151
|
Ikeda M, Hojo Y, Komatsuzaki Y, Okamoto M, Kato A, Takeda T, Kawato S. Hippocampal spine changes across the sleep-wake cycle: corticosterone and kinases. J Endocrinol 2015; 226:M13-27. [PMID: 26034071 DOI: 10.1530/joe-15-0078] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/01/2015] [Indexed: 12/22/2022]
Abstract
The corticosterone (CORT) level changes along the circadian rhythm. Hippocampus is sensitive to CORT, since glucocorticoid receptors are highly expressed. In rat hippocampus fixed in a living state every 3 h, we found that the dendritic spine density of CA1 pyramidal neurons increased upon waking (within 3 h), as compared with the spine density in the sleep state. Particularly, the large-head spines increased. The observed change in the spine density may be due to the change in the hippocampal CORT level, since the CORT level at awake state (∼30 nM) in cerebrospinal fluid was higher than that at sleep state (∼3 nM), as observed from our earlier study. In adrenalectomized (ADX) rats, such a wake-induced increase of the spine density disappeared. S.c. administration of CORT into ADX rats rescued the decreased spine density. By using isolated hippocampal slices, we found that the application of 30 nM CORT increased the spine density within 1 h and that the spine increase was mediated via PKA, PKC, ERK MAPK, and LIMK signaling pathways. These findings suggest that the moderately rapid increase of the spine density on waking might mainly be caused by the CORT-driven kinase networks.
Collapse
Affiliation(s)
- Muneki Ikeda
- Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan
| | - Yasushi Hojo
- Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan
| | - Yoshimasa Komatsuzaki
- Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan
| | - Masahiro Okamoto
- Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan
| | - Asami Kato
- Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan
| | - Taishi Takeda
- Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan
| | - Suguru Kawato
- Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan Department of Biophysics and Life SciencesGraduate School of Arts and Sciences, University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 152-8902, JapanBioinformatics Project of Japan Science and Technology AgencyUniversity of Tokyo, Tokyo, JapanLaboratory of Exercise Biochemistry and NeuroendocrinologyFaculty of Health and Sports Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, JapanDepartment of UrologyGraduate School of Medicine, Juntendo University, 2-1-1 Hongo, Tokyo 113-8424, Japan
| |
Collapse
|
152
|
Sandi C, Haller J. Stress and the social brain: behavioural effects and neurobiological mechanisms. Nat Rev Neurosci 2015; 16:290-304. [PMID: 25891510 DOI: 10.1038/nrn3918] [Citation(s) in RCA: 400] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Stress often affects our social lives. When undergoing high-level or persistent stress, individuals frequently retract from social interactions and become irritable and hostile. Predisposition to antisocial behaviours - including social detachment and violence - is also modulated by early life adversity; however, the effects of early life stress depend on the timing of exposure and genetic factors. Research in animals and humans has revealed some of the structural, functional and molecular changes in the brain that underlie the effects of stress on social behaviour. Findings in this emerging field will have implications both for the clinic and for society.
Collapse
Affiliation(s)
- Carmen Sandi
- Brain Mind Institute, School of Life Sciences, École Polytechnique Federale de Lausanne (EPFL), Lausanne CH-1050, Switzerland
| | - József Haller
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1450, Hungary
| |
Collapse
|
153
|
Colciago A, Casati L, Negri-Cesi P, Celotti F. Learning and memory: Steroids and epigenetics. J Steroid Biochem Mol Biol 2015; 150:64-85. [PMID: 25766520 DOI: 10.1016/j.jsbmb.2015.02.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 02/11/2015] [Accepted: 02/12/2015] [Indexed: 12/19/2022]
Abstract
Memory formation and utilization is a complex process involving several brain structures in conjunction as the hippocampus, the amygdala and the adjacent cortical areas, usually defined as medial temporal lobe structures (MTL). The memory processes depend on the formation and modulation of synaptic connectivity affecting synaptic strength, synaptic plasticity and synaptic consolidation. The basic neurocognitive mechanisms of learning and memory are shortly recalled in the initial section of this paper. The effect of sex hormones (estrogens, androgens and progesterone) and of adrenocortical steroids on several aspects of memory processes are then analyzed on the basis of animal and human studies. A specific attention has been devoted to the different types of steroid receptors (membrane or nuclear) involved and on local metabolic transformations when required. The review is concluded by a short excursus on the steroid activated epigenetic mechanisms involved in memory formation.
Collapse
Affiliation(s)
- Alessandra Colciago
- Department of Pharmacological and Biomolecular Sciences, Section of Biomedicine and Endocrinology, Via Balzaretti 9, 20133 Milano, Italy
| | - Lavinia Casati
- Department of Medical Biotechnologies and Translational Medicine, Via Vanvitelli 32, 20129 Milano, Italy
| | - Paola Negri-Cesi
- Department of Pharmacological and Biomolecular Sciences, Section of Biomedicine and Endocrinology, Via Balzaretti 9, 20133 Milano, Italy
| | - Fabio Celotti
- Department of Pharmacological and Biomolecular Sciences, Section of Biomedicine and Endocrinology, Via Balzaretti 9, 20133 Milano, Italy
| |
Collapse
|
154
|
Effects of mineralocorticoid receptor stimulation via fludrocortisone on memory in women with borderline personality disorder. Neurobiol Learn Mem 2015; 120:94-100. [DOI: 10.1016/j.nlm.2015.02.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 02/09/2015] [Accepted: 02/21/2015] [Indexed: 01/11/2023]
|
155
|
ter Heegde F, De Rijk RH, Vinkers CH. The brain mineralocorticoid receptor and stress resilience. Psychoneuroendocrinology 2015; 52:92-110. [PMID: 25459896 DOI: 10.1016/j.psyneuen.2014.10.022] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 10/23/2014] [Accepted: 10/27/2014] [Indexed: 12/11/2022]
Abstract
Stress exposure activates the HPA-axis and results in the release of corticosteroids which bind to two receptor types in the brain: the mineralocorticoid receptor (MR) and the glucocorticoid receptor (GR). While the role of the GR in stress reactivity has been extensively studied, the MR has received less attention. Nevertheless, pioneering in-depth studies over the past two decades have shown the importance of the brain MR in the processing of stressful information. Moreover, a membrane-bound MR mediating the rapid effects of cortisol was recently discovered. This review summarizes how the MR may play a role in stress resilience. Both preclinical and clinical studies suggest that the MR is an important stress modulator and influences basal as well as stress-induced HPA-axis activity, stress appraisal, and fear-related memories. These MR effects are mediated by both genomic and non-genomic MRs and appear to be at least partially sex-dependent. Moreover, the majority of studies indicate that high MR functionality or expression may confer resilience to traumatic stress. This has direct clinical implications. First, increasing activity or expression of brain MRs may prevent or reverse symptoms of stress-related depression. Second, individuals with a relatively low MR functionality may possess an increased stress susceptibility for depression. Nevertheless, the number of clinical MR studies is currently limited. In conclusion, the recent emergence of the MR as a putative stress resilience factor is important and may open up new avenues for the prevention and treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Freija ter Heegde
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roel H De Rijk
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands; Department of Clinical Psychology, Leiden, The Netherlands
| | - Christiaan H Vinkers
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
156
|
Bohacek J, Manuella F, Roszkowski M, Mansuy IM. Hippocampal gene expression induced by cold swim stress depends on sex and handling. Psychoneuroendocrinology 2015; 52:1-12. [PMID: 25459888 DOI: 10.1016/j.psyneuen.2014.10.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/03/2014] [Accepted: 10/31/2014] [Indexed: 01/03/2023]
Abstract
Stress-related disorders such as PTSD and depression are more prevalent in women than men. One reason for such discordance may be that brain regions involved in stress responses are more sensitive to stress in females. Here, we compared the effects of acute stress on gene transcription in the hippocampus of female and male mice, and also examined the involvement of two key stress-related hormones, corticosterone and corticotropin releasing hormone (Crh). Using quantitative reverse transcription polymerase chain reaction (RT-qPCR), we measured gene expression of Fos, Per1 and Sgk1 45 min after exposure to brief cold swim stress. Stress induced a stronger increase in Fos and Per1 expression in females than males. The handling control procedure increased Fos in both sexes, but occluded the effects of stress in males. Further, handling increased Per1 only in males. Sgk1 was insensitive to handling, and increased in response to stress similarly in males and females. The transcriptional changes observed after swim stress were not mimicked by corticosterone injections, and the stress-induced increase in Fos, Per1 and Sgk1 could neither be prevented by pharmacologically blocking glucocorticoid receptor (GR) nor by blocking Crh receptor 1 (Crhr1) before stress exposure. Finally, we demonstrate that the effects are stressor-specific, as the expression of target genes could not be increased by brief restraint stress in either sex. In summary, we find strong effects of acute swim stress on hippocampal gene expression, complex interactions between handling and sex, and a remarkably unique response pattern for each gene. Overall, females respond to a cold swim challenge with stronger hippocampal gene transcription than males, independent of two classic mediators of the stress response, corticosterone and Crh. These findings may have important implications for understanding the higher vulnerability of women to certain stress-related neuropsychiatric diseases.
Collapse
Affiliation(s)
- Johannes Bohacek
- Brain Research Institute, Neuroscience Center Zürich, University of Zurich/ETH Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| | - Francesca Manuella
- Brain Research Institute, Neuroscience Center Zürich, University of Zurich/ETH Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Martin Roszkowski
- Brain Research Institute, Neuroscience Center Zürich, University of Zurich/ETH Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Isabelle M Mansuy
- Brain Research Institute, Neuroscience Center Zürich, University of Zurich/ETH Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
157
|
Ventral hippocampal nicotinic acetylcholine receptors mediate stress-induced analgesia in mice. Prog Neuropsychopharmacol Biol Psychiatry 2015; 56:235-42. [PMID: 25281932 DOI: 10.1016/j.pnpbp.2014.09.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 09/10/2014] [Accepted: 09/10/2014] [Indexed: 12/21/2022]
Abstract
Evidence suggests that various stressful procedures induce an analgesic effect in laboratory animals commonly referred to as stress-induced analgesia (SIA). The aim of the present study was to assess the role of ventral hippocampal (VH) nicotinic acetylcholine receptors (nAChRs) in SIA in adult male NMRI mice. The VHs of animals were bilaterally cannulated and nociceptive threshold was measured using infrared source in a tail-flick apparatus. Acute stress was evoked by placing the animals on an elevated platform for 10, 20 and 30 min. The results showed that exposure to 20 and 30 min acute stress produced analgesia, while exposure to 10 min stress had no effect on the pain response. Intra-VH microinjection of nicotine (0.001-0.1 μg/mouse), 5 min before an ineffective stress (10 min stress), induced analgesia, suggesting the potentiative effect of nicotine on SIA. It is important to note that bilateral intra-VH microinjections of the same doses of nicotine without stress had no effect on the tail-flick test. On the other hand, intra-VH microinjection of mecamylamine (0.5-1 μg/mouse) 5 min before 20-min stress inhibited SIA. However, bilateral intra-VH microinjections of the same doses of mecamylamine without stress had no effect on the tail-flick response. In addition, the microinjection of mecamylamine into the VH reversed the potentiative effect of nicotine on SIA. Taken together, it can be concluded that exposure to acute stress induces SIA in a time-dependent manner and the ventral hippocampal cholinergic system may be involved in SIA via nAChRs.
Collapse
|
158
|
Laryea G, Muglia L, Arnett M, Muglia LJ. Dissection of glucocorticoid receptor-mediated inhibition of the hypothalamic-pituitary-adrenal axis by gene targeting in mice. Front Neuroendocrinol 2015; 36:150-64. [PMID: 25256348 PMCID: PMC4342273 DOI: 10.1016/j.yfrne.2014.09.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 08/05/2014] [Accepted: 09/11/2014] [Indexed: 12/17/2022]
Abstract
Negative feedback regulation of glucocorticoid (GC) synthesis and secretion occurs through the function of glucocorticoid receptor (GR) at sites in the hypothalamic-pituitary-adrenal (HPA) axis, as well as in brain regions such as the hippocampus, prefrontal cortex, and sympathetic nervous system. This function of GRs in negative feedback coordinates basal glucocorticoid secretion and stress-induced increases in secretion that integrate GC production with the magnitude and duration of the stressor. This review describes the effects of GR loss along major sites of negative feedback including the entire brain, the paraventricular nucleus of the hypothalamus (PVN), and the pituitary. In genetic mouse models, we evaluate circadian regulation of the HPA axis, stress-stimulated neuroendocrine response and behavioral activity, as well as the integrated response of organism metabolism. Our analysis provides information on contributions of region-specific GR-mediated negative feedback to provide insight in understanding HPA axis dysregulation and the pathogenesis of psychiatric and metabolic disorders.
Collapse
Affiliation(s)
- Gloria Laryea
- Neuroscience Graduate Program, School of Medicine, Vanderbilt University, Nashville, TN, United States; Center for Preterm Birth Research, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, ML 7009, Cincinnati, OH 45229, United States.
| | - Lisa Muglia
- Center for Preterm Birth Research, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, ML 7009, Cincinnati, OH 45229, United States.
| | - Melinda Arnett
- Center for Preterm Birth Research, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, ML 7009, Cincinnati, OH 45229, United States.
| | - Louis J Muglia
- Center for Preterm Birth Research, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, ML 7009, Cincinnati, OH 45229, United States; Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, ML 7009, Cincinnati, OH 45229, United States.
| |
Collapse
|
159
|
Reul JM, Collins A, Saliba RS, Mifsud KR, Carter SD, Gutierrez-Mecinas M, Qian X, Linthorst AC. Glucocorticoids, epigenetic control and stress resilience. Neurobiol Stress 2015; 1:44-59. [PMID: 27589660 PMCID: PMC4721318 DOI: 10.1016/j.ynstr.2014.10.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 10/01/2014] [Accepted: 10/04/2014] [Indexed: 11/25/2022] Open
Abstract
Glucocorticoid hormones play a pivotal role in the response to stressful challenges. The surge in glucocorticoid hormone secretion after stress needs to be tightly controlled with characteristics like peak height, curvature and duration depending on the nature and severity of the challenge. This is important as chronic hyper- or hypo-responses are detrimental to health due to increasing the risk for developing a stress-related mental disorder. Proper glucocorticoid responses to stress are critical for adaptation. Therefore, the tight control of baseline and stress-evoked glucocorticoid secretion are important constituents of an organism's resilience. Here, we address a number of mechanisms that illustrate the multitude and complexity of measures safeguarding the control of glucocorticoid function. These mechanisms include the control of mineralocorticoid (MR) and glucocorticoid receptor (GR) occupancy and concentration, the dynamic control of free glucocorticoid hormone availability by corticosteroid-binding globulin (CBG), and the control exerted by glucocorticoids at the signaling, epigenetic and genomic level on gene transcriptional responses to stress. We review the beneficial effects of regular exercise on HPA axis and sleep physiology, and cognitive and anxiety-related behavior. Furthermore, we describe that, possibly through changes in the GABAergic system, exercise reduces the impact of stress on a signaling pathway specifically in the dentate gyrus that is strongly implicated in the behavioral response to that stressor. These observations underline the impact of life style on stress resilience. Finally, we address how single nucleotide polymorphisms (SNPs) affecting glucocorticoid action can compromise stress resilience, which becomes most apparent under conditions of childhood abuse.
Collapse
Affiliation(s)
- Johannes M.H.M. Reul
- Neuro-Epigenetics Research Group, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, United Kingdom
| | - Andrew Collins
- Neuro-Epigenetics Research Group, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, United Kingdom
| | - Richard S. Saliba
- Neuro-Epigenetics Research Group, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, United Kingdom
| | - Karen R. Mifsud
- Neuro-Epigenetics Research Group, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, United Kingdom
| | - Sylvia D. Carter
- Neuro-Epigenetics Research Group, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, United Kingdom
| | - Maria Gutierrez-Mecinas
- Neuro-Epigenetics Research Group, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, United Kingdom
| | - Xiaoxiao Qian
- Neurobiology of Stress and Behaviour Research Group, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, United Kingdom
| | - Astrid C.E. Linthorst
- Neurobiology of Stress and Behaviour Research Group, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, United Kingdom
| |
Collapse
|
160
|
Gomez-Sanchez EP. Brain mineralocorticoid receptors in cognition and cardiovascular homeostasis. Steroids 2014; 91:20-31. [PMID: 25173821 PMCID: PMC4302001 DOI: 10.1016/j.steroids.2014.08.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 07/10/2014] [Accepted: 08/04/2014] [Indexed: 12/20/2022]
Abstract
Mineralocorticoid receptors (MR) mediate diverse functions supporting osmotic and hemodynamic homeostasis, response to injury and inflammation, and neuronal changes required for learning and memory. Inappropriate MR activation in kidneys, heart, vessels, and brain hemodynamic control centers results in cardiovascular and renal pathology and hypertension. MR binds aldosterone, cortisol and corticosterone with similar affinity, while the glucocorticoid receptor (GR) has less affinity for cortisol and corticosterone. As glucocorticoids are more abundant than aldosterone, aldosterone activates MR in cells co-expressing enzymes with 11β-hydroxydehydrogenase activity to inactivate them. MR and GR co-expressed in the same cell interact at the molecular and functional level and these functions may be complementary or opposing depending on the cell type. Thus the balance between MR and GR expression and activation is crucial for normal function. Where 11β-hydroxydehydrogenase 2 (11β-HSD2) that inactivates cortisol and corticosterone in aldosterone target cells of the kidney and nucleus tractus solitarius (NTS) is not expressed, as in most neurons, MR are activated at basal glucocorticoid concentrations, GR at stress concentrations. An exception may be pre-autonomic neurons of the PVN which express MR and 11β-HSD1 in the absence of hexose-6-phosphate dehydrogenase required to generate the requisite cofactor for reductase activity, thus it acts as a dehydrogenase. MR antagonists, valuable adjuncts to the treatment of cardiovascular disease, also inhibit MR in the brain that are crucial for memory formation and exacerbate detrimental effects of excessive GR activation on cognition and mood. 11β-HSD1 inhibitors combat metabolic and cognitive diseases related to glucocorticoid excess, but may exacerbate MR action where 11β-HSD1 acts as a dehydrogenase, while non-selective 11β-HSD1&2 inhibitors cause injurious disruption of MR hemodynamic control. MR functions in the brain are multifaceted and optimal MR:GR activity is crucial. Therefore selectively targeting down-stream effectors of MR specific actions may be a better therapeutic goal.
Collapse
Affiliation(s)
- Elise P Gomez-Sanchez
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| |
Collapse
|
161
|
Spironolactone decreases the somatic signs of opiate withdrawal by blocking the mineralocorticoid receptors (MR). Toxicology 2014; 326:36-43. [DOI: 10.1016/j.tox.2014.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/03/2014] [Accepted: 10/03/2014] [Indexed: 12/29/2022]
|
162
|
Sase S, Meyer K, Lubec G, Korz V. Different expression of membrane-bound and cytosolic hippocampal steroid receptor complexes during spatial training in young male rats. Eur Neuropsychopharmacol 2014; 24:1819-27. [PMID: 25258178 DOI: 10.1016/j.euroneuro.2014.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/31/2014] [Accepted: 09/03/2014] [Indexed: 10/24/2022]
Abstract
Brain steroid receptors are involved in mediating stress responses and cognitive processes throughfast non-genomic signaling of membrane-bound receptors or through the slower genomic actions of cytosolic receptors. Although the contribution of these different pathways in the formation and maintenance of memories has been widely discussed, little is known about the regulation of membrane versus cytosolic receptors during a learning task. Besides the relatively well studied corticosterone-binding glucocorticoid (GR) and mineralocorticoid (MR) receptors, sex steroid hormone receptors, such as the androgen and estrogen (ERα and ERβ) receptors, have also been shown to be involved in the regulation of stress and cognition. Moreover, the latter receptors are known to be functional in both sexes. Therefore, we studied the expression of hippocampal receptors in both cellular fractions during spatial learning in male rats. Membrane and cytosolic GR were shown to be downregulated after memory acquisition and unaffected after consolidation, whereas membrane MR was upregulated after both learning phases and unaffected in the cytosol. Cytosolic ERα was downregulated after both phases and unaffected in the membrane. The remaining receptors were not regulated. The data suggest a specific role of MR and ERα during training as fast and slow mediators, respectively.
