Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

European Journal of Integrative Medicine 42 (2021) 101286

Contents lists available at ScienceDirect

European Journal of Integrative Medicine


journal homepage: www.elsevier.com/locate/eujim

Research paper

Photoprotective potential of a Tunisian halophyte plant Carpobrotus edulis L


Mohamed Ali Lassoued a,∗, Nour El Houda Ben Fatma a, Mariem Haj Romdhane b, Adel Faidi a,
Hatem Majdoub b, Souad Sfar a
a
University of Monastir, Laboratory of Pharmaceutical, Chemical and Pharmacological Drug Development - LR12ES09, Faculty of Pharmacy of Monastir, Rue Ibn Sina,
5000 Monastir, Tunisia
b
University of Monastir, Laboratory of Interfaces and Advanced Materials - LR11ES55, Faculty of Sciences of Monastir, Avenue de l’environnement, 5019 Monastir,
Tunisia

a r t i c l e i n f o a b s t r a c t

Keywords: Introduction: Different strategies are applicable to protect skin against the harmful effects which may appear
Carpobrotus edulis L. due to an overexposure to UV radiation including the use of sunscreens. Natural polyphenols have shown an
Chemical analysis interesting activity as photoprotectors. This study aimed to assess the potentiality of a Tunisian halophyte plant,
Phenolic compounds
Carpobrotus edulis L. (CE) as a candidate for topical photoprotection.
Antioxidant
Sun Protection Factor Methods: A phytochemical characterization of CE aqueous leaf extract was processed by quantification of total
Photoprotection polyphenols and flavonoids contents as well as by FT-IR, UV and LC-MS analysis. The plant extract was, then, in-
corporated into an oil-in-water (O/W) cream and assessed for its in vitro antioxidant properties and sun protection
factor (SPF).
Results: Phytochemical characterization highlighted the richness of the aqueous extract in polyphenols. LC-MS
analysis permitted the identification of 20 phenolic compounds. Catechin, procyanidin B2, dihydromyricetin
and syringetin-3-O-rutinoside were the most predominant components (19.39%, 16.04%, 12.85% and 14.28%
respectively). UV spectrophotometric analyses showed that CE extract absorbed in the UV region (190–400 nm).
C.E crude extract exhibited, at the concentration of 1000 μg/ml, a high photoprotective effect with a SPF value
of 6.996 ± 0.174 corresponding to a percent of UV blockage ranging from 80 to 90%. Besides, when it was
incorporated into an O/W emulsion at a concentration of 10% (m/m), it presented a good antioxidant activity
(DPPH inhibition (%) ≈ 70%) and an in vitro SPF value of 1.626 ± 0.466 corresponding to a percent of UV
blockage ≈50%.
Conclusion: These results suggest that Carpobrotus edulis L. could be a good topical photoprotective agent.

1. Introduction chronic reactions are mediated by the generation of reactive oxygen and
nitrogen species (ROS and RNS, respectively), singlet oxygen and free
Nowadays, sun exposure has increased due to changes in lifestyle. radicals, which damage lipids, proteins, nucleic acids (DNA, RNA) and
The electromagnetic radiations provided by the sun have a wide spec- others cutaneous cell components; and by an impairment or an overflow
trum of wavelengths including the ultraviolet (UV) region (100- 400 nm) of biological antioxidant systems (endogenous enzymatic and nonen-
[1,2]. An overexposure to UV-B (290 – 320 nm) and UV-A (320 - zymatic antioxidants) [3-6]. Various strategies can protect skin from
400 nm) lights without suitable protection may cause, in human skin, UV-induced damage including the use of broad-spectrum sunscreens.
a variety of undesirable effects such as oxidative stress induction, im- Sunscreen agents such as zinc oxide or titanium dioxide (physical sun-
mune response alteration, erythema, keratosis, sunburn reaction, pre- screens) act by reflecting UV radiation away from the skin. Chemical
mature skin aging and, in extreme cases, carcinogenesis. These acute and sunscreens such as methylene bis-benzotriazolyl tetramethylbutylphe-

Abbreviations: UV, Ultraviolet; ROS, Reactive oxygen species; RNS, Reactive nitrogen species; C.E, Carpobrotus edulis; FT-IR, Fourier-transform infrared; LC-MS,
Liquid chromatography–mass spectrometry; O/W, Oil-in-water; SFP, Sun protection factor; TPC, Total phenols content; GAE, Gallic acid equivalent; TFC, Total
flavonoid content; QE, Quercetin equivalent; PAD, photodiode array detector; DPPH, 2,2-diphenyl-1-picrylhydrazyl.

Corresponding author: Faculty of Pharmacy of Monastir, Laboratory of Pharmaceutical, Chemical and Pharmacological Drug Development LR12ES09, Rue
Avicenne, 5000 Monastir, Tunisia.
E-mail addresses: lassoued98@yahoo.fr (M.A. Lassoued), nour.benfatma@gmail.com (N.E.H. Ben Fatma), maryem.hajromdhane92@gmail.com (M. Haj Romd-
hane), adelfaidi_mima@yahoo.fr (A. Faidi), hatemmajdoub.fsm@gmail.com (H. Majdoub), souad.sfar@laposte.net (S. Sfar).

https://doi.org/10.1016/j.eujim.2021.101286
Received 1 September 2020; Received in revised form 6 January 2021; Accepted 8 January 2021
1876-3820/© 2021 Elsevier GmbH. All rights reserved.
M.A. Lassoued, N.E.H. Ben Fatma, M. Haj Romdhane et al. European Journal of Integrative Medicine 42 (2021) 101286

nol or isoamyl 4-methoxycinnamate act by absorbing UV rays. However, Data analysis was performed using Hyper® 1.57 software (Shimadzu
these synthetic agents may interact with cutaneous cells causing various Corporation, Japan).
skin effects such as contact dermatitis, photo-irritation or photosensitiv-
ity reactions [4]. For these reasons, in recent years, there has been an 2.5. Polyphenolic compounds analysis
increasing interest in finding natural compounds from plants such as
herbal oils, plant extracts, etc. as photoprotective agents [4,7,8]. Carpo- 2.5.1. Determination of total phenolic content
brotus edulis L. specie (Order: Caryophyllales, Family: Aizoaceae), also Total phenols content (TPC) was determined spectrophotometri-
known as “sea fig”, is a medicinal and an edible ground creeping halo- cally (Evolution® 60 UV-Visible spectrophotometer, USA), according
phyte plant growing, mainly, in sandy coastal areas [9-11]. In Tunisia, to Folin-Ciocalteu method, using gallic acid as standard [20]. 0.5 ml
its leaves were used, in traditional medicine, for their wound healing of extract stock solution (1 mg/ml) were mixed with 2.5 ml of 10%
properties and for treating warts [9]. Moreover, several published stud- Folin-Ciocalteu reagent and 2.5 ml of 7.5% NaHCO3 solution. Next, the
ies reported that this specie has several in vitro biological activities such mixture was kept for 45 min at 45 °C before measuring absorbance at
as antioxidant, antibacterial, anti-tumoral, anticholinestrease and anti- 765 nm. The experiment was performed in triplicate (n = 3). Results, ex-
inflammatory properties [12-16]. This halophyte plant is known to be pressed as μg gallic acid equivalent per ml of sample (μg GAE/ml), were
rich in phenolic compounds. Natural polyphenols, applied topically, determined from a calibration curve of gallic acid (calibration curve
have shown promising photoprotective effects against photo-damage equation: y = 8.6231x – 0.0197, r2 = 0.9988).
and inflammatory response produced by UV irradiation [4,17-19]. In
the present study, we investigated the potential photoprotective effect 2.5.2. Determination of total flavonoid content
of Carpobrotus edulis L. (C.E) aqueous leaf extract, collected from a Total flavonoid content (TFC) was assessed according to Lamaison
Tunisian biotope. The phytochemical characterization of the plant mate- and Carnat method (flavonoids-aluminium’s complex formation), us-
rial was carried out by Fourier-transform infrared spectroscopy (FT-IR), ing quercetin as standard [21]. 2 ml aliquots of extract stock solution
by spectrophotometric methods (total contents of polyphenols (TPC) (1 mg/ml) were mixed with 2 ml of 2% AlCl3 solution. Next, absorbance
and flavonoids (TFC)) and by Liquid chromatography–mass spectrom- was measured at 430 nm (Evolution® 60 UV-Visible spectrophotome-
etry (LC-MS). The plant extract was, then, incorporated into an oil-in- ter, USA), after 10 min of incubation at room temperature. Experiment
water (O/W) emulsion and its effects on the physicochemical proper- was repeated 3 times (n = 3). Results, expressed as μg quercetin equiv-
ties of the formulation (pH, viscosity and spreadability) were studied. alent per ml of sample (μg QE/ml), were determined from a calibration
Finally, its in vitro antioxidant properties and photoprotective activity curve of quercetin (calibration curve equation: y = 0.045x – 0.0328;
(UV spectrum analysis, sun protection factor (SPF)) were evaluated. r2 = 0.9994).

2. Material and methods 2.5.3. Characterization of phenolic compounds by LC-MS


The aqueous extract (5 mg/ml in distilled water) was analyzed us-
2.1. Chemical reagents ing a Shimadzu Europe - LCMS-2010EV system (Germany) equipped
with an auto-sampler (kept at 5 °C), a degasser, a quaternary pump,
Extra virgin olive oil was purchased from the local market. POE a photodiode array detector (PAD) coupled to an electrospray ioniza-
(20) sorbitan monostearate (HLB=14.9) was purchased from PROLABO tion mass detector (LC-DAD-ESI/MS) and an automated thermostatted
(France). Sorbitan monostearate (HLB=4.7) and Sepicide® HB were ob- column compartment. Chromatographic separation was achieved with
tained from SEPPIC (France). Glycerin purified was purchased from CDH a Waters Spherisorb S3 ODS-2C18 3 𝜇m (4.6 mm × 150 mm) column
(India). Cetyl alcohol was purchased from AppliChem (Germany). Par- thermostatted at 35 °C. The solvents used were: (A) Methanol, (B) 0.1%
sol® MCX (Octyl methoxycinnamate) was purchased from ROCHE Inc formic acid in water. The elution gradient established was isocratic 15%
(Switzerland). Ethanol absolute was purchased from LOBA Chemie (In- A for 5 min, 15–30% A over 5 min, 30–50% A over 5 min, 50–60% B
dia). over 5 min, 60–70% A over 5 min, 70–80% A over 5 min, 80–90% A
over 5 min, 90–50% A over 5 min and re-equilibration of the column,
2.2. Plant material using a flow rate of 0.5 ml/min. Detection was carried out in the PAD
using 280 nm as preferred wavelength and in mass spectrometer (MS).
Carpobrotus edulis L. (C.E) was collected from its natural biotope The injection volume was 50 μl. MS detection was performed in negative
(Falaise, Monastir; Tunisia) in January 2018. The plant material was mode, using a Shimadzu Europe - LCMS-2010EV system equipped with
identified by Dr. Amer Elaissi, Ph.D. (Department of the botany, Fac- an electrospray ionization (ESI) source. Nitrogen served as the sheath
ulty of Pharmacy of Monastir, Tunisia). A voucher specimen (No. 0241) gas (50 psi); the system was operated with a spray voltage of 5 kV, a
was deposited at the herbarium of this Department. source temperature of 325 °C and a capillary voltage of −20 V. The tube
lens offset was kept at a voltage of - 66 V. The full scan covered the mass
2.3. Preparation of the extract range from m/z 100–1500. The collision energy used was 35 (arbitrary
units). Data acquisition was carried out with Xcalibur® data system.
C.E aqueous leaf extract was prepared according to the protocol de- Phenolic compounds were identified according to their retention times,
scribed by Bouftira et al. [12]: Fresh leaves were well rinsed with dis- mass spectra and UV–Vis spectra compared with standards when avail-
tilled water, cut into small pieces, boiled in distilled water for 15 min able, or, were tentatively identified by comparing the obtained results
followed by filtration using a filter paper (Munktell® 3 m/N, Germany). with available data reported in the literature.
The obtained filtrate was, then, lyophilized (FLEXI-DRY® Freeze Dryer,
USA) and stored at −10 °C, in a hermetically sealed glass bottle, until 2.6. Preparation of O/W emulsions
further use. The ratio of C.E leaf to water, for decoction, was 2:1 (w/v).
Extraction yield (%) was 0.965%. 2.6.1. Preparation of Carpobrotus edulis L. leaf extract formulations
Qualitative and quantitative compositions of the cream base (0% of
2.4. Fourier transform infrared spectroscopy analysis C.E leaf extract (F1 )) and the formulations containing various plant ex-
tract concentrations (2% and 10% of plant extract for F2 and F3 , re-
Fourier transform infrared spectrum of C.E leaf extract was obtained spectively) are reported in Table 1. O/W emulsions were prepared as
on a Shimadzu 8400 FT-IR Spectrophotometer. Data were collected over follow: Both phases A (oil phase) and B (aqueous phase) were heated
a spectral region from 4000 to 400 cm−1 with a resolution of 4 cm−1 . separately to 75 °C under gentle stirring (250 rpm). Next, the aqueous

2
M.A. Lassoued, N.E.H. Ben Fatma, M. Haj Romdhane et al. European Journal of Integrative Medicine 42 (2021) 101286

Table 1
Qualitative and quantitative compositions of cream formulations containing various
concentrations of Carpobrotus Edulis L. extract.

Amount (%)
Compound F1 F2 F3 Reference

Phase Olive oil 15 15 15 12.3


A Cetyl alcohol 5.5 5.5 5.5 5.5
Octyl methoxycinnamate 0 0 0 2.7
Sorbitan monostearate 2.75 2.75 2.75 2.75
Phase POE (20) sorbitan monostearate 2.25 2.25 2.25 2.25
B Glycerin 5 5 5 5
Carpobrotus Edulis extract 0 2 10 0
Water up to 100
Phase C Sepicide HB 0.5

