Next Article in Journal
The Highly Regioselective Synthesis of Novel Imidazolidin-2-Ones via the Intramolecular Cyclization/Electrophilic Substitution of Urea Derivatives and the Evaluation of Their Anticancer Activity
Next Article in Special Issue
Artemisia anomala Herba Alleviates 2,4-Dinitrochlorobenzene-Induced Atopic Dermatitis-Like Skin Lesions in Mice and the Production of Pro-Inflammatory Mediators in Tumor Necrosis Factor Alpha-/Interferon Gamma-Induced HaCaT Cells
Previous Article in Journal
Bioactive Molecules for Discriminating Robinia and Helianthus Honey: High-Performance Liquid Chromatography–Electron Spray Ionization–Mass Spectrometry Polyphenolic Profile and Physicochemical Determinations
Previous Article in Special Issue
Santamarine Shows Anti-Photoaging Properties via Inhibition of MAPK/AP-1 and Stimulation of TGF-β/Smad Signaling in UVA-Irradiated HDFs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advantages of Hyaluronic Acid and Its Combination with Other Bioactive Ingredients in Cosmeceuticals

by
Anca Maria Juncan
1,2,3,*,
Dana Georgiana Moisă
3,*,
Antonello Santini
4,
Claudiu Morgovan
3,*,
Luca-Liviu Rus
3,
Andreea Loredana Vonica-Țincu
3 and
Felicia Loghin
1
1
Department of Toxicology, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 6 Pasteur Str., 400349 Cluj-Napoca, Romania
2
SC Aviva Cosmetics SRL, 71A Kövari Str., 400217 Cluj-Napoca, Romania
3
Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 2A Lucian Blaga Str., 550169 Sibiu, Romania
4
Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Napoli, Italy
*
Authors to whom correspondence should be addressed.
Molecules 2021, 26(15), 4429; https://doi.org/10.3390/molecules26154429
Submission received: 7 June 2021 / Revised: 14 July 2021 / Accepted: 20 July 2021 / Published: 22 July 2021
(This article belongs to the Special Issue Bioactive Compounds for Cosmeceuticals against Skin Diseases)

Abstract

:
This study proposes a review on hyaluronic acid (HA) known as hyaluronan or hyaluronate and its derivates and their application in cosmetic formulations. HA is a glycosaminoglycan constituted from two disaccharides (N-acetylglucosamine and D-glucuronic acid), isolated initially from the vitreous humour of the eye, and subsequently discovered in different tissues or fluids (especially in the articular cartilage and the synovial fluid). It is ubiquitous in vertebrates, including humans, and it is involved in diverse biological processes, such as cell differentiation, embryological development, inflammation, wound healing, etc. HA has many qualities that recommend it over other substances used in skin regeneration, with moisturizing and anti-ageing effects. HA molecular weight influences its penetration into the skin and its biological activity. Considering that, nowadays, hyaluronic acid has a wide use and a multitude of applications (in ophthalmology, arthrology, pneumology, rhinology, aesthetic medicine, oncology, nutrition, and cosmetics), the present study describes the main aspects related to its use in cosmetology. The biological effect of HA on the skin level and its potential adverse effects are discussed. Some available cosmetic products containing HA have been identified from the brand portfolio of most known manufacturers and their composition was evaluated. Further, additional biological effects due to the other active ingredients (plant extracts, vitamins, amino acids, peptides, proteins, saccharides, probiotics, etc.) are presented, as well as a description of their possible toxic effects.

Graphical Abstract

1. Introduction

Hyaluronic acid (HA) is a polysaccharide belonging to the glycosaminoglycans, made up of disaccharide units constituted of N-acetylglucosamine and D-glucuronic acid (Figure 1). It is a component of the connective, epithelial, and neural tissues and it represents a substantial constituent of the extracellular matrix (ECM) [1,2,3,4,5,6]. HA was discovered for the first time in the vitreous humour of the eye in 1934, and in 1964 it was synthesized in vitro [7,8,9]. HA has a wide range of molecular weights ranging from 2 × 105 to 107 Da [10,11,12,13]. The HA average molecular weight can influence its physico-chemical properties [3,14].
Among the many biological effects, HA is involved in cell differentiation, embryological development, inflammation, wound healing, viscoelasticity, etc. [15]. As it has been observed, the molecular mass and the mode of its synthesis or degradation define the HA biological effects [3,16,17]. By a passive mechanism, high molecular weight HA (HMW-HA) permits the tissue hydration, contributes to the osmotic balance, and stabilizes the ECM structure. On the other hand, HA interacts with different receptor binding proteins, and its molecular weight can influence the receptor affinity or its uptake by the cells, leading to opposite effects. For example, HMW-HA inhibits the cell growth (angiogenetic activity) and protects the articular cartilage due to its lubrication properties. Low molecular weight HA (LMW-HA) has angiogenetic activity and can induce tumor progression or presents pro-inflammatory activity [15,16]. Thus, the biological activity of HA is due to its binding to different receptors. For example, the binding HA-CD44 transmembrane receptor mediates cell adhesion and migration in many physiological or pathophysiological processes: (a) angiogenesis; (b) ECM structure (linking the HA with cytoskeleton); (c) inflammation (upregulation of the receptors overexpresses the interleukin-1); (d) wound healing; (e) malignant tumors (e.g., pancreatic, breast, lung, etc.). The CD-168 receptor (Receptor for Hyaluronan-Mediated Motility, RHAMM) localized on the cell surface has an important relevance in cell migration. When the receptor is situated intracellularly, it affects the activity of the mitotic spindle. As a result, the HA-RHAMM links can influence the inflammation and tissue repair processes. The HARE (Hyaluronan Receptor for Endocytosis) receptors modulate the glycosaminoglycans clearance. The lymph absorption of HA, implicitly the HA turnover, is controlled by LYVE1 (Lymphatic Vessel Endothelial Hyaluronan receptor-1). As a result, HA-LYVE1 interaction influences the tissue biomechanical properties, including its hydration. Referring to the HA interaction with TLRs (Toll-Like Receptor), it is noticed that LMW-HA has an inflammatory effect, because of its agonist activity on TLR-2 and TLR-4. On the other hand, a high mass of HA decreases the binding capacity to the receptors, forming a dense coat around the cell and covering the receptor surface [15,16,18,19].
Nowadays, there are a lot of studies conducted in order to elucidate the mechanism of action and the biosynthetic pathways of HA, or to optimize its biotechnological production, in order to synthesize derivatives with superior properties and to improve its therapeutic utilization [16].
The list of substances that are restricted or prohibited in the EU for use in cosmetic products does not include hyaluronic acid and sodium hyaluronate (NaHA). As an example, using hyaluronic acid, sodium hyaluronate, or potassium hyaluronate (KHA) in cosmetics is not restricted in Japan [20].
Some studies realized by the Cosmetic Ingredient Review (CIR) experts panel, based on the application of cosmetic HA in various concentrations, showed acute, short-term, or chronic toxicity [20]. Additionally, some side tolerable effects (scaling, erythema, and pruritus) were observed, after the use of a topical product with hyaluronic acid (0.01%), hydroquinone (4%), and glycolic acid (10%) for melasma treatment [4]. Subsequently, HA and NaHA can be nebulized and used in cosmetic products which can be applied as aerosols (e.g., hair spray) [20]. The nebulized particles cand be stored at different levels of the respiratory system, depending on their size and concentration. Because of this fact, safety assessment of cosmetic aerosols is an important issue. The protective effect of HA on the respiratory system was noticed in some studies [21], however the propellant gas, vapors, and other soluble compounds (e.g., alkanes, alcohols, stabilization polymers, bentonite, aluminium chlorhydrate, perfume oils, cosmetic colorings, complexation agents, lanolin derivates, plant extracts, etc.) associated with hyaluronan in cosmetic aerosols could induce respiratory sensitization effects such as: rhinitis, conjunctivitis, wheeze, dyspnea, or asthma. Moreover, the insoluble particles from aerosols could be responsible for pulmonary overload, leading to chronic toxicity (e.g., chronic inflammation, fibrosis, including lung tumor). These effects are related to their concentration, exposure duration, or particle size. For example, the assessment of the inhalation toxicity of products with insoluble particles with a size below 10 μm is recommended. Regarding these aerosols, an exposure duration of 5 min is indicated, and also it is necessary to avoid the exposure to fine droplets of lipophilic substances, which could produce “acute respiratory syndrome” [22].
Due to various biological activities, HA products are increasingly in demand. Thus, in 2016, the total market of HA (pharmaceuticals, beauty, and personal care) exceeded 141 tones and it is expected to grow more than 30% in 2021 (Figure 2). The most significant increasing of HA market is estimated to be in Europe and Asia [23].
Some formulations containing HA are already available on the market, with a large experience in their use. At the same time, for other products it is necessary to perform subsequent investigations to confirm their efficacy. HA is a special moisturizing active ingredient, used in cosmetics, particularly formulated as emulsions or serums, claiming hydration and skin elasticity effect. These skin biophysical parameters are closely related to anti-wrinkle effect, but no rigorous scientific evidence does justify this statement completely. Additionally, it should be taken into consideration that the efficacy of hyaluronic acid depends largely on the molecular weight [10].
Hyaluronic acid is one of the most efficient and safe ingredients used frequently in cosmetics. HA properties can be improved by other bioactive ingredients (e.g., plant extracts, vitamins, amino acids, peptides, proteins, minerals, saccharides, probiotics, etc.). Nowadays, there are a multitude of cosmetics containing HA, marketed by different manufacturers. The previously published papers present separately these advantages of HA or bioactive ingredients. In our paper, we present firstly the biological effect of HA on skin level, after which the portfolio of some popular manufacturers was analyzed, commercially cosmetic brands and products containing HA were identified, and their declared qualitative composition was evaluated. Subsequently, the additional biologic effects and the toxicological potential of the other active ingredients were presented.

2. Applications of Hyaluronic Acid

Taking into account its biological actions, physico-chemical properties, its biocompatibility or safety profile, HA has multiple applications. Figure 3 depicts the utilization of HA and its derivates in: medical (arthrology, cancer therapy, pneumology, odontology, ophthalmology, otolaryngology, rhinology, soft tissue regeneration, urology, wound treatment, etc.), pharmaceutical (e.g., drug delivery systems), nutritional (nutraceuticals, nutricosmeceuticals), or cosmetic field [3,8,9].
Being an important component of the ECM and due to its available derivatization scenarios, HA is widely used in drug delivery through several routes: cutaneous, ocular (intravitreal, periocular, subretinal), topical, nasal, oral, etc. HA can be conjugated with drug molecules (in the form of prodrugs) or can be incorporated in several molecular architectures (nanoparticles, microparticles, microspheres, gels, polyplexes, polymersomes, liposomes, micelles, implants, etc.). The resulting HA structures possess superior physico-chemical properties and higher therapeutic efficacy. A brief list of HA applications in drug delivery includes: targeting for skin diseases, cancer therapy, and controlled release of proteins, antiseptics, and antibiotics [16,24,25,26,27,28,29,30].
Normal body cells have a poor expression of HA receptors while many tumor cells generate overexpressed receptors that bind HA. This fact can lead to several approaches in cancer therapy, involving HA. Firstly, the conjugation of paclitaxel (PXT) and docetaxel (DOX), should be mentioned. PXT alone is not suitable for intravenous injection due to its hydrophobicity and adverse events. The PXT-HA conjugate is hydrophilic enough and seems to overcome limitations. Hydrophobic drug molecules can be loaded in HA micelles in order to achieve target delivery to cancer cells. Both lipophilic and hydrophilic drugs can be loaded in polymersomes. The main advantages of the previously mentioned structure modulations are solubility increase and targeting CD44 receptors on tumor cells. Modifying mesoporous silica nanoparticles with HA leads to an increased uptake in case of CD44 over expressing cells. Other nanomaterials with a potential efficiency in cancer therapy include dendrimers and liposomes. Additionally, HA coated nanoparticles (NP) are of considerable interest in cancer therapy. Several HA based nanomaterials are used in hyperthermia (an increase in the temperature of the cancer cells at about 42–46°C): NIR-loaded nanoparticles, gold nanoparticles, functionalized graphene, oxide nanoparticles, Prussian Blue nanoparticles, and other particles (related to magnetic hyperthermia treatment). In addition, HA based nanoparticles were used in photodynamic therapy, immunotherapy, and sonodynamic therapy [31,32,33,34,35,36,37].
Two major steps in wound healing (in which HA is involved) are inflammation and angiogenesis. Several biomaterials (wound dressings based on HA or combinations of HA with other biopolymers) were synthetized and tested: sponges, films, hydrogels, and electrospun membranes. The main advantages of incorporating HA in these biomaterials are: porosity and swelling improvement, together with exudate absorption. Tissue engineering uses HA for the regeneration and reconstruction of several tissues: cartilage, ocular tissues, skin, vascular tissue, adipose tissue, and peripheral nerve [38,39,40,41,42,43,44,45].
The intended use of HA in dentistry is mainly the regeneration of soft tissues, but also wound healing and regeneration of hard tissues. HA may be used as a co-material (together with other biopolymers) in several procedures related to dentistry: papilla reconstruction, osseointegration of implants, sinus lifting, periodontitis, and stomatitis therapy [46,47,48,49].
Orthokeratology is a treatment for correcting patients’ refractive error by means of wearing a special lens overnight. Viscous artificial tears (based on HA) proved to be superior (in terms of patients’ comfort) in comparison to saline solution, when used for the fitting of the orthokeratology lenses. Nisin was grafted on HA by means of amide bonds. Biocidal capacity of this modified polysaccharide (incorporated in solutions or gels) was tested on Gram positive organisms with promising results. Additionally, HA conjugated with ciprofloxacin and vancomycin were used for the prevention of the infections in ophthalmic surgery. Ophthalmic viscoelastic devices (OVDs) are used during cataract surgery due to their multiple advantages. However, long retention times of OVDs can lead to an increase in intraocular pressure (IOP). The use of two OVDs (Healon GV—1.8% sodium hyaluronate and Healon 5—2.3% sodium hyaluronate) showed a non-significant increase of IOP. The use of artificial tears is the most common therapeutic solution for dry eye syndrome. Tears based on HA and carmellose (carboxymethylcellulose) proved to be superior in terms of stability of the tear film and quality of vision in comparison with normal saline solution [26,50,51,52,53,54].
Physical and physico-chemical properties of HA are highly dependent on its molecular weight (MW). The main benefits of HA use in arthrology are related to the treatment of osteoarthritis, rheumatoid arthritis, and bone cancers. In the case of advanced osteoarthritis, knee joint distraction is a promising procedure for spontaneous cartilage repair, in about 8 weeks. The key factors are HA (in synovial fluid) and mesenchymal stromal cells (MSCs). MSCs are able to adhere to cartilage under the influence of HA (especially the ones with MW > 9 MDa) [55,56,57,58].
Interstitial Cystitis/Bladder Pain Syndrome is a chronic inflammatory syndrome and seems to be related to the destruction of bladder mucosa, especially glycosaminoglycan coating, both Chondroitin Sulphate (CS) and non-sulphated–HA [59,60]. A therapeutic approach is the intravesical instillation of CS or a combination of CS and HA while evaluating: Female Sexual Function Index (FSFI), Visual Analog Pain Scale, Interstitial Cystitis Syndrome, and Interstitial Cystitis Problem Index. FSFI was higher for the control group (CS group). The last three parameters were improved in a higher manner when comparing CS/HA and CS groups [60]. No adverse reactions were reported during CS or CS/HA instillations [61,62].
Vesicoureteral Reflux (VUR) affects children and is linked to the patient’s Urinary Tract Infection (UTI) history [63]. Surgery is the main therapeutic approach for VUR, but in this case complications may occur. Another approach is endoscopic injection therapy with teflon, polydimethylsiloxane, dextranomer/hyaluronic acid copolymer (Dx/HA), and polyacrylate polyalcohol copolymer. In the case of Dx/HA combination the short time success rate was high, but more studies are needed regarding long-term success rate [64].
Cystic fibrosis is an inflammatory lung disease linked with high airway levels of neutrophil elastase. Polysulfated GAGs (including polysulfated HA) are currently used for neutrophil elastase inhibition and because of their anti-inflammatory properties [65]. HA is also used in other airway related diseases: chronic sinusitis, asthma, bronchiectasis, and chronic obstructive pulmonary disease [66]. HA was successfully usedin post-operative recovery of nasal mucosa after sinus surgery when administered topically or by means of nebulisation [67,68].
Due to its anti-inflammatory and tissue regeneration properties, HA has been used for gene delivery in otology and there are some promising results in tympanic membrane perforation treatment [68,69].
The activity of fibroblasts (in the epidermis) and keratinocytes (in the dermis) seems to slow down together with age and also became less responsive to growth factors [70]. The ageing process consists of both intrinsic and extrinsic ageing which leads to a reduction of HA in the skin [71]. Because of the great number of polar groups present in its molecule, hyaluronic acid is a hydrophilic macromolecule with anti-ageing and hydrating claims. In aqueous solutions it can form viscoelastic gels, and when it is applied to the skin it ensures moisturizing, firming, rejuvenation, and has improved wound healing effects [10,12]. In contact with water, HA has the capacity to augment its volume, having the effect of softening the wrinkles by filling the spaces between the cells of the skin forming a viscid gel matrix. The half-life of HA in the tissues, in its natural form, is of just 12–24 h. As a result, crosslinked forms of HA are used in topical and cosmetic preparations [72,73,74]. Nevertheless, the high molecular weight of HA does not allow it to penetrate the deeper layers of the skin which restricts its benefits to topical effects [75]. Many studies showed the exogenous HA significant role in the epidermis and especially in the dermis, and its involvement in remodelling, tissue repair, and healing [2,76,77,78,79,80]. In a randomized, placebo-controlled, single-blind trial (daily oral intake, for 60 days, of 200 mg of hyaluronic acid, 500 mg of L-carnosine, and 400 mg of methylsulfonyl methane) it was proven that skin hydration and elasticity were improved and glabellar sebaceous secretion decreased [81]. Ingestion of HA/hyaluronans can improve skin moisture content and reduce ageing symptoms and signs [82,83].

3. Use of Hyaluronic Acid in Cosmetology

Nowadays, HA is one of the most widely used active ingredients in cosmetic formulations. General perception about skin regeneration is of constant interest for both industry professionals and consumers. It is evident that the skin is an indicator of individuals’ health and HA is one of the main factors for healthy skin [84]. As shown above, hyaluronic acid is a biopolymer considered of primary interest from a scientific point of view, due to its multitude of applications in cosmetic and biomedical fields. Such being the case, exploration on this ingredient is increasing in many interdisciplinary domains targeting, on the one hand, the improvement of production processes in terms of biotechnology and on the other hand the development of new formulations incorporating hyaluronan or HA-based innovative ingredients. Scientific efforts are moving nowadays towards the production of appropriate molecular weight biopolymers. This specific aspect relies precisely to the biological function, as indicated by bibliographic studies. Although HA was synthesized a very long time ago, it is still needed to investigate this active ingredient in terms of physico-chemical and biological properties [16].
HA has a multitude of applications based on specific properties such as: (1) high hygroscopicity; (2) viscoelastic nature; (3) biocompatibility; (4) non-immunogenicity. Nevertheless, the HA skin penetration mechanism is still barely understood. A multitude of factors are studied, including the existence of HA receptors for an active transport and a particular structure of the hydrated HA. The general hydration effect of the skin may also optimize dermal absorption of active ingredients and can assist their retention within the moisturized epidermal layers. HA is appropriate for biomacromolecules because it ensures protein stabilizing properties. However, the precise mechanism for the transdermal transport of HA remains to be elucidated [10,85,86].
In wound regeneration, HA has mainly cosmetic applications. In skin care formulations, it can be used as a moisturizing component, because of its hydrophilic nature. Using cosmetic products such as creams or lotions that contain HA helps to moisturize the skin and to improve elasticity, thereby decreasing the depth of wrinkles. It is assumed that, when applied onto the surface of the skin, HA solutions form an occlusive layer, absorb moisture, thereby hydrating the skin, and default wrinkles filling occurs. HA is assumed to stimulate the migration of epidermal cells. Additionally, the occlusive properties given by HA may allow the biologically active substances incorporated in cosmetics to persist in the skin layers and possibly make it easier for them to penetrate the epidermis. According to previous studies, some cosmetic HA products have been proven efficient in protecting the skin from UV irradiation. At the same time, sunscreen products containing hyaluronic acid help to maintain a firmer skin, protecting it from the injurious impact of UV radiation, due to the potential antioxidant effect of HA [87].
In cosmetic formulations, hyaluronic acid has the function of a viscosity modifier and/or a skin conditioning agent. HA is mainly used in anti-ageing cosmetic products. LMW-HA has the ability to enhance the level of moisture of the skin and expedite regeneration. HMW-HA forms a viscoelastic film when applied onto the skin and has a moisturizing effect. The main action of the HMW-HA polymer is film forming and it reduces evaporation of water from the skin and thus possessesan occlusive effect. Additionally, HMW-HA, Medium molecular weight (MMW-HA), and LMW-HA hygroscopic properties justify the ability to maintain skin hydration [87,88].
HA is also of particular importance as a delivery system of active ingredients. Currently, there are some commercially available formulations incorporating actives in different concentrations. These products are designated for the topical treatment of actinic keratosis and skin inflammatory diseases. In fact, it has been proven that HA enhances the penetration of the active ingredient through the stratum corneum (SC), which behaves as a barrier to the entry of the molecule into the deeper layers of the skin, and the holding and locating the active ingredient in the epidermis. Topical preparations containing HA in formulation are used for their healing properties, decreasing the skin irritation. A topical preparation that contains HA (0.2% w/w sodium hyaluronate (NaHA)) as a main component is currently available for the amelioration of acute and chronic wounds (areas of grafted skin, post-surgical incisions, etc.) [13,77,88].
A significant number of in vitro and in vivo studies have shown the effectiveness of HA treatment as: anti-inflammatory, skin regeneration and chondro-protective effect, anti-ageing and immunosuppressive effects, etc.
Although hyaluronan has various applications, subsequent research and technological development are needed, because there are currently certain issues to be elucidated. Firstly, further consideration of aspects regarding HA metabolism and receptor clustering analysis is necessary in order to explain the various biological actions and to foresee the effects that canvary with the molecular weight of HA. Some pharmaceuticals and/or cosmetics can incorporate HA with different molecular weight. Thus, studies are necessary for assessing the implications of molecular weight in the HA effects. Next-generation products with derivatives of crosslinked HA-conjugated polymer-delivery systems and drug substances should be developed, granting a high level of biocompatibility, prolonged half-life, and permanent in situ performance. Therefore, clinical exploration is imperative to fully characterize the safety and efficacy profile of these substances. So far, recent in vitro studies have shown promising results regarding the safety and efficacy of these promising and novel compounds: for example, HA-CL (urea-crosslinked hyaluronic acid) showed a significant biocompatibility with human corneal epithelial cell, having antioxidant, anti-inflammatory, and skin regeneration properties [16,24].
HA is used in cosmetic formulations in concentrations ranging from 0.2 to 1%. The maximum concentration of NaHA in a body lotion is 2%. When a rate of 1 mg/cm2 of a product is applied, the contribution of hyaluronic acid is 0.02 mg/cm2 of skin [20].
Interest in using hyaluronic acid as a cosmetic ingredient in skin care products occurred with the discovery that the amount of HA found in natural skin diminishes with age, and when reintroduced into the skin care products, it keeps skin hydrated, attenuates the appearance of wrinkles, and smooths the skin. HA has many qualities that make it superior to other substances used in skin regeneration, with pronounced moisturizing and anti-ageing effects [87,88]. Biological activity and HA penetration into the skin depends on the molecular weight of this substance showing different effects on the skin, as presented in Figure 4.
It has been demonstrated by some researchers, that HA has extraordinary cosmetic and nutricosmetic efficacy in improving diverse skin imperfections such as wrinkles, periorbital and nasolabial folds, and skin ageing. These types of effects of HA have been correlated with their capacity to induce the augmentation of soft tissue, to hydrate the skin, stimulate collagen, and rejuvenate the faceas summarized in Figure 5 [89].

3.1. Hydratation Effect of HA in Cosmetic Formulations

The amount of hyaluronic acid synthesized is more substantial in the epidermis than in the dermis. Since the dermis is much thicker than the epidermis, it comprises four to nine times more HA, but it was demonstrated that for equivalent tissue quantities, the epidermis synthesizes four times more hyaluronic acid than the dermis. In the epidermis, HA is located in the intercellular matrix of the basal and spinous layers. Similarly as in the dermis, the hygroscopic properties of the hyaluronic acid are of substantial relevance in hydrating the deep layers of the epidermis, but its contribution goes further than conventional hydration [90,91,92].
HA, which has the ability to bind water up to 1000 times its volume, has a relevant contribution to cellular growth, adhesion, and membrane receptor function. The major biologic role of HA in the intercellular matrix is to reinforce the intercellular structures and to produce the elastoviscous fluid matrix that firmly envelops collagen and elastin fibers. HA holds moisture, and provides firmness and radiance to the skin as well [93,94]. HA can be used topically to regenerate the skin and support hydration, although its very high molecular weight prevents its penetration through the SC [95,96].

3.2. Anti-Ageing Effect of HA in Cosmetic Formulations

HA also has an important role regarding skin ageing. Cells lose their ability to produce HA with ageing. The skin becomes drier, thinner, and looser, leading to wrinkling, among other significant changes [97]. Skin ageing is also associated with a decrease of skin moisture. Hyaluronic acid (hyaluronan) has a unique capacity to link and retain water molecules [98]. As it was shown, hyaluronic acid is a natural component that is present in the whole body. In a 70 kg individual there are 15 g of hyaluronic acid, 5 g of which are replaced daily. HA is naturally and constantly renewed because of its rapid degradation, but its renewal tends to slow with age and external aggressions. Therefore it is necessary to act very early, sustaining an optimal hyaluronic acid turnover, similar to that of young skin, in order to prevent the signs of ageing [76,99,100,101].
In relation to its biological effects at skin level, it is known that hyaluronic acid is actively involved in skin cell signaling (by binding the CD44 and LYVE-1 receptors) and thus influences the ECM stability. It has been noticed that HA has an impact on the growth of keratinocytes which protect the epidermis from ageing [10,16,79,93,102]. Hyaluronic acid is used in cosmetic preparations for its elasticity effect and for giving shape to the periorbital area after HA cosmetic treatment [103]. Additionally, the chemical double binding structure of the D-glucuronic acid unit confers antioxidant properties to hyaluronic acid. Furthermore, HA restrains the proliferation of the skin cells via the CD44 receptor and HA also has anti-inflammatory properties on the skin [76,104].
Hyaluronic acid is applied in a multitude of anti-ageing products. For example, Figure 6 presents the effect of an anti-ageing cream incorporating 0.5% (w/w) LMW-HA (20–50 KDa) and 3% (w/w) encapsulated HMW-HA (1–1.4 MDa) on periorbital wrinkles before treatment and after 28 days of treatment (Protocol Report No. 300924/19/JSHR/Agreement No. 331/30 August 2019 JS. Hamilton Romania S.R.L) [105].

4. Cosmetic Products with HA and Its Derivates Available on the Market

The cosmetic industry has been using HA for over 20 years for its great skin moisturizing properties. In 2016, over 5900 end products launched on the market contained either HA or hydrolyzed HA, with more than 70% of these products now dedicated to the mass market and masstige market [106]. According to the price, cosmetic companies can position their products on the market as: (1) premium products; (2) mass-premium products; (3) mass-market products [107]. For marketing, it is known that the price plays a psychological role in a collective imagination, for a product with a high price, consumers attribute a high value. Premium cosmetics include products with a higher purchasing price, while mass-market products have, in general, a lower price.
HA and its sodium and potassium salts are important cosmetic ingredients that are incorporated in moisturizing and anti-ageing products. Additionally, products that contain HA represent only 5%, while more than 95% of the total products contain sodium hyaluronate [106]. Hyaluronic acid and its derivates are incorporated in a multitude of cosmetic products for eye contour, lips, facial, and neck care, anti-cellulite body care, or cosmetic color conditioning in different cosmetic categories: creams, lotions, serums, masks [93,101]. A significant number of cosmetics based on hyaluronan have been launched on the market in the last years. Some examples of last products launched in the period 2015–2020 are listed in Table 1, depending on product category or proposed use, trade name and producer, and the incorporated HA forms in the cosmetic formulation. Additionally, the market segment of HA cosmetic products is indicated.
Some manufacturers launched cosmetic products on the market, containing HA or hyaluronates in combination with other active ingredients, like botanical extracts, vitamins, probiotics, amino acids, peptides, proteins, etc. These compounds improve the cosmetic formulation qualities and benefits, awarding additional claims.

4.1. Bioactive Compounds in Cosmetics with HA and HA Derivates

Different vegetal extracts incorporated in HA available cosmetics can claim different additional effects such as antioxidant, anti-inflammatory, skin conditioning, hydrating, anti-wrinkle, skin whitening, or photoprotective properties. From the cosmetovigilance point of view, vegetal extracts are mostly considered as safe for cosmetic use, but some minor adverse effects (e.g., irritation, sensitization, allergic contact reactions) have been reported.
Literature data contains a wealth of information describing the aspects regarding composition, effects, and also adverse reactions of diverse bioactive ingredients incorporated into commercially available HA or HA derivates cosmetics. Some examples are mentioned below, describing their bioactive components, cosmetic claims, and benefits, as well as reported adverse effects as indicated in Table 2.

4.2. Other Active Ingredients in Commercialy Available HA and HA Derivates Cosmetics

Besides plant extracts, commercially available cosmetics containing HA or NaHA incorporate different categories of active ingredients (e.g., probiotics, amino acids, peptides, proteins, vitamins, saccharides, or other active compounds like allantoin, lactic acid, lecithin, urea, Superoxide Dismutase (SOD), gold, malachite extract) claiming additional effects, such as moisturizing, anti-ageing, antioxidant, keratolytic, skin lightening, depigmenting, etc. These active ingredients are considered safe when used in cosmetic products and show good skin compatibility. Minor adverse reactions like contact dermatitis were reported.
More data indicating the category of other active ingredients, the cosmetic claims, skin benefits, and reported adverse effects in commercially available hyaluronan cosmetics are presented in Table 3.

5. Conclusions

Presently, the cosmetic industry is increasingly focusing on the development and formulation of active cosmetic products and cosmeceuticals. Although HA and its derivates have a large applicability in the cosmetic practice, with a multitude of useful and interesting applications, further exploration and technological development are imperative. As shown, the efficacy of hyaluronic acid depends largely on the molecular weight, claiming different effects like hydrating, regenerating, and anti-ageing. Further consideration of aspects regarding HA metabolism and receptor clustering analysis and explanations regarding various biological changes and foreseeable effects related to the molecular weight of HA are needed.
Lately, there have been a multitude of commercially available cosmetic formulations which incorporate HA or HA derivates. Categorized from mass-market to prestige or luxury products, it is important to mention that finished products containing HA represent only a small percentage, and the majority of the total products contain sodium hyaluronate.
Cosmetics incorporating HA or NaHA also contain in their formulation different plant extracts, vitamins, amino acids, peptides, proteins, saccharides, probiotics, and even gold or malachite extract. Although these additional active ingredients can cause some minor side effects, they can raise the market price, and sustain additional claims of the cosmetic product containing HA or HA derivates.

Author Contributions

Conceptualization, A.M.J., C.M. and F.L.; methodology, A.M.J., C.M. and L.-L.R.; software, D.G.M., A.M.J. and L.-L.R.; resources, A.M.J., D.G.M., C.M., L.-L.R. and A.L.V.-Ț.; writing—original draft preparation, A.M.J., D.G.M., C.M., L.-L.R. and A.L.V.-Ț.; writing—review and editing, A.M.J., D.G.M., C.M. and L.-L.R.; visualization, A.M.J., C.M., F.L. and A.S.; supervision, F.L. and A.S.; project administration, A.M.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADAtopic dermatitis
CIRCosmetic Ingredient Review
CSChondroitin sulphate
DEJDermoepidermal junction
DOXDocetaxel
Dx/HADextranomer/hyaluronic acid copolymer
ECMExtracellular matrix
FSFIFemale sexual function index
HAHyaluronic acid
HAREHyaluronan receptor for endocytosis
HMW-HAHigh molecular weight hyaluronic acid
IOPIntraocular pressure
KHAPotassium hyaluronate
LMW-HALow molecular weight hyaluronic acid
LYVE1Lymphatic vessel endothelial hyaluronan receptor-1
MMW-HAMedium molecular weight hyaluronic acid
MSCsMesenchymal stronal cells
NaHASodium hyaluronate
NPNanoparticles
OVDOphthalmic viscoelastic device
PLOD 3Procollagen-lysine, oxoglutarate 5-dioxygenase 3
PTXPaclitaxel
RHAMMReceptor for hyaluronan-mediated motility
SCStratum corneum
SODSuperoxide dismutase
TEWLTransepidermal water loss
TLRToll-like receptor
TNF-αTumor necrosis factor-α
UTIUrinary tract infection
VURVesicoureteral reflux

References

  1. Juhlin, L. Hyaluronan in skin. J. Intern. Med. 1997, 242, 61–66. [Google Scholar] [CrossRef]
  2. Ghersetich, I.; Lotti, T.; Campanile, G.; Grappone, C.; Dini, G. Hyaluronic acid in cutaneous intrinsec aging. Int. J. Dermatol. 1994, 33, 119–122. [Google Scholar] [CrossRef]
  3. Liao, Y.H.; Jones, S.A.; Forbes, B.; Martin, G.P.; Brown, M.B. Hyaluronan: Pharmaceutical characterization and drug delivery. Drug Deliv. J. Deliv. Target. Ther. Agents 2005, 12, 327–342. [Google Scholar] [CrossRef]
  4. Ibrahim, Z.A.; Gheida, S.F.; El Maghraby, G.M.; Farag, Z.E. Evaluation of the efficacy and safety of combinations of hydroquinone, glycolic acid, and hyaluronic acid in the treatment of melasma. J. Cosmet. Dermatol. 2015, 14, 113–123. [Google Scholar] [CrossRef]
  5. Turlier, V.; Rouquier, A.; Black, D.; Josse, G.; Auvergnat, A.; Briant, A.; Dahan, S.; Gassia, V.; Saint-Martory, C.; Zakaria, W.; et al. Assessment of the clinical efficacy of a hyaluronic acid-based deep wrinkle filler using new instrumental methods. J. Cosmet. Laser Ther. 2010, 12, 195–202. [Google Scholar] [CrossRef] [PubMed]
  6. Muntean, A.C.; Juncan, A.M.; Moisa, D.G.; Vonica, A.L.; Rus, L.L.; Morgovan, C.; Gligor, F.G.; Butuca, A.; Stanila, A. Primary packaging and stability evaluation of a serum used for the periorbital area of the sensitive eye. Mater. Plast. 2019, 56. [Google Scholar] [CrossRef]
  7. Price, R.D.; Berry, M.G.; Navsaria, H.A. Hyaluronic acid: The scientific and clinical evidence. J. Plast. Reconstr. Aesthetic Surg. 2007, 60, 1110–1119. [Google Scholar] [CrossRef]
  8. Robert, L. Hyaluronan, a truly “youthful” polysaccharide. Its medical applications. Pathol. Biol. 2015, 63, 32–34. [Google Scholar] [CrossRef]
  9. Andre, P. Hyaluronic acid and its use as a “rejuvenation” agent in cosmetic dermatology. Semin. Cutan. Med. Surg. 2004, 23, 218–222. [Google Scholar] [CrossRef] [PubMed]
  10. Witting, M.; Boreham, A.; Brodwolf, R.; Vávrová, K.; Alexiev, U.; Friess, W.; Hedtrich, S. Interactions of hyaluronic acid with the skin and implications for the dermal delivery of biomacromolecules. Mol. Pharm. 2015, 12, 1391–1401. [Google Scholar] [CrossRef] [PubMed]
  11. Brown, M.B.; Jones, S.A. Hyaluronic acid: A unique topical vehicle for the localized delivery of drugs to the skin. J. Eur. Acad. Dermatol. Venereol. 2005, 19, 308–318. [Google Scholar] [CrossRef] [PubMed]
  12. Voigt, J.; Driver, V.R. Hyaluronic acid and wound healing. Wound Repair Regen. 2012, 20, 317–331. [Google Scholar] [CrossRef] [PubMed]
  13. Ferguson, E.L.; Roberts, J.L.; Moseley, R.; Griffiths, P.C.; Thomas, D.W. Evaluation of the physical and biological properties of hyaluronan and hyaluronan fragments. Int. J. Pharm. 2011, 420, 84–92. [Google Scholar] [CrossRef] [PubMed]
  14. Kakehi, K.; Kinoshita, M.; Yasueda, S.I. Hyaluronic acid: Separation and biological implications. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2003, 797, 347–355. [Google Scholar] [CrossRef]
  15. Girish, K.S.; Kemparaju, K. The magic glue hyaluronan and its eraser hyaluronidase: A biological overview. Life Sci. 2007, 80, 1921–1943. [Google Scholar] [CrossRef]
  16. Fallacara, A.; Baldini, E.; Manfredini, S.; Vertuani, S. Hyaluronic acid in the third millennium. Polymers 2018, 10, 701. [Google Scholar] [CrossRef] [Green Version]
  17. Salwowska, N.M.; Bebenek, K.A.; Żądło, D.A.; Wcisło-Dziadecka, D.L. Physiochemical properties and application of hyaluronic acid: A systematic review. J. Cosmet. Dermatol. 2016, 15, 520–526. [Google Scholar] [CrossRef]
  18. Altman, R.D.; Manjoo, A.; Fierlinger, A.; Niazi, F.; Nicholls, M. The mechanism of action for hyaluronic acid treatment in the osteoarthritic knee: A systematic review. BMC Musculoskelet. Disord. 2015, 16, 321. [Google Scholar] [CrossRef] [Green Version]
  19. Gupta, R.C.; Lall, R.; Srivastava, A.; Sinha, A. Hyaluronic Acid: Molecular Mechanisms and Therapeutic Trajectory. Front. Vet. Sci. 2019, 6, 1–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Becker, L.C.; Bergfeld, W.F.; Belsito, D.V.; Klaassen, C.D.; Marks, J.G.; Shank, R.C.; Slaga, T.J.; Snyder, P.W.; Ingredient, C.; Expert, R.; et al. Final Report of the Safety Assessment of Hyaluronic Acid, Potassium Hyaluronate, and Sodium Hyaluronate. Int. J. Toxicol. 2009, 28, 5–67. [Google Scholar] [CrossRef]
  21. Scuri, M.; Abraham, W.M. Hyaluronan blocks human neutrophil elastase (HNE)-induced airway responses in sheep. Pulm. Pharmacol. Ther. 2003, 16, 335–340. [Google Scholar] [CrossRef]
  22. Rothe, H.; Fautz, R.; Gerber, E.; Neumann, L.; Rettinger, K.; Schuh, W.; Gronewold, C. Special aspects of cosmetic spray safety evaluations: Principles on inhalation risk assessment. Toxicol. Lett. 2011, 205, 97–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Global Hyaluronic Acid Products Market Size, Share, Trends and Growth Analysis Report—Segmented By Product Type, Application and Region—Industry Forecast (2020 to 2025). Available online: https://www.marketdataforecast.com/market-reports/hyaluronic-acid-products-market (accessed on 15 April 2020).
  24. Vasvani, S.; Kulkarni, P.; Rawtani, D. Hyaluronic acid: A review on its biology, aspects of drug delivery, route of administrations and a special emphasis on its approved marketed products and recent clinical studies. Int. J. Biol. Macromol. 2019, 151, 1012–1029. [Google Scholar] [CrossRef]
  25. Nien, H.K.; Yap, W.H.; Lai, C.; Lim, H.; Goh, B.H. Hyaluronic Acid-Mediated Drug Delivery System Targeting for In fl ammatory Skin Diseases: A Mini Review. Front. Pharmacol. 2020, 11, 1–8. [Google Scholar] [CrossRef]
  26. Bayer, I.S. Hyaluronic Acid and Controlled Release: A Review. Molecules 2020, 25, 2649. [Google Scholar] [CrossRef] [PubMed]
  27. Dubashynskaya, N.; Poshina, D.; Raik, S.; Urtti, A.; Skorik, Y.A. Polysaccharides in Ocular Drug Delivery. Pharmaceutics 2019, 12, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Huang, G.; Huang, H. Application of hyaluronic acid as carriers in drug delivery. Drug Deliv. 2018, 25, 766–772. [Google Scholar] [CrossRef]
  29. Huang, G.; Huang, H. Hyaluronic acid-based biopharmaceutical delivery and tumor-targeted drug delivery system. J. Control. Release 2018, 278, 122–126. [Google Scholar] [CrossRef] [PubMed]
  30. Trombino, S.; Servidio, C.; Curcio, F.; Cassano, R. Strategies for Hyaluronic Acid-Based Hydrogel Design in Drug Delivery. Pharmaceutics 2019, 11, 407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Lee, S.Y.; Kang, M.S.; Jeong, W.Y.; Han, D.; Kim, K.S. Hyaluronic Acid-Based Theranostic Nanomedicines for Targeted Cancer Therapy. Cancers 2020, 12, 940. [Google Scholar] [CrossRef] [Green Version]
  32. Kim, J.H.; Moon, M.J.; Kim, D.Y.; Heo, Y.; Suk, H.H.; Jeong, Y.Y. Hyaluronic Acid-Based Nanomaterials for Cancer Therapy. Polymers 2018, 10, 1133. [Google Scholar] [CrossRef] [Green Version]
  33. Kim, S.; Moon, M.; Surendran, S.P.; Jeong, Y.Y. Biomedical Applications of Hyaluronic Acid-Based Nanomaterials in Hyperthermic Cancer Therapy. Pharmaceutics 2019, 11, 306. [Google Scholar] [CrossRef] [Green Version]
  34. Kim, K.; Choi, H.; Choi, E.S.; Park, M. Hyaluronic Acid-Coated Nanomedicine for Targeted Cancer Therapy. Pharmaceutics 2019, 11, 301. [Google Scholar] [CrossRef] [Green Version]
  35. Li, M.; Sun, J.; Zhang, W.; Zhao, Y.; Zhang, S.; Zhang, S.; Car, C. Drug delivery systems based on CD44-targeted glycosaminoglycans for cancer therapy. Carbohydr. Polym. 2021, 251, 117103. [Google Scholar] [CrossRef]
  36. Chis, A.A.; Dobrea, C.; Morgovan, C.; Arseniu, A.M.; Rus, L.L.; Butuca, A.; Juncan, A.M.; Totan, M.; Vonica-tincu, A.L.; Cormos, G.; et al. Applications and Limitations of Dendrimers in Biomedicine. Molecules 2020, 25, 3982. [Google Scholar] [CrossRef] [PubMed]
  37. Wickens, J.M.; Alsaab, H.O.; Kesharwani, P.; Bhise, K.; Amin, M.C.I.; Tekade, R.K.; Gupta, U.; Iyer, A.K. Recent advances in hyaluronic acid-decorated nanocarriers for targeted cancer therapy. Drug Discov. Today 2016, 22, 665–680. [Google Scholar] [CrossRef] [Green Version]
  38. Litwiniuk, M.; Krejner-Bienias, A.; Gauto, A.R.; Tomasz, G. Hyaluronic Acid in Inflammation and Tissue Regeneration. Wounds 2016, 28, 78–88. [Google Scholar] [PubMed]
  39. Schneider, H.P.; Landsman, A. Preclinical and Clinical Studies of Hyaluronic Acid in Wound Care: A Case Series and Literature Review. Wounds 2019, 31, 41–48. [Google Scholar] [PubMed]
  40. Graça, M.F.P.; Miguel, S.P.; Cabral, C.S.D.; Correia, I.J. Hyaluronic acid—Based wound dressings: A review. Carbohydr. Polym. 2020, 241, 116364. [Google Scholar] [CrossRef]
  41. Abatangelo, G.; Vindigni, V.; Avruscio, G.; Pandis, L.; Brun, P. Hyaluronic Acid: Redefining Its Role. Cells 2020, 9, 1743. [Google Scholar] [CrossRef]
  42. Ahmadian, E.; Dizaj, S.M.; Eftekhari, A.; Dalir, E.; Vahedi, P.; Hasanzadeh, A.; Samiei, M. The Potential Applications of Hyaluronic Acid Hydrogels in Biomedicine. Drug Res. 2020, 70, 6–11. [Google Scholar] [CrossRef] [PubMed]
  43. Sahana, T.G.; Rekha, P.D. Biopolymers: Applications in wound healing and skin tissue engineering. Mol. Biol. Rep. 2018, 45, 2857–2867. [Google Scholar] [CrossRef] [PubMed]
  44. Shaharudin, A.; Aziz, Z. Effectiveness of hyaluronic acid and its derivatives on chronic wounds: A systematic review. J. Wound Care 2016, 25, 585–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Vigani, B.; Rossi, S.; Sandri, G.; Bonferoni, M.C.; Caramella, C.M.; Ferrari, F. Expert Opinion on Drug Delivery Hyaluronic acid and chitosan-based nanosystems: A new dressing generation for wound care wound care. Expert Opin. Drug Deliv. 2019, 16, 715–740. [Google Scholar] [CrossRef]
  46. Al-Khateeb, R.; Olszewska-Czyz, I. Heliyon Biological molecules in dental applications: Hyaluronic acid as a companion biomaterial for diverse dental applications. Heliyon 2020, 6, e03722. [Google Scholar] [CrossRef]
  47. Casale, M.; Moffa, A.; Vella, P.; Sabatino, L.; Capuano, F.; Salvinelli, B.; Lopez, M.A.; Carinci, F.; Salvinelli, F. Hyaluronic acid: Perspectives in dentistry. A systematic review. Int. J. Immunopathol. Pharmacol. 2016, 29, 572–582. [Google Scholar] [CrossRef]
  48. Vasilyev, A.V.; Kuznetsova, V.S.; Bukharova, T.B.; Grigoriev, T.E.; Zagoskin, Y.; Korolenkova, M.V.; Zorina, O.A.; Chvalun, S.N.; Goldshtein, D.V.; Kulakov, A.A. Development prospects of curable osteoplastic materials in dentistry and maxillofacial surgery. Heliyon 2020, 6, e04686. [Google Scholar] [CrossRef]
  49. Zhao, N.; Wang, X.; Qin, L.; Zhai, M.; Yuan, J.; Chen, J.; Li, D. Effect of hyaluronic acid in bone formation and its applications in dentistry. J. Biomed. Mater. Res. Part A 2016, 104, 1560–1569. [Google Scholar] [CrossRef]
  50. Carracedo, G.; Villa-Collar, C.; Martin-Gil, A.; Serramito, M.; Santamaria, L. Comparison Between Viscous Teardrops and Saline Solution to Fill Orthokeratology Contact Lenses Before Overnight Wear. Eye Contact Lens Sci. Clin. Pract. 2017, 44, S307–S311. [Google Scholar] [CrossRef]
  51. Lequeux, I.; Ducasse, E.; Jouenne, T.; Thebault, P. Addition of antimicrobial properties to hyaluronic acid by grafting of antimicrobial peptide. Eur. Polym. J. 2014, 51, 182–190. [Google Scholar] [CrossRef]
  52. Malvankar-Mehta, M.S.; Fu, A.; Subramanian, Y.; Hutnik, C. Impact of Ophthalmic Viscosurgical Devices in Cataract Surgery. J. Ophthalmol. 2020, 1–17. [Google Scholar] [CrossRef] [PubMed]
  53. Vandermeer, G.; Chamy, Y.; Pisella, P.-J. Comparison of objective optical quality measured by double-pass aberrometry in patients with moderate dry eye: Normal saline vs. artificial tears: A pilot study. J. Fr. Ophtalmol. 2018, 41, e51–e57. [Google Scholar] [CrossRef]
  54. Zhang, Z.; Suner, S.S.; Blake, D.A.; Ramesh, A.S.; Sahiner, N. Antimicrobial activity and biocompatibility of slow-release hyaluronic acid- antibiotic conjugated particles. Int. J. Pharm. 2020, 576, 119024. [Google Scholar] [CrossRef]
  55. Baboolal, T.G.; Mastbergen, S.C.; Jones, E.; Calder, S.J.; Lafeber, F.P.J.G.; Mcgonagle, D. Synovial fluid hyaluronan mediates MSC attachment to cartilage, a potential novel mechanism contributing to cartilage repair in osteoarthritis using knee joint distraction. Ann. Rheum. Dis. 2015, 75, 908–915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Li, C.; Cao, Z.; Li, W.; Liu, R.; Chen, Y.; Song, Y.; Liu, G.; Song, Z.; Liu, Z.; Lu, C.; et al. A review on the wide range applications of hyaluronic acid as a promising rejuvenating biomacromolecule in the treatments of bone related diseases. Int. J. Biol. Macromol. 2020, 165, 1264–1275. [Google Scholar] [CrossRef]
  57. Snetkov, P.; Zakharova, K.; Morozkina, S.; Olekhnovich, R.; Uspenskaya, M. Hyaluronic Acid: The Influence of Molecular Weight on Structural, Physical, Physico-Chemical, and Degradable Properties of Biopolymer. Polymers 2020, 12, 1800. [Google Scholar] [CrossRef] [PubMed]
  58. Kosiński, J.; Jarecki, J.; Przepiórka-Kosińska, J.; Ratajczak, M. Hyaluronic Acid in Orthopedics. Wiad Lek. 2020, LXXIII, 1878–1881. [Google Scholar] [CrossRef]
  59. Van de Merwe, J.P.; Nordling, J.; Bouchelouche, P.; Bouchelouche, K.; Cervigni, M.; Kurosch Daha, L.; Elneil, S.; Fall, M.; Hohlbrugger, G.; Irwin, P.; et al. Diagnostic Criteria, Classification, and Nomenclature for Painful Bladder Syndrome/Interstitial Cystitis: An ESSIC Proposal. Eur. Urol. 2008, 53, 60–67. [Google Scholar] [CrossRef]
  60. Arslan, B.; Gönültaş, S.; Gökmen, E.; Özman, O.; Asım Avci, M.; Özdemir, E. Outcomes of intravesical chondroitin-sulfate and combined hyaluronic-acid/chondroitin-sulfate therapy on female sexual function in bladder pain syndrome. Int. Urogynecol. J. 2019, 30, 1857–1862. [Google Scholar] [CrossRef]
  61. Pyo, J.-S.; Cho, W.J. Systematic Review and Meta-Analysis of Intravesical Hyaluronic Acid and Hyaluronic Acid/Chondroitin Sulfate Instillation for Interstitial Cystitis/Painful Bladder Syndrome. Cell. Physiol. Biochem. 2016, 39, 1618–1625. [Google Scholar] [CrossRef] [Green Version]
  62. Riedl, C.R.; Engelhardt, P.F.; Daha, K.L.; Morakis, N.; Pflüger, H. Hyaluronan treatment of interstitial cystitis/painful bladder syndrome. Int. Urogynecol. J. 2008, 19, 717–721. [Google Scholar] [CrossRef]
  63. Edwards, A.; Peters, C.A. Managing vesicoureteral reflux in children: Making sense of all the data. F1000Research 2019, 8, F1000. [Google Scholar] [CrossRef] [Green Version]
  64. Kim, S.W.; Lee, Y.S.; Han, S.W. Endoscopic injection therapy. Investig. Clin. Urol. 2017, 58, S38–S45. [Google Scholar] [CrossRef] [Green Version]
  65. Voynow, J.A.; Zheng, S.; Kummarapurugu, A.B. Glycosaminoglycans as Multifunctional Anti-Elastase and Anti-In fl ammatory Drugs in Cystic Fibrosis Lung Disease. Front. Pharmacol. 2020, 11, 1011. [Google Scholar] [CrossRef]
  66. Máiz Carro, L.; Martínez-García, M.A. Use of Hyaluronic Acid (HA) in Chronic Airway Diseases. Cells 2020, 9, 2210. [Google Scholar] [CrossRef]
  67. Fong, E.; Garcia, M.; Woods, C.M.; Ooi, E. Hyaluronic acid for post sinus surgery care: Systematic review and meta-analysis. J. Laryngol. Otol. 2017, 131, S2–S11. [Google Scholar] [CrossRef]
  68. Pignataro, L.; Marchisio, P.; Ibba, T.; Torretta, S. Topically administered hyaluronic acid in the upper airway: A narrative review. Immunopathol. Pharmacol. 2018, 32. [Google Scholar] [CrossRef]
  69. Abi Zeid Daou, C.; Bassim, M. Hyaluronic acid in otology: Its uses, advantages and drawbacks—A review. Am. J. Otolaryngol. 2020, 41, 102375. [Google Scholar] [CrossRef]
  70. Baumann, L. How to Use Oral and Topical Cosmeceuticals to Prevent and Treat Skin Aging. Facial Plast. Surg. Clin. N. Am. 2018, 26, 407–413. [Google Scholar] [CrossRef]
  71. Genovese, L.; Sibilla, S. Innovative Nutraceutical Approaches to Counteract the Signs of Aging. In Textbook of Aging Skin; Farage, M.A., Miller, K.W., Maibach, H.I., Eds.; Springer: Berlin/Heidelberg, Germany, 2017; pp. 1967–1991. ISBN 9783662473986. [Google Scholar]
  72. Janiš, R.; Pata, V.; Egner, P.; Pavlačková, J.; Zapletalová, A.; Kejlová, K. Comparison of metrological techniques for evaluation of the impact of a cosmetic product containing hyaluronic acid on the properties of skin surface. Biointerphases 2017, 12, 021006. [Google Scholar] [CrossRef]
  73. Papakonstantinou, E.; Roth, M.; Karakiulakis, G. Hyaluronic acid: A key molecule in skin aging. Dermatoendocrinology 2012, 4. [Google Scholar] [CrossRef] [Green Version]
  74. Nobile, V.; Buonocore, D.; Michelotti, A.; Marzatico, F. Anti-aging and filling efficacy of six types hyaluronic acid based dermo-cosmetic treatment: Double blind, randomized clinical trial of efficacy and safety. J. Cosmet. Dermatol. 2014, 13, 277–287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Sakulwech, S.; Lourith, N.; Ruktanonchai, U.; Kanlayavattanakul, M. Preparation and characterization of nanoparticles from quaternized cyclodextrin-grafted chitosan associated with hyaluronic acid for cosmetics. Asian J. Pharm. Sci. 2018, 13, 498–504. [Google Scholar] [CrossRef]
  76. Mondon, P.; Doridot, E.; Ringenbach, C.; Gracioso, O. Hyaluronic acid: History and future potential. Pers. Care 2015, 6, 27–30. [Google Scholar]
  77. Neuman, M.G.; Nanau, R.M.; Oruña-Sanchez, L.; Coto, G. Hyaluronic acid and wound healing. J. Pharm. Pharm. Sci. 2015, 18, 53–60. [Google Scholar] [CrossRef] [Green Version]
  78. Elsner, P.; Maibach, H.I. Cosmeceuticals: Drugs vs. Cosmetics, 1st ed.; Marcel Dekker: New York, NY, USA, 2000; Volume 23, ISBN 0824703057. [Google Scholar]
  79. Manuskiatti, W.; Maibach, H.I. Hyaluronic acid and skin: Wound healing and aging. Int. J. Dermatol. 1996, 35, 539–544. [Google Scholar] [CrossRef]
  80. Oh, J.H.; Kim, Y.K.; Jung, J.Y.; Shin, J.; Chung, J.H. Changes in glycosaminoglycans and related proteoglycans in intrinsically aged human skin in vivo. Exp. Dermatol. 2011, 20, 454–456. [Google Scholar] [CrossRef]
  81. Guaitolini, E.; Cavezzi, A.; Cocchi, S.; Roberto, C. Randomized, Placebo-controlled Study of a Nutraceutical Based on Hyaluronic Acid, L-carnosine, and Methylsulfonylmethane in Facial Skin Aesthetics and Well-being. J. Clin. Aesthet. Dermatol. 2021, 12, 40–45. [Google Scholar]
  82. Kawada, C.; Yoshida, T.; Yoshida, H.; Matsuoka, R.; Sakamoto, W.; Odanaka, W.; Sato, T.; Yamasaki, T.; Kanemitsu, T.; Masuda, Y.; et al. Ingested hyaluronan moisturizes dry skin. Nutr. J. 2014, 13, 70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Kawada, C.; Yoshida, T.; Yoshida, H.; Sakamoto, W.; Odanaka, W.; Sato, T.; Yamasaki, T.; Kanemitsu, T.; Masuda, Y.; Urushibata, O. Ingestion of hyaluronans (molecular weights 800 k and 300 k) improves dry skin conditions: A randomized, double blind, controlled study. J. Clin. Biochem. Nutr. 2015, 56, 66–73. [Google Scholar] [CrossRef] [Green Version]
  84. Baumann, L. Cosmetic Dermatology. Principles and Practice, 2nd ed.; McGraw-Hill: New York, NY, USA, 2009; ISBN 9780071641289. [Google Scholar]
  85. Gaffney, J.; Matou-Nasri, S.; Grau-Olivares, M.; Slevin, M. Therapeutic applications of hyaluronan. Mol. Biosyst. 2010, 6, 437–443. [Google Scholar] [CrossRef]
  86. Brown, T.J.; Alcorn, D.; Fraser, J.R.E. Absorption of Hyaluronan Applied to the Surface of Intact Skin. J. Investig. Dermatol. 1999, 113, 740–746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Schiraldi, C.; La Gatta, A.; De Rosa, M. Biotechnological Production and Application of Hyaluronan. In Biopolymers; Elnashar, M.M., Ed.; InTech Europe: Rijeka, Croatia, 2010; pp. 388–412. ISBN 9789533071091. [Google Scholar]
  88. Essendoubi, M.; Gobinet, C.; Reynaud, R.; Angiboust, J.F.; Manfait, M.; Piot, O. Human skin penetration of hyaluronic acid of different molecular weights as probed by Raman spectroscopy. Ski. Res. Technol. 2016, 22, 55–62. [Google Scholar] [CrossRef] [PubMed]
  89. Bukhari, N.S.; Roswandi, N.L.; Waqas, M.; Habib, H.; Hussain, F.; Khan, S.; Sohail, M.; Ramli, N.A.; Thu, H.E.; Hussain, Z. Hyaluronic acid, a promising skin rejuvenating biomedicine: A review of recent updates and pre-clinical and clinical investigations on cosmetic and nutricosmetic effects. Int. J. Biol. Macromol. 2018, 120 Pt B, 1682–1695. [Google Scholar] [CrossRef]
  90. Morro, G.; Morvan, P.-Y.; Vallee, R. Epidermal hyaluronic acid: A new look at hydration. Pers. Care 2013, 11, 56–58. [Google Scholar]
  91. Tammi, R.; Säämämen, A.-M.; Maibach, H.I.; Tammi, M. Degradation of Newly Synthesized High Molecular Mass Hyaluronan in the Epidermal and Dermal Compartments of Human Skin in Organ Culture. J. Investig. Dermatol. 1991, 97, 126–130. [Google Scholar] [CrossRef] [Green Version]
  92. Rao, S.; Muia, F.; Bennett, S.; Lonza, J.V.G. Improving barrier function to address premature ageing. Pers. Care 2013, 9, 77–82. [Google Scholar]
  93. Pavicic, T.; Gauglitz, G.G.; Lersch, P.; Schwach-Abdellaoui, K.; Malle, B.; Korting, H.C.; Farwick, M. Efficacy of Cream-Based Novel Formulations of Hyaluronic Acid of Different Molecular Weights in Anti-Wrinkle Treatment. J. Drugs Dermatol. 2011, 10, 990–1000. [Google Scholar]
  94. Souto, E.B.; Fernandes, A.R.; Martins-Gomes, C.; Coutinho, T.E.; Durazzo, A.; Lucarini, M.; Souto, S.B.; Silva, A.M.; Santini, A. Nanomaterials for skin delivery of cosmeceuticals and pharmaceuticals. Appl. Sci. 2020, 10, 1594. [Google Scholar] [CrossRef] [Green Version]
  95. Dayan, N. Skin Aging Handbook. An Integrated Approach to Biochemistry and Product Development; Dayan, N., Ed.; William Andrew Inc.: New York, NY, USA, 2008; ISBN 9780815515845. [Google Scholar]
  96. Weindl, G.; Schaller, M.; Schäfer-Korting, M.; Korting, H.C. Hyaluronic Acid in the Treatment and Prevention of Skin Diseases: Molecular Biological, Pharmaceutical and Clinical Aspects. Skin Pharmacol. Physiol. 2004, 17, 207–213. [Google Scholar] [CrossRef] [PubMed]
  97. Lee, D.H.; Oh, J.H.; Chung, J.H. Glycosaminoglycan and proteoglycan in skin aging. J. Dermatol. Sci. 2016, 83, 174–181. [Google Scholar] [CrossRef]
  98. Mourelle, M.; Gonzalez, J. Can a cosmetic have similar impact as dermal fillers? Pers. Care 2015, 11, 73–76. [Google Scholar]
  99. Fraser, J.R.E.; Laurent, T.C.; Laurent, U.B.G. Hyaluronan: Its nature, distribution, functions and turnover. J. Intern. Med. 1997, 242, 27–33. [Google Scholar] [CrossRef]
  100. Tzellos, T.G.; Klagas, I.; Vahtsevanos, K.; Triaridis, S.; Printza, A.; Kyrgidis, A.; Karakiulakis, G.; Zouboulis, C.C.; Papakonstantinou, E. Extrinsic ageing in the human skin is associated with alterations in the expression of hyaluronic acid and its metabolizing enzymes. Exp. Dermatol. 2009, 18, 1028–1035. [Google Scholar] [CrossRef]
  101. Olejnik, A.; Gościańska, J.; Nowak, I. Significance of hyaluronic acid in cosmetic industry and aesthetic medicine. Chemik 2012, 66, 129–135. [Google Scholar]
  102. Haeusler, H. Efficacy of Hyaluronic Acid Gel to Improve Skin Properties. SOFW J. 2015, 9, 16–18. [Google Scholar]
  103. Burgess, C.M. Soft Tissue Augmentation. In Cosmetic Dermatology; Burgess, C.M., Ed.; Springer: Berlin/Heidelberg, Germany, 2005; pp. 93–109. ISBN 3540230645. [Google Scholar]
  104. Cutting, K.F. Wound healing through synergy of hyaluronan and an iodine complex. J. Wound Care 2011, 20, 424–430. [Google Scholar] [CrossRef]
  105. Juncan, A.M. Visioline VL 650. The Images of Skin Texture before Product Application (D0) and after 28 Days (D28); Report No. 300924/19/JSHR Table 3; Courage+Khazaka Electronic GmbH: Köln, Germany, 2019. [Google Scholar]
  106. Reynaud, R.; Scandolera, A.; Dinant, C.; Lefèvre, F.; Bourgon, O. A new generation of oil-compatible hydrated HA. Pers. Care 2017, 9, 61–63. [Google Scholar]
  107. Tang, C.S.; Teo, C.-P.; Wei, K.K. Supply Chain Analysis: A Handbook on the Interaction of Information, System and Optimization; Springer Science+Business Media: New York, NY, USA, 2008; ISBN 9780387752396. [Google Scholar]
  108. Available online: https://www.fresh.com/us/skincare/categories/essences-serums/rose-deep-hydration-face-serum-H00003685.html (accessed on 20 May 2021).
  109. Available online: https://www.cultbeauty.co.uk/the-ordinary-buffet.html (accessed on 1 June 2021).
  110. Available online: https://www.cultbeauty.co.uk/the-ordinary-hyaluronic-acid-2-b5.html (accessed on 20 January 2021).
  111. Available online: https://www.apivita.com/en/intensive-care-eye-serum-10-22-01-615.html (accessed on 20 January 2021).
  112. Available online: https://www.farmec.eu/products/skin/hyaluronic-acid-ampoules-5-gerovital-h3-evolution-1119.html (accessed on 1 June 2021).
  113. Available online: https://infinitumcosmetics.ro/produs/deep-wrinkles-anti-aging-serum/ (accessed on 1 June 2021).
  114. Available online: https://www.skinsociety.me/collections/skin-care-anti-aging-day-night-care/products/mysterieux-mille-et-un-jours-anti-ageing-day-emulsion-combination-to-oily-skin-garancia (accessed on 15 May 2021).
  115. Available online: https://www.balanceme.com/gb/skincare/eye-creams/ (accessed on 1 June 2021).
  116. Available online: https://earthsciencebeauty.com/products/apricot-night-cream?_pos=1&_sid=7ac9e205a&_ss=r (accessed on 15 May 2021).
  117. Available online: http://www.cosmeticplant.com/skin-type/normal-skin/lift-up-anti-wrinkle-day-cream-with-hyaluronic-acid-liftonin-xpress-and-magnolia-extract-50-ml/ (accessed on 15 May 2021).
  118. Available online: https://infinitumcosmetics.ro/produs/cellular-regenerating-cream/ (accessed on 1 June 2021).
  119. Available online: https://www.gerocossen.ro/crema-antirid-de-zi-spf-10-hyaluron-anti-age-50-ml.html (accessed on 15 May 2021).
  120. Available online: https://infinitumcosmetics.ro/produs/golden-elixir-anti-ageing-cream/ (accessed on 1 June 2021).
  121. Available online: https://www.farmec.eu/products/skin/anti-wrinkle-cream-concentrated-with-hyaluronic-acid-3-684.html (accessed on 1 June 2021).
  122. Available online: https://www.arbonne.com/Pws/homeoffice/store/AMCA/product/RE9-Advanced-for-Men-Anti-Aging-Moisturizer-Broad-1094 Spectrum-SPF-15-CA-6513,8782.aspx (accessed on 15 May 2021).
  123. Available online: https://infinitumcosmetics.ro/produs/anti-aging-cleansing-emulsion/ (accessed on 15 May 2021).
  124. Available online: https://en.eauthermalejonzac.com/product/dermo-repair-cream-40-ml/ (accessed on 15 May 2021).
  125. Available online: https://www.everglowcosmetics.com/ (accessed on 1 June 2021).
  126. Available online: https://www.naturerepuliceurope.com/it/i-nostri-prodotti/ (accessed on 1 June 2021).
  127. Available online: https://www.dm.de/search?query=Alverde Handcreme&searchType=product (accessed on 15 May 2021).
  128. Available online: https://www.innisfree.com/hk/en/product/productView.do?prdSeq=16287 (accessed on 15 May 2021).
  129. Available online: https://www.innisfree.com/sg/en/product/productView.do?prdSeq=10837 (accessed on 15 May 2021).
  130. Available online: https://jjj-shop.com/etude-house-berry-aha-bright-peel-bubble-wash-review/ (accessed on 1 June 2021).
  131. Available online: https://www.illamasqua.com/liquid-lip-lure/11283816.html (accessed on 15 May 2021).
  132. Available online: https://www.clinique.com/product/1592/41442/makeup/sun-kissed-face-gelee-complexion-multitasker?size=1.0_fl_oz (accessed on 15 May 2021).
  133. Available online: https://www.paulandjoe-beaute.hk/ProductDetails.aspx?master_sku=APAAVN (accessed on 1 June 2021).
  134. Kato, A.; Koyama, J.; Shinzawa, K.; Imaeda, S.; Adachi, I.; Nash, R.J.; Fleet, G.W.J.; Shintani, M.; Takeuchi, C.; Ishikawa, F. Ginnalin B induces differentiation markers and modulates the proliferation/differentiation balance via the upregulation of NOTCH1 in human epidermal keratinocytes. Bioorg. Med. Chem. 2019, 27, 2172–2180. [Google Scholar] [CrossRef]
  135. Muhsinah, A.B.; Ma, H.; DaSilva, N.A.; Yuan, T.; Seeram, N.P. Bioactive Glucitol-Core Containing Gallotannins and other Phytochemicals from Silver Maple (Acer saccharinum) Leaves. Nat. Prod. Commun. 2017, 12, 83–84. [Google Scholar] [CrossRef] [Green Version]
  136. Liu, C.; Guo, H.; Dain, J.; Wan, Y.; Gao, X.-H.; Chen, H.-D.; Seeram, N.P.; Ma, H. Cytoprotective Effects of A Proprietary Red Maple Leaves Extract and Its Major Polyphenol, Ginnalin A, against Hydrogen Peroxide and Methylglyoxal Induced Oxidative Stress in Human Keratinocytes. Food Funct. 2020, 11, 5105–5114. [Google Scholar] [CrossRef] [PubMed]
  137. Ma, H.; Liu, W.; Frost, L.; Kirschenbaum, L.J.; Dain, J.A.; Seeram, N.P. Glucitol-core containing gallotannins inhibit the formation of advanced glycation end-products mediated by their antioxidant potential. Food Funct. 2016, 7, 2213–2222. [Google Scholar] [CrossRef] [PubMed]
  138. Santos, G.A.; Doty, M.S. IR Studies on Carrageenan of Ahnfeltia concinna, a Marine Red Alga. J. Pharm. Sci. 1975, 64, 704–1706. [Google Scholar] [CrossRef]
  139. Cheong, K.L.; Qiu, H.M.; Du, H.; Liu, Y.; Khan, B.M. Oligosaccharides Derived from Red Seaweed: Production, Properties, and Potential Health and Cosmetic Applications. Molecules 2018, 23, 2451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Cunha, L.; Grenha, A. Sulfated Seaweed Polysaccharides as Multifunctional Materials in Drug Delivery Applications. Mar. Drugs 2016, 14, 42. [Google Scholar] [CrossRef]
  141. Yun, E.J.; Lee, S.; Kim, J.H.; Kim, B.B.; Kim, H.T.; Lee, S.H.; Pelton, J.G.; Kang, N.J.; Choi, I.; Kim, K.H. Enzymatic production of 3, 6-anhydro-L-galactose from agarose and its purification and in vitro skin whitening and anti-inflammatory activities. Appl. Microbiol. Biotechnol. 2013, 97, 2961–2970. [Google Scholar] [CrossRef]
  142. Pimentel, F.B.; Alves, R.C.; Rodrigues, F.; Oliveira, M.B.P.P. Macroalgae-Derived Ingredients for Cosmetic Industry—An Update. Cosmetics 2018, 5, 2. [Google Scholar] [CrossRef] [Green Version]
  143. Verdy, C.; Branka, J.E.; Mekideche, N. Quantitative assessment of lactate and progerin production in normal human cutaneous cells during normal ageing: Effect of an Alaria esculenta extract. Int. J. Cosmet. Sci. 2011, 33, 462–466. [Google Scholar] [CrossRef]
  144. De la Moneda, A.; Carro, M.D.; Weisbjerg, M.R.; Roleda, M.Y.; Lind, V.; Novoa-Garrido, M.; Molina-Alcaide, E. Variability and Potential of Seaweeds as Ingredients of Ruminant Diets: An In Vitro Study. Animals 2019, 9, 851. [Google Scholar] [CrossRef] [Green Version]
  145. Rahnasto-Rilla, M.K.; McLoughlin, P.; Kulikowicz, T.; Doyle, M.; Bohr, V.A.; Lahtela-Kakkonen, M.; Ferrucci, L.; Hayes, M.; Moaddel, R. The Identification of a SIRT6 Activator from Brown Algae Fucus Distichus. Mar. Drugs 2017, 15, 190. [Google Scholar] [CrossRef] [Green Version]
  146. Couteau, C.; Coiffard, L. Seaweed Application in Cosmetics. In Seaweed in Health and Disease Prevention; Fleurence, J., Levine, I., Eds.; Elsevier, Inc.: London, UK, 2016; pp. 423–441. ISBN 9780128027936. [Google Scholar]
  147. Verdy, C.; Branka, J.; Mekideche, N. Melanosome transfer evaluation by quantitative measurement of Pmel 17 in human normal melanocyte-keratinocyte co-cultures: Effect of an Alaria esculenta extract. J. Cosmet. Sci. 2012, 63, 197–203. [Google Scholar]
  148. Rajauria, G. In-Vitro Antioxidant Properties of Lipophilic Antioxidant Compounds from 3 Brown Seaweed. Antioxidants 2019, 8, 596. [Google Scholar] [CrossRef] [Green Version]
  149. Heffernan, N.; Smyth, T.J.; Soler-Villa, A.; Fitzgerald, R.J.; Brunton, N.P. Phenolic content and antioxidant activity of fractions obtained from selected Irish macroalgae species (Laminaria digitata, Fucus serratus, Gracilaria gracilis and Codium fragile). J. Appl. Phycol. 2014, 27, 519–530. [Google Scholar] [CrossRef]
  150. Janssen Cosmetics. Ingredients Information Algae. Available online: https://www.janssen-cosmetics.com/Uploads/_UNTERGRUPPE/1590_Ocean_Treasure/1950_Ingredients_Information_Algae_Ritual.pdf (accessed on 18 March 2021).
  151. Guo, X.; Mei, N. Aloe vera: A review of toxicity and adverse clinical effects. J. Environ. Sci. Heal. Part C Environ. Carcinog. Ecotoxicol. Rev. 2016, 34, 77–96. [Google Scholar] [CrossRef] [PubMed]
  152. Cosmetic Ingredient Review Expert Panel. Final Report on the Safety Assessment of Aloe andongensis Extract, Aloe andongensis Leaf Juice, Aloe arborescens Leaf Extract, Aloe arborescens Leaf Juice, Aloe arborescens Leaf Protoplasts, Aloe barbadensis Flower Extract, Aloe barbadensis Leaf, Aloe Bar. Int. J. Toxicol. 2007, 26, 1–50. [Google Scholar] [CrossRef]
  153. Dal’Belo, S.E.; Rigo Gaspar, L.; Maia Campos, P.M.B.G. Moisturizing effect of cosmetic formulations containing Aloe vera extract in different concentrations assessed by skin bioengineering techniques. Skin Res. Technol. 2006, 12, 241–246. [Google Scholar] [CrossRef]
  154. Hamman, J.H. Composition and applications of Aloe vera leaf gel. Molecules 2008, 13, 1599–1616. [Google Scholar] [CrossRef] [Green Version]
  155. Herman, A. Comparison of Antimicrobial Activity of Essential Oils, Plant Extracts and Methylparaben in Cosmetic Emulsions: 2 Months Study. Indian J. Microbiol. 2014, 54, 361–364. [Google Scholar] [CrossRef] [Green Version]
  156. Miroddi, M.; Navarra, M.; Calapai, F.; Mancari, F.; Giofrè, S.V.; Gangemi, S.; Calapai, G. Review of clinical pharmacology of Aloe vera L. in the treatment of psoriasis. Phyther. Res. 2015, 29, 648–655. [Google Scholar] [CrossRef]
  157. Ganesan, P.; Choi, D.K. Current application of phytocompound-based nanocosmeceuticals for beauty and skin therapy. Int. J. Nanomed. 2016, 11, 1987–2007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Rigat, M.; Vallès, J.; D’Ambrosio, U.; Gras, A.; Iglésias, J.; Garnatje, T. Plants with topical uses in the Ripollès district (Pyrenees, Catalonia, Iberian Peninsula): Ethnobotanical survey and pharmacological validation in the literature. J. Ethnopharmacol. 2015, 164, 162–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Casetti, F.; Wölfle, U.; Gehring, W.; Schempp, C.M. Dermocosmetics for dry skin: A new role for botanical extracts. Skin Pharmacol. Physiol. 2011, 24, 289–293. [Google Scholar] [CrossRef]
  160. Beringhs, A.O.R.; Rosa, J.M.; Stulzer, H.K.; Budal, R.M.; Sonaglio, D. Green Clay and Aloe vera Peel-Off Facial Masks: Response Surface Methodology Applied to the Formulation Design. AAPS PharmSciTech. 2013, 14, 445–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Krulj, J.; Brlek, T.; Pezo, L.; Brkljača, J.; Popović, S.; Zeković, Z.; Bodroža Solarov, M. Extraction methods of Amaranthus sp. grain oil isolation. J. Sci. Food Agric. 2016, 96, 3552–3558. [Google Scholar] [CrossRef]
  162. Huang, Z.R.; Lin, Y.K.; Fang, J.Y. Biological and Pharmacological Activities of Squalene and Related Compounds: Potential Uses in Cosmetic Dermatology. Molecules 2009, 14, 540–554. [Google Scholar] [CrossRef] [PubMed]
  163. Wołosik, K.; Knas, M.; Zalewska, A.; Niczyporuk, M.; Przystupa, A.W. The importance and perspective of plant-based squalene in cosmetology. J. Cosmet. Sci. 2013, 64, 59–65. [Google Scholar] [PubMed]
  164. De Vita, D.; Messore, A.; Toniolo, C.; Frezza, C.; Scipione, L.; Bertea, C.M.; Micera, M.; Di Sarno, V.; Madia, V.N.; Pindinello, I.; et al. Towards a new application of amaranth seed oil as an agent against Candida albicans. Nat. Prod. Res. 2019, 1–6. [Google Scholar] [CrossRef]
  165. Cho, Y.H.; Kim, J.H.; Park, S.M.; Lee, B.C.; Pyo, H.B.; Park, H.D. New cosmetic agents for skin whitening from Angelica dahurica. J. Cosmet. Sci. 2006, 57, 11–21. [Google Scholar]
  166. Kil, Y.; Pham, S.T.; Seo, K.E.; Jafari, M. Angelica keiskei, an emerging medicinal herb with various bioactive constituents and biological activities. Arch. Pharm. Res. 2017, 40, 655–675. [Google Scholar] [CrossRef] [PubMed]
  167. Son, H.-U.; Yoon, E.-K.; Cha, Y.-S.; Kim, M.-A.; Shin, Y.-K.; Kim, J.-M.; Choi, Y.-H.; Lee, S.-H. Comparison of the toxicity of aqueous and ethanol fractions of Angelica keiskei leaf using the eye irritancy test. Exp. Ther. Med. 2012, 4, 820–824. [Google Scholar] [CrossRef]
  168. Lee, S. Evaluation of acute skin irritation and phototoxicity by aqueous and ethanol fractions of Angelica keiskei. Exp. Ther. Med. 2012, 5, 45–50. [Google Scholar] [CrossRef]
  169. Paulsen, E. Contact sensitization from Compositae-containing herbal remedies and cosmetics. Contact Dermat. 2002, 47, 189–198. [Google Scholar] [CrossRef]
  170. Anonymous. Final Report on the Safety Assessment of Arnica Montana Extract and Arnica Montana. Int. J. Toxicol. 2001, 20, 1–11. [Google Scholar] [CrossRef] [PubMed]
  171. Baumann, L.S. Less-known botanical cosmeceuticals. Dermatol. Ther. 2007, 20, 330–342. [Google Scholar] [CrossRef] [PubMed]
  172. Cizauskaite, U.; Bernatoniene, J. Innovative Natural ingredients-Based Multiple Emulsions: The Effect on Human Skin Moisture, Sebum Content, Pore Size and Pigmentation. Molecules 2018, 23, 1428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Vaseli-Hagh, N.; Deezagi, A.; Shahraki, M.K. Anti-aging effects of the proteins from artemia extract on human fibroblasts cell proliferation and collagen expression in induced aging conditions. Ann. Biotechnol. 2018, 3, 1015. [Google Scholar] [CrossRef]
  174. Macwan, C.; Patel, H.V.; Kalia, K. A comparative evaluation of in vitro antioxidant properties of bamboo Bambusa arundinacea leaves extracts. J. Cell Tissue Res. 2010, 10, 2413–2418. [Google Scholar]
  175. Arora, D.; Rani, A.; Sharma, A. A review on phytochemistry and ethnopharmacological aspects of genus Calendula. Pharmacogn. Rev. 2013, 7, 179–187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Jadoon, S.; Karim, S.; Asad, M.H.H.B.; Akram, M.R.; Kalsoom Khan, A.; Malik, A.; Chen, C.; Murtaza, G. Anti-Aging Potential of Phytoextract Loaded-Pharmaceutical Creams for Human Skin Cell Longetivity. Oxid. Med. Cell. Longev. 2015, 1–17. [Google Scholar] [CrossRef] [Green Version]
  177. Andresen, F.A. Final report on the safety assessment of Calendula officinalis extract and Calendula officinalis. Int. J. Toxicol. 2001, 20, 13–20. [Google Scholar] [CrossRef]
  178. Re, T.A.; Mooney, D.; Antignac, E.; Dufour, E.; Bark, I.; Srinivasan, V.; Nohynek, G. Application of the threshold of toxicological concern approach for the safety evaluation of calendula flower (Calendula officinalis) petals and extracts used in cosmetic and personal care products. Food Chem. Toxicol. 2009, 47, 1246–1254. [Google Scholar] [CrossRef] [PubMed]
  179. Lohani, A.; Mishra, A.K.; Verma, A. Cosmeceutical potential of geranium and calendula essential oil: Determination of antioxidant activity and in vitro sun protection factor. J. Cosmet. Dermatol. 2018, 18, 1–8. [Google Scholar] [CrossRef]
  180. Fonseca, Y.M.; Catini, C.D.; Vicentini, F.T.M.C.; Cardoso, J.C.; Cavalcanti De Albuquerque, R.L., Jr.; Vieira Fonseca, M.J. Efficacy of Marigold Extract-Loaded Formulations Against UV-induced Oxidative Stress. J. Pharm. Sci. 2011, 100, 2182–2193. [Google Scholar] [CrossRef]
  181. Akhtar, N.; Zaman, S.U.; Khan, B.A.; Amir, M.N.; Ebrahimzadeh, M.A. Calendula extract: Effects on mechanical parameters of human skin. Acta Pol. Pharm. Drug Res. 2011, 68, 693–701. [Google Scholar]
  182. Andersen, F.A.; Bergfeld, W.F.; Belsito, D.V.; Hill, R.A.; Klaassen, C.D.; Liebler, D.C.; Marks, J.G.; Shank, R.C.; Slaga, T.J.; Snyder, P.W. Final report of the cosmetic ingredient review expert panel amended safety assessment of Calendula officinalis-Derived cosmetic ingredients. Int. J. Toxicol. 2010, 29, 221S–243S. [Google Scholar] [CrossRef]
  183. Mahmood, T.; Akhtar, N. Combined Topical Application of Lotus and Green Tea Improves Facial Skin Surface Parameters. Rejuvenation Res. 2013, 16, 91–97. [Google Scholar] [CrossRef] [Green Version]
  184. Mahmood, T.; Akhtar, N.; Khan, B.A.; Khan, H.M.S.; Saeed, T. Outcomes of 3% green tea emulsion on skin sebum production in male volunteers. Bosn. J. Basic Med. Sci. 2010, 10, 260–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Koch, W.; Zagórska, J.; Marzec, Z.; Kukula-Koch, W. Applications of tea (Camellia sinensis) and its Active Constituents in Cosmetics. Molecules 2019, 24, 4277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Hsu, S. Green tea and the skin. J. Am. Acad. Dermatol. 2005, 52, 1049–1059. [Google Scholar] [CrossRef] [PubMed]
  187. Gianeti, M.D.; Mercurio, D.G.; Maia Campos, P.M.B.G. The use of green tea extract in cosmetic formulations: Not only an antioxidant active ingredient. Dermatol. Ther. 2013, 26, 267–271. [Google Scholar] [CrossRef] [PubMed]
  188. Nobrega, A.T.; Wagemaker, T.A.L.; Maia Campos, P.M.B.G. Antioxidant activity of Matricaria chamomilla L. extract and clinical efficacy of cosmetic formulations containing this extract and its isolated compounds. J. Biomed. Biopharm. Res. 2013, 10, 249–261. [Google Scholar] [CrossRef]
  189. Srivastava, J.K.; Shankar, E.; Gupta, S. Chamomile: A herbal medicine of the past with a bright future (Review). Mol. Med. Rep. 2010, 3, 895–901. [Google Scholar] [CrossRef] [PubMed]
  190. Avonto, C.; Rua, D.; Lasonkar, P.B.; Chittiboyina, A.G.; Khan, I.A. Identification of a compound isolated from German chamomile (Matricaria chamomilla) with dermal sensitization potential. Toxicol. Appl. Pharmacol. 2017, 318, 16–22. [Google Scholar] [CrossRef] [PubMed]
  191. Ratz-Łyko, A.; Arct, J.; Pytkowska, K. Moisturizing and Antiinflammatory Properties of Cosmetic Formulations Containing Centella asiatica Extract. Indian J. Pharm. Sci. 2016, 78, 27–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Bylka, W.; Znajdek-awiżeń, P.; Studzińska-sroka, E.; Brzezińska, M. Centella asiatica in cosmetology. Adv. Dermatology Allergol. 2013, 1, 46–49. [Google Scholar] [CrossRef]
  193. Lall, N.; Kishore, N.; Momtaz, S.; Hussein, A.; Naidoo, S.; Nqephe, M.; Crampton, B. Extract from Ceratonia siliqua Exhibits Depigmentation Properties. Phyther. Res. 2015, 29, 1729–1736. [Google Scholar] [CrossRef]
  194. Azab, A. CAROB (Ceratonia siliqua): Health, Medicine and Chemistry. Eur. Chem. Bull. 2017, 61, 456–469. [Google Scholar] [CrossRef] [Green Version]
  195. Krokou, A.; Stylianou, M.; Agapiou, A. Assessing the volatile profile of carob tree (Ceratonia siliqua L.). Environ. Sci. Pollut. Res. 2019. [Google Scholar] [CrossRef]
  196. Botto, J.-M.; Domloge, N.; Portolan, F. Cosmetic Use of a Carob Seed Extract as a Slimming Active Agent. European Patent No. EP2931231A2, 21 October 2015. [Google Scholar]
  197. Dosoky, N.S.; Setzer, W.N. Biological Activities and Safety of Citrus spp. Essential Oils. Int. J. Mol. Sci. 2018, 19, 1966. [Google Scholar] [CrossRef] [Green Version]
  198. Burnett, C.L.; Fiume, M.M.; Bergfeld, W.F.; Belsito, D.V.; Hill, R.A.; Klaassen, C.D.; Liebler, D.C.; Marks, J.G., Jr.; Shank, R.C.; Slaga, T.J.; et al. Safety Assessment of Citrus-Derived Peel Oils as Used in Cosmetics. Int. J. Toxicol. 2019, 38, 33S–59S. [Google Scholar] [CrossRef]
  199. Navarra, M.; Mannucci, C.; Delbò, M.; Calapai, G. Citrus bergamia essential oil: From basic research to clinical application. Front. Pharmacol. 2015, 6, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Ravichandran, C.; Badgujar, P.C.; Gundev, P.; Upadhyay, A. Review of toxicological assessment of d-limonene, a food and cosmetics additive. Food Chem. Toxicol. 2018, 120, 668–680. [Google Scholar] [CrossRef] [PubMed]
  201. Sotiroudis, G.; Melliou, E.; Sotiroudis, T.G.; Chinou, I. Chemical Analysis, Antioxidant and Antimicrobial Activity of Three Greek Cucumber (Cucumis sativus) Cultivars. J. Food Biochem. 2009, 34, 61–78. [Google Scholar] [CrossRef]
  202. Fiume, M.M.; Bergfeld, W.F.; Belsito, D.V.; Hill, R.A.; Klaassen, C.D.; Liebler, D.C.; Marks, J.; Shank, R.C.; Slaga, T.J.; Snyder, P.W.; et al. Safety Assessment of Cucumis sativus (Cucumber)-Derived Ingredients as Used in Cosmetics. Int. J. Toxicol. 2014, 33, 47S–64S. [Google Scholar] [CrossRef]
  203. Kawahara, T.; Tsutsui, K.; Nakanishi, E.; Inoue, T.; Hamauzu, Y. Effect of the topical application of an ethanol extract of quince seeds on the development of atopic dermatitis-like symptoms in NC/Nga mice. Complement. Altern. Med. 2017, 17, 80. [Google Scholar] [CrossRef] [Green Version]
  204. Muzykiewicz, A.; Zielonka-brzezicka, J.; Klimowicz, A. Quince (Cydonia oblonga Mill.) as a useful source of antioxidants–antioxidant activity evaluation. Herba Pol. 2018, 64, 23–33. [Google Scholar] [CrossRef] [Green Version]
  205. Tamri, P.; Hemmati, A.; Boroujerdnia, G.M. Wound healing properties of quince seed mucilage: In vivo evaluation in rabbit full-thickness wound model. Int. J. Surg. 2014, 12, 843–847. [Google Scholar] [CrossRef] [Green Version]
  206. Monka, A.; Grygorieva, O.; Chlebo, P.; Brindza, J. Morphological and antioxidant characteristics of quince (Cydonia oblonga Mill.) and chinese quince fruit (Pseudocydonia sinensis Schneid.). Potravinarstvo 2014, 8, 333–340. [Google Scholar] [CrossRef]
  207. Aghmiuni, A.I.; Keshel, S.H.; Sefat, F.; Khiyavi, A.A. Quince seed mucilage-based scaffold as a smart biological substrate to mimic mechanobiological behavior of skin and promote fibroblasts proliferation and h-ASCs differentiation into keratinocytes. Int. J. Biol. Macromol. 2019, 142, 668–679. [Google Scholar] [CrossRef]
  208. Ghafourian, M.; Tamri, P.; Hemmati, A.A. Enhancement of Human Skin Fibroblasts Proliferation as a Result Treating With Quince Seed Mucilage. Jundishapur J. Nat. Pharm. Prod. 2015, 10, e18820. [Google Scholar] [CrossRef] [Green Version]
  209. Li, Y.; Huang, J.; Lu, J.; Ding, Y.; Jiang, L.; Hu, S.; Chen, J. The role and mechanism of Asian medicinal plants in treating skin pigmentary disorders. J. Ethnopharmacol. 2019, 112173. [Google Scholar] [CrossRef]
  210. Xu, P.; Su, S.; Tan, C.; Lai, R.; Min, Z. Effects of aqueous extracts of Ecliptae herba, Polygoni multiflori radix praeparata and Rehmanniae radix praeparata on melanogenesis and the migration of human melanocytes. J. Ethnopharmacol. 2016, 195, 89–95. [Google Scholar] [CrossRef]
  211. Chung, I.; Rajakumar, G.; Lee, J.; Kim, S. Ethnopharmacological uses, phytochemistry, biological activities, and biotechnological applications of Eclipta prostrata. Appl. Microbiol. Biotechnol. 2017, 101, 5247–5257. [Google Scholar] [CrossRef]
  212. Chan, C.; Huang, W.; Guo, H.; Wang, B.R. Potent Antioxidative and UVB Protective Effect of Water Extract of Eclipta prostrata L. Sci. World J. 2014, 1–8. [Google Scholar] [CrossRef] [Green Version]
  213. Jahan, R.; Al-nahain, A.; Majumder, S.; Rahmatullah, M. Ethnopharmacological Significance of Eclipta alba (L.) Hassk. (Asteraceae). Int. Sch. Res. Not. 2014, 1–22. [Google Scholar] [CrossRef] [Green Version]
  214. Liu, Y.; Hwang, E.; Ngo, H.T.T.; Perumalsamy, H.; Kim, Y.J.; Li, L. Protective Effects of Euphrasia officinalis Extract against Ultraviolet B-Induced Photoaging in Normal Human Dermal Fibroblasts. Int. J. Mol. Sci. 2018, 19, 3327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Petrichenko, V.M.; Sukhinina, T.V.; Babiyan, L.K.; Shramm, N.I. Chemical composition and antioxidant properties of biologically active compounds from Euphrasia brevipila. Pharm. Chem. J. 2006, 40, 312–316. [Google Scholar] [CrossRef]
  216. Bigagli, E.; Cinci, L.; D’Ambrosio, M.; Luceri, C. Pharmacological activities of an eye drop containing Matricaria chamomilla and Euphrasia officinalis extracts in UVB-induced oxidative stress and inflammation of human corneal cells. J. Photochem. Photobiol. B Biol. 2017, 173, 618–625. [Google Scholar] [CrossRef] [PubMed]
  217. Laekeman, G.; Houdart, M.; Vervisch, P. EMA Assessment Report on Euphrasia officinalis L. and Euphrasia rostkoviana Hayne, Herba. Available online: https://www.ema.europa.eu/en/documents/herbal-report/final-assessment-report-euphrasia-officinalis-l-euphrasia-rostkoviana-hayne-herba_en.pdf (accessed on 8 July 2020).
  218. Badgujar, S.B.; Patel, V.V.; Bandivdekar, A.H.; Mahajan, R.T. Traditional uses, phytochemistry and pharmacology of Ficus carica: A review. Pharm. Biol. 2014, 52, 1487–1503. [Google Scholar] [CrossRef] [Green Version]
  219. Khan, H.; Akhtar, N.; Ali, A. Effects of Cream Containing Ficus carica L. Fruit Extract on Skin Parameters: In vivo Evaluation. Indian J. Pharm. Sci. 2014, 76, 560–564. [Google Scholar]
  220. Abbasi, S.; Kamalinejad, M.; Babaie, D.; Shams, S.M.; Sadr, Z.; Gheysarif, M.; Askarig, V.R.; Rakhshandeh, H. Complementary Therapies in Medicine A new topical treatment of atopic dermatitis in pediatric patients based on Ficus carica L. (Fig): A randomized, placebo-controlled clinical trial. Complement. Ther. Med. 2017, 35, 85–91. [Google Scholar] [CrossRef]
  221. Azadbakht, M.; Monadi, T.; Esmaeili, Z.; Chabra, A.; Tavakoli, N. Formulation and evaluation of licorice shampoo in comparison with commercial shampoo. J. Pharm. Bioallied Sci. 2018, 10, 208–215. [Google Scholar] [CrossRef] [PubMed]
  222. Pastorino, G.; Cornara, L.; Rodrigues, F.; Oliveira, M.B.P.P. Liquorice (Glycyrrhiza glabra): A phytochemical and pharmacological review. Phyther. Res. 2018, 32, 2323–2339. [Google Scholar] [CrossRef] [PubMed]
  223. Schoelermann, A.M.; Weber, T.M.; Arrowitz, C.; Rizer, R.L.; Qian, K.; Babcock, M. Skin compatibility and ef fi cacy of a cosmetic skin care regimen with licochalcone A and 4-t-butylcyclohexanol in patients with rosacea subtype I. J. Eur. Acad. Dermatol. Venereol. 2016, 30, 21–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Castangia, C.; Caddeo, M.; Manca, L.; Casu, L.; Latorre, A.C.; Díez-Sales, O.; Ruiz-Saurí, A.; Bacchetta, G.; Fadda, A.M.; Manconi, M. Delivery of liquorice extract by liposomes and hyalurosomes to protect the skin against oxidative stress injuries. Carbohydr. Polym. 2015, 134, 663. [Google Scholar] [CrossRef]
  225. Waqas, M.K.; Akhtar, N.; Mustafa, R.; Jamshaid, M.; Khan, H.M.S.; Murtaza, G. Review Dermatological and Cosmeceutical Benefits of Glycine Max (Soybean) and its Active Components. Acta Pol. Pharm. Drug Res. 2015, 72, 3–11. [Google Scholar]
  226. Lai, J.; Xin, C.; Zhao, Y.; Feng, B.; He, C.; Dong, Y.; Fang, Y.; Wei, S. Study of Active Ingredients in Black Soybean Sprouts and Their Safety in Cosmetic Use. Molecules 2012, 17, 11669–11679. [Google Scholar] [CrossRef]
  227. Bhattacharyya, T.K.; Bueller, H.; Hsia, Y.; Thomas, J.R. Dermal Histology in Mouse Skin Exposed to Cosmeceuticals. Facial Plast. Surg. 2017, 33, 545–550. [Google Scholar] [CrossRef]
  228. Jhan, J.; Chung, Y.; Chen, G.; Chang, C.; Lu, Y.; Hsu, C. Anthocyanin contents in the seed coat of black soya bean and their anti-human tyrosinase activity and antioxidative activity. Int. J. Cosmet. Sci. 2016, 38, 319–324. [Google Scholar] [CrossRef]
  229. Bazin, R.; Flament, F.; Colonna, A.; Harzic, L.; Bückle, R.; Piot, B.; Laize, F.; Kaaty, M.; König, K.; Fluhr, J.W. Clinical study on the effects of a cosmetic product on dermal extracellular matrix components using a high-resolution multiphoton tomograph. Skin Res. Technol. 2010, 16, 305–310. [Google Scholar] [CrossRef] [PubMed]
  230. Wallo, W.; Nebus, J.; Leyden, J.J. Efficacy of a soy moisturizer in photoaging: A double-blind, vehicle-controlled, 12-week study. J. Drugs Dermatol. 2007, 6, 917–922. [Google Scholar]
  231. Choi, S.; Jung, T.-D.; Cho, B.-Y.; Choi, S.-H.; Sim, W.-S.; Han, X.; Lee, S.J.; Kim, Y.-C.; Lee, O.-H. Anti-photoaging effect of fermented agricultural by-products on ultraviolet B-irradiated hairless mouse skin. Int. J. Mol. Med. 2019, 44, 559–568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Hooker, E. Final Report of the Amended Safety Assessment of PEG-5, -10, -16, -25, -30, and -40 Soy Sterol. Int. J. Toxicol. 2004, 23, 23–47. [Google Scholar] [CrossRef]
  233. Iijima, S.; Ito, M.; Makabe, K.; Murakami, Y.; Yokooji, T.; Matsuo, H. Case of anaphylactic reaction to soy following percutaneous sensitization by soy-based ingredients in cosmetic products. J. Dermatol. 2015, 42, 917–918. [Google Scholar] [CrossRef]
  234. Lutsenko, Y.; Bylka, W.; Matławska, I.; Darmohray, R. Hedera helix as a medicinal plant. Herba Pol. 2010, 56, 4–10. [Google Scholar]
  235. Facino, R.M.; Carini, M.; Stefani, R.; Aldini, G.; Saibene, L. Anti-Elastase and Anti-Hyaluronidase Activities of Saponins and Ruscus aculeatus: Factors Contributing to their Efficacy in the Sapogenins from Hedera helix, Aesculus hippocastanurn, and Treatment of Venous Insufficiency. Arch. Pharm. 1995, 328, 720–724. [Google Scholar] [CrossRef] [PubMed]
  236. Eberlin, S.; del Carmen Velazquez Pereda, M.; de Campos Dieamant, G.; Nogueira, C.; Werka, R.M.; de Souza, M.L. Effects of a Brazilian herbal compound as a cosmetic eyecare for periorbital hyperchromia (“dark circles”). J. Cosmet. Dermatol. 2009, 8, 127–135. [Google Scholar] [CrossRef]
  237. Mucaji, P.; Haladová, M.; Eisenreichová, E.; Sersen, F.; Ubik, K.; Granca, D. Constituents of Lilium candidum L. and their antioxidative activity. Ces. Slov. Farm. 2007, 56, 27–29. [Google Scholar]
  238. Golz-Berner, K.; Zastrow, L. Cosmetic Cleansing and Skin Care Preparation Containing Plant and Algae Extracts. U.S. Patent No. 6,221,372, 24 April 2001. [Google Scholar]
  239. Kanlayavattanakul, M.; Lourith, N. An update on cutaneous aging treatment using herbs: An update on cutaneous aging treatment using herbs. J. Cosmet. Laser Ther. 2015, 17, 343–352. [Google Scholar] [CrossRef]
  240. Active Concepts LLC. Safety Statement SilDerm® Conditioning (Cyclopentasiloxane & Dimethicone/Silsesquioxane Copolymer & Silk & Malva sylvestris (Mallow) Extract & Lilium candidum Bulb Extract & Lactobacillus/Eriodictyon Californicum Ferment Extract & Cymbidium grandiflorum F. Available online: https://activeconceptsllc.com/wp-content/uploads/2015/12/30341-SilDerm-Conditioning-Safety-Statement-v1.pdf (accessed on 20 March 2021).
  241. Bajpai, V.K.; Rahman, A.; Dung, N.T.; Huh, M.K.; Kang, S.C. In vitro Inhibition of Food Spoilage and Foodborne Pathogenic Bacteria by Essential Oil and Leaf Extracts of Magnolia liliflora Desr. J. Food Sci. 2008, 73, 314–320. [Google Scholar] [CrossRef]
  242. Bajpai, V.K.; Yoon, J.I.; Kang, S.C. Antioxidant and antidermatophytic activities of essential oil and extracts of Magnolia liliflora Desr. Food Chem. Toxicol. 2009, 47, 2606–2612. [Google Scholar] [CrossRef]
  243. Park, C.; Park, S.-Y.; Lee, S.; Kim, J.; Park, S. Analysis of Metabolites in White Flowers of Magnolia denudata Desr. and Violet Flowers of Magnolia liliiflora Desr. Molecules 2018, 23, 1558. [Google Scholar] [CrossRef] [Green Version]
  244. Martins, R.M.; de Alves Dias Assis, G.; De Siqueira Martins, S.; de Freitas, A.P.L.; Rochette, P.J.; Moulin, V.J.; Fonseca, M.J.V. Apple extract (Malus sp.) and rutin as photochemopreventive agents: Evaluation of UVB-induced alterations on skin biopsies and tissue-engineered skin. Rejuvenation Res. 2020, 23, 465–475. [Google Scholar] [CrossRef]
  245. Nešić, I.; Stojiljković, D.; Savić, S.; Tasić-Kostov, M.; Tadić, V. Stability, antioxidant activity, in vivo safety and efficacy of creams with standardized wild apple fruit extract: A comparison of conventional and biodegradable emulsifiers. Int. J. Cosmet. Sci. 2019, 41, 300–310. [Google Scholar] [CrossRef]
  246. Baldisserotto, A.; Malisardi, G.; Scalambra, E.; Andreotti, E.; Romagnoli, C.; Vicentini, C.B.; Manfredini, S.; Vertuani, S. Synthesis, Antioxidant and Antimicrobial Activity of a New Phloridzin Derivative for Dermo-Cosmetic Applications. Molecules 2012, 17, 13275–13289. [Google Scholar] [CrossRef] [Green Version]
  247. Moruś, M.; Baran, M.; Rost-Roszkowska, M.; Skotnicka-Graca, U. Plant Stem Cells as Innovation in Cosmetics. Acta Pol. Pharm. Drug Res. 2014, 71, 701–707. [Google Scholar]
  248. Shin, S.; Kum, H.; Ryu, D.; Kim, M.; Jung, E.; Park, D. Protective Effects of a New Phloretin Derivative against UVB-Induced Damage in Skin Cell Model and Human Volunteers. Int. J. Mol. Sci. 2014, 15, 18919–18940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  249. Sampaio, G.G.; Leódido, G.; Machado Gonçalves, L.; Paschoa Benini, M.A. In vitro antimicrobial potential of infant mouthwashes against streptococcus mutans biofilm: A preliminary study. Indian J. Dent. Res. 2019, 30, 399–402. [Google Scholar] [CrossRef]
  250. Medellín-Luna, M.F. Castañeda-Delgado, J.E.; Martínez-Balderas, V.Y. Cervantes-Villagrana, A.R. Medicinal Plant Extracts and Their Use as Wound Closure Inducing Agents. J. Med. Food 2019, 22, 1–9. [Google Scholar] [CrossRef] [PubMed]
  251. Braga, A.S.; Pires, J.G.; Magalhães, A.C. Effect of a mouthrinse containing Malva sylvestris on the viability and activity of microcosm biofilm and on enamel demineralization compared to known antimicrobials mouthrinses. Biofouling 2018, 34, 252–261. [Google Scholar] [CrossRef] [PubMed]
  252. Afshar, M.; Ravarian, B.; Zardast, M.; Adel, S.; Fard, M.H.; Valavi, M. Evaluation of cutaneous wound healing activity of Malva sylvestris aqueous extract in BALB/c mice. Iran. J. Basic Med. Sci. 2021, 18, 616–622. [Google Scholar]
  253. Nasiri, E.; Hosseinimehr, S.J.; Azadbakht, M.; Akbari, J.; Enayati-fard, R.; Azizi, S. Effect of Malva sylvestris cream on burn injury and wounds in rats. Avicenna J. Phytomed. 2021, 5, 341–354. [Google Scholar]
  254. Barros, L.; Carvalho, A.M.; Ferreira, I.C.F.R. Leaves, flowers, immature fruits and leafy flowered stems of Malva sylvestris: A comparative study of the nutraceutical potential and composition. Food Chem. Toxicol. 2010, 48, 1466–1472. [Google Scholar] [CrossRef]
  255. Pirbalouti, G.A.; Koohpyeh, A. Wound Healing Activity of Extracts of Malva sylvestris and Stachys lavandulifolia. Int. J. Biol. 2011, 3, 174–179. [Google Scholar] [CrossRef] [Green Version]
  256. Prudente, A.S.; Sponchiado, G.; Mendes, D.A.G.B.; Soley, B.S.; Cabrini, D.A.; Otuki, M.F. Pre-clinical efficacy assessment of Malva sylvestris on chronic skin inflammation. Biomed. Pharmacother. 2017, 93, 852–860. [Google Scholar] [CrossRef] [PubMed]
  257. Cudalbeanu, M.; Ghinea, I.O.; Furdui, B.; Dah-nouvlessounon, D.; Raclea, R.; Costache, T.; Cucolea, I.E.; Urlan, F.; Dinica, R.M. Exploring New Antioxidant and Mineral Compounds from Nymphaea alba Wild-Grown in Danube Delta Biosphere. Molecules 2018, 23, 1247. [Google Scholar] [CrossRef] [Green Version]
  258. Zhao, Y.; Fan, Y.-Y.; Yu, W.-G.; Wang, J.; Lu, W.; Song, X.-Q. Ultrasound-Enhanced Subcritical Fluid Extraction of Essential Oil from Nymphaea alba var and Its Antioxidant Activity. J. AOAC Int. 2019, 102, 1448–1454. [Google Scholar] [CrossRef]
  259. Bakr, R.O.; El-naa, M.M.; Zaghloul, S.S.; Omar, M.M. Profile of bioactive compounds in Nymphaea alba L. leaves growing in Egypt: Hepatoprotective, antioxidant and anti-inflammatory activity. BMC Complement. Altern. Med. 2017, 17, 52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  260. Laughlin, T.; Tan, Y.; Jarrold, B.; Chen, J.; Li, L.; Fang, B.; Zhao, W.; Tamura, M.; Matsubara, A.; Deng, G.; et al. Autophagy activators stimulate the removal of advanced glycation end products in human keratinocytes. J. Eur. Acad. Dermatol. Venereol. 2020, 34, 12–18. [Google Scholar] [CrossRef]
  261. Monrroy, M.; García, E.; Ríos, K.; García, J.R. Extraction and Physicochemical Characterization of Mucilage from Opuntia cochenillifera (L.) Miller. J. Chem. 2017, 1–9. [Google Scholar] [CrossRef] [Green Version]
  262. Da Cruz Filho, I.J.; da Silva Barros, B.R.; de Souza Aguiar, L.M.; Navarro, C.D.C.; Ruas, J.S.; de Lorena, V.M.B.; de Moares Rocha, G.J.; Verecesi, A.E.; Moutinho Lagos de Melo, C.; Souto Maior, A.M. Lignins isolated from Prickly pear cladodes of the species Opuntia fícus-indica (Linnaeus) Miller and Opuntia cochenillifera (Linnaeus) Miller induces mice splenocytes activation, proliferation and cytokines production. Int. J. Biol. Macromol. 2019, 123, 1331–1339. [Google Scholar] [CrossRef]
  263. Stintzing, F.C.; Carle, R. Review Cactus stems (Opuntia spp.): A review on their chemistry, technology, and uses. Mol. Nutr. Food Res. 2005, 49, 175–194. [Google Scholar] [CrossRef]
  264. Aruwa, E.C.; Amoo, S.O.; Kudanga, T. Opuntia (Cactaceae) plant compounds, biological activities and prospects—A comprehensive review. Food Res. Int. 2018, 112, 328–344. [Google Scholar] [CrossRef] [PubMed]
  265. Kanlayavattanakul, M.; Lourith, N. Orchid Extracts and Cosmetic Benefits. In Orchids Phytochemistry, Biology and Horticulture; Mérillon, J.-M., Kodja, H., Eds.; Springer International Publishing: Cham, Switzerland, 2020; pp. 1–18. ISBN 9783030112578. [Google Scholar]
  266. Bose, B.; Choudhury, H.; Tandon, P.; Kumaria, S. Studies on secondary metabolite profiling, anti-inflammatory potential, in vitro photoprotective and skin-aging related enzyme inhibitory activities of Malaxis acuminata, a threatened orchid of nutraceutical importance. J. Photochem. Photobiol. B Biol. 2017, 173, 686–695. [Google Scholar] [CrossRef]
  267. Zhu, Y.; Pan, W.; Ku, C.F.; Zhang, H.; Tsang, S.W. Design, synthesis and evaluation of novel dihydrostilbene derivatives as potential anti-melanogenic skin-protecting agents. Eur. J. Med. Chem. 2018, 143, 1254–1260. [Google Scholar] [CrossRef]
  268. Hadi, H.; Razali, S.N.S.; Awadh, A.I. A Comprehensive Review of the Cosmeceutical Benefits of Vanda Species (Orchidaceae). Nat. Prod. Commun. 2015, 10, 1483–1488. [Google Scholar] [CrossRef] [Green Version]
  269. Tadokoro, T.; Bonte, F.; Archambault, J.C.; Cauchard, J.H.; Neveu, M.; Ozawa, K.; Noguchi, F.; Ikeda, A.; Nagamatsu, M.; Shinn, S. Whitening efficacy of plant extracts including orchid extracts on Japanese female skin with melasma and lentigo senilis. J. Dermatol. 2010, 37, 522–530. [Google Scholar] [CrossRef] [PubMed]
  270. MacAulay, J.C. Orchid allergy. Contact Dermat. 1987, 17, 112–113. [Google Scholar] [CrossRef] [PubMed]
  271. Mazzanti, G.; Braghiroli, L. Analgesic Antiinflammatory Action of Pfaffia paniculata (Martius) Kuntze. Phyther. Res. 1994, 8, 413–416. [Google Scholar] [CrossRef]
  272. Angelis, A.; Hubert, J.; Aligiannis, N.; Michalea, R.; Abedini, A.; Nuzillard, J.-M.; Gangloff, S.C.; Skaltsounis, A.-L.; Renault, J.-H. Bio-Guided Isolation of Methanol-Soluble by-Products and Investigation of Their Dermo-Cosmetic Properties. Molecules 2016, 21, 1586. [Google Scholar] [CrossRef] [Green Version]
  273. Hubert, J.; Angelis, A.; Aligiannis, N.; Rosalia, M.; Abedini, A.; Bakiri, A.; Reynaud, R.; Nuzillard, J.-M.; Gangloff, S.C.; Skaltsounis, A.-L.; et al. In Vitro Dermo-Cosmetic Evaluation of Bark Extracts from Common Temperate Trees. Planta Med. 2016, 82, 1351–1358. [Google Scholar] [CrossRef] [Green Version]
  274. Burčová, Z.; Kreps, F.; Greifová, M.; Jablonský, M.; Ház, A.; Schmidt, Š.; Šurina, I. Antibacterial and antifungal activity of phytosterols and methyl dehydroabietate of Norway spruce bark extracts. J. Biotechnol. 2018, 282, 18–24. [Google Scholar] [CrossRef]
  275. Sipponen, A.; Peltola, R.; Jokinen, J.J.; Laitinen, K.; Lohi, J.; Rautio, M.; Sipponen, P.; Lounatmaa, K. Effects of Norway Spruce (Picea abies) Resin on Cell Wall and Cell Membrane of Staphylococcus aureus. Ultrastruct. Pathol. 2009, 33, 128–135. [Google Scholar] [CrossRef]
  276. Jokinen, J.J.; Sipponen, A. Refined Spruce Resin to Treat Chronic Wounds: Rebirth of an Old Folkloristic Therapy. Adv. Wound Care 2016, 5, 198–207. [Google Scholar] [CrossRef] [Green Version]
  277. Marcati, A.; Ursu, V.A.; Laroche, C.; Soanen, N.; Marchal, L.; Jubeau, S.; Djelveh, G.; Michaud, P. Extraction and fractionation of polysaccharides and B-phycoerythrin from the microalga Porphyridium cruentum by membrane technology. Algal Res. 2014, 5, 258–263. [Google Scholar] [CrossRef]
  278. Arad, M.; Yaron, A. Natural pigments from red microalgae for use in foods and cosmetics. Trends Food Sci. Technol. 1992, 3, 92–97. [Google Scholar] [CrossRef]
  279. Servel, M.-O.; Claire, C.; Derrien, A.; Coiffard, L.; De Roeck-Holtzhauer, Y. Fatty acid composition of some Marine Microalge. Phytochemistry 1994, 36, 691–693. [Google Scholar] [CrossRef]
  280. Huang, J.J.; Xu, W.; Lin, S.; Cheung, P.C.-K. Phytochemical profiles of marine phytoplanktons: An evaluation of their in vitro antioxidant and anti-proliferative activities. Food Funct. 2016, 7, 5002–5017. [Google Scholar] [CrossRef]
  281. De Jesus Raposo, F.M.; de Morais, M.A.B.; de Morais, R.M.S.C. Marine Polysaccharides from Algae with Potential Biomedical Applications. Mar. Drugs 2015, 13, 2967–3028. [Google Scholar] [CrossRef] [PubMed]
  282. Mourelle, M.L.; Gómez, C.P.; Legido, J.L. The Potential Use of Marine Microalgae and Cyanobacteria in Cosmetics and Thalassotherapy. Cosmetics 2017, 4, 46. [Google Scholar] [CrossRef] [Green Version]
  283. Baby, A.R.; Maciel, C.P.M.; Kaneko, T.M.; Velasco, M.V.R. UV Spectrophotometric Determination of Bioflavonoids from a Semisolid Pharmaceutical Dosage Form Containing Trichilia catigua Adr. Juss and Ptychopetalum olacoides Bentham Standardized Extract: Analytical Method Validation and Statistical Procedures. J. AOAC Int. 2006, 89, 1532–1537. [Google Scholar] [CrossRef] [Green Version]
  284. Bogdan, C.; Iurian, S.; Tomuta, I.; Moldovan, M. Improvement of skin condition in striae distensae: Development, characterization and clinical efficacy of a cosmetic product containing Punica granatum seed oil and Croton lechleri resin extract. Drug Des. Devel. Ther. 2017, 11, 521–531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  285. Fleck, A.; Cabral, P.F.G.; Vieira, F.F.M.; Pinheiro, D.A.; Pereira, C.R.; Santos, W.C.; Machado, T.B. Punica granatum L. Hydrogel for Wound Care Treatment: From Case Study to Phytomedicine Standardization. Molecules 2016, 21, 1059. [Google Scholar] [CrossRef] [Green Version]
  286. Prasad, D.; Kunnaiah, R. Punica granatum: A review on its potential role in treating periodontal disease. J. Indian Soc. Periodontol. 2014, 18, 428–432. [Google Scholar] [CrossRef]
  287. Javanmard, M.; Asadi-Gharneh, H.A.; Nikneshan, P. Characterization of biochemical traits of dog rose (Rosa canina L.) ecotypes in the central part of Iran. Nat. Prod. Res. 2018, 32, 1738–1743. [Google Scholar] [CrossRef] [PubMed]
  288. Ochando-Ibernón, G.; Schneller-Pavelescu, L.; Silvestre-Salvador, J.F. Allergic contact dermatitis caused by “Rosa mosqueta” oil. Contact Dermat. 2018, 79, 259–260. [Google Scholar] [CrossRef] [PubMed]
  289. Hwang, D.H.; Lee, D.Y.; Koh, P.O.; Yang, H.R.; Kang, C.; Kim, E. Rosa davurica pall. Improves Propionibacterium acnes-induced inflammatory responses in mouse ear edema model and suppresses pro-inflammatory chemokine production via MAPK and NF-κB pathways in HaCaT cells. Int. J. Mol. Sci. 2020, 21, 1717. [Google Scholar] [CrossRef] [Green Version]
  290. Olech, M.; Pietrzak, W.; Nowak, R. Characterization of Free and Bound Phenolic Acids and Flavonoid Aglycones in Rosa rugosa Thunb. Leaves and Achenes using LC-ESI-MS/MS-MRM Methods. Molecules 2020, 25, 1804. [Google Scholar] [CrossRef] [Green Version]
  291. Kılıç, S.; Okullu, S.Ö.; Kurt, Ö.; Sevinç, H.; Dündar, C.; Altınordu, F.; Türkoğlu, M. Efficacy of two plant extracts against acne vulgaris: Initial results of microbiological tests and cell culture studies. J. Cosmet. Dermatol. 2018, 10, 1061–1065. [Google Scholar] [CrossRef]
  292. Boskabady, M.H.; Shafei, M.N.; Saberi, Z.; Amini, S. Pharmacological effects of Rosa Damascena. Iran. J. Basic Med. Sci. 2011, 14, 295–307. [Google Scholar] [CrossRef]
  293. Basim, E.; Basim, H. Antibacterial activity of Rosa damascena essential oil. Fitoterapia 2003, 74, 394–396. [Google Scholar] [CrossRef]
  294. Baydar, N.G.; Baydar, H. Phenolic compounds, antiradical activity and antioxidant capacity of oil-bearing rose (Rosa damascena Mill.) extracts. Ind. Crops Prod. 2013, 41, 375–380. [Google Scholar] [CrossRef]
  295. Martínez, M.C.; Santiago, J.L.; Boso, S.; Gago, P.; Álvarez-Acero, I.; De Vega, M.E.; Martínez-Bartolomé, M.; Álvarez-Nogal, R.; Molíst, P.; Caser, M.; et al. Narcea—An unknown, ancient cultivated rose variety from northern Spain. Hortic. Res. 2020, 7, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  296. Palshetkar, A.; Pathare, N.; Jadhav, N.; Pawar, M.; Wadhwani, A.; Kulkarni, S.; Singh, K.K. In vitro anti-HIV activity of some Indian medicinal plant extracts. BMC Complement. Med. Ther. 2020, 20, 69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  297. De Macedo, L.M.; Dos Santos, É.M.; Militão, L.; Tundisi, L.L.; Ataide, J.A.; Souto, E.B.; Mazzola, P.G. Rosemary (Rosmarinus officinalis L., syn Salvia rosmarinus Spenn.) and Its Topical Applications: A review. Plants 2020, 9, 651. [Google Scholar] [CrossRef]
  298. Nobile, V.; Michelotti, A.; Cestone, E.; Caturla, N.; Castillo, J.; Benavente-García, O.; Pérez-Sánchez, A.; Micol, V. Skin photoprotective and antiageing effects of a combination of rosemary (Rosmarinus officinalis) and grapefruit (Citrus paradisi) polyphenols. Food Nutr. Res. 2016, 60, 31871. [Google Scholar] [CrossRef] [Green Version]
  299. Miroddi, M.; Calapai, G.; Isola, S.; Minciullo, P.L.; Gangemi, S. Rosmarinus officinalis L. as cause of contact dermatitis. Allergol. Immunopathol. 2014, 42, 616–619. [Google Scholar] [CrossRef]
  300. Puupponen-Pimiä, R.; Nohynek, L.; Alakomi, H.-L.; Oksman-Caldentey, K.-M. Bioactive berry compounds—Novel tools against human pathogens. Appl. Microbiol. Biotechnol. 2004, 67, 8–18. [Google Scholar] [CrossRef] [PubMed]
  301. Hummer, K.E. Rubus Pharmacology: Antiquity to the Present. Hortic. Sci. 2010, 45, 1587–1591. [Google Scholar] [CrossRef] [Green Version]
  302. Final Report Plant-Derived Fatty Acid Oils as Used in Cosmetics. Available online: https://purelyprofessional.dk/wp-content/uploads/inci/persea-gratissima-oil.pdf (accessed on 15 May 2021).
  303. Singh, A.; Lal, U.R.; Mukhtar, H.M.; Singh, P.S.; Shah, G.; Dhawan, R.K. Phytochemical profile of sugarcane and its potential health aspects. Pharmacogn. Rev. 2015, 9, 45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  304. Alves, P.E.; Gomes, A.C.C.; Gomes, A.K.C.; Nigro, F.; Kuster, R.M.; de Freitas, Z.M.F.; Coutinho, C.S.C.; de S.B. Monteiro, M.S.; Pereira dos Santos, E.; Simas, N.K. Development and Characterization of Phytocosmetic Formulations with Saccharum officinarum. Rev. Bras. Farmacogn. 2020, 30, 406–415. [Google Scholar] [CrossRef]
  305. Ali, S.E.; El Gedaily, R.A.; Mocan, A.; Farag, M.A.; El-seedi, H.R. Sugarcane (Saccharum officinarum Linn.) Juice and Its Product Molasses via a Multiplex Metabolomics Approach. Molecules 2019, 24, 934. [Google Scholar] [CrossRef] [Green Version]
  306. Tundis, R.; Ursino, C.; Bonesi, M.; Loizzo, M.R.; Sicari, V.; Pellican, T.; Manfredi, I.L.; Figoli, A.; Cassano, A. Flower and Leaf Extracts of Sambucus nigra L.: Application of Membrane Processes to Obtain Fractions with Antioxidant and Antityrosinase Properties. Membranes 2019, 9, 127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  307. Jarzycka, A.; Lewin, A.; Gancarz, R.; Wilk, K.A. Assessment of extracts of Helichrysum arenarium, Crataegus monogyna, Sambucus nigra in photoprotective UVA and UVB; photostability in cosmetic emulsions q. J. Photochem. Photobiol. B Biol. 2013, 128, 50–57. [Google Scholar] [CrossRef] [PubMed]
  308. Jarić, S.; Kostić, O.; Mataruga, Z.; Pavlović, D.; Pavlović, M.; Pavlović, P. Traditional wound-healing plants used in the Balkan region (Southeast Europe). J. Ethnopharmacol. 2017, 211, 311–328. [Google Scholar] [CrossRef] [PubMed]
  309. Örs, G.; İz Gülçe, S. Cytoprotective effect of a functional antipollutant blend through reducing B [a] P-induced intracellular oxidative stress and UVA exposure. Turk. J. Biol. 2018, 42, 453–462. [Google Scholar] [CrossRef]
  310. Lin, P.; Hwang, E.; Ngo, H.T.T.; Seo, S.A.; Yi, T.-H. Sambucus nigra L. ameliorates UVB-induced photoaging and inflammatory response in human skin keratinocytes. Cytotechnology 2019, 71, 1003–1017. [Google Scholar] [CrossRef]
  311. Mogoşanu, G.D.; Popescu, F.C.; Busuioc, C.J.; Pop, O.T.; Mogoantă, L.; Pârvănescu, H.; Rău, G.; Lascăr, I. Effects of a Topical Preparation Containing Sambuci Folium Extract in Experimental Model of Thermal Skin Burns on Rats. Farmacia 2014, 62, 693–703. [Google Scholar]
  312. Crisan, M.; David, L.; Moldovan, B.; Vulcu, A.; Dreve, S.; Perde-schrepler, M.; Tatomir, C.; Filip, G.; Bolfa, P. New nanomaterials for the improvement of psoriatic lesions. J. Mater. Chem. B 2013, 1, 3152. [Google Scholar] [CrossRef] [PubMed]
  313. Lall, N.; Chrysargyris, A.; Lambrechts, I.; Fibrich, B.; Van Staden, A.B.; Twilley, D.; de Canha, M.N.; Oosthuizen, C.B.; Bodiba, D.; Tzortzakis, N. Sideritis perfoliata (Subsp. Perfoliata) Nutritive Value and Its Potential Medicinal Properties. Antioxidants 2019, 8, 521. [Google Scholar] [CrossRef] [Green Version]
  314. Charami, M.-T.; Lazari, D.; Karioti, A.; Skaltsa, H.; Hadjipavlou-Litina, D. Souleles, C. Antioxidant and Antiinflammatory Activities of Sideritis perfoliata subsp. perfoliata (Lamiaceae). Phyther. Res. 2008, 22, 450–454. [Google Scholar] [CrossRef]
  315. Lytra, K.; Tomou, E.; Chrysargyris, A.; Drouza, C.; Skaltsa, H.; Tzortzakis, N. Traditionally Used Sideritis cypria Post.: Phytochemistry, Nutritional Content, Bioactive Compounds of Cultivated Populations. Front. Pharmacol. 2020, 11, 650. [Google Scholar] [CrossRef] [PubMed]
  316. Kirkan, B.; Locatelli, M.; Mocan, A.; Zengin, G.; Sarikurucu, C. Phenolic profile and bioactivities of Sideritis perfoliata L.: From the plant to its most active extract and its broad biological properties. Front. Pharmacol. 2020, 10, 1642. [Google Scholar] [CrossRef] [Green Version]
  317. Romanucci, V.; Di Fabio, G.; D’Alonzo, D.; Guaragna, A.; Scapagninib, G.; Zarrelli, A. Traditional uses, chemical composition and biological activities of Sideritis raeseri Boiss. & Heldr. J. Sci. Food Agric. 2016, 97, 373–383. [Google Scholar] [CrossRef]
  318. He, X.; Bai, Y.; Zhao, Z.; Wang, X.; Fang, J.; Huang, L.; Zeng, M.; Zhang, Q.; Zhang, Y.; Zheng, X. Local and traditional uses, phytochemistry, and pharmacology of Sophora japonica L.: A review. J. Ethnopharmacol. 2016, 187, 160–182. [Google Scholar] [CrossRef]
  319. Li, L.; Huang, T.; Lan, C.; Ding, H.; Yan, C.; Dou, Y. Protective effect of polysaccharide from Sophora japonica L. flower buds against UVB radiation in a human keratinocyte cell line (HaCaT cells). J. Photochem. Photobiol. B Biol. 2019, 191, 135–142. [Google Scholar] [CrossRef]
  320. Lo, Y.-H.; Lin, R.-D.; Lin, Y.-P.; Liu, Y.-L.; Lee, M.-H. Active constituents from Sophora japonica exhibiting cellular tyrosinase inhibition in human epidermal melanocytes. J. Ethnopharmacol. 2009, 124, 625–629. [Google Scholar] [CrossRef] [PubMed]
  321. Wang, K.-H.; Lin, R.-D.; Hsu, F.-L.; Huang, Y.-H.; Chang, H.-C.; Huang, C.-Y.; Lee, M.-H. Cosmetic applications of selected traditional Chinese herbal medicines. J. Ethnopharmacol. 2006, 106, 353–359. [Google Scholar] [CrossRef]
  322. Sanguigno, L.; Minale, M.; Vannini, E.; Arato, G.; Riccio, R.; Casapullo, A.; Monti, M.C.; Riccio, R.; Formisano, S.; Di Rezo, G.; et al. Oligosaccharidic fractions derived from Triticum vulgare extract accelerate tissutal repairing processes in in vitro and in vivo models of skin lesions. J. Ethnopharmacol. 2015, 159, 198–208. [Google Scholar] [CrossRef] [Green Version]
  323. Tito, A.; Minale, M.; Riccio, S.; Grieco, F.; Colucci, M.G.; Apone, F. A Triticum vulgare Extract Exhibits Regenerating Activity During the Wound Healing Process. Clin. Cosmet. Investig. Dermatol. 2020, 13, 21–30. [Google Scholar] [CrossRef] [Green Version]
  324. D’Agostino, A.D.; Pirozzi, A.V.A.; Finamore, R.; Grieco, F.; Minale, M.; Schiraldi, C. Molecular Mechanisms at the Basis of Pharmaceutical Grade Triticum vulgare Extract Efficacy in Prompting Keratinocytes Healing. Molecules 2020, 25, 431. [Google Scholar] [CrossRef] [Green Version]
  325. Martini, P.; Mazzatenta, C.; Saponati, G. Efficacy and Tolerability of Fitostimoline in Two Different Forms (Soaked Gauzes and Cream) and Citrizan Gel in the Topical Treatment of Second-Degree Superficial Cutaneous Burns. Dermatol. Res. Pract. 2011, 1–8. [Google Scholar] [CrossRef] [Green Version]
  326. Burnett, C.; Bergfeld, W.F.; Belsito, D.V.; Hill, R.A.; Klaassen, C.D.; Liebler, D.C.; Marks, J.G.; Shank, R.C.; Slaga, T.J.; Snyder, P.W.; et al. Safety Assessment of Hydrolyzed Wheat Protein and Hydrolyzed Wheat Gluten as Used in Cosmetics. Int. J. Toxicol. 2018, 37, 55S–66S. [Google Scholar] [CrossRef] [PubMed]
  327. Eom, S.Y.; Chung, C.B.; Kim, Y.S.; Kim, J.H.; Kim, K.S.; Kim, Y.H.; Park, S.H.; Hwang, Y.; Kim, K.H. Cosmeceutical properties of polysaccharides from the root bark of Ulmus davidiana var. japonica. J. Cosmet. Sci. 2006, 57, 355–367. [Google Scholar] [PubMed]
  328. Yang, H.H.; Son, J.-K.; Jung, B.; Zheng, M.; Kim, J.-R. Epifriedelanol from the Root Bark of Ulmus davidiana Inhibits Cellular Senescence in Human Primary Cells. Planta Med. 2011, 77, 441–449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  329. Choi, Y.-R.; Lee, Y.-K.; Chang, Y.H. Structural and rheological properties of pectic polysaccharide extracted from Ulmus davidiana esterified by succinic acid. Int. J. Biol. Macromol. 2018, 120, 245–254. [Google Scholar] [CrossRef]
  330. Svobodová, A.; Zdařilová, A.; Vostálová, J. Lonicera caerulea and Vaccinium myrtillus fruit polyphenols protect HaCaT keratinocytes against UVB-induced phototoxic stress and DNA damage. J. Dermatol. Sci. 2009, 56, 196–204. [Google Scholar] [CrossRef]
  331. Maramaldi, G.; Togni, S.; Di Pierro, F.; Biondi, M. A cosmeceutical formulation based on boswellic acids for the treatment of erythematous eczema and psoriasis. Clin. Cosmet. Investig. Dermatol. 2014, 7, 321–327. [Google Scholar] [CrossRef] [Green Version]
  332. Bucci, P.; Prieto, M.J.; Milla, L.; Calienni, M.N.; Martinez, L.; Rivarola, V.; Alonso, A.; Montanari, J. Skin penetration and UV-damage prevention by nanoberries. J. Cosmet. Dermatol. 2018, 17, 889–899. [Google Scholar] [CrossRef]
  333. Skarupova, D.; Vostalova, J.; Svobodova, A.R. Ultraviolet A protective potential of plant extracts and phytochemicals. Biomed. Pap. Med. Fac. Univ. Palacky. Olomouc. Czech. Repub. 2020, 164, 1–22. [Google Scholar] [CrossRef] [Green Version]
  334. Calò, R.; Marabini, L. Protective effect of Vaccinium myrtillus extract against UVA- and UVB-induced damage in a human keratinocyte cell line (HaCaT cells). J. Photochem. Photobiol. B Biol. 2014, 132, 27–35. [Google Scholar] [CrossRef]
  335. Widy-Tyszkiewicz, E. EMA Assessment Report on Vaccinium myrtillus L., Fructus. Available online: https://www.ema.europa.eu/en/documents/herbal-report/draft-assessment-report-vaccinium-myrtillus-l-fructus_en.pdf (accessed on 8 July 2020).
  336. Fiume, M.M.; Bergfeld, W.F.; Belsito, D.V.; Hill, R.A.; Klaassen, C.D.; Liebler, D.C.; Marks, J.G.; Shank, R.C.; Slaga, T.J.; Snyder, P.W.; et al. Safety Assessment of Vitis vinifera (Grape)-Derived Ingredients as Used in Cosmetics. Int. J. Toxicol. 2014, 33, 48S–83S. [Google Scholar] [CrossRef]
  337. Durazzo, A.; Lucarini, M.; Santini, A. Nutraceuticals in Human Health. Foods 2020, 9, 370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  338. Dresch, R.R.; Dresch, M.T.K.; Biegelmeyer, R.; Argenta, D.F.; da Rocha, R.F.; Teixeira, H.F.; Moreira, J.C.F.; Henriques, A.T. Potential use of secondary products of the agri-food industry for topical formulations and comparative analysis of antioxidant activity of grape leaf polyphenols. Nat. Prod. Res. 2018, 32, 486–492. [Google Scholar] [CrossRef] [PubMed]
  339. Boo, Y.C. Human Skin Lightening Efficacy of Resveratrol and Its Analogs: From in Vitro Studies to Cosmetic Applications. Antioxidants 2019, 8, 332. [Google Scholar] [CrossRef] [Green Version]
  340. Minciullo, P.L.; Calapai, G.; Miroddi, M.; Mannucci, C.; Chinou, I.; Gangemi, S.; Schmidt, R.J. Contact dermatitis as an adverse reaction to some topically used European herbal medicinal products—Part 4: Solidago virgaureaVitis vinifera. Contact Dermat. 2017, 77, 67–87. [Google Scholar] [CrossRef] [PubMed]
  341. Enwere, N.J.; Hung, Y.-C. Some chemical and physical properties of bambara groundnut (Voundzeia subterrunea Thouars) seed and products. Int. J. Food Sci. Nutr. 1996, 47, 469–475. [Google Scholar] [CrossRef]
  342. Marcel, A.; Bienvenu, M.J. Chemical and phytochemical compositions of Voandzeia subterranea seeds. Pak. J. Biol. Sci. 2014, 17, 1083–1088. [Google Scholar] [CrossRef] [Green Version]
  343. Gilles Pauly, N. Cosmetic Composition containing an Extract from the seed of Bambara (Voandzeia subterranea) Nut. U.S. Patent No. 6,391,320, 21 May 2002. [Google Scholar]
  344. Roudsari, M.R.; Karimi, R.; Sohrabvandi, S.; Mortazavian, A.M. Health Effects of Probiotics on the Skin. Crit. Rev. Food Sci. Nutr. 2015, 55, 1219–1240. [Google Scholar] [CrossRef]
  345. Bustamante, M.; Oomah, B.D.; Oliveira, W.P.; Burgos-Díaz, C.; Rubilar, M.; Shene, C. Probiotics and prebiotics potential for the care of skin, female urogenital tract, and respiratory tract. Folia Microbiol. 2020, 65, 245–264. [Google Scholar] [CrossRef] [Green Version]
  346. Maguire, G. The role of microbiota, and probiotics and prebiotics in skin health. Arch. Dermatol. Res. 2017, 309, 411–421. [Google Scholar] [CrossRef]
  347. Al-Ghazzewi, F.H.; Tester, R.F. Impact of prebiotics and probiotics on skin health. Benef. Microbes. 2014, 5, 99–107. [Google Scholar] [CrossRef] [PubMed]
  348. Krutmann, J. Pre- and probiotics for human skin. J. Dermatol. Sci. 2009, 54, 1–5. [Google Scholar] [CrossRef]
  349. Knackstedt, R.; Knackstedt, T.; Gatherwright, J. The role of topical probiotics in skin conditions: A systematic review of animal and human studies and implications for future therapies. Exp. Dermatol. 2019, 29, 15–21. [Google Scholar] [CrossRef] [Green Version]
  350. Bindurani, S. Review: Probiotics in dermatology. J. Skin Sex. Transm. Dis. 2019, 1, 66–71. [Google Scholar] [CrossRef]
  351. Van der Hoeven, H. The skin microbiome, probiotics and skin care. Pers. Care 2017, 9, 25–27. [Google Scholar]
  352. Isolauri, E.; Arvola, T.; Sütas, Y.; Moilanen, E.; Salminen, S. Probiotics in the management of atopic eczema. Clin. Exp. Allergy 2000, 30, 1605–1610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  353. Caramia, G.; Atzei, A.; Fanos, V. Probiotics and the skin. Clin. Dermatol. 2008, 26, 4–11. [Google Scholar] [CrossRef] [PubMed]
  354. Mottin, V.H.M.; Suyenaga, E.S. An approach on the potential use of probiotics in the treatment of skin conditions: Acne and atopic dermatitis. Int. J. Dermatol. 2018, 57, 1425–1432. [Google Scholar] [CrossRef] [PubMed]
  355. Guéniche, A.; Bastien, P.; Ovigne, J.M.; Kermici, M.; Courchay, G.; Chevalier, V.; Breton, L.; Castiel-Higounenc, I. Bifidobacterium longum lysate, a new ingredient for reactive skin. Exp. Dermatol. 2010, 19, e1–e8. [Google Scholar] [CrossRef]
  356. Repair Complex CLR™ PF. Available online: https://www.ulprospector.com/documents/987102.pdf?bs=1382&b=90742&st=1&sl=92671889&crit=a2V5d29yZDpbQklGSURBIEZFUk1FTlQgTFlTQVRFXQ%3D%3D&k=BIFIDA%7CFERMENT%7CLYSATE&r=eu&ind=personalcare (accessed on 24 March 2021).
  357. Van der Hoeven, H.; Prade, H. Epidermal anti-ageing with a probiotic skin care approach. Pers. Care 2015, 9, 63–66. [Google Scholar]
  358. ProRenew Complex CLR™ NP. Available online: https://www.ulprospector.com/documents/1542305.pdf?bs=1382&b=736219&st=1&sl=92726643&crit=TGFjdG9jb2NjdXMgRmVybWVudCBMeXNhdGU%3D&r=eu&ind=personalcare (accessed on 24 March 2021).
  359. Mateu, M.; Davi, C.; Cañadas, E.; Soley, A.; Delgado, R. Sebum production and pore size finally under control. Pers. Care 2015, 4, 58–62. [Google Scholar]
  360. Lolou, V.; Panayiotidis, M.I. Functional Role of Probiotics and Prebiotics on Skin Health and Disease. Fermentation 2019, 5, 41. [Google Scholar] [CrossRef] [Green Version]
  361. Matmarine™ Biotech Ingredient G, Lipotec S.A.U. Available online: https://www.ulprospector.com/documents/1343813.pdf?bs=2316&b=525392&st=1&sl=92726092&crit=UHNldWRvYWx0ZXJvbW9uYXMgRmVybWVudCBFeHRyYWN0&r=eu&ind=personalcare (accessed on 24 March 2021).
  362. Ahsan, H. Immunopharmacology and immunopathology of peptides and proteins in personal products. J. Immunoass. Immunochem. 2019, 40, 439–447. [Google Scholar] [CrossRef]
  363. Burnett, C.L.; Heldreth, B.; Bergfeld, W.F.; Belsito, D.V.; Hill, R.A.; Klaassen, C.D.; Liebler, D.C.; Marks, J.G.J.; Shank, R.C.; Slaga, T.J.; et al. Safety Assessment of α-Amino Acids as Used in Cosmetics. Int. J. Toxicol. 2013, 32, 41S–64S. [Google Scholar] [CrossRef]
  364. Federici, A.; Federici, G.; Milani, M. An urea, arginine and carnosine based cream (Ureadin Rx Db ISDIN) shows greater efficacy in the treatment of severe xerosis of the feet in Type 2 diabetic patients in comparison with glycerol-based emollient cream. A randomized, assessor-blinded, controll. BMC Dermatol. 2012, 12, 16. [Google Scholar] [CrossRef] [Green Version]
  365. Marseglia, A.; Licari, A.; Agostinis, F.; Barcella, A.; Bonamonte, D.; Puviani, M.; Milani, M.; Marseglia, G.; Matteo, P.S. Local rhamnosoft, ceramides and L-isoleucine in atopic eczema: A randomized, placebo controlled trial. Pediatr. Allergy Immunol. Orig. 2014, 25, 271–275. [Google Scholar] [CrossRef] [Green Version]
  366. Lungu, C.; Considine, E.; Zahir, S.; Ponsati, B.; Arrastia, S.; Hallett, M. Pilot study of topical acetyl hexapeptide-8 in the treatment for blepharospasm in patients receiving botulinum toxin therapy. Eur. J. Neurol. 2013, 20, 515–518. [Google Scholar] [CrossRef] [Green Version]
  367. Lim, S.H.; Sun, Y.; Thiruvallur, T.M.; Rosa, V.; Kang, L. Enhanced Skin Permeation of Anti-wrinkle Peptides via Molecular Modification. Sci. Rep. 2018, 8, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  368. Kraeling, M.E.K.; Zhou, W.; Wang, P.; Ogunsola, O.A. In vitro skin penetration of acetyl hexapeptide-8 from a cosmetic formulation. Cutan. Ocul. Toxicol. 2014, 34, 46–52. [Google Scholar] [CrossRef]
  369. Campos, V.; Kalil, C.; Reinehr, C.; Canavaci, G.; Beltrao, F. Observational studies with confirmation of safety, tolerance and efficacy of a facial sterile solution for the skin rejuvenation containing hyaluronic acid, acetyl hexapeptide-8, carnitine and pyruvic acid in association to laser procedure. J. Am. Acad. Dermatol. 2017, 76, AB182. [Google Scholar] [CrossRef]
  370. Rull, M.; Davi, C.; Cañadas, E.; Cebrián, J.; Delgado, R. Reversing signs of ageing in mature skin. Pers. Care 2012, 9, 75–77. [Google Scholar]
  371. Park, J.; Jung, H.; Jang, B.; Song, H.; Han, I.; Oh, E. D-tyrosine adds an anti-melanogenic effect to cosmetic peptides. Sci. Rep. 2020, 10, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  372. Dragomirescu, A.O.; Andoni, M.; Ionescu, D.; Andrei, F. The Efficiency and Safety of Leuphasyl—A Botox-Like Peptide. Cosmetics 2014, 1, 75–81. [Google Scholar] [CrossRef] [Green Version]
  373. Dipeptide Diaminobutyroyl Benzylamide Diacetate. Available online: https://www.ulprospector.com/en/eu/PersonalCare/search?k=dipeptide+diaminobutyroyl benzylamide diacetate&st=1 (accessed on 24 March 2021).
  374. Hahn, H.J.; Jung, H.J.; Schrammek-Drusios, C.; Lee, S.N.; Kim, J.I.H.; Kwon, S.B.; An, I.-S.; An, S.; Ahn, K.J. Instrumental evaluation of anti-aging effects of cosmetic formulations containing palmitoyl peptides, Silybum marianum seed oil, vitamin E and other functional ingredients on aged human skin. Exp. Ther. Med. 2016, 12, 1171–1176. [Google Scholar] [CrossRef] [Green Version]
  375. Johnson, W., Jr.; Bergfeld, W.F.; Belsito, D.V.; Hill, R.A.; Klaassen, C.D.; Liebler, D.C.; James, M.G.; Shank, R.C.; Slaga, T.J.; Snyder, P.W.; et al. Safety Assessment of Tripeptide-1, Hexapeptide-12, Their Metal Salts and Fatty Acyl Derivatives, and Palmitoyl Tetrapeptide-7 as Used in Cosmetics. Int. J. Toxicol. 2018, 37, 90S–102S. [Google Scholar] [CrossRef]
  376. Gianeti, M.D.; Gaspar, L.R.; Bueno de Camarago Júnior, F.; Berardo Gonçalves Maia Campos, P.M. Benefits of Combinations of Vitamin A, C and E Derivatives in the Stability of Cosmetic Formulations. Molecules 2012, 17, 2219–2230. [Google Scholar] [CrossRef]
  377. Narda, M.; Brown, A.; Muscatelli-Groux, B.; Grimaud, J.A.; Granger, C. Epidermal and Dermal Hallmarks of Photoaging are Prevented by Treatment with Night Serum Containing Melatonin, Bakuchiol, and Ascorbyl Tetraisopalmitate: In Vitro and Ex Vivo Studies. Dermatol. Ther. 2020, 10, 191–202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  378. Assier, H.; Wolkenstein, P.; Grille, C.; Chosidow, O. Contact dermatitis caused by ascorbyl tetraisopalmitate in a cream used for the management of atopic dermatitis. Conta 2014, 71, 60–61. [Google Scholar] [CrossRef]
  379. Swinnen, I.; Goossens, A. Allergic contact dermatitis caused by ascorbyl tetraisopalmitate. Contact Dermat. 2011, 64, 241–242. [Google Scholar] [CrossRef] [PubMed]
  380. Wohlrab, J. Niacinamide—Mechanisms of Action and Its Topical Use in Dermatology. Skin Pharmacol. Physiol. 2014, 27, 311–315. [Google Scholar] [CrossRef] [PubMed]
  381. Panel, C.I.R.E. Final Report of the Safety Assessment of Niacinamide and Niacin. Int. J. Toxicol. 2014, 24, 1–31. [Google Scholar] [CrossRef]
  382. Pavlačková, J.; Egner, P.; Sedláček, T.; Mokrejš, P.; Sedlaříková, J.; Polášková, J. In vivo efficacy and properties of semisolid formulations containing panthenol. J. Cosmet. Dermatol. 2018, 18, 346–354. [Google Scholar] [CrossRef] [Green Version]
  383. Fernandes, R.A.; Santiago, L.; Gouveia, M.; Gonçalo, M. Allergic contact dermatitis caused by dexpanthenol—Probably a frequent allergen. Contact Dermat. 2018, 79, 276–280. [Google Scholar] [CrossRef]
  384. Miroux-Catarino, A.; Silva, L.; Amaro, C.; Viana, I. Allergic contact dermatitis caused dexpanthenol—But is that all? Contact Dermat. 2019, 81, 391–392. [Google Scholar] [CrossRef]
  385. Silva, S.; Ferreira, M.; Oliveira, A.S.; Magalhães, C.; Sousa, M.E.; Pinto, M.; Lobo Sousa, J.M.; Almeida, I.F. Evolution of the use of antioxidants in anti-ageing cosmetics. Int. J. Cosmet. Sci. 2019, 41, 378–386. [Google Scholar] [CrossRef] [PubMed]
  386. Mukherjee, S.; Date, A.; Patravale, V.; Korting, H.C.; Roeder, A.; Weindl, G. Retinoids in the treatment of skin aging: An overview of clinical efficacy and safety. Clin. Interv. Aging 2006, 1, 327–348. [Google Scholar] [CrossRef]
  387. Fiume, M.M.; Bergfeld, W.F.; Belsito, D.V.; Ronald, A.H.; Klaassen, C.D.; Liebler, D.C.; Marks, G.J.J.; Shank, R.C.; Slaga, T.J.; Snyder, P.W.; et al. Safety Assessment of Tocopherols and Tocotrienols as Used in Cosmetics. Int. J. Toxicol. 2018, 37, 61S–94S. [Google Scholar] [CrossRef] [Green Version]
  388. Ohtake, S.; Wang, Y.J. Trehalose: Current Use and Future Applications. J. Pharm. Sci. 2011, 100, 2020–2053. [Google Scholar] [CrossRef]
  389. Cai, X.; Seitl, I.; Mu, W.; Zhang, T.; Stressler, T.; Fischer, L.; Jiang, B. Biotechnical production of trehalose through the trehalose synthase pathway: Current status and future prospects. Appl. Microbiol. Biotechnol. 2018, 102, 2965–2976. [Google Scholar] [CrossRef]
  390. Cornara, L.; Biagi, M.; Xiao, J.; Burlando, B. Therapeutic Properties of Bioactive Compounds from Different Honeybee Products. Front. Pharmacol. 2017, 8. [Google Scholar] [CrossRef] [PubMed]
  391. Burlando, B.; Cornara, L. Honey in dermatology and skin care: A review. J. Cosmet. Dermatol. 2013, 12, 306–313. [Google Scholar] [CrossRef]
  392. Viuda-Martos, M.; Ruiz-Navajas, Y.; Fernández-López, J.; Pérez-Álvarez, J.A. Functional Properties of Honey, Propolis, and Royal Jelly. J. Food Sci. 2008, 73, 117–124. [Google Scholar] [CrossRef] [PubMed]
  393. Cole, N.; Sou, P.W.; Ngo, A.; Tsang, K.H.; Severino, J.A.J.; Arun, S.J.; Duke, C.C.; Reeve, V.E. Topical ‘Sydney’ Propolis Protects against UV-Radiation-Induced Inflammation, Lipid Peroxidation and Immune Suppression in Mouse Skin. Int. Arch. Allergy Immunol. 2010, 152, 87–97. [Google Scholar] [CrossRef]
  394. Duplan, H.; Questel, E.; Hernandez-Pigeon, H.; Galliano, M.F.; Caruana, A.; Ceruti, I.; Ambonati, M.; Mejean, C.; Damour, O.; Castex-Rizzi, N.; et al. Effects of Hydroxydecine® (10-hydroxy-2-decenoic acid) on skin barrier structure and function. Eur. J. Dermatol. 2011, 2, 906–915. [Google Scholar] [CrossRef]
  395. Park, H.M.; Hwang, E.; Lee, K.G.; Han, S.-M.; Cho, Y.; Kim, S.Y. Royal Jelly Protects Against Ultraviolet B–Induced Photoaging in Human Skin Fibroblasts via Enhancing Collagen Production. J. Med. Food 2011, 14, 899–906. [Google Scholar] [CrossRef] [Green Version]
  396. Rosmilah, M.; Shahnaz, M.; Patel, G.; Lock, J.; Rahman, D.; Masita, A.; Noormalin, A. Characterization of major allergens of royal jelly Apis mellifera. Trop. Biomed. 2009, 25, 243–251. [Google Scholar]
  397. Walgrave, S.E.; Warshaw, E.M.; Glesne, L.A. Allergic Contact Dermatitis from Propolis. Dermatitis 2005, 16, 209–215. [Google Scholar] [CrossRef] [PubMed]
  398. Nishinami, S.; Yoshizawa, S.; Arakawa, T.; Shiraki, K. Allantoin and hydantoin as new protein aggregation suppressors and their mechanisms of action. Int. J. Biol. Macromol. 2018, 114. [Google Scholar] [CrossRef]
  399. Becker, L.C.; Bergfeld, W.F.; Belsito, D.V.; Klaassen, C.D.; Marks, J.G.J.; Shank, R.C.; Slaga, T.J.; Snyder, P.W.; Andersen, F.A. Final Report of the Safety Assessment of Allantoin and Its Related Complexes. Int. J. Toxicol. 2010, 29, 84S–97S. [Google Scholar] [CrossRef]
  400. Angelova-Fischer, I.; Rippke, F.; Richter, D.; Filbry, A.; Arrowitz, C.; Weber, T.; Fischer, T.W.; Zillikens, D. Stand-alone Emollient Treatment Reduces Flares After Discontinuation of Topical Steroid Treatment in Atopic Dermatitis: A Double-blind, Randomized, Vehicle-controlled, Left-right Comparison Study. Acta Derm. Venereol. 2018, 98, 517–523. [Google Scholar] [CrossRef] [Green Version]
  401. Yilmaz, E.; Borchert, H.-H. Effect of lipid-containing, positively charged nanoemulsions on skin hydration, elasticity and erythema—An in vivo study. Int. J. Pharm. 2006, 307, 232–238. [Google Scholar] [CrossRef] [PubMed]
  402. Choi, S.M.; Lee, B.-M. Safety and risk assessment of ceramide 3 in cosmetic products. Food Chem. Toxicol. 2015, 84, 8–17. [Google Scholar] [CrossRef]
  403. Cao, M.; Li, J.; Tang, J.; Chen, C.; Zhao, Y. Gold Nanomaterials in Consumer Cosmetics Nanoproducts: Analyses, Characterization, and Dermal Safety Assessment. Small 2016, 12, 5488–5496. [Google Scholar] [CrossRef]
  404. Cornier, J.; Keck, C.M.; Van de Voorde, M. Nanocosmetics. From Ideas to Products; Springer Nature Switzerland: Cham, Switzerland, 2019; ISBN 9783030165727. [Google Scholar]
  405. Pulit-Prociak, J.; Grabowska, A.; Majka, T.M. Safety of the application of nanosilver and nanogold in topical cosmetic preparations. Colloids Surf. B Biointerfaces 2019, 183. [Google Scholar] [CrossRef]
  406. Wee, Y.-J.; Kim, J.-N.; Ryu, H.-W. Biotechnological Production of Lactic Acid and Its Recent Applications. Food Technol. Biotechnol. 2006, 44, 163–172. [Google Scholar]
  407. Scherdin, U.; Presto, S.; Rippke, F.; Nielsen, J.; Strassner, M.; Imadojemun, A.; Gärtner, E.; Herpens, A.; Korting, H.C.; Bielfeldt, S. In vivo assessment of the efficacy of an innovative face care system in subjects with mild acne vulgaris. Int. J. Cosmet. Sci. 2004, 26, 221–229. [Google Scholar] [CrossRef] [PubMed]
  408. Singh, R.; Goyal, S.; Ahmed, Q.R.; Gupta, N.; Singh, S. Effect of 82 % Lactic Acid in Treatment of Melasma. Int. Sch. Res. Not. 2014, 1–7. [Google Scholar] [CrossRef] [Green Version]
  409. Fiume, Z. Final Report on the Safety Assessment of Lecithin and Hydrogenated Lecithin. Int. J. Toxicol. 2001, 20, 20–45. [Google Scholar] [CrossRef]
  410. Mala’Kîte™ Protective Mineral Complex. Available online: https://www.gattefosse.com/personal-care-actives/malakite (accessed on 25 March 2021).
  411. Lin, S.-Y.; Lin, T.-C. In Vitro Repairability for the Disordered Skin by Pyrrolidone-Carboxylate Sodium. Drug Dev. Ind. Pharm. 1994, 20, 665–676. [Google Scholar] [CrossRef]
  412. Fiume, M.M.; Bergfeld, W.F.; Belsito, D.V.; Hill, R.A.; Klaassen, C.D.; Liebler, D.C.; Marks, J.G., Jr.; Shank, R.C.; Slaga, T.J.; Snyder, P.W.; et al. Safety Assessment of PCA (2-Pyrrolidone-5-Carboxylic Acid) and Its Salts as Used in Cosmetics. Int. J. Toxicol. 2019, 38, 5S–11S. [Google Scholar] [CrossRef]
  413. Lods, L.M.; Dres, C.; Johnson, C.; Scholz, D.B.; Brooks, G.J. The future of enzymes in cosmetics. Int. J. Cosmet. Sci. 2000, 22, 85–94. [Google Scholar] [CrossRef]
  414. Ahmad Nasrollahi, S.; Ayatollahi, A.; Yazdanparast, T.; Samadi, A.; Hosseini, H.; Shamsipour, M.; Akhlaghi, A.A.; Yadangi, S.; Abels, C.; Firoozz, A. Comparison of linoleic acid-containing water- in-oil emulsion with urea-containing water-in-oil emulsion in the treatment of atopic dermatitis: A randomized clinical trial. Clin. Cosmet. Investig. Dermatol. 2018, 11, 21–28. [Google Scholar] [CrossRef] [Green Version]
  415. Yamarik, T.A.; Elmore, A.R. Final Report of the Safety Assessment of Urea. Int. J. Toxicol. 2005, 24, 1–56. [Google Scholar] [CrossRef]
  416. Bissonnette, R.; Maari, C.; Provost, N.; Bolduc, C.; Nigen, S.; Rougier, A.; Seite, S. A double-blind study of tolerance and efficacy of a new urea-containing moisturizer in patients with atopic dermatitis. J. Cosmet. Dermatol. 2010, 9, 16–21. [Google Scholar] [CrossRef] [PubMed]
  417. Celleno, L. Topical urea in skincare: A review. Dermatol. Ther. 2018, e12690. [Google Scholar] [CrossRef]
Figure 1. Chemical structure of hyaluronic acid (HA).
Figure 1. Chemical structure of hyaluronic acid (HA).
Molecules 26 04429 g001
Figure 2. Hyaluronic acid market-regional comparison.
Figure 2. Hyaluronic acid market-regional comparison.
Molecules 26 04429 g002
Figure 3. Cosmetic, pharmaceutical, and medical applications of HA and its derivates.
Figure 3. Cosmetic, pharmaceutical, and medical applications of HA and its derivates.
Molecules 26 04429 g003
Figure 4. Hyaluronic acid activity, molecular weight dependence and claimed effect of HA. (TEWL—Transepidermal Water Loss; DEJ—Dermoepidermal junction).
Figure 4. Hyaluronic acid activity, molecular weight dependence and claimed effect of HA. (TEWL—Transepidermal Water Loss; DEJ—Dermoepidermal junction).
Molecules 26 04429 g004
Figure 5. Cosmetic and nutricosmetic effects of HA.
Figure 5. Cosmetic and nutricosmetic effects of HA.
Molecules 26 04429 g005
Figure 6. The images of skin texture before product application (D0) and after 28 days (D28) of regular application of an anti-ageing cream incorporating 0.5% (w/w) LMW-HA (20–50 KDa) and 3% (w/w) encapsulated HMW-HA (1–1.4 MDa) [105].
Figure 6. The images of skin texture before product application (D0) and after 28 days (D28) of regular application of an anti-ageing cream incorporating 0.5% (w/w) LMW-HA (20–50 KDa) and 3% (w/w) encapsulated HMW-HA (1–1.4 MDa) [105].
Molecules 26 04429 g006
Table 1. Commercially available cosmetics incorporating HA and HA derivatives.
Table 1. Commercially available cosmetics incorporating HA and HA derivatives.
Cosmetic Product Category/Proposed UseProducer (Country of Origin)/Cosmetic Product Trade NameHA FormOther Active Ingredients Incorporated in the Cosmetic Formulation (INCI—International Nomenclature of Cosmetic Ingredients Denomination)Cosmetic ClaimMarket SegmentReference
Skin care cosmeticsFRESH (USA)
Deep Hydration
Face Serum
Serum
NaHAPorphyridium Cruentum Extract, Pyrus Cydonia Seed Extract, Angelica Keiskei Extract, Voandzeia Subterra Nea Seed Extract, Cucumis Sativus (Cucumber) Fruit Extract, Tocopheryl Acetate24-h moisturePremium Market[108]
DECIEM (Canada)
The ORDINARY
“Buffet” Multi-Technology Peptide Serum
Serum
NaHALactococcus Ferment Lysate, Acetyl Hexapeptide-8, Pentapeptide-18, Palmitoyl Tripeptide-1, Palmitoyl Tetrapeptide-7, Palmitoyl Tripeptide-38, Dipeptide Diaminobutyroyl BenzylamideDiacetate, Acetylarginyltryptophyl Diphenylglycine, Allantoin, Glycine, Alanine, Serine, Valine, Isoleucine,
Proline, Threonine, Histidine, Phenylalanin, Arginine, Aspartic, Acid, Trehalose, Fructose, Glucose, Maltose, Urea, Sodium PCA, PCA, Hydroxypropyl Cyclodextrin
anti-ageingPremium Market[109]
DECIEM (Canada)
THE ORDINARY Hyaluronic Acid 2% + B5
Serum
NaHAPanthenol, Ahnfeltia Concinna Extractmoisturizing,
anti-ageing
Premium Market[110]
APIVITA (Greece)
5-action Eye Serum Advanced eye care
Serum
Hydrolyzed HALilium Candidum Extract, PfaffiaPaniculata Extract, PtychopetalumOlacoides Extract, CopperLysinate/Prolinate, Propolis Extract, Mel Extract, Methylglucoside Phosphate, Euphrasia Officinalis Extract, Lecithin, Hydroxypropyl Cyclodextrin, Ascorbyl Tetraisopalmitate, Panthenol, SideritisPerfoliata Extract, Aloe Barbadensis Extract, SideritisScardica Extract, SideritisRaeseri Extract, Bisabololanti-ageing, moisturizingPremium Market[111]
FARMEC (Romania)
Hyaluronic Acid ampoules 5%
Serum
NaHASuperoxide Dismutase, Lecithinanti-wrinkle,
intensive moisturizing
Mass Market[112]
AVIVA COSMETICS (Romania)
INFINITUM Deep Wrinkles Anti-Ageing Serum
Hydrolyzed HAAesculus Hippocastanum Extractfirming, anti-ageingPremium Market[113]
GARANCIA (France) MYSTÉRIEUX MILLE ET UN JOURS Anti-Ageing Day EmulsionHydrolyzed HAAlaria Esculenta Extract, Cyathea Cumingii Extract, Dipeptide Diaminobutyroyl Benzylamide Diacetate, Hydroxypropyl Cyclodextrin, Palmitoyl Tripeptide-38relax expression linesPremium Market[114]
BALANCE ME (UK)
Tinted Wonder Eye Cream Eye cream complexion perfection
Day Cream
Hydrolyzed HAPicea Abies Extract, Rubus Chamaemorus Seed Extract, Aloe Barbadensis Extract, Styrax Benzoin Extract, Rosmarinus Officinalis Extract, Vitis Vinifera Seed Extract, Tocopherolsoothing,
anti-ageing
Mass Market[115]
EARTH SCIENCE (USA)
Apricot
Intensive night cream
Night Cream
NaHAPyrus Malus Extract, Glycyrrhiza Glabra Extract, Glycine Soja Seed Extract, Tocopherol, Citrus Grandis Seed Extract, Calendula Officinalis Extract, Bambusa Arundinacea Extract, Sambucus Nigra Extract, Cucumis Sativus Extract, Arnica Montana Extract, Hedera Helix Extract, Lactic acid, Allantoinreduces the appearance of fine lines and wrinklesMass Market[116]
COSMETIC PLANT (Romania)
Lift Up—Hyaluronic Acid Day Anti-Ageing Cream
Day Cream
Hydrolyzed HAPorphyridium Cruentum Extract, Magnolia Liliflora Extract, Tocopherolanti-ageingMass Market[117]
AVIVA COSMETICS (Romania)
INFINITUM Cellular Regenerating Cream
Hydrolyzed HAAesculus Hippocastanum Extract, Tocopheryl Acetatefirming, regenerating, rejuvenatingPremium Market[118]
GEROCOSSEN (Romania)
Hyaluron Anti-Age CreamSPF 10 Day Cream
NaHALecithin, Tocopheryl acetate, Retinyl palmitateanti-ageingMass Market[119]
AVIVA COSMETICS (Romania)
INFINITUM Golden Elixir Anti-Ageing Cream SPF 15 Day Cream
Hydrolyzed HAAesculus Hippocastanum Extract, Gold (CI 777480), Ectoinfirming,
anti-ageing,
SPF 15
Premium Market[120]
FARMEC (Romania)
Anti-Wrinkle Cream Concentrated with Hyaluronic Acid 3%
Day Cream
NaHAPseudoalteromonas Ferment Extract, Hydrolyzed Wheat Protein, Hydrolyzed Soy Protein, Tripeptide-10 Citrulline, Tripeptide-1, Lecithin, Artemia Salina Extract, Superoxide Dismutaseanti-wrinkle, line filling and smoothing, intense hydrationMass Market[121]
ARBONNE RE9 (USA) Advanced For Men
Anti-Aging Moisturizer Broad Spectrum
Men Care
NaHAAloe Barbadensis Gel, Ceratonia Siliqua Extract, Laminaria Digitata Extract, Malachite Extract, Chamomilla Recutita Extract, Sophora Japonica Flower Extract, Tripleurospermum Maritimum Extractmoisturizing, SPF 15Premium Market[122]
AVIVA COSMETICS (Romania)
INFINITUM Anti-Ageing
Cleansing Emulsion
Hydrolyzed HAAesculus Hippocastanum Extractfirming,
anti-ageing
Premium Market[123]
JONZAC (France)
Bébé Bio Dermo-Repair Cream
Baby Care
Hydrolyzed HAMalva Sylvestris Extract, Tocopherol, Hydrogenated Lecithinsoothing, regenerating, protectingMass Market[124]
Body cosmeticsLES MERVEILLEUSES LADUREE (France)
Sun Protection Body Cream
Body Cream
NaHARosa Centifolia Extract, Rosa Damascena Extracthydrating,
SPF 50
Premium Market[125]
NATURE REPUBLIC (South Korea)
Ice Sun, Ice Puff Sun
Sun/Sunbed Exposure
NaHACalendula Officinalis Extractanti-wrinkle and skin bleftening, SPF 50Mass Market[126]
DM ALVERDE NATURKOSMETIK (Germany)
Ivital +
Hand cream
NaHAAmaranthus Caudatus Extract, Triticum Aestivum Germ Extract, Punica Granatum Extract, Tocopherolsmoothes lines and wrinklesMass Market[127]
INNISFREE (South Korea)
My Body Tangerine Blossom
Body lotion
Hydrolyzed HACitrus Unshiu Peel Extract, Algae Extract, Eclipta Prostrata Extract, Orhid Extract, Camelia Sinensis Extract, Camelia Japonica Extract, Opuntia Coccinellifera Extract, Tocopherys AcetatemoisturizingMass Market[128]
INNISFREE (South Korea)
Wine Peeling Jelly Scrub
Peeling Scrub
NaHAVitis ViniferaExtract, Ulmus Davidiana Root Extract, Amaranthus Caudatus Extract, Centella Asiatica Extract, Ficus Carica Extract, Citrus Unshiu Extract, Orchis Extract, Camelia Sinensis Extract, Camelia Japonica Extract, Opuntia Coccinellifera Extract, Niacinamide, Allantoin, Ceramide 3moisturizingMass Market[129]
TUDE HOUSE (South Korea)
Berry AHA! Bleft Peel Bubble Wash
Hydrolyzed HAAlgae Extract, Eclipta Prostrata Extract, Vaccinium Myrtillus Extract, Saccharum Officinarum Extract, Citrus Limonum Extract, Citrus Aurantium Dulcis Extract, Ulmus Davidiana Extract, Amaranthus Caudatus Extract, Acer Saccharinum Extract, Nymphaea Alba Extract, Bifida Ferment Lysate, LecitinblefteningMass Market[130]
Makeup cosmeticsILLAMASQUA (UK) HYPNOTICA Lipe Lure
Colour intense Liquid lipstick
NaHANlong-wearing, picture-perfect finishPremium Market[131]
CLINIQUE (USA)
Sun-Kissed Face Gelee
Complexion multitasker
NaHANmoisturizingPremium Market[132]
PAUL& JOE (Japan)
Pore SmoothingPrimer
Face primer
Hydrolyzed HACalendula Officianlis Extract, Rosa Canina Fruit Extract, TocopherolmoisturizingPremium Market[133]
NaHA—Sodium Hyaluronate; Hydrolized HA—Hydrolized Hyaluronic Acid; N—Not known.
Table 2. The benefits and potential adverse effects of botanical extracts used in cosmetic products associated with hyaluronic acid and HA derivates.
Table 2. The benefits and potential adverse effects of botanical extracts used in cosmetic products associated with hyaluronic acid and HA derivates.
Active Ingredient/Plant SpeciesBioactive ComponentsBiological Role and Benefits for SkinToxicity of Topical Products
Acer saccharumphenolic compounds (maplexins, ginnalins) [134,135]increases the intracellular ceramide level
stimulates the proliferation and differentiation of keratinocytes
maintains thehomeostasis of the epidermis
antioxidant capacity [134,136,137]
* N
Ahnfeltia concinna
(red algae)
polysaccharides (carrageenan, agar), amino acids, minerals, vitamins, trace elements [138,139,140]antioxidant capacity, anti-wrinkle, skin-whitening (suppresses the melanin production), skin moisturizing [139,140,141,142]not toxic in topical applications
possible irritation, sensitization, or photoreactions [142]
Alaria esculenta
(brown algae)
polyphenols, diterpenoids, fatty acids, polysaccharides, fucosterol, fucoxhantin (e.g., retinoic acid), amino acids, minerals, vitamins, trace elements [143,144,145]hyperpigmentation amelioration
increases skin firmness and elasticity
stimulates collagen and keratin synthesis, reducing the progerin production (increased in aged skin) of ‘aged’ fibroblasts
skin-whitening, antioxidant capacity [142,143,146,147]
anti-cellulite and antiedema activity [142,148,149,150]
possible local irritation, sensitization, or photoreactions [142]
Aloe sp.
(Aloe barbadensis)
antraquinones, polysaccharides, phenolic compounds, organic acids [151,152,153,154]antimicrobial activity [155,156]
antioxidant capacity, anti-inflammatory, anti-pyrotic [156,157,158]
improves skin moisture and water retention in the SC [153,156,157,159,160]
phototoxicity, eczema, contact dermatitis [151,152]
Amaranthus caudatusamino-acids, proteins, amylopectin, minerals, vitamins (vitamin A, E, K), fibers, essential fatty acids, triterpenes (squalene) [161,162]antioxidant capacity, moisturizing, skin hydration, suppressing the pigmentation [162,163,164]* N
Angelica keiskeicoumarins, terpens, phenolic compounds [165,166,167,168]antioxidant capacity, astringent, emollient, skin conditioning and protecting, skin whitening and lightening, sunlight protection [165]* N
Arnica montanafatty acids (palmitic, linoleic, myristic), essential oil, triterpenic alcohols, sugars, phytosterols, phenol acids, tannins, choline, inulin, flavonoids, carotenoids, coumarins, sesquiterpene lactones [169,170]anti-inflammatory effect [158,171]
reduces the ecchymosis and oedema [158]
contact dermatitis [169,172]
rarely ocular irritation [170]
Artemia salinadiguanosine-tetraphosphate, D-myo-inosite-1,4,5-triphosphate, proteins, glucan [173]stimulates skin regeneration, strengthens the immune system of the skin, sustains epidermal cell proliferation, anti-ageing (inhibiting the cells senescence, stimulating the collagen expression) and photo-protection effect [173]* N
Bambusa arundinaceaflavonoids, phenolic compounds, chlorogenic acid, caffeic acid, ferulic acid, 8-C-glucosyl apigenin, luteolin derivatives [174]antioxidant capacity, anti-ageing, photo-protection, skin pigmentation modulating effect, anti-allergic effect (inhibiting the production of IgE and lymphocytes) [174]* N
Calendula officinalistriterpenes, polyphenolic compounds, polysaccharides, vitamin C, tocopherols, quinones, carotenoids saponins, sterols [175,176,177,178]anti-inflammatory effect [175,179]
cell rejuvenation effect [179,180]
skin smoothing and softening effect [179]
prevents skin alteration and early ageing (improving the skin elasticity) [176,180,181]
anti-irritant, anti-psoriatic and callus treating [158]
stimulates the regeneration and epithelisation of wounded skin [175,179,180,181]
antioxidant capacity [175,176,179,180]
antimicrobial activity [155,179]
rare sensitization or allergic contact reactions [169,175,182]
Camelia sp.
(C. sinensis,
C. japonica)
polyphenols, catechins [176]antioxidant capacity, reduces the sebum production, improves skin hydration, skin smoothing and softening, photoprotective, and anti-inflammatory effect [176,183,184,185,186,187]contact dermatitis [172]
Chamomila recutitapolysaccharides, flavonoids (α-bisabolol, apigenin), sesquiterpene lactones [169,176]anti-inflammatory effect, antioxidant capacity [188]
repairs the SC, wound healing, anti-ageing activity
skin smoothing and softening, also being used in the treatment of eczema [176,188,189]
skin sensitization, contact dermatitis [169,189,190]
Centella asiaticasaponins, flavonoids (quercetin, kaempferol, apigenil, naringenin, phenolic acids (chlorogenic acid), triterpenic steroids, amino acids, sugars [191,192]antioxidant capacity, anti-inflammatory, anti-ageing effect
stimulates the fibroblast proliferation, increasing the collagen synthesis
increases the SC hydration and the epidermal barrier function
anti-cellulite effect [191,192]
local allergic reactions, burning, eczemas, vesicles, pruritus [192]
Ceratonia siliquaflavonoids, phytosterols, acids, esters, terpenoids, fenolic compounds [193,194,195]anti-cellulite effect (increasing the aquaglyceroporines activity, stimulating lipolysis, reducing the localized fat overload), antioxidant capacity, depigmentation effect (anti-tyrosinase activity), skin lightening effect [193,194,196]non-irritation potential [193]
Citrus sp.
(C. aurantium dulcis,
C. grandis,
C. limon,
C. unshiu peel)
bergapten, bergamotin, isopimpinellin, coumarins, psoralenes, angelical, volatile oil (limonene, linalool, linalyl acetate, terpineol, terpinene, terpinolene, ocimene, pinene etc.) [197,198,199]antioxidant capacity, anti-inflammatory, antiseptic and anti-verrucous effect, used in acne treatment, wound healing properties, used as fragrance [158,197,198,199]possible irritation, skin sensitization, hyperpigmentation, redness, oedema, photo-toxicity [197,198,199,200]
Cucumis sativusflavonoids, saponins, sterols, carbohydrates, triterpenes, vitamins (C, B), fatty acid, proteins [201,202]used in the treatment of periorbital edema, soothing emollient, anti-wrinkle, anti-ageing, anti-itching effects depigmentant and antioxidant capacity, anti-hyaluronidase and anti-elastase actions [201]erythema after semi occlusive patch testing and conjunctival hyperemia/ocular irritation after a using of an eye lotion containing 1% C. sativus (cucumber) fruit extract [202]
Cydonia oblonga seedcellulose, polysaccharides, polyphenols [203,204]stimulates fibroblasts proliferation, wound, and burns healing
maintains skin barrier function
anti-inflammatory, anti-allergic and protective effects antioxidant capacity [203,204,205,206,207,208]
* N
Eclipta prostrataflavonoids (luteolin, apigenin), wedelolactone, terpenoids, sterols, alkaloids, volatile oils [209,210,211,212,213]depigmentant effect (anti-tyrosinase activity)
hair revitalizing, dye in cosmetic products
antioxidant and photoprotective capacity [209,210,211,212]
* N
Euphrasia officinalisiridoids, flavonoids, polyphenols, saponins, alkaloids, tannins, etheric oils [214,215]anti-inflammatory and astringent effect
antioxidant capacity
photo-protection, protective effect against photo-ageing, stimulates collagen synthesis [214,216,217]
* N
Ficus caricaphytosterols, anthocyanins, amino acids, atty acids, phenolic acids, flavonoids, volatile components [218,219]antioxidant capacity, anti-warts and anti-verrucous activity
used in the treatment of dry skin, eczema, acne
anti-inflammatory, anti-ageing, anti-wrinkle and skin protection effects [158,218,219,220]
no side effects observed [220]
Glycyrrhiza glabrasaponins (glycyrrhizin), flavonoids (liquiritigenin, liquiritin), glycosides, isoflavons (glabridin, coumarins, stilbenoids) [221,222]anti-inflammatory effect, antioxidant capacity, photo-protection effect
reduces erythema and hyperpigmentation
improves epidermal hydration and transepidermal water loss
antibacterial and antifungal effect
anti-dandruff effect [221,222,223,224]
eye sensitivity (burning, itch, redness), scalp pruritus (itch, dandruff) [221]
Glycine maxphenolic acids (ferulic, syringic, sinapic, flavonoids (isoflavons), soy proteins (β-conglycinin, glycinin), lipids, vitamin E [225,226,227]anti-inflammatory effect, antioxidant capacity
skin lightening and depigmentant
protective effect against photo-ageing
stimulates collagen and elastin synthesis, increasing the hyaluronic acid levels in aged skin
skin regeneration effect, providing nutrients for cell renewal [225,226,227,228,229,230,231]
none allergic reactions to human skin toxicity evaluation [226,232]
itching eczema after several months of exposure to a soy-based ingredients in cosmetic lotions [233]
Hedera helixsaponins (hederin, hederagenin, hederacosides, hederacolchisides etc.), flavonoids, anthocyanins, coumarins and phenolic acids, steroids, vitamins, volatile and fixed oils [234]antiseptic, anti-elastase and anti-hyaluronidase effect indicated in cellulitis, cicatrisation, and wound healing [158,234,235]contact dermatitis [234]
Lilium candidumsaponins, flavonoids, glycosides, nitrogenous compounds [236,237]amelioration of skin redness, burn healing, hyperpigmentation, edema/skin inflammation
anti-irritant, anti-inflammatory, antioxidant effect, and also emollient and sebostatic effect
capacity of reducing ecchymosis, providing keratinocytes proliferation [158,236,238,239]
no irritations in vitro dermal or ocular irritation model [240]
Magnolia lilifloraterpenes, flavonoids, chlorogenic acid, rutin, quercetin, kaempferol, volatile oil (phenyethyl alcohol, levoxine, pinene, caryophyllene oxide, caryophyllene, bourbonene, farnesene, aerpineol, humulene) [241,242,243]antioxidant capacity, anti-dermatophytes action, anti-inflammatory effect [241,242,243]* N
Malus domestica/sylvestrispolyphenols (phloridizin, phloretin, quercetin, glycosides, rutin), acids (citric, malic, lactic, pyruvic, ascorbic etc.) [244,245,246]confers cell protection, anti-ageing and photoprotective effect, antioxidant capacity
increases moisture, reduces tyrosinase activity (depigmentation effect) [245,246,247,248]
absence of skin irritation of a cosmetic product containing 6% Malus sylvestris fruit extract under 24 h occlusive patch testing [245]
Malva sylvestrismucilaginous polysaccharides, tannins, polyphenols, vitamin C, vitamin E, β-carotene, anthocyanidines, terpenes, naphthoquinones, flavonoids, fatty acids (e.g., α-linolenic acid) and minerals [158,249,250,251,252,253,254,255]anti-pruritus, anti-psoriatic and antiseptic effect emollient, anti-inflammatory and antimicrobial capacity
wound healing (stimulates the formation of free granulation tissue and reepithelization) [158,249,250,251,252,253,255]
low toxicity of Malva sylvestris hydro-alcoholic extract on topical application [256]
Nymphaea albapolyphenols, flavonoids, essential oil, vitamin E, ellagitannins, sterols, fatty acids [257,258,259]stimulates the autophagy (reduces the accumulation of advanced glycation end products which determine inflammatory response, destroys the protein structureand colors the skin in yellow or brown)
antioxidant and photo-protection capacity
reduces the hyperpigmentation [258,260]
* N
Opuntia cochenilliferaflavonoids, terpenes, cellulose, hemicellulose, lignins, pectines, malic acid, citric acid, ascorbic acid, oligosaccharides (fructose, glucose, sucrose, maltose etc.), amino-acids, fatty-acids [261,262,263,264]antioxidant capacity, immunomodulation, hydration and cooling effects, wound healing [262,263]* N
Orchid sp.flavonoids, anthocyanins, fatty acids, organic acids, sterols, stilbenoids (resveratrol), amino-acids [265,266,267]anti-inflammatory effect, antioxidant capacity, anti-tyrosinase activity, inhibits collagenase and elastase photo-protection, wound healing capacity
cooling agent
astringent, anti-ageing, emollient, skin moisturizing and whitening effect [265,266,268,269]
allergic contact dermatitis [270]
Pfaffia paniculatasaponins (pfaffosides), pfaffic acid, sterols, allantoin [236,271]anti-inflammatory and anti-cellulite effect,
antioxidant capacity, protective effect against photo-ageing [236,271]
* N
Picea abiesditerpene, stilbene glycosides (E-astringin, E-isorhapontin, flavonoids (catechin, taxifolin, taxifolin-3′-O-glucoside), lignin (pinoresinol) phenolic acid derivates, saccharose [272,273]antioxidant capacity, antibacterial activity
inhibits collagenase (anti-ageing activity), elastase (anti-ageing, anti-inflammatory and anti-psoriatic effect) and tyrosinase (whitening activity)
wound healing and skin regeneration properties (cellular regeneration and epithelialization) [272,273,274,275,276]
possible allergic skin reactions (e.g., contact dermatitis) [276]
Porphyridium cruentumexopolysaccharide, phycoerythrins, phycoerythrocyanins, phycocyanins, sterols, superoxide dismutase, polyunsaturated fatty acids [277,278,279]antioxidant capacity, anti-inflammatory, anti-redness effect
restores skin barrier permeability, photo-protection, regenerative and anti-wrinkles effect
natural dye for cosmetics (creams, powders, lipsticks, make-up, eye shadows) [278,279,280,281,282]
* N
Ptychopetalum olacoidesfatty acids, saponins, flavonoids (rutin), sterols, aromatic oil, alkaloids, lupeol [236,283]antioxidant capacity,
anti-cellulite effect [236]
* N
Punica granatumanthocyanins, ellagic acid, tannins [176]improves viscoelasticity,
anti-ageing effect,
anti-inflammatory and antioxidant capacity
antimicrobial activity [176,284,285]
no adverse effects [286]
Rosa sp.
(R. damascena,
R. canina,
R. centifolia,
R. davurica,
R. mosqueta,
R. rugosa)
carotenoids, sterols, anthocyanins, catechins, polyphenolic compounds (tocopherol), vitamin C, fatty acids (trans-retinoic acid, arachidonic acid, and unsaturated fatty acids, such as oleic, linoleic and linolenic acids) [287,288]anti-inflammatory effect (suppressing proinflammatory cytokines) [289,290,291,292]
antibacterial against skin bacteria (Propionibacterium acnes) [289,290,291,292,293]
antioxidant capacity [290,292,294]
antiproliferative and chemopreventive effects [290]
fragrance in the perfumery industry [295]
well tolerate [296]
no toxicity on human keratinocyte HaCaT cells [289]
contact dermatitis to Rosa mosqueta oil applied for 3 days [288]
Rosmarinus officinalistriterpes (rosmarinic acid, oleanolic acid, ursolic acid) [172,297]antioxidant capacity, photoprotective effect,
anti-aging, wound healing, anti-inflammatory, increases skin elasticity, wrinkles reducing effect [157,297,298]
eczema, contact dermatitis [172,299]
Rubus chamaemorusellagitannins, proanthocyanidins, fenolic acids, flavonoids, lignans, vitamin C, fatty acids (oleic, linoleic, linolenic etc.) [300]antioxidant capacity
antimicrobial effect (Staphylococcus aureus skin infections)
reduces periorbital inflammations and ameliorates pruritus [300,301]
no irritation after an 48 h occlusive patch testin of a cosmetic product incorporating 2.5% Rubus chamaemorus Seed Oil (25 μL testing product quantity) [302]
Saccharum officinarumfatty acids, fatty alcohols, acids, esters, aldehydes, ketones, sterols, terpoides, phenolic acids, flavonoids (flavones glycosides) [303,304,305]antioxidant capacity, anti-tyrosinase activity
photo-protection effect [304,305]
low toxicity after in vitro cytotoxicity assay [304]
Sambucus nigraphenolic acids (caffeic, chlorogenic, ferulic, rosmarinic), flavonoids (quercetin, rutin, kaempferol), anthocyanins, tannins, triterpenes, organic acids [306,307,308]antioxidant capacity, reduces tyrosinase activity antiseptic, anti-inflammatory, anti-psoriatic, photoprotective effects [306,307,308,309,310,311,312]* N
Sideritis sp.
(S. perfoliata,
S. raeseri)
terpenes, phenolic compounds (flavonoids, phenylpropanid glycosides), tannins [313,314,315,316,317]antioxidant capacity, anti-elastase, anti-melanin and anti-tyrosinase activity (anti-wrinkle and depigmentation effect)
anti-inflammatory and antibacterial capacity
photo-protection effect [313,314,316,317]
* N
Sophora japonicaflavonoids (rutin, quercetin etc.), tripterpenoids, alkaloids, saponins, polysaccharides [318,319,320]antioxidant capacity, photo-protection, anti-melanin and anti-tyrosinase activity (skin-whitening) [318,319,320,321]* N
Triticum aestivumoligosaccharides [322]stimulates hyaluronan synthase, fibronectin synthesys, restores the skin barrier integrity (wound healing: decubitus ulcers, skin lesions and burns), anti-inflammatory effect, antioxidant capacity [322,323,324,325]possible hypersensitivity reactions [326]
Ulmus davidianasaccharides (galactose, rhamnose, glucose), succinic acid, terpenoids (epifriedenalol), lignans, flavonoids (catechin) [327,328,329]moisturizing, photoprotective, anti-ageing, anti-allergic and anti-inflammatory effect [327,328]* N
Vaccinium myrtillusanthocyanins (cyanidin, delphinidin, malvidin, peonidin, petunidin), flavonoids, phenolic acids [330]antioxidant capacity, photoprotective and anti-inflammatory effect (anti-psoriatic, anti-erythematous),
astringent properties,
wound healing, restores and protects the skin barrier function [330,331,332,333,334,335]
* N
Vitis viniferapolyphenols (resveratrol, anthocyanins), coumarn, carotenoids, flavonoids, tartaric acid, tannins, terpene alcohols [336,337,338]anti-ageing effect,
antioxidant capacity,
anti-melanogenic activities,
sunscreen protection [157,176,338,339]
allergic dermatitis [336,340]
Voandzeia subterraneanalkaloids, flavonoids, glycosides, saponins, steroids, triterpenoids, phenols, anthocyanins, carotenoids
proteins, crude fiber, carbohydrates [341,342]
anti-wrinkle effects, photoprotective, anti-irritant, anti-pollution, hydrating effects,
antioxidant capacity,
anti-ageing activity, inhibits proteases, elastases, collagenases and catalase [343]
* N
* N—No article reported.
Table 3. Benefits, cosmetic claim, and toxicity of some active ingredients used in cosmetics together with hyaluronic acid and HA derivates.
Table 3. Benefits, cosmetic claim, and toxicity of some active ingredients used in cosmetics together with hyaluronic acid and HA derivates.
Active IngredientCosmetic Claim and Skin BenefitsToxicity of Topical Products
Probioticsprevention and improvement of skin conditions (e.g., external signs of ageing, acne, rosacea, yeast and bacterial infections, psoriasis, dermatitis) [344,345,346,347,348,349,350]no side effects [344,350]
Bifida Ferment Lysateimproves the skin’s epidermal renewal process [351]
diminishes clinical signs and symptoms of atopic eczema [352,353,354]
regulates skin reactivity and dryness [355]
non-sensitizing [356]
Lactococcus Ferment Lysatestimulates the skin’s self-renewal [357]
significant improvement of atopic dermatitis (AD) [354]
non-irritant, no allergenic potential [358]
Pseudoalteromonas Ferment Extractreduces skin shininess, pores, spots, and sebum [359,360]non-sensitizing, non-irritating [361]
Amino acids,
Peptides,
Proteins
common ingredients in cosmetics, as they function as natural moisturizing factors which act as water-binding molecules
stimulating collagen and elastin synthesis in skin and improving surface healing
anti-ageing effect,
increase hydration of the skin which helps to reduce wrinkles and improves the functions of the skin barrier [362]
safe in cosmetics [362]
Alaninenatural moisturizing factor which acts as a water binding molecule [362]safe for cosmetic use
no dermal irritation or sensitization [363]
Argininemoisturizing effect
increases skin hydration and alleviates the condition of skin dryness [364]
no dermal irritation or sensitization [363]
Isoleucinean essential amino acid known for its ability to support tissue repair
effective in the treatment of mild to moderate chronic lesion of atopic eczema of the face [365]
safe for cosmetic use [363]
Acetyl Hexapeptide-8cosmetic application for wrinkles, working by relaxing of superficial dermal muscles [366]
decreases hyperkinetic facial lines or expression wrinkles, effective in reducing skin roughness [367]
neurotransmitter inhibitor peptide, used in anti-wrinkle formulations applied to the periorbital area [368]
well tolerated, with lack of burning and discomfort to the patient
safe for topical application [369]
Acetylarginyltryptophyl Diphenylglycineenhances skin elasticity and tightness,
increases type I collagen synthesis [370]
* N
Pentapeptide-18neurotransmitter-inhibiting peptide that decreases neuronal activity and catecholamine release, giving it Botox-like effects in reducing fine lines and wrinkles, and improving skin firmness [371]safe for cosmetic use [372]
Dipeptide Diaminobutyroyl Benzylamide Diacetatea small tripetide with anti-ageing and mattifying effect,
stimulates PLOD 3 (procollagen-lysine, oxoglutarate 5-dioxygenase 3), enzyme which is known to be of importance for the intermolecular collagen crosslink stability, increasing the stability of collagen, especially in mature skin [373]
* N
Palmitoyl Tripeptideimprovement of facial wrinkles, elasticity, dermal density and skin tone [374]safe in cosmetic products [375]
Vitaminsuseful role in the treatment of skin ageing and protection of human skin against UV induced ageing [376]
Ascorbyl Tetraisopalmitateantioxidant and anti-inflammatory properties,
increases skin hydration and smoothness [377]
contact dermatitis [378,379]
Niacinamideantipruritic, antimicrobial, vasoactive, photoprotective, sebostatic capacity,
lightening effect depending on its concentration [380]
safe for cosmetic use [381]
Panthenolhydrating and softening potential,
significantly accelerating epidermal regeneration [382]
allergic contact dermatitis [383,384]
Retinoidsantioxidant capacity, anti-wrinkle, depigmentation, anti-roughness, keratolytic effects, improve skin texture and laxity, protective effect against photo-ageing [385,386]photosensitization, irritation, erythema, dermatitis, pruritus, burning sensation [386]
Tocopherol
Tocotrienols
antioxidant capacity, anti-wrinkle effect, improving skin moisture [157,385,387]erythema, oedema, local irritation [387]
Saccharides
Trehalosemoisture retaining agent in several cosmetic creams and lotions [388,389]safe for use in cosmetic products [388]
Bee products (Honey, Propolis)
Honey Extract
Propolis Extract
antimicrobial and immunomodulatory effect, antioxidant capacity, wound healing (stimulates angiogenesis, granulation, epithelialization, TNF-α (tumor necrosis factor-α) production, increasing collagen and fibroblasts synthesis),
emollient, humectant, moisturizing, nourishing and protective effects against photo-ageing,
restoration capacity of skin barrier function [390,391,392,393,394,395,396,397]
contact dermatitis, acute sensitization [391,396,397]
Other Chemical Compounds
Allantoin
(5-ureidohydantoin)
anti-inflammatory effect and anti-irritation, wound healing and keratolytic properties,
hydration effect, tissue regeneration and cell proliferation capacity [398,399]
no adverse effects [399]
Ceramide 3increases SC hydration, enhances the barrier function and reduces inflammation [400,401]safe for use in cosmetic products [402]
Goldanti-inflammatory and antioxidant effect,
tissue regeneration capacity,
restores skin elasticity, reduces signs of stress and ageing [403,404,405]
particles larger than 30 nm can be considered as safe but there is a need to comprehensively study the effects of gold nanoparticles on the basis of their size distribution for their safe application in cosmetics [404]
Lactic acidprimarily used as moisturizer and pH regulator in cosmetics,
antimicrobial activity,
skin lightening effect,
keratolytic properties, possesses high water-binding capacity,
antibacterial effect [406,407]
peeling agent used in the amelioration of acne vulgaris and in the treatment of melasma [408]
good skin compatibility, showing only minor reactions [407]
Lecithinantioxidant effect,
dispersing agent for pigments [409]
safe as used in rinse-off products
safe for use in leave-on products at concentrations of 15%
insufficient data to determine the safety for use in cosmetic products where Lecithin or Hydrogenated Lecithin are likely to be inhaled [409]
Malachite Extracta copper complex extracted from the malachite stone, being a powerful free radical scavenger, boosting cellular defenses, offering protection and detoxifying benefits [410]* N
Sodium Pyrrolidone Carboxylateskin moisturizing effect [411]safe in cosmetics
non-irritating in a reconstructed human epidermis model test using the EpiSkin model [412]
Superoxide Dismutase (SOD)reduces UV-induced erythema, free radical scavenger, anti-irritant effect,
anti-ageing capacity [413]
non-irritating and non-sensitizing [413]
Ureahumectant, decreases TEWL in normal skin and especially in xerotic skin disorders (AD patients) [414]
moisturizing effect, desquamating actions (urea dissolves the intercellular cementing substance in the SC),
antimicrobial action [415,416,417]
safe as used in cosmetic products [415].
* N—No article reported. We used background color for cosmetic ingredients classes which represent examples of ingredients from the class presented above.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Juncan, A.M.; Moisă, D.G.; Santini, A.; Morgovan, C.; Rus, L.-L.; Vonica-Țincu, A.L.; Loghin, F. Advantages of Hyaluronic Acid and Its Combination with Other Bioactive Ingredients in Cosmeceuticals. Molecules 2021, 26, 4429. https://doi.org/10.3390/molecules26154429

AMA Style

Juncan AM, Moisă DG, Santini A, Morgovan C, Rus L-L, Vonica-Țincu AL, Loghin F. Advantages of Hyaluronic Acid and Its Combination with Other Bioactive Ingredients in Cosmeceuticals. Molecules. 2021; 26(15):4429. https://doi.org/10.3390/molecules26154429

Chicago/Turabian Style

Juncan, Anca Maria, Dana Georgiana Moisă, Antonello Santini, Claudiu Morgovan, Luca-Liviu Rus, Andreea Loredana Vonica-Țincu, and Felicia Loghin. 2021. "Advantages of Hyaluronic Acid and Its Combination with Other Bioactive Ingredients in Cosmeceuticals" Molecules 26, no. 15: 4429. https://doi.org/10.3390/molecules26154429

Article Metrics

Back to TopTop