1
|
Chen Z, Wei D, Zhao M, Shi J, Ma C, Zhang C, Lin H, Huo W, Wang C, Fan C, Mao Z. Associations of serum glucocorticoid levels on hypertension and blood pressure-related indicators: a nested case-control study in rural China. J Hypertens 2024; 42:1555-1565. [PMID: 38747439 DOI: 10.1097/hjh.0000000000003758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
BACKGROUND The relationship between glucocorticoids and hypertension has shown inconsistent findings in previous studies. To address this, our study employed a nested case-control design in rural areas to further investigate the association between serum glucocorticoid levels and hypertension, and blood pressure-related indicators. METHODS This study employed a nested case-control design, involving 560 pairs of hypertensive cases and matched controls. The concentrations of serum cortisol (F), cortisone (E) and 11-deoxycortisol (S) were determined using liquid chromatography-tandem mass spectrometry. We employed various methods, including generalized linear model (GLM), conditional logistic regression model, restricted cubic spline regression, subgroup analysis, interaction, and joint effects, with adjustments for multiple covariates to analyze the relationships between glucocorticoids, hypertension, and blood pressure-related indicators. RESULTS After multivariable adjustments, ln-F, ln-F/E, and ln-S were positively associated with SBP, DBP, pulse pressure (PP), and mean arterial pressure (MAP), while ln-E was negatively associated with DBP and MAP ( P < 0.05). Interestingly, ln-S showed no statistically significant association with hypertension prevalence ( P > 0.05), whereas ln-F and ln-F/E were positively associated with it ( P < 0.05). The adjusted odds ratios (ORs) and 95% confidence intervals (CIs) were 1.153 (1.011-1.315) for ln-F and 2.072 (1.622-2.645) for ln-F/E, respectively. In contrast, ln-E exhibited a negative association with hypertension prevalence (adjusted OR = 0.837, 95% CI 0.714-0.982). Moreover, a significant association was observed between the combined use of high-dose F/E and high-dose S with hypertension prevalence (adjusted OR = 3.273, 95% CI 2.013-5.321). Blood pressure indicators and hypertension prevalence significantly increased with elevated serum F and F/E concentrations ( P < 0.05). Interaction analysis further revealed that among women, the positive association between F/E and hypertension prevalence was more pronounced than in men ( P < 0.05), and S exhibited a more significant positive association with hypertension prevalence in the overweight population ( P < 0.05). CONCLUSION Serum F/E and S levels demonstrated positive associations with hypertension and blood pressure-related indicators, and their combined influence exhibited a synergistic effect on hypertension. Notably, F, F/E, and S were associated with heightened hypertension risk, warranting particular attention in women and overweight populations.
Collapse
Affiliation(s)
- Zhiwei Chen
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan
| | - Dandan Wei
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan
| | - Mengzhen Zhao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan
| | - Jiayu Shi
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan
| | - Cuicui Ma
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan
| | - Caiyun Zhang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan
| | - Hualiang Lin
- Department of Epidemiology, School of Public Health, Sun Yat Sen University, Guangzhou
| | - Wenqian Huo
- Department of Occupational and Environmental Health Sciences, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Chongjian Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan
| | - Caini Fan
- Department of Hypertension, Henan Provincial People's Hospital, Zhengzhou
| | - Zhenxing Mao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan
| |
Collapse
|
2
|
Zhang D, Xu D, Chen X, Zhou H, Xu G. Divergent Entry to Walsucochin Nortriterpenoids: Total Syntheses of (±)-Walsucochin A and (±)-Walsucochinoids C-F. J Org Chem 2021; 86:7271-7279. [PMID: 33978408 DOI: 10.1021/acs.joc.1c00401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Nortriterpenoids isolated from Walsura cochinchinensis have attracted much attention from both synthetic and medicinal chemists, yet only recently have efficient synthetic approaches to any members appeared. Shown here is that the common intermediate with a 6/6/5/6-fused tetracyclic ring nucleus can be converted to walsucochin family members. The first total syntheses of (±)-walsucochin A, (±)-walsucochinoids C-F, and their analogues were achieved in this work.
Collapse
Affiliation(s)
- Danyang Zhang
- College of Plant Protection, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China
| | - Dan Xu
- College of Chemistry & Pharmacy, Northwest A&F University, 22 Xinong Road, Yangling 712100, Shaanxi, China.,State Key Laboratory of Crop Stress Biology for Arid Areas, Key Laboratory of Botanical Pesticide R&D in Shaanxi Province, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Yangling 712100, Shaanxi, China
| | - Xinyue Chen
- College of Plant Protection, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China
| | - Huan Zhou
- College of Plant Protection, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China.,State Key Laboratory of Crop Stress Biology for Arid Areas, Key Laboratory of Botanical Pesticide R&D in Shaanxi Province, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Yangling 712100, Shaanxi, China
| | - Gong Xu
- College of Plant Protection, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China.,State Key Laboratory of Crop Stress Biology for Arid Areas, Key Laboratory of Botanical Pesticide R&D in Shaanxi Province, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Yangling 712100, Shaanxi, China
| |
Collapse
|
3
|
Gomez-Sanchez EP, Gomez-Sanchez CE. 11β-hydroxysteroid dehydrogenases: A growing multi-tasking family. Mol Cell Endocrinol 2021; 526:111210. [PMID: 33607268 PMCID: PMC8108011 DOI: 10.1016/j.mce.2021.111210] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/02/2021] [Accepted: 02/07/2021] [Indexed: 02/06/2023]
Abstract
This review briefly addresses the history of the discovery and elucidation of the three cloned 11β-hydroxysteroid dehydrogenase (11βHSD) enzymes in the human, 11βHSD1, 11βHSD2 and 11βHSD3, an NADP+-dependent dehydrogenase also called the 11βHSD1-like dehydrogenase (11βHSD1L), as well as evidence for yet identified 11βHSDs. Attention is devoted to more recently described aspects of this multi-functional family. The importance of 11βHSD substrates other than glucocorticoids including bile acids, 7-keto sterols, neurosteroids, and xenobiotics is discussed, along with examples of pathology when functions of these multi-tasking enzymes are disrupted. 11βHSDs modulate the intracellular concentration of glucocorticoids, thereby regulating the activation of the glucocorticoid and mineralocorticoid receptors, and 7β-27-hydroxycholesterol, an agonist of the retinoid-related orphan receptor gamma (RORγ). Key functions of this nuclear transcription factor include regulation of immune cell differentiation, cytokine production and inflammation at the cell level. 11βHSD1 expression and/or glucocorticoid reductase activity are inappropriately increased with age and in obesity and metabolic syndrome (MetS). Potential causes for disappointing results of the clinical trials of selective inhibitors of 11βHSD1 in the treatment of these disorders are discussed, as well as the potential for more targeted use of inhibitors of 11βHSD1 and 11βHSD2.
Collapse
Affiliation(s)
| | - Celso E Gomez-Sanchez
- Department of Pharmacology and Toxicology, Jackson, MS, USA; Medicine (Endocrinology), Jackson, MS, USA; University of Mississippi Medical Center and G.V. (Sonny) Montgomery VA Medical Center(3), Jackson, MS, USA
| |
Collapse
|
4
|
Verma M, Sooy K, Just G, Nixon M, Morgan R, Andrew R, Chapman KE, Homer NZ. Quantitative analysis of 11-dehydrocorticosterone and corticosterone for preclinical studies by liquid chromatography/triple quadrupole mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2020; 34 Suppl 4:e8610. [PMID: 31677354 PMCID: PMC7540072 DOI: 10.1002/rcm.8610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/17/2019] [Accepted: 09/23/2019] [Indexed: 06/10/2023]
Abstract
RATIONALE The activity of the glucocorticoid activating enzyme 11β-hydroxysteroid dehydrogenase type-1 (11βHSD1) is altered in diseases such as obesity, inflammation and psychiatric disorders. In rodents 11βHSD1 converts inert 11-dehydrocorticosterone (11-DHC) into the active form, corticosterone (CORT). A sensitive, specific liquid chromatography/tandem mass spectrometry method was sought to simultaneously quantify total 11-DHC and total and free CORT in murine plasma for simple assessment of 11βHSD1 activity in murine models. METHODS Mass spectrometry parameters were optimised and a method for the chromatographic separation of CORT and 11-DHC was developed. Murine plasma was prepared by 10:1 chloroform liquid-liquid extraction (LLE) for analysis. Limits of quantitation (LOQs), linearity and other method criteria were assessed, according to bioanalytical method validation guidelines. RESULTS Reliable separation of 11-DHC and CORT was achieved using an ACE Excel 2 C18-AR (2.1 × 150 mm; 2 μm) fused core column at 25°C, with an acidified water/acetonitrile gradient over 10 min. Analytes were detected by multiple reaction monitoring after positive electrospray ionisation (m/z 345.1.1 ➔ 121.2, m/z 347.1 ➔ 121.1 for 11-DHC and CORT, respectively). The LOQs were 0.25 and 0.20 ng/mL for 11-DHC and CORT, respectively. CONCLUSIONS This LC/MS method is suitable for the reliable analysis of 11-DHC and CORT following simple LLE of murine plasma, bringing preclinical analysis in line with recommendations for clinical endocrinology and biochemistry.
Collapse
Affiliation(s)
- Manu Verma
- University/BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh47 Little France CrescentEdinburghEH16 4TJUK
| | - Karen Sooy
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research InstituteUniversity of Edinburgh47 Little France CrescentEdinburghEH16 4TJUK
| | - George Just
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research InstituteUniversity of Edinburgh47 Little France CrescentEdinburghEH16 4TJUK
| | - Mark Nixon
- University/BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh47 Little France CrescentEdinburghEH16 4TJUK
| | - Ruth Morgan
- University/BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh47 Little France CrescentEdinburghEH16 4TJUK
| | - Ruth Andrew
- University/BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh47 Little France CrescentEdinburghEH16 4TJUK
| | - Karen E. Chapman
- University/BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh47 Little France CrescentEdinburghEH16 4TJUK
| | - Natalie Z.M. Homer
- University/BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh47 Little France CrescentEdinburghEH16 4TJUK
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research InstituteUniversity of Edinburgh47 Little France CrescentEdinburghEH16 4TJUK
| |
Collapse
|
5
|
Ryu JH, Lee JA, Kim S, Shin YA, Yang J, Han HY, Son HJ, Kim YH, Sa JH, Kim JS, Lee J, Lee J, Park HG. Discovery of 2-((R)-4-(2-Fluoro-4-(methylsulfonyl)phenyl)-2-methylpiperazin-1-yl)-N-((1R,2s,3S,5S,7S)-5-hydroxyadamantan-2-yl)pyrimidine-4-carboxamide (SKI2852): A Highly Potent, Selective, and Orally Bioavailable Inhibitor of 11β-Hydroxysteroid Dehydrogenase Type 1 (11β-HSD1). J Med Chem 2016; 59:10176-10189. [DOI: 10.1021/acs.jmedchem.6b01122] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Je Ho Ryu
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
- Research
Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, San 56-1, Shillim-Dong, Kwanak-Gu, Seoul 151-742, Korea
| | - Jung A Lee
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
| | - Shinae Kim
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
| | - Young Ah Shin
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
| | - Jewon Yang
- Research
Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, San 56-1, Shillim-Dong, Kwanak-Gu, Seoul 151-742, Korea
| | - Hye Young Han
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
| | - Hyun Joo Son
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
| | - Yong Hyuk Kim
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
| | - Joon Ho Sa
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
| | - Jae-Sun Kim
- Life Science R&D Center, SK Chemicals, Seongnam-Si, Bundang-Gu, Sampyeong-Dong 686, Gyeonggi-Do 463-400, Korea
| | - Jungeun Lee
- Research
Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, San 56-1, Shillim-Dong, Kwanak-Gu, Seoul 151-742, Korea
| | - Jeeyeon Lee
- Research
Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, San 56-1, Shillim-Dong, Kwanak-Gu, Seoul 151-742, Korea
| | - Hyeung-geun Park
- Research
Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, San 56-1, Shillim-Dong, Kwanak-Gu, Seoul 151-742, Korea
| |
Collapse
|
6
|
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
|
7
|
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]
|
8
|
Gomez-Sanchez EP. Brain mineralocorticoid receptors in cognition and cardiovascular homeostasis. Steroids 2014; 91:20-31. [PMID: 25173821 PMCID: PMC4302001 DOI: 10.1016/j.steroids.2014.08.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 07/10/2014] [Accepted: 08/04/2014] [Indexed: 12/20/2022]
Abstract
Mineralocorticoid receptors (MR) mediate diverse functions supporting osmotic and hemodynamic homeostasis, response to injury and inflammation, and neuronal changes required for learning and memory. Inappropriate MR activation in kidneys, heart, vessels, and brain hemodynamic control centers results in cardiovascular and renal pathology and hypertension. MR binds aldosterone, cortisol and corticosterone with similar affinity, while the glucocorticoid receptor (GR) has less affinity for cortisol and corticosterone. As glucocorticoids are more abundant than aldosterone, aldosterone activates MR in cells co-expressing enzymes with 11β-hydroxydehydrogenase activity to inactivate them. MR and GR co-expressed in the same cell interact at the molecular and functional level and these functions may be complementary or opposing depending on the cell type. Thus the balance between MR and GR expression and activation is crucial for normal function. Where 11β-hydroxydehydrogenase 2 (11β-HSD2) that inactivates cortisol and corticosterone in aldosterone target cells of the kidney and nucleus tractus solitarius (NTS) is not expressed, as in most neurons, MR are activated at basal glucocorticoid concentrations, GR at stress concentrations. An exception may be pre-autonomic neurons of the PVN which express MR and 11β-HSD1 in the absence of hexose-6-phosphate dehydrogenase required to generate the requisite cofactor for reductase activity, thus it acts as a dehydrogenase. MR antagonists, valuable adjuncts to the treatment of cardiovascular disease, also inhibit MR in the brain that are crucial for memory formation and exacerbate detrimental effects of excessive GR activation on cognition and mood. 11β-HSD1 inhibitors combat metabolic and cognitive diseases related to glucocorticoid excess, but may exacerbate MR action where 11β-HSD1 acts as a dehydrogenase, while non-selective 11β-HSD1&2 inhibitors cause injurious disruption of MR hemodynamic control. MR functions in the brain are multifaceted and optimal MR:GR activity is crucial. Therefore selectively targeting down-stream effectors of MR specific actions may be a better therapeutic goal.
Collapse
Affiliation(s)
- Elise P Gomez-Sanchez
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| |
Collapse
|
9
|
Abstract
The primary adrenal cortical steroid hormones, aldosterone, and the glucocorticoids cortisol and corticosterone, act through the structurally similar mineralocorticoid (MR) and glucocorticoid receptors (GRs). Aldosterone is crucial for fluid, electrolyte, and hemodynamic homeostasis and tissue repair; the significantly more abundant glucocorticoids are indispensable for energy homeostasis, appropriate responses to stress, and limiting inflammation. Steroid receptors initiate gene transcription for proteins that effect their actions as well as rapid non-genomic effects through classical cell signaling pathways. GR and MR are expressed in many tissues types, often in the same cells, where they interact at molecular and functional levels, at times in synergy, others in opposition. Thus the appropriate balance of MR and GR activation is crucial for homeostasis. MR has the same binding affinity for aldosterone, cortisol, and corticosterone. Glucocorticoids activate MR in most tissues at basal levels and GR at stress levels. Inactivation of cortisol and corticosterone by 11β-HSD2 allows aldosterone to activate MR within aldosterone target cells and limits activation of the GR. Under most conditions, 11β-HSD1 acts as a reductase and activates cortisol/corticosterone, amplifying circulating levels. 11β-HSD1 and MR antagonists mitigate inappropriate activation of MR under conditions of oxidative stress that contributes to the pathophysiology of the cardiometabolic syndrome; however, MR antagonists decrease normal MR/GR functional interactions, a particular concern for neurons mediating cognition, memory, and affect.
Collapse
Affiliation(s)
- Elise Gomez-Sanchez
- G.V.(Sonny) Montgomery V.A. Medical Center and Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Celso E. Gomez-Sanchez
- G.V.(Sonny) Montgomery V.A. Medical Center and Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
10
|
Deng X, Shen Y, Yang J, He J, Zhao Y, Peng LY, Leng Y, Zhao QS. Discovery and structure–activity relationships of ent-Kaurene diterpenoids as potent and selective 11β-HSD1 inhibitors: Potential impact in diabetes. Eur J Med Chem 2013; 65:403-14. [DOI: 10.1016/j.ejmech.2013.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 05/08/2013] [Accepted: 05/09/2013] [Indexed: 01/22/2023]
|
11
|
Sawada T, Nakada M. Enantioselective Total Synthesis of (+)-Colletoic Acid via Catalytic Asymmetric Intramolecular Cyclopropanation of an α-Diazo-β-keto Diphenylphosphine Oxide. Org Lett 2013; 15:1004-7. [DOI: 10.1021/ol303459x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Takashi Sawada
- Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Masahisa Nakada
- Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku-ku, Tokyo 169-8555, Japan
| |
Collapse
|
12
|
Wan ZK, Chenail E, Li HQ, Ipek M, Xiang J, Suri V, Hahm S, Bard J, Svenson K, Xu X, Tian X, Wang M, Li X, Johnson CE, Qadri A, Panza D, Perreault M, Mansour TS, Tobin JF, Saiah E. Discovery of HSD-621 as a Potential Agent for the Treatment of Type 2 Diabetes. ACS Med Chem Lett 2013; 4:118-23. [PMID: 24900572 DOI: 10.1021/ml300352x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 11/23/2012] [Indexed: 12/20/2022] Open
Abstract
11β-Hydroxysteroid dehydrogenase type 1 (11β-HSD1) catalyzes the conversion of inactive glucocorticoid cortisone to its active form, cortisol. The glucocorticoid receptor (GR) signaling pathway has been linked to the pathophysiology of diabetes and metabolic syndrome. Herein, the structure-activity relationship of a series of piperazine sulfonamide-based 11β-HSD1 inhibitors is described. (R)-3,3,3-Trifluoro-2-(5-(((R)-4-(4-fluoro-2-(trifluoromethyl)phenyl)-2-methylpiperazin-1-yl)sulfonyl)thiophen-2-yl)-2-hydroxypropanamide 18a (HSD-621) was identified as a potent and selective 11β-HSD1 inhibitor and was ultimately selected as a clinical development candidate. HSD-621 has an attractive overall pharmaceutical profile and demonstrates good oral bioavailability in mouse, rat, and dog. When orally dosed in C57/BL6 diet-induced obesity (DIO) mice, HSD-621 was efficacious and showed a significant reduction in both fed and fasting glucose and insulin levels. Furthermore, HSD-621 was well tolerated in drug safety assessment studies.
Collapse
Affiliation(s)
- Zhao-Kui Wan
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Eva Chenail
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Huan-Qiu Li
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Manus Ipek
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Jason Xiang
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Vipin Suri
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Seung Hahm
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Joel Bard
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Kristine Svenson
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Xin Xu
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Xianbin Tian
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Mengmeng Wang
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Xiangping Li
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Christian E. Johnson
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Ariful Qadri
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Darrell Panza
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Mylene Perreault
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Tarek S. Mansour
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - James F. Tobin
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| | - Eddine Saiah
- Worldwide
Research and Development, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, Massachusetts
02140, United States
| |
Collapse
|
13
|
Véniant MM, Hale C, Hungate RW, Gahm K, Emery MG, Jona J, Joseph S, Adams J, Hague A, Moniz G, Zhang J, Bartberger MD, Li V, Syed R, Jordan S, Komorowski R, Chen MM, Cupples R, Kim KW, St. Jean DJ, Johansson L, Henriksson MA, Williams M, Vallgårda J, Fotsch C, Wang M. Discovery of a Potent, Orally Active 11β-Hydroxysteroid Dehydrogenase Type 1 Inhibitor for Clinical Study: Identification of (S)-2-((1S,2S,4R)-Bicyclo[2.2.1]heptan-2-ylamino)-5-isopropyl-5-methylthiazol-4(5H)-one (AMG 221). J Med Chem 2010; 53:4481-7. [DOI: 10.1021/jm100242d] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
14
|
Hult M, Ortsäter H, Schuster G, Graedler F, Beckers J, Adamski J, Ploner A, Jörnvall H, Bergsten P, Oppermann U. Short-term glucocorticoid treatment increases insulin secretion in islets derived from lean mice through multiple pathways and mechanisms. Mol Cell Endocrinol 2009; 301:109-16. [PMID: 18984029 DOI: 10.1016/j.mce.2008.09.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2008] [Revised: 09/30/2008] [Accepted: 09/30/2008] [Indexed: 11/15/2022]
Abstract
Chronic exposure to elevated levels of glucocorticoids leads to metabolic dysfunctions with hyperglycemia and insulin resistance. Long-term treatment with glucocorticoids induces severe impairment of glucose-stimulated insulin secretion. We analyzed the effects of short-, and medium-term (2-120h) treatment with 50-200nM glucocorticoids on primary pancreatic islet cultures derived from lean C57BL/6J mice. In contrast to animal models of insulin resistance, beta-cells from lean mice respond with an increased glucose-stimulated insulin secretion, with a peak effect around 18-24h of treatment. Analyses of the insulin secretion response reveal that early and late phase responses are dissociated upon glucocorticoid treatment. Whereas late phase responses return to basal levels after long treatment, early phase responses remain increased over several days. Increased insulin secretion is also obtained by incubation with the inactive glucocorticoid dehydrocorticosterone, pointing to an important role of the enzyme 11beta-hydroxysteroid dehydrogenase type 1 in mediating glucocorticoid effects in beta-cells. Transcript profiling revealed differential regulation of genes involved in mediation of signal transduction, insulin secretion, stress and inflammatory responses. The results show that short- to medium-term glucocorticoid treatment of pancreatic islets derived from lean mice leads to an increased insulin release and may constitute an important parameter in changing towards a pro-diabetic phenotype.
Collapse
Affiliation(s)
- Malin Hult
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Blum A, Favia AD, Maser E. 11beta-Hydroxysteroid dehydrogenase type 1 inhibitors with oleanan and ursan scaffolds. Mol Cell Endocrinol 2009; 301:132-6. [PMID: 18822345 DOI: 10.1016/j.mce.2008.08.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2008] [Revised: 08/15/2008] [Accepted: 08/25/2008] [Indexed: 11/26/2022]
Abstract
The enzyme 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) converts cortisone to the active glucocorticoid cortisol, thereby acting as a cellular switch to mediate glucocorticoid action in many tissues. Several studies have indicated that 11beta-HSD1 plays a crucial role in the onset of type 2 diabetes and central obesity. As a consequence, selective inhibition of 11beta-HSD1 in humans might become a new and promising approach for lowering blood glucose concentrations and for counteracting the accumulation of visceral fat and its related metabolic abnormalities in type 2 diabetes. In this study, we present the synthesis and the biological evaluation of ursan or oleanan type triterpenoids which may act as selective 11beta-HSD1 inhibitors in liver as well as in peripheral tissues, like adipocytes and muscle cells. In order to rationalise the outcomes of the inhibition data, docking simulations of the ligands were performed on the experimentally determined structure of 11beta-HSD1. Furthermore, we discuss the structural determinants that confer enzymatic specificity. From our investigation, valuable information has been obtained to design selective 11beta-HSD1 blockers based on the oleanan and ursan scaffold.
Collapse
Affiliation(s)
- Andreas Blum
- Institute of Toxicology and Pharmacology for Natural Scientists, University Medical School Schleswig-Holstein, Kiel, Germany
| | | | | |
Collapse
|
16
|
Véniant MM, Hale C, Komorowski R, Chen MM, St Jean DJ, Fotsch C, Wang M. Time of the day for 11beta-HSD1 inhibition plays a role in improving glucose homeostasis in DIO mice. Diabetes Obes Metab 2009; 11:109-17. [PMID: 18479468 DOI: 10.1111/j.1463-1326.2008.00911.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIMS The physiological effects of glucocorticoids in a given tissue are driven by the local level of the active glucocorticoid, which is determined by two sources: the plasma cortisol in human (or corticosterone in rodents) and the cortisol produced locally through 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) activity. Because of the circadian variation of plasma glucocorticoids, the pharmacological efficacy of 11beta-HSD1 inhibition may depend on the time of the day for inhibitor administration. METHODS The circadian profile of corticosterone was established in lean and diet-induced obesity (DIO) C57BL/6 mice from blood collected at different time of the day. 11beta-HSD1 enzyme activity was also measured throughout the day in DIO mice. To determine the optimal timing for administration of an 11beta-HSD1 inhibitor to obtain maximum efficacy, we used a DIO mouse model and a small molecule inhibitor of 11beta-HSD1 from our thiazolinone series. Based on the circadian profile of corticosterone obtained, we administered the 11beta-HSD1 inhibitor to these animals at different times of the day and evaluated the effects on plasma glucose levels and glucose tolerance. RESULTS We report that corticosterone circadian rhythm was similar between lean and DIO C57BL/6 mice, and 11beta-HSD1 enzyme activity undergoes minimal variations throughout the day. Interestingly, the compound exhibited maximum efficacy if dosed in the afternoon when plasma corticosterone is high; the morning dosing when plasma corticosterone is low did not lead to efficacy. CONCLUSION These data suggest that because of the circadian rhythm of circulating glucocorticoids, the time of the day for 11beta-HSD1 inhibitor administration is important in achieving efficacy.
Collapse
Affiliation(s)
- M M Véniant
- Department of Metabolic Disorders, Amgen Inc., Thousand Oaks, CA 91320, USA.
| | | | | | | | | | | | | |
Collapse
|
17
|
Heinig K, Wirz T, Bucheli F. Determination of Glucocorticoids in Animal Plasma, Urine and Tissues by Column-Switching LC–Tandem-MS. Chromatographia 2008. [DOI: 10.1365/s10337-008-0776-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
18
|
Distinctive molecular inhibition mechanisms for selective inhibitors of human 11beta-hydroxysteroid dehydrogenase type 1. Bioorg Med Chem 2008; 16:8922-31. [PMID: 18789704 DOI: 10.1016/j.bmc.2008.08.065] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Revised: 08/20/2008] [Accepted: 08/26/2008] [Indexed: 11/22/2022]
Abstract
11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) catalyzes the NADPH dependent interconversion of inactive cortisone to active cortisol. Excess 11beta-HSD1 or cortisol leads to insulin resistance and metabolic syndrome in animal models and in humans. Inhibiting 11beta-HSD1 activity signifies a promising therapeutic strategy in the treatment of Type 2 diabetes and related diseases. Herein, we report two highly potent and selective small molecule inhibitors of human 11beta-HSD1. While compound 1, a sulfonamide, functions as a simple substrate competitive inhibitor, compound 2, a triazole, shows the kinetic profile of a mixed inhibitor. Co-crystal structures reveal that both compounds occupy the 11beta-HSD1 catalytic site, but present distinct molecular interactions with the protein. Strikingly, compound 2 interacts much closer to the cofactor NADP+ and likely modifies its binding. Together, the structural and kinetic analyses demonstrate two distinctive molecular inhibition mechanisms, providing valuable information for future inhibitor design.
Collapse
|
19
|
Johansson L, Fotsch C, Bartberger MD, Castro VM, Chen M, Emery M, Gustafsson S, Hale C, Hickman D, Homan E, Jordan SR, Komorowski R, Li A, McRae K, Moniz G, Matsumoto G, Orihuela C, Palm G, Veniant M, Wang M, Williams M, Zhang J. 2-Amino-1,3-thiazol-4(5H)-ones as Potent and Selective 11β-Hydroxysteroid Dehydrogenase Type 1 Inhibitors: Enzyme−Ligand Co-Crystal Structure and Demonstration of Pharmacodynamic Effects in C57Bl/6 Mice. J Med Chem 2008; 51:2933-43. [DOI: 10.1021/jm701551j] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Lars Johansson
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Christopher Fotsch
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Michael D. Bartberger
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Victor M. Castro
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Michelle Chen
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Maurice Emery
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Sonja Gustafsson
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Clarence Hale
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Dean Hickman
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Evert Homan
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Steven R. Jordan
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Renee Komorowski
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Aiwen Li
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Kenneth McRae
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - George Moniz
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Guy Matsumoto
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Carlos Orihuela
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Gunnar Palm
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Murielle Veniant
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Minghan Wang
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Meredith Williams
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| | - Jiandong Zhang
- Biovitrum AB, SE-112 76 Stockholm, Sweden, and Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320
| |
Collapse
|
20
|
Aster SD, Graham DW, Kharbanda D, Patel G, Ponpipom M, Santorelli GM, Szymonifka MJ, Mundt SS, Shah K, Springer MS, Thieringer R, Hermanowski-Vosatka A, Wright SD, Xiao J, Zokian H, Balkovec JM. Bis-aryl triazoles as selective inhibitors of 11beta-hydroxysteroid dehydrogenase type 1. Bioorg Med Chem Lett 2008; 18:2799-804. [PMID: 18434143 DOI: 10.1016/j.bmcl.2008.04.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2008] [Revised: 04/02/2008] [Accepted: 04/02/2008] [Indexed: 10/22/2022]
Abstract
3-Aryl-5-phenyl-(1,2,4)-triazoles were identified as selective inhibitors of 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1). They are active in both in vitro and an in vivo mouse pharmacodynamic (PD) model. The synthesis and structure activity relationships are presented.
Collapse
Affiliation(s)
- Susan D Aster
- Department of Medicinal Chemistry, Merck & Co., Inc., PO Box 2000, Rahway, NJ 07065, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Lee JH, Kang NS, Yoo SE. Docking-based 3D-QSAR study for 11β-HSD1 inhibitors. Bioorg Med Chem Lett 2008; 18:2479-90. [DOI: 10.1016/j.bmcl.2008.02.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2007] [Revised: 01/21/2008] [Accepted: 02/14/2008] [Indexed: 12/01/2022]
|
22
|
Myers M, Lamont MC, van den Driesche S, Mary N, Thong KJ, Hillier SG, Duncan WC. Role of luteal glucocorticoid metabolism during maternal recognition of pregnancy in women. Endocrinology 2007; 148:5769-79. [PMID: 17872369 DOI: 10.1210/en.2007-0742] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The human corpus luteum (hCL) is an active, transient, and dynamic endocrine gland. It will experience extensive tissue and vascular remodeling followed by 1) demise of the whole gland without any apparent scarring or 2) maintenance of structural and functional integrity dependent on conceptus-derived human chorionic gonadotropin (hCG). Because cortisol has well-characterized roles in tissue remodeling and repair, we hypothesized that it may have a role in controlling luteal dissolution during luteolysis and would be locally produced toward the end of the luteal cycle. Glucocorticoid-metabolizing enzymes [11beta-hydroxysteroid dehydrogenase (11betaHSD) types 1 and 2] and the glucocorticoid receptor (GR) were assessed in hCL and cultures of luteinized granulosa cells (LGC) using immunofluorescence and quantitative RT-PCR. Furthermore, the effect of cortisol on steroidogenic cell survival and fibroblast-like cell activity was explored in vitro. The hCL expressed 11betaHSD isoenzymes in LGC and nuclear GR in several cell types. hCG up-regulated the expression and activity of 11betaHSD type 1 (P < 0.05) and down-regulated type 2 enzyme (P < 0.05) in vitro and tended to do the same in vivo. Cortisol increased the survival of LGC treated with RU486 (P < 0.05) and suppressed the activity of a proteolytic enzyme associated with luteolysis in fibroblast-like cells (P < 0.05). Our results suggest that, rather than during luteolysis, it is luteal rescue with hCG that is associated with increased local cortisol generation by 11betaHSD type 1. Locally generated cortisol may therefore act on the hCL through GR to have a luteotropic role in the regulation of luteal tissue remodeling during maternal recognition of pregnancy.
Collapse
Affiliation(s)
- Michelle Myers
- Obstetrics and Gynaecology, The Queen's Medical Research Institute Centre for Reproductive Biology, 47 Little France Crescent, Edinburgh, Scotland, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
23
|
The scientific impact of the Structural Genomics Consortium: a protein family and ligand-centered approach to medically-relevant human proteins. ACTA ACUST UNITED AC 2007; 8:107-19. [PMID: 17932789 PMCID: PMC2140095 DOI: 10.1007/s10969-007-9027-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2007] [Accepted: 09/22/2007] [Indexed: 11/04/2022]
Abstract
As many of the structural genomics centers have ended their first phase of operation, it is a good point to evaluate the scientific impact of this endeavour. The Structural Genomics Consortium (SGC), operating from three centers across the Atlantic, investigates human proteins involved in disease processes and proteins from Plasmodium falciparum and related organisms. We present here some of the scientific output of the Oxford node of the SGC, where the target areas include protein kinases, phosphatases, oxidoreductases and other metabolic enzymes, as well as signal transduction proteins. The SGC has aimed to achieve extensive coverage of human gene families with a focus on protein–ligand interactions. The methods employed for effective protein expression, crystallization and structure determination by X-ray crystallography are summarized. In addition to the cumulative impact of accelerated delivery of protein structures, we demonstrate how family coverage, generic screening methodology, and the availability of abundant purified protein samples, allow a level of discovery that is difficult to achieve otherwise. The contribution of NMR to structure determination and protein characterization is discussed. To make this information available to a wide scientific audience, a new tool for disseminating annotated structural information was created that also represents an interactive platform allowing for a continuous update of the annotation by the scientific community.
Collapse
|
24
|
Sutin L, Andersson S, Bergquist L, Castro VM, Danielsson E, James S, Henriksson M, Johansson L, Kaiser C, Flyrén K, Williams M. Oxazolones as potent inhibitors of 11β-hydroxysteroid dehydrogenase type 1. Bioorg Med Chem Lett 2007; 17:4837-40. [PMID: 17600707 DOI: 10.1016/j.bmcl.2007.06.054] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2007] [Revised: 06/13/2007] [Accepted: 06/13/2007] [Indexed: 11/15/2022]
Abstract
2,5,5-Trisubstituted oxazolones were identified as potent inhibitors of 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1). The synthesis, structure-activity relationship and metabolic stability of these compounds are presented.
Collapse
Affiliation(s)
- Lori Sutin
- Biovitrum AB, SE-112 76, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Flyrén K, Bergquist LO, Castro VM, Fotsch C, Johansson L, St Jean DJ, Sutin L, Williams M. Piperidine amides as 11beta-hydroxysteroid dehydrogenase type 1 inhibitors. Bioorg Med Chem Lett 2007; 17:3421-5. [PMID: 17442567 DOI: 10.1016/j.bmcl.2007.03.083] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Revised: 03/27/2007] [Accepted: 03/27/2007] [Indexed: 10/23/2022]
Abstract
A series of piperidine amide inhibitors of human 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) were identified via modifications of the HTS hit compound 1. The synthesis, in vitro biological evaluation, and structure-activity relationship of these compounds are presented.
Collapse
|
26
|
Wu X, Lukacik P, Kavanagh KL, Oppermann U. SDR-type human hydroxysteroid dehydrogenases involved in steroid hormone activation. Mol Cell Endocrinol 2007; 265-266:71-6. [PMID: 17234335 DOI: 10.1016/j.mce.2006.12.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Hydroxysteroid dehydrogenases catalyze the NAD(P)(H)-dependent oxidoreduction of hydroxyl and oxo-functions at distinct positions of steroid hormones. This reversible reaction constitutes an important pre-receptor control mechanism for nuclear receptor ligands of the androgen, estrogen and glucocorticoid classes, since the conversion "switches" between receptor ligands and their inactive metabolites. The major reversible activities found in mammals acting on steroid hormones comprise 3alpha-, 11beta- and 17beta-hydroxysteroid dehydrogenases, and for each group several distinct isozymes have been described. The enzymes differ in their expression pattern, nucleotide cofactor preference, steroid substrate specificity and subcellular localization, and thus constitute a complex system ensuring cell-specific adaptation and regulation of steroid hormone levels. Several isoforms constitute promising drug targets, of particular importance in cancer, metabolic diseases, neurodegeneration and immunity.
Collapse
Affiliation(s)
- Xiaoqiu Wu
- Structural Genomics Consortium, University of Oxford, Oxford OX3 7LD, United Kingdom
| | | | | | | |
Collapse
|
27
|
Drolet B, Simard C, Poirier P. Impact of weight-loss medications on the cardiovascular system: focus on current and future anti-obesity drugs. Am J Cardiovasc Drugs 2007; 7:273-88. [PMID: 17696568 DOI: 10.2165/00129784-200707040-00005] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Overweight and obesity have been rising dramatically worldwide and are associated with numerous co-morbidities such as cardiovascular disease (CVD), type 2 diabetes mellitus, hypertension, certain cancers, and sleep apnea. In fact, obesity is an independent risk factor for CVD and CVD risks have also been documented in obese children. The majority of overweight and obese patients who achieve a significant short-term weight loss do not maintain their lower bodyweight in the long term. This may be due to a lack of intensive counseling and support from a facilitating environment including dedicated healthcare professionals such as nutritionists, kinesiologists, and behavior specialists. As a result, there has been a considerable focus on the role of adjunctive therapy such as pharmacotherapy for long-term weight loss and weight maintenance. Beyond an unfavorable risk factor profile, overweight and obesity also impact upon heart structure and function. Since the beginning, the quest for weight loss drugs has encountered warnings from regulatory agencies and the withdrawal from the market of efficient but unsafe medications. Fenfluramine was withdrawn from the market because of unacceptable pulmonary and cardiac adverse effects. Nevertheless, there is extensive research directed at the development of new anti-obesity compounds. The effect of these molecules on CVD risk factors has been studied and reported but information regarding their impact on the cardiovascular system is sparse. Thus, instead of looking at the benefit of weight loss on metabolism and risk factor management, this article discusses the impact of weight loss medications on the cardiovascular system. The potential interaction of available and potential new weight loss drugs with heart function and structure is reviewed.
Collapse
Affiliation(s)
- Benoit Drolet
- Institut Universitaire de Cardiologie et de Pneumologie, Laval Hospital, Quebec City, Quebec, Canada
| | | | | |
Collapse
|
28
|
Witchel SF, DeFranco DB. Mechanisms of Disease: regulation of glucocorticoid and receptor levels—impact on the metabolic syndrome. ACTA ACUST UNITED AC 2006; 2:621-31. [PMID: 17082809 DOI: 10.1038/ncpendmet0323] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2006] [Accepted: 06/12/2006] [Indexed: 12/18/2022]
Abstract
Glucocorticoids exert their effects in target tissues predominantly through their interaction with the glucocorticoid receptor, a member of the nuclear receptor superfamily of transcription factors. Over the years many studies have linked hormone responsiveness, both in vitro and in vivo, to the levels of both glucocorticoid and glucocorticoid receptor; furthermore, an impact of glucocorticoid receptor subcellular trafficking on hormone response has been revealed. This review will focus on the molecular mechanisms responsible for the regulation of glucocorticoid receptor trafficking and expression, and will highlight work that revealed selective physiological effects of altered glucocorticoid receptor expression. The role of alterations in glucocorticoid levels and glucocorticoid receptor function in the metabolic syndrome will also be discussed.
Collapse
Affiliation(s)
- Selma F Witchel
- Division of Pediatric Endocrinology, Children's Hospital of Pittsburgh, and University of Pittsburgh School of Medicine, Pittsburg, PA 15261, USA
| | | |
Collapse
|
29
|
Sorensen B, Winn M, Rohde J, Shuai Q, Wang J, Fung S, Monzon K, Chiou W, Stolarik D, Imade H, Pan L, Deng X, Chovan L, Longenecker K, Judge R, Qin W, Brune M, Camp H, Frevert EU, Jacobson P, Link JT. Adamantane sulfone and sulfonamide 11-beta-HSD1 Inhibitors. Bioorg Med Chem Lett 2006; 17:527-32. [PMID: 17070044 DOI: 10.1016/j.bmcl.2006.10.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Revised: 10/03/2006] [Accepted: 10/05/2006] [Indexed: 10/24/2022]
Abstract
Potent and selective adamantane sulfone and sulfonamide inhibitors of 11-beta-HSD-1 have been discovered. Selected compounds from these series have robust pharmacokinetic profiles and strongly inhibit liver, fat, and brain HSD1 for extended periods after oral dosing.
Collapse
Affiliation(s)
- Bryan Sorensen
- Abbott, Dept. 47F, Bldg. AP-10, Rm. 301, 100 Abbott Park Road, Abbott Park, IL 60064-6098, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Sorensen B, Rohde J, Wang J, Fung S, Monzon K, Chiou W, Pan L, Deng X, Stolarik D, Frevert EU, Jacobson P, Link JT. Adamantane 11-beta-HSD-1 inhibitors: Application of an isocyanide multicomponent reaction. Bioorg Med Chem Lett 2006; 16:5958-62. [PMID: 16996734 DOI: 10.1016/j.bmcl.2006.08.129] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2006] [Revised: 08/30/2006] [Accepted: 08/31/2006] [Indexed: 01/06/2023]
Abstract
A series of potent and selective adamantane aminoamide 11-beta-HSD-1 inhibitors has been optimized. Chemically these studies were expedited by utilizing readily obtained amino acids as starting materials or an isocyanide multicomponent reaction. Structure-activity relationship studies resulted in the discovery of dual human and mouse 11-beta-HSD-1 potent and selective inhibitors like adamantane 11 and related compounds with high metabolic stability and robust pharmacokinetic profiles.
Collapse
Affiliation(s)
- Bryan Sorensen
- Metabolic Disease Research, Abbott, 100 Abbott Park Road, Abbott Park, IL 60064, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Hult M, Shafqat N, Elleby B, Mitschke D, Svensson S, Forsgren M, Barf T, Vallgårda J, Abrahmsen L, Oppermann U. Active site variability of type 1 11beta-hydroxysteroid dehydrogenase revealed by selective inhibitors and cross-species comparisons. Mol Cell Endocrinol 2006; 248:26-33. [PMID: 16431016 DOI: 10.1016/j.mce.2005.11.043] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The NADPH-dependent enzyme type 1 11beta-hydroxysteroid dehydrogenase (11beta-HSD1) activates in a tissue-specific manner circulating pro-glucocorticoid hormones (cortisone in humans) to the 11beta-OH ligand (cortisol in humans), which is able to bind to its cognate receptor and regulate gene transcription. Modulation of this pre-receptor activation mechanism by selective enzyme inhibitors is a desirable goal in the treatment of insulin resistance and related metabolic disorders. Like most other hydroxysteroid dehydrogenases 11beta-HSD1 belongs to the evolutionarily conserved enzyme superfamily of short-chain dehydrogenases/reductases (SDR). The enzyme is anchored within the endoplasmic reticulum through an N-terminal transmembrane domain. In this study we aimed to characterize the active site of mammalian 11beta-HSD1 by determining primary structures from several mammalian lines (cat, hamster, cynomolgus, chimpanzee, dog) thus increasing substantially available sequence information, and allowing us to determine highly variable and constant parts within the primary structure. These regions were mapped to the recently determined three-dimensional structure and are mostly found around the substrate binding site. Furthermore we performed inhibition studies by using different series of inhibitors, comprising 11beta-HSD1 selective arylsulfonamidothiazoles and the unselective steroid-based compound carbenoxolone. The different arylsulfonamidothiazoles display distinct inhibition profiles versus the mammalian species tested, with several tight binding inhibitors for the human enzyme (Ki approximately 50 nM), intermediate for mouse, and weak or not binding inhibitors for rat and guinea pig (Ki>3 microM). Analysis of the inhibition mode reveals that the tight binding inhibitor BVT.528 is a competitive inhibitor for the human form, whereas the related compound BVT.2733 displays a mixed-type inhibition pattern versus the mouse enzyme. Taken together, this structure-activity study provides increased insight into active site complexity and catalytic mechanism of 11beta-HSD1, useful for further inhibitor design.
Collapse
Affiliation(s)
- Malin Hult
- Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
11β-Hydroxysteroid Dehydrogenase Type 1 Inhibitors. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2006. [DOI: 10.1016/s0065-7743(06)41007-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|
33
|
Koch MA, Waldmann H. Protein structure similarity clustering and natural product structure as guiding principles for chemical genomics. ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2006:89-109. [PMID: 16709001 DOI: 10.1007/978-3-540-37635-4_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The majority of all proteins are modularly built from a limited set of approximately 1,000 structural domains. The knowledge of a common protein fold topology in the ligand-sensing cores of protein domains can be exploited for the design of small-molecule libraries in the development of inhibitors and ligands. Thus, a novel strategy of clustering protein domain cores based exclusively on structure similarity considerations (protein structure similarity clustering, PSSC) has been successfully applied to the development of small-molecule inhibitors of acetylcholinesterase and the 11beta-hydroxysteroid dehydrogenases based on the structure of a naturally occurring Cdc25 inhibitor. The efficiency of making use of the scaffolds of natural products as biologically prevalidated starting points for the design of compound libraries is further highlighted by the development of benzopyran-based FXR ligands.
Collapse
Affiliation(s)
- M A Koch
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Dortmund, Germany
| | | |
Collapse
|
34
|
Hermanowski-Vosatka A, Balkovec JM, Cheng K, Chen HY, Hernandez M, Koo GC, Le Grand CB, Li Z, Metzger JM, Mundt SS, Noonan H, Nunes CN, Olson SH, Pikounis B, Ren N, Robertson N, Schaeffer JM, Shah K, Springer MS, Strack AM, Strowski M, Wu K, Wu T, Xiao J, Zhang BB, Wright SD, Thieringer R. 11beta-HSD1 inhibition ameliorates metabolic syndrome and prevents progression of atherosclerosis in mice. ACTA ACUST UNITED AC 2005; 202:517-27. [PMID: 16103409 PMCID: PMC2212859 DOI: 10.1084/jem.20050119] [Citation(s) in RCA: 302] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The enzyme 11β–hydroxysteroid dehydrogenase (HSD) type 1 converts inactive cortisone into active cortisol in cells, thereby raising the effective glucocorticoid (GC) tone above serum levels. We report that pharmacologic inhibition of 11β-HSD1 has a therapeutic effect in mouse models of metabolic syndrome. Administration of a selective, potent 11β-HSD1 inhibitor lowered body weight, insulin, fasting glucose, triglycerides, and cholesterol in diet-induced obese mice and lowered fasting glucose, insulin, glucagon, triglycerides, and free fatty acids, as well as improved glucose tolerance, in a mouse model of type 2 diabetes. Most importantly, inhibition of 11β-HSD1 slowed plaque progression in a murine model of atherosclerosis, the key clinical sequela of metabolic syndrome. Mice with a targeted deletion of apolipoprotein E exhibited 84% less accumulation of aortic total cholesterol, as well as lower serum cholesterol and triglycerides, when treated with an 11β-HSD1 inhibitor. These data provide the first evidence that pharmacologic inhibition of intracellular GC activation can effectively treat atherosclerosis, the key clinical consequence of metabolic syndrome, in addition to its salutary effect on multiple aspects of the metabolic syndrome itself.
Collapse
|
35
|
Olson S, Aster SD, Brown K, Carbin L, Graham DW, Hermanowski-Vosatka A, LeGrand CB, Mundt SS, Robbins MA, Schaeffer JM, Slossberg LH, Szymonifka MJ, Thieringer R, Wright SD, Balkovec JM. Adamantyl triazoles as selective inhibitors of 11β-hydroxysteroid dehydrogenase type 1. Bioorg Med Chem Lett 2005; 15:4359-62. [PMID: 16039856 DOI: 10.1016/j.bmcl.2005.06.040] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2005] [Revised: 06/08/2005] [Accepted: 06/10/2005] [Indexed: 10/25/2022]
Abstract
Adamantyl triazoles were identified as selective inhibitors of 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1). They are active both in in vitro and in in vivo pharmacodynamic models. The synthesis and structure-activity relationships of these inhibitors are presented.
Collapse
Affiliation(s)
- Steven Olson
- Department of Medicinal Chemistry, Merck Research Laboratories, PO Box 2000, Rahway, NJ 07065, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Small GR, Hadoke PWF, Sharif I, Dover AR, Armour D, Kenyon CJ, Gray GA, Walker BR. Preventing local regeneration of glucocorticoids by 11beta-hydroxysteroid dehydrogenase type 1 enhances angiogenesis. Proc Natl Acad Sci U S A 2005; 102:12165-70. [PMID: 16093320 PMCID: PMC1189304 DOI: 10.1073/pnas.0500641102] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Angiogenesis restores blood flow to healing tissues, a process that is inhibited by high doses of glucocorticoids. However, the role of endogenous glucocorticoids and the potential for antiglucocorticoid therapy to enhance angiogenesis is unknown. Using in vitro and in vivo models of angiogenesis in mice, we examined effects of (i) endogenous glucocorticoids, (ii) blocking endogenous glucocorticoid action with the glucocorticoid receptor antagonist RU38486, and (iii) abolishing local regeneration of glucocorticoids by the enzyme 11beta-hydroxysteroid dehydrogenase type 1 (11betaHSD1). Glucocorticoids, administered at physiological concentrations, inhibited angiogenesis in an in vitro aortic ring model and in vivo in polyurethane sponges implanted s.c. RU38486-enhanced angiogenesis in s.c. sponges, in healing surgical wounds, and in the myocardium of mice 7 days after myocardial infarction induced by coronary artery ligation. 11betaHSD1 knockout mice showed enhanced angiogenesis in vitro and in vivo within sponges, wounds, and infarcted myocardium. Endogenous glucocorticoids, including those generated locally by 11betaHSD1, exert tonic inhibition of angiogenesis. Inhibition of 11betaHSD1 in liver and adipose has been advocated to reduce cardiovascular risk in the metabolic syndrome: these data suggest that 11betaHSD1 inhibition offers a previously uncharacterized therapeutic approach to improve healing of ischemic or injured tissue.
Collapse
Affiliation(s)
- Gary R Small
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, Scotland, UK
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Dekker FJ, Koch MA, Waldmann H. Protein structure similarity clustering (PSSC) and natural product structure as inspiration sources for drug development and chemical genomics. Curr Opin Chem Biol 2005; 9:232-9. [PMID: 15939324 DOI: 10.1016/j.cbpa.2005.03.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2005] [Accepted: 03/22/2005] [Indexed: 02/04/2023]
Abstract
Finding small molecules that modulate protein function is of primary importance in drug development and in the emerging field of chemical genomics. To facilitate the identification of such molecules, we developed a novel strategy making use of structural conservatism found in protein domain architecture and natural product inspired compound library design. Domains and proteins identified as being structurally similar in their ligand-sensing cores are grouped in a protein structure similarity cluster (PSSC). Natural products can be considered as evolutionary pre-validated ligands for multiple proteins and therefore natural products that are known to interact with one of the PSSC member proteins are selected as guiding structures for compound library synthesis. Application of this novel strategy for compound library design provided enhanced hit rates in small compound libraries for structurally similar proteins.
Collapse
Affiliation(s)
- Frank J Dekker
- Department of Chemical Biology, Max-Planck Institute of Molecular Physiology, Otto-Hahn Str. 11, D-44227 Dortmund, Germany
| | | | | |
Collapse
|
38
|
Fotsch C, Askew BC, Chen JJ. 11β-Hydroxysteroid dehydrogenase-1 as a therapeutic target for metabolic diseases. Expert Opin Ther Pat 2005. [DOI: 10.1517/13543776.15.3.289] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
39
|
Balamurugan R, Dekker FJ, Waldmann H. Design of compound libraries based on natural product scaffolds and protein structure similarity clustering (PSSC). MOLECULAR BIOSYSTEMS 2005; 1:36-45. [PMID: 16880961 DOI: 10.1039/b503623b] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Recent advances in structural biology, bioinformatics and combinatorial chemistry have significantly impacted the discovery of small molecules that modulate protein functions. Natural products which have evolved to bind to proteins may serve as biologically validated starting points for the design of focused libraries that might provide protein ligands with enhanced quality and probability. The combined application of natural product derived scaffolds with a new approach that clusters proteins according to structural similarity of their ligand sensing cores provides a new principle for the design and synthesis of such libraries. This article discusses recent advances in the synthesis of natural product inspired compound collections and the application of protein structure similarity clustering for the development of such libraries.
Collapse
Affiliation(s)
- Rengarajan Balamurugan
- Department of Chemical Biology, Max-Planck Institute of Molecular Physiology, Dortmund, Germany
| | | | | |
Collapse
|
40
|
Sandeep TC, Andrew R, Homer NZM, Andrews RC, Smith K, Walker BR. Increased in vivo regeneration of cortisol in adipose tissue in human obesity and effects of the 11beta-hydroxysteroid dehydrogenase type 1 inhibitor carbenoxolone. Diabetes 2005; 54:872-9. [PMID: 15734867 DOI: 10.2337/diabetes.54.3.872] [Citation(s) in RCA: 156] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
11beta-Hydroxysteroid dehydrogenase type 1 (11HSD1) regenerates cortisol from cortisone within adipose tissue and liver. 11HSD1 inhibitors may enhance insulin sensitivity in type 2 diabetes and be most efficacious in obesity when 11HSD1 is increased in subcutaneous adipose biopsies. We examined the regeneration of cortisol in vivo in obesity, and the effects of the 11HSD1 inhibitor carbenoxolone. We compared six lean and six obese men and performed a randomized, placebo-controlled crossover study of carbenoxolone in obese men. The obese men had no difference in their whole-body rate of regenerating cortisol (measured with 9,11,12,12-[(2)H(4)]cortisol tracer), but had more rapid conversion of [(3)H]cortisone to [(3)H]cortisol in abdominal subcutaneous adipose tissue (measured with microdialysis). During insulin infusion, adipose 11HSD1 activity fell markedly in lean but not in obese men. Carbenoxolone inhibited whole-body cortisol regeneration, but did not significantly inhibit adipose 11HSD1 and had no effects on insulin sensitivity (measured by [(2)H(2)]glucose infusion with or without hyperinsulinemia). Thus, in vivo cortisol generation is increased selectively within adipose tissue in obesity, perhaps reflecting resistance to insulin-mediated downregulation of 11HSD1. However, obese men are less susceptible than lean men to the insulin-sensitizing effects of carbenoxolone. To be useful in obese patients, 11HSD1 inhibitors will need to inhibit the enzyme more effectively in adipose tissue.
Collapse
Affiliation(s)
- Thekkepat C Sandeep
- Endocrinology Unit, School of MolecularClinical Medicine, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, U.K
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
Metabolic syndrome (MS), a condition characterized by multiple related clinical disorders including insulin resistance, central obesity, hyperlipidemia, hypertension, and heart disease, is an increasingly prevalent disease in industrialized societies. The intense research interest in the peroxisome proliferator-activated receptors (PPARs), by both the pharmaceutical industry and academia, stems largely from the well-documented therapeutic actions of their synthetic agonists in alleviating several of the maladies associated with MS. This report focuses on the current understanding of the mechanisms of action of PPAR agents and their clinical use in the context of MS.
Collapse
Affiliation(s)
- Taro E Akiyama
- Merck Research Laboratories, PO Box 2000, RY80N-C31, 126 East Lincoln Avenue, Rahway, NJ 07065, USA.
| | | | | |
Collapse
|
42
|
Ogg D, Elleby B, Norström C, Stefansson K, Abrahmsén L, Oppermann U, Svensson S. The Crystal Structure of Guinea Pig 11β-Hydroxysteroid Dehydrogenase Type 1 Provides a Model for Enzyme-Lipid Bilayer Interactions. J Biol Chem 2005; 280:3789-94. [PMID: 15542590 DOI: 10.1074/jbc.m412463200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The metabolic reduction of 11-keto groups in glucocorticoid steroids such as cortisone leads to the nuclear receptor ligand cortisol. This conversion is an example of pre-receptor regulation and constitutes a novel pharmacological target for the treatment of metabolic disorders such as insulin resistance and possibly other derangements observed in the metabolic syndrome, such as hyperlipidemia, hypertension, and lowered insulin secretion. This reaction is carried out by the NADPH-dependent type 1 11beta-hydroxysteroid dehydrogenase (11beta-HSD1), an enzyme attached through an integral N-terminal transmembrane helix to the lipid bilayer and located with its active site within the lumen of the endoplasmic reticulum. Here we report the crystal structure of recombinant guinea pig 11beta-HSD1. This variant was determined in complex with NADP at 2.5 A resolution and crystallized in the presence of detergent and guanidinium hydrochloride. The overall structure of guinea pig 11beta-HSD1 shows a clear relationship to other members of the superfamily of short-chain dehydrogenases/reductases but harbors a unique C-terminal helical segment that fulfills three essential functions and accordingly is involved in subunit interactions, contributes to active site architecture, and is necessary for lipid-membrane interactions. The structure provides a model for enzyme-lipid bilayer interactions and suggests a funneling of lipophilic substrates such as steroid hormones from the hydrophobic membrane environment to the enzyme active site.
Collapse
Affiliation(s)
- Derek Ogg
- Department of Structural Chemistry, Biovitrum, SE-112 76 Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Insulin resistance is a common phenomenon of the metabolic syndrome, which is clinically characterized by a clustering of various cardiovascular risk factors in a single individual and a higher prevalence of respective complications, such as coronary heart disease and stroke. At the cellular level, insulin resistance is defined as a reduced insulin action, which can affect not only glucose uptake, but also gene regulation. Elucidation of novel signaling networks within the cell which are mediating and affecting insulin action will reveal many new genes and drug targets that are potentially of clinical relevance in the future. In this chapter, we propose that the metabolic syndrome might be a clinical consequence of altered gene regulation. This is illuminated in the context of transcription factors, e.g., sterol regulatory element binding proteins (SREBPs), coupling signals from nutrients, metabolites, and hormones at the gene regulatory level with pathobiochemical features of increased lipid accumulation in lean nonadipose tissues. The phenomenon of ectopic lipid accumulation (lipotoxicity) appears to be a novel link between insulin resistance, obesity, and possibly other features of the metabolic syndrome. Therefore, the investigation of specific gene regulatory networks and their alterations might be a clue to understanding the development and clustering of different cardiovascular risk factors in different individuals. As cellular sensors transcription factors--as common denominators of gene regulatory networks--might thereby also determine the susceptibility of individuals to cardiovascular risk factors and their complications.
Collapse
Affiliation(s)
- D Müller-Wieland
- Deutsches Diabetes-Zentrum, Institut für Klinische Biochemie und Pathobiochemie, Düsseldorf, Germany.
| | | |
Collapse
|
44
|
Koch MA, Wittenberg LO, Basu S, Jeyaraj DA, Gourzoulidou E, Reinecke K, Odermatt A, Waldmann H. Compound library development guided by protein structure similarity clustering and natural product structure. Proc Natl Acad Sci U S A 2004; 101:16721-6. [PMID: 15548605 PMCID: PMC534721 DOI: 10.1073/pnas.0404719101] [Citation(s) in RCA: 156] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
To identify biologically relevant and drug-like protein ligands for medicinal chemistry and chemical biology research the grouping of proteins according to evolutionary relationships and conservation of molecular recognition is an established method. We propose to employ structure similarity clustering of the ligand-sensing cores of protein domains (PSSC) in conjunction with natural product guided compound library development as a synergistic approach for the identification of biologically prevalidated ligands with high fidelity. This is supported by the concepts that (i) in nature spatial structure is more conserved than amino acid sequence, (ii) the number of fold types characteristic for all protein domains is limited, and (iii) the underlying frameworks of natural product classes with multiple biological activities provide evolutionarily selected starting points in structural space. On the basis of domain core similarity considerations and irrespective of sequence similarity, Cdc25A phosphatase, acetylcholinesterase, and 11beta-hydroxysteroid dehydrogenases type 1 and type 2 were grouped into a similarity cluster. A 147-member compound collection derived from the naturally occurring Cdc25A inhibitor dysidiolide yielded potent and selective inhibitors of the other members of the similarity cluster with a hit rate of 2-3%. Protein structure similarity clustering may provide an experimental opportunity to identify supersites in proteins.
Collapse
Affiliation(s)
- Marcus A Koch
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, D-44227 Dortmund, Germany
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Rosemond MJC, Walsh JS. Human Carbonyl Reduction Pathways and a Strategy for Their Study In Vitro. Drug Metab Rev 2004; 36:335-61. [PMID: 15237858 DOI: 10.1081/dmr-120034154] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Carbonyl reduction plays a significant role in physiological processes throughout the body. Although much is known about endogenous carbonyl metabolism, much less is known about the roles of carbonyl-reducing enzymes in xenobiotic metabolism. Multiple pathways exist in humans for metabolizing carbonyl moieties of xenobiotics to their corresponding alcohols, readying these molecules for subsequent conjugation and/or excretion. When exploring carbonyl reduction clearance pathways for a drug development candidate, it is possible to assess the relative contributions of these enzymes due to their differences in subcellular locations, cofactor dependence, and inhibitor profiles. In addition, the contributions of these enzymes may be explored by varying incubation conditions, such as pH. Presently, individual isoforms of carbonyl-reducing enzymes are not widely available, either in recombinant or purified form. However, it is possible to study carbonyl reduction clearance pathways from simple experiments with commercially available reagents. This article provides an overview of carbonyl-reducing enzymes, including some kinetic data for substrates and inhibitors. In addition, an experimental strategy for the study of these enzymes in vitro is presented.
Collapse
Affiliation(s)
- M Jane Cox Rosemond
- Worldwide Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, North Carolina, USA.
| | | |
Collapse
|