1
|
Kang H, Hwang HJ, Kim E, Lim SH, Choi EH. Activation of Local 11β-Hydroxysteroid Dehydrogenase Type 1 by Diosmetin Enhances Endogenous Glucocorticoid Levels to Alleviate Skin Inflammation: Insights Into a Novel Therapeutic Strategy for Atopic Dermatitis. Exp Dermatol 2025; 34:e70039. [PMID: 39887444 DOI: 10.1111/exd.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 12/10/2024] [Accepted: 01/10/2025] [Indexed: 02/01/2025]
Abstract
Glucocorticoids (GCs) are synthesised de novo by peripheral tissues and the adrenal cortex of the hypothalamic-pituitary-adrenal axis. Skin expresses an enzyme called 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), which reduces cortisone to the active hormone cortisol which activates GC receptors. 11β-HSD1 plays a significant role in alleviating atopic inflammation through the elevation of the concentrations of active GC in the skin. This study aimed to investigate the role of diosmetin as an activator of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1). In human keratinocytes, diosmetin was found to upregulate 11β-HSD1 protein expression and cortisol levels, as well as the transcriptional expression of 11β-HSD1 mRNA. However, this upregulation of 11β-HSD1 mRNA was abrogated in keratinocytes transfected with 11β-HSD1 small interfering RNA (siRNA). In an atopic dermatitis (AD) murine model, topical administration of diosmetin significantly attenuated basal transepidermal water loss (TEWL) and the Eczema Area and Severity Index (EASI), while enhancing stratum corneum (SC) hydration. Diosmetin also increased corticosterone levels in the SC and upregulated 11β-HSD1 expression in both the serum and epidermis. Furthermore, diosmetin treatment led to a marked reduction in serum immunoglobulin E (IgE) and tumour necrosis factor-α (TNF-α) levels, and suppressed mRNA expression of thymic stromal lymphopoietin (TSLP), interleukin-1β (IL-1β), IL-4, and IL-13 in the epidermis. Collectively, these findings suggest that diosmetin promotes the endogenous activation of glucocorticoids via local 11β-HSD1 activation, underscoring its potential as a novel topical therapeutic agent for the management of inflammatory skin disorders, such as AD.
Collapse
Affiliation(s)
- Hyun Kang
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Hyun Jee Hwang
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Eunjung Kim
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Sung Ha Lim
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Eung Ho Choi
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Research Institute of Metabolism and Inflammation, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| |
Collapse
|
2
|
Ding X, Huang H, Chen Y, Wu J, Yan X, Ding Y, Dong J, Wang Y, Wang L, Tan Q, Yang C. Electrospun 11β-HSD1 Inhibitor-Loaded Scaffolds for Accelerating Diabetic Ulcer Healing. ACS APPLIED BIO MATERIALS 2025; 8:435-445. [PMID: 39690109 DOI: 10.1021/acsabm.4c01397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Diabetic ulcers (DUs) are a common and severe complication of diabetes, characterized by impaired wound healing due to a complex pathophysiological mechanism. Elevated levels of 11β-hydroxysteroid dehydrogenase type I (11β-HSD1) in wounds have been demonstrated to modulate glucocorticoid activity, leading to delayed skin cell proliferation and restricted angiogenesis, ultimately hindering wound healing. In this study, we propose an electrospun poly(ε-caprolactone) (PCL) nanofiber scaffold doped with the 11β-HSD1 inhibitor BVT2733 (BPs) to prevent 11β-HSD1 activity during the diabetic wound healing process. The electrospun scaffold loaded with BVT2733 is designed to achieve localized inhibition of 11β-HSD1 in DUs. This scaffold exhibited a porous morphology and desirable drug-loading capacity, meeting the requirements for wound coverage and effective delivery of BVT2733 BPs. In vitro studies demonstrated that the sustained release of BVT2733 from the scaffold promoted skin cell proliferation and migration while stimulating angiogenesis by upregulating HIF1-α/VEGF expression. The therapeutic effect of the scaffold was further confirmed in a full-thickness wound model using diabetic mice. The mice treated with the scaffolds exhibited an accelerated wound healing rate, increased neovascularization, enhanced collagen deposition, and regeneration of skin appendages within 2 weeks postinjury. The findings here provide evidence for the use of 11β-HSD1 inhibitor-integrated biomaterials in treating DUs and represent a novel biological platform for modulating dysregulated mechanisms in DUs.
Collapse
Affiliation(s)
- Xiaofeng Ding
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Heyan Huang
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yutong Chen
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Junchao Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Xin Yan
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Youjun Ding
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jie Dong
- Jiangsu Provincial Engineering Research Centre of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yiwei Wang
- Jiangsu Provincial Engineering Research Centre of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lili Wang
- Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Qian Tan
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chenxi Yang
- Jiangsu Provincial Engineering Research Centre of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Department of Immunology, School of Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| |
Collapse
|
3
|
Hall L, Hart R. Role of corticosteroids in skin physiology and therapeutic potential of an 11β-HSD1 inhibitor: A review. Int J Dermatol 2024; 63:443-454. [PMID: 38146184 DOI: 10.1111/ijd.16967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/15/2023] [Accepted: 11/29/2023] [Indexed: 12/27/2023]
Abstract
Skin is a major site of cortisol bioconversion by 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) enzymes which catalyze intracellular inactive cortisone into physiologically active cortisol. 11β-HSD1 is highly expressed in skin, especially in dermal fibroblasts, epidermal keratinocytes, melanocytes, and hair follicles, and plays important roles in regulating keratinocytes, fibroblast proliferation, and has roles in skin aging. Inhibition of 11β-HSD1 may reverse decreased collagen levels observed in extrinsically and intrinsically aged skin. Inhibitors of 11β-HSD1 may also have the potential to reverse decreased collagen observed in skin atrophy induced by glucocorticoid treatment. This systematic review aimed to summarize the current knowledge of roles for 11β-HSD1 inhibitor in skin physiology and potential for future use in medications. Studies have demonstrated that immediately following experimental insult in an animal model, there is increased expression of 11β-HSD1, and that topical application of an 11β-HSD1 inhibitor increases the rate of healing, increases skin collagen content, increases dermal fibroblasts, and increases dermal thickness. Furthermore, in patients with type 2 diabetes mellitus, 11β-HSD1 inhibitors reduce wound diameter after injury. Further development of 11β-HSD1 inhibitors appears to be a promising area for treating aging skin, aiding wound healing, and mitigating effects of systemic glucocorticoid use. Both topically and orally administered 11β-HSD1 inhibitors appear to be viable avenues for future research.
Collapse
Affiliation(s)
- Larissa Hall
- Faculty of Science and Agriculture, Business and Law, School of Science and Technology, University of New England, Armidale, NSW, Australia
| | - Robert Hart
- Faculty of Science and Agriculture, Business and Law, School of Science and Technology, University of New England, Armidale, NSW, Australia
| |
Collapse
|
4
|
Livingstone DEW, Sooy K, Sykes C, Webster SP, Walker BR, Andrew R. 5α-Tetrahydrocorticosterone: A topical anti-inflammatory glucocorticoid with an improved therapeutic index in a murine model of dermatitis. Br J Pharmacol 2024; 181:1256-1267. [PMID: 37990638 DOI: 10.1111/bph.16285] [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: 12/20/2022] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND AND PURPOSE Glucocorticoids are powerful anti-inflammatory drugs, but are associated with many side-effects. Topical application in atopic dermatitis leads to skin thinning, metabolic changes, and adrenal suppression. 5α-Tetrahydrocorticosterone (5αTHB) is a potential selective anti-inflammatory with reduced metabolic effects. Here, the efficacy and side-effect profile of 5αTHB were compared with hydrocortisone in preclinical models of irritant dermatitis. EXPERIMENTAL APPROACH Acute irritant dermatitis was invoked in ear skin of male C57BL/6 mice with a single topical application of croton oil. Inflammation was assessed as oedema via ear weight following treatment with 5αTHB and hydrocortisone. Side-effects of 5αTHB and hydrocortisone were assessed following chronic topical steroid treatment (28 days) to non-irritated skin. Skin thinning was quantified longitudinally by caliper measurements and summarily by qPCR for transcripts for genes involved in extracellular matrix homeostasis; systemic effects of topical steroid administration also were assessed. Clearance of 5αTHB and hydrocortisone were measured following intravenous and oral administration. KEY RESULTS 5αTHB suppressed ear swelling in mice, with ED50 similar to hydrocortisone (23 μg vs. 13 μg). Chronic application of 5αTHB did not cause skin thinning, adrenal atrophy, weight loss, thymic involution, or raised insulin levels, all of which were observed with topical hydrocortisone. Transcripts for genes involved in collagen synthesis and stability were adversely affected by all doses of hydrocortisone, but only by the highest dose of 5αTHB (8× ED50 ). 5αTHB was rapidly cleared from the systemic circulation. CONCLUSIONS AND IMPLICATIONS Topical 5αTHB has potential to treat inflammatory skin conditions, particularly in areas of delicate skin.
Collapse
Affiliation(s)
- Dawn Elizabeth Watson Livingstone
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Science, University of Edinburgh, Edinburgh, UK
| | - Karen Sooy
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Catherine Sykes
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Scott Peter Webster
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Brian Robert Walker
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Ruth Andrew
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
5
|
Jin SP, Suh JH, Kim CE, Oh IG, Seo EY, Kim MK, Yoon KN, Chung JH. Functionally similar genes exhibit comparable/similar time-course expression kinetics in the UV-induced photoaged mouse model. PLoS One 2023; 18:e0290358. [PMID: 37943888 PMCID: PMC10635544 DOI: 10.1371/journal.pone.0290358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/07/2023] [Indexed: 11/12/2023] Open
Abstract
Skin photoaging induced by ultraviolet (UV) irradiation contributes to the formation of thick and coarse wrinkles. Humans are exposed to UV light throughout their lives. Therefore, it is crucial to determine the time-sequential effects of UV on the skin. In this study, we irradiated the mouse back skin with UV light for eight weeks and observed the changes in gene expressions via microarray analysis every week. There were more downregulated genes (514) than upregulated genes (123). The downregulated genes had more functional diversity than the upregulated genes. Additionally, the number of downregulated genes did not increase in a time-dependent manner. Instead, time-dependent kinetic patterns were observed. Interestingly, each kinetic cluster harbored functionally enriched gene sets. Since collagen changes in the dermis are considered to be a major cause of photoaging, we hypothesized that other gene sets contributing to photoaging would exhibit kinetics similar to those of the collagen-regulatory genes identified in this study. Accordingly, co-expression network analysis was conducted using 11 well-known collagen-regulatory seed genes to predict genes with similar kinetics. We ranked all downregulated genes from 1 to 504 based on their expression levels, and the top 50 genes were suggested to be involved in the photoaging process. Additionally, to validate and support our identified top 50 gene lists, we demonstrated that the genes (FN1, CCDC80, PRELP, and TGFBR3) we discovered are downregulated by UV irradiation in cultured human fibroblasts, leading to decreased collagen levels, which is indicative of photoaging processes. Overall, this study demonstrated the time-sequential genetic changes in chronically UV-irradiated skin and proposed 50 genes that are involved in the mechanisms of photoaging.
Collapse
Affiliation(s)
- Seon-Pil Jin
- Department of Dermatology, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Dermatology, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Joong Heon Suh
- Department of Dermatology, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Dermatology, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea
- Department of Biomedical Sciences, Graduate School, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Chang-Eop Kim
- Department of Physiology, Department of Physiology, Gachon University College of Korean Medicine, Seongnam, Republic of Korea
| | - Inn Gyung Oh
- Department of Dermatology, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Dermatology, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Eun Young Seo
- Department of Dermatology, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Dermatology, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Min-Kyoung Kim
- Department of Dermatology, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Dermatology, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Kyeong-No Yoon
- Department of Dermatology, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Dermatology, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea
- Department of Biomedical Sciences, Graduate School, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Jin Ho Chung
- Department of Dermatology, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Dermatology, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea
- Department of Biomedical Sciences, Graduate School, Seoul National University Graduate School, Seoul, Republic of Korea
| |
Collapse
|
6
|
Pofi R, Caratti G, Ray DW, Tomlinson JW. Treating the Side Effects of Exogenous Glucocorticoids; Can We Separate the Good From the Bad? Endocr Rev 2023; 44:975-1011. [PMID: 37253115 PMCID: PMC10638606 DOI: 10.1210/endrev/bnad016] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/25/2023] [Accepted: 05/26/2023] [Indexed: 06/01/2023]
Abstract
It is estimated that 2% to 3% of the population are currently prescribed systemic or topical glucocorticoid treatment. The potent anti-inflammatory action of glucocorticoids to deliver therapeutic benefit is not in doubt. However, the side effects associated with their use, including central weight gain, hypertension, insulin resistance, type 2 diabetes (T2D), and osteoporosis, often collectively termed iatrogenic Cushing's syndrome, are associated with a significant health and economic burden. The precise cellular mechanisms underpinning the differential action of glucocorticoids to drive the desirable and undesirable effects are still not completely understood. Faced with the unmet clinical need to limit glucocorticoid-induced adverse effects alongside ensuring the preservation of anti-inflammatory actions, several strategies have been pursued. The coprescription of existing licensed drugs to treat incident adverse effects can be effective, but data examining the prevention of adverse effects are limited. Novel selective glucocorticoid receptor agonists and selective glucocorticoid receptor modulators have been designed that aim to specifically and selectively activate anti-inflammatory responses based upon their interaction with the glucocorticoid receptor. Several of these compounds are currently in clinical trials to evaluate their efficacy. More recently, strategies exploiting tissue-specific glucocorticoid metabolism through the isoforms of 11β-hydroxysteroid dehydrogenase has shown early potential, although data from clinical trials are limited. The aim of any treatment is to maximize benefit while minimizing risk, and within this review we define the adverse effect profile associated with glucocorticoid use and evaluate current and developing strategies that aim to limit side effects but preserve desirable therapeutic efficacy.
Collapse
Affiliation(s)
- Riccardo Pofi
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Giorgio Caratti
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - David W Ray
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Oxford Kavli Centre for Nanoscience Discovery, University of Oxford, Oxford OX37LE, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| |
Collapse
|
7
|
Lee YB, Hwang HJ, Kim E, Lim SH, Chung CH, Choi EH. Hyperglycemia-activated 11β-hydroxysteroid dehydrogenase type 1 increases endoplasmic reticulum stress and skin barrier dysfunction. Sci Rep 2023; 13:9206. [PMID: 37280272 PMCID: PMC10244460 DOI: 10.1038/s41598-023-36294-y] [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: 05/06/2023] [Accepted: 05/31/2023] [Indexed: 06/08/2023] Open
Abstract
The diabetes mellitus (DM) skin shows skin barrier dysfunction and skin lipid abnormality, similar to conditions induced by systemic or local glucocorticoid excess and aged skin. Inactive glucocorticoid (GC) is converted into active glucocorticoid by 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1). Hyperglycemia in DM and excessive GC are known to increase endoplasmic reticulum (ER) stress. We hypothesized that hyperglycemia affects systemic GC homeostasis and that the action of skin 11β-HSD1 and GC contributes to increased ER stress and barrier defects in DM. We compared 11β-HSD1, active GC, and ER stress between hyperglycemic and normoglycemic conditions in normal human keratinocytes and db/db mice. 11β-HSD1 and cortisol increased with time in keratinocyte culture under hyperglycemic conditions. 11β-HSD1 siRNA-transfected cells did not induce cortisol elevation in hyperglycemic condition. The production of 11β-HSD1 and cortisol was suppressed in cell culture treated with an ER stress-inhibitor. The 14-week-old db/db mice showed higher stratum corneum (SC) corticosterone, and skin 11β-HSD1 levels than 8-week-old db/db mice. Topical 11β-HSD1 inhibitor application in db/db mice decreased SC corticosterone levels and improved skin barrier function. Hyperglycemia in DM may affect systemic GC homeostasis, activate skin 11β-HSD1, and induce local GC excess, which increases ER stress and adversely affects skin barrier function.
Collapse
Affiliation(s)
- Young Bin Lee
- Department of Dermatology, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, 26426, Republic of Korea
| | - Hyun Jee Hwang
- Department of Dermatology, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, 26426, Republic of Korea
| | - Eunjung Kim
- Department of Dermatology, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, 26426, Republic of Korea
| | - Sung Ha Lim
- Department of Dermatology, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, 26426, Republic of Korea
| | - Choon Hee Chung
- Department of Endocrinology and Metabolism, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Research Institute of Metabolism and Inflammation, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Eung Ho Choi
- Department of Dermatology, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, 26426, Republic of Korea.
- Research Institute of Metabolism and Inflammation, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea.
| |
Collapse
|
8
|
Ross SW, Malcolm J, Maitz J, Li Z, Wang Y, Issler-Fisher AC. Fractional ablative laser therapy for the treatment of severe burn scars: A pilot study of the underlying mechanisms. Burns 2023; 49:573-582. [PMID: 36642662 DOI: 10.1016/j.burns.2022.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 12/12/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023]
Abstract
Ablative fractional resurfacing is clinically an efficient treatment for burn scar management. The aim of this pilot study was to investigate the poorly understood mechanisms underlying ablative fractional CO2 laser (AFL-CO2) therapy in relation to biomarkers S100 and 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1). S100 stains for Langerhans cells and neuronal cells, potentially representing the pruritus experienced. 11β-HSD1 catalyses the interconversion of cortisol and cortisone in cells, promoting tissue remodelling. Immunohistochemical analysis of S100 and 11β-HSD1 protein expression in the dermis and epidermis of the skin was performed on normal skin, before and after AFL-CO2 therapy. Data assessing outcome parameters was collected concurrently with the skin biopsies. 13 patients were treated with AFL-CO2 therapy. Langerhans cells decreased by 39% after 2nd treatment. Neuronal cells were overexpressed before treatment in the scar tissue by 91% but levels returned to that resembling normal skin. 11β-HSD1 expression in keratinocytes was significantly higher after laser treatment compared to before in scar tissue (p <0.01). No clear correlation was found in dermal fibroblast numbers throughout the treatment course. Whilst the role of the explored mechanisms and their association with clinical outcomes cannot conclusively be stated, this pilot study demonstrates promising trends that encourages investigation into this relationship.
Collapse
Affiliation(s)
- Stewart W Ross
- Faculty of Engineering, University of Sydney, Sydney, Australia; ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, Australia.
| | - Josephine Malcolm
- Burns Unit, Concord Repatriation General Hospital, Sydney, Australia; ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, Australia
| | - Joanneke Maitz
- Burns Unit, Concord Repatriation General Hospital, Sydney, Australia; ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, Australia
| | - Zhe Li
- Burns Unit, Concord Repatriation General Hospital, Sydney, Australia; ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, Australia; Concord Clinical School, University of Sydney, Sydney, Australia
| | - Yiwei Wang
- Faculty of Engineering, University of Sydney, Sydney, Australia; Burns Unit, Concord Repatriation General Hospital, Sydney, Australia; ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, Australia
| | - Andrea C Issler-Fisher
- Burns Unit, Concord Repatriation General Hospital, Sydney, Australia; ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, Australia; Concord Clinical School, University of Sydney, Sydney, Australia
| |
Collapse
|
9
|
Kley M, Moser SO, Winter DV, Odermatt A. In vitro methods to assess 11β-hydroxysteroid dehydrogenase type 1 activity. Methods Enzymol 2023; 689:121-165. [PMID: 37802569 DOI: 10.1016/bs.mie.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
11β-Hydroxysteroid dehydrogenase type 1 (11β-HSD1) converts inactive 11-keto-glucocorticoids to their active 11β-hydroxylated forms. It also catalyzes the oxoreduction of other endogenous and exogenous substrates. The ubiquitously expressed 11β-HSD1 shows high levels in liver and other metabolically active tissues such as brain and adipose tissue. Pharmacological inhibition of 11β-HSD1 was found to ameliorate adverse metabolic effects of elevated glucocorticoids in rodents and humans, improve wound healing and delay skin aging, and enhance memory and cognition in rodent Alzheimer's disease models. Thus, there is an interest to develop 11β-HSD1 inhibitors for therapeutic purposes. This chapter describes in vitro methods to assess 11β-HSD1 enzyme activity for different purposes, be it in disease models, for the assessment of the kinetics of novel substrates or for the screening and characterization of inhibitors. 11β-HSD1 protein expression and preparations of the different biological samples are discussed first, followed by a description of a well-established and easily adaptable 11β-HSD1 enzyme activity assay. Finally, different readout methods are shortly described. This chapter should provide the reader with a toolbox of methods to assess 11β-HSD1 activity with instructions in the form of a decision tree for the choice and implementation of an appropriate enzyme activity assay.
Collapse
Affiliation(s)
- Manuel Kley
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland; Swiss Centre for Applied Human Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Seraina O Moser
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland; Swiss Centre for Applied Human Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Denise V Winter
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland; Swiss Centre for Applied Human Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| |
Collapse
|
10
|
Davidson CT, Miller E, Muir M, Dawson JC, Lee M, Aitken S, Serrels A, Webster SP, Homer NZM, Andrew R, Brunton VG, Hadoke PWF, Walker BR. 11β-HSD1 inhibition does not affect murine tumour angiogenesis but may exert a selective effect on tumour growth by modulating inflammation and fibrosis. PLoS One 2023; 18:e0255709. [PMID: 36940215 PMCID: PMC10027213 DOI: 10.1371/journal.pone.0255709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 12/05/2022] [Indexed: 03/21/2023] Open
Abstract
Glucocorticoids inhibit angiogenesis by activating the glucocorticoid receptor. Inhibition of the glucocorticoid-activating enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) reduces tissue-specific glucocorticoid action and promotes angiogenesis in murine models of myocardial infarction. Angiogenesis is important in the growth of some solid tumours. This study used murine models of squamous cell carcinoma (SCC) and pancreatic ductal adenocarcinoma (PDAC) to test the hypothesis that 11β-HSD1 inhibition promotes angiogenesis and subsequent tumour growth. SCC or PDAC cells were injected into female FVB/N or C57BL6/J mice fed either standard diet, or diet containing the 11β-HSD1 inhibitor UE2316. SCC tumours grew more rapidly in UE2316-treated mice, reaching a larger (P<0.01) final volume (0.158 ± 0.037 cm3) than in control mice (0.051 ± 0.007 cm3). However, PDAC tumour growth was unaffected. Immunofluorescent analysis of SCC tumours did not show differences in vessel density (CD31/alpha-smooth muscle actin) or cell proliferation (Ki67) after 11β-HSD1 inhibition, and immunohistochemistry of SCC tumours did not show changes in inflammatory cell (CD3- or F4/80-positive) infiltration. In culture, the growth/viability (assessed by live cell imaging) of SCC cells was not affected by UE2316 or corticosterone. Second Harmonic Generation microscopy showed that UE2316 reduced Type I collagen (P<0.001), whilst RNA-sequencing revealed that multiple factors involved in the innate immune/inflammatory response were reduced in UE2316-treated SCC tumours. 11β-HSD1 inhibition increases SCC tumour growth, likely via suppression of inflammatory/immune cell signalling and extracellular matrix deposition, but does not promote tumour angiogenesis or growth of all solid tumours.
Collapse
Affiliation(s)
- Callam T. Davidson
- BHF Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Eileen Miller
- BHF Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Morwenna Muir
- Cancer Research UK Edinburgh Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - John C. Dawson
- Cancer Research UK Edinburgh Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Martin Lee
- Cancer Research UK Edinburgh Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Stuart Aitken
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Alan Serrels
- Cancer Research UK Edinburgh Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Scott P. Webster
- BHF Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Natalie Z. M. Homer
- BHF Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Mass Spectrometry Core, Clinical Research Facility, University of Edinburgh, Edinburgh, United Kingdom
| | - Ruth Andrew
- BHF Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Valerie G. Brunton
- Cancer Research UK Edinburgh Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Patrick W. F. Hadoke
- BHF Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Brian R. Walker
- BHF Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Institute of Genetic Medicine, Newcastle University, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
11
|
Tsai KHY, Shi H, Parungao RJ, Naficy S, Ding X, Ding X, Hew JJ, Wang X, Chrzanowski W, Lavery GG, Li Z, Issler-Fisher AC, Chen J, Tan Q, Maitz PK, Cooper MS, Wang Y. Skin 11β-hydroxysteroid dehydrogenase type 1 enzyme expression regulates burn wound healing and can be targeted to modify scar characteristics. BURNS & TRAUMA 2023; 11:tkac052. [PMID: 36694861 PMCID: PMC9862341 DOI: 10.1093/burnst/tkac052] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/29/2022] [Accepted: 11/16/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Excessive scarring and fibrosis are the most severe and common complications of burn injury. Prolonged exposure to high levels of glucocorticoids detrimentally impacts on skin, leading to skin thinning and impaired wound healing. Skin can generate active glucocorticoids locally through expression and activity of the 11β-hydroxysteroid dehydrogenase type 1 enzyme (11β-HSD1). We hypothesised that burn injury would induce 11β-HSD1 expression and local glucocorticoid metabolism, which would have important impacts on wound healing, fibrosis and scarring. We additionally proposed that pharmacological manipulation of this system could improve aspects of post-burn scarring. METHODS Skin 11β-HSD1 expression in burns patients and mice was examined. The impacts of 11β-HSD1 mediating glucocorticoid metabolism on burn wound healing, scar formation and scar elasticity and quality were additionally examined using a murine 11β-HSD1 genetic knockout model. Slow-release scaffolds containing therapeutic agents, including active and inactive glucocorticoids, were developed and pre-clinically tested in mice with burn injury. RESULTS We demonstrate that 11β-HSD1 expression levels increased substantially in both human and mouse skin after burn injury. 11β-HSD1 knockout mice experienced faster wound healing than wild type mice but the healed wounds manifested significantly more collagen deposition, tensile strength and stiffness, features characteristic of excessive scarring. Application of slow-release prednisone, an inactive glucocorticoid, slowed the initial rate of wound closure but significantly reduced post-burn scarring via reductions in inflammation, myofibroblast generation, collagen production and scar stiffness. CONCLUSIONS Skin 11β-HSD1 expression is a key regulator of wound healing and scarring after burn injury. Application of an inactive glucocorticoid capable of activation by local 11β-HSD1 in skin slows the initial rate of wound closure but significantlyimproves scar characteristics post burn injury.
Collapse
Affiliation(s)
- Kevin H-Y Tsai
- Adrenal Steroid Group, ANZAC Research Institute, Concord Hospital, The University of Sydney, Sydney, NSW 2137, Australia
- Burns and Reconstructive Surgery Research Group, ANZAC Research Institute, Concord Hospital, The University of Sydney, Sydney, NSW 2137, Australia
| | - Huaikai Shi
- Burns and Reconstructive Surgery Research Group, ANZAC Research Institute, Concord Hospital, The University of Sydney, Sydney, NSW 2137, Australia
| | - Roxanne J Parungao
- Burns and Reconstructive Surgery Research Group, ANZAC Research Institute, Concord Hospital, The University of Sydney, Sydney, NSW 2137, Australia
| | - Sina Naficy
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, NSW 2006, Australia
| | - Xiaotong Ding
- Jiangsu Provincial Engineering Research Centre of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Xiaofeng Ding
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China
| | - Jonathan J Hew
- Burns and Reconstructive Surgery Research Group, ANZAC Research Institute, Concord Hospital, The University of Sydney, Sydney, NSW 2137, Australia
| | - Xiaosuo Wang
- Heart Research Institute, The University of Sydney, Sydney, NSW 2006 , Australia
| | - Wojciech Chrzanowski
- Sydney Nano Institute, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Gareth G Lavery
- Department of Biosciences, Centre for Healthy Ageing and Understanding Disease, Nottingham Trent University, NG1 4BU, UK
| | - Zhe Li
- Burns and Reconstructive Surgery Unit, Concord Repatriation General Hospital, Sydney, NSW 2137, Australia
| | - Andrea C Issler-Fisher
- Burns and Reconstructive Surgery Unit, Concord Repatriation General Hospital, Sydney, NSW 2137, Australia
| | - Jun Chen
- Jiangsu Provincial Engineering Research Centre of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Qian Tan
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China
| | - Peter K Maitz
- Burns and Reconstructive Surgery Research Group, ANZAC Research Institute, Concord Hospital, The University of Sydney, Sydney, NSW 2137, Australia
- Burns and Reconstructive Surgery Unit, Concord Repatriation General Hospital, Sydney, NSW 2137, Australia
| | | | | |
Collapse
|
12
|
Lv S, Shen Q, Li H, Chen Q, Xie W, Li Y, Wang X, Ding G. Caloric restriction delays age-related muscle atrophy by inhibiting 11β-HSD1 to promote the differentiation of muscle stem cells. Front Med (Lausanne) 2023; 9:1027055. [PMID: 36687405 PMCID: PMC9849809 DOI: 10.3389/fmed.2022.1027055] [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: 08/24/2022] [Accepted: 12/08/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction Calorie restriction (CR) is an important direction for the delay of sarcopenia in elderly individuals. However, the specific mechanisms of CR against aging are still unclear. Methods In this study, we used a CR model of elderly mice with muscle-specific 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1) knockout mice and 11β-HSD1 overexpression mice to confirm that CR can delay muscle aging by inhibiting 11β-HSD1 which can transform inactive GC(cortisone) into active GC(cortisol). The ability of self-proliferation and differentiation into muscle fibers of these mouse muscle stem cells (MuSCs) was observed in vitro. Additionally, the mitochondrial function and mitochondrial ATP production capacity of MuSCs were measured by mitochondrial oxygen consumption. Results It was found that the 11β-HSD1 expression level was increased in age-related muscle atrophy. Overexpression of 11β-HSD1 led to muscle atrophy in young mice, and 11β-HSD1 knockout rescued age-related muscle atrophy. Moreover, CR in aged mice reduced the local effective concentration of glucocorticoid (GC) through 11β-HSD1, thereby promoting the mitochondrial function and differentiation ability of MuSCs. Conclusions Together, our findings highlight promising sarcopenia protection with 40% CR in older ages. Furthermore, we speculated that targeting an 11β-HSD1-dependent metabolic pathway may represent a novel strategy for developing therapeutics against age-related muscle atrophy.
Collapse
Affiliation(s)
- Shan Lv
- Department of Geriatric Endocrinology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Qianjin Shen
- Department of Emergency Medicine, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hengzhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qun Chen
- Department of Orthopedics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Wenqing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China,*Correspondence: Yusheng Li,
| | - Xiaodong Wang
- Department of Geriatric Endocrinology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China,Xiaodong Wang,
| | - Guoxian Ding
- Department of Geriatric Endocrinology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China,Guoxian Ding,
| |
Collapse
|
13
|
Choi Y, Takasugi M, Takemura K, Yoshida Y, Kamiya T, Adachi J, Tsuruta D, Ohtani N. Characterization of Transcriptomic and Proteomic Changes in the Skin after Chronic Fluocinolone Acetonide Treatment. Biomolecules 2022; 12:biom12121822. [PMID: 36551249 PMCID: PMC9775701 DOI: 10.3390/biom12121822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
While topical corticosteroid (TCS) treatment is widely used for many skin diseases, it can trigger adverse side effects, and some of such effects can last for a long time after stopping the treatment. However, molecular changes induced by TCS treatment remain largely unexplored, although transient changes in histology and some major ECM components have been documented. Here, we investigated transcriptomic and proteomic changes induced by fluocinolone acetonide (FA) treatment in the mouse skin by conducting RNA-Seq and quantitative proteomics. Chronic FA treatment affected the expression of 4229 genes, where downregulated genes were involved in cell-cycle progression and ECM organization, and upregulated genes were involved in lipid metabolism. The effects of FA on transcriptome and histology of the skin largely returned to normal by two weeks after the treatment. Only a fraction of transcriptomic changes were reflected by proteomic changes, and the expression of 46 proteins was affected one day after chronic FA treatment. A comparable number of proteins were differentially expressed between control and FA-treated skin samples even at 15 and 30 days after stopping chronic FA treatment. Interestingly, proteins affected during and after chronic FA treatment were largely different. Our results provide fundamental information of molecular changes induced by FA treatment in the skin.
Collapse
Affiliation(s)
- Yongsu Choi
- Department of Pathophysiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan
- Department of Dermatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan
| | - Masaki Takasugi
- Department of Pathophysiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan
- Correspondence: (M.T.); (N.O.)
| | - Kazuaki Takemura
- Department of Pathophysiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan
| | - Yuya Yoshida
- Department of Pathophysiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan
| | - Tomonori Kamiya
- Department of Pathophysiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan
| | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki City 567-0085, Japan
| | - Daisuke Tsuruta
- Department of Dermatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan
| | - Naoko Ohtani
- Department of Pathophysiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan
- Correspondence: (M.T.); (N.O.)
| |
Collapse
|
14
|
A stem cell aging framework, from mechanisms to interventions. Cell Rep 2022; 41:111451. [DOI: 10.1016/j.celrep.2022.111451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/04/2022] [Accepted: 09/14/2022] [Indexed: 11/19/2022] Open
|
15
|
Tognetti L, Damiani F, Marrocco C, Mariotti G, Trovato E, Cinotti E, Marcolongo P, Pellegrino M, Rubegni P. Evaluation of 11-β hydroxysteroid dehydrogenase type 1 in cutaneous fibroblasts cultures of psoriatic lesional skin before and after narrow band-UVB phototherapy. PHOTODERMATOLOGY, PHOTOIMMUNOLOGY & PHOTOMEDICINE 2022; 38:391-394. [PMID: 34743342 PMCID: PMC9541928 DOI: 10.1111/phpp.12752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/29/2021] [Accepted: 11/04/2021] [Indexed: 06/13/2023]
Affiliation(s)
- Linda Tognetti
- Department of ClinicalDermatology Unit and Skin Bank UnitSurgical and NeurosciencesUniversity of SienaSienaItaly
| | - Francesco Damiani
- Department of Molecular and Developmental MedicineUniversity of SienaSienaItaly
| | - Camilla Marrocco
- Department of ClinicalDermatology Unit and Skin Bank UnitSurgical and NeurosciencesUniversity of SienaSienaItaly
| | - Giancarlo Mariotti
- Department of ClinicalDermatology Unit and Skin Bank UnitSurgical and NeurosciencesUniversity of SienaSienaItaly
| | - Emanuele Trovato
- Department of ClinicalDermatology Unit and Skin Bank UnitSurgical and NeurosciencesUniversity of SienaSienaItaly
| | - Elisa Cinotti
- Department of ClinicalDermatology Unit and Skin Bank UnitSurgical and NeurosciencesUniversity of SienaSienaItaly
| | - Paola Marcolongo
- Department of Molecular and Developmental MedicineUniversity of SienaSienaItaly
| | | | - Pietro Rubegni
- Department of ClinicalDermatology Unit and Skin Bank UnitSurgical and NeurosciencesUniversity of SienaSienaItaly
| |
Collapse
|
16
|
Abbas A, Schini M, Ainsworth G, Brown SR, Oughton J, Crowley RK, Cooper MS, Fairclough RJ, Eastell R, Stewart PM. Effect of AZD4017, a Selective 11β-HSD1 Inhibitor, on Bone Turnover Markers in Postmenopausal Osteopenia. J Clin Endocrinol Metab 2022; 107:2026-2035. [PMID: 35275196 PMCID: PMC9202729 DOI: 10.1210/clinem/dgac100] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT The causative link between circulating glucocorticoid excess and osteoporosis is well-established. The enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), which increases local cortisol production, is expressed in human osteoblasts and its activity increases with age. OBJECTIVE We hypothesized that local 11β-HSD1 might mediate an age-related decrease in bone formation and that selective 11β-HSD1 inhibition may enhance bone formation. METHODS A dual-center, phase II, randomized, double-blind, placebo-controlled trial of 90 days' treatment with AZD4017 (a selective 11β-HSD1 inhibitor) was conducted in 55 postmenopausal women with osteopenia. Participants received 400 mg oral AZD4017 twice daily vs matched placebo over 90 days. The primary outcome measure was the impact on the bone formation marker osteocalcin. Secondary objectives included correlation with 11β-HSD1 activity. RESULTS At 90 days, osteocalcin levels did not differ between treatment groups: active (mean 22.3 [SD 8.6] ng/mL, n = 22) and placebo (21.7 [SD 9.2] ng/mL, n = 24), with a baseline-adjusted treatment effect of 0.95 (95% CI: -2.69, 4.60). The results from the urinary [THF + alloTHF]/THE ratio (index of 11β-HSD1 activity) and the urinary cortisol/cortisone ratio (index of 11β-HSD2 activity) confirmed a > 90% inhibition of 11β-HSD1 but no change in activity of 11β-HSD2. CONCLUSION This trial demonstrates that AZD4017 selectively inhibits 11β-HSD1 activity in vivo in a safe and reversible manner. Following 90 days of treatment, there is no effect on bone formation, indicating that the relative impairment of bone mineral density in postmenopausal women is not mediated by local intracellular production of cortisol under normal physiological concentrations.
Collapse
Affiliation(s)
- Afroze Abbas
- Faculty of Medicine and Health, University of Leeds, and Leeds Teaching Hospitals NHS Trust, Leeds LS9 7TF, UK
| | - Marian Schini
- Academic Unit of Bone Metabolism, University of Sheffield, Sheffield S5 7AU, UK
| | - Gemma Ainsworth
- Clinical Trials Research Unit, University of Leeds, Leeds LS2 9JT, UK
| | - Sarah R Brown
- Clinical Trials Research Unit, University of Leeds, Leeds LS2 9JT, UK
| | - Jamie Oughton
- Clinical Trials Research Unit, University of Leeds, Leeds LS2 9JT, UK
| | - Rachel K Crowley
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Mark S Cooper
- Concord Clinical School, Faculty of Medicine & Health, University of Sydney, NSW 2139, Australia
| | - Rebecca J Fairclough
- Emerging Innovations Unit, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Richard Eastell
- Academic Unit of Bone Metabolism, University of Sheffield, Sheffield S5 7AU, UK
| | - Paul M Stewart
- Faculty of Medicine and Health, University of Leeds, and Leeds Teaching Hospitals NHS Trust, Leeds LS9 7TF, UK
| |
Collapse
|
17
|
Téblick A, Van Dyck L, Van Aerde N, Van der Perre S, Pauwels L, Derese I, Debaveye Y, Wouters PJ, Vanhorebeek I, Langouche L, Van den Berghe G. Impact of duration of critical illness and level of systemic glucocorticoid availability on tissue-specific glucocorticoid receptor expression and actions: A prospective, observational, cross-sectional human and two translational mouse studies. EBioMedicine 2022; 80:104057. [PMID: 35584557 PMCID: PMC9117556 DOI: 10.1016/j.ebiom.2022.104057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/19/2022] [Accepted: 04/27/2022] [Indexed: 12/24/2022] Open
Abstract
Background Reduced glucocorticoid-receptor (GR) expression in blood suggested that critically ill patients become glucocorticoid-resistant necessitating stress-doses of glucocorticoids. We hypothesised that critical illness evokes a tissue-specific, time-dependent expression of regulators of GR-action which adaptively guides glucocorticoid action to sites of need. Methods We performed a prospective, observational, cross-sectional human study and two translational mouse studies. In freshly-isolated neutrophils and monocytes and in skeletal muscle and subcutaneous adipose tissue of 137 critically ill patients and 20 healthy controls and in skeletal muscle and adipose tissue as well as in vital tissues (heart, lung, diaphragm, liver, kidney) of 88 septic and 26 healthy mice, we quantified gene expression of cortisone-reductase 11β-HSD1, glucocorticoid-receptor-isoforms GRα and GRβ, GRα-sensitivity-regulating-co-chaperone FKBP51, and GR-action-marker GILZ. Expression profiles were compared in relation to illness-duration and systemic-glucocorticoid-availability. Findings In patients’ neutrophils, GRα and GILZ were substantially suppressed (p≤0·05) throughout intensive care unit (ICU)-stay, while in monocytes low/normal GRα coincided with increased GILZ (p≤0·05). FKBP51 was increased transiently (neutrophils) or always (monocytes,p≤0·05). In patients’ muscle, 11β-HSD1 and GRα were low-normal (p≤0·05) and substantially suppressed in adipose tissue (p≤0·05); FKBP51 and GILZ were increased in skeletal muscle (p≤0·05) but normal in adipose tissue. GRβ was undetectable. Increasing systemic glucocorticoid availability in patients independently associated with further suppressed muscle 11β-HSD1 and GRα, further increased FKBP51 and unaltered GILZ (p≤0·05). In septic mouse heart and lung, 11β-HSD1, FKBP51 and GILZ were always high (p≤0·01). In heart, GRα was suppressed (p≤0·05), while normal or high in lung (all p≤0·05). In diaphragm, 11β-HSD1 was high/normal, GRα low/normal and FKBP51 and GILZ high (p≤0·01). In kidney, 11β-HSD1 transiently increased but decreased thereafter, GRα was normal and FKBP51 and GILZ high (p≤0·01). In liver, 11β-HSD1 was suppressed (p≤0·01), GRα normal and FKBP51 high (p≤0·01) whereas GILZ was transiently decreased but elevated thereafter (p≤0·05). Only in lung and diaphragm, treatment with hydrocortisone further increased GILZ. Interpretation Tissue-specific, time-independent adaptations to critical illness guided GR-action predominantly to vital tissues such as lung, while (partially) protecting against collateral harm in other cells and tissues, such as neutrophils. These findings argue against maladaptive generalised glucocorticoid-resistance necessitating glucocorticoid-treatment. Funding Research-Foundation-Flanders, Methusalem-Program-Flemish-Government, European-Research-Council, European-Respiratory-Society.
Collapse
Affiliation(s)
- Arno Téblick
- Clinical division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Lisa Van Dyck
- Clinical division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Nathalie Van Aerde
- Clinical division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Sarah Van der Perre
- Clinical division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Lies Pauwels
- Clinical division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Inge Derese
- Clinical division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Yves Debaveye
- Clinical division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Pieter J Wouters
- Clinical division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Ilse Vanhorebeek
- Clinical division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Lies Langouche
- Clinical division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Greet Van den Berghe
- Clinical division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
| |
Collapse
|
18
|
Ajjan RA, Hensor EMA, Del Galdo F, Shams K, Abbas A, Fairclough RJ, Webber L, Pegg L, Freeman A, Taylor AE, Arlt W, Morgan AW, Tahrani AA, Stewart PM, Russell DA, Tiganescu A. Oral 11β-HSD1 inhibitor AZD4017 improves wound healing and skin integrity in adults with type 2 diabetes mellitus: a pilot randomized controlled trial. Eur J Endocrinol 2022; 186:441-455. [PMID: 35113805 PMCID: PMC8942338 DOI: 10.1530/eje-21-1197] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/03/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Chronic wounds (e.g. diabetic foot ulcers) reduce the quality of life, yet treatments remain limited. Glucocorticoids (activated by the enzyme 11β-hydroxysteroid dehydrogenase type 1, 11β-HSD1) impair wound healing. OBJECTIVES Efficacy, safety, and feasibility of 11β-HSD1 inhibition for skin function and wound healing. DESIGN Investigator-initiated, double-blind, randomized, placebo-controlled, parallel-group phase 2b pilot trial. METHODS Single-center secondary care setting. Adults with type 2 diabetes mellitus without foot ulcers were administered 400 mg oral 11β-HSD1 inhibitor AZD4017 (n = 14) or placebo (n = 14) bi-daily for 35 days. Participants underwent 3-mm full-thickness punch skin biopsies at baseline and on day 28; wound healing was monitored after 2 and 7 days. Computer-generated 1:1 randomization was pharmacy-administered. Analysis was descriptive and focused on CI estimation. Of the 36 participants screened, 28 were randomized. RESULTS Exploratory proof-of-concept efficacy analysis suggested AZD4017 did not inhibit 24-h ex vivoskin 11β-HSD1 activity (primary outcome; difference in percentage conversion per 24 h 1.1% (90% CI: -3.4 to 5.5) but reduced systemic 11β-HSD1 activity by 87% (69-104%). Wound diameter was 34% (7-63%) smaller with AZD4017 at day 2, and 48% (12-85%) smaller after repeat wounding at day 30. AZD4017 improved epidermal integrity but modestly impaired barrier function. Minimal adverse events were comparable to placebo. Recruitment rate, retention, and data completeness were 2.9/month, 27/28, and 95.3%, respectively. CONCLUSION A phase 2 trial is feasible, and preliminary proof-of-concept data suggests AZD4017 warrants further investigation in conditions of delayed healing, for example in diabetic foot ulcers. SIGNIFICANCE STATEMENT Stress hormone activation by the enzyme 11β-HSD type 1 impairs skin function (e.g. integrity) and delays wound healing in animal models of diabetes, but effects in human skin were previously unknown. Skin function was evaluated in response to treatment with a 11β-HSD type 1 inhibitor (AZD4017), or placebo, in people with type 2 diabetes. Importantly, AZD4017 was safe and well tolerated. This first-in-human randomized, controlled, clinical trial found novel evidence that 11β-HSD type 1 regulates skin function in humans, including improved wound healing, epidermal integrity, and increased water loss. Results warrant further studies in conditions of impaired wound healing, for example, diabetic foot ulcers to evaluate 11β-HSD type 1 as a novel therapeutic target forchronic wounds.
Collapse
Affiliation(s)
- R A Ajjan
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - E M A Hensor
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Center, Leeds Teaching Hospitals, NHS Trust, Leeds, UK
| | - F Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Center, Leeds Teaching Hospitals, NHS Trust, Leeds, UK
| | - K Shams
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Center, Leeds Teaching Hospitals, NHS Trust, Leeds, UK
| | - A Abbas
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - R J Fairclough
- Emerging Innovations Unit, Discovery Sciences, BioPharmaceuticals R&D
| | - L Webber
- Emerging Portfolio Development, Late Oncology, Oncology R&D, AstraZeneca, Cambridge, UK
| | - L Pegg
- Emerging Portfolio Development, Late Oncology, Oncology R&D, AstraZeneca, Cambridge, UK
| | - A Freeman
- Emerging Innovations Unit, Discovery Sciences, BioPharmaceuticals R&D
| | - A E Taylor
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - W Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - A W Morgan
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Center, Leeds Teaching Hospitals, NHS Trust, Leeds, UK
| | - A A Tahrani
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - P M Stewart
- NIHR Leeds Biomedical Research Center, Leeds Teaching Hospitals, NHS Trust, Leeds, UK
- Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - D A Russell
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- Leeds Vascular Institute, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - A Tiganescu
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Center, Leeds Teaching Hospitals, NHS Trust, Leeds, UK
- Correspondence should be addressed to A Tiganescu;
| |
Collapse
|
19
|
Radstake WE, Baselet B, Baatout S, Verslegers M. Spaceflight Stressors and Skin Health. Biomedicines 2022; 10:364. [PMID: 35203572 PMCID: PMC8962330 DOI: 10.3390/biomedicines10020364] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/28/2022] [Accepted: 01/31/2022] [Indexed: 02/06/2023] Open
Abstract
Traveling to space puts astronauts at risk of developing serious health problems. Of particular interest is the skin, which is vitally important in protecting the body from harmful environmental factors. Although data obtained from long-duration spaceflight studies are inconsistent, there have been indications of increased skin sensitivity and signs of dermal atrophy in astronauts. To better understand the effects of spaceflight stressors including microgravity, ionizing radiation and psychological stress on the skin, researchers have turned to in vitro and in vivo simulation models mimicking certain aspects of the spaceflight environment. In this review, we provide an overview of these simulation models and highlight studies that have improved our understanding on the effect of simulation spaceflight stressors on skin function. Data show that all aforementioned spaceflight stressors can affect skin health. Nevertheless, there remains a knowledge gap regarding how different spaceflight stressors in combination may interact and affect skin health. In future, efforts should be made to better simulate the spaceflight environment and reduce uncertainties related to long-duration spaceflight health effects.
Collapse
Affiliation(s)
- Wilhelmina E. Radstake
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400 Mol, Belgium; (W.E.R.); (S.B.); (M.V.)
- Department of Molecular Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Bjorn Baselet
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400 Mol, Belgium; (W.E.R.); (S.B.); (M.V.)
| | - Sarah Baatout
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400 Mol, Belgium; (W.E.R.); (S.B.); (M.V.)
- Department of Molecular Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Mieke Verslegers
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400 Mol, Belgium; (W.E.R.); (S.B.); (M.V.)
| |
Collapse
|
20
|
A novel mineralocorticoid receptor antagonist, 7,3',4'-trihydroxyisoflavone improves skin barrier function impaired by endogenous or exogenous glucocorticoids. Sci Rep 2021; 11:11920. [PMID: 34099793 PMCID: PMC8184959 DOI: 10.1038/s41598-021-91450-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 05/10/2021] [Indexed: 11/08/2022] Open
Abstract
Excess glucocorticoids (GCs) with either endogenous or exogenous origins deteriorate skin barrier function. GCs bind to mineralocorticoid and GC receptors (MRs and GRs) in normal human epidermal keratinocytes (NHEKs). Inappropriate MR activation by GCs mediates various GC-induced cutaneous adverse events. We examined whether MR antagonists can ameliorate GC-mediated skin barrier dysfunction in NHEKs, reconstructed human epidermis (RHE), and subjects under psychological stress (PS). In a preliminary clinical investigation, topical MR antagonists improved skin barrier function in topical GC-treated subjects. In NHEKs, cortisol induced nuclear translocation of GR and MR, and GR and MR antagonists inhibited cortisol-induced reductions of keratinocyte differentiation. We identified 7,3',4'-trihydroxyisoflavone (7,3',4'-THIF) as a novel compound that inhibits MR transcriptional activity by screening 30 cosmetic compounds. 7,3',4'-THIF ameliorated the cortisol effect which decreases keratinocyte differentiation in NHEKs and RHE. In a clinical study on PS subjects, 7,3',4'-THIF (0.1%)-containing cream improved skin barrier function, including skin surface pH, barrier recovery rate, and stratum corneum lipids. In conclusion, skin barrier dysfunction owing to excess GC is mediated by MR and GR; thus, it could be prevented by treatment with MR antagonists. Therefore, topical MR antagonists are a promising therapeutic option for skin barrier dysfunction after topical GC treatment or PS.
Collapse
|
21
|
Li X, Hu S, Zhu Q, Yao G, Yao J, Li J, Wang Y, Ding Y, Qi J, Xu R, Zhao H, Zhu Z, Du Y, Sun K, Sun Y. Addressing the role of 11β-hydroxysteroid dehydrogenase type 1 in the development of polycystic ovary syndrome and the putative therapeutic effects of its selective inhibition in a preclinical model. Metabolism 2021; 119:154749. [PMID: 33722534 DOI: 10.1016/j.metabol.2021.154749] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 02/20/2021] [Accepted: 03/02/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is the most common metabolic and endocrine disorder among reproductive-age women, and the leading cause of anovulatory infertility. 11β-hydroxysteroid dehydrogenases-1 (11β-HSD1) catalysing the conversion of inactive cortisone to active cortisol plays a crucial role in various metabolic diseases. However, whether 11β-HSD1 is associated with the pathogenesis of PCOS and whether 11β-HSD1 can be a treating target of PCOS remain unknown. METHODS This study was first designed to explore the role of 11β-HSD1 in PCOS development and the effect of selective 11β-HSD1 inhibitor administration on PCOS treatment. Follicular fluid and granulosa cells (GCs) were collected from 32 non-PCOS patients and 37 patients with PCOS to measure cortisol and 11β-HSDs levels. Female Sprague-Dawley rats (3-week-old) were injected with dehydroepiandrosterone (DHEA) to induce PCOS and their ovaries were collected to measure the abundance of corticosterone (CORT) and 11β-HSDs. To determine the role of 11β-HSD1 in PCOS development, we overexpressed 11β-HSD1 in the ovaries of female rats (5-week-old) or knocked down the expression of 11β-HSD1 in the ovaries from PCOS rats via lentivirus injection. After lentivirus infection, the body weights, ovarian weights, estrous cycles, reproductive hormones and morphology of the ovary were analysed in rats from different experimental groups. Then to figure out the translational potential of the selective 11β-HSD1 inhibitor in treating PCOS, PCOS rats were treated with BVT.2733, a selective 11β-HSD1 inhibitor and a cluster of PCOS-like traits were analysed, including insulin sensitivity, ovulatory function and fertility of rats from the Control, PCOS and PCOS+BVT groups. Rat ovarian explants and human GCs were used to explore the effect of CORT or cortisol on ovarian extracellular matrix remodelling. RESULTS The elevated expression of 11β-HSD1 contributed to the increased cortisol and corticosterone (CORT) concentrations observed in the ovaries of PCOS patients and PCOS rats respectively. Our results showed that ovarian overexpression of 11β-HSD1 induced a cluster of PCOS phenotypes in rats including irregular estrous cycles, reproductive hormone dysfunction and polycystic ovaries. While knockdown of ovarian 11β-HSD1 of PCOS rats reversed these PCOS-like changes. Additionally, the selective 11β-HSD1 inhibitor BVT.2733 alleviated PCOS symptoms such as insulin resistance (IR), irregular estrous cycles, reproductive hormone dysfunction, polycystic ovaries, ovulatory dysfunction and subfertility. Moreover, we showed that cortisol target ovarian insulin signalling pathway and ovarian extracellular matrix (ECM) remodelling in vivo, in ovarian explants and in GCs. CONCLUSION Elevated 11β-HSD1 abundance in ovarian is involved in the pathogenesis of PCOS by impairing insulin signalling pathway and ECM remodelling. Selective inhibition of 11β-HSD1 ameliorates a cluster of PCOS phenotypes. Our study demonstrates the selective 11β-HSD1 inhibitor as a novel and promising strategy for the treatment of PCOS.
Collapse
Affiliation(s)
- Xinyu Li
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Shuanggang Hu
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Qinling Zhu
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Guangxin Yao
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Jufang Yao
- Animal Laboratory, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
| | - Jiaxing Li
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Yuan Wang
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Ying Ding
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Jia Qi
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Rui Xu
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Hanting Zhao
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Zhenyi Zhu
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Yanzhi Du
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Kang Sun
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Yun Sun
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China.
| |
Collapse
|
22
|
Kim BJ, Lee NR, Lee CH, Lee YB, Choe SJ, Lee S, Hwang HJ, Kim E, Lavery GG, Shin KO, Park K, Choi EH. Increased Expression of 11β-Hydroxysteroid Dehydrogenase Type 1 Contributes to Epidermal Permeability Barrier Dysfunction in Aged Skin. Int J Mol Sci 2021; 22:ijms22115750. [PMID: 34072239 PMCID: PMC8198579 DOI: 10.3390/ijms22115750] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
Inactive cortisone is converted into active cortisol by 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1). Excessive levels of active glucocorticoids could deteriorate skin barrier function; barrier impairment is also observed in aged skin. In this study, we aimed to determine whether permeability barrier impairment in the aged skin could be related to increased 11β-HSD1 expression. Aged humans (n = 10) showed increased cortisol in the stratum corneum (SC) and oral epithelium, compared to young subjects (n = 10). 11β-HSD1 expression (as assessed via immunohistochemical staining) was higher in the aged murine skin. Aged hairless mice (56-week-old, n = 5) manifested greater transepidermal water loss, lower SC hydration, and higher levels of serum inflammatory cytokines than the young mice (8-week-old, n = 5). Aged 11β-HSD1 knockout mice (n = 11), 11β-HSD1 inhibitor (INHI)-treated aged wild type (WT) mice (n = 5) and young WT mice (n = 10) exhibited reduced SC corticosterone level. Corneodesmosome density was low in WT aged mice (n = 5), but high in aged 11β-HSD1 knockout and aged INHI-treated WT mice. Aged mice exhibited lower SC lipid levels; this effect was reversed by INHI treatment. Therefore, upregulation of 11β-HSD1 in the aged skin increases the active-glucocorticoid levels; this suppresses SC lipid biosynthesis, leading to impaired epidermal permeability barrier.
Collapse
Affiliation(s)
- Beom Jun Kim
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju 26426, Korea; (B.J.K.); (N.R.L.); (C.H.L.); (Y.B.L.); (S.J.C.); (S.L.); (H.J.H.); (E.K.)
| | - Noo Ri Lee
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju 26426, Korea; (B.J.K.); (N.R.L.); (C.H.L.); (Y.B.L.); (S.J.C.); (S.L.); (H.J.H.); (E.K.)
| | - Chung Hyeok Lee
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju 26426, Korea; (B.J.K.); (N.R.L.); (C.H.L.); (Y.B.L.); (S.J.C.); (S.L.); (H.J.H.); (E.K.)
| | - Young Bin Lee
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju 26426, Korea; (B.J.K.); (N.R.L.); (C.H.L.); (Y.B.L.); (S.J.C.); (S.L.); (H.J.H.); (E.K.)
| | - Sung Jay Choe
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju 26426, Korea; (B.J.K.); (N.R.L.); (C.H.L.); (Y.B.L.); (S.J.C.); (S.L.); (H.J.H.); (E.K.)
| | - Solam Lee
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju 26426, Korea; (B.J.K.); (N.R.L.); (C.H.L.); (Y.B.L.); (S.J.C.); (S.L.); (H.J.H.); (E.K.)
| | - Hyun Jee Hwang
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju 26426, Korea; (B.J.K.); (N.R.L.); (C.H.L.); (Y.B.L.); (S.J.C.); (S.L.); (H.J.H.); (E.K.)
| | - Eunjung Kim
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju 26426, Korea; (B.J.K.); (N.R.L.); (C.H.L.); (Y.B.L.); (S.J.C.); (S.L.); (H.J.H.); (E.K.)
| | - Gareth G. Lavery
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK;
| | - Kyong-Oh Shin
- Department of Food Science and Nutrition, Convergence Program of Materials Science for Medicine and Pharmaceutics, Hallym University, Chuncheon 24252, Korea; (K.-O.S.); (K.P.)
| | - Kyungho Park
- Department of Food Science and Nutrition, Convergence Program of Materials Science for Medicine and Pharmaceutics, Hallym University, Chuncheon 24252, Korea; (K.-O.S.); (K.P.)
| | - Eung Ho Choi
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju 26426, Korea; (B.J.K.); (N.R.L.); (C.H.L.); (Y.B.L.); (S.J.C.); (S.L.); (H.J.H.); (E.K.)
- Correspondence: ; Tel.: +82-33-748-2650
| |
Collapse
|
23
|
Chae M, Bae IH, Lim S, Jung K, Roh J, Kim W. AP Collagen Peptides Prevent Cortisol-Induced Decrease of Collagen Type I in Human Dermal Fibroblasts. Int J Mol Sci 2021; 22:ijms22094788. [PMID: 33946465 PMCID: PMC8125628 DOI: 10.3390/ijms22094788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 11/16/2022] Open
Abstract
Cortisol is an endogenous glucocorticoid (GC) and primary stress hormone that regulates a wide range of stress responses in humans. The adverse effects of cortisol on the skin have been extensively documented but the underlying mechanism of cortisol-induced signaling is still unclear. In the present study, we investigate the effect of cortisol on collagen type I expression and the effect of AP collagen peptides, collagen tripeptide-rich hydrolysates containing 3% glycine-proline- hydroxyproline (Gly-Pro-Hyp, GPH) from the fish skin, on the cortisol-mediated inhibition of collagen type I and the cortisol-induced signaling that regulates collagen type I production in human dermal fibroblasts (HDFs). We determine that cortisol downregulates the expression of collagen type I. AP collagen peptides or GC receptor (GR) inhibitors recover the cortisol-mediated inhibition of collagen type I and GR activation. AP collagen peptides or GR inhibitors also prevent the cortisol-dependent inhibition of transforming growth factor (TGF)-β signaling. AP collagen peptides or GR inhibitors are effective in the prevention of collagen type I inhibition mediated by cortisol in senescent HDFs and reconstituted human skin models. Taken together, GR signaling might be responsible for the cortisol-mediated inhibition of TGF-β. AP collagen peptides act as GR-mediated signaling blockers, preventing the cortisol-dependent inhibition of collagen type I. Therefore, AP collagen peptides have the potential to improve skin health.
Collapse
|
24
|
Gomez-Sanchez EP, Gomez-Sanchez CE. 11β-hydroxysteroid dehydrogenases: A growing multi-tasking family. Mol Cell Endocrinol 2021; 526:111210. [PMID: 33607268 PMCID: PMC8108011 DOI: 10.1016/j.mce.2021.111210] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/02/2021] [Accepted: 02/07/2021] [Indexed: 02/06/2023]
Abstract
This review briefly addresses the history of the discovery and elucidation of the three cloned 11β-hydroxysteroid dehydrogenase (11βHSD) enzymes in the human, 11βHSD1, 11βHSD2 and 11βHSD3, an NADP+-dependent dehydrogenase also called the 11βHSD1-like dehydrogenase (11βHSD1L), as well as evidence for yet identified 11βHSDs. Attention is devoted to more recently described aspects of this multi-functional family. The importance of 11βHSD substrates other than glucocorticoids including bile acids, 7-keto sterols, neurosteroids, and xenobiotics is discussed, along with examples of pathology when functions of these multi-tasking enzymes are disrupted. 11βHSDs modulate the intracellular concentration of glucocorticoids, thereby regulating the activation of the glucocorticoid and mineralocorticoid receptors, and 7β-27-hydroxycholesterol, an agonist of the retinoid-related orphan receptor gamma (RORγ). Key functions of this nuclear transcription factor include regulation of immune cell differentiation, cytokine production and inflammation at the cell level. 11βHSD1 expression and/or glucocorticoid reductase activity are inappropriately increased with age and in obesity and metabolic syndrome (MetS). Potential causes for disappointing results of the clinical trials of selective inhibitors of 11βHSD1 in the treatment of these disorders are discussed, as well as the potential for more targeted use of inhibitors of 11βHSD1 and 11βHSD2.
Collapse
Affiliation(s)
| | - Celso E Gomez-Sanchez
- Department of Pharmacology and Toxicology, Jackson, MS, USA; Medicine (Endocrinology), Jackson, MS, USA; University of Mississippi Medical Center and G.V. (Sonny) Montgomery VA Medical Center(3), Jackson, MS, USA
| |
Collapse
|
25
|
Ajjan R, Hensor EM, Shams K, Del Galdo F, Abbas A, Woods J, Fairclough RJ, Webber L, Pegg L, Freeman A, Morgan A, Stewart PM, Taylor AE, Arlt W, Tahrani A, Russell D, Tiganescu A. A randomised controlled pilot trial of oral 11β-HSD1 inhibitor AZD4017 for wound healing in adults with type 2 diabetes mellitus.. [DOI: 10.1101/2021.03.23.21254200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
AbstractChronic wounds (e.g. diabetic foot ulcers) have a major impact on quality of life, yet treatments remain limited. Glucocorticoids impair wound healing; preclinical research suggests that blocking glucocorticoid activation by the enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) improves wound repair. This investigator-initiated double-blind, randomised, placebo-controlled parallel-group phase 2b pilot trial investigated efficacy, safety and feasibility of 11β-HSD1 inhibition for 35 days by oral AZD4017 (AZD) treatment in adults with type 2 diabetes (n=14) compared to placebo (PCB, n=14) in a single-centre secondary care setting. Computer-generated 1:1 randomisation was pharmacy-administered. From 300 screening invitations, 36 attended, 28 were randomised. There was no proof-of-concept that AZD inhibited 24 hour skin 11β-HSD1 activity at day 28 (primary outcome: adjusted difference AZD-PCB 90% CI (diffCI)=-3.4,5.5) but systemic 11β-HSD1 activity (median urinary [THF+alloTHF]/THE ratio) was 87% lower with AZD at day 35 (PCB 1.00, AZD 0.13, diffCI=-1.04,-0.69). Mean wound gap diameter (mm) following baseline 2mm punch biopsy was 34% smaller at day 2 (PCB 1.51, AZD 0.98, diffCI=-0.95,-0.10) and 48% smaller after repeat wounding at day 30 (PCB 1.35, AZD 0.70, diffCI=-1.15,-0.16); results also suggested greater epidermal integrity but modestly impaired barrier function with AZD. AZD was well-tolerated with minimal side effects and comparable adverse events between treatments. Staff availability restricted recruitment (2.9/month); retention (27/28) and data completeness (95.3%) were excellent. These preliminary findings suggest that AZD may improve wound healing in patients with type 2 diabetes and warrant a fully-powered trial in patients with active ulcers. [Trial Registry: www.isrctn.com/ISRCTN74621291.FundingMRC Confidence in Concept and NIHR Senior Investigator Award.]Single Sentence SummaryAZD4017 was safe; data suggested improved skin healing / integrity, and modestly reduced epidermal barrier function in patients with type 2 diabetes.Disclosure SummaryI certify that neither I nor my co-authors have a conflict of interest as described above that is relevant to the subject matter or materials included in this Work.
Collapse
|
26
|
Beck KR, Odermatt A. Antifungal therapy with azoles and the syndrome of acquired mineralocorticoid excess. Mol Cell Endocrinol 2021; 524:111168. [PMID: 33484741 DOI: 10.1016/j.mce.2021.111168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 12/31/2020] [Accepted: 01/06/2021] [Indexed: 10/22/2022]
Abstract
The syndromes of mineralocorticoid excess describe a heterogeneous group of clinical manifestations leading to endocrine hypertension, typically either through direct activation of mineralocorticoid receptors or indirectly by impaired pre-receptor enzymatic regulation or through disturbed renal sodium homeostasis. The phenotypes of these disorders can be caused by inherited gene variants and somatic mutations or may be acquired upon exposures to exogenous substances. Regarding the latter, the symptoms of an acquired mineralocorticoid excess have been reported during treatment with azole antifungal drugs. The current review describes the occurrence of mineralocorticoid excess particularly during the therapy with posaconazole and itraconazole, addresses the underlying mechanisms as well as inter- and intra-individual differences, and proposes a therapeutic drug monitoring strategy for these two azole antifungals. Moreover, other therapeutically used azole antifungals and ongoing efforts to avoid adverse mineralocorticoid effects of azole compounds are shortly discussed.
Collapse
Affiliation(s)
- Katharina R Beck
- Swiss Centre for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Alex Odermatt
- Swiss Centre for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| |
Collapse
|
27
|
Inhibitors of 11β-Hydroxysteroid Dehydrogenase Type 1 as Potential Drugs for Type 2 Diabetes Mellitus—A Systematic Review of Clinical and In Vivo Preclinical Studies. Sci Pharm 2021. [DOI: 10.3390/scipharm89010005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Diabetes mellitus is a pathology with increasing frequency in society, being one of the main causes of death worldwide. For this reason, new therapeutic targets have been studied over the years. 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) is an enzyme responsible for reducing cortisone to its active form cortisol, which can lead to metabolic changes such as insulin resistance and hyperglycemia. Therefore, 11β-HSD1 inhibition may offer a new therapeutic approach for type 2 diabetes mellitus. This work intends to systematically review the available scientific evidence on this subject. For this, a search was conducted in three databases and 15 clinical and in vivo preclinical studies were included in this review. Despite the high inhibitory and selectivity levels achieved with several molecules and the demonstrated clinical efficacy in diabetes treatment, no phase III clinical trials have yet been conducted. This is important because the long-term effects of 11β-HSD1 inhibitors including the consequences in hypothalamic–pituitary–adrenal axis must be evaluated. However, this enzyme remains a promising target for drug development, including due to its effectiveness in controlling various factors that constitute the metabolic syndrome and its potential for multiple indications in patients with diabetes, including wound healing and weight loss.
Collapse
|
28
|
Brazel CB, Simon JC, Tuckermann JP, Saalbach A. Inhibition of 11β-HSD1 Expression by Insulin in Skin: Impact for Diabetic Wound Healing. J Clin Med 2020; 9:jcm9123878. [PMID: 33260645 PMCID: PMC7760287 DOI: 10.3390/jcm9123878] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 01/20/2023] Open
Abstract
Chronic, non-healing wounds impose a great burden on patients, professionals and health care systems worldwide. Diabetes mellitus (DM) and obesity are globally highly prevalent metabolic disorders and increase the risk for developing chronic wounds. Glucocorticoids (GCs) are endogenous stress hormones that exert profound effects on inflammation and repair systems. 11-beta-hydroxysteroid dehydrogenase 1 (11β-HSD1) is the key enzyme which controls local GC availability in target tissues such as skin. Since treatment with GCs has detrimental side effects on skin integrity, causing atrophy and delayed wound healing, we asked whether the dysregulated expression of 11β-HSD1 and consequently local GC levels in skin contribute to delayed wound healing in obese, diabetic db/db mice. We found increased expression of 11β-HSD1 during disturbed wound healing and in the healthy skin of obese, diabetic db/db mice. Cell analysis revealed increased expression of 11β-HSD1 in fibroblasts, myeloid cells and dermal white adipose tissue from db/db mice, while expression in keratinocytes was unaffected. Among diabetes- and obesity-related factors, insulin and insulin-like growth factor 1 down-regulated 11β-HSD1 expression in fibroblasts and myeloid cells, while glucose, fatty acids, TNF-α and IL-1β did not affect it. Insulin exerted its inhibitory effect on 11β-HSD1 expression by activating PI3-kinase/Akt-signalling. Consequently, the inhibitory effect of insulin is attenuated in fibroblasts from insulin-resistant db/db mice. We conclude that insulin resistance in obesity and diabetes prevents the down-regulation of 11β-HSD1, leading to elevated endogenous GC levels in diabetic skin, which could contribute to impaired wound healing in patients with DM.
Collapse
Affiliation(s)
- Christina B. Brazel
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany; (C.B.B.); (J.C.S.)
| | - Jan C. Simon
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany; (C.B.B.); (J.C.S.)
| | - Jan P. Tuckermann
- Institute of Comparative Molecular Endocrinology, Ulm University, 89081 Ulm, Germany;
- Klinikum der Universität München, Ludwig-Maximilian University of Munich, 80336 Munich, Germany
| | - Anja Saalbach
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany; (C.B.B.); (J.C.S.)
- Correspondence: ; Tel.: +49-341-9725880; Fax: +49-341-9725878
| |
Collapse
|
29
|
Role of 11β-hydroxysteroid dehydrogenase type 1 in the development of atopic dermatitis. Sci Rep 2020; 10:20237. [PMID: 33214595 PMCID: PMC7678864 DOI: 10.1038/s41598-020-77281-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/06/2020] [Indexed: 12/14/2022] Open
Abstract
Glucocorticoids (GCs) are potent anti-inflammatory drugs, the secretion of which is mediated and controlled by the hypothalamic–pituitary–adrenal axis. However, they are also secreted de novo by peripheral tissues for local use. Several tissues express 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1), including the skin. The inactive GC cortisone is converted by 11β-HSD1 to active GC cortisol, which is responsible for delayed wound healing during a systemic excess of GC. However, the role of 11β-HSD1 in inflammation is unclear. We assessed whether 11β-HSD1 affects the development of atopic dermatitis (AD) in vitro and in vivo. The expression of 11β-HSD1 in the epidermis of AD lesions was higher than that in the epidermis of healthy controls. Knockdown of 11β-HSD1 in human epidermal keratinocytes increased the production of thymic stromal lymphopoietin. In an oxazolone-induced mouse model of AD, localized inhibition of 11β-HSD1 aggravated the development of AD and increased serum cytokine levels associated with AD. Mice with whole-body knockout (KO) of 11β-HSD1 developed significantly worse AD upon induction by oxazolone. We propose that 11β-HSD1 is a major factor affecting AD pathophysiology via suppression of atopic inflammation due to the modulation of active GC in the skin.
Collapse
|
30
|
Synthesis of Kisspeptin-Mimicking Fragments and Investigation of their Skin Anti-Aging Effects. Int J Mol Sci 2020; 21:ijms21228439. [PMID: 33182726 PMCID: PMC7698007 DOI: 10.3390/ijms21228439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/30/2020] [Accepted: 11/06/2020] [Indexed: 11/16/2022] Open
Abstract
In recent years, a number of active materials have been developed to provide anti-aging benefits for skin and, among them, peptides have been considered the most promising candidate due to their remarkable and long-lasting anti-wrinkle activity. Recent studies have begun to elucidate the relationship between the secretion of emotion-related hormones and skin aging. Kisspeptin, a neuropeptide encoded by the KISS1 gene, has gained attention in reproductive endocrinology since it stimulates the reproductive axis in the hypothalamus; however, the effects of Kisspeptin on skin have not been studied yet. In this study, we synthesized Kisspeptin-10 and Kisspeptin-E, which are biologically active fragments, to mimic the action of Kisspeptin. Next, we demonstrated the anti-aging effects of the Kisspeptin-mimicking fragments using UV-induced skin aging models, such as UV-induced human dermal fibroblasts (Hs68) and human skin explants. Kisspeptin-E suppressed UV-induced 11 beta-hydroxysteroid dehydrogenase type 1 (11β-HSD1) stimulation leading to a regulation of skin aging related genes, including type I procollagen, matrix metalloproteinases-1 (MMP-1), interleukin-6 (IL-6), and IL-8, and rescued the skin integrity. Taken together, these results suggest that Kisspeptin-E could be useful to improve UV-induced skin aging by modulating expression of stress related genes, such as 11β-HSD1.
Collapse
|
31
|
Aging-associated alterations in epidermal function and their clinical significance. Aging (Albany NY) 2020; 12:5551-5565. [PMID: 32217811 PMCID: PMC7138575 DOI: 10.18632/aging.102946] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 03/10/2020] [Indexed: 12/11/2022]
Abstract
Chronologically-aged skin displays multiple functional changes in both the dermis and the epidermis. It appears that epidermal dysfunction, compromised permeability homeostasis, reduced stratum corneum hydration and elevated skin surface pH predispose to the development of aging-associated cutaneous and extracutaneous disorders. Improvements in epidermal function have been shown to be an effective alternative therapy in the prevention and treatment of some aging-associated cutaneous disorders, including eczematous dermatitis, pruritus, and xerosis. Recent studies demonstrated that epidermal dysfunction leads to the development of chronic, low-grade systemic inflammation, termed ‘inflammaging,’ which is linked to the development of aging-associated systemic disorders. Thus, correction of epidermal dysfunction could comprise a novel strategy in the prevention and treatment of aging-associated systemic disorders as well. In this review, we summarize aging-associated alterations in epidermal function, their underlying mechanisms, and their clinical significance. Regimens to improve epidermal function in the elderly are also discussed.
Collapse
|
32
|
Bigas J, Sevilla LM, Pérez P. Epidermal Mineralocorticoid Receptor Inactivation Affects the Homeostasis of All Skin Layers in Chronologically Aged Mice. J Invest Dermatol 2020; 140:1899-1908. [PMID: 32199993 DOI: 10.1016/j.jid.2020.03.933] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/17/2020] [Accepted: 03/02/2020] [Indexed: 12/26/2022]
Abstract
The increased production of endogenous glucocorticoids (GCs) in the skin of the elderly population contributes to age-related defects strikingly similar to those occurring after pharmacologic treatments with GCs. GCs act through the ligand-dependent transcription factors GC receptor (GR) and mineralocorticoid receptor (MR). We reported that epidermal MR plays nonredundant roles relative to GR in adult mouse skin homeostasis; however, its relative contribution to natural skin aging has not been previously investigated. A 13-month-old MR epidermal knockout (MREKO) mice showed differential features of aging relative to controls (CO) in all skin compartments. MREKO mice were resistant to age-induced epidermal atrophy but showed reduced dermal thickness, with decreased collagen deposition and decreased SMAD2 and 3 activity. Importantly, the dermal white adipose tissue (dWAT) was 2.5-fold enlarged in 13-month MREKO versus CO, featuring adipocyte hyperplasia and hypertrophy at least in part through early increases in Pparg. These changes correlated with compartment-specific alterations in GC signaling. In addition, conditioned medium from MREKO keratinocytes increased adipocyte differentiation, indicating paracrine regulation of adipogenesis through mechanisms that include activation of β-catenin signaling. These findings highlight the importance of epidermal MR in regulating cross-talk among skin compartments in naturally aged skin through GC and β-catenin signaling pathways.
Collapse
Affiliation(s)
- Judit Bigas
- Instituto de Biomedicina de Valencia-Consejo Superior de Investigaciones Científicas (IBV-CSIC), Jaime Roig, Valencia, Spain
| | - Lisa M Sevilla
- Instituto de Biomedicina de Valencia-Consejo Superior de Investigaciones Científicas (IBV-CSIC), Jaime Roig, Valencia, Spain
| | - Paloma Pérez
- Instituto de Biomedicina de Valencia-Consejo Superior de Investigaciones Científicas (IBV-CSIC), Jaime Roig, Valencia, Spain.
| |
Collapse
|
33
|
Foster MA, Taylor AE, Hill NE, Bentley C, Bishop J, Gilligan LC, Shaheen F, Bion JF, Fallowfield JL, Woods DR, Bancos I, Midwinter MM, Lord JM, Arlt W. Mapping the Steroid Response to Major Trauma From Injury to Recovery: A Prospective Cohort Study. J Clin Endocrinol Metab 2020; 105:dgz302. [PMID: 32101296 PMCID: PMC7043227 DOI: 10.1210/clinem/dgz302] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 01/31/2020] [Indexed: 12/31/2022]
Abstract
CONTEXT Survival rates after severe injury are improving, but complication rates and outcomes are variable. OBJECTIVE This cohort study addressed the lack of longitudinal data on the steroid response to major trauma and during recovery. DESIGN We undertook a prospective, observational cohort study from time of injury to 6 months postinjury at a major UK trauma centre and a military rehabilitation unit, studying patients within 24 hours of major trauma (estimated New Injury Severity Score (NISS) > 15). MAIN OUTCOME MEASURES We measured adrenal and gonadal steroids in serum and 24-hour urine by mass spectrometry, assessed muscle loss by ultrasound and nitrogen excretion, and recorded clinical outcomes (ventilator days, length of hospital stay, opioid use, incidence of organ dysfunction, and sepsis); results were analyzed by generalized mixed-effect linear models. FINDINGS We screened 996 multiple injured adults, approached 106, and recruited 95 eligible patients; 87 survived. We analyzed all male survivors <50 years not treated with steroids (N = 60; median age 27 [interquartile range 24-31] years; median NISS 34 [29-44]). Urinary nitrogen excretion and muscle loss peaked after 1 and 6 weeks, respectively. Serum testosterone, dehydroepiandrosterone, and dehydroepiandrosterone sulfate decreased immediately after trauma and took 2, 4, and more than 6 months, respectively, to recover; opioid treatment delayed dehydroepiandrosterone recovery in a dose-dependent fashion. Androgens and precursors correlated with SOFA score and probability of sepsis. CONCLUSION The catabolic response to severe injury was accompanied by acute and sustained androgen suppression. Whether androgen supplementation improves health outcomes after major trauma requires further investigation.
Collapse
Affiliation(s)
- Mark A Foster
- NIHR-Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Royal Centre for Defence Medicine, Queen Elizabeth Hospital Birmingham, UK
| | - Angela E Taylor
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, University of Birmingham, Birmingham, UK
| | - Neil E Hill
- Section of Investigative Medicine, Imperial College London, UK
| | - Conor Bentley
- NIHR-Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Royal Centre for Defence Medicine, Queen Elizabeth Hospital Birmingham, UK
| | - Jon Bishop
- NIHR-Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Royal Centre for Defence Medicine, Queen Elizabeth Hospital Birmingham, UK
| | - Lorna C Gilligan
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, University of Birmingham, Birmingham, UK
| | - Fozia Shaheen
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, University of Birmingham, Birmingham, UK
| | - Julian F Bion
- Intensive Care Medicine, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | - David R Woods
- Royal Centre for Defence Medicine, Queen Elizabeth Hospital Birmingham, UK
- Leeds Beckett University, Leeds, UK
| | - Irina Bancos
- Division of Endocrinology, Metabolism and Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Mark M Midwinter
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Janet M Lord
- NIHR-Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- MRC-ARUK Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| |
Collapse
|
34
|
Nguyen VT, Farman N, Palacios-Ramirez R, Sbeih M, Behar-Cohen F, Aractingi S, Jaisser F. Cutaneous Wound Healing in Diabetic Mice Is Improved by Topical Mineralocorticoid Receptor Blockade. J Invest Dermatol 2019; 140:223-234.e7. [PMID: 31278904 DOI: 10.1016/j.jid.2019.04.030] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 03/20/2019] [Accepted: 04/08/2019] [Indexed: 12/19/2022]
Abstract
Skin ulcers resulting from impaired wound healing are a serious complication of diabetes. Unresolved inflammation, associated with the dysregulation of both the phenotype and function of macrophages, is involved in the poor healing of diabetic wounds. Here, we report that topical pharmacological inhibition of the mineralocorticoid receptor (MR) by canrenoate or MR small interfering RNA can resolve inflammation to improve delayed skin wound healing in diabetic mouse models; importantly, wounds from normal mice are unaffected. The beneficial effect of canrenoate is associated with an increased ratio of anti-inflammatory M2 macrophages to proinflammatory M1 macrophages in diabetic wounds. Furthermore, we show that MR blockade leads to downregulation of the MR target, LCN2, which may facilitate macrophage polarization toward the M2 phenotype and improve impaired angiogenesis in diabetic wounds. Indeed, diabetic LCN2-deficient mice showed improved wound healing associated with macrophage M2 polarization and angiogenesis. In addition, recombinant LCN2 protein prevented IL-4-induced macrophage switch from M1 to M2 phenotype. In conclusion, topical MR blockade accelerates skin wound healing in diabetic mice via LCN2 reduction, M2 macrophage polarization, prevention of inflammation, and induction of angiogenesis.
Collapse
Affiliation(s)
- Van Tuan Nguyen
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, Paris, France; Laboratory of progenitors and endothelial cells during and after pregnancy, INSERM UMR 938, Centre de Recherche St Antoine, Sorbonne Université, Paris, France; Department of Basic Science, Thai Nguyen University of Agriculture and Forestry, Thainguyen, Vietnam
| | - Nicolette Farman
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, Paris, France
| | - Roberto Palacios-Ramirez
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, Paris, France
| | - Maria Sbeih
- Laboratory of progenitors and endothelial cells during and after pregnancy, INSERM UMR 938, Centre de Recherche St Antoine, Sorbonne Université, Paris, France
| | - Francine Behar-Cohen
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, Paris, France; Faculty of Medicine, Université Paris Descartes, Paris, France
| | - Sélim Aractingi
- Laboratory of progenitors and endothelial cells during and after pregnancy, INSERM UMR 938, Centre de Recherche St Antoine, Sorbonne Université, Paris, France; Faculty of Medicine, Université Paris Descartes, Paris, France; Department of Dermatology, Hôpital Cochin-Tarnier, Paris, France
| | - Frederic Jaisser
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, Paris, France; INSERM, Clinical Investigation Centre 1433, Vandoeuvre-lès-Nancy, France.
| |
Collapse
|
35
|
Chemali D, Roumeliotis N, Cater C, Dulsrud L, Jeffs L, Taddio A, Frndova H, Parshuram C. Describing drug and fluid therapy in the paediatric intensive care unit: A pilot study. J Crit Care 2019; 52:53-57. [PMID: 30974315 DOI: 10.1016/j.jcrc.2019.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 03/28/2019] [Accepted: 04/01/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Care in the paediatric intensive care unit (PICU) involves many clinical activities. The objectives of this study were to evaluate the feasibility of a novel observation method, the reliability of data abstraction, and to report the initial findings from application of this approach. MATERIALS AND METHODS Bedside activities of patients and clinical staff were recorded by direct observational study using video recording and audio annotation. Data were abstracted into 9 broad clinical activities and 12 specific drug-fluid activities. Enrolment rates, agreement between abstractors, clinical activity durations and interruptions are reported. RESULTS We enrolled 42 healthcare professionals, 12 family members of 13 patients, and recorded 12 patients (consent rates of 70%-92%). There were 884 clinical activity episodes. Each hour was comprised of a median (IQR) of 11.9 (4.8-16.5) minutes of drug and fluid related tasks. The 682 drug and fluid related activities were mainly preparation and administration. Interruptions occurred on average 7 times per hour. Data abstraction for 8 h had intra-class correlation co-efficient (95% CI) of 0.91 (079-0.96). CONCLUSIONS Real-time recording of clinical tasks in the PICU using a direct observation model combined with video recording is feasible. Preliminary results suggest abundant and diverse activity is routine.
Collapse
Affiliation(s)
- Dana Chemali
- Department of Critical Care Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Nadia Roumeliotis
- Department of Critical Care Medicine, Hospital for Sick Children, Toronto, ON, Canada; Child Health Evaluative Sciences, The Research Institute, Hospital for Sick Children, Toronto, ON, Canada
| | - Caitlyn Cater
- Department of Critical Care Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Lianne Dulsrud
- Department of Critical Care Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Lianne Jeffs
- Lawrence S. Bloomberg Faculty of Nursing, University of Toronto, Toronto, ON, Canada
| | - Anna Taddio
- Child Health Evaluative Sciences, The Research Institute, Hospital for Sick Children, Toronto, ON, Canada
| | - Helena Frndova
- Department of Critical Care Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Christopher Parshuram
- Department of Critical Care Medicine, Hospital for Sick Children, Toronto, ON, Canada; Child Health Evaluative Sciences, The Research Institute, Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
36
|
Abstract
The skin provides the primary protection for the body against external injuries and is essential in the maintenance of general homeostasis. During ageing, resident cells become senescent and the extracellular matrix, mainly in the dermis, is progressively damaged affecting the normal organization of the skin and its capacity for repair. In parallel, extrinsic factors such as ultraviolet irradiation, pollution, and intrinsic factors such as diabetes or vascular disease can further accelerate this phenomenon. Indeed, numerous mechanisms are involved in age-induced degradation of the skin and these also relate to non-healing or chronic wounds in the elderly. In particular, the generation of reactive oxygen species seems to play a major role in age-related skin modifications. Certainly, targeting both the hormonal status of the skin or its surface nutrition can slow down age-induced degradation of the skin and improve healing of skin damage in the elderly. Skin care regimens that prevent radiation and pollution damage, and reinforce the skin surface and its microbiota are among the different approaches able to minimize the effects of ageing on the skin.
Collapse
|
37
|
Yiallouris A, Tsioutis C, Agapidaki E, Zafeiri M, Agouridis AP, Ntourakis D, Johnson EO. Adrenal Aging and Its Implications on Stress Responsiveness in Humans. Front Endocrinol (Lausanne) 2019; 10:54. [PMID: 30792695 PMCID: PMC6374303 DOI: 10.3389/fendo.2019.00054] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/21/2019] [Indexed: 11/13/2022] Open
Abstract
Normal aging results in subtle changes both in ACTH and cortisol secretion. Most notable is the general increase in mean daily serum cortisol levels in the elderly, without a noteworthy alteration in the normal circadian rhythm pattern. Glucocorticoid excess seen in the elderly population can have serious consequences in both the structural and functional integrity of various key areas in the brain, including the hippocampus, amygdala, prefrontal cortex, with consequent impairment in normal memory, cognitive function, and sleep cycles. The chronically elevated glucocorticoid levels also impinge on the normal stress response in the elderly, leading to an impaired ability to recover from stressful stimuli. In addition to the effects on the brain, glucocorticoid excess is associated with other age-related changes, including loss of muscle mass, hypertension, osteopenia, visceral obesity, and diabetes, among others. In contrast to the increase in glucocorticoid levels, other adrenocortical hormones, particularly serum aldosterone and DHEA (the precursor to androgens and estrogens) show significant decreases in the elderly. The underlying mechanisms for their decrease remain unclear. While the adrenomedullary hormone, norephinephrine, shows an increase in plasma levels, associated with a decrease in clearance, no notable changes observed in plasma epinephrine levels in the elderly. The multiplicity and complexity of the adrenal hormone changes observed throughout the normal aging process, suggests that age-related alterations in cellular growth, differentiation, and senescence specific to the adrenal gland must also be considered.
Collapse
Affiliation(s)
- Andreas Yiallouris
- School of Medicine, European University Cyprus, Nicosia, Cyprus
- Laboratory of Education & Research Neuroscience, Department of Anatomy, School of Medicine, National and Kapodistrian University Athens, Athens, Greece
| | - Constantinos Tsioutis
- School of Medicine, European University Cyprus, Nicosia, Cyprus
- Society of Junior Doctors, Athens, Greece
| | | | - Maria Zafeiri
- Society of Junior Doctors, Athens, Greece
- Diabetes and Obesity Center, Konstantopouleio Hospital, Athens, Greece
| | | | | | - Elizabeth O. Johnson
- School of Medicine, European University Cyprus, Nicosia, Cyprus
- Laboratory of Education & Research Neuroscience, Department of Anatomy, School of Medicine, National and Kapodistrian University Athens, Athens, Greece
- *Correspondence: Elizabeth O. Johnson
| |
Collapse
|
38
|
Inhibitory Effects of Prunella vulgaris L. Extract on 11 β-HSD1 in Human Skin Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:1762478. [PMID: 30402116 PMCID: PMC6193348 DOI: 10.1155/2018/1762478] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 06/05/2018] [Accepted: 09/03/2018] [Indexed: 11/30/2022]
Abstract
Glucocorticoids are a risk factor for age-induced skin structure and function defects, and the glucocorticoid-activating enzyme, 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1), represents a promising therapeutic target. Prunella vulgaris L. (PV) is a perennial and an edible herbaceous plant normally cultivated in Asia and Europe. A recent study demonstrated a broad range of biological activities of PV including immune modulatory, antiviral, antiallergic, anti-inflammatory, antioxidant, and antidiabetic. However, little is known about the inhibitory effect of PV on 11β-HSD1. In this study, we investigated the inhibitory effect of Prunella vulgaris L. extract (PVE) and the underlying mechanism of 11β-HSD11 inhibition. Consistent with these results, cortisol levels were also reduced by PVE in vitro. The cortisone-induced translocation of glucocorticoids receptor (GR) was also attenuated. In addition, PVE inhibited a cortisone-mediated decrease in collagen content in skin. Collectively, these results suggest the beneficial effects of PVE in maintaining skin integrity.
Collapse
|
39
|
Qiao N, Swearingen B, Tritos NA. Cushing's disease in older patients: Presentation and outcome. Clin Endocrinol (Oxf) 2018; 89:444-453. [PMID: 29939400 DOI: 10.1111/cen.13799] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 06/22/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND To define the symptoms, signs and treatment outcomes in a population of older patients with Cushing's disease (CD). METHODS We analysed the clinical presentation and treatment outcomes in 45 CD patients older than 60 years, in comparison with 90 CD patients younger than 60, and a control group of 45 older patients with nonfunctioning pituitary adenomas. We reviewed preoperative clinical characteristics, medical comorbidities, imaging findings and endocrine testing as well as surgical and endocrine outcomes. RESULTS Older CD patients had significantly lower body mass index (BMI) (P = 0.031), were more likely to have muscle wasting (P = 0.006) and women were less likely to have hirsutism (P = 0.033). Older patients with CD had more medical comorbidities than younger patients, which correlated with a higher ASA grade (P < 0.001), but the surgical complication rates were similar in both groups. Surgical remission was achieved in 38/45 (84.4%) older patients and 78/90 (86.7%) younger patients (P = NS). Recurrent disease was more frequent in younger patients (19.2%) in comparison with older patients (2.6%, P = 0.019). CONCLUSIONS Older patients with CD appear to have a distinct phenotype with a more catabolic picture, including a lower BMI and greater prevalence of muscle wasting. Surgical outcomes are similar without a significant age-related increase in complications.
Collapse
Affiliation(s)
- Nidan Qiao
- Department of Neurosurgery, Huashan Hospital, Shanghai, China
- Harvard Medical School, Boston, Massachusetts
| | - Brooke Swearingen
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Nicholas A Tritos
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
40
|
Bunte K, Smith DJ, Chappell MJ, Hassan-Smith ZK, Tomlinson JW, Arlt W, Tiňo P. Learning pharmacokinetic models for in vivo glucocorticoid activation. J Theor Biol 2018; 455:222-231. [PMID: 30048717 DOI: 10.1016/j.jtbi.2018.07.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 07/03/2018] [Accepted: 07/21/2018] [Indexed: 10/28/2022]
Abstract
To understand trends in individual responses to medication, one can take a purely data-driven machine learning approach, or alternatively apply pharmacokinetics combined with mixed-effects statistical modelling. To take advantage of the predictive power of machine learning and the explanatory power of pharmacokinetics, we propose a latent variable mixture model for learning clusters of pharmacokinetic models demonstrated on a clinical data set investigating 11β-hydroxysteroid dehydrogenase enzymes (11β-HSD) activity in healthy adults. The proposed strategy automatically constructs different population models that are not based on prior knowledge or experimental design, but result naturally as mixture component models of the global latent variable mixture model. We study the parameter of the underlying multi-compartment ordinary differential equation model via identifiability analysis on the observable measurements, which reveals the model is structurally locally identifiable. Further approximation with a perturbation technique enables efficient training of the proposed probabilistic latent variable mixture clustering technique using Estimation Maximization. The training on the clinical data results in 4 clusters reflecting the prednisone conversion rate over a period of 4 h based on venous blood samples taken at 20-min intervals. The learned clusters differ in prednisone absorption as well as prednisone/prednisolone conversion. In the discussion section we include a detailed investigation of the relationship of the pharmacokinetic parameters of the trained cluster models for possible or plausible physiological explanation and correlations analysis using additional phenotypic participant measurements.
Collapse
Affiliation(s)
- Kerstin Bunte
- School of Computer Science, The University of Birmingham, Birmingham B15 2TT, UK; Faculty of Science and Engineering, University of Groningen, P.O. Box 407, Groningen 9700 AK, Netherlands.
| | - David J Smith
- School of Mathematics, The University of Birmingham, Birmingham B15 2TT, UK; Institute of Metabolism and Systems Research, University of Birmingham, UK
| | | | - Zaki K Hassan-Smith
- Centre for Applied Biological and Exercise Science, Coventry University, Coventry, UK; Departments of Endocrinology and Acute Internal Medicine, Queen Elizabeth Hospital Birmingham, Birmingham B15 2TH, UK; Centre of Endocrinology, Diabetes and Metabolism, Queen Elizabeth Hospital Birmingham, Birmingham Health Partners, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, UK; Centre of Endocrinology, Diabetes and Metabolism, Queen Elizabeth Hospital Birmingham, Birmingham Health Partners, UK
| | - Peter Tiňo
- School of Computer Science, The University of Birmingham, Birmingham B15 2TT, UK; Institute of Metabolism and Systems Research, University of Birmingham, UK
| |
Collapse
|
41
|
Sevilla LM, Pérez P. Roles of the Glucocorticoid and Mineralocorticoid Receptors in Skin Pathophysiology. Int J Mol Sci 2018; 19:ijms19071906. [PMID: 29966221 PMCID: PMC6073661 DOI: 10.3390/ijms19071906] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/25/2018] [Accepted: 06/27/2018] [Indexed: 12/15/2022] Open
Abstract
The nuclear hormone receptor (NR) superfamily comprises approximately 50 evolutionarily conserved proteins that play major roles in gene regulation by prototypically acting as ligand-dependent transcription factors. Besides their central role in physiology, NRs have been largely used as therapeutic drug targets in many chronic inflammatory conditions and derivatives of their specific ligands, alone or in combination, are frequently prescribed for the treatment of skin diseases. In particular, glucocorticoids (GCs) are the most commonly used compounds for treating prevalent skin diseases such as psoriasis due to their anti-proliferative and anti-inflammatory actions. However, and despite their therapeutic efficacy, the long-term use of GCs is limited because of the cutaneous adverse effects including atrophy, delayed wound healing, and increased susceptibility to stress and infections. The GC receptor (GR/NR3C1) and the mineralocorticoid receptor (MR/NR3C2) are members of the NR subclass NR3C that are highly related, both structurally and functionally. While the GR is ubiquitously expressed and is almost exclusively activated by GCs; an MR has a more restricted tissue expression pattern and can bind GCs and the mineralocorticoid aldosterone with similar high affinity. As these receptors share 95% identity in their DNA binding domains; both can recognize the same hormone response elements; theoretically resulting in transcriptional regulation of the same target genes. However, a major mechanism for specific activation of GRs and/or MRs is at the pre-receptor level by modulating the local availability of active GCs. Furthermore, the selective interactions of each receptor with spatio-temporally regulated transcription factors and co-regulators are crucial for the final transcriptional outcome. While there are abundant genome wide studies identifying GR transcriptional targets in a variety of tissue and cell types; including keratinocytes; the data for MR is more limited thus far. Our group and others have studied the role of GRs and MRs in skin development and disease by generating and characterizing mouse and cellular models with gain- and loss-of-function for each receptor. Both NRs are required for skin barrier competence during mouse development and also play a role in adult skin homeostasis. Moreover, the combined loss of epidermal GRs and MRs caused a more severe skin phenotype relative to single knock-outs (KOs) in developing skin and in acute inflammation and psoriasis, indicating that these corticosteroid receptors play cooperative roles. Understanding GR- and MR-mediated signaling in skin should contribute to deciphering their tissue-specific relative roles and ultimately help to improve GC-based therapies.
Collapse
Affiliation(s)
- Lisa M Sevilla
- Instituto de Biomedicina de Valencia (IBV)-CSIC, 46010 Valencia, Spain.
| | - Paloma Pérez
- Instituto de Biomedicina de Valencia (IBV)-CSIC, 46010 Valencia, Spain.
| |
Collapse
|
42
|
Wang W, Chen ZJ, Myatt L, Sun K. 11β-HSD1 in Human Fetal Membranes as a Potential Therapeutic Target for Preterm Birth. Endocr Rev 2018; 39:241-260. [PMID: 29385440 DOI: 10.1210/er.2017-00188] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 01/23/2018] [Indexed: 12/18/2022]
Abstract
Human parturition is a complex process involving interactions between the myometrium and signals derived from the placenta, fetal membranes, and fetus. Signals originating from fetal membranes are crucial components that trigger parturition, which is clearly illustrated by the labor-initiating consequence of membrane rupture. It has been recognized for a long time that among fetal tissues in late gestation the fetal membranes possess the highest capacity for cortisol regeneration by 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1). However, the exact role of this unique feature remains a mystery. Accumulating evidence indicates that this extra-adrenal source of cortisol may serve as an upstream signal for critical events in human parturition, including enhanced prostaglandin and estrogen synthesis as well as extracellular matrix remodeling. This may explain why such high capacity for cortisol regeneration develops in human fetal membranes at late gestation. Therefore, inhibition of 11β-HSD1 may provide a potential therapeutic target for prevention of preterm birth. This review summarizes the current understanding of the functional role of cortisol regeneration by 11β-HSD1 in human fetal membranes.
Collapse
Affiliation(s)
- Wangsheng Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, Oregon
| | - Kang Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| |
Collapse
|
43
|
Choe SJ, Kim D, Kim EJ, Ahn JS, Choi EJ, Son ED, Lee TR, Choi EH. Psychological Stress Deteriorates Skin Barrier Function by Activating 11β-Hydroxysteroid Dehydrogenase 1 and the HPA Axis. Sci Rep 2018; 8:6334. [PMID: 29679067 PMCID: PMC5910426 DOI: 10.1038/s41598-018-24653-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 04/03/2018] [Indexed: 01/04/2023] Open
Abstract
Psychological stress (PS) increases endogenous glucocorticoids (GC) by activating the hypothalamic-pituitary-adrenal axis. The negative effects of GC on skin barrier function under PS have been well-established. However, endogenous GC can also be active when cortisone (inactive form) is converted to cortisol (active form) by 11β-hydroxysteroid dehydrogenase type I (11ß-HSD1) in the peripheral tissue. Here, we evaluated the changes in 11ß-HSD1 and barrier function under PS. Elevated 11ß-HSD1 in oral mucosa correlated with increased cortisol in the stratum corneum and deteriorated barrier function. Expression of 11ß-HSD1 in the oral mucosa correlated with that in the epidermal keratinocytes. We further investigated whether barrier function improved when PS was relieved using a selective serotonin reuptake inhibitor (SSRI) in patients with anxiety. Decreased 11ß-HSD1 and improved barrier function were observed after SSRI treatment. The collective findings suggest that elevated 11ß-HSD1 under PS increases the level of cutaneous GC and eventually impairs barrier function. PS-alleviating drugs, such as SSRI, may help to treat PS-aggravated skin diseases.
Collapse
Affiliation(s)
- Sung Jay Choe
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Donghye Kim
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Eun Jung Kim
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Joung-Sook Ahn
- Department of Psychiatry, Yonsei University Wonju College of Medicine, Wonju, Korea
| | | | | | | | - Eung Ho Choi
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju, Korea.
| |
Collapse
|
44
|
Kishibe M, Baida G, Bhalla P, Lavker RM, Schlosser B, Iinuma S, Yoshida S, Dudley JT, Budunova I. Important role of kallikrein 6 for the development of keratinocyte proliferative resistance to topical glucocorticoids. Oncotarget 2018; 7:69479-69488. [PMID: 27283773 PMCID: PMC5342492 DOI: 10.18632/oncotarget.9926] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 05/13/2016] [Indexed: 11/25/2022] Open
Abstract
One of the major adverse effects of topical glucocorticoids is cutaneous atrophy often followed by development of resistance to steroids (tachyphylaxis). Previously we showed that after two weeks, interfollicular mouse keratinocytes acquired resistance to anti-proliferative effects of glucocorticoid fluocinolone acetonide (FA). One of the top genes activated by FA during tachyphylaxis was Klk6 encoding kallikrein-related peptidase 6, known to enhance keratinocyte proliferation. KLK6 was also strongly induced by chronic glucocorticoids in human skin. Double immunostaining showed that KLK6+ keratinocytes, localized in suprabasal layer of mouse skin, were frequently adjacent to proliferating 5-bromo-2'-deoxyuridine-positive basal keratinocytes. We used KLK6 knockout (KO) mice to evaluate KLK6 role in skin regeneration after steroid-induced atrophy. KLK6 KOs had thinner epidermis and decreased keratinocyte proliferation. The keratinocytes in wild type and KLK6 KO epidermis were equally sensitive to acute anti-proliferative effect of FA. However, the development of proliferative resistance during chronic treatment was reduced in KO epidermis. This was not due to the changes in glucocorticoid receptor (GR) expression or function as GR protein level and induction of GR-target genes were similar in wild type and KLK6 KO skin. Overall, these results suggest a novel mechanism of epidermal regeneration after glucocorticoid-induced atrophy via KLK6 activation.
Collapse
Affiliation(s)
- Mari Kishibe
- Department of Dermatology, Asahikawa Medical University, Japan
| | - Gleb Baida
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| | - Pankaj Bhalla
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| | - Robert M Lavker
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| | | | - Sin Iinuma
- Department of Dermatology, Asahikawa Medical University, Japan
| | - Shigetaka Yoshida
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Japan
| | - Joel T Dudley
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Irina Budunova
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| |
Collapse
|
45
|
Mi Y, Wang W, Lu J, Zhang C, Wang Y, Ying H, Sun K. Proteasome-mediated degradation of collagen III by cortisol in amnion fibroblasts. J Mol Endocrinol 2018; 60:45-54. [PMID: 29191827 DOI: 10.1530/jme-17-0215] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 11/30/2017] [Indexed: 12/19/2022]
Abstract
Rupture of fetal membranes (ROM) can initiate parturition at both term and preterm. Collagen III in the compact layer of the amnion contributes to the tensile strength of fetal membranes. However, the upstream signals triggering collagen III degradation remain mostly elusive. In this study, we investigated the role of cortisol regenerated by 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1) in collagen III degradation in human amnion fibroblasts with an aim to seek novel targets for the prevention of preterm premature ROM (pPROM)-elicited preterm birth. Human amnion tissue and cultured amnion tissue explants and amnion fibroblasts were used to study the regulation of collagen III, which is composed of three identical 3α 1 chains (COL3A1), by cortisol. Cortisol decreased COL3A1 protein but not mRNA abundance in a concentration-dependent manner. Cortisone also decreased COL3A1 protein, which was blocked by 11β-HSD1 inhibition. The reduction in COL3A1 protein by cortisol was not affected by a transcription inhibitor but was further enhanced by a translation inhibitor. Autophagic pathway inhibitor chloroquine or siRNA-mediated knock-down of ATG7, an essential protein for autophagy, failed to block cortisol-induced reduction in COL3A1 protein abundance, whereas proteasome pathway inhibitors MG132 and bortezomib significantly attenuated cortisol-induced reduction in COL3A1 protein abundance. Moreover, cortisol increased COL3A1 ubiquitination and the reduction of COL3A1 protein by cortisol was blocked by PYR-41, a ubiquitin-activating enzyme inhibitor. Conclusively, cortisol regenerated in amnion fibroblasts may be associated with ROM at parturition by reducing collagen III protein abundance through a ubiquitin-proteasome pathway.
Collapse
Affiliation(s)
- Yabing Mi
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Wangsheng Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Jiangwen Lu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Chuyue Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Yawei Wang
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Hao Ying
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Kang Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| |
Collapse
|
46
|
Tiganescu A, Hupe M, Uchida Y, Mauro T, Elias PM, Holleran WM. Topical 11β-Hydroxysteroid Dehydrogenase Type 1 Inhibition Corrects Cutaneous Features of Systemic Glucocorticoid Excess in Female Mice. Endocrinology 2018; 159:547-556. [PMID: 29087473 PMCID: PMC6459061 DOI: 10.1210/en.2017-00607] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 10/23/2017] [Indexed: 01/13/2023]
Abstract
Glucocorticoid (GC) excess drives multiple cutaneous adverse effects, including skin thinning and poor wound healing. The ubiquitously expressed enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) activates mouse corticosterone from 11-dehydrocorticosterone (and human cortisol from cortisone). We previously demonstrated elevated 11β-HSD1 activity during mouse wound healing, but the interplay between cutaneous 11β-HSD1 and systemic GC excess is unexplored. Here, we examined effects of 11β-HSD1 inhibition by carbenoxolone (CBX) in mice treated with corticosterone (CORT) or vehicle for 6 weeks. Mice were treated bidaily with topical CBX or vehicle (VEH) 7 days before wounding and during wound healing. CORT mice displayed skin thinning and impaired wound healing but also increased epidermal integrity. 11β-HSD1 activity was elevated in unwounded CORT skin and was inhibited by CBX. CORT mice treated with CBX displayed 51%, 59%, and 100% normalization of wound healing, epidermal thickness, and epidermal integrity, respectively. Gene expression studies revealed normalization of interleukin 6, keratinocyte growth factor, collagen 1, collagen 3, matrix metalloproteinase 9, and tissue inhibitor of matrix metalloproteinase 4 by CBX during wound healing. Importantly, proinflammatory cytokine expression and resolution of inflammation were unaffected by 11β-HSD1 inhibition. CBX did not regulate skin function or wound healing in the absence of CORT. Our findings demonstrate that 11β-HSD1 inhibition can limit the cutaneous effects of GC excess, which may improve the safety profile of systemic steroids and the prognosis of chronic wounds.
Collapse
Affiliation(s)
- Ana Tiganescu
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, England
- Department of Dermatology, University of California San Francisco, San Francisco, California
| | - Melanie Hupe
- Department of Dermatology, University of California San Francisco, San Francisco, California
| | - Yoshikazu Uchida
- Department of Dermatology, University of California San Francisco, San Francisco, California
| | - Theadora Mauro
- Department of Dermatology, University of California San Francisco, San Francisco, California
| | - Peter M Elias
- Department of Dermatology, University of California San Francisco, San Francisco, California
| | - Walter M Holleran
- Department of Dermatology, University of California San Francisco, San Francisco, California
| |
Collapse
|
47
|
Gadd45b deficiency promotes premature senescence and skin aging. Oncotarget 2017; 7:26935-48. [PMID: 27105496 PMCID: PMC5053623 DOI: 10.18632/oncotarget.8854] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 04/12/2016] [Indexed: 12/14/2022] Open
Abstract
The GADD45 family of proteins functions as stress sensors in response to various physiological and environmental stressors. Here we show that primary mouse embryo fibroblasts (MEFs) from Gadd45b null mice proliferate slowly, accumulate increased levels of DNA damage, and senesce prematurely. The impaired proliferation and increased senescence in Gadd45b null MEFs is partially reversed by culturing at physiological oxygen levels, indicating that Gadd45b deficiency leads to decreased ability to cope with oxidative stress. Interestingly, Gadd45b null MEFs arrest at the G2/M phase of cell cycle, in contrast to other senescent MEFs, which arrest at G1. FACS analysis of phospho-histone H3 staining showed that Gadd45b null MEFs are arrested in G2 phase rather than M phase. H2O2 and UV irradiation, known to increase oxidative stress, also triggered increased senescence in Gadd45b null MEFs compared to wild type MEFs. In vivo evidence for increased senescence in Gadd45b null mice includes the observation that embryos from Gadd45b null mice exhibit increased senescence staining compared to wild type embryos. Furthermore, it is shown that Gadd45b deficiency promotes senescence and aging phenotypes in mouse skin. Together, these results highlight a novel role for Gadd45b in stress-induced senescence and in tissue aging.
Collapse
|
48
|
Loerz C, Maser E. The cortisol-activating enzyme 11β-hydroxysteroid dehydrogenase type 1 in skeletal muscle in the pathogenesis of the metabolic syndrome. J Steroid Biochem Mol Biol 2017; 174:65-71. [PMID: 28765040 DOI: 10.1016/j.jsbmb.2017.07.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/24/2017] [Accepted: 07/25/2017] [Indexed: 12/25/2022]
Abstract
The enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) contributes to intracellular glucocorticoid action by converting inactive cortisone to its receptor-active form cortisol (11-dehydrocorticosterone and corticosterone in mice and rats). The potential role of 11β-HSD1 in the pathogenesis of the metabolic syndrome has emerged over the past three decades. However, the precise impact of 11β-HSD1 in obesity-related diseases remains uncertain. Many studies from animal experiments to clinical studies have investigated liver and adipose tissue 11β-HSD1 in relation to obesity and its metabolic disorders including insulin resistance. But the relevance of 11β-HSD1 in skeletal muscle has been less extensively studied. On the other hand, skeletal muscle is assumed to be the main site of peripheral insulin resistance, but the biological relevance of 11β-HSD1 in skeletal muscle is unclear. This mini-review will focus on 11β-HSD1 in skeletal muscle and its postulated link to obesity and insulin-resistance.
Collapse
Affiliation(s)
- Christine Loerz
- Institute of Toxicology and Pharmacology for Natural Scientists, University Medical School Schleswig-Holstein, Kiel, Germany
| | - Edmund Maser
- Institute of Toxicology and Pharmacology for Natural Scientists, University Medical School Schleswig-Holstein, Kiel, Germany.
| |
Collapse
|
49
|
Stojadinovic O, Lindley LE, Jozic I, Tomic-Canic M. Mineralocorticoid Receptor Antagonists-A New Sprinkle of Salt and Youth. J Invest Dermatol 2017; 136:1938-1941. [PMID: 27664711 DOI: 10.1016/j.jid.2016.07.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 07/13/2016] [Indexed: 01/17/2023]
Abstract
Skin atrophy and impaired cutaneous wound healing are the recognized side effects of topical glucocorticoid (GC) therapy. Although GCs have high affinity for the glucocorticoid receptor, they also bind and activate the mineralocorticoid receptor. In light of this, one can speculate that some of the GC-mediated side effects can be remedied by blocking activation of the mineralocorticoid receptor. Indeed, according to Nguyen et al., local inhibition of the mineralocorticoid receptor via antagonists (spironolactone, canrenoate, and eplerenone) rescues GC-induced delayed epithelialization and accelerates wound closure in diabetic animals by targeting epithelial sodium channels and stimulating keratinocyte proliferation. These findings suggest that the use of mineralocorticoid receptor antagonists coupled with GC therapy may be beneficial in overcoming at least some of the GC-mediated side effects.
Collapse
Affiliation(s)
- Olivera Stojadinovic
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA; Graduate Program in Biomedical Sciences, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Linsey E Lindley
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ivan Jozic
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA; John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida, USA.
| |
Collapse
|
50
|
Nam JJ, Min JE, Son MH, Oh JH, Kang S. Ultraviolet- and infrared-induced 11 beta-hydroxysteroid dehydrogenase type 1 activating skin photoaging is inhibited by red ginseng extract containing high concentration of ginsenoside Rg3(S). PHOTODERMATOLOGY PHOTOIMMUNOLOGY & PHOTOMEDICINE 2017; 33:311-320. [PMID: 28793178 DOI: 10.1111/phpp.12337] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/03/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Sun irradiation is one of major extrinsic stressors responsible for premature skin aging through activation and expression of 11 beta-hydroxysteroid dehydrogenase type 1 (11β-HSD1), which converts inactive cortisone to active cortisol. The aim of this study was to evaluate the inhibitory effects of red ginseng extract containing high concentrations of ginsenoside Rg3 (S) (GERg3) on 11β-HSD1-induced skin photoaging. METHODS To evaluate the inhibitory effects of GERg3 on ultraviolet- (UV) or infrared (IR)-induced skin photoaging, human dermal fibroblasts or a normal human 3D skin model was exposed to UV or an IR. RT-PCR, ELISA, Western blot, and H&E staining were used for evaluations. GERg3 was isolated from crude red ginseng. RESULTS GERg3 inhibited the increased expressions of 11β-HSD1, interleukin (IL)-6, and matrix metalloproteinase-1 (MMP-1) in UVB- or IR-exposed Hs68 cells. Additionally, the increased cortisol, IL-6, and MMP-1 expressions were effectively reduced by GERg3 in UVA-exposed 3D skin models. The photoinduced decrease in type 1 procollagen also recovered as a result of GERg3 treatment in Hs68 cells and the 3D skin model. In addition, the UVA-exposed dermal thickness was decreased in comparison with the UVA-protected 3D skin model, recovered with GERg3 treatment. CONCLUSION GERg3 had antiphotoaging effects in UV- or IR-exposed human dermal fibroblasts and normal human 3D skin model.
Collapse
Affiliation(s)
- Jin-Ju Nam
- R&I Center, COSMAX BTI, Seongnam, South Korea
| | - Ji-Eun Min
- R&I Center, COSMAX, Seongnam, South Korea
| | - Min-Ho Son
- R&I Center, COSMAX, Seongnam, South Korea
| | | | | |
Collapse
|