1
|
Yu P, Yuan H, Li X, Chen H. Impact of cortisol on liver fat and metabolic health in adrenal incidentalomas and Cushing's syndrome. Endocrine 2024:10.1007/s12020-024-04043-4. [PMID: 39320593 DOI: 10.1007/s12020-024-04043-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/14/2024] [Indexed: 09/26/2024]
Abstract
OBJECTIVE To evaluate liver fat content in patients with non-functional adrenal incidentalomas (NFAI), mild autonomous cortisol secretion (MACS), and Cushing's syndrome (CS), and assess its relationship with cortisol levels. METHODS This cross-sectional study used retrospective data from 103 NFAI patients, 100 MACS (serum cortisol after a 1-mg dexamethasone test >50 nmol/L), and 59 with CS. Abdominal CT scans measured hepatic and splenic CT values to calculate the liver-to-spleen (L/S) ratio. Metabolic indicators including fasting plasma glucose (FPG), LDL-c, HDL-c, HbA1c, etc were measured. Mediation analysis was used to explore the indirect effects of metabolic traits on the cortisol-liver fat relationship. RESULTS Patients included 103 NFAI, 100 MACS, and 59 CS. MACS patients had higher NAFLD prevalence (57%) than NFAI (26.2%, p < 0.001) but lower than CS (66.1%, p < 0.001). MACS and CS were associated with NAFLD (OR 3.83 and OR 5.73, p < 0.01), adjusted for age, body mass index (BMI), and covariates. Midnight serum cortisol correlated with L/S ratio (p < 0.001). HbA1c and Triglyceride-glucose index (TyG) mediated 24.5% and 49.5% of the cortisol and L/S ratio association, respectively. FPG, HbA1c, HDL-c, and TyG mediated the association between MACS or CS and the L/S ratio. Homeostasis model assessment of insulin resistance (HOMA-IR), fructosamine, and triglycerides mediated for MACS, while alkaline phosphatase did so for CS. Total cholesterol, LDL-c, ALT, AST, γ-GT, insulin, and uric acid did not mediate the association. CONCLUSION MACS and CS are linked to significant metabolic disturbances, including increased liver fat and impaired glucose and lipid metabolism, contributing to fatty liver.
Collapse
Affiliation(s)
- Peng Yu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Endocrinology and Metabolism, Shanghai Geriatric Medical Center, Shanghai, China
| | - Haoyue Yuan
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaomu Li
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Hong Chen
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Martinez GJ, Appleton M, Kipp ZA, Loria AS, Min B, Hinds TD. Glucocorticoids, their uses, sexual dimorphisms, and diseases: new concepts, mechanisms, and discoveries. Physiol Rev 2024; 104:473-532. [PMID: 37732829 PMCID: PMC11281820 DOI: 10.1152/physrev.00021.2023] [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: 05/22/2023] [Revised: 08/07/2023] [Accepted: 09/10/2023] [Indexed: 09/22/2023] Open
Abstract
The normal stress response in humans is governed by the hypothalamic-pituitary-adrenal (HPA) axis through heightened mechanisms during stress, raising blood levels of the glucocorticoid hormone cortisol. Glucocorticoids are quintessential compounds that balance the proper functioning of numerous systems in the mammalian body. They are also generated synthetically and are the preeminent therapy for inflammatory diseases. They act by binding to the nuclear receptor transcription factor glucocorticoid receptor (GR), which has two main isoforms (GRα and GRβ). Our classical understanding of glucocorticoid signaling is from the GRα isoform, which binds the hormone, whereas GRβ has no known ligands. With glucocorticoids being involved in many physiological and cellular processes, even small disruptions in their release via the HPA axis, or changes in GR isoform expression, can have dire ramifications on health. Long-term chronic glucocorticoid therapy can lead to a glucocorticoid-resistant state, and we deliberate how this impacts disease treatment. Chronic glucocorticoid treatment can lead to noticeable side effects such as weight gain, adiposity, diabetes, and others that we discuss in detail. There are sexually dimorphic responses to glucocorticoids, and women tend to have a more hyperresponsive HPA axis than men. This review summarizes our understanding of glucocorticoids and critically analyzes the GR isoforms and their beneficial and deleterious mechanisms and the sexual differences that cause a dichotomy in responses. We also discuss the future of glucocorticoid therapy and propose a new concept of dual GR isoform agonist and postulate why activating both isoforms may prevent glucocorticoid resistance.
Collapse
Affiliation(s)
- Genesee J Martinez
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Malik Appleton
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Zachary A Kipp
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Analia S Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
- Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Booki Min
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
- Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
3
|
Yoon JY, Kim JY, Kim HJ, Ka NL, Lee SH, Lee MO. LncRNA Ctcflos modulates glucocorticoid receptor-mediated induction of hepatic phosphoenolpyruvate carboxykinase in mice. Life Sci 2022; 312:121254. [PMID: 36470542 DOI: 10.1016/j.lfs.2022.121254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/16/2022] [Accepted: 11/26/2022] [Indexed: 12/12/2022]
Affiliation(s)
- Jae-Yeun Yoon
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Ju-Yeon Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Hyeon-Ji Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Na-Lee Ka
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sang-Heon Lee
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Mi-Ock Lee
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea; Bio-MAX institute, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Nuclear Receptors in Energy Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1390:61-82. [DOI: 10.1007/978-3-031-11836-4_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
5
|
Kokkinopoulou I, Diakoumi A, Moutsatsou P. Glucocorticoid Receptor Signaling in Diabetes. Int J Mol Sci 2021; 22:ijms222011173. [PMID: 34681832 PMCID: PMC8537243 DOI: 10.3390/ijms222011173] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/04/2021] [Accepted: 10/12/2021] [Indexed: 12/20/2022] Open
Abstract
Stress and depression increase the risk of Type 2 Diabetes (T2D) development. Evidence demonstrates that the Glucocorticoid (GC) negative feedback is impaired (GC resistance) in T2D patients resulting in Hypothalamic-Pituitary-Adrenal (HPA) axis hyperactivity and hypercortisolism. High GCs, in turn, activate multiple aspects of glucose homeostasis in peripheral tissues leading to hyperglycemia. Elucidation of the underlying molecular mechanisms revealed that Glucocorticoid Receptor (GR) mediates the GC-induced dysregulation of glucose production, uptake and insulin signaling in GC-sensitive peripheral tissues, such as liver, skeletal muscle, adipose tissue, and pancreas. In contrast to increased GR peripheral sensitivity, an impaired GR signaling in Peripheral Blood Mononuclear Cells (PBMCs) of T2D patients, associated with hyperglycemia, hyperlipidemia, and increased inflammation, has been shown. Given that GR changes in immune cells parallel those in brain, the above data implicate that a reduced brain GR function may be the biological link among stress, HPA hyperactivity, hypercortisolism and hyperglycemia. GR polymorphisms have also been associated with metabolic disturbances in T2D while dysregulation of micro-RNAs—known to target GR mRNA—has been described. Collectively, GR has a crucial role in T2D, acting in a cell-type and context-specific manner, leading to either GC sensitivity or GC resistance. Selective modulation of GR signaling in T2D therapy warrants further investigation.
Collapse
|
6
|
Molecular Mechanisms of Glucocorticoid-Induced Insulin Resistance. Int J Mol Sci 2021; 22:ijms22020623. [PMID: 33435513 PMCID: PMC7827500 DOI: 10.3390/ijms22020623] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/29/2020] [Accepted: 01/02/2021] [Indexed: 12/12/2022] Open
Abstract
Glucocorticoids (GCs) are steroids secreted by the adrenal cortex under the hypothalamic-pituitary-adrenal axis control, one of the major neuro-endocrine systems of the organism. These hormones are involved in tissue repair, immune stability, and metabolic processes, such as the regulation of carbohydrate, lipid, and protein metabolism. Globally, GCs are presented as ‘flight and fight’ hormones and, in that purpose, they are catabolic hormones required to mobilize storage to provide energy for the organism. If acute GC secretion allows fast metabolic adaptations to respond to danger, stress, or metabolic imbalance, long-term GC exposure arising from treatment or Cushing’s syndrome, progressively leads to insulin resistance and, in fine, cardiometabolic disorders. In this review, we briefly summarize the pharmacological actions of GC and metabolic dysregulations observed in patients exposed to an excess of GCs. Next, we describe in detail the molecular mechanisms underlying GC-induced insulin resistance in adipose tissue, liver, muscle, and to a lesser extent in gut, bone, and brain, mainly identified by numerous studies performed in animal models. Finally, we present the paradoxical effects of GCs on beta cell mass and insulin secretion by the pancreas with a specific focus on the direct and indirect (through insulin-sensitive organs) effects of GCs. Overall, a better knowledge of the specific action of GCs on several organs and their molecular targets may help foster the understanding of GCs’ side effects and design new drugs that possess therapeutic benefits without metabolic adverse effects.
Collapse
|
7
|
Chen S, Sbuh N, Veedu RN. Antisense Oligonucleotides as Potential Therapeutics for Type 2 Diabetes. Nucleic Acid Ther 2020; 31:39-57. [PMID: 33026966 DOI: 10.1089/nat.2020.0891] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes (T2D) is a chronic metabolic disorder characterized by persistent hyperglycemia resulting from inefficient signaling and insufficient production of insulin. Conventional management of T2D has largely relied on small molecule-based oral hypoglycemic medicines, which do not halt the progression of the disease due to limited efficacy and induce adverse effects as well. To this end, antisense oligonucleotide has attracted immense attention in developing antidiabetic agents because of their ability to downregulate the expression of disease-causing genes at the RNA and protein level. To date, seven antisense agents have been approved by the United States Food and Drug Administration for therapies of a variety of human maladies, including genetic disorders. Herein, we provide a comprehensive review of antisense molecules developed for suppressing the causative genes believed to be responsible for insulin resistance and hyperglycemia toward preventing and treating T2D.
Collapse
Affiliation(s)
- Suxiang Chen
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Australia.,Perron Institute for Neurological and Translational Science, Perth, Australia
| | - Nabayet Sbuh
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Australia.,Perron Institute for Neurological and Translational Science, Perth, Australia
| | - Rakesh N Veedu
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Australia.,Perron Institute for Neurological and Translational Science, Perth, Australia
| |
Collapse
|
8
|
Lakshmi J, Mukhopadhyay K, Ramaswamy P, Mahadevan S. A Systematic Review on Organophosphate Pesticide and Type II Diabetes Mellitus. Curr Diabetes Rev 2020; 16:586-597. [PMID: 31544698 DOI: 10.2174/1573399815666190712192844] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/25/2019] [Accepted: 06/22/2019] [Indexed: 12/17/2022]
Abstract
Organophosphate (OP) pesticides are extremely poisonous and they affect the glucose breakdown in numerous and mechanism. There are higher evidence of stimulating diabetes mellitus through OP pesticides especially the type II diabetes. The upsurge in the level of glucose (hyperglycemia), and insulin resistance along with their related outcomes are discussed in this review. The data related to investigational and clinical techniques endorse a connection amid such molecular mechanism and compounds of OPs. Numerous studies conducted till March 2018 have reported OP' exposures and diabetes-related outcomes. The acute and chronic exposure in case of these insecticides and diabetesrelated outcomes are defined in this study. Initially, it was declared that OPs prompt to hyperglycemia. Then, a high association of glucose in blood beside insulin was found out. The affirmation from some clinical as well as investigational studies supported a connection amid exposure to OP and diabetes, yet in maximum number of instances, non-specific diabetes occurs.
Collapse
Affiliation(s)
- Jothi Lakshmi
- Department of Environmental Health Engineering, Sri Ramachandra Medical College and Research Institute, Chennai, India
| | - Krishnendu Mukhopadhyay
- Department of Environmental Health Engineering, Sri Ramachandra Medical College and Research Institute, Chennai, India
| | - Padmavathi Ramaswamy
- Department of Physiology, Sri Ramachandra Medical College and Research Institute, Chennai, India
| | - Shriraam Mahadevan
- Department of Endocrinology, Sri Ramachandra Medical College and Research Institute, Chennai, India
| |
Collapse
|
9
|
Czajka M, Matysiak-Kucharek M, Jodłowska-Jędrych B, Sawicki K, Fal B, Drop B, Kruszewski M, Kapka-Skrzypczak L. Organophosphorus pesticides can influence the development of obesity and type 2 diabetes with concomitant metabolic changes. ENVIRONMENTAL RESEARCH 2019; 178:108685. [PMID: 31479978 DOI: 10.1016/j.envres.2019.108685] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 08/09/2019] [Accepted: 08/21/2019] [Indexed: 06/10/2023]
Abstract
Widespread use and the bioaccumulation of pesticides in the environment lead to the contamination of air, water, soil and agricultural resources. A huge body of evidence points to the association between the pesticide exposure and increase in the incidence of chronic diseases, e.g. cancer, birth defects, reproductive disorders, neurodegenerative, cardiovascular and respiratory diseases, developmental disorders, metabolic disorders, chronic renal disorders or autoimmune diseases. Organophosphorus compounds are among the most widely used pesticides. A growing body of evidence is suggesting the potential interdependence between the organophosphorus pesticides (OPs) exposure and risk of obesity and type 2 diabetes mellitus (T2DM). This article reviews the current literature to highlight the latest in vitro and in vivo evidences on the possible influence of OPs on obesity and T2DM development, as well as epidemiological evidence for the metabolic toxicity of OPs in humans. The article also draws attention to the influence of maternal OPs exposure on offspring. Summarized studies suggest that OPs exposure is associated with metabolic changes linked with obesity and T2DM indicated that such exposures may increase risk or vulnerability to other contributory components.
Collapse
Affiliation(s)
- Magdalena Czajka
- Department of Molecular Biology and Translational Research, Institute of Rural Health, 20-090, Lublin, Poland.
| | - Magdalena Matysiak-Kucharek
- Department of Molecular Biology and Translational Research, Institute of Rural Health, 20-090, Lublin, Poland
| | - Barbara Jodłowska-Jędrych
- Department of Histology and Embryology with Experimental Cytology Unit, Medical University of Lublin, 20-080, Lublin, Poland
| | - Krzysztof Sawicki
- Department of Molecular Biology and Translational Research, Institute of Rural Health, 20-090, Lublin, Poland
| | - Berta Fal
- Department of Molecular Biology and Translational Research, Institute of Rural Health, 20-090, Lublin, Poland
| | - Bartłomiej Drop
- Department of Medical Informatics and Statistics with E-learning Lab, Medical University of Lublin, 20-090, Lublin, Poland
| | - Marcin Kruszewski
- Department of Molecular Biology and Translational Research, Institute of Rural Health, 20-090, Lublin, Poland; Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195, Warsaw, Poland
| | - Lucyna Kapka-Skrzypczak
- Department of Molecular Biology and Translational Research, Institute of Rural Health, 20-090, Lublin, Poland.
| |
Collapse
|
10
|
Joshi AKR, Kandlakunta B, Kotturu SK, Ghosh S. Antiglucocorticoid potential of nutraceuticals: In silico molecular docking and in vitro assessment. J Food Biochem 2018. [DOI: 10.1111/jfbc.12522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Apurva Kumar Ramesh Joshi
- Food Chemistry Division; National Institute of Nutrition, Jamai-Osmania; Telangana Hyderabad 500007 India
| | - Bhaskarachary Kandlakunta
- Food Chemistry Division; National Institute of Nutrition, Jamai-Osmania; Telangana Hyderabad 500007 India
| | - Sandeep Kumar Kotturu
- Division of Molecular Biology; National Institute of Nutrition, Jamai-Osmania; Telangana Hyderabad 500007 India
| | - Sudip Ghosh
- Division of Molecular Biology; National Institute of Nutrition, Jamai-Osmania; Telangana Hyderabad 500007 India
| |
Collapse
|
11
|
Abstract
Glucocorticoid hormones (GC) regulate essential physiological functions including energy homeostasis, embryonic and postembryonic development, and the stress response. From the biomedical perspective, GC have garnered a tremendous amount of attention as highly potent anti-inflammatory and immunosuppressive medications indispensable in the clinic. GC signal through the GC receptor (GR), a ligand-dependent transcription factor whose structure, DNA binding, and the molecular partners that it employs to regulate transcription have been under intense investigation for decades. In particular, next-generation sequencing-based approaches have revolutionized the field by introducing a unified platform for a simultaneous genome-wide analysis of cellular activities at the level of RNA production, binding of transcription factors to DNA and RNA, and chromatin landscape and topology. Here we describe fundamental concepts of GC/GR function as established through traditional molecular and in vivo approaches and focus on the novel insights of GC biology that have emerged over the last 10 years from the rapidly expanding arsenal of system-wide genomic methodologies.
Collapse
Affiliation(s)
- Maria A Sacta
- Hospital for Special Surgery, The David Rosensweig Genomics Center, New York, NY 10021; .,Weill Cornell/Rockefeller/Sloan Kettering MD/PhD program, New York, NY 10021
| | - Yurii Chinenov
- Hospital for Special Surgery, The David Rosensweig Genomics Center, New York, NY 10021;
| | - Inez Rogatsky
- Hospital for Special Surgery, The David Rosensweig Genomics Center, New York, NY 10021; .,Weill Cornell/Rockefeller/Sloan Kettering MD/PhD program, New York, NY 10021
| |
Collapse
|
12
|
Priyadarshini E, Anuradha CV. Glucocorticoid Antagonism Reduces Insulin Resistance and Associated Lipid Abnormalities in High-Fructose-Fed Mice. Can J Diabetes 2016; 41:41-51. [PMID: 27614803 DOI: 10.1016/j.jcjd.2016.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 06/07/2016] [Accepted: 06/13/2016] [Indexed: 01/17/2023]
Abstract
OBJECTIVES High intake of dietary fructose causes perturbation in lipid metabolism and provokes lipid-induced insulin resistance. A rise in glucocorticoids (GCs) has recently been suggested to be involved in fructose-induced insulin resistance. The objective of the study was to investigate the effect of GC blockade on lipid abnormalities in insulin-resistant mice. METHODS Insulin resistance was induced in mice by administering a high-fructose diet (HFrD) for 60 days. Mifepristone (RU486), a GC antagonist, was administered to HFrD-fed mice for the last 18 days, and the intracellular and extracellular GC levels, the glucocorticoid receptor (GR) activation and the expression of GC-regulated genes involved in lipid metabolism were examined. RESULTS HFrD elevated the intracellular GC content in both liver and adipose tissue and enhanced the GR nuclear translocation. The plasma GC level remained unchanged. The levels of free fatty acids and triglycerides in plasma were elevated, accompanied by increased plasma insulin and glucose levels and decreased hepatic glycogen content. Treatment with RU486 reduced plasma lipid levels, tissue GC levels and the expression of GC-targeted genes involved in lipid accumulation, and it improved insulin sensitivity. CONCLUSIONS This study demonstrated that HFrD-induced lipid accumulation and insulin resistance are mediated by enhanced GC in liver and adipose tissue and that GC antagonism might reduce fructose-induced lipid abnormalities and insulin resistance.
Collapse
|
13
|
Ribonuclease H1-dependent hepatotoxicity caused by locked nucleic acid-modified gapmer antisense oligonucleotides. Sci Rep 2016; 6:30377. [PMID: 27461380 PMCID: PMC4961955 DOI: 10.1038/srep30377] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 06/30/2016] [Indexed: 12/16/2022] Open
Abstract
Gapmer antisense oligonucleotides cleave target RNA effectively in vivo, and is considered as promising therapeutics. Especially, gapmers modified with locked nucleic acid (LNA) shows potent knockdown activity; however, they also cause hepatotoxic side effects. For developing safe and effective gapmer drugs, a deeper understanding of the mechanisms of hepatotoxicity is required. Here, we investigated the cause of hepatotoxicity derived from LNA-modified gapmers. Chemical modification of gapmer’s gap region completely suppressed both knockdown activity and hepatotoxicity, indicating that the root cause of hepatotoxicity is related to intracellular gapmer activity. Gene silencing of hepatic ribonuclease H1 (RNaseH1), which catalyses gapmer-mediated RNA knockdown, strongly supressed hepatotoxic effects. Small interfering RNA (siRNA)-mediated knockdown of a target mRNA did not result in any hepatotoxic effects, while the gapmer targeting the same position on mRNA as does the siRNA showed acute toxicity. Microarray analysis revealed that several pre-mRNAs containing a sequence similar to the gapmer target were also knocked down. These results suggest that hepatotoxicity of LNA gapmer is caused by RNAseH1 activity, presumably because of off-target cleavage of RNAs inside nuclei.
Collapse
|
14
|
Woods CP, Hazlehurst JM, Tomlinson JW. Glucocorticoids and non-alcoholic fatty liver disease. J Steroid Biochem Mol Biol 2015; 154:94-103. [PMID: 26241028 DOI: 10.1016/j.jsbmb.2015.07.020] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 07/28/2015] [Accepted: 07/29/2015] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the hepatic manifestation of the global obesity and metabolic disease epidemic and is rapidly becoming the leading cause of liver cirrhosis and indication for liver transplantation worldwide. The hallmark pathological finding in NAFLD is excess lipid accumulation within hepatocytes, but it is a spectrum of disease ranging from benign hepatic steatosis to steatohepatitis through to fibrosis, cirrhosis and risk of hepatocellular carcinoma. The exact pathophysiology remains unclear with a multi-hit hypothesis generally accepted as being required for inflammation and fibrosis to develop after initial steatosis. Glucocorticoids have been implicated in the pathogenesis of NAFLD across all stages. They have a diverse array of metabolic functions that have the potential to drive NAFLD acting on both liver and adipose tissue. In the fasting state, they are able to mobilize lipid, increasing fatty acid delivery and in the fed state can promote lipid accumulation. Their action is controlled at multiple levels and in this review will outline the evidence base for the role of GCs in the pathogenesis of NAFLD from cell systems, rodent models and clinical studies and describe interventional strategies that have been employed to modulate glucocorticoid action as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Conor P Woods
- Oxford Centre for Diabetes Endocrinology & Metabolism (OCDEM), Churchill Hospital, Headington, Oxford, OX3 7LJ, UK
| | - Jonathon M Hazlehurst
- Oxford Centre for Diabetes Endocrinology & Metabolism (OCDEM), Churchill Hospital, Headington, Oxford, OX3 7LJ, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes Endocrinology & Metabolism (OCDEM), Churchill Hospital, Headington, Oxford, OX3 7LJ, UK.
| |
Collapse
|
15
|
Gragnoli C. Hypothesis of the neuroendocrine cortisol pathway gene role in the comorbidity of depression, type 2 diabetes, and metabolic syndrome. APPLICATION OF CLINICAL GENETICS 2014; 7:43-53. [PMID: 24817815 PMCID: PMC4012344 DOI: 10.2147/tacg.s39993] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Depression, type 2 diabetes (T2D), and metabolic syndrome (MetS) are often comorbid. Depression per se increases the risk for T2D by 60%. This risk is not accounted for by the use of antidepressant therapy. Stress causes hyperactivation of the hypothalamic–pituitary–adrenal (HPA) axis, by triggering the hypothalamic corticotropin-releasing hormone (CRH) secretion, which stimulates the anterior pituitary to release the adrenocorticotropin hormone (ACTH), which causes the adrenal secretion of cortisol. Depression is associated with an increased level of cortisol, and CRH and ACTH at inappropriately “normal” levels, that is too high compared to their expected lower levels due to cortisol negative feedback. T2D and MetS are also associated with hypercortisolism. High levels of cortisol can impair mood as well as cause hyperglycemia and insulin resistance and other traits typical of T2D and MetS. We hypothesize that HPA axis hyperactivation may be due to variants in the genes of the CRH receptors (CRHR1, CRHR2), corticotropin receptors (or melanocortin receptors, MC1R-MC5R), glucocorticoid receptor (NR3C1), mineralocorticoid receptor (NR3C2), and of the FK506 binding protein 51 (FKBP5), and that these variants may be partially responsible for the clinical association of depression, T2D and MetS. In this review, we will focus on the correlation of stress, HPA axis hyperactivation, and the possible genetic role of the CRHR1, CRHR2, MCR1–5, NR3C1, and NR3C2 receptors and FKBP5 in the susceptibility to the comorbidity of depression, T2D, and MetS. New studies are needed to confirm the hypothesized role of these genes in the clinical association of depression, T2D, and MetS.
Collapse
Affiliation(s)
- Claudia Gragnoli
- Center for Biotechnology and Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, USA ; Molecular Biology Laboratory, Bios Biotech Multi-Diagnostic Health Center, Rome, Italy
| |
Collapse
|
16
|
Rose AJ, Herzig S. Metabolic control through glucocorticoid hormones: an update. Mol Cell Endocrinol 2013; 380:65-78. [PMID: 23523966 DOI: 10.1016/j.mce.2013.03.007] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 02/21/2013] [Accepted: 03/08/2013] [Indexed: 01/28/2023]
Abstract
In the past decades, glucocorticoid (GC) hormones and their cognate, intracellular receptor, the glucocorticoid receptor (GR), have been well established as critical checkpoints in mammalian energy homeostasis. Whereas many aspects in healthy nutrient metabolism require physiological levels and/or action of GC, aberrant GC/GR signalling has been linked to severe metabolic dysfunction, including obesity, insulin resistance and type 2 diabetes. Consequently, studies of the molecular mechanisms within the GC signalling axis have become a major focus in biomedical research, up-to-date particularly focusing on systemic glucose and lipid handling. However, with the availability of novel high throughput technologies and more sophisticated metabolic phenotyping capabilities, as-yet non-appreciated, metabolic functions of GC have been recently discovered, including regulatory roles of the GC/GR axis in protein and bile acid homeostasis as well as metabolic inter-organ communication. Therefore, this review summarises recent advances in GC/GR biology, and summarises findings relevant for basic and translational metabolic research.
Collapse
Affiliation(s)
- Adam J Rose
- Joint Research Division, Molecular Metabolic Control, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH), Heidelberg University, Network Aging Research, University Hospital Heidelberg, Germany
| | | |
Collapse
|
17
|
Mueller KM, Themanns M, Friedbichler K, Kornfeld JW, Esterbauer H, Tuckermann JP, Moriggl R. Hepatic growth hormone and glucocorticoid receptor signaling in body growth, steatosis and metabolic liver cancer development. Mol Cell Endocrinol 2012; 361:1-11. [PMID: 22564914 PMCID: PMC3419266 DOI: 10.1016/j.mce.2012.03.026] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 03/30/2012] [Indexed: 01/07/2023]
Abstract
Growth hormone (GH) and glucocorticoids (GCs) are involved in the control of processes that are essential for the maintenance of vital body functions including energy supply and growth control. GH and GCs have been well characterized to regulate systemic energy homeostasis, particular during certain conditions of physical stress. However, dysfunctional signaling in both pathways is linked to various metabolic disorders associated with aberrant carbohydrate and lipid metabolism. In liver, GH-dependent activation of the transcription factor signal transducer and activator of transcription (STAT) 5 controls a variety of physiologic functions within hepatocytes. Similarly, GCs, through activation of the glucocorticoid receptor (GR), influence many important liver functions such as gluconeogenesis. Studies in hepatic Stat5 or GR knockout mice have revealed that they similarly control liver function on their target gene level and indeed, the GR functions often as a cofactor of STAT5 for GH-induced genes. Gene sets, which require physical STAT5-GR interaction, include those controlling body growth and maturation. More recently, it has become evident that impairment of GH-STAT5 signaling in different experimental models correlates with metabolic liver disease, ranging from hepatic steatosis to hepatocellular carcinoma (HCC). While GH-activated STAT5 has a protective role in chronic liver disease, experimental disruption of GC-GR signaling rather seems to ameliorate metabolic disorders under metabolic challenge. In this review, we focus on the current knowledge about hepatic GH-STAT5 and GC-GR signaling in body growth, metabolism, and protection from fatty liver disease and HCC development.
Collapse
Affiliation(s)
| | | | | | - Jan-Wilhelm Kornfeld
- Institute for Genetics, Department of Mouse Genetics and Metabolism, University of Cologne, Cologne, Germany
| | - Harald Esterbauer
- Department of Laboratory Medicine, Medical University Vienna, Vienna, Austria
| | - Jan P. Tuckermann
- Tissue-Specific Hormone Action, Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany
- Institute for General Zoology and Endocrinology, University of Ulm, Ulm, Germany
| | - Richard Moriggl
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
- Corresponding author. Address: Ludwig Boltzmann Institute for Cancer Research, Waehringerstrasse 13a, 1090 Vienna, Austria. Tel.: +43 14277 64111; fax: +43 14277 9641.
| |
Collapse
|
18
|
Stanton R, Sciabola S, Salatto C, Weng Y, Moshinsky D, Little J, Walters E, Kreeger J, DiMattia D, Chen T, Clark T, Liu M, Qian J, Roy M, Dullea R. Chemical modification study of antisense gapmers. Nucleic Acid Ther 2012; 22:344-59. [PMID: 22852836 DOI: 10.1089/nat.2012.0366] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
A series of insertion patterns for chemically modified nucleotides [2'-O-methyl (2'-OMe), 2'-fluoro (2'-F), methoxyethyl (MOE), locked nucleic acid (LNA), and G-Clamp] within antisense gapmers is studied in vitro and in vivo in the context of the glucocorticoid receptor. Correlation between lipid transfection and unassisted (gymnotic--using no transfection agent) in vitro assays is seen to be dependent on the chemical modification, with the in vivo results corresponding to the unassisted assay in vitro. While in vitro mRNA knockdown assays are typically reasonable predictors of in vivo results, G-Clamp modified antisense oligonucleotides have poor in vivo mRNA knockdown as compared to transfected cell based assays. For LNA gapmers, knockdown is seen to be highly sensitive to the length of the antisense and number of LNA insertions, with longer 5LNA-10DNA-5LNA compounds giving less activity than 3LNA-10DNA-3LNA derivatives. Additionally, the degree of hepatoxicity for antisense gapmers with identical sequences was seen to vary widely with only subtle changes in the chemical modification pattern. While the optimization of knockdown and hepatic effects remains a sequence specific exercise, general trends emerge around preferred physical properties and modification patterns.
Collapse
Affiliation(s)
- Robert Stanton
- Oligonucleotide Therapeutic Unit, Pfizer, Cambridge, MA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Yang R, Lacson RG, Castriota G, Zhang XD, Liu Y, Zhao W, Einstein M, Camargo LM, Qureshi S, Wong KK, Zhang BB, Ferrer M, Berger JP. A genome-wide siRNA screen to identify modulators of insulin sensitivity and gluconeogenesis. PLoS One 2012; 7:e36384. [PMID: 22590537 PMCID: PMC3348929 DOI: 10.1371/journal.pone.0036384] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 03/30/2012] [Indexed: 01/03/2023] Open
Abstract
Background Hepatic insulin resistance impairs insulin’s ability to suppress hepatic glucose production (HGP) and contributes to the development of type 2 diabetes (T2D). Although the interests to discover novel genes that modulate insulin sensitivity and HGP are high, it remains challenging to have a human cell based system to identify novel genes. Methodology/Principal Findings To identify genes that modulate hepatic insulin signaling and HGP, we generated a human cell line stably expressing beta-lactamase under the control of the human glucose-6-phosphatase (G6PC) promoter (AH-G6PC cells). Both beta-lactamase activity and endogenous G6PC mRNA were increased in AH-G6PC cells by a combination of dexamethasone and pCPT-cAMP, and reduced by insulin. A 4-gene High-Throughput-Genomics assay was developed to concomitantly measure G6PC and pyruvate-dehydrogenase-kinase-4 (PDK4) mRNA levels. Using this assay, we screened an siRNA library containing pooled siRNA targeting 6650 druggable genes and identified 614 hits that lowered G6PC expression without increasing PDK4 mRNA levels. Pathway analysis indicated that siRNA-mediated knockdown (KD) of genes known to positively or negatively affect insulin signaling increased or decreased G6PC mRNA expression, respectively, thus validating our screening platform. A subset of 270 primary screen hits was selected and 149 hits were confirmed by target gene KD by pooled siRNA and 7 single siRNA for each gene to reduce G6PC expression in 4-gene HTG assay. Subsequently, pooled siRNA KD of 113 genes decreased PEPCK and/or PGC1alpha mRNA expression thereby demonstrating their role in regulating key gluconeogenic genes in addition to G6PC. Last, KD of 61 of the above 113 genes potentiated insulin-stimulated Akt phosphorylation, suggesting that they suppress gluconeogenic gene by enhancing insulin signaling. Conclusions/Significance These results support the proposition that the proteins encoded by the genes identified in our cell-based druggable genome siRNA screen hold the potential to serve as novel pharmacological targets for the treatment of T2D.
Collapse
Affiliation(s)
- Ruojing Yang
- Department of Metebolic Disorders-Diabetes, Merck Research Laboratories, Rahway, New Jersey, United States of America
- * E-mail: (RY); (JPB)
| | - Raul G. Lacson
- Cell Based HTS, Merck Research Laboratories, North Wales, Pennsylvania, United States of America
| | - Gino Castriota
- Department of Metebolic Disorders-Diabetes, Merck Research Laboratories, Rahway, New Jersey, United States of America
| | - Xiaohua D. Zhang
- Biometrics Research, Merck Research Laboratories, West Point, Pennsylvania, United States of America
| | - Yaping Liu
- Cell Based HTS, Merck Research Laboratories, North Wales, Pennsylvania, United States of America
| | - Wenqing Zhao
- Department of Guided Solutions, Merck Research Laboratories, Rahway, New Jersey, United States of America
| | - Monica Einstein
- Department of Metebolic Disorders-Diabetes, Merck Research Laboratories, Rahway, New Jersey, United States of America
| | - Luiz Miguel Camargo
- Department of Metebolic Disorders-Diabetes, Merck Research Laboratories, Rahway, New Jersey, United States of America
| | - Sajjad Qureshi
- Department of Metebolic Disorders-Diabetes, Merck Research Laboratories, Rahway, New Jersey, United States of America
| | - Kenny K. Wong
- Department of Atherosclerosis, Merck Research Laboratories, Rahway, New Jersey, United States of America
| | - Bei B. Zhang
- Department of Metebolic Disorders-Diabetes, Merck Research Laboratories, Rahway, New Jersey, United States of America
| | - Marc Ferrer
- Cell Based HTS, Merck Research Laboratories, North Wales, Pennsylvania, United States of America
| | - Joel P. Berger
- Department of Metebolic Disorders-Diabetes, Merck Research Laboratories, Rahway, New Jersey, United States of America
- * E-mail: (RY); (JPB)
| |
Collapse
|
20
|
Joshi AKR, Nagaraju R, Rajini PS. Insights into the mechanisms mediating hyperglycemic and stressogenic outcomes in rats treated with monocrotophos, an organophosphorus insecticide. Toxicology 2012; 294:9-16. [DOI: 10.1016/j.tox.2012.01.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 01/16/2012] [Accepted: 01/17/2012] [Indexed: 10/14/2022]
|
21
|
Rose AJ, Vegiopoulos A, Herzig S. Role of glucocorticoids and the glucocorticoid receptor in metabolism: insights from genetic manipulations. J Steroid Biochem Mol Biol 2010; 122:10-20. [PMID: 20170729 DOI: 10.1016/j.jsbmb.2010.02.010] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 12/14/2009] [Accepted: 02/10/2010] [Indexed: 12/21/2022]
Abstract
Since the discovery of the beneficial effects of adrenocortical extracts for treating adrenal insufficiency more than 80 years ago, glucocorticoids and their cognate, intracellular receptor, the glucocorticoid receptor have been characterized as critical checkpoints in the delicate hormonal control of energy homeostasis in mammals. Whereas physiological levels of glucocorticoids are required for proper metabolic control, aberrant glucocorticoid action has been linked to a variety of pandemic metabolic diseases, such as type II diabetes and obesity. Based on its importance for human health, studies of the molecular mechanisms of within the glucocorticoid signaling axis have become a major focus in biomedical research. In particular, the understanding of tissue-specific functions of the glucocorticoid receptor pathway has been proven to be of substantial value for the development of novel therapies in the treatment of chronic metabolic disorders. Therefore, this review focuses on the consequences of endogenous and experimental modulation of glucocorticoid receptor expression for metabolic homeostasis and dysregulation, particularly emphasizing tissue-specific contributions of the glucocorticoid pathway to the control of energy metabolism.
Collapse
Affiliation(s)
- Adam J Rose
- Molecular Metabolic Control, DKFZ-ZMBH Alliance, German Cancer Research Center, Heidelberg, Germany
| | | | | |
Collapse
|
22
|
Edgerton DS, Basu R, Ramnanan CJ, Farmer TD, Neal D, Scott M, Jacobson P, Rizza RA, Cherrington AD. Effect of 11 beta-hydroxysteroid dehydrogenase-1 inhibition on hepatic glucose metabolism in the conscious dog. Am J Physiol Endocrinol Metab 2010; 298:E1019-26. [PMID: 20159854 PMCID: PMC2867371 DOI: 10.1152/ajpendo.00740.2009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Inactive cortisone is converted to active cortisol within the liver by 11 beta-hydroxysteroid dehydrogenase-1 (11 beta-HSD1), and impaired regulation of this process may be related to increased hepatic glucose production (HGP) in individuals with type 2 diabetes. The primary aim of this study was to investigate the effect of acute 11 beta-HSD1 inhibition on HGP and fat metabolism during insulin deficiency. Sixteen conscious, 42-h-fasted, lean, healthy dogs were studied. Somatostatin was infused to create insulin deficiency, and the animals were treated with a specific 11 beta-HSD1 inhibitor (compound 531) or placebo for 5 h. 11 beta-HSD1 inhibition completely suppressed hepatic cortisol production, and this attenuated the increase in HGP that occurred during insulin deficiency. PEPCK and glucose-6-phosphatase expression were decreased when 11 beta-HSD1 was inhibited, but gluconeogenic flux was unchanged, implying an effect on glycogenolysis. Since inhibition of hepatic cortisol production reduces HGP during insulin deficiency, 11 beta-HSD1 is a potential therapeutic target for the treatment of excess glucose production that occurs in diabetes.
Collapse
Affiliation(s)
- Dale S Edgerton
- Molecular Physiology and Biophysics, Vanderbilt University Medical Center, 710 Robinson Research Bldg., Nashville, TN 37232-0615, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Prakash TP, Siwkowski A, Allerson CR, Migawa MT, Lee S, Gaus HJ, Black C, Seth PP, Swayze EE, Bhat B. Antisense oligonucleotides containing conformationally constrained 2',4'-(N-methoxy)aminomethylene and 2',4'-aminooxymethylene and 2'-O,4'-C-aminomethylene bridged nucleoside analogues show improved potency in animal models. J Med Chem 2010; 53:1636-50. [PMID: 20108935 DOI: 10.1021/jm9013295] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
To identify chemistries and strategies to improve the potency of MOE second generation ASOs, we have evaluated gapmer antisense oligonucleotides containing BNAs having N-O bonds. These modifications include N-MeO-amino BNA, N-Me-aminooxy BNA, 2',4'-BNA(NC)[NMe], and 2',4'-BNA(NC) bridged nucleoside analogues. These modifications provided increased thermal stability and improved in vitro activity compared to the corresponding ASO containing the MOE modification. Additionally, ASOs containing N-MeO-amino BNA, N-Me-aminooxy BNA, and 2',4'-BNA(NC)[NMe] modifications showed improved in vivo activity (>5-fold) compared to MOE ASO. Importantly, toxicity parameters, such as AST, ALT, liver, kidney, and body weights, were found to be normal for N-MeO-amino BNA, N-Me-aminooxy BNA, and 2',4'-BNA(NC)[NMe] ASO treated animals. The data generated in these experiments suggest that N-MeO-amino BNA, N-Me-aminooxy BNA, and 2',4'-BNA(NC)[NMe] are useful modifications for applications in both antisense and other oligonucleotide based drug discovery efforts.
Collapse
Affiliation(s)
- Thazha P Prakash
- Department of Medicinal Chemistry and Antisense Core Research, Isis Pharmaceuticals Inc., 1896 Rutherford Road, Carlsbad, California 92008, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Tomlinson JW, Stewart PM. Modulation of glucocorticoid action and the treatment of type-2 diabetes. Best Pract Res Clin Endocrinol Metab 2007; 21:607-19. [PMID: 18054738 DOI: 10.1016/j.beem.2007.07.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The global epidemic of obesity and type-2 diabetes has heightened the need to understand the mechanisms that contribute to its pathogenesis and also to design and trial novel treatments. Patients with glucocorticoid (GC) excess--'Cushing's syndrome'--are phenotypically similar to patients with simple obesity. As such, much research has focused on the manipulation of local GC action as a therapeutic strategy. The majority of the classical actions of GCs are mediated via activation of the glucocorticoid receptor (GR). 11beta-Hydroxysteroid dehydrogenase type 1 (11beta-HSD1) converts inactive cortisone to cortisol and therefore amplifies local GC action. There is now a wealth of data from rodent and clinical studies implicating this conversion in the pathogenesis of obesity, type-2 diabetes, and the metabolic syndrome. Selective 11beta-HSD1 inhibitors (selective in that they block the activity of 11beta-HSD1 and not 11beta-HSD2 which inactivates cortisone to cortisol in mineralocorticoid target tissues) are currently in development although not yet available for use in clinical studies. Rodent studies utilizing these compounds have shown dramatic improvements in insulin sensitivity as well as improvements in lipid profiles and atherogenesis. A further experimental approach has been to design drugs that antagonize GR activation, and again these compounds appear to improve insulin sensitivity and lower glucose production rates. The key test for both of these research strategies is whether they will translate into clinical studies, and results from these trials are now eagerly awaited.
Collapse
Affiliation(s)
- Jeremy W Tomlinson
- Division of Medical Sciences, Institute of Biomedical Research, University of Birmingham, Queen Elizabeth Hospital, Edgbaston, Birmingham B15 2TT, UK.
| | | |
Collapse
|
25
|
Vegiopoulos A, Herzig S. Glucocorticoids, metabolism and metabolic diseases. Mol Cell Endocrinol 2007; 275:43-61. [PMID: 17624658 DOI: 10.1016/j.mce.2007.05.015] [Citation(s) in RCA: 339] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2007] [Revised: 05/14/2007] [Accepted: 05/17/2007] [Indexed: 12/14/2022]
Abstract
Since the discovery of the beneficial effects of adrenocortical extracts for treating adrenal insufficiency more than 80 years ago, glucocorticoids (GC) and their cognate, intracellular receptor, the glucocorticoid receptor (GR) have been characterized as critical components of the delicate hormonal control system that determines energy homeostasis in mammals. Whereas physiological levels of GCs are required for proper metabolic control, excessive GC action has been tied to a variety of pandemic metabolic diseases, such as type II diabetes and obesity. Highlighted by its importance for human health, the investigation of molecular mechanisms of GC/GR action has become a major focus in biomedical research. In particular, the understanding of tissue-specific functions of the GC-GR pathway has been proven to be of substantial value for the identification of novel therapeutic options in the treatment of severe metabolic disorders. Therefore, this review focuses on the role of the GC-GR axis for metabolic homeostasis and dysregulation, emphasizing tissue-specific functions of GCs in the control of energy metabolism.
Collapse
|
26
|
Zinker B, Mika A, Nguyen P, Wilcox D, Ohman L, von Geldern TW, Opgenorth T, Jacobson P. Liver-selective glucocorticoid receptor antagonism decreases glucose production and increases glucose disposal, ameliorating insulin resistance. Metabolism 2007; 56:380-7. [PMID: 17292727 DOI: 10.1016/j.metabol.2006.10.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Accepted: 10/17/2006] [Indexed: 12/29/2022]
Abstract
It is unclear how hepatic glucocorticoid receptor (GR) function and hypothalamic-pituitary-adrenal axis tone contribute to the diabetic state and in particular whole-body glucose fluxes. We have previously demonstrated that long-term exposure to hepatic GR inhibition lowers glucose levels in ob/ob mice (J Pharmacol Exp Ther 2005;314:191). The purpose of this study was to determine the effects of a novel GR antagonist (A-348441) on whole-body glucose fluxes in a model of insulin resistance, the Zucker fatty (fa/fa) rat. After an overnight fast, euglycemic-hyperinsulinemic clamp studies were performed 2 hours after single oral dosing as follows: (1) A-348441 at 100 mg/kg or (2) vehicle. Furthermore, effects of 1 week of treatment with either vehicle or A-348441 (3, 10, 30, or 100 mg/kg PO, once per day) were investigated in separate groups of rats fasted overnight and given a final dose of their respective compound, followed 2 hours later by a euglycemic-hyperinsulinemic clamp. One week after catheter implantation, body weight returned to presurgery levels, with no difference between groups. A single, 100-mg/kg dose of A-348441 significantly increased glucose infusion rate 4-fold (P < .05) and reduced endogenous glucose production by 37% (P < .05) but did not change glucose disposal. After 1 week of sub-long-term dosing, fasting glucose levels were reduced dose-dependently with A-348441 vs vehicle (-8%, not significant; -14%, -20%, and -25%, P < .05, at 3, 10, 30, and 100 mg/kg, respectively) with no observed hypoglycemia or change in fasting insulin levels. A-348441 increased the glucose infusion rates after 1-week treatment by 1.3-, 5.7-, 7.3-, and 6.4-fold (P < .05). Endogenous glucose production was decreased (-25%, -44%, -50%, and -61%, P < .05), whereas glucose disposal was increased (29% and 13%, not significant; 23% and 34%, P < .05), with A-348441. In summary, single-dose treatment with the liver-selective GR antagonist A-348441 decreases glucose production with no effect on glucose disposal or fasting glucose levels. After 1 week of treatment with A-348441, (1) there was no effect on body weight, (2) fasting glucose levels decreased, (3) both glucose disposal and glucose infusion rate increased during clamping, and (4) endogenous glucose production was greatly reduced. In addition, hepatic glucose production was highly correlated with fasting glucose levels (r = 0.97). In conclusion, these results indicate that A-348441 increases insulin sensitivity at both the liver and peripheral tissues, leading toward a normalization of the insulin resistant state. Furthermore, with 1-week vs single-dose liver-selective glucocorticoid antagonism, we have determined that the peripheral effect is secondary to the primary event of reduced hepatic glucose production. The approach of inhibiting the hepatic GR may be an advantageous treatment paradigm for individuals with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Bradley Zinker
- Metabolic Diseases Research, Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Qi D, Rodrigues B. Glucocorticoids produce whole body insulin resistance with changes in cardiac metabolism. Am J Physiol Endocrinol Metab 2007; 292:E654-67. [PMID: 17077342 DOI: 10.1152/ajpendo.00453.2006] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Insulin resistance is viewed as an insufficiency in insulin action, with glucocorticoids being recognized to play a key role in its pathogenesis. With insulin resistance, metabolism in multiple organ systems such as skeletal muscle, liver, and adipose tissue is altered. These metabolic alterations are widely believed to be important factors in the morbidity and mortality of cardiovascular disease. More importantly, clinical and experimental studies have established that metabolic abnormalities in the heart per se also play a crucial role in the development of heart failure. Following glucocorticoids, glucose utilization is compromised in the heart. This attenuated glucose metabolism is associated with altered fatty acid supply, composition, and utilization. In the heart, elevated fatty acid use has been implicated in a number of metabolic, morphological, and mechanical changes and, more recently, in "lipotoxicity". In the present article, we review the action of glucocorticoids, their role in insulin resistance, and their influence in modulating peripheral and cardiac metabolism and heart disease.
Collapse
Affiliation(s)
- Dake Qi
- Division of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, The University of British Columbia, 2146 East Mall, Vancouver, BC, Canada V6T 1Z3
| | | |
Collapse
|
28
|
Swayze EE, Siwkowski AM, Wancewicz EV, Migawa MT, Wyrzykiewicz TK, Hung G, Monia BP, Bennett CF. Antisense oligonucleotides containing locked nucleic acid improve potency but cause significant hepatotoxicity in animals. Nucleic Acids Res 2006; 35:687-700. [PMID: 17182632 PMCID: PMC1802611 DOI: 10.1093/nar/gkl1071] [Citation(s) in RCA: 321] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
A series of antisense oligonucleotides (ASOs) containing either 2′-O-methoxyethylribose (MOE) or locked nucleic acid (LNA) modifications were designed to investigate whether LNA antisense oligonucleotides (ASOs) have the potential to improve upon MOE based ASO therapeutics. Some, but not all, LNA containing oligonucleotides increased potency for reducing target mRNA in mouse liver up to 5-fold relative to the corresponding MOE containing ASOs. However, they also showed profound hepatotoxicity as measured by serum transaminases, organ weights and body weights. This toxicity was evident for multiple sequences targeting three different biological targets, as well as in mismatch control sequences having no known mRNA targets. Histopathological evaluation of tissues from LNA treated animals confirmed the hepatocellular involvement. Toxicity was observed as early as 4 days after a single administration. In contrast, the corresponding MOE ASOs showed no evidence for toxicity while maintaining the ability to reduce target mRNA. These studies suggest that while LNA ASOs have the potential to improve potency, they impose a significant risk of hepatotoxicity.
Collapse
Affiliation(s)
- Eric E Swayze
- Isis Pharmaceuticals, Inc., 1896 Rutherford Road, Carlsbad, CA 92008, USA.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Basu R, Edgerton DS, Singh RJ, Cherrington A, Rizza RA. Splanchnic cortisol production in dogs occurs primarily in the liver: evidence for substantial hepatic specific 11beta hydroxysteroid dehydrogenase type 1 activity. Diabetes 2006; 55:3013-9. [PMID: 17065337 DOI: 10.2337/db06-0601] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Eight dogs underwent combined hepatic/portal vein catheterization and infusion of D4-cortisol in order to determine the relative contributions of the viscera and liver to splanchnic cortisol production. D4-cortisol concentrations progressively decreased from 2.6 +/- 0.1 to 2.4 +/- 0.1 to 1.7 +/- 0.1 microg/dl (P < 0.001 by ANOVA) from hepatic artery to portal vein to hepatic vein, respectively, indicating 8 +/- 3 and 28 +/- 3% extraction across the viscera and liver, respectively. On the other hand, hepatic artery, portal vein, and hepatic vein cortisol concentrations did not differ (0.31 +/- 0.12 vs. 0.28 +/- 0.11 vs. 0.27 +/- 0.10 microg/dl, respectively), indicating zero net cortisol balance. This meant that 1.0 +/- 0.1 microg/min of cortisol was produced within the splanchnic bed, all of which occurred within the liver (1.2 +/- 0.1 microg/min). On the other hand, visceral cortisol production did not differ from zero (-0.2 +/- 0.2 microg/min; P < 0.001 vs. liver). Flux through the 11beta hydroxysteroid dehydrogenase (HSD) type 1 pathway can be measured by determining the rate of conversion of D4-cortisol to D3-cortisol. D3-cortisol concentrations were lower in the portal vein than hepatic artery (0.45 +/- 0.03 vs. 0.48 +/- 0.02, respectively; P < 0.01) but did not differ in the portal vein and hepatic vein, indicating net uptake across the viscera but zero balance across the liver. D3-cortisol production with the viscera and liver averaged 0.2 +/- 0.1 microg/min (P = NS vs. zero production) and 0.6 +/- 0.1 microg/min (P < 0.001 vs. zero production; P < 0.001 vs. viscera production), respectively. We conclude that most, if not all, of splanchnic cortisol production occurs within the liver. Taken together, these data suggest that the high local cortisol concentrations generated via the 11beta HSD type 1 pathway within the liver likely contribute to the regulation of hepatic glucose, fat, and protein metabolism.
Collapse
Affiliation(s)
- Rita Basu
- Mayo Clinic, 200 1st St. SW, Rm 5-194 Joseph, Rochester, MN 55905, USA
| | | | | | | | | |
Collapse
|
30
|
Edgerton DS, Jacobson PB, Opgenorth TJ, Zinker B, Beno D, von Geldern T, Ohman L, Scott M, Neal D, Cherrington AD. Selective antagonism of the hepatic glucocorticoid receptor reduces hepatic glucose production. Metabolism 2006; 55:1255-62. [PMID: 16919547 DOI: 10.1016/j.metabol.2006.05.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2006] [Accepted: 05/29/2006] [Indexed: 01/07/2023]
Abstract
A liver-selective glucocorticoid (GC) receptor antagonist (A-348441) was used to determine the effect of reduced hepatic GC signaling on hepatic glucose production. Fasted conscious dogs were studied in the presence (GRA, n = 6) or absence (CON, n = 6) of the intraduodenally administered GC receptor antagonist (100 mg/kg). All dogs were maintained on a pancreatic clamp and in a euglycemic state for 7 hours to ensure that any changes in glucose metabolism were the direct result of the effects of A-348441, which was given at the start of a 5-hour experimental period. In the GRA group, the arterial plasma insulin level was 4.6 +/- 0.7 and 4.8 +/- 0.6 microU/mL during the basal and the last 30 minutes of the experimental periods, respectively. In the CON group, it was 4.0 +/- 0.3 and 4.5 +/- 0.5 microU/mL in the 2 periods, respectively. The arterial plasma glucagon level was 49 +/- 4 and 46 +/- 3 pg/mL in the 2 periods in the GRA group, and 45 +/- 3 and 42 +/- 3 pg/mL in the CON group. Net hepatic glucose balance progressively decreased in the GRA group from 1.31 +/- 0.18 to 0.49 +/- 0.30 mg/kg per minute, whereas in the CON group, net hepatic glucose balance was 1.17 +/- 0.09 and 1.43 +/- 0.18 mg/kg per minute during the basal and last 30 minutes of the experimental periods, respectively. No significant change in net renal or gut glucose balance or nonhepatic glucose uptake was observed in either group. This study demonstrates that the GC receptor plays an important role in the regulation of basal hepatic glucose production and represents a significant potential therapeutic target.
Collapse
Affiliation(s)
- Dale S Edgerton
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Hanniman EA, Lambert G, Inoue Y, Gonzalez FJ, Sinal CJ. Apolipoprotein A-IV is regulated by nutritional and metabolic stress: involvement of glucocorticoids, HNF-4 alpha, and PGC-1 alpha. J Lipid Res 2006; 47:2503-14. [PMID: 16929032 DOI: 10.1194/jlr.m600303-jlr200] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Apolipoprotein A-IV (apoA-IV) is a 46 kDa glycoprotein that associates with triglyceride-rich and high density lipoproteins. Blood levels of apoA-IV generally correlate with triglyceride levels and are increased in diabetic patients. This study investigated the mechanisms regulating the in vivo expression of apoA-IV in the liver and intestine of mice in response to changes in nutritional status. Fasting markedly increased liver and ileal apoA-IV mRNA and plasma protein concentrations. This induction was associated with increased serum glucocorticoid levels and was abolished by adrenalectomy. Treatment with dexamethasone increased apoA-IV expression in adrenalectomized mice. Marked increases of apoA-IV expression were also observed in two murine models of diabetes. Reporter gene analysis of the murine and human apoA-IV/C-III promoters revealed a conserved cooperative activation by the hepatic nuclear factor-4 alpha (HNF-4 alpha) and the peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1 alpha) but no evidence of a direct regulatory role for the glucocorticoid receptor. Consistent with these in vitro data, induction of apoA-IV in response to fasting was accompanied by increases in HNF-4 alpha and PGC-1 alpha expression and was abolished in liver-specific HNF-4 alpha-deficient mice. Together, these results indicate that the induction of apoA-IV expression in fasting and diabetes likely involves PGC-1 alpha-mediated coactivation of HNF-4 alpha in addition to glucocorticoid-dependent actions.
Collapse
Affiliation(s)
- Elyhisha A Hanniman
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | | | | | | |
Collapse
|
32
|
Wang S, Cheng L, Yu F, Pan W, Zhang J. Delivery of different length poly(L-lysine)-conjugated ODN to HepG2 cells using N-stearyllactobionamide-modified liposomes and their enhanced cellular biological effects. Int J Pharm 2006; 311:82-8. [PMID: 16427225 DOI: 10.1016/j.ijpharm.2005.12.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2005] [Revised: 11/22/2005] [Accepted: 12/10/2005] [Indexed: 10/25/2022]
Abstract
Short (14-20-mer range) synthetic oligodeoxynucleotides (ODNs) allow specific modulation of cellular gene expression at various stages, thus providing a versatile tool for fundamental studies and a rational approach to anticancer chemotherapy. However, several problems, such as metabolic stability, efficient cell internalization of ODNs and their efficient entrapment into liposomes continue to markedly limit this approach. To improve the target specificity and biological activity of ODN, three different length of poly(L-lysine) (PLL) were conjugated to ODN and these conjugates were encapsulated in N-stearyllactobionamide (N-SLBA)-modified liposomes, N-SLBA is a ligand for the asialoglycoprotein receptor. Then, we investigated their effects on cell cycle and survivin protein levels of HepG2 cells. The results showed that the encapsulation efficiency was improved because the polycationic charges of PLL neutralized the polyanionic charges of ODN. Among them, PLL (M(W) 2000 and 10,000)-conjugated ODN encapsulated in N-SLBA liposomes induced apoptosis of HepG2 cells and highly inhibited survivin gene expression.
Collapse
Affiliation(s)
- Siling Wang
- Department of Pharmaceutics and Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | | | | | | | | |
Collapse
|