1
|
Rolan P, Seckl J, Taylor J, Harrison J, Maruff P, Woodward M, Mills R, Jaros M, Hilt D. Clinical Pharmacology and Approach to Dose Selection of Emestedastat, a Novel Tissue Cortisol Synthesis Inhibitor for the Treatment of Central Nervous System Disease. Clin Pharmacol Drug Dev 2025. [PMID: 39748632 DOI: 10.1002/cpdd.1496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025]
Abstract
This review demonstrates the value of central pharmacodynamics (PD), including positron emission tomography (PET) and computerized cognitive testing, to supplement pharmacokinetic (PK) and peripheral PD for determining the target dose range for clinical efficacy testing of emestedastat, an 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1) inhibitor. Combined data from 6 clinical trials in cognitively normal volunteers and patients with Alzheimer disease included a population PK model, endocrine PD, a human PET trial (11β-HSD1 brain imaging), and computerized cognitive testing. PK and PET findings were similar in volunteers and patients with Alzheimer disease. PK modeling suggested that 20 mg daily would be optimal to maintain cerebrospinal fluid concentrations above the brain half maximal inhibitory concentration. However, subsequent PET scanning suggested that emestedastat doses of 10 or even 5 mg daily may be sufficient to adequately inhibit 11β-HSD1. With once-daily doses of 5-20 mg in cognitively normal, older volunteers, a consistent pattern of pro-cognitive benefit, without dose-response, was seen as improvement in attention and working memory but not episodic memory. Thus, emestedastat therapeutic activity might be attained at doses lower than those predicted from cerebrospinal fluid drug levels. Doses as low as 5 mg daily may be efficacious and were studied in subsequent trials.
Collapse
Affiliation(s)
- Paul Rolan
- Actinogen Medical Ltd, Sydney, New South Wales, Australia
- University of Adelaide Medical School, Adelaide, South Australia, Australia
| | - Jonathan Seckl
- Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Jack Taylor
- Actinogen Medical Ltd, Sydney, New South Wales, Australia
| | - John Harrison
- Scottish Brain Sciences, Edinburgh, UK
- King's College, London, UK
- Alzheimercentrum, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Paul Maruff
- Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Michael Woodward
- Medical Health and Cognitive Research Unit, Austin Health, Heidelberg Repatriation Hospital, Heidelberg West, Victoria, Australia
| | | | | | - Dana Hilt
- Actinogen Medical Ltd, Sydney, New South Wales, Australia
| |
Collapse
|
2
|
Agbaria M, Egbaria N, Nairoukh Z. Dearomative spirocyclization of ynamides. Chem Sci 2024:d4sc05541a. [PMID: 39502502 PMCID: PMC11533057 DOI: 10.1039/d4sc05541a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024] Open
Abstract
Spiro N-heterocycles, particularly aza-spiro piperidines, have shown significant promise in pharmaceutical applications due to their ability to enhance physicochemical properties. Despite their potential, the preparation of these complex structures poses significant challenges. To address this, we propose a one-pot dearomative spirocyclization reaction of ynamides. This method involves a copper-catalyzed carbomagnesiation reaction, achieving chemo-, regio-, and stereoselective formation of vinyl metal intermediates. Upon the addition of a Lewis acid, these intermediates undergo a regioselective nucleophilic dearomatization event, facilitating the synthesis of diverse aza-spiro dihydropyridine scaffolds with multiple functional handles. Various Grignard reagents, diverse ynamides, and acylating reagents have been explored. A subsequent hydrogenation reaction provides access to both partially and fully reduced spirocyclic frameworks, broadening the scope of spirocyclic structures with potential medicinal applications.
Collapse
Affiliation(s)
- Mohamed Agbaria
- Institute of Chemistry, Casali Center of Applied Chemistry, The Hebrew University of Jerusalem Jerusalem 9190401 Israel
| | - Nwar Egbaria
- Institute of Chemistry, Casali Center of Applied Chemistry, The Hebrew University of Jerusalem Jerusalem 9190401 Israel
| | - Zackaria Nairoukh
- Institute of Chemistry, Casali Center of Applied Chemistry, The Hebrew University of Jerusalem Jerusalem 9190401 Israel
| |
Collapse
|
3
|
Zhong C, Li N, Wang S, Li D, Yang Z, Du L, Huang G, Li H, Yeung WS, He S, Ma S, Wang Z, Jiang H, Zhang H, Li Z, Wen X, Xue S, Tao X, Li H, Xie D, Zhang Y, Chen Z, Wang J, Yan J, Liang Z, Zhang Z, Zhong Z, Wu Z, Wan C, Liang C, Wang L, Yu S, Ma Y, Yu Y, Li F, Chen Y, Zhang B, Lyu A, Ren F, Zhou H, Liu J, Zhang G. Targeting osteoblastic 11β-HSD1 to combat high-fat diet-induced bone loss and obesity. Nat Commun 2024; 15:8588. [PMID: 39362888 PMCID: PMC11449908 DOI: 10.1038/s41467-024-52965-4] [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: 03/16/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024] Open
Abstract
Excessive glucocorticoid (GC) action is linked to various metabolic disorders. Recent findings suggest that disrupting skeletal GC signaling prevents bone loss and alleviates metabolic disorders in high-fat diet (HFD)-fed obese mice, underpinning the neglected contribution of skeletal GC action to obesity and related bone loss. Here, we show that the elevated expression of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), the enzyme driving local GC activation, and GC signaling in osteoblasts, are associated with bone loss and obesity in HFD-fed male mice. Osteoblast-specific 11β-HSD1 knockout male mice exhibit resistance to HFD-induced bone loss and metabolic disorders. Mechanistically, elevated 11β-HSD1 restrains glucose uptake and osteogenic activity in osteoblast. Pharmacologically inhibiting osteoblastic 11β-HSD1 by using bone-targeted 11β-HSD1 inhibitor markedly promotes bone formation, ameliorates glucose handling and mitigated obesity in HFD-fed male mice. Taken together, our study demonstrates that osteoblastic 11β-HSD1 directly contributes to HFD-induced bone loss, glucose handling impairment and obesity.
Collapse
Affiliation(s)
- Chuanxin Zhong
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Nanxi Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shengzheng Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Dijie Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangxi Universities Key Laboratory of Stem cell and Biopharmaceutical Technology, College of Life Sciences, Guangxi Normal University, Gui Lin, China
| | - Zhihua Yang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lin Du
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Guangxin Huang
- Department of Joint Surgery, The Third Affiliated Hospital of Southern Medical University, The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Haitian Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Wing Sze Yeung
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shan He
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Shuting Ma
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zhuqian Wang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Hewen Jiang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Huarui Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhanghao Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xiaoxin Wen
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Song Xue
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xiaohui Tao
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Haorui Li
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Duoli Xie
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yihao Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zefeng Chen
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Junqin Wang
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jianfeng Yan
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Zhengming Liang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zongkang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhigang Zhong
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Zeting Wu
- International Medical Service Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Chao Wan
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Chao Liang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Luyao Wang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Sifan Yu
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yuan Ma
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Fangfei Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yang Chen
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Baoting Zhang
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Aiping Lyu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China.
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Hong Kong, China.
| | - Fuzeng Ren
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Hong Zhou
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, Australia.
| | - Jin Liu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China.
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China.
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China.
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
4
|
Gómez C, Alimajstorovic Z, Othonos N, Winter DV, White S, Lavery GG, Tomlinson JW, Sinclair AJ, Odermatt A. Identification of a human blood biomarker of pharmacological 11β-hydroxysteroid dehydrogenase 1 inhibition. Br J Pharmacol 2024; 181:698-711. [PMID: 37740611 DOI: 10.1111/bph.16251] [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: 04/20/2023] [Revised: 08/16/2023] [Accepted: 09/12/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND AND PURPOSE 11β-Hydroxysteroid dehydrogenase-1 (11β-HSD1) catalyses the oxoreduction of cortisone to cortisol, amplifying levels of active glucocorticoids. It is a pharmaceutical target in metabolic disease and cognitive impairments. 11β-HSD1 also converts some 7oxo-steroids to their 7β-hydroxy forms. A recent study in mice described the ratio of tauroursodeoxycholic acid (TUDCA)/tauro-7oxolithocholic acid (T7oxoLCA) as a biomarker for decreased 11β-HSD1 activity. The present study evaluates the equivalent bile acid ratio of glycoursodeoxycholic acid (GUDCA)/glyco-7oxolithocholic acid (G7oxoLCA) as a biomarker for pharmacological 11β-HSD1 inhibition in humans and compares it with the currently applied urinary (5α-tetrahydrocortisol + tetrahydrocortisol)/tetrahydrocortisone ((5αTHF + THF)/THE) ratio. EXPERIMENTAL APPROACH Bile acid profiles were analysed by ultra-HPLC tandem-MS in blood samples from two independent, double-blind placebo-controlled clinical studies of the orally administered selective 11β-HSD1 inhibitor AZD4017. The blood GUDCA/G7oxoLCA ratio was compared with the urinary tetrahydro-glucocorticoid ratio for ability to detect 11β-HSD1 inhibition. KEY RESULTS No significant alterations were observed in bile acid profiles following 11β-HSD1 inhibition by AZD4017, except for an increase of the secondary bile acid G7oxoLCA. The enzyme product/substrate ratio GUDCA/G7oxoLCA was found to be more reliable to detect 11β-HSD1 inhibition than the absolute G7oxoLCA concentration in both cohorts. Comparison of the blood GUDCA/G7oxoLCA ratio with the urinary (5αTHF + THF)/THE ratio revealed that both successfully detect 11β-HSD1 inhibition. CONCLUSIONS AND IMPLICATIONS 11β-HSD1 inhibition does not cause major alterations in bile acid homeostasis. The GUDCA/G7oxoLCA ratio represents the first blood biomarker of pharmacological 11β-HSD1 inhibition and may replace or complement the urinary (5αTHF + THF)/THE ratio biomarker.
Collapse
Affiliation(s)
- Cristina Gómez
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Zerin Alimajstorovic
- Metabolic Neurology, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Nantia Othonos
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, UK
| | - Denise V Winter
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Sarah White
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, UK
| | - Gareth G Lavery
- Department for Biosciences, Nottingham Trent University, Nottingham, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, UK
| | - Alexandra J Sinclair
- Metabolic Neurology, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Department of Neurology, University Hospitals Birmingham, Birmingham, UK
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| |
Collapse
|
5
|
Prabhakar PK, Batiha GES. Potential Therapeutic Targets for the Management of Diabetes Mellitus Type 2. Curr Med Chem 2024; 31:3167-3181. [PMID: 37125833 DOI: 10.2174/0929867330666230501172557] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/14/2023] [Accepted: 03/26/2023] [Indexed: 05/02/2023]
Abstract
Diabetes is one of the lifelong chronic metabolic diseases which is prevalent globally. There is a continuous rise in the number of people suffering from this disease with time. It is characterized by hyperglycemia, which leads to severe damage to the body's system, such as blood vessels and nerves. Diabetes occurs due to the dysfunction of pancreatic β -cell which leads to the reduction in the production of insulin or body cells unable to use insulin produce efficiently. As per the data shared International diabetes federation (IDF), there are around 415 million affected by this disease worldwide. There are a number of hit targets available that can be focused on treating diabetes. There are many drugs available and still under development for the treatment of type 2 diabetes. Inhibition of gluconeogenesis, lipolysis, fatty acid oxidation, and glucokinase activator is emerging targets for type 2 diabetes treatment. Diabetes management can be supplemented with drug intervention for obesity. The antidiabetic drug sale is the second-largest in the world, trailing only that of cancer. The future of managing diabetes will be guided by research on various novel targets and the development of various therapeutic leads, such as GLP-1 agonists, DPP-IV inhibitors, and SGLT2 inhibitors that have recently completed their different phases of clinical trials. Among these therapeutic targets associated with type 2 diabetes, this review focused on some common therapeutic targets for developing novel drug candidates of the newer generation with better safety and efficacy.
Collapse
Affiliation(s)
- Pranav Kumar Prabhakar
- Division of Research and Development, Lovely Professional University, Phagwara (Punjab) 144411, India
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt
| |
Collapse
|
6
|
Abstract
11-beta-hydroxysteroid dehydrogenases (11β-HSDs) catalyse the conversion of active 11-hydroxy glucocorticoids (cortisol, corticosterone) and their inert 11-keto forms (cortisone, 11-dehydrocorticosterone). They were first reported in the body and brain 70 years ago, but only recently have they become of interest. 11β-HSD2 is a dehydrogenase, potently inactivating glucocorticoids. In the kidney, 11β-HSD2 generates the aldosterone-specificity of intrinsically non-selective mineralocorticoid receptors. 11β-HSD2 also protects the developing foetal brain and body from premature glucocorticoid exposure, which otherwise engenders the programming of neuropsychiatric and cardio-metabolic disease risks. In the adult CNS, 11β-HSD2 is confined to a part of the brain stem where it generates aldosterone-specific central control of salt appetite and perhaps blood pressure. 11β-HSD1 is a reductase, amplifying active glucocorticoid levels within brain cells, notably in the cortex, hippocampus and amygdala, paralleling its metabolic functions in peripheral tissues. 11β-HSD1 is elevated in the ageing rodent and, less certainly, human forebrain. Transgenic models show this rise contributes to age-related cognitive decline, at least in mice. 11β-HSD1 inhibition robustly improves memory in healthy and pathological ageing rodent models and is showing initial promising results in phase II studies of healthy elderly people. Larger trials are needed to confirm and clarify the magnitude of effect and define target populations. The next decade will be crucial in determining how this tale ends - in new treatments or disappointment.
Collapse
Affiliation(s)
- Jonathan Seckl
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
7
|
Patel VK, Vaishnaw A, Shirbhate E, Kore R, Singh V, Veerasamy R, Rajak H. Cortisol as a Target for Treating Mental Disorders: A Promising Avenue for Therapy. Mini Rev Med Chem 2024; 24:588-600. [PMID: 37861053 DOI: 10.2174/0113895575262104230928042150] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/19/2023] [Accepted: 08/05/2023] [Indexed: 10/21/2023]
Abstract
Cortisol, commonly known as the "stress hormone," plays a critical role in the body's response to stress. Elevated cortisol levels have been associated with various mental disorders, including anxiety, depression, and post-traumatic stress disorder. Consequently, researchers have explored cortisol modulation as a promising avenue for treating these conditions. However, the availability of research on cortisol as a therapeutic option for mental disorders is limited, and existing studies employ diverse methodologies and outcome measures. This review article aimed to provide insights into different treatment approaches, both pharmacological and non-pharmacological, which can effectively modulate cortisol levels. Pharmacological interventions involve the use of substances, such as somatostatin analogs, dopamine agonists, corticotropin-releasing hormone antagonists, and cortisol synthesis inhibitors. Additionally, non-pharmacological techniques, including cognitivebehavioral therapy, herbs and supplements, transcranial magnetic stimulation, lifestyle changes, and surgery, have been investigated to reduce cortisol levels. The emerging evidence suggests that cortisol modulation could be a promising treatment option for mental disorders. However, more research is needed to fully understand the effectiveness and safety of these therapies.
Collapse
Affiliation(s)
- Vijay K Patel
- Pushpendra College of Pharmacy, Ambikapur, Surguja 497101, (C.G.), India
| | - Aayush Vaishnaw
- Dr. C.V. Raman Institute of Pharmacy, Dr. C.V. Raman University, Bilaspur, C.G. 495113, India
| | - Ekta Shirbhate
- Department of Pharmacy, Guru Ghasidas University, Bilaspur 495 009, (C.G.), India
| | - Rakesh Kore
- Department of Pharmacy, Guru Ghasidas University, Bilaspur 495 009, (C.G.), India
| | - Vaibhav Singh
- Department of Pharmacy, Guru Ghasidas University, Bilaspur 495 009, (C.G.), India
| | - Ravichandran Veerasamy
- Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Harish Rajak
- Department of Pharmacy, Guru Ghasidas University, Bilaspur 495 009, (C.G.), India
| |
Collapse
|
8
|
Taves MD, Otsuka S, Taylor MA, Donahue KM, Meyer TJ, Cam MC, Ashwell JD. Tumors produce glucocorticoids by metabolite recycling, not synthesis, and activate Tregs to promote growth. J Clin Invest 2023; 133:e164599. [PMID: 37471141 PMCID: PMC10503810 DOI: 10.1172/jci164599] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 07/18/2023] [Indexed: 07/22/2023] Open
Abstract
Glucocorticoids are steroid hormones with potent immunosuppressive properties. Their primary source is the adrenals, where they are generated via de novo synthesis from cholesterol. In addition, many tissues have a recycling pathway in which glucocorticoids are regenerated from inactive metabolites by the enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1, encoded by Hsd11b1). Here, we find that multiple tumor types express Hsd11b1 and produce active glucocorticoids. Genetic ablation of Hsd11b1 in such cells had no effect on in vitro growth, but reduced in vivo tumor progression, which corresponded with increased frequencies of CD8+ tumor-infiltrating lymphocytes (TILs) expressing activation markers and producing effector cytokines. Tumor-derived glucocorticoids were found to promote signatures of Treg activation and suppress signatures of conventional T cell activation in tumor-infiltrating Tregs. Indeed, CD8+ T cell activation was restored and tumor growth reduced in mice with Treg-specific glucocorticoid receptor deficiency. Importantly, pharmacologic inhibition of 11β-HSD1 reduced tumor growth to the same degree as gene knockout and rendered immunotherapy-resistant tumors susceptible to PD-1 blockade. Given that HSD11B1 expression is upregulated in many human tumors and that inhibition of 11β-HSD1 is well tolerated in clinical studies, these data suggest that targeting 11β-HSD1 may be a beneficial adjunct in cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | - Thomas J. Meyer
- CCR Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Margaret C. Cam
- CCR Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | | |
Collapse
|
9
|
Martins Nascentes Melo L, Herrera-Rios D, Hinze D, Löffek S, Oezel I, Turiello R, Klein J, Leonardelli S, Westedt IV, Al-Matary Y, Egea-Rodriguez S, Brenzel A, Bau M, Sucker A, Hadaschik E, Wirsdörfer F, Hanenberg H, Uhlenbrock N, Rauh D, Poźniak J, Rambow F, Marine JC, Effern M, Glodde N, Schadendorf D, Jablonska J, Hölzel M, Helfrich I. Glucocorticoid activation by HSD11B1 limits T cell-driven interferon signaling and response to PD-1 blockade in melanoma. J Immunother Cancer 2023; 11:e004150. [PMID: 37028818 PMCID: PMC10083881 DOI: 10.1136/jitc-2021-004150] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2023] [Indexed: 04/09/2023] Open
Abstract
BACKGROUND Immune responses against tumors are subject to negative feedback regulation. Immune checkpoint inhibitors (ICIs) blocking Programmed cell death protein 1 (PD-1), a receptor expressed on T cells, or its ligand PD-L1 have significantly improved the treatment of cancer, in particular malignant melanoma. Nevertheless, responses and durability are variables, suggesting that additional critical negative feedback mechanisms exist and need to be targeted to improve therapeutic efficacy. METHODS We used different syngeneic melanoma mouse models and performed PD-1 blockade to identify novel mechanisms of negative immune regulation. Genetic gain-of-function and loss-of-function approaches as well as small molecule inhibitor applications were used for target validation in our melanoma models. We analyzed mouse melanoma tissues from treated and untreated mice by RNA-seq, immunofluorescence and flow cytometry to detect changes in pathway activities and immune cell composition of the tumor microenvironment. We analyzed tissue sections of patients with melanoma by immunohistochemistry as well as publicly available single-cell RNA-seq data and correlated target expression with clinical responses to ICIs. RESULTS Here, we identified 11-beta-hydroxysteroid dehydrogenase-1 (HSD11B1), an enzyme that converts inert glucocorticoids into active forms in tissues, as negative feedback mechanism in response to T cell immunotherapies. Glucocorticoids are potent suppressors of immune responses. HSD11B1 was expressed in different cellular compartments of melanomas, most notably myeloid cells but also T cells and melanoma cells. Enforced expression of HSD11B1 in mouse melanomas limited the efficacy of PD-1 blockade, whereas small molecule HSD11B1 inhibitors improved responses in a CD8+ T cell-dependent manner. Mechanistically, HSD11B1 inhibition in combination with PD-1 blockade augmented the production of interferon-γ by T cells. Interferon pathway activation correlated with sensitivity to PD-1 blockade linked to anti-proliferative effects on melanoma cells. Furthermore, high levels of HSD11B1, predominantly expressed by tumor-associated macrophages, were associated with poor responses to ICI therapy in two independent cohorts of patients with advanced melanomas analyzed by different methods (scRNA-seq, immunohistochemistry). CONCLUSION As HSD11B1 inhibitors are in the focus of drug development for metabolic diseases, our data suggest a drug repurposing strategy combining HSD11B1 inhibitors with ICIs to improve melanoma immunotherapy. Furthermore, our work also delineated potential caveats emphasizing the need for careful patient stratification.
Collapse
Affiliation(s)
- Luiza Martins Nascentes Melo
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Dayana Herrera-Rios
- Department of Dermatology and Allergy, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Daniel Hinze
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Stefanie Löffek
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Irem Oezel
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Roberta Turiello
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Juliane Klein
- Department of Dermatology and Allergy, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Sonia Leonardelli
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Isa-Vanessa Westedt
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Yahya Al-Matary
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Sara Egea-Rodriguez
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- Department of Dermatology and Allergy, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Alexandra Brenzel
- Institute for Experimental Immunology and Imaging, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Maja Bau
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Antje Sucker
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Eva Hadaschik
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany
| | - Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Helmut Hanenberg
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Niklas Uhlenbrock
- Drug Discovery Hub Dortmund (DDHD) am Zentrum für integrierte Wirkstoffforschung (ZIW), Dortmund, Belgium
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Daniel Rauh
- Drug Discovery Hub Dortmund (DDHD) am Zentrum für integrierte Wirkstoffforschung (ZIW), Dortmund, Belgium
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Joanna Poźniak
- Center for Cancer Biology, VIB-KU Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Florian Rambow
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
- Center for Cancer Biology, VIB-KU Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jean-Christophe Marine
- Center for Cancer Biology, VIB-KU Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Maike Effern
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Nicole Glodde
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Dirk Schadendorf
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Jadwiga Jablonska
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Michael Hölzel
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Iris Helfrich
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany
- Department of Dermatology and Allergy, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| |
Collapse
|
10
|
Chen Q, Zhou X, Rehmel J, Steele JP, Svensson KA, Beck JP, Hembre EJ, Hao J. Ensemble Docking Approach to Mitigate Pregnane X Receptor-Mediated CYP3A4 Induction Risk. J Chem Inf Model 2023; 63:173-186. [PMID: 36473234 DOI: 10.1021/acs.jcim.2c01175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Three structurally closely related dopamine D1 receptor positive allosteric modulators (D1 PAMs) based on a tetrahydroisoquinoline (THIQ) scaffold were profiled for their CYP3A4 induction potentials. It was found that the length of the linker at the C5 position greatly affected the potentials of these D1 PAMs as CYP3A4 inducers, and the level of induction correlated well with the activation of the pregnane X receptor (PXR). Based on the published PXR X-ray crystal structures, we built a binding model specifically for these THIQ-scaffold-based D1 PAMs in the PXR ligand-binding pocket via an ensemble docking approach and found the model could explain the observed CYP induction disparity. Combined with our previously reported D1 receptor homology model, which identified the C5 position as pointing toward the solvent-exposed space, our PXR-binding model coincidentally suggested that structural modifications at the C5 position could productively modulate the CYP induction potential while maintaining the D1 PAM potency of these THIQ-based PAMs.
Collapse
Affiliation(s)
- Qi Chen
- Discovery Chemistry Research and Technologies, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - Xin Zhou
- Drug Disposition, Lilly Biotechnology Center, Eli Lilly and Company, 10290 Campus Point Drive, San Diego, California92121, United States
| | - Jessica Rehmel
- Drug Disposition, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - James P Steele
- Quantitative Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - Kjell A Svensson
- Neuroscience Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - James P Beck
- Discovery Chemistry Research and Technologies, Lilly Biotechnology Center, Eli Lilly and Company, 10290 Campus Point Drive, San Diego, California92121, United States
| | - Erik J Hembre
- Discovery Chemistry Research and Technologies, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - Junliang Hao
- Discovery Chemistry Research and Technologies, Lilly Biotechnology Center, Eli Lilly and Company, 10290 Campus Point Drive, San Diego, California92121, United States
| |
Collapse
|
11
|
Modern Photocatalytic Strategies in Natural Product Synthesis. PROGRESS IN THE CHEMISTRY OF ORGANIC NATURAL PRODUCTS 2023; 120:1-104. [PMID: 36587307 DOI: 10.1007/978-3-031-11783-1_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Modern photocatalysis has proven its generality for the development and functionalization of native functionalities. To date, the field has found broad applications in diverse research areas, including the total synthesis of natural products. This contribution covers recent reports of total syntheses involving as a key step a photocatalytic reaction. Among the selected examples, the photocatalytic processes proceed in a highly chemo-, regio-, and stereoselective manner, thereby allowing the rapid access to structurally complex architectures under light-driven conditions.
Collapse
|
12
|
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
|
13
|
Deng X, Huang SL, Ren J, Pan ZH, Shen Y, Zhou HF, Zuo ZL, Leng Y, Zhao QS. Development and structure-activity relationships of tanshinones as selective 11β-hydroxysteroid dehydrogenase 1 inhibitors. NATURAL PRODUCTS AND BIOPROSPECTING 2022; 12:36. [PMID: 36131216 PMCID: PMC9492458 DOI: 10.1007/s13659-022-00358-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/26/2022] [Indexed: 06/15/2023]
Abstract
11β-Hydroxysteroid dehydrogenase 1 (11β-HSD1) represents a promising drug target for metabolic syndrome, including obesity and type 2 diabetes. Our initial screen of a collection of natural products from Danshen led to the identification of tanshinones as the potent and selective 11β-HSD1 inhibitors. To improve the druggability and explore the structure-activity relationships (SARs), more than 40 derivatives have been designed and synthesized using tanshinone IIA and cryptotanshinone as the starting materials. More than 10 derivatives exhibited potent in vitro 11β-HSD1 inhibitory activity and good selectivity over 11β-HSD2 across human and mouse species. Based on the biological results, SARs were further discussed, which was also partially rationalized by a molecular docking model of 1 bound to the 11β-HSD1. Remarkably, compounds 1, 17 and 30 significantly inhibited 11β-HSD1 in 3T3-L1 adipocyte and in livers of ob/ob mice, which merits further investigations as anti-diabetic agents. This study not only provides a series of novel selective 11β-HSD1 inhibitors with promising therapeutic potentials in metabolic syndromes, but also expands the boundaries of the chemical and biological spaces of tanshinones.
Collapse
Affiliation(s)
- Xu Deng
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650204, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Su-Ling Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jian Ren
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650204, China
| | - Zheng-Hong Pan
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650204, China
- Guangxi Key Laboratory of Functional Phytochemicals Research and Utilization, Guangxi Institute of Botany, Chinese Academy of Sciences, Guilin, 541006, China
| | - Yu Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hao-Feng Zhou
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650204, China
| | - Zhi-Li Zuo
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650204, China.
| | - Ying Leng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Qin-Shi Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650204, China.
| |
Collapse
|
14
|
Burns DM, He C, Li YL, Zhuo J, Qian DQ, Chen L, Jalluri R, Diamond S, Covington MB, Li Y, Wynn R, Scherle P, Yeleswaram S, Hollis G, Friedman S, Metcalf B, Yao W. Discovery of a novel 2-spiroproline steroid mimetic scaffold for the potent inhibition of 11β-HSD1. Bioorg Med Chem Lett 2022; 73:128884. [PMID: 35835377 DOI: 10.1016/j.bmcl.2022.128884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 11/30/2022]
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 a variety of ailments including 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 3-point pharmacophore for 11β-HSD1 that was utilized to design a 2-spiroproline derivative as a steroid mimetic scaffold. Reiterative optimization provided valuable insight into the bioactive conformation of our novel scaffold and led to the discovery of several leads, such as compounds 39 and 51. Importantly, deleterious hERG inhibition and pregnane X receptor induction were mitigated by the introduction of a 4-hydroxyl group to the proline ring system.
Collapse
Affiliation(s)
- David M Burns
- 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
- Prelude Therapeutics, 200 Powder Mill Road, Wilmington, DE 19803, USA
| | - Ding-Quan Qian
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | | | | | - Sharon Diamond
- 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
- Prelude Therapeutics, 200 Powder Mill Road, Wilmington, DE 19803, USA
| | - Swamy Yeleswaram
- Incyte Research Institute, 1801 Augustine Cut-off, Wilmington, DE 19880, USA
| | | | | | | | | |
Collapse
|
15
|
Singh R, Bhardwaj VK, Das P, Purohit R. Identification of 11β-HSD1 inhibitors through enhanced sampling methods. Chem Commun (Camb) 2022; 58:5005-5008. [PMID: 35362492 DOI: 10.1039/d1cc06894f] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Aminoarylbenzosuberene (AAB) molecules were chosen for in silico analysis to develop effective and more competent 11β-hydroxysteroid dehydrogenase (11β-HSD1) protein inhibitors. The AAB4 molecule was shown to have stronger interactions and binding affinity than standard inhibitors (co-crystallized molecules). These results were based on conventional, steered and enhanced umbrella sampling simulations.
Collapse
Affiliation(s)
- Rahul Singh
- Structural Bioinformatics Lab, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, HP, 176061, India. .,Biotechnology division, CSIR-IHBT, Palampur, HP, 176061, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Vijay Kumar Bhardwaj
- Structural Bioinformatics Lab, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, HP, 176061, India. .,Biotechnology division, CSIR-IHBT, Palampur, HP, 176061, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Pralay Das
- Biotechnology division, CSIR-IHBT, Palampur, HP, 176061, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad-201002, India.,Natural Product Chemistry and Process Development, CSIR-IHBT, Palampur, HP, 176061, India
| | - Rituraj Purohit
- Structural Bioinformatics Lab, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, HP, 176061, India. .,Biotechnology division, CSIR-IHBT, Palampur, HP, 176061, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad-201002, India
| |
Collapse
|
16
|
Kittikool T, Phakdeeyothin K, Chantarojsiri T, Yotphan S. Manganese‐Promoted Regioselective Direct
C3
‐Phosphinoylation of 2‐Pyridones. European J Org Chem 2021. [DOI: 10.1002/ejoc.202100336] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Tanakorn Kittikool
- Department of Chemistry and Center of Excellence for Innovation in Chemistry Faculty of Science Mahidol University Rama VI Road 10400 Bangkok Thailand
| | - Kunita Phakdeeyothin
- Department of Chemistry and Center of Excellence for Innovation in Chemistry Faculty of Science Mahidol University Rama VI Road 10400 Bangkok Thailand
| | - Teera Chantarojsiri
- Department of Chemistry and Center of Excellence for Innovation in Chemistry Faculty of Science Mahidol University Rama VI Road 10400 Bangkok Thailand
| | - Sirilata Yotphan
- Department of Chemistry and Center of Excellence for Innovation in Chemistry Faculty of Science Mahidol University Rama VI Road 10400 Bangkok Thailand
| |
Collapse
|
17
|
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
|
18
|
Weingartner M, Stücheli S, Kratschmar DV, Birk J, Klusonova P, Chapman KE, Lavery GG, Odermatt A. The ratio of ursodeoxycholyltaurine to 7-oxolithocholyltaurine serves as a biomarker of decreased 11β-hydroxysteroid dehydrogenase 1 activity in mouse. Br J Pharmacol 2021; 178:3309-3326. [PMID: 33450045 PMCID: PMC8359391 DOI: 10.1111/bph.15367] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 12/06/2020] [Accepted: 12/27/2020] [Indexed: 11/30/2022] Open
Abstract
Background and Purpose 11β‐Hydroxysteroid dehydrogenase 1 (11β‐HSD1) regulates tissue‐specific glucocorticoid metabolism and its impaired expression and activity are associated with major diseases. Pharmacological inhibition of 11β‐HSD1 is considered a promising therapeutic strategy. This study investigated whether alternative 7‐oxo bile acid substrates of 11β‐HSD1 or the ratios to their 7‐hydroxy products can serve as biomarkers for decreased enzymatic activity. Experimental Approach Bile acid profiles were measured by ultra‐HPLC tandem‐MS in plasma and liver tissue samples of four different mouse models with decreased 11β‐HSD1 activity: global (11KO) and liver‐specific 11β‐HSD1 knockout mice (11LKO), mice lacking hexose‐6‐phosphate dehydrogenase (H6pdKO) that provides cofactor NADPH for 11β‐HSD1 and mice treated with the pharmacological inhibitor carbenoxolone. Additionally, 11β‐HSD1 expression and activity were assessed in H6pdKO‐ and carbenoxolone‐treated mice. Key Results The enzyme product to substrate ratios were more reliable markers of 11β‐HSD1 activity than absolute levels due to large inter‐individual variations in bile acid concentrations. The ratio of the 7β‐hydroxylated ursodeoxycholyltaurine (UDC‐Tau) to 7‐oxolithocholyltaurine (7oxoLC‐Tau) was diminished in plasma and liver tissue of all four mouse models and decreased in H6pdKO‐ and carbenoxolone‐treated mice with moderately reduced 11β‐HSD1 activity. The persistence of 11β‐HSD1 oxoreduction activity in the face of H6PD loss indicates the existence of an alternative NADPH source in the endoplasmic reticulum. Conclusions and Implications The plasma UDC‐Tau/7oxo‐LC‐Tau ratio detects decreased 11β‐HSD1 oxoreduction activity in different mouse models. This ratio may be a useful biomarker of decreased 11β‐HSD1 activity in pathophysiological situations or upon pharmacological inhibition. LINKED ARTICLES This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc
Collapse
Affiliation(s)
- Michael Weingartner
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Simon Stücheli
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Denise V Kratschmar
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Julia Birk
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Petra Klusonova
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Karen E Chapman
- Queen's Medical Research Institute, University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| |
Collapse
|
19
|
Yang Z. Navigating the Pauson-Khand Reaction in Total Syntheses of Complex Natural Products. Acc Chem Res 2021; 54:556-568. [PMID: 33412841 DOI: 10.1021/acs.accounts.0c00709] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
"Total synthesis endeavors provide wonderful opportunities to discover and invent new synthetic reactions as a means to advance organic synthesis in general. Such discoveries and inventions can occur when the practitioner faces intransigent problems that cannot be solved by known methods and/or when method improvements are desired in terms of elegance, efficiency, cost-effectiveness, practicality, or environmental friendliness" (K. C. Nicolaou et al. from their review in CCS Chem. 2019, 1, 3-37). To date tens of thousands of bioactive compounds have been isolated from plants, microbes, marine invertebrates, and other sources. These chemical structures have been studied by chemists who scanned the breadth of natural diversity toward drug discovery efforts. Drug-likeness of natural products often possesses common features including molecular complexity, protein-binding ability, structural rigidity, and three-dimensionality. Considering certain biologically important natural products are scarce from natural supply, total synthesis may provide an alternative solution to generating these compounds and their derivatives for the purpose of probing their biological functions. Natural products bearing quaternary carbon stereocenters represent a group of biologically important natural entities that are lead compounds in the development of pharmacological agents and biological probes. However, the stereocontrolled introduction of quaternary carbons, with vicinal patterns that substantially expand the complexity of molecular architectures and chemical space in particular, presents distinct challenges because of the high steric repulsion between substituents. Though remarkable advance has been seen for quaternary carbon stereocenter generation, the process remains a daunting challenge given that the formation of highly congested stereocenters increases the difficulty in achieving orbital overlap.In the past two decades, our group has initiated a program to develop synthetic strategies and methods with the aim of advancing the frontiers of the total syntheses of biologically important complex natural products bearing all-carbon quaternary stereogenic centers. Typical endeavors have involved the use of a Pauson-Khand (PK) reaction as a key step in constructing core structures with all-carbon quaternary stereogenic center(s), with the aid of well-orchestrated thiourea-Co- and thiourea-Pd-catalyzed PK reactions. These methodological advances have enabled us to achieve total syntheses of a series of topologically complex natural products with diverse structural features. These methods will enable the assembly of molecules with improved biological functions and provide tool compounds for elucidation of mechanism of action or identification of potential cellular targets.
Collapse
Affiliation(s)
- Zhen Yang
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Beijing National Laboratory for Molecular Science and Key Laboratory of Bioorganic Chemistry and Molecular Engineering of the Ministry of Education, College of Chemistry and Molecular Engineering and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
- Shenzhen Bay Laboratory, Shenzhen 518055, China
| |
Collapse
|
20
|
Yan H, Li X, Ni W, Zhao Q, Leng Y, Liu HY. Phytochemicals from the Leaves of Cyclocarya paliurus and their 11β-HSD1 Enzyme Inhibitory Effects. Chem Biodivers 2020; 18:e2000772. [PMID: 33369207 DOI: 10.1002/cbdv.202000772] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/18/2020] [Indexed: 11/11/2022]
Abstract
Two new dammarane-type triterpenoid saponins, 3β-(α-l-arabinopyranosyloxy)-24,25-dihydroxydammar-20-en-12α-yl 6-deoxy-β-d-glucopyranoside (1) and (24R)-3β-[(4-O-acetyl-α-l-arabinopyranosyl)oxy]-25-hydroxy-20,24-epoxydammaran-12β-yl 6-deoxy-β-d-glucopyranoside (2), and fourteen known triterpenoids were isolated from the 70 % MeOH extract of the leaves of Cyclocarya paliurus. Their structures were established based on analyses of spectroscopic data. All compounds were tested for their inhibitory activities against the 11β-HSD1 enzyme. Hederagenin (13) exhibited moderate inhibitory effect for mouse 11β-HSD1 with an IC50 value of 0.16±0.04 μM.
Collapse
Affiliation(s)
- Huan Yan
- State Key Laboratory of Phytochemistry and Plant Resources in West China, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, P. R. China
| | - Xiang Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, P. R. China.,Yunnan University of Chinese Medicine, Kunming, 650500, P. R. China
| | - Wei Ni
- State Key Laboratory of Phytochemistry and Plant Resources in West China, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, P. R. China
| | - Qing Zhao
- Yunnan University of Chinese Medicine, Kunming, 650500, P. R. China
| | - Ying Leng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, P. R. China
| | - Hai-Yang Liu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, P. R. China
| |
Collapse
|
21
|
Zhang W, Zhang Z, Tang JC, Che JT, Zhang HY, Chen JH, Yang Z. Total Synthesis of (+)-Haperforin G. J Am Chem Soc 2020; 142:19487-19492. [DOI: 10.1021/jacs.0c10122] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Wei Zhang
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education and Beijing National Laboratory for Molecular Science (BNLMS), and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Zhenyu Zhang
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education and Beijing National Laboratory for Molecular Science (BNLMS), and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Jun-Chen Tang
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education and Beijing National Laboratory for Molecular Science (BNLMS), and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Jin-Teng Che
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education and Beijing National Laboratory for Molecular Science (BNLMS), and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Hao-Yu Zhang
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education and Beijing National Laboratory for Molecular Science (BNLMS), and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Jia-Hua Chen
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education and Beijing National Laboratory for Molecular Science (BNLMS), and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Zhen Yang
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education and Beijing National Laboratory for Molecular Science (BNLMS), and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Shenzhen Bay Laboratory, Shenzhen 518055, China
| |
Collapse
|
22
|
Ganjoo A, Tripathi A, Chetti P. Structural assessment and identification of 11β-hydroxysteroid dehydrogenase type 1 inhibitors. J Biomol Struct Dyn 2020; 38:4928-4937. [DOI: 10.1080/07391102.2019.1688193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Ananta Ganjoo
- Department of Chemistry, National Institute of Technology, Kurukshetra, India
| | - Anuj Tripathi
- Department of Chemistry, National Institute of Technology, Kurukshetra, India
| | - Prabhakar Chetti
- Department of Chemistry, National Institute of Technology, Kurukshetra, India
| |
Collapse
|
23
|
Kupczyk D, Studzińska R, Bilski R, Baumgart S, Kołodziejska R, Woźniak A. Synthesis of Novel 2-(Isopropylamino)thiazol-4(5 H)-one Derivatives and Their Inhibitory Activity of 11β-HSD1 and 11β-HSD2 in Aspect of Carcinogenesis Prevention. Molecules 2020; 25:E4233. [PMID: 32942682 PMCID: PMC7570983 DOI: 10.3390/molecules25184233] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 01/24/2023] Open
Abstract
Glucocorticoid metabolism at the tissue level is regulated by two isoenzymes 11β-hydroxysteroid dehydrogenase (11β-HSD), which mutually convert biologically active cortisol and inactive cortisone. Recent research is focused on the role of 11β-HSD1 and 11β-HSD2 as autocrine factors of tumor cell proliferation and differentiation. Herein, we report the synthesis of novel 2-(isopropylamino)thiazol-4(5H)-one derivatives and their inhibitory activity for 11β-HSD1 and 11β-HSD2. The derivative containing the spiro system of thiazole and cyclohexane rings shows the highest degree of 11β-HSD1 inhibition (54.53% at 10 µM) and is the most selective inhibitor of this enzyme among the tested compounds. In turn, derivatives containing ethyl and n-propyl group at C-5 of thiazole ring inhibit the activity of 11β-HSD2 to a high degree (47.08 and 54.59% at 10 µM respectively) and are completely selective. Inhibition of the activity of these enzymes may have a significant impact on the process of formation and course of tumors. Therefore, these compounds can be considered as potential pharmaceuticals supporting anti-cancer therapy.
Collapse
Affiliation(s)
- Daria Kupczyk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland; (R.B.); (R.K.); (A.W.)
| | - Renata Studzińska
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-089 Bydgoszcz, Poland;
| | - Rafał Bilski
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland; (R.B.); (R.K.); (A.W.)
| | - Szymon Baumgart
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-089 Bydgoszcz, Poland;
| | - Renata Kołodziejska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland; (R.B.); (R.K.); (A.W.)
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland; (R.B.); (R.K.); (A.W.)
| |
Collapse
|
24
|
He Y, Liu Y, Liu Y, Kou X, Li Q, Li J, Jiang H, Leng H, Peng C, Li J. Diastereodivergent Formal 1,3‐Dipolar Cycloaddition of 5‐alkenyl thiazolones to Access Stereochemically Diverse Pyrrolidinyl Spirooxindoles. Adv Synth Catal 2020. [DOI: 10.1002/adsc.201901541] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yu He
- School of Life Science and EngineeringSouthwest Jiaotong University Chengdu 610031 People's Republic of China
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of AntibioticsChengdu University Chengdu 610052 People's Republic of China
| | - Yue Liu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of AntibioticsChengdu University Chengdu 610052 People's Republic of China
| | - Yu Liu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of AntibioticsChengdu University Chengdu 610052 People's Republic of China
| | - Xin‐Xin Kou
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of AntibioticsChengdu University Chengdu 610052 People's Republic of China
| | - Qing‐Zhu Li
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of AntibioticsChengdu University Chengdu 610052 People's Republic of China
| | - Jia‐Hong Li
- School of Life Science and EngineeringSouthwest Jiaotong University Chengdu 610031 People's Republic of China
| | - He‐Zhong Jiang
- School of Life Science and EngineeringSouthwest Jiaotong University Chengdu 610031 People's Republic of China
| | - Hai‐Jun Leng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of PharmacyChengdu University of Traditional Chinese Medicine Chengdu 611137 People's Republic of China
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of AntibioticsChengdu University Chengdu 610052 People's Republic of China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of PharmacyChengdu University of Traditional Chinese Medicine Chengdu 611137 People's Republic of China
| | - Jun‐Long Li
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of AntibioticsChengdu University Chengdu 610052 People's Republic of China
| |
Collapse
|
25
|
Ling T, Miller DJ, Lang WH, Griffith E, Rodriguez-Cortes A, El Ayachi I, Palacios G, Min J, Miranda-Carboni G, Lee RE, Rivas F. Mechanistic Insight on the Mode of Action of Colletoic Acid. J Med Chem 2019; 62:6925-6940. [PMID: 31294974 DOI: 10.1021/acs.jmedchem.9b00187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The natural product colletoic acid (CA) is a selective inhibitor of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), which primarily converts cortisone to the active glucocorticoid (GC) cortisol. Here, CA's mode of action and its potential as a chemical tool to study intracellular GC signaling in adipogenesis are disclosed. 11β-HSD1 biochemical studies of CA indicated that its functional groups at C-1, C-4, and C-9 were important for enzymatic activity; an X-ray crystal structure of 11β-HSD1 bound to CA at 2.6 Å resolution revealed the nature of those interactions, namely, a close-fitting and favorable interactions between the constrained CA spirocycle and the catalytic triad of 11β-HSD1. Structure-activity relationship studies culminated in the development of a superior CA analogue with improved target engagement. Furthermore, we demonstrate that CA selectively inhibits preadipocyte differentiation through 11β-HSD1 inhibition, suppressing other relevant key drivers of adipogenesis (i.e., PPARγ, PGC-1α), presumably by negatively modulating the glucocorticoid signaling pathway. The combined findings provide an in-depth evaluation of the mode of action of CA and its potential as a tool compound to study adipose tissue and its implications in metabolic syndrome.
Collapse
Affiliation(s)
| | | | | | | | | | - Ikbale El Ayachi
- Department of Medicine , The University of Tennessee Health Science Center , Memphis , Tennessee 38163 , United States
| | | | | | - Gustavo Miranda-Carboni
- Department of Medicine , The University of Tennessee Health Science Center , Memphis , Tennessee 38163 , United States
| | | | | |
Collapse
|
26
|
Liu Z, Singh SB, Zheng Y, Lindblom P, Tice C, Dong C, Zhuang L, Zhao Y, Kruk BA, Lala D, Claremon DA, McGeehan GM, Gregg RD, Cain R. Discovery of Potent Inhibitors of 11β-Hydroxysteroid Dehydrogenase Type 1 Using a Novel Growth-Based Protocol of in Silico Screening and Optimization in CONTOUR. J Chem Inf Model 2019; 59:3422-3436. [PMID: 31355641 DOI: 10.1021/acs.jcim.9b00198] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Zhijie Liu
- Allergan Plc, 2525 Dupont Drive, Irvine, California 92612, United States
- Vitae Pharmaceuticals, Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Suresh B. Singh
- Vitae Pharmaceuticals, Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Yajun Zheng
- Allergan Plc, 2525 Dupont Drive, Irvine, California 92612, United States
- Vitae Pharmaceuticals, Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Peter Lindblom
- Vitae Pharmaceuticals, Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Colin Tice
- Vitae Pharmaceuticals, Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Chengguo Dong
- Allergan Plc, 2525 Dupont Drive, Irvine, California 92612, United States
- Vitae Pharmaceuticals, Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Linghang Zhuang
- Vitae Pharmaceuticals, Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Yi Zhao
- Allergan Plc, 2525 Dupont Drive, Irvine, California 92612, United States
- Vitae Pharmaceuticals, Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Barbara A. Kruk
- Vitae Pharmaceuticals, Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Deepak Lala
- Vitae Pharmaceuticals, Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - David A. Claremon
- Vitae Pharmaceuticals, Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Gerard M. McGeehan
- Vitae Pharmaceuticals, Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Richard D. Gregg
- Vitae Pharmaceuticals, Inc., 502 West Office Center Drive, Fort Washington, Pennsylvania 19034, United States
| | - Robert Cain
- Allergan Plc, 2525 Dupont Drive, Irvine, California 92612, United States
| |
Collapse
|
27
|
Phakdeeyothin K, Yotphan S. Metal-free regioselective direct thiolation of 2-pyridones. Org Biomol Chem 2019; 17:6432-6440. [PMID: 31218319 DOI: 10.1039/c9ob01061k] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A highly regioselective metal-free direct C-H thiolation of 2-pyridones with disulfides or thiols has been developed. A combination of persulfate and a commercially available halide source such as LiCl, NCS or I2 enables the successful direct incorporation of a sulfide moiety into the 5-position of pyridone under mild conditions, providing a useful and convenient approach for the preparation of a diverse array of 5-thio-substituted pyridones in moderate to excellent yields.
Collapse
Affiliation(s)
- Kunita Phakdeeyothin
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| | - Sirilata Yotphan
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| |
Collapse
|
28
|
Studzińska R, Kołodziejska R, Płaziński W, Kupczyk D, Kosmalski T, Jasieniecka K, Modzelewska-Banachiewicz B. Synthesis of the N-methyl Derivatives of 2-Aminothiazol-4(5H)-one and Their Interactions with 11βHSD1-Molecular Modeling and in Vitro Studies. Chem Biodivers 2019; 16:e1900065. [PMID: 31012543 DOI: 10.1002/cbdv.201900065] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/23/2019] [Indexed: 11/11/2022]
Abstract
11β-Hydroxysteroid dehydrogenase type 1 (11β-HSD1) is an enzyme that affects the body's cortisol levels. The inhibition of its activity can be used in the treatment of Cushing's syndrome, metabolic syndrome and type 2 diabetes. In this study, we synthesized new derivatives of 2-(methylamino)thiazol-4(5H)-one and tested their activity towards inhibition of 11β-HSD1 and its isoform - 11β-HSD2. The results were compared with the previously tested allyl derivatives. We found out that methyl derivatives are weaker inhibitors of 11β-HSD1 in comparison to their allyl analogs. Due to significant differences in the activity of the compounds, molecular modeling was performed, which was aimed at comparing the interactions between 11β-HSD1 and ligands differing by substituent at the amine group (allyl vs. methyl). Modeling showed that the absence of the allyl group can lead to the rotation of whole ligand molecule which affects its interaction with the enzyme.
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
| | - 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
| | - Wojciech Płaziński
- J. Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek Str. 8, 30-239, Cracow, 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
| | - Tomasz Kosmalski
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Jurasza 2, 85-089, Bydgoszcz, Poland
| | - Katarzyna Jasieniecka
- 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
|
29
|
Application of ELISA Technique and Human Microsomes in the Search for 11 β-Hydroxysteroid Dehydrogenase Inhibitors. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5747436. [PMID: 31214617 PMCID: PMC6535869 DOI: 10.1155/2019/5747436] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/02/2019] [Indexed: 01/18/2023]
Abstract
The metabolic syndrome is defined by impaired carbohydrate metabolism and lipid disorders and often accompanied by hypertension, all of which will lead to obesity and insulin resistance. Glucocorticoids play a regulatory role in the metabolism of proteins, lipids, and carbohydrates. There is growing evidence for a role of glucocorticoids in the development of the metabolic syndrome. The most important factor that regulates the access of endogenous glucocorticoids to receptors after release of glucocorticoids and their diffusion into the cytoplasm of target cells is the steroid metabolism involving a microsomal enzyme, 11β-hydroxysteroid dehydrogenase (11β-HSD). The changes in intracellular glucocorticoid metabolism in the pathogenesis of obesity indicate the participation of modulation by 11β-HSD1, which may represent a new therapeutic target for the treatment of diseases such as type 2 diabetes, visceral obesity, or atherosclerosis. The aim of our study was to determine the fast and effective method to assess inhibition activity of compounds in relation with 11β-hydroxysteroid dehydrogenase. The material for this study was human liver and kidney microsomes. In this study we used ELISA technique using 96-well microplates coated with antibodies which were specific for analyzed enzymes. The method can quickly and efficiently measure the inhibition of both 11β-HSD1 and 11β-HSD2. This method can be used to search for and determine inhibitors of this enzyme. Cortisone and cortisol were used as the substrates for corresponding enzyme assays. Furthermore, 3-N-allyl-2-thiouracil derivatives were used by us for comparison purposes in developing the method, although, due to their structure, those derivatives have not previously been considered as potential inhibitors of 11β-HSD1. 3-N-Allyl-2-thiouracil derivatives are a group worth considering, because by modifying their structure (e.g., by introducing other substituents into the pyrimidine ring) it will be possible to obtain an increase in the activity of compounds in this regard. In conclusion, this study shows an efficient and fast method of determining inhibition activity of compounds in relation with 11β-hydroxysteroid dehydrogenase.
Collapse
|
30
|
Sandström J, Kratschmar DV, Broyer A, Poirot O, Marbet P, Chantong B, Zufferey F, Dos Santos T, Boccard J, Chrast R, Odermatt A, Monnet-Tschudi F. In vitro models to study insulin and glucocorticoids modulation of trimethyltin (TMT)-induced neuroinflammation and neurodegeneration, and in vivo validation in db/db mice. Arch Toxicol 2019; 93:1649-1664. [PMID: 30993381 DOI: 10.1007/s00204-019-02455-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023]
Abstract
Brain susceptibility to a neurotoxic insult may be increased in a compromised health status, such as metabolic syndrome. Both metabolic syndrome and exposure to trimethyltin (TMT) are known to promote neurodegeneration. In combination the two factors may elicit additive or compensatory/regulatory mechanisms. Combined effects of TMT exposure (0.5-1 μM) and mimicked metabolic syndrome-through modulation of insulin and glucocorticoid (GC) levels-were investigated in three models: tridimensional rat brain cell cultures for neuron-glia effects; murine microglial cell line BV-2 for a mechanistic analysis of microglial reactivity; and db/db mice as an in vivo model of metabolic syndrome. In 3D cultures, low insulin condition significantly exacerbated TMT's effect on GABAergic neurons and promoted TMT-induced neuroinflammation, with increased expression of cytokines and of the regulator of intracellular GC activity, 11β-hydroxysteroid dehydrogenase 1 (11β-Hsd1). Microglial reactivity increased upon TMT exposure in medium combining low insulin and high GC. These results were corroborated in BV-2 microglial cells where lack of insulin exacerbated the TMT-induced increase in 11β-Hsd1 expression. Furthermore, TMT-induced microglial reactivity seems to depend on mineralocorticoid receptor activation. In diabetic BKS db mice, a discrete exacerbation of TMT neurotoxic effects on GABAergic neurons was observed, together with an increase of interleukin-6 (IL-6) and of basal 11β-Hsd1 expression as compared to controls. These results suggest only minor additive effects of the two brain insults, neurotoxicant TMT exposure and metabolic syndrome conditions, where 11β-Hsd1 appears to play a key role in the regulation of neuroinflammation and of its protective or neurodegenerative consequences.
Collapse
Affiliation(s)
- Jenny Sandström
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 7, 1005, Lausanne, Switzerland.,Swiss Centre for Applied Human Toxicology, Basel, Switzerland
| | - Denise V Kratschmar
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.,Swiss Centre for Applied Human Toxicology, Basel, Switzerland
| | - Alexandra Broyer
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 7, 1005, Lausanne, Switzerland
| | - Olivier Poirot
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
| | - Philippe Marbet
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Boonrat Chantong
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Fanny Zufferey
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 7, 1005, Lausanne, Switzerland.,Swiss Centre for Applied Human Toxicology, Basel, Switzerland
| | - Tania Dos Santos
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 7, 1005, Lausanne, Switzerland
| | - Julien Boccard
- Swiss Centre for Applied Human Toxicology, Basel, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Roman Chrast
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland.,Department of Neuroscience and Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.,Swiss Centre for Applied Human Toxicology, Basel, Switzerland
| | - Florianne Monnet-Tschudi
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 7, 1005, Lausanne, Switzerland. .,Swiss Centre for Applied Human Toxicology, Basel, Switzerland.
| |
Collapse
|
31
|
Bellaire S, Walzer M, Wang T, Krauwinkel W, Yuan N, Marek GJ. Safety, Pharmacokinetics, and Pharmacodynamics of ASP3662, a Novel 11β-Hydroxysteroid Dehydrogenase Type 1 Inhibitor, in Healthy Young and Elderly Subjects. Clin Transl Sci 2019; 12:291-301. [PMID: 30740895 PMCID: PMC6510378 DOI: 10.1111/cts.12618] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/31/2018] [Indexed: 11/28/2022] Open
Abstract
Inhibition of the enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) represents a potential mechanism for improving pain conditions. ASP3662 is a potent and selective inhibitor of 11β-HSD1. Two phase I clinical studies were conducted to assess the safety, tolerability, pharmacokinetics (PKs), and pharmacodynamics (PDs) of single and multiple ascending doses of ASP3662 in healthy young and elderly non-Japanese and young Japanese subjects. Nonlinear, more than dose-proportional PKs were observed for ASP3662 after single-dose administration, particularly at lower doses (≤ 6 mg); the PKs at steady state were dose proportional, although the time to ASP3662 steady state was dose dependent at lower doses (≤ 2 mg). Similar PKs were observed among young Japanese, young non-Japanese, and elderly non-Japanese subjects. Specific inhibition of 11β-HSD1 occurred after both single and multiple doses of ASP3662. A marked dissociation between PKs and PDs was observed after single but not multiple doses of ASP3662. ASP3662 was generally safe and well tolerated.
Collapse
Affiliation(s)
- Susan Bellaire
- Formerly with Astellas Pharma Europe BV, Leiden, The Netherlands
| | - Mark Walzer
- Astellas Pharma Global Development, Northbrook, Illinois, USA
| | - Tianli Wang
- Formerly with Astellas Pharma, Inc., Northbrook, Illinois, USA
| | | | - Nancy Yuan
- Formerly with Astellas Pharma, Inc., Northbrook, Illinois, USA
| | | |
Collapse
|
32
|
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
|
33
|
Li J, Kennedy LJ, Walker SJ, Wang H, Li JJ, Hong Z, O’Connor SP, Ye XY, Chen S, Wu S, Yoon DS, Nayeem A, Camac DM, Ramamurthy V, Morin PE, Sheriff S, Wang M, Harper TW, Golla R, Seethala R, Harrity T, Ponticiello RP, Morgan NN, Taylor JR, Zebo R, Maxwell B, Moulin F, Gordon DA, Robl JA. Discovery of Clinical Candidate BMS-823778 as an Inhibitor of Human 11β-Hydroxysteroid Dehydrogenase Type 1 (11β-HSD-1). ACS Med Chem Lett 2018; 9:1170-1174. [PMID: 30613321 DOI: 10.1021/acsmedchemlett.8b00307] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/13/2018] [Indexed: 12/18/2022] Open
Abstract
BMS-823778 (2), a 1,2,4-triazolopyridinyl-methanol derived analog, was identified as a potent and selective inhibitor of human 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD-1) enzyme (IC50 = 2.3 nM) with >10,000-fold selectivity over 11β-HSD-2. Compound 2 exhibits robust acute pharmacodynamic effects in cynomolgus monkeys (ED50 = 0.6 mg/kg) and in diet-induced obese (DIO) mice (ED50 = 34 mg/kg). Compound 2 also showed excellent inhibition in an ex vivo adipose DIO mouse model (ED50 = 5.2 mg/kg). Oral bioavailability ranges from 44% to 100% in preclinical species. Its favorable development properties, pharmacokinetics, high adipose-to-plasma concentration ratio, and preclinical pharmacology profile have prompted the evaluation of 2 for the treatment of type 2 diabetes and metabolic syndrome in phase 2 clinical trials.
Collapse
Affiliation(s)
- Jun Li
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Lawrence J. Kennedy
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Steven J. Walker
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Haixia Wang
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - James J. Li
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Zhenqiu Hong
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Stephen P. O’Connor
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Xiang-yang Ye
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Stephanie Chen
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Shung Wu
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - David S. Yoon
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Akbar Nayeem
- Research & Development, Bristol-Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Daniel M. Camac
- Research & Development, Bristol-Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Vidhyashankar Ramamurthy
- Research & Development, Bristol-Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Paul E. Morin
- Research & Development, Bristol-Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Steven Sheriff
- Research & Development, Bristol-Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Mengmeng Wang
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Timothy W. Harper
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Rajasree Golla
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Ramakrishna Seethala
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Thomas Harrity
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Randolph P. Ponticiello
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Nathan N. Morgan
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Joseph R. Taylor
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Rachel Zebo
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Brad Maxwell
- Research & Development, Bristol-Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Frederick Moulin
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - David A. Gordon
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| | - Jeffrey A. Robl
- Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543, United States
| |
Collapse
|
34
|
White PC. Alterations of Cortisol Metabolism in Human Disorders. Horm Res Paediatr 2018; 89:320-330. [PMID: 29843121 DOI: 10.1159/000485508] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/21/2017] [Indexed: 11/19/2022] Open
Abstract
The interconversion of active and inactive corticosteroids - cortisol and cortisone, respectively, in humans - is modulated by isozymes of 11β-hydroxysteroid dehydrogenase (11-HSD). Studies of this process have provided crucial insights into glucocorticoid effects in a wide variety of tissues. The 11-HSD1 isozyme functions mainly as an oxoreductase (cortisone to cortisol) and is expressed at high levels in the liver and other glucocorticoid target tissues. Because it is required for full physiological effects of cortisol, it has emerged as a drug target for metabolic syndrome and type 2 diabetes. Mutations in the corresponding HSD11B1 gene, or in the H6PD gene encoding hexose-6-phosphate dehydrogenase (which supplies the NADPH required for the oxoreductase activity of 11-HSD1), cause apparent cortisone reductase deficiency, a rare syndrome of adrenocortical hyperactivity and hyperandrogenism. In contrast, the 11-HSD2 isozyme functions as a dehydrogenase (cortisol to cortisone) and is expressed mainly in mineralocorticoid target tissues, where it bars access of cortisol to the mineralocorticoid receptor. Mutations in the HSD11B2 gene encoding 11-HSD2 cause the syndrome of apparent mineralocorticoid excess, a severe form of familial hypertension. The role of this enzyme in the pathogenesis of common forms of low-renin hypertension remains uncertain.
Collapse
|
35
|
Boudon S, Heidl M, Vuorinen A, Wandeler E, Campiche R, Odermatt A, Jackson E. Design, synthesis, and biological evaluation of novel selective peptide inhibitors of 11β-hydroxysteroid dehydrogenase 1. Bioorg Med Chem 2018; 26:5128-5139. [PMID: 30245006 DOI: 10.1016/j.bmc.2018.09.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 11/17/2022]
Abstract
The enzyme 11β-HSD1 plays a crucial role in the tissue-specific regulation of cortisol levels and it has been associated with various diseases. Inhibition of 11β-HSD1 is an attractive intervention strategy and the discovery of novel selective 11β-HSD1 inhibitors is of high relevance. In this study, we identified and evaluated a new series of selective peptide 11β-HSD1 inhibitors with potential for skin care applications. This novel scaffold was designed with the aid of molecular modeling and two previously reported inhibitors. SAR optimization yielded highly active peptides (IC50 below 400 nM) that were inactive at 1 µM concentration against structurally related enzymes (11β-HSD2, 17β-HSD1 and 17β-HSD2). The best performing peptides inhibited the conversion of cortisone into cortisol in primary human keratinocytes and the most active compound, 5d, was further shown to reverse cortisone-induced collagen damage in human ex-vivo tissue.
Collapse
Affiliation(s)
- Stephanie Boudon
- DSM Nutritional Products Ltd., Wurmisweg 576, 4303 Kaiseraugst, Switzerland
| | - Marc Heidl
- DSM Nutritional Products Ltd., Wurmisweg 576, 4303 Kaiseraugst, Switzerland
| | - Anna Vuorinen
- Division of Molecular and Systems Toxicology, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Eliane Wandeler
- DSM Nutritional Products Ltd., Wurmisweg 576, 4303 Kaiseraugst, Switzerland
| | - Remo Campiche
- DSM Nutritional Products Ltd., Wurmisweg 576, 4303 Kaiseraugst, Switzerland
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| | - Eileen Jackson
- DSM Nutritional Products Ltd., Wurmisweg 576, 4303 Kaiseraugst, Switzerland.
| |
Collapse
|
36
|
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
|
37
|
Mosquera C, Panay AJ, Montoya G. Pentacyclic Triterpenes from Cecropia telenitida Can Function as Inhibitors of 11β-Hydroxysteroid Dehydrogenase Type 1. Molecules 2018; 23:molecules23061444. [PMID: 29899225 PMCID: PMC6099733 DOI: 10.3390/molecules23061444] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/05/2018] [Accepted: 06/08/2018] [Indexed: 11/16/2022] Open
Abstract
Plant extracts from the genus Cecropia have been used by Latin-American traditional medicine to treat metabolic disorders and diabetes. Previous results have shown that roots of Cecropia telenitida contain pentacyclic triterpenes and these molecules display a hypoglycemic effect in an insulin-resistant murine model. The pharmacological target of these molecules, however, remains unknown. Several lines of evidence indicate that pentacyclic triterpenes inhibit the 11β-hydroxysteroid dehydrogenase type 1 enzyme, which highlights the potential use of this type of natural product as phytotherapeutic or botanical dietary supplements. The main goal of the study was the evaluation of the inhibitory effect of Cecropia telenitida molecules on 11β-hydroxysteroid dehydrogenase type 1 enzyme activity. A pre-fractionated chemical library was obtained from the roots of Cecropia telenitida using several automated chromatography separation steps and a homogeneous time resolved fluorescence assay was used for the bio-guided isolation of inhibiting molecules. The screening of a chemical library consisting of 125 chemical purified fractions obtained from Cecropia telenitida roots identified one fraction displaying 82% inhibition of the formation of cortisol by the 11β-hydroxysteroid dehydrogenase type 1 enzyme. Furthermore, a molecule displaying IC50 of 0.95 ± 0.09 µM was isolated from this purified fraction and structurally characterized, which confirms that a pentacyclic triterpene scaffold was responsible for the observed inhibition. Our results support the hypothesis that pentacyclic triterpene molecules from Cecropia telenitida can inhibit 11β-hydroxysteroid dehydrogenase type 1 enzyme activity. These findings highlight the potential ethnopharmacological use of plants from the genus Cecropia for the treatment of metabolic disorders and diabetes.
Collapse
Affiliation(s)
- Catalina Mosquera
- Department of Chemical Sciences, Faculty of Natural Sciences, Universidad Icesi, Cali, Valle del Cauca 760031, Colombia.
| | - Aram J Panay
- Department of Chemical Sciences, Faculty of Natural Sciences, Universidad Icesi, Cali, Valle del Cauca 760031, Colombia.
| | - Guillermo Montoya
- Department of Pharmaceutical Sciences, Faculty of Natural Sciences, Universidad Icesi, Calle 18 # 122⁻135, Cali, Valle del Cauca 760031, Colombia.
| |
Collapse
|
38
|
Yang Y, Fei C, Wang K, Liu B, Jiang D, Yin B. Access to Densely Functionalized Chalcone Derivatives with a 2-Pyridone Subunit via Pd/Cu-Catalyzed Oxidative Furan–Yne Cyclization of N-(2-Furanylmethyl) Alkynamides under Air. Org Lett 2018; 20:2273-2277. [DOI: 10.1021/acs.orglett.8b00618] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Yongjie Yang
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, P. R. China
| | - ChengCheng Fei
- Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, Laboratory of Insect Toxicology, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Kai Wang
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510006, P. R. China
| | - Bo Liu
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510006, P. R. China
| | - Dingxin Jiang
- Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, Laboratory of Insect Toxicology, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Biaolin Yin
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, P. R. China
| |
Collapse
|
39
|
Zhang L, Zheng X, Chen J, Cheng K, Jin L, Jiang X, Yu C. Ru(ii)-Catalyzed C6-selective C–H amidation of 2-pyridones. Org Chem Front 2018. [DOI: 10.1039/c8qo00795k] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
An efficient Ru-catalyzed C6 site-selective amidation of 2-pyridones has been accomplished with dioxazolone under mild conditions.
Collapse
Affiliation(s)
- Lei Zhang
- College of Pharmaceutical Sciences
- Zhejiang University of Technology
- Hangzhou 310014
- P. R. China
| | - Xiangyun Zheng
- College of Pharmaceutical Sciences
- Zhejiang University of Technology
- Hangzhou 310014
- P. R. China
| | - Jinkang Chen
- College of Pharmaceutical Sciences
- Zhejiang University of Technology
- Hangzhou 310014
- P. R. China
| | - Kang Cheng
- College of Pharmaceutical Sciences
- Zhejiang University of Technology
- Hangzhou 310014
- P. R. China
| | - Licheng Jin
- College of Pharmaceutical Sciences
- Zhejiang University of Technology
- Hangzhou 310014
- P. R. China
| | - Xinpeng Jiang
- College of Pharmaceutical Sciences
- Zhejiang University of Technology
- Hangzhou 310014
- P. R. China
| | - Chuanming Yu
- College of Pharmaceutical Sciences
- Zhejiang University of Technology
- Hangzhou 310014
- P. R. China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals
| |
Collapse
|
40
|
Wnuk A, Kajta M. Steroid and Xenobiotic Receptor Signalling in Apoptosis and Autophagy of the Nervous System. Int J Mol Sci 2017; 18:ijms18112394. [PMID: 29137141 PMCID: PMC5713362 DOI: 10.3390/ijms18112394] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/06/2017] [Accepted: 11/09/2017] [Indexed: 12/15/2022] Open
Abstract
Apoptosis and autophagy are involved in neural development and in the response of the nervous system to a variety of insults. Apoptosis is responsible for cell elimination, whereas autophagy can eliminate the cells or keep them alive, even in conditions lacking trophic factors. Therefore, both processes may function synergistically or antagonistically. Steroid and xenobiotic receptors are regulators of apoptosis and autophagy; however, their actions in various pathologies are complex. In general, the estrogen (ER), progesterone (PR), and mineralocorticoid (MR) receptors mediate anti-apoptotic signalling, whereas the androgen (AR) and glucocorticoid (GR) receptors participate in pro-apoptotic pathways. ER-mediated neuroprotection is attributed to estrogen and selective ER modulators in apoptosis- and autophagy-related neurodegenerative diseases, such as Alzheimer’s and Parkinson’s diseases, stroke, multiple sclerosis, and retinopathies. PR activation appeared particularly effective in treating traumatic brain and spinal cord injuries and ischemic stroke. Except for in the retina, activated GR is engaged in neuronal cell death, whereas MR signalling appeared to be associated with neuroprotection. In addition to steroid receptors, the aryl hydrocarbon receptor (AHR) mediates the induction and propagation of apoptosis, whereas the peroxisome proliferator-activated receptors (PPARs) inhibit this programmed cell death. Most of the retinoid X receptor-related xenobiotic receptors stimulate apoptotic processes that accompany neural pathologies. Among the possible therapeutic strategies based on targeting apoptosis via steroid and xenobiotic receptors, the most promising are the selective modulators of the ER, AR, AHR, PPARγ agonists, flavonoids, and miRNAs. The prospective therapies to overcome neuronal cell death by targeting autophagy via steroid and xenobiotic receptors are much less recognized.
Collapse
Affiliation(s)
- Agnieszka Wnuk
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Smetna Street 12, 31-343 Krakow, Poland.
| | - Małgorzata Kajta
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Smetna Street 12, 31-343 Krakow, Poland.
| |
Collapse
|
41
|
Loerz C, Staab-Weijnitz C, Huebbe P, Giller K, Metges C, Rimbach G, Maser E. Regulation of 11β-hydroxysteroid dehydrogenase type 1 following caloric restriction and re-feeding is species dependent. Chem Biol Interact 2017; 276:95-104. [DOI: 10.1016/j.cbi.2017.02.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/17/2017] [Accepted: 02/26/2017] [Indexed: 01/22/2023]
|
42
|
Shao LD, Bao Y, Shen Y, Su J, Leng Y, Zhao QS. Synthesis of selective 11β-HSD1 inhibitors based on dammarane scaffold. Eur J Med Chem 2017; 135:324-338. [PMID: 28458137 DOI: 10.1016/j.ejmech.2017.04.059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/21/2017] [Accepted: 04/22/2017] [Indexed: 11/26/2022]
Abstract
Inspired by natural 11β-HSD1 inhibitors hupehenols A - E, a ring-focused strategy was applied for the synthesis of 35 structurally diverse dammarane-type derivatives. These derivatives were effectively prepared from protopanaxadiol based on the modification of rings A and D. Among these compounds, ten were identified as selective 11β-HSD1 inhibitors (IC50 range: 101-1047 nM, SI range: 8-169) which exhibited inhibitory activities against human or mouse 11β-HSD1. Otherwise, we found 23a could selectively inhibit both human and mouse 11β-HSD1 with IC50 value of 994 and 213 nM (SI > 10 and > 47), respectively. Additionally, the molecular modelling results of 23a docking into the human and mouse 11β-HSD1 were in good agreement with the results from the enzyme inhibitory experiment. Moreover, valuable structural-activity relationship (SAR) information of dammarane-type 11β-HSD1 inhibitor was summarized.
Collapse
Affiliation(s)
- Li-Dong Shao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650204, China
| | - Ying Bao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650204, China; University of Chinese Academy of Science, Beijing 100049, China
| | - Yu Shen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jia Su
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650204, China
| | - Ying Leng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Qin-Shi Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650204, China.
| |
Collapse
|
43
|
Choi KJ, Na YJ, Park SB, Jung WH, Sung HR, Kim KY. Carbenoxolone prevents chemical eye ischemia-reperfusion-induced cell death via 11β-hydroxysteroid dehydrogenase type 1 inhibition. Pharmacol Res 2017; 123:62-72. [PMID: 28687341 DOI: 10.1016/j.phrs.2017.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 04/07/2017] [Accepted: 07/03/2017] [Indexed: 01/09/2023]
Abstract
Glaucoma is one of the leading causes of preventable blindness diseases, affecting more than 2 million people in the United States. Recently, 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) inhibitors were found to exert preventive effects against glaucoma. Therefore, we investigated whether carbenoxolone (CBX), an 11β-HSD1 inhibitor, prevents chemical ischemia-reperfusion-induced cell death in human trabecular meshwork (HTM) cells. The present study demonstrated that CBX inhibited cell death caused by iodoacetic acid (IAA)-induced ischemia-reperfusion, and its effect was associated with the inhibition of 11β-HSD1 expression and activity. Furthermore, CBX reversed the IAA-induced structural damage on filamentous actin in HTM cells. In IAA-treated cells, the levels of 11β-HSD1 and the apoptosis-related factors Bax and FASL were increased throughout the reperfusion period, and CBX was able to attenuate the expression of 11β-HSD1 and the apoptosis-related factors. CBX also effectively suppressed IAA-induced intracellular ROS formation and cytochrome c release, which are involved in the mitochondrial apoptosis pathway. In addition, IAA-induced chemical ischemia-reperfusion stimulated TNF-α expression and NF-κB p65 phosphorylation, and these effects were attenuated by CBX. 11β-HSD1 RNAi also suppressed IAA-induced cell apoptosis via reduction of oxidative stress and inhibition of the pro-inflammatory pathway. Taken together, the present study demonstrated that the inhibition of 11β-HSD1 protected the TM against chemical ischemia-reperfusion injury, suggesting that the use of 11β-HSD1 inhibitors could be a useful strategy for glaucoma therapy.
Collapse
Affiliation(s)
- Kyoung-Jin Choi
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, P.O. Box 107, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Yoon-Ju Na
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, P.O. Box 107, Yuseong-gu, Daejeon 34114, Republic of Korea; Department of New Drug Discovery and Development, Chungnam National University, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Sung Bum Park
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, P.O. Box 107, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Won Hoon Jung
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, P.O. Box 107, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Hye-Rim Sung
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, P.O. Box 107, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Ki Young Kim
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, P.O. Box 107, Yuseong-gu, Daejeon 34114, Republic of Korea; Department of New Drug Discovery and Development, Chungnam National University, Yuseong-gu, Daejeon 34134, Republic of Korea.
| |
Collapse
|
44
|
Zhuang L, Tice CM, Xu Z, Zhao W, Cacatian S, Ye YJ, Singh SB, Lindblom P, McKeever BM, Krosky PM, Zhao Y, Lala D, Kruk BA, Meng S, Howard L, Johnson JA, Bukhtiyarov Y, Panemangalore R, Guo J, Guo R, Himmelsbach F, Hamilton B, Schuler-Metz A, Schauerte H, Gregg R, McGeehan GM, Leftheris K, Claremon DA. Discovery of BI 135585, an in vivo efficacious oxazinanone-based 11β hydroxysteroid dehydrogenase type 1 inhibitor. Bioorg Med Chem 2017; 25:3649-3657. [DOI: 10.1016/j.bmc.2017.04.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 04/24/2017] [Accepted: 04/26/2017] [Indexed: 11/16/2022]
|
45
|
Beck KR, Kaserer T, Schuster D, Odermatt A. Virtual screening applications in short-chain dehydrogenase/reductase research. J Steroid Biochem Mol Biol 2017; 171:157-177. [PMID: 28286207 PMCID: PMC6831487 DOI: 10.1016/j.jsbmb.2017.03.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/06/2017] [Accepted: 03/08/2017] [Indexed: 02/06/2023]
Abstract
Several members of the short-chain dehydrogenase/reductase (SDR) enzyme family play fundamental roles in adrenal and gonadal steroidogenesis as well as in the metabolism of steroids, oxysterols, bile acids, and retinoids in peripheral tissues, thereby controlling the local activation of their cognate receptors. Some of these SDRs are considered as promising therapeutic targets, for example to treat estrogen-/androgen-dependent and corticosteroid-related diseases, whereas others are considered as anti-targets as their inhibition may lead to disturbances of endocrine functions, thereby contributing to the development and progression of diseases. Nevertheless, the physiological functions of about half of all SDR members are still unknown. In this respect, in silico tools are highly valuable in drug discovery for lead molecule identification, in toxicology screenings to facilitate the identification of hazardous chemicals, and in fundamental research for substrate identification and enzyme characterization. Regarding SDRs, computational methods have been employed for a variety of applications including drug discovery, enzyme characterization and substrate identification, as well as identification of potential endocrine disrupting chemicals (EDC). This review provides an overview of the efforts undertaken in the field of virtual screening supported identification of bioactive molecules in SDR research. In addition, it presents an outlook and addresses the opportunities and limitations of computational modeling and in vitro validation methods.
Collapse
Affiliation(s)
- Katharina R Beck
- Swiss Center for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Teresa Kaserer
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), Computer Aided Molecular Design Group, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Daniela Schuster
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), Computer Aided Molecular Design Group, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| | - Alex Odermatt
- Swiss Center for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| |
Collapse
|
46
|
Ye XY, Chen SY, Wu S, Yoon DS, Wang H, Hong Z, O'Connor SP, Li J, Li JJ, Kennedy LJ, Walker SJ, Nayeem A, Sheriff S, Camac DM, Ramamurthy V, Morin PE, Zebo R, Taylor JR, Morgan NN, Ponticiello RP, Harrity T, Apedo A, Golla R, Seethala R, Wang M, Harper TW, Sleczka BG, He B, Kirby M, Leahy DK, Li J, Hanson RL, Guo Z, Li YX, DiMarco JD, Scaringe R, Maxwell B, Moulin F, Barrish JC, Gordon DA, Robl JA. Discovery of Clinical Candidate 2-((2S,6S)-2-Phenyl-6-hydroxyadamantan-2-yl)-1-(3'-hydroxyazetidin-1-yl)ethanone [BMS-816336], an Orally Active Novel Selective 11β-Hydroxysteroid Dehydrogenase Type 1 Inhibitor. J Med Chem 2017; 60:4932-4948. [PMID: 28537398 DOI: 10.1021/acs.jmedchem.7b00211] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BMS-816336 (6n-2), a hydroxy-substituted adamantyl acetamide, has been identified as a novel, potent inhibitor against human 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) enzyme (IC50 3.0 nM) with >10000-fold selectivity over human 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2). 6n-2 exhibits a robust acute pharmacodynamic effect in cynomolgus monkeys (ED50 0.12 mg/kg) and in DIO mice. It is orally bioavailable (%F ranges from 20 to 72% in preclinical species) and has a predicted pharmacokinetic profile of a high peak to trough ratio and short half-life in humans. This ADME profile met our selection criteria for once daily administration, targeting robust inhibition of 11β-HSD1 enzyme for the first 12 h period after dosing followed by an "inhibition holiday" so that the potential for hypothalamic-pituitary-adrenal (HPA) axis activation might be mitigated. 6n-2 was found to be well-tolerated in phase 1 clinical studies and represents a potential new treatment for type 2 diabetes, metabolic syndrome, and other human diseases modulated by glucocorticoid control.
Collapse
Affiliation(s)
- Xiang-Yang Ye
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Stephanie Y Chen
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Shung Wu
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - David S Yoon
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Haixia Wang
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Zhenqiu Hong
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Stephen P O'Connor
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Jun Li
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - James J Li
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Lawrence J Kennedy
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Steven J Walker
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Akbar Nayeem
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Steven Sheriff
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Daniel M Camac
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Vidyhashankar Ramamurthy
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Paul E Morin
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Rachel Zebo
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Joseph R Taylor
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Nathan N Morgan
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Randolph P Ponticiello
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Thomas Harrity
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Atsu Apedo
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Rajasree Golla
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Ramakrishna Seethala
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Mengmeng Wang
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Timothy W Harper
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Bogdan G Sleczka
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Bin He
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Mark Kirby
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - David K Leahy
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Jianqing Li
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Ronald L Hanson
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Zhiwei Guo
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Yi-Xin Li
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - John D DiMarco
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Raymond Scaringe
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Brad Maxwell
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Frederick Moulin
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Joel C Barrish
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - David A Gordon
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Jeffrey A Robl
- Discovery Chemistry, ‡Pharmaceutical Candidate Optimization, §Computer-Assisted Drug Design, ∥Metabolic Diseases Biology, ⊥Lead Evaluation, #Process Chemistry, ∇Chemical Synthesis, ○Discovery Toxicology, Research and Development, Bristol-Myers Squibb , 350 Carter Road, Princeton, New Jersey 08540, United States.,Molecular Structure and Design, ¶Protein Science, +Solid State Chemistry, Research and Development, Bristol-Myers Squibb , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| |
Collapse
|
47
|
Affiliation(s)
- Cristina M. Alcántara
- Organic & Pharmaceutical Chemistry Department, Complutense University of Madrid, Madrid, Spain
| | - Andrés R. Alcántara
- Biotransformations Group, Organic & Pharmaceutical Chemistry Department, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
48
|
Novel 11β-hydroxysteroid dehydrogenase 1 inhibitors reduce cortisol levels in keratinocytes and improve dermal collagen content in human ex vivo skin after exposure to cortisone and UV. PLoS One 2017; 12:e0171079. [PMID: 28152550 PMCID: PMC5289826 DOI: 10.1371/journal.pone.0171079] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 01/17/2017] [Indexed: 01/03/2023] Open
Abstract
Activity and selectivity assessment of new bi-aryl amide 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1) inhibitors, prepared in a modular manner via Suzuki cross-coupling, are described. Several compounds inhibiting 11β-HSD1 at nanomolar concentrations were identified. Compounds 2b, 3e, 7b and 12e were shown to selectively inhibit 11β-HSD1 over 11β-HSD2, 17β-HSD1 and 17β-HSD2. These inhibitors also potently inhibited 11β-HSD1 activity in intact HEK-293 cells expressing the recombinant enzyme and in intact primary human keratinocytes expressing endogenous 11β-HSD1. Moreover, compounds 2b, 3e and 12e were tested for their activity in human skin biopsies. They were able to prevent, at least in part, both the cortisone- and the UV-mediated decreases in collagen content. Thus, inhibition of 11β-HSD1 by these compounds can be further investigated to delay or prevent UV-mediated skin damage and skin aging.
Collapse
|
49
|
Chourasiya SS, Patel DR, Nagaraja CM, Chakraborti AK, Bharatam PV. Sulfonamide vs. sulfonimide: tautomerism and electronic structure analysis of N-heterocyclic arenesulfonamides. NEW J CHEM 2017. [DOI: 10.1039/c7nj01353a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Experimental and computational studies suggest a preference toward the sulfonimide tautomer in N-heterocyclic arenesulfonamide.
Collapse
Affiliation(s)
- Sumit S. Chourasiya
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research
- Punjab – 160 062
- India
| | - Dhara R. Patel
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research
- Punjab – 160 062
- India
| | - C. M. Nagaraja
- Department of Chemistry
- Indian Institute of Technology (IIT) Ropar
- Roopnagar – 140 001
- India
| | - Asit K. Chakraborti
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research
- Punjab – 160 062
- India
| | - Prasad V. Bharatam
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research
- Punjab – 160 062
- India
| |
Collapse
|
50
|
Kasimov EM, Aghaeva FA. [Cortisol levels in plasma and aqueous humour of patients with steroid induced and other glaucomas]. Vestn Oftalmol 2017; 133:39-45. [PMID: 28524138 DOI: 10.17116/oftalma2017133239-44] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
UNLABELLED The paper describes the role of endogenous glucocorticoids in the pathogenesis of open-angle glaucoma. As to steroid-induced glaucoma (SG), however, indicators of systemic and local cortisol metabolism are yet underinvestigated. Aim - to determine and compare systemic (plasma) and local (aqueous humour) cortisol concentrations in patients with SG and other types of glaucoma (OTG). MATERIAL AND METHODS A prospective analysis was conducted in 64 patients who underwent plasma and aqueous humor sampling. All the patients were divided into 4 groups: group 1 - 11 patients with steroid induced ocular hypertension (SOH) and SG; group 2 - 31 patients with OTG; group 3 - 10 patients on steroid therapy (ST) without a so called 'steroid response'; group 4 - the controls (12 patients with cataract). RESULTS The mean cortisol levels in plasma and aqueous humour were the following: 177.5±29.4 ng/ml and 53.1±12.2 ng/ml in patients with SOH and SG; 277.7±33.6 ng/ml and 44.6±4.4 ng/ml in patients with OTG; 125.2±28.1 ng/ml in patients on ST; 147.2±19.1 ng/ml and 29.8±3.2 ng/ml in the controls. The highest plasma and aqueous humour cortisol levels were found in groups 2 and 1, respectively. CONCLUSION Plasma and aqueous humour levels of cortisol appeared to be higher in glaucoma patients than in the controls. Taking into account the fact that plasma cortisol levels are higher in steroid responders, it is recommended that all patients on ST undergo cortisol testing.
Collapse
Affiliation(s)
- E M Kasimov
- National Centre of Ophthalmology named after acad. Zarifa Aliyeva, 32/15 Javadkhan St., Baku, Azerbaijan, AZ 1114
| | - F A Aghaeva
- National Centre of Ophthalmology named after acad. Zarifa Aliyeva, 32/15 Javadkhan St., Baku, Azerbaijan, AZ 1114
| |
Collapse
|