Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31.004
Filter
1.
Acta Derm Venereol ; 104: adv40601, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39101340

ABSTRACT

Keratinocyte-derived skin cancers comprise basal cell carcinoma, squamous cell carcinoma, its precursor actinic keratosis, and Bowen's disease. Historically, this group of neoplasms has been subsumed under the term non-melanoma skin cancer. However, the term non-melanoma skin cancer can be misleading and lacks precision. Therefore, more precise and reasonable terminology, valuing the relevance of keratinocyte-derived cancer, appears pertinent to meet its clinical and scientific significance. A group of experienced dermato-oncologists initiated a consensus approach to promote the use of the term "keratinocyte cancer" instead of "non-melanoma skin cancer" when referring to carcinomas and their precursors that are derived from keratinocytes. The vote among members of the consensus group indicated unanimous agreement on the consistent use of the term "keratinocyte cancer" instead of "non-melanoma skin cancer". International delegates also voted in favour of the revised terminology. The more precise and, by means of etiopathogenesis, correct term "keratinocyte cancer" should be consistently used for malignancies originated from keratinocytes. This is expected to have a positive impact on patient-physician communication and gives better justice to this important group of keratinocyte-derived cancers.


Subject(s)
Carcinoma, Basal Cell , Carcinoma, Squamous Cell , Consensus , Keratinocytes , Keratosis, Actinic , Skin Neoplasms , Terminology as Topic , Humans , Skin Neoplasms/pathology , Keratinocytes/pathology , Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/pathology , Keratosis, Actinic/pathology , Keratosis, Actinic/diagnosis , Bowen's Disease/pathology , Europe
2.
Virol J ; 21(1): 172, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095779

ABSTRACT

Human papillomavirus (HPV) 11/16 E6/E7 proteins have been recognized to be pivotal in viral pathogenesis. This study sought to uncover the potential mechanisms of how HPV11/16 E6/E7-transfected keratinocytes inhibit cytokine secretion in peripheral blood mononuclear cells (PBMC). Upon co-culturing HPV11/16 E6/E7-transfected keratinocytes with PBMC in a non-contact manner, we observed a marked decrease in various cytokines secreted by PBMC. To determine if this suppression was mediated by specific common secreted factors, we conducted transcriptomic sequencing on these transfected cells. This analysis identified 53 common differentially secreted genes in all four HPV-transfected cells. Bioinformatics analysis demonstrated these genes were predominantly involved in immune regulation. Results from quantitative PCR (qPCR) and an extensive literature review suggested the downregulation of 12 genes (ACE2, BMP3, BPIFB1, CLU, CST6, CTF1, HMGB2, MMP12, PDGFA, RNASE7, SULF2, TGM2), and upregulation of 7 genes (CCL17, CCL22, FBLN1, PLAU, S100A7, S100A8, S100A9), may be crucial in modulating tumor immunity and combating pathogenic infections, with genes S100A8 and S100A9, and IL-17 signaling pathway being particularly noteworthy. Thus, HPV11/16 E6/E7 proteins may inhibit cytokine secretion of immune cells by altering the expression of host-secreted genes. Further exploration of these genes may yield new insights into the complex dynamics of HPV infection.


Subject(s)
Cytokines , Leukocytes, Mononuclear , Oncogene Proteins, Viral , Humans , Cytokines/metabolism , Cytokines/genetics , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Leukocytes, Mononuclear/metabolism , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Oncogene Proteins, Viral/immunology , Keratinocytes/virology , Keratinocytes/immunology , Keratinocytes/metabolism , Human papillomavirus 16/genetics , Human papillomavirus 16/immunology , Human papillomavirus 11/genetics , Human papillomavirus 11/immunology , Gene Expression Profiling , Papillomavirus Infections/virology , Papillomavirus Infections/immunology , Papillomavirus Infections/genetics , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/metabolism , Papillomavirus E7 Proteins/immunology , Coculture Techniques , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics
3.
J Biomed Opt ; 29(8): 086003, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39099678

ABSTRACT

Significance: Accurate identification of epidermal cells on reflectance confocal microscopy (RCM) images is important in the study of epidermal architecture and topology of both healthy and diseased skin. However, analysis of these images is currently done manually and therefore time-consuming and subject to human error and inter-expert interpretation. It is also hindered by low image quality due to noise and heterogeneity. Aim: We aimed to design an automated pipeline for the analysis of the epidermal structure from RCM images. Approach: Two attempts have been made at automatically localizing epidermal cells, called keratinocytes, on RCM images: the first is based on a rotationally symmetric error function mask, and the second on cell morphological features. Here, we propose a dual-task network to automatically identify keratinocytes on RCM images. Each task consists of a cycle generative adversarial network. The first task aims to translate real RCM images into binary images, thus learning the noise and texture model of RCM images, whereas the second task maps Gabor-filtered RCM images into binary images, learning the epidermal structure visible on RCM images. The combination of the two tasks allows one task to constrict the solution space of the other, thus improving overall results. We refine our cell identification by applying the pre-trained StarDist algorithm to detect star-convex shapes, thus closing any incomplete membranes and separating neighboring cells. Results: The results are evaluated both on simulated data and manually annotated real RCM data. Accuracy is measured using recall and precision metrics, which is summarized as the F 1 -score. Conclusions: We demonstrate that the proposed fully unsupervised method successfully identifies keratinocytes on RCM images of the epidermis, with an accuracy on par with experts' cell identification, is not constrained by limited available annotated data, and can be extended to images acquired using various imaging techniques without retraining.


Subject(s)
Epidermis , Keratinocytes , Microscopy, Confocal , Humans , Microscopy, Confocal/methods , Epidermis/diagnostic imaging , Keratinocytes/cytology , Image Processing, Computer-Assisted/methods , Algorithms , Epidermal Cells , Neural Networks, Computer , Unsupervised Machine Learning
4.
Cell Commun Signal ; 22(1): 386, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090602

ABSTRACT

BACKGROUND: T-LAK cell-oriented protein kinase (TOPK) strongly promotes the malignant proliferation of cancer cells and is recognized as a promising biomarker of tumor progression. Psoriasis is a common inflammatory skin disease featured by excessive proliferation of keratinocytes. Although we have previously reported that topically inhibiting TOPK suppressed psoriatic manifestations in psoriasis-like model mice, the exact role of TOPK in psoriatic inflammation and the underlying mechanism remains elusive. METHODS: GEO datasets were analyzed to investigate the association of TOPK with psoriasis. Skin immunohistochemical (IHC) staining was performed to clarify the major cells expressing TOPK. TOPK conditional knockout (cko) mice were used to investigate the role of TOPK-specific deletion in IMQ-induced psoriasis-like dermatitis in mice. Flow cytometry was used to analyze the alteration of psoriasis-related immune cells in the lesional skin. Next, the M5-induced psoriasis cell model was used to identify the potential mechanism by RNA-seq, RT-RCR, and western blotting. Finally, the neutrophil-neutralizing antibody was used to confirm the relationship between TOPK and neutrophils in psoriasis-like dermatitis in mice. RESULTS: We found that TOPK levels were strongly associated with the progression of psoriasis. TOPK was predominantly increased in the epidermal keratinocytes of psoriatic lesions, and conditional knockout of TOPK in keratinocytes suppressed neutrophils infiltration and attenuated psoriatic inflammation. Neutrophils deletion by neutralizing antibody greatly diminished the suppressive effect of TOPK cko in psoriasis-like dermatitis in mice. In addition, topical application of TOPK inhibitor OTS514 effectively attenuated already-established psoriasis-like dermatitis in mice. Mechanismly, RNA-seq revealed that TOPK regulated the expression of some genes in the IL-17 signaling pathway, such as neutrophils chemokines CXCL1, CXCL2, and CXCL8. TOPK modulated the expression of neutrophils chemokines via activating transcription factors STAT3 and NF-κB p65 in keratinocytes, thereby promoting neutrophils infiltration and psoriasis progression. CONCLUSIONS: This study identified a crucial role of TOPK in psoriasis by regulating neutrophils infiltration, providing new insights into the pathogenesis of psoriasis.


Subject(s)
Keratinocytes , Mitogen-Activated Protein Kinase Kinases , Neutrophil Infiltration , Psoriasis , Animals , Humans , Mice , Disease Models, Animal , Disease Progression , Imiquimod , Keratinocytes/pathology , Keratinocytes/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Neutrophils/pathology , Psoriasis/pathology , Psoriasis/genetics , Signal Transduction , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Up-Regulation , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism
5.
Sci Adv ; 10(27): eado2365, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38959302

ABSTRACT

Pityriasis rubra pilaris (PRP) is a rare inflammatory skin disease with a poorly understood pathogenesis. Through a molecularly driven precision medicine approach and an extensive mechanistic pathway analysis in PRP skin samples, compared to psoriasis, atopic dermatitis, healed PRP, and healthy controls, we identified IL-1ß as a key mediator, orchestrating an NF-κB-mediated IL-1ß-CCL20 axis, including activation of CARD14 and NOD2. Treatment of three patients with the IL-1 antagonists anakinra and canakinumab resulted in rapid clinical improvement and reversal of the PRP-associated molecular signature with a 50% improvement in skin lesions after 2 to 3 weeks. This transcriptional signature was consistent with in vitro stimulation of keratinocytes with IL-1ß. With the central role of IL-1ß underscoring its potential as a therapeutic target, our findings propose a redefinition of PRP as an autoinflammatory keratinization disorder. Further clinical trials are needed to validate the efficacy of IL-1ß antagonists in PRP.


Subject(s)
Antibodies, Monoclonal, Humanized , Interleukin 1 Receptor Antagonist Protein , Interleukin-1beta , Keratinocytes , Pityriasis Rubra Pilaris , Humans , Pityriasis Rubra Pilaris/drug therapy , Pityriasis Rubra Pilaris/pathology , Pityriasis Rubra Pilaris/genetics , Interleukin-1beta/metabolism , Interleukin-1beta/antagonists & inhibitors , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Keratinocytes/metabolism , Keratinocytes/drug effects , Keratinocytes/pathology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Male , NF-kappa B/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Female , CARD Signaling Adaptor Proteins/metabolism , CARD Signaling Adaptor Proteins/genetics , Skin/pathology , Skin/metabolism , Skin/drug effects , Interleukin-1/antagonists & inhibitors , Interleukin-1/metabolism , Interleukin-1/genetics , Middle Aged , Guanylate Cyclase/metabolism , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/genetics , Adult , Signal Transduction/drug effects , Membrane Proteins
6.
Int J Mol Sci ; 25(14)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-39062816

ABSTRACT

To assess the effects of hydroxysafflor yellow A (HSYA) on ultraviolet A (UVA)-induced damage in HaCaT keratinocytes. HaCaT keratinocytes were UVA-irradiated, and the effects of HSYA on cell viability, reactive oxygen species (ROS) generation, lipid peroxidation, and messenger (m)RNA expression were measured. mRNA expressions of matrix metalloproteinase (MMP)-1, MMP-2, MMP-9, and cyclooxygenase (COX)-2 were determined by a real-time polymerase chain reaction (RT-PCR). UVA exposure led to a decrease in cell viability and an increase in ROS generation in HaCaT keratinocytes. HSYA effectively increased the viability of HaCaT keratinocytes after UVA exposure and protected them from UVA-induced oxidative stress. Moreover, HSYA inhibited expressions of MMP-1, MMP-2, MMP-9, and COX-2 by HaCaT keratinocytes with UVA-induced photodamage. Our results suggest that HSYA can act as a free radical scavenger when keratinocytes are photodamaged. HSYA has the potential to be a skin-protective ingredient against UVA-induced photodamage.


Subject(s)
Cell Survival , Chalcone , HaCaT Cells , Keratinocytes , Quinones , Reactive Oxygen Species , Ultraviolet Rays , Humans , Quinones/pharmacology , Ultraviolet Rays/adverse effects , Keratinocytes/drug effects , Keratinocytes/radiation effects , Keratinocytes/metabolism , Chalcone/pharmacology , Chalcone/analogs & derivatives , Cell Survival/drug effects , Cell Survival/radiation effects , Reactive Oxygen Species/metabolism , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Lipid Peroxidation/drug effects , Cell Line , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/genetics
7.
Int J Mol Sci ; 25(14)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39062847

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) and their extracellular vesicles (MSC-EVs) have been described to have important roles in tissue regeneration, including tissue repair, control of inflammation, enhancing angiogenesis, and regulating extracellular matrix remodeling. MSC-EVs have many advantages for use in regeneration therapies such as facility for dosage, histocompatibility, and low immunogenicity, thus possessing a lower possibility of rejection. In this work, we address the potential activity of MSC-EVs isolated from adipose-derived MSCs (ADMSC-EVs) cultured on cross-linked dextran microcarriers, applied to test the scalability and reproducibility of EV production. Isolated ADMSC-EVs were added into cultured human dermal fibroblasts (NHDF-1), keratinocytes (HaCat), endothelial cells (HUVEC), and THP-1 cell-derived macrophages to evaluate cellular responses (i.e., cell proliferation, cell migration, angiogenesis induction, and macrophage phenotype-switching). ADMSC viability and phenotype were assessed during cell culture and isolated ADMSC-EVs were monitored by nanotracking particle analysis, electron microscopy, and immunophenotyping. We observed an enhancement of HaCat proliferation; NHDF-1 and HaCat migration; endothelial tube formation on HUVEC; and the expression of inflammatory cytokines in THP-1-derived macrophages. The increased expression of TGF-ß and IL-1ß was observed in M1 macrophages treated with higher doses of ADMSC-EVs. Hence, EVs from microcarrier-cultivated ADMSCs are shown to modulate cell behavior, being able to induce skin tissue related cells to migrate and proliferate as well as stimulate angiogenesis and cause balance between pro- and anti-inflammatory responses in macrophages. Based on these findings, we suggest that the isolation of EVs from ADMSC suspension cultures makes it possible to induce in vitro cellular responses of interest and obtain sufficient particle numbers for the development of in vivo concept tests for tissue regeneration studies.


Subject(s)
Cell Proliferation , Extracellular Vesicles , Macrophages , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Extracellular Vesicles/metabolism , Macrophages/metabolism , Macrophages/cytology , Cell Movement , THP-1 Cells , Fibroblasts/metabolism , Fibroblasts/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Cell Culture Techniques/methods , Cells, Cultured , Keratinocytes/metabolism , Keratinocytes/cytology , Cytokines/metabolism
8.
Int J Mol Sci ; 25(14)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39062857

ABSTRACT

The delay in wound healing caused by chronic wounds or pathological scars is a pressing issue in clinical practice, imposing significant economic and psychological burdens on patients. In particular, with the aging of the population and the increasing incidence of diseases such as diabetes, impaired wound healing is one of the growing health problems. MicroRNA (miRNA) plays a crucial role in wound healing and regulates various biological processes. Our results show that miR-618 was significantly upregulated during the inflammatory phase of wound healing.Subsequently, miR-618 promotes the secretion of pro-inflammatory cytokines and regulates the proliferation and migration of keratinocytes. Mechanistically, miR-618 binds to the target gene-Atp11b and inhibits the PI3K-Akt signaling pathway, inhibiting the epithelial-mesenchymal transition (EMT) of keratinocytes. In addition, the PI3K-Akt signaling pathway induces the enrichment of nuclear miR-618, and miR-618 binds to the promoter of Lin7a to regulate gene transcription. Intradermal injection of miR-618 antagomir around full-thickness wounds in peridermal mice effectively accelerates wound closure compared to control. In conclusion, miR-618 antagomir can be a potential therapeutic agent for wound healing.


Subject(s)
Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Keratinocytes , MicroRNAs , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Wound Healing , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Keratinocytes/metabolism , Wound Healing/genetics , Mice , Cell Movement/genetics , Phosphatidylinositol 3-Kinases/metabolism , Epithelial-Mesenchymal Transition/genetics , Proto-Oncogene Proteins c-akt/metabolism , Humans , Antagomirs/metabolism , Antagomirs/pharmacology
9.
Int J Mol Sci ; 25(14)2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39062960

ABSTRACT

Human papillomavirus (HPV) infection poses a significant health challenge, particularly in low- and middle-income countries (LMIC), where limited healthcare access and awareness hinder vaccine accessibility. To identify alternative HPV targeting interventions, we previously reported on surfactant protein A (SP-A) as a novel molecule capable of recognising HPV16 pseudovirions (HPV16-PsVs) and reducing infection in a murine cervicovaginal HPV challenge model. Building on these findings, our current study aimed to assess SP-A's suitability as a broad-spectrum HPV-targeting molecule and its impact on innate immune responses. We demonstrate SP-A's ability to agglutinate and opsonise multiple oncogenic HPV-PsVs types, enhancing their uptake and clearance by RAW264.7 murine macrophages and THP-1 human-derived immune cells. The SP-A opsonisation of HPV not only led to increased lysosomal accumulation in macrophages and HaCaT keratinocytes but also resulted in a decreased infection of HaCaT cells, which was further decreased when co-cultured with innate immune cells. An analysis of human innate immune cell cytokine profiles revealed a significant inflammatory response upon SP-A exposure, potentially contributing to the overall inhibition of HPV infection. These results highlight the multi-layered impact of SP-A on HPV, innate immune cells and keratinocytes and lay the basis for the development of alternative prophylactic interventions against diverse HPV types.


Subject(s)
Macrophages , Papillomavirus Infections , Pulmonary Surfactant-Associated Protein A , Humans , Animals , Mice , Papillomavirus Infections/prevention & control , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactant-Associated Protein A/immunology , RAW 264.7 Cells , Macrophages/immunology , Macrophages/metabolism , Immunity, Innate , Keratinocytes/metabolism , Keratinocytes/virology , Keratinocytes/immunology , Cytokines/metabolism , HaCaT Cells , THP-1 Cells , Female
10.
Int J Mol Sci ; 25(14)2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39063153

ABSTRACT

Difamilast, a phosphodiesterase 4 (PDE4) inhibitor, has been shown to be effective in the treatment of atopic dermatitis (AD), although the mechanism involved remains unclear. Since IL-33 plays an important role in the pathogenesis of AD, we investigated the effect of difamilast on IL-33 activity. Since an in vitro model of cultured normal human epidermal keratinocytes (NHEKs) has been utilized to evaluate the pharmacological potential of adjunctive treatment of AD, we treated NHEKs with difamilast and analyzed the expression of the suppression of tumorigenicity 2 protein (ST2), an IL-33 receptor with transmembrane (ST2L) and soluble (sST2) isoforms. Difamilast treatment increased mRNA and protein levels of sST2, a decoy receptor suppressing IL-33 signal transduction, without affecting ST2L expression. Furthermore, supernatants from difamilast-treated NHEKs inhibited IL-33-induced upregulation of TNF-α, IL-5, and IL-13 in KU812 cells, a basophil cell line sensitive to IL-33. We also found that difamilast activated the aryl hydrocarbon receptor (AHR)-nuclear factor erythroid 2-related factor 2 (NRF2) axis. Additionally, the knockdown of AHR or NRF2 abolished the difamilast-induced sST2 production. These results indicate that difamilast treatment produces sST2 via the AHR-NRF2 axis, contributing to improving AD symptoms by inhibiting IL-33 activity.


Subject(s)
Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Keratinocytes , NF-E2-Related Factor 2 , Phosphodiesterase 4 Inhibitors , Receptors, Aryl Hydrocarbon , Signal Transduction , Humans , NF-E2-Related Factor 2/metabolism , Keratinocytes/metabolism , Keratinocytes/drug effects , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-1 Receptor-Like 1 Protein/genetics , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Phosphodiesterase 4 Inhibitors/pharmacology , Interleukin-33/metabolism , Signal Transduction/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line
11.
Biomed Pharmacother ; 177: 117129, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39018874

ABSTRACT

Atopic dermatitis (AD) is a chronic inflammatory skin condition with complex causes involving immune factors. The presence of essential trace elements that support immune system function can influence the development of this condition. This study investigated how serum trace elements impact the pathogenesis of atopic dermatitis. Upon analyzing serum microelements in AD patients and control subjects, it was observed that patients with AD had notably lower zinc levels. Genomic analysis of AD skin revealed distinct gene expression patterns, specifically the increased expression of CXCL10 in the epidermis. The heightened levels of CXCL10 in AD skin lesions were found to correlate with reduced serum zinc levels. Treatment with zinc gluconate showed reduced chemotactic response and CXCL10 release, suggesting its potential to regulate CXCL10 expression of keratinocytes in AD. The mechanism behind this involved the downregulation of STAT phosphorylation through activating PPARα. In the AD-like dermatitis mouse model, zinc gluconate therapy decreased serum IgE levels, alleviated skin lesion severity, reduced skin thickness, and lowered CXCL10 expression, demonstrating its efficacy in managing AD-like skin conditions. These findings indicate that zinc gluconate can reduce inflammation in keratinocytes by activating PPARα, inhibiting STAT signaling, and decreasing CXCL10 release, thus highlighting its potential as a therapeutic target for AD.


Subject(s)
Chemokine CXCL10 , Dermatitis, Atopic , Gluconates , Keratinocytes , PPAR alpha , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/pathology , Keratinocytes/drug effects , Keratinocytes/metabolism , Chemokine CXCL10/metabolism , Animals , Humans , Gluconates/pharmacology , PPAR alpha/metabolism , Male , Female , Mice , Adult , Skin/drug effects , Skin/pathology , Skin/metabolism , Disease Models, Animal , Mice, Inbred BALB C , Signal Transduction/drug effects
12.
Cell Signal ; 121: 111285, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38969192

ABSTRACT

TST has been mainly studied for its anti-tumor proliferation and antimicrobial effects, but not widely used in dermatological diseases. The mechanism of cellular damage by TST in response to H2O2-mediated oxidative stress was investigated in human skin immortalized keratinocytes (HaCaT) as an in vitro model. The findings reveal that TST treatment leads to increased oxidative stress in the cells by reducing levels of superoxide dismutase (SOD), glutathione (GSH), and catalase (CAT). This effect is further supported by an upsurge in the expression of malondialdehyde (MDA, a pivotal marker of lipid peroxidation). Additionally, dysregulation of FoxM1 at both gene and protein levels corroborates its involvement TST associated effects. Analysis of ferroptosis-related genes confirms dysregulation following TST treatment in HaCaT cells. Furthermore, TST treatment exhibits effects on mitochondrial morphology and function, affirming its induction of apoptosis in the cells through heightened oxidative stress due to mitochondrial damage and dysregulation of mitochondrial membrane potential.


Subject(s)
Ferroptosis , HaCaT Cells , Mitochondria , Oxidative Stress , Humans , Oxidative Stress/drug effects , Ferroptosis/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Membrane Potential, Mitochondrial/drug effects , Keratinocytes/metabolism , Keratinocytes/drug effects , Hydrogen Peroxide , Lipid Peroxidation/drug effects , Superoxide Dismutase/metabolism , Glutathione/metabolism
13.
PeerJ ; 12: e17608, 2024.
Article in English | MEDLINE | ID: mdl-38978756

ABSTRACT

According to our preliminary study, melatonin and its N-amide derivatives (N-(2-(1-4-bromobenzoyl-5-methoxy-1H-indol-3-yl)ethyl)acetamide (BBM) and 4-bromo-N-(2-(5-methoxy-1H-indol-3-yl)ethyl)benzamide (EBM)) inhibited the marker of acute inflammation in tests in vitro and in vivo. The anti-inflammatory agent is intended for the prevention and treatment of chemotherapy-induced toxicity. In this study aimed to evaluate the effect of melatonin and its derivatives on mechanisms related to chemotherapy-induced oral mucositis by in vitro ROS and 5-FU-induced human keratinocyte cells as well as in vivo oral mucositis model. In in vitro H2O2-induced HaCaT cells, BBM had the highest level of protection (34.57%) at a concentration 50 µM, followed by EBM (26.41%), and melatonin (7.9%). BBM also protected cells against 5-FU-induced to 37.69-27.25% at 12.5-100 µM while EBM was 36.93-29.33% and melatonin was 22.5-11.39%. In in vivo 5-FU-induced oral mucositis in mice, melatonin, BBM, and EBM gel formulations protected tissue damage from 5-FU similar to the standard compound, benzydamine. Moreover, the weight of mice and food consumption recovered more quickly in the BBM group. These findings suggested that it was possible to develop BBM and EBM as new therapeutic agents for the treatment of oral mucositis.


Subject(s)
Melatonin , Stomatitis , Melatonin/pharmacology , Melatonin/therapeutic use , Stomatitis/chemically induced , Stomatitis/drug therapy , Stomatitis/prevention & control , Stomatitis/pathology , Animals , Humans , Mice , Keratinocytes/drug effects , Fluorouracil/adverse effects , Fluorouracil/toxicity , Male , Reactive Oxygen Species/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antioxidants/pharmacology
14.
Drug Des Devel Ther ; 18: 2793-2812, 2024.
Article in English | MEDLINE | ID: mdl-38979400

ABSTRACT

Purpose: Zoledronate (ZA) stands as a highly effective antiresorptive agent known to trigger medication-related osteonecrosis of the jaw (MRONJ). Its clinical dosages primarily encompass those used for oncologic and osteoporosis treatments. While inflammation is recognized as a potential disruptor of mucosal healing processes associated with ZA, prior research has overlooked the influence of varying ZA dosages on tissue adaptability. Therefore, a deeper understanding of the specific mechanisms by which inflammation exacerbates ZA-induced MRONJ, particularly when inflammation acts as a risk factor, remains crucial. Methods: Cell proliferation and migration of human oral keratinocytes (HOK) was analyzed after treatment with different doses of ZA and/or lipopolysaccharide (LPS) to assess their possible effect on mucosal healing of extraction wounds. Mouse periodontitis models were established using LPS, and histological changes in extraction wounds were observed after the administration of oncologic dose ZA. Hematoxylin and eosin (HE) staining and immunofluorescence were used to evaluate mucosal healing. Results: In vitro, LPS did not exacerbate the effects of osteoporosis therapeutic dose of ZA on the proliferation and migration of HOK cells, while aggravated these with the oncologic dose of ZA treatment by inducing mitochondrial dysfunction and oxidative stress via regulating SIRT1 expression. Furthermore, SIRT1 overexpression can alleviate this process. In vivo, local injection of LPS increased the nonunion of mucous membranes in MRONJ and decreased the expression of SIRT1, PGC-1α, and MnSOD. Conclusion: Inflammation aggravates oncologic dose of ZA-induced mitochondrial dysfunction and oxidative stress via a SIRT1-dependent pathway, enhancing the risk of impaired mucosal healing in MRONJ. Our study implies that inflammation becomes a critical risk factor for MRONJ development at higher ZA concentrations. Elucidating the mechanisms of inflammation as a risk factor for mucosal non-healing in MRONJ could inform the development of SIRT1-targeted therapies.


Subject(s)
Cell Proliferation , Dose-Response Relationship, Drug , Inflammation , Signal Transduction , Sirtuin 1 , Zoledronic Acid , Sirtuin 1/metabolism , Animals , Mice , Humans , Cell Proliferation/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/chemically induced , Inflammation/pathology , Signal Transduction/drug effects , Zoledronic Acid/pharmacology , Zoledronic Acid/administration & dosage , Risk Factors , Cell Movement/drug effects , Bisphosphonate-Associated Osteonecrosis of the Jaw/pathology , Bisphosphonate-Associated Osteonecrosis of the Jaw/metabolism , Bisphosphonate-Associated Osteonecrosis of the Jaw/drug therapy , Mice, Inbred C57BL , Cells, Cultured , Male , Keratinocytes/drug effects , Keratinocytes/metabolism , Lipopolysaccharides/pharmacology
16.
Immun Inflamm Dis ; 12(7): e1303, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38967379

ABSTRACT

BACKGROUND: Psoriasis refers to a highly prevalent and immunologically mediated dermatosis with considerable deterioration in life quality. Wogonin, a sort of flavonoid, has been mentioned to elicit protective activities in skin diseases. However, whether Wogonin is implicated in the treatment of psoriasis and its specific mechanisms are not fully understood. AIM: The present work attempted to elaborate the role of Wogonin during the process of psoriasis and to concentrate on the associated action mechanism. METHODS: Cell counting kit-8 (CCK-8) method was initially applied to assay the viability of human keratinocyte HaCaT cells treated by varying concentrations of Wogonin. To mimic psoriasis in vitro, HaCaT cells were exposed to M5 cytokines. CCK-8 and 5-Ethynyl-2'-deoxyuridine  assays were adopted for the measurement of cell proliferation. Inflammatory levels were examined with enzyme-linked immunosorbent assay. Immunofluorescence staining tested nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) and Caspase-1 expressions. Western blot examined the protein expressions of proliferation-, inflammation-, pyroptosis-associated factors, and NLRP3. RESULTS: Wogonin treatment antagonized the proliferation, inflammatory response, and NLRP3/caspase-1/Gasdermin-D (GSDMD)-mediated pyroptosis in M5-challenged HaCaT cells. Besides, NLRP3 elevation partially abrogated the effects of Wogonin on M5-induced proliferation, inflammatory response, and NLRP3/caspase-1/GSDMD-mediated pyroptosis in HaCaT cells. CONCLUSION: In a word, Wogonin might exert anti-proliferation, anti-inflammatory and anti-pyroptosis activities in M5-induced cell model of psoriasis and the blockade of NLRP3/Caspase-1/GSDMD pathway might be recognized as a potential mechanism underlying the protective mechanism of Wogonin in psoriasis, suggesting Wogonin as a prospective anti-psoriasis drug.


Subject(s)
Caspase 1 , Cell Proliferation , Flavanones , Keratinocytes , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Signal Transduction , Humans , Flavanones/pharmacology , Pyroptosis/drug effects , Keratinocytes/drug effects , Keratinocytes/metabolism , Cell Proliferation/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Caspase 1/metabolism , Signal Transduction/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Psoriasis/drug therapy , Psoriasis/metabolism , Psoriasis/pathology , Inflammation/metabolism , Inflammation/drug therapy , HaCaT Cells , Cell Line , Gasdermins , Phosphate-Binding Proteins
17.
Science ; 385(6705): 194-200, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38991070

ABSTRACT

Millions of hibernating bats across North America have died from white-nose syndrome (WNS), an emerging disease caused by a psychrophilic (cold-loving) fungus, Pseudogymnoascus destructans, that invades their skin. Mechanisms of P. destructans invasion of bat epidermis remain obscure. Guided by our in vivo observations, we modeled hibernation with a newly generated little brown bat (Myotis lucifugus) keratinocyte cell line. We uncovered the stealth intracellular lifestyle of P. destructans, which inhibits apoptosis of keratinocytes and spreads through the cells by two epidermal growth factor receptor (EGFR)-dependent mechanisms: active penetration during torpor and induced endocytosis during arousal. Melanin of endocytosed P. destructans blocks endolysosomal maturation, facilitating P. destructans survival and germination after return to torpor. Blockade of EGFR aborts P. destructans entry into keratinocytes.


Subject(s)
Arousal , Ascomycota , Chiroptera , ErbB Receptors , Hibernation , Keratinocytes , Animals , Apoptosis , Ascomycota/physiology , Ascomycota/pathogenicity , Cell Line , Chiroptera/microbiology , Chiroptera/physiology , Endocytosis , ErbB Receptors/metabolism , Keratinocytes/microbiology , Melanins/metabolism
18.
Autoimmunity ; 57(1): 2377138, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39037929

ABSTRACT

Keratinocytes in mucosal and skin tissues maintain tissue integrity via desmosomes and desmoglein-3 (Dsg3). Pemphigus Vulgaris (PV) is a life-threatening autoimmune blistering disease characterized by autoantibodies against Dsg3, disrupting desmosomes. Nuclear factor erythroid 2-related factor 2 (Nrf2) regulates oxidative stress responses crucial for skin tissue protection. Although the pathogenesis of PV is known, the detailed molecular events remain unclear. This study investigates changes in Nrf2 expression in keratinocytes following pathogenic anti-Dsg3 antibody AK23 exposure, using dose- and time-dependent studies employing immunofluorescence analysis. N/TERT keratinocytes were cultured in keratinocytes serum-free medium and treated with AK23 at varying doses (5 µg/mL,40µg/mL,75µg/mL) and durations (2, 6, 24 h). Immunofluorescence staining was performed to assess the expression of Nrf2 and Dsg3. All fluorescent images were analyzed using ImageJ software. A dose-dependent increase in Dsg3 was noted following AK23 treatment, while Nrf2 expression and subcellular localization varied. Time-course analyses showed decreased Nrf2 at 24 h and increased Dsg3 levels. Early time-point (2 and 6 h) variations were evident in Nrf2 levels. This study highlights the impact of AK23 on Nrf2 expression, potentially disrupting Nrf2-mediated cytoprotection and implicating oxidative stress (ROS generation) in PV pathogenesis. Further investigation is necessary to validate the findings.


Subject(s)
Desmoglein 3 , Keratinocytes , NF-E2-Related Factor 2 , Pemphigus , Reactive Oxygen Species , Pemphigus/immunology , Pemphigus/metabolism , Pemphigus/pathology , NF-E2-Related Factor 2/metabolism , Keratinocytes/metabolism , Keratinocytes/immunology , Desmoglein 3/immunology , Desmoglein 3/metabolism , Humans , Reactive Oxygen Species/metabolism , Oxidative Stress , Antibodies, Monoclonal/pharmacology , Cell Line , Autoantibodies/immunology
19.
Exp Dermatol ; 33(7): e15142, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39032085

ABSTRACT

Frequent itching and incessant scratching are commonly observed in various chronic inflammatory skin conditions, including atopic dermatitis and psoriasis. The persistent and prolonged nature of pruritus can worsen one's quality of life. Keratinocytes (KCs), the predominant cells of the epidermis, have been confirmed to interact with sensory neurons and immune cells and be involved in chronic skin inflammatory diseases associated with pruritus. Initially, KCs and sensory neurons form a unique synapse-like connection within the epidermis, serving as the structural foundation for their interaction. Additionally, several receptors, including toll-like receptors and protease-activated receptor 2, expressed on KCs, become activated in an inflammatory milieu. On the one hand, activated KCs are sources of pro-inflammatory cytokines and neurotrophic factors, such as adenosine triphosphate, thymic stromal lymphopoietin, and nerve growth factor, which directly or indirectly participate in stimulating sensory neurons, thereby contributing to the itch sensations. On the other hand, KCs also function as primary transducers alongside intraepidermal nerve endings, directly initiating pruritic responses. This review summarizes the current literature and highlights the critical role of KCs in the development and persistence of chronic itch in inflammatory skin disorders.


Subject(s)
Keratinocytes , Pruritus , Humans , Pruritus/etiology , Pruritus/physiopathology , Keratinocytes/metabolism , Chronic Disease , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Dermatitis, Atopic/complications , Animals , Cytokines/metabolism , Psoriasis/complications
20.
Nat Commun ; 15(1): 5834, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992003

ABSTRACT

We present Dystrophic Epidermolysis Bullosa Cell Therapy (DEBCT), a scalable platform producing autologous organotypic iPS cell-derived induced skin composite (iSC) grafts for definitive treatment. Clinical-grade manufacturing integrates CRISPR-mediated genetic correction with reprogramming into one step, accelerating derivation of COL7A1-edited iPS cells from patients. Differentiation into epidermal, dermal and melanocyte progenitors is followed by CD49f-enrichment, minimizing maturation heterogeneity. Mouse xenografting of iSCs from four patients with different mutations demonstrates disease modifying activity at 1 month. Next-generation sequencing, biodistribution and tumorigenicity assays establish a favorable safety profile at 1-9 months. Single cell transcriptomics reveals that iSCs are composed of the major skin cell lineages and include prominent holoclone stem cell-like signatures of keratinocytes, and the recently described Gibbin-dependent signature of fibroblasts. The latter correlates with enhanced graftability of iSCs. In conclusion, DEBCT overcomes manufacturing and safety roadblocks and establishes a reproducible, safe, and cGMP-compatible therapeutic approach to heal lesions of DEB patients.


Subject(s)
Cell- and Tissue-Based Therapy , Collagen Type VII , Epidermolysis Bullosa Dystrophica , Induced Pluripotent Stem Cells , Humans , Epidermolysis Bullosa Dystrophica/therapy , Epidermolysis Bullosa Dystrophica/genetics , Animals , Induced Pluripotent Stem Cells/transplantation , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Mice , Collagen Type VII/genetics , Collagen Type VII/metabolism , Cell- and Tissue-Based Therapy/methods , Fibroblasts/metabolism , Cell Differentiation , Keratinocytes/metabolism , Keratinocytes/transplantation , Skin/metabolism , Transplantation, Autologous , Male , Mutation , Female , Skin Transplantation/methods , Gene Editing/methods , CRISPR-Cas Systems
SELECTION OF CITATIONS
SEARCH DETAIL