1
|
Wu A, Yang G, Liu G, Zhang J. SGK1 upregulation in GFAP + neurons in the frontal association cortex protects against neuronal apoptosis after spinal cord injury. Cell Death Dis 2025; 16:237. [PMID: 40175324 PMCID: PMC11965300 DOI: 10.1038/s41419-025-07542-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 02/23/2025] [Accepted: 03/17/2025] [Indexed: 04/04/2025]
Abstract
Spinal cord injury (SCI) casts devastating and long-lasting impacts on the well-being of patients. Cognitive deficits and emotional disorders are common in individuals with SCI, yet the underlying mechanisms are not completely understood. Astrogliosis and glial scar formation occur during the subacute phase post-injury, playing complicated roles in remyelination and neurite regrowth. Therefore, we constructed a GFAP-IRES-Venus-AkaLuc knock-in mouse model for the corresponding studies. Surprisingly, complete spinal cord transection (SCT) surgery led to earlier and more prominent augmentation of bioluminescence in the brain than in the spinal cord. Bulk RNA sequencing revealed the activation of apoptotic signaling and the upregulation of serum and glucocorticoid-regulated kinase 1 (SGK1). The pattern of GFAP signals changed throughout the brain after SCT, as indicated by tissue clearing and immunostaining. Specifically, GFAP signals were intensified in the frontal association cortex (FrA), an encephalic region involved in associative learning and recognition memory processes. Further exploration unraveled that intensified GFAP signals in the FrA were attributed to apoptotic neurons with SGK1 upregulation, which was induced by sustained high glucocorticoid levels after SCT. The introduction of SGK1 silencing vectors confirmed that SGK upregulation in these FrA neurons exerted anti-apoptotic effects through NRF2/HO-1 signaling. In addition, SGK1 knockdown in FrA neurons aggravated the post-SCI depressive-like behaviors. Thus, ectopic SGK1 expression designated for limbic neurons could serve as a promising therapeutic target for the future development of treatments for spinal cord injuries.
Collapse
Affiliation(s)
- Anbiao Wu
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Guang Yang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Genyu Liu
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jiyan Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, China.
| |
Collapse
|
2
|
Joëls M, Karst H, Tasker JG. The emerging role of rapid corticosteroid actions on excitatory and inhibitory synaptic signaling in the brain. Front Neuroendocrinol 2024; 74:101146. [PMID: 39004314 DOI: 10.1016/j.yfrne.2024.101146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/26/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
Over the past two decades, there has been increasing evidence for the importance of rapid-onset actions of corticosteroid hormones in the brain. Here, we highlight the distinct rapid corticosteroid actions that regulate excitatory and inhibitory synaptic transmission in the hypothalamus, the hippocampus, basolateral amygdala, and prefrontal cortex. The receptors that mediate rapid corticosteroid actions are located at or close to the plasma membrane, though many of the receptor characteristics remain unresolved. Rapid-onset corticosteroid effects play a role in fast neuroendocrine feedback as well as in higher brain functions, including increased aggression and anxiety, and impaired memory retrieval. The rapid non-genomic corticosteroid actions precede and complement slow-onset, long-lasting transcriptional actions of the steroids. Both rapid and slow corticosteroid actions appear to be indispensable to adapt to a continuously changing environment, and their imbalance can increase an individual's susceptibility to psychopathology.
Collapse
Affiliation(s)
- Marian Joëls
- University Medical Center Groningen, University of Groningen, the Netherlands; University Medical Center Utrecht, Utrecht University, the Netherlands.
| | - Henk Karst
- University Medical Center Utrecht, Utrecht University, the Netherlands; SILS-CNS. University of Amsterdam, the Netherlands.
| | - Jeffrey G Tasker
- Department of Cell and Molecular Biology and Tulane Brain Institute, Tulane University, and Southeast Louisiana Veterans Affairs Healthcare System, New Orleans, USA.
| |
Collapse
|
3
|
de Kloet ER, Joëls M. The cortisol switch between vulnerability and resilience. Mol Psychiatry 2024; 29:20-34. [PMID: 36599967 DOI: 10.1038/s41380-022-01934-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023]
Abstract
In concert with neuropeptides and transmitters, the end products of the hypothalamus-pituitary-adrenal (HPA) axis, the glucocorticoid hormones cortisol and corticosterone (CORT), promote resilience: i.e., the ability to cope with threats, adversity, and trauma. To exert this protective action, CORT activates mineralocorticoid receptors (MR) and glucocorticoid receptors (GR) that operate in a complementary manner -as an on/off switch- to coordinate circadian events, stress-coping, and adaptation. The evolutionary older limbic MR facilitates contextual memory retrieval and supports an on-switch in the selection of stress-coping styles at a low cost. The rise in circulating CORT concentration after stress subsequently activates a GR-mediated off-switch underlying recovery of homeostasis by providing the energy for restraining the primary stress reactions and promoting cognitive control over emotional reactivity. GR activation facilitates contextual memory storage of the experience to enable future stress-coping. Such complementary MR-GR-mediated actions involve rapid non-genomic and slower gene-mediated mechanisms; they are time-dependent, conditional, and sexually dimorphic, and depend on genetic background and prior experience. If coping fails, GR activation impairs cognitive control and promotes emotional arousal which eventually may compromise resilience. Such breakdown of resilience involves a transition to a chronic stress construct, where information processing is crashed; it leads to an imbalanced MR-GR switch and hence increased vulnerability. Novel MR-GR modulators are becoming available that may reset a dysregulated stress response system to reinstate the cognitive flexibility required for resilience.
Collapse
Affiliation(s)
- E Ronald de Kloet
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden University, Leiden, The Netherlands.
- Leiden/Amsterdam Center of Drug Research, Leiden University, Leiden, The Netherlands.
| | - Marian Joëls
- Dept. Translational Neuroscience, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
4
|
de Kloet ER. Glucocorticoid feedback paradox: a homage to Mary Dallman. Stress 2023; 26:2247090. [PMID: 37589046 DOI: 10.1080/10253890.2023.2247090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/07/2023] [Indexed: 08/18/2023] Open
Abstract
As the end product of the hypothalamus-pituitary-adrenal (HPA) axis, the glucocorticoid hormones cortisol and corticosterone coordinate circadian activities, stress-coping, and adaptation to change. For this purpose, the hormone promotes energy metabolism and controls defense reactions in the body and brain. This life-sustaining action exerted by glucocorticoids occurs in concert with the autonomic nervous and immune systems, transmitters, growth factors/cytokines, and neuropeptides. The current contribution will focus on the glucocorticoid feedback paradox in the HPA-axis: the phenomenon that stress responsivity remains resilient if preceded by stress-induced secretion of glucocorticoid hormone, but not if this hormone is previously administered. Furthermore, in animal studies, the mixed progesterone/glucocorticoid antagonist RU486 or mifepristone switches to an apparent partial agonist upon repeated administration. To address these enigmas several interesting phenomena are highlighted. These include the conditional nature of the excitation/inhibition balance in feedback regulation, the role of glucose as a determinant of stress responsivity, and the potential of glucocorticoids in resetting the stress response system. The analysis of the feedback paradox provides also a golden opportunity to review the progress in understanding the role of glucocorticoid hormone in resilience and vulnerability during stress, the science that was burned deeply in Mary Dallman's emotions.
Collapse
Affiliation(s)
- Edo Ronald de Kloet
- Department of Clinical Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
5
|
Meijer OC, Kooijman S, Kroon J, Winter EM. The importance of the circadian trough in glucocorticoid signaling: a variation on B-flat. Stress 2023; 26:2275210. [PMID: 37874158 DOI: 10.1080/10253890.2023.2275210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 09/28/2023] [Indexed: 10/25/2023] Open
Abstract
Glucocorticoid hormones are essential for health, but overexposure may lead to many detrimental effects, including metabolic, psychiatric, and bone disease. These effects may not only be due to increased overall exposure to glucocorticoids, but also to elevated hormone levels at the time of the physiological circadian trough of glucocorticoid levels. The late Mary Dallman developed a model that allows the differentiation between the effects of overall 24-hour glucocorticoid overexposure and the effects of a lack of circadian rhythmicity. For this, she continuously treated rats with a low dose of corticosterone (or "B"), which leads to a constant hormone level, without 24-hour overexposure using subcutaneously implanted pellets. The data from this "B-flat" model suggest that even modest elevations of glucocorticoid signaling during the time of the normal circadian trough of hormone secretion are a substantial contributor to the negative effects of glucocorticoids on health.
Collapse
Affiliation(s)
- Onno C Meijer
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Sander Kooijman
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Kroon
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Elizabeth M Winter
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
6
|
Yoshimura M, Flynn BP, Kershaw YM, Zhao Z, Ueta Y, Lightman SL, Conway-Campbell BL. Phase-shifting the circadian glucocorticoid profile induces disordered feeding behaviour by dysregulating hypothalamic neuropeptide gene expression. Commun Biol 2023; 6:998. [PMID: 37775688 PMCID: PMC10541449 DOI: 10.1038/s42003-023-05347-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 09/12/2023] [Indexed: 10/01/2023] Open
Abstract
Here we demonstrate, in rodents, how the timing of feeding behaviour becomes disordered when circulating glucocorticoid rhythms are dissociated from lighting cues; a phenomenon most commonly associated with shift-work and transmeridian travel 'jetlag'. Adrenalectomized rats are infused with physiological patterns of corticosterone modelled on the endogenous adrenal secretory profile, either in-phase or out-of-phase with lighting cues. For the in-phase group, food intake is significantly greater during the rats' active period compared to their inactive period; a feeding pattern similar to adrenal-intact control rats. In contrast, the feeding pattern of the out-of-phase group is significantly dysregulated. Consistent with a direct hypothalamic modulation of feeding behaviour, this altered timing is accompanied by dysregulated timing of anorexigenic and orexigenic neuropeptide gene expression. For Neuropeptide Y (Npy), we report a glucocorticoid-dependent direct transcriptional regulation mechanism mediated by the glucocorticoid receptor (GR). Taken together, our data highlight the adverse behavioural outcomes that can arise when two circadian systems have anti-phasic cues, in this case impacting on the glucocorticoid-regulation of a process as fundamental to health as feeding behaviour. Our findings further highlight the need for development of rational approaches in the prevention of metabolic dysfunction in circadian-disrupting activities such as transmeridian travel and shift-work.
Collapse
Affiliation(s)
- Mitsuhiro Yoshimura
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
- Department of Physiology, University of Occupational and Environmental Health, Japan 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Benjamin P Flynn
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
| | - Yvonne M Kershaw
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
| | - Zidong Zhao
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
| | - Yoichi Ueta
- Department of Physiology, University of Occupational and Environmental Health, Japan 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Stafford L Lightman
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
| | - Becky L Conway-Campbell
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK.
| |
Collapse
|
7
|
Sarafoglou K, Merke DP, Reisch N, Claahsen-van der Grinten H, Falhammar H, Auchus RJ. Interpretation of Steroid Biomarkers in 21-Hydroxylase Deficiency and Their Use in Disease Management. J Clin Endocrinol Metab 2023; 108:2154-2175. [PMID: 36950738 PMCID: PMC10438890 DOI: 10.1210/clinem/dgad134] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/07/2023] [Indexed: 03/24/2023]
Abstract
The most common form of congenital adrenal hyperplasia is 21-hydroxylase deficiency (21OHD), which in the classic (severe) form occurs in roughly 1:16 000 newborns worldwide. Lifelong treatment consists of replacing cortisol and aldosterone deficiencies, and supraphysiological dosing schedules are typically employed to simultaneously attenuate production of adrenal-derived androgens. Glucocorticoid titration in 21OHD is challenging as it must balance the consequences of androgen excess vs those from chronic high glucocorticoid exposure, which are further complicated by interindividual variability in cortisol kinetics and glucocorticoid sensitivity. Clinical assessment and biochemical parameters are both used to guide therapy, but the specific purpose and goals of each biomarker vary with age and clinical context. Here we review the approach to medication titration for children and adults with classic 21OHD, with an emphasis on how to interpret adrenal biomarker values in guiding this process. In parallel, we illustrate how an understanding of the pathophysiologic and pharmacologic principles can be used to avoid and to correct complications of this disease and consequences of its management using existing treatment options.
Collapse
Affiliation(s)
- Kyriakie Sarafoglou
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Minnesota Medical School, Minneapolis, MN 55454, USA
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
| | - Deborah P Merke
- Department of Pediatrics, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Nicole Reisch
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, 80336 Munich, Germany
| | - Hedi Claahsen-van der Grinten
- Department of Pediatrics, Amalia Children's Hospital, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Henrik Falhammar
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE-17176, Stockholm, Sweden
- Department of Endocrinology, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Richard J Auchus
- Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
8
|
Koorneef LL, Viho EMG, Wahl LF, Meijer OC. Do Corticosteroid Receptor mRNA Levels Predict the Expression of Their Target Genes? J Endocr Soc 2022; 7:bvac188. [PMID: 36578881 PMCID: PMC9791178 DOI: 10.1210/jendso/bvac188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Indexed: 12/14/2022] Open
Abstract
The glucocorticoid stress hormones affect brain function via high-affinity mineralocorticoid receptors (MRs) and lower-affinity glucocorticoid receptors (GRs). MR and GR not only differ in affinity for ligands, but also have distinct, sometimes opposite, actions on neuronal excitability and other cellular and higher-order parameters related to cerebral function. GR and MR messenger RNA (mRNA) levels are often used as a proxy for the responsiveness to glucocorticoids, assuming proportionality between mRNA and protein levels. This may be especially relevant for the MR, which because of its high affinity is already largely occupied at low basal (trough) hormone levels. Here we explore how GR and MR mRNA levels are associated with the expression of a shared target gene, glucocorticoid-induced leucine zipper (GILZ, coded by Tsc22d3) with basal and elevated levels of corticosterone in male mice, using in situ hybridization. Depending on the hippocampal subfield and the corticosterone levels, mRNA levels of MR rather than GR mostly correlated with GILZ mRNA in the hippocampus and hypothalamus at the bulk tissue level. At the individual cell level, these correlations were much weaker. Using publicly available single-cell RNA sequencing data, we again observed that MR and GR mRNA levels were only weakly correlated with target gene expression in glutamatergic and GABAergic neurons. We conclude that MR mRNA levels can be limiting for receptor action, but many other cell-specific and region-specific factors ultimately determine corticosteroid receptor action. Altogether, our results argue for caution while interpreting the consequences of changed receptor expression for the response to glucocorticoids.
Collapse
Affiliation(s)
- Lisa L Koorneef
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Eva M G Viho
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Lucas F Wahl
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Onno C Meijer
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| |
Collapse
|
9
|
Srinivasan M, Walker C. Circadian Clock, Glucocorticoids and NF-κB Signaling in Neuroinflammation- Implicating Glucocorticoid Induced Leucine Zipper as a Molecular Link. ASN Neuro 2022; 14:17590914221120190. [PMID: 36317290 PMCID: PMC9629546 DOI: 10.1177/17590914221120190] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Inflammation including neuroinflammation is considered a protective response and is directed to repair, regenerate, and restore damaged tissues in the central nervous system. Persistent inflammation due to chronic stress, age related accrual of free radicals, subclinical infections or other factors lead to reduced survival and increased neuronal death. Circadian abnormalities secondary to altered sleep/wake cycles is one of the earliest signs of neurodegenerative diseases. Brain specific or global deficiency of core circadian trans-activator brain and muscle ARNT (Arylhydrocarbon Receptor Nuclear Translocator)-like protein 1 (BMAL1) or that of the transrepressor REV-ERBα, impaired neural function and cognitive performance in rodents. Consistently, transcripts of inflammatory cytokines and host immune responses have been shown to exhibit diurnal variation, in parallel with the disruption of the circadian rhythm. Glucocorticoids that exhibit both a circadian rhythm similar to that of the core clock transactivator BMAL1 and tissue specific ultradian rhythm are critical in the control of neuroinflammation and re-establishment of homeostasis. It is widely accepted that the glucocorticoids suppress nuclear factor-kappa B (NF-κB) mediated transactivation and suppress inflammation. Recent mechanistic elucidations suggest that the core clock components also modulate NF-κB mediated transactivation in the brain and peripheral tissues. In this review we discuss evidence for interactions between the circadian clock components, glucocorticoids and NF-κB signaling responses in the brain and propose glucocorticoid induced leucine zipper (GILZ) encoded by Tsc22d3, as a molecular link that connect all three pathways in the maintenance of CNS homeostasis as well as in the pathogenesis of neuroinflammation-neurodegeneration.
Collapse
Affiliation(s)
- Mythily Srinivasan
- Department of Oral Pathology, Medicine and Radiology, Indiana University School of Dentistry, Indiana University Purdue University at Indianapolis, Indiana, USA,Provaidya LLC, Indiana Center for Biomedical Innovation, Indianapolis, Indiana, USA,Mythily Srinivasan, Oral Pathology, Radiology and Medicine, Indiana University School of Dentistry, Indianapolis, Indiana, United States; Provaidya LLC, Indiana Center for Biomedical Innovation, 1800 North Capitol Av, Indianapolis, IN 46202, United States.
;
| | - Chandler Walker
- Department of Oral Pathology, Medicine and Radiology, Indiana University School of Dentistry, Indiana University Purdue University at Indianapolis, Indiana, USA,Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indiana University Purdue University at Indianapolis, Indiana, USA
| |
Collapse
|
10
|
Giannos P, Prokopidis K, Forbes SC, Celoch K, Candow DG, Tartar JL. Gene Expression Changes of Murine Cortex Homeostasis in Response to Sleep Deprivation Hint Dysregulated Aging-like Transcriptional Responses. Brain Sci 2022; 12:825. [PMID: 35884632 PMCID: PMC9313387 DOI: 10.3390/brainsci12070825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 12/04/2022] Open
Abstract
Sleep deprivation leads to the deterioration in the physiological functioning of the brain, cognitive decline, and many neurodegenerative diseases, all of which progress with advancing age. Sleep insufficiency and impairments in cognitive function are characterized by progressive neuronal losses in the cerebral cortex. In this study, we analyze gene expression profiles following sleep-deprived murine models and circadian matched controls to identify genes that might underlie cortical homeostasis in response to sleep deprivation. Screening of the literature resulted in three murine (Mus musculus) gene expression datasets (GSE6514, GSE78215, and GSE33491) that included cortical tissue biopsies from mice that are sleep deprived for 6 h (n = 15) and from circadian controls that are left undisturbed (n = 15). Cortical differentially expressed genes are used to construct a network of encoded proteins that are ranked based on their interactome according to 11 topological algorithms. The analysis revealed three genes-NFKBIA, EZR, and SGK1-which exhibited the highest multi-algorithmic topological significance. These genes are strong markers of increased brain inflammation, cytoskeletal aberrations, and glucocorticoid resistance, changes that imply aging-like transcriptional responses during sleep deprivation in the murine cortex. Their potential role as candidate markers of local homeostatic response to sleep loss in the murine cortex warrants further experimental validation.
Collapse
Affiliation(s)
- Panagiotis Giannos
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, South Kensington, London SW7 2AZ, UK
- Society of Meta-Research and Biomedical Innovation, London W12 0BZ, UK;
| | - Konstantinos Prokopidis
- Society of Meta-Research and Biomedical Innovation, London W12 0BZ, UK;
- Department of Musculoskeletal Biology, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L69 3BX, UK
| | - Scott C. Forbes
- Department of Physical Education Studies, Faculty of Education, Brandon University, Brandon, MB R7A 6A9, Canada;
| | - Kamil Celoch
- Department of Psychology and Neuroscience, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (K.C.); (J.L.T.)
| | - Darren G. Candow
- Faculty of Kinesiology and Health Studies, University of Regina, Regina, SK S4S 0A2, Canada;
| | - Jaime L. Tartar
- Department of Psychology and Neuroscience, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (K.C.); (J.L.T.)
| |
Collapse
|
11
|
Viho EMG, Buurstede JC, Berkhout JB, Mahfouz A, Meijer OC. Cell type specificity of glucocorticoid signaling in the adult mouse hippocampus. J Neuroendocrinol 2022; 34:e13072. [PMID: 34939259 PMCID: PMC9286676 DOI: 10.1111/jne.13072] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/14/2021] [Accepted: 11/18/2021] [Indexed: 12/14/2022]
Abstract
Glucocorticoid stress hormones are powerful modulators of brain function and can affect mood and cognitive processes. The hippocampus is a prominent glucocorticoid target and expresses both the glucocorticoid receptor (GR: Nr3c1) and the mineralocorticoid receptor (MR: Nr3c2). These nuclear steroid receptors act as ligand-dependent transcription factors. Transcriptional effects of glucocorticoids have often been deduced from bulk mRNA measurements or spatially informed individual gene expression. However, only sparse data exists allowing insights on glucocorticoid-driven gene transcription at the cell type level. Here, we used publicly available single-cell RNA sequencing data to assess the cell-type specificity of GR and MR signaling in the adult mouse hippocampus. The data confirmed that Nr3c1 and Nr3c2 expression differs across neuronal and non-neuronal cell populations. We analyzed co-expression with sex hormones receptors, transcriptional coregulators, and receptors for neurotransmitters and neuropeptides. Our results provide insights in the cellular basis of previous bulk mRNA results and allow the formulation of more defined hypotheses on the effects of glucocorticoids on hippocampal function.
Collapse
Affiliation(s)
- Eva M. G. Viho
- Division of EndocrinologyDepartment of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Jacobus C. Buurstede
- Division of EndocrinologyDepartment of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Jari B. Berkhout
- Division of EndocrinologyDepartment of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
| | - Ahmed Mahfouz
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
- Delft Bioinformatics LaboratoryDelft University of TechnologyDelftThe Netherlands
- Leiden Computational Biology CenterLeiden University Medical CenterLeidenThe Netherlands
| | - Onno C. Meijer
- Division of EndocrinologyDepartment of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
12
|
Danan D, Todder D, Zohar J, Cohen H. Is PTSD-Phenotype Associated with HPA-Axis Sensitivity? Feedback Inhibition and Other Modulating Factors of Glucocorticoid Signaling Dynamics. Int J Mol Sci 2021; 22:ijms22116050. [PMID: 34205191 PMCID: PMC8200046 DOI: 10.3390/ijms22116050] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 01/12/2023] Open
Abstract
Previously, we found that basal corticosterone pulsatility significantly impacts the vulnerability for developing post-traumatic stress disorder (PTSD). Rats that exhibited PTSD-phenotype were characterized by blunted basal corticosterone pulsatility amplitude and a blunted corticosterone response to a stressor. This study sought to identify the mechanisms underlining both the loss of pulsatility and differences in downstream responses. Serial blood samples were collected manually via jugular vein cannula at 10-min intervals to evaluate suppression of corticosterone following methylprednisolone administration. The rats were exposed to predator scent stress (PSS) after 24 h, and behavioral responses were assessed 7 days post-exposure for retrospective classification into behavioral response groups. Brains were harvested for measurements of the glucocorticoid receptor, mineralocorticoid receptor, FK506-binding protein-51 and arginine vasopressin in specific brain regions to assess changes in hypothalamus–pituitary–adrenal axis (HPA) regulating factors. Methylprednisolone produced greater suppression of corticosterone in the PTSD-phenotype group. During the suppression, the PTSD-phenotype rats showed a significantly more pronounced pulsatile activity. In addition, the PTSD-phenotype group showed distinct changes in the ventral and dorsal CA1, dentate gyrus as well as in the paraventricular nucleus and supra-optic nucleus. These results demonstrate a pre-trauma vulnerability state that is characterized by an over-reactivity of the HPA and changes in its regulating factors.
Collapse
Affiliation(s)
- Dor Danan
- Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, Ministry of Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84170, Israel; (D.D.); (D.T.)
| | - Doron Todder
- Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, Ministry of Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84170, Israel; (D.D.); (D.T.)
| | - Joseph Zohar
- Post-Trauma Center, Sheba Medical Center, Tel Aviv 52621, Israel;
| | - Hagit Cohen
- Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, Ministry of Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84170, Israel; (D.D.); (D.T.)
- Correspondence: ; Tel.: +972-544-369106
| |
Collapse
|
13
|
Abstract
Learning to respond appropriately to one's surrounding environment is fundamental to survival. Importantly, however, individuals vary in how they respond to cues in the environment and this variation may be a key determinant of psychopathology. The ability of seemingly neutral cues to promote maladaptive behavior is a hallmark of several psychiatric disorders including, substance use disorder, post-traumatic stress disorder, eating disorders and obsessive-compulsive disorder. Thus, it is important to uncover the neural mechanisms by which such cues are able to attain inordinate control and promote psychopathological behavior. Here, we suggest that glucocorticoids play a critical role in this process. Glucocorticoids are primarily recognized as the main hormone secreted in response to stress but are known to exert their effects across the body and the brain, and to affect learning and memory, cognition and reward-related behaviors, among other things. Here we speculate that glucocorticoids act to facilitate a dopamine-dependent form of cue-reward learning that appears to be relevant to a number of psychiatric conditions. Specifically, we propose to utilize the sign-tracker/goal-tracker animal model as a means to capture individual variation in stimulus-reward learning and to isolate the role of glucocorticoid-dopamine interactions in mediating these individual differences. It is hoped that this framework will lead to the discovery of novel mechanisms that contribute to complex neuropsychiatric disorders and their comorbidity.
Collapse
Affiliation(s)
- Sofia A. Lopez
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Shelly B. Flagel
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
14
|
Bläuer M, Sand J, Laukkarinen J. Regulation of p38 MAPK and glucocorticoid receptor activation by hydrocortisone in mono-and co-cultured pancreatic acinar and stellate cells. Pancreatology 2021; 21:384-389. [PMID: 33454208 DOI: 10.1016/j.pan.2020.12.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/17/2020] [Accepted: 12/29/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND/OBJECTIVES Acute pancreatitis develops as an inflammatory response to pancreatic tissue injury. Postoperative pancreatitis has recently been associated with increased occurrence of complications. Activation of the mitogen-activated protein kinase p38 (p38 MAPK) pathway occurs early in acute pancreatitis and its inhibition has been suggested to alleviate pancreatic inflammation. Glucocorticoids are potent anti-inflammatory steroids whose use in the management of acute pancreatitis remains controversial. Our aim was to examine the effect of crosstalk between pancreatic acinar cells (PACs) and stellate cells (PSCs) on p38 MAPK and glucocorticoid receptor (GR) activation and to assess the impact of hydrocortisone on these events. METHODS The long-term co-culture setting for mouse PACs and PSCs developed in our laboratory was used. Parallel 4d mono- and co-cultures with or without 10 nM hydrocortisone were performed followed by immunocytochemical analysis of nuclear GR and phospho-p38 MAPK (pp38 MAPK). RESULTS Hydrocortisone inhibited pp38 MAPK up-regulation evoked by co-culture in PACs and PSCs and increased nuclear translocation of GR in PAC monocultures and in co-cultured PACs and PSCs. In PSC monocultures and co-cultured PACs, ligand-independent expression of nuclear GR was observed. In the former no change in nuclear GR but a significant decrease in total GR as analyzed by Western blot was caused by hydrocortisone. CONCLUSIONS Cellular microenvironment plays a significant role on p38 MAPK and GR activation in PACs and PSCs. Hydrocortisone is an effective means to inhibit p38 MAPK activation in PACs and PSCs. Both ligand-dependent and -independent regulatory roles for GR are suggested in the exocrine pancreas.
