Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Mol Psychiatry ; 24(4): 576-587, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-29955162

RESUMEN

The United States is in the midst of an opioid addiction and overdose crisis precipitated and exacerbated by use of prescription opioid medicines. The majority of opioid prescriptions are dispensed to patients with comorbid mood disorders including major depressive disorder (MDD). A growing body of research indicates that the endogenous opioid system is directly involved in the regulation of mood and is dysregulated in MDD. This involvement of the endogenous opioid system may underlie the disproportionate use of opioids among patients with mood disorders. Emerging approaches to address endogenous opioid dysregulation in MDD may yield novel therapeutics that have a low or absent risk of abuse and addiction relative to µ-opioid agonists. Moreover, agents targeting the endogenous opioid system would be expected to yield clinical benefits qualitatively different from conventional monaminergic antidepressants. The development of safe and effective agents to treat MDD-associated endogenous opioid dysregulation may represent a distinct and currently underappreciated means of addressing treatment resistant depression with the potential to attenuate the on-going opioid crisis.


Asunto(s)
Analgésicos Opioides/metabolismo , Depresión/tratamiento farmacológico , Depresión/fisiopatología , Analgésicos Opioides/farmacología , Analgésicos Opioides/uso terapéutico , Depresión/metabolismo , Trastorno Depresivo Mayor/tratamiento farmacológico , Trastorno Depresivo Mayor/fisiopatología , Sobredosis de Droga , Humanos , Trastornos del Humor/tratamiento farmacológico , Trastornos Relacionados con Opioides , Medicamentos bajo Prescripción , Estados Unidos
2.
Pain Med ; 21(8): 1538-1545, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31633789

RESUMEN

OBJECTIVE: Serotonin norepinephrine reuptake inhibitors (SNRIs) are commonly co-prescribed with opioids for chronic pain. The purpose of this study was to describe pain and mood response to venlafaxine among older adults with chronic low back pain (CLBP) and depression relative to opioid exposure. DESIGN: Secondary analyses were collected from a randomized clinical trial testing a stepped-care approach to comorbid pain and depression in older patients: the Addressing Depression and Pain Together study (ADAPT: 2010-2016). SETTING: University-based late-life mental health research clinic. SUBJECTS: Two hundred twenty-seven adults aged 65+ years with CLBP and depression. METHODS: Participants received six weeks of lower-dose venlafaxine (≤150 mg/d). Pain and depression were measured each week. Response for both pain and depression at the end of six weeks was defined by a ≥30% improvement on a 0-20 numeric rating scale for low back pain and a Patient Health Questionnaire-9 score ≤5. Opioid exposure was analyzed as prescribed (yes or no) and by morphine equivalent dosing (MED). RESULTS: Patients co-prescribed an opioid were less likely to report a pain response to venlafaxine. MED was negatively correlated with pain response. Depression response was not impacted. CONCLUSIONS: Opioids are negatively associated with older adults' early analgesic response to lower-dose venlafaxine. These findings suggest that clinicians may wish to consider either nonopioid or alternative antidepressant approaches to pain management in these complex patients. It is reassuring that opioids do not prevent depression response. Future research should examine both longer duration of treatment and a wider range of doses.


Asunto(s)
Dolor Crónico , Dolor de la Región Lumbar , Anciano , Analgésicos Opioides/uso terapéutico , Antidepresivos , Dolor Crónico/tratamiento farmacológico , Depresión/tratamiento farmacológico , Humanos , Dolor de la Región Lumbar/tratamiento farmacológico , Clorhidrato de Venlafaxina
3.
J Neurosci ; 35(27): 9957-65, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-26156996

RESUMEN

Back pain is common in the general population, but only a subgroup of back pain patients develops a disabling chronic pain state. The reasons for this are incompletely understood, but recent evidence implies that both preexisting and pain-related variations in the structure and function of the nervous system may contribute significantly to the development of chronic pain. Here, we addressed the role of striatal dopamine (DA) D2/D3 receptor (D2/D3R) function in chronic non-neuropathic back pain (CNBP) by comparing CNBP patients and healthy controls using PET and the D2/D3R-selective radioligand [(11)C]raclopride. D2/D3R availability was measured at baseline and during a pain challenge, yielding in vivo measures of receptor availability (binding potential, BPND) and DA release (change in BPND from baseline to activated state). At baseline, CNBP patients demonstrated reductions in D2/D3R BPND in the ventral striatum compared with controls. These reductions were associated with greater positive affect scores and pain tolerance measures. The reductions in D2/D3R BPND were also correlated with µ-opioid receptor BPND and pain-induced endogenous opioid system activation in the amygdala, further associated with measures of positive affect, the affective component of back pain and pain tolerance. During the pain challenge, lower magnitudes of DA release, and therefore D2/D3R activation, were also found in the ventral striatum in the CNBP sample compared with controls. Our results show that CNBP is associated with adaptations in ventral striatal D2/D3R function, which, together with endogenous opioid system function, contribute to the sensory and affective-motivational features of CNBP. SIGNIFICANCE STATEMENT: The neural systems that underlie chronic pain remain poorly understood. Here, using PET, we provide insight into the molecular mechanisms that regulate sensory and affective dimensions of pain in chronic back pain patients. We found that patients with back pain have alterations in brain dopamine function that are associated with measures of pain sensitivity and affective state, but also with brain endogenous opioid system functional measures. These findings suggest that brain dopamine-opioid interactions are involved in the pathophysiology of chronic pain, which has potential therapeutic implications. Our results may also help to explain individual variation in susceptibility to opioid medication misuse and eventual addiction in the context of chronic pain.


Asunto(s)
Dolor de Espalda/patología , Dopamina/metabolismo , Transmisión Sináptica/fisiología , Estriado Ventral/metabolismo , Estriado Ventral/fisiopatología , Adulto , Analgésicos Opioides/farmacocinética , Mapeo Encefálico , Radioisótopos de Carbono/farmacocinética , Enfermedad Crónica , Antagonistas de Dopamina/farmacocinética , Femenino , Fentanilo/análogos & derivados , Fentanilo/farmacocinética , Humanos , Masculino , Dimensión del Dolor , Unión Proteica/efectos de los fármacos , Racloprida/farmacocinética , Cintigrafía , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D3/metabolismo , Estriado Ventral/diagnóstico por imagen , Adulto Joven
4.
J Int Neuropsychol Soc ; 22(4): 412-25, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26831638

RESUMEN

OBJECTIVES: Individuals with major depressive disorder (MDD) demonstrate poorer learning and memory skills relative to never-depressed comparisons (NDC). Previous studies report decreased volume and disrupted function of frontal lobes and hippocampi in MDD during memory challenge. However, it has been difficult to dissociate contributions of short-term memory and executive functioning to memory difficulties from those that might be attributable to long-term memory deficits. METHODS: Adult males (MDD, n=19; NDC, n=22) and females (MDD, n=23; NDC, n=19) performed the Semantic List Learning Task (SLLT) during functional magnetic resonance imaging. The SLLT Encoding condition consists of 15 lists, each containing 14 words. After each list, a Distractor condition occurs, followed by cued Silent Rehearsal instructions. Post-scan recall and recognition were collected. Groups were compared using block (Encoding-Silent Rehearsal) and event-related (Words Recalled) models. RESULTS: MDD displayed lower recall relative to NDC. NDC displayed greater activation in several temporal, frontal, and parietal regions, for both Encoding-Silent Rehearsal and the Words Recalled analyses. Groups also differed in activation patterns in regions of the Papez circuit in planned analyses. The majority of activation differences were not related to performance, presence of medications, presence of comorbid anxiety disorder, or decreased gray matter volume in MDD. CONCLUSIONS: Adults with MDD exhibit memory difficulties during a task designed to reduce the contribution of individual variability from short-term memory and executive functioning processes, parallel with decreased activation in memory and executive functioning circuits. Ecologically valid long-term memory tasks are imperative for uncovering neural correlates of memory performance deficits in adults with MDD.


Asunto(s)
Aprendizaje por Asociación/fisiología , Corteza Cerebral/diagnóstico por imagen , Señales (Psicología) , Trastorno Depresivo Mayor/complicaciones , Trastorno Depresivo Mayor/patología , Discapacidades para el Aprendizaje/etiología , Sistema Límbico/diagnóstico por imagen , Semántica , Adolescente , Adulto , Anciano , Análisis de Varianza , Mapeo Encefálico , Trastorno Depresivo Mayor/diagnóstico por imagen , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Discapacidades para el Aprendizaje/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Trastornos de la Memoria/diagnóstico por imagen , Trastornos de la Memoria/etiología , Persona de Mediana Edad , Pruebas Neuropsicológicas , Adulto Joven
5.
J Neurosci ; 34(17): 5874-81, 2014 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-24760847

RESUMEN

Brain-derived neurotrophic factor (BDNF) levels in dopaminergic (DA) cells within the ventral tegmental area (VTA)/nucleus accumbens (NAc) circuitry appear to be a candidate mechanism for the neuroadaptive changes that follow stress and reward responses in animal models. However, the role of the BDNF gene variants in responses to salient cues through DA neurotransmission in humans remains unexplored. Here, we studied the effect of the common functional BDNF Val(66)Met (rs6265) polymorphism on rewarding experiences in the striatum and DA-mediated responses to stress. Seventy-two healthy controls were genotyped for the BDNF Val(66)Met polymorphism and underwent the monetary incentive delay task during an functional magnetic resonance imaging (fMRI) session. Forty-nine of them also underwent a sustained pain challenge with and without placebo administration with potential analgesic properties during PET measures of DA D2/3-receptor-mediated neurotransmission. Neuroimaging results revealed a significant effect of BDNF (Met(66) carriers > Val/Val) on brain responses during the anticipation of monetary losses, baseline D2/3 receptor availability, and pain-stress-induced DA release in the NAc. Conversely, BDNF Met(66) carriers showed no activation in response to monetary gains and a blunted DA response to the analgesic placebo in the NAc. These results provide initial human evidence regarding the effect of the BDNF Val(66)Met polymorphism on DA-mediated responses to stress, its cognitive regulation by positive expectations, and the anticipatory responses to monetary gains and losses in the VTA-NAc pathway. Our results are of relevance to the neurobiology of stress and reward interactions and the pathophysiology of stress-related disorders.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Cuerpo Estriado/fisiología , Neuronas Dopaminérgicas/fisiología , Núcleo Accumbens/fisiología , Polimorfismo de Nucleótido Simple , Recompensa , Estrés Fisiológico/fisiología , Adulto , Mapeo Encefálico , Femenino , Genotipo , Humanos , Imagen por Resonancia Magnética , Masculino , Pruebas Neuropsicológicas , Dolor/fisiopatología , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D3/genética , Receptores de Dopamina D3/metabolismo , Transmisión Sináptica/fisiología , Área Tegmental Ventral/fisiología
6.
J Neurosci ; 33(37): 14729-37, 2013 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-24027273

RESUMEN

The absence of consistent end organ abnormalities in many chronic pain syndromes has led to a search for maladaptive CNS mechanisms that may explain their clinical presentations and course. Here, we addressed the role of brain regional µ-opioid receptor-mediated neurotransmission, one of the best recognized mechanisms of pain regulation, in chronic back pain in human subjects. We compared µ-opioid receptor availability in vivo at baseline, during pain expectation, and with moderate levels of sustained pain in 16 patients with chronic nonspecific back pain (CNBP) and in 16 age- and gender-matched healthy control subjects, using the µ-opioid receptor-selective radioligand [(11)C]carfentanil and positron emission tomography. We found that CNBP patients showed baseline increases in thalamic µ-opioid receptor availability, contrary to a previously studied sample of patients diagnosed with fibromyalgia. During both pain expectation and sustained pain challenges, CNBP patients showed regional reductions in the capacity to activate this neurotransmitter system compared with their control sample, further associated with clinical pain and affective state ratings. Our results demonstrate heterogeneity in endogenous opioid system functional measures across pain conditions, and alterations in both receptor availability and endogenous opioid function in CNBP that are relevant to the clinical presentation of these patients and the effects of opioid analgesics on µ-opioid receptors.


Asunto(s)
Analgésicos Opioides/metabolismo , Dolor de Espalda/patología , Encéfalo/metabolismo , Adulto , Análisis de Varianza , Dolor de Espalda/diagnóstico por imagen , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Mapeo Encefálico , Radioisótopos de Carbono , Estudios de Casos y Controles , Enfermedad Crónica , Femenino , Fentanilo/análogos & derivados , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Dimensión del Dolor , Umbral del Dolor/efectos de los fármacos , Tomografía de Emisión de Positrones , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
7.
J Neurosci ; 32(9): 3253-60, 2012 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-22378896

RESUMEN

The corticotropin-releasing hormone (CRH) system coordinates neuroendocrine and behavioral responses to stress and has been implicated in the development of major depressive disorder (MDD). Recent reports suggest that GG-homozygous individuals of a single nucleotide polymorphism (rs110402) in the CRH receptor 1 (CRHR1) gene show behavioral and neuroendocrine evidence of stress vulnerability. The present study explores whether those observations extend to the neuronal processing of emotional stimuli in humans. CRHR1 was genotyped in 83 controls and a preliminary sample of 16 unmedicated patients with MDD who completed a functional magnetic resonance imaging scan while viewing blocks of positive, negative, and neutral words. In addition, potential mediating factors such as early life stress, sex, personality traits, and negative memory bias were examined. Robust differences in blood oxygenation level-dependent (BOLD) signal were found in healthy controls (A allele carriers > GG-homozygotes) in the right middle temporal/angular gyrus while subjects were viewing negative versus neutral words. Among GG-homozygotes, BOLD signal in the subgenual cingulate was greater in MDD participants (n = 9) compared with controls (n = 33). Conversely, among A-carriers, BOLD signal was smaller in MDD (n = 7) compared with controls (n = 50) in the hypothalamus, bilateral amygdala, and left nucleus accumbens. Early life stress, personality traits, and levels of negative memory bias were associated with brain activity depending on genotype. Results from healthy controls and a preliminary sample of MDD participants show that CRHR1 single nucleotide polymorphism rs110402 moderates neural responses to emotional stimuli, suggesting a potential mechanism of vulnerability for the development of MDD.


Asunto(s)
Emociones/fisiología , Variación Genética/fisiología , Imagen por Resonancia Magnética/métodos , Receptores de Hormona Liberadora de Corticotropina/genética , Transducción de Señal/genética , Adulto , Trastorno Depresivo Mayor/genética , Trastorno Depresivo Mayor/psicología , Femenino , Variación Genética/genética , Humanos , Masculino , Persona de Mediana Edad , Receptores de Hormona Liberadora de Corticotropina/fisiología , Transducción de Señal/fisiología , Adulto Joven
8.
JAMA Psychiatry ; 80(4): 389-398, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36857039

RESUMEN

Importance: Despite high antidepressant placebo response rates, the mechanisms underlying the persistence of antidepressant placebo effects are still poorly understood. Objective: To investigate the neurobehavioral mechanisms underlying the evolution of antidepressant placebo effects using a reinforcement learning (RL) framework. Design, Setting, and Participants: In this acute within-patient cross-sectional study of antidepressant placebos, patients aged 18 to 55 years not receiving medication for major depressive disorder (MDD) were recruited at the University of Pittsburgh between February 21, 2017, to March 1, 2021. Interventions: The antidepressant placebo functional magnetic resonance imaging task manipulates placebo-associated expectancies using visually cued fast-acting antidepressant infusions and controls their reinforcement with sham visual neurofeedback while assessing expected and experienced mood improvement. Main Outcomes and Measures: The trial-by-trial evolution of expectancies and mood was examined using multilevel modeling and RL, relating model-predicted signals to spatiotemporal dynamics of blood oxygenation level-dependent (BOLD) response. Results: A bayesian RL model comparison in 60 individuals (mean [SE] age, 24.5 [0.8] years; 51 females [85%]) with MDD revealed that antidepressant placebo trial-wise expectancies were updated by composite learning signals multiplexing sensory evidence (neurofeedback) and trial-wise mood (bayesian omnibus risk <0.001; exceedance probability = 97%). Placebo expectancy, neurofeedback manipulations, and composite learning signals modulated the visual cortex and dorsal attention network (threshold-free cluster enhancement [TFCE] = 1 - P >.95). As participants anticipated antidepressant infusions, learned placebo expectancies modulated the salience network (SN, TFCE = 1 - P >.95), positively scaling with depression severity. Conclusions and Relevance: Results of this cross-sectional study suggest that on a timescale of minutes, antidepressant placebo effects were maintained by positive feedback loops between expectancies and mood improvement. During learning, representations of placebos and their perceived effects were enhanced in primary and secondary sensory cortices. Latent learned placebo expectancies were encoded in the SN.


Asunto(s)
Trastorno Depresivo Mayor , Adulto , Femenino , Humanos , Adulto Joven , Antidepresivos/uso terapéutico , Teorema de Bayes , Estudios Transversales , Trastorno Depresivo Mayor/tratamiento farmacológico , Retroalimentación
9.
Neuroimage Clin ; 36: 103157, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36027717

RESUMEN

Major depressive disorder is among the most prevalent psychiatric disorders, exacting a substantial personal, social, and economic toll. Antidepressant treatment typically involves an individualized trial and error approach with an inconsistent success rate. Despite a pressing need, no reliable biomarkers for predicting treatment outcome have yet been discovered. Brain MRI measures hold promise in this regard, though clinical translation remains elusive. In this review, we summarize structural MRI and functional MRI (fMRI) measures that have been investigated as predictors of treatment outcome. We broadly divide these into five categories including three structural measures: volumetric, white matter burden, and white matter integrity; and two functional measures: resting state fMRI and task fMRI. Currently, larger hippocampal volume is the most widely replicated predictor of successful treatment. Lower white matter hyperintensity burden has shown robustness in late life depression. However, both have modest discriminative power. Higher fractional anisotropy of the cingulum bundle and frontal white matter, amygdala hypoactivation and anterior cingulate cortex hyperactivation in response to negative emotional stimuli, and hyperconnectivity within the default mode network (DMN) and between the DMN and executive control network also show promise as predictors of successful treatment. Such network-focused measures may ultimately provide a higher-dimensional measure of treatment response with closer ties to the underlying neurobiology.


Asunto(s)
Trastorno Depresivo Mayor , Sustancia Blanca , Humanos , Trastorno Depresivo Mayor/diagnóstico por imagen , Trastorno Depresivo Mayor/tratamiento farmacológico , Mapeo Encefálico , Encéfalo/diagnóstico por imagen , Imagen por Resonancia Magnética
10.
Contemp Clin Trials ; 107: 106503, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34237458

RESUMEN

The desire to reduce high placebo response rates in clinical trials is a popular concept. However, few studies have rigorously examined the effectiveness of methods to control for placebo responses that are relevant to randomized controlled trials. The primary objective of this review was to evaluate the effect of experimental placebo manipulations in randomized controlled trials (RCTs). We critically reviewed studies designed to manipulate placebo responses including positive expectations regarding the effectiveness of the placebo treatment, manipulating the time spent with subjects, and training study staff and subjects to accurately report symptom severity. These efforts have generally resulted in reduced placebo response and improved discrimination between drug and placebo. Interventions that neutralize staff and subject expectations and improve the ability of subjects to accurately report symptom severity have shown the most promise. Reduction of the placebo response has the potential to accelerate the development of new therapeutics.


Asunto(s)
Efecto Placebo , Humanos
11.
Artículo en Inglés | MEDLINE | ID: mdl-33732892

RESUMEN

Over the last two decades, neuroscientists have used antidepressant placebo probes to examine the biological mechanisms implicated in antidepressant placebo effects. However, findings from these studies have not yet elucidated a model-based theory that would explain the mechanism through which antidepressant expectancies evolve to induce persistent mood changes. Emerging evidence suggests that antidepressant placebo effects may be informed by models of reinforcement learning (RL). Such that an individual's expectation of improvement is updated with the arrival of new sensory evidence, by incorporating a reward prediction error (RPE), which signals the mismatch between the expected (expected value) and perceived improvement. Consistent with this framework, neuroimaging studies of antidepressant placebo effects have demonstrated placebo-induced µ-opioid activation and increased blood-oxygen-level dependent (BOLD) responses in regions tracking expected values (e.g., ventromedial prefrontal cortex (vmPFC)) and RPEs (e.g., ventral striatum (VS)). In this study, we will demonstrate the causal contribution of reward learning signals (expected values and RPEs) to antidepressant placebo effects by experimentally manipulating expected values using transcranial magnetic stimulation (TMS) targeting the vmPFC and µ-opioid striatal RPE signal using pharmacological approaches. We hypothesized that antidepressant placebo expectancies are represented in the vmPFC (expected value) and updated by means of µ-opioid-modulated striatal learning signal. In a 3 × 3 factorial double-blind design, we will randomize 120 antidepressant-free individuals with depressive symptoms to one of three between-subject opioid conditions: the µ-opioid agonist buprenorphine, the µ-opioid antagonist naltrexone, or an inert pill. Within each arm, individuals will be assigned to receive three within-subject counterbalanced forms of TMS targeting the vmPFC-intermittent Theta Burst Stimulation (TBS) expected to potentiate the vmPFC, continuous TBS expected to de-potentiate the vmPFC, or sham TBS. These experimental manipulations will be used to modulate trial-by-trial reward learning signals and related brain activity during the Antidepressant Placebo functional MRI (fMRI) Task to address the following aims: (1) investigate the relationship between reward learning signals within the vmPFC-VS circuit and antidepressant placebo effects; (2) examine the causal contribution of vmPFC expected value computations to antidepressant placebo effects; and (3) investigate the causal contribution of µ-opioid-modulated striatal RPEs to antidepressant placebo effects. The proposed study will be the first to investigate the causal contribution of µ-opioid-modulated vmPFC-VS learning signals to antidepressant placebo responses, paving the way for developing novel treatments modulating learning processes and objective means of quantifying and potentially reducing placebo effects during drug development. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT04276259.

12.
Artículo en Inglés | MEDLINE | ID: mdl-33684624

RESUMEN

BACKGROUND: Like placebo analgesia, the antidepressant placebo effect appears to involve cortical and subcortical endogenous opioid signaling, yet the mechanism through which opioid release affects mood remains unclear. The orbitofrontal cortex (OFC)-which integrates various attributes of a stimulus to predict associated outcomes-has been implicated in placebo effects and is rich in µ opioid receptors. We hypothesized that naltrexone blockade of µ opioid receptors would blunt OFC-dependent antidepressant placebo effects. METHODS: Twenty psychotropic-free patients with major depressive disorder completed a randomized, double-blind, placebo-controlled crossover study of 1 oral dose of 50 mg of naltrexone or matching placebo immediately before completing 2 sessions of the antidepressant placebo functional magnetic resonance imaging task. This task manipulates placebo-associated expectancies and their reinforcement while assessing expected and actual mood improvement. RESULTS: Behaviorally, manipulations of antidepressant placebo expectancies and their reinforcement had positive, interactive effects on participants' expectancy and mood ratings. The high-expectancy condition recruited the dorsolateral and ventrolateral prefrontal cortex, as well as dorsal attention stream regions. Interestingly, increased dorsolateral and ventrolateral prefrontal cortex brain responses appeared to attenuate the antidepressant placebo effect. The administration of 1 oral dose of naltrexone, compared with placebo, partially abolished the interaction of the expectancy and reinforcement manipulation on mood and blocked reinforcement-induced responses in the right central OFC. CONCLUSIONS: Our results show preliminary evidence for the role of µ opioid central OFC modulation in antidepressant placebo effects by positively biasing the value of placebo based on reinforcement and enhancing subsequent hedonic experiences.


Asunto(s)
Trastorno Depresivo Mayor , Naltrexona , Antidepresivos/uso terapéutico , Estudios Cruzados , Trastorno Depresivo Mayor/tratamiento farmacológico , Humanos , Naltrexona/farmacología , Naltrexona/uso terapéutico , Antagonistas de Narcóticos/farmacología , Antagonistas de Narcóticos/uso terapéutico , Efecto Placebo , Corteza Prefrontal
13.
Front Psychiatry ; 10: 669, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31616327

RESUMEN

Over the last two decades, neuroscientists have used antidepressant placebo probes to examine the biological mechanisms implicated in expectancies of mood improvement.However, findings from these studies have yet to elucidate a model-based theory that would explain the mechanisms through which antidepressant expectancies evolve to induce persistent mood changes. Compared to other fields, the development of experimental models of antidepressant placebo effects faces significant challenges, such as the delayed mechanism of action of conventional antidepressants and the complex internal dynamics of mood. Still, recent neuroimaging studies of antidepressant placebo effects have shown remarkable similarities to those observed in other disciplines (e.g., placebo analgesia), such as placebo-induced increased µ-opioid signaling and blood-oxygen-level dependent (BOLD) responses in areas involved in cognitive control, the representation of expected values and reward and emotional processing. This review will summarize these findings and the challenges and opportunities that arise from applying methodologies used in the field of placebo analgesia into the field of antidepressant placebo effects.

14.
Neuroimage Clin ; 21: 101679, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30685701

RESUMEN

Buprenorphine is currently being studied for treatment-resistant depression because of its rapid effect, relative safety, and unique pharmacodynamics. To understand the neural impact of buprenorphine in depression, we examined acute limbic and reward circuit changes during an intervention with low-dose buprenorphine augmentation pharmacotherapy. Mid and late-life adults with major depression (N = 31) who did not completely respond to an adequate trial of venlafaxine were randomized to augmentation with low-dose buprenorphine or matching placebo. We investigated early neural changes using functional magnetic resonance imaging (fMRI) from pre-randomization to 3 weeks using both an emotional reactivity task and a gambling task. We tested if: 1) there were significant neural changes acutely per intervention group, and 2) if acute neural changes were associated with depressive symptom change over 8 weeks using both the total score and the dysphoria subscale of the Montgomery Asberg Depression Rating Scale. Participants in both the buprenorphine and placebo groups showed similar changes in depressive symptoms. Neither the emotional reactivity nor gambling task resulted in significant neural activation changes from pre-randomization to 3-weeks. In both groups, increases in rostral anterior cingulate (rACC) and ventromedial prefrontal cortex (vmPFC) activation during the emotional reactivity task were associated with overall symptom improvement. In the buprenorphine but not the placebo group, increased activation in left anterior insula (aINS) and bilateral middle frontal gyrus (MFG) was associated with improvement on the dysphoria subscale. Activation changes in the reward task were not associated with buprenorphine. This is the first study to show an association between acute neural changes during emotion reactivity and changes in depression severity with buprenorphine treatment.


Asunto(s)
Buprenorfina/uso terapéutico , Depresión/tratamiento farmacológico , Trastorno Depresivo Mayor/tratamiento farmacológico , Recompensa , Anciano , Antidepresivos/uso terapéutico , Encéfalo/fisiopatología , Trastorno Depresivo Mayor/fisiopatología , Método Doble Ciego , Emociones/efectos de los fármacos , Emociones/fisiología , Femenino , Humanos , Imagen por Resonancia Magnética/métodos , Masculino
15.
Int Rev Neurobiol ; 138: 17-37, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29681324

RESUMEN

Expectancies are powerful modulators of cognitive and emotional experiences, as well as the neurobiological responses linked to these processes. In medicine, placebo effects are a clear example of how expectancies activate opioid neurotransmission in a treatment context, leading to the experience of analgesia and the improvement of emotional states, among other symptoms. Molecular neuroimaging techniques using positron emission tomography (PET) and the selective µ-opioid receptor tracer [11C]carfentanil 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 effects and provided the technical tools to examine inter-individual differences in the function of placebo responsive mechanisms. As a consequence, the capacity to activate endogenous opioid networks during the administration of placebos has been 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. This evidence has contributed to the understanding of the biological bases of the cognitive and psychological mechanisms implicated in the response to treatments, and opened up new opportunities for drug development and the enhancement of treatment responses. Further, delineation of these processes within and across diseases is critical to understand neural systems that could be enhanced to promote symptomatic improvement and modify disease progression.


Asunto(s)
Analgesia , Encéfalo/fisiología , Trastorno Depresivo Mayor/metabolismo , Conocimientos, Actitudes y Práctica en Salud , Nocicepción/fisiología , Personalidad/fisiología , Efecto Placebo , Receptores Opioides mu/fisiología , Transmisión Sináptica/fisiología , Analgesia/psicología , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Trastorno Depresivo Mayor/diagnóstico por imagen , Trastorno Depresivo Mayor/fisiopatología , Humanos , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo
16.
Eur Neuropsychopharmacol ; 27(10): 977-986, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28870407

RESUMEN

Dopamine (DA) neurotransmission within the brain's reward circuit has been implicated in the pathophysiology of depression and in both, cognitive and pharmacological mechanisms of treatment response. Still, a direct relationship between measures of DA neurotransmission and reward-related deficits in patients with depression has not been demonstrated. To gain insight into the symptom-specific alterations in the DA system in patients with depression, we used positron emission tomography (PET) and the D2/3 receptor-selective radiotracer [11C]raclopride in twenty-three non-smoking un-medicated Major Depressive Disorder (MDD) patients and sixteen healthy controls (HC). We investigated the relationship between D2/3 receptor availability and baseline measures of depression severity, anxiety, anhedonia, and cognitive and pharmacological mechanisms of treatment response. We found that, compared to controls, patients with depression showed greater D2/3 receptor availability in several striatal regions, including the bilateral ventral pallidum/nucleus accumbens (vPAL/NAc), and the right ventral caudate and putamen. In the depressed sample, D2/3 receptor availability in the caudal portion of the ventral striatum (NAc/vPAL) correlated with higher anxiety symptoms, whereas D2/3 receptor availability in the rostral area of the ventral striatum correlated negatively with the severity of motivational anhedonia. Finally, MDD non-remitters showed greater baseline anxiety, greater D2/3 availability in the NAc/vPAL, and greater placebo-induced DA release in the bilateral NAc. Our results demonstrate abnormally high D2/3 receptor availability in the ventral striatum of patients with MDD, which seem to be associated with comorbid anxiety symptoms and lack of response to antidepressants.


Asunto(s)
Trastorno Depresivo Mayor/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D3/metabolismo , Transmisión Sináptica/fisiología , Estriado Ventral/metabolismo , Adolescente , Adulto , Anhedonia/efectos de los fármacos , Anhedonia/fisiología , Antidepresivos/uso terapéutico , Ansiedad/diagnóstico por imagen , Ansiedad/tratamiento farmacológico , Ansiedad/metabolismo , Mapeo Encefálico , Estudios Cruzados , Trastorno Depresivo Mayor/diagnóstico por imagen , Trastorno Depresivo Mayor/tratamiento farmacológico , Trastorno Depresivo Mayor/psicología , Femenino , Fentanilo/análogos & derivados , Humanos , Masculino , Persona de Mediana Edad , Motivación/efectos de los fármacos , Motivación/fisiología , Tomografía de Emisión de Positrones , Radiofármacos , Transmisión Sináptica/efectos de los fármacos , Resultado del Tratamiento , Estriado Ventral/diagnóstico por imagen , Estriado Ventral/efectos de los fármacos , Adulto Joven
17.
Artículo en Inglés | MEDLINE | ID: mdl-26709390

RESUMEN

BACKGROUND: Recent neuroimaging studies have demonstrated resting-state functional connectivity (rsFC) abnormalities among intrinsic brain networks in Major Depressive Disorder (MDD); however, their role as predictors of treatment response has not yet been explored. Here, we investigate whether network-based rsFC predicts antidepressant and placebo effects in MDD. METHODS: We performed a randomized controlled trial of two weeklong, identical placebos (described as having either "active" fast-acting, antidepressant effects or as "inactive") followed by a ten-week open-label antidepressant medication treatment. Twenty-nine participants underwent a rsFC fMRI scan at the completion of each placebo condition. Networks were isolated from resting-state blood-oxygen-level-dependent signal fluctuations using independent component analysis. Baseline and placebo-induced changes in rsFC within the default-mode, salience, and executive networks were examined for associations with placebo and antidepressant treatment response. RESULTS: Increased baseline rsFC in the rostral anterior cingulate (rACC) within the salience network, a region classically implicated in the formation of placebo analgesia and the prediction of treatment response in MDD, was associated with greater response to one week of active placebo and ten weeks of antidepressant treatment. Machine learning further demonstrated that increased salience network rsFC, mainly within the rACC, significantly predicts individual responses to placebo administration. CONCLUSIONS: These data demonstrate that baseline rsFC within the salience network is linked to clinical placebo responses. This information could be employed to identify patients who would benefit from lower doses of antidepressant medication or non-pharmacological approaches, or to develop biomarkers of placebo effects in clinical trials.

18.
Psychopharmacology (Berl) ; 233(23-24): 3905-3919, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27614896

RESUMEN

RATIONALE: Oxytocin is a neuropeptide widely recognized for its role in regulating social and reproductive behavior. Increasing evidence from animal models suggests that oxytocin also modulates reward circuitry in non-social contexts, but evidence in humans is lacking. OBJECTIVES: We examined the effects of oxytocin administration on reward circuit function in 18 healthy men as they performed a monetary incentive task. METHODS: The blood oxygenation level-dependent (BOLD) signal was measured using functional magnetic resonance imaging in the context of a randomized, double-blind, placebo-controlled, crossover trial of intranasal oxytocin. RESULTS: We found that oxytocin increases the BOLD signal in the midbrain (substantia nigra and ventral tegmental area) during the late phase of the hemodynamic response to incentive stimuli. Oxytocin's effects on midbrain responses correlated positively with its effects on positive emotional state. We did not detect an effect of oxytocin on responses in the nucleus accumbens. Whole-brain analyses revealed that oxytocin attenuated medial prefrontal cortical deactivation specifically during anticipation of loss. CONCLUSIONS: Our findings demonstrate that intranasal administration of oxytocin modulates human midbrain and medial prefrontal function during motivated behavior. These findings suggest that endogenous oxytocin is a neurochemical mediator of reward behaviors in humans-even in a non-social context-and that the oxytocinergic system is a potential target of pharmacotherapy for psychiatric disorders that involve dysfunction of reward circuitry.


Asunto(s)
Hemodinámica/efectos de los fármacos , Motivación/efectos de los fármacos , Oxitócicos/farmacología , Oxitocina/farmacología , Corteza Prefrontal/efectos de los fármacos , Recompensa , Administración Intranasal , Adulto , Análisis de Varianza , Animales , Método Doble Ciego , Emociones/efectos de los fármacos , Humanos , Imagen por Resonancia Magnética , Masculino , Mesencéfalo/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Oxígeno/sangre , Área Tegmental Ventral/efectos de los fármacos , Adulto Joven
19.
Int J Clin Health Psychol ; 16(3): 266-275, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-30487870

RESUMEN

Background/Objective: The heterogeneous clinical presentations of individuals with Autism Spectrum Disorders (ASD) pose a significant challenge for sample characterization. Therefore the main goal of DSM-5 must be to identify subgroups of ASD, including comorbidity disorders and severity. The main goal of this study is to explore the psychiatric comorbidities and the severity of symptoms that could be relevant for the phenotype characterization in ASD and also to compare these results according to the different classification criteria between the DSM-IV-TR and the DSM-5. Method: A comparative study of severity and psychiatric comorbidities was carried out between a sample of participants that only met criteria for Pervasive Developmental Disorder (PDD) according to the DSM-IV-TR and a sample of participants that also met ASD criteria according to DSM-5 classification. The recruitment of children was via educational (N = 123). The psychiatric symptoms, comorbid disorders and severity of symptoms were assessed through The Nisonger Child Behavior Rating Form, clinical interview and The Inventory of Autism Spectrum Disorder, respectively. The psychiatric comorbidities considered were: anxiety, eating behavioural problems, self-aggressiveness, hetero-aggressiveness, self-harm, obsessive compulsive disorder and attention deficit and hyperactivity disorder. Results: Statistically significant differences between both groups were found regarding obsessive compulsive disorder, eating behavioural problems and severity. Conclusions: The results support the hypothesis that patients who meet the DSM-5 criteria have more severe symptoms, not only regarding the core autistic symptoms but also in relation with psychiatric comorbidities.


Antecedentes/Objetivo: Los Trastornos del Espectro Autista (TEA) incluyen un grupo heterogéneo en cuanto a su presentación clínica, que supone un desafío a nivel de caracterización diagnóstica. Por consiguiente, el objetivo principal de la clasificación DSM-5 debería de ser identificar subgrupos de TEA incluyendo severidad y comorbilidades psiquiátricas. El objetivo principal de este estudio es explorar las comorbilidades diagnósticas que pueden ser relevantes como descriptores de fenotipos autistas así como la severidad de los síntomas de autismo y comparar los resultados de las diferentes criterios de clasificación entre el DSM-IV-TR y el DSM-5. Método: Se realiza un estudio comparativo de severidad y comorbilidades psiquiátricas entre una muestra con diagnóstico de Trastorno Generalizado del Desarrollo, según criterios DSM-IV-TR, y una muestra que cumplía también criterios para TEA según la clasificación DSM-5. La muestra fue obtenida en centros educativos (N = 123). Las comorbilidades psiquiátricas y la severidad de los síntomas se evaluaron a través del The Nisonger Child Behavior Rating Form, entrevista clínica y el Inventario de Trastorno del Espectro Autista, respectivamente. Las comorbilidades estudiadas fueron ansiedad, alteraciones de la conducta alimentaria, auto-agresividad, hetero-agresividad, autolesiones, trastorno obsesivo-compulsivo y déficit de atención e hiperactividad. Resultados: Se encontraron diferencias estadísticamente significativas entre ambos grupos para trastorno obsesivo-compulsivo, alteraciones de la conducta alimentaria y severidad. Conclusiones: Se apoya la hipótesis de que los individuos que cumplen criterios diagnósticos según DSM-5 tienen mayor severidad sintomática, no sólo con respecto a los síntomas autistas centrales, sino también en relación con comorbilidades psiquiátricas.

20.
Neuropsychopharmacology ; 40(4): 957-65, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25308352

RESUMEN

Mu-opioid receptors (MOPRs) are critically involved in the modulation of pain and analgesia, and represent a candidate mechanism for the development of biomarkers of pain conditions and their responses to treatment. To further understand the human implications of genetic variation within the opioid system in pain and opioid-mediated placebo responses, we investigated the association between the functional single-nucleotide polymorphism (SNP) in the µ-opioid receptor gene (OPRM1), A118G, and psychophysical responses, personality traits, and neurotransmitter systems (dopamine (DA), opioid) related to pain and placebo analgesia. OPRM1 G carriers, compared with AA homozygotes, showed an overall reduction of baseline µ-opioid receptor availability in regions implicated in pain and affective regulation. In response to a sustained painful stimulus, we found no effect of A118G on pain-induced endogenous opioid release. Instead, AA homozygotes showed a blunted DA response in the nucleus accumbens (NAc) in response to the pain challenge. After placebo administration, G carriers showed more pronounced mood disturbances and lower placebo-induced µ-opioid system activation in the anterior insula (aINS), the amygdala (AMY), the NAc, the thalamus (THA), and the brainstem, as well as lower levels of DA D2/3 activation in the NAc. At a trait level, G carriers reported higher NEO-Neuroticism scores; a personality trait previously associated with increased pain and lower placebo responses, which were negatively correlated with baseline µ-opioid receptor availability in the aINS and subgenual anterior cingulate cortex (sgACC). Our results demonstrate that the A118G OPRM1 polymorphism contributes to interindividual variations in the function of neurotransmitters responsive to pain (endogenous opioid and dopamine), as well as their regulation through cognitive-emotional influences in the context of therapeutic expectations, the so-called placebo effect. These effects are relevant to human vulnerability to disease processes where these neurotransmitters have a role, such as persistent pain, mood, and substance use disorders, and responses to their treatments.


Asunto(s)
Mialgia/genética , Mialgia/psicología , Personalidad/genética , Polimorfismo de Nucleótido Simple/genética , Receptores Opioides mu/genética , Adulto , Afecto/efectos de los fármacos , Analgésicos Opioides/farmacocinética , Antagonistas de Dopamina/farmacocinética , Femenino , Fentanilo/análogos & derivados , Fentanilo/farmacocinética , Genotipo , Humanos , Masculino , Mialgia/tratamiento farmacológico , Mialgia/etiología , Dimensión del Dolor , Inventario de Personalidad , Estimulación Física/efectos adversos , Efecto Placebo , Tomografía de Emisión de Positrones , Racloprida/farmacocinética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda