Next Article in Journal
Protopine and Allocryptopine Interactions with Plasma Proteins
Next Article in Special Issue
Antimicrobial Activity of Arthrospira (Former Spirulina) and Dunaliella Related to Recognized Antimicrobial Bioactive Compounds
Previous Article in Journal
Metabolic Composition of Methanolic Extract of the Balkan Endemic Species Micromeria frivaldszkyana (Degen) Velen and Its Anti-Inflammatory Effect on Male Wistar Rats
Previous Article in Special Issue
Regioselective and Stereoselective Synthesis of Parthenolide Analogs by Acyl Nitroso-Ene Reaction and Their Biological Evaluation against Mycobacterium tuberculosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Multiplicative Effects of Essential Oils and Other Active Components on Skin Tissue and Skin Cancers

Department of Physiology, College of Medicine, Gachon University, Lee Gil Ya Cancer and Diabetes Institute, 155 Getbeolro, Yeonsu-gu, Incheon 21999, Republic of Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(10), 5397; https://doi.org/10.3390/ijms25105397
Submission received: 23 March 2024 / Revised: 12 May 2024 / Accepted: 13 May 2024 / Published: 15 May 2024
(This article belongs to the Special Issue Current Research in Antimicrobial Natural Products)

Abstract

:
Naturally derived essential oils and their active components are known to possess various properties, ranging from anti-oxidant, anti-inflammatory, anti-bacterial, anti-fungal, and anti-cancer activities. Numerous types of essential oils and active components have been discovered, and their permissive roles have been addressed in various fields. In this comprehensive review, we focused on the roles of essential oils and active components in skin diseases and cancers as discovered over the past three decades. In particular, we opted to highlight the effectiveness of essential oils and their active components in developing strategies against various skin diseases and skin cancers and to describe the effects of the identified essential-oil-derived major components from physiological and pathological perspectives. Overall, this review provides a basis for the development of novel therapies for skin diseases and cancers, especially melanoma.

1. Introduction

Essential oils (EO)s are natural oils secreted as secondary metabolites or concentrated plant extracts from many parts of aromatic plants (especially the bark, fruits, and flowers) [1]. EOs and their active components possess many biological properties, including anti-bacterial, anti-virus, anti-fungal, anti-inflammatory, and anti-cancer properties [2,3]. The skin is the first barrier that protects against external stimuli and microorganisms. Moreover, the skin possesses unique characteristics, such as its stratified structure, various cell types, and the pigmentation process, which are distinct from other tissues. Notably, EOs are used to treat several diseases, including skin diseases such as eczema, psoriasis, dermatitis, and skin cancers. In this review, we aimed to highlight the benefits of various EOs and several plant oils (POs) in terms of skin reactivity and the treatment of skin cancers, especially melanoma, with a PubMed-based selection of studies published in the last three decades which address the effects of EOs/POs on skin diseases and skin cancers. Overall, a summary of the effectiveness of EOs would help alleviate skin diseases and promote the development of therapeutic strategies against various skin diseases and skin cancers.

2. Anti-Inflammatory Role of EOs

2.1. Citrus limetta Peel Essential Oil (Cl-EO)

Citrus limetta (C. limetta) Risso peels exhibit various medicinal activities, such as anti-oxidant and anti-inflammatory activities [4], due to the presence of large quantities of flavonoids [5]. EO, a widely used ingredient in cosmetic and pharmaceutical products, is an important product of citrus fruit peels [6,7]. EOs and their constituents from citrus fruits exhibit anti-inflammatory activities in vitro by inhibiting the biosynthesis of inflammatory cytokines [8]. Inflammatory cytokines, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6, are found to be dose-dependently reduced by treatment with C. limetta peel EO (Cl-EO) in the lipopolysaccharide (LPS)-exposed primary macrophages or the 12-O-tetradecanoylphorbol-13-acetate (TPA), used as protein kinase C (PKC) activator, -exposed mouse ear [9]. In addition, Cl-EO is found to inhibit, dose-dependently, oxidative stress in hydrogen peroxide (H2O2)-exposed primary macrophages and TPA-exposed mice [9].

2.2. Baccharis dracunculifolia Essential Oil (BD-EO)

Baccharis dracunculifolia (B. dracunculifolia) is a domestic plant which is widely used as an immunomodulator and an anti-bacterial and anti-diabetic agent, but it possesses many other properties [10,11]. Several studies have evaluated the medicinal actions of B. dracunculifolia, especially its anti-microbial effects [12,13,14]. Dos Santos et al. reported the anti-inflammatory effects of B. dracunculifolia leaves extract on a paw edema model, which is induced by carrageenan and formalin [15]. Additionally, B. dracunculifolia leaf extract is found to exhibit an inhibitory effect on LPS-challenged murine macrophages by inhibiting the biosynthesis of IL-6 and IL-10 [16,17]. Ear edema, infiltration, and proliferation/differentiation of keratinocytes, as well as the activities of myeloperoxidase and N-acetylglutamate synthase, are downregulated by BD-EO administration in a TPA-induced acute and chronic inflammatory mouse model [18].

2.3. Perilla frutescens L. Britt Essential oil (PF-EO)

Perilla frutescens (P. frutescens) L. Britt is a herbaceous plant that plays a critical role in Chinese medicine and is used to treat various pathological symptoms, including abdominal pain, nausea, cold, constipation, food poisoning, and cough [19]. The EOs from P. frutescens have been noted to possess anti-bacterial, anti-inflammatory, and anti-oxidant properties [20,21]. In addition, psoriasis symptoms, such as erythema, scaling, and epidermal thickening, have previously found to be alleviated by treatment with P. frutescens L. Britt EO (PF-EO) in a mouse model of imiquimod (IMQ)-induced psoriasis [22]. Inflammation-related factors, such as inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2, IL-1, IL-6, and neutrophil proliferation, are downregulated by PF-EO treatment in the full skin and epidermis of an IMQ-induced psoriasis in mouse [22]. IMQ induces dendritic cell activation, which is related to the activation of macrophages and T-lymphocytes [23,24,25]. The upregulated mRNA expression levels of IL-17, IL-22, IL-23, interferon (IFN)-α, and IFN-γ due to IMQ administration are found to be reduced by PF-EO treatment in the skin [22]. The production of psoriasis-development-associated cytokines, such as IL-1, IL-6, IL-17, IL-23, and nuclear factor kappa B (NF-κB), are downregulated by PF-EO treatment in the serum of an IMQ-induced mouse [22] (Figure 1).

2.4. Grapefruit Essential Oil (G-EO)

Citrus EO exhibits various biological activities, such as anti-microbial [26], anti-oxidant [27], and anti-fungal activities [28]. Grapefruit (Citrus maxima (Burm). Merr) is one of the citrus producers, and the major components of grapefruit EO (G-EO), which are extracted from the grapefruit peel, are terpenes and terpene oxides [29,30,31]. G-EO is a valuable ingredient because of its characteristic flavor and fragrance [32,33]. G-EO exhibits various biological activities, including free radical scavenging [29,34,35,36,37] and anti-inflammatory [38], anti-bacterial [39,40,41,42], anti-microbial [43], anti-proliferative [37], and anti-cancer activities [44,45,46], and is a positive regulator of sympathetic nervous system activity [47]. The main component of citrus EO is limonene, an important anti-oxidant [48] and anti-inflammatory agent [49]. Inflammatory cell damage and reactive oxygen species (ROS) biosynthesis are inhibited by G-EO treatment through the downregulated expression of IL-1 and COX-2 in Staphylococcus aureus (S. aureus)-exposed HaCaT keratinocytes [50]. These results indicate that G-EO exhibits a remarkable protective effect in HaCaT keratinocytes by inhibiting inflammatory-stimulus-mediated ROS production. In addition, skin-barrier-structure-associated proteins, such as filaggrin (FLG) and loricrin, are found to be recovered by treatment with G-EO in an S. aureus-degraded 3D skin model [50].

2.5. Oregano Essential Oil (O-EO)

Origanum vulgare (O. vulgare), also known as oregano, is a ubiquitous aromatic plant of the Lamiaceae family and a typical Mediterranean flora [51]. Assuming no toxicity, O. vulgare is used not only in therapeutic regimens [52,53,54] but also in the food, agricultural, and veterinary fields [55,56]. Moreover, oregano EOs (O-EOs) exhibit a unique function in the prevention of neurodegenerative disorders [57]. The major components of O-EO are carvacrol (CRV) and thymol [51], which exhibit anti-oxidant, immunomodulatory, anti-cancerous, anti-melanogenesis, anti-inflammatory, and anti-microbial activities [52,54,58]. The anti-cancer effects of the EOs, such as their effects against melanoma, are discussed in Section 3. O-EO is found to reduce ROS production, DNA damage, and the expression of inflammatory factors, such as inter-cellular adhesion molecule (ICAM)-1, iNOS, and COX-2, in IFN-γ/histamine-induced NCTC254 cells, a normal human keratinocyte cell line [51]. In addition, the expression levels of extracellular matrix (ECM) agents—such as matrix metalloproteinase (MMP)-1 and MMP-12—and cell proliferation are found to be reduced by O-EO treatment in IFN-γ/histamine-induced NCTC254 cells [51].

2.6. Satureja sahendica Essential Oil (SS-EO)

Satureja sahendica (S. sahendica) is a perennial plant and belongs to the Lamiaceae family [59]. The major constituents of S. sahendica EO (SS-EO) are CRV, thymol, p-cymene, ß-caryophyllene, linalool, and other terpenoids with anti-bacterial properties [60]. Assuming no toxicity, the Satureja genus is known to exhibit anti-oxidant, anti-bacterial, and anti-inflammatory effects in the management of diarrhea and wound healing [61,62,63]. Topical SS-EO treatment is found to reduce the wound area, bacterial count, and cell infiltration in wound-induced mouse [59]. Mechanically, SS-EO enhances the levels of various factors, such as the anti-inflammatory-associated cytokine, IL-10 [64]; proliferation-associated factors, insulin-like growth factor (IGF)-1, fibroblast growth factor (FGF)-2, and chemokine (C-X-C motif) ligand (CXCL)-1 [65,66,67,68]; and angiogenesis-associated factors, vascular endothelial growth factor (VEGF), and transforming growth factor (TGF)-β [65,69], in isolated primary skin tissues from mice [59]. In addition, SS-EO is also found to downregulate the level of IL-1β in mouse skin tissues [59].

2.7. Matricaria chamomilla Essential Oil (MC-EO)

Matricaria chamomilla L., also known as German chamomile, is a member of the Compositae family [70]. Matricaria chamomilla (M. chamomilla) is reported as possessing traditional uses; it is used in the treatment of gastrointestinal conditions [71] and has anti-inflammatory [72] and anti-spasmodic properties [73]. In addition, it is reported that M. chamomilla EO (MC-EO) promotes wound healing and repairs the wounded skin barrier [74]. Moreover, Wang et al. found that MC-EO decreased the pro-inflammatory factors such as TNF-α and IL-6 during eczema [75]. The major component of MC-EO is azulene [70]. The inflammatory cytokines—IL-1β, IL-6, and TGF-β—are attenuated by via MC-EO treatment through downregulation of phosphorylated (p)-Akt, the p-mammalian target of rapamycin (p-mTOR), and the p-p38 pathway in IL-22, TNF-α, or LPS-stimulated HaCaT cells [70]. In addition, not only the clinical symptoms of psoriasis, such as erythema, thickening, and scaling, but also the skin inflammation cytokines are alleviated through the downregulation of p-phosphoinositide 3-kinase (PI3K) and p-mTOR by treatment with MC-EO in a mouse model of IMQ-induced psoriasis [70]. Moreover, treatment with MC-EO decreases scratch frequency and serum levels of immunoglobulin (Ig)E, IgG, and histamine in a mouse model of 2,4-dinitrochlorobenzene (DNCB)-stimulated atopic dermatitis (AD) [76]. Moreover, the symptoms of eczema, such as skin erythema, exudation, thickening, rough surface, and swelling, are reduced by treatment with MC-EO in a DNCB-induced eczema mouse model [75]. The serum levels of IL-6, IL-17, and TNF-α are also downregulated through the downregulation of mitogen-activated protein kinase (MAPK) and the NF-κB pathway by treatment with MC-EO in a DNCB-induced eczema mouse model [75].

2.8. Helianthus annuus Plant Oil (HA-PO)

Helianthus annuus (H. annuus), also known as common sunflower, belongs to the Asteraceae family [77]. The major components of H. annuus plant oil (HA-PO) are oleic and linoleic acids [77]. The HA-EO moisturizes and protects damaged skin barriers. In particular, ozonated HA-PO is an active-components mixture obtained from the partial ozonation of HA-PO. Ozonated HA-PO possesses an anti-inflammatory effect on the skin diseases of mice and humans [78]. The symptoms of AD, such as scaling, excoriation, erythema, edema, epidermal thickness, infiltration of mast cell, serum levels of IgE, and spleen weight and lymph node length are decreased by treatment with ozonated HA-PO in an oxazolone (Oxz)-induced AD mouse model [79]. In addition, skin hydration function and FLG are recovered by treatment with ozonated HA-PO through the downregulation of IL-4/signal transducers and activators of the transcription (STAT) 3/extracellular signal-regulated kinase (ERK) pathway in an Oxz-induced AD mouse model [79]. Moreover, the levels of nitrite oxide (NO) and iNOS are inhibited by treatment with ozonated HA-PO through downregulation of IL-1β and TNF-α via MAPK and the NF-κB pathway in serum and skin tissues in an Oxz-induced AD mouse model [79].

2.9. Mentha arvensis Essential Oil (MA-EO)

Mentha arvensis (M. arvensis), commonly known as mint, belongs to Lamiaceae family and is a flowering plant species [80]. Mentha species are native to Asia, Europe, Africa, Australia, and North America [81]. M. arvensis has been commonly used in many medicines for its anti-inflammatory and anti-oxidant activities [82]. The major components of M. arvensis EO (MA-EO) are menthol, menthone, and piperitone [80]. The inflammation mediators, such as PGE2 and NO, IL-1β, and IL-6, are decreased by treatment with MA-EO through the downregulation of iNOS and COX-2 in LPS-exposed HaCaT keratinocytes [80]. In addition, the clinical symptoms of AD, such as erythema, edema, and ear thickness, as well as the scoring AD index, are improved by treatment with MA-EO in a DNCB-induced AD mouse model [80]. Moreover, infiltration of mast cell and epidermal layer thickness are attenuated by treatment with MA-EO in a DNCB-induced AD mouse model [80]. Furthermore, the formation of the nucleotide-binding oligomerization-domain-like receptor (NLR) family pyrin-domain-containing 3 (NLRP3) inflammasome is attenuated by treatment with MA-EO in the macrophage of a DNCB-induced AD mouse [83].

2.10. Rosmarinus officinalis Essential Oil (RO-EO)

Rosmarinus officinalis (R. officinalis) L., also known as rosemary, is a perennial evergreen shrub [84]. Rosemary is often used to treat digestive problems, the nervous system, and allergies in Morocco [85]. Takaki et al. assessed the anti-inflammatory effect of R. officinalis EO (RO-EO) and reported that RO-EO treatment reduced effusion volume and leukocyte migration in a carrageenan-stimulated rat model [86]. The major components of RO-EO are camphor and eucalyptol [84]. The symptoms of AD are improved by treatment with RO-EO in a DNCB-induced AD mouse model [84]. In addition, serum levels of IL-6 and TNF-α are attenuated by treatment with RO-EO through regulation of the Janus kinase (JAK)/STAT/MAPK pathway in a DNCB-induced AD mouse model [84].

2.11. Curcuma longa Essential Oil (Cl-EO)

Curcuma longa (C. longa), is a member of the Zingiberaceae family and is grown mainly in Asia and India [87]. The skin penetration effects of the rhizome extracts, molecules, and EO of Curcuma species in skin diseases are reported, assuming no toxicity [88]. The major components of C. longa EO (Cl-EO) are terpinolene and α-phellandrene [89]. The levels of inflammatory cytokines, including, IL-6, IL-1β, and TNF-α, are downregulated by treatment with Cl-EO in LPS- or TPA-stimulated HaCaT cells [89]. In in vivo systems, the levels of IL-1β, IL-6, and TNF-α are decreased by treatment with Cl-EO in the serum of a TPA-induced inflammatory mouse model [89]. In addition, ear edema and leucocyte infiltration are reduced by treatment with Cl-EO in a TPA-induced inflammatory mouse model [89].

2.12. Artemisia argyi Essential Oil (AA-EO)

Artemisia argyi (A. argyi) is a species of herbaceous plant distributed in China, Japan, and many parts of Taiwan [90,91]. A. argyi has been determined to possess various biological properties, including an anti-mutagenic [92], anti-cancer [93,94,95,96], anti-inflammatory [97], and anti-oxidant [91] properties. Additionally, the biological properties of A. argyi EO (AA-EO) have been studied, such as its anti-asthmatic [98] and anti-fungal activity [99]. The major components of AA-EO are cineole, camphor, eucalyptol, and caryophyllene [90,91]. Ear edema, as well as neutrophil infiltration, hyperplasia of the epidermal layer, and disruption of connective tissue, are reduced by treatment with AA-EO through downregulation of the JAK/STAT/COX-2 pathway in a TPA-induced inflammatory mouse model [90].

2.13. Lavender-Essential Oil (L-EO)

Lavandula angustifolia, also known as lavender, belongs to the Labiatae family and has been used as either a form of dried plant or for its volatile oils due to its diverse therapeutic and cosmetic properties [100]. Lavender EO (L-EO) has been reported to possess numerous biological properties, including anxiolytic [101], neuroprotective [102], anti-oxidant [103], analgesic [103], anti-inflammatory [103,104], anti-microbial [105,106], wound healing [107], and anti-joint-pain properties [108]. The symptoms of psoriasis, such as thickness, erythema, and scaling, and the inflammatory cytokines levels of T-helper (Th)-17-specific cells, such as IL-17 and IL-22, and Th-1-specific cells, such as TNF-α and IL-1β, are reduced by treatment with L-EO in an IMQ-induced psoriasis mouse model [109]. In addition, the major compounds of L-EO are linalool and linalyl acetate [107], which represent the alleviation effects of psoriasis, such as the reduction of erythema, thickness, scaling, keratin, pigmentation, and curvature, as well as the inflammatory cytokines levels of Th-17- and Th-1- specific cells by downregulating C-C motif chemokine receptor (CCR) 6 and IL-17 expression in an IMQ-induced psoriasis mouse model [109]. In addition, linalool and linalyl acetate exhibit an anti-inflammatory effect against a carrageenan-induced edema model [110].

2.14. Zanthoxylum coreanum Essential Oil (ZC-EO)

The Zanthoxylum species belongs to the Rutaceae family and has been used as a source of spices in Asian cuisine and traditional Asian medicine [111,112]. Zanthoxylum coreanum (Z. coreanum) has been shown to have anti-viral activity against picornaviruses [113]. The major components of Z. coreanum EO (ZC-EO) are β-Ocimene and α-pinene [114]. The AD-like skin lesions, such as large ulcers, ear swelling, and hyperkeratosis, the thickness of the epidermis and dermis, and inflammatory cell infiltration, are inhibited by treatment with ZC-EO through the downregulation of NF-κB and the phosphorylated MAPK pathway in a DNCB-induced AD mouse model [114].

3. Anti-Cancer Effect of EOs

EOs are concentrated hydrophobic liquids from aromatic plants that exert anti-cancer effects on various cellular targets [115]. Melanogenesis is a pivotal process in melanocytes, which possess melanosomes to synthesize and store melanin pigment [116]. This process is tightly regulated by several enzymes, including tyrosinase and tyrosinase-related proteins (TRPs)-1 and -2 [117]. These enzymes are responsible for initiating and regulating melanogenesis [117]. In addition, these enzymes contribute to the completion of the process and act as modifiers to regulate pathway velocity [118]. Among melanogenesis-associated enzymes, TRP-1 and TRP-2 stabilize tyrosinase activity and maintain the structural integrity of melanosome [118]. Moreover, melanogenesis is associated with H2O2 production through the enzymatic and non-enzymatic reactions and subsequently induces oxidative stress in melanocytes [119,120]. ROS production is derived by α-melanocyte, stimulating hormone (MSH)-induced melanogenesis [121]. Treatment of several ROS scavengers and ROS inhibitors reduces the UV-induced melanogenesis [122,123]. Therefore, the development of melanogenesis inhibitors, anti-oxidants, and ROS scavengers has been revealed to be beneficial in the treatment of hyperpigmentation in skin care fields. This section is focused on EO’s anti-cancer effects, including their anti-melanoma effects.

3.1. Aloysia citrodora Essential Oil (AC-EO)

Aloysia citrodora (A. citrodora) Paláu (Lippia citriodora Kunth), also known as Aloysia. triphylla (L’Hér.) Britton, Aloysia. citridora, Aloysia. citriodora Paláu, Lippia citriodora Kunth, Lippia. citrodora Kunth, Lippia. triphylla (L’Hér.) Kuntze, Lippia triphylla, Verbena triphylla, Aloysia triphylla, Verbena citriodora, or lemon verbena, is derived from America, Africa, and Europe, and is used in pharmacological purposes to alleviate flatulence, diarrhea, rheumatoid arthritis, and insomnia [124,125]. The A. citrodora EO (AC-EO) contains the chemical metabolites of aromatic plants, such as neral, geranial, limonene, citral, and 1,8-cineole [124,126]. AC-EO and its components (geranial, neral, flavonoid, and phenol) have been reported to possess anti-cancer effects in breast cancer, human chronic myelogenous erythroleukemia, malignant tumor cell lines, a 4T1 breast cancer xenograft mouse model, murine mastocytoma cell line, colorectal adenocarcinoma, hepatocellular carcinoma, breast adenocarcinoma, and malignant melanoma [127,128,129,130]; anti-oxidant effects [131,132]; and anti-bacterial effects [131]. In the presence of AC-EO, cell viability is dose-dependently decreased through the downregulation of p-ERK expression and the upregulation of apoptotic factors such as Bax and caspases-3 and -9 in human epidermoid carcinoma A431, human SK-MEL-28 melanoma, and murine B16F10 melanoma [133]. In addition, the tumor volume is found to be reduced by treatment with AC-EO in a melanoma mouse model [133]. ECM-degrading proteases, including disintegrin and metalloprotease-9 (ADAM-9), MMP-2, -7, and -9, play pivotal roles in cancer processes, including the growth, migration, invasion, adhesion, proliferation, and apoptosis of melanoma [134,135,136,137,138,139]. ECM degradation and MMP expression are modulated by epidermal growth factor receptor (EGFR) signaling [140,141]. The plasma and expression levels of MMP-7 and -9 and ADAM-9 are reduced by AC-EO treatment in B16F10-injected mice and in B16F10 melanoma [133]. EGFR overexpression or heparin-binding (HB)-EGF treatment-mediated cell proliferation and MMP-7, -9, and ADAM-9 expression levels are also found to be reduced by treatment with AC-EO [133]. The recombinant chimeric EGFR monoclonal antibody, cetuximab, induces EGFR silencing [133]. Furthermore, co-treatment with AC-EO and siRNA-EGFR or cetuximab leads to a more effective downregulation of cell proliferation and the expression level of MMP-7 and -9 than individual treatment in B16F10 melanoma [133]. The anti-melanoma effects of AC-EO are shown in Figure 2. Overall, AC-EO is involved in the inhibition of EGFR signaling and may be a therapeutic agent against melanoma.
More effective carriers are available for maintaining the solubility of EOs. Chitosan nanoparticles, which possess biodegradability, biocompatibility, and mucosal adhesion properties, are among the most-common carriers [142,143]. Nanoparticle-based structures containing EOs are promising for improving the solubility and efficacy of EOs [144,145]. DPPH radical-scavenging activity is found to be enhanced by treatment with chitosan nanoparticles containing citral and Lippia citriodora EO (LC-EO) [126]. Several researchers have reported the anti-cancer effects of chitosan nanoparticles containing EOs, such as Syzygium aromaticum EO in A375 melanoma cells [146], Morinda citrifolia EO in A549 cells [147], and celandine (Chelidonium majus L.) EO in MCF-7 cells [148]. Cell viability has been found to decrease in a dose-dependent manner following chitosan nanoparticle-conjugated citral or LC-EO treatment in A375 cells [126].

3.2. Origanum majorana Essential Oil (OM-EO)

The major constituents of Origanum majorana (O. majorana) EO (OM-EO) are Terpinen-4-o1 and L-α-Terpineol [149]. OM-EO exhibits anti-bacterial, anti-oxidant, and anti-fungal effects [150]. Nanoemulsions are dispersions of oil in water or water in oil, formulated using amphiphilic materials (surfactants), where the droplets are on the nanometer scale [151]. Nanoemulsions have important advantages, including high stability, bioavailability, biocompatibility, and biodegradability [149], and are among the most suitable formulations with which to enhance the efficiency and stability of EOs [152]. Recently, several studies have reported the anti-cancer effects of nanoemulsions containing EOs, such as Cinnamomum cassia EO, Zingiber ottensii EO, and Citrus aurantium in A549 [153,154,155]. Notably, treatment with nanoemulsions containing OM-EO enhances DPPH radical-scavenging activity and apoptotic effects in A375 melanoma cells [149].

3.3. Artemisia capillaris Grass Clumps Essential Oil (AC-EO)

The ethanol extract and ethyl acetate fraction of Artemisia capillaris (A. capillaris) possess anti-cancer [156] and anti-oxidant functions [157], respectively. Treatment with A. capillaris grass clumps EO (AC-EO) is found to reduce both intracellular and extracellular melanin contents via the downregulation of TRP-1 signaling in melanoma B16F10 melanoma [118]. The skin is exposed to various external stresses, such as ROS, which induce various deleterious effects and the apoptosis of keratinocytes [158,159]. AC-EO treatment is found to upregulate the proliferation of H2O2-exposed B16F10 melanoma [118].

3.4. Camellia japonica Seed Essential Oil (CJ-EO)

Camellia japonica (C. japonica) seed EO (CJ-EO) possesses various biological activities, including anti-oxidant [160,161], anti-bacterial [162,163], anti-inflammatory [164], and skin barrier function [165]. Tyrosinase is a critical enzyme for melanin synthesis [166]. Therefore, inhibition of substances of tyrosinase, TRP-1, and TRP-2 reduces melanin synthesis [167]. More recently, the major components of CJ-EO are identified as hexamethylcyclotrisiloxane and octamethylcyclotetrasiloxane [168]. The activities of melanin synthesis-associated components, tyrosinase, and TRP-1 and -2, as well as the content of melanin, are dose-dependently reduced by CJ-EO treatment in α-MSH-exposed B16F10 melanoma [168]. Thus, CJ-EO could be considered as a potential whitening agent.

3.5. Origanum syriacum (OS) and Origanum Ehrenbergii (OE)

Origanum syriacum (OS) and Origanum ehrenbergii (OE) are two naturally growing plants in Lebanon that belong to the Lamiaceae family [57]. These plants are used in maceration for rheumatism and neuralgic treatments [169]. The major components of OS and OE are aromatic terpenoids, quinones, and CRV [170]. The cell viability of B16-F1 melanoma is found to be dose-dependently decreased following treatment with OS-EO and OE-EO [171]. Melanin levels are downregulated by treatment with OS-EO, OE-EO, or CRV without any alterations to tyrosinase activity in B16-F1 melanoma [171]. These results indicate that both EOs and CRV exhibit anti-melanogenic activity by competing with tyrosine as a tyrosinase substance.

3.6. Calocedrus formosana Essential Oil (CF-EO)

Calocedrus formosana (C. formosana), also known as Taiwan incense cedar, belongs to the Cupressaceae family [172]. The compounds of C. formosana possess the diverse biological effects, such as anti-cancer [173], anti-oxidative [174,175], anti-inflammation [176], and anti-fungal [177] effects. The major components of C. formosana EO (CF-EO) are α-Terpineol, Terpinen-4-o1, and thymol [172]. The treatment with CF-EO inhibits melanogenesis by inhibiting tyrosinase activity and by reducing TRP-1 and TRP-2, co-stimulated with α-MSH and forskolin (FSK), in B16F10 melanoma [172]. Furthermore, thymol inhibits melanogenesis in the presence of co-stimulation with α-MSH and FSK in B16F10 melanoma [172].

3.7. Melaleuca quinquenervia Essential Oil (MQ-EO)

Melaleuca quinquenervia (M. quinquenervia) belongs to the Myrtaceae family and is native to northern Australia [178]. The major compositions of M. quinquenervia EO (MQ-EO) are the monoterpene family, such as 1,8-cineole, α-pinene, and α-terpineol [179]. Among them, 1,8-cineole is responsible for the anti-bacterial activity of MQ-EO [180]. The melanin production is inhibited by treatment with MQ-EO but also its major compositions, 1,8-cineole, α-pinene, and α-terpineol, through the downregulation of tyrosinase activity in α-MSH-exposed B16 melanoma [179]. It is well known that α-MSH-exposed melanogenesis is involved in ROS production [121,181]. Oxidative damage and lipid peroxidation is recovered by treatment with MQ-EO through the upregulation of glutathione (GSH) levels or anti-oxidants, such as superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT), in α-MSH-stimulated B16 melanoma [179]. GSH plays vital role in anti-melanogenesis and in the maintenance of cellular redox status [182]. The anti-melanogenesis effect of MQ-EO might be regulated by the regulation of tyrosinase activity and anti-oxidant ability.

3.8. Cinnamomum cassia Essential Oil (CC-EO)

Cinnamomum cassia (C. cassia) Presl is broadly grown in China [167]. Assuming no toxicity, the dried form of C. cassia stem bark is known to exhibit multiple biological activities, such as anti-bacterial [183], anti-inflammatory [184], and anti-diabetic properties [185], and its traditional uses are reviewed in [186]. Additionally, it has been found that the extraction of C. cassia twigs suppresses tyrosinase activity [187]. The C. cassia EO (CC-EO) is known to possess hypouricemic [188] and anti-fungal activities [189]. The major components of CC-EO are cis-2-methoxycinnamic acid and cinnamaldehyde [167]. Melanin content is reduced by treatment with CC-EO or its major components through the inhibition of tyrosinase activity and its expression in α-MSH-induced B16 melanoma [167]. α-MSH-mediated oxidative stress and lipid peroxidation are attenuated by treatment with CC-EO or cinnamaldehyde through the regulation of GSH level and CAT activity in α-MSH-induced B16 melanoma [167].

3.9. Leaf of Alpinia nantoensis Essential Oil (LAN-EO) and Rhizome of Alpinia nantoensis Essential Oil (RAN-EO)

Alpinia is the largest genus in Zingiberaceae, which are found in tropical/sub-tropical regions of China, India, and Polynesia [190]. Extractions from the various parts of Alpinia nantoensis (A. nantoensis), such as the leaf, stem, and rhizome, exhibit anti-metastatic properties in lung cancer [191] and breast cancer cells [192]. Treatment with the leaf of A. nantoensis EO (LAN-EO) or rhizome of A. nantoensis EO (RAN-EO) reduces melanogenesis through ERK1/2-activation-mediated MITF ubiquitination and proteasomal degradation, which downregulate tyrosinase and TRP-1 in FSK-stimulated B16F10 cells [193]. The major compositions of LAN-EO and RAN-EO are camphor, camphene, β-pinene, p-cymene, α-pinene, and D-limonene [193]. Among them, α-pinene and D-limonene attenuate melanogenesis by downregulating tyrosinase activity in FSK-stimulated B16F10 melanoma [193].

3.10. Chrysanthemum boreale MAKINO Essential Oil (CB-EO)

Chrysanthemum boreale (C. boreale) MAKINO is a perennial herbaceous plant and is a member of the Compositae family [194]. The C. boreale species, including C. boreale MAKINO, are broadly grown in East Asia, including Korea, China, and Japan [195,196], and, assuming no toxicity, they are widely used as medicinal plants due to their biological properties, such as anti-inflammation and anti-bacterial action, skin-regenerative properties, and wound healing properties [194,196,197]. The major compounds of CB-EO are camphor, germacrene-D, and β-thujone [196]. Moreover, treatment with C. boreale MAKINO EO (CB-EO) from the flower reduces melanogenesis through the downregulation of tyrosinase via the ERK and MAPK pathway in α-MSH-stimulated B16BL6 melanoma [198].

3.11. Vitex negundo Essential Oil (VN-EO)

Vitex negundo (V. negundo) Linn is a large aromatic shrub belonging to the Verbenaceae family, mainly distributed in the Indochina region [199]. V. negundo has been revealed to possess various pharmacological properties, including anti-nociceptive [200,201], anti-convulsant [202], and anti-inflammatory [201] properties. In addition, the extracts of V. negundo Linn are known to possess analgesic [203], anti-inflammatory [203], and tyrosinase inhibition properties [204]. The major components of V. negundo EO (VN-EO) are sesquiterpene and monoterpene [199]. Treatment with VN-EO reduces melanogenesis through the downregulation of tyrosinase activity in α-MSH-induced B16F10 cells [199]. Moreover, the anti-oxidant effects of VN-EO are revealed through its DPPH radical-scavenging activity [199].

3.12. Achillea millefolium Essential Oil (AM-EO)

The Achillea genus of the Asteraceae family is broadly distributed throughout the world [205], and among this genus is Achillea millefolium (A. millefolium) L., which is used to treat gastrointestinal disorders [206], hepatobiliary conditions [207], and cardiovascular [208] and respiratory infections [209]. The major components of A. millefolium EO (AM-EO) are camphor, artemisia ketone, linalyl acetate, and 1,8-cineole [210]. Among them, linalyl acetate inhibits melanin contents by downregulating tyrosinase activity in α-MSH-induced B16 melanoma [211]. Treatment with AM-EO reduces melanogenesis through the regulation of tyrosinase activity and the Jun N-terminal kinase -ERK pathway in α-MSH-stimulated B16 melanoma cells [211]. Moreover, oxidative stress and lipid peroxidation are attenuated by treatment with AM-EO through the regulation of the GSH level and anti-oxidant enzymes in α-MSH-stimulated B16 cells [211].

3.13. Artemisia argyi Essential Oil (AA-EO)

The anti-inflammatory activity of AA-EO has been addressed in Section 2.12. Melanogenesis is attenuated by treatment with AA-EO through the downregulation of tyrosinase activity or oxidative stress in α-MSH-stimulated B16F10 cells [91].

3.14. Vetiveria zizanioides Essential Oil (VZ-EO)

Vetiveria zizanioides (V. zizanioides) is a perennial tussock grass that belongs to of the Gramineae family [212,213]. Moreover, the various parts or volatile oils of V. zizanioides possesses diverse biological properties, including anti-oxidant [214], anti-microbial [215], and anti-inflammatory properties [216]. The major components of V. zizanioides EO (VZ-EO) are Cedr-8-en-13-ol and α-Amorphene [217]. The melanin production is attenuated by treatment with VZ-EO through the inhibition of tyrosinase activity and anti-oxidant activity via the regulation of GSH level and anti-oxidant enzymes, GPx and SOD, in α-MSH-stimulated B16 melanoma cells [217].

3.15. Eucalyptus camaldulensis Essential Oil (EC-EO)

Eucalypt trees belong to the Eucalyptus genus and Myrtaceae family [218]. The Eucalyptus genus is derived from Australia [219]. The diverse pharmacological activities of Eucalyptus species leaf EO, such as its anti-bacterial [220], anti-termitic [221], and anti-oxidant activities [222], have been reported. The major components of Eucalyptus camaldulensis EO (EC-EO) are eucalyptol and γ-terpinene [223]. Melanogenesis is reduced by treatment with EC-EO through the downregulation of MAPK/PKA signaling pathways and TRP-1 activities in α-MSH-stimulated B16F10 cells [223].

3.16. Acorus macrospadiceus Essential Oil (AM-EO)

Plants of the Acorus species belong to the Acoraceae family and are distributed in India, Europe, and Asia [224]. Assuming non-toxicity, the biological activities of the EOs extracted from Acorus species are reported. The volatile oils from Acorus (A.) calamus rhizomes possess diverse biological properties, including anthelmintic activities [225] and acetylcholinesterase-inhibitory activities [226]. In addition to Acorus calamus, the volatile oil from A. gramineus rhizomes possesses a neuroprotection function [227]. The major components of A. macrospadiceus EO (AM-EO) are chavicol methyl ether and nootkatone [228]. Melanogenesis is reduced by treatment with AM-EO through the downregulation of tyrosinase activity or oxidative stress in α-MSH-stimulated B16F10 cells [228].

4. Modulation of Skin Proliferation

Wound healing is complex physiological process which consists of three dynamic steps, including an inflammatory step, a proliferative step, and a maturation step involving the development of cellular connective tissue and the formation of newly generated epithelial tissue [229,230]. The cellular connective tissue is supported by the presence of collagen [231]. In addition, dysregulated keratinocyte growth and differentiation lead to aberrant cellular processes, including hyperproliferation, abnormal differentiation, and inflammatory infiltration [232]. The hyperproliferation of epidermal keratinocytes leads to the pathogenesis of several cutaneous disorders, such as psoriasis and AD [233]. These diseases are characterized by dysregulated epidermal homeostasis, which causes disordered skin proliferation and differentiation [234]. This section is focused on the effects of EOs on skin proliferation and regeneration in maintaining epidermal homeostasis.

4.1. Artemisia montana Pampan Essential Oil (AM-EO)

The Artemisia genus has been used to treat cancer, malaria, inflammation, and virus infection [235,236,237]. In particular, Artemisia montana (A. montana) Pampan engages in various biological activities, including anti-diabetic [238], anti-inflammatory [239], and anti-oxidant activities [240]. The major components of A. montana EO (AM-EO) are β-caryophyllene, germacrene D, 1,8-cineole, and camphor [241]. AM-EO treatment is found to enhance the levels of p-Akt and p-ERK1/2 in HaCaT keratinocytes [241]. In addition, type-IV collagen synthesis promotes basement membrane formation in the skin [242]. Type-IV collagen synthesis is upregulated by AM-EO treatment in HaCaT keratinocytes [241]. AM-EO treatment increases wound closure in the rat tail [241], suggesting that AM-EO reveals a potential positive effect on skin regeneration.

4.2. Pistacia lentiscus L. Plant Oil (PL-PO)

Pistacia lentiscus L., which is known as the lentisk or mastic tree in Greece, belongs to the Anacardiaceae family [243]. Pistacia lentiscus PO (PL-PO) is mainly used as a flavor agent in cuisines but is also administered as an ointment to treat wounds [244]. The major components of PL-PO are oleic, palmitic, linoleic acids, and phenolic compounds, including tocopherols, carotenoids, and anthocyanins [245,246]. PL-PO possesses several biological activities, including anti-bacterial, anti-oxidant, proliferation, and wound healing effects [247,248,249,250]. Treatment with PL-PO liposomes induces the proliferation of HaCaT keratinocytes [251] and enhances the migration of HaCaT keratinocytes [251]. The application of liposomes with PL-PO might be a useful approach in the field of cosmetics.

4.3. Lavender-Essential Oil (L-EO)

L-EO is derived from the blossoms of Lavandula angustifolia [252]. As described in Section 2.13, the major components of L-EO are linalool and linalyl acetate [107]. Based on various reports, L-EO is expected to exhibit beneficial effects in wound healing [253,254,255,256]. Wound area is found to be rapidly reduced by treatment with L-EO compared with that of the control in a rat model [107]. The synthesis of type-III collagen (Col III) a1, an essential component for the formation of granulation tissue in the early phase of wound healing, is upregulated by L-EO treatment in rat wound lesions [107]. In addition, the mRNA levels of Col IIIa1 and Col Ia2 are enhanced by L-EO treatment in a wounded rat model [107]. TGF-β is known to induce the fibroblasts proliferation and production of both Col I and Col III [257]. The level of TGF-β is upregulated by L-EO treatment in the wound lesions of rats [107]. In addition, TGF-β induces the differentiation of fibroblasts to myofibroblasts in wound granulation tissue [258]. Differentiation into myofibroblasts and wound contraction are observed in rat wound lesions treated with L-EO [107].

4.4. Calophyllum inophyllum Plant Oil (CI-PO)

Calophyllum inophyllum (C. inophyllum), a member of the mangosteen family, is a large, evergreen tree of South India, Malaysia, Africa, Polynesia, and the Philippines [259]. C. inophyllum PO (CI-PO; syn: Tamanu oil) is a plant oil acquired from the seeds of the C. inophyllum tree [260]. CI-PO possesses anti-inflammatory [261], anti-microbial [262,263], and anti-fungal [263]. The components of C. inophyllum are calophyllolide, calophyllic acid, inophyllum, and polyphenols that possess wound healing and anti-oxidant properties [264]. Wound area is reduced by treatment with CI-PO through the enhancement of mature granulation and the density of fibrosis and collagen in wounded skin in a rat model [265].

4.5. Parrotiopsis jacquemontiana Plant Oil (PJ-PO)

Parrotiopsis jacquemontiana (P. jacquemontiana) belongs to the Hamamelidaceae family [266] and is reported to possess medicinal properties such as anti-microbial [266] and anti-cancer properties [267]. The major compounds of P. jacquemontiana PO (PJ-PO) are 2, 6-dimethyl-8-oxoocta-2, 6-dienoic acid, syringol, and catechol [268]. Wound contraction is inhibited by treatment with PJ-PO through collagen synthesis in wounded skin in a rat model [268]. In addition, the thickness of the epidermis layer is accompanied by faster fibroblast cells migration from the dermal layer to the epidermal layer in the treated groups [268].

4.6. Chrysanthemum boreale Makino Essential Oil (CB-EO)

The anti-melanogenesis activity of CB-EO has been addressed in Section 3.10. The keratinocyte proliferation is enhanced by treatment with CB-EO through the upregulated Akt and ERK1/2 pathway in HaCaT keratinocyte [196]. In addition to the in vitro model, wound area is decreased by treatment with CB-EO in a wounded tail in a rat model [196].

4.7. Salvia aurea L. Essential Oil (SA-EO)

Salvia aurea L. (syn. S. africana-lutea L.) belongs to the Lamiaceae family [269], and, assuming non-toxicity, Salvia L. genus represents various pharmacological activities, including anti-fungal [270], anti-bacterial [271], anti-inflammatory [272], anti-cancer [273], and anti-oxidant activities [271]. In addition, the Salvia species’ essential oil is reported to possess anti-fungal [274] and anti-viral activities [275]. The major compounds of Salvia aurea L. EO (SA-EO) are 1,8-cineole, β-pinene, cis-thujone, and camphor [276]. Wound closure is increased by treatment with SA-EO in NIH 3T3 fibroblasts [276].

4.8. Rose Plant Oil

Rosa damascena Mill. F. trigintipetala Dieck (Rosaceae) is a plant rich in polyphenolic compounds with various pharmacological properties, including anti-virus, anti-bacterial, anti-oxidant, anti-tussive, anti-diabetic, anti-plasmodial, and anti-inflammatory properties [277]. Polyphenols, especially flavonoids, are well known to possess a broad range of biological activities, such as anti-oxidant, anti-cancer, anti-inflammatory, anti-mutagenic, and anti-proliferative [278]. Quercetin and ellagic acid, which are polyphenols and flavonoids enriched in Carya illinoinensis (Wangenh.) K. Koch, Juglans nigra L., Rosa rugosa Thunb., Prunus domestica (Rosaceae), Malus domestica (Rosaceae), and Prunus avium (Rosaceae), are found to exert anti-proliferative effects on keratinocytes, liver cancer, and breast cancer [232,279,280,281,282,283,284]. In addition, the combined administration of quercetin and ellagic acid induces synergistic effects on apoptosis and anti-proliferation in MOLT-4 human leukemia cells [285]. Thus, treatment with rose plant oil distillation wastewater (RODW) decreases the proliferation and migration of HaCaT keratinocytes [232]. Further, the secretion of the proliferation factor, VEGF, is inhibited by RODW treatment in TNF-α-exposed HaCaT keratinocytes [232]. Apoptotic vesicles are induced by RODW treatment in HaCaT keratinocytes [232]. Thus, RODW might be a useful factor in the alleviation of hyperproliferative skin diseases, such as psoriasis.

4.9. Zanthoxylum bungeanum Maxim Essential Oil (ZB-EO)

Zanthoxylum bungeanum (Z. bungeanum) Maxim is widely distributed in most parts of China and several Southeast Asian countries [286]. Z. bungeanum Maxim EO (ZB-EO) possesses diverse pharmacological properties, such as anti-oxidant [287], anti-cancer [288], anti-inflammatory [289], anti-microbial [290], and insecticidal properties [291]. The major components of ZB-EO are D-limonene and β-myrcene [286]. Treatment with ZB-EO is found to reduce the viability and proliferation of HaCaT keratinocytes through cell cycle arrest [286]. Moreover, the apoptotic effect of ZB-EO is recognized to be mediated by the regulation of apoptosis-associated factors, such as Bax, Bcl-2, and cleaved caspase-3, -8, and -9 in HaCaT keratinocytes [286]. ZB-EO is also considered a potential anti-proliferative drug against hyperproliferative diseases.

4.10. Prunus armeniaca Essential Oil (PA-EO)

Prunus armeniaca L. (P. armeniaca), also known as bitter apricot, belongs to the Rosaceae family and is native to Eurasia and America [292]. Bitter apricot seeds have been used for the alleviation of several skin diseases, including dandruff, acne vulgaris, and furuncle [293]. In addition, Bitter apricot seed possesses a broad range of biological effects, such as anti-cancer [294], anti-oxidant [295,296], anti-microbial [294,295,297], anti-inflammatory [298], and anti-asthmatic [299] activities. Moreover, EO from the apricot seed is reported to possess anti-microbial activity and induce the apoptosis process through regulation of apoptotic factors Bax and Bcl-2 expression [293,300]. The major compounds of P. armeniaca EO (PA-EO) are benzaldehyde, benzoic acid, mandelonitrile [301]. The keratinocyte proliferation is inhibited by treatment with PA-EO or benzaldehyde through upregulated apoptosis-associated factors, such as cleaved Poly(ADP-ribose) polymerase (PARP), caspase-3/8/9, and Bax, and downregulated apoptosis and proliferation-associated factors, such as Bcl-2 and NF-κB, in HaCaT keratinocyte [301].

5. Summary

Naturally derived EOs and other plant-based active components are believed to possess various biological pharmacological properties and are used as raw materials in drug development. Owing to the various functions of EOs and their active components in the skin, they are considered to be potential treatments for different diseases (Table 1). Although EOs have various and positive effects not only on the skin but also on other tissues and organs, EOs are associated with several drawbacks, such as hypersensitivity reactions, appropriate doses, individual reaction differences, toxicity, and side effects. Because of the stratified structure, various cell types, and the presence of pigmentation in the skin tissue, research on the modulation of skin homeostasis should be dealt with in particular. Thus, scientific evidence pertaining to EOs, such as the identification of the biological and physiological mechanisms of EOs, should be developed and reinforced for skin safety concerns. Moreover, it is necessary to overcome skin cancer and skin diseases, including psoriasis, eczema, and fungal infections, through the discovery of numerous potential EO substances as alternatives to synthetic drugs.

Author Contributions

H.J.K. and J.H.H. conceptualized and designed the study and acquired and interpreted the information from PubMed; H.J.K. generated all figures and tables; J.H.H. drafted the manuscript; J.H.H. revised the manuscript critically for important intellectual content, contributed to funding acquisition and final approval of the published version, and was responsible for all aspects of the work, including its accuracy and integrity. All authors have read and agreed to the published version of the manuscript.

Funding

This research was financially supported by a National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT; 2022R1A2C1003890: J.H.H.).

Acknowledgments

All figures were developed by the authors. Generative AI or AI-assisted technologies were not used in the creation of this review.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as potential conflicts of interest.

References

  1. Noorpisheh Ghadimi, S.; Sharifi, N.; Osanloo, M. The leishmanicidal activity of essential oils: A systematic review. J. Herbmed Pharmacol. 2020, 9, 300–308. [Google Scholar] [CrossRef]
  2. Osanloo, M.; Yousefpoor, Y.; Alipanah, H.; Ghanbariasad, A.; Jalilvand, M.; Amani, A. In-vitro Assessment of Essential Oils as Anticancer Therapeutic Agents: A Systematic Literature Review. Jordan J. Pharm. Sci. 2022, 15, 173–203. [Google Scholar] [CrossRef]
  3. Osanloo, M.; Ghaznavi, G.; Abdollahi, A. Surveying the chemical composition and antibacterial activity of essential oils from selected medicinal plants against human pathogens. Iran. J. Microbiol. 2020, 12, 577–583. [Google Scholar] [CrossRef] [PubMed]
  4. Bharti, S.; Rani, N.; Krishnamurthy, B.; Arya, D.S. Preclinical evidence for the pharmacological actions of naringin: A review. Planta Med. 2014, 80, 437–451. [Google Scholar] [CrossRef]
  5. Mohanty, S.; Maurya, A.K.; Jyotshna; Saxena, A.; Shanker, K.; Pal, A.; Bawankule, D.U. Flavonoids rich fraction of Citrus limetta fruit peels reduces proinflammatory cytokine production and attenuates malaria pathogenesis. Curr. Pharm. Biotechnol. 2015, 16, 544–552. [Google Scholar] [CrossRef]
  6. Kondo, M.; Goto, M.; Kodama, A.; Hirose, T. Fractional extraction by supercritical carbon dioxide for the deterpenation of bergamot oil. Ind. Eng. Chem. Res. 2000, 39, 4745–4748. [Google Scholar] [CrossRef]
  7. Misharina, T.A.; Samusenko, A.L. Antioxidant properties of essential oils from lemon, grapefruit, coriander, clove, and their mixtures. Appl. Biochem. Microbiol. 2008, 44, 438–442. [Google Scholar] [CrossRef]
  8. Mitoshi, M.; Kuriyama, I.; Nakayama, H.; Miyazato, H.; Sugimoto, K.; Kobayashi, Y.; Jippo, T.; Kuramochi, K.; Yoshida, H.; Mizushina, Y. Suppression of allergic and inflammatory responses by essential oils derived from herbal plants and citrus fruits. Int. J. Mol. Med. 2014, 33, 1643–1651. [Google Scholar] [CrossRef]
  9. Maurya, A.K.; Mohanty, S.; Pal, A.; Chanotiya, C.S.; Bawankule, D.U. The essential oil from Citrus limetta Risso peels alleviates skin inflammation: In-vitro and in-vivo study. J. Ethnopharmacol. 2018, 212, 86–94. [Google Scholar] [CrossRef]
  10. Freires, I.A.; Denny, C.; Benso, B.; de Alencar, S.M.; Rosalen, P.L. Antibacterial Activity of Essential Oils and Their Isolated Constituents against Cariogenic Bacteria: A Systematic Review. Molecules 2015, 20, 7329–7358. [Google Scholar] [CrossRef]
  11. Trojan-Rodrigues, M.; Alves, T.L.S.; Soares, G.L.G.; Ritter, M.R. Plants used as antidiabetics in popular medicine in Rio Grande do Sul, southern Brazil. J. Ethnopharmacol. 2012, 139, 155–163. [Google Scholar] [CrossRef]
  12. Aga, H.; Shibuya, T.; Sugimoto, T.; Kurimoto, M.; Nakajima, S. Isolation and Identification of Antimicrobial Compounds in Brazilian Propolis. Biosci. Biotechnol. Biochem. 1994, 58, 945–946. [Google Scholar] [CrossRef]
  13. da Silva Filho, A.A.; de Sousa, J.P.; Soares, S.; Furtado, N.A.; Andrade e Silva, M.L.; Cunha, W.R.; Gregorio, L.E.; Nanayakkara, N.P.; Bastos, J.K. Antimicrobial activity of the extract and isolated compounds from Baccharis dracunculifolia D. C. (Asteraceae). Z. Naturforsch. C J. Biosci. 2008, 63, 40–46. [Google Scholar] [CrossRef]
  14. Barbosa, E.V.; Assumpçäo, Y.M.; Teixeira, I.M.; Pereira, R.F.A.; Ribeiro, V.P.; Bastos, J.K.; Cardoso, C.V.; Liberal, M.H.T.; Penna, B.A.; Rocha, L.M. In vitro comparison between antimicrobial and antibiofilm effects of Green Propolis and Baccharis dracunculifolia against Staphylococcus pseudintermedius isolate. Acad. Bras. Cienc. 2022, 94, e20211103. [Google Scholar] [CrossRef]
  15. dos Santos, D.A.; Fukui Mde, J.; Dhammika Nanayakkara, N.P.; Khan, S.I.; Sousa, J.P.; Bastos, J.K.; Andrade, S.F.; da Silva Filho, A.A.; Quintao, N.L. Anti-inflammatory and antinociceptive effects of Baccharis dracunculifolia DC (Asteraceae) in different experimental models. J. Ethnopharmacol. 2010, 127, 543–550. [Google Scholar] [CrossRef]
  16. Bachiega, T.F.; de Sousa, J.P.; Bastos, J.K.; Sforcin, J.M. Immunomodulatory/anti-inflammatory effects of Baccharis dracunculifolia leaves. Nat. Prod. Res. 2013, 27, 1646–1650. [Google Scholar] [CrossRef] [PubMed]
  17. Missima, F.; da Silva Filho, A.A.; Nunes, G.A.; Bueno, P.C.; de Sousa, J.P.; Bastos, J.K.; Sforcin, J.M. Effect of Baccharis dracunculifolia D.C. (Asteraceae) extracts and its isolated compounds on macrophage activation. J. Pharm. Pharmacol. 2007, 59, 463–468. [Google Scholar] [CrossRef]
  18. Brandenburg, M.M.; Rocha, F.G.; Pawloski, P.L.; Soley, B.D.S.; Rockenbach, A.; Scharf, D.R.; Heiden, G.; Ascari, J.; Cabrini, D.A.; Otuki, M.F. Baccharis dracunculifolia (Asteraceae) essential oil displays anti-inflammatory activity in models of skin inflammation. J. Ethnopharmacol. 2020, 259, 112840. [Google Scholar] [CrossRef] [PubMed]
  19. Seema Singh, S.S.; Verma, S.K. Biological activities and therapeutic potential of Perilla frutescens (purple mint): A review. Int. J. Pharm. Sci. Res. 2022, 13, 645–653. [Google Scholar] [CrossRef]
  20. Zi, Y.; Yao, M.; Lu, Z.; Lu, F.; Bie, X.; Zhang, C.; Zhao, H. Glycoglycerolipids from the leaves of Perilla frutescens (L.) Britton (Labiatae) and their anti-inflammatory activities in lipopolysaccharide-stimulated RAW264.7 cells. Phytochemistry 2021, 184, 112679. [Google Scholar] [CrossRef]
  21. Wang, H.S.; Guo, L.; Liu, L.; Han, B.Q.; Niu, X.D. Composite chitosan films prepared using nisin and Perilla frutescense essential oil and their use to extend strawberry shelf life. Food Biosci. 2021, 41, 101037. [Google Scholar] [CrossRef]
  22. Xu, Y.; Shi, Y.; Huang, J.; Gu, H.; Li, C.; Zhang, L.; Liu, G.; Zhou, W.; Du, Z. The Essential Oil Derived from Perilla frutescens (L.) Britt. Attenuates Imiquimod-Induced Psoriasis-like Skin Lesions in BALB/c Mice. Molecules 2022, 27, 2996. [Google Scholar] [CrossRef] [PubMed]
  23. Dillmann, C.; Ringel, C.; Ringleb, J.; Mora, J.; Olesch, C.; Fink, A.F.; Roberts, E.; Brune, B.; Weigert, A. S1PR4 Signaling Attenuates ILT 7 Internalization To Limit IFN-alpha Production by Human Plasmacytoid Dendritic Cells. J. Immunol. 2016, 196, 1579–1590. [Google Scholar] [CrossRef] [PubMed]
  24. Arakura, F.; Hida, S.; Ichikawa, E.; Yajima, C.; Nakajima, S.; Saida, T.; Taki, S. Genetic control directed toward spontaneous IFN-alpha/IFN-beta responses and downstream IFN-gamma expression influences the pathogenesis of a murine psoriasis-like skin disease. J. Immunol. 2007, 179, 3249–3257. [Google Scholar] [CrossRef] [PubMed]
  25. Works, M.G.; Yin, F.; Yin, C.C.; Yiu, Y.; Shew, K.; Tran, T.T.; Dunlap, N.; Lam, J.; Mitchell, T.; Reader, J.; et al. Inhibition of TYK2 and JAK1 ameliorates imiquimod-induced psoriasis-like dermatitis by inhibiting IL-22 and the IL-23/IL-17 axis. J. Immunol. 2014, 193, 3278–3287. [Google Scholar] [CrossRef]
  26. Ajayi-Moses, O.B.; Ogidi, C.O.; Akinyele, B.J. Bioactivity of Citrus essential oils (CEOs) against microorganisms associated with spoilage of some fruits. Chem. Biol. Technol. Ag. 2019, 6, 22. [Google Scholar] [CrossRef]
  27. Raspo, M.A.; Vignola, M.B.; Andreatta, A.E.; Juliani, H.R. Antioxidant and antimicrobial activities of citrus essential oils from Argentina and the United States. Food Biosci. 2020, 36, 100651. [Google Scholar] [CrossRef]
  28. Jing, L.; Lei, Z.; Li, L.; Xie, R.; Xi, W.; Guan, Y.; Sumner, L.W.; Zhou, Z. Antifungal Activity of Citrus Essential Oils. J. Agric. Food Chem. 2014, 62, 3011–3033. [Google Scholar] [CrossRef] [PubMed]
  29. Yang, S.A.; Jeon, S.K.; Lee, E.J.; Shim, C.H.; Lee, I.S. Comparative study of the chemical composition and antioxidant activity of six essential oils and their components. Nat. Prod. Res. 2010, 24, 140–151. [Google Scholar] [CrossRef]
  30. Gonzalez-Mas, M.C.; Rambla, J.L.; Lopez-Gresa, M.P.; Blazquez, M.A.; Granell, A. Volatile Compounds in Citrus Essential Oils: A Comprehensive Review. Front. Plant Sci. 2019, 10, 12. [Google Scholar] [CrossRef]
  31. Njoroge, S.M.; Koaze, H.; Karanja, P.N.; Sawamura, M. Volatile constituents of redblush grapefruit (Citrus paradisi) and pummelo (Citrus grandis) peel essential oils from Kenya. J. Agric. Food Chem. 2005, 53, 9790–9794. [Google Scholar] [CrossRef] [PubMed]
  32. Nelson, E.K.; Mottern, H.H. Florida grapefruit oil. J. Ind. Eng. Chem 1934, 26, 634–637. [Google Scholar] [CrossRef]
  33. Flamini, G.; Cioni, P.L. Odour gradients and patterns in volatile emission of different plant parts and developing fruits of grapefruit (Citrus paradisi L.). Food Chem. 2010, 120, 984–992. [Google Scholar] [CrossRef]
  34. Wang, J.; Rong, L.; Jiang, Z.T. Chemical Composition as Identified by GC-MS and Antioxidant Activity of Grapefruit Mint Essential Oil. Food Sci. 2013, 34, 91–94. [Google Scholar] [CrossRef]
  35. Teixeira, B.; Marques, A.; Ramos, C.; Neng, N.R.; Nogueira, J.M.F.; Saraiva, J.A.; Nunes, M.L. Chemical composition and antibacterial and antioxidant properties of commercial essential oils. Ind. Crop. Prod. 2013, 43, 587–595. [Google Scholar] [CrossRef]
  36. Ahmed, S.; Rattanpal, H.S.; Gul, K.; Dar, R.A.; Sharma, A. Chemical composition, antioxidant activity and GC-MS analysis of juice and peel oil of grapefruit varieties cultivated in India. J. Integr. Agric. 2019, 18, 1634–1642. [Google Scholar] [CrossRef]
  37. Sun, J.; Chu, Y.F.; Wu, X.; Liu, R.H. Antioxidant and antiproliferative activities of common fruits. J. Agric. Food Chem. 2002, 50, 7449–7454. [Google Scholar] [CrossRef] [PubMed]
  38. Nikolic, D.; Bosco, L.; Moschetti, M.; Tinnirello, V.; Pucci, M.; Corleone, V.; Raimondo, S.; Alessandro, R.; Fontana, S. Anti-inflammatory properties of an aldehydes-enriched fraction of grapefruit essential oil. J. Food Sci. 2023, 88, 1172–1187. [Google Scholar] [CrossRef] [PubMed]
  39. Randazzo, W.; Jiménez-Belenguer, A.; Settanni, L.; Perdones, A.; Moschetti, M.; Palazzolo, E.; Guarrasi, V.; Vargas, M.; Germanà, M.A.; Moschetti, G. Antilisterial effect of citrus essential oils and their performance in edible film formulations. Food Control 2016, 59, 750–758. [Google Scholar] [CrossRef]
  40. Negi, P.; Jayaprakasha, G. Antibacterial activity of grapefruit (Citrus paradisi) peel extracts. Eur. Food Res. Technol. 2001, 213, 484–487. [Google Scholar]
  41. Viuda-Martos, M.; Ruiz-Navajas, Y.; Fernández-López, J.; Perez-Álvarez, J. Antibacterial activity of lemon (Citrus lemon L.), mandarin (Citrus reticulata L.), grapefruit (Citrus paradisi L.) and orange (Citrus sinensis L.) essential oils. J. Food Saf. 2008, 28, 567–576. [Google Scholar] [CrossRef]
  42. Uysal, B.; Sozmen, F.; Aktas, O.; Oksal, B.S.; Kose, E.O. Essential oil composition and antibacterial activity of the grapefruit (Citrus paradisi L.) peel essential oils obtained by solvent-free microwave extraction: Comparison with hydrodistillation. Int. J. Food Sci. Technol. 2011, 46, 1455–1461. [Google Scholar] [CrossRef]
  43. Okunowo, W.O.; Oyedeji, O.; Afolabi, L.O.; Matanmi, E. Essential Oil of Grape Fruit (Citrus paradisi) Peels and Its Antimicrobial Activities. Am. J. Plant Sci. 2013, 4, 1–9. [Google Scholar] [CrossRef]
  44. Deng, W.; Liu, K.; Cao, S.; Sun, J.; Zhong, B.; Chun, J. Chemical Composition, Antimicrobial, Antioxidant, and Antiproliferative Properties of Grapefruit Essential Oil Prepared by Molecular Distillation. Molecules 2020, 25, 217. [Google Scholar] [CrossRef] [PubMed]
  45. Stanly, C.; Alfieri, M.; Ambrosone, A.; Leone, A.; Fiume, I.; Pocsfalvi, G. Grapefruit-Derived Micro and Nanovesicles Show Distinct Metabolome Profiles and Anticancer Activities in the A375 Human Melanoma Cell Line. Cells 2020, 9, 2722. [Google Scholar] [CrossRef] [PubMed]
  46. Diab, K.A. In Vitro Studies on Phytochemical Content, Antioxidant, Anticancer, Immunomodulatory, and Antigenotoxic Activities of Lemon, Grapefruit, and Mandarin Citrus Peels. Asian Pac. J. Cancer Prev. 2016, 17, 3559–3567. [Google Scholar] [PubMed]
  47. Kawai, E.; Takeda, R.; Ota, A.; Morita, E.; Imai, D.; Suzuki, Y.; Yokoyama, H.; Ueda, S.; Nakahara, H.; Miyamoto, T.; et al. Increase in diastolic blood pressure induced by fragrance inhalation of grapefruit essential oil is positively correlated with muscle sympathetic nerve activity. J. Physiol. Sci. 2020, 70, 2. [Google Scholar] [CrossRef]
  48. Denkova-Kostova, R.; Teneva, D.; Tomova, T.; Goranov, B.; Denkova, Z.; Shopska, V.; Slavchev, A.; Hristova-Ivanova, Y. Chemical composition, antioxidant and antimicrobial activity of essential oils from tangerine (Citrus reticulata L.), grapefruit (Citrus paradisi L.), lemon (Citrus lemon L.) and cinnamon (Cinnamomum zeylanicum Blume). Z. Naturforsch. C J. Biosci. 2021, 76, 175–185. [Google Scholar] [CrossRef]
  49. d’alessio, P.A.; Mirshahi, M.; Bisson, J.F.; Bene, M.C. Skin repair properties of d-Limonene and perillyl alcohol in murine models. Antiinflamm. Antiallergy Agents Med. Chem. 2014, 13, 29–35. [Google Scholar] [CrossRef]
  50. Zhang, X.; Xu, H.; Hua, J.; Zhu, Z.; Wang, M. Protective Effects of Grapefruit Essential Oil against Staphylococcus aureus-Induced Inflammation and Cell Damage in Human Epidermal Keratinocytes. Chem. Biodivers. 2022, 19, e202200205. [Google Scholar] [CrossRef]
  51. Avola, R.; Granata, G.; Geraci, C.; Napoli, E.; Graziano, A.C.E.; Cardile, V. Oregano (Origanum vulgare L.) essential oil provides anti-inflammatory activity and facilitates wound healing in a human keratinocytes cell model. Food Chem. Toxicol. 2020, 144, 111586. [Google Scholar] [CrossRef] [PubMed]
  52. Chun, S.S.; Vattem, D.A.; Lin, Y.T.; Shetty, K. Phenolic antioxidants from clonal oregano (Origanum vulgare) with antimicrobial activity against Helicobacter pylori. Process Biochem. 2005, 40, 809–816. [Google Scholar] [CrossRef]
  53. Wojdylo, A.; Oszmianski, J.; Czemerys, R. Antioxidant activity and phenolic compounds in 32 selected herbs. Food Chem. 2007, 105, 940–949. [Google Scholar] [CrossRef]
  54. Ocana-Fuentes, A.; Arranz-Gutierrez, E.; Senorans, F.J.; Reglero, G. Supercritical fluid extraction of oregano (Origanum vulgare) essentials oils: Anti-inflammatory properties based on cytokine response on THP-1 macrophages. Food Chem. Toxicol. 2010, 48, 1568–1575. [Google Scholar] [CrossRef]
  55. Sahin, F.; Güllüce, M.; Daferera, D.; Sökmen, A.; Sökmen, M.; Polissiou, M.; Agar, G.; Özer, H. Biological activities of the essential oils and methanol extract of Origanum vulgare ssp vulgare in the Eastern Anatolia region of Turkey. Food Control 2004, 15, 549–557. [Google Scholar] [CrossRef]
  56. Granata, G.; Stracquadanio, S.; Leonardi, M.; Napoli, E.; Consoli, G.M.L.; Cafiso, V.; Stefani, S.; Geraci, C. Essential oils encapsulated in polymer-based nanocapsules as potential candidates for application in food preservation. Food Chem. 2018, 269, 286–292. [Google Scholar] [CrossRef] [PubMed]
  57. Loizzo, M.R.; Menichini, F.; Conforti, F.; Tundis, R.; Bonesi, M.; Saab, A.M.; Statti, G.A.; Cindio, B.d.; Houghton, P.J.; Menichini, F.; et al. Chemical analysis, antioxidant, antiinflammatory and anticholinesterase activities of Origanum ehrenbergii Boiss and Origanum syriacum L. essential oils. Food Chem. 2009, 117, 174–180. [Google Scholar] [CrossRef]
  58. Bukovska, A.; Cikos, S.; Juhas, S.; Il’kova, G.; Rehak, P.; Koppel, J. Effects of a combination of thyme and oregano essential oils on TNBS-induced colitis in mice. Mediat. Inflamm. 2007, 2007, 23296. [Google Scholar] [CrossRef] [PubMed]
  59. Omarizadeh, K.; Farahpour, M.R.; Alipour, M. Topical Administration of an Ointment Prepared From Satureja sahendica Essential Oil Accelerated Infected Full-Thickness Wound Healing by Modulating Inflammatory Response in a Mouse Model. Wounds 2021, 33, 321–328. [Google Scholar] [CrossRef]
  60. Sefidkon, F.; Jamzad, Z.; Mirza, M. Chemical variation in the essential oil of Satureja sahendica from Iran. Food Chem. 2004, 88, 325–328. [Google Scholar] [CrossRef]
  61. Eftekhar, F.; Raei, F.; Yousefzadi, M.; Ebrahimi, S.N.; Hadian, J. Antibacterial activity and essential oil composition of Satureja spicigera from Iran. Z. Naturforsch. C J. Biosci. 2009, 64, 20–24. [Google Scholar] [CrossRef] [PubMed]
  62. Ghazanfari, G.; Minaie, B.; Yasa, N.; Nakhai, L.A.; Mohammadirad, A.; Nikfar, S.; Dehghan, G.; Boushehri, V.S.; Jamshidi, H.; Khorasani, R.; et al. Biochemical and histopathological evidences for beneficial effects of Satureja khuzestanica jamzad essential oil on the mouse model of inflammatory bowel diseases. Toxicol. Mech. Methods 2006, 16, 365–372. [Google Scholar] [CrossRef]
  63. Hadian, N.; Rezaei, M.; Mosayebi, Z.; Meshkani, F. CO2 reforming of methane over nickel catalysts supported on nanocrystalline MgAl2O4 with high surface area. J. Nat. Gas Chem. 2012, 21, 200–206. [Google Scholar] [CrossRef]
  64. Hajilou, H.; Farahpour, M.R.; Hamishehkar, H. Polycaprolactone nanofiber coated with chitosan and functionalized as a novel wound dressing for healing infected wounds. Int. J. Biol. Macromol. 2020, 164, 2358–2369. [Google Scholar] [CrossRef] [PubMed]
  65. Pakyari, M.; Farrokhi, A.; Maharlooei, M.K.; Ghahary, A. Critical Role of Transforming Growth Factor Beta in Different Phases of Wound Healing. Adv. Wound Care 2013, 2, 215–224. [Google Scholar] [CrossRef] [PubMed]
  66. Haleagrahara, N.; Chakravarthi, S.; Mathews, L. Insulin like growth factor-1 (IGF-1) causes overproduction of IL-8, an angiogenic cytokine and stimulates neovascularization in isoproterenol-induced myocardial infarction in rats. Int. J. Mol. Sci. 2011, 12, 8562–8574. [Google Scholar] [CrossRef]
  67. Metcalfe, A.D.; Ferguson, M.W. Tissue engineering of replacement skin: The crossroads of biomaterials, wound healing, embryonic development, stem cells and regeneration. J. R. Soc. Interface 2007, 4, 413–437. [Google Scholar] [CrossRef] [PubMed]
  68. O’Neill, B.T.; Lauritzen, H.P.; Hirshman, M.F.; Smyth, G.; Goodyear, L.J.; Kahn, C.R. Differential Role of Insulin/IGF-1 Receptor Signaling in Muscle Growth and Glucose Homeostasis. Cell Rep. 2015, 11, 1220–1235. [Google Scholar] [CrossRef]
  69. Ramirez, H.; Patel, S.B.; Pastar, I. The Role of TGFbeta Signaling in Wound Epithelialization. Adv. Wound Care 2014, 3, 482–491. [Google Scholar] [CrossRef]
  70. Chen, G.; Lv, C.; Nie, Q.; Li, X.; Lv, Y.; Liao, G.; Liu, S.; Ge, W.; Chen, J.; Du, Y. Essential Oil of Matricaria chamomilla Alleviate Psoriatic-Like Skin Inflammation by Inhibiting PI3K/Akt/mTOR and p38MAPK Signaling Pathway. Clin. Cosmet. Investig. Dermatol. 2024, 17, 59–77. [Google Scholar] [CrossRef]
  71. Kroll, U.; Cordes, C. Pharmaceutical prerequisites for a multi-target therapy. Phytomedicine 2006, 13 (Suppl. 5), 12–19. [Google Scholar] [CrossRef] [PubMed]
  72. De Cicco, P.; Ercolano, G.; Sirignano, C.; Rubino, V.; Rigano, D.; Ianaro, A.; Formisano, C. Chamomile essential oils exert anti-inflammatory effects involving human and murine macrophages: Evidence to support a therapeutic action. J. Ethnopharmacol. 2023, 311, 116391. [Google Scholar] [CrossRef] [PubMed]
  73. Forster, H.B.; Niklas, H.; Lutz, S. Antispasmodic effects of some medicinal plants. Planta Med. 1980, 40, 309–319. [Google Scholar] [CrossRef] [PubMed]
  74. Lin, T.K.; Zhong, L.; Santiago, J.L. Anti-Inflammatory and Skin Barrier Repair Effects of Topical Application of Some Plant Oils. Int. J. Mol. Sci. 2017, 19, 70. [Google Scholar] [CrossRef] [PubMed]
  75. Wang, W.; Wang, Y.; Zou, J.; Jia, Y.; Wang, Y.; Li, J.; Wang, C.; Sun, J.; Guo, D.; Wang, F.; et al. The Mechanism Action of German Chamomile (Matricaria recutita L.) in the Treatment of Eczema: Based on Dose-Effect Weight Coefficient Network Pharmacology. Front. Pharmacol. 2021, 12, 706836. [Google Scholar] [CrossRef] [PubMed]
  76. Lee, S.H.; Heo, Y.; Kim, Y.C. Effect of German chamomile oil application on alleviating atopic dermatitis-like immune alterations in mice. J. Vet. Sci. 2010, 11, 35–41. [Google Scholar] [CrossRef]
  77. Fecker, R.; Magyari-Pavel, I.Z.; Cocan, I.; Alexa, E.; Popescu, I.M.; Lombrea, A.; Bora, L.; Dehelean, C.A.; Buda, V.; Folescu, R.; et al. Oxidative Stability and Protective Effect of the Mixture between Helianthus annuus L. and Oenothera biennis L. Oils on 3D Tissue Models of Skin Irritation and Phototoxicity. Plants 2022, 11, 2977. [Google Scholar] [CrossRef] [PubMed]
  78. Zeng, J.; Lei, L.; Zeng, Q.; Yao, Y.; Wu, Y.; Li, Q.; Gao, L.; Du, H.; Xie, Y.; Huang, J.; et al. Ozone Therapy Attenuates NF-kappaB-Mediated Local Inflammatory Response and Activation of Th17 Cells in Treatment for Psoriasis. Int. J. Biol. Sci. 2020, 16, 1833–1845. [Google Scholar] [CrossRef] [PubMed]
  79. Kim, S.Y.; Lee, J.O.; Lee, S.; Heo, J.; Cho, K.H.; Bahuguna, A.; Yoo, K.H.; Kim, B.J. Ozonated Sunflower Oil (OSO) Alleviates Inflammatory Responses in Oxazolone-Induced Atopic Dermatitis (AD)-Like Mice and LPS-Treated RAW 264.7 Cells. J. Microbiol. Biotechnol. 2024, 34, 765–773. [Google Scholar] [CrossRef]
  80. Kim, S.Y.; Han, S.D.; Kim, M.; Mony, T.J.; Lee, E.S.; Kim, K.M.; Choi, S.H.; Hong, S.H.; Choi, J.W.; Park, S.J. Mentha arvensis Essential Oil Exerts Anti-Inflammatory in LPS-Stimulated Inflammatory Responses via Inhibition of ERK/NF-kappaB Signaling Pathway and Anti-Atopic Dermatitis-like Effects in 2,4-Dinitrochlorobezene-Induced BALB/c Mice. Antioxidants 2021, 10, 1941. [Google Scholar] [CrossRef]
  81. Salehi, B.; Stojanovic-Radic, Z.; Matejic, J.; Sharopov, F.; Antolak, H.; Kregiel, D.; Sen, S.; Sharifi-Rad, M.; Acharya, K.; Sharifi-Rad, R.; et al. Plants of Genus Mentha: From Farm to Food Factory. Plants 2018, 7, 70. [Google Scholar] [CrossRef]
  82. Dong-Hun Choi, O.-T.S.; Lim, M.-H. Comparative study of the biological activities effect of Mentha arvensis L. extracts from water and 80% ethanol. J. Korean Appl. Sci. Technol. 2019, 36, 208–216. [Google Scholar] [CrossRef]
  83. Kim, S.Y.; Sapkota, A.; Bae, Y.J.; Choi, S.H.; Bae, H.J.; Kim, H.J.; Cho, Y.E.; Choi, Y.Y.; An, J.Y.; Cho, S.Y.; et al. The Anti-Atopic Dermatitis Effects of Mentha arvensis Essential Oil Are Involved in the Inhibition of the NLRP3 Inflammasome in DNCB-Challenged Atopic Dermatitis BALB/c Mice. Int. J. Mol. Sci. 2023, 24, 7720. [Google Scholar] [CrossRef] [PubMed]
  84. Li, J.K.; Duan, J.W.; Wang, Y.J.; Zhou, P.J.; Wang, X.; Xia, N.; Wang, J.; Li, J.; Wang, W.F.; Wang, X.; et al. The JAK/STAT/NF-κB signaling pathway can be regulated by rosemary essential oil, thereby providing a potential treatment for DNCB-induced in mice. Biomed. Pharmacother. 2023, 168, 115727. [Google Scholar] [CrossRef]
  85. Jamila, F.; Mostafa, E. Ethnobotanical survey of medicinal plants used by people in Oriental Morocco to manage various ailments. J. Ethnopharmacol. 2014, 154, 76–87. [Google Scholar] [CrossRef]
  86. Takaki, I.; Bersani-Amado, L.E.; Vendruscolo, A.; Sartoretto, S.M.; Diniz, S.P.; Bersani-Amado, C.A.; Cuman, R.K. Anti-inflammatory and antinociceptive effects of Rosmarinus officinalis L. essential oil in experimental animal models. J. Med. Food 2008, 11, 741–746. [Google Scholar] [CrossRef] [PubMed]
  87. Sanjay Jain, S.S.; Satish Nayak, S. Sumbhate. Plant Review Recent trends in Curcuma Longa Linn. Pharmacogn. Rev. 2007, 1, 119–128. [Google Scholar]
  88. Srivilai, J.; Waranuch, N.; Tangsumranjit, A.; Khorana, N.; Ingkaninan, K. Germacrone and sesquiterpene-enriched extracts from Curcuma aeruginosa Roxb. increase skin penetration of minoxidil, a hair growth promoter. Drug Deliv. Transl. Res. 2018, 8, 140–149. [Google Scholar] [CrossRef] [PubMed]
  89. Kumar, A.; Agarwal, K.; Singh, M.; Saxena, A.; Yadav, P.; Maurya, A.K.; Yadav, A.; Tandon, S.; Chanda, D.; Bawankule, D.U. Essential oil from waste leaves of Curcuma longa L. alleviates skin inflammation. Inflammopharmacology 2018, 26, 1245–1255. [Google Scholar] [CrossRef]
  90. Chen, L.L.; Zhang, H.J.; Chao, J.; Liu, J.F. Essential oil of Artemisia argyi suppresses inflammatory responses by inhibiting JAK/STATs activation. J. Ethnopharmacol. 2017, 204, 107–117. [Google Scholar] [CrossRef]
  91. Huang, H.C.; Wang, H.F.; Yih, K.H.; Chang, L.Z.; Chang, T.M. Dual Bioactivities of Essential Oil Extracted from the Leaves of Artemisia argyi as an Antimelanogenic versus Antioxidant Agent and Chemical Composition Analysis by GC/MS. Int. J. Mol. Sci. 2012, 13, 14679–14697. [Google Scholar] [CrossRef] [PubMed]
  92. Nakasugi, T.; Nakashima, M.; Komai, K. Antimutagens in gaiyou (Artemisia argyi levl. et vant.). J. Agric. Food Chem. 2000, 48, 3256–3266. [Google Scholar] [CrossRef] [PubMed]
  93. Adams, M.; Efferth, T.; Bauer, R. Activity-guided isolation of scopoletin and isoscopoletin, the inhibitory active principles towards CCRF-CEM leukaemia cells and multi-drug resistant CEM/ADR5000 cells, from Artemisia argyi. Planta Med. 2006, 72, 862–864. [Google Scholar] [CrossRef] [PubMed]
  94. Jeong, M.A.; Lee, K.W.; Yoon, D.Y.; Lee, H.J. Jaceosidin, a pharmacologically active flavone derived from Artemisia argyi, inhibits phorbol-ester-induced upregulation of COX-2 and MMP-9 by blocking phosphorylation of ERK-1 and -2 in cultured human mammary epithelial cells. Ann. N. Y. Acad. Sci. 2007, 1095, 458–466. [Google Scholar] [CrossRef] [PubMed]
  95. Seo, J.M.; Kang, H.M.; Son, K.H.; Kim, J.H.; Lee, C.W.; Kim, H.M.; Chang, S.I.; Kwon, B.M. Antitumor activity of flavones isolated from Artemisia argyi. Planta Med. 2003, 69, 218–222. [Google Scholar] [CrossRef] [PubMed]
  96. Bao, X.; Yuan, H.; Wang, C.; Liu, J.; Lan, M. Antitumor and immunomodulatory activities of a polysaccharide from Artemisia argyi. Carbohydr. Polym. 2013, 98, 1236–1243. [Google Scholar] [CrossRef]
  97. Ge, Y.B.; Wang, Z.G.; Xiong, Y.; Huang, X.J.; Mei, Z.N.; Hong, Z.G. Anti-inflammatory and blood stasis activities of essential oil extracted from Artemisia argyi leaf in animals. J. Nat. Med. 2016, 70, 531–538. [Google Scholar] [CrossRef] [PubMed]
  98. Bian, R.L. Pharmacological studies on the antiasthmatic principle of essential oil of Artemisia argyi--terpinenol-4 (author’s transl). Zhonghua Jie He He Hu Xi Xi Ji Bing Za Zhi 1981, 4, 203–206. [Google Scholar] [PubMed]
  99. Wenqiang, G.; Shufen, L.; Ruixiang, Y.; Yanfeng, H. Comparison of composition and antifungal activity of Artemisia argyi Levl. et Vant inflorescence essential oil extracted by hydrodistillation and supercritical carbon dioxide. Nat. Prod. Res. 2006, 20, 992–998. [Google Scholar] [CrossRef]
  100. Cavanagh, H.M.; Wilkinson, J.M. Biological activities of lavender essential oil. Phytother. Res. 2002, 16, 301–308. [Google Scholar] [CrossRef]
  101. Chioca, L.R.; Ferro, M.M.; Baretta, I.P.; Oliveira, S.M.; Silva, C.R.; Ferreira, J.; Losso, E.M.; Andreatini, R. Anxiolytic-like effect of lavender essential oil inhalation in mice: Participation of serotonergic but not GABAA/benzodiazepine neurotransmission. J. Ethnopharmacol. 2013, 147, 412–418. [Google Scholar] [CrossRef] [PubMed]
  102. Xu, P.; Wang, K.; Lu, C.; Dong, L.; Gao, L.; Yan, M.; Aibai, S.; Liu, X. Protective effect of lavender oil on scopolamine induced cognitive deficits in mice and H(2)O(2) induced cytotoxicity in PC12 cells. J. Ethnopharmacol. 2016, 193, 408–415. [Google Scholar] [CrossRef] [PubMed]
  103. Silva, G.L.; Luft, C.; Lunardelli, A.; Amaral, R.H.; Melo, D.A.; Donadio, M.V.; Nunes, F.B.; de Azambuja, M.S.; Santana, J.C.; Moraes, C.M.; et al. Antioxidant, analgesic and anti-inflammatory effects of lavender essential oil. Acad. Bras. Cienc. 2015, 87, 1397–1408. [Google Scholar] [CrossRef] [PubMed]
  104. Cardia, G.F.E.; Silva-Filho, S.E.; Silva, E.L.; Uchida, N.S.; Cavalcante, H.A.O.; Cassarotti, L.L.; Salvadego, V.E.C.; Spironello, R.A.; Bersani-Amado, C.A.; Cuman, R.K.N. Effect of Lavender (Lavandula angustifolia) Essential Oil on Acute Inflammatory Response. Evid. Based Complement. Altern. Med. 2018, 2018, 1413940. [Google Scholar] [CrossRef] [PubMed]
  105. Adaszynska-Skwirzynska, M.; Szczerbinska, D. The antimicrobial activity of lavender essential oil (Lavandula angustifolia) and its influence on the production performance of broiler chickens. J. Anim. Physiol. Anim. Nutr. 2018, 102, 1020–1025. [Google Scholar] [CrossRef] [PubMed]
  106. de Rapper, S.; Kamatou, G.; Viljoen, A.; van Vuuren, S. The In Vitro Antimicrobial Activity of Lavandula angustifolia Essential Oil in Combination with Other Aroma-Therapeutic Oils. Evid. Based Complement. Altern. Med. 2013, 2013, 852049. [Google Scholar] [CrossRef] [PubMed]
  107. Mori, H.M.; Kawanami, H.; Kawahata, H.; Aoki, M. Wound healing potential of lavender oil by acceleration of granulation and wound contraction through induction of TGF-beta in a rat model. BMC Complement. Altern. Med. 2016, 16, 144. [Google Scholar] [CrossRef] [PubMed]
  108. Nasiri, A.; Mahmodi, M.A.; Nobakht, Z. Effect of aromatherapy massage with lavender essential oil on pain in patients with osteoarthritis of the knee: A randomized controlled clinical trial. Complement. Ther. Clin. Pract. 2016, 25, 75–80. [Google Scholar] [CrossRef] [PubMed]
  109. Rai, V.K.; Sinha, P.; Yadav, K.S.; Shukla, A.; Saxena, A.; Bawankule, D.U.; Tandon, S.; Khan, F.; Chanotiya, C.S.; Yadav, N.P. Anti-psoriatic effect of Lavandula angustifolia essential oil and its major components linalool and linalyl acetate. J. Ethnopharmacol. 2020, 261, 113127. [Google Scholar] [CrossRef]
  110. Peana, A.T.; D’Aquila, P.S.; Panin, F.; Serra, G.; Pippia, P.; Moretti, M.D. Anti-inflammatory activity of linalool and linalyl acetate constituents of essential oils. Phytomedicine 2002, 9, 721–726. [Google Scholar] [CrossRef]
  111. Shibata, C.; Sasaki, I.; Naito, H.; Ueno, T.; Matsuno, S. The herbal medicine Dai-Kenchu-Tou stimulates upper gut motility through cholinergic and 5-hydroxytryptamine 3 receptors in conscious dogs. Surgery 1999, 126, 918–924. [Google Scholar] [CrossRef] [PubMed]
  112. Cho, M.G.; Chang, C.S.; Chae, Y.A. Variation of volatile composition in the leaf of Zanthoxylum schinofolium siebold et zucc. and Zanthoxylum piperitum DC. Korean J. Med. Crop. Sci. 2002, 10, 162–166. [Google Scholar]
  113. Choi, H.J. Evaluation of Antiviral Activity of Zanthoxylum Species Against Picornaviruses. Osong. Public Health Res. Perspect. 2016, 7, 400–403. [Google Scholar] [CrossRef] [PubMed]
  114. Guo, R.H.; Park, J.U.; Jo, S.J.; Ahn, J.H.; Park, J.H.; Yang, J.Y.; Lee, S.S.; Park, M.J.; Kim, Y.R. Anti-allergic Inflammatory Effects of the Essential Oil From Fruits of Zanthoxylum coreanum Nakai. Front. Pharmacol. 2018, 9, 1441. [Google Scholar] [CrossRef] [PubMed]
  115. Gautam, N.; Mantha, A.K.; Mittal, S. Essential Oils and Their Constituents as Anticancer Agents: A Mechanistic View. Biomed Res. Int. 2014, 2014, 154106. [Google Scholar] [CrossRef] [PubMed]
  116. Tayarani-Najaran, Z.; Akaberi, M.; Vatani, M.; Emami, S.A. Evaluation of antioxidant and anti-melanogenic activities of different extracts from aerial parts of Nepeta binaludensis Jamzad in murine melanoma B16F10 cells. Iran. J. Basic Med. Sci. 2016, 19, 662–669. [Google Scholar]
  117. Videira, I.F.; Moura, D.F.; Magina, S. Mechanisms regulating melanogenesis. Bras. Dermatol. 2013, 88, 76–83. [Google Scholar] [CrossRef]
  118. Kim, M.J.; Mohamed, E.A.; Kim, D.S.; Park, M.J.; Ahn, B.J.; Jeung, E.B.; An, B.S. Inhibitory effects and underlying mechanisms of Artemisia capillaris essential oil on melanogenesis in the B16F10 cell line. Mol. Med. Rep. 2022, 25, 1–12. [Google Scholar] [CrossRef]
  119. Munoz-Munoz, J.L.; Garcia-Molina, F.; Varon, R.; Tudela, J.; Garcia-Canovas, F.; Rodriguez-Lopez, J.N. Generation of hydrogen peroxide in the melanin biosynthesis pathway. Biochim. Biophys. Acta 2009, 1794, 1017–1029. [Google Scholar] [CrossRef]
  120. Perluigi, M.; De Marco, F.; Foppoli, C.; Coccia, R.; Blarzino, C.; Marcante, M.L.; Cini, C. Tyrosinase protects human melanocytes from ROS-generating compounds. Biochem. Biophys. Res. Commun. 2003, 305, 250–256. [Google Scholar] [CrossRef]
  121. Liu, G.S.; Peshavariya, H.; Higuchi, M.; Brewer, A.C.; Chang, C.W.; Chan, E.C.; Dusting, G.J. Microphthalmia-associated transcription factor modulates expression of NADPH oxidase type 4: A negative regulator of melanogenesis. Free Radic. Biol. Med. 2012, 52, 1835–1843. [Google Scholar] [CrossRef]
  122. Yasui, H.; Sakurai, H. Age-dependent generation of reactive oxygen species in the skin of live hairless rats exposed to UVA light. Exp. Dermatol. 2003, 12, 655–661. [Google Scholar] [CrossRef] [PubMed]
  123. Funasaka, Y.; Komoto, M.; Ichihashi, M. Depigmenting effect of alpha-tocopheryl ferulate on normal human melanocytes. Pigment. Cell Res. 2000, 13 (Suppl. 8), 170–174. [Google Scholar] [CrossRef]
  124. Bahramsoltani, R.; Rostamiasrabadi, P.; Shahpiri, Z.; Marques, A.M.; Rahimi, R.; Farzaei, M.H. Aloysia citrodora Paláu (Lemon verbena): A review of phytochemistry and pharmacology. J. Ethnopharmacol. 2018, 222, 34–51. [Google Scholar] [CrossRef]
  125. Spyridopoulou, K.; Aravidou, T.; Lampri, E.; Effraimidou, E.; Pappa, A.; Chlichlia, K. Antitumor Potential of Lippia citriodora Essential Oil in Breast Tumor-Bearing Mice. Antioxidants 2021, 10, 875. [Google Scholar] [CrossRef]
  126. Karimivaselabadi, A.; Osanloo, M.; Ghanbariasad, A.; Zarenezhad, E.; Hosseini, H. Comparison of chitosan nanoparticles containing Lippia citriodora essential oil and citral on the induction of apoptosis in A375 melanoma cells. BMC Complement. Med. Ther. 2023, 23, 435. [Google Scholar] [CrossRef] [PubMed]
  127. Najar, B.; Shortrede, J.E.; Pistelli, L.; Buhagiar, J. Chemical Composition and in Vitro Cytotoxic Screening of Sixteen Commercial Essential Oils on Five Cancer Cell Lines. Chem. Biodivers. 2020, 17, e1900478. [Google Scholar] [CrossRef]
  128. Zeng, S.; Kapur, A.; Patankar, M.S.; Xiong, M.P. Formulation, Characterization, and Antitumor Properties of Trans- and Cis-Citral in the 4T1 Breast Cancer Xenograft Mouse Model. Pharm. Res. 2015, 32, 2548–2558. [Google Scholar] [CrossRef] [PubMed]
  129. Oukerrou, M.A.; Tilaoui, M.; Mouse, H.A.; Bouchmaa, N.; Zyad, A. Differential Cytotoxic Activity of Essential Oil of Lippia citriodora from Different Regions in Morocco. Chem. Biodivers. 2017, 14, 1–11. [Google Scholar] [CrossRef]
  130. Fitsiou, E.; Mitropoulou, G.; Spyridopoulou, K.; Vamvakias, M.; Bardouki, H.; Galanis, A.; Chlichlia, K.; Kourkoutas, Y.; Panayiotidis, M.; Pappa, A. Chemical Composition and Evaluation of the Biological Properties of the Essential Oil of the Dietary Phytochemical Lippia citriodora. Molecules 2018, 23, 123. [Google Scholar] [CrossRef]
  131. Lertsatitthanakorn, P.; Taweechaisupapong, S.; Aromdee, C.; Khunkitti, W. In vitro bioactivities of essential oils used for acne control. Int. J. Aromather. 2006, 16, 43–49. [Google Scholar] [CrossRef]
  132. Farahmandfar, R.; Asnaashari, M.; Pourshayegan, M.; Maghsoudi, S.; Moniri, H. Evaluation of antioxidant properties of lemon verbena (Lippia citriodora) essential oil and its capacity in sunflower oil stabilization during storage time. Food Sci. Nutr. 2018, 6, 983–990. [Google Scholar] [CrossRef] [PubMed]
  133. Salama, Y.; Jaradat, N.; Hattori, K.; Heissig, B. Aloysia Citrodora Essential Oil Inhibits Melanoma Cell Growth and Migration by Targeting HB-EGF-EGFR Signaling. Int. J. Mol. Sci. 2021, 22, 8151. [Google Scholar] [CrossRef] [PubMed]
  134. Kawasaki, K.; Kawakami, T.; Watabe, H.; Itoh, F.; Mizoguchi, M.; Soma, Y. Expression of matrilysin (matrix metalloproteinase-7) in primary cutaneous and metastatic melanoma. Br. J. Dermatol. 2007, 156, 613–619. [Google Scholar] [CrossRef] [PubMed]
  135. Zigrino, P.; Nischt, R.; Mauch, C. The Disintegrin-like and Cysteine-rich domains of ADAM-9 Mediate Interactions between Melanoma Cells and Fibroblasts. J. Biol. Chem. 2011, 286, 6801–6807. [Google Scholar] [CrossRef] [PubMed]
  136. Sternlicht, M.D.; Werb, Z. How matrix metalloproteinases regulate cell behavior. Annu. Rev. Cell Dev. Biol. 2001, 17, 463–516. [Google Scholar] [CrossRef] [PubMed]
  137. Heissig, B.; Hattori, K.; Friedrich, M.; Rafii, S.; Werb, Z. Angiogenesis: Vascular remodeling of the extracellular matrix involves metalloproteinases. Curr. Opin. Hematol. 2003, 10, 136–141. [Google Scholar] [CrossRef]
  138. Villanueva, J.; Herlyn, M. Melanoma and the tumor microenvironment. Curr. Oncol. Rep. 2008, 10, 439–446. [Google Scholar] [CrossRef]
  139. Winkler, J.; Abisoye-Ogunniyan, A.; Metcalf, K.J.; Werb, Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat. Commun. 2020, 11, 5120. [Google Scholar] [CrossRef]
  140. Hudson, L.G.; Moss, N.M.; Stack, M.S. EGF-receptor regulation of matrix metalloproteinases in epithelial ovarian carcinoma. Future Oncol. 2009, 5, 323–338. [Google Scholar] [CrossRef]
  141. Mader, C.C.; Oser, M.; Magalhaes, M.A.; Bravo-Cordero, J.J.; Condeelis, J.; Koleske, A.J.; Gil-Henn, H. An EGFR-Src-Arg-cortactin pathway mediates functional maturation of invadopodia and breast cancer cell invasion. Cancer Res. 2011, 71, 1730–1741. [Google Scholar] [CrossRef] [PubMed]
  142. Osanloo, M.; Alipanah, H.; Farjam, M.; Taheri, A.; Zarenezhad, E. Anticancer activity of Chitosan nanoparticles containing Satureja khuzistanica essential oil, and carvacrol against human Melanoma and Breast Cancer. Russ. J. Bioorg. Chem. 2023, 49, 594–601. [Google Scholar] [CrossRef]
  143. Alipanah, H.; Farjam, M.; Zarenezhad, E.; Roozitalab, G.; Osanloo, M. Chitosan nanoparticles containing limonene and limonene-rich essential oils: Potential phytotherapy agents for the treatment of melanoma and breast cancers. BMC Complement. Med. Ther. 2021, 21, 186. [Google Scholar] [CrossRef] [PubMed]
  144. Qasemi, H.; Fereidouni, Z.; Karimi, J.; Abdollahi, A.; Zarenezhad, E.; Rasti, F.; Osanloo, M. Promising antibacterial effect of impregnated nanofiber mats with a green nanogel against clinical and standard strains of Pseudomonas aeruginosa and Staphylococcus aureus. J. Drug Deliv. Sci. Technol. 2021, 66, 102844. [Google Scholar] [CrossRef]
  145. Rasti, F.; Yousefpoor, Y.; Abdollahi, A.; Safari, M.; Roozitalab, G.; Osanloo, M. Antioxidative, anticancer, and antibacterial activities of a nanogel containing Mentha spicata L. essential oil and electrospun nanofibers of polycaprolactone-hydroxypropyl methylcellulose. BMC Complement. Med. Ther. 2022, 22, 261. [Google Scholar] [CrossRef]
  146. Valizadeh, A.; Khaleghi, A.A.; Alipanah, H.; Zarenezhad, E.; Osanloo, M. Anticarcinogenic Effect of Chitosan Nanoparticles Containing Syzygium aromaticum Essential Oil or Eugenol Toward Breast and Skin Cancer Cell Lines. Bionanoscience 2021, 11, 678–686. [Google Scholar] [CrossRef]
  147. Rajivgandhi, G.; Saravanan, K.; Ramachandran, G.; Li, J.L.; Yin, L.Z.; Quero, F.; Alharbi, N.S.; Kadaikunnan, S.; Khaled, J.M.; Manoharan, N.; et al. Enhanced anti-cancer activity of chitosan loaded Morinda citrifolia essential oil against A549 human lung cancer cells. Int. J. Biol. Macromol. 2020, 164, 4010–4021. [Google Scholar] [CrossRef]
  148. Hesami, S.; Safi, S.; Larijani, K.; Badi, H.N.; Abdossi, V.; Hadidi, M. Synthesis and characterization of chitosan nanoparticles loaded with greater celandine (Chelidonium majus L.) essential oil as an anticancer agent on MCF-7 cell line. Int. J. Biol. Macromol. 2022, 194, 974–981. [Google Scholar] [CrossRef] [PubMed]
  149. Rasti, F.; Ahmadi, E.; Safari, M.; Abdollahi, A.; Satvati, S.; Ranjbar, R.; Osanloo, M. Anticancer, antioxidant, and antibacterial effects of nanoemulsion of Origanum majorana essential oil. Iran. J. Microbiol. 2023, 15, 565–573. [Google Scholar] [CrossRef]
  150. Prakash, B.; Singh, P.; Kedia, A.; Dubey, N.K. Assessment of some essential oils as food preservatives based on antifungal, antiaflatoxin, antioxidant activities and in vivo efficacy in food system. Food Res. Int. 2012, 49, 201–208. [Google Scholar] [CrossRef]
  151. Maleki, H.; Azadi, H.; Yousefpoor, Y.; Doostan, M.; Doostan, M.; Farzaei, M.H. Encapsulation of Ginger Extract in Nanoemulsions: Preparation, Characterization and in vivo Evaluation in Rheumatoid Arthritis. J. Pharm. Sci. 2023, 112, 1687–1697. [Google Scholar] [CrossRef] [PubMed]
  152. Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; Rodriguez-Torres, M.D.P.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano based drug delivery systems: Recent developments and future prospects. J. Nanobiotechnol. 2018, 16, 71. [Google Scholar] [CrossRef]
  153. Alam, A.; Ansari, M.J.; Alqarni, M.H.; Salkini, M.A.; Raish, M. Antioxidant, Antibacterial, and Anticancer Activity of Ultrasonic Nanoemulsion of Cinnamomum cassia L. Essential Oil. Plants 2023, 12, 834. [Google Scholar] [CrossRef]
  154. Panyajai, P.; Chueahongthong, F.; Viriyaadhammaa, N.; Nirachonkul, W.; Tima, S.; Chiampanichayakul, S.; Anuchapreeda, S.; Okonogi, S. Anticancer activity of Zingiber ottensii essential oil and its nanoformulations. PLoS ONE 2022, 17, e0262335. [Google Scholar] [CrossRef]
  155. Navaei Shoorvarzi, S.; Shahraki, F.; Shafaei, N.; Karimi, E.; Oskoueian, E. Citrus aurantium L. bloom essential oil nanoemulsion: Synthesis, characterization, cytotoxicity, and its potential health impacts on mice. J. Food Biochem. 2020, 44, e13181. [Google Scholar] [CrossRef]
  156. Kim, J.; Jung, K.H.; Yan, H.H.; Cheon, M.J.; Kang, S.; Jin, X.; Park, S.; Oh, M.S.; Hong, S.S. Artemisia Capillaris leaves inhibit cell proliferation and induce apoptosis in hepatocellular carcinoma. BMC Complement. Altern. Med. 2018, 18, 147. [Google Scholar] [CrossRef]
  157. Hong, J.H.; Lee, J.W.; Park, J.H.; Lee, I.S. Antioxidative and cytoprotective effects of Artemisia capillaris fractions. Biofactors 2007, 31, 43–53. [Google Scholar] [CrossRef] [PubMed]
  158. Baldea, I.; Mocan, T.; Cosgarea, R. The role of ultraviolet radiation and tyrosine stimulated melanogenesis in the induction of oxidative stress alterations in fair skin melanocytes. Exp. Oncol. 2009, 31, 200–208. [Google Scholar] [PubMed]
  159. Kim, E.S.; Park, S.J.; Goh, M.J.; Na, Y.J.; Jo, D.S.; Jo, Y.K.; Shin, J.H.; Choi, E.S.; Lee, H.K.; Kim, J.Y.; et al. Mitochondrial dynamics regulate melanogenesis through proteasomal degradation of MITF via ROS-ERK activation. Pigm. Cell Melanoma Res. 2014, 27, 1051–1062. [Google Scholar] [CrossRef] [PubMed]
  160. Choi, M.-H.; Min, M.-J.; Oh, D.-S.; Shin, H.-J. Antimicrobial and Antioxidant Activity of Camellia japonica Extracts for Cosmetic Applications. KSBB J. 2013, 28, 99–105. [Google Scholar] [CrossRef]
  161. Lee, H.H.; Cho, J.Y.; Moon, J.H.; Park, K.H. Isolation and identification of antioxidative phenolic acids and flavonoid glycosides from Camellia japonica flowers. Hortic. Environ. Biotechnol. 2011, 52, 270–277. [Google Scholar] [CrossRef]
  162. Kim, K.Y.; Davidson, P.M.; Chung, H.J. Antibacterial activity in extracts of Camellia japonica L. petals and its application to a model food system. J. Food Prot. 2001, 64, 1255–1260. [Google Scholar] [CrossRef] [PubMed]
  163. Kong, Y.; Wang, G.; Wang, X.; Wang, T.; Shen, J.; Zhang, A.; Zheng, L.; Zhang, Y. Essential Oils from The Dropped Flowers of Camellia japonica: Extraction Optimization, Chemical Profile and Antibacterial Property. Am. J. Biochem. Biotechnol. 2021, 17, 40–49. [Google Scholar] [CrossRef]
  164. Kim, S.; Jung, E.; Shin, S.; Kim, M.; Kim, Y.S.; Lee, J.; Park, D. Anti-inflammatory activity of Camellia japonica oil. BMB Rep. 2012, 45, 177–182. [Google Scholar] [CrossRef] [PubMed]
  165. Jung, E.; Lee, J.; Baek, J.; Jung, K.; Lee, J.; Huh, S.; Kim, S.; Koh, J.; Park, D. Effect of Camellia japonica oil on human type I procollagen production and skin barrier function. J. Ethnopharmacol. 2007, 112, 127–131. [Google Scholar] [CrossRef]
  166. Tahany, M.; Abdel-Rahman, N.M.K.; Abd El-Ghany, M.N.; Yosef, E. Purification, characterization and medicinal application of tyrosinase extracted from Saccharomyces cerevisiae. J. Innov. Pharm. Biol. Sci. 2019, 6, 1–11. [Google Scholar]
  167. Chou, S.T.; Chang, W.L.; Chang, C.T.; Hsu, S.L.; Lin, Y.C.; Shih, Y. Cinnamomum cassia essential oil inhibits alpha-MSH-induced melanin production and oxidative stress in murine B16 melanoma cells. Int. J. Mol. Sci. 2013, 14, 19186–19201. [Google Scholar] [CrossRef]
  168. Ha, S.Y.; Jung, J.Y.; Yang, J.K. Camellia japonica Essential Oil Inhibits alpha-MSH-Induced Melanin Production and Tyrosinase Activity in B16F10 Melanoma Cells. Evid. Based Complement. Altern. Med. 2021, 2021, 6328767. [Google Scholar] [CrossRef]
  169. Marc, E.B.; Nelly, A.; Annick, D.D.; Frederic, D. Plants used as remedies antirheumatic and antineuralgic in the traditional medicine of Lebanon. J. Ethnopharmacol. 2008, 120, 315–334. [Google Scholar] [CrossRef]
  170. El Khoury, R.; Michael Jubeli, R.; El Beyrouthy, M.; Baillet Guffroy, A.; Rizk, T.; Tfayli, A.; Lteif, R. Phytochemical screening and antityrosinase activity of carvacrol, thymoquinone, and four essential oils of Lebanese plants. J. Cosmet. Dermatol. 2019, 18, 944–952. [Google Scholar] [CrossRef]
  171. El Khoury, R.; Michael-Jubeli, R.; Bakar, J.; Dakroub, H.; Rizk, T.; Baillet-Guffroy, A.; Lteif, R.; Tfayli, A. Origanum essential oils reduce the level of melanin in B16-F1 melanocytes. Eur. J. Dermatol. 2019, 29, 596–602. [Google Scholar] [CrossRef] [PubMed]
  172. Hsiao, W.W.; Kumar, K.J.S.; Lee, H.J.; Tsao, N.W.; Wang, S.Y. Anti-Melanogenic Activity of Calocedrus formosana Wood Essential Oil and Its Chemical Composition Analysis. Plants 2021, 11, 62. [Google Scholar] [CrossRef] [PubMed]
  173. Yuan, S.Y.; Lin, C.C.; Hsu, S.L.; Cheng, Y.W.; Wu, J.H.; Cheng, C.L.; Yang, C.R. Leaf Extracts of Calocedrus formosana (Florin) Induce G2/M Cell Cycle Arrest and Apoptosis in Human Bladder Cancer Cells. Evid. Based Complement. Altern. Med. 2011, 2011, 380923. [Google Scholar] [CrossRef]
  174. Ho, C.L.; Tseng, Y.H.; Wang, E.I.; Liao, P.C.; Chou, J.C.; Lin, C.N.; Su, Y.C. Composition, antioxidant and antimicrobial activities of the seed essential oil of Calocedrus formosana from Taiwan. Nat. Prod. Commun. 2011, 6, 133–136. [Google Scholar] [CrossRef] [PubMed]
  175. Wang, S.Y.; Wu, J.H.; Cheng, S.S.; Lo, C.P.; Chang, H.N.; Shyur, L.F.; Chang, S.T. Antioxidant activity of extracts from Calocedrus formosana leaf, bark, and heartwood. J. Wood Sci. 2004, 50, 422–426. [Google Scholar] [CrossRef]
  176. Chao, K.P.; Hua, K.F.; Hsu, H.Y.; Su, Y.C.; Chang, S.T. Anti-inflammatory activity of sugiol, a diterpene isolated from Calocedrus formosana bark. Planta Med. 2005, 71, 300–305. [Google Scholar] [CrossRef] [PubMed]
  177. Cheng, S.S.; Wu, C.L.; Chang, H.T.; Kao, Y.T.; Chang, S.T. Antitermitic and antifungal activities of essential oil of Calocedrus formosana leaf and its composition. J. Chem. Ecol. 2004, 30, 1957–1967. [Google Scholar] [CrossRef] [PubMed]
  178. Hellyer, R.O.; Lassak, E.V. The steam-volatile constituents of Melaleuca viridiflora sol. ex gaertn. Aust. J. Chem. 1968, 10, 2585–2587. [Google Scholar] [CrossRef]
  179. Chao, W.W.; Su, C.C.; Peng, H.Y.; Chou, S.T. Melaleuca quinquenervia essential oil inhibits alpha-melanocyte-stimulating hormone-induced melanin production and oxidative stress in B16 melanoma cells. Phytomedicine 2017, 34, 191–201. [Google Scholar] [CrossRef]
  180. Mann, M.N.; Fisher, E.R. Investigation of Antibacterial 1,8-Cineole-Derived Thin Films Formed via Plasma-Enhanced Chemical Vapor Deposition. ACS Appl. Mater. Interfaces 2017, 9, 36548–36560. [Google Scholar] [CrossRef]
  181. Pillaiyar, T.; Manickam, M.; Jung, S.H. Downregulation of melanogenesis: Drug discovery and therapeutic options. Drug Discov. Today 2017, 22, 282–298. [Google Scholar] [CrossRef] [PubMed]
  182. Villarama, C.D.; Maibach, H.I. Glutathione as a depigmenting agent: An overview. Int. J. Cosmet. Sci. 2005, 27, 147–153. [Google Scholar] [CrossRef] [PubMed]
  183. Lee, H.S.; Ahn, Y.J. Growth-Inhibiting Effects of Cinnamomum cassia Bark-Derived Materials on Human Intestinal Bacteria. J. Agric. Food Chem. 1998, 46, 8–12. [Google Scholar] [CrossRef] [PubMed]
  184. Lee, H.S.; Kim, B.S.; Kim, M.K. Suppression effect of Cinnamomum cassia bark-derived component on nitric oxide synthase. J. Agric. Food Chem. 2002, 50, 7700–7703. [Google Scholar] [CrossRef] [PubMed]
  185. Verspohl, E.J.; Bauer, K.; Neddermann, E. Antidiabetic effect of Cinnamomum cassia and Cinnamomum zeylanicum in vivo and in vitro. Phytother. Res. 2005, 19, 203–206. [Google Scholar] [CrossRef] [PubMed]
  186. Zhang, C.; Fan, L.; Fan, S.; Wang, J.; Luo, T.; Tang, Y.; Chen, Z.; Yu, L. Cinnamomum cassia Presl: A Review of Its Traditional Uses, Phytochemistry, Pharmacology and Toxicology. Molecules 2019, 24, 3473. [Google Scholar] [CrossRef] [PubMed]
  187. Ngoc, T.M.; Lee, I.; Ha, D.T.; Kim, H.; Min, B.; Bae, K. Tyrosinase-inhibitory constituents from the twigs of Cinnamomum cassia. J. Nat. Prod. 2009, 72, 1205–1208. [Google Scholar] [CrossRef] [PubMed]
  188. Zhao, X.; Zhu, J.X.; Mo, S.F.; Pan, Y.; Kong, L.D. Effects of cassia oil on serum and hepatic uric acid levels in oxonate-induced mice and xanthine dehydrogenase and xanthine oxidase activities in mouse liver. J. Ethnopharmacol. 2006, 103, 357–365. [Google Scholar] [CrossRef] [PubMed]
  189. Giordani, R.; Regli, P.; Kaloustian, J.; Portugal, H. Potentiation of antifungal activity of amphotericin B by essential oil from Cinnamomum cassia. Phytother. Res. 2006, 20, 58–61. [Google Scholar] [CrossRef]
  190. Padalia, R.C.; Verma, R.S.; Sundaresan, V.; Chanotiya, C.S. Chemical diversity in the genus Alpinia (Zingiberaceae): Comparative composition of four Alpinia species grown in Northern India. Chem. Biodivers. 2010, 7, 2076–2087. [Google Scholar] [CrossRef]
  191. Kumar, K.J.S.; Wang, S.H.; Tseng, Y.H.; Tsao, N.W.; Kuo, Y.H.; Wang, S.Y. trans-3-Methoxy-5-hydroxystilbene (MHS) from the rhizome of Alpinia nantonensis inhibits metastasis in human lung cancer cells. Phytomedicine 2018, 50, 223–230. [Google Scholar] [CrossRef] [PubMed]
  192. Kuo, C.Y.; Weng, T.S.; Kumar, K.J.S.; Tseng, Y.H.; Tung, T.W.; Wang, S.Y.; Wang, H.C. Ethanol Extracts of Dietary Herb, Alpinia nantoensis, Exhibit Anticancer Potential in Human Breast Cancer Cells. Integr. Cancer Ther. 2019, 18, 1–12. [Google Scholar] [CrossRef] [PubMed]
  193. Kumar, K.J.S.; Vani, M.G.; Wu, P.C.; Lee, H.J.; Tseng, Y.H.; Wang, S.Y. Essential Oils of Alpinia nantoensis Retard Forskolin-Induced Melanogenesis via ERK1/2-Mediated Proteasomal Degradation of MITF. Plants 2020, 9, 1672. [Google Scholar] [CrossRef] [PubMed]
  194. Kim, K.J.; Kim, Y.H.; Yu, H.H.; Jeong, S.I.; Cha, J.D.; Kil, B.S.; You, Y.O. Antibacterial activity and chemical composition of essential oil of Chrysanthemum boreale. Planta Med. 2003, 69, 274–277. [Google Scholar] [CrossRef] [PubMed]
  195. Kim, D.Y.; Won, K.J.; Yoon, M.S.; Yu, H.J.; Park, J.H.; Kim, B.; Lee, H.M. Chrysanthemum boreale flower floral water inhibits platelet-derived growth factor-stimulated migration and proliferation in vascular smooth muscle cells. Pharm. Biol. 2015, 53, 725–734. [Google Scholar] [CrossRef] [PubMed]
  196. Kim, D.Y.; Won, K.J.; Yoon, M.S.; Hwang, D.I.; Yoon, S.W.; Park, J.H.; Kim, B.; Lee, H.M. Chrysanthemum boreale Makino essential oil induces keratinocyte proliferation and skin regeneration. Nat. Prod. Res. 2015, 29, 562–564. [Google Scholar] [CrossRef] [PubMed]
  197. Kim, Y.; Sung, J.; Sung, M.; Choi, Y.; Jeong, H.S.; Lee, J. Involvement of heme oxygenase-1 in the anti-inflammatory activity of Chrysanthemum boreale Makino extracts on the expression of inducible nitric oxide synthase in RAW264.7 macrophages. J. Ethnopharmacol. 2010, 131, 550–554. [Google Scholar] [CrossRef] [PubMed]
  198. Kim, D.Y.; Won, K.J.; Hwang, D.I.; Park, S.M.; Kim, B.; Lee, H.M. Chemical Composition, Antioxidant and Anti melanogenic Activities of Essential Oils from Chrysanthemum boreale Makino at Different Harvesting Stages. Chem. Biodivers. 2018, 15, e1700506. [Google Scholar] [CrossRef]
  199. Huang, H.C.; Chang, T.Y.; Chang, L.Z.; Wang, H.F.; Yih, K.H.; Hsieh, W.Y.; Chang, T.M. Inhibition of melanogenesis versus antioxidant properties of essential oil extracted from leaves of Vitex negundo Linn and chemical composition analysis by GC-MS. Molecules 2012, 17, 3902–3916. [Google Scholar] [CrossRef]
  200. Gupta, R.K.; Tandon, V.R. Antinociceptive activity of Vitex-negundo Linn leaf extract. Indian J. Physiol. Pharmacol. 2005, 49, 163–170. [Google Scholar]
  201. Zheng, C.J.; Huang, B.K.; Han, T.; Zhang, Q.Y.; Zhang, H.; Rahman, K.; Qin, L.P. Antinociceptive activities of the liposoluble fraction from Vitex negundo seeds. Pharm. Biol. 2010, 48, 651–658. [Google Scholar] [CrossRef] [PubMed]
  202. Tandon, V.R.; Gupta, R.K. An experimental evaluation of anticonvulsant activity of Vitex-negundo. Indian J. Physiol. Pharmacol. 2005, 49, 199–205. [Google Scholar] [PubMed]
  203. Dharmasiri, M.G.; Jayakody, J.R.; Galhena, G.; Liyanage, S.S.; Ratnasooriya, W.D. Anti-inflammatory and analgesic activities of mature fresh leaves of Vitex negundo. J. Ethnopharmacol. 2003, 87, 199–206. [Google Scholar] [CrossRef] [PubMed]
  204. Haq, A.U.; Malik, A.; Khan, M.T.H.; Anwar-ul-Haq; Khan, S.B.; Ahmad, A.; Choudhary, M.I. Tyrosinase inhibitory lignans from the methanol extract of the roots of Vitex negundo Linn. and their structure-activity relationship. Phytomedicine 2006, 13, 255–260. [Google Scholar] [CrossRef] [PubMed]
  205. Feizpour, A.; Boskabady, M.H.; Byrami, G.; Golamnezhad, Z.; Shafei, M.N. The effect of hydro-ethanolic extract of Achillea millefolium on muscarinic receptors of guinea pig tracheal smooth muscle. Indian J. Pharmacol. 2013, 45, 13–17. [Google Scholar] [CrossRef] [PubMed]
  206. Mahady, G.B.; Pendland, S.L.; Stoia, A.; Hamill, F.A.; Fabricant, D.; Dietz, B.M.; Chadwick, L.R. In vitro susceptibility of Helicobacter pylori to botanical extracts used traditionally for the treatment of gastrointestinal disorders. Phytother. Res. 2005, 19, 988–991. [Google Scholar] [CrossRef]
  207. Benedek, B.; Geisz, N.; Jager, W.; Thalhammer, T.; Kopp, B. Choleretic effects of yarrow (Achillea millefolium s.l.) in the isolated perfused rat liver. Phytomedicine 2006, 13, 702–706. [Google Scholar] [CrossRef]
  208. Khan, A.U.; Gilani, A.H. Blood pressure lowering, cardiovascular inhibitory and bronchodilatory actions of Achillea millefolium. Phytother. Res. 2011, 25, 577–583. [Google Scholar] [CrossRef]
  209. Farasati Far, B.; Behzad, G.; Khalili, H. Achillea millefolium: Mechanism of action, pharmacokinetic, clinical drug-drug interactions and tolerability. Heliyon 2023, 9, e22841. [Google Scholar] [CrossRef]
  210. Chou, S.T.; Peng, H.Y.; Hsu, J.C.; Lin, C.C.; Shih, Y. Achillea millefolium L. essential oil inhibits LPS-induced oxidative stress and nitric oxide production in RAW 264.7 Macrophages. Int. J. Mol. Sci. 2013, 14, 12978–12993. [Google Scholar] [CrossRef]
  211. Peng, H.Y.; Lin, C.C.; Wang, H.Y.; Shih, Y.; Chou, S.T. The melanogenesis alteration effects of Achillea millefolium L. essential oil and linalyl acetate: Involvement of oxidative stress and the JNK and ERK signaling pathways in melanoma cells. PLoS ONE 2014, 9, e95186. [Google Scholar] [CrossRef]
  212. Aibibu, N.; Liu, Y.; Zeng, G.; Wang, X.; Chen, B.; Song, H.; Xu, L. Cadmium accumulation in vetiveria zizanioides and its effects on growth, physiological and biochemical characters. Bioresour. Technol. 2010, 101, 6297–6303. [Google Scholar] [CrossRef] [PubMed]
  213. Amarasiri, S.S.; Attanayake, A.P.; Arawwawala, L.; Jayatilaka, K.; Mudduwa, L.K.B. Nephroprotective activity of Vetiveria zizanioides (L.) Nash supplement in doxorubicin-induced nephrotoxicity model of Wistar rats. J. Food Biochem. 2021, 45, e13901. [Google Scholar] [CrossRef] [PubMed]
  214. Luqman, S.; Kumar, R.; Kaushik, S.; Srivastava, S.; Darokar, M.P.; Khanuja, S.P. Antioxidant potential of the root of Vetiveria zizanioides (L.) Nash. Indian J. Biochem. Biophys. 2009, 46, 122–125. [Google Scholar] [PubMed]
  215. David, A.; Wang, F.; Sun, X.; Li, H.; Lin, J.; Li, P.; Deng, G. Chemical Composition, Antioxidant, and Antimicrobial Activities of Vetiveria zizanioides (L.) Nash Essential Oil Extracted by Carbon Dioxide Expanded Ethanol. Molecules 2019, 24, 1897. [Google Scholar] [CrossRef]
  216. Chou, S.T.; Lai, C.P.; Lin, C.C.; Shih, Y. Study of the chemical composition, antioxidant activity and anti-inflammatory activity of essential oil from Vetiveria zizanioides. Food Chem. 2012, 134, 262–268. [Google Scholar] [CrossRef]
  217. Peng, H.Y.; Lai, C.C.; Lin, C.C.; Chou, S.T. Effect of Vetiveria zizanioides essential oil on melanogenesis in melanoma cells: Downregulation of tyrosinase expression and suppression of oxidative stress. Sci. World J. 2014, 2014, 213013. [Google Scholar] [CrossRef] [PubMed]
  218. Borzoui, E.; Khaghani, R.; Nouri-Ganbalani, G. Lethal and Sublethal Effects of Eucalyptus camaldulensis and Mentha piperita Essential Oils on the Khapra Beetle (Coleoptera: Dermestidae) in Terms of Feeding Inhibition, Oviposition, and Seed Damage. Environ. Entomol. 2021, 50, 692–698. [Google Scholar] [CrossRef]
  219. Butcher, P.A.; Otero, A.; McDonald, M.W.; Moran, G.F. Nuclear RFLP variation in Eucalyptus camaldulensis Dehnh. from northern Australia. Heredity 2002, 88, 402–412. [Google Scholar] [CrossRef]
  220. Cimanga, K.; Kambu, K.; Tona, L.; Apers, S.; De Bruyne, T.; Hermans, N.; Totté, J.; Pieters, L.; Vlietinck, A.J. Correlation between chemical composition and antibacterial activity of essential oils of some aromatic medicinal plants growing in the Democratic Republic of Congo. J. Ethnopharmacol. 2002, 79, 213–220. [Google Scholar] [CrossRef]
  221. Siramon, P.; Ohtani, Y.; Ichiura, H. Biological performance of Eucalyptus camaldulensis leaf oils from Thailand against the subterranean termite Coptotermes formosanus Shiraki. J. Wood Sci. 2009, 55, 41–46. [Google Scholar] [CrossRef]
  222. Siramon, P.; Ohtani, Y. Antioxidative and antiradical activities of Eucalyptus camaldulensis leaf oils from Thailand. J. Wood Sci. 2007, 53, 498–504. [Google Scholar] [CrossRef]
  223. Huang, H.C.; Ho, Y.C.; Lim, J.M.; Chang, T.Y.; Ho, C.L.; Chang, T.M. Investigation of the Anti-Melanogenic and Antioxidant Characteristics of Eucalyptus camaldulensis Flower Essential Oil and Determination of Its Chemical Composition. Int. J. Mol. Sci. 2015, 16, 10470–10490. [Google Scholar] [CrossRef]
  224. Zhao, Y.; Li, J.; Cao, G.; Zhao, D.; Li, G.; Zhang, H.; Yan, M. Ethnic, Botanic, Phytochemistry and Pharmacology of the Acorus L. Genus: A Review. Molecules 2023, 28, 7117. [Google Scholar] [CrossRef]
  225. Kumar, R.; Prakash, O.; Pan, A.K.; Hore, S.K.; Chanotiya, C.S.; Mathela, C.S. Compositional variations and anthelmentic activity of essential oils from rhizomes of different wild populations of Acorus calamus L. and its major component, beta-Asarone. Nat. Prod. Commun. 2009, 4, 275–278. [Google Scholar] [CrossRef]
  226. Mukherjee, P.K.; Kumar, V.; Mal, M.; Houghton, P.J. In vitro acetylcholinesterase inhibitory activity of the essential oil from Acorus calamus and its main constituents. Planta Med. 2007, 73, 283–285. [Google Scholar] [CrossRef]
  227. Cho, J.; Kong, J.Y.; Jeong, D.Y.; Lee, K.D.; Lee, D.U.; Kang, B.S. NMDA recepter-mediated neuroprotection by essential oils from the rhizomes of Acorus gramineus. Life Sci. 2001, 68, 1567–1573. [Google Scholar] [CrossRef]
  228. Huang, H.C.; Wang, H.F.; Yih, K.H.; Chang, L.Z.; Chang, T.M. The Dual Antimelanogenic and Antioxidant Activities of the Essential Oil Extracted from the Leaves of Acorus macrospadiceus (Yamamoto) F. N. Wei et Y. K. Li. Evid. Based Complement. Altern. Med. 2012, 2012, 781280. [Google Scholar] [CrossRef]
  229. Chereddy, K.K.; Coco, R.; Memvanga, P.B.; Ucakar, B.; des Rieux, A.; Vandermeulen, G.; Preat, V. Combined effect of PLGA and curcumin on wound healing activity. J. Control Release 2013, 171, 208–215. [Google Scholar] [CrossRef]
  230. Dwivedi, D.; Dwivedi, M.; Malviya, S.; Singh, V. Evaluation of wound healing, anti-microbial and antioxidant potential of Pongamia pinnata in wistar rats. J. Tradit. Complement. Med. 2017, 7, 79–85. [Google Scholar] [CrossRef]
  231. Nayak, B.S.; Sandiford, S.; Maxwell, A. Evaluation of the Wound-healing Activity of Ethanolic Extract of Morinda citrifolia L. Leaf. Evid. Based Complement. Altern. Med. 2009, 6, 351–356. [Google Scholar] [CrossRef]
  232. Wedler, J.; Rusanov, K.; Atanassov, I.; Butterweck, V. A Polyphenol-Enriched Fraction of Rose Oil Distillation Wastewater Inhibits Cell Proliferation, Migration and TNF-alpha-Induced VEGF Secretion in Human Immortalized Keratinocytes. Planta Med. 2016, 82, 1000–1008. [Google Scholar] [CrossRef]
  233. Mercurio, L.; Albanesi, C.; Madonna, S. Recent Updates on the Involvement of PI3K/AKT/mTOR Molecular Cascade in the Pathogenesis of Hyperproliferative Skin Disorders. Front. Med. 2021, 8, 665647. [Google Scholar] [CrossRef]
  234. Quadri, M.; Pellegrini, C.; Efimova, T.; Palazzo, E. Editorial: New tools and molecular advances in hyperproliferative skin disorders. Front. Med. 2022, 9, 1002872. [Google Scholar] [CrossRef]
  235. Choi, E.; Park, H.; Lee, J.; Kim, G. Anticancer, antiobesity, and anti-inflammatory activity of Artemisia species in vitro. J. Tradit. Chin. Med. 2013, 33, 92–97. [Google Scholar] [CrossRef]
  236. Elfawal, M.A.; Towler, M.J.; Reich, N.G.; Golenbock, D.; Weathers, P.J.; Rich, S.M. Dried Whole Plant Artemisia annua as an Antimalarial Therapy. PLoS ONE 2012, 7, e52746. [Google Scholar] [CrossRef]
  237. Xiao, J.; Liu, P.; Hu, Y.; Liu, T.; Guo, Y.; Sun, P.; Zheng, J.; Ren, Z.; Wang, Y. Antiviral activities of Artemisia vulgaris L. extract against herpes simplex virus. Chin. Med. 2023, 18, 21. [Google Scholar] [CrossRef]
  238. Ali, M.Y.; Park, S.E.; Seong, S.H.; Zamponi, G.W.; Jung, H.A.; Choi, J.S. Ursonic acid from Artemisia montana exerts anti-diabetic effects through anti-glycating properties, and by inhibiting PTP1B and activating the PI3K/Akt signaling pathway in insulin-resistant C2C12 cells. Chem.-Biol. Interact. 2023, 376, 110452. [Google Scholar] [CrossRef]
  239. Jeong, S.H.; Kim, J.; Min, H. In vitro anti-inflammatory activity of the Artemisia montana leaf ethanol extract in macrophage RAW 264.7 cells. Food Agric. Immunol. 2018, 29, 688–698. [Google Scholar] [CrossRef]
  240. Lee, G.-D.; Kim, J.-S.; Bae, J.-O.; Yoon, H.-S. Antioxidative Effectiveness of Water Extract and Ether Extract in Wormwood(Artemisia montana Pampan). J. Korean Soc. Food Sci. Nutr. 1992, 21, 17–22. [Google Scholar]
  241. Yoon, M.S.; Won, K.J.; Kim, D.Y.; Hwang, D.I.; Yoon, S.W.; Kim, B.; Lee, H.M. Skin regeneration effect and chemical composition of essential oil from Artemisia Montana. Nat. Prod. Commun. 2014, 9, 1619–1622. [Google Scholar] [CrossRef]
  242. Tang, L.; Sierra, J.O.; Kelly, R.; Kirsner, R.S.; Li, J. Wool-derived keratin stimulates human keratinocyte migration and types IV and VII collagen expression. Exp. Dermatol. 2012, 21, 458–460. [Google Scholar] [CrossRef]
  243. Pachi, V.K.; Mikropoulou, E.V.; Gkiouvetidis, P.; Siafakas, K.; Argyropoulou, A.; Angelis, A.; Mitakou, S.; Halabalaki, M. Traditional uses, phytochemistry and pharmacology of Chios mastic gum (Pistacia lentiscus var. Chia, Anacardiaceae): A review. J. Ethnopharmacol. 2020, 254, 112485. [Google Scholar] [CrossRef]
  244. Milia, E.; Bullitta, S.M.; Mastandrea, G.; Szotakova, B.; Schoubben, A.; Langhansova, L.; Quartu, M.; Bortone, A.; Eick, S. Leaves and Fruits Preparations of Pistacia lentiscus L.: A Review on the Ethnopharmacological Uses and Implications in Inflammation and Infection. Antibiotics 2021, 10, 425. [Google Scholar] [CrossRef]
  245. Mezni, F.; Khouja, M.L.; Gregoire, S.; Martine, L.; Khaldi, A.; Berdeaux, O. Effect of growing area on tocopherols, carotenoids and fatty acid composition of Pistacia lentiscus edible oil. Nat. Prod. Res. 2014, 28, 1225–1230. [Google Scholar] [CrossRef]
  246. Longo, L.; Scardino, A.; Vasapollo, G. Identification and quantification of anthocyanins in the berries of Pistacia lentiscus L., Phillyrea latifolia L. and Rubia peregrina L. Innov. Food Sci. Emerg. 2007, 8, 360–364. [Google Scholar] [CrossRef]
  247. Mharti, F.Z.; Lyoussi, B.; Abdellaoui, A. Antibacterial activity of the essential oils of Pistacia lentiscus used in Moroccan folkloric medicine. Nat. Prod. Commun. 2011, 6, 1505–1506. [Google Scholar] [CrossRef]
  248. Rahman, H.S. Phytochemical analysis and antioxidant and anticancer activities of mastic gum resin from Pistacia atlantica subspecies kurdica. Onco Targets Ther. 2018, 11, 4559–4572. [Google Scholar] [CrossRef]
  249. Siano, F.; Cutignano, A.; Moccia, S.; Russo, G.L.; Volpe, M.G.; Picariello, G. Phytochemical Characterization and Effects on Cell Proliferation of Lentisk (Pistacia lentiscus) Berry Oil: A Revalued Source of Phenolics. Plant Food Hum. Nutr. 2020, 75, 487–494. [Google Scholar] [CrossRef]
  250. Mezni, F.; Miled, K.; Khaldi, A.; Khouja, M.L.; Boubaker, S.; Maaroufi, A. Wound healing effect of Pistacia lentiscus L. seed oil: Confirmation of its uses in Mediterranean traditional medicine. Boletín Latinoam. Y Del Caribe De Plantas Med. Y Aromáticas 2020, 19, 314–320. [Google Scholar] [CrossRef]
  251. Allaw, M.; Manconi, M.; Caboni, P.; Bacchetta, G.; Escribano-Ferrer, E.; Peris, J.E.; Nacher, A.; Diez-Sales, O.; Manca, M.L. Formulation of liposomes loading lentisk oil to ameliorate topical delivery, attenuate oxidative stress damage and improve cell migration in scratch assay. Biomed. Pharmacother. 2021, 144, 112351. [Google Scholar] [CrossRef]
  252. Fadel, B.A.; Elwakil, B.H.; Fawzy, E.E.; Shaaban, M.M.; Olama, Z.A. Nanoemulsion of Lavandula angustifolia Essential Oil/Gold Nanoparticles: Antibacterial Effect against Multidrug-Resistant Wound-Causing Bacteria. Molecules 2023, 28, 6988. [Google Scholar] [CrossRef]
  253. Woollard, A.C.; Tatham, K.C.; Barker, S. The influence of essential oils on the process of wound healing: A review of the current evidence. J. Wound Care 2007, 16, 255–257. [Google Scholar] [CrossRef]
  254. Vakilian, K.; Atarha, M.; Bekhradi, R.; Chaman, R. Healing advantages of lavender essential oil during episiotomy recovery: A clinical trial. Complement. Ther. Clin. Pract. 2011, 17, 50–53. [Google Scholar] [CrossRef]
  255. Altaei, D.T. Topical lavender oil for the treatment of recurrent aphthous ulceration. Am. J. Dent. 2012, 25, 39–43. [Google Scholar]
  256. Koca Kutlu, A.; Cecen, D.; Gurgen, S.G.; Sayin, O.; Cetin, F. A Comparison Study of Growth Factor Expression following Treatment with Transcutaneous Electrical Nerve Stimulation, Saline Solution, Povidone-Iodine, and Lavender Oil in Wounds Healing. Evid. Based Complement. Altern. Med. 2013, 2013, 361832. [Google Scholar] [CrossRef]
  257. Clark, R.A.; Nielsen, L.D.; Welch, M.P.; McPherson, J.M. Collagen matrices attenuate the collagen-synthetic response of cultured fibroblasts to TGF-beta. J. Cell Sci. 1995, 108 Pt 3, 1251–1261. [Google Scholar] [CrossRef]
  258. Desmouliere, A.; Geinoz, A.; Gabbiani, F.; Gabbiani, G. Transforming growth factor-beta 1 induces alpha-smooth muscle actin expression in granulation tissue myofibroblasts and in quiescent and growing cultured fibroblasts. J. Cell Biol. 1993, 122, 103–111. [Google Scholar] [CrossRef]
  259. Crane, S.; Aurore, G.; Joseph, H.; Mouloungui, Z.; Bourgeois, P. Composition of fatty acids triacylglycerols and unsaponifiable matter in Calophyllum calaba L. oil from Guadeloupe. Phytochemistry 2005, 66, 1825–1831. [Google Scholar] [CrossRef]
  260. Ansel, J.L.; Lupo, E.; Mijouin, L.; Guillot, S.; Butaud, J.F.; Ho, R.; Lecellier, G.; Raharivelomanana, P.; Pichon, C. Biological Activity of Polynesian Calophyllum inophyllum Oil Extract on Human Skin Cells. Planta Med. 2016, 82, 961–966. [Google Scholar] [CrossRef]
  261. Bhalla, T.N.; Saxena, R.C.; Nigam, S.K.; Misra, G.; Bhargava, K.P. Calophyllolide--a new non-steroidal anti-inflammatory agent. Indian J. Med. Res. 1980, 72, 762–765. [Google Scholar]
  262. Yimdjo, M.C.; Azebaze, A.G.; Nkengfack, A.E.; Meyer, A.M.; Bodo, B.; Fomum, Z.T. Antimicrobial and cytotoxic agents from Calophyllum inophyllum. Phytochemistry 2004, 65, 2789–2795. [Google Scholar] [CrossRef] [PubMed]
  263. Saravanan, R.; Dhachinamoorthi, D.; Senthilkumar, K.; Thamizhvanan, K. Antimicrobial activity of various extracts from various parts of Calophyllum inophyllum L. J. Appl. Pharm. Sci. 2011, 1, 1–12. [Google Scholar]
  264. Dweck, A.C.; Meadows, T. Tamanu (Calophyllum inophyllum)—The African, Asian, Polynesian and Pacific Panacea. Int. J. Cosmet. Sci. 2002, 24, 341–348. [Google Scholar] [CrossRef] [PubMed]
  265. Erdogan, S.S.; Gur, T.F.; Terzi, N.K.; Dogan, B. Evaluation of the cutaneous wound healing potential of tamanu oil in wounds induced in rats. J. Wound Care 2021, 30, Vi–Vx. [Google Scholar] [CrossRef]
  266. Ali, S.; Khan, M.R.; Irfanullah; Sajid, M.; Zahra, Z. Phytochemical investigation and antimicrobial appraisal of Parrotiopsis jacquemontiana (Decne) Rehder. BMC Complement. Altern. Med. 2018, 18, 43. [Google Scholar] [CrossRef] [PubMed]
  267. Ali, S.; Khan, M.R.; Iqbal, J.; Batool, R.; Naz, I.; Yaseen, T.; Abbasi, B.A.; Nasir, J.A.; El-Serehy, H.A. Chemical composition and pharmacological bio-efficacy of Parrotiopsis jacquemontiana (Decne) Rehder for anticancer activity. Saudi J. Biol. Sci. 2021, 28, 4969–4986. [Google Scholar] [CrossRef] [PubMed]
  268. Ali, S.; Khan, M.R.; Batool, R.; Maryam, S.; Majid, M. Wound healing potential of oil extracted from Parrotiopsis jacquemontiana (Decne) Rehder. J. Ethnopharmacol. 2019, 236, 354–365. [Google Scholar] [CrossRef] [PubMed]
  269. Walker, J.B.; Sytsma, K.J. Staminal evolution in the genus Salvia (Lamiaceae): Molecular phylogenetic evidence for multiple origins of the staminal lever. Ann. Bot. 2007, 100, 375–391. [Google Scholar] [CrossRef]
  270. Badiee, P.; Nasirzadeh, A.R.; Motaffaf, M. Comparison of Salvia officinalis L. essential oil and antifungal agents against candida species. J. Pharm. Technol. Drug Res. 2012, 1, 7. [Google Scholar] [CrossRef]
  271. Miguel, G.; Cruz, C.; Faleiro, M.L.; Simoes, M.T.; Figueiredo, A.C.; Barroso, J.G.; Pedro, L.G. Salvia officinalis L. essential oils: Effect of hydrodistillation time on the chemical composition, antioxidant and antimicrobial activities. Nat. Prod. Res. 2011, 25, 526–541. [Google Scholar] [CrossRef] [PubMed]
  272. Dal Prá, V.; Bisol, L.B.; Detoni, S.; Denti, M.; Grando, J.; Pollo, C.; Pasquali, T.R.; Hofmann Júnio, A.E.; Mazzuti, M.A.; Macedo, S.M. Antiinflammatory activity of fracionated extracts of Salvia officinalis L. J. Appl. Pharm. Sci. 2011, 1, 67–71. [Google Scholar]
  273. Hamidpour, M.; Hamidpour, R.; Hamidpour, S.; Shahlari, M. Chemistry, Pharmacology, and Medicinal Property of Sage (Salvia) to Prevent and Cure Illnesses such as Obesity, Diabetes, Depression, Dementia, Lupus, Autism, Heart Disease, and Cancer. J. Tradit. Complement. Med. 2014, 4, 82–88. [Google Scholar] [CrossRef] [PubMed]
  274. Medjahed, F.; Merouane, A.; Saadi, A.; Bader, A.; Cioni, P.L.; Flamini, G. Chemical profile and antifungal potential of essential oils from leaves and flowers of Salvia algeriensis (Desf.): A comparative study. Chil. J. Agric. Res. 2016, 76, 195–200. [Google Scholar] [CrossRef]
  275. Cagno, V.; Sgorbini, B.; Sanna, C.; Cagliero, C.; Ballero, M.; Civra, A.; Donalisio, M.; Bicchi, C.; Lembo, D.; Rubiolo, P. In vitro anti-herpes simplex virus-2 activity of Salvia desoleana Atzei & V. Picci essential oil. PLoS ONE 2017, 12, e0172322. [Google Scholar] [CrossRef] [PubMed]
  276. Alves-Silva, J.M.; Maccioni, D.; Cocco, E.; Goncalves, M.J.; Porcedda, S.; Piras, A.; Cruz, M.T.; Salgueiro, L.; Maxia, A. Advances in the Phytochemical Characterisation and Bioactivities of Salvia aurea L. Essential Oil. Plants 2023, 12, 1247. [Google Scholar] [CrossRef]
  277. Boskabady, M.H.; Shafei, M.N.; Saberi, Z.; Amini, S. Pharmacological effects of rosa damascena. Iran. J. Basic Med. Sci. 2011, 14, 295–307. [Google Scholar] [PubMed]
  278. Fresco, P.; Borges, F.; Marques, M.P.; Diniz, C. The anticancer properties of dietary polyphenols and its relation with apoptosis. Curr. Pharm. Des. 2010, 16, 114–134. [Google Scholar] [CrossRef]
  279. Hisaka, T.; Sakai, H.; Sato, T.; Goto, Y.; Nomura, Y.; Fukutomi, S.; Fujita, F.; Mizobe, T.; Nakashima, O.; Tanigawa, M.; et al. Quercetin Suppresses Proliferation of Liver Cancer Cell Lines In Vitro. Anticancer. Res. 2020, 40, 4695–4700. [Google Scholar] [CrossRef]
  280. Zhang, T.; Chen, H.S.; Wang, L.F.; Bai, M.H.; Wang, Y.C.; Jiang, X.F.; Liu, M. Ellagic acid exerts anti-proliferation effects via modulation of Tgf-beta/Smad3 signaling in MCF-7 breast cancer cells. Asian Pac. J. Cancer Prev. 2014, 15, 273–276. [Google Scholar] [CrossRef]
  281. Singh, P.; Arif, Y.; Bajguz, A.; Hayat, S. The role of quercetin in plants. Plant Physiol. Biochem. 2021, 166, 10–19. [Google Scholar] [CrossRef] [PubMed]
  282. Villarreal-Lozoya, J.E.; Lombardini, L.; Cisneros-Zevallos, L. Phytochemical constituents and antioxidant capacity of different pecan [Carya illinoinensis (Wangenh.) K. Koch] cultivars. Food Chem 2007, 102, 1241–1249. [Google Scholar] [CrossRef]
  283. Daniel, E.M.; Krupnick, A.S.; Heur, Y.H.; Blinzler, J.A.; Nims, R.W.; Stoner, G.D. Extraction, stability, and quantitation of ellagic acid in various fruits and nuts. J. Food Compos. Anal. 1989, 2, 338–349. [Google Scholar] [CrossRef]
  284. Koponen, J.M.; Happonen, A.M.; Mattila, P.H.; Torronen, A.R. Contents of anthocyanins and ellagitannins in selected foods consumed in Finland. J. Agric. Food Chem. 2007, 55, 1612–1629. [Google Scholar] [CrossRef] [PubMed]
  285. Mertens-Talcott, S.U.; Talcott, S.T.; Percival, S.S. Low concentrations of quercetin and ellagic acid synergistically influence proliferation, cytotoxicity and apoptosis in MOLT-4 human leukemia cells. J. Nutr. 2003, 133, 2669–2674. [Google Scholar] [CrossRef]
  286. Li, K.; Zhou, R.; Wang Jia, W.; Li, Z.; Li, J.; Zhang, P.; Xiao, T. Zanthoxylum bungeanum essential oil induces apoptosis of HaCaT human keratinocytes. J. Ethnopharmacol. 2016, 186, 351–361. [Google Scholar] [CrossRef]
  287. Xia, L.A.; You, J.M.; Li, G.L.; Sun, Z.W.; Suo, Y.R. Compositional and Antioxidant Activity Analysis of Zanthoxylum bungeanum Seed Oil Obtained by Supercritical CO2 Fluid Extraction. J. Am. Oil Chem. Soc. 2011, 88, 23–32. [Google Scholar] [CrossRef]
  288. Li, Z.D.; Han, S.N.; Jiang, J.L.; Zhang, X.H.; Li, Y.; Chen, H.; Yuan, Y.J. Antitumor compound identification from Zanthoxylum bungeanum essential oil based on composition-activity relationship. Chem. Res. Chin. Univ. 2013, 29, 1068–1071. [Google Scholar] [CrossRef]
  289. Tezuka, Y.; Irikawa, S.; Kaneko, T.; Banskota, A.H.; Nagaoka, T.; Xiong, Q.; Hase, K.; Kadota, S. Screening of Chinese herbal drug extracts for inhibitory activity on nitric oxide production and identification of an active compound of Zanthoxylum bungeanum. J. Ethnopharmacol. 2001, 77, 209–217. [Google Scholar] [CrossRef]
  290. Zhu, R.X.; Zhong, K.; Zeng, W.C.; He, X.Y.; Gu, X.Q.; Zhao, Z.F.; Gao, H. Essential oil composition and antibacterial activity of Zanthoxylum bungeanum. Afr. J. Microbiol. Res. 2011, 5, 4631–4637. [Google Scholar] [CrossRef]
  291. Wang, C.F.; Yang, K.; Zhang, H.M.; Cao, J.; Fang, R.; Liu, Z.L.; Du, S.S.; Wang, Y.Y.; Deng, Z.W.; Zhou, L. Components and insecticidal activity against the maize weevils of Zanthoxylum schinifolium fruits and leaves. Molecules 2011, 16, 3077–3088. [Google Scholar] [CrossRef]
  292. Lim, T.K. Edible Medicinal and Non-Medicinal Plants; Springer: Dordrecht, The Netherlands; New York, NY, USA, 2012; pp. 442–450. [Google Scholar]
  293. Lee, H.H.; Ahn, J.H.; Kwon, A.R.; Lee, E.S.; Kwak, J.H.; Min, Y.H. Chemical composition and antimicrobial activity of the essential oil of apricot seed. Phytother. Res. 2014, 28, 1867–1872. [Google Scholar] [CrossRef]
  294. Gomaa, E.Z. In vitro antioxidant, antimicrobial, and antitumor activities of bitter almond and sweet apricot (Prunus armeniaca L.) kernels. Food Sci. Biotechnol. 2013, 22, 455–463. [Google Scholar] [CrossRef]
  295. Yigit, D.; Yigit, N.; Mavi, A. Antioxidant and antimicrobial activities of bitter and sweet apricot (Prunus armeniaca L.) kernels. Braz. J. Med. Biol. Res. 2009, 42, 346–352. [Google Scholar] [CrossRef]
  296. Korekar, G.; Stobdan, T.; Arora, R.; Yadav, A.; Singh, S.B. Antioxidant capacity and phenolics content of apricot (Prunus armeniaca L.) kernel as a function of genotype. Plant Foods Hum. Nutr. 2011, 66, 376–383. [Google Scholar] [CrossRef] [PubMed]
  297. Abtahi, H.; Ghazavi, A.; Karimi, M.; Mollaghase, S.; Mosayebi, G. Antimicrobial Activities of Water and Methanol Extracts of Bitter Apricot Seeds. J. Med. Sci. 2008, 8, 433–436. [Google Scholar] [CrossRef]
  298. Chang, H.K.; Yang, H.Y.; Lee, T.H.; Shin, M.C.; Lee, M.H.; Shin, M.S.; Kim, C.J.; Kim, O.J.; Hong, S.P.; Cho, S. Armeniacae semen extract suppresses lipopolysaccharide-induced expressions of cyclooxygenase [correction of cycloosygenase]-2 and inducible nitric oxide synthase in mouse BV2 microglial cells. Biol. Pharm. Bull. 2005, 28, 449–454. [Google Scholar] [CrossRef]
  299. Do, J.S.; Hwang, J.K.; Seo, H.J.; Woo, W.H.; Nam, S.Y. Antiasthmatic activity and selective inhibition of type 2 helper T cell response by aqueous extract of semen armeniacae amarum. Immunopharmacol. Immunotoxicol. 2006, 28, 213–225. [Google Scholar] [CrossRef]
  300. Chang, H.K.; Shin, M.S.; Yang, H.Y.; Lee, J.W.; Kim, Y.S.; Lee, M.H.; Kim, J.; Kim, K.H.; Kim, C.J. Amygdalin induces apoptosis through regulation of Bax and Bcl-2 expressions in human DU145 and LNCaP prostate cancer cells. Biol. Pharm. Bull. 2006, 29, 1597–1602. [Google Scholar] [CrossRef]
  301. Li, K.; Yang, W.; Li, Z.; Jia, W.; Li, J.; Zhang, P.; Xiao, T. Bitter apricot essential oil induces apoptosis of human HaCaT keratinocytes. Int. Immunopharmacol. 2016, 34, 189–198. [Google Scholar] [CrossRef]
Figure 1. Schematic of the alleviation effect of PF-EO treatment in an imiquimod-mediated mouse model of psoriasis. PF-EO treatment induces the downregulation of psoriasis symptoms, such as erythema, scaling, and epidermal thickening, and the expression of inflammatory factors and psoriasis-related factors [22]. The red arrow indicates downregulated expression in the epidermis of a PF-EO-treated mouse. PF-EO: Perilla frutescens L. Britt essential oil; IL: interleukin; iNOS: inducible nitric oxide synthase; COX-2: cyclooxygenase-2; IFN: interferon.
Figure 1. Schematic of the alleviation effect of PF-EO treatment in an imiquimod-mediated mouse model of psoriasis. PF-EO treatment induces the downregulation of psoriasis symptoms, such as erythema, scaling, and epidermal thickening, and the expression of inflammatory factors and psoriasis-related factors [22]. The red arrow indicates downregulated expression in the epidermis of a PF-EO-treated mouse. PF-EO: Perilla frutescens L. Britt essential oil; IL: interleukin; iNOS: inducible nitric oxide synthase; COX-2: cyclooxygenase-2; IFN: interferon.
Ijms 25 05397 g001
Figure 2. Schematic of the anti-melanoma effect of AC-EO treatment on B16F10 melanoma cells. Cell viability is regulated by AC-EO-mediated apoptosis or the downregulation of proliferation [133]. HB-EGF or EGFR overexpression affects MMPs-7 and -9 and ADAM-9 signaling, whereas AC-EO, Cetuximab, or siRNA-EGFR inhibits signaling [133]. AC-EO: Aloysia citrodora-essential oil; Bax: Bcl-2-associated X; Cas: caspase; p-ERK: phosphoextracellular signal-regulated kinase; MMP: matrix metalloproteinase; ADAM: a disintegrin and metalloprotease; HB-EGF: heparin-binding epidermal growth factor; EGFR: epidermal growth factor receptor.
Figure 2. Schematic of the anti-melanoma effect of AC-EO treatment on B16F10 melanoma cells. Cell viability is regulated by AC-EO-mediated apoptosis or the downregulation of proliferation [133]. HB-EGF or EGFR overexpression affects MMPs-7 and -9 and ADAM-9 signaling, whereas AC-EO, Cetuximab, or siRNA-EGFR inhibits signaling [133]. AC-EO: Aloysia citrodora-essential oil; Bax: Bcl-2-associated X; Cas: caspase; p-ERK: phosphoextracellular signal-regulated kinase; MMP: matrix metalloproteinase; ADAM: a disintegrin and metalloprotease; HB-EGF: heparin-binding epidermal growth factor; EGFR: epidermal growth factor receptor.
Ijms 25 05397 g002
Table 1. Different functions of essential oils in the skin and their natural sources.
Table 1. Different functions of essential oils in the skin and their natural sources.
Essential Oil (EO)/Plant Oil (PO)EffectsSourcesParts (Components)Refs.
Citrus limetta peels EOAnti-inflammatory and anti-oxidantC. limetta RissoFruit peels (flavonoids)[4,5,8,9]
Baccharis dracunculifolia EOAnti-inflammatory, immunomodulator, anti-bacterial, anti-diabetic, and anti-microbialB. dracunculifoliaLeaves[10,11,12,13,14,15,16,17,18]
Perilla frutescens L. Britt EOAnti-inflammatory, anti-bacterial, anti-oxidant, alleviation of cold, cough, nausea, vomiting, abdominal pain, constipation, asthma, and food poisoningP. frutescens L. BrittStems and leaves[19,20,21,22]
Grapefruit EOAnti-inflammatory, anti-microbial, anti-oxidants, anti-bacterial, anti-proliferation, anti-cancer, and muscle sympathetic nerve activity regulatorC. maxima Burm. MerrGrapefruit peel (terpenes and terpene oxides)[29,30,31,34,35,36,37,38,39,40,41,42,43,44,45,46,47,50]
Oregano EOAnti-inflammatory, prevention of neurodegenerative disorders, anti-oxidant, immunomodulatory, anti-cancer, anti-melanogenesis, and anti-microbialO. vulgareCRV, thymol[51,52,53,54,57,58]
Satureja sahendica EOAnti-inflammatory, anti-bacterialS. sahendicaCRV, thymol, p-cymene, ß-caryophyllene, linalool, and other terpenoids[59,60,64]
Matricaria chamomilla EOAnti-inflammatory, alleviation of gastrointestinal conditions, anti-spasmodic, and wound healingM. chamomillaAzulene[71,72,73,74]
Helianthus annuus POAnti-inflammatory, skin hydrationH. annuusOleic acid, linoleic acids[77,79]
Mentha arvensis EOAnti-inflammatory, anti-oxidant, M. arvensisMenthol, menthone, and piperitone[80,82]
Rosmarinus officinalis EOAnti-inflammatory, treat digestive problem, nervous system, and allergyR. officinalisCamphor, eucalyptol[84,85,86]
Curcuma longa EOAnti-inflammatory, skin penetrationC. longaRhizome (terpinolene and α-phellandrene)[87,88,89]
Artemisia argyi EOAnti-inflammatory, anti-mutagenic, anti-tumor, anti-oxidant, anti-asthmatic, anti-fungal, and anti-melanogenesisA. argyiLeaves (cineole, camphor, eucalyptol, and caryophyllene)[90,91,92,93,94,95,96,97,98,99]
Lavender EOAnti-inflammatory, skin protection and wound healing, anxiolytic, neuroprotective, anti-oxidant, analgesic, anti-microbial, and alleviation of joint painL. angustifoliaBlossoms (Linalool and linalyl acetate)[100,101,102,103,104,105,106,107,108,109,110,252,253,254,255,256]
Zanthoxylum coreanum EOAnti-inflammatory, anti-viralZ. coreanumβ-Ocimene, α-pinene[111,112,113,114]
Aloysia citrodora EO
(Lippia citriodora EO)
Anti-melanoma, anti-cancer, anti-oxidant, and anti-bacterialA. citrodora
(L. citriodora)
Geranial, neral, flavonoid, phenol, 1,8-cineole, limonene, and citral[124,125,126,127,128,129,130,131,132,133]
Origanum majorana EOAnti-melanoma, anti-bacterial, anti-oxidant, and anti-fungalO. majoranaTerpinen-4-o1, L-α-Terpineol[149,150]
Artemisia capillaris EOAnti-melanogenesis, anti-cancer, and anti-oxidantA. capillarisGrass clumps[118,156,157]
Camellia japonica seed EOAnti-melanogenesis, anti-oxidant, anti-bacterial, anti-inflammatory, and skin barrier functionC. japonicaSeed (Hexamethylcyclotrisiloxane and octamethylcyclotetrasiloxane)[160,161,162,163,164,165,168]
Origanum syriacum EO and Origanum ehrenbergii EOAnti-melanogenesis, rheumatism maceration, and neuralgic treatmentO. syriacum and O. ehrenbergiiTerpenoids, quinones, and CRV[57,169,170,171]
Calocedrus formosana EOAnti-melanogenesis, anti-oxidant, anti-fungal, anti-inflammatory, and anti-cancerC. formosanaα-Terpineol, Terpinen-4-o1, and thymol[172,173,174,175,176,177]
Melaleuca quinquenervia EOAnti-melanogenesis, anti-bacterialM. quinquenervia1,8-Cineole, α-Pinene, and α-Terpineol[178,179,180]
Cinnamomum cassia EOAnti-melanogenesis, anti-bacterial, anti-inflammatory, anti-diabetic, hypouricemic, and anti-fungalC. cassiaStem bark and twig (cis-2-methoxycinnamic acid and cinnamaldehyde)[167,183,184,185,187,188,189]
Leaf of Alpinia nantoensis EO and rhizome of Alpinia nantoensis EOAnti-melanogenesis, anti-cancer,A. nantoensisLeaf, rhizome, and stem (camphor, camphene, β-pinene, p-cymene, α-pinene, and D-limonene)[191,192,193]
Chrysanthemum boreale MAKINO EOAnti-melanogenesis, anti-inflammatory, anti-bacterial, skin regeneration, and wound healingC. borealeFlower[194,196,197]
Vitex negundo EOAnti-melanogenesis, anti-nociceptive, anti-convulsant, anti-inflammatory, and analgesicV. negundoleaves and roots (sesquiterpene and monoterpene)[199,200,201,202,203,204]
Achillea millefolium EOAnti-melanogenesis, alleviation of inflammatory, gastrointestinal disorders, hepatobiliary conditions, overactive cardiovascular, and respiratory infectionA. millefoliumArtemisia ketone, camphor, linalyl acetate, and 1,8-cineole[206,207,208,209,210,211]
Vetiveria zizanioides EOAnti-melanogenesis, anti-oxidants, antimicrobial, and anti-inflammatoryV. zizanioidesRoots (Cedr-8-en-13-ol and α-Amorphene)[212,213,214,215,216,217]
Eucalyptus camaldulensis EOAnti-melanogenesis, anti-bacterial, anti-termitic, and anti-oxidantE. camaldulensisLeaf (Eucalyptol and γ-Terpinene)[220,221,222,223]
Acorus macrospadiceus EOAnti-melanogenesisA. macrospadiceusChavicol methyl ether and nootkatone[228]
Artemisia montana Pampan EOWound healing, anti-diabetic, anti-inflammatory, and anti-oxidantA. montanaβ-caryophyllene, germacrene D, 1,8-cineole, and camphor[238,239,240,241]
Pistacia lentiscus POWound healing, anti-bacterial, anti-oxidants, and proliferationP. lentiscus L.Oleic, palmitic, linoleic, tocopherols, carotenoids, and anthocyanins[243,244,245,246,247,248,249,250,251]
Calophyllum inophyllum POAnti-inflammatory, anti-microbial, anti-fungal, and wound healingC. inophyllumCalophyllolide, calophyllic acid, inophyllum, and polyphenols[261,262,263,264,265]
Parrotiopsis jacquemontiana POWound healing, anti-microbial, and anti-cancerP. jacquemontiana2, 6-dimethyl-8-oxoocta-2, 6-dienoic acid, syringol, and catechol[266,267,268]
Salvia aurea L. EOWound healingS. aurea1,8-cineole, β-pinene, cis-thujone, and camphor[276]
Polyphenol-enriched fraction of Rose plant oil distillation wastewaterHyperproliferation inhibition, anti-HIV, anti-bacterial, anti-oxidant, anti-tussive, anti-diabetic, anti-inflammatory, and anti-plasmodialR. damascena Mill. F. trigintipetala DieckPolyphenols, flavonoid[232,277,278]
Zanthoxylum bungeanum Maxim EOHyperproliferation inhibition, anti-oxidants, anti-cancer, anti-inflammatory, anti-microbial, and anti-insecticidalZ. bungeanumD-Limonene, β-Myrcene[286,287,288,289,290,291]
Prunus armeniaca EOHyperproliferation inhibition, anti-cancer, anti-oxidant, anti-microbial, anti-inflammatory, and anti-asthmatic, P. armeniacaSeeds (Benzaldehyde, benzoic acid, mandelonitrile)[294,295,296,297,298,299,301]
Abbreviation: C. limetta, Citrus limetta; B. dracunculifolia, Baccharis dracunculifolia; P. frutescens, Perilla frutescens; C. maxima, Citrus maxima; O. vulgare, Origanum vulgare; CRV, Carvacrol; S. sahendica, Satureja sahendica; M. chamomilla, Matricaria chamomilla; H. annuus, Helianthus annuus; M. arvensis, Mentha arvensis; R. officinalis, Rosmarinus officinalis; C. longa, Curcuma longa; A. argyi, Artemisia argyi; L. angustifolia, Lavandula angustifolia; Z. coreanum, Zanthoxylum coreanum; A. citrodora, Aloysia citrodora; L. citriodora, Lippia citriodora; O. majorana, Origanum majorana; A. capillaris, Artemisia capillaris; C. japonica, Camellia japonica; O. syriacum, Origanum syriacum; O. ehrenbergii, Origanum ehrenbergii; C. formosana, Calocedrus formosana; M. quinquenervia; Melaleuca quinquenervia; C. cassia; Cinnamomum cassia; A. nantoensis, Alpinia nantoensis; C. boreale, Chrysanthemum boreale; V. negundo, Vitex negundo; A. millefolium, Achillea millefolium; V. zizanioides, Vetiveria zizanioides; E. camaldulensis, Eucalyptus camaldulensis, A. macrospadiceus, Acorus macrospadiceus; A. montana, Artemisia montana; P. lentiscus, Pistacia lentiscus; C. inophyllum, Calophyllum inophyllum; P. jacquemontiana, Parrotiopsis jacquemontiana; S. aurea, Salvia aurea; R. damascena, Rosa damascene; Z. bungeanum, Zanthoxylum bungeanum; P. armeniaca, Prunus armeniaca.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kim, H.J.; Hong, J.H. Multiplicative Effects of Essential Oils and Other Active Components on Skin Tissue and Skin Cancers. Int. J. Mol. Sci. 2024, 25, 5397. https://doi.org/10.3390/ijms25105397

AMA Style

Kim HJ, Hong JH. Multiplicative Effects of Essential Oils and Other Active Components on Skin Tissue and Skin Cancers. International Journal of Molecular Sciences. 2024; 25(10):5397. https://doi.org/10.3390/ijms25105397

Chicago/Turabian Style

Kim, Hyeong Jae, and Jeong Hee Hong. 2024. "Multiplicative Effects of Essential Oils and Other Active Components on Skin Tissue and Skin Cancers" International Journal of Molecular Sciences 25, no. 10: 5397. https://doi.org/10.3390/ijms25105397

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