1
|
Wei YF, Zhang HL, Li LZ, Lv Y, Li H, Li Z, Yu FL, Jiang T, Zhang TY, Xin F, Ma C, Ren YX. Sirt1 blocks nucleus pulposus and macrophages crosstalk by inhibiting RelA/Lipocalin 2 axis. J Orthop Translat 2025; 50:30-43. [PMID: 39758288 PMCID: PMC11699611 DOI: 10.1016/j.jot.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/26/2024] [Accepted: 11/25/2024] [Indexed: 01/07/2025] Open
Abstract
Background Intervertebral disc degeneration (IVDD) stands as a primary pathophysiological driver of low back pain, yet no therapeutic intervention effectively arrests its progression. Evidence shows that certain Sirt1 agonists may confer protective effects on intervertebral discs, but the underlying mechanisms remain unclear. This study aims to delineate the interaction between Sirt1 and the inflammatory microenvironment, offering potential novel avenues for IVDD prevention and treatment. Methods In vitro IL-1β-induced nucleus pulposus cells (NPCs) degenerative model and in vivo a mouse annulus fibrosus needle puncture model in Sirt1 transgenic (Sirt1TG) and the same litter WT mice were used to investigate the role of Sirt1 in homeostasis and inflammation. Mechanistic insights were obtained through RNA sequencing, co-immunoprecipitation (Co-IP), luciferase assays, and chromatin immunoprecipitation-(ChIP)-PCR. A co-culture system of Raw264.7 and NPCs was employed to assess the involvement of Lipocalin 2. Results Our study demonstrated reduced Sirt1 expression in degenerating human nucleus pulposus (NP) tissue. Both in vitro and in vivo data revealed that NP-specific overexpression of Sirt1 inhibited extracellular matrix degradation and inflammation. Mechanistically, Sirt1 suppressed the acetylation of RelA/p65 at lysine 310 and phosphorylation at serine 536, with the C-terminus of Sirt1 and the RHD-NLS domain of RelA mediating to their interaction. Furthermore, NPCs-derived Lipocalin 2 was identified as a cytokine involved in macrophage chemotaxis and M1 polarization to exacerbate inflammation. Conclusion Our work revealed that Sirt1 negatively regulates Lipocalin 2, thereby ameliorating the inflammatory milieu and blocking NPCs and macrophages crosstalk. The Translational Potential of this Article This study illuminates the crucial role and molecular mechanisms of Sirt1 in regulating the NP microenvironment. These insights shed light on strategies for the prevention and treatment of IVDD-related herniation and low back pain. By pinpointing specific biological targets, the screening of smallmolecule compounds with significant clinical implications can be facilitated. This translational innovation promises to optimize cells communication within intervertebral disc microenvironment via localized drug delivery, potentially improving patient outcomes and satisfaction following spinal fusion or discectomy surgeries.
Collapse
Affiliation(s)
- Yi-Fan Wei
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - He-Long Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - Ling-Zhi Li
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - You Lv
- Department of Orthopaedics, Lianyungang Clinical College of Nanjing Medical University, 6 Zhenhua East Rd, Lianyungang, 221000, China
| | - He Li
- Department of Sports Medicine, Lianyungang Clinical College of Nanjing Medical University, 6 Zhenhua East Rd, Lianyungang, 221000, China
| | - Zhi Li
- Department of Orthopaedics, Geriatric Hospital of Nanjing Medical University, 65 Jiangsu Rd, Nanjing, 210024, China
| | - Feng-Lei Yu
- Department of Trauma and Orthopaedics, The First People's Hospital of Kunshan, 566 East Qianjin Rd, Suzhou, 215000, China
| | - Tao Jiang
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - Tian-You Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - Feng Xin
- Department of Orthopaedics, Xuzhou Cancer Hospital, 131 Huancheng Rd, Xuzhou, 221005, China
| | - Cheng Ma
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - Yong-Xin Ren
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| |
Collapse
|
2
|
Xue Y, Xia Y, Cheng D, Shi T, Mei P, Hong S. Association between genetically proxied PPARG activation and psoriasis vulgaris: a Mendelian randomization study. J DERMATOL TREAT 2024; 35:2381763. [PMID: 39034037 DOI: 10.1080/09546634.2024.2381763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/03/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Psoriasis is a common autoimmune disease in clinical practice, and previous observational studies have suggested that PPARG agonists such as Pioglitazone may be potential therapeutic agents. However, due to interference from various confounding factors, different observational studies have not reached a unified conclusion. We aim to evaluate the potential use of PPARG agonists for treating psoriasis from a new perspective through drug-targeted Mendelian randomization (MR) analysis. MATERIALS AND METHODS This study includes data on 8,876 individuals for acute myocardial infarction from GWAS, and LDL cholesterol data from 343,621 Europeans. FinnGen contributed psoriasis vulgaris data for 403,972 individuals. The DrugBank10 databases function to identify genes encoding protein products targeted by active constituents of lipid-modifying targets. A two-sample MR analysis and summary-data-based MR (SMR) analysis estimated the associations between expressions of drug target genes and symptoms of psoriasis vulgaris. A multivariable MR study was further conducted to examine if the observed association was direct association. RESULTS SMR analysis revealed that enhanced PPARG gene expression in the blood (equivalent to a one standard deviation increase) was a protective factor for psoriasis vulgaris (beta = -0.2017, se = 0.0723, p = 0.0053). Besides, there exists an MR association between LDL mediated by PPARG and psoriasis vulgaris outcomes (beta = -3.9169, se = 0.5676, p = 5.17E-12). These results indicate that PPARG is a therapeutic target for psoriasis, suggesting that psoriasis may be a potential indication for PPARG agonists. CONCLUSION This study confirms that therapeutic activation of PPARG helps suppress the development of psoriasis. Psoriasis may be a new indication for PPARG agonists, such as Pioglitazone. In the future, new anti-psoriatic drugs could be developed targeting PPARG.
Collapse
Affiliation(s)
- Yan Xue
- Department of Dermatology, First Affiliated Hospital of Naval Military Medical University, Shanghai, China
| | - Yuning Xia
- First Clinical Medical College (First Affiliated Hospital), Anhui Medical University, Hefei, China
| | - Donghao Cheng
- First Clinical Medical College (First Affiliated Hospital), Anhui Medical University, Hefei, China
| | - Taiyu Shi
- First Clinical Medical College (First Affiliated Hospital), Anhui Medical University, Hefei, China
| | - Ping Mei
- Department of Radiology, Anqing Municipal Hospital, Anqing, China
| | - Sheng Hong
- Department of Dermatology, First Affiliated Hospital of Naval Military Medical University, Shanghai, China
| |
Collapse
|
3
|
Alhelf M, Rashed L, Doss RW, Mohamed SM, Abd Elazeem NA. Long noncoding RNA (taurine upregulated gene 1) and micro RNA-377: emerging players in the development of metabolic syndrome among psoriasis patients. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2023; 12:68. [DOI: 10.1186/s43088-023-00404-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/29/2023] [Indexed: 09/02/2023] Open
Abstract
Abstract
Background
Psoriasis (PsO) is an immune-mediated dermatosis and systemic inflammatory condition that can affect the skin, joints, and other organs and tissues with a range of comorbidities. The activation of proinflammatory cytokines is the primary cause of the development of skin lesions in PsO. Patients with PsO have a higher risk of developing cardiovascular metabolic comorbidities; among these is the metabolic syndrome (MetS). Particularly, MetS is characterized by abdominal obesity, hypertension, hyperglycemia, and hyperlipidemia, has been linked to PsO. The connection between PsO and MetS is believed to be caused by PsO generating systemic inflammation, which then results in elevated inflammatory adipokines, endothelial dysfunction, and insulin resistance. Micro RNA-377 and long noncoding RNA taurine upregulated 1 (TUG1) are both involved in the control of a variety of inflammatory disorders in humans and can be employed as biomarkers for the diagnosis and prognosis of psoriasis. The aim of the present study is to establish a panel of biomarkers for the early diagnosis of MetS incidence in psoriasis and thereby, reducing its lethal consequences.
Results
In this study, 120 patients: 40 psoriatic patients, 40 psoriatic patients with metabolic syndrome, and 40 healthy subjects were conducted. Expressions of Long noncoding RNA Taurine Upregulated Gene-1 (TUG1), miRNA-377 and Peroxisome Proliferator-Activated Receptor-γ (PPAR-γ) were assessed in tissue lesion by real-time PCR. ELISA technique was carried out for the detection of serum levels of plasminogen activator inhibitor-1 (PAI-1) and transforming growth factor β (TGFβ). Moreover, miRNA-377 expression was significantly elevated with the simultaneous down-regulation of both TUG-1 and PPAR-γ in PsO-MetS group when compared to those of PsO and control groups. Furthermore, PAI-1 and TGFβ levels were higher in PsO-MetS than PsO.
Conclusions
The dysregulated levels of TUG-1, miRNA-377, PPAR-γ, PAI-1, and TGFβ, biomarkers may provide information about their potential role in the emergence of MetS in psoriasis patients.
Collapse
|
4
|
Saika A, Tiwari P, Nagatake T, Node E, Hosomi K, Honda T, Kabashima K, Kunisawa J. Mead acid inhibits retinol-induced irritant contact dermatitis via peroxisome proliferator-activated receptor alpha. Front Mol Biosci 2023; 10:1097955. [PMID: 36825199 PMCID: PMC9941550 DOI: 10.3389/fmolb.2023.1097955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
Retinol is widely used in topical skincare products to ameliorate skin aging and treat acne and wrinkles; however, retinol and its derivatives occasionally have adverse side effects, including the induction of irritant contact dermatitis. Previously, we reported that mead acid (5,8,11-eicosatrienoic acid), an oleic acid metabolite, ameliorated skin inflammation in dinitrofluorobenzene-induced allergic contact hypersensitivity by inhibiting neutrophil infiltration and leukotriene B4 production by neutrophils. Here, we showed that mead acid also suppresses retinol-induced irritant contact dermatitis. In a murine model, we revealed that mead acid inhibited keratinocyte abnormalities such as keratinocyte hyperproliferation. Consistently, mead acid inhibited p38 MAPK (mitogen-activated protein kinase) phosphorylation, which is an essential signaling pathway in the keratinocyte hyperplasia induced by retinol. These inhibitory effects of mead acid were associated with the prevention of both keratinocyte hyperproliferation and the gene expression of neutrophil chemoattractants, including Cxcl1 and Cxcl2, and they were mediated by a PPAR (peroxisome proliferator-activated receptor)-α pathway. Our findings identified the anti-inflammatory effects of mead acid, the use of which can be expected to minimize the risk of adverse side effects associated with topical retinoid application.
Collapse
Affiliation(s)
- Azusa Saika
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan
| | - Prabha Tiwari
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan,Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Takahiro Nagatake
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan,Laboratory of Functional Anatomy, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| | - Eri Node
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan
| | - Tetsuya Honda
- Department of Dermatology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Kenji Kabashima
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan,International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Minato, Tokyo, Japan,Graduate School of Medicine, Graduate School of Dentistry, Graduate School of Pharmaceutical Sciences, Graduate School of Science, Osaka University, Suita, Osaka, Japan,Department of Microbiology and Immunology, Graduate School of Medicine, Kobe University, Kobe, Hyogo, Japan,Research Organization for Nano and Life Innovation, Waseda University, Shinjuku, Tokyo, Japan,Graduate School of Biomedical and Health Sciences, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan,*Correspondence: Jun Kunisawa,
| |
Collapse
|
5
|
Ramer R, Hinz B. Cannabinoid Compounds as a Pharmacotherapeutic Option for the Treatment of Non-Cancer Skin Diseases. Cells 2022; 11:4102. [PMID: 36552866 PMCID: PMC9777118 DOI: 10.3390/cells11244102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 12/23/2022] Open
Abstract
The endocannabinoid system has been shown to be involved in various skin functions, such as melanogenesis and the maintenance of redox balance in skin cells exposed to UV radiation, as well as barrier functions, sebaceous gland activity, wound healing and the skin's immune response. In addition to the potential use of cannabinoids in the treatment and prevention of skin cancer, cannabinoid compounds and derivatives are of interest as potential systemic and topical applications for the treatment of various inflammatory, fibrotic and pruritic skin conditions. In this context, cannabinoid compounds have been successfully tested as a therapeutic option for the treatment of androgenetic alopecia, atopic and seborrhoeic dermatitis, dermatomyositis, asteatotic and atopic eczema, uraemic pruritis, scalp psoriasis, systemic sclerosis and venous leg ulcers. This review provides an insight into the current literature on cannabinoid compounds as potential medicines for the treatment of skin diseases.
Collapse
Affiliation(s)
| | - Burkhard Hinz
- Institute of Pharmacology and Toxicology, Rostock University Medical Centre, Schillingallee 70, D-18057 Rostock, Germany
| |
Collapse
|
6
|
Lin X, Meng X, Song Z, Lin J. Peroxisome proliferator-activator receptor γ and psoriasis, molecular and cellular biochemistry. Mol Cell Biochem 2022; 477:1905-1920. [PMID: 35348980 DOI: 10.1007/s11010-022-04417-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 03/16/2022] [Indexed: 10/18/2022]
Abstract
The pathophysiology of psoriasis is complex and has not been completely elucidated. Better understanding of the pathogenesis may contribute to further improvement of our therapeutic strategies controlling psoriasis. Emerging evidence points to a causative relationship between altered activity of peroxisome proliferator-activated receptor γ (PPARγ) and psoriasis. The present review focuses on deeper understanding of the possible role of PPARγ in the pathogenesis of psoriasis and the potential of PPARγ agonist to improve the treatment of psoriasis. PPARγ is decreased in psoriasis. PPARγ possibly has effects on the multiple aspects of the pathogenesis of psoriasis, including abnormal lipid metabolism, insulin resistance, immune cells, pro-inflammatory cytokines, keratinocytes, angiogenesis, oxidative stress, microRNAs and nuclear factor kappa B. As defective activation of PPARγ is involved in psoriasis development, PPARγ agonists may be promising agents for treatment of psoriasis. Pioglitazone appears an effective and safe option in the treatment of patients with psoriasis, but there are still concerns about its potential side effects. Research effort has recently been undertaken to explore the PPARγ-activating potential of natural products. Among them some have been studied clinically or preclinically for treatment of psoriasis with promising results.
Collapse
Affiliation(s)
- Xiran Lin
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Lu, Dalian, 116011, China.
| | - Xianmin Meng
- Department of Pathology and Laboratory Medicine, Axia Women's Health, 450 Cresson BLVD, Oaks, PA, 19456, USA
| | - Zhiqi Song
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Lu, Dalian, 116011, China
| | - Jingrong Lin
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Lu, Dalian, 116011, China
| |
Collapse
|
7
|
Chang CJ, Zhang J, Tsai YL, Chen CB, Lu CW, Huo YP, Liou HM, Ji C, Chung WH. Compositional Features of Distinct Microbiota Base on Serum Extracellular Vesicle Metagenomics Analysis in Moderate to Severe Psoriasis Patients. Cells 2021; 10:2349. [PMID: 34571998 PMCID: PMC8467001 DOI: 10.3390/cells10092349] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 01/14/2023] Open
Abstract
The bacterial microbiota in the skin and intestine of patients with psoriasis were different compared with that of healthy individuals. However, the presence of a distinct blood microbiome in patients with psoriasis is yet to be investigated. In this study, we investigated the differences in bacterial communities in plasma-derived extracellular vesicles (EVs) between patients with moderate to severe psoriasis (PSOs) and healthy controls (HCs). The plasma EVs from the PSO (PASI > 10) (n = 20) and HC (n = 8) groups were obtained via a series of centrifugations, and patterns were examined and confirmed using transmission electron microscopy (TEM) and EV-specific markers. The taxonomic composition of the microbiota was determined by using full-length 16S ribosomal RNA gene sequencing. The PSO group had lower bacterial diversity and richness compared with HC group. Principal coordinate analysis (PCoA)-based clustering was used to assess diversity and validated dysbiosis for both groups. Differences at the level of amplicon sequence variant (ASV) were observed, suggesting alterations in specific ASVs according to health conditions. The HC group had higher levels of the phylum Firmicutes and Fusobacteria than in the PSO group. The order Lactobacillales, family Brucellaceae, genera Streptococcus, and species Kingella oralis and Aquabacterium parvum were highly abundant in the HC group compared with the PSO group. Conversely, the order Bacillales and the genera Staphylococcus and Sphihgomonas, as well as Ralstonia insidiosa, were more abundant in the PSO group. We further predicted the microbiota functional capacities, which revealed significant differences between the PSO and HC groups. In addition to previous studies on microbiome changes in the skin and gut, we demonstrated compositional differences in the microbe-derived EVs in the plasma of PSO patients. Plasma EVs could be an indicator for assessing the composition of the microbiome of PSO patients.
Collapse
Affiliation(s)
- Chih-Jung Chang
- Medical Research Center and Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen 361028, China;
- Drug Hypersensitivity Clinical and Research Center, Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Taoyuan 333423, Taiwan; (C.-B.C.); (C.-W.L.)
| | - Jing Zhang
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China;
| | - Yu-Ling Tsai
- Department of Pathology, Tri-Service General Hospital, Taipei 114202, Taiwan;
| | - Chun-Bing Chen
- Drug Hypersensitivity Clinical and Research Center, Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Taoyuan 333423, Taiwan; (C.-B.C.); (C.-W.L.)
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Department of Medical Research, Chang Gung Memorial Hospital, Linkou, Taoyuan 333423, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung 20445, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan 333323, Taiwan
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen 361028, China; (Y.-P.H.); (H.-M.L.)
| | - Chun-Wei Lu
- Drug Hypersensitivity Clinical and Research Center, Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Taoyuan 333423, Taiwan; (C.-B.C.); (C.-W.L.)
- College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan 333323, Taiwan
| | - Yu-Ping Huo
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen 361028, China; (Y.-P.H.); (H.-M.L.)
| | - Huey-Ming Liou
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen 361028, China; (Y.-P.H.); (H.-M.L.)
| | - Chao Ji
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China;
| | - Wen-Hung Chung
- Medical Research Center and Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen 361028, China;
- Drug Hypersensitivity Clinical and Research Center, Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Taoyuan 333423, Taiwan; (C.-B.C.); (C.-W.L.)
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Department of Medical Research, Chang Gung Memorial Hospital, Linkou, Taoyuan 333423, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung 20445, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan 333323, Taiwan
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen 361028, China; (Y.-P.H.); (H.-M.L.)
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
8
|
Blunder S, Krimbacher T, Moosbrugger‐Martinz V, Gruber R, Schmuth M, Dubrac S. Keratinocyte-derived IL-1β induces PPARG downregulation and PPARD upregulation in human reconstructed epidermis following barrier impairment. Exp Dermatol 2021; 30:1298-1308. [PMID: 33683743 PMCID: PMC8451818 DOI: 10.1111/exd.14323] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 02/15/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are a family of nuclear hormone receptors. In skin, PPARs modulate inflammation, lipid synthesis, keratinocyte differentiation and proliferation and thus are important for skin barrier homeostasis. Accordingly, PPAR expression is altered in various skin conditions that entail epidermal barrier impairment, that is atopic dermatitis (AD) and psoriasis. Using human epidermal equivalents (HEEs), we established models of acute epidermal barrier impairment devoid of immune cells. We assessed PPAR and cytokine expression after barrier perturbation and examined effects of keratinocyte-derived cytokines on PPAR expression. We show that acetone or SDS treatment causes graded impairment of epidermal barrier function. Furthermore, we demonstrate that besides IL-1β and TNFα, IL-33 and TSLP are highly relevant markers for acute epidermal barrier impairment. Both SDS- and acetone-mediated epidermal barrier impairment reduce PPARG expression levels, whereas only SDS enhances PPARD expression. In line with findings in IL-1β and TNFα-treated HEEs, abrogation of IL-1 signalling restores PPARG expression and limits the increase of PPARD expression in SDS-induced epidermal barrier impairment. Thus, following epidermal barrier perturbation, keratinocyte-derived IL-1β and partly TNFα modulate PPARG and PPARD expression. These results emphasize a role for PPARγ and PPARβ/δ in acute epidermal barrier impairment with possible implications for diseases such as AD and psoriasis.
Collapse
Affiliation(s)
- Stefan Blunder
- Department of Dermatology, Venereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Thomas Krimbacher
- Department of Dermatology, Venereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| | | | - Robert Gruber
- Department of Dermatology, Venereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Matthias Schmuth
- Department of Dermatology, Venereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| |
Collapse
|
9
|
Morin S, Simard M, Flamand N, Pouliot R. Biological action of docosahexaenoic acid in a 3D tissue-engineered psoriatic skin model: Focus on the PPAR signaling pathway. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159032. [PMID: 34428549 DOI: 10.1016/j.bbalip.2021.159032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/02/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022]
Abstract
N-3 polyunsaturated fatty acids (n-3 PUFAs), and in particular docosahexaenoic acid (DHA), have many beneficial metabolic effects, including reducing epidermal thickness in patients with psoriasis. The positive impacts of DHA in psoriasis could be mediated by its interactions with the PPAR signaling pathway, as well as by its secretion of anti-inflammatory bioactive metabolites, but the detailed metabolism is still not understood. In the present study, we evaluated the influence of DHA on the main features of psoriasis and its effects on the PPAR signaling pathway, in a psoriatic in vitro skin model. Healthy and psoriatic skin substitutes were produced according to the tissue-engineered self-assembly method, using culture media supplemented with 10 μM of DHA. The presence of DHA led to a reduction in the abnormal cell differentiation of psoriatic keratinocytes, seen in the increased expression of filaggrin and keratin 10. DHA was incorporated into the membrane phospholipids of the epidermis and transformed principally into eicosapentaenoic acid (EPA). Furthermore, the addition of DHA into the culture medium led to a decrease in the levels of lipid mediators derived from n-6 PUFAs, mainly prostaglandin E2 (PGE2) and 12-hydroxyeicosatetraenoic acid (12-HETE). Finally, DHA supplementation rebalanced the expression of PPAR receptors and caused a decrease in the secretion of TNF-α. Altogether, our results show that DHA possesses the ability to attenuate the psoriatic characteristics of psoriatic skin substitutes, mostly by restoring epidermal cell differentiation and proliferation, as well as by reducing inflammation.
Collapse
Affiliation(s)
- Sophie Morin
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Axe médecine régénératrice, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1J 1Z4, Canada; Faculté de pharmacie de l'Université Laval, Québec, QC G1J 1A4, Canada.
| | - Mélissa Simard
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Axe médecine régénératrice, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1J 1Z4, Canada; Faculté de pharmacie de l'Université Laval, Québec, QC G1J 1A4, Canada.
| | - Nicolas Flamand
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, QC G1V 4G5, Canada; Département de médecine, Faculté de médecine de l'Université Laval, Québec, QC G1V 0A6, Canada.
| | - Roxane Pouliot
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Axe médecine régénératrice, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1J 1Z4, Canada; Faculté de pharmacie de l'Université Laval, Québec, QC G1J 1A4, Canada.
| |
Collapse
|
10
|
Blunder S, Pavel P, Minzaghi D, Dubrac S. PPARdelta in Affected Atopic Dermatitis and Psoriasis: A Possible Role in Metabolic Reprograming. Int J Mol Sci 2021; 22:7354. [PMID: 34298981 PMCID: PMC8303290 DOI: 10.3390/ijms22147354] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 12/16/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear hormone receptors expressed in the skin. Three PPAR isotypes, α (NRC1C1), β or δ (NRC1C2) and γ (NRC1C3), have been identified. After activation through ligand binding, PPARs heterodimerize with the 9-cis-retinoic acid receptor (RXR), another nuclear hormone receptor, to bind to specific PPAR-responsive elements in regulatory regions of target genes mainly involved in organogenesis, cell proliferation, cell differentiation, inflammation and metabolism of lipids or carbohydrates. Endogenous PPAR ligands are fatty acids and fatty acid metabolites. In past years, much emphasis has been given to PPARα and γ in skin diseases. PPARβ/δ is the least studied PPAR family member in the skin despite its key role in several important pathways regulating inflammation, keratinocyte proliferation and differentiation, metabolism and the oxidative stress response. This review focuses on the role of PPARβ/δ in keratinocytes and its involvement in psoriasis and atopic dermatitis. Moreover, the relevance of targeting PPARβ/δ to alleviate skin inflammation is discussed.
Collapse
Affiliation(s)
| | | | | | - Sandrine Dubrac
- Epidermal Biology Laboratory, Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (S.B.); (P.P.); (D.M.)
| |
Collapse
|
11
|
Madden SK, Flanagan KL, Jones G. How lifestyle factors and their associated pathogenetic mechanisms impact psoriasis. Clin Nutr 2019; 39:1026-1040. [PMID: 31155371 DOI: 10.1016/j.clnu.2019.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 04/11/2019] [Accepted: 05/06/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUNDS AND AIMS Psoriasis is a skin disorder affecting approximately 2-3% of the global population. While research has revealed a strong genetic component, there are few studies exploring the extent to which lifestyle factors influence psoriasis pathogenesis. The aim of this review was to describe the role of lifestyle factors as both a potential cause and treatment for psoriasis. The review also examines the underlying mechanisms through which these lifestyle factors may operate. METHODS This narrative review aims to incorporate current knowledge relating to both lifestyle and pathogenetic factors that contribute to and alleviate psoriasis presentation. Studies reporting the effect of an inflammatory diet and potential dietary benefits are reported, as well as insights into the effects of stress, smoking and alcohol, insulin resistance and exercise. RESULTS Poor nutrition and low Omega 3 fatty acid intake, likely combined with fat malabsorption caused by gut dysbiosis and systemic inflammation, are associated with psoriasis. The data strongly suggest that improvements to disease severity can be made through dietary and lifestyle interventions and increased physical activity. Less conclusive, although worthy of mention, is the beneficial effect of bile acid supplementation. CONCLUSIONS Lifestyle interventions are a promising treatment for psoriasis and its associated co-morbidities. However, gaps and inadequacies exist within the literature, e.g. methodology, absence of a unified scoring system, lack of controlled clinical data and lack of studies without simultaneous usage of biologics or alternative therapies. Future directions should focus on high quality cohort studies and clinical trials.
Collapse
Affiliation(s)
- Seonad K Madden
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Australia.
| | - Katie L Flanagan
- School of Medicine, College of Health and Medicine, University of Tasmania, Australia; School of Health & Biomedical Science, Royal Melbourne Institute of Technology, Australia; Infectious Diseases Service, Launceston General Hospital, Australia; Dept. of Immunology and Pathology, Monash University, Australia
| | - Graeme Jones
- Menzies Institute for Medical Research, University of Tasmania, Australia
| |
Collapse
|
12
|
Chimenti MS, Sunzini F, Fiorucci L, Botti E, Fonti GL, Conigliaro P, Triggianese P, Costa L, Caso F, Giunta A, Esposito M, Bianchi L, Santucci R, Perricone R. Potential Role of Cytochrome c and Tryptase in Psoriasis and Psoriatic Arthritis Pathogenesis: Focus on Resistance to Apoptosis and Oxidative Stress. Front Immunol 2018; 9:2363. [PMID: 30429845 PMCID: PMC6220124 DOI: 10.3389/fimmu.2018.02363] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 09/24/2018] [Indexed: 02/06/2023] Open
Abstract
Psoriasis (PsO) is an autoimmune disease characterized by keratinocyte proliferation, chronic inflammation and mast cell activation. Up to 42% of patients with PsO may present psoriatic arthritis (PsA). PsO and PsA share common pathophysiological mechanisms: keratinocytes and fibroblast-like synoviocytes are resistant to apoptosis: this is one of the mechanism facilitating their hyperplasic growth, and at joint level, the destruction of articular cartilage, and bone erosion and/or proliferation. Several clinical studies regarding diseases characterized by impairment of cell death, either due to apoptosis or necrosis, reported cytochrome c release from the mitochondria into the extracellular space and finally into the circulation. The presence of elevated cytochrome c levels in serum has been demonstrated in diseases as inflammatory arthritis, myocardial infarction and stroke, and liver diseases. Cytochrome c is a signaling molecule essential for apoptotic cell death released from mitochondria to the cytosol allowing the interaction with protease, as the apoptosis protease activation factor, which lead to the activation of factor-1 and procaspase 9. It has been demonstrated that this efflux from the mitochondria is crucial to start the intracellular signaling responsible for apoptosis, then to the activation of the inflammatory process. Another inflammatory marker, the tryptase, a trypsin-like serine protease produced by mast cells, is released during inflammation, leading to the activation of several immune cells through proteinase-activated receptor-2. In this review, we aimed at discussing the role played by cytochrome c and tryptase in PsO and PsA pathogenesis. To this purpose, we searched pathogenetic mechanisms in PUBMED database and review on oxidative stress, cytochrome c and tryptase and their potential role during inflammation in PsO and PsA. To this regard, the cytochrome c release into the extracellular space and tryptase may have a role in skin and joint inflammation.
Collapse
Affiliation(s)
- Maria Sole Chimenti
- Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| | - Flavia Sunzini
- Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| | - Laura Fiorucci
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Giulia Lavinia Fonti
- Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| | - Paola Conigliaro
- Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| | - Paola Triggianese
- Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| | - Luisa Costa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | | | - Maria Esposito
- Dermatology, University of Rome Tor Vergata, Rome, Italy
| | - Luca Bianchi
- Dermatology, University of Rome Tor Vergata, Rome, Italy
| | - Roberto Santucci
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Roberto Perricone
- Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
13
|
Miragliotta V, Ricci PL, Albanese F, Pirone A, Tognotti D, Abramo F. Cannabinoid receptor types 1 and 2 and peroxisome proliferator-activated receptor-α: distribution in the skin of clinically healthy cats and cats with hypersensitivity dermatitis. Vet Dermatol 2018; 29:316. [PMID: 29920828 DOI: 10.1111/vde.12658] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2018] [Indexed: 01/17/2023]
Abstract
BACKGROUND Cannabinoid receptors and peroxisome proliferator-activated receptor-alpha (PPAR-α) are gaining recognition as potential therapeutic targets for the treatment of skin disorders. HYPOTHESIS/OBJECTIVES The aim of this study was to investigate the distribution of cannabinoid type 1 and 2 receptors (CBR1 and CBR2) and PPAR-α in feline skin and verify whether changes occur in the course of hypersensitivity dermatitis. ANIMALS Twelve privately owned cats. Skin samples were obtained from five healthy cats with no skin lesions and seven cats clinically diagnosed with hypersensitivity dermatitis. METHODS AND MATERIALS Haematoxylin and eosin stained skin sections were investigated for histopathological changes. Indirect immunofluorescence for CBR1, CBR2 and PPAR-α was performed on paraffin-embedded sections, and antibody specificity tested by Western blot analysis. RESULTS Skin samples from cats with hypersensitivity dermatitis were all histopathologically diagnosed with eosinophilic dermatitis. CB receptors and PPAR-α were distributed throughout the skin in both healthy and allergic cats. In normal feline skin, these receptors were mainly distributed in the epithelial compartment. Receptor expression increased in hypersensitivity compared to healthy skin, with the main distribution changes being suprabasal for CBR1, dermal for CBR2 and marked expression of PPAR-α in hyperplastic epidermis and perivascular infiltrate. CONCLUSIONS AND CLINICAL IMPORTANCE Increased expression of cannabinoid receptors in the skin of cats with hypersensitivity dermatitis suggests an endogenous protective strategy and may support the use of natural cannabinoid receptor or PPAR-α agonists to treat feline hypersensitivity dermatitis.
Collapse
Affiliation(s)
- Vincenzo Miragliotta
- Department of Veterinary Sciences, University of Pisa, Viale delle Piagge 2, I-56124, Pisa, Italy
| | - Pier Luca Ricci
- Department of Veterinary Sciences, University of Pisa, Viale delle Piagge 2, I-56124, Pisa, Italy
| | - Francesco Albanese
- Private Veterinary Laboratory "LaVallonea", Via Giuseppe Sirtori, 9, 20017, Passirana di Rho, Italy
| | - Andrea Pirone
- Department of Veterinary Sciences, University of Pisa, Viale delle Piagge 2, I-56124, Pisa, Italy
| | - Danika Tognotti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Francesca Abramo
- Department of Veterinary Sciences, University of Pisa, Viale delle Piagge 2, I-56124, Pisa, Italy
| |
Collapse
|
14
|
Jørgensen E, Lazzarini G, Pirone A, Jacobsen S, Miragliotta V. Normal microscopic anatomy of equine body and limb skin: A morphological and immunohistochemical study. Ann Anat 2018; 218:205-212. [PMID: 29730469 DOI: 10.1016/j.aanat.2018.03.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 03/12/2018] [Accepted: 03/29/2018] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Information on microscopic anatomy of equine skin is sparse. In horses, limb wounds often become chronic and/or non-healing whereas body wounds heal normally. These dissimilarities in healing patterns might be a product of different phenotypic characteristics of body and limb skin. The objective of this study was to investigate microscopic anatomy, epidermal thickness, keratinocyte proliferation and differentiation as well as the presence of mast cells in normal equine skin of body and limb. MATERIALS AND METHODS The study involved body and limb skin biopsies from six horses. Histological characteristics of the epidermis were assessed and epithelial thickness measured. Immunohistochemistry was performed to investigate epidermal differentiation patterns of cytokeratin (CK) 10, CK14, CK16, loricrin, and peroxisome proliferator-activated receptor alpha (PPAR-α), epidermal proliferation (Ki-67 immunostaining), and mast cells distribution in the skin. RESULTS The epidermis was significantly thicker in the limb skin compared to body skin (p<0.01). Epidermal proliferation and CK distribution did not show differences in the two anatomical areas. Loricrin presence was focally found in the spinous layer in four out of six limb skin samples but not in body skin samples. Tryptase positive mast cells were detected in the dermis and their density (cell/mm2) was not different between body and limb. DISCUSSION AND CONCLUSION Here we report for the first time about the normal distribution of CK10, CK14, CK16, PPAR-α, and loricrin in equine limb and body skin as well as about epidermal proliferation rate and mast cell count. It will be relevant to investigate the distribution of the investigated epithelial differentiation markers and the role of mast cells during equine wound healing and/or other skin diseases.
Collapse
Affiliation(s)
- Elin Jørgensen
- Department of Veterinary Clinical Sciences, University of Copenhagen, Højbakkegaard Alle 5, DK-2630 Taastrup, Denmark
| | - Giulia Lazzarini
- Department of Veterinary Sciences, University of Pisa, viale delle Piagge 2, 56124 Pisa, Italy
| | - Andrea Pirone
- Department of Veterinary Sciences, University of Pisa, viale delle Piagge 2, 56124 Pisa, Italy
| | - Stine Jacobsen
- Department of Veterinary Clinical Sciences, University of Copenhagen, Højbakkegaard Alle 5, DK-2630 Taastrup, Denmark
| | - Vincenzo Miragliotta
- Department of Veterinary Sciences, University of Pisa, viale delle Piagge 2, 56124 Pisa, Italy.
| |
Collapse
|
15
|
Tohyama M, Shirakata Y, Hanakawa Y, Dai X, Shiraishi K, Murakami M, Miyawaki S, Mori H, Utsunomiya R, Masuda K, Hashimoto K, Sayama K. Bcl-3 induced by IL-22 via STAT3 activation acts as a potentiator of psoriasis-related gene expression in epidermal keratinocytes. Eur J Immunol 2018; 48:168-179. [PMID: 28901004 DOI: 10.1002/eji.201747017] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 07/26/2017] [Accepted: 09/08/2017] [Indexed: 12/25/2022]
Abstract
IL-22 induces STAT3 phosphorylation and mediates psoriasis-related gene expression. However, the signaling mechanism leading from pSTAT3 to the expression of these genes remains unclear. We focused on Bcl-3, which is induced by STAT3 activation and mediates gene expression. In cultured human epidermal keratinocytes, IL-22 increased Bcl-3, which was translocated to the nucleus with p50 via STAT3 activation. The increases in CXCL8, S100As and human β-defensin 2 mRNA expression caused by IL-22 were abolished by siRNA against Bcl-3. Although CCL20 expression was also augmented by IL-22, the knockdown of Bcl-3 increased its level. Moreover, the combination of IL-22 and IL-17A enhanced Bcl-3 production, IL-22-induced gene expression, and the expression of other psoriasis-related genes, including those encoding IL-17C, IL-19, and IL-36γ. The expression of these genes (except for CCL20) was also suppressed by the knockdown of Bcl-3. Bcl-3 overexpression induced CXCL8 and HBD2 expression but not S100As expression. We also compared Bcl-3 expression between psoriatic skin lesions and normal skin. Immunostaining revealed strong signals for Bcl-3 and p50 in the nucleus of epidermal keratinocytes from psoriatic skin. The IL-22-STAT3-Bcl-3 pathway may be important in the pathogenesis of psoriasis.
Collapse
Affiliation(s)
- Mikiko Tohyama
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Yuji Shirakata
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Yasushi Hanakawa
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Xiuju Dai
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ken Shiraishi
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Masamoto Murakami
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Saori Miyawaki
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Hideki Mori
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ryo Utsunomiya
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Kana Masuda
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Koji Hashimoto
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Koji Sayama
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| |
Collapse
|
16
|
Müller R. PPARβ/δ in human cancer. Biochimie 2016; 136:90-99. [PMID: 27916645 DOI: 10.1016/j.biochi.2016.10.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 10/06/2016] [Accepted: 10/19/2016] [Indexed: 12/31/2022]
Abstract
The nuclear receptor factor peroxisome proliferator-activated receptor (PPARβ/δ) can regulate its target genes by transcriptional activation or repression through both ligand-dependent and independent mechanism as well as by interactions with other transcription factors. PPARβ/δ exerts essential regulatory functions in intermediary metabolism that have been elucidated in detail, but clearly also plays a role in inflammation, differentiation, apoptosis and other cancer-associated processes, which is, however, mechanistically only partly understood. Consistent with these functions clinical associations link the expression of PPARβ/δ and its target genes to an unfavorable outcome of several human cancers. However, the available data do not yield a clear picture of PPARβ/δ's role in cancer-associated processes and are in fact partly controversial. This article provides an overview of this research area and discusses the role of PPARβ/δ in cancer in light of the complex mechanisms of its transcriptional regulation and its potential as a druggable anti-cancer target.
Collapse
Affiliation(s)
- Rolf Müller
- Institute of Molecular Biology and Tumor Research, Center for Tumor Biology and Immunology, Philipps University, Hans-Meerwein-Str. 3, 35043 Marburg, Germany.
| |
Collapse
|
17
|
Abstract
Progenitor/stem cell populations of epithelium are known to reside in the small-sized cell population. Our objective was to physically isolate and characterize an oral keratinocyte-enriched population of small-sized progenitor/stem cells. Primary human oral mucosal keratinocytes cultured in a chemically defined serum-free culture system, devoid of animal-derived feeder cells, were sorted by relative cell size and characterized by immunolabeling for β1 integrin, nuclear transcription factor, peroxisome proliferator-activated receptor-gamma, and cell-cycle analysis. Sorted cells were distinguished as progenitor/stem cells by functional assays and their ability to regenerate an oral mucosal graft. Small-sized cells demonstrated the lowest expression of peroxisome proliferator-activated receptor-gamma, the highest colony-forming efficiency, a longer long-term proliferative potential, an enriched quiescent cell population, and the ability to regenerate an oral mucosal graft, implying that the small-sized cultured oral keratinocytes contained an enriched population of progenitor/stem cells.
Collapse
Affiliation(s)
- K Izumi
- Department of Oral and Maxillofacial Surgery, University of Michigan Health System, B1-208 TC, Box 0018, 1500 East Medical Center Drive, Ann Arbor, MI 48109-0018, USA
| | | | | |
Collapse
|
18
|
Yin K, Smith AG. Nuclear receptor function in skin health and disease: therapeutic opportunities in the orphan and adopted receptor classes. Cell Mol Life Sci 2016; 73:3789-800. [PMID: 27544210 PMCID: PMC11108460 DOI: 10.1007/s00018-016-2329-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 08/04/2016] [Indexed: 12/12/2022]
Abstract
The skin forms a vital barrier between an organism's external environment, providing protection from pathogens and numerous physical and chemical threats. Moreover, the intact barrier is essential to prevent water and electrolyte loss without which terrestrial life could not be maintained. Accordingly, acute disruption of the skin through physical or chemical trauma needs to be repaired timely and efficiently as sustained skin pathologies ranging from mild irritations and inflammation through to malignancy impact considerably on morbidity and mortality. The Nuclear Hormone Receptor Family of transcriptional regulators has proven to be highly valuable targets for addressing a range of pathologies, including metabolic syndrome and cancer. Indeed members of the classic endocrine sub-group, such as the glucocorticoid, retinoid, and Vitamin D receptors, represent mainstay treatment strategies for numerous inflammatory skin disorders, though side effects from prolonged use are common. Emerging evidence has now highlighted important functional roles for nuclear receptors belonging to the adopted and orphan subgroups in skin physiology and patho-physiology. This review will focus on these subgroups and explore the current evidence that suggests these nuclear receptor hold great promise as future stand-alone or complementary drug targets in treating common skin diseases and maintaining skin homeostasis.
Collapse
Affiliation(s)
- Kelvin Yin
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Aaron G Smith
- Dermatology Research Centre, School of Medicine, University of Queensland, Brisbane, QLD, 4072, Australia.
- School of Biomedical Science, Institute of Health and Biomedical Innovation at the Translational Research Institute, Queensland University of Technology, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
19
|
Wang X, Hao Y, Wang X, Wang L, Chen Y, Sun J, Hu J. A PPARδ-selective antagonist ameliorates IMQ-induced psoriasis-like inflammation in mice. Int Immunopharmacol 2016; 40:73-78. [PMID: 27584056 DOI: 10.1016/j.intimp.2016.08.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 08/22/2016] [Accepted: 08/24/2016] [Indexed: 11/16/2022]
Abstract
PPARδ is highly expressed in skin, especially keratinocytes, and its expression is increased in psoriatic lesions. However, the potential role of PPARδ in the pathogenesis of psoriasis remains undefined. Mice treated with Imiquimod (IMQ) to induce psoriasis can be used to evaluate the pathogenesis of psoriasis, and this model has become one of the most important in vivo research tools for research on the disease. In the current study, we showed that PPARδ was highly expressed in the skin of IMQ-induced psoriasis mice. To further understand the impact of PPARδ in psoriasis, we used these mice in a series of experiments to evaluate the pathogenesis of psoriasis. We found that PPARδ was highly expressed in both psoriatic lesions and normal skin in IMQ-induced psoriasis mice. Furthermore, the expression of PPARδ-relevant lipases was also significantly increased. The PPARδ-selective antagonist GSK3787 ameliorated the observed inflammation in the skin of the experimental mice. Based on these results, PPARδ may be a potential target for the effective treatment of psoriasis.
Collapse
Affiliation(s)
- Xuguo Wang
- Department of Pharmacy, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Yangyang Hao
- Department of Dermatology, 98th Hospital of People's Liberation Army, Anhui Medical University, Huzhou, Zhejiang, China
| | - Xiaohuan Wang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Fudan University, China; Shanghai Public Health Clinical Center, Fudan University, China
| | - Lumei Wang
- Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Yongchun Chen
- Department of Health Toxicology, Department of Basic Courses, The Second Military Medical University, Shanghai, China
| | - Jun Sun
- Department of Pharmacy, General Hospital of Jinan Military Area, Jinan, Shandong Province, China
| | - Jinhong Hu
- Department of Pharmacy, Changhai Hospital, The Second Military Medical University, Shanghai, China.
| |
Collapse
|
20
|
Ramot Y, Mastrofrancesco A, Camera E, Desreumaux P, Paus R, Picardo M. The role of PPARγ-mediated signalling in skin biology and pathology: new targets and opportunities for clinical dermatology. Exp Dermatol 2016; 24:245-51. [PMID: 25644500 DOI: 10.1111/exd.12647] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2015] [Indexed: 12/19/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that modulate the expression of multiple different genes involved in the regulation of lipid, glucose and amino acid metabolism. PPARs and cognate ligands also regulate important cellular functions, including cell proliferation and differentiation, as well as inflammatory responses. This includes a role in mediating skin and pilosebaceous unit homoeostasis: PPARs appear to be essential for maintaining skin barrier permeability, inhibit keratinocyte cell growth, promote keratinocyte terminal differentiation and regulate skin inflammation. They also may have protective effects on human hair follicle (HFs) epithelial stem cells, while defects in PPARγ-mediated signalling may promote the death of these stem cells and thus facilitate the development of cicatricial alopecia (lichen planopilaris). Overall, however, selected PPARγ modulators appear to act as hair growth inhibitors that reduce the proliferation and promote apoptosis of hair matrix keratinocytes. The fact that commonly prescribed PPARγ-modulatory drugs of the thiazolidine-2,4-dione class can exhibit a battery of adverse cutaneous effects underscores the importance of distinguishing beneficial from clinically undesired cutaneous activities of PPARγ ligands and to better understand on the molecular level how PPARγ-regulated cutaneous lipid metabolism and PPARγ-mediated signalling impact on human skin physiology and pathology. Surely, the therapeutic potential that endogenous and exogenous PPARγ modulators may possess in selected skin diseases, ranging from chronic inflammatory hyperproliferative dermatoses like psoriasis and atopic dermatitis, via scarring alopecia and acne can only be harnessed if the complexities of PPARγ signalling in human skin and its appendages are systematically dissected.
Collapse
Affiliation(s)
- Yuval Ramot
- Department of Dermatology, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | |
Collapse
|
21
|
Chon SH, Tannahill R, Yao X, Southall MD, Pappas A. Keratinocyte differentiation and upregulation of ceramide synthesis induced by an oat lipid extract via the activation of PPAR pathways. Exp Dermatol 2016; 24:290-5. [PMID: 25651930 DOI: 10.1111/exd.12658] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2015] [Indexed: 12/13/2022]
Abstract
Activation of peroxisome proliferator-activated receptors (PPARs) has been shown to have an important role in skin barrier function by regulating differentiation and lipid synthesis in keratinocytes. Oat (Avena sativa) has long been used as a soothing agent to relieve skin irritations, and the clinical benefits of topical oat formulations have been proven; however, the mechanistic understanding of oat's mode of action remains unknown. We investigated whether an oat lipid extract could activate PPARs and subsequently increase epidermal lipid synthesis and differentiation markers. Primary human epidermal keratinocytes and transformed cell lines were treated with PPAR agonists and oat lipid extracts to investigate the PPAR agonism. PPAR target genes and epidermal differentiation markers were analysed using quantitative real-time PCR and HPTLC analysis. Oat lipid extract demonstrated robust dual agonism for PPARα and PPARβ/δ, and increased direct PPAR target gene induction in primary human keratinocytes. In addition, oat oil treatment increased both receptor expression and, consistent with the literature on PPARs, oat oil treatment resulted in a significant upregulation of differentiation genes (involucrin, SPRRs and transglutaminase 1) and ceramide processing genes (β-glucocerebrosidase, sphingomyelinases 3 and ABCA12). Further, oat oil treatment in keratinocytes significantly increased ceramide levels (70%), suggesting a functional translation of PPAR activation by oat oil in keratinocytes. Taken together, these results demonstrate that oat lipids possess robust dual agonistic activities for PPARα and PPARβ/δ, increase their gene expression and induce differentiation and ceramide synthesis in keratinocytes, which can collectively improve skin barrier function.
Collapse
Affiliation(s)
- Su-Hyoun Chon
- Johnson & Johnson Skin Research Center, CPPW, A Division of Johnson & Johnson Consumer Companies, Inc., Skillman, NJ, USA
| | | | | | | | | |
Collapse
|
22
|
Schumann T, Adhikary T, Wortmann A, Finkernagel F, Lieber S, Schnitzer E, Legrand N, Schober Y, Nockher WA, Toth PM, Diederich WE, Nist A, Stiewe T, Wagner U, Reinartz S, Müller-Brüsselbach S, Müller R. Deregulation of PPARβ/δ target genes in tumor-associated macrophages by fatty acid ligands in the ovarian cancer microenvironment. Oncotarget 2016; 6:13416-33. [PMID: 25968567 PMCID: PMC4537024 DOI: 10.18632/oncotarget.3826] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/29/2015] [Indexed: 01/04/2023] Open
Abstract
The nuclear receptor peroxisome proliferator-activated receptor β/δ (PPARβ/δ) is a lipid ligand-inducible transcription factor associated with macrophage polarization. However, its function in tumor-associated macrophages (TAMs) has not been investigated to date. Here, we report the PPARβ/δ-regulated transcriptome and cistrome for TAMs from ovarian carcinoma patients. Comparison with monocyte-derived macrophages shows that the vast majority of direct PPARβ/δ target genes are upregulated in TAMs and largely refractory to synthetic agonists, but repressible by inverse agonists. Besides genes with metabolic functions, these include cell type-selective genes associated with immune regulation and tumor progression, e.g., LRP5, CD300A, MAP3K8 and ANGPTL4. This deregulation is not due to increased expression of PPARβ/δ or its enhanced recruitment to target genes. Instead, lipidomic analysis of malignancy-associated ascites revealed high concentrations of polyunsaturated fatty acids, in particular linoleic acid, acting as potent PPARβ/δ agonists in macrophages. These fatty acid ligands accumulate in lipid droplets in TAMs, thereby providing a reservoir of PPARβ/δ ligands. These observations suggest that the deregulation of PPARβ/δ target genes by ligands of the tumor microenvironment contributes to the pro-tumorigenic polarization of ovarian carcinoma TAMs. This conclusion is supported by the association of high ANGPTL4 expression with a shorter relapse-free survival in serous ovarian carcinoma.
Collapse
Affiliation(s)
- Tim Schumann
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Till Adhikary
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Annika Wortmann
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Florian Finkernagel
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Sonja Lieber
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Evelyn Schnitzer
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Nathalie Legrand
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Yvonne Schober
- Metabolomics Core Facility and Institute of Laboratory Medicine and Pathobiochemistry, Philipps University, Marburg, Germany
| | - W Andreas Nockher
- Metabolomics Core Facility and Institute of Laboratory Medicine and Pathobiochemistry, Philipps University, Marburg, Germany
| | - Philipp M Toth
- Medicinal Chemistry Core Facility and Institute of Pharmaceutical Chemistry, Philipps University, Marburg, Germany
| | - Wibke E Diederich
- Medicinal Chemistry Core Facility and Institute of Pharmaceutical Chemistry, Philipps University, Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Silke Reinartz
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | | | - Rolf Müller
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| |
Collapse
|
23
|
Su X, Zhou G, Wang Y, Yang X, Li L, Yu R, Li D. The PPARβ/δ agonist GW501516 attenuates peritonitis in peritoneal fibrosis via inhibition of TAK1-NFκB pathway in rats. Inflammation 2015; 37:729-37. [PMID: 24337677 DOI: 10.1007/s10753-013-9791-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Peritoneal fibrosis is a common consequence of long-term peritoneal dialysis (PD), and peritonitis is a factor in its onset. Agonist-bound peroxisome proliferator-activated receptors (PPARs) function as key regulators of energy metabolism and inflammation. Here, we examined the effects of PPARβ/δ agonist GW501516 on peritonitis in a rat peritoneal fibrosis model. Peritoneal fibrosis secondary to inflammation was induced into uremic rats by daily injection of Dianeal 4.25% PD solutions along with six doses of lipopolysaccharide before commencement of GW501516 treatment. Normal non-uremic rats served as control, and all rats were fed with a control diet or a GW501516-containing diet. Compared to control group, exposure to PD fluids caused peritoneal fibrosis that was accompanied by increased mRNA levels of monocyte chemoattractant protein-1, tumor necrotic factor-α, and interleukin-6 in the uremic rats, and these effects were prevented by GW501516 treatment. Moreover, GW501516 was found to attenuate glucose-stimulated inflammation in cultured rat peritoneal mesothelial cells via inhibition of transforming growth factor-β-activated kinase 1 (TAK1), and nuclear factor kappa B (NFκB) signaling pathway (TAK1-NFκB pathway), a main inflammation regulatory pathway. In conclusion, inhibition of TAK1-NFκB pathway with GW501516 may represent a novel therapeutic approach to ameliorate peritonitis-induced peritoneal fibrosis for patients on PD.
Collapse
Affiliation(s)
- Xuesong Su
- Department of Nephrology, Shengjing Hospital, China Medical University, 36 Sanhao Street, Shenyang, 110004, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
24
|
Cross Talk between Proliferative, Angiogenic, and Cellular Mechanisms Orchestred by HIF-1α in Psoriasis. Mediators Inflamm 2015; 2015:607363. [PMID: 26136626 PMCID: PMC4475568 DOI: 10.1155/2015/607363] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/21/2015] [Indexed: 02/08/2023] Open
Abstract
Psoriasis is a chronic inflammatory skin disease where the altered regulation in angiogenesis, inflammation, and proliferation of keratinocytes are the possible causes of the disease, and the transcription factor “hypoxia-inducible factor 1-alpha” (HIF-1α) is involved in the homeostasis of these three biological phenomena. In this review, the role of HIF-1α in the cross talk between the cytokines and cells of the immunological system involved in the pathogenesis of psoriasis is discussed.
Collapse
|
25
|
Adhikary T, Wortmann A, Schumann T, Finkernagel F, Lieber S, Roth K, Toth PM, Diederich WE, Nist A, Stiewe T, Kleinesudeik L, Reinartz S, Müller-Brüsselbach S, Müller R. The transcriptional PPARβ/δ network in human macrophages defines a unique agonist-induced activation state. Nucleic Acids Res 2015; 43:5033-51. [PMID: 25934804 PMCID: PMC4446423 DOI: 10.1093/nar/gkv331] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/01/2015] [Indexed: 02/06/2023] Open
Abstract
Peroxisome proliferator-activated receptor β/δ (PPARβ/δ) is a lipid ligand-inducible transcription factor with established metabolic functions, whereas its anti-inflammatory function is poorly understood. To address this issue, we determined the global PPARβ/δ-regulated signaling network in human monocyte-derived macrophages. Besides cell type-independent, canonical target genes with metabolic and immune regulatory functions we identified a large number of inflammation-associated NFκB and STAT1 target genes that are repressed by agonists. Accordingly, PPARβ/δ agonists inhibited the expression of multiple pro-inflammatory mediators and induced an anti-inflammatory, IL-4-like morphological phenotype. Surprisingly, bioinformatic analyses also identified immune stimulatory effects. Consistent with this prediction, PPARβ/δ agonists enhanced macrophage survival under hypoxic stress and stimulated CD8+ T cell activation, concomitantly with the repression of immune suppressive target genes and their encoded products CD274 (PD-1 ligand), CD32B (inhibitory Fcγ receptor IIB) and indoleamine 2,3-dioxygenase 1 (IDO-1), as well as a diminished release of the immune suppressive IDO-1 metabolite kynurenine. Comparison with published data revealed a significant overlap of the PPARβ/δ transcriptome with coexpression modules characteristic of both anti-inflammatory and pro-inflammatory cytokines. Our findings indicate that PPARβ/δ agonists induce a unique macrophage activation state with strong anti-inflammatory but also specific immune stimulatory components, pointing to a context-dependent function of PPARβ/δ in immune regulation.
Collapse
Affiliation(s)
- Till Adhikary
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Annika Wortmann
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Tim Schumann
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Florian Finkernagel
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Sonja Lieber
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Katrin Roth
- Cellular Imaging Core Facility, Philipps University, Center for Tumor Biology and Immunology (ZTI), 35043 Marburg, Germany
| | - Philipp M Toth
- Medicinal Chemistry Core Facility and Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Wibke E Diederich
- Medicinal Chemistry Core Facility and Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Lara Kleinesudeik
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Silke Reinartz
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Sabine Müller-Brüsselbach
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| | - Rolf Müller
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, 35043 Marburg, Germany
| |
Collapse
|
26
|
Kopp TI, Lundqvist J, Petersen RK, Oskarsson A, Kristiansen K, Nellemann C, Vogel U. In vitro screening of inhibition of PPAR-γ activity as a first step in identification of potential breast carcinogens. Hum Exp Toxicol 2015; 34:1106-18. [DOI: 10.1177/0960327115569811] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Alcohol consumption and increased estrogen levels are major risk factors for breast cancer, and peroxisome proliferator-activated receptor γ (PPAR-γ) plays an important role in alcohol-induced breast cancer. PPAR-γ activity is inhibited by ethanol, leading to increased aromatase activity and estrogen biosynthesis ultimately leading to breast cancer. If other organic solvents inhibit PPAR-γ activity, they should also lead to increased oestrogen biosynthesis and thus be potential breast carcinogens. Ten commonly used hydrophilic organic solvents were first tested in a cell-based screening assay for inhibitory effects on PPAR-γ transactivation. The chemicals shown to inhibit PPAR-γ were tested with vectors encoding PPAR-γ with deleted AB domains and only the ligand-binding domain to rule out unspecific toxicity. Next, the effects on biosynthesis of estradiol, testosterone and oestrone sulphate were measured in the H295R steroidogenesis assay after incubation with the chemicals. Ethylene glycol, ethyl acetate, and dimethyl sulphoxide inhibited PPAR-γ transactivation in a dose-dependent manner. The inhibitory effect on PPAR-γ was specific for PPAR-γ since the AB domain of PPAR-γ was required for the inhibitory effect. In the second step, ethylene glycol significantly increased production of oestradiol by 19% ( p < 0.05) and ethyl acetate inhibited production of testosterone ( p < 0.05). We here show that screening of 10 commonly used organic solvents for the ability to inhibit PPAR-γ transactivation followed by a well-established steroidogenesis assay for production of sex hormones in exposed H295 R cells may provide a screening tool for potential breast carcinogens. This initial screening thus identified ethylene glycol and possibly ethyl acetate as potential breast carcinogens.
Collapse
Affiliation(s)
- TI Kopp
- National Food Institute, Technical University of Denmark, Søborg, Denmark
| | - J Lundqvist
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Denmark
| | - RK Petersen
- Department of Biology, University of Copenhagen, Copenhagen N, Denmark
| | - A Oskarsson
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Denmark
| | - K Kristiansen
- Department of Biology, University of Copenhagen, Copenhagen N, Denmark
| | - C Nellemann
- National Food Institute, Technical University of Denmark, Søborg, Denmark
| | - U Vogel
- National Research Centre for the Working Environment, Copenhagen Ø, Denmark
| |
Collapse
|
27
|
Sertznig P, Reichrath J. Peroxisome proliferator-activated receptors (PPARs) in dermatology. DERMATO-ENDOCRINOLOGY 2014. [DOI: 10.4161/derm.15025] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
28
|
Kubanov AA, Mineyeva AA. Molecular and genetic studies of the predisposition to the development of psoriasis among the population of the Russian Federation: a study of polymorphisms of TNFAIP3, TNIP1, TYK2 and REL genes. VESTNIK DERMATOLOGII I VENEROLOGII 2014. [DOI: 10.25208/0042-4609-2014-90-5-73-80] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Goal. To study the role of polymorphic variants of genes encoding proteins of the signaling pathway of the nuclear transcription factor kappa-B, NF-kB (TNFAIP3 (rs610604), TNIP1 (rs17728338), TYK2 (rs12720356) and REL (rs702873)) in the predisposition to psoriasis among the Caucasian population of the Russian Federation. Materials and methods. The authors studied markers of the predisposition to the development of psoriasis such as TNFAIP3 (rs610604), TNIP1 (rs17728338), TYK2 (rs12720356) and REL (rs702873) using Real-Time PCR methods and allele-specific oligonucleotide probes and PFLP (Restriction Fragment Length Polymorphism) assay in whole blood samples taken from psoriatic patients (n = 301). To compare the frequency of genotypes, the authors used data obtained as a result of the genotyping of DNAs taken from healthy volunteers (n = 109). Results. The authors analyzed particular features of the distribution of polymorphic variants of TNFAIP3 (rs610604), TNIP1 (rs17728338), TYK2 (rs1272035) и REL (rs702873) genes in a sample of 301 psoriatic patients and 109 healthy volunteers from the Russian Federation. They revealed reliable differences in the frequency of registration of genotypes of these genes. The authors also determined genotypes considered as risk factors with regard to the development of psoriasis (TNFAIP3-A/C, TNIP1-A/G, TNIP1-A/A and TYK2-T/T) as well as protector genotypes (TNFAIP3-A/Ä, REL-A/A and TYK2-T/G)
Collapse
|
29
|
Neels JG, Grimaldi PA. Physiological functions of peroxisome proliferator-activated receptor β. Physiol Rev 2014; 94:795-858. [PMID: 24987006 DOI: 10.1152/physrev.00027.2013] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The peroxisome proliferator-activated receptors, PPARα, PPARβ, and PPARγ, are a family of transcription factors activated by a diversity of molecules including fatty acids and fatty acid metabolites. PPARs regulate the transcription of a large variety of genes implicated in metabolism, inflammation, proliferation, and differentiation in different cell types. These transcriptional regulations involve both direct transactivation and interaction with other transcriptional regulatory pathways. The functions of PPARα and PPARγ have been extensively documented mainly because these isoforms are activated by molecules clinically used as hypolipidemic and antidiabetic compounds. The physiological functions of PPARβ remained for a while less investigated, but the finding that specific synthetic agonists exert beneficial actions in obese subjects uplifted the studies aimed to elucidate the roles of this PPAR isoform. Intensive work based on pharmacological and genetic approaches and on the use of both in vitro and in vivo models has considerably improved our knowledge on the physiological roles of PPARβ in various cell types. This review will summarize the accumulated evidence for the implication of PPARβ in the regulation of development, metabolism, and inflammation in several tissues, including skeletal muscle, heart, skin, and intestine. Some of these findings indicate that pharmacological activation of PPARβ could be envisioned as a therapeutic option for the correction of metabolic disorders and a variety of inflammatory conditions. However, other experimental data suggesting that activation of PPARβ could result in serious adverse effects, such as carcinogenesis and psoriasis, raise concerns about the clinical use of potent PPARβ agonists.
Collapse
Affiliation(s)
- Jaap G Neels
- Institut National de la Santé et de la Recherche Médicale U 1065, Mediterranean Center of Molecular Medicine (C3M), Team "Adaptive Responses to Immuno-metabolic Dysregulations," Nice, France; and Faculty of Medicine, University of Nice Sophia-Antipolis, Nice, France
| | - Paul A Grimaldi
- Institut National de la Santé et de la Recherche Médicale U 1065, Mediterranean Center of Molecular Medicine (C3M), Team "Adaptive Responses to Immuno-metabolic Dysregulations," Nice, France; and Faculty of Medicine, University of Nice Sophia-Antipolis, Nice, France
| |
Collapse
|
30
|
Montagner A, Delgado MB, Tallichet-Blanc C, Chan JSK, Sng MK, Mottaz H, Degueurce G, Lippi Y, Moret C, Baruchet M, Antsiferova M, Werner S, Hohl D, Al Saati T, Farmer PJ, Tan NS, Michalik L, Wahli W. Src is activated by the nuclear receptor peroxisome proliferator-activated receptor β/δ in ultraviolet radiation-induced skin cancer. EMBO Mol Med 2014; 6:80-98. [PMID: 24203162 PMCID: PMC3936491 DOI: 10.1002/emmm.201302666] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 09/25/2013] [Accepted: 09/26/2013] [Indexed: 02/04/2023] Open
Abstract
Although non-melanoma skin cancer (NMSC) is the most common human cancer and its incidence continues to rise worldwide, the mechanisms underlying its development remain incompletely understood. Here, we unveil a cascade of events involving peroxisome proliferator-activated receptor (PPAR) β/δ and the oncogene Src, which promotes the development of ultraviolet (UV)-induced skin cancer in mice. UV-induced PPARβ/δ activity, which directly stimulated Src expression, increased Src kinase activity and enhanced the EGFR/Erk1/2 signalling pathway, resulting in increased epithelial-to-mesenchymal transition (EMT) marker expression. Consistent with these observations, PPARβ/δ-null mice developed fewer and smaller skin tumours, and a PPARβ/δ antagonist prevented UV-dependent Src stimulation. Furthermore, the expression of PPARβ/δ positively correlated with the expression of SRC and EMT markers in human skin squamous cell carcinoma (SCC), and critically, linear models applied to several human epithelial cancers revealed an interaction between PPARβ/δ and SRC and TGFβ1 transcriptional levels. Taken together, these observations motivate the future evaluation of PPARβ/δ modulators to attenuate the development of several epithelial cancers.
Collapse
Affiliation(s)
- Alexandra Montagner
- Center for Integrative Genomics, National Research Center Frontiers in Genetics, University of LausanneLe Genopode, Lausanne, Switzerland
| | - Maria B Delgado
- Center for Integrative Genomics, National Research Center Frontiers in Genetics, University of LausanneLe Genopode, Lausanne, Switzerland
| | - Corinne Tallichet-Blanc
- Center for Integrative Genomics, National Research Center Frontiers in Genetics, University of LausanneLe Genopode, Lausanne, Switzerland
| | - Jeremy S K Chan
- School of Biological Sciences, Nanyang Technological UniversityNanyang Drive, Singapore, Singapore
| | - Ming K Sng
- School of Biological Sciences, Nanyang Technological UniversityNanyang Drive, Singapore, Singapore
| | - Hélène Mottaz
- Center for Integrative Genomics, National Research Center Frontiers in Genetics, University of LausanneLe Genopode, Lausanne, Switzerland
| | - Gwendoline Degueurce
- Center for Integrative Genomics, National Research Center Frontiers in Genetics, University of LausanneLe Genopode, Lausanne, Switzerland
| | - Yannick Lippi
- GeT-TRiX Facility, INRA ToxAlim, UMR1331Chemin de Tournefeuille, Toulouse Cedex, France
| | - Catherine Moret
- Center for Integrative Genomics, National Research Center Frontiers in Genetics, University of LausanneLe Genopode, Lausanne, Switzerland
| | - Michael Baruchet
- Center for Integrative Genomics, National Research Center Frontiers in Genetics, University of LausanneLe Genopode, Lausanne, Switzerland
| | - Maria Antsiferova
- Department of Biology, Institute of Molecular Health Sciences, ETH ZurichSchafmattstrasse, Zurich, Switzerland
| | - Sabine Werner
- Department of Biology, Institute of Molecular Health Sciences, ETH ZurichSchafmattstrasse, Zurich, Switzerland
| | - Daniel Hohl
- Department of Dermatology, University Hospital of Lausanne (CHUV)Lausanne, Switzerland
| | - Talal Al Saati
- INSERM/UPS, US006/CREFRE, Histopathology Facility, Place du Docteur BaylacCHU Purpan, Toulouse Cedex, France
| | - Pierre J Farmer
- Exploratory Biomarker Analysis, Biomarker Technologies, Bioinformatics, Non Clinical Development, Merck Serono International S.A. SwitzerlandChemin des Mines, Geneva, Switzerland
| | - Nguan S Tan
- School of Biological Sciences, Nanyang Technological UniversityNanyang Drive, Singapore, Singapore
- Institute of Molecular and Cell Biology, Biopolis DriveProteos, Singapore, Singapore
| | - Liliane Michalik
- Center for Integrative Genomics, National Research Center Frontiers in Genetics, University of LausanneLe Genopode, Lausanne, Switzerland
- *Corresponding author: Tel: +41 21 692 41 10; Fax: +41 21 692 41 15; E-mail:
| | - Walter Wahli
- Center for Integrative Genomics, National Research Center Frontiers in Genetics, University of LausanneLe Genopode, Lausanne, Switzerland
- Lee Kong Chian School of Medicine, Imperial College London, Nanyang Technological UniversitySingapore, Singapore
- **Corresponding author: Tel: +41 21 692 41 10; Fax: +41 21 692 41 15; E-mail:
| |
Collapse
|
31
|
Valente Duarte de Sousa IC. Novel pharmacological approaches for the treatment of acne vulgaris. Expert Opin Investig Drugs 2014; 23:1389-410. [PMID: 24890096 DOI: 10.1517/13543784.2014.923401] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Acne vulgaris is the most common skin disease worldwide; yet, current treatment options, although effective, are associated with unwanted side effects, chronicity, relapses and recurrences. The adequate control of the four pathogenic mechanisms, involved in the appearance of acne lesions, is paramount to treatment success. AREAS COVERED The authors discuss and evaluate the pathogenic pathways related to the mechanisms of action of novel molecules, which are currently under investigation for the treatment of acne vulgaris. The manuscript is based on comprehensive searches made through PubMed, GoogleScholar and ClinicalTrial.gov, using different combination of key words, which include acne vulgaris, pathogenesis, treatment, sebogenesis and Propionibacterium acnes. EXPERT OPINION In the near future, more effective treatments with fewer side effects are expected. The use of topical antiandrogens, acetylcholine inhibitors and PPAR modulators seem to be promising options for controlling sebum production. Retinoic acid metabolism-blocking agents and IL-1α inhibitors have the potential to become legitimate alternative options to retinoid therapy in the management of infundibular dyskeratosis. Indeed, the authors believe that there will likely be a decline in the use of antibiotics for controlling P. acnes colonization and targeting the inflammation cascade.
Collapse
|
32
|
Preclinical Studies of a Specific PPARγ Modulator in the Control of Skin Inflammation. J Invest Dermatol 2014; 134:1001-1011. [DOI: 10.1038/jid.2013.448] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 08/27/2013] [Accepted: 09/10/2013] [Indexed: 12/29/2022]
|
33
|
Rodríguez-Cerdeira C, Molares-Vila A, Sánchez-Blanco E, Sánchez-Blanco B. Study on Certain Biomarkers of Inflammation in Psoriasis Through "OMICS" Platforms. Open Biochem J 2014; 8:21-34. [PMID: 24688608 PMCID: PMC3970352 DOI: 10.2174/1874091x01408010021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Revised: 12/11/2013] [Accepted: 12/14/2013] [Indexed: 12/14/2022] Open
Abstract
Background: In recent years, research on psoriasis has focused on the identification of biomarkers for the diagnosis, pathogenesis, prognosis, or therapeutic response of the disease. These studies could provide insights into the susceptibility and natural history of psoriasis. The identification of biomarkers related to comorbidities in psoriasis, such as arthritis, cardiovascular disease, and the metabolic syndrome, is of special clinical interest. Materials and Methods: We performed an extensive review on psoriasis biomarkers, including cytokine and growth factors, in the literature published between 1997 and 2013, including cross-references of any retrieved articles. We also included some data from our own studies. Results: This review presents current knowledge of soluble biomarkers in psoriasis, including cytokines, chemokines, proangiogenic mediators, growth factors, antimicrobial proteins, neuropeptides, and oxidative stress markers. Conclusion: In conclusion, a number of studies have been conducted with the aim of establishing soluble biomarkers for psoriasis. Most of the biomarkers that have been studied do not meet the criteria for a clinically useful biomarker. Further work is needed to establish a role for soluble biomarkers in the diagnosis and treatment of psoriasis, with a special focus on biomarkers for psoriasis comorbidities, such as arthritis, cardiovascular disease, and the metabolic syndrome.
Collapse
Affiliation(s)
| | - A Molares-Vila
- Department of Analytical Chemistry, University of Vigo, Spain
| | | | - B Sánchez-Blanco
- Postgraduate researcher, Department of Emergency, CHUVI, Vigo, Spain
| |
Collapse
|
34
|
An integrated epigenetic and transcriptomic analysis reveals distinct tissue-specific patterns of DNA methylation associated with atopic dermatitis. J Invest Dermatol 2014; 134:1873-1883. [PMID: 24739813 DOI: 10.1038/jid.2014.87] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 01/29/2014] [Accepted: 01/29/2014] [Indexed: 12/22/2022]
Abstract
Epigenetic alterations are increasingly recognized as mechanisms for disease-associated changes in genome function and important risk factors for complex diseases. The epigenome differs between cell types and so far has been characterized in few human tissues only. In order to identify disease-associated DNA methylation differences for atopic dermatitis (AD), we investigated DNA from whole blood, T cells, B cells, as well as lesional and non-lesional epidermis from AD patients and healthy controls. To elicit functional links, we examined epidermal mRNA expression profiles. No genome-wide significant DNA methylation differences between AD cases and controls were observed in whole blood, T cells, and B cells, and, in general, intra-individual differences in DNA methylation were larger than interindividual differences. However, striking methylation differences were observed between lesional epidermis from patients and healthy control epidermis for various CpG sites, which partly correlated with altered transcript levels of genes predominantly relevant for epidermal differentiation and innate immune response. Significant DNA methylation differences were discordant in skin and blood samples, suggesting that blood is not an ideal surrogate for skin tissue. Our pilot study provides preliminary evidence for functionally relevant DNA methylation differences associated with AD, particularly in the epidermis, and represents a starting point for future investigations of epigenetic mechanisms in AD.
Collapse
|
35
|
Schmuth M, Moosbrugger-Martinz V, Blunder S, Dubrac S. Role of PPAR, LXR, and PXR in epidermal homeostasis and inflammation. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:463-73. [PMID: 24315978 DOI: 10.1016/j.bbalip.2013.11.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Revised: 11/18/2013] [Accepted: 11/23/2013] [Indexed: 12/19/2022]
Abstract
Epidermal lipid synthesis and metabolism are regulated by nuclear hormone receptors (NHR) and in turn epidermal lipid metabolites can serve as ligands to NHR. NHR form a large superfamily of receptors modulating gene transcription through DNA binding. A subgroup of these receptors is ligand-activated and heterodimerizes with the retinoid X receptor including peroxisome proliferator-activated receptor (PPAR), liver X receptor (LXR) and pregnane X receptor (PXR). Several isotypes of these receptors exist, all of which are expressed in skin. In keratinocytes, ligand activation of PPARs and LXRs stimulates differentiation, induces lipid accumulation, and accelerates epidermal barrier regeneration. In the cutaneous immune system, ligand activation of all three receptors, PPAR, LXR, and PXR, has inhibitory properties, partially mediated by downregulation of the NF-kappaB pathway. PXR also has antifibrotic effects in the skin correlating with TGF-beta inhibition. In summary, ligands of PPAR, LXR and PXR exert beneficial therapeutic effects in skin disease and represent promising targets for future therapeutic approaches in dermatology. This article is part of a Special Issue entitled The Important Role of Lipids in the Epidermis and their Role in the Formation and Maintenance of the Cutaneous Barrier. Guest Editors: Kenneth R. Feingold and Peter Elias.
Collapse
Affiliation(s)
- Matthias Schmuth
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria.
| | | | - Stefan Blunder
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria
| | - Sandrine Dubrac
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria.
| |
Collapse
|
36
|
Allergen-induced dermatitis causes alterations in cutaneous retinoid-mediated signaling in mice. PLoS One 2013; 8:e71244. [PMID: 23977003 PMCID: PMC3744553 DOI: 10.1371/journal.pone.0071244] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 06/27/2013] [Indexed: 12/11/2022] Open
Abstract
Nuclear receptor-mediated signaling via RARs and PPARδ is involved in the regulation of skin homeostasis. Moreover, activation of both RAR and PPARδ was shown to alter skin inflammation. Endogenous all-trans retinoic acid (ATRA) can activate both receptors depending on specific transport proteins: Fabp5 initiates PPARδ signaling whereas Crabp2 promotes RAR signaling. Repetitive topical applications of ovalbumin (OVA) in combination with intraperitoneal injections of OVA or only intraperitoneal OVA applications were used to induce allergic dermatitis. In our mouse model, expression of IL-4, and Hbegf increased whereas expression of involucrin, Abca12 and Spink5 decreased in inflamed skin, demonstrating altered immune response and epidermal barrier homeostasis. Comprehensive gene expression analysis showed alterations of the cutaneous retinoid metabolism and retinoid-mediated signaling in allergic skin immune response. Notably, ATRA synthesis was increased as indicated by the elevated expression of retinaldehyde dehydrogenases and increased levels of ATRA. Consequently, the expression pattern of genes downstream to RAR was altered. Furthermore, the increased ratio of Fabp5 vs. Crabp2 may indicate an up-regulation of the PPARδ pathway in allergen-induced dermatitis in addition to the altered RAR signaling. Thus, our findings suggest that ATRA levels, RAR-mediated signaling and signaling involved in PPARδ pathways are mainly increased in allergen-induced dermatitis and may contribute to the development and/or maintenance of allergic skin diseases.
Collapse
|
37
|
Kim T, Yang Q. Peroxisome-proliferator-activated receptors regulate redox signaling in the cardiovascular system. World J Cardiol 2013; 5:164-174. [PMID: 23802046 PMCID: PMC3691497 DOI: 10.4330/wjc.v5.i6.164] [Citation(s) in RCA: 280] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/06/2013] [Accepted: 05/17/2013] [Indexed: 02/06/2023] Open
Abstract
Peroxisome-proliferator-activated receptors (PPARs) comprise three subtypes (PPARα, δ and γ) to form a nuclear receptor superfamily. PPARs act as key transcriptional regulators of lipid metabolism, mitochondrial biogenesis, and anti-oxidant defense. While their roles in regulating lipid metabolism have been well established, the role of PPARs in regulating redox activity remains incompletely understood. Since redox activity is an integral part of oxidative metabolism, it is not surprising that changes in PPAR signaling in a specific cell or tissue will lead to alteration of redox state. The effects of PPAR signaling are directly related to PPAR expression, protein activities and PPAR interactions with their coregulators. The three subtypes of PPARs regulate cellular lipid and energy metabolism in most tissues in the body with overlapping and preferential effects on different metabolic steps depending on a specific tissue. Adding to the complexity, specific ligands of each PPAR subtype may also display different potencies and specificities of their role on regulating the redox pathways. Moreover, the intensity and extension of redox regulation by each PPAR subtype are varied depending on different tissues and cell types. Both beneficial and adverse effects of PPAR ligands against cardiovascular disorders have been extensively studied by many groups. The purpose of the review is to summarize the effects of each PPAR on regulating redox and the underlying mechanisms, as well as to discuss the implications in the cardiovascular system.
Collapse
|
38
|
Stockert J, Wolf A, Kaddatz K, Schnitzer E, Finkernagel F, Meissner W, Müller-Brüsselbach S, Kracht M, Müller R. Regulation of TAK1/TAB1-mediated IL-1β signaling by cytoplasmic PPARβ/δ. PLoS One 2013; 8:e63011. [PMID: 23646170 PMCID: PMC3639976 DOI: 10.1371/journal.pone.0063011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 03/27/2013] [Indexed: 11/19/2022] Open
Abstract
The peroxisome proliferator-activated receptor subtypes PPARα, PPARβ/δ, PPARγ are members of the steroid hormone receptor superfamily with well-established functions in transcriptional regulation. Here, we describe an unexpected cytoplasmic function of PPARβ/δ. Silencing of PPARβ/δ expression interferes with the expression of a large subset of interleukin-1β (IL-1β)-induced target genes in HeLa cells, which is preceded by an inhibition of the IL-1β-induced phosphorylation of TAK1 and its downstream effectors, including the NFκBα inhibitor IκBα (NFKBIA) and the NFκBα subunit p65 (RELA). PPARβ/δ enhances the interaction between TAK1 and the small heat-shock protein HSP27, a known positive modulator of TAK1-mediated IL-1β signaling. Consistent with these findings, PPARβ/δ physically interacts with both the endogenous cytoplasmic TAK1/TAB1 complex and HSP27, and PPARβ/δ overexpression increases the TAK1-induced transcriptional activity of NFκB. These observations suggest that PPARβ/δ plays a role in the assembly of a cytoplasmic multi-protein complex containing TAK1, TAB1, HSP27 and PPARβ/δ, and thereby participates in the NFκB response to IL-1β.
Collapse
Affiliation(s)
- Josefine Stockert
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Alexander Wolf
- Rudolf Buchheim Institute for Pharmacology, Giessen, Germany
| | - Kerstin Kaddatz
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Evelyn Schnitzer
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Florian Finkernagel
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | - Wolfgang Meissner
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
| | | | - Michael Kracht
- Rudolf Buchheim Institute for Pharmacology, Giessen, Germany
| | - Rolf Müller
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Marburg, Germany
- * E-mail:
| |
Collapse
|
39
|
Remels AHV, Gosker HR, Bakker J, Guttridge DC, Schols AMWJ, Langen RCJ. Regulation of skeletal muscle oxidative phenotype by classical NF-κB signalling. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1313-25. [PMID: 23563317 DOI: 10.1016/j.bbadis.2013.03.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 03/04/2013] [Accepted: 03/26/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND Impairments in skeletal muscle oxidative phenotype (OXPHEN) have been linked to the development of insulin resistance, metabolic inflexibility and progression of the metabolic syndrome and have been associated with progressive disability in diseases associated with chronic systemic inflammation. We previously showed that the inflammatory cytokine tumour necrosis factor-α (TNF-α) directly impairs muscle OXPHEN but underlying molecular mechanisms remained unknown. Interestingly, the inflammatory signalling pathway classical nuclear factor-κB (NF-κB) is activated in muscle in abovementioned disorders. Therefore, we hypothesised that muscle activation of classical NF-κB signalling is sufficient and required for inflammation-induced impairment of muscle OXPHEN. METHODS Myotubes from mouse and human muscle cell lines were subjected to activation or blockade of the classical NF-κB pathway. In addition, wild-type and MISR (muscle-specific inhibition of classical NF-κB) mice were injected intra-muscularly with TNF-α. Markers and key regulators of muscle OXPHEN were investigated. RESULTS Classical NF-κB activation diminished expression of oxidative phosphorylation (OXPHOS) sub-units, slow myosin heavy chain expression, activity of mitochondrial enzymes and potently reduced intra-cellular ATP levels. Accordingly, PGC-1/PPAR/NRF-1/Tfam signalling, the main pathway controlling muscle OXPHEN, was impaired upon classical NF-κB activation which required intact p65 trans-activation domains and depended on de novo gene transcription. Unlike wild-type myotubes, IκBα-SR myotubes (blocked classical NF-κB signalling) were refractory to TNF-α-induced impairments in OXPHEN and its regulation by the PGC-1/PPAR/NRF-1/Tfam cascade. In line with in vitro data, NF-κB blockade in vivo abrogated TNF-α-induced reductions in PGC-1α expression. CONCLUSION Classical NF-κB activation impairs skeletal muscle OXPHEN.
Collapse
Affiliation(s)
- A H V Remels
- NUTRIM School for Nutrition, Toxicology and Metabolism, Department of Respiratory Medicine, Maastricht University Medical Centre +, Maastricht, The Netherlands.
| | | | | | | | | | | |
Collapse
|
40
|
Hyter S, Indra AK. Nuclear hormone receptor functions in keratinocyte and melanocyte homeostasis, epidermal carcinogenesis and melanomagenesis. FEBS Lett 2013; 587:529-41. [PMID: 23395795 PMCID: PMC3670764 DOI: 10.1016/j.febslet.2013.01.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 12/12/2012] [Accepted: 01/18/2013] [Indexed: 12/19/2022]
Abstract
Skin homeostasis is maintained, in part, through regulation of gene expression orchestrated by type II nuclear hormone receptors in a cell and context specific manner. This group of transcriptional regulators is implicated in various cellular processes including epidermal proliferation, differentiation, permeability barrier formation, follicular cycling and inflammatory responses. Endogenous ligands for the receptors regulate actions during skin development and maintenance of tissue homeostasis. Type II nuclear receptor signaling is also important for cellular crosstalk between multiple cell types in the skin. Overall, these nuclear receptors are critical players in keratinocyte and melanocyte biology and present targets for cutaneous disease management.
Collapse
Affiliation(s)
- Stephen Hyter
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, USA
- Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon, USA
| | - Arup K Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, USA
- Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon, USA
- Environmental Health Science Center, Oregon State University, Corvallis, Oregon, USA
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
41
|
Tian S, Krueger JG, Li K, Jabbari A, Brodmerkel C, Lowes MA, Suárez-Fariñas M. Meta-analysis derived (MAD) transcriptome of psoriasis defines the "core" pathogenesis of disease. PLoS One 2012; 7:e44274. [PMID: 22957057 PMCID: PMC3434204 DOI: 10.1371/journal.pone.0044274] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 07/31/2012] [Indexed: 12/14/2022] Open
Abstract
The cause of psoriasis, a common chronic inflammatory skin disease, is not fully understood. Microarray experiments have been widely used in recent years to identify genes associated with psoriasis pathology, by comparing expression levels of lesional (LS) with adjacent non-lesional (NL) skin. It is commonly observed that the differentially expressed genes (DEGs) differ greatly across experiments, due to variations introduced in the microarray experiment pipeline. Therefore, a statistically based meta-analytic approach, which combines the results of individual studies, is warranted. In this study, a meta-analysis was conducted on 5 microarray data sets, including 193 LS and NL pairs. We termed this the Meta-Analysis Derived (MAD) transcriptome. In “MAD-5” transcriptome, 677 genes were up-regulated and 443 were down-regulated in LS skin compared to NL skin. This represents a much larger set than the intersection of DEGs of these 5 studies, which consisted of 100 DEGs. We also analyzed 3 of the studies conducted on the Affymetrix hgu133plus2 chips and found a greater number of DEGs (1084 up- and 748 down-regulated). Top canonical pathways over-represented in the MAD transcriptome include Atherosclerosis Signaling and Fatty Acid Metabolism, while several “new” genes identified are involved in Cardiovascular Development and Lipid Metabolism. These findings highlight the relationship between psoriasis and systemic manifestations such as the metabolic syndrome and cardiovascular disease. Then, the Meta Threshold Gradient Descent Regularization (MTGDR) algorithm was used to select potential markers distinguishing LS and NL skin. The resulting set (20 genes) contained many genes that were part of the residual disease genomic profile (RDGP) or “molecular scar” after successful treatment, and also genes subject to differential methylation in LS tissues. To conclude, this MAD transcriptome yielded a reference list of reliable psoriasis DEGs, and represents a robust pool of candidates for further discovery of pathogenesis and treatment evaluation.
Collapse
Affiliation(s)
- Suyan Tian
- Center for Clinical and Translational Science, The Rockefeller University, New York, New York, United States of America
| | - James G. Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, United States of America
| | - Katherine Li
- Immunology & Biomarkers, Janssen Research & Development, Radnor, Pennsylvania, United States of America
| | - Ali Jabbari
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, United States of America
| | - Carrie Brodmerkel
- Immunology & Biomarkers, Janssen Research & Development, Radnor, Pennsylvania, United States of America
| | - Michelle A. Lowes
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, United States of America
| | - Mayte Suárez-Fariñas
- Center for Clinical and Translational Science, The Rockefeller University, New York, New York, United States of America
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
42
|
MINEEVA AA, KOZHUSHNAYA OS, VOLNUKHIN VA, FRIGO NV, ZNAMENSKAYA LF, KUBANOV AA, MELEKHINA LE. Study of the genetic factors predisposing to the development of psoriasis. VESTNIK DERMATOLOGII I VENEROLOGII 2012. [DOI: 10.25208/vdv681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Background papers on psoriasis epidemiology, pathogenesis and genetics are presented. Special attention is given to genetic factors of the aptitude to psoriasis development. Were analysed researches, dedicated to the genome-wide screening of associations of polymorphic genetic locus with psoriasis development. Obtained results allow to reveal pathogenic psoriasis mechanisms, to forecast the character of the clinical course of the disease, as well as the efficiency of therapy and forecast the risk of psoriasis origination at patient’s relatives.
Collapse
|
43
|
Sahu RP, DaSilva SC, Rashid B, Martel KC, Jernigan D, Mehta SR, Mohamed DR, Rezania S, Bradish JR, Armstrong AB, Warren S, Konger RL. Mice lacking epidermal PPARγ exhibit a marked augmentation in photocarcinogenesis associated with increased UVB-induced apoptosis, inflammation and barrier dysfunction. Int J Cancer 2012; 131:E1055-66. [PMID: 22467332 DOI: 10.1002/ijc.27562] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 02/16/2012] [Indexed: 12/21/2022]
Abstract
Recent studies suggest that peroxisome proliferator-activated receptor gamma (PPARγ) agonists may have cancer chemopreventive activity. Other studies have shown that loss of epidermal PPARγ results in enhanced chemical carcinogenesis in mice via unknown mechanisms. However, ultraviolet B (UVB) exposure represents the primary etiological agent for skin cancer formation and the role of PPARγ in photobiology and photocarcinogenesis is unknown. In previous studies, we demonstrated that UVB irradiation of cells results in the formation of oxidized glycerophosphocholines that exhibit PPARγ ligand activity. We therefore hypothesized that PPARγ would prove to be a chemopreventive target in photocarcinogenesis. We first showed that UVB irradiation of mouse skin causes generation of PPARγ agonist species in vivo. We then generated SKH-1 hairless, albino mice deficient in epidermal Pparg (Pparg-/-(epi)) using a cytokeratin 14 driven Cre-LoxP strategy. Using a chronic model of UVB photocarcinogenesis, we next showed that Pparg-/-(epi) mice exhibit an earlier onset of tumor formation, increased tumor burden and tumor progression. Increased tumor burden in Pparg-/-(epi) mice was accompanied by a significant increase in epidermal hyperplasia and p53 positive epidermal cells in surrounding skin lacking tumors. After acute UVB irradiation, Pparg-/-(epi) mice exhibited an augmentation of both UVB-induced Caspase 3/7 activity and inflammation. Increased apoptosis and inflammation was also observed after treatment with the PPARγ antagonist GW9662. With chronic UVB irradiation, Pparg-/-(epi) mice exhibited a sustained increase in erythema and transepidermal water loss relative to wildtype littermates. This suggests that PPARγ agonists could have possible chemopreventive activity in non-melanoma skin cancer.
Collapse
Affiliation(s)
- Ravi P Sahu
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Montagner A, Rando G, Degueurce G, Leuenberger N, Michalik L, Wahli W. New insights into the role of PPARs. Prostaglandins Leukot Essent Fatty Acids 2011; 85:235-43. [PMID: 21778043 DOI: 10.1016/j.plefa.2011.04.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are fatty acid-activated transcription factors belonging to the nuclear hormone receptor family. While PPARs are best known as regulators of energy homeostasis, evidence also has accumulated recently for their involvement in basic cellular functions. We review novel insights into PPAR functions in skin wound healing and liver, with emphasis on PPARβ/δ and PPARα, respectively. Activation of PPARβ/δ expression in response to injury promotes keratinocyte survival, directional sensing, and migration over the wound bed. In addition, interleukin (IL)-1 produced by the keratinocytes activates PPARβ/δ expression in the underlying fibroblasts, which hinders the mitotic activity of keratinocytes via inhibition of IL-1 signaling. Initially, roles were identified for PPARα in fatty acid catabolism. However, PPARα is also involved in downregulating many genes in female mammals. We have elucidated the mechanism of this repression, which requires sumoylation of PPARα. Physiologically, this control confers protection against estrogen-induced intrahepatic cholestasis.
Collapse
Affiliation(s)
- Alexandra Montagner
- Center for Integrative Genomics, National Research Center "Frontiers in Genetics", University of Lausanne, Genopode Building, CH-1015 Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
45
|
Di R, Huang MT, Ho CT. Anti-inflammatory activities of mogrosides from Momordica grosvenori in murine macrophages and a murine ear edema model. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2011; 59:7474-7481. [PMID: 21631112 DOI: 10.1021/jf201207m] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Momordica grosvenori (Luo Han Guo), grown primarily in Guangxi province in China, has been traditionally used for thousands of years by the Chinese to make hot drinks for the treatment of sore throat and the removal of phlegm. The natural noncaloric sweetening triterpenoid glycosides (mogrosides) contained in the M. grosvenori fruits are also antioxidative, anticarcinogenic, and helpful in preventing diabetic complications. The aim of this study was to assess the anti-inflammatory properties of mogrosides in both murine macrophage RAW 264.7 cells and a murine ear edema model. The results indicate that mogrosides can inhibit inflammation induced by lipopolysaccharides (LPS) in RAW 264.7 cells by down-regulating the expression of key inflammatory genes iNOS, COX-2, and IL-6 and up-regulating some inflammation protective genes such as PARP1, BCL2l1, TRP53, and MAPK9. Similarly, in the murine ear edema model, 12-O-tetradecanoylphorbol-13-acetate-induced inflammation was inhibited by mogrosides by down-regulating COX-2 and IL-6 and up-regulating PARP1, BCL2l1, TRP53, MAPK9, and PPARδ gene expression. This study shows that the anticancer and antidiabetic effects of M. grosvenori may result in part from its anti-inflammatory activity.
Collapse
Affiliation(s)
- Rong Di
- Department of Plant Biology and Pathology, Rutgers University, New Brunswick, NJ 08901, USA.
| | | | | |
Collapse
|
46
|
Sertznig P, Reichrath J. Peroxisome proliferator-activated receptors (PPARs) in dermatology: Challenge and promise. DERMATO-ENDOCRINOLOGY 2011; 3:130-5. [PMID: 22110772 DOI: 10.4161/derm.3.3.15025] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 01/17/2011] [Accepted: 02/01/2011] [Indexed: 02/03/2023]
Abstract
Since their discovery it has become clear that peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors involved in the genetic regulation of the lipid metabolism and energy homoeostasis. Subsequently, accumulating evidence suggests a role of PPARs in genomic pathways including the regulation of cell growth, apoptosis and differentiation. These findings indicate that PPARs and PPAR agonists play an important role in inflammatory responses and tumor promotion. Because of their diverse biologic activities on keratinocytes and other skin cells, PPARs represent a major research target for the understanding and treatment of many skin pathologies, such as hyperproliferative and inflammatory diseases. Overmore recent clinical trials identified PPARs as promising drug targets for the prevention and treatment of various diseases in the field of dermatology. The present review summarizes the current knowledge of PPAR functions in various skin disorders particularly those involving inflammation and epidermal hyperproliferation (i.e., psoriasis, atopic dermatitis, acne, scleroderma, skin malignancies).
Collapse
Affiliation(s)
- Pit Sertznig
- Department of Dermatology; RWTH Aachen University Hospital; Aachen
| | | |
Collapse
|
47
|
Nakai K, Yoneda K, Ishihara Y, Ohmori K, Moriue T, Igarashi J, Kohno M, Kosaka H, Kubota Y. Lipid peroxidation-induced VEGF expression in the skin of KKAy obese mice. Exp Dermatol 2011; 20:388-93. [PMID: 21355888 DOI: 10.1111/j.1600-0625.2010.01223.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Obesity is known to be associated with a number of effects on skin physiology. KKA(y) obese mouse is a model of type 2 diabetes characterized by systemic oxidative stress because of severe obesity, hypertriglyceridaemia, hyperglycaemia and hyperinsulinaemia. We investigated lipid peroxidation and vascular endothelial growth factor (VEGF) expression in the skin of KKA(y) obese mice. We also investigated the effect of lipid peroxidation derivatives on VEGF production and proliferation in human epidermal keratinocyte cell line (HaCaT). The lipid peroxidation level in the mouse skin tissue was determined by measuring the levels of thiobarbituric acid-reactive substances. The levels of VEGF expression, p44/p42 mitogen-activated protein kinase (MAPK) activation and CD36 expression were analysed by Western blot. Their localization was examined by immunofluorescence. For the in vitro experiments, an enzyme-linked immunosorbent assay was utilized to measure VEGF secretion in the medium. In vitro experiments demonstrated that lipid peroxidation derivatives increased VEGF production in HaCaT cells, which was blocked by a p44/p42 MAPK inhibitor and anti-CD36 antibody. We observed increased levels of lipid peroxidation derivatives, p44/p42 MAPK activation and VEGF expression in the skin of KKA(y) obese mice. Notably, pitavastatin, an inhibitor of competitive 3-hydroxy-3-methylglutaryl coenzyme A reductase, suppressed all of these processes. Our results suggest that lipid peroxidation induces VEGF expression via CD36 and p44/p42 MAPK pathway in the skin of obese mice.
Collapse
Affiliation(s)
- Kozo Nakai
- Department of Dermatology, Faculty of Medicine, Kagawa University, Kita-gun, Kagawa, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Dubrac S, Schmuth M. PPAR-alpha in cutaneous inflammation. DERMATO-ENDOCRINOLOGY 2011; 3:23-6. [PMID: 21519405 PMCID: PMC3051849 DOI: 10.4161/derm.3.1.14615] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Accepted: 12/21/2010] [Indexed: 12/20/2022]
Abstract
Peroxisome proliferator-activated receptor (PPAR)-alpha is a fatty acid activated transcription factors that belongs to the nuclear hormone receptor family. Primarily PPAR-alpha serves as a lipid sensor. While PPAR-alpha controls enzymes from the lipid and glucose metabolism in the liver, heart and muscles, PPAR-alpha is also involved in skin homeostasis. PPAR-alpha controls keratinocyte proliferation/differentiation, contributes to wound healing and regulates skin inflammation. PPAR-alpha activation exerts anti-inflammatory effects in various skin conditions such as irritant and allergic contact dermatitis, atopic dermatitis and UV-induced erythema, rendering investigations into the functions of PPAR-alpha necessary to provide better understandings to treat many inflammatory skin disorders.
Collapse
Affiliation(s)
- Sandrine Dubrac
- Molecular Biology Laboratory; Department of Dermatology; Innsbruck Medical University; Innsbruck Austria
| | | |
Collapse
|
49
|
Barroso E, Eyre E, Palomer X, Vázquez-Carrera M. The peroxisome proliferator-activated receptor β/δ (PPARβ/δ) agonist GW501516 prevents TNF-α-induced NF-κB activation in human HaCaT cells by reducing p65 acetylation through AMPK and SIRT1. Biochem Pharmacol 2010; 81:534-43. [PMID: 21146504 DOI: 10.1016/j.bcp.2010.12.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 11/30/2010] [Accepted: 12/01/2010] [Indexed: 12/19/2022]
Abstract
Nuclear factor (NF)-κB is a ubiquitously expressed transcription factor controlling the expression of numerous genes involved in inflammation. The aim of this study was to evaluate whether activation of the peroxisome proliferator-activated receptor (PPAR) β/δ prevented TNF-α-induced NF-κB activation in human HaCaT keratinocytes and, if so, to determine the mechanism involved. The PPARβ/δ agonist GW501516 inhibited the increase caused by TNF-α in the mRNA levels of the NF-κB target genes interleukin 8 (IL-8), TNF-α and thymic stromal lymphopoietin (TSLP). Likewise, GW501516 prevented the increase in NF-κB DNA-binding activity observed in cells exposed to TNF-α. The reduction in NF-κB activity following GW501516 treatment in cells stimulated with TNF-α did not involve either increased IκBα protein levels or a reduction in the translocation of the p65 subunit of NF-κB. In contrast, GW501516 treatment decreased TNF-α-induced p65 acetylation. Acetylation of p65 is mainly regulated by p300, a transcriptional co-activator that binds to and acetylates p65. Of note, AMP kinase (AMPK) activation phosphorylates p300 and reduces its binding to p65. GW501516 increased AMPK phosphorylation and the subsequent p300 phosphorylation, leading to a marked reduction in the association between p65 and this transcriptional co-activator. In addition, treatment with the PPARβ/δ agonist increased SIRT1 protein levels. Finally, the reduction in IL-8 mRNA levels following GW501516 treatment in TNF-α-stimulated cells was abolished in the presence of the PPARβ/δ antagonist GSK0660, the AMPK inhibitor compound C and the SIRT1 inhibitor sirtinol, indicating that the effects of GW501516 on NF-κB activity were dependent on PPARβ/δ, AMPK and SIRT1, respectively.
Collapse
Affiliation(s)
- Emma Barroso
- Department of Pharmacology and Therapeutic Chemistry, Faculty of Pharmacy, University of Barcelona, Institut de Biomedicina de la UB (IBUB), Spain
| | | | | | | |
Collapse
|
50
|
McCusker MM, Grant-Kels JM. Healing fats of the skin: the structural and immunologic roles of the omega-6 and omega-3 fatty acids. Clin Dermatol 2010; 28:440-51. [PMID: 20620762 DOI: 10.1016/j.clindermatol.2010.03.020] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Linoleic acid (18:2omega6) and alpha-linolenic acid (18:3omega3) represent the parent fats of the two main classes of polyunsaturated fatty acids: the omega-6 (n-6) and the omega-3 (n-3) fatty acids, respectively. Linoleic acid and alpha-linolenic acid both give rise to other long-chain fatty acid derivatives, including gamma-linolenic acid and arachidonic acid (omega-6 fatty acids) and docosahexaenoic acid and eicosapentaenoic acid (omega-3 fatty acids). These fatty acids are showing promise as safe adjunctive treatments for many skin disorders, including atopic dermatitis, psoriasis, acne vulgaris, systemic lupus erythematosus, nonmelanoma skin cancer, and melanoma. Their roles are diverse and include maintenance of the stratum corneum permeability barrier, maturation and differentiation of the stratum corneum, formation and secretion of lamellar bodies, inhibition of proinflammatory eicosanoids, elevation of the sunburn threshold, inhibition of proinflammatory cytokines (tumor necrosis factor-alpha, interferon-gamma, and interleukin-12), inhibition of lipoxygenase, promotion of wound healing, and promotion of apoptosis in malignant cells, including melanoma. They fulfill these functions independently and through the modulation of peroxisome proliferator-activated receptors and Toll-like receptors.
Collapse
Affiliation(s)
- Meagen M McCusker
- Department of Dermatology University of Connecticut Health Center, 263 Farmington Avenue, MC 6230, Farmington, CT 06030, USA
| | | |
Collapse
|