Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Mol Psychiatry ; 21(2): 243-51, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26283642

RESUMO

Although evidence shows depressed moods enhance risk for somatic diseases, molecular mechanisms underlying enhanced somatic susceptibility are ill-defined. Knowledge of these molecular mechanisms will inform development of treatment and prevention strategies across comorbid depressive and somatic illnesses. Existing evidence suggests that interleukin-18 (IL-18; an IL-1 family cytokine) is elevated in depression and implicated in pathophysiology underlying comorbid medical illnesses. We previously identified strong associations between baseline IL-18 and µ-opioid receptor availability in major depressive disorder (MDD) volunteers. Combined with the evidence in animal models, we hypothesized that experimental mood induction would change IL-18, the extent proportional to opioid neurotransmitter release. Using the Velten technique in a [(11)C]carfentanil positron emission tomography neuroimaging study, we examined the impact of experimentally induced mood (sad, neutral) on plasma IL-18 and relationships with concurrent changes in the central opioid neurotransmission in 28 volunteers (healthy, MDD). Results showed mood induction impacted IL-18 (F2,25=12.2, P<0.001), sadness increasing IL-18 (T27=2.6, P=0.01) and neutral mood reducing IL-18 (T27=-4.1, P<0.001). In depressed volunteers, changes in IL-18 were more pronounced (F2,25=3.6, P=0.03) and linearly proportional to sadness-induced µ-opioid activation (left ventral pallidum, bilateral anterior cingulate cortices, right hypothalamus and bilateral amygdala). These data demonstrate that dynamic changes of a pro-inflammatory IL-1 superfamily cytokine, IL-18, and its relationship to µ-opioid neurotransmission in response to experimentally induced sadness. Further testing is warranted to delineate the role of neuroimmune interactions involving IL-18 in enhancing susceptibility to medical illness (that is, diabetes, heart disease and persistent pain states) in depressed individuals.


Assuntos
Interleucina-18/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Receptores Opioides mu/metabolismo , Adulto , Afeto/fisiologia , Tonsila do Cerebelo/metabolismo , Encéfalo/fisiopatologia , Transtorno Depressivo Maior/metabolismo , Emoções , Feminino , Giro do Cíngulo/metabolismo , Humanos , Fatores Imunológicos , Dor/fisiopatologia , Medição da Dor , Transmissão Sináptica/fisiologia
2.
Psychol Med ; 46(5): 1055-67, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26784396

RESUMO

BACKGROUND: Recent meta-analyses of resting-state networks in major depressive disorder (MDD) implicate network disruptions underlying cognitive and affective features of illness. Heterogeneity of findings to date may stem from the relative lack of data parsing clinical features of MDD such as phase of illness and the burden of multiple episodes. METHOD: Resting-state functional magnetic resonance imaging data were collected from 17 active MDD and 34 remitted MDD patients, and 26 healthy controls (HCs) across two sites. Participants were medication-free and further subdivided into those with single v. multiple episodes to examine disease burden. Seed-based connectivity using the posterior cingulate cortex (PCC) seed to probe the default mode network as well as the amygdala and subgenual anterior cingulate cortex (sgACC) seeds to probe the salience network (SN) were conducted. RESULTS: Young adults with remitted MDD demonstrated hyperconnectivity of the left PCC to the left inferior frontal gyrus and of the left sgACC to the right ventromedial prefrontal cortex (PFC) and left hippocampus compared with HCs. Episode-independent effects were observed between the left PCC and the right dorsolateral PFC, as well as between the left amygdala and right insula and caudate, whereas the burden of multiple episodes was associated with hypoconnectivity of the left PCC to multiple cognitive control regions as well as hypoconnectivity of the amygdala to large portions of the SN. CONCLUSIONS: This is the first study of a homogeneous sample of unmedicated young adults with a history of adolescent-onset MDD illustrating brain-based episodic features of illness.


Assuntos
Tonsila do Cerebelo/fisiopatologia , Transtorno Depressivo Maior/diagnóstico por imagem , Transtorno Depressivo Maior/fisiopatologia , Vias Neurais/fisiopatologia , Adolescente , Adulto , Mapeamento Encefálico/métodos , Estudos de Casos e Controles , Função Executiva , Feminino , Lobo Frontal/fisiopatologia , Giro do Cíngulo/fisiopatologia , Humanos , Imageamento por Ressonância Magnética , Masculino , Recidiva , Adulto Jovem
3.
Mol Psychiatry ; 20(4): 416-23, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25510510

RESUMO

Endogenous opioid and non-opioid mechanisms (for example, dopamine (DA), endocannabinoids (eCB)) have been implicated in the formation of placebo analgesic effects, with initial reports dating back three decades. Besides the perspective that placebo effects confound randomized clinical trials, the information so far acquired points to neurobiological systems that when activated by positive expectations and maintained through conditioning and reward learning are capable of inducing physiological changes that lead to the experience of analgesia and improvements in emotional state. Molecular neuroimaging techniques with positron emission tomography and the selective µ-opioid and D2/3 radiotracers [(11)C]carfentanil and [(11)C]raclopride have significantly contributed to our understanding of the neurobiological systems involved in the formation of placebo effects. This line of research has described neural and neurotransmitter networks implicated in placebo responses and provided the technical tools to examine inter-individual differences in the function of placebo-responsive mechanisms, and potential surrogates (biomarkers). As a consequence, the formation of biological placebo effects is now being linked to the concept of resiliency mechanisms, partially determined by genetic factors, and uncovered by the cognitive emotional integration of the expectations created by the therapeutic environment and its maintenance through learning mechanisms. Further work needs to extend this research into clinical conditions where the rates of placebo responses are high and its neurobiological mechanisms have been largely unexplored (for example, mood and anxiety disorders, persistent pain syndromes or even Parkinson disease and multiple sclerosis). The delineation of these processes within and across diseases would point to biological targets that have not been contemplated in traditional drug development.


Assuntos
Analgésicos Opioides/metabolismo , Encéfalo/fisiologia , Neuroimagem , Neurotransmissores/genética , Efeito Placebo , Encéfalo/efeitos dos fármacos , Humanos , Neurotransmissores/metabolismo , Personalidade
4.
Mol Psychiatry ; 20(2): 193-200, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25600108

RESUMO

The µ-opioid receptor (MOR) system, well known for dampening physical pain, is also hypothesized to dampen 'social pain.' We used positron emission tomography scanning with the selective MOR radioligand [(11)C]carfentanil to test the hypothesis that MOR system activation (reflecting endogenous opioid release) in response to social rejection and acceptance is altered in medication-free patients diagnosed with current major depressive disorder (MDD, n=17) compared with healthy controls (HCs, n=18). During rejection, MDD patients showed reduced endogenous opioid release in brain regions regulating stress, mood and motivation, and slower emotional recovery compared with HCs. During acceptance, only HCs showed increased social motivation, which was positively correlated with endogenous opioid release in the nucleus accumbens, a reward structure. Altered endogenous opioid activity in MDD may hinder emotional recovery from negative social interactions and decrease pleasure derived from positive interactions. Both effects may reinforce depression, trigger relapse and contribute to poor treatment outcomes.


Assuntos
Encéfalo/metabolismo , Transtorno Depressivo Maior/patologia , Transtorno Depressivo Maior/psicologia , Distância Psicológica , Receptores Opioides mu/metabolismo , Facilitação Social , Adulto , Analgésicos Opioides/farmacocinética , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Radioisótopos de Carbono/farmacocinética , Emoções , Retroalimentação , Feminino , Fentanila/análogos & derivados , Fentanila/farmacocinética , Humanos , Hidrocortisona/sangue , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Masculino , Tomografia por Emissão de Pósitrons , Ligação Proteica/efeitos dos fármacos , Escalas de Graduação Psiquiátrica , Radiografia , Adulto Jovem
5.
Mol Psychiatry ; 19(3): 385-91, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24042479

RESUMO

Endogenous opioid and cannabinoid systems are thought to act synergistically regulating antinociceptive and reward mechanisms. To further understand the human implications of the interaction between these two systems, we investigated the role of the common, functional missense variant Pro129Thr of the gene coding fatty acid amide hydrolase (FAAH), the major degrading enzyme of endocannabinoids, on psychophysical and neurotransmitter (dopaminergic, opioid) responses to pain and placebo-induced analgesia in humans. FAAH Pro129/Pro129 homozygotes, who constitute nearly half of the population, reported higher placebo analgesia and more positive affective states immediately and 24 h after placebo administration; no effects on pain report in the absence of placebo were observed. Pro129/Pro129 homozygotes also showed greater placebo-induced µ-opioid, but not D(2/3) dopaminergic, enhancements in neurotransmission in regions known involved in placebo effects. These results show that a common genetic variation affecting the function of the cannabinoid system is serving as a probe to demonstrate the involvement of cannabinoid and opioid transmitters on the formation of placebo effects.


Assuntos
Amidoidrolases/genética , Encéfalo/metabolismo , Efeito Placebo , Receptores de Dopamina D2/metabolismo , Receptores Opioides mu/metabolismo , Adulto , Afeto , Encéfalo/diagnóstico por imagem , Feminino , Neuroimagem Funcional , Homozigoto , Humanos , Masculino , Mutação de Sentido Incorreto/genética , Medição da Dor , Tomografia por Emissão de Pósitrons , Ensaio Radioligante , Receptores de Dopamina D3/metabolismo , Transmissão Sináptica/genética , Adulto Jovem
6.
Mol Psychiatry ; 19(1): 129-39, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23337945

RESUMO

Emotional behavior is in part heritable and often disrupted in psychopathology. Identification of specific genetic variants that drive this heritability may provide important new insight into molecular and neurobiological mechanisms involved in emotionality. Our results demonstrate that the presynaptic vesicular monoamine transporter 1 (VMAT1) Thr136Ile (rs1390938) polymorphism is functional in vitro, with the Ile allele leading to increased monoamine transport into presynaptic vesicles. Moreover, we show that the Thr136Ile variant predicts differential responses in emotional brain circuits consistent with its effects in vitro. Lastly, deep sequencing of bipolar disorder (BPD) patients and controls identified several rare novel VMAT1 variants. The variant Phe84Ser was only present in individuals with BPD and leads to marked increase monoamine transport in vitro. Taken together, our data show that VMAT1 polymorphisms influence monoamine signaling, the functional response of emotional brain circuits and risk for psychopathology.


Assuntos
Sintomas Afetivos/genética , Emoções/fisiologia , Polimorfismo Genético/genética , Proteínas Vesiculares de Transporte de Monoamina/genética , Adolescente , Sintomas Afetivos/patologia , Animais , Monoaminas Biogênicas/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Encéfalo/patologia , Estudos de Casos e Controles , Linhagem Celular Transformada , Chlorocebus aethiops , Feminino , Estudos de Associação Genética , Genótipo , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Transfecção , Proteínas Vesiculares de Transporte de Monoamina/metabolismo , Adulto Jovem
7.
Mol Psychiatry ; 18(11): 1211-7, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23958960

RESUMO

The endogenous opioid system, which alleviates physical pain, is also known to regulate social distress and reward in animal models. To test this hypothesis in humans (n=18), we used an µ-opioid receptor (MOR) radiotracer to measure changes in MOR availability in vivo with positron emission tomography during social rejection (not being liked by others) and acceptance (being liked by others). Social rejection significantly activated the MOR system (i.e., reduced receptor availability relative to baseline) in the ventral striatum, amygdala, midline thalamus and periaqueductal gray (PAG). This pattern of activation is consistent with the hypothesis that the endogenous opioids have a role in reducing the experience of social pain. Greater trait resiliency was positively correlated with MOR activation during rejection in the amygdala, PAG and subgenual anterior cingulate cortex (sgACC), suggesting that MOR activation in these areas is protective or adaptive. In addition, MOR activation in the pregenual ACC was correlated with reduced negative affect during rejection. In contrast, social acceptance resulted in MOR activation in the amygdala and anterior insula, and MOR deactivation in the midline thalamus and sgACC. In the left ventral striatum, MOR activation during acceptance predicted a greater desire for social interaction, suggesting a role for the MOR system in social reward. The ventral striatum, amygdala, midline thalamus, PAG, anterior insula and ACC are rich in MORs and comprise a pathway by which social cues may influence mood and motivation. MOR regulation of this pathway may preserve and promote emotional well being in the social environment.


Assuntos
Encéfalo/metabolismo , Voluntários Saudáveis/psicologia , Distância Psicológica , Receptores Opioides mu/metabolismo , Adaptação Psicológica , Adulto , Afeto , Encéfalo/diagnóstico por imagem , Mapeamento Encefálico , Feminino , Fentanila/análogos & derivados , Humanos , Masculino , Cintilografia
8.
Psychol Med ; 43(7): 1433-45, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23298715

RESUMO

BACKGROUND: Facial emotion perception (FEP) is a critical human skill for successful social interaction, and a substantial body of literature suggests that explicit FEP is disrupted in major depressive disorder (MDD). Prior research suggests that weakness in FEP may be an important phenomenon underlying patterns of emotion-processing challenges in MDD and the disproportionate frequency of MDD in women. Method Women with (n = 24) and without (n = 22) MDD, equivalent in age and education, completed a FEP task during functional magnetic resonance imaging. RESULTS: The MDD group exhibited greater extents of frontal, parietal and subcortical activation compared with the control group during FEP. Activation in the inferior frontal gyrus (IFG) appeared shifted from a left >right pattern observed in healthy women to a bilateral pattern in MDD women. The ratio of left to right suprathreshold IFG voxels in healthy controls was nearly 3:1, whereas in the MDD group, there was a greater percentage of suprathreshold IFG voxels bilaterally, with no leftward bias. In MDD, relatively greater activation in right IFG compared with left IFG (ratio score) was present and predicted FEP accuracy (r = 0.56, p < 0.004), with an inverse relationship observed between FEP and subgenual cingulate activation (r = - 0.46, p = 0.02). CONCLUSIONS: This study links, for the first time, disrupted IFG activation laterality and increased subgenual cingulate activation with deficient FEP in women with MDD, providing an avenue for imaging-to-assessment translational applications in MDD.


Assuntos
Encéfalo/fisiopatologia , Transtorno Depressivo Maior/fisiopatologia , Emoções/fisiologia , Expressão Facial , Lobo Frontal/fisiopatologia , Lateralidade Funcional , Reconhecimento Visual de Modelos/fisiologia , Adulto , Tonsila do Cerebelo/fisiopatologia , Mapeamento Encefálico , Estudos de Casos e Controles , Córtex Cerebral/fisiopatologia , Feminino , Neuroimagem Funcional , Giro do Cíngulo/fisiopatologia , Humanos , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Pessoa de Meia-Idade , Adulto Jovem
9.
Mol Psychiatry ; 17(5): 511-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-21483437

RESUMO

Genetic factors, externalizing personality traits such as impulsivity, and brain processing of salient stimuli all can affect individual risk for alcoholism. One of very few confirmed genetic association findings differentiating alcoholics from non-alcoholics is with variants in the inhibitory γ-amino butyric acid α2 receptor subunit (GABRA2) gene. Here we report the association of two of these GABRA2 variants with measures of alcohol symptoms, impulsivity and with insula cortex activation during anticipation of reward or loss using functional magnetic resonance imaging (fMRI). In a sample of 173 families (449 subjects), 129 of whom had at least one member diagnosed with alcohol dependence or abuse, carriers for the G allele in two single-nucleotide polymorphisms (SNPs) and haplotypes were more likely to have alcohol dependence symptoms (rs279858, P=0.01; rs279826, P=0.05; haplotype, P=0.02) and higher NEO Personality Inventory-Revised (NEO-PI-R) Impulsiveness scores (rs279858, P=0.016; rs279826, P=0.012; haplotype, P=0.032) with a stronger effect in women (rs279858, P=0.011; rs279826, P=0.002; haplotype, P=0.006), all P-values are corrected for family history and age. A subset of offspring from these families (n=44, 20 females), genotyped for GABRA2, participated in an fMRI study using a monetary incentive delay task. Increased insula activation during reward (r(2)=0.4; P=0.026) and loss (r(2)=0.38; P=0.039) anticipation was correlated with NEO-PI-R Impulsiveness and further associated with the GG genotype for both SNPs (P's<0.04). Our results suggest that GABRA2 genetic variation is associated with Impulsiveness through variation of insula activity responses, here evidenced during anticipatory responses.


Assuntos
Alcoolismo/fisiopatologia , Antecipação Psicológica/fisiologia , Córtex Cerebral/fisiopatologia , Neuroimagem Funcional/psicologia , Comportamento Impulsivo/fisiopatologia , Receptores de GABA-A/fisiologia , Recompensa , Adolescente , Adulto , Idoso , Alcoolismo/diagnóstico , Alcoolismo/genética , Alelos , Saúde da Família , Feminino , Neuroimagem Funcional/métodos , Predisposição Genética para Doença/psicologia , Haplótipos/fisiologia , Humanos , Comportamento Impulsivo/genética , Imageamento por Ressonância Magnética/métodos , Imageamento por Ressonância Magnética/psicologia , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/fisiologia , Receptores de GABA-A/genética , Caracteres Sexuais
12.
Nat Med ; 2(11): 1225-9, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8898749

RESUMO

The endogenous opioid system has been recently implicated in the reinforcing actions of cocaine and other addictive drugs. In this study we examined mu opioid receptor binding in ten cocaine-dependent men and seven nonaddicted controls using positron emission tomography and [11C] carfentanil. Mu opioid binding was increased in several brain regions of the cocaine addicts studied 1-4 days after their last use of cocaine. Binding was positively correlated with the severity of cocaine craving experienced at the time. The upregulation of mu opioid receptor binding persisted after 4 weeks of monitored cocaine abstinence. These findings demonstrate for the first time the involvement of the endogenous opioid system in cocaine dependence and cocaine craving in living human subjects.


Assuntos
Analgésicos Opioides/metabolismo , Encéfalo/metabolismo , Cocaína/farmacologia , Fentanila/análogos & derivados , Transtornos Relacionados ao Uso de Opioides/metabolismo , Receptores Opioides mu/metabolismo , Tomografia Computadorizada de Emissão/métodos , Adulto , Analgésicos Opioides/farmacologia , Feminino , Fentanila/metabolismo , Fentanila/farmacologia , Humanos , Masculino , Síndrome de Abstinência a Substâncias , Fatores de Tempo
15.
Science ; 293(5528): 311-5, 2001 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-11452128

RESUMO

The endogenous opioid system is involved in stress responses, in the regulation of the experience of pain, and in the action of analgesic opiate drugs. We examined the function of the opioid system and mu-opioid receptors in the brains of healthy human subjects undergoing sustained pain. Sustained pain induced the regional release of endogenous opioids interacting with mu-opioid receptors in a number of cortical and subcortical brain regions. The activation of the mu-opioid receptor system was associated with reductions in the sensory and affective ratings of the pain experience, with distinct neuroanatomical involvements. These data demonstrate the central role of the mu-opioid receptors and their endogenous ligands in the regulation of sensory and affective components of the pain experience.


Assuntos
Encéfalo/fisiologia , Fentanila/análogos & derivados , Dor , Receptores Opioides mu/fisiologia , Adulto , Tonsila do Cerebelo/fisiologia , Analgésicos Opioides/administração & dosagem , Mapeamento Encefálico , Feminino , Fentanila/administração & dosagem , Humanos , Imageamento por Ressonância Magnética , Masculino , Músculo Masseter , Peptídeos Opioides/fisiologia , Medição da Dor , Tálamo/fisiologia , Tomografia Computadorizada de Emissão
16.
J Dent Res ; 98(12): 1324-1331, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31490699

RESUMO

Clinicians have the dilemma of prescribing opioid or nonopioid analgesics to chronic pain patients; however, the impact of pain on our endogenous µ-opioid system and how our genetic profile (specifically catechol-O-methyltransferase [COMT] polymorphisms) impacts its activation are currently unknown. Twelve chronic temporomandibular disorder (TMD) patients and 12 healthy controls (HCs) were scanned using positron emission tomography (PET) with [11C]carfentanil, a selective radioligand for µ-opioid receptors (µORs). The first 45 min of each PET measured the µOR nondisplaceable binding potential (BPND) at resting state, and the last 45 min consisted of a 20-min masseteric pain challenge with an injection of 5% hypertonic saline. Participants were also genotyped for different COMT alleles. There were no group differences in µOR BPND at resting state (early phase). However, during the masseteric pain challenge (late phase), TMD patients exhibited significant reductions in µOR BPND (decreased [11C]carfentanil binding) in the contralateral parahippocampus (P = 0.002) compared to HCs. The µOR BPND was also significantly lower in TMD patients with longer pain chronicity (P < 0.001). When considering COMT genotype and chronic pain suffering, TMD patients with the COMT158Met substitution had higher pain sensitivity and longer pain chronicity with a 5-y threshold for µOR BPND changes to occur in the parahippocampus. Together, the TMD diagnosis, COMT158Met substitution, and pain chronicity explained 52% of µOR BPND variance in the parahippocampus (cumulative R2 = 52%, P < 0.003, and HC vs. TMD Cohen's effect size d = 1.33 SD). There is strong evidence of dysregulation of our main analgesic and limbic systems in chronic TMD pain. The data also support precision medicine by helping identify TMD patients who may be more susceptible to chronic pain sensitivity and opioid dysfunction based on their genetic profile.


Assuntos
Catecol O-Metiltransferase/genética , Dor Crônica/genética , Receptores Opioides mu/fisiologia , Transtornos da Articulação Temporomandibular/fisiopatologia , Adulto , Analgésicos Opioides , Estudos de Casos e Controles , Feminino , Genótipo , Humanos , Masculino , Limiar da Dor , Polimorfismo de Nucleotídeo Único , Tomografia por Emissão de Pósitrons , Transtornos da Articulação Temporomandibular/genética , Adulto Jovem
17.
Transl Psychiatry ; 8(1): 222, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30323205

RESUMO

Placebo responses in depression exemplify how expectancies and appraisals impact mood. Cognitive and neural mechanisms underlying these responses are still poorly understood, partly due to the difficulty of simulating antidepressant effects and manipulating mood experimentally. To address these challenges, we developed an acute antidepressant placebo experiment involving the intravenous administration of a "fast-acting antidepressant" and a trial-by-trial sham fMRI "neurofeedback" manipulation, purporting to reveal mood-relevant neural responses. Twenty volunteers with major depression underwent this experiment while rating their expected and actual mood improvement. Mixed-effects analyses of trial-by-trial ratings revealed that the "drug" infusion cues induced higher expectancies of mood improvement, while both the "drug" infusion cue and the sham neurofeedback induced a reported mood improvement. Neurofeedback of greater magnitude, compared to lower magnitude, recruited the lateral prefrontal cortex (lPFC). Individuals with greater lPFC responses to neurofeedback displayed: (1) greater effect of previous mood improvement on expectancy ratings and (2) greater effect of sham neurofeedback on mood improvement. Behavioral antidepressant placebo effects were additionally moderated by changes in peripheral ß-endorphin plasma levels and depressive symptomatology. These data demonstrate the feasibility of trial-by-trial manipulation of antidepressant placebo-associated expectancies and their reinforcement. We provide initial insights into the role of the lPFC in the interplay between placebo-induced expectancies and mood, as well as preliminary evidence for the role of the opioid system in antidepressant placebo effects.


Assuntos
Antidepressivos/uso terapêutico , Transtorno Depressivo Maior/fisiopatologia , Transtorno Depressivo Maior/terapia , Efeito Placebo , Córtex Pré-Frontal/fisiopatologia , Reforço Psicológico , Adolescente , Adulto , Mapeamento Encefálico , Transtorno Depressivo Maior/tratamento farmacológico , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Neurorretroalimentação , Resultado do Tratamento , Adulto Jovem
18.
Addict Behav ; 41: 210-7, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25452067

RESUMO

INTRODUCTION: Previous research has demonstrated that chronic cigarette smoking and major depressive disorder (MDD) are each associated with cognitive decrements. Further, these conditions co-occur commonly, though mechanisms in the comorbid condition are poorly understood. There may be distinct, additive, or overlapping factors underlying comorbid cigarette smoking and MDD. The present study investigated the impact of smoking and MDD on executive function and emotion processing. METHODS: Participants (N=198) were grouped by diagnostic category (MDD and healthy controls, HC) and smoking status (ever-smokers, ES and never-smokers, NS). Participants completed the Facial Emotion Perception Test (FEPT), a measure of emotional processing, and the parametric Go/No-go task (PGNG), a measure of executive function. RESULTS: FEPT performance was analyzed using ANCOVA with accuracy and reaction time as separate dependent variables. Repeated measures MANCOVA was conducted for PGNG with performance measure and task level as dependent variables. Analyses for each task included diagnostic and smoking group as independent variables, and gender was controlled for. Results for FEPT reveal that lower overall accuracy was found for ES relative to NS, though MDD did not differ from HC. Post-hoc analyses revealed that ES were poorer at identifying happy and sad, but not fearful or angry, faces. For PGNG, poorer performance was observed in MDD relative to HC in response time to Go targets, but there were no differences for ES and NS. Interaction of diagnosis and smoking group was not observed for performance on either task. CONCLUSIONS: The results of this study provide preliminary evidence for distinctive cognitive decrements in smokers and individuals with depression.


Assuntos
Transtorno Depressivo Maior/epidemiologia , Transtorno Depressivo Maior/psicologia , Expressão Facial , Felicidade , Fumar/epidemiologia , Percepção Visual/fisiologia , Adulto , Análise de Variância , Transtorno Depressivo Maior/fisiopatologia , Emoções , Feminino , Humanos , Masculino , Michigan/epidemiologia , Estudos Retrospectivos
19.
J Clin Endocrinol Metab ; 86(2): 679-84, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11158031

RESUMO

Experimental evidence suggests that gonadal steroids regulate brain neurochemical systems associated with cognitive function, such as the cholinergic system. This study examines the effect of long-term postmenopausal hormone therapy on the brain concentrations of cholinergic synaptic terminals in women using single photon emission computed tomography and the radiotracer [(123)I]iodobenzovesamicol ([(123)I]IBVM). [(123)I]IBVM labels the vesicular acetylcholine transporter (VAChT) located in the presynaptic terminals of these neurons. Sixteen healthy women treated with hormone therapy since the menopause and 12 women not treated with hormones were studied. There were no significant differences in regional IBVM binding indexes between the 2 groups. The length of hormone replacement therapy correlated positively with VAChT binding indexes in multiple cortical areas (P < 0.05): frontal cortex (Spearman rank correlation: rho = 0.79), parietal cortex (rho = 0.62), temporal cortex (rho = 0.80), anterior cingulate (rho = 0.71), and posterior cingulate (rho = 0.63), but not in the basal ganglia (rho = 0.35; P = 0.2). An earlier onset of menopause in hormone-treated women was associated with higher VAChT indexes in the anterior cingulate (rho = -0.56; P = 0.02) and posterior cingulate (rho = -0.63; P = 0.01). The opposite was found in the posterior cingulate of women not treated with hormones (rho = 0.58; P = 0.04). Women treated with estrogen alone also showed higher VAChT indexes than women treated with estrogen and progestin in the posterior cingulate cortex (by Mann-Whitney U test: z = 2.42; P = 0.015). Although an overall effect of postmenopausal hormone therapy was not found, associations between an index of cortical cholinergic terminal concentrations and the length of hormonal replacement suggest that hormone therapy may influence the survival or plasticity of these cells in postmenopausal women. The data also suggest possible differential effects of estrogen and estrogen with progestin treatments in brain areas critical for cognitive processing.


Assuntos
Encéfalo/fisiologia , Proteínas de Transporte/análise , Terapia de Reposição de Estrogênios , Proteínas de Membrana Transportadoras , Piperidinas/farmacocinética , Pós-Menopausa/fisiologia , Vesículas Sinápticas/fisiologia , Tetra-Hidronaftalenos/farmacocinética , Proteínas de Transporte Vesicular , Idoso , Encéfalo/diagnóstico por imagem , Estrogênios , Feminino , Humanos , Radioisótopos do Iodo/farmacocinética , Pessoa de Meia-Idade , Especificidade de Órgãos , Progestinas , Valores de Referência , Análise de Regressão , Vesículas Sinápticas/diagnóstico por imagem , Fatores de Tempo , Tomografia Computadorizada de Emissão de Fóton Único , Proteínas Vesiculares de Transporte de Acetilcolina
20.
J Clin Endocrinol Metab ; 83(12): 4498-505, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9851799

RESUMO

The regulation of central mu-opioid receptors in women during the menstrual cycle was explored with positron emission tomography and the selective radiotracer [11C]carfentanil. Ten healthy women were studied twice, during their follicular and luteal phases. Plasma concentrations of estradiol, progesterone, testosterone, and beta-endorphin were determined immediately before scanning. LH pulsatility was measured over the 9 h preceding each of the two positron emission tomography scans. No significant differences in the binding potential of mu-opioid receptors (binding capacity/Kd) were observed between phases of the menstrual cycle. However, significant negative correlations were observed between circulating levels of estradiol during the follicular phase and mu-receptor binding measures in the amygdala and hypothalamus, two regions thought to be involved in the regulation of GnRH pulsatility. LH pulse amplitude was positively correlated with mu binding in the amygdala, whereas LH pulse number was negatively correlated with binding in this same region. No significant associations were noted between LH pulse measures and the hypothalamus for this sample. These results suggest that amygdalar mu-opioid receptors exert a modulatory effect on GnRH pulsatility, and that circulating levels of estradiol also regulate central mu-opioid function.


Assuntos
Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Ciclo Menstrual/fisiologia , Receptores Opioides/metabolismo , Tomografia Computadorizada de Emissão , Adulto , Anovulação/metabolismo , Feminino , Hormônios Esteroides Gonadais/metabolismo , Humanos , Hormônio Luteinizante/metabolismo , Ovulação/metabolismo , Fluxo Pulsátil , Receptores Opioides mu/metabolismo , Valores de Referência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA