Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.393
Filter
2.
Exp Dermatol ; 33(5): e15083, 2024 May.
Article in English | MEDLINE | ID: mdl-38794808

ABSTRACT

Interferons (IFNs) are signalling proteins primarily involved in initiating innate immune responses against pathogens and promoting the maturation of immune cells. Interferon Regulatory Factor 7 (IRF7) plays a pivotal role in the IFNs signalling pathway. The activation process of IRF7 is incited by exogenous or abnormal nucleic acids, which is followed by the identification via pattern recognition receptors (PRRs) and the ensuing signalling cascades. Upon activation, IRF7 modulates the expression of both IFNs and inflammatory gene regulation. As a multifunctional transcription factor, IRF7 is mainly expressed in immune cells, yet its presence is also detected in keratinocytes, fibroblasts, and various dermal cell types. In these cells, IRF7 is critical for skin immunity, inflammation, and fibrosis. IRF7 dysregulation may lead to autoimmune and inflammatory skin conditions, including systemic scleroderma (SSc), systemic lupus erythematosus (SLE), Atopic dermatitis (AD) and Psoriasis. This comprehensive review aims to extensively elucidate the role of IRF7 and its signalling pathways in immune cells and keratinocytes, highlighting its significance in skin-related and connective tissue diseases.


Subject(s)
Connective Tissue Diseases , Interferon Regulatory Factor-7 , Keratinocytes , Signal Transduction , Skin Diseases , Humans , Interferon Regulatory Factor-7/metabolism , Interferon Regulatory Factor-7/genetics , Skin Diseases/immunology , Skin Diseases/metabolism , Keratinocytes/metabolism , Keratinocytes/immunology , Connective Tissue Diseases/metabolism , Connective Tissue Diseases/immunology , Psoriasis/immunology , Psoriasis/metabolism , Animals , Skin/metabolism , Skin/immunology , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/immunology , Scleroderma, Systemic/genetics , Immunity, Innate
3.
Exp Dermatol ; 33(5): e15087, 2024 May.
Article in English | MEDLINE | ID: mdl-38685821

ABSTRACT

Hidradenitis Suppurativa (HS) is a chronic autoinflammatory skin disease with activated keratinocytes, tunnel formation and a complex immune infiltrate in tissue. The HS microbiome is polymicrobial with an abundance of commensal gram-positive facultative (GPs) Staphylococcus species and gram-negative anaerobic (GNA) bacteria like Prevotella, Fusobacterium and Porphyromonas with increasing predominance of GNAs with disease severity. We sought to define the keratinocyte response to bacteria commonly isolated from HS lesions to probe pathogenic relationships between HS and the microbiome. Type strains of Prevotella nigrescens, Prevotella melaninogenica, Prevotella intermedia, Prevotella asaccharolytica, Fusobacterium nucleatum, as well as Staphylococcus aureus and the normal skin commensal Staphylococcus epidermidis were heat-killed and co-incubated with normal human keratinocytes. RNA was collected and analysed using RNAseq and RT-qPCR. The supernatant was collected from cell culture for protein quantification. Transcriptomic profiles between HS clinical samples and stimulated keratinocytes were compared. Co-staining of patient HS frozen sections was used to localize bacteria in lesions. A mouse intradermal injection model was used to investigate early immune recruitment. TLR4 and JAK inhibitors were used to investigate mechanistic avenues of bacterial response inhibition. GNAs, especially F. nucleatum, stimulated vastly higher CXCL8, IL17C, CCL20, IL6, TNF and IL36γ transcription in normal skin keratinocytes than the GPs S. epidermidis and S. aureus. Using RNAseq, we found that F. nucleatum (and Prevotella) strongly induced the IL-17 pathway in keratinocytes and overlapped with transcriptome profiles of HS patient clinical samples. Bacteria were juxtaposed to activated keratinocytes in vivo, and F. nucleatum strongly recruited murine neutrophil and macrophage migration. Both the TLR4 and pan-JAK inhibitors reduced cytokine production. Detailed transcriptomic profiling of healthy skin keratinocytes exposed to GNAs prevalent in HS revealed a potent, extensive inflammatory response vastly stronger than GPs. GNAs stimulated HS-relevant genes, including many genes in the IL-17 response pathway, and were significantly associated with HS tissue transcriptomes. The close association of activated keratinocytes with bacteria in HS lesions and innate infiltration in murine skin cemented GNA pathogenic potential. These novel mechanistic insights could drive future targeted therapies.


Subject(s)
Hidradenitis Suppurativa , Keratinocytes , Keratinocytes/immunology , Keratinocytes/microbiology , Keratinocytes/metabolism , Humans , Animals , Mice , Hidradenitis Suppurativa/microbiology , Hidradenitis Suppurativa/immunology , Staphylococcus aureus/immunology , Staphylococcus epidermidis/immunology , Fusobacterium nucleatum/immunology , Transcriptome , Cytokines/metabolism , Bacteria, Anaerobic , Interleukin-17/metabolism , Microbiota , Prevotella/immunology
4.
Clin Immunol ; 263: 110199, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38565329

ABSTRACT

Cell-cell communication is crucial for regulating signaling and cellular function. However, the precise cellular and molecular changes remain poorly understood in skin aging. Based on single-cell and bulk RNA data, we explored the role of cell-cell ligand-receptor interaction in skin aging. We found that the macrophage migration inhibitory factor (MIF)/CD74 ligand-receptor complex was significantly upregulatedin aged skin, showing the predominant paracrine effect of keratinocytes on fibroblasts. Enrichment analysis and in vitro experiment revealed a close association of the activation of the MIF/CD74 with inflammatory pathways and immune response. Mechanistically, MIF/CD74 could significantly inhibit PPARγ protein, which thus significantly increased the degree of fibroblast senescence, and significantly up-regulated the expression of senescence-associated secretory phenotype (SASP) factors and FOS gene. Therefore, our study reveals that MIF/CD74 inhibits the activation of the PPAR signaling pathway, subsequently inducing the production of SASP factors and the upregulation of FOS expression, ultimately accelerating fibroblast senescence.


Subject(s)
Antigens, Differentiation, B-Lymphocyte , Fibroblasts , Histocompatibility Antigens Class II , Macrophage Migration-Inhibitory Factors , Single-Cell Analysis , Skin Aging , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/metabolism , Humans , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Fibroblasts/metabolism , Skin Aging/genetics , Skin Aging/physiology , Single-Cell Analysis/methods , Signal Transduction , Cellular Senescence/genetics , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Sequence Analysis, RNA , Keratinocytes/metabolism , Keratinocytes/immunology , PPAR gamma/metabolism , PPAR gamma/genetics , Middle Aged , Male , Female , Skin/metabolism , Skin/immunology , Cells, Cultured , Adult
5.
mSystems ; 9(5): e0017924, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38656122

ABSTRACT

The utilization of ATP within cells plays a fundamental role in cellular processes that are essential for the regulation of host-pathogen dynamics and the subsequent immune response. This study focuses on ATP-binding proteins to dissect the complex interplay between Staphylococcus aureus and human cells, particularly macrophages (THP-1) and keratinocytes (HaCaT), during an intracellular infection. A snapshot of the various protein activity and function is provided using a desthiobiotin-ATP probe, which targets ATP-interacting proteins. In S. aureus, we observe enrichment in pathways required for nutrient acquisition, biosynthesis and metabolism of amino acids, and energy metabolism when located inside human cells. Additionally, the direct profiling of the protein activity revealed specific adaptations of S. aureus to the keratinocytes and macrophages. Mapping the differentially activated proteins to biochemical pathways in the human cells with intracellular bacteria revealed cell-type-specific adaptations to bacterial challenges where THP-1 cells prioritized immune defenses, autophagic cell death, and inflammation. In contrast, HaCaT cells emphasized barrier integrity and immune activation. We also observe bacterial modulation of host processes and metabolic shifts. These findings offer valuable insights into the dynamics of S. aureus-host cell interactions, shedding light on modulating host immune responses to S. aureus, which could involve developing immunomodulatory therapies. IMPORTANCE: This study uses a chemoproteomic approach to target active ATP-interacting proteins and examines the dynamic proteomic interactions between Staphylococcus aureus and human cell lines THP-1 and HaCaT. It uncovers the distinct responses of macrophages and keratinocytes during bacterial infection. S. aureus demonstrated a tailored response to the intracellular environment of each cell type and adaptation during exposure to professional and non-professional phagocytes. It also highlights strategies employed by S. aureus to persist within host cells. This study offers significant insights into the human cell response to S. aureus infection, illuminating the complex proteomic shifts that underlie the defense mechanisms of macrophages and keratinocytes. Notably, the study underscores the nuanced interplay between the host's metabolic reprogramming and immune strategy, suggesting potential therapeutic targets for enhancing host defense and inhibiting bacterial survival. The findings enhance our understanding of host-pathogen interactions and can inform the development of targeted therapies against S. aureus infections.