Collapse
Affiliation(s)
- Merja Bläuer
- Tampere Pancreas Laboratory and Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Juhani Sand
- Tampere Pancreas Laboratory and Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Johanna Laukkarinen
- Tampere Pancreas Laboratory and Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
15
|
HUZARD D, RAPPENEAU V, MEIJER OC, TOUMA C, ARANGO-LIEVANO M, GARABEDIAN MJ, JEANNETEAU F. Experience and activity-dependent control of glucocorticoid receptors during the stress response in large-scale brain networks. Stress 2021; 24:130-153. [PMID: 32755268 PMCID: PMC7907260 DOI: 10.1080/10253890.2020.1806226] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The diversity of actions of the glucocorticoid stress hormones among individuals and within organs, tissues and cells is shaped by age, gender, genetics, metabolism, and the quantity of exposure. However, such factors cannot explain the heterogeneity of responses in the brain within cells of the same lineage, or similar tissue environment, or in the same individual. Here, we argue that the stress response is continuously updated by synchronized neural activity on large-scale brain networks. This occurs at the molecular, cellular and behavioral levels by crosstalk communication between activity-dependent and glucocorticoid signaling pathways, which updates the diversity of responses based on prior experience. Such a Bayesian process determines adaptation to the demands of the body and external world. We propose a framework for understanding how the diversity of glucocorticoid actions throughout brain networks is essential for supporting optimal health, while its disruption may contribute to the pathophysiology of stress-related disorders, such as major depression, and resistance to therapeutic treatments.
Collapse
Affiliation(s)
- Damien HUZARD
- Department of Neuroscience and Physiology, University of Montpellier, CNRS, INSERM, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Virginie RAPPENEAU
- Department of Behavioural Biology, University of Osnabrück, Osnabrück, Germany
| | - Onno C. MEIJER
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Chadi TOUMA
- Department of Behavioural Biology, University of Osnabrück, Osnabrück, Germany
| | - Margarita ARANGO-LIEVANO
- Department of Neuroscience and Physiology, University of Montpellier, CNRS, INSERM, Institut de Génomique Fonctionnelle, Montpellier, France
| | | | - Freddy JEANNETEAU
- Department of Neuroscience and Physiology, University of Montpellier, CNRS, INSERM, Institut de Génomique Fonctionnelle, Montpellier, France
- Corresponding author:
| |
Collapse
|
16
|
Herman JP, Nawreen N, Smail MA, Cotella EM. Brain mechanisms of HPA axis regulation: neurocircuitry and feedback in context Richard Kvetnansky lecture. Stress 2020; 23:617-632. [PMID: 33345670 PMCID: PMC8034599 DOI: 10.1080/10253890.2020.1859475] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 11/28/2020] [Indexed: 12/11/2022] Open
Abstract
Regulation of stress reactivity is a fundamental priority of all organisms. Stress responses are critical for survival, yet can also cause physical and psychological damage. This review provides a synopsis of brain mechanisms designed to control physiological responses to stress, focusing primarily on glucocorticoid secretion via the hypothalamo-pituitary-adrenocortical (HPA) axis. The literature provides strong support for multi-faceted control of HPA axis responses, involving both direct and indirect actions at paraventricular nucleus (PVN) corticotropin releasing hormone neurons driving the secretory cascade. The PVN is directly excited by afferents from brainstem and hypothalamic circuits, likely relaying information on homeostatic challenge. Amygdala subnuclei drive HPA axis responses indirectly via disinhibition, mediated by GABAergic relays onto PVN-projecting neurons in the hypothalamus and bed nucleus of the stria terminalis (BST). Inhibition of stressor-evoked HPA axis responses is mediated by an elaborate network of glucocorticoid receptor (GR)-containing circuits, providing a distributed negative feedback signal that inhibits PVN neurons. Prefrontal and hippocampal neurons play a major role in HPA axis inhibition, again mediated by hypothalamic and BST GABAergic relays to the PVN. The complexity of the regulatory process suggests that information on stressors is integrated across functional disparate brain circuits prior to accessing the PVN, with regions such as the BST in prime position to relay contextual information provided by these sources into appropriate HPA activation. Dysregulation of the HPA in disease is likely a product of inappropriate checks and balances between excitatory and inhibitory inputs ultimately impacting PVN output.
Collapse
Affiliation(s)
- James P Herman
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, USA
- Cincinnati Veterans Administration Medical Center, Cincinnati, OH, USA
| | - Nawshaba Nawreen
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Marissa A Smail
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Evelin M Cotella
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
- Cincinnati Veterans Administration Medical Center, Cincinnati, OH, USA
| |
Collapse
|
17
|
Dattilo V, Amato R, Perrotti N, Gennarelli M. The Emerging Role of SGK1 (Serum- and Glucocorticoid-Regulated Kinase 1) in Major Depressive Disorder: Hypothesis and Mechanisms. Front Genet 2020; 11:826. [PMID: 32849818 PMCID: PMC7419621 DOI: 10.3389/fgene.2020.00826] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/09/2020] [Indexed: 12/28/2022] Open
Abstract
Major depressive disorder (MDD) is a heterogeneous psychiatric disease characterized by persistent low mood, diminished interests, and impaired cognitive and social functions. The multifactorial etiology of MDD is still largely unknown because of the complex genetic and environmental interactions involved. Therefore, no established mechanism can explain all the aspects of the disease. In this light, an extensive research about the pathophysiology of MDD has been carried out. Several pathogenic hypotheses, such as monoamines deficiency and neurobiological alterations in the stress-responsive system, including the hypothalamic-pituitary-adrenal (HPA) axis and the immune system, have been proposed for MDD. Over time, remarkable studies, mainly on preclinical rodent models, linked the serum- and glucocorticoid-regulated kinase 1 (SGK1) to the main features of MDD. SGK1 is a serine/threonine kinase belonging to the AGK Kinase family. SGK1 is ubiquitously expressed, which plays a pivotal role in the hormonal regulation of several ion channels, carriers, pumps, and transcription factors or regulators. SGK1 expression is modulated by cell stress and hormones, including gluco- and mineralocorticoids. Compelling evidence suggests that increased SGK1 expression or function is related to the pathogenic stress hypothesis of major depression. Therefore, the first part of the present review highlights the putative role of SGK1 as a critical mediator in the dysregulation of the HPA axis, observed under chronic stress conditions, and its controversial role in the neuroinflammation as well. The second part depicts the negative regulation exerted by SGK1 in the expression of both the brain-derived neurotrophic factor (BDNF) and the vascular endothelial growth factor (VEGF), resulting in an anti-neurogenic activity. Finally, the review focuses on the antidepressant-like effects of anti-oxidative nutraceuticals in several preclinical model of depression, resulting from the restoration of the physiological expression and/or activity of SGK1, which leads to an increase in neurogenesis. In summary, the purpose of this review is a systematic analysis of literature depicting SGK1 as molecular junction of the complex mechanisms underlying the MDD in an effort to suggest the kinase as a potential biomarker and strategic target in modern molecular antidepressant therapy.
Collapse
Affiliation(s)
- Vincenzo Dattilo
- Genetic Unit, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Rosario Amato
- Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy.,Medical Genetics Unit, Mater Domini University Hospital, Catanzaro, Italy
| | - Nicola Perrotti
- Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy.,Medical Genetics Unit, Mater Domini University Hospital, Catanzaro, Italy
| | - Massimo Gennarelli
- Genetic Unit, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
18
|
Schouten M, Bielefeld P, Garcia-Corzo L, Passchier EMJ, Gradari S, Jungenitz T, Pons-Espinal M, Gebara E, Martín-Suárez S, Lucassen PJ, De Vries HE, Trejo JL, Schwarzacher SW, De Pietri Tonelli D, Toni N, Mira H, Encinas JM, Fitzsimons CP. Circadian glucocorticoid oscillations preserve a population of adult hippocampal neural stem cells in the aging brain. Mol Psychiatry 2020; 25:1382-1405. [PMID: 31222184 PMCID: PMC7303016 DOI: 10.1038/s41380-019-0440-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 04/09/2019] [Accepted: 04/29/2019] [Indexed: 12/17/2022]
Abstract
A decrease in adult hippocampal neurogenesis has been linked to age-related cognitive impairment. However, the mechanisms involved in this age-related reduction remain elusive. Glucocorticoid hormones (GC) are important regulators of neural stem/precursor cells (NSPC) proliferation. GC are released from the adrenal glands in ultradian secretory pulses that generate characteristic circadian oscillations. Here, we investigated the hypothesis that GC oscillations prevent NSPC activation and preserve a quiescent NSPC pool in the aging hippocampus. We found that hippocampal NSPC populations lacking expression of the glucocorticoid receptor (GR) decayed exponentially with age, while GR-positive populations decayed linearly and predominated in the hippocampus from middle age onwards. Importantly, GC oscillations controlled NSPC activation and GR knockdown reactivated NSPC proliferation in aged mice. When modeled in primary hippocampal NSPC cultures, GC oscillations control cell cycle progression and induce specific genome-wide DNA methylation profiles. GC oscillations induced lasting changes in the methylation state of a group of gene promoters associated with cell cycle regulation and the canonical Wnt signaling pathway. Finally, in a mouse model of accelerated aging, we show that disruption of GC oscillations induces lasting changes in dendritic complexity, spine numbers and morphology of newborn granule neurons. Together, these results indicate that GC oscillations preserve a population of GR-expressing NSPC during aging, preventing their activation possibly by epigenetic programming through methylation of specific gene promoters. Our observations suggest a novel mechanism mediated by GC that controls NSPC proliferation and preserves a dormant NSPC pool, possibly contributing to a neuroplasticity reserve in the aging brain.
Collapse
Affiliation(s)
- M Schouten
- Neuroscience Collaboration, Swammerdam Institute for Life Sciences, Faculty of Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - P Bielefeld
- Neuroscience Collaboration, Swammerdam Institute for Life Sciences, Faculty of Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - L Garcia-Corzo
- Biomedicine Institute of Valencia (IBV), Consejo Superior de Investigaciones Científicas (CSIC), Valencia, Spain
| | - E M J Passchier
- Neuroscience Collaboration, Swammerdam Institute for Life Sciences, Faculty of Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - S Gradari
- Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - T Jungenitz
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - M Pons-Espinal
- Neurobiology of miRNA Lab, Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genoa, Italy
| | - E Gebara
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | | | - P J Lucassen
- Neuroscience Collaboration, Swammerdam Institute for Life Sciences, Faculty of Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - H E De Vries
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - J L Trejo
- Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - S W Schwarzacher
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - D De Pietri Tonelli
- Neurobiology of miRNA Lab, Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genoa, Italy
| | - N Toni
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - H Mira
- Biomedicine Institute of Valencia (IBV), Consejo Superior de Investigaciones Científicas (CSIC), Valencia, Spain
| | - J M Encinas
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Ikerbasque, The Basque Foundation for Science, Bilbao, Spain
- University of the Basque Country (UPV/EHU), Leioa, Spain
| | - C P Fitzsimons
- Neuroscience Collaboration, Swammerdam Institute for Life Sciences, Faculty of Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
19
|
van der Sluis RJ, Hoekstra M. Glucocorticoids are active players and therapeutic targets in atherosclerotic cardiovascular disease. Mol Cell Endocrinol 2020; 504:110728. [PMID: 31968221 DOI: 10.1016/j.mce.2020.110728] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/19/2019] [Accepted: 01/17/2020] [Indexed: 02/07/2023]
Abstract
Adrenal-derived glucocorticoids mediate the physiological response to stress. Chronic disturbances in glucocorticoid homeostasis, i.e. in Addison's and Cushing's disease patients, predispose to the development of atherosclerotic cardiovascular disease. Here we review preclinical and clinical findings regarding the relation between changes in plasma glucocorticoid levels and the atherosclerosis extent. It appears that, although the altered glucocorticoid function can in most cases be restored in the different patient groups, current therapies do not necessarily reverse the associated risk for atherosclerotic cardiovascular disease. In our opinion much attention should therefore be given to the development of a Cushing's disease mouse model that can (1) effectively replicate the effect of hypercortisolemia on atherosclerosis outcome observed in humans and (2) be used to investigate, in a preclinical setting, the relative impact on atherosclerosis susceptibility of already available (e.g. metyrapone) and potentially novel (i.e. SR-BI activity modulators) therapeutic agents that target the adrenal glucocorticoid output.
Collapse
Affiliation(s)
- Ronald J van der Sluis
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, 2333CC, Leiden, the Netherlands
| | - Menno Hoekstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, 2333CC, Leiden, the Netherlands.
| |
Collapse
|
20
|
Bonapersona V, Damsteegt R, Adams ML, van Weert LTCM, Meijer OC, Joëls M, Sarabdjitsingh RA. Sex-Dependent Modulation of Acute Stress Reactivity After Early Life Stress in Mice: Relevance of Mineralocorticoid Receptor Expression. Front Behav Neurosci 2019; 13:181. [PMID: 31440147 PMCID: PMC6693524 DOI: 10.3389/fnbeh.2019.00181] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 07/19/2019] [Indexed: 01/11/2023] Open
Abstract
Early life stress (ELS) is considered a major risk factor for developing psychopathology. Increasing evidence points towards sex-dependent dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis as a contributing mechanism. Additionally, clinical studies suggest that the mineralocorticoid receptor (MR) may further confer genetic vulnerability/resilience on a background of ELS. The link between ELS, sex and the HPA axis and how this interacts with MR genotype is understudied, yet important to understand vulnerability/resilience to stress. We used the early life-limited nesting and bedding model to test the effect of ELS on HPA properties in adult female and male mice carrying a forebrain-specific heterozygous knockout for MR. Basal HPA axis activity was measured by circadian peak and nadir corticosterone levels, in addition to body weight and weight of stress-sensitive tissues. HPA axis reactivity was assessed by mapping corticosterone levels after 10 min immobilization. Additionally, we measured the effects of ELS on steroid receptor [MR and glucocorticoid receptor (GR)] levels in the dorsal hippocampus and medial prefrontal cortex (mPFC) with western blot. Finally, behavioral reactivity towards a novel environment was measured as a proxy for anxiety-like behavior. Results show that HPA axis activity under rest conditions was not affected by ELS. HPA axis reactivity after immobilization was decreased by ELS in females and increased, at trend-level in males. This effect in females was further exacerbated by low expression of the MR. We also observed a sex*ELS interaction regarding MR and GR expression in the dorsal hippocampus, with a significant upregulation of MR in males only. The sex-dependent interaction with ELS was not reflected in the behavioral response to novel environment and time spent in a sheltered compartment. We did find increased locomotor activity in all groups after a history of ELS, which attenuated after 4 h in males but not females regardless of condition. Our findings support that ELS alters HPA axis functioning sex-dependently. Genetic predisposition to low MR function may render females more susceptible to the harmful effect of ELS whereas in males low MR function promotes resilience. We propose that this model may be a useful tool to investigate the underlying mechanisms of sex-dependent and genetic vulnerability/resilience to stress-related psychopathology.
Collapse
Affiliation(s)
- Valeria Bonapersona
- Department of Translational Neuroscience, UMC Utrecht Brain Center, Utrecht University, Utrecht, Netherlands
| | - Ruth Damsteegt
- Department of Translational Neuroscience, UMC Utrecht Brain Center, Utrecht University, Utrecht, Netherlands
| | - Mirjam L Adams
- Department of Translational Neuroscience, UMC Utrecht Brain Center, Utrecht University, Utrecht, Netherlands
| | - Lisa T C M van Weert
- Department of Internal Medicine, Leiden University Medical Center, Division of Endocrinology, Leiden, Netherlands
| | - Onno C Meijer
- Department of Internal Medicine, Leiden University Medical Center, Division of Endocrinology, Leiden, Netherlands
| | - Marian Joëls
- Department of Translational Neuroscience, UMC Utrecht Brain Center, Utrecht University, Utrecht, Netherlands.,University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Ratna Angela Sarabdjitsingh
- Department of Translational Neuroscience, UMC Utrecht Brain Center, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
21
|
de Kloet ER, de Kloet SF, de Kloet CS, de Kloet AD. Top-down and bottom-up control of stress-coping. J Neuroendocrinol 2019; 31:e12675. [PMID: 30578574 PMCID: PMC6519262 DOI: 10.1111/jne.12675] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 12/17/2022]
Abstract
In this 30th anniversary issue review, we focus on the glucocorticoid modulation of limbic-prefrontocortical circuitry during stress-coping. This action of the stress hormone is mediated by mineralocorticoid receptors (MRs) and glucocorticoid receptors (GRs) that are co-expressed abundantly in these higher brain regions. Via both receptor types, the glucocorticoids demonstrate, in various contexts, rapid nongenomic and slower genomic actions that coordinate consecutive stages of information processing. MR-mediated action optimises stress-coping, whereas, in a complementary fashion, the memory storage of the selected coping strategy is promoted via GR. We highlight the involvement of adipose tissue in the allocation of energy resources to central regulation of stress reactions, point to still poorly understood neuronal ensembles in the prefrontal cortex that underlie cognitive flexibility critical for effective coping, and evaluate the role of cortisol as a pleiotropic regulator in vulnerability to, and treatment of, trauma-related psychiatric disorders.
Collapse
Affiliation(s)
- Edo R. de Kloet
- Division of EndocrinologyDepartment of MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Sybren F. de Kloet
- Department of Integrative NeurophysiologyCenter for Neurogenomics and Cognitive ResearchVU‐University of AmsterdamAmsterdamThe Netherlands
| | | | - Annette D. de Kloet
- Department of Physiology and Functional GenomicsUniversity of FloridaGainesvilleFlorida
| |
Collapse
|
22
|
Brain Mineralocorticoid Receptors and Resilience to Stress. VITAMINS AND HORMONES 2019; 109:341-359. [DOI: 10.1016/bs.vh.2018.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
|
23
|
de Kloet ER, Meijer OC, de Nicola AF, de Rijk RH, Joëls M. Importance of the brain corticosteroid receptor balance in metaplasticity, cognitive performance and neuro-inflammation. Front Neuroendocrinol 2018; 49:124-145. [PMID: 29428549 DOI: 10.1016/j.yfrne.2018.02.003] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/25/2018] [Accepted: 02/07/2018] [Indexed: 01/14/2023]
Abstract
Bruce McEwen's discovery of receptors for corticosterone in the rat hippocampus introduced higher brain circuits in the neuroendocrinology of stress. Subsequently, these receptors were identified as mineralocorticoid receptors (MRs) that are involved in appraisal processes, choice of coping style, encoding and retrieval. The MR-mediated actions on cognition are complemented by slower actions via glucocorticoid receptors (GRs) on contextualization, rationalization and memory storage of the experience. These sequential phases in cognitive performance depend on synaptic metaplasticity that is regulated by coordinate MR- and GR activation. The receptor activation includes recruitment of coregulators and transcription factors as determinants of context-dependent specificity in steroid action; they can be modulated by genetic variation and (early) experience. Interestingly, inflammatory responses to damage seem to be governed by a similarly balanced MR:GR-mediated action as the initiating, terminating and priming mechanisms involved in stress-adaptation. We conclude with five questions challenging the MR:GR balance hypothesis.
Collapse
Affiliation(s)
- E R de Kloet
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| | - O C Meijer
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| | - A F de Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina.
| | - R H de Rijk
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands & Department of Clinical Psychology, Leiden University, The Netherlands.
| | - M Joëls
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands; University of Groningen, University Medical Center Groningen, The Netherlands.
| |
Collapse
|
24
|
Blunted basal corticosterone pulsatility predicts post-exposure susceptibility to PTSD phenotype in rats. Psychoneuroendocrinology 2018; 87:35-42. [PMID: 29035710 DOI: 10.1016/j.psyneuen.2017.09.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/24/2017] [Accepted: 09/28/2017] [Indexed: 12/24/2022]
Abstract
The basal activity of the hypothalamic-pituitary-adrenal axis is highly dynamic and is characterized by both circadian and ultradian (pulsatile) patterns of hormone secretion. Pulsatility of glucocorticoids has been determined to be critical for optimal transcriptional, neuroendocrine, and behavioral responses. We used an animal model of post-traumatic stress disorder (PTSD) to assess whether stress-induced impairment of behavioral responses is correlated with aberrant secretion of corticosterone. Serial blood samples were collected manually via the jugular vein cannula during the light-(inactive)-phase in conscious male rats at 20-min intervals for a period of 5h before and 6.5h after exposure to predator scent stress. The outcome measures included behavior in an elevated plus-maze and acoustic startle response 7days after exposure. Individual animals were retrospectively classified as having "extreme", "partial", or "minimal" behavioral responses according to pre-set cut-off criteria for behavioral response patterns. Corticosterone secretion patterns were analyzed retrospectively. Under basal conditions, the amplitude of ultradian oscillations of corticosterone levels, rather than the mean corticosterone level or the frequency of corticosterone pulsatility, was significantly reduced in individuals who displayed PTSD-phenotype 8days later. In addition, extreme disruption of behavior on day 8 post-exposure was also characterized by a blunting of corticosterone response to the stressor. Animals with behavior that was only partially affected or unaffected displayed none of the above changes. Blunted basal corticosterone pulse amplitude is a pre-existing susceptibility or risk factor for PTSD, which originates from prior (life) experiences and may therefore predict post-exposure PTSD-phenotype in rats.
Collapse
|
25
|
den Boon FS, Sarabdjitsingh RA. Circadian and ultradian patterns of HPA-axis activity in rodents: Significance for brain functionality. Best Pract Res Clin Endocrinol Metab 2017; 31:445-457. [PMID: 29223280 DOI: 10.1016/j.beem.2017.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The hypothalamo-pituitary-adrenal (HPA) axis comprises interactions between the hypothalamus, the pituitary and the adrenal glands and its activation results in the release of corticosteroid hormones. Corticosteroids are secreted from the adrenal gland in a distinct 24-h circadian rhythm overarching an ultradian rhythm, which consists of hourly corticosteroid pulses exposing target tissues to rapidly changing steroid levels. On top of these rhythms surges can take place after stress. HPA-axis rhythms promote adaptation to predictable (i.e. the earth's rotation) and unpredictable (i.e. stressors) changes in environmental factors. Two steroid hormone receptors, the mineralocorticoid receptor (MR) and the glucocorticoid receptor (GR), are activated by corticosteroids and mediate effects at fast and slow timescales on e.g. glucose availability, gene transcription and synaptic plasticity. The current review discusses the origin of the circadian and ultradian corticosteroid rhythms and their relevance for gene regulation, neuroendocrine and physiological responses to stress and the involvement in the maintenance of brain functionality in rodents.
Collapse
Affiliation(s)
- Femke S den Boon
- Dept. Translational Neuroscience, Brain Center Rudolf Magnus, UMC Utrecht, The Netherlands
| | | |
Collapse
|
26
|
Hinds LR, Chun LE, Woodruff ER, Christensen JA, Hartsock MJ, Spencer RL. Dynamic glucocorticoid-dependent regulation of Sgk1 expression in oligodendrocytes of adult male rat brain by acute stress and time of day. PLoS One 2017; 12:e0175075. [PMID: 28376115 PMCID: PMC5380358 DOI: 10.1371/journal.pone.0175075] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/19/2017] [Indexed: 01/08/2023] Open
Abstract
Recent studies support plasticity in adult brain white matter structure and myelination in response to various experiential factors. One possible contributor to this plasticity may be activity-dependent modulation of serum- and glucocorticoid-inducible kinase 1 (Sgk1) expression in oligodendrocytes. We examined whether Sgk1 expression in adult rat brain white matter is increased by acute stress-induced elevations in endogenous corticosterone and whether it fluctuates with diurnal variations in corticosterone. We observed rapid increases (within 30 min) in Sgk1 mRNA in the corpus callosum in response to acute stress, as well as large increases at the beginning of the rat's active period (the time of peak corticosterone secretion). These increases were absent in adrenalectomized rats. Corticosterone treatment of adrenalectomized rats also rapidly increased corpus callosum Sgk1 mRNA. The majority of Sgk1 mRNA in corpus callosum was co-localized with myelin basic protein mRNA, suggesting that mature oligodendrocytes respond dynamically to acute stress and circadian rhythms. The regulation of Sgk1 expression by acute stress and time of day was selective for white matter, with limited alteration of Sgk1 expression by these factors in hippocampus and somatosensory cortex. These results indicate a unique sensitivity of oligodendrocyte Sgk1 expression to activity-dependent fluctuations in corticosterone hormone secretion, and raises the prospect that hypothalamic-pituitary-adrenal axis dysregulation or glucocorticoid pharmacotherapy may compromise the normal activity-dependent interactions between oligodendrocytes and neurons.
Collapse
Affiliation(s)
- Laura R. Hinds
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Lauren E. Chun
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Elizabeth R. Woodruff
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Jennifer A. Christensen
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Matthew J. Hartsock
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Robert L. Spencer
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, United States of America
| |
Collapse
|
27
|
Chen X, Gianferante D, Hanlin L, Fiksdal A, Breines JG, Thoma MV, Rohleder N. HPA-axis and inflammatory reactivity to acute stress is related with basal HPA-axis activity. Psychoneuroendocrinology 2017; 78:168-176. [PMID: 28209543 PMCID: PMC5375039 DOI: 10.1016/j.psyneuen.2017.01.035] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 01/17/2017] [Accepted: 01/28/2017] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Inflammation is drawing attention as pathway between psychosocial stress and health, and basal HPA axis activity has been suggested to exert a consistent regulatory influence on peripheral inflammation. Here we studied the relationship between basal HPA axis activity and inflammatory and HPA axis acute stress reactivity. METHODS We recruited 48 healthy individuals and collected saliva for diurnal cortisol sampling at 6 points. Participants were previously exposed to the Trier Social Stress Test (TSST) on two consecutive days. Plasma interleukin-6 (IL-6) and salivary cortisol reactivity to acute stress were measured, and their relationships with basal HPA axis activity were analyzed. RESULTS Steeper cortisol awakening response (CAR) linear increase was related with stronger cortisol stress reactivity (γ=0.015; p=0.042) and marginally significantly with greater habituation (γ=0.01; p=0.066). Greater curvilinearity of CAR was related with stronger cortisol reactivity (γ=-0.014; p=0.021) and greater cortisol habituation (γ=-0.011; p=0.006). Steeper daily linear decline was related with significant or marginally significantly stronger cortisol and IL-6 reactivity (cortisol: γ=-0.0004; p=0.06; IL-6: γ=-0.028; p=0.031) and greater habituation (cortisol: γ=-0.002; p=0.009, IL-6: γ=-0.015; p=0.033). Greater curvilinearity of daily decline was related with stronger IL-6 reactivity (γ=0.002; p=0.024) and also greater cortisol and IL-6 habituation (cortisol: γ=0.00009; p=0.03, IL-6: γ=0.001; p=0.024). CONCLUSIONS Patterns of basal HPA axis activity that are related with healthier outcomes were found to be related with stronger initial cortisol and IL-6 reactivity and greater habituation. This is an important step in understanding the long-term health implications of acute stress responsiveness, and future studies should employ longitudinal designs to identify the direction of these relationships.
