1
|
Astor MC, Løvås K, Methlie P, Simunkova K, Assmus J, Husebye ES. Corticosteroid rhythms in hypoparathyroid patients. Eur J Endocrinol 2024; 191:271-278. [PMID: 39167533 DOI: 10.1093/ejendo/lvae102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/16/2024] [Accepted: 08/19/2024] [Indexed: 08/23/2024]
Abstract
OBJECTIVE Previous studies indicate a possible bidirectional stimulatory relationship between parathyroid hormone (PTH) and adrenocortical hormones, but the pattern of adrenocortical secretion in hypoparathyroidism is unknown. We aimed to characterize the adrenocortical secretion in patients with postsurgical hypoparathyroidism, and whether continuous subcutaneous PTH (1-34) infusion alters secretion patterns. DESIGN Crossover interventional study. METHODS We recruited 10 patients with postsurgical hypoparathyroidism with very low PTH levels on stable treatment with active vitamin D and calcium. Cortisol, cortisone, and aldosterone levels were measured in microdialysate from subcutaneous tissue over 24 h, before and during continuous subcutaneous PTH (1-34) infusion. Cortisol was also assayed in serum, saliva, and urine, and aldosterone and ACTH in serum and plasma, respectively. Ten patients with primary hyperparathyroidism and 10 healthy volunteers matched for sex and age served as controls. RESULTS Hypoparathyroid patients displayed both ultradian and circadian rhythmicity for tissue cortisol, cortisone, and aldosterone. Tissue aldosterone and cortisone levels were significantly lower in hypoparathyroid patients than in healthy controls, with no difference in tissue cortisol, but a higher cortisol to cortisone ratio. Treatment with PTH (1-34) increased tissue levels of aldosterone, cortisol, and cortisone and reduced the ratio of cortisol to cortisone. CONCLUSION Adrenocortical hormone levels are reduced in postsurgical hypoparathyroidism, and partly restored by short-term continuous subcutaneous PTH (1-34) therapy. CLINICAL TRIAL REGISTRATION NUMBER NCT02986607.
Collapse
Affiliation(s)
- Marianne C Astor
- Department of Clinical Medicine and KG Jebsen-Center for Autoimmune Diseases, University of Bergen, 5020 Bergen, Norway
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Kristian Løvås
- Department of Clinical Medicine and KG Jebsen-Center for Autoimmune Diseases, University of Bergen, 5020 Bergen, Norway
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Paal Methlie
- Department of Clinical Medicine and KG Jebsen-Center for Autoimmune Diseases, University of Bergen, 5020 Bergen, Norway
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Katerina Simunkova
- Department of Clinical Medicine and KG Jebsen-Center for Autoimmune Diseases, University of Bergen, 5020 Bergen, Norway
| | - Jörg Assmus
- Centre for Clinical Research, Haukeland University Hospital, 5021 Bergen, Norway
| | - Eystein S Husebye
- Department of Clinical Medicine and KG Jebsen-Center for Autoimmune Diseases, University of Bergen, 5020 Bergen, Norway
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| |
Collapse
|
2
|
Potter O, Menon V, Mollan SP. Risk factors and disease associations in people living with idiopathic intracranial hypertension. Expert Rev Neurother 2024; 24:681-689. [PMID: 38803205 DOI: 10.1080/14737175.2024.2359420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
INTRODUCTION Idiopathic intracranial hypertension is a neurological condition characterized by a raised intracranial pressure and papilledema, leading to chronic headaches and visual disturbances. By recognizing modifiable risk factors and deploying evidence-based interventions, healthcare providers have the potential to mitigate the burden of the disease and improve patient outcomes. AREAS COVERED It is well known that the condition occurs in predominantly pre-menopausal females who live with obesity particularly in the context of recent weight gain. This review discusses what risk factors may contribute to the metabolic underpinnings of cerebrospinal fluid dysregulation. There are a number of disease associations that are important to screen for as they can alter management. EXPERT OPINION There is emerging evidence to suggest that idiopathic intracranial hypertension is a systemic metabolic disease and it is unknown what are all the risk factors are that precipitate the condition. Targeting certain hardwired risk factors is unachievable. However, as recent weight gain has been identified as a predominant risk factor for the development of the disease and relapse, modification of body weight should be the primary aim of management. Insightful research into the involvement of the neuroendocrine axis driving cerebrospinal fluid dysregulation now has the potential for the development of therapeutic targets.
Collapse
Affiliation(s)
- Olivia Potter
- Birmingham Neuro-Ophthalmology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Vaishnavi Menon
- Birmingham Neuro-Ophthalmology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Susan P Mollan
- Birmingham Neuro-Ophthalmology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Translational Brain Science, Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, UK
| |
Collapse
|
3
|
Suzuki M, Funasaka N, Sato Y, Inamori D, Watanabe Y, Ozaki M, Hosono M, Shindo H, Kawamura K, Tatsukawa T, Yoshioka M. Association of seasonal changes in circulating cortisol concentrations with the expression of cortisol biosynthetic enzymes and a glucocorticoid receptor in the blubber of common bottlenose dolphin. Gen Comp Endocrinol 2024; 352:114516. [PMID: 38593942 DOI: 10.1016/j.ygcen.2024.114516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 03/15/2024] [Accepted: 04/06/2024] [Indexed: 04/11/2024]
Abstract
Cortisol is secreted from the adrenal cortex in response to stress, and its circulating levels are used as robust physiological indicators of stress intensity in various animals. Cortisol is also produced locally in adipose tissue by the conversion of steroid hormones such as cortisone, which is related to fat accumulation. Circulating cortisol levels, probably induced by cold stress, increase in cetaceans under cold conditions. However, whether cortisol production in subcutaneous adipose tissue is enhanced when fat accumulation is renewed during the cold season remains unclear. Therefore, in this study, we examine the effect of environmental temperature on the expression of cortisol synthesis-related enzymes and a glucocorticoid receptor in the subcutaneous fat (blubber) and explore the association between these expressions and fluctuations in circulating cortisol levels in common bottlenose dolphins (Tursiops truncatus). Skin biopsies were obtained seasonally from eight female dolphins, and seasonal differences in the expression of target genes in the blubber were analyzed. Blood samples were collected throughout the year, and cortisol levels were measured. We found that the expressions of cytochrome P450 family 21 subfamily A member 2 (CYP21A2) and nuclear receptor subfamily 3 group C member 1 (NR3C1), a glucocorticoid receptor, were increased in the cold season, and 11 beta-hydroxysteroid dehydrogenase type 1 (HSD11B1) showed a similar trend. Blood cortisol levels increased when the water temperature decreased. These results suggest that the conversion of 17-hydroxyprogesterone to cortisol via 11-deoxycortisol and/or of cortisone to cortisol is enhanced under cold conditions, and the physiological effects of cortisol in subcutaneous adipose tissue may contribute to on-site lipid accumulation and increase the circulating cortisol concentrations. The results obtained in this study highlight the role of cortisol in the regulation of the blubber that has developed to adapt to aquatic life.
Collapse
Affiliation(s)
- Miwa Suzuki
- College of Bioresource Sciences, Nihon University, Fujisawa, Kanagawa 252-0880, Japan.
| | - Noriko Funasaka
- Cetacean Research Center, Graduate School of Bioresources, Mie University, Tsu, Mie 514-8507 Japan
| | - Yuki Sato
- College of Bioresource Sciences, Nihon University, Fujisawa, Kanagawa 252-0880, Japan
| | - Daiki Inamori
- Taiji Whale Museum, Higashimuro, Wakayama 649-5171 Japan
| | - Yurie Watanabe
- Taiji Whale Museum, Higashimuro, Wakayama 649-5171 Japan
| | - Miki Ozaki
- Adventure World, Nishimuro, Wakayama 649-2201 Japan
| | | | - Hideaki Shindo
- Shimonoseki Marine Science Museum, Shimonoseki, Yamaguchi 750-0036 Japan
| | - Keiko Kawamura
- Shimonoseki Marine Science Museum, Shimonoseki, Yamaguchi 750-0036 Japan
| | | | - Motoi Yoshioka
- Cetacean Research Center, Graduate School of Bioresources, Mie University, Tsu, Mie 514-8507 Japan.
| |
Collapse
|
4
|
Vignesh V, Castro-Dominguez B, James TD, Gamble-Turner JM, Lightman S, Reis NM. Advancements in Cortisol Detection: From Conventional Methods to Next-Generation Technologies for Enhanced Hormone Monitoring. ACS Sens 2024; 9:1666-1681. [PMID: 38551608 PMCID: PMC11059103 DOI: 10.1021/acssensors.3c01912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/22/2024] [Accepted: 03/08/2024] [Indexed: 05/02/2024]
Abstract
The hormone cortisol, released as the end-product of the hypothalamic-pituitary-adrenal (HPA) axis, has a well-characterized circadian rhythm that enables an allostatic response to external stressors. When the pattern of secretion is disrupted, cortisol levels are chronically elevated, contributing to diseases such as heart attacks, strokes, mental health disorders, and diabetes. The diagnosis of chronic stress and stress related disorders depends upon accurate measurement of cortisol levels; currently, it is quantified using mass spectroscopy or immunoassay, in specialized laboratories with trained personnel. However, these methods are time-consuming, expensive and are unable to capture the dynamic biorhythm of the hormone. This critical review traces the path of cortisol detection from traditional laboratory-based methods to decentralised cortisol monitoring biosensors. A complete picture of cortisol biology and pathophysiology is provided, and the importance of precision medicine style monitoring of cortisol is highlighted. Antibody-based immunoassays still dominate the pipeline of development of point-of-care biosensors; new capture molecules such as aptamers and molecularly imprinted polymers (MIPs) combined with technologies such as microfluidics, wearable electronics, and quantum dots offer improvements to limit of detection (LoD), specificity, and a shift toward rapid or continuous measurements. While a variety of different sensors and devices have been proposed, there still exists a need to produce quantitative tests for cortisol ─ using either rapid or continuous monitoring devices that can enable a personalized medicine approach to stress management. This can be addressed by synergistic combinations of technologies that can leverage low sample volumes, relevant limit of detection and rapid testing time, to better account for cortisol's shifting biorhythm. Trends in cortisol diagnostics toward rapid and continuous monitoring of hormones are highlighted, along with insights into choice of sample matrix.
Collapse
Affiliation(s)
- Visesh Vignesh
- Department
of Chemical Engineering and Centre for Bioengineering and Biomedical
Technologies (CBio) University of Bath, BA2 7AY Bath, U.K.
| | - Bernardo Castro-Dominguez
- Department
of Chemical and Engineering and Digital Manufacturing and Design University
of Bath, BA2 7AY Bath, U.K.
| | - Tony D. James
- Department
of Chemistry, University of Bath, BA2 7AY Bath, U.K.
| | | | - Stafford Lightman
- Translational
Health Sciences, Bristol Medical School, University of Bristol, BS1 3NY Bristol, U.K.
| | - Nuno M. Reis
- Department
of Chemical Engineering and Centre for Bioengineering and Biomedical
Technologies (CBio) University of Bath, BA2 7AY Bath, U.K.
| |
Collapse
|
5
|
Schiffer L, Oestlund I, Snoep JL, Gilligan LC, Taylor AE, Sinclair AJ, Singhal R, Freeman A, Ajjan R, Tiganescu A, Arlt W, Storbeck KH. Inhibition of the glucocorticoid-activating enzyme 11β-hydroxysteroid dehydrogenase type 1 drives concurrent 11-oxygenated androgen excess. FASEB J 2024; 38:e23574. [PMID: 38551804 DOI: 10.1096/fj.202302131r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/19/2024] [Accepted: 03/11/2024] [Indexed: 04/02/2024]
Abstract
Aldo-keto reductase 1C3 (AKR1C3) is a key enzyme in the activation of both classic and 11-oxygenated androgens. In adipose tissue, AKR1C3 is co-expressed with 11β-hydroxysteroid dehydrogenase type 1 (HSD11B1), which catalyzes not only the local activation of glucocorticoids but also the inactivation of 11-oxygenated androgens, and thus has the potential to counteract AKR1C3. Using a combination of in vitro assays and in silico modeling we show that HSD11B1 attenuates the biosynthesis of the potent 11-oxygenated androgen, 11-ketotestosterone (11KT), by AKR1C3. Employing ex vivo incubations of human female adipose tissue samples we show that inhibition of HSD11B1 results in the increased peripheral biosynthesis of 11KT. Moreover, circulating 11KT increased 2-3 fold in individuals with type 2 diabetes after receiving the selective oral HSD11B1 inhibitor AZD4017 for 35 days, thus confirming that HSD11B1 inhibition results in systemic increases in 11KT concentrations. Our findings show that HSD11B1 protects against excess 11KT production by adipose tissue, a finding of particular significance when considering the evidence for adverse metabolic effects of androgens in women. Therefore, when targeting glucocorticoid activation by HSD11B1 inhibitor treatment in women, the consequently increased generation of 11KT may offset beneficial effects of decreased glucocorticoid activation.
Collapse
Affiliation(s)
- Lina Schiffer
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Imken Oestlund
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Jacky L Snoep
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
- Molecular Cell Biology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Lorna C Gilligan
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Angela E Taylor
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Alexandra J Sinclair
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Rishi Singhal
- Upper GI Unit and Minimally Invasive Unit, Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Adrian Freeman
- Emerging Innovations Unit, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ramzi Ajjan
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Center, Leeds Teaching Hospitals, NHS Trust, Leeds, UK
| | - Ana Tiganescu
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Center, Leeds Teaching Hospitals, NHS Trust, Leeds, UK
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London, UK
- Medical Research Council Laboratory of Medical Sciences, London, UK
| | - Karl-Heinz Storbeck
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
6
|
Dineen RA, Martin-Grace J, Ahmed KMS, Taylor AE, Shaheen F, Schiffer L, Gilligan LC, Lavery GG, Frizelle I, Gunness A, Garrahy A, Hannon AM, Methlie P, Eystein SH, Stewart PM, Tomlinson JW, Hawley JM, Keevil BG, O’Reilly MW, Smith D, McDermott J, Healy ML, Agha A, Pazderska A, Gibney J, Behan LA, Thompson CJ, Arlt W, Sherlock M. Tissue Glucocorticoid Metabolism in Adrenal Insufficiency: A Prospective Study of Dual-release Hydrocortisone Therapy. J Clin Endocrinol Metab 2023; 108:3178-3189. [PMID: 37339332 PMCID: PMC10673701 DOI: 10.1210/clinem/dgad370] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/01/2023] [Accepted: 06/16/2023] [Indexed: 06/22/2023]
Abstract
BACKGROUND Patients with adrenal insufficiency (AI) require life-long glucocorticoid (GC) replacement therapy. Within tissues, cortisol (F) availability is under the control of the isozymes of 11β-hydroxysteroid dehydrogenase (11β-HSD). We hypothesize that corticosteroid metabolism is altered in patients with AI because of the nonphysiological pattern of current immediate release hydrocortisone (IR-HC) replacement therapy. The use of a once-daily dual-release hydrocortisone (DR-HC) preparation, (Plenadren®), offers a more physiological cortisol profile and may alter corticosteroid metabolism in vivo. STUDY DESIGN AND METHODS Prospective crossover study assessing the impact of 12 weeks of DR-HC on systemic GC metabolism (urinary steroid metabolome profiling), cortisol activation in the liver (cortisone acetate challenge test), and subcutaneous adipose tissue (microdialysis, biopsy for gene expression analysis) in 51 patients with AI (primary and secondary) in comparison to IR-HC treatment and age- and BMI-matched controls. RESULTS Patients with AI receiving IR-HC had a higher median 24-hour urinary excretion of cortisol compared with healthy controls (72.1 µg/24 hours [IQR 43.6-124.2] vs 51.9 µg/24 hours [35.5-72.3], P = .02), with lower global activity of 11β-HSD2 and higher 5-alpha reductase activity. Following the switch from IR-HC to DR-HC therapy, there was a significant reduction in urinary cortisol and total GC metabolite excretion, which was most significant in the evening. There was an increase in 11β-HSD2 activity. Hepatic 11β-HSD1 activity was not significantly altered after switching to DR-HC, but there was a significant reduction in the expression and activity of 11β-HSD1 in subcutaneous adipose tissue. CONCLUSION Using comprehensive in vivo techniques, we have demonstrated abnormalities in corticosteroid metabolism in patients with primary and secondary AI receiving IR-HC. This dysregulation of pre-receptor glucocorticoid metabolism results in enhanced glucocorticoid activation in adipose tissue, which was ameliorated by treatment with DR-HC.
Collapse
Affiliation(s)
- Rosemary A Dineen
- Academic Department of Endocrinology, Beaumont Hospital/Royal College of Surgeons in Ireland, Dublin, D09 YD60, Ireland
| | - Julie Martin-Grace
- Academic Department of Endocrinology, Beaumont Hospital/Royal College of Surgeons in Ireland, Dublin, D09 YD60, Ireland
| | | | - Angela E Taylor
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, UK
| | - Fozia Shaheen
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, UK
| | - Lina Schiffer
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, UK
| | - Lorna C Gilligan
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, UK
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, UK
| | - Isolda Frizelle
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, D24 TP66, Ireland
| | - Anjuli Gunness
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, D24 TP66, Ireland
| | - Aoife Garrahy
- Academic Department of Endocrinology, Beaumont Hospital/Royal College of Surgeons in Ireland, Dublin, D09 YD60, Ireland
| | - Anne Marie Hannon
- Academic Department of Endocrinology, Beaumont Hospital/Royal College of Surgeons in Ireland, Dublin, D09 YD60, Ireland
| | - Paal Methlie
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| | | | - Paul M Stewart
- Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, Churchill Hospital, University of Oxford, Oxford OX3 7LE, UK
| | - James M Hawley
- Department of Clinical Biochemistry, University Hospital of South Manchester, Manchester Academic Health Science Centre, The University of Manchester, Manchester M23 9LT, UK
| | - Brian G Keevil
- Department of Clinical Biochemistry, University Hospital of South Manchester, Manchester Academic Health Science Centre, The University of Manchester, Manchester M23 9LT, UK
| | - Michael W O’Reilly
- Academic Department of Endocrinology, Beaumont Hospital/Royal College of Surgeons in Ireland, Dublin, D09 YD60, Ireland
| | - Diarmuid Smith
- Academic Department of Endocrinology, Beaumont Hospital/Royal College of Surgeons in Ireland, Dublin, D09 YD60, Ireland
| | - John McDermott
- Department of Endocrinology, Connolly Hospital, Dublin, D15 X40D, Ireland
| | - Marie-Louise Healy
- Department of Endocrinology, St James Hospital, Dublin, D08 K0Y5, Ireland
| | - Amar Agha
- Academic Department of Endocrinology, Beaumont Hospital/Royal College of Surgeons in Ireland, Dublin, D09 YD60, Ireland
| | | | - James Gibney
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, D24 TP66, Ireland
| | - Lucy-Ann Behan
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, D24 TP66, Ireland
| | - Chris J Thompson
- Academic Department of Endocrinology, Beaumont Hospital/Royal College of Surgeons in Ireland, Dublin, D09 YD60, Ireland
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, UK
- Medical Research Council London, Institute of Medical Sciences, London W12 0NN, UK
| | - Mark Sherlock
- Academic Department of Endocrinology, Beaumont Hospital/Royal College of Surgeons in Ireland, Dublin, D09 YD60, Ireland
| |
Collapse
|
7
|
Khatkar P, Hubbard JC, Hill L, Sinclair AJ, Mollan SP. Experimental drugs for the treatment of idiopathic intracranial hypertension (IIH): shedding light on phase I and II trials. Expert Opin Investig Drugs 2023; 32:1123-1131. [PMID: 38006580 DOI: 10.1080/13543784.2023.2288073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/22/2023] [Indexed: 11/27/2023]
Abstract
INTRODUCTION Idiopathic intracranial hypertension is a neurological condition characterized by a raised intracranial pressure and papilledema that causes debilitating headaches. While the extent of the pathophysiology is being discovered, the condition is emerging as a systemic metabolic disease distinct to people living with obesity alone. Idiopathic intracranial hypertension is becoming more common and therefore establishing licensed therapeutics is a key priority. AREA COVERED The translation of preclinical work in idiopathic intracranial hypertension is evident by the two early phase trials evaluating 11-β-hydroxysteroid dehydrogenase inhibitor, AZD4017, and a glucagon like peptide-1 receptor agonist, Exenatide. This review summarizes these two early phase trials evaluating targeted medicines for the treatment of intracranial pressure. The modulation of these two distinct mechanisms have potential for therapeutic intervention in people living with idiopathic intracranial hypertension. EXPERT OPINION The clinical trial landscape in idiopathic intracranial hypertension is a challenge due to the rarity of the disease and the lack of agreed meaningful trial outcomes. Further preclinical work to fully understand the pathogenesis is required to enable personalized targeted drug treatment.
Collapse
Affiliation(s)
- Pavan Khatkar
- Medical school Imperial College London, UK
- Birmingham Neuro-Ophthalmology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Jess C Hubbard
- School of Biomedical Sciences, Institute of Clinical Sciences, University of Birmingham, Birmingham, UK
- Translational Brain Science, Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, UK
| | - Lisa Hill
- School of Biomedical Sciences, Institute of Clinical Sciences, University of Birmingham, Birmingham, UK
- Translational Brain Science, Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, UK
| | - Alexandra J Sinclair
- Translational Brain Science, Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
- Department of Neurology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Susan P Mollan
- Birmingham Neuro-Ophthalmology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Translational Brain Science, Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, UK
| |
Collapse
|
8
|
Kappes C, Stein R, Körner A, Merkenschlager A, Kiess W. Stress, Stress Reduction and Obesity in Childhood and Adolescence. Horm Res Paediatr 2023; 96:88-96. [PMID: 34469895 DOI: 10.1159/000519284] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/11/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Obesity in childhood and adolescence remains a great global health challenge. Stress exposure during childhood and adolescence is associated with a higher risk for obesity, yet the linkage between stress and obesity is multidimensional, and its biological and behavioral mechanisms are still not fully understood. SUMMARY In this literature review, we identified different types of stress exposure in children and adolescents, including first studied effects of the COVID-19 pandemic as a prolonged stress exposure and their association with obesity risk. We investigated studies on the connection of altered stress biology and behavioral pathways as well as intervention programs on stress reduction in children and adolescents with obesity. KEY MESSAGES There is evidence that stress exposure in childhood and adolescence promotes biological and behavioral alterations that contribute to the multifactorial pathogenesis of obesity. COVID-19 related-stress presents the most current example of a negative influence on weight development in children and adolescents. However, longitudinal studies on the linkage between environmental, behavioral, and biological factors across development are few, and results are partly equivocal. Intervention programs to reduce stress in children through mindfulness might be a promising adjunctive tool in the prevention and treatment of childhood and adolescent obesity that could further offer proof of concept of theoretically elaborated cause-and-effect relationships.
Collapse
Affiliation(s)
- Claudia Kappes
- University Hospital for Children and Adolescents, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Robert Stein
- University Hospital for Children and Adolescents, Medical Faculty, University of Leipzig, Leipzig, Germany.,Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Antje Körner
- University Hospital for Children and Adolescents, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Andreas Merkenschlager
- University Hospital for Children and Adolescents, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Wieland Kiess
- University Hospital for Children and Adolescents, Medical Faculty, University of Leipzig, Leipzig, Germany
| |
Collapse
|
9
|
Bianzano S, Nordaby M, Plum-Mörschel L, Peil B, Heise T. Safety, tolerability, pharmacodynamics and pharmacokinetics following once-daily doses of BI 187004, an inhibitor of 11 beta-hydroxysteroid dehydrogenase-1, over 28 days in patients with type 2 diabetes mellitus and overweight or obesity. Diabetes Obes Metab 2023; 25:832-843. [PMID: 36478142 PMCID: PMC10107759 DOI: 10.1111/dom.14932] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/17/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
AIMS To study the oral 11 beta-hydroxysteroid dehydrogenase-1 (11β-HSD1) inhibitor BI 187004 (NCT02150824), as monotherapy and in combination with metformin, versus placebo in patients with type 2 diabetes mellitus (T2DM) affected by overweight or obesity. MATERIALS AND METHODS This Phase II, randomized controlled trial investigated multiple rising doses of BI 187004 as monotherapy (Arm 1: 20, 80 or 240 mg) and in combination with metformin (Arm 2: 240 mg), in adults with T2DM and a body mass index of 28-40 kg/m2 . RESULTS In total, 103 patients (Arm 1: n = 62, Arm 2: n = 41) were included in this study. BI 187004 was rapidly absorbed and exposure increased approximately dose-dependently. Target engagement of 11β-HSD1 was observed with near-full inhibition of 11β-HSD1 in the liver [decreased (5α-tetrahydrocortisol + 5β-tetrahydrocortisol)/tetrahydrocortisone ratio]; hypothalamic-pituitary-adrenal axis activation was also seen (increased total urinary corticosteroids). No clinically relevant changes from baseline with BI 187004 treatment were observed for bodyweight or meal tolerance test parameters, or in most efficacy endpoints testing glucose and lipid metabolism; a significant increase was observed in weighted mean plasma glucose (p < .05 for 80 and 240 mg BI 187004) but not fasting plasma glucose. Drug-related adverse events were reported for 14 patients (22.6%) in Arm 1 and 10 patients (24.4%) in Arm 2, most frequently headache, diarrhoea, flushing and dizziness. A dose-dependent increase in heart rate was seen with BI 187004 treatment. CONCLUSIONS BI 187004 was generally well tolerated in patients with T2DM. Despite complete 11β-HSD1 inhibition, no clinically relevant effects were observed with BI 187004.
Collapse
Affiliation(s)
| | - Matias Nordaby
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | | | - Barbara Peil
- Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim, Germany
| | | |
Collapse
|
10
|
Kupczyk D, Studzińska R, Kołodziejska R, Baumgart S, Modrzejewska M, Woźniak A. 11β-Hydroxysteroid Dehydrogenase Type 1 as a Potential Treatment Target in Cardiovascular Diseases. J Clin Med 2022; 11:jcm11206190. [PMID: 36294507 PMCID: PMC9605099 DOI: 10.3390/jcm11206190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/27/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
Glucocorticoids (GCs) belong to the group of steroid hormones. Their representative in humans is cortisol. GCs are involved in most physiological processes of the body and play a significant role in important biological processes, including reproduction, growth, immune responses, metabolism, maintenance of water and electrolyte balance, functioning of the central nervous system and the cardiovascular system. The availability of cortisol to the glucocorticoid receptor is locally controlled by the enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1). Evidence of changes in intracellular GC metabolism in the pathogenesis of obesity, metabolic syndrome (MetS) and cardiovascular complications highlights the role of selective 11β-HSD1 inhibition in the pharmacotherapy of these diseases. This paper discusses the role of 11β-HSD1 in MetS and its cardiovascular complications and the importance of selective inhibition of 11β-HSD1.
Collapse
Affiliation(s)
- Daria Kupczyk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland
- Correspondence: (D.K.); (R.S.)
| | - Renata Studzińska
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Jurasza 2, 85-089 Bydgoszcz, Poland
- Correspondence: (D.K.); (R.S.)
| | - Renata Kołodziejska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland
| | - Szymon Baumgart
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Jurasza 2, 85-089 Bydgoszcz, Poland
| | - Martyna Modrzejewska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland
| |
Collapse
|
11
|
Naredo-Gonzalez G, Upreti R, Jansen MA, Semple S, Sutcliffe OB, Marshall I, Walker BR, Andrew R. Non-invasive in vivo assessment of 11β-hydroxysteroid dehydrogenase type 1 activity by 19F-Magnetic Resonance Spectroscopy. Sci Rep 2022; 12:16268. [PMID: 36175417 PMCID: PMC9523021 DOI: 10.1038/s41598-022-18740-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 08/18/2022] [Indexed: 11/24/2022] Open
Abstract
11β-Hydroxysteroid dehydrogenase type 1 (11β-HSD1) amplifies tissue glucocorticoid levels and is a pharmaceutical target in diabetes and cognitive decline. Clinical translation of inhibitors is hampered by lack of in vivo pharmacodynamic biomarkers. Our goal was to monitor substrates and products of 11β-HSD1 non-invasively in liver via 19Fluorine magnetic resonance spectroscopy (19F-MRS). Interconversion of mono/poly-fluorinated substrate/product pairs was studied in Wistar rats (male, n = 6) and healthy men (n = 3) using 7T and 3T MRI scanners, respectively. Here we show that the in vitro limit of detection, as absolute fluorine content, was 0.625 μmole in blood. Mono-fluorinated steroids, dexamethasone and 11-dehydrodexamethasone, were detected in phantoms but not in vivo in human liver following oral dosing. A non-steroidal polyfluorinated tracer, 2-(phenylsulfonyl)-1-(4-(trifluoromethyl)phenyl)ethanone and its metabolic product were detected in vivo in rat liver after oral administration of the keto-substrate, reading out reductase activity. Administration of a selective 11β-HSD1 inhibitor in vivo in rats altered total liver 19F-MRS signal. We conclude that there is insufficient sensitivity to measure mono-fluorinated tracers in vivo in man with current dosage regimens and clinical scanners. However, since reductase activity was observed in rats using poly-fluorinated tracers, this concept could be pursued for translation to man with further development.
Collapse
Affiliation(s)
- Gregorio Naredo-Gonzalez
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, Scotland, UK
| | - Rita Upreti
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, Scotland, UK
| | - Maurits A Jansen
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, Scotland, UK.,Edinburgh Imaging, Queen's Medical Research Institute, 47 Little France Crescent, University of Edinburgh, Edinburgh, EH16 4TJ, Scotland, UK
| | - Scott Semple
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, Scotland, UK.,Edinburgh Imaging, Queen's Medical Research Institute, 47 Little France Crescent, University of Edinburgh, Edinburgh, EH16 4TJ, Scotland, UK
| | - Oliver B Sutcliffe
- Department of Natural Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK
| | - Ian Marshall
- Edinburgh Imaging, Queen's Medical Research Institute, 47 Little France Crescent, University of Edinburgh, Edinburgh, EH16 4TJ, Scotland, UK.,Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, University of Edinburgh, Edinburgh, EH16 4SB, Scotland, UK
| | - Brian R Walker
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, Scotland, UK.,Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| | - Ruth Andrew
- University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, Scotland, UK.
| |
Collapse
|
12
|
Kupczyk D, Bilski R, Kozakiewicz M, Studzińska R, Kędziora-Kornatowska K, Kosmalski T, Pedrycz-Wieczorska A, Głowacka M. 11β-HSD as a New Target in Pharmacotherapy of Metabolic Diseases. Int J Mol Sci 2022; 23:ijms23168984. [PMID: 36012251 PMCID: PMC9409048 DOI: 10.3390/ijms23168984] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Glucocorticoids (GCs), which are secreted by the adrenal cortex, are important regulators in the metabolism of carbohydrates, lipids, and proteins. For the proper functioning of the body, strict control of their release is necessary, as increased GCs levels may contribute to the development of obesity, type 2 diabetes mellitus, hypertension, cardiovascular diseases, and other pathological conditions contributing to the development of metabolic syndrome. 11β-hydroxysteroid dehydrogenase type I (11β-HSD1) locally controls the availability of the active glucocorticoid, namely cortisol and corticosterone, for the glucocorticoid receptor. Therefore, the participation of 11β-HSD1 in the development of metabolic diseases makes both this enzyme and its inhibitors attractive targets in the pharmacotherapy of the above-mentioned diseases.
Collapse
Affiliation(s)
- Daria Kupczyk
- Department of Medical Biology and Biochemistry, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland
- Correspondence: (D.K.); (R.S.)
| | - Rafał Bilski
- Department of Medical Biology and Biochemistry, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland
| | - Mariusz Kozakiewicz
- Department of Geriatrics, Nicolaus Copernicus University in Toruń, L. Rydygier Collegium Medicum in Bydgoszcz, Dębowa 3, 85-626 Bydgoszcz, Poland
| | - Renata Studzińska
- Department of Organic Chemistry, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Jurasza 2, 85-089 Bydgoszcz, Poland
- Correspondence: (D.K.); (R.S.)
| | - Kornelia Kędziora-Kornatowska
- Department of Geriatrics, Nicolaus Copernicus University in Toruń, L. Rydygier Collegium Medicum in Bydgoszcz, Dębowa 3, 85-626 Bydgoszcz, Poland
| | - Tomasz Kosmalski
- Department of Organic Chemistry, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Jurasza 2, 85-089 Bydgoszcz, Poland
| | | | - Mariola Głowacka
- Faculty of Health Sciences, Mazovian State University in Płock, Plac Dąbrowskiego 2, 09-402 Płock, Poland
| |
Collapse
|
13
|
Zhang C, Xu M, He C, Zhuo J, Burns DM, Qian DQ, Lin Q, Li YL, Chen L, Shi E, Agrios C, Weng L, Sharief V, Jalluri R, Li Y, Scherle P, Diamond S, Hunter D, Covington M, Marando C, Wynn R, Katiyar K, Contel N, Vaddi K, Yeleswaram S, Hollis G, Huber R, Friedman S, Metcalf B, Yao W. Discovery of 1'-(1-phenylcyclopropane-carbonyl)-3H-spiro[isobenzofuran-1,3'-pyrrolidin]-3-one as a novel steroid mimetic scaffold for the potent and tissue-specific inhibition of 11β-HSD1 using a scaffold-hopping approach. Bioorg Med Chem Lett 2022; 69:128782. [PMID: 35537608 DOI: 10.1016/j.bmcl.2022.128782] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/19/2022] [Accepted: 05/02/2022] [Indexed: 11/15/2022]
Abstract
11β-hydroxysteroid dehydrogenase 1 (11β-HSD1) has been identified as the primary enzyme responsible for the activation of hepatic cortisone to cortisol in specific peripheral tissues resulting in the concomitant antagonism of insulin action within these tissues. Dysregulation of 11β-HSD1, particularly in adipose tissues, has been associated with metabolic syndrome and type 2 diabetes mellitus. Therefore, inhibition of 11β-HSD1 with a small nonsteroidal molecule is therapeutically desirable. Implementation of a scaffold-hopping approach revealed a three-point pharmacophore for 11β-HSD1 that was utilized to design a steroid mimetic scaffold. Reiterative optimization provided valuable insight into the bioactive conformation of our novel scaffold and led to the discovery of INCB13739. Clinical evaluation of INCB13739 confirmed for the first time that tissue-specific inhibition of 11β-HSD1 in patients with type 2 diabetes mellitus was efficacious in controlling glucose levels and reducing cardiovascular risk factors.
Collapse
Affiliation(s)
- Colin Zhang
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Meizhong Xu
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Chunhong He
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Jincong Zhuo
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - David M Burns
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Ding-Quan Qian
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Qiyan Lin
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Yun-Long Li
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Lihua Chen
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Eric Shi
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Costas Agrios
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Linkai Weng
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Vaqar Sharief
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Ravi Jalluri
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Yanlong Li
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Peggy Scherle
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Sharon Diamond
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Deborah Hunter
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Maryanne Covington
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Cindy Marando
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Richard Wynn
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Kamna Katiyar
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Nancy Contel
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Kris Vaddi
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Swamy Yeleswaram
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Gregory Hollis
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Reid Huber
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Steve Friedman
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Brian Metcalf
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | - Wenqing Yao
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA.
| |
Collapse
|
14
|
Anderson AJ, Andrew R, Homer NZM, Hughes KA, Boyle LD, Nixon M, Karpe F, Stimson RH, Walker BR. Effects of Obesity and Insulin on Tissue-Specific Recycling Between Cortisol and Cortisone in Men. J Clin Endocrinol Metab 2021; 106:e1206-e1220. [PMID: 33270115 PMCID: PMC7947841 DOI: 10.1210/clinem/dgaa896] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Indexed: 11/19/2022]
Abstract
CONTEXT 11β-Hydroxysteroid dehydrogenase 1 (11βHSD1) reduces inert cortisone into active cortisol but also catalyzes reverse dehydrogenase activity. Drivers of cortisol/cortisone equilibrium are unclear. With obesity, 11βHSD1 transcripts are more abundant in adipose, but the consequences for oxidation vs reduction remain unknown. OBJECTIVE This work aimed to determine whether 11βHSD1 equilibrium in metabolic tissues is regulated by insulin and obesity. METHODS A 2-phase, randomized, crossover, single-blinded study in a clinical research facility was conducted of 10 lean and obese healthy men. 11β-Reductase and 11β-dehydrogenase activities were measured during infusion of 9,11,12,12-[2H]4-cortisol and 1,2-[2H]2-cortisone, respectively, on 2 occasions: once during saline infusion and once during a hyperinsulinemic-euglycemic clamp. Arterialized and venous samples were obtained across forearm skeletal muscle and abdominal subcutaneous adipose. Steroids were quantified by liquid chromatography-tandem mass spectrometry and adipose tissue transcripts by quantitative polymerase chain reaction. RESULTS Neither whole-body nor tissue-specific rates of production of cortisol or cortisone differed between lean and obese men, however insulin attenuated the diurnal decrease. Whole-body 11β-HSD1 reductase activity tended to be higher in obesity (~ 10%) and was further increased by insulin. Across adipose tissue, 11β-reductase activity was detected in obese individuals only and increased in the presence of insulin (18.99 ± 9.62 vs placebo 11.68 ± 3.63 pmol/100 g/minute; P < .05). Across skeletal muscle, 11β-dehydrogenase activity was reduced by insulin in lean men only (2.55 ± 0.90 vs 4.50 ± 1.42 pmol/100 g/minute, P < .05). CONCLUSIONS Regeneration of cortisol is upregulated by insulin in adipose tissue but not skeletal muscle. In obesity, the equilibrium between 11β-reductase and 11β-dehydrogenase activities likely promotes cortisol accumulation in adipose, which may lead to adverse metabolic consequences.
Collapse
Affiliation(s)
- Anna J Anderson
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ruth Andrew
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Mass Spectrometry Core Laboratory, Edinburgh Clinical Research Facility, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Correspondence: Ruth Andrew, PhD, Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, EH16 4TJ Edinburgh, Scotland, UK.
| | - Natalie Z M Homer
- Mass Spectrometry Core Laboratory, Edinburgh Clinical Research Facility, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Katherine A Hughes
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Luke D Boyle
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Mark Nixon
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, University of Oxford, Headington, Oxford, UK
| | - Roland H Stimson
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Brian R Walker
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
15
|
Hardy RS, Botfield H, Markey K, Mitchell JL, Alimajstorovic Z, Westgate CSJ, Sagmeister M, Fairclough RJ, Ottridge RS, Yiangou A, Storbeck KHH, Taylor AE, Gilligan LC, Arlt W, Stewart PM, Tomlinson JW, Mollan SP, Lavery GG, Sinclair AJ. 11βHSD1 Inhibition with AZD4017 Improves Lipid Profiles and Lean Muscle Mass in Idiopathic Intracranial Hypertension. J Clin Endocrinol Metab 2021; 106:174-187. [PMID: 33098644 PMCID: PMC7765633 DOI: 10.1210/clinem/dgaa766] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Indexed: 02/02/2023]
Abstract
BACKGROUND The enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) determines prereceptor metabolism and activation of glucocorticoids within peripheral tissues. Its dysregulation has been implicated in a wide array of metabolic diseases, leading to the development of selective 11β-HSD1 inhibitors. We examined the impact of the reversible competitive 11β-HSD1 inhibitor, AZD4017, on the metabolic profile in an overweight female cohort with idiopathic intracranial hypertension (IIH). METHODS We conducted a UK multicenter phase II randomized, double-blind, placebo-controlled trial of 12-week treatment with AZD4017. Serum markers of glucose homeostasis, lipid metabolism, renal and hepatic function, inflammation and androgen profiles were determined and examined in relation to changes in fat and lean mass by dual-energy X-ray absorptiometry. RESULTS Patients receiving AZD4017 showed significant improvements in lipid profiles (decreased cholesterol, increased high-density lipoprotein [HDL] and cholesterol/HDL ratio), markers of hepatic function (decreased alkaline phosphatase and gamma-glutamyl transferase), and increased lean muscle mass (1.8%, P < .001). No changes in body mass index, fat mass, and markers of glucose metabolism or inflammation were observed. Patients receiving AZD4017 demonstrated increased levels of circulating androgens, positively correlated with changes in total lean muscle mass. CONCLUSIONS These beneficial metabolic changes represent a reduction in risk factors associated with raised intracranial pressure and represent further beneficial therapeutic outcomes of 11β-HSD1 inhibition by AZD4017 in this overweight IIH cohort. In particular, beneficial changes in lean muscle mass associated with AZD4017 may reflect new applications for this nature of inhibitor in the management of conditions such as sarcopenia.
Collapse
Affiliation(s)
- Rowan S Hardy
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, UK
| | - Hannah Botfield
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Keira Markey
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - James L Mitchell
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
- Department of Neurology, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Birmingham, UK
| | - Zerin Alimajstorovic
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Connar S J Westgate
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Michael Sagmeister
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Rebecca J Fairclough
- Emerging Innovations Unit, Discovery Sciences. BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ryan S Ottridge
- Birmingham Clinical Trials Unit, Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Andreas Yiangou
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
- Department of Neurology, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Birmingham, UK
| | - Karl-Heinz H Storbeck
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Biochemistry, Stellenbosch University, Stellenbosch, Matieland, South Africa
| | - Angela E Taylor
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Lorna C Gilligan
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University of Birmingham and University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology & Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, UK
| | - Susan P Mollan
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Alexandra J Sinclair
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
- Department of Neurology, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Birmingham, UK
- Correspondence and Reprint Requests: Alexandra Sinclair, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK. E-mail:
| |
Collapse
|
16
|
Grech O, Mollan SP, Wakerley BR, Alimajstorovic Z, Lavery GG, Sinclair AJ. Emerging themes in idiopathic intracranial hypertension. J Neurol 2020; 267:3776-3784. [PMID: 32700012 PMCID: PMC7674362 DOI: 10.1007/s00415-020-10090-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE Idiopathic intracranial hypertension (IIH) is a rare disorder characterised by raised intracranial pressure. The underlying pathophysiology is mostly unknown and effective treatment is an unmet clinical need in this disease. This review evaluates key emerging themes regarding disease characteristics, mechanisms contributing to raised intracranial pressure and advances in potential therapeutic targets. FINDINGS IIH is becoming more common, with the incidence rising in parallel with the global obesity epidemic. Current medical management remains centred around weight management, which is challenging. Metabolic investigations of patients have identified specific androgen profiles in cerebrospinal fluid (CSF), which suggest an endocrine dysfunction impacting CSF secretion in IIH. Glucagon-like peptide-1 (GLP-1) and 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) have been found to play a role in CSF dynamics in IIH and have formed the basis of the first clinical trials looking at new treatments. CONCLUSIONS Identification of novel molecular targets thought to underlie IIH pathology is now being translated to clinical trials.
Collapse
Affiliation(s)
- Olivia Grech
- Metabolic Neurology, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, B15 2TH UK
| | - Susan P. Mollan
- Birmingham Neuro-Ophthalmology, Ophthalmology Department, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2TH UK
| | | | - Zerin Alimajstorovic
- Metabolic Neurology, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, B15 2TH UK
| | - Gareth G. Lavery
- Metabolic Neurology, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, B15 2TH UK
| | - Alexandra J. Sinclair
- Metabolic Neurology, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, B15 2TH UK
- Department of Neurology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2TH UK
| |
Collapse
|
17
|
Wilson RE, Jaquins-Gerstl A, Chen J, Rerick M, Weber SG. Electroosmotic Perfusion-Microdialysis Probe Created by Direct Laser Writing for Quantitative Assessment of Leucine Enkephalin Hydrolysis by Insulin-Regulated Aminopeptidase in Vivo. Anal Chem 2020; 92:14558-14567. [PMID: 32961052 PMCID: PMC11027065 DOI: 10.1021/acs.analchem.0c02799] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
There are many processes that actively alter the concentrations of solutes in the extracellular space. Enzymatic reactions, either by soluble enzymes or membrane-bound ectoenzymes, and uptake or clearance are two such processes. Investigations of ectoenzymatic reactions in vivo is challenging, particularly in the brain. Studies using microdialysis have revealed some qualitative information about what enzymes may be present, but microdialysis is a sampling technique so it is not designed to control conditions such as a substrate concentration outside the probe. Micropush-pull perfusion has been used to determine which nitric oxide synthase enzymes are active in discrete regions of the rat retina. Ectopeptidases are a particularly important class of ectoenzymes. As far as it is known, the extracellular activity of active peptides in the brain is controlled by ectopeptidases. To understand ectopeptidase activity, we developed a physical probe and an accompanying method. The probe has a two-channel source that supplies substrate or substrate plus inhibitor using electroosmotic perfusion (EOP). It also has a microdialysis probe to collect products and unreacted substrate. The method provides quantitative estimates of substrate-to-product conversion and the influence of inhibitors on this process. The quantitative estimates are made possible by including a d-amino acid-containing peptide analog of the substrate in the substrate-containing solution infused. Quantitative analysis of substrate, substrate analog, and products is carried out by quantitative, online capillary liquid chromatography-tandem mass spectrometry. The electroosmotic perfusion-microdialysis probe and associated method were used to determine the effect of the selective inhibitor HFI-419 on insulin-regulated aminopeptidase (EC 3.4.11.3) in the rat neocortex.
Collapse
Affiliation(s)
- Rachael E Wilson
- Department of Chemistry University of Pittsburgh Pittsburgh Pennsylvania 15260, United States
| | - Andrea Jaquins-Gerstl
- Department of Chemistry University of Pittsburgh Pittsburgh Pennsylvania 15260, United States
| | - Jun Chen
- Department of Electrical and Computer Engineering, and Petersen Institute of NanoScience and Engineering University of Pittsburgh Pittsburgh Pennsylvania 15260, United States
| | - Michael Rerick
- Department of Chemistry University of Pittsburgh Pittsburgh Pennsylvania 15260, United States
| | - Stephen G Weber
- Department of Chemistry University of Pittsburgh Pittsburgh Pennsylvania 15260, United States
| |
Collapse
|
18
|
Gregory S, Hill D, Grey B, Ketelbey W, Miller T, Muniz-Terrera G, Ritchie CW. 11β-hydroxysteroid dehydrogenase type 1 inhibitor use in human disease-a systematic review and narrative synthesis. Metabolism 2020; 108:154246. [PMID: 32333937 DOI: 10.1016/j.metabol.2020.154246] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/01/2020] [Accepted: 04/20/2020] [Indexed: 11/20/2022]
Abstract
INTRODUCTION 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) is an intracellular enzyme that catalyses conversion of cortisone into cortisol; correspondingly, 11β-HSD1 inhibitors inhibit this conversion. This systematic review focuses on the use of 11β-HSD1 inhibitors in diseases known to be associated with abnormalities in hypothalamic pituitary adrenal (HPA) axis function. METHODS The databases screened for suitable papers were: MedLine, EMBASE, Web of Science, ClinicalTrials.gov, and Cochrane Central. RESULTS 1925 papers were identified, of which 29 were included in the final narrative synthesis. 11β-HSD1 and its inhibitors have been studied in diabetes, obesity, metabolic syndrome (MetS), and Alzheimer's disease (AD). Higher expression of 11β-HSD1 is seen in obesity and MetS, but has not yet been described in obesity or AD. Genetic studies identify 11β-HSD1 SNPs of interest in populations with diabetes, MetS, and AD. One phase II trial successfully reduced HbA1c in a diabetic population, however trials in MetS, obesity, and AD have not met primary endpoints. CONCLUSIONS Translation of this research from preclinical studies has proved challenging so far, however this is a growing area of research and more studies should focus on understanding the complex relationships between 11β-HSD1 and disease pathology, especially given the therapeutic potential of 11β-HSD1 inhibitors in development.
Collapse
Affiliation(s)
- Sarah Gregory
- Centre for Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.
| | - David Hill
- Centre for Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Ben Grey
- Centre for Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | | | | | - Graciela Muniz-Terrera
- Centre for Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Craig W Ritchie
- Centre for Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
19
|
Metabolic Concepts in Idiopathic Intracranial Hypertension and Their Potential for Therapeutic Intervention. J Neuroophthalmol 2020; 38:522-530. [PMID: 29985799 PMCID: PMC6215484 DOI: 10.1097/wno.0000000000000684] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Traditional risk factors associated with idiopathic intracranial hypertension (IIH) include obesity, weight gain, and female sex. The incidence of IIH is increasing and yet the underlying trigger and the fueling pathological mechanisms are still poorly understood. EVIDENCE ACQUISITION Review of ophthalmology, neurology, general surgery, obesity, endocrinology, nutrition, and neurosurgery literature was made. RESULTS The facts that implicate sex and obesity in IIH and headache are examined. The role of fat distribution in IIH is questioned, and the concept of adipose tissue functioning as an endocrine organ driving IIH is discussed. The impact of androgen metabolism in IIH is reviewed as is the emerging role of glucagon-like-peptide-1 analogues in modulating intracranial pressure. This introduces the concept of developing targeted disease-modifying therapeutic strategies for IIH. CONCLUSIONS This review will discuss the possible role of the adipose/gut/brain metabolism axis in IIH and speculate how this may impact the pathogenesis of IIH and therapeutic opportunities.
Collapse
|
20
|
Markey K, Mitchell J, Botfield H, Ottridge RS, Matthews T, Krishnan A, Woolley R, Westgate C, Yiangou A, Alimajstorovic Z, Shah P, Rick C, Ives N, Taylor AE, Gilligan LC, Jenkinson C, Arlt W, Scotton W, Fairclough RJ, Singhal R, Stewart PM, Tomlinson JW, Lavery GG, Mollan SP, Sinclair AJ. 11β-Hydroxysteroid dehydrogenase type 1 inhibition in idiopathic intracranial hypertension: a double-blind randomized controlled trial. Brain Commun 2020; 2:fcz050. [PMID: 32954315 PMCID: PMC7425517 DOI: 10.1093/braincomms/fcz050] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 10/05/2019] [Accepted: 10/25/2019] [Indexed: 11/13/2022] Open
Abstract
Treatment options for idiopathic intracranial hypertension are limited. The enzyme 11β-hydroxysteroid dehydrogenase type 1 has been implicated in regulating cerebrospinal fluid secretion, and its activity is associated with alterations in intracranial pressure in idiopathic intracranial hypertension. We assessed therapeutic efficacy, safety and tolerability and investigated indicators of in vivo efficacy of the 11β-hydroxysteroid dehydrogenase type 1 inhibitor AZD4017 compared with placebo in idiopathic intracranial hypertension. A multicenter, UK, 16-week phase II randomized, double-blind, placebo-controlled trial of 12-week treatment with AZD4017 or placebo was conducted. Women aged 18–55 years with active idiopathic intracranial hypertension (>25 cmH2O lumbar puncture opening pressure and active papilledema) were included. Participants received 400 mg of oral AZD4017 twice daily compared with matching placebo over 12 weeks. The outcome measures were initial efficacy, safety and tolerability. The primary clinical outcome was lumbar puncture opening pressure at 12 weeks analysed by intention-to-treat. Secondary clinical outcomes were symptoms, visual function, papilledema, headache and anthropometric measures. In vivo efficacy was evaluated in the central nervous system and systemically. A total of 31 subjects [mean age 31.2 (SD = 6.9) years and body mass index 39.2 (SD = 12.6) kg/m2] were randomized to AZD4017 (n = 17) or placebo (n = 14). At 12 weeks, lumbar puncture pressure was lower in the AZD4017 group (29.7 cmH2O) compared with placebo (31.3 cmH2O), but the difference between groups was not statistically significant (mean difference: −2.8, 95% confidence interval: −7.1 to 1.5; P = 0.2). An exploratory analysis assessing mean change in lumbar puncture pressure within each group found a significant decrease in the AZD4017 group [mean change: −4.3 cmH2O (SD = 5.7); P = 0.009] but not in the placebo group [mean change: −0.3 cmH2O (SD = 5.9); P = 0.8]. AZD4017 was safe, with no withdrawals related to adverse effects. Nine transient drug-related adverse events were reported. One serious adverse event occurred in the placebo group (deterioration requiring shunt surgery). In vivo biomarkers of 11β-hydroxysteroid dehydrogenase type 1 activity (urinary glucocorticoid metabolites, hepatic prednisolone generation, serum and cerebrospinal fluid cortisol:cortisone ratios) demonstrated significant enzyme inhibition with the reduction in serum cortisol:cortisone ratio correlating significantly with reduction in lumbar puncture pressure (P = 0.005, R = 0.70). This is the first phase II randomized controlled trial in idiopathic intracranial hypertension evaluating a novel therapeutic target. AZD4017 was safe and well tolerated and inhibited 11β-hydroxysteroid dehydrogenase type 1 activity in vivo. Reduction in serum cortisol:cortisone correlated with decreased intracranial pressure. Possible clinical benefits were noted in this small cohort. A longer, larger study would now be of interest.
Collapse
Affiliation(s)
- Keira Markey
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - James Mitchell
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK.,Department of Neurology, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Birmingham B15 2WB, UK
| | - Hannah Botfield
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Ryan S Ottridge
- Birmingham Clinical Trials Unit, Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Tim Matthews
- Birmingham Neuro-Ophthalmology, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Birmingham B15 2WB, UK
| | - Anita Krishnan
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool L9 7LJ, UK
| | - Rebecca Woolley
- Birmingham Clinical Trials Unit, Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Connar Westgate
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
| | - Andreas Yiangou
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK.,Department of Neurology, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Birmingham B15 2WB, UK
| | - Zerin Alimajstorovic
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
| | - Pushkar Shah
- Institute of Neurological Sciences, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde, Glasgow G51 4TF, UK
| | - Caroline Rick
- Nottingham Clinical Trials Unit, Queens Medical Centre, Nottingham NG7 2UH, UK
| | - Natalie Ives
- Birmingham Clinical Trials Unit, Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Angela E Taylor
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
| | - Lorna C Gilligan
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
| | - Carl Jenkinson
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
| | - William Scotton
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK.,Department of Neurology, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Birmingham B15 2WB, UK
| | - Rebecca J Fairclough
- Emerging Innovations Unit, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0SL, UK
| | - Rishi Singhal
- Upper GI Unit and Minimally Invasive Unit, Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B9 5SS, UK
| | | | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology & Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Headington, Oxford OX3 7LJ, UK
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
| | - Susan P Mollan
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.,Birmingham Neuro-Ophthalmology, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Birmingham B15 2WB, UK
| | - Alexandra J Sinclair
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK.,Department of Neurology, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Birmingham B15 2WB, UK
| |
Collapse
|
21
|
Abulizi A, Camporez JP, Zhang D, Samuel VT, Shulman GI, Vatner DF. Ectopic lipid deposition mediates insulin resistance in adipose specific 11β-hydroxysteroid dehydrogenase type 1 transgenic mice. Metabolism 2019; 93:1-9. [PMID: 30576689 PMCID: PMC6401251 DOI: 10.1016/j.metabol.2018.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 11/28/2018] [Accepted: 12/14/2018] [Indexed: 12/17/2022]
Abstract
CONTEXT Excessive adipose glucocorticoid action is associated with insulin resistance, but the mechanisms linking adipose glucocorticoid action to insulin resistance are still debated. We hypothesized that insulin resistance from excess glucocorticoid action may be attributed in part to increased ectopic lipid deposition in liver. METHODS We tested this hypothesis in the adipose specific 11β-hydroxysteroid dehydrogenase-1 (HSD11B1) transgenic mouse, an established model of adipose glucocorticoid excess. Tissue specific insulin action was assessed by hyperinsulinemic-euglycemic clamps, hepatic lipid content was measured, hepatic insulin signaling was assessed by immunoblotting. The role of hepatic lipid content was further probed by administration of the functionally liver-targeted mitochondrial uncoupler, Controlled Release Mitochondrial Protonophore (CRMP). FINDINGS High fat diet fed HSD11B1 transgenic mice developed more severe hepatic insulin resistance than littermate controls (endogenous suppression of hepatic glucose production was reduced by 3.8-fold, P < 0.05); this was reflected by decreased insulin-stimulated hepatic insulin receptor kinase tyrosine phosphorylation and AKT serine phosphorylation. Hepatic insulin resistance was associated with a 53% increase (P < 0.05) in hepatic triglyceride content, a 73% increase in diacylglycerol content (P < 0.01), and a 66% increase in PKCε translocation (P < 0.05). Hepatic insulin resistance was prevented with administration of CRMP by reversal of hepatic steatosis and prevention of hepatic diacylglycerol accumulation and PKCε activation. CONCLUSIONS These findings are consistent with excess adipose glucocorticoid activity being a predisposing factor for the development of lipid (diacylglycerol-PKCε)-induced hepatic insulin resistance.
Collapse
Affiliation(s)
- Abudukadier Abulizi
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - João-Paulo Camporez
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Dongyan Zhang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Varman T Samuel
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Veterans Affairs Medical Center, West Haven, CT 06516, USA.
| | - Gerald I Shulman
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Daniel F Vatner
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
22
|
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: 10] [Impact Index Per Article: 1.7] [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
|
23
|
Studzińska R, Kupczyk D, Płazińska A, Kołodziejska R, Kosmalski T, Modzelewska-Banachiewicz B. Thiazolo[3,2-a]pyrimidin-5-one derivatives as a novel class of 11β-hydroxysteroid dehydrogenase inhibitors. Bioorg Chem 2018; 81:21-26. [PMID: 30086417 DOI: 10.1016/j.bioorg.2018.07.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/26/2018] [Accepted: 07/30/2018] [Indexed: 10/28/2022]
Abstract
11β-hydroxysteroid type 1 dehydrogenase (11β-HSD1) is an enzyme that increases tissue concentrations of cortisol. Selective inhibitors of this enzyme regulate the level of cortisol and thus play a key role in the treatment of Cushing's syndrome, metabolic syndrome and type 2 diabetes. In this study the inhibitory activity of 29 thiazolo[3,2-a]pyrimidin-5-one derivatives on 11β-HSD1 were investigated. Studies were carried out with pooled human liver microsomes. A lot of analyzed compounds show activity for inhibiting 11β-HSD1 (up to 59.15% at concentration 10 µmol/l). Molecular docking simulation show that the molecule of the most active compound: 7-(cyclohexylmethyl)-2-iodomethyl-2,3-dihydrothiazolo[3,2-a]pyrimidin-5-one forms hydrogen bonds with Ala172, Leu171, Leu215 or Tyr177. In addition, the cycloalkane moiety can create the hydrophobic contacts with NADP+. For this compound also the most favourable Docking Score value was obtained. The most active compound only in the slight degree inhibits 11β-HSD2 activity and is a selective inhibitor of 11β-hydroxysteroid dehydrogenase type 1. Consequently it can have a real effect on the regulation of the cortisol level in the body.
Collapse
Affiliation(s)
- Renata Studzińska
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Jurasza 2, 85-089 Bydgoszcz, Poland.
| | - Daria Kupczyk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland.
| | - Anita Płazińska
- Department of Biopharmacy, Faculty of Pharmacy, Medical University of Lublin, W. Chodzki Str. 4a, 20-093 Lublin, Poland.
| | - Renata Kołodziejska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland.
| | - Tomasz Kosmalski
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Jurasza 2, 85-089 Bydgoszcz, Poland.
| | - Bożena Modzelewska-Banachiewicz
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Jurasza 2, 85-089 Bydgoszcz, Poland.
| |
Collapse
|
24
|
Ou Y, Wilson RE, Weber SG. Methods of Measuring Enzyme Activity Ex Vivo and In Vivo. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2018; 11:509-533. [PMID: 29505726 PMCID: PMC6147230 DOI: 10.1146/annurev-anchem-061417-125619] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Enzymes catalyze a variety of biochemical reactions in the body and, in conjunction with transporters and receptors, control virtually all physiological processes. There is great value in measuring enzyme activity ex vivo and in vivo. Spatial and temporal differences or changes in enzyme activity can be related to a variety of natural and pathological processes. Several analytical approaches have been developed to meet this need. They can be classified broadly as methods either based on artificial substrates, with the goal of creating images of diseased tissue, or based on natural substrates, with the goal of understanding natural processes. This review covers a selection of these methods, including optical, magnetic resonance, mass spectrometry, and physical sampling approaches, with a focus on creative chemistry and method development that make ex vivo and in vivo measurements of enzyme activity possible.
Collapse
Affiliation(s)
| | - Rachael E Wilson
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA;
| | - Stephen G Weber
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA;
| |
Collapse
|
25
|
Stirrat LI, Walker JJ, Stryjakowska K, Jones N, Homer NZM, Andrew R, Norman JE, Lightman SL, Reynolds RM. Pulsatility of glucocorticoid hormones in pregnancy: Changes with gestation and obesity. Clin Endocrinol (Oxf) 2018; 88:592-600. [PMID: 29314170 PMCID: PMC5887976 DOI: 10.1111/cen.13548] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/14/2017] [Accepted: 12/28/2017] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Hypothalamic-pituitary-adrenal axis (HPA) activity is decreased in obese pregnancy and associates with increased foetal size. Pulsatile release of glucocorticoid hormones regulates their action in target tissues. Glucocorticoids are essential for normal foetal growth, but little is known about glucocorticoid pulsatility in pregnancy. We aimed to investigate the ultradian rhythm of glucocorticoid secretion during obese and lean pregnancy and nonpregnancy. DESIGN Serum cortisol, cortisone, corticosterone and 11-dehydrocorticosterone were measured by LC-MS/MS from samples obtained at 10-minute intervals between 08.00-11.00 hours and 16.00-19.00 hours, from 8 lean (BMI <25 kg/m2 ) and 7 obese (BMI > 35 kg/m2 ) pregnant women between 16-24 weeks gestation and again at 30-36 weeks), and nonpregnant controls (lean n = 3, obese n = 4) during the luteal phase of their menstrual cycle. Interstitial fluid cortisol was measured by ELISA, from samples obtained using a portable microdialysis and automated collection device at 20-minute intervals over 24 hours. RESULTS Serum cortisol AUC, highest peak and lowest trough increased significantly with gestation in lean and obese pregnant compared with nonpregnant subjects. Pulsatility of cortisol was detected in interstitial fluid. In pregnant subjects, interstitial fluid pulse frequency was significantly lower with advancing gestation in obese, but not in lean. CONCLUSIONS We demonstrate cortisol pulsatility in interstitial fluid. Pulse frequency is altered with increased gestation and BMI. This may be a novel mechanism to explain decreased HPA activity in obese pregnancy.
Collapse
Affiliation(s)
- Laura I. Stirrat
- Tommy's Centre for Maternal and Fetal HealthMedical Research Council Centre for Reproductive HealthUniversity of EdinburghEdinburghUK
| | - Jamie J. Walker
- Henry Wellcome Laboratories for Integrative Neuroscience and EndocrinologyUniversity of BristolBristolUK
- Wellcome Trust Centre for Biomedical Modelling and AnalysisUniversity of ExeterExeterUK
- EPSRC Centre for Predictive Modelling in HealthcareUniversity of ExeterExeterUK
- College of Engineering, Mathematics and Physical SciencesUniversity of ExeterExeterUK
| | - Ksenia Stryjakowska
- Tommy's Centre for Maternal and Fetal HealthMedical Research Council Centre for Reproductive HealthUniversity of EdinburghEdinburghUK
| | - Natalie Jones
- University/BHF Centre for Cardiovascular ScienceUniversity of EdinburghEdinburghUK
| | - Natalie Z. M. Homer
- Mass Spectrometry CoreEdinburgh Clinical Research FacilityUniversity of EdinburghEdinburghUK
| | - Ruth Andrew
- University/BHF Centre for Cardiovascular ScienceUniversity of EdinburghEdinburghUK
| | - Jane E. Norman
- Tommy's Centre for Maternal and Fetal HealthMedical Research Council Centre for Reproductive HealthUniversity of EdinburghEdinburghUK
| | - Stafford L. Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience and EndocrinologyUniversity of BristolBristolUK
| | - Rebecca M. Reynolds
- Tommy's Centre for Maternal and Fetal HealthMedical Research Council Centre for Reproductive HealthUniversity of EdinburghEdinburghUK
- University/BHF Centre for Cardiovascular ScienceUniversity of EdinburghEdinburghUK
| |
Collapse
|
26
|
Herhaus B, Petrowski K. Cortisol Stress Reactivity to the Trier Social Stress Test in Obese Adults. Obes Facts 2018; 11:491-500. [PMID: 30537716 PMCID: PMC6341320 DOI: 10.1159/000493533] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/05/2018] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Approximately 600 million adults worldwide suffer from obesity. In addition to individual's eating behavior and lack of physical activity in the development of obesity and overweight, psychosocial stress as well as hormonal stress reactivity must also be considered as important contributing factors. In the current study we compared the cortisol stress response pathway in a psychosocial stress induction (Trier Social Stress Test; TSST) with obese individuals and normal-weight controls. METHOD 32 obese individuals (17 females; mean age = 33.94 years, SD = 11.31 years) and 32 normal-weight controls (17 females; mean age = 29.09 years, SD = 10.46 years) underwent the TSST. The salivary cortisol responses and three appraisal questionnaires (Primary Appraisal Secondary Appraisal, Visual Analogue Scale, Trier Inventory for Chronic Stress) were measured. RESULTS After stress induction, there was a significant main group difference between the obese individuals and the normal-weight controls for cortisol, with lower baseline and post-stress cortisol levels in the obese individuals. Nevertheless, the obese individuals as well as the normal-weight controls showed no significant difference in the self-reported assessment of the stress condition but some significant differences in the cognitive appraisal of the TSST. CONCLUSION In conclusion, the induction of psychosocial stress showed differences in the cortisol patterns between the obese individuals and the normal-weight controls. Furthermore, the present data suggest that obesity leads to lower cortisol activity, which may indicate alterations in the Hypothalamic-pituitary-adrencortical (HPA) axis.
Collapse
Affiliation(s)
- Benedict Herhaus
- Medical Psychology and Medical Sociology, Clinic and Policlinic for Psychosomatic Medicine and Psychotherapy, University Medicine Mainz, Mainz, Germany,
| | - Katja Petrowski
- Medical Psychology and Medical Sociology, Clinic and Policlinic for Psychosomatic Medicine and Psychotherapy, University Medicine Mainz, Mainz, Germany
| |
Collapse
|
27
|
Geiker NRW, Astrup A, Hjorth MF, Sjödin A, Pijls L, Markus CR. Does stress influence sleep patterns, food intake, weight gain, abdominal obesity and weight loss interventions and vice versa? Obes Rev 2018; 19:81-97. [PMID: 28849612 DOI: 10.1111/obr.12603] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 07/06/2017] [Accepted: 07/22/2017] [Indexed: 12/29/2022]
Abstract
Decades of research have reported only weak associations between the intakes of specific foods or drinks and weight gain and obesity. Randomized controlled dietary intervention trials have only shown very modest effects of changes in nutrient intake and diet composition on body weight in obese subjects. This review summarizes the scientific evidence on the role mental stress (either in or not in association with impaired sleep) may play in poor sleep, enhanced appetite, cravings and decreased motivation for physical activity. All these factors contribute to weight gain and obesity, possibly via decreasing the efficacy of weight loss interventions. We also review evidence for the role that lifestyle and stress management may play in achieving weight loss in stress-vulnerable individuals with overweight.
Collapse
Affiliation(s)
- N R W Geiker
- Clinical Nutrition Research Unit, Copenhagen University Hospital Herlev and Gentofte, Hellerup, Denmark
| | - A Astrup
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - M F Hjorth
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - A Sjödin
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - L Pijls
- Loekintofood-gcv/scs, Brussels, Belgium
| | - C Rob Markus
- Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
28
|
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
|
29
|
Loerz C, Maser E. The cortisol-activating enzyme 11β-hydroxysteroid dehydrogenase type 1 in skeletal muscle in the pathogenesis of the metabolic syndrome. J Steroid Biochem Mol Biol 2017; 174:65-71. [PMID: 28765040 DOI: 10.1016/j.jsbmb.2017.07.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/24/2017] [Accepted: 07/25/2017] [Indexed: 12/25/2022]
Abstract
The enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) contributes to intracellular glucocorticoid action by converting inactive cortisone to its receptor-active form cortisol (11-dehydrocorticosterone and corticosterone in mice and rats). The potential role of 11β-HSD1 in the pathogenesis of the metabolic syndrome has emerged over the past three decades. However, the precise impact of 11β-HSD1 in obesity-related diseases remains uncertain. Many studies from animal experiments to clinical studies have investigated liver and adipose tissue 11β-HSD1 in relation to obesity and its metabolic disorders including insulin resistance. But the relevance of 11β-HSD1 in skeletal muscle has been less extensively studied. On the other hand, skeletal muscle is assumed to be the main site of peripheral insulin resistance, but the biological relevance of 11β-HSD1 in skeletal muscle is unclear. This mini-review will focus on 11β-HSD1 in skeletal muscle and its postulated link to obesity and insulin-resistance.
Collapse
Affiliation(s)
- Christine Loerz
- Institute of Toxicology and Pharmacology for Natural Scientists, University Medical School Schleswig-Holstein, Kiel, Germany
| | - Edmund Maser
- Institute of Toxicology and Pharmacology for Natural Scientists, University Medical School Schleswig-Holstein, Kiel, Germany.
| |
Collapse
|
30
|
Li X, Wang J, Yang Q, Shao S. 11β-Hydroxysteroid Dehydrogenase Type 1 in Obese Subjects With Type 2 Diabetes Mellitus. Am J Med Sci 2017; 354:408-414. [PMID: 29078846 DOI: 10.1016/j.amjms.2017.03.023] [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] [Received: 08/13/2016] [Revised: 03/13/2017] [Accepted: 03/14/2017] [Indexed: 01/03/2023]
Abstract
Obesity is one of the most significant contributors to the development of type 2 diabetes mellitus. Tissue-specific glucocorticoids regulated by 11β-hydroxysteroid dehydrogenase enzyme (11β-HSD) type 1 are involved in central obesity and obesity-related comorbidities. Moderate downregulation of 11β-HSD1 can attenuate insulin insensitivity and the impairment of glucose-stimulated insulin secretion. Some of the beneficial effects of 11β-HSD1 inhibition may be mediated, at least in part, through inactivation of tissue-specific glucocorticoid action related to insulin signaling mechanisms, alleviation of abnormal cytokine profile and the improvement of β-cell function. Thus, 11β-HSD1 is a promising target for the treatment and prevention of type 2 diabetes mellitus with obesity.
Collapse
Affiliation(s)
- Xia Li
- Division of Endocrinology, The First People׳s Hospital of Yichang, Three Gorges University People׳s Hospital, Yichang, P.R. China
| | - Jingli Wang
- Division of Endocrinology, Jingzhou Central Hospital, Jingzhou, P.R. China
| | - Qin Yang
- Division of Pathology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, P.R. China.
| | - Shiying Shao
- Division of Endocrinology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, P.R. China.
| |
Collapse
|
31
|
Yehuda S, Rabinovitz S. The Role of Essential Fatty Acids in Anorexia Nervosa and Obesity. Crit Rev Food Sci Nutr 2017; 56:2021-35. [PMID: 26068122 DOI: 10.1080/10408398.2013.809690] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The two basic questions in food intake study are what we eat, and how much do we eat. Most research is directed toward the control of how much is eaten. This is likely the result of the increased number of individuals with eating disorders in the Western world. Feeding behavior is highly complex, and is controlled by many psychological, physiological, biochemical, and immunological factors. The aim of this review is to clarify the involvement of fatty acids in eating disorders such as anorexia and binge eating disorder. The review will describe the modified fatty acid profile observed in individuals with anorexia or binge eating disorder, and discuss on what factors fatty acids can exert beneficial effects. In addition, the differences and similarities between anorexia and binge eating disorder will be discussed. We suggest that beneficial effects of essential fatty acids on both anorexia and binge eating disorder can be explained by the stabilizing effect of those fatty acids on the neuronal membrane fluidity index.
Collapse
Affiliation(s)
- Shlomo Yehuda
- a Psychopharmacology Lab , Department of Psychology, Bar Ilan University , Ramat Gan , Israel
| | - Sharon Rabinovitz
- a Psychopharmacology Lab , Department of Psychology, Bar Ilan University , Ramat Gan , Israel.,b School of Criminology, University of Haifa , Mount Carmel , Israel
| |
Collapse
|
32
|
Mass spectrometry and its evolving role in assessing tissue specific steroid metabolism. Biochem Soc Trans 2016; 44:645-51. [PMID: 27068983 DOI: 10.1042/bst20150234] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Indexed: 12/21/2022]
Abstract
Glucocorticoid hormones play vital roles in regulating diverse biological processes in health and disease. Tissue levels are regulated by enzymes which activate and inactivate hormones. The enzyme, 11β-hydroxysteroid dehydrogenase type 1 (11βHSD1), in particular, has been identified as a potential drug target; inhibiting this enzyme attenuates glucocorticoid action by lowering local levels of active hormone. A variety of mass spectrometric approaches have been developed to characterize this enzymein vivo Endogenous glucocorticoids and their metabolites can be profiled in urine by GC-MS and circulating steroids are now more commonly quantified by liquid chromatography tandem mass spectrometry. Tracer dilution studies have allowed rates of generation of glucocorticoids by the enzyme to be distinguished from hormone generated directly by the adrenal glands and, in combination with arterio-venous (AV) sampling, rates of production by specific tissues have been quantified. This has allowed the contribution of liver, adipose, muscle and brain to cortisol production in metabolic disease and hence prioritized drug targets. Most recently MS imaging in combination with on-tissue derivatization has been developed to profile oxo-steroids in tissue sections, allowing molecular maps to be generated across complex tissues, where regional functions are important. The review provides a synopsis of how measurement of steroids by MS has evolved with technological advances and this has provided insight into the dynamic turnover of glucocorticoidsin vivo, highlighting the milestones that have advanced the field and identifying the remaining challenges for researchers, in terms of analytical chemistry and endocrine physiology and biochemistry.
Collapse
|
33
|
Anderson AJ, Andrew R, Homer NZ, Jones GC, Smith K, Livingstone DE, Walker BR, Stimson RH. Metformin Increases Cortisol Regeneration by 11βHSD1 in Obese Men With and Without Type 2 Diabetes Mellitus. J Clin Endocrinol Metab 2016; 101:3787-3793. [PMID: 27459533 PMCID: PMC5052341 DOI: 10.1210/jc.2016-2069] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
CONTEXT The mechanism of action of metformin remains unclear. Given the regulation of the cortisol-regenerating enzyme 11βhydroxysteroid dehydrogenase 1 (11βHSD1) by insulin and the limited efficacy of selective 11βHSD1 inhibitors to lower blood glucose when co-prescribed with metformin, we hypothesized that metformin reduces 11βHSD1 activity. OBJECTIVE To determine whether metformin regulates 11βHSD1 activity in vivo in obese men with and without type 2 diabetes mellitus. DESIGN Double-blind, randomized, placebo-controlled, crossover study. SETTING A hospital clinical research facility. PARTICIPANTS Eight obese nondiabetic (OND) men and eight obese men with type 2 diabetes (ODM). INTERVENTION Participants received 28 days of metformin (1 g twice daily), placebo, or (in the ODM group) gliclazide (80 mg twice daily) in random order. A deuterated cortisol infusion at the end of each phase measured cortisol regeneration by 11βHSD1. Oral cortisone was given to measure hepatic 11βHSD1 activity in the ODM group. The effect of metformin on 11βHSD1 was also assessed in human hepatocytes and Simpson-Golabi-Behmel syndrome adipocytes. MAIN OUTCOME MEASURES The effect of metformin on whole-body and hepatic 11βHSD1 activity. RESULTS Whole-body 11βHSD1 activity was approximately 25% higher in the ODM group than the OND group. Metformin increased whole-body cortisol regeneration by 11βHSD1 in both groups compared with placebo and gliclazide and tended to increase hepatic 11βHSD1 activity. In vitro, metformin did not increase 11βHSD1 activity in hepatocytes or adipocytes. CONCLUSIONS Metformin increases whole-body cortisol generation by 11βHSD1 probably through an indirect mechanism, potentially offsetting other metabolic benefits of metformin. Co-prescription with metformin should provide a greater target for selective 11βHSD1 inhibitors.
Collapse
Affiliation(s)
- Anna J Anderson
- University/British Heart Foundation Centre for Cardiovascular Science (A.J.A., R.A., N.Z.H., G.C.J., K.S., D.E.L., B.R.W., R.H.S), University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom; Diabetes Centre, Gartnavel General Hospital (G.C.J.), Glasgow, Scotland, United Kingdom; and Division of Medical Sciences and Graduate Entry Medicine (K.S.), School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| | - Ruth Andrew
- University/British Heart Foundation Centre for Cardiovascular Science (A.J.A., R.A., N.Z.H., G.C.J., K.S., D.E.L., B.R.W., R.H.S), University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom; Diabetes Centre, Gartnavel General Hospital (G.C.J.), Glasgow, Scotland, United Kingdom; and Division of Medical Sciences and Graduate Entry Medicine (K.S.), School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| | - Natalie Z Homer
- University/British Heart Foundation Centre for Cardiovascular Science (A.J.A., R.A., N.Z.H., G.C.J., K.S., D.E.L., B.R.W., R.H.S), University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom; Diabetes Centre, Gartnavel General Hospital (G.C.J.), Glasgow, Scotland, United Kingdom; and Division of Medical Sciences and Graduate Entry Medicine (K.S.), School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| | - Gregory C Jones
- University/British Heart Foundation Centre for Cardiovascular Science (A.J.A., R.A., N.Z.H., G.C.J., K.S., D.E.L., B.R.W., R.H.S), University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom; Diabetes Centre, Gartnavel General Hospital (G.C.J.), Glasgow, Scotland, United Kingdom; and Division of Medical Sciences and Graduate Entry Medicine (K.S.), School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| | - Kenneth Smith
- University/British Heart Foundation Centre for Cardiovascular Science (A.J.A., R.A., N.Z.H., G.C.J., K.S., D.E.L., B.R.W., R.H.S), University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom; Diabetes Centre, Gartnavel General Hospital (G.C.J.), Glasgow, Scotland, United Kingdom; and Division of Medical Sciences and Graduate Entry Medicine (K.S.), School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| | - Dawn E Livingstone
- University/British Heart Foundation Centre for Cardiovascular Science (A.J.A., R.A., N.Z.H., G.C.J., K.S., D.E.L., B.R.W., R.H.S), University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom; Diabetes Centre, Gartnavel General Hospital (G.C.J.), Glasgow, Scotland, United Kingdom; and Division of Medical Sciences and Graduate Entry Medicine (K.S.), School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| | - Brian R Walker
- University/British Heart Foundation Centre for Cardiovascular Science (A.J.A., R.A., N.Z.H., G.C.J., K.S., D.E.L., B.R.W., R.H.S), University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom; Diabetes Centre, Gartnavel General Hospital (G.C.J.), Glasgow, Scotland, United Kingdom; and Division of Medical Sciences and Graduate Entry Medicine (K.S.), School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| | - Roland H Stimson
- University/British Heart Foundation Centre for Cardiovascular Science (A.J.A., R.A., N.Z.H., G.C.J., K.S., D.E.L., B.R.W., R.H.S), University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom; Diabetes Centre, Gartnavel General Hospital (G.C.J.), Glasgow, Scotland, United Kingdom; and Division of Medical Sciences and Graduate Entry Medicine (K.S.), School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| |
Collapse
|
34
|
Gibb FW, Homer NZM, Faqehi AMM, Upreti R, Livingstone DE, McInnes KJ, Andrew R, Walker BR. Aromatase Inhibition Reduces Insulin Sensitivity in Healthy Men. J Clin Endocrinol Metab 2016; 101:2040-6. [PMID: 26967690 PMCID: PMC4870856 DOI: 10.1210/jc.2015-4146] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
CONTEXT Deficiency of aromatase, the enzyme that catalyzes the conversion of androgens to estrogens, is associated with insulin resistance in humans and mice. OBJECTIVE We hypothesized that pharmacological aromatase inhibition results in peripheral insulin resistance in humans. DESIGN This was a double-blind, randomized, controlled, crossover study. SETTING The study was conducted at a clinical research facility. PARTICIPANTS Seventeen healthy male volunteers (18-50 y) participated in the study. INTERVENTION The intervention included oral anastrozole (1 mg daily) and placebo, each for 6 weeks with a 2-week washout period. MAIN OUTCOME MEASURE Glucose disposal and rates of lipolysis were measured during a stepwise hyperinsulinemic euglycemic clamp. Data are mean (SEM). RESULTS Anastrozole therapy resulted in significant estradiol suppression (59.9 ± 3.6 vs 102.0 ± 5.7 pmol/L, P = < .001) and a more modest elevation of total T (25.8 ± 1.2 vs 21.4 ± 0.7 nmol/L, P = .003). Glucose infusion rate, during the low-dose insulin infusion, was lower after anastrozole administration (12.16 ± 1.33 vs 14.15 ± 1.55 μmol/kg·min, P = .024). No differences in hepatic glucose production or rate of lipolysis were observed. CONCLUSION Aromatase inhibition reduces insulin sensitivity, with respect to peripheral glucose disposal, in healthy men. Local generation and action of estradiol, at the level of skeletal muscle, is likely to be an important determinant of insulin sensitivity.
Collapse
Affiliation(s)
- Fraser W Gibb
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Natalie Z M Homer
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Abdullah M M Faqehi
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Rita Upreti
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Dawn E Livingstone
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Kerry J McInnes
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Ruth Andrew
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Brian R Walker
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| |
Collapse
|
35
|
Markey KA, Uldall M, Botfield H, Cato LD, Miah MAL, Hassan-Smith G, Jensen RH, Gonzalez AM, Sinclair AJ. Idiopathic intracranial hypertension, hormones, and 11β-hydroxysteroid dehydrogenases. J Pain Res 2016; 9:223-32. [PMID: 27186074 PMCID: PMC4847593 DOI: 10.2147/jpr.s80824] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Idiopathic intracranial hypertension (IIH) results in raised intracranial pressure (ICP) leading to papilledema, visual dysfunction, and headaches. Obese females of reproductive age are predominantly affected, but the underlying pathological mechanisms behind IIH remain unknown. This review provides an overview of pathogenic factors that could result in IIH with particular focus on hormones and the impact of obesity, including its role in neuroendocrine signaling and driving inflammation. Despite occurring almost exclusively in obese women, there have been a few studies evaluating the mechanisms by which hormones and adipokines exert their effects on ICP regulation in IIH. Research involving 11β-hydroxysteroid dehydrogenase type 1, a modulator of glucocorticoids, suggests a potential role in IIH. Improved understanding of the complex interplay between adipose signaling factors such as adipokines, steroid hormones, and ICP regulation may be key to the understanding and future management of IIH.
Collapse
Affiliation(s)
- Keira A Markey
- Neurometabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Maria Uldall
- Danish Headache Center, Clinic of Neurology, Rigshospitalet-Glostrup, University of Copenhagen, Glostrup, Denmark
| | - Hannah Botfield
- Neurometabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Liam D Cato
- Neurometabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Mohammed A L Miah
- Neurometabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Ghaniah Hassan-Smith
- Neurometabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Rigmor H Jensen
- Danish Headache Center, Clinic of Neurology, Rigshospitalet-Glostrup, University of Copenhagen, Glostrup, Denmark
| | - Ana M Gonzalez
- Neurometabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Alexandra J Sinclair
- Neurometabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
36
|
Incollingo Rodriguez AC, Epel ES, White ML, Standen EC, Seckl JR, Tomiyama AJ. Hypothalamic-pituitary-adrenal axis dysregulation and cortisol activity in obesity: A systematic review. Psychoneuroendocrinology 2015; 62:301-18. [PMID: 26356039 DOI: 10.1016/j.psyneuen.2015.08.014] [Citation(s) in RCA: 266] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 08/18/2015] [Accepted: 08/18/2015] [Indexed: 12/29/2022]
Abstract
BACKGROUND Although there is substantial evidence of differential hypothalamic-pituitary-adrenal (HPA) axis activity in both generalized and abdominal obesity, consistent trends in obesity-related HPA axis perturbations have yet to be identified. OBJECTIVES To systematically review the existing literature on HPA activity in obesity, identify possible explanations for inconsistencies in the literature, and suggest methodological improvements for future study. DATA SOURCES Included papers used Pubmed, Google Scholar, and the University of California Library search engines with search terms body mass index (BMI), waist-to-hip ratio (WHR), waist circumference, sagittal diameter, abdominal versus peripheral body fat distribution, body fat percentage, DEXA, abdominal obesity, and cortisol with terms awakening response, slope, total daily output, reactivity, feedback sensitivity, long-term output, and 11β-HSD expression. STUDY ELIGIBILITY CRITERIA Empirical research papers were eligible provided that they included at least one type of obesity (general or abdominal), measured at least one relevant cortisol parameter, and a priori tested for a relationship between obesity and cortisol. RESULTS A general pattern of findings emerged where greater abdominal fat is associated with greater responsivity of the HPA axis, reflected in morning awakening and acute stress reactivity, but some studies did show underresponsiveness. When examined in adipocytes, there is a clear upregulation of cortisol output (due to greater expression of 11β-HSD1), but in hepatic tissue this cortisol is downregulated. Overall obesity (BMI) appears to also be related to a hyperresponsive HPA axis in many but not all studies, such as when acute reactivity is examined. LIMITATIONS The reviewed literature contains numerous inconsistencies and contradictions in research methodologies, sample characteristics, and results, which partially precluded the development of clear and reliable patterns of dysregulation in each investigated cortisol parameter. CONCLUSIONS AND IMPLICATIONS The literature to date is inconclusive, which may well arise from differential effects of generalized obesity vs. abdominal obesity or from modulators such as sex, sex hormones, and chronic stress. While the relationship between obesity and adipocyte cortisol seems to be clear, further research is warranted to understand how adipocyte cortisol metabolism influences circulating cortisol levels and to establish consistent patterns of perturbations in adrenal cortisol activity in both generalized and abdominal obesity.
Collapse
Affiliation(s)
| | - Elissa S Epel
- University of California, San Francisco, CA 94118, USA
| | - Megan L White
- University of California, Los Angeles, CA 90095, USA
| | | | - Jonathan R Seckl
- University of Edinburgh, Edinburgh EH1 1HT, Scotland, United Kingdom
| | | |
Collapse
|
37
|
A potent and selective 11β-hydroxysteroid dehydrogenase type 1 inhibitor, SKI2852, ameliorates metabolic syndrome in diabetic mice models. Eur J Pharmacol 2015; 768:139-48. [PMID: 26519792 DOI: 10.1016/j.ejphar.2015.10.042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 10/22/2015] [Accepted: 10/26/2015] [Indexed: 11/22/2022]
Abstract
11β-Hydroxysteroid dehydrogenase type 1 (11βHSD1) has been targeted for new drugs to treat type 2 diabetes and metabolic syndrome. In this study, we determined whether the inhibition of 11βHSD1 with a new selective inhibitor, SKI2852, could improve lipid profiles, glucose levels, and insulin sensitivity in type 2 diabetic and obese conditions. SKI2852 showed a potent inhibition of cortisone to cortisol conversion for over 80% in both liver and adipose tissue ex vivo from orally administered C57BL/6 mice, and in vivo analysis results were consistent with this. Repeated oral administrations of SKI2852 in diet-induced obesity (DIO) and ob/ob mice revealed a partially beneficial effect of SKI2852 in improving levels of cholesterols, triglycerides, free fatty acids, postprandial glucose, and/or blood hemoglobinA1c. SKI2852 significantly reduced body weight increase in ob/ob mice, and efficiently suppressed hepatic mRNA levels of gluconeogenic enzymes in DIO mice. Moreover, SKI2852 enhanced hepatic and whole body insulin sensitivities in hyperinsulinemic-euglycemic clamp experiment in DIO mice. In conclusion, these results indicate that selective and potent inhibition of 11βHSD1 by SKI2852, thus blockade of active glucocorticoid conversion, may improve many aspects of metabolic parameters in type 2 diabetes and metabolic diseases, mainly by inhibitions of hepatic gluconeogenesis and partial improvements of lipid profiles. Our study strongly support that SKI2852 may have a great potential as a novel candidate drug for the treatment of diabetes and metabolic diseases.
Collapse
|
38
|
Teshima T, Matsumoto H, Okusa T, Nakamura Y, Koyama H. Effects of Carbenoxolone on the Canine Pituitary-Adrenal Axis. PLoS One 2015; 10:e0135516. [PMID: 26262685 PMCID: PMC4532459 DOI: 10.1371/journal.pone.0135516] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/13/2015] [Indexed: 11/25/2022] Open
Abstract
Cushing’s disease caused by pituitary corticotroph adenoma is a common endocrine disease in dogs. A characteristic biochemical feature of corticotroph adenomas is their relative resistance to suppressive negative feedback by glucocorticoids. The abnormal expression of 11beta-hydroxysteroid dehydrogenase (11HSD), which is a cortisol metabolic enzyme, is found in human and murine corticotroph adenomas. Our recent studies demonstrated that canine corticotroph adenomas also have abnormal expression of 11HSD. 11HSD has two isoforms in dogs, 11HSD type1 (HSD11B1), which converts cortisone into active cortisol, and 11HSD type2 (HSD11B2), which converts cortisol into inactive cortisone. It has been suggested that glucocorticoid resistance in corticotroph tumors is related to the overexpression of HSD11B2. Therefore it was our aim to investigate the effects of carbenoxolone (CBX), an 11HSD inhibitor, on the healthy dog’s pituitary-adrenal axis. Dogs were administered 50 mg/kg of CBX twice each day for 15 days. During CBX administration, no adverse effects were observed in any dogs. The plasma adrenocorticotropic hormone (ACTH), and serum cortisol and cortisone concentrations were significantly lower at day 7 and 15 following corticotropin releasing hormone stimulation. After completion of CBX administration, the HSD11B1 mRNA expression was higher, and HSD11B2 mRNA expression was significantly lower in the pituitaries. Moreover, proopiomelanocortin mRNA expression was lower, and the ratio of ACTH-positive cells in the anterior pituitary was also significantly lower after CBX treatment. In adrenal glands treated with CBX, HSD11B1 and HSD11B2 mRNA expression were both lower compared to normal canine adrenal glands. The results of this study suggested that CBX inhibits ACTH secretion from pituitary due to altered 11HSD expressions, and is potentially useful for the treatment of canine Cushing’s disease.
Collapse
Affiliation(s)
- Takahiro Teshima
- Division of Therapeutic Science I, Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602, Japan
- * E-mail:
| | - Hirotaka Matsumoto
- Division of Therapeutic Science I, Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602, Japan
| | - Tomoko Okusa
- Division of Therapeutic Science I, Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602, Japan
| | - Yumi Nakamura
- Division of Therapeutic Science I, Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602, Japan
| | - Hidekazu Koyama
- Division of Therapeutic Science I, Department of Veterinary Clinical Medicine, School of Veterinary Medicine, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602, Japan
| |
Collapse
|
39
|
Byun SY, Shin YJ, Nam KY, Hong SP, Ahn SK. A novel highly potent and selective 11β-hydroxysteroid dehydrogenase type 1 inhibitor, UI-1499. Life Sci 2015; 120:1-7. [DOI: 10.1016/j.lfs.2014.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 10/15/2014] [Accepted: 11/01/2014] [Indexed: 01/27/2023]
|
40
|
Tagawa N, Kubota S, Kobayashi Y, Kato I. Genistein inhibits glucocorticoid amplification in adipose tissue by suppression of 11β-hydroxysteroid dehydrogenase type 1. Steroids 2015; 93:77-86. [PMID: 25447798 DOI: 10.1016/j.steroids.2014.11.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 10/15/2014] [Accepted: 11/18/2014] [Indexed: 11/20/2022]
Abstract
Excess glucocorticoids promote visceral obesity, hyperlipidemia, and insulin resistance. The main regulator of intracellular glucocorticoid levels is 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), which converts inactive glucocorticoids into bioactive forms such as cortisol in humans and corticosterone in rodents. Hexose-6-phosphate dehydrogenase (H6PD), which is colocalized with 11β-HSD1 in the intralumen of the endoplasmic reticulum, supplies a crucial coenzyme, NADPH, for full reductase activity of 11β-HSD1. Therefore, it is possible that inhibition of 11β-HSD1 will become a considerable medical treatment for metabolic diseases including obesity and diabetes. Genistein, a soy isoflavone, has received attention for its therapeutic potential for obesity, diabetes, and cardiovascular disease, and has been proposed as a promising compound for the treatment of metabolic disorders. However, the mechanisms underlying the pleiotropic anti-obesity effects of genistein have not been fully clarified. Here, we demonstrate that genistein was able to inhibit 11β-HSD1 and H6PD activities within 10 or 20min, in dose- and time-dependent manners. Inhibition of 11β-HSD2 activity was not observed in rat kidney microsomes. The inhibition was not reversed by two estrogen receptor antagonists, tamoxifen and ICI182,780. A kinetic study revealed that genistein acted as a non-competitive inhibitor of 11β-HSD1, and its apparent Km value for 11-dehydrocorticosterone was 0.5μM. Genistein also acted as a non-competitive inhibitor of H6PD, and its apparent Km values for G6P and NADP were 0.9 and 3.3μM, respectively. These results suggest that genistein may exert its inhibitory effect by interacting with these enzymes.
Collapse
Affiliation(s)
- Noriko Tagawa
- Department of Medical Biochemistry, Kobe Pharmaceutical University, Kobe, Japan.
| | - Sayaka Kubota
- Department of Medical Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Yoshiharu Kobayashi
- Department of Medical Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Ikuo Kato
- Department of Medical Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| |
Collapse
|
41
|
Dube S, Norby BJ, Pattan V, Carter RE, Basu A, Basu R. 11β-hydroxysteroid dehydrogenase types 1 and 2 activity in subcutaneous adipose tissue in humans: implications in obesity and diabetes. J Clin Endocrinol Metab 2015; 100:E70-6. [PMID: 25303491 PMCID: PMC4283013 DOI: 10.1210/jc.2014-3017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
CONTEXT The role of 11β-hydroxysteroid dehydrogenase types 1 (11β-HSD-1) and 2 (11β-HSD-2) enzymes in sc adipose tissue is controversial. OBJECTIVE The objective of the study was to determine the activity of 11β-HSD-1 and -2 enzymes in the abdominal and leg sc adipose tissue in obesity and diabetes. DESIGN 11β-HSD-1 and -2 enzyme activities in abdominal and leg sc adipose tissue were measured by infusing [2,2,4,6,6,12,12-(2)H7] cortisone (D7 cortisone) and [9,12,12-(2)H3] cortisol (D3 cortisol) via microdialysis catheters placed in sc fat depots. SETTING The study was conducted at the Mayo Clinic Clinical Research Unit. PARTICIPANTS Lean nondiabetic (n = 13), overweight/obese nondiabetic (n = 15), and overweight/obese participants with type 2 diabetes mellitus (n = 15) participated in the study. MAIN OUTCOME MEASURES The conversion of infused D7 cortisone to D7 cortisol (via 11β-HSD reductase activity) and D3 cortisol to D3 cortisone (via 11β-HSD dehydrogenase activity) in sc adipose tissue. RESULTS Enrichment of D7 cortisone and D3 cortisol were similar in the effluents from both sites in all groups. D3 cortisone enrichment did not differ in the three cohorts, indicating that 11β-HSD-2 enzyme activity (conversion of cortisol to cortisone) occurs equally in all groups. However, D7 cortisol enrichment was detectable in abdominal sc fat of overweight/obese participants with type 2 diabetes mellitus only, implying 11β-HSD-1 reductase activity (conversion of cortisone to cortisol) occurs in obese subjects with type 2 diabetes. CONCLUSIONS There is conversion of cortisone to cortisol via the 11β-HSD-1 enzyme pathway in abdominal sc fat depots in overweight/obese participants with type 2 diabetes mellitus. This observation has significant implications for developing tissue-specific 11β-HSD-1 inhibitors in type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Simmi Dube
- Endocrine Research Unit (S.D., B.J.N., V.P., A.B., R.B.), Division of Endocrinology, Diabetes, Metabolism, and Nutrition, and Division of Biomedical Statistics and Informatics (R.E.C.), Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota 55905
| | | | | | | | | | | |
Collapse
|
42
|
Corey SM, Epel E, Schembri M, Pawlowsky SB, Cole RJ, Araneta MRG, Barrett-Connor E, Kanaya AM. Effect of restorative yoga vs. stretching on diurnal cortisol dynamics and psychosocial outcomes in individuals with the metabolic syndrome: the PRYSMS randomized controlled trial. Psychoneuroendocrinology 2014; 49:260-71. [PMID: 25127084 PMCID: PMC4174464 DOI: 10.1016/j.psyneuen.2014.07.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 07/11/2014] [Accepted: 07/12/2014] [Indexed: 11/25/2022]
Abstract
PURPOSE Chronic stimulation and dysregulation of the neuroendocrine system by stress may cause metabolic abnormalities. We estimated how much cortisol and psychosocial outcomes improved with a restorative yoga (relaxation) versus a low impact stretching intervention for individuals with the metabolic syndrome. METHODS We conducted a 1-year multi-center randomized controlled trial (6-month intervention and 6-month maintenance phase) of restorative yoga vs. stretching. Participants completed surveys to assess depression, social support, positive affect, and stress at baseline, 6 months and 12 months. For each assessment, we collected saliva at four points daily for three days and collected response to dexamethasone on the fourth day for analysis of diurnal cortisol dynamics. We analyzed our data using multivariate regression models, controlling for study site, medications (antidepressants, hormone therapy), body mass index, and baseline cortisol values. RESULTS Psychosocial outcome measures were available for 171 study participants at baseline, 140 at 6 months, and 132 at 1 year. Complete cortisol data were available for 136 of 171 study participants (72 in restorative yoga and 64 in stretching) and were only available at baseline and 6 months. At 6 months, the stretching group had decreased cortisol at waking and bedtime compared to the restorative yoga group. The pattern of changes in stress mirrored this improvement, with the stretching group showing reductions in chronic stress severity and perseverative thoughts about their stress. Perceived stress decreased by 1.5 points (-0.4; 3.3, p=0.11) at 6 months, and by 2.0 points (0.1; 3.9, p=0.04) at 1 year in the stretching compared to restorative yoga groups. Post hoc analyses suggest that in the stretching group only, perceived increases in social support (particularly feelings of belonging), but not changes in stress were related to improved cortisol dynamics. CONCLUSIONS We found significant decreases in salivary cortisol, chronic stress severity, and stress perception in the stretching group compared to the restorative yoga group. Group support during the interactive stretch classes may have contributed to these changes.
Collapse
Affiliation(s)
- Sarah M Corey
- University of California, San Francisco, CA 94115, USA.
| | - Elissa Epel
- University of California, San Francisco, CA 94115, USA
| | | | | | - Roger J Cole
- Synchrony Applied Health Sciences, Del Mar, CA 92014, USA
| | | | | | - Alka M Kanaya
- University of California, San Francisco, CA 94115, USA
| |
Collapse
|
43
|
Valsamakis G, Lois K, Kumar S, Mastorakos G. New molecular targets in the pathophysiology of obesity and available treatment options under investigation. Clin Obes 2014; 4:209-19. [PMID: 25826792 DOI: 10.1111/cob.12064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 05/12/2014] [Accepted: 05/14/2014] [Indexed: 12/22/2022]
Abstract
The pharmacotherapy of obesity has historically recorded an overall poor safety and efficacy profile largely because of the complex mechanisms involved in the pathophysiology of obesity. It is hoped that a better understanding of the regulation of body weight will lead us to the development of effective and safer drugs. Recent advances in our understanding of the regulation of energy homeostasis has allowed the design of novel anti-obesity drugs targeting specific molecules crucial for the modulation of energy balance, including drugs that induce satiety, modulate nutrient absorption or influence metabolism or lipogenesis. Almost a decade after the Food and Drug Administration approved the first weight loss medication, it recently approved two novel anti-obesity drugs Belviq (lorcaserin) and Qsymia (topiramate and phentermine), thus signalling the beginning of a new era in the pharmacotherapy of obesity. It is believed that the next generation of weight-loss drugs will be based on combination treatments with gut hormones in a manner that mimics the changes underlying surgically induced weight loss thus introducing the so called 'bariatric pharmacotherapy'. An in-depth understanding of the interrelated physiological and behavioural effects of these new molecules together with the development of new treatment paradigms is needed so that future disappointments in the field of obesity pharmacotherapy may be avoided.
Collapse
Affiliation(s)
- G Valsamakis
- Endocrine Unit, 2nd Department of Obs and Gynae, Areteeion University Hospital, Athens Medical School National and Kapodistrian University of Athens, Athens, Greece; WISDEM Centre for Diabetes, Endocrinology and Metabolism, Warwick Medical School, University of Warwick, Coventry, UK
| | | | | | | |
Collapse
|
44
|
Upreti R, Hughes KA, Livingstone DEW, Gray CD, Minns FC, Macfarlane DP, Marshall I, Stewart LH, Walker BR, Andrew R. 5α-reductase type 1 modulates insulin sensitivity in men. J Clin Endocrinol Metab 2014; 99:E1397-406. [PMID: 24823464 PMCID: PMC4207930 DOI: 10.1210/jc.2014-1395] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
CONTEXT 5α-Reductase (5αR) types 1 and 2 catalyze the A-ring reduction of steroids, including androgens and glucocorticoids. 5α-R inhibitors lower dihydrotestosterone in benign prostatic hyperplasia; finasteride inhibits 5αR2, and dutasteride inhibits both 5αR2 and 5αR1. In rodents, loss of 5αR1 promotes fatty liver. OBJECTIVE Our objective was to test the hypothesis that inhibition of 5αR1 causes metabolic dysfunction in humans. DESIGN, SETTING, AND PARTICIPANTS This double-blind randomized controlled parallel group study at a clinical research facility included 46 men (20-85 years) studied before and after intervention. INTERVENTION Oral dutasteride (0.5 mg daily; n = 16), finasteride (5 mg daily; n = 16), or control (tamsulosin; 0.4 mg daily; n = 14) was administered for 3 months. MAIN OUTCOME MEASURE Glucose disposal was measured during a stepwise hyperinsulinemic-euglycemic clamp. Data are mean (SEM). RESULTS Dutasteride and finasteride had similar effects on steroid profiles, with reduced urinary androgen and glucocorticoid metabolites and reduced circulating DHT but no change in plasma or salivary cortisol. Dutasteride, but not finasteride, reduced stimulation of glucose disposal by high-dose insulin (dutasteride by -5.7 [3.2] μmol/kg fat-free mass/min, versus finasteride +7.2 [3.0], and tamsulosin +7.0 [2.0]). Dutasteride also reduced suppression of nonesterified fatty acids by insulin and increased body fat (by 1.6% [0.6%]). Glucose production and glycerol turnover were unchanged. Consistent with metabolic effects of dutasteride being mediated in peripheral tissues, mRNA for 5αR1 but not 5αR2 was detected in human adipose tissue. CONCLUSION Dual inhibition of 5αRs, but not inhibition of 5αR2 alone, modulates insulin sensitivity in human peripheral tissues rather than liver. This may have important implications for patients prescribed dutasteride for prostatic disease.
Collapse
Affiliation(s)
- Rita Upreti
- University/British Heart Foundation Centre for Cardiovascular Science (R.U., K.A.H., D.E.W.L., D.P.M., I.M., B.R.W., R.A.) and Clinical Research Imaging Centre (C.D.G.), University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom; and Radiology (F.C.M.) and Urology (L.H.S.) Departments, National Health Service Lothian University Hospitals Division, Western General Hospital, Edinburgh EH4 2XU, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Gambineri A, Fanelli F, Tomassoni F, Munarini A, Pagotto U, Andrew R, Walker BR, Pasquali R. Tissue-specific dysregulation of 11β-hydroxysteroid dehydrogenase type 1 in overweight/obese women with polycystic ovary syndrome compared with weight-matched controls. Eur J Endocrinol 2014; 171:47-57. [PMID: 24743397 DOI: 10.1530/eje-13-1030] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
CONTEXT Abnormal cortisol metabolism in polycystic ovary syndrome (PCOS) has been invoked as a cause of secondary activation of the hypothalamic-pituitary-adrenal axis and hence androgen excess. However, this is based on urinary excretion of cortisol metabolites, which cannot detect tissue-specific changes in metabolism and may be confounded by obesity. OBJECTIVE To assess cortisol clearance and whole-body and tissue-specific activities of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1 (HSD11B1)) in PCOS. DESIGN Case-control study. SETTING Medical center. PATIENTS A total of 20 overweight-obese unmedicated Caucasian women with PCOS, aged 18-45 years, and 20 Caucasian controls matched for age, BMI, body fat distribution, and HSD11B1 genotypes (rs846910 and rs12086634). MAIN OUTCOME MEASURES Cortisol metabolites were measured in 24 h urine. During steady-state 9,11,12,12-[(2)H]4-cortisol infusion, cortisol clearance was calculated and whole-body HSD11B1 activity was assessed as the rate of appearance of 9,12,12-(2)H3-cortisol (d3-cortisol). Hepatic HSD11B1 activity was quantified as the generation of plasma cortisol following an oral dose of cortisone. Subcutaneous adipose HSD11B1 activity and HSD11B1 mRNA were measured, ex vivo, in biopsies. RESULTS Urinary cortisol metabolite excretion, deuterated cortisol clearance, and the rate of appearance of d3-cortisol did not differ between patients with PCOS and controls. However, hepatic HSD11B1 conversion of oral cortisone to cortisol was impaired (P<0.05), whereas subcutaneous abdominal adipose tissue HSD11B1 mRNA levels and activity were increased (P<0.05) in women with PCOS when compared with controls. CONCLUSIONS Tissue-specific dysregulation of HSD11B1 is a feature of PCOS, over and above obesity, whereas increased clearance of cortisol may result from obesity rather than PCOS.
Collapse
Affiliation(s)
- Alessandra Gambineri
- Division of EndocrinologyDepartment of Medical and Surgical Science, Centre for Applied Biomedical Research (C.R.B.A.), S. Orsola-Malpighi Hospital, University of Bologna, University Alma Mater Studiorum, Via Massarenti 9, 40138 Bologna, ItalyEndocrinology UnitQueen's Medical Research Institute, University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | - Flaminia Fanelli
- Division of EndocrinologyDepartment of Medical and Surgical Science, Centre for Applied Biomedical Research (C.R.B.A.), S. Orsola-Malpighi Hospital, University of Bologna, University Alma Mater Studiorum, Via Massarenti 9, 40138 Bologna, ItalyEndocrinology UnitQueen's Medical Research Institute, University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | - Federica Tomassoni
- Division of EndocrinologyDepartment of Medical and Surgical Science, Centre for Applied Biomedical Research (C.R.B.A.), S. Orsola-Malpighi Hospital, University of Bologna, University Alma Mater Studiorum, Via Massarenti 9, 40138 Bologna, ItalyEndocrinology UnitQueen's Medical Research Institute, University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | - Alessandra Munarini
- Division of EndocrinologyDepartment of Medical and Surgical Science, Centre for Applied Biomedical Research (C.R.B.A.), S. Orsola-Malpighi Hospital, University of Bologna, University Alma Mater Studiorum, Via Massarenti 9, 40138 Bologna, ItalyEndocrinology UnitQueen's Medical Research Institute, University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | - Uberto Pagotto
- Division of EndocrinologyDepartment of Medical and Surgical Science, Centre for Applied Biomedical Research (C.R.B.A.), S. Orsola-Malpighi Hospital, University of Bologna, University Alma Mater Studiorum, Via Massarenti 9, 40138 Bologna, ItalyEndocrinology UnitQueen's Medical Research Institute, University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | - Ruth Andrew
- Division of EndocrinologyDepartment of Medical and Surgical Science, Centre for Applied Biomedical Research (C.R.B.A.), S. Orsola-Malpighi Hospital, University of Bologna, University Alma Mater Studiorum, Via Massarenti 9, 40138 Bologna, ItalyEndocrinology UnitQueen's Medical Research Institute, University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | - Brian R Walker
- Division of EndocrinologyDepartment of Medical and Surgical Science, Centre for Applied Biomedical Research (C.R.B.A.), S. Orsola-Malpighi Hospital, University of Bologna, University Alma Mater Studiorum, Via Massarenti 9, 40138 Bologna, ItalyEndocrinology UnitQueen's Medical Research Institute, University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | - Renato Pasquali
- Division of EndocrinologyDepartment of Medical and Surgical Science, Centre for Applied Biomedical Research (C.R.B.A.), S. Orsola-Malpighi Hospital, University of Bologna, University Alma Mater Studiorum, Via Massarenti 9, 40138 Bologna, ItalyEndocrinology UnitQueen's Medical Research Institute, University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| |
Collapse
|
46
|
Grolmusz VK, Ács OD, Feldman-Kovács K, Szappanos Á, Stenczer B, Fekete T, Szendei G, Reismann P, Rácz K, Patócs A. Genetic variants of the HSD11B1 gene promoter may be protective against polycystic ovary syndrome. Mol Biol Rep 2014; 41:5961-9. [DOI: 10.1007/s11033-014-3473-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 06/14/2014] [Indexed: 01/08/2023]
|
47
|
Stomby A, Andrew R, Walker BR, Olsson T. Tissue-specific dysregulation of cortisol regeneration by 11βHSD1 in obesity: has it promised too much? Diabetologia 2014; 57:1100-10. [PMID: 24710966 DOI: 10.1007/s00125-014-3228-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 03/11/2014] [Indexed: 01/24/2023]
Abstract
Cushing's syndrome, caused by increased production of cortisol, leads to metabolic dysfunction including visceral adiposity, hypertension, hyperlipidaemia and type 2 diabetes. The similarities with the metabolic syndrome are striking and major efforts have been made to find obesity-associated changes in the regulation of glucocorticoid action and synthesis, both at a systemic level and tissue level. Obesity is associated with tissue-specific alterations in glucocorticoid metabolism, with increased activity of the glucocorticoid-regenerating enzyme 11β-hydroxysteroid dehydrogenase type 1 (11βHSD1) in subcutaneous adipose tissue and decreased conversion of cortisone to cortisol, interpreted as decreased 11βHSD1 activity, in the liver. In addition, genetic manipulation of 11βHSD1 activity in rodents can either induce (by overexpression of Hsd11b1, the gene encoding 11βHSD1) or prevent (by knocking out Hsd11b1) obesity and metabolic dysfunction. Taken together with earlier evidence that non-selective inhibitors of 11βHSD1 enhance insulin sensitivity, these results led to the hypothesis that inhibition of 11βHSD1 might be a promising target for treatment of the metabolic syndrome. Several selective 11βHSD1 inhibitors have now been developed and shown to improve metabolic dysfunction in patients with type 2 diabetes, but the small magnitude of the glucose-lowering effect has precluded their further commercial development.This review focuses on the role of 11βHSD1 as a tissue-specific regulator of cortisol exposure in obesity and type 2 diabetes in humans. We consider the potential of inhibition of 11βHSD1 as a therapeutic strategy that might address multiple complications in patients with type 2 diabetes, and provide our thoughts on future directions in this field.
Collapse
Affiliation(s)
- Andreas Stomby
- Department for Public Health and Clinical Medicine, Medicine, Umeå University, Umeå, Sweden
| | | | | | | |
Collapse
|
48
|
Anderson A, Walker BR. 11β-HSD1 inhibitors for the treatment of type 2 diabetes and cardiovascular disease. Drugs 2014; 73:1385-93. [PMID: 23990334 DOI: 10.1007/s40265-013-0112-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Inhibition of the enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) has been proposed as a novel therapeutic target for the treatment of type 2 diabetes mellitus. Over 170 new compounds targeting 11β-HSD1 have been developed. This article reviews the current published literature on compounds that have reached phase II clinical trials in patients with type 2 diabetes, and summarises the preclinical evidence that such agents may be useful for associated conditions, including peripheral vascular disease, coronary artery disease and cognitive decline. In clinical trials, 11β-HSD1 inhibitors have been well tolerated and have improved glycaemic control, lipid profile and blood pressure, and induced modest weight loss. The magnitude of the effects are small relative to other agents, so that further development of 11β-HSD1 inhibitors for the primary therapeutic indication of type 2 diabetes has stalled. Ongoing programmes are focused on additional benefits for cognitive function and other cardiovascular risk factors.
Collapse
Affiliation(s)
- Anna Anderson
- University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | | |
Collapse
|
49
|
Scott JS, Goldberg FW, Turnbull AV. Medicinal Chemistry of Inhibitors of 11β-Hydroxysteroid Dehydrogenase Type 1 (11β-HSD1). J Med Chem 2013; 57:4466-86. [DOI: 10.1021/jm4014746] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- James S. Scott
- AstraZeneca Innovative Medicines, Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, U.K
| | - Frederick W. Goldberg
- AstraZeneca Innovative Medicines, Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, U.K
| | - Andrew V. Turnbull
- AstraZeneca Innovative Medicines, Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, U.K
| |
Collapse
|
50
|
Methlie P, Dankel S, Myhra T, Christensen B, Gjerde J, Fadnes D, Våge V, Løvås K, Mellgren G. Changes in adipose glucocorticoid metabolism before and after bariatric surgery assessed by direct hormone measurements. Obesity (Silver Spring) 2013; 21:2495-503. [PMID: 23512832 DOI: 10.1002/oby.20449] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 02/21/2013] [Indexed: 12/29/2022]
Abstract
OBJECTIVES Increased intra-adipose cortisol is thought to promote obesity, but few human studies have investigated intra-adipose glucocorticoid hormones and none have demonstrated prospective changes with fat loss. DESIGN AND METHODS Subcutaneous adipose tissue (SAT) was obtained from obese subjects before and 1-year after surgery-induced fat loss, and from nonobese controls. In a second similar cohort of obese subjects, adipocytes and stromal-vascular fraction were isolated. Intra-adipose cortisol and cortisone levels were analyzed by liquid chromatography mass spectrometry and HSD11B1/HSD11B2 mRNA by qPCR. RESULTS SAT cortisol/cortisone ratio before fat loss, median 4.8 (interquartile range, 4.1-5.7), was higher than after fat loss, 1.9 (1.0-2.7) (P = 0.001), and compared to nonobese controls, 3.2 (2.4-3.9) (P = 0.005). Cortisone before fat loss, 2.3 (1.2-2.9) nmol/kg, was lower than after fat loss, 5.8 (3.0-10.2) nmol/kg (P = 0.042), and compared to controls, 5.1 (3.8-6.7) nmol/kg (P = 0.013). HSD11B1 was predominantly expressed in mature adipocytes, whereas HSD11B2 was expressed at a higher level in stromal-vascular fraction. CONCLUSIONS The intra-adipose glucocorticoid metabolism was markedly altered in the extremely obese state with increased cortisol levels relative to cortisone, whereas fat loss restored this balance approximating nonobese subjects. Changes were more pronounced for cortisone than cortisol, suggesting an adaptive response to insufficient intra-adipose cortisol levels in obesity.
Collapse
Affiliation(s)
- Paal Methlie
- Department of Clinical Science, University of Bergen, Bergen, Norway; Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|