Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 156
Filter
1.
J Affect Disord ; 239: 242-246, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30025313

ABSTRACT

BACKGROUND: Previous studies have shown that individuals with mood disorders have a higher prevalence of both hypercortisolemia and insulin resistance. Insulin resistance is posited to contribute to the cognitive deficits observed in individuals who have depression. However, the mechanistic relationship between cortisol and insulin within the central nervous system remains to be further elucidated. This study aimed to evaluate the effects of the antiglucocorticoid agent, mifepristone, on metabolic function and cognitive performance in individuals receiving treatment for depressive disorders who were euthymic at baseline. METHODS: Participants were administered a 600 mg/day dose of mifepristone for 28 days. Oral glucose tolerance tests (OGTTs) and cognitive assessments measuring verbal memory and executive functioning were administered at baseline and after 28 days of treatment. RESULTS: Improvements in attention and verbal learning were associated with reduction of fasting plasma glucose (FPG) in response to mifepristone treatment. LIMITATIONS: Limitations include the open-label design of this study and a small sample size. CONCLUSIONS: The findings from this study suggest that improvement in fasting plasma glucose levels, upon administration of mifepristone, is associated with the improvement in early input of verbal information. Further studies are warranted in order to better evaluate the use of mifepristone or other antiglucocorticoid agents in treatment of mood disorders characterized by metabolic dysfunction.


Subject(s)
Blood Glucose/drug effects , Cognition/drug effects , Depressive Disorder, Major/metabolism , Hormone Antagonists/pharmacology , Mifepristone/pharmacology , Overweight/metabolism , Aged , Attention/drug effects , Blood Glucose/metabolism , Depressive Disorder, Major/complications , Executive Function/drug effects , Female , Glucose Tolerance Test , Humans , Hydrocortisone/metabolism , Male , Memory/drug effects , Middle Aged , Neuropsychological Tests , Overweight/complications , Verbal Learning/drug effects
2.
Brain Struct Funct ; 223(2): 987-999, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29243106

ABSTRACT

The CLARITY technique enables three-dimensional visualization of fluorescent-labeled biomolecules in clarified intact brain samples, affording a unique view of molecular neuroanatomy and neurocircuitry. It is therefore, essential to find the ideal combination for clearing tissue and detecting the fluorescent-labeled signal. This method requires the formation of a formaldehyde-acrylamide fixative-generated hydrogel mesh through which cellular lipid is removed with sodium dodecyl sulfate. Several laboratories have used differential acrylamide and detergent concentrations to achieve better tissue clearing and antibody penetration, but the potential effects upon fluorescent signal retention is largely unknown. In an effort to optimize CLARITY processing procedures we performed quantitative parvalbumin immunofluorescence and lectin-based vasculature staining using either 4 or 8% sodium dodecyl sulfate detergent in combination with different acrylamide formulas in mouse brain slices. Using both confocal and CLARITY-optimized lightsheet microscope-acquired images, we demonstrate that 2% acrylamide monomer combined with 0.0125% bis-acrylamide and cleared with 4% sodium dodecyl sulfate generally provides the most optimal signal visualization amongst various hydrogel monomer concentrations, lipid removal times, and detergent concentrations.


Subject(s)
Acrylamide/metabolism , Brain/anatomy & histology , Fluorescent Antibody Technique/methods , Lectins/metabolism , Animals , Brain/metabolism , Dose-Response Relationship, Drug , Imaging, Three-Dimensional , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Parvalbumins/metabolism , Staining and Labeling/methods , Time Factors
3.
Mol Psychiatry ; 22(4): 527-536, 2017 04.
Article in English | MEDLINE | ID: mdl-27528460

ABSTRACT

The hypothalamic-pituitary-adrenal (HPA) axis has been implicated in the pathophysiology of a variety of mood and cognitive disorders. Neuroendocrine studies have demonstrated HPA axis overactivity in major depression, a relationship of HPA axis activity to cognitive performance and a potential role of HPA axis genetic variation in cognition. The present study investigated the simultaneous roles HPA axis activity, clinical symptomatology and HPA genetic variation play in cognitive performance. Patients with major depression with psychotic major depression (PMD) and with nonpsychotic major depression (NPMD) and healthy controls (HC) were studied. All participants underwent a diagnostic interview and psychiatric ratings, a comprehensive neuropsychological battery, overnight hourly blood sampling for cortisol and genetic assessment. Cognitive performance differed as a function of depression subtype. Across all subjects, cognitive performance was negatively correlated with higher cortisol, and PMD patients had higher cortisol than did NPMDs and HCs. Cortisol, clinical symptoms and variation in genes, NR3C1 (glucocorticoid receptor; GR) and NR3C2 (mineralocorticoid receptor; MR) that encode for GRs and MRs, predicted cognitive performance. Beyond the effects of cortisol, demographics and clinical symptoms, NR3C1 variation predicted attention and working memory, whereas NR3C2 polymorphisms predicted memory performance. These findings parallel the distribution of GR and MR in primate brain and their putative roles in specific cognitive tasks. HPA axis genetic variation and activity were important predictors of cognition across the entire sample of depressed subjects and HR. GR and MR genetic variation predicted unique cognitive functions, beyond the influence of cortisol and clinical symptoms. GR genetic variation was implicated in attention and working memory, whereas MR was implicated in verbal memory.


