Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 166
Filter
1.
J Pain ; 25(4): 1070-1081, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37956741

ABSTRACT

This study explored the association between experimentally-induced pain sensitivity and µ-opioid receptor (µOR) availability in patients with temporomandibular disorder (TMD) and further investigated any changes in the pain and µOR availability following high-definition transcranial direct current stimulation (HD-tDCS) over the primary motor cortex (M1) with pilot randomized clinical trials. Seven patients with TMD completed either active (n = 3) or sham treatment (n = 4) for 10 daily sessions and underwent positron emission tomography (PET) scans with [11C]carfentanil, a selective µOR agonist, a week before and after treatment. PET imaging consisted of an early resting and late phase with the sustained masseteric pain challenge by computer-controlled injection of 5% hypertonic saline. We also included 12 patients with TMD, obtained from our previous study, for baseline PET analysis. We observed that patients with more sensitivity to pain, indicated by lower infusion rate, had less µOR availability in the right amygdala during the late phase. Moreover, active M1 HD-tDCS, compared to sham, increased µOR availability post-treatment in the thalamus during the early resting phase and the amygdala, hippocampus, and parahippocampal gyrus during the late pain challenge phase. Importantly, increased µOR availability post-treatment in limbic structures including the amygdala and hippocampus was associated with decreased pain sensitivity. The findings underscore the role of the µOR system in pain regulation and the therapeutic potential of HD-tDCS for TMD. Nonetheless, large-scale studies are necessary to establish the clinical significance of these results. TRIAL REGISTRATION: ClinicalTrial.gov (NCT03724032) PERSPECTIVE: This study links pain sensitivity and µ-opioid receptors in patients with TMD. HD-tDCS over M1 improved µOR availability, which was associated with reduced pain sensitivity. Implications for TMD pain management are promising, but larger clinical trials are essential for validation.


Subject(s)
Temporomandibular Joint Disorders , Transcranial Direct Current Stimulation , Humans , Transcranial Direct Current Stimulation/methods , Pilot Projects , Pain Threshold/physiology , Pain , Temporomandibular Joint Disorders/diagnostic imaging , Temporomandibular Joint Disorders/therapy
2.
J Pain Res ; 16: 2509-2523, 2023.
Article in English | MEDLINE | ID: mdl-37497372

ABSTRACT

Objective: The current understanding of utilizing HD-tDCS as a targeted approach to improve headache attacks and modulate endogenous opioid systems in episodic migraine is relatively limited. This study aimed to determine whether high-definition transcranial direct current stimulation (HD-tDCS) over the primary motor cortex (M1) can improve clinical outcomes and endogenous µ-opioid receptor (µOR) availability for episodic migraineurs. Methods: In a randomized, double-blind, and sham-controlled trial, 25 patients completed 10-daily 20-min M1 HD-tDCS, repeated Positron Emission Tomography (PET) scans with a selective agonist for µOR. Twelve age- and sex-matched healthy controls participated in the baseline PET/MRI scan without neuromodulation. The primary endpoints were moderate-to-severe (M/S) headache days and responder rate (≥50% reduction on M/S headache days from baseline), and secondary endpoints included the presence of M/S headache intensity and the use of rescue medication over 1-month after treatment. Results: In a one-month follow-up, at initial analysis, both the active and sham groups exhibited no significant differences in their primary outcomes (M/S headache days and responder rates). Similarly, secondary outcomes (M/S headache intensity and the usage of rescue medication) also revealed no significant differences between the two groups. However, subsequent analyses showed that active M1 HD-tDCS, compared to sham, resulted in a more beneficial response predominantly in higher-frequency individuals (>3 attacks/month), as demonstrated by the interaction between treatment indicator and baseline frequency of migraine attacks on the primary outcomes. These favorable outcomes were also confirmed for the secondary endpoints in higher-frequency patients. Active treatment also resulted in increased µOR concentration compared to sham in the limbic and descending pain modulatory pathway. Our exploratory mediation analysis suggests that the observed clinical efficacy of HD-tDCS in patients with higher-frequency conditions might be potentially mediated through an increase in µOR availability. Conclusion: The 10-daily M1 HD-tDCS can improve clinical outcomes in episodic migraineurs with a higher baseline frequency of migraine attacks (>3 attacks/month). This improvement may be, in part, facilitated by the increase in the endogenous µOR availability. Clinical Trial Registration: www.ClinicalTrials.gov, identifier - NCT02964741.

3.
Front Neurosci ; 16: 889849, 2022.
Article in English | MEDLINE | ID: mdl-35747210

ABSTRACT

Chronic pain and reward processing are understood to be reciprocally related to one another. Previous studies of reward processing in chronic pain patients have reported incongruent findings. While several factors likely contribute to these disparate findings, these previous studies did not stratify their analyses by sex-a factor previously shown to robustly impact reward-related responses. Thus, we examined sex as a factor of interest in level of striatal activation during anticipation of monetary incentives among patients with chronic non-specific back pain and healthy controls (HC). This study utilized functional magnetic resonance imaging during a monetary incentive delay task to evaluate reward and loss responsivity in the striatum among males and females with and without chronic pain (N = 90). Group, sex, and group-by-sex interactions were analyzed via repeated measures analysis of variance. Among HC, males exhibited significantly greater blood oxygen level dependent (BOLD) signal in the striatum during reward anticipation, particularly during large reward trials. By contrast, no significant sex differences were observed among patients. A significant group-by-sex interaction was also observed, revealing diminished BOLD responses among males with chronic pain relative to control males. These results provide novel evidence of sex-specific reductions in anticipatory responses to reward in patients with chronic pain. Altered striatal reward responsivity among males, but not females, suggests that the reward systems of males and females are uniquely disrupted by chronic pain, and highlights the value of including sex as a factor of interest in future studies of reward responsivity in the context of persistent pain.

4.
Neuropsychopharmacology ; 47(10): 1784-1790, 2022 09.
Article in English | MEDLINE | ID: mdl-35545664

ABSTRACT

Major depressive disorder (MDD) and opioid use disorder (OUD) are common, potentially fatal, polygenic disorders that are moderately heritable and often co-occur. We examined the unique and shared associations of polygenic risk scores (PRS) for these disorders with µ-opioid receptor (MOR) concentration and endogenous opioid response during a stressful stimulus. Participants were 144 healthy European-ancestry (EA) subjects (88 females) who underwent MOR quantification scans with [11C]carfentanil and PET and provided DNA for genotyping. MOR non-displaceable binding potential (BPND) was measured in 5 regions of interest (ROIs) related to mood and addiction. We examined associations of PRS both at baseline and following opioid release calculated as the ratio of baseline and stress-challenge scans, first in the entire sample and then separately by sex. MOR availability at baseline was positively associated with MDD PRS in the amygdala and ventral pallidum. MDD and OUD PRS were significantly associated with stress-induced opioid system activation in multiple ROIs, accounting for up to 14.5% and 5.4%, respectively, of the variance in regional activation. The associations were most robust among females, where combined they accounted for up to 25.0% of the variance among the ROIs. We conclude that there is a pathophysiologic link between polygenic risk for MDD and OUD and opioid system activity, as evidenced by PRS with unique and overlapping regional associations with this neurotransmitter system. This link could help to explain the high rate of comorbidity of MDD and OUD and suggests that opioid-modulating interventions could be useful in treating MDD and OUD, both individually and jointly.


Subject(s)
Depressive Disorder, Major , Opioid-Related Disorders , Analgesics, Opioid/metabolism , Depression , Depressive Disorder, Major/diagnostic imaging , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/genetics , Female , Humans , Multifactorial Inheritance , Opioid Peptides , Opioid-Related Disorders/diagnostic imaging , Opioid-Related Disorders/drug therapy , Opioid-Related Disorders/genetics
5.
Transl Psychiatry ; 12(1): 20, 2022 01 12.
Article in English | MEDLINE | ID: mdl-35022382

ABSTRACT

µ-Opioid receptors (MOR) are a major target of endogenous and exogenous opioids, including opioid pain medications. The µ-opioid neurotransmitter system is heavily implicated in the pathophysiology of chronic pain and opioid use disorder and, as such, central measures of µ-opioid system functioning are increasingly being considered as putative biomarkers for risk to misuse opioids. To explore the relationship between MOR system function and risk for opioid misuse, 28 subjects with chronic nonspecific back pain completed a clinically validated measure of opioid misuse risk, the Pain Medication Questionnaire (PMQ), and were subsequently separated into high (PMQ > 21) and low (PMQ ≤ 21) opioid misuse risk groups. Chronic pain patients along with 15 control participants underwent two separate [11C]-carfentanil positron emission tomography scans to explore MOR functional measures: one at baseline and one during a sustained pain-stress challenge, with the difference between the two providing an indirect measure of stress-induced endogenous opioid release. We found that chronic pain participants at high risk for opioid misuse displayed higher baseline MOR availability within the right amygdala relative to those at low risk. By contrast, patients at low risk for opioid misuse showed less pain-induced activation of MOR-mediated, endogenous opioid neurotransmission in the nucleus accumbens. This study links human in vivo MOR system functional measures to the development of addictive disorders and provides novel evidence that MORs and µ-opioid system responsivity may underlie risk to misuse opioids among chronic pain patients.


Subject(s)
Chronic Pain , Opioid-Related Disorders , Analgesics, Opioid/adverse effects , Chronic Pain/drug therapy , Humans , Opioid-Related Disorders/complications , Opioid-Related Disorders/epidemiology , Positron-Emission Tomography , Surveys and Questionnaires
6.
Mol Psychiatry ; 27(2): 831-839, 2022 02.
Article in English | MEDLINE | ID: mdl-34716408

ABSTRACT

Behavioral conditioning and expectation can have profound impact on animal and human physiology. Placebo, administered under positive expectation in clinical trials, can have potent effects on disease pathology, obscuring active medications. Emerging evidence suggests placebo-responsive neurotransmitter systems (e.g., endogenous opioid) regulate immune function by manipulating inflammatory proteins including IL-18, a potent pro-inflammatory, nociceptive cytokine implicated in pathophysiology of various diseases. Validation that neuroimmune interactions involving brain µ-opioid receptor (MOR) activity and plasma IL-18 underlie placebo analgesic expectation could have widespread clinical applications. Unfortunately, current lack of mechanistic clarity obfuscates clinical translation. To elucidate neuroimmune interactions underlying placebo analgesia, we exposed 37 healthy human volunteers to a standardized pain challenge on each of 2 days within a Positron Emission Tomography (PET) neuroimaging paradigm using the MOR selective radiotracer, 11C-Carfentanil (CFN). Each day volunteers received an intervention (placebo under analgesic expectation or no treatment), completed PET scanning, and rated their pain experience. MOR BPND parametric maps were generated from PET scans using standard methods. Results showed placebo reduced plasma IL-18 during pain (W74 = -3.7, p < 0.001), the extent correlating with reduction in pain scores. Placebo reduction in IL-18 covaried with placebo-induced endogenous opioid release in the left nucleus accumbens (T148 = 3.33; puncorr < 0.001) and left amygdala (T148 = 3.30; puncorr < 0.001). These findings are consistent with a modulating effect of placebo (under analgesic expectation in humans) on a potent nociceptive, pro-inflammatory cytokine (IL-18) and underlying relationships with endogenous opioid activity, a neurotransmitter system critically involved in pain, stress, and mood regulation.


Subject(s)
Analgesics, Opioid , Receptors, Opioid, mu , Analgesics , Analgesics, Opioid/metabolism , Brain/metabolism , Humans , Interleukin-18/metabolism , Neurotransmitter Agents/metabolism , Opioid Peptides/metabolism , Pain/metabolism , Positron-Emission Tomography/methods , Receptors, Opioid, mu/metabolism , Synaptic Transmission/physiology
7.
Appl Neuropsychol Adult ; 27(2): 134-142, 2020.
Article in English | MEDLINE | ID: mdl-30811264

ABSTRACT

Memory difficulties are consistently reported in Major Depressive Disorder (MDD). Nonetheless, it has not been thoroughly investigated as to whether these deficits persist during remission from MDD. A group of 32 healthy young adults with no history of a mood disorder (Mage = 20.8, SD = 2.1) and 62 remitted depressed young adults (Mage = 21.1, SD = 1.9) completed a neuropsychological battery. The test battery included two measures of nonverbal memory, two measures of verbal memory, and a measure of performance validity. The testing session was repeated three to six weeks later to determine performance stability. No differences were found between healthy controls and remitted depressed patients in either memory domain (all ps > .05) and improvement in performance was exhibited over time for both groups (p = 0.004). Potential practice effects are examined. We found a stronger performance for women than men (p = 0.003), particularly for the Semantic List Learning Task (SLLT) (p = .047). Verbal and nonverbal memory and effort may not be impacted in those who are in a remitted state of MDD, early in the course of the illness. Women demonstrated auditory memory superiority over men, similar to prior research.


Subject(s)
Cognitive Dysfunction/physiopathology , Depressive Disorder, Major/physiopathology , Memory Disorders/physiopathology , Psychomotor Performance/physiology , Adult , Cognitive Dysfunction/etiology , Depressive Disorder, Major/complications , Female , Follow-Up Studies , Humans , Male , Memory Disorders/complications , Remission Induction , Sex Factors , Young Adult
8.
Addict Biol ; 25(2): e12743, 2020 03.
Article in English | MEDLINE | ID: mdl-30945801

ABSTRACT

Preclinical studies have shown effects of chronic exposure to addictive drugs on glutamatergic-mediated neuroplasticity in frontostriatal circuitry. These initial findings have been paralleled by human functional magnetic resonance imaging (fMRI) research demonstrating weaker frontostriatal resting-state functional connectivity (rsFC) among individuals with psychostimulant use disorders. However, there is a dearth of human imaging literature describing associations between long-term prescription opioid use, frontostriatal rsFC, and brain morphology among chronic pain patients. We hypothesized that prescription opioid users with chronic pain, as compared with healthy control subjects, would evidence weaker frontostriatal rsFC coupled with less frontostriatal gray matter volume (GMV). Further, those opioid use-related deficits in frontostriatal circuitry would be associated with negative affect and drug misuse. Prescription opioid users with chronic pain (n = 31) and drug-free healthy controls (n = 30) underwent a high-resolution anatomical and an eyes-closed resting-state functional scan. The opioid group, relative to controls, exhibited weaker frontostriatal rsFC, and less frontostriatal GMV in both L.NAc and L.vmPFC. Frontostriatal rsFC partially mediated group differences in negative affect. Within opioid users, L.NAc GMV predicted opioid misuse severity. The current study revealed that prescription opioid use in the context of chronic pain is associated with functional and structural abnormalities in frontostriatal circuitry. These results suggest that opioid use-related abnormalities in frontostriatal circuitry may undergird disturbances in affect that may contribute to the ongoing maintenance of opioid use and misuse. These findings warrant further examination of interventions to treat opioid pathophysiology in frontostriatal circuitry over the course of treatment.


Subject(s)
Affect/drug effects , Analgesics, Opioid/adverse effects , Chronic Pain/drug therapy , Corpus Striatum/drug effects , Corpus Striatum/physiopathology , Frontal Lobe/drug effects , Frontal Lobe/physiopathology , Adult , Analgesics, Opioid/therapeutic use , Chronic Pain/physiopathology , Corpus Striatum/diagnostic imaging , Female , Frontal Lobe/diagnostic imaging , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged
9.
Sci Adv ; 5(10): eaax1569, 2019 10.
Article in English | MEDLINE | ID: mdl-31663023

ABSTRACT

Addiction neuroscience models posit that recurrent drug use increases reactivity to drug-related cues and blunts responsiveness to natural rewards, propelling a cycle of hedonic dysregulation that drives addictive behavior. Here, we assessed whether a cognitive intervention for addiction, Mindfulness-Oriented Recovery Enhancement (MORE), could restructure reward responsiveness from valuation of drug-related reward back to valuation of natural reward. Before and after 8 weeks of MORE or a support group control, prescription opioid users (N = 135) viewed opioid and natural reward cues while an electroencephalogram biomarker of target engagement was assessed. MORE was associated with decreased opioid cue-reactivity and enhanced capacity to regulate responses to opioid and natural reward cues. Increased positive affective responses to natural reward cues were associated with decreased craving and mediated MORE's therapeutic effects on opioid misuse. This series of randomized experiments provide the first neurophysiological evidence that an integrative behavioral treatment can remediate hedonic dysregulation among chronic opioid users.


Subject(s)
Opioid-Related Disorders/psychology , Analgesics, Opioid/therapeutic use , Chronic Pain/drug therapy , Chronic Pain/psychology , Cues , Female , Humans , Male , Middle Aged , Mindfulness , Reward
10.
Pain Rep ; 4(4): e769, 2019.
Article in English | MEDLINE | ID: mdl-31579860

ABSTRACT

The enormous advancements in the medical imaging methods witnessed in the past decades have allowed clinical researchers to study the function of the human brain in vivo, both in health and disease. In addition, a better understanding of brain responses to different modalities of stimuli such as pain, reward, or the administration of active or placebo interventions has been achieved through neuroimaging methods. Although magnetic resonance imaging has provided important information regarding structural, hemodynamic, and metabolic changes in the central nervous system related to pain, magnetic resonance imaging does not address modulatory pain systems at the molecular level (eg, endogenous opioid). Such important information has been obtained through positron emission tomography, bringing insights into the neuroplastic changes that occur in the context of the pain experience. Positron emission tomography studies have not only confirmed the brain structures involved in pain processing and modulation but also have helped elucidate the neural mechanisms that underlie healthy and pathological pain regulation. These data have shown some of the biological basis of the interindividual variability in pain perception and regulation. In addition, they provide crucial information to the mechanisms that drive placebo and nocebo effects, as well as represent an important source of variability in clinical trials. Positron emission tomography studies have also permitted exploration of the dynamic interaction between behavior and genetic factors and between different pain modulatory systems. This narrative review will present a summary of the main findings of the positron emission tomography studies that evaluated the functioning of the opioidergic system in the context of pain.

11.
Neuroimage Clin ; 23: 101905, 2019.
Article in English | MEDLINE | ID: mdl-31279240

ABSTRACT

OBJECTIVE: To evaluate, in vivo, the impact of ongoing chronic migraine (CM) attacks on the endogenous µ-opioid neurotransmission. BACKGROUND: CM is associated with cognitive-emotional dysfunction. CM is commonly associated with frequent acute medication use, including opioids. METHODS: We scanned 15 migraine patients during the spontaneous headache attack (ictal phase): 7 individuals with CM and 8 with episodic migraine (EM), as well as 7 healthy controls (HC), using positron emission tomography (PET) with the selective µ-opioid receptor (µOR) radiotracer [11C]carfentanil. Migraineurs were scanned in two paradigms, one with thermal pain threshold challenge applied to the site of the headache, and one without thermal challenge. Multivariable analysis was performed between the µ-opioid receptor availability and the clinical data. RESULTS: µOR availability, measured with [11C]carfentanil nondisplaceable binding potential (BPND), in the left thalamus (P-value = 0.005) and left caudate (P-value = 0.003) were decreased in CM patients with thermal pain threshold during the ictal phase relative to HC. Lower µOR BPND in the right parahippocampal region (P-value = 0.001) and right amygdala (P-value = 0.002) were seen in CM relative to EM patients. Lower µOR BPND values indicate either a decrease in µOR concentration or an increase in endogenous µ-opioid release in CM patients. In the right amygdala, 71% of the overall variance in µOR BPND levels was explained by the type of migraine (CM vs. EM: partial-R2 = 0.47, P-value<0.001, Cohen's effect size d = 2.6SD), the severity of the attack (pain area and intensity number summation [P.A.I.N.S.]: partial-R2 = 0.16, P-value = 0.031), and the thermal pain threshold (allodynia: partial-R2 = 0.08). CONCLUSIONS: Increased endogenous µ-opioid receptor-mediated neurotransmission is seen in the limbic system of CM patients, especially in right amygdala, which is highly modulated by the attack frequency, pain severity, and sensitivity. This study demonstrates for the first time the negative impact of chronification and exacerbation of headache attacks on the endogenous µ-opioid mechanisms of migraine patients. ClinicalTrials.gov identifier: NCT03004313.


Subject(s)
Amygdala/metabolism , Migraine Disorders/metabolism , Migraine Disorders/physiopathology , Nociception/physiology , Pain Threshold/physiology , Parahippocampal Gyrus/metabolism , Receptors, Opioid, mu/metabolism , Adult , Amygdala/diagnostic imaging , Analgesics, Opioid/pharmacokinetics , Caudate Nucleus/diagnostic imaging , Caudate Nucleus/metabolism , Chronic Disease , Female , Fentanyl/analogs & derivatives , Fentanyl/pharmacokinetics , Humans , Male , Middle Aged , Migraine Disorders/diagnostic imaging , Parahippocampal Gyrus/diagnostic imaging , Physical Stimulation , Positron-Emission Tomography , Radiopharmaceuticals/pharmacokinetics , Severity of Illness Index , Thalamus/diagnostic imaging , Thalamus/metabolism , Young Adult
12.
Biol Psychol ; 146: 107709, 2019 09.
Article in English | MEDLINE | ID: mdl-31170437

ABSTRACT

Reward anticipation dysfunction is associated with major depressive disorder (MDD), but is not universally observed in individuals with MDD. Reward anticipation deficits have also been linked to childhood adversity (CA) and approach/avoidance traits. The present study evaluated whether severity of CA (as measured by the Childhood Trauma Questionnaire) and approach/avoidance traits predict individual differences in blood oxygen level dependent (BOLD) response to reward anticipation beyond MDD diagnosis alone. Participants were individuals with MDD (n = 23) and healthy controls (n = 27). Multiple regression was conducted using CTQ scores, trait approach/avoidance scores, and diagnosis to predict activation during reward anticipation in a monetary incentive delay fMRI task. Across groups, higher trait reward responsiveness predicted increased activation in the hippocampus, cingulate cortex, and medial frontal gyrus. Greater CTQ scores predicted increased reward network activation. Overall, CTQ and reward responsiveness scores predicted more variance in reward anticipation activation than diagnosis. These findings suggest that clinicians should assess history of childhood adversity and trait reward responsiveness when treating individuals with MDD.


Subject(s)
Adult Survivors of Child Adverse Events/psychology , Depression/psychology , Motivation/physiology , Reward , Adult , Child , Female , Gyrus Cinguli/diagnostic imaging , Hippocampus/diagnostic imaging , Humans , Individuality , Magnetic Resonance Imaging , Male , Multivariate Analysis , Prefrontal Cortex/diagnostic imaging , Regression Analysis , Young Adult
13.
Addict Behav Rep ; 9: 100181, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31193775

ABSTRACT

INTRODUCTION: Smoking rates are up to 2-4 times higher among individuals with mental illness. Hospital readmissions for patients with psychiatric illness within a year of discharge are also high, and there is limited evidence of associations between smoking and these readmissions. METHODS: This study was a secondary data analysis using clinical data of psychiatric inpatients with initial admissions between the years 2000 and 2015. Following a descriptive analysis, logistic regression models were fitted to explore relationships between smoking and psychiatric hospital readmission within 30 days and a year of discharge. RESULTS: A total of 5439 patients with average age of 30.18 ±â€¯15.97 were identified. Of this number, 47.0% were current smokers and 53.0% were never smokers. Within 30 days of discharge, 11% of the current smokers were readmitted compared to 9% of never smokers. The primary diagnoses with highest proportion of smokers were, opioid or substance use disorders (80.0%), schizophrenia (70.7%), alcohol dependence (68.2%), and bipolar disorders (50.8%). About 31% of current smokers were readmitted within one year of discharge compared to 26% of never smokers. Adjusted odds ratios for readmission within 1-year of discharge were, bipolar disorders (1.41, p = 0.01), schizophrenia (2.33, p < 0.001), and opioid/substance dependence (1.55, p = 0.01). CONCLUSION: Significant relationships exist between smoking and readmission for patients with psychiatric illness. Smokers are more likely to be readmitted within 30 days or one year after discharge. Interaction of smoking and certain specific diagnoses significantly increases readmission.

14.
Front Psychol ; 10: 691, 2019.
Article in English | MEDLINE | ID: mdl-30984083

ABSTRACT

Heterogeneity within Major Depressive Disorder (MDD) has hampered identification of biological markers (e.g., intermediate phenotypes, IPs) that might increase risk for the disorder or reflect closer links to the genes underlying the disease process. The newer characterizations of dimensions of MDD within Research Domain Criteria (RDoC) domains may align well with the goal of defining IPs. We compare a sample of 25 individuals with MDD compared to 29 age and education matched controls in multimodal assessment. The multimodal RDoC assessment included the primary IP biomarker, positron emission tomography (PET) with a selective radiotracer for 5-HT1A [(11C)WAY-100635], as well as event-related functional MRI with a Go/No-go task targeting the Cognitive Control network, neuropsychological assessment of affective perception, negative memory bias and Cognitive Control domains. There was also an exploratory genetic analysis with the serotonin transporter (5-HTTLPR) and monamine oxidase A (MAO-A) genes. In regression analyses, lower 5-HT1A binding potential (BP) in the MDD group was related to diminished engagement of the Cognitive Control network, slowed resolution of interfering cognitive stimuli, one element of Cognitive Control. In contrast, higher/normative levels of 5-HT1A BP in MDD (only) was related to a substantial memory bias toward negative information, but intact resolution of interfering cognitive stimuli and greater engagement of Cognitive Control circuitry. The serotonin transporter risk allele was associated with lower 1a BP and the corresponding imaging and cognitive IPs in MDD. Lowered 5HT 1a BP was present in half of the MDD group relative to the control group. Lowered 5HT 1a BP may represent a subtype including decreased engagement of Cognitive Control network and impaired resolution of interfering cognitive stimuli. Future investigations might link lowered 1a BP to neurobiological pathways and markers, as well as probing subtype-specific treatment targets.

15.
Mol Psychiatry ; 24(4): 576-587, 2019 04.
Article in English | MEDLINE | ID: mdl-29955162

ABSTRACT

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.


Subject(s)
Analgesics, Opioid/metabolism , Depression/drug therapy , Depression/physiopathology , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Depression/metabolism , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/physiopathology , Drug Overdose , Humans , Mood Disorders/drug therapy , Opioid-Related Disorders , Prescription Drugs , United States
16.
Article in English | MEDLINE | ID: mdl-30009871

ABSTRACT

A large number of studies have attempted to use neuroimaging tools to aid in treatment prediction models for major depressive disorder (MDD). Most such studies have reported on only one dimension of function and prediction at a time. In this study, we used three different tasks across domains of function (emotion processing, reward anticipation, and cognitive control, plus resting state connectivity completed prior to start of medication to predict treatment response in 13-36 adults with MDD. For each experiment, adults with MDD were prescribed only label duloxetine (all experiments), whereas another subset were prescribed escitalopram. We used a KeyNet (both Task derived masks and Key intrinsic Network derived masks) approach to targeting brain systems in a specific match to tasks. The most robust predictors were (Dichter et al., 2010) positive response to anger and (Gong et al., 2011) negative response to fear within relevant anger and fear TaskNets and Salience and Emotion KeyNet (Langenecker et al., 2018) cognitive control (correct rejections) within Inhibition TaskNet (negative) and Cognitive Control KeyNet (positive). Resting state analyses were most robust for Cognitive control Network (positive) and Salience and Emotion Network (negative). Results differed by whether an -fwhm or -acf (more conservative) adjustment for multiple comparisons was used. Together, these results implicate the importance of future studies with larger sample sizes, multidimensional predictive models, and the importance of using empirically derived masks for search areas.


Subject(s)
Antidepressive Agents/therapeutic use , Depressive Disorder, Major/diagnostic imaging , Depressive Disorder, Major/drug therapy , Emotions/physiology , Magnetic Resonance Imaging/methods , Photic Stimulation/methods , Adult , Brain/diagnostic imaging , Brain/drug effects , Citalopram/therapeutic use , Depressive Disorder, Major/psychology , Duloxetine Hydrochloride/therapeutic use , Emotions/drug effects , Female , Humans , Male , Middle Aged , Neuroimaging/methods , Predictive Value of Tests , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Treatment Outcome
17.
Neuropsychopharmacology ; 44(3): 495-502, 2019 02.
Article in English | MEDLINE | ID: mdl-30337638

ABSTRACT

Neuropeptide Y (NPY) produces anxiolytic effects in rodent models, and naturally occurring low NPY expression in humans has been associated with negative emotional phenotypes. Studies in rodent models have also demonstrated that NPY elicits reward behaviors through its action in the nucleus accumbens (NAc), but the impact of NPY on the human NAc is largely unexplored. We recruited 222 healthy young adults of either sex and genetically selected 53 of these subjects at the extremes of NPY expression (Low-NPY and High-NPY) to participate in functional magnetic resonance imaging. Responses of the NAc and surrounding ventral striatum were quantified during a monetary incentive delay task in which stimuli varied by salience (high versus low) and valence (win versus loss). We found that bilateral NAc responses to high-salience versus low-salience stimuli were greater for Low-NPY subjects relative to High-NPY subjects, regardless of stimulus valence. To our knowledge, these results provide the first evidence in humans linking NPY with salience sensitivity of the NAc, raising the possibility that individual differences in NPY expression moderate the risk for disorders of mesoaccumbal function such as addictions and mood disorders. Additionally, we found that head motion was greater among High-NPY subjects, consistent with previous reports linking NPY with hyperactivity. Future studies in animal models are warranted to elucidate the neural mechanisms through which NPY influences NAc function and related behaviors.


Subject(s)
Delay Discounting/physiology , Head Movements/physiology , Neuropeptide Y/physiology , Nucleus Accumbens/physiology , Reward , Adolescent , Adult , Female , Functional Neuroimaging , Gene Expression/genetics , Humans , Magnetic Resonance Imaging , Male , Neuropeptide Y/genetics , Nucleus Accumbens/diagnostic imaging , Young Adult
19.
J Affect Disord ; 241: 371-380, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30145507

ABSTRACT

BACKGROUND: Major depressive disorder (MDD) and anxiety disorders are highly comorbid, sharing many similar symptoms, including impairments in cognitive control. Deficits in cognitive control could be a potential mechanism underlying impaired emotion regulation in mood disorders. METHODS: Participants were 44 individuals with no history of mental illness (healthy controls, HC), 31 individuals in the remitted state of MDD (rMDD), and 18 individuals who met lifetime DSM-IV-TR criteria for rMDD and an anxiety disorder in remission (Comorbid). Participants completed a Parametric Go/No-Go (PGNG) test during fMRI. Event-related analyses modeled activity for cognitive control successes (Hits for Targets, Rejections for Lures) and failures (Commissions on Lures) on the PGNG task. RESULTS: The rMDD group showed significantly reduced activity within the cognitive control network (CCN) during Commission errors, including the middle frontal gyrus and inferior parietal lobule (IPL). The Comorbid group showed significantly reduced activity in several clusters within the CCN during correct Rejections, including the left IPL and right inferior frontal gyrus and greater subgenual cingulate. Notably, during correct Rejections, 60% of activation for the Comorbid group was within the Salience and Emotion Network (SEN), with 0% within the CCN. LIMITATIONS: The size of the Comorbid subgroup was modest, preventing subanalysis of the different AD subtypes. CONCLUSIONS: There is evidence that CCN activity declines in rMDD and that there may be compensatory SEN activity in individuals with Comorbid rMDD and anxiety. Our findings support the identification of comorbid anxiety as a meaningful subtype of MDD that may obscure group differences between rMDD and HCs.


Subject(s)
Anxiety Disorders/physiopathology , Cognition/physiology , Depressive Disorder, Major/physiopathology , Gyrus Cinguli/physiology , Adolescent , Brain Mapping , Comorbidity , Emotions/physiology , Executive Function , Female , Humans , Magnetic Resonance Imaging , Male , Neuropsychological Tests , Young Adult
20.
Menopause ; 25(12): 1424-1431, 2018 12.
Article in English | MEDLINE | ID: mdl-29994967

ABSTRACT

OBJECTIVE: Cognitive outcomes in trials of postmenopausal hormone treatment have been inconsistent. Differing outcomes may be attributed to hormone formulation, treatment duration and timing, and differential cognitive domain effects. We previously demonstrated treatment benefits on visual cognitive function. In the present study, we describe the effects of hormone treatment on verbal outcomes in the same women, seeking to understand the effects of prior versus current hormone treatment on verbal function. METHODS: This is a cross-sectional evaluation of 57 women (38 hormone users [25 prior long-term users and 13 current users] and 19 never-users). Hormone users took identical formulations of estrogen or estrogen + progestin (0.625 mg/d conjugated equine estrogens with or without medroxyprogesterone acetate) for at least 10 years, beginning within 2 years of menopause. Women were evaluated with tests of verbal function and functional magnetic resonance imaging (fMRI) of a verbal discrimination task. RESULTS: All women scored similarly on assessments of verbal function (Hopkins Verbal Learning Test and a verbal discrimination task performed during the fMRI scanning session); however, women ever treated with hormones had more left inferior frontal (T = 3.72; P < 0.001) and right prefrontal cortex (T = 3.53; P < 0.001) activation during the verbal task. Hormone-treated women performed slightly worse on the verbal discrimination task (mean accuracy 81.72 ±â€Š11.57 ever-treated, 85.30 ±â€Š5.87 never-treated, P = 0.14), took longer to respond (mean reaction time 1.10 ±â€Š0.17 s ever-treated, 1.02 ±â€Š0.11 never-treated, P = 0.03), and remembered fewer previously viewed words (mean accuracy 62.21 ±â€Š8.73 ever-treated, 65.45 ±â€Š7.49 never-treated, P = 0.18). Increased posterior cingulate activity was associated with longer response times (R = 0.323, P = 0.015) and worse delayed verbal recall (R = -0.328, P = 0.048), suggesting that increased activation was associated with less efficient cognitive processing. We did not detect between group differences in activation in the left prefrontal cortex, superior frontal cortex, thalamus, or occipital/parietal junction. CONCLUSIONS: Although current and past hormone treatment was associated with differences in neural pathways used during verbal discrimination, verbal function was not higher than never-users.


Subject(s)
Cognition/drug effects , Estrogen Receptor Modulators/pharmacology , Estrogen Replacement Therapy/psychology , Estrogens, Conjugated (USP)/pharmacology , Estrogens/pharmacology , Medroxyprogesterone Acetate/pharmacology , Neural Pathways/drug effects , Postmenopause/drug effects , Aged , Cross-Sectional Studies , Drug Combinations , Female , Humans , Magnetic Resonance Imaging , Memory, Short-Term/physiology , Mental Recall/physiology , Middle Aged , Neuropsychological Tests , Prefrontal Cortex/diagnostic imaging , Prefrontal Cortex/metabolism , Reaction Time , Treatment Outcome , Verbal Learning
SELECTION OF CITATIONS
SEARCH DETAIL
...