Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 562(7727): 429-433, 2018 10.
Article in English | MEDLINE | ID: mdl-30297801

ABSTRACT

Despite the efficacy of Hedgehog pathway inhibitors in the treatment of basal cell carcinoma (BCC)1, residual disease persists in some patients and may contribute to relapse when treatment is discontinued2. Here, to study the effect of the Smoothened inhibitor vismodegib on tumour clearance, we have used a Ptch1-Trp53 mouse model of BCC3 and found that mice treated with vismodegib harbour quiescent residual tumours that regrow upon cessation of treatment. Profiling experiments revealed that residual BCCs initiate a transcriptional program that closely resembles that of stem cells of the interfollicular epidermis and isthmus, whereas untreated BCCs are more similar to the hair follicle bulge. This cell identity switch was enabled by a mostly permissive chromatin state accompanied by rapid Wnt pathway activation and reprogramming of super enhancers to drive activation of key transcription factors involved in cellular identity. Accordingly, treatment of BCC with both vismodegib and a Wnt pathway inhibitor reduced the residual tumour burden and enhanced differentiation. Our study identifies a resistance mechanism in which tumour cells evade treatment by adopting an alternative identity that does not rely on the original oncogenic driver for survival.


Subject(s)
Anilides/pharmacology , Carcinoma, Basal Cell/pathology , Cell Differentiation/drug effects , Hedgehog Proteins/antagonists & inhibitors , Pyridines/pharmacology , Signal Transduction/drug effects , Skin Neoplasms/pathology , Anilides/administration & dosage , Anilides/therapeutic use , Animals , Carcinoma, Basal Cell/drug therapy , Carcinoma, Basal Cell/metabolism , Cell Proliferation/drug effects , Epidermal Cells/drug effects , Epidermal Cells/metabolism , Epidermal Cells/pathology , Hair Follicle/drug effects , Hair Follicle/metabolism , Hair Follicle/pathology , Hedgehog Proteins/metabolism , Humans , Mice , Pyridines/administration & dosage , Pyridines/therapeutic use , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Smoothened Receptor/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/pathology , Wnt Signaling Pathway/drug effects
2.
Acta Biochim Biophys Sin (Shanghai) ; 56(7): 1072-1084, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38779766

ABSTRACT

Epidermal stem cells (EpSCs) play a vital role in skin wound healing through re-epithelialization. Identifying chemicals that can promote EpSC proliferation is helpful for treating skin wounds. This study investigates the effect of morroniside on cutaneous wound healing in mice and explores the underlying mechanisms. Application of 10‒50 µg/mL of morroniside to the skin wound promotes wound healing in mice. In vitro studies demonstrate that morroniside stimulates the proliferation of mouse and human EpSCs in a time- and dose-dependent manner. Mechanistic studies reveal that morroniside promotes the proliferation of EpSCs by facilitating the cell cycle transition from the G1 to S phase. Morroniside increases the expression of ß-catenin via the glucagon-like peptide-1 receptor (GLP-1R)-mediated PKA, PKA/PI3K/AKT and PKA/ERK signaling pathways, resulting in an increase in cyclin D1 and cyclin E1 expression, either directly or by upregulating c-Myc expression. This process ultimately leads to EpSC proliferation. Administration of morroniside to mouse skin wounds increases the phosphorylation of AKT and ERK, the expressions of ß-catenin, c-Myc, cyclin D1, and cyclin E1, as well as the proliferation of EpSCs, in periwound skin tissue, and accelerates wound re-epithelialization. These effects of morroniside are mediated by the GLP-1R. Overall, these results indicate that morroniside promotes skin wound healing by stimulating the proliferation of EpSCs via increasing ß-catenin expression and subsequently upregulating c-Myc, cyclin D1, and cyclin E1 expressions through GLP-1R signaling pathways. Morroniside has clinical potential for treating skin wounds.


Subject(s)
Cell Proliferation , Re-Epithelialization , Stem Cells , Up-Regulation , beta Catenin , Animals , beta Catenin/metabolism , beta Catenin/genetics , Cell Proliferation/drug effects , Mice , Up-Regulation/drug effects , Humans , Re-Epithelialization/drug effects , Stem Cells/metabolism , Stem Cells/drug effects , Stem Cells/cytology , Wound Healing/drug effects , Signal Transduction/drug effects , Male , Epidermal Cells/metabolism , Epidermal Cells/drug effects , Epidermal Cells/cytology , Cyclin D1/metabolism , Cyclin D1/genetics
3.
Int J Mol Sci ; 25(8)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38674064

ABSTRACT

Olive leaf contains plenty of phenolic compounds, among which oleuropein (OP) is the main component and belongs to the group of secoiridoids. Additionally, phenolic compounds such as oleocanthal (OL) and oleacein (OC), which share a structural similarity with OP and two aldehyde groups, are also present in olive leaves. These compounds have been studied for several health benefits, such as anti-cancer and antioxidant effects. However, their impact on the skin remains unknown. Therefore, this study aims to compare the effects of these three compounds on melanogenesis using B16F10 cells and human epidermal cells. Thousands of gene expressions were measured by global gene expression profiling with B16F10 cells. We found that glutaraldehyde compounds derived from olive leaves have a potential effect on the activation of the melanogenesis pathway and inducing differentiation in B16F10 cells. Accordingly, the pro-melanogenesis effect was investigated by means of melanin quantification, mRNA, and protein expression using human epidermal melanocytes (HEM). This study suggests that secoiridoid and its derivates have an impact on skin protection by promoting melanin production in both human and mouse cell lines.