phase was added to the oil phase at a stirring rate of 500 rpm and mixing 2.8. Antioxidant activity of cream formulations by DPPH radical
was maintained for 10 min. During this step, the temperature was kept scavenging
at 75 °C. Next, the formed emulsion was passed through a homogenizer
(ERWEKA® HO, Germany) and kept under stirring until cooling to room 1 g of each cream formulation was weighted, transferred to 100 ml
temperature. Sepicide® HB (preservative) (phase C) was added at the volumetric flask and diluted to volume with ethanol absolute for F1 or
end when temperature was within 40 °C. Stirring was, then, maintained ethanol-water mixture (40:60 v/v) for F2 and F3 . Next, samples were,
for 5 more minutes. first, vortexed for 1 min, then kept for ultrasonication for 10 min (Pro-
labo® Transsonic 275, T460 model, 35 khz, Germany) and finally mixed
using a magnetic stirring at 500 rpm for 10 min. Afterward, they were
2.6.2. Preparation of the reference Octyl methoxycinnamate (2.7%) as the
filtered through a pleated filter paper (Munktell® 3 m/N, Germany).
standard sunscreen
Free radical scavenging activity was measured from the bleaching
A reference formulation (Table 1) containing 2.7% of Octyl
of purple-coloured ethanol solution of 2,2-diphenyl-1-picrylhydrazyl
methoxycinnamate (UV-B filter) was realized according to the same pro-
(DPPH), a stable free radical, according to Bouftira et al. [24]. 1 ml
cedure described in Section 2.6.1. It was prepared to ensure the repro-
of each sample was mixed with 1 ml of DPPH ethanolic solution and in-
ducibility of results in SPF determination. Expected SPF value of the
cubated in darkness at room temperature for 30 min. Absorbance values
reference formulation is 4 [22].
of the different samples were determined at 517 nm (Evolution® 60 UV-
Visible spectrophotometer, USA) against a blank solution containing a
2.7. Characterization of formulated creams mixture of 1 ml of DPPH solution and 1 ml of ethanol. DPPH quenching
ability was determined according to Falleh et al. [25] as follow:
Cream formulations with and without 2% of CE extract (F1 and F2 , Ablank − Aassay
respectively) were evaluated for their pH, viscosity and spreadability %DPPH inhibition = × 100
Ablank
in the aim to assess the plant extract effect on these physicochemical
properties. Where: A blank and A assay are the absorbance values, at 517 nm, of
the blank and the samples solutions, respectively.
All measurements were repeated in triplicate (n = 3) and results
2.7.1. pH were expressed as percentage inhibition of DPPH ± s.d. ANOVA test
1 g of sample was dispersed in 9 ml of distilled water and pH mea- and Fisher’s least significant difference (LSD) multiple range test at 95%
surements were carried out at room temperature. Results were expressed were applied to determine significant differences between groups. Stat-
as mean ± standard deviation (s.d.) of 6 repetitions (n = 6). graphics Centurion XV® software (StatPoint, version 15.1.02) was used
for statistical analysis.
2.7.2. Apparent viscosity
2.9. In vitro photoprotective assessment
Apparent viscosity was determined, at room temperature, using
Brookfield® DV-III Ultra Rheometer (USA) equipped with an RV5 and
2.9.1. UV spectrophotometric analyses
RV2 spindle for F1 and F2 , respectively. Measurements were obtained
Absorbances of C.E crude extract in water were measured with Evo-
at a spindle speed rate of 1 rpm. Results (n = 6) were expressed, in cP,
lution® 201 UV-Visible spectrophotometer (USA). Scans were realized
as mean ± s.d.
in the UV region (190–400 nm) after appropriate dilution of the samples.

2.7.3. Spreadability 2.9.2. In vitro SPF determination


Spreadability was evaluated at room temperature using the scanned 2.9.2.1. Carpobrotus edulis L. crude extract. Lyophilized crude extract
image analysis method developed by Rigo et al. [23]. A total of 20 wt was dissolved in water to a final concentration of 200 and 1000 μg/ml.
plates were used. Scanned images were obtained by a HP® Deskjet 1050 Afterward, the samples were assayed spectrophotometrically (Evolu-
scanner (China) and pictures were treated using ImageJ® software (ver- tion® 201 UV-Visible spectrophotometer, USA) by measuring the ab-
sion 1.50i, National Institutes of Health, USA). Spreadability factor (Sf ), sorbance in UV-B wavelength range (290–320 nm) with 5-nm incre-
expressed in mm2 .g−1 , was calculated as follow: ments.
In vitro SPF values were calculated by applying Mansur et al. equa-
𝐴
𝑆𝑓 = tion [26] and results were expressed as mean ± s.d of 6 repetitions
𝑊
(n = 6).
where: A (mm2 ) corresponds to the maximum spread area after addition ∑320
of all the plate weights and W (g) corresponds to the total weight added SPF = CF x EE(𝜆) x I(𝜆) x Abs(𝜆)
290
in the experiment. Results were expressed as mean ± s.d. of 3 repetitions Where: EE (𝜆) corresponds to the erythemogenic effect of radiation at
(n = 3). 𝜆; I (𝜆) is the intensity of solar light at 𝜆; Abs (𝜆) is the absorbance of the

3
M.A. Lassoued, N.E.H. Ben Fatma, M. Haj Romdhane et al. European Journal of Integrative Medicine 42 (2021) 101286

Table 2 standard compounds. Moreover, compounds were tentatively charac-


Normalized EE (𝜆) × I (𝜆) values used terized comparing the obtained information with data reported in lit-
in SPF calculation [27]. erature. In the current study, 21 phenolic compounds were recorded
𝜆 (nm) EE (𝜆) × I (𝜆) (normalized) but only 20 compounds were successfully identified (Table 3). The de-
tected phenolic compounds in C.E aqueous extract belong mostly to the
290 0.015
295 0.0817
group of flavonoids with catechin, procyanidin B2 and dihydromyricetin
300 0.2874 being the most predominant flavonoids (19.39, 16.04 and 12.85%, re-
305 0.3278 spectively), followed by the group of flavonoid-O-glycosides, essentially
310 0.1864 syringetin-3-O-rutinoside (14.28%).
315 0.0837
320 0.018
Total 1 3.3. Physicochemical parameters of cream formulations

3.3.1. pH measurements
pH measurements were done after appropriate dilution of samples
preparation at 𝜆 and CF represents the correction factor determined so
with distilled water. pH values of F1 and F2 formulations were found
that the reference formulation containing 2.7% octyl methoxycinnamate
6.50 ± 0.20 and 5.61 ± 0.17, respectively. The presence of C.E leaf
presented a SPF value of 4. EE(𝜆) × I(𝜆) values are reported in Table 2
extract in the formulation led to a significant decrease in the pH value
[26].
in comparison with the cream base (p < 0.05).

2.9.2.2. O/W cream formulations. 100 mg of cream samples were


3.3.2. Apparent viscosity
weighted in test tubes, added with 10 ml of absolute ethanol for F1
Apparent viscosity, expressed in cP, was assessed using a Brook-
and the reference formulation (containing 2.7% of Octyl methoxycin-
field® DV-III Ultra Rheometer equipped with an RV5 (for F1 ) and RV2
namate) or ethanol-water mixture (40:60 v/v) for F2 and F3 and vor-
(for F2 ) spindles at a speed rate of 1 rpm. Apparent viscosity values were
texed for 1 min. Next, samples were ultrasonicated for 10 min, vor-
found 43.792 × 103 ± 5.751 × 103 cP and 14.770 × 103 ± 0.515 × 103 cP
texed for 1 min and filtered through a pleated filter paper (Munk-
for F1 and F2 respectively. The addition of C.E extract (F2 ) led to a sta-
tell® 3 m/N, Germany). Afterward, they were appropriately di-
tistically significant decrease in apparent viscosity value in comparison
luted using the same solvent for their preparation and assayed spec-
with the cream base (F1 ) (p < 0.05).
trophotometrically (Evolution® 201 UV-Visible spectrophotometer,
USA).
3.3.3. Spreadability
Absorbance measurements as well as SPF values of each cream sam-
The scanned image analysis method developed by Rigo et al.
ple were determined as described above (Section 2.9.2.1.). SPF results
[23] was used to evaluate the spreadability of the different formulations.
corresponded to mean ± s.d. of 6 determinations.
Experiments were performed at room temperature. The formulation F2
showed a higher spreadability, and thus an easier skin application, than
2.10. Statistical analysis the formulation F1 with a calculated spreadability factor (Sf ) equal to
3.557 ± 0.464 mm2 .g − 1 and 2.169 ± 0.098 mm2 .g − 1 , respectively.
Statistical analysis was computed with Microsoft Office® Excel 2007
software. Student’s t-test was used to compare average values for F1 3.4. Anti-radical activity of formulated creams
versus F2 formulations. Differences observed were considered significant
at p ≤ 0.05. The results of DPPH free-radical scavenging activity of the cream
base (F1 ) and of those containing 2% and 10% of C.E leaf extract (F2
3. Results and F3 , respectively) are reported in Fig. 3. DPPH inhibition percentage
was found 3.05 ± 0.82%, 24.11 ± 3.71% and 69.02 ± 2.81% for F1 ,
3.1. Infrared spectroscopy analysis F2 and F3 , respectively. According to these results, there is a statisti-
cally significant difference in DPPH inhibition percentage between the
C.E leaf extract functional groups were determined by FT-IR spec- three formulations (p < 0.05). The cream base (F1 ) showed a very low
troscopy. The main vibrations detected in the FT-IR spectrum of the anti-radical activity, whereas F3 (10% of Carpobrotus edulis L. leaf ex-
plant extract were as follow (Fig. 1): a broad and intense absorp- tract) exhibited the highest percent of inhibition in comparison to other
tion band due to O–H stretching vibrations at 3324 cm−1, an ab- formulations.
sorption band due to C = O stretching vibrations at 1606 cm−1 , an
absorption band due to O–H bending vibrations at 1434 cm−1 and 3.5. In vitro photoprotective assessment: SPF determination
two absorption bands due to C–O stretching vibrations at 1249 and
1067 cm−1 . To be effective in preventing UV-induced skin damage, C.E extract
should have a wide range of absorbance in the UV region, viz, between
3.2. Phytochemical analysis of Carpobrotus edulis L. aqueous leaf extract 290 nm and 400 nm. For this purpose, both UV spectrum analysis and
sun protection factor (SPF) estimation were conducted.
Phytochemical studies of C.E extract included the determination of UV spectrophotometric analyses in UV region (Fig. 4) indicated that
total phenolic content (TPC) and total flavonoid content (TFC), as well C.E aqueous leaf extract absorbs in the UV-C (190–290 nm), UV-B (290–
as an LC-MS analysis. 320 nm) and UV-A (320–400 nm) ranges. This result suggests that C.E
According to the linear regression line equations of the calibration may provide protection against both UV-B and UV-A radiations with a
curves of gallic acid and quercetin, total phenol and total flavonoid good score.
concentrations were found to be 81.84 ± 27.92 μg of GAE / ml and In vitro SPF values were determined spectrophotometrically. SPF
3.07 ± 0.51 μg of QE / ml of extract, respectively. value of the reference formulation (2.7% OMC) was 3.664 ± 0.320
The investigation of the chromatographic profile of C.E aqueous ex- which corresponds to 91.6% when compared to the expected SPF (=
tract (Fig. 2) and the mass spectrum data revealed that this plant is rich 4). This result indicates the good reliability of this in vitro method to
in phenolic compounds. These later were identified by comparing their measure SPF. The correction factor (C.F), calculated, on the basis of the
retention time, mass spectra, and UV spectra with those obtained from standard sunscreen result, by applying Mansur et al. equation [26] is

