Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

A New Anti-Depressive Strategy for the Elderly: Ablation of FKBP5/FKBP51

  • John C. O'Leary III,

    Affiliation Department of Molecular Medicine, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, Florida, United States of America

  • Sheetal Dharia,

    Affiliation College of Pharmacy, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, Florida, United States of America

  • Laura J. Blair,

    Affiliation Department of Molecular Medicine, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, Florida, United States of America

  • Sarah Brady,

    Affiliation Department of Molecular Medicine, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, Florida, United States of America

  • Amelia G. Johnson,

    Affiliation Department of Molecular Medicine, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, Florida, United States of America

  • Melinda Peters,

    Affiliation Department of Physiology and Pharmacology, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, Florida, United States of America

  • Joyce Cheung-Flynn,

    Affiliation Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America

  • Marc B. Cox,

    Affiliations Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas, United States of America, Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas, United States of America

  • Gabriel de Erausquin,

    Affiliation Department of Psychiatry and Neuroscience, University of South Florida, Tampa, Florida, United States of America

  • Edwin J. Weeber,

    Affiliation Department of Physiology and Pharmacology, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, Florida, United States of America

  • Umesh K. Jinwal,

    Affiliation College of Pharmacy, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, Florida, United States of America

  • Chad A. Dickey

    cdickey@health.usf.edu

    Affiliation Department of Molecular Medicine, Byrd Alzheimer's Research Institute, University of South Florida, Tampa, Florida, United States of America

Abstract

The gene FKBP5 codes for FKBP51, a co-chaperone protein of the Hsp90 complex that increases with age. Through its association with Hsp90, FKBP51 regulates the glucocorticoid receptor (GR). Single nucleotide polymorphisms (SNPs) in the FKBP5 gene associate with increased recurrence of depressive episodes, increased susceptibility to post-traumatic stress disorder, bipolar disorder, attempt of suicide, and major depressive disorder in HIV patients. Variation in one of these SNPs correlates with increased levels of FKBP51. FKBP51 is also increased in HIV patients. Moreover, increases in FKBP51 in the amygdala produce an anxiety phenotype in mice. Therefore, we tested the behavioral consequences of FKBP5 deletion in aged mice. Similar to that of naïve animals treated with classical antidepressants FKBP5−/− mice showed antidepressant behavior without affecting cognition and other basic motor functions. Reduced corticosterone levels following stress accompanied these observed effects on depression. Age-dependent anxiety was also modulated by FKBP5 deletion. Therefore, drug discovery efforts focused on depleting FKBP51 levels may yield novel antidepressant therapies.

Introduction

Genes regulating the hypothalamus-pituitary-adrenal (HPA) axis are associated with susceptibility to depression as well as antidepressant efficacy [1], [2], [3]. The HPA axis has a well-characterized role as a regulator of the neuroendocrine stress response [4]. Its activation leads to the production of glucocorticoids in the adrenal axis, of which the major constituent in humans is cortisol and in rodents is corticosterone. Over the past decade, genome wide association studies for single nucleotide polymorphisms (SNPs) revealed significant associations between susceptibility to depressive episodes and variants in both the NR3C1, that encodes the glucocorticoid receptor (GR), and FKBP5, that encodes a GR binding protein thought to attenuate GR activity [2], [5]. While most studies have focused on the variants in GR because of its role as a transcriptional regulator [6], the involvement of FKBP5 and its gene product, FKBP51, have received little attention. This is largely due to uncertainty about how to approach this relatively unknown protein. In fact, it remains to be proven whether FKBP51 is a valid therapeutic target for treating depression, despite its clear genetic link.

Since the initial discovery of the association between FKBP5 SNPs and depression, other psychiatric disorders have been found to be associated with FKBP5 SNPs including PTSD [7], bipolar disorder [8], anxiety [9], peritraumatic dissociation [10], and major depression in HIV patients [11]. The TT variant of the rs1360780 SNP was associated with both an increased incidence of depressive episodes throughout a carrier's lifetime, and increased sensitivity to common neurotransmitter-based anti-depressants [2]. Interestingly, individuals with the rs1360780 TT SNP had significantly higher FKBP51 protein levels in their lymphocytes. FKBP51 levels are also elevated in patients with HIV infection, perhaps playing a role in the depression that commonly occurs with chronic highly active antiretroviral therapies (HAART) [12]. Recently, stress was shown to induce neuropsin activity in the amygdala, inducing anxiety in mice through an FKBP51-dependent mechanism [13]. How FKBP51 directly modulates GR has been investigated in vitro. In these systems, upregulation of FKBP51 decreases the affinity of GR for its substrate. This in turn decreases the amount of GR that becomes transcriptionally active [9]. In new world monkeys a naturally occurring glucocorticoid resistance has been attributed to higher than normal levels of FKBP51. However, reduced GR activity in a transgenic mouse model of FKBP51 overexpression has never been shown [14], [15].

While the causes of major depressive disorders are unknown, there is an emerging genetic diathesis for its occurrence within genes regulating the HPA axis; however few animal models have been developed or utilized for aetiologic validation studies. Genetic variation in FKBP51 appears to be one factor that facilitates liability to anxiety and mood disorders. Thus, the goal of this study was to determine whether decreasing FKBP51 expression could make mice less susceptible to inducible “depression-like” states through a corticosterone-dependent mechanism in vivo in well established models with high predictive value [16]. Indeed, aged FKBP5-deficient mice were resistant to stress-induced depressive-like behavior. Moreover, despite robust hippocampal and forebrain expression patterns, deletion of FKBP5 did not result in cognitive impairment or other behavioral abnormalities. Circulating levels of corticosterone in the same FKBP5−/− mice were also reduced after stress, confirming the proposed mechanism previously described [9]. These data suggest that not only is FKBP51 a valid therapeutic target, but targeting this protein may also have minimal consequences for other behavioral characteristics.

Results

FKBP5−/− mice

FKBP5−/− mice were used to determine the effect of gene deletion on behavior. The mice contain a β-galactosidase reporter cassette, which expresses wherever the FKBP5 gene is normally expressed. To confirm gene knockout and establish cerebral distribution of FKBP51, tissue from 5.5 and 20-month-old FKBP5−/− mice was stained using an X-gal kit that produces a blue product when β-galactosidase is present. An age-dependent increase in β-gal expression was observed particularly in the upper cortical layers of the forebrain (Figure 1A). This was consistent with the age-dependent increase in FKBP51 expression that had been previously reported in normal mice [17]. To confirm the absence of FKBP51 protein, whole brain homogenates were analyzed by immunoblot and probed using an FKBP51 antibody. No detectable FKBP51 was observed (Figure 1B). To verify that FKBP51 mRNA was absent, oligo d(T) RT-PCR was performed using reverse transcriptase to produce cDNA. No FKBP5 PCR product was detected in the FKBP5−/− mice, confirming that no FKBP51 mRNA was present (Figure 1C).

thumbnail
Figure 1. Distribution and confirmation of gene knockout.

(A) Representative images of β-gal staining in horizontal brain slices of 5.5 and 20 month old FKBP5−/− mice. (B) Western blot analysis of 20-month old wildtype and FKBP5−/− whole brain homogenates. (C) FKBP5 primer-specific PCR of cDNA synthesized from brain-isolated mRNA by reverse transcription.

https://doi.org/10.1371/journal.pone.0024840.g001

Antidepressive behavior in FKBP5−/− mice

FKBP5−/− mice aged 17–20 months were submitted to two behavioral models of depressive-like activity; the classical forced swim test (FST) and the tail suspension test (TST) [18], [19]. Results from these tests are based on the total time spent immobile over a 6-minute period, interpreted as despair. These tests are typical for assessing antidepressant efficacy [18]. Aged FKBP5−/− mice displayed a shorter immobility time than their wildtype counterparts (Figure 2A–2B). The weight, activity, and physical fitness of the mice were evaluated to account for possible confounding factors, but these variables did not contribute to the effect (Figure 2C–2E).

thumbnail
Figure 2. FKBP5−/− mice display resistance to depression inducing stimulus.

(A) Total time immobile in the tail suspension test; *p<0.05, t = 2.870, df = 16. (B) Total time immobile in the forced swim test; **p<0.01, t = 2.982, df = 16. (C) No significant difference was observed between mass measurements of groups. (D) Latency to fall from a gradually accelerating rotorod apparatus displays no difference between groups. (E) Activity levels in the open field display no differences between groups.

https://doi.org/10.1371/journal.pone.0024840.g002

Effect of FKBP5−/− on corticosterone production

Stress causes increased levels of cortisol in humans and depressed patients have higher-than-normal levels of cortisol in the blood [20]. The dexamethasone-corticotropin releasing hormone (DEX-CRH) test, a commonly used tool to detect HPA system changes, has been used to link depressive behavior with glucocorticoid receptor (GR) insensitivity [21]. To determine if FKBP5 deletion was altering glucocorticoid levels, circulating corticosterone levels were assessed in the same aged cohort of FKBP5−/− mice that were used for behavioral analyses. Early morning blood draws were collected from both FKBP5−/− and wildtype littermates before and 30 minutes after being placed in a restrainer for 10 minutes. Levels of basal corticosterone were predictably low, consistent with previous reports showing low levels of corticosterone production in the earliest part of the murine diurnal cycle (Figure 3) [22]. After stress corticosterone levels rose in both wildtype and FKBP5−/− mice, but this induction was attenuated in the FKBP5−/− mice. These findings suggest that the lack of FKBP51 permits unrestrained GR transcription activity, which decreases HPA-axis activity and corticosterone levels, improving resilience to depressive-like behavior.

thumbnail
Figure 3. Effects of FKBP5−/− on corticosterone production.

Levels of corticosterone in serum right before a 10-minute tube restraint and 30 minute after (wt n = 5, FKBP5−/− n = 6); interaction F(1,18) = 11.54, **p<0.01; genotype F(1,18) = 11.72, **p<0.01; stress F(1,18) = 40.5, ***p<0.0001; Bonferroni multiple comparisons wt vs FKBP5−/− stress, t = 4.82, ***p<0.001.

https://doi.org/10.1371/journal.pone.0024840.g003

Characterization of anxious behavior in the FKBP5−/− model

Brain-specific knockout of GR in mice reduces anxiety [23]. Also, FKBP51 expression was recently correlated to anxious behavior [13]. Based on this and the novel link now established between FKBP51, GR activity and glucocorticoid production, the effects of FKBP5 deletion on anxiety were tested. The longitudinal impact of FKBP5 deletion on anxiety was assessed in mice aged 11–14 months and then again in the same mice aged 18–22 months using the elevated plus maze (EPM). A repeated measures two-way analysis of variance was conducted to examine the effect of time and genotype on anxiety. The behavioral correlates of anxiety in the EPM were significantly affected by age, and this change depended on the genotype at FKBP5 as reflected by a significant age*genotype interaction, (Figure 4A–4B). Indeed, in wildtype mice behavioral correlates of anxiety decreased with age, whereas they appeared to increase in FKBP5−/− mice. To further examine whether anxiety was indeed being modified in these mice, the 18–22 month-old mice were subjected to the light-dark chamber paradigm, which is another standard measure of anxiety. The mice were allowed to explore between a well-lit and unprotected area and a dark covered area that the mice had access to through a small opening. Normal mice typically move to the dark chamber. The light-dark maze did not reveal any statistical differences between the groups (Figure 4C–4D). These data suggest that suppression of FKBP51 may have a differential effect on anxiety depending on the developmental stage; however, the effect is not pervasive enough to manifest itself through several indices of anxiety.

thumbnail
Figure 4. Age-associated changes in anxious behavior in FKBP5−/− mice.

(A) Time spent in closed arms of the EPM (n = 8 for each group); interaction F(1,14) = 4.59, p = 0.0502; genotype F(1,14) = 0.14, p = 0.713; time F(1,14) = 8.38, *p<0.05. (B) Time spent in open arms of the EPM (n = 8 for each group); interaction F(1,14) = 8.30, *p<0.05; genotype F(1,14) = 0.0, p = 0.999; time F(1,14) = 6.33, *p<0.05. (C) Time spent in the areas of the dark-light chamber (wt n = 8, FKBP5−/− n = 8); interaction F(1,28) = 6.13, *p<0.05; genotype F(1,28) = 0, p = 0.999; amount of light in chamber F(1,28) = 57.9, ***p<0.0001. (D) Number of entries into the areas of the dark-light chamber (wt n = 8, FKBP5−/− n = 8); interaction F(1,26) = 0.01, p = 0.933; genotype F(1,26) = 5.82, *p<0.05; amount of light in chamber F(1,26) = 0.02, p = 0.879.

https://doi.org/10.1371/journal.pone.0024840.g004

Memory and general behavioral characterization of FKBP5−/− mice

Untoward consequences of FKBP51 ablation were a distinct possibility given the ubiquitous expression of FKBP51 throughout the brain. Given the high levels of expression of FKBP5 in the hippocampus (Figure 1A) memory formation was assessed. Therefore, short-term memory in the FKBP5−/− mice was tested with the use of the novel object recognition and Y-maze paradigms. Both tests are performed in a short period of time to capture the working memory ability of the mice. Neither test showed statistically significant differences between FKBP5−/− and wildtype littermates (Figure 5A–5B). Long-term spatial memory function was then tested using the Morris water maze (MWM). Memory retention was tested 24 hours after the training was completed. The training phase of the MWM displayed that wildtype and FKBP5−/− mice were equally capable of learning the location of a hidden platform (Figure 5C). Moreover wildtype and FKBP5−/− mice displayed an equivalent capacity to locate the hidden platform 24 hours after training (Figure 5D).

thumbnail
Figure 5. Deletion of the FKBP5 gene does not alter learning and memory despite robust expression of FKBP5 in the hippocampus.

(A) Discrimination index between familiar and novel object in the novel object recognition test. (B) Percent of spontaneous alternation shows no difference between the groups. (C) Learning of the location of the hidden platform in the MWM (interaction F(5,80) = 0.14, p = 0.98; genotype F(1,16) = 0.0, p = 0.98; time F(5,80) = 12.71, p<0.0001). (D) Percent time spent in quadrant during the probe trial of MWM. (E) Software generated heat plot representing amount of time spent in an area. (F) Percent freezing during training portion of fear conditioning; interaction F(6,90) = 4.1, **p = 0.001; genotype F(1,15) = 5.66, *p = 0.031; time F(6,90) = 79.89, ***p<0.0001; Bonferroni multiple comparisons wt vs FKBP5−/− at 240 s, t = 2.42, ***p<0.001. (G) Percent freezing during contextual exposure to fear-conditioning chamber. (H) Percent freezing during the cued fear-conditioning test; interaction F(1,30) = 0.05, p = 0.828; genotype F(1,30) = 0.25, p = 0.624; tone F(1,30) = 55.75, ***p<0.0001.

https://doi.org/10.1371/journal.pone.0024840.g005

The amygdala controls emotional learning in the brain and FKBP5 expression can be upregulated by GR activation [24]. As a consequence, function of the amygdala could be affected by FKBP5 deletion. To test the impact of FKBP5−/− on emotionally derived memory function, a fear-conditioning paradigm was employed. In this test, a tone played for 30 seconds is followed by a small foot shock. Thus the animal learns to associate the tone with the shock. The amygdala-associated fear response caused by the tone is able to bypass the hippocampus once the association is made, allowing for assessment of emotionally driven memory formation. Surprisingly, no differences between wildtype and FKBP5−/− mice were observed in either contextual (environmentally-based) or cued (tone-based) fear conditioning paradigms (Figure 5E–5G). These findings suggest that neither spatially nor emotionally driven long-term memory is affected by FKBP5 deletion. In addition to these learning and memory tasks, no observable differences between wildtype and FKBP5−/− mice were noted in tasks designed to assess activity, motor performance, motor coordination, motor learning, hearing, or prepulse inhibition, which measures startle response inhibition (Figure 6).

thumbnail
Figure 6. Behavioral characterization of FKBP5−/− mice.

(A) Distance traveled in the open field test. (B) Assessment of motor learning from comparison between the average of all four trials of rotorod from day one with those of day two; interaction F(1,16) = 1.28, p = .275; genotype F(1,16) = 1.37, p = 0.259; time F(1,16) = 80.3, ***p<0.0001. (C) Latency to fall from the rotorod apparatus; interacton F(7,112) = 1.28, p = 0.267; genotype F(1,16) = 1.37, p = 0.259; time F(7,112) = 17.97, ***p<0.0001. (D) Percent inhibition of startle response by increasing prepulse intensity; interaction F(4,70) = 0.61, p = 0.656; genotype F(1,70) = 0.33,p = 0.565; prepulse intensity F(4,70) = 3.07, *p<0.05. (E) Comparison of no pulse (no tone) with a 120 dB pulse (tone) to test hearing; interaction F(1,32) = 0.11, p = 0.74; genotype F(1,32) = 0.16, p = 0.694; pulse F(1,32) = 147.58, ***p<0.0001.

https://doi.org/10.1371/journal.pone.0024840.g006

Discussion

Major depression is a devastating disease with a course that is frequently chronic or recurrent and affects millions of people. Research in the last decade has shown that variation in the FKBP5 gene is associated with depression and several other mood and anxiety disorders. And although in vitro data suggests the possibility of a causal relationship between FKBP5 expression levels and depression, this has never been tested in vivo. Here we show for the first time that ablation of FKBP5 in mice led to reduced immobility in two behavioral models that are routinely used to assess anti-depressant efficacy. This behavioral effect coincided with attenuation of corticosterone production after a stressful episode. Moreover, no defects in locomotion, somato-sensation or learning and memory were observed. Thus, therapies developed to reduce FKBP51 levels may be highly efficacious as next generation anti-depressants. Furthermore, because FKBP51 ablation results in reduced anxiety-like behavior in mice [13], we provide experimental support for the notion that genetically-driven variations in expression of FKBP51 may underlie susceptibility to anxiety and mood disorders, as suggested by association studies in humans [2], [9].

FKBP51 is a peptidyl-prolyl cis-trans isomerase (PPIase) enzyme that also associates with the chaperone Hsp90 and is distributed ubiquitously throughout the brain. This isomerase activity is thought to be important for structural rearrangements and phosphorylation dynamics of client proteins bound by Hsp90. Several diseases in addition to psychiatric conditions have implicated FKBP51 as having a role in their pathogenesis. These include prostate cancer [25], and neurodegenerative diseases, specifically tauopathies [17]. In fact, these effects on tau may somehow be tied to the manifestation of these psychiatric conditions. Indeed, depleting FKBP51 levels was shown to also reduce tau levels, while inhibiting its PPIase activity actually lead to increased stability of phosphorylated tau. Thus it is certainly possible that FKBP51 is involved in Alzheimer's disease progression, since one of its earliest clinical features is depression. More recently, the extracellular protease neuropsin was shown to mediate anxiety-like behavior via an FKBP51 dependent mechanism [13]. Thus, an important role for FKBP51 in maintaining proper brain function is emerging. Its relationship with major depressive disorder in HIV, bipolar disorder and possibly anxiety and Alzheimer's disease further underlie its significance.

Current treatment for depression includes the use of medications that extend the amount of time neurotransmitters are present in the synaptic cleft including serotonin, norepinephrine, and dopamine. It is estimated that 60–70% of patients reach remission with the use of anti-depressant drugs [26]. These low rates of efficacy have prompted research into other potential therapeutic targets in the HPA axis, particularly GR. However, there are many different isoforms of GR, making selective targeting with compounds challenging. Therefore, the results presented here show that FKBP51 may be the most appropriate target for treating depression via the modulation of the HPA axis in terms of its risk/benefit equation and potential therapeutic window. Also, and most noteworthy, because FKBP51 may act on the genetic liability to abnormal mood and anxiety states, it may provide a much needed treatment tool for secondary prophylaxis of depression recurrence and relapse.

Materials and Methods

Generation of FKBP5−/− mice

The mice have been generated as published previously [27]. Briefly, by PCR screening the 129SvJ mouse BAC library (Genome Systems, St. Louis, MO), bacterial artificial chromosome (BAC) clones that contained genomic regions for FKBP5 were isolated. Restriction fragments were subcloned into pBluescript (pBS; Stratagene, La Jolla, CA) or pZero (Invitrogen, Carlsbad, CA) cloning vectors. The PCR products were amplified from the BAC clones and were then used to construct a targeting vector in the pPGKneo vector (a generous gift of James Lee, Mayo Clinic Scottsdale). The targeting vector contained a beta-galactosidase/neomycin cassette flanked by regions homologous to the FKBP5 gene. Due to the size of the protein it is more practical to partially delete the gene. Thus, when the targeting vector integrates into the chromosome through homologous recombination it removes all of exon 2, which is the first coding exon. Since the only deleted portion of the gene is exon 2 the expression of the beta-galactosidase protein is dependent on the FKBP5 promoter and transcription machinery and expresses in frame with the initiation codon. ES cells were isolated from the 129SvJ mouse and cultured in Knockout DMEM media (Invitrogen) supplemented with 10% FBS, penicillin/streptomycin, essential amino acids, and ESGRO (103 U/ml; Chemicon, Temecula, CA) with irradiated embryonic fibroblast feeder cells. The ES cells were then electroporated at 0.2 kV, 950 µF (Gene Pulser II; Bio-Rad, Hercules, CA) with linearized targeting vectors and selected with G418. DNA from G418-resistant clones was isolated for Southern blot analysis. A DNA probe was used to distinguish PstI restriction fragments from wildtype allele (∼7.5 kb) and targeting vector (∼10 kb). Appropriate homologous recombination in ES cell clones was confirmed by PCR using primers complementary to sequences within the neomycin cassette and to 3′ FKBP5 sequences downstream from the recombination site. ES cell clones containing the targeting vector were injected into C57BL/6 blastocysts and implanted into pseudopregnant 129SvJ females. Chimeric offspring were identified by coat patterns and mated to C57BL/6 mice to obtain germline transmission of the targeting vector. For colony maintenance mice were crossed from C57BL6 onto Swiss-Webster for purposes of fecundity and genetic diversity to be more representative of a human population.

Brain Tissue Fractionation and Western Blot Analysis

Brain tissue fractionation and western blot analysis were done as previously described [28].

PCR

mRNA was isolated and purified from the brain of four wildtype and four FKBP5−/− mice using RNAeasy kit (QIAGEN, Valencia, CA). cDNA was synthesized from isolated mRNA by reverse transcription using Super Script III First-Strand cDNA Sythesis Kit (Invitrogen, Carlsbad, CA) from 50 ng of isolated mRNA. PCR was performed with synthesized cDNA and FKBP5 specific primers to confirm presence or absence of FKBP5 gene.

Antibodies

Horseradish peroxidase conjugated secondary antibodies (Southern Biotech, Birmingham, AL), Glyceraldehyde-3-phosphate dehydrogenase antibody (Meridian Life Science, Saco, ME), Anti-FKBP51 was provided by Drs. David F. Smith and Marc Cox (Mayo Clinic, Scottsdale, AZ).

Immunohistochemistry

Fixed mouse brains were processed for sectioning as previously described [29]. β-galactosidase staining was performed using the in situ β-gal staining kit (Stratagene, La Jolla, CA).

Behavior

N = 9 unless otherwise noted. Video tracking software was used in several tests (ANY-Maze, Stoelting, Illinois).

Open Field

Animals were monitored for 15 min in an open field with video tracking software.

Rotorod test

Testing started at an initial rotation of 4 rpm accelerating to 40 rpm over 5 min. Mice were tested for 4 trials per day, for 2 consecutive days with a 30-min intertrial interval. Latency to fall from the rod onto a spring-cushioned lever was measured.

Morris water maze (MWM)

Mice were trained to locate an escape platform hidden beneath the water (3 centimeter). Each mouse was given 4 trials per day with an intertrial interval of 1 hour for 6 consecutive days. Each animal was given 60 seconds to find the platform. Afterwards the mice were placed on the platform for 30 s. On day 7, mice were subjected to a trial in which the platform was removed, and had 60 s to search for it.

Associative Fear Conditioning

Two mild foot shocks (0.5 milliamps) were paired with an auditory conditioned stimulus (CS, white noise, 70 decibels) within a novel environment. The CS was given for 30 s before each foot shock (2 s). Twenty-four hours later, the mice were placed in the chamber and monitored for freezing for 3 min (no shocks or CS). Immediately after the test, mice were placed into a novel context for 3 min without CS and then exposed to the CS for 3 min (cued).

Prepulse Inhibition (PPI)

Mice were placed in a restrainer (Panlab, Barcelona Spain) and placed inside a sound attenuation chamber. The test consisted of 7 trial types in pseudorandom order: 1) 40 ms, 120 decibels sound burst (startle); 2–6) 5 different acoustic prepulses 100 ms in length, a 20 ms duration at 74, 78, 82, 86, and 90 dB; 7) no stimulus for baseline measurement. The intertrial interval was 15 s. The startle response peak was measured within a second after the stimulus.

Elevated Plus Maze (EPM)

EPM consisted of 2 open arms facing each other and two enclosed arms also facing each other. Each arm is attached to the center platform and elevated 40 centimeters off the floor. The mouse was placed on the platform and allowed to explore for 5 min. Video tracking software measured movement.

Porsolt forced swim test (FST)

Each mouse was placed in a 45 centimeters high and 20 centimeters diameter clear Plexiglas cylinder filled with room temperature water to a depth of 12 centimeters for 6 min. Amount of time spent immobile was recorded.

Tail Suspension Test (TST)

Mice were suspended from their tail for 6 min. Amount of time spent immobile was recorded.

Novel Object

Mice were placed in an area with two objects similar in scale to the mouse. Each animal was given 3 acclimation trials of 5 min with a 5-min intertrial interval. Then one acclimated object was replaced with a novel object. Animals were given a 5-min exploratory trial monitored by video recording.

Y-maze

Animals were started at the center of the Y and allowed to explore for 8 min. Each session was video-monitored. The number of arm entries was recorded. The percent of spontaneous alternation was calculated as the number of triads containing entries into all three arms divided by the maximum possible of alternations (total number of entries minus 2).

Corticosterone Assay, blood collection and stress paradigm

The levels of corticosterone were measured using an ELISA kit (Enzo Life Sciences, Plymouth Meeting, PA). Blood from mice was collected in the morning one hour after the light cycle began and 30 min after a 10-min tube restraint using the submandibular vein puncture method.

Statistics

The student's t-test was used to compare 2 groups. The paired t-test was used to compare paired observations within 2 groups. The 2-way RMANOVA was used to compare the interaction between two dependent variables and an independent variable. The Bonferroni post test was used to correct for multiple testing. All error bars represent S.EM.

Animal Study Approval

All animal procedures were approved by the University of South Florida Institutional Animal Care and Use Committee (IACUC), protocol #R3848.

Acknowledgments

We would like to acknowledge Dr. David F. Smith for antibodies and for the FKBP5−/− mice.

Author Contributions

Conceived and designed the experiments: CAD JCO. Performed the experiments: JCO LJB SB AGJ UKJ JC-F MBC. Analyzed the data: JCO GdE. Contributed reagents/materials/analysis tools: MP EJW. Wrote the paper: CAD JCO JC-F MBC EJW GdE SD.

References

  1. 1. van Rossum EF, Binder EB, Majer M, Koper JW, Ising M, et al. (2006) Polymorphisms of the glucocorticoid receptor gene and major depression. Biological psychiatry 59: 681–688.
  2. 2. Binder EB, Salyakina D, Lichtner P, Wochnik GM, Ising M, et al. (2004) Polymorphisms in FKBP5 are associated with increased recurrence of depressive episodes and rapid response to antidepressant treatment. Nat Genet 36: 1319–1325.
  3. 3. Liu Z, Zhu F, Wang G, Xiao Z, Wang H, et al. (2006) Association of corticotropin-releasing hormone receptor1 gene SNP and haplotype with major depression. Neuroscience letters 404: 358–362.
  4. 4. Lupien SJ, McEwen BS, Gunnar MR, Heim C (2009) Effects of stress throughout the lifespan on the brain, behaviour and cognition. Nature reviews Neuroscience 10: 434–445.
  5. 5. Lahti J, Raikkonen K, Bruce S, Heinonen K, Pesonen AK, et al. (2011) Glucocorticoid receptor gene haplotype predicts increased risk of hospital admission for depressive disorders in the Helsinki birth cohort study. J Psychiatr Res 45: 1160–1164.
  6. 6. Derijk RH, van Leeuwen N, Klok MD, Zitman FG (2008) Corticosteroid receptor-gene variants: modulators of the stress-response and implications for mental health. European journal of pharmacology 585: 492–501.
  7. 7. Xie P, Kranzler HR, Poling J, Stein MB, Anton RF, et al. (2010) Interaction of FKBP5 with childhood adversity on risk for post-traumatic stress disorder. Neuropsychopharmacology: official publication of the American College of Neuropsychopharmacology 35: 1684–1692.
  8. 8. Willour VL, Chen H, Toolan J, Belmonte P, Cutler DJ, et al. (2009) Family-based association of FKBP5 in bipolar disorder. Molecular psychiatry 14: 261–268.
  9. 9. Binder EB (2009) The role of FKBP5, a co-chaperone of the glucocorticoid receptor in the pathogenesis and therapy of affective and anxiety disorders. Psychoneuroendocrinology 34: Suppl 1S186–195.
  10. 10. Koenen KC, Saxe G, Purcell S, Smoller JW, Bartholomew D, et al. (2005) Polymorphisms in FKBP5 are associated with peritraumatic dissociation in medically injured children. Molecular psychiatry 10: 1058–1059.
  11. 11. Tatro ET, Nguyen TB, Bousman CA, Masliah E, Grant I, et al. (2010) Correlation of major depressive disorder symptoms with FKBP5 but not FKBP4 expression in human immunodeficiency virus-infected individuals. Journal of neurovirology 16: 399–404.
  12. 12. Tatro ET, Everall IP, Masliah E, Hult BJ, Lucero G, et al. (2009) Differential expression of immunophilins FKBP51 and FKBP52 in the frontal cortex of HIV-infected patients with major depressive disorder. Journal of neuroimmune pharmacology: the official journal of the Society on NeuroImmune Pharmacology 4: 218–226.
  13. 13. Attwood BK, Bourgognon JM, Patel S, Mucha M, Schiavon E, et al. (2011) Neuropsin cleaves EphB2 in the amygdala to control anxiety. Nature 464: 1201–1204.
  14. 14. Denny WB, Prapapanich V, Smith DF, Scammell JG (2005) Structure-function analysis of squirrel monkey FK506-binding protein 51, a potent inhibitor of glucocorticoid receptor activity. Endocrinology 146: 3194–3201.
  15. 15. Westberry JM, Sadosky PW, Hubler TR, Gross KL, Scammell JG (2006) Glucocorticoid resistance in squirrel monkeys results from a combination of a transcriptionally incompetent glucocorticoid receptor and overexpression of the glucocorticoid receptor co-chaperone FKBP51. J Steroid Biochem Mol Biol 100: 34–41.
  16. 16. Frazer A, Morilak DA (2005) What should animal models of depression model? Neuroscience and biobehavioral reviews 29: 515–523.
  17. 17. Jinwal UK, Koren J 3rd, Borysov SI, Schmid AB, Abisambra JF, et al. (2010) The Hsp90 cochaperone, FKBP51, increases Tau stability and polymerizes microtubules. The Journal of neuroscience: the official journal of the Society for Neuroscience 30: 591–599.
  18. 18. Steru L, Chermat R, Thierry B, Simon P (1985) The tail suspension test: a new method for screening antidepressants in mice. Psychopharmacology 85: 367–370.
  19. 19. Porsolt RD, Le Pichon M, Jalfre M (1977) Depression: a new animal model sensitive to antidepressant treatments. Nature 266: 730–732.
  20. 20. Burke HM, Davis MC, Otte C, Mohr DC (2005) Depression and cortisol responses to psychological stress: a meta-analysis. Psychoneuroendocrinology 30: 846–856.
  21. 21. Holsboer F, von Bardeleben U, Wiedemann K, Muller OA, Stalla GK (1987) Serial assessment of corticotropin-releasing hormone response after dexamethasone in depression. Implications for pathophysiology of DST nonsuppression. Biological psychiatry 22: 228–234.
  22. 22. Kakihana R, Moore JA (1976) Circadian rhythm of corticosterone in mice: the effect of chronic consumption of alcohol. Psychopharmacologia 46: 301–305.
  23. 23. Tronche F, Kellendonk C, Kretz O, Gass P, Anlag K, et al. (1999) Disruption of the glucocorticoid receptor gene in the nervous system results in reduced anxiety. Nature genetics 23: 99–103.
  24. 24. Scharf SH, Liebl C, Binder EB, Schmidt MV, Muller MB (2011) Expression and regulation of the Fkbp5 gene in the adult mouse brain. PLoS One 6: e16883.
  25. 25. Ni L, Yang CS, Gioeli D, Frierson H, Toft DO, et al. (2010) FKBP51 promotes assembly of the Hsp90 chaperone complex and regulates androgen receptor signaling in prostate cancer cells. Mol Cell Biol 30: 1243–1253.
  26. 26. Rush AJ, Trivedi MH, Wisniewski SR, Nierenberg AA, Stewart JW, et al. (2006) Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. The American journal of psychiatry 163: 1905–1917.
  27. 27. Tranguch S, Cheung-Flynn J, Daikoku T, Prapapanich V, Cox MB, et al. (2005) Cochaperone immunophilin FKBP52 is critical to uterine receptivity for embryo implantation. Proc Natl Acad Sci U S A 102: 14326–14331.
  28. 28. Dickey CA, Yue M, Lin WL, Dickson DW, Dunmore JH, et al. (2006) Deletion of the ubiquitin ligase CHIP leads to the accumulation, but not the aggregation, of both endogenous phospho- and caspase-3-cleaved tau species. J Neurosci 26: 6985–6996.
  29. 29. Gordon MN, Holcomb LA, Jantzen PT, DiCarlo G, Wilcock D, et al. (2002) Time course of the development of Alzheimer-like pathology in the doubly transgenic PS1+APP mouse. Exp Neurol 173: 183–195.