1
|
Kuralay A, McDonough MC, Resch JM. Control of sodium appetite by hindbrain aldosterone-sensitive neurons. Mol Cell Endocrinol 2024; 592:112323. [PMID: 38936597 PMCID: PMC11381173 DOI: 10.1016/j.mce.2024.112323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/25/2024] [Indexed: 06/29/2024]
Abstract
Mineralocorticoids play a key role in hydromineral balance by regulating sodium retention and potassium wasting. Through favoring sodium, mineralocorticoids can cause hypertension from fluid overload under conditions of hyperaldosteronism, such as aldosterone-secreting tumors. An often-overlooked mechanism by which aldosterone functions to increase sodium is through stimulation of salt appetite. To drive sodium intake, aldosterone targets neurons in the hindbrain which uniquely express 11β-hydroxysteroid dehydrogenase type 2 (HSD2). This enzyme is a necessary precondition for aldosterone-sensing cells as it metabolizes glucocorticoids - preventing their activation of the mineralocorticoid receptor. In this review, we will consider the role of hindbrain HSD2 neurons in regulating sodium appetite by discussing HSD2 expression in the brain, regulation of hindbrain HSD2 neuron activity, and the circuitry mediating the effects of these aldosterone-sensitive neurons. Reducing the activity of hindbrain HSD2 neurons may be a viable strategy to reduce sodium intake and cardiovascular risk, particularly for conditions of hyperaldosteronism.
Collapse
Affiliation(s)
- Ahmet Kuralay
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, USA
| | - Miriam C McDonough
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA; Molecular Medicine Graduate Program, University of Iowa, Iowa City, IA, USA
| | - Jon M Resch
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, USA; Molecular Medicine Graduate Program, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
2
|
Ha K, Mundt-Machado N, Bisignano P, Pinedo A, Raleigh DR, Loeb G, Reiter JF, Cao E, Delling M. Cilia-enriched oxysterol 7β,27-DHC is required for polycystin ion channel activation. Nat Commun 2024; 15:6468. [PMID: 39085216 PMCID: PMC11291729 DOI: 10.1038/s41467-024-50318-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 06/28/2024] [Indexed: 08/02/2024] Open
Abstract
Polycystin-1 (PC-1) and PC-2 form a heteromeric ion channel complex that is abundantly expressed in primary cilia of renal epithelial cells. This complex functions as a non-selective cation channel, and mutations within the polycystin complex cause autosomal dominant polycystic kidney disease (ADPKD). The spatial and temporal regulation of the polycystin complex within the ciliary membrane remains poorly understood. Using both whole-cell and ciliary patch-clamp recordings, we identify a cilia-enriched oxysterol, 7β,27-dihydroxycholesterol (DHC), that serves as a necessary activator of the polycystin complex. We further identify an oxysterol-binding pocket within PC-2 and showed that mutations within this binding pocket disrupt 7β,27-DHC-dependent polycystin activation. Pharmacologic and genetic inhibition of oxysterol synthesis reduces channel activity in primary cilia. In summary, our findings reveal a regulator of the polycystin complex. This oxysterol-binding pocket in PC-2 may provide a specific target for potential ADPKD therapeutics.
Collapse
Affiliation(s)
- Kodaji Ha
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Nadine Mundt-Machado
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Paola Bisignano
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Aide Pinedo
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - David R Raleigh
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
| | - Gabriel Loeb
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Erhu Cao
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Markus Delling
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
3
|
Hu C, Zhai Y, Lin H, Lu H, Zheng J, Wen C, Li X, Ge RS, Liu Y, Zhu Q. Resveratrol analogues and metabolites selectively inhibit human and rat 11β-hydroxysteroid dehydrogenase 1 as the therapeutic drugs: structure-activity relationship and molecular dynamics analysis. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2024; 35:641-663. [PMID: 39139138 DOI: 10.1080/1062936x.2024.2389817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/03/2024] [Indexed: 08/15/2024]
Abstract
Resveratrol is converted to various metabolites by gut microbiota. Human and rat liver 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1) are critical for glucocorticoid activation, while 11β-HSD2 in the kidney does the opposite reaction. It is still uncertain whether resveratrol and its analogues selectively inhibit 11β-HSD1. In this study, the inhibitory strength, mode of action, structure-activity relationship (SAR), and docking analysis of resveratrol analogues on human, rat, and mouse 11β-HSD1 and 11β-HSD2 were performed. The inhibitory strength of these chemicals on human 11β-HSD1 was dihydropinosylvin (6.91 μM) > lunularin (45.44 μM) > pinostilbene (46.82 μM) > resveratrol (171.1 μM) > pinosylvin (193.8 μM) > others. The inhibitory strength of inhibiting rat 11β-HSD1 was pinostilbene (9.67 μM) > lunularin (17.39 μM) > dihydropinosylvin (19.83 μM) > dihydroresveratrol (23.07 μM) > dihydroxystilbene (27.84 μM) > others and dihydropinosylvin (85.09 μM) and pinostilbene (>100 μM) inhibited mouse 11β-HSD1. All chemicals did not inhibit human, rat, and mouse 11β-HSD2. It was found that dihydropinosylvin, lunularin, and pinostilbene were competitive inhibitors of human 11β-HSD1 and that pinostilbene, lunularin, dihydropinosylvin, dihydropinosylvin and dihydroxystilbene were mixed inhibitors of rat 11β-HSD1. Docking analysis showed that they bind to the steroid-binding site of human and rat 11β-HSD1. In conclusion, resveratrol and its analogues can selectively inhibit human and rat 11β-HSD1, and mouse 11β-HSD1 is insensitive to the inhibition of resveratrol analogues.
Collapse
Affiliation(s)
- C Hu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Y Zhai
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - H Lin
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - H Lu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - J Zheng
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - C Wen
- Department of Neonatal Paediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - X Li
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - R S Ge
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Y Liu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Q Zhu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
4
|
Lin H, Su M, Wen C, Tang Y, Li H, Wu Y, Ge RS, Li XW, Lin H. Chalcones from plants cause toxicity by inhibiting human and rat 11β-hydroxysteroid dehydrogenase 2: 3D-quantitative structure-activity relationship (3D-QSAR) and in silico docking analysis. Food Chem Toxicol 2024; 184:114415. [PMID: 38141941 DOI: 10.1016/j.fct.2023.114415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 12/25/2023]
Abstract
Chalcones from licorice and its related plants have many pharmacological effects. However, the effects of chalcones on the activity of human and rat 11β-hydroxysteroid dehydrogenase 2 (11β-HSD2), and associated side effects remain unclear. The inhibition of 11 chalcones on human and rat 11β-HSD2 were evaluated in microsomes and a 3D-quantitative structure-activity relationship (3D-QSAR) was analyzed. Screening revealed that bavachalcone, echinatin, isobavachalcone, isobavachromene, isoliquiritigenin, licochalcone A, and licochalcone B significantly inhibited human 11β-HSD2 with IC50 values ranging from 15.62 (licochalcone A) to 38.33 (echinatin) μM. Screening showed that the above chemicals and 4-hydroxychalcone significantly inhibited rat 11β-HSD2 with IC50 values ranging from 6.82 (isobavachalcone) to 72.26 (4-hydroxychalcone) μM. These chalcones acted as noncompetitive/mixed inhibitors for both enzymes. Comparative analysis revealed that inhibition of 11β-HSD2 depended on the species. Most chemicals bind to the NAD+ binding site or both the NAD+ and substrate binding sites. Bivariate correlation analysis showed that lipophilicity and molecular weight determine inhibitory strength. Through our 3D-QSAR models, we identified that the hydrophobic region, hydrophobic aliphatic groups, and hydrogen bond acceptors are pivotal factors in inhibiting 11β-HSD2. In conclusion, many chalcones inhibit human and rat 11β-HSD2, possibly causing side effects and there is structure-dependent and species-dependent inhibition on 11β-HSD2.
Collapse
Affiliation(s)
- Hang Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, 325027, China
| | - Ming Su
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, 325027, China
| | - Chao Wen
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, 325027, China
| | - Yunbing Tang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Huitao Li
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, 325027, China
| | - Yandan Wu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, 325027, China
| | - Ren-Shan Ge
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, 325027, China; Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| | - Xing-Wang Li
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, 325027, China.
| | - Han Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, 325027, China.
| |
Collapse
|
5
|
Chen L, Zhao L, Han J, Xiao P, Zhao M, Zhang S, Duan J. Biosynthesis of Chryseno[2,1,c]oxepin-12-Carboxylic Acid from Glycyrrhizic Acid in Aspergillus terreus TMZ05-2, and Analysis of Its Anti-inflammatory Activity. J Microbiol 2024; 62:113-124. [PMID: 38411880 DOI: 10.1007/s12275-024-00105-4] [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: 10/10/2023] [Revised: 12/26/2023] [Accepted: 01/01/2024] [Indexed: 02/28/2024]
Abstract
Glycyrrhizic acid, glycyrrhetinic acid, and their oxo, ester, lactone, and other derivatives, are known for their anti-inflammatory, anti-oxidant, and hypoglycemic pharmacological activities. In this study, chryseno[2,1-c]oxepin-12-carboxylic acid (MG) was first biosynthesized from glycyrrhizic acid through sequential hydrolysis, oxidation, and esterification using Aspergillus terreus TMZ05-2, providing a novel in vitro biosynthetic pathway for glycyrrhizic acid derivatives. Assessing the influence of fermentation conditions and variation of strains during culture under stress-induction strategies enhanced the final molar yield to 88.3% (5 g/L glycyrrhizic acid). CCK8 assays showed no cytotoxicity and good cell proliferation, and anti-inflammatory experiments demonstrated strong inhibition of NO release (36.3%, low-dose MG vs. model), transcriptional downregulation of classical effective cellular factors tumor necrosis factor-α (TNF-α; 72.2%, low-dose MG vs. model), interleukin-6 (IL-6; 58.3%, low-dose MG vs. model) and interleukin-1β (IL-1β; 76.4%, low-dose MG vs. model), and decreased abundance of P-IKK-α, P-IKB-α, and P-P65 proteins, thereby alleviating inflammatory responses through the NF-κB pathway in LPS-induced RAW264.7 cells. The findings provide a reference for the biosynthesis of lactone compounds from medicinal plants.
Collapse
Affiliation(s)
- Liangliang Chen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Key Laboratory of Chinese Medicinal Resources Recycling Utilization of State Administration of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Lin Zhao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Key Laboratory of Chinese Medicinal Resources Recycling Utilization of State Administration of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Ju Han
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Key Laboratory of Chinese Medicinal Resources Recycling Utilization of State Administration of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Ping Xiao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Key Laboratory of Chinese Medicinal Resources Recycling Utilization of State Administration of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, People's Republic of China.
| | - Mingzhe Zhao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Key Laboratory of Chinese Medicinal Resources Recycling Utilization of State Administration of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Sen Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Key Laboratory of Chinese Medicinal Resources Recycling Utilization of State Administration of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, People's Republic of China.
| | - Jinao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Key Laboratory of Chinese Medicinal Resources Recycling Utilization of State Administration of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, People's Republic of China
| |
Collapse
|
6
|
Crompton M, Skinner LJ, Satchell SC, Butler MJ. Aldosterone: Essential for Life but Damaging to the Vascular Endothelium. Biomolecules 2023; 13:1004. [PMID: 37371584 PMCID: PMC10296074 DOI: 10.3390/biom13061004] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
The renin angiotensin aldosterone system is a key regulator of blood pressure. Aldosterone is the final effector of this pathway, acting predominantly via mineralocorticoid receptors. Aldosterone facilitates the conservation of sodium and, with it, water and acts as a powerful stimulus for potassium excretion. However, evidence for the pathological impact of excess mineralocorticoid receptor stimulation is increasing. Here, we discussed how in the heart, hyperaldosteronism is associated with fibrosis, cardiac dysfunction, and maladaptive hypertrophy. In the kidney, aldosterone was shown to cause proteinuria and fibrosis and may contribute to the progression of kidney disease. More recently, studies suggested that aldosterone excess damaged endothelial cells. Here, we reviewed how damage to the endothelial glycocalyx may contribute to this process. The endothelial glycocalyx is a heterogenous, negatively charged layer on the luminal surface of cells. Aldosterone exposure alters this layer. The resulting structural changes reduced endothelial reactivity in response to protective shear stress, altered permeability, and increased immune cell trafficking. Finally, we reviewed current therapeutic strategies for limiting endothelial damage and suggested that preventing glycocalyx remodelling in response to aldosterone exposure may provide a novel strategy, free from the serious adverse effect of hyperkalaemia seen in response to mineralocorticoid blockade.
Collapse
Affiliation(s)
| | | | | | - Matthew J. Butler
- Bristol Renal, Dorothy Hodgkin Building, University of Bristol, Whitson Street, Bristol BS1 3NY, UK
| |
Collapse
|
7
|
Mukherji ST, Brambilla L, Stuart KB, Mayes I, Kutz LC, Chen Y, Barbosa LA, Elmadbouh I, McDermott JP, Haller ST, Romero MF, Soleimani M, Liu J, Shapiro JI, Blanco GV, Xie Z, Pierre SV. Na/K-ATPase signaling tonically inhibits sodium reabsorption in the renal proximal tubule. FASEB J 2023; 37:e22835. [PMID: 36856735 PMCID: PMC10028530 DOI: 10.1096/fj.202200785rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 03/02/2023]
Abstract
Through its classic ATP-dependent ion-pumping function, basolateral Na/K-ATPase (NKA) generates the Na+ gradient that drives apical Na+ reabsorption in the renal proximal tubule (RPT), primarily through the Na+ /H+ exchanger (NHE3). Accordingly, activation of NKA-mediated ion transport decreases natriuresis through activation of basolateral (NKA) and apical (NHE3) Na+ reabsorption. In contrast, activation of the more recently discovered NKA signaling function triggers cellular redistribution of RPT NKA and NHE3 and decreases Na+ reabsorption. We used gene targeting to test the respective contributions of NKA signaling and ion pumping to the overall regulation of RPT Na+ reabsorption. Knockdown of RPT NKA in cells and mice increased membrane NHE3 and Na+ /HCO3 - cotransporter (NBCe1A). Urine output and absolute Na+ excretion decreased by 65%, driven by increased RPT Na+ reabsorption (as indicated by decreased lithium clearance and unchanged glomerular filtration rate), and accompanied by elevated blood pressure. This hyper reabsorptive phenotype was rescued upon crossing with RPT NHE3-/- mice, confirming the importance of NKA/NHE3 coupling. Hence, NKA signaling exerts a tonic inhibition on Na+ reabsorption by regulating key apical and basolateral Na+ transporters. This action, lifted upon NKA genetic suppression, tonically counteracts NKA's ATP-driven function of basolateral Na+ reabsorption. Strikingly, NKA signaling is not only physiologically relevant but it also appears to be functionally dominant over NKA ion pumping in the control of RPT reabsorption.
Collapse
Affiliation(s)
- Shreya T. Mukherji
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Luca Brambilla
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Kailey B. Stuart
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Isabella Mayes
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Laura C. Kutz
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Yiliang Chen
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Blood Research Institute, Versiti, WI
| | - Leandro A Barbosa
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
- Universidade Federal de São João del-Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, MG, Brazil
| | - Ibrahim Elmadbouh
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Jeff P. McDermott
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS
| | - Steven T. Haller
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Michael F. Romero
- Physiology & Biomedical Engineering and Nephrology & Hypertension, Mayo Clinic College of Medicine & Science, Rochester, MN
| | - Manoocher Soleimani
- Department of Medicine, The University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Jiang Liu
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | - Joseph I. Shapiro
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | - Gustavo V. Blanco
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| | - Sandrine V. Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV
| |
Collapse
|
8
|
Johnston JG, Welch AK, Cain BD, Sayeski PP, Gumz ML, Wingo CS. Aldosterone: Renal Action and Physiological Effects. Compr Physiol 2023; 13:4409-4491. [PMID: 36994769 PMCID: PMC11472823 DOI: 10.1002/cphy.c190043] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Aldosterone exerts profound effects on renal and cardiovascular physiology. In the kidney, aldosterone acts to preserve electrolyte and acid-base balance in response to changes in dietary sodium (Na+ ) or potassium (K+ ) intake. These physiological actions, principally through activation of mineralocorticoid receptors (MRs), have important effects particularly in patients with renal and cardiovascular disease as demonstrated by multiple clinical trials. Multiple factors, be they genetic, humoral, dietary, or otherwise, can play a role in influencing the rate of aldosterone synthesis and secretion from the adrenal cortex. Normally, aldosterone secretion and action respond to dietary Na+ intake. In the kidney, the distal nephron and collecting duct are the main targets of aldosterone and MR action, which stimulates Na+ absorption in part via the epithelial Na+ channel (ENaC), the principal channel responsible for the fine-tuning of Na+ balance. Our understanding of the regulatory factors that allow aldosterone, via multiple signaling pathways, to function properly clearly implicates this hormone as central to many pathophysiological effects that become dysfunctional in disease states. Numerous pathologies that affect blood pressure (BP), electrolyte balance, and overall cardiovascular health are due to abnormal secretion of aldosterone, mutations in MR, ENaC, or effectors and modulators of their action. Study of the mechanisms of these pathologies has allowed researchers and clinicians to create novel dietary and pharmacological targets to improve human health. This article covers the regulation of aldosterone synthesis and secretion, receptors, effector molecules, and signaling pathways that modulate its action in the kidney. We also consider the role of aldosterone in disease and the benefit of mineralocorticoid antagonists. © 2023 American Physiological Society. Compr Physiol 13:4409-4491, 2023.
Collapse
Affiliation(s)
- Jermaine G Johnston
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
- Nephrology Section, Veteran Administration Medical Center, North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Amanda K Welch
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA
- Nephrology Section, Veteran Administration Medical Center, North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Brian D Cain
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | - Peter P Sayeski
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
| | - Michelle L Gumz
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
- Nephrology Section, Veteran Administration Medical Center, North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Charles S Wingo
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
- Nephrology Section, Veteran Administration Medical Center, North Florida/South Georgia Malcom Randall Department of Veterans Affairs Medical Center, Gainesville, Florida, USA
| |
Collapse
|
9
|
Loughlin S, Costello HM, Roe AJ, Buckley C, Wilson SM, Bailey MA, Mansley MK. Mapping the Transcriptome Underpinning Acute Corticosteroid Action within the Cortical Collecting Duct. KIDNEY360 2023; 4:226-240. [PMID: 36821614 PMCID: PMC10103384 DOI: 10.34067/kid.0003582022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/08/2022] [Indexed: 11/10/2022]
Abstract
Key Points We report the transcriptomes associated with acute corticosteroid regulation of ENaC activity in polarized mCCDcl1 collecting duct cells. Nine genes were regulated by aldosterone (ALDO), 0 with corticosterone alone, and 151 with corticosterone when 11βHSD2 activity was inhibited. We validated three novel ALDO-induced genes, Rasd1 , Sult1d1 , and Gm43305 , in primary cells isolated from a novel principal cell reporter mouse. Background Corticosteroids regulate distal nephron and collecting duct (CD) Na+ reabsorption, contributing to fluid-volume and blood pressure homeostasis. The transcriptional landscape underpinning the acute stimulation of the epithelial sodium channel (ENaC) by physiological concentrations of corticosteroids remains unclear. Methods Transcriptomic profiles underlying corticosteroid-stimulated ENaC activity in polarized mCCDcl1 cells were generated by coupling electrophysiological measurements of amiloride-sensitive currents with RNAseq. Generation of a principal cell-specific reporter mouse line, mT/mG -Aqp2Cre, enabled isolation of primary CD principal cells by FACS, and ENaC activity was measured in cultured primary cells after acute application of corticosteroids. Expression of target genes was assessed by qRT-PCR in cultured cells or freshly isolated cells after the acute elevation of steroid hormones in mT/mG -Aqp2Cre mice. Results Physiological relevance of the mCCDcl1 model was confirmed with aldosterone (ALDO)-specific stimulation of SGK1 and ENaC activity. Corticosterone (CORT) only modulated these responses at supraphysiological concentrations or when 11βHSD2 was inhibited. When 11βHSD2 protection was intact, CORT caused no significant change in transcripts. We identified a small number of ALDO-induced transcripts associated with stimulated ENaC activity in mCCDcl1 cells and a much larger number with CORT in the absence of 11βHSD2 activity. Principal cells isolated from mT/mG -Aqp2Cre mice were validated and assessment of identified ALDO-induced genes revealed that Sgk1 , Zbtbt16 , Sult1d1 , Rasd1 , and Gm43305 are acutely upregulated by corticosteroids both in vitro and in vivo . Conclusions This study reports the transcriptome of mCCDcl1 cells and identifies a small number of ALDO-induced genes associated with acute stimulation of ENaC, including three previously undescribed genes.
Collapse
Affiliation(s)
- Struan Loughlin
- Cellular Medicine Research Division, University of St Andrews, St Andrews, United Kingdom
- Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Hannah M. Costello
- Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew J. Roe
- Division of Pharmacy, School of Medicine, Pharmacy and Health, Durham University Queen's Campus, Stockton-on-Tees, United Kingdom
| | - Charlotte Buckley
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Stuart M. Wilson
- Division of Pharmacy, School of Medicine, Pharmacy and Health, Durham University Queen's Campus, Stockton-on-Tees, United Kingdom
| | - Matthew A. Bailey
- Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Morag K. Mansley
- Cellular Medicine Research Division, University of St Andrews, St Andrews, United Kingdom
- Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Division of Pharmacy, School of Medicine, Pharmacy and Health, Durham University Queen's Campus, Stockton-on-Tees, United Kingdom
| |
Collapse
|
10
|
Korokin MV, Gudyrev OS, Lebedev PR, Kuzubova EV, Radchenko AI, Koklin IS, Taran EI, Kochkarov AA. Characteristics of the state of bone tissue in genetically modified mice with impaired enzymatic regulation of steroid hormone metabolism. RESEARCH RESULTS IN PHARMACOLOGY 2022. [DOI: 10.3897/rrpharmacology.8.98779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Introduction: The aim was to evaluate the structural and functional changes of bone tissue in mice with null expression of 11β-HSD2 or both 11β-HSD2 and Apolipoprotein E.
Materials and methods: The experimental study was performed in 60 male mice, weighting 24–30 g. The animals were kept in accordance with the rules of laboratory practice for preclinical studies on the territory of the Russian Federation. Mice lacking 11β-HSD2 (Hsd2-/-) and male mice lacking 11β-HSD2 and Apolipoprotein E (Hsd2-/-/Apoe-/-) were used in the study. We studied and characterized the state of bone tissue, indicators of bone density, microcirculation in bone tissue, endothelial dysfunction coefficient, width of bone trabeculae, as well as serum concentrations of bone alkaline phosphatase, hydroxyproline, deoxyprinoline and expression levels of p53, Bcl2, Bax, eNOS genes.
Results and discussion: We showed that mice with the Hsd2-/- genotype with no expression of 11ß-HSD2 by the 6th month of life showed a statistically significant decrease in bone density, which progresses to the 7th and 8th months of life. At the 8th month of animal life, a decrease in bone density is accompanied by a statistically significant decrease in the level of microcirculation in the bone and an increase in the coefficient of endothelial dysfunction. Taking into account the relationship of endothelial dysfunction, atherogenesis and disorders in the processes of bone remodeling, in the framework of this study, we also assessed the state of bone tissue in double transgenes with the genotype Hsd2-/-/Apoe-/-, which lack the expression of both 11ß-HSD2 and Apolipoprotein E. In this study, we also saw increased activation of processes leading to disruption of bone remodeling processes. In the group of the animals with the genotype Hsd2-/-/Apoe-/-, we found statistically significant differences from the mice with no expression of 11ß-HSD2 in bone density and microcirculation, and the width of bone trabeculae. Also, a statistically significant increase in hydroxyproline and deoxyprinoline was found in the group of double transgenes, in the absence of significant changes in the concentration of bone alkaline phosphatase. This fact indicates a pronounced activation of bone resorption processes in the absence of activation of osteosynthesis processes, which leads to the detected violation of bone remodeling processes.
Conclusion: Thus, we have shown that a violation of the metabolic regulation of steroid hormone metabolism in animals with null expression of the 11ß-HSD2 (Hsd2-/- genotype) leads to the development of signs of osteoporosis – bone density decreases, which is accompanied by a decrease in the width of bone trabeculae, the level of microcirculation in bone tissue decreases simultaneously with an increase in the coefficient of endothelial dysfunction. The additional null expression of ApoE gene in double transgenes with the genotype Hsd2-/-/Apoe-/- leads to an increase in the severity of changes associated with a violation of bone remodeling processes and, in addition to a more pronounced change in bone tissue density, bone trabecular width, microcirculation and the coefficient of endothelial dysfunction leads to an increase in the concentration of biochemical markers of bone resorption. These changes indicate the important role of the enzyme 11ß-hydroxysteroid dehydrogenase type 2 in the processes of bone remodeling disorders.
Graphical abstract
Collapse
|
11
|
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
|
12
|
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
|
13
|
Equisetin is an anti-obesity candidate through targeting 11 β-HSD1. Acta Pharm Sin B 2022; 12:2358-2373. [PMID: 35646525 PMCID: PMC9136616 DOI: 10.1016/j.apsb.2022.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/25/2022] Open
Abstract
Obesity is increasingly prevalent globally, searching for therapeutic agents acting on adipose tissue is of great importance. Equisetin (EQST), a meroterpenoid isolated from a marine sponge-derived fungus, has been reported to display antibacterial and antiviral activities. Here, we revealed that EQST displayed anti-obesity effects acting on adipose tissue through inhibiting adipogenesis in vitro and attenuating HFD-induced obesity in mice, doing so without affecting food intake, blood pressure or heart rate. We demonstrated that EQST inhibited the enzyme activity of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), a therapeutic target of obesity in adipose tissue. Anti-obesity properties of EQST were all offset by applying excessive 11β-HSD1's substrates and 11β-HSD1 inhibition through knockdown in vitro or 11β-HSD1 knockout in vivo. In the 11β-HSD1 bypass model constructed by adding excess 11β-HSD1 products, EQST's anti-obesity effects disappeared. Furthermore, EQST directly bond to 11β-HSD1 protein and presented remarkable better intensity on 11β-HSD1 inhibition and better efficacy on anti-obesity than known 11β-HSD1 inhibitor. Therefore, EQST can be developed into anti-obesity candidate compound, and this study may provide more clues for developing higher effective 11β-HSD1 inhibitors.
Collapse
|
14
|
Liu Y, Ding Q, Guo W. Life Course Impact of Glucocorticoids During Pregnancy on Muscle Development and Function. FRONTIERS IN ANIMAL SCIENCE 2021; 2. [PMID: 36325303 PMCID: PMC9624510 DOI: 10.3389/fanim.2021.788930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Maternal stress, such as maternal obesity, can induce severe gestational disease and hormonal disorder which may disrupt fetal organ maturation and further cause endangered early or future health in offspring. During fetal development, glucocorticoids are essential for the maturation of organ systems. For instance, in clinical applications, glucocorticoids are commonly utilized to pregnant women with the risk of preterm delivery to reduce mortality of the newborns. However, exposure of excessive glucocorticoids at embryonic and fetal developmental stages can cause diseases such as cardiovascular disease and muscle atrophy in adulthood. Effects of excessive glucocorticoids on human health are well-recognized and extensively studied. Nonetheless, effects of these hormones on farm animal growth and development, particularly on prenatal muscle development, and postnatal growth, did not attract much attention until the last decade. Here, we provided a short review of the recent progress relating to the effect of glucocorticoids on prenatal skeletal muscle development and postnatal muscle growth as well as heart muscle development and cardiovascular disease during life course.
Collapse
|
15
|
Knockout of the hsd11b2 Gene Extends the Cortisol Stress Response in Both Zebrafish Larvae and Adults. Int J Mol Sci 2021; 22:ijms222212525. [PMID: 34830405 PMCID: PMC8619348 DOI: 10.3390/ijms222212525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/14/2021] [Accepted: 11/17/2021] [Indexed: 11/30/2022] Open
Abstract
The Hsd11b2 enzyme converts cortisol into its inactive form, cortisone and regulates cortisol levels, in particular in response to stress. Taking advantage of CRISPR/Cas9 technology, we generated a hsd11b2 zebrafish mutant line to evaluate the involvement of this gene in stress response regulation. The absence of a functional Hsd11b2 affects survival of zebrafish, although homozygous hsd11b2−/− mutants can reach adulthood. Reproductive capability of hsd11b2−/− homozygous adult males is almost completely abrogated, while that of females is reduced. Interestingly, basal cortisol levels and glucocorticoid-dependent transcriptional activities are not affected by the mutation. In agreement with basal cortisol results, we also demonstrated that basal response to light (LMR-L/D) or mechanical (VSRA) stimuli is not significantly different in wild-type (hsd11b2+/+) compared to mutant larvae. However, after exposure to an acute stressor, the cortisol temporal patterns of synthesis and release are prolonged in both 5 days post fertilization larvae and one-year-old adult hsd11b2−/− zebrafish compared to wild-type siblings, showing at the same time, at 5 dpf, a higher magnitude in the stress response at 10 min post stress. All in all, this new zebrafish model represents a good tool for studying response to different stressors and to identify mechanisms that are induced by cortisol during stress response.
Collapse
|
16
|
Yang J, Chen Y, Li X, Xu D. New insights into the roles of glucocorticoid signaling dysregulation in pathological cardiac hypertrophy. Heart Fail Rev 2021; 27:1431-1441. [PMID: 34455516 DOI: 10.1007/s10741-021-10158-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/09/2021] [Indexed: 01/02/2023]
Abstract
Pathological cardiac hypertrophy is a process of abnormal remodeling of the myocardium in response to stress overload or ischemia that results in myocardial injury, which is an independent risk factor for the increased morbidity and mortality of heart failure. Elevated circulating glucocorticoids (GCs) levels are associated with an increased risk of pathological cardiac hypertrophy, but the exact role remains unclear. In the heart, GCs exerts physiological and pharmacological effects by binding the glucocorticoid receptor (GR, NR3C1). However, under the state of tissue damage or oxidative stress, GCs can also bind the closely related mineralocorticoid receptor (MR, NR3C2) to exert a detrimental effect on cardiac function. In addition, the bioavailability of GCs at the cellular level is mainly regulated by tissue-specific metabolic enzymes 11β-hydroxysteroid dehydrogenases (11β-HSDs), including 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) and type 2 (11β-HSD2), which catalyze the interconversion of active GCs. In this paper, we provide an overview of GC signaling and its physiological roles in the heart and highlight the dynamic and diverse roles of GC signaling dysregulation, mediated by excessive ligand GCs levels, GR/MR deficiency or overexpression, and local GCs metabolic disorder by 11β-HSDs, in the pathology of cardiac hypertrophy. Our findings will provide new ideas and insights for the search for appropriate intervention targets for pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Jingmin Yang
- Department of Cardiology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410000, Hunan, China
| | - Yanying Chen
- Department of Cardiology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410000, Hunan, China
| | - Xiao Li
- Department of Cardiology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410000, Hunan, China
| | - Danyan Xu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410000, Hunan, China.
| |
Collapse
|
17
|
Assmus A, Mullins L, Ward M, Dobie R, Hunter R, Henderson NC, Mullins JJ. Loss of Adam10 Disrupts Ion Transport in Immortalized Kidney Collecting Duct Cells. FUNCTION 2021; 2:zqab024. [PMID: 34131651 PMCID: PMC8187228 DOI: 10.1093/function/zqab024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 01/06/2023] Open
Abstract
The kidney cortical collecting duct (CCD) comprises principal cells (PCs), intercalated cells (IC), and the recently discovered intermediate cell type. Kidney pathology in a mouse model of the syndrome of apparent aldosterone excess revealed plasticity of the CCD, with altered PC:intermediate cell:IC ratio. The self-immortalized mouse CCD cell line, mCCDcl1, shows functional characteristics of PCs, but displays a range of cell types, including intermediate cells, making it ideal to study plasticity. We knocked out Adam10, a key component of the Notch pathway, in mCCDcl1 cells, using CRISPR-Cas9 technology, and isolated independent clones, which exhibited severely affected sodium transport capacity and loss of aldosterone response. Single-cell RNA sequencing revealed significantly reduced expression of major PC-specific markers, such as Scnn1g (γ-ENaC) and Hsd11b2 (11βHSD2), but no significant changes in transcription of components of the Notch pathway were observed. Immunostaining in the knockout clone confirmed the decrease in expression of γ-ENaC and importantly, showed an altered, diffuse distribution of PC and IC markers, suggesting altered trafficking in the Adam10 knockout clone as an explanation for the loss of polarization.
Collapse
Affiliation(s)
- Adrienne Assmus
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Linda Mullins
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Mairi Ward
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Ross Dobie
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Robert Hunter
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Neil C Henderson
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - John J Mullins
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| |
Collapse
|
18
|
Wang Y, Wang J, Yang R, Wang P, Porche R, Kim S, Lutfy K, Liu L, Friedman TC, Jiang M, Liu Y. Decreased 11β-Hydroxysteroid Dehydrogenase Type 2 Expression in the Kidney May Contribute to Nicotine/Smoking-Induced Blood Pressure Elevation in Mice. Hypertension 2021; 77:1940-1952. [PMID: 33813843 DOI: 10.1161/hypertensionaha.120.16458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Ying Wang
- Department of Pediatrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (Y.W., R.Y., Y.L.).,Division of Endocrinology, Metabolism and Molecular Medicine, Charles R. Drew University of Medicine and Sciences, Los Angeles, CA (Y.W., J.W., P.W., R.P., S.K., K.L., T.C.F., Y.L.)
| | - Jian Wang
- Division of Endocrinology, Metabolism and Molecular Medicine, Charles R. Drew University of Medicine and Sciences, Los Angeles, CA (Y.W., J.W., P.W., R.P., S.K., K.L., T.C.F., Y.L.).,Department of Neonatology, The First Hospital of Jilin University, Changchun, China (J.W.)
| | - Rong Yang
- Department of Pediatrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (Y.W., R.Y., Y.L.)
| | - Piwen Wang
- Division of Endocrinology, Metabolism and Molecular Medicine, Charles R. Drew University of Medicine and Sciences, Los Angeles, CA (Y.W., J.W., P.W., R.P., S.K., K.L., T.C.F., Y.L.)
| | - Rene Porche
- Division of Endocrinology, Metabolism and Molecular Medicine, Charles R. Drew University of Medicine and Sciences, Los Angeles, CA (Y.W., J.W., P.W., R.P., S.K., K.L., T.C.F., Y.L.)
| | - Samuel Kim
- Division of Endocrinology, Metabolism and Molecular Medicine, Charles R. Drew University of Medicine and Sciences, Los Angeles, CA (Y.W., J.W., P.W., R.P., S.K., K.L., T.C.F., Y.L.)
| | - Kabirullah Lutfy
- Division of Endocrinology, Metabolism and Molecular Medicine, Charles R. Drew University of Medicine and Sciences, Los Angeles, CA (Y.W., J.W., P.W., R.P., S.K., K.L., T.C.F., Y.L.).,College of Pharmacy, Western University of Health Sciences, Pomona, CA (K.L.)
| | - Limei Liu
- Department of Endocrinology and Metabolism, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, China (L.L.)
| | - Theodore C Friedman
- Division of Endocrinology, Metabolism and Molecular Medicine, Charles R. Drew University of Medicine and Sciences, Los Angeles, CA (Y.W., J.W., P.W., R.P., S.K., K.L., T.C.F., Y.L.).,David Geffen School of Medicine at University of California, Los Angeles (T.C.F., Y.L.)
| | - Meisheng Jiang
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles (M.J.)
| | - Yanjun Liu
- Department of Pediatrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (Y.W., R.Y., Y.L.).,Division of Endocrinology, Metabolism and Molecular Medicine, Charles R. Drew University of Medicine and Sciences, Los Angeles, CA (Y.W., J.W., P.W., R.P., S.K., K.L., T.C.F., Y.L.).,David Geffen School of Medicine at University of California, Los Angeles (T.C.F., Y.L.)
| |
Collapse
|
19
|
Ferguson D, Hutson I, Tycksen E, Pietka TA, Bauerle K, Harris CA. Role of Mineralocorticoid Receptor in Adipogenesis and Obesity in Male Mice. Endocrinology 2020; 161:bqz010. [PMID: 32036385 PMCID: PMC7007880 DOI: 10.1210/endocr/bqz010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 10/30/2019] [Indexed: 02/07/2023]
Abstract
Increased visceral adiposity and hyperglycemia, 2 characteristics of metabolic syndrome, are also present in conditions of excess glucocorticoids (GCs). GCs are hormones thought to act primarily via the glucocorticoid receptor (GR). GCs are commonly prescribed for inflammatory disorders, yet their use is limited due to many adverse metabolic side effects. In addition to GR, GCs also bind the mineralocorticoid receptor (MR), but there are many conflicting studies about the exact role of MR in metabolic disease. Using MR knockout mice (MRKO), we find that both white and brown adipose depots form normally when compared with wild-type mice at P5. We created mice with adipocyte-specific deletion of MR (FMRKO) to better understand the role of MR in metabolic dysfunction. Treatment of mice with excess GCs for 4 weeks, via corticosterone in drinking water, induced increased fat mass and glucose intolerance to similar levels in FMRKO and floxed control mice. Separately, when fed a high-fat diet for 16 weeks, FMRKO mice had reduced body weight, fat mass, and hepatic steatosis, relative to floxed control mice. Decreased adiposity likely resulted from increased energy expenditure since food intake was not different. RNA sequencing analysis revealed decreased enrichment of genes associated with adipogenesis in inguinal white adipose of FMRKO mice. Differentiation of mouse embryonic fibroblasts (MEFs) showed modestly impaired adipogenesis in MRKO MEFs compared with wild type, but this was rescued upon the addition of peroxisome proliferator-activated receptor gamma (PPARγ) agonist or PPARγ overexpression. Collectively, these studies provide further evidence supporting the potential value of MR as a therapeutic target for conditions associated with metabolic syndrome.
Collapse
Affiliation(s)
- Daniel Ferguson
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
| | - Irina Hutson
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
| | - Eric Tycksen
- Genome Technology Access Center, McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Terri A Pietka
- Nutrition and Geriatrics Division, Washington University School of Medicine, St. Louis, Missouri
| | - Kevin Bauerle
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
| | - Charles A Harris
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
- Department of Medicine, Veterans Affairs St Louis Healthcare System, John Cochran Division, St. Louis, Missouri
| |
Collapse
|
20
|
Lerman LO, Kurtz TW, Touyz RM, Ellison DH, Chade AR, Crowley SD, Mattson DL, Mullins JJ, Osborn J, Eirin A, Reckelhoff JF, Iadecola C, Coffman TM. Animal Models of Hypertension: A Scientific Statement From the American Heart Association. Hypertension 2019; 73:e87-e120. [PMID: 30866654 DOI: 10.1161/hyp.0000000000000090] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hypertension is the most common chronic disease in the world, yet the precise cause of elevated blood pressure often cannot be determined. Animal models have been useful for unraveling the pathogenesis of hypertension and for testing novel therapeutic strategies. The utility of animal models for improving the understanding of the pathogenesis, prevention, and treatment of hypertension and its comorbidities depends on their validity for representing human forms of hypertension, including responses to therapy, and on the quality of studies in those models (such as reproducibility and experimental design). Important unmet needs in this field include the development of models that mimic the discrete hypertensive syndromes that now populate the clinic, resolution of ongoing controversies in the pathogenesis of hypertension, and the development of new avenues for preventing and treating hypertension and its complications. Animal models may indeed be useful for addressing these unmet needs.
Collapse
|
21
|
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: 3.0] [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
|
22
|
Feng Q, Hui J, Tang N, Liu YM, Zhong H, Li Z, Wang LM, Qu YY, Deng FM, He F. Unexpected role of the human cytomegalovirus contribute to essential hypertension in the Kazakh Chinese population of Xinjiang. Biosci Rep 2018; 38:BSR20171522. [PMID: 29752343 PMCID: PMC6019381 DOI: 10.1042/bsr20171522] [Citation(s) in RCA: 5] [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: 11/09/2017] [Revised: 04/29/2018] [Accepted: 05/10/2018] [Indexed: 02/06/2023] Open
Abstract
Human cytomegalovirus (HCMV) infection, chronic inflammation and oxidative stress, the renin-angiotensin system (RAS), endothelial function, and DNA methylation play roles in the pathogenesis of essential hypertension (EH); however, the mechanism by which HCMV predisposes patients to hypertension remain unclear. Our group previously demonstrated an association between EH and HCMV infection in Kazakh Chinese. Here, we investigated the relationship between HCMV infection and other clinicopathological features in 720 Kazakh individuals with or without hypertension (n=360 each; age: 18-80). Multiple linear and logistic regression analyses were used to determine the associations between HCMV infection, clinical characteristics, and EH. Notably, patients with EH, particularly those with HCMV infection, exhibited a marked increase in tumor necrosis factor-α (TNF-α) and 8-hydroxy-2-deoxyguanosine (8-OHDG) levels, but a decrease in endothelial nitric oxide synthase (eNOS) and renin levels. Similarly, elevated TNF-α and 8-OHDG levels were independent predictors of increased HCMV antibody titers, whereas eNOS and renin were negatively correlated with the latter. Moreover, serum angiotensin-converting enzyme (sACE, ACE) methylation was increased, whereas 11-β hydroxysteroid dehydrogenase 2 (HSD11β2; HSD3B2) methylation was decreased in patients with EH who were also infected with HCMV. A positive correlation between HSD3B2 methylation and HCMV IgG titer and blood pressure was additionally observed, whereas angiotensin-converting enzyme (ACE) methylation was inversely correlated with blood pressure. Collectively, these data indicate that HCMV may contribute to EH development in the Kazakh Chinese by increasing TNF-α and 8-OHDG levels, suppressing eNOS and renin, and manipulating HSD3B2 and ACE methylation.
Collapse
Affiliation(s)
- Qian Feng
- Department of Pathophysiology/Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, China
| | - Jing Hui
- Department of Pathophysiology/Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, China
| | - Na Tang
- Department of Pathophysiology/Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, China
| | - Yong-Min Liu
- Department of Pathophysiology/Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, China
| | - Hua Zhong
- Department of Pathophysiology/Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, China
| | - Zhen Li
- Department of Pathophysiology/Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, China
| | - La-Mei Wang
- Centre of Medical Functional Experiments, Medical College of Shihezi University, Shihezi, China
| | - Yuan-Yuan Qu
- Department of Respiration Medicine, The First Affiliated Hospital of Medical College of Shihezi University, Shihezi, Xijiang, China
| | - Feng-Mei Deng
- Department of Pathophysiology, Chengdu Medical College, Sichuan, China
| | - Fang He
- Department of Pathophysiology/Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, Shihezi, China
| |
Collapse
|
23
|
A novel derivatives of thiazol-4(5H)-one and their activity in the inhibition of 11β-hydroxysteroid dehydrogenase type 1. Bioorg Chem 2018; 79:115-121. [PMID: 29738970 DOI: 10.1016/j.bioorg.2018.04.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/18/2018] [Accepted: 04/20/2018] [Indexed: 11/22/2022]
Abstract
11β-Hydroxysteroid dehydrogenase type 1 (11β-HSD1) is an enzyme that catalyzes the conversion of inactive cortisone into physiologically active cortisol. Inhibiting the activity of this enzyme plays a key role in the treatment of Cushing's syndrome, metabolic syndrome and type 2 diabetes. Therefore, new compounds that are selective inhibitors of this enzyme are constantly being looked for. In this work we present the synthesis of 2-(allylamino)thiazol-4(5H)-one derivatives by the reaction of N-allylthiourea with appropriate α-bromoesters. In the case of using of aliphatic α-bromoesters and α-bromo-β-phenylesters, the reactions were carried out in a basic medium (sodium ethoxide) and the products were isolated with a yield of up to 68%. Derivatives containing spiro systems in which carbon C-5 of the thiazole ring is the linker atom were obtained in the presence of N,N-diisopropylethylamine. Some of the obtained compounds, at a concentration of 10 μM have activity in the inhibition of 11β-HSD1 up to 71%. IC50 value for the most active compound: 2-(allylamino)-1-thia-3-azaspiro[4.5]dec-2-en-4-one is 2.5 µM. With a high degree of 11β-HSD1 inhibition and a relatively large difference in the inhibition of 11β-HSD1 and 11β-HSD2 activity, this compound appears to be promising and should be subjected to further testing.
Collapse
|
24
|
In search of selective 11β-HSD type 1 inhibitors without nephrotoxicity: An approach to resolve the metabolic syndrome by virtual based screening. ARAB J CHEM 2018. [DOI: 10.1016/j.arabjc.2015.08.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
25
|
Wu C, Cao S, Hong T, Dong Y, Li C, Wang Q, Sun J, Ge RS. Taxifolin inhibits rat and human 11β-hydroxysteroid dehydrogenase 2. Fitoterapia 2017; 121:112-117. [DOI: 10.1016/j.fitote.2017.07.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 01/21/2017] [Accepted: 07/07/2017] [Indexed: 01/04/2023]
|
26
|
Whirledge S, Cidlowski JA. Glucocorticoids and Reproduction: Traffic Control on the Road to Reproduction. Trends Endocrinol Metab 2017; 28:399-415. [PMID: 28274682 PMCID: PMC5438761 DOI: 10.1016/j.tem.2017.02.005] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/06/2017] [Accepted: 02/12/2017] [Indexed: 02/06/2023]
Abstract
Glucocorticoids are steroid hormones that regulate diverse cellular functions and are essential to facilitate normal physiology. However, stress-induced levels of glucocorticoids result in several pathologies including profound reproductive dysfunction. Compelling new evidence indicates that glucocorticoids are crucial to the establishment and maintenance of reproductive function. The fertility-promoting or -inhibiting activity of glucocorticoids depends on timing, dose, and glucocorticoid responsiveness within a given tissue, which is mediated by the glucocorticoid receptor (GR). The GR gene and protein are subject to cellular processing, contributing to signaling diversity and providing a mechanism by which both physiological and stress-induced levels of glucocorticoids function in a cell-specific manner. Understanding how glucocorticoids regulate fertility and infertility may lead to novel approaches to the regulation of reproductive function.
Collapse
Affiliation(s)
- Shannon Whirledge
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - John A Cidlowski
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 TW Alexander Drive, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
27
|
Abstract
Glucocorticoids are primary stress hormones produced by the adrenal cortex. The concentration of serum glucocorticoids in the fetus is low throughout most of gestation but surge in the weeks prior to birth. While their most well-known function is to stimulate differentiation and functional development of the lungs, glucocorticoids also play crucial roles in the development of several other organ systems. Mothers at risk of preterm delivery are administered glucocorticoids to accelerate fetal lung development and prevent respiratory distress. Conversely, excessive glucocorticoid signaling is detrimental for fetal development; slowing fetal and placental growth and programming the individual for disease later in adult life. This review explores the mechanisms that control glucocorticoid signaling during pregnancy and provides an overview of the impact of glucocorticoid signaling on fetal development.
Collapse
Affiliation(s)
- Jonathan T Busada
- Molecular Endocrinology Group, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - John A Cidlowski
- Molecular Endocrinology Group, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States.
| |
Collapse
|
28
|
Gray GA, White CI, Castellan RFP, McSweeney SJ, Chapman KE. Getting to the heart of intracellular glucocorticoid regeneration: 11β-HSD1 in the myocardium. J Mol Endocrinol 2017; 58:R1-R13. [PMID: 27553202 PMCID: PMC5148800 DOI: 10.1530/jme-16-0128] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 08/19/2016] [Indexed: 12/11/2022]
Abstract
Corticosteroids influence the development and function of the heart and its response to injury and pressure overload via actions on glucocorticoid (GR) and mineralocorticoid (MR) receptors. Systemic corticosteroid concentration depends largely on the activity of the hypothalamic-pituitary-adrenal (HPA) axis, but glucocorticoid can also be regenerated from intrinsically inert metabolites by the enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), selectively increasing glucocorticoid levels within cells and tissues. Extensive studies have revealed the roles for glucocorticoid regeneration by 11β-HSD1 in liver, adipose, brain and other tissues, but until recently, there has been little focus on the heart. This article reviews the evidence for glucocorticoid metabolism by 11β-HSD1 in the heart and for a role of 11β-HSD1 activity in determining the myocardial growth and physiological function. We also consider the potential of 11β-HSD1 as a therapeutic target to enhance repair after myocardial infarction and to prevent the development of cardiac remodelling and heart failure.
Collapse
Affiliation(s)
- Gillian A Gray
- University/BHF Centre for Cardiovascular ScienceQueen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Christopher I White
- University/BHF Centre for Cardiovascular ScienceQueen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Raphael F P Castellan
- University/BHF Centre for Cardiovascular ScienceQueen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Sara J McSweeney
- University/BHF Centre for Cardiovascular ScienceQueen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Karen E Chapman
- University/BHF Centre for Cardiovascular ScienceQueen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
29
|
Pravastatin ameliorates placental vascular defects, fetal growth, and cardiac function in a model of glucocorticoid excess. Proc Natl Acad Sci U S A 2016; 113:6265-70. [PMID: 27185937 DOI: 10.1073/pnas.1520356113] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Fetoplacental glucocorticoid overexposure is a significant mechanism underlying fetal growth restriction and the programming of adverse health outcomes in the adult. Placental glucocorticoid inactivation by 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2) plays a key role. We previously discovered that Hsd11b2(-/-) mice, lacking 11β-HSD2, show marked underdevelopment of the placental vasculature. We now explore the consequences for fetal cardiovascular development and whether this is reversible. We studied Hsd11b2(+/+), Hsd11b2(+/-), and Hsd11b2(-/-) littermates from heterozygous (Hsd11b(+/-)) matings at embryonic day (E)14.5 and E17.5, where all three genotypes were present to control for maternal effects. Using high-resolution ultrasound, we found that umbilical vein blood velocity in Hsd11b2(-/-) fetuses did not undergo the normal gestational increase seen in Hsd11b2(+/+) littermates. Similarly, the resistance index in the umbilical artery did not show the normal gestational decline. Surprisingly, given that 11β-HSD2 absence is predicted to initiate early maturation, the E/A wave ratio was reduced at E17.5 in Hsd11b2(-/-) fetuses, suggesting impaired cardiac function. Pravastatin administration from E6.5, which increases placental vascular endothelial growth factor A and, thus, vascularization, increased placental fetal capillary volume, ameliorated the aberrant umbilical cord velocity, normalized fetal weight, and improved the cardiac function of Hsd11b2(-/-) fetuses. This improved cardiac function occurred despite persisting indications of increased glucocorticoid exposure in the Hsd11b2(-/-) fetal heart. Thus, the pravastatin-induced enhancement of fetal capillaries within the placenta and the resultant hemodynamic changes correspond with restored fetal cardiac function. Statins may represent a useful therapeutic approach to intrauterine growth retardation due to placental vascular hypofunction.
Collapse
|
30
|
Murumkar PR, Shinde AC, Sharma MK, Yamaguchi H, Miniyar PB, Yadav MR. Development of a credible 3D-QSAR CoMSIA model and docking studies for a series of triazoles and tetrazoles containing 11β-HSD1 inhibitors. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2016; 27:265-292. [PMID: 27094303 DOI: 10.1080/1062936x.2016.1167774] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Type 2 diabetes mellitus is described by insulin resistance and high fasting blood glucose. Increased levels of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) enzyme result in insulin resistance and metabolic syndrome. Inhibition of 11β-HSD1 decreases glucose production and increases hepatic insulin sensitivity. Use of selective 11β-HSD1 inhibitors could prove to be an effective strategy for the treatment of the disease. It was decided to identify the essential structural features required by any compound to possess 11β-HSD1 inhibitory activity. A dataset of 139 triazoles and tetrazoles having 11β-HSD1 inhibitory activity was used for the development of a 3D-QSAR model. The best comparative molecular field analysis (CoMFA) model was generated with databased alignment, which was further used for comparative molecular similarity indices analysis (CoMSIA). The optimal CoMSIA model showed [Formula: see text] = 0.809 with five components, [Formula: see text] = 0.931, SEE = 0.323 and F-value = 249.126. The CoMSIA model offered better prediction than the CoMFA model with [Formula: see text] = 0.522 and 0.439, respectively, indicating that the CoMSIA model appeared to be a better one for the prediction of activity for the newly designed 11β-HSD1 inhibitors. The selectivity aspect of 11β-HSD1 over 11β-HSD2 was studied with the help of docking studies.
Collapse
Affiliation(s)
- P R Murumkar
- a Faculty of Pharmacy , The Maharaja Sayajirao University of Baroda , Vadodara , India
- b Sinhgad Institute of Pharmacy , Narhe , India
| | - A C Shinde
- b Sinhgad Institute of Pharmacy , Narhe , India
| | - M K Sharma
- a Faculty of Pharmacy , The Maharaja Sayajirao University of Baroda , Vadodara , India
- b Sinhgad Institute of Pharmacy , Narhe , India
| | - H Yamaguchi
- c Department of Pharmacy , Meijo University , Nagoya , Japan
| | - P B Miniyar
- b Sinhgad Institute of Pharmacy , Narhe , India
| | - M R Yadav
- a Faculty of Pharmacy , The Maharaja Sayajirao University of Baroda , Vadodara , India
| |
Collapse
|
31
|
CZARNECKA ANNAM, NIEDZWIEDZKA MAGDALENA, PORTA CAMILLO, SZCZYLIK CEZARY. Hormone signaling pathways as treatment targets in renal cell cancer (Review). Int J Oncol 2016; 48:2221-35. [DOI: 10.3892/ijo.2016.3460] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/14/2016] [Indexed: 11/06/2022] Open
|
32
|
Evans LC, Ivy JR, Wyrwoll C, McNairn JA, Menzies RI, Christensen TH, Al-Dujaili EAS, Kenyon CJ, Mullins JJ, Seckl JR, Holmes MC, Bailey MA. Conditional Deletion of Hsd11b2 in the Brain Causes Salt Appetite and Hypertension. Circulation 2016; 133:1360-70. [PMID: 26951843 PMCID: PMC4819772 DOI: 10.1161/circulationaha.115.019341] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 02/12/2016] [Indexed: 11/30/2022]
Abstract
Supplemental Digital Content is available in the text. Background— The hypertensive syndrome of Apparent Mineralocorticoid Excess is caused by loss-of-function mutations in the gene encoding 11β-hydroxysteroid dehydrogenase type 2 (11βHSD2), allowing inappropriate activation of the mineralocorticoid receptor by endogenous glucocorticoid. Hypertension is attributed to sodium retention in the distal nephron, but 11βHSD2 is also expressed in the brain. However, the central contribution to Apparent Mineralocorticoid Excess and other hypertensive states is often overlooked and is unresolved. We therefore used a Cre-Lox strategy to generate 11βHSD2 brain-specific knockout (Hsd11b2.BKO) mice, measuring blood pressure and salt appetite in adults. Methods and Results— Basal blood pressure, electrolytes, and circulating corticosteroids were unaffected in Hsd11b2.BKO mice. When offered saline to drink, Hsd11b2.BKO mice consumed 3 times more sodium than controls and became hypertensive. Salt appetite was inhibited by spironolactone. Control mice fed the same daily sodium intake remained normotensive, showing the intrinsic salt resistance of the background strain. Dexamethasone suppressed endogenous glucocorticoid and abolished the salt-induced blood pressure differential between genotypes. Salt sensitivity in Hsd11b2.BKO mice was not caused by impaired renal sodium excretion or volume expansion; pressor responses to phenylephrine were enhanced and baroreflexes impaired in these animals. Conclusions— Reduced 11βHSD2 activity in the brain does not intrinsically cause hypertension, but it promotes a hunger for salt and a transition from salt resistance to salt sensitivity. Our data suggest that 11βHSD2-positive neurons integrate salt appetite and the blood pressure response to dietary sodium through a mineralocorticoid receptor–dependent pathway. Therefore, central mineralocorticoid receptor antagonism could increase compliance to low-sodium regimens and help blood pressure management in cardiovascular disease.
Collapse
Affiliation(s)
- Louise C Evans
- From British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, United Kingdom (L.C.E., J.R.I., C.W., J.A.M., R.I.M., T.H.C., C.J.K., J.J.M., J.R.S., M.C.H., M.A.B.); and Dietetics, Nutrition and Biological Sciences Department, Queen Margaret University, Edinburgh, United Kingdom (E.A.S.Al-D.). The current address for Dr Evans is Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee; the current address for Dr Wyrwoll is School of Anatomy, Physiology & Human Biology, The University of Western Australia, Crawley, Australia; and the current address for Dr Christensen is Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense
| | - Jessica R Ivy
- From British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, United Kingdom (L.C.E., J.R.I., C.W., J.A.M., R.I.M., T.H.C., C.J.K., J.J.M., J.R.S., M.C.H., M.A.B.); and Dietetics, Nutrition and Biological Sciences Department, Queen Margaret University, Edinburgh, United Kingdom (E.A.S.Al-D.). The current address for Dr Evans is Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee; the current address for Dr Wyrwoll is School of Anatomy, Physiology & Human Biology, The University of Western Australia, Crawley, Australia; and the current address for Dr Christensen is Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense
| | - Caitlin Wyrwoll
- From British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, United Kingdom (L.C.E., J.R.I., C.W., J.A.M., R.I.M., T.H.C., C.J.K., J.J.M., J.R.S., M.C.H., M.A.B.); and Dietetics, Nutrition and Biological Sciences Department, Queen Margaret University, Edinburgh, United Kingdom (E.A.S.Al-D.). The current address for Dr Evans is Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee; the current address for Dr Wyrwoll is School of Anatomy, Physiology & Human Biology, The University of Western Australia, Crawley, Australia; and the current address for Dr Christensen is Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense
| | - Julie A McNairn
- From British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, United Kingdom (L.C.E., J.R.I., C.W., J.A.M., R.I.M., T.H.C., C.J.K., J.J.M., J.R.S., M.C.H., M.A.B.); and Dietetics, Nutrition and Biological Sciences Department, Queen Margaret University, Edinburgh, United Kingdom (E.A.S.Al-D.). The current address for Dr Evans is Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee; the current address for Dr Wyrwoll is School of Anatomy, Physiology & Human Biology, The University of Western Australia, Crawley, Australia; and the current address for Dr Christensen is Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense
| | - Robert I Menzies
- From British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, United Kingdom (L.C.E., J.R.I., C.W., J.A.M., R.I.M., T.H.C., C.J.K., J.J.M., J.R.S., M.C.H., M.A.B.); and Dietetics, Nutrition and Biological Sciences Department, Queen Margaret University, Edinburgh, United Kingdom (E.A.S.Al-D.). The current address for Dr Evans is Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee; the current address for Dr Wyrwoll is School of Anatomy, Physiology & Human Biology, The University of Western Australia, Crawley, Australia; and the current address for Dr Christensen is Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense
| | - Thorbjørn H Christensen
- From British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, United Kingdom (L.C.E., J.R.I., C.W., J.A.M., R.I.M., T.H.C., C.J.K., J.J.M., J.R.S., M.C.H., M.A.B.); and Dietetics, Nutrition and Biological Sciences Department, Queen Margaret University, Edinburgh, United Kingdom (E.A.S.Al-D.). The current address for Dr Evans is Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee; the current address for Dr Wyrwoll is School of Anatomy, Physiology & Human Biology, The University of Western Australia, Crawley, Australia; and the current address for Dr Christensen is Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense
| | - Emad A S Al-Dujaili
- From British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, United Kingdom (L.C.E., J.R.I., C.W., J.A.M., R.I.M., T.H.C., C.J.K., J.J.M., J.R.S., M.C.H., M.A.B.); and Dietetics, Nutrition and Biological Sciences Department, Queen Margaret University, Edinburgh, United Kingdom (E.A.S.Al-D.). The current address for Dr Evans is Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee; the current address for Dr Wyrwoll is School of Anatomy, Physiology & Human Biology, The University of Western Australia, Crawley, Australia; and the current address for Dr Christensen is Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense
| | - Christopher J Kenyon
- From British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, United Kingdom (L.C.E., J.R.I., C.W., J.A.M., R.I.M., T.H.C., C.J.K., J.J.M., J.R.S., M.C.H., M.A.B.); and Dietetics, Nutrition and Biological Sciences Department, Queen Margaret University, Edinburgh, United Kingdom (E.A.S.Al-D.). The current address for Dr Evans is Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee; the current address for Dr Wyrwoll is School of Anatomy, Physiology & Human Biology, The University of Western Australia, Crawley, Australia; and the current address for Dr Christensen is Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense
| | - John J Mullins
- From British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, United Kingdom (L.C.E., J.R.I., C.W., J.A.M., R.I.M., T.H.C., C.J.K., J.J.M., J.R.S., M.C.H., M.A.B.); and Dietetics, Nutrition and Biological Sciences Department, Queen Margaret University, Edinburgh, United Kingdom (E.A.S.Al-D.). The current address for Dr Evans is Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee; the current address for Dr Wyrwoll is School of Anatomy, Physiology & Human Biology, The University of Western Australia, Crawley, Australia; and the current address for Dr Christensen is Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense
| | - Jonathan R Seckl
- From British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, United Kingdom (L.C.E., J.R.I., C.W., J.A.M., R.I.M., T.H.C., C.J.K., J.J.M., J.R.S., M.C.H., M.A.B.); and Dietetics, Nutrition and Biological Sciences Department, Queen Margaret University, Edinburgh, United Kingdom (E.A.S.Al-D.). The current address for Dr Evans is Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee; the current address for Dr Wyrwoll is School of Anatomy, Physiology & Human Biology, The University of Western Australia, Crawley, Australia; and the current address for Dr Christensen is Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense
| | - Megan C Holmes
- From British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, United Kingdom (L.C.E., J.R.I., C.W., J.A.M., R.I.M., T.H.C., C.J.K., J.J.M., J.R.S., M.C.H., M.A.B.); and Dietetics, Nutrition and Biological Sciences Department, Queen Margaret University, Edinburgh, United Kingdom (E.A.S.Al-D.). The current address for Dr Evans is Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee; the current address for Dr Wyrwoll is School of Anatomy, Physiology & Human Biology, The University of Western Australia, Crawley, Australia; and the current address for Dr Christensen is Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense
| | - Matthew A Bailey
- From British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, United Kingdom (L.C.E., J.R.I., C.W., J.A.M., R.I.M., T.H.C., C.J.K., J.J.M., J.R.S., M.C.H., M.A.B.); and Dietetics, Nutrition and Biological Sciences Department, Queen Margaret University, Edinburgh, United Kingdom (E.A.S.Al-D.). The current address for Dr Evans is Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee; the current address for Dr Wyrwoll is School of Anatomy, Physiology & Human Biology, The University of Western Australia, Crawley, Australia; and the current address for Dr Christensen is Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense.
| |
Collapse
|
33
|
Glucocorticoid-induced fetal origins of adult hypertension: Association with epigenetic events. Vascul Pharmacol 2016; 82:41-50. [PMID: 26903240 DOI: 10.1016/j.vph.2016.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/15/2016] [Accepted: 02/18/2016] [Indexed: 02/05/2023]
Abstract
Hypertension is a predominant risk factor for cardiovascular diseases and a major health care burden. Accumulating epidemiological and experimental evidence suggest that adult-onset hypertension may have its origins during early development. Upon exposure to glucocorticoids, the fetus develops hypertension, and the offspring may be programmed to continue the hypertensive trajectory into adulthood. Elevated oxidative stress and deranged nitric oxide system are not only hallmarks of adult hypertension but are also observed earlier in life. Endothelial dysfunction and remodeling of the vasculature, which are robustly associated with increased incidence of hypertension, are likely to have been pre-programmed during fetal life. Apparently, genomic, non-genomic, and epigenomic factors play a significant role in the development of hypertension, including glucocorticoid-driven effects on blood pressure. In this review, we discuss the involvement of the aforementioned participants in the pathophysiology of hypertension and suggest therapeutic opportunities for targeting epigenome modifiers, potentially for personalized medicine.
Collapse
|
34
|
Arnett MG, Muglia LM, Laryea G, Muglia LJ. Genetic Approaches to Hypothalamic-Pituitary-Adrenal Axis Regulation. Neuropsychopharmacology 2016; 41:245-60. [PMID: 26189452 PMCID: PMC4677126 DOI: 10.1038/npp.2015.215] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 07/09/2015] [Accepted: 07/09/2015] [Indexed: 01/12/2023]
Abstract
The normal function of the hypothalamic-pituitary-adrenal (HPA) axis, and resultant glucocorticoid (GC) secretion, is essential for human health. Disruption of GC regulation is associated with pathologic, psychological, and physiological disease states such as depression, post-traumatic stress disorder, hypertension, diabetes, and osteopenia, among others. As such, understanding the mechanisms by which HPA output is tightly regulated in its responses to environmental stressors and circadian cues has been an active area of investigation for decades. Over the last 20 years, however, advances in gene targeting and genome modification in rodent models have allowed the detailed dissection of roles for key molecular mediators and brain regions responsible for this control in vivo to emerge. Here, we summarize work done to elucidate the function of critical neuropeptide systems, GC-signaling targets, and inflammation-associated pathways in HPA axis regulation and behavior, and highlight areas for future investigation.
Collapse
Affiliation(s)
- Melinda G Arnett
- Cincinnati Children's Hospital Medical Center, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati, OH, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA,Cincinnati Children's Hospital Medical Center, Center for Prevention of Preterm Birth, Perinatal Institute, 3333 Burnet Avenue, MLC 7009, Attention Melinda Arnett, Cincinnati, OH 45229, USA, Tel: +1 513 803 8040, Fax: +1 513 803 5009, E-mail:
| | - Lisa M Muglia
- Cincinnati Children's Hospital Medical Center, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati, OH, USA
| | - Gloria Laryea
- Cincinnati Children's Hospital Medical Center, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati, OH, USA,Neuroscience Graduate Program Vanderbilt University, Nashville, TN, USA
| | - Louis J Muglia
- Cincinnati Children's Hospital Medical Center, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati, OH, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
35
|
Haque M, Wilson R, Sharma K, Mills NJ, Teruyama R. Localisation of 11β-Hydroxysteroid Dehydrogenase Type 2 in Mineralocorticoid Receptor Expressing Magnocellular Neurosecretory Neurones of the Rat Supraoptic and Paraventricular Nuclei. J Neuroendocrinol 2015; 27:835-49. [PMID: 26403275 PMCID: PMC5019266 DOI: 10.1111/jne.12325] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 09/15/2015] [Accepted: 09/17/2015] [Indexed: 01/29/2023]
Abstract
An accumulating body of evidence suggests that the activity of the mineralocorticoid, aldosterone, in the brain via the mineralocorticoid receptor (MR) plays an important role in the regulation of blood pressure. MR was recently found in vasopressin and oxytocin synthesising magnocellular neurosecretory cells (MNCs) in both the paraventricular (PVN) and supraoptic (SON) nuclei in the hypothalamus. Considering the physiological effects of these hormones, MR in these neurones may be an important site mediating the action of aldosterone in blood pressure regulation within the brain. However, aldosterone activation of MR in the hypothalamus remains controversial as a result of the high binding affinity of glucocorticoids to MR at substantially higher concentrations compared to aldosterone. In aldosterone-sensitive epithelia, the enzyme 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2) prevents glucocorticoids from binding to MR by converting glucocorticoids into inactive metabolites. The present study aimed to determine whether 11β-HSD2, which increases aldosterone selectivity, is expressed in MNCs. Specific 11β-HSD2 immunoreactivity was found in the cytoplasm of the MNCs in both the SON and PVN. In addition, double-fluorescence confocal microscopy demonstrated that MR-immunoreactivity and 11β-HSD2-in situ hybridised products are colocalised in MNCs. Lastly, single-cell reverse transcriptase-polymerase chain reaction detected MR and 11β-HSD2 mRNAs from cDNA libraries derived from single identified MNCs. These findings strongly suggest that MNCs in the SON and PVN are aldosterone-sensitive neurones.
Collapse
Affiliation(s)
- M Haque
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - R Wilson
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - K Sharma
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - N J Mills
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - R Teruyama
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|
36
|
Hunter RW, Ivy JR, Flatman PW, Kenyon CJ, Craigie E, Mullins LJ, Bailey MA, Mullins JJ. Hypertrophy in the Distal Convoluted Tubule of an 11β-Hydroxysteroid Dehydrogenase Type 2 Knockout Model. J Am Soc Nephrol 2015; 26:1537-48. [PMID: 25349206 PMCID: PMC4483573 DOI: 10.1681/asn.2013060634] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 08/12/2014] [Indexed: 11/03/2022] Open
Abstract
Na(+) transport in the renal distal convoluted tubule (DCT) by the thiazide-sensitive NaCl cotransporter (NCC) is a major determinant of total body Na(+) and BP. NCC-mediated transport is stimulated by aldosterone, the dominant regulator of chronic Na(+) homeostasis, but the mechanism is controversial. Transport may also be affected by epithelial remodeling, which occurs in the DCT in response to chronic perturbations in electrolyte homeostasis. Hsd11b2(-/-) mice, which lack the enzyme 11β-hydroxysteroid dehydrogenase type 2 (11βHSD2) and thus exhibit the syndrome of apparent mineralocorticoid excess, provided an ideal model in which to investigate the potential for DCT hypertrophy to contribute to Na(+) retention in a hypertensive condition. The DCTs of Hsd11b2(-/-) mice exhibited hypertrophy and hyperplasia and the kidneys expressed higher levels of total and phosphorylated NCC compared with those of wild-type mice. However, the striking structural and molecular phenotypes were not associated with an increase in the natriuretic effect of thiazide. In wild-type mice, Hsd11b2 mRNA was detected in some tubule segments expressing Slc12a3, but 11βHSD2 and NCC did not colocalize at the protein level. Thus, the phosphorylation status of NCC may not necessarily equate to its activity in vivo, and the structural remodeling of the DCT in the knockout mouse may not be a direct consequence of aberrant corticosteroid signaling in DCT cells. These observations suggest that the conventional concept of mineralocorticoid signaling in the DCT should be revised to recognize the complexity of NCC regulation by corticosteroids.
Collapse
Affiliation(s)
- Robert W Hunter
- British Heart Foundation Centre for Cardiovascular Science and
| | - Jessica R Ivy
- British Heart Foundation Centre for Cardiovascular Science and
| | - Peter W Flatman
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Eilidh Craigie
- British Heart Foundation Centre for Cardiovascular Science and
| | - Linda J Mullins
- British Heart Foundation Centre for Cardiovascular Science and
| | | | - John J Mullins
- British Heart Foundation Centre for Cardiovascular Science and
| |
Collapse
|
37
|
Mullins LJ, Kenyon CJ, Bailey MA, Conway BR, Diaz ME, Mullins JJ. Mineralocorticoid Excess or Glucocorticoid Insufficiency: Renal and Metabolic Phenotypes in a Rat Hsd11b2 Knockout Model. Hypertension 2015; 66:667-73. [PMID: 26077568 PMCID: PMC4847935 DOI: 10.1161/hypertensionaha.115.05262] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 05/27/2015] [Indexed: 01/06/2023]
Abstract
Obesity and hypertension are 2 major health issues of the 21st century. The syndrome of apparent mineralocorticoid excess is caused by deficiency of 11β-hydroxysteroid dehydrogenase type 2 (Hsd11b2), which normally inactivates glucocorticoids, rendering the mineralocorticoid receptor aldosterone–specific. The metabolic consequences of Hsd11b2 knockout in the rat are investigated in parallel with electrolyte homeostasis. Hsd11b2 was knocked out, by pronuclear microinjection of targeted zinc-finger nuclease mRNAs, and 1 line was characterized for its response to renal and metabolic challenges. Plasma 11-dehydrocorticosterone was below detection thresholds, and Hsd11b2 protein was undetected by Western blot, indicating complete ablation. Homozygotes were 13% smaller than wild-type littermates, and were polydipsic and polyuric. Their kidneys, adrenals, and hearts were significantly enlarged, but mesenteric fat pads and liver were significantly smaller. On a 0.3% Na diet, mean arterial blood pressure was ≈65 mm Hg higher than controls but only 25 mm Hg higher on a 0.03% Na+ diet. Urinary Na/K ratio of homozygotes was similar to controls on 0.3% Na+ diet but urinary albumin and calcium were elevated. Corticosterone and aldosterone levels showed normal circadian variation on both a 0.3% and 0.03% Na+ diet, but plasma renin was suppressed in homozygotes on both diets. Plasma glucose responses to an oral glucose challenge were reduced despite low circulating insulin, indicating much greater sensitivity to insulin in homozygotes. The rat model reveals mechanisms linking electrolyte homeostasis and metabolic control through the restriction of Hsd11b1 substrate availability.
Collapse
Affiliation(s)
- Linda J Mullins
- From the Molecular Physiology Laboratory, University of Edinburgh/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, Edinburgh, United Kingdom.
| | - Christopher J Kenyon
- From the Molecular Physiology Laboratory, University of Edinburgh/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Matthew A Bailey
- From the Molecular Physiology Laboratory, University of Edinburgh/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Bryan R Conway
- From the Molecular Physiology Laboratory, University of Edinburgh/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Mary E Diaz
- From the Molecular Physiology Laboratory, University of Edinburgh/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - John J Mullins
- From the Molecular Physiology Laboratory, University of Edinburgh/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, Edinburgh, United Kingdom
| |
Collapse
|
38
|
Ryu JH, Kim S, Lee JA, Han HY, Son HJ, Lee HJ, Kim YH, Kim JS, Park HG. Synthesis and optimization of picolinamide derivatives as a novel class of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) inhibitors. Bioorg Med Chem Lett 2015; 25:1679-1683. [DOI: 10.1016/j.bmcl.2015.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 02/23/2015] [Accepted: 03/03/2015] [Indexed: 11/15/2022]
|
39
|
Hunter RW, Bailey MA. Glucocorticoids and 11β-hydroxysteroid dehydrogenases: mechanisms for hypertension. Curr Opin Pharmacol 2015; 21:105-14. [DOI: 10.1016/j.coph.2015.01.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 01/20/2015] [Accepted: 01/22/2015] [Indexed: 11/26/2022]
|
40
|
Chen XQ, Shao LD, Pal M, Shen Y, Cheng X, Xu G, Peng LY, Wang K, Pan ZH, Li MM, Leng Y, He J, Zhao QS. Hupehenols A-E, selective 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) inhibitors from Viburnum hupehense. JOURNAL OF NATURAL PRODUCTS 2015; 78:330-334. [PMID: 25590374 DOI: 10.1021/np500896n] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Five selective 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) competitive inhibitors, hupehenols A-E (1-5), were isolated from Viburnum hupehense. The structure elucidation indicated that compounds 1-5 are new 20,21,22,23,24,25,26,27-octanordammarane triterpenoids. Their structures were established on the basis of NMR spectroscopic and mass spectrometric analysis. Hupehenols A-E (1-5) showed inhibition against human 11β-HSD1, with hupehenols B (2) and E (5) having IC50 values of 15.3 and 34.0 nM, respectively. Moreover, hupehenols C (3) and D (4) are highly selective inhibitors of human 11β-HSD1 when compared to murine 11β-HSD1.
Collapse
Affiliation(s)
- Xuan-Qin Chen
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences , Kunming 650204, Yunnan, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Livingstone DEW, Di Rollo EM, Yang C, Codrington LE, Mathews JA, Kara M, Hughes KA, Kenyon CJ, Walker BR, Andrew R. Relative adrenal insufficiency in mice deficient in 5α-reductase 1. J Endocrinol 2014; 222:257-66. [PMID: 24872577 PMCID: PMC4104038 DOI: 10.1530/joe-13-0563] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Patients with critical illness or hepatic failure exhibit impaired cortisol responses to ACTH, a phenomenon known as 'relative adrenal insufficiency'. A putative mechanism is that elevated bile acids inhibit inactivation of cortisol in liver by 5α-reductases type 1 and type 2 and 5β-reductase, resulting in compensatory downregulation of the hypothalamic-pituitary-adrenal axis and adrenocortical atrophy. To test the hypothesis that impaired glucocorticoid clearance can cause relative adrenal insufficiency, we investigated the consequences of 5α-reductase type 1 deficiency in mice. In adrenalectomised male mice with targeted disruption of 5α-reductase type 1, clearance of corticosterone was lower after acute or chronic (eightfold, P<0.05) administration, compared with WT control mice. In intact 5α-reductase-deficient male mice, although resting plasma corticosterone levels were maintained, corticosterone responses were impaired after ACTH administration (26% lower, P<0.05), handling stress (2.5-fold lower, P<0.05) and restraint stress (43% lower, P<0.05) compared with WT mice. mRNA levels of Nr3c1 (glucocorticoid receptor), Crh and Avp in pituitary or hypothalamus were altered, consistent with enhanced negative feedback. These findings confirm that impaired peripheral clearance of glucocorticoids can cause 'relative adrenal insufficiency' in mice, an observation with important implications for patients with critical illness or hepatic failure, and for patients receiving 5α-reductase inhibitors for prostatic disease.
Collapse
Affiliation(s)
- Dawn E W Livingstone
- EndocrinologyQueen's Medical Research Institute, University and British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Emma M Di Rollo
- EndocrinologyQueen's Medical Research Institute, University and British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Chenjing Yang
- EndocrinologyQueen's Medical Research Institute, University and British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Lucy E Codrington
- EndocrinologyQueen's Medical Research Institute, University and British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - John A Mathews
- EndocrinologyQueen's Medical Research Institute, University and British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Madina Kara
- EndocrinologyQueen's Medical Research Institute, University and British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Katherine A Hughes
- EndocrinologyQueen's Medical Research Institute, University and British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Christopher J Kenyon
- EndocrinologyQueen's Medical Research Institute, University and British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Brian R Walker
- EndocrinologyQueen's Medical Research Institute, University and British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Ruth Andrew
- EndocrinologyQueen's Medical Research Institute, University and British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
42
|
Vuorinen A, Engeli R, Meyer A, Bachmann F, Griesser UJ, Schuster D, Odermatt A. Ligand-based pharmacophore modeling and virtual screening for the discovery of novel 17β-hydroxysteroid dehydrogenase 2 inhibitors. J Med Chem 2014; 57:5995-6007. [PMID: 24960438 PMCID: PMC4111740 DOI: 10.1021/jm5004914] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
17β-Hydroxysteroid dehydrogenase 2 (17β-HSD2) catalyzes the inactivation of estradiol into estrone. This enzyme is expressed only in a few tissues, and therefore its inhibition is considered as a treatment option for osteoporosis to ameliorate estrogen deficiency. In this study, ligand-based pharmacophore models for 17β-HSD2 inhibitors were constructed and employed for virtual screening. From the virtual screening hits, 29 substances were evaluated in vitro for 17β-HSD2 inhibition. Seven compounds inhibited 17β-HSD2 with low micromolar IC50 values. To investigate structure-activity relationships (SAR), 30 more derivatives of the original hits were tested. The three most potent hits, 12, 22, and 15, had IC50 values of 240 nM, 1 μM, and 1.5 μM, respectively. All but 1 of the 13 identified inhibitors were selective over 17β-HSD1, the enzyme catalyzing conversion of estrone into estradiol. Three of the new, small, synthetic 17β-HSD2 inhibitors showed acceptable selectivity over other related HSDs, and six of them did not affect other HSDs.
Collapse
Affiliation(s)
- Anna Vuorinen
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck - CMBI, University of Innsbruck , Innrain 80/82, 6020 Innsbruck, Austria
| | | | | | | | | | | | | |
Collapse
|
43
|
Branchi I, Cirulli F. Early experiences: Building up the tools to face the challenges of adult life. Dev Psychobiol 2014; 56:1661-74. [DOI: 10.1002/dev.21235] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 06/06/2014] [Indexed: 12/17/2022]
Affiliation(s)
- Igor Branchi
- Section of Behavioural Neurosciences; Department of Cell Biology and Neurosciences; Istituto Superiore di Sanità, Viale Regina Elena 299 00161 Rome Italy
- Institute of Anatomy; University of Zurich; 190 Winterthurestrasse 8057 Zurich Switzerland
| | - Francesca Cirulli
- Section of Behavioural Neurosciences; Department of Cell Biology and Neurosciences; Istituto Superiore di Sanità, Viale Regina Elena 299 00161 Rome Italy
| |
Collapse
|
44
|
Rezaei M, Andrieu T, Neuenschwander S, Bruggmann R, Mordasini D, Frey FJ, Vogt B, Frey BM. Regulation of 11β-hydroxysteroid dehydrogenase type 2 by microRNA. Hypertension 2014; 64:860-6. [PMID: 24980668 DOI: 10.1161/hypertensionaha.114.00002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The enzyme 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2) is selectively expressed in aldosterone target tissues, conferring aldosterone selectivity for the mineralocorticoid receptor. A diminished activity causes salt-sensitive hypertension. The mechanism of the variable and distinct 11β-hydroxysteroid dehydrogenase type 2 gene (HSD11B2) expression in the cortical collecting duct is poorly understood. Here, we analyzed for the first time whether the 11β-HSD2 expression is modulated by microRNAs (miRNAs). In silico analysis revealed 53 and 27 miRNAs with potential binding sites on human or rat HSD11B2 3'-untranslated region. A reporter assay demonstrated 3'-untranslated region-dependent regulation of human and rodent HSD11B2. miRNAs were profiled from cortical collecting ducts and proximal convoluted tubules. Bioinformatic analyses showed a distinct clustering for cortical collecting ducts and proximal convoluted tubules with 53 of 375 miRNAs, where 13 were predicted to bind to the rat HSD11B2 3'-untranslated region. To gain insight into potentially relevant miRNAs in vivo, we investigated 2 models with differential 11β-HSD2 activity linked with salt-sensitive hypertension. (1) Comparing Sprague-Dawley with low and Wistar rats with high 11β-HSD2 activity revealed rno-miR-20a-5p, rno-miR-19b-3p, and rno-miR-190a-5p to be differentially expressed. (2) Uninephrectomy lowered 11β-HSD2 activity in the residual kidney with differentially expressed rno-miR-19b-3p, rno-miR-29b-3p, and rno-miR-26-5p. In conclusion, miRNA-dependent mechanisms seem to modulate 11β-HSD2 dosage in health and disease states.
Collapse
Affiliation(s)
- Mina Rezaei
- From the Department of Nephrology, Hypertension, and Clinical Pharmacology (M.R., T.A., D.M., F.J.F., B.V., B.M.F.) and Department of Biology and Bioinformatics (S.N., R.B.), University of Bern, Bern, Switzerland; Vital-IT, Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland (S.N.); and Department of Clinical Research, University Hospital Bern, Bern, Switzerland (B.M.F.)
| | - Thomas Andrieu
- From the Department of Nephrology, Hypertension, and Clinical Pharmacology (M.R., T.A., D.M., F.J.F., B.V., B.M.F.) and Department of Biology and Bioinformatics (S.N., R.B.), University of Bern, Bern, Switzerland; Vital-IT, Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland (S.N.); and Department of Clinical Research, University Hospital Bern, Bern, Switzerland (B.M.F.)
| | - Samuel Neuenschwander
- From the Department of Nephrology, Hypertension, and Clinical Pharmacology (M.R., T.A., D.M., F.J.F., B.V., B.M.F.) and Department of Biology and Bioinformatics (S.N., R.B.), University of Bern, Bern, Switzerland; Vital-IT, Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland (S.N.); and Department of Clinical Research, University Hospital Bern, Bern, Switzerland (B.M.F.)
| | - Rémy Bruggmann
- From the Department of Nephrology, Hypertension, and Clinical Pharmacology (M.R., T.A., D.M., F.J.F., B.V., B.M.F.) and Department of Biology and Bioinformatics (S.N., R.B.), University of Bern, Bern, Switzerland; Vital-IT, Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland (S.N.); and Department of Clinical Research, University Hospital Bern, Bern, Switzerland (B.M.F.)
| | - David Mordasini
- From the Department of Nephrology, Hypertension, and Clinical Pharmacology (M.R., T.A., D.M., F.J.F., B.V., B.M.F.) and Department of Biology and Bioinformatics (S.N., R.B.), University of Bern, Bern, Switzerland; Vital-IT, Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland (S.N.); and Department of Clinical Research, University Hospital Bern, Bern, Switzerland (B.M.F.)
| | - Felix J Frey
- From the Department of Nephrology, Hypertension, and Clinical Pharmacology (M.R., T.A., D.M., F.J.F., B.V., B.M.F.) and Department of Biology and Bioinformatics (S.N., R.B.), University of Bern, Bern, Switzerland; Vital-IT, Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland (S.N.); and Department of Clinical Research, University Hospital Bern, Bern, Switzerland (B.M.F.)
| | - Bruno Vogt
- From the Department of Nephrology, Hypertension, and Clinical Pharmacology (M.R., T.A., D.M., F.J.F., B.V., B.M.F.) and Department of Biology and Bioinformatics (S.N., R.B.), University of Bern, Bern, Switzerland; Vital-IT, Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland (S.N.); and Department of Clinical Research, University Hospital Bern, Bern, Switzerland (B.M.F.)
| | - Brigitte M Frey
- From the Department of Nephrology, Hypertension, and Clinical Pharmacology (M.R., T.A., D.M., F.J.F., B.V., B.M.F.) and Department of Biology and Bioinformatics (S.N., R.B.), University of Bern, Bern, Switzerland; Vital-IT, Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland (S.N.); and Department of Clinical Research, University Hospital Bern, Bern, Switzerland (B.M.F.)
| |
Collapse
|
45
|
King S, Bray S, Galbraith S, Christie L, Fleming S. Evidence for aldosterone-dependent growth of renal cell carcinoma. Int J Exp Pathol 2014; 95:244-50. [PMID: 24802662 DOI: 10.1111/iep.12074] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 01/29/2014] [Indexed: 12/12/2022] Open
Abstract
The aim if this study was to investigate the hypothesis that K-RAS 4A is upregulated in a mineralocorticoid-dependent manner in renal cell carcinoma and that this supports the proliferation and survival of some renal cancers. Expression of the K-RAS in renal tumour tissues and cell lines was examined by real-time PCR and Western blot and mineralocorticoid receptor, and its gatekeeper enzyme 11β-hydroxysteroid dehydrogenase-2 was examined by immunocytochemistry on a tissue microarray of 27 cases of renal cell carcinoma. Renal cancer cells lines 04A018 (RCC4 plus VHL) and 04A019 (RCC4 plus vector alone) were examined for the expression of K-RAS4A and for the effect on K-RAS expression of spironolactone blockade of the mineralocorticoid receptor. K-RAS4A was suppressed by siRNA, and the effect on cell survival, proliferation and activation of the Akt and Raf signalling pathways was investigated in vitro. K-RAS4A was expressed in RCC tissue and in the renal cancer cell lines but K-RAS was downregulated by spironolactone and upregulated by aldosterone. Spironolactone treatment and K-RAS suppression both led to a reduction in cell number in vitro. Both Akt and Raf pathways showed activation which was dependent on K-RAS expression. K-RAS expression in renal cell carcinoma is at least partially induced by aldosterone. Aldosterone supports the survival and proliferation of RCC cells by upregulation of K-RAS acting through the Akt and Raf pathways.
Collapse
Affiliation(s)
- Sharon King
- Division of Cancer Research, Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | | | | | | | | |
Collapse
|
46
|
Lathe R, Kotelevtsev Y. Steroid signaling: ligand-binding promiscuity, molecular symmetry, and the need for gating. Steroids 2014; 82:14-22. [PMID: 24462647 DOI: 10.1016/j.steroids.2014.01.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 12/03/2013] [Accepted: 01/06/2014] [Indexed: 11/28/2022]
Abstract
Steroid/sterol-binding receptors and enzymes are remarkably promiscuous in the range of ligands they can bind to and, in the case of enzymes, modify - raising the question of how specific receptor activation is achieved in vivo. Estrogen receptors (ER) are modulated by 27-hydroxycholesterol and 5α-androstane-3β,17β-diol (Adiol), in addition to estradiol (E2), and respond to diverse small molecules such as bisphenol A. Steroid-modifying enzymes are also highly promiscuous in ligand binding and metabolism. The specificity problem is compounded by the fact that the steroid core (hydrogenated cyclopentophenanthrene ring system) has several planes of symmetry. Ligand binding can be in symmetrical East-West (rotation) and North-South (inversion) orientations. Hydroxysteroid dehydrogenases (HSDs) can modify symmetrical 7 and 11, also 3 and 17/20, positions, exemplified here by yeast 3α,20β-HSD and mammalian 11β-HSD and 17β-HSD enzymes. Faced with promiscuity and symmetry, other strategies are clearly necessary to promote signaling selectivity in vivo. Gating regulates hormone access via enzymes that preferentially inactivate (or activate) a subclass of ligands, thereby governing which ligands gain receptor access - exemplified by 11β-HSD gating cortisol access to the mineralocorticoid receptor, and P450 CYP7B1 gating Adiol access to ER. Counter-intuitively, the specificity of steroid/sterol action is achieved not by intrinsic binding selectivity but by the combination of local metabolism and binding affinity.
Collapse
Affiliation(s)
- Richard Lathe
- State University of Pushchino, Prospekt Nauki, Pushchino 142290, Moscow Region, Russia; Pushchino Branch of the Institute of Bio-Organic Chemistry, Russian Academy of Sciences, Pushchino 142290, Moscow Region, Russia; Pieta Research, PO Box 27069, Edinburgh EH10 5YW, UK.
| | - Yuri Kotelevtsev
- State University of Pushchino, Prospekt Nauki, Pushchino 142290, Moscow Region, Russia; Pushchino Branch of the Institute of Bio-Organic Chemistry, Russian Academy of Sciences, Pushchino 142290, Moscow Region, Russia; Biomedical Centre for Research Education and Innovation (CREI), Skolkovo Institute of Science and Technology, 143025 Skolkovo, Russia; Queens Medical Research Institute, University of Edinburgh, Little France, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
47
|
Cottrell EC, Seckl JR, Holmes MC, Wyrwoll CS. Foetal and placental 11β-HSD2: a hub for developmental programming. Acta Physiol (Oxf) 2014; 210:288-95. [PMID: 24330050 DOI: 10.1111/apha.12187] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 07/31/2013] [Accepted: 10/23/2013] [Indexed: 01/01/2023]
Abstract
Foetal growth restriction (FGR), reflective of an adverse intrauterine environment, confers a significantly increased risk of perinatal mortality and morbidity. In addition, low birthweight associates with adult diseases including hypertension, metabolic dysfunction and behavioural disorders. A key mechanism underlying FGR is exposure of the foetus to glucocorticoids which, while critical for foetal development, in excess can reduce foetal growth and permanently alter organ structure and function, predisposing to disease in later life. Foetal glucocorticoid exposure is regulated, at least in part, by the enzyme 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2), which catalyses the intracellular inactivation of glucocorticoids. This enzyme is highly expressed within the placenta at the maternal-foetal interface, limiting the passage of glucocorticoids to the foetus. Expression of 11β-HSD2 is also high in foetal tissues, particularly within the developing central nervous system. Down-regulation or genetic deficiency of placental 11β-HSD2 is associated with significant reductions in foetal growth and birth weight, and programmed outcomes in adulthood. To unravel the direct significance of 11β-HSD2 for developmental programming, placental function, neurodevelopment and adult behaviour have been extensively investigated in a mouse knockout of 11β-HSD2. This review highlights the evidence obtained from this mouse model for a critical role of feto-placental 11β-HSD2 in determining the adverse programming outcomes.
Collapse
Affiliation(s)
- E. C. Cottrell
- Maternal and Fetal Health Research Centre; Institute of Human Development; University of Manchester; Manchester UK
| | - J. R. Seckl
- University/BHF Centre for Cardiovascular Science; University of Edinburgh; Edinburgh UK
| | - M. C. Holmes
- University/BHF Centre for Cardiovascular Science; University of Edinburgh; Edinburgh UK
| | - C. S. Wyrwoll
- School of Anatomy, Physiology and Human Biology; University of Western Australia; Crawley WA Australia
| |
Collapse
|
48
|
Goldberg FW, Dossetter AG, Scott JS, Robb GR, Boyd S, Groombridge SD, Kemmitt PD, Sjögren T, Gutierrez PM, deSchoolmeester J, Swales JG, Turnbull AV, Wild MJ. Optimization of Brain Penetrant 11β-Hydroxysteroid Dehydrogenase Type I Inhibitors and in Vivo Testing in Diet-Induced Obese Mice. J Med Chem 2014; 57:970-86. [DOI: 10.1021/jm4016729] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
| | | | - James S. Scott
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Graeme R. Robb
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Scott Boyd
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Sam D. Groombridge
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Paul D. Kemmitt
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Tove Sjögren
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | | | | | - John G. Swales
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Andrew V. Turnbull
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Martin J. Wild
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| |
Collapse
|
49
|
Ye L, Guo J, Ge RS. Environmental pollutants and hydroxysteroid dehydrogenases. VITAMINS AND HORMONES 2014; 94:349-90. [PMID: 24388197 DOI: 10.1016/b978-0-12-800095-3.00013-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hydroxysteroid dehydrogenases (HSD) are a group of steroidogenic enzymes that are involved in the steroid biosynthesis and metabolism. Four classes of HSDs, namely, 3β-, 11β-, 17β-, and 20α-HSDs, are discussed. 3β-HSDs catalyze the conversion of pregnenolone, 17α-hydroxypregnenolone, and dehydroepiandrosterone to progesterone, 17α-hydroxyprogesterone, and androstenedione, respectively. 11β-HSDs catalyze the interconversion between active cortisol and inactive cortisone. 17β-HSDs catalyze the interconversion between 17β-hydroxyl steroids and 17-ketoandrogens and estrogens. 20α-HSDs catalyze the conversion of progesterone into 20α-hydroxyprogesterone. Many environmental pollutants directly inhibit one or more enzymes of these HSDs, thus interfering with endogenous active steroid hormone levels. These chemicals include industrial materials (perfluoroalkyl compounds, phthalates, bisphenol A, and benzophenone), pesticides/biocides (methoxychlor, organotins, 1,2-dibromo-3-chloropropane, and prochloraz), and plant constituents (genistein, gossypol, and licorice). This chapter reviews these inhibitors targeting on HSDs.
Collapse
Affiliation(s)
- Leping Ye
- The 2nd Affiliated Hospital and Research Academy of Reproductive Biomedicine of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Jingjing Guo
- The 2nd Affiliated Hospital and Research Academy of Reproductive Biomedicine of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Ren-Shan Ge
- The 2nd Affiliated Hospital and Research Academy of Reproductive Biomedicine of Wenzhou Medical University, Wenzhou, Zhejiang, PR China.
| |
Collapse
|
50
|
McCoull W, Augustin M, Blake C, Ertan A, Kilgour E, Krapp S, Moore JE, Newcombe NJ, Packer MJ, Rees A, Revill J, Scott JS, Selmi N, Gerhardt S, Ogg DJ, Steinbacher S, Whittamore PRO. Identification and optimisation of 3,3-dimethyl-azetidin-2-ones as potent and selective inhibitors of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1). MEDCHEMCOMM 2014. [DOI: 10.1039/c3md00234a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|