Collapse
Affiliation(s)
- Sunetra Sase
- University of Vienna, Department of Pediatrics, Vienna, Austria
| | - Katrin Meyer
- Leibniz Institute for Neurobiology, Magdeburg, Germany; University of Magdeburg, Institute of Biology, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Gert Lubec
- University of Vienna, Department of Pediatrics, Vienna, Austria
| | - Volker Korz
- University of Magdeburg, Institute of Biology, Leipziger Str. 44, 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany.
| |
Collapse
|
163
|
G protein-coupled receptors: extranuclear mediators for the non-genomic actions of steroids. Int J Mol Sci 2014; 15:15412-25. [PMID: 25257522 PMCID: PMC4200746 DOI: 10.3390/ijms150915412] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 07/26/2014] [Accepted: 08/20/2014] [Indexed: 02/06/2023] Open
Abstract
Steroids hormones possess two distinct actions, a delayed genomic effect and a rapid non-genomic effect. Rapid steroid-triggered signaling is mediated by specific receptors localized most often to the plasma membrane. The nature of these receptors is of great interest and accumulated data suggest that G protein-coupled receptors (GPCRs) are appealing candidates. Increasing evidence regarding the interaction between steroids and specific membrane proteins, as well as the involvement of G protein and corresponding downstream signaling, have led to identification of physiologically relevant GPCRs as steroid extranuclear receptors. Examples include G protein-coupled receptor 30 (GPR30) for estrogen, membrane progestin receptor for progesterone, G protein-coupled receptor family C group 6 member A (GPRC6A) and zinc transporter member 9 (ZIP9) for androgen, and trace amine associated receptor 1 (TAAR1) for thyroid hormone. These receptor-mediated biological effects have been extended to reproductive development, cardiovascular function, neuroendocrinology and cancer pathophysiology. However, although great progress have been achieved, there are still important questions that need to be answered, including the identities of GPCRs responsible for the remaining steroids (e.g., glucocorticoid), the structural basis of steroids and GPCRs' interaction and the integration of extranuclear and nuclear signaling to the final physiological function. Here, we reviewed the several significant developments in this field and highlighted a hypothesis that attempts to explain the general interaction between steroids and GPCRs.
Collapse
|
164
|
Enhanced emotional empathy after mineralocorticoid receptor stimulation in women with borderline personality disorder and healthy women. Neuropsychopharmacology 2014; 39:1799-804. [PMID: 24535100 PMCID: PMC4059897 DOI: 10.1038/npp.2014.36] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 01/10/2014] [Accepted: 02/12/2014] [Indexed: 01/10/2023]
Abstract
The mineralocorticoid receptor (MR) is highly expressed in the hippocampus and prefrontal cortex. MR have an important role in appraisal processes and in modulating stress-associated emotional reactions but it is not known whether the MR affects empathy. Borderline personality disorder (BPD) is characterized by disturbed emotion regulation and alterations in empathy. In the current study, we examined whether stimulation of the MR enhances empathy in patients with BPD and healthy individuals. In a placebo-controlled study, we randomized 38 women with BPD and without psychotropic medication, and 35 healthy women to either placebo or 0.4 mg fludrocortisone, an MR agonist. Subsequently, all participants underwent two tests of social cognition, the Multifaceted Empathy Test (MET) and the Movie for the Assessment of Social Cognition (MASC), measuring cognitive and emotional facets of empathy. Eighteen BPD patients and 18 healthy women received placebo, whereas 20 BPD patients and 17 healthy women received fludrocortisone. In the MET, fludrocortisone enhanced emotional empathy across groups, whereas cognitive empathy was not affected. In the MASC, no effect of fludrocortisone could be revealed. In both tests, BPD patients and healthy women did not differ significantly in cognitive and emotional empathy and in their response to fludrocortisone. Stimulation of MR enhanced emotional empathy in healthy women and in BPD patients. Whether fludrocortisone might have a therapeutic role in psychotherapeutic processes, remains to be elucidated.
Collapse
|
165
|
Abstract
The primary adrenal cortical steroid hormones, aldosterone, and the glucocorticoids cortisol and corticosterone, act through the structurally similar mineralocorticoid (MR) and glucocorticoid receptors (GRs). Aldosterone is crucial for fluid, electrolyte, and hemodynamic homeostasis and tissue repair; the significantly more abundant glucocorticoids are indispensable for energy homeostasis, appropriate responses to stress, and limiting inflammation. Steroid receptors initiate gene transcription for proteins that effect their actions as well as rapid non-genomic effects through classical cell signaling pathways. GR and MR are expressed in many tissues types, often in the same cells, where they interact at molecular and functional levels, at times in synergy, others in opposition. Thus the appropriate balance of MR and GR activation is crucial for homeostasis. MR has the same binding affinity for aldosterone, cortisol, and corticosterone. Glucocorticoids activate MR in most tissues at basal levels and GR at stress levels. Inactivation of cortisol and corticosterone by 11β-HSD2 allows aldosterone to activate MR within aldosterone target cells and limits activation of the GR. Under most conditions, 11β-HSD1 acts as a reductase and activates cortisol/corticosterone, amplifying circulating levels. 11β-HSD1 and MR antagonists mitigate inappropriate activation of MR under conditions of oxidative stress that contributes to the pathophysiology of the cardiometabolic syndrome; however, MR antagonists decrease normal MR/GR functional interactions, a particular concern for neurons mediating cognition, memory, and affect.
Collapse
Affiliation(s)
- Elise Gomez-Sanchez
- G.V.(Sonny) Montgomery V.A. Medical Center and Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Celso E. Gomez-Sanchez
- G.V.(Sonny) Montgomery V.A. Medical Center and Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
166
|
An organoselenium compound improves behavioral, endocrinal and neurochemical changes induced by corticosterone in mice. Psychopharmacology (Berl) 2014; 231:2119-30. [PMID: 24306280 DOI: 10.1007/s00213-013-3361-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 11/14/2013] [Indexed: 12/14/2022]
Abstract
RATIONALE 3-(4-Fluorophenylselenyl)-2,5-diphenylselenophene (F-DPS) is a promising organoselenium compound that shows antidepressant-like properties related to interaction with the serotonergic system. OBJECTIVES In this study, a mouse model of anxiety/depressant-like behavior induced by long-term corticosterone treatment was used to evaluate behavioral, endocrinal, and neurochemical changes in mice and their possible modulation of F-DPS treatment. METHODS Swiss mice were subjected to 4 weeks of corticosterone administration (20 μg/ml in drinking water) and a new therapeutic approach with F-DPS (0.1 mg/kg/day, intragastric route, during 1 week) was employed to modulate changes induced by corticosterone exposure. RESULTS Treatment with corticosterone caused a significant depressant-like behavior in the forced swimming test and tail suspension test, which was accompanied by anxiety-like condition in the light-dark test and novelty suppressed-feeding; similarly to the classical antidepressant drug paroxetine, F-DPS treatment was effective in reversing these behavioral changes. Further, F-DPS normalized serum levels of corticosterone and adrenocorticotropic hormone, which were increased after corticosterone exposure. Corticosterone also significantly inhibited glutamate uptake in the prefrontal cortex of mice, whereas glutamate release was not modified. Besides normalizing glutamate uptake in the corticosterone-exposed mice, F-DPS promoted an inhibition of 5-HT uptake in the prefrontal cortex and hippocampus. In addition, hippocampal monoamine oxidase-A activity was also inhibited by F-DPS treatment. CONCLUSIONS These findings suggest a modulation of both serotonergic and glutamatergic systems by F-DPS after a long-term corticosterone exposure in mice, which may be involved in the antidepressant- and anxiolytic-like actions of this organoselenium compound.
Collapse
|
167
|
Bellavance MA, Rivest S. The HPA - Immune Axis and the Immunomodulatory Actions of Glucocorticoids in the Brain. Front Immunol 2014; 5:136. [PMID: 24744759 PMCID: PMC3978367 DOI: 10.3389/fimmu.2014.00136] [Citation(s) in RCA: 287] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 03/18/2014] [Indexed: 12/20/2022] Open
Abstract
In response to physiological and psychogenic stressors, the hypothalamic–pituitary–adrenal (HPA) axis orchestrates the systemic release of glucocorticoids (GCs). By virtue of nearly ubiquitous expression of the GC receptor and the multifaceted metabolic, cardiovascular, cognitive, and immunologic functions of GCs, this system plays an essential role in the response to stress and restoration of an homeostatic state. GCs act on almost all types of immune cells and were long recognized to perform salient immunosuppressive and anti-inflammatory functions through various genomic and non-genomic mechanisms. These renowned effects of the steroid hormone have been exploited in the clinic for the past 70 years and synthetic GC derivatives are commonly used for the therapy of various allergic, autoimmune, inflammatory, and hematological disorders. The role of the HPA axis and GCs in restraining immune responses across the organism is however still debated in light of accumulating evidence suggesting that GCs can also have both permissive and stimulatory effects on the immune system under specific conditions. Such paradoxical actions of GCs are particularly evident in the brain, where substantial data support either a beneficial or detrimental role of the steroid hormone. In this review, we examine the roles of GCs on the innate immune system with a particular focus on the CNS compartment. We also dissect the numerous molecular mechanisms through which GCs exert their effects and discuss the various parameters influencing the paradoxical immunomodulatory functions of GCs in the brain.
Collapse
Affiliation(s)
- Marc-André Bellavance
- Faculty of medicine, Department of Molecular Medicine, Neuroscience Laboratory, CHU de Québec Research Center, Laval University , Québec, QC , Canada
| | - Serge Rivest
- Faculty of medicine, Department of Molecular Medicine, Neuroscience Laboratory, CHU de Québec Research Center, Laval University , Québec, QC , Canada
| |
Collapse
|
168
|
Nogami H, Yamamoto N, Hiraoka Y, Aiso S, Sugimoto K, Yoshida S, Shutoh F, Hisano S. Rapid induction of the growth hormone gene transcription by glucocorticoids in vitro: possible involvement of membrane glucocorticoid receptors and phosphatidylinositol 3-kinase activation. J Neuroendocrinol 2014; 26:195-204. [PMID: 24428719 DOI: 10.1111/jne.12132] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 12/19/2013] [Accepted: 01/07/2014] [Indexed: 12/25/2022]
Abstract
The regulation of transcription of the growth hormone (GH) gene by glucocorticoids was studied in MtT/S cells, a cell line derived from an oestrogen-induced mammotrophic tumour in the rat, and in the primary culture of the anterior pituitary gland of adult mice. The levels of the GH heteronuclear RNA (GH hnRNA), which are mainly determined by the transcription rate, increased by 25-fold during 24 h in response to dexamethasone (DEX; 1 μM) in MtT/S cells that were cultured in the medium containing charcoal-stripped serum for 7 days. The stimulatory effect of DEX on the GH hnRNA levels was detectable as early as 30 min. This rapid effect of DEX did not require on-going protein synthesis, whereas it was considered that DEX requires the presence of unknown cellular proteins produced independently of DEX stimulation. By contrast, on-going protein synthesis was required for DEX action when incubated for 6 h, as has been observed in the previous studies. The specific inhibitor of glucocorticoid receptor, RU486, inhibited both rapid (30 min) and delayed (6 h) the effects of glucocorticoids on GH hnRNA levels. Membrane impermeable glucocorticoid, corticosterone-bovine serum albumin conjugate (CSBSA), was found to have effects similar to those of DEX and free corticosterone (CS), suggesting that glucocorticoids regulate GH gene transcription at least in part through the membrane bound receptors. From pharmacological studies, it was suggested that phosphatidylinositol 3-kinase (PI3K) activation is involved in the rapid effects but not in the delayed effects of glucocorticoids. This also suggests that the delayed effects of glucocorticoids depend on mechanisms other than the activation of PI3-kinase. Finally, both rapid and delayed effects of CS and CSBSA were observed not only in MtT/S cells, but also in the mouse pituitary cells in primary culture. Therefore, it is possible that the membrane initiated action of glucocorticoids is involved in the regulation of GH transcription in normal pituitary cells, as well as in pituitary tumour cells.