4
M.A. Lassoued, N.E.H. Ben Fatma, M. Haj Romdhane et al. European Journal of Integrative Medicine 42 (2021) 101286

Fig. 1. FT-IR spectrum of aqueous extract of Carpobrotus


Edulis L. leaves.

Fig. 2. Chromatographic profile of Carpobrotus Edulis L. aqueous leaves extract, at 280 nm, analyzed by HPLC-MS in negative ionization mode. The numbers
correspond to the compounds listed in Table 3.

10.98. This latter was used to determine the SPF values of C.E. crude 4. Discussion
extract samples (200 and 1000 μg/ml) as well as those of F1 , F2 and F3
formulations. This study aimed to assess the potential photoprotective activ-
SPF values of C.E crude extract were 1.426 ± 0.031 and ity of Carpobrotus edulis L. (CE) aqueous leaf extract which could
6.996 ± 0.174, for a concentration of 200 and 1000 μg/ml, re- justify its use as ingredient for pharmaceutical formulations. This
spectively. Cream base formulation (F1 ) exhibited the lowest SFP work was divided, mainly, into two parts, viz, 1) a phytochemical
value (0.206 ± 0.026) when compared to the others formulations characterization of CE aqueous leaf extract by quantification of to-
(0.646 ± 0.151 and 1.626 ± 0.466 for F2 and F3 , respectively). The tal polyphenols and flavonoids contents (TPC and TPC, respectively)
highest value was obtained when 10% (m/m) of plant extract was in- as well as by FT-IR and LC-MS analysis; and 2) the assessment of
corporated. its in vitro antioxidant properties (DPPH inhibition percent) and pho-

5
M.A. Lassoued, N.E.H. Ben Fatma, M. Haj Romdhane et al. European Journal of Integrative Medicine 42 (2021) 101286

Table 3
Tentative identification of bioactive compounds detected in Carpobrotus Edulis L. leaves aqueous extract obtained using LC-MS in negative
ionization mode, according to their retention times (Rt ), wavelengths of maximum absorption (𝜆max ) and mass spectral data.

Peak Rt (min) 𝜆max (nm) [M-H]− m/z Tentative identification Classification Identified compound (%)

1 3.20 264 329 Vanillic acid-glucoside Hydrolyzable tannins 0.64


2 3.35 264 533 Kaempferol 3-O-(6″-malonyl-glucoside) Flavonoid-O-glycosides 4.75
3 4.75 – 391 not identified ——————————- 0.47
4 5.08 257 443 Rothindin Flavonoid glucosides 0.95
5 5.50 274 685 Niazinin A Thiocarbamate glycosides 0.19
6 6.28 257 475 Diosmetin-7-O-glucuronide Flavonoid-O- glycosides 0.95
7 6.52 248 613 Catechin-di-O-glucoside Flavonoid O-glycosides 1.66
8 6.73 276 455 Epicatechin 3-(3-methylgallate) Flavonoids 0.24
9 10.25 277 315 Quercetin 3-O-methyl-ether Methylated flavonoids 0.47
10 12.50 278 577 Procyanidin B2 Flavonoids 4.04
11 13.03 279 491 Isorhamnetin 4′-glucuronide Flavonoid glycosides 6.42
12 14.08 279 289 (+) – Catechin Flavonoids 9.16
13 14.59 279 577 Procyanidin B2 Flavonoids 12.00
14 15.23 279 319 (±) - Dihydromyricetin Flavonoids 12.85
15 16.42 279 289 (+) – Catechin Flavonoids 10.23
16 17.37 279 413 Asperuloside Monoterpenoid glycosides 6.66
17 19.63 350 623 Isorhamnetin-O-rutinoside Flavonoid O-glycosides 3.80
18 20.05 350 623 Isorhamnetin-O-rutinoside Flavonoid O-glycosides 4.52
19 20.62 349 639 Laricitrin-3-O-rutinose Flavonoid O-glycosides 3.34
20 21.99 351 653 Syringetin-3-O-rutinoside Flavonoid O-glycosides 14.28
21 22.53 349 507 Syringetin-O-glucoside Flavonoid O-glycosides 2.38
Σ=100%

[14,25,31,32] conducted several studies to assess C.E phytochemical


composition from the region of Jerba (south of Tunisia, arid bioclimatic
stage). Methanolic extracts analysis showed qualitative and quantita-
tive variation as function of used plant organs (shoots, roots, leaves and
stems) and harvest period (spring, summer, autumn and winter). TPC
ranged from 68.75 ± 1.07 mg GAE/g dry weight (DW) to 122.3 ± 6.4 mg
GAE/g DW and TFC varied from 22.21 ± 0.45 mg Catechin equivalent
(CE)/g DW to 66.0 ± 2.1 mg CE/g DW. Phloretin, Quercitrin, Avicularin,
(+)-Catechin, Procyanidin B2, (-)-Epicatechin, reyotrin and hydroxycin-
namic acid (in shoots methanolic extract) and Procyanidin B2 and (-)-
Epicatechin (in roots methanolic extract) were the major phenolic com-
pounds identified. The same authors [31,32] studied the phytochemical
Fig. 3. Graphic representation of the antiradical activity, measured by the per- composition of C.E shoots from Monastir (center of Tunisia, superior
centage of DPPH inhibition, for F1 (formulation without Carpobrotus Edulis L. semi-arid bioclimatic stage) and from Bizerte (North of Tunisia, humid
extract), F2 (formulation containing 2% of plant leaf extract) and F3 (formula- bioclimatic stage). Harvest periods were summer and spring, for Monas-
tion containing 10% of plant leaf extract). Experiments were repeated in trip- tir and Bizerte, respectively. For material plant harvested from Monastir,
licate (n = 3) and results are expressed as mean ± S.D. (For interpretation of TPC and TFC were 48.5 ± 0.4 mg GAE/g DW and 35.7 ± 0.7 mg CE/g
the references to colour in this figure legend, the reader is referred to the web DW, respectively. Quercitrin, avicularin, phloretin were the main phe-
version of this article.) nolic compounds found in the methanolic extract for the plant collected
from the region of Monastir, meanwhile, avicularin, isorhamnoside ruti-
nol, Reyotrin, Catechin and epicatechin were the major ones identified
toprotective effect (UV spectrum analysis and sun protection factor from shoots collected from the region of Bizerte. Hafsa et al. [33] de-
estimation). termined the phytochemical content of C.E aqueous (AQe) and ethanol-
Phytochemical analysis results (TPC, TFC and LC-MS) revealed the water (EWe) leaf extracts harvested from the region of Sousse (center of
richness of C.E leaf extract in phenolic compounds. Catechin, procyani- Tunisia, superior semi-arid bioclimatic stage). TPC was much higher in
din B2, dihydromyricetin and syringetin-3-O-rutinoside were the most hydroalcoholic extract compared to the aqueous extract (66.3 ± 6.2 mg
predominant components among the 20 phenolic compounds identi- GAE/g DW and 151.9 ± 4.1 mg GAE/g DW for AQe and EWe, respec-
fied by LC-MS analysis. Catechin (peaks 12 and 15), which belongs to tively). Same finding was noticed for TFC (21.9 ± 1.7 mg Quercetin
the family of flavonoids (group of flavan-3-ol), represents 19.39% from equivalent (QE)/g DW and 38.84 ± 0.8 mg QE/g DW for AQe and EWe,
the total phenolic composition. This compound is known for its anti- respectively). HPLC analysis showed the presence of seven major com-
aging potential, as well as preventing and repairing of damaged skin pounds, including, isoquercitin (6.442 and 3.963 mg/g residue for EWe
[27]. Besides, catechin is widely used as a positive standard in differ- and AQe, respectively), luteolin7-O-glucoside (4.013 and 3.741 mg/g
ent antioxidant bioassays [28]. Peaks 10 and 13, which were assigned residue for EWe and AQe, respectively) and ferulic acid (1.556 and
to procyanidin B2, represents 16.04% of the total polyphenol composi- 1.580 mg/g residue for EWe and AQe, respectively). Finally, a HPLC
tion. This latter compound possesses strong antioxidant properties to- analysis, conducted by Meddeb et al. [34], on water-acetone extracts
wards Lipoperoxidation and Anti-Elastase Activity [29]. Syringetin-3- of C.E leaves harvested from Monastir (Collection period: spring) high-
O-rutinoside (14.28% of the total phenolic composition of C.E aqueous lighted the presence of 4 major phenolic compounds, viz, Chlorogenic
extract), which belongs to flavonol group, has shown a protective effect acid, Catechin, 1,3-Dicaffeoylquinic acid and Gallic acid. TPC was found
of the skin cells against oxidative stress and cellular aging [30]. 184 ± 5 mg GAE/100 g of fresh matter.
When comparing our results to those reported in literature, These differences, in terms of polyphenols composition, may be at-
qualitative and quantitative differences were noticed. Falleh et al. tributed to several factors such as environmental conditions (Bioclimatic

6
M.A. Lassoued, N.E.H. Ben Fatma, M. Haj Romdhane et al. European Journal of Integrative Medicine 42 (2021) 101286

Fig. 4. Ultraviolet absorption spectra for Carpobro-


tus Edulis L. aqueous leaf extract in UV region (190–
400 nm): (A) UV-C (𝜆 = 190 – 290 nm), (B) UV-B+UV-A
(𝜆 = 290 – 400 nm) absorption bands.

stage; temperature, solar radiation, water stress, soil factors, viz, type, surements [38]. According to the results, pH value of the cream for-
chemistry, pH, salinity and nutritional status; altitude; harvest period; mulation decreased significantly in presence of C.E extract. This varia-
etc.), biological conditions (phases of growth; plant organ, viz, roots, tion may be attributed to the chemical composition of the added aque-
stems, leaves, flowers, etc.) and technical factors (extraction methods, ous extract. However, the obtained value remained within the range
solvents used, etc.) [31,35-37]]. of the physiological skin pH, which is acidic (pH ranging from 4 to 6)
In order to assess its effectiveness as topical photoprotective agent, [39].
C.E aqueous extract was incorporated into a cream formulation at dif- Viscosity is an important physical property to assess when topical
ferent concentrations (2% and 10%). The physicochemical character- formulations are prepared since it plays a key role on their use (ease of
istics (pH, apparent viscosity and spreadability) of the formulations application, uniformity of spreading, maintaining on the skin after ap-
without (F1) and with C.E extract (F2 and F3) were explored in order plication); on their stability (high viscosity interferes with the migration
to evaluate the effect of the added plant extract on those properties. of the oil droplets at the top of the emulsion under the influence of the
pH determination was preceded by a dilution of the samples to one- density difference between the two phases); on the applied dose amount;
tenth (1:10 dilution) of their original concentration with distilled wa- and on the delivery of the active ingredient to the skin, its efficiency and
ter. As reported in literature, such dilution factor does not affect mea- its transdermal diffusion capacity [38,40].