Subject(s)
Iridoid Glucosides , Melanins , Melanocytes , Olea , Phenols , Humans , Melanocytes/drug effects , Melanocytes/metabolism , Olea/chemistry , Animals , Melanins/biosynthesis , Melanins/metabolism , Mice , Phenols/pharmacology , Iridoid Glucosides/pharmacology , Iridoids/pharmacology , Aldehydes/pharmacology , Cell Differentiation/drug effects , Cyclopentane Monoterpenes , Epidermal Cells/metabolism , Epidermal Cells/drug effects , Plant Extracts/pharmacology , Plant Extracts/chemistry , Epidermis/metabolism , Epidermis/drug effects , Cell Line, Tumor , Plant Leaves/chemistry , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanogenesis
4.
Int J Mol Sci ; 25(16)2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39201318

ABSTRACT

The skin is a direct target of the air pollutant benzo[a]pyrene (BaP). While its carcinogenic qualities are well-studied, the immunotoxicity of BaP after dermal exposure is less understood. This study examines the immunomodulatory effects of a 10-day epicutaneous BaP application, in environmentally/occupationally relevant doses, by analyzing ex vivo skin immune response (skin explant, epidermal cells and draining lymph node/DLN cell activity), alongside the skin's reaction to sensitization with experimental hapten dinitrochlorobenzene (DNCB). The results show that BaP application disrupts the structure of the epidermal layer and promotes immune cell infiltration in the dermis. BaP exposure led to oxidative stress in epidermal cells, characterized by decreased reduced glutathione and increased AHR and Cyp1A1 expression. Production and gene expression of proinflammatory cytokines (TNF, IL-1ß) by epidermal cells decreased, while IL-10 response increased. Decreased spontaneous production of IFN-γ and IL-17, along with unchanged IL-10, was observed in DLC cells, whereas ConA-stimulated production of these cytokines was elevated. Local immunosuppression caused by BaP application seems to reduce the skin's response to an additional stimulus, evidenced by decreased effector activity of DLN cells three days after sensitization with DNCB. These findings provide new insight into the immunomodulatory effects and health risks associated with skin exposure to BaP.


Subject(s)
Benzo(a)pyrene , Cytokines , Lymph Nodes , Benzo(a)pyrene/toxicity , Animals , Rats , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/metabolism , Cytokines/metabolism , Skin/drug effects , Skin/metabolism , Skin/immunology , Epidermal Cells/drug effects , Epidermal Cells/metabolism , Epidermis/drug effects , Epidermis/metabolism , Epidermis/immunology , Oxidative Stress/drug effects , Dinitrochlorobenzene , Male , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A1/genetics
5.
J Allergy Clin Immunol ; 147(6): 2202-2212.e8, 2021 06.
Article in English | MEDLINE | ID: mdl-33338537

ABSTRACT

BACKGROUND: Janus kinase (JAK) inhibitors are a new class of therapeutic compounds for dermatological diseases. In atopic dermatitis (AD), data of clinical phase III trials show rapid improvement of pruritus and significant reduction of inflammation within the first weeks with a favorable safety profile. However, their mode of action in AD is not fully understood. OBJECTIVES: In our study, we investigate the effect of different JAK inhibitors on cell differentiation, phenotype, and function of inflammatory dendritic epidermal cells (IDECs). METHODS: We analyzed the JAK expression in IDEC from ex vivo skin and in vitro generated IDECs using flow cytometry and PCR. Further, we studied in vitro the effect of different JAK inhibitors on IDEC cell differentiation, phenotype, and maturation. RESULTS: IDECs express JAK1 and JAK2 ex vivo and in vitro. We found that JAK1 and JAK2 were upregulated during the differentiation from monocytes to IDECs. Conversely, JAK2 inhibition by ruxolitinib (JAK1/2 inhibitor) or BMS-911543 (JAK2 inhibitor) abrogated the differentiation from monocytes into IDECs. Differentiated IDECs can redifferentiate into a more monocyte-like phenotype in the presence of ruxolitinib or BMS-911543. Furthermore, we showed that concomitant inhibition of JAK1/2 rather than blocking JAK1 or JAK2 alone, impaired maturation and the release of proinflammatory cytokines on lipopolysaccharide stimulation. CONCLUSIONS: Our results suggest that inhibition of JAK1/2 impairs IDEC differentiation and function. We provide new insight into the mode of action of JAK inhibitors in AD and highlight the role of JAK1/2 inhibitors for the treatment of patients with AD.


Subject(s)
Dermatitis, Atopic/etiology , Dermatitis, Atopic/metabolism , Epidermal Cells/drug effects , Epidermal Cells/metabolism , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Cell Differentiation , Cytokines/metabolism , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/pathology , Epidermal Cells/pathology , Gene Expression , Humans , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Protein Kinase Inhibitors/therapeutic use
6.
Biochem Biophys Res Commun ; 534: 802-807, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33162034

ABSTRACT

To overcome dermatological concerns causing abnormally excessive melanin synthesis, highly effective and safe skin depigmentation compounds have been identified in the cosmetic and pharmaceutical industries. Among several methods used to achieve skin depigmentation, inhibition of tyrosinase is one of the most effective, since tyrosinase is a crucial enzyme in melanogenesis. Herein, isolindleyin, a novel inhibitor of human tyrosinase, was introduced and evaluated for its anti-melanogenic effects in human epidermal melanocytes. The results revealed that isolindleyin was directly bound to tyrosinase and it suppressed melanin synthesis. The binding mode between isolindleyin and the active sites of human tyrosinase was investigated using computational molecular docking at the atomic level. Isolindleyin binding was found to be stabilized by hydrophobic interactions between His 367 and Val 377 and by hydrogen bonds between Ser 380 and Asn 364. The results of this study revealed the anti-melanogenic effects of isolindleyin that could contribute toward overcoming dermatological concerns that cause abnormally excessive melanin synthesis.


Subject(s)
Glucosides/pharmacology , Melanocytes/drug effects , Monophenol Monooxygenase/antagonists & inhibitors , Monophenol Monooxygenase/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Epidermal Cells/drug effects , Glucosides/chemistry , Glucosides/metabolism , Humans , Melanins/metabolism , Melanocytes/metabolism , Molecular Docking Simulation , Monophenol Monooxygenase/chemistry , Surface Plasmon Resonance
7.
Arch Toxicol ; 95(6): 2201-2221, 2021 06.
Article in English | MEDLINE | ID: mdl-33890134

ABSTRACT

Deoxynivalenol (vomitoxin, DON) is a secondary metabolite produced by Fusarium spp. fungi and it is one of the most prevalent mycotoxins worldwide. Crop infestation results not only in food and feed contamination, but also in direct dermal exposure, especially during harvest and food processing. To investigate the potential dermotoxicity of DON, epidermoid squamous cell carcinoma cells A431 were compared to primary human neonatal keratinocytes (HEKn) cells via proteome/phosphoproteome profiling. In A431 cells, 10 µM DON significantly down-regulated ribosomal proteins, as well as mitochondrial respiratory chain elements (OXPHOS regulation) and transport proteins (TOMM22; TOMM40; TOMM70A). Mitochondrial impairment was reflected in altered metabolic competence, apparently combined with interference of the lipid biosynthesis machinery. Functional effects on the cell membrane were confirmed by live cell imaging and membrane fluidity assays (0.1-10 µM DON). Moreover, a common denominator for both A431 and HEKn cells was a significant downregulation of the squalene synthase (FDFT1). In sum, proteome alterations could be traced back to the transcription factor Klf4, a crucial regulator of skin barrier function. Overall, these results describe decisive molecular events sustaining the capability of DON to impair skin barrier function. Proteome data generated in the study are fully accessible via ProteomeXchange with the accession numbers PXD011474 and PXD013613.


Subject(s)
Epidermal Cells/drug effects , Keratinocytes/drug effects , Lipids/biosynthesis , Trichothecenes/toxicity , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cells, Cultured , Dose-Response Relationship, Drug , Epidermal Cells/pathology , Fusarium/metabolism , Humans , Keratinocytes/pathology , Mitochondria/drug effects , Mitochondria/pathology , Proteomics , Secondary Metabolism , Trichothecenes/administration & dosage , Trichothecenes/isolation & purification
8.
Skin Pharmacol Physiol ; 34(2): 103-114, 2021.
Article in English | MEDLINE | ID: mdl-33721861

ABSTRACT

INTRODUCTION: Lactic fermentation products (LFPs) are thought to affect "good" bacteria in the gut. We previously reported that oral administration of LFPs has beneficial therapeutic effects in a mouse model of atopic dermatitis. However, it is unclear how LFPs affect human epidermal cell differentiation, ceramide (Cer), and amino acid production. OBJECTIVE: The aim of this study was to determine the effects of LFPs on epidermal cell differentiation, by assessing amino acid and Cer production. METHODS: A 3-dimensional cultured human epidermis model and normal human epidermal keratinocytes were used. Cytotoxicity tests were performed using alamar Blue. Transepidermal water loss (TEWL) was used as an index to assess barrier function. Keratin 1 (K1), keratin 5 (K5), keratin 10 (K10), involucrin (INV), calpain 1, and transglutaminase (TGase) (markers of differentiation) and profilaggrin (proFLG) and bleomycin hydrolase (amino acid synthesis-related genes) expression levels were quantified by RT-PCR. In addition, TGase protein levels were measured by Western blotting. The intercellular lipid content of the stratum corneum was measured by high-performance thin-layer chromatography. Amino acids were quantified using an amino acid analyzer. Finally, bound water content in the stratum corneum was measured by differential scanning calorimetry. RESULTS: Cell viability did not change, but TEWL was significantly decreased in the cells treated with LFPs compared with the control cells. Treatment with LFPs significantly increased expression of the late-differentiation markers INV and TGase at the RNA level. Furthermore, TGase protein expression was significantly increased by treatment with LFPs. Treating a 3-dimensional cultured epidermis model with LFPs significantly increased the intercellular lipid content of the stratum corneum and production of the amino acid arginine (Arg). The amount of bound water in the stratum corneum was increased significantly in the LFP application group. CONCLUSION: Treatment with LFPs promotes human epidermal cell differentiation and increases the intercellular content of the free fatty acid, Chol, Cer [NS], Cer [AS], and Cer [AP]. This may result in improved skin barrier function. The increased amount of Arg observed in keratinocytes may help improve water retention.


Subject(s)
Amino Acids/drug effects , Cell Differentiation/drug effects , Ceramides/metabolism , Epidermal Cells/drug effects , Keratinocytes/drug effects , Lactobacillales/metabolism , Amino Acids/metabolism , Cell Differentiation/physiology , Cell Survival , Epidermal Cells/metabolism , Fermentation/physiology , Gene Expression , Humans , Keratinocytes/metabolism , Lactic Acid , Water/metabolism
9.
Wound Repair Regen ; 28(2): 177-184, 2020 03.
Article in English | MEDLINE | ID: mdl-31667902

ABSTRACT

The purpose of this study was to investigate the effect of the peptide analgesic hybrid compounds: AWL3106 analog of dermorphin and substance P (7-11), and biphalin enkephalin analog on wound healing in streptozotocin-induced diabetic rats. The diabetes was induced in 6-7 week-old male Wistar rats by intraperitoneal injection of streptozotocin. After 70 days, the wounds were created on the back of the rats and then, once a day for 21 days, the dressing containing lanolin ointment, 10% of keratin scaffolds, and 1 mM of AWL3106 or biphalin was applied. The wounds histology were analyzed by hematoxylin and eosin staining. The orientation and organization of collagen was analyzed by Masson's trichome staining. The number of macrophages, blood vessels, and fibroblasts were visualized by CD68, CD34, and vimentin immunoreactivity, respectively. Our results demonstrated that the wound area of AWL3106- and biphalin-treated groups was greatly reduced (up to 47% on the 7 day) in comparison with untreated diabetic groups. The immunohistochemical staining of macrophages demonstrated that AWL3106 and biphalin accelerated inflammatory progression and subsequently decreased persistent inflammation. The histological analysis showed that the structure of tissue in the groups under the study was very similar to the one of wound tissue in N-DM group. The H&E and Masson's trichome staining demonstrated that the orientation and organization of collagen as well as the number and shape of blood vessels were better in 3106- and BIF-treated group than in DM group. In conclusion, the obtained data suggested that our hybrid peptides enhanced wound healing, particularly by accelerating the inflammatory phase and promoted the wound closure.


Subject(s)
Analgesics/pharmacology , Diabetes Mellitus, Experimental , Enkephalins/pharmacology , Macrophages/drug effects , Neovascularization, Physiologic/drug effects , Opioid Peptides/pharmacology , Peptide Fragments/pharmacology , Substance P/pharmacology , Wound Healing/drug effects , Animals , Collagen/drug effects , Collagen/metabolism , Epidermal Cells/drug effects , Fibroblasts/drug effects , Male , Rats , Rats, Wistar
10.
Wound Repair Regen ; 28(1): 16-25, 2020 01.
Article in English | MEDLINE | ID: mdl-31270882

ABSTRACT

Reepithelialization is an important step of wound healing, which is mainly completed by proliferation and migration of epidermal cells. Akermanite is a Ca-, Mg-, and Si-containing bioceramic. This study evaluated the effects of Akermanite on wound healing and investigated the mechanisms. Using scald burn mice models, we demonstrated that local Akermanite treatment significantly accelerated wound healing by increasing reepithelialization and the stemness of epidermal cells. Epidermal cells were cultured in medium containing Akermanite extracts to explore the cellular mechanism of reepithelialization. Akermanite promoted the cell proliferation and migration, maintaining more cells in the S and G2 /M phases of the cell cycle. An additional study showed that Akermanite enhanced the expressions of integrinß1, Lgr4, Lgr5, and Lgr6, which are specific molecular markers of epidermal stem cells, accompanied by the activation of the Wnt/ß-catenin pathway. These results suggested that Akermanite accelerated reepithelialization by increasing the proliferation, migration, and stemness of epidermal cells in a manner related to the Wnt/ß-catenin pathway, which might contribute, at least partially, to accelerated wound healing by Akermanite therapy.


