Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Brain Res ; 1715: 106-114, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30880118

ABSTRACT

In previous studies, early-life fibroblast growth factor-2 (FGF2) administration conferred resilience to developing anxiety-like behavior in vulnerable animals in adulthood. To follow up on this work, we administered FGF2 the day after birth to animals that differ in emotional behavior and further explored its long-term effects on affective behavior and circuitry. Selectively-bred "high responder" rats (bHRs) exhibit low levels of anxiety-like and depression-like behavior, whereas selectively-bred "low responders" (bLRs) display high levels of anxiety-like and depression-like behavior. We found that early-life administration of FGF2 decreased negative affect in bLRs during the early post-natal period, as indexed by 40 kHz ultrasonic vocalizations (USVs) in response to a brief maternal separation on PND11. FGF2 also increased positive affect during the juvenile period, as measured by 50 kHz USVs in response to heterospecific hand play ("tickling") after weaning. In general, we found that bHRs produced more 50 kHz USVs than bLRs. In adulthood, we measured opioid ligand and receptor expression in brain regions implicated in USV production and affect regulation by mRNA in situ hybridization. Within multiple affective brain regions, bHRs had greater expression of the mu opioid receptor than bLRs. FGF2 increased mu opioid expression in bLRs. The bLRs had more kappa and less delta receptor expression than bHRs, and FGF2 increased prodynorphin in bLRs. Our results provide support for further investigations into the role of growth factors and endogenous opioids in the treatment of disorders characterized by altered affect, such as anxiety and depression.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Receptors, Opioid, mu/metabolism , Vocalization, Animal/physiology , Animals , Anxiety/metabolism , Behavior, Animal/drug effects , Emotions , Exploratory Behavior/drug effects , Fibroblast Growth Factor 2/metabolism , Male , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley , Ultrasonic Waves , Vocalization, Animal/drug effects
2.
Neuropsychopharmacology ; 44(2): 356-363, 2019 01.
Article in English | MEDLINE | ID: mdl-29703997

ABSTRACT

Recent evidence highlights the fibroblast growth factor (FGF) family in emotion modulation. Although ligands that activate FGF receptors have antidepressant and anxiolytic effects in animal models, FGF ligands have a broad range of actions both in the brain and the periphery. Therefore, identifying molecular partners that may function as allosteric modulators could offer new avenues for drug development. Since neural cell adhesion molecule (NCAM) activates FGF receptors, we asked whether peripherally administered NCAM peptide mimetics penetrate the brain and alter the behavior of standardized tests that have predictive validity for drug treatments of anxiety or depression. The NCAM peptide mimetic, plannexin, acutely increased and chronically decreased anxiety, but did not have antidepressant effects in rats. Another NCAM peptide mimetic, FGLL, had acute anxiogenic effects and chronic antidepressant effects in rats. A related NCAM peptide mimetic, FGLS, had antidepressant effects without modulating anxiety-like behavior, and these antidepressant effects were blocked by an AMPA receptor antagonist. Cisternal cerebrospinal fluid (CSF) levels of FGLs correlated with blood plasma levels in rats and non-human primates, and CSF-to-blood ratios of FGLS were comparable in both species. Results indicate that NCAM peptide mimetics penetrate the brain and support the suggestion that FGLS may be a candidate for further development as a novel treatment for major depressive disorder in humans.


Subject(s)
Anxiety/drug therapy , Behavior, Animal/drug effects , Depression/drug therapy , Emotions/drug effects , Neural Cell Adhesion Molecules/pharmacology , Oligopeptides/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Anxiety/metabolism , Brain/drug effects , Depression/metabolism , Male , Motor Activity/drug effects , Neural Cell Adhesion Molecules/metabolism , Neural Cell Adhesion Molecules/therapeutic use , Oligopeptides/metabolism , Oligopeptides/therapeutic use , Rats , Rats, Sprague-Dawley , Saimiri
3.
Neuropsychopharmacology ; 42(8): 1706-1714, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28205604

ABSTRACT

Individuals respond differently to traumatic experiences, including their propensity to develop posttraumatic stress disorder (PTSD). Understanding individual differences in PTSD vulnerability will allow the development of improved prevention and treatment options. Here we characterized fear conditioning and extinction in rats selectively bred for differences in their locomotor response to a novel environment. Selectively bred high-responder (bHR) and low-responder (bLR) male rats are known to differ in their emotional reactivity on a range of measures of spontaneous anxiety- and depressive-like behaviors. We demonstrate that bHRs have facilitated extinction learning and retention compared with outbred Sprague Dawley rats, whereas bLRs show reduced extinction learning and retention. This indicates that bLRs are more vulnerable to PTSD-like behavior. Fibroblast growth factor 2 (FGF2) has previously been implicated in the development of these behavioral phenotypes and facilitates extinction learning in outbred animals, therefore we examined the effects of early-life FGF2 on bHR and bLR behavior. FGF2 administered on the day after birth facilitated extinction learning and retention in bHRs, but not in bLRs or control rats, during adulthood. This indicates that, under the current fear conditioning paradigm, early-life FGF2 has protective effects only in resilient animals. This stands in contrast to FGF2's ability to protect vulnerable animals in milder tests of anxiety. These results provide a unique animal model of individual differences in PTSD-like behavior, allowing the study of genetic, developmental, and environmental factors in its expression.


Subject(s)
Behavior, Animal , Disease Susceptibility/psychology , Fibroblast Growth Factor 2/pharmacology , Individuality , Stress Disorders, Post-Traumatic/psychology , Aging/drug effects , Animals , Conditioning, Psychological , Environment , Extinction, Psychological , Fear , Male , Motor Activity , Rats , Rats, Inbred Strains
4.
Proc Natl Acad Sci U S A ; 112(38): 11953-8, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26351673

ABSTRACT

Both gene expression profiling in postmortem human brain and studies using animal models have implicated the fibroblast growth factor (FGF) family in affect regulation and suggest a potential role in the pathophysiology of major depressive disorder (MDD). FGF2, the most widely characterized family member, is down-regulated in the depressed brain and plays a protective role in rodent models of affective disorders. By contrast, using three microarray analyses followed by quantitative RT-PCR confirmation, we show that FGF9 expression is up-regulated in the hippocampus of individuals with MDD, and that FGF9 expression is inversely related to the expression of FGF2. Because little is known about FGF9's function in emotion regulation, we used animal models to shed light on its potential role in affective function. We found that chronic social defeat stress, an animal model recapitulating some aspects of MDD, leads to a significant increase in hippocampal FGF9 expression, paralleling the elevations seen in postmortem human brain tissue. Chronic intracerebroventricular administration of FGF9 increased both anxiety- and depression-like behaviors. In contrast, knocking down FGF9 expression in the dentate gyrus of the hippocampus using a lentiviral vector produced a decrease in FGF9 expression and ameliorated anxiety-like behavior. Collectively, these results suggest that high levels of hippocampal FGF9 play an important role in the development or expression of mood and anxiety disorders. We propose that the relative levels of FGF9 in relation to other members of the FGF family may prove key to understanding vulnerability or resilience in affective disorders.


Subject(s)
Affect , Fibroblast Growth Factor 9/metabolism , Adult , Affect/drug effects , Aged , Aged, 80 and over , Animals , Anxiety/complications , Anxiety/metabolism , Avoidance Learning/drug effects , Case-Control Studies , Demography , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Depressive Disorder, Major/complications , Depressive Disorder, Major/metabolism , Female , Fibroblast Growth Factor 9/administration & dosage , Fibroblast Growth Factor 9/genetics , Fibroblast Growth Factor 9/pharmacology , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Humans , Lentivirus/metabolism , Male , Microinjections , Middle Aged , Postmortem Changes , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Stress, Psychological/complications , Stress, Psychological/genetics , Young Adult
5.
Proc Natl Acad Sci U S A ; 111(32): 11834-9, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25071177

ABSTRACT

Posttranslational modifications of histone tails in chromatin template can result from environmental experiences such as stress and substance abuse. However, the role of epigenetic modifications as potential predisposing factors in affective behavior is less well established. To address this question, we used our selectively bred lines of high responder (bHR) and low responder (bLR) rats that show profound and stable differences in affective responses, with bLRs being prone to anxiety- and depression-like behavior and bHRs prone to addictive behavior. We first asked whether these phenotypes are associated with basal differences in epigenetic profiles. Our results reveal broad between-group differences in basal levels of trimethylated histone protein H3 at lysine 9 (H3K9me3) in hippocampus (HC), amygdala, and nucleus accumbens. Moreover, levels of association of H3K9me3 at Glucocorticoid Receptor (GR) and Fibroblast growth Factor 2 (FGF2) promoters differ reciprocally between bHRs and bLRs in these regions, consistent with these genes' opposing levels of expression and roles in modulating anxiety behavior. Importantly, this basal epigenetic pattern is modifiable by FGF2, a factor that modulates anxiety behavior. Thus, early-life FGF2, which decreases anxiety, altered the levels of H3K9me3 and its binding at FGF2 and GR promoters of bLRs rendering them more similar to bHRs. Conversely, knockdown of HC FGF2 altered both anxiety behavior and levels of H3K9me3 in bHRs, rendering them more bLR-like. These findings implicate FGF2 as a modifier of epigenetic mechanisms associated with emotional responsiveness, and point to H3K9me3 as a key player in the regulation of affective vulnerability.


Subject(s)
Affect/physiology , Behavior, Animal/physiology , Emotions/physiology , Epigenesis, Genetic , Fibroblast Growth Factor 2/genetics , Histones/chemistry , Histones/metabolism , Animals , Chromatin Assembly and Disassembly , DNA Methylation , Fibroblast Growth Factor 2/deficiency , Gene Knockdown Techniques , Hippocampus/metabolism , Promoter Regions, Genetic , Rats , Receptors, Glucocorticoid/genetics
6.
Neuropsychopharmacology ; 39(12): 2816-23, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24896613

ABSTRACT

Addiction is characterized by a high propensity for relapse, in part because cues associated with drugs can acquire Pavlovian incentive motivational properties, and acting as incentive stimuli, such cues can instigate and invigorate drug-seeking behavior. There is, however, considerable individual variation in the propensity to attribute incentive salience to reward cues. Discrete and localizable reward cues act as much more effective incentive stimuli in some rats ('sign-trackers', STs), than others ('goal-trackers', GTs). We asked whether similar individual variation exists for contextual cues associated with cocaine. Cocaine context conditioned motivation was quantified in two ways: (1) the ability of a cocaine context to evoke conditioned hyperactivity and (2) the ability of a context in which cocaine was previously self-administered to renew cocaine-seeking behavior. Finally, we assessed the effects of intra-accumbens core flupenthixol, a nonselective dopamine receptor antagonist, on context renewal. In contrast to studies using discrete cues, a cocaine context spurred greater conditioned hyperactivity, and more robustly renewed extinguished cocaine seeking in GTs than STs. In addition, cocaine context renewal was blocked by antagonism of dopamine receptors in the accumbens core. Thus, contextual cues associated with cocaine preferentially acquire motivational control over behavior in different individuals than do discrete cues, and in these individuals the ability of a cocaine context to create conditioned motivation for cocaine requires dopamine in the core of the nucleus accumbens. We speculate that different individuals may be preferentially sensitive to different 'triggers' of relapse.


Subject(s)
Cocaine-Related Disorders/physiopathology , Cocaine-Related Disorders/psychology , Cues , Drug-Seeking Behavior/physiology , Extinction, Psychological/physiology , Individuality , Animals , Cocaine/administration & dosage , Cocaine-Related Disorders/drug therapy , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Dopamine Antagonists/pharmacology , Dopamine Uptake Inhibitors/administration & dosage , Drug-Seeking Behavior/drug effects , Extinction, Psychological/drug effects , Flupenthixol/pharmacology , Male , Motivation , Motor Activity/drug effects , Motor Activity/physiology , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiopathology , Rats, Sprague-Dawley , Self Administration , Spatial Behavior/drug effects , Spatial Behavior/physiology
7.
Neuropsychologia ; 51(2): 372-6, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23182725

ABSTRACT

Research in schizophrenia has tended to emphasize deficits in higher cognitive abilities, such as attention, memory, and executive function. Here we provide evidence for dysfunction at a more fundamental level of perceptual processing, temporal integration. On a measure of flicker fusion, patients with schizophrenia exhibited significantly lower thresholds than age and education matched healthy controls. We reasoned that this finding could result from a longer window of temporal integration or could reflect diminished repetition suppression: if every frame of the repeating stimulus were represented as novel, its perceived duration would be accordingly longer. To tease apart these non-exclusive hypotheses, we asked patients to report the number of stimuli perceived on the screen at once (numerosity) as they watched rapidly flashing stimuli that were either repeated or novel. Patients reported significantly higher numerosity than controls in all conditions, again indicating a longer window of temporal integration in schizophrenia. Further, patients showed the largest difference from controls in the repeated condition, suggesting a possible effect of weaker repetition suppression. Finally, we establish that our findings generalize to several different classes of stimuli (letters, pictures, faces, words, and pseudo-words), demonstrating a non-specific effect of a lengthened window of integration. We conclude that the visual system in schizophrenics integrates input over longer periods of time, and that repetition suppression may also be deficient. We suggest that these abnormalities in the processing of temporal information may underlie higher-level deficits in schizophrenia and account for the disturbed sense of continuity and fragmentation of events in time reported by patients.


Subject(s)
Pattern Recognition, Visual/physiology , Perceptual Disorders/etiology , Schizophrenia/complications , Adult , Case-Control Studies , Female , Flicker Fusion , Humans , Male , Middle Aged , Perceptual Disorders/diagnosis , Psychophysics , Reaction Time/physiology , Vocabulary
8.
Exp Brain Res ; 193(4): 591-601, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19039579

ABSTRACT

A large body of research indicates a critical role for the left mid-fusiform cortex in reading, however, the extent to which this area is dedicated exclusively to the processing of words and letters has been debated. Two questions regarding left mid-fusiform function are critical to this debate: (1) Are letters stored preferentially compared to visually equivalent non-letters (letter selectivity)? (2) Are letter representations abstract with respect to changes in letter case (e.g., A/a; case invariance)? The present study addressed these questions by comparing priming for letters and pseudoletters in left and right mid-fusiform regions using functional magnetic resonance imaging while subjects performed a same/different matching task. Results revealed priming for letters but not pseudoletters in the left mid-fusiform region, suggesting that representations are letter selective. However, no priming for different-case-primed letters was observed in this region, indicating that representations are not case invariant. In addition, priming for pseudoletters but not letters was observed in the homologous right mid-fusiform region. Overall, findings contradict strongly modular theories of letter/word processing and suggest that left and right mid-fusiform regions support generic object processes that are differentially effective for representing disparate types of visual stimuli.


Subject(s)
Psycholinguistics , Reading , Temporal Lobe/physiology , Analysis of Variance , Female , Humans , Magnetic Resonance Imaging , Male , Occipital Lobe/physiology , Pattern Recognition, Visual/physiology , Photic Stimulation , Reaction Time , Task Performance and Analysis , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...