Collapse
Affiliation(s)
- Xuejie Chen
- Department of Psychology and Volen National Center for Complex Systems, Brandeis University, Waltham, MA, United States
| | - Danielle Gianferante
- Department of Psychology and Volen National Center for Complex Systems, Brandeis University, Waltham, MA, United States
| | - Luke Hanlin
- Department of Psychology and Volen National Center for Complex Systems, Brandeis University, Waltham, MA, United States
| | - Alexander Fiksdal
- Department of Psychology and Volen National Center for Complex Systems, Brandeis University, Waltham, MA, United States
| | - Juliana G. Breines
- Department of Psychology and Volen National Center for Complex Systems, Brandeis University, Waltham, MA, United States
| | - Myriam V. Thoma
- Department of Psychology and Volen National Center for Complex Systems, Brandeis University, Waltham, MA, United States,Division of Psychopathology and Clinical Intervention, University of Zurich, Zurich, Switzerland
| | - Nicolas Rohleder
- Department of Psychology and Volen National Center for Complex Systems, Brandeis University, Waltham, MA, United States; Chair of Health Psychology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
28
|
Bielefeld P, Schouten M, Lucassen PJ, Fitzsimons CP. Transcription factor oscillations in neural stem cells: Implications for accurate control of gene expression. NEUROGENESIS 2017; 4:e1262934. [PMID: 28321433 PMCID: PMC5345753 DOI: 10.1080/23262133.2016.1262934] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/14/2016] [Accepted: 11/15/2016] [Indexed: 01/14/2023]
Abstract
Naturally occurring oscillations in glucocorticoids induce a cyclic activation of the glucocorticoid receptor (GR), a well-characterized ligand-activated transcription factor. These cycles of GR activation/deactivation result in rapid GR exchange at genomic response elements and GR recycling through the chaperone machinery, ultimately generating pulses of GR-mediated transcriptional activity of target genes. In a recent article we have discussed the implications of circadian and high-frequency (ultradian) glucocorticoid oscillations for the dynamic control of gene expression in hippocampal neural stem/progenitor cells (NSPCs) (Fitzsimons et al., Front. Neuroendocrinol., 2016). Interestingly, this oscillatory transcriptional activity is common to other transcription factors, many of which regulate key biological functions in NSPCs, such as NF-kB, p53, Wnt and Notch. Here, we discuss the oscillatory behavior of these transcription factors, their role in a biologically accurate target regulation and the potential importance for a dynamic control of transcription activity and gene expression in NSPCs.
Collapse
Affiliation(s)
- Pascal Bielefeld
- Neuroscience Program, Swammerdam Institute for Life Sciences, Faculty of Sciences, University of Amsterdam
| | - Marijn Schouten
- Neuroscience Program, Swammerdam Institute for Life Sciences, Faculty of Sciences, University of Amsterdam
| | - Paul J Lucassen
- Neuroscience Program, Swammerdam Institute for Life Sciences, Faculty of Sciences, University of Amsterdam
| | - Carlos P Fitzsimons
- Neuroscience Program, Swammerdam Institute for Life Sciences, Faculty of Sciences, University of Amsterdam
| |
Collapse
|
29
|
George CL, Birnie MT, Flynn BP, Kershaw YM, Lightman SL, Conway-Campbell BL. Ultradian glucocorticoid exposure directs gene-dependent and tissue-specific mRNA expression patterns in vivo. Mol Cell Endocrinol 2017; 439:46-53. [PMID: 27769714 PMCID: PMC5131830 DOI: 10.1016/j.mce.2016.10.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 10/17/2016] [Accepted: 10/18/2016] [Indexed: 10/28/2022]
Abstract
In this paper we report differential decoding of the ultradian corticosterone signal by glucocorticoid target tissues. Pulsatile corticosterone replacement in adrenalectomised rats resulted in different dynamics of Sgk1 mRNA production, with a distinct pulsatile mRNA induction profile observed in the pituitary in contrast to a non-pulsatile induction in the prefrontal cortex (PFC). We further report the first evidence for pulsatile transcriptional repression of a glucocorticoid-target gene in vivo, with pulsatile regulation of Pomc transcription in pituitary. We have explored a potential mechanism for differences in the induction dynamics of the same transcript (Sgk1) between the PFC and pituitary. Glucocorticoid receptor (GR) activation profiles were strikingly different in pituitary and prefrontal cortex, with a significantly greater dynamic range and shorter duration of GR activity detected in the pituitary, consistent with the more pronounced gene pulsing effect observed. In the prefrontal cortex, expression of Gilz mRNA was also non-pulsatile and exhibited a significantly delayed timecourse of increase and decrease when compared to Sgk1, additionally highlighting gene-specific regulatory dynamics during ultradian glucocorticoid treatment.
Collapse
Affiliation(s)
- Charlotte L George
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK; CGAT, MRC Weatherall Institute of Molecular Medicine Centre for Computational Biology, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK.
| | - Matthew T Birnie
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK.
| | - Benjamin P Flynn
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK.
| | - Yvonne M Kershaw
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK.
| | - Stafford L Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK.
| | - Becky L Conway-Campbell
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK.
| |
Collapse
|
30
|
The activity of the glucocorticoid receptor is regulated by SUMO conjugation to FKBP51. Cell Death Differ 2016; 23:1579-91. [PMID: 27177020 DOI: 10.1038/cdd.2016.44] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/01/2016] [Accepted: 04/08/2016] [Indexed: 02/06/2023] Open
Abstract
FK506-binding protein 51 (FKBP51) regulates the activity of the glucocorticoid receptor (GR), and is therefore a key mediator of the biological actions of glucocorticoids. However, the understanding of the molecular mechanisms that govern its activity remains limited. Here, we uncover a novel regulatory switch for GR activity by the post-translational modification of FKBP51 with small ubiquitin-like modifier (SUMO). The major SUMO-attachment site, lysine 422, is required for FKBP51-mediated inhibition of GR activity in hippocampal neuronal cells. Importantly, impairment of SUMO conjugation to FKBP51 impacts on GR-dependent neuronal signaling and differentiation. We demonstrate that SUMO conjugation to FKBP51 is enhanced by the E3 ligase PIAS4 and by environmental stresses such as heat shock, which impact on GR-dependent transcription. SUMO conjugation to FKBP51 regulates GR hormone-binding affinity and nuclear translocation by promoting FKBP51 interaction within the GR complex. SUMOylation-deficient FKBP51 fails to interact with Hsp90 and GR thus facilitating the recruitment of the closely related protein, FKBP52, which enhances GR transcriptional activity. Moreover, we show that the modification of FKBP51 with SUMO modulates its binding to Hsp90. Our data establish SUMO conjugation as a novel regulatory mechanism in the Hsp90 cochaperone activity of FKBP51 with a functional impact on GR signaling in a neuronal context.
Collapse
|
31
|
Fitzsimons CP, Herbert J, Schouten M, Meijer OC, Lucassen PJ, Lightman S. Circadian and ultradian glucocorticoid rhythmicity: Implications for the effects of glucocorticoids on neural stem cells and adult hippocampal neurogenesis. Front Neuroendocrinol 2016; 41:44-58. [PMID: 27234350 DOI: 10.1016/j.yfrne.2016.05.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 05/01/2016] [Accepted: 05/03/2016] [Indexed: 01/01/2023]
Abstract
Psychosocial stress, and within the neuroendocrine reaction to stress specifically the glucocorticoid hormones, are well-characterized inhibitors of neural stem/progenitor cell proliferation in the adult hippocampus, resulting in a marked reduction in the production of new neurons in this brain area relevant for learning and memory. However, the mechanisms by which stress, and particularly glucocorticoids, inhibit neural stem/progenitor cell proliferation remain unclear and under debate. Here we review the literature on the topic and discuss the evidence for direct and indirect effects of glucocorticoids on neural stem/progenitor cell proliferation and adult neurogenesis. Further, we discuss the hypothesis that glucocorticoid rhythmicity and oscillations originating from the activity of the hypothalamus-pituitary-adrenal axis, may be crucial for the regulation of neural stem/progenitor cells in the hippocampus, as well as the implications of this hypothesis for pathophysiological conditions in which glucocorticoid oscillations are affected.
Collapse
Affiliation(s)
- Carlos P Fitzsimons
- Neuroscience Program, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098XH Amsterdam, The Netherlands.
| | - Joe Herbert
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Marijn Schouten
- Neuroscience Program, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098XH Amsterdam, The Netherlands
| | - Onno C Meijer
- Leiden University Medical Centre, Department of Endocrinology, Leiden, The Netherlands
| | - Paul J Lucassen
- Neuroscience Program, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098XH Amsterdam, The Netherlands.
| | - Stafford Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, United Kingdom
| |
Collapse
|
32
|
Gil-Lozano M, Wu WK, Martchenko A, Brubaker PL. High-Fat Diet and Palmitate Alter the Rhythmic Secretion of Glucagon-Like Peptide-1 by the Rodent L-cell. Endocrinology 2016; 157:586-99. [PMID: 26646204 DOI: 10.1210/en.2015-1732] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Secretion of the incretin hormone, glucagon-like peptide-1 (GLP-1), by the intestinal L-cell is rhythmically regulated by an independent molecular clock. However, the impact of factors known to affect the activity of similar cell-autonomous clocks, such as circulating glucocorticoids and high-fat feeding, on GLP-1 secretory patterns remains to be elucidated. Herein the role of the endogenous corticosterone rhythm on the pattern of GLP-1 and insulin nutrient-induced responses was examined in corticosterone pellet-implanted rats. Moreover, the impact of nutrient excess on the time-dependent secretion of both hormones was assessed in rats fed a high-fat, high-sucrose diet. Finally, the effects of the saturated fatty acid, palmitate, on the L-cell molecular clock and GLP-1 secretion were investigated in vitro using murine GLUTag L-cells. Diurnal variations in GLP-1 and insulin nutrient-induced responses were maintained in animals lacking an endogenous corticosterone rhythm, suggesting that glucocorticoids are not the predominant entrainment factor for L-cell rhythmic activity. In addition to hyperglycemia, hyperinsulinemia, insulin resistance, and disorganization of feeding behavior, high-fat high-sucrose-fed rats showed a total abrogation of the diurnal variation in GLP-1 and insulin nutrient-induced responses, with comparable levels of both hormones at the normal peak (5:00 pm) and trough (5:00 am) of their daily pattern. Finally, palmitate incubation induced profound derangements in the rhythmic expression of circadian oscillators in GLUTag L-cells and severely impaired the secretory activity of these cells. Collectively our findings demonstrate that obesogenic diets disrupt the rhythmic activity of the L-cell, partially through a direct effect of specific nutritional components.
Collapse
Affiliation(s)
- Manuel Gil-Lozano
- Departments of Physiology (M.G.-L., W.K.W., A.M., P.L.B.) and Medicine (P.L.B.), University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - W Kelly Wu
- Departments of Physiology (M.G.-L., W.K.W., A.M., P.L.B.) and Medicine (P.L.B.), University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Alexandre Martchenko
- Departments of Physiology (M.G.-L., W.K.W., A.M., P.L.B.) and Medicine (P.L.B.), University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Patricia L Brubaker
- Departments of Physiology (M.G.-L., W.K.W., A.M., P.L.B.) and Medicine (P.L.B.), University of Toronto, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
33
|
Lee MS, Kim YH, Park WS, Park OK, Kwon SH, Hong KS, Rhim H, Shim I, Morita K, Wong DL, Patel PD, Lyons DM, Schatzberg AF, Her S. Temporal variability of glucocorticoid receptor activity is functionally important for the therapeutic action of fluoxetine in the hippocampus. Mol Psychiatry 2016; 21:252-60. [PMID: 25330740 PMCID: PMC5189925 DOI: 10.1038/mp.2014.137] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/11/2014] [Accepted: 09/04/2014] [Indexed: 12/19/2022]
Abstract
Previous studies have shown inconsistent results regarding the actions of antidepressants on glucocorticoid receptor (GR) signalling. To resolve these inconsistencies, we used a lentiviral-based reporter system to directly monitor rat hippocampal GR activity during stress adaptation. Temporal GR activation was induced significantly by acute stress, as demonstrated by an increase in the intra-individual variability of the acute stress group compared with the variability of the non-stress group. However, the increased intra-individual variability was dampened by exposure to chronic stress, which was partly restored by fluoxetine treatment without affecting glucocorticoid secretion. Immobility in the forced-swim test was negatively correlated with the intra-individual variability, but was not correlated with the quantitative GR activity during fluoxetine therapy; this highlights the temporal variability in the neurobiological links between GR signalling and the therapeutic action of fluoxetine. Furthermore, we demonstrated sequential phosphorylation between GR (S224) and (S232) following fluoxetine treatment, showing a molecular basis for hormone-independent nuclear translocation and transcriptional enhancement. Collectively, these results suggest a neurobiological mechanism by which fluoxetine treatment confers resilience to the chronic stress-mediated attenuation of hypothalamic-pituitary-adrenal axis activity.
Collapse
Affiliation(s)
- M-S Lee
- Bio-Imaging Centre, Korea Basic Science Institute, Chuncheon, South Korea
| | - Y-H Kim
- Bio-Imaging Centre, Korea Basic Science Institute, Chuncheon, South Korea
| | - W-S Park
- Bio-Imaging Centre, Korea Basic Science Institute, Chuncheon, South Korea
| | - O-K Park
- Bio-Imaging Centre, Korea Basic Science Institute, Chuncheon, South Korea
| | - S-H Kwon
- Bio-Imaging Centre, Korea Basic Science Institute, Chuncheon, South Korea
| | - K S Hong
- Division of MR Research, Korea Basic Science Institute, Cheongwon, South Korea
| | - H Rhim
- Neuroscience Centre, Korea Institute of Science and Technology, Seoul, South Korea
| | - I Shim
- Acupuncture and Meridian Science Research Centre, Kyung Hee University, Seoul, South Korea
| | - K Morita
- Department of Nursing, Shikoku University, School of Health Sciences, Tokushima, Japan
| | - D L Wong
- Department of Psychiatry, Harvard Medical School and Laboratory of Molecular and Developmental Neurobiology, McLean Hospital, Belmont, MA, USA
| | - P D Patel
- Department of Psychiatry, Molecular and Behavioral Neuroscience Institute, University of Michigan Medical Centre, Ann Arbor, MI, USA
| | - D M Lyons
- Departments of Psychiatry, Stanford University Medical Centre, Stanford, CA, USA
| | - A F Schatzberg
- Departments of Psychiatry, Stanford University Medical Centre, Stanford, CA, USA
| | - S Her
- Bio-Imaging Centre, Korea Basic Science Institute, Chuncheon, South Korea,Bio-Imaging Centre, Korea Basic Science Institute, 192-1 Hyoja 2-Dong, Chuncheon, Gangwon-Do 200-701, South Korea. E-mail:
| |
Collapse
|
34
|
Sarabdjitsingh RA, Pasricha N, Smeets JAS, Kerkhofs A, Mikasova L, Karst H, Groc L, Joëls M. Hippocampal Fast Glutamatergic Transmission Is Transiently Regulated by Corticosterone Pulsatility. PLoS One 2016; 11:e0145858. [PMID: 26741493 PMCID: PMC4712151 DOI: 10.1371/journal.pone.0145858] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 12/09/2015] [Indexed: 12/22/2022] Open
Abstract
In recent years it has become clear that corticosteroid hormones (such as corticosterone) are released in ultradian pulses as a natural consequence of pituitary-adrenal interactions. All organs, including the brain, are thus exposed to pulsatile changes in corticosteroid hormone level, important to ensure full genomic responsiveness to stress-induced surges. However, corticosterone also changes neuronal excitability through rapid non-genomic pathways, particularly in the hippocampus. Potentially, background excitability of hippocampal neurons could thus be changed by pulsatile exposure to corticosteroids. It is currently unknown, though, how neuronal activity alters during a sequence of corticosterone pulses. To test this, hippocampal cells were exposed in vitro to four consecutive corticosterone pulses with a 60 min inter-pulse interval. During the pulses we examined four features of hippocampal signal transfer by the main excitatory transmitter glutamate—i.e., postsynaptic responses to spontaneous release of presynaptic vesicles, postsynaptic GluA2-AMPA receptor dynamics, basal (evoked) field responses, and synaptic plasticity, using a set of high resolution imaging and electrophysiological approaches. We show that the first pulse of corticosterone causes a transient increase in miniature EPSC frequency, AMPA receptor trafficking and synaptic plasticity, while basal evoked field responses are unaffected. This pattern is not maintained during subsequent applications: responses become more variable, attenuate or even reverse over time, albeit with different kinetics for the various experimental endpoints. This may indicate that the beneficial effect of ultradian pulses on transcriptional regulation in the hippocampus is not consistently accompanied by short-term perturbations in background excitability. In general, this could be interpreted as a means to keep hippocampal neurons responsive to incoming signals related to environmental challenges.
Collapse
Affiliation(s)
- R. Angela Sarabdjitsingh
- Dept. Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| | - Natasha Pasricha
- Dept. Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
- Universite de Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000 Bordeaux, France
- CNRS, IINS UMR 5297, Bordeaux, France
| | - Johanna A. S. Smeets
- Dept. Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Amber Kerkhofs
- Dept. Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
- Universite de Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000 Bordeaux, France
- CNRS, IINS UMR 5297, Bordeaux, France
| | - Lenka Mikasova
- Universite de Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000 Bordeaux, France
- CNRS, IINS UMR 5297, Bordeaux, France
| | - Henk Karst
- Dept. Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Laurent Groc
- Universite de Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000 Bordeaux, France
- CNRS, IINS UMR 5297, Bordeaux, France
| | - Marian Joëls
- Dept. Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
35
|
Indomethacin induced gene regulation in the rat hippocampus. Mol Brain 2015; 8:59. [PMID: 26438564 PMCID: PMC4595115 DOI: 10.1186/s13041-015-0150-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 09/30/2015] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Non-steroidal anti-inflammatory drugs such as indomethacin are widely used to treat inflammatory diseases and manage pain, fever and inflammation in several conditions, including neuropsychiatric disorders. Although they predominantly function by inhibiting cyclooxygenase (COX) activity, important COX-independent actions also occur. These actions could be responsible for the adverse side effects associated with chronic and/or high dose usage of this popular drug class. RESULTS We examined gene regulation in the hippocampus after peripheral administration of indomethacin by employing a microarray approach. Secondary confirmation and the brain expression pattern of regulated genes was examined by in situ hybridization and immunohistochemistry. Transglutaminase 2, serum glucocorticoid inducible kinase, Inhibitor of NF-kappa B and vascular endothelial growth factor were among genes that were prominently upregulated, while G-protein coupled receptor 56 and neuropeptide Y were among genes that were downregulated by indomethacin. Co-localization studies using blood vessel markers revealed that transglutaminase 2 was induced specifically in brain vasculature. CONCLUSIONS The data demonstrate that COX-inhibitors can differentially regulate gene transcription in multiple, functionally distinctly cell types in the brain. The results provide additional insight into the molecular actions of COX-inhibitors and indicate that their effects on vasculature could influence cerebral blood flow mechanisms.
Collapse
|
36
|
Ayyar VS, Almon RR, Jusko WJ, DuBois DC. Quantitative tissue-specific dynamics of in vivo GILZ mRNA expression and regulation by endogenous and exogenous glucocorticoids. Physiol Rep 2015; 3:3/6/e12382. [PMID: 26056061 PMCID: PMC4510616 DOI: 10.14814/phy2.12382] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Glucocorticoids (GC) are steroid hormones, which regulate metabolism and immune function. Synthetic GCs, or corticosteroids (CS), have appreciable clinical utility via their ability to suppress inflammation in immune-mediated diseases like asthma and rheumatoid arthritis. Recent work has provided insight to novel GC-induced genes that mediate their anti-inflammatory effects, including glucocorticoid-induced leucine zipper (GILZ). Since GILZ comprises an important part of GC action, its regulation by both drug and hormone will influence CS therapy. In addition, GILZ expression is often employed as a biomarker of GC action, which requires judicious selection of sampling time. Understanding the in vivo regulation of GILZ mRNA expression over time will provide insight into both the physiological regulation of GILZ by endogenous GC and the dynamics of its enhancement by CS. A highly quantitative qRT-PCR assay was developed for measuring GILZ mRNA expression in tissues obtained from normal and CS-treated rats. This assay was applied to measure GILZ mRNA expression in eight tissues; to determine its endogenous regulation over time; and to characterize its dynamics in adipose tissue, muscle, and liver following treatment with CS. We demonstrate that GILZ mRNA is expressed in several tissues. GILZ mRNA expression in adipose tissue displayed a robust circadian rhythm that was entrained with the circadian oscillation of endogenous corticosterone; and is strongly enhanced by acute and chronic dosing. Single dosing also enhanced GILZ mRNA in muscle and liver, but the dynamics varied. In conclusion, GILZ is widely expressed in the rat and highly regulated by endogenous and exogenous GCs.
Collapse
Affiliation(s)
- Vivaswath S Ayyar
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York
| | - Richard R Almon
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York Department of Biological Sciences, State University of New York at Buffalo, Buffalo, New York New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York
| | - William J Jusko
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York
| | - Debra C DuBois
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York Department of Biological Sciences, State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
37
|
Hall BS, Moda RN, Liston C. Glucocorticoid Mechanisms of Functional Connectivity Changes in Stress-Related Neuropsychiatric Disorders. Neurobiol Stress 2015; 1:174-183. [PMID: 25729760 PMCID: PMC4340078 DOI: 10.1016/j.ynstr.2014.10.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Stress—especially chronic, uncontrollable stress—is an important risk factor for many neuropsychiatric disorders. The underlying mechanisms are complex and multifactorial, but they involve correlated changes in structural and functional measures of neuronal connectivity within cortical microcircuits and across neuroanatomically distributed brain networks. Here, we review evidence from animal models and human neuroimaging studies implicating stress-associated changes in functional connectivity in the pathogenesis of PTSD, depression, and other neuropsychiatric conditions. Changes in fMRI measures of corticocortical connectivity across distributed networks may be caused by specific structural alterations that have been observed in the prefrontal cortex, hippocampus, and other vulnerable brain regions. These effects are mediated in part by glucocorticoids, which are released from the adrenal gland in response to a stressor and also oscillate in synchrony with diurnal rhythms. Recent work indicates that circadian glucocorticoid oscillations act to balance synapse formation and pruning after learning and during development, and chronic stress disrupts this balance. We conclude by considering how disrupted glucocorticoid oscillations may contribute to the pathophysiology of depression and PTSD in vulnerable individuals, and how circadian rhythm disturbances may affect non-psychiatric populations, including frequent travelers, shift workers, and patients undergoing treatment for autoimmune disorders.
Collapse
Affiliation(s)
- Baila S Hall
- Brain and Mind Research Institute and Program in Neuroscience, Weill Cornell Medical College, 413 East 69 Street, Box 240, New York, NY 10021
| | - Rachel N Moda
- Brain and Mind Research Institute and Program in Neuroscience, Weill Cornell Medical College, 413 East 69 Street, Box 240, New York, NY 10021
| | - Conor Liston
- Brain and Mind Research Institute and Program in Neuroscience, Weill Cornell Medical College, 413 East 69 Street, Box 240, New York, NY 10021 ; Sackler Institute for Developmental Psychobiology, Weill Cornell Medical College, 413 East 69 Street, Box 240, New York, NY 10021 ; Department of Psychiatry, Weill Cornell Medical College, 413 East 69 Street, Box 240, New York, NY 10021
| |
Collapse
|
38
|
Expression of glucocorticoid inducible genes is associated with reductions in cornu ammonis and dentate gyrus volumes in patients with major depressive disorder. Dev Psychopathol 2014; 26:1209-17. [DOI: 10.1017/s0954579414000972] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
AbstractAlterations of the glucocorticoid system and of hippocampal volumes have consistently been reported in patients with major depressive disorders (MDD). The aim of the present study was to investigate whether the messenger RNA (mRNA) expression of glucocorticoid inducible genes is associated with changes in the cornu ammonis (CA) and dentate gyrus subfields. Forty-three patients with MDD and 43 healthy controls were recruited and investigated with high resolution magnetic resonance imaging. Hippocampal subfields were measured using freesurfer. Measurement of whole blood mRNA expression of glucocorticoid inducible genes serum and glucocorticoid-regulated kinase 1 (SGK1), FK506 binding protein 5 (FKBP5), and glucocorticoid induced leucine zipper (GILZ) was performed. Patients with MDD had significantly smaller volumes of CA1, CA2/3, CA4/DG, and subiculum compared to healthy controls. In the regression analysis, the factor diagnosis had a significant moderating effect on the association of SGK1 and hippocampal volumes. Patients with low expression of SGK1 had significantly smaller CA2/3 and CA4/DG volumes compared to patients with high expression of SGK1 mRNA and to healthy controls with low/high expression of SGK1, respectively. Therefore, a lack of mRNA expression of glucocorticoid inducible genes in patients with MDD that seems to correspond to a blunted cortisol response is associated with smaller hippocampal CA and dentate gyrus volumes. SGK1 seems to be particularly relevant for stress-related mental disorders.
Collapse
|
39
|
Shyti R, Eikermann-Haerter K, van Heiningen SH, Meijer OC, Ayata C, Joëls M, Ferrari MD, van den Maagdenberg AMJM, Tolner EA. Stress hormone corticosterone enhances susceptibility to cortical spreading depression in familial hemiplegic migraine type 1 mutant mice. Exp Neurol 2014; 263:214-20. [PMID: 25447936 DOI: 10.1016/j.expneurol.2014.10.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 09/25/2014] [Accepted: 10/22/2014] [Indexed: 01/10/2023]
Abstract
Stress is a putative migraine trigger, but the pathogenic mechanisms involved are unknown. Stress and stress hormones increase neuronal excitability by enhancing glutamatergic neurotransmission, but inhibitory effects have also been reported. We hypothesise that an acute rise in stress hormones, such as corticosteroids which are released after stress, increase neuronal excitability and thereby may increase susceptibility to cortical spreading depression (CSD), the mechanism underlying the migraine aura. Here we investigated effects of acute restraint stress and of the stress hormone corticosterone on CSD susceptibility as surrogate migraine marker, in a transgenic mouse model of familial hemiplegic migraine type 1 (FHM1), which displays increased glutamatergic cortical neurotransmission and increased propensity for CSD. We found that 20-min and 3-h restraint stress did not influence CSD susceptibility in mutant or wild-type mice, despite elevated levels of plasma corticosterone. By contrast, subcutaneous administration of 20mg/kg corticosterone increased CSD frequency exclusively in mutant mice, while corticosterone plasma levels were similarly elevated in mutants and wild types. The effect of corticosterone on CSD frequency was normalised by pre-administration of the glucocorticoid receptor (GR) antagonist mifepristone. These findings suggest that corticosteroid-induced GR activation can enhance susceptibility to CSD in genetically susceptible individuals, and may predispose to attacks of migraine. Although corticosterone levels rise also during acute stress, the latter likely triggers a spatiotemporally more complex biological response with multiple positive and negative modulators which may not be adequately modeled by exogenous administration of corticosterone alone.
Collapse
Affiliation(s)
- Reinald Shyti
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Katharina Eikermann-Haerter
- Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, MA General Hospital, Harvard Medical School, Charlestown, USA
| | | | - Onno C Meijer
- Department of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Cenk Ayata
- Neurovascular Research Laboratory, Department of Radiology, MA General Hospital, Harvard Medical School, Charlestown, USA; Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, MA General Hospital, Harvard Medical School, Charlestown, USA
| | - Marian Joëls
- Department of Neuroscience and Pharmacology, University Medical Center Utrecht, Rudolf Magnus Institute of Neuroscience, Utrecht, The Netherlands
| | - Michel D Ferrari
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Arn M J M van den Maagdenberg
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands; Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Else A Tolner
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
40
|
Ultradian corticosterone pulses balance glutamatergic transmission and synaptic plasticity. Proc Natl Acad Sci U S A 2014; 111:14265-70. [PMID: 25225407 DOI: 10.1073/pnas.1411216111] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The rodent adrenal hormone corticosterone (CORT) reaches the brain in hourly ultradian pulses, with a steep rise in amplitude before awakening. The impact of a single CORT pulse on glutamatergic transmission is well documented, but it remains poorly understood how consecutive pulses impact on glutamate receptor trafficking and synaptic plasticity. By using high-resolution imaging and electrophysiological approaches, we report that a single pulse of CORT to hippocampal networks causes synaptic enrichment of glutamate receptors and increased responses to spontaneously released glutamatergic vesicles, collectively abrogating the ability to subsequently induce synaptic long-term potentiation. Strikingly, a second pulse of CORT one hour after the first--mimicking ultradian pulses--completely normalizes all aspects of glutamate transmission investigated, restoring the plastic range of the synapse. The effect of the second pulse is precisely timed and depends on a nongenomic glucocorticoid receptor-dependent pathway. This normalizing effect through a sequence of CORT pulses--as seen around awakening--may ensure that hippocampal glutamatergic synapses remain fully responsive and able to encode new stress-related information when daily activities start.
Collapse
|
41
|
Li ZY, Jiang YM, Liu YM, Guo Z, Shen SN, Liu XM, Pan RL. Saikosaponin D acts against corticosterone-induced apoptosis via regulation of mitochondrial GR translocation and a GR-dependent pathway. Prog Neuropsychopharmacol Biol Psychiatry 2014; 53:80-9. [PMID: 24636912 DOI: 10.1016/j.pnpbp.2014.02.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 02/13/2014] [Accepted: 02/25/2014] [Indexed: 11/17/2022]
Abstract
Saikosaponin D is an agonist of the glucocorticoid receptor (GR), and our preliminary study showed that it possesses neuroprotective effects in corticosterone-treated PC12 cells. However, further proof is required, and the molecular mechanisms of this neuroprotection remain unclear. This study sought to further examine the cytoprotective efficiency and potential mechanisms of action of Saikosaponin D in corticosterone-treated PC12 cells. The cells were treated with 250 μM corticosterone in the absence or presence of Saikosaponin D for 24 h; cell viability was then determined, and Hoechst 33342/propidium iodide (PI) and annexin/PI double staining, and TUNEL staining were performed. Next, mPTP, MMP, [Ca(2+)]i, translocation of the GR to the nucleus and Western blot analyses for caspase-3, caspase-9, cytochrome C, GR, GILZ, SGK-1, NF-Κb (P65), IκB-α, Bad, Akt, Hsp90 and HDAC-6 were investigated. The neuroprotective effects of Saikosaponin D were further confirmed by Hoechst 33342/PI, annexin/PI and TUNEL staining assays. These additional data suggested that Saikosaponin D partially reversed the physiological changes induced by corticosterone by inhibiting the translocation of the GR to the mitochondria, restoring mitochondrial function, down-regulating the expression of pro-apoptotic-related signalling events and up-regulating anti-apoptotic-related signalling events. These findings suggest that SSD exhibited its anti-apoptotic effects via differential regulation of mitochondrial and nuclear GR translocation, partial reversal of mitochondrial dysfunction, inhibition of the mitochondrial apoptotic pathway, and selective activation of the GR-dependent survival pathway.
Collapse
Affiliation(s)
- Zong-Yang Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100193, China
| | - Yu-Mao Jiang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100193, China
| | - Ya-Min Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100193, China
| | - Zhi Guo
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100193, China
| | - Sheng-Nan Shen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100193, China
| | - Xin-Min Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100193, China
| | - Rui-Le Pan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, Beijing 100193, China.
| |
Collapse
|
42
|
Abstract
Corticosteroids secreted as end product of the hypothalamic-pituitary-adrenal axis act like a double-edged sword in the brain. The hormones coordinate appraisal processes and decision making during the initial phase of a stressful experience and promote subsequently cognitive performance underlying the management of stress adaptation. This action exerted by the steroids on the initiation and termination of the stress response is mediated by 2 related receptor systems: mineralocorticoid receptors (MRs) and glucocorticoid receptors (GRs). The receptor types are unevenly distributed but colocalized in abundance in neurons of the limbic brain to enable these complementary hormone actions. This contribution starts from a historical perspective with the observation that phasic occupancy of GR during ultradian rhythmicity is needed to maintain responsiveness to corticosteroids. Then, during stress, initially MR activation enhances excitability of limbic networks that are engaged in appraisal and emotion regulation. Next, the rising hormone concentration occupies GR, resulting in reallocation of energy to limbic-cortical circuits with a role in behavioral adaptation and memory storage. Upon MR:GR imbalance, dysregulation of the hypothalamic-pituitary-adrenal axis occurs, which can enhance an individual's vulnerability. Imbalance is characteristic for chronic stress experience and depression but also occurs during exposure to synthetic glucocorticoids. Hence, glucocorticoid psychopathology may develop in susceptible individuals because of suppression of ultradian/circadian rhythmicity and depletion of endogenous corticosterone from brain MR. This knowledge generated from testing the balance hypothesis can be translated to a rational glucocorticoid therapy.
Collapse
Affiliation(s)
- E Ron de Kloet
- Department of Medical Pharmacology, Leiden Academic Centre for Drug Research, Leiden University and Department of Endocrinology and Metabolism, Leiden University Medical Center, 2300 RA Leiden, The Netherlands
| |
Collapse
|
43
|
|
44
|
Sarabdjitsingh RA, Zhou M, Yau JL, Webster SP, Walker BR, Seckl JR, Joëls M, Krugers HJ. Inhibiting 11β-hydroxysteroid dehydrogenase type 1 prevents stress effects on hippocampal synaptic plasticity and impairs contextual fear conditioning. Neuropharmacology 2014; 81:231-6. [DOI: 10.1016/j.neuropharm.2014.01.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 01/04/2014] [Accepted: 01/23/2014] [Indexed: 12/25/2022]
|
45
|
Henley DE, Lightman SL. Cardio-metabolic consequences of glucocorticoid replacement: relevance of ultradian signalling. Clin Endocrinol (Oxf) 2014; 80:621-8. [PMID: 24611992 DOI: 10.1111/cen.12422] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 12/31/2013] [Accepted: 01/30/2014] [Indexed: 01/01/2023]
Abstract
Chronic exposure to elevated glucocorticoid levels is associated with obesity, insulin resistance, impaired glucose tolerance, hypertension and dyslipidaemia, manifest classically in Cushing's syndrome and with high-dose glucocorticoid therapy. However, cardiovascular events are also reportedly higher in patients with primary and secondary hypoadrenalism receiving 'replacement' glucocorticoid doses. This has been attributed to an inability to mimic accurately the diurnal rhythm of cortisol with current oral replacement therapy and subsequent glucocorticoid excess. Although development of delayed release oral preparations has sought to overcome this problem, there has been little attention on the ultradian rhythm of glucocorticoids and its relevance for replacement therapy and associated cardio-metabolic comorbidity. Endogenous glucocorticoids are released in a pulsatile manner, and this ultradian rhythm is important in maintaining homeostatic control through glucocorticoid-receptor (GR)-dependent transcription regulation that rapidly responds to circulating hormone levels. Constant glucocorticoid exposure can result in continuous transcription, aberrant mRNA accumulation and abnormal protein levels. GR regulation of transcription programmes is highly cell and tissue specific, binding to distinct genomic loci in different cellular contexts. GR also interacts with a large cohort of DNA-binding factors with cell-specific interactions. The relevance of kinetic patterns of GR-dependent gene expression in vivo is not yet fully elucidated. However, given that GR gene variants are associated with cardiovascular disease, it is possible that ultradian delivery of glucocorticoid replacement may become important, at least in selected patients.
Collapse
Affiliation(s)
- David E Henley
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia; Faculty of Medicine, Dentistry and Health Sciences, School of Medicine and Pharmacology, University of Western Australia, Crawley, WA, Australia
| | | |
Collapse
|
46
|
Schumann G, Binder EB, Holte A, de Kloet ER, Oedegaard KJ, Robbins TW, Walker-Tilley TR, Bitter I, Brown VJ, Buitelaar J, Ciccocioppo R, Cools R, Escera C, Fleischhacker W, Flor H, Frith CD, Heinz A, Johnsen E, Kirschbaum C, Klingberg T, Lesch KP, Lewis S, Maier W, Mann K, Martinot JL, Meyer-Lindenberg A, Müller CP, Müller WE, Nutt DJ, Persico A, Perugi G, Pessiglione M, Preuss UW, Roiser JP, Rossini PM, Rybakowski JK, Sandi C, Stephan KE, Undurraga J, Vieta E, van der Wee N, Wykes T, Haro JM, Wittchen HU. Stratified medicine for mental disorders. Eur Neuropsychopharmacol 2014; 24:5-50. [PMID: 24176673 DOI: 10.1016/j.euroneuro.2013.09.010] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 09/09/2013] [Accepted: 09/26/2013] [Indexed: 12/13/2022]
Abstract
There is recognition that biomedical research into the causes of mental disorders and their treatment needs to adopt new approaches to research. Novel biomedical techniques have advanced our understanding of how the brain develops and is shaped by behaviour and environment. This has led to the advent of stratified medicine, which translates advances in basic research by targeting aetiological mechanisms underlying mental disorder. The resulting increase in diagnostic precision and targeted treatments may provide a window of opportunity to address the large public health burden, and individual suffering associated with mental disorders. While mental health and mental disorders have significant representation in the "health, demographic change and wellbeing" challenge identified in Horizon 2020, the framework programme for research and innovation of the European Commission (2014-2020), and in national funding agencies, clear advice on a potential strategy for mental health research investment is needed. The development of such a strategy is supported by the EC-funded "Roadmap for Mental Health Research" (ROAMER) which will provide recommendations for a European mental health research strategy integrating the areas of biomedicine, psychology, public health well being, research integration and structuring, and stakeholder participation. Leading experts on biomedical research on mental disorders have provided an assessment of the state of the art in core psychopathological domains, including arousal and stress regulation, affect, cognition social processes, comorbidity and pharmacotherapy. They have identified major advances and promising methods and pointed out gaps to be addressed in order to achieve the promise of a stratified medicine for mental disorders.
Collapse
Affiliation(s)
- Gunter Schumann
- MRC-Social Genetic Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, PO80, 16 De Crespigny Park, London SE5 8AF, UK.
| | | | - Arne Holte
- Norwegian Institute of Public Health, Oslo, Norway
| | - E Ronald de Kloet
- Department of Endocrinology and Metabolism, Leiden University Medical Centre and Medical Pharmacology, LACDR, Leiden University, The Netherlands
| | - Ketil J Oedegaard
- Department of Clinical Medicine, Section of Psychiatry, University of Bergen and Psychiatric division, Health Bergen, Norway
| | - Trevor W Robbins
- Behavioural and Clinical Neuroscience Institute and Department of Psychology, Cambridge University, Cambridge, UK
| | - Tom R Walker-Tilley
- MRC-Social Genetic Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, PO80, 16 De Crespigny Park, London SE5 8AF, UK
| | - Istvan Bitter
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| | - Verity J Brown
- Department of Psychology, University of St Andrews, St Andrews, UK
| | - Jan Buitelaar
- Department of Cognitive Neuroscience, University Medical Center, St Radboud and Karakter Child and Adolescent Psychiatry University Center, Nijmegen, The Netherlands
| | - Roberto Ciccocioppo
- Department of Experimental Medicine and Public Health, University of Camerino, Camerino, Macerata, Italy
| | | | - Carles Escera
- Department of Psychiatry and Clinical Psychobiology, University of Barcelona, Barcelona, Spain
| | - Wolfgang Fleischhacker
- Department of Psychiatry and Psychotherapy, Medical University Innsbruck, Innsbruck, Austria
| | - Herta Flor
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Chris D Frith
- Wellcome Trust Centre for Neuroimaging, University College London, London, UK
| | - Andreas Heinz
- Berlin School of Mind and Brain, Bernstein Center for Computational Neuroscience (BCCN), Clinic for Psychiatry and Psychotherapy, Charité - Universitätsmedizin, Berlin, Germany
| | - Erik Johnsen
- Department of Clinical Medicine, Section of Psychiatry, University of Bergen and Psychiatric division, Health Bergen, Norway
| | - Clemens Kirschbaum
- Technische Universität Dresden, Department of Psychology, Dresden, Germany
| | | | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Laboratory of Translational Neuroscience, University of Würzburg, Würzburg, Germany and Department of Neuroscience, School of Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, The Netherlands
| | - Shon Lewis
- University of Manchester, Manchester, UK
| | - Wolfgang Maier
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Karl Mann
- Department of Addictive Behaviour and Addiction Medicine, Central Institute of Mental Health, Mannheim, Germany
| | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM CEA Unit 1000 "Imaging & Psychiatry", University Paris Sud, Orsay; AP-HP Department of Adolescent Psychopathology and Medicine, Maison de Solenn, University Paris Descartes, Paris, France
| | - Andreas Meyer-Lindenberg
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christian P Müller
- Psychiatric University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Walter E Müller
- Department of Pharmacology, Biocenter Niederursel, University of Frankfurt, Frankfurt, Germany
| | - David J Nutt
- Neuropsychopharmacology Unit, Division of Brain Sciences, Imperial College, London, UK
| | - Antonio Persico
- Child and Adolescent Neuropsychiatry Unit & Laboratory of Molecular Psychiatry and Neurogenetics, University Campus Bio-Medico, Rome, Italy
| | - Giulio Perugi
- Department of Psychiatry, University of Pisa, Pisa, Italy
| | - Mathias Pessiglione
- Institut du Cerveau et de la Moelle épinière (ICM), Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Ulrich W Preuss
- Department of Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University of Halle-Wittenberg, Halle/Saale, Germany
| | - Jonathan P Roiser
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - Paolo M Rossini
- Department of Geriatrics, Neuroscience & Orthopaedics, Catholic University of Sacred Heart, Policlinico A. Gemelli, Rome, Italy
| | - Janusz K Rybakowski
- Department of Adult Psychiatry, Poznan University of Medical Sciences, Poznan, Poland
| | - Carmen Sandi
- Laboratory of Behavioural Genetics, Brain Mind Institute, EPFL, Lausanne, Switzerland
| | - Klaas E Stephan
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Juan Undurraga
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Eduard Vieta
- Bipolar Disorders Programme, Institute of Neuroscience, Hospital Clínic Barcelona, IDIBAPS, CIBERSAM, University of Barcelona, Barcelona, Catalonia, Spain
| | - Nic van der Wee
- Leiden Institute for Brain and Cogntion/Psychiatric Neuroimaging, Dept. of Psychiatry, Leiden University Medical Center, The Netherlands
| | - Til Wykes
- Department of Psychology, Institute of Psychiatry, King's College London, UK
| | - Josep Maria Haro
- Parc Sanitari Sant Joan de Déu, University of Barcelona, CIBERSAM, Barcelona, Spain
| | - Hans Ulrich Wittchen
- Institute of Clinical Psychology and Psychotherapy, TU Dresden, Dresden, Germany
| |
Collapse
|
47
|
Pandey GN, Rizavi HS, Ren X, Dwivedi Y, Palkovits M. Region-specific alterations in glucocorticoid receptor expression in the postmortem brain of teenage suicide victims. Psychoneuroendocrinology 2013; 38:2628-39. [PMID: 23845513 PMCID: PMC3812306 DOI: 10.1016/j.psyneuen.2013.06.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 06/05/2013] [Accepted: 06/17/2013] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Abnormal function of the hypothalamic-pituitary-adrenal (HPA) axis has been implicated in the pathophysiology of depression and suicide. The purpose of this study was to test the hypothesis that the reported dysregulation of the HPA axis in suicide may be related to a disturbed feedback inhibition caused by decreased corticoid receptors in the brain. We therefore determined the protein and gene expression of glucocorticoid (GR) and mineralocorticoid receptors (MR) in the postmortem brain of teenage suicide victims and matched normal controls. METHODS Protein and mRNA expression of GR (GR-α and GR-β) and MR and the mRNA expression of glucocorticoid-induced leucine zipper (GILZ), a target gene for GR were determined by immunolabeling using Western blot technique and the real-time RT-polymerase chain reaction (qPCR) technique in the prefrontal cortex (PFC), hippocampus, subiculum, and amygdala obtained from 24 teenage suicide victims and 24 teenage control subjects. RESULTS We observed that protein and gene expression of GR-α was significantly decreased in the PFC and amygdala, but not in the hippocampus or subiculum, of teenage suicide victims compared with normal control subjects. Also, the mRNA levels of GR inducible target gene GILZ was significantly decreased in PFC and amygdaloid nuclei but not in hippocampus compared with controls. In contrast, no significant differences were observed in protein or gene expression of MR in any of the areas studied between teenage suicide victims and normal control subjects. There was no difference in the expression of GR-β in the PFC between suicide victims and normal controls. CONCLUSIONS These results suggested that the observed dysregulation of the HPA axis in suicide may be related to a decreased expression of GR-α and GR inducible genes in the PFC and amygdala of teenage suicide victims. The reason why GR receptors are not dysregulated in the hippocampus or subiculum, presumably two sites of stress action, are not clear at this time.
Collapse
Affiliation(s)
- Ghanshyam N. Pandey
- University of Illinois at Chicago, Department of Psychiatry, Chicago, IL 60612, USA,Corresponding Author: Ghanshyam N. Pandey, Ph.D., University of Illinois at Chicago, 1601 West Taylor Street, Chicago, IL 60612, USA, Phone (312) 413-4540, Fax: (312) 413-4547,
| | - Hooriyah S. Rizavi
- University of Illinois at Chicago, Department of Psychiatry, Chicago, IL 60612, USA
| | - Xinguo Ren
- University of Illinois at Chicago, Department of Psychiatry, Chicago, IL 60612, USA
| | - Yogesh Dwivedi
- University of Illinois at Chicago, Department of Psychiatry, Chicago, IL 60612, USA
| | | |
Collapse
|
48
|
Carter BS, Hamilton DE, Thompson RC. Acute and chronic glucocorticoid treatments regulate astrocyte-enriched mRNAs in multiple brain regions in vivo. Front Neurosci 2013; 7:139. [PMID: 23966905 PMCID: PMC3736049 DOI: 10.3389/fnins.2013.00139] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/19/2013] [Indexed: 12/03/2022] Open
Abstract
Previous studies have primarily interpreted gene expression regulation by glucocorticoids in the brain in terms of impact on neurons; however, less is known about the corresponding impact of glucocorticoids on glia and specifically astrocytes in vivo. Recent microarray experiments have identified glucocorticoid-sensitive mRNAs in primary astrocyte cell culture, including a number of mRNAs that have reported astrocyte-enriched expression patterns relative to other brain cell types. Here, we have tested whether elevations of glucocorticoids regulate a subset of these mRNAs in vivo following acute and chronic corticosterone exposure in adult mice. Acute corticosterone exposure was achieved by a single injection of 10 mg/kg corticosterone, and tissue samples were harvested 2 h post-injection. Chronic corticosterone exposure was achieved by administering 10 mg/mL corticosterone via drinking water for 2 weeks. Gene expression was then assessed in two brain regions associated with glucocorticoid action (prefrontal cortex and hippocampus) by qPCR and by in situ hybridization. The majority of measured mRNAs regulated by glucocorticoids in astrocytes in vitro were similarly regulated by acute and/or chronic glucocorticoid exposure in vivo. In addition, the expression levels for mRNAs regulated in at least one corticosterone exposure condition (acute/chronic) demonstrated moderate positive correlation between the two conditions by brain region. In situ hybridization analyses suggest that select mRNAs are regulated by chronic corticosterone exposure specifically in astroctyes based on (1) similar general expression patterns between corticosterone-treated and vehicle-treated animals and (2) similar expression patterns to the pan-astrocyte marker Aldh1l1. Our findings demonstrate that glucocorticoids regulate astrocyte-enriched mRNAs in vivo and suggest that glucocorticoids regulate gene expression in the brain in a cell type-dependent fashion.
Collapse
Affiliation(s)
- Bradley S Carter
- Neuroscience Graduate Program, University of Michigan Ann Arbor, MI, USA ; Molecular and Behavioral Neuroscience Institute, University of Michigan Ann Arbor, MI, USA
| | | | | |
Collapse
|
49
|
Robertson S, Rohwer JM, Hapgood JP, Louw A. Impact of glucocorticoid receptor density on ligand-independent dimerization, cooperative ligand-binding and basal priming of transactivation: a cell culture model. PLoS One 2013; 8:e64831. [PMID: 23717665 PMCID: PMC3661511 DOI: 10.1371/journal.pone.0064831] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 04/18/2013] [Indexed: 11/26/2022] Open
Abstract
Glucocorticoid receptor (GR) levels vary between tissues and individuals and are altered by physiological and pharmacological effectors. However, the effects and implications of differences in GR concentration have not been fully elucidated. Using three statistically different GR concentrations in transiently transfected COS-1 cells, we demonstrate, using co-immunoprecipitation (CoIP) and fluorescent resonance energy transfer (FRET), that high levels of wild type GR (wtGR), but not of dimerization deficient GR (GRdim), display ligand-independent dimerization. Whole-cell saturation ligand-binding experiments furthermore establish that positive cooperative ligand-binding, with a concomitant increased ligand-binding affinity, is facilitated by ligand-independent dimerization at high concentrations of wtGR, but not GRdim. The down-stream consequences of ligand-independent dimerization at high concentrations of wtGR, but not GRdim, are shown to include basal priming of the system as witnessed by ligand-independent transactivation of both a GRE-containing promoter-reporter and the endogenous glucocorticoid (GC)-responsive gene, GILZ, as well as ligand-independent loading of GR onto the GILZ promoter. Pursuant to the basal priming of the system, addition of ligand results in a significantly greater modulation of transactivation potency than would be expected solely from the increase in ligand-binding affinity. Thus ligand-independent dimerization of the GR at high concentrations primes the system, through ligand-independent DNA loading and transactivation, which together with positive cooperative ligand-binding increases the potency of GR agonists and shifts the bio-character of partial GR agonists. Clearly GR-levels are a major factor in determining the sensitivity to GCs and a critical factor regulating transcriptional programs.
Collapse
Affiliation(s)
- Steven Robertson
- Department of Biochemistry, University of Stellenbosch, Matieland, Stellenbosch, Republic of South Africa
| | - Johann M. Rohwer
- Department of Biochemistry, University of Stellenbosch, Matieland, Stellenbosch, Republic of South Africa
| | - Janet P. Hapgood
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, Republic of South Africa
| | - Ann Louw
- Department of Biochemistry, University of Stellenbosch, Matieland, Stellenbosch, Republic of South Africa
| |
Collapse
|
50
|
Gil-Lozano M, Romaní-Pérez M, Outeiriño-Iglesias V, Vigo E, Brubaker PL, González-Matías LC, Mallo F. Effects of prolonged exendin-4 administration on hypothalamic-pituitary-adrenal axis activity and water balance. Am J Physiol Endocrinol Metab 2013; 304:E1105-17. [PMID: 23531615 DOI: 10.1152/ajpendo.00529.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Exendin-4 (Ex-4) is a natural agonist of the glucagon-like peptide-1 (GLP-1) receptor, currently being used as a treatment for type 2 diabetes mellitus due to its insulinotropic properties. Previous studies have revealed that acute administration of both GLP-1 and, in particular, Ex-4 potently stimulates hypothalamic-pituitary-adrenal (HPA) axis activity. In this work, the effects of prolonged Ex-4 exposure on HPA function were explored. To this end, Sprague-Dawley rats were subjected to a daily regimen of two Ex-4 injections (5 μg/kg sc) for a minimum of 7 days. We found that subchronic Ex-4 administration produced a number of effects that resemble chronic stress situations, including hyperactivation of the HPA axis during the trough hours, disruption of glucocorticoid circadian secretion, hypertrophy of the adrenal gland, decreased adrenal gland sensitivity, impaired pituitary-adrenal stress responses, and reductions in both food intake and body weight. In addition, a threefold increase in diuresis was observed followed by a 1.5-fold increase in water intake; these latter effects were abolished by adrenalectomy. Together, these findings indicate that Ex-4 induces a profound dysregulation of HPA axis activity that may also affect renal function.
Collapse
Affiliation(s)
- Manuel Gil-Lozano
- Laboratory of Endocrinology, Center for Biomedical Research, Campus As Lagoas-Marcosende, University of Vigo, Vigo, Spain
| | | | | | | | | | | | | |
Collapse
|