7
M.A. Lassoued, N.E.H. Ben Fatma, M. Haj Romdhane et al. European Journal of Integrative Medicine 42 (2021) 101286

In this study, the addition of the extract led to a decrease of the vis- thema than UVA [4]. Several in vivo and in vitro methods were developed
cosity of the cream when compared to the cream base. This finding is in for measurement of the UV protection factor. In vivo SPF measurement
good accordance with the literature. In fact, previous studies reported on volunteers involves the exposure to source of radiation which may
that the addition of plant extract, generally, affect the formulation con- present risks to human subjects, are tedious, time consuming process
sistency. This phenomenon can be attributed to its chemical composition and present ethical issues. In comparison, in vitro tests are easy-to-use,
which may interact with the thickener and causes a potential internal quick, simple and economical methods which can be considered as a
disarrangement of the emulsion or to its viscosity since the addition of useful tool at development stage of a novel product as a supplement of
a plant extract to the continuous phase could result in a system dilution the measurement of in vivo SPF. They, also, permit to reduce the number
and likely to a destabilization of the emulsion structure [24,41,42]. of in vivo experiments [8,51,52].
Spreadability, which indicates the area surface on which a semi-solid According to the obtained SPF values, C.E crude extract exhibited a
formulation will expand after a specified time, is closely related to the high photoprotective effect, at the concentration of 1000 μg/ml, corre-
qualitative and quantitative composition of the formulation and to its sponding to a percent of UV blockage ranging from 80 to 90% [8]. F3
viscosity [43]. This parameter has a determining influence on both user formulation showed a SPF value near 2 which corresponds to a percent
acceptance and application efficiency of the product since a poor spread- of UV blockage around 50% [8]. These results suggest that C.E aqueous
ability can lead to an uneven distribution of the topical formulation and leaf extract possesses an interesting photoprotective effect which is di-
be perceived as weakness by the consumer [38,43]. Spreadability was rectly proportional to its concentration. This activity may be explained
assessed using the scanned image analysis method [23]. The addition of by its richness in phenolic compounds (mainly flavonoids). In fact, sev-
C.E extract resulted in a significant increase of the spreadability value of eral studies highlighted the multiple functions of natural flavonoids in
the formulation in comparison with the cream base. This phenomenon photoprotection and it has been reported that, generally, the efficiency
could be explained by the difference in viscosity between F1 and F2 . of these compounds, as photoprotector, was related to their abundance
The lower is the viscosity the higher is the spreadability [44]. Hence, in the plant material [4,53-55].
the chemical composition of an added extract needs to be taken into
consideration when formulating a preparation for a topical use to get
the expected physicochemical parameters. 5. Limitations
In vitro anti-radical activity of formulated creams (F1 to F3 ) was as-
sessed by DPPH radical scavenging method. DPPH assay is considered In this current study, C.E aqueous extract was obtained by a decoc-
as a simple, rapid, economic and reliable method to estimate antirad- tion of a fresh matter for 20 min. It is well known that experimental con-
ical properties of both hydrophilic and hydrophobic compounds since ditions (methods of extraction, temperature, nature (polar and nonpo-
it may be used in different solvent systems (aqueous and nonpolar or- lar) and concentration of solvent, acidification, etc.) may have an impact
ganic solvents). Besides, free radical scavenging activity is determined on qualitative and quantitative phytocomposition of an extract. Hence,
at room temperature which avoids thermal degradation of tested com- a potential limitation of our work is the extraction process. Indeed, in
pounds [45]. a previous published study [33], the authors prepared an aqueous ex-
Antioxidant compounds protect against oxidative damages mediated tract of C.E using a dried leaf powder as matter. The extraction was
by free radicals, including, reactive oxygen and nitrogen species (ROS realized under magnetic stirring for 30 min at 60 °C. They found a total
and RNS, respectively). These latter are products of cellular metabolism flavonoid content (TFC) (21.9 ± 1.7 μg of quercetin/mg of dry extract)
(metabolic processes in biological systems) and environmental oxidative higher than that obtained in our case (3.07 ± 0.51 μg of QE / ml). Hence,
stress [45-47]. Elevated intracellular levels of reactive species can alter further studies are necessary in order to optimize the extraction process.
biomolecules (lipids, proteins and DNA) structure which may lead to Another limitation is that the photoprotective activity was assessed by
cellular dysfunction. Such deleterious changes are linked to a myriad mean of in vitro methods (UV spectrum analysis and SPF determination).
of health disorders including immune response alteration, premature Although these in-vitro results are promising and highlighted the poten-
aging, cardiovascular diseases, mutagenesis and carcinogenesis [6,45- tial photoprotective effect of C.E aqueous leaf extract, in-vivo studies
47]. must be undertaken to support this current work.
According to the obtained results, F1 (cream base) showed the lowest
DPPH activity which is most probably due to the presence of olive oil in
the formulation. In fact, in a previous study, Valavanidis et al. [48] re- 6. Conclusion
ported the radical scavenging activity of olive oil which was explained
by the presence of components with high antioxidant potential such as The aim of this present study was to assess the potential photopro-
phenolic compounds. The authors found that the amount of oil sample tective effect of a Tunisian halophyte plant Carpobrotus edulis L. Phy-
that can reduce DPPH activity by 50% (IC50 ) was about 11 ± 0.6 mg/ml tochemical characterization (FT-IR, TPC, TFC and LC-MS) indicated its
and 17.5 ± 1.2 mg/ml for extra virgin olive oil and commercial olive richness in phenolic compounds. UV spectrophotometric analyses in the
oil, respectively [48]. UV region showed that the plant extract absorbed in both UV-B and UV-
The formulation containing the highest amount of C.E extract (F3 ) A ranges. C.E crude extract exhibited a great photoprotective activity
presented the greatest activity in comparison to the two other formu- with a SPF value of ≈7 at the concentration of 1000 μg/ml correspond-
lations. This may be explained by the fact that an increase in plant ex- ing to a percent of UV blockage ranging from 80% to 90%. This plant
tract concentration led to an entrapment of more free radicals formed extract, when incorporated into an O/W emulsion at a concentration of
by DPPH and, thus, to a greater anti-radical activity. DPPH radical scav- 10% (m/m), presented a good antioxidant activity (≈70%) and an in
enging capacity of C.E may be explained by its richness in phenolics vitro SPF value of ≈1.7 which corresponds to a percent of UV blockage
compounds, particularly catechin and procyanidin B2, which are strong around 50%. Those results could be explained by the presence of phe-
free radical scavengers that act as electron donors [25,32,49]. The other nolic compounds, mainly flavonoids, in its composition. Overall, this
identified compounds, in C.E aqueous extract, should not be neglected present work highlighted the potential of Carpobrotus edulis L., as an
since synergy between the phenolic compounds could affect the global interesting photoprotective and antioxidant source. However, on basis
biological activity of plant organs [50]. of previously published data, which demonstrated the impact of envi-
Sunscreen formulations must protect the skin against the harmful ronmental, biological and experimental factors on extraction yield, as
effects of both short and long wavelength UV radiations (UV-B+UV-A) well as on the qualitative and quantitative phytocomposition of an ex-
and, thus, should cover a board-spectrum, viz, between 290 nm and tract, further studies should be conducted in the aim to explore more
400 nm. UV-B radiation is 1000 times capable of causing actinic ery- this activity taking into consideration all the factors implied.

8
M.A. Lassoued, N.E.H. Ben Fatma, M. Haj Romdhane et al. European Journal of Integrative Medicine 42 (2021) 101286

Authors contribution [15] A. Martins, A. Vasas, Z. Schelz, M. Viveiros, J. Molnár, J. Hohmann, L. Amaral, Con-
stituents of Carpobrotus edulis Inhibit P-Glycoprotein of MDR1-transfected Mouse
Lymphoma Cells, Anticancer Res 30 (2010) 829–835.
Mohamed Ali Lassoued: Conceived the project; designed and per- [16] M. Ondua, E.M. Njoya, M.A. Abdalla, L.J. McGaw, Anti-inflammatory and an-
formed the experiments; wrote the manuscript. tioxidant properties of leaf extracts of eleven South African medicinal plants
Nour El Houda Ben Fatma: Contributed to the preparation of the used traditionally to treat inflammation, J. Ethnopharmacol. 234 (2019) 27–35,
doi:10.1016/j.jep.2018.12.030.
plant extract and to the phytochemical characterization. [17] A. García Forero, D.A. Villamizar Mantilla, L.A. Núñez, R.E. Ocazionez,
Mariem Haj Romdhane: Carried out the LC-MS analysis; contributed E.E. Stashenko, J.L. Fuentes, Photoprotective and Antigenotoxic Effects of the
to the interpretation of the results and to the drafting of the manuscript. Flavonoids Apigenin, Naringenin and Pinocembrin, Photochem. Photobiol. 95 (4)
(2019) 1010–1018, doi:10.1111/php.13085.
Adel Faidi: Contributed to the interpretation of the results.
[18] J.A. Nichols, S.K. Katiyar, Skin photoprotection by natural polyphenols: anti-
Hatem Majdoub: Contributed to the interpretation of the results and inflammatory, antioxidant and DNA repair mechanisms, Arch. Dermatol. Res. 302
to the correction of the manuscript. (2) (2010) 71–83, doi:10.1007/s00403-009-1001-3.
[19] H.C. Polonini, L.L. Lima, K.M. Gonçalves, A.M.R. do Carmo, A.D. da Silva, N.R.B. Ra-
Souad Sfar: Supervised the project and contributed to the correction
poso, Photoprotective activity of resveratrol analogues, Bioorg. Med. Chem. 21 (4)
of the manuscript. (2013) 964–968, doi:10.1016/j.bmc.2012.11.052.
[20] M.S. Stankovic, Total phenolic content, flavonoid concentration and antioxidant ac-
Financial support tivity of Marrubium peregrinum L. Extracts, Kragujevac J. Sci. 33 (2011) 63–72.
[21] N. Chahmi, J. Anissi, S. Jennan, A. Farah, K. Sendide, M.El Hassouni, Antioxi-
dant activities and total phenol content of Inula viscosa extracts selected from
This research received no specific grant from any funding agency in three regions of Morocco, Asian Pac, J. Trop. Biomed. 5 (3) (2015) 228–233,
the public, commercial, or not-for-profit sectors. doi:10.1016/S2221-1691(15)30010-1.
[22] H. Gers-Barlag, E. Klette, R. Bimczok, C. Springob, P. Finkel, T. Rudolph,
H.U. Gonzenbach, P.H. Schneider, D. Kockott, U. Heinrich, H. Tronnier, R. Bernklau,
Declaration of Competing Interest W. Johncock, R. Langner, H.J. Driller, H. Westenfelder, In vitro testing to assess the
UVA protection performance of sun care products, Int. J. Cosmet. Sci. 23 (1) (2001)
The authors have no conflicts of interest to disclose. 3–14, doi:10.1046/j.1467-2494.2001.00048.x.
[23] L.A. Rigo, J. Weber, C.B. Silva, R.C.R. Beck, Evaluation of the spreadability of phar-
maceutical or cosmetic semisolid formulations using scanned images, Lat, Am. J.
Acknowledgements Pharm. 31 (10) (2012) 1387–1391.
[24] I. Bouftira, C. Abdelly, S. Sfar, Characterization of cosmetic cream with Mesem-
We thank Pr. Evangelia Varella and Dr. Eleni Koliarmou from the bryanthemum crystallinum plant extract: influence of formulation composition on
physical stability and anti-oxidant activity, Int. J. Cosmet. Sci. 30 (2008) 443–452,
laboratory of Organic chemistry in university of Aristotle of Thessaloniki doi:10.1111/j.1468-2494.2008.00469.x.
(Greece) for providing the necessary equipment for the LC-MS analysis. [25] H. Falleh, R. Ksouri, S. Oueslati, S. Guyot, C. Magné, C. Abdelly, Interspecific
variability of antioxidant activities and phenolic composition in Mesembryanthe-
Data availability mum genus, Food Chem. Toxicol. 47 (9) (2009) 2308–2313, doi:10.1016/j.fct.2009.
06.025.
[26] P. Shenekar, P. Ukirade, S. Salunkhe, S.T. Sutar, C.S. Magdum, S.K. Mohite,
The study information can be obtained from the author on request. S.G. Lokapure, S.M. Metri, In vitro evaluation of sun protection factor of fruit ex-
tract of Carica papaya L. as a lotion formulation, Eur. J. Exp. Biol. 4 (2) (2014)
References 44–47.
[27] O. Olayemi, C. Isimi, K. Ekere, M.A. Gbate, M. Emeje, Determination of sun protec-
[1] G.J. Clydesdale, G.W. Dandie, H.K. Muller, Ultraviolet light induced injury: im- tion factor number: an emerging in – vitro tool for predicting UV protection capa-
munological and inflammatory effects, Immunol. Cell. Biol. 79 (2001) 547–568, bilities, Int. J. Herb. Med. 5 (2017) 6–9.
doi:10.1046/j.1440-1711.2001.01047.x. [28] Y.S. Shyu, J.T. Lin, Y.T. Chang, C.J. Chiang, D.J. Yang, Evaluation of antioxidant
[2] S.E. Ullrich, M.L. Kripke, H.N. Ananthaswamy, Mechanisms underlying UV-induced ability of ethanolic extract from dill (Anethum graveolens L.) flower, Food Chem
immune suppression: implications for sunscreen design, Exp. Dermatol. 11 (1) 115 (2) (2009) 515–521, doi:10.1016/j.foodchem.2008.12.039.
(2002) 13–16, doi:10.1034/j.1600-0625.11.s.1.4.x. [29] M. Bos, B. Vennat, M. Meunier, M. Pouget, A. Pourrat, J. Fialip, Procyanidins from
[3] J. Fuchs, Potentials and limitations of the natural antioxidants RRR-alpha- toco- Tormentil: antioxidant Properties towards Lipoperoxidation and Anti-Elastase Ac-
pherol, l-ascorbic acid and 𝛽-carotene in cutaneous photoprotection, Free Radic, tivity, Biol. Pharm. Bull. 19 (1) (1996) 146–148, doi:10.1248/bpb.19.146.
Biol. Med. 25 (7) (1998) 848–873, doi:10.1016/s0891-5849(98)00161-0. [30] N. Min Kyun, B.Ki Hwan, H.Nam Doo, M. Nobuhiko, Antioxidant activity of cercis
[4] N. Saewan, A. Jimtaisong, Photoprotection of natural flavonoids, J. Appl. Pharm. chinensis and its protective effect on skin aging, Proc. SCSK. Conf. (2003) 291–312.
Sci. 3 (09) (2013) 129–141, doi:10.7324/JAPS.2013.3923. [31] H. Falleh, I. Jalleli, R. Ksouri, M. Boulaaba, S. Guyot, C. Magné, C. Abdelly, Ef-
[5] R. Stevanato, M. Bertelle, S. Fabris, Photoprotective characteristics of nat- fect of salt treatment on phenolic compounds and antioxidant activity of two
ural antioxidant polyphenols, Regul. Toxicol. Pharmacol. 69 (2014) 71–77, Mesembryanthemum edule provenances, Plant Physiol. Biochem. 52 (2012) 1–8,
doi:10.1016/j.yrtph.2014.02.014. doi:10.1016/j.plaphy.2011.11.001.
[6] A.C. Tedesco, L. Martínez, S. González, Photochemistry and photobiology of actinic [32] H. Falleh, R. Ksouri, M. Boulaaba, S. Guyot, C. Magné, C. Abdelly, Phenolic nature,
erythema: defensive and reparative cutaneous mechanisms, Brazilian J. Med. Biol. occurrence and polymerization degree as marker of environmental adaptation in
Res. 30 (5) (1997) 561–575, doi:10.1590/S0100-879X1997000500002. the edible halophyte Mesembryanthemum edule, South African J. Bot. 79 (2012)
[7] B. Choquenet, C. Couteau, E. Paparis, L.J.M. Coiffard, Quercetin and rutin as poten- 117–124, doi:10.1016/j.sajb.2011.10.001.
tial sunscreen agents: determination of efficacy by an in vitro method, J. Nat. Prod. [33] J. Hafsa, K.M. Hammi, M.R. Ben Khedher, M.A. Smach, B. Charfeddine, K. Limem,
71 (6) (2008) 1117–1118, doi:10.1021/np7007297. H. Majdoub, Inhibition of protein glycation, antioxidant and antiproliferative activ-
[8] S. Imam, I. Azhar, Z. Alam Mahmood, In-Vitro evaluation of Sun Protection Factor of ities of Carpobrotus edulis extracts, Biomed. Pharmacother. 84 (2016) 1496–1503,
a cream formulation prepared from extracts of Musa Accuminata (L.), Psidium Gu- doi:10.1016/j.biopha.2016.11.046.
java (L.) And Pyrus Communis (L.), Asian J. Pharm. Clin. Res. 8 (3) (2015) 234–237. [34] E. Meddeb, M. Charni, T. Ghazouani, A. Cozzolino, F. Fratianni, F. Raboudi,
[9] M.K. Boukef, Plants in traditional Tunisian medicine, Agence de coopération cul- F. Nazzaro, S. Fattouch, Biochemical and molecular study of carpobrotus edulis
turelle et technique, Tunis (1986). bioactive properties and their effects on dugesia sicula (turbellaria, tricla-
[10] E. Le Floc’h, Contribution à une étude ethnobotanique de la flore tunisienne, Im- dida) regeneration, Appl. Biochem. Biotechnol. 182 (3) (2017) 1131–1143,
primerie officielle de la République tunisienne, Tunis (1983). doi:10.1007/s12010-016-2387-y.
[11] T.N. Mudimba, J.M. Nguta, Traditional uses, phytochemistry and pharmacological [35] S. Ahmed, T.S. Griffin, D. Kraner, M.K. Schaffner, D. Sharma, M. Hazel, A.R. Leitch,
activity of Carpobrotus edulis: a global perspective, J. Phytopharm. 8 (3) (2019) C.M. Orians, W. Han, J.R. Stepp, A. Robbat, C. Matyas, C. Long, D. Xue,
111–116, doi:10.31254/phyto.2019.8305. R.F. Houser, S.B. Cash, Environmental factors variably impact tea secondary
[12] I. Bouftira, C. Abdelly, S. Sfar, Antioxidant and Antibacterial Properties of Mesem- metabolites in the context of climate change, Front. Plant Sci. 10 (2019) 939,
bryanthemum crystallinum and Carpobrotus edulis Extracts, Adv. Chem. Eng. Sci. doi:10.3389/fpls.2019.00939.
02 (2012) 359–365, doi:10.4236/aces.2012.23042. [36] R. Ksouri, W. Megdiche, H. Falleh, N. Trabelsi, M. Boulaaba, A. Smaoui, C. Abdelly,
[13] L. Custódio, A.C. Ferreira, H. Pereira, L. Silvestre, C. Vizetto-Duarte, L. Barreira, Influence of biological, environmental and technical factors on phenolic content and
A.P. Rauter, F. Albericio, J. Varela, The marine halophytes Carpobrotus edulis L. antioxidant activities of Tunisian halophytes, C. R. Biol. 331 (11) (2008) 865–873,
and Arthrocnemum macrostachyum L. are potential sources of nutritionally impor- doi:10.1016/j.crvi.2008.07.024.
tant PUFAs and metabolites with antioxidant, metal chelating and anticholinesterase [37] W. Liu, J. Liu, D. Yin, X. Zhao, Influence of Ecological Factors on the Production
inhibitory activities, Bot. Mar. 55 (2012) 281–288, doi:10.1515/bot-2012-0098. of Active Substances in the Anti-Cancer Plant Sinopodophyllum hexandrum (Royle)
[14] H. Falleh, R. Ksouri, F. Medini, S. Guyot, C. Abdelly, C. Magné, Antiox- T.S. Ying, PLoS ONE 10 (4) (2015) e0122981, doi:10.1371/journal.pone.0122981.
idant activity and phenolic composition of the medicinal and edible halo- [38] L. Montenegro, L. Rapisarda, C. Ministeri, G. Puglisi, Effects of lipids and emulsifiers
phyte Mesembryanthemum edule L., Ind. Crops Prod. 34 (2011) 1066–1071, on the physicochemical and sensory properties of cosmetic emulsions containing vi-
doi:10.1016/j.indcrop.2011.03.018. tamin E, Cosmetics 2(1) (2015) 35–47. https://doi.org/10.3390/cosmetics2010035.

9
M.A. Lassoued, N.E.H. Ben Fatma, M. Haj Romdhane et al. European Journal of Integrative Medicine 42 (2021) 101286

[39] S.M. Ali, G. Yosipovitch, Skin pH: from basic science to basic skin care, Acta. Derm. [48] A. Valavanidis, C. Nisiotou, Y. Papageorgiou, I. Kremli, N. Satravelas, N. Zinieris,
Venereol. 93 (2013) 261–267, doi:10.2340/00015555-1531. H. Zygalaki, Comparison of the radical scavenging potential of polar and lipidic frac-
[40] R.L. Schnaare, L.H. Block, L.C. Rohan, Rheology, in: D.B. Troy, P. Beringer (Eds.), tions of olive oil and other vegetable oils under normal conditions and after thermal
Remington: The Science and Practice of Pharmacy, Lippincott Williams & Wilkins, treatment, J. Agric. Food. Chem. 52 (8) (2004) 2358–2365, doi:10.1021/jf030491h.
Maryland, 2006, pp. 338–360. [49] E. Bendary, R.R. Francis, H.M.G. Ali, M.I. Sarwat, S. El Hady, Antioxidant and
[41] V.M. Di Mambro, M.J.V. Fonseca, Assays of physical stability and antioxidant activ- structure–activity relationships (SARs) of some phenolic and anilines compounds,
ity of a topical formulation added with different plant extracts, J. Pharm. Biomed. Ann. Agric. Sci. 58 (2) (2013) 173–181, doi:10.1016/J.AOAS.2013.07.002.
Anal. 37 (2) (2005) 287–295, doi:10.1016/j.jpba.2004.10.030. [50] A. Djeridane, M. Yousfi, B. Nadjemi, D. Boutassouna, P. Stocker, N. Vidal, Antiox-
[42] M. Karsheva, S. Georgieva, Flow properties of phytocosmetic formulations: effect idant activity of some algerian medicinal plants extracts containing phenolic com-
of plant extracts and thickeners, Compt. Rend. Acad. Bulg. Sci. 63 (12) (2010) pounds, Food Chem 97 (2006) 654–660, doi:10.1016/j.foodchem.2005.04.028.
1725–1732. [51] C.D. Kaur, S. Saraf, In vitro sun protection factor determination of herbal oils used in
[43] P. Rosa, J. Dos Santos, T.F. Lehmen, J. Weber, F.C. Flores, C. De Bona Da Silva, cosmetics, Pharmacognosy Res 2 (1) (2010) 22–25, doi:10.4103/0974-8490.60586.
S.M. Oliveira, I. Brusco, G.B. Milani, A. Horn Adams, In vitro and in vivo evaluation of [52] A. Wawrzynczak, I. Nowak, Spectrophotometric determination of UV protection pro-
a desonide gel-cream photostabilized with benzophenone-3, Drug Dev. Ind. Pharm. vided by cosmetic products with sunscreen properties, Chemik 65 (7) (2011) 655–
42 (1) (2016) 19–27, doi:10.3109/03639045.2015.1022554. 660, doi:10.4172/hccr.1000108.
[44] V.C.K.N. Deuschle, R.A. Norbert Deuschle, M.R. Bortoluzzi, M.L. Athayde, [53] A. Svobodová, J. Psotová, D. Walterová, Natural phenolics in the prevention
Physical chemistry evaluation of stability, spreadability, in vitro antioxidant, of UV-induced skin damage: a Review, Biomed. Pap. 147 (2) (2003) 137–145,
and photo-protective capacities of topical formulations containing calendula doi:10.5507/bp.2003.019.
officinalis L. Leaf extract, Brazilian J. Pharm. Sci. 51 (1) (2015) 63–75, [54] G. Agati, M. Tattini, Multiple functional roles of flavonoids in photoprotection, New
doi:10.1590/S1984-82502015000100007. Phytol 186 (4) (2010) 786–793, doi:10.1111/j.1469-8137.2010.03269.x.
[45] S.B. Kedare, R.P. Singh, Genesis and development of DPPH method of antioxidant as- [55] A.R. Nunes, I.G.P. Vieira, D.B. Queiroz, A.L. Alves, B. Leal, S.M. Morais, D.F. Mu-
say, J. Food Sci. Technol. 48 (4) (2011) 412–422, doi:10.1007/s13197-011-0251-1. niz, J.T. Calixto-Junior, H.D.M. Coutinho, Use of flavonoids and cinnamates, the
[46] K. Makarova, K. Zawada, D. Wagner, J. Skowyra, Optimization of antioxidant prop- main photoprotectors with natural origin, Adv. Pharmacol. Sci. 2018 (2018) 1–9,
erties of creams with berry extracts by artificial neural networks, Acta Phys. Pol. doi:10.1155/2018/5341487.
Series a 132 (1) (2017) 44–51, doi:10.12693/APhysPolA.132.44.
[47] A.K. Mishra, A. Mishra, P. Chattopadhyay, Assessment of in vitro sun protection fac-
tor of Calendula officinalis L. (asteraceae) essential oil formulation, J. Young Pharm.
4 (1) (2012) 17–21, doi:10.4103/0975-1483.93575.

10

You might also like