51
|
Lack of adipose-specific hexose-6-phosphate dehydrogenase causes inactivation of adipose glucocorticoids and improves metabolic phenotype in mice. Clin Sci (Lond) 2020; 133:2189-2202. [PMID: 31696216 DOI: 10.1042/cs20190679] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/07/2019] [Accepted: 10/18/2019] [Indexed: 12/11/2022]
Abstract
Excessive glucocorticoid (GC) production in adipose tissue promotes the development of visceral obesity and metabolic syndrome (MS). 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) is critical for controlling intracellular GC production, and this process is tightly regulated by hexose-6-phosphate dehydrogenase (H6PDH). To better understand the integrated molecular physiological effects of adipose H6PDH, we created a tissue-specific knockout of the H6PDH gene mouse model in adipocytes (adipocyte-specific conditional knockout of H6PDH (H6PDHAcKO) mice). H6PDHAcKO mice exhibited almost complete absence of H6PDH expression and decreased intra-adipose corticosterone production with a reduction in 11β-HSD1 activity in adipose tissue. These mice also had decreased abdominal fat mass, which was paralleled by decreased adipose lipogenic acetyl-CoA carboxylase (ACC) and ATP-citrate lyase (ACL) gene expression and reduction in their transcription factor C/EBPα mRNA levels. Moreover, H6PDHAcKO mice also had reduced fasting blood glucose levels, increased glucose tolerance, and increased insulin sensitivity. In addition, plasma free fatty acid (FFA) levels were decreased with a concomitant decrease in the expression of lipase adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL) in adipose tissue. These results indicate that inactivation of adipocyte H6PDH expression is sufficient to cause intra-adipose GC inactivation that leads to a favorable pattern of metabolic phenotypes. These data suggest that H6PDHAcKO mice may provide a good model for studying the potential contributions of fat-specific H6PDH inhibition to improve the metabolic phenotype in vivo. Our study suggests that suppression or inactivation of H6PDH expression in adipocytes could be an effective intervention for treating obesity and diabetes.
Collapse
|
52
|
Rossi A, Simeoli C, Salerno M, Ferrigno R, Della Casa R, Colao A, Strisciuglio P, Parenti G, Pivonello R, Melis D. Imbalanced cortisol concentrations in glycogen storage disease type I: evidence for a possible link between endocrine regulation and metabolic derangement. Orphanet J Rare Dis 2020; 15:99. [PMID: 32306986 PMCID: PMC7169016 DOI: 10.1186/s13023-020-01377-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 04/07/2020] [Indexed: 12/13/2022] Open
Abstract
Background Glycogen storage disease type I (GSDI) is an inborn error of carbohydrate metabolism caused by mutations of either the G6PC gene (GSDIa) or the SLC37A4 gene (GSDIb). Glucose 6-phosphate (G6P) availability has been shown to modulate 11β-hydroxysteroid dehydrogenase type 1 (11βHSD1), an ER-bound enzyme catalyzing the local conversion of inactive cortisone into active cortisol. Adrenal cortex assessment has never been performed in GSDI. The aim of the current study was to evaluate the adrenal cortex hormones levels in GSDI patients. Methods Seventeen GSDI (10 GSDIa and 7 GSDIb) patients and thirty-four age and sex-matched controls were enrolled. Baseline adrenal cortex hormones and biochemical markers of metabolic control serum levels were analyzed. Low dose ACTH stimulation test was also performed. Results Baseline cortisol serum levels were higher in GSDIa patients (p = 0.042) and lower in GSDIb patients (p = 0.041) than controls. GSDIa patients also showed higher peak cortisol response (p = 0.000) and Cortisol AUC (p = 0.029). In GSDIa patients, serum cholesterol (p = 0.000), triglycerides (p = 0.000), lactate (p = 0.000) and uric acid (p = 0.008) levels were higher and bicarbonate (p = 0.000) levels were lower than controls. In GSDIb patients, serum cholesterol levels (p = 0.016) were lower and lactate (p = 0.000) and uric acid (p = 0.000) levels were higher than controls. Baseline cortisol serum levels directly correlated with cholesterol (ρ = 0.65, p = 0.005) and triglycerides (ρ = 0.60, p = 0.012) serum levels in GSDI patients. Conclusions The present study showed impaired cortisol levels in GSDI patients, with opposite trend between GSDIa and GSDIb. The otherwise preserved adrenal cortex function suggests that this finding might be secondary to local deregulation rather than hypothalamo-pituitary-adrenal axis dysfunction in GSDI patients. We hypothesize that 11βHSD1 might represent the link between endocrine regulation and metabolic derangement in GSDI, constituting new potential therapeutic target in GSDI patients.
Collapse
Affiliation(s)
- Alessandro Rossi
- Department of Translational Medicine, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - Chiara Simeoli
- Dipartmento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, University of Naples "Federico II", Naples, Italy
| | - Mariacarolina Salerno
- Department of Translational Medicine, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - Rosario Ferrigno
- Dipartmento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, University of Naples "Federico II", Naples, Italy
| | - Roberto Della Casa
- Maternal-Infant Department, Pediatrics Unit, "San Pio" Hospital, Benevento, Italy
| | - Annamaria Colao
- Dipartmento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, University of Naples "Federico II", Naples, Italy
| | - Pietro Strisciuglio
- Department of Translational Medicine, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - Giancarlo Parenti
- Department of Translational Medicine, Section of Pediatrics, University of Naples "Federico II", Naples, Italy.,Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Rosario Pivonello
- Dipartmento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, University of Naples "Federico II", Naples, Italy
| | - Daniela Melis
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Section of Pediatrics, University of Salerno, Via Salvador Allende, 43 84081, Baronissi (Salerno), Italy.
| |
Collapse
|
53
|
Wang K, Zhu H, Zhao H, Zhang K, Tian Y. Application of carbamyl in structural optimization. Bioorg Chem 2020; 98:103757. [PMID: 32217370 DOI: 10.1016/j.bioorg.2020.103757] [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: 12/10/2019] [Revised: 02/07/2020] [Accepted: 03/12/2020] [Indexed: 12/11/2022]
Abstract
Carbamyl is considered a privileged structure in medicinal chemistry. It has a wide range of biological activities such as antimicrobial, anticancer, anti-epilepsy, for which the best evidence is a number of marketed carbamyl-containing drugs. Carbamyl is formed of primary amine and carbonyl moieties that act as hydrogen bond donors and hydrogen acceptors with residues of targets respectively, which are benefit for improving pharmacological activities. In other cases, the introduced carbamyl improves drug-like properties including oral bioavailability. In this review, we introduce the carbamyl-containing drugs and the application of carbamyl in structural optimization as a result of enhancing activities or/and drug-like properties.
Collapse
Affiliation(s)
- Kuanglei Wang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, PR China; International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, PR China; School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Hongxi Zhu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Hongqian Zhao
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Kun Zhang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, PR China; International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, PR China; School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, PR China.
| | - Yongshou Tian
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
54
|
A heretical view: rather than a solely placental protective function, placental 11β hydroxysteroid dehydrogenase 2 also provides substrate for fetal peripheral cortisol synthesis in obese pregnant ewes. J Dev Orig Health Dis 2020; 12:94-100. [PMID: 32151296 DOI: 10.1017/s2040174420000112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Exposure to glucocorticoid levels higher than appropriate for current developmental stages induces offspring metabolic dysfunction. Overfed/obese (OB) ewes and their fetuses display elevated blood cortisol, while fetal Adrenocorticotropic hormone (ACTH) remains unchanged. We hypothesized that OB pregnancies would show increased placental 11β hydroxysteroid dehydrogenase 2 (11β-HSD2) that converts maternal cortisol to fetal cortisone as it crosses the placenta and increased 11β-HSD system components responsible for peripheral tissue cortisol production, providing a mechanism for ACTH-independent increase in circulating fetal cortisol. Control ewes ate 100% National Research Council recommendations (CON) and OB ewes ate 150% CON diet from 60 days before conception until necropsy at day 135 gestation. At necropsy, maternal jugular and umbilical venous blood, fetal liver, perirenal fat, and cotyledonary tissues were harvested. Maternal plasma cortisol and fetal cortisol and cortisone were measured. Fetal liver, perirenal fat, cotyledonary 11β-HSD1, hexose-6-phosphate dehydrogenase (H6PD), and 11β-HSD2 protein abundance were determined by Western blot. Maternal plasma cortisol, fetal plasma cortisol, and cortisone were higher in OB vs. CON (p < 0.01). 11β-HSD2 protein was greater (p < 0.05) in OB cotyledonary tissue than CON. 11β-HSD1 abundance increased (p < 0.05) in OB vs. CON fetal liver and perirenal fat. Fetal H6PD, an 11β-HSD1 cofactor, also increased (p < 0.05) in OB vs. CON perirenal fat and tended to be elevated in OB liver (p < 0.10). Our data provide evidence for increased 11β-HSD system components responsible for peripheral tissue cortisol production in fetal liver and adipose tissue, thereby providing a mechanism for an ACTH-independent increase in circulating fetal cortisol in OB fetuses.
Collapse
|
55
|
Ferguson D, Hutson I, Tycksen E, Pietka TA, Bauerle K, Harris CA. Role of Mineralocorticoid Receptor in Adipogenesis and Obesity in Male Mice. Endocrinology 2020; 161:bqz010. [PMID: 32036385 PMCID: PMC7007880 DOI: 10.1210/endocr/bqz010] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 10/30/2019] [Indexed: 02/07/2023]
Abstract
Increased visceral adiposity and hyperglycemia, 2 characteristics of metabolic syndrome, are also present in conditions of excess glucocorticoids (GCs). GCs are hormones thought to act primarily via the glucocorticoid receptor (GR). GCs are commonly prescribed for inflammatory disorders, yet their use is limited due to many adverse metabolic side effects. In addition to GR, GCs also bind the mineralocorticoid receptor (MR), but there are many conflicting studies about the exact role of MR in metabolic disease. Using MR knockout mice (MRKO), we find that both white and brown adipose depots form normally when compared with wild-type mice at P5. We created mice with adipocyte-specific deletion of MR (FMRKO) to better understand the role of MR in metabolic dysfunction. Treatment of mice with excess GCs for 4 weeks, via corticosterone in drinking water, induced increased fat mass and glucose intolerance to similar levels in FMRKO and floxed control mice. Separately, when fed a high-fat diet for 16 weeks, FMRKO mice had reduced body weight, fat mass, and hepatic steatosis, relative to floxed control mice. Decreased adiposity likely resulted from increased energy expenditure since food intake was not different. RNA sequencing analysis revealed decreased enrichment of genes associated with adipogenesis in inguinal white adipose of FMRKO mice. Differentiation of mouse embryonic fibroblasts (MEFs) showed modestly impaired adipogenesis in MRKO MEFs compared with wild type, but this was rescued upon the addition of peroxisome proliferator-activated receptor gamma (PPARγ) agonist or PPARγ overexpression. Collectively, these studies provide further evidence supporting the potential value of MR as a therapeutic target for conditions associated with metabolic syndrome.
Collapse
Affiliation(s)
- Daniel Ferguson
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
| | - Irina Hutson
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
| | - Eric Tycksen
- Genome Technology Access Center, McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Terri A Pietka
- Nutrition and Geriatrics Division, Washington University School of Medicine, St. Louis, Missouri
| | - Kevin Bauerle
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
| | - Charles A Harris
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
- Department of Medicine, Veterans Affairs St Louis Healthcare System, John Cochran Division, St. Louis, Missouri
| |
Collapse
|
56
|
Pérez JH, Swanson RE, Lau HJ, Cheah J, Bishop VR, Snell KRS, Reid AMA, Meddle SL, Wingfield JC, Krause JS. Tissue-specific expression of 11β-HSD and its effects on plasma corticosterone during the stress response. ACTA ACUST UNITED AC 2020; 223:jeb.209346. [PMID: 31796607 DOI: 10.1242/jeb.209346] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 11/21/2019] [Indexed: 01/14/2023]
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis is under complex regulatory control at multiple levels. Enzymatic regulation plays an important role in both circulating levels of glucocorticoids and target tissue exposure. Three key enzyme pathways are responsible for the immediate control of glucocorticoids. De novo synthesis of glucocorticoid from cholesterol involves a multistep enzymatic cascade. This cascade terminates with 11β-hydroxylase, responsible for the final conversion of 11-deoxy precursors into active glucocorticoids. Additionally, 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) controls regeneration of glucocorticoids from inactive metabolites, providing a secondary source of active glucocorticoids. Localized inactivation of glucocorticoids is under the control of Type 2 11β-HSD (11β-HSD2). The function of these enzymes is largely unexplored in wild species, particularly songbirds. Here, we investigated the contribution of both clearance and generation of glucocorticoids to regulation of the hormonal stress response via the use of pharmacological antagonists. Additionally, we mapped 11β-HSD gene expression. We found 11β-HSD1 primarily in liver, kidney and adrenal glands, although it was detectable across all tissue types. 11β-HSD2 was predominately expressed in the adrenal glands and kidney with moderate gonadal and liver expression. Inhibition of glucocorticoid generation by metyrapone was found to decrease levels peripherally, while both peripheral and central administration of the 11β-HSD2 inhibitor DETC resulted in elevated concentrations of corticosterone. These data suggest that during the stress response, peripheral antagonism of the 11β-HSD system has a greater impact on circulating glucocorticoid levels than central control. Further studies should aim to elucidate the respective roles of the 11β-HSD and 11β-hydroxylase enzymes.
Collapse
Affiliation(s)
- Jonathan H Pérez
- Department of Neurobiology, Physiology and Behavior, University of California Davis, One Shields Avenue, Davis, CA 95616, USA .,The Institute of Biodiversity, Animal Health & Comparative Medicine, University of Glasgow, Glasgow, G12 8QQ, UK.,The Roslin Institute, The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Ryan E Swanson
- Department of Neurobiology, Physiology and Behavior, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Hannah J Lau
- Department of Neurobiology, Physiology and Behavior, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Jeffrey Cheah
- Department of Neurobiology, Physiology and Behavior, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Valerie R Bishop
- The Roslin Institute, The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Katherine R S Snell
- Center for Macroecology, Evolution and Climate, Natural History Museum of Denmark, University of Copenhagen, Universitetsparken 15, DK-2100 Copenhagen, Denmark
| | - Angus M A Reid
- The Roslin Institute, The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK.,MRC HGU, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Simone L Meddle
- The Roslin Institute, The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - John C Wingfield
- Department of Neurobiology, Physiology and Behavior, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Jesse S Krause
- Department of Neurobiology, Physiology and Behavior, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| |
Collapse
|
57
|
Akalestou E, Genser L, Rutter GA. Glucocorticoid Metabolism in Obesity and Following Weight Loss. Front Endocrinol (Lausanne) 2020; 11:59. [PMID: 32153504 PMCID: PMC7045057 DOI: 10.3389/fendo.2020.00059] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/30/2020] [Indexed: 01/08/2023] Open
Abstract
Glucocorticoids are steroid hormones produced by the adrenal cortex and are essential for the maintenance of various metabolic and homeostatic functions. Their function is regulated at the tissue level by 11β-hydroxysteroid dehydrogenases and they signal through the glucocorticoid receptor, a ligand-dependent transcription factor. Clinical observations have linked excess glucocorticoid levels with profound metabolic disturbances of intermediate metabolism resulting in abdominal obesity, insulin resistance and dyslipidaemia. In this review, we discuss the physiological mechanisms of glucocorticoid secretion, regulation and function, and survey the metabolic consequences of excess glucocorticoid action resulting from elevated release and activation or up-regulated signaling. Finally, we summarize the reported impact of weight loss by diet, exercise, or bariatric surgery on circulating and tissue-specific glucocorticoid levels and examine the therapeutic possibility of reversing glucocorticoid-associated metabolic disorders.
Collapse
Affiliation(s)
- Elina Akalestou
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
| | - Laurent Genser
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
- Department of Digestive and Hepato-Pancreato-Biliary Surgery, Liver Transplantation, Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière University Hospital, Institut Hospitalo-Universitaire ICAN, Sorbonne Université, Paris, France
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
- *Correspondence: Guy A. Rutter
| |
Collapse
|
58
|
Kwon YJ, Son DH, Chung TH, Lee YJ. A Review of the Pharmacological Efficacy and Safety of Licorice Root from Corroborative Clinical Trial Findings. J Med Food 2019; 23:12-20. [PMID: 31874059 DOI: 10.1089/jmf.2019.4459] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Since ancient times, licorice, the root of Glycyrrhiza glabra, has been known to have a wide spectrum of therapeutic effects. Glycyrrhizin is cleaved to glycyrrhizic acid, which is subsequently converted to glycyrrhetic acid by human intestinal microflora. Glycyrrhetic acid is a potent inhibitor of 11β-hydroxysteroid dehydrogenase (11β-HSD) and performs a range of corticosteroid-like activities. The pharmacologic effects of licorice contribute to its anti-inflammatory, antioxidative, anti-allergenic, and antimicrobial properties. Licorice has been used to treat liver disease, gastrointestinal disorders, oral disease, and various skin disorders and has been used in gum, candy, herbs, alcoholic beverages, and food supplements. Licorice and its extracts, especially glycyrrhizin, can be taken orally, through the skin (in the form of gels and oils), and intravenously. Licorice demonstrates mineralocorticoid-like activity not only by inhibiting 11β-HSD2, but also by binding to a mineralocorticoid receptor, leading to potentially adverse risks of mineralocorticoid-like overactivity. Chronic use of licorice can lead to hypokalemia and hypertension, and some people are more sensitive to licorice exposure. Based on clinical trials, this review summarizes the positive effects of licorice and other reported side effects.
Collapse
Affiliation(s)
- Yu-Jin Kwon
- Department of Family Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Gyeongi, Korea.,Department of Medicine, Graduate School, Yonsei University, Seoul, Korea
| | - Da-Hye Son
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Tae-Ha Chung
- Department of Medicine, Graduate School, Yonsei University, Seoul, Korea.,Department of Health Medicine, Severance Hospital, Severance Check-Up, Yonsei University Health System, Seoul, Korea
| | - Yong-Jae Lee
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
59
|
Zhang J, Yang Y, Liu W, Schlenk D, Liu J. Glucocorticoid and mineralocorticoid receptors and corticosteroid homeostasis are potential targets for endocrine-disrupting chemicals. ENVIRONMENT INTERNATIONAL 2019; 133:105133. [PMID: 31520960 DOI: 10.1016/j.envint.2019.105133] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/19/2019] [Accepted: 08/26/2019] [Indexed: 05/16/2023]
Abstract
Endocrine-disrupting chemicals (EDCs) have received significant concern, since they ubiquitously exist in the environment and are able to induce adverse health effects on human and wildlife. Increasing evidence shows that the glucocorticoid receptor (GR) and the mineralocorticoid receptor (MR), members of the steroid receptor subfamily, are potential targets for EDCs. GR and MR mediate the actions of glucocorticoids and mineralocorticoids, respectively, which are two main classes of corticosteroids involved in many physiological processes. The effects of EDCs on the homeostasis of these two classes of corticosteroids have also gained more attention recently. This review summarized the effects of environmental GR/MR ligands on receptor activity, and disruption of corticosteroid homeostasis. More than 130 chemicals classified into 7 main categories were reviewed, including metals, metalloids, pesticides, bisphenol analogues, flame retardants, other industrial chemicals and pharmaceuticals. The mechanisms by which EDCs interfere with GR/MR activity are primarily involved in ligand-receptor binding, nuclear translocation of the receptor complex, DNA-receptor binding, and changes in the expression of endogenous GR/MR genes. Besides directly interfering with receptors, enzyme-catalyzed synthesis and prereceptor regulation pathways of corticosteroids are also important targets for EDCs. The collected evidence suggests that corticosteroids and their receptors should be considered as potential targets for safety assessment of EDCs. The recognition of relevant xenobiotics and their underlying mechanisms of action is still a challenge in this emerging field of research.
Collapse
Affiliation(s)
- Jianyun Zhang
- MOE Key Laboratory of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China; Department of Public Health, School of Medicine, Hangzhou Normal University, Hangzhou 310036, China
| | - Ye Yang
- Institute of Hygiene, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Weiping Liu
- MOE Key Laboratory of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Daniel Schlenk
- Department of Environmental Sciences, University of California, Riverside, 900 University Avenue, Riverside, CA 92521, United States
| | - Jing Liu
- MOE Key Laboratory of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
60
|
Chedid MF, do Nascimento FV, de Oliveira FS, de Souza BM, Kruel CRP, Gurski RR, Canani LH, Crispim D, Gerchman F. Interaction of HSD11B1 and H6PD polymorphisms in subjects with type 2 diabetes are protective factors against obesity: a cross-sectional study. Diabetol Metab Syndr 2019; 11:78. [PMID: 31558916 PMCID: PMC6755690 DOI: 10.1186/s13098-019-0474-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/12/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The enzyme 11-beta hydroxysteroid dehydrogenase type 1 (HSD11B1) converts inactive cortisone to active cortisol in a process mediated by the enzyme hexose-6-phosphate dehydrogenase (H6PD). The generation of cortisol from this reaction may increase intra-abdominal cortisol levels and contribute to the physiopathogenesis of obesity and metabolic syndrome (MetS). The relationship of HSD11B1 rs45487298 and H6PD rs6688832 polymorphisms with obesity and MetS was studied. We also studied how HSD11B1 abdominal subcutaneous (SAT) and visceral adipose tissue (VAT) gene expression is related to body fat distribution. METHODS Rates of obesity and MetS features were cross-sectionally analyzed according to these polymorphisms in 1006 Brazilian white patients with type 2 diabetes (T2DM). Additionally, HSD11B1 expression was analyzed in VAT and SAT in a different cohort of 28 participants with and without obesity who underwent elective abdominal operations. RESULTS Although polymorphisms of the two genes were not individually associated with MetS features, a synergistic effect was observed between both. Carriers of at least three minor alleles exhibited lower BMI compared to those with two or fewer minor alleles adjusting for gender and age (27.4 ± 4.9 vs. 29.3 ± 5.3 kg/m2; P = 0.005; mean ± SD). Obesity frequency was also lower in the first group (24.4% vs. 41.6%, OR = 0.43, 95% CI 0.21-0.87; P = 0.019). In the second cohort of 28 subjects, HSD11B1 gene expression in VAT was inversely correlated with BMI (r = - 0.435, P = 0.034), waist circumference (r = - 0.584, P = 0.003) and waist-to-height ratio (r = - 0.526, P = 0.010). CONCLUSIONS These polymorphisms might interact in the protection against obesity in T2DM individuals. Obese individuals may have decreased intra-abdominal VAT HSD11B1 gene expression resulting in decreasing intra-abdominal cortisol levels as a compensatory mechanism against central and general adiposity.
Collapse
Affiliation(s)
- Marcio F. Chedid
- Postgraduate Program of Surgical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Division of Gastrointestinal Surgery, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Filipe V. do Nascimento
- Postgraduate Program in Medical Science: Endocrinology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Fernanda S. de Oliveira
- Postgraduate Program in Medical Science: Endocrinology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Bianca M. de Souza
- Postgraduate Program of Surgical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Cleber R. P. Kruel
- Postgraduate Program of Surgical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Division of Gastrointestinal Surgery, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Richard R. Gurski
- Postgraduate Program of Surgical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Division of Gastrointestinal Surgery, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luis H. Canani
- Postgraduate Program in Medical Science: Endocrinology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Division of Endocrinology and Metabolism, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos 2350, Prédio 12, 4° andar, Bairro Santana, Porto Alegre, RS 90035-003 Brazil
| | - Daisy Crispim
- Postgraduate Program in Medical Science: Endocrinology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Division of Endocrinology and Metabolism, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos 2350, Prédio 12, 4° andar, Bairro Santana, Porto Alegre, RS 90035-003 Brazil
| | - Fernando Gerchman
- Postgraduate Program in Medical Science: Endocrinology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Division of Endocrinology and Metabolism, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos 2350, Prédio 12, 4° andar, Bairro Santana, Porto Alegre, RS 90035-003 Brazil
| |
Collapse
|
61
|
Praveenkumar E, Gurrapu N, Kumar Kolluri P, Yerragunta V, Reddy Kunduru B, Subhashini N. Synthesis, anti-diabetic evaluation and molecular docking studies of 4-(1-aryl-1H-1, 2, 3-triazol-4-yl)-1,4-dihydropyridine derivatives as novel 11-β hydroxysteroid dehydrogenase-1 (11β-HSD1) inhibitors. Bioorg Chem 2019; 90:103056. [DOI: 10.1016/j.bioorg.2019.103056] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/29/2019] [Accepted: 06/06/2019] [Indexed: 12/13/2022]
|
62
|
Fenton CG, Doig CL, Fareed S, Naylor A, Morrell AP, Addison O, Wehmeyer C, Buckley CD, Cooper MS, Lavery GG, Raza K, Hardy RS. 11β-HSD1 plays a critical role in trabecular bone loss associated with systemic glucocorticoid therapy. Arthritis Res Ther 2019; 21:188. [PMID: 31420008 PMCID: PMC6698000 DOI: 10.1186/s13075-019-1972-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 08/07/2019] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Despite their efficacy in the treatment of chronic inflammation, the prolonged application of therapeutic glucocorticoids (GCs) is limited by significant systemic side effects including glucocorticoid-induced osteoporosis (GIOP). 11β-Hydroxysteroid dehydrogenase type 1 (11β-HSD1) is a bi-directional enzyme that primarily activates GCs in vivo, regulating tissue-specific exposure to active GC. We aimed to determine the contribution of 11β-HSD1 to GIOP. METHODS Wild type (WT) and 11β-HSD1 knockout (KO) mice were treated with corticosterone (100 μg/ml, 0.66% ethanol) or vehicle (0.66% ethanol) in drinking water over 4 weeks (six animals per group). Bone parameters were assessed by micro-CT, sub-micron absorption tomography and serum markers of bone metabolism. Osteoblast and osteoclast gene expression was assessed by quantitative RT-PCR. RESULTS Wild type mice receiving corticosterone developed marked trabecular bone loss with reduced bone volume to tissue volume (BV/TV), trabecular thickness (Tb.Th) and trabecular number (Tb.N). Histomorphometric analysis revealed a dramatic reduction in osteoblast numbers. This was matched by a significant reduction in the serum marker of osteoblast bone formation P1NP and gene expression of the osteoblast markers Alp and Bglap. In contrast, 11β-HSD1 KO mice receiving corticosterone demonstrated almost complete protection from trabecular bone loss, with partial protection from the decrease in osteoblast numbers and markers of bone formation relative to WT counterparts receiving corticosterone. CONCLUSIONS This study demonstrates that 11β-HSD1 plays a critical role in GIOP, mediating GC suppression of anabolic bone formation and reduced bone volume secondary to a decrease in osteoblast numbers. This raises the intriguing possibility that therapeutic inhibitors of 11β-HSD1 may be effective in preventing GIOP in patients receiving therapeutic steroids.
Collapse
Affiliation(s)
- C. G. Fenton
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - C. L. Doig
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - S. Fareed
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - A. Naylor
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - A. P. Morrell
- Aston Institute of Materials Research, Aston University, Birmingham, UK
| | - O. Addison
- Institute of Clinical Sciences, University of Birmingham, Birmingham, UK
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - C. Wehmeyer
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - C. D. Buckley
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - M. S. Cooper
- ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - G. G. Lavery
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - K. Raza
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, UK
| | - R. S. Hardy
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
63
|
Ling T, Miller DJ, Lang WH, Griffith E, Rodriguez-Cortes A, El Ayachi I, Palacios G, Min J, Miranda-Carboni G, Lee RE, Rivas F. Mechanistic Insight on the Mode of Action of Colletoic Acid. J Med Chem 2019; 62:6925-6940. [PMID: 31294974 DOI: 10.1021/acs.jmedchem.9b00187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The natural product colletoic acid (CA) is a selective inhibitor of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), which primarily converts cortisone to the active glucocorticoid (GC) cortisol. Here, CA's mode of action and its potential as a chemical tool to study intracellular GC signaling in adipogenesis are disclosed. 11β-HSD1 biochemical studies of CA indicated that its functional groups at C-1, C-4, and C-9 were important for enzymatic activity; an X-ray crystal structure of 11β-HSD1 bound to CA at 2.6 Å resolution revealed the nature of those interactions, namely, a close-fitting and favorable interactions between the constrained CA spirocycle and the catalytic triad of 11β-HSD1. Structure-activity relationship studies culminated in the development of a superior CA analogue with improved target engagement. Furthermore, we demonstrate that CA selectively inhibits preadipocyte differentiation through 11β-HSD1 inhibition, suppressing other relevant key drivers of adipogenesis (i.e., PPARγ, PGC-1α), presumably by negatively modulating the glucocorticoid signaling pathway. The combined findings provide an in-depth evaluation of the mode of action of CA and its potential as a tool compound to study adipose tissue and its implications in metabolic syndrome.
Collapse
Affiliation(s)
| | | | | | | | | | - Ikbale El Ayachi
- Department of Medicine , The University of Tennessee Health Science Center , Memphis , Tennessee 38163 , United States
| | | | | | - Gustavo Miranda-Carboni
- Department of Medicine , The University of Tennessee Health Science Center , Memphis , Tennessee 38163 , United States
| | | | | |
Collapse
|
64
|
Luijten IHN, Cannon B, Nedergaard J. Glucocorticoids and Brown Adipose Tissue: Do glucocorticoids really inhibit thermogenesis? Mol Aspects Med 2019; 68:42-59. [PMID: 31323252 DOI: 10.1016/j.mam.2019.07.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 07/11/2019] [Indexed: 12/13/2022]
Abstract
A reduction in the thermogenic activity of brown adipose tissue (BAT) is presently discussed as a possible determinant for the development of obesity in humans. One group of endogenous factors that could potentially affect BAT activity is the glucocorticoids (e.g. cortisol). We analyse here studies examining the effects of alterations in glucocorticoid signaling on BAT recruitment and thermogenic capacity. We find that irrespective of which manipulation of glucocorticoid signaling is examined, a seemingly homogeneous picture of lowered thermogenic capacity due to glucocorticoid stimulation is apparently obtained: e.g. lowered uncoupling protein 1 (UCP1) protein levels per mg protein, and an increased lipid accumulation in BAT. However, further analyses generally indicate that these effects result from a dilution effect rather than a true decrease in total capacity; the tissue may thus be said to be in a state of pseudo-atrophy. However, under conditions of very low physiological stimulation of BAT, glucocorticoids may truly inhibit Ucp1 gene expression and consequently lower total UCP1 protein levels, but the metabolic effects of this reduction are probably minor. It is thus unlikely that glucocorticoids affect organismal metabolism and induce the development of obesity through alterations of BAT activity.
Collapse
Affiliation(s)
- Ineke H N Luijten
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.
| |
Collapse
|
65
|
Agnew EJ, Garcia-Burgos A, Richardson RV, Manos H, Thomson AJW, Sooy K, Just G, Homer NZM, Moran CM, Brunton PJ, Gray GA, Chapman KE. Antenatal dexamethasone treatment transiently alters diastolic function in the mouse fetal heart. J Endocrinol 2019; 241:279-292. [PMID: 31013474 PMCID: PMC6541236 DOI: 10.1530/joe-18-0666] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 04/23/2019] [Indexed: 12/27/2022]
Abstract
Endogenous glucocorticoid action is important in the structural and functional maturation of the fetal heart. In fetal mice, although glucocorticoid concentrations are extremely low before E14.5, glucocorticoid receptor (GR) is expressed in the heart from E10.5. To investigate whether activation of cardiac GR prior to E14.5 induces precocious fetal heart maturation, we administered dexamethasone in the drinking water of pregnant dams from E12.5 to E15.5. To test the direct effects of glucocorticoids upon the cardiovascular system we used SMGRKO mice, with Sm22-Cre-mediated disruption of GR in cardiomyocytes and vascular smooth muscle. Contrary to expectations, echocardiography showed no advancement of functional maturation of the fetal heart. Moreover, litter size was decreased 2 days following cessation of antenatal glucocorticoid exposure, irrespective of fetal genotype. The myocardial performance index and E/A wave ratio, markers of fetal heart maturation, were not significantly affected by dexamethasone treatment in either genotype. Dexamethasone treatment transiently decreased the myocardial deceleration index (MDI; a marker of diastolic function), in control fetuses at E15.5, with recovery by E17.5, 2 days after cessation of treatment. MDI was lower in SMGRKO than in control fetuses and was unaffected by dexamethasone. The transient decrease in MDI was associated with repression of cardiac GR in control fetuses following dexamethasone treatment. Measurement of glucocorticoid levels in fetal tissue and hypothalamic corticotropin-releasing hormone (Crh) mRNA levels suggest complex and differential effects of dexamethasone treatment upon the hypothalamic-pituitary-adrenal axis between genotypes. These data suggest potentially detrimental and direct effects of antenatal glucocorticoid treatment upon fetal heart function.
Collapse
Affiliation(s)
- E J Agnew
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - A Garcia-Burgos
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - R V Richardson
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - H Manos
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - A J W Thomson
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - K Sooy
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - G Just
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - N Z M Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - C M Moran
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - P J Brunton
- Centre for Discovery Brain Sciences, The University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, UK
| | - G A Gray
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
| | - K E Chapman
- Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK
- Correspondence should be addressed to K E Chapman:
| |
Collapse
|
66
|
Szelényi P, Somogyi A, Sarnyai F, Zámbó V, Simon-Szabó L, Kereszturi É, Csala M. Microsomal pre-receptor cortisol production is inhibited by resveratrol and epigallocatechin gallate through different mechanisms. Biofactors 2019; 45:236-243. [PMID: 30496642 DOI: 10.1002/biof.1477] [Citation(s) in RCA: 9] [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: 07/07/2018] [Revised: 09/24/2018] [Accepted: 10/22/2018] [Indexed: 01/11/2023]
Abstract
Local activation of cortisol in hormone target tissues is a major determinant of glucocorticoid effect. Disorders in this peripheral cortisol metabolism play an important role in the development of metabolic diseases, such as obesity or type 2 diabetes mellitus. Hence, dietary factors influencing the activity of the involved enzymes can have major impacts on the risk of the above diseases. Resveratrol and epigallocatechin gallate (EGCG), two natural polyphenols found in several nutriments and in green tea, respectively, are well-known for their antiobesity and antidiabetic activities. EGCG has been shown to interfere with microsomal cortisol production through decreasing the luminal NADPH:NADP+ ratio. The aim of this study was to clarify if resveratrol also induces such a redox shift or causes any direct enzyme inhibition that influences local cortisol production. Cortisone-cortisol conversions and changes in NADPH levels were monitored in rat liver microsomal vesicles. Cortisol production was inhibited by resveratrol in a concentration dependent manner while the intrinsic reducing and oxidizing capacity as well as the NADPH level inside the ER-derived vesicles remained unaffected. Activity measurements performed in permeabilized microsomes confirmed that resveratrol, unlike EGCG, inhibits 11β-hydroxysteroid dehydrogenase type 1 directly. Long-term moderation of pre-receptor cortisol production likely contributes to the beneficial health effects of both polyphenols. © 2018 BioFactors, 45(2):236-243, 2019.
Collapse
Affiliation(s)
- Péter Szelényi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Anna Somogyi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Farkas Sarnyai
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Veronika Zámbó
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Laura Simon-Szabó
- Pathobiochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University (MTA-SE), Budapest, Hungary
| | - Éva Kereszturi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Miklós Csala
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| |
Collapse
|
67
|
Dammann C, Stapelfeld C, Maser E. Expression and activity of the cortisol-activating enzyme 11β-hydroxysteroid dehydrogenase type 1 is tissue and species-specific. Chem Biol Interact 2019; 303:57-61. [PMID: 30796905 DOI: 10.1016/j.cbi.2019.02.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 02/03/2019] [Accepted: 02/19/2019] [Indexed: 10/27/2022]
Abstract
The microsomal enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) interconverts glucocorticoid receptor-inert cortisone (11-dehydrocorticosterone in rodents) to its receptor-active form cortisol (corticosterone in rodents). Thus, 11β-HSD1 amplifies glucocorticoid action at the tissue level. According to the current literature, dysregulation of glucocorticoid signaling may contribute to the pathogenesis of the metabolic syndrome in which regeneration of cortisol by 11β-HSD1 may be an important factor. This is why the enzyme has been very intensely investigated as a potential therapeutic target to treat metabolic complications such as obesity and diabetes type 2. However, due to controversial results from the various animal and human studies as well as from different findings with regard to tissue-specific expression and activity, the varied results unfortunately do not yield a consistent picture. Therefore, the precise role of 11β-HSD1 in the development of complications associated with the metabolic syndrome has still not been deciphered yet. Overall, the prominent role of this enzyme in the pathogenesis of the metabolic syndrome becomes more and more dubious and therefore further studies are necessary to clarify its role finally. This short review gives an overview on the main contradicting findings on the role of 11β-HSD1 in the development of visceral obesity and diabetes type 2.
Collapse
Affiliation(s)
- Christine Dammann
- Institute of Toxicology and Pharmacology for Natural Scientists, University Medical School Schleswig-Holstein, Kiel, Germany
| | - Claudia Stapelfeld
- 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
|
68
|
Shukla R, Basu AK, Mandal B, Mukhopadhyay P, Maity A, Chakraborty S, Devrabhai PK. 11β Hydroxysteroid dehydrogenase - 1 activity in type 2 diabetes mellitus: a comparative study. BMC Endocr Disord 2019; 19:15. [PMID: 30678666 PMCID: PMC6345010 DOI: 10.1186/s12902-019-0344-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 01/16/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND A comparative study of 11 β HSD 1 activity in type 2 diabetes mellitus subjects with respect to fasting blood glucose and other metabolic parameters was conducted. METHODS A case control experimental study was performed enrolling thirty type 2 diabetes mellitus patients and thirty age, gender and BMI matched controls using cortisone acetate test. RESULTS The rise of serum cortisol after oral 25 mg cortisone acetate from baseline (dexamethasone suppressed level) is higher in subjects with type 2 diabetes and is associated with exercise, BMI, SGOT but not daily calorie intake, lipid parameters and thyroid status. Fasting blood glucose after overnight 1 mg oral dexamethasone is a strong predictor of 11HSD1 activity, irrespective of presence of type 2 diabetes. CONCLUSION 11β HSD 1 activity is higher in type 2 diabetes mellitus subjects, especially those who are lean. Future 11 β HSD 1 inhibitors targeting metabolic syndrome, will be most useful in those with increased fasting blood glucose. The role of DHEAS and vitamin D status needs to be explored.
Collapse
Affiliation(s)
- Ravindra Shukla
- Department of Endocrinology & Metabolism, AIIMS, Jodhpur, 342005 India
| | | | | | | | | | | | | |
Collapse
|
69
|
Trends in Analysis of Cortisol and Its Derivatives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1140:649-664. [DOI: 10.1007/978-3-030-15950-4_39] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
70
|
Wu C, Jiang F, Wei K, Lin F, Jiang Z. Effects of Exercise Combined with Finasteride on Hormone and Ovarian Function in Polycystic Ovary Syndrome Rats. Int J Endocrinol 2019; 2019:8405796. [PMID: 31001337 PMCID: PMC6436361 DOI: 10.1155/2019/8405796] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 01/25/2019] [Accepted: 02/06/2019] [Indexed: 01/01/2023] Open
Abstract
Exercise can reduce androgen and insulin levels in polycystic ovary syndrome (PCOS) patients. Finasteride is also presumed to improve the developing follicle environment. Therefore, the aim of this study was to observe the effects of the combination of exercise and finasteride therapy on hormone levels and ovarian morphology in rats with polycystic ovary syndrome. Forty female rats were randomly divided into five groups (n = 8 each group): the PCOS sedentary group (P-Sed), PCOS exercise group (P-Ex), PCOS finasteride and sedentary group (P-FSed), and PCOS finasteride and exercise group (P-FEx), and healthy, age-matched rats were used as controls (CO). The results indicated that the levels of FINS in the P-FEx group were significantly lower than those in the P-Sed and P-FSed groups, while the ratio of fasting blood glucose (FBG)/fasting serum levels of insulin (FINS) in the P-FEx group was significantly higher than that in the P-Sed and P-FSed groups. Compared to the P-FEx group, serum levels of TT (total testosterone) in the P-Sed and P-FSed groups were significantly increased. The thickness of the follicular membrane and the number of atresia follicles in the P-FEx and CO groups were significantly lower than those in the P-Sed group, but there was no significant difference between the P-Ex and P-Sed groups. These results show that the combined usage of exercise and finasteride does not alter the effects of exercise on increasing insulin sensitivity and reducing androgen levels. There is also a synergistic effect of exercise and finasteride on the morphology of the ovary, including a reduced number of atresia follicles and thickness of the follicular membrane.
Collapse
Affiliation(s)
- Chuyan Wu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Feng Jiang
- Neonatal Department, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Ke Wei
- Medical Service Section, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Feng Lin
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhongli Jiang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
71
|
Sagmeister MS, Taylor AE, Fenton A, Wall NA, Chanouzas D, Nightingale PG, Ferro CJ, Arlt W, Cockwell P, Hardy RS, Harper L. Glucocorticoid activation by 11β-hydroxysteroid dehydrogenase enzymes in relation to inflammation and glycaemic control in chronic kidney disease: A cross-sectional study. Clin Endocrinol (Oxf) 2019; 90:241-249. [PMID: 30358903 PMCID: PMC6334281 DOI: 10.1111/cen.13889] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/02/2018] [Accepted: 10/22/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Patients with chronic kidney disease (CKD) have dysregulated cortisol metabolism secondary to changes in 11β-hydroxysteroid dehydrogenase (11β-HSD) enzymes. The determinants of this and its clinical implications are poorly defined. METHODS We performed a cross-sectional study to characterize shifts in cortisol metabolism in relation to renal function, inflammation and glycaemic control. Systemic activation of cortisol by 11β-HSD was measured as the metabolite ratio (tetrahydrocortisol [THF]+5α-tetrahydrocortisol [5αTHF])/tetrahydrocortisone (THE) in urine. RESULTS The cohort included 342 participants with a median age of 63 years, median estimated glomerular filtration rate (eGFR) of 28 mL/min/1.73 m2 and median urine albumin-creatinine ratio of 35.5 mg/mmol. (THF+5αTHF)/THE correlated negatively with eGFR (Spearman's ρ = -0.116, P = 0.032) and positively with C-reactive protein (ρ = 0.208, P < 0.001). In multivariable analysis, C-reactive protein remained a significant independent predictor of (THF+5αTHF)/THE, but eGFR did not. Elevated (THF+5αTHF)/THE was associated with HbA1c (ρ = 0.144, P = 0.008) and diabetes mellitus (odds ratio for high vs low tertile of (THF+5αTHF)/THE 2.57, 95% confidence interval 1.47-4.47). Associations with diabetes mellitus and with HbA1c among the diabetic subgroup were independent of eGFR, C-reactive protein, age, sex and ethnicity. CONCLUSIONS In summary, glucocorticoid activation by 11β-HSD in our cohort comprising a spectrum of renal function was associated with inflammation and impaired glucose control.
Collapse
Affiliation(s)
- Michael S. Sagmeister
- Institute of Inflammation and AgeingUniversity of BirminghamBirminghamUK
- Department of Renal MedicineUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | - Angela E. Taylor
- Institute of Metabolism and Systems ResearchUniversity of BirminghamBirminghamUK
| | - Anthony Fenton
- Institute of Inflammation and AgeingUniversity of BirminghamBirminghamUK
- Department of Renal MedicineUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | - Nadezhda A. Wall
- Institute of Clinical SciencesUniversity of BirminghamBirminghamUK
| | - Dimitrios Chanouzas
- Institute of Inflammation and AgeingUniversity of BirminghamBirminghamUK
- Department of Renal MedicineUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | - Peter G. Nightingale
- Institute of Translational MedicineUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | - Charles J. Ferro
- Department of Renal MedicineUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | - Wiebke Arlt
- Institute of Metabolism and Systems ResearchUniversity of BirminghamBirminghamUK
| | - Paul Cockwell
- Department of Renal MedicineUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | - Rowan S. Hardy
- Institute of Metabolism and Systems ResearchUniversity of BirminghamBirminghamUK
- Institute of Inflammation and Ageing, ARUK Rheumatoid Arthritis Centre of Excellence, and MRC ARUK Centre for Musculoskeletal AgeingUniversity of BirminghamBirminghamUK
| | - Lorraine Harper
- Department of Renal MedicineUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
- Institute of Clinical SciencesUniversity of BirminghamBirminghamUK
| |
Collapse
|
72
|
Huang M, Liu J, Sheng Y, Lv Y, Yu J, Qi H, Di W, Lv S, Zhou S, Ding G. 11β-hydroxysteroid dehydrogenase type 1 inhibitor attenuates high-fat diet induced cardiomyopathy. J Mol Cell Cardiol 2018; 125:106-116. [DOI: 10.1016/j.yjmcc.2018.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 09/06/2018] [Accepted: 10/02/2018] [Indexed: 12/29/2022]
|
73
|
White PC. Alterations of Cortisol Metabolism in Human Disorders. Horm Res Paediatr 2018; 89:320-330. [PMID: 29843121 DOI: 10.1159/000485508] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/21/2017] [Indexed: 11/19/2022] Open
Abstract
The interconversion of active and inactive corticosteroids - cortisol and cortisone, respectively, in humans - is modulated by isozymes of 11β-hydroxysteroid dehydrogenase (11-HSD). Studies of this process have provided crucial insights into glucocorticoid effects in a wide variety of tissues. The 11-HSD1 isozyme functions mainly as an oxoreductase (cortisone to cortisol) and is expressed at high levels in the liver and other glucocorticoid target tissues. Because it is required for full physiological effects of cortisol, it has emerged as a drug target for metabolic syndrome and type 2 diabetes. Mutations in the corresponding HSD11B1 gene, or in the H6PD gene encoding hexose-6-phosphate dehydrogenase (which supplies the NADPH required for the oxoreductase activity of 11-HSD1), cause apparent cortisone reductase deficiency, a rare syndrome of adrenocortical hyperactivity and hyperandrogenism. In contrast, the 11-HSD2 isozyme functions as a dehydrogenase (cortisol to cortisone) and is expressed mainly in mineralocorticoid target tissues, where it bars access of cortisol to the mineralocorticoid receptor. Mutations in the HSD11B2 gene encoding 11-HSD2 cause the syndrome of apparent mineralocorticoid excess, a severe form of familial hypertension. The role of this enzyme in the pathogenesis of common forms of low-renin hypertension remains uncertain.
Collapse
|
74
|
Hardy RS, Zhou H, Seibel MJ, Cooper MS. Glucocorticoids and Bone: Consequences of Endogenous and Exogenous Excess and Replacement Therapy. Endocr Rev 2018; 39:519-548. [PMID: 29905835 DOI: 10.1210/er.2018-00097] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/08/2018] [Indexed: 02/02/2023]
Abstract
Osteoporosis associated with long-term glucocorticoid therapy remains a common and serious bone disease. Additionally, in recent years it has become clear that more subtle states of endogenous glucocorticoid excess may have a major impact on bone health. Adverse effects can be seen with mild systemic glucocorticoid excess, but there is also evidence of tissue-specific regulation of glucocorticoid action within bone as a mechanism of disease. This review article examines (1) the role of endogenous glucocorticoids in normal bone physiology, (2) the skeletal effects of endogenous glucocorticoid excess in the context of endocrine conditions such as Cushing disease/syndrome and autonomous cortisol secretion (subclinical Cushing syndrome), and (3) the actions of therapeutic (exogenous) glucocorticoids on bone. We review the extent to which the effect of glucocorticoids on bone is influenced by variations in tissue metabolizing enzymes and glucocorticoid receptor expression and sensitivity. We consider how the effects of therapeutic glucocorticoids on bone are complicated by the effects of the underlying inflammatory disease being treated. We also examine the impact that glucocorticoid replacement regimens have on bone in the context of primary and secondary adrenal insufficiency. We conclude that even subtle excess of endogenous or moderate doses of therapeutic glucocorticoids are detrimental to bone. However, in patients with inflammatory disorders there is a complex interplay between glucocorticoid treatment and underlying inflammation, with the underlying condition frequently representing the major component underpinning bone damage.
Collapse
Affiliation(s)
- Rowan S Hardy
- University of Birmingham, Birmingham, United Kingdom
| | - Hong Zhou
- Bone Research Program, ANZAC Research Institute, Sydney, New South Wales, Australia
| | - Markus J Seibel
- Bone Research Program, ANZAC Research Institute, Sydney, New South Wales, Australia.,Department of Endocrinology and Metabolism, Concord Repatriation General Hospital, Sydney, New South Wales, Australia.,Concord Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Mark S Cooper
- Department of Endocrinology and Metabolism, Concord Repatriation General Hospital, Sydney, New South Wales, Australia.,Concord Clinical School, University of Sydney, Sydney, New South Wales, Australia.,Adrenal Steroid Laboratory, ANZAC Research Institute, Sydney, New South Wales, Australia
| |
Collapse
|
75
|
Kiso T, Sekizawa T, Uchino H, Tsukamoto M, Kakimoto S. Analgesic effects of ASP3662, a novel 11β-hydroxysteroid dehydrogenase 1 inhibitor, in rat models of neuropathic and dysfunctional pain. Br J Pharmacol 2018; 175:3784-3796. [PMID: 30006998 DOI: 10.1111/bph.14448] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 07/03/2018] [Accepted: 07/09/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Glucocorticoids are a major class of stress hormones known to participate in stress-induced hyperalgesia. Although 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1) is a key enzyme in the intracellular regeneration of glucocorticoids in the CNS, its role in pain perception has not been assessed. Here, we examined the effects of ASP3662, a novel 11β-HSD1 inhibitor, on neuropathic and dysfunctional pain. EXPERIMENTAL APPROACH The enzyme inhibitory activities and pharmacokinetics of ASP3662 were examined, and its antinociceptive effects were evaluated in models of neuropathic pain, fibromyalgia and inflammatory pain in Sprague-Dawley rats. KEY RESULTS ASP3662 inhibited human, mouse and rat 11β-HSD1 but not human 11β-HSD2, in vitro. ASP3662 had no significant effect on 87 other possible targets (enzymes, transporters and receptors). ASP3662 inhibited in vitro conversion of glucocorticoid from its inactive to active form in extracts of rat brain and spinal cord. Pharmacokinetic analysis in Sprague-Dawley rats showed that ASP3662 has CNS-penetrability and long-lasting pharmacokinetic properties. Single oral administration of ASP3662 ameliorated mechanical allodynia in spinal nerve ligation (SNL) and streptozotocin-induced diabetic rats and thermal hyperalgesia in chronic constriction nerve injury rats. ASP3662 also restored muscle pressure thresholds in reserpine-induced myalgia rats. Intrathecal administration of ASP3662 was also effective in SNL rats. However, ASP3662 had no analgesic effects in adjuvant-induced arthritis rats. CONCLUSIONS AND IMPLICATIONS ASP3662 is a potent, selective and CNS-penetrable inhibitor of 11β-HSD1. The effects of ASP3662 suggest that selective inhibition of 11β-HSD1 may be an attractive approach for the treatment of neuropathic and dysfunctional pain, as observed in fibromyalgia.
Collapse
Affiliation(s)
- Tetsuo Kiso
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - Toshihiro Sekizawa
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - Hiroshi Uchino
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - Mina Tsukamoto
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - Shuichiro Kakimoto
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| |
Collapse
|
76
|
Zou X, Ramachandran P, Kendall TJ, Pellicoro A, Dora E, Aucott RL, Manwani K, Man TY, Chapman KE, Henderson NC, Forbes SJ, Webster SP, Iredale JP, Walker BR, Michailidou Z. 11Beta-hydroxysteroid dehydrogenase-1 deficiency or inhibition enhances hepatic myofibroblast activation in murine liver fibrosis. Hepatology 2018; 67:2167-2181. [PMID: 29251794 PMCID: PMC6001805 DOI: 10.1002/hep.29734] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 11/16/2017] [Accepted: 12/07/2017] [Indexed: 12/13/2022]
Abstract
A hallmark of chronic liver injury is fibrosis, with accumulation of extracellular matrix orchestrated by activated hepatic stellate cells (HSCs). Glucocorticoids limit HSC activation in vitro, and tissue glucocorticoid levels are amplified by 11beta-hydroxysteroid dehydrogenase-1 (11βHSD1). Although 11βHSD1 inhibitors have been developed for type 2 diabetes mellitus and improve diet-induced fatty liver in various mouse models, effects on the progression and/or resolution of liver injury and consequent fibrosis have not been characterized. We have used the reversible carbon tetrachloride-induced model of hepatocyte injury and liver fibrosis to show that in two models of genetic 11βHSD1 deficiency (global, Hsd11b1-/- , and hepatic myofibroblast-specific, Hsd11b1fl/fl /Pdgfrb-cre) 11βHSD1 pharmacological inhibition in vivo exacerbates hepatic myofibroblast activation and liver fibrosis. In contrast, liver injury and fibrosis in hepatocyte-specific Hsd11b1fl/fl /albumin-cre mice did not differ from that of controls, ruling out 11βHSD1 deficiency in hepatocytes as the cause of the increased fibrosis. In primary HSC culture, glucocorticoids inhibited expression of the key profibrotic genes Acta2 and Col1α1, an effect attenuated by the 11βHSD1 inhibitor [4-(2-chlorophenyl-4-fluoro-1-piperidinyl][5-(1H-pyrazol-4-yl)-3-thienyl]-methanone. HSCs from Hsd11b1-/- and Hsd11b1fl/fl /Pdgfrb-cre mice expressed higher levels of Acta2 and Col1α1 and were correspondingly more potently activated. In vivo [4-(2-chlorophenyl-4-fluoro-1-piperidinyl][5-(1H-pyrazol-4-yl)-3-thienyl]-methanone administration prior to chemical injury recapitulated findings in Hsd11b1-/- mice, including greater fibrosis. CONCLUSION 11βHSD1 deficiency enhances myofibroblast activation and promotes initial fibrosis following chemical liver injury; hence, the effects of 11βHSD1 inhibitors on liver injury and repair are likely to be context-dependent and deserve careful scrutiny as these compounds are developed for chronic diseases including metabolic syndrome and dementia. (Hepatology 2018;67:2167-2181).
Collapse
Affiliation(s)
- Xiantong Zou
- BHF Centre for Cardiovascular ScienceThe University of EdinburghEdinburghUK
- MRC Centre for Inflammation ResearchThe University of EdinburghEdinburghUK
| | | | - Timothy J. Kendall
- MRC Centre for Inflammation ResearchThe University of EdinburghEdinburghUK
| | | | - Elena Dora
- MRC Centre for Inflammation ResearchThe University of EdinburghEdinburghUK
| | - Rebecca L. Aucott
- MRC Centre for Inflammation ResearchThe University of EdinburghEdinburghUK
| | - Kajal Manwani
- BHF Centre for Cardiovascular ScienceThe University of EdinburghEdinburghUK
| | - Tak Yung Man
- BHF Centre for Cardiovascular ScienceThe University of EdinburghEdinburghUK
| | - Karen E. Chapman
- BHF Centre for Cardiovascular ScienceThe University of EdinburghEdinburghUK
| | - Neil C. Henderson
- MRC Centre for Inflammation ResearchThe University of EdinburghEdinburghUK
| | - Stuart J. Forbes
- MRC Centre for Regenerative MedicineQueen's Medical Research InstituteEdinburghUK
| | - Scott P. Webster
- BHF Centre for Cardiovascular ScienceThe University of EdinburghEdinburghUK
| | - John P. Iredale
- MRC Centre for Inflammation ResearchThe University of EdinburghEdinburghUK
| | - Brian R. Walker
- BHF Centre for Cardiovascular ScienceThe University of EdinburghEdinburghUK
| | - Zoi Michailidou
- BHF Centre for Cardiovascular ScienceThe University of EdinburghEdinburghUK
- MRC Centre for Inflammation ResearchThe University of EdinburghEdinburghUK
| |
Collapse
|
77
|
Exercise activates the PI3K-AKT signal pathway by decreasing the expression of 5α-reductase type 1 in PCOS rats. Sci Rep 2018; 8:7982. [PMID: 29789599 PMCID: PMC5964186 DOI: 10.1038/s41598-018-26210-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 05/03/2018] [Indexed: 12/31/2022] Open
Abstract
Hyperandrogenism and hyperinsulinemia are main clinical endocrine features of PCOS. Exercise can adjust the androgen level, as well as increase the sensitivity of insulin by activating PI3K-Akt insulin signaling pathways. 5αR1 has certain effects on insulin resistance and can synthesize dihydrotestosterone by metabolizing testosterone. So 5αR1 may be the target of androgen and insulin for exercise-induced regulation. To investigate the role of 5αR1 in the PI3K-Akt signaling pathway in skeletal muscle of PCOS rats activated by exercise, fifty-four female rats were randomly divided into the PCOS group (n = 42) and the control group(n = 12). After injection of testosterone propionate for 28 days, the remaining 36 rats in the PCOS group were randomly assigned to six groups: the sedentary group (PS, n = 6), sedentary and 5αRI (5α-reductase inhibitor) group (PS + RI, n = 6), sedentary and 5αR2I (5α-reductase type 2 selective inhibitor) group (PS + R2I, n = 6), exercise group (PE, n = 6), exercise and 5αRI group (PE + RI, n = 6), and exercise and 5αR2I group (PE + R2I, n = 6). The rats undergoing exercise were trained to swim for 14 days. Finasteride (5α-reductase type 2 selective inhibitor) and dutasteride (5α-reductase inhibitor) were administered once daily and were dosed based on weight. At the end, the expression of 5αR1 proteins, the phosphorylation level of PI3K and AKT, were determined by Western blot. The PCOS non-exercise group and the PE + RI group displayed significantly lower phosphorylation of Akt, PI3K p85 and GLUT4 expression, while in the PE + R2I group, the level of Akt phosphorylation and PI3K p85 expression was significantly higher than that of the PCOS non-exercise group and the PE + RI group. In summary, our study demonstrated that exercise can activate the PI3K/AKT signal pathway of PCOS rats by decreasing the expression of 5αR1.
Collapse
|
78
|
Abstract
PURPOSE OF REVIEW The use of 5-alpha reductase inhibitors (5ARIs) for the treatment of benign prostatic hyperplasia (BPH) and other diseases has been proposed and studied. However, the controversy about its benefits and harms for other diseases has persisted. In this review, we will discuss the newly identified effects of 5ARIs based on recently published studies. RECENT FINDINGS These drugs are currently recommended in clinical guidelines for BPH. However, the reporting of adverse effects, including sexual dysfunction as well as neurologic, endocrine, and cardiovascular effects, have been controversial. There are reports of additional effects of 5ARI in prostate cancer and bladder cancer. Although 5ARIs have been prescribed for the treatment of androgenic alopecia (AGA), postfinasteride syndrome can result, with symptoms that range from sexual dysfunction to muscle atrophy. SUMMARY Clinical applications of 5ARIs have been established for the treatment of BPH and AGA from a series of randomized controlled trials. The adverse effects of 5ARIs affect only a small proportion of treated patients and can be resolved with discontinued treatment. It will be necessary to establish the mechanism by which 5ARIs elicit these effects through better designed studies.
Collapse
|
79
|
Seo M, Islam SA, Moon SS. Acute anti-obesity effects of intracerebroventricular 11β-HSD1 inhibitor administration in diet-induced obese mice. J Neuroendocrinol 2018; 30:e12580. [PMID: 29418022 DOI: 10.1111/jne.12580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 01/09/2018] [Accepted: 02/01/2018] [Indexed: 11/30/2022]
Abstract
The hypothalamus is the regulatory centre of both appetite and energy balance and endoplasmic reticulum (ER) stress in the hypothalamus is involved in the pathogenesis of obesity. Recently, inhibition of 11 β hydroxysteroid dehydrogenase type1 (11β-HSD1) was reported to have an anti-obesity effect by reducing fat mass. However, the link between the role of 11β-HSD1 in the hypothalamus and obesity has yet to be determined. In the present study, embryonal primary hypothalamic neurones and high-fat diet (HFD) fed mice were used to investigate the anorexigenic effects of 11β-HSD1 inhibitors both in vitro and in vivo. In hypothalamic neurones, carbenoxolone (a non selecitve 11β-HSD inhibitor) alleviated ER stress and ER stress-induced neuropeptide alterations. In HFD mice, i.c.v. administration of carbenoxolone or KR67500 (nonselective and selective 11β-HSD1 inhibitors, respectively) was associated with less weight gain compared to control mice for 24 hours after treatment, presumably by reducing food intake. Furthermore, glucose regulated protein (Grp78), spliced X-box binding protein (Xbp-1s), c/EBP homologous protein (chop) and ER DnaJ homologue protein (Erdj4) expression was decreased in the hypothalami of mice administrated 11β-HSD1 inhibitors compared to controls. Conversely, the phosphorylation of protein kinase B (PKB/Akt), signal transducer and activator of transcription 3 (Stat3), mitogen-activated protein kinase (MAPK/ERK) and S6 kinase1 (S6K1) in the hypothalamus was induced more in mice treated using the same regimes. In conclusion, acute 11β-HSD1 inhibition in the hypothalamus could reduce food intake by decreasing ER stress and increasing insulin, leptin, and mammalian target of rapamycin complex 1 (mTORC1) signalling.
Collapse
Affiliation(s)
- M Seo
- Medical Institute of Dongguk University, Gyeongju, South Korea
- Department of Agricultural Biology, National Institute of Agricultural Sciences, Rural Development Administration, Wanju-gun, South Korea
| | - S A Islam
- Department of Internal Medicine, College of Medicine, Dongguk University, Gyeongju, South Korea
| | - S-S Moon
- Medical Institute of Dongguk University, Gyeongju, South Korea
- Department of Internal Medicine, College of Medicine, Dongguk University, Gyeongju, South Korea
| |
Collapse
|
80
|
Enhanced hexose-6-phosphate dehydrogenase expression in adipose tissue may contribute to diet-induced visceral adiposity. Int J Obes (Lond) 2018; 42:1999-2011. [PMID: 29568102 PMCID: PMC6105561 DOI: 10.1038/s41366-018-0041-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/30/2017] [Accepted: 12/27/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Visceral fat accumulation increases the risk of developing type 2 diabetes and metabolic syndrome, and is associated with excessive glucocorticoids (GCs). Fat depot-specific GC action is tightly controlled by 11ß-hydroxysteroid dehydrogenase (11ß-HSD1) coupled with the enzyme hexose-6-phosphate dehydrogenase (H6PDH). Mice with inactivation or activation of H6PDH genes show altered adipose 11ß-HSD1 activity and lipid storage. We hypothesized that adipose tissue H6PDH activation is a leading cause for the visceral obesity and insulin resistance. Here, we explored the role and possible mechanism of enhancing adipose H6PDH in the development of visceral adiposity in vivo. METHODS We investigated the potential contribution of adipose H6PDH activation to the accumulation of visceral fat by characterization of visceral fat obese gene expression profiles, fat distribution, adipocyte metabolic molecules, and abdominal fat-specific GC signaling mechanisms underlying the diet-induced visceral obesity and insulin resistance in H6PDH transgenic mice fed a standard of high-fat diet (HFD). RESULTS Transgenic H6PDH mice display increased abdominal fat accumulation, which is paralleled by elevated lipid synthesis associated with induction of lipogenic transcriptor C/EBPα and PPARγ mRNA levels within adipose tissue. Transgenic H6PDH mice fed a high-fat diet (HFD) gained more abdominal visceral fat mass coupled with activation of GSK3β and induction of XBP1/IRE1α, but reduced pThr308 Akt/PKB content and browning gene CD137 and GLUT4 mRNA levels within the visceral adipose tissue than WT controls. HFD-fed H6PDH transgenic mice also had impaired insulin sensitivity and exhibited elevated levels of intra-adipose GCs with induction of adipose 11ß-HSD1. CONCLUSION These data provide the first in vivo mechanistic evidence for the adverse metabolic effects of adipose H6PDH activation on visceral fat distribution, fat metabolism, and adipocyte function through enhancing 11ß-HSD1-driven intra-adipose GC action.
Collapse
|
81
|
Intrauterine growth restriction combined with a maternal high-fat diet increased adiposity and serum corticosterone levels in adult rat offspring. J Dev Orig Health Dis 2018; 9:315-328. [DOI: 10.1017/s2040174418000016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
AbstractIntrauterine growth restriction (IUGR) and fetal exposure to a maternal high-fat diet (HFD) independently increase the risk of developing obesity in adulthood. Excess glucocorticoids increase obesity. We hypothesized that surgically induced IUGR combined with an HFD would increase adiposity and glucocorticoids more than in non-IUGR offspring combined with the same HFD, findings that would persist despite weaning to a regular diet. Non-IUGR (N) and IUGR (I) rat offspring from dams fed either regular rat chow (R) or an HFD (H) were weaned to either a regular rat chow or an HFD. For non-IUGR and IUGR rats, this study design resulted in three diet groups: offspring from dams fed a regular diet and weaned to a regular diet (NRR and IRR), offspring rats from dams fed an HFD and weaned to a regular diet (NHR and IHR) and offspring from dams fed an HFD and weaned to an HFD (NHH and IHH). Magnetic resonance imaging or fasting visceral and subcutaneous adipose tissue collection occurred at postnatal day 60. IHH male rats had greater adiposity than NHH males, findings that were only partly normalized by weaning to a regular chow. IHH male rats had a 10-fold increase in serum corticosterone levels. IHH female rats had increased adiposity and serum triglycerides. We conclude that IUGR combined with an HFD throughout life increased adiposity, glucocorticoids and triglycerides in a sex-specific manner. Our data suggest that one mechanism through which the perinatal environment programs increased adiposity in IHH male rats may be via increased systemic glucocorticoids.
Collapse
|
82
|
Fine NHF, Doig CL, Elhassan YS, Vierra NC, Marchetti P, Bugliani M, Nano R, Piemonti L, Rutter GA, Jacobson DA, Lavery GG, Hodson DJ. Glucocorticoids Reprogram β-Cell Signaling to Preserve Insulin Secretion. Diabetes 2018; 67:278-290. [PMID: 29203512 PMCID: PMC5780059 DOI: 10.2337/db16-1356] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 11/16/2017] [Indexed: 12/19/2022]
Abstract
Excessive glucocorticoid exposure has been shown to be deleterious for pancreatic β-cell function and insulin release. However, glucocorticoids at physiological levels are essential for many homeostatic processes, including glycemic control. We show that corticosterone and cortisol and their less active precursors 11-dehydrocorticosterone (11-DHC) and cortisone suppress voltage-dependent Ca2+ channel function and Ca2+ fluxes in rodent as well as in human β-cells. However, insulin secretion, maximal ATP/ADP responses to glucose, and β-cell identity were all unaffected. Further examination revealed the upregulation of parallel amplifying cAMP signals and an increase in the number of membrane-docked insulin secretory granules. Effects of 11-DHC could be prevented by lipotoxicity and were associated with paracrine regulation of glucocorticoid activity because global deletion of 11β-hydroxysteroid dehydrogenase type 1 normalized Ca2+ and cAMP responses. Thus, we have identified an enzymatically amplified feedback loop whereby glucocorticoids boost cAMP to maintain insulin secretion in the face of perturbed ionic signals. Failure of this protective mechanism may contribute to diabetes in states of glucocorticoid excess, such as Cushing syndrome, which are associated with frank dyslipidemia.
Collapse
Affiliation(s)
- Nicholas H F Fine
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, U.K
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, U.K
| | - Craig L Doig
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, U.K
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, U.K
| | - Yasir S Elhassan
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, U.K
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, U.K
| | - Nicholas C Vierra
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Marco Bugliani
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rita Nano
- Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Department of Medicine, Imperial College London, London, U.K
| | - David A Jacobson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, U.K
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, U.K
| | - David J Hodson
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, U.K.
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, U.K
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, U.K
| |
Collapse
|
83
|
Hariri M, Haghighatdoost F. Effect of Curcumin on Anthropometric Measures: A Systematic Review on Randomized Clinical Trials. J Am Coll Nutr 2018; 37:215-222. [PMID: 29313748 DOI: 10.1080/07315724.2017.1392263] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Curcumin is an active constituent of turmeric. Recently, scientists have suggested that curcumin can be used in weight reduction. We performed a systematic review based on randomized controlled trials to assess the effects of curcumin supplementation on anthropometric variables. METHODS We searched databases including PubMed, Embase, Web of Science, Scopus, and Google Scholar up to August 2017. Randomized clinical trials assessing the effects of curcumin on anthropometric parameters in human adults were included. RESULT Eight randomized clinical trials were allowed to be included in the systematic review. Five articles used the regular form of curcumin with short follow-up duration and did not indicate any significant effect on anthropometric measures, while three articles with significant results used either the more bioavailable form of curcumin or a longer intervention duration. CONCLUSION Randomized clinical trials related to curcumin effect on weight are limited but their result indicated useful effect of curcumin on weight. It seems that the bioavailable form of curcumin can reduce obesity and overweight. Further articles with longer duration of intervention and different forms of curcumin supplementation are necessary before any recommendation is made for clinical use of these interventions.
Collapse
Affiliation(s)
- Mitra Hariri
- a Department of Basic Medical Sciences , Neyshabur University of Medical Sciences , Neyshabur , Iran
| | - Fahimeh Haghighatdoost
- b Food Security Research Center , Isfahan University of Medical Sciences , Isfahan , Iran.,c Department of Community Nutrition, School of Nutrition and Food Science , Isfahan University of Medical Sciences , Isfahan , Iran
| |
Collapse
|
84
|
de Kloet AD, Herman JP. Fat-brain connections: Adipocyte glucocorticoid control of stress and metabolism. Front Neuroendocrinol 2018; 48:50-57. [PMID: 29042142 DOI: 10.1016/j.yfrne.2017.10.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 01/08/2023]
Abstract
Glucocorticoids act via multiple mechanisms to mobilize energy for maintenance and restoration of homeostasis. In adipose tissue, glucocorticoids can promote lipolysis and facilitate adipocyte differentiation/growth, serving both energy-mobilizing and restorative processes during negative energy balance. Recent data suggest that adipose-dependent feedback may also be involved in regulation of stress responses. Adipocyte glucocorticoid receptor (GR) deletion causes increased HPA axis stress reactivity, due to a loss of negative feedback signals into the CNS. The fat-to-brain signal may be mediated by neuronal mechanisms, release of adipokines or increased lipolysis. The ability of adipose GRs to inhibit psychogenic as well as metabolic stress responses suggests that (1) feedback regulation of the HPA axis occurs across multiple bodily compartments, and (2) fat tissue integrates psychogenic stress signals. These studies support a link between stress biology and energy metabolism, a connection that has clear relevance for numerous disease states and their comorbidities.
Collapse
Affiliation(s)
- Annette D de Kloet
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32611, United States
| | - James P Herman
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, United States.
| |
Collapse
|
85
|
Quantification of 11β-hydroxysteroid dehydrogenase 1 kinetics and pharmacodynamic effects of inhibitors in brain using mass spectrometry imaging and stable-isotope tracers in mice. Biochem Pharmacol 2017; 148:88-99. [PMID: 29248595 PMCID: PMC5821700 DOI: 10.1016/j.bcp.2017.12.013] [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: 10/26/2017] [Accepted: 12/13/2017] [Indexed: 12/22/2022]
Abstract
11β-Hydroxysteroid dehydrogenase 1 (11β-HSD1; EC 1.1.1.146) generates active glucocorticoid hormones. Small molecule inhibitors have been developed to target 11β-HSD1 for the treatment of dementia; these must enter brain subregions, such as the hippocampus, to be effective. We previously reported mass spectrometry imaging measurement of murine tissue steroids, and deuterated steroid tracer infusion quantification of 11β-HSD1 turnover in humans. Here, these tools are combined to assess tissue pharmacokinetics and pharmacodynamics of an 11β-HSD1 inhibitor that accesses the brain. [9,11,12,12-2H]4-Cortisol was infused (1.75 mg/day) by minipump for 2 days into C57Bl6 mice (male, age 12 weeks, n = 3/group) after which an 11β-HSD1 inhibitor (UE2316) was administered (25 mg/kg oral gavage) and animals culled immediately or 1, 2 and 4 h post-dosing. Mice with global genetic disruption of Hsd11B1 were studied similarly. Turnover of d4-cortisol to d3-cortisone (by loss of the 11-deuterium) and regeneration of d3-cortisol (by 11β-HSD1-mediated reduction) were assessed in plasma, liver and brain using matrix assisted laser desorption ionization coupled to Fourier transform cyclotron resonance mass spectrometry. The tracer d4-cortisol was detected in liver and brain following a two day infusion. Turnover to d3-cortisone and on to d3-cortisol was slower in brain than liver. In contrast, d3-cortisol was not detected in mice lacking 11β-HSD1. UE2316 impaired d3-cortisol generation measured in whole body (assessed in plasma; 53.1% suppression in rate of appearance in d3-cortisol), liver and brain. Differential inhibition in brain regions was observed; active glucocorticoids were suppressed to a greater in extent hippocampus or cortex than in amygdala. These data confirm that the contribution of 11β-HSD1 to the tissue glucocorticoid pool, and the consequences of enzyme inhibition on active glucocorticoid concentrations, are substantial, including in the brain. They further demonstrate the value of mass spectrometry imaging in pharmacokinetic and pharmacodynamic studies.
Collapse
|
86
|
Abstract
The metabolic syndrome describes a clustering of risk factors—visceral obesity, dyslipidaemia, insulin resistance, and salt-sensitive hypertension—that increases mortality related to cardiovascular disease, type 2 diabetes, cancer, and non-alcoholic fatty liver disease. The prevalence of these concurrent comorbidities is ~ 25–30% worldwide, and metabolic syndrome therefore presents a significant global public health burden. Evidence from clinical and preclinical studies indicates that glucocorticoid excess is a key causal feature of metabolic syndrome. This is not increased systemic in circulating cortisol, rather increased bioavailability of active glucocorticoids within tissues. This review examines the role of covert glucocorticoid excess on the hypertension of the metabolic syndrome. Here, the role of the 11β-hydroxysteroid dehydrogenase enzymes, which exert intracrine and paracrine control over glucocorticoid signalling, is examined. 11βHSD1 amplifies glucocorticoid action in cells and contributes to hypertension through direct and indirect effects on the kidney and vasculature. The deactivation of glucocorticoid by 11βHSD2 controls ligand access to glucocorticoid and mineralocorticoid receptors: loss of function promotes salt retention and hypertension. As for hypertension in general, high blood pressure in the metabolic syndrome reflects a complex interaction between multiple systems. The clear association between high dietary salt, glucocorticoid production, and metabolic disorders has major relevance for human health and warrants systematic evaluation.
Collapse
Affiliation(s)
- Matthew A Bailey
- The British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
87
|
Vandermosten L, De Geest C, Knoops S, Thijs G, Chapman KE, De Bosscher K, Opdenakker G, Van den Steen PE. 11β-hydroxysteroid dehydrogenase type 1 has no effect on survival during experimental malaria but affects parasitemia in a parasite strain-specific manner. Sci Rep 2017; 7:13835. [PMID: 29062028 PMCID: PMC5653823 DOI: 10.1038/s41598-017-14288-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 10/06/2017] [Indexed: 01/01/2023] Open
Abstract
Malaria is a global disease associated with considerable mortality and morbidity. An appropriately balanced immune response is crucial in determining the outcome of malarial infection. The glucocorticoid (GC) metabolising enzyme, 11β-hydroxysteroid dehydrogenase-1 (11β-HSD1) converts intrinsically inert GCs into active GCs. 11β-HSD1 shapes endogenous GC action and is immunomodulatory. We investigated the role of 11β-HSD1 in two mouse models of malaria. 11β-HSD1 deficiency did not affect survival after malaria infection, but it increased disease severity and parasitemia in mice infected with Plasmodium chabaudi AS. In contrast, 11β-HSD1 deficiency rather decreased parasitemia in mice infected with the reticulocyte-restricted parasite Plasmodium berghei NK65 1556Cl1. Malaria-induced antibody production and pathology were unaltered by 11β-HSD1 deficiency though plasma levels of IL-4, IL-6 and TNF-α were slightly affected by 11β-HSD1 deficiency, dependent on the infecting parasite. These data suggest that 11β-HSD1 is not crucial for survival of experimental malaria, but alters its progression in a parasite strain-specific manner.
Collapse
Affiliation(s)
- L Vandermosten
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven - University of Leuven, Leuven, Belgium
| | - C De Geest
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven - University of Leuven, Leuven, Belgium
| | - S Knoops
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven - University of Leuven, Leuven, Belgium
| | - G Thijs
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven - University of Leuven, Leuven, Belgium
| | - K E Chapman
- University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - K De Bosscher
- Receptor Research Laboratories, Nuclear Receptor Lab, VIB-UGent Center for Medical Biotechnology, Gent, Belgium
| | - G Opdenakker
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven - University of Leuven, Leuven, Belgium
| | - P E Van den Steen
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven - University of Leuven, Leuven, Belgium.
| |
Collapse
|
88
|
Loerz C, Staab-Weijnitz C, Huebbe P, Giller K, Metges C, Rimbach G, Maser E. Regulation of 11β-hydroxysteroid dehydrogenase type 1 following caloric restriction and re-feeding is species dependent. Chem Biol Interact 2017; 276:95-104. [DOI: 10.1016/j.cbi.2017.02.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/17/2017] [Accepted: 02/26/2017] [Indexed: 01/22/2023]
|
89
|
Yao F, Chen L, Fan Z, Teng F, Zhao Y, Guan F, Zhang M, Liu Y. Interplay between H6PDH and 11β-HSD1 implicated in the pathogenesis of type 2 diabetes mellitus. Bioorg Med Chem Lett 2017; 27:4107-4113. [PMID: 28751144 DOI: 10.1016/j.bmcl.2017.07.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/08/2017] [Accepted: 07/14/2017] [Indexed: 11/19/2022]
Abstract
Extensive studies have been performed on the role of 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1) in metabolic diseases. Our previous study reported glucose could directly regulate hexose-6-phosphate dehydrogenase (H6PDH) and 11β-HSD1. Recently, we further investigated the interplay of H6PDH and 11β-HSD1 and their roles in hepatic gluconeogenesis and insulin resistance to elucidate the importance of H6PDH and 11β-HSD1 in pathogenesis of type 2 diabetes mellitus (T2DM). T2DM rats model and H6PDH or 11β-HSD1 siRNA transfected in CBRH-7919 cells were used to explore the effect of H6PDH and 11β-HSD1 in T2DM. The results showed that the expression and activity of H6PDH and 11β-HSD1 in livers of diabetic rats were increased, with the expressions of PEPCK and G6Pase or liver corticosterone increased apparently. It also showed that H6PDH siRNA and 11β-HSD1 siRNA could inhibit the protein expression and enzyme activity by each other. With H6PDH siRNA, the enhancement of gluconeogenesis was blocked and insulin resistance stimulated by corticosterone was reduced. H6PDH and 11β-HSD1 might be the effective and prospective targets for T2DM and metabolic syndromes, based on the interplay between these two enzymes.
Collapse
Affiliation(s)
- Fan Yao
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Li Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; School of Nursing, Jilin University, Changchun 130021, China
| | - Zheng Fan
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Fei Teng
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yali Zhao
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Fengying Guan
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Ming Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Yanjun Liu
- Division of Endocrinology, Metabolism and Molecular Medicine, Charles R. Drew University of Medicine and Sciences, UCLA School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
90
|
Teich T, Pivovarov JA, Porras DP, Dunford EC, Riddell MC. Curcumin limits weight gain, adipose tissue growth, and glucose intolerance following the cessation of exercise and caloric restriction in rats. J Appl Physiol (1985) 2017; 123:1625-1634. [PMID: 28839007 DOI: 10.1152/japplphysiol.01115.2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Weight regain, adipose tissue growth, and insulin resistance can occur within days after the cessation of regular dieting and exercise. This phenomenon has been attributed, in part, to the actions of stress hormones as well as local and systemic inflammation. We investigated the effect of curcumin, a naturally occurring polyphenol known for its anti-inflammatory properties and inhibitory action on 11β-HSD1 activity, on preserving metabolic health and limiting adipose tissue growth following the cessation of daily exercise and caloric restriction (CR). Sprague-Dawley rats (6-7 wk old) underwent a "training" protocol of 24-h voluntary running wheel access and CR (15-20 g/day; ~50-65% of ad libitum intake) for 3 wk ("All Trained") or were sedentary and fed ad libitum ("Sed"). After 3 wk, All Trained were randomly divided into one group which was terminated immediately ("Trained"), and two detrained groups which had their wheels locked and were reintroduced to ad libitum feeding for 1 wk. The wheel locked groups received either a daily gavage of a placebo ("Detrained + Placebo") or curcumin (200 mg/kg) ("Detrained + Curcumin"). Cessation of daily CR and exercise caused an increase in body mass, as well as a 9- to 14-fold increase in epididymal, perirenal, and inguinal adipose tissue mass, all of which were attenuated by curcumin ( P < 0.05). Insulin area under the curve (AUC) during an oral glucose tolerance test, HOMA-IR, and C-reactive protein (CRP) were elevated 6-, 9-, and 2-fold, respectively, in the Detrained + Placebo group vs. the Trained group (all P < 0.05). Curcumin reduced insulin AUC, HOMA-IR, and CRP vs. the placebo group (all P < 0.05). Our results indicate that curcumin has a protective effect against weight regain and impaired metabolic control following a successful period of weight loss through diet and exercise, perhaps via inhibition of glucocorticoid action and inflammation. NEW & NOTEWORTHY Weight regain after dieting and exercise is a common phenomenon plaguing many individuals. The biological mechanisms underlying weight regain are incompletely understood and are likely multifactorial. In this paper, we examined the metabolic implications of curcumin, a compound known for its anti-inflammatory properties and inhibitory action on the enzyme 11β-HSD1, in a rodent model of adiposity rebound after the cessation of diet and exercise.
Collapse
Affiliation(s)
- Trevor Teich
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University , Toronto, Ontario , Canada
| | - Jacklyn A Pivovarov
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University , Toronto, Ontario , Canada
| | - Deanna P Porras
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University , Toronto, Ontario , Canada
| | - Emily C Dunford
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University , Toronto, Ontario , Canada
| | - Michael C Riddell
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University , Toronto, Ontario , Canada
| |
Collapse
|
91
|
Rocamora-Reverte L, Reichardt HM, Villunger A, Wiegers GJ. T-cell autonomous death induced by regeneration of inert glucocorticoid metabolites. Cell Death Dis 2017; 8:e2948. [PMID: 28726773 PMCID: PMC5550885 DOI: 10.1038/cddis.2017.344] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 06/14/2017] [Accepted: 06/20/2017] [Indexed: 11/09/2022]
Abstract
Glucocorticoids (GC) are essential regulators of T-cell development and function. Activation of the immune system increases systemic adrenal-derived GC levels which downregulate immune activity as part of a negative feedback control system. Increasing evidence shows, however, that GC can also be derived from extra-adrenal sources such as the thymus or intestine, thus providing local control of GC-mediated effects. The thymus reportedly produces GC, but whether thymic epithelial cells or thymocytes produce GC acting either in an autocrine or paracrine fashion is not clear. We studied the expression of two main enzymes involved in de novo GC synthesis, CYP11A1 and CYP11B1, as well as the expression and activity of HSD11B1, an enzyme catalyzing interconversion of inert GC metabolites with active GC. While we found no evidence of de novo GC synthesis in both thymocytes and peripheral T cells, abundant regeneration of GC from the inactive metabolite 11-dehydrocorticosterone was detectable. Irrespective of their maturation stage, T cells that produced GC in this manner undergo autonomous cell death as this was blocked when glucocorticoid receptor-deficient T cells were treated with GC metabolites. These results indicate that both immature and mature T cells possess the capacity to undergo apoptosis in response to intrinsically generated GC. Consequently, positive selection of thymocytes, as well as survival of peripheral T cells may depend on TCR-induced escape of otherwise HSD11B1-driven autonomous T-cell death.
Collapse
Affiliation(s)
| | - Holger M Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Andreas Villunger
- Biocenter, Division of Developmental Immunology, Medical University, Innsbruck, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - GJan Wiegers
- Biocenter, Division of Developmental Immunology, Medical University, Innsbruck, Austria
| |
Collapse
|
92
|
Geisler CE, Renquist BJ. Hepatic lipid accumulation: cause and consequence of dysregulated glucoregulatory hormones. J Endocrinol 2017; 234:R1-R21. [PMID: 28428362 DOI: 10.1530/joe-16-0513] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 04/20/2017] [Indexed: 12/11/2022]
Abstract
Fatty liver can be diet, endocrine, drug, virus or genetically induced. Independent of cause, hepatic lipid accumulation promotes systemic metabolic dysfunction. By acting as peroxisome proliferator-activated receptor (PPAR) ligands, hepatic non-esterified fatty acids upregulate expression of gluconeogenic, beta-oxidative, lipogenic and ketogenic genes, promoting hyperglycemia, hyperlipidemia and ketosis. The typical hormonal environment in fatty liver disease consists of hyperinsulinemia, hyperglucagonemia, hypercortisolemia, growth hormone deficiency and elevated sympathetic tone. These endocrine and metabolic changes further encourage hepatic steatosis by regulating adipose tissue lipolysis, liver lipid uptake, de novo lipogenesis (DNL), beta-oxidation, ketogenesis and lipid export. Hepatic lipid accumulation may be induced by 4 separate mechanisms: (1) increased hepatic uptake of circulating fatty acids, (2) increased hepatic de novo fatty acid synthesis, (3) decreased hepatic beta-oxidation and (4) decreased hepatic lipid export. This review will discuss the hormonal regulation of each mechanism comparing multiple physiological models of hepatic lipid accumulation. Nonalcoholic fatty liver disease (NAFLD) is typified by increased hepatic lipid uptake, synthesis, oxidation and export. Chronic hepatic lipid signaling through PPARgamma results in gene expression changes that allow concurrent activity of DNL and beta-oxidation. The importance of hepatic steatosis in driving systemic metabolic dysfunction is highlighted by the common endocrine and metabolic disturbances across many conditions that result in fatty liver. Understanding the mechanisms underlying the metabolic dysfunction that develops as a consequence of hepatic lipid accumulation is critical to identifying points of intervention in this increasingly prevalent disease state.
Collapse
Affiliation(s)
- Caroline E Geisler
- School of Animal and Comparative Biomedical SciencesUniversity of Arizona, Tucson, Arizona, USA
| | - Benjamin J Renquist
- School of Animal and Comparative Biomedical SciencesUniversity of Arizona, Tucson, Arizona, USA
| |
Collapse
|
93
|
Beck KR, Kaserer T, Schuster D, Odermatt A. Virtual screening applications in short-chain dehydrogenase/reductase research. J Steroid Biochem Mol Biol 2017; 171:157-177. [PMID: 28286207 PMCID: PMC6831487 DOI: 10.1016/j.jsbmb.2017.03.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/06/2017] [Accepted: 03/08/2017] [Indexed: 02/06/2023]
Abstract
Several members of the short-chain dehydrogenase/reductase (SDR) enzyme family play fundamental roles in adrenal and gonadal steroidogenesis as well as in the metabolism of steroids, oxysterols, bile acids, and retinoids in peripheral tissues, thereby controlling the local activation of their cognate receptors. Some of these SDRs are considered as promising therapeutic targets, for example to treat estrogen-/androgen-dependent and corticosteroid-related diseases, whereas others are considered as anti-targets as their inhibition may lead to disturbances of endocrine functions, thereby contributing to the development and progression of diseases. Nevertheless, the physiological functions of about half of all SDR members are still unknown. In this respect, in silico tools are highly valuable in drug discovery for lead molecule identification, in toxicology screenings to facilitate the identification of hazardous chemicals, and in fundamental research for substrate identification and enzyme characterization. Regarding SDRs, computational methods have been employed for a variety of applications including drug discovery, enzyme characterization and substrate identification, as well as identification of potential endocrine disrupting chemicals (EDC). This review provides an overview of the efforts undertaken in the field of virtual screening supported identification of bioactive molecules in SDR research. In addition, it presents an outlook and addresses the opportunities and limitations of computational modeling and in vitro validation methods.
Collapse
Affiliation(s)
- Katharina R Beck
- Swiss Center for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Teresa Kaserer
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), Computer Aided Molecular Design Group, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Daniela Schuster
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), Computer Aided Molecular Design Group, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| | - Alex Odermatt
- Swiss Center for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| |
Collapse
|
94
|
Mylonas KJ, Turner NA, Bageghni SA, Kenyon CJ, White CI, McGregor K, Kimmitt RA, Sulston R, Kelly V, Walker BR, Porter KE, Chapman KE, Gray GA. 11β-HSD1 suppresses cardiac fibroblast CXCL2, CXCL5 and neutrophil recruitment to the heart post MI. J Endocrinol 2017; 233:315-327. [PMID: 28522730 PMCID: PMC5457506 DOI: 10.1530/joe-16-0501] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 04/11/2017] [Indexed: 12/20/2022]
Abstract
We have previously demonstrated that neutrophil recruitment to the heart following myocardial infarction (MI) is enhanced in mice lacking 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) that regenerates active glucocorticoid within cells from intrinsically inert metabolites. The present study aimed to identify the mechanism of regulation. In a mouse model of MI, neutrophil mobilization to blood and recruitment to the heart were higher in 11β-HSD1-deficient (Hsd11b1-/- ) relative to wild-type (WT) mice, despite similar initial injury and circulating glucocorticoid. In bone marrow chimeric mice, neutrophil mobilization was increased when 11β-HSD1 was absent from host cells, but not when absent from donor bone marrow-derived cells. Consistent with a role for 11β-HSD1 in 'host' myocardium, gene expression of a subset of neutrophil chemoattractants, including the chemokines Cxcl2 and Cxcl5, was selectively increased in the myocardium of Hsd11b1-/- mice relative to WT. SM22α-Cre directed disruption of Hsd11b1 in smooth muscle and cardiomyocytes had no effect on neutrophil recruitment. Expression of Cxcl2 and Cxcl5 was elevated in fibroblast fractions isolated from hearts of Hsd11b1-/- mice post MI and provision of either corticosterone or of the 11β-HSD1 substrate, 11-dehydrocorticosterone, to cultured murine cardiac fibroblasts suppressed IL-1α-induced expression of Cxcl2 and Cxcl5 These data identify suppression of CXCL2 and CXCL5 chemoattractant expression by 11β-HSD1 as a novel mechanism with potential for regulation of neutrophil recruitment to the injured myocardium, and cardiac fibroblasts as a key site for intracellular glucocorticoid regeneration during acute inflammation following myocardial injury.
Collapse
Affiliation(s)
- Katie J Mylonas
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - Neil A Turner
- Division of Cardiovascular & Diabetes ResearchLeeds Institute of Cardiovascular & Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds, UK
| | - Sumia A Bageghni
- Division of Cardiovascular & Diabetes ResearchLeeds Institute of Cardiovascular & Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds, UK
| | - Christopher J Kenyon
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - Christopher I White
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - Kieran McGregor
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - Robert A Kimmitt
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - Richard Sulston
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - Valerie Kelly
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - Brian R Walker
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - Karen E Porter
- Division of Cardiovascular & Diabetes ResearchLeeds Institute of Cardiovascular & Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds, UK
| | - Karen E Chapman
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - Gillian A Gray
- University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| |
Collapse
|
95
|
Whirledge S, Cidlowski JA. Glucocorticoids and Reproduction: Traffic Control on the Road to Reproduction. Trends Endocrinol Metab 2017; 28:399-415. [PMID: 28274682 PMCID: PMC5438761 DOI: 10.1016/j.tem.2017.02.005] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/06/2017] [Accepted: 02/12/2017] [Indexed: 02/06/2023]
Abstract
Glucocorticoids are steroid hormones that regulate diverse cellular functions and are essential to facilitate normal physiology. However, stress-induced levels of glucocorticoids result in several pathologies including profound reproductive dysfunction. Compelling new evidence indicates that glucocorticoids are crucial to the establishment and maintenance of reproductive function. The fertility-promoting or -inhibiting activity of glucocorticoids depends on timing, dose, and glucocorticoid responsiveness within a given tissue, which is mediated by the glucocorticoid receptor (GR). The GR gene and protein are subject to cellular processing, contributing to signaling diversity and providing a mechanism by which both physiological and stress-induced levels of glucocorticoids function in a cell-specific manner. Understanding how glucocorticoids regulate fertility and infertility may lead to novel approaches to the regulation of reproductive function.
Collapse
Affiliation(s)
- Shannon Whirledge
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - John A Cidlowski
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 TW Alexander Drive, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
96
|
Legeza B, Marcolongo P, Gamberucci A, Varga V, Bánhegyi G, Benedetti A, Odermatt A. Fructose, Glucocorticoids and Adipose Tissue: Implications for the Metabolic Syndrome. Nutrients 2017; 9:nu9050426. [PMID: 28445389 PMCID: PMC5452156 DOI: 10.3390/nu9050426] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/16/2017] [Accepted: 04/20/2017] [Indexed: 12/13/2022] Open
Abstract
The modern Western society lifestyle is characterized by a hyperenergetic, high sugar containing food intake. Sugar intake increased dramatically during the last few decades, due to the excessive consumption of high-sugar drinks and high-fructose corn syrup. Current evidence suggests that high fructose intake when combined with overeating and adiposity promotes adverse metabolic health effects including dyslipidemia, insulin resistance, type II diabetes, and inflammation. Similarly, elevated glucocorticoid levels, especially the enhanced generation of active glucocorticoids in the adipose tissue due to increased 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1) activity, have been associated with metabolic diseases. Moreover, recent evidence suggests that fructose stimulates the 11β-HSD1-mediated glucocorticoid activation by enhancing the availability of its cofactor NADPH. In adipocytes, fructose was found to stimulate 11β-HSD1 expression and activity, thereby promoting the adipogenic effects of glucocorticoids. This article aims to highlight the interconnections between overwhelmed fructose metabolism, intracellular glucocorticoid activation in adipose tissue, and their metabolic effects on the progression of the metabolic syndrome.
Collapse
Affiliation(s)
- Balázs Legeza
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest 1085, Hungary.
- First Department of Pediatrics, Semmelweis University, Budapest 1085, Hungary.
| | - Paola Marcolongo
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy.
| | - Alessandra Gamberucci
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy.
| | - Viola Varga
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest 1085, Hungary.
| | - Gábor Bánhegyi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest 1085, Hungary.
- Pathobiochemistry Research Group of the Hungarian Academy of Sciences and Semmelweis University, Budapest 1085, Hungary.
| | - Angiolo Benedetti
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy.
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| |
Collapse
|
97
|
Carnevali O, Notarstefano V, Olivotto I, Graziano M, Gallo P, Di Marco Pisciottano I, Vaccari L, Mandich A, Giorgini E, Maradonna F. Dietary administration of EDC mixtures: A focus on fish lipid metabolism. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2017; 185:95-104. [PMID: 28208108 DOI: 10.1016/j.aquatox.2017.02.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 01/30/2017] [Accepted: 02/05/2017] [Indexed: 05/26/2023]
Abstract
Many man-made chemical compounds are recognized as endocrine disruptors and once released into the environment are likely to spread and bioaccumulate in wild species. Due to their lipophilic nature, these substances pass through the cell membrane or bind to specific receptors activating physiological responses that in the long run can cause reproductive impairment, physiological disorders, including the occurrence of metabolic syndromes. One significant source of contamination is represented by the consumption of polluted food. As a consequence, different environmental pollutants, with similar or different modes of action, can accumulate in organisms and biomagnify along the food web, finally targeting humans. The aim of this study was to analyze, under controlled conditions, the effects induced by the consumption of contaminated diets, focusing on the effects exerted at hepatic level. Juvenile seabream were fed for 21days a diet enriched with different combinations of pollutants, nonylphenol (NP), tert-octylphenol (t-OP) and bisphenol A (BPA). The different diets containing 5mg/kg bw of each contaminant, were formulated as follows: NP+tOP, BPA+NP, BPA+tOP and NP+BPA+tOP (NBO). EDCs, at the doses administered, showed low biomagnification factor (BMF), suggesting that these pollutants hardly accumulate in muscles. The results obtained at hepatic level pinpointed the steatotic effect of all the administered diets, associated to a modulation of the expression of genes involved in lipid metabolism (ppars, fas, lpl, and hsl). Results were compared to those obtained in previous studies in which fish were fed single pollutants evidencing that the administration of mixture of contaminants exerts a milder lipogenic effect, highlighting the contrasting/antagonistic interaction establishing among chemicals. Noteworthy was the setup of a new chromatographic method to detect the presence of the selected chemical in fish muscle and the application of Fourier Transform Infrared (FT-IR) analysis to evaluate pollutant-induced changes in the liver macromolecular building.
Collapse
Affiliation(s)
- O Carnevali
- Dipartimento di Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, 60131 Ancona, Italy; INBB Consorzio Interuniversitario Istituto Nazionale Biostrutture e Biosistemi, 00136 Roma, Italy
| | - V Notarstefano
- Dipartimento di Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, 60131 Ancona, Italy
| | - I Olivotto
- Dipartimento di Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, 60131 Ancona, Italy
| | - M Graziano
- Dipartimento di Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, 60131 Ancona, Italy
| | - P Gallo
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, Università di Genova, 16132 Genova, Italy; Dipartimento di Chimica, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, NA, Italy
| | - I Di Marco Pisciottano
- Dipartimento di Chimica, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, NA, Italy
| | - L Vaccari
- SISSI Beamline, Elettra Synchrotron Light Laboratory, 34149 Basovizza, Trieste, Italy
| | - A Mandich
- INBB Consorzio Interuniversitario Istituto Nazionale Biostrutture e Biosistemi, 00136 Roma, Italy; Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, Università di Genova, 16132 Genova, Italy
| | - E Giorgini
- Dipartimento di Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, 60131 Ancona, Italy
| | - F Maradonna
- Dipartimento di Scienze della Vita e dell'Ambiente, Università Politecnica delle Marche, 60131 Ancona, Italy; INBB Consorzio Interuniversitario Istituto Nazionale Biostrutture e Biosistemi, 00136 Roma, Italy.
| |
Collapse
|
98
|
Hocher B, Adamski J. Metabolomics for clinical use and research in chronic kidney disease. Nat Rev Nephrol 2017; 13:269-284. [PMID: 28262773 DOI: 10.1038/nrneph.2017.30] [Citation(s) in RCA: 232] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Chronic kidney disease (CKD) has a high prevalence in the general population and is associated with high mortality; a need therefore exists for better biomarkers for diagnosis, monitoring of disease progression and therapy stratification. Moreover, very sensitive biomarkers are needed in drug development and clinical research to increase understanding of the efficacy and safety of potential and existing therapies. Metabolomics analyses can identify and quantify all metabolites present in a given sample, covering hundreds to thousands of metabolites. Sample preparation for metabolomics requires a very fast arrest of biochemical processes. Present key technologies for metabolomics are mass spectrometry and proton nuclear magnetic resonance spectroscopy, which require sophisticated biostatistic and bioinformatic data analyses. The use of metabolomics has been instrumental in identifying new biomarkers of CKD such as acylcarnitines, glycerolipids, dimethylarginines and metabolites of tryptophan, the citric acid cycle and the urea cycle. Biomarkers such as c-mannosyl tryptophan and pseudouridine have better performance in CKD stratification than does creatinine. Future challenges in metabolomics analyses are prospective studies and deconvolution of CKD biomarkers from those of other diseases such as metabolic syndrome, diabetes mellitus, inflammatory conditions, stress and cancer.
Collapse
Affiliation(s)
- Berthold Hocher
- Department of Basic Medicine, Medical College of Hunan University, 410006 Changsha, China
| | - Jerzy Adamski
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| |
Collapse
|
99
|
Martins CS, Elias D, Colli LM, Couri CE, Souza MCLA, Moreira AC, Foss MC, Elias LLK, de Castro M. HPA axis dysregulation, NR3C1 polymorphisms and glucocorticoid receptor isoforms imbalance in metabolic syndrome. Diabetes Metab Res Rev 2017; 33. [PMID: 27507764 DOI: 10.1002/dmrr.2842] [Citation(s) in RCA: 25] [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: 03/03/2016] [Revised: 06/19/2016] [Accepted: 08/01/2016] [Indexed: 01/08/2023]
Abstract
CONTEXT Metabolic syndrome (MetS) shares several similarities with hypercortisolism. OBJECTIVES To evaluate hypothalamic-pituitary-adrenal (HPA) axis sensitivity to dexamethasone (DEX), NR3C1 single nucleotide polymorphisms (SNPs), and expression of glucocorticoid receptor (GR) isoforms and cytokines in peripheral immune cells of MetS patients and controls. DESIGN Prospective study with 40 MetS patients and 40 controls was conducted at the Ribeirão Preto Medical School University Hospital. METHODS Plasma and salivary cortisol were measured in basal conditions and after 0.25, 0.5, and 1 mg of DEX given at 2300 h. In addition, p.N363S (rs6195), p.ER22/23EK (rs6189-6190), and BclI (rs41423247) SNPs were evaluated by quantitative polymerase chain reaction allelic discrimination. Exons 3 to 9 and exon/intron boundaries of NR3C1 were sequenced. GR isoforms and cytokines (IL1B, IL2, IL4, IL6, IL8, IL10, IFNγ, TNFα) expression were assessed by quantitative polymerase chain reaction. RESULTS Plasma and salivary cortisol (nmol/L) after 1-mg DEX were higher in MetS patients compared with controls (PF: 70.2 ± 17.3 vs 37.9 ± 2.6, P = .02, and SF: 4.9 ± 1.7 vs 2.2 ± 0.3, P < .0001). After all DEX doses, a lower number of MetS patients suppressed plasma and salivary cortisol compared with controls. The BclI genotypic frequencies (%) differed between patients (CC:56/CG:44) and controls (CC:50/CG:32.5/GG:17.5) (P = .03). The GRβ was overexpressed (fold = 100.0; P = .002) and IL4 (fold = -265.0; P < .0001) was underexpressed in MetS. CONCLUSION MetS patients exhibited decreased HPA sensitivity to glucocorticoid feedback. Moreover, the BclI polymorphism lower frequency, GRβ overexpression, and IL4 underexpression might underlie the molecular mechanism of glucocorticoid resistance in MetS. Thus, HPA axis dysregulation might contribute to MetS pathogenesis.
Collapse
Affiliation(s)
- Clarissa Silva Martins
- Department of Internal Medicine - Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Daniel Elias
- Department of Internal Medicine - Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Leandro Machado Colli
- Department of Internal Medicine - Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Carlos Eduardo Couri
- Department of Internal Medicine - Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Manoel Carlos L A Souza
- Department of Internal Medicine - Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Ayrton C Moreira
- Department of Internal Medicine - Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Milton C Foss
- Department of Internal Medicine - Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Lucila L K Elias
- Department of Physiology - Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Margaret de Castro
- Department of Internal Medicine - Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
100
|
Abstract
Glucocorticoids are primary stress hormones produced by the adrenal cortex. The concentration of serum glucocorticoids in the fetus is low throughout most of gestation but surge in the weeks prior to birth. While their most well-known function is to stimulate differentiation and functional development of the lungs, glucocorticoids also play crucial roles in the development of several other organ systems. Mothers at risk of preterm delivery are administered glucocorticoids to accelerate fetal lung development and prevent respiratory distress. Conversely, excessive glucocorticoid signaling is detrimental for fetal development; slowing fetal and placental growth and programming the individual for disease later in adult life. This review explores the mechanisms that control glucocorticoid signaling during pregnancy and provides an overview of the impact of glucocorticoid signaling on fetal development.
Collapse
Affiliation(s)
- Jonathan T Busada
- Molecular Endocrinology Group, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - John A Cidlowski
- Molecular Endocrinology Group, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States.
| |
Collapse
|