Back to Journals » Clinical, Cosmetic and Investigational Dermatology » Volume 17

Ex vivo Evaluation of a Liposome-Mediated Antioxidant Delivery System on Markers of Skin Photoaging and Skin Penetration

Authors Min M , Egli C, Bartolome RA, Sivamani RK

Received 28 January 2024

Accepted for publication 14 May 2024

Published 21 June 2024 Volume 2024:17 Pages 1481—1494

DOI https://doi.org/10.2147/CCID.S461753

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 3

Editor who approved publication: Dr Jeffrey Weinberg



Mildred Min,1,2 Caitlin Egli,1,3 Rebecca Alonzo Bartolome,4 Raja K Sivamani1,2,5,6

1Integrative Skin Science and Research Sacramento, Sacramento, CA, USA; 2College of Medicine, California Northstate University, Elk Grove, CA, USA; 3College of Medicine, University of St. George’s, University Centre, West Indies, Grenada; 4Nalón Innova, Asturias, Spain; 5Department of Dermatology, University of California-Davis, Sacramento, CA, USA; 6Pacific Skin Institute, Sacramento, CA, USA

Correspondence: Raja K Sivamani, Integrative Skin Science and Research Sacramento, 1451 River Park Dr. Suite 222, Sacramento, CA, 95815, USA, Tel +1 916-750-2463, Email [email protected]

Purpose: The topical application of antioxidants has been shown to augment the skin’s innate antioxidant system and enhance photoprotection. A challenge of topical antioxidant formulation is stability and penetrability. The use of a targeted drug delivery system may improve the bioavailability and delivery of antioxidants. In this ex vivo study, we assessed the effects of the topical application of a liposome-encapsulated antioxidant complex versus a free antioxidant complex alone on skin photoaging parameters and penetrability in human skin explants.
Patients and Methods: Human organotypic skin explant cultures (hOSEC) were irradiated to mimic photoaging. The encapsulated antioxidant complex and free antioxidant complex were applied topically onto the irradiated hOSEC daily for 7 days. The two control groups were healthy untreated hOSEC and irradiated hOSEC. Photoprotective efficacy was measured with pro-inflammatory cytokine (IL-6 and IL-8) and matrix metalloproteinase 9 (MMP-9) secretion. Cell viability and metabolic activity were measured via resazurin assay. Tissue damage was evaluated via lactate dehydrogenase (LDH) cytotoxicity assay. Skin penetration of the encapsulated antioxidant complex was assessed via fluorescent dye and confocal microscopy.
Results: Compared to healthy skin, irradiated skin experienced increases in IL-6, IL-8 (p < 0.05), and MMP-9 (p < 0.05) secretion. After treatment with the encapsulated antioxidant complex, there was a 39.3% reduction in IL-6 secretion, 49.8% reduction in IL-8 (p < 0.05), and 38.5% reduction in MMP-9 (p < 0.05). After treatment with the free antioxidant complex, there were no significant differences in IL-6, IL-8, or MMP-9 secretion. Neither treatment group experienced significant LDH leakage or reductions in metabolic activity. Liposomes passed through the stratum corneum and into the epidermis.
Conclusion: The topical application of a liposome-encapsulated antioxidant complex containing ectoin, astaxanthin-rich microalgae Haematococcus pluvialis extract, and THDA improves penetrability and restored IL-6, IL-8, and MMP-9 levels in irradiated human skin explants, which was not seen in the comparator free antioxidant complex group.

Keywords: skin explants, ectoin, astaxanthin, THDA, tetrahexyldecyl ascorbate

Introduction

Oxidative damage induced by reactive oxygen species (ROS) and free radicals have been implicated in numerous dermatological conditions. Although the skin has a natural antioxidant system composed of various enzymes (eg, glutathione, catalase, isocitrate dehydrogenase) and non-enzymatic antioxidants (eg, ascorbic acid, alpha-tocopherol, uric acid). These antioxidants are readily depleted by ultraviolet (UV) exposure, and the use of supplementary antioxidants to scavenge excess free radicals have been reported to act as therapeutic agents against oxidative stress.1–3 For example, oral supplementation of turmeric/curcumin products have been shown to improve disease severity in various dermatoses, partly via its antioxidative properties.4 Carotenoids, such as astaxanthin-rich microalgae Haematococcus pluvialis extract, beta-carotene, and lycopene have also been shown to have antioxidative properties leading to photoprotective effects when taken orally.5–7 The consumption of foods containing antioxidants such as almonds containing alpha-tocopherol8 and carotenoid-rich mangos9 have also been shown to improve measures of photoaging in postmenopausal women.

Moreover, the topical delivery of antioxidants has been shown to support the skin’s innate antioxidant system. In one study, the topical application of tocopherol (vitamin E) in combination with L-ascorbic acid and other ingredients was shown to reduce signs of skin aging.10 Topical formulations including ascorbic acid, otherwise known as vitamin C, have also been shown to reduce UV-induced expressed of MMP-1 and 9, which are implicated in photoprotection.11 Acetyl zingerone is a novel antioxidant that, when applied topically, has been shown to improve the appearance of winkles, dyspigmentation, and redness intensity while maintaining photostability.12

A challenge of topical antioxidant formulation is stability and penetrability. Antioxidants that degrade rapidly may not have the bioavailability needed to exert their beneficial effects.13 Furthermore, antioxidants that are more hydrophilic may not be permeable enough to bypass the stratum corneum and get into the deeper layers of the skin.14 For example, ascorbic acid and vitamin E have been shown to degrade rapidly under UV exposure.15,16 Ascorbic acid is also hydrophilic in its natural state, thereby hindering its ability to bypass the stratum corneum.17 Alternatively, acetyl zingerone is an antioxidant which has been shown to be photostable and is able to stabilize the ascorbic acid precursor tetrahexyldecyl ascorbate (THDA).18 Tocopherol is lipophilic, allowing it to easily penetrate the lipid-rich stratum corneum.19 However, tocopherol in its pure form is poorly water soluble, photosensitive, and may be irritating to the skin.20 To overcome obstacles such as these, recent approaches to topical antioxidant delivery design have focused on methods that preserve antioxidant stability and enhance the penetrability of the antioxidant to improve bioavailability.

Emerging approaches to modulate the delivery of antioxidants to the skin have utilized specialized delivery carriers such as nanoparticles or liposomes.21,22 In these strategies, antioxidants are encapsulated in a variety of different vesicular, lipidic, and polymeric systems to prevent degradation and improve penetration.22 For molecules to pass through the stratum corneum, there are physiochemical properties that must be met. For example, the partition coefficient (log P) must be high enough for the molecule to pass through the lipid-rich stratum corneum.23 Moreover, the molecular weight would ideally be less than 500 Da and the melting point less than 200C to effectively bypass the stratum corneum.23 Physiological factors such as age, anatomical location, race, gender, and environmental conditions also influence the rate of permeability.24 Thus, the nanomaterials utilized may increase both the bioavailability and distribution of antioxidants to the skin.25 Other benefits that nanocarriers may provide include passive delivery, increased surface area, controlled release, and decreased skin irritation.26 These systems may include solid lipid nanoparticles, nanostructured lipid carriers, nanoemulsions, polymer nanoparticles, and more.27 For example, the topical application of solid lipid nanoparticles and nanostructured lipid carrier systems have been utilized with active compounds such as vitamin E (tocopherol),28 retinoic acid,29 and more.30,31 Additionally, liposomes have been utilized to load anti-aging compounds such as type I collagen,32 ferulic acid,33 apigenin and doxycycline,34 polyvinyl alcohol and gelatin,35 and ligustrazine hydrochloride,36 all of which have shown improved photoprotective effects with the utilization of the liposomal technology. Complexing lipids with polyphenols, termed lipophenols, have also been shown to be effective.37,38 Moreover, nanocarriers have been utilized to stabilize vitamin C and its derivatives, thereby avoiding rapid oxidation and degradation.39

The topical administration of antioxidants to the skin is an important component of preventing oxidative stress that accelerates photoaging. However, some antioxidants are limited by their stability and permeability. Preliminary studies have shown that various drug delivery systems may enhance the bioavailability and delivery of these antioxidants.22 Thus, the objective of this ex-vivo study was to assess the effects of a topical liposome-mediated drug delivery system with antioxidants (XOSM™ technology, Image Skincare, Lantana, FL, USA) versus free antioxidants alone on parameters of skin photoaging in human skin explants. Additionally, this study aims to test the penetrability of the liposome-mediated drug delivery system.

Materials and Methods

Human ex vivo Skin Model

Human organotypic skin explant cultures (hOSECs) were obtained with informed consent from healthy donors undergoing plastic surgery (authorization granted by French government ethical committee according to French law L.1245 CSP). Skin samples were obtained from a 42-year-old Caucasian female of Fitzpatrick skin type II from the abdomen with no marks or scars. The skin was cut into 0.8 cm2 pieces and shipped in transport medium. Upon receipt, samples were placed with dermis facing down and epidermis facing up in culture plates containing skin culture medium without animal components and supplemented with antibiotics (1% pen-strep). Tissue cultures were incubated for at least 48 hours at 37°C under 5% CO2 for recovery prior to study initiation.

Materials

The two test items utilized in this study included a free antioxidant complex and an encapsulated antioxidant complex. The test items were stored in a dark at room temperature, according to NANOVEX (Nanovex Biotechnologies, Asturias, Spain) recommendations. In all experimental assays, the test items were applied topically over skin explant surface. In both cases, the ratio of components of the complex was as follows: ectoin, tetrahexyldecyl ascorbate, and Haematococcus pluvialis extract. The THDA utilized in this study is commercially produced in Parsippany, NJ, USA and has a purity of about 99%. The Haematococcus pluvialis extract utilized in this study is commercially produced in Frejus, France. The ectoin utilized in this study is commercially produced in Germany. They were provided as a final product (emulsion) and applied topically at 2 mg/cm² (10 µL).

Liposome Obtention

The Materials for the lipid mix are dissolved in ethanol (Phase A). This solution is then evaporated at low pressure to form a thin lipid layer on the inside of a round-bottomed flask or a glass vial. The purpose of this step is to create a lipid layer that will eventually shape the liposome structure.

Once the lipid layer is ready, it is rehydrated with an aqueous solution (Phase B) that has hydrophilic substances. This rehydration step involves adding the aqueous solution to the lipid layer while gently swirling or shaking the container with the obtention of large multilamellar vesicles (MLVs) To make small liposomes, the MLVs obtained in the rehydration step were homogenized to reduce their size. This method helps to break the larger vesicles into smaller, more uniform liposomes. The composition of liposomes is included in Table 1.

Table 1 Composition of Liposomes

Treatment Groups and Regimen

In order to mimic skin photo-aging, sun-like light irradiation (5 J/cm²) was applied daily to the hOSEC for a total of 7 applications. Repeated irradiation at 5 J/cm2 has been shown to act as a photoaging model for in vitro studies.40,41 The wavelength range of the UV radiation applied was mainly in the ultraviolet, visible, and near infrared bands, with wavelengths between 0.2 and 3.0 micrometers (200 to 3000 nm) (Figure 1) and the instrument utilized was the Sol 500 Solar Simulator (Dr. Honle AG, Gilching, Germany). After the hOSEC was irradiated, the test products were administered topically at 2 mg/cm2, for a total of 7 applications as detailed in Figure 2. The test products were in contact with the hOSEC throughout the study. The four groups were as follows: 1) healthy control (untreated hOSEC); 2) photo-aged hOSEC (irradiated); 3) photo-aged skin + encapsulated antioxidant complex; and 4) photo-aged skin + free antioxidant complex complex.

Figure 1 Emission spectra of a SOL Solar Simulator.

Figure 2 Graphical scheme of treatment regimen.

Points of Assay

The experimental design described above was replicated in order to obtain the following end points: LDH leakage, resazurin reduction activity, pro-inflammatory cytokines (IL-6 and IL-8) and matrix metalloproteinases (MMP-9) secretion.

Lactate Dehydrogenase Cytotoxicity Assay

A total of 50 µL of supernatant were removed from each sample and transferred into a 96-well microplate. Subsequently, 50 µL of formazan dye was added to each sample and after a 30-minute incubation period, absorbance was read using a standard ELISA plate reader at 490 nm. The released LDH in the culture medium supernatants was then measured. LDH oxidizes lactate to pyruvate which then reacts with the tetrazolium salt WST-1 to form formazan. The increase in the amount of formazan measured in the culture supernatant directly correlates to the increase in the number of lysed cells (damage) in the skin explant.

Resazurin Assay

The resazurin dye (7-hydroxy-3H-phenoxazin-3-one 10-oxide) was used in this study as an indicator of cell viability in proliferation and cytotoxicity. Prior to the topical application of the test items, the skin explants were treated with 6 µM of resazurin solution for 1 hour. Subsequently, a volume of 100 µL sample was removed from each sample and transferred into a 96-well microplate. The resorufin formed was quantified in a fluorometer plate reader. The fluorescent signal was monitored using 530 nm excitation wavelength and 590 nm emission wavelength.

Pro-Inflammatory Cytokines Quantifications: IL-6 and IL-8

Quantification of the pro-inflammatory cytokines, interleukin (IL)-6 and IL-8, was conducted from tissue culture supernatant using commercial sandwich ELISA kits from R&D systems (Bio-Techne, Minneapolis, MN, USA) following manufacturer’s instructions to measure UV-induced damage in the hOSEC.

Matrix Metalloproteinases Quantification: MMP-9

Quantification of matrix metalloproteinase 9 (MMP-9) was conducted from tissue culture supernatant using commercial sandwich ELISA kit from R&D Systems (Bio-Techne) following manufacturer’s instructions. The assay measures the remodeling protein matrix metalloproteinase MMP-9.

Statistical Analysis

Values are given as mean ± standard deviation (SD). The homogeneity of variance was confirmed by the Brown-Forsythe test, and the normality was confirmed by the D’Agostino-Pearson omnibus test. One factor analysis of variance (ANOVA) with Fisher’s LSD post-hoc tests were performed to assess differences among groups means. When homogeneity of variances could not be assumed, Welch’s correction was used, and when parametric assumptions were not met Kruskal–Wallis with Dunn’s post-hoc test was used. p-values < 0.05 were considered statistically significant. Values were compared against healthy control group values (*), against photo-aged skin group values (#) or between treatments ($).

Skin Penetration Study

Liposomes were synthesized including rhodamine-labelled phospholipids (18:1 PE CF) in the membrane of the liposomes. The full chemical name of the phospholipid(s) utilized is as follows: 1.2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-(carboxyfluorescein)(ammonium salt). It is a phosphatidylethanolamine (PE) with an 18-carbon chain and one unsaturation which is labeled with the fluorescent dye (CF). The product was characterized prior to the analysis, to assure that it was in accordance with the specifications regarding size and polydispersity index. Frozen skin samples were hydrated and set to room temperature using a saline buffer. The skin was then mounted on the Franz diffusion cells, and the water bath was set at 37°C. A total of 100 µL of fluorescent vegan liposomes were added to the receptor compartment of the Franz diffusion cells, and the compound was let to diffuse for 16 hours. After that time, the skin sample was rinsed off with ultrapure water and fixed with paraformaldehyde (PFA) for 5 hours. Finally, the sample was embedded in OCT, cryopreserved, and sectioned using a cryostat, obtaining a cross section of the skin. These skin sections were dyed with DAPI, a fluorescent stain with specificity for cellular nuclei with an emission maximum at 461 nm (blue) and observed with a confocal microscope. The sample was also observed at the emission wavelength of rhodamine and contrasted with the transmission image of the sample, which was able to provide information about the penetrability of the liposomes.

Results

LDH Assay

Untreated healthy human skin explants (healthy) were set as 100% of the LDH concentration. Tissue damage is represented by increased leakage of LDH. Compared to healthy skin, there were no significant differences in the amount of LDH leaked in irradiated skin (control) or in either treatment group (Figure 3).

Figure 3 Tissue damage (LDH leakage) of non-irradiated hOSEC (Healthy), irradiated (UV Control) or irradiated and treated with either the free antioxidant complex or the encapsulated antioxidant complex.

Resazurin Assay

The resazurin reduction assay was performed to measure cell viability and metabolic state. UV radiation reduced metabolic activity in irradiated skin (control) compared to healthy skin, however this difference was not statistically significant. When treated with the encapsulated antioxidant complex, metabolic activity was normalized (Figure 4); however, this effect was not seen in the free antioxidant complex group.

Figure 4 Metabolic activity (resazurin reduction) of non-irradiated hOSEC (Healthy), irradiated (UV Control) or irradiated and treated with either the free antioxidant or encapsulated antioxidant complex.

Pro-Inflammatory Cytokine Secretion

Compared to healthy skin, there was an increase in IL-6 secretion in irradiated skin (control). Treatment with the free antioxidant complex reduced IL-6 secretion by 9.9% compared to the control group. However, when treated with the encapsulated antioxidant complex, there was a 39.3% reduction in IL-6 compared to the control group (Figure 5).

Figure 5 IL-6 secretion of non-irradiated hOSEC (Healthy), irradiated (UV Control) or irradiated and treated with either the free antioxidant complex or the encapsulated antioxidant complex.

Compared to healthy skin, there was a significant increase in IL-8 secretion in irradiated skin (control) (p < 0.05). When treated with the free antioxidant complex, there was a 0.9% reduction in IL-8 secretion compared to the control group. When treated with the encapsulated antioxidant complex, there was a significant decrease in IL-8 secretion by 49.8% compared to the control group (p < 0.05) (Figure 6).

Figure 6 IL-8 secretion of non-irradiated hOSEC (Healthy), irradiated (UV Control) or irradiated and treated with either the free antioxidant complex or encapsulated antioxidant complex. Astericks *Indicate a statistically significant difference (p<0.05) compared to the Healthy group, hashes #Compared to the UV Control group, and dollar sign $Compared to the free antioxidant complex group.

MMP-9 Secretion

UV radiation increased the secretion of MMP-9 in irradiated skin (control) compared to healthy skin (p < 0.05). Treatment with Free antioxidant complex led to a 7.7% increase in MMP-9 levels, which was not statistically significant when compared to the control group. Treatment with Encapsulated antioxidant complex led to a 38.5% decrease in MMP-9 levels when compared to the control group (p < 0.05) (Figure 7).

Figure 7 Matrix Metalloproteinase 9 (MMP-9) secretion of non-irradiated hOSEC (Healthy), irradiated (UV Control) or irradiated and treated with either the free antioxidant complex or encapsulated antioxidant complex. Astericks *Indicate a statistically significant difference (p<0.05) compared to the Healthy group, hashes #Compared to the UV Control group, and dollar sign $Compared to the free antioxidant complex group.

Penetration Study

Fluorescent dye and confocal microscopy were utilized to visualize the penetrability of the encapsulated antioxidant complex. The liposomal distribution can be visualized in Figure 8, which shows that liposomes were able to pass through the stratum corneum and reach the epidermis.

Figure 8 (A) Cross section of the skin sample. (B) Confocal image of encapsulated antioxidant complex liposomes (red) after penetration in a skin sample (grey) and (C) Confocal image of encapsulated antioxidant complex liposomes (red) after penetration in a skin sample (grey) stained with DAPI (blue).

Abbreviations: SC, stratum corneum; EP, epidermis; D, dermis.

Discussion

Our study found that the topical application of a targeted, encapsulated antioxidant complex containing ectoin, astaxanthin-rich microalgae Haematococcus pluvialis, and THDA may restore the photoprotective capabilities of photoaged skin whereas the topical application of free antioxidants alone may not provide any significant photoprotective effects. Furthermore, the encapsulated antioxidant complex was shown to be able to pass through the stratum corneum to exert its effects in the epidermal layer. Previous studies have shown that the use of targeted delivery systems may improve the photoprotective activity of antioxidants.42–44 In our study, we visualized the penetrability of the delivery system utilized in our study to further validate our findings.

There were several notable findings in this study. Our study found that UV radiation increases the secretion of pro-inflammatory cytokines (IL-6 and IL-8) as well as MMP-9 in human skin explants. This agrees with studies showing that UVA-1 and UVB exposure in vivo significantly increases IL-6 levels,45,46 thereby highlighting its role in skin photoaging pathology. UVA exposure in human keratinocytes have also been shown to up-regulate IL-8 mRNA expression,47 which is also in agreement with our findings. MMP-9, a matrix metalloproteinase, also plays a crucial role in UV-induced skin inflammation and photoaging.48 Other pro-inflammatory mediators including IL-1, epidermal growth factor (EGF), and tumor necrosis factor receptors are also activated upon exposure to UVA and UVB and these pro-inflammatory markers may be interesting to investigate in future studies.

When irradiated skin was treated with the free antioxidant complex, there were no significant effects on IL-6, IL-8, or MMP-9 levels. Although the topical application of ectoin, astaxanthin, and THDA have been shown to improve skin photoprotection,49–53 antioxidants such as these may have their respective Limitations in terms of stability and penetrability without the use of stabilizing ingredients or targeted delivery technology. For example, astaxanthin is difficult to apply topically to the skin due to its poor water solubility,54 and the use of a nanostructure lipid carrier for astaxanthin was shown to improve the permeability and retention of astaxanthin.55 Moreover, THDA degrades rapidly when exposed to singlet oxygen and upregulates inflammatory type I interferon signaling when applied by itself; however, these effects may be mitigated with the addition of stabilizing antioxidants such as acetyl zingerone.16 Without the addition of targeted delivery technology or stabilizing antioxidants, our findings are consistent with the established limitations of free antioxidants to the skin.

Treatment with the encapsulated antioxidant complex normalized IL-6, IL-8, and MMP-9 levels to basal levels like those found in healthy skin. Our penetration study showed that the encapsulated antioxidant complex (XOSM™ technology), which utilizes liposomes to encapsulate the antioxidants, was able to bypass the stratum corneum and deliver product to the epidermal layer of the skin. Liposomes are spherical vesicles composed of one or more concentric phospholipid bilayers that act as a delivery tool for therapeutic agents.56 Liposomes have been shown to increase drug loading, prevent degradation of product, and improve the penetrability of agents with limited penetrability.57 Furthermore, liposome composition may influence permeability and stability of the liposome. For example, liposomes formulated from unsaturated phosphatidylcholine, such as in our study, tend to have highly permeable and low stable properties, whereas liposomes formulated using saturated phospholipids, such as dipalmitoyl phosphatidylcholine, tend to be more impermeable.56 Thus, the addition of the encapsulated delivery system for ectoin, astaxanthin-rich microalgae Haematococcus pluvialis extract, and THDA likely improved the bioavailability of these antioxidants, thereby improving the antioxidative and photoprotective capabilities of these ingredients on the skin. We chose these antioxidants due to their well-documented anti-photoaging effects, which have demonstrated that these antioxidants counteract UV radiation and oxidative stress on the skin. For example, ectoin has been used in anti-aging topicals due to its inhibitory effects on tyrosinase activity, anti-inflammatory effects, and ability to absorb UV radiation, thereby protecting DNA from photodamage.58 The topical application of astaxanthin has been shown to protect against UV irradiation,59,60 thereby improving wrinkle severity and skin hydration.61 Furthermore, THDA has been shown to improve hyperpigmentation and photodamaged skin.51 Therefore, the normalization of photoaging markers after treatment with an antioxidant complex with improved penetrability is consistent with expectations based on previous literature.

In addition to the role of IL-6 in photoaged skin, some studies have found that increased levels of IL-6 are implicated in atopic dermatitis,62 psoriasis-like inflammation,63 and basal cell carcinoma,64 and that therapies that reduce IL-6 mediated signaling may improve these conditions. However, IL-6 is not always a symptom of inflammation. For example, IL-6 is involved in the regulation of the immune response, cell differentiation, and tissue repair and thus its effects can be context-dependent.65 Similarly, IL-8 has been shown to be a key inflammatory cytokine involved in atopic dermatitis,66 psoriatic disorders,67–69 and acne vulgaris.70 Furthermore, the overexpression of MMP-9 has been found to contribute to psoriatic disease,71 atopic dermatitis,72 connective tissue diseases,73 and chronic wounds.74 Thus, the downregulation of these markers by the encapsulated antioxidant complex also signifies the therapeutic potential of these antioxidants in numerous inflammatory dermatoses.

In our study, there was a reduction in metabolic activity in irradiated skin that was normalized with the encapsulated antioxidant complex but not with the free antioxidant complex alone. However, these differences were not statistically significant. Moreover, LDH leakage is an indicator of UV damage.75 However, there were no significant changes in LDH release in either treatment group compared to the control group. Thus, neither treatment had significant cytotoxic effects on human skin explants. Although there were no noted cytotoxic effects on the human skin explants in our study, liposomal agents may impart toxicity to tissues and cause immunogenic responses depending on their chemical properties.76 Cationic phospholipids, such as those utilized in our study, have been shown to influence macrophage uptake, have cytotoxic effects via ROS production, contribute to IL-6 and TNFα production, and activate the complement pathway.76

Limitations

The experimental system of this study was focused on ex vivo findings. The human skin explants utilized were derived from a single subject with replicates and the results may not be extrapolated to the general population without future clinical work in an expanded set of participants. Human skin explants have a lack of physiological processes such as circulation and desquamation that may influence the results. In vivo models allow studies that are close to reality over ex vivo models. Clinical studies should be completed to verify the results of the ex vivo model. Although our study visualized skin penetrability of the liposomes using staining and confocal microscopy, future studies may consider testing skin permeation by quantifying the bioactive compounds remaining in each Franz compartment. Furthermore, the skin has been shown to react differently to different UV wavelengths,77 for example UVA versus UVB, and utilizing a different spectrum of irradiation may produce different results.

Conclusion

Sun-like light irradiation to human skin explants led to increases in pro-inflammatory cytokines IL-6 and IL-8 as well as MMP-9, therefore mimicking photoaged skin. The topical application of a liposome-encapsulated antioxidant complex (XOSM™ technology) containing ectoin, astaxanthin-rich microalgae Haematococcus pluvialis extract, and THDA to human skin explants restored IL-6, IL-8, and MMP-9 levels in irradiated human skin explants to basal levels, which was not seen in the comparator free antioxidant complex group. Neither treatment was found to be significantly cytotoxic based on LDH leakage or metabolic activity. Furthermore, the cationic liposomal technology in the delivery system utilized bypassed the stratum corneum to get into the epidermis, thereby emphasizing the potential of targeted delivery systems to improve the efficacy and safety of drugs and other therapeutic agents such as antioxidants. Future research on diversified skin samples and in vivo studies are warranted to confirm and expand on these results.

Abbreviations

hOSEC, human organotypic skin explant cultures; IL-, interleukin; MMP, matrix metalloproteinase; LDH, lactate dehydrogenase; THDA, tetrahexyldecyl ascorbate; ROS, reactive oxygen species; UV, ultraviolet.

Institutional Review Board Statement

This study complies with the Declaration of Helsinki. The study was conducted with skin explants that were obtained from a third-party supplier (Biopredic International, Saint-Gregoire, France) that has authorization granted by the French government ethics committee in accordance with the French law L.1245 CSP. Therefore, this study did not need formal IRB approval.

Data Sharing Statement

The data is not publicly available.

Informed Consent Statement

Human organotypic skin explant cultures (hOSECs) were obtained with informed consent from healthy donors undergoing plastic surgery (authorization granted by French government ethical committee according to French law L.1245 CSP).

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Funding

Funding for this study was provided by Image Skincare. The sponsor had a role in the design and conduct of the study but no role in the data analysis or the decision to publish.

Disclosure

RKS serves as a scientific advisor for LearnHealth, Arbonne, and Codex Labs and has served as a consultant or speaker for Almirall, Burt’s Bees, Novozymes, Novartis, Sanofi, Bristol Myers Squibb, Pfizer, Nutrafol, Galderma, Novartis, Abbvie, Lilly, Incyte, Image Skincare, Leo, UCB, Sun and Regeneron Pharmaceuticals. All other authors declare no conflicts of interest in this work.

References

1. Fuchs J, Huflejt ME, Rothfuss LM, Wilson DS, Carcamo G, Packer L. Impairment of enzymic and nonenzymic antioxidants in skin by UVB irradiation. J Invest Dermatol. 1989;93(6):769–773. doi:10.1111/1523-1747.ep12284412

2. Lohana P, Suryaprawira A, Woods EL, et al. Role of enzymic antioxidants in mediating oxidative stress and contrasting wound healing capabilities in oral mucosal/skin fibroblasts and tissues. Antioxidants (Basel). 2023;12(7):1.

3. Shindo Y, Witt E, Han D, Epstein W, Packer L. Enzymic and non-enzymic antioxidants in epidermis and dermis of human skin. J Invest Dermatol. 1994;102(1):122–124. doi:10.1111/1523-1747.ep12371744

4. Vaughn AR, Branum A, Sivamani RK. Effects of turmeric (Curcuma longa) on skin health: a systematic review of the clinical evidence. Phytother Res. 2016;30(8):1243–1264. doi:10.1002/ptr.5640

5. Ito N, Seki S, Ueda F. The protective role of astaxanthin for UV-induced skin deterioration in healthy people-a randomized, double-blind, placebo-controlled trial. Nutrients. 2018;10(7):817. doi:10.3390/nu10070817

6. Li J, Zeng X, Yang X, Ding H. Lycopene ameliorates skin aging by regulating the insulin resistance pathway and activating SIRT1. Food Funct. 2022;13(21):11307–11320. doi:10.1039/d2fo01111e

7. Stahl W, Sies H. beta-Carotene and other carotenoids in protection from sunlight. Am J Clin Nutr. 2012;96(5):1179S–84S. doi:10.3945/ajcn.112.034819

8. Rybak I, Carrington AE, Dhaliwal S, et al. Prospective randomized controlled trial on the effects of almonds on facial wrinkles and pigmentation. Nutrients. 2021;13(3):785. doi:10.3390/nu13030785

9. Fam VW, Holt RR, Keen CL, Sivamani RK, Hackman RM. Prospective evaluation of mango fruit intake on facial wrinkles and erythema in postmenopausal women: a randomized clinical pilot study. Nutrients. 2020;12(11):3381. doi:10.3390/nu12113381

10. Neves JR, Grether-Beck S, Krutmann J, et al. Efficacy of a topical serum containing L-ascorbic acid, neohesperidin, pycnogenol, tocopherol, and hyaluronic acid in relation to skin aging signs. J Cosmet Dermatol. 2022;21(10):4462–4469. doi:10.1111/jocd.14837

11. Greul AK, Grundmann JU, Heinrich F, et al. Photoprotection of UV-irradiated human skin: an antioxidative combination of vitamins E and C, carotenoids, selenium and proanthocyanidins. Skin Pharmacol Appl Skin Physiol. 2002;15(5):307–315. doi:10.1159/000064534

12. Dhaliwal S, Rybak I, Pourang A, et al. Randomized double-blind vehicle controlled study of the effects of topical acetyl zingerone on photoaging. J Cosmet Dermatol. 2021;20(1):166–173. doi:10.1111/jocd.13464

13. Ascenso A, Ribeiro HM, Marques HC, Simoes S. Topical delivery of antioxidants. Curr Drug Deliv. 2011;8(6):640–660. doi:10.2174/156720111797635487

14. Belhaj N, Arab-Tehrany E, Loing E, Bezivin C. Skin delivery of hydrophilic molecules from liposomes and polysaccharide-coated liposomes. Int J Cosmet Sci. 2017;39(4):435–441. doi:10.1111/ics.12394

15. Al-Niaimi F, Chiang NYZ. Topical vitamin C and the skin: mechanisms of action and clinical applications. J Clin Aesthet Dermatol. 2017;10(7):14–17.

16. Swindell WR, Randhawa M, Quijas G, Bojanowski K, Chaudhuri RK. Tetrahexyldecyl Ascorbate (THDC) degrades rapidly under oxidative stress but can be stabilized by acetyl zingerone to enhance collagen production and antioxidant effects. Int J Mol Sci. 2021;22(16):1.

17. Stamford NP. Stability, transdermal penetration, and cutaneous effects of ascorbic acid and its derivatives. J Cosmet Dermatol. 2012;11(4):310–317. doi:10.1111/jocd.12006

18. Meyer TA, Swindell WR, Chaudhuri RK. Acetyl zingerone: a photostable multifunctional skincare ingredient that combats features of intrinsic and extrinsic skin aging. Antioxidants. 2023;12(6):1.

19. Teo CWL, Tay SHY, Tey HL, Ung YW, Yap WN. Vitamin E in atopic dermatitis: from preclinical to clinical studies. Dermatology. 2021;237(4):553–564. doi:10.1159/000510653

20. Abla MJ, Banga AK. Formulation of tocopherol nanocarriers and in vitro delivery into human skin. Int J Cosmet Sci. 2014;36(3):239–246. doi:10.1111/ics.12119

21. Hatem S, Nasr M, Elkheshen SA, Geneidi AS. Recent advances in antioxidant cosmeceutical topical delivery. Curr Drug Deliv. 2018;15(7):953–964. doi:10.2174/1567201815666180214143551

22. Pozos-Nonato S, Dominguez-Delgado CL, Campos-Santander KA, et al. Novel nanotechnological strategies for skin anti-aging. Curr Pharm Biotechnol. 2023;24(11):1397–1419. doi:10.2174/1389201024666221223095315

23. Khan NR, Harun MS, Nawaz A, Harjoh N, Wong TW. Nanocarriers and their actions to improve skin permeability and transdermal drug delivery. Curr Pharm Des. 2015;21(20):2848–2866. doi:10.2174/1381612821666150428145216

24. N’Da DD. Prodrug strategies for enhancing the percutaneous absorption of drugs. Molecules. 2014;19(12):20780–20807. doi:10.3390/molecules191220780

25. Tetik G, Rencber S, Ozoglu ET, Yapar EA, Karavana SY, Ozer O. Design and evaluation of topical antioxidant nanogel formulations. J Cosmet Sci. 2021;72(1):47–62.

26. Phatale V, Vaiphei KK, Jha S, Patil D, Agrawal M, Alexander A. Overcoming skin barriers through advanced transdermal drug delivery approaches. J Control Release. 2022;351:361–380. doi:10.1016/j.jconrel.2022.09.025

27. Ghasemiyeh P, Mohammadi-Samani S. Solid lipid nanoparticles and nanostructured lipid carriers as novel drug delivery systems: applications, advantages and disadvantages. Res Pharm Sci. 2018;13(4):288–303. doi:10.4103/1735-5362.235156

28. Ijaz M, Akhtar N. Fatty acids based alpha-Tocopherol loaded nanostructured lipid carrier gel: in vitro and in vivo evaluation for moisturizing and anti-aging effects. J Cosmet Dermatol. 2020;19(11):3067–3076. doi:10.1111/jocd.13346

29. Ghate VM, Lewis SA, Prabhu P, Dubey A, Patel N. Nanostructured lipid carriers for the topical delivery of tretinoin. Eur J Pharm Biopharm. 2016;108:253–261. doi:10.1016/j.ejpb.2016.07.026

30. Intagliata S, Modica MN, Santagati LM, Montenegro L. Strategies to improve resveratrol systemic and topical bioavailability: an update. Antioxidants. 2019;8(8). doi:10.3390/antiox8080244

31. Montenegro L, Panico AM, Santagati LM, Siciliano EA, Intagliata S, Modica MN. Solid lipid nanoparticles loading idebenone ester with pyroglutamic acid: in vitro antioxidant activity and in vivo topical efficacy. Nanomaterials (Basel). 2018;9(1):1.

32. Li M, Li M, Li X, et al. Preparation, characterization and ex vivo skin permeability evaluation of type I collagen-loaded liposomes. Int J Nanomed. 2023;18:1853–1871. doi:10.2147/IJN.S404494

33. Yücel Ç, Şeker Karatoprak G, Ilbasmis-Tamer S, Değim İT. Ferulic acid-loaded aspasomes: a new approach to enhance the skin permeation, anti-aging and antioxidant effects. J Drug Delivery Sci Technol. 2023;86:104748. doi:10.1016/j.jddst.2023.104748

34. Liu C, Guo X, Chen Y, et al. Anti-photoaging effect and mechanism of flexible liposomes co-loaded with apigenin and doxycycline. Biomed Pharmacother. 2023;164:114998. doi:10.1016/j.biopha.2023.114998

35. Ali S, Ranjha NM, Ahmad B, et al. Sustained release of drug facilitated through chemically crosslinked polyvinyl alcohol-gelatin (PVA-GE) hydrogels. A sustainable biomedical approach. Pol J Chem Technol. 2023;25(2):56–65. doi:10.2478/pjct-2023-0017

36. Liu C, Xia Y, Li Y, et al. Ligustrazine as an extract from medicinal and edible plant chuanxiong encapsulated in liposome-hydrogel exerting antioxidant effect on preventing skin photoaging. Polymers. 2022;14(21). doi:10.3390/polym14214778

37. Benincasa C, La Torre C, Fazio A, et al. Identification of tyrosyl oleate as a novel olive oil lipophenol with proliferative and antioxidant properties in human keratinocytes. Antioxidants. 2021;10(7):1.

38. Benincasa C, La Torre C, Plastina P, et al. Hydroxytyrosyl oleate: improved extraction procedure from olive oil and by-products, and in vitro antioxidant and skin regenerative properties. Antioxidants. 2019;8(7):1.

39. Comunian T, Babazadeh A, Rehman A, et al. Protection and controlled release of vitamin C by different micro/nanocarriers. Crit Rev Food Sci Nutr. 2022;62(12):3301–3322. doi:10.1080/10408398.2020.1865258

40. Nakyai W, Saraphanchotiwitthaya A, Viennet C, Humbert P, Viyoch J. An in vitro model for fibroblast photoaging comparing single and repeated UVA irradiations. Photochem Photobiol. 2017;93(6):1462–1471. doi:10.1111/php.12801

41. Zhang Y, Zhang M, Yao A, et al. Circ_0011129 encapsulated by the small extracellular vesicles derived from human stem cells ameliorate skin photoaging. Int J Mol Sci. 2022;23(23):1.

42. Lu B, Huang Y, Chen Z, et al. Niosomal nanocarriers for enhanced skin delivery of quercetin with functions of anti-tyrosinase and antioxidant. Molecules. 2019;24(12):2322. doi:10.3390/molecules24122322

43. Nastiti C, Ponto T, Mohammed Y, Roberts MS, Benson HAE. Novel nanocarriers for targeted topical skin delivery of the antioxidant resveratrol. Pharmaceutics. 2020;12(2):108. doi:10.3390/pharmaceutics12020108

44. Szulc-Musiol B, Sarecka-Hujar B. The use of micro- and nanocarriers for resveratrol delivery into and across the skin in different skin diseases-A literature review. Pharmaceutics. 2021;13(4):451. doi:10.3390/pharmaceutics13040451

45. Schneider LA, Raizner K, Wlaschek M, Brenneisen P, Gethoffer K, Scharffetter-Kochanek K. UVA-1 exposure in vivo leads to an IL-6 surge within the skin. Exp Dermatol. 2017;26(9):830–832. doi:10.1111/exd.13286

46. Vostalova J, Rajnochova Svobodova A, Galandakova A, Sianska J, Dolezal D, Ulrichova J. Differential modulation of inflammatory markers in plasma and skin after single exposures to UVA or UVB radiation in vivo. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 2013;157(2):137–145. doi:10.5507/bp.2013.036

47. Wu S, Gao J, Dinh QT, Chen C, Fimmel S. IL-8 production and AP-1 transactivation induced by UVA in human keratinocytes: roles of D-alpha-tocopherol. Mol Immunol. 2008;45(8):2288–2296. doi:10.1016/j.molimm.2007.11.019

48. Kim S, Kim Y, Kim JE, Cho KH, Chung JH. Berberine inhibits TPA-induced MMP-9 and IL-6 expression in normal human keratinocytes. Phytomedicine. 2008;15(5):340–347. doi:10.1016/j.phymed.2007.09.011

49. Buenger J, Driller H. Ectoin: an effective natural substance to prevent UVA-induced premature photoaging. Skin Pharmacol Physiol. 2004;17(5):232–237. doi:10.1159/000080216

50. Duteil L, Queille-Roussel C, Aladren S, et al. Prevention of polymorphic light eruption afforded by a very high broad-spectrum protection sunscreen containing ectoin. Dermatol Ther. 2022;12(7):1603–1613. doi:10.1007/s13555-022-00755-5

51. Herndon JH, Jiang LI, Kononov T, Fox T. An open label clinical trial to evaluate the efficacy and tolerance of a retinol and vitamin C facial regimen in women with mild-to-moderate hyperpigmentation and photodamaged facial skin. J Drugs Dermatol. 2016;15(4):476–482.

52. Kelm RC, Zahr AS, Kononov T, Ibrahim O. Effective lightening of facial melasma during the summer with a dual regimen: a prospective, open-label, evaluator-blinded study. J Cosmet Dermatol. 2020;19(12):3251–3257. doi:10.1111/jocd.13787

53. Zhou X, Cao Q, Orfila C, Zhao J, Zhang L. Systematic review and meta-analysis on the effects of astaxanthin on human skin ageing. Nutrients. 2021;13(9):2917. doi:10.3390/nu13092917

54. Lima SGM, Freire M, Oliveira VDS, Solisio C, Converti A, de Lima AAN. Astaxanthin delivery systems for skin application: a review. Mar Drugs. 2021;19(9):511. doi:10.3390/md19090511

55. Geng Q, Zhao Y, Wang L, Xu L, Chen X, Han J. Development and evaluation of astaxanthin as nanostructure lipid carriers in topical delivery. AAPS Pharm Sci Tech. 2020;21(8):318. doi:10.1208/s12249-020-01822-w

56. Guimaraes D, Cavaco-Paulo A, Nogueira E. Design of liposomes as drug delivery system for therapeutic applications. Int J Pharm. 2021;601:120571. doi:10.1016/j.ijpharm.2021.120571

57. Gorain B, Al-Dhubiab BE, Nair A, Kesharwani P, Pandey M, Choudhury H. Multivesicular liposome: a lipid-based drug delivery system for efficient drug delivery. Curr Pharm Des. 2021;27(43):4404–4415. doi:10.2174/1381612827666210830095941

58. Bownik A, Stepniewska Z. Ectoine as a promising protective agent in humans and animals. Arh Hig Rada Toksikol. 2016;67(4):260–265. doi:10.1515/aiht-2016-67-2837

59. Hama S, Takahashi K, Inai Y, et al. Protective effects of topical application of a poorly soluble antioxidant astaxanthin liposomal formulation on ultraviolet-induced skin damage. J Pharm Sci. 2012;101(8):2909–2916. doi:10.1002/jps.23216

60. Yoshihisa Y, Rehman MU, Shimizu T. Astaxanthin, a xanthophyll carotenoid, inhibits ultraviolet-induced apoptosis in keratinocytes. Exp Dermatol. 2014;23(3):178–183. doi:10.1111/exd.12347

61. Tominaga K, Hongo N, Karato M, Yamashita E. Cosmetic benefits of astaxanthin on humans subjects. Acta Biochim Pol. 2012;59(1):43–47. doi:10.18388/abp.2012_2168

62. He H, Olesen CM, Pavel AB, et al. Tape-strip proteomic profiling of atopic dermatitis on dupilumab identifies minimally invasive biomarkers. Front Immunol. 2020;11:1768. doi:10.3389/fimmu.2020.01768

63. Sun J, Zhao Y, Hu J. Curcumin inhibits imiquimod-induced psoriasis-like inflammation by inhibiting IL-1beta and IL-6 production in mice. PLoS One. 2013;8(6):e67078. doi:10.1371/journal.pone.0067078

64. Slawinska M, Zablotna M, Glen J, Lakomy J, Nowicki RJ, Sobjanek M. STAT3 polymorphisms and IL-6 polymorphism are associated with the risk of basal cell carcinoma in patients from northern Poland. Arch Dermatol Res. 2019;311(9):697–704. doi:10.1007/s00403-019-01952-7

65. Rose-John S. Interleukin-6 Family Cytokines. Cold Spring Harb Perspect Biol. 2018;10(2):a028415. doi:10.1101/cshperspect.a028415

66. Guttman-Yassky E, Diaz A, Pavel AB, et al. Use of tape strips to detect immune and barrier abnormalities in the skin of children with early-onset atopic dermatitis. JAMA Dermatol. 2019;155(12):1358–1370. doi:10.1001/jamadermatol.2019.2983

67. Lemster BH, Carroll PB, Rilo HR, Johnson N, Nikaein A, Thomson AW. IL-8/IL-8 receptor expression in psoriasis and the response to systemic tacrolimus (FK506) therapy. Clin Exp Immunol. 1995;99(2):148–154. doi:10.1111/j.1365-2249.1995.tb05525.x

68. Michel G, Mirmohammadsadegh A, Olasz E, et al. Demonstration and functional analysis of IL-10 receptors in human epidermal cells: decreased expression in psoriatic skin, down-modulation by IL-8, and up-regulation by an antipsoriatic glucocorticosteroid in normal cultured keratinocytes. J Immunol. 1997;159(12):6291–6297. doi:10.4049/jimmunol.159.12.6291

69. Ogilvie AL, Luftl M, Antoni C, Schuler G, Kalden JR, Lorenz HM. Leukocyte infiltration and mRNA expression of IL-20, IL-8 and TNF-R P60 in psoriatic skin is driven by TNF-alpha. Int J Immunopathol Pharmacol. 2006;19(2):271–278. doi:10.1177/039463200601900204

70. Grange PA, Raingeaud J, Calvez V, Dupin N. Nicotinamide inhibits Propionibacterium acnes-induced IL-8 production in keratinocytes through the NF-kappaB and MAPK pathways. J Dermatol Sci. 2009;56(2):106–112. doi:10.1016/j.jdermsci.2009.08.001

71. Alves-Filho JC, Marcel Silva Melo B, Ryffel B. MMP-9 mediates cross-talk between neutrophils and endothelial cells in psoriasis. J Invest Dermatol. 2021;141(4):716–718. doi:10.1016/j.jid.2020.09.006

72. Harper JI, Godwin H, Green A, et al. A study of matrix metalloproteinase expression and activity in atopic dermatitis using a novel skin wash sampling assay for functional biomarker analysis. Br J Dermatol. 2010;162(2):397–403. doi:10.1111/j.1365-2133.2009.09467.x

73. Meng C, Chen X, Li J, Wu Y, Liu H. Expression of MMP-9 and TIMP-1 in lesions of systemic sclerosis and its implications. J Huazhong Univ Sci Technolog Med Sci. 2008;28(4):480–482. doi:10.1007/s11596-008-0424-y

74. Zhou W, Duan Z, Zhao J, Fu R, Zhu C, Fan D. Glucose and MMP-9 dual-responsive hydrogel with temperature sensitive self-adaptive shape and controlled drug release accelerates diabetic wound healing. Bioact Mater. 2022;17:1–17. doi:10.1016/j.bioactmat.2022.01.004

75. Potapovich AI, Kostyuk TV, Ishutina OV, Shutava TG, Kostyuk VA. Effects of native and particulate polyphenols on DNA damage and cell viability after UV-C exposure. Naunyn Schmiedebergs Arch Pharmacol. 2023;396(9):1923–1930. doi:10.1007/s00210-023-02443-3

76. Inglut CT, Sorrin AJ, Kuruppu T, et al. Immunological and toxicological considerations for the design of liposomes. Nanomaterials. 2020;10(2):190. doi:10.3390/nano10020190

77. Ikehata H. Mechanistic considerations on the wavelength-dependent variations of UVR genotoxicity and mutagenesis in skin: the discrimination of UVA-signature from UV-signature mutation. Photochem Photobiol Sci. 2018;17(12):1861–1871. doi:10.1039/c7pp00360a

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.