Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167195, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38648901

ABSTRACT

Acne is a common chronic inflammatory disease of the pilosebaceous unit. Transient receptor potential vanilloid 3 (TRPV3) is an ion channel that is involved in inflammatory dermatosis development. However, the involvement of TRPV3 in acne-related inflammation remains unclear. Here, we used acne-like mice and human sebocytes to examine the role of TRPV3 in the development of acne. We found that TRPV3 expression increased in the skin lesions of Propionibacterium acnes (P. acnes)-injected acne-like mice and the facial sebaceous glands (SGs) of acne patients. TRPV3 promoted inflammatory cytokines and chemokines secretion in human sebocytes and led to neutrophil infiltration surrounding the SGs in acne lesions, further exacerbating sebaceous inflammation and participating in acne development. Mechanistically, TRPV3 enhanced TLR2 level by promoting transcriptional factor phosphorylated-FOS-like antigen-1 (p-FOSL1) expression and its binding to the TLR2 promoter, leading to TLR2 upregulation and downstream NF-κB signaling activation. Genetic or pharmacological inhibition of TRPV3 both alleviated acne-like skin inflammation in mice via the TLR2-NF-κB axis. Thus, our study revealed the critical role of TRPV3 in sebaceous inflammation and indicated its potential as an acne therapeutic target.


Subject(s)
Acne Vulgaris , Sebaceous Glands , TRPV Cation Channels , Toll-Like Receptor 2 , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/genetics , Animals , Acne Vulgaris/metabolism , Acne Vulgaris/pathology , Acne Vulgaris/genetics , Acne Vulgaris/immunology , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Humans , Mice , Sebaceous Glands/metabolism , Sebaceous Glands/pathology , Sebaceous Glands/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics , Propionibacterium acnes , Male , NF-kappa B/metabolism , Signal Transduction , Mice, Inbred C57BL , Female
2.
Sci Rep ; 11(1): 21510, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34728702

ABSTRACT

Activation of Toll-like receptors (TLR) 1/2 and 4 are central in inducing inflammation in sebocytes by regulating the expression of protein coding mRNAs, however the microRNA (miRNA) profile in response to TLR activation and thus the possible role of miRNAs in modulating sebocyte functions has not been elucidated. In this work we identified miR-146a to have the highest induction in the TLR1/2 and 4 activated SZ95 sebocytes and found that its increased levels led to the down-regulation of IL-8 secretion, decreased the chemoattractant potential and stimulated the proliferation of sebocytes. Assessing the gene expression profile of SZ95 sebocytes treated with a miR-146a inhibitor, the induction of GNG7 was one of the highest, while when cells were treated with a miR-146a mimic, the expression of GNG7 was down-regulated. These findings correlated with our in situ hybridization results, that compared with control, miR-146a showed an increased, while GNG7 a decreased expression in sebaceous glands of acne samples. Further studies revealed, that when inhibiting the levels of GNG7 in SZ95 sebocytes, cells increased their lipid content and decreased their proliferation. Our findings suggest, that miR-146a could be a potential player in acne pathogenesis by regulating inflammation, inducing proliferation and, through the indirect down-regulation of GNG7, promoting the lipid production of sebocytes.


Subject(s)
Acne Vulgaris/pathology , GTP-Binding Protein gamma Subunits/metabolism , Inflammation/pathology , Lipids/analysis , Lipogenesis , MicroRNAs/genetics , Sebaceous Glands/pathology , Acne Vulgaris/genetics , Acne Vulgaris/immunology , Acne Vulgaris/metabolism , Adult , Case-Control Studies , Cell Proliferation , Cells, Cultured , GTP-Binding Protein gamma Subunits/genetics , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Male , RNA-Seq , Sebaceous Glands/immunology , Sebaceous Glands/metabolism , Toll-Like Receptor 1/genetics , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
3.
J Dermatol Sci ; 103(3): 156-166, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34334258

ABSTRACT

BACKGROUND: Acne vulgaris is a prevalent skin disease lacking effective and well-tolerated treatment. An earlier study indicated that resveratrol (RVT) has therapeutic effects in acne patients through unknown mechanisms. OBJECTIVES: To evaluate the effects of RVT on linoleic acid (LA)-induced lipogenesis and peptidoglycan (PGN)-induced inflammation in cultured SZ95 sebocytes in vitro, and to investigate the underlying mechanisms. METHODS: RNA-sequencing was used to analyze the whole transcriptome. Nile red staining was used to detect intracellular neutral lipids, whereas lipidomics was used to investigate changes in the lipid profile in sebocytes. Interleukin (IL)-1ß and IL-6 mRNA and protein levels were assessed through quantitative real-time PCR and Enzyme-linked immunosorbent assay, respectively. Western blot was used to evaluate the expression of lipogenesis-related proteins, the inflammatory signaling pathway, and the AMP-activated protein kinase (AMPK) pathway. Further, specific small interfering RNA (siRNA) was used to knockdown sirtuin-1 (SIRT1) expression. RESULTS: RVT inhibited the lipogenesis-related pathway and nuclear factor-kappa B (NF-κB) signaling pathway in SZ95 sebocytes. It also downregulated LA-induced lipogenesis, the expression of lipid-related proteins, and the contents of unsaturated fatty acids. Besides, RVT promoted SIRT1 expression and deacetylation of the NF-κB p65 subunit, thereby lowering IL-1ß and IL-6 secretion under PGN induction. Furthermore, pretreatment with AMPK inhibitor Compound C abolished RVT-mediated sebosuppressive and anti-inflammation effects. Meanwhile, SIRT1 silencing abrogated the anti-inflammatory potential of RVT. CONCLUSION: In human SZ95 sebocytes, RVT exhibits sebosuppressive and anti-inflammatory effects partially through the AMPK pathway, which may justify the role of RVT treatment in acne vulgaris.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Acne Vulgaris/drug therapy , Anti-Inflammatory Agents/pharmacology , Resveratrol/pharmacology , Sebaceous Glands/drug effects , Acne Vulgaris/immunology , Acne Vulgaris/pathology , Anti-Inflammatory Agents/therapeutic use , Cell Line , Gene Knockdown Techniques , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/pathology , Linoleic Acid/pharmacology , Lipogenesis/drug effects , Lipogenesis/immunology , Peptidoglycan/immunology , Resveratrol/therapeutic use , Sebaceous Glands/cytology , Sebaceous Glands/immunology , Sebaceous Glands/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Sirtuin 1/genetics , Sirtuin 1/metabolism
4.
Front Immunol ; 12: 600017, 2021.
Article in English | MEDLINE | ID: mdl-34025636

ABSTRACT

Epidermal growth factor (EGF) acts as a paracrine and autocrine mediator of cell proliferation and differentiation in various types of epithelial cells, such as sebocytes, which produce the lipid-rich sebum to moisturize the skin. However, sebum lipids via direct contact and by penetrating through the epidermis may have regulatory roles on epidermal and dermal cells as well. As EGF receptor (EGFR) is expressed throughout the proliferating and the lipid-producing layers of sebaceous glands (SGs) in healthy and acne-involved skin, we investigated the effect of EGF on SZ95 sebocytes and how it may alter the changes induced by palmitic acid (PA), a major sebum component with bioactive roles. We found that EGF is not only a potent stimulator of sebocyte proliferation, but also induces the secretion of interleukin (IL)6 and down-regulates the expression of genes involved in steroid and retinoid metabolism. Importantly, when applied in combination with PA, the PA-induced lipid accumulation was decreased and the cells secreted increased IL6 levels. Functional clustering of the differentially regulated genes in SZ95 sebocytes treated with EGF, PA or co-treated with EGF+PA further confirmed that EGF may be a potent inducer of hyperproliferative/inflammatory pathways (IL1 signaling), an effect being more pronounced in the presence of PA. However, while a group of inflammatory genes was up-regulated significantly in EGF+PA co-treated sebocytes, PA treatment in the absence of EGF, regulated genes only related to cell homeostasis. Meta-analysis of the gene expression profiles of whole acne tissue samples and EGF- and EGF+PA -treated SZ95 sebocytes showed that the EGF+PA co-activation of sebocytes may also have implications in disease. Altogether, our results reveal that PA-induced lipid accumulation and inflammation can be modulated by EGF in sebocytes, which also highlights the need for system biological approaches to better understand sebaceous (immuno)biology.


Subject(s)
Epidermal Growth Factor/immunology , Epithelial Cells/immunology , Palmitic Acid/pharmacology , Sebaceous Glands/immunology , Signal Transduction/drug effects , Cell Line , Epithelial Cells/pathology , Humans , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Interleukin-6/immunology , Palmitic Acid/adverse effects , Sebaceous Glands/pathology
5.
JAMA Dermatol ; 156(12): 1307-1314, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33146669

ABSTRACT

Importance: Risk of sebaceous carcinoma (SC), a rare skin cancer associated with Muir-Torre syndrome, is elevated among solid organ transplant recipients (SOTRs). However, population studies evaluating this association and assessing survival for posttransplant cases are lacking, and further understanding of SC epidemiology in this immunosuppressed population could provide etiologic and clinical insights. Objective: To assess SC incidence and patient survival after solid organ transplantation. Design, Setting, and Participants: This cohort study, conducted from January 1, 1987, to December 31, 2017, used data from the Transplant Cancer Match Study, which links transplant and cancer registry data for 17 states and 1 metropolitan area in the United States. Altogether, these registries account for approximately 46% of all US transplants. Data on demographic and transplant characteristics as well as induction and initial maintenance immunosuppressive therapies were obtained from the transplant registry. Standardized incidence ratios (SIRs) comparing SC incidence among SOTRs to the general population were calculated. Incidence rate ratios (IRRs) comparing SC risk between SOTR subgroups were calculated using multivariate Poisson regression. Cox regression was used to compare overall survival between SC cases in SOTRs and other individuals. Main Outcomes and Measures: Sebaceous carcinoma incidence and overall patient survival after transplantation compared with the general population. Results: A total of 326 282 transplant procedures were performed for 301 075 patients (No. [%] age at transplant, 126 550 [38.8%] aged 0-44 years; 82 394 [25.3%] aged 45-54 years; 82 082 [25.5%] aged 55-64 years; 35 256 [10.8%] aged ≥65 years; 201 354 male patients [61.7%]; 202 557 White patients [62.1%]). A total of 102 SCs were diagnosed in 301 075 SOTRs, corresponding to a 25-fold increased incidence (SIR, 24.8; 95% CI, 20.2-30.1). Incidence was especially elevated among lung recipients (SIR, 47.7; 95% CI, 20.6-94.0) and after a posttransplant diagnosis of cutaneous squamous cell carcinoma (SIR, 104.0; 95% CI, 62.8-163.0). Among SOTRs, factors independently associated with SC risk included male sex (IRR, 2.46; 95% CI, 1.48-4.07; P < .001), race/ethnicity (non-Hispanic Black vs non-Hispanic White, IRR, 0.28; 95% CI, 0.10-0.77; P = .01), older age (IRR, 7.85; 95% CI, 3.85-16.0; ≥65 vs 0-44 years; P < .001 for trend), use of thymoglobulin induction (IRR, 1.82; 95% CI, 1.16-2.86; P = .009), posttransplant cutaneous squamous cell carcinoma (IRR, 4.60; 95% CI, 2.67-7.94; P < .001), and longer time since transplant (IRR, 8.40; 95% CI, 3.94-17.90; ≥10 vs 0-1.9 years; P < .001 for trend). Muir-Torre syndrome-associated cancers were rare among both SOTRs and others with SC (3.3%-4.1%). Among patients with SC, prior transplantation was associated with increased overall mortality (adjusted hazard ratio, 2.09; 95% CI, 1.45-3.01), although few deaths were attributed to SC (4 of 92 SOTRs [4.3%]; 235 of 3585 non-SOTRs [6.6%]). Conclusions and Relevance: Among SOTRs, results of this large cohort study suggest that SC was associated with measures of immunosuppression, and overall survival was worse than for other patients with SC. Findings also suggest a possible role for UV radiation in carcinogenesis.


Subject(s)
Carcinoma, Squamous Cell/epidemiology , Muir-Torre Syndrome/epidemiology , Organ Transplantation/adverse effects , Sebaceous Gland Neoplasms/epidemiology , Transplant Recipients/statistics & numerical data , Adolescent , Adult , Aged , Carcinogenesis/immunology , Carcinogenesis/radiation effects , Carcinoma, Squamous Cell/etiology , Child , Child, Preschool , Female , Humans , Immunocompromised Host , Incidence , Infant , Infant, Newborn , Male , Middle Aged , Muir-Torre Syndrome/complications , Registries/statistics & numerical data , Risk Factors , Sebaceous Gland Neoplasms/etiology , Sebaceous Glands/immunology , Sebaceous Glands/pathology , Sebaceous Glands/radiation effects , Survival Analysis , Ultraviolet Rays/adverse effects , United States/epidemiology , Young Adult
6.
J Invest Dermatol ; 140(10): 1909-1918.e8, 2020 10.
Article in English | MEDLINE | ID: mdl-32142797

ABSTRACT

We have shown previously that endocannabinoids promote sebaceous lipogenesis, and sebocytes are involved in the metabolism of the endocannabinoid-like substance oleoylethanolamide (OEA). OEA is an endogenous activator of GPR119, a recently deorphanized receptor, which currently is being investigated as a promising antidiabetic drug target. In this study, we investigated the effects of OEA as well as the expression and role of GPR119 in human sebocytes. We found that OEA promoted differentiation of human SZ95 sebocytes (elevated lipogenesis, enhanced granulation, and the induction of early apoptotic events), and it switched the cells to a proinflammatory phenotype (increased expression and release of several proinflammatory cytokines). Moreover, we could also demonstrate that GPR119 was expressed in human sebocytes, and its small interfering RNA-mediated gene silencing suppressed OEA-induced sebaceous lipogenesis, which was mediated via c-Jun N-terminal kinase, extracellular signal-regulated kinase 1/2, protein kinase B, and CRE-binding protein activation. Finally, our pilot data demonstrated that GPR119 was downregulated in the sebaceous glands of patients with acne, arguing that GPR119 signaling may indeed be disturbed in acne. Collectively, our findings introduce the OEA/GPR119 signaling as a positive regulator of sebocyte differentiation and highlight the possibility that dysregulation of this pathway may contribute to the development of seborrhea and acne.


Subject(s)
Receptors, G-Protein-Coupled/physiology , Sebaceous Glands/cytology , Sebaceous Glands/physiology , Cell Differentiation/drug effects , Cells, Cultured , Cytokines/biosynthesis , Endocannabinoids/pharmacology , Humans , Oleic Acids/pharmacology , PPAR alpha/physiology , Sebaceous Glands/immunology , Signal Transduction/physiology
7.
J Mol Histol ; 50(4): 335-342, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31062203

ABSTRACT

Eccrine sweat glands and hair follicles are two primary skin appendages that serve different functions. Although the two appendages exhibit unique morphological patterns in adults, it is difficult to distinguish them morphologically in the early stages of development and regeneration. To research and compare the development, differentiation and regeneration between eccrine sweat glands and hair follicles/pilosebaceous units, specific antigen markers must be found first. Human skin samples were fixed, paraffin-embedded, and cut. The expression of K5, K7, K8, K14, K27, K31, K73, AE13, α-smooth muscle actin (α-SMA), epithelial membrane antigen (EMA), carcinoembryonic antigen (CEA), Na+/K+-ATPase α and Na+-K+-2Cl cotransporter 1 (NKCC1) in eccrine sweat glands, hair follicles and sebaceous glands was detected by immunofluorescence staining. The results showed that eccrine sweat glands expressed K5, K7, K8, K14, K31, α-SMA, CEA, EMA, Na+/K+-ATPase α and NKCC1, but did not express K27, K73 or K31. Hair follicles expressed K5, K8, K14, K27, K31, K73, α-SMA and AE13, but did not express K7, CEA, Na+/K+-ATPase α or NKCC1. Sebaceous glands expressed K5, K14, K73, and EMA, but did not express K7, K8, K31, α-SMA, CEA, EMA, Na+/K+-ATPase α or NKCC1. We concluded that K7, CEA, Na+/K+-ATPase and NKCC1 can be used as specific markers for eccrine sweat glands, K27 and AE13 can be used as specific markers for hair follicles, and K73 can be used as a specific marker for pilosebaceous unit. These specific markers may contribute to differentiate between eccrine sweat glands and hair follicle/pilosebaceous units.


Subject(s)
Antigens, Surface/analysis , Eccrine Glands/cytology , Hair Follicle/cytology , Sebaceous Glands/cytology , Skin/cytology , Antigens, Surface/immunology , Antigens, Surface/metabolism , Biomarkers/analysis , Eccrine Glands/immunology , Fluorescent Antibody Technique/methods , Hair Follicle/immunology , Humans , Sebaceous Glands/immunology , Skin/immunology
8.
Arch Dermatol Res ; 311(7): 563-571, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31127384

ABSTRACT

When anti-acne alternatives from dietary and plant sources are ingested, systemic alterations of interleukin (IL)-4, IL-10, IL-12 and interferon (IFN)-γ, individually or simultaneously, are induced at a 0.1-10.0-fold (×) range of normal physiological concentrations (1×). However, little is known about the effects of these cytokines on excess sebum, a pathophysiological factor of acne development. In this study, human sebocytes were treated with 0.1-10.0× of IL-4, IL-10, IL-12 and IFN-γ for 3 or 5 days to elucidate the effects on lipid content. Treatment with individual cytokines decreased the lipid content at specific concentrations rather than in a concentration-dependent manner. Specifically, 5.0× of IL-4, 5.0× of IFN-γ (5.0IFN), and 0.5×, 5.0× and 10.0× of IL-10 for 3 days, and 0.5× of IL-4 (0.5IL4) for 5 days decreased lipid content to 87.6-93.0% of the control. Treatment with other concentrations of IL-4, IL-10 and IFN-γ, and 0.1-10.0× of IL-12 did not alter lipid content. Combined treatment with 0.5IL4, 5.0IFN and 0.5× of IL-10 for 3 or 5 days decreased the lipid content more than each individual treatment. However, this effect was more evident after 3 days, in parallel with decreased levels of triglycerides, cholesterol esters and free fatty acids, the major lipid compositions of sebocytes, and decreased protein expression of fatty acid synthase (FAS) and mature sterol response element-binding protein-1 (SREBP-1), the lipogenesis-related factors, without altered cell proliferation. We demonstrated that suppressed IL-4 and IL-10 with enhanced IFN-γ synergistically decreased lipid content and protein expression of FAS and mature SREBP-1 in human sebocytes.


Subject(s)
Fatty Acid Synthases/metabolism , Sebaceous Glands/metabolism , Sebum/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Acne Vulgaris/diet therapy , Acne Vulgaris/immunology , Acne Vulgaris/pathology , Cell Line , Cell Proliferation , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Lipids/analysis , Lipogenesis/immunology , Primary Cell Culture , Sebaceous Glands/cytology , Sebaceous Glands/immunology , Sebum/chemistry , Sebum/immunology
9.
Br J Dermatol ; 181(4): 677-690, 2019 10.
Article in English | MEDLINE | ID: mdl-31056753

ABSTRACT

BACKGROUND: Sebaceous glands (SGs) are appendages of mammalian skin that produce a mixture of lipids known as sebum. Acne vulgaris is an exceptionally common skin condition, characterized by elevated sebum production, altered sebum composition, and the formation of infundibular cysts, called comedones. Comedo-associated SGs are atrophic, suggesting that comedo formation involves abnormal differentiation of progenitor cells that generate the SG and infundibulum: the 'comedo switch'. Understanding the biological processes that govern SG homeostasis promises to highlight potential aetiological mechanisms underlying acne and other SG-associated skin disorders. RESULTS: In this review, we discuss the clinical data, genetic mouse models and in vitro research that have highlighted major hormones, paracrine factors, transcription factors and signalling pathways that control SG homeostasis. These include, but are not limited to androgens, progestogens and oestrogens; retinoids; receptor tyrosine kinases such as ErbB family receptors, fibroblast growth factor receptor 2 and insulin/insulin-like growth factor 1 receptors; peroxisome proliferator-activated receptor γ; aryl hydrocarbon receptor; and the Wnt signalling pathway. Where possible, the cellular and molecular mechanisms by which these regulatory factors control SG biology are indicated, along with considerations as to how they might contribute to acne pathogenesis. CONCLUSIONS: Future research should seek to establish the relative importance, and causative relationships, of altered sebum production, sebum composition, inflammation and abnormal differentiation of sebaceous progenitors to the process of comedo formation in acne. Such an understanding will allow for therapeutic targeting of regulatory factors that control SG homeostasis, with the aim of treating acne.


Subject(s)
Acne Vulgaris/immunology , Sebaceous Glands/pathology , Sebum/metabolism , Acne Vulgaris/pathology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Sebaceous Glands/immunology , Sebaceous Glands/metabolism , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/immunology
10.
Arch Dermatol Res ; 311(5): 337-349, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30859308

ABSTRACT

Acne vulgaris is a cutaneous chronic inflammatory disorder with complex pathogenesis. Four factors play vital roles in acne pathophysiology: hyperseborrhea and dysseborrhea, altered keratinization of the pilosebaceous duct, Cutibacterium acnes (C. acnes) and inflammation. The main hormones responsible for the development of acne vulgaris include androgens, insulin and insulin-like growth factor-1. Other factors involved in this process are corticotropin-releasing hormone, α-melanocyte-stimulating hormone and substance P. Wnt/ß-catenin signaling pathway, phosphoinositide 3-kinase (PI3K)/Akt pathway, mitogen-activated protein kinase pathway, adenosine 5'-monophosphate-activated protein kinase pathway and nuclear factor kappa B pathway participate in the modulation of sebocyte, keratinocyte and inflammatory cell (e.g. lymphocytes, monocytes, macrophages, neutrophils) activity. Among all the triggers and pathways mentioned above, IGF-1-induced PI3K/Akt/Forkhead box protein O1/mammalian target of rapamycin (mTOR) C1 pathway is the most important signaling responsible for acne pathogenesis. Commonly used anti-acne agents include retinoids, benzoyl peroxide, antibiotics and hormonal agents (e.g. spironolactone, combination oral contraceptive and flutamide). New approaches including peroxisome proliferator-activated receptor γ modifier, melanocortin receptor antagonists, epigallocatechin-3-gallate, metformin, olumacostat glasaretil, stearoyl-CoA desaturase inhibitor omiganan pentahydrochloride, KDPT, afamelanotide, apremilast and biologics have been developed as promising treatments for acne vulgaris. Although these anti-acne agents have various pharmacological effects against the diverse pathogenesis of acne, all of them have a synergistic mode of action, the attenuation of Akt/mTORC1 signaling and enhancement of p53 signal transduction. In addition to drug therapy, diet with no hyperglycemic carbohydrates, no milk and dairy products is also beneficial for treatment of acne.


Subject(s)
Acne Vulgaris/therapy , Anti-Bacterial Agents/therapeutic use , Dermatologic Agents/therapeutic use , Sebaceous Glands/metabolism , Acne Vulgaris/etiology , Anti-Bacterial Agents/pharmacology , Dairy Products/adverse effects , Dermatologic Agents/pharmacology , Dietary Carbohydrates/adverse effects , Drug Synergism , Humans , Propionibacterium acnes/drug effects , Propionibacterium acnes/immunology , Propionibacterium acnes/isolation & purification , Sebaceous Glands/drug effects , Sebaceous Glands/immunology , Sebum/metabolism , Signal Transduction/drug effects
11.
J Immunol ; 202(6): 1767-1776, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30737272

ABSTRACT

The regulation of cutaneous inflammatory processes is essential for the human skin to maintain homeostasis in the presence of the dense communities of resident microbes that normally populate this organ. Forming the hair follicle-associated sebaceous gland, sebocytes are specialized lipid-producing cells that can release inflammatory mediators. Cytokine and chemokine expression by pilosebaceous epithelial cells (i.e., sebocytes and follicular keratinocytes) has been proposed to contribute to the common human skin disease acne vulgaris. The underlying mechanisms that drive inflammatory gene expression in acne-involved pilosebaceous epithelial cells are still unknown because almost all sebaceous follicles contain dense concentrations of bacteria yet only some show an inflammatory reaction. In this study, we hypothesized that metabolites from the abundant skin-resident microbe Propionibacterium acnes can influence cytokine expression from human sebocytes. We show that short-chain fatty acids produced by P. acnes under environmental conditions that favor fermentation will drive inflammatory gene expression from sebocytes. These molecules are shown to influence sebocyte behavior through two distinct mechanisms: the inhibition of histone deacetylase (HDAC) activity and the activation of fatty acid receptors. Depletion of HDAC8 and HDAC9 in human sebocytes resulted in an enhanced cytokine response to TLR-2 activation that resembled the transcriptional profile of an acne lesion. These data provide a new insight into the regulation of inflammatory gene expression in the skin, further characterize the contribution of sebocytes to epidermal immunity, and demonstrate how changes in the metabolic state of the skin microbiome can promote inflammatory acne.


Subject(s)
Epithelial Cells/metabolism , Fatty Acids, Volatile/metabolism , Gene Expression Regulation/physiology , Propionibacteriaceae/metabolism , Sebaceous Glands/metabolism , Acne Vulgaris/immunology , Acne Vulgaris/metabolism , Cell Line , Epigenesis, Genetic , Epithelial Cells/immunology , Humans , Inflammation/immunology , Inflammation/metabolism , Sebaceous Glands/immunology , Skin/immunology , Skin/metabolism , Skin/microbiology
12.
Cell ; 176(5): 982-997.e16, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30712873

ABSTRACT

Immune cells and epithelium form sophisticated barrier systems in symbiotic relationships with microbiota. Evidence suggests that immune cells can sense microbes through intact barriers, but regulation of microbial commensalism remain largely unexplored. Here, we uncovered spatial compartmentalization of skin-resident innate lymphoid cells (ILCs) and modulation of sebaceous glands by a subset of RORγt+ ILCs residing within hair follicles in close proximity to sebaceous glands. Their persistence in skin required IL-7 and thymic stromal lymphopoietin, and localization was dependent on the chemokine receptor CCR6. ILC subsets expressed TNF receptor ligands, which limited sebocyte growth by repressing Notch signaling pathway. Consequently, loss of ILCs resulted in sebaceous hyperplasia with increased production of antimicrobial lipids and restricted commensalism of Gram-positive bacterial communities. Thus, epithelia-derived signals maintain skin-resident ILCs that regulate microbial commensalism through sebaceous gland-mediated tuning of the barrier surface, highlighting an immune-epithelia circuitry that facilitates host-microbe symbiosis.


Subject(s)
Lymphocytes/immunology , Sebaceous Glands/metabolism , Sebaceous Glands/microbiology , Animals , Bacteria/metabolism , Cytokines/metabolism , Epithelium/immunology , Hair Follicle/metabolism , Hair Follicle/microbiology , Immunity, Innate , Interleukin-7/metabolism , Lymphocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/immunology , Receptors, CCR6/metabolism , Receptors, Notch/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Sebaceous Glands/immunology , Skin/metabolism , Skin Physiological Phenomena , Symbiosis , Thymic Stromal Lymphopoietin
13.
Am J Clin Dermatol ; 20(3): 335-344, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30632097

ABSTRACT

Acne vulgaris is a chronic skin disorder involving hair follicles and sebaceous glands. Multiple factors contribute to the disease, including skin microbes. The skin microbiome in the follicle is composed of a diverse group of microorganisms. Among them, Propionibacterium acnes and Malassezia spp. have been linked to acne development through their influence on sebum secretion, comedone formation, and inflammatory response. Antibiotics targeting P. acnes have been the mainstay in acne treatment for the past four decades. Among them, macrolides, clindamycin, and tetracyclines are the most widely prescribed. As antibiotic resistance becomes an increasing concern in clinical practice, understanding the skin microbiome associated with acne and the effects of antibiotic use on the skin commensals is highly relevant and critical to clinicians. In this review, we summarize recent studies of the composition and dynamics of the skin microbiome in acne and the effects of antibiotic treatment on skin microbes.


Subject(s)
Acne Vulgaris/drug therapy , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Microbiota/drug effects , Acne Vulgaris/immunology , Acne Vulgaris/microbiology , Anti-Bacterial Agents/therapeutic use , Hair Follicle/immunology , Hair Follicle/microbiology , Humans , Malassezia/isolation & purification , Malassezia/physiology , Microbiota/immunology , Propionibacterium acnes/isolation & purification , Propionibacterium acnes/physiology , Sebaceous Glands/immunology , Sebaceous Glands/microbiology , Symbiosis/drug effects , Treatment Outcome
14.
J Innate Immun ; 11(1): 41-51, 2019.
Article in English | MEDLINE | ID: mdl-30056444

ABSTRACT

Activation of Toll-like receptor (TLR)-2 and subsequent inflammatory response contribute to lesion development in acne vulgaris. A cross-talk between aryl hydrocarbon receptor (AhR), a cytosolic receptor protein that responds to environmental and physiological stress, and TLRs has recently been reported. In this study, we explored the possible role of AhR in the effects induced on cultured human SZ95 sebocytes by peptidoglycan (PGN), a classic TLR2 agonist. PGN-induced secretion of inflammatory factors TNF-α and IL-8 in human SZ95 sebocytes was suppressed after knockdown of AhR and pretreatment with the AhR antagonist CH223191. In addition, the AhR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) enhanced TNF-α and IL-8 secretion in PGN-pretreated sebocytes. Furthermore, PGN-induced expression of myeloid differentiation factor 88 (MyD88), phospho-p38MAPK (p-p38MAPK), and p-p65NF-κB was strengthened by TCDD and repressed by CH223191. AhR inhibition by transfecting shRNA blocked the ability of PGN to stimulate phosphorylation of p38MAPK and p65NF-κB in SZ95 sebocytes. Overall, these data demonstrate that AhR is able to modulate PGN-induced expression of TNF-α and IL-8 in human SZ95 sebocytes involving the MyD88-p65NF-κB/p38MAPK signaling pathway, which probably indicates a new mechanism in TLR2-mediated acne.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Interleukin-8/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Tumor Necrosis Factor-alpha/metabolism , Acne Vulgaris/physiopathology , Basic Helix-Loop-Helix Transcription Factors/immunology , Cells, Cultured , Epithelial Cells/immunology , Epithelial Cells/metabolism , Humans , Inflammation , Myeloid Differentiation Factor 88/metabolism , Peptidoglycan/immunology , Peptidoglycan/metabolism , Polychlorinated Dibenzodioxins/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/immunology , Sebaceous Glands/immunology , Sebaceous Glands/metabolism , Signal Transduction , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Sci Transl Med ; 10(451)2018 07 25.
Article in English | MEDLINE | ID: mdl-30045980

ABSTRACT

Meibomian glands (MGs) are sebaceous glands of the eyelid margin that secrete lipids needed to avert tear evaporation and to help maintain ocular surface homeostasis. Obstruction of MGs or other forms of MG dysfunction can promote chronic diseases of the ocular surface. Although chronic eyelid inflammation, such as allergic eye disease, is an associated risk factor for obstructive MG dysfunction, it is not clear whether inflammatory processes contribute to the pathophysiology of MG obstruction. We show that polymorphonuclear neutrophils (PMNs) promoted MG obstruction in a chronic inflammatory model of allergic eye disease in mice. Analysis of leukocytes in tears of patients with MG dysfunction showed an increase in PMN numbers compared to healthy subjects. Moreover, PMN numbers in tears positively correlated with clinical severity of MG dysfunction. Our findings point to a role for PMNs in the pathogenesis and progression of MG dysfunction.


Subject(s)
Eyelid Diseases/immunology , Eyelid Diseases/pathology , Meibomian Glands/immunology , Meibomian Glands/pathology , Sebaceous Glands/immunology , Sebaceous Glands/pathology , Animals , Mice , Mice, Inbred C57BL , Neutrophils/metabolism
16.
Br J Dermatol ; 179(4): 906-917, 2018 10.
Article in English | MEDLINE | ID: mdl-29663317

ABSTRACT

BACKGROUND: Possible outcomes of acne lesions are atrophic scars, which may cause serious psychological distress. Current treatments for postacne scarring often require invasive procedures. Pathophysiological studies on acne scarring have only investigated the first week of papule life. OBJECTIVES: To study the pathophysiology of atrophic scar formation to identify molecular and cellular pathways that can lead to new therapies for the prevention of acne scarring. METHODS: Large-scale gene expression profiling and immunohistochemistry analysis were performed on uninvolved skin and papules in both scar-prone (SP) and non-scar-prone (NSP) patients with acne, at different time points. RESULTS: Gene expression and immunohistochemistry analyses showed a very similar immune response in 48-h-old papules in SP and NSP populations, characterized by elevated numbers of T cells, neutrophils and macrophages. However, the immune response only persisted in SP patients in 3-week-old papules, and was characterized by an important B-cell infiltrate. Transient downmodulation of sebaceous gland markers related to lipid metabolism was observed in 48-h-old papules in NSP patients, followed by normalization after 3 weeks. In contrast, in SP patients a drastic reduction of these markers persisted in 3-week-old papules, suggesting an irreversible destruction of sebaceous gland structures after inflammatory remodelling in SP patients with acne. CONCLUSIONS: Long-lived acne papules are characterized by a B-cell infiltrate. A relationship exists between the duration and severity of inflammation and the alteration of sebaceous gland structures, leading to atrophic scar formation in acne.


Subject(s)
Acne Vulgaris/complications , Cicatrix/immunology , Plasma Cells/immunology , Sebaceous Glands/pathology , Atrophy/etiology , Atrophy/immunology , Biopsy , Cicatrix/etiology , Cicatrix/pathology , Epidermis/immunology , Epidermis/pathology , Gene Expression Profiling , Humans , Sebaceous Glands/cytology , Sebaceous Glands/immunology
17.
Br J Dermatol ; 179(2): 420-430, 2018 08.
Article in English | MEDLINE | ID: mdl-29532463

ABSTRACT

BACKGROUND: Rosacea is a common facial skin disorder mainly affecting middle-aged adults. Its aetiology is unknown and pathogenesis uncertain. Activation of the host innate immune response has been identified as an important factor. The Demodex mite population in the skin of rosacea patients is significantly higher than in patients with normal skin, suggesting that they may be of aetiological importance in this disorder. OBJECTIVES: To determine the potential of Demodex mites to interact with the host immune system. METHODS: Live Demodex mites were extracted from normal facial skin of control subjects and used in cell stimulation experiments with the immortalized SZ95 sebocyte line. Time- and mite-dose-dependent experiments were performed. Direct effects of Demodex and effects of the medium in which Demodex had been cultured were evaluated on the Toll-like receptor (TLR) signalling pathway on both a gene and protein expression level. RESULTS: Mites modulated TLR signalling events on both mRNA and protein levels in SZ95 sebocytes. An initial trend towards downmodulation of genes in this pathway was observed. A subsequent switch to positive gene upregulation was recorded after 48 h of coculture. Demodex secreted bioactive molecules that affected TLR2 receptor expression by sebocytes. High numbers of Demodex induced proinflammatory cytokine secretion, whereas lower numbers did not. CONCLUSIONS: Demodex mites have the capacity to modulate the TLR signalling pathway of an immortalized human sebocyte line. Mites have the capacity to secrete bioactive molecules that affect the immune reactivity of sebocytes. Increasing mite numbers influenced interleukin-8 secretion by these cells.


Subject(s)
Facial Dermatoses/immunology , Mite Infestations/immunology , Mites/immunology , Rosacea/immunology , Sebaceous Glands/immunology , Animals , Biopsy , Cell Line , Cytokines/metabolism , Epithelial Cells/immunology , Epithelial Cells/parasitology , Epithelial Cells/pathology , Facial Dermatoses/parasitology , Facial Dermatoses/pathology , Host-Parasite Interactions/immunology , Humans , Mite Infestations/parasitology , Mite Infestations/pathology , Rosacea/parasitology , Rosacea/pathology , Sebaceous Glands/cytology , Sebaceous Glands/parasitology , Sebaceous Glands/pathology , Signal Transduction/immunology , Toll-Like Receptors/metabolism , Up-Regulation
18.
J Invest Dermatol ; 138(8): 1699-1706, 2018 08.
Article in English | MEDLINE | ID: mdl-29501385

ABSTRACT

We have previously shown that endocannabinoids (eCBs) (e.g., anandamide) are involved in the maintenance of homeostatic sebaceous lipid production in human sebaceous glands and that eCB treatment dramatically increases sebaceous lipid production. Here, we aimed to investigate the expression of the major eCB synthesizing and degrading enzymes and to study the effects of eCB uptake inhibitors on human SZ95 sebocytes, thus exploring the role of the putative eCB membrane transporter, which has been hypothesized to facilitate the cellular uptake and subsequent degradation of eCBs. We found that the major eCB synthesizing (N-acyl phosphatidylethanolamine-specific phospholipase D, and diacylglycerol lipase-α and -ß) and degrading (fatty acid amide hydrolase, monoacylglycerol lipase) enzymes are expressed in SZ95 sebocytes and also in sebaceous glands (except for diacylglycerol lipase-α, the staining of which was dubious in histological preparations). eCB uptake-inhibition with VDM11 induced a moderate increase in sebaceous lipid production and also elevated the levels of various eCBs and related acylethanolamides. Finally, we found that VDM11 was able to interfere with the proinflammatory action of the TLR4 activator lipopolysaccharide. Collectively, our data suggest that inhibition of eCB uptake exerts anti-inflammatory actions and elevates both sebaceous lipid production and eCB levels; thus, these inhibitors might be beneficial in cutaneous inflammatory conditions accompanied by dry skin.


Subject(s)
Arachidonic Acids/pharmacology , Endocannabinoids/metabolism , Epithelial Cells/metabolism , Sebaceous Glands/metabolism , Amidohydrolases/metabolism , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Humans , Lipid Metabolism/drug effects , Lipoprotein Lipase/metabolism , Monoacylglycerol Lipases/metabolism , Phospholipase D/metabolism , Sebaceous Glands/cytology , Sebaceous Glands/drug effects , Sebaceous Glands/immunology
19.
Biochem Pharmacol ; 138: 96-106, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28461124

ABSTRACT

The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) controls the expression of genes involved in the regulation of lipid and glucose metabolism, cell proliferation/differentiation as well as inflammatory pathways. Pivotal studies in human sebocytes and isolated sebaceous glands have raised the interesting possibility that compounds acting on PPARγ can modulate sebaceous lipids and inflammation and, as such, may be useful in the treatment of acne. To investigate the role of this receptor in the regulation of lipid synthesis, proliferation and inflammation, we used the SZ95 sebaceous gland cell line stimulated with insulin. In sebocytes, insulin signaling activated the phosphatidylinositide 3-kinase-Akt (PI3K/Akt) and mammalian target of rapamycin (mTOR) pathways, which, in turn, induced high protein/lipid synthesis, increased cell growth and proliferation as well as inflammation. As regards lipogenesis, insulin initially stimulated the formation of unsaturated lipids and then the neosynthesis of lipids. The results showed, that the modulation of PPARγ, counteracted the insulin-induced altered lipogenesis, evident through a decrease in gene expression of key enzymes responsible for the synthesis of fatty acids, and through a reduction of lipid species synthesis analyzed by Oil/Nile Red staining and GC-MS. PPARγ modulation also regulated the insulin-induced proliferation, inhibiting the cell cycle progression and p21WAF1/CIP1 (p21) protein reduction. Moreover, the expression of inflammatory cytokines, induced by insulin or lipopolysaccharide (LPS), was down-modulated. In PPARγ-deficient cells or in the presence of GW9662 antagonist, all these observed effects were abolished, indicating that PPARγ activation plays a role in regulating alteration of lipogenesis, cell proliferation and inflammatory signaling. We demonstrated that selective modulation of PPARγ activity is likely to represent a therapeutic strategy for the treatment of acne.


Subject(s)
Gene Expression Regulation , Lipogenesis , PPAR gamma/metabolism , Sebaceous Glands/metabolism , Sebum/metabolism , Signal Transduction , Acetanilides/adverse effects , Acetanilides/pharmacology , Anilides/adverse effects , Anilides/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Cycle/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytokines/agonists , Cytokines/metabolism , Dermatologic Agents/adverse effects , Dermatologic Agents/pharmacology , Gene Expression Regulation/drug effects , Humans , Hypoglycemic Agents/antagonists & inhibitors , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Insulin Antagonists/adverse effects , Insulin Antagonists/pharmacology , Lipogenesis/drug effects , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/toxicity , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , Phenylpropionates/adverse effects , Phenylpropionates/pharmacology , RNA Interference , Sebaceous Glands/cytology , Sebaceous Glands/drug effects , Sebaceous Glands/immunology , Sebum/drug effects , Signal Transduction/drug effects
20.
Nagoya J Med Sci ; 79(1): 85-90, 2017 02.
Article in English | MEDLINE | ID: mdl-28303065

ABSTRACT

A 65-year-old man was referred to our hospital for the treatment of a lesion on the medial lacrimal canthus of both eyes. He had a history of perinuclear anti-neutrophil cytoplasmic antibodies, i.e., pANCA-positive interstitial pneumonia. Orbital magnetic resonance imaging excluded space occupying lesions, and laboratory testing excluded thyroid-related diseases. The masses were excised, and histopathological examinations showed sebaceous gland hyperplasia and inflammatory changes around the gland. In addition, the specimen from the left eye showed a retention cyst possibly caused by an infection. It was also possible that the use of steroid was involved in the development of the lesions. A relationship between the ANCA and the lesions was not completely eliminated.


Subject(s)
Antibodies, Antineutrophil Cytoplasmic/immunology , Lacrimal Apparatus Diseases/diagnosis , Lung Diseases, Interstitial/diagnosis , Aged , Humans , Inflammation/immunology , Inflammation/metabolism , Lacrimal Apparatus Diseases/immunology , Lacrimal Apparatus Diseases/metabolism , Lung Diseases, Interstitial/immunology , Lung Diseases, Interstitial/metabolism , Male , Sebaceous Glands/immunology , Sebaceous Glands/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...