Subject(s)
Cell Proliferation/drug effects , Ceramics/pharmacology , Epidermal Cells/drug effects , Re-Epithelialization/drug effects , Stem Cells/metabolism , Animals , Biocompatible Materials , Cell Cycle/drug effects , Cell Movement/drug effects , Cells, Cultured , Epidermal Cells/metabolism , Humans , Integrin beta1/drug effects , Integrin beta1/genetics , Integrin beta1/metabolism , Mice , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Wnt Signaling Pathway , Wound Healing
11.
Acta Biochim Biophys Sin (Shanghai) ; 52(10): 1102-1110, 2020 Oct 19.
Article in English | MEDLINE | ID: mdl-32840291

ABSTRACT

Skin epidermal stem cells (EpSCs) play an important role in wound healing. Quercetin is a phytoestrogen reported to accelerate skin wound healing, but its effect on EpSCs is unknown. In this study, we investigated the effect of quercetin on human EpSC proliferation and explored the underlying mechanisms. We found that quercetin at 0.1~1 µM significantly promoted EpSC proliferation and increased the number of cells in S phase. The pro-proliferative effect of quercetin on EpSCs was confirmed in cultured human skin tissue. Mechanistic studies showed that quercetin significantly upregulated the expressions of ß-catenin, c-Myc, and cyclins A2 and E1. Inhibitor for ß-catenin or c-Myc significantly inhibited quercetin-induced EpSC proliferation. The ß-catenin inhibitor XAV-939 suppressed quercetin-induced expressions of ß-catenin, c-Myc, and cyclins A2 and E1. The c-Myc inhibitor 10058-F4 inhibited the upregulation of c-Myc and cyclin A2 by quercetin. Pretreatment of EpSCs with estrogen receptor (ER) antagonist ICI182780, but not the G protein-coupled ER1 antagonist G15, reversed quercetin-induced cell proliferation and upregulation of ß-catenin, c-Myc, and cyclin A2. Collectively, these results indicate that quercetin promotes EpSC proliferation through ER-mediated activation of ß-catenin/c-Myc/cyclinA2 signaling pathway and ER-independent upregulation of cyclin E1 and that quercetin may accelerate skin wound healing through promoting EpSC proliferation. As EpSCs are used not only in clinic to treat skin wounds but also as seed cells in skin tissue engineering, quercetin is a useful reagent to expand EpSCs for basic research, skin wound treatment, and skin tissue engineering.


Subject(s)
Epidermal Cells/metabolism , Quercetin/pharmacology , Signal Transduction/drug effects , Stem Cells/metabolism , Adult , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cyclin A2/metabolism , Cyclin E/metabolism , Epidermal Cells/drug effects , Humans , Male , Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Estrogen/metabolism , Skin/cytology , Skin/drug effects , Skin/metabolism , Skin/pathology , Stem Cells/drug effects , beta Catenin/metabolism
12.
Int J Mol Sci ; 21(14)2020 Jul 13.
Article in English | MEDLINE | ID: mdl-32668737

ABSTRACT

The BC-box motif in suppressor of cytokine signaling 6 (SOCS6) promotes the neuronal differentiation of somatic stem cells, including epidermal stem cells. SOCS6 protein belongs to the group of SOCS proteins and inhibits cytokine signaling. Here we showed that epidermal stem cells were induced to differentiate into GABAnergic neurons by the intracellular delivery of a peptide composed of the amino-acid sequences encoded by the BC-box motif in SOCS6 protein. The BC-box motif (SLQYLCRFVI) in SOCS6 corresponded to the binding site of elongin BC. GABAnergic differentiation mediated by the BC-box motif in SOCS6 protein was caused by ubiquitination of JAK2 and inhibition of the JAK2-STAT3 pathway. Furthermore, GABAnergic neuron-like cells generated from epidermal stem cells were transplanted into the brain of a rodent ischemic model. Then, we demonstrated that these transplanted cells were GAD positive and that the cognitive function of the ischemic model rodents with the transplanted cells was improved. This study could contribute to not only elucidating the mechanism of GABAnergic neuronal differentiation but also to neuronal regenerative medicine utilizing GABAnergic neurons.


Subject(s)
Epidermal Cells/drug effects , GABAergic Neurons/cytology , Neurogenesis/drug effects , Pluripotent Stem Cells/drug effects , Suppressor of Cytokine Signaling Proteins/pharmacology , Amino Acid Motifs , Amino Acid Sequence , Animals , Cells, Cultured , Cognition Disorders/etiology , Epidermal Cells/cytology , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/surgery , Janus Kinase 2/metabolism , Microscopy, Fluorescence , Morris Water Maze Test , Patch-Clamp Techniques , Peptide Fragments/administration & dosage , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Pluripotent Stem Cells/transplantation , Protein Processing, Post-Translational , Rats , STAT3 Transcription Factor/metabolism , Signal Transduction , Suppressor of Cytokine Signaling Proteins/administration & dosage , Suppressor of Cytokine Signaling Proteins/chemistry , Ubiquitination
13.
Int J Mol Sci ; 21(22)2020 Nov 11.
Article in English | MEDLINE | ID: mdl-33187177

ABSTRACT

A human skin equivalent (HSE) composed of the epidermis and dermis is cultured using a pumpless skin-on-a-chip system to supply cultures the desired flow rate using gravity flow without a pump or an external tube connection. Coenzyme Q10 efficacy is tested by adjusting its concentration, as it is known to have anti-aging and antioxidant effects in culture solutions. The relationship between the contraction rate of a full-thickness human skin equivalent and secreted transforming growth factor (TGF) ß-1 is analyzed via enzyme-linked immunosorbent assay (ELISA). Following hematoxylin and eosin (H&E) staining, an image of the skin equivalent is analyzed to measure the epidermal layer's thickness. The cell density and differentiation of the dermis layer are investigated. Gene and protein expression in the dermal and epidermal layers are quantitatively analyzed using quantitative real time polymerase chain reaction (qPCR) and immunohistochemical staining. As the coenzyme Q10 treatment concentration increased, the number of cells per unit area and the thickness of the epidermal layer increased, the expression level of filaggrin increased, and the contraction rate of full-thickness HSE was proportional to the amount of TGF ß-1 secreted.


Subject(s)
Skin/drug effects , Ubiquinone/analogs & derivatives , Animals , Cell Count/methods , Cell Differentiation/drug effects , Cells, Cultured , Epidermal Cells/drug effects , Epidermal Cells/metabolism , Epidermis/drug effects , Epidermis/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Filaggrin Proteins , Gene Expression/drug effects , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Lab-On-A-Chip Devices , Rats , Skin/metabolism , Transforming Growth Factor beta1/metabolism , Ubiquinone/pharmacology
14.
Int J Mol Sci ; 21(3)2020 Jan 25.
Article in English | MEDLINE | ID: mdl-31991834

ABSTRACT

The ß-blocker carvedilol prevents ultraviolet (UV)-induced skin cancer, but the mechanism is unknown. Since carvedilol possesses antioxidant activity, this study investigated whether carvedilol prevents oxidative photodamage of skin, a precursor event in skin carcinogenesis. The effects of carvedilol, metoprolol (a ß-blocker without antioxidant property), and 4-hydroxycarbazole (4-OHC, a carvedilol synthesis intermediate and a free radical scavenger) were compared on UV- or H2O2-induced cell death and reactive oxygen species (ROS) production in murine epidermal JB6 P+ cells. Although carvedilol attenuated cell death, metoprolol and 4-OHC failed to show protective effects. As expected, increased cellular ROS induced by H2O2 or UV was abolished by carvedilol and 4-OHC, but not by metoprolol. Consistently, carvedilol attenuated the formation of UV-induced cyclobutane pyrimidine dimers (CPDs) and release of prostaglandin E2 in JB6 P+ cells. Carvedilol's activity was further confirmed in full thickness 3D human reconstituted skin, where carvedilol attenuated UV-mediated epidermal thickening, the number of Ki-67 and p53 positive cells as well as CPD formation. Based on pathway-specific Polymerase Chain Reaction (PCR) Array analysis, carvedilol treatment in many cases normalized UV-induced expression changes in DNA repair genes. Thus, carvedilol's photoprotective activity is not attributed to ß-blockade or direct ROS-scavenging capacity, but likely via DNA repair regulation.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Carvedilol/pharmacology , Epidermal Cells/drug effects , Epidermal Cells/radiation effects , Ultraviolet Rays/adverse effects , Animals , Cell Culture Techniques , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/radiation effects , Cytokines/metabolism , DNA Damage/drug effects , Dinoprostone/metabolism , Epidermal Cells/metabolism , Humans , Hydrogen Peroxide , Inflammation Mediators , Mice , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
15.
Molecules ; 25(23)2020 Nov 26.
Article in English | MEDLINE | ID: mdl-33256158

ABSTRACT

Rosacea is a common and chronic inflammatory skin disease that is characterized by dysfunction of the immune and vascular system. The excessive production and activation of kallikerin 5 (KLK5) and cathelicidin have been implicated in the pathogenesis of rosacea. Coptis chinensis Franch (CC) has been used as a medicinal herb in traditional oriental medicine. However, little is known about the efficacy and mechanism of action of CC in rosacea. In this study, we evaluate the effect of CC and its molecular mechanism on rosacea in human epidermal keratinocytes. CC has the capacity to downregulate the expression of KLK5 and cathelicidin, and also inhibits KLK5 protease activity, which leads to reduced processing of inactive cathelicidin into active LL-37. It was determined that CC ameliorates the expression of pro-inflammatory cytokines through the inhibition of LL-37 processing. In addition, it was confirmed that chitin, an exoskeleton of Demodex mites, mediates an immune response through TLR2 activation, and CC inhibits TLR2 expression and downstream signal transduction. Furthermore, CC was shown to inhibit the proliferation of human microvascular endothelial cells induced by LL-37, the cause of erythematous rosacea. These results demonstrate that CC improved rosacea by regulating the immune response and angiogenesis, and revealed its mechanism of action, indicating that CC may be a useful therapeutic agent for rosacea.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Coptis/chemistry , Epidermal Cells/drug effects , Epidermal Cells/metabolism , Kallikreins/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Plant Extracts/pharmacology , Cell Line , Cytokines/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Inflammation Mediators/metabolism , Models, Biological , Phytochemicals/chemistry , Phytochemicals/pharmacology , Plant Extracts/chemistry , Proteolysis , Rosacea/drug therapy , Cathelicidins
16.
J Recept Signal Transduct Res ; 39(2): 134-145, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31354004

ABSTRACT

Context: Epidermal cells play an important role in regulating the regeneration of skin after burns and wounds. Objective: The aim of our study is to explore the role of Tanshinone IIA (Tan IIA) in the apoptosis of epidermal HaCaT cells induced by H2O2, with a focus on mitochondrial homeostasis and inverted formin-2 (INF2). Materials and methods: Cellular viability was determined using the MTT assay, TUNEL staining, western blot analysis and LDH release assay. Adenovirus-loaded INF2 was transfected into HaCaT cells to overexpress INF2 in the presence of Tan IIA treatment. Mitochondrial function was determined using JC-1 staining, mitochondrial ROS staining, immunofluorescence and western blotting. Results: Oxidative stress promoted the death of HaCaT cells and this effect could be reversed by Tan IIA. At the molecular levels, Tan IIA treatment sustained mitochondrial energy metabolism, repressed mitochondrial ROS generation, stabilized mitochondrial potential, and blocked the mitochondrial apoptotic pathway. Furthermore, we demonstrated that Tan IIA modulated mitochondrial homeostasis via affecting INF2-related mitochondrial stress. Overexpression of INF2 could abolish the protective effects of Tan IIA on HaCaT cells viability and mitochondrial function. Besides, we also reported that Tan IIA regulated INF2 expression via the ERK pathway; inhibition of this pathway abrogated the beneficial effects of Tan IIA on HaCaT cells survival and mitochondrial homeostasis. Conclusions: Overall, our results indicated that oxidative stress-mediated HaCaT cells apoptosis could be reversed by Tan IIA treatment via reducing INF2-related mitochondrial stress in a manner dependent on the ERK signaling pathway.


Subject(s)
Abietanes/pharmacology , Burns/drug therapy , Formins/genetics , Oxidative Stress/drug effects , Apoptosis/drug effects , Burns/genetics , Burns/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cellular Microenvironment/drug effects , Energy Metabolism/drug effects , Epidermal Cells/drug effects , Humans , Hydrogen Peroxide/pharmacology , Keratinocytes/drug effects , Keratinocytes/metabolism , MAP Kinase Signaling System/drug effects , Mitochondria/drug effects , Mitochondria/genetics , Reactive Oxygen Species/metabolism , Skin/drug effects , Skin/injuries , Skin/pathology
17.
Biol Pharm Bull ; 42(9): 1446-1449, 2019.
Article in English | MEDLINE | ID: mdl-31474706

ABSTRACT

During the process of skin regeneration following a skin injury, de novo hair follicle regeneration is initiated after wounding; however, these regenerated hairs are mostly unpigmented. The activation of epidermal melanocyte stem cells and their differentiation into regenerating hair follicles have been shown to be necessary for the pigmented hair regeneration after wounding. To determine the role of flavonoids in the regeneration of pigmented hairs, we applied the candidate flavonoids to the regenerating hair follicles after wounding and identified the flavonoid species that maximally induced pigmented hair regeneration. Flavonoids with two OH groups in the B-ring, such as sterubin, luteolin, and hydroxygenkwanin, showed promising effects in regenerating black pigmented hairs, while those with one OH group in the B-ring showed no significant change. Thus, flavonoids with two OH groups in their B-ring could be studied further as potential wound healing agents with the ability to regenerate pigmented hair.


Subject(s)
Flavonoids/pharmacology , Hair Color , Hair Follicle/drug effects , Regeneration/drug effects , Skin/injuries , Wound Healing/drug effects , Animals , Epidermal Cells/drug effects , Epidermal Cells/physiology , Flavonoids/chemistry , Hair Follicle/physiology , Luteolin/chemistry , Luteolin/pharmacology , Melanocytes/drug effects , Melanocytes/physiology , Mice, Inbred C57BL , Skin/drug effects , Structure-Activity Relationship
18.
Biochemistry (Mosc) ; 84(9): 1047-1056, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31693464

