.S.; Kwon, J.H.; Arshad, M.U.; Sultan, M.T. Mangiferin: A natural miracle bioactive compound
against lifestyle related disorders. Lipids Health Dis. 2017,16, 84–101. [CrossRef]
35.
Márquez, L.; García-Bueno, B.; Madrigal, J.L.; Leza, J.C. Mangiferin decreases inflammation and oxidative damage in rat brain
after stress. Eur. J. Nutr. 2012,51, 729–739. [CrossRef]
36.
Du, W.; An, Y.; He, X.; Zhang, D.; He, W. Protection of kaempferol on oxidative stress-induced retinal pigment epithelial cell
damage. Oxid. Med. Cell Longev. 2018,2018, 1610751. [CrossRef] [PubMed]
37.
Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D.; et al.
Oxidative stress, aging and diseases. Clin. Interv. Aging 2018,13, 757–772. [CrossRef]
38.
Ma, H.; Chen, H.; Sun, L.; Tong, L.; Zhang, T. Improving permeability and oral absorption of mangiferin by phospholipid
complexation. Fitoterapia 2014,93, 54–61. [CrossRef] [PubMed]
39.
Han, D.; Chen, C.; Zhang, C.; Zhang, Y.; Tang, X. Determination of mangiferin in rat plasma by liquid-liquid extraction with
UPLC-MS/MS. J. Pharm. Biomed. Anal. 2010,51, 260–263. [CrossRef]
40.
Ochocka, R.; Hering, A.; Stefanowicz–Hajduk, J.; Cal, K.; Bara´nska, H. The effect of mangiferin on skin: Penetration, permeation
and inhibition of ECM enzymes. PLoS ONE 2017,12, e0181542. [CrossRef]
41.
Kim, H.S.; Song, J.H.; Youn, U.J.; Hyun, J.W.; Jeong, W.S.; Lee, M.Y.; Choi, H.J.; Lee, H.K.; Chae, S. Inhibition of UVB-induced
wrinkle formation and MMP-9 expression by mangiferin isolated from Anemarrhena asphodeloides.Eur. J. Pharmacol.
2012
,689,
38–44. [CrossRef]
42.
Allaw, M.; Pleguezuelos-Villa, M.; Manca, M.L.; Caddeo, C.; Aroffu, M.; Nacher, A.; Diez-Sales, O.; Saurí, A.R.; Ferrer, E.E.; Fadda,
A.M.; et al. Innovative strategies to treat skin wounds with mangiferin: Fabrication of transfersomes modified with glycols and
mucin. Nanomedicine 2020,15, 1671–1685. [CrossRef]
43.
Omidbaigi, R.; Faghih, N.M. Quantitative distribution of hesperidin in Citrus species, during fruit maturation and optimal harvest
time. Nat. Prod. Radiance 2004,4, 12–15.
44.
Chen, J.; Wang, Z.Z.; Kong, L.L.; Chen, N.H. Hesperidin. In Natural Small Molecule Drugs from Plants, 1st ed.; Du, G.H., Ed.;
Springer: Singapore, 2018; pp. 81–86.
45.
Parhiz, H.; Roohbakhsh, A.; Soltani, F.; Rezaee, R.; Iranshahi, M. Antioxidant and anti-inflammatory properties of the citrus
flavonoids hesperidin and hesperetin: An updated review of their molecular mechanisms and experimental models. Phytother.
Res. 2015,29, 323–331. [CrossRef]
46.
Roohbakhsh, A.; Parhiz, H.; Soltani, F.; Rezaee, R.; Iranshahi, M. Molecular mechanisms behind the biological effects of hesperidin
and hesperetin for the prevention of cancer and cardiovascular diseases. Life Sci. 2015,124, 64–74. [CrossRef]
47.
Ganeshpurkar, A.; Saluja, A. In silico interaction of hesperidin with some immunomodulatory targets: A docking analysis. Indian
J. Biochem. Biophys. 2019,56, 28–33.
48.
Nectoux, A.M.; Abe, C.; Huang, S.W.; Ohno, N.; Tabata, J.; Miyata, Y.; Tanaka, K.; Tanaka, T.; Yamamura, H.; Matsui, T. Absorption
and metabolic behaviour of hesperidin (rutinosylated hesperetin) after single oral administration to Sprague-Dawley rats. J.
Agric. Food Chem. 2019,67, 9812–9819. [CrossRef] [PubMed]
49.
Rémésy, C. Bioavailability in humans of the flavanones hesperidin and narirutin after the ingestion of two doses of orange juice.
Eur. J. Clin. Nutr. 2003,57, 235–242.
50.
Li, C.; Schluesener, H. Health-promoting effects of the citrus flavanone hesperidin. Crit. Rev. Food Sci. Nutr.
2017
,57, 613–631.
[CrossRef]
Molecules 2021,26, 6547 15 of 16
51.
Ávila-Gálvez, M.Á.; Giménez-Bastida, J.A.; González-Sarrías, A.; Espín, J.C. New insights into the metabolism of the flavanones
eriocitrin and hesperidin: A comparative human pharmacokinetic study. Antioxidants 2021,10, 435. [CrossRef] [PubMed]
52.
Man, M.Q.; Yang, B.; Elias, P.M. Benefits of hesperidin for cutaneous functions. Evid. Based Complement. Alternat. Med.
2019
,2019,
1–19. [CrossRef] [PubMed]
53.
Schafer, M.; Werner, S. Oxidative stress in normal and impaired wound repair. Pharmacol. Res.
2008
,58, 165–171. [CrossRef]
[PubMed]
54.
Li, M.; Lin, X.F.; Lu, J.; Zhou, B.R.; Luo, D. Hesperidin ameliorates UV radiation-induced skin damage by abrogation of oxidative
stress and infammatory in HaCaT cells. J. Photochem. Photobiol. 2016,165, 240–245. [CrossRef] [PubMed]
55.
Martinez, R.M.; Pinho-Ribeiro, F.A.; Steffen, V.S.; Caviglione, C.V.; Pala, D.; Baracat, M.M.; Georgetti, S.R.; Verri, W.A.; Casagrande,
R. Topical formulation containing hesperidin methyl chalcone inhibits skin oxidative stress and inflammation induced by
ultraviolet B irradiation. Photochem. Photobiol. Sci. 2016,15, 554–563. [CrossRef]
56.
Lee, H.J.; Im, A.R.; Kim, S.M.; Kang, H.S.; Lee, J.D.; Chae, S. The favonoid hesperidin exerts anti-photoaging effect by down-
regulating matrix metalloproteinase (MMP)-9 expression via mitogen activated protein kinase (MAPK)-dependent signalling
pathways. BMC Complement. Altern. Med. 2018,18, 39–47. [CrossRef]
57.
Moon, P.D.; Kim, H.M. Anti-inflammatory effects of traditional Korean medicine, JinPi-tang and its active ingredient, hesperidin
in HaCaT cells. Phytother. Res. 2012,26, 657–662. [CrossRef]
58.
Lee, H.J.; Lee, W.J.; Chang, S.E.; Lee, G.Y. Hesperidin, a popular antioxidant inhibits melanogenesis via Erk1/2 mediated MITF
degradation. Int. J. Mol. Sci. 2015,16, 18384–18395. [CrossRef]
59.
Kiefer, S.; Weibel, M.; Smits, J.; Juch, M.; Tiedke, J.; Herbst, N. Citrus flavonoids with skin lightening effects—Safety and efficacy
studies. Int. J. Appl. Sci. 2010,136, 46–54.
60.
Zhang, C.; Lu, Y.; Tao, L.; Tao, X.; Su, X.; Wei, D. Tyrosinase inhibitory effects and inhibition mechanisms of nobiletin and
hesperidin from citrus peel crude extracts. J. Enzym. Inhib. Med. Chem. 2007,22, 83–90. [CrossRef]
61.
Hou, M.; Man, M.; Man, W.; Zhu, W.; Hupe, M.; Park, K.; Crumrine, D.; Elias, P.M.; Man, M.Q. Topical hesperidin improves
epidermal permeability barrier function and epidermal differentiation in normal murine skin. Exp. Dermatol.
2012
,21, 337–340.
[CrossRef]
62.
Bentli, R.; Ciftci, O.; Cetin, A.; Unlu, M.; Basak, N.; Cay, M. Oral administration of hesperidin, a citrus flavonone, in rats
counteracts the oxidative stress, the inflammatory cytokine production, and the hepatotoxicity induced by the ingestion of
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Eur. Cytokine Netw. 2013,24, 91–96. [CrossRef]
63.
Kaliˇcanin, B.; Velimirovi´c, D. A study of the possible harmful effects of cosmetic beauty products on human health. Biol. Trace
Elem. Res. 2016,170, 476–484. [CrossRef]
64.
Cospite, M. Double-blind, placebo-controlled evaluation of clinical activity and safety of Daflon 500 mg in the treatment of acute
hemorrhoids. Angiology 1994,45, 566–573. [PubMed]
65.
Lucca, J.M.; Joseph, R.; Al Kubaish, Z.H.; Al-Maskeen, S.M.; Alokaili, Z.A. An observational study on adverse reactions of
cosmetics: The need of practice the Cosmetovigilance system. Saudi Pharm. J 2020,28, 746–753. [CrossRef]
66.
Pavun, L.A.; Dimitric-Markovic, J.M.; Durdevic, P.T.; Jelikic-Stankov, M.D.; Dikanovic, D.B.; Ciric, A.R.; Malesev, D.L. Develop-
ment and validation of a fluorimetric method for the determination of hesperidin in human plasma and farmaceutical forms. J.
Serb. Chem. Soc. 2012,77, 1625–1640. [CrossRef]
67.
Cheong, W.; Prahl, S.A.; Welch, A.J. A review of optical properties of biological tissues. IEEE J. Quantum Electron.
1990
,26,
2166–2185. [CrossRef]
68.
Xie, M.X.; Xu, X.Y.; Wang, Y.D. Interaction between hesperetin and human serum albumin revealed by spectroscopic methods.
Biochim. Biophys. Acta 2005,1724, 215–224. [CrossRef] [PubMed]
69.
Jiang, M.; Xie, M.X.; Zheng, D.; Liu, Y.; Li, X.Y.; Chen, X. Spectroscopic studies on the interaction of cinnamic acid and its hydroxyl
derivatives with human serum albumin. J. Mol. Struct. 2004,692, 71–80.
70.
Majumdar, S.; Srirangam, R. Solubility, stability physicochemical characteristics and
in vitro
ocular tissue permeability of
hesperidin: A natural bioflavonoid. Pharm. Res. 2009,26, 1217–1225. [CrossRef] [PubMed]
71. Nuñez-Selles, A.J. Antioxidant therapy: Myth or reality? J. Braz. Chem. Soc. 2005,16, 699–710. [CrossRef]
72.
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]
73. Nott, P.E.; Roberts, J.C. The structure of mangiferin. Phytochemistry 1967,6, 741–747. [CrossRef]
74.
Takayama, K.; Kikuchi, K.; Obata, Y.; Machina, Y.; Nagai, T. Terpens as percutaneous absorption promote. STP Pharma Sci.
1991,1, 83–88.
75.
Atrahimovich, D.; Avni, D.; Khatib, S. Flavonoids-macromolecules interactions in human diseases with focus on Alzheimer,
atherosclerosis and cancer. Antioxidants 2021,10, 423. [CrossRef] [PubMed]
76. Hegde, A.H.; Sandhya, B.; Seetharamappa, J. Evaluation of binding and thermodynamic characteristics of interactions between
a citrus flavonoid hesperitin with protein and effects of metal ions on binding. Mol. Biol. Rep.
2011
,38, 4921–4929. [CrossRef]
[PubMed]
77.
Schultz, G.S.; Ladwig, G.; Wysocki, A. Extracellular matrix: Review of its roles in acute and chronic wounds. World Wide Wounds
2005,2005, 1–20.
Molecules 2021,26, 6547 16 of 16
78.
Shirshin, E.A.; Gurfinkel, Y.I.; Priezzhev, A.V.; Fadeev, V.V.; Lademann, J.; Darvin, M.E. Two-photon autofluorescence lifetime
imaging of human skin papillary dermis
in vivo
: Assessment of blood capillaries and structural proteins localization. Sci. Rep.
2017,7, 1171–1180. [CrossRef] [PubMed]
79.
Hermsmeier, M.; Jeong, S.; Yamamoto, A.; Chen, X.; Nagavarapu, U.; Evans, C.L.; Chan, K.F. Characterization of human cutaneous
tissue autofluorescence: Implications in topical drug delivery studies with fluorescence microscopy. Biomed. Opt. Express
2018
,9,
5400–5418. [CrossRef]
80.
Lin, Y.S.; Huang, W.Y.; Ho, P.Y.; Hu, S.Y.; Lin, Y.Y.; Chen, C.Y.; Chang, M.Y.; Huang, S.L. Effects of storage time and temperature on
antioxidants in juice from Momordica charantia L. and Momordica charantia L. var Abbreviata Ser.Molecules
2020
,25, 3614. [CrossRef]
[PubMed]
81.
Makarova, K.; Sajkowska-Kozielewicz, J.J.; Zawada, K.; Olchowik-Grabarek, E.; Ciach, M.A.; Gogolewski, K.; Dobros, N.;
Ciechowicz, P.; Freichels, H.; Gambin, A. Harvest time affects antioxidant capacity, total polyphenol and flavonoid content of
Polish St John’s wort’s (Hypericum perforatum L.) flowers. Sci. Rep. 2021,11, 3989–4000. [CrossRef]
82.
Juhaimi, F.A.; Ghafoor, K.; Uslu, N.; Mohamed Ahmed, I.A.; Babiker, E.E.; Özcan, M.M.; Fadimu, G.J. The effect of harvest times
on bioactive properties and fatty acid compositions of prickly pear (Opuntia ficus-barbarica A. Berger) fruits. Food Chem.
2020
,303,
125387–125393. [CrossRef] [PubMed]
83.
Joubert, E.; Botha, M.; Maicu, C.; De Beer, D.; Manley, M. Rapid screening methods for estimation of mangiferin and xanthone
contents of Cyclopia subternata plant material. S. Afr. J. Bot. 2012,82, 113–122. [CrossRef]
84.
Freeman, B.L.; Eggett, D.L.; Parker, T.L. Synergistic and antagonistic interactions of phenolic compounds found in navel oranges.
J. Food Sci. 2010,75, 570–576. [CrossRef] [PubMed]
85.
Rutkowska, M.; Olszewska, M.A.; Kolodziejczyk-Czepas, J.; Nowak, P.; Owczarek, A. Sorbus domestica leaf extracts and their
activity markers: Antioxidant potential and synergy effects in scavenging assays of multiple oxidants. Molecules
2019
,24, 2289.
[CrossRef] [PubMed]
86.
Kligman, A.M.; Christophers, E. Preparation of isolated sheets of human stratum corneum. Arch. Dermatol.
1963
,88, 702–705.
[CrossRef]
87.
Xia, J.; Kotani, A.; Hakamata, H.; Kusu, F. Determination of hesperidin in Pericarpium Citri Reticulatae by semi-micro HPLC with
electrochemical detection. J. Pharm. Biomed. Anal. 2006,41, 1401–1405. [CrossRef]
88.
Perez-Ruiz, T.; Martinez-Lozano, C.; Tomas, V.; Fenoll, J. Spectrofotometric determination of hesperidin by manual and flow-
injection methods. Fresenius J. Anal. Chem. 1999,364, 279–283.
89.
Sethiya, N.K.; Nahata, A.; Dixit, V.K. Simultaneous spectrofluorimetric determination of scopoletin and mangiferin in methanolic
extract of Conscora decussata.Asian J. Tradit. Med. 2008,3, 224–229.
90.
Doose, S.; Neuweiler, H.; Sauer, M. Fluorescence quenching by photoinduced electron transfer: A reporter for conformational
dynamics of macromolecules. ChemPhysChem 2009,10, 1389–1398. [CrossRef]
... Among the four major flavonoids in yuzu including hesperidin, naringin, narirutin, and neohesperidin, hesperidin and narirutin have anti-inflammatory activity, whitening activity, and anti-wrinkle properties [4,5]. Hesperidin also combats high cholesterol levels and fatty liver by inhibiting cholesterol synthesis and absorption [6]. ...
... As hesperidin and narirutin possess antioxidant, anti-inflammatory, and anti-wrinkle properties, the extracts containing these flavonoids have been traditionally used to treat inflammation, allergies, and liver diseases [4,5]. Flavanones, flavones, and isoflavones are considered safe to use internally and externally protect skin macromolecules from degradation [5]. ...
... As hesperidin and narirutin possess antioxidant, anti-inflammatory, and anti-wrinkle properties, the extracts containing these flavonoids have been traditionally used to treat inflammation, allergies, and liver diseases [4,5]. Flavanones, flavones, and isoflavones are considered safe to use internally and externally protect skin macromolecules from degradation [5]. Hesperidin enhances skin function by reducing oxidative stress around wound, protecting keratinocytes against ultraviolet ray induced damage, inhibiting inflammatory reactions and melanogenesis, and lightening skin [5]. ...
Article
This study examines how to obtain high levels of two major flavonoids, hesperidin and narirutin, from GY (green yuzu) powder and investigate their biological properties. We selected solvent extraction and enzyme treatment conditions before purification with an HP-20 Diaion column. We determined the appropriate extracted condition as 5 % GY with a 50 % ethanol solvent, and the enzyme treatment condition as 0.05 % RF enzyme for 24 h. Purification using 50 % ethanol as an eluent resulted in highly-enriched narirutin and hesperidin (50.78 % and 42.43 %, respectively). We also investigated the biological activities of extracts from each step, including the assessment of MMP-1 and tyrosinase inhibition activities, antibacterial properties, cytotoxicity, and anti- inflammatory activities. The extracts exhibited desirable properties such as skin elasticity, whitening effects, and anti-inflammatory effects in LPS-simulated RAW 264.7 cells. Highly purified extracts showed better anti- bacterial activity against food and dermatological pathogens. The results suggested that narirutin and hesperidin concentration and purity may influence the extracts’ biological effects. Overall, this study provides valuable insights into the improved extraction and purification of narirutin and hesperidin from GY powder and their biological activities, highlighting their potential as a functional food and health product ingredients.
... The ex vivo experiments indicated that hesperidin penetrates the skin from water and ethanol solutions with higher efficiency than from honeybush extracts. In addition, hesperidin indicates a lack of ability to interact with skin macromolecules, but, being unbound, may be capable of protecting cells against UV radiation and oxidative stress [24,25]. Mangiferin, in contrast to hesperidin, from honeybush extracts has shown a greater ability to penetrate the skin [21,24] and exhibited an ability to interact with skin extracellular matrix (ECM) molecules, incompetently inhibiting elastase and collagenase activity [21]. ...
... In addition, hesperidin indicates a lack of ability to interact with skin macromolecules, but, being unbound, may be capable of protecting cells against UV radiation and oxidative stress [24,25]. Mangiferin, in contrast to hesperidin, from honeybush extracts has shown a greater ability to penetrate the skin [21,24] and exhibited an ability to interact with skin extracellular matrix (ECM) molecules, incompetently inhibiting elastase and collagenase activity [21]. ECM compounds, such as collagen and elastin, are the structural and elasticity fibres of many organs and tissues [26,27], and they are also presented in abundant amounts among skin layers, where, with hyaluronic acid, they determine the skin functions [28][29][30]. ...
... The HPLC system and the course and conditions of the analysis were described previously [21,24,45]. Briefly, the analyses were performed on a Dionex HPLC system (a Dionex P580 gradient pump, a Dionex TCC-100 column oven, a Dionex ASI-100T autosampler and a Dionex UltiMate 3000 Photodiode Array Detector (Dionex Corporation, Sunnyvale, CA, USA)). ...
Article
Full-text available
Cyclopia sp. (honeybush) is an African shrub known as a rich source of polyphenols. The biological effects of fermented honeybush extracts were investigated. The influence of honeybush extracts on extracellular matrix (ECM) enzymes responsible for the skin malfunction and aging process—collagenase, elastase, tyrosinase and hyaluronidase—was analysed. The research also included assessment of the in vitro photoprotection efficiency of honeybush extracts and their contribution to the wound healing process. Antioxidant properties of the prepared extracts were evaluated, and quantification of the main compounds in the extracts was achieved. The research showed that the analysed extracts had a significant ability to inhibit collagenase, tyrosinase and hyaluronidase and a weak influence on elastase activity. Tyrosinase was inhibited effectively by honeybush acetone (IC50 26.18 ± 1.45 µg/mL), ethanol (IC50 45.99 ± 0.76 µg/mL) and water (IC50 67.42 ± 1.75 µg/mL) extracts. Significant hyaluronidase inhibition was observed for ethanol, acetone and water extracts (IC50 were 10.99 ± 1.56, 13.21 ± 0.39 and 14.62 ± 0.21µg/mL, respectively). Collagenase activity was inhibited effectively by honeybush acetone extract (IC50 42.5 ± 1.05 μg/mL). The wound healing properties of the honeybush extracts, estimated in vitro in human keratinocytes (HaCaTs), were indicated for water and ethanol extracts. In vitro sun protection factor (SPF in vitro) showed medium photoprotection potential for all the honeybush extracts. The quantity of polyphenolic compounds was estimated with the use of high-performance liquid chromatography equipped with diode-array detection (HPLC-DAD), indicating the highest mangiferin contents in ethanol, acetone and n-butanol extracts, while in the water extract hesperidin was the dominant compound. The antioxidant properties of the honeybush extracts were estimated with FRAP (2,4,6-Tris(2-pyridyl)-s-triazine) and DPPH (2,2-diphenyl-1-picrylhydrazyl) tests, indicating their strong antioxidant activity, similar to ascorbic acid for the acetone extract in both tests. The wound healing abilities, estimation of SPF in vitro and the direct influence on selected enzymes (elastase, tyrosinase, collagenase and hyaluronidase) of the tested honeybush extracts were analysed for the first time, indicating a high potential of these well-known herbal tea for antiaging, anti-inflammation, regeneration and protection of the skin.
... Transdermal distribution of hesperidin has also been demonstrated. The results of the study by Hering et al. (2021) showed that hesperidin was able to cross the stratum corneum, the upper layer of the epidermis, and penetrate the deeper layers of the epidermis and dermis [11]. Although the bioavailability of hesperidin and hesperetin is relatively low, it can be increased by modifying a number of substances for topical application. ...
... Transdermal distribution of hesperidin has also been demonstrated. The results of the study by Hering et al. (2021) showed that hesperidin was able to cross the stratum corneum, the upper layer of the epidermis, and penetrate the deeper layers of the epidermis and dermis [11]. Although the bioavailability of hesperidin and hesperetin is relatively low, it can be increased by modifying a number of substances for topical application. ...
... In our study, the anti-aging potential of the flavonoids hesperidin and hesperetin and their carbohydrate components rutinose, and rhamnose were investigated. The transdermal distribution of hesperidin has already been demonstrated, so we focused on its biological activity during skin aging [11]. First, their ability to inhibit the activities of enzymes associated with skin aging, specifically hyaluronidase, collagenase, and elastase, which degrade the components of the ECM, was determined in the tube test. ...
Article
Full-text available
Aging is a complex physiological process that can be accelerated by chemical (high blood glucose levels) or physical (solar exposure) factors. It is accompanied by the accumulation of altered molecules in the human body. The accumulation of oxidatively modified and glycated proteins is associated with inflammation and the progression of chronic diseases (aging). The use of antiglycating agents is one of the recent approaches in the preventive strategy of aging and natural compounds seem to be promising candidates. Our study focused on the anti-aging effect of the flavonoid hesperetin, its glycoside hesperidin and its carbohydrate moieties rutinose and rhamnose on young and physiologically aged normal human dermal fibroblasts (NHDFs). The anti-aging activity of the test compounds was evaluated by measuring matrix metalloproteinases (MMPs) and inflammatory interleukins by ELISA. The modulation of elastase, hyaluronidase, and collagenase activity by the tested substances was evaluated spectrophotometrically by tube tests. Rutinose and rhamnose inhibited the activity of pure elastase, hyaluronidase, and collagenase. Hesperidin and hesperetin inhibited elastase and hyaluronidase activity. In skin aging models, MMP-1 and MMP-2 levels were reduced after application of all tested substances. Collagen I production was increased after the application of rhamnose and rutinose.
... -Flavanones, especially naringenin and hesperidin, are inserted into TP with an antiinflammatory purpose. They are used for atopic dermatitis, WD nanoemulsions or nanogels on different polymeric supports, or lipidic nanocarriers [186][187][188]. -Flavones represented by apigenin, which is a hydrophobic flavone, have been used in several formulations so far for TD, including liposomes, nanocrystal gel formulations, and self-micro-emulsifying delivery systems for anti-inflammatory and breast cancer treatment [189,190]. ...
Article
Full-text available
Transdermal delivery devices and wound dressing materials are constantly improved and upgraded with the aim of enhancing their beneficial effects, biocompatibility, biodegradability, and cost effectiveness. Therefore, researchers in the field have shown an increasing interest in using natural compounds as constituents for such systems. Plants, as an important source of so-called “natural products” with an enormous variety and structural diversity that still exceeds the capacity of present-day sciences to define or even discover them, have been part of medicine since ancient times. However, their benefits are just at the beginning of being fully exploited in modern dermal and transdermal delivery systems. Thus, plant-based primary compounds, with or without biological activity, contained in gums and mucilages, traditionally used as gelling and texturing agents in the food industry, are now being explored as valuable and cost-effective natural components in the biomedical field. Their biodegradability, biocompatibility, and non-toxicity compensate for local availability and compositional variations. Also, secondary metabolites, classified based on their chemical structure, are being intensively investigated for their wide pharmacological and toxicological effects. Their impact on medicine is highlighted in detail through the most recent reported studies. Innovative isolation and purification techniques, new drug delivery devices and systems, and advanced evaluation procedures are presented.
... Mangiferin does not show dermal toxic effects [16]. Moreover, our previous analysis revealed that after application on the skin surface, mangiferin can pass through the stratum corneum and penetrate the dermis and epidermis layers [13,45]. On the basis of these findings and following additional, comprehensive in vitro and in vivo analysis with human tyrosinase and appropriate cell cultures, mangiferin could be considered a candidate for further research in the field of skin pathological conditions as well as for application in the cosmetic industry to maintain regular skin color. ...
Article
Full-text available
Citation: Hering, A.; Stefanowicz-Hajduk, J.; Dziomba, S.; Halasa, R.; Krzemieniecki, R.; Sappati, S.; Baginski, M.; Ochocka, J.R. Mangiferin Affects Melanin Synthesis by an Influence on Tyrosinase: Inhibition, Mechanism of Action and Molecular Docking Studies. Abstract: Mangiferin is a strong antioxidant that presents a wide range of biological activities. The aim of this study was to evaluate, for the first time, the influence of mangiferin on tyrosinase, an enzyme responsible for melanin synthesis and the unwanted browning process of food. The research included both the kinetics and molecular interactions between tyrosinase and mangiferin. The research proved that mangiferin inhibits tyrosinase activity in a dose-dependent manner with IC 50 290 +/− 6.04 µM, which was found comparable with the standard kojic acid (IC 50 217.45 +/− 2.54 µM). The mechanism of inhibition was described as mixed inhibition. The interaction between tyrosinase enzyme and mangiferin was confirmed with capillary electrophoresis (CE). The analysis indicated the formation of two main, and four less significant complexes. These results have also been supported by the molecular docking studies. It was indicated that mangiferin binds to tyrosinase, similarly to L-DOPA molecule, both in the active center and peripheral site. As it was presented in molecular docking studies, mangiferin and L-DOPA molecules can interact in a similar way with surrounding amino acid residues of tyrosinase. Additionally, hydroxyl groups of mangiferin may interact with amino acids on the tyrosinase external surface causing non-specific interaction.
... The penetration of hesperidin through the skin was found greater from water solution (451.02 ± 14.5 ng/cm 2 ) compared to ethanol (357.39 ± 4.51 ng/cm 2 ), and was also found to be significantly higher than mangiferin. Further, the distribution of hesperidin and mangiferin through the skin could be achieved in the same manner regardless of solutions or honeybush extract preparation, thus revealing the percutaneous absorption of hesperidin [102]. ...
Article
Flavonoids are natural polyphenolic compounds and constitute a major class of plant secondary metabolites. To date, structures of more than 10,000 different flavonoids have been elucidated, and most of them are present in cells and tissues of plant parts. Flavonoids have been reported to exert multiple physiological activities and are also consumed as dietary supplements. Flavonoids have been extensively explored as anticancer, anti-inflammatory, antidiabetic, antirheumatic, antioxidant, antimalarial, neuroprotective, cardioprotective, anti-angiogenic, and antiproliferative agents. Most of the flavonoids are biosynthesized in plants via the phenylpropanoid pathway. However, they are associated with some limitations. Chemical synthesis is an alternative strategy to improve the yield and obtain purified products but is hampered by drawbacks, such as intolerance to stressful lab conditions. Pharmacokinetics is the rate-limiting step defining the bioavailability and metabolism of flavonoids, though greatly influenced by their chemical structure. However, nanoformulation is an emerging technique to improve biopharmaceutical fate and achieve target drug delivery. Thus, much attention should be given to identifying other possible chemical approaches for synthesizing flavonoids and improving their pharmacokinetic profiling, hence potentiating their efficacy in clinic.
Article
Objective Excessive skin exposure to deleterious environmental variables results in inflammation as well as molecular and cellular impairments that compromise its functionality, aesthetic qualities, and overall well‐being. The implementation of topical administration of antioxidants and other compounds as a method for preventing or reversing damage is a rational approach. Numerous phenolic compounds derived from plants have demonstrated capabilities such as scavenging free radicals and promoting tissue healing. However, the primary obstacle lies in effectively delivering these compounds to the specific place on the skin, and accurately forecasting their diffusion through the skin can assist in determining the most effective tactics. Hence, this article provides a comprehensive analysis of recent literature pertaining to the in vitro skin diffusion characteristics of plant phenolics. The aim is to gain a deeper understanding of their behaviour when present in various forms such as solutions, suspensions, and formulations. Method The data on plant extracts and isolated plant phenolic compounds in vitro skin diffusion assays published over the last six years were compiled and discussed. Results Even though the gold standard Franz diffusion cell is the most commonly used in the assessment of in vitro plant phenolic skin diffusion profiles, a plethora of skin models and assay conditions are reported for a variety of compounds and extracts in different vehicles. Conclusion The presence of numerous models and vehicles poses a challenge in creating correlations among the existing data on plant phenolic compounds. However, it is possible to draw some general conclusions regarding molecular, vehicle, and skin characteristics based on the gathered information.
Article
Full-text available
Arnica tincture is a traditional herbal medicine used to treat blunt injuries, e.g., bruises and squeezes. In addition, a potential new use in the treatment of cutaneous leishmaniasis is currently under investigation. Therefore, detailed information about the dermal absorption of the tincture and especially its bioactive constituents, sesquiterpene lactones (STLs) of the helenalin- and 11α,13-dihydrohelenalin type, is mandatory. Consequently, this article reports on dermal absorption studies of Arnica tincture using diffusion cells and porcine skin as well as two human skin samples with different permeability. The amounts of STLs on the skin surfaces, in skin extracts and in the receptor fluids were quantified by ultra-high-performance liquid chromatography with high-resolution mass spectrometry (UHPLC-HRMS). It was found that Arnica STLs permeated into the receptor fluid already 4 h after the application, but the amount was rather low. Within 48 h, a maximum of 8.4%, 14.6% and 36.4% of STLs permeated through porcine skin, human skin A (trans-epidermal water loss (TEWL) = 11.518 g·m−2·h−1) and the more permeable human skin B (TEWL = 17.271 g·m−2·h−1), respectively. The majority of STLs was absorbed (penetrated into the skin; 97.6%, 97.8% and 99.3%) after 48 h but a huge portion could not be extracted from skin and is expected to be irreversibly bound to skin proteins. To better visualize the analytes in different skin layers, a fluorescence-labeled STL, helenalin 3,4-dimethoxycinnamate, was synthesized. Fluorescence microscopic images depict an accumulation of the fluorescent derivative in the epidermis. For the treatment of local, cutaneous complaints, an enrichment of the bioactive substances in the skin may be considered beneficial.
Article
Full-text available
Aging and, particularly, the onset of age-related diseases are associated with tissue dys-function and macromolecular damage, some of which can be attributed to accumulation of oxidative damage. Recently, growing interest has emerged on the beneficial effects of plant-based diets for the prevention of chronic diseases including obesity, diabetes, and cardiovascular disease. Several studies collectively suggests that the intake of polyphenols and their major food sources may exert beneficial effects on improving insulin resistance and related diabetes risk factors, such as inflammation and oxidative stress. They are the most abundant antioxidants in the diet, and their intake has been associated with a reduced aging in humans. Polyphenolic intake has been shown to be effective at ameliorating several age-related phenotypes, including oxidative stress, inflammation, impaired proteostasis, and cellular senescence, both in vitro and in vivo. In this paper, effects of these phytochemicals (either pure forms or polyphenolic-food) are reviewed and summarized according to affected cellular signaling pathways. Finally, the effectiveness of the anti-aging preventive action of nutritional interventions based on diets rich in polyphenolic food, such as the diets of the Blue zones, are discussed.
Article
Full-text available
The intake of hesperidin-rich sources, mostly found in orange juice, can decrease cardiometabolic risk, potentially linked to the gut microbial phase-II hesperetin derivatives. However, the low hesperidin solubility hampers its bioavailability and microbial metabolism, yielding a high inter-individual variability (high vs. low-producers) that prevents consistent health-related evidence. Contrarily, the human metabolism of (lemon) eriocitrin is hardly known. We hypothesize that the higher solubility of (lemon) eriocitrin vs. (orange) hesperidin might yield more bioavailable metabolites than hesperidin. A randomized-crossover human pharmacokinetic study (n = 16) compared the bioavailability and metabolism of flavanones from lemon and orange extracts and postprandial changes in oxidative, inflammatory, and metabolic markers after a high-fat-high-sugars meal. A total of 17 phase-II flavanone-derived metabolites were identified. No significant biomarker changes were observed. Plasma and urinary concentrations of all metabolites, including hesperetin metabolites, were higher after lemon extract intake. Total plasma metabolites showed significantly mean lower Tmax (6.0 ± 0.4 vs. 8.0 ± 0.5 h) and higher Cmax and AUC values after lemon extract intake. We provide new insights on hesperetin-eriodictyol interconversion and naringenin formation from hesperidin in humans. Our results suggest that regular consumption of a soluble and eco-friendly eriocitrin-rich lemon extract could provide a circulating concentration metabolites threshold to exert health benefits, even in the so-called low-producers.
Article
Full-text available
Flavonoids, a class of polyphenols, consumed daily in our diet, are associated with a reduced risk for oxidative stress (OS)-related chronic diseases, such as cardiovascular disease, neurodegenerative diseases, cancer, and inflammation. The involvement of flavonoids with OS-related chronic diseases have been traditionally attributed to their antioxidant activity. However, evidence from recent studies indicate that flavonoids' beneficial impact may be assigned to their interaction with cellular macromolecules, rather than exerting a direct antioxidant effect. This review provides an overview of the recent evolving research on interactions between the flavonoids and lipoproteins, proteins, chromatin, DNA, and cell-signaling molecules that are involved in the OS-related chronic diseases; it focuses on the mechanisms by which flavonoids attenuate the development of the aforementioned chronic diseases via direct and indirect effects on gene expression and cellular functions. The current review summarizes data from the literature and from our recent research and then compares specific flavonoids’ interactions with their targets, focusing on flavonoid structure–activity relationships. In addition, the various methods of evaluating flavonoid–protein and flavonoid–DNA interactions are presented. Our aim is to shed light on flavonoids action in the body, beyond their well-established, direct antioxidant activity, and to provide insights into the mechanisms by which these small molecules, consumed daily, influence cellular functions.
Article
Full-text available
The polyphenol content and antioxidant capacity of hyperforin and hypericin-standardized H. perforatum L. extracts may vary due to the harvest time. In this work, ethanol and ethanol–water extracts of air-dried and lyophilized flowers of H. perforatum L., collected throughout a vegetation season in central Poland, were studied. Air-dried flowers extracts had higher polyphenol (371 mg GAE/g) and flavonoid (160 mg CAE/g) content, DPPH radical scavenging (1672 mg DPPH/g), ORAC (5214 µmol TE/g) and FRAP (2.54 mmol Fe²⁺/g) than lyophilized flowers extracts (238 mg GAE/g, 107 mg CAE/g, 1287 mg DPPH/g, 3313 µmol TE/g and 0.31 mmol Fe²⁺/g, respectively). Principal component analysis showed that the collection date influenced the flavonoid and polyphenol contents and FRAP of ethanol extracts, and DPPH and ORAC values of ethanol–water extracts. The ethanol extracts with the highest polyphenol and flavonoid content protected human erythrocytes against bisphenol A-induced damage. Both high field and benchtop NMR spectra of selected extracts, revealed differences in composition caused by extraction solvent and raw material collection date. Moreover, we have shown that benchtop NMR can be used to detect the compositional variation of extracts if the assignment of signals is done previously.
Article
Full-text available
This study determined the antioxidant activities of juice from Momordica charantia L. (MC) and MC var. abbreviata Ser. (MCVAS) by analyzing 1,1-diphenyl-2-picrylhydrazyl (DPPH) scavenging ability, ferric reducing power (FRP), and total phenolic content (TPC). The effects of storage time and storage temperature on these antioxidant activities were investigated. Liquid chromatography–mass spectrometry was conducted to identify the major components of MC and MCVAS. The results revealed that the antioxidant activity of MCVAS was better than that of MC, possibly because of richer components of MCVAS. For MC and MCVAS, the scavenging concentrations of 50% DPPH were 3.33 and 1.19 mg/mL, respectively; moreover, the FRP values were 68.93 and 118.14 mg ascorbic acid equivalent/g dry weight, respectively; and the TPC values were 8.15 and 11.47 mg gallic acid equivalent/g dry weight, respectively. The antioxidant activities of MC and MCVAS decreased with storage time. High storage temperature decreased antioxidant activity more quickly than a low temperature. In addition, MC had exhibited a faster decline in DPPH scavenging ability and FRP than MCVAS during 24-day storage, but no difference was observed in TPC.
Article
Full-text available
Mitochondrial dysfunction plays a major role not only in the pathogenesis of many oxidative stress or age-related diseases such as neurodegenerative as well as mental disorders but also in normal aging. There is evidence that oxidative stress and mitochondrial dysfunction are the most upstream and common events in the pathomechanisms of neurodegeneration. Cyclopia species are endemic South African plants and some have a long tradition of use as herbal tea, known as honeybush tea. Extracts of the tea are gaining more scientific attention due to their phenolic composition. In the present study, we tested not only the in vitro mitochondria-enhancing properties of honeybush extracts under physiological conditions but also their ameliorative properties under oxidative stress situations. Hot water and ethanolic extracts of C. subternata, C. genistoides, and C. longifolia were investigated. Pretreatment of human neuroblastoma SH-SY5Y cells with honeybush extracts, at a concentration range of 0.1-1 ng/ml, had a beneficial effect on bioenergetics as it increased ATP production, respiration, and mitochondrial membrane potential (MMP) after 24 hours under physiological conditions. The aqueous extracts of C. subternata and C. genistoides, in particular, showed a protective effect by rescuing the bioenergetic and mitochondrial deficits under oxidative stress conditions (400 μM H2O2 for 3 hours). These findings indicate that honeybush extracts could constitute candidates for the prevention of oxidative stress with an impact on aging processes and age-related neurodegenerative disorders potentially leading to the development of a condition-specific nutraceutical.
Article
Full-text available
Aim: The moisturizing properties of glycerol, the penetration enhancing capability of propylene glycol and the bioadhesive properties of mucin were combined to improve the carrier capabilities of transfersomes and the efficacy of mangiferin in the treatment of skin lesions. Materials & methods: Mangiferin was incorporated in transfersomes and glycoltransfersomes, which were also modified with mucin. The physico–chemical features were assessed, along with the efficacy against oxidative stress and skin wounds in vitro and in vivo. Results: Glycoltransfersomes promoted the deposition of mangiferin in epidermis and dermis, protected fibroblasts from oxidative stress and stimulated their proliferation. The wound healing and anti-inflammatory efficacy of glycoltransfersomes were confirmed in vivo. Conclusion: Results confirmed the potential of glycoltransfersomes in preventing/treating of skin lesions.
Article
Full-text available
Oxidative stress is associated with skin ageing and disease in humans. However, it is difficult to evaluate the effects of oxidative stress on the skin in vivo using conventional invasive methods. In this study, we performed two-dimensional imaging of ultra-weak photon emission (UPE) generated by excited species in oxidative reaction to determine regional variations in oxidative stress in human facial skin and analysed the relationship between UPE intensity and biophysical properties in vivo. UPE imaging of the facial skin of volunteers revealed regional variations in oxidative stress. The nose, its surrounding regions, and the area between eyebrows showed higher UPE intensity than other facial regions, indicating high oxidative stress in these regions. In contrast, only the region surrounding the eyes showed age-related alterations in UPE intensity; moreover, wrinkle score in these regions was correlated with UPE intensity. These results suggest that oxidative stress in the skin induces wrinkle formation. UPE intensity was correlated with porphyrin score in the skin; however, no correlation was observed between UPE intensity and skin colour parameters. This study provides insights into the treatment of facial skin areas vulnerable to ageing and helps improve our understanding of topical skin diseases related to oxidative stress.
Article
Full-text available
Introduction Cosmetovigilance is public health surveillance on cosmetic products with a public health objective. Since the radical development in beautifying products in Saudi Arabia, the Saudi Food and Drug Authority takes the responsibility of regulating cosmetic products and issuing guidelines to ensure its safety. Despite this, there exists a lacuna of practice on cosmetics-related adverse reactions in the Saudi population. We aimed to assess self-reported adverse reactions in the general public of the Eastern Province, Saudi Arabia. Materials and Method A cross-sectional study was conducted for three months. United States Food and Drug Authority cosmetics adverse reaction reporting form for consumers were used for collecting the information Results Among the 425 participants, 50.6% reported that they had at least one adverse reaction in the past two years. Redness of the skin (19%), pimples (15%), and itching (13%) were the most reported adverse reactions. The majority of the adverse reactions were reported with hair care (29%) and skincare products (25%). Surprisingly, 30.9% of adverse reactions led to hospital visits. The univariate analysis found that gender, age, allergic to medications and food, family history of allergy, mixing cosmetics, and frequent switching of cosmetic brands were associated with adverse events. However, the adjusted analysis found that allergic to medication (adjusted OR: 3.9), family history of allergy (adjusted OR: 1.91), and mixing cosmetics (adjusted OR: 1.70) were significantly associated with cosmetics-related adverse reactions. Conclusion Cosmetovigilance is a brand-new model of safety monitoring of cosmetics in Saudi Arabia. It can be considered as a crucial element in public health activities. Pharmacists should be more vigil on this issue in the near future. To strengthen the findings further, a national wide prevalence study should be conducted prospectively and analyses causality and report to the Saudi FDA.
Article
A wide range of natural products including, flavonoids have been studied for their therapeutic potential in different diseases. Mangiferin (2-c-b-d-glucopyranosyl 1,3,6,7-tetrahydroxyxanthone), a natural plant xanthonoid polyphenol present mainly in the leaves and bark of Mangifera indica exhibited therapeutic activities in the treatment of various illnesses. Mounting evidence suggested mangiferin as a potent compound having diverse pharmacological activities, including antioxidant, anti-inflammatory, neuroprotective, cardioprotective, nephroprotective, hepatoprotective, anti-diabetic, anti-asthamtic, gastroprotective, immunomodulatory, anti-cancer, and hypocholesterolemic effects. Mangiferin can be considered powerful free radical scavenging activity along with multifaceted molecular targets, such as NF-ҡB, Nrf2-HO-1, PI3K/Akt, MAPK/TGF-β, SIRT-1, mTOR, AMPK, and PPARγ signaling pathways in different pathological conditions. The current review article presents an overview of major pharmacological actions and underlying pleotropic molecular mechanisms of mangiferin that have been reported in the recent literature that sheds the light on the potential of this phytoconstituent as intriguing therapeutic approach for the management of different pathological conditions. A narrative review of all the relevant papers published right from the first article on mangiferin to articles of 2020 was conducted and required data was extracted from selected papers. For the same pubmed, and other digital repository sites were explored.