Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 8.228
1.
J Nanobiotechnology ; 22(1): 307, 2024 Jun 02.
Article En | MEDLINE | ID: mdl-38825668

Skin aging is characterized by the disruption of skin homeostasis and impaired skin injury repair. Treatment of aging skin has long been limited by the unclear intervention targets and delivery techniques. Engineering extracellular vesicles (EVs) as an upgraded version of natural EVs holds great potential in regenerative medicine. In this study, we found that the expression of the critical antioxidant and detoxification gene Gstm2 was significantly reduced in aging skin. Thus, we constructed the skin primary fibroblasts-derived EVs encapsulating Gstm2 mRNA (EVsGstm2), and found that EVsGstm2 could significantly improve skin homeostasis and accelerate wound healing in aged mice. Mechanistically, we found that EVsGstm2 alleviated oxidative stress damage of aging dermal fibroblasts by modulating mitochondrial oxidative phosphorylation, and promoted dermal fibroblasts to regulate skin epidermal cell function by paracrine secretion of Nascent Polypeptide-Associated Complex Alpha subunit (NACA). Furthermore, we confirmed that NACA is a novel skin epidermal cell protective molecule that regulates skin epidermal cell turnover through the ROS-ERK-ETS-Cyclin D pathway. Our findings demonstrate the feasibility and efficacy of EVs-mediated delivery of Gstm2 for aged skin treatment and unveil novel roles of GSTM2 and NACA for improving aging skin.


Extracellular Vesicles , Fibroblasts , Glutathione Transferase , RNA, Messenger , Skin Aging , Wound Healing , Animals , Mice , Fibroblasts/metabolism , Glutathione Transferase/metabolism , Extracellular Vesicles/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics , Epidermis/metabolism , Mice, Inbred C57BL , Oxidative Stress , Skin/metabolism , Male , Humans , Epidermal Cells/metabolism , Cells, Cultured
2.
J Nanobiotechnology ; 22(1): 312, 2024 Jun 05.
Article En | MEDLINE | ID: mdl-38840221

Zinc oxide nanoparticles (ZNPs) are widely used in sunscreens and nanomedicines, and it was recently confirmed that ZNPs can penetrate stratum corneum into deep epidermis. Therefore, it is necessary to determine the impact of ZNPs on epidermis. In this study, ZNPs were applied to mouse skin at a relatively low concentration for one week. As a result, desmosomes in epidermal tissues were depolymerized, epidermal mechanical strain resistance was reduced, and the levels of desmosomal cadherins were decreased in cell membrane lysates and increased in cytoplasmic lysates. This finding suggested that ZNPs promote desmosomal cadherin endocytosis, which causes desmosome depolymerization. In further studies, ZNPs were proved to decrease mammalian target of rapamycin complex 1 (mTORC1) activity, activate transcription factor EB (TFEB), upregulate biogenesis of lysosome-related organelle complex 1 subunit 3 (BLOC1S3) and consequently promote desmosomal cadherin endocytosis. In addition, the key role of mTORC1 in ZNP-induced decrease in mechanical strain resistance was determined both in vitro and in vivo. It can be concluded that ZNPs reduce epidermal mechanical strain resistance by promoting desmosomal cadherin endocytosis via the mTORC1-TFEB-BLOC1S3 axis. This study helps elucidate the biological effects of ZNPs and suggests that ZNPs increase the risk of epidermal fragmentation.


Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cadherins , Endocytosis , Epidermis , Mechanistic Target of Rapamycin Complex 1 , Zinc Oxide , Animals , Mechanistic Target of Rapamycin Complex 1/metabolism , Endocytosis/drug effects , Mice , Cadherins/metabolism , Epidermis/metabolism , Epidermis/drug effects , Zinc Oxide/pharmacology , Zinc Oxide/chemistry , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Desmosomes/metabolism , Nanoparticles/chemistry , Stress, Mechanical
3.
Exp Dermatol ; 33(6): e15107, 2024 Jun.
Article En | MEDLINE | ID: mdl-38840418

The skin plays an essential role in preventing the entry of external environmental threats and the loss of internal substances, depending on the epidermal permeability barrier. Nuclear receptors (NRs), present in various tissues and organs including full-thickness skin, have been demonstrated to exert significant effects on the epidermal lipid barrier. Formation of the lipid lamellar membrane and the normal proliferation and differentiation of keratinocytes (KCs) are crucial for the development of the epidermal permeability barrier and is regulated by specific NRs such as PPAR, LXR, VDR, RAR/RXR, AHR, PXR and FXR. These receptors play a key role in regulating KC differentiation and the entire process of epidermal lipid synthesis, processing and secretion. Lipids derived from sebaceous glands are influenced by NRs as well and participate in regulation of the epidermal lipid barrier. Furthermore, intricate interplay exists between these receptors. Disturbance of barrier function leads to a range of diseases, including psoriasis, atopic dermatitis and acne. Targeting these NRs with agonists or antagonists modulate pathways involved in lipid synthesis and cell differentiation, suggesting potential therapeutic approaches for dermatosis associated with barrier damage. This review focuses on the regulatory role of NRs in the maintenance and processing of the epidermal lipid barrier through their effects on skin lipid synthesis and KC differentiation, providing novel insights for drug targets to facilitate precision medicine strategies.


Cell Differentiation , Epidermis , Keratinocytes , Lipid Metabolism , Receptors, Cytoplasmic and Nuclear , Humans , Epidermis/metabolism , Keratinocytes/metabolism , Keratinocytes/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Permeability
4.
Front Immunol ; 15: 1365430, 2024.
Article En | MEDLINE | ID: mdl-38840912

The presence of the blood group H2 antigen on the membrane of red blood cells determines blood type O in individuals and this H2 antigen serves as a precursor to the A and B antigens expressed in blood types A and B, respectively. However, the specific involvement of ABH antigens in skin diseases is unknown. Therefore, we aim to investigate the expression of ABH antigens in skin tissue of patients with atopic dermatitis (AD) and MC903-induced AD-like mice. We demonstrated that the expression of ABH antigen is primarily located in the granular and horny layers of the skin in healthy control individuals. However, in patients with AD, the expression of the ABH antigen was absent or diminished in these layers, while the H2 antigen expression increased in the spinous layers of the affected skin lesions. Then, we investigated the biological function of blood group H antigen mediated by fucosyltransferase 1 (Fut1) in the skin, utilizing an AD mouse model induced by MC903 in wild-type (WT) and Fut1-knockout mice. After the application of MC903, Fut1-deficient mice, with no H2 antigen expression on their skin, exhibited more severe clinical signs, increased ear swelling, and elevated serum IgE levels compared with those of WT mice. Additionally, the MC903-induced thickening of both the epidermis and dermis was more pronounced in Fut1-deficient mice than that in WT mice. Furthermore, Fut1-deficient mice showed a significantly higher production of interleukin-4 (IL-4) and IL-6 in skin lesions compared with that of their WT counterparts. The expression of chemokines, particularly Ccl2 and Ccl8, was notably higher in Fut1-deficient mice compared with those of WT mice. The infiltration of CD4+ T cells, eosinophils, and mast cells into the lesional skin was significantly elevated in Fut1-deficient mice compared with that in WT mice. These findings demonstrate the protective role of H2 antigen expression against AD-like inflammation and highlight its potential therapeutic impact on AD through the regulation of blood group antigens.


Dermatitis, Atopic , Fucosyltransferases , Galactoside 2-alpha-L-fucosyltransferase , Mice, Knockout , Dermatitis, Atopic/immunology , Animals , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Mice , Humans , Female , Male , Disease Models, Animal , Cytokines/metabolism , Epidermis/immunology , Epidermis/pathology , Epidermis/metabolism , Adult , Mice, Inbred C57BL
5.
J Drugs Dermatol ; 23(6): 466-471, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38834224

Xerosis is experienced by almost everyone at some time in their lives and the foundation of management of dry skin (both consumer- and healthcare professional--directed) rests on the use of moisturizers. Given the wide range of available moisturizers, counseling patients about selecting the optimum moisturizer for their individual situation relies on knowledge of ingredients and formulations. Traditionally, the main focus for many moisturizers centered on the core functional and structural role of ceramides within the epidermal barrier.  However, while a key aspect of transepidermal water loss and other skin barrier functions, components other than ceramides are equally essential in increasing moisturization. The skin's natural moisturizing factors (NMFs) are a complex mixture of water-attracting compounds such as amino acids, urea, lactate, pyrrolidone carboxylic acid (PCA), and electrolytes which play a fundamental role in preserving physiologic function by regulating the water content of the stratum corneum. By facilitating water retention, NMFs contribute significantly to the suppleness, elasticity, normal desquamation, and overall integrity of the skin barrier. Incorporation of NMFs into moisturizers addresses critical deficiencies in the skin's moisture balance that exist in xerotic and atopic skin, and in many skin disorders, mitigating signs and symptoms associated with xerosis and promoting optimal skin health. The biochemical composition of NMFs and the intricate interplay with epidermal homeostasis translate to a central role in moisturizers used for prophylactic and therapeutic management of various dry skin conditions, beyond ceramides alone. J Drugs Dermatol. 2024;23(6):466-471.     doi:10.36849/JDD.8358.


Ceramides , Emollients , Water Loss, Insensible , Humans , Ceramides/administration & dosage , Water Loss, Insensible/drug effects , Emollients/administration & dosage , Skin Cream/administration & dosage , Administration, Cutaneous , Epidermis/drug effects , Epidermis/metabolism , Epidermis/physiology , Urea/administration & dosage
6.
Cells ; 13(9)2024 Apr 29.
Article En | MEDLINE | ID: mdl-38727296

Derangement of the epidermal barrier lipids and dysregulated immune responses are key pathogenic features of atopic dermatitis (AD). The Th2-type cytokines interleukin IL-4 and IL-13 play a prominent role in AD by activating the Janus Kinase/Signal Transduction and Activator of Transcription (JAK/STAT) intracellular signaling axis. This study aimed to investigate the role of JAK/STAT in the lipid perturbations induced by Th2 signaling in 3D epidermal equivalents. Tofacitinib, a low-molecular-mass JAK inhibitor, was used to screen for JAK/STAT-mediated deregulation of lipid metabolism. Th2 cytokines decreased the expression of elongases 1, 3, and 4 and serine-palmitoyl-transferase and increased that of sphingolipid delta(4)-desaturase and carbonic anhydrase 2. Th2 cytokines inhibited the synthesis of palmitoleic acid and caused depletion of triglycerides, in association with altered phosphatidylcholine profiles and fatty acid (FA) metabolism. Overall, the ceramide profiles were minimally affected. Except for most sphingolipids and very-long-chain FAs, the effects of Th2 on lipid pathways were reversed by co-treatment with tofacitinib. An increase in the mRNA levels of CPT1A and ACAT1, reduced by tofacitinib, suggests that Th2 cytokines promote FA beta-oxidation. In conclusion, pharmacological inhibition of JAK/STAT activation prevents the lipid disruption caused by the halted homeostasis of FA metabolism.


Cytokines , Janus Kinases , Lipid Metabolism , STAT Transcription Factors , Th2 Cells , Humans , Th2 Cells/metabolism , Th2 Cells/drug effects , STAT Transcription Factors/metabolism , Janus Kinases/metabolism , Cytokines/metabolism , Lipid Metabolism/drug effects , Epidermis/metabolism , Epidermis/drug effects , Signal Transduction/drug effects , Piperidines/pharmacology , Pyrimidines/pharmacology , Janus Kinase Inhibitors/pharmacology , Interleukin-4/metabolism , Fatty Acids/metabolism
7.
BMC Biotechnol ; 24(1): 28, 2024 May 04.
Article En | MEDLINE | ID: mdl-38702622

Scientists know very little about the mechanisms underlying fish skin mucus, despite the fact that it is a component of the immune system. Fish skin mucus is an important component of defence against invasive infections. Recently, Fish skin and its mucus are gaining interest among immunologists. Characterization was done on the obtained silver nanoparticles Ag combined with Clarias gariepinus catfish epidermal mucus proteins (EMP-Ag-NPs) through UV-vis, FTIR, XRD, TEM, and SEM. Ag-NPs ranged in size from 4 to 20 nm, spherical in form and the angles were 38.10°, 44.20°, 64.40°, and 77.20°, Where wavelength change after formation of EMP-Ag-NPs as indicate of dark brown, the broad band recorded at wavelength at 391 nm. Additionally, the antimicrobial, antibiofilm and anticancer activities of EMP-Ag-NPs was assessed. The present results demonstrate high activity against unicellular fungi C. albicans, followed by E. faecalis. Antibiofilm results showed strong activity against both S. aureus and P. aeruginosa pathogens in a dose-dependent manner, without affecting planktonic cell growth. Also, cytotoxicity effect was investigated against normal cells (Vero), breast cancer cells (Mcf7) and hepatic carcinoma (HepG2) cell lines at concentrations (200-6.25 µg/mL) and current results showed highly anticancer effect of Ag-NPs at concentrations 100, 5 and 25 µg/mL exhibited rounding, shrinkage, deformation and granulation of Mcf7 and HepG2 with IC50 19.34 and 31.16 µg/mL respectively while Vero cells appeared rounded at concentration 50 µg/mL and normal shape at concentration 25, 12.5 and 6.25 µg/ml with IC50 35.85 µg/mL. This study evidence the potential efficacy of biologically generated Ag-NPs as a substitute medicinal agent against harmful microorganisms. Furthermore, it highlights their inhibitory effect on cancer cell lines.


Biofilms , Catfishes , Metal Nanoparticles , Silver , Metal Nanoparticles/chemistry , Biofilms/drug effects , Biofilms/growth & development , Silver/chemistry , Silver/pharmacology , Animals , Humans , Mucus/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Vero Cells , Fish Proteins/pharmacology , Fish Proteins/chemistry , Fish Proteins/metabolism , Chlorocebus aethiops , Cell Line, Tumor , Microbial Sensitivity Tests , Pseudomonas aeruginosa/drug effects , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Candida albicans/drug effects , Epidermis/metabolism
8.
Lipids Health Dis ; 23(1): 138, 2024 May 11.
Article En | MEDLINE | ID: mdl-38734619

BACKGROUND: Skin barrier alterations play a crucial function in melasma development. Past researches have demonstrated variations in lipid content between the epidermis of melasma lesions and normal tissues, along with the varied expression of lipid-related genes in melasma. This study aimed to analyze the lipidome profiles of skin surface lipids (SSL) in patients with melasma before and after treatment to understand associated abnormalities. METHODS: Melasma was treated with tranexamic acid orally and hydroquinone cream topically. Disease was assessed using the Melasma Area and Severity Index (MASI), and the impact to life was evaluated with Melasma Quality of Life (MELASQoL) score. Epidermal melanin particles were observed using reflection confocal microscopy (RCM), whereas epidermal pigment and blood vessel morphology were observed using dermoscopy, and SSL samples were collected. Specific information regarding alterations in lipid composition was obtained through multivariate analysis of the liquid chromatography-mass spectrometry data. RESULTS: After treatment, patients with melasma exhibited decreased MASI and MELASQoL scores (P < 0.001); RCM revealed reduced melanin content in the lesions, and dermoscopy revealed fewer blood vessels. Fifteen lipid subclasses and 382 lipid molecules were identified using lipidomic assays. The expression levels of total lipids, phosphatidylcholine, and phosphatidylethanolamine in the melasma lesions decreased after treatment (P < 0.05). CONCLUSION: This study revealed alterations in the SSL composition after effective melasma treatment, suggesting a compensatory role for lipids in melasma barrier function. The mechanism involving SSL and the lipid barrier, which influences melasma's occurrence, needs further elucidation.


Hydroquinones , Lipidomics , Melanosis , Quality of Life , Humans , Melanosis/drug therapy , Female , Adult , Hydroquinones/therapeutic use , Hydroquinones/administration & dosage , Tranexamic Acid/therapeutic use , Middle Aged , Melanins/metabolism , Male , Lipids/blood , Lipids/analysis , Epidermis/metabolism , Epidermis/drug effects , Epidermis/pathology , Phosphatidylethanolamines/metabolism , Phosphatidylcholines/metabolism , Skin/pathology , Skin/drug effects , Skin/metabolism , Lipid Metabolism/drug effects
9.
Bull Exp Biol Med ; 176(5): 636-639, 2024 Mar.
Article En | MEDLINE | ID: mdl-38727953

Immunohistochemical detection of the LYVE-1 marker in healthy human full-thickness skin (the epidermis and the dermis) was carried out. LYVE-1 expression was found in the endothelium of lymphatic capillaries located in the papillary dermis, in the endothelium of larger lymphatic vessels of the reticular dermis, and in fibroblasts, which indicates their joint participation in hyaluronan metabolism. LYVE-1+ staining detected for the first time in cells of the stratum basale, the stratum spinosum, and the stratum granulosum of healthy human epidermis indicates their participation in hyaluronan metabolism and allows us to consider the spaces between epidermis cells as prelimphatics.


Epidermis , Hyaluronic Acid , Lymphatic Vessels , Skin , Vesicular Transport Proteins , Humans , Hyaluronic Acid/metabolism , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Skin/metabolism , Lymphatic Vessels/metabolism , Epidermis/metabolism , Ligands , Fibroblasts/metabolism , Dermis/metabolism , Lymphatic System/metabolism , Adult , Female , Male , Immunohistochemistry
10.
Exp Dermatol ; 33(5): e15084, 2024 May.
Article En | MEDLINE | ID: mdl-38711223

The transmembrane protein claudin-1 is critical for formation of the epidermal barrier structure called tight junctions (TJ) and has been shown to be important in multiple disease states. These include neonatal ichthyosis and sclerosing cholangitis syndrome, atopic dermatitis and various viral infections. To develop a model to investigate the role of claudin-1 in different disease settings, we used CRISPR/Cas9 to generate human immortalized keratinocyte (KC) lines lacking claudin-1 (CLDN1 KO). We then determined whether loss of claudin-1 expression affects epidermal barrier formation/function and KC differentiation/stratification. The absence of claudin-1 resulted in significantly reduced barrier function in both monolayer and organotypic cultures. CLDN1 KO cells demonstrated decreases in gene transcripts encoding the barrier protein filaggrin and the differentiation marker cytokeratin-10. Marked morphological differences were also observed in CLDN1 KO organotypic cultures including diminished stratification and reduced formation of the stratum granulosum. We also detected increased proliferative KC in the basale layer of CLDN1 KO organotypic cultures. These results further support the role of claudin-1 in epidermal barrier and suggest an additional role of this protein in appropriate stratification of the epidermis.


Cell Differentiation , Claudin-1 , Epidermis , Filaggrin Proteins , Keratinocytes , Keratinocytes/metabolism , Claudin-1/metabolism , Claudin-1/genetics , Humans , Filaggrin Proteins/metabolism , Epidermis/metabolism , Epidermis/pathology , Skin Diseases/genetics , Skin Diseases/metabolism , Tight Junctions/metabolism , Keratin-10/metabolism , Keratin-10/genetics , Gene Knockout Techniques , Cell Proliferation , CRISPR-Cas Systems
11.
Nat Commun ; 15(1): 4062, 2024 May 15.
Article En | MEDLINE | ID: mdl-38750035

The stratum corneum is the outermost skin layer with a vital role in skin barrier function. It is comprised of dead keratinocytes (corneocytes) and is known to maintain its thickness by shedding cells, although, the precise mechanisms that safeguard stratum corneum maturation and homeostasis remain unclear. Previous ex vivo studies have suggested a neutral-to-acidic pH gradient in the stratum corneum. Here, we use intravital pH imaging at single-corneocyte resolution to demonstrate that corneocytes actually undergo differentiation to develop three distinct zones in the stratum corneum, each with a distinct pH value. We identified a moderately acidic lower, an acidic middle, and a pH-neutral upper layer in the stratum corneum, with tight junctions playing a key role in their development. The upper pH neutral zone can adjust its pH according to the external environment and has a neutral pH under steady-state conditions owing to the influence of skin microbiota. The middle acidic pH zone provides a defensive barrier against pathogens. With mathematical modeling, we demonstrate the controlled protease activation of kallikrein-related peptidases on the stratum corneum surface that results in proper corneocyte shedding in desquamation. This work adds crucial information to our understanding of how stratum corneum homeostasis is maintained.


Epidermis , Homeostasis , Keratinocytes , Hydrogen-Ion Concentration , Animals , Keratinocytes/metabolism , Epidermis/metabolism , Skin/metabolism , Mice , Humans , Cell Differentiation , Tight Junctions/metabolism , Male , Female , Mice, Inbred C57BL
13.
Exp Dermatol ; 33(5): e15109, 2024 May.
Article En | MEDLINE | ID: mdl-38794812

Cornulin (CRNN) and repetin (RPTN) belong to the fused-type S100 protein family. Although these proteins have been reported to be expressed in the granular layer of the epidermis and have been suggested to be associated with barrier formation in the epidermis, their exact function remains unclear. This study examined the effects of ultraviolet B (UVB) irradiation on CRNN and RPTN expression in human skin xenotransplantation. The CRNN expression increased in the granular layer of UVB-irradiated skin 2 days after UVB irradiation compared to that in sham-irradiated skin. Interestingly, CRNN signals were observed not only in the cytoplasm, but also in the peripheral regions of granular keratinocytes. In contrast, RPTN was rarely expressed in sham-irradiated skin; however, RPTN signals were markedly increased in the granular layer of the UVB-irradiated skin. In addition, activation of ERK1/2 and STAT3 was observed in UVB-irradiated skin. Accordingly, the present study demonstrated that CRNN and RPTN are novel proteins whose expression can be increased by UVB irradiation. The activation of ERK1/2 and STAT3 may be associated with the regeneration of a UVB-damaged epidermis, and CRNN and RPTN may be induced to repair any dysfunction in the epidermal barrier during this regeneration process.


STAT3 Transcription Factor , Ultraviolet Rays , Humans , STAT3 Transcription Factor/metabolism , Transplantation, Heterologous , Keratinocytes/metabolism , Keratinocytes/radiation effects , Animals , Skin/metabolism , Skin/radiation effects , Epidermis/metabolism , Epidermis/radiation effects , Skin Transplantation , Cornified Envelope Proline-Rich Proteins/metabolism , Cornified Envelope Proline-Rich Proteins/genetics , Heterografts , S100 Proteins/metabolism , S100 Proteins/genetics , Mice
14.
Sci Rep ; 14(1): 12383, 2024 05 29.
Article En | MEDLINE | ID: mdl-38811772

Mesotrypsin, encoded by the PRSS3 gene, is a distinctive trypsin isoform renowned for its exceptional resistance to traditional trypsin inhibitors and unique substrate specificity. Within the skin epidermis, this protein primarily expresses in the upper layers of the stratified epidermis and plays a crucial role in processing pro-filaggrin (Pro-FLG). Although prior studies have partially elucidated its functions using primary cultured keratinocytes, challenges persist due to these cells' differentiation-activated cell death program. In the present study, HaCaT keratinocytes, characterized by minimal endogenous mesotrypsin expression and sustained proliferation in differentiated states, were utilized to further scrutinize the function of mesotrypsin. Despite the ready degradation of the intact form of active mesotrypsin in these cells, fusion with Venus, flanked by a peptide linker, enables evasion from the protein elimination machinery, thus facilitating activation of the Pro-FLG processing system. Inducing Venus-mesotrypsin expression in the cells resulted in a flattened phenotype and reduced proliferative capacity. Moreover, these cells displayed altered F-actin assembly, enhanced E-cadherin adhesive activity, and facilitated tight junction formation without overtly influencing epidermal differentiation. These findings underscore mesotrypsin's potentially pivotal role in shaping the characteristic cellular morphology of upper epidermal layers.


Cadherins , Cell Differentiation , Cell Proliferation , Filaggrin Proteins , Keratinocytes , Trypsin , Keratinocytes/metabolism , Humans , Trypsin/metabolism , Filaggrin Proteins/metabolism , Cadherins/metabolism , Epidermis/metabolism , Actins/metabolism , HaCaT Cells , Tight Junctions/metabolism , Cell Adhesion , Cell Line , Epidermal Cells/metabolism
15.
Int J Mol Sci ; 25(10)2024 May 09.
Article En | MEDLINE | ID: mdl-38791217

The dermal-epidermal junction (DEJ) is essential for maintaining skin structural integrity and regulating cell survival and proliferation. Thus, DEJ rejuvenation is key for skin revitalization, particularly in age-related DEJ deterioration. Radiofrequency (RF) treatment, known for its ability to enhance collagen fiber production through thermal mechanisms and increase heat shock protein (HSP) expression, has emerged as a promising method for skin rejuvenation. Additionally, RF activates Piezo1, an ion channel implicated in macrophage polarization toward an M2 phenotype and enhanced TGF-ß production. This study investigated the impact of RF treatment on HSP47 and HSP90 expression, known stimulators of DEJ protein expression. Furthermore, using in vitro and aged animal skin models, we assessed whether RF-induced Piezo1 activation and the subsequent M2 polarization could counter age-related DEJ changes. The RF treatment of H2O2-induced senescent keratinocytes upregulated the expression of HSP47, HSP90, TGF-ß, and DEJ proteins, including collagen XVII. Similarly, the RF treatment of senescent macrophages increased Piezo1 and CD206 (M2 marker) expression. Conditioned media from RF-treated senescent macrophages enhanced the expression of TGF-ß and DEJ proteins, such as nidogen and collagen IV, in senescent fibroblasts. In aged animal skin, RF treatment increased the expression of HSP47, HSP90, Piezo1, markers associated with M2 polarization, IL-10, and TGF-ß. Additionally, RF treatment enhanced DEJ protein expression. Moreover, RF reduced lamina densa replication, disrupted lesions, promoted hemidesmosome formation, and increased epidermal thickness. Overall, RF treatment effectively enhanced DEJ protein expression and mitigated age-related DEJ structural changes by increasing HSP levels and activating Piezo1.


Epidermis , Animals , Epidermis/metabolism , Epidermis/radiation effects , Mice , Dermis/metabolism , Keratinocytes/metabolism , Macrophages/metabolism , Skin Aging/radiation effects , Skin/metabolism , Skin/radiation effects , Skin/pathology , Humans , Aging/metabolism , Transforming Growth Factor beta/metabolism , HSP90 Heat-Shock Proteins/metabolism , HSP47 Heat-Shock Proteins/metabolism , HSP47 Heat-Shock Proteins/genetics
16.
Int J Mol Sci ; 25(10)2024 May 10.
Article En | MEDLINE | ID: mdl-38791225

Epidermal melanin synthesis determines an individual's skin color. In humans, melanin is formed by melanocytes within the epidermis. The process of melanin synthesis strongly depends on a range of cellular factors, including the fine-tuned interplay with reactive oxygen species (ROS). In this context, a role of cold atmospheric plasma (CAP) on melanin synthesis was proposed due to its tunable ROS generation. Herein, the argon-driven plasma jet kINPen® MED was employed, and its impact on melanin synthesis was evaluated by comparison with known stimulants such as the phosphodiesterase inhibitor IBMX and UV radiation. Different available model systems were employed, and the melanin content of both cultured human melanocytes (in vitro) and full-thickness human skin biopsies (in situ) were analyzed. A histochemical method detected melanin in skin tissue. Cellular melanin was measured by NIR autofluorescence using flow cytometry, and a highly sensitive HPLC-MS method was applied, which enabled the differentiation of eu- and pheomelanin by their degradation products. The melanin content in full-thickness human skin biopsies increased after repeated CAP exposure, while there were only minor effects in cultured melanocytes compared to UV radiation and IBMX treatment. Based on these findings, CAP does not appear to be a useful option for treating skin pigmentation disorders. On the other hand, the risk of hyperpigmentation as an adverse effect of CAP application for wound healing or other dermatological diseases seems to be neglectable.


Epidermis , Melanins , Melanocytes , Plasma Gases , Humans , Melanins/metabolism , Melanins/biosynthesis , Melanocytes/metabolism , Melanocytes/drug effects , Plasma Gases/pharmacology , Epidermis/metabolism , Epidermis/drug effects , Epidermis/radiation effects , Ultraviolet Rays , Skin Pigmentation/drug effects , Skin Pigmentation/radiation effects , Cells, Cultured , Reactive Oxygen Species/metabolism , Biopsy , Melanogenesis
17.
Int J Mol Sci ; 25(9)2024 May 03.
Article En | MEDLINE | ID: mdl-38732212

The skin wound healing process consists of hemostatic, inflammatory, proliferative, and maturation phases, with a complex cellular response by multiple cell types in the epidermis, dermis, and immune system. Magnesium is a mineral essential for life, and although magnesium treatment promotes cutaneous wound healing, the molecular mechanism and timing of action of the healing process are unknown. This study, using human epidermal-derived HaCaT cells and human normal epidermal keratinocyte cells, was performed to investigate the mechanism involved in the effect of magnesium on wound healing. The expression levels of epidermal differentiation-promoting factors were reduced by MgCl2, suggesting an inhibitory effect on epidermal differentiation in the remodeling stage of the late wound healing process. On the other hand, MgCl2 treatment increased the expression of matrix metalloproteinase-7 (MMP7), a cell migration-promoting factor, and enhanced cell migration via the MEK/ERK pathway activation. The enhancement of cell migration by MgCl2 was inhibited by MMP7 knockdown, suggesting that MgCl2 enhances cell migration which is mediated by increased MMP7 expression. Our results revealed that MgCl2 inhibits epidermal differentiation but promotes cell migration, suggesting that applying magnesium to the early wound healing process could be beneficial.


Cell Differentiation , Cell Movement , Keratinocytes , Magnesium , Matrix Metalloproteinase 7 , Wound Healing , Wound Healing/drug effects , Humans , Cell Movement/drug effects , Keratinocytes/drug effects , Keratinocytes/metabolism , Cell Differentiation/drug effects , Magnesium/pharmacology , Magnesium/metabolism , Matrix Metalloproteinase 7/metabolism , Matrix Metalloproteinase 7/genetics , Skin/metabolism , Skin/drug effects , Skin/injuries , MAP Kinase Signaling System/drug effects , Cell Line , Epidermis/drug effects , Epidermis/metabolism , Magnesium Chloride/pharmacology
18.
Clin Exp Pharmacol Physiol ; 51(7): e13874, 2024 Jul.
Article En | MEDLINE | ID: mdl-38797519

Glycolysis is vital for the excessive proliferation of keratinocytes in psoriasis, and uridine phosphorylase-1 (UPP1) functions as an enhancer of cancer cell proliferation. However, little is known about whether UPP1 promotes keratinocyte proliferation and accelerates psoriasis development. This study revealed that UPP1 facilitates cell viability and cell-cycle progression in human epidermal keratinocytes (HEKs) by modulating the glycolytic pathway. Bioinformatics analysis of UPP1 gene expression and its correlation with the Reactome revealed that UPP1 mRNA expression, cell-cycle progression, the interleukin-6 (IL-6)/Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) pathway and glycolysis were positively associated with psoriasis. Cell proliferation, the cell cycle and glycolysis were evaluated after UPP1 was silenced or overexpressed. The results showed that UPP1 overexpression increased cell proliferation, cell-cycle progression and glycolysis, which was contrary to the effects of UPP1 silencing. However, the STAT3 inhibitor diminished UPP1 expression because STAT3 can bind to the UPP1 promoter. In conclusion, UPP1 was significantly activated by the IL-6/STAT3 pathway and could modulate glycolysis to regulate cell proliferation and cell-cycle progression in keratinocytes during the development of psoriasis.


Cell Cycle , Cell Survival , Glycolysis , Keratinocytes , STAT3 Transcription Factor , Uridine Phosphorylase , Humans , Cell Proliferation , Epidermis/metabolism , Epidermis/pathology , Interleukin-6/metabolism , Interleukin-6/genetics , Keratinocytes/metabolism , Psoriasis/pathology , Psoriasis/metabolism , Psoriasis/genetics , Signal Transduction , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Uridine Phosphorylase/metabolism , Uridine Phosphorylase/genetics
19.
PLoS One ; 19(5): e0302781, 2024.
Article En | MEDLINE | ID: mdl-38713650

Atopic dermatitis is a multi-pathogenic disease characterized by chronic skin inflammation and barrier dysfunction. Therefore, improving the skin's ability to form an epidermal barrier and suppressing the production of cytokines that induce type 2 inflammatory responses are important for controlling atopic dermatitis symptoms. (-)-Blebbistatin, a non-muscle myosin II inhibitor, has been suggested to improve pulmonary endothelial barrier function and control inflammation by suppressing immune cell migration; however, its efficacy in atopic dermatitis is unknown. In this study, we investigated whether (S)-(-)-blebbistatin O-benzoate, a derivative of (-)-blebbistatin, improves dermatitis symptoms in a mite antigen-induced atopic dermatitis model using NC/Nga mice. The efficacy of the compound was confirmed using dermatitis scores, ear thickness measurements, serum IgE levels, histological analysis of lesions, and filaggrin expression analysis, which is important for barrier function. (S)-(-)-Blebbistatin O-benzoate treatment significantly reduced the dermatitis score and serum IgE levels compared to those in the vehicle group (p < 0.05). Furthermore, the histological analysis revealed enhanced filaggrin production and a decreased number of mast cells (p < 0.05), indicating that (S)-(-)-blebbistatin O-benzoate improved atopic dermatitis symptoms in a pathological model. In vitro analysis using cultured keratinocytes revealed increased expression of filaggrin, loricrin, involucrin, and ceramide production pathway-related genes, suggesting that (S)-(-)-blebbistatin O-benzoate promotes epidermal barrier formation. Furthermore, the effect of (S)-(-)-blebbistatin O-benzoate on type 2 alarmin cytokines, which are secreted from epidermal cells upon scratching or allergen stimulation and are involved in the pathogenesis of atopic dermatitis, was evaluated using antigens derived from mite feces. The results showed that (S)-(-)-blebbistatin O-benzoate inhibited the upregulation of these cytokines. Based on the above, (S)-(-)-blebbistatin O-benzoate has the potential to be developed as an atopic dermatitis treatment option that controls dermatitis symptoms by suppressing inflammation and improving barrier function by acting on multiple aspects of the pathogenesis of atopic dermatitis.


Benzoates , Cytokines , Dermatitis, Atopic , Epidermis , Filaggrin Proteins , Heterocyclic Compounds, 4 or More Rings , Animals , Humans , Male , Mice , Antigens, Dermatophagoides/immunology , Benzoates/pharmacology , Benzoates/therapeutic use , Cytokines/metabolism , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/pathology , Dermatitis, Atopic/metabolism , Disease Models, Animal , Epidermis/drug effects , Epidermis/metabolism , Epidermis/pathology , Filaggrin Proteins/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Immunoglobulin E/blood , Intermediate Filament Proteins/metabolism , Intermediate Filament Proteins/genetics , Keratinocytes/drug effects , Keratinocytes/metabolism , Alarmins/drug effects
20.
J Phys Chem B ; 128(22): 5378-5386, 2024 Jun 06.
Article En | MEDLINE | ID: mdl-38805566

The extracellular lipid matrix in the stratum corneum (SC) plays a critical role in skin barrier functionality, comprising three primary components: ceramides, cholesterol, and free fatty acids. The diverse ceramides, differentiated by molecular structures such as hydroxylations and varying chain lengths, are essential for the lipid matrix's structural integrity. Recently, a new subclass of ceramide, 1-O-acylceramide NP (CerENP), has been identified; however, its precise role in the lipid matrix of the SC is still elusive. Herein, we investigate the role of CerENP on the structure and permeability of the SC using molecular dynamics simulations. Our findings indicate that CerENP contributes to a compact lipid matrix in the lateral dimension of our SC model with a repeat distance of about 13 nm. Additionally, ethanol permeability assessments show that CerENP effectively reduces molecular penetration through the lipid matrix. This study provides an insight into the role of a new subclass of ceramide in the SC, enhancing our understanding of skin structure and the mechanisms behind barrier dysfunction in skin diseases.


Ceramides , Molecular Dynamics Simulation , Ceramides/chemistry , Epidermis/metabolism , Epidermis/chemistry , Permeability , Humans , Skin/metabolism , Skin/chemistry , Lipids/chemistry , Ethanol/chemistry
...