Download PDF
Review  |  Open Access  |  17 Sep 2024

Role of skin enzymes in metabolism of topical drugs

Views: 158 |  Downloads: 15 |  Cited:  0 0
Metab Target Organ Damage 2024;4:32.
10.20517/mtod.2024.17 |  © The Author(s) 2024.
Author Information
Article Notes
Cite This Article

Abstract

Topical drugs have gained a lot of interest with their massive market growth and are available in various dosage forms. Prodrug compounds of transdermal delivery systems can be very different and designed to convert into the form of active pharmaceutical ingredients (APIs) through enzymatic action once they enter the body. The skin, as an interfacial barrier between the body and surroundings, has demonstrated critical roles in metabolizing, filtering, and detoxifying to minimize certain side effects and improve the medication benefits of topically administered products. It is well recognized that the drug pharmacokinetics can be altered by the presence of skin enzymes driven by biotransformation reactions. To evaluate the effectiveness of a topical generic drug product, its safety, and bioequivalence with the reference one, models assessing enzyme metabolic activity are highly required for testing the amount of drugs that are metabolized or can potentially be metabolized in both healthy and compromised skin. Thus, knowledge of skin composition and enzyme expression levels is of paramount importance in mapping the relevant metabolism that may have occurred. Regulatory authorities have also been making efforts to develop efficient and harmonizable protocols to evaluate the metabolism of transdermal products. This review is a compilation of reported skin metabolizing enzymes, including their role in both drug metabolism and homeostasis regulation, along with their localization and quantification in skin equivalents (and/or membrane layers). Various aspects that potentially affect the skin enzyme metabolism study were also discussed with respect to drug development considerations.

Keywords

Topical drugs, skin enzymes, metabolism, drug-metabolizing enzymes, homeostasis, skin models, vitamins, drug development

INTRODUCTION

Global topical drug delivery has gained extensive interest in the pharmaceutical and dermatological industry, signifying a market growth from USD $109.1 billion in 2023 to USD $176.8 billion by 2030[1,2]. Topical drugs are medication products that are directly administered to the spot of the outer skin surface[3,4], where the drug molecules can penetrate through skin layers, producing non-systemic (e.g., local effects within skin layers) or, to a lesser extent, systemic effects (e.g., enter blood circulation)[5]. It is well known that topical administration can avoid the hepatic first-pass metabolism that could otherwise reduce drug absorption and bioavailability. Over the last decades, however, studies have revealed significant expression of skin enzymes in the metabolic activity of topical drugs[6-8]. These enzymes are described as drug-metabolizing enzymes (DMEs), which are capable of performing biotransformation and detoxification of drug molecules, and also potentially result in activation of toxic metabolites and cutaneous reactions[6,9].

Despite numerous topical products on the market, the enzymatic profile of DMEs and apprehension of other skin enzymes (e.g., which enzymes are present, where exactly they are localized, and the underlying biotransformation mechanisms) remain unclear. Assessing the roles of skin enzymes and downstream metabolic pathways following drug exposure is an essential approach for drug development and targeted drug delivery to ensure equivalent safety and efficacy in clinical translation [from in vitro data to in vivo or in vitro-in vivo correlation (IVIVC)]. Thus, this review aims to provide an overview of: (i) skin metabolic enzymes (DMEs), as well as their interactions with substrates and drugs, (ii) the role of epidermal enzymes in maintaining homeostasis, (iii) the evaluation of DMEs cellular locations, molecular functions, and their abundances using advance analytical and modeling techniques, (iv) drugs and vitamins metabolism in skin, and (v) key considerations in drug development. The work is expected to provide updates on skin DMEs and support the development of new research models, strategies and regulatory authorities toward the stage of standardized clinical practice and drug testing improvement.

SKIN MORPHOLOGY

The skin is composed of a multilayered structure, including the epidermis, dermis, and hypodermis, and features an intricate network of cells, nerves, and receptors. The epidermis, which is a thin stratified layer of epithelium with five different cell strata, is divided into two regions: the stratum corneum (SC) or non-viable epidermis, and viable epidermis consisting of stratum lucidum, stratum granulosum, stratum spinosum, and stratum basale[10]. Keratinocytes are the major cell type in the epidermis, making up 95% of the total, and play a vital role in sustaining the integrity of the skin barrier and homeostasis. More details of the roles of keratinocytes in homeostasis will be discussed in the following section. Other important cells such as Langerhans cells, melanocytes, and Merkel cells are distributed throughout the viable layers. Starting from the innermost layer of the epidermis, the stratum basale (SB) is anchored to the basement membrane, which is also known as the dermal-epidermal junction, by integrins. This layer consists of a large volume of highly proliferative cells, including the epidermal progenitor and stem cells. Continuous proliferation of these stem cells gives rise to specified cells such as melanocytes and, in particular, keratinocytes that can undergo stratification, differentiate and migrate upwards (from basale to corneum layer) to displace the surficial cells, a process called keratinization[11]. Several transcription factors (TFs), such as the activator protein-1 (AP-1), nuclear factor erythroid-related factor 2 (Nrf2), and aryl hydrocarbon receptor (AhR), are thought to regulate the signaling pathway of keratinocyte differentiation genes and govern the epithelial development[12]. Among the skin TFs, AhR is found to be expressed in every type of skin cell, including keratinocytes, melanocytes, fibroblasts, and immune cells, and it has been identified as a promiscuous binding site for many endogenous and exogenous compounds. Following the keratinocyte differentiation, the nuclei eventually degrade, and other organelles are catabolized by various epidermal enzymes[13]. Finally, the terminally differentiated cells form the outermost SC, which is made up of anucleated and flattened dead keratinocytes (also termed corneocytes) embedded in the lipid matrix (such as ceramides, cholesterol, free fatty acids, and phospholipids). The arrangement of corneocytes in a dense lamellar structure in SC provides an efficient physical barrier of skin against the ingress of compounds from reaching viable tissue. The dermis, located underneath the SB layer, primarily consists of fibroblasts, which are responsible for the synthesis and secretion of collagen, elastin, and hyaluronic acid into the intercellular matrix. In the dermis, there are also other cells such as adipocytes, endothelial cells, and reticular fiber networks such as hair follicles, eccrine glands, and sebaceous glands. The hypodermis, the deepest layer of the skin, is composed of subcutaneous fat and loose connective tissue[14]. The compositions of skin with different stages of cell differentiation and expression reflect distinct barrier properties of the multilayered skin structure and, subsequently, the extent of metabolism.

DRUG-METABOLIZING ENZYMES

Once drugs pass the physical barrier, DMEs act as a biochemical barrier, metabolizing and detoxifying (or toxifying) drug compounds from skin cells[15]. DMEs are typically found in the viable epidermis, with the majority found in keratinocytes (the primary cells of the epidermis)[15-17]. These enzymes are classified into phase I (i.e., oxidation, reduction, hydrolysis) and phase II (i.e., conjugation)[18-20].

Phase I enzymes include cytochromes CYPs, alcohol dehydrogenases (ADHs), aldehyde dehydrogenase (ALDHs), aldehyde oxidases, amine oxidases (AOCs), carboxylesterase (CESs), cyclooxygenases, esterases, epoxide hydrolases (EPHX), leukotriene A4 hydrolases, and flavin-containing monooxygenases (FMOs)[8]. As one of the most versatile enzyme families, CYPs (especially CYP1-3) are responsible for > 75% of phase I metabolic activity of general drug types[19,21]. Along with the distinct classification of isozymes, most dermatological drugs are either substrates, inducers, or inhibitors of CYPs, resulting in a myriad of potential reactions (e.g., drug-drug interactions)[20]. CYPs and other enzymes from phase I facilitate detoxification through (i) the biotransformation of prodrugs to their active metabolites or by (ii) increasing the excretion rate that involves the conversion of lipophilic compounds into more hydrophilic compounds. Of importance, the former process has been associated with the potential generation of procarcinogens (which can then be metabolized into toxic carcinogens)[20,22]. On the other hand, the latter process may result in insufficient drug potency, leading to reduced therapeutic efficacy (or a low threshold for effectiveness). As a member of the cytochrome P450 group (phase I enzymes), CYP11A1 plays significant roles in the human skin, such as initiation of local steroidogenesis and metabolism of Vitamin D, as well as regulation of the protective barrier and skin immune functions. Skin pathology could happen with disturbances in CYP11A1 activity[23]. Action of CYP11A1 was observed on the unmodified side chain of Vitamin D3, which produces 20-hydroxyvitamin D3 [20(OH)D3] (considered as the major product), and further oxidizes the side chain to generate 20,23-dihydroxyvitamin D3 [20,23(OH)2D3]; 20,22-dihydroxyvitamin D3 [20,22(OH)2D3]; and 17,20-dihydroxyvitamin D3, with 20-hydroxyvitamin D3; 17,20,23-trihydroxyvitamin D3 [17,20,23(OH)3D3]; and 17-hydroxyvitamin D3 as minor products[24,25]. Other CYP enzymes such as CYP27A1, CYP2R1, CYP3A4, CYP24A1, and CYP27B1 could further metabolize the CYP11A1-derived hydroxyderivatives with the production of at least 18 hydroxy metabolites[25,26].

Phase II enzymes such as hydroxysteroid dehydrogenase (HSD), glutathione S-transferase (GST), UDP-glucuronosyltransferase (UGT), N-acetyltransferase (NAT), sulfotransferase, and acetyltransferase further promote the metabolism and elimination of drugs or intermediates through conjugation of more polar functional groups to the drugs. The GST is identified as the most prominent phase II enzyme, comprising three superfamilies (cytosolic, microsomal, mitochondrial)[27]. Altogether, the biotransformation of DMEs is characterized as the rate-determining steps in drug metabolism. Common DMEs found in human skin, including ex vivo native skin and reconstructed skin equivalents, are listed in Tables 1 and 2, though this list is not exhaustive.

Table 1

Common phase I drug metabolic enzymes detected in skin, its substrates or reactions performed, and topical drugs metabolized by the enzyme

EnzymeSubstrates/reactions performedTopical drugReference
ADH1AAldophosphamide, aliphatic alcoholsZiagen, cyclophosphamides[28]
ADH1BAliphatic alcohols, hydroxysteroids, retinolRetinyl palmitate[28]
ADH1C*Aliphatic alcohols, retinol, steroidsN/A[28]
ADH5Pentanol, ω-hydroxyl fatty acidsN/A[28]
ALDH1A1Acetaldehyde, aldophosphamide, aliphatic aldehydes, retinalN/A[29]
ALDH1B1Acetaldehyde, aliphatic aldehydes, lipid peroxidation-derived aldehydesN/A[29]
ALDH1L110-formyl-tetrahydrofolateN/A[29]
ALDH2Aldehydes Nitro-glycerine[29]
ALDH3A1Medium-chain aliphatic and aromatic aldehydesN/A[29]
ALDH3A2Fatty and long-chain aliphatic aldehydesN/A[29]
ALDH4A1Glutamate-ϒ-semialdehydeN/A[29]
ALDH6A1Methylmalonate semialdehyde, oxidative decarboxylation of methylmalonate semialdehyde to acetyl-CoA and propionyl-CoAN/A[30]
AOAza- (and oxa-) heterocycles, oxidize endogenous/exogenous aldehydes to carboxylic acidCarbazeran, citalopram, pyridoxal, tamoxifen, vanillin, zoniporide[31]
AKR1A1Aldehyde
N/A[32]
AKR1B1AldoseN/A[32]
AKR1C1-3Trans-dihydrodiols of aromatic hydrocarbonsN/A[32]
AKR1D1Δ4-3-ketosteroid 5β-stereospecificN/A[32]
AKR7A2Azole (e.g., posaconazole, voriconazole)Azole antifungal drug[33,34]
CBR1Aldehydes and ketones to hydroxylLaxoprofen, pentoxifylline[35]
CYP1A1Dibenzo [a, l] pyrene, 7,12-dimethybenz[α]anthraceneDacarbazine[36]
CYP1A2Dibenzo [a, l] pyrene, 7,12-dimethybenz[α]anthraceneAcetaminophen, caffeine, imipramine, pirfenidone, theophylline[36]
CYP1B1Dibenzo [a, l] pyrene, 17β-estradiol, fatty acid, polycyclic aromatic hydrocarbonsEstradiol, melatonin, retinol[37]
CYP2B6Bupropion, O-deethylation of 7 ethoxy-4-trifluoromethyl coumarinAminopyrine, clonazepam, diazepam[38]
CYP2E1Chlorzoxazone hydroxylation, para-nitrophenol hydroxylaseTretinoin[39]
CYP3A4/5/7Erytchromycin N-demethylase, cyclosporine Bexarotene, benzodiazepines, clindamycin, lidocaine, tretinoin, opioids, sandimmune [40]
CYP4B1N-hydroxylation of aromatic amines, W-hydroxylation of lauric acidN/A[41]
CYP4F819-Hydroxylation of prostaglandin H1 and H2N/A[41]
CYP4F12Oxidation of arachidonic acids into 18-hydroxy arachidonic acidN/A[41]
CYP7B125-hydroxycholesterol, 27-hydroxycholesterol, DHEAPrasterone[41]
CYP8A1EicosanoidsN/A[8]
CYP11A1Hydroxylation of cholesterol to pregnenolone, hydroxylation of Vitamin D3Lanosterol, lumisterol 3[26]
CYP2R125-hydroxylation of Vitamin D3N/A[42]
CYP24A1Hydroxylation of 1α, 25(OH)2D3 and 25(OH)D3N/A[42]
CYP26B1Retinoic acid (Vitamin A)Tretinoin[43]
CYP27A127-hydroxylation of cholesterol and 25-hydroxylation Vitamin D3 N/A[44]
CYP27B1Vitamin D3 metabolism N/A[45]
CYP3A4 and CYP39A17α-hydroxylation of 24-hydroxy cholesterol N/A[41]
CYP51A14β-desmethyllanosterol, 24,25-dihydrolanosterol, 24-methylenedihydrolanosaterol, obtusifoliolImidazole[46,47]
CYB5AN/ARuxolitinib[48]
FMO1N-oxygenation of secondary and tertiary amines (benzydamine)Benzydamine, ketoconazole, rifampicin[49]
FMO3Sulfides and tertiary amines to sulfoxide and N-oxideCimetidine, nicotine[50]
FMO5Ketones to estersN/A[50]
EPHX1 (dermis) and EPHX2 (epidermis)Hydrolysis of arene and aliphatic epoxide to less reactive and become more water-soluble dihydrodiol by the addition of water (Trans)N/A[51]
STS (epidermis)Hydrolysis of aryl and alkyl sulfateEstrone sulfate, lovastatin[52]
Table 2

Common phase II drug metabolic enzymes in skin, its substrates or reactions performed, with examples of inducers or inhibitors

EnzymeSubstrates/reactions performedTopical drugReference
17β-HSD115-androstane-3,17-diol (3-diol) into androsteroneGlucocorticoids[53]
GSTK1 (κ)CDNBN/A[54]
GSTM2 (µ)CDNB, glutathioneN/A[55]
GSTO1 (Ω)Ascorbic acid, CDNBN/A[56]
GSTP1 (π)CDNB, ethacrynic acid glutathione N/A[57]
GSTT1 (θ)CDNB S-hexylglutathioneN/A[58]
MGST11-S-(glutathionyl)-2,4,6-trinitrocyclohexadienate; glutathione; glutathionyl-S-dinitrobenzeneN/A[57]
SULT1A4N/AN/A[59]
SULT2B1DHEA, pregnenoloneViramune[59]
COMPTAscorbic acid, caffeic acid, daphnetin, flavonoids, gallic acid, green tea catechins, hydroxyquinolineBenserazide[60]
MPST3-mercaptopyruvate (with dihydrolipoic acid and thioredoxin) to hydrogen sulfideHydrogen sulfide[61]
TGM3First undergoes proteolysis to its active form
Acyl-transfer between glutamine and other primary amines essential for the formation of epithelial and hair follicle
N/A[62]
TPMTS-methylation of aromatic and heterocyclic sulphydrylAzathioprine[63]
TSTThiosulfateHydrogen sulfide, sodium thiosulfate[64]

IDENTIFICATION, CHARACTERIZATION AND ABUNDANCE OF SKIN ENZYMES

The advent of new approach methodologies (NAMs) such as omics- (e.g., metabolomics, genomics, proteomics and transcriptomics) and onics-based (e.g., electronics and photonics) tools have eased the bioprospection of a massive data for the enzymes, including the study of physicochemical properties and clinical information. Mass spectrometry (MS) is the most comprehensive proteomic tool and is often used in conjunction with other methods to manifest both qualitative and quantitative profiling at the single-cell level, as well as to discover novel enzymes.

Representing the most vital and largest interface between the body and surroundings, however, the skin is highly heterogeneous across intra- and inter-individual (or skin equivalents), resulting in various enzymatic activities and their cellular localization[16]. Kazem et al. have summarized the DMEs’ activities in different skin equivalents[65]. Overall, mRNA expression of phase I enzymes is more plentiful than phase II[65,66]. The mRNA of CYP1A1 and CYP1B1 was found to be expressed in all tested skin models, including ex vivo native human skin (NHS). Specifically, both CYP1A1 and CYP1B1 are colocalized within the epidermis (matured keratinocytes), fibroblasts, and sebaceous gland, with little expression in sweat producing cells[67]. Dysregulation of CYP1A1 and CYP1B1 has been observed to mediate reactions that result in mutagenic and carcinogenic metabolites[20,68]. Their clinical implications should be cautiously scrutinized during drug development. Although mRNA expression was detected in other CYP subfamilies (e.g., CYP4, CYP7, CYP8, CYP11, CYP21, CYP26, CYP27, CYP39 and CYP51), they showed no significant cellular functionality[20,65,68].

The fact is that there is a low correlation between the levels of mRNA, protein expression, and molecular activities. A recent study performed by Couto et al. has observed and quantified 2,000 skin proteins using MS with label-free quantification (MaxQuant)[8]. Herein, instead of phase I, a higher phase II enzyme expression was reported at the proteomic level in both NHS and skin equivalents. GSTP1 (π) was the highest abundant (62.65 ± 17.78 pmol·mg-1) phase II enzyme, followed by GSTM4 (µ) and GSTM3 (µ) at concentrations of 21.75 ± 0.63 pmol·mg-1 and 19.68 ± 10.29 pmol·mg-1, respectively. This finding corresponds to the data reported by Götz et al., Hewitt et al., and Eijl et al., where GSTP1 (π) protein (2-fold) and activity levels (8-fold) are higher in skin compared to liver[27,54,69]. The GSTP1 (π), in particular, has been demonstrated to play important roles in regulating epidermal cell growth and cellular apoptosis. Differentiated keratinocytes appeared to promote high expression of GSTP1 (π) and increase drug clearance[70]. In contrast to liver, UGT was not actively detected in any NHS and skin equivalents, suggesting that skin is a minor site for glucuronidation reaction[54]. Upon ligand binding to AhR, however, it may enhance UGT (UGT1A3) transcription in response to xenobiotic drug molecules. On the other hand, the most abundant enzymes from phase I are ADH1B, ADH1C, ALDH1A1, and carbonyl reductase (CBR), which are mainly found in the epidermis layers[71]. Among these enzymes, ADH1C is observed to be the highest expressed phase I enzyme (37.46 ± 15.05 pmol·mg-1), yet the concentration is much lower as compared to GSTP1 (π).

Unexpectedly, CBR3, which was understood to be expressed highly in skin, was reported to have lower expression levels compared to CBR1, which is predominantly expressed in liver, kidney, and intestines. Only three CYPs enzymes, which are CYP8A1 (5.52 ± 4.70 pmol·mg-1), CYP7B1 (2.57 ± 0.45 pmol·mg-1), and CYP51A1 (0.98 ± 0.09 pmol·mg-1), were detected. These CYPs enzymes are primarily involved in endogenous metabolism (i.e., cholesterol and eicosanoid metabolism). The result showed a controversy with the work of Kazem et al., where no enzyme activity was assessed for the above CYPs enzymes[65]. Being the most abundant phase I DME in liver, CYP3A4 has low or no expression in all tested skin models[7,66]. Meanwhile, FMO1 and FMO5 were highly detected in engineered models, whereas FMO3 was only found expressed in NHS[7]. Other phase I enzymes have been detected, including AOs, AOCs, CES, and EPHX. In addition, Couto et al. have also identified and quantified redox enzymes, proteases, and nucleases[8]. Thioredoxin and peroxiredoxin 1 were reported to be the most abundant antioxidant enzymes in skin. Another study done by Liu et al. combined liquid chromatography with tandem MS (LC/MS/MS), non-invasive tap strips method, and the label-free MaxLFQ to quantify skin proteins in volunteers[72]. Herein, up to 1,157 epidermal proteins, including TGM1, TGM3, ALOX12B, and ALOXE3, were matched to the UniProt database[72]. As one of the most crucial enzymes in cornification, TGM was identified to be expressed in the granulosum and spinosum layers[73].

The epidermis was recognized as the major reservoir site for skin esterase, which is responsible for the conversion of methyl salicylate to salicylic acid[74,75]. Telaprolu et al., instead, have identified greater metabolism in the dermis layer derived from the same donors, with greater area stained with esterase specific dye and a greater density, supporting higher dermal esterase prevalence[76]. The evidence further highlights the indispensable relationship between the biological complexity of skin models and enzyme activities. Nonetheless, the enzymes were mostly close or below the limit of detection[7]. The observed data can be explained by the discrepancy between the mRNA turnover rate and the protein translation level. Moreover, this can also be due to a limited metabolic pathway (i.e., a high saturation rate of metabolism). Skin metabolism is approximately only 10%-30% of that activity in liver, albeit most DMEs are detected in skin[16,65,77].

ROLE OF EPIDERMAL ENZYMES IN HOMEOSTASIS

The skin is continuously exposed to a number of endogenous and external compounds. In addition to serving as an effective barrier, it is capable of maintaining local and global homeostatic mechanisms (with respect to physical, chemical, immune, microbial and neuronal functional levels), i.e., pH, plasma concentration, and neuronal plasticity[78]. Apart from DMEs, there are other intra- and extracellular epidermal enzymes (EEs) responsible for regulating redox balance and dynamic equilibrium by enhancing the barrier structure functions[79,80]. As mentioned above, most enzymes are localized in the epidermal, and the driven biotransformation activity is substantially important in inducing the differentiation of organelle (i.e., melanin and keratin). For instance, TGMs mainly located in the spinosum and granulosum layers can synthesize and upregulate the expression of barrier-related proteins such as keratin, profilaggrin, loricrin and involurin, accelerating the keratinization and cornification. TGM-3, specifically, is a cross-linking protein for the formation of epidermal and reticular fibers[81]. Filaggrin is the major intermediate for the compaction of keratinocytes and the regulation of the epidermal terminal differentiation. During this process, the keratinocytes can modulate the expression and secretion of EEs, such as the kallikrein (KLK)-related proteases, enabling direct cleavage of extracellular and transmembrane epidermal proteins (e.g., corneodesmosin and desmocollin) in SC layer[80,82]. In healthy skin, desquamation and degradation of these proteins enable continual self-renewal of epidermal cells to maintain intact barrier homeostasis[82].

Moreover, essential lipids consisting of ceramides, cholesterol, and fatty acids (FA) are key components of the epidermis[78]. Common EEs involved in the lipids metabolism, that are important for keeping a functional skin barrier include β-glucocerebrosidase (converts glycosylceramides into ceramides), acidic sphingomyelinase (converts sphingomyelin into ceramides), and phospholipase A2 (converts phospholipids to free FAs and glycerol). Another relevant aspect is the effect of pH, which is directly relevant for cell differentiation, formation of epidermal lipids (and other lipid compartments), maintenance of skin microbiome, and the promotion of suboptimal substrate turnover[83]. The epidermis has a low pH of 4.0-6.0. Both β-glucocerebrosidase and sphingomyelinase are reported to work optimally at pH 5.0-5.5[83]. The free FAs formed by phospholipid breakdown contribute to the acidic environment in SC, which is required for the regulating activity of many of the enzymes in SC and the barrier function. In reaction with the main stressors in skin, such as ultraviolet radiation (UVR), the phase 1-dependent skin cells and DMEs may generate carcinogenic stressors, including the reactive oxygen species (ROS) and nitric oxide (NO)[84]. The UVR can be classified into UV-A, UV-B, and UV-C, each with different electrophysical properties, and their penetration into skin is dependent on the wavelength. UV-A tends to penetrate to the innermost epidermis layer and to the dermis, while UV-B can be absorbed fully by the epidermis melanocytes[11]. The resulting radicals are responsible for the induction of inflammation cascade and degenerative photoaging[11]. These radicals can also alter nucleic acids (DNA and RNA) and protein expression, which result in mutations and are the leading factor in melanoma or non-melanoma cancers and drug resistance[85,86]. To detoxify and eliminate these radicals, the skin is equipped with antioxidant EEs such as glutathione peroxidases (GPxs), superoxide dismutases (SODs), and catalases (CATs), which are expressed abundantly in the viable layers.

As a protective barrier between the internal human body and external environments, the sensory and adaptive capacity of skin could sustain local and global body homeostasis against harmful/unpleasant factors. The ability of skin to synthesize melatonin in response to external stress is critical and is considered one of its essential physiological functions[87]. Slominski et al. presented that melatonin (N-acetyl-5-methoxytryptamine) is a product of multistep tryptophan metabolism via serotonin and N-acetylserotonin[88]. Melatonin metabolism occurs rapidly through indolic and kynuric pathways caused by UVR or ROS. Antioxidative responses and DNA repair pathways can be stimulated by melatonin and metabolite products of melatonin. These properties of melatonin could be derived from the binding to quinone reductase 2 (NQO2), calmodulin (CaM) or the regulation of mitochondrial functions impacting cellular homeostasis. At pharmacological concentrations, melatonin can be used to counteract a number of damages caused by MT1- and MT2-independent mechanisms; this effect is mediated by the aryl hydrocarbon receptor (AhR), which is attributed to the structural similarity between melatonin and its natural ligands, such as tryptophan metabolites and indolic compounds[88].

In the research of Slominski et al., UV energy can touch the central neuroendocrine system and reset the body homeostasis, including that of skin (cutaneous homeostasis)[89]. UV absorption by the skin triggers mechanisms defending skin integrity, and also causes skin pathology, such as cancer, aging, and autoimmune responses. Melanins, known as skin colors or biopolymeric pigments, are produced by melanocytes (which are specialized pigment cells)[90]. Melanins are also considered polymorphous and multifunctional biopolymers, represented by eumelanin, pheomelanin, neuromelanin, and mixed melanin pigment[89]. L-tyrosine and L-DOPA play significant roles as the substrates and intermediates of melanogenesis. They also act as positive regulators of melanogenesis and other cellular functions of the body, depending on internal and external factors[91]. The optical and chemical filtering properties of melanins, together with their cytoprotective ability, could help protect skin cells against environmental factors such as solar/UV radiation, free radicals, and toxic chemicals. Melanins achieve this by binding to these harmful substances and then transferring them into more stable complexes[90,92]. Nevertheless, according to Karkoszka et al., owing to the formation of such complexes, locally and systemically administered drugs bound to melanins might not directly interact with receptors or enzymes.This could result in a lack of certain pharmacological and/or pharmacodynamic parameters, increasing the likelihood of side effects[92]. Additionally, there are risk factors for melanoma, deriving from the malignant transformation of melanin-producing melanocytes, which is a formidable malignancy[93].

Additionally, EEs are reported to be involved in hydration-induced changes (e.g., maturation of skin cells, desquamation process, and expression of protein) in different epidermal cells. For instance, a decrease in glycerol metabolism combined with reduced sebaceous gland activity leads to water loss in SC, causing an abnormal skin barrier and altered molecular mobility[94]. Several studies have also identified the role of EEs in antioxidant defense and redox metabolism; for instance, protein glutathione peroxidase 4 (GPX4) and thioredoxin reductase 1 (TXNRD1) are responsible for protecting skin against membrane lipid peroxidation[95].

METABOLISM OF DRUGS AND VITAMINS IN SKIN

Prodrugs and soft drugs

The entity of metabolites and the quantification of metabolomes have been a standard protocol to identify the amount of drugs permeated to different skin layers, and potentially predict the levels of metabolic activity. Prodrugs are intentionally designed to be inactive until they reach the site of action, in this case mainly within skin layers, and are converted to the active parent drugs by DMEs to exert pharmacological effects. These prodrugs can avoid extensive metabolism by skin enzymes, reduce off-target toxicity, and enhance the bioavailability of drugs at local sites. Telaprolu et al. have identified the presence of esterase enzymes in ex vivo human skin membranes by using methyl salicylate (in Metsal™ cream) as substrates[76]. Methyl salicylate is a widely available over-the-counter topical drug. It is a prodrug that is pharmacologically inert until it is metabolized into the therapeutically active salicylic acid (Aspirin) and methanol by the skin carboxylesterases in the viable epidermis and dermis layers [Figure 1]. The salicylic acid will be further metabolized (in liver) for clearance by phase II DMEs, i.e., CYP2C9, NAT2, and UGT1A6 through hydroxylation, acetylation, and glucuronidation, respectively[96].

Role of skin enzymes in metabolism of topical drugs

Figure 1. Metabolism of methyl salicylate by skin esterases. (Reproduced from “Human skin drug metabolism: relationships between methyl salicylate metabolism and esterase activities in ivpt skin membranes” by Krishna et al., Metabolites 2023).

Another example is tamoxifen, which is traditionally used as first-line treatment in breast cancer but has more recently been identified as a potential drug for exophytic dermatitis[97,98]. The parent drug is bio-transformed by CYP2C9 to its potent form, 4-hydroxytamoxifen, with minimal off-target issues. Pre-formulation with nano-emulgel further increases its lipid solubility and enhances its topical SC penetration[97]. Hyaluronic acid (HA) is broadly incorporated in many cosmetics formulations to enhance skin rejuvenation and hydration, along with the capability to potentially support the wound healing process and prevent skin necrosis[99]. Topical HA, in its non-cross-linked form, however, is highly susceptible to enzymatic breakdown (e.g., hyaluronidase). Fortunately, the nano-prodrug formulation enhances its bioavailability by enabling controlled bioactivation and release upon reaching the target site, such as radicals production, overexpressed enzymes, and low pH intracellular environment[99]. Naturally-derived drug compounds (e.g. silibinin) exhibit low chemical stability and undergo rapid metabolism (mainly mediated by UGT1A1, UGT1A6, UGT1A9, GST and SULT), leading to rapid clearance from the body[100]. Silibinin is known for its photo-protective capabilities against UV-B to prevent hyperproliferation and differentiation of epidermal keratinocytes, and regulate inflammation signaling cascades[101]. Several research studies have been conducted to improve silibinin bioavailability through prodrug modification[102]. Silibinin loaded in hydrogel polymer with a combination of chitosan and fucoidan has been shown to favor longer drug retention time and skin distribution[103].

Contrary to prodrugs, soft drugs are therapeutically active molecules that undergo rapid systemic metabolism into inactive metabolites after exerting their local effects. The active drugs, thus, do not enter the systemic circulation and are promptly cleared from the body[104]. For example, crisaborole is a topical soft drug used to treat atopic dermatitis (AD). The drug is metabolized into 5-(4-cyanophenoxy)-2-hydroxyl benzylalcohol (AN7602) by CYP enzymes (e.g., CYP1A1/2 and CYP3A4) and hydrolases. It was then further metabolized to 5-(4-cyanophenoxy)-2-hydroxyl benzoic acid and AN7602-sulfate through oxidation and sulfation[105]. Both metabolites are inactive products lacking glucocorticoid activity[106]. Tom et al. have demonstrated relatively low levels of crisaborole topical ointment in human plasma, suggesting systemic detoxification through enzyme metabolism[106].

Vitamins

Vitamins (i.e., Vitamins A, C, D, and E) are actively used as important ingredients in many topical products. Endogenous retinoic acid (RA) is triggered during wound-induced hair follicle neogenesis (WIHN) and other skin damage responses (e.g., laser treatments)[107]. Bioactivation of Vitamin A (retinol) is through two-step oxidation, forming retinaldehyde and later to the biologically active RA by retinol dehydrogenase (ALDH1 family) and CYP26 enzymes, respectively[108]. Retinol can also undergo esterification with fatty acid to form retinyl esters by retinol acyltransferase (LRAT) [Figure 2].

Role of skin enzymes in metabolism of topical drugs

Figure 2. Enzyme-catalyzed Vitamin A metabolism. (Reproduced from “Retinol and retinyl esters: biochemistry and physiology” by Sheila M and William S, J Lipid Res 2013).

Synthesis of Vitamin D mainly occurs in the epidermis, where epidermal keratinocytes carry the most metabolic enzyme activity. Vitamin D3 is produced from 7-dehydrocholesterol (7-DHC) (photolysis) or derived from diet. CYP27A is reported to metabolize Vitamin D3 to 25OHD and further to its active metabolite 1,25(OH)2D or 24,25(OH)2D by CYP27B1 and CYP24A1, respectively[44,45].

DRUG DEVELOPMENT CONSIDERATIONS

The key considerations in topical drug development are to test whether the product is safe, potent and capable of reaching its desired clinical efficacy. Following the phase-out of animal trials in research, as mandated by FDA Modernization Act 2.0, a number of in vitro and in vivo skin permeation models, as well as NAMs, have been implemented to evaluate skin metabolism under controlled settings to allow for the optimization and delivery control[109]. Generic topical dermatological products can be safe, effective and affordable alternatives to branded medications. Yet, the complex nature of skin can impact skin enzyme expression and metabolic activity, causing high variability in the pharmacokinetics and pharmacodynamics (PK/PD) of the drugs[7]. Important scientific questions, thus, need to be addressed for drug assessment.

Roles of skin enzymes

The enzymes involved in the biotransformation of topically administered drugs, as well as their biotransformation mechanism and pathway, should be thoroughly investigated. This includes the reaction kinetics (Vmax/Km/Kcat/CLint), and toxicology profiles with potential induction and inhibition of enzymes. The enzyme saturation state is essential for determining the reaction rate and the amount of parent drugs available, especially when prodrugs or soft drugs are applied. Drug interaction following the enzyme biotransformation (or drug combinations) generates reactive metabolites that could potentially induce toxicity, causing pre- or post-marketing withdrawals. Although most DMEs in skin showed low basal activity, leave-on topical products can have significant time-concentration effects following prolonged exposure.

Genetic variation

Intra- (e.g., transcriptomic heterogeneity across different skin regions) and inter- (e.g., age, gender, racial) individual differences can lead to case-to-case variation in drug activities.

Skin nature

The skin biological complexity, maturation stage, culture and isolation methods (from selection, preparation to storage conditions) can make huge disparities in drug permeation and enzyme expression. The reconstructed skin has an overall lower number of enzymes detected compared to ex vivo native human skin (NHS)[77]. The NHS and synthetic reconstructed skin comprise only an approximate 32% overlap in the identification and quantification of enzymes[8].

Correlation between mRNA-protein-cellular activities of enzymes

Levels of mRNA were shown to have poor proxies toward the quantification of protein and activity[69]. Plus, many DME activities are close to the limits of detection and quantification, which could reduce accuracy[8,69].

Skin equivalents

The skin models should have the ability to reflect in vivo conditions of drug metabolism. The selection, preparation, and isolation methods (e.g., heat and frozen treatments) are shown to affect enzyme turnover and metabolic activity[76].

There are also investigations regarding comorbidities associated with metabolic dysfunction that result in changes in DME expression and activities[110]. The alterations due to the disease state itself, as well as the treatments and comedication (e.g., drug- and/or enzyme-drug interactions)[110,111], can lead to a loss of enzyme activity, causing the enzymes to fail to respond properly to the drugs. At the same time, many autoimmune and inflammatory-mediated reactions can also lead to abnormal mRNA and protein turnover. For instance, Sumantran et al. observed a meaningful reduction in DME levels in both melanomas and psoriasis[112]. Another study done by Candi et al. showed overexpression of TGM-5 in patients with ichthyosis vulgaris, while nearly absent in Darier’s disease-affected skin[73]. Literature studies also suggested that various skin diseases can manifest the generation of free radicals (e.g., ROS) and trigger subsequent inflammatory responses, eventually resulting in the destruction of homeostasis and redox balance. Perihan et al. reported that in patients with acne vulgaris, there were significantly exacerbated levels of antioxidant EEs, as compared to healthy controls, due to the increase in oxidative stressors[113]. Conversely, the expression of EEs was found to decrease in severe acne than mild or acute conditions[113]. This could probably be attributed to the overproduction of ROS that leads to cellular senescence of enzyme-related proteins[95]. While evidence linking comorbidities status with enzyme expression, the exact underlying mechanism remains to be discovered.

Additionally, skin diseases could also alter the epidermal keratinocyte differentiation program, causing a defective skin barrier and affecting the delivery of topically administered drugs[95]. Psoriasis features hyperproliferation and upregulation of keratinocyte differentiation, resulting in abnormal protein metabolism and redox regulation. This disturbance contributes to the improper development of skin adhesion and junction proteins essential for intact barrier formation[114,115]. Meanwhile, the levels of filaggrin, together with the EEs involved in SC lipid biosynthesis, were significantly low in atopic dermatitis[116]. Alterations in the expression of barrier-related protein (or nucleic acids) are believed to contribute to epidermal cell degradation, increased lipids peroxidation and membrane permeability of topical drugs. Skin affected with vitiligo has selective loss of melanin resulting from autoimmune destruction of melanocytes in the epidermis and hair follicles. This disrupts the balance of the cutaneous antioxidant system, including both enzymatic and non-enzymatic molecules, inducing a pre-senescent status and compromised metabolic function[117]. Progressively, irregular expression of these enzymes and barrier-related proteins could result in skin barrier dysfunction, reduced drug biotransformation, and eventually result in excessive drug accumulation at local sites or in systemic circulation.

Hence, it is always important to understand the comprehensive role of enzymes in skin to ensure the safety and efficacy of the topically administered drugs. Information on metabolizing enzymes in skin is also highly helpful in assessing and comprehending the bioequivalence of topical products in terms of qualitative (Q1) and quantitative (Q2) composition, as well as physical and microstructural arrangement of matter (Q3). Even if Q1/Q2 similarity is achieved, the grade of excipients and enhancers needs to be considered, as they can have a significant impact on enzyme metabolism and final drug product quality attributes.

CONCLUSION

Numerous works have demonstrated the presence of DMEs with significant skin metabolizing activity. Although enzyme expression (both transcriptional and translational levels) and activity in the skin are generally much lower than in the liver, the skin is still the largest interface between the body and surroundings; hence, the skin’s net capacity for metabolic processes and other cellular effects can be considerable. Owing to their ability to induce biotransformation and bioactivation, these DMEs can bring both positive and adverse impacts. A comprehensive understanding of these enzymes could provide a basis for promising in vitro to in vivo extrapolation, including dose optimization and targeted drug delivery. However, there are no verified and validated standard protocols for quantification parameters of enzymes in the skin compartment, hampering the efficiency and reliability of research findings, thereby reducing the accuracy of assessment tools. Furthermore, the complexity of skin morphology, the scarcity of studies on the relationship between skin enzymes and topical drugs, and the low correlation among different types of data derived from mRNA, protein, and activity studies pose further challenges in drug development. Given these issues, future research shall continue to fill the gaps relating to complications of metabolic profiling, improve data incorporation into modeling systems, and refine existing predictive frameworks.

DECLARATIONS

Authors’ contributions

Writing - original draft preparation: Lau N

Writing - review and editing: Phan K

Supervision, revisions and proofing: Mohammed Y

Availability of data and materials

Not applicable.

Financial support and sponsorship

None.

Conflicts of interest

The authors declared that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2024.

REFERENCES

1. Global topical drug delivery market (2023-2028) by products, routes of administration, end-user, and geography, competitive analysis, impact of covid-19 with ansoff analysis. Available from: https://www.researchandmarkets.com/reports/5696819/global-drug-delivery-devices-market-2023-2028. [Last accessed on 10 Sep 2024].

2. Topical drug delivery - global strategic business report. Available from: https://www.researchandmarkets.com/reports/5303550/topical-drug-delivery-global-strategic. [Last accessed on 10 Sep 2024].

3. Raina N, Rani R, Thakur VK, Gupta M. New insights in topical drug delivery for skin disorders: from a nanotechnological perspective. ACS Omega 19;8:19145-167.

4. Buhse L, Kolinski R, Westenberger B, et al. Topical drug classification. Int J Pharm 2005;295:101-12.

5. Hmingthansanga V, Singh N, Banerjee S, Manickam S, Velayutham R, Natesan S. Improved topical drug delivery: role of permeation enhancers and advanced approaches. Pharmaceutics 2022;14:2818.

6. Eilstein J, Léreaux G, Arbey E, Daronnat E, Wilkinson S, Duché D. Xenobiotic metabolizing enzymes in human skin and SkinEthic reconstructed human skin models. Exp Dermatol 2015;24:547-9.

7. Wiegand C, Hewitt NJ, Merk HF, Reisinger K. Dermal xenobiotic metabolism: a comparison between native human skin, four in vitro skin test systems and a liver system. Skin Pharmacol Physiol 2014;27:263-75.

8. Couto N, Newton JRA, Russo C, et al. Label-free quantitative proteomics and substrate-based mass spectrometry imaging of xenobiotic metabolizing enzymes in ex vivo human skin and a human living skin equivalent model. Drug Metab Dispos 2021;49:39-52.

9. Leake CD. Annual review of pharmacology and toxicology: review of reviews. Annu Rev Pharmacol Toxicol 1978;18:581-8.

10. Almeida M, Diogo R. Human enhancement: genetic engineering and evolution. Evol Med Public Health 2019;2019:183-9.

11. D’Orazio J, Jarrett S, Amaro-Ortiz A, Scott T. UV radiation and the skin. Int J Mol Sci 2013;14:12222-48.

12. Steinhoff M, Alam M, Ahmad A, Uddin S, Buddenkotte J. Targeting oncogenic transcription factors in skin malignancies: an update on cancer stemness and therapeutic outcomes. Semin Cancer Biol 2022;87:98-116.

13. Simpson CL, Patel DM, Green KJ. Deconstructing the skin: cytoarchitectural determinants of epidermal morphogenesis. Nat Rev Mol Cell Biol 2011;12:565-80.

14. Kolarsick PAJ, Kolarsick MA, Goodwin C. Anatomy and physiology of the skin. J Dermat Nurses Asso 2011;3:203-13.

15. Oesch F, Fabian E, Oesch-Bartlomowicz B, Werner C, Landsiedel R. Drug-metabolizing enzymes in the skin of man, rat, and pig. Drug Metab Rev 2007;39:659-98.

16. Oesch F, Fabian E, Landsiedel R. Xenobiotica-metabolizing enzymes in the skin of rat, mouse, pig, guinea pig, man, and in human skin models. Arch Toxicol 2018;92:2411-56.

17. Baron JM, Höller D, Schiffer R, et al. Expression of multiple cytochrome p450 enzymes and multidrug resistance-associated transport proteins in human skin keratinocytes. J Invest Dermatol 2001;116:541-8.

18. Pyo SM, Maibach HI. Maibach, skin metabolism: relevance of skin enzymes for rational drug design. Skin Pharmacol Physiol 2019;32:283-94.

19. Rekka EA, Kourounakis PN, Pantelidou M. Xenobiotic metabolising enzymes: impact on pathologic conditions, drug interactions and drug design. Curr Top Med Chem 2019;19:276-91.

20. Ahmad N, Mukhtar H. Cytochrome p450: a target for drug development for skin diseases. J Invest Dermatol 2004;123:417-25.

21. Rendic S, Guengerich FP. Guengerich, survey of human oxidoreductases and cytochrome P450 enzymes involved in the metabolism of xenobiotic and natural chemicals. Chem Res Toxicol 2015;28:38-42.

22. Neis MM, Wendel A, Wiederholt T, et al. Expression and induction of cytochrome P450 isoenzymes in human skin equivalents. Skin Pharmacol Physiol 2010;23:29-39.

23. Slominski RM, Raman C, Elmets C, Jetten AM, Slominski AT, Tuckey RC. The significance of CYP11A1 expression in skin physiology and pathology. Mol Cell Endocrinol 2021;530:111238.

24. Slominski A, Semak I, Zjawiony J, et al. The cytochrome P450scc system opens an alternate pathway of vitamin D3 metabolism. FEBS J 2005;272:4080-90.

25. Slominski AT, Kim TK, Janjetovic Z, et al. Biological effects of CYP11A1-derived Vitamin D and lumisterol metabolites in the skin. J Invest Dermatol 2024;11:S0022-202X(24)00386.

26. Slominski AT, Li W, Kim TK, et al. Novel activities of CYP11A1 and their potential physiological significance. J Steroid Biochem Mol Biol 2015;151:25-37.

27. van Eijl S, Zhu Z, Cupitt J, et al. Elucidation of xenobiotic metabolism pathways in human skin and human skin models by proteomic profiling. PLoS One 2012;7:e41721.

28. Di L, Balesano A, Jordan S, Shi SM. The role of alcohol dehydrogenase in drug metabolism: beyond ethanol oxidation. AAPS J 2021;23:20.

29. Muzio G, Maggiora M, Paiuzzi E, Oraldi M, Canuto RA. Aldehyde dehydrogenases and cell proliferation. Free Radic Biol Med 2012;52:735-46.

30. Xia J, Li S, Liu S, Zhang L. Aldehyde dehydrogenase in solid tumors and other diseases: potential biomarkers and therapeutic targets. MedComm (2020) 2023;4:e195.

31. Manevski N, Balavenkatraman KK, Bertschi B, et al. Aldehyde oxidase activity in fresh human skin. Drug Metab Dispos 2014;42:2049-57.

32. Barski OA, Tipparaju SM, Bhatnagar A. Bhatnagar, the aldo-keto reductase superfamily and its role in drug metabolism and detoxification. Drug Metab Rev 2008;40:553-624.

33. Wu W, Jiang T, Lin H, Chen C, et al. The specific binding and promotion effect of azoles on human aldo-keto reductase 7A2. Metabolites 2023;13:601.

34. Lee A, Tysome JR, Saeed SR. Topical azole treatments for otomycosis. Cochrane Database Syst Rev 2021;5:Cd009289.

35. Shi SM, Di L. The role of carbonyl reductase 1 in drug discovery and development. Expert Opin Drug Metab Toxicol 2017;13:859-70.

36. Kapelyukh Y, Henderson CJ, Scheer N, et al. Defining the contribution of CYP1A1 and CYP1A2 to Drug metabolism using humanized CYP1A1/1A2 and Cyp1a1/Cyp1a2 knockout mice. Drug Metab Dispos 2019;47:907-18.

37. Li F, Zhu W, Gonzalez FJ. Gonzalez, potential role of CYP1B1 in the development and treatment of metabolic diseases. Pharmacol Ther 2017;178:18-30.

38. Hedrich WD, Hassan HE, Wang H. Insights into CYP2B6-mediated drug-drug interactions. Acta Pharm Sin B 2016;6:413-25.

39. Szymański Ł, Skopek R, Palusińska M, et al. Retinoic acid and its derivatives in skin. Cells 2020;9:2660.

40. Hakkola J, Hukkanen J, Turpeinen M, Pelkonen O. Inhibition and induction of CYP enzymes in humans: an update. Arch Toxicol 2020;94:3671-722.

41. Nebert DW, Wikvall K, Miller WL. Human cytochromes P450 in health and disease. Philos Trans R Soc Lond B Biol Sci 2013;368:20120431.

42. Tuckey RC, Cheng CYS, Slominski AT. Slominski, the serum vitamin d metabolome: what we know and what is still to discover. J Steroid Biochem Mol Biol 2019;186:4-21.

43. O’Byrne SM, Blaner WS. Retinol and retinyl esters: biochemistry and physiology. J Lipid Res 2013;54:1731-43.

44. Gottfried E, Rehli M, Hahn J, Holler E, Andreesen R, Kreutz M. Monocyte-derived cells express CYP27A1 and convert vitamin D3 into its active metabolite. Biochem Biophys Res Commun 2006;349:209-13.

45. Bikle DD. Vitamin D metabolism and function in the skin. Mol Cell Endocrinol 2011;347:80-9.

46. Lepesheva GI, Waterman MR. Sterol 14alpha-demethylase cytochrome P450 (CYP51), a P450 in all biological kingdoms. Biochim Biophys Acta 2007;1770:467-77.

47. Lepesheva GI, Hargrove TY, Kleshchenko Y, Nes WD, Villalta F, Waterman MR. CYP51: a major drug target in the cytochrome P450 superfamily. Lipids 2008;43:1117-25.

48. Guo H, Liang S, Wang Y, et al. Cytochrome B5 type A alleviates HCC metastasis via regulating STOML2 related autophagy and promoting sensitivity to ruxolitinib. Cell Death Dis 2022;13:623.

49. Phillips IR, Shephard EA. Drug metabolism by flavin-containing monooxygenases of human and mouse. Expert Opin Drug Metab Toxicol 2017;13:167-181.

50. Fukami T, Yokoi T, Nakajima M. Non-P450 drug-metabolizing enzymes: contribution to drug disposition, toxicity, and development. Annu Rev Pharmacol Toxicol 2022;62:405-25.

51. Gautheron J, Jéru I. The multifaceted role of epoxide hydrolases in human health and disease. Int J Mol Sci 2020;22:13.

52. Rižner TL. The important roles of steroid sulfatase and sulfotransferases in gynecological diseases. Front Pharmacol 2016;7:30.

53. Marchais-Oberwinkler S, Henn C, Möller G, et al. 17β-hydroxysteroid dehydrogenases (17β-HSDs) as therapeutic targets: protein structures, functions, and recent progress in inhibitor development. J Steroid Biochem Mol Biol 2011;125:66-82.

54. Götz C, Pfeiffer R, Tigges J, et al. Xenobiotic metabolism capacities of human skin in comparison with a 3D epidermis model and keratinocyte-based cell culture as in vitro alternatives for chemical testing: activating enzymes (Phase I). Exp Dermatol 2012;21:358-63.

55. Federici L, Lo Sterzo C, Pezzola S, et al. Structural basis for the binding of the anticancer compound 6-(7-nitro-2,1,3-benzoxadiazol-4-ylthio)hexanol to human glutathione s-transferases. Cancer Res 2009;69:8025-34.

56. Zhou H, Brock J, Liu D, Board PG, Oakley AJ. Structural insights into the dehydroascorbate reductase activity of human omega-class glutathione transferases. J Mol Biol 2012;420:190-203.

57. Patskovsky Y, Patskovska L, Almo SC, Listowsky I. Transition state model and mechanism of nucleophilic aromatic substitution reactions catalyzed by human glutathione S-transferase M1a-1a. Biochemistry 2006;45:3852-62.

58. Tars K, Larsson AK, Shokeer A, Olin B, Mannervik B, Kleywegt GJ. Structural basis of the suppressed catalytic activity of wild-type human glutathione transferase T1-1 compared to its W234R mutant. J Mol Biol 2006;355:96-105.

59. Gamage N, Barnett A, Hempel N, et al. Human sulfotransferases and their role in chemical metabolism. Toxicol Sci 2006;90:5-22.

60. Wang FY, Wang P, Zhao DF, et al. Analytical methodologies for sensing catechol-O-methyltransferase activity and their applications. J Pharm Anal 2021;11:15-27.

61. Rao SP, Dobariya P, Bellamkonda H, More SS. Role of 3-mercaptopyruvate sulfurtransferase (3-MST) in physiology and disease. Antioxidants (Basel) 2023;12:603.

62. Chermnykh ES, Alpeeva EV, Vorotelyak EA. Vorotelyak, transglutaminase 3: the involvement in epithelial differentiation and cancer. Cells 2020;9:1996.

63. Lee M, Seo J, Bang D, Kim DY. Thiopurine S-methyltransferase polymorphisms in korean dermatologic patients. Ann Dermatol 2017;29:529-35.

64. Kruithof PD, Lunev S, Aguilar Lozano SP, et al. Unraveling the role of thiosulfate sulfurtransferase in metabolic diseases. Biochim Biophys Acta Mol Basis Dis 2020;1866:165716.

65. Kazem S, Linssen EC, Gibbs S. Skin metabolism phase I and phase II enzymes in native and reconstructed human skin: a short review. Drug Discov Today 2019;24:1899-1910.

66. Hu T, Khambatta ZS, Hayden PJ, et al. Xenobiotic metabolism gene expression in the EpiDerm™ in vitro 3D human epidermis model compared to human skin. Toxicology in Vitro 2010;24:1450-63.

67. Janmohamed A, Dolphin CT, Phillips IR, Shephard EA. Quantification and cellular localization of expression in human skin of genes encoding flavin-containing monooxygenases and cytochromes P450. Biochem Pharmacol 2001;62:777-86.

68. Shimizu Y, Sasaki T, Yonekawa E. Association of CYP1A1 and CYP1B1 inhibition in in vitro assays with drug-induced liver injury. J Toxicol Sci 2021;46:167-76.

69. Hewitt NJ, Edwards RJ, Fritsche E, et al. Use of human in vitro skin models for accurate and ethical risk assessment: metabolic considerations. Toxicol Sci 2013;133:209-17.

70. Ściskalska M, Milnerowicz H. The role of GSTπ isoform in the cells signalling and anticancer therapy. Eur Rev Med Pharmacol Sci 2020;24:8537-50.

71. Lockley DJ, Howes D, Williams FM. Cutaneous metabolism of glycol ethers. Arch Toxicol 2005;79:160-8.

72. Liu M, Zhang J, Wang Y, et al. Non-invasive proteome-wide quantification of skin barrier-related proteins using label-free LC-MS/MS analysis. Mol Med Rep 2020;21:2227-35.

73. Candi E, Oddi S, Paradisi A, et al. Expression of transglutaminase 5 in normal and pathologic human epidermis. J Invest Dermatol 2002;119:670-7.

74. Tokudome Y, Katayanagi M, Hashimoto F. Esterase activity and intracellular localization in reconstructed human epidermal cultured skin models. Ann Dermatol 2015;27:269-74.

75. Lau WM, Ng KW, Sakenyte K, Heard CM. Distribution of esterase activity in porcine ear skin, and the effects of freezing and heat separation. Int J Pharm 2012;433:10-15.

76. Telaprolu KC, Grice JE, Mohammed YH, Roberts MS. Human skin drug metabolism: relationships between methyl salicylate metabolism and esterase activities in ivpt skin membranes. Metabolite 2023;13:934.

77. Eilstein J, Léreaux G, Budimir N, Hussler G, Wilkinson S, Duché D. Comparison of xenobiotic metabolizing enzyme activities in ex vivo human skin and reconstructed human skin models from SkinEthic. Arch Toxicol 2014;88:1681-94.

78. Lefèvre-Utile A, Braun C, Haftek M, Aubin F. Five functional aspects of the epidermal barrier. Int J Mol Sc 2021;22:11676.

79. Redoules D, Tarroux R, Périé J. Epidermal enzymes: their role in homeostasis and their relationships with dermatoses. Skin Pharmacol Appl Skin Physiol 1998;11:183-92.

80. Nauroy P, Nyström A. Kallikreins: essential epidermal messengers for regulation of the skin microenvironment during homeostasis, repair and disease. Matrix Biol Plus 2020;6-7:100019.

81. Tabolacci C, Lentini A, Provenzano B, Beninati S. Evidences for a role of protein cross-links in transglutaminase-related disease. Amino Acids 2012;42:975-86.

82. Wu Z, Hansmann B, Meyer-Hoffert U, Gläser R, Schröder JM. Molecular identification and expression analysis of filaggrin-2, a member of the S100 fused-type protein family. PLoS One 2009;4:e5227.

83. Lukić M, Pantelić I, Savić SD. Towards optimal ph of the skin and topical formulations: from the current state of the art to tailored products. Cosmetics 2021;8:69.

84. Slominski AT, Zmijewski MA, Skobowiat C, Zbytek B, Slominski RM, Steketee JD. Introduction, in sensing the environment: regulation of local and global homeostasis by the skin’s neuroendocrine system. Adv Anat Embryol Cell Biol 2012;212:1-115.

85. Veith A, Moorthy B. Role of cytochrome P450s in the generation and metabolism of reactive oxygen species. Curr Opin Toxicol 2018;7:44-51.

86. Slominski RM, Chen JY, Raman C, Slominski AT. Photo-neuro-immuno-endocrinology: how the ultraviolet radiation regulates the body, brain, and immune system. Proc Natl Acad Sci U S A 2024;121:e2308374121.

87. Slominski AT, Zmijewski MA, Semak I, et al. Melatonin, mitochondria, and the skin. Cell Mol Life Sci 2017;74:3913-25.

88. Slominski AT, Kim TK, Slominski RM. Melatonin and its metabolites can serve as agonists on the aryl hydrocarbon receptor and peroxisome proliferator-activated receptor gamma. Int J Mol Sci 2023;24:15496.

89. Slominski AT, Zmijewski MA, Plonka PM, Szaflarski JP, Paus R. How UV light touches the brain and endocrine system through skin, and why. Endocrinology 2018;159:1992-2007.

90. Slominski A, Tobin DJ, Shibahara S, Wortsman J. Melanin pigmentation in mammalian skin and its hormonal regulation. Physiol Rev 2004;84:1155-228.

91. Slominski A, Zmijewski MA, Pawelek J. L-tyrosine and L-dihydroxyphenylalanine as hormone-like regulators of melanocyte functions. Pigment Cell Melanoma Res 2012;25:14-27.

92. Karkoszka M, Rok J, Wrzesniok D. Melanin biopolymers in pharmacology and medicine-skin pigmentation disorders, implications for drug action, adverse effects and therapy. Pharmaceuticals (Basel) 2024;17:521.

93. Slominski RM, Kim TK, Janjetovic Z, et al. Malignant melanoma: an overview, new perspectives, and Vitamin D signaling. Cancers (Basel) 2024;16:2262.

94. Feingold KR. Thematic review series: skin lipids. The role of epidermal lipids in cutaneous permeability barrier homeostasis. J Lipid Res 2007;48:2531-46.

95. Jaffri J. Reactive oxygen species and antioxidant system in selected skin disorders. Malays J Med Sci 2023;30:7-20.

96. Palikhe NS, Kim SH, Nam YH, Ye YM, Park HS. Polymorphisms of aspirin-metabolizing enzymes CYP2C9, NAT2 and UGT1A6 in aspirin-intolerant urticaria. Allergy Asthma Immunol Res 2011;3:273-6.

97. Alyami MH, Alyami HS, Alshehri AA, Alsharif WK, Shaikh IA, Algahtani TS. Tamoxifen citrate containing topical nanoemulgel prepared by ultrasonication technique: formulation design and in vitro evaluation. Gels 2022;8:456.

98. Soares-Lopes LR, Soares-Lopes IM, Filho LL, Alencar AP, da Silva BB. Morphological and morphometric analysis of the effects of intralesional tamoxifen on keloids. Exp Biol Med (Maywood) 2017;242:926-9.

99. Machado V, Morais M, Medeiros R. Hyaluronic acid-based nanomaterials applied to cancer: where are we now? Pharmaceutics 2022;14:2092.

100. Zhao J, Agarwal R. Tissue distribution of silibinin, the major active constituent of silymarin, in mice and its association with enhancement of phase II enzymes: implications in cancer chemoprevention. Carcinogenesis 1999;20:2101-8.

101. Singh RP, Agarwal R. Cosmeceuticals and silibinin. Clinics in Dermatology 2009;27:479-84.

102. Romanucci V, Giordano M, Pagano R, et al. Investigation on the solid-phase synthesis of silybin prodrugs and their timed-release. Bioorg Med Chem 2021;50:116478.

103. Karami M, Sharif Makhmalzadeh B, Pooranian M, Rezai A. Preparation and optimization of silibinin-loaded chitosan-fucoidan hydrogel: an in vivo evaluation of skin protection against UVB. Pharm Dev Technol 2021;26:209-19.

104. Bell M, Foley D, Naylor C, et al. Discovery of soft-drug topical tool modulators of sphingosine-1-phosphate receptor 1 (S1PR1). ACS Med Chem Lett 2019;10:341-7.

105. Aprile S, Serafini M, Pirali T. Soft drugs for dermatological applications: recent trends. Drug Discovery Today 2019;24:2234-46.

106. Tom WL, Van Syoc M, Chanda S, Zane LT. Pharmacokinetic profile, safety, and tolerability of crisaborole topical ointment, 2% in adolescents with atopic dermatitis: an open-label phase 2a study. Pediatr Dermatol 2016;33:150-9.

107. Kim D, Chen R, Sheu M, et al. Noncoding dsRNA induces retinoic acid synthesis to stimulate hair follicle regeneration via TLR3. Nat Commun 2019;10:2811.

108. Gudas LJ. Retinoid metabolism: new insights. J Mol Endocrinol 2022;69:T37-49.

109. Stresser DM, Kopec AK, Hewitt P, et al. Towards in vitro models for reducing or replacing the use of animals in drug testing. Nat Biomed Eng 2024;8:930-5.

110. Cobbina E, Akhlaghi F. Non-alcoholic fatty liver disease (NAFLD) - pathogenesis, classification, and effect on drug metabolizing enzymes and transporters. Drug Metab Rev 2017;49:197-211.

111. Bhutani P, Joshi G, Raja N, et al. U.S. FDA approved drugs from 2015-June 2020: a perspective. J Med Chem 2021;64:2339-81.

112. Sumantran VN, Mishra P, Bera R, Sudhakar N. Microarray analysis of differentially-expressed genes encoding CYPs and phase II drug metabolizing enzymes in psoriasis and melanoma. Pharmaceutics 2016;8:4.

113. Perihan O, Ergul KB, Neslihan D, Filiz A. The activity of adenosine deaminase and oxidative stress biomarkers in scraping samples of acne lesions. J Cosmet Dermatol 2012;11:323-8.

114. Visconti B, Paolino G, Carotti S, et al. Immunohistochemical expression of VDR is associated with reduced integrity of tight junction complex in psoriatic skin. J Eur Acad Dermatol Venereol 2015;29:2038-42.

115. Ortiz-Lopez LI, Choudhary V, Bollag WB. Updated perspectives on keratinocytes and psoriasis: keratinocytes are more than innocent bystanders. Psoriasis (Auckl) 2022;12:73-87.

116. Danso M, Boiten W, van Drongelen V, et al. Altered expression of epidermal lipid bio-synthesis enzymes in atopic dermatitis skin is accompanied by changes in stratum corneum lipid composition. J Dermatol Sci 2017;88:57-66.

117. Boniface K, Seneschal J, Picardo M, Taïeb A. Vitiligo: focus on clinical aspects, immunopathogenesis, and therapy. Clin Rev Allergy Immunol 2018;54:52-67.

Cite This Article

Review
Open Access
Role of skin enzymes in metabolism of topical drugs
Nikki Lau, ... Yousuf MohammedYousuf Mohammed

How to Cite

Lau, N.; Phan K.; Mohammed Y. Role of skin enzymes in metabolism of topical drugs. Metab. Target. Organ. Damage. 2024, 4, 32. http://dx.doi.org/10.20517/mtod.2024.17

Download Citation

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click on download.

Export Citation File:

Type of Import

Tips on Downloading Citation

This feature enables you to download the bibliographic information (also called citation data, header data, or metadata) for the articles on our site.

Citation Manager File Format

Use the radio buttons to choose how to format the bibliographic data you're harvesting. Several citation manager formats are available, including EndNote and BibTex.

Type of Import

If you have citation management software installed on your computer your Web browser should be able to import metadata directly into your reference database.

Direct Import: When the Direct Import option is selected (the default state), a dialogue box will give you the option to Save or Open the downloaded citation data. Choosing Open will either launch your citation manager or give you a choice of applications with which to use the metadata. The Save option saves the file locally for later use.

Indirect Import: When the Indirect Import option is selected, the metadata is displayed and may be copied and pasted as needed.

About This Article

© The Author(s) 2024. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
158
Downloads
15
Citations
0
Citations
0
Comments
0
0

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Metabolism and Target Organ Damage
ISSN 2769-6375 (Online)

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/