Subject(s)
Adenosine Triphosphate , Host-Pathogen Interactions , Keratinocytes , Macrophages , Staphylococcus aureus , Humans , Staphylococcus aureus/metabolism , Adenosine Triphosphate/metabolism , Host-Pathogen Interactions/immunology , Macrophages/microbiology , Macrophages/metabolism , Macrophages/immunology , Keratinocytes/microbiology , Keratinocytes/metabolism , Keratinocytes/immunology , THP-1 Cells , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Proteomics/methods , Bacterial Proteins/metabolism , HaCaT Cells
6.
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
7.
Int Immunopharmacol ; 132: 111993, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38565044

ABSTRACT

OBJECTIVE: Psoriasis is an immune-mediated skin disease where the IL-17 signaling pathway plays a crucial role in its development. Chronic circadian rhythm disorder in psoriasis pathogenesis is gaining more attention. The relationship between IL and 17 signaling pathway and skin clock genes remains poorly understood. METHODS: GSE121212 with psoriatic lesion and healthy controls was used as the exploration cohort for searching analysis. Datasets GSE54456, GSE13355, GSE14905, GSE117239, GSE51440, and GSE137218 were applied to validation analysis. Single-cell RNA sequencing (scRNA-seq) dataset GSE173706 was used to explore the F3 expression and related pathway activities in single-cell levels. Through intersecting with high-expression DEGs, F3 was selected as the signature skin circadian gene in psoriasis for further investigation. Functional analyses, including correlation analyses, prediction of transcription factors, protein-protein interaction, and single gene GSEA to explore the potential roles of F3. ssGSEA algorithm was performed to uncover the immune-related characteristics of psoriasis. We further explored F3 expression in the specific cell population in scRNA-seq dataset, besides this, AUCell analysis was performed to explore the pathway activities and the results were further compared between the specific cell cluster. Immunohistochemistry experiment, RT-qPCR was used to validate the location and expression of F3, small interfering RNA (siRNA) transfection experiment in HaCaT, and transcriptome sequencing analysis were applied to explore the potential function of F3. RESULTS: F3 was significantly down-regulated in psoriasis and interacted with IL-17 signaling pathway. Low expression of F3 could upregulate the receptor of JAK-STAT signaling, thereby promoting keratinocyte inflammation. CONCLUSION: Our research revealed a bidirectional link between the skin circadian gene F3 and the IL-17 signaling pathway in psoriasis, suggesting that F3 may interact with the IL-17 pathway by activating JAK-STAT within keratinocytes and inducing abnormal intracellular inflammation.


Subject(s)
Interleukin-17 , Keratinocytes , Psoriasis , Signal Transduction , Skin , Psoriasis/genetics , Psoriasis/immunology , Humans , Interleukin-17/metabolism , Interleukin-17/genetics , Keratinocytes/metabolism , Keratinocytes/immunology , Skin/pathology , Skin/immunology , Skin/metabolism , Circadian Clocks/genetics , Biomarkers/metabolism , Severity of Illness Index , HaCaT Cells
8.
J Dermatol Sci ; 114(1): 44-51, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38508975

ABSTRACT

BACKGROUND: Bullous pemphigoid (BP) is an antibody-mediated blistering disease predominantly affecting the elderly. The pathogenesis involves both complement-dependent and complement-independent mechanisms. The therapeutic potential of targeting complement-independent mechanism has not yet been determined. The mainstay of treatment, corticosteroid, has many side effects, indicating the needs of better treatments. OBJECTIVE: We tempted to establish an in vitro model of BP which resembles complement-independent mechanism and to examine the therapeutic potential of a novel anti-inflammatory agent, diacerein. METHODS: Cultured HaCaT cells were treated with purified antibodies from BP patients, with or without diacerein to measure the cell interface presence of BP180, protein kinase C, and the production of proinflammatory cytokines. An open-label, randomized, phase 2 trial was conducted to compare topical diacerein and clobetasol ointments in patients with mild-to-moderate BP (NCT03286582). RESULTS: The reduced presentation of BP180 at cell interface after treating with BP autoantibodies was noticed in immunofluorescence and western blotting studies. The phenomenon was restored by diacerein. Diacerein also reduced the autoantibody-induced increase of pro-inflammatory cytokines. Reciprocal changes of BP180 and protein kinase C at the cell interface were found after treating with BP autoantibodies. This phenomenon was also reversed by diacerein in a dose-dependent manner. The phase 2 trial showed that topical diacerein reduced the clinical symptoms which were comparable to those of topical clobetasol. CONCLUSION: Diacerein inhibited BP autoantibody-induced reduction of BP180 and production of proinflammatory cytokines in vitro and showed therapeutic potential in patients with BP. It is a novel drug worthy of further investigations.