Collapse
Affiliation(s)
- H Nogami
- Laboratry of Neuroendocrinology, Institute of Basic Medical Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | | | | | | | | | | | | | | |
Collapse
|
169
|
Zouikr I, Tadros MA, Barouei J, Beagley KW, Clifton VL, Callister RJ, Hodgson DM. Altered nociceptive, endocrine, and dorsal horn neuron responses in rats following a neonatal immune challenge. Psychoneuroendocrinology 2014; 41:1-12. [PMID: 24495603 DOI: 10.1016/j.psyneuen.2013.11.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 11/18/2013] [Accepted: 11/18/2013] [Indexed: 01/16/2023]
Abstract
The neonatal period is characterized by significant plasticity where the immune, endocrine, and nociceptive systems undergo fine-tuning and maturation. Painful experiences during this period can result in long-term alterations in the neurocircuitry underlying nociception, including increased sensitivity to mechanical or thermal stimuli. Less is known about the impact of neonatal exposure to mild inflammatory stimuli, such as lipopolysaccharide (LPS), on subsequent inflammatory pain responses. Here we examine the impact of neonatal LPS exposure on inflammatory pain sensitivity and HPA axis activity during the first three postnatal weeks. Wistar rats were injected with LPS (0.05mg/kg IP, Salmonella enteritidis) or saline on postnatal days (PNDs) 3 and 5 and later subjected to the formalin test at PNDs 7, 13, and 22. One hour after formalin injection, blood was collected to assess corticosterone responses. Transverse spinal cord slices were also prepared for whole-cell patch clamp recording from lumbar superficial dorsal horn neurons (SDH). Brains were obtained at PND 22 and the hypothalamus was isolated to measure glucocorticoid (GR) and mineralocorticoid receptor (MR) transcript expression using qRT-PCR. Behavioural analyses indicate that at PND 7, no significant differences were observed between saline- or LPS-challenged rats. At PND 13, LPS-challenged rats exhibited enhanced licking (p<.01), and at PND 22, increased flinching in response to formalin injection (p<.05). LPS-challenged rats also displayed increased plasma corticosterone at PND 7 and PND 22 (p<.001) but not at PND 13 following formalin administration. Furthermore, at PND 22 neonatal LPS exposure induced decreased levels of GR mRNA and increased levels of MR mRNA in the hypothalamus. The intrinsic properties of SDH neurons were similar at PND 7 and PND 13. However, at PND 22, ipsilateral SDH neurons in LPS-challenged rats had a lower input resistance compared to their saline-challenged counterparts (p<.05). These data suggest neonatal LPS exposure produces developmentally regulated changes in formalin-induced behavioural responses, corticosterone levels, and dorsal horn neuron properties following noxious stimulation later in life. These findings highlight the importance of immune activation during the neonatal period in shaping pain sensitivity later in life. This programming involves both spinal cord neurons and the HPA axis.
Collapse
Affiliation(s)
- Ihssane Zouikr
- Laboratory of Neuroimmunology, School of Psychology, University of Newcastle, Newcastle, New South Wales, Australia
| | - Melissa A Tadros
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Javad Barouei
- Laboratory of Neuroimmunology, School of Psychology, University of Newcastle, Newcastle, New South Wales, Australia
| | - Kenneth W Beagley
- Institute of Health Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Vicki L Clifton
- Robinson Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Robert J Callister
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Deborah M Hodgson
- Laboratory of Neuroimmunology, School of Psychology, University of Newcastle, Newcastle, New South Wales, Australia.
| |
Collapse
|
170
|
Strehl C, Buttgereit F. Unraveling the functions of the membrane-bound glucocorticoid receptors: first clues on origin and functional activity. Ann N Y Acad Sci 2014; 1318:1-6. [PMID: 24611742 DOI: 10.1111/nyas.12364] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Glucocorticoids (GCs) are routinely used to treat a wide range of rheumatic and other inflammatory diseases. GCs are steroidal drugs that exert their strong anti-inflammatory and immunosuppressive effects via genomic mechanisms, primarily by signaling through the cytosolic glucocorticoid receptor. In addition, rapid, nongenomic responses following GC treatment have been reported to involve signaling via the membrane-bound glucocorticoid receptor (mGR). Since an important clinical role of this receptor has been proposed, investigations regarding the origin and function of the mGR are currently performed in order to understand rapid GC signaling and to optimize treatment strategies with GCs. Here, we summarize the current knowledge on the mGR and compare these findings to results obtained for other membrane-bound receptors, such as membrane forms of the estrogen and progesterone receptors.
Collapse
Affiliation(s)
- Cindy Strehl
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | | |
Collapse
|
171
|
Saaltink DJ, Vreugdenhil E. Stress, glucocorticoid receptors, and adult neurogenesis: a balance between excitation and inhibition? Cell Mol Life Sci 2014; 71:2499-515. [PMID: 24522255 PMCID: PMC4055840 DOI: 10.1007/s00018-014-1568-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 12/26/2013] [Accepted: 01/16/2014] [Indexed: 02/06/2023]
Abstract
Adult neurogenesis, the birth of new neurons in the mature brain, has attracted considerable attention in the last decade. One of the earliest identified and most profound factors that affect adult neurogenesis both positively and negatively is stress. Here, we review the complex interplay between stress and adult neurogenesis. In particular, we review the role of the glucocorticoid receptor, the main mediator of the stress response in the proliferation, differentiation, migration, and functional integration of newborn neurons in the hippocampus. We review a multitude of mechanisms regulating glucocorticoid receptor activity in relationship to adult neurogenesis. We postulate a novel concept in which the level of glucocorticoid receptor expression directly regulates the excitation-inhibition balance, which is key for proper neurogenesis. We furthermore argue that an excitation-inhibition dis-balance may underlie aberrant functional integration of newborn neurons that is associated with psychiatric and paroxysmal brain disorders.
Collapse
Affiliation(s)
- Dirk-Jan Saaltink
- Department of Medical Pharmacology, Leiden University Medical Center/Leiden Amsterdam Center for Drug Research, 2300 RC, Leiden, The Netherlands
| | | |
Collapse
|
172
|
Reul JMHM. Making memories of stressful events: a journey along epigenetic, gene transcription, and signaling pathways. Front Psychiatry 2014; 5:5. [PMID: 24478733 PMCID: PMC3897878 DOI: 10.3389/fpsyt.2014.00005] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 01/09/2014] [Indexed: 01/19/2023] Open
Abstract
Strong psychologically stressful events are known to have a long-lasting impact on behavior. The consolidation of such, largely adaptive, behavioral responses to stressful events involves changes in gene expression in limbic brain regions such as the hippocampus and amygdala. However, the underlying molecular mechanisms were until recently unresolved. More than a decade ago, we started to investigate the role of these hormones in signaling and epigenetic mechanisms participating in the effects of stress on gene transcription in hippocampal neurons. We discovered a novel, rapid non-genomic mechanism in which glucocorticoids via glucocorticoid receptors facilitate signaling of the ERK-MAPK signaling pathway to the downstream nuclear kinases MSK1 and Elk-1 in dentate gyrus granule neurons. Activation of this signaling pathway results in serine10 (S10) phosphorylation and lysine14 (K14) acetylation at histone H3 (H3S10p-K14ac), leading to the induction of the immediate-early genes c-Fos and Egr-1. In addition, we found a role of the DNA methylation status of gene promoters. A series of studies showed that these molecular mechanisms play a critical role in the long-lasting consolidation of behavioral responses in the forced swim test and Morris water maze. Furthermore, an important role of GABA was found in controlling the epigenetic and gene transcriptional responses to psychological stress. Thus, psychologically stressful events evoke a long-term impact on behavior through changes in hippocampal function brought about by distinct glutamatergic and glucocorticoid-driven changes in epigenetic regulation of gene transcription, which are modulated by (local) GABAergic interneurons and limbic afferent inputs. These epigenetic processes may play an important role in the etiology of stress-related mental disorders such as major depressive and anxiety disorders like post-traumatic stress disorder.
Collapse
Affiliation(s)
- Johannes M. H. M. Reul
- Neuro-Epigenetics Research Group, School of Clinical Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
173
|
Schwabe L, Tegenthoff M, Höffken O, Wolf OT. Mineralocorticoid receptor blockade prevents stress-induced modulation of multiple memory systems in the human brain. Biol Psychiatry 2013; 74:801-8. [PMID: 23871473 DOI: 10.1016/j.biopsych.2013.06.001] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 05/16/2013] [Accepted: 06/02/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND Accumulating evidence suggests that stress may orchestrate the engagement of multiple memory systems in the brain. In particular, stress is thought to favor dorsal striatum-dependent procedural over hippocampus-dependent declarative memory. However, the neuroendocrine mechanisms underlying these modulatory effects of stress remain elusive, especially in humans. Here, we targeted the role of the mineralocorticoid receptor (MR) in the stress-induced modulation of dorsal striatal and hippocampal memory systems in the human brain using a combination of event-related functional magnetic resonance imaging and pharmacologic blockade of the MR. METHODS Eighty healthy participants received the MR antagonist spironolactone (300 mg) or a placebo and underwent a stressor or control manipulation before they performed, in the scanner, a classification task that can be supported by the hippocampus and the dorsal striatum. RESULTS Stress after placebo did not affect learning performance but reduced explicit task knowledge and led to a relative increase in the use of more procedural learning strategies. At the neural level, stress promoted striatum-based learning at the expense of hippocampus-based learning. Functional connectivity analyses showed that this shift was associated with altered coupling of the amygdala with the hippocampus and dorsal striatum. Mineralocorticoid receptor blockade before stress prevented the stress-induced shift toward dorsal striatal procedural learning, same as the stress-induced alterations of amygdala connectivity with hippocampus and dorsal striatum, but resulted in significantly impaired performance. CONCLUSIONS Our findings indicate that the stress-induced shift from hippocampal to dorsal striatal memory systems is mediated by the amygdala, required to preserve performance after stress, and dependent on the MR.
Collapse
Affiliation(s)
- Lars Schwabe
- Institute of Cognitive Neuroscience, Department of Cognitive Psychology, Ruhr-University Bochum, Bochum, Germany.
| | | | | | | |
Collapse
|
174
|
Rivers-Auty J, Ashton JC. Neuroinflammation in ischemic brain injury as an adaptive process. Med Hypotheses 2013; 82:151-8. [PMID: 24345344 DOI: 10.1016/j.mehy.2013.11.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 11/12/2013] [Accepted: 11/19/2013] [Indexed: 12/12/2022]
Abstract
Cerebral ischaemia triggers various physiological processes, some of which have been considered deleterious and others beneficial. These processes have been characterized in one influential model as being part of a transition from injury to repair processes. We argue that another important distinction is between dysregulated and regulated processes. Although intervening in the course of dysregulated processes may be neuroprotective, this is unlikely to be true for regulated processes. This is because from an evolutionary perspective, regulated complex processes that are conserved across many species are likely to be adaptive and provide a survival advantage. We argue that the neuroinflammatory cascade is an adaptive process in this sense, and contrast this with a currently popular theory which we term the maladaptive immune response theory. We review the evidence from clinical and preclinical pharmacology with respect to this theory, and deduced that the evidence is inconclusive at best, and probably falsifies the theory. We argue that this is why there are no anti-inflammatory treatments for cerebral ischaemia, despite 30 years of seemingly promising preclinical results. We therefore propose an opposing theory, which we call the adaptive immune response hypothesis.
Collapse
Affiliation(s)
- Jack Rivers-Auty
- Department of Pharmacology and Toxicology, School of Medical Sciences, University of Otago, P.O. Box 913, Dunedin, New Zealand
| | - John C Ashton
- Department of Pharmacology and Toxicology, School of Medical Sciences, University of Otago, P.O. Box 913, Dunedin, New Zealand.
| |
Collapse
|
175
|
Strehl C, Buttgereit F. Optimized glucocorticoid therapy: teaching old drugs new tricks. Mol Cell Endocrinol 2013; 380:32-40. [PMID: 23403055 DOI: 10.1016/j.mce.2013.01.026] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 01/31/2013] [Accepted: 01/31/2013] [Indexed: 01/01/2023]
Abstract
Glucocorticoids (GCs) are commonly used in the treatment of a wide range of rheumatic and other inflammatory diseases. They exert their potent anti-inflammatory and immunosuppressive effects primarily via so called genomic mechanisms, mediated by the cytosolic glucocorticoid receptor (cGR). This mechanism of GC action can be divided into the transactivation and the transrepression processes. However, also rapid effects of GCs exist which are mediated by specific and unspecific non-genomic mechanisms. A clinical relevance of this mode of GC action is assumed for effects mediated by membrane-bound glucocorticoid receptors, but detailed knowledge on the underlying mechanisms is still missing. Great efforts have been made in the past to diminish GC-induced adverse effects, thus improving the benefit/risk ratio of the drugs. Besides approaches to improve the treatment with conventional glucocorticoids currently available to clinicians, new innovative GCs or GC receptor ligands are also being developed.
Collapse
Affiliation(s)
- Cindy Strehl
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany.
| | | |
Collapse
|
176
|
Ratman D, Vanden Berghe W, Dejager L, Libert C, Tavernier J, Beck IM, De Bosscher K. How glucocorticoid receptors modulate the activity of other transcription factors: a scope beyond tethering. Mol Cell Endocrinol 2013; 380:41-54. [PMID: 23267834 DOI: 10.1016/j.mce.2012.12.014] [Citation(s) in RCA: 289] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 12/13/2012] [Accepted: 12/16/2012] [Indexed: 01/11/2023]
Abstract
The activity of the glucocorticoid receptor (GR), a nuclear receptor transcription factor belonging to subclass 3C of the steroid/thyroid hormone receptor superfamily, is typically triggered by glucocorticoid hormones. Apart from driving gene transcription via binding onto glucocorticoid response elements in regulatory regions of particular target genes, GR can also inhibit gene expression via transrepression, a mechanism largely based on protein:protein interactions. Hereby GR can influence the activity of other transcription factors, without contacting DNA itself. GR is known to inhibit the activity of a growing list of immune-regulating transcription factors. Hence, GCs still rule the clinic for treatments of inflammatory disorders, notwithstanding concomitant deleterious side effects. Although patience is a virtue when it comes to deciphering the many mechanisms GR uses to influence various signaling pathways, the current review is testimony of the fact that groundbreaking mechanistic work has been accumulating over the past years and steadily continues to grow.
Collapse
Affiliation(s)
- Dariusz Ratman
- Cytokine Receptor Lab, VIB Department of Medical Protein Research, VIB, UGent, Albert Baertsoenkaai 3, B-9000 Gent, Belgium.
| | | | | | | | | | | | | |
Collapse
|
177
|
Boguszewski CL. Are patients in remission from Cushing's syndrome mentally healthy? Clin Endocrinol (Oxf) 2013; 79:615-6. [PMID: 23808843 DOI: 10.1111/cen.12278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 06/22/2013] [Indexed: 11/27/2022]
Affiliation(s)
- Cesar L Boguszewski
- Endocrine Division (SEMPR), Department of Internal Medicine, Federal University Hospital of Parana, Curitiba, Brazil
| |
Collapse
|
178
|
Ishimoto H, Wang Z, Rao Y, Wu CF, Kitamoto T. A novel role for ecdysone in Drosophila conditioned behavior: linking GPCR-mediated non-canonical steroid action to cAMP signaling in the adult brain. PLoS Genet 2013; 9:e1003843. [PMID: 24130506 PMCID: PMC3794910 DOI: 10.1371/journal.pgen.1003843] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 08/15/2013] [Indexed: 12/19/2022] Open
Abstract
The biological actions of steroid hormones are mediated primarily by their cognate nuclear receptors, which serve as steroid-dependent transcription factors. However, steroids can also execute their functions by modulating intracellular signaling cascades rapidly and independently of transcriptional regulation. Despite the potential significance of such "non-genomic" steroid actions, their biological roles and the underlying molecular mechanisms are not well understood, particularly with regard to their effects on behavioral regulation. The major steroid hormone in the fruit fly Drosophila is 20-hydroxy-ecdysone (20E), which plays a variety of pivotal roles during development via the nuclear ecdysone receptors. Here we report that DopEcR, a G-protein coupled receptor for ecdysteroids, is involved in activity- and experience-dependent plasticity of the adult central nervous system. Remarkably, a courtship memory defect in rutabaga (Ca²⁺/calmodulin-responsive adenylate cyclase) mutants was rescued by DopEcR overexpression or acute 20E feeding, whereas a memory defect in dunce (cAMP-specific phosphodiestrase) mutants was counteracted when a loss-of-function DopEcR mutation was introduced. A memory defect caused by suppressing dopamine synthesis was also restored through enhanced DopEcR-mediated ecdysone signaling, and rescue and phenocopy experiments revealed that the mushroom body (MB)--a brain region central to learning and memory in Drosophila--is critical for the DopEcR-dependent processing of courtship memory. Consistent with this finding, acute 20E feeding induced a rapid, DopEcR-dependent increase in cAMP levels in the MB. Our multidisciplinary approach demonstrates that DopEcR mediates the non-canonical actions of 20E and rapidly modulates adult conditioned behavior through cAMP signaling, which is universally important for neural plasticity. This study provides novel insights into non-genomic actions of steroids, and opens a new avenue for genetic investigation into an underappreciated mechanism critical to behavioral control by steroids.
Collapse
Affiliation(s)
- Hiroshi Ishimoto
- Department of Anesthesia and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Zhe Wang
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, Iowa, United States of America
| | - Yi Rao
- National Institute of Biological Sciences, Beijing, People's Republic of China
- Peking-Tsinghua Center for Life Sciences, Peking University School of Life Sciences, Beijing, People's Republic of China
| | - Chun-Fang Wu
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Programs in Genetics and Neuroscience, University of Iowa, Iowa City, Iowa, United States of America
| | - Toshihiro Kitamoto
- Department of Anesthesia and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Programs in Genetics and Neuroscience, University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
179
|
The biology of the glucocorticoid receptor: new signaling mechanisms in health and disease. J Allergy Clin Immunol 2013; 132:1033-44. [PMID: 24084075 DOI: 10.1016/j.jaci.2013.09.007] [Citation(s) in RCA: 697] [Impact Index Per Article: 58.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 09/05/2013] [Accepted: 09/06/2013] [Indexed: 12/20/2022]
Abstract
Glucocorticoids are primary stress hormones necessary for life that regulate numerous physiologic processes in an effort to maintain homeostasis. Synthetic derivatives of these hormones have been mainstays in the clinic for treating inflammatory diseases, autoimmune disorders, and hematologic cancers. The physiologic and pharmacologic actions of glucocorticoids are mediated by the glucocorticoid receptor (GR), a member of the nuclear receptor superfamily of ligand-dependent transcription factors. Ligand-occupied GR induces or represses the transcription of thousands of genes through direct binding to DNA response elements, physically associating with other transcription factors, or both. The traditional view that glucocorticoids act through a single GR protein has changed dramatically with the discovery of a large cohort of receptor isoforms with unique expression, gene-regulatory, and functional profiles. These GR subtypes are derived from a single gene by means of alternative splicing and alternative translation initiation mechanisms. Posttranslational modification of these GR isoforms further expands the diversity of glucocorticoid responses. Here we discuss the origin and molecular properties of the GR isoforms and their contribution to the specificity and sensitivity of glucocorticoid signaling in healthy and diseased tissues.
Collapse
|
180
|
Muto A, Taylor MR, Suzawa M, Korenbrot JI, Baier H. Glucocorticoid receptor activity regulates light adaptation in the zebrafish retina. Front Neural Circuits 2013; 7:145. [PMID: 24068988 PMCID: PMC3781318 DOI: 10.3389/fncir.2013.00145] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 08/28/2013] [Indexed: 11/13/2022] Open
Abstract
Glucocorticoids modulate diverse aspects of physiology and behavior, including energy homeostasis, stress response, and memory, through activation of the glucocorticoid receptor (GR). Light perception has profound effects on the production of glucocorticoids via functional connections of the retina to the hypothalamus-pituitary-adrenal axis. We report here that glucocorticoids can also signal in the reverse direction, i. e., regulate visual function in zebrafish, Danio rerio. The zebrafish GR mutant, gr (s357) , harbors a missense mutation that completely blocks the transcriptional activity of GR. In this mutant, visual behavior was abolished following a period of darkness and recovered sluggishly after return to the light. Electrophysiological measurements showed that the photoresponse of the dark-adapted retina was reduced in the mutant and re-adapted to light with a substantial delay. Several gene products, including some that are important for dopaminergic signaling, were misregulated in gr (s357) mutants. We suggest that GR controls a gene network required for visual adaptation in the zebrafish retina and potentially integrates neuroendocrine and sensory responses to environmental changes.
Collapse
Affiliation(s)
- Akira Muto
- Department of Physiology, University of California at San Francisco San Francisco, CA, USA
| | | | | | | | | |
Collapse
|
181
|
Lau T, Heimann F, Bartsch D, Schloss P, Weber T. Nongenomic, glucocorticoid receptor-mediated regulation of serotonin transporter cell surface expression in embryonic stem cell derived serotonergic neurons. Neurosci Lett 2013; 554:115-20. [PMID: 24021805 DOI: 10.1016/j.neulet.2013.08.070] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 07/26/2013] [Accepted: 08/29/2013] [Indexed: 01/30/2023]
Abstract
Depressive disorders have been linked to the combined dysregulation of the hypothalamus-pituitary-adrenal (HPA)-axis and the serotonergic system. The HPA-axis and serotonergic (5-HT) neurons exert reciprocal regulatory actions. It has been reported that glucocorticoid-glucocorticoid receptor (GR) signaling influences serotonin transporter (5-HTT) transcription but data also points to the fact that 5-HTT expression is regulated nongenomically via redistribution of 5-HTT from the cell surface into intracellular compartments. In order to analyze the acute effects of glucocorticoids on 5-HTT cell surface localization we differentiated serotonergic neurons from mouse embryonic stem (ES) cells derived from the C57BL/6N blastocysts. These postmitotic 5-HT neurons express all relevant serotonergic markers following the application of a growth factor-based differentiation protocol. Increasing concentrations of the GR agonist dexamethasone (Dex) resulted in enhanced, dose-dependent 5-HTT cell surface localization in the presence of the protein synthesis inhibitor cycloheximide already 1h after incubation. Inhibition of GR function by the specific GR-antagonist mifepristone abolished the increase in 5-HTT cell surface localization. Hence, our data account for a nongenomic upregulation of 5-HTT cell surface expression by glucocorticoid-GR interaction which likely constitutes a rapid physiological response to increased levels of glucocorticoids as seen during stress. Taken together, we provide a cellular model to analyze and dissect glucocorticoid-5HTT interactions on a molecular level that corresponds to in vivo animal models using C57BL/6N mice.
Collapse
Affiliation(s)
- Thorsten Lau
- Department of Psychiatry and Psychotherapy, Biochemical Laboratory, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, J5, 68159 Mannheim, Germany.
| | | | | | | | | |
Collapse
|
182
|
Gibbison B, Angelini G, Lightman S. Dynamic output and control of the hypothalamic-pituitary-adrenal axis in critical illness and major surgery. Br J Anaesth 2013; 111:347-60. [DOI: 10.1093/bja/aet077] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
183
|
Beste C, Yildiz A, Meissner TW, Wolf OT. Stress improves task processing efficiency in dual-tasks. Behav Brain Res 2013; 252:260-5. [DOI: 10.1016/j.bbr.2013.06.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 06/03/2013] [Accepted: 06/06/2013] [Indexed: 01/20/2023]
|
184
|
Glucocorticoid receptors in the prefrontal cortex regulate dopamine efflux to stress via descending glutamatergic feedback to the ventral tegmental area. Int J Neuropsychopharmacol 2013; 16:1799-807. [PMID: 23590841 DOI: 10.1017/s1461145713000187] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Enhanced dopamine (DA) efflux in the medial prefrontal cortex (mPFC) is a well-documented response to acute stress. We have previously shown that glucocorticoid receptors in the mPFC regulate stress-evoked DA efflux but the underlying mechanism is unknown. DA neurons in the ventral tegmental area (VTA) receive excitatory input from and send reciprocal projections to the mPFC. We hypothesize that blockade of prefrontal glucocorticoid receptors can reduce activity of descending glutamatergic input to the VTA, thereby attenuating stress-evoked DA efflux in the mPFC. Using in vivo microdialysis, we demonstrate that acute tail-pinch stress leads to a significant increase in glutamate efflux in the VTA. Blockade of prefrontal glucocorticoid receptors with the selective antagonist CORT 108297 attenuates stress-evoked glutamate efflux in the VTA together with DA efflux in the mPFC. Furthermore, blockade of ionotrophic glutamate receptors in the VTA attenuates stress-evoked DA efflux in the mPFC. We also examine the possible role of glucocorticoid-induced synthesis and release of endocannabinoids acting presynaptically via cannabinoid CB1 receptors to inhibit GABA release onto prefrontal pyramidal cells, thus enhancing descending glutamatergic input to the VTA leading to an increase in mPFC DA efflux during stress. However, administration of the cannabinoid CB1 receptor antagonist into the mPFC does not attenuate stress-evoked DA efflux in the mPFC. Taken together, our data indicate that glucocorticoids act locally within the mPFC to modulate mesocortical DA efflux by potentiation of glutamatergic drive onto DA neurons in the VTA.
Collapse
|
185
|
Merz CJ, Hermann A, Stark R, Wolf OT. Cortisol modifies extinction learning of recently acquired fear in men. Soc Cogn Affect Neurosci 2013; 9:1426-34. [PMID: 23945999 DOI: 10.1093/scan/nst137] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Exposure therapy builds on the mechanism of fear extinction leading to decreased fear responses. How the stress hormone cortisol affects brain regions involved in fear extinction in humans is unknown. For this reason, we tested 32 men randomly assigned to receive either 30 mg hydrocortisone or placebo 45 min before fear extinction. In fear acquisition, a picture of a geometrical figure was either partially paired (conditioned stimulus; CS+) or not paired (CS-) with an electrical stimulation (unconditioned stimulus; UCS). In fear extinction, each CS was presented again, but no UCS occurred. Cortisol increased conditioned skin conductance responses in early and late extinction. In early extinction, higher activation towards the CS- than to the CS+ was found in the amygdala, hippocampus and posterior parahippocampal gyrus. This pattern might be associated with the establishment of a new memory trace. In late extinction, the placebo compared with the cortisol group displayed enhanced CS+/CS- differentiation in the amygdala, medial frontal cortex and nucleus accumbens. A change from early deactivation to late activation of the extinction circuit as seen in the placebo group seems to be needed to enhance extinction and to reduce fear. Cortisol appears to interfere with this process thereby impairing extinction of recently acquired conditioned fear.
Collapse
Affiliation(s)
- Christian Josef Merz
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Ruhr-University Bochum, Universitätsstr. 150, 44780 Bochum, Bender Institute of Neuroimaging, and Department of Psychotherapy and Systems Neuroscience, Justus Liebig University Giessen, Otto-Behaghel-Str. 10F, 35394 Giessen, Germany Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Ruhr-University Bochum, Universitätsstr. 150, 44780 Bochum, Bender Institute of Neuroimaging, and Department of Psychotherapy and Systems Neuroscience, Justus Liebig University Giessen, Otto-Behaghel-Str. 10F, 35394 Giessen, Germany
| | - Andrea Hermann
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Ruhr-University Bochum, Universitätsstr. 150, 44780 Bochum, Bender Institute of Neuroimaging, and Department of Psychotherapy and Systems Neuroscience, Justus Liebig University Giessen, Otto-Behaghel-Str. 10F, 35394 Giessen, Germany Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Ruhr-University Bochum, Universitätsstr. 150, 44780 Bochum, Bender Institute of Neuroimaging, and Department of Psychotherapy and Systems Neuroscience, Justus Liebig University Giessen, Otto-Behaghel-Str. 10F, 35394 Giessen, Germany
| | - Rudolf Stark
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Ruhr-University Bochum, Universitätsstr. 150, 44780 Bochum, Bender Institute of Neuroimaging, and Department of Psychotherapy and Systems Neuroscience, Justus Liebig University Giessen, Otto-Behaghel-Str. 10F, 35394 Giessen, Germany Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Ruhr-University Bochum, Universitätsstr. 150, 44780 Bochum, Bender Institute of Neuroimaging, and Department of Psychotherapy and Systems Neuroscience, Justus Liebig University Giessen, Otto-Behaghel-Str. 10F, 35394 Giessen, Germany
| | - Oliver Tobias Wolf
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Ruhr-University Bochum, Universitätsstr. 150, 44780 Bochum, Bender Institute of Neuroimaging, and Department of Psychotherapy and Systems Neuroscience, Justus Liebig University Giessen, Otto-Behaghel-Str. 10F, 35394 Giessen, Germany
| |
Collapse
|
186
|
Trotman HD, Holtzman CW, Ryan AT, Shapiro DI, MacDonald AN, Goulding SM, Brasfield JL, Walker EF. The development of psychotic disorders in adolescence: a potential role for hormones. Horm Behav 2013; 64:411-9. [PMID: 23998682 PMCID: PMC4070947 DOI: 10.1016/j.yhbeh.2013.02.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 01/20/2013] [Accepted: 02/26/2013] [Indexed: 12/14/2022]
Abstract
This article is part of a Special Issue "Puberty and Adolescence". The notion that adolescence is characterized by dramatic changes in behavior, and often by emotional upheaval, is widespread and longstanding in popular western culture. In recent decades, this notion has gained increasing support from empirical research showing that the peri- and post-pubertal developmental stages are associated with a significant rise in the rate of psychiatric symptoms and syndromes. As a result, interest in adolescent development has burgeoned among researchers focused on the origins of schizophrenia and other psychotic disorders. Two factors have fueled this trend: 1) increasing evidence from longitudinal research that adolescence is the modal period for the emergence of "prodromal" manifestations, or precursors of psychotic symptoms, and 2) the rapidly accumulating scientific findings on brain structural and functional changes occurring during adolescence and young adulthood. Further, gonadal and adrenal hormones are beginning to play a more prominent role in conceptualizations of adolescent brain development, as well as in the origins of psychiatric symptoms during this period (Walker and Bollini, 2002; Walker et al., 2008). In this paper, we begin by providing an overview of the nature and course of psychotic disorders during adolescence/young adulthood. We then turn to the role of hormones in modulating normal brain development, and the potential role they might play in the abnormal brain changes that characterize youth at clinical high-risk (CHR) for psychosis. The activational and organizational effects of hormones are explored, with a focus on how hormone-induced changes might be linked with neuropathological processes in the emergence of psychosis.
Collapse
Affiliation(s)
- Hanan D Trotman
- Department of Psychology, Emory University, Atlanta, GA 30322, USA.
| | | | | | | | | | | | | | | |
Collapse
|
187
|
Dindia L, Faught E, Leonenko Z, Thomas R, Vijayan MM. Rapid cortisol signaling in response to acute stress involves changes in plasma membrane order in rainbow trout liver. Am J Physiol Endocrinol Metab 2013; 304:E1157-66. [PMID: 23531621 DOI: 10.1152/ajpendo.00500.2012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The activation of genomic signaling in response to stressor-mediated cortisol elevation has been studied extensively in teleosts. However, very little is known about the rapid signaling events elicited by this steroid. We tested the hypothesis that cortisol modulates key stress-related signaling pathways in response to an acute stressor in fish liver. To this end, we investigated the effect of an acute stressor on biophysical properties of plasma membrane and on stressor-related protein phosphorylation in rainbow trout (Oncorhynchus mykiss) liver. A role for cortisol in modulating the acute cellular stress response was ascertained by blocking the stressor-induced elevation of this steroid by metyrapone. The acute stressor exposure increased plasma cortisol levels and liver membrane fluidity (measured by anisotropy of 1,6-diphenyl-1,3,5-hexatriene), but these responses were abolished by metyrapone. Atomic force microscopy further confirmed biophysical alterations in liver plasma membrane in response to stress, including changes in membrane domain topography. The changes in membrane order did not correspond to any changes in membrane fatty acid components after stress, suggesting that changes in membrane structure may be associated with cortisol incorporation into the lipid bilayer. Plasma cortisol elevation poststress correlated positively with activation of intracellular stress signaling pathways, including increased phosphorylation of extracellular signal-related kinases as well as several putative PKA and PKC but not Akt substrate proteins. Together, our results indicate that stressor-induced elevation of plasma cortisol level is associated with alterations in plasma membrane fluidity and rapid activation of stress-related signaling pathways in trout liver.
Collapse
Affiliation(s)
- Laura Dindia
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | | | | | | | | |
Collapse
|
188
|
Teng Z, Zhang M, Zhao M, Zhang W. Glucocorticoid exerts its non-genomic effect on IPSC by activation of a phospholipase C-dependent pathway in prefrontal cortex of rats. J Physiol 2013; 591:3341-53. [PMID: 23652592 DOI: 10.1113/jphysiol.2013.254961] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In response to stressor, the brain activates a comprehensive stress system. Among others, this stress system causes release of glucocorticoids that also feed back to the brain. Glucocorticoids affect brain function by activation of both delayed, genomic and rapid, non-genomic mechanisms in rodents. Here we report that application of the potent glucocorticoid receptor agonist dexamethasone (DEX) caused a rapid increase of spontaneous and miniature inhibitory postsynaptic currents (IPSCs) and elicited intermittent burst activities through a non-genomic pathway, involving membrane-located receptors. The onset of the rapid effect in prefrontal cortex (PFC, <15 min) was much slower than in hippocampus (<5 min). The intermittent burst activities were abolished in the presence of TTX. Furthermore, the nitric oxide (NO) pathway was present and endogenously activated in PFC. Part of the rapid DEX effect in PFC remained after blocking NO-sensitive guanylyl cyclase that was due to activation of a phospholipase C-diacylglycerol-dependent signalling pathway. Thus, our data demonstrated that glucocorticoids could rapidly enhance IPSCs and evoke burst activities by activation of at least two different signalling pathways in hippocampus and PFC of rats.
Collapse
Affiliation(s)
- Zenghui Teng
- Laboratory of Molecular Psychiatry, Department of Psychiatry, Westfälische Wilhelms-University of Münster, Albert-Schweitzer-Campus 1, Gebäude 9a, 48149 Münster, Germany
| | | | | | | |
Collapse
|
189
|
Benes J, Tomankova H, Novakova M, Rohan Z, Kvetnansky R, Myslivecek J. Corticotropin-releasing hormone affects short immobilization stress-induced changes in lung cytosolic and membrane glucocorticoid binding sites. Cell Mol Neurobiol 2013; 33:503-11. [PMID: 23430272 PMCID: PMC11497924 DOI: 10.1007/s10571-013-9916-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 02/06/2013] [Indexed: 12/22/2022]
Abstract
Glucocorticoids act via glucocorticoid receptors (GR), typically localized in the cytosol (cGR). Rapid action is probably mediated via membrane receptors (mGR). In corticotropin-releasing hormone knockouts (CRH-KO), basal plasma glucocorticoid levels do differ from wild type levels (WT), but are approximately ten times lower during exposure to immobilization stress (IMMO) in comparison to WT. We tested the following hypotheses: (1) the mice lung tissue GR basal numbers would not be changed in CRH-KO (because of similar glucocorticoid levels), (2) the number of GR would be changed in WT but not in KO during short (30, 90, and 120 min) IMMO (because of higher increase of glucocorticoid levels in WT). The basal levels of cGR were not changed in CRH-KO (compared to WT), while mGR were significantly lower (62 %) in CRH-KO. In WT, there was the only decrease (to 32 %) in cGR after 120 min when we also found an increase in mGR in WT (to 201 %). In CRH-KO, IMMO caused gradual decrease in cGR (to 52 % after 30 min, to 46 % after 90 min, and to 32 % after 120 min). In CRH-KO, the only increase in mGR appeared already at 30 min of IMMO. These data suggest, on the contrary to our hypotheses, that CRH-KO are more susceptible to GR changes in early phases of stress.
Collapse
Affiliation(s)
- Jan Benes
- Institute of Physiology, 1st Faculty of Medicine, Charles University, Albertov 5, 12800 Prague, Czech Republic
| | - Hana Tomankova
- Institute of Physiology, 1st Faculty of Medicine, Charles University, Albertov 5, 12800 Prague, Czech Republic
| | - Martina Novakova
- Institute of Physiology, 1st Faculty of Medicine, Charles University, Albertov 5, 12800 Prague, Czech Republic
| | - Zdeněk Rohan
- Institute of Physiology, 1st Faculty of Medicine, Charles University, Albertov 5, 12800 Prague, Czech Republic
| | - Richard Kvetnansky
- Institute of Experimental Endocrinology, Centre of Excellence for Cardiovascular Research and CENDO, Slovak Academy of Sciences, Vlarska 3, 833 06 Bratislava, Slovakia
| | - Jaromir Myslivecek
- Institute of Physiology, 1st Faculty of Medicine, Charles University, Albertov 5, 12800 Prague, Czech Republic
| |
Collapse
|
190
|
Abstract
Mineralocorticoid receptors (MR) exist in many tissues, in which they mediate diverse functions crucial to normal physiology, including tissue repair and electrolyte and fluid homeostasis. However, inappropriate activation of MR within these tissues, and especially in the brain, causes hypertension and pathological vascular, cardiac, and renal remodeling. MR binds aldosterone, cortisol and corticosterone with equal affinity. In aldosterone-target cells, co-expression with the 11β-hydroxysteroid dehydrogenase 2 (HSD2) allows aldosterone specifically to activate MR. Aldosterone levels are excessive in primary aldosteronism, but in conditions with increased oxidative stress, like CHF, obesity and diabetes, MR may also be inappropriately activated by glucocorticoids. Unlike thiazide diuretics, MR antagonists are diuretics that do not cause insulin resistance. Addition of MR antagonists to standard treatment for hypertension and cardiac or renal disease decreases end-organ pathology and sympathetic nerve activation (SNA), and increases quality of life indices.
Collapse
|
191
|
Solas M, Gerenu G, Gil-Bea FJ, Ramírez MJ. Mineralocorticoid receptor activation induces insulin resistance through c-Jun N-terminal kinases in response to chronic corticosterone: cognitive implications. J Neuroendocrinol 2013. [PMID: 23181759 DOI: 10.1111/jne.12006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
It is becoming evident that chronic exposure to glucocorticoids might not only result in insulin resistance or cognitive deficits, but also is considered as a risk factor for pathologies such as depression or Alzheimer's disease. In the present study, in vivo experiments using a non-invasive method of chronic administration of corticosterone in drinking water demonstrated that chronic corticosterone administration led to cognitive impairment in the novel object recognition test and insulin resistance, as shown by significant increases in plasma insulin levels and the homeostatic model assessment index, and decreased insulin receptor phosphorylation. Corticosterone treatment induced an increased expression of stress-activated c-Jun N-terminal kinase (JNK) in the hippocampus, accompanied by decreases in glycogen synthase kinase 3β, increases in pTau levels and increased neuronal cell death (caspase-3 activity). All these effects were reversed by the administration of a JNK1 inhibitor or by the mineralocorticoid receptor antagonist spironolactone. It is suggested that the mineralocorticoid receptors and JNK-mediated pathways are involved in the interaction of glucocorticoid-insulin resistance and the development of relevant cellular processes for Alzheimer's disease.
Collapse
Affiliation(s)
- M Solas
- Department of Pharmacology, University of Navarra, Pamplona, Spain
| | | | | | | |
Collapse
|
192
|
Zalachoras I, Houtman R, Meijer OC. Understanding stress-effects in the brain via transcriptional signal transduction pathways. Neuroscience 2013; 242:97-109. [PMID: 23545270 DOI: 10.1016/j.neuroscience.2013.03.038] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 03/22/2013] [Accepted: 03/23/2013] [Indexed: 12/22/2022]
Abstract
Glucocorticoid hormones exert crucial effects on the brain in relation to physiology, endocrine regulation, mood and cognition. Their two receptor types, glucocorticoid and mineralocorticoid receptors (GR and MR), are members of the nuclear receptor superfamily and act in large measure as transcription factors. The outcome of MR/GR action on the genome depends on interaction with members from different protein families, which are of crucial importance for cross-talk with other neuronal and hormonal signals that impinge on the glucocorticoid sensitive circuitry. Relevant interacting proteins include other transcription factors that may either tether the receptor to the DNA, or that bind in the vicinity of GR and MR to tune the transcriptional response. In addition, transcriptional coregulator proteins constitute the actual signal transduction pathway to the transcription machinery. We review the current evidence for involvement of individual coregulators in GR-dependent effects on stress responses, and learning and memory. We discuss the use of in vitro and in silico tools to predict those coregulators that are of importance for particular brain processes. Finally, we discuss the potential of selective receptor modulators that may only allow a subset of all interactions, thus allowing more selective targeting of glucocorticoid-dependent processes in the brain.
Collapse
Affiliation(s)
- I Zalachoras
- Department of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | |
Collapse
|
193
|
Sandi C. Stress and cognition. WILEY INTERDISCIPLINARY REVIEWS. COGNITIVE SCIENCE 2013; 4:245-261. [DOI: 10.1002/wcs.1222] [Citation(s) in RCA: 179] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Carmen Sandi
- Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| |
Collapse
|
194
|
Anacker C, Pariante CM. New models to investigate complex glucocorticoid receptor functions. Front Behav Neurosci 2012; 6:90. [PMID: 23293592 PMCID: PMC3533204 DOI: 10.3389/fnbeh.2012.00090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 12/12/2012] [Indexed: 01/11/2023] Open
Affiliation(s)
- Christoph Anacker
- Department of Psychological Medicine, Section of Perinatal Psychiatry and Stress, Psychiatry and Immunology (SPI-Lab), Institute of Psychiatry, King's College London London, UK
| | | |
Collapse
|
195
|
Endothelial glucocorticoid receptor is required for protection against sepsis. Proc Natl Acad Sci U S A 2012; 110:306-11. [PMID: 23248291 DOI: 10.1073/pnas.1210200110] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The glucocorticoid receptor (GR) is ubiquitously expressed on nearly all cell types, but tissue-specific deletion of this receptor can produce dramatic whole organism phenotypes. In this study we investigated the role of the endothelial GR in sepsis in vivo and in vitro. Mice with an endothelial-specific GR deletion and controls were treated with 12.5 mg/kg LPS and phenotyped. Mice lacking GR showed significantly increased mortality, more hemodynamic instability, higher nitric oxide levels, and higher levels of the inflammatory cytokines, tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) compared with controls. There were no differences in rates of apoptosis or macrophage recruitment between the two groups. Both endothelial nitric oxide synthase (eNOS) and inducible nitric oxide synthase (iNOS) expression were increased after LPS challenge in mice with endothelial GR deficiency, and aminoguanidine, a specific iNOS inhibitor in mice was able to rescue hemodynamic collapse in these animals. In vitro, human umbilical vein cells (HUVECs) subjected to GR knockdown by siRNA showed increased expression of eNOS at baseline that persisted after treatment with LPS. Both eNOS and iNOS mRNA was increased by qPCR. In HUVECs lacking GR, NF-κB levels and NF-κB-dependent genes tissue factor and IL-6 were increased compared with controls. Thus, endothelial GR is a critical regulator of NF-κB activation and nitric oxide synthesis in sepsis.
Collapse
|
196
|
Chantong B, Kratschmar DV, Nashev LG, Balazs Z, Odermatt A. Mineralocorticoid and glucocorticoid receptors differentially regulate NF-kappaB activity and pro-inflammatory cytokine production in murine BV-2 microglial cells. J Neuroinflammation 2012. [PMID: 23190711 PMCID: PMC3526453 DOI: 10.1186/1742-2094-9-260] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background Microglia, the resident macrophage-like cells in the brain, regulate innate immune responses in the CNS to protect neurons. However, excessive activation of microglia contributes to neurodegenerative diseases. Corticosteroids are potent modulators of inflammation and mediate their effects by binding to mineralocorticoid receptors (MR) and glucocorticoid receptors (GR). Here, the coordinated activities of GR and MR on the modulation of the nuclear factor-κB (NF-κB) pathway in murine BV-2 microglial cells were studied. Methods BV-2 cells were treated with different corticosteroids in the presence or absence of MR and GR antagonists. The impact of the glucocorticoid-activating enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) was determined by incubating cells with 11-dehydrocorticosterone, with or without selective inhibitors. Expression of interleukin-6 (IL-6), tumor necrosis factor receptor 2 (TNFR2), and 11β-HSD1 mRNA was analyzed by RT-PCR and IL-6 protein expression by ELISA. NF-κB activation and translocation upon treatment with various corticosteroids were visualized by western blotting, immunofluorescence microscopy, and translocation assays. Results GR and MR differentially regulate NF-κB activation and neuroinflammatory parameters in BV-2 cells. By converting inactive 11-dehydrocorticosterone to active corticosterone, 11β-HSD1 essentially modulates the coordinated action of GR and MR. Biphasic effects were observed for 11-dehydrocorticosterone and corticosterone, with an MR-dependent potentiation of IL-6 and tumor necrosis factor-α (TNF-α) expression and NF-κB activation at low/moderate concentrations and a GR-dependent suppression at high concentrations. The respective effects were confirmed using the MR ligand aldosterone and the antagonist spironolactone as well as the GR ligand dexamethasone and the antagonist RU-486. NF-κB activation could be blocked by spironolactone and the inhibitor of NF-κB translocation Cay-10512. Moreover, an increased expression of TNFR2 was observed upon treatment with 11-dehydrocorticosterone and aldosterone, which was reversed by 11β-HSD1 inhibitors and/or spironolactone and Cay-10512. Conclusions A tightly coordinated GR and MR activity regulates the NF-κB pathway and the control of inflammatory mediators in microglia cells. The balance of GR and MR activity is locally modulated by the action of 11β-HSD1, which is upregulated by pro-inflammatory mediators and may represent an important feedback mechanism involved in resolution of inflammation.
Collapse
Affiliation(s)
- Boonrat Chantong
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | | | | | | | | |
Collapse
|
197
|
Brain-derived neurotrophic factor and glucocorticoids: reciprocal influence on the central nervous system. Neuroscience 2012; 239:157-72. [PMID: 23069755 DOI: 10.1016/j.neuroscience.2012.09.073] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 09/14/2012] [Accepted: 09/29/2012] [Indexed: 12/25/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) has multiple roles in the central nervous system (CNS), including maintaining cell survival and regulation of synaptic function. In CNS neurons, BDNF triggers activation of phospholipase Cγ (PLCγ), mitogen-activated protein/extracellular signal-regulated kinase (MAPK/ERK), and phosphoinositide 3-kinase (PI3K)/Akt pathways, influencing neuronal cells beneficially through these intracellular signaling cascades. There is evidence to suggest that decreased BDNF expression or function is related to the pathophysiology of brain diseases including psychiatric disorders. Additionally, glucocorticoids, which are critical stress hormones, also influence neuronal function in the CNS, and are putatively involved in the onset of depression when levels are abnormally high. In animal models of depression, changes in glucocorticoid levels, expression of glucocorticoid receptor (GR), and alterations in BDNF signaling are observed. Interestingly, several studies using in vivo and in vitro systems suggest that glucocorticoids interact with BDNF to ultimately affect CNS function. In the present review, we provide an overview of recent evidence concerning the interaction between BDNF and glucocorticoids.
Collapse
|
198
|
Virtual screening as a strategy for the identification of xenobiotics disrupting corticosteroid action. PLoS One 2012; 7:e46958. [PMID: 23056542 PMCID: PMC3464284 DOI: 10.1371/journal.pone.0046958] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 09/06/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Impaired corticosteroid action caused by genetic and environmental influence, including exposure to hazardous xenobiotics, contributes to the development and progression of metabolic diseases, cardiovascular complications and immune disorders. Novel strategies are thus needed for identifying xenobiotics that interfere with corticosteroid homeostasis. 11β-hydroxysteroid dehydrogenase 2 (11β-HSD2) and mineralocorticoid receptors (MR) are major regulators of corticosteroid action. 11β-HSD2 converts the active glucocorticoid cortisol to the inactive cortisone and protects MR from activation by glucocorticoids. 11β-HSD2 has also an essential role in the placenta to protect the fetus from high maternal cortisol concentrations. METHODS AND PRINCIPAL FINDINGS We employed a previously constructed 3D-structural library of chemicals with proven and suspected endocrine disrupting effects for virtual screening using a chemical feature-based 11β-HSD pharmacophore. We tested several in silico predicted chemicals in a 11β-HSD2 bioassay. The identified antibiotic lasalocid and the silane-coupling agent AB110873 were found to concentration-dependently inhibit 11β-HSD2. Moreover, the silane AB110873 was shown to activate MR and stimulate mitochondrial ROS generation and the production of the proinflammatory cytokine interleukin-6 (IL-6). Finally, we constructed a MR pharmacophore, which successfully identified the silane AB110873. CONCLUSIONS Screening of virtual chemical structure libraries can facilitate the identification of xenobiotics inhibiting 11β-HSD2 and/or activating MR. Lasalocid and AB110873 belong to new classes of 11β-HSD2 inhibitors. The silane AB110873 represents to the best of our knowledge the first industrial chemical shown to activate MR. Furthermore, the MR pharmacophore can now be used for future screening purposes.
Collapse
|
199
|
Sousa N, Almeida OFX. Disconnection and reconnection: the morphological basis of (mal)adaptation to stress. Trends Neurosci 2012; 35:742-51. [PMID: 23000140 DOI: 10.1016/j.tins.2012.08.006] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Revised: 07/15/2012] [Accepted: 08/20/2012] [Indexed: 12/26/2022]
Abstract
Maladaptive responses to stress and the associated hypersecretion of glucocorticoids cause psychopathologies ranging from hyperemotional states and mood dysfunction to cognitive impairments. Research in both humans and animal models has begun to identify morphological correlates of these functional changes. These include dendritic and synaptic reorganization, glial remodeling, and altered cell fate in cortical and subcortical structures. The emerging view is that stress induces a 'disconnection syndrome' whereby the transmission and integration of information that are critical for orchestrating appropriate physiological and behavioral responses are perturbed. High-resolution spatiotemporal mapping of the complete neural circuitry and identification of the cellular processes impacted by stress will help to advance discovery of strategies to reduce or reverse the burden of stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Nuno Sousa
- Life and Health Science Research Institute, School of Health Sciences, University of Minho, Braga, Portugal.
| | | |
Collapse
|
200
|
Exposure of Wistar rats to 24-h psycho-social stress alters gene expression in the inferior colliculus. Neurosci Lett 2012; 527:40-5. [PMID: 22922217 DOI: 10.1016/j.neulet.2012.08.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 07/16/2012] [Accepted: 08/09/2012] [Indexed: 11/22/2022]
Abstract
Recently, we have demonstrated that the exposure of Wistar rats to psycho-social stress results in a transient auditory hypersensitivity. Here, to learn more about modifications occurring in auditory brainstem, we have analyzed gene expression pattern in inferior colliculus using quantitative RT-PCR. As targets, we have chosen genes associated with: neural activity (FBJ osteosarcoma viral oncogene, cFos), hypoxia (nitric oxide synthase inducible, iNos; superoxide dismutase 2, Sod2), neuroprotection (nerve growth factor beta, Ngfb; heat shock factor 1, Hsf1; heat shock protein 70, Hsp70) and inflammation (tumor necrosis factor alpha, Tnfa; tumor necrosis factor alpha receptor, Tnfar; substance P, Sp; cyclooxygenase 2, Cox2). We found that the expression of all genes was modified following stress, as compared to the controls. Immediately after stress, the number of transcripts encoding iNos, Sod2, Hsf1, Ngfb, Tnfa, Tnfar and Sp was significantly increased, suggesting possible modulation during exposure to stressor. Interestingly, we found that expression of Hsf1 and Ngfb at this particular time was left-right asymmetrical: there were more transcripts of both genes found in the left colliculi, as compared to the right colliculi. Three hours post-stress, iNos, Hsf1, Tnfa and Tnfar were still upregulated, Sod2, Ngfb and Sp went back to baseline and Cox2 was upregulated. Six hours post-stress, cFos mRNA became downregulated. The number of Hsp70 mRNA increased 24h post-stress. Except for the reduced number of cFos transcripts, expression of all other genes tested reached the baseline seven days post-stress. Presented results corroborate the concept of auditory system responding to the psycho-social stress. Post-stress changes in the IC gene expression could likely indicate shift from allostasis to homeostasis in the auditory brainstem.
Collapse
|