Subject(s)
Depressive Disorder, Major/metabolism , Hydrocortisone/metabolism , Hydrocortisone/physiology , Receptors, Glucocorticoid/genetics , Adult , Bipolar Disorder/physiopathology , Brain/physiopathology , Cognition/physiology , Cognition Disorders/physiopathology , Depressive Disorder, Major/blood , Depressive Disorder, Major/physiopathology , Female , Genetic Variation/genetics , Humans , Hydrocortisone/blood , Hypothalamo-Hypophyseal System/physiopathology , Male , Middle Aged , Pituitary-Adrenal System/physiopathology , Polymorphism, Single Nucleotide/genetics , Psychotic Disorders/physiopathology , Receptors, Mineralocorticoid/genetics
5.
Transl Psychiatry ; 6: e799, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27138798

ABSTRACT

Few reliable predictors indicate which depressed individuals respond to antidepressants. Several studies suggest that a history of early-life trauma predicts poorer response to antidepressant therapy but results are variable and limited in adults. The major goal of the present study was to evaluate the role of early-life trauma in predicting acute response outcomes to antidepressants in a large sample of well-characterized patients with major depressive disorder (MDD). The international Study to Predict Optimized Treatment for Depression (iSPOT-D) is a randomized clinical trial with enrollment from December 2008 to January 2012 at eight academic and nine private clinical settings in five countries. Patients (n=1008) meeting DSM-IV criteria for MDD and 336 matched healthy controls comprised the study sample. Six participants withdrew due to serious adverse events. Randomization was to 8 weeks of treatment with escitalopram, sertraline or venlafaxine with dosage adjusted by the participant's treating clinician per routine clinical practice. Exposure to 18 types of traumatic events before the age of 18 was assessed using the Early-Life Stress Questionnaire. Impact of early-life stressors-overall trauma 'load' and specific type of abuse-on treatment outcomes measures: response: (⩾50% improvement on the 17-item Hamilton Rating Scale for Depression, HRSD17 or on the 16-item Quick Inventory of Depressive Symptomatology-Self-Rated, QIDS_SR16) and remission (score ⩽7 on the HRSD17 and ⩽5 on the QIDS_SR16). Trauma prevalence in MDD was compared with controls. Depressed participants were significantly more likely to report early-life stress than controls; 62.5% of MDD participants reported more than two traumatic events compared with 28.4% of controls. The higher rate of early-life trauma was most apparent for experiences of interpersonal violation (emotional, sexual and physical abuses). Abuse and notably abuse occurring at ⩽7 years of age predicted poorer outcomes after 8 weeks of antidepressants, across the three treatment arms. In addition, the abuses occurring between ages 4 and 7 years differentially predicted the poorest outcome following the treatment with sertraline. Specific types of early-life trauma, particularly physical, emotional and sexual abuse, especially when occurring at ⩽7 years of age are important moderators of subsequent response to antidepressant therapy for MDD.


Subject(s)
Antidepressive Agents/therapeutic use , Child Abuse/psychology , Depressive Disorder, Major/complications , Depressive Disorder, Major/drug therapy , Stress, Psychological/complications , Stress, Psychological/psychology , Adolescent , Adult , Aged , Child , Depressive Disorder, Major/psychology , Female , Humans , Internationality , Male , Middle Aged , Surveys and Questionnaires , Treatment Outcome , Young Adult
6.
Transl Psychiatry ; 6: e810, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27187232

ABSTRACT

Anhedonia, the reduced ability to experience pleasure in response to otherwise rewarding stimuli, is a core symptom of major depressive disorder (MDD). Although the posterior ventromedial prefrontal cortex (pVMPFC) and its functional connections have been consistently implicated in MDD, their roles in anhedonia remain poorly understood. Furthermore, it is unknown whether anhedonia is primarily associated with intrinsic 'resting-state' pVMPFC functional connectivity or an inability to modulate connectivity in a context-specific manner. To address these gaps, a pVMPFC region of interest was first identified using activation likelihood estimation meta-analysis. pVMPFC connectivity was then examined in relation to anhedonia and general distress symptoms of depression, using both resting-state and task-based functional magnetic resonance imaging involving pleasant music, in current MDD and healthy control groups. In MDD, pVMPFC connectivity was negatively correlated with anhedonia but not general distress during music listening in key reward- and emotion-processing regions, including nucleus accumbens, ventral tegmental area/substantia nigra, orbitofrontal cortex and insula, as well as fronto-temporal regions involved in tracking complex sound sequences, including middle temporal gyrus and inferior frontal gyrus. No such dissociations were observed in the healthy controls, and resting-state pVMPFC connectivity did not dissociate anhedonia from general distress in either group. Our findings demonstrate that anhedonia in MDD is associated with context-specific deficits in pVMPFC connectivity with the mesolimbic reward system when encountering pleasurable stimuli, rather than a static deficit in intrinsic resting-state connectivity. Critically, identification of functional circuits associated with anhedonia better characterizes MDD heterogeneity and may help track of one of its core symptoms.


Subject(s)
Anhedonia , Depressive Disorder, Major/psychology , Prefrontal Cortex/physiopathology , Stress, Psychological/psychology , Adult , Brain/diagnostic imaging , Brain/physiopathology , Case-Control Studies , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/physiopathology , Depressive Disorder, Major/diagnostic imaging , Depressive Disorder, Major/physiopathology , Female , Functional Neuroimaging , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neural Pathways/diagnostic imaging , Neural Pathways/physiopathology , Nucleus Accumbens/diagnostic imaging , Nucleus Accumbens/physiopathology , Prefrontal Cortex/diagnostic imaging , Reward , Stress, Psychological/diagnostic imaging , Stress, Psychological/physiopathology , Substantia Nigra/diagnostic imaging , Substantia Nigra/physiopathology , Temporal Lobe/diagnostic imaging , Temporal Lobe/physiopathology , Ventral Tegmental Area/diagnostic imaging , Ventral Tegmental Area/physiopathology
7.
Mol Psychiatry ; 21(2): 252-60, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25330740

ABSTRACT

Previous studies have shown inconsistent results regarding the actions of antidepressants on glucocorticoid receptor (GR) signalling. To resolve these inconsistencies, we used a lentiviral-based reporter system to directly monitor rat hippocampal GR activity during stress adaptation. Temporal GR activation was induced significantly by acute stress, as demonstrated by an increase in the intra-individual variability of the acute stress group compared with the variability of the non-stress group. However, the increased intra-individual variability was dampened by exposure to chronic stress, which was partly restored by fluoxetine treatment without affecting glucocorticoid secretion. Immobility in the forced-swim test was negatively correlated with the intra-individual variability, but was not correlated with the quantitative GR activity during fluoxetine therapy; this highlights the temporal variability in the neurobiological links between GR signalling and the therapeutic action of fluoxetine. Furthermore, we demonstrated sequential phosphorylation between GR (S224) and (S232) following fluoxetine treatment, showing a molecular basis for hormone-independent nuclear translocation and transcriptional enhancement. Collectively, these results suggest a neurobiological mechanism by which fluoxetine treatment confers resilience to the chronic stress-mediated attenuation of hypothalamic-pituitary-adrenal axis activity.


Subject(s)
Fluoxetine/pharmacology , Receptors, Glucocorticoid/metabolism , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents, Second-Generation/pharmacology , Corticosterone/pharmacology , Hippocampus/metabolism , Hypothalamo-Hypophyseal System/metabolism , Male , Phosphorylation , Pituitary-Adrenal System/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Stress, Psychological
8.
Transl Psychiatry ; 5: e636, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26371764

ABSTRACT

Stress can be a predisposing factor to psychiatric disorders and has been associated with decreased neurogenesis and reduced hippocampal volume especially in depression. Similarly, in white blood cells chronic psychological stress has been associated with telomere shortening and with mood disorders and schizophrenia (SZ). However, in previous post-mortem brain studies from occipital cortex and cerebellum, no difference in telomere length was observed in depression. We hypothesized that in psychiatric disorders, stress-driven accelerated cellular aging can be observed in brain regions particularly sensitive to stress. Telomere length was measured by quantitative-PCR in five brain regions (dorsolateral prefrontal cortex, hippocampus (HIPP), amygdala, nucleus accumbens and substantia nigra (SN)) in major depressive disorder (MDD), bipolar disorder, SZ and normal control subjects (N = 40, 10 subjects per group). We observed significant differences in telomere length across brain regions suggesting variable levels of cell aging, with SN and HIPP having the longest telomeres and the dorsolateral prefrontal cortex the shortest. A significant decrease (P < 0.02) in telomere length was observed specifically in the HIPP of MDD subjects even after controlling for age. In the HIPP of MDD subjects, several genes involved in neuroprotection and in stress response (FKBP5, CRH) showed altered levels of mRNA. Our results suggest the presence of hippocampal stress-mediated accelerated cellular aging in depression. Further studies are needed to investigate the cellular specificity of these findings.


Subject(s)
Depressive Disorder, Major/genetics , Depressive Disorder, Major/pathology , Hippocampus/pathology , Telomere/genetics , Telomere/pathology , Analysis of Variance , Brain/pathology , Cadaver , Dissection , Female , Genetic Techniques , Humans , Male , Middle Aged , Polymerase Chain Reaction
9.
Transl Psychiatry ; 5: e537, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25826112

ABSTRACT

Stress inoculation entails intermittent exposure to mildly stressful situations that present opportunities to learn, practice and improve coping in the context of exposure psychotherapies and resiliency training. Here we investigate behavioral and hormonal aspects of stress inoculation modeled in mice. Mice randomized to stress inoculation or a control treatment condition were assessed for corticosterone stress hormone responses and behavior during open-field, object-exploration and tail-suspension tests. Stress inoculation training sessions that acutely increased plasma levels of corticosterone diminished subsequent immobility as a measure of behavioral despair on tail-suspension tests. Stress inoculation also decreased subsequent freezing in the open field despite comparable levels of thigmotaxis in mice from both treatment conditions. Stress inoculation subsequently decreased novel-object exploration latencies and reduced corticosterone responses to repeated restraint. These results demonstrate that stress inoculation acutely stimulates glucocorticoid signaling and then enhances subsequent indications of active coping behavior in mice. Unlike mouse models that screen for the absence of vulnerability to stress or presence of traits that occur in resilient individuals, stress inoculation training reflects an experience-dependent learning-like process that resembles interventions designed to build resilience in humans. Mouse models of stress inoculation may provide novel insights for new preventive strategies or therapeutic treatments of human psychiatric disorders that are triggered and exacerbated by stressful life events.


Subject(s)
Adaptation, Psychological/physiology , Behavior, Animal/physiology , Corticosterone/blood , Stress, Psychological/blood , Stress, Psychological/physiopathology , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Stress, Psychological/psychology
10.
Pharmacogenomics J ; 15(4): 332-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25487678

ABSTRACT

The hypothesis that allelic variation in the multidrug resistance-1 (MDR1 or ABCB1) gene encoding the P-glycoprotein (P-gp) blood-brain barrier efflux pump is associated with remission and side effects was tested in chronic major depression patients treated with P-gp substrates. In 83 patients from the REVAMP trial, frequency of and time to remission as well as side effects was tested among genotype groups at 6 ABCB1 single nucleotide polymorphisms (SNPs). These six SNPs are significantly associated with remission and time to remission, with minor allele carriers on rs2235040 and rs9282564 attaining statistical significance after controlling for the other ABCB1 SNPs. The six ABCB1 SNPs are also significantly associated with the average side effects. However, here common homozygotes on rs2235040 and rs9282564 demonstrated significantly higher side effects after controlling for the effects of the other ABCB1 SNPs. These findings confirm and extend previous observations that minor alleles of two ABCB1 SNPs predict remission to treatment with substrates and demonstrate that common homozygotes on these SNPs experience greater side effects. Results point to the potential importance of ABCB1 variation for personalized medicine approaches to treating depression.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Antidepressive Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/genetics , ATP Binding Cassette Transporter, Subfamily B/genetics , Adult , Alleles , Antidepressive Agents/administration & dosage , Antidepressive Agents/adverse effects , Chronic Disease , Dose-Response Relationship, Drug , Female , Genotype , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Polymorphism, Single Nucleotide , Sertraline/therapeutic use , Treatment Outcome , White People
11.
Mol Psychiatry ; 20(1): 48-55, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25349171

ABSTRACT

Conventional antidepressants require 2-8 weeks for a full clinical response. In contrast, two rapidly acting antidepressant interventions, low-dose ketamine and sleep deprivation (SD) therapy, act within hours to robustly decrease depressive symptoms in a subgroup of major depressive disorder (MDD) patients. Evidence that MDD may be a circadian-related illness is based, in part, on a large set of clinical data showing that diurnal rhythmicity (sleep, temperature, mood and hormone secretion) is altered during depressive episodes. In a microarray study, we observed widespread changes in cyclic gene expression in six regions of postmortem brain tissue of depressed patients matched with controls for time-of-death (TOD). We screened 12 000 transcripts and observed that the core clock genes, essential for controlling virtually all rhythms in the body, showed robust 24-h sinusoidal expression patterns in six brain regions in control subjects. In MDD patients matched for TOD with controls, the expression patterns of the clock genes in brain were significantly dysregulated. Some of the most robust changes were seen in anterior cingulate (ACC). These findings suggest that in addition to structural abnormalities, lesion studies, and the large body of functional brain imaging studies reporting increased activation in the ACC of depressed patients who respond to a wide range of therapies, there may be a circadian dysregulation in clock gene expression in a subgroup of MDDs. Here, we review human, animal and neuronal cell culture data suggesting that both low-dose ketamine and SD can modulate circadian rhythms. We hypothesize that the rapid antidepressant actions of ketamine and SD may act, in part, to reset abnormal clock genes in MDD to restore and stabilize circadian rhythmicity. Conversely, clinical relapse may reflect a desynchronization of the clock, indicative of a reactivation of abnormal clock gene function. Future work could involve identifying specific small molecules capable of resetting and stabilizing clock genes to evaluate if they can rapidly relieve symptoms and sustain improvement.


Subject(s)
Antidepressive Agents/therapeutic use , CLOCK Proteins/genetics , Chronobiology Disorders/complications , Chronobiology Disorders/genetics , Depressive Disorder, Major , Animals , Depressive Disorder, Major/etiology , Depressive Disorder, Major/genetics , Depressive Disorder, Major/therapy , Excitatory Amino Acid Antagonists/therapeutic use , Gyrus Cinguli/metabolism , Humans , Ketamine/therapeutic use , Sleep Deprivation
12.
Mol Psychiatry ; 19(2): 220-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24166410

ABSTRACT

Genetic variation underlying hypothalamic pituitary adrenal (HPA) axis overactivity in healthy controls (HCs) and patients with severe forms of major depression has not been well explored, but could explain risk for cortisol dysregulation. In total, 95 participants were studied: 40 patients with psychotic major depression (PMD); 26 patients with non-psychotic major depression (NPMD); and 29 HCs. Collection of genetic material was added one third of the way into a larger study on cortisol, cognition and psychosis in major depression. Subjects were assessed using the Brief Psychiatric Rating Scale, the Hamilton Depression Rating Scale and the Structured Clinical Interview for Diagnostic and Statistical Manual of Mental Disorders. Blood was collected hourly for determination of cortisol from 1800 to 0900 h and for the assessment of alleles for six genes involved in HPA axis regulation. Two of the six genes contributed significantly to cortisol levels, psychosis measures or depression severity. After accounting for age, depression and psychosis, and medication status, only allelic variation for the glucocorticoid receptor (GR) gene accounted for a significant variance for mean cortisol levels from 1800 to 0100 h (r(2)=0.288) and from 0100 to 0900 h (r(2)=0.171). In addition, GR and corticotropin-releasing hormone receptor 1 (CRHR1) genotypes contributed significantly to psychosis measures and CRHR1 contributed significantly to depression severity rating.


Subject(s)
Affective Disorders, Psychotic/genetics , Depressive Disorder, Major/genetics , Hydrocortisone/blood , Hypothalamo-Hypophyseal System/physiopathology , Pituitary-Adrenal System/physiopathology , Adult , Affective Disorders, Psychotic/diagnosis , Affective Disorders, Psychotic/physiopathology , Corticotropin-Releasing Hormone/genetics , Depressive Disorder, Major/diagnosis , Depressive Disorder, Major/physiopathology , Female , Humans , Interview, Psychological , Linkage Disequilibrium , Male , Psychiatric Status Rating Scales , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Glucocorticoid/genetics , Receptors, Mineralocorticoid/genetics , Tacrolimus Binding Proteins/genetics
13.
Psychol Med ; 41(10): 2089-97, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21349239

ABSTRACT

BACKGROUND: Selective serotonin reuptake inhibitors take several weeks to achieve their full antidepressant effects. Post-synaptic 5-HT2A receptor activation is thought to be involved in this delayed therapeutic effect. Pipamperone acts as a highly selective 5-HT2A/D4 antagonist when administered in low doses. The purpose of this study was to compare citalopram 40 mg once daily plus pipamperone 5 mg twice daily (PipCit) versus citalopram plus placebo twice daily for magnitude and onset of therapeutic effect. METHOD: An 8-week, randomized, double-blind study in patients with major depressive disorder was carried out. RESULTS: The study population comprised 165 patients (citalopram and placebo, n=82; PipCit, n=83) with a mean baseline Montgomery-Asberg Depression Rating Scale (MADRS) score of 32.6 (s.d.=5.5). In the first 4 weeks, more citalopram and placebo than PipCit patients discontinued treatment (18% v. 4%, respectively, p=0.003). PipCit patients had significantly greater improvement in MADRS score at week 1 [observed cases (OC), p=0.021; last observation carried forward (LOCF), p=0.007] and week 4 (LOCF, p=0.025) but not at week 8 compared with citalopram and placebo patients. Significant differences in MADRS scores favoured PipCit in reduced sleep, reduced appetite, concentration difficulties and pessimistic thoughts. Mean Clinical Global Impression-Improvement scores were significantly improved after 1 week of PipCit compared with citalopram and placebo (OC and LOCF, p=0.002). CONCLUSIONS: Although the MADRS score from baseline to 8 weeks did not differ between groups, PipCit provided superior antidepressant effects and fewer discontinuations compared with citalopram and placebo during the first 4 weeks of treatment, especially in the first week.


Subject(s)
Butyrophenones/therapeutic use , Citalopram/therapeutic use , Depressive Disorder, Major/drug therapy , Selective Serotonin Reuptake Inhibitors/therapeutic use , Serotonin Antagonists/therapeutic use , Adolescent , Adult , Aged , Butyrophenones/administration & dosage , Butyrophenones/adverse effects , Butyrophenones/standards , Citalopram/administration & dosage , Citalopram/adverse effects , Citalopram/standards , Depressive Disorder, Major/diagnosis , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Placebos , Psychiatric Status Rating Scales , Scotland , Serotonin Antagonists/administration & dosage , Serotonin Antagonists/adverse effects , Serotonin Antagonists/standards , Selective Serotonin Reuptake Inhibitors/administration & dosage , Selective Serotonin Reuptake Inhibitors/adverse effects , Selective Serotonin Reuptake Inhibitors/standards , Treatment Outcome , Young Adult
14.
Mol Psychiatry ; 16(6): 634-46, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20386568

ABSTRACT

Several studies have proposed that brain glutamate signaling abnormalities and glial pathology have a role in the etiology of major depressive disorder (MDD). These conclusions were primarily drawn from post-mortem studies in which forebrain brain regions were examined. The locus coeruleus (LC) is the primary source of extensive noradrenergic innervation of the forebrain and as such exerts a powerful regulatory role over cognitive and affective functions, which are dysregulated in MDD. Furthermore, altered noradrenergic neurotransmission is associated with depressive symptoms and is thought to have a role in the pathophysiology of MDD. In the present study we used laser-capture microdissection (LCM) to selectively harvest LC tissue from post-mortem brains of MDD patients, patients with bipolar disorder (BPD) and from psychiatrically normal subjects. Using microarray technology we examined global patterns of gene expression. Differential mRNA expression of select candidate genes was then interrogated using quantitative real-time PCR (qPCR) and in situ hybridization (ISH). Our findings reveal multiple signaling pathway alterations in the LC of MDD but not BPD subjects. These include glutamate signaling genes, SLC1A2, SLC1A3 and GLUL, growth factor genes FGFR3 and TrkB, and several genes exclusively expressed in astroglia. Our data extend previous findings of altered glutamate, astroglial and growth factor functions in MDD for the first time to the brainstem. These findings indicate that such alterations: (1) are unique to MDD and distinguishable from BPD, and (2) affect multiple brain regions, suggesting a whole-brain dysregulation of such functions.


Subject(s)
Depressive Disorder, Major/pathology , Gene Expression Regulation , Glutamic Acid/metabolism , Locus Coeruleus/metabolism , Neuroglia/metabolism , Signal Transduction/physiology , Adolescent , Adult , Aged , Female , Gene Expression Profiling/methods , Glutamate Plasma Membrane Transport Proteins , Glutamic Acid/genetics , Humans , Intercellular Signaling Peptides and Proteins , Locus Coeruleus/pathology , Male , Microdissection , Middle Aged , Models, Biological , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis/methods , RNA, Messenger/metabolism , Young Adult
16.
Mol Psychiatry ; 12(12): 1089-102, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17893703

ABSTRACT

Stressful experiences that consistently increase cortisol levels appear to alter the expression of hundreds of genes in prefrontal limbic brain regions. Here, we investigate this hypothesis in monkeys exposed to intermittent social stress-induced episodes of hypercortisolism or a no-stress control condition. Prefrontal profiles of gene expression compiled from Affymetrix microarray data for monkeys randomized to the no-stress condition were consistent with microarray results published for healthy humans. In monkeys exposed to intermittent social stress, more genes than expected by chance appeared to be differentially expressed in ventromedial prefrontal cortex compared to monkeys not exposed to adult social stress. Most of these stress responsive candidate genes were modestly downregulated, including ubiquitin conjugation enzymes and ligases involved in synaptic plasticity, cell cycle progression and nuclear receptor signaling. Social stress did not affect gene expression beyond that expected by chance in dorsolateral prefrontal cortex or prefrontal white matter. Thirty four of 48 comparisons chosen for verification by quantitative real-time polymerase chain reaction (qPCR) were consistent with the microarray-predicted result. Furthermore, qPCR and microarray data were highly correlated. These results provide new insights on the regulation of gene expression in a prefrontal corticolimbic region involved in the pathophysiology of stress and major depression. Comparisons between these data from monkeys and those for ventromedial prefrontal cortex in humans with a history of major depression may help to distinguish the molecular signature of stress from other confounding factors in human postmortem brain research.


Subject(s)
Gene Expression Regulation/physiology , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Stress, Physiological/pathology , Animals , Gene Expression/physiology , Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , Primates/anatomy & histology , Stress, Physiological/genetics , Stress, Physiological/physiopathology
17.
Mol Psychiatry ; 12(6): 544-55, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17353910

ABSTRACT

The s allele variant of the serotonin transporter gene (5-HTT) has recently been observed to moderate the relationship of stress to depression and anxiety. To date no study has considered interactive effects of 5-HTT genotype, stress and hypothalamic-pituitary-adrenal (HPA) function on cognition in healthy, older adults, which may reflect developmental, functional or neurodegenerative effects of the serotonin transporter polymorphism. We investigated whether 5-HTT genotype interacts with cumulative life stress and HPA-axis measures of waking and diurnal cortisol slope to impact cognition in 154 non-depressed, older adults. Structural images of hippocampal volume were acquired on a subsample of 56 participants. The 5-HTT s allele was associated with both significantly lower delayed recall and higher waking cortisol levels. Presence of the s allele interacted with higher waking cortisol to negatively impact memory. We also observed a significant interaction of higher waking cortisol and the s allele on lower hippocampal volume. Smaller hippocampi and higher cortisol were associated with lower delayed recall only in s allele carriers. No impact or interactions of cumulative life stress with 5-HTT or cortisol were observed. This is the first investigation to identify an association of the 5-HTT s allele with poorer memory function in older adults. The interactive effects of the s allele and waking cortisol levels on reduced hippocampal volume and lower memory suggest that the negative effect of the serotonin polymorphism on memory is mediated by the HPA axis. Further, given the significant association of the s allele with higher waking cortisol in our investigation, future studies may be needed to evaluate the impact of the serotonin transporter polymorphism on any neuropsychiatric or behavioral outcome which is influenced by HPA axis function in older adults.


Subject(s)
Aging/genetics , Cognition/physiology , Hippocampus/anatomy & histology , Hydrocortisone/blood , Memory/physiology , Serotonin Plasma Membrane Transport Proteins/genetics , Aged , Aged, 80 and over , Aging/metabolism , Female , Hippocampus/metabolism , Humans , Hypothalamo-Hypophyseal System/metabolism , Male , Middle Aged , Organ Size , Pituitary-Adrenal System/metabolism , Polymorphism, Genetic , Reference Values , Regression Analysis , Serotonin Plasma Membrane Transport Proteins/metabolism , Stress, Psychological/genetics , Stress, Psychological/metabolism
18.
Neurology ; 65(4): 642-4, 2005 Aug 23.
Article in English | MEDLINE | ID: mdl-16116137

ABSTRACT

The authors investigated the relationship between obstructive sleep apnea/hypopnea (OSAH) and cognition in 36 older adults, 18 APOE epsilon4 carriers, and 18 non-carriers. Greater numbers of respiratory events negatively impacted memory function in epsilon4 carriers only. This is the first study to provide preliminary evidence for a negative interaction of APOE epsilon4 and OSAH on memory in older adults, which may have important implications for treating cognitive decline and delaying dementia onset.


Subject(s)
Apolipoproteins E/genetics , Genetic Predisposition to Disease/genetics , Heterozygote , Memory Disorders/genetics , Sleep Apnea Syndromes/complications , Sleep Apnea Syndromes/genetics , Aged , Apolipoprotein E4 , Brain/metabolism , Brain/physiopathology , Cognition Disorders/genetics , Cognition Disorders/physiopathology , DNA Mutational Analysis , Disease Progression , Female , Genetic Testing , Genetic Variation/genetics , Humans , Male , Memory Disorders/physiopathology , Memory Disorders/psychology , Middle Aged , Neuropsychological Tests , Oxidative Stress/genetics , Sleep Apnea Syndromes/physiopathology , Sleep Stages/genetics
19.
Neuroimage ; 20(1): 257-64, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14527586

ABSTRACT

As a step toward bridging the gap between human and animal studies of olfactory brain systems, we report results from an fMRI study of olfaction in squirrel monkeys. High-resolution fMRI images at 3 T with 1.25 x 1.25 x 1.2 mm(3) voxels were obtained covering the whole brain using an 8-cm-diameter birdcage coil and a gradient-echo spiral pulse sequence. Data were acquired from six sedated adult males using a standard block design. All fMRI data were spatially normalized to a common template and analyzed at the individual and group levels with statistical parametric and nonparametric methods. Robust odorant-induced activations were detected in several brain regions previously implicated in conscious human olfactory processing, including the orbitofrontal cortex, cerebellum, and piriform cortex. Consistent with human data, no stimulus intensity effects were observed in any of these regions. Average signal changes in these regions exceeded 0.6%, more than three times the expected signal change based on human fMRI studies of olfaction adjusting for differences in voxel size. These results demonstrate the feasibility of studying olfaction in sedated monkeys with imaging techniques commonly used at 3 T in humans and help promote direct comparisons between humans and nonhuman primates. Our findings, for example, provide novel support for the hypothesis that the cerebellum is involved in sensory acquisition. More broadly, this study suggests that olfactory processing in sedated monkeys and nonsedated humans shares similar neural substrates both within and beyond the primary olfactory system.


Subject(s)
Brain/physiology , Smell/physiology , Animals , Conscious Sedation , Fourier Analysis , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Odorants , Pilot Projects , Saimiri
20.
Arch Gen Psychiatry ; 58(12): 1145-51, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11735843

ABSTRACT

BACKGROUND: Opportunities for research on the causes and consequences of stress-related hippocampal atrophy are limited in human psychiatric disorders. Therefore, this longitudinal study investigated early life stress and inherited variation in monkey hippocampal volumes. METHODS: Paternal half-siblings raised apart from one another by different mothers in the absence of fathers were randomized to 1 of 3 postnatal conditions that disrupted diverse aspects of early maternal care (n = 13 monkeys per condition). These conditions were previously shown to produce differences in social behavior, emotional reactivity, and neuroendocrine stress physiology. Hippocampal volumes were subsequently determined in adulthood by high-resolution magnetic resonance imaging. RESULTS: Adult hippocampal volumes did not differ with respect to the stressful postnatal conditions. Based on paternal half-sibling effects, the estimated proportion of genetic variance, ie, heritability, was 54% for hippocampal size. Paternal half-siblings with small adult hippocampal volumes responded to the removal of all mothers after weaning with initially larger relative increases in cortisol levels. Plasma cortisol levels 3 and 7 days later, and measures of cortisol-negative feedback in adulthood were not, however, correlated with hippocampal size. CONCLUSIONS: In humans with mood and anxiety disorders, small hippocampal volumes have been taken as evidence that excessive stress levels of cortisol induce hippocampal volume loss. Results from this study of monkeys suggest that small hippocampi also reflect an inherited characteristic of the brain. Genetically informed clinical studies should assess whether inherited variation in hippocampal morphology contributes to excessive stress levels of cortisol through diminished neuroendocrine regulation.


Subject(s)
Arousal/genetics , Genetic Variation , Hippocampus/pathology , Maternal Deprivation , Stress, Psychological/complications , Animals , Arousal/physiology , Atrophy , Female , Hydrocortisone/blood , Magnetic Resonance Imaging , Male , Organ Size/genetics , Phenotype , Saimiri
SELECTION OF CITATIONS
SEARCH DETAIL
...