Subject(s)
Anthraquinones , Autoantibodies , Cytokines , Non-Fibrillar Collagens , Pemphigoid, Bullous , Humans , Pemphigoid, Bullous/immunology , Pemphigoid, Bullous/drug therapy , Pemphigoid, Bullous/pathology , Anthraquinones/pharmacology , Anthraquinones/therapeutic use , Autoantibodies/immunology , Autoantibodies/blood , Non-Fibrillar Collagens/immunology , Cytokines/metabolism , Cytokines/immunology , Collagen Type XVII , Autoantigens/immunology , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Clobetasol/therapeutic use , Clobetasol/pharmacology , Aged , Male , HaCaT Cells , Female , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase C/immunology , Complement System Proteins/immunology , Cell Line , Treatment Outcome , Keratinocytes/immunology , Keratinocytes/drug effects
9.
Eur J Immunol ; 54(5): e2250279, 2024 May.
Article in English | MEDLINE | ID: mdl-38356200

ABSTRACT

Although biologics have achieved tremendous success in the treatment of psoriasis and revolutionized the clinical management of the disease, certain issues arise during treatments, including the phenotypic switch from psoriasis to other skin disorders and the recurrence of psoriasis after the cessation of biologic treatment. Here we provide a concise overview of the roles of keratinocytes in the pathogenesis of psoriasis, elucidate the involvement of keratinocytes in the phenotypic switch and relapse of psoriasis, and address the challenges encountered in both basic and clinical research on psoriasis.


Subject(s)
Keratinocytes , Phenotype , Psoriasis , Recurrence , Psoriasis/immunology , Psoriasis/pathology , Humans , Keratinocytes/immunology , Animals
10.
J Innate Immun ; 15(1): 822-835, 2023.
Article in English | MEDLINE | ID: mdl-37903473

ABSTRACT

INTRODUCTION: CircRNAs are closely related to many human diseases; however, their role in acne remains unclear. This study aimed to determine the role of hsa_circ_0102678 in regulating inflammation of acne. METHODS: First, microarray analysis was performed to study the expression of circRNAs in acne. Subsequently, RNase R digestion assay and fluorescence in situ hybridization assay were utilized to confirm the characteristics of hsa_circ_0102678. Finally, qRT-PCR, Western blotting analysis, immunoprecipitation, luciferase reporter assay, circRNA probe pull-down assay, biotin-labeled miRNA pull-down assay, RNA immunoprecipitation assay, and m6A dot blot assay were utilized to reveal the functional roles of hsa_circ_0102678 on inflammation induced by C. acnes biofilm in human primary keratinocytes. RESULTS: Our investigations showed that the expression of hsa_circ_0102678 was significantly decreased in acne tissues, and hsa_circ_0102678 was a type of circRNAs, which was mainly localized in the cytoplasm of primary human keratinocytes. Moreover, hsa_circ_0102678 remarkably affected the expression of IL-8, IL-6, and TNF-α, which induced by C. acnes biofilm. Importantly, mechanistic studies indicated that the YTHDC1 could bind directly to hsa_circ_0102678 and promote the export of N6-methyladenosine-modified hsa_circ_0102678 to the cytoplasm. Besides, hsa_circ_0102678 could bind to miR-146a and sponge miR-146a to promote the expression of IRAK1 and TRAF6. CONCLUSION: Our findings revealed a previously unknown process by which hsa_circ_0102678 promoted keratinocyte inflammation induced by C. acnes biofilm via regulating miR-146a/TRAF6 and IRAK1 axis.


Subject(s)
Acne Vulgaris , Intracellular Signaling Peptides and Proteins , Nerve Tissue Proteins , Propionibacteriaceae , RNA Splicing Factors , RNA, Circular , Humans , Propionibacteriaceae/physiology , Acne Vulgaris/immunology , Acne Vulgaris/microbiology , Cells, Cultured , Keratinocytes/immunology , Keratinocytes/microbiology , RNA, Circular/genetics , Down-Regulation , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Biological Transport, Active , RNA Splicing Factors/metabolism , Nerve Tissue Proteins/metabolism
11.
Front Immunol ; 13: 884248, 2022.
Article in English | MEDLINE | ID: mdl-35844545

ABSTRACT

The severe autoimmune blistering disease Pemphigus vulgaris (PV) is mainly caused by autoantibodies (IgG) against desmoglein (Dsg) 3 and Dsg1. The mechanisms leading to the development of blisters are not fully understood, but intracellular signaling seems to play an important role. Sheddases ADAM10 and ADAM17 are involved in the turnover of the desmosomal cadherin Dsg2 and ADAM10 has been shown to contribute to acantholysis in a murine pemphigus model. In the present study, we further examined the role of ADAM10 and ADAM17 both in keratinocyte adhesion and in the pathogenesis of PV. First, we found that inhibition of ADAM10 enhanced adhesion of primary human keratinocytes but not of immortalized keratinocytes. In dissociation assays, inhibition of ADAM10 shifted keratinocyte adhesion towards a hyperadhesive state. However, ADAM inhibition did neither modulate protein levels of Dsg1 and Dsg3 nor activation of EGFR at Y1068 and Y845. In primary human keratinocytes, inhibition of ADAM10, but not ADAM17, reduced loss of cell adhesion and fragmentation of Dsg1 and Dsg3 immunostaining in response to a PV1-IgG from a mucocutaneous PV patient. Similarly, inhibition of ADAM10 in dissociation assay decreased fragmentation of primary keratinocytes induced by a monoclonal antibody against Dsg3 and by PV-IgG from two other patients both suffering from mucosal PV. However, such protective effect was not observed in both cultured cells and ex vivo disease models, when another mucocutaneous PV4-IgG containing more Dsg1 autoantibodies was used. Taken together, ADAM10 modulates both hyperadhesion and PV-IgG-induced loss of cell adhesion dependent on the autoantibody profile.


Subject(s)
ADAM10 Protein , ADAM17 Protein , Keratinocytes , Pemphigus , ADAM10 Protein/immunology , ADAM17 Protein/immunology , Amyloid Precursor Protein Secretases , Animals , Autoantibodies/immunology , Cell Adhesion/immunology , Desmoglein 1/immunology , Desmoglein 3/immunology , Humans , Immunoglobulin G/immunology , Keratinocytes/immunology , Keratinocytes/pathology , Membrane Proteins/metabolism , Mice , Pemphigus/immunology , Pemphigus/pathology
12.
Proc Natl Acad Sci U S A ; 119(25): e2116027119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35704759

ABSTRACT

The epidermis is the outermost layer of the skin and the body's primary barrier to external pathogens; however, the early epidermal immune response remains to be mechanistically understood. We show that the chemokine CXCL14, produced by epidermal keratinocytes, exhibits robust circadian fluctuations and initiates innate immunity. Clearance of the skin pathogen Staphylococcus aureus in nocturnal mice was associated with CXCL14 expression, which was high during subjective daytime and low at night. In contrast, in marmosets, a diurnal primate, circadian CXCL14 expression was reversed. Rhythmically expressed CXCL14 binds to S. aureus DNA and induces inflammatory cytokine production by activating Toll-like receptor (TLR)9-dependent innate pathways in dendritic cells and macrophages underneath the epidermis. CXCL14 also promoted phagocytosis by macrophages in a TLR9-independent manner. These data indicate that circadian production of the epidermal chemokine CXCL14 rhythmically suppresses skin bacterial proliferation in mammals by activating the innate immune system.


Subject(s)
Epidermis , Immunity, Innate , Skin Diseases, Bacterial , Animals , Chemokines, CXC/genetics , Chemokines, CXC/immunology , Circadian Clocks/immunology , Epidermis/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Keratinocytes/immunology , Mammals , Mice , Skin Diseases, Bacterial/immunology , Skin Diseases, Bacterial/metabolism , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology
13.
J Invest Dermatol ; 142(2): 402-413, 2022 02.
Article in English | MEDLINE | ID: mdl-34333017

ABSTRACT

Dermal fibroblasts are strategically positioned underneath the basal epidermis layer to support keratinocyte proliferation and extracellular matrix production. In inflammatory conditions, these fibroblasts produce cytokines and chemokines that promote the chemoattraction of immune cells into the dermis and the hyperplasia of the epidermis, two characteristic hallmarks of psoriasis. However, how dermal fibroblasts specifically contribute to psoriasis development remains largely uncharacterized. In this study, we investigated through which cytokines and signaling pathways dermal fibroblasts contribute to the inflammatory features of psoriatic skin. We show that dermal fibroblasts from lesional psoriatic skin are important producers of inflammatory mediators, including IL-6, CXCL8, and CXCL2. This increased cytokine production was found to be regulated by ZFP36 family members ZFP36, ZFP36L1, and ZFP36L2, RNA-binding proteins with mRNA-degrading properties. In addition, the expression of ZFP36 family proteins was found to be reduced in chronic inflammatory conditions that mimic psoriatic lesional skin. Collectively, these results indicate that dermal fibroblasts are important producers of cytokines in psoriatic skin and that reduced expression of ZFP36 members in psoriasis dermal fibroblasts contributes to their inflammatory phenotype.


