1
|
Luo Y, Bollag WB. The Role of PGC-1α in Aging Skin Barrier Function. Cells 2024; 13:1135. [PMID: 38994987 PMCID: PMC11240425 DOI: 10.3390/cells13131135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/13/2024] Open
Abstract
Skin provides a physical and immune barrier to protect the body from foreign substances, microbial invasion, and desiccation. Aging reduces the barrier function of skin and its rate of repair. Aged skin exhibits decreased mitochondrial function and prolonged low-level inflammation that can be seen in other organs with aging. Peroxisome proliferator-activated receptor (PPAR)-γ coactivator-1α (PGC-1α), an important transcriptional coactivator, plays a central role in modulating mitochondrial function and antioxidant production. Mitochondrial function and inflammation have been linked to epidermal function, but the mechanisms are unclear. The aim of this review is to discuss the mechanisms by which PGC-1α might exert a positive effect on aged skin barrier function. Initially, we provide an overview of the function of skin under physiological and aging conditions, focusing on the epidermis. We then discuss mitochondrial function, oxidative stress, cellular senescence, and inflamm-aging, the chronic low-level inflammation observed in aging individuals. Finally, we discuss the effects of PGC-1α on mitochondrial function, as well as the regulation and role of PGC-1α in the aging epidermis.
Collapse
Affiliation(s)
- Yonghong Luo
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Wendy B. Bollag
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
- Department of Dermatology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
2
|
Yin X, Yan Y, Li J, Cao Z, Shen S, Chang Q, Zhao Y, Wang X, Wang P. Nuclear receptors for epidermal lipid barrier: Advances in mechanisms and applications. Exp Dermatol 2024; 33:e15107. [PMID: 38840418 DOI: 10.1111/exd.15107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 04/20/2024] [Accepted: 05/19/2024] [Indexed: 06/07/2024]
Abstract
The skin plays an essential role in preventing the entry of external environmental threats and the loss of internal substances, depending on the epidermal permeability barrier. Nuclear receptors (NRs), present in various tissues and organs including full-thickness skin, have been demonstrated to exert significant effects on the epidermal lipid barrier. Formation of the lipid lamellar membrane and the normal proliferation and differentiation of keratinocytes (KCs) are crucial for the development of the epidermal permeability barrier and is regulated by specific NRs such as PPAR, LXR, VDR, RAR/RXR, AHR, PXR and FXR. These receptors play a key role in regulating KC differentiation and the entire process of epidermal lipid synthesis, processing and secretion. Lipids derived from sebaceous glands are influenced by NRs as well and participate in regulation of the epidermal lipid barrier. Furthermore, intricate interplay exists between these receptors. Disturbance of barrier function leads to a range of diseases, including psoriasis, atopic dermatitis and acne. Targeting these NRs with agonists or antagonists modulate pathways involved in lipid synthesis and cell differentiation, suggesting potential therapeutic approaches for dermatosis associated with barrier damage. This review focuses on the regulatory role of NRs in the maintenance and processing of the epidermal lipid barrier through their effects on skin lipid synthesis and KC differentiation, providing novel insights for drug targets to facilitate precision medicine strategies.
Collapse
Affiliation(s)
- Xidie Yin
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yu Yan
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiandan Li
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhi Cao
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shuzhan Shen
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qihang Chang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yiting Zhao
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiuli Wang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Peiru Wang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
3
|
Zhu B, Zhu X, Borland MG, Ralph DH, Chiaro CR, Krausz KW, Ntambi JM, Glick AB, Patterson AD, Perdew GH, Gonzalez FJ, Peters JM. Activation of Peroxisome Proliferator-Activated Receptor-β/δ (PPARβ/δ) in Keratinocytes by Endogenous Fatty Acids. Biomolecules 2024; 14:606. [PMID: 38927010 PMCID: PMC11201440 DOI: 10.3390/biom14060606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 06/28/2024] Open
Abstract
Nuclear hormone receptors exist in dynamic equilibrium between transcriptionally active and inactive complexes dependent on interactions with ligands, proteins, and chromatin. The present studies examined the hypothesis that endogenous ligands activate peroxisome proliferator-activated receptor-β/δ (PPARβ/δ) in keratinocytes. The phorbol ester treatment or HRAS infection of primary keratinocytes increased fatty acids that were associated with enhanced PPARβ/δ activity. Fatty acids caused PPARβ/δ-dependent increases in chromatin occupancy and the expression of angiopoietin-like protein 4 (Angptl4) mRNA. Analyses demonstrated that stearoyl Co-A desaturase 1 (Scd1) mediates an increase in intracellular monounsaturated fatty acids in keratinocytes that act as PPARβ/δ ligands. The activation of PPARβ/δ with palmitoleic or oleic acid causes arrest at the G2/M phase of the cell cycle of HRAS-expressing keratinocytes that is not found in similarly treated HRAS-expressing Pparb/d-null keratinocytes. HRAS-expressing Scd1-null mouse keratinocytes exhibit enhanced cell proliferation, an effect that is mitigated by treatment with palmitoleic or oleic acid. Consistent with these findings, the ligand activation of PPARβ/δ with GW0742 or oleic acid prevented UVB-induced non-melanoma skin carcinogenesis, an effect that required PPARβ/δ. The results from these studies demonstrate that PPARβ/δ has endogenous roles in keratinocytes and can be activated by lipids found in diet and cellular components.
Collapse
Affiliation(s)
- Bokai Zhu
- Department of Veterinary and Biomedical Sciences, The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA; (B.Z.); (X.Z.); (M.G.B.); (D.H.R.); (C.R.C.); (A.B.G.); (A.D.P.); (G.H.P.)
| | - Xiaoyang Zhu
- Department of Veterinary and Biomedical Sciences, The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA; (B.Z.); (X.Z.); (M.G.B.); (D.H.R.); (C.R.C.); (A.B.G.); (A.D.P.); (G.H.P.)
| | - Michael G. Borland
- Department of Veterinary and Biomedical Sciences, The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA; (B.Z.); (X.Z.); (M.G.B.); (D.H.R.); (C.R.C.); (A.B.G.); (A.D.P.); (G.H.P.)
- Department of Biochemistry, Microbiology and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Douglas H. Ralph
- Department of Veterinary and Biomedical Sciences, The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA; (B.Z.); (X.Z.); (M.G.B.); (D.H.R.); (C.R.C.); (A.B.G.); (A.D.P.); (G.H.P.)
| | - Christopher R. Chiaro
- Department of Veterinary and Biomedical Sciences, The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA; (B.Z.); (X.Z.); (M.G.B.); (D.H.R.); (C.R.C.); (A.B.G.); (A.D.P.); (G.H.P.)
- Department of Genetics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Kristopher W. Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA; (K.W.K.); (F.J.G.)
| | - James M. Ntambi
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA;
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Adam B. Glick
- Department of Veterinary and Biomedical Sciences, The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA; (B.Z.); (X.Z.); (M.G.B.); (D.H.R.); (C.R.C.); (A.B.G.); (A.D.P.); (G.H.P.)
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences, The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA; (B.Z.); (X.Z.); (M.G.B.); (D.H.R.); (C.R.C.); (A.B.G.); (A.D.P.); (G.H.P.)
| | - Gary H. Perdew
- Department of Veterinary and Biomedical Sciences, The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA; (B.Z.); (X.Z.); (M.G.B.); (D.H.R.); (C.R.C.); (A.B.G.); (A.D.P.); (G.H.P.)
- Department of Biochemistry, Microbiology and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Genetics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA; (K.W.K.); (F.J.G.)
| | - Jeffrey M. Peters
- Department of Veterinary and Biomedical Sciences, The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA; (B.Z.); (X.Z.); (M.G.B.); (D.H.R.); (C.R.C.); (A.B.G.); (A.D.P.); (G.H.P.)
- Department of Biochemistry, Microbiology and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Genetics, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
4
|
Butera A, Agostini M, Cassandri M, De Nicola F, Fanciulli M, D’Ambrosio L, Falasca L, Nardacci R, Wang L, Piacentini M, Knight RA, Jia W, Sun Q, Shi Y, Wang Y, Candi E, Melino G. ZFP750 affects the cutaneous barrier through regulating lipid metabolism. SCIENCE ADVANCES 2023; 9:eadg5423. [PMID: 37115925 PMCID: PMC10146900 DOI: 10.1126/sciadv.adg5423] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
An essential function of the epidermis is to provide a physical barrier that prevents the loss of water. Essential mediators of this barrier function include ceramides, cholesterol, and very long chain fatty acids, and their alteration causes human pathologies, including psoriasis and atopic dermatitis. A frameshift mutation in the human ZNF750 gene, which encodes a zinc finger transcription factor, has been shown to cause a seborrhea-like dermatitis. Here, we show that genetic deletion of the mouse homolog ZFP750 results in loss of epidermal barrier function, which is associated with a substantial reduction of ceramides, nonpolar lipids. The alteration of epidermal lipid homeostasis is directly linked to the transcriptional activity of ZFP750. ZFP750 directly and/or indirectly regulates the expression of crucial enzymes primarily involved in the biosynthesis of ceramides. Overall, our study identifies the transcription factor ZFP750 as a master regulator epidermal homeostasis through lipid biosynthesis and thus contributing to our understanding of the pathogenesis of several human skin diseases.
Collapse
Affiliation(s)
- Alessio Butera
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Massimiliano Agostini
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Matteo Cassandri
- Department of Oncohematology, Bambino Gesù Children’s Hospital, 00146 Rome, Italy
| | - Francesca De Nicola
- Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maurizio Fanciulli
- Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Lorenzo D’Ambrosio
- Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Laura Falasca
- Laboratory of Electron Microscopy, National Institute for Infectious Diseases “L. Spallanzani,” IRCCS, Rome Italy
| | - Roberta Nardacci
- Laboratory of Electron Microscopy, National Institute for Infectious Diseases “L. Spallanzani,” IRCCS, Rome Italy
- Departmental Faculty of Medicine and Surgery, Saint Camillus International University of Health Sciences (UniCamillus), Rome, Italy
| | - Lu Wang
- University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Mauro Piacentini
- Laboratory of Electron Microscopy, National Institute for Infectious Diseases “L. Spallanzani,” IRCCS, Rome Italy
| | - Richard A. Knight
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Wei Jia
- University of Hawaii Cancer Center, Honolulu, HI 96813, USA
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Qiang Sun
- Laboratory of Cell Engineering, Institute of Biotechnology, Research Unit of Cell Death Mechanism, 2021RU008, Chinese Academy of Medical Science, 20 Dongda Street, Beijing, 100071, China
| | - Yufang Shi
- The Third Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou 215123, China
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences/Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy
- IDI-IRCCS, via Monti di Creta, 106, 00166 Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy
- Corresponding author.
| |
Collapse
|
5
|
Horse-Derived Ceramide Accentuates Glucosylceramide Synthase and Ceramide Synthase 3 by Activating PPARβ/δ and/or PPARγ to Stimulate Ceramide Synthesis. Biomedicines 2023; 11:biomedicines11020548. [PMID: 36831084 PMCID: PMC9953238 DOI: 10.3390/biomedicines11020548] [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: 12/05/2022] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Horse-derived ceramide (HC), which contains galactosylceramides as its main component, significantly improves skin symptoms when applied topically to patients with atopic dermatitis. We speculated that efficacy resulted from the amelioration of epidermal ceramide metabolism, and we characterized those effects using reconstructed human epidermal equivalents. Lipid analysis, RT-PCR and Western blotting revealed that HC significantly increased the total ceramide content of the stratum corneum (SC), accompanied by significantly increased gene and/or protein expression levels of ceramide synthase (CERS) 3, fatty acid elongase (ELOVL) 4, glucosylceramide synthase (GCS), β-glucocerebrosidase, sphingomyelin synthase and acid sphingomyelinase. Mechanistic analyses using cultures of primary human keratinocytes revealed the marked stimulatory effects of HC on the mRNA expression levels of CERS3, ELOVL4 and GCS under high calcium-derived differentiation conditions. Signaling analyses demonstrated that an antagonist of PPARβ/δ significantly abrogated the HC-stimulated mRNA expression levels of GCS, CERS3 and ELOVL4. GW9662, an antagonist of PPARγ, significantly abolished the HC-up-regulated mRNA expression levels of GCS and ELOVL4, but not of CERS3. These findings suggest that HC has the distinct potential to accentuate the expression of GCS, CERS3 and ELOVL4 via the activation of PPARβ/δ and/or PPARγ to accelerate ceramide synthesis in the SC.
Collapse
|
6
|
Murashkin NN, Opryatin LA, Epishev RV, Materikin AI, Ambarchian ET, Ivanov RA, Savelova AA, Nezhvedilova RY, Rusakova LL. Filaggrin Defect at Atopic Dermatitis: Modern Treatment Options. CURRENT PEDIATRICS 2022. [DOI: 10.15690/vsp.v21i5.2452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Atopic dermatitis is a common chronic skin disease, its pathogenesis is associated with congenital or acquired deficiency of filaggrin protein. In recent years, extensive evidence on the causes of filaggrin deficiency has been obtained. The structure and functions of this protein are described, that opens new approaches for atopic dermatitis management.
Collapse
Affiliation(s)
- Nikolay N. Murashkin
- National Medical Research Center of Children’s Health; Sechenov First Moscow State Medical University; Central State Medical Academy of Department of Presidential Affairs
| | | | | | | | - Eduard T. Ambarchian
- Pediatrics and Child Health Research Institute in Petrovsky National Research Centre of Surgery
| | | | | | | | | |
Collapse
|
7
|
Sun Z, Guo Y, Zhang D, Zhang G, Zhang Y, Wang X. FABP7 inhibits proliferation and invasion abilities of cutaneous squamous cell carcinoma cells via the Notch signaling pathway. Oncol Lett 2022; 24:254. [PMID: 35765272 PMCID: PMC9219017 DOI: 10.3892/ol.2022.13374] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 05/24/2021] [Indexed: 11/15/2022] Open
Abstract
Cutaneous squamous cell carcinoma (CSCC) is one of the most common non-melanoma skin cancers worldwide. Fatty acid-binding protein 7 (FABP7) has been reported to be involved in the occurrence, development, metastasis and prognosis of various tumors. In addition, downregulated FABP7 expression was demonstrated in cutaneous malignant melanoma in a previous study. Therefore, we speculated that FABP7 may be a biomarker for CSCC diagnosis. The aim of the present study was to determine the molecular mechanism underlying the effects of FABP7 in CSCC, which may provide a new diagnostic biomarker or treatment target for CSCC. Reverse transcription-PCR, western blotting and immunohistochemistry assays were performed to detect the expression levels of FABP7 in CSCC tissues and cells. Overexpression of FABP7 was achieved in A431 and colo-16 cell lines by transfection with an overexpression vector (oeFABP7). Cell proliferation, colony formation, migration and invasion were detected by Cell Counting Kit-8, crystal violet, scratch and Transwell assays, respectively. Following FABP7 overexpression, western blotting was used to determine the expression levels of proliferation-, invasion- and Notch pathway-associated proteins, including Snail, N-cadherin, Twist, matrix metalloproteinase (MMP)-2, MMP-7, Notch 1 and Notch 3. In addition a CSCC model in nude mice was constructed. Immunohistochemistry was used to determine the expression levels of FABP7, Ki67, Notch 1 and Notch 3. It was demonstrated that FABP7 expression levels were significantly reduced in human CSCC tissues and cells compared with normal samples. Overexpression of FABP7 inhibited the proliferation, invasion and migration abilities of A431 and colo-16 cells compared with those in the negative control group. In addition, transfection with oeFABP7 reduced the expression levels of proliferation-, invasion- and Notch pathway-associated proteins compared with those in the negative control group. Overexpression of FABP7 also reduced the growth of CSCC tumors in vivo and inhibited the expression of Ki67, Notch 1 and Notch 3. Therefore, the results of the present study suggested that FABP7 may inhibit the proliferation and invasion of CSCC cells via the Notch signaling pathway.
Collapse
Affiliation(s)
- Zhonghui Sun
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200050, P.R. China.,Department of Dermatology, Fengxian Institute of Dermatosis Prevention and Treatment, Shanghai 201408, P.R. China
| | - Yunyi Guo
- Department of Dermatology, Fengxian Institute of Dermatosis Prevention and Treatment, Shanghai 201408, P.R. China
| | - Danlu Zhang
- Department of Dermatology, Fengxian Institute of Dermatosis Prevention and Treatment, Shanghai 201408, P.R. China
| | - Guolong Zhang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200050, P.R. China
| | - Ying Zhang
- Department of Dermatology, Fengxian Institute of Dermatosis Prevention and Treatment, Shanghai 201408, P.R. China
| | - Xiuli Wang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200050, P.R. China
| |
Collapse
|
8
|
PPARβ/δ Augments IL-1β-Induced COX-2 Expression and PGE2 Biosynthesis in Human Mesangial Cells via the Activation of SIRT1. Metabolites 2022; 12:metabo12070595. [PMID: 35888719 PMCID: PMC9320509 DOI: 10.3390/metabo12070595] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/11/2022] [Accepted: 06/22/2022] [Indexed: 12/10/2022] Open
Abstract
Peroxisome proliferator-activated receptor β/δ (PPARβ/δ), a ligand-activated nuclear receptor, regulates lipid and glucose metabolism and inflammation. PPARβ/δ can exert an anti-inflammatory effect by suppressing proinflammatory cytokine production. Cyclooxygenase-2 (COX-2)-triggered inflammation plays a crucial role in the development of many inflammatory diseases, including glomerulonephritis. However, the effect of PPARβ/δ on the expression of COX-2 in the kidney has not been fully elucidated. The present study showed that PPARβ/δ was functionally expressed in human mesangial cells (hMCs), where its expression was increased by interleukin-1β (IL-1β) treatment concomitant with enhanced COX-2 expression and prostaglandin E2 (PGE2) biosynthesis. The treatment of hMCs with GW0742, a selective agonist of PPARβ/δ, or the overexpression of PPARβ/δ via an adenovirus-mediated approach significantly increased COX-2 expression and PGE2 production. PPARβ/δ could further augment the IL-1β-induced COX-2 expression and PGE2 production in hMCs. Moreover, both PPARβ/δ activation and overexpression markedly increased sirtuin 1 (SIRT1) expression. The inhibition or knockdown of SIRT1 significantly attenuated the effects of PPARβ/δ on the IL-1β-induced expression of COX-2 and PGE2 biosynthesis. Taken together, PPARβ/δ could augment the IL-1β-induced COX-2 expression and PGE2 production in hMCs via the SIRT1 pathway. Given the critical role of COX-2 in glomerulonephritis, PPARβ/δ may represent a novel target for the treatment of renal inflammatory diseases.
Collapse
|
9
|
Escandon P, Vasini B, Whelchel AE, Nicholas SE, Matlock HG, Ma JX, Karamichos D. The role of peroxisome proliferator-activated receptors in healthy and diseased eyes. Exp Eye Res 2021; 208:108617. [PMID: 34010603 PMCID: PMC8594540 DOI: 10.1016/j.exer.2021.108617] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/23/2022]
Abstract
Peroxisome Proliferator-Activated Receptors (PPARs) are a family of nuclear receptors that play essential roles in modulating cell differentiation, inflammation, and metabolism. Three subtypes of PPARs are known: PPAR-alpha (PPARα), PPAR-gamma (PPARγ), and PPAR-beta/delta (PPARβ/δ). PPARα activation reduces lipid levels and regulates energy homeostasis, activation of PPARγ results in regulation of adipogenesis, and PPARβ/δ activation increases fatty acid metabolism and lipolysis. PPARs are linked to various diseases, including but not limited to diabetes, non-alcoholic fatty liver disease, glaucoma and atherosclerosis. In the past decade, numerous studies have assessed the functional properties of PPARs in the eye and key PPAR mechanisms have been discovered, particularly regarding the retina and cornea. PPARγ and PPARα are well established in their functions in ocular homeostasis regarding neuroprotection, neovascularization, and inflammation, whereas PPARβ/δ isoform function remains understudied. Naturally, studies on PPAR agonists and antagonists, associated with ocular pathology, have also gained traction with the development of PPAR synthetic ligands. Studies on PPARs has significantly influenced novel therapeutics for diabetic eye disease, ocular neuropathy, dry eye, and age-related macular degeneration (AMD). In this review, therapeutic potentials and implications will be highlighted, as well as reported adverse effects. Further investigations are necessary before any of the PPARs ligands can be utilized, in the clinics, to treat eye diseases. Future research on the prominent role of PPARs will help unravel the complex mechanisms involved in order to prevent and treat ocular diseases.
Collapse
Affiliation(s)
- Paulina Escandon
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Brenda Vasini
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Amy E Whelchel
- Department of Physiology, University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA
| | - Sarah E Nicholas
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - H Greg Matlock
- Department of Physiology, University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA
| | - Jian-Xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA; Harold Hamm Oklahoma Diabetes Center, 1000 N Lincoln Blvd, Oklahoma City, OK, USA
| | - Dimitrios Karamichos
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA.
| |
Collapse
|
10
|
Peters JM, Walter V, Patterson AD, Gonzalez FJ. Unraveling the role of peroxisome proliferator-activated receptor-β/δ (PPARβ/δ) expression in colon carcinogenesis. NPJ Precis Oncol 2019; 3:26. [PMID: 31602402 PMCID: PMC6779880 DOI: 10.1038/s41698-019-0098-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/17/2019] [Indexed: 01/09/2023] Open
Abstract
The peroxisome proliferator-activated-β/δ (PPARβ/δ) was identified in 1994, but not until 1999 was PPARβ/δ suggested to be involved in carcinogenesis. Initially, it was hypothesized that expression of PPARβ/δ was increased during colon cancer progression, which led to increased transcription of yet-to-be confirmed target genes that promote cell proliferation and tumorigenesis. It was also hypothesized at this time that lipid-metabolizing enzymes generated lipid metabolites that served as ligands for PPARβ/δ. These hypothetical mechanisms were attractive because they potentially explained how non-steroidal anti-inflammatory drugs inhibited tumorigenesis by potentially limiting the concentration of endogenous PPARβ/δ ligands that could activate this receptor that was increased in cancer cells. However, during the last 20 years, considerable research was undertaken describing expression of PPARβ/δ in normal and cancer cells that has led to a significant impact on the mechanisms by which PPARβ/δ functions in carcinogenesis. Whereas results from earlier studies led to much uncertainty about the role of PPARβ/δ in cancer, more recent analyses of large databases have revealed a more consistent understanding. The focus of this review is on the fundamental level of PPARβ/δ expression in normal tissues and cancerous tissue as described by studies during the past two decades and what has been delineated during this timeframe about how PPARβ/δ expression influences carcinogenesis, with an emphasis on colon cancer.
Collapse
Affiliation(s)
- Jeffrey M. Peters
- Department of Veterinary and Biomedical Sciences, The Center of Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, State College, PA 16801 USA
| | - Vonn Walter
- Departments of Public Health Sciences and Biochemistry, The Pennsylvania State University, College of Medicine, Hershey, State College, PA 16801 USA
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences, The Center of Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, State College, PA 16801 USA
| | - Frank J. Gonzalez
- Laboratory of Metabolism, National Cancer Institute, Bethesda, MD USA
| |
Collapse
|
11
|
Peters JM, Kim DJ, Bility MT, Borland MG, Zhu B, Gonzalez FJ. Regulatory mechanisms mediated by peroxisome proliferator-activated receptor-β/δ in skin cancer. Mol Carcinog 2019; 58:1612-1622. [PMID: 31062422 DOI: 10.1002/mc.23033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 04/12/2019] [Accepted: 04/15/2019] [Indexed: 12/21/2022]
Abstract
Considerable progress has been made during the past 20 years towards elucidating the role of peroxisome proliferator-activated receptor-β/δ (PPARβ/δ) in skin cancer. In 1999, the original notion that PPARβ/δ was involved with epithelial cell function was postulated based on a correlation between PPARβ/δ expression and the induction of messenger RNAs encoding proteins that mediate terminal differentiation in keratinocytes. Subsequent studies definitively revealed that PPARβ/δ could induce terminal differentiation and inhibit proliferation of keratinocytes. Molecular mechanisms have since been discovered to explain how this nuclear receptor can be targeted for preventing and treating skin cancer. This includes the regulation of terminal differentiation, mitotic signaling, endoplasmic reticulum stress, and cellular senescence. Interestingly, the effects of activating PPARβ/δ can preferentially target keratinocytes with genetic mutations associated with skin cancer. This review provides the history and current understanding of how PPARβ/δ can be targeted for both nonmelanoma skin cancer and melanoma and postulates how future approaches that modulate PPARβ/δ signaling may be developed for the prevention and treatment of these diseases.
Collapse
Affiliation(s)
- Jeffrey M Peters
- Department of Veterinary and Biomedical Sciences, The Center of Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania
| | - Dae J Kim
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas
| | - Moses T Bility
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael G Borland
- Department of Chemistry & Biochemistry, Bloomsburg University of Pennsylvania, Bloomsburg, Pennsylvania
| | - Bokai Zhu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
12
|
Muresan XM, Narzt MS, Woodby B, Ferrara F, Gruber F, Valacchi G. Involvement of cutaneous SR-B1 in skin lipid homeostasis. Arch Biochem Biophys 2019; 666:1-7. [DOI: 10.1016/j.abb.2019.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/06/2019] [Accepted: 03/09/2019] [Indexed: 12/16/2022]
|
13
|
Aster glehni Extract Containing Caffeoylquinic Compounds Protects Human Keratinocytes through the TRPV4-PPAR δ-AMPK Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2018:9616574. [PMID: 30622619 PMCID: PMC6304624 DOI: 10.1155/2018/9616574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/21/2018] [Accepted: 11/12/2018] [Indexed: 11/17/2022]
Abstract
Aster glehni (AG) has been used in cooking and as a medicine to treat various diseases for over hundreds of years in Korea. To speculate the protective effects of AG on skin barrier, we estimated the protein levels of biomarkers related to skin barrier protection in human keratinocytes, HaCaT cells treated with sodium dodecyl sulfate (SDS), or 2,4-dinitrochlorobenzene (DNCB). The protein levels for keratin, involucrin, defensin, tumor necrosis factor alpha (TNFα), peroxisome proliferator-activated receptor delta (PPARδ), 5′ adenosine monophosphate-activated protein kinase (AMPK), serine palmitoyltransferase long chain base subunit 2 (SPTLC2), and transient receptor potential cation channel subfamily V member 4 (TRPV4) were evaluated using western blotting or immunocytochemistry in HaCaT cells. AG extract increased the protein levels of PPARδ, phosphorylated AMPK, SPTLC2, keratin, involucrin, and defensin compared to the SDS or DNCB control group. However, TNFα expression increased by SDS or DNCB was decreased with AG extract. The order of action of each regulatory biomarker in AG pathway was identified TRPV4→PPARδ→AMPK from antagonist and siRNA treatment studies. AG can ameliorate the injury of keratinocytes caused by SDS or DNCB through the sequential regulation of TRPV4→PPARδ→AMPK pathway.
Collapse
|
14
|
Local Burn Injury Promotes Defects in the Epidermal Lipid and Antimicrobial Peptide Barriers in Human Autograft Skin and Burn Margin: Implications for Burn Wound Healing and Graft Survival. J Burn Care Res 2018; 38:e212-e226. [PMID: 27183442 DOI: 10.1097/bcr.0000000000000357] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Burn injury increases the risk of morbidity and mortality by promoting severe hemodynamic shock and risk for local or systemic infection. Graft failure due to poor wound healing or infection remains a significant problem for burn subjects. The mechanisms by which local burn injury compromises the epithelial antimicrobial barrier function in the burn margin, containing the elements necessary for healing of the burn site, and in distal unburned skin, which serves as potential donor tissue, are largely unknown. The objective of this study was to establish defects in epidermal barrier function in human donor skin and burn margin, to identify potential mechanisms that may lead to graft failure and/or impaired burn wound healing. In this study, we established that epidermal lipids and respective lipid synthesis enzymes were significantly reduced in both donor skin and burn margin. We further identified diverse changes in the gene expression and protein production of several candidate skin antimicrobial peptides (AMPs) in both donor skin and burn margin. These results also parallel changes in cutaneous AMP activity against common burn wound pathogens, aberrant production of epidermal proteases known to regulate barrier permeability and AMP activity, and greater production of proinflammatory cytokines known to be induced by AMPs. These findings suggest that impaired epidermal lipid and AMP regulation could contribute to graft failure and infectious complications in subjects with burn or other traumatic injury.
Collapse
|
15
|
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
|
16
|
Man MQ, Sun R, Man G, Lee D, Hill Z, Elias PM. Commonly Employed African Neonatal Skin Care Products Compromise Epidermal Function in Mice. Pediatr Dermatol 2016; 33:493-500. [PMID: 27396436 DOI: 10.1111/pde.12901] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Neonatal mortality is much higher in the developing world than in developed countries. Infections are a major cause of neonatal death, particularly in preterm infants, in whom defective epidermal permeability barrier function facilitates transcutaneous pathogen invasion. The objective was to determine whether neonatal skin care products commonly used in Africa benefit or compromise epidermal functions in murine skin. METHODS After twice-daily treatment of 6- to 8-week-old hairless mice with each skin care product for 3 days, epidermal permeability barrier function, skin surface pH, stratum corneum hydration, and barrier recovery were measured using a multiprobe adapter system physiology monitor. For products showing some benefits in these initial tests, the epidermal permeability barrier homeostasis was assessed 1 and 5 hours after a single application to acutely disrupted skin. RESULTS All of the skin care products compromised basal permeability barrier function and barrier repair kinetics. Moreover, after 3 days of treatment, most of the products also reduced stratum corneum hydration while elevating skin surface pH to abnormal levels. CONCLUSION Some neonatal skin care products that are widely used in Africa perturb important epidermal functions, including permeability barrier homeostasis in mice. Should these products have similar effects on newborn human skin, they could cause a defective epidermal permeability barrier, which can increase body fluid loss, impair thermoregulation, and contribute to the high rates of neonatal morbidity and mortality seen in Africa. Accordingly, alternative products that enhance permeability barrier function should be identified, particularly for use in preterm infants.
Collapse
Affiliation(s)
- Mao-Qiang Man
- Dermatology Service, Veterans Affairs Medical Center, San Francisco, California.
- Department of Dermatology, University of California, San Francisco, San Francisco, California.
| | - Richard Sun
- Dermatology Service, Veterans Affairs Medical Center, San Francisco, California
- Department of Dermatology, University of California, San Francisco, San Francisco, California
| | - George Man
- Dermatology Service, Veterans Affairs Medical Center, San Francisco, California
- Department of Dermatology, University of California, San Francisco, San Francisco, California
| | - Dale Lee
- Dermatology Service, Veterans Affairs Medical Center, San Francisco, California
- Department of Dermatology, University of California, San Francisco, San Francisco, California
| | - Zelee Hill
- Institute for Global Health, Faculty of Population Health Sciences, University College London, London, UK
| | - Peter M Elias
- Dermatology Service, Veterans Affairs Medical Center, San Francisco, California
- Department of Dermatology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
17
|
Degueurce G, D'Errico I, Pich C, Ibberson M, Schütz F, Montagner A, Sgandurra M, Mury L, Jafari P, Boda A, Meunier J, Rezzonico R, Brembilla NC, Hohl D, Kolios A, Hofbauer G, Xenarios I, Michalik L. Identification of a novel PPARβ/δ/miR-21-3p axis in UV-induced skin inflammation. EMBO Mol Med 2016; 8:919-36. [PMID: 27250636 PMCID: PMC4967944 DOI: 10.15252/emmm.201505384] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Although excessive exposure to UV is widely recognized as a major factor leading to skin perturbations and cancer, the complex mechanisms underlying inflammatory skin disorders resulting from UV exposure remain incompletely characterized. The nuclear hormone receptor PPARβ/δ is known to control mouse cutaneous repair and UV-induced skin cancer development. Here, we describe a novel PPARβ/δ-dependent molecular cascade involving TGFβ1 and miR-21-3p, which is activated in the epidermis in response to UV exposure. We establish that the passenger miRNA miR-21-3p, that we identify as a novel UV-induced miRNA in the epidermis, plays a pro-inflammatory function in keratinocytes and that its high level of expression in human skin is associated with psoriasis and squamous cell carcinomas. Finally, we provide evidence that inhibition of miR-21-3p reduces UV-induced cutaneous inflammation in ex vivo human skin biopsies, thereby underlining the clinical relevance of miRNA-based topical therapies for cutaneous disorders.
Collapse
Affiliation(s)
- Gwendoline Degueurce
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Ilenia D'Errico
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Christine Pich
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Mark Ibberson
- SIB Swiss Institute of Bioinformatics University of Lausanne, Lausanne, Switzerland
| | - Frédéric Schütz
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland SIB Swiss Institute of Bioinformatics University of Lausanne, Lausanne, Switzerland
| | - Alexandra Montagner
- INRA ToxAlim, Integrative Toxicology and Metabolism, UMR1331, Toulouse, France
| | - Marie Sgandurra
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Lionel Mury
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Paris Jafari
- Department of Musculoskeletal Medicine, Service of Plastic and Reconstructive Surgery CHUV, Epalinges, Switzerland
| | - Akash Boda
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Julien Meunier
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Roger Rezzonico
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, UMR 7275, Valbonne, France
| | - Nicolò Costantino Brembilla
- Dermatology, University Hospital and School of Medicine, Geneva, Switzerland Immunology and Allergy, University Hospital and School of Medicine, Geneva Switzerland
| | - Daniel Hohl
- Service de dermatologie et venereology, Hôpital de Beaumont CHUV, Lausanne, Switzerland
| | - Antonios Kolios
- Department of Immunology, University Hospital, University of Zürich, Zürich, Switzerland Department of Dermatology, University Hospital, University of Zürich, Zürich, Switzerland
| | - Günther Hofbauer
- Department of Dermatology, University Hospital, University of Zürich, Zürich, Switzerland
| | - Ioannis Xenarios
- SIB Swiss Institute of Bioinformatics University of Lausanne, Lausanne, Switzerland
| | - Liliane Michalik
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
18
|
Yasuda T, Fukada T, Nishida K, Nakayama M, Matsuda M, Miura I, Dainichi T, Fukuda S, Kabashima K, Nakaoka S, Bin BH, Kubo M, Ohno H, Hasegawa T, Ohara O, Koseki H, Wakana S, Yoshida H. Hyperactivation of JAK1 tyrosine kinase induces stepwise, progressive pruritic dermatitis. J Clin Invest 2016; 126:2064-76. [PMID: 27111231 DOI: 10.1172/jci82887] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 03/03/2016] [Indexed: 01/12/2023] Open
Abstract
Skin homeostasis is maintained by the continuous proliferation and differentiation of epidermal cells. The skin forms a strong but flexible barrier against microorganisms as well as physical and chemical insults; however, the physiological mechanisms that maintain this barrier are not fully understood. Here, we have described a mutant mouse that spontaneously develops pruritic dermatitis as the result of an initial defect in skin homeostasis that is followed by induction of a Th2-biased immune response. These mice harbor a mutation that results in a single aa substitution in the JAK1 tyrosine kinase that results in hyperactivation, thereby leading to skin serine protease overexpression and disruption of skin barrier function. Accordingly, treatment with an ointment to maintain normal skin barrier function protected mutant mice from dermatitis onset. Pharmacological inhibition of JAK1 also delayed disease onset. Together, these findings indicate that JAK1-mediated signaling cascades in skin regulate the expression of proteases associated with the maintenance of skin barrier function and demonstrate that perturbation of these pathways can lead to the development of spontaneous pruritic dermatitis.
Collapse
|
19
|
The role of barrier genes in epidermal malignancy. Oncogene 2016; 35:5705-5712. [PMID: 27041586 DOI: 10.1038/onc.2016.84] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 12/24/2022]
Abstract
The outermost layer of the mammalian skin, the epidermis, forms a protective barrier against pathogenic microbes and tissue dehydration. This barrier is formed and maintained by complex genetic networks that connect cellular differentiation processes, enzymatic activities and cellular junctions. Disruption in these networks affects the balance between keratinocyte proliferation and differentiation resulting in barrier function impairment, epidermal hyperproliferation and in some cases, squamous cell carcinoma (SCC). Recent studies in wound-induced inflammation-mediated cancers in mice have identified dysregulation of core barrier components as tumor drivers. We therefore propose a hypothesis in which loss of key barrier genes, induce barrier dysfunction, and promote inflammation-driven epidermal hyperplasia and carcinogenesis over time. This emerging vision suggests that under specific genetic circumstances, localized barrier impairment could be considered as a hallmark of initiating lesions in epidermal SCC.
Collapse
|
20
|
Chen WM, Shaw LH, Chang PJ, Tung SY, Chang TS, Shen CH, Hsieh YY, Wei KL. Hepatoprotective effect of resveratrol against ethanol-induced oxidative stress through induction of superoxide dismutase in vivo and in vitro. Exp Ther Med 2016; 11:1231-1238. [PMID: 27073428 PMCID: PMC4812565 DOI: 10.3892/etm.2016.3077] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 12/01/2015] [Indexed: 12/14/2022] Open
Abstract
The present study aimed to investigate the hepatoprotective effect of resveratrol (RSV) against ethanol-induced oxidative stress in vivo, and investigate the underlying mechanisms by which RSV exerts its anti-oxidative effects on hepatic cells. C57BL/6J mice were divided into four groups: Untreated control, ethanol-treated, RSV-treated, and ethanol + RSV-treated. The plasma lipid profile, hepatic lipid accumulation and antioxidative enzyme activities were analyzed. HepG2 cells were used as a cellular model to analyze the effects of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) and peroxisome proliferator-activated receptors (PPARs) in the RSV-mediated protection of ethanol-induced oxidative stress. In C57BL/6J mice, ethanol caused a significant increase in plasma triglyceride levels and hepatic lipid accumulation (P<0.05), whereas RSV notably increased SOD activity. In HepG2 cells, SOD activity was enhanced in the RSV-treated HepG2 cells, whereas the activity of CAT and GPx was not affected. Western blot and quantitative polymerase chain reaction analyses demonstrated that RSV significantly increased SOD protein and mRNA expression levels (P<0.05). Using a transient transfection assay, PPARγ was observed to participate in the regulation of SOD gene expression in RSV-administered HepG2 cells. To conclude, the results from the present study suggest that RSV may contribute towards the protection of hepatic cells from ethanol-induced oxidative stress via the induction of SOD activity and gene expression.
Collapse
Affiliation(s)
- Wei-Ming Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Puzi, Chiayi 61363, Taiwan, R.O.C.; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, R.O.C.; College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, R.O.C
| | - Lee-Hsin Shaw
- Institute of Traditional Medicine, National Yang-Ming University, Taipei 11221, Taiwan, R.O.C
| | - Pey-Jium Chang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, R.O.C
| | - Shui-Yi Tung
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Puzi, Chiayi 61363, Taiwan, R.O.C.; College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, R.O.C
| | - Te-Sheng Chang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Puzi, Chiayi 61363, Taiwan, R.O.C.; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, R.O.C
| | - Chein-Heng Shen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Puzi, Chiayi 61363, Taiwan, R.O.C.; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, R.O.C
| | - Yung-Yu Hsieh
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Puzi, Chiayi 61363, Taiwan, R.O.C.; College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, R.O.C
| | - Kuo-Liang Wei
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Puzi, Chiayi 61363, Taiwan, R.O.C.; College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, R.O.C
| |
Collapse
|
21
|
Gupta M, Mahajan VK, Mehta KS, Chauhan PS, Rawat R. Peroxisome proliferator-activated receptors (PPARs) and PPAR agonists: the 'future' in dermatology therapeutics? Arch Dermatol Res 2015; 307:767-780. [PMID: 25986745 DOI: 10.1007/s00403-015-1571-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/11/2015] [Accepted: 05/05/2015] [Indexed: 01/10/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear hormone receptors and comprise three different isoforms namely PPARα, PPARγ, and PPARβ/δ with PPARβ/δ being the predominant subtype in human keratinocytes. After binding with specific ligands, PPARs regulate gene expression, cell growth and differentiation, apoptosis, inflammatory responses, and tumorogenesis. PPARs also modulate a wide variety of skin functions including keratinocyte proliferation, epidermal barrier formation, wound healing, melanocyte proliferation, and sebum production. Recent studies have shown the importance of PPARs in the pathogenesis of many dermatological disorders. Clinical trials have suggested possible role of PPAR agonists in the management of various dermatoses ranging from acne vulgaris, psoriasis, hirsutism, and lipodystrophy to cutaneous malignancies including melanoma. This article is intended to be a primer for dermatologists in their understanding of clinical relevance of PPARs and PPAR agonists in dermatology therapeutics.
Collapse
Affiliation(s)
- Mrinal Gupta
- Department of Dermatology, Venereology and Leprosy, Dr. R. P. Govt. Medical College, Kangra, Tanda, 176001, Himachal Pradesh, India
| | - Vikram K Mahajan
- Department of Dermatology, Venereology and Leprosy, Dr. R. P. Govt. Medical College, Kangra, Tanda, 176001, Himachal Pradesh, India.
| | - Karaninder S Mehta
- Department of Dermatology, Venereology and Leprosy, Dr. R. P. Govt. Medical College, Kangra, Tanda, 176001, Himachal Pradesh, India
| | - Pushpinder S Chauhan
- Department of Dermatology, Venereology and Leprosy, Dr. R. P. Govt. Medical College, Kangra, Tanda, 176001, Himachal Pradesh, India
| | - Ritu Rawat
- Department of Dermatology, Venereology and Leprosy, Dr. R. P. Govt. Medical College, Kangra, Tanda, 176001, Himachal Pradesh, India
| |
Collapse
|
22
|
|
23
|
Man G, Cheung C, Crumrine D, Hupe M, Hill Z, Man MQ, Elias PM. An optimized inexpensive emollient mixture improves barrier repair in murine skin. DERMATOL SIN 2015. [DOI: 10.1016/j.dsi.2015.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
24
|
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: 124] [Impact Index Per Article: 11.3] [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
|
25
|
Borland MG, Krishnan P, Lee C, Albrecht PP, Shan W, Bility MT, Marcus CB, Lin JM, Amin S, Gonzalez FJ, Perdew GH, Peters JM. Modulation of aryl hydrocarbon receptor (AHR)-dependent signaling by peroxisome proliferator-activated receptor β/δ (PPARβ/δ) in keratinocytes. Carcinogenesis 2014; 35:1602-12. [PMID: 24639079 PMCID: PMC4076811 DOI: 10.1093/carcin/bgu067] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 02/26/2014] [Accepted: 03/10/2014] [Indexed: 12/11/2022] Open
Abstract
Whether peroxisome proliferator-activated receptor β/δ (PPARβ/δ) reduces skin tumorigenesis by altering aryl hydrocarbon receptor (AHR)-dependent activities was examined. Polycyclic aromatic hydrocarbons (PAH) increased expression of cytochrome P4501A1 (CYP1A1), CYP1B1 and phase II xenobiotic metabolizing enzymes in wild-type skin and keratinocytes. Surprisingly, this effect was not found in Pparβ/δ-null skin and keratinocytes. Pparβ/δ-null keratinocytes exhibited decreased AHR occupancy and histone acetylation on the Cyp1a1 promoter in response to a PAH compared with wild-type keratinocytes. Bisulfite sequencing of the Cyp1a1 promoter and studies using a DNA methylation inhibitor suggest that PPARβ/δ promotes demethylation of the Cyp1a1 promoter. Experiments with human HaCaT keratinocytes stably expressing shRNA against PPARβ/δ also support this conclusion. Consistent with the lower AHR-dependent activities in Pparβ/δ-null mice compared with wild-type mice, 7,12-dimethylbenz[a]anthracene (DMBA)-induced skin tumorigenesis was inhibited in Pparβ/δ-null mice compared with wild-type. Results from these studies demonstrate that PPARβ/δ is required to mediate complete carcinogenesis by DMBA. The mechanisms underlying this PPARβ/δ-dependent reduction of AHR signaling by PAH are not due to alterations in the expression of AHR auxiliary proteins, ligand binding or AHR nuclear translocation between genotypes, but are likely influenced by PPARβ/δ-dependent demethylation of AHR target gene promoters including Cyp1a1 that reduces AHR accessibility as shown by reduced promoter occupancy. This PPARβ/δ/AHR crosstalk is unique to keratinocytes and conserved between mice and humans.
Collapse
MESH Headings
- 9,10-Dimethyl-1,2-benzanthracene/toxicity
- Animals
- Basic Helix-Loop-Helix Transcription Factors/physiology
- Blotting, Western
- Carcinogens/toxicity
- Cell Transformation, Neoplastic/chemically induced
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Cells, Cultured
- Chromatin Immunoprecipitation
- Dermis/cytology
- Dermis/metabolism
- Female
- Fibroblasts/cytology
- Fibroblasts/metabolism
- Humans
- Immunoenzyme Techniques
- Keratinocytes/cytology
- Keratinocytes/metabolism
- Mice
- Mice, Knockout
- PPAR delta/physiology
- PPAR-beta/physiology
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Receptors, Aryl Hydrocarbon/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Skin Neoplasms/chemically induced
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
Collapse
Affiliation(s)
- Michael G Borland
- Department of Veterinary and Biomedical Sciences and the Center of Molecular Toxicology and Carcinogenesis and The Graduate Program in Biochemistry, Microbiology, and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Prasad Krishnan
- Department of Veterinary and Biomedical Sciences and the Center of Molecular Toxicology and Carcinogenesis and
| | - Christina Lee
- Department of Veterinary and Biomedical Sciences and the Center of Molecular Toxicology and Carcinogenesis and
| | - Prajakta P Albrecht
- Department of Veterinary and Biomedical Sciences and the Center of Molecular Toxicology and Carcinogenesis and
| | - Weiwei Shan
- Department of Veterinary and Biomedical Sciences and the Center of Molecular Toxicology and Carcinogenesis and
| | - Moses T Bility
- Department of Veterinary and Biomedical Sciences and the Center of Molecular Toxicology and Carcinogenesis and
| | - Craig B Marcus
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Jyh M Lin
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University, Milton S. Hershey Medical Center, Hershey, PA 17033, USA and
| | - Shantu Amin
- Department of Pharmacology, Penn State Cancer Institute, The Pennsylvania State University, Milton S. Hershey Medical Center, Hershey, PA 17033, USA and
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, Bethesda, MD 20892, USA
| | - Gary H Perdew
- Department of Veterinary and Biomedical Sciences and the Center of Molecular Toxicology and Carcinogenesis and The Graduate Program in Biochemistry, Microbiology, and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Jeffrey M Peters
- Department of Veterinary and Biomedical Sciences and the Center of Molecular Toxicology and Carcinogenesis and The Graduate Program in Biochemistry, Microbiology, and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA,
| |
Collapse
|
26
|
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
|
27
|
Ceramide synthesis in the epidermis. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:422-34. [PMID: 23988654 DOI: 10.1016/j.bbalip.2013.08.011] [Citation(s) in RCA: 180] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 08/09/2013] [Accepted: 08/12/2013] [Indexed: 11/20/2022]
Abstract
The epidermis and in particular its outermost layer the stratum corneum provides terrestrial vertebrates with a pivotal defensive barrier against water loss, xenobiotics and harmful pathogens. A vital demand for this epidermal permeability barrier is the lipid-enriched lamellar matrix that embeds the enucleated corneocytes. Ceramides are the major components of these highly ordered intercellular lamellar structures, in which linoleic acid- and protein-esterified ceramides are crucial for structuring and maintaining skin barrier integrity. In this review, we describe the fascinating diversity of epidermal ceramides including 1-O-acylceramides. We focus on epidermal ceramide biosynthesis emphasizing its metabolic and topological requirements and discuss enzymes that may be involved in α- and ω-hydroxylation. Finally, we turn to epidermal ceramide regulation, highlighting transcription factors and liposensors recently described to play crucial roles in modulating skin lipid metabolism and epidermal barrier homeostasis. 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.
Collapse
|
28
|
Mizutani Y, Sun H, Ohno Y, Sassa T, Wakashima T, Obara M, Yuyama K, Kihara A, Igarashi Y. Cooperative Synthesis of Ultra Long-Chain Fatty Acid and Ceramide during Keratinocyte Differentiation. PLoS One 2013; 8:e67317. [PMID: 23826266 PMCID: PMC3694974 DOI: 10.1371/journal.pone.0067317] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 05/16/2013] [Indexed: 12/17/2022] Open
Abstract
The lipid lamellae in the stratum corneum is important for the epidermal permeability barrier. The lipid lamellae component ceramide (CER), comprising an ultra long-chain (ULC) fatty acid (FA) of ≥26 carbons (ULC CER), plays an essential role in barrier formation. ULC acyl-CoAs, produced by the FA elongase ELOVL4, are converted to ULC CERs by the CER synthase CERS3. In the presented study, we observed that ELOVL4 and CERS3 mRNAs increased during keratinocyte differentiation in vivo and in vitro. We also determined that peroxisome proliferator-activated receptor β/δ is involved in the up-regulation of the mRNAs. Knockdown of CERS3 caused a reduction in the elongase activities toward ULC acyl-CoAs, suggesting that CERS3 positively regulates ULCFA. Thus, we reveal that the two key players in ULC CER production in epidermis, CERS3 and ELOVL4, are coordinately regulated at both the transcriptional and enzymatic levels.
Collapse
Affiliation(s)
- Yukiko Mizutani
- Laboratory of Biomembrane and Biofunctional Chemistry, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Hui Sun
- Laboratory of Biomembrane and Biofunctional Chemistry, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Yusuke Ohno
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Takayuki Sassa
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Takeshi Wakashima
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Mari Obara
- Laboratory of Biomembrane and Biofunctional Chemistry, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Kohei Yuyama
- Laboratory of Biomembrane and Biofunctional Chemistry, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Akio Kihara
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
- * E-mail: (YI); (AK)
| | - Yasuyuki Igarashi
- Laboratory of Biomembrane and Biofunctional Chemistry, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
- * E-mail: (YI); (AK)
| |
Collapse
|
29
|
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
|
30
|
Khozoie C, Borland MG, Zhu B, Baek S, John S, Hager GL, Shah YM, Gonzalez FJ, Peters JM. Analysis of the peroxisome proliferator-activated receptor-β/δ (PPARβ/δ) cistrome reveals novel co-regulatory role of ATF4. BMC Genomics 2012; 13:665. [PMID: 23176727 PMCID: PMC3556323 DOI: 10.1186/1471-2164-13-665] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 11/22/2012] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The present study coupled expression profiling with chromatin immunoprecipitation sequencing (ChIP-seq) to examine peroxisome proliferator-activated receptor-β/δ (PPARβ/δ)-dependent regulation of gene expression in mouse keratinocytes, a cell type that expresses PPARβ/δ in high concentration. RESULTS Microarray analysis elucidated eight different types of regulation that modulated PPARβ/δ-dependent gene expression of 612 genes ranging from repression or activation without an exogenous ligand, repression or activation with an exogenous ligand, or a combination of these effects. Bioinformatic analysis of ChIP-seq data demonstrated promoter occupancy of PPARβ/δ for some of these genes, and also identified the presence of other transcription factor binding sites in close proximity to PPARβ/δ bound to chromatin. For some types of regulation, ATF4 is required for ligand-dependent induction of PPARβ/δ target genes. CONCLUSIONS PPARβ/δ regulates constitutive expression of genes in keratinocytes, thus suggesting the presence of one or more endogenous ligands. The diversity in the types of gene regulation carried out by PPARβ/δ is consistent with dynamic binding and interactions with chromatin and indicates the presence of complex regulatory networks in cells expressing high levels of this nuclear receptor such as keratinocytes. Results from these studies are the first to demonstrate that differences in DNA binding of other transcription factors can directly influence the transcriptional activity of PPARβ/δ.
Collapse
Affiliation(s)
- Combiz Khozoie
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Michael G Borland
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, 16802, USA
- Present address: Department of Chemistry and Biochemistry, Bloomsburg University of Pennsylvania, Bloomsburg, PA, USA
| | - Bokai Zhu
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Songjoon Baek
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Sam John
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, Bethesda, MD, 20892, USA
- Present address: Genome Sciences, University of Washington, Seattle, WA, USA
| | - Gordon L Hager
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Yatrik M Shah
- Laboratory of Metabolism, National Cancer Institute, Bethesda, MD, 20892, USA
- Present address: Department of Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Jeffrey M Peters
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
31
|
Janssens M, van Smeden J, Gooris GS, Bras W, Portale G, Caspers PJ, Vreeken RJ, Hankemeier T, Kezic S, Wolterbeek R, Lavrijsen AP, Bouwstra JA. Increase in short-chain ceramides correlates with an altered lipid organization and decreased barrier function in atopic eczema patients. J Lipid Res 2012; 53:2755-66. [PMID: 23024286 DOI: 10.1194/jlr.p030338] [Citation(s) in RCA: 332] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A hallmark of atopic eczema (AE) is skin barrier dysfunction. Lipids in the stratum corneum (SC), primarily ceramides, fatty acids, and cholesterol, are crucial for the barrier function, but their role in relation to AE is indistinct. Filaggrin is an epithelial barrier protein with a central role in the pathogenesis of AE. Nevertheless, the precise causes of AE-associated barrier dysfunction are largely unknown. In this study, a comprehensive analysis of ceramide composition and lipid organization in nonlesional SC of AE patients and control subjects was performed by means of mass spectrometry, infrared spectroscopy, and X-ray diffraction. In addition, the skin barrier and clinical state of the disease were examined. The level of ceramides with an extreme short chain length is drastically increased in SC of AE patients, which leads to an aberrant lipid organization and a decreased skin barrier function. Changes in SC lipid properties correlate with disease severity but are independent of filaggrin mutations. We demonstrate for the first time that changes in ceramide chain length and lipid organization are directly correlated with the skin barrier defects in nonlesional skin of AE patients. We envisage that these insights will provide a new therapeutic entry in therapy and prevention of AE.
Collapse
Affiliation(s)
- Michelle Janssens
- Department of Drug Delivery Technology, Leiden/Amsterdam Center for Drug Research, Leiden University, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Despite the acknowledged contributions of a defective epidermal permeability barrier, dryness of the skin, and the propensity to develop secondary infections to the etiology and pathophysiology of atopic dermatitis (AD), these epidermal changes have, until recently, been assumed to reflect downstream consequences that are secondary phenomena of the primary immunologic abnormality--the historical "inside-outside" view that AD is basically an intrinsic inflammatory disease. In this review, we focused on the role of the epidermal barrier function in the pathophysiology of AD. Specifically, we presented data in support of a barrier-initiated pathogenesis of AD, ie, the "outside-inside" concept. First, we reviewed the evidence on the existence of inherited barrier abnormalities in AD. Reported studies on the possible association of mutations in the filaggrin gene (FLG) and data on human tissue kallikreins (KLKs) and AD have been addressed. We then dealt with the question of the causal link between impaired epidermal barrier and inflammation. Finally, the association between innate immune defense system and the increased avidity of Staphylococcus aureus for atopic skin was examined. Despite very convincing evidence to support the barrier-initiated pathogenesis of AD, the view that AD reflects the downstream consequences of a primary immunologic abnormality cannot be dismissed out of hand. Almost every line of evidence in support of the role of the epidermal barrier as the "driver" of the disease activity can be challenged and at least partially contradicted by opposing evidence. Until more data are available and until all the dust settles around this issue, we should take advantage of what we already know and use our knowledge for practical purposes. Deployment of specific strategies to restore the barrier function in AD means the use of moisturizers as first-line therapy.
Collapse
Affiliation(s)
- Ronni Wolf
- Dermatology Unit, Kaplan Medical Center, Rehovot 76100, Israel.
| | | |
Collapse
|
33
|
Abstract
A major function of the skin is to provide a barrier to the movement of water and electrolytes, which is required for life in a terrestrial environment. This permeability barrier is localized to the stratum corneum and is mediated by extracellular lipid-enriched lamellar membranes, which are delivered to the extracellular spaces by the secretion of lamellar bodies by stratum granulosum cells. A large number of factors have been shown to regulate the formation of this permeability barrier. Specifically, lamellar body secretion and permeability barrier formation are accelerated by decreases in the calcium content in the stratum granulosum layer of the epidermis. In addition, increased expression of cytokines and growth factors and the activation of nuclear hormone receptors (peroxisome proliferator-activated receptors, liver X receptors, vitamin D receptor) accelerate permeability barrier formation. In contrast, nitric oxide, protease-activated receptor 2 activation, glucocorticoids, and testosterone inhibit permeability barrier formation. The ability of a variety of factors to regulate permeability barrier formation allows for a more precise and nuanced regulation.
Collapse
|
34
|
Therapeutic implications of a barrier-based pathogenesis of atopic dermatitis. Clin Rev Allergy Immunol 2012; 41:282-95. [PMID: 21174234 DOI: 10.1007/s12016-010-8231-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Excessive Th2 cell signaling and IgE production play key roles in the pathogenesis of atopic dermatitis (AD). Yet, recent information suggests that the inflammation in AD instead is initiated by inherited insults to the barrier, including a strong association between mutations in FILAGGRIN and SPINK5 in Netherton syndrome, the latter of which provides an important clue that AD is provoked by excess serine protease activity. But acquired stressors to the barrier may also be required to initiate inflammation in AD, and in addition, microbial colonization by Staphylococcus aureus both amplifies inflammation, but also further stresses the barrier in AD. Therapeutic implications of these insights are as follows: While current therapy has been largely directed toward ameliorating Th2-mediated inflammation and/or pruritus, these therapies are fraught with short-term and potential long-term risks. In contrast, "barrier repair" therapy, with a ceramide-dominant triple-lipid mixture of stratum corneum lipids, is more logical, of proven efficacy, and it provides a far-improved safety profile.
Collapse
|
35
|
Peters JM, Foreman JE, Gonzalez FJ. Dissecting the role of peroxisome proliferator-activated receptor-β/δ (PPARβ/δ) in colon, breast, and lung carcinogenesis. Cancer Metastasis Rev 2012; 30:619-40. [PMID: 22037942 DOI: 10.1007/s10555-011-9320-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Peroxisome proliferator-activated receptor-β/δ (PPARβ/δ) is a promising drug target since its agonists increase serum high-density lipoprotein; decrease low-density lipoprotein, triglycerides, and insulin associated with metabolic syndrome; improve insulin sensitivity; and decrease high fat diet-induced obesity. PPARβ/δ agonists also promote terminal differentiation and elicit anti-inflammatory activities in many cell types. However, it remains to be determined whether PPARβ/δ agonists can be developed as therapeutics because there are reports showing either pro- or anti-carcinogenic effects of PPARβ/δ in cancer models. This review examines studies reporting the role of PPARβ/δ in colon, breast, and lung cancers. The prevailing evidence would suggest that targeting PPARβ/δ is not only safe but could have anti-carcinogenic protective effects.
Collapse
Affiliation(s)
- Jeffrey M Peters
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA.
| | | | | |
Collapse
|
36
|
Schäfer M, Farwanah H, Willrodt AH, Huebner AJ, Sandhoff K, Roop D, Hohl D, Bloch W, Werner S. Nrf2 links epidermal barrier function with antioxidant defense. EMBO Mol Med 2012; 4:364-79. [PMID: 22383093 PMCID: PMC3403295 DOI: 10.1002/emmm.201200219] [Citation(s) in RCA: 144] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2011] [Revised: 01/17/2012] [Accepted: 01/20/2012] [Indexed: 12/18/2022] Open
Abstract
The skin provides an efficient permeability barrier and protects from microbial invasion and oxidative stress. Here, we show that these essential functions are linked through the Nrf2 transcription factor. To test the hypothesis that activation of Nrf2 provides skin protection under stress conditions, we determined the consequences of pharmacological or genetic activation of Nrf2 in keratinocytes. Surprisingly, mice with enhanced Nrf2 activity in keratinocytes developed epidermal thickening, hyperkeratosis and inflammation resembling lamellar ichthyosis. This resulted from upregulation of the cornified envelope proteins small proline-rich proteins (Sprr) 2d and 2h and of secretory leukocyte peptidase inhibitor (Slpi), which we identified as novel Nrf2 targets in keratinocytes. Since Sprrs are potent scavengers of reactive oxygen species and since Slpi has antimicrobial activities, their upregulation contributes to Nrf2's protective function. However, it also caused corneocyte fragility and impaired desquamation, followed by alterations in the epidermal lipid barrier, inflammation and overexpression of mitogens that induced keratinocyte hyperproliferation. These results identify an unexpected role of Nrf2 in epidermal barrier function, which needs to be considered for pharmacological use of Nrf2 activators.
Collapse
Affiliation(s)
- Matthias Schäfer
- Department of Biology, Institute of Cell Biology, ETH Zurich, Zurich, Switzerland.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Nakamura Y, Nakamura T, Tarui T, Inoue J, Kinoshita S. Functional role of PPARδ in corneal epithelial wound healing. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 180:583-98. [PMID: 22119718 DOI: 10.1016/j.ajpath.2011.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 10/02/2011] [Accepted: 10/11/2011] [Indexed: 12/23/2022]
Abstract
The peroxisome proliferator-activated receptor (PPAR) δ is involved in tissue repair. In this study, we investigated the functional role of PPARδ in corneal epithelial wound healing. In an in vivo corneal wound-healing model, the changes of PPARδ expression in corneal epithelia were examined by immunofluorescence microscopy, and the effect of topical administrations of a PPARδ agonist on corneal wound healing was also evaluated. The inhibitory effect of a PPARδ agonist on the cytokine-induced death of human corneal epithelial cells was evaluated using a DNA fragmentation assay kit. The changes of PPARδ expression and epithelial cell death were also investigated using human corneoscleral tissues ex vivo. Our findings showed that PPARδ expression was temporally up-regulated in corneal epithelial cells during experimental wound healing and that topical administration of a PPARδ agonist significantly promoted the healing of experimental corneal epithelial wounds. In human corneal epithelial cells, up-regulation of PPARδ and DNA fragmentation was demonstrated by stimulation with cytokines, and the DNA fragmentation was significantly inhibited by pretreatment with a PPARδ agonist. By using human corneoscleral tissues ex vivo, PPARδ was up-regulated in both healthy corneal epithelia (during re-epithelialization) and diseased corneal epithelia. Inflammatory stimulation-induced corneal epithelial cell death was inhibited by pretreatment with a PPARδ agonist. These results strongly suggest that PPARδ is involved in the corneal epithelial wound healing.
Collapse
|
38
|
Murakami I, Wakasa Y, Yamashita S, Kurihara T, Zama K, Kobayashi N, Mizutani Y, Mitsutake S, Shigyo T, Igarashi Y. Phytoceramide and sphingoid bases derived from brewer's yeast Saccharomyces pastorianus activate peroxisome proliferator-activated receptors. Lipids Health Dis 2011; 10:150. [PMID: 21861924 PMCID: PMC3176198 DOI: 10.1186/1476-511x-10-150] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 08/24/2011] [Indexed: 12/27/2022] Open
Abstract
Background Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that regulate lipid and glucose metabolism. PPARα is highly expressed in the liver and controls genes involved in lipid catabolism. We previously reported that synthetic sphingolipid analogs, part of which contains shorter-length fatty acid chains than natural sphingolipids, stimulated the transcriptional activities of PPARs. Sphingosine and dihydrosphingosine (DHS) are abundant sphingoid bases, and ceramide and dihydroceramide are major ceramide species in mammals. In contrast, phytosphingosine (PHS) and DHS are the main sphingoid bases in fungi. PHS and phytoceramide exist in particular tissues such as the epidermis in mammals, and involvement of ceramide species in PPARβ activation in cultured keratinocytes has been reported. The purpose of the present study is to investigate whether natural sphingolipids with C18 fatty acid and yeast-derived sphingoid bases activate PPARs as PPAR agonists. Method Lipids of brewer's yeast contain PHS- and DHS-based sphingolipids. To obtain the sphingoid bases, lipids were extracted from brewer's yeast and acid-hydrolyzed. The sphingoid base fraction was purified and quantified. To assess the effects of sphingolipids on PPAR activation, luciferase reporter assay was carried out. NIH/3T3 and human hepatoma (HepG2) cells were transfected with expression vectors for PPARs and retinoid × receptors, and PPAR responsive element reporter vector. When indicated, the PPAR/Gal4 chimera system was performed to enhance the credibility of experiments. Sphingolipids were added to the cells and the dual luciferase reporter assay was performed to determine the transcriptional activity of PPARs. Results We observed that phytoceramide increased the transcriptional activities of PPARs significantly, whereas ceramide and dihydroceramide did not change PPAR activities. Phytoceramide also increased transactivation of PPAR/Gal4 chimera receptors. Yeast-derived sphingoid base fraction, which contained PHS and DHS, or authentic PHS or DHS increased PPAR-dependent transcription. Additionally, phytoceramide stimulated PPARα activity in HepG2 hepatocytes, suggesting that phytoceramide activates genes regulated by PPARα. Conclusions Phytoceramide and yeast-derived sphingoid bases activate PPARs, whereas ceramide and dihydroceramide do not change the PPAR activity. The present findings suggest that phytoceramide acts as a PPAR ligand that would regulate PPAR-targeted genes.
Collapse
Affiliation(s)
- Itsuo Murakami
- Department of Biomembrane and Biofunctional Chemistry, Faculty of Advanced Life Sciences, Hokkaido University, Nishi 11, Kita 21, Kita-ku, Sapporo 001-0021, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Crack LR, Chan HW, McPherson T, Ogg GS. Phenotypic analysis of perennial airborne allergen-specific CD4+ T cells in atopic and non-atopic individuals. Clin Exp Allergy 2011; 41:1555-67. [PMID: 21729183 DOI: 10.1111/j.1365-2222.2011.03819.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Accumulating evidence suggests that T cells play an important role in the pathogenesis of atopic dermatitis (AD); yet, little is known of the differentiation status of CD4+ T cells specific for common environmental allergens, such as the major cat allergen, Fel d 1. OBJECTIVE To determine the frequency, differentiation phenotype and function of circulating Fel d 1-specific CD4+ T cells in adult individuals with severe persistent AD in comparison with healthy controls. METHODS Using HLA class II tetrameric complexes based on a HLA-DPB1*0401-restricted Fel d 1 epitope, ex vivo and cultured T cell frequency and phenotype were analysed in individuals with AD and healthy controls. Cytokine secretion was measured by ex vivo and cultured IL-4 and IFN-γ ELISpots. RESULTS Ex vivo Fel d 1-specific DPB1*0401-restricted CD4+ T cells in both atopics and non-atopics express high levels of CCR7, CD62L, CD27 and CD28, placing the cells largely within the central memory subgroup. However, the functional phenotype was distinct, with greater IL-4 production from the cells derived from atopics, which correlated with disease severity. CONCLUSIONS AND CLINICAL RELEVANCE Circulating Fel d 1-specific DPB1*0401-restricted CD4+ T cells in both atopic and non-atopic donors maintain a central memory phenotype; however in atopics, the cells had greater Th2 effector function, compatible with a disease model of altered antigen delivery in atopic individuals.
Collapse
Affiliation(s)
- L R Crack
- MRC Human Immunology Unit, University of Oxford, Weatherall Institute of Molecular Medicine, Oxford, UK
| | | | | | | |
Collapse
|
40
|
Jiang YJ, Kim P, Lu YF, Feingold KR. PPARgamma activators stimulate aquaporin 3 expression in keratinocytes/epidermis. Exp Dermatol 2011; 20:595-9. [PMID: 21457357 DOI: 10.1111/j.1600-0625.2011.01269.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Aquaporin 3 (AQP3), a member of the aquaglyceroporin family, which transports water and glycerol, is robustly expressed in epidermis and plays an important role in stratum corneum hydration, permeability barrier function and wound healing. PPAR and LXR activation regulates the expression of many proteins in the epidermis and thereby can affect epidermal function. Here, we report that PPARgamma activators markedly stimulate AQP3 mRNA expression in both undifferentiated and differentiated cultured human keratinocytes (CHKs). The increase in AQP3 mRNA by PPARgamma activator occurs in a dose- and time-dependent fashion. Increased AQP3 mRNA levels are accompanied by an increase in AQP3 protein in undifferentiated keratinocytes and a significant increase in glycerol uptake. Activation of LXR, RAR and RXR also increases AQP3 mRNA levels in undifferentiated and differentiated CHKs, but to a lesser extent. PPARdelta activation stimulates AQP3 expression in undifferentiated CHKs but decreases expression in differentiated CHKs. In contrast, PPARalpha activators do not alter AQP3 expression. AQP9 and AQP10, other members of aquaglyceroporin family, are less abundantly expressed in CHKs, and their expression levels are not significantly altered by treatment with LXR, PPAR, RAR or RXR activators. Finally, when topically applied, the PPARgamma activator, ciglitazone, induces AQP3 but not AQP9 gene expression in mouse epidermis. Our data demonstrate that PPAR and LXR activators stimulate AQP3 expression, providing an additional mechanism by which PPAR and LXR activators regulate epidermal function.
Collapse
Affiliation(s)
- Yan J Jiang
- Metabolism Section, Veterans Affairs Medical Center, Northern California Institute for Research and Education, University of California at San Francisco, San Francisco, CA, USA
| | | | | | | |
Collapse
|
41
|
Feingold KR, Jiang YJ. The mechanisms by which lipids coordinately regulate the formation of the protein and lipid domains of the stratum corneum: Role of fatty acids, oxysterols, cholesterol sulfate and ceramides as signaling molecules. DERMATO-ENDOCRINOLOGY 2011; 3:113-8. [PMID: 21695021 PMCID: PMC3117011 DOI: 10.4161/derm.3.2.14996] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Accepted: 12/10/2010] [Indexed: 12/14/2022]
Abstract
The formation of a permeability barrier between the external environment and the host is essential for survival. To provide this barrier keratinocytes undergo a complex pathway of differentiation, which culminates in keratinocyte cornification and the formation of extracellular lipid enriched lamellar membranes in the stratum corneum. The mechanisms that coordinately regulate the parallel formation of the corneocytes and lamellar membranes are unknown. The extracellular lamellar membranes are derived from the exocytosis of lamellar bodies and to synthesize lamellar bodies the keratinocyte must have abundant quantities of cholesterol, fatty acids and ceramides. These lipids could serve as signaling molecules and thereby coordinately regulate the formation of the stratum corneum. Fatty acids activate PPARs and studies have shown that PPAR activation stimulates keratinocyte differentiation. Cholesterol is converted to oxysterols that activate LXR and studies have shown that LXR activation also stimulates keratinocyte differentation. Additionally, PPAR and LXR activation also facilitates the formation of the lipid enriched lamellar membranes. Ceramides, via a number of mechanisms also stimulate keratinocyte differentiation. Recently, studies have shown that ceramides by increasing PPAR delta also increase the expression of ABCA12, which would facilitate the formation of lamellar bodies. Finally, keratinocytes accumulate a large quantity of cholesterol sulfate, which plays a key role in regulating desquamation. Cholesterol sulfate has also been shown to stimulate keratinocyte differentiation. Thus, cholesterol, cholesterol sulfate, fatty acids and ceramides all stimulate keratinocyte differentiation and thereby could coordinately regulate the formation of the stratum corneum.
Collapse
Affiliation(s)
- Kenneth R Feingold
- Metabolism Section and Dermatology Service; Department of Veterans Affairs Medical Center; University of California, San Francisco; San Francisco, CA USA
| | | |
Collapse
|
42
|
Man M, Hupe M, Mackenzie D, Kim H, Oda Y, Crumrine D, Lee SH, Martin-Ezquerra G, Trullas C, Mauro TM, Feingold KR, Elias PM, Man MQ. A topical Chinese herbal mixture improves epidermal permeability barrier function in normal murine skin. Exp Dermatol 2011; 20:285-288. [PMID: 21323748 PMCID: PMC4548955 DOI: 10.1111/j.1600-0625.2010.01205.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Chinese herbal medicine (CHM) has been shown to have beneficial effects for both skin disorders with barrier abnormality and as skin care ingredients. Yet, how CHM exerts their benefits is unclear. As most, if not all, inflammatory dermatoses are accompanied by abnormal permeability barrier function, we assessed the effects of topical CHM extracts on epidermal permeability barrier function and their potential mechanisms. Topical CHM accelerated barrier recovery following acute barrier disruption. Epidermal lipid content and mRNA expression of fatty acid and ceramide synthetic enzymes increased following topical CHM treatment in addition to mRNA levels for the epidermal glucosylceramide transport protein, ATP-binding cassette A12. Likewise, CHM extract increased mRNA expression of antimicrobial peptides both in vivo and in vitro. These results demonstrate that the topical CHM extract enhances epidermal permeability barrier function, suggesting that topical CHM could provide an alternative regimen for the prevention/treatment of inflammatory dermatoses accompanied by barrier abnormalities.
Collapse
Affiliation(s)
- Mona Man
- Dermatology Service, VAMC, and Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Melanie Hupe
- Dermatology Service, VAMC, and Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Donald Mackenzie
- Dermatology Service, VAMC, and Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Hyunjung Kim
- Department of Dermatology, Yonsei University College of Medicine, Yongdong Severance Hospital, Seoul, Korea
| | - Yuko Oda
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Debra Crumrine
- Dermatology Service, VAMC, and Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Seung Hun Lee
- Department of Dermatology, Yonsei University College of Medicine, Yongdong Severance Hospital, Seoul, Korea
| | | | | | - Theodora M. Mauro
- Dermatology Service, VAMC, and Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Kenneth R. Feingold
- Dermatology Service, VAMC, and Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Peter M. Elias
- Dermatology Service, VAMC, and Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Mao-Qiang Man
- Dermatology Service, VAMC, and Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
43
|
Qin H, Zheng X, Zhong X, Shetty AK, Elias PM, Bollag WB. Aquaporin-3 in keratinocytes and skin: its role and interaction with phospholipase D2. Arch Biochem Biophys 2011; 508:138-43. [PMID: 21276418 DOI: 10.1016/j.abb.2011.01.014] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 01/20/2011] [Accepted: 01/21/2011] [Indexed: 12/30/2022]
Abstract
Aquaporin 3 (AQP3) is an aquaglyceroporin that transports water and glycerol and is expressed in the epidermis, among other epithelial tissues. We have recently shown that there is an association between this glycerol channel and phospholipase D2 (PLD2) in caveolin-rich membrane microdomains. While PLD2 is able to hydrolyze membrane phospholipids to generate phosphatidic acid, this enzyme also catalyzes, in the presence of primary alcohols, a transphosphatidylation reaction to produce a phosphatidylalcohol. We have proposed that AQP3 associated with PLD2 provides the physiological primary alcohol glycerol to PLD2 for use in the transphosphatidylation reaction to generate phosphatidylglycerol (PG). Further, we have proposed that PG functions as a signaling molecule to mediate early epidermal keratinocyte differentiation, and manipulation of this signaling module inhibits keratinocyte proliferation and enhances differentiation. In contrast, other investigators have suggested a proliferative role for AQP3 in keratinocytes. In addition, AQP3 knockout mice exhibit an epidermal phenotype, characterized by dry skin, decreased elasticity and delayed barrier repair and wound healing, which can be corrected by glycerol but not other humectants. AQP3 levels have also been found to be altered in human skin diseases. In this article the evidence supporting a role for AQP3 in the epidermis will be discussed.
Collapse
Affiliation(s)
- Haixia Qin
- Institute of Molecular Medicine and Genetics, Georgia Health Sciences University (formerly Medical College of Georgia), Augusta, GA 30912, USA
| | | | | | | | | | | |
Collapse
|
44
|
Lass A, Zimmermann R, Oberer M, Zechner R. Lipolysis - a highly regulated multi-enzyme complex mediates the catabolism of cellular fat stores. Prog Lipid Res 2010; 50:14-27. [PMID: 21087632 PMCID: PMC3031774 DOI: 10.1016/j.plipres.2010.10.004] [Citation(s) in RCA: 460] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 10/12/2010] [Accepted: 10/13/2010] [Indexed: 12/17/2022]
Abstract
Lipolysis is the biochemical pathway responsible for the catabolism of triacylglycerol (TAG) stored in cellular lipid droplets. The hydrolytic cleavage of TAG generates non-esterified fatty acids, which are subsequently used as energy substrates, essential precursors for lipid and membrane synthesis, or mediators in cell signaling processes. Consistent with its central importance in lipid and energy homeostasis, lipolysis occurs in essentially all tissues and cell types, it is most abundant, however, in white and brown adipose tissue. Over the last 5years, important enzymes and regulatory protein factors involved in lipolysis have been identified. These include an essential TAG hydrolase named adipose triglyceride lipase (ATGL) [annotated as patatin-like phospholipase domain-containing protein A2], the ATGL activator comparative gene identification-58 [annotated as α/β hydrolase containing protein 5], and the ATGL inhibitor G0/G1 switch gene 2. Together with the established hormone-sensitive lipase [annotated as lipase E] and monoglyceride lipase, these proteins constitute the basic "lipolytic machinery". Additionally, a large number of hormonal signaling pathways and lipid droplet-associated protein factors regulate substrate access and the activity of the "lipolysome". This review summarizes the current knowledge concerning the enzymes and regulatory processes governing lipolysis of fat stores in adipose and non-adipose tissues. Special emphasis will be given to ATGL, its regulation, and physiological function.
Collapse
Affiliation(s)
- Achim Lass
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | | | | | | |
Collapse
|
45
|
Elias PM. Therapeutic Implications of a Barrier-based Pathogenesis of Atopic Dermatitis. Ann Dermatol 2010; 22:245-54. [PMID: 20711259 DOI: 10.5021/ad.2010.22.3.245] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 06/18/2010] [Accepted: 06/21/2010] [Indexed: 12/15/2022] Open
Abstract
In this review, I first provide relevant background information about normal epidermal barrier structure and function. I then update recent information about how inherited defects in either filaggrin and/or in the serine protease inhibitor, lymphoepithelial Kazal-type inhibitor 1, converge to stimulate the development of atopic dermatitis (AD). Next I explain the multiple mechanisms whereby a primary barrier abnormality in AD can lead to inflammation. Furthermore, I explore how certain acquired stressors, such as a reduced external humidity, high pH soaps/surfactants, psychological stress, as well as secondary Staphylococcus aureus infections initiate or further aggravate AD. Finally, and most importantly, I compare various therapeutic paradigms for AD, highlighting the risks and benefits of glucocorticoids and immunomodulators vs. corrective, lipid replacement therapy.
Collapse
Affiliation(s)
- Peter M Elias
- Dermatology Service, Veterans Affairs Medical Center, and Department of Dermatology, University of California, San Francisco, CA, USA
| |
Collapse
|
46
|
Jiang YJ, Lu B, Tarling EJ, Kim P, Man MQ, Crumrine D, Edwards PA, Elias PM, Feingold KR. Regulation of ABCG1 expression in human keratinocytes and murine epidermis. J Lipid Res 2010; 51:3185-95. [PMID: 20675829 DOI: 10.1194/jlr.m006445] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
ABCG1, a member of the ATP binding cassette superfamily, facilitates the efflux of cholesterol from cells to HDL. In this study, we demonstrate that ABCG1 is expressed in cultured human keratinocytes and murine epidermis, and induced during keratinocyte differentiation, with increased levels in the outer epidermis. ABCG1 is regulated by liver X receptor (LXR) and peroxisome proliferator-activated receptor-δ (PPAR-δ) activators, cellular sterol levels, and acute barrier disruption. Both LXR and PPAR-δ activators markedly stimulate ABCG1 expression in a dose- and time-dependent fashion. PPAR-γ activators also increase ABCG1 expression, but to a lesser degree. In contrast, activators of PPAR-α, retinoic acid receptor, retinoid X receptor, and vitamin D receptor do not alter ABCG1 expression. In response to increased intracellular sterol levels, ABCG1 expression increases, whereas inhibition of cholesterol biosynthesis decreases ABCG1 expression. In vivo, ABCG1 is stimulated 3-6 h after acute barrier disruption by either tape stripping or acetone treatment, an increase that can be inhibited by occlusion, suggesting a potential role of ABCG1 in permeability barrier homeostasis. Although Abcg1-null mice display normal epidermal permeability barrier function and gross morphology, abnormal lamellar body (LB) contents and secretion leading to impaired lamellar bilayer formation could be demonstrated by electron microscopy, indicating a potential role of ABCG1 in normal LB formation and secretion.
Collapse
Affiliation(s)
- Yan J Jiang
- Metabolism Section, Veterans Affairs Medical Center, Northern California Institute for Research and Education, University of California at San Francisco, San Francisco, CA 94121, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Liu J, Man WY, Lv CZ, Song SP, Shi YJ, Elias PM, Man MQ. Epidermal permeability barrier recovery is delayed in vitiligo-involved sites. Skin Pharmacol Physiol 2010; 23:193-200. [PMID: 20185976 DOI: 10.1159/000288166] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Accepted: 01/07/2010] [Indexed: 01/17/2023]
Abstract
BACKGROUND/OBJECTIVES Prior studies have demonstrated that both the skin surface pH and epidermal permeability barrier function vary with skin pigmentation types. Although melanin deficiency is the main feature of vitiligo, alterations in cutaneous biophysical properties in vitiligo have not yet been well defined. In the present study, stratum corneum (SC) hydration, the skin surface pH and epidermal permeability barrier function in vitiligo were evaluated. METHODS A total of 30 volunteers with vitiligo comprising 19 males and 11 females aged 13-51 years (mean age: 27.91 +/- 2.06 years) were enrolled in this study. The skin surface pH, SC hydration, melanin/erythema index and transepidermal water loss (TEWL) were measured by respective probes connected to a Courage-Khazaka MPA5. SC integrity was determined by measuring the TEWL following each D-Squame application. The barrier recovery rate was assessed at 5 h following barrier disruption by repeated tape stripping. RESULTS In addition to SC hydration, both melanin and erythema index were significantly lower in vitiligo lesions than in contralateral, nonlesional sites, while no difference in skin surface pH between vitiligo-involved and uninvolved areas was observed. In addition, neither the basal TEWL nor SC integrity in the involved areas differed significantly from that in the uninvolved areas. However, barrier recovery in vitiligo-involved sites was significantly delayed in comparison with uninvolved sites (40.83 +/- 5.39% vs. 58.30 +/- 4.71%; t = 2.441; p < 0.02). CONCLUSION Barrier recovery following tape stripping of the SC is delayed in vitiligo. Therefore, improvement in epidermal permeability barrier function may be an important unrecognized factor to be considered in treating patients with vitiligo.
Collapse
Affiliation(s)
- J Liu
- Dalian Skin Disease Hospital, Dalian, The People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
48
|
Genovese S, Foreman JE, Borland MG, Epifano F, Gonzalez FJ, Curini M, Peters JM. A natural propenoic acid derivative activates peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta). Life Sci 2010; 86:493-8. [PMID: 20153754 DOI: 10.1016/j.lfs.2010.02.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Revised: 01/27/2010] [Accepted: 02/02/2010] [Indexed: 12/13/2022]
Abstract
AIMS Previous studies showed that natural prenyloxyphenylpropanoid derivatives have potent biological properties in vivo. Given the structural similarities between these compounds and known peroxisome proliferator-activated receptor (PPAR) agonists, the present study examined the hypothesis that propenoic acid derivatives activate PPARs. MAIN METHODS Chimeric reporter assays were performed to identify propenoic acid derivates that could activate PPARs. Quantitative polymerase chain reaction (qPCR) analysis of wild-type and Pparbeta/delta-null mouse primary keratinocytes was performed to determine if a test compound could specifically activate PPARbeta/delta. A human epithelial carcinoma cell line and primary mouse keratinocytes were used to determine the effect of the compound on cell proliferation. KEY FINDINGS Three of the propenoic acid derivatives activated PPARs, with the greatest efficacy being observed with prenyloxycinnamic acid derivatives 4'-geranyloxyferulic acid (compound 1) for PPARbeta/delta. Compound 1 increased expression of a known PPARbeta/delta target gene through a mechanism that requires PPARbeta/delta. Inhibition of cell proliferation by compound 1 was found in a human epithelial carcinoma cell line. SIGNIFICANCE Results from these studies demonstrate that compound 1 can activate PPARbeta/delta and inhibit cell proliferation of a human skin cancer cell line, suggesting that the biological effects of 4'-geranyloxyferulic acid may be mediated in part by activating this PPAR isoform.
Collapse
Affiliation(s)
- Salvatore Genovese
- Dipartimento di Scienze del Farmaco, Università G. D'Annunzio, Chieti-Pescara, Via dei Vestini 31, 66100 Chieti Scalo, Italy
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW Many recent studies have revealed the key roles played by Th1/Th2 cell dysregulation, IgE production, mast cell hyperactivity, and dendritic cell signaling in the pathogenesis of atopic dermatitis. Accordingly, current therapy has been largely directed towards ameliorating Th2-mediated inflammation and/or pruritus. We will review here emerging evidence that the inflammation in atopic dermatitis results from inherited and acquired insults to the barrier and the therapeutic implications of this new paradigm. RECENT FINDINGS Recent molecular genetic studies have shown a strong association between mutations in FILAGGRIN and atopic dermatitis, particularly in Northern Europeans. But additional acquired stressors to the barrier are required to initiate inflammation. Sustained hapten access through a defective barrier stimulates a Th1 --> Th2 shift in immunophenotype, which in turn further aggravates the barrier. Secondary Staphylococcus aureus colonization not only amplifies inflammation but also further stresses the barrier in atopic dermatitis. SUMMARY These results suggest a new 'outside-to-inside, back to outside' paradigm for the pathogenesis of atopic dermatitis. This new concept is providing impetus for the development of new categories of 'barrier repair' therapy.
Collapse
|
50
|
Törmä H, Berne B. Sodium lauryl sulphate alters the mRNA expression of lipid-metabolizing enzymes and PPAR signalling in normal human skinin vivo. Exp Dermatol 2009; 18:1010-5. [DOI: 10.1111/j.1600-0625.2009.00877.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|