1
|
Hisaoka-Nakashima K, Takeuchi Y, Saito Y, Shimoda T, Nakamura Y, Wang D, Liu K, Nishibori M, Morioka N. Glucocorticoids induce HMGB1 release in primary cultured rat cortical microglia. Neuroscience 2024; 560:56-66. [PMID: 39304023 DOI: 10.1016/j.neuroscience.2024.09.031] [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: 06/30/2024] [Revised: 09/01/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024]
Abstract
Stress, a risk factor for major depressive disorder and Alzheimer disease, leads to the release of high-mobility group box-1 (HMGB1) protein, which in turn causes neuroinflammation. The mechanism underlying stress-induced HMGB1 release is unknown, but stress-associated glucocorticoids could be involved. Primary cultured rat cortical microglia and neurons were treated with corticosterone, a stress-associated glucocorticoid, and HMGB1 release was measured by ELISA and western blotting to test this hypothesis. With corticosterone treatment, significant HMGB1 was released in microglia but not in neuronal cell cultures. HMGB1 mRNA expression and HMGB1 protein expression in microglia were not affected by corticosterone treatment. Thus, the source of extracellular HMGB1 released into the medium is likely to be existing nuclear HMGB1 rather than newly synthesized HMGB1. Corticosterone-induced HMGB1 release in microglia culture was significantly attenuated by blocking glucocorticoid receptors but not mineralocorticoid receptors. Dexamethasone, a selective glucocorticoid receptor agonist, and dexamethasone-bovine serum albumin (BSA), a membrane-impermeable glucocorticoid receptor agonist used to confirm the membrane receptor-mediated effects of glucocorticoids, increased the release of HMGB1. Immunocytochemistry showed that HMGB1 translocated from the nucleus to the cytoplasm following dexamethasone or dexamethasone-BSA treatment through glucocorticoid receptors. The present findings suggest that glucocorticoids stimulate microglial membrane glucocorticoid receptors and trigger cytoplasmic translocation and extracellular release of nuclear HMGB1. Thus, under stress conditions, glucocorticoids induce microglial HMGB1 release, leading to a neuroinflammatory state that could mediate neurological disorders.
Collapse
Affiliation(s)
- Kazue Hisaoka-Nakashima
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Yuka Takeuchi
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Yukino Saito
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Takahisa Shimoda
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan; Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Co. Ltd., 632-1 Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| | - Yoki Nakamura
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Dengli Wang
- Department of Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata, Okayama, Japan
| | - Keyue Liu
- Department of Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata, Okayama, Japan
| | - Masahiro Nishibori
- Department of Translational Research & Drug Development, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata, Okayama, Japan
| | - Norimitsu Morioka
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan.
| |
Collapse
|
2
|
Duan T, Cao Z, Huang X, Wang X, Sun T, Xu C. Association of social health with all-cause mortality and cause-specific mortality: A population-based cohort study. J Affect Disord 2024; 359:49-58. [PMID: 38768818 DOI: 10.1016/j.jad.2024.05.081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND Previous studies only focused on the individual social factors, without considering the overall social health patterns. The present study aimed to develop an integrated social health score (SHS) and investigate its associations with all-cause, cardiovascular disease (CVD), cancer mortality. METHODS A total of 330,716 participants (mean age 56.3 years; 52.4 % female) from UK Biobank was included between 2006 and 2010, and thereafter followed up to 2021. SHS was calculated by using information on social connections, social engagement and social support. Cox proportional hazards models was used to estimate the hazard ratios and 95 % confidence intervals (CIs) of the association between SHS and all-cause and cause-specific mortality and the 4-way decomposition was used to quantify the mediating effect of lifestyle factors. RESULTS During a median follow-up period of 12.4 years, 37,897 death cases were recorded, including 4347 CVD and 10,380 cancer cases. The SHS was inversely associated with the risks of all-cause, CVD and cancer mortality in a dose-dependent manner (P for trend <0.001). The association between SHS with all-cause mortality was mediated by lifestyle factors including diet score, smoking status and alcohol consumption. CONCLUSION Integrated SHS was inversely associated with risks of all-cause, CVD and cancer mortality, and the associations were partially mediated by lifestyle factors. Our study highlights the importance of maintaining high levels of social health by jointly enhancing social involvement, expanding social networks, and cultivating enduring intimate relationships across the life course.
Collapse
Affiliation(s)
- Tingshan Duan
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Zhi Cao
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianhong Huang
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Xiaohe Wang
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Tao Sun
- School of Public Health, Hangzhou Normal University, Hangzhou, China.
| | - Chenjie Xu
- School of Public Health, Hangzhou Normal University, Hangzhou, China.
| |
Collapse
|
3
|
Wang F, Gao Y, Han Z, Yu Y, Long Z, Jiang X, Wu Y, Pei B, Cao Y, Ye J, Wang M, Zhao Y. A systematic review and meta-analysis of 90 cohort studies of social isolation, loneliness and mortality. Nat Hum Behav 2023; 7:1307-1319. [PMID: 37337095 DOI: 10.1038/s41562-023-01617-6] [Citation(s) in RCA: 107] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 04/27/2023] [Indexed: 06/21/2023]
Abstract
The associations between social isolation, loneliness and the risk of mortality from all causes, cardiovascular disease (CVD) and cancer are controversial. We systematically reviewed prospective studies on the association between social isolation, loneliness and mortality outcomes in adults aged 18 years or older, as well as studies on these relationships in individuals with CVD or cancer, and conducted a meta-analysis. The study protocol was registered with PROSPERO (reg. no. CRD42022299959). A total of 90 prospective cohort studies including 2,205,199 individuals were included. Here we show that, in the general population, both social isolation and loneliness were significantly associated with an increased risk of all-cause mortality (pooled effect size for social isolation, 1.32; 95% confidence interval (CI), 1.26 to 1.39; P < 0.001; pooled effect size for loneliness, 1.14; 95% CI, 1.08 to 1.20; P < 0.001) and cancer mortality (pooled effect size for social isolation, 1.24; 95% CI, 1.19 to 1.28; P < 0.001; pooled effect size for loneliness, 1.09; 95% CI, 1.01 to 1.17; P = 0.030). Social isolation also increased the risk of CVD mortality (1.34; 95% CI, 1.25 to 1.44; P < 0.001). There was an increased risk of all-cause mortality in socially isolated individuals with CVD (1.28; 95% CI, 1.10 to 1.48; P = 0.001) or breast cancer (1.51; 95% CI, 1.34 to 1.70; P < 0.001), and individuals with breast cancer had a higher cancer-specific mortality owing to social isolation (1.33; 95% CI, 1.02 to 1.75; P = 0.038). Greater focus on social isolation and loneliness may help improve people's well-being and mortality risk.
Collapse
Affiliation(s)
- Fan Wang
- Department of Epidemiology, School of Public Health, Harbin Medical University, Harbin, P. R. China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, P. R. China
| | - Yu Gao
- Department of Epidemiology, School of Public Health, Harbin Medical University, Harbin, P. R. China
| | - Zhen Han
- Department of Epidemiology, School of Public Health, Harbin Medical University, Harbin, P. R. China
| | - Yue Yu
- Department of Epidemiology, School of Public Health, Harbin Medical University, Harbin, P. R. China
| | - Zhiping Long
- Department of Epidemiology, School of Public Health, Harbin Medical University, Harbin, P. R. China
| | - Xianchen Jiang
- Department of Chronic Disease Prevention and Control, Quzhou Center for Disease Control and Prevention, Quzhou, P. R. China
| | - Yi Wu
- Department of Epidemiology, School of Public Health, Harbin Medical University, Harbin, P. R. China
| | - Bing Pei
- Department of Epidemiology, School of Public Health, Harbin Medical University, Harbin, P. R. China
| | - Yukun Cao
- Department of Epidemiology, School of Public Health, Harbin Medical University, Harbin, P. R. China
| | - Jingyu Ye
- Department of Epidemiology, School of Public Health, Harbin Medical University, Harbin, P. R. China
| | - Maoqing Wang
- National Key Disciplines of Nutrition and Food Hygiene, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China.
| | - Yashuang Zhao
- Department of Epidemiology, School of Public Health, Harbin Medical University, Harbin, P. R. China.
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, P. R. China.
| |
Collapse
|
4
|
Razzaghi A, Ghaffari MH, Rico DE. The impact of environmental and nutritional stresses on milk fat synthesis in dairy cows. Domest Anim Endocrinol 2022; 83:106784. [PMID: 36586193 DOI: 10.1016/j.domaniend.2022.106784] [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] [Received: 06/15/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Stress reduces milk and milk components synthesis and increases maintenance requirements of cows. The major stress-related alterations involve enhanced secretion of glucocorticoids and increased sympathetic nervous system activity, which results in biochemical and physiologic changes. In dairy cows exposed to social (ie housing conditions, overstocking, regrouping, feed delivery), physiological (ie initiation of lactation and parturition), or physical (ie heat or cold stress) stressors, responses involve alterations in energy balance and nutrient partitioning. The capacity of the animal to synthesize milk fat largely depends on the availability of substrates for lipid synthesis from the diet, ruminal fermentation or adipose tissue stores, all of which can be altered under stress conditions. Indeed, milk fat concentration is particularly responsive to diet and environment modifications, where a wide range of nutritional and non-nutritional factors influence milk fat output. Milk fat synthesis is an energy demanding process, and extremely sensitive to stress factors during lactation and the involvement of multiple organs. Recent studies examining social, physical, and physiological stressors have provided important insights into how differences in milk yield and milk components may be associated with biological responses to stress factors in dairy cows. This review focuses primarily on the role of stress sources and indicators to which the dairy cow is exposed in regulating milk fat synthesis. We will review the role of nutritional and non-nutritional factors on milk fat synthesis in dairy cows under stress conditions.
Collapse
Affiliation(s)
- A Razzaghi
- Innovation Center, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - M H Ghaffari
- Institute of Animal Science, University of Bonn, Bonn, Germany
| | - D E Rico
- Centre de recherche en sciences animales de Deschambault (CRSAD), Deschambault, QC, Canada, G0A 1S0
| |
Collapse
|
5
|
Zheng Y, Karnoub AE. Endocrine regulation of cancer stem cell compartments in breast tumors. Mol Cell Endocrinol 2021; 535:111374. [PMID: 34242715 DOI: 10.1016/j.mce.2021.111374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 10/20/2022]
Abstract
Cancer cells within breast tumors exist within a hierarchy in which only a small and rare subset of cells is able to regenerate growths with the heterogeneity of the original tumor. These highly malignant cancer cells, which behave like stem cells for new cancers and are called "cancer stem cells" or CSCs, have also been shown to possess increased resistance to therapeutics, and represent the root cause underlying therapy failures, persistence of residual disease, and relapse. As >90% of cancer deaths are due to refractory tumors, identification of critical molecular drivers of the CSC-state would reveal vulnerabilities that can be leveraged in designing therapeutics that eradicate advanced disease and improve patient survival outcomes. An expanding and complex body of work has now described the exquisite susceptibility of CSC pools to the regulatory influences of local and systemic hormones. Indeed, breast CSCs express a plethora of hormonal receptors, which funnel hormonal influences over every aspect of breast neoplasia - be it tumor onset, growth, survival, invasion, metastasis, or therapy resistance - via directly impacting CSC behavior. This article is intended to shed light on this active area of investigation by attempting to provide a systematic and comprehensive overview of the available evidence directly linking hormones to breast CSC biology.
Collapse
Affiliation(s)
- Yurong Zheng
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Antoine E Karnoub
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA; Harvard Stem Cell Institute, Cambridge, MA, 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
6
|
Ye R, Sun L, Peng J, Wu A, Chen X, Wen L, Bai C, Chen G. Design, Synthesis, and Biological Evaluation of Dexamethasone-Salvianolic Acid B Conjugates and Nanodrug Delivery against Cisplatin-Induced Hearing Loss. J Med Chem 2021; 64:3115-3130. [PMID: 33666428 DOI: 10.1021/acs.jmedchem.0c01916] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cisplatin (CDDP) is an extensively used chemotherapeutic agent but has a high incidence of severe ototoxicity. Although a few molecules have entered clinical trials, none have been approved to prevent or treat CDDP-induced hearing loss by the Food and Drug Administration. In this study, an amphiphilic drug-drug conjugate was synthesized by covalently linking dexamethasone (DEX) and salvianolic acid B (SAL) through an ester or amide bond. The conjugates could self-assemble into nanoparticles (NPs) with ultrahigh drug loading capacity and favorable stability. Compared with DEX, SAL, or their physical mixture at the same concentrations, both conjugates and NPs showed enhanced otoprotection in vitro and in vivo. More importantly, the conjugates and NPs almost completely restored hearing in a guinea pig model with good biocompatibility. Immunohistochemical analyses suggested that conjugates and NPs activated the glucocorticoid receptor, which may work as one of the major mechanisms for their protective effects.
Collapse
Affiliation(s)
- Ruiqin Ye
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lifang Sun
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jinghui Peng
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Aixin Wu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiaozhu Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lu Wen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chuan Bai
- Institute of Human Virology, Sun Yat-sen University, Guangzhou 510080, China
| | - Gang Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.,Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou 510006, China.,Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
7
|
Luo QQ, Wang B, Chen X, Qiu HY, Li WT, Yan XJ, Chen SL. Acute stress induces visceral hypersensitivity via glucocorticoid receptor-mediated membrane insertion of TRPM8: Involvement of a non-receptor tyrosine kinase Pyk2. Neurogastroenterol Motil 2020; 32:1514-1528. [PMID: 32391653 DOI: 10.1111/nmo.13877] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 03/05/2020] [Accepted: 04/17/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Psychological stress is an important factor for the development and recurrence of irritable bowel syndrome (IBS). The mechanisms underlying stress-induced visceral hypersensitivity (VH), a key pathophysiological component in IBS, are still incompletely understood. We aimed to test whether transient receptor potential melastatin 8 (TRPM8) participates in acute stress-induced VH. METHODS Rats were subjected to 1-hour water avoidance stress (WAS). Visceral sensitivity was measured with visceromotor response to colorectal distension. Western blot and immunofluorescence were applied to evaluate the expression of GR and TRPM8 and activation of PKA, Akt, and PKC pathways. RESULTS WAS-caused VH depended on glucocorticoid receptors (GRs) and TRPM8 channels. In a dorsal root ganglion (DRG)-derived cell line, corticosterone rapidly (within 30 minutes) induced membrane expression of TRPM8. This effect was inhibited by GR antagonism and was mimicked by membrane-impermeable corticosterone. PKA, PI3K/Akt, and PKC pathways, which lied downstream of GR and acted in parallel to promote membrane expression of TRPM8, contributed to WAS-induced VH. The non-receptor tyrosine kinase Pyk2, which may serve as a convergence point for PKA, PI3K/Akt, and PKC pathways, facilitated membrane insertion of TRPM8 via tyrosine-phosphorylating TRPM8 in L6-S2 DRGs and participated in WAS-induced VH. CONCLUSIONS Collectively, acute stress-induced VH could involve membrane-bound GR-dependent enhancement of TRPM8 function in nociceptive DRG neurons. Mechanistically, Pyk2 could act as a key mediator that coordinates multiple protein kinase signaling and triggers phosphorylation and membrane insertion of TRPM8.
Collapse
Affiliation(s)
- Qing-Qing Luo
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Bo Wang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Xin Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Hong-Yi Qiu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Wen-Ting Li
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Xiu-Juan Yan
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Sheng-Liang Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| |
Collapse
|
8
|
Akalestou E, Genser L, Rutter GA. Glucocorticoid Metabolism in Obesity and Following Weight Loss. Front Endocrinol (Lausanne) 2020; 11:59. [PMID: 32153504 PMCID: PMC7045057 DOI: 10.3389/fendo.2020.00059] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/30/2020] [Indexed: 01/08/2023] Open
Abstract
Glucocorticoids are steroid hormones produced by the adrenal cortex and are essential for the maintenance of various metabolic and homeostatic functions. Their function is regulated at the tissue level by 11β-hydroxysteroid dehydrogenases and they signal through the glucocorticoid receptor, a ligand-dependent transcription factor. Clinical observations have linked excess glucocorticoid levels with profound metabolic disturbances of intermediate metabolism resulting in abdominal obesity, insulin resistance and dyslipidaemia. In this review, we discuss the physiological mechanisms of glucocorticoid secretion, regulation and function, and survey the metabolic consequences of excess glucocorticoid action resulting from elevated release and activation or up-regulated signaling. Finally, we summarize the reported impact of weight loss by diet, exercise, or bariatric surgery on circulating and tissue-specific glucocorticoid levels and examine the therapeutic possibility of reversing glucocorticoid-associated metabolic disorders.
Collapse
Affiliation(s)
- Elina Akalestou
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
| | - Laurent Genser
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
- Department of Digestive and Hepato-Pancreato-Biliary Surgery, Liver Transplantation, Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière University Hospital, Institut Hospitalo-Universitaire ICAN, Sorbonne Université, Paris, France
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
- *Correspondence: Guy A. Rutter
| |
Collapse
|
9
|
Kim JS, Iremonger KJ. Temporally Tuned Corticosteroid Feedback Regulation of the Stress Axis. Trends Endocrinol Metab 2019; 30:783-792. [PMID: 31699237 DOI: 10.1016/j.tem.2019.07.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/23/2019] [Accepted: 07/08/2019] [Indexed: 01/28/2023]
Abstract
Activity of the hypothalamic-pituitary-adrenal (HPA) axis is tuned by corticosteroid feedback. Corticosteroids regulate cellular function via genomic and nongenomic mechanisms, which operate over diverse time scales. This review summarizes recent advances in our understanding of how corticosteroid feedback regulates hypothalamic stress neuron function and output through synaptic plasticity, changes in intrinsic excitability, and modulation of neuropeptide production. The temporal kinetics of corticosteroid actions in the brain versus the pituitary have important implications for how organisms respond to stress. Furthermore, we will discuss, some of the technical limitations and missing links in the field, and the potential implications these may have on our interpretations of corticosteroid negative feedback experiments.
Collapse
Affiliation(s)
- Joon S Kim
- Centre for Neuroendocrinology, Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Karl J Iremonger
- Centre for Neuroendocrinology, Department of Physiology, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
10
|
Aedo JE, Zuloaga R, Bastías-Molina M, Meneses C, Boltaña S, Molina A, Valdés JA. Early transcriptomic responses associated with the membrane-initiated action of cortisol in the skeletal muscle of rainbow trout (Oncorhynchus mykiss). Physiol Genomics 2019; 51:596-606. [DOI: 10.1152/physiolgenomics.00042.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cortisol is a critical neuroendocrine regulator of the stress response in fish. Cortisol practically affects all tissues by interacting with an intracellular receptor and modulating target gene expression. However, cortisol also interacts with components of the plasma membrane in a nongenomic process that activates rapid signaling. Until now, the implication of this novel cortisol signaling for the global transcriptional response has not been explored. In the present work, we evaluated the effects of the membrane-initiated actions of cortisol on the in vivo transcriptome of rainbow trout ( Oncorhynchus mykiss) skeletal muscle. RNA-Seq analyses were performed to examine the transcriptomic changes in rainbow trout stimulated by physiological concentrations of cortisol and cortisol coupled with bovine serum albumin (cortisol-BSA), a membrane-impermeable analog of cortisol. A total of 660 million paired-ends reads were generated. Reads mapped onto the reference genome revealed that 1,737; 897; and 1,012 transcripts were differentially expressed after 1, 3, and 9 h of cortisol-BSA treatment, respectively. Gene Ontology analysis showed that this novel action of cortisol modulates several biological processes, such as mRNA processing, ubiquitin-dependent protein catabolic processes, and transcription regulation. In addition, a KEGG analysis revealed that focal adhesion was the main signaling pathway that was upregulated at all the times tested. Taking these results together, we propose that the membrane-initiated cortisol action contributes significantly in the regulation of stress-mediated gene expression.
Collapse
Affiliation(s)
- Jorge E. Aedo
- Universidad Andrés Bello, Departamento Ciencias Biológicas, Facultad de Ciencias de la Vida, Santiago, Chile
- Interdisciplinary Center for Aquaculture Research (INCAR), Concepción, Chile
| | - Rodrigo Zuloaga
- Universidad Andrés Bello, Departamento Ciencias Biológicas, Facultad de Ciencias de la Vida, Santiago, Chile
| | - Macarena Bastías-Molina
- Universidad Andrés Bello, Centro de Biotecnología Vegetal, FONDAP Center for Genome Regulation, Facultad de Ciencias de la Vida, Santiago, Chile
| | - Claudio Meneses
- Universidad Andrés Bello, Centro de Biotecnología Vegetal, FONDAP Center for Genome Regulation, Facultad de Ciencias de la Vida, Santiago, Chile
| | - Sebastián Boltaña
- Interdisciplinary Center for Aquaculture Research (INCAR), Concepción, Chile
- Universidad de Concepción, ThermoFish Lab, Biotechnology Center, Concepción, Chile
| | - Alfredo Molina
- Universidad Andrés Bello, Departamento Ciencias Biológicas, Facultad de Ciencias de la Vida, Santiago, Chile
- Universidad de Concepción, ThermoFish Lab, Biotechnology Center, Concepción, Chile
| | - Juan Antonio Valdés
- Universidad Andrés Bello, Departamento Ciencias Biológicas, Facultad de Ciencias de la Vida, Santiago, Chile
- Universidad de Concepción, ThermoFish Lab, Biotechnology Center, Concepción, Chile
| |
Collapse
|
11
|
Jaćević V, Wu Q, Nepovimova E, Kuča K. Efficacy of methylprednisolone on T-2 toxin-induced cardiotoxicity in vivo: A pathohistological study. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2019; 71:103221. [PMID: 31365892 DOI: 10.1016/j.etap.2019.103221] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 07/11/2019] [Accepted: 07/15/2019] [Indexed: 06/10/2023]
Abstract
Our aim was to compare the protective efficacy of two different formulations of methylprednisolone in T-2 toxin-induced cardiomyopathy. Methylprednisolone (soluble form, Lemod-solu® and/or depot form, Lemod-depo®, a total single dose of 40 mg/kg im) was given immediately after T-2 toxin (1 LD50 0.23 mg/kg sc). The myocardial tissue samples were examinated by using histopathology, semiquantitative and imaging analyses on day 1, 7, 14, 21, 28 and 60 of the study. Therapeutic application of Lemod-solu® significantly decreased the intensity of myocardial degeneration and haemorrhages, distribution of glycogen granules in the endo- and perimysium, a total number of mast cells and the degree of their degranulation was in correlation with the reversible heart structural lesions (p < 0.01 vs. T-2 toxin). These changes were completely abolished by the therapeutic use of Lemod-solu® plus Lemod-depo® (p < 0.001 vs. T-2 toxin). Our results show that a significant cardioprotective efficacy of methylprednisolone is mediated by its anti-inflammatory activity.
Collapse
Affiliation(s)
- Vesna Jaćević
- National Poison Control Centre, Military Medical Academy, 17 Crnotravska St, 11000, Belgrade, Serbia; Medical Faculty of the Military Medical Academy, University of Defence, 1 Pavla Jurišića-Šturma St, 11000, Belgrade, Serbia; Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanského 62, 500 03, Hradec Králové, Czechia
| | - Qinghua Wu
- College of Life Science, Yangtze University, 1 Nanhuan Road, 434023, Jingzhou, Hubei, China; Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanského 62, 500 03, Hradec Králové, Czechia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanského 62, 500 03, Hradec Králové, Czechia
| | - Kamil Kuča
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanského 62, 500 03, Hradec Králové, Czechia; Malaysia-Japan International Institute of Technology (MJIIT), University Teknologi Malaysia, Jalan Sultan Yahya Petra, 54100, Kuala Lumpur, Malaysia.
| |
Collapse
|
12
|
MacDougall-Shackleton SA, Bonier F, Romero LM, Moore IT. Glucocorticoids and "Stress" Are Not Synonymous. Integr Org Biol 2019; 1:obz017. [PMID: 33791532 PMCID: PMC7671118 DOI: 10.1093/iob/obz017] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Reference to glucocorticoids as “stress hormones” has been growing in prevalence in the literature, including in comparative and environmental endocrinology. Although glucocorticoids are elevated in response to a variety of stressors in vertebrate animals, the primary functions of glucocorticoids are not responding to stressors and they are only one component of complex suite of physiological and behavioral responses to stressors. Thus, the use of the short-hand phrase “stress hormone” can be misleading. Further, simply measuring glucocorticoids is not equivalent to measuring a stress response, nor is manipulating glucocorticoids equivalent to exposing an animal to a stressor. In this commentary we highlight the problems with using functional names for hormones, and of treating cortisol or corticosterone as synonymous with stress. We provide recommendations to add clarity to the presentation of research on this topic, and to avoid conflation of glucocorticoids with stressors and the stress response in the design of experiments.
Collapse
Affiliation(s)
| | - F Bonier
- Biology Department, Queen's University, Kingston, Ontario, Canada K7L 3N6
| | - L M Romero
- Department of Biology, Tufts University, Medford, MA 02155, USA
| | - I T Moore
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
13
|
Aedo JE, Zuloaga R, Boltaña S, Molina A, Valdés JA. Membrane-initiated cortisol action modulates early pyruvate dehydrogenase kinase 2 (pdk2) expression in fish skeletal muscle. Comp Biochem Physiol A Mol Integr Physiol 2019; 233:24-29. [DOI: 10.1016/j.cbpa.2019.03.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/27/2019] [Accepted: 03/25/2019] [Indexed: 10/27/2022]
|
14
|
von Krogh K, Bjørndal GT, Nourizadeh-Lillabadi R, Ropstad E, Haug TM, Weltzien FA. Cortisol differentially affects cell viability and reproduction-related gene expression in Atlantic cod pituitary cultures dependent on stage of sexual maturation. Comp Biochem Physiol A Mol Integr Physiol 2019; 236:110517. [PMID: 31254635 DOI: 10.1016/j.cbpa.2019.06.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/16/2019] [Accepted: 06/24/2019] [Indexed: 01/03/2023]
Abstract
Through the action of cortisol, stress can affect reproductive biology with behavioural and physiological alterations. Using mixed sex primary pituitary cultures from Atlantic cod (Gadus morhua), the present study aimed to investigate potential direct effects of basal and stress level cortisol on the pituitary in terms of cell viability and reproduction-related gene expression at different stages of sexual maturity. Stress level of cortisol stimulated cell viability in cells derived from sexually maturing and mature fish. In cells from spent fish, high cortisol levels did not affect cell viability in terms of metabolic activity, but did stimulate viability in terms of membrane integrity. Basal cortisol levels did not affect cell viability. Ethanol, used as solvent for cortisol, decreased cell viability at all maturity stages, but did generally not affect gene expression. Genes investigated were fshb, lhb and two Gnrh receptors expressed in cod gonadotropes (gnrhr1b and gnrhr2a). Cortisol had dual effects on fshb expression; stimulating expression in cells from mature fish at stress dose, while inhibiting expression in cells from spent fish at both doses. In contrast, cortisol had no direct effect on lhb expression. While gnrhr2a transcript levels largely increased following cortisol treatment, gnrhr1b expression decreased in cells from spent fish and was unaffected at other maturity stages. These findings demonstrate that cortisol can act directly and differentially at the pituitary level in Atlantic cod and that factors facilitating these actions are dose-dependently activated and vary with level of sexual maturity.
Collapse
Affiliation(s)
- Kristine von Krogh
- Norwegian University of Life Sciences, Faculty of Veterinary Medicine, Department of Basic Science and Aquatic Medicine, Oslo, Norway
| | - Gunnveig Toft Bjørndal
- Norwegian University of Life Sciences, Faculty of Veterinary Medicine, Department of Basic Science and Aquatic Medicine, Oslo, Norway
| | - Rasoul Nourizadeh-Lillabadi
- Norwegian University of Life Sciences, Faculty of Veterinary Medicine, Department of Basic Science and Aquatic Medicine, Oslo, Norway
| | - Erik Ropstad
- Norwegian University of Life Sciences, Faculty of Veterinary Medicine, Department of Production Animal Clinical Sciences, Oslo, Norway
| | - Trude M Haug
- University of Oslo, Faculty of Dentistry, Department of Oral Biology, Oslo, Norway
| | - Finn-Arne Weltzien
- Norwegian University of Life Sciences, Faculty of Veterinary Medicine, Department of Basic Science and Aquatic Medicine, Oslo, Norway.
| |
Collapse
|
15
|
Johnstone WM, Honeycutt JL, Deck CA, Borski RJ. Nongenomic glucocorticoid effects and their mechanisms of action in vertebrates. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 346:51-96. [PMID: 31122395 DOI: 10.1016/bs.ircmb.2019.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Glucocorticoids (GC) act on multiple organ systems to regulate a variety of physiological processes in vertebrates. Due to their immunosuppressive and anti-inflammatory actions, glucocorticoids are an attractive target for pharmaceutical development. Accordingly, they are one of the most widely prescribed classes of therapeutics. Through the classical mechanism of steroid action, glucocorticoids are thought to mainly affect gene transcription, both in a stimulatory and suppressive fashion, regulating de novo protein synthesis that subsequently leads to the physiological response. However, over the past three decades multiple lines of evidence demonstrate that glucocorticoids may work through rapid, nonclassical mechanisms that do not require alterations in gene transcription or translation. This review assimilates evidence across the vertebrate taxa on the diversity of nongenomic actions of glucocorticoids and the membrane-associated cellular mechanisms that may underlie rapid glucocorticoid responses to include potential binding sites characterized to date.
Collapse
Affiliation(s)
- William M Johnstone
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - Jamie L Honeycutt
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - Courtney A Deck
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - Russell J Borski
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States.
| |
Collapse
|
16
|
Bastos LFS, Vago JP, Caux TR, Costa BL, Godin AM, Menezes RR, Pena RR, Silva-Cunha A, Sousa LP, Machado RR, Moraes MFD, Fialho SL, Coelho MM. Delay of neuropathic pain sensitization after application of dexamethasone-loaded implant in sciatic nerve-injured rats. BRAZ J PHARM SCI 2019. [DOI: 10.1590/s2175-97902019000218112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
17
|
Aedo JE, Ruiz-Jarabo I, Martínez-Rodríguez G, Boltaña S, Molina A, Valdés JA, Mancera JM. Contribution of Non-canonical Cortisol Actions in the Early Modulation of Glucose Metabolism of Gilthead Sea Bream ( Sparus aurata). Front Endocrinol (Lausanne) 2019; 10:779. [PMID: 31798534 PMCID: PMC6863068 DOI: 10.3389/fendo.2019.00779] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/24/2019] [Indexed: 11/24/2022] Open
Abstract
Teleost fish are exposed to diverse stressors in farming and wildlife conditions during their lifespan. Cortisol is the main glucocorticoid hormone involved in the regulation of their metabolic acclimation under physiological stressful conditions. In this context, increased plasma cortisol is associated with energy substrate mobilization from metabolic tissues, such as liver and skeletal muscle, to rapidly obtain energy and cope with stress. The metabolic actions of cortisol have primarily been attributed to its genomic/classic action mechanism involving the interaction with intracellular receptors, and regulation of stress-responsive genes. However, cortisol can also interact with membrane components to activate rapid signaling pathways. In this work, using the teleost fish gilthead sea bream (Sparus aurata) as a model, we evaluated the effects of membrane-initiated cortisol actions on the early modulation of glucose metabolism. For this purpose, S. aurata juveniles were intraperitoneally administrated with cortisol and with its membrane impermeable analog, cortisol-BSA. After 1 and 6 h of each treatment, plasma cortisol levels were measured, together with glucose, glycogen and lactate in plasma, liver and skeletal muscle. Transcript levels of corticosteroids receptors (gr1, gr2, and mr) and key gluconeogenesis (g6pc and pepck)- and glycolysis (pgam1 and aldo) related genes in the liver were also measured. Cortisol and cortisol-BSA administration increased plasma cortisol levels in S. aurata 1 h after administration. Plasma glucose levels enhanced 6 h after each treatment. Hepatic glycogen content decreased in the liver at 1 h of both cortisol and cortisol-BSA administration, while increased at 6 h due to cortisol but not in response to cortisol-BSA. Expression of gr1, g6pc, pgam1, and aldo were preferentially increased by cortisol-BSA in the liver. Taking all these results in consideration, we suggest that non-canonical cortisol mechanisms contribute to the regulation of the early glucose metabolism responses to stress in S. aurata.
Collapse
Affiliation(s)
- Jorge E. Aedo
- Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, Concepción, Chile
| | - Ignacio Ruiz-Jarabo
- Department of Biology, Faculty of Marine and Environmental Sciences, Instituto Universitario de Investigación Marina (INMAR), Campus de Excelencia Internacional del Mar (CEI-MAR), University of Cádiz, Cádiz, Spain
| | - Gonzalo Martínez-Rodríguez
- Department of Marine Biology and Aquaculture, Instituto de Ciencias Marinas de Andalucía (ICMAN-CSIC), Puerto Real, Spain
| | - Sebastián Boltaña
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, Concepción, Chile
| | - Alfredo Molina
- Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, Concepción, Chile
| | - Juan A. Valdés
- Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, Concepción, Chile
- *Correspondence: Juan A. Valdés
| | - Juan M. Mancera
- Department of Biology, Faculty of Marine and Environmental Sciences, Instituto Universitario de Investigación Marina (INMAR), Campus de Excelencia Internacional del Mar (CEI-MAR), University of Cádiz, Cádiz, Spain
| |
Collapse
|
18
|
Czuchlej SC, Volonteri MC, Regueira E, Ceballos NR. Effect of glucocorticoids on androgen biosynthesis in the testes of the toad Rhinella arenarum (Amphibia, Anura). JOURNAL OF EXPERIMENTAL ZOOLOGY PART 2018; 331:17-26. [PMID: 30218550 DOI: 10.1002/jez.2232] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/23/2018] [Accepted: 08/27/2018] [Indexed: 11/11/2022]
Abstract
In rat Leydig cells, glucocorticoids (GCs) inhibit testosterone production through the interaction with the glucocorticoid receptor (GR). However, the sensitivity of those cells to GCs is regulated by the enzyme 11β-hydroxysteroid dehydrogenase Type 1 (11β-HSD1). In the testes of the toad Rhinella arenarum, the presence of an 11β-HSD similar to type 2 and a cytosolic GR has also been described. However, there is a lack of information regarding the effects of GCs on amphibian testicular steroidogenesis. In this study, the effects of corticosterone on androgen production, and the activity of two steroidogenic enzymes in toad testes were reported. Corticosterone inhibits androgen production via the GR because the GR antagonist RU486 prevents corticosterone-induced inhibition of testosterone. Corticosterone also reduced the activity of the cytochrome P450 17-hydroxylase, C17,20-lyase (Cyp450 c17 ) without affecting the 3β-hydroxysteroid dehydrogenase/isomerase activity. This effect on Cyp450 c17 was likewise inhibited by RU486. On the other hand, corticosterone had no effect on the amount of steroidogenic acute regulator protein. These results suggest that GCs inhibit steroidogenesis in toad testes by reducing of Cyp450 c17 activity via a GR-mediated mechanism.
Collapse
Affiliation(s)
- Silvia Cristina Czuchlej
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Clara Volonteri
- Instituto de Diversidad y Evolución Austral (IDEAus CENPAT-CONICET), Puerto Madryn, Chubut, Argentina
| | - Eleonora Regueira
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA UBA- CONICET), Buenos Aires, Argentina
| | - Nora Raquel Ceballos
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
19
|
Das C, Thraya M, Vijayan MM. Nongenomic cortisol signaling in fish. Gen Comp Endocrinol 2018; 265:121-127. [PMID: 29673844 DOI: 10.1016/j.ygcen.2018.04.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/12/2018] [Accepted: 04/14/2018] [Indexed: 10/17/2022]
Abstract
Glucocorticoids are critical regulators of the cellular processes that allow animals to cope with stressors. In teleosts, cortisol is the primary circulating glucocorticoid and this hormone mediates a suite of physiological responses, most importantly energy substrate mobilization that is essential for acute stress adaptation. Cortisol signaling has been extensively studied and the majority of work has been on the activation of the glucocorticoid receptor (GR), a ligand-bound transcription factor, and the associated downstream transcriptional and protein responses. Despite the role of this hormone in acute stress adaptation, very few studies have examined the rapid effects of this hormone on cellular function. The nongenomic corticosteroid effects, which are rapid (seconds to minutes) and independent of transcription and translation, involve changes to second-messenger pathways and effector proteins, but the primary receptors involved in this pathway activation remain elusive. In teleosts, a few studies suggested the possibility that GR located on the membrane may be initiating a rapid response based on the abrogation of this effect with RU486, a GR antagonist. However, studies have also proposed other signaling mechanisms, including a putative novel membrane receptor and changes to membrane biophysical properties as initiators of rapid signaling in response to cortisol stimulation. Emerging evidence suggests that cortisol activates multiple signaling pathways in cells to bring about rapid effects, but the underlying physiological implications on acute stress adaptation are far from clear.
Collapse
Affiliation(s)
- Chinmayee Das
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Marwa Thraya
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Mathilakath M Vijayan
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada.
| |
Collapse
|
20
|
Godoy LD, Rossignoli MT, Delfino-Pereira P, Garcia-Cairasco N, de Lima Umeoka EH. A Comprehensive Overview on Stress Neurobiology: Basic Concepts and Clinical Implications. Front Behav Neurosci 2018; 12:127. [PMID: 30034327 PMCID: PMC6043787 DOI: 10.3389/fnbeh.2018.00127] [Citation(s) in RCA: 389] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/06/2018] [Indexed: 12/20/2022] Open
Abstract
Stress is recognized as an important issue in basic and clinical neuroscience research, based upon the founding historical studies by Walter Canon and Hans Selye in the past century, when the concept of stress emerged in a biological and adaptive perspective. A lot of research after that period has expanded the knowledge in the stress field. Since then, it was discovered that the response to stressful stimuli is elaborated and triggered by the, now known, stress system, which integrates a wide diversity of brain structures that, collectively, are able to detect events and interpret them as real or potential threats. However, different types of stressors engage different brain networks, requiring a fine-tuned functional neuroanatomical processing. This integration of information from the stressor itself may result in a rapid activation of the Sympathetic-Adreno-Medullar (SAM) axis and the Hypothalamus-Pituitary-Adrenal (HPA) axis, the two major components involved in the stress response. The complexity of the stress response is not restricted to neuroanatomy or to SAM and HPA axes mediators, but also diverge according to timing and duration of stressor exposure, as well as its short- and/or long-term consequences. The identification of neuronal circuits of stress, as well as their interaction with mediator molecules over time is critical, not only for understanding the physiological stress responses, but also to understand their implications on mental health.
Collapse
Affiliation(s)
- Lívea Dornela Godoy
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Matheus Teixeira Rossignoli
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Polianna Delfino-Pereira
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Norberto Garcia-Cairasco
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, São Paulo, Brazil
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Eduardo Henrique de Lima Umeoka
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
21
|
Jones DK, Hintz WD, Schuler MS, Yates EK, Mattes BM, Relyea RA. Inducible Tolerance to Agrochemicals Was Paved by Evolutionary Responses to Predators. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2017; 51:13913-13919. [PMID: 29087697 DOI: 10.1021/acs.est.7b03816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Recent research has reported increased tolerance to agrochemicals in target and nontarget organisms following acute physiological changes induced through phenotypic plasticity. Moreover, the most inducible populations are those from more pristine locations, far from agrochemical use. We asked why do populations with no known history of pesticide exposure have the ability to induce adaptive responses to novel agrochemicals? We hypothesized that increased pesticide tolerance results from a generalized stressor response in organisms, and would be induced following sublethal exposure to natural and anthropogenic stressors. We exposed larval wood frogs (Lithobates sylvaticus) to one of seven natural or anthropogenic stressors (predator cue (Anax spp.), 0.5 or 1.0 mg carbaryl/L, road salt (200 or 1000 mg Cl-/L), ethanol-vehicle control, or no-stressor control) and subsequently tested their tolerance to a lethal carbaryl concentration using time-to-death assays. We observed induced carbaryl tolerance in tadpoles exposed to 0.5 mg/L carbaryl and also in tadpoles exposed to predator cues. Our results suggest that the ability to induce pesticide tolerance likely arose through evolved antipredator responses. Given that antipredator responses are widespread among species, many animals might possess inducible pesticide tolerance, buffering them from agrochemical exposure.
Collapse
Affiliation(s)
- Devin K Jones
- Darrin Fresh Water Institute, Department of Biological Sciences, Rensselaer Polytechnic Institute , Troy, New York 12180, United States
| | - William D Hintz
- Darrin Fresh Water Institute, Department of Biological Sciences, Rensselaer Polytechnic Institute , Troy, New York 12180, United States
| | - Matthew S Schuler
- Darrin Fresh Water Institute, Department of Biological Sciences, Rensselaer Polytechnic Institute , Troy, New York 12180, United States
| | - Erika K Yates
- Darrin Fresh Water Institute, Department of Biological Sciences, Rensselaer Polytechnic Institute , Troy, New York 12180, United States
| | - Brian M Mattes
- Darrin Fresh Water Institute, Department of Biological Sciences, Rensselaer Polytechnic Institute , Troy, New York 12180, United States
| | - Rick A Relyea
- Darrin Fresh Water Institute, Department of Biological Sciences, Rensselaer Polytechnic Institute , Troy, New York 12180, United States
| |
Collapse
|
22
|
Georgatza D, Gorgogietas VA, Kylindri P, Charalambous MC, Papadopoulou KK, Hayes JM, Psarra AMG. The triterpene echinocystic acid and its 3-O-glucoside derivative are revealed as potent and selective glucocorticoid receptor agonists. Int J Biochem Cell Biol 2016; 79:277-287. [DOI: 10.1016/j.biocel.2016.08.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 07/19/2016] [Accepted: 08/29/2016] [Indexed: 12/20/2022]
|
23
|
Mechanisms of cortisol action in fish hepatocytes. Comp Biochem Physiol B Biochem Mol Biol 2016; 199:136-145. [DOI: 10.1016/j.cbpb.2016.06.012] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/21/2016] [Accepted: 06/27/2016] [Indexed: 12/17/2022]
|
24
|
Hossny E, Rosario N, Lee BW, Singh M, El-Ghoneimy D, SOH JY, Le Souef P. The use of inhaled corticosteroids in pediatric asthma: update. World Allergy Organ J 2016; 9:26. [PMID: 27551328 PMCID: PMC4982274 DOI: 10.1186/s40413-016-0117-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 07/21/2016] [Indexed: 02/07/2023] Open
Abstract
Despite the availability of several formulations of inhaled corticosteroids (ICS) and delivery devices for treatment of childhood asthma and despite the development of evidence-based guidelines, childhood asthma control remains suboptimal. Improving uptake of asthma management plans, both by families and practitioners, is needed. Adherence to daily ICS therapy is a key determinant of asthma control and this mandates that asthma education follow a repetitive pattern and involve literal explanation and physical demonstration of the optimal use of inhaler devices. The potential adverse effects of ICS need to be weighed against the benefit of these drugs to control persistent asthma especially that its safety profile is markedly better than oral glucocorticoids. This article reviews the key mechanisms of inhaled corticosteroid action; recommendations on dosage and therapeutic regimens; potential optimization of effectiveness by addressing inhaler technique and adherence to therapy; and updated knowledge on the real magnitude of adverse events.
Collapse
Affiliation(s)
- Elham Hossny
- Pediatric Allergy and Immunology Unit, Children’s Hospital, Ain Shams University, Cairo, 11566 Egypt
| | | | - Bee Wah Lee
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Meenu Singh
- Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Dalia El-Ghoneimy
- Pediatric Allergy and Immunology Unit, Children’s Hospital, Ain Shams University, Cairo, 11566 Egypt
| | - Jian Yi SOH
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Peter Le Souef
- Winthrop Professor of Paediatrics & Child Health, School of Paediatrics & Child Health, University of Western Australia, Crawley, Australia
| |
Collapse
|
25
|
Pulina G, Nudda A, Macciotta NPP, Battacone G, Giacomo Rassu SP, Cannas A. Non-nutritional factors affecting lactation persistency in dairy ewes: a review. ITALIAN JOURNAL OF ANIMAL SCIENCE 2016. [DOI: 10.4081/ijas.2007.115] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
26
|
|
27
|
Jiang CL, Liu L, Li Z, Buttgereit F. The novel strategy of glucocorticoid drug development via targeting nongenomic mechanisms. Steroids 2015; 102:27-31. [PMID: 26122209 DOI: 10.1016/j.steroids.2015.06.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Revised: 06/13/2015] [Accepted: 06/22/2015] [Indexed: 12/20/2022]
Abstract
Glucocorticoids (GCs) are widely used in clinical practice as potent anti-inflammatory and immunosuppressive agents. Unfortunately, they can also produce numerous and potentially serious side effects that limit their usage. This problem represents the driving force for the intensive search for novel GCs with a better benefit-risk ratio compared to conventional GCs. GCs are believed to take effects mainly through classical genomic mechanisms, which are also largely responsible for GCs' side effects. However, in addition to these genomic effects, GCs also demonstrate rapid genomic-independent activities. It has become increasingly evident that some of the anti-inflammatory, immunosuppressive, anti-allergic and anti-shock effects of GCs could be mediated through nongenomic mechanisms. Thus, theoretically, trying to use nongenomic mechanisms of GCs more intensively may represent a novel strategy for development of GCs with low side effect profile. The new GCs' drugs will take clinical effects mainly via nongenomic mechanisms and do not execute the classical genomic mechanism to reduce side effects.
Collapse
Affiliation(s)
- Chun-Lei Jiang
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, 200433 Shanghai, PR China.
| | - Lei Liu
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, 200433 Shanghai, PR China
| | - Zhen Li
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, 201203 Shanghai, PR China
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, 10117 Berlin, Germany.
| |
Collapse
|
28
|
Yu J, Park MH, Choi SY, Jo SH. Cortisone and hydrocortisone inhibit human Kv1.3 activity in a non-genomic manner. Naunyn Schmiedebergs Arch Pharmacol 2015; 388:653-61. [PMID: 25743574 DOI: 10.1007/s00210-015-1109-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/18/2015] [Indexed: 11/24/2022]
Abstract
Glucocorticoids are hormones released in response to stress that are involved in various physiological processes including immune functions. One immune-modulating mechanism is achieved by the Kv1.3 voltage-dependent potassium channel, which is expressed highly in lymphocytes including effector memory T lymphocytes (TEM). Although glucocorticoids are known to inhibit Kv1.3 function, the detailed inhibitory mechanism is not yet fully understood. Here we studied the rapid non-genomic effects of cortisone and hydrocortisone on the human Kv1.3 channel expressed in Xenopus oocytes. Both cortisone and hydrocortisone reduced the amplitude of the Kv1.3 channel current in a concentration-dependent manner. Both cortisone and hydrocortisone rapidly and irreversibly inhibited Kv1.3 currents, eliminating the possibility of genomic regulation. Inhibition rate was stable relative to the degree of depolarization. Kinetically, cortisone altered the activating gate of Kv1.3 and hydrocortisone interacted with this channel in an open state. These results suggest that cortisone and hydrocortisone inhibit Kv1.3 currents via a non-genomic mechanism, providing a mechanism for the immunosuppressive effects of glucocorticoids.
Collapse
Affiliation(s)
- Jing Yu
- Department of Physiology, Institute of Bioscience and Biotechnology, BK21 plus Graduate Program, Kangwon National University College of Medicine, Hyoja-Dong, Chuncheon, 200-701, Republic of Korea
| | | | | | | |
Collapse
|
29
|
Yu J, Park MH, Jo SH. Inhibitory effects of cortisone and hydrocortisone on human Kv1.5 channel currents. Eur J Pharmacol 2014; 746:158-66. [PMID: 25449034 DOI: 10.1016/j.ejphar.2014.11.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 10/31/2014] [Accepted: 11/07/2014] [Indexed: 01/23/2023]
Abstract
Glucocorticoids are the primary hormones that respond to stress and protect organisms from dangerous situations. The glucocorticoids hydrocortisone and its dormant form, cortisone, affect the cardiovascular system with changes such as increased blood pressure and cardioprotection. Kv1.5 channels play a critical role in the maintenance of cellular membrane potential and are widely expressed in pancreatic β-cells, neurons, myocytes, and smooth muscle cells of the pulmonary vasculature. We examined the electrophysiological effects of both cortisone and hydrocortisone on human Kv1.5 channels expressed in Xenopus oocytes using a two-microelectrode voltage clamp technique. Both cortisone and hydrocortisone rapidly and irreversibly suppressed the amplitude of Kv1.5 channel current with IC50 values of 50.2±4.2μM and 33.4±3.2μM, respectively, while sustained the current trace shape of Kv1.5 current. The inhibitory effect of cortisone on Kv1.5 decreased progressively from -10mV to +30mV, while hydrocortisone׳s inhibition of the channel did not change across the same voltage range. Both cortisone and hydrocortisone blocked Kv1.5 channel currents in a non-use-dependent manner and neither altered the channel׳s steady-state activation or inactivation curves. These results show that cortisone and hydrocortisone inhibited Kv1.5 channel currents differently, and that Kv1.5 channels were more sensitive to hydrocortisone than to cortisone.
Collapse
Affiliation(s)
- Jing Yu
- Department of Physiology, Institute of Bioscience and Biotechnology, BK21 plus Graduate Program, Kangwon National University School of Medicine, Chuncheon 200-701, Republic of Korea
| | - Mi-Hyeong Park
- Department of Physiology, Institute of Bioscience and Biotechnology, BK21 plus Graduate Program, Kangwon National University School of Medicine, Chuncheon 200-701, Republic of Korea
| | - Su-Hyun Jo
- Department of Physiology, Institute of Bioscience and Biotechnology, BK21 plus Graduate Program, Kangwon National University School of Medicine, Chuncheon 200-701, Republic of Korea.
| |
Collapse
|
30
|
Boardman C, Chachi L, Gavrila A, Keenan CR, Perry MM, Xia YC, Meurs H, Sharma P. Mechanisms of glucocorticoid action and insensitivity in airways disease. Pulm Pharmacol Ther 2014; 29:129-43. [PMID: 25218650 DOI: 10.1016/j.pupt.2014.08.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 08/18/2014] [Accepted: 08/25/2014] [Indexed: 01/04/2023]
Abstract
Glucocorticoids are the mainstay for the treatment of chronic inflammatory diseases including asthma and chronic obstructive pulmonary disease (COPD). However, it has been recognized that glucocorticoids do not work well in certain patient populations suggesting reduced sensitivity. The ultimate biologic responses to glucocorticoids are determined by not only the concentration of glucocorticoids but also the differences between individuals in glucocorticoid sensitivity, which is influenced by multiple factors. Studies are emerging to understand these mechanisms in detail, which would help in increasing glucocorticoid sensitivity in patients with chronic airways disease. This review aims to highlight both classical and emerging concepts of the anti-inflammatory mechanisms of glucocorticoids and also review some novel strategies to overcome steroid insensitivity in airways disease.
Collapse
Affiliation(s)
- C Boardman
- Airway Disease, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - L Chachi
- Institute for Lung Health, Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - A Gavrila
- Institute for Lung Health, Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - C R Keenan
- Department of Pharmacology, University of Melbourne, Parkville, Victoria, Australia
| | - M M Perry
- Airway Disease, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Y C Xia
- Department of Pharmacology, University of Melbourne, Parkville, Victoria, Australia
| | - H Meurs
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands
| | - P Sharma
- Department of Physiology and Pharmacology, Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, 4C46 HRIC, 3280 Hospital Dr NW, Calgary, AB, Canada T2N 4N1.
| |
Collapse
|
31
|
Abstract
Social isolation has been recognized as a major risk factor for morbidity and mortality in humans for more than a quarter of a century. Although the focus of research has been on objective social roles and health behavior, the brain is the key organ for forming, monitoring, maintaining, repairing, and replacing salutary connections with others. Accordingly, population-based longitudinal research indicates that perceived social isolation (loneliness) is a risk factor for morbidity and mortality independent of objective social isolation and health behavior. Human and animal investigations of neuroendocrine stress mechanisms that may be involved suggest that (a) chronic social isolation increases the activation of the hypothalamic pituitary adrenocortical axis, and (b) these effects are more dependent on the disruption of a social bond between a significant pair than objective isolation per se. The relational factors and neuroendocrine, neurobiological, and genetic mechanisms that may contribute to the association between perceived isolation and mortality are reviewed.
Collapse
Affiliation(s)
- John T Cacioppo
- Department of Psychology, University of Chicago, Chicago, Illinois 60637;
| | | | | | | |
Collapse
|
32
|
Dominguez G, Faucher P, Henkous N, Krazem A, Piérard C, Béracochéa D. Stress induced a shift from dorsal hippocampus to prefrontal cortex dependent memory retrieval: role of regional corticosterone. Front Behav Neurosci 2014; 8:166. [PMID: 24860451 PMCID: PMC4030165 DOI: 10.3389/fnbeh.2014.00166] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 04/18/2014] [Indexed: 12/25/2022] Open
Abstract
Most of the deleterious effects of stress on memory retrieval are due to a dysfunction of the hippocampo-prefrontal cortex interplay. The role of the stress-induced regional corticosterone increase in such dysfunction remains however unclear, since there is no published study as yet dedicated to measuring corticosterone concentrations simultaneously in both the prefrontal cortex (mPFC) and the hippocampus (dHPC) in relation with memory impairments. To that aim, we first showed in Experiment 1 that an acute stress (3 electric footschocks; 0.9 mA each) delivered before memory testing reversed the memory retrieval pattern (MRP) in a serial discrimination task in which mice learned two successive discriminations. More precisely, whereas non-stressed animals remembered accurately the first learned discrimination and not the second one, stressed mice remembered more accurately the second discrimination but not the first one. We demonstrated that local inactivation of dHPC or mPFC with the anesthetic lidocaine recruited the dHPC activity in non-stress conditions whereas the stress-induced MRP inversion recruited the mPFC activity. In a second experiment, we showed that acute stress induced a very similar time-course evolution of corticosterone rises within both the mPFC and dHPC. In a 3rd experiment, we found however that in situ injections of corticosterone either within the mPFC or the dHPC before memory testing favored the emergence of the mPFC-dependent MRP but blocked the emergence of the dHPC-dependent one. Overall, our study evidences that the simultaneous increase of corticosterone after stress in both areas induces a shift from dHPC (non-stress condition) to mPFC-dependent MRP and that corticosterone is critically involved in mediating the deleterious effects of stress on cognitive functions involving the mPFC-HPC interplay.
Collapse
Affiliation(s)
- Gaelle Dominguez
- INSERM U-930, Université François Rabelais, Parc Grandmont Tours, France
| | - Pierre Faucher
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, CNRS UMR 5287, Nouvelle Université de Bordeaux Talence, France
| | - Nadia Henkous
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, CNRS UMR 5287, Nouvelle Université de Bordeaux Talence, France
| | - Ali Krazem
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, CNRS UMR 5287, Nouvelle Université de Bordeaux Talence, France
| | | | - Daniel Béracochéa
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, CNRS UMR 5287, Nouvelle Université de Bordeaux Talence, France
| |
Collapse
|
33
|
Moisan MP, Minni AM, Dominguez G, Helbling JC, Foury A, Henkous N, Dorey R, Béracochéa D. Role of corticosteroid binding globulin in the fast actions of glucocorticoids on the brain. Steroids 2014; 81:109-15. [PMID: 24252379 DOI: 10.1016/j.steroids.2013.10.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Corticosteroid binding globulin (CBG) is a glycoprotein synthesized in liver and secreted in the blood where it binds with a high affinity but low capacity glucocorticoid hormones, cortisol in humans and corticosterone in laboratory rodents. In mammals, 95% of circulating glucocorticoids are bound to either CBG (80%) or albumin (15%) and only the 5% free fraction is able to enter the brain. During stress, the concentration of glucocorticoids rises significantly and the free fraction increases even more because CBG becomes saturated. However, glucocorticoids unbound to CBG are cleared from the blood more quickly. Our studies on mice totally devoid of CBG (Cbg k.o.) showed that during stress these mutant mice display a lower rise of glucocorticoids than the wild-type controls associated with altered emotional reactivity. These data suggested that CBG played a role in the fast actions of glucocorticoids on behavior. Further analyses demonstrated that stress-induced memory retrieval impairment, an example of the fast action of glucocorticoids on the brain is abolished in the Cbg k.o. mice. This effect of stress on memory retrieval could be restored in the Cbg k.o. mice by infusing corticosterone directly in the hippocampus. The mechanisms explaining these effects involved an increased clearance but no difference in corticosterone production. Thus, CBG seems to have an important role in maintaining in blood a glucocorticoid pool that will be able to access the brain for the fast effects of glucocorticoids.
Collapse
Affiliation(s)
- M P Moisan
- INRA, Nutrition & Neurobiologie Intégrée (NutriNeuro), UMR 1286, 33076 Bordeaux, France; Univ Bordeaux, Nutrition & Neurobiologie Intégrée (NutriNeuro), UMR 1286, 33076 Bordeaux, France.
| | - A M Minni
- INRA, Nutrition & Neurobiologie Intégrée (NutriNeuro), UMR 1286, 33076 Bordeaux, France; Univ Bordeaux, Nutrition & Neurobiologie Intégrée (NutriNeuro), UMR 1286, 33076 Bordeaux, France
| | - G Dominguez
- CNRS, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), UMR 5287, Universités de Bordeaux1 et 2, 33400 Talence, France; UFR Sciences et Technique, Université de Tours, Parc de Grandmont, 37200 Tours, France
| | - J C Helbling
- INRA, Nutrition & Neurobiologie Intégrée (NutriNeuro), UMR 1286, 33076 Bordeaux, France; Univ Bordeaux, Nutrition & Neurobiologie Intégrée (NutriNeuro), UMR 1286, 33076 Bordeaux, France
| | - A Foury
- INRA, Nutrition & Neurobiologie Intégrée (NutriNeuro), UMR 1286, 33076 Bordeaux, France; Univ Bordeaux, Nutrition & Neurobiologie Intégrée (NutriNeuro), UMR 1286, 33076 Bordeaux, France
| | - N Henkous
- CNRS, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), UMR 5287, Universités de Bordeaux1 et 2, 33400 Talence, France
| | - R Dorey
- CNRS, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), UMR 5287, Universités de Bordeaux1 et 2, 33400 Talence, France
| | - D Béracochéa
- CNRS, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), UMR 5287, Universités de Bordeaux1 et 2, 33400 Talence, France
| |
Collapse
|
34
|
Santana PPB, Carvalho CMF, da Costa NN, Silva TVG, Ramos PCA, Cordeiro MS, Santos SSD, Khayat AS, Ohashi OM, Miranda MS. Effect of dexamethasone on development of in vitro-produced bovine embryos. Theriogenology 2014; 82:10-6. [PMID: 24656431 DOI: 10.1016/j.theriogenology.2014.02.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 02/05/2014] [Accepted: 02/15/2014] [Indexed: 11/15/2022]
Abstract
Studies in somatic cells have shown that glucocorticoids such as dexamethasone (DEX) may trigger or prevent apoptosis depending on the cell type in culture. Because the dysregulation of apoptosis may lower in vitro embryo production efficiency, we sought to investigate the effects of supplementing IVC medium with DEX (0.1 μg/mL) on embryo morphology, development kinetics, and apoptosis rates of in vitro-produced bovine preimplantation embryos. Embryo morphology was graded on Day 7, and development rates were assessed on Days 4 and 7 of IVC. Apoptosis was evaluated via annexin/propidium iodide staining under fluorescence microscopy where a cell labeled with annexin, propidium iodide, or both would be considered apoptotic. An embryo was counted in the apoptosis rates, if it displayed at least one such labeled cell. Although DEX supplementation did not reduce apoptosis rates, it had a positive impact on developmental kinetics and cell number both on Days 4 and 7 of embryo culture. Presumably, such effect resulted from increased cell proliferation rather than a direct inhibition of apoptosis. Further studies may evaluate the mechanisms by which glucocorticoids may affect embryo development, as DEX supplementation could become a tool to improve in vitro embryo yield in mammalian species.
Collapse
Affiliation(s)
- Priscila P B Santana
- Institute of Biological Sciences, Federal University of Pará, Belém, Para, Brazil.
| | - Carla M F Carvalho
- Institute of Biological Sciences, Federal University of Pará, Belém, Para, Brazil
| | - Nathália N da Costa
- Institute of Biological Sciences, Federal University of Pará, Belém, Para, Brazil
| | - Thiago V G Silva
- Institute of Biological Sciences, Federal University of Pará, Belém, Para, Brazil
| | - Priscilla C A Ramos
- Institute of Biological Sciences, Federal University of Pará, Belém, Para, Brazil
| | - Marcela S Cordeiro
- Federal Institute of Education, Science and Technology of Pará, Abaetetuba, Para, Brazil
| | - Simone S D Santos
- Institute of Biological Sciences, Federal University of Pará, Belém, Para, Brazil
| | - André S Khayat
- Institute of Biological Sciences, Federal University of Pará, Belém, Para, Brazil
| | - Otávio M Ohashi
- Institute of Biological Sciences, Federal University of Pará, Belém, Para, Brazil
| | - Moysés S Miranda
- Institute of Biological Sciences, Federal University of Pará, Belém, Para, Brazil
| |
Collapse
|
35
|
Jiang CL, Liu L, Tasker JG. Why do we need nongenomic glucocorticoid mechanisms? Front Neuroendocrinol 2014; 35:72-5. [PMID: 24103541 DOI: 10.1016/j.yfrne.2013.09.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 09/11/2013] [Accepted: 09/30/2013] [Indexed: 10/26/2022]
Abstract
Glucocorticoids (GCs) are a class of steroid hormones that have been known to be involved in various physiological processes and to play a pivotal role in preserving basal and stress-related homeostasis. GCs are also widely used clinically as anti-inflammatory, immunosuppressive, anti-shock drugs. It is believed traditionally that GCs exert most of their effects genomically. In addition to the well-known classical genomic mechanisms, GCs also affect various functions via rapid, nongenomic mechanisms. The therapeutic benefits of nongenomic GC actions have been exploited in clinical medicine, especially with high-dose pulsed glucocorticoid administration. However, it is certainly not the case that the inherent nongenomic glucocorticoid mechanisms evolved only for their clinical utility. Here, we review the recent literature on nongenomic actions of GCs related to stress and the physiological significance of these actions, and we propose reasons why nongenomic mechanisms of GC actions are needed.
Collapse
Affiliation(s)
- Chun-Lei Jiang
- Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China; Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, PR China.
| | - Lei Liu
- Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Jeffrey G Tasker
- Neurobiology Division, Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| |
Collapse
|
36
|
Török Z, Crul T, Maresca B, Schütz GJ, Viana F, Dindia L, Piotto S, Brameshuber M, Balogh G, Péter M, Porta A, Trapani A, Gombos I, Glatz A, Gungor B, Peksel B, Vigh L, Csoboz B, Horváth I, Vijayan MM, Hooper PL, Harwood JL, Vigh L. Plasma membranes as heat stress sensors: from lipid-controlled molecular switches to therapeutic applications. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:1594-618. [PMID: 24374314 DOI: 10.1016/j.bbamem.2013.12.015] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 12/09/2013] [Accepted: 12/18/2013] [Indexed: 12/31/2022]
Abstract
The classic heat shock (stress) response (HSR) was originally attributed to protein denaturation. However, heat shock protein (Hsp) induction occurs in many circumstances where no protein denaturation is observed. Recently considerable evidence has been accumulated to the favor of the "Membrane Sensor Hypothesis" which predicts that the level of Hsps can be changed as a result of alterations to the plasma membrane. This is especially pertinent to mild heat shock, such as occurs in fever. In this condition the sensitivity of many transient receptor potential (TRP) channels is particularly notable. Small temperature stresses can modulate TRP gating significantly and this is influenced by lipids. In addition, stress hormones often modify plasma membrane structure and function and thus initiate a cascade of events, which may affect HSR. The major transactivator heat shock factor-1 integrates the signals originating from the plasma membrane and orchestrates the expression of individual heat shock genes. We describe how these observations can be tested at the molecular level, for example, with the use of membrane perturbers and through computational calculations. An important fact which now starts to be addressed is that membranes are not homogeneous nor do all cells react identically. Lipidomics and cell profiling are beginning to address the above two points. Finally, we observe that a deregulated HSR is found in a large number of important diseases where more detailed knowledge of the molecular mechanisms involved may offer timely opportunities for clinical interventions and new, innovative drug treatments. This article is part of a Special Issue entitled: Membrane Structure and Function: Relevance in the Cell's Physiology, Pathology and Therapy.
Collapse
Affiliation(s)
- Zsolt Török
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary.
| | - Tim Crul
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Bruno Maresca
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Salerno, Italy
| | - Gerhard J Schütz
- Institute of Applied Physics, Vienna University of Technology, Wiedner Hauptstrasse 8-10, 1040 Vienna, Austria
| | - Felix Viana
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550 San Juan de Alicante, Spain
| | - Laura Dindia
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Stefano Piotto
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Salerno, Italy
| | - Mario Brameshuber
- Institute of Applied Physics, Vienna University of Technology, Wiedner Hauptstrasse 8-10, 1040 Vienna, Austria
| | - Gábor Balogh
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Mária Péter
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Amalia Porta
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Salerno, Italy
| | - Alfonso Trapani
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Salerno, Italy
| | - Imre Gombos
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Attila Glatz
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Burcin Gungor
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Begüm Peksel
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - László Vigh
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Bálint Csoboz
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Ibolya Horváth
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary
| | - Mathilakath M Vijayan
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada; Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Phillip L Hooper
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Medical School, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - John L Harwood
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, Wales, UK
| | - László Vigh
- Institute of Biochemistry, Biological Research Centre of the Hung. Acad. Sci., Szeged H-6726, Hungary.
| |
Collapse
|
37
|
Harris BN, de Jong TR, Yang V, Saltzman W. Chronic variable stress in fathers alters paternal and social behavior but not pup development in the biparental California mouse (Peromyscus californicus). Horm Behav 2013; 64:799-811. [PMID: 24157379 PMCID: PMC3894746 DOI: 10.1016/j.yhbeh.2013.10.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 10/09/2013] [Accepted: 10/11/2013] [Indexed: 02/02/2023]
Abstract
Stress and chronically elevated glucocorticoid levels have been shown to disrupt parental behavior in mothers; however, almost no studies have investigated corresponding effects in fathers. The present experiment tested the hypothesis that chronic variable stress inhibits paternal behavior and consequently alters pup development in the monogamous, biparental California mouse (Peromyscus californicus). First-time fathers were assigned to one of three experimental groups: chronic variable stress (CVS, n=8), separation control (SC, n=7), or unmanipulated control (UC, n=8). The CVS paradigm (3 stressors per day for 7 days) successfully stressed mice, as evidenced by increased baseline plasma corticosterone concentrations, increased adrenal mass, decreased thymus mass, and a decrease in body mass over time. CVS altered paternal and social behavior of fathers, but major differences were observed only on day 6 of the 7-day paradigm. At that time point, CVS fathers spent less time with their pairmate and pups, and more time autogrooming, as compared to UC fathers; SC fathers spent more time behaving paternally and grooming the female mate than CVS and UC fathers. Thus, CVS blocked the separation-induced increase in social behaviors observed in the SC fathers. Nonetheless, chronic stress in fathers did not appear to alter survival or development of their offspring: pups from the three experimental conditions did not differ in body mass gain over time, in the day of eye opening, or in basal or post-stress corticosterone levels. These results demonstrate that chronic stress can transiently disrupt paternal and social behavior in P. californicus fathers, but does not alter pup development or survival under controlled, non-challenging laboratory conditions.
Collapse
Affiliation(s)
- Breanna N Harris
- Department of Biology, University of California, Riverside, CA 92521, USA; Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA.
| | | | | | | |
Collapse
|
38
|
Johnstone WM, Mills KA, Alyea RA, Thomas P, Borski RJ. Characterization of membrane receptor binding activity for cortisol in the liver and kidney of the euryhaline teleost, Mozambique tilapia (Oreochromis mossambicus). Gen Comp Endocrinol 2013; 192:107-14. [PMID: 23851043 DOI: 10.1016/j.ygcen.2013.06.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 06/11/2013] [Accepted: 06/19/2013] [Indexed: 11/22/2022]
Abstract
Glucocorticoids (GCs) regulate an array of physiological responses in vertebrates. Genomic GC actions mediated by nuclear steroid receptors require a lag time on the order of hours to days to generate an appreciable physiological response. Experimental evidence has accumulated that GCs, can also act rapidly through a nongenomic mechanism to modulate cellular physiology in vertebrates. Causal evidence in the Mozambique tilapia (Oreochromis mossambicus) suggests that the GC cortisol exerts rapid, nongenomic actions in the gills, liver, and pituitary of this euryhaline teleost, but the membrane receptor mediating these actions has not been characterized. Radioreceptor binding assays were conducted to identify a putative GC membrane receptor site in O. mossambicus. The tissue distribution, binding kinetics, and pharmacological signature of the GC membrane-binding activity were characterized. High affinity (Kd=9.527±0.001 nM), low-capacity (Bmax=1.008±0.116 fmol/mg protein) [(3)H] cortisol binding was identified on plasma membranes prepared from the livers and a lower affinity (Kd=30.08±2.373 nM), low capacity (Bmax=4.690±2.373 fmol/mg protein) binding was found in kidney membrane preparations. Competitors with high binding affinity for nuclear GC receptors, mifepristone (RU486), dexamethasone, and 11-deoxycorticosterone, displayed no affinity for the membrane GC receptor. The association and dissociation kinetics of [(3)H] cortisol binding to membranes were orders of magnitude faster (t1/2=1.7-2.6 min) than those for the intracellular (nuclear) GC receptor (t1/2=10.2h). Specific [(3)H] cortisol membrane binding was also detected in the gill and pituitary but not in brain tissue. This study represents the first characterization of a membrane GC receptor in fishes and one of only a few characterized in vertebrates.
Collapse
Affiliation(s)
- William M Johnstone
- Department of Biology, North Carolina State University, Raleigh, NC 27695, USA
| | | | | | | | | |
Collapse
|
39
|
Stress-induced memory retrieval impairments: different time-course involvement of corticosterone and glucocorticoid receptors in dorsal and ventral hippocampus. Neuropsychopharmacology 2012; 37:2870-80. [PMID: 22948976 PMCID: PMC3499833 DOI: 10.1038/npp.2012.170] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The present study was aimed at determining the relative contribution of the dorsal (DH) and ventral (VH) hippocampus in stress-induced memory retrieval impairments. Thus, we studied the temporal involvement of corticosterone and its receptors, i.e. mineralocorticoid (MR) and glucocorticoid (GR) in the DH and VH, in relation with the time-course evolution of stress-induced memory retrieval impairments. In a first experiment, double microdialysis allowed showing on the same animal that an acute stress (electric footshocks) induced an earlier corticosterone rise in the DH (15-60 min post-stress) and then in the VH (90-105 min post-stress). The return to baseline was faster in the DH (105 min) than in the VH (120 min). Memory deficits assessed by delayed alternation occurred at 15-, 60-, and 105-min delays after stress and were closely related to the kinetic of corticosterone rises within the DH and VH. In a second experiment, the GR antagonist RU-38486 and the MR antagonist RU-28318 were administered in the DH or VH 15 min before stress. RU-38486 restored memory at 60 but not at 105 min post-stress delays in the DH, whereas the opposite pattern was observed in the VH. By contrast, RU-28318 had no effect on memory impairments at both the 60- and 105-min post-stress delays, showing that MR receptors are not involved at these delays. However, RU-28318 administered in the DH restored memory when administered at a shorter post-stress delay (15 min). Overall, our data are first to evidence that stress induces a functional switch from the DH to VH via different corticosterone time-course evolutions in these areas and the sequential GR receptors involvement in the DH and then in the VH, as regards the persistence of stress-induced memory retrieval deficits over time.
Collapse
|
40
|
Dindia L, Murray J, Faught E, Davis TL, Leonenko Z, Vijayan MM. Novel nongenomic signaling by glucocorticoid may involve changes to liver membrane order in rainbow trout. PLoS One 2012; 7:e46859. [PMID: 23056491 PMCID: PMC3466178 DOI: 10.1371/journal.pone.0046859] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 09/06/2012] [Indexed: 11/21/2022] Open
Abstract
Stress-induced glucocorticoid elevation is a highly conserved response among vertebrates. This facilitates stress adaptation and the mode of action involves activation of the intracellular glucocorticoid receptor leading to the modulation of target gene expression. However, this genomic effect is slow acting and, therefore, a role for glucocorticoid in the rapid response to stress is unclear. Here we show that stress levels of cortisol, the primary glucocorticoid in teleosts, rapidly fluidizes rainbow trout (Oncorhynchus mykiss) liver plasma membranes in vitro. This involved incorporation of the steroid into the lipid domains, as cortisol coupled to a membrane impermeable peptide moiety, did not affect membrane order. Studies confirmed that cortisol, but not sex steroids, increases liver plasma membrane fluidity. Atomic force microscopy revealed cortisol-mediated changes to membrane surface topography and viscoelasticity confirming changes to membrane order. Treating trout hepatocytes with stress levels of cortisol led to the modulation of cell signaling pathways, including the phosphorylation status of putative PKA, PKC and AKT substrate proteins within 10 minutes. The phosphorylation by protein kinases in the presence of cortisol was consistent with that seen with benzyl alcohol, a known membrane fluidizer. Our results suggest that biophysical changes to plasma membrane properties, triggered by stressor-induced glucocorticoid elevation, act as a nonspecific stress response and may rapidly modulate acute stress-signaling pathways.
Collapse
Affiliation(s)
- Laura Dindia
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Josh Murray
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Erin Faught
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Tracy L. Davis
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Zoya Leonenko
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | | |
Collapse
|
41
|
Joëls M, Sarabdjitsingh RA, Karst H. Unraveling the time domains of corticosteroid hormone influences on brain activity: rapid, slow, and chronic modes. Pharmacol Rev 2012; 64:901-38. [PMID: 23023031 DOI: 10.1124/pr.112.005892] [Citation(s) in RCA: 306] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Brain cells are continuously exposed to corticosteroid hormones, although the levels vary (e.g., after stress). Corticosteroids alter neural activity via two receptor types, mineralocorticoid (MR) and glucocorticoid receptors (GR). These receptors regulate gene transcription but also, as we now know, act nongenomically. Via nongenomic pathways, MRs enhance and GRs suppress neural activity. In the hypothalamus, inhibitory GR effects contribute to negative feedback regulation of the stress axis. Nongenomic MR actions are also important extrahypothalamically and help organisms to immediately select an appropriate response strategy. Via genomic mechanisms, corticosteroid actions in the basolateral amygdala and ventral-most part of the cornu ammonis 1 hippocampal area are generally excitatory, providing an extended window for encoding of emotional aspects of a stressful event. GRs in hippocampal and prefrontal pyramidal cells increase surface expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors and strengthen glutamatergic signaling through pathways partly overlapping with those involved in long-term potentiation. This raises the threshold for subsequent induction of synaptic potentiation and promotes long-term depression. Synapses activated during stress are thus presumably strengthened but protected against excitatory inputs reaching the cells later. This restores higher cognitive control and promotes, for example, consolidation of stress-related contextual information. When an organism experiences stress early in life or repeatedly in adulthood, the ability to induce synaptic potentiation is strongly reduced and the likelihood to induce depression enhanced, even under rest. Treatment with antiglucocorticoids can ameliorate cellular effects after chronic stress and thus provide an interesting lead for treatment of stress-related disorders.
Collapse
Affiliation(s)
- Marian Joëls
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | |
Collapse
|
42
|
Hawkley LC, Cole SW, Capitanio JP, Norman GJ, Cacioppo JT. Effects of social isolation on glucocorticoid regulation in social mammals. Horm Behav 2012; 62:314-23. [PMID: 22663934 PMCID: PMC3449017 DOI: 10.1016/j.yhbeh.2012.05.011] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 05/23/2012] [Accepted: 05/24/2012] [Indexed: 12/24/2022]
Abstract
The regulation and function of the hypothalamic-pituitary-adrenocortical (HPA) axis and glucocorticoids have been well conserved across vertebrate species. Glucocorticoids influence a wide range of physiological functions that include glucose regulation, metabolism, inflammatory control, as well as cardiovascular, reproductive, and neuronal effects. Some of these are relatively quick-acting non-genomic effects, but most are slower-acting genomic effects. Thus, any stimulus that affects HPA function has the potential to exert wide-ranging short-term and long-term effects on much of vertebrate physiology. Here, we review the effects of social isolation on the functioning of the HPA axis in social species, and on glucocorticoid physiology in social mammals in particular. Evidence indicates that objective and perceived social isolation alter HPA regulation, although the nature and direction of the HPA response differs among species and across age. The inconsistencies in the direction and nature of HPA effects have implications for drawing cross-species conclusions about the effects of social isolation, and are particularly problematic for understanding HPA-related physiological processes in humans. The animal and human data are incommensurate because, for example, animal studies of objective isolation have typically not been modeled on, or for comparability with, the subjective experience of isolation in humans. An animal model of human isolation must be taken more seriously if we want to advance our understanding of the mechanisms for the effects of objective and perceived isolation in humans.
Collapse
Affiliation(s)
- Louise C Hawkley
- Department of Psychology and Center for Cognitive and Social Neuroscience, University of Chicago, Chicago, IL, USA.
| | | | | | | | | |
Collapse
|
43
|
Park KM, Yoo JH, Shin YJ. Effects of Egg Shell Membrane Hydrolysates on Skin Whitening, Wound Healing, and UV-Protection. Korean J Food Sci Anim Resour 2012. [DOI: 10.5851/kosfa.2012.32.3.308] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
44
|
Tesone AJ, Regueira E, Canosa LF, Ceballos NR. 5α-Reductase, an enzyme regulating glucocorticoid action in the testis of Rhinella arenarum (Amphibia: Anura). Gen Comp Endocrinol 2012; 176:500-6. [PMID: 22285601 DOI: 10.1016/j.ygcen.2012.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Revised: 01/03/2012] [Accepted: 01/07/2012] [Indexed: 11/26/2022]
Abstract
The reduction of A-ring of glucocorticoids to produce 5α-dihydro-derivatives by 5α-reductases has been considered as a pathway of irreversible inactivation. However, 5α-reduced metabolites of corticosterone and testosterone have significant biological activity. In this paper, we investigated whether toad testicular 5α-reductase (5α-Red) is able to transform corticosterone into 5α-dihydrocorticosterone. Furthermore, we studied the role of 5α-reduced metabolite of corticosterone as a glucocorticoid receptor (GR) agonist. The activity of 5α-Red was assayed in subcellular fractions with [(3)H]corticosterone or [(3)H]testosterone as substrate. The enzyme localizes in microsomes and its optimal pH is between 7 and 8. The activity is not inhibited by finasteride. These results support the conclusion that toad 5α-Red resembles mammalian type 1 isoenzyme. Kinetic studies indicate that neither K(m) nor V(max) for both corticosterone and testosterone were significantly different among reproductive periods. The K(m) value for testosterone was significantly higher than that for corticosterone, indicating that the C-21 steroid is the preferred substrate for the enzyme. Studies of the binding capacity of 5α-dihydrocorticosterone (5α-DHB) to the testicular GR show that 5α-DHB is able to displace the binding of [(3)H]dexamethasone to testicular cytosol with a similar potency than corticosterone. The inhibition constant (Ki) values for corticosterone and 5α-DHB were similar, 31.33±2.9 nM and 35.24±2.3 nM, respectively. In vitro experiments suggest that 5α-DHB is an agonist of toad testicular GR, decreasing the activity of the key enzyme for androgen synthesis, the cytochrome P450 17-hydroxylase, C17,20-lyase.
Collapse
Affiliation(s)
- Amelia J Tesone
- Laboratorio de Endocrinología Comparada, Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | | | | | | |
Collapse
|
45
|
Teleost fish larvae adapt to dietary arachidonic acid supply through modulation of the expression of lipid metabolism and stress response genes. Br J Nutr 2011; 108:864-74. [PMID: 22172151 DOI: 10.1017/s0007114511006143] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Dietary fatty acid supply can affect stress response in fish during early development. Although knowledge on the mechanisms involved in fatty acid regulation of stress tolerance is scarce, it has often been hypothesised that eicosanoid profiles can influence cortisol production. Genomic cortisol actions are mediated by cytosolic receptors which may respond to cellular fatty acid signalling. An experiment was designed to test the effects of feeding gilthead sea-bream larvae with four microdiets, containing graded arachidonic acid (ARA) levels (0·4, 0·8, 1·5 and 3·0 %), on the expression of genes involved in stress response (steroidogenic acute regulatory protein, glucocorticoid receptor and phosphoenolpyruvate carboxykinase), lipid and, particularly, eicosanoid metabolism (hormone-sensitive lipase, PPARα, phospholipase A2, cyclo-oxygenase-2 and 5-lipoxygenase), as determined by real-time quantitative PCR. Fish fatty acid phenotypes reflected dietary fatty acid profiles. Growth performance, survival after acute stress and similar whole-body basal cortisol levels suggested that sea-bream larvae could tolerate a wide range of dietary ARA levels. Transcription of all genes analysed was significantly reduced at dietary ARA levels above 0·4 %. Nonetheless, despite practical suppression of phospholipase A2 transcription, higher leukotriene B4 levels were detected in larvae fed 3·0 % ARA, whereas a similar trend was observed regarding PGE2 production. The present study demonstrates that adaptation to a wide range of dietary ARA levels in gilthead sea-bream larvae involves the modulation of the expression of genes related to eicosanoid synthesis, lipid metabolism and stress response. The roles of ARA, other polyunsaturates and eicosanoids as signals in this process are discussed.
Collapse
|
46
|
Dorey R, Piérard C, Shinkaruk S, Tronche C, Chauveau F, Baudonnat M, Béracochéa D. Membrane mineralocorticoid but not glucocorticoid receptors of the dorsal hippocampus mediate the rapid effects of corticosterone on memory retrieval. Neuropsychopharmacology 2011; 36:2639-49. [PMID: 21814189 PMCID: PMC3230488 DOI: 10.1038/npp.2011.152] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
This study was aimed at determining the type of the glucocorticoid membrane receptors (mineralocorticoid receptors (MRs) or glucocorticoid receptors (GRs)) in the dorsal hippocampus (dHPC) involved in the rapid effects of corticosterone or stress on memory retrieval. For that purpose, we synthesized corticosterone-3-O-carboxymethyloxime-bovine serum albumin conjugate (Cort-3CMO-BSA) conjugate (a high MW complex that cannot cross the cell membrane) totally devoid of free corticosterone, stable in physiological conditions. In a first experiment, we evidenced that an acute stress (electric footshocks) induced both a dHPC corticosterone rise measured by microdialysis and memory retrieval impairment on delayed alternation task. Both the endocrinal and cognitive effects of stress were blocked by metyrapone (a corticosterone synthesis inhibitor). In a second experiment, we showed that bilateral injections of either corticosterone or Cort-3CMO-BSA in dHPC 15 min before memory testing produced impairments similar to those resulting from acute stress. Furthermore, we showed that anisomycin (a protein synthesis inhibitor) failed to block the deleterious effect of Cort-3CMO-BSA on memory. In a third experiment, we evidenced that intra-hippocampal injection of RU-28318 (MR antagonist) but not of RU-38486 (GR antagonist) totally blocked the Cort-3CMO-BSA-induced memory retrieval deficit. In a fourth experiment, we demonstrated that RU-28318 administered 15 min before stress blocked the stress-induced memory impairments when behavioral testing occurred 15 min but not 60 min after stress. Overall, this study provides strong in vivo evidence that the dHPC membrane GRs, mediating the rapid and non-genomic effects of acute stress on memory retrieval, are of MR but not GR type.
Collapse
Affiliation(s)
- Rodolphe Dorey
- CNRS, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA) UMR 5287, Universités de Bordeaux, Talence, France,Institut de Recherche Biomédicale des Armées (IRBA)—Unité Fatigue et Vigilance, Brétigny sur Orge, France
| | - Christophe Piérard
- Institut de Recherche Biomédicale des Armées (IRBA)—Unité Fatigue et Vigilance, Brétigny sur Orge, France
| | - Svitlana Shinkaruk
- U862 Inserm Physiopathologie de la plasticité neuronale, Institut François Magendie, Universités de Bordeaux, Bordeaux, France,ENITA de Bordeaux, Gradignan, France
| | - Christophe Tronche
- CNRS, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA) UMR 5287, Universités de Bordeaux, Talence, France,Institut de Recherche Biomédicale des Armées (IRBA)—Unité Fatigue et Vigilance, Brétigny sur Orge, France
| | - Frédéric Chauveau
- Institut de Recherche Biomédicale des Armées (IRBA)—Unité Fatigue et Vigilance, Brétigny sur Orge, France
| | - Mathieu Baudonnat
- CNRS, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA) UMR 5287, Universités de Bordeaux, Talence, France
| | - Daniel Béracochéa
- CNRS, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA) UMR 5287, Universités de Bordeaux, Talence, France,CNRS, Institut de Neurosciences Cognitives et Integratives d'Aquitaine (INCIA) UMR 5287, Universités de Bordeaux, Talence 33400, France. Tel: +33 54 000 2439; Fax: +33 54 000 8743; E-mail:
| |
Collapse
|
47
|
Harris BN, Perea-Rodriguez JP, Saltzman W. Acute effects of corticosterone injection on paternal behavior in California mouse (Peromyscus californicus) fathers. Horm Behav 2011; 60:666-75. [PMID: 21939660 DOI: 10.1016/j.yhbeh.2011.09.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 09/03/2011] [Accepted: 09/06/2011] [Indexed: 10/17/2022]
Abstract
Glucocorticoids are thought to mediate the disruption of parental behavior in response to acute and chronic stress. Previous research supports their role in chronic stress; however, no study has experimentally tested the effects of acute glucocorticoid elevation on paternal behavior. We tested the prediction that acute corticosterone (CORT) increases would decrease paternal behavior in California mouse fathers and would lead to longer-term effects on reproductive success, as even short-term increases in CORT have been shown to produce lasting effects on the hypothalamic-pituitary-adrenal axis. First-time fathers were injected with 30 mg/kg CORT, 60 mg/kg CORT or vehicle, or left unmanipulated. Interactions between the male and its pup(s) were recorded 1.5-2h after injection and scored for paternal and non-paternal behavior. Treatment groups were combined into control (unmanipulated + vehicle, n = 15) and CORT (30 mg/kg + 60 mg/kg, n = 16) for analysis based on resulting plasma CORT concentrations. CORT treatment did not alter paternal or non-paternal behaviors or any long-term measures (male body mass or temperature, pup growth rate, pup survival, interbirth interval, number or mass of pups born in the second litter). Fathers showed a significant rise in body mass at day 30 postpartum, followed by a decrease in body mass after the birth of the second litter; however, this pattern did not differ between the CORT and control groups. In summary, acute elevation of plasma CORT did not alter direct paternal behavior, body mass, or reproductive outcomes, suggesting that acute CORT elevation alone does not overtly disrupt paternal care in this biparental mammal.
Collapse
Affiliation(s)
- Breanna N Harris
- Department of Biology, University of California, Riverside, CA 92521, USA.
| | | | | |
Collapse
|
48
|
Prevoo B, Miller DS, van de Water FM, Wever KE, Russel FGM, Flik G, Masereeuw R. Rapid, nongenomic stimulation of multidrug resistance protein 2 (Mrp2) activity by glucocorticoids in renal proximal tubule. J Pharmacol Exp Ther 2011; 338:362-71. [PMID: 21515814 PMCID: PMC3126637 DOI: 10.1124/jpet.111.179689] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Accepted: 04/21/2011] [Indexed: 12/21/2022] Open
Abstract
In renal proximal tubule, multidrug resistance protein 2 (Mrp2) actively transports many organic anions into urine, including drugs and metabolic wastes. Upon exposure to nephrotoxicants or during endotoxemia, both Mrp2 activity and expression are up-regulated. This may result from induced de novo synthesis of Mrp2 or post-transcriptional events involving specific signaling pathways. Here, we investigated glucocorticoid signaling to Mrp2 in killifish renal proximal tubules, a model system in which transport activity can be measured using a fluorescent substrate and confocal imaging. Exposure of tubules to dexamethasone rapidly increased Mrp2-mediated fluorescein methotrexate transport. Other glucocorticoid receptor (GR) ligands, cortisol and triamcinolone acetonide, also stimulated Mrp2-mediated transport. The GR antagonist, mifepristone 17β-hydroxy-11β-[4-dimethylamino phenyl]-17α-[1-propynyl]estra-4,9-dien-3-one (RU486), abolished stimulation by all three ligands, whereas the mineralocorticoid receptor antagonist, spironolactone, was ineffective. Consistent with action through a nongenomic mechanism, dexamethasone stimulation of Mrp2-mediated transport was insensitive to cycloheximide and actinomycin D, and immunohistochemistry revealed no alterations in Mrp2 expression at the luminal membrane. (9S-(9α,10β,12α))-2,3,9,10,11,12-hexahydro-10-hydroxy-10-(methoxycarbonyl)-9-methyl-9,12-epoxy-1H-diindolo[1,2,3-fg:3',2',1'-kl]pyrrolo[3,4-i][1,6]benzodiazocin-1-one (K252a), an inhibitor of the tyrosine receptor kinase subfamily, reduced the dexamethasone effect, as did the specific hepatocyte growth factor receptor (c-Met) tyrosine kinase inhibitor, (2R)-1-[[5-[(Z)-[5-[[(2,6-dichlorophenyl)methyl]sulfonyl]-1,2-dihydro-2-oxo-3H-indol-3-ylidene]methyl]-2,4-dimethyl-1H-pyrrol-3-yl]carbonyl]-2-(1-pyrrolidinylmethyl)pyrrolidine (PHA-665752). Hepatocyte growth factor (HGF), an endogenous ligand for c-Met, stimulated Mrp2-mediated transport. This effect was reversed by PHA-665752 but not by RU486. Inhibition of mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (MEK 1/2) also abolished the effects of dexamethasone and HGF. Our results disclose a novel mechanism by which glucocorticoids acting through GR, c-Met, and MEK1/2 cause rapid, nongenomic stimulation of Mrp2-mediated transport in renal proximal tubules. This up-regulation may be nephroprotective, enhancing efflux of metabolic wastes and toxicants during cell and tissue stress.
Collapse
Affiliation(s)
- Brigitte Prevoo
- Department of Pharmacology and Toxicology, Radboud University Nijmegen Medical Centre/Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
49
|
Zhou J, Li M, Sheng CQ, Liu L, Li Z, Wang Y, Zhou JR, Jing ZP, Chen YZ, Jiang CL. A novel strategy for development of glucocorticoids through non-genomic mechanism. Cell Mol Life Sci 2011; 68:1405-14. [PMID: 20853130 PMCID: PMC11114530 DOI: 10.1007/s00018-010-0526-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 07/28/2010] [Accepted: 08/31/2010] [Indexed: 12/21/2022]
Abstract
Glucocorticoids (GCs) are routinely believed to take effect through genomic mechanisms, which are also largely responsible for GCs' side effects. Beneficial non-genomic effects of GCs have been reported as being independent of the genomic pathway. Here, we synthesized a new type of GCs, which took effect mainly via non-genomic mechanisms. Hydrocortisone was conjugated with glycine, lysine and phenylalanine to get a bigger molecular structure, which could hardly go through the cell membrane. Evaluation of the anti-inflammatory efficacy showed that hydrocortisone-conjugated glycine (HG) and lysine could inhibit neutrophil degranulation within 15 min. HG could inhibit IgE-mediated histamine release from mast cells via a non-genomic pathway, and rapidly alleviate allergic reaction. Luciferase reporter assay showed that HG would not activate the glucocorticoid response element within 30 min, which verified the rapid effects independent of the genomic pathway. The work proposes a novel insight into the development of novel GCs, and provides new tools for experimental study on non-genomic mechanisms.
Collapse
Affiliation(s)
- Jian Zhou
- Laboratory of Stress Medicine, Department of Nautical Medicine, Second Military Medical University, Shanghai, 200433 People’s Republic of China
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433 People’s Republic of China
| | - Min Li
- Laboratory of Stress Medicine, Department of Nautical Medicine, Second Military Medical University, Shanghai, 200433 People’s Republic of China
| | - Chun-Quan Sheng
- Department of Medicinal Chemistry, College of Pharmacy, Second Military Medical University, Shanghai, 200433 People’s Republic of China
| | - Lei Liu
- Laboratory of Stress Medicine, Department of Nautical Medicine, Second Military Medical University, Shanghai, 200433 People’s Republic of China
| | - Zhen Li
- Department of Clinical Pharmacology, Changhai Hospital, Second Military Medical University, Shanghai, 200433 People’s Republic of China
| | - Yan Wang
- Department of Pathophysiology, Second Military Medical University, Shanghai, 200433 People’s Republic of China
| | - Jiang-Rui Zhou
- Laboratory of Stress Medicine, Department of Nautical Medicine, Second Military Medical University, Shanghai, 200433 People’s Republic of China
| | - Zai-Ping Jing
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433 People’s Republic of China
| | - Yi-Zhang Chen
- Institute of Neuroscience, Second Military Medical University, Shanghai, 200433 People’s Republic of China
| | - Chun-Lei Jiang
- Laboratory of Stress Medicine, Department of Nautical Medicine, Second Military Medical University, Shanghai, 200433 People’s Republic of China
| |
Collapse
|
50
|
Tabuchi K, Hara A. [Glucocorticoid treatment for cochlear ischemic and acoustic injuries]. NIHON JIBIINKOKA GAKKAI KAIHO 2010; 113:831-837. [PMID: 21174729 DOI: 10.3950/jibiinkoka.113.831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
The effect of glucocorticoids on sensorineural hearing loss of sudden onset remains to be controversial although glucocorticoids have been used for treatment of sudden sensorineural hearing loss. We review recent findings about the effect of glucocorticoids on cochlear ischemic and acoustic injuries obtained from animal experiments. Systemically administered glucocorticoids penetrate the blood-cochlear barrier well. Glucocorticoids ameliorated the cochlear ischemic and acoustic injuries at a relatively wide range of doses, and they protect cochlear hair cells in these types of injury. The therapeutic actions of glucocorticoids in cochlear injuries were considered to be mediated via both genomic and non-genomic pathways. Based on the results obtained in acoustic injury, therapeutic time window of glucocorticoids is considered to be short after the onset of injury. These findings obtained from animal experiments are important in considering clinical usage of glucocorticoids for the treatment of sensorineural hearing loss.
Collapse
Affiliation(s)
- Keiji Tabuchi
- Department of Otolaryngology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba
| | | |
Collapse
|