当前位置: 首页 > 期刊 > 《内分泌学杂志》 > 2005年第12期 > 正文
编号:11411134
Low Doses of Dexamethasone Can Produce a Hypocorticosteroid State in t
http://www.100md.com 《内分泌学杂志》
     Division of Medical Pharmacology, Leiden/Amsterdam Center for Drug Research and Leiden University Medical Center, 2300 RA Leiden, The Netherlands

    Abstract

    The synthetic glucocorticoid dexamethasone (dex) blocks stress-induced hypothalamic-pituitary-adrenal (HPA) activation primarily at the level of the anterior pituitary because multidrug resistance P-glycoprotein hampers its penetration in the brain. Here, we tested the hypothesis that central components of the HPA axis would escape dex suppression under conditions of potent peripheral glucocorticoid action. We subchronically treated rats with low or high doses of dex. The animals were subjected on the last day of treatment for 30 min to a restraint stressor after which central and peripheral markers of HPA axis activity were measured. Basal and stress-induced corticosterone secretion, body weight gain, adrenal and thymus weight, as well as proopiomelanocortin mRNA in the anterior pituitary were reduced in a dose-dependent manner by dex administered either 5 d sc or 3 wk orally. In the brain, the highest dose dex suppressed CRH mRNA and CRH heteronuclear RNA in the paraventricular nucleus (PVN). However, in the peripherally active low-dose range of dex CRH mRNA and heteronuclear RNA showed resistance to suppression, and CRH mRNA expression in the PVN was in fact enhanced under the long-term treatment condition. In the PVN, c-fos mRNA was suppressed by the highest dose of dex, but this effect showed a degree of resistance after long-term oral treatment. c-fos mRNA responses in the anterior pituitary followed those in PVN and reflect central drive of the HPA axis even if corticosterone responses are strongly reduced. The results support the concept that low doses of dex can create a hypocorticoid state in the brain.

    Introduction

    THE SYNTHETIC GLUCOCORTICOID dexamethasone (dex) is a potent and rather selective glucocorticoid receptor (GR) ligand in vivo (1). It has profound effects on energy metabolism, the immune system, and hypothalamic-pituitary-adrenal (HPA) axis activity. In the clinic, it is commonly used, often in combination with a CRH challenge, for diagnostic and research purposes to test HPA axis function in, e.g. Cushing’s disease and affective disorders. Several studies have demonstrated a pituitary rather than a brain site of action in the suppression of HPA axis activity if moderate amounts of dex are administered (2, 3, 4, 5, 6). In support of these findings, it was demonstrated that the efflux transporter P-glycoprotein, expressed at the luminal side of the blood-brain barrier (7), hampers the penetration of dex into the brain (8, 9).

    In the present study, the hypothesis was tested that dex exclusion from the rat brain by P-glycoprotein provides a subtle way to create a brain-selective alternative for adrenalectomy, dissociated from the abundant exposure of peripheral corticosteroid targets to glucocorticoids, with adrenal medullary functions and aldosterone secretion intact. This state would arise because treatment with small amounts of dex results in suppression of endogenous corticosterone secretion, and, at the same time, dex would poorly substitute for corticosterone in the brain. As a consequence, a reduced feedback of glucocorticoids to its central targets should ensue and create in the brain a tissue-specific hypocorticoid condition.

    All doses of dex are expected to inhibit adrenal output of corticosterone and, thus, lead to decreased occupancy of the high-affinity mineralocorticoid receptor (MR) in the brain. Although high doses of dex lead to the aphysiological situation in which GR gets occupied, but MR becomes depleted from endogenous corticosterone (10), the low poorly penetrating dose would lead to lower activation of both MR and GR. This could yield a model to study the effects of a hypocorticoid brain state, dissociated from peripheral glucocorticoid exposure.

    To test this hypothesis, male rats were treated with different doses of dex for either 5 d sc or 3 wk orally. The rats treated with dex were compared with two different control groups. Adrenalectomized (ADX) rats served as control for the central (and peripheral) hypocorticoid state, and a high-dose dex group represented chronic GR activation as present in hypercorticism. To reveal glucocorticoid feedback, the animals were exposed the last day to a restraint stressor. As markers for peripheral glucocorticoid effects, body, adrenal and thymus weight, and plasma corticosterone levels were measured as well as the expression of anterior pituitary proopiomelanocortin (POMC) mRNA. As central markers, the CRH mRNA and heteronuclear RNA (hnRNA) were measured in the parvocellular part of the hypothalamic paraventricular nucleus (PVN). cFos mRNA was used as marker for the excitatory input to the PVN and the anterior pituitary (11).

    Materials and Methods

    Animals

    Young adult male Wistar rats (Charles River, Sulzfeld, Germany), weighing around 185–265 g at the time of arrival, were used. Animals were group-housed except for the drinking water experiment during which they were solitary housed, under a 12-h light, 12-h dark cycle with lights off at 20 h in a temperature (21 C) and humidity-controlled room. They had free access to food and drinking water. The experiments took place 2 wk after arrival at the lab. During this period, rats were handled daily. All experiments were carried out in accordance with the European Communities Council Directive 86/609/EEC and with approval from the animal care committee of the Faculty of Medicine, Leiden University (Leiden, The Netherlands).

    Experiment 1: 5-d treatment with dex sc

    At the start of experiment, the rats weighed 303 ± 15 g (mean ± SD). They were divided into four groups each consisting of six to 10 animals. One group (ADX) was ADX under gas anesthesia (isoflurane) by dorsal approach at the start of the experiment. After ADX, animals had free access to 0.9% saline and normal drinking water. All other animals were sham-operated. For 5 d, one group (DEXlow) was sc injected with a low dose of 50 μg/kg dex-21-phosphate (Sigma-Aldrich, St. Louis, MO; dissolved in 0.9% saline) twice daily at 0900 and 2100 h. A second group (DEXhigh) was treated with a high dose of dex (500 μg/kg twice daily). Sham-operated (VEH) and ADX groups were treated with vehicle. During the whole experimental period, body weight was monitored. At d 4, blood was sampled 1 h after lights off to determine corticosterone plasma levels at the circadian peak. At d 6, the animals received one last injection in the morning. Six hours later at 1500 h, half of each group (three to five rats per group) was exposed to restraint stress in a wire mesh/plastic cylinder for 30 min. At the start and after 15 min, blood samples were taken using the tail incision method (12). After 30 min, animals were removed from the restrainers and immediately decapitated. Nonstressed animals (the second half of each group) were decapitated directly after removal from their home cage. Trunk blood was collected in EDTA-coated tubes and centrifuged. Plasma was kept at –20 C until determination of corticosterone plasma levels. Brains were rapidly removed from the skull and quickly frozen in isopentane precooled on dry ice/ethanol. Thymus, pituitary, and adrenal glands were dissected and frozen on dry ice. All tissues were stored at –80 C until further use. Thymus and adrenals were cleaned and weighed.

    Experiment 2: 3-wk treatment with dex in drinking water

    At the start of the drinking water experiment, rats weighed 225 ± 9 g (mean ± SD). They were divided into six treatment groups consisting of five to eight animals. Three groups were treated with different concentrations (0.5, 1.0, and 10 μg/ml) of dex 21-phosphate in their drinking water for 3 wk (DEX0.5, DEX1.0, and DEX10 groups). One group was ADX at the start of the experiment. After adrenalectomy, animals had free access to 0.9% saline and normal drinking water. All other animals were SHAM operated including control animals (VEH) that received normal drinking water during the whole experiment. Each day, animals and bottles were weighed to determine the body weight gain and volume of drinking solution that each animal had drunk over 24 h. One animal of the highest concentration group died before the end of the experiment.

    After 3 wk, rats were stressed by restraint in a wire mesh/plastic cylinder and decapitated after 30 min as described for experiment 1. A control nonstress, nontreated group (VEH nonstress) was decapitated immediately after removal from the home cage.

    In situ hybridization

    Coronal sections of 14 μm through the PVN of the hypothalamus and hippocampus were cut in a cryostat. Pituitaries were sectioned at 12 μm. Sections were thaw-mounted on poly-L-lysine-coated microscopic slides. These slides were stored at –80 C until hybridization. The sections were postfixed in a freshly prepared 4% paraformaldehyde solution (pH 7.2) for 60 min at room temperature and rinsed twice in PBS for 5 min at room temperature. In case of in situ mRNA hybridizations, sections were permeabilized with proteinase K [1 μg/ml in 0.1 M Tris (pH 8.0)] at 37 C for 10 min. After a brief rinse in diethyl pyrocarbonate-treated water, they were treated with 0.25% acetic anhydride in 0.1 M triethanolamine (pH 8.0) for 10 min at room temperature and finally rinsed in 2x standard sodium citrate (SSC) (SSC = 0.15 M NaCl and 0.015 M sodium citrate) for 10 min at room temperature. Subsequently, the sections were dehydrated through a graded series of ethanol and air dried.

    Preparation of probes

    To visualize mRNAs, in situ mRNA and oligonucleotide hybridizations were performed. Different 33P- and 35S-labeled cRNA antisense probes were employed to hybridize with complementary brain tissue c-fos and CRH mRNA and CRH hnRNA. The c-fos mRNA probe was transcribed from a pBluescript (pBS) KS plasmid containing a 2.1-kb full-length rat c-fos cDNA sequence (courtesy of Dr. T. Curran, St. Jude Children’s Research Hospital, Lauderdale, Memphis, TN) in the presence of 33P-UTP (ICN Biomedicals, Costa Mesa, CA; Isoblue stabilized, specific activity. 3000 Ci/mmol). This probe was hydrolyzed by incubation in 90 mM 0.2 M Na2CO3 and 60 mM 0.2 M NaHCO3 at 60 C for 10 min to facilitate the tissue penetration. The CRH hnRNA 33P-UTP-labeled probe was transcribed from a 687-bp fragment (courtesy of P. Sawchenko, The Salk Institute, San Diego, CA) covering the single intron of the rat CRH gene subcloned into a pBS vector. A full-length probe for CRH mRNA (1.2 kb subcloned into pBS; courtesy of Dr. K. Mayo, Northwestern University, Evanston, IL) was synthesized in presence of 35S-UTP. Incorporation of labeled UTP was at least 75%.

    A 42-nucleotide mouse POMC oligonucleotide (GGT-TTT-CAG-TCA-GGG-GCT-GTT-CAT-CTC-CGT-TGC-CAG-GAA-ACA; 90% homology with rat POMC; Eurogentec, Seraing, Belgium) was end-labeled with 33P-dATP (NEN Life Science Products, Hoofddorp, The Netherlands; 2000 Ci/mmol, 10 mCi/ml) using terminal transferase with the manufacturer’s protocol (Roche Molecular Biochemicals, Almere, The Netherlands). A 0.33-pmol oligonucleotide was labeled at molar ratio of 1:20 (oligo:label). Incorporation was typically between 50 and 75%, resulting in a tail of 5- to 7.5-A residues per oligonucleotide.

    Hybridization procedures

    For riboprobes, each slide, containing four sections, was loaded with a 100-μl mix containing 70% deionized formamide, 10% dextran sulfate, 3x SSC, 50 mM dithiothreitol, 1x Denhardt’s solution, 0.1 mg/ml yeast tRNA, 0.1 mg/ml sheared herring sperm DNA, and 1–3 x 106 dpm of the probe, and covered with microscopic coverslips. Overnight hybridization was performed in a moist chamber at 55 C. As a control, a few slides were hybridized with sense probe. The next day, coverslips were removed, and the slides were washed in 2x SSC at room temperature for 10 min, treated with Rnase A [2 mg/100 ml in 0.5 M NaCl (pH 7.5)] at 37 C for 10 min, and washed three times in 2x SSC/50% formamide at 60 C for 15 min. After a short wash with 2x SSC, sections were dehydrated in an ethanol series and air dried. Finally, the slides were put in an x-ray exposure holder and apposed to Biomax MR film (Kodak, Rochester, NY) for 3–12 d (13).

    In situ hybridization using oligonucleotides was performed essentially as described (14). Labeled oligonucleotide (0.5–0.8 x 106 dpm) per 100 μl hybridization mix was applied to each slide. Hybridization mix consisted of 50% formamide, 10% dextran sulfate, 4x SSC, 25 mM sodium phosphate (pH 7.0), 1 mM sodium pyrophosphate, 20 mM DTT, 5x Denhardt’s, 100 μg/ml poly-A, and 100 μg/ml sheared herring sperm DNA. Sections were coverslipped and hybridized overnight in a moist chamber at 42 C. The next morning, coverslips were removed, rinsed in 1x SSC at room temperature, washed twice for 30 min in 1x SSC at 50 C, washed for 5 min in 1x SSC at room temperature, dehydrated in an ethanol series, and air dried. Then, sections were apposed to Kodak Biomax MR film that were developed after 17–20 h.

    Densitometric quantification

    Optical density was quantified with analysis performed on a Macintosh computer using the public domain NIH Image program (http://rsb.info.nih.gov/nih-image). Nissl staining and light microscopy were used to confirm the presence of the PVN in the section. Optical densities were determined by outlining the parvocellular part of the PVN. The OD of the area dorsolateral from the PVN was used to correct for tissue background. Measurements of three to five sections were averaged per animal with the mean value from each animal used in subsequent statistical analysis.

    Corticosterone plasma levels

    The plasma corticosterone concentration was determined using a standard in-house RIA procedure (15). Antiserum raised in sheep against corticosterone-21-hemisuccinate BSA was a gift from Dr. F. Sweep (University of Nijmegen, The Netherlands). The detection limit was 0.2 μg/dl. Animals were considered ADX when basal trough plasma corticosterone levels were lower than 1.0 μg/dl.

    Determination of apoptosis

    The presence of apoptotic granule cells in the dentate gyrus was evaluated by qualitatively scoring cell nuclei with fragmented DNA (i.e. pyknotic cells) in Nissl-stained sections of the hippocampus at a magnification of 400x (Fig. 1D).

    Statistics

    Completeness of ADX was determined based on corticosterone plasma levels and on relative saline intake (Richter, 1941); one animal was excluded from the analysis because of incomplete ADX. Plasma corticosterone data were analyzed using one-way ANOVA or repeated measurements ANOVA, where appropriate. All other data were analyzed using two-way ANOVA with treatment and stress as factors (experiment 1) or one-way ANOVA with treatment as factor and stress as covariate (experiment 2; only one nonstress control group was included in this case). When a significant stress effect or treatment x stress interaction was absent, data of stress and nonstress groups were combined for further analysis on treatment effects. In case stress or interaction effects were significant, stress and nonstress groups were separately analyzed applying one-way ANOVA. When appropriate, post hoc comparisons were performed using Tukey’s honestly significant difference (HSD) test. Statistical difference was accepted at P < 0.05. All data are expressed as mean ± SEM.

    Results

    In both experiments, we evaluated basal as well as restraint stress-induced parameters of the HPA axis. Because POMC mRNA in the pituitary and CRH mRNA in the PVN did not react to the 30-min stressor, we have included these in the basal, nonstress-reactive parameters.

    Experiment 1: 5-d treatment with dex sc

    Basal activity: peripheral.

    Two daily sc injections of dex for 5 d suppressed basal plasma corticosterone levels both in the morning immediately before the stress response and at the circadian peak (P < 0.05, Table 1). Further, adrenal and thymus weights were significantly reduced in both DEXlow and DEXhigh groups (P < 0.05; Table 1, Fig. 1A). Body weight decreased from the start of the treatment in contrast to vehicle-treated animals that gained weight at a normal rate resulting in a significant difference between both groups at the end of the treatment (Table 1). Two-way ANOVA did not reveal an effect of 30-min restraint stress on POMC mRNA expression in the anterior pituitary [F(1,21) = 0.319; P > 0.05], which is consistent with previous studies (16), but there was a significant treatment effect; dex treatment significantly reduced POMC mRNA expression in the anterior pituitary in a dose-dependent manner (P < 0.05, Fig. 1B). ADX animals had undetectable corticosterone plasma levels, increased POMC mRNA expression, and increased thymus weight (P < 0.05, Table 1 and Fig. 1, A and B). Pituitary intermediate lobe POMC mRNA expression was not affected by treatment nor by stress (data not shown).

    Basal activity: central.

    CRH mRNA expression was not affected by 30-min restraint stress. Basal CRH mRNA levels were strongly reduced in the DEXhigh group but in DEXlow group were not different from vehicle-treated animals (Fig. 1C). No significant up-regulation was observed after ADX. In 70% of the ADX animals, pyknotic cells, which are indicative of apoptosis, were abundantly present in the granule cell layer of the dentate gyrus, mainly located in the tip of the inner blade (Fig. 1D). Apoptosis of dentate gyrus cells was not found in the brains of any of the DEXlow animals and in DEXhigh animals (0%).

    Restraint stress-induced activity: peripheral.

    Repeated measurements ANOVA on the stress-induced corticosterone response revealed a significant interaction effect of stress and treatment [F(6,24) = 46.7; P < 0.05; Fig. 2A]. The levels at 30 min after the initiation of stress were considerably lower in the DEXlow group (P < 0.05) compared with those in VEH group (Fig. 2A), although the stress-induced rise of corticosterone was not completely blocked to the extent observed in the DEXhigh group. In agreement with a previous report (17), restraint stress-induced a response of c-fos mRNA expression in the anterior pituitary [F(1,21) = 17.5; P < 0.05], which was significant in low-dose dex and control rats but reduced in the DEXhigh group (Fig. 2B).

    Restraint stress-induced activity: central.

    In the brain, the stress-induced responses of c-fos mRNA and CRH hnRNA expression in the parvocellular part of the PVN were strongly reduced in the DEXhigh group (Fig. 2, C and D). However, in the DEXlow animals, the rapid increases of c-fos mRNA and CRH hnRNA after stress were not different from the VEH animals or the 5-d ADX animals. With respect to the control measurements for stress induction, dex treatment had no effect on the basal level of c-fos mRNA in the PVN or on basal CRH hnRNA levels, although the latter showed a tendency toward differences, possibly reflecting lower expression in the DEXhigh group [F(3,13) = 3.236, P = 0.06; Fig. 2D].

    Experiment 2: 3-wk treatment with dex in drinking water

    As described above, the 5-d treatment with a low dose of dex led to a clear dissociation between peripheral and central effects of corticosterone. However, we hoped to create by dex administration a model in which we would induce an ADX-like central state in terms of glucocorticoid exposure, rather than a low-corticosterone one. Because we could not distinguish between the two states and because the 5-d low-dose dex treatment was insufficient to completely block stress-induced increase in corticosterone levels, we treated rats for 3 wk with low and high doses of dex via the drinking water and subjected the animals to restraint stress after this period.

    Basal activity: peripheral effects.

    dex treatment did not affect the volume of drinking solution drunk by the animals (Table 2). The amounts of dex ingested by the animals were calculated based on the volume drunk and the concentration of dex. Animals treated with 0.5, 1.0, or 10 μg dex/ml in their drinking water ingested per day 42 ± 1, 91 ± 4, and 1447 ± 145 μg dex/kg body weight, respectively. Control animals gained 27–33% body weight during the experimental period. DEX0.5 and DEX1.0 lost weight during the first 5 d and stabilized later on, such that they had not gained substantial weight at the end of the experimental period. In the DEX10 animals, the treatment resulted in a severe loss of body weight (Table 2).

    ADX and treatment with dex in the drinking water had the expected effects on peripheral glucocorticoid markers. In all dex treatment groups, basal trough levels of plasma corticosterone were below detection level of the assay. Also, circadian peak corticosterone levels in the DEX10 and DEX1.0 groups were completely suppressed, whereas hormones levels in the DEX0.5 group were significantly lowered compared with control rats, but not suppressed completely. Adrenal and thymus weights were dose dependently decreased by dex treatment, whereas thymus weight was increased in ADX animals (P < 0.05, Table 2 and Fig. 3A).

    Anterior pituitary POMC mRNA levels were significantly and strongly reduced in the DEX10 group and strongly increased in ADX animals. Although the average anterior pituitary POMC mRNA expression was reduced by 35% in the Dex1.0 groups, this difference did not reach statistical significance (P = 0.06; Fig. 3B).

    Basal parameters: central.

    Similar to the injection study, central markers were differentially affected by treatment with small amounts of dex compared with treatment with large amounts of dex. Both DEX0.5 and ADX animals showed increased levels of CRH mRNA in the PVN compared with the control group (P < 0.05), whereas the DEX10 animals showed a clear reduction in CRH mRNA levels (Fig. 3C). The CRH mRNA expression in the DEX1.0 animals was not changed compared with untreated control animals. No signs of apoptosis within the granule cell layer of the dentate gyrus could be found in any of the non-ADX treatment groups.

    Restraint stress-induced: peripheral.

    The stress-induced increase of corticosterone was completely blocked in the DEX10 and the DEX1.0 groups. In the DEX0.5 group, dex treatment reduced corticosterone plasma levels at 30 min after onset of stress to 35% of control levels (Fig. 4A).

    The c-fos mRNA expression in the anterior pituitary clearly responded to stress in all groups (Fig. 4B). Treatment also significantly affected expression levels between different groups of stressed animals [F(4,27) = 3.676, P < 0.05]. Low doses of dex treatment did not suppress the stress-induced c-fos expression in anterior pituitary but if anything seemed to induce an increased c-fos mRNA (although this effect was only found to be statistically significant when compared with the ADX group).

    Restraint stress-induced: central.

    c-fos mRNA showed a strong response to stress in all groups. This response showed a significant attenuation in the DEX10 animals (Fig. 4C), whereas it was not different from control animals in the two other dex-treated groups and in ADX animals. Stress induced a modest increase in CRH hnRNA in the PVN of untreated control animals, which in fact did not reach statistical significance by Tukey HSD post hoc test (P = 0.12; Fig. 4D). Only the DEX0.5 group showed a highly significant increase in CRH hnRNA levels after stress. There was a very clear dissociation between the lowest and highest dose of dex because CRH hnRNA levels clearly did not react to stress in the DEX 10 group.

    Discussion

    The present study demonstrates that hypothalamic CRH mRNA as well as restraint stress-induced CRH hnRNA and c-fos mRNA expression are resistant to dex under conditions in which peripheral glucocorticoid targets like anterior pituitary POMC expression, adrenal corticosteroid secretion, and thymus weight are suppressed. Although we were not able achieve a true brain-selective adrenalectomy, this finding supports the concept that, using the right conditions, the partial exclusion of dex from the brain combined with the suppression of peripheral pituitary-adrenal activity can create the state of a central hypocorticoid condition.

    Although in both experiments the effects of low dex clearly dissociated between brain and periphery, the case for an actual hypocorticoid central state, based on centrally measured parameters, can only be made for the long-term oral treatment, when CRH mRNA was increased relative to the stressed control group to a similar extent in the DEX0.5 and ADX groups. Interpretation of the first 5-d experiment is hampered by the fact that ADX group did not function as the positive control we expected. Apart from the unexpected lack of significantly increased thymus weight, we also did not observe any increase in brain markers, such as CRH mRNA. These atypical results remain unexplained. They do not reflect incomplete ADX, because other markers did react: plasma hormone levels, amount of ingested saline, POMC mRNA levels, and apoptosis in the dentate gyrus of the hippocampus. Importantly, the other tested paradigm of 3-wk oral low-dex administration most likely does lead to a hypocorticoid state, as is evident from the increased CRH mRNA levels and the CRH hnRNA response to restraint stress.

    The underpinning of the concept of exclusion of dex from brain originates from studies demonstrating that in vivo 3H-corticosterone administered in the circulation can easily enter the brain (4, 18, 19), whereas the entrance of 3H-dex is hampered. The brain does express high amounts of GR (18, 20), and in vitro brain GRs retain both 3H-dex and 3H-corticosterone (4, 21). Accordingly, the existence of a blood-brain barrier limiting the in vivo uptake of dex was postulated (2, 4, 22). Conclusive evidence for this barrier was obtained more recently, when it was demonstrated that the efflux transporter P-glycoprotein at the blood-brain barrier hampered the penetration of dex, prednisone, and cortisol, which are exogenous steroids for mice and rats (8, 9, 19, 23, 24). If the P-glycoprotein gene was disrupted as in the mdr1a knockout mice, tracer amounts of 3H-dex and 3H-cortisol were taken up and retained in the brain of these mutants, accumulating in hippocampal neurons as 3H-corticosterone did (9, 19). However, in intact animals, as a result of its poor central access to the brain, moderate doses of dex primarily act on the anterior pituitary to suppress pituitary-adrenal activity (3). Our data corroborate therefore a pituitary site of dex action on stress-induced ACTH release. However, our data also suggest that for the presently used stressor in rats a complete chronic suppression of the HPA axis requires doses of dex that can also act at (and may involve) central binding sites.

    Our findings are consistent with reports studying receptor occupancy in pituitary and brain tissue after either short-term or acute dex treatment (5, 6, 10, 11, 25). The latter studies have demonstrated that low doses of dex substantially occupy GR in the pituitary and other peripheral target tissues, whereas GR activity in the brain was relatively little affected. With regards to the MR, data have demonstrated that it will become to some extent depleted after a single dose of dex that suppresses endogenous corticosterone secretion (10). Using chronic high doses of dex, the responses to restraint stress were clearly reduced, and CRH mRNA expression was diminished after administration of large amounts of dex. This indicates that the barrier formed by P-glycoprotein is of course relative (8) and can be overcome by high amounts of dex to activate the GR (10, 25, 26). Indeed, studies reporting effects of dex on glucocorticoid targets in the brain used very high systemic doses or local brain implants of dex (27, 28, 29, 30, 31).

    Qualitative analysis revealed that pyknotic cells in the dentate gyrus were clearly visible in the ADX animals, reflecting apoptosis as a consequence of lack of MR activation (32, 33). However, no dose of dex treatment caused apoptosis, even though high doses of dex failed to protect against ADX-induced apoptosis (34), and in fact have been reported to induce apoptosis in the hippocampus in rats (6 months but not 1 month old) (35). In the low-dex groups, the PM levels of corticosterone were still elevated significantly above the detection limit, which likely protects against apoptosis via MR occupation. The lack of apoptosis even in the high-dex groups may indicate that corticosterone levels were not suppressed to the level that no MR activation was present, or that aldosterone levels were high enough to activate MR and protect against apoptosis.

    Although not all changes as observed after ADX were found in the brains of dex-treated rats, the amount of circulating corticosterone in blood and brain was strongly reduced after dex, also after low-dose treatment. Accordingly, the animals will be characterized by a profound change in the activation of the MR and GR. Such changes in MR/GR balance are considered a risk for coping with stress and are thought to enhance vulnerability to stress-related disease (36, 37). The low-dose dex-treated animal in particular may therefore present an interesting animal model because it ensures a relatively stable hypocorticoid climate in the brain that is not disturbed by (acute) stress-induced rises in corticosterone. Such a model may be useful in further defining the input of metabolic factors and cognitive processes in glucocorticoid-dependent brain mechanisms (38, 39, 40, 41).

    c-fos mRNA and CRH hnRNA on the PVN were resistant to dex suppression at low doses. In these groups as well as in the ADX groups, the responses were not augmented compared with those of the control groups in the present study. The latter is at variance with the previously reported amplified central stress response 5 d to 1 wk after ADX (29, 42) or after acute glucocorticoid withdrawal (43). However, in agreement with our finding are several other studies reporting no effect of 5-d ADX on induction of c-fos mRNA in hypothalamus after stress (44, 45, 46). As was pointed out in a recent report (11), an acute injection of the glucocorticoid RU28362 was capable to suppress stress-induced CRH expression rather than c-fos mRNA. Thus, acute suppression by RU28362 may target CRH and does not affect the excitatory input activating c-fos. It may be relevant that although CRH may be a direct target gene for glucocorticoids, at least in vitro (47), this has to our knowledge not been shown for c-fos. Our present data show that PVN c-fos expression does not escape blockade with high doses of dex, suggesting that under those conditions, these excitatory inputs are also suppressed.

    Interestingly, c-fos mRNA induction in the anterior pituitary in the 5-d experiment reacted similar to central markers and was only suppressed by the high dose of dex. Possibly (parallel to the PVN discussed above) the pituitary c-fos response is governed primarily by hypothalamic secretagogs, rather than subject to direct negative feedback. Support for the dissociation of pituitary POMC and c-fos regulation comes from a study that used AtT-20 cells to show that induction of c-fos was less strongly suppressed by dex than POMC mRNA (48). After 3 wk of administration through the drinking water, no suppression of this c-fos response was observed, not even in the DEX10 group, whereas at lower doses, the c-fos response was even enhanced. This is striking because the measures for PVN activity, c-fos, and CRH hnRNA were suppressed by the highest dose of dex. Possibly, the chronically disturbed MR/GR balance led to adaptations in parameters that were not evaluated here, such as vasopressin or other secretagogs. Alternatively, the assumption that the anterior pituitary c-fos response to restraint stress reflects solely corticotropic cells can be questioned. Anterior pituitary POMC mRNA expression is reduced after treatment with dex for 5 d, but after treatment for 3 wk, the effects of low doses of dex were surprisingly less dramatic. This to us is also suggestive of adaptive responses to the imbalanced activation of the two corticosteroid receptor types, with consequences for pituitary function.

    The impaired penetration of dex into the brain has important implications for the interpretation of the dex suppression test. This test is used in the clinic, often in combination with a CRH challenge, to evaluate the dysregulation of the HPA axis in, e.g. depressive patients (49, 50). A characteristic feature is the hyperactive HPA axis, which is not suppressed by a low dose of dex administered to these patients the night before. This escape of dex suppression observed in hypercortisolemic depressive patients is exaggerated by exogenous CRH. The present study offers an explanation for this phenomenon. The low-dex concentration will create a hypocorticoid condition in the brain and therefore trigger a response from the CRH-producing neurons of the PVN, which may be reflected by the enhanced response in the anterior pituitary. dex suppression in healthy subjects can indeed be overcome by concurrent infusion of CRH and vasopressin (51).

    In contrast to dex, the naturally occurring glucocorticoid corticosterone can easily enter the brain. In the mdr1a knockout and wild-type mice, 3H-corticosterone was taken up in brain and retained by brain MR to a similar extent (9, 19). Mdr1b-coded P-glycoprotein has been suggested to hamper corticosterone entrance to rodent brain, based on studies using mdr1a/1b knockout animal (24). It should be pointed out however that the mdr1b gene is expressed at the blood-brain barrier at much lower levels than mdr1a (52, 53) but is highly expressed in the adrenal. We would argue that the small effect in the mutants reported previously by others (24) probably is caused by factors beyond the brain affecting the distribution and metabolism of corticosterone. Interestingly, evidence based on measurement of steroid concentrations in postmortem brain points to preferential brain uptake of corticosterone also in humans (19). Thus, the much better penetration of corticosterone than dex in brain makes this steroid a much better candidate to MR and GR function (37, 54, 55, 56, 57), perhaps also in humans.

    In conclusion, our findings suggest that treatment with small amounts of dex can produce a hypocorticoid state selectively in the brain concomitant with feature of modestly increased glucocorticoid action in the periphery. This condition will specifically affect through imbalance of MR and GR central glucocorticoid target areas modulating HPA axis, behavioral adaptation, and synaptic plasticity, particularly after stress. Thus, divergence of peripheral and central glucocorticoid effects due to hampered access of small amounts of dex to the brain may serve as a model for states with disturbed glucocorticoid signaling in the brain.

    Acknowledgments

    The skillful assistance of Peter Steenbergen and Marleen Visser is highly appreciated. We also are grateful to Sergiu Dalm, Heidi Lesscher, Maaike van der Mark, and Servane Lachize for technical assistance. We thank Liesbeth de Lange for critical reading of the manuscript.

    Footnotes

    This work was supported by The Netherlands Organisation for Scientific Research (NWO Vidi Grant to O.C.M.), by the European Union ERASMUS (European Community Action Scheme for the Mobility of University Students) exchange program (to R.S.P.), and by the Royal Netherlands Academy of Arts and Sciences (to E.R.d.K.).

    Present address for A.M.K.: Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California 94304-5485.

    First Published Online September 8, 2005

    Abbreviations: ADX, Adrenalectomized; dex, dexamethasone; GR, glucocorticoid receptor; hnRNA, heteronuclear RNA; HPA, hypothalamic-pituitary-adrenal; HSD, honestly significant difference; MR, mineralocorticoid receptor; pBS, pBluescript; POMC, proopiomelanocortin; PVN, paraventricular nucleus; SSC, standard sodium citrate.

    Accepted for publication August 31, 2005.

    References

    Reul JM, Gesing A, Droste S, Stec IS, Weber A, Bachmann C, Bilang-Bleuel A, Holsboer F, Linthorst AC 2000 The brain mineralocorticoid receptor: greedy for ligand, mysterious in function. Eur J Pharmacol 405:235–249

    Rees HD, Stumpf WE, Sar M 1975 Autoradiographic studies with 3H dexamethasone in the rat brain and pituitary. In: Stumpf WE, Grant L, eds. Anatomical neuroendocrinology. Basel: S. Karger; 262–269

    De Kloet ER, van der Vies J, de Wied D 1974 The site of suppressive action of dexamethasone on pituitary-adrenal activity. Endocrinology 94:61–73

    De Kloet ER, Wallach G, McEwen BS 1975 Differences in corticosterone and dexamethasone binding to rat brain and pituitary. Endocrinology 96:598–609

    Miller AH, Spencer RL, Pulera M, Kang S, McEwen BS, Stein M 1992 Adrenal steroid receptor activation in rat brain and pituitary following dexamethasone: implications for the dexamethasone suppression test. Biol Psychiatry 32:850–869

    Cole MA, Kim PJ, Kalman BA, Spencer RL 2000 Dexamethasone suppression of corticosteroid secretion: evaluation of the site of action by receptor measures and functional studies. Psychoneuroendocrinology 25:151–167

    Cordon-Cardo C, O’Brien JP, Casals D, Rittman-Grauer L, Biedler JL, Melamed MR, Bertino JR 1989 Multidrug-resistance gene (P-glycoprotein) is expressed by endothelial cells at blood-brain barrier sites. Proc Natl Acad Sci USA 86:695–698

    Schinkel AH, Wagenaar E, van Deemter L, Mol CAAM, Borst P 1995 Absence of the mdr1a P-Glycoprotein in mice affects tissue distribution and pharmacokinetics of dexamethasone, digoxin, and cyclosporin A. J Clin Invest 96:1698–1705

    Meijer OC, de Lange ECM, Breimer DD, de Boer AG, Workel JO, De Kloet ER 1998 Penetration of dexamethasone into brain glucocorticoid targets is enhanced in mdr1a P-glycoprotein knockout mice. Endocrinology 139:1789–1793

    Reul JMHM, van den Bosch JR, De Kloet ER 1987 Relative occupation of type-I and type-II corticosteroid receptors in the rat brain following stress and dexamethasone treatment: functional implications. J Endocrinology 115:459–467

    Ginsberg AB, Campeau S, Day HE, Spencer RL 2003 Acute glucocorticoid pretreatment suppresses stress-induced hypothalamic-pituitary-adrenal axis hormone secretion and expression of corticotropin-releasing hormone hnRNA but does not affect c-fos mRNA or fos protein expression in the paraventricular nucleus of the hypothalamus. J Neuroendocrinol 15:1075–1083

    Fluttert M, Dalm S, Oitzl MS 2000 A refined method for sequential blood sampling by tail incision in rats. Lab Anim 34:372–378

    Sibug RM, Compaan JC, Meijer OC, van der Gugten J, Olivier B, De Kloet ER 1998 Flesinoxan treatment reduces 5-HT1A receptor mRNA in the dentate gyrus independently of high plasma corticosterone levels. Eur J Pharmacol 353:207–214

    Meijer OC, Steenbergen PJ, De Kloet ER 2000 Differential expression and regional distribution of steroid receptor coactivators SRC-1 and SRC-2 in brain and pituitary. Endocrinology 141:2192–2199

    Veldhuis HD, van Koppen C, van Ittersum M, De Kloet ER 1982 Specificity of adrenal steroid receptor system in the rat hippocampus. Endocrinology 110:2044–2051

    Harbuz MS, Lightman SL 1989 Responses of hypothalamic and pituitary mRNA to physical and psychological stress in the rat. J Endocrinol 122:705–711

    Senba E, Umemoto S, Kawai Y, Noguchi K 1994 Differential expression of fos family and jun family mRNAs in the rat hypothalamo-pituitary-adrenal axis after immobilization stress. Brain Res Mol Brain Res 24:283–294

    Stumpf WE, Heiss C, Sar M, Duncan GE, Craver C 1989 Dexamethasone and corticosterone receptor sites. Differential topographic distribution in rat hippocampus revealed by high resolution autoradiography. Histochemistry 92:201–210

    Karssen AM, Meijer OC, van der Sandt IC, Lucassen PJ, de Lange EC, de Boer AG, de Kloet ER 2001 Multidrug resistance P-glycoprotein hampers the access of cortisol but not of corticosterone to mouse and human brain. Endocrinology 142:2686–2694

    Reul JMHM, De Kloet ER 1985 Two receptor systems for corticosterone in rat brain: microdistribution and differential occupation. Endocrinology 117:2505–2511

    McEwen BS, de Kloet R, Wallach G 1976 Interactions in vivo and in vitro of corticoids and progesterone with cell nuclei and soluble macromolecules from rat brain regions and pituitary. Brain Res 105:129–136

    Coutard M, Osborne-Pellegrin MJ, Funder JW 1978 Tissue distribution and specific binding of tritiated dexamethasone in vivo: autoradiographic and cell fractionation studies in the mouse. Endocrinology 103:1144–1152

    Karssen AM, Meijer OC, Van Der Sandt IC, De Boer AG, De Lange EC, De Kloet ER 2002 The role of the efflux transporter P-glycoprotein in brain penetration of prednisolone. J Endocrinol 175:251–260

    Uhr M, Holsboer F, Muller MB 2002 Penetration of endogenous steroid hormones corticosterone, cortisol, aldosterone and progesterone into the brain is enhanced in mice deficient for both mdr1a and mdr1b P-glycoproteins. J Neuroendocrinol 14:753–759

    Miller AH, Spencer RL, Stein M, McEwen BS 1990 Adrenal steroid receptor binding in spleen and thymus after stress or dexamethasone. Am J Physiol 259:E405–E412

    Spencer RL, Young EA, Choo PH, McEwen BS 1990 Adrenal steroid type I and type II receptor binding: estimates of in vivo receptor number, occupancy, and activation with varying level of steroid. Brain Res 514:37–48

    Kovacs KJ, Mezey E 1987 Dexamethasone inhibits corticotropin-releasing factor gene expression in the rat paraventricular nucleus. Neuroendocrinology 46:365–368

    Sawchenko PE 1987 Evidence for a local site of action for glucocorticoids in inhibiting CRF and vasopressin expression in the paraventricular nucleus. Brain Res 403:213–223

    Imaki T, Xiao-Quan W, Shibasaki T, Yamada K, Harada S, Chikada N, Naruse M, Demura H 1995 Stress-induced activation of neuronal activity and corticotropin-releasing factor gene expression in the paraventricular nucleus is modulated by glucocorticoids in rats. J Clin Invest 96:231–238

    Roozendaal B, McGaugh JL 1996 The memory-modulatory effects of glucocorticoids depend on an intact stria terminalis. Brain Res 709:243–250

    Feldman S, Weidenfeld J 2002 Further evidence for the central effect of dexamethasone at the hypothalamic level in the negative feedback mechanism. Brain Res 958:291–296

    Gass P, Kretz O, Wolfer DP, Berger S, Tronche F, Reichardt HM, Kellendonk C, Lipp HP, Schmid W, Schutz G 2000 Genetic disruption of mineralocorticoid receptor leads to impaired neurogenesis and granule cell degeneration in the hippocampus of adult mice. EMBO Rep 1:447–451

    Sloviter RS, Valiquette G, Abrams GM, Ronk EC, Sollas AL, Paul LA, Neubort S 1989 Selective loss of hippocampal granule cells in the mature rat brain after adrenalectomy. Science 243:535–538

    Hornsby CD, Grootendorst J, de Kloet ER 1996 Dexamethasone does not prevent seven-day ADX-induced apoptosis in the dentate gyrus of the rat hippocampus. Stress 1:51–64

    Hassan AH, von Rosenstiel P, Patchev VK, Holsboer F, Almeida OF 1996 Exacerbation of apoptosis in the dentate gyrus of the aged rat by dexamethasone and the protective role of corticosterone. Exp Neurol 140:43–52

    De Kloet ER, Vreugdenhil E, Oitzl MS, Joels M 1998 Brain corticosteroid receptor balance in health and disease. Endocr Rev 19:269–301

    De Kloet ER 1991 Brain corticosteroid receptor balance and homeostatic control. Front Neuroendocrinol 12:95–164

    Laugero KD, Gomez F, Manalo S, Dallman MF 2002 Corticosterone infused intracerebroventricularly inhibits energy storage and stimulates the hypothalamo-pituitary axis in adrenalectomized rats drinking sucrose. Endocrinology 143:4552–4562

    Laugero KD, Bell ME, Bhatnagar S, Soriano L, Dallman MF 2001 Sucrose ingestion normalizes central expression of corticotropin-releasing-factor messenger ribonucleic acid and energy balance in adrenalectomized rats: a glucocorticoid-metabolic-brain axis Endocrinology 142:2796–2804

    Oitzl MS, De Kloet ER 1992 Selective corticosteroid antagonists modulate specific aspects of spatial orientation learning. Behav Neurosci 106:62–71

    Sandi C 1998 The role and mechanisms of action of glucocorticoid involvement in memory storage. Neural Plast 6:41–52

    Kovacs KJ, Foldes A, Sawchenko PE 2000 Glucocorticoid negative feedback selectively targets vasopressin transcription in parvocellular neurosecretory neurons. J Neurosci 20:3843–3852

    Herman JP, Schafer MK, Thompson RC, Watson SJ 1992 Rapid regulation of corticotropin-releasing hormone gene transcription in vivo. Mol Endocrinol 6:1061–1069

    Melia KR, Ryabinin AE, Schroeder R, Bloom FE, Wilson MC 1994 Induction and habituation of immediate early gene expression in rat brain by acute and repeated restraint stress. J Neurosci 14:5929–5938

    Helmreich DL, Cullinan WE, Watson SJ 1996 The effect of adrenalectomy on stress-induced c-fos mRNA expression in the rat brain. Brain Res 706:137–144

    Brown ER, Sawchenko PE 1997 Hypophysiotropic CRF neurons display a sustained immediate-early gene response to chronic stress but not to adrenalectomy. J Neuroendocrinol 9:307–316

    Malkoski SP, Handanos CM, Dorin RI 1997 Localization of a negative glucocorticoid response element of the human corticotropin releasing hormone gene. Mol Cell Endocrinol 127:189–199

    Autelitano DJ 1994 Glucocorticoid regulation of c-fos, c-jun and transcription factor AP-1 in the AtT-20 corticotrope cell. J Neuroendocrinology 6:627–637

    Holsboer F, Barden N 1996 Antidepressants and hypothalamo-pituitary-adrenal regulation. Endocr Rev 17:187–205

    Holsboer F 2000 The corticosteroid receptor hypothesis of depression. Neuropsychopharmacology 23:477–501

    von Bardeleben U, Holsboer F, Stalla GK, Muller OA 1985 Combined administration of human corticotropin-releasing factor and lysine vasopressin induces cortisol escape from dexamethasone suppression in healthy subjects. Life Sci 37:1613–1618

    Mei Q, Richards K, Strong-Basalyga K, Fauty SE, Taylor A, Yamazaki M, Prueksaritanont T, Lin JH, Hochman J 2004 Using real-time quantitative TaqMan RT-PCR to evaluate the role of dexamethasone in gene regulation of rat P-glycoproteins mdr1a/1b and cytochrome P450 3A1/2. J Pharm Sci 93:2488–2496

    Kwan P, Sills GJ, Butler E, Gant TW, Brodie MJ 2003 Differential expression of multidrug resistance genes in naive rat brain. Neurosci Lett 339:33–36

    Dallman MF, Akana SF, Cascio CS, Darlington DN, Jacobson L, Levin N 1987 Regulation of ACTH secretion: variations on a theme of B. Recent Prog Horm Res 43:113–173

    Levin N, Shinsako J, Dallman MF 1988 Corticosterone acts on the brain to inhibit adrenalectomy-induced adrenocorticotropin secretion. Endocrinology 122:694–701

    Diorio D, Viau V, Meaney MJ 1993 The role of the medial prefrontal cortex (cingulate gyrus) in the regulation of hypothalamic-pituitary-adrenal responses to stress. J Neurosci 13:3839–3847

    Dallman MF, Akana SF, Levin N, Walker CD, Bradbury MJ, Suemaru S, Scribner KS 1994 Corticosteroids and the control of function in the hypothalamo-pituitary-adrenal (HPA) axis. Ann N Y Acad Sci 746:22–31;discussion 31- 2, 64–67(A. M. Karssen, O. C. Meijer, A. Berry, R)