ABSTRACT

Plant hormones produce cytotoxic effect on human cells and can trigger the processes unrelated to cell death, e.g., biosynthetic system stress. The goal of this study was to investigate activation of the endoplasmic reticulum (ER) stress by jasmonic acid (JA) and to distinguish between the responses of cultured immortalized non-tumorigenic HaCaT cells and epidermoid carcinoma A431 cells to this plant hormone. JA was used in the concentration of 2 mM, as it suppressed cell proliferation in both cell lines. We analyzed expression of genes associated with the activation of ER stress (GRP78, ATF4, CHOP), the structure of the ER and Golgi complex, and synthetic processes in the HaCaT and A431 cell lines. JA induced expression of genes responsible for the activation of ER stress and caused hypertrophic changes in the Golgi complex in both cell lines. However, the patterns of gene expression in the HaCaT and A431 cells were different, and higher levels of involucrin synthesis were observed in A431 but not in HaCaT cells, suggesting that JA activated differentiation of the tumor A431 cells only. Therefore, JA induced ER stress in both cell lines, but the consequences of ER stress were different for the epidermal immortalized non-tumorigenic and tumor cells.


Subject(s)
Cyclopentanes/pharmacology , Endoplasmic Reticulum Stress/drug effects , Epidermal Cells/drug effects , Epidermal Cells/pathology , Neoplasms/pathology , Oxylipins/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Endoplasmic Reticulum Chaperone BiP , Epidermal Cells/cytology , Humans , Neoplasms/metabolism , Structure-Activity Relationship
19.
Clin Exp Dermatol ; 44(5): 512-519, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30456849

ABSTRACT

BACKGROUND: Ultraviolet (UV)B radiation has long been considered a carcinogen in both epidemiological surveys and experimental studies. However, recent work has suggested that different dosages of UVB exert different influences on cells. There are also co-carcinogenesis factors such as arsenic that affect the role of UVB. AIM: To explore the co-carcinogenesis effect of UVB and arsenic on the mouse epidermal cell line JB6 and the mechanism underlying it. METHODS: Growth of JB6 cells was measured by MTT assay. We carried out a comet assay to determine the DNA damage caused by UVB and arsenic, and tested the expression of DNA repair protein by western blotting. Reactive oxygen species (ROS) were measured using DCF and DHE staining, and changes in antioxidant enzymes were assessed using western blotting. RESULTS: Viability assays showed that arsenic increased the UVB-induced death rate. Arsenic enhanced DNA damage caused by UVB both directly by injury to double-stranded DNA and indirectly by reducing the capability of DNA repair in JB6 cells. All of these effects are the results of increased ROS generation and reduced expression of the antioxidant enzyme superoxide dismutase (SOD)1. CONCLUSION: Arsenic was found to enhance UVB-induced production of ROS and to downregulate SOD1 expression, leading to DNA damage and apoptosis in mouse skin cells. The combination of arsenic and UVB exposure was found to differentially regulate the expression of SOD1 and SOD2.


Subject(s)
Apoptosis/drug effects , Arsenic/pharmacology , DNA Damage/drug effects , Epidermal Cells/drug effects , Reactive Oxygen Species/metabolism , Ultraviolet Rays/adverse effects , Animals , Apoptosis/radiation effects , Cell Death/drug effects , Cell Death/radiation effects , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Damage/radiation effects , Epidermal Cells/metabolism , Epidermal Cells/radiation effects , Mice , Reactive Oxygen Species/radiation effects , Superoxide Dismutase-1/drug effects , Superoxide Dismutase-1/metabolism
20.
Phytother Res ; 33(2): 422-430, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30461085

ABSTRACT

We aimed to explore the effect of curcumin on epidermal stem cells (ESCs) in regulating wound healing and the underlying molecular mechanism. We treated mouse ESCs isolated from skin tissues with curcumin, and then assessed the proliferation ability of cells induced by epidermal growth factor using cell counting kit-8 assay. The pluripotency of ESCs was evaluated as well through examination of Nanog expression in ESCs. Further, mice with skin burns were treated with ESCs with or without curcumin pretreatments. Histological evaluations were then preformed to determine wound scores, cell proliferation, reepithelialization, and capillary density in wounds. Curcumin treatment promoted the proliferative ability of ESCs and conditioned medium from curcumin-treated ESCs enhanced human umbilical vein endothelial cell (HUVEC) tube formation. We also found curcumin treatment elevated caveolin-1 expression in ESCs, which was required for the beneficial effect of curcumin on ESC proliferation and HUVEC tube formation. Next, using a mouse model of burn wound healing, curcumin-treated ESCs exhibited enhanced wound closure, which also required caveolin-1 expression. Our current study demonstrates the beneficial effect of curcumin on burn wound healing in mice, which is mediated by upregulating caveolin-1 in ESCs, and supports the potential therapeutic role of curcumin in ESC-based treatment against skin wound healing.


Subject(s)
Burns/drug therapy , Caveolin 1/genetics , Curcumin/pharmacology , Wound Healing/drug effects , Animals , Cell Proliferation/drug effects , Epidermal Cells/drug effects , Epidermis/drug effects , Female , Humans , Male , Mice , Mice, Inbred BALB C , Skin/pathology , Stem Cells/drug effects , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL