Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Exp Dermatol ; 26(8): 744-747, 2017 08.
Article in English | MEDLINE | ID: mdl-27943416

ABSTRACT

Exposure of the skin to ultraviolet (UV) irradiation causes various consequences such as inflammation and photoageing. Galanin is an active neuropeptide expressed widely in the central nervous system and peripheral tissues including the skin. Galanin promotes or inhibits inflammation in a context-dependent manner, but its role in UV irradiation-induced responses in human skin was still unknown. UV irradiation induced a substantial expression of galanin in primary epidermal keratinocytes in vitro and in human epidermis in vivo. Galanin knock-down by siRNA transfection markedly inhibited UV irradiation-induced expression of matrix metalloproteinase (MMP)-1, interleukin (IL)-1ß, IL-6 and cyclooxygenase (COX)-2. Moreover, siRNA-mediated knock-down of GAL2 , a principal galanin receptor in the skin, led to a considerable decrease in these mediators in keratinocytes. Collectively, our findings suggest that galanin is an important messenger between the neuroendocrine system and UV irradiation-damaged skin.


Subject(s)
Epidermis/radiation effects , Galanin/metabolism , Keratinocytes/drug effects , Radiodermatitis/metabolism , Epidermis/metabolism , Humans , Keratinocytes/metabolism , Ultraviolet Rays
2.
Biochem Biophys Res Commun ; 477(3): 336-42, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27343558

ABSTRACT

Vasoactive intestinal peptide (VIP), one of the major skin neuropeptides, has been suggested to have active roles in the pathogenesis of inflammatory skin disorders such as atopic dermatitis and psoriasis, which can commonly cause post-inflammatory hyperpigmentation. However, the effect of VIP on melanogenesis remains unknown. In this study, we showed that the melanin contents, tyrosinase activity, and gene expression of tyrosinase and microphthalmia-associated transcription factor (MITF) were significantly increased by treatment with VIP in B16F10 mouse melanoma cells and the stimulatory melanogenic effect was further examined in human epidermal melanocytes (HEMns). In addition, phosphorylated levels of CRE-binding protein (CREB) and protein kinase A (PKA) were markedly increased after VIP treatment, but not p38 mitogen-activated protein kinase (p38 MAPK), extracellular signal-regulated kinase (ERK), or Akt, indicating the possible PKA-CREB signaling pathway involved in VIP-induced melanogenesis. This result was further verified by the fact that VIP induced increased melanin synthesis, and protein levels of phosphorylated CREB, MITF, tyrosinase were significantly attenuated by H89 (a specific PKA inhibitor). These data suggest that VIP-induced upregulation of tyrosinase through the CREB-MITF signaling pathway plays an important role in finding new treatment strategy for skin inflammatory diseases related pigmentation disorders.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Melanins/biosynthesis , Melanoma, Experimental/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Monophenol Monooxygenase/metabolism , Signal Transduction , Vasoactive Intestinal Peptide/pharmacology , Animals , Melanoma, Experimental/pathology , Mice , Microphthalmia-Associated Transcription Factor/genetics , RNA, Messenger/genetics
3.
Exp Dermatol ; 25(7): 526-31, 2016 07.
Article in English | MEDLINE | ID: mdl-26900010

ABSTRACT

Ultraviolet (UV) irradiation on skin triggers photoageing-related phenotypes such as formation of wrinkles. UV ray upregulates matrix metalloproteinase-1 (MMP-1), which in turn degrades extracellular matrix proteins, mostly collagens. Serum amyloid A1 (SAA1) is an acute-phase protein of which plasma concentration increases in response to inflammation. Although the expression of SAA1 in the skin was reported, its function in the skin is yet to be studied. In this research, we found that the expression of SAA1 was increased in acute UV-irradiated buttock skin and photoaged forearm skin in vivo. UV irradiation also increased SAA1 in normal human epidermal keratinocytes (NHEK), and treatment of recombinant human SAA1 (rhSAA1) induced MMP-1 in normal human dermal fibroblasts (NHDF) but not in NHEK. Next, we demonstrated that NHDF treated with UV-irradiated keratinocyte-conditioned media showed the increased MMP-1 expression; however, this increase of MMP-1 in NHDF was inhibited by knockdown of SAA1 in NHEK. In addition, knockdown of Toll-like receptor 4 (TLR4) inhibited rhSAA1-induced MMP-1 expression in NHDF. Taken together, our data showed that UV-induced SAA1 production in NHEK, and this secreted SAA1 induced MMP-1 expression in NHDF in a paracrine manner through TLR4 signalling pathway. Therefore, our results suggest that SAA1 can be a potential mediator for UV-induced MMP-1 expression in human skin.


Subject(s)
Fibroblasts/metabolism , Keratinocytes/radiation effects , Matrix Metalloproteinase 1/metabolism , Serum Amyloid A Protein/metabolism , Skin Aging/radiation effects , Adult , Aged , Aged, 80 and over , Healthy Volunteers , Humans , Keratinocytes/metabolism , Middle Aged , Signal Transduction , Toll-Like Receptor 4/metabolism , Ultraviolet Rays/adverse effects
4.
Acta Pharmacol Sin ; 34(2): 289-94, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23123645

ABSTRACT

AIM: To investigate the effect of [6]-shogaol, an active ingredient in ginger, on melanogenesis and the underlying mechanisms. METHODS: B16F10 mouse melanoma cells were tested. Cell viability was determined with the MTT assay. Melanin content and tyrosinase activity were analyzed with a spectrophotometer. The protein expression of tyrosinase and microphthalmia associated transcription factor (MITF), as well as phosphorylated or total ERK1/2 and Akt were measured using Western blot. RESULTS: Treatment of the cells with [6]-shogaol (1, 5, 10 µmol/L) reduced the melanin content in a concentration-dependent manner. [6]-Shogaol (5 and 10 µmol/L) significantly decreased the intracellular tyrosinase activity, and markedly suppressed the expression levels of tyrosinase and MITF proteins in the cells. Furthermore, [6]-shogaol (10 µmol/L) activated ERK, which was known to negatively regulate melanin synthesis in these cells. Pretreatment with the specific ERK pathway inhibitor PD98059 (20 µmol/L) greatly attenuated the inhibition of melanin synthesis by [6]-shogaol (10 µmol/L). CONCLUSION: The results demonstrate that [6]-shogaol inhibits melanogenesis in B16F10 mouse melanoma cells via activating the ERK pathway.


Subject(s)
Catechols/pharmacology , Enzyme Activation/drug effects , MAP Kinase Signaling System/drug effects , Melanins/antagonists & inhibitors , Melanins/metabolism , Melanoma, Experimental/metabolism , Animals , Cell Line, Tumor , Zingiber officinale/chemistry , Mice , Monophenol Monooxygenase/antagonists & inhibitors , Monophenol Monooxygenase/metabolism
5.
Aging Cell ; 22(11): e14000, 2023 11.
Article in English | MEDLINE | ID: mdl-37828898

ABSTRACT

Aging is accompanied by impaired mitochondrial function and accumulation of senescent cells. Mitochondrial dysfunction contributes to senescence by increasing the levels of reactive oxygen species and compromising energy metabolism. Senescent cells secrete a senescence-associated secretory phenotype (SASP) and stimulate chronic low-grade inflammation, ultimately inducing inflammaging. Mitochondrial dysfunction and cellular senescence are two closely related hallmarks of aging; however, the key driver genes that link mitochondrial dysfunction and cellular senescence remain unclear. Here, we aimed to elucidate a novel role of carnitine acetyltransferase (CRAT) in the development of mitochondrial dysfunction and cellular senescence in dermal fibroblasts. Transcriptomic analysis of skin tissues from young and aged participants showed significantly decreased CRAT expression in intrinsically aged skin. CRAT downregulation in human dermal fibroblasts recapitulated mitochondrial changes in senescent cells and induced SASP secretion. Specifically, CRAT knockdown caused mitochondrial dysfunction, as indicated by increased oxidative stress, disruption of mitochondrial morphology, and a metabolic shift from oxidative phosphorylation to glycolysis. Mitochondrial damage induced the release of mitochondrial DNA into the cytosol, which activated the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) and NF-ĸB pathways to induce SASPs. Consistently, fibroblast-specific CRAT-knockout mice showed increased skin aging phenotypes in vivo, including decreased cell proliferation, increased SASP expression, increased inflammation, and decreased collagen density. Our results suggest that CRAT deficiency contributes to aging by mediating mitochondrial dysfunction-induced senescence.


Subject(s)
Carnitine O-Acetyltransferase , Cellular Senescence , Animals , Mice , Humans , Aged , Carnitine O-Acetyltransferase/metabolism , Cellular Senescence/physiology , Mitochondria/metabolism , NF-kappa B/metabolism , Inflammation/metabolism , Fibroblasts/metabolism
6.
Biomed Pharmacother ; 150: 113034, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35489284

ABSTRACT

Photoaging mainly occurs due to ultraviolet (UV) radiation, and is accompanied by increased secretion of matrix metalloproteinases (MMPs) and degradation of collagen. UV radiation induces cell senescence in the skin; however, the role of senescent cells in photoaging remains unclear. Therefore, to elucidate the role of senescent cells in photoaging, we evaluated the effect of senolytics in a photoaging mouse model and investigated the underlying mechanism of their antiaging effect. Both UV-induced senescent human dermal fibroblasts and a photoaging mouse model, ABT-263 and ABT-737, demonstrated senolytic effects on senescent fibroblasts. Moreover, we found that several senescence-associated secretory phenotype factors, such as IL-6, CCL5, CCL7, CXCL12, and SCF, induced MMP-1 expression in dermal fibroblasts, which decreased after treatment with ABT-263 and ABT-737 in vivo and in vitro. Both senolytic drugs attenuated the induction of MMPs and decreased collagen density in the photoaging mouse model. Our data suggest that senolytic agents reduce UV-induced photoaging, making strategies for targeting senescent dermal fibroblasts promising options for the treatment of photoaging.


Subject(s)
Skin Aging , Skin Diseases , Animals , Cells, Cultured , Collagen/metabolism , Fibroblasts , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinases/metabolism , Mice , Senotherapeutics , Skin , Skin Diseases/metabolism , Ultraviolet Rays
7.
Commun Biol ; 5(1): 702, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35835838

ABSTRACT

The multifaceted nature of senescent cell cycle arrest necessitates the targeting of multiple factors arresting or promoting the cell cycle. We report that co-inhibition of ATM and ROCK by KU-60019 and Y-27632, respectively, synergistically increases the proliferation of human diploid fibroblasts undergoing replicative senescence through activation of the transcription factors E2F1 and FOXM1. Time-course transcriptome analysis identified FOXM1 and E2F1 as crucial factors promoting proliferation. Co-inhibition of the kinases ATM and ROCK first promotes the G2/M transition via FOXM1 activation, leading to accumulation of cells undergoing the G1/S transition via E2F1 activation. The combination of both inhibitors increased this effect more significantly than either inhibitor alone, suggesting synergism. Our results demonstrate a FOXM1- and E2F1-mediated molecular pathway enhancing cell cycle progression in cells with proliferative potential under replicative senescence conditions, and treatment with the inhibitors can be tested for senomorphic effect in vivo.


Subject(s)
Cellular Senescence , E2F1 Transcription Factor , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Proliferation , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/pharmacology , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/metabolism , Forkhead Box Protein M1/pharmacology , Humans
8.
J Dermatol Sci ; 103(1): 16-24, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34030962

ABSTRACT

BACKGROUND: Melanin plays important roles in determining human skin color and protecting human skin cells against harmful ultraviolet light. However, abnormal hyperpigmentation in some areas of the skin may become aesthetically unpleasing, resulting in the need for effective agents or methods to regulate undesirable hyperpigmentation. OBJECTIVE: We investigated the effect of harmine, a natural harmala alkaloid belonging to the beta-carboline family, on melanin synthesis and further explored the signaling pathways involved in its mechanism of action. METHODS: Human MNT-1 melanoma cells and human primary melanocytes were treated with harmine, chemical inhibitors, small interfering RNAs, or mammalian expression vectors. Cell viability, melanin content, and expression of various target molecules were assessed. RESULTS: Harmine decreased melanin synthesis and tyrosinase expression in human MNT-1 melanoma cells. Inhibition of DYRK1A, a harmine target, decreased melanin synthesis and tyrosinase expression. Further studies revealed that nuclear translocation of NFATC3, a potential DYRK1A substrate, was induced via the harmine/DYRK1A pathway and that NFATC3 knockdown increased melanin synthesis and tyrosinase expression. Suppression of melanin synthesis and tyrosinase expression via the harmine/DYRK1A pathway was significantly attenuated by NFATC3 knockdown. Furthermore, harmine also decreased melanin synthesis and tyrosinase expression through regulation of NFATC3 in human primary melanocytes. CONCLUSION: Our results indicate that harmine decreases melanin synthesis through regulation of the DYRK1A/NFATC3 pathway and suggest that the DYRK1A/NFATC3 pathway may be a potential target for the development of depigmenting agents.


Subject(s)
Harmine/pharmacology , Melanins/antagonists & inhibitors , NFATC Transcription Factors/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Skin Lightening Preparations/pharmacology , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Melanins/biosynthesis , Melanocytes/drug effects , Melanocytes/metabolism , NFATC Transcription Factors/genetics , Primary Cell Culture , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Skin/cytology , Skin/metabolism , Skin Pigmentation/drug effects , Dyrk Kinases
9.
J Dermatol Sci ; 101(2): 107-114, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33309320

ABSTRACT

BACKGROUND: Ultraviolet (UV) irradiation is the main contributing factor for skin aging. UV irradiation induces epigenetic changes in skin. It increases the activity of histone acetylases (HATs) but decreases that of histone deacetylases (HDACs). OBJECTIVE: We aimed to investigate alterations in all classes of HDACs and sirtuins (SIRTs) in response to UV irradiation, and determine the HDACs regulating the expressions of matrix metalloproteinase 1 (MMP-1) and type I procollagen. METHODS: Primary human dermal fibroblasts were UV irradiated. HDAC4 was knocked-down or overexpressed to investigate its effect on the expression of MMP-1 and type I procollagen. The mRNA and protein levels were analyzed by quantitative real-time polymerase chain reaction and western blotting. RESULTS: Among 11 HDACs and 7 SIRTs, we found that the expression of HDAC4, HDAC5, HDAC6, HDAC7, HDAC8, HDAC11, SIRT2, and SIRT3 were significantly and consistently reduced by UV at both mRNA and protein levels. Among these, the reduction of HDAC4 was responsible for the basal and UV-induced increase in the expression of MMP-1 and decrease in that of type I procollagen. Furthermore, the reduced HDAC4 could activate c-Jun N-terminal kinase (JNK), resulting in an increase in MMP-1 and decrease in type I procollagen. CONCLUSIONS: UV treatment decreases the expression of HDACs and SIRTs in dermal fibroblasts; in particular, the UV-induced reduction in the expression of HDAC4 might play an important role in regulating the expression of MMP-1 and type I procollagen.


Subject(s)
Collagen Type I/metabolism , Histone Deacetylases/genetics , Matrix Metalloproteinase 1/genetics , Procollagen/metabolism , Repressor Proteins/genetics , Skin Aging/radiation effects , Ultraviolet Rays/adverse effects , Cells, Cultured , Down-Regulation/radiation effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Gene Knockdown Techniques , Healthy Volunteers , Histone Deacetylases/metabolism , Humans , MAP Kinase Signaling System/radiation effects , Primary Cell Culture , Repressor Proteins/metabolism , Sirtuins/metabolism , Skin/cytology , Skin/metabolism , Skin/radiation effects , Skin Aging/genetics , Up-Regulation
10.
Exp Mol Med ; 53(6): 1092-1108, 2021 06.
Article in English | MEDLINE | ID: mdl-34188179

ABSTRACT

Senescent cells exhibit a reduced response to intrinsic and extrinsic stimuli. This diminished reaction may be explained by the disrupted transmission of nuclear signals. However, this hypothesis requires more evidence before it can be accepted as a mechanism of cellular senescence. A proteomic analysis of the cytoplasmic and nuclear fractions obtained from young and senescent cells revealed disruption of nucleocytoplasmic trafficking (NCT) as an essential feature of replicative senescence (RS) at the global level. Blocking NCT either chemically or genetically induced the acquisition of an RS-like senescence phenotype, named nuclear barrier-induced senescence (NBIS). A transcriptome analysis revealed that, among various types of cellular senescence, NBIS exhibited a gene expression pattern most similar to that of RS. Core proteomic and transcriptomic patterns common to both RS and NBIS included upregulation of the endocytosis-lysosome network and downregulation of NCT in senescent cells, patterns also observed in an aging yeast model. These results imply coordinated aging-dependent reduction in the transmission of extrinsic signals to the nucleus and in the nucleus-to-cytoplasm supply of proteins/RNAs. We further showed that the aging-associated decrease in Sp1 transcription factor expression was critical for the downregulation of NCT. Our results suggest that NBIS is a modality of cellular senescence that may represent the nature of physiological aging in eukaryotes.


Subject(s)
Cellular Senescence , Proteomics , Cell Nucleus/metabolism , Cellular Senescence/genetics , Down-Regulation
11.
J Korean Med Sci ; 25(6): 930-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20514317

ABSTRACT

Polyunsaturated fatty acids (PUFAs) are known to play important roles in various physiological and pathological processes. Recent studies have shown that some omega-3 (omega-3) PUFAs, such as eicosapentaenoic acid (EPA) and dodecahexaenoic acid (DHA), have protective effects on acute and chronic UV-induced changes. However, the effects of other omega-3 PUFAs including 11,14,17-eicosatrienoic acid (20:3) (ETA) on UV-induced skin damages are poorly understood. In this study, we investigated the cutaneous photoprotective effects of ETA in hairless mice in vivo. Female HR-1 hairless mice were topically treated with vehicle (ethanol:polyethylene glycol=30:70) only, 0.1% ETA, or 1% ETA once a day for 3 successive days after one time UV irradiation (200 mJ/cm(2)) on dorsal skins. Skin biopsy was carried out on the fourth day (72 hr after UV irradiation). We found that topical treatment with ETA attenuated UV-induced epidermal and dermal thickness and infiltration of inflammatory cells, and impairment of skin barrier function. In addition, ETA suppressed the expression of IL-1beta, COX-2, and MMP-13 induced by UV irradiation. Our results show that the topical application of ETA protects against UV-induced skin damage in hairless mice and suggest that ETA can be a potential agent for preventing and/or treating UV-induced inflammation and photoaging.


Subject(s)
8,11,14-Eicosatrienoic Acid/administration & dosage , Radiation-Protective Agents/administration & dosage , Skin Diseases/prevention & control , Skin/radiation effects , Ultraviolet Rays , Administration, Topical , Animals , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Female , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Mice , Mice, Hairless , Skin/drug effects , Skin/pathology , Skin Diseases/pathology
12.
J Investig Dermatol Symp Proc ; 14(1): 15-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19675547

ABSTRACT

Sunlight damages human skin, resulting in a wrinkled appearance. Since natural sunlight is polychromatic, its ultimate effects on the human skin are the result of not only the action of each wavelength separately, but also interactions among the many wavelengths, including UV, visible light, and infrared (IR). In direct sunlight, the temperature of human skin rises to about 40 degrees C following the conversion of absorbed IR into heat. So far, our knowledge of the effects of IR radiation or heat on skin aging is limited. Recent work demonstrates that IR and heat exposure each induces cutaneous angiogenesis and inflammatory cellular infiltration, disrupts the dermal extracellular matrix by inducing matrix metalloproteinases, and alters dermal structural proteins, thereby adding to premature skin aging. This review provides a summary of current research on the effects of IR radiation and heat on aging in human skin in vivo.Journal of Investigative Dermatology Symposium Proceedings (2009) 14, 15-19; doi:10.1038/jidsymp.2009.7.


Subject(s)
Skin Aging/radiation effects , Collagen/metabolism , Cytokines/metabolism , DNA Damage , Fibrillins , Hot Temperature/adverse effects , Humans , Infrared Rays/adverse effects , Keratinocytes/metabolism , Mast Cells/metabolism , Mast Cells/pathology , Mast Cells/radiation effects , Matrix Metalloproteinases/metabolism , Microfilament Proteins/metabolism , Neovascularization, Pathologic/etiology , Reactive Oxygen Species/metabolism , Skin/blood supply , Skin/injuries , Skin/metabolism , Skin/radiation effects , Skin Aging/physiology , TRPV Cation Channels/metabolism , Tropoelastin/metabolism , Tryptases/metabolism
13.
Exp Gerontol ; 106: 8-15, 2018 06.
Article in English | MEDLINE | ID: mdl-29474864

ABSTRACT

In our previous study, we uncovered a novel mechanism in which amelioration of Hutchinson-Gilford progeria syndrome (HGPS) phenotype is mediated by mitochondrial functional recovery upon rho-associated protein kinase (ROCK) inhibition. However, it remains elusive whether this mechanism is also applied to the amelioration of normal aging cells. In this study, we used Y-27632 and fasudil as effective ROCK inhibitors, and examined their role in senescence. We found that ROCK inhibition induced the functional recovery of the mitochondria as well as the metabolic reprogramming, which are two salient features that are altered in normal aging cells. Moreover, microarray analysis revealed that the up-regulated pathway upon ROCK inhibition is enriched for chromatin remodeling genes, which may play an important role in the alleviation of senescence-associated cell cycle arrest. Indeed, ROCK inhibition induced cellular proliferation, concomitant with the amelioration of senescent phenotype. Furthermore, the restorative effect by ROCK inhibition was observed in vivo as evidenced by the facilitated cutaneous wound healing. Taken together, our data indicate that ROCK inhibition might be utilized to ameliorate normal aging process and to treat age-related disease.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Amides/pharmacology , Cellular Senescence/drug effects , Pyridines/pharmacology , Wound Healing/drug effects , rho-Associated Kinases/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Cell Proliferation/drug effects , Child , Chromatin/genetics , Fibroblasts/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Phenotype , Progeria/pathology
14.
J Dermatol Sci ; 90(2): 180-189, 2018 May.
Article in English | MEDLINE | ID: mdl-29428815

ABSTRACT

BACKGROUND: Ultraviolet (UV) radiation plays important roles in various skin diseases including premature aging and cancer. UV has been shown to regulate the expressions of many genes including matrix metalloproteinases (MMPs). Gasdermin C (GSDMC) belongs to Gasdermin family and is known to be expressed in the epithelial cells of many tissues including the skin. However, the functions of GSDMC remain poorly understood. OBJECTIVE: We aimed to investigate the role of GSDMC in UV-induced MMP-1, MMP-3, and MMP-9 expressions in human skin keratinocytes. METHODS: Primary human skin keratinocytes and an immortalized human skin keratinocyte cell line (HaCaT cells) were irradiated with UV. Knockdown and overexpression of GSDMC were performed to study the effect of GSDMC. The mRNA and protein levels were analyzed by quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting, respectively. RESULTS: We found that GSDMC expression is increased by UV irradiation in human skin keratinocytes. Further studies showed that GSDMC expression is increased at relatively late time points after UV irradiation and that this GSDMC induction plays important roles in the expressions of MMP-1, but not of MMP-3 and MMP-9, and the activations of ERK and JNK induced by UV. In addition, we found that overexpression of GSDMC increases the MMP-1 expression and the activities of ERK and JNK and that GSDMC-induced MMP-1 expression is suppressed by inhibition of ERK or JNK activities. CONCLUSIONS: Our results suggest that GSDMC is increased by UV radiation and contributes to UV-induced MMP-1 expression through the activation of ERK and JNK pathways.


Subject(s)
Biomarkers, Tumor/metabolism , DNA-Binding Proteins/metabolism , MAP Kinase Signaling System/radiation effects , Matrix Metalloproteinase 1/metabolism , Ultraviolet Rays/adverse effects , Biomarkers, Tumor/genetics , Cell Line , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , Healthy Volunteers , Humans , Keratinocytes , Male , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 9/metabolism , Primary Cell Culture , Skin/cytology , Skin/pathology , Skin/radiation effects , Skin Diseases/etiology , Skin Diseases/pathology
15.
Mol Med Rep ; 17(2): 2581-2585, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29207132

ABSTRACT

Pattern recognition receptors (PRRs) are part of the immune system. They can recognize pathogen­associated molecular patterns (PAMPs). Toll­like receptors (TLRs) and retinoic acid­inducible gene 1 (RIG­1)­like receptors (RLRs) are 2 types of PRR in the innate immune system. Double­stranded RNA (dsRNA) can exist as a PAMP, including dsRNA viruses. dsRNA is known as a ligand not only for TLR3 but also for RLRs, including melanoma differentiation­associated gene 5 and RIG­1. Collagen is the main structural protein in the extracellular space in the skin. Recently, it was reported that treatment of a synthetic dsRNA, poly(I:C), decreases procollagen expression in skin fibroblasts. However, signaling pathways involved in this process have not yet been fully elucidated. The present study further explored the underlying signaling pathways involved in the processes. It was demonstrated by western blotting that treatment of poly(I:C), but not another PAMP, Pam3CSK4, inhibited procollagen expression in cultured human skin fibroblasts. Treatment of poly(I:C)and Pam3CSK4 induced activation of the mitogen­activated protein kinases and the nuclear factor­κB pathways. However, only poly(I:C), but not Pam3CSK4, induced the activation of the interferon regulatory factor 3 (IRF3) pathway. By using specific inhibitors, it was demonstrated that inhibition of IRF3 pathway relieved poly(I:C)­induced procollagen reduction. In conclusion, IRF3 signaling pathway serves an important role in poly(I:C)­induced procollagen reduction in skin fibroblasts. This suggests that the IRF3 signaling pathway may be a key target for collagen regulation in the skin.


Subject(s)
Fibroblasts/drug effects , Fibroblasts/metabolism , Interferon Regulatory Factor-3/metabolism , Poly I-C/pharmacology , Procollagen/metabolism , Signal Transduction/drug effects , Skin/cytology , Cells, Cultured , Humans , Lipopeptides/pharmacology
16.
Int J Mol Med ; 42(5): 2859-2866, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30226565

ABSTRACT

Gasdermin (GSDM)­C is a member of the GSDM gene family and is expressed in the epithelial cells of various tissue types, including skin. GSDMC expression is induced by ultraviolet (UV) irradiation and contributes to UV­induced matrix metalloproteinase 1 expression in human skin keratinocytes. However, how UV irradiation induces GSDMC expression remains unclear. The present study aimed to investigate the role of transient receptor potential cation channel subfamily V member 1 (TRPV1) and a calcium/calcineurin­signaling pathway in UV­induced GSDMC expression in human skin keratinocytes. Suppression of TRPV1 activity by treatment with the TRPV1 antagonists capsazepine and ruthenium red significantly reduced UV­induced GSDMC expression, whereas direct activation of TRPV1 by capsaicin, a TRPV1 agonist, increased GSDMC expression. The results demonstrated that extracellular calcium and calcineurin activity may be necessary for UV­induced GSDMC expression in HaCaT cells. In addition, UV­induced GSDMC expression was either decreased or increased following knockdown or overexpression of nuclear factor of activated T­cells, cytoplasmic 1 (NFATc1), respectively. These data suggested that TRPV1 may serve an important role in the induction of GSDMC expression by UV and that UV­induced GSDMC expression may be mediated via a calcium/calcineurin/NFATc1 pathway.


Subject(s)
Biomarkers, Tumor/genetics , Calcineurin/metabolism , Calcium/metabolism , DNA-Binding Proteins/genetics , Keratinocytes/radiation effects , NFATC Transcription Factors/metabolism , Signal Transduction/radiation effects , TRPV Cation Channels/metabolism , Cell Line , Humans , Keratinocytes/metabolism , Ultraviolet Rays , Up-Regulation/radiation effects
17.
J Invest Dermatol ; 137(8): 1757-1765, 2017 08.
Article in English | MEDLINE | ID: mdl-28392346

ABSTRACT

Ubiquitin carboxyl-terminal hydrolase L1 (UCHL1) is involved in many signaling pathways via the ubiquitin-proteasome system. UCHL1 is expressed in the human skin and serves as a neuronal marker; however, its functions in melanogenesis remain unknown. Here, we investigated the role of UCHL1 in melanogenesis and elucidated the underlying mechanism using human melanocytes. UCHL1 downregulation by small interfering RNA resulted in upregulation of microphthalmia-associated transcription factor (MITF), tyrosinase, dopachrome tautomerase, tyrosinase-related protein-1, and melanin. In contrast, overexpression of UCHL1 in melanocytes via adenovirus transfection led to downregulation of tyrosinase, dopachrome tautomerase, and tyrosinase-related protein-1 and decreased melanin contents. Furthermore, UCHL1 reduced the protein, but not mRNA, levels of MITF, the upstream regulator of tyrosinase, dopachrome tautomerase, and tyrosinase-related protein-1. Inhibition of de novo protein synthesis and treatment of normal human primary epidermal melanocytes with proteasome inhibitor MG132 revealed that UCHL1 negatively regulates the stability of MITF by binding to the ubiquitinated protein. Finally, overexpression of MITF via an adenovirus restored the level of melanogenesis reduced by UCHL1. Collectively, our findings indicate a role of UCHL1 in regulating skin pigmentation. Suppression of MITF activity by UCHL1 via protein degradation might aid in the development of new therapeutic approaches for melanoma or dyspigmentation disorders.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanocytes/metabolism , Melanoma/genetics , Microphthalmia-Associated Transcription Factor/genetics , RNA, Neoplasm/genetics , Skin Neoplasms/genetics , Ubiquitin Thiolesterase/genetics , Blotting, Western , Humans , Male , Melanocytes/pathology , Melanoma/metabolism , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/biosynthesis , Real-Time Polymerase Chain Reaction , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Cells, Cultured , Ubiquitin Thiolesterase/biosynthesis , Melanoma, Cutaneous Malignant
18.
J Dermatol Sci ; 88(1): 126-133, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28551094

ABSTRACT

BACKGROUND: Transient receptor potential type 1 (TRPV1) can be activated by ultraviolet (UV) irradiation, and mediates UV-induced matrix metalloproteinase (MMP)-1 and proinflammatory cytokines in keratinocytes. Various chemicals and compounds targeting TRPV1 activation have been developed, but are not in clinical use mostly due to their safety issues. OBJECTIVE: We aimed to develop a novel TRPV1-targeting peptide to inhibit UV-induced responses in human skin. METHODS: We designed and generated a novel TRPV1 inhibitory peptide (TIP) which mimics the specific site in TRPV1 (aa 701-709: Gln-Arg-Ala-Ile-Thr-Ile-Leu-Asp-Thr, QRAITILDT), Thr705, and tested its efficacy of blocking UV-induced responses in HaCaT, mouse, and human skin. RESULTS: TIP effectively inhibited capsaicin-induced calcium influx and TRPV1 activation. Treatment of HaCaT with TIP prevented UV-induced increases of MMP-1 and pro-inflammatory cytokines such as interleukin (IL)-6 and tumor necrosis factor-α. In mouse skin in vivo, TIP inhibited UV-induced skin thickening and prevented UV-induced expression of MMP-13 and MMP-9. Moreover, TIP attenuated UV-induced erythema and the expression of MMP-1, MMP-2, IL-6, and IL-8 in human skin in vivo. CONCLUSION: The novel synthetic peptide targeting TRPV1 can ameliorate UV-induced skin responses in vitro and in vivo, providing a promising therapeutic approach against UV-induced inflammation and photoaging.


Subject(s)
Erythema/drug therapy , Peptides/pharmacology , Skin Aging/drug effects , Skin/metabolism , TRPV Cation Channels/antagonists & inhibitors , Ultraviolet Rays/adverse effects , Adult , Animals , Back , Biopsy , Calcium/metabolism , Capsaicin/pharmacology , Cell Line , Collagenases/metabolism , Disease Models, Animal , Erythema/etiology , Female , Healthy Volunteers , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/radiation effects , Male , Mice , Mice, Hairless , Peptides/chemical synthesis , Peptides/therapeutic use , Phosphorylation , Skin/drug effects , Skin/pathology , Skin/radiation effects , Skin Aging/pathology , TRPV Cation Channels/metabolism , Threonine/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
Photochem Photobiol ; 82(2): 574-8, 2006.
Article in English | MEDLINE | ID: mdl-16613515

ABSTRACT

Repetitive exposure of the skin to UV radiation induces various harmful changes, such as thickening, wrinkle formation, inflammation and carcinogenesis. A variety of natural compounds and synthetic compounds have been studied to determine whether they can prevent UV-induced harmful effects. In this study, we investigated the effect of a novel compound, Melanocin A, which was isolated from Eupenicillium shearii F80695, on UV-induced premature skin aging. First, we studied the effect of Melanocin A on UV-induced matrix metalloproteinase (MMP)-9 expression in an immortalized human keratinocyte cell line, HaCaT, in vitro. Acute UV irradiation induced MMP-9 expression at both the mRNA and protein levels and Melanocin A suppressed this expression in a dose-dependent manner. We then investigated the effect of Melanocin A on UV-induced skin changes in hairless mice in vivo. Chronic exposure of hairless mouse dorsal skin to UV increased skin thickness and induced wrinkle formation and the gelatinase activities of MMP-2 and MMP-9. Moreover, Melanocin A significantly suppressed UV-induced morphologic skin changes and MMP-2 and MMP-9 expression. Taken together, these results show that Melanocin A can prevent the harmful effects of UV that lead to skin aging. Therefore, we suggest that Melanocin A should be viewed as a potential therapeutic agent for preventing and/or treating premature skin aging.


Subject(s)
Cyanides/pharmacology , Eurotiales/chemistry , Keratinocytes/drug effects , Skin Aging/drug effects , Ultraviolet Rays/adverse effects , Animals , Cell Line , Dose-Response Relationship, Drug , Humans , Keratinocytes/cytology , Keratinocytes/physiology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Hairless , Phototherapy/methods , Proteins/analysis , Proteins/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Skin Aging/physiology , Skin Aging/radiation effects , Time Factors
20.
J Invest Dermatol ; 124(2): 315-23, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15675949

ABSTRACT

Dehydroepiandrosterone (DHEA) and its sulfate conjugate (DHEA-S) are the most abundantly produced human adrenal steroids to be reduced with age. DHEA may be related to the process of skin aging through the regulation and degradation of extracelluar matrix protein. In this study, we demonstrate that DHEA can increase procollagen synthesis and inhibit collagen degradation by decreasing matrix metalloproteinases (MMP)-1 synthesis and increasing tisuue inhibitor of matrix metalloprotease (TIMP-1) production in cultured dermal fibroblasts. DHEA was found to inhibit ultraviolet (UV)-induced MMP-1 production and the UV-induced decrease of procollagen synthesis, probably due to the inhibition of UV-induced AP-1 activity. DHEA (5%) in ethanol:olive oil (1:2) was topically applied to buttock skin of volunteers 12 times over 4 weeks, and was found to significantly increase the expression of procollagen alpha1(I) mRNA and protein in both aged and young skin. On the other hand, topical DHEA significantly decreased the basal expression of MMP-1 mRNA and protein, but increased the expression of TIMP-1 protein in aged skin. We also found that DHEA induced the expressions of transforming growth factor-beta1 and connective tissue growth factor mRNA in cultured fibroblasts and aged skin, which may play a role in the DHEA-induced changes of procollagen and MMP-1 expression. Our results suggest the possibility of using DHEA as an anti-skin aging agent.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Collagen Type I/metabolism , Dehydroepiandrosterone/administration & dosage , Dermis/drug effects , Dermis/metabolism , Administration, Topical , Adult , Aged , Aged, 80 and over , Cells, Cultured , Collagen Type I/genetics , Connective Tissue Growth Factor , Dermis/cytology , Fibroblasts/cytology , Fibroblasts/radiation effects , Gene Expression/drug effects , Humans , Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Male , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , RNA, Messenger/analysis , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transcription Factor AP-1/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1 , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL