Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Psychoneuroendocrinology ; 75: 164-172, 2017 01.
Article in English | MEDLINE | ID: mdl-27835807

ABSTRACT

Deficits in cognitive control are a hallmark characteristic of depression, however less is known about the degree to which they persist beyond symptom remission and might contribute to symptom recurrence in remitted individuals (rMDD). Evidence indicates that stress interferes with cognitive control, highlighting a potential mechanism by which stress precipitates depression relapse. Therefore, this study examined whether stress exposure elicits deficits in error monitoring - a component of cognitive control thought to be particularly implicated in the ability to adaptively respond to negative feedback - in individuals with rMDD. Unmedicated individuals with rMDD (n=30) and healthy controls (n=34) performed an Eriksen Flanker task before and 45min after an acute stressor while 128-channel event-related potentials (ERPs) were recorded. Flanker interference effects and post-error adjustments were examined, and ERP analyses focused on the error-related negativity (ERN) and error positivity (Pe). Standardized low resolution electromagnetic tomography (sLORETA) was used to examine stress-induced changes in current source density. Individuals with rMDD showed blunted cortisol reactivity to the stressor, coupled with heightened self-reported stress reactivity. Although no significant effects of group or stress were observed in scalp-level ERPs, source-level analyses indicated that among the rMDD group only, stress caused a reduction in activation in frontocingulate regions critically implicated in error monitoring. The magnitude of stress-induced decreases in frontocingulate activation correlated with heightened self-reported stress reactivity, and also predicted heightened levels of stress and depression 18 months later in the entire sample. These findings suggest that individuals with rMDD show a stress-induced disruption in frontocingulate function that is linked to heightened stress reactivity, and this disruption prospectively predicts heightened levels of future stress and depressive symptomatology.


Subject(s)
Depressive Disorder, Major/physiopathology , Evoked Potentials/physiology , Executive Function/physiology , Frontal Lobe/physiopathology , Gyrus Cinguli/physiopathology , Hydrocortisone/metabolism , Stress, Psychological/physiopathology , Adult , Depressive Disorder, Major/etiology , Depressive Disorder, Major/metabolism , Female , Humans , Male , Recurrence , Stress, Psychological/complications , Stress, Psychological/metabolism , Young Adult
2.
Brain Behav ; 6(9): e00523, 2016 09.
Article in English | MEDLINE | ID: mdl-27688945

ABSTRACT

INTRODUCTION: Studies in laboratory animals and humans indicate that endogenous opioids play an important role in regulating the rewarding value of various drugs, including ethanol (EtOH). Indeed, opioid antagonists are currently a front-line treatment for alcoholism in humans. Although roles for mu- and delta-opioid receptors have been characterized, the contribution of kappa-opioid receptors (KORs) is less clear. There is evidence that changes in KOR system function can decrease or increase EtOH drinking, depending on test conditions. For example, female mice lacking preprodynorphin - the precursor to the endogenous KOR ligand dynorphin - have reduced EtOH intake. Considering that KORs can regulate dopamine (DA) transmission, we hypothesized that KORs expressed on DA neurons would play a prominent role in EtOH intake in females. METHODS: We used a Cre/loxP recombination strategy to ablate KORs throughout the body or specifically on dopamine uptake transporter (DAT)-expressing neurons to investigate the role of KORs on preference for and intake of EtOH (2-bottle choice), the transition from moderate to excessive EtOH drinking (intermittent EtOH access), and binge EtOH drinking (drinking in the dark [DID]). RESULTS: KOR deletion decreased preference for EtOH, although this effect was less pronounced when EtOH intake increased beyond relatively low levels. DISCUSSION: Our findings indicate that KOR activation increases EtOH drinking via effects mediated, at least in part, by KORs on DA neurons. While the mechanisms of this regulation remain unknown, previous work suggests that alterations in negative reinforcement processes or sensitivity to the sensory properties of EtOH can affect preference and intake.

3.
Parkinsonism Relat Disord ; 33: 27-35, 2016 12.
Article in English | MEDLINE | ID: mdl-27769649

ABSTRACT

Essential tremor (ET) is a common cause of significant disability, but its etiologies and pathogenesis are poorly understood. Research has been hampered by the variable definition of ET and by non-standardized research approaches. The National Institute of Neurological Disorders and Stroke (USA) invited experts in ET and related fields to discuss current knowledge, controversies, and gaps in our understanding of ET and to develop recommendations for future research. Discussion focused on phenomenology and phenotypes, therapies and clinical trials, pathophysiology, pathology, and genetics. Across all areas, the need for collaborative and coordinated research on a multinational level was expressed. Standardized data collection using common data elements for genetic, clinical, neurophysiological, and pathological studies was recommended. Large cohorts of patients should be studied prospectively to collect bio-samples, characterize the natural history of the clinical syndrome including patient-oriented outcomes, investigate potential etiologies of various phenotypes, and identify pathophysiological mechanisms. In particular, cellular and system-level mechanisms of tremor oscillations should be elucidated because they may yield effective therapeutic targets and biomarkers. A neuropathology consortium was recommended to standardize postmortem analysis and further characterize neuropathological observations in the cerebellum and elsewhere. Furthermore, genome-wide association studies on large patient cohorts (>10,000 patients) may allow the identification of common genes contributing to risk, and whole exome or genome sequencing may enable the identification of genetic risk and causal mutations in cohorts and well-characterized families.


Subject(s)
Biomedical Research , Essential Tremor , Knowledge , Animals , Biomedical Research/methods , Biomedical Research/standards , Essential Tremor/genetics , Essential Tremor/pathology , Essential Tremor/physiopathology , Humans
5.
Article in English | MEDLINE | ID: mdl-26858994

ABSTRACT

BACKGROUND: Major depressive disorder (MDD) is a highly recurrent condition, and improving our understanding of the abnormalities that persist in remitted MDD (rMDD) may provide insight into mechanisms that contribute to relapse. MDD has been characterized by reward learning deficits linked to dysfunction in frontostriatal regions. Although initial behavioral evidence of reward learning deficits in rMDD has recently emerged, it is unclear whether these reflect impairments in neural reward processing that persist into remission. METHODS: We examined behavioral reward learning and 128-channel event-related potentials (ERP) during a well-validated probabilistic reward task in 26 rMDD individuals and 34 never-depressed controls. Temporo-spatial principal components analysis (PCA) was used to separate overlapping ERP components, and group differences in neural activity in a priori regions were examined using low-resolution electromagnetic tomography (LORETA). RESULTS: Individuals with rMDD displayed reduced behavioral reward learning, as well as blunted ERP amplitude to reward feedback. Importantly, the reduction in ERP amplitude occurred at a PCA factor that peaked during the time at which phasic reward feedback-related signaling - hypothesized to originate in the striatum and project to the anterior cingulate cortex (ACC) - are thought to modulate scalp-recorded activity. Consistent with this, LORETA analyses revealed reduced activity in the ACC in the rMDD group, and this blunting correlated with poorer reward learning. CONCLUSION: These findings suggest that the reward learning impairment observed in acute MDD persists into full remission and that these impairments may be attributable to abnormalities in the neural processes that support reward feedback monitoring, particularly within the ACC.

6.
PLoS One ; 10(3): e0120693, 2015.
Article in English | MEDLINE | ID: mdl-25822989

ABSTRACT

Both the development and relief of stress-related psychiatric conditions such as major depression (MD) and post-traumatic stress disorder (PTSD) have been linked to neuroplastic changes in the brain. One such change involves the birth of new neurons (neurogenesis), which occurs throughout adulthood within discrete areas of the mammalian brain, including the dorsal hippocampus (HIP). Stress can trigger MD and PTSD in humans, and there is considerable evidence that it can decrease HIP neurogenesis in laboratory animals. In contrast, antidepressant treatments increase HIP neurogenesis, and their efficacy is eliminated by ablation of this process. These findings have led to the working hypothesis that HIP neurogenesis serves as a biomarker of neuroplasticity and stress resistance. Here we report that local alterations in the expression of Sprouty2 (SPRY2), an intracellular inhibitor of growth factor function, produces profound effects on both HIP neurogenesis and behaviors that reflect sensitivity to stressors. Viral vector-mediated disruption of endogenous Sprouty2 function (via a dominant negative construct) within the dorsal HIP of adult rats stimulates neurogenesis and produces signs of stress resilience including enhanced extinction of conditioned fear. Conversely, viral vector-mediated elevation of SPRY2 expression intensifies the behavioral consequences of stress. Studies of these manipulations in HIP primary cultures indicate that SPRY2 negatively regulates fibroblast growth factor-2 (FGF2), which has been previously shown to produce antidepressant- and anxiolytic-like effects via actions in the HIP. Our findings strengthen the relationship between HIP plasticity and stress responsiveness, and identify a specific intracellular pathway that could be targeted to study and treat stress-related disorders.


Subject(s)
Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Stress, Psychological/metabolism , Animals , Depression/metabolism , Depression/physiopathology , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/physiopathology , Fibroblast Growth Factor 2/metabolism , Hippocampus/physiopathology , Male , Neuronal Plasticity/physiology , Neurons/metabolism , Neurons/physiology , Rats , Rats, Sprague-Dawley , Stress Disorders, Post-Traumatic/metabolism , Stress Disorders, Post-Traumatic/physiopathology , Stress, Psychological/physiopathology
7.
PLoS One ; 8(8): e70701, 2013.
Article in English | MEDLINE | ID: mdl-23976952

ABSTRACT

BACKGROUND: Nor-BNI, GNTI and JDTic induce selective κ opioid antagonism that is delayed and extremely prolonged, but some other effects are of rapid onset and brief duration. The transient effects of these compounds differ, suggesting that some of them may be mediated by other targets. RESULTS: In binding assays, the three antagonists showed no detectable affinity (K(i)≥10 µM) for most non-opioid receptors and transporters (26 of 43 tested). There was no non-opioid target for which all three compounds shared detectable affinity, or for which any two shared sub-micromolar affinity. All three compounds showed low nanomolar affinity for κ opioid receptors, with moderate selectivity over µ and δ (3 to 44-fold). Nor-BNI bound weakly to the α(2C)-adrenoceptor (K(i) = 630 nM). GNTI enhanced calcium mobilization by noradrenaline at the α(1A)-adrenoceptor (EC50 = 41 nM), but did not activate the receptor, displace radioligands, or enhance PI hydrolysis. This suggests that it is a functionally-selective allosteric enhancer. GNTI was also a weak M1 receptor antagonist (K(B) = 3.7 µM). JDTic bound to the noradrenaline transporter (K(i) = 54 nM), but only weakly inhibited transport (IC50 = 1.1 µM). JDTic also bound to the opioid-like receptor NOP (K(i) = 12 nM), but gave little antagonism even at 30 µM. All three compounds exhibited rapid permeation and active efflux across Caco-2 cell monolayers. CONCLUSIONS: Across 43 non-opioid CNS targets, only GNTI exhibited a potent functional effect (allosteric enhancement of α(1A)-adrenoceptors). This may contribute to GNTI's severe transient effects. Plasma concentrations of nor-BNI and GNTI may be high enough to affect some peripheral non-opioid targets. Nonetheless, κ opioid antagonism persists for weeks or months after these transient effects dissipate. With an adequate pre-administration interval, our results therefore strengthen the evidence that nor-BNI, GNTI and JDTic are highly selective κ opioid antagonists.


Subject(s)
Guanidines/metabolism , Morphinans/metabolism , Naltrexone/analogs & derivatives , Narcotic Antagonists/metabolism , Piperidines/metabolism , Receptors, Opioid, kappa/metabolism , Tetrahydroisoquinolines/metabolism , Allosteric Regulation , Biological Transport , Caco-2 Cells , Calcium/metabolism , Guanidines/pharmacology , Humans , Kinetics , Morphinans/pharmacology , Naltrexone/metabolism , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Norepinephrine/metabolism , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Piperidines/pharmacology , Protein Binding , Receptors, Adrenergic, alpha/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, mu/metabolism , Tetrahydroisoquinolines/pharmacology
8.
Psychopharmacology (Berl) ; 229(3): 435-52, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23836029

ABSTRACT

RATIONALE: Accumulating evidence indicates that brain kappa-opioid receptors (KORs) and dynorphin, the endogenous ligand that binds at these receptors, are involved in regulating states of motivation and emotion. These findings have stimulated interest in the development of KOR-targeted ligands as therapeutic agents. As one example, it has been suggested that KOR antagonists might have a wide range of indications, including the treatment of depressive, anxiety, and addictive disorders, as well as conditions characterized by co-morbidity of these disorders (e.g., post-traumatic stress disorder) A general effect of reducing the impact of stress may explain how KOR antagonists can have efficacy in such a variety of animal models that would appear to represent different disease states. OBJECTIVE: Here, we review evidence that disruption of KOR function attenuates prominent effects of stress. We will describe behavioral and molecular endpoints including those from studies that characterize the effects of KOR antagonists and KOR ablation on the effects of stress itself, as well as on the effects of exogenously delivered corticotropin-releasing factor, a brain peptide that mediates key effects of stress. CONCLUSION: Collectively, available data suggest that KOR disruption produces anti-stress effects and under some conditions can prevent the development of stress-induced adaptations. As such, KOR antagonists may have unique potential as therapeutic agents for the treatment and even prevention of stress-related psychiatric illness, a therapeutic niche that is currently unfilled.


Subject(s)
Anxiety Disorders/metabolism , Brain/metabolism , Receptors, Opioid, kappa/metabolism , Stress, Psychological/metabolism , Animals , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Anxiety Disorders/drug therapy , Anxiety Disorders/psychology , Behavior, Animal/drug effects , Brain/drug effects , Brain/physiopathology , Corticotropin-Releasing Hormone/metabolism , Dynorphins/genetics , Dynorphins/metabolism , Humans , Ligands , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, kappa/genetics , Stress, Psychological/drug therapy , Stress, Psychological/psychology
9.
Neuropsychopharmacology ; 38(8): 1585-97, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23446450

ABSTRACT

Brain kappa-opioid receptors (KORs) are implicated in states of motivation and emotion. Activation of KORs negatively regulates mesolimbic dopamine (DA) neurons, and KOR agonists produce depressive-like behavioral effects. To further evaluate how KOR function affects behavior, we developed mutant mice in which exon 3 of the KOR gene (Oprk1) was flanked with Cre-lox recombination (loxP) sites. By breeding these mice with lines that express Cre-recombinase (Cre) in early embryogenesis (EIIa-Cre) or only in DA neurons (dopamine transporter (DAT)-Cre), we developed constitutive KOR knockouts (KOR(-/-)) and conditional knockouts that lack KORs in DA-containing neurons (DAT-KOR(lox/lox)). Autoradiography demonstrated complete ablation of KOR binding in the KOR(-/-) mutants, and reduced binding in the DAT-KOR(lox/lox) mutants. Quantitative reverse transcription PCR (qPCR) studies confirmed that KOR mRNA is undetectable in the constitutive mutants and reduced in the midbrain DA systems of the conditional mutants. Behavioral characterization demonstrated that these mutant lines do not differ from controls in metrics, including hearing, vision, weight, and locomotor activity. Whereas KOR(-/-) mice appeared normal in the open field and light/dark box tests, DAT-KOR(lox/lox) mice showed reduced anxiety-like behavior, an effect that is broadly consistent with previously reported effects of KOR antagonists. Sensitization to the locomotor-stimulating effects of cocaine appeared normal in KOR(-/-) mutants, but was exaggerated in DAT-KOR(lox/lox) mutants. Increased sensitivity to cocaine in the DAT-KOR(lox/lox) mutants is consistent with a role for KORs in negative regulation of DA function, whereas the lack of differences in the KOR(-/-) mutants suggests compensatory adaptations after constitutive receptor ablation. These mouse lines may be useful in future studies of KOR function.


Subject(s)
Anti-Anxiety Agents/metabolism , Brain/metabolism , Cocaine/pharmacology , Dopaminergic Neurons/metabolism , Neuronal Plasticity/physiology , Receptors, Opioid, kappa/deficiency , Animals , Benzeneacetamides/metabolism , Benzeneacetamides/pharmacology , Brain/drug effects , Dopaminergic Neurons/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neuronal Plasticity/drug effects , Protein Binding/physiology , Pyrrolidines/metabolism , Pyrrolidines/pharmacology , Receptors, Opioid, kappa/agonists
10.
Neuropsychopharmacology ; 37(13): 2809-16, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22948977

ABSTRACT

Stress often disrupts behavior and can lead to psychiatric illness. Considerable evidence suggests that corticotropin-releasing factor (CRF) plays an important role in regulating the effects of stress. CRF administration produces stress-like effects in humans and laboratory animals, and CRF levels are elevated in individuals with stress-related illness. Recent work indicates that κ-opioid receptor (KOR) antagonists can block CRF effects, raising the possibility that at least some of the effects of stress are mediated via KORs. Here we examined the effects of CRF on performance in the 5-choice serial reaction time task (5CSRTT), a test used to quantify attention in rodents, as well as functional interactions between CRF and KORs. Male Sprague-Dawley rats were trained in the 5CSRTT and then each was implanted with an intracerebroventricular (ICV) cannula. After recovery and restabilization of performance, they received a single intraperitoneal (IP) injection of vehicle or JDTic (10 mg/kg), a KOR antagonist with long-lasting (>14 days) effects. In subsequent sessions, rats received ICV infusions of CRF (0.25-1.0 µg) or vehicle and were tested 60 min later. CRF dose-dependently disrupted performance as reflected by decreases in correct responding, increases in omission errors, increases in latencies to respond correctly, and increases in time to complete the session. JDTic attenuated each of these CRF-induced deficits while having no effects on its own. The persistent ability of JDTic to disrupt KOR function was confirmed using the tail immersion assay. These findings indicate that KOR antagonists can prevent acute stress-related effects that degrade performance in tasks requiring attention.


Subject(s)
Attention/drug effects , Corticotropin-Releasing Hormone/administration & dosage , Narcotic Antagonists/administration & dosage , Piperidines/administration & dosage , Receptors, Opioid, kappa/antagonists & inhibitors , Tetrahydroisoquinolines/administration & dosage , Animals , Attention/physiology , Corticotropin-Releasing Hormone/antagonists & inhibitors , Injections, Intraventricular , Male , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/physiology
11.
Neuropsychopharmacology ; 37(11): 2467-75, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22739467

ABSTRACT

Major depression is associated with both dysregulated glutamatergic neurotransmission and fewer astrocytes in limbic areas including the prefrontal cortex (PFC). These deficits may be functionally related. Notably, astrocytes regulate glutamate levels by removing glutamate from the synapse via the glutamate transporter (GLT-1). Previously, we demonstrated that central blockade of GLT-1 induces anhedonia and c-Fos expression in the PFC. Given the role of the PFC in regulating mood, we hypothesized that GLT-1 blockade in the PFC alone would be sufficient to induce anhedonia in rats. We microinjected the GLT-1 inhibitor, dihydrokainic acid (DHK), into the PFC and examined the effects on mood using intracranial self-stimulation (ICSS). At lower doses, intra-PFC DHK produced modest increases in ICSS thresholds, reflecting a depressive-like effect. At higher doses, intra-PFC DHK resulted in cessation of responding. We conducted further tests to clarify whether this total cessation of responding was related to an anhedonic state (tested by sucrose intake), a nonspecific result of motor impairment (measured by the tape test), or seizure activity (measured with electroencephalogram (EEG)). The highest dose of DHK increased latency to begin drinking without altering total sucrose intake. Furthermore, neither motor impairment nor evidence of seizure activity was observed in the tape test or EEG recordings. A decrease in reward value followed by complete cessation of ICSS responding suggests an anhedonic-like effect of intra-PFC DHK; a conclusion that was substantiated by an increased latency to begin sucrose drinking. Overall, these results suggest that blockade of astrocytic glutamate uptake in the PFC is sufficient to produce anhedonia, a core symptom of depression.


Subject(s)
Anhedonia/physiology , Astrocytes/metabolism , Glutamic Acid/metabolism , Prefrontal Cortex/cytology , Analysis of Variance , Anhedonia/drug effects , Animals , Astrocytes/drug effects , Brain Waves/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Electric Stimulation , Electroencephalography , Electromyography , Excitatory Amino Acid Agonists/pharmacology , Food Preferences/drug effects , Kainic Acid/analogs & derivatives , Kainic Acid/pharmacology , Male , Prefrontal Cortex/drug effects , Psychomotor Performance/drug effects , Rats , Rats, Sprague-Dawley , Self Stimulation , Sucrose/administration & dosage
12.
BMC Pharmacol ; 12: 5, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22642416

ABSTRACT

BACKGROUND: Nor-BNI, GNTI and JDTic induce κ opioid antagonism that is delayed by hours and can persist for months. Other effects are transient. It has been proposed that these drugs may be slowly absorbed or distributed, and may dissolve in cell membranes, thus slowing elimination and prolonging their effects. Recent evidence suggests, instead, that they induce prolonged desensitization of the κ opioid receptor. METHODS: To evaluate these hypotheses, we measured relevant physicochemical properties of nor-BNI, GNTI and JDTic, and the timecourse of brain and plasma concentrations in mice after intraperitoneal administration (using LC-MS-MS). RESULTS: In each case, plasma levels were maximal within 30 min and declined by >80% within four hours, correlating well with previously reported transient effects. A strong negative correlation was observed between plasma levels and the delayed, prolonged timecourse of κ antagonism. Brain levels of nor-BNI and JDTic peaked within 30 min, but while nor-BNI was largely eliminated within hours, JDTic declined gradually over a week. Brain uptake of GNTI was too low to measure accurately, and higher doses proved lethal. None of the drugs were highly lipophilic, showing high water solubility (> 45 mM) and low distribution into octanol (log D7.4 < 2). Brain homogenate binding was within the range of many shorter-acting drugs (>7% unbound). JDTic showed P-gp-mediated efflux; nor- BNI and GNTI did not, but their low unbound brain uptake suggests efflux by another mechanism. CONCLUSIONS: The negative plasma concentration-effect relationship we observed is difficult to reconcile with simple competitive antagonism, but is consistent with desensitization. The very slow elimination of JDTic from brain is surprising given that it undergoes active efflux, has modest affinity for homogenate, and has a shorter duration of action than nor-BNI under these conditions. We propose that this persistence may result from entrapment in cellular compartments such as lysosomes.


Subject(s)
Guanidines/pharmacokinetics , Morphinans/pharmacokinetics , Naltrexone/analogs & derivatives , Narcotic Antagonists/pharmacokinetics , Piperidines/pharmacokinetics , Receptors, Opioid, kappa/metabolism , Tetrahydroisoquinolines/pharmacokinetics , Animals , Biological Transport , Brain/metabolism , Guanidines/blood , Hydrophobic and Hydrophilic Interactions , Injections, Intraperitoneal , LLC-PK1 Cells , Male , Mice , Morphinans/blood , Naltrexone/blood , Naltrexone/pharmacokinetics , Narcotic Antagonists/blood , Permeability , Piperidines/blood , Swine , Tetrahydroisoquinolines/blood
13.
Neuron ; 71(3): 383-5, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21835335

ABSTRACT

In this issue, Bruchas et al. (2011) uncover a novel stress-induced p38α MAPK signaling cascade within serotonergic neurons of the dorsal raphe nucleus that mediates depressive and drug-seeking behaviors. Their findings have potentially important implications for medication development.

14.
J Neurosci ; 31(8): 3095-103, 2011 Feb 23.
Article in English | MEDLINE | ID: mdl-21414930

ABSTRACT

Stress triggers psychiatric conditions including depressive and anxiety disorders. The mechanisms by which stress produces persistent changes in behavior are not fully understood. Here we show in rats that stress (footshock) activates the transcription factor cAMP response element binding protein (CREB) within the nucleus accumbens shell (NAS), a brain area involved in encoding reward and aversion. To examine the behavioral significance of altered CREB function in the NAS, we used viral vectors to elevate or disrupt CREB in this region. Elevated CREB produced increases in intracranial self-stimulation thresholds, a depressive-like sign reflecting anhedonia (decreased sensitivity to reward), whereas disruption of CREB function by expression of a dominant-negative CREB had the opposite effect. To determine whether neuroadaptations that produce anhedonia subsequently affect vulnerability to stress-induced behavioral adaptations, we subjected rats with altered CREB function in the NAS to fear conditioning. Although neither elevation nor disruption of CREB function altered the development of conditioned fear, elevation of CREB impaired extinction of conditioned fear. To mimic downstream effects of CREB activation on expression of the opioid peptide dynorphin, we microinjected the κ-opioid receptor (KOR) agonist U50,488 directly into the NAS. KOR stimulation produced anhedonia but had no effect on expression or extinction of conditioned fear. These findings demonstrate that activation of CREB in the NAS produces multiple behavioral signs (anhedonia, impaired extinction) characteristic of experience-dependent psychiatric conditions such as posttraumatic stress disorder. Although CREB activation is a common trigger, expression of these individual signs appears to involve divergent downstream mechanisms.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Extinction, Psychological/physiology , Fear/physiology , Nucleus Accumbens/metabolism , Reward , Stress, Psychological/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/physiology , Male , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiopathology , Rats , Rats, Sprague-Dawley , Stress Disorders, Post-Traumatic/genetics , Stress Disorders, Post-Traumatic/metabolism , Stress Disorders, Post-Traumatic/physiopathology , Stress, Psychological/genetics , Stress, Psychological/physiopathology
15.
Psychopharmacology (Berl) ; 213(4): 697-706, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20922362

ABSTRACT

RATIONALE: Anxiety disorders affect 18% of the United States adult population annually. Recent surges in the diagnosis of posttraumatic stress disorder (PTSD) from combat-exposed veterans have prompted an urgent need to understand the pathophysiology underlying this debilitating condition. OBJECTIVES: Anxiety and fear responses are partly modulated by gamma aminobutyric acid type A (GABA(A)) receptor-mediated synaptic inhibition; benzodiazepines potentiate GABAergic inhibition and are effective anxiolytics. Many genetically modified mouse lines are generated and/or maintained on the C57BL/6J background, a strain where manipulation of anxiety-like behavior using benzodiazepines is difficult. Fear-potentiated startle (FPS), a test of conditioned fear, is a useful preclinical tool to study PTSD-like responses but has been difficult to establish in C57BL/6J mice. METHODS: We modified several FPS experimental parameters and developed a paradigm to assess conditioned fear in C57BL/6J mice. The 6-day protocol consisted of three startle Acclimation days, a Pre-Test day followed by Training and Testing for FPS. Subject responses to the effects of three benzodiazepines were also examined. RESULTS: C57BL/6J mice had low levels of unconditioned fear assessed during Pre-Test (15-18%) but showed robust FPS (80-120%) during the Test session. Conditioned fear responses extinguished over repeated test sessions. Administration of the benzodiazepines alprazolam (0.5 and 1 mg/kg, i.p.), chlordiazepoxide (5 and 10 mg/kg, i.p.), and diazepam (1, 2, and 4 mg/kg, i.p.) significantly reduced FPS to Pre-Test levels. CONCLUSIONS: We used a modified and pharmacologically-validated paradigm to assess FPS in mice thereby providing a powerful tool to examine the neurobiology of PTSD in genetic models of anxiety generated on the C57BL/6J background.


Subject(s)
Anti-Anxiety Agents/pharmacology , Benzodiazepines/pharmacology , Fear/drug effects , Reflex, Startle/drug effects , Alprazolam/administration & dosage , Alprazolam/pharmacology , Animals , Anti-Anxiety Agents/administration & dosage , Benzodiazepines/administration & dosage , Chlordiazepoxide/administration & dosage , Chlordiazepoxide/pharmacology , Diazepam/administration & dosage , Diazepam/pharmacology , Dose-Response Relationship, Drug , GABA Modulators/administration & dosage , GABA Modulators/pharmacology , Male , Mice , Mice, Inbred C57BL , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism
16.
Cell ; 137(4): 761-72, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19450521

ABSTRACT

The transient receptor potential channel 5 (TRPC5) is predominantly expressed in the brain where it can form heterotetrameric complexes with TRPC1 and TRPC4 channel subunits. These excitatory, nonselective cationic channels are regulated by G protein, phospholipase C-coupled receptors. Here, we show that TRPC5(-/-) mice exhibit diminished innate fear levels in response to innately aversive stimuli. Moreover, mutant mice exhibited significant reductions in responses mediated by synaptic activation of Group I metabotropic glutamate and cholecystokinin 2 receptors in neurons of the amygdala. Synaptic strength at afferent inputs to the amygdala was diminished in P10-P13 null mice. In contrast, baseline synaptic transmission, membrane excitability, and spike timing-dependent long-term potentiation at cortical and thalamic inputs to the amygdala were largely normal in older null mice. These experiments provide genetic evidence that TRPC5, activated via G protein-coupled neuronal receptors, has an essential function in innate fear.


Subject(s)
Amygdala/physiology , Fear , TRPC Cation Channels/physiology , Animals , Brain , Conditioning, Psychological , Long-Term Potentiation , Male , Mice , Mice, Knockout , Receptors, Metabotropic Glutamate/physiology , Synaptic Transmission , TRPC Cation Channels/genetics
17.
J Neurosci Res ; 87(1): 69-78, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18752299

ABSTRACT

Previous work has indicated that BDNF increases the differentiation of basal forebrain (BF) oligodendrocytes (OLGs) in culture through the mediation of trkB and the MAPK pathway (Du et al. [ 2006a, b] Mol. Cell. Neurosci. 31:366-375; J. Neurosci. Res. 84:1692-1702). In the present work, effects of BDNF on BF OLG progenitor cells (OPCs) were examined. BDNF increased DNA synthesis of OPCs, as assessed by thymidine and bromodeoxyuridine incorporation. Effects of BDNF on DNA synthesis were mediated through the trkB receptor and not the p75 receptor, as shown by inhibitors that block neurotrophin binding to the receptors and by the phosphorylation of trkB. TrkB can activate the mitogen- activated protein kinase (MAPK), phosphatidylinositol-3 kinase (PI3-K), and phospholipase C-gamma (PLC-gamma) pathways. BDNF elicited the phosphorylation of MAPK and Akt, a kinase downstream of PI3K, but not PLC-gamma in OPCs. Through the use of specific inhibitors to the MAPK and PI3-K pathways, it was found that the MAPK pathway was responsible for the effect of BDNF on DNA synthesis. These data indicate that BDNF affects OPC proliferation and development through the mediation of trkB and the MAPK pathway.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Mitogen-Activated Protein Kinase Kinases/metabolism , Oligodendroglia/physiology , Prosencephalon/cytology , Receptor, trkB/metabolism , Stem Cells/drug effects , Animals , Antibodies/pharmacology , Bromodeoxyuridine/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Female , Gangliosides/immunology , Mitogen-Activated Protein Kinase Kinases/immunology , Pregnancy , Rats , Receptor, trkB/immunology , Thymidine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...