51
|
Cognitive impairment induced by permanent bilateral common carotid occlusion exacerbates depression-related behavioral, biochemical, immunological and neuronal markers. Brain Res 2015; 1596:58-68. [DOI: 10.1016/j.brainres.2014.09.056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 09/01/2014] [Accepted: 09/23/2014] [Indexed: 01/08/2023]
|
52
|
Differential effects of acute cortisol administration on deep and shallow episodic memory traces: A study on healthy males. Neurobiol Learn Mem 2014; 114:186-92. [DOI: 10.1016/j.nlm.2014.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 05/30/2014] [Accepted: 06/11/2014] [Indexed: 11/23/2022]
|
53
|
Li H, Scholl JL, Tu W, Hassell JE, Watt MJ, Forster GL, Renner KJ. Serotonergic responses to stress are enhanced in the central amygdala and inhibited in the ventral hippocampus during amphetamine withdrawal. Eur J Neurosci 2014; 40:3684-92. [PMID: 25234335 DOI: 10.1111/ejn.12735] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 08/05/2014] [Accepted: 08/20/2014] [Indexed: 12/26/2022]
Abstract
Withdrawal from amphetamine increases anxiety and reduces the ability to cope with stress, which are factors that are believed to contribute to drug relapse. Stress-induced serotonergic transmission in the central nucleus of the amygdala is associated with anxiety states and fear. Conversely, stress-induced increases in ventral hippocampal serotonin (5-HT) levels have been linked to coping mechanisms. The goal of this study was to investigate the neurobiological changes induced by amphetamine that contribute to stress sensitivity during withdrawal. We tested the hypothesis that limbic serotonergic responses to restraint stress would be altered in male Sprague-Dawley rats chronically pretreated with amphetamine (2.5 mg/kg, intraperitoneal) and then subjected to 2 weeks of withdrawal. Amphetamine withdrawal resulted in increased stress-induced behavioral arousal relative to control treatment, suggesting that drug withdrawal induced greater sensitivity to the stressor. When microdialysis was used to determine the effects of restraint on extracellular 5-HT, stress-induced increases in 5-HT levels were abolished in the ventral hippocampus and augmented in the central amygdala during amphetamine withdrawal. Reverse dialysis of the glucocorticoid receptor antagonist mifepristone into the ventral hippocampus blocked the stress-induced increase in 5-HT levels in saline-pretreated rats, suggesting that glucocorticoid receptors mediate stress-induced increases in 5-HT levels in the ventral hippocampus. However, mifepristone had no effect on stress-induced increases in 5-HT levels in the central amygdala, indicating that stress increases 5-HT levels in this region independently of glucocorticoid receptors. During amphetamine withdrawal, the absence of stress-induced increases in ventral hippocampal 5-HT levels combined with enhanced stress-induced serotonergic responses in the central amygdala may contribute to drug relapse by decreasing stress-coping ability and heightening stress responsiveness.
Collapse
Affiliation(s)
- Hao Li
- Department of Biology & Center for Brain and Behavior Research, University of South Dakota, 414 E. Clark St, Vermillion, SD, 57069, USA
| | | | | | | | | | | | | |
Collapse
|
54
|
Uchoa ET, Aguilera G, Herman JP, Fiedler JL, Deak T, Cordeiro de Sousa MB. Novel aspects of glucocorticoid actions. J Neuroendocrinol 2014; 26:557-72. [PMID: 24724595 PMCID: PMC4161987 DOI: 10.1111/jne.12157] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 04/06/2014] [Accepted: 04/08/2014] [Indexed: 12/20/2022]
Abstract
Normal hypothalamic-pituitary-adrenal (HPA) axis activity leading to the rhythmic and episodic release of adrenal glucocorticoids (GCs) is essential for body homeostasis and survival during stress. Acting through specific intracellular receptors in the brain and periphery, GCs regulate behaviour, as well as metabolic, cardiovascular, immune and neuroendocrine activities. By contrast to chronic elevated levels, circadian and acute stress-induced increases in GCs are necessary for hippocampal neuronal survival and memory acquisition and consolidation, as a result of the inhibition of apoptosis, the facilitation of glutamatergic neurotransmission and the formation of excitatory synapses, and the induction of immediate early genes and dendritic spine formation. In addition to metabolic actions leading to increased energy availability, GCs have profound effects on feeding behaviour, mainly via the modulation of orexigenic and anorixegenic neuropeptides. Evidence is also emerging that, in addition to the recognised immune suppressive actions of GCs by counteracting adrenergic pro-inflammatory actions, circadian elevations have priming effects in the immune system, potentiating acute defensive responses. In addition, negative-feedback by GCs involves multiple mechanisms leading to limited HPA axis activation and prevention of the deleterious effects of excessive GC production. Adequate GC secretion to meet body demands is tightly regulated by a complex neural circuitry controlling hypothalamic corticotrophin-releasing hormone (CRH) and vasopressin secretion, which are the main regulators of pituitary adrenocorticotrophic hormone (ACTH). Rapid feedback mechanisms, likely involving nongenomic actions of GCs, mediate the immediate inhibition of hypothalamic CRH and ACTH secretion, whereas intermediate and delayed mechanisms mediated by genomic actions involve the modulation of limbic circuitry and peripheral metabolic messengers. Consistent with their key adaptive roles, HPA axis components are evolutionarily conserved, being present in the earliest vertebrates. An understanding of these basic mechanisms may lead to novel approaches for the development of diagnostic and therapeutic tools for disorders related to stress and alterations of GC secretion.
Collapse
Affiliation(s)
- Ernane Torres Uchoa
- Department of Physiology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Greti Aguilera
- Section on Endocrine Physiology, National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - James P. Herman
- Department of Psychiatry and Behavioural Neuroscience, University of Cincinnati, Metabolic Diseases Institute, Cincinnati, OH, USA
| | - Jenny L. Fiedler
- Department of Biochemistry and Molecular Biology, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
| | - Terrence Deak
- Department of Psychology, Binghamton University, Binghamton, NY, USA
| | | |
Collapse
|
55
|
Revest JM, Le Roux A, Roullot-Lacarrière V, Kaouane N, Vallée M, Kasanetz F, Rougé-Pont F, Tronche F, Desmedt A, Piazza PV. BDNF-TrkB signaling through Erk1/2 MAPK phosphorylation mediates the enhancement of fear memory induced by glucocorticoids. Mol Psychiatry 2014; 19:1001-9. [PMID: 24126929 PMCID: PMC4195976 DOI: 10.1038/mp.2013.134] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 08/27/2013] [Accepted: 08/28/2013] [Indexed: 12/25/2022]
Abstract
Activation of glucocorticoid receptors (GR) by glucocorticoid hormones (GC) enhances contextual fear memories through the activation of the Erk1/2(MAPK) signaling pathway. However, the molecular mechanism mediating this effect of GC remains unknown. Here we used complementary molecular and behavioral approaches in mice and rats and in genetically modified mice in which the GR was conditionally deleted (GR(NesCre)). We identified the tPA-BDNF-TrkB signaling pathway as the upstream molecular effectors of GR-mediated phosphorylation of Erk1/2(MAPK) responsible for the enhancement of contextual fear memory. These findings complete our knowledge of the molecular cascade through which GC enhance contextual fear memory and highlight the role of tPA-BDNF-TrkB-Erk1/2(MAPK) signaling pathways as one of the core effectors of stress-related effects of GC.
Collapse
Affiliation(s)
- J-M Revest
- INSERM U862, Neurocentre Magendie, Pathophysiology of Addiction, Bordeaux, France,Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France,Pathophysiology of Addiction, Neurocentre Magendie, INSERM-U862, 146 rue Léo Saignat, Bordeaux F-33077, France. E-mail:
| | - A Le Roux
- INSERM U862, Neurocentre Magendie, Pathophysiology of Addiction, Bordeaux, France,Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France
| | - V Roullot-Lacarrière
- INSERM U862, Neurocentre Magendie, Pathophysiology of Addiction, Bordeaux, France,Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France
| | - N Kaouane
- Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France,INSERM U862, Neurocentre Magendie, Pathophysiology of Declarative Memory, Bordeaux, France
| | - M Vallée
- INSERM U862, Neurocentre Magendie, Pathophysiology of Addiction, Bordeaux, France,Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France
| | - F Kasanetz
- INSERM U862, Neurocentre Magendie, Pathophysiology of Addiction, Bordeaux, France,Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France
| | - F Rougé-Pont
- INSERM U862, Neurocentre Magendie, Pathophysiology of Addiction, Bordeaux, France,Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France
| | - F Tronche
- CNRS UMR7224, UPMC Université Pierre et Marie Curie, Molecular Genetics, Neurophysiology and Behavior, Paris, France
| | - A Desmedt
- Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France,INSERM U862, Neurocentre Magendie, Pathophysiology of Declarative Memory, Bordeaux, France
| | - P V Piazza
- INSERM U862, Neurocentre Magendie, Pathophysiology of Addiction, Bordeaux, France,Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France
| |
Collapse
|
56
|
Stress hormone exposure reduces mGluR5 expression in the nucleus accumbens: functional implications for interoceptive sensitivity to alcohol. Neuropsychopharmacology 2014; 39:2376-86. [PMID: 24713611 PMCID: PMC4138747 DOI: 10.1038/npp.2014.85] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 03/11/2014] [Accepted: 04/02/2014] [Indexed: 12/19/2022]
Abstract
Escalations in alcohol drinking associated with experiencing stressful life events and chronic life stressors may be related to altered sensitivity to the interoceptive/subjective effects of alcohol. Indeed, through the use of drug discrimination methods, rats show decreased sensitivity to the discriminative stimulus (interoceptive) effects of alcohol following exposure to the stress hormone corticosterone (CORT). This exposure produces heightened elevations in plasma CORT levels (eg, as may be experienced by an individual during stressful episodes). We hypothesized that decreased sensitivity to alcohol may be related, in part, to changes in metabotropic glutamate receptors-subtype 5 (mGluR5) in the nucleus accumbens, as these receptors in this brain region are known to regulate the discriminative stimulus effects of alcohol. In the accumbens, we found reduced mGluR5 expression (immunohistochemistry and Western blot) and decreased neural activation (as measured by c-Fos immunohistochemistry) in response to a moderate alcohol dose (1 g/kg) following CORT exposure (7 days). The functional role of these CORT-induced adaptations in relation to the discriminative stimulus effects of alcohol was confirmed, as both the systemic administration of 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB) an mGluR5 positive allosteric modulator and the intra-accumbens administration of (R,S)-2-Amino-2-(2-chloro-5-hydroxyphenyl)acetic acid sodium salt (CHPG) an mGluR5 agonist restored sensitivity to alcohol in discrimination-trained rats. These results suggest that activation of mGluR5 may alleviate the functional impact of the CORT-induced downregulation of mGluR5 in relation to sensitivity to alcohol. Understanding the contribution of such neuroadaptations to the interoceptive effects of alcohol may enrich our understanding of potential changes in subjective sensitivity to alcohol during stressful episodes.
Collapse
|
57
|
Vermetten E, Zhohar J, Krugers HJ. Pharmacotherapy in the aftermath of trauma; opportunities in the 'golden hours'. Curr Psychiatry Rep 2014; 16:455. [PMID: 24890991 DOI: 10.1007/s11920-014-0455-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Several lines of research have demonstrated that memories for fearful events become transiently labile upon re-exposure. Activation of molecular mechanisms is required in order to maintain retrieved information. This process is called reconsolidation. Targeting reconsolidation - as in exposure-based psychotherapy - offers therefore a potentially interesting tool to manipulate fear memories, and subsequently to treat disorders such as post-traumatic stress disorder (PTSD). In this paper we discuss the evidence for reconsolidation in rodents and humans and highlight recent studies in which clinical research on normal and abnormal fear extinction reduction of the expression of fear was obtained by targeting the process of reconsolidation. We conclude that reconsolidation presents an interesting opportunity to modify or alter fear and fear-related memories. More clinical research on normal and abnormal fear extinction is required.
Collapse
Affiliation(s)
- Eric Vermetten
- Department Psychiatry, Leiden University Medical Center Utrecht, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands,
| | | | | |
Collapse
|
58
|
Koe AS, Salzberg MR, Morris MJ, O'Brien TJ, Jones NC. Early life maternal separation stress augmentation of limbic epileptogenesis: the role of corticosterone and HPA axis programming. Psychoneuroendocrinology 2014; 42:124-33. [PMID: 24636509 DOI: 10.1016/j.psyneuen.2014.01.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 01/12/2014] [Accepted: 01/13/2014] [Indexed: 10/25/2022]
Abstract
Early life stress causes long-lasting effects on the limbic system that may be relevant to the development of mesial temporal lobe epilepsy (MTLE) and its associated psychopathology. Recent studies in rats suggest that maternal separation (MS), a model of early life stress, confers enduring vulnerability to amygdala kindling limbic epileptogenesis. However, the mechanisms underlying this remain unknown. Here, we tested whether hypothalamic-pituitary-adrenal (HPA) axis hyper-reactivity induced by MS - specifically the excessive secretion of corticosterone following a seizure - was involved in this vulnerability. In adult female rats subjected to MS from postnatal days 2-14, seizure-induced corticosterone responses were significantly augmented and prolonged for at least two hours post-seizure, compared to control early-handled (EH) rats. This was accompanied by reduced seizure threshold (p<0.05) and increased vulnerability to the kindling-induced progression of seizure duration (p<0.05) in MS rats. Pre-seizure treatment with the corticosterone synthesis inhibitor, metyrapone (MET) (50mg/kgsc) effectively blocked seizure-induced corticosterone responses. When delivered throughout kindling, MET treatment also reversed the MS-induced reduction in seizure threshold and the lengthened seizure duration back to levels of EH rats. These observations suggest that adverse early life environments induce a vulnerability to kindling epileptogenesis mediated by HPA axis hyper-reactivity, which could have relevance for the pathogenesis of MTLE.
Collapse
Affiliation(s)
- Amelia S Koe
- Department of Medicine, Royal Melbourne Hospital, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| | - Michael R Salzberg
- St Vincent's Mental Health Service, St Vincent's Hospital, Fitzroy, VIC, Australia; Department of Psychiatry, St Vincent's Hospital, University of Melbourne, Fitzroy, VIC, Australia
| | - Margaret J Morris
- Department of Pharmacology, University of New South Wales, Sydney, NSW, Australia
| | - Terence J O'Brien
- Department of Medicine, Royal Melbourne Hospital, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia; Department of Neurology, University of Melbourne, Parkville, VIC, Australia
| | - Nigel C Jones
- Department of Medicine, Royal Melbourne Hospital, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
59
|
Treccani G, Musazzi L, Perego C, Milanese M, Nava N, Bonifacino T, Lamanna J, Malgaroli A, Drago F, Racagni G, Nyengaard JR, Wegener G, Bonanno G, Popoli M. Stress and corticosterone increase the readily releasable pool of glutamate vesicles in synaptic terminals of prefrontal and frontal cortex. Mol Psychiatry 2014; 19:433-43. [PMID: 24535456 DOI: 10.1038/mp.2014.5] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 12/18/2013] [Accepted: 01/06/2014] [Indexed: 02/06/2023]
Abstract
Stress and glucocorticoids alter glutamatergic transmission, and the outcome of stress may range from plasticity enhancing effects to noxious, maladaptive changes. We have previously demonstrated that acute stress rapidly increases glutamate release in prefrontal and frontal cortex via glucocorticoid receptor and accumulation of presynaptic SNARE complex. Here we compared the ex vivo effects of acute stress on glutamate release with those of in vitro application of corticosterone, to analyze whether acute effect of stress on glutamatergic transmission is mediated by local synaptic action of corticosterone. We found that acute stress increases both the readily releasable pool (RRP) of vesicles and depolarization-evoked glutamate release, while application in vitro of corticosterone rapidly increases the RRP, an effect dependent on synaptic receptors for the hormone, but does not induce glutamate release for up to 20 min. These findings indicate that corticosterone mediates the enhancement of glutamate release induced by acute stress, and the rapid non-genomic action of the hormone is necessary but not sufficient for this effect.
Collapse
Affiliation(s)
- G Treccani
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics-Dipartimento di Scienze Farmacologiche e Biomolecolari and CEND, Università di Milano, Milano, Italy
| | - L Musazzi
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics-Dipartimento di Scienze Farmacologiche e Biomolecolari and CEND, Università di Milano, Milano, Italy
| | - C Perego
- Laboratory of Cell Physiology-Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Milano, Italy
| | - M Milanese
- Department of Pharmacy-Unit of Pharmacology and Toxicology, Center of Excellence for Biomedical Research, Università di Genova, Genova, Italy
| | - N Nava
- 1] Stereology and Electron Microscopy Laboratory, Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University Hospital, Aarhus, Denmark [2] Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - T Bonifacino
- Department of Pharmacy-Unit of Pharmacology and Toxicology, Center of Excellence for Biomedical Research, Università di Genova, Genova, Italy
| | - J Lamanna
- Neurobiology of Learning Unit, Scientific Institute San Raffaele and Università Vita e Salute San Raffaele, Milano, Italy
| | - A Malgaroli
- Neurobiology of Learning Unit, Scientific Institute San Raffaele and Università Vita e Salute San Raffaele, Milano, Italy
| | - F Drago
- Department of Clinical and Molecular Biomedicine, Section of Pharmacology and Biochemistry, Università di Catania, Catania, Italy
| | - G Racagni
- 1] Laboratory of Neuropsychopharmacology and Functional Neurogenomics-Dipartimento di Scienze Farmacologiche e Biomolecolari and CEND, Università di Milano, Milano, Italy [2] IRCCS San Giovanni di Dio-Fatebenefratelli, Brescia, Italy
| | - J R Nyengaard
- Stereology and Electron Microscopy Laboratory, Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University Hospital, Aarhus, Denmark
| | - G Wegener
- 1] Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark [2] Centre of Excellence for Pharmaceutical Sciences, North West University, Potchefstroom, South Africa
| | - G Bonanno
- Department of Pharmacy-Unit of Pharmacology and Toxicology, Center of Excellence for Biomedical Research, Università di Genova, Genova, Italy
| | - M Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics-Dipartimento di Scienze Farmacologiche e Biomolecolari and CEND, Università di Milano, Milano, Italy
| |
Collapse
|
60
|
Takeda A, Tamano H. Cognitive decline due to excess synaptic Zn(2+) signaling in the hippocampus. Front Aging Neurosci 2014; 6:26. [PMID: 24578691 PMCID: PMC3936311 DOI: 10.3389/fnagi.2014.00026] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 02/13/2014] [Indexed: 12/02/2022] Open
Abstract
Zinc is an essential component of physiological brain function. Vesicular zinc is released from glutamatergic (zincergic) neuron terminals and serves as a signal factor (Zn2+ signal) in both the intracellular (cytosol) compartment and the extracellular compartment. Synaptic Zn2+ signaling is dynamically linked to neurotransmission and is involved in processes of synaptic plasticity such as long-term potentiation and cognitive activity. On the other hand, the activity of the hypothalamic–pituitary–adrenal (HPA) axis, i.e., glucocorticoid secretion, which can potentiate glutamatergic neuron activity, is linked to cognitive function. HPA axis activity modifies synaptic Zn2+ dynamics at zincergic synapses. An increase in HPA axis activity, which occurs after exposure to stress, may induce excess intracellular Zn2+ signaling in the hippocampus, followed by hippocampus-dependent memory deficit. Excessive excitation of zincergic neurons in the hippocampus can contribute to cognitive decline under stressful and/or pathological conditions. This paper provides an overview of the ``Hypothesis and Theory'' of Zn2+-mediated modification of cognitive activity.
Collapse
Affiliation(s)
- Atsushi Takeda
- Department of Bioorganic Chemistry, School of Pharmaceutical Sciences, University of Shizuoka Shizuoka, Japan
| | - Haruna Tamano
- Department of Bioorganic Chemistry, School of Pharmaceutical Sciences, University of Shizuoka Shizuoka, Japan
| |
Collapse
|
61
|
Yılmaz T, Akça M, Turan Y, Ocak H, Kamaşak K, Yildirim M. Efficacy of dexamethasone on penicillin-induced epileptiform activity in rats: an electrophysiological study. Brain Res 2014; 1554:67-72. [PMID: 24495842 DOI: 10.1016/j.brainres.2014.01.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 01/05/2014] [Accepted: 01/24/2014] [Indexed: 11/18/2022]
Abstract
Corticosteroids are extensively used in treatment of many diseases. In neurosurgery practice, dexamethasone (DEX) is commonly used particularly in cerebral edema secondary to brain tumors, head trauma, and central nervous system infections. There are some uncertainties surrounding the secure use of DEX in patients with epilepsy or seizures induced by diseases of the central nervous system such as head trauma and brain tumors. Despite its extensive use, the effect of DEX on epileptiform activity is unclear. In this study the effect of DEX on epileptiform activity was investigated in rats. The effects of 1, 3, and 10mg/kg DEX on epileptiform activity was compared with effects of antiepileptic drugs commonly employed in treatment of epilepsy, namely phenytoin (PHT) 50mg/kg and levetiracetam (LEV) 50mg/kg that were administered intraperitoneally for 1 week. All groups were administered intracortical penicillin (500IU) to induce epileptiform activity. DEX at the doses of 3mg/kg and 10mg/kg significantly reduced spike frequencies compared to the initial values. In conclusion, we think that DEX can effectively decrease the epileptiform activity.
Collapse
Affiliation(s)
- Tevfik Yılmaz
- Department of Neurosurgery, Faculty of Medicine, Dicle University, Yenişehir 21280, Diyarbakir, Turkey.
| | - Metehan Akça
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Yahya Turan
- Department of Neurosurgery, Faculty of Medicine, Dicle University, Yenişehir 21280, Diyarbakir, Turkey
| | - Hakan Ocak
- Department of Anatomy, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Kağan Kamaşak
- Department of Neurosurgery, Faculty of Medicine, Dicle University, Yenişehir 21280, Diyarbakir, Turkey
| | - Mehmet Yildirim
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
62
|
Daulatzai MA. Role of stress, depression, and aging in cognitive decline and Alzheimer's disease. Curr Top Behav Neurosci 2014; 18:265-96. [PMID: 25167923 DOI: 10.1007/7854_2014_350] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Late-onset Alzheimer's disease (AD) is a chronic neurodegenerative disorder and the most common cause of progressive cognitive dysfunction and dementia. Despite considerable progress in elucidating the molecular pathology of this disease, we are not yet close to unraveling its etiopathogenesis. A battery of neurotoxic modifiers may underpin neurocognitive pathology via deleterious heterogeneous pathologic impact in brain regions, including the hippocampus. Three important neurotoxic factors being addressed here include aging, stress, and depression. Unraveling "upstream pathologies" due to these disparate neurotoxic entities, vis-à-vis cognitive impairment involving hippocampal dysfunction, is of paramount importance. Persistent systemic inflammation triggers and sustains neuroinflammation. The latter targets several brain regions including the hippocampus causing upregulation of amyloid beta and neurofibrillary tangles, synaptic and neuronal degeneration, gray matter volume atrophy, and progressive cognitive decline. However, what is the fundamental source of this peripheral inflammation in aging, stress, and depression? This chapter highlights and delineates the inflammatory involvement-i.e., from its inception from gut to systemic inflammation to neuroinflammation. It highlights an upregulated cascade in which gut-microbiota-related dysbiosis generates lipopolysaccharides (LPS), which enhances inflammation and gut's leakiness, and through a Web of interactions, it induces stress and depression. This may increase neuronal dysfunction and apoptosis, promote learning and memory impairment, and enhance vulnerability to cognitive decline.
Collapse
Affiliation(s)
- Mak Adam Daulatzai
- Sleep Disorders Group, EEE Department, Melbourne School of Engineering, The University of Melbourne, Building 193, 3rd Floor, Room no. 3/344, Parkville, VIC, 3010, Australia,
| |
Collapse
|
63
|
Whitehead G, Jo J, Hogg EL, Piers T, Kim DH, Seaton G, Seok H, Bru-Mercier G, Son GH, Regan P, Hildebrandt L, Waite E, Kim BC, Kerrigan TL, Kim K, Whitcomb DJ, Collingridge GL, Lightman SL, Cho K. Acute stress causes rapid synaptic insertion of Ca2+ -permeable AMPA receptors to facilitate long-term potentiation in the hippocampus. ACTA ACUST UNITED AC 2013; 136:3753-65. [PMID: 24271563 PMCID: PMC3859225 DOI: 10.1093/brain/awt293] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The neuroendocrine response to episodes of acute stress is crucial for survival whereas the prolonged response to chronic stress can be detrimental. Learning and memory are particularly susceptible to stress with cognitive deficits being well characterized consequences of chronic stress. Although there is good evidence that acute stress can enhance cognitive performance, the mechanism(s) for this are unclear. We find that hippocampal slices, either prepared from rats following 30 min restraint stress or directly exposed to glucocorticoids, exhibit an N-methyl-d-aspartic acid receptor-independent form of long-term potentiation. We demonstrate that the mechanism involves an NMDA receptor and PKA-dependent insertion of Ca2+-permeable AMPA receptors into synapses. These then trigger the additional NMDA receptor-independent form of LTP during high frequency stimulation.
Collapse
Affiliation(s)
- Garry Whitehead
- 1 Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, Whitson Street, Bristol BS1 3NY, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Finsterwald C, Alberini CM. Stress and glucocorticoid receptor-dependent mechanisms in long-term memory: from adaptive responses to psychopathologies. Neurobiol Learn Mem 2013; 112:17-29. [PMID: 24113652 DOI: 10.1016/j.nlm.2013.09.017] [Citation(s) in RCA: 222] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 09/20/2013] [Accepted: 09/25/2013] [Indexed: 12/17/2022]
Abstract
A proper response against stressors is critical for survival. In mammals, the stress response is primarily mediated by secretion of glucocorticoids via the hypothalamic-pituitary-adrenocortical (HPA) axis and release of catecholamines through adrenergic neurotransmission. Activation of these pathways results in a quick physical response to the stress and, in adaptive conditions, mediates long-term changes in the brain that lead to the formation of long-term memories of the experience. These long-term memories are an essential adaptive mechanism that allows an animal to effectively face similar demands again. Indeed, a moderate stress level has a strong positive effect on memory and cognition, as a single arousing or moderately stressful event can be remembered for up to a lifetime. Conversely, exposure to extreme, traumatic, or chronic stress can have the opposite effect and cause memory loss, cognitive impairments, and stress-related psychopathologies such as anxiety disorders, depression and post-traumatic stress disorder (PTSD). While more effort has been devoted to the understanding of the negative effects of chronic stress, much less has been done thus far on the identification of the mechanisms engaged in the brain when stress promotes long-term memory formation. Understanding these mechanisms will provide critical information for use in ameliorating memory processes in both normal and pathological conditions. Here, we will review the role of glucocorticoids and glucocorticoid receptors (GRs) in memory formation and modulation. Furthermore, we will discuss recent findings on the molecular cascade of events underlying the effect of GR activation in adaptive levels of stress that leads to strong, long-lasting memories. Our recent data indicate that the positive effects of GR activation on memory consolidation critically engage the brain-derived neurotrophic factor (BDNF) pathway. We propose and will discuss the hypothesis that stress promotes the formation of strong long-term memories because the activation of hippocampal GRs after learning is coupled to the recruitment of the growth and pro-survival BDNF/cAMP response element-binding protein (CREB) pathway, which is well-know to be a general mechanism required for long-term memory formation. We will then speculate about how these results may explain the negative effects of traumatic or chronic stress on memory and cognitive functions.
Collapse
|
65
|
Tao L, Zheng Y, Shen Z, Li Y, Tian X, Dou X, Qian J, Shen H. Psychological stress-induced lower serum zinc and zinc redistribution in rats. Biol Trace Elem Res 2013; 155:65-71. [PMID: 23975576 DOI: 10.1007/s12011-013-9762-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 07/11/2013] [Indexed: 12/29/2022]
Abstract
In humans, long-term exposure to uncontrollable and unpredictable life stressors is a major precipitant in the development of depressive disorders. There are strong evidences that depression is accompanied by lower serum zinc. The aim of present study is to assess the effects of repeated psychological stress (PS) on the zinc metabolism in rat. The rats were divided into control group and PS group which were subdivided into three subgroups: 7-day group, 14-day group, and recovery group (ten rats in each subgroup). PS model was created by a communication box which contains room A and room B. Rats in room A were only exposed to the responses of rats which were randomly given electrical shock for 30 min in room B. PS was given to rats for 30 min every morning for 14 days. The serum corticosterone (CORT), zinc in serum and tissues, and zinc apparent absorption after PS exposure were investigated. The results showed that the serum CORT increased and serum zinc decreased after 7 and 14 days of PS treatment. The zinc concentration in the liver was increased by 14 days PS exposure, whereas its concentration in the hippocampus was decreased by 7 and 14 days of PS exposure. There were no significant changes in zinc concentration in the heart, spleen, kidney, duodenum, cortex, and cerebellum. A decrease in the zinc apparent absorption was observed in the 7- and 14-day PS groups. The increased serum CORT and liver zinc concentrations and decreased serum zinc and apparent absorption of zinc recovered to normal concentrations 7 days away from PS exposure. The results suggest that PS could induce lower serum zinc, which might be correlated with decreased zinc absorption in the small intestine and increased liver zinc accumulation after PS exposure. The consequent effects of decreased hippocampal and serum zinc and increased CORT concentration after PS exposure on stress-related diseases await further research.
Collapse
Affiliation(s)
- Liping Tao
- Department of Military Hygiene, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
66
|
Yang S, Roselli F, Patchev AV, Yu S, Almeida OFX. Non-receptor-tyrosine kinases integrate fast glucocorticoid signaling in hippocampal neurons. J Biol Chem 2013; 288:23725-39. [PMID: 23818519 DOI: 10.1074/jbc.m113.470146] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Despite numerous descriptions of rapid effects of corticosterone on neuronal function, the intracellular mechanisms responsible for these changes remain elusive. The present comprehensive analysis reveals that signaling from a membrane-located G protein-coupled receptor activates PKC, Akt/PKB, and PKA, which subsequently trigger the phosphorylation of the tyrosine kinases Pyk2, Src, and Abl. These changes induce rapid cytoskeletal rearrangements (increased PSD-95 co-clustering) within the post-synaptic density; these events are accompanied by increased surface NMDA receptor expression, reflecting corticosterone-induced inhibition of NMDA receptor endocytosis. Notably, none of these signaling mechanisms require de novo protein synthesis. The observed up-regulation of ERK1/2 (downstream of NMDA receptor signaling) together with the fact that c-Abl integrates cytoplasmic and nuclear functions introduces a potential mechanism through which rapid signaling initiated at the plasma membrane may eventually determine the long term integrated response to corticosterone by impacting on the transcriptional machinery that is regulated by classical, nuclear mineralocorticoid, and glucocorticoid receptors.
Collapse
Affiliation(s)
- Silei Yang
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | | | | | | | | |
Collapse
|
67
|
Jeanneteau F, Chao MV. Are BDNF and glucocorticoid activities calibrated? Neuroscience 2013; 239:173-95. [PMID: 23022538 PMCID: PMC3581703 DOI: 10.1016/j.neuroscience.2012.09.017] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 09/04/2012] [Accepted: 09/06/2012] [Indexed: 12/22/2022]
Abstract
One hypothesis to account for the onset and severity of neurological disorders is the loss of trophic support. Indeed, changes in the levels and activities of brain-derived neurotrophic factor (BDNF) occur in numerous neurodegenerative and neuropsychiatric diseases. A deficit promotes vulnerability whereas a gain of function facilitates recovery by enhancing survival, synapse formation and synaptic plasticity. Implementation of 'BDNF therapies', however, faces numerous methodological and pharmacokinetic issues. Identifying BDNF mimetics that activate the BDNF receptor or downstream targets of BDNF signaling represent an alternative approach. One mechanism that shows great promise is to study the interplay of BDNF and glucocorticoid hormones, a major class of natural steroid secreted during stress reactions and in synchrony with circadian rhythms. While small amounts of glucocorticoids support normal brain function, excess stimulation by these steroid hormones precipitates stress-related affective disorders. To date, however, because of the paucity of knowledge of underlying cellular mechanisms, deleterious effects of glucocorticoids are not prevented following extreme stress. In the present review, we will discuss the complementary roles shared by BDNF and glucocorticoids in synaptic plasticity, and delineate possible signaling mechanisms mediating these effects.
Collapse
Affiliation(s)
- F Jeanneteau
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA.
| | | |
Collapse
|
68
|
Stress and excitatory synapses: from health to disease. Neuroscience 2013; 248:626-36. [PMID: 23727506 DOI: 10.1016/j.neuroscience.2013.05.043] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 05/01/2013] [Accepted: 05/21/2013] [Indexed: 01/20/2023]
Abstract
Individuals are exposed to stressful events in their daily life. The effects of stress on brain function ranges from highly adaptive to increasing the risk to develop psychopathology. For example, stressful experiences are remembered well which can be seen as a highly appropriate behavioral adaptation. On the other hand, stress is an important risk factor, in susceptible individuals, for depression and anxiety. An important question that remains to be addressed is how stress regulates brain function and what determines the threshold between adaptive and maladaptive responses. Excitatory synapses play a crucial role in synaptic transmission, synaptic plasticity and behavioral adaptation. In this review we discuss how brief and prolonged exposure to stress, in adulthood and early life, regulate the function of these synapses, and how these effects may contribute to behavioral adaptation and psychopathology.
Collapse
|
69
|
Jett JD, Morilak DA. Too much of a good thing: blocking noradrenergic facilitation in medial prefrontal cortex prevents the detrimental effects of chronic stress on cognition. Neuropsychopharmacology 2013; 38:585-95. [PMID: 23132268 PMCID: PMC3572455 DOI: 10.1038/npp.2012.216] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cognitive impairments associated with dysfunction of the medial prefrontal cortex (mPFC) are prominent in stress-related psychiatric disorders. We have shown that enhancing noradrenergic tone acutely in the rat mPFC facilitated extra-dimensional (ED) set-shifting on the attentional set-shifting test (AST), whereas chronic unpredictable stress (CUS) impaired ED. In this study, we tested the hypothesis that the acute facilitatory effect of norepinephrine (NE) in mPFC becomes detrimental when activated repeatedly during CUS. Using microdialysis, we showed that the release of NE evoked in mPFC by acute stress was unchanged at the end of CUS treatment. Thus, to then determine if repeated elicitation of this NE activity in mPFC during CUS may have contributed to the ED deficit, we infused a cocktail of α(1)-, β(1)-, and β(2)-adrenergic receptor antagonists into the mPFC prior to each CUS session, then tested animals drug free on the AST. Antagonist treatment prevented the CUS-induced ED deficit, suggesting that NE signaling during CUS compromised mPFC function. We confirmed that this was not attributable to sensitization of adrenergic receptor function following chronic antagonist treatment, by administering an additional microinjection into the mPFC immediately prior to ED testing. Acute antagonist treatment did not reverse the beneficial effects of chronic drug treatment during CUS, nor have any effect on baseline ED performance in chronic vehicle controls. Thus, we conclude that blockade of noradrenergic receptors in mPFC protected against the detrimental cognitive effects of CUS, and that repeated elicitation of noradrenergic facilitatory activity is one mechanism by which chronic stress may promote mPFC cognitive dysfunction.
Collapse
Affiliation(s)
- Julianne D Jett
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, USA
| | - David A Morilak
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, USA,Department of Pharmacology, MC 7764, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA, Tel: +1 210 567 4174, Fax: +1 210 567 4300, E-mail:
| |
Collapse
|
70
|
Liu W, Katz DA, Locke C, Daszkowski D, Wang Y, Rieser MJ, Awni W, Marek GJ, Dutta S. Clinical Safety, Pharmacokinetics, and Pharmacodynamics of the 11β-Hydroxysteroid Dehydrogenase Type 1 Inhibitor ABT-384 in Healthy Volunteers and Elderly Adults. Clin Pharmacol Drug Dev 2013; 2:133-51. [PMID: 27121668 DOI: 10.1002/cpdd.5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 10/10/2012] [Indexed: 11/10/2022]
Abstract
ABT-384 is a potent and selective inhibitor of 11β-hydroxysteroid dehydrogenase type 1 (HSD-1), the enzyme that regenerates cortisol in several tissues. Two clinical studies of ABT-384 were undertaken to assess its safety, pharmacokinetics, target engagement, and pharmacologic effects in healthy subjects. Single doses from 1 to 240 mg, and multiple doses from 1 to 100 mg once daily for 7-14 days, were administered to healthy adults. Multiple doses from 10 to 100 mg once daily for 21 days were administered to elderly subjects. A total of 103 subjects received at least 1 dose of ABT-384. A maximum-tolerated dose was not defined in either study. The pharmacokinetic profiles of ABT-384 and its active metabolite support once daily dosing. Analysis of urine cortisol metabolites demonstrated full hepatic HSD-1 inhibition with regimens from 1 mg daily, and confirmed in vitro target selectivity. Pharmacologic effects included increases of adrenocorticotrophic hormone levels, cortisol production and androgen and estradiol levels. ABT-384 has a wide therapeutic index relative to full hepatic target engagement which is relevant for indications such as diabetes and metabolic syndrome. Its therapeutic index for other potential indications such as Alzheimer's disease remains to be established.
Collapse
Affiliation(s)
- Wei Liu
- AbbVie, North Chicago, IL, USA
| | | | | | | | - Yi Wang
- AbbVie, North Chicago, IL, USA
| | | | | | | | | |
Collapse
|
71
|
Impaired structural hippocampal plasticity is associated with emotional and memory deficits in the olfactory bulbectomized rat. Neuroscience 2013; 236:233-43. [PMID: 23357118 DOI: 10.1016/j.neuroscience.2013.01.037] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 01/15/2013] [Accepted: 01/16/2013] [Indexed: 12/12/2022]
Abstract
Disturbances in olfactory circuitry have been associated with depression in humans. The olfactory bulbectomized (OBX lesion) has been largely used as a model of depression-like behavior in the rat. However, quantitative neuronal rearrangements in key brain regions in this animal model have not been evaluated yet. Accordingly, we investigated changes in hippocampal plasticity as well as behavioral deficits in this animal model. OBX-induced behavioral deficits were studied in a battery of tests, namely the open field test (OFT), forced swim test (FST), and spatial memory disturbances in the Morris water maze (MWM). To characterize the neuronal remodeling, neuroanatomical rearrangements were investigated in the CA1 hippocampus and piriform cortex (PirC), brain regions receiving inputs from the olfactory bulbs and associated with emotional or olfactory processes. Additionally, cell proliferation and survival of newborn cells in the adult dentate gyrus (DG) of the hippocampus were also determined. OBX induced hyperlocomotion and enhanced rearing and grooming in the OFT, increased immobility in the FST as well as required a longer time to find the hidden platform in the MWM. OBX also induced dendritic atrophy in the hippocampus and PirC. In addition, cell proliferation was decreased while the survival remained unchanged in the DG of these animals. These various features are also observed in depressed subjects, adding further support to the validity and usefulness of this model to evaluate potential novel antidepressants.
Collapse
|
72
|
Zhang X, Kan Q, Fu Y, Liu S, Dai Z, Dong Y. Noradrenergic activity regulated dexamethasone-induced increase of 5-HT3 receptor-mediated glutamate release in the rat's prelimbic cortex. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:2157-67. [DOI: 10.1016/j.bbamcr.2012.08.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 08/04/2012] [Accepted: 08/08/2012] [Indexed: 10/28/2022]
|
73
|
Stress-induced memory retrieval impairments: different time-course involvement of corticosterone and glucocorticoid receptors in dorsal and ventral hippocampus. Neuropsychopharmacology 2012; 37:2870-80. [PMID: 22948976 PMCID: PMC3499833 DOI: 10.1038/npp.2012.170] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The present study was aimed at determining the relative contribution of the dorsal (DH) and ventral (VH) hippocampus in stress-induced memory retrieval impairments. Thus, we studied the temporal involvement of corticosterone and its receptors, i.e. mineralocorticoid (MR) and glucocorticoid (GR) in the DH and VH, in relation with the time-course evolution of stress-induced memory retrieval impairments. In a first experiment, double microdialysis allowed showing on the same animal that an acute stress (electric footshocks) induced an earlier corticosterone rise in the DH (15-60 min post-stress) and then in the VH (90-105 min post-stress). The return to baseline was faster in the DH (105 min) than in the VH (120 min). Memory deficits assessed by delayed alternation occurred at 15-, 60-, and 105-min delays after stress and were closely related to the kinetic of corticosterone rises within the DH and VH. In a second experiment, the GR antagonist RU-38486 and the MR antagonist RU-28318 were administered in the DH or VH 15 min before stress. RU-38486 restored memory at 60 but not at 105 min post-stress delays in the DH, whereas the opposite pattern was observed in the VH. By contrast, RU-28318 had no effect on memory impairments at both the 60- and 105-min post-stress delays, showing that MR receptors are not involved at these delays. However, RU-28318 administered in the DH restored memory when administered at a shorter post-stress delay (15 min). Overall, our data are first to evidence that stress induces a functional switch from the DH to VH via different corticosterone time-course evolutions in these areas and the sequential GR receptors involvement in the DH and then in the VH, as regards the persistence of stress-induced memory retrieval deficits over time.
Collapse
|
74
|
Symonds CS, McKie S, Elliott R, William Deakin JF, Anderson IM. Detection of the acute effects of hydrocortisone in the hippocampus using pharmacological fMRI. Eur Neuropsychopharmacol 2012; 22:867-74. [PMID: 22521875 DOI: 10.1016/j.euroneuro.2012.03.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 02/27/2012] [Accepted: 03/24/2012] [Indexed: 01/09/2023]
Abstract
Impaired hippocampal function is believed to be important in the pathogenesis of depression. The hippocampus contains a high concentration of both mineralocorticoid (MR) and glucocorticoid receptors (GR), and the experimental administration of corticosteroids has been reported to mimic memory impairments seen in depression. Using pharmacological functional magnetic resonance imaging (phMRI) we investigated whether hippocampal function is altered after acute administration of hydrocortisone. Changes in BOLD signal following infusion of 100mg hydrocortisone given as a rapid intravenous bolus were measured in 14 healthy volunteers in a within-subject placebo-controlled crossover design. Subsequently, subjects completed an n-back task during an fMRI scan. Hydrocortisone infusion caused a significant, time-dependent increase in fMRI BOLD signal in hippocampus reaching a maximal effect at 11-19min. The n-back task increased BOLD signal in prefrontal and parietal cortical areas and decreased it in the hippocampus. After hydrocortisone the left hippocampal decrease in BOLD signal was attenuated with the magnitude of attenuation correlating with the increase seen after hydrocortisone infusion. No difference in behavioural task performance was observed. The results suggest acute hydrocortisone has rapid direct and modulatory influences on hippocampal function, probably acting through non-genomic GR or MR signalling. Hydrocortisone infusion phMRI may be a useful tool to investigate hippocampal corticosteroid receptor function in depression.
Collapse
Affiliation(s)
- Catherine S Symonds
- Neuroscience and Psychiatry Unit, The University of Manchester and Manchester Academic Health Sciences Centre, Manchester, United Kingdom.
| | | | | | | | | |
Collapse
|
75
|
McEwen BS. The ever-changing brain: cellular and molecular mechanisms for the effects of stressful experiences. Dev Neurobiol 2012; 72:878-90. [PMID: 21898852 DOI: 10.1002/dneu.20968] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The adult brain is capable of considerable structural and functional plasticity and the study of hormone actions in brain has contributed to our understanding of this important phenomenon. In particular, stress and stress-related hormones such as glucocorticoids and mineralocorticoids play a key role in the ability of acute and chronic stress to cause reversible remodeling of neuronal connections in the hippocampus, prefrontal cortex, and amygdala. To produce this plasticity, these hormones act by both genomic and non-genomic mechanisms together with ongoing, experience-driven neural activity mediated by excitatory amino acid neurotransmitters, neurotrophic factors such as brain derived neurotrophic factor, extracellular molecules such as neural cell adhesion molecule, neuropeptides such as corticotrophin releasing factor, and endocannabinoids. The result is a dynamic brain architecture that can be modified by experience. Under this view, the role of pharmaceutical agents, such as antidepressants, is to facilitate such plasticity that must also be guided by experiences.
Collapse
Affiliation(s)
- Bruce S McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York 10065, USA.
| |
Collapse
|
76
|
Krugers HJ, Karst H, Joels M. Interactions between noradrenaline and corticosteroids in the brain: from electrical activity to cognitive performance. Front Cell Neurosci 2012; 6:15. [PMID: 22509154 PMCID: PMC3321636 DOI: 10.3389/fncel.2012.00015] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 03/20/2012] [Indexed: 11/13/2022] Open
Abstract
One of the core reactions in response to a stressful situation is the activation of the hypothalamus-pituitary-adrenal axis which increases the release of glucocorticoid hormones from the adrenal glands. In concert with other neuro-modulators, such as (nor)adrenaline, these hormones enable and promote cognitive adaptation to stressful events. Recent studies have demonstrated that glucocorticoid hormones and noradrenaline, via their receptors, can both rapidly and persistently regulate the function of excitatory synapses which are critical for storage of information. Here we will review how glucocorticoids and noradrenaline alone and in synergy dynamically tune these synapses in the hippocampus and amygdala, and discuss how these hormones interact to promote behavioral adaptation to stressful situations.
Collapse
Affiliation(s)
- Harm J Krugers
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | | | | |
Collapse
|
77
|
Magariños AM, Li CJ, Gal Toth J, Bath KG, Jing D, Lee FS, McEwen BS. Effect of brain-derived neurotrophic factor haploinsufficiency on stress-induced remodeling of hippocampal neurons. Hippocampus 2012; 21:253-64. [PMID: 20095008 DOI: 10.1002/hipo.20744] [Citation(s) in RCA: 189] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Chronic restraint stress (CRS) induces the remodeling (i.e., retraction and simplification) of the apical dendrites of hippocampal CA3 pyramidal neurons in rats, suggesting that intrahippocampal connectivity can be affected by a prolonged stressful challenge. Since the structural maintenance of neuronal dendritic arborizations and synaptic connectivity requires neurotrophic support, we investigated the potential role of brain derived neurotrophic factor (BDNF), a neurotrophin enriched in the hippocampus and released from neurons in an activity-dependent manner, as a mediator of the stress-induced dendritic remodeling. The analysis of Golgi-impregnated hippocampal sections revealed that wild type (WT) C57BL/6 male mice showed a similar CA3 apical dendritic remodeling in response to three weeks of CRS to that previously described for rats. Haploinsufficient BDNF mice (BDNF(±) ) did not show such remodeling, but, even without CRS, they presented shorter and simplified CA3 apical dendritic arbors, like those observed in stressed WT mice. Furthermore, unstressed BDNF(±) mice showed a significant decrease in total hippocampal volume. The dendritic arborization of CA1 pyramidal neurons was not affected by CRS or genotype. However, only in WT mice, CRS induced changes in the density of dendritic spine shape subtypes in both CA1 and CA3 apical dendrites. These results suggest a complex role of BDNF in maintaining the dendritic and spine morphology of hippocampal neurons and the associated volume of the hippocampal formation. The inability of CRS to modify the dendritic structure of CA3 pyramidal neurons in BDNF(±) mice suggests an indirect, perhaps permissive, role of BDNF in mediating hippocampal dendritic remodeling.
Collapse
Affiliation(s)
- A M Magariños
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, USA.
| | | | | | | | | | | | | |
Collapse
|
78
|
Groeneweg FL, Karst H, de Kloet ER, Joëls M. Mineralocorticoid and glucocorticoid receptors at the neuronal membrane, regulators of nongenomic corticosteroid signalling. Mol Cell Endocrinol 2012; 350:299-309. [PMID: 21736918 DOI: 10.1016/j.mce.2011.06.020] [Citation(s) in RCA: 202] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 06/15/2011] [Accepted: 06/20/2011] [Indexed: 02/06/2023]
Abstract
The balance between corticosteroid actions induced via activation of the mineralocorticoid receptor (MR) and the glucocorticoid receptor (GR) determines the brain's response to stress. While both receptors are best known for their delayed genomic role, it has become increasingly evident that they can also associate with the plasma membrane and act as mediators of rapid, nongenomic signalling. Nongenomic corticosteroid actions in the brain are required for the coordination of a rapid adaptive response to stress; membrane-associated MRs and GRs play a major role herein. However, many questions regarding the underlying mechanism are still unresolved. How do MR and GR translocate to the membrane and what are their downstream signalling partners? In this review we discuss these issues based on insights obtained from related receptors, most notably the estrogen receptor α.
Collapse
Affiliation(s)
- Femke L Groeneweg
- Department of Medical Pharmacology, Leiden Amsterdam Centre for Drug Research, Leiden University Medical Centre, Leiden University, Einsteinweg 55, Leiden, The Netherlands.
| | | | | | | |
Collapse
|
79
|
Tse YC, Bagot RC, Wong TP. Dynamic regulation of NMDAR function in the adult brain by the stress hormone corticosterone. Front Cell Neurosci 2012; 6:9. [PMID: 22408607 PMCID: PMC3294281 DOI: 10.3389/fncel.2012.00009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 02/17/2012] [Indexed: 12/18/2022] Open
Abstract
Stress and corticosteroids dynamically modulate the expression of synaptic plasticity at glutamatergic synapses in the developed brain. Together with alpha-amino-3-hydroxy-methyl-4-isoxazole propionic acid receptors (AMPAR), N-methyl-D-aspartate receptors (NMDAR) are critical mediators of synaptic function and are essential for the induction of many forms of synaptic plasticity. Regulation of NMDAR function by cortisol/corticosterone (CORT) may be fundamental to the effects of stress on synaptic plasticity. Recent reports of the efficacy of NMDAR antagonists in treating certain stress-associated psychopathologies further highlight the importance of understanding the regulation of NMDAR function by CORT. Knowledge of how corticosteroids regulate NMDAR function within the adult brain is relatively sparse, perhaps due to a common belief that NMDAR function is stable in the adult brain. We review recent results from our laboratory and others demonstrating dynamic regulation of NMDAR function by CORT in the adult brain. In addition, we consider the issue of how differences in the early life environment may program differential sensitivity to modulation of NMDAR function by CORT and how this may influence synaptic function during stress. Findings from these studies demonstrate that NMDAR function in the adult hippocampus remains sensitive to even brief exposures to CORT and that the capacity for modulation of NMDAR may be programmed, in part, by the early life environment. Modulation of NMDAR function may contribute to dynamic regulation of synaptic plasticity and adaptation in the face of stress, however, enhanced NMDAR function may be implicated in mechanisms of stress-related psychopathologies including depression.
Collapse
Affiliation(s)
- Yiu Chung Tse
- Neuroscience Division, Douglas Mental Health University Institute, McGill University, Montreal QC, Canada
| | | | | |
Collapse
|
80
|
Involvement of glucocorticoid-mediated Zn2+ signaling in attenuation of hippocampal CA1 LTP by acute stress. Neurochem Int 2012; 60:394-9. [DOI: 10.1016/j.neuint.2012.01.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Revised: 01/12/2012] [Accepted: 01/18/2012] [Indexed: 01/14/2023]
|
81
|
Martisova E, Solas M, Horrillo I, Ortega JE, Meana JJ, Tordera RM, Ramírez MJ. Long lasting effects of early-life stress on glutamatergic/GABAergic circuitry in the rat hippocampus. Neuropharmacology 2012; 62:1944-53. [PMID: 22245561 DOI: 10.1016/j.neuropharm.2011.12.019] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 12/14/2011] [Accepted: 12/19/2011] [Indexed: 11/18/2022]
Abstract
The objective of the present work was to study the effects of an early-life stress (maternal separation, MS) in the excitatory/inhibitory ratio as a potential factor contributing to the ageing process, and the purported normalizing effects of chronic treatment with the antidepressant venlafaxine. MS induced depressive-like behaviour in the Porsolt forced swimming test that was reversed by venlafaxine, and that persisted until senescence. Aged MS rats showed a downregulation of vesicular glutamate transporter 1 and 2 (VGlut1 and VGlut2) and GABA transporter (VGAT) and increased expression of excitatory amino acid transporter 2 (EAAT2) in the hippocampus. Aged rats showed decreased expression of glutamic acid decarboxylase 65 (GAD65), while the excitatory amino acid transporter 1 (EAAT1) was affected only by stress. Glutamate receptor subunits NR1 and NR2A and GluR4 were upregulated in stressed rats, and this effect was reversed by venlafaxine. NR2B, GluR1 and GluR2/3 were not affected by either stress or age. MS, both in young and aged rats, induced an increase in the circulating levels of corticosterone. Corticosterone induced an increase glutamate and a decrease in GABA release in hippocampal slices, which was reversed by venlafaxine. Chronic treatment with corticosterone recapitulated the main biochemical findings observed in MS. The different effects that chronic stress exerts in young and adult animals on expression of proteins responsible for glutamate/GABA cycling may explain the involvement of glucocorticoids in ageing-related diseases. Modulation of glutamate/GABA release may be a relevant component of the therapeutic action of antidepressants, such as venlafaxine.
Collapse
Affiliation(s)
- Eva Martisova
- Department of Pharmacology, University of Navarra, C/ Irunlarrea 1, 31008 Pamplona, Spain
| | | | | | | | | | | | | |
Collapse
|
82
|
Mora F, Segovia G, Del Arco A, de Blas M, Garrido P. Stress, neurotransmitters, corticosterone and body-brain integration. Brain Res 2012; 1476:71-85. [PMID: 22285436 DOI: 10.1016/j.brainres.2011.12.049] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 12/22/2011] [Accepted: 12/23/2011] [Indexed: 12/26/2022]
Abstract
Stress can be defined as a brain-body reaction towards stimuli arising from the environment or from internal cues that are interpreted as a disruption of homeostasis. The organization of the response to a stressful situation involves not only the activity of different types of neurotransmitter systems in several areas of the limbic system, but also the response of neurons in these areas to several other chemicals and hormones, chiefly glucocorticoids, released from peripheral organs and glands. Thus, stress is probably the process through which body-brain integration plays a major role. Here we review first the responses to an acute stress in terms of neurotransmitters such as dopamine, acetylcholine, glutamate and GABA in areas of the brain involved in the regulation of stress responses. These areas include the prefrontal cortex, amygdala, hippocampus and nucleus accumbens and the interaction among those areas. Then, we consider the role of glucocorticoids and review some recent data about the interaction of these steroids with several neurotransmitters in those same areas of the brain. Also the actions of other substances (neuromodulators) released from peripheral organs such as the pancreas, liver or gonads (insulin, IGF-1, estrogens) are reviewed. The role of an environmental enrichment on these same responses is also discussed. Finally a section is devoted to put into perspective all these environmental-brain-body-brain interactions during stress and their consequences on aging. It is concluded that the integrative perspective framed in this review is relevant for better understanding of how the organism responds to stressful challenges and how this can be modified through different environmental conditions during the process of aging. This article is part of a Special Issue entitled: Brain Integration.
Collapse
Affiliation(s)
- Francisco Mora
- Department of Physiology, Faculty of Medicine, Universidad Complutense, Madrid, Spain.
| | | | | | | | | |
Collapse
|
83
|
Takeda A, Tamano H. Proposed glucocorticoid-mediated zinc signaling in the hippocampus. Metallomics 2012; 4:614-8. [DOI: 10.1039/c2mt20018j] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
84
|
Wang M, Hill MN, Zhang L, Gorzalka BB, Hillard CJ, Alger BE. Acute restraint stress enhances hippocampal endocannabinoid function via glucocorticoid receptor activation. J Psychopharmacol 2012; 26:56-70. [PMID: 21890595 PMCID: PMC3373303 DOI: 10.1177/0269881111409606] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Exposure to behavioural stress normally triggers a complex, multilevel response of the hypothalamic-pituitary-adrenal (HPA) axis that helps maintain homeostatic balance. Although the endocannabinoid (eCB) system (ECS) is sensitive to chronic stress, few studies have directly addressed its response to acute stress. Here we show that acute restraint stress enhances eCB-dependent modulation of GABA release measured by whole-cell voltage clamp of inhibitory postsynaptic currents (IPSCs) in rat hippocampal CA1 pyramidal cells in vitro. Both Ca(2+)-dependent, eCB-mediated depolarization-induced suppression of inhibition (DSI), and muscarinic cholinergic receptor (mAChR)-mediated eCB mobilization are enhanced following acute stress exposure. DSI enhancement is dependent on the activation of glucocorticoid receptors (GRs) and is mimicked by both in vivo and in vitro corticosterone treatment. This effect does not appear to involve cyclooxygenase-2 (COX-2), an enzyme that can degrade eCBs; however, treatment of hippocampal slices with the L-type calcium (Ca(2+)) channel inhibitor, nifedipine, reverses while an agonist of these channels mimics the effect of in vivo stress. Finally, we find that acute stress produces a delayed (by 30 min) increase in the hippocampal content of 2-arachidonoylglycerol, the eCB responsible for DSI. These results support the hypothesis that the ECS is a biochemical effector of glucocorticoids in the brain, linking stress with changes in synaptic strength.
Collapse
Affiliation(s)
- Meina Wang
- Department of Physiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA 21201
| | - Matthew N. Hill
- Department of Psychology, University of British Columbia, Vancouver, BC Canada
| | - Longhua Zhang
- Department of Physiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA 21201
| | - Boris B. Gorzalka
- Department of Psychology, University of British Columbia, Vancouver, BC Canada
| | - Cecilia J. Hillard
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA 53226
| | - Bradley E. Alger
- Department of Physiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA 21201
| |
Collapse
|
85
|
Dorey R, Piérard C, Shinkaruk S, Tronche C, Chauveau F, Baudonnat M, Béracochéa D. Membrane mineralocorticoid but not glucocorticoid receptors of the dorsal hippocampus mediate the rapid effects of corticosterone on memory retrieval. Neuropsychopharmacology 2011; 36:2639-49. [PMID: 21814189 PMCID: PMC3230488 DOI: 10.1038/npp.2011.152] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
This study was aimed at determining the type of the glucocorticoid membrane receptors (mineralocorticoid receptors (MRs) or glucocorticoid receptors (GRs)) in the dorsal hippocampus (dHPC) involved in the rapid effects of corticosterone or stress on memory retrieval. For that purpose, we synthesized corticosterone-3-O-carboxymethyloxime-bovine serum albumin conjugate (Cort-3CMO-BSA) conjugate (a high MW complex that cannot cross the cell membrane) totally devoid of free corticosterone, stable in physiological conditions. In a first experiment, we evidenced that an acute stress (electric footshocks) induced both a dHPC corticosterone rise measured by microdialysis and memory retrieval impairment on delayed alternation task. Both the endocrinal and cognitive effects of stress were blocked by metyrapone (a corticosterone synthesis inhibitor). In a second experiment, we showed that bilateral injections of either corticosterone or Cort-3CMO-BSA in dHPC 15 min before memory testing produced impairments similar to those resulting from acute stress. Furthermore, we showed that anisomycin (a protein synthesis inhibitor) failed to block the deleterious effect of Cort-3CMO-BSA on memory. In a third experiment, we evidenced that intra-hippocampal injection of RU-28318 (MR antagonist) but not of RU-38486 (GR antagonist) totally blocked the Cort-3CMO-BSA-induced memory retrieval deficit. In a fourth experiment, we demonstrated that RU-28318 administered 15 min before stress blocked the stress-induced memory impairments when behavioral testing occurred 15 min but not 60 min after stress. Overall, this study provides strong in vivo evidence that the dHPC membrane GRs, mediating the rapid and non-genomic effects of acute stress on memory retrieval, are of MR but not GR type.
Collapse
Affiliation(s)
- Rodolphe Dorey
- CNRS, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA) UMR 5287, Universités de Bordeaux, Talence, France,Institut de Recherche Biomédicale des Armées (IRBA)—Unité Fatigue et Vigilance, Brétigny sur Orge, France
| | - Christophe Piérard
- Institut de Recherche Biomédicale des Armées (IRBA)—Unité Fatigue et Vigilance, Brétigny sur Orge, France
| | - Svitlana Shinkaruk
- U862 Inserm Physiopathologie de la plasticité neuronale, Institut François Magendie, Universités de Bordeaux, Bordeaux, France,ENITA de Bordeaux, Gradignan, France
| | - Christophe Tronche
- CNRS, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA) UMR 5287, Universités de Bordeaux, Talence, France,Institut de Recherche Biomédicale des Armées (IRBA)—Unité Fatigue et Vigilance, Brétigny sur Orge, France
| | - Frédéric Chauveau
- Institut de Recherche Biomédicale des Armées (IRBA)—Unité Fatigue et Vigilance, Brétigny sur Orge, France
| | - Mathieu Baudonnat
- CNRS, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA) UMR 5287, Universités de Bordeaux, Talence, France
| | - Daniel Béracochéa
- CNRS, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA) UMR 5287, Universités de Bordeaux, Talence, France,CNRS, Institut de Neurosciences Cognitives et Integratives d'Aquitaine (INCIA) UMR 5287, Universités de Bordeaux, Talence 33400, France. Tel: +33 54 000 2439; Fax: +33 54 000 8743; E-mail:
| |
Collapse
|
86
|
The stressed synapse: the impact of stress and glucocorticoids on glutamate transmission. Nat Rev Neurosci 2011; 13:22-37. [PMID: 22127301 DOI: 10.1038/nrn3138] [Citation(s) in RCA: 971] [Impact Index Per Article: 69.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mounting evidence suggests that acute and chronic stress, especially the stress-induced release of glucocorticoids, induces changes in glutamate neurotransmission in the prefrontal cortex and the hippocampus, thereby influencing some aspects of cognitive processing. In addition, dysfunction of glutamatergic neurotransmission is increasingly considered to be a core feature of stress-related mental illnesses. Recent studies have shed light on the mechanisms by which stress and glucocorticoids affect glutamate transmission, including effects on glutamate release, glutamate receptors and glutamate clearance and metabolism. This new understanding provides insights into normal brain functioning, as well as the pathophysiology and potential new treatments of stress-related neuropsychiatric disorders.
Collapse
|
87
|
Krugers HJ, Zhou M, Joëls M, Kindt M. Regulation of excitatory synapses and fearful memories by stress hormones. Front Behav Neurosci 2011; 5:62. [PMID: 22013419 PMCID: PMC3190121 DOI: 10.3389/fnbeh.2011.00062] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2011] [Accepted: 09/05/2011] [Indexed: 12/18/2022] Open
Abstract
Memories for emotionally arousing and fearful events are generally well retained. From the evolutionary point of view this is a highly adaptive behavioral response aimed to remember relevant information. However, fearful memories can also be inappropriately and vividly (re)expressed, such as in posttraumatic stress disorder. The memory formation of emotionally arousing events is largely modulated by hormones, peptides, and neurotransmitters which are released during and after exposure to these conditions. One of the core reactions in response to a stressful situation is the rapid activation of the autonomic nervous system, which results in the release of norepinephrine in the brain. In addition, stressful events stimulate the hypothalamus-pituitary-adrenal axis which slowly increases the release of glucocorticoid hormones from the adrenal glands. Here we will review how glucocorticoids and norepinephrine regulate the formation of fearful memories in rodents and humans and how these hormones can facilitate the storage of information by regulating excitatory synapses.
Collapse
Affiliation(s)
- Harm J. Krugers
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, Netherlands
| | - Ming Zhou
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, Netherlands
| | - Marian Joëls
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, Netherlands
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, University Medical Center UtrechtUtrecht, Netherlands
| | - Merel Kindt
- Department of Clinical Psychology, University of AmsterdamAmsterdam, Netherlands
| |
Collapse
|
88
|
Farley S, Dumas S, El Mestikawy S, Giros B. Increased expression of the Vesicular Glutamate Transporter-1 (VGLUT1) in the prefrontal cortex correlates with differential vulnerability to chronic stress in various mouse strains: effects of fluoxetine and MK-801. Neuropharmacology 2011; 62:503-17. [PMID: 21945287 DOI: 10.1016/j.neuropharm.2011.09.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 09/07/2011] [Accepted: 09/08/2011] [Indexed: 12/27/2022]
Abstract
Major depression is a chronic psychiatric illness that is highly prevalent and disabling. The available medications are ineffective for many patients suggesting that differents molecular pathways could be specifically altered in the unresponsive patients. Recently, the glutamatergic system has emerged as a target in the research on depression and acute NMDA receptor blockade has been shown to produce strong antidepressant effects. We have studied the adaptations of four mice strains (C57BL/6, DBA/2, C3H and BALB/c) to a chronic unpredictable stress protocol, a widely used model of depression in rodents. BALB/c mice displayed strikingly different behavioral and neurochemical adaptations compared to the other strains tested, suggesting that different molecular pathways are involved in their specific vulnerability. They became hyperactive during the dark period, anhedonic-like and displayed no alterations in the tail suspension test (TST). After chronic stress, only the BALB/c displayed an increased frontocortical VGLUT1 expression which is suggestive of a dysregulation of their prefrontal glutamatergic system, and no BDNF mRNA alteration, although the acute stress modulation of this mRNA is similar to the other strains. Chronic administration of an antagonist of NMDA receptors, MK-801, induced antidepressant-like effects in the TST for stressed BALB/c, but was ineffective for the hyperactivity and anhedonia-like behavior, in contrast to fluoxetine. Chronic MK-801 was totally inactive on the behavior of stressed C57BL/6 mice. MK-801, but not fluoxetine, inhibited the VGLUT1 prefrontal increase in BALB/c. Fluoxetine increased VGLUT1 and BDNF mRNA expression in the hippocampus of the C57BL/6 but not in the BALB/c strain, suggesting a different reactivity in-between strain to both stress and antidepressant. Interestingly enough, the BDNF or VGLUT1 increase is not necessary to reverse the stress induced behavioral alterations in our experimental settings. This observation supports the conclusion that BDNF and VGLUT1 are depressive state markers, but not involved in its etiology. Finally, there is a substantial similarity between the phenotypes that are observed in the BALB/c mice and endogenous depression in humans, as well as between C57BL/6 mice and atypical depression. To have a better understanding of the variability of depression etiologies in human, and the implication of the glutamatergic system, it may be suggested that future animal studies in the mouse would systematically compare the two strains BALB/c and C57BL/6 for the identification of relevant biological mechanisms. This article is part of a special Issue entitled 'Anxiety and Depression'.
Collapse
Affiliation(s)
- Séverine Farley
- INSERM U952 Physiopathologie des Maladies du Système nerveux Central, 9 Quai St Bernard, 75005 Paris, France
| | | | | | | |
Collapse
|
89
|
Towards a glutamate hypothesis of depression: an emerging frontier of neuropsychopharmacology for mood disorders. Neuropharmacology 2011; 62:63-77. [PMID: 21827775 DOI: 10.1016/j.neuropharm.2011.07.036] [Citation(s) in RCA: 779] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Revised: 07/22/2011] [Accepted: 07/23/2011] [Indexed: 12/12/2022]
Abstract
Half a century after the first formulation of the monoamine hypothesis, compelling evidence implies that long-term changes in an array of brain areas and circuits mediating complex cognitive-emotional behaviors represent the biological underpinnings of mood/anxiety disorders. A large number of clinical studies suggest that pathophysiology is associated with dysfunction of the predominant glutamatergic system, malfunction in the mechanisms regulating clearance and metabolism of glutamate, and cytoarchitectural/morphological maladaptive changes in a number of brain areas mediating cognitive-emotional behaviors. Concurrently, a wealth of data from animal models have shown that different types of environmental stress enhance glutamate release/transmission in limbic/cortical areas and exert powerful structural effects, inducing dendritic remodeling, reduction of synapses and possibly volumetric reductions resembling those observed in depressed patients. Because a vast majority of neurons and synapses in these areas and circuits use glutamate as neurotransmitter, it would be limiting to maintain that glutamate is in some way 'involved' in mood/anxiety disorders; rather it should be recognized that the glutamatergic system is a primary mediator of psychiatric pathology and, potentially, also a final common pathway for the therapeutic action of antidepressant agents. A paradigm shift from a monoamine hypothesis of depression to a neuroplasticity hypothesis focused on glutamate may represent a substantial advancement in the working hypothesis that drives research for new drugs and therapies. Importantly, despite the availability of multiple classes of drugs with monoamine-based mechanisms of action, there remains a large percentage of patients who fail to achieve a sustained remission of depressive symptoms. The unmet need for improved pharmacotherapies for treatment-resistant depression means there is a large space for the development of new compounds with novel mechanisms of action such as glutamate transmission and related pathways. This article is part of a Special Issue entitled 'Anxiety and Depression'.
Collapse
|
90
|
McEwen BS, Eiland L, Hunter RG, Miller MM. Stress and anxiety: structural plasticity and epigenetic regulation as a consequence of stress. Neuropharmacology 2011; 62:3-12. [PMID: 21807003 DOI: 10.1016/j.neuropharm.2011.07.014] [Citation(s) in RCA: 384] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 07/09/2011] [Accepted: 07/13/2011] [Indexed: 12/31/2022]
Abstract
The brain is the central organ of stress and adaptation to stress because it perceives and determines what is threatening, as well as the behavioral and physiological responses to the stressor. The adult, as well as developing brain, possess a remarkable ability to show reversible structural and functional plasticity in response to stressful and other experiences, including neuronal replacement, dendritic remodeling, and synapse turnover. This is particularly evident in the hippocampus, where all three types of structural plasticity have been recognized and investigated, using a combination of morphological, molecular, pharmacological, electrophysiological and behavioral approaches. The amygdala and the prefrontal cortex, brain regions involved in anxiety and fear, mood, cognitive function and behavioral control, also show structural plasticity. Acute and chronic stress cause an imbalance of neural circuitry subserving cognition, decision making, anxiety and mood that can increase or decrease expression of those behaviors and behavioral states. In the short term, such as for increased fearful vigilance and anxiety in a threatening environment, these changes may be adaptive; but, if the danger passes and the behavioral state persists along with the changes in neural circuitry, such maladaptation may need intervention with a combination of pharmacological and behavioral therapies, as is the case for chronic or mood anxiety disorders. We shall review cellular and molecular mechanisms, as well as recent work on individual differences in anxiety-like behavior and also developmental influences that bias how the brain responds to stressors. Finally, we suggest that such an approach needs to be extended to other brain areas that are also involved in anxiety and mood. This article is part of a Special Issue entitled 'Anxiety and Depression'.
Collapse
Affiliation(s)
- Bruce S McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| | | | | | | |
Collapse
|
91
|
Stress by noise produces differential effects on the proliferation rate of radial astrocytes and survival of neuroblasts in the adult subgranular zone. Neurosci Res 2011; 70:243-50. [DOI: 10.1016/j.neures.2011.03.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 03/28/2011] [Accepted: 03/31/2011] [Indexed: 01/22/2023]
|
92
|
Takeda A, Tamano H. Zinc signaling through glucocorticoid and glutamate signaling in stressful circumstances. J Neurosci Res 2011; 88:3002-10. [PMID: 20568287 DOI: 10.1002/jnr.22456] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Humans and animals are constantly exposed to environmental stress. The hypothalamic-pituitary-adrenal (HPA) axis responds to stress, followed by glucocorticoid secretion from the adrenal glands. This response serves to maintain homeostasis in the living body through energy mobilization or to restore it. The brain is an important target for glucocorticoids. The hippocampus participates in the regulation of the HPA axis. Stress activates glutamatergic neurons in the hippocampus, and serious stress induces dyshomeostasis of extracellular glutamate. This dyshomeostasis, which is potentiated by glucocorticoids, modifies cognitive and emotional behavior. On the other hand, zinc is necessary for glucocorticoid signaling and is released from glutamatergic (zincergic) neurons to modulate synaptic glutamate signaling. Stress also induces dyshomeostasis of extracellular zinc, which may be linked to dyshomeostasis of extracellular glutamate. Thus, glucocorticoid signaling might also contribute to dyshomeostasis of extracellular zinc. It is likely that zinc signaling participates in cognitive and emotional behavior through glucocorticoid and glutamate signaling under stressful circumstances. This Mini-Review analyzes the relationship among signals of glucocorticoid, glutamate, and zinc under stressful circumstances to elucidate the significance of the zinc signaling in response to stress.
Collapse
Affiliation(s)
- Atsushi Takeda
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan.
| | | |
Collapse
|
93
|
Prins J, Olivier B, Korte SM. Triple reuptake inhibitors for treating subtypes of major depressive disorder: the monoamine hypothesis revisited. Expert Opin Investig Drugs 2011; 20:1107-30. [DOI: 10.1517/13543784.2011.594039] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
94
|
Musazzi L, Racagni G, Popoli M. Stress, glucocorticoids and glutamate release: effects of antidepressant drugs. Neurochem Int 2011; 59:138-49. [PMID: 21689704 DOI: 10.1016/j.neuint.2011.05.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 05/26/2011] [Accepted: 05/30/2011] [Indexed: 12/21/2022]
Abstract
Stressful life events impact on memory, cognition and emotional responses, and are known to precipitate mood/anxiety disorders. It is increasingly recognized that stress and its neurochemical and endocrine mediators induce changes in glutamate synapses and circuitry, and this in turn modify mental states. Half a century after the monoamine hypothesis, it is widely accepted that maladaptive changes in excitatory/inhibitory circuitry have a primary role in the pathophysiology of mood/anxiety disorders. The neuroplasticity hypothesis posits that volumetric changes consistently found in limbic and cortical areas of depressed subjects are in good part due to remodeling of neuronal dendritic arbors and loss of synaptic spines. A considerable body of work, carried out with in vivo microdialysis as well as alternative methodologies, has shown that both stress and corticosterone treatment induce enhancement of activity-dependent glutamate release. Accordingly, results from preclinical studies suggest that stress- and glucocorticoid-induced enhancement of glutamate release and transmission plays a main role in the induction of maladaptive cellular effects, in turn responsible for dendritic remodeling. Additional recent work has showed that drugs employed for therapy of mood/anxiety disorders (antidepressants) prevent the enhancement of glutamate release induced by stress. Understanding the action of traditional drugs on glutamate transmission could be of great help in developing drugs that may work directly at this level.
Collapse
Affiliation(s)
- Laura Musazzi
- Center of Neuropharmacology, Department of Pharmacological Sciences, University of Milano, Italy
| | | | | |
Collapse
|
95
|
Acute inhibition of 11beta-hydroxysteroid dehydrogenase type-1 improves memory in rodent models of cognition. J Neurosci 2011; 31:5406-13. [PMID: 21471376 DOI: 10.1523/jneurosci.4046-10.2011] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mounting evidence suggests excessive glucocorticoid activity may contribute to Alzheimer's disease (AD) and age-associated memory impairment. 11β-hydroxysteroid dehydrogenase type-1 (HSD1) regulates conversion of glucocorticoids from inactive to active forms. HSD1 knock-out mice have improved cognition, and the nonselective inhibitor carbenoxolone improved verbal memory in elderly men. Together, these data suggest that HSD1 inhibition may be a potential therapy for cognitive deficits, such as those associated with AD. To investigate this, we characterized two novel and selective HSD1 inhibitors, A-918446 and A-801195. Learning, memory consolidation, and recall were evaluated in mouse 24 h inhibitory avoidance. Inhibition of brain cortisol production and phosphorylation of cAMP response element-binding protein (CREB), a transcription factor involved in cognition, were also examined. Rats were tested in a short-term memory model, social recognition, and in a separate group cortical and hippocampal acetylcholine release was measured via in vivo microdialysis. Acute treatment with A-801195 (10-30 mg/kg) or A-918446 (3-30 mg/kg) inhibited cortisol production in the ex vivo assay by ∼ 35-90%. Acute treatment with A-918446 improved memory consolidation and recall in inhibitory avoidance and increased CREB phosphorylation in the cingulate cortex. Acute treatment with A-801195 significantly improved short-term memory in rat social recognition that was not likely due to alterations of the cholinergic system, as acetylcholine release was not increased in a separate set of rats. These studies suggest that selective HSD1 inhibitors work through a novel, noncholinergic mechanism to facilitate cognitive processing.
Collapse
|
96
|
Effects of early life stress on neuroendocrine and neurobehavior: mechanisms and implications. Pediatr Neonatol 2011; 52:122-9. [PMID: 21703552 DOI: 10.1016/j.pedneo.2011.03.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 11/30/2010] [Accepted: 12/23/2010] [Indexed: 12/12/2022] Open
Abstract
Evidence continues to mount that adverse experiences early in life have an impact on brain functions. Early life stress can program the development of the hypothalamic-pituitary-adrenal axis and cause alterations of neurochemistry and signaling pathways involved in regulating neuroplasticity, with resultant neurobehavioral changes. Early life experiences and genetic factors appear to interact in determining the individual vulnerability to mental health disorders. We reviewed the effects of early life stress on neuroendocrine regulation and the relevance to neurobehavioral development.
Collapse
|
97
|
Serotonergic neurotransmission in the ventral hippocampus is enhanced by corticosterone and altered by chronic amphetamine treatment. Neuroscience 2011; 182:105-14. [PMID: 21420472 DOI: 10.1016/j.neuroscience.2011.03.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 03/14/2011] [Accepted: 03/14/2011] [Indexed: 01/13/2023]
Abstract
The ventral hippocampus modulates anxiety-like behavior in rats, and serotonergic transmission within the hippocampus facilitates adaptation to stress. Chronic amphetamine treatment results in anxiety-like behavior in rats and reduced monoamine concentrations in the ventral hippocampus. Since reduced hippocampal serotonergic transmission in response to stress is observed in rats that display high anxiety-like behavior, anxiety states in amphetamine-treated rats may be associated with reduced stress-related serotonergic transmission in the hippocampus. Therefore, using in vivo microdialysis in anesthetized rats, we investigated the effect of corticosterone infused locally into the ventral hippocampus on serotonergic transmission, and the effect of chronic amphetamine pretreatment on corticosteroid receptor protein expression and the corticosterone-induced serotonergic response. Extracellular serotonin in the ventral hippocampus was increased by corticosterone in drug naive rats, and this corticosterone-induced serotonin augmentation was blocked by the glucocorticoid receptor antagonist mifepristone. Furthermore, chronic pretreatment with amphetamine abolished the serotonin response to physiologically relevant corticosterone levels and reduced glucocorticoid receptor protein expression. Together, our results suggest that chronic amphetamine exposure reduces serotonergic neurotransmission, in part via alterations to glucocorticoid receptor-facilitation of serotonin release in the rat ventral hippocampus. Reduced serotonergic activity in the ventral hippocampus may contribute to altered stress responses and adaptive coping following repeated drug exposure.
Collapse
|
98
|
Cazakoff BN, Howland JG. Acute stress disrupts paired pulse facilitation and long-term potentiation in rat dorsal hippocampus through activation of glucocorticoid receptors. Hippocampus 2011; 20:1327-31. [PMID: 20043285 DOI: 10.1002/hipo.20738] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cognitive functions such as learning and memory are widely believed to depend on patterns of short- and long-term synaptic plasticity. Factors, such as acute stress, which affect learning and memory, may do so by altering patterns of synaptic plasticity in distinct neural circuits. Numerous studies have examined the effects of acute stress on long-term synaptic plasticity; however, few have examined its influence on short-term plasticity. The present experiments directly assessed the effects of acute stress on short-term synaptic plasticity as measured by paired pulse facilitation (PPF) of excitatory postsynaptic potentials recorded from rat dorsal hippocampus (dHip) in vivo. Long-term potentiation (LTP) was also examined. Acute stress induced by exposure to an elevated platform impaired PPF and LTP in the dHip. Pretreatment of rats exposed to stress with mifepristone (RU38486; 10 mg kg⁻¹) blocked the stress-induced impairment of both PPF and LTP. These data demonstrate that activation of glucocorticoid receptors during acute stress disrupts normal patterns of both PPF and LTP in the dHip.
Collapse
Affiliation(s)
- Brittany N Cazakoff
- Department of Physiology, Neural Systems and Plasticity Research Group, University of Saskatchewan, 9 Campus Drive, Saskatoon, SK, S7N 5A5, Canada
| | | |
Collapse
|
99
|
Sandi C. Glucocorticoids act on glutamatergic pathways to affect memory processes. Trends Neurosci 2011; 34:165-76. [PMID: 21377221 DOI: 10.1016/j.tins.2011.01.006] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2010] [Revised: 01/24/2011] [Accepted: 01/24/2011] [Indexed: 12/22/2022]
Abstract
Glucocorticoids can acutely affect memory processes, with both facilitating and impairing effects having been described. Recent work has revealed that glucocorticoids may affect learning and memory processes by interacting with glutamatergic mechanisms. In this opinion article I describe different glutamatergic pathways that glucocorticoids can affect to modulate memory processes. Furthermore, glucocorticoid-glutamatergic interactions during information processing are proposed as a potential model to explain many of the diverse actions of glucocorticoids on cognition. The model suggests that direct modulation of glutamatergic pathways by glucocorticoids could serve as an important mechanism for these hormones to directly alter cognitive functions.
Collapse
Affiliation(s)
- Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
100
|
Chronic stress and lithium treatments alter hippocampal glutamate uptake and release in the rat and potentiate necrotic cellular death after oxygen and glucose deprivation. Neurochem Res 2011; 36:793-800. [PMID: 21253855 DOI: 10.1007/s11064-011-0404-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2011] [Indexed: 01/02/2023]
Abstract
This study was undertaken to evaluate the effects of chronic variate stress and lithium treatment on glutamatergic activity and neuronal vulnerability of rat hippocampus. Male Wistar rats were simultaneously treated with lithium and submitted to a chronic variate stress protocol during 40 days, and afterwards the hippocampal glutamatergic uptake and release, measured in slices and synaptosomes, were evaluated. We observed an increased synaptosomal [(3)H]glutamate uptake and an increase in [(3)H]glutamate stimulated release in hippocampus of lithium-treated rats. Chronic stress increased basal [(3)H]glutamate release by synaptosomes, and decreased [(3)H]glutamate uptake in hippocampal slices. When evaluating cellular vulnerability, both stress and lithium increased cellular death after oxygen and glucose deprivation (OGD). We suggest that the manipulation of glutamatergic activity induced by stress may be in part responsible for the neuroendangerment observed after stress exposure, and that, in spite of the described neuroprotective effects of lithium, it increased the neuronal vulnerability after OGD.
Collapse
|