Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Clin Exp Allergy ; 49(1): 68-81, 2019 01.
Article in English | MEDLINE | ID: mdl-30288827

ABSTRACT

BACKGROUND: Atopic dermatitis (AD) represents the most common inflammatory skin disorder in children showing massive infiltration of immune cells. The colonization of AD-afflicted skin by Staphylococcus aureus and S.Ā aureus-derived extracellular vesicles (SEVs) has been associated with AD pathogenesis; however, the molecular mechanism underlying SEV-mediated inflammatory responses remains unclear. OBJECTIVE: We investigated how SEVs can mediate inflammatory responses in AD pathogenesis by examining the effect of SEVs on human dermal microvascular endothelia cells (HDMECs). METHODS: HDMECs were treated with SEVs, and the expression of cell adhesion molecules or cytokines was assessed using RT-qPCR, Western blot or cytokine array analyses. The receptor for SEVs and related signalling molecules in HDMECs were addressed and verified via gene knockdown or inhibitor experiments. The recruitment assay of human THP-1 monocytic cells on HDMECs was performed after SEV treatment in the presence or absence of the verified receptor or signalling molecule. RESULTS: SEVs, but not other gram-positive bacteria-derived extracellular vesicles, directly activated HDMECs by increasing the expression of cell adhesion molecules (E-selectin, VCAM1 and ICAM1) and that of IL-6, the inflammatory cytokine; consequently, they enhanced the recruitment of THP-1 monocytic cells to HDMECs. The SEV-induced HDMEC activation was dependent on Toll-like receptor 4 and the NF-κB signalling pathway, which was rapidly activated within 1Ā hour post-treatment and followed by an upregulation of cell adhesion molecules and IL-6 at later time-points. Moreover, SEV-mediated HDMEC responses were more rapid and intense than those induced by the same protein concentrations of S.Ā aureus extracts. CONCLUSIONS & CLINICAL RELEVANCE: SEVs as proinflammatory factors could mediate immune cell infiltration in AD by efficiently inducing endothelial cell activation and monocyte recruitment, which may provide insights into alleviating the S.Ā aureus-mediated onset or progression of AD and its phenotypes.


Subject(s)
Dermatitis, Atopic/immunology , Dermis/immunology , Endothelial Cells/immunology , Extracellular Vesicles/immunology , Microvessels/immunology , Monocytes/immunology , Staphylococcus aureus/immunology , Cell Line , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/pathology , Dermis/pathology , Endothelial Cells/pathology , Humans , Microvessels/pathology , Monocytes/pathology
2.
Exp Dermatol ; 28(6): 738-741, 2019 06.
Article in English | MEDLINE | ID: mdl-30408247

ABSTRACT

Hyperpigmentation is caused by excessive production of melanin in melanocytes. Mannosylerythritol lipids (MELs) are glycolipid biosurfactants that are abundantly produced by yeasts and used commercially in cosmetics. However, the potential depigmenting efficacy of MELs has not been evaluated. In this study, the depigmentary effect of MELs was tested in primary normal human melanocytes (NHMs), α-melanocyte-stimulating hormone (MSH)-stimulated B16 cells (murine melanoma cells) and a human skin equivalent (MelanoDerm) using photography, Fontana-Masson (F&M) staining and two-photon microscopy. Mannosylerythritol lipids significantly decreased the melanin contents in NHMs and α-MSH-stimulated B16 cells. Consistent with these findings, MELs treatment had a clear whitening effect in a human skin equivalent, brightening the tissue colour and reducing the melanin content. The molecular mechanism underlying the anti-melanogenic effect of MELs treatment was examined by real-time PCR and Western blotting. Mechanistically, MELs clearly suppressed the gene expression levels of representative melanogenic enzymes, including tyrosinase, Tyrp-1 and Tyrp-2, by inhibiting the ERK/CREB/MiTF signalling pathway in NHMs. This work demonstrates for the first time that MELs exert whitening effects on human melanocytes and skin equivalent. Thus, we suggest that MELs could be developed as a potent anti-melanogenic agent for effective whitening, beyond their use as a biosurfactant in cosmetics.


Subject(s)
Glycolipids/pharmacology , Hyperpigmentation/drug therapy , MAP Kinase Signaling System/drug effects , Melanocytes/drug effects , Animals , Cell Line , Drug Evaluation, Preclinical , Glycolipids/therapeutic use , Humans , Melanins/biosynthesis , Melanocytes/metabolism , Mice , Primary Cell Culture
3.
FASEB J ; 32(3): 1510-1523, 2018 03.
Article in English | MEDLINE | ID: mdl-29180440

ABSTRACT

As the outermost physical barrier of an organism, the skin is diurnally exposed to UV radiation (UVR). Recent studies have revealed that the skin exhibits a circadian rhythm in various functions, and this oscillation is disturbed and reset via a strong environmental cue, the UVR. However, a molecular link between circadian perturbation by UVR and UVR-induced cellular responses has not been investigated. We identified tissue inhibitor of metalloproteinase ( TIMP)- 3 as a novel circadian locomotor output cycles kaput (CLOCK)-dependent diurnal gene by using a CLOCK-knockdown strategy in human keratinocytes. Among dozens of identified transcripts down-regulated by CLOCK knockdown, TIMP3 displayed a rhythmic expression in a CLOCK-dependent manner, in which the expression of matrix metalloproteinase (MMP)-1 and inflammatory cytokines, such as TNF-α, chemokine (C-X-C motif) ligand (CXCL)-1, and IL-8, were inversely regulated. Upon UVB exposure, the expression of CLOCK and TIMP3 was down-regulated, which led to an up-regulation of secretion of MMP1 and TNF-α proteins and in the transcription of CXCL1 and IL-8 via CCAAT-enhancer binding protein (C/EBP)-α. UVB-induced TNF-α secretion increased further or decreased by knockdown or overexpression of TIMP3, respectively, as well as by CLOCK. As a novel CLOCK-dependent diurnal gene, TIMP3 inhibits the expression of inflammatory cytokines that are up-regulated by UV irradiation in human keratinocytes. Thus, our work suggests a molecular link between circadian perturbation by UVR and UVR-induced inflammation.-Park, S., Kim, K., Bae, I.-H., Lee, S. H., Jung, J., Lee, T. R., Cho, E.-G. TIMP3 is a CLOCK-dependent diurnal gene that inhibits the expression of UVB-induced inflammatory cytokines in human keratinocytes.


Subject(s)
CLOCK Proteins/metabolism , Cytokines/biosynthesis , Gene Expression Regulation/radiation effects , Keratinocytes/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , Ultraviolet Rays/adverse effects , CLOCK Proteins/genetics , Cytokines/genetics , Humans , Keratinocytes/pathology , Tissue Inhibitor of Metalloproteinase-3/genetics
4.
Skin Pharmacol Physiol ; 32(2): 109-116, 2019.
Article in English | MEDLINE | ID: mdl-30840954

ABSTRACT

BACKGROUND/AIMS: Dehydroabietic acid (DAA) is a natural phytochemical found in red pine trees and herbal plants. While DAA and its derivatives are known for improving diabetes and hyperlipidemia, the antiaging effect and its underlying mechanisms of DAA on skin have not been fully examined. Here, we assessed the antiaging effects of DAA on human dermal fibroblasts and skin equivalents. METHODS: We investigated the effect of DAA on the secretion of type I procollagen and matrix metalloproteinase-1 (MMP-1) in ultraviolet B (UVB)-irradiated neonatal normal human dermal fibroblasts (NHDFn). Using nonlinear optical imaging techniques, we visualized quantitative and qualitative changes of collagen fibers by DAA treatment in human skin equivalent models. RESULTS: DAA induces increases in type I procollagen secretion when treated on UVB-irradiated NHDFn. DAA also downregulates secretion of MMP-1 through the inhibition of the JNK signaling pathway. In human skin equivalent models, we successfully visualized the spatial distribution of collagen fibers in the dermis and found that quantity, diameter, and arrangement of collagen fibers in the dermis were significantly improved by DAA treatment. CONCLUSION: Our results suggest that DAA could be a useful agent for improving skin photoaging through the protection and regeneration of collagen fibers in skin.


Subject(s)
Abietanes/pharmacology , Collagen/metabolism , Fibroblasts/drug effects , Radiation-Protective Agents/pharmacology , Skin/drug effects , Ultraviolet Rays/adverse effects , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/radiation effects , Humans , In Vitro Techniques , Matrix Metalloproteinase 1/metabolism , Skin/cytology , Skin/metabolism , Skin/radiation effects , Skin Aging
5.
Int J Mol Sci ; 20(7)2019 Mar 29.
Article in English | MEDLINE | ID: mdl-30934981

ABSTRACT

Dehydroabietic acid (DAA) is a naturally occurring diterpene resin acid derived from coniferous plants such as Pinus and Picea. Various bioactive effects of DAA have been studied including antibacterial, antifungal, and anticancer activities. However, the anti-inflammatory mechanism of DAA remains unclear. We evaluated the anti-inflammatory effect of DAA in macrophage cell lines. Dehydroabietic acid clearly reduced nitric oxide (NO) production and inflammatory gene expression decreased according to RT-PCR results. Dehydroabietic acid displayed anti-inflammatory activity at the transcriptional level in results from NF-κB- or AP-1-mediated luciferase assays. To identify the DAA target protein, we investigated NF-κB and AP-1 pathways by Western blotting analysis. Dehydroabietic acid suppressed the activity of proto-oncogene tyrosine protein kinase (Src) and spleen tyrosine kinase (Syk) in the NF-κB cascade and transforming growth factor beta-activated kinase 1 (TAK1) in the AP-1 cascade. Using overexpression strategies, we confirmed that DAA targeted these kinases. Our findings demonstrate the anti-inflammatory effects and molecular mechanism of DAA. This suggests that DAA has potential as a drug or supplement to ameliorate inflammation.


Subject(s)
Abietanes/pharmacology , Inflammation/pathology , MAP Kinase Kinase Kinases/metabolism , Signal Transduction , Syk Kinase/metabolism , src-Family Kinases/metabolism , Abietanes/chemistry , Animals , Anti-Inflammatory Agents/pharmacology , Cell Death/drug effects , Mice , Models, Biological , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , RAW 264.7 Cells , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism , Transcription, Genetic/drug effects
6.
Korean J Physiol Pharmacol ; 23(2): 113-120, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30820155

ABSTRACT

Mannosylerythritol lipids (MELs) are glycolipids and have several pharmacological efficacies. MELs also show skin-moisturizing efficacy through a yet-unknown underlying mechanism. Aquaporin-3 (AQP3) is a membrane protein that contributes to the water homeostasis of the epidermis, and decreased AQP3 expression following ultraviolet (UV)-irradiation of the skin is associated with reduced skin moisture. No previous study has examined whether the skin-moisturizing effect of MELs might act through the modulation of AQP3 expression. Here, we report for the first time that MELs ameliorate the UVA-induced downregulation of AQP3 in cultured human epidermal keratinocytes (HaCaT keratinocytes). Our results revealed that UVA irradiation decreases AQP3 expression at the protein and messenger RNA (mRNA) levels, but that MEL treatment significantly ameliorated these effects. Our mitogen-activated protein kinase inhibitor analysis revealed that phosphorylation of c-Jun N-terminal kinase (JNK), but not extracellular signal-regulated kinase or p38, mediates UVA-induced AQP3 downregulation, and that MEL treatment significantly suppressed the UVA-induced phosphorylation of JNK. To explore a possible mechanism, we tested whether MELs could regulate the expression of peroxidase proliferator-activated receptor gamma (PPAR-ƎĀ³), which acts as a potent transcription factor for AQP3 expression. Interestingly, UVA irradiation significantly inhibited the mRNA expression of PPAR-ƎĀ³ in HaCaT keratinocytes, whereas a JNK inhibitor and MELs significantly rescued this effect. Taken together, these findings suggest that MELs ameliorate UVA-induced AQP3 downregulation in HaCaT keratinocytes by suppressing JNK activation to block the decrease of PPAR-ƎĀ³. Collectively, our findings suggest that MELs can be used as a potential ingredient that modulates AQP3 expression to improve skin moisturization following UVA irradiation-induced damage.

7.
Cytokine ; 110: 126-130, 2018 10.
Article in English | MEDLINE | ID: mdl-29730385

ABSTRACT

Melanin synthesis in melanocytes is affected by various cytokines. Here, we reported for the first time that tumor necrosis factor superfamily member 14 (TNFSF14) inhibits melanogenesis in the primary culture of human epidermal melanocytes. TNFSF14 is known to bind to its receptors herpes virus entry mediator (HVEM) and lymphotoxin Ɵ receptor (LTƟR) for signal transduction, but TNFSF14-induced hypopigmentation was independent of HVEM and LTƟR in melanocytes. To explore signaling in melanocytes treated with TNFSF14, we performed RNA-seq and found that nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) signaling is activated by TNFSF14. Further, we observed that inhibition of NF-kB effectively blocks the hypopigmentation induced by TNFSF14. We conclude that TNFSF14 inhibits melanogenesis in melanocytes via NF-κB signaling and could be applied in the treatment of cutaneous pigment disorders.


Subject(s)
Melanocytes/metabolism , NF-kappa B/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Cell Line , Humans , Lymphocyte Activation/physiology , Lymphotoxin beta Receptor/metabolism , Melanins/metabolism , Receptors, Tumor Necrosis Factor, Member 14/metabolism
8.
Toxicol Appl Pharmacol ; 338: 174-181, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29183759

ABSTRACT

Benzophenone-3 (BP-3), which is extensively used in organic sunscreen, has phototoxic potential in human skin. Phosphodiesterase 4B (PDE4B) has a well-established role in inflammatory responses in immune cells. Currently, it is unknown if PDE4B is associated with BP-3-induced phototoxicity in normal human keratinocytes (NHKs). We found that BP-3 significantly increased PDE4B expression in ultraviolet B (UVB)-irradiated NHKs. Notably, BP-8, a sunscreen agent that shares the 2-hydroxy-4-methoxyphenyl methanone moiety with BP-3, also upregulated PDE4B expression in NHKs. Upon UVB irradiation, BP-3 upregulated the expression of pro-inflammatory factors, such as prostaglandin endoperoxide synthase 2, tumor necrosis factor α, interleukin 8, and S100A7, and downregulated the level of cornified envelope associated proteins, which are important in the development of the epidermal permeability barrier. The additive effects of UVB-activated BP-3 on the expression of both pro-inflammatory mediators and cornified envelope associated proteins were antagonized by treatment with the PDE4 inhibitor rolipram. The BP-3 and UVB co-stimulation-induced PDE4B upregulation and its association with the upregulation of pro-inflammatory mediators and the downregulation of epidermal differentiation markers were confirmed in a reconstituted three dimensional human epidermis model. Therefore, PDE4B has a role in the mechanism of BP-3-induced phototoxicity.


Subject(s)
Benzophenones/toxicity , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Dermatitis, Phototoxic/etiology , Keratinocytes/drug effects , Cyclic AMP/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Dinoprostone/biosynthesis , Humans , Interleukin-8/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Ultraviolet Rays
9.
Exp Dermatol ; 27(5): 453-459, 2018 05.
Article in English | MEDLINE | ID: mdl-28453913

ABSTRACT

This study reports a three-dimensional (3D) bioprinting technique that is capable of producing a full-thickness skin model containing pigmentation. Multiple layers of fibroblast (FB)-containing collagen hydrogel precursor were printed and crosslinked through neutralization using sodium bicarbonate, constituting the dermal layer. Melanocytes (MCs) and keratinocytes (KCs) were sequentially printed on top of the dermal layer to induce skin pigmentation upon subsequent air-liquid interface culture. Histological analysis was performed not only to confirm the formation of distinct skin layers, but also to identify the presence of pigmentation. The bioprinted skin structure showed the dermal and epidermal layers as well as the terminal differentiation of the KC that formed the stratum corneum. Moreover, the MC-containing epidermal layer showed freckle-like pigmentations at the dermal-epidermal junction, without the use of external ultraviolet light or chemical stimuli. The presented method offers the capability of producing engineered ephelides in biomimetic skin, thus rendering 3D bioprinting techniques as productive on-demand options for the creation of skin models available for therapeutic or research use.


Subject(s)
Bioprinting , Melanocytes , Printing, Three-Dimensional , Skin Pigmentation , Skin , Cells, Cultured , Humans
10.
Exp Dermatol ; 27(10): 1126-1133, 2018 10.
Article in English | MEDLINE | ID: mdl-30044012

ABSTRACT

The receptor-interacting protein kinase 4 (RIP4), a serine/threonine kinase, is an important modulator of epidermal growth and cutaneous inflammation. We found that RIP4 expression was significantly increased in the lesional skin of psoriasis. However, the role and regulatory mechanism of RIP4 in psoriasis have not been characterized. After treatment with IL-17, RIP4 mRNA and protein levels were increased in HaCaT cells. IL-17 also activated the RIP4 promoter. To understand the functional role of RIP4 in keratinocyte and to investigate the genes regulated by RIP4, RNA-based microarray analysis was performed. Among immune response-related genes, CCL20 expression was significantly changed by RIP4. To identify RIP4-interacting protein, an immunoprecipitation assay was performed. As a result, STAT3 was identified as a new protein that interacts with RIP4. The interaction of RIP4 and STAT3 enhanced STAT3 phosphorylation. In addition, the transcriptional activity of STAT3 induced by RIP4 regulated IL-17-mediated CCL20 expression in HaCaT cells. Taken together, these findings indicate that IL-17 increased RIP4-mediated STAT3 phosphorylation by directly interacting with STAT3. Thus, transcriptional activation of STAT3 promotes the expression of CCL20. Thus, activations of these signalling pathways by RIP4 may contribute to epithermal inflammation in psoriatic keratinocytes.


Subject(s)
Chemokine CCL20/genetics , Interleukin-17/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Psoriasis/genetics , STAT3 Transcription Factor/metabolism , Adult , HEK293 Cells , Humans , Keratinocytes , Oligonucleotide Array Sequence Analysis , Phosphorylation , Promoter Regions, Genetic/drug effects , Psoriasis/metabolism , Psoriasis/pathology , RNA, Messenger/analysis , RNA, Messenger/metabolism , STAT3 Transcription Factor/genetics , Transcription, Genetic , Up-Regulation/drug effects
11.
Exp Dermatol ; 27(11): 1287-1293, 2018 11.
Article in English | MEDLINE | ID: mdl-30230035

ABSTRACT

Hypoxia-inducible factor-1α (HIF-1α) has been reported to be up-regulated in psoriatic epidermis, resulting in increased proliferation and abnormal differentiation of human keratinocytes (KCs). However, the role of HIF-1α in psoriatic epidermis, which is mainly composed of KCs, is poorly understood. Here, we show that morphogenic protein 6 (BMP6) is down-regulated when HIF-1α is upregulated in patients with psoriasis skin lesions. HIF-1α overexpression in primary human KCs promoted proliferation and inhibited terminal differentiation. Furthermore, HIF1-α repressed the expression of BMP6 by binding directly to the hypoxia-response element (HRE) in the BMP6 promotor region, which shows that BMP6 is a novel target gene of HIF-1α. We also found that HIF-1α-mediated BMP6 suppression could alter the proliferation status by modulating the expression levels of cell cycle regulatory proteins and also affect the early differentiation of KCs. Therefore, we suggest that HIF-1α-dependent BMP6 suppression has a critical role in the induction of hyper-proliferation and abnormal differentiation in psoriatic KCs.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Psoriasis/genetics , Antigens, Neoplasm/metabolism , Bone Morphogenetic Protein 6/pharmacology , Carbonic Anhydrase IX/metabolism , Cell Cycle/genetics , Cell Differentiation/drug effects , Cell Hypoxia , Cell Proliferation/drug effects , Cells, Cultured , Down-Regulation , Glucose Transporter Type 1/metabolism , Humans , Keratinocytes/physiology , Primary Cell Culture , Promoter Regions, Genetic , Psoriasis/metabolism , Transfection
12.
Int J Mol Sci ; 19(7)2018 06 21.
Article in English | MEDLINE | ID: mdl-29933606

ABSTRACT

8-Hydroxydaidzein (8-HD) is a daidzein metabolite isolated from soybeans. This compound has been studied for its anti-proliferation, depigmentation, and antioxidant activities. However, the anti-inflammatory activities of 8-HD are not well-understood. Through its antioxidant effects in ABTS and DPPH assays, 8-HD reduces the production of sodium nitroprusside (SNP)-induced radical oxygen species (ROS). By triggering various Toll-like receptors (TLRs), 8-HD suppresses the inflammatory mediator nitric oxide (NO) without cytotoxicity. We examined the regulatory mechanism of 8-HD in lipopolysaccharide (LPS)-induced conditions. We found that 8-HD diminishes inflammatory gene expression (e.g., inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2, and tumor necrosis factor (TNF)-α) by regulating the transcriptional activities of nuclear factor (NF)-κB and activator protein 1 (AP-1). To find the potential targets of 8-HD, signaling pathways were investigated by immunoblotting analyses. These analyses revealed that 8-HD inhibits the activation of TAK1 and that phosphorylated levels of downstream molecules decrease in sequence. Together, our results demonstrate the antioxidant and anti-inflammatory actions of 8-HD and suggest its potential use in cosmetics or anti-inflammatory drugs.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Gene Expression Regulation/drug effects , Isoflavones/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/isolation & purification , Antioxidants/isolation & purification , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Isoflavones/isolation & purification , Lipopolysaccharides/pharmacology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/immunology , Macrophage Activation/drug effects , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/immunology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , RAW 264.7 Cells , Signal Transduction , Glycine max/chemistry , Transcription Factor AP-1/genetics , Transcription Factor AP-1/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
13.
Nucleic Acids Res ; 43(15): 7462-79, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26202967

ABSTRACT

ΔNp63 is required for both the proliferation and differentiation of keratinocytes, but its role in the differentiation of these cells is poorly understood. The corresponding gene, TP63, harbors the MIR944 sequence within its intron. However, the mechanism of biogenesis and the function of miR-944 are unknown. We found that miR-944 is highly expressed in keratinocytes, in a manner that is concordant with that of ΔNp63 mRNA, but the regulation of miR-944 expression under various conditions did not correspond with that of ΔNp63. Bioinformatics analysis and functional studies demonstrated that MIR944 has its own promoter. We demonstrate here that MIR944 is a target of ΔNp63. Promoter analysis revealed that the activity of the MIR944 promoter was markedly enhanced by the binding of ΔNp63, which was maintained by the supportive action of AP-2 during keratinocyte differentiation. Our results indicated that miR-944 biogenesis is dependent on ΔNp63 protein, even though it is generated from ΔNp63 mRNA-independent transcripts. We also demonstrated that miR-944 induces keratin 1 and keratin 10 expression by inhibiting ERK signaling and upregulating p53 expression. Our findings suggested that miR-944, as an intronic miRNA and a direct target of ΔNp63, contributes to the function of ΔNp63 in the induction of epidermal differentiation.


Subject(s)
Cell Differentiation/genetics , Epidermal Cells , MicroRNAs/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Cells, Cultured , Gene Expression Regulation , Humans , Introns , Keratinocytes/metabolism , MicroRNAs/biosynthesis , MicroRNAs/genetics , Promoter Regions, Genetic
14.
J Lipid Res ; 57(4): 563-73, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26658689

ABSTRACT

Lipin-1 is an Mg(2+)-dependent phosphatidate phosphatase that facilitates the dephosphorylation of phosphatidic acid to generate diacylglycerol. Little is known about the expression and function of lipin-1 in normal human epidermal keratinocytes (NHEKs). Here, we demonstrate that lipin-1 is present in basal and spinous layers of the normal human epidermis, and lipin-1 expression is gradually downregulated during NHEK differentiation. Interestingly, lipin-1 knockdown (KD) inhibited keratinocyte differentiation and caused G1 arrest by upregulating p21 expression. Cell cycle arrest by p21 is required for commitment of keratinocytes to differentiation, but must be downregulated for the progress of keratinocyte differentiation. Therefore, reduced keratinocyte differentiation results from sustained upregulation of p21 by lipin-1 KD. Lipin-1 KD also decreased the phosphorylation/activation of protein kinase C (PKC)α, whereas lipin-1 overexpression increased PKCα phosphorylation. Treatment with PKCα inhibitors, like lipin-1 KD, stimulated p21 expression, while lipin-1 overexpression reduced p21 expression, implicating PKCα in lipin-1-induced regulation of p21 expression. Taken together, these results suggest that lipin-1-mediated downregulation of p21 is critical for the progress of keratinocyte differentiation after the initial commitment of keratinocytes to differentiation induced by p21, and that PKCα is involved in p21 expression regulation by lipin-1.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Enzymologic , Keratinocytes/cytology , Phosphatidate Phosphatase/genetics , Phosphatidate Phosphatase/metabolism , Cell Cycle , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Diglycerides/metabolism , Down-Regulation , Enzyme Activation , Epidermal Cells , Gene Knockdown Techniques , Humans , Infant, Newborn , Phosphatidate Phosphatase/deficiency , Protein Kinase C-alpha/metabolism , Tumor Suppressor Protein p53/metabolism
15.
J Lipid Res ; 57(4): 607-15, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26851298

ABSTRACT

NO regulates a variety of physiological processes, including cell proliferation, differentiation, and inflammation. S-nitrosylation, a NO-mediated reversible protein modification, leads to changes in the activity and function of proteins. In particular, the role of S-nitrosylation during adipogenesis is largely unknown. We hypothesized that the normal physiological levels of NO, but not the excess levels generated under severe conditions, such as inflammation, may be critically involved in the proper regulation of adipogenesis. We found that endogenous S-nitrosylation of proteins was required for adipocyte differentiation. By performing a biotin-switch assay, we identified FAS, a key lipogenic enzyme in adipocytes, as a target of S-nitrosylation during adipogenesis. Interestingly, we also observed that the dimerization of FAS increased in parallel with the amount of S-nitrosylated FAS during adipogenesis. In addition, we found that exogenous NO enhanced the dimerization and the enzymatic activity of FAS. Moreover, site-directed mutagenesis of three predicted S-nitrosylation sites indicated that S-nitrosylation of FAS at Cys(1471)and Cys(2091), but not at Cys(1127), increased its enzymatic activity. Taken together, these results suggest that the S-nitrosylation of FAS at normal physiological levels of NO increases its activity through dimerization and may contribute to the proper regulation of adipogenesis.


Subject(s)
Fatty Acid Synthases/chemistry , Fatty Acid Synthases/metabolism , Nitric Oxide/metabolism , Protein Multimerization , Protein Processing, Post-Translational , Adipocytes/cytology , Adipogenesis , Cysteine/metabolism , Enzyme Activation , HEK293 Cells , Humans , Protein Structure, Quaternary , Sulfur/metabolism
16.
Biochem Biophys Res Commun ; 469(2): 251-6, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26646290

ABSTRACT

Atmospheric oxygen is important for the epidermis, as the skin epidermis is not greatly affected by blood circulation. Therefore, it is necessary to understand the effect of hypoxic signals on the epidermis as some environmental stimuli can induce skin hypoxia. Here, we investigated how hypoxia (1% O2) affected skin equivalents (SEs) and normal human epidermal keratinocytes. We found that hypoxia specifically decreased the protein levels of keratin 1 (K1)/keratin 10 (K10), a representative marker of the epidermal spinous layer in the epidermis. However, hypoxia-inducible factors, the major regulators of hypoxia, did not affect hypoxia-induced down-regulation of K1/K10. We also found that N-acetyl-l-cysteine (NAC), a reactive oxygen species scavenger, antagonized the hypoxia-induced reduction of K1/K10 in keratinocytes and SEs. In contrast to the findings for NAC, inhibitors that blocked reactive oxygen species generation did not cause recovery of K1/K10 protein levels under hypoxic conditions. Taken together, these results indicate that hypoxia leads to abnormal keratinocyte differentiation by down-regulating K1/K10 and that this phenomenon can be ameliorated by NAC.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Keratins, Type II/metabolism , Oxygen/metabolism , Signal Transduction/physiology , Cell Differentiation/physiology , Cell Hypoxia/physiology , Cells, Cultured , Humans
17.
Exp Dermatol ; 25(8): 636-41, 2016 08.
Article in English | MEDLINE | ID: mdl-27060579

ABSTRACT

Psoriasin (S100A7), a member of the S100 protein family, is a well-known antimicrobial peptide and a signalling molecule which regulates cellular function and is highly expressed in hyperproliferative skin conditions such as atopic dermatitis (AD) and psoriasis with disrupted skin barrier function. However, its role in epidermal differentiation remains unknown. We examined the effect of S100A7 on epidermal differentiation in normal human keratinocytes (NHKs) and on a reconstituted human epidermis model. When NHKs were exposed to disruptive stimuli such as Staphylococcus aureus, ultraviolet irradiation and retinoic acid, the secretion of S100A7 into the culture medium increased and the expression of epidermal differentiation markers decreased. Treatment of NHKs with S100A7 significantly inhibited epidermal differentiation by reducing the expression of keratin 1, keratin 10, involucrin and loricrin and by increasing the expression of abnormal differentiation markers (keratin 6 and keratin 16). We verified that the MyD88-IκB/NF-κB signal cascade was activated via RAGE after S100A7 treatment, resulting in the upregulation of interleukin-6. Finally, we confirmed that S100A7 is a negative regulator of epidermal differentiation using a reconstituted human epidermis model. This study suggests that S100A7-related signalling molecules could be potent targets for recovering skin barrier function in AD and psoriasis where S100A7 is accumulated excessively.


Subject(s)
Cell Differentiation , Epidermis/metabolism , Interleukin-6/metabolism , Keratinocytes/metabolism , S100 Calcium Binding Protein A7/metabolism , Cells, Cultured , Epidermal Cells , Humans , Keratinocytes/cytology , Signal Transduction , Stress, Physiological
18.
Acta Derm Venereol ; 96(5): 635-40, 2016 Jun 15.
Article in English | MEDLINE | ID: mdl-26671310

ABSTRACT

Basement membrane (BM) disruption and dermal changes (elastosis, collagenolysis, vascular ectasia) have been reported in melasma. Although ultraviolet (UV) irradiation can induce these changes, UV is not always necessary for melasma development. Cadherin 11 (CDH11), which is upregulated in some melasma patients, has previously been shown to stimulate melanogenesis. Because CDH11 action requires cell-cell adhesion between fibroblasts and melanocytes, BM disruption in vivo should facilitate this. The aim of this study was to examine whether CDH11 overexpression leads to BM disruption and dermal changes, independent of UV irradiation. Immunohistochemistry/immunofluorescence, real-time PCR, Western blotting, and zymography suggested that BM disruption/dermal changes and related factors were present in the hyperpigmented skin of CDH11-upregulated melasma patients and in CDH11-overexpressing fibroblasts/keratinocytes. The opposite was seen in CDH11-knockdown cells. UV irradiation of the cultured cells did not increase CDH11 expression. Collectively, these data demonstrate that CDH11 overexpression could induce BM disruption and dermal changes in melasma, regardless of UV exposure.


Subject(s)
Basement Membrane/metabolism , Basement Membrane/pathology , Cadherins/metabolism , Melanosis/metabolism , Melanosis/pathology , Blotting, Western , Cells, Cultured , Female , Fibroblasts/metabolism , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Keratinocytes/metabolism , Melanocytes/metabolism , Real-Time Polymerase Chain Reaction , Up-Regulation
19.
Int J Mol Sci ; 17(4): 583, 2016 Apr 16.
Article in English | MEDLINE | ID: mdl-27092497

ABSTRACT

The regulation of melanin production is important for managing skin darkness and hyperpigmentary disorders. Numerous anti-melanogenic agents that target tyrosinase activity/stability, melanosome maturation/transfer, or melanogenesis-related signaling pathways have been developed. As a rate-limiting enzyme in melanogenesis, tyrosinase has been the most attractive target, but tyrosinase-targeted treatments still pose serious potential risks, indicating the necessity of developing lower-risk anti-melanogenic agents. Sugars are ubiquitous natural compounds found in humans and other organisms. Here, we review the recent advances in research on the roles of sugars and sugar-related agents in melanogenesis and in the development of sugar-based anti-melanogenic agents. The proposed mechanisms of action of these agents include: (a) (natural sugars) disturbing proper melanosome maturation by inducing osmotic stress and inhibiting the PI3 kinase pathway and (b) (sugar derivatives) inhibiting tyrosinase maturation by blocking N-glycosylation. Finally, we propose an alternative strategy for developing anti-melanogenic sugars that theoretically reduce melanosomal pH by inhibiting a sucrose transporter and reduce tyrosinase activity by inhibiting copper incorporation into an active site. These studies provide evidence of the utility of sugar-based anti-melanogenic agents in managing skin darkness and curing pigmentary disorders and suggest a future direction for the development of physiologically favorable anti-melanogenic agents.


Subject(s)
Carbohydrates/chemistry , Carbohydrates/pharmacology , Melanins/antagonists & inhibitors , Melanins/metabolism , Skin Pigmentation/drug effects , Animals , Antigens, Neoplasm/metabolism , Humans , Membrane Transport Proteins/metabolism
20.
J Biol Chem ; 289(42): 28835-45, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25164816

ABSTRACT

Testosterone is an endocrine hormone with functions in reproductive organs, anabolic events, and skin homeostasis. We report here that GPRC6A serves as a sensor and mediator of the rapid action of testosterone in epidermal keratinocytes. The silencing of GPRC6A inhibited testosterone-induced intracellular calcium ([Ca(2+)]i) mobilization and H2O2 generation. These results indicated that a testosterone-GPRC6A complex is required for activation of Gq protein, IP3 generation, and [Ca(2+)]i mobilization, leading to Duox1 activation. H2O2 generation by testosterone stimulated the apoptosis of keratinocytes through the activation of caspase-3. The application of testosterone into three-dimensional skin equivalents increased the apoptosis of keratinocytes between the granular and stratified corneum layers. These results support an understanding of the molecular mechanism of testosterone-dependent apoptosis in which testosterone stimulates H2O2 generation through the activation of Duox1.


Subject(s)
Gene Expression Regulation, Enzymologic , Keratinocytes/metabolism , NADPH Oxidases/metabolism , Receptors, G-Protein-Coupled/metabolism , Skin/metabolism , Testosterone/chemistry , Apoptosis , Calcium/metabolism , Caspase 3/metabolism , Cell Line , Dual Oxidases , Gene Expression Profiling , Humans , Hydrogen Peroxide/metabolism , Membrane Potential, Mitochondrial , Oxidation-Reduction , Signal Transduction , Skin/cytology , Skin/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL