Next Article in Journal
Melatonin and Pathological Cell Interactions: Mitochondrial Glucose Processing in Cancer Cells
Next Article in Special Issue
Unraveling the Role of Sex Hormones on Keratinocyte Functions in Human Inflammatory Skin Diseases
Previous Article in Journal
HLA-G Is Widely Expressed by Mast Cells in Regions of Organ Fibrosis in the Liver, Lung and Kidney
Previous Article in Special Issue
Keratins as an Inflammation Trigger Point in Epidermolysis Bullosa Simplex
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Modulatory Influence of Plant-Derived Compounds on Human Keratinocyte Function

1
Department of Computer Science in Economics, University of Lodz, 90-214 Lodz, Poland
2
Department of Biology and Pharmaceutical Botany, Medical University of Lodz, 90-151 Lodz, Poland
3
Department of Medical Informatics and Statistics, Medical University of Lodz, 90-645 Lodz, Poland
4
Chair of Gerontology, Geriatrics and Social Work at the Faculty of Pedagogy, Ignatianum Academy in Cracow, 31-501 Cracow, Poland
5
Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(22), 12488; https://doi.org/10.3390/ijms222212488
Submission received: 5 November 2021 / Revised: 16 November 2021 / Accepted: 16 November 2021 / Published: 19 November 2021
(This article belongs to the Special Issue Functional Defects of Keratinocytes in Inflammatory Skin Diseases)

Abstract

:
The plant kingdom is a rich source of secondary metabolites with numerous properties, including the potential to modify keratinocyte biology. Keratinocytes are important epithelial cells that play a protective role against various chemical, physical and biological stimuli, and participate in reactive oxygen scavenging and inflammation and wound healing processes. The epidermal cell response may be modulated by phytochemicals via changes in signal transduction pathways. Plant extracts and single secondary compounds can possess a high antioxidant capacity and may suppress reactive oxygen species release, inhibit pro-apoptotic proteins and apoptosis and activate antioxidant enzymes in keratinocytes. Moreover, selected plant extracts and single compounds also exhibit anti-inflammatory properties and exposure may result in limited production of adhesion molecules, pro-inflammatory cytokines and chemokines in keratinocytes. In addition, plant extracts and single compounds may promote keratinocyte motility and proliferation via the regulation of growth factor production and enhance wound healing. While such plant compounds may modulate keratinocyte functions, further in vitro and in vivo studies are needed on their mechanisms of action, and more specific toxicity and clinical studies are needed to ensure their effectiveness and safety for use on human skin.

1. Introduction

Plants produce a range of secondary metabolites. These not only play a crucial role in the adaptation of plants to the environment but also have a strong impact on other living organisms, including humans. Various groups of molecules with a plant origin such as phenolics, alkaloids, saponins, terpenes, lipids and carbohydrates, are involved in a plethora of biological activities. Selected phytochemicals may modulate several cell-signaling pathways in different cell types, including those of epidermal cells [1].
The skin creates a barrier protecting the host from the outside environment. The outer layer of the human skin, the epidermis, is formed by keratinocytes. These cells are on the first line of defense against various harmful chemical, physical and biological factors and are particularly vulnerable to ultraviolet (UV)-radiation, a strong source of reactive oxygen species (ROS). In response, keratinocytes produce a range of biologically active molecules that enhance ROS scavenging, inflammatory responses and wound healing [2].
Numerous plant extracts, as well as their component compounds, exert antioxidant activity. Phenolic compounds are particularly effective ROS scavengers. These chemicals are able to neutralize ROS via their ability to donate hydrogen atoms or electrons to radicals and chelate metal cations. Selected plant extracts and their component compounds have been found to suppress ROS release, decrease the activity of inducible nitric oxide synthase and nitric oxide, inhibit pro-apoptotic proteins and apoptosis and activate antioxidant enzymes in keratinocytes following exposure to hydrogen peroxide (H2O2) [3]. A wide range of secondary metabolites is also known to play an anti-inflammatory role. Following stimulation with pro-inflammatory factors, keratinocytes produce numerous molecules including adhesion molecules, interleukins and chemokines, with this production being suppressed by exposure to plant extracts and compounds [4]. Moreover, keratinocytes treated with UV radiation are exposed to the action of both ROS and pro-inflammatory factors; indeed, selected plant extracts and single compounds may play a dual, antioxidant and anti-inflammatory role in epidermal cells exposed to UV radiation [5,6]. In addition, plant extracts and single compounds may also modulate the release of various growth factors, chemokines or neuropeptides by keratinocytes, with exposure to molecules of plant origin possibly enhancing keratinocyte migratory and proliferation rates. This effect also accelerates the wound healing process [7].
The aim of the present paper is to review the role of selected plant extracts and single compounds as modulators of keratinocyte biology, paying particular attention to stimuli, specific receptors, protein release and signal transduction pathways. It examines the potential of phytochemicals as ROS scavengers, important anti-inflammatory agents and as factors for mediating wound healing. The role, contributions and usefulness of plants were summarized.

2. Criteria for Paper Selection

To give an overview of the current state of knowledge regarding the topic, i.e., in vitro treatment of human keratinocytes with oxidants, pro-inflammatory agents and UV radiation followed by exposure to different plant extracts, papers were selected from those included in the electronic databases PubMed/MEDLINE, Google Scholar, Scopus and Web of Science over the past 10 years. The following search terms were used: keratinocytes, plant extract, plant-derived compounds, reactive oxygen species, inflammation, UV-irradiation and wound healing. Moreover, the impact of plant secondary compounds on keratinocyte motility and proliferation rate was evaluated. Papers reporting articles published in languages other than English, those with only an abstract or lacking full-text access, those published earlier than 10 years ago, those without identified compounds in extracts and those examining cells other than human keratinocytes were excluded. Each selected document was analyzed and the following data were extracted and presented in tables: species name, plant part, type of extract, type of cell lines, identified compounds, mechanism of action and final effect. The main text includes a characterization of the signaling cascades involved in keratinocyte biology.

3. Plant Secondary Metabolites

The plant kingdom is a rich source of secondary metabolites. Over 50,000 molecules have been discovered [8]. These metabolites can be divided into the following structural classes: phenolics, alkaloids, saponins, terpenes, lipids and carbohydrates [9]. These compounds play an essential role for the plant itself by allowing adaptation to the environment. Phytochemicals contribute to perennial growth, deciduous behavior, flowering, fruit set and abscission, and are known to demonstrate antimicrobial properties [8]. Many of these compounds also exhibit antioxidant [10], anti-inflammatory [11], anticancer [12], including against keratinocyte carcinomas [13], antipathogenic [14,15] or antiobesity [16] properties; as such, they are widely used in various industrial sectors, including pharmaceutical, cosmetic or food [17]. In addition, there is a strong trend towards using herbal formulations in health and wellness [18].
Secondary metabolites may be obtained from the leaves, roots, stems, bark and aerial parts of the plants; however, the plants require careful preparation, extraction and quantitative and qualitative determination of active compounds. The first step involves the selection of an appropriate solvent. The most popular are polar ones such as water and alcohols, intermediate polar ones such as acetone and dichloromethane, and nonpolar ones such as chloroform, n-hexane and ether. Following this, an appropriate extraction method must be identified. Various options exist including maceration, decoction, digestion, percolation, infusion, Soxhlet extraction, superficial extraction, microwave-assisted extractions or ultrasound-assisted extraction. The isolated compounds must then be purified via chromatographic methods and identified via spectroscopy [19]. However, the greatest challenges are those posed by the determination of the molecular mechanism of action and the later conduct of clinical trials where necessary [20].

4. Keratinocyte Characteristics

The skin is the largest organ in the human body. In addition to serving as a barrier for the inner environment, it also regulates body temperature, enhances metabolic functions and enables contact with the outer environment by its host of nerve endings.
Human skin consists of three layers: an outer stratified epithelium, a middle dermis and an underlying subcutaneous tissue. The epidermis is formed mostly (at least 80%) by keratinocytes. Newly-synthesized keratinocytes build up a supply of keratin in the cytoplasm and then undergo structural and biochemical changes until terminal differentiation and cell death. Keratinocytes play numerous roles including the creation of a physical barrier. That barrier provides protection against water loss, biological agents such as pathogens, physical agents such as UV radiation and various chemical agents. Moreover, epithelial cells accumulate melanin derived from melanocytes, migrate and proliferate to heal wounds when skin integrity is disrupted and participate in skin immunity. In addition, keratinocytes are involved in molecular interactions between various surrounding skin cells via different signal transduction pathways [21,22].
During normal epidermal differentiation, the following types of keratinocytes are distinguished: basal, spinous, granular and cornified. These types vary in their phenotypic and biochemical properties based on changes in gene expression. Many of these genes are regulated by p63 transcription factors, including keratins, involucrin and loricrin [23]. Basal cells that are in contact with the basement membrane through hemidesmosomes and focal adhesions possess mitotic activity and express keratins 5 (K5) and K14. During the progress of differentiation, keratinocytes demonstrate basal membrane detachment and loss of mitotic activity and migrate into suprabasal layers. These suprabasal cells attach to their neighboring cells through desmosomes, an attachment that must be broken during the progress of epithelization.
Spinous keratinocytes begin to express keratins K1 and K10, and this expression continues fully in the granular forms. They also express involucrin and loricrin, the precursors of cornified envelope proteins. These play a role in corneocyte formation, the final stage of differentiation preceded by loss of nuclei and organelles. The corneocytes are connected by corneodesmosomes and covered with lipid layers secured by a protein structure. These form a barrier to prevent water evaporation [24].
The differentiation process of keratinocytes is regulated by the mitogen-activated protein kinase (MAPK) pathway, which is induced by various stimuli including epidermal growth factor (EGF), tumor necrosis factor (TNF) and calcium influx [25]. The signaling transition pathway uses different isoforms of protein kinase C (PKC) [26]. In normal human epidermal keratinocytes (NHEKs), the inhibition of the MAPK pathway resulted in the decreased expression of proteins directly implicated in the differentiation process. For example, the suppression of p38 signaling resulted in the inhibition of K5, K14, ST14 transmembrane serine protease matriptase (ST14), small proline-rich protein 3 (SPRR3), serine/threonine kinase (Akt) expression and suppressed the nuclear factor of kappa in B cells (NF-κB) light polypeptide gene enhancer; suppression of c-Jun N-terminal kinase (JNK) signaling resulted in the inhibition of K14, SPRR3, Akt expression and suppression of NF-κB. In addition, the suppression of extracellular signal-regulated kinase 1/2 (ERK1/2) signaling resulted in the inhibition of SPRR3 and Akt expression [27].
Being located directly on the edge of the internal and external environment, epidermal keratinocytes are exposed to mechanical stress. Stretching the skin results in hyperproliferation via the induction of calcium influx followed by phosphorylation of essential growth-related factors, including the epidermal growth factor receptor (EGFR) [28]. Moreover, keratinocytes can also become activated in response to exposure to ROS, inflammatory factors, UV-radiation and skin barrier disruption. Such cells generally express various surface receptors which translate the stimulus into biological effects [29]. Ligand/receptor binding induces signal transduction pathways and changes in protein phosphorylation [30]. Plant-derived compounds are believed to modulate various signaling pathways in various cell lines, including keratinocytes, some of which include signaling cascades related to proliferation and apoptosis [31].

5. Modulatory Effect of Plant Secondary Metabolites on Keratinocytes Exposed to ROS

UV radiation is the main source of ROS in keratinocytes, with heat shock and drugs being others. Oxidative stress may be involved in the inflammatory condition and apoptosis of the skin. In addition, ROS may modulate various signaling cascades in human cells in vitro. ROS have also been found to mediate MAPK activities, and an oxidant state has been associated with an elevated level of activator protein 1 (AP-1) and NFκB in mouse keratinocytes [32]. However, there is a need to better identify the role of ROS in the signal transduction pathway using antioxidants including plant-derived compounds [33].
Many plants’ secondary metabolites, especially phenolic compounds, act as antioxidants. This has been attributed to their ability to donate hydrogen atoms or electrons to radicals to form nonreactive phenoxyl radicals or stable radical cations, respectively [34]. They are also believed to chelate metal cations. The oxidized metabolites of phenolic compounds have also been found to up-regulate the expression of antioxidant enzymes, and are believed to bestow considerable beneficial biological effects related to their antioxidant activity, including anti-inflammatory, anti-aging and anti-cancer properties [35,36]. Table 1 presents the impact of the selected plant extracts on H2O2-stimulated human keratinocytes. Cellular exposure to H2O2 is strictly connected with ROS induction; therefore, the presented phytochemicals exhibit important antioxidant properties such as suppressing ROS release, limiting damage to DNA and lipid oxidation, inhibiting iNOS and NO, decreasing the level of pro-apoptotic proteins and apoptosis and activating antioxidant enzymes.

6. The Modulatory Effect of Plant Secondary Metabolites on Keratinocytes Involved in the Inflammation Process Triggered by Physical, Chemical or Biological Agents

Unperturbed keratinocytes do not release inflammatory mediators; however, their expression may be enhanced by various stimuli, including UV radiation, wounding, pathogens and pathogen products. ROS play a special role in skin inflammation and may trigger a chronic skin response. Following the activation, the upregulation of the adhesion molecule, cytokine and chemokine expression is observed [47].
Adhesion molecules that belong to the immunoglobin superfamily, including intercellular adhesion molecule-1 (ICAM-1) and vascular adhesion molecule-1 (VCAM-1) play an essential role in inflammatory mechanisms. ICAM-1 is a ligand for the B-2 integrin leukocyte function-associated antigen-1 (LFA-1). This molecule promotes cellular interaction in the immune response. ICAM-1 and LFA-1 interaction is essential for T lymphocytes activation. Elevated ICAM-1 expression is observed in keratinocytes after interferon-gamma (IFN-γ) exposure. Tumor necrosis factor-α (TNF-α) and IL-17 act synergistically with INF- γ in the induction of ICAM-1 expression. VCAM-1 interacts with various integrins [48].
Cytokine action is initiated by the activation of NFκB pathways by Toll-Like Receptors (TLRs), which recognize pathogen-associated molecular patterns [49]. It has been shown that keratinocytes express various TLRs including 1–6 and 9 [50,51], indicating that the induced TLRs stimulate cytokines [52,53]. TLR ligands and cytokines mostly have overlapping or synergistic influences on keratinocytes [49]. Keratinocytes produce a number of cytokines, including interleukin (IL)-1, -6, -7, -8, -10, -12, -15, -18 and -20, and TNF-α. Keratinocytes also express various cytokine receptors including IL-1R, IL-2R, IL-4R, IL-6R, IL-10R, IL-13R, IL-17R, IL-18R, IL-20R, IL-24R and TNF-R [54].
IL-1, -6, -8 and TNF-α are well-known pro-inflammatory cytokines. IL-1 acts as a chemoattractant for keratinocytes [55], activates expression of keratin 6 [56], decreases adherence of selected pathogens to keratinocytes [57] and protects transformed keratinocytes from tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis [58]. A study conducted on mice revealed that keratinocytes secrete high amounts of IL-1α, which is followed by a skin inflammatory response. IL-1α is essential for caspase-1 activation in an inflammasome-dependent manner. It is hypothesized that epidermal cells may play a critical role in skin immunity [59], and that IL-6 regulates normal keratinocyte growth [60]. However, a study of injury-induced keratinocyte stimulation indicated that IL-6 activity may regulate keratinocyte differentiation rather than proliferation [61]. The data indicate that IL-6 is involved in the restoration of the epidermal barrier after wounding [62]. Another cytokine, IL-8, is released by external stimuli, including UV radiation [63] and is believed to stimulate keratinocyte migration [64].
TNFα plays a differential role in epidermal keratinocytes. On the one hand, TNFα activates modulators of the actin cytoskeleton and integrins, regulates their degree of motility or attachment and stimulates the expression of collagen-degrading proteases and basement membrane components, followed by tissue repair. On the other hand, TNFα stimulates the nuclear translocation of NFκB. It is also believed to induce the expression of various genes, including those of cytokines, chemokines, growth factors and cell-surface receptors that may attract neutrophils, macrophages and T lymphocytes. Additionally, genes regulated by TNFα are involved in the modulation of the cell cycle and apoptosis [65].
IL-7 and IL-15 are involved in T cell homeostasis. IL-7 participates in the development of mature T lymphocytes in the thymus. In addition, IL-7 also induces the expression of the anti-apoptotic protein Bcl-2, followed by the survival of both naïve and memory T lymphocytes. IL-15 stimulates the proliferation of CD8+ T cells in a process independent of antigens. While both cytokines may help to defend organisms against pathogens and tumors, elevated production may be linked to several autoimmune diseases [66]. Research based on mouse keratinocytes indicates that elevated IL-7 levels can predispose to lymphoproliferative skin disease [67]. IL-7 and IL-15 production is reduced by UVB radiation [68,69].
Cytokines IL-10, IL-12 and IL-18 act as immunomodulators. IL-10 stimulates the Th2 immune response [70] and its level in keratinocytes is upregulated after exposure to UVB. Therefore, it has been hypothesized that they may play a role in immunosuppression [71]. IL-12 stimulates the Th1 immune response [72]. IL-12 is also believed to take part in limiting skin inflammation [73] and blocking the release of TNFα induced by UVB [74]. IL-18 stimulates the Th1 immune responses in collaboration with IL-12. The Th1 response is related to the host defense against pathogens by the stimulation of IFN-γ production. Without IL-12 collaboration, IL-18 induces the Th2 response [75].
IL-20 induces keratinocyte hyperproliferation and therefore may act as a modulator of skin inflammation [76]. Its release by keratinocytes may be followed by the production of various chemokines [49]. The chemokine structure is maintained by disulfide bonds formed between cysteine residues. The monomer is composed of an α-helix in the C terminus, a three-stranded β-sheet in the central region and an unstructured region in the N terminus. The chemokines can be divided into four groups based on the configuration of two cysteines closer to the N-termini: CC, CXC, CX3C and XC. Following stimulation with IFN-α, IFN-γ, IL-13, IL-17, TNF-α or IL-4, keratinocytes release a plethora of different chemokines belonging to three different classes, including CC, CXC and CX3C [77].
Plant extracts and their component compounds may modulate the inflammatory keratinocyte response in vitro via different mechanisms. Keratinocytes exposed to TNF-α or IFN-γ exhibit an elevated production of pro-inflammatory cytokines. To counteract this, treatment with plant extracts may downregulate the expression of ICAM-1, IL-1, IL-6, IL-8, TNF-α and selected chemokines, and suppress the NFκB and MAPKs pathways. Table 2 presents the impact of selected plant extracts on human keratinocytes stimulated with TNF-α or IFN-γ. As cellular exposure to pro-inflammatory agents is closely connected with induction by various cytokines or chemokines, the presented plant-derived compounds are believed to exhibit important anti-inflammatory properties.
Ampelopsis glandulosa [83] extract was found to inhibit TNF-α, IFN-γ, IL-4, IL-13 and IL-31 expression in the ear tissue of mice in vivo. This observation was in response to exposure to the inflammation inducers 2, 4-dinitrochlorobenzene (DNCB).
Studies based on single compounds indicate that HaCaT cells stimulated with TNF-α/IFN-γ exhibit a decreased IL-6, IL-8, ICAM-1 and selected chemokine production, suppression of NFκB translocation to the nucleus and lowered p38 and ERK1/2 activity following treatment with selected compounds derived from Cudrania tricuspidate: dihydrokaempferol, steppogenin, cudraflavanone D, cudraflavanone B, cudraflavone C, kuwanon C, cudraxanthone L, macluraxanthone B, 1,6,7-trihydroxy-2-(1,1-dimethyl-2-propenyl)-3- methoxyxanthone, cudratricusxanthone L, cudracuspixanthone A [93]. Elsewhere HaCaT keratinocytes stimulated with LPS demonstrated reduced expression of chemokine MCP-1 following exposure to damsin isolated from Ambrosia arborescens [94]. In addition, human keratinocytes stimulated with TNF-α/IFN-γ exhibit downregulation of the signal transducer and activatorof transcription-1 (STAT-1), IL-33, ICAM-1 and chemokine TARC/CCL17 following treatment with isosecotanapartholide obtained from Artemisia princeps [95].

7. The Effect of Plant Secondary Metabolite Treatment on Keratinocytes Exposed to UV-Radiation

UV radiation is an environmental-damaging agent that causes oxidative stress, resulting in damage to cellular components and apoptosis. It is also known to induce inflammatory conditions that lead to skin disorders.
Keratinocytes are the first line of defense against UV radiation [96]. Exposure to UVB has been associated with changes in gene expression in epidermal cells [97]. A study of NHEKs found changes in expression in 249 of 539 studied genes four hours after exposure and in all 539 genes 24 h after exposure. The early cell response is mainly related to transcriptional arrest; however, the late cell response is more complex [98].
Following exposure to UV radiation (UVA, UVB and UVC mixed), keratinocytes prepared from newborn CD-1 mouse skin demonstrated EGFR receptor activation by phosphorylation of Tyr-992/Tyr-1045/Tyr-1068/Tyr-1173 residues via mechanisms associated with ROS. The EGFR receptor is strongly mitogenic and directly regulates the response of keratinocytes to UV radiation [99]. This activation may result in the induction of MAPK, phosphoinositide 3-kinase (PI3K)/Akt and NFκB signal transduction pathways [100]. These cascades regulate cell proliferation, differentiation and death [27,101,102]. It has been found that Egfr knockout mice are able to stay alive for only a few days after birth [99].
UVA and UVB exposure induces different groups of factors. UVB exposure induces the activation of c-Jun amino-terminal kinases (JNK), a member of the MAPKs in NHEKs [103], as well as JNK1 and JNK2 Th-183/Tyr-185 phosphorylation followed by the increased c-Fos expression [104]. It has also been found to induce transcription factor AP1 via the JNK pathway [105], and to induce Ser-473 phosphorylation of Akt [104] and Ser-727 phosphorylation of STAT3, known to play crucial roles in cell division, survival as well as migration [106]. UVA exposure of NHEKs cells induces Th-202/Tyr-424 phosphorylation of ERK1 and ERK2 and Th-180/Tyr-240 phosphorylation of p38, another member of the MAPKs family, followed by c-Jun overexpression. Moreover, mTOR is phosphorylated at Thr-2448 and p70S6k at Thr-421/Ser-424, both being downstream targets of Akt, and PI3K expression was found to be upregulated (p85), this being an upstream target of Akt. STAT 3 is phosphorylated at Tyr-705. In addition, nuclear translocation of NFκB proteins appears to be responsible for the induction of apoptosis [104].
NHEKs and immortalized human HaCaT keratinocytes also exhibit MAPKs signaling activation following exposure to both UVA and UVB radiation. The HaCaT cells demonstrate two-fold or even four-fold higher activity of p38 and JNK than NHEK, but the latter release more TNFα. The data also indicate that immortalized cells do not demonstrate NFκB pathway induction [107].
HaCaT keratinocytes exposed to both UVA and UVB radiation exhibit different gene expression profiles depending on a single or repetitive dose. Exposure to a single dose was associated with upregulation of the G/S checkpoint of the cell cycle, as well as NFκB, IL-1, death receptor and p38 signaling and downregulation of genes that regulate G2/M checkpoint of cell cycle and ATM serine/threonine kinase signaling. Additionally, numerous regulators including forkhead box M1 (FOXM1), forkhead box O1 (FOXO1), activating transcription factor 4 (ATF4), ATF6, melanocyte inducing transcription factor (MITF) and ETS homologous factor (EHF) were inhibited, while lysine demethylase 5B (KDM5B), SMAD family member 4 (SMAD4), tumor protein p63 (TP63), nuclear protein 1, transcriptional regulator (NUPR1), cyclin-dependent kinase inhibitor 2A (CDKN2A), NOTCH11 and STAT3 were activated. After repeated doses, interferon signaling was downregulated. Specific regulators such as FOXM1, TP73, NFE2 like bZIP transcription factor 2 (NFE2L2), MITF and EHF were inhibited, whereas STAT3, NUPR1, chromobox 5 (CBX5), ATF3, SMAD4 and MYC proto-oncogene, bHLH transcription factor (MYC), were activated [108]. In addition, HaCaT cells demonstrate strong activation of JNK but weak activation of ERKs following exposure to UVB [109].
Exposure to UV radiation also causes collagen destruction. The enzymes responsible for such effects are named matrix metalloproteinases (MMPs). In vivo data indicate that UV irradiation induces the expression of MMP-1, MMP-3 and MMP-9 mainly in the human epidermis [110]. Keratinocytes exposed to UVB exhibit accelerated MMP-1 expression, which may be mediated by the PKC-dependent induction of transient receptor potential vanilloid type 1 (TRPV1) followed by Ca2+-influx. This receptor is believed to be activated by various stimuli including capsaicin, heat or acids [111]. Moreover, keratinocytes treated with UVB exhibit elevated levels of MMP-1. These levels are driven by cascades dependent on ROS and ERK activation of the BLT2 receptor for leukotriene B4 (LTB4) and 12(S)-hydroxyeicosatetraenoic acid (12(S)-HETE) [112]. Additionally, UVB-irradiated keratinocytes may mediate a signal to the fibroblasts and enhance MMP-1 production [113]. The induction of MMP-9 and involucrin production were also observed on cultured keratinocytes after UVB treatment [114].
Human keratinocytes exposed to UV radiation result in ROS accumulation, as well as apoptosis by the intrinsic and extrinsic pathways [115]. Induction of programmed cell death is an important mechanism that may provide protection against skin cancer [116]. ROS generation and DNA damage induce p53 protein activation and cell cycle arrest followed by DNA repair or apoptosis [117]. TP53 knockout mice exhibit lower amounts of sunburn cells in the epidermis after exposure to UV radiation in comparison to wild-type mice [118]. In NHEKs, elevated TP53 expression after irradiation is related to phosphorylation of Ser-9/Ser15 followed by Apaf-1 and caspase-3 activation [119]. It also has been shown that p53 can interact with Bcl-2 and Bcl-xL proteins and regulate the intrinsic apoptosis pathway [120]. However, it has been found that combined UVB and heat treatment of NHEKs causes DNA damage but reduces the events of apoptosis in comparison to UVB alone, probably by the downregulation of the p-53-mediated response [121]. However, the process of apoptosis in basal keratinocytes exposed to UV radiation may also be independent of p-53 protein activity [115]. Studies suggest that the basal keratinocyte layer repairs DNA damage or undergoes apoptosis more rapidly than the suprabasal layer [119].
UV-induced HaCaT cells express the TNF-family receptor CD95 and may undergo apoptosis via the extrinsic pathway [122]. The data also show that UVB radiation induces caspase 8 in NHEKs and promotes the extrinsic pathway, as well as suppresses Bcl-2 and promotes the intrinsic pathway [123]. Similar results were obtained for HaCaT cells exposed to UVB radiation, in which both intrinsic and extrinsic apoptosis was induced [124]. In NHEKs irradiated with UVB, caspase activation occurs in the following order: caspase 3, caspase 9 then caspase 8 [125]. UVB irradiated HaCaT cells overexpressing Bcl-2 prevent ROS release and procaspase 3 and procaspase 8 activation [126]. The activation of caspases in UV-exposed NHEKs depends on protein kinase C (PKC). Caspase-3 and caspase-8 activation is prevented in blocked PKC [127].
Plant-derived compounds may counteract the detrimental effects of UV exposure on human skin. Data indicate that oil in water (O/W) emulsion containing 5% (w/w) Euterpe oleracea extract demonstrated a PF-UVA value (i.e., protection against UVA) of about 15 and a ratio of SPF (i.e., protection against UVB) to PF-UVA of about 1.5 [128]. Other extracts demonstrating great potential against UV radiation were obtained from Helichrysum arenarium, Sambucus nigra and Crataegus monogyna. All of them exhibit SPF values above 6 when used alone in O/W emulsion at 10% (wt.%), and above 16.5 and PF-UVA above 7.5 when used in combination [129]. Hylocereus polyrhizus at 1 mg/mL demonstrated an SPF of about 35 [130]. Pterodon emarginatus extract at 10% (w/w) in three different formulations (Lanette®, Polawax® and Focus Gel®) exhibited an SPF value of about 8 [131]. Furthermore, 1% (w/w) extract of Olea europaea standardized to 20% oleuropein exhibited an SPF value of about 21 used in the special formulation [132]. A methanolic extract of Washingtonia filifera exhibits an SPF value of about 3.4 [133]. A phenolic compound-enriched fraction of Salicornia ramosissima extract at 10% (w/w) was found to have both SPF and PF-UVA values of about 13.5 when applied as O/W emulsion [134], whereas a flavonoid-enriched fraction of the Vitis vinifera extract at a concentration of 250 μg/mL had an SPF of about 18.6 and a PF-UVA of about 3.2 [135].
The most important group of compounds that may be used as sunscreen agents are phenolic compounds. These molecules equipped with aromatic rings are able to absorb UVA and UVB radiation at wavelengths of 200–400 nm and scavenge ROS and modulate signaling processes. Two flavonoids, rutin and quercitin, provide SPF values of about 12 when used in 10% (w/w) O/W emulsion, whereas PF-UVA provides a value of about 14.5 [136]. Combining 0.1% (w/w) rutin with 6% benzophenone, a synthetic organic filter, increased the SPF value from about 24 to 33 [137]. Similarly, synergistic effects were observed between 0.1% rutin (w/w), 1% benzophenone (w/w) and 3.5% (w/w) ethylhexyl methoxycinnamate [138]. Another promising sunscreen agent is named silymarin: a 50 µmol/L ethanol solution of silymarin yielded an SPF value of about 5.5, and its main constituent silybin provided a value of 6.0. PF-UVA was about 1.5 for both [139].
Choquenet tested twelve phenolic compounds at various concentrations, including myricetin, luteolin, apigenin, puerarin, baicalin, baicalein, hesperidin, hesperetin, naringenin, diosmin, caffeic acid and chlorogenic acid. Of these, chlorogenic acid and apigenin were found to be the best UVB and UVA filters, with SPF values at about 10 and 7 and PF-UVA values at about 9 and 6, respectively [140]. Similarly, among fifteen tested phenolic compounds at 7% (w/v) concentrations (resveratrol, piceid, catechin, quercetin, kaempferol, galangin, apigenin, naringenin, chrysin, pinocembrin, coumaric acid, ferulic acid, caffeic acid, caffeic acid phenylethyl ester and dimethyl caffeic acid), the highest SPF value was observed for apigenin, i.e., about 29 [141].
The plant extracts and single compounds may also modulate the UV-radiated keratinocytes response via different mechanisms. Both may downregulate the ROS level and upregulate the antioxidative enzyme level in keratinocytes exposed to UV. Typically, the nuclear factor erythroid 2–related factor 2 (Nrf-2) pathway is induced, with NF-κB, p53, MAPK and Akt signaling being reduced. In addition, MMPs and prooxidative cytokine expression are suppressed. Table 3 presents the impact of selected plant extracts on UV-irradiated human keratinocytes. The presented phytochemicals clearly exhibit significant antioxidant and anti-inflammatory properties.
Mice treated orally with a red raspberry extract prior to exposure to UVB irradiation demonstrate less skin wrinkling, water loss and epidermal thickening in comparison to those that were not treated [143]. Rhus javanica extract suppresses COX-2 expression in mice exposed to UVB and has anti-wrinkle effects [151]. In addition, Rosa multiflora extract reduces TNF-α and MMP-13 production [154], and Paeonia × suffruticosa extract reduces MMP-1 production in mice exposed to UVB [160]. Moringa oleifera extract is found to protect against UVB-induced oxidative stress injuries in the epidermis of the mouse in vivo. PPARα induction is involved in the protective effect of the extract [39].
Studies have examined the protective roles of single compounds on epidermal cells, including 3,5-dicaffeoyl-epiquinic acid obtained from Atriplex gmelinii [167], quercetin 3,7-dimethyl ether 40-glucoside from Nymphoides indica [163], youngiasides A and C from Youngia denticulatum [146], ixerisoside A from Ixeris dentata [168], α-, β-, γ-mangostins and gartanin from Garcinia mangostana [169]. Of these, 3,5-dicaffeoyl-epiquinic acid downregulates the expression of MMP-1, -2 and -9, whereas α-, β-, γ-mangostins and gartanin downregulate the expression of MMP-1 and -9. In addition to these, youngiasides A and C also increased SOD1, Nrf2 and heme oxygenase-1 (HO-1) expression and downregulated MAPK and NF-κB pathways. Quercetin 3,7-dimethyl ether 40-glucoside also suppressed NF-κB as well as TNF-α, IL-1β, IL-6, IL-8, myelodysplastic syndromes (MDS) and thymus and activation-regulated chemokine (TARC). Ixerisoside A also blocked pro-inflammatory cytokines including IL-6 and IL-8, inhibited COX-2 expression and downregulated MAPKs. Keratinocytes exposed to UVA irradiation and then treated with prenylated phenols from Artocarpus communis exhibited reduced cell damage in contrast to untreated ones [170].

8. Modulatory Effect of Plant Secondary Metabolites on Keratinocytes Involved in the Wound Healing Process Triggered by Disruption of the Epidermal Barrier

Wound healing occurs as a consequence of skin barrier disruption. It is a complex process consisting of a series of phases, including hemostasis, inflammation, proliferation and remodeling [171]. The first phase is related to the formation of blood clots that prevent blood loss. The next phases are connected with the recruitment of numerous cells, including keratinocytes, to the wound site [172]. During inflammation, the debris is removed by neutrophils and macrophages. The proliferation phase is characterized by re-epithelialization and granulation performed by a mixture of stem cells, endothelial cells and keratinocytes. Both the inflammation and proliferation phases are related to new blood vessel formation. Finally, the wound healing is completed by the remodeling stage, in which the collagen matrix is restructured by fibroblasts [173].
Keratinocytes migrate, proliferate, and cross-talk with fibroblasts following wound contraction [174]. Activated keratinocytes are characterized by cytoskeleton changes, downregulation of K1 and K10 expression and upregulation of K6, K16 and K17 keratin expression, enabling them to migrate and restore the epidermal barrier [175,176]. The migratory rate is closely related with the disruption of adhesion between cells (desmosomes) as well as between cells and substratum (hemidesmosomes).
Desmosomes are structures that are essential for cell–cell adhesion and skin integrity. The core desmosome protein is named desmoplakin. Desmosomal adhesion in response to wounding is regulated by PKCα [177]. It has been suggested that the transcription factor Slug is related to effective re-epithelialization and desmosomal disruption [178]. In addition, retinitis pigmentosa GTPase regulator interacting protein 1-like (RPGRIP1L) is believed to be an epithelialization factor, and Rpgrip1l knockout mice exhibit impaired desmosomal structure [179]. Problems with desmosome function may also be associated with loss of desmoplakin [180] and flotillin expression [181], as well as cadherin disruption [182]. Desmoplakin is essential for cadherin cluster formation [183], which is crucial for desmosome formation, as well as intercellular adhesion. In turn, cluster formation is regulated by flotillins [184].
Hemidesmosomes are structures that link cells into the basal layer and are mediated by integrins [185]. Integrins are considered as regulators of growth factor receptor pathways, and their activity is believed to elevate growth factor activity [186]. One integrin expressed by keratinocytes, α6β4, binds to laminin-5. α6β4 knockout mice demonstrate an absence of hemidesmosomes [187]. The modulation of α6β4 binding affinities is involved in keratinocyte motility [188]. In cultured keratinocytes, PKC or PKA stimulate hemidesmosome turnover by integrin β4 subunit phosphorylation [189]. In turn, this phosphorylation is modulated by the presence of EGF and macrophage-stimulating proteins in the wound [190].
Numerous other regulators take part in the process of keratinocyte migration and proliferation, including various growth factors from the tyrosine kinase, insulin, FGF, vascular endothelial growth factor and scatter factor families [191]. In addition, cytokines, chemokines, MMPs and extracellular macromolecules also play various roles [192].
The epidermal growth factor family of receptor tyrosine kinases (HER) are also known to play a role in keratinocyte activity. Keratinocytes express 1, 2 and 3 HER receptors. Their ligand, including epidermal growth factor (EGF), heparin-binding EGF like growth factor (HB-EGF) and TGF-α stimulate keratinocyte migration [191] and proliferation [193]. EGF agonists have been found to completely prevent keratinocyte migration [194].
Moreover, insulin secreted by β-cells in the pancreas is able to circulate into the skin, where it can then bind to insulin receptors (IR) expressed in skin keratinocytes. Insulin mediates keratinocyte migration and proliferation. A combination of insulin and TGF-α was found to act synergistically in this regard. An in vivo study in a mouse model found that topical application of insulin results in accelerated wound healing [195]. A similar effect was observed in a diabetic mouse model [196].
Additionally, keratinocyte insulin-like growth factor 1 (IGF-1) receptor and its ligand, IGF-1, synthesized by fibroblasts or hepatocytes, has been found to stimulate the motogenic effect in keratinocytes. That signaling is downregulated in diabetic wounds. IGF-1 and EGF act synergistically; the first by induction of the PI3K pathway, the second by the MAPK/ERK pathway [197]. IGF-1 also acts synergistically with HB-EGF in stimulating keratinocyte proliferation [198].
Fibroblast growth factor receptor 1 (FGFR1) and FGFR2 have been shown to stimulate keratinocyte migration and proliferation following binding by several ligands, including fibroblast growth factor 1 (FGF-1), -2, -7, -10 and FGF-1 -10, respectively. FGFR1 and FGFR2 knockout keratinocytes have reduced the motility that may result from a lack of expression of focal adhesion components due to the absence of an FGFR signal transduction pathway [199]. FGF-7 is highly expressed at the beginning of the wound healing process. This factor probably acts by compensatory or overlapping mechanisms because inhibiting their expression does not directly influence the overall rate of the process. FGF-7 knockout diabetic mice exhibit delayed wound healing [200]. FGF-10 with dermatan sulfate synergistically increased keratinocyte migration in a wound [201].
Keratinocytes also express VEGFR-1, -2, and -3. Their ligand, VEGF-A, is produced by keratinocytes and macrophages. In addition to granulation tissue formation and angiogenesis, that factor also promotes keratinocyte migration [202].
Keratinocytes also express the MET receptor tyrosine kinase (RTK). Their ligand, hepatocyte growth factor (HGF), is produced by fibroblasts and by keratinocytes after skin injury. Throughout the stimulation of STAT3 signaling, HGF induces keratinocyte motility. In addition, HGF activates VEGF-A expression and influences cell proliferation. The RON RTK receptor expressed by keratinocytes is activated by a macrophage-stimulating protein (MSP) produced by hepatocytes. RON activates the PI3K/Akt pathway; it also induces phosphorylation of both RON and integrin α6β4 at 14-3-3 binding sites, allowing the formation of RON and α6β4 complex by 14-3-3. Following this, α6β4 is relocated to the lamellipodia from hemidesmosomes. The induction of α3β1 is associated with keratinocyte spread on laminin-5, as well as activation of p38 and NF-κB signaling, which is required for cell migration [192].
Keratinocyte proliferation is also stimulated by granulocyte macrophage-colony stimulating factor (GM-CSF) produced by keratinocytes, among others. Following binding to the CD116 receptor, GM-CSF activates the JAK/STAT signaling pathway. During wound healing, GM-CSF plays an indirect role by the induction of secondary cytokines. Studies of transgenic mice overexpressing the GM-SCF factor indicate that keratinocyte-derived GM-CSF overexpression takes place in the basal layer and is related to accelerated wound contracting. In the first phase of that process, significant mitogenic keratinocyte growth and granulation tissue formation were observed, as well as different regulation of TGF-β, IFN-γ, and iIL-6 release [203].
Another factor, angiopoietin-related growth factor (AGF), is able to bind to keratinocytes αv integrin via RGD-motif. Transgenic mice overexpressing AGF revealed increased proliferation of keratinocytes and more rapid wound closure compared to wild-type controls. AGF is also produced by platelets and mast cells; however, AGF release takes place only on wounded skin. AGF is profusely expressed by hepatocytes [204].
High mobility group protein β1 (HMGB1) enhances keratinocyte migration by an unknown receptor. That factor is released by macrophages and monocytes. HaCaT keratinocytes exposed to HMGB1 exhibit accelerated migration and proliferation, as well as ERK1/2 pathway induction [205]. An in vivo study found that diabetic mice demonstrate lower levels of HMGB1 than non-diabetic mice. In addition, topical application of HMGB1 resulted in accelerated wound healing in diabetic mice, but not in normoglycemic mice. Moreover, it was observed that HMGB1 plays a chemotactic role on keratinocytes in vitro [206].
Hypoxia-inducible factor-1 (HIF-1), a regulator of oxygen homeostasis, is released during the hypoxia accompanying wound healing. The presence of HIF-1 results in elevated expression of heat shock protein 90 (HSP90) by keratinocytes and binding to LDL receptor-related protein 1 (LRP-1) followed by an increase in cell migration. HSP90 regulates the initial phase of wound healing [207,208] and acts synergistically with TGF-α [209]. An in vitro heat shock assay performed with HaCatT cells increases keratinocyte motility, whereas an in vivo assay performed on mice with thermal burns shows that topical application of HSP90 increases tissue granulation and reduces inflammation [210].
Cytokines including IL-1, IL-6 and TNF-α also regulate keratinocyte migration. IL-1 promotes the secretion of FGF-7 [211], while IL-6 stimulates motility via the STAT-3 dependent pathway [212].
Chemokines produced by various types of cells may also propagate keratinocyte motility. The N terminus takes part in receptor activation. Keratinocytes express CXCL1, 8, 10, 11 and CCR14, 17, 27, which stimulate their migration, whereas CXCL1, 8 and 12 accelerate their proliferation. Reduced keratinocyte motility and proliferation has been observed in CXCR2 knockout mice. CXCR2 is a receptor for CXCL1 (growth-related oncogene-α) and CXCL8 (IL-8) chemokines, which are essential for keratinocyte activity [208]. CXCL10 and CXCL11 are expressed by basal keratinocytes during the re-epithelialization phase [213]. CXCR3 is a receptor for CXCL10 and CXCL11. CXCR3 knockout mice demonstrate delayed re-epithelization [214]. Interestingly, keratinocyte migration and wound closure were accelerated in mice with thick wounds in the presence of CXCL11 [215]. Conversely, reduced proliferation was observed in keratinocytes exposed to an elevated level of CXCL8 [216].
Neuropeptides, molecules released, among others, by sensory neurons during cutaneous injury, can also accelerate keratinocyte motility and proliferation [217]. The G protein-coupled receptor (GCRP) increases the mitogenic potential of keratinocytes [218]. Vasoactive intestinal peptide (VIP) was found to directly induce keratinocyte migration in vitro [219], or by the upregulation of the production of TGF-α [220] and VEGF [221]. Substance P (SP), which binds to the neurokinin 1 receptor (NK1R), and calcitonin gene-related peptide (CGRP) stimulate the production of inflammatory agents including IL-1, IL-6, TNF-α and nerve growth factor (NGF) in keratinocytes, as well as their motility. Similarly, SP and NK1R have been found to promote human and murine keratinocyte proliferation [222,223,224,225,226]. SP and NK1R SP activate all three members of the MAPK family, whereas CGRP only induces p38 and ERK1/2 [227]. Keratinocytes under hypoxia and poor nutrient environments similar to chronic wounds exhibit accelerated cell proliferation when exposed to SP, and mice with full-thickness wounds demonstrated elevated wound closure after topical application of SP [228]. Diabetic wounded mice also exhibited accelerated re-epithelialization after SP exposure [229]; however, an SP damage agent named neutral endopeptidase (NEP) was also found to be upregulated in diabetic mice wounds. This may be due to the impaired activities of keratinocytes under the influence of NEP inhibitors [230]. It was observed that, under hyperglycemic conditions, keratinocytes demonstrate lower expression of the neuropeptide neurotensin (NT) and neurotensin receptor (NTR); however, the addition of exogenous NT do not impact keratinocyte proliferation but even reduced migration [231]. An in vitro study on keratinocytes indicated increased expression of IL-1α, IL-8, TNF-α and NGF mRNA following treatment with SP, CGRP, VIP and galanin (GAL) [232]. It was also found that keratinocyte expresses the GAL receptor GALR2, which is known to influence keratinocyte proliferation [233]. In addition, β-endorphin also appears to enhance keratinocyte motility by binding with mu-opiate receptor [234].
Keratinocytes also express cholinergic receptors, including nicotinic (nAChR), including α3, α5, α7, α9, α10, β1, β2, and β4 and muscarinic acetylcholine (ACh) (mAChR) forms, including M1, 2, 3, 4, and 5. Increased keratinocyte migration is observed after the activation of the following receptors: α3 by PKCδ and RhoA signaling; α7 by PI3K and Rac/Cdc42 signaling; α9 by modulating adhesion between cells and between the cell and extracellular matrix (ECM) and M4 by integrins α5β1, αvβ5 and αvβ6. Conversely, M3 activation upregulates integrins α2β1 and α3β1, related to adhesion, and suppresses keratinocyte migration [235,236,237].
In wounds, keratinocyte migration onto the dermal matrix and MMP-1 expression is mediated by native type I collagen [238]. In vitro data indicate that MMP-1 fragmented the collagen, which was followed by impaired keratinocyte function and slow spread [239]. Interestingly, it has been suggested that chronic wounds, characterized by accelerated IL-1β and TNF-α expression, result in elevated levels of MMPs and the secretion of growth factors such as FGFs. This results in reduced keratinocyte migration due to degradation of ECM components and the greater availability of selected growth factors [172]. Throughout proteolytic activity, MMPs convert the latent form of growth factors into active ones, including IGF-1 [240].
The plant extracts and their component compounds may modulate keratinocyte migration and proliferation rates via various mechanisms, including stimulation of the MAPK and PI3K/AKT signal transduction pathways. In some studies, a higher expression of β1-, α6-, β4-integrin and E-cadherin were observed after extract treatment. Table 4 presents the impact of selected plant extracts on the migration and proliferation rates of human keratinocytes. As cellular motility is closely connected with the induction of various growth factors, the presented plant-derived compounds exhibit important properties that modulate the production of growth factors and enhance migration followed by accelerated wound closure.
A study of the abovementioned Aegle marmelos [253], Boerhavia diffusa [254] and Stellera chamaejasme [260] extracts found them to reduce wound area in rats. Annona reticulata [255] and Centella asiatica [259] also demonstrated acceleration of wound healing after topical application in mice and rabbits, respectively.
Finally, cyanidin-3-glucoside derived from blackberry [263] and chlorogenic acid derived from Parrotia persica [264] significantly accelerate the wound closure process in keratinocytes in vitro.

9. Conclusions

Plants are key sources of secondary metabolites that exert various antioxidant and anti-inflammatory effects, among others. These compounds are able to modulate signaling pathways in numerous cells, including epidermal cells. Keratinocytes placed in the outer layer of the skin create a physical barrier against harmful stimuli. They are particularly vulnerable to UV-radiation, related oxidative stress and inflammation. Any disruption of the epidermis stimulates cell migration, proliferation and participation in wound healing. It is suggested that such compounds of plant origin may be used to modulate keratinocyte function by improving ROS scavenging, inhibiting inflammation and accelerating wound healing via influencing signal transduction pathways. Hence, plant extracts and their component compounds may have an impact on keratinocyte biology and its ability to maintain homeostasis. However, further in vitro and in vivo studies of the mechanisms of the action of phytochemicals, and more specific toxicity and clinical studies are needed to ensure the effectiveness and safety of plant compounds for use on human skin.

Author Contributions

Conceptualization, A.M.-S. and P.S.; Formal Analysis, E.K.; K.Z. and R.Z.; Data curation, E.K.; K.Z. and R.Z.; Writing—original draft preparation, A.M.-S.; Writing—review and editing, A.M.-S., P.S. and T.K.; Supervision—R.Z. 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

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Erb, M.; Kliebenstein, D.J. Plant Secondary Metabolites as Defenses, Regulators, and Primary Metabolites: The Blurred Functional Trichotomy. Plant Physiol. 2020, 184, 39–52. [Google Scholar] [CrossRef] [PubMed]
  2. Wickett, R.R.; Visscher, M.O. Structure and function of the epidermal barrier. Am. J. Infect. Control 2006, 34, S98–S110. [Google Scholar] [CrossRef]
  3. Kasote, D.M.; Katyare, S.S.; Hegde, M.V.; Bae, H. Significance of antioxidant potential of plants and its relevance to therapeutic applications. Int. J. Biol. Sci. 2015, 11, 982–991. [Google Scholar] [CrossRef] [Green Version]
  4. Nunes, C.R.; Arantes, M.B.; de Faria Pereira, S.M.; da Cruz, L.L.; de Souza Passos, M.; de Moraes, L.P.; Vieira, I.J.C.; de Oliveira, D.B. Plants as Sources of Anti-Inflammatory Agents. Molecules 2020, 25, 3726. [Google Scholar] [CrossRef]
  5. De Jager, T.L.; Cockrell, A.E.; Du Plessis, S.S. Ultraviolet light induced generation of reactive oxygen species. Adv. Exp. Med. Biol. 2017, 996, 15–23. [Google Scholar] [CrossRef] [PubMed]
  6. Ciążyńska, M.; Olejniczak-Staruch, I.; Sobolewska-Sztychny, D.; Narbutt, J.; Skibińska, M.; Lesiak, A. Ultraviolet radiation and chronic inflammation-molecules and mechanisms involved in skin carcinogenesis: A narrative review. Life 2021, 11, 326. [Google Scholar] [CrossRef]
  7. Pastar, I.; Stojadinovic, O.; Tomic-Canic, M. Role of keratinocytes in healing of chronic wounds. Surg. Technol. Int. 2008, 17, 105–112. [Google Scholar]
  8. Teoh, E.S. Secondary Metabolites of Plants. In Medicinal Orchids of Asia; Springer: Berlin/Heidelberg, Germany, 2016; pp. 59–73. [Google Scholar] [CrossRef]
  9. Hussein, A.R.; El-Anssary, A.A. Plants Secondary Metabolites: The Key Drivers of the Pharmacological Actions of Medicinal Plants. In Herbal Medicine; IntechOpen Limited: London, UK, 2019. [Google Scholar]
  10. Merecz-Sadowska, A.; Sitarek, P.; Kucharska, E.; Kowalczyk, T.; Zajdel, K.; Cegliński, T.; Zajdel, R. Antioxidant properties of plant-derived phenolic compounds and their effect on skin fibroblast cells. Antioxidants 2021, 10, 726. [Google Scholar] [CrossRef]
  11. Merecz-Sadowska, A.; Sitarek, P.; Śliwiński, T.; Zajdel, R. Anti-inflammatory activity of extracts and pure compounds derived from plants via modulation of signaling pathways, especially PI3K/akt in macrophages. Int. J. Mol. Sci. 2020, 21, 9605. [Google Scholar] [CrossRef]
  12. Sitarek, P.; Merecz-Sadowska, A.; Śliwiński, T.; Zajdel, R.; Kowalczyk, T. An in vitro evaluation of the molecular mechanisms of action of medical plants from the lamiaceae family as effective sources of active compounds against human cancer cell lines. Cancers 2020, 12, 2957. [Google Scholar] [CrossRef] [PubMed]
  13. Josiah, A.J.; Twilley, D.; Pillai, S.K.; Ray, S.S.; Lall, N. Pathogenesis of Keratinocyte Carcinomas and the Therapeutic Potential of Medicinal Plants and Phytochemicals. Molecules 2021, 26, 1979. [Google Scholar] [CrossRef]
  14. Sitarek, P.; Merecz-Sadowska, A.; Kowalczyk, T.; Wieczfinska, J.; Zajdel, R.; Śliwiński, T. Potential synergistic action of bioactive compounds from plant extracts against skin infecting microorganisms. Int. J. Mol. Sci. 2020, 21, 5105. [Google Scholar] [CrossRef]
  15. Sitarek, P.; Kowalczyk, T.; Wieczfinska, J.; Merecz-Sadowska, A.; Górski, K.; Śliwiński, T.; Skała, E. Plant extracts as a natural source of bioactive compounds and potential remedy for the treatment of certain skin diseases. Curr. Pharm. Des. 2020, 26, 2859–2875. [Google Scholar] [CrossRef]
  16. Zielinska-Blizniewska, H.; Sitarek, P.; Merecz-Sadowska, A.; Malinowska, K.; Zajdel, K.; Jablonska, M.; Sliwinski, T.; Zajdel, R. Plant extracts and reactive oxygen species as two counteracting agents with anti- and pro-obesity properties. Int. J. Mol. Sci. 2019, 20, 4556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Chiocchio, I.; Mandrone, M.; Tomasi, P.; Marincich, L.; Poli, F. Plant secondary metabolites: An opportunity for circular economy. Molecules 2021, 26, 495. [Google Scholar] [CrossRef]
  18. Ekor, M. The growing use of herbal medicines: Issues relating to adverse reactions and challenges in monitoring safety. Front. Neurol. 2014, 4, 177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Abubakar, A.R.; Haque, M. Preparation of medicinal plants: Basic extraction and fractionation procedures for experimental purposes. J. Pharm. Bioallied Sci. 2020, 12, 1–10. [Google Scholar] [CrossRef]
  20. Atanasov, A.G.; Waltenberger, B.; Pferschy-Wenzig, E.M.; Linder, T.; Wawrosch, C.; Uhrin, P.; Temml, V.; Wang, L.; Schwaiger, S.; Heiss, E.H.; et al. Discovery and resupply of pharmacologically active plant-derived natural products: A review. Biotechnol. Adv. 2015, 33, 1582–1614. [Google Scholar] [CrossRef] [Green Version]
  21. Abdo, J.M.; Sopko, N.A.; Milner, S.M. The applied anatomy of human skin: A model for regeneration. Wound Med. 2020, 28, 100179. [Google Scholar] [CrossRef]
  22. Apalla, Z.; Nashan, D.; Weller, R.B.; Castellsagué, X. Skin Cancer: Epidemiology, Disease Burden, Pathophysiology, Diagnosis, and Therapeutic Approaches. Dermatol. Ther. 2017, 7 (Suppl. 1), 5–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Botchkarev, V.A.; Gdula, M.R.; Mardaryev, A.N.; Sharov, A.A.; Fessing, M.Y. Epigenetic regulation of gene expression in keratinocytes. J. Investig. Dermatol. 2012, 132, 2505–2521. [Google Scholar] [CrossRef] [Green Version]
  24. Tomic-Canic, M.; Komine, M.; Freedberg, I.M.; Blumenberg, M. Epidermal signal transduction and transcription factor activation in activated keratinocytes. J. Dermatol. Sci. 1998, 17, 167–181. [Google Scholar] [CrossRef]
  25. Jost, M.; Huggett, T.M.; Kari, C.; Rodeck, U. Matrix-independent survival of human keratinocytes through an EGF receptor/MAPK-kinase-dependent pathway. Mol. Biol. Cell. 2001, 12, 1519–1527. [Google Scholar] [CrossRef] [Green Version]
  26. Deucher, A.; Efimova, T.; Eckert, R.L. Calcium-dependent involucrin expression is inversely regulated by protein kinase C (PKC)α and PKCδ. J. Biol. Chem. 2002, 277, 17032–17040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Meng, X.; Qiu, L.; Song, H.; Dang, N. MAPK pathway involved in epidermal terminal differentiation of normal human epidermal keratinocytes. Open Med. 2018, 13, 189–195. [Google Scholar] [CrossRef]
  28. Yano, S.; Komine, M.; Fujimoto, M.; Okochi, H.; Tamaki, K. Mechanical stretching in vitro regulates signal transduction pathways and cellular proliferation in human epidermal keratinocytes. J. Investig. Dermatol. 2004, 122, 783–790. [Google Scholar] [CrossRef] [Green Version]
  29. El Darzi, E.; Bazzi, S.; Daoud, S.; Echtay, K.S.; Bahr, G.M. Differential regulation of surface receptor expression, proliferation, and apoptosis in HaCaT cells stimulated with interferon-3, interleukin-4, tumor necrosis factor-α, or muramyl dipeptide. Int. J. Immunopathol. Pharmacol. 2017, 30, 130–145. [Google Scholar] [CrossRef] [Green Version]
  30. Mitev, V.; Miteva, L. Signal transduction in keratinocytes. Exp. Dermatol. 1999, 8, 96–108. [Google Scholar] [CrossRef] [Green Version]
  31. Sun, L.R.; Zhou, W.; Zhang, H.M.; Guo, Q.S.; Yang, W.; Li, B.J.; Sun, Z.H.; Gao, S.H.; Cui, R.J. Modulation of Multiple Signaling Pathways of the Plant-Derived Natural Products in Cancer. Front. Oncol. 2019, 9, 1153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Gupta, A.; Rosenberger, S.F.; Bowden, G.T. Increased ROS levels contribute to elevated transcription factor and MAP kinase activities in malignantly progressed mouse keratinocyte cell lines. Carcinogenesis 1999, 20, 2063–2073. [Google Scholar] [CrossRef] [Green Version]
  33. Bito, T.; Nishigori, C. Impact of reactive oxygen species on keratinocyte signaling pathways. J. Dermatol. Sci. 2012, 68, 3–8. [Google Scholar] [CrossRef] [PubMed]
  34. Leopoldini, M.; Marino, T.; Russo, N.; Toscano, M. Antioxidant properties of phenolic compounds: H-atom versus electron transfer mechanism. J. Phys. Chem. A 2004, 108, 4916–4922. [Google Scholar] [CrossRef]
  35. Dangles, O. Antioxidant Activity of Plant Phenols: Chemical Mechanisms and Biological Significance. Curr. Org. Chem. 2012, 16, 692–714. [Google Scholar] [CrossRef]
  36. Minatel, I.O.; Borges, C.V.; Ferreira, M.I.; Gomez, H.A.G.; Chen, C.-Y.O.; Lima, G.P.P. Phenolic Compounds: Functional Properties, Impact of Processing and Bioavailability. In Phenolic Compounds—Biological Activity; IntechOpen Limited: London, UK, 2017. [Google Scholar]
  37. Mussard, E.; Jousselin, S.; Cesaro, A.; Legrain, B.; Lespessailles, E.; Esteve, E.; Berteina-raboin, S.; Toumi, H. Andrographis paniculata and its bioactive diterpenoids against inflammation and oxidative stress in keratinocytes. Antioxidants 2020, 9, 530. [Google Scholar] [CrossRef]
  38. Liu, C.; Guo, H.; Dain, J.A.; Wan, Y.; Gao, X.H.; Chen, H.D.; Seeram, N.P.; Ma, H. Cytoprotective effects of a proprietary red maple leaf 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]
  39. Zhou, Y.; Yang, W.; Li, Z.; Luo, D.; Li, W.; Zhang, Y.; Wang, X.; Fang, M.; Chen, Q.; Jin, X. Moringa oleifera stem extract protect skin keratinocytes against oxidative stress injury by enhancement of antioxidant defense systems and activation of PPARα. Biomed. Pharmacother. 2018, 107, 44–53. [Google Scholar] [CrossRef]
  40. Kolakul, P.; Sripanidkulchai, B. Phytochemicals and anti-aging potentials of the extracts from Lagerstroemia speciosa and Lagerstroemia floribunda. Ind. Crops Prod. 2017, 109, 707–716. [Google Scholar] [CrossRef]
  41. Liu, C.; Guo, H.; DaSilva, N.A.; Li, D.; Zhang, K.; Wan, Y.; Gao, X.H.; Chen, H.D.; Seeram, N.P.; Ma, H. Pomegranate (Punica granatum) phenolics ameliorate hydrogen peroxide-induced oxidative stress and cytotoxicity in human keratinocytes. J. Funct. Foods 2019, 54, 559–567. [Google Scholar] [CrossRef]
  42. Squillaci, G.; Apone, F.; Sena, L.M.; Carola, A.; Tito, A.; Bimonte, M.; Lucia, A.D.; Colucci, G.; Cara, F.L.; Morana, A. Chestnut (Castanea sativa Mill.) industrial wastes as a valued bioresource for the production of active ingredients. Process Biochem. 2018, 64, 228–236. [Google Scholar] [CrossRef]
  43. Lee, S.Y.; Kim, C.H.; Hwang, B.S.; Choi, K.M.; Yang, I.J.; Kim, G.Y.; Choi, Y.H.; Park, C.; Jeong, J.W. Protective effects of Oenothera biennis against hydrogen peroxide-induced oxidative stress and cell death in skin keratinocytes. Life 2020, 10, 255. [Google Scholar] [CrossRef] [PubMed]
  44. Bazzicalupo, M.; Burlando, B.; Denaro, M.; Barreca, D.; Trombetta, D.; Smeriglio, A.; Cornara, L. Polyphenol characterization and skin-preserving properties of hydroalcoholic flower extract from Himantoglossum robertianum (Orchidaceae). Plants 2019, 8, 502. [Google Scholar] [CrossRef] [Green Version]
  45. Do, N.Q.; Zheng, S.; Park, B.; Nguyen, Q.T.N.; Choi, B.R.; Fang, M.; Kim, M.; Jeong, J.; Choi, J.; Yang, S.J.; et al. Camu-camu fruit extract inhibits oxidative stress and inflammatory responses by regulating NFAT and Nrf2 signaling pathways in high glucose-induced human keratinocytes. Molecules 2021, 26, 3174. [Google Scholar] [CrossRef]
  46. Zakaria, N.N.A.; Okello, E.J.; Howes, M.J.; Birch-Machin, M.A.; Bowman, A. In vitro protective effects of an aqueous extract of Clitoria ternatea L. flower against hydrogen peroxide-induced cytotoxicity and UV-induced mtDNA damage in human keratinocytes. Phyther. Res. 2018, 32, 1064–1072. [Google Scholar] [CrossRef] [Green Version]
  47. Pastore, S.; Mascia, F.; Mariani, V.; Girolomoni, G. Keratinocytes in skin inflammation. Expert Rev. Dermatol. 2006, 1, 279–291. [Google Scholar] [CrossRef]
  48. Ćabrijan, L.; Lipozenćić, J. Adhesion molecules in keratinocytes. Clin. Dermatol. 2011, 29, 427–431. [Google Scholar] [CrossRef] [PubMed]
  49. Bernard, F.-X.; Morel, F.; Camus, M.; Pedretti, N.; Barrault, C.; Garnier, J.; Lecron, J.-C. Keratinocytes under Fire of Proinflammatory Cytokines: Bona Fide Innate Immune Cells Involved in the Physiopathology of Chronic Atopic Dermatitis and Psoriasis. J. Allergy 2012, 2012, 718725. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Lebre, M.C.; Van Der Aar, A.M.G.; Van Baarsen, L.; Van Capel, T.M.M.; Schuitemaker, J.H.N.; Kapsenberg, M.L.; De Jong, E.C. Human keratinocytes express functional toll-like receptor 3, 4, 5, and 9. J. Investig. Dermatol. 2007, 127, 331–341. [Google Scholar] [CrossRef] [Green Version]
  51. Miller, L.S. Toll-Like Receptors in Skin. Adv. Dermatol. 2008, 24, 71–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Li, Z.J.; Sohn, K.C.; Choi, D.K.; Shi, G.; Hong, D.; Lee, H.E.; Whang, K.U.; Lee, Y.H.; Im, M.; Lee, Y.; et al. Roles of TLR7 in Activation of NF-κB Signaling of Keratinocytes by Imiquimod. PLoS ONE 2013, 8, e77159. [Google Scholar] [CrossRef] [Green Version]
  53. Pivarcsi, A.; Bodai, L.; Réthi, B.; Kenderessy-Szabó, A.; Koreck, A.; Széll, M.; Beer, Z.; Bata-Csörgo, Z.; Magócsi, M.; Rajnavölgyi, E.; et al. Expression and function of Toll-like receptors 2 and 4 in human keratinocytes. Int. Immunol. 2003, 15, 721–730. [Google Scholar] [CrossRef] [Green Version]
  54. Gröne, A. Keratinocytes and cytokines. Vet. Immunol. Immunopathol. 2002, 88, 1–12. [Google Scholar] [CrossRef]
  55. Sauder, D.N.; Orr, F.W.; Matic, S.; Stetsko, D.; Parker, K.P.; Chizzonite, R.; Kilian, P.L. Human interleukin-1α is chemotactic for normal human keratinocytes. Immunol. Lett. 1989, 22, 123–127. [Google Scholar] [CrossRef]
  56. Komine, M.; Rao, L.S.; Freedberg, I.M.; Simon, M.; Milisavljevic, V.; Blumenberg, M. Interleukin-1 induces transcription of keratin K6 in human epidermal keratinocytes. J. Investig. Dermatol. 2001, 116, 330–338. [Google Scholar] [CrossRef] [Green Version]
  57. Darmstadt, G.L.; Fleckman, P.; Rubens, C.E. Tumor necrosis factor-α and interleukin-1α decrease the adherence of Streptococcus pyogenes to cultured keratinocytes. J. Infect. Dis. 1999, 180, 1718–1721. [Google Scholar] [CrossRef]
  58. Kothny-Wilkes, G.; Kulms, D.; Pöppelmann, B.; Luger, T.A.; Kubin, M.; Schwarz, T. Interleukin-1 protects transformed keratinocytes from tumor necrosis factor-related apoptosis-inducing ligand. J. Biol. Chem. 1998, 273, 29247–29253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Feldmeyer, L.; Werner, S.; French, L.E.; Beer, H.D. Interleukin-1, inflammasomes and the skin. Eur. J. Cell Biol. 2010, 89, 638–644. [Google Scholar] [CrossRef]
  60. Yoshizaki, K.; Nishimoto, N.; Matsumoto, K.; Tagoh, H.; Taga, T.; Deguchi, Y.; Kuritani, T.; Hirano, T.; Hashimoto, K.; Okada, N.; et al. Interleukin 6 and expression of its receptor on epidermal keratinocytes. Cytokine 1990, 2, 381–387. [Google Scholar] [CrossRef]
  61. Sugawara, T.; Gallucci, R.M.; Simeonova, P.P.; Luster, M.I. Regulation and role of interleukin 6 in wounded human epithelial keratinocytes. Cytokine 2001, 15, 328–336. [Google Scholar] [CrossRef]
  62. Wang, X.P.; Schunck, M.; Kallen, K.J.; Neumann, C.; Trautwein, C.; Rose-John, S.; Proksch, E. The interleukin-6 cytokine system regulates epidermal permeability barrier homeostasis. J. Investig. Dermatol. 2004, 123, 124–131. [Google Scholar] [CrossRef] [Green Version]
  63. Kondo, S.; Kono, T.; Sauder, D.N.; McKenzie, R.C. IL-8 gene expression and production in human keratinocytes and their modulation by UVB. J. Investig. Dermatol. 1993, 101, 690–694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Jiang, W.G.; Sanders, A.J.; Ruge, F.; Harding, K.G. Influence of interleukin-8 (IL-8) and IL-8 receptors on the migration of human keratinocytes, the role of plc-γ and potential clinical implications. Exp. Ther. Med. 2012, 3, 231–236. [Google Scholar] [CrossRef] [Green Version]
  65. Banno, T.; Gazel, A.; Blumenberg, M. Effects of tumor necrosis factor-α (TNFα) in epidermal keratinocytes revealed using global transcriptional profiling. J. Biol. Chem. 2004, 279, 32633–32642. [Google Scholar] [CrossRef] [Green Version]
  66. Kim, H.R.; Hwang, K.A.; Park, S.H.; Kang, I. IL-7 and IL-15: Biology and roles in T-cell immunity in health and disease. Crit. Rev. Immunol. 2008, 28, 325–339. [Google Scholar] [CrossRef]
  67. Williams, I.R.; Rawson, E.A.; Manning, L.; Karaoli, T.; Rich, B.E.; Kupper, T.S. IL-7 overexpression in transgenic mouse keratinocytes causes a lymphoproliferative skin disease dominated by intermediate TCR cells: Evidence for a hierarchy in IL-7 responsiveness among cutaneous T cells. J. Immunol. 1997, 159, 3044–30456. [Google Scholar]
  68. Takashima, A.; Matsue, H.; Bergstresser, P.R.; Ariizumi, K. Interleukin-7-dependent interaction of dendritic epidermal t cells with keratinocytes. J. Investig. Dematol. 1995, 105 (Suppl. 1), S50–S53. [Google Scholar] [CrossRef] [Green Version]
  69. Blauvelt, A.; Asada, H.; Klaus-Kovtun, V.; Altman, D.J.; Lucey, D.R.; Katz, S.I. Interleukin-15 mRNA is expressed by human keratinocytes, langerhans cells, and blood-derived dendritic cells and is downregulated by ultraviolet B radiation. J. Investig. Dermatol. 1996, 106, 1047–1052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Moore, K.W.; De Waal Malefyt, R.; Coffman, R.L.; O’Garra, A. Interleukin-10 and the interleukin-10 receptor. Annu. Rev. Immunol. 2001, 19, 683–765. [Google Scholar] [CrossRef]
  71. Enk, C.D.; Sredni, D.; Blauvelt, A.; Katz, S.I. Induction of IL-10 gene expression in human keratinocytes by UVB exposure in vivo and in vitro. J. Immunol. 1995, 154, 4851–4856. [Google Scholar] [CrossRef]
  72. Hirohata, S. Human Th1 responses driven by IL-12 are associated with enhanced expression of CD40 ligand. Clin. Exp. Immunol. 1999, 115, 78–85. [Google Scholar] [CrossRef] [PubMed]
  73. Kulig, P.; Musiol, S.; Freiberger, S.N.; Schreiner, B.; Gyu’lveszi, G.; Russo, G.; Pantelyushin, S.; Kishihara, K.; Alessandrini, F.; Ku’ndig, T.; et al. IL-12 protects from psoriasiform skin inflammation. Nat. Commun. 2016, 7, 13466. [Google Scholar] [CrossRef] [PubMed]
  74. Werth, V.P.; Bashir, M.M.; Zhang, W. IL-12 completely blocks ultraviolet-induced secretion of tumor necrosis factor α from cultured skin fibroblasts and keratinocytes. J. Investig. Dermatol. 2003, 120, 116–122. [Google Scholar] [CrossRef] [Green Version]
  75. Nakanishi, K.; Yoshimoto, T.; Tsutsui, H.; Okamura, H. Interleukin-18 regulates both Th1 and Th2 responses. Annu. Rev. Immunol. 2001, 19, 423–474. [Google Scholar] [CrossRef]
  76. Rich, B.E.; Kupper, T.S. Cytokines: IL-20—A new effector in skin inflammation. Curr. Biol. 2001, 11, R531–R534. [Google Scholar] [CrossRef] [Green Version]
  77. Jiang, Y.; Tsoi, L.C.; Billi, A.C.; Ward, N.L.; Harms, P.W.; Zeng, C.; Maverakis, E.; Michelle Kahlenberg, J.; Gudjonsson, J.E. Cytokinocytes: The diverse contribution of keratinocytes to immune responses in skin. JCI Insight 2020, 5, e142067. [Google Scholar] [CrossRef] [PubMed]
  78. Alilou, M.; Marzocco, S.; Hofer, D.; Rapa, S.F.; Asadpour, R.; Schwaiger, S.; Troppmair, J.; Stuppner, H. Labdane-Type Diterpenes from the Aerial Parts of Rydingia persica: Their Absolute Configurations and Protective Effects on LPS-Induced Inflammation in Keratinocytes. J. Nat. Prod. 2020, 83, 2456–2468. [Google Scholar] [CrossRef]
  79. Pintatum, A.; Maneerat, W.; Logie, E.; Tuenter, E.; Sakavitsi, M.E.; Pieters, L.; Berghe, W.V.; Sripisut, T.; Deachathai, S.; Laphookhieo, S. In vitro anti-inflammatory, anti-oxidant, and cytotoxic activities of four curcuma species and the isolation of compounds from Curcuma aromatica rhizome. Biomolecules 2020, 10, 799. [Google Scholar] [CrossRef] [PubMed]
  80. Ahama-Esseh, K.; Bodet, C.; Quashie-Mensah-Attoh, A.; Garcia, M.; Théry-Koné, I.; Dorat, J.; De Souza, C.; Enguehard-Gueiffier, C.; Boudesocque-Delaye, L. Anti-inflammatory activity of Crateva adansonii DC on keratinocytes infected by Staphylococcus aureus: From traditional practice to scientific approach using HPTLC-densitometry. J. Ethnopharmacol. 2017, 204, 26–35. [Google Scholar] [CrossRef] [PubMed]
  81. Yang, J.H.; Yoo, J.M.; Lee, E.; Lee, B.H.; Cho, W.K.; Park, K.I.; Yeul Ma, J. Anti-inflammatory effects of Perillae herba ethanolic extract against TNF-α/IFN-γ-stimulated human keratinocyte HaCaT cells. J. Ethnopharmacol. 2018, 211, 217–223. [Google Scholar] [CrossRef]
  82. Khalilpour, S.; Sangiovanni, E.; Piazza, S.; Fumagalli, M.; Beretta, G.; Dell’Agli, M. In vitro evidences of the traditional use of Rhus coriaria L. fruits against skin inflammatory conditions. J. Ethnopharmacol. 2019, 238, 111829. [Google Scholar] [CrossRef]
  83. Choi, Y.A.; Yu, J.H.; Jung, H.D.; Lee, S.; Park, P.H.; Lee, H.S.; Kwon, T.K.; Shin, T.Y.; Lee, S.W.; Rho, M.C.; et al. Inhibitory effect of ethanol extract of Ampelopsis brevipedunculata rhizomes on atopic dermatitis-like skin inflammation. J. Ethnopharmacol. 2019, 238, 111850. [Google Scholar] [CrossRef]
  84. Yang, J.H.; Hwang, Y.H.; Gu, M.J.; Cho, W.K.; Ma, J.Y. Ethanol extracts of Sanguisorba officinalis L. suppress TNF-α/IFN-γ-induced pro-inflammatory chemokine production in HaCaT cells. Phytomedicine 2015, 22, 1262–1268. [Google Scholar] [CrossRef]
  85. Seo, C.S.; Lim, H.S.; Ha, H.; Jin, S.E.; Shin, H.K. Quantitative analysis and anti-inflammatory effects of Gleditsia sinensis thorns in RAW 264.7 macrophages and HaCaT keratinocytes. Mol. Med. Rep. 2015, 12, 4773–4781. [Google Scholar] [CrossRef]
  86. Hardianti, B.; Umeyama, L.; Li, F.; Yokoyama, S.; Hayakawa, Y. Anti-inflammatory compounds moracin O and P from Morus alba Linn. (Sohakuhi) target the NF-κB pathway. Mol. Med. Rep. 2020, 22, 5385–5391. [Google Scholar] [CrossRef]
  87. Jin, S.E.; Ha, H.; Shin, H.K.; Seo, C.S. Anti-allergic and anti-inflammatory effects of Kuwanon G and Morusin on MC/9 mast cells and HaCaT keratinocytes. Molecules 2019, 24, 265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Marquardt, P.; Seide, R.; Vissiennon, C.; Schubert, A.; Birkemeyer, C.; Ahyi, V.; Fester, K. Phytochemical characterization and in vitro anti-inflammatory, antioxidant and antimicrobial activity of Combretum collinum Fresen leaves extracts from Benin. Molecules 2020, 25, 288. [Google Scholar] [CrossRef] [Green Version]
  89. Lim, H.S.; Jin, S.E.; Kim, O.S.; Shin, H.K.; Jeong, S.J. Alantolactone from Saussurea lappa Exerts Antiinflammatory Effects by Inhibiting Chemokine Production and STAT1 Phosphorylation in TNF-α and IFN-γ-induced in HaCaT cells. Phyther. Res. 2015, 29, 1088–1096. [Google Scholar] [CrossRef] [PubMed]
  90. Kim, M.; Yin, J.; Hwang, I.H.; Park, D.H.; Lee, E.K.; Kim, M.J.; Lee, M.W. Anti-Acne vulgaris effects of pedunculagin from the leaves of Quercus mongolica by anti-inflammatory activity and 5α-reductase inhibition. Molecules 2020, 25, 2154. [Google Scholar] [CrossRef]
  91. Albouchi, F.; Avola, R.; Dico, G.M.L.; Calabrese, V.; Graziano, A.C.E.; Abderrabba, M.; Cardile, V. Melaleuca styphelioides Sm. Polyphenols modulate interferon gamma/histamine-induced inflammation in human NCTC 2544 keratinocytes. Molecules 2018, 23, 2526. [Google Scholar] [CrossRef] [Green Version]
  92. Yin, J.; Hwang, I.H.; Lee, M.W. Anti-acne vulgaris effect including skin barrier improvement and 5α-reductase inhibition by tellimagrandin I from Carpinus tschonoskii. BMC Complement. Altern. Med. 2019, 19, 323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Ko, W.; Kim, N.; Lee, H.; Woo, E.R.; Kim, Y.C.; Oh, H.; Lee, D.S. Anti-inflammatory effects of compounds from Cudrania tricuspidata in hacat human keratinocytes. Int. J. Mol. Sci. 2021, 22, 7472. [Google Scholar] [CrossRef]
  94. Svensson, D.; Lozano, M.; Almanza, G.R.; Nilsson, B.O.; Sterner, O.; Villagomez, R. Sesquiterpene lactones from Ambrosia arborescens Mill. inhibit pro-inflammatory cytokine expression and modulate NF-κB signaling in human skin cells. Phytomedicine 2018, 50, 118–126. [Google Scholar] [CrossRef] [PubMed]
  95. Oh, C.T.; Jang, Y.J.; Kwon, T.R.; Im, S.; Kim, S.R.; Seok, J.; Kim, G.Y.; Kim, Y.H.; Mun, S.K.; Kim, B.J. Effect of isosecotanapartholide isolated from Artemisia princeps Pampanini on IL-33 production and STAT-1 activation in HaCaT keratinocytes. Mol. Med. Rep. 2017, 15, 2681–2688. [Google Scholar] [CrossRef] [PubMed]
  96. D’Orazio, J.; Jarrett, S.; Amaro-Ortiz, A.; Scott, T. UV radiation and the skin. Int. J. Mol. Sci. 2013, 14, 12222–12248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Sun, X.; Kim, A.; Nakatani, M.; Shen, Y.; Liu, L. Distinctive molecular responses to ultraviolet radiation between keratinocytes and melanocytes. Exp. Dermatol. 2016, 25, 708–713. [Google Scholar] [CrossRef] [Green Version]
  98. Sesto, A.; Navarro, M.; Burslem, F.; Jorcano, J.L. Analysis of the ultraviolet B response in primary human keratinocytes using oligonucleotide microarrays. Proc. Natl. Acad. Sci. USA 2002, 99, 2965–2970. [Google Scholar] [CrossRef] [Green Version]
  99. El-Abaseri, T.B.; Putta, S.; Hansen, L.A. Ultraviolet irradiation induces keratinocyte proliferation and epidermal hyperplasia through the activation of the epidermal growth factor receptor. Carcinogenesis 2006, 27, 225–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Oda, K.; Matsuoka, Y.; Funahashi, A.; Kitano, H. A comprehensive pathway map of epidermal growth factor receptor signaling. Mol. Syst. Biol. 2005, 1, 2005.0010. [Google Scholar] [CrossRef] [Green Version]
  101. Calautti, E.; Li, J.; Saoncella, S.; Brissette, J.L.; Goetinck, P.F. Phosphoinositide 3-kinase signaling to Akt promotes keratinocyte differentiation versus death. J. Biol. Chem. 2005, 280, 32856–32865. [Google Scholar] [CrossRef] [Green Version]
  102. Takao, J.; Yudate, T.; Das, A.; Shikano, S.; Bonkobara, M.; Ariizumi, K.; Cruz, P.D. Expression of NF-κB in epidermis and the relationship between NF-κB activation and inhibition of keratinocyte growth. Br. J. Dermatol. 2003, 148, 680–688. [Google Scholar] [CrossRef]
  103. Barr, R.K.; Bogoyevitch, M.A. The c-Jun N-terminal protein kinase family of mitogen-activated protein kinases (JNK MAPKs). Int. J. Biochem. Cell Biol. 2001, 33, 1047–1063. [Google Scholar] [CrossRef]
  104. Syed, D.N.; Afaq, F.; Mukhtar, H. Differential activation of signaling pathways by UVA and UVB radiation in normal human epidermal keratinocytes. Photochem. Photobiol. 2012, 88, 1184–1190. [Google Scholar] [CrossRef] [PubMed]
  105. Adachi, M.; Gazel, A.; Pintucci, G.; Shuck, A.; Shifteh, S.; Ginsburg, D.; Rao, L.S.; Kaneko, T.; Freedberg, I.M.; Tamaki, K.; et al. Specificity in Stress Response: Epidermal Keratinocytes Exhibit Specialized UV-Responsive Signal Transduction Pathways. DNA Cell Biol. 2003, 22, 665–677. [Google Scholar] [CrossRef] [PubMed]
  106. Sano, S.; Chan, K.S.; Carbajal, S.; Clifford, J.; Peavey, M.; Kiguchi, K.; Itami, S.; Nickoloff, B.J.; DiGiovanni, J. Stat3 links activated keratinocytes and immunocytes required for development of psoriasis in a novel transgenic mouse model. Nat. Med. 2005, 11, 43–49. [Google Scholar] [CrossRef]
  107. Muthusamy, V.; Piva, T.J. A comparative study of UV-induced cell signalling pathways in human keratinocyte-derived cell lines. Arch. Dermatol. Res. 2013, 305, 817–833. [Google Scholar] [CrossRef]
  108. Marais, T.L.D.; Kluz, T.; Xu, D.; Zhang, X.; Gesumaria, L.; Matsui, M.S.; Costa, M.; Sun, H. Transcription factors and stress response gene alterations in human keratinocytes following Solar Simulated Ultra Violet Radiation. Sci. Rep. 2017, 7, 13622. [Google Scholar] [CrossRef]
  109. Assefa, Z.; Garmyn, M.; Bouillon, R.; Merlevede, W.; Vandenheede, J.R.; Agostinis, P. Differential stimulation of ERK and JNK activities by ultraviolet B irradiation and epidermal growth factor in human keratinocytes. J. Investig. Dermatol. 1997, 108, 886–891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Quan, T.; Qin, Z.; Xia, W.; Shao, Y.; Voorhees, J.J.; Fisher, G.J. Matrix-degrading metalloproteinases in photoaging. J. Investig. Dermatol. Symp. Proc. 2009, 14, 20–24. [Google Scholar] [CrossRef] [Green Version]
  111. Young, M.L.; Yeon, K.K.; Kyu, H.K.; Su, J.P.; Sung, J.K.; Jin, H.C. A novel role for the TRPV1 channel in UV-induced matrix metalloproteinase (MMP)-1 expression in HaCaT cells. J. Cell. Physiol. 2009, 219, 766–775. [Google Scholar] [CrossRef]
  112. Kim, C.; Ryu, H.C.; Kim, J.H. Low-dose UVB irradiation stimulates matrix metalloproteinase-1 expression via a BLT2-linked pathway in HaCaT cells. Exp. Mol. Med. 2010, 42, 833–841. [Google Scholar] [CrossRef] [Green Version]
  113. Dong, K.K.; Damaghi, N.; Picart, S.D.; Markova, N.G.; Obayashi, K.; Okano, Y.; Masaki, H.; Grether-Beck, S.; Krutmann, J.; Smiles, K.A.; et al. UV-induced DNA damage initiates release of MMP-1 in human skin. Exp. Dermatol. 2008, 17, 1037–1044. [Google Scholar] [CrossRef]
  114. Onoue, S.; Kobayashi, T.; Takemoto, Y.; Sasaki, I.; Shinkai, H. Induction of matrix metalloproteinase-9 secretion from human keratinocytes in culture by ultraviolet B irradiation. J. Dermatol. Sci. 2003, 33, 105–111. [Google Scholar] [CrossRef]
  115. Lee, C.H.; Wu, S.B.; Hong, C.H.; Yu, H.S.; Wei, Y.H. Molecular mechanisms of UV-induced apoptosis and its effects on skin residential cells: The implication in UV-based phototherapy. Int. J. Mol. Sci. 2013, 14, 6414–6435. [Google Scholar] [CrossRef] [Green Version]
  116. Adewale, F.O.; Basiru, A.O.; Ayorinde, O.O.; Israel, O.I.; Oluwafemi, O.A. Regulation of Apoptotic and Necroptotic Cell Death in Skin Cancer. J. Cancer Biol. Res. 2017, 5, 1108. [Google Scholar]
  117. Chen, H.; Weng, Q.Y.; Fisher, D.E. UV signaling pathways within the skin. J. Investig. Dermatol. 2014, 134, 2080–2085. [Google Scholar] [CrossRef] [Green Version]
  118. Tron, V.A.; Trotter, M.J.; Tang, L.; Krajewska, M.; Reed, J.C.; Ho, V.C.; Li, G. p53-regulated apoptosis is differentiation dependent in ultraviolet B- irradiated mouse keratinocytes. Am. J. Pathol. 1998, 153, 579–585. [Google Scholar] [CrossRef] [Green Version]
  119. Qin, J.Z.; Chaturvedi, V.; Denning, M.F.; Bacon, P.; Panella, J.; Choubey, D.; Nickoloff, B.J. Regulation of apoptosis by p53 in UV-irradiated human epidermis, psoriatic plaques and senescent keratinocytes. Oncogene 2002, 21, 2991–3002. [Google Scholar] [CrossRef] [Green Version]
  120. Holley, A.K.; St Clair, D.K. Watching the watcher: Regulation of p53 by mitochondria. Futur. Oncol. 2009, 5, 117–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Calapre, L.; Gray, E.S.; Kurdykowski, S.; David, A.; Hart, P.; Descargues, P.; Ziman, M. Heat-mediated reduction of apoptosis in UVB-damaged keratinocytes in vitro and in human skin ex vivo. BMC Dermatol. 2016, 16, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Aragane, Y.; Kulms, D.; Metze, D.; Wilkes, G.; Pöppelmann, B.; Luger, T.A.; Schwarz, T. Ultraviolet light induces apoptosis via direct activation of CD95 (Fas/APO-1) independently of its ligand CD95L. J. Cell Biol. 1998, 140, 171–182. [Google Scholar] [CrossRef] [Green Version]
  123. Lee, C.H.; Yu, C.L.; Liao, W.T.; Kao, Y.H.; Chai, C.Y.; Chen, G.S.; Yu, H.S. Effects and interactions of low doses of arsenic and UVB on keratinocyte apoptosis. Chem. Res. Toxicol. 2004, 17, 1199–1205. [Google Scholar] [CrossRef]
  124. Takasawa, R.; Nakamura, H.; Mori, T.; Tanuma, S. Differential apoptotic pathways in human keratinocyte HaCaT cells exposed to UVB and UVC. Apoptosis 2005, 10, 1121–1130. [Google Scholar] [CrossRef]
  125. Sitailo, L.A.; Tibudan, S.S.; Denning, M.F. Activation of caspase-9 is required for UV-induced apoptosis of human keratinocytes. J. Biol. Chem. 2002, 277, 19346–19352. [Google Scholar] [CrossRef] [Green Version]
  126. Assefa, Z.; Garmyn, M.; Vantieghem, A.; Declercq, W.; Vandenabeele, P.; Vandenheede, J.R.; Agostinis, P. Ultraviolet B radiation-induced apoptosis in human keratinocytes: Cytosolic activation of procaspase-8 and the role of Bcl-2. FEBS Lett. 2003, 540, 125–132. [Google Scholar] [CrossRef] [Green Version]
  127. Denning, M.F.; Wang, Y.; Tibudan, S.; Alkan, S.; Nickoloff, B.J.; Qin, J.Z. Caspase activation and disruption of mitochondrial membrane potential during UV radiation-induced apoptosis of human keratinocytes requires activation of protein kinase C. Cell Death Differ. 2002, 9, 40–52. [Google Scholar] [CrossRef]
  128. Daher, C.C.; Fontes, I.S.; De Oliveira Rodrigues, R.; Azevedo De Brito Damasceno, G.; Dos Santos Soares, D.; Flávio Soares Aragão, C.; Barreto Gomes, P.A.; Ferrari, M. Development of O/W emulsions containing euterpe oleracea extract and evaluation of photoprotective efficacy. Braz. J. Pharm. Sci. 2014, 50, 639–652. [Google Scholar] [CrossRef] [Green Version]
  129. Jarzycka, A.; Lewińska, 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. J. Photochem. Photobiol. B Biol. 2013, 128, 50–57. [Google Scholar] [CrossRef]
  130. Vijayakumar, R.; Abd Gani, S.S.; Zaidan, U.H.; Halmi, M.I.E.; Karunakaran, T.; Hamdan, M.R. Exploring the Potential Use of Hylocereus polyrhizus Peels as a Source of Cosmeceutical Sunscreen Agent for Its Antioxidant and Photoprotective Properties. Evid.-Based Complement. Altern. Med. 2020, 2020, 7520736. [Google Scholar] [CrossRef]
  131. Rocha de Carvalho, W.; Ceres Moreira, L.; Valadares, M.; Diniz, A.D.; Freitas Bara, M. Pterodon emarginatus hydroalcoholic extract: Antioxidant and photoprotective activities, noncytotoxic effect, and perspective of obtaining formulations with photochemoprotective activity. Pharmacogn. Mag. 2019, 15, 176–182. [Google Scholar] [CrossRef]
  132. Da Silva, A.C.P.; Paiva, J.P.; Diniz, R.R.; dos Anjos, V.M.; Silva, A.B.S.M.; Pinto, A.V.; dos Santos, E.P.; Leitão, A.C.; Cabral, L.M.; Rodrigues, C.R.; et al. Photoprotection assessment of olive (Olea europaea L.) leaves extract standardized to oleuropein: In vitro and in silico approach for improved sunscreens. J. Photochem. Photobiol. B Biol. 2019, 193, 162–171. [Google Scholar] [CrossRef]
  133. Era, B.; Floris, S.; Sogos, V.; Porcedda, C.; Piras, A.; Medda, R.; Fais, A.; Pintus, F. Anti-Aging Potential of Extracts from Washingtonia filifera Seeds. Plants 2021, 10, 151. [Google Scholar] [CrossRef]
  134. Surget, G.; Stiger-Pouvreau, V.; Le Lann, K.; Kervarec, N.; Couteau, C.; Coiffard, L.J.M.; Gaillard, F.; Cahier, K.; Guérard, F.; Poupart, N. Structural elucidation, in vitro antioxidant and photoprotective capacities of a purified polyphenolic-enriched fraction from a saltmarsh plant. J. Photochem. Photobiol. B Biol. 2015, 143, 52–60. [Google Scholar] [CrossRef]
  135. Cefali, L.C.; Ataide, J.A.; de Sousa, I.M.O.; Figueiredo, M.C.; Ruiz, A.L.T.G.; Foglio, M.A.; Mazzola, P.G. In vitro solar protection factor, antioxidant activity, and stability of a topical formulation containing Benitaka grape (Vitis vinifera L.) peel extract. Nat. Prod. Res. 2020, 34, 2677–2682. [Google Scholar] [CrossRef]
  136. Choquenet, B.; Couteau, C.; Paparis, E.; Coiffard, L.J.M. Quercetin and rutin as potential sunscreen agents: Determination of efficacy by an in vitro method. J. Nat. Prod. 2008, 71, 1117–1118. [Google Scholar] [CrossRef] [PubMed]
  137. Kazumy De Lima Yamaguchi, K.; Dos, L.; Santarém, S.; Lamarão, C.V.; Lima, E.S.; Florêncio Da Veiga-Junior, V. Avaliação in vitro da Atividade Fotoprotetora de Resíduos de Frutas Amazônicas. Sci. Amaz. 2016, 5, 109–116. [Google Scholar]
  138. Velasco, M.V.R.; Balogh, T.S.; Pedriali, C.A.; Sarruf, F.D.; Pinto, C.A.S.O.; Kaneko, T.M.; Baby, A.R. Rutin association with ethylhexyl methoxycinnamate and benzophenone-3: In vitro evaluation of the photoprotection effectiveness by reflectance spectrophotometry. Lat. Am. J. Pharm. 2008, 27, 23–27. [Google Scholar]
  139. Rajnochová Svobodová, A.; Gabrielová, E.; Michaelides, L.; Kosina, P.; Ryšavá, A.; Ulrichová, J.; Zálešák, B.; Vostálová, J. UVA-photoprotective potential of silymarin and silybin. Arch. Dermatol. Res. 2018, 310, 413–424. [Google Scholar] [CrossRef]
  140. Choquenet, B.; Couteau, C.; Paparis, E.; Coiffard, L.J.M. Flavonoids and polyphenols, molecular families with sunscreen potential: Determining effectiveness with an in vitro method. Nat. Prod. Commun. 2009, 4, 227–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Stevanato, R.; Bertelle, M.; Fabris, S. Photoprotective characteristics of natural antioxidant polyphenols. Regul. Toxicol. Pharmacol. 2014, 69, 71–77. [Google Scholar] [CrossRef] [PubMed]
  142. Kim, K.M.; Im, A.R.; Lee, S.; Chae, S. Dual protective effects of flavonoids from Petasites japonicus against UVB-induced apoptosis mediated via HSF-1 activated heat shock proteins and Nrf2-activated heme oxygenase-1 pathways. Biol. Pharm. Bull. 2017, 40, 765–773. [Google Scholar] [CrossRef] [Green Version]
  143. Wang, P.W.; Cheng, Y.C.; Hung, Y.C.; Lee, C.H.; Fang, J.Y.; Li, W.T.; Wu, Y.R.; Pan, T.L. Red raspberry extract protects the skin against UVB-induced damage with antioxidative and anti-inflammatory properties. Oxid. Med. Cell. Longev. 2019, 2019, 9529676. [Google Scholar] [CrossRef] [Green Version]
  144. Cerulli, A.; Masullo, M.; Mari, A.; Balato, A.; Filosa, R.; Lembo, S.; Napolitano, A.; Piacente, S. Phenolics from Castanea sativa leaves and their effects on UVB-induced damage. Nat. Prod. Res. 2018, 32, 1170–1175. [Google Scholar] [CrossRef]
  145. Xuan, S.H.; Hong, I.K.; Lee, Y.J.; Kim, J.W.; Park, S.N. Biological activities and chemical components of Potentilla kleiniana Wight & Arn. Nat. Prod. Res. 2020, 34, 3262–3266. [Google Scholar] [CrossRef] [PubMed]
  146. Kim, M.; Park, Y.G.; Lee, H.J.; Lim, S.J.; Nho, C.W. Youngiasides A and C Isolated from Youngia denticulatum Inhibit UVB-Induced MMP Expression and Promote Type I Procollagen Production via Repression of MAPK/AP-1/NF-κB and Activation of AMPK/Nrf2 in HaCaT Cells and Human Dermal Fibroblasts. J. Agric. Food Chem. 2015, 63, 5428–5438. [Google Scholar] [CrossRef]
  147. Sangiovanni, E.; Di Lorenzo, C.; Piazza, S.; Manzoni, Y.; Brunelli, C.; Fumagalli, M.; Magnavacca, A.; Martinelli, G.; Colombo, F.; Casiraghi, A.; et al. Vitis vinifera L. Leaf extract inhibits in vitro mediators of inflammation and oxidative stress involved in inflammatory-based skin diseases. Antioxidants 2019, 8, 134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Ham, S.A.; Hwang, J.S.; Kang, E.S.; Yoo, T.; Lim, H.H.; Lee, W.J.; Paek, K.S.; Seo, H.G. Ethanol extract of Dalbergia odorifera protects skin keratinocytes against ultraviolet B-induced photoaging by suppressing production of reactive oxygen species. Biosci. Biotechnol. Biochem. 2015, 79, 760–766. [Google Scholar] [CrossRef]
  149. Petruk, G.; Di Lorenzo, F.; Imbimbo, P.; Silipo, A.; Bonina, A.; Rizza, L.; Piccoli, R.; Monti, D.M.; Lanzetta, R. Protective effect of Opuntia ficus-indica L. cladodes against UVA-induced oxidative stress in normal human keratinocytes. Bioorg. Med. Chem. Lett. 2017, 27, 5485–5489. [Google Scholar] [CrossRef] [Green Version]
  150. Pérez-Sánchez, A.; Barrajón-Catalán, E.; Herranz-López, M.; Castillo, J.; Micol, V. Lemon balm extract (Melissa officinalis, L.) promotes melanogenesis and prevents UVB-induced oxidative stress and DNA damage in a skin cell model. J. Dermatol. Sci. 2016, 84, 169–177. [Google Scholar] [CrossRef] [PubMed]
  151. Ha, S.J.; Lee, J.; Kim, H.; Song, K.M.; Lee, N.H.; Kim, Y.E.; Lee, H.; Kim, Y.H.; Jung, S.K. Preventive effect of Rhus javanica extract on UVB-induced skin inflammation and photoaging. J. Funct. Foods 2016, 27, 589–599. [Google Scholar] [CrossRef]
  152. Muzaffer, U.; Paul, V.I.; Prasad, N.R.; Karthikeyan, R.; Agilan, B. Protective effect of Juglans regia L. against ultraviolet B radiation induced inflammatory responses in human epidermal keratinocytes. Phytomedicine 2018, 42, 100–111. [Google Scholar] [CrossRef] [PubMed]
  153. Hwan, J.; Youngwan, O.; Kong, C.S. Anti-photoaging effects of solvent—Partitioned fractions from Portulaca oleracea L. on UVB -stressed human keratinocytes. J. Food Biochem. 2019, 43, e12814. [Google Scholar] [CrossRef]
  154. Kwak, C.S.; Yang, J.; Shin, C.Y.; Chung, J.H. Rosa multiflora Thunb Flower Extract Attenuates Ultraviolet-Induced Photoaging in Skin Cells and Hairless Mice. J. Med. Food 2020, 23, 988–997. [Google Scholar] [CrossRef]
  155. Shiratake, S.; Nakahara, T.; Iwahashi, H.; Onodera, T.; Mizushina, Y. Rose myrtle (Rhodomyrtus tomentosa) extract and its component, piceatannol, enhance the activity of DNA polymerase and suppress the inflammatory response elicited by UVB-induced DNA damage in skin cells. Mol. Med. Rep. 2015, 12, 5857–5864. [Google Scholar] [CrossRef]
  156. De Assis Dias Alves, G.; De Souza, R.O.; Rogez, H.L.G.; Masaki, H.; Fonseca, M.J.V. Cecropia obtusa extract and chlorogenic acid exhibit anti aging effect in human fibroblasts and keratinocytes cells exposed to UV radiation. PLoS ONE 2019, 14, e0216501. [Google Scholar] [CrossRef]
  157. Wang, Y.S.; Cho, J.G.; Hwang, E.S.; Yang, J.E.; Gao, W.; Fang, M.Z.; Zheng, S.; Yi, T.H. Enhancement of Protective Effects of Radix scutellariae on UVB-induced Photo Damage in Human HaCaT Keratinocytes. Appl. Biochem. Biotechnol. 2018, 184, 1073–1093. [Google Scholar] [CrossRef]
  158. Kwon, K.R.; Alam, M.B.; Park, J.H.; Kim, T.H.; Lee, S.H. Attenuation of UVB-induced photo-aging by polyphenolic-rich Spatholobus suberectus stem extract via modulation of MAPK/AP-1/MMPs signaling in human keratinocytes. Nutrients 2019, 11, 1341. [Google Scholar] [CrossRef] [Green Version]
  159. Ahn, H.S.; Kim, H.J.; Na, C.; Jang, D.S.; Shin, Y.K.; Lee, S.H. The protective effect of Adenocaulon himalaicum Edgew. And its bioactive compound neochlorogenic acid against uvb-induced skin damage in human dermal fibroblasts and epidermal keratinocytes. Plants 2021, 10, 1669. [Google Scholar] [CrossRef]
  160. Sun, Z.W.; Du, J.; Hwang, E.; Yi, T.H. Paeonol extracted from Paeonia suffruticosa Andr. ameliorated UVB-induced skin photoaging via DLD/Nrf2/ARE and MAPK/AP-1 pathway. Phyther. Res. 2018, 32, 1741–1749. [Google Scholar] [CrossRef] [PubMed]
  161. Wongwad, E.; Pingyod, C.; Saesong, T.; Waranuch, N.; Wisuitiprot, W.; Sritularak, B.; Temkitthawon, P.; Ingkaninan, K. Assessment of the bioactive components, antioxidant, antiglycation and anti-inflammatory properties of Aquilaria crassna Pierre ex Lecomte leaves. Ind. Crops Prod. 2019, 138, 111448. [Google Scholar] [CrossRef]
  162. Razia, S.; Park, H.; Shin, E.; Shim, K.S.; Cho, E.; Kim, S.Y. Effects of Aloe vera flower extract and its active constituent isoorientin on skin moisturization via regulating involucrin expression: In vitro and molecular docking studies. Molecules 2021, 26, 2626. [Google Scholar] [CrossRef]
  163. Kim, Y.A.; Kim, D.H.; Park, C.B.; Park, T.S.; Park, B.J. Anti-inflammatory and skin-moisturizing effects of a flavonoid glycoside extracted from the aquatic plant Nymphoides indica in human keratinocytes. Molecules 2018, 23, 2342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Hwang, H.S.; Shim, J.H. Brazilin and Caesalpinia sappan L. extract protect epidermal keratinocytes from oxidative stress by inducing the expression of GPX7. Chin. J. Nat. Med. 2018, 16, 203–209. [Google Scholar] [CrossRef]
  165. Park, H.-A.; Kim, M.Y.; Lee, N.-Y.; Lim, J.; Park, K.-B.; Lee, C.-K.; Nguyen, V.D.; Kim, J.; Park, J.-T.; Park, J.-I. Variation of Triterpenic Acids in 12 Wild Syzygium formosum and Anti-Inflammation Activity on Human Keratinocyte HaCaT. Plants 2021, 10, 2428. [Google Scholar] [CrossRef]
  166. Kim, W.S.; Seo, J.H.; Lee, J.-I.; Ko, E.-S.; Cho, S.-M.; Kang, J.-R.; Jeong, J.-H.; Jeong, Y.J.; Kim, C.Y.; Cha, J.-D.; et al. The Metabolite Profile in Culture Supernatant of Aster yomena Callus and Its Anti-Photoaging Effect in Skin Cells Exposed to UVB. Plants 2021, 10, 659. [Google Scholar] [CrossRef]
  167. Oh, J.H.; Lee, J.I.; Karadeniz, F.; Park, S.Y.; Seo, Y.; Kong, C.S. Antiphotoaging Effects of 3,5-Dicaffeoyl-epi-quinic Acid via Inhibition of Matrix Metalloproteinases in UVB-Irradiated Human Keratinocytes. Evid.-Based Complement. Altern. Med. 2020, 2020, 8949272. [Google Scholar] [CrossRef]
  168. Kim, S.B.; Kim, J.E.; Kang, O.H.; Mun, S.H.; Seo, Y.S.; Kang, D.H.; Yang, D.W.; Ryu, S.Y.; Lee, Y.M.; Kwon, D.Y. Protective effect of ixerisoside A against UVB-induced pro-infammatory cytokine production in human keratinocytes. Int. J. Mol. Med. 2015, 35, 1411–1418. [Google Scholar] [CrossRef] [Green Version]
  169. Im, A.R.; Kim, Y.M.; Chin, Y.W.; Chae, S. Protective effects of compounds from Garcinia mangostana L. (mangosteen) against UVB damage in HaCaT cells and hairless mice. Int. J. Mol. Med. 2017, 40, 1941–1949. [Google Scholar] [CrossRef] [Green Version]
  170. Lin, K.W.; Wang, B.W.; Wu, C.M.; Yen, M.H.; Wei, B.L.; Hung, C.F.; Lin, C.N. Antioxidant prenylated phenols of Artocarpus plants attenuate ultraviolet radiation-induced damage on human keratinocytes and fibroblasts. Phytochem. Lett. 2015, 14, 190–197. [Google Scholar] [CrossRef]
  171. Landén, N.X.; Li, D.; Ståhle, M. Transition from inflammation to proliferation: A critical step during wound healing. Cell. Mol. Life Sci. 2016, 73, 3861–3885. [Google Scholar] [CrossRef] [Green Version]
  172. Barrientos, S.; Stojadinovic, O.; Golinko, M.S.; Brem, H.; Tomic-Canic, M. Growth factors and cytokines in wound healing. Wound Repair Regen. 2008, 16, 585–601. [Google Scholar] [CrossRef]
  173. Ridiandries, A.; Tan, J.T.M.; Bursill, C.A. The role of chemokines in wound healing. Int. J. Mol. Sci. 2018, 19, 3217. [Google Scholar] [CrossRef] [Green Version]
  174. Wojtowicz, A.M.; Oliveira, S.; Carlson, M.W.; Zawadzka, A.; Rousseau, C.F.; Baksh, D. The importance of both fibroblasts and keratinocytes in a bilayered living cellular construct used in wound healing. Wound Repair Regen. 2014, 22, 246–255. [Google Scholar] [CrossRef] [Green Version]
  175. Coulombe, P.A. Towards a molecular definition of keratinocyte activation after acute injury to stratified epithelia. Biochem. Biophys. Res. Commun. 1997, 236, 231–238. [Google Scholar] [CrossRef]
  176. Pora, A.; Yoon, S.; Dreissen, G.; Hoffmann, B.; Merkel, R.; Windoffer, R.; Leube, R.E. Regulation of keratin network dynamics by the mechanical properties of the environment in migrating cells. Sci. Rep. 2020, 10, 4574. [Google Scholar] [CrossRef] [Green Version]
  177. Wallis, S.; Lloyd, S.; Wise, I.; Ireland, G.; Fleming, T.P.; Garrod, D. The α isoform of protein kinase C is involved in signaling the response of desmosomes to wounding in cultured epithelial cells. Mol. Biol. Cell. 2000, 11, 1077–1092. [Google Scholar] [CrossRef]
  178. Savagner, P.; Kusewitt, D.F.; Carver, E.A.; Magnino, F.; Choi, C.; Gridley, T.; Hudson, L.G. Developmental transcription factor slug is required for effective re-epithelialization by adult keratinocytes. J. Cell. Physiol. 2005, 202, 858–866. [Google Scholar] [CrossRef] [Green Version]
  179. Choi, Y.J.; Laclef, C.; Yang, N.; Andreu-Cervera, A.; Lewis, J.; Mao, X.; Li, L.; Snedecor, E.R.; Takemaru, K.I.; Qin, C.; et al. RPGRIP1L is required for stabilizing epidermal keratinocyte adhesion through regulating desmoglein endocytosis. PLoS Genet. 2019, 15, e1007914. [Google Scholar] [CrossRef]
  180. Sumigray, K.; Zhou, K.; Lechler, T. Cell-cell adhesions and cell contractility are upregulated upon desmosome disruption. PLoS ONE 2014, 9, e101824. [Google Scholar] [CrossRef] [Green Version]
  181. Völlner, F.; Ali, J.; Kurrle, N.; Exner, Y.; Eming, R.; Hertl, M.; Banning, A.; Tikkanen, R. Loss of flotillin expression results in weakened desmosomal adhesion and Pemphigus vulgaris-like localisation of desmoglein-3 in human keratinocytes. Sci. Rep. 2016, 6, 28820. [Google Scholar] [CrossRef]
  182. Amagai, M.; Fujimori, T.; Masunaga, T.; Shimizu, H.; Nishikawa, T.; Shimizu, N.; Takeichi, M.; Hashimoto, T. Delayed assembly of desmosomes in keratinocytes with disrupted classic-cadherin-mediated cell adhesion by a dominant negative mutant. J. Investig. Dermatol. 1995, 104, 27–32. [Google Scholar] [CrossRef] [Green Version]
  183. Wanuske, M.T.; Brantschen, D.; Schinner, C.; Stüdle, C.; Walter, E.; Hiermaier, M.; Vielmuth, F.; Waschke, J.; Spindler, V. Clustering of desmosomal cadherins by desmoplakin is essential for cell-cell adhesion. Acta Physiol. 2021, 231, e13609. [Google Scholar] [CrossRef] [PubMed]
  184. Bodin, S.; Planchon, D.; Morris, E.R.; Comunale, F.; Gauthier-Rouviére, C. Flotillins in intercellular adhesion—From cellular physiology to human diseases. J. Cell Sci. 2014, 127, 5139–5147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Litjens, S.H.M.; de Pereda, J.M.; Sonnenberg, A. Current insights into the formation and breakdown of hemidesmosomes. Trends Cell Biol. 2006, 16, 376–383. [Google Scholar] [CrossRef]
  186. Miranti, C.K.; Brugge, J.S. Sensing the environment: A historical perspective on integrin signal transduction. Nat. Cell Biol. 2002, 4, E83–E90. [Google Scholar] [CrossRef]
  187. Niculescu, C.; Ganguli-Indra, G.; Pfister, V.; Dupé, V.; Messaddeq, N.; De Arcangelis, A.; Georges-Labouesse, E. Conditional ablation of integrin alpha-6 in mouse epidermis leads to skin fragility and inflammation. Eur. J. Cell Biol. 2011, 90, 270–277. [Google Scholar] [CrossRef]
  188. Nguyen, B.P.; Ryan, M.C.; Gil, S.G.; Carter, W.G. Deposition of laminin 5 in epidermal wounds regulates integrin signaling and adhesion. Curr. Opin. Cell Biol. 2000, 12, 554–562. [Google Scholar] [CrossRef]
  189. Wilhelmsen, K.; Litjens, S.H.M.; Kuikman, I.; Margadant, C.; Van Rheenen, J.; Sonnenberg, A. Serine phosphorylation of the integrin β4 subunit is necessary for epidermal growth factor receptor-induced hemidesmosome disruption. Mol. Biol. Cell. 2007, 18, 3512–3522. [Google Scholar] [CrossRef]
  190. Santoro, M.M.; Gaudino, G.; Marchisio, P.C. The MSP receptor regulates alpha6beta4 and alpha3beta1 integrins via 14-3-3 proteins in keratinocyte migration. Dev. Cell. 2003, 5, 257–271. [Google Scholar] [CrossRef] [Green Version]
  191. Seeger, M.A.; Paller, A.S. The Roles of Growth Factors in Keratinocyte Migration. Adv. Wound Care 2015, 4, 213–224. [Google Scholar] [CrossRef] [Green Version]
  192. Sivamani, R.K.; Garcia, M.S.; Rivkah Isseroff, R. Wound re-epithelialization: Modulating keratinocyte migration in wound healing. Front. Biosci. 2007, 12, 2849–2868. [Google Scholar] [CrossRef] [Green Version]
  193. Gniadecki, R. Regulation of keratinocyte proliferation. Gen. Pharmacol. 1998, 30, 619–622. [Google Scholar] [CrossRef]
  194. Ando, Y.; Jensen, P.J. Epidermal growth factor and insulin-like growth factor I enhance keratinocyte migration. J. Investig. Dermatol. 1993, 100, 633–639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Liu, Y.; Petreaca, M.; Yao, M.; Martins-Green, M. Cell and molecular mechanisms of keratinocyte function stimulated by insulin during wound healing. BMC Cell Biol. 2009, 10, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Aijaz, A.; Faulknor, R.; Berthiaume, F.; Olabisi, R.M. Hydrogel Microencapsulated Insulin-Secreting Cells Increase Keratinocyte Migration, Epidermal Thickness, Collagen Fiber Density, and Wound Closure in a Diabetic Mouse Model of Wound Healing. Tissue Eng. Part A 2015, 21, 2723–2732. [Google Scholar] [CrossRef] [Green Version]
  197. Haase, I.; Evans, R.; Pofahl, R.; Watt, F.M. Regulation of keratinocyte shape, migration and wound epithelialization by IGF-1 and EGF-dependent signalling pathways. J. Cell Sci. 2003, 116, 3227–3238. [Google Scholar] [CrossRef] [Green Version]
  198. Marikovsky, M.; Vogt, P.; Eriksson, E.; Rubin, J.S.; Taylor, W.G.; Sasse, J.; Klagsbrun, M. Wound fluid-derived heparin-binding EGF-like growth factor (HB-EGF) is synergistic with insulin-like growth factor-I for Balb/MK keratinocyte proliferation. J. Investig. Dermatol. 1996, 106, 616–621. [Google Scholar] [CrossRef] [Green Version]
  199. Meyer, M.; Müller, A.K.; Yang, J.; Moik, D.; Ponzio, G.; Ornitz, D.M.; Grose, R.; Werner, S. FGF receptors 1 and 2 are key regulators of keratinocyte migration in vitro and in wounded skin. J. Cell Sci. 2012, 125, 5690–5701. [Google Scholar] [CrossRef] [Green Version]
  200. Peng, C.; Chen, B.; Kao, H.K.; Murphy, G.; Orgill, D.P.; Guo, L. Lack of FGF-7 further delays cutaneous wound healing in diabetic mice. Plast. Reconstr. Surg. 2011, 128, 673e–684e. [Google Scholar] [CrossRef]
  201. Radek, K.A.; Taylor, K.R.; Gallo, R.L. FGF-10 and specific structural elements of dermatan sulfate size and sulfation promote maximal keratinocyte migration and cellular proliferation. Wound Repair Regen. 2009, 17, 118–126. [Google Scholar] [CrossRef] [Green Version]
  202. Viac, J.; Palacio, S.; Schmitt, D.; Claudy, A. Expression of vascular endothelial growth factor in normal epidermis, epithelial tumors and cultured keratinocytes. Arch. Dermatol. Res. 1997, 289, 158–163. [Google Scholar] [CrossRef]
  203. Mann, A.; Breuhahn, K.; Schirmacher, P.; Blessing, M. Keratinocyte-derived granulocyte-macrophage colony stimulating factor accelerates wound healing: Stimulation of keratinocyte proliferation, granulation tissue formation, and vascularization. J. Investig. Dermatol. 2001, 117, 1382–1390. [Google Scholar] [CrossRef] [Green Version]
  204. Oike, Y.; Yasunaga, K.; Ito, Y.; Matsumoto, S.; Maekawa, H.; Morisada, T.; Arai, F.; Nakagata, N.; Takeya, M.; Masuho, Y.; et al. Angiopoietin-related growth factor (AGF) promotes epidermal proliferation, remodeling, and regeneration. Proc. Natl. Acad. Sci. USA 2003, 100, 9494–9499. [Google Scholar] [CrossRef] [Green Version]
  205. Ranzato, E.; Patrone, M.; Pedrazzi, M.; Burlando, B. HMGb1 promotes scratch wound closure of HaCaT keratinocytes via ERK1/2 activation. Mol. Cell. Biochem. 2009, 332, 199–205. [Google Scholar] [CrossRef]
  206. Straino, S.; Di Carlo, A.; Mangoni, A.; De Mori, R.; Guerra, L.; Maurelli, R.; Panacchia, L.; Di Giacomo, F.; Palumbo, R.; Di Campli, C.; et al. High-mobility group box 1 protein in human and murine skin: Involvement in wound healing. J. Investig. Dermatol. 2008, 128, 1545–1553. [Google Scholar] [CrossRef] [PubMed]
  207. Woodley, D.T.; Wysong, A.; DeClerck, B.; Chen, M.; Li, W. Keratinocyte Migration and a Hypothetical New Role for Extracellular Heat Shock Protein 90 Alpha in Orchestrating Skin Wound Healing. Adv. Wound Care 2015, 4, 203–212. [Google Scholar] [CrossRef] [Green Version]
  208. Guo, J.; Chang, C.; Li, W. The role of secreted heat shock protein-90 (Hsp90) in wound healing—How could it shape future therapeutics? Expert Rev. Proteom. 2017, 14, 665–675. [Google Scholar] [CrossRef]
  209. Cheng, C.-F.; Fan, J.; Fedesco, M.; Guan, S.; Li, Y.; Bandyopadhyay, B.; Bright, A.M.; Yerushalmi, D.; Liang, M.; Chen, M.; et al. Transforming Growth Factor α (TGFα)-Stimulated Secretion of HSP90α: Using the Receptor LRP-1/CD91 To Promote Human Skin Cell Migration against a TGFβ-Rich Environment during Wound Healing. Mol. Cell. Biol. 2008, 28, 3344–3358. [Google Scholar] [CrossRef] [Green Version]
  210. Zhang, Y.; Bai, X.; Wang, Y.; Li, N.; Li, X.; Han, F.; Su, L.; Hu, D. Role for heat shock protein 90α in the proliferation and migration of HaCaT cells and in the deep second-degree burn wound healing in mice. PLoS ONE 2014, 9, e103723. [Google Scholar] [CrossRef] [PubMed]
  211. Tang, A.; Gilchrest, B.A. Regulation of keratinocyte growth factor gene expression in human skin fibroblasts. J. Dermatol. Sci. 1996, 11, 41–50. [Google Scholar] [CrossRef]
  212. Gallucci, R.M.; Sloan, D.K.; Heck, J.M.; Murray, A.R.; O’Dell, S.J. Interleukin 6 indirectly induces keratinocyte migration. J. Investig. Dermatol. 2004, 122, 764–772. [Google Scholar] [CrossRef] [Green Version]
  213. Devalaraja, R.M.; Nanney, L.B.; Qian, Q.; Du, J.; Yu, Y.; Devalaraja, M.N.; Richmond, A. Delayed wound healing in CXCR 2 knockout mice. J. Investig. Dermatol. 2000, 115, 234–244. [Google Scholar] [CrossRef] [Green Version]
  214. Yates, C.C.; Whaley, D.; Hooda, S.; Hebda, P.A.; Bodnar, R.J.; Wells, A. Delayed reepithelialization and basement membrane regeneration after wounding in mice lacking CXCR3. Wound Repair Regen. 2009, 17, 34–41. [Google Scholar] [CrossRef] [Green Version]
  215. Tortelli, F.; Pisano, M.; Briquez, P.S.; Martino, M.M.; Hubbell, J.A. Fibronectin Binding Modulates CXCL11 Activity and Facilitates Wound Healing. PLoS ONE 2013, 8, e79610. [Google Scholar] [CrossRef] [Green Version]
  216. Iocono, J.A.; Colleran, K.R.; Remick, D.G.; Gillespie, B.W.; Ehrlich, H.P.; Garner, W.L. Interleukin-8 levels and activity in delayed-healing human thermal wounds. Wound Repair Regen. 2000, 8, 216–225. [Google Scholar] [CrossRef]
  217. Da Silva, L.; Carvalho, E.; Cruz, M.T. Role of neuropeptides in skin inflammation and its involvement in diabetic wound healing. Expert Opin. Biol. Ther. 2010, 10, 1427–1439. [Google Scholar] [CrossRef]
  218. Roggenkamp, D.; Köpnick, S.; Stäb, F.; Wenck, H.; Schmelz, M.; Neufang, G. Epidermal nerve fibers modulate keratinocyte growth via neuropeptide signaling in an innervated skin model. J. Investig. Dermatol. 2013, 133, 1620–1628. [Google Scholar] [CrossRef] [Green Version]
  219. Wollina, U.; Huschenbeck, J.; Knöll, B.; Sternberg, B.; Hipler, U.C. Vasoactive intestinal peptide supports induced migration of human keratinocytes and their colonization of an artificial polyurethane matrix. Regul. Pept. 1997, 70, 29–36. [Google Scholar] [CrossRef]
  220. Sung, K.J.; Chang, S.E.; Paik, E.M.; Lee, M.W.; Choi, J.H. Vasoactive intestinal polypeptide stimulates the proliferation of HaCat cell via TGF-α. Neuropeptides 1999, 33, 435–446. [Google Scholar] [CrossRef]
  221. Kakurai, M.; Demitsu, T.; Umemoto, N.; Kobayashi, Y.; Inoue-Narita, T.; Fujita, N.; Ohtsuki, M.; Furukawa, Y. Vasoactive intestinal peptide and inflammatory cytokines enhance vascular endothelial growth factor production from epidermal keratinocytes. Br. J. Dermatol. 2009, 161, 1232–1238. [Google Scholar] [CrossRef]
  222. Altun, V.; Hakvoort, T.E.; van Zuijlen, P.P.M.; van der Kwast, T.H.; Prens, E.P. Nerve outgrowth and neuropeptide expression during the remodeling of human burn wound scars. Burns 2001, 27, 717–722. [Google Scholar] [CrossRef]
  223. Delgado, A.V.; McManus, A.T.; Chambers, J.P. Exogenous administration of substance P enhances wound healing in a novel skin-injury model. Exp. Biol. Med. 2005, 230, 271–280. [Google Scholar] [CrossRef]
  224. Gibran, N.S.; Tamura, R.; Tsou, R.; Isik, F.F. Human dermal microvascular endothelial cells produce nerve growth factor: Implications for wound repair. Shock 2003, 19, 127–130. [Google Scholar] [CrossRef] [Green Version]
  225. McGovern, U.B.; Jones, K.T.; Sharpe, G.R. Intracellular calcium as a second messenger following growth stimulation of human keratinocytes. Br. J. Dermatol. 1995, 132, 892–896. [Google Scholar] [CrossRef]
  226. Tanaka, T.; Danno, K.; Ikai, K.; Imamura, S. Effects of substance P and substance K on the growth of cultured keratinocytes. J. Investig. Dermatol. 1988, 90, 399–401. [Google Scholar] [CrossRef] [Green Version]
  227. Shi, X.; Wang, L.; Clark, J.D.; Kingery, W.S. Keratinocytes express cytokines and nerve growth factor in response to neuropeptide activation of the ERK1/2 and JNK MAPK transcription pathways. Regul. Pept. 2013, 186, 92–103. [Google Scholar] [CrossRef] [Green Version]
  228. Kumar, S.; Tan, Y.; Berthiaume, F. Neuropeptide substance p enhances skin wound healing in vitro and in vivo under hypoxia. Biomedicines 2021, 9, 222. [Google Scholar] [CrossRef]
  229. Gibran, N.S.; Jang, Y.C.; Isik, F.F.; Greenhalgh, D.G.; Muffley, L.A.; Underwood, R.A.; Usui, M.L.; Larsen, J.; Smith, D.G.; Bunnett, N.; et al. Diminished neuropeptide levels contribute to the impaired cutaneous healing response associated with diabetes mellitus. J. Surg. Res. 2002, 108, 122–128. [Google Scholar] [CrossRef]
  230. Spenny, M.L.; Muangman, P.; Sullivan, S.R.; Bunnett, N.W.; Ansel, J.C.; Olerud, J.E.; Gibran, N.S. Neutral endopeptidase inhibition in diabetic wound repair. Wound Repair Regen. 2002, 10, 295–301. [Google Scholar] [CrossRef]
  231. Moura, L.I.; Cruz, M.T.; Carvalho, E. The effect of neurotensin in human keratinocytes—Implication on impaired wound healing in diabetes. Exp. Biol. Med. 2014, 239, 6–12. [Google Scholar] [CrossRef]
  232. Dallos, A.; Kiss, M.; Polyánka, H.; Dobozy, A.; Kemény, L.; Husz, S. Effects of the neuropeptides substance P, calcitonin gene-related peptide, vasoactive intestinal polypeptide and galanin on the production of nerve growth factor and inflammatory cytokines in cultured human keratinocytes. Neuropeptides 2006, 40, 251–263. [Google Scholar] [CrossRef]
  233. Dallos, A.; Kiss, M.; Polyánka, H.; Dobozy, A.; Kemény, L.; Husz, S. Galanin receptor expression in cultured human keratinocytes and in normal human skin. J. Peripher. Nerv. Syst. 2006, 11, 156–164. [Google Scholar] [CrossRef]
  234. Bigliardi, P.L.; Büchner, S.; Rufli, T.; Bigliardi-Qi, M. Specific stimulation of migration of human keratinocytes by μ-opiate receptor agonists. J. Recept. Signal Transduct. Res. 2002, 22, 191–199. [Google Scholar] [CrossRef]
  235. Chernyavsky, A.I.; Arrendondo, J.; Marubio, L.M.; Grando, S.A. Differential regulation of keratinocyte chemokinesis and chemotaxis through distinct nicotinic receptor subtypes. J. Cell Sci. 2004, 117, 5665–5679. [Google Scholar] [CrossRef] [Green Version]
  236. Chernyavsky, A.I.; Arredondo, J.; Vetter, D.E.; Grando, S.A. Central role of α9 acetylcholine receptor in coordinating keratinocyte adhesion and motility at the initiation of epithelialization. Exp. Cell Res. 2007, 313, 3542–3555. [Google Scholar] [CrossRef] [Green Version]
  237. Chernyavsky, A.I.; Arredondo, J.; Wess, J.; Karlsson, E.; Grando, S.A. Novel signaling pathways mediating reciprocal control of keratinocyte migration and wound epithelialization through M3 and M4 muscarinic receptors. J. Cell Biol. 2004, 166, 261–272. [Google Scholar] [CrossRef] [Green Version]
  238. Sudbeck, B.D.; Pilcher, B.K.; Welgus, H.G.; Parks, W.C. Induction and repression of collagenase-1 by keratinocytes is controlled by distinct components of different extracellular matrix compartments. J. Biol. Chem. 1997, 272, 22103–22110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Varani, J.; Perone, P.; Deming, M.O.B.; Warner, R.L.; Aslam, M.N.; Bhagavathula, N.; Dame, M.K.; Voorhees, J.J. Impaired keratinocyte function on matrix metalloproteinase-1 (MMP-1) damaged collagen. Arch. Dermatol. Res. 2009, 301, 497–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  240. Imai, K.; Hiramatsu, A.; Fukushima, D.; Pierschbacher, M.D.; Okada, Y. Degradation of decorin by matrix metalloproteinases: Identification of the cleavage sites, kinetic analyses and transforming growth factor-β1 release. Biochem. J. 1997, 322, 809–814. [Google Scholar] [CrossRef]
  241. Ruttanapattanakul, J.; Wikan, N.; Okonogi, S.; Na Takuathung, M.; Buacheen, P.; Pitchakarn, P.; Potikanond, S.; Nimlamool, W. Boesenbergia rotunda extract accelerates human keratinocyte proliferation through activating ERK1/2 and PI3K/Akt kinases. Biomed. Pharmacother. 2021, 133, 111002. [Google Scholar] [CrossRef]
  242. Ziemlewska, A.; Zagórska-Dziok, M.; Nizioł-Łukaszewska, Z. Assessment of cytotoxicity and antioxidant properties of berry leaves as by-products with potential application in cosmetic and pharmaceutical products. Sci. Rep. 2021, 11, 3240. [Google Scholar] [CrossRef]
  243. Muniandy, K.; Gothai, S.; Tan, W.S.; Kumar, S.S.; Mohd Esa, N.; Chandramohan, G.; Al-Numair, K.S.; Arulselvan, P. In Vitro Wound Healing Potential of Stem Extract of Alternanthera sessilis. Evid.-Based Complement. Altern. Med. 2018, 2018, 142073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Di Martino, O.; Tito, A.; De Lucia, A.; Cimmino, A.; Cicotti, F.; Apone, F.; Colucci, G.; Calabrò, V. Hibiscus syriacus Extract from an Established Cell Culture Stimulates Skin Wound Healing. Biomed. Res. Int. 2017, 2017, 7932019. [Google Scholar] [CrossRef] [Green Version]
  245. Park, S.M.; Won, K.J.; Hwang, D.I.; Kim, D.Y.; Kim, H.B.; Li, Y.; Lee, H.M. Potential Beneficial Effects of Digitaria ciliaris Flower Absolute on the Wound Healing-Linked Activities of Fibroblasts and Keratinocytes. Planta Med. 2020, 86, 348–355. [Google Scholar] [CrossRef] [PubMed]
  246. Csepregi, R.; Temesfői, V.; Das, S.; Alberti, Á.; Tóth, C.A.; Herczeg, R.; Papp, N.; Kőszegi, T. Cytotoxic, antimicrobial, antioxidant properties and effects on cell migration of phenolic compounds of selected transylvanian medicinal plants. Antioxidants 2020, 9, 166. [Google Scholar] [CrossRef] [Green Version]
  247. Paudel, S.B.; Park, J.; Kim, N.H.; Choi, H.; Seo, E.K.; Woo, H.A.; Nam, J.W. Constituents of the leaves and twigs of Elaeagnus umbellata and their proliferative effects on human keratinocyte HaCaT cells. Fitoterapia 2019, 139, 104374. [Google Scholar] [CrossRef]
  248. Prado, L.G.; Arruda, H.S.; Peixoto Araujo, N.M.; de Oliveira Braga, L.E.; Banzato, T.P.; Pereira, G.A.; Figueiredo, M.C.; Ruiz, A.L.T.G.; Eberlin, M.N.; de Carvalho, J.E.; et al. Antioxidant, antiproliferative and healing properties of araticum (Annona crassiflora Mart.) peel and seed. Food Res. Int. 2020, 133, 109168. [Google Scholar] [CrossRef]
  249. Kisseih, E.; Lechtenberg, M.; Petereit, F.; Sendker, J.; Zacharski, D.; Brandt, S.; Agyare, C.; Hensel, A. Phytochemical characterization and in vitro wound healing activity of leaf extracts from Combretum mucronatum Schum. & Thonn.: Oligomeric procyanidins as strong inductors of cellular differentiation. J. Ethnopharmacol. 2015, 174, 628–636. [Google Scholar] [CrossRef] [PubMed]
  250. Dorjsembe, B.; Lee, H.J.; Kim, M.; Dulamjav, B.; Jigjid, T.; Nho, C.W. Achillea asiatica extract and its active compounds induce cutaneous wound healing. J. Ethnopharmacol. 2017, 206, 306–314. [Google Scholar] [CrossRef]
  251. Chin, C.Y.; Jalil, J.; Ng, P.Y.; Ng, S.F. Development and formulation of Moringa oleifera standardised leaf extract film dressing for wound healing application. J. Ethnopharmacol. 2018, 212, 188–199. [Google Scholar] [CrossRef] [PubMed]
  252. De Moura Sperotto, N.D.; Steffens, L.; Veríssimo, R.M.; Henn, J.G.; Péres, V.F.; Vianna, P.; Chies, J.A.B.; Roehe, A.; Saffi, J.; Moura, D.J. Wound healing and anti-inflammatory activities induced by a Plantago australis hydroethanolic extract standardized in verbascoside. J. Ethnopharmacol. 2018, 225, 178–188. [Google Scholar] [CrossRef]
  253. Azmi, L.; Shukla, I.; Goutam, A.; Rao, C.V.; Jawaid, T.; Kamal, M.; Awaad, A.S.; Alqasoumi, S.I.; AlKhamees, O.A. In vitro wound healing activity of 1-hydroxy-5,7-dimethoxy-2-naphthalene-carboxaldehyde (HDNC)and other isolates of Aegle marmelos L.: Enhances keratinocytes motility via Wnt/β-catenin and RAS-ERK pathways. Saudi Pharm. J. 2019, 27, 532–539. [Google Scholar] [CrossRef]
  254. Juneja, K.; Mishra, R.; Chauhan, S.; Gupta, S.; Roy, P.; Sircar, D. Metabolite profiling and wound-healing activity of Boerhavia diffusa leaf extracts using in vitro and in vivo models. J. Tradit. Complement. Med. 2020, 10, 52–59. [Google Scholar] [CrossRef]
  255. Mazumdar, S.; Ghosh, A.K.; Dinda, M.; Das, A.K.; Das, S.; Jana, K.; Karmakar, P. Evaluation of wound healing activity of ethanol extract of Annona reticulata L. leaf both in vitro and in diabetic mice model. J. Tradit. Complement. Med. 2021, 11, 27–37. [Google Scholar] [CrossRef] [PubMed]
  256. Azis, H.A.; Taher, M.; Ahmed, A.S.; Sulaiman, W.M.A.W.; Susanti, D.; Chowdhury, S.R.; Zakaria, Z.A. In vitro and In vivo wound healing studies of methanolic fraction of Centella asiatica extract. S. Afr. J. Bot. 2017, 108, 163–174. [Google Scholar] [CrossRef]
  257. Petpiroon, N.; Suktap, C.; Pongsamart, S.; Chanvorachote, P.; Sukrong, S. Kaempferol-3-O-rutinoside from Afgekia mahidoliae promotes keratinocyte migration through FAK and Rac1 activation. J. Nat. Med. 2015, 69, 340–348. [Google Scholar] [CrossRef]
  258. Moriyama, M.; Moriyama, H.; Uda, J.; Kubo, H.; Nakajima, Y.; Goto, A.; Akaki, J.; Yoshida, I.; Matsuoka, N.; Hayakawa, T. Beneficial effects of the genus Aloe on wound healing, cell proliferation, and differentiation of epidermal keratinocytes. PLoS ONE 2016, 11, e0164799. [Google Scholar] [CrossRef] [PubMed]
  259. Girija, D.M.; Kalachaveedu, M.; Subbarayan, R.; Jenifer, P.; Rao, S.R. Aristolochia bracteolata enhances wound healing in vitro through anti-inflammatory and proliferative effect on human dermal fibroblasts and keratinocytes. Pharmacogn. J. 2017, 9, s129–s136. [Google Scholar] [CrossRef] [Green Version]
  260. Kim, M.; Lee, H.J.; Randy, A.; Yun, J.H.; Oh, S.R.; Nho, C.W. Stellera chamaejasme and its constituents induce cutaneous wound healing and anti-inflammatory activities. Sci. Rep. 2017, 7, 42490. [Google Scholar] [CrossRef] [Green Version]
  261. Seo, S.H.; Lee, S.H.; Cha, P.H.; Kim, M.Y.; Min, D.S.; Choi, K.Y. Polygonum aviculare L. And its active compounds, quercitrin hydrate, caffeic acid, and rutin, activate the Wnt/β-catenin pathway and induce cutaneous wound healing. Phyther. Res. 2016, 30, 848–854. [Google Scholar] [CrossRef] [PubMed]
  262. Bridi, H.; Beckenkamp, A.; Ccana-Ccapatinta, G.V.; de Loreto Bordignon, S.A.; Buffon, A.; von Poser, G.L. Characterization of Phloroglucinol-enriched Fractions of Brazilian Hypericum Species and Evaluation of Their Effect on Human Keratinocytes Proliferation. Phyther. Res. 2017, 31, 62–68. [Google Scholar] [CrossRef] [Green Version]
  263. Évora, A.; De Freitas, V.; Mateus, N.; Fernandes, I. The effect of anthocyanins from red wine and blackberry on the integrity of a keratinocyte model using ECIS. Food Funct. 2017, 8, 3989–3998. [Google Scholar] [CrossRef]
  264. Moghadam, S.E.; Ebrahimi, S.N.; Salehi, P.; Farimani, M.M.; Hamburger, M.; Jabbarzadeh, E. Wound healing potential of chlorogenic acid and myricetin-3-o-β-rhamnoside isolated from Parrotia persica. Molecules 2017, 22, 1501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. Selected plant extracts from different species with identified compounds and their in vitro effect on H2O2-stimulated human keratinocytes.
Table 1. Selected plant extracts from different species with identified compounds and their in vitro effect on H2O2-stimulated human keratinocytes.
Name of the Species/FamilyPart of the PlantType of ExtractCell LineIdentified CompoundsMechanism of ActionEffectsRef.
Andrographis paniculata (Burm.f.) Nees
(Acanthaceae)
leavesmethanolicHaCaT stimulated by hydrogen peroxideandrographolideInhibition: ROS
production
antioxidant[37]
Acer rubrum L.
(Sapindaceae)
leavesMaplifaTMHaCaT stimulated by hydrogen peroxide and methylglyoxalginnalin AInduction: cell viability
Inhibition: ROS production, caspases-3/7 and -8 release
antioxidant
cytoprotective
anti-apoptotic
[38]
Moringa oleifera Lam. (Moringaceae)stemethanolicHaCaT stimulated by hydrogen peroxideluteolin, rutin, quercetinInduction: SOD and CAT, activation of PPARα
Inhibition: ROS production
antioxidant[39]
Lagerstroemia speciosa (L.) Pers. and Lagerstroemia floribunda Jack
(Lythraceae)
flowersethanolicHaCaT stimulated by hydrogen peroxideellagic acid, epicatechin gallate, and quercetinInhibition: hydrogen peroxide-induced cell deathantioxidant
anti-apoptotic
[40]
Punica granatum L.
(Lythraceae)
fruitsPomella®HaCaT stimulated by hydrogen peroxidepunicalagins, ellagic acid and urolithin AInhibition: ROS production, apoptotic cells formation, caspase 3 productionanti-oxidant
anti-apoptotic
[41]
Castanea sativa Mill.
(Fagaceae)
chestnut shells, and inner chestnut shellsaqueousHaCaT stimulated by hydrogen peroxidegallic acidInhibition: oxidized lipids, NO and
iNOS production, collagen degradation
cytoprotective
antioxidant
[42]
Oenothera biennis L.
(Onagraceae)
aerial partsethanolicHaCaT stimulated by hydrogen peroxide3-caeoylquinic acid, ellagic acid, and quercetin 3-O-glucuronide, quercetinInduction: cell viability, heme oxygenase-1 (HO-1)
Inhibition: DNA damages, caspase-3, PARP
cytoprotective
antioxidant
anti-apoptotic
[43]
Himantoglossum robertianum (Loisel.) P.Delforge (Orchidaceae)flowersethanolicHaCaT stimulated by hydrogen peroxideflavones and flavan-3-ols, scopoletin, and phenolic acidsInduction: cell viability and motility
Inhibition: elastase and collagenase
cytoprotective
antioxidant
stimulate migration
[44]
Myrciaria dubia (Kunth) McVaugh (Myrtaceae)fruitethanolicHaCaT stimulated by high glucoseellagic acid and quercetinInduction: Nrf2
Inhibition: MAPK/AP-1, NF-κB
antioxidant
anti-inflammatory
[45]
Clitoria ternatea L.
(Fabaceae)
flowersaqueousHaCaT stimulated by hydrogen peroxideanthocyanins derived from delphinidin, including polyacylated ternatins, and flavonol glycosides derived from quercetin and kaempferolInhibition: cytotoxicity effects of H2O2antioxidant[46]
Table 2. Selected plant extracts from different species with identified compounds and their in vitro effect on TNF-α or IFN-γ-stimulated human keratinocytes.
Table 2. Selected plant extracts from different species with identified compounds and their in vitro effect on TNF-α or IFN-γ-stimulated human keratinocytes.
Name of the Species/FamilyPart of the PlantType of ExtractCell LineIdentified CompoundsMechanism of ActionEffectsRef.
Rydingia persica (Burm.f.) Scheen & V.A.Albert
(Lamiaceae)
aerial partsmethanolicHaCaT stimulated by LPSlabdane-type diterpenoidsInhibition: IL-6 and TNF-α releaseanti-inflammatory[78]
Andrographis paniculata (Burm.f.) Nees
(Acanthaceae)
leavesmethanolicHaCaT stimulated by LPS/TNF-αandrographolideInduction: IL-8 secretion
Inhibition: TNF-α expression
anti-inflammatory[37]
Curcuma aromatica Salisb. (Zingiberaceae)rhizomeethanolicHaCaT stimulated by TNF-αgermacrone, curdione, dehydrocurdione, zederone, curcumenol, curcuminInhibition: NF-κB activationanti-inflammatory[79]
Crateva adansonii DC. (Capparaceae)leavesaqueousHPEKs infected by Staphylococcus aureusquercitrin, isoquercitrin, quercetin-3-O-(b-d-xylopyranosyl-a-l-rhamnopyranoside)Inhibition: IL-6, IL-8 and TNFα expressionanti-inflammatory[80]
Perilla frutescens var. crispa (Thunb.) H.Deane (Lamiaceae)leavesethanolicHaCaT stimulated by TNF-α/IFN-γcaffeic acid, rosmarinic acid, luteolinInhibition: p38, ERK, and JNK expression; STAT-1 and NK-κB activationanti-inflammatory[81]
Rhus coriaria L.
(Anacardiaceae)
fruitsethanolicHaCaT stimulated by TNF-αrutin, quercetin derivative, gallotanninsInhibition: NF-κB activation; ICAM-1, and MMP-9 secretionanti-inflammatory[82]
Ampelopsis glandulosa (Wall.) Momiy.
(Vitaceae)
rhizomeethanolicHaCaT stimulated by TNF-α/IFN-γbetulin, betulinic acid, β-sitosterol, β-5 sitosterol glucoside, dihydrokaempferol, dihydrokaempferol 3-O-glycoside, catechin, gallic acid, vanillic acid, ethyl gallate, ethyl gallate 4-O-β-D7glucopyranoside, syringic acid, benzyl 6ʹ-O-galloyl-β-d-glucopyranoside, ellagic acid, 3ʹ-O-methylellagic acid 4-O-α-l-rhamnopyranoside, 3,3′4′-O-tri-methylellagic acid 4-O-β-d-glucopyranoside, and resveratrolInhibition: TNF-α, IL-6, IL-1β, and CCL17 expression; STAT-1, NK-κB, ERK and p38 activationanti-inflammatory[83]
Sanguisorba officinalis L. (Rosaceae)rootsethanolicHaCaT stimulated by TNF-α/IFN-γ(+)-catechin, (–)-epicatechin, ziyuglycosideInhibition: macrophage-derived chemokine (MDC), normal T-cell expressed and secreted (RANTES), IL-8 and thymus and activation regulated chemokine (TARC) production; STAT-1, ERK and NF-κB activationanti-inflammatory[84]
Gleditsia sinensis Lam. (Fabaceae)thornsethanolicHaCaT stimulated by TNF-α/IFN-γ(+)catechin, epicatechin, eriodictyol and quercetin, caffeic acid and ethyl gallateInhibition: MDC and TARC productionanti-inflammatory[85]
Morus alba L. (Moraceae)barksaqueousHaCaT stimulated by TRAILmoracin O and PInduction: antiapoptotic proteins Bcl-xL and Bcl-2
Inhibition: NFκB activation
anti-inflammatory
anti-apoptotic
[86]
Morus alba L. (Moraceae)root barkethanolicHaCaT stimulated by TNF-
α/IFN-γ
kuwanon G and morusinInhibition: RANTES/CCL5, TARC/CCL17, and MDC/CCL22 secretion; STAT 1 and NF-κB activationanti-inflammatory[87]
Combretum collinum Fresen. (Combretaceae)leavesaqueousHaCaT stimulated by TNF-αmyricetin-3-O-rhamnoside and myricetin-3-O-glucosideInhibition: IL-8 secretionanti-inflammatory[88]
Aucklandia lappa DC. (Asteraceae)whole extractmethanolicHaCaT stimulated by TNF-α/IFN-γalantolactone, caryophyllene, costic acid, costunolide, and dehydrocostuslactoneInhibition: TARC, RANTES, MDC and IL-8 production; STAT1 activationanti-inflammatory[89]
Quercus mongolica Fisch. ex Ledeb. (Fagaceae)leavesacetoneHaCaT stimulated by LPSpedunculaginInhibition: IL-6 and IL-8 productionanti-inflammatory[90]
Melaleuca styphelioides Sm. (Myrtaceae)leavesmethanolicNCTC 2544 keratinocytes stimulated by IFN-γ/histaminequercetin, gallic acid, ellagic acidInhibition: ICAM-1, iNOS, COX-2, NF-κBanti-inflammatory
antioxidant
[91]
Carpinus tschonoskii Maxim. (Betulaceae) leavesethanolic HaCaT cells stimulated by LPS tellimagrandin I Inhibition: IL-6 production anti-inflammatory[92]
Table 3. Selected plant extracts from different species with identified compounds and their in vitro effect on UV-radiation-stimulated human keratinocytes.
Table 3. Selected plant extracts from different species with identified compounds and their in vitro effect on UV-radiation-stimulated human keratinocytes.
Name of the Species/FamilyPart of the PlantType of ExtractCell LineIdentified CompoundsMechanism of ActionEffectsRef.
Petasites japonicus (Siebold & Zucc.) Maxim. (Asteraceae)leavesmethanolicNHEKs exposed to UVB irradiationkaempferol-3-O-(6″-acetyl)-β-d-glucoside, quercetin-3-O-(6″-acetyl)-β-d-glucoside, kaempferol-3-O-β-d-glucoside, and quercetin-3-O-β-d-glucosideInduction:
Nrf2 and heat-shock response transcription elements (HSE) that resulted in the induction
of heme oxygenase-1 (HO-1) and HSP70, respectively
Protection against UV-induced cell damages, anti-apoptotic[142]
Rubus idaeus L. (Rosaceae)fruitsethanolicHaCaT exposed to UVB radiationcyanidin, ellagic acid, pelagonidin-3-sophoroside, methylquercetin-pentose conjugate, and cyanidin-3-rutinosideInduction: SOD, Nrf2, and HO-1.
Inhibition: caspase-3,
c-jun modulation; NF-κB and COX-2 activation
antioxidant, anti-apoptototic
anti-inflammatory
[143]
Castanea sativa Mill.
(Fagaceae)
leavesmethanolicHaCaT exposed to UVB radiationcrenatin, chestanin,
gallic acid, cretanin, 5-O-p-coumaroylquinic acid, p-methylgallic acid and quercetin-3-O-glucoside
Inhibition: p53 expressionprotection against UVB-induced cell damages, antioxidant[144]
Potentilla kleiniana Wight et Arn (Rosaceae)whole plantethanolicHaCaT exposed to UVB radiationdiosmetin-7-O-neohesperidoside, dimethylellagic acid hexose, zizybeoside I, 4-O-[b-d-xylopyranosyl]-3,30-di-O-methylellagic acid, and buddlenol AInhibition: caspase-3cytoprotective effect[145]
Crepidiastrum denticulatum (Houtt.) Pak & Kawano (Asteraceae)whole plantethanolicHaCaT exposed to UVB radiationchicoric acid, 3,5dicaffeoylquinic acid, chlorogenic acid, luteolin 7-O-glucuronide, youngiaside A, youngiaside B, youngiaside CInduction: antioxidant enzymes expression
Inhibition: ROS release, MAPKs, AP-1 and NF-κB activation
antioxidant, anti-inflammatory[146]
Vitis vinifera L. (Vitaceae)leavesaqueousHaCaT exposed to UVB radiationcaftaric acid, rutin, hyperoside, quercetin 3-O-glucoside, quercetin 3-O-glucuronide, kaempferol 3-O-glucoside, delphinidin 3-O-glucoside, cyanidin 3-O-glucoside, petunidin 3-O-glucoside, peonidin 3-O-glucoside, malvidin 3-O-glucosideInhibition: IL-8 secretionanti-inflammatory[147]
Dalbergia odorifera T.C.Chen (Fabaceae)heartwoodethanolicHPEKs exposed to UVB radiationsativanoneInhibition: ROS release, p53 and p21 protein productionantioxidant, anti-senescence[148]
Opuntia ficus-indica (L.) Mill. (Cactaceae)stemsaqueousHaCaT exposed to UVA radiationeucomic and piscidic acidsInhibition: ROS production, lipid
peroxidation and GSH depletion
antioxidant[149]
Melissa officinalis L.
(Lamiaceae)
leavesethanolicHaCaT exposed to UVB radiationrosmarinic acid, salvianolic acid derivatives, caffeic acid and luteolin glucuronideInhibition: ROS production, DNA damage and DNA damage responsecytoprotective
antioxidant
[150]
Rhus javanica L.
(Anacardiaceae)
whole plantethanolicHaCaT exposed to UVB radiationgallic acid, 5-O-galloyl-β-d-glucose, Methyl gallate, Syringic acid, Protocatechuic acidInhibition: COX-1, MMP-1 exprwssion; MAPK, AKT, EGFR activityantioxidant, anti-inflammatory[151]
Juglans regia L.
(Juglandaceae)
flowersmethanolicHaCaT exposed to UVB radiation3,7-dimethyl-1,6-octadiene, pentadecanoic acid, 14-methyl, methyl ester, 2-(2,6-dimethoxy-benzoylamino)-propionic acid, ethyl ester, hexadecanoic acid, ethyl ester (palmitic acid), 10-octadecenoic acid, methyl ester, erucic acid; 1,2,3-benzothiadiazole; estra-1,3,5(10),6-tetraene-3,17-diol, (17β)-; 17-acetate, 2,2,4-trimethyl-3- (3,8,12,16-tetramethyl-heptadeca-3,7,11,15-tetraenyl)-cyclohexanol and oleic acid, trimethylsilyl esterInhibition: ROS production, lipid peroxidation,
TNF-α, IL-1, IL-6, NF-κB, COX-2 activation
antioxidant, anti-inflammatory[152]
Portulaca oleracea L.
(Portulacaceae)
whole plantmethanolicHaCaT exposed to UVB radiationportulacanone A and portulacanon DInduction: SOD expression, and HO-1 via Nrf2 pathway
Inhibition: ROS production
antioxidant[153]
Rosa multiflora Thunb. (Rosaceae)flowersethanolicHaCaT exposed to UVB radiationquercitrin, hyperin, and isoquercetinInhibition: ROS production, IL-6, IL-8 MMP1; NF-κB activationanti-oxidant
anti-inflammatory
[154]
Rhodomyrtus tomentosa (Aiton) Hassk.
(Myrtaceae)
fruitsethanolicNHEKs exposed to UVB radiationpiceatannolInhibition: cyclobutane pyrimidine dimers formation, prostaglandin E2 secretion
Induction: enzyme activity of the DNA polymerases
cytoprotective
anti-inflammatory
[155]
Cecropia obtusa Trécul (Urticaceae)leavesmethanolHaCaT exposed to UVB radiationchlorogenic acid, luteolin-C-hexoside, luteolin-Chexose-O-deoxy-hexose, and apigenin-C-hexose-O-deoxy-hexoseInhibition: MMP-1, IL-1β and IL-6anti-inflammatory[156]
Scutellaria baicalensis Georgi (Lamiaceae) rootsethanolicHaCaT exposed to UVB radiationbaicalin, wogonoside, baicalein and wogoninInduction: HO-1; Nrf2 activation
Inhibition: MMP-1, IL-6; MAPK, AP-1 and NF-κB activation
cytoprotective
anti-inflammatory
antioxidant
[157]
Spatholobus suberectus Dunn (Fabaceae)stemaqueous and ethanolicHaCaT exposed to UVB radiationgallic acid, catechin, vanillic acid, syringic acid and epicatechinInhibition: ROS production; MAPKs, NF-κB, c-Jun activationanti-inflammatory
antioxidant
[158]
Adenocaulon himalaicum Edgew. (Asteraceae) leaves ethanolHaCaT exposed to UVB radiationneochlorogenic acidInduction: filaggrin, involucrin, loricrin expression
Inhibition: MMP-1; MAPK, AP-1 activation
hydration
anti-inflammatory
antioxidant
[159]
Paeonia × suffruticosa Andrews (Paeoniaceae) roots ethanolHaCaT exposed to UVB radiationpaeonolInduction HO-1; Nrf2 activation
Inhibition: MAPK
cytoprotective,
antioxidant
[160]
Aquilaria crassna Pierre ex Lecomte
(Thymelaeaceae)
leavesaqueous/ethanolicNHEKs exposed to UVB radiationiriflophenone 3,5-C-β-d-diglucoside, iriflophenone 3-C-β-d-glucoside, mangiferin and genkwanin 5-O-β-primevosideInhibition: IL-1α, IL-8 and
prostaglandin E2 (PGE2)
anti-inflammatory[161]
Aloe vera (L.) Burm.f.
(Asphlodelaceae)
flowersaqueousHaCaT exposed to UVB radiationisoorientinInduction: involucrin expressionhydration[162]
Nymphoides indica (L.) Kuntze (Menyanthaceae)whole plantmethanolicHaCaT exposed to UVB radiationquercetin 3,7-dimethyl ether 4′-glucosideInduction: filaggrin, involucrin, loricrin expression
Inhibition: MAPK, NF-κB activation
hydration
cytoprotective
antioxidant
[163]
Biancaea sappan (L.) Tod. (Fabaceae)whole plantmethanolicNHDKs exposed to UVBA radiationbrazilinInduction: glutathione peroxidase 7antioxidant[164]
Clitoria ternatea L.
(Fabaceae)
flowersaqueousHaCaT exposed to UVB radiationdelphinidin, including polyacylated ternatins, and flavonol glycosides derived from quercetin and kaempferolInhibition: mtDNA damagecytoprotective[46]
Syzygium formosum (Wall.) Masam
(Myrtaceae)
leavesethanolicHaCaT exposed to UVB radiationtriterpenic acidsInhibition: IL-1β, IL-6, IL-8 and COX-2 expressionanti-inflammatory[165]
Aster yomena (Kitam.) Honda.
(Astereae)
callusaqueousHaCaT exposed to UVB radiationrobustic acid, 3,5-Di-O-methyl-8-prenylafzelechin-4beta-ol, acetylpterosin C and pterosin N, L-thyronine, 3,4-dicaffeoyl-1,5-quinolactone, dehydrophytosphingosine and phytosphingosine, α-linolenic acid, palmitic amide, olemaide, and 13Z-docosenamide, and glycerophospholipidsInducttion: type I procollagen synthesis, TGF-β expression
Inhibition: ROS production, elastase production, MMP-1, MMP-3, MMP-9, TNF-α, IL-1β, IL-8 expression
cytoprotective
antioxidant
anti-inflammatory
[166]
Table 4. Selected plant extracts from different species with identified compounds and their in vitro effect on human keratinocyte migration and proliferation rates.
Table 4. Selected plant extracts from different species with identified compounds and their in vitro effect on human keratinocyte migration and proliferation rates.
Name of the Species/FamilyPart of the PlantType of ExtractCell LineIdentified CompoundsMechanism of ActionEffectsRef.
Boesenbergia rotunda (L.) Mansf. (Zingiberaceae)rhizomesethanolicHaCaTkaempferolInduction: ERK 1/2, Akt
Activation: MAPK and PI3K/Akt pathways
stimulate proliferation[241]
Rubus fruticosus L. (Rosaceae)leavesaqueousHaCaTphenolic compounds-stimulate migration[242]
Alternanthera sessilis (L.) R.Br. ex DC. (Amaranthaceae)stemsethanolicHaCaT2,4-dihydroxy-2,5-dimethyl-3(2H)-furan-3-one, hexadecanoic acid <n->, 2-1,2,4-trioxolane,3-phenyl-, palmitate <ethyl- and L-glutamic acid-stimulate migration and proliferation[243]
Hibiscus syriacus L.
(Malvaceae)
leavesethanolicHaCaTflavonoids, coumarins-stimulate migration[244]
Digitaria sanguinalis (L.) Scop. (Poaceae)flowersethanolicHaCaTxycaine, hexadecanoic acid, linolenic acid, octadecanoic acid, phenol, 2,2′-methylenebis [6-(1,1-dimethylethyl)-4-methyl-, pentacosane, heptacosane, squalene, 1-docosene, cyclooctacosane, campesterol, stigmasterol, lanosterol, multiflora-7,9(11)-dien-3β-ol, sitostenone-stimulate proliferation[245]
Fuchsia magellanica Lam. (Onagraceae)leavesaqueous, ethanolicHaCaTgallic acid derivatives, hydroxycinnamic acid derivatives and flavonoid glycosides, anthocyanins-stimulate migration[246]
Elaeagnus umbellata Thunb. (Elaeagnaceae)leaves and twigsacetoneHaCaTN-[2-(5-hydroxyl-1H- indol-3-yl)ethyl]-butanamide, kaempferol-3-O-β-d-xylopyranosyl(1→2)-β-d-galactopyranoside-7-O-α-l-rhamnopyranoside, kaempferol-3-O-β-d-galactopyranoside-7-O-α-Lrhamnopyranoside, kaempferol-3-O-α-l-rhamnopyranosyl(1→6)-β-d-galactopyranoside-7-O-α-l-rhamnopyranoside, kaempferol-3-O-β-d-xylopyranosyl(1→2)-β-d-galactopyranoside-stimulate proliferation[247]
Annona crassiflora Mart. (Annonaceae)seedsaqueousHaCaTcatechin, epicatechin, rutin, quercetin, naringenin, protocatechuic acid, 4-hydroxybenzoic acid, vanillic acid, chlorogenic acid, caffeic acid, p-coumaric acid, ferulic acid-stimulate migration[248]
Combretum mucronatum Schumach. & Thonn. (Combretaceae)leafaqueousNHEKsepicatechin, procyanidinB2, vitexin and isovitexin-stimulate migration and differentiation[249]
Achillea asiatica Serg. (Asteraceae)aerial partethanolicHaCaTchlorogenic acid, schaftoside, quercetin-3-O arabinosyl(1→6)glucoside, apigenin-7-O-glucoside, luteolin, and apigeninInduction: β-catenin, Aktstimulate migration[250]
Moringa oleifera Lam. (Moringaceae)leavesaqueousNHEKsvicenin-2, chlorogenic acid, gallic acid, quercetin, kaempferol, rosmarinic acid and rutin-stimulate migration and proliferation[251]
Plantago australis Lam. (Plantaginaceae)leavesethanolicHaCaTverbascoside-stimulate migration[252]
Aegle marmelos (L.) Corrêa (Rutaceae)flowerethanolicHaCaTcineol, aegelin, cuminaldehyde, luvangetin, 1-hydroxy-5,7-dimethoxy-2-naphthalene-carboxaldehyde, eugenol-stimulate migration[253]
Boerhavia diffusa L.
(Nyctaginaceae)
leavesmethanolicHaCaTcaffeic acid, ferulic acid and D-pinitol-stimulate migration[254]
Annona reticulata L.
(Annonaceae)
leavesethanolicHaCaTquercetin and β-sitosterolIncreased: VEGF and Aktstimulate migration and proliferation[255]
Centella asiatica (L.) Urb. (Apiaceae)whole plantmethanolicHaCaTasiaticoside-stimulate migration[256]
Afgekia mahidoliae B. L. Burtt & Chermsir.
(Fabaceae)
leaveschloroform/methanolHaCaTkaempferol-3-O-arabinoside, kaempferol-3-O-glucoside, and kaempferol-3-O-rutinoside,Induction: filopodia and lamellipodia formation, Aktstimulate migration[257]
Aloe vera (L.) Burm.f. (Asphodelaceae)leavesaqueousHPEKsanthraquinonesInduction: β1-, α6-, β4-integrin, and E-cadherin expressionstimulate migration[258]
Aristolochia bracteolata Lam. (Aristolochiaceae)leavesmethanolicHaCaTquercetin-stimulate migration[259]
Stellera chamaejasme L. (Thymelaeaceae)aerial partsethanolicHaCaTdaphnin, daphnetin-8-O-glucoside, daphnetin, rutarensin, isoquercitrin, chamechromone and daphnoretinInduction: β-catenin, ERK and Aktstimulate migration[260]
Polygonum aviculare L (Polygonaceae)whole plantethanolicHaCaTquercitrin hydrate, caffeic acid, and rutinInduction: Wnt/β-catenin signalingstimulate migration[261]
Hypericum carinatum Griseb. (Hypericaceae)aerial partsn-hexaneHaCat cellsuliginosin A, japonicin A, uliginosin B, hyperbrasilol B, and the three benzopyrans, that is, 6-isobutyryl-5,7-dimethoxy-2,2-dimethyl-benzopyran, 7-hydroxy-6-isobutyryl-5-methoxy-2,2-dimethyl-benzopyran, and 5-hydroxy-6-isobutyryl-7-methoxy-2,2-dimethyl-benzopyran-stimulate migration[262]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Merecz-Sadowska, A.; Sitarek, P.; Zajdel, K.; Kucharska, E.; Kowalczyk, T.; Zajdel, R. The Modulatory Influence of Plant-Derived Compounds on Human Keratinocyte Function. Int. J. Mol. Sci. 2021, 22, 12488. https://doi.org/10.3390/ijms222212488

AMA Style

Merecz-Sadowska A, Sitarek P, Zajdel K, Kucharska E, Kowalczyk T, Zajdel R. The Modulatory Influence of Plant-Derived Compounds on Human Keratinocyte Function. International Journal of Molecular Sciences. 2021; 22(22):12488. https://doi.org/10.3390/ijms222212488

Chicago/Turabian Style

Merecz-Sadowska, Anna, Przemysław Sitarek, Karolina Zajdel, Ewa Kucharska, Tomasz Kowalczyk, and Radosław Zajdel. 2021. "The Modulatory Influence of Plant-Derived Compounds on Human Keratinocyte Function" International Journal of Molecular Sciences 22, no. 22: 12488. https://doi.org/10.3390/ijms222212488

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop