Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Psychoneuroendocrinology ; 146: 105945, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36183622

ABSTRACT

Post-traumatic stress disorder (PTSD) is characterized by the co-existence of a persistent strong memory of the traumatic experience and amnesia for the peritraumatic context. Most animal models, however, fail to account for the contextual amnesia which is considered to play a critical role in the etiology of PTSD intrusive memories. It is also unclear how aging affects PTSD-like memory. Glucocorticoids alter the formation and retention of fear-associated memory. Here, we investigated whether a deficiency of 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) (an intracellular glucocorticoid generating enzyme) and aging modulates fear conditioning and PTSD-like memory in mice. We first measured memory in 6 months and 24 months old 11ß-HSD1 deficient (HSD1 KO) and wildtype (WT) mice following paired tone-shock fear conditioning. Then, separate groups of mice were exposed to restraint stress immediately after unpaired tone-shock contextual fear conditioning. Compared with young controls, aged WT mice exhibited enhanced auditory cued fear memory, but contextual fear memory was not different. Contextual fear memory retention was attenuated in both young and aged HSD1 KO mice. In contrast, auditory cued fear memory was reduced 24 h after training only in aged HSD1 KO mice. When fear conditioned with stress, WT mice displayed PTSD-like memory (i.e., increased fear to tone not predictive of shock and reduced fear to 'aversive' conditioning context); this was unchanged with aging. In contrast, young HSD1 KO mice fear conditioned with stress showed normal fear memory (i.e., increased fear response to conditioning context), as observed in WT mice fear conditioned alone. While aged HSD1 KO mice fear conditioned with stress also displayed normal contextual fear memory, the fear response to the 'safe' tone remained. Thus, a deficiency of 11ß-HSD1 protects against both amnesia for the conditioning context and hypermnesia for a salient tone in young adult mice but only contextual amnesia is prevented in aged mice. These results suggest that brain 11ß-HSD1 generated glucocorticoids make a significant contribution to fear conditioning and PTSD-like memory. 11ß-HSD1 inhibition may be useful in prevention and/or treatment of PTSD.

2.
Psychoneuroendocrinology ; 89: 13-22, 2018 03.
Article in English | MEDLINE | ID: mdl-29306773

ABSTRACT

Chronic exposure to stress during midlife associates with subsequent age-related cognitive decline and may increase the vulnerability to develop psychiatric conditions. Increased hypothalamic-pituitary-adrenal (HPA) axis activity has been implicated in pathogenesis though any causative role for glucocorticoids is unestablished. This study investigated the contribution of local glucocorticoid regeneration by the intracellular enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1), in persisting midlife stress-induced behavioral effects in mice. Middle-aged (10 months old) 11ß-HSD1-deficient mice and wild-type congenic controls were randomly assigned to 28 days of chronic unpredictable stress or left undisturbed (non-stressed). All mice underwent behavioral testing at the end of the stress/non-stress period and again 6-7 months later. Chronic stress impaired spatial memory in middle-aged wild-type mice. The effects, involving a wide spectrum of behavioral modalities, persisted for 6-7 months after cessation of stress into early senescence. Enduring effects after midlife stress included impaired spatial memory, enhanced contextual fear memory, impaired fear extinction, heightened anxiety, depressive-like behavior, as well as reduced hippocampal glucocorticoid receptor mRNA expression. In contrast, 11ß-HSD1 deficient mice resisted both immediate and enduring effects of chronic stress, despite similar stress-induced increases in systemic glucocorticoid activity during midlife stress. In conclusion, chronic stress in midlife exerts persisting effects leading to cognitive and affective dysfunction in old age via mechanisms that depend, at least in part, on brain glucocorticoids generated locally by 11ß-HSD1. This finding supports selective 11ß-HSD1 inhibition as a novel therapeutic target to ameliorate the long-term consequences of stress-related psychiatric disorders in midlife.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Affect/physiology , Stress, Psychological/physiopathology , Animals , Corticosterone/metabolism , Fear/physiology , Glucocorticoids/metabolism , Hippocampus/metabolism , Hypothalamo-Hypophyseal System/metabolism , Male , Memory/physiology , Memory Disorders/metabolism , Mice , Mice, Inbred C57BL , Pituitary-Adrenal System/metabolism , Spatial Memory/physiology
3.
Endocrinology ; 156(12): 4592-603, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26305888

ABSTRACT

Chronic exposure to elevated levels of glucocorticoids has been linked to age-related cognitive decline and may play a role in Alzheimer's disease. In the brain, 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) amplifies intracellular glucocorticoid levels. We show that short-term treatment of aged, cognitively impaired C57BL/6 mice with the potent and selective 11ß-HSD1 inhibitor UE2316 improves memory, including after intracerebroventricular drug administration to the central nervous system alone. In the Tg2576 mouse model of Alzheimer's disease, UE2316 treatment of mice aged 14 months for 4 weeks also decreased the number of ß-amyloid (Aß) plaques in the cerebral cortex, associated with a selective increase in local insulin-degrading enzyme (involved in Aß breakdown and known to be glucocorticoid regulated). Chronic treatment of young Tg2576 mice with UE2316 for up to 13 months prevented cognitive decline but did not prevent Aß plaque formation. We conclude that reducing glucocorticoid regeneration in the brain improves cognition independently of reduced Aß plaque pathology and that 11ß-HSD1 inhibitors have potential as cognitive enhancers in age-associated memory impairment and Alzheimer's dementia.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Alzheimer Disease/genetics , Amyloid beta-Peptides/drug effects , Cerebral Cortex/drug effects , Cognition/drug effects , Memory/drug effects , Plaque, Amyloid/pathology , Pyrazoles/pharmacology , Thiophenes/pharmacology , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Behavior, Animal/drug effects , Cerebral Cortex/pathology , Disease Models, Animal , Humans , Insulysin/drug effects , Insulysin/metabolism , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic
4.
Eur J Neurosci ; 41(6): 787-92, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25614240

ABSTRACT

11ß-Hydroxysteroid dehydrogenase type 1 (11ß-HSD1) locally regenerates active glucocorticoids from their inert forms thereby amplifying intracellular levels within target tissues including the brain. We previously showed greater increases in intra-hippocampal corticosterone (CORT) levels upon Y-maze testing in aged wild-type than in 11ß-HSD1(-/-) mice coinciding with impaired and intact spatial memory, respectively. Here we examined whether ageing influences 11ß-HSD1 regulation of CORT in the dorsal hippocampus under basal conditions during the diurnal cycle and following stress. Intra-hippocampal CORT levels measured by in vivo microdialysis in freely behaving wild-type mice displayed a diurnal variation with peak levels in the evening that were significantly elevated with ageing. In contrast, the diurnal rise in intra-hippocampal CORT levels was greatly diminished in 11ß-HSD1(-/-) mice and there was no rise with ageing; basal intra-hippocampal CORT levels were similar to wild-type controls. Furthermore, a short (3 min) swim stress induced a longer lasting increase in intra-hippocampal CORT levels in wild-type mice than in 11ß-HSD1(-/-) mice despite no genotypic differences in elevation of plasma CORT. These data indicate that 11ß-HSD1 activity contributes substantially to diurnal and stress-induced increases in hippocampal CORT levels. This contribution is even greater with ageing. Thus, 11ß-HSD1 inhibition may be an attractive target for treating cognitive impairments associated with stress or ageing.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/physiology , Aging , Circadian Rhythm , Corticosterone/physiology , Hippocampus/physiology , Stress, Psychological , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Animals , Corticosterone/analysis , Hippocampus/chemistry , Male , Mice , Mice, Knockout , Microdialysis
5.
Neurobiol Aging ; 36(1): 334-43, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25109766

ABSTRACT

11Beta-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) locally amplifies active glucocorticoids within specific tissues including in brain. In the hippocampus, 11ß-HSD1 messenger RNA increases with aging. Here, we report significantly greater increases in intrahippocampal corticosterone (CORT) levels in aged wild-type (WT) mice during the acquisition and retrieval trials in a Y-maze than age-matched 11ß-HSD1(-/-) mice, corresponding to impaired and intact spatial memory, respectively. Acute stress applied to young WT mice led to increases in intrahippocampal CORT levels similar to the effects of aging and impaired retrieval of spatial memory. 11ß-HSD1(-/-) mice resisted the stress-induced memory impairment. Pharmacologic inhibition of 11ß-HSD1 abolished increases in intrahippocampal CORT levels during the Y-maze trials and prevented spatial memory impairments in aged WT mice. These data provide the first in vivo evidence that dynamic increases in hippocampal 11ß-HSD1 regenerated CORT levels during learning and retrieval play a key role in age- and stress-associated impairments of spatial memory.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/physiology , Aging/psychology , Glucocorticoids/metabolism , Hippocampus/metabolism , Spatial Memory/physiology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Aging/genetics , Aging/metabolism , Animals , Male , Maze Learning/physiology , Memory Disorders/drug therapy , Memory Disorders/genetics , Mice, Inbred C57BL , Molecular Targeted Therapy , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , RNA, Messenger/metabolism , Stress, Psychological/psychology , Thiophenes/pharmacology , Thiophenes/therapeutic use
6.
Neuropharmacology ; 91: 71-6, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25497454

ABSTRACT

High glucocorticoid levels induced by stress enhance the memory of fearful events and may contribute to the development of anxiety and posttraumatic stress disorder. In contrast, elevated glucocorticoids associated with ageing impair spatial memory. We have previously shown that pharmacological inhibition of the intracellular glucocorticoid-amplifying enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) improves spatial memory in aged mice. However, it is not known whether inhibition of 11ß-HSD1 will have any beneficial effects on contextual fear memories in aged mice. Here, we examined the effects of UE2316, a selective 11ß-HSD1 inhibitor which accesses the brain, on both spatial and contextual fear memories in aged mice using a vehicle-controlled crossover study design. Short-term UE2316 treatment improved spatial memory in aged mice, an effect which was reversed when UE2316 was substituted with vehicle. In contrast, contextual fear memory induced by foot-shock conditioning was significantly reduced by UE2316 in a non-reversible manner. When the order of treatment was reversed following extinction of the original fear memory, and a second foot-shock conditioning was given in a novel context, UE2316 treated aged mice (previously on vehicle) now showed increased fear memory compared to vehicle-treated aged mice (previously on UE2316). Renewal of the original extinguished fear memory triggered by exposure to a new environmental context may explain these effects. Thus 11ß-HSD1 inhibition reverses spatial memory impairments with ageing while reducing the strength and persistence of new contextual fear memories. Potentially this could help prevent anxiety-related disorders in vulnerable elderly individuals.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/physiology , Fear/physiology , Memory/physiology , Spatial Memory/physiology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Age Factors , Animals , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Corticosterone/blood , Cross-Over Studies , Fear/drug effects , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Pyrazoles/pharmacology , Spatial Memory/drug effects , Thiophenes/pharmacology
7.
Neuropharmacology ; 81: 231-6, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24495397

ABSTRACT

11ß-Hydroxysteroid dehydrogenase type 1 (11ß-HSD1) catalyzes intracellular regeneration of corticosterone and cortisol, thereby enhancing glucocorticoid action. Inhibition of 11ß-HSD1 reverses the deficits in cognition with aging, a state of elevated glucocorticoid levels. However, any impact of 11ß-HSD1 inhibition during high glucocorticoid states in younger animals is unknown. Here we examined whether a single injection of the selective 11ß-HSD1 inhibitor UE2316 modifies the effect of stress on hippocampal long-term potentiation and fear conditioning, a learning paradigm that is strongly modulated by glucocorticoids. We found that novelty stress suppresses hippocampal synaptic potentiation. This effect was completely prevented by administration of UE2316 one hour before stress exposure. A single injection of UE2316 also impaired contextual, but not tone-cue-fear conditioning. These observations suggest that local metabolism of glucocorticoids is relevant for the outcome of stress effects on hippocampal synaptic plasticity and contextual fear conditioning. Selective 11ß-HSD1 inhibitors may be an interesting new approach to the prevention of trauma-associated psychopathology.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Conditioning, Psychological/physiology , Learning Disabilities/etiology , Neuronal Plasticity/physiology , Pyrazoles/therapeutic use , Stress, Psychological/complications , Thiophenes/therapeutic use , Acoustic Stimulation/adverse effects , Analysis of Variance , Animals , Conditioning, Psychological/drug effects , Corticosterone/blood , Cues , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Fear/drug effects , Hippocampus/drug effects , In Vitro Techniques , Long-Term Potentiation/drug effects , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Pyrazoles/administration & dosage , Stress, Psychological/blood , Stress, Psychological/pathology , Stress, Psychological/prevention & control , Thiophenes/administration & dosage
8.
Front Aging Neurosci ; 4: 24, 2012.
Article in English | MEDLINE | ID: mdl-22952463

ABSTRACT

The hippocampus is a prime target for glucocorticoids (GCs) and a brain structure particularly vulnerable to aging. Prolonged exposure to excess GCs compromises hippocampal electrophysiology, structure, and function. Blood GC levels tend to increase with aging and correlate with impaired spatial memory in aging rodents and humans. The magnitude of GC action within tissues depends not only on levels of steroid hormone that enter the cells from the periphery and the density of intracellular receptors but also on the local metabolism of GCs by 11ß-hydroxysteroid dehydrogenases (11ß-HSD). The predominant isozyme in the adult brain, 11ß-HSD1, locally regenerates active GCs from inert 11-keto forms thus amplifying GC levels within specific target cells including in the hippocampus and cortex. Aging associates with elevated hippocampal and neocortical 11ß-HSD1 and impaired spatial learning while deficiency of 11ß-HSD1 in knockout (KO) mice prevents the emergence of cognitive decline with age. Furthermore, short-term pharmacological inhibition of 11ß-HSD1 in already aged mice reverses spatial memory impairments. Here, we review research findings that support a key role for GCs with special emphasis on their intracellular regulation by 11ß-HSD1 in the emergence of spatial memory deficits with aging, and discuss the use of 11ß-HSD1 inhibitors as a promising novel treatment in ameliorating/improving age-related memory impairments.

9.
J Neurosci ; 31(11): 4188-93, 2011 Mar 16.
Article in English | MEDLINE | ID: mdl-21411659

ABSTRACT

Local brain amplification of glucocorticoids (GCs) by 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) plays a pivotal role in age-related memory deficits. 11ß-HSD1 deficient mice are protected from spatial memory impairments with aging, but the underlying mechanisms are unknown. To determine which brain receptors [high-affinity mineralocorticoid receptors (MRs) or low-affinity glucocorticoid receptors (GRs)] are involved, spatial memory was measured in aged 11ß-HSD1(-/-) mice before and during intracerebroventricular infusion (10 d) of spironolactone (MR antagonist) or RU486 (GR antagonist). Aged C57BL/6J control mice showed impaired spatial memory in the Y-maze; this improved with GR blockade, while MR blockade had no effect. In contrast, aged 11ß-HSD1(-/-) mice showed intact spatial memory that became impaired with MR blockade, but not GR blockade. Hippocampal MR and GR mRNA expression and plasma corticosterone levels were not significantly altered with spironolactone or RU486 in either genotype. These data support the notion that 11ß-HSD1 deficiency in aging mice leads to lower intracellular GC concentrations in brain, particularly in the hippocampus, which activate predominantly MRs to enhance memory, while in aging C57BL/6J controls, the increased intracellular GCs saturate MRs and activate predominantly GRs, thus impairing memory, an effect reversed by GR blockade.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Aging/physiology , Hippocampus/metabolism , Memory/physiology , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Analysis of Variance , Animals , Cognition/drug effects , Cognition/physiology , Corticosterone/blood , Hippocampus/drug effects , Infusions, Intraventricular , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , Mice , Mice, Knockout , Mineralocorticoid Receptor Antagonists/pharmacology , Radioimmunoassay , Receptors, Glucocorticoid/genetics , Receptors, Mineralocorticoid/genetics , Spironolactone/pharmacology
10.
Behav Brain Res ; 216(1): 293-300, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20713092

ABSTRACT

Manganese (Mn(2+))-enhanced magnetic resonance (MR) imaging (MEMRI) in rodents offers unique opportunities for the longitudinal study of hippocampal structure and function in parallel with cognitive testing. However, Mn(2+) is a potent toxin and there is evidence that it can interfere with neuronal function. Thus, apart from causing adverse peripheral side effects, Mn(2+) may disrupt the function of brain areas where it accumulates to produce signal enhancement and, thereby, Mn(2+) administration may confound cognitive testing. Here, we examined in male adult Lister hooded rats if a moderate systemic dose of MnCl2 (200 µmol/kg; two intraperitoneal injections of 100 µmol/kg separated by 1 h) that produces hippocampal MR signal enhancement would disrupt hippocampal function. To this end, we used a delayed-matching-to-place (DMP) watermaze task, which requires rapid allocentric place learning and is highly sensitive to interference with hippocampal function. Tested on the DMP task 1 h and 24 h after MnCl2 injection, rats did not show any impairment in indices of memory performance (latencies, search preference) or any sensorimotor effects. However, MnCl2 injection caused acute peripheral effects (severe ataxia and erythema, i.e. redness of paws, ears, and nose) which subsided over 30 min. Additionally, rats injected with MnCl2 showed reduced weight 1 day after injection and failed to reach the normal weight-growth curve of control rats within the 16 days monitored. Our results indicate that 200 µmol/kg MnCl2 produces hippocampal MR signal enhancement without disrupting hippocampus-dependent behavior on a rapid place learning task, even though attention must be paid to peripheral side effects.


Subject(s)
Chlorides/administration & dosage , Hippocampus/physiology , Image Enhancement/methods , Magnetic Resonance Imaging/methods , Manganese Compounds/administration & dosage , Manganese , Maze Learning/physiology , Analysis of Variance , Animals , Ataxia/chemically induced , Brain Mapping , Chlorides/adverse effects , Contrast Media/administration & dosage , Erythema/chemically induced , Male , Manganese Compounds/adverse effects , Rats
11.
J Neurosci ; 30(41): 13867-72, 2010 Oct 13.
Article in English | MEDLINE | ID: mdl-20943927

ABSTRACT

11ß-Hydroxysteroid dehydrogenase type 1 (11ß-HSD1) regenerates active glucocorticoids (GCs) from intrinsically inert 11-keto substrates inside cells, including neurons, thus amplifying steroid action. Excess GC action exerts deleterious effects on the hippocampus and causes impaired spatial memory, a key feature of age-related cognitive dysfunction. Mice with complete deficiency of 11ß-HSD1 are protected from spatial memory impairments with aging. Here, we tested whether lifelong or short-term decreases in 11ß-HSD1 activity are sufficient to alter cognitive function in aged mice. Aged (24 months old) heterozygous male 11ß-HSD1 knock-out mice, with ∼60% reduction in hippocampal 11ß-reductase activity throughout life, were protected against spatial memory impairments in the Y-maze compared to age-matched congenic C57BL/6J controls. Pharmacological treatment of aged C57BL/6J mice with a selective 11ß-HSD1 inhibitor (UE1961) for 10 d improved spatial memory performance in the Y-maze (59% greater time in novel arm than vehicle control). These data support the use of selective 11ß-HSD1 inhibitors in the treatment of age-related cognitive impairments.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Aging/metabolism , Cognition/physiology , Hippocampus/metabolism , Maze Learning/physiology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Analysis of Variance , Animals , Corticosterone/blood , Male , Mice , Mice, Knockout , Radioimmunoassay , Time Factors
12.
J Neurosci ; 30(20): 6916-20, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20484633

ABSTRACT

Increased neuronal glucocorticoid exposure may underlie interindividual variation in cognitive function with aging in rodents and humans. 11beta-Hydroxysteroid dehydrogenase type 1 (11beta-HSD1) catalyzes the regeneration of active glucocorticoids within cells (in brain and other tissues), thus amplifying steroid action. We examined whether 11beta-HSD1 plays a role in the pathogenesis of cognitive deficits associated with aging in male C57BL/6J mice. We show that 11beta-HSD1 levels increase with age in CA3 hippocampus and parietal cortex, correlating with impaired cognitive performance in the water maze. In contrast, neither circulating corticosterone levels nor tissue corticosteroid receptor expression correlates with cognition. 11beta-HSD1 elevation appears causal, since aging (18 months) male transgenic mice with forebrain-specific 11beta-HSD1 overexpression ( approximately 50% in hippocampus) exhibit premature age-associated cognitive decline in the absence of altered circulating glucocorticoid levels or other behavioral (affective) deficits. Thus, excess 11beta-HSD1 in forebrain is a cause of as well as a therapeutic target in memory impairments with aging.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Aging , Gene Expression Regulation/physiology , Hippocampus/metabolism , Memory Disorders/genetics , Parietal Lobe/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Age Factors , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Male , Maze Learning/physiology , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/metabolism
13.
J Neurosci ; 27(39): 10487-96, 2007 Sep 26.
Article in English | MEDLINE | ID: mdl-17898220

ABSTRACT

Glucocorticoids are pivotal in the maintenance of memory and cognitive functions as well as other essential physiological processes including energy metabolism, stress responses, and cell proliferation. Normal aging in both rodents and humans is often characterized by elevated glucocorticoid levels that correlate with hippocampus-dependent memory impairments. 11Beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) amplifies local intracellular ("intracrine") glucocorticoid action; in the brain it is highly expressed in the hippocampus. We investigated whether the impact of 11beta-HSD1 deficiency in knock-out mice (congenic on C57BL/6J strain) on cognitive function with aging reflects direct CNS or indirect effects of altered peripheral insulin-glucose metabolism. Spatial learning and memory was enhanced in 12 month "middle-aged" and 24 month "aged" 11beta-HSD1(-/-) mice compared with age-matched congenic controls. These effects were not caused by alterations in other cognitive (working memory in a spontaneous alternation task) or affective domains (anxiety-related behaviors), to changes in plasma corticosterone or glucose levels, or to altered age-related pathologies in 11beta-HSD1(-/-) mice. Young 11beta-HSD1(-/-) mice showed significantly increased newborn cell proliferation in the dentate gyrus, but this was not maintained into aging. Long-term potentiation was significantly enhanced in subfield CA1 of hippocampal slices from aged 11beta-HSD1(-/-) mice. These data suggest that 11beta-HSD1 deficiency enhances synaptic potentiation in the aged hippocampus and this may underlie the better maintenance of learning and memory with aging, which occurs in the absence of increased neurogenesis.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/physiology , Hippocampus/physiology , Long-Term Potentiation/physiology , Maze Learning/physiology , Memory/physiology , Age Factors , Aging/physiology , Animals , Cognition/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout
14.
Neuropsychopharmacology ; 32(12): 2520-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17356567

ABSTRACT

The mechanisms by which antidepressants regulate the hypothalamic-pituitary-adrenal (HPA) axis are still unknown. The ABCB1-type multiple drug resistance (MDR) p-glycoprotein (PGP) regulates the HPA axis by limiting the access of glucocorticoids to the brain in mice and humans. Previous work in cell cultures has found that antidepressants enhance glucocorticoid receptor (GR) function in vitro by inhibiting MDR PGP, and therefore by increasing the intracellular concentration of glucocorticoids-but this model has never been tested directly in animals. Here, the tricyclic antidepressant, desipramine (20 mg/kg/day, i.p., for seven days), was administered to abcb1ab MDR PGP knockout mice (congenic on the FVB/N background strain) and to FVB/N controls. The hippocampal mRNA expression of GR, mineralocorticoid receptor (MR), MDR (Mdr1a) PGP, and 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) were measured, together with plasma corticosterone levels. In FVB/N controls, desipramine induced a significant upregulation of GR mRNA in the CA1 region (+31%; p=0.045); in contrast, in abcb1ab (-/-) mice, desipramine induced a significant downregulation of GR mRNA in the CA1 region (-45%; p=0.004). MR mRNA expression was unaltered. Desipramine decreased corticosterone levels in both FVB/N controls and in abcb1ab (-/-) mice, but in abcb1ab (-/-) mice the effects were smaller. Specifically, in FVB/N controls (but not in abcb1ab (-/-) mice), desipramine reduced corticosterone levels not only compared with saline-treated mice but also compared with the 'physiological' levels of untreated mice (-39%; p=0.05). Finally, desipramine reduced Mdr1a mRNA expression across all hippocampus areas (-9 to -23%), but had no effect on 11beta-HSD1 mRNA expression. These data support the notion that the MDR PGP is one of the molecular targets through which antidepressants regulate the HPA axis.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Antidepressive Agents, Tricyclic/pharmacology , Desipramine/pharmacology , Receptors, Glucocorticoid/metabolism , Up-Regulation/drug effects , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1 , ATP-Binding Cassette Transporters/genetics , Analysis of Variance , Animals , Corticosterone/blood , Hippocampus/drug effects , Hippocampus/metabolism , In Situ Hybridization/methods , Male , Mice , Mice, Knockout , RNA, Messenger/metabolism , Receptors, Glucocorticoid/genetics
15.
J Neurosci ; 26(43): 11034-40, 2006 Oct 25.
Article in English | MEDLINE | ID: mdl-17065445

ABSTRACT

Pregnenolone (PREG) and dehydroepiandrosterone (DHEA) have been reported to improve memory in aged rodents. In brain, these neurosteroids are transformed predominantly into 7alpha-hydroxylated metabolites by the cytochrome P450-7B1 (CYP7B). The biological role of steroid B-ring hydroxylation is unclear. It has been proposed to generate bioactive derivatives that enhance cognition, immune, and other physiological processes. In support, 7alpha-hydroxylated DHEA increases the immune response in mice with greater potency than the parent steroid. Whether the memory-enhancing effects of PREG in rats is mediated via its 7alpha-hydroxylated metabolite 7alpha-hydroxyPREG is not known. We investigated this by treating memory-impaired aged rats (identified by their spatial memory performances in the Morris water maze task compared with young controls) with 7alpha-hydroxyPREG or PREG administered intracerebroventricularly using osmotic minipumps and then tested the rats during week 2 of steroid treatment in the eight-arm radial-arm version of the water maze (RAWM) that allows repeated assessment of learning. CYP7B bioactivity in hippocampal tissue (percentage conversion of [14C]DHEA to [14C]7alpha-hydroxyDHEA) was decreased selectively in memory-impaired aged rats compared with both young and memory-intact aged rats. 7alpha-hydroxyPREG (100 ng/h) but not PREG (100 ng/h) administration to memory-impaired aged rats for 11 d enhanced spatial memory retention (after a 30 min delay between an exposure trial 1 and test trial 2) in the RAWM. These data provide evidence for a biologically active enzyme product 7alpha-hydroxyPREG and suggests that reduced CYP7B function in the hippocampus of memory-impaired aged rats may, in part, be overcome by administration of 7alpha-hydroxyPREG.


Subject(s)
17-alpha-Hydroxypregnenolone/analogs & derivatives , Aging/drug effects , Cytochrome P-450 Enzyme System/administration & dosage , Retention, Psychology/drug effects , Spatial Behavior/drug effects , Steroid Hydroxylases/administration & dosage , 17-alpha-Hydroxypregnenolone/administration & dosage , Aging/physiology , Animals , Cytochrome P450 Family 7 , Injections, Intraventricular , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , Memory/physiology , Rats , Retention, Psychology/physiology , Spatial Behavior/physiology
16.
Brain Res Mol Brain Res ; 129(1-2): 189-92, 2004 Oct 22.
Article in English | MEDLINE | ID: mdl-15469896

ABSTRACT

Adult rats were given antidepressant drugs orally. Fluoxetine, but not moclobemide, venlafaxine, tianeptine or desipramine, increased total glucocorticoid receptor (GR) mRNA in the hippocampus after 4 weeks. Further examination revealed that GR mRNA containing the brain-specific exon 1(7) was increased across all hippocampal subregions. In contrast, expression of the major exon 1(10) and another brain-specific exon 1(5)-containing GR mRNAs were unchanged. Tissue-specific first exon usage may contribute to the differential regulation of GR by fluoxetine in brain subregions.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Fluoxetine/pharmacology , Hippocampus , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Antidepressive Agents, Second-Generation/administration & dosage , Brain/anatomy & histology , Brain/metabolism , Exons , Fluoxetine/administration & dosage , Gene Expression Regulation , Hippocampus/drug effects , Hippocampus/metabolism , Protein Isoforms/genetics , Rats , Receptors, Glucocorticoid/genetics
17.
Proc Natl Acad Sci U S A ; 101(17): 6734-9, 2004 Apr 27.
Article in English | MEDLINE | ID: mdl-15071189

ABSTRACT

In aging humans and rodents, inter-individual differences in cognitive function have been ascribed to variations in long-term glucocorticoid exposure. 11beta-Hydroxysteroid dehydrogenase type 1 (11beta-HSD1) regenerates the active glucocorticoid cortisol from circulating inert cortisone, thus amplifying intracellular glucocorticoid levels in some tissues. We show that 11beta-HSD1, but not 11beta-HSD2, mRNA is expressed in the human hippocampus, frontal cortex, and cerebellum. In two randomized, double-blind, placebo-controlled crossover studies, administration of the 11beta-HSD inhibitor carbenoxolone (100 mg three times per day) improved verbal fluency (P < 0.01) after 4 weeks in 10 healthy elderly men (aged 55-75 y) and improved verbal memory (P < 0.01) after 6 weeks in 12 patients with type 2 diabetes (52-70 y). Although carbenoxolone has been reported to enhance hepatic insulin sensitivity in short-term studies, there were no changes in glycemic control or serum lipid profile, nor was plasma cortisol altered. 11beta-HSD1 inhibition may be a new approach to prevent/ameliorate cognitive decline.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Cognition , Diabetes Mellitus, Type 2/physiopathology , 11-beta-Hydroxysteroid Dehydrogenases/genetics , 11-beta-Hydroxysteroid Dehydrogenases/metabolism , Aged , Case-Control Studies , Cross-Over Studies , Diabetes Mellitus, Type 2/enzymology , Female , Humans , Male , Middle Aged , Placebos , RNA, Messenger/genetics
18.
Ann N Y Acad Sci ; 1007: 357-66, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14993069

ABSTRACT

Glucocorticoids affect a wide range of processes in the brain, altering neurotransmission, electrophysiological activity, metabolism, cell division, and death. These actions are mediated by corticosteroid receptors (glucocorticoid and mineralocorticoid) that modify transcriptional activity of target genes. The amount of steroid available to activate these receptors is not only dependent on the circulating levels but also on pre-receptor metabolism of glucocorticoids occurring intracellularly. This metabolism is carried out by the enzymes 11beta-hydroxysteroid dehydrogenases (11beta-HSDs). There are two distinct isozymes, the products of distantly related genes. 11beta-HSD type 2 inactivates glucocorticoids to its inert 11-keto derivative, while 11alpha-HSD type 1 elevates intracellular glucocorticoid levels by regenerating active glucocorticoids from circulating 11-dehydrocorticosterone or cortisone. This review highlights the important and very different roles the two enzymes play in the brain, outlining recent results obtained from studying mice with a targeted gene deletion in the 11beta-HSD1 or 11beta-HSD2 genes.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/physiology , 11-beta-Hydroxysteroid Dehydrogenase Type 2/physiology , Brain/enzymology , Animals , Humans
19.
Brain Res Mol Brain Res ; 106(1-2): 117-23, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12393271

ABSTRACT

As rats age, a subgroup will show spatial memory impairments, along with decreased corticosteroid receptors (MR and/or GR) in the hippocampus and a hyperactive hypothalamic-pituitary-adrenal axis. In previous work, we have shown that amitriptyline treatment increases hippocampal MR mRNA and improves spatial memory in young rats but had no effect in aged rats. Here, we examine the effect of 1-month treatment with the selective 5-HT re-uptake inhibitor, fluoxetine (10 mg/kg, p.o.) on hippocampal corticosteroid receptor mRNA and spatial memory in young 4-month-old and aged 24-month-old rats. Aged rats were impaired in spatial memory compared to young controls. MR mRNA expression was reduced with ageing in all hippocampal subfields except CA4 (35% decrease in dentate gyrus (DG) and CA2, P<0.05) and GR mRNA was decreased selectively in CA1 (17% decrease, P<0.05). Fluoxetine treatment increased GR mRNA in the hippocampus of young rats (24 and 46% increase in DG and CA3, respectively, P<0.01) but had no effect on hippocampal MR mRNA expression. In contrast, in aged rats, fluoxetine treatment increased hippocampal MR mRNA selectively in CA2 (43% increase, P<0.05), but had no effect on hippocampal GR mRNA. Fluoxetine treatment did not alter watermaze performance in either young or aged rats. It appears that increased hippocampal MR (at least in the CA2 region) which may underlie the enhancement in memory processing in young rats, is insufficient to improve memory in already cognitively impaired aged rats.


Subject(s)
Aging/physiology , Brain/drug effects , Fluoxetine/pharmacology , Memory/physiology , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Brain/metabolism , In Situ Hybridization , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Receptors, Glucocorticoid/genetics , Receptors, Mineralocorticoid/genetics
20.
J Neurosci ; 22(4): 1436-42, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11850470

ABSTRACT

Increasing evidence links chronically elevated glucocorticoid levels and cognitive impairments in a subpopulation of aged rodents and humans. Antidepressant drugs improve hypothalamic-pituitary-adrenal axis feedback regulation and reduce plasma glucocorticoid levels. Decreasing the cumulative lifetime exposure to glucocorticoid excess by long-term exposure to antidepressants may prevent the emergence of cognitive impairments in aged rats. To test this hypothesis, we treated middle-aged male Lister hooded rats (16 months) with amitriptyline until they were 24 months of age, and their cognitive function was assessed in the water maze. Performance in the spatial learning task declined significantly with aging (p < 0.01), with 33% of aged controls showing poorer (<2.5 SD) probe test performance than young controls. Amitriptyline treatment from midlife preserved water maze performance with aging (p < 0.01 compared with aged controls) and significantly (p < 0.01) reduced the proportion of poor performers (7%). Measures of anxiety-related behaviors in the elevated plus-maze were significantly (p < 0.05) decreased in the aged rats after amitriptyline. Furthermore, evening plasma corticosterone levels were reduced (30% decrease; p < 0.01 compared with aged controls) after 6 months of amitriptyline. These data suggest that long-term treatment with amitriptyline decreases the prevalence of cognitive impairment in aged rats and that this may, in part, be a consequence of reduced plasma corticosterone levels and reduced anxiety.


Subject(s)
Aging , Amitriptyline/administration & dosage , Antidepressive Agents, Tricyclic/administration & dosage , Maze Learning/drug effects , Memory Disorders/prevention & control , Administration, Oral , Adrenal Glands/drug effects , Aging/blood , Animals , Anxiety/drug therapy , Behavior, Animal/drug effects , Cognition/drug effects , Corticosterone/blood , Drug Administration Schedule , Male , Memory Disorders/blood , Organ Size/drug effects , Rats , Rats, Inbred Strains , Reaction Time/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...