Subject(s)
Butyrate Response Factor 1/metabolism , Fibroblasts/metabolism , Psoriasis/immunology , Transcription Factors/metabolism , Tristetraprolin/metabolism , Biopsy , Butyrate Response Factor 1/genetics , Case-Control Studies , Epidermis/immunology , Epidermis/metabolism , Epidermis/pathology , Gene Knockdown Techniques , Healthy Volunteers , Humans , Inflammation Mediators/metabolism , Keratinocytes/immunology , Keratinocytes/metabolism , Psoriasis/pathology , Transcription Factors/genetics , Tristetraprolin/genetics
14.
J Invest Dermatol ; 142(2): 390-401, 2022 02.
Article in English | MEDLINE | ID: mdl-34293351

ABSTRACT

Psoriasis is a chronic immune-mediated disease characterized by excessive proliferation of epidermal keratinocytes and increased immune cell infiltration to the skin. Although it is well-known that psoriasis pathogenesis is driven by aberrant production of proinflammatory cytokines, the mechanisms underlying the imbalance between proinflammatory and anti-inflammatory cytokine expression are incompletely understood. In this study, we report that the transcriptional coregulators CtBP1 and 2 can transactivate a common set of proinflammatory genes both in the skin of imiquimod-induced mouse psoriasis model and in human keratinocytes and macrophages stimulated by imiquimod. We find that mice overexpressing CtBP1 in epidermal keratinocytes display severe skin inflammation phenotypes with increased expression of T helper type 1 and T helper type 17 cytokines. We also find that the expression of CtBPs and CtBP-target genes is elevated both in human psoriatic lesions and in the mouse imiquimod psoriasis model. Moreover, we were able to show that topical treatment with a peptidic inhibitor of CtBP effectively suppresses the CtBP-regulated proinflammatory gene expression and thus attenuates psoriatic inflammation in the imiquimod mouse model. Together, our findings suggest to our knowledge previously unreported strategies for therapeutic modulation of the immune response in inflammatory skin diseases.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Psoriasis/drug therapy , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , HaCaT Cells , Humans , Imiquimod/immunology , Keratinocytes/drug effects , Keratinocytes/immunology , Keratinocytes/pathology , Mice , Mice, Transgenic , Psoriasis/genetics , Psoriasis/immunology , Psoriasis/pathology , Transcriptional Activation/drug effects , Transcriptional Activation/immunology
15.
J Leukoc Biol ; 111(1): 135-145, 2022 01.
Article in English | MEDLINE | ID: mdl-33847413

ABSTRACT

Tissue-resident γδ T cells form the first line of defense at barrier surfaces where they survey host tissue for signs of stress or damage. Following recognition of injury, γδ T cells play a crucial role in the wound-healing response through the production of growth factors and cytokines that promote proliferation in surrounding epithelial cells. To initiate this response, γδ T cells require interactions with a variety of epithelial-expressed costimulatory molecules in addition to primary signaling through their TCR. In the epidermis these signals include the coxsackie and adenovirus receptor (CAR), histocompatibility antigen 60c (H60c), and plexin B2, which interact with γδ T cell-expressed junctional adhesion molecule-like protein (JAML), NKG2D, and CD100, respectively. Here we identify heat shock protein family A member 8 (Hspa8) and ICAM-1 as two additional keratinocyte-expressed costimulatory molecules for epidermal resident γδ T cells (termed DETC). These molecules were rapidly up-regulated in the epidermis following wounding in both mouse and human tissue. Both Hspa8 and ICAM-1 had a costimulatory effect on DETC, inducing proliferation, CD25 up-regulation, and IL-2 production. We also provide evidence that DETC can be activated through the potential ICAM-1 and Hspa8 receptors LFA-1 and CD316. Finally, knockdown of Hspa8 in keratinocytes reduced their ability to activate DETC in culture and ICAM-1-/- mice exhibited impaired rates of healing in skin-organ culture suggesting a role for these proteins in the DETC-mediated damage response. Together with previous work on CAR, H60c, and plexin B2, these results add to a picture of a complex keratinocyte wound signature that is required for efficient DETC activation.


Subject(s)
HSC70 Heat-Shock Proteins/immunology , Intercellular Adhesion Molecule-1/immunology , Lymphocyte Activation , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Animals , Cell Proliferation , Cells, Cultured , Humans , Keratinocytes/immunology , Mice, Inbred C57BL , T-Lymphocytes/cytology
16.
Oral Dis ; 28(1): 150-163, 2022 Jan.
Article in English | MEDLINE | ID: mdl-33200485

ABSTRACT

OBJECTIVE: Double-strand (ds) DNA-enveloped viruses can cause oral infection. Our aim is to investigate whether oral mucosal cells participate in immune response against cytosolic dsDNA invasion. METHODS: We examined the response to transfected herpes simplex virus (HSV) dsDNA via intracellular receptors in oral keratinocytes (RT7) and fibroblasts (GT1), and the effect of TNF-α on those responses. RESULTS: Transfected dsDNA increased CXCL10 expression via NF-κB activation in both cell types, while those responses were inhibited by knockdown of RIG-I, an RNA sensor. Although IFI16, a DNA sensor, was expressed in the nuclei of both types, its knockdown decreased transfected dsDNA-induced CXCL10 expression in GT1 but not RT7 cells. IFI16 in GT1 cells was translocated into cytoplasm from nuclei, which was attributed to immune response to cytosolic dsDNA. TNF-α enhanced transfected dsDNA-induced CXCL10, and knockdown of IFI16 decreased TNF-α and dsDNA-driven CXCL10 expression in both RT7 and GT1 cells. Finally, the combination of TNF-α and transfected dsDNA resulted in translocation of IFI16 from nuclei to cytoplasm in RT7 cells. CONCLUSION: RIG-I and IFI16 in oral mucosal cells may play important roles in host immune response against DNA viral infection, while TNF-α contributes to development of an antiviral system via those intracellular receptors.


Subject(s)
DNA, Viral/immunology , Fibroblasts , Keratinocytes , Simplexvirus/immunology , Antiviral Restriction Factors/immunology , Cell Line , Chemokine CXCL10/immunology , Cytoplasm , Fibroblasts/immunology , Humans , Immunity , Keratinocytes/immunology , Nuclear Proteins/immunology , Phosphoproteins/immunology , Receptors, Retinoic Acid/immunology , Tumor Necrosis Factor-alpha/immunology
17.
J Clin Invest ; 132(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-34905516

ABSTRACT

Desmoglein 1 (Dsg1) is a cadherin restricted to stratified tissues of terrestrial vertebrates, which serve as essential physical and immune barriers. Dsg1 loss-of-function mutations in humans result in skin lesions and multiple allergies, and isolated patient keratinocytes exhibit increased proallergic cytokine expression. However, the mechanism by which genetic deficiency of Dsg1 causes chronic inflammation is unknown. To determine the systemic response to Dsg1 loss, we deleted the 3 tandem Dsg1 genes in mice. Whole transcriptome analysis of embryonic Dsg1-/- skin showed a delay in expression of adhesion/differentiation/keratinization genes at E17.5, a subset of which recovered or increased by E18.5. Comparing epidermal transcriptomes from Dsg1-deficient mice and humans revealed a shared IL-17-skewed inflammatory signature. Although the impaired intercellular adhesion observed in Dsg1-/- mice resembles that resulting from anti-Dsg1 pemphigus foliaceus antibodies, pemphigus skin lesions exhibit a weaker IL-17 signature. Consistent with the clinical importance of these findings, treatment of 2 Dsg1-deficient patients with an IL-12/IL-23 antagonist originally developed for psoriasis resulted in improvement of skin lesions. Thus, beyond impairing the physical barrier, loss of Dsg1 function through gene mutation results in a psoriatic-like inflammatory signature before birth, and treatment with a targeted therapy significantly improved skin lesions in patients.


Subject(s)
Desmoglein 1/immunology , Desmosomes/immunology , Keratinocytes/immunology , Pemphigus/immunology , Th17 Cells/immunology , Animals , Desmoglein 1/genetics , Desmosomes/genetics , Mice , Pemphigus/genetics
18.
Biochem Biophys Res Commun ; 586: 100-106, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34837833

ABSTRACT

Lipopolysaccharide (LPS) is the principal component of the outer membrane of gram-negative bacteria. The prior oral administration of LPS attenuates inflammatory responses, such as intestinal injury and atopic dermatitis, in mouse models; however, the underlying mechanism remains unclear. Here, we examined the effect of topical LPS application on allergic contact dermatitis and its mechanism of action using a murine contact hypersensitivity (CHS) model. Prolonged LPS application to the skin significantly suppressed 2,4-dinitrofluorobenzene (DNFB)-induced CHS. LPS application to the skin also reduced the phagocytosis of fluorescein isothiocyanate (FITC)-dextran by Langerhans and dendritic cells. Cutaneous cell migration into the skin-draining lymph nodes (LNs) induced by FITC painting was reduced by LPS application. During the CHS response, DNFB application induced T-cell proliferation and inflammatory cytokine production in skin-draining LNs, whereas prolonged LPS application inhibited DNFB-induced T-cell growth and interferon gamma production, indicating suppression of DNFB-induced sensitization. These results suggest that prolonged LPS application suppressed DNFB-induced sensitization and subsequently CHS response. Our findings imply that topical application of LPS may prevent allergic dermatitis such as CHS.


Subject(s)
Dermatitis, Contact/drug therapy , Immunologic Factors/pharmacology , Lipopolysaccharides/pharmacology , Lymphocytes/drug effects , Skin/drug effects , Administration, Cutaneous , Animals , Cell Line , Cell Movement/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dermatitis, Contact/etiology , Dermatitis, Contact/immunology , Dermatitis, Contact/pathology , Dextrans/metabolism , Dinitrofluorobenzene/administration & dosage , Ear , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/immunology , Langerhans Cells/cytology , Langerhans Cells/drug effects , Langerhans Cells/immunology , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Activation/drug effects , Lymphocytes/cytology , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Phagocytosis/drug effects , Primary Cell Culture , Skin/immunology , Skin/pathology
19.
Front Immunol ; 12: 772516, 2021.
Article in English | MEDLINE | ID: mdl-34938293

ABSTRACT

Although the role of epidermal cells in skin regeneration has been extensively documented, their functions in immunity and tolerance mechanisms are largely underestimated. The aim of the present review was to outline the state of knowledge on resident immune cells of hematopoietic origin hosted in the epidermis, and then to focus on the involvement of keratinocytes in the complex skin immune networks acting in homeostasis and regeneration conditions. Based on this knowledge, the mechanisms of immune tolerance are reviewed. In particular, strategies based on immunosuppression mediated by HLA-G are highlighted, as recent advances in this field open up perspectives in epidermis-substitute bioengineering for temporary and permanent skin replacement strategies.


Subject(s)
HLA-G Antigens/immunology , Keratinocytes/immunology , Skin/immunology , Animals , Genetic Therapy , Homeostasis , Humans , Immune Tolerance , Skin/cytology
20.
Front Immunol ; 12: 773720, 2021.
Article in English | MEDLINE | ID: mdl-34899726

ABSTRACT

Anti-laminin 332 mucous membrane pemphigoid (MMP) is an autoimmune blistering disease characterized by predominant mucosal lesions and autoantibodies against laminin 332. The exact diagnosis of anti-laminin 332 MMP is important since nearly 30% of patients develop solid cancers. This study compared two independently developed diagnostic indirect immunofluorescence (IF) tests based on recombinant laminin 332 expressed in HEK239 cells (biochip mosaic assay) and the migration trails of cultured keratinocytes rich in laminin 332 (footprint assay). The sera of 54 anti-laminin 332 MMP, 35 non-anti-laminin 332 MMP, and 30 pemphigus vulgaris patients as well as 20 healthy blood donors were analyzed blindly and independently. Fifty-two of 54 and 54/54 anti-laminin 332 MMP sera were positive in the biochip mosaic and the footprint assay, respectively. In the 35 non-anti-laminin 332 MMP sera, 3 were positive in both tests and 4 others showed weak reactivity in the footprint assay. In conclusion, both assays are easy to perform, highly sensitive, and specific, which will further facilitate the diagnosis of anti-laminin 332 MMP.


Subject(s)
Autoantibodies/blood , Autoantigens/immunology , Biological Assay , Cell Adhesion Molecules/immunology , Pemphigoid, Benign Mucous Membrane/diagnosis , Adult , Aged , Aged, 80 and over , Autoantigens/genetics , Autoantigens/metabolism , Biomarkers/blood , Case-Control Studies , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Female , Fluorescent Antibody Technique, Indirect , Germany , HEK293 Cells , Humans , Japan , Keratinocytes/immunology , Keratinocytes/metabolism , Male , Middle Aged , Netherlands , Pemphigoid, Benign Mucous Membrane/blood , Pemphigoid, Benign Mucous Membrane/immunology , Predictive Value of Tests , Reproducibility of Results , Kalinin
SELECTION OF CITATIONS
SEARCH DETAIL
...