Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.351
Filter
1.
Cell Death Dis ; 15(6): 392, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834617

ABSTRACT

Keratinocyte proliferation and differentiation in epidermis are well-controlled and essential for reacting to stimuli such as ultraviolet light. Imbalance between proliferation and differentiation is a characteristic feature of major human skin diseases such as psoriasis and squamous cell carcinoma. However, the effect of keratinocyte metabolism on proliferation and differentiation remains largely elusive. We show here that the gluconeogenic enzyme fructose-1,6-bisphosphatase 1 (FBP1) promotes differentiation while inhibits proliferation of keratinocyte and suppresses psoriasis development. FBP1 is identified among the most upregulated genes induced by UVB using transcriptome sequencing and is elevated especially in upper epidermis. Fbp1 heterozygous mice exhibit aberrant epidermis phenotypes with local hyperplasia and dedifferentiation. Loss of FBP1 promotes proliferation and inhibits differentiation of keratinocytes in vitro. Mechanistically, FBP1 loss facilitates glycolysis-mediated acetyl-CoA production, which increases histone H3 acetylation at lysine 9, resulting in enhanced transcription of proliferation genes. We further find that the expression of FBP1 is dramatically reduced in human psoriatic lesions and in skin of mouse imiquimod psoriasis model. Fbp1 deficiency in mice facilitates psoriasis-like skin lesions development through glycolysis and acetyl-CoA production. Collectively, our findings reveal a previously unrecognized role of FBP1 in epidermal homeostasis and provide evidence for FBP1 as a metabolic psoriasis suppressor.


Subject(s)
Cell Differentiation , Cell Proliferation , Fructose-Bisphosphatase , Histones , Keratinocytes , Psoriasis , Psoriasis/pathology , Psoriasis/metabolism , Psoriasis/genetics , Animals , Keratinocytes/metabolism , Keratinocytes/pathology , Humans , Acetylation , Histones/metabolism , Fructose-Bisphosphatase/metabolism , Fructose-Bisphosphatase/genetics , Mice , Glycolysis , Mice, Inbred C57BL , Acetyl Coenzyme A/metabolism , Disease Models, Animal
4.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167207, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38701954

ABSTRACT

PURPOSE: In this study, we identified and diagnosed a novel inherited condition called Dyschromatosis, Ichthyosis, Deafness, and Atopic Disease (DIDA) syndrome. We present a series of studies to clarify the pathogenic variants and specific mechanism. METHODS: Exome sequencing and Sanger sequencing was conducted in affected and unaffected family members. A variety of human and cell studies were performed to explore the pathogenic process of keratosis. RESULTS: Our finding indicated that DIDA syndrome was caused by compound heterozygous variants in the oxysterol-binding protein-related protein 2 (OSBPL2) gene. Furthermore, our findings revealed a direct interaction between OSBPL2 and Phosphoinositide phospholipase C-beta-3 (PLCB3), a key player in hyperkeratosis. OSBPL2 effectively inhibits the ubiquitylation of PLCB3, thereby stabilizing PLCB3. Conversely, OSBPL2 variants lead to enhanced ubiquitination and subsequent degradation of PLCB3, leading to epidermal hyperkeratosis, characterized by aberrant proliferation and delayed terminal differentiation of keratinocytes. CONCLUSIONS: Our study not only unveiled the association between OSBPL2 variants and the newly identified DIDA syndrome but also shed light on the underlying mechanism.


Subject(s)
Deafness , Ichthyosis , Pedigree , Phospholipase C beta , Humans , Deafness/genetics , Deafness/pathology , Phospholipase C beta/genetics , Phospholipase C beta/metabolism , Female , Male , Ichthyosis/genetics , Ichthyosis/pathology , Ichthyosis/metabolism , Heterozygote , Ubiquitination , Keratinocytes/metabolism , Keratinocytes/pathology , Exome Sequencing , Adult , Syndrome , HEK293 Cells , Receptors, Steroid
5.
Int Immunopharmacol ; 135: 112244, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38776847

ABSTRACT

Psoriasis is a common and prevalent chronic papulosquamous cutaneous disorder characterized by sustained inflammation, uncontrolled keratinocyte proliferation, dysfunctional differentiation, and angiogenesis. Autophagy, an intracellular catabolic process, can be induced in response to nutrient stress. It entails the degradation of cellular constituents through the lysosomal machinery, and its association with psoriasis has been well-documented. Nevertheless, there remains a notable dearth of research concerning the involvement of autophagy in the pathogenesis of psoriasis within human skin. This review provides a comprehensive overview of autophagy in psoriasis pathogenesis, focusing on its involvement in two key pathological manifestations: sustained inflammation and uncontrolled keratinocyte proliferation and differentiation. Additionally, it discusses potential avenues for disease management.


Subject(s)
Autophagy , Cell Differentiation , Inflammation , Keratinocytes , Psoriasis , Humans , Psoriasis/immunology , Psoriasis/pathology , Keratinocytes/immunology , Keratinocytes/pathology , Keratinocytes/physiology , Inflammation/immunology , Animals , Cell Proliferation , Skin/pathology , Skin/immunology
6.
J Med Virol ; 96(4): e29580, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38566572

ABSTRACT

A persistent infection with human papillomavirus (HPV) can induce precancerous lesions of the cervix that may ultimately develop into cancer. Cervical cancer development has been linked to altered microRNA (miRNA) expression, with miRNAs regulating anchorage-independent growth being particularly important for the progression of precancerous lesions to cancer. In this study, we set out to identify and validate targets of miR-129-5p, a previously identified tumor suppressive miRNA involved in anchorage-independent growth and HPV-induced carcinogenesis. We predicted 26 potential miR-129-5p targets using online databases, followed by KEGG pathway enrichment analysis. RT-qPCR and luciferase assays confirmed that 3'UTR regions of six genes (ACTN1, BMPR2, CAMK4, ELK4, EP300, and GNAQ) were targeted by miR-129-5p. Expressions of ACTN1, CAMK4, and ELK4 were inversely correlated to miR-129-5p expression in HPV-transformed keratinocytes, and their silencing reduced anchorage-independent growth. Concordantly, miR-129-5p overexpression decreased protein levels of ACTN1, BMPR2, CAMK4 and ELK4 in anchorage-independent conditions. Additionally, c-FOS, a downstream target of ELK4, was downregulated upon miR-129-5p overexpression, suggesting regulation through the ELK4/c-FOS axis. ACTN1 and ELK4 expression was also upregulated in high-grade precancerous lesions and cervical cancers, supporting their clinical relevance. In conclusion, we identified six targets of miR-129-5p involved in the regulation of anchorage-independent growth, with ACTN1, BMPR2, ELK4, EP300, and GNAQ representing novel targets for miR-129-5p. For both ACTN1 and ELK4 functional and clinical relevance was confirmed, indicating that miR-129-5p-regulated ACTN1 and ELK4 expression contributes to HPV-induced carcinogenesis.


Subject(s)
MicroRNAs , Papillomavirus Infections , Precancerous Conditions , Uterine Cervical Neoplasms , Female , Humans , Human Papillomavirus Viruses , Papillomavirus Infections/genetics , Papillomavirus Infections/pathology , Cell Line, Tumor , MicroRNAs/genetics , MicroRNAs/metabolism , Keratinocytes/metabolism , Keratinocytes/pathology , Carcinogenesis/genetics , Carcinogenesis/pathology , Precancerous Conditions/pathology , Cell Proliferation/genetics , ets-Domain Protein Elk-4 , Actinin/genetics
7.
Cells ; 13(8)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38667326

ABSTRACT

Precancerous cells in the oral cavity may appear as oral potentially malignant disorders, but they may also present as dysplasia without visual manifestation in tumor-adjacent tissue. As it is currently not possible to prevent the malignant transformation of these oral precancers, new treatments are urgently awaited. Here, we generated precancer culture models using a previously established method for the generation of oral keratinocyte cultures and incorporated CRISPR/Cas9 editing. The generated cell lines were used to investigate the efficacy of a set of small molecule inhibitors. Tumor-adjacent mucosa and oral leukoplakia biopsies were cultured and genetically characterized. Mutations were introduced in CDKN2A and TP53 using CRISPR/Cas9 and combined with the ectopic activation of telomerase to generate cell lines with prolonged proliferation. The method was tested in normal oral keratinocytes and tumor-adjacent biopsies and subsequently applied to a large set of oral leukoplakia biopsies. Finally, a subset of the immortalized cell lines was used to assess the efficacy of a set of small molecule inhibitors. Culturing and genomic engineering was highly efficient for normal and tumor-adjacent oral keratinocytes, but success rates in oral leukoplakia were remarkably low. Knock-out of CDKN2A in combination with either the activation of telomerase or knock-out of TP53 seemed a prerequisite for immortalization. Prolonged culturing was accompanied by additional genetic aberrations in these cultures. The generated cell lines were more sensitive than normal keratinocytes to small molecule inhibitors of previously identified targets. In conclusion, while very effective for normal keratinocytes and tumor-adjacent biopsies, the success rate of oral leukoplakia cell culturing methods was very low. Genomic engineering enabled the prolonged culturing of OL-derived keratinocytes but was associated with acquired genetic changes. Further studies are required to assess to what extent the immortalized cultures faithfully represent characteristics of the cells in vivo.


Subject(s)
Keratinocytes , Leukoplakia, Oral , Mouth Neoplasms , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Leukoplakia, Oral/genetics , Leukoplakia, Oral/pathology , Telomerase/genetics , Telomerase/metabolism , Genetic Engineering , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , CRISPR-Cas Systems/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Mouth Mucosa/pathology , Precancerous Conditions/pathology , Precancerous Conditions/genetics
8.
Am J Hum Genet ; 111(5): 896-912, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38653249

ABSTRACT

Porokeratosis is a clonal keratinization disorder characterized by solitary, linearly arranged, or generally distributed multiple skin lesions. Previous studies showed that genetic alterations in MVK, PMVK, MVD, or FDPS-genes in the mevalonate pathway-cause hereditary porokeratosis, with skin lesions harboring germline and lesion-specific somatic variants on opposite alleles. Here, we identified non-hereditary porokeratosis associated with epigenetic silencing of FDFT1, another gene in the mevalonate pathway. Skin lesions of the generalized form had germline and lesion-specific somatic variants on opposite alleles in FDFT1, representing FDFT1-associated hereditary porokeratosis identified in this study. Conversely, lesions of the solitary or linearly arranged localized form had somatic bi-allelic promoter hypermethylation or mono-allelic promoter hypermethylation with somatic genetic alterations on opposite alleles in FDFT1, indicating non-hereditary porokeratosis. FDFT1 localization was uniformly diminished within the lesions, and lesion-derived keratinocytes showed cholesterol dependence for cell growth and altered expression of genes related to cell-cycle and epidermal development, confirming that lesions form by clonal expansion of FDFT1-deficient keratinocytes. In some individuals with the localized form, gene-specific promoter hypermethylation of FDFT1 was detected in morphologically normal epidermis adjacent to methylation-related lesions but not distal to these lesions, suggesting that asymptomatic somatic epigenetic mosaicism of FDFT1 predisposes certain skin areas to the disease. Finally, consistent with its genetic etiology, topical statin treatment ameliorated lesions in FDFT1-deficient porokeratosis. In conclusion, we identified bi-allelic genetic and/or epigenetic alterations of FDFT1 as a cause of porokeratosis and shed light on the pathogenesis of skin mosaicism involving clonal expansion of epigenetically altered cells.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Keratinocytes , Mosaicism , Porokeratosis , Promoter Regions, Genetic , Porokeratosis/genetics , Porokeratosis/pathology , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Promoter Regions, Genetic/genetics , Male , Alleles , Female
9.
Med J Aust ; 220(9): 472-479, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38685827

ABSTRACT

OBJECTIVES: To examine recent changes in the numbers of Medicare-subsidised keratinocyte cancer excisions, particularly for younger people exposed to primary prevention campaigns since the early 1980s. STUDY DESIGN: Retrospective cohort study; analysis of administrative data. SETTING, PARTICIPANTS: Analysis of Medicare Benefits Schedule (MBS) claims data for procedures related to the diagnosis and treatment of keratinocyte cancer in Australia, 2012-2021. MAIN OUTCOME MEASURES: Age-standardised rates for MBS-subsidised claims for first surgical squamous cell carcinoma (SCC) and basal cell carcinoma (BCC) excisions, Mohs surgery, surgical excisions of benign lesions, skin biopsies, and cryotherapy or serial curettage of premalignant and malignant lesions, overall, and by sex, state/territory, and age group; average annual percentage change in rate for time intervals determined by joinpoint regression. RESULTS: In men, the age-standardised rate of BCC/SCC excisions increased by 1.9% (95% confidence interval [CI], 1.4-2.4%) per year during 2012-2019 (from 2931 to 3371 per 100 000 men) and then declined by 3.8% (95% CI, 0.5-7.0%) per year during 2019-2021 (to 3152 per 100 000). In women, the age-standardised rate increased by 2.2% (95% CI, 1.7-2.8%) per year during 2012-2019 (from 1798 to 2093 per 100 000 women); the decline to 1967 excisions per 100 000 women in 2021 was not statistically significant. BCC/SCC excision rates declined for men under 55 years of age (by 1.0-3.4% per year) and women under 45 years of age (by 1.7-2.3% per year). Age-standardised biopsy rates increased during 2012-2021 in all age groups (by 2.8-6.9% per year). CONCLUSIONS: Rates of MBS-subsidised treatment for keratinocyte cancers increased during 2012-2019, but BCC/SCC treatment rates declined among younger Australians, who have probably been exposed to less sunlight than earlier generations because of public health interventions and population-wide lifestyle changes related to technology use.


Subject(s)
Carcinoma, Basal Cell , Carcinoma, Squamous Cell , Skin Neoplasms , Humans , Retrospective Studies , Male , Skin Neoplasms/pathology , Skin Neoplasms/epidemiology , Skin Neoplasms/surgery , Skin Neoplasms/therapy , Female , Australia/epidemiology , Carcinoma, Basal Cell/epidemiology , Carcinoma, Basal Cell/therapy , Carcinoma, Basal Cell/pathology , Carcinoma, Basal Cell/surgery , Middle Aged , Carcinoma, Squamous Cell/epidemiology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Aged , Adult , Keratinocytes/pathology , Aged, 80 and over , Mohs Surgery/statistics & numerical data , Young Adult , Cryotherapy/statistics & numerical data , Age Factors
10.
J Invest Dermatol ; 144(5): 989-1000.e1, 2024 May.
Article in English | MEDLINE | ID: mdl-38643989

ABSTRACT

A compromised permeability barrier is a hallmark of atopic dermatitis (AD). Localized to the outermost skin layer, the stratum corneum (SC) is critically dependent on terminal differentiation of epidermal keratinocytes, which transform into protein-rich corneocytes surrounded by extracellular lamellae of unique epidermal lipids, conferring permeability barrier function. These structures are disrupted in AD. A leaky barrier is prone to environmental insult, which in AD elicits type 2-dominant inflammation, in turn resulting in a vicious cycle further impairing the SC structure. Therapies directed at enforcing SC structure and anti-inflammatory strategies administered by topical and systemic route as well as UV therapy have differential effects on the permeability barrier. The expanding armamentarium of therapeutic modalities for AD treatment warrants optimization of their effects on permeability barrier function.


Subject(s)
Dermatitis, Atopic , Keratinocytes , Dermatitis, Atopic/therapy , Dermatitis, Atopic/pathology , Humans , Keratinocytes/pathology , Permeability , Epidermis/pathology , Epidermis/metabolism , Skin/pathology , Skin/metabolism , Animals , Cell Differentiation
11.
Int J Mol Sci ; 25(8)2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38674130

ABSTRACT

IQGAP3 (IQ Motif Containing GTPase Activating Protein 3) is member of the IQGAP family of scaffold proteins, which are essential for assembling multiprotein complexes that coordinate various intracellular signaling pathways. Previous research has shown that IQGAP3 is overexpressed in psoriatic skin lesions. Given its involvement in processes like cell proliferation and chemokine signaling, we sought to explore its molecular role in driving the psoriatic phenotype of keratinocytes. By conducting transcriptome profiling of HaCaT keratinocytes, we identified numerous psoriasis-associated pathways that were affected when IQGAP3 was knocked down. These included alterations in NFkB signaling, EGFR signaling, activation of p38/MAPK and ERK1/ERK2, lipid metabolism, cytokine production, and the response to inflammatory cytokine stimulation. Real-time analysis further revealed changes in cell growth dynamics, including proliferation and wound healing. The balance between cell proliferation and apoptosis was altered, as were skin barrier functions and the production of IL-6 and IFNγ. Despite these significant findings, the diversity of the alterations observed in the knockdown cells led us to conclude that IQGAP3 may not be the best target for the therapeutic inhibition to normalize the phenotype of keratinocytes in psoriasis.


Subject(s)
Cell Proliferation , GTPase-Activating Proteins , Keratinocytes , Psoriasis , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Psoriasis/metabolism , Psoriasis/pathology , Psoriasis/genetics , ras GTPase-Activating Proteins/metabolism , ras GTPase-Activating Proteins/genetics , Signal Transduction , HaCaT Cells , Cytokines/metabolism , Apoptosis , Skin/metabolism , Skin/pathology , Cell Line , Gene Expression Profiling
12.
Cutis ; 113(2): 82-91, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38593108

ABSTRACT

Psoriasis is a chronic heterogeneous condition with multiple available treatment options that have resulted in dramatic disease improvements for patients. IL-23/IL-17 signaling is the central immune signaling pathway driving psoriasis, though recent research has uncovered other key contributing signals such as IL-17C, IL-17F, IL-36, and tyrosine kinase 2 (TYK2). Novel therapeutic targets inhibiting these cytokines have expanded our understanding of the pathogenesis of psoriasis. IL-23/IL-17 signaling is critical for the development of epidermal hyperplasia and the mature psoriatic plaque in susceptible individuals. Increased IL-17 and IL-23 expression works synergistically with other cytokines, such as IL-12, IL-22, IL-36, tumor necrosis factor (TNF), and interferon (IFN), to help create a self-sustaining, feed-forward circuit in keratinocytes, which contributes to the chronicity of the disease. This clinical review highlights recent discoveries in the immunopathogenesis of psoriasis and summarizes new antipsoriasis therapies targeting IL-36, IL-17F, aryl hydrocarbon receptors (AHRs), phosphodiesterase 4 (PDE4), and TYK2 signaling. Despite recent success in the treatment of psoriasis, continued research is needed to further advance disease understanding and shape management strategies.


Subject(s)
Interleukin-17 , Psoriasis , Humans , Psoriasis/drug therapy , Keratinocytes/metabolism , Keratinocytes/pathology , Cytokines , Interleukin-23
13.
J Dermatol ; 51(5): 621-631, 2024 May.
Article in English | MEDLINE | ID: mdl-38605467

ABSTRACT

Alopecia areata refers to an autoimmune illness indicated by persistent inflammation. The key requirement for alopecia areata occurrence is the disruption of immune-privileged regions within the hair follicles. Recent research has indicated that neuropeptides play a role in the damage to hair follicles by triggering neurogenic inflammation, stimulating mast cells ambient the follicles, and promoting apoptotic processes in keratinocytes. However, the exact pathogenesis of alopecia areata requires further investigation. Recently, there has been an increasing focus on understanding the mechanisms of immune diseases resulting from the interplay between the nervous and the immune system. Neurogenic inflammation due to neuroimmune disorders of the skin system may disrupt the inflammatory microenvironment of the hair follicle, which plays a crucial part in the progression of alopecia areata.


Subject(s)
Alopecia Areata , Hair Follicle , Neurogenic Inflammation , Alopecia Areata/immunology , Alopecia Areata/etiology , Alopecia Areata/pathology , Humans , Hair Follicle/immunology , Hair Follicle/pathology , Neurogenic Inflammation/immunology , Neurogenic Inflammation/etiology , Neuropeptides/metabolism , Neuropeptides/immunology , Mast Cells/immunology , Keratinocytes/immunology , Keratinocytes/pathology , Apoptosis/immunology , Animals
14.
Cell Cycle ; 23(5): 537-554, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38662954

ABSTRACT

Cholesteatoma is a common disease of the middle ear. Currently, surgical removal is the only treatment option and patients face a high risk of relapse. The molecular basis of cholesteatoma remains largely unknown. Here, we show that Osteopontin (OPN), a predominantly secreted protein, plays a crucial role in the development of middle ear cholesteatoma. Global transcriptome analysis revealed the loss of epithelial features and an enhanced immune response in human cholesteatoma tissues. Quantitative RT-PCR and immunohistochemical staining of middle ear cholesteatoma validated the reduced expression of epithelial markers, as well as the elevated expression of mesenchymal markers including Vimentin and Fibronectin, but not N-Cadherin, α-smooth muscle actin (α-SMA) or ferroptosis suppressor protein 1 (FSP1), indicating a partial epithelial-mesenchymal transition (EMT) state. Besides, the expression of OPN was significantly elevated in human cholesteatoma tissues. Treatment with OPN promoted cell proliferation, survival and migration and led to a partial EMT in immortalized human keratinocyte cells. Importantly, blockade of OPN signaling could remarkably improve the cholesteatoma-like symptoms in SD rats. Our mechanistic study demonstrated that the AKT-zinc finger E-box binding homeobox 2 (ZEB2) axis mediated the effects of OPN. Overall, these findings suggest that targeting the OPN signaling represents a promising strategy for the treatment of middle ear cholesteatoma.


Subject(s)
Cell Proliferation , Cholesteatoma, Middle Ear , Epithelial-Mesenchymal Transition , Osteopontin , Rats, Sprague-Dawley , Epithelial-Mesenchymal Transition/genetics , Humans , Osteopontin/metabolism , Osteopontin/genetics , Animals , Cholesteatoma, Middle Ear/metabolism , Cholesteatoma, Middle Ear/pathology , Cholesteatoma, Middle Ear/genetics , Rats , Cell Proliferation/genetics , Cell Movement/genetics , Signal Transduction , Male , Proto-Oncogene Proteins c-akt/metabolism , Zinc Finger E-box Binding Homeobox 2/metabolism , Zinc Finger E-box Binding Homeobox 2/genetics , Keratinocytes/metabolism , Keratinocytes/pathology , Female
15.
J Prim Health Care ; 16(1): 90-95, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38546776

ABSTRACT

Introduction The New Zealand population has one of the highest incidences of skin cancer in the world. Hospital waiting lists for surgical excision of keratinocytic skin cancers (basal cell carcinoma and squamous cell carcinoma) are lengthy, and increasingly, excisions are undertaken in primary care. Teledermatology, in response to general practitioners' electronic referrals (e-referrals), can improve clinical communication between general practitioners and dermatologists. Aim The aim of this study was to evaluate an excision pathway for keratinocytic cancers diagnosed by teledermatology. Methods A retrospective observational descriptive review of a 3-month cohort of primary care e-referrals was undertaken. Results Three hundred and fifty eight suspected keratinocytic cancers (KCs) were diagnosed by teledermatology; histology reports confirmed KC in 201 of 267 excisions (75%). The majority (77.2%) were excised by general practitioners an average of 25 days after the dermatologist's recommendation. The rest were excised by plastic surgeons in private (3.4%) or at a public hospital (19.5%) after an average of 40 or 134 days, respectively. Discussion E-referral pathways are now widely implemented. However, the ideal workflow for skin cancer management is unknown. We have demonstrated in New Zealand that surgery can be undertaken in primary care within a month of a teledermatology diagnosis and excision recommendation. Conclusion This study reports prompt excision of KCs by general practitioners after an e-referral and a teledermatology response.


Subject(s)
Carcinoma, Basal Cell , Dermatology , Skin Neoplasms , Telemedicine , Humans , Retrospective Studies , Dermatology/methods , Skin Neoplasms/diagnosis , Skin Neoplasms/surgery , Skin Neoplasms/epidemiology , Carcinoma, Basal Cell/diagnosis , Carcinoma, Basal Cell/surgery , Carcinoma, Basal Cell/epidemiology , Keratinocytes/pathology , Telemedicine/methods
16.
Acta Derm Venereol ; 104: adv19460, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38483083

ABSTRACT

Since December 2019, the COVID-19 pandemic has profoundly affected healthcare. The real effects of the COVID-19 pandemic on skin cancer are still unclear, more than 3 years later. This study aims to summarise the pandemic's impact on skin cancer diagnosis and outcome. A systematic review and meta-analysis was conducted, selecting studies comparing skin cancer diagnosis and prognosis post-pandemic with pre-pandemic data. A total of 27 papers were reviewed including 102,263 melanomas and 271,483 keratinocyte carcinomas. During the initial pandemic months (January-July 2020), melanoma surgeries dropped by 29.7% and keratinocyte carcinomas surgeries by 50.8%. Early pandemic tumours exhibited greater thickness and stage. In a long-term period beyond the initial months, melanoma surgeries decreased by 9.3%, keratinocyte carcinomas by 16.6%. No significant differences were observed in the Breslow thickness of melanomas after the start of the pandemic (mean difference 0.06, 95% confidence interval -0.46, 0.58). Melanomas operated on post-pandemic onset had an increased risk of ulceration (odds ratio 1.35, 95% confidence interval 1.22-1.50). Keratinocyte carcinomas showed increased thickness and worsened stage post-pandemic. However, studies included were mostly retrospective and cross-sectional, reporting diverse data. This review indicates that the pandemic likely caused delays in skin cancer diagnosis and treatment, potentially impacting patient outcomes.


Subject(s)
COVID-19 , Carcinoma , Melanoma , Skin Neoplasms , Humans , Melanoma/diagnosis , Melanoma/epidemiology , Melanoma/surgery , Pandemics , Retrospective Studies , Cross-Sectional Studies , COVID-19/epidemiology , Skin Neoplasms/diagnosis , Skin Neoplasms/epidemiology , Skin Neoplasms/surgery , Keratinocytes/pathology , COVID-19 Testing
17.
J Biol Chem ; 300(4): 107173, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38499149

ABSTRACT

Sunlight exposure results in an inflammatory reaction of the skin commonly known as sunburn, which increases skin cancer risk. In particular, the ultraviolet B (UVB) component of sunlight induces inflammasome activation in keratinocytes to instigate the cutaneous inflammatory responses. Here, we explore the intracellular machinery that maintains skin homeostasis by suppressing UVB-induced inflammasome activation in human keratinocytes. We found that pharmacological inhibition of autophagy promoted UVB-induced NLRP3 inflammasome activation. Unexpectedly, however, gene silencing of Atg5 or Atg7, which are critical for conventional autophagy, had no effect, whereas gene silencing of Beclin1, which is essential not only for conventional autophagy but also for Atg5/Atg7-independent alternative autophagy, promoted UVB-induced inflammasome activation, indicating an involvement of alternative autophagy. We found that damaged mitochondria were highly accumulated in UVB-irradiated keratinocytes when alternative autophagy was inhibited, and they appear to be recognized by NLRP3. Overall, our findings indicate that alternative autophagy, rather than conventional autophagy, suppresses UVB-induced NLRP3 inflammasome activation through the clearance of damaged mitochondria in human keratinocytes and illustrate a previously unknown involvement of alternative autophagy in inflammation. Alternative autophagy may be a new therapeutic target for sunburn and associated cutaneous disorders.


Subject(s)
Autophagy , Inflammasomes , Keratinocytes , Mitochondria , NLR Family, Pyrin Domain-Containing 3 Protein , Ultraviolet Rays , Humans , Autophagy/radiation effects , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Beclin-1/metabolism , Beclin-1/genetics , Inflammasomes/metabolism , Keratinocytes/metabolism , Keratinocytes/pathology , Keratinocytes/radiation effects , Mitochondria/metabolism , Mitochondria/radiation effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Ultraviolet Rays/adverse effects , Cells, Cultured
18.
Cell Death Dis ; 15(3): 180, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38429278

ABSTRACT

Gasdermin E (GSDME) has recently been identified as a critical executioner to mediate pyroptosis. While epidermal keratinocytes can initiate GSDME-mediated pyroptosis, the role of keratinocyte GSDME in psoriatic dermatitis remains poorly characterized. Through analysis of GEO datasets, we found elevated GSDME levels in psoriatic lesional skin. Additionally, GSDME levels correlated with both psoriasis severity and response to biologics treatments. Single-cell RNA sequencing (scRNA-seq) from a GEO dataset revealed GSDME upregulation in keratinocytes of psoriasis patients. In the imiquimod (IMQ)-induced psoriasis-like dermatitis mouse model, both full-length and cleaved forms of caspase-3 and GSDME were elevated in the epidermis. Abnormal proliferation and differentiation of keratinocytes and dermatitis were attenuated in Gsdme-/- mice and keratinocyte-specific Gsdme conditional knockout mice after IMQ stimulation. Exposure of keratinocytes to mixed cytokines (M5), mimicking psoriatic conditions, led to GSDME cleavage. Moreover, the interaction between GSDME-FL and p65 or c-jun was significantly increased after M5 stimulation. GSDME knockdown inhibited nuclear translocation of p65 and c-jun and decreased upregulation of psoriatic inflammatory mediators such as IL1ß, CCL20, CXCL1, CXCL8, S100A8, and S100A9 in M5-challenged keratinocytes. In conclusion, GSDME in keratinocytes contributes to the pathogenesis and progression of psoriasis, potentially in a pyroptosis-independent manner by interacting and promoting translocation of p65 and c-jun. These findings suggest that keratinocyte GSDME could serve as a potential therapeutic target for psoriasis treatment.


Subject(s)
Dermatitis , Gasdermins , Psoriasis , Animals , Humans , Mice , Dermatitis/metabolism , Dermatitis/pathology , Gasdermins/metabolism , Imiquimod/adverse effects , Inflammation/pathology , Keratinocytes/pathology , Psoriasis/metabolism , Psoriasis/pathology , Transcription Factor RelA/metabolism , Proto-Oncogene Proteins c-jun/metabolism
19.
J Cutan Med Surg ; 28(2): NP19-NP35, 2024.
Article in English | MEDLINE | ID: mdl-38314729

ABSTRACT

Psoriasis is an immune-mediated chronic inflammatory skin disease that affects 2% to 3% of the world's population. It is widely assumed that immune cells and cytokines acting together play a crucial part in the pathophysiology of psoriasis by promoting the excessive proliferation of skin keratinocytes and inflammatory infiltration. Interleukins (ILs), as a critical component of cytokines, have been closely associated with the pathogenesis and progression of psoriasis. This review summarizes the current contribution of ILs to psoriasis and describes the role each IL performs in psoriasis. Furthermore, the paper presents the therapeutic effects and application prospects of biologics developed for ILs in clinical treatment and experiments. The study aims to further the research on ILs in the treatment of psoriasis.


Subject(s)
Interleukins , Psoriasis , Humans , Psoriasis/drug therapy , Psoriasis/pathology , Skin/pathology , Keratinocytes/pathology , Cytokines
20.
Am J Pathol ; 194(5): 708-720, 2024 May.
Article in English | MEDLINE | ID: mdl-38320628

ABSTRACT

Psoriasis is a chronic inflammatory skin disease characterized by the activation of keratinocytes and the infiltration of immune cells. Overexpression of the transcription factor LIM-domain only protein 4 (LMO4) promoted by IL-23 has critical roles in regulating the proliferation and differentiation of psoriatic keratinocytes. IL-6, an autocrine cytokine in psoriatic epidermis, is a key mediator of IL-23/T helper 17-driven cutaneous inflammation. However, little is known about how IL-6 regulates the up-regulation of LMO4 expression in psoriatic lesions. In this study, human immortalized keratinocyte cells, clinical biopsy specimens, and an animal model of psoriasis induced by imiquimod cream were used to investigate the role of IL-6 in the regulation of keratinocyte proliferation and differentiation. Psoriatic epidermis showed abnormal expression of IL-6 and LMO4. IL-6 up-regulated the expression of LMO4 and promoted keratinocyte proliferation and differentiation. Furthermore, in vitro and in vivo studies showed that IL-6 up-regulates LMO4 expression by activating the mitogen-activated extracellular signal-regulated kinase (MEK)/extracellular signal-regulated kinase (ERK)/NF-κB signaling pathway. These results suggest that IL-6 can activate the NF-κB signaling pathway, up-regulate the expression of LMO4, lead to abnormal proliferation and differentiation of keratinocytes, and promote the occurrence and development of psoriasis.


Subject(s)
Extracellular Signal-Regulated MAP Kinases , Psoriasis , Animals , Humans , Adaptor Proteins, Signal Transducing/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-23/adverse effects , Interleukin-23/metabolism , Interleukin-6/metabolism , Keratinocytes/pathology , LIM Domain Proteins/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Psoriasis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...