Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 185
Filter
1.
Eur Arch Psychiatry Clin Neurosci ; 271(5): 835-845, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33001272

ABSTRACT

The Habenula is increasingly being investigated in addiction. Reduced volumes of other relevant brain regions in addiction, such as nucleus accumbens, globus pallidus and hypothalamus have been reported. Reduced volumes of the habenula as well as reduced neuronal cell count in the habenula have also been reported in mood disorders and an overlap between mood disorders and addiction is clinically widely recognized. Thus, our aim was to investigate possible volume and neuronal cell count differences in heroin addicts compared to healthy controls. Volumes of the medial (MHB) and lateral habenula (LHB) in heroin addicts (n = 12) and healthy controls (n = 12) were assessed by morphometry of 20 µm serial whole brain sections. Total brain volume was larger in the heroin group (mean 1466.6 ± 58.5 cm3 vs. mean 1331.5 ± 98.8 cm3), possibly because the heroin group was about 15 years younger (p = 0.001). Despite larger mean whole brain volume, the mean relative volume of the MHB was smaller than in healthy non-addicted controls (6.94 ± 2.38 × 10-6 vs.10.64 ± 3.22 × 10-6; p = 0.004). A similar finding was observed regarding relative volumes of the LHB (46.62 ± 10.90 × 10-6 vs. 63.05 ± 16.42 × 10-6 p = 0.009). In parallel, neuronal cell numbers were reduced in the MHB of heroin-addicted subjects (395,966 ± 184,178 vs. 644,149 ± 131,140; p < 0.001). These findings were not significantly confounded by age and duration of autolysis. Our results provide further evidence for brain-structural deficits in heroin addiction.


Subject(s)
Habenula , Heroin Dependence , Neurons , Autopsy , Case-Control Studies , Cell Count , Habenula/pathology , Heroin Dependence/pathology , Humans , Male , Neurons/pathology , Organ Size
2.
J Cell Biochem ; 121(2): 1870-1879, 2020 02.
Article in English | MEDLINE | ID: mdl-31692007

ABSTRACT

Heroin dependence is a complex behavioral disease, and a chronic encephalopathy with the important feature of relapse. The purpose of the study was to identify the regulatory mechanism of the nucleus accumbens (NAc) in heroin dependence. We used weighted gene co-expression network analysis to analyze the GSE87823 data package, which included 27 heroin users and 22 controls of human NAc tissue. Modules were correlated with basic information of samples and enrichment analyses used to identify biological function and transcription factors and online tools were used to perform the gene ontology of significant genes. We identified one gene module from the total data (blue) and the male data (turquoise), respectively. The overlap genes of top 10 hub genes in significant modules (PRR11, SLC35E1, LPP, ZNF721, ZNF611, LRRFIP1) were selected to identify as candidate genes in the regulation mechanism of NAc in heroin dependence. Then, we accorded the results to further explore that miRNA-hsa-miR-155-5p in male and total may be a potential marker. The candidate genes may serve as novel prognostic markers and treatment targets. Hsa-miR-155-5p may be a promising regulatory point for the treatment of heroin addiction.


Subject(s)
Biomarkers/analysis , Computational Biology/methods , Gene Expression Regulation , Gene Regulatory Networks , Heroin Dependence/genetics , Nucleus Accumbens/metabolism , Gene Expression Profiling , Gene Ontology , Heroin Dependence/pathology , Humans , Male , MicroRNAs/genetics
3.
J Neurosci ; 38(22): 5182-5195, 2018 05 30.
Article in English | MEDLINE | ID: mdl-29760180

ABSTRACT

Previous studies have shown that individuals with heroin and cocaine addiction prefer to use these drugs in distinct settings: mostly at home in the case of heroin and mostly outside the home in the case of cocaine. Here we investigated whether the context would modulate the affective and neural responses to these drugs in a similar way. First, we used a novel emotional task to assess the affective state produced by heroin or cocaine in different settings, based on the recollections of male and female drug users. Then we used fMRI to monitor neural activity during drug imagery (re-creating the setting of drug use) in male drug users. Consistent with our working hypothesis, the majority of participants reported a shift in the affective valence of heroin from mostly pleasant at home to mostly unpleasant outside the home (p < 0.0001). The opposite shift was observed for cocaine; that is, most participants who found cocaine pleasant outside the home found it unpleasant when taken at home (p < 0.0014). Furthermore, we found a double dissociation, as a function of drug and setting imagery, in BOLD signal changes in the left PFC and caudate, and bilaterally in the cerebellum (all p values <0.01), suggesting that the fronto-striatal-cerebellar network is implicated in the contextualization of drug-induced affect. In summary, we report that the same setting can influence in opposite directions the affective and neural response to psychostimulants versus opiates in humans, adding to growing evidence of distinct substrates for the rewarding effects of these two drug classes.SIGNIFICANCE STATEMENT The rewarding effects of addictive drugs are often thought to depend on shared substrates. Yet, environmental influences can unmask striking differences between psychostimulants and opiates. Here we used emotional tasks and fMRI to explore the influence of setting on the response to heroin versus cocaine in individuals with addiction. Simply moving from one setting to another significantly decreased heroin pleasure but increased cocaine pleasure, and vice versa. Similar double dissociation was observed in the activity of the fronto-striatal-cerebellar network. These findings suggest that the effects of opiates and psychostimulants depend on dissociable psychological and neural substrates and that therapeutic approaches to addiction should take into account the peculiarities of different drug classes and the settings of drug use.


Subject(s)
Affect/drug effects , Cocaine-Related Disorders/pathology , Cocaine-Related Disorders/psychology , Environment , Heroin Dependence/pathology , Heroin Dependence/psychology , Neurons/drug effects , Adult , Brain Mapping , Caudate Nucleus/diagnostic imaging , Caudate Nucleus/drug effects , Cerebellum/diagnostic imaging , Cerebellum/drug effects , Corpus Striatum/diagnostic imaging , Corpus Striatum/physiology , Humans , Imagination/drug effects , Magnetic Resonance Imaging , Male , Middle Aged , Nerve Net/diagnostic imaging , Nerve Net/drug effects , Photic Stimulation , Prefrontal Cortex/diagnostic imaging , Prefrontal Cortex/drug effects , Social Environment
4.
Eur Arch Psychiatry Clin Neurosci ; 269(3): 317-324, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30173319

ABSTRACT

Deep brain stimulation (DBS) of the globus pallidus internus was recently proposed as a potential new treatment target for opioid addiction. DBS requires computer-assisted-3D planning to implant the stimulation electrode precisely. As volumes of brain regions may differ in addiction compared to healthy controls, our aim was to investigate possible volume differences in addicts compared to healthy controls. Volumes of the globus pallidus externus (PE) and internus (PI) in heroin addicts (n = 14) and healthy controls (n = 12) were assessed using morphometry of serial whole-brain sections. Total brain volume was larger in the heroin group (mean 1479 ± 62 cm3 vs. mean 1352 ± 103 cm3), as the heroin group was more than 10 years younger (p = 0.001). Despite larger mean whole brain volume, the mean relative volume of the PE and PI was smaller in addicted subjects compared to healthy controls (PE 0.658 ± 0.183 × 10-3 vs. 0.901 ± 0.284 × 10-3; ANOVA F(1, 24) = 6.945, p = 0.014, η2 = 0.224; PI 0.253 ± 0.095 × 10-3 vs. 0.345 ± 0.107 × 10-3; ANOVA F(1, 24) = 5.374, p = 0.029, η2 = 0.183). These findings were not significantly confounded by age, duration of autolysis, and fixation time. Our results provide further evidence for structural and not only functional deficits of the globus pallidus in addiction. In the context of previous studies, our findings support the idea of shared pathophysiological processes between comorbid depression and impulsivity in opioid addiction.


Subject(s)
Globus Pallidus/pathology , Heroin Dependence/pathology , Adult , Autopsy , Humans , Male , Middle Aged , Young Adult
5.
PLoS Genet ; 12(3): e1005910, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27010727

ABSTRACT

Methadone maintenance treatment (MMT) is commonly used for controlling opioid dependence, preventing withdrawal symptoms, and improving the quality of life of heroin-dependent patients. A steady-state plasma concentration of methadone enantiomers, a measure of methadone metabolism, is an index of treatment response and efficacy of MMT. Although the methadone metabolism pathway has been partially revealed, no genome-wide pharmacogenomic study has been performed to identify genetic determinants and characterize genetic mechanisms for the plasma concentrations of methadone R- and S-enantiomers. This study was the first genome-wide pharmacogenomic study to identify genes associated with the plasma concentrations of methadone R- and S-enantiomers and their respective metabolites in a methadone maintenance cohort. After data quality control was ensured, a dataset of 344 heroin-dependent patients in the Han Chinese population of Taiwan who underwent MMT was analyzed. Genome-wide single-locus and haplotype-based association tests were performed to analyze four quantitative traits: the plasma concentrations of methadone R- and S-enantiomers and their respective metabolites. A significant single nucleotide polymorphism (SNP), rs17180299 (raw p = 2.24 × 10(-8)), was identified, accounting for 9.541% of the variation in the plasma concentration of the methadone R-enantiomer. In addition, 17 haplotypes were identified on SPON1, GSG1L, and CYP450 genes associated with the plasma concentration of methadone S-enantiomer. These haplotypes accounted for approximately one-fourth of the variation of the overall S-methadone plasma concentration. The association between the S-methadone plasma concentration and CYP2B6, SPON1, and GSG1L were replicated in another independent study. A gene expression experiment revealed that CYP2B6, SPON1, and GSG1L can be activated concomitantly through a constitutive androstane receptor (CAR) activation pathway. In conclusion, this study revealed new genes associated with the plasma concentration of methadone, providing insight into the genetic foundation of methadone metabolism. The results can be applied to predict treatment responses and methadone-related deaths for individualized MMTs.


Subject(s)
Cytochrome P-450 CYP2B6/genetics , Extracellular Matrix Proteins/genetics , Heroin Dependence/genetics , Methadone/administration & dosage , Adult , Androstanes/metabolism , Female , Genome-Wide Association Study , Haplotypes/genetics , Heroin/metabolism , Heroin/toxicity , Heroin Dependence/metabolism , Heroin Dependence/pathology , Humans , Male , Methadone/metabolism , Middle Aged , Opiate Substitution Treatment , Pharmacogenetics , Polymorphism, Single Nucleotide , Stereoisomerism
6.
Eur Arch Psychiatry Clin Neurosci ; 268(3): 243-248, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28534187

ABSTRACT

The hypothalamus is at the core of the stress responses systems of the brain. Most interestingly, even though changes of HPA-function have been observed in opiate addiction not much is known about structural changes of the hypothalamus. Volumes of hypothalamus in heroin addicts (n = 14) and healthy controls (n = 12) were assessed by using morphometry of serial whole-brain sections. Total brain volume was larger in the heroin group (mean 1478.85 ± 62.34 cm3 vs. mean 1352.38 ± 103.24 cm3), as the heroin group was more than 10 years younger (p = 0.001). Thus, diagnosis-related effects in the hypothalamus were assessed using the hypothalamus volume relative to whole brain volume showing reduced volumes of the hypothalamus in the heroin group (0.201 ± 0.074 × 10-3 vs. 0.267 ± 0.048 × 10-3; ANOVA: F(1,23) = 6.211, p = 0.020) with a strong hemispheric effect (left side: about 20% reduction 0.209 ± 0.080 × 10-3 vs. 0.264 ± 0.049 × 10-3; F = 4.109; p = 0.054; right side: about 27% reduction, 0.198 ± 0.069 × 10-3 vs. 0.271 ± 0.050 × 10-3; F = -8.800; p = 0.007). Our results provide further evidence for structural and not only functional deficits of the hypothalamus in addiction.


Subject(s)
Diagnosis , Heroin Dependence/pathology , Hypothalamus/pathology , Adult , Autopsy , Follow-Up Studies , Functional Laterality , Humans , Male , Middle Aged , Statistics, Nonparametric , Young Adult
7.
Psychiatry Clin Neurosci ; 72(6): 435-444, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29582514

ABSTRACT

AIM: Chronic heroin use can cause various neuropathological characteristics that may compromise brain function. The present study evaluated the alteration of gray matter volume (GMV) and its resting-state functional connectivity (rsFC) over the dorsolateral prefrontal cortex (DLPFC) among male heroin users. METHODS: Thirty heroin-dependent men undergoing methadone maintenance therapy and 30 educational-level- and age-matched male controls were recruited for this study. To assess their GMV and rsFC, the participants were evaluated using spoiled gradient echo and gradient-recalled echo planar imaging sequences with a 3-Tesla General Electric MR scanner under resting state. RESULTS: The heroin-dependent men showed lower GMV over the right DLPFC in comparison with the controls. Further evaluation of the rsFC of the right DLPFC revealed a marked decrease in interhemispheric DLPFC connectivity among those with heroin dependence under control of head movement and GMV of the right DLPFC. CONCLUSION: Although the mechanism remains unclear, the present study shows that chronic heroin use is associated with alteration of morphology as well as rsFC over the right DLPFC. As the DLPFC plays an imperative role in various domains of cognitive function, service providers for heroin users should consider the impacts of possible DLPFC-related cognitive deficits on treatment effectiveness.


Subject(s)
Connectome/methods , Gray Matter/pathology , Heroin Dependence/pathology , Heroin Dependence/physiopathology , Prefrontal Cortex/physiopathology , Adult , Gray Matter/diagnostic imaging , Heroin Dependence/diagnostic imaging , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Prefrontal Cortex/diagnostic imaging
8.
Brain ; 138(Pt 1): 217-28, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25414039

ABSTRACT

Recent evidence has shown that a single maintenance dose of heroin attenuates psychophysiological stress responses in heroin-dependent patients, probably reflecting the effectiveness of heroin-assisted therapies for the treatment of severe heroin addiction. However, the underlying neural circuitry of these effects has not yet been investigated. Using a cross-over, double-blind, vehicle-controlled design, 22 heroin-dependent and heroin-maintained outpatients from the Centre of Substance Use Disorders at the University Hospital of Psychiatry in Basel were studied after heroin and placebo administration, while 17 healthy controls from the general population were included for placebo administration only. Functional magnetic resonance imaging was used to detect brain responses to fearful faces and dynamic causal modelling was applied to compute fear-induced modulation of connectivity within the emotional face network. Stress responses were assessed by hormone releases and subjective ratings. Relative to placebo, heroin acutely reduced the fear-induced modulation of connectivity from the left fusiform gyrus to the left amygdala and from the right amygdala to the right orbitofrontal cortex in dependent patients. Both of these amygdala-related connectivity strengths were significantly increased in patients after placebo treatment (acute withdrawal) compared to healthy controls, whose connectivity estimates did not differ from those of patients after heroin injection. Moreover, we found positive correlations between the left fusiform gyrus to amygdala connectivity and different stress responses, as well as between the right amygdala to orbitofrontal cortex connectivity and levels of craving. Our findings indicate that the increased amygdala-related connectivity during fearful face processing after the placebo treatment in heroin-dependent patients transiently normalizes after acute heroin maintenance treatment. Furthermore, this study suggests that the assessment of amygdala-related connectivity during fear processing may provide a prognostic tool to assess stress levels in heroin-dependent patients and to quantify the efficacy of maintenance treatments in drug addiction.


Subject(s)
Analgesics, Opioid/therapeutic use , Heroin Dependence/drug therapy , Heroin/therapeutic use , Stress, Psychological/pathology , Adrenocorticotropic Hormone/blood , Adult , Amygdala/pathology , Bayes Theorem , Brain/drug effects , Brain/pathology , Double-Blind Method , Fear/drug effects , Fear/psychology , Female , Heroin Dependence/pathology , Heroin Dependence/psychology , Humans , Male , Middle Aged , Models, Neurological , Stress, Psychological/blood , Stress, Psychological/etiology , Surveys and Questionnaires , Time Factors
9.
J Neurosci ; 34(16): 5649-57, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24741055

ABSTRACT

Reducing the enduring vulnerability to relapse is a therapeutic goal in treating drug addiction. Studies with animal models of drug addiction show a marked increase in extrasynaptic glutamate in the core subcompartment of the nucleus accumbens (NAcore) during reinstated drug seeking. However, the synaptic mechanisms linking drug-induced changes in extrasynaptic glutamate to relapse are poorly understood. Here, we discovered impaired glutamate elimination in rats extinguished from heroin self-administration that leads to spillover of synaptically released glutamate into the nonsynaptic extracellular space in NAcore and investigated whether restoration of glutamate transport prevented reinstated heroin seeking. Through multiple functional assays of glutamate uptake and analyzing NMDA receptor-mediated currents, we show that heroin self-administration produced long-lasting downregulation of glutamate uptake and surface expression of the transporter GLT-1. This downregulation was associated with spillover of synaptic glutamate to extrasynaptic NMDA receptors within the NAcore. Ceftriaxone restored glutamate uptake and prevented synaptic glutamate spillover and cue-induced heroin seeking. Ceftriaxone-induced inhibition of reinstated heroin seeking was blocked by morpholino-antisense targeting GLT-1 synthesis. These data reveal that the synaptic glutamate spillover in the NAcore results from reduced glutamate transport and is a critical pathophysiological mechanism underling reinstated drug seeking in rats extinguished from heroin self-administration.


Subject(s)
Drug-Seeking Behavior/physiology , Glutamic Acid/metabolism , Heroin Dependence/metabolism , Heroin Dependence/prevention & control , Heroin/administration & dosage , Synapses/metabolism , Animals , Aspartic Acid/pharmacology , Ceftriaxone/pharmacology , Conditioning, Operant/drug effects , Disease Models, Animal , Down-Regulation/drug effects , Drug-Seeking Behavior/drug effects , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Heroin/adverse effects , Heroin Dependence/etiology , Heroin Dependence/pathology , In Vitro Techniques , Male , Morpholinos/pharmacology , Neurons/drug effects , Neurons/physiology , Nucleus Accumbens/cytology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Potassium/pharmacology , Rats , Rats, Sprague-Dawley , Secondary Prevention , Synapses/drug effects
10.
Neuroimage ; 115: 76-84, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25944613

ABSTRACT

Advanced neuroimaging studies have identified brain correlates of pathological impulsivity in a variety of neuropsychiatric disorders. However, whether and how these spatially separate and functionally integrated neural correlates collectively contribute to aberrant impulsive behaviors remains unclear. Building on recent progress in neuroeconomics toward determining a biological account of human behaviors, we employed resting-state functional MRI to characterize the nature of the links between these neural correlates and to investigate their impact on impulsivity. We demonstrated that through functional connectivity with the ventral medial prefrontal cortex, the δ-network (regions of the executive control system, such as the dorsolateral prefrontal cortex) and the ß-network (regions of the reward system involved in the mesocorticolimbic pathway), jointly influence impulsivity measured by the Barratt impulsiveness scale scores. In control nondrug-using subjects, the functional link between the ß- and δ-networks is balanced, and the δ-network competitively controls impulsivity. However, in abstinent heroin-dependent subjects, the link is imbalanced, with stronger ß-network connectivity and weaker δ-network connectivity. The imbalanced link is associated with impulsivity, indicating that the ß- and δ-networks may mutually reinforce each other in abstinent heroin-dependent subjects. These findings of an aberrant link between the ß- and δ-networks in abstinent heroin-dependent subjects may shed light on the mechanism of aberrant behaviors of drug addiction and may serve as an endophenotype to mark individual subjects' self-control capacity.


Subject(s)
Heroin Dependence/pathology , Heroin Dependence/psychology , Impulsive Behavior , Nerve Net/pathology , Neural Pathways/pathology , Adult , Executive Function , Female , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Prefrontal Cortex/pathology
11.
Hum Brain Mapp ; 36(12): 5287-300, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26441146

ABSTRACT

Heroin addiction is a severe relapsing brain disorder associated with impaired cognitive control, including deficits in attention allocation. The thalamus has a high density of opiate receptors and is critically involved in orchestrating cortical activity during cognitive control. However, there have been no studies on how acute heroin treatment modulates thalamic activity. In a cross-over, double-blind, vehicle-controlled study, 29 heroin-maintained outpatients were studied after heroin and placebo administration, while 20 healthy controls were included for the placebo condition only. Resting-state functional magnetic resonance imaging was used to analyze functional integration of the thalamus by three different resting state analysis techniques. Thalamocortical functional connectivity (FC) was analyzed by seed-based correlation, while intrinsic thalamic oscillation was assessed by analysis of regional homogeneity (ReHo) and the fractional amplitude of low frequency fluctuations (fALFF). Relative to the placebo treatment and healthy controls, acute heroin administration reduced thalamocortical FC to cortical regions, including the frontal cortex, while the reductions in FC to the mediofrontal cortex, orbitofrontal cortex, and frontal pole were positively correlated with the plasma level of morphine, the main psychoactive metabolite of heroin. Furthermore, heroin treatment was associated with increased thalamic ReHo and fALFF values, whereas fALFF following heroin exposure correlated negatively with scores of attentional control. The heroin-associated increase in fALFF was mainly dominated by slow-4 (0.027-0.073 Hz) oscillations. Our findings show that there are acute effects of heroin within the thalamocortical system and may shed new light on the role of the thalamus in cognitive control in heroin addiction. Future research is needed to determine the underlying physiological mechanisms and their role in heroin addiction.


Subject(s)
Cerebral Cortex/pathology , Heroin Dependence/drug therapy , Heroin/therapeutic use , Narcotics/therapeutic use , Thalamus/drug effects , Thalamus/pathology , Adult , Cerebral Cortex/drug effects , Cross-Over Studies , Double-Blind Method , Echo-Planar Imaging , Female , Functional Laterality/drug effects , Functional Laterality/physiology , Heroin/blood , Heroin Dependence/blood , Heroin Dependence/pathology , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Middle Aged , Neural Pathways/drug effects , Neural Pathways/pathology , Outpatients , Oxygen/blood , Psychiatric Status Rating Scales , Statistics as Topic , Thalamus/blood supply , Young Adult
12.
J Neurosci Res ; 93(11): 1693-702, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26280556

ABSTRACT

Functional neuroimaging studies suggest that abnormal brain functional connectivity may be the neural underpinning of addiction to illicit drugs and of relapse after successful cessation therapy. Aberrant brain networks have been demonstrated in addicted patients and in newly abstinent addicts. However, it is not known whether abnormal brain connectivity patterns persist after prolonged abstinence. In this cross-sectional study, whole-brain resting-state functional magnetic resonance images (8 min) were collected from 30 heroin-addicted individuals after a long period of abstinence (more than 3 years) and from 30 healthy controls. We first examined the group differences in the resting-state functional connectivity of the nucleus accumbens (NAc), a brain region implicated in relapse-related processes, including craving and reactivity to stress following acute and protracted withdrawal from heroin. We then examined the relation between the duration of abstinence and the altered NAc functional connectivity in the heroin group. We found that, compared with controls, heroin-dependent participants exhibited significantly greater functional connectivity between the right ventromedial prefrontal cortex and the NAc and weaker functional connectivity between the NAc and the left putamen, left precuneus, and supplementary motor area. However, with longer abstinence time, the strength of NAc functional connectivity with the left putamen increased. These results indicate that dysfunction of the NAc functional network is still present in long-term-abstinent heroin-dependent individuals.


Subject(s)
Heroin Dependence/pathology , Neural Pathways/pathology , Nucleus Accumbens/pathology , Adult , Cross-Sectional Studies , Female , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male
13.
J Cutan Pathol ; 42(11): 815-23, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26345172

ABSTRACT

BACKGROUND: The rate of intravenous drug use (IVDU) has been increasing nationally; however, cutaneous manifestations of IVDU have infrequently been investigated. We report a series of the clinicopathological correlation of IVDU in the skin. METHODS: A search of surgical pathology files between the years 2000 and 2014 was performed for cutaneous specimens from patients with a reported history of IVDU for which the histopathological findings could not be attributed to another etiology. Ten cases for which slides were available were included in the study. RESULTS: Patients had an average age of 39.7 years and had active or recent history of IVDU. Clinical impressions included ulcer, granulomatous dermatitis, vasculitis, pyoderma gangrenosum and hyperpigmentation at injection sites. Histopathology revealed leukocytoclastic vasculitis (n = 2), dermal pigment deposition (n = 3), non-specific ulceration/scarring (n = 4) and necrobiosis lipoidica-like dermatitis (n = 1). No infectious etiology or polarizable foreign material was identified in any case. CONCLUSIONS: Cutaneous manifestations of IVDU should be considered in the differential as an etiology for dermatopathologic findings in high-risk patients. We report histopathological findings beyond the scope of those most commonly associated with IVDU. We aim to raise awareness of the cutaneous manifestations of IVDU to improve clinicopathological correlation and patient management in light of the ongoing epidemic.


Subject(s)
Skin Diseases/chemically induced , Skin Diseases/pathology , Substance Abuse, Intravenous/pathology , Adult , Female , Heroin Dependence/pathology , Humans , Male , Middle Aged , Pregnancy , Skin Pigmentation/drug effects
14.
Eur Arch Psychiatry Clin Neurosci ; 265(8): 647-53, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26189034

ABSTRACT

Deep brain stimulation (DBS) of the nucleus accumbens (NAc) is increasingly investigated in neuropsychiatric disorders. DBS requires computer-assisted 3D planning to implant the stimulation electrode precisely. Recently, there has been a debate about the true dimensions of NAc in healthy as well as in mentally ill individuals. Knowing its true dimensions in different neuropsychiatric disorders may improve even more precise targeting of NAc for therapeutic DBS. Volumes of NAc of heroin addicts (n = 14) and healthy controls (n = 12) were calculated by using morphometry of serial whole-brain sections. Total brain volume was larger in the heroin group (mean 1478.85 ± 62.34 vs. mean 1352.38 ± 103.24 cm(3)), as the heroin group was more than 10 years younger (p = 0.001). However, the mean volume of the NAc in heroin addicts was smaller than in controls (0.528 ± 0.166 vs. 0.623 ± 0.196 cm(3); p = 0.019). This group effect did not significantly differ between the hemispheres. When assessed separately, left-hemispheric NAc volume was 15 % lower (p = 0.020), while right-hemispheric NAc volume was 16 % lower (p = 0.047) in the heroin-addicted group compared to controls. Based on these diagnosis-related differences, we believe it is important to further analyze NAc volumes in different psychiatric disorders to further improve precise targeting and electrode placement.


Subject(s)
Abducens Nucleus/pathology , Diagnosis , Heroin Dependence/pathology , Adult , Analysis of Variance , Functional Laterality , Humans , Imaging, Three-Dimensional , Magnetic Resonance Imaging , Male , Middle Aged , Young Adult
15.
Eur Arch Psychiatry Clin Neurosci ; 265(8): 637-45, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25467383

ABSTRACT

The neural mechanisms of heroin addiction are still incompletely understood, even though modern neuroimaging techniques offer insights into disease-related changes in vivo. While changes on cortical structure have been reported in heroin addiction, evidence from subcortical areas remains underrepresented. Functional imaging studies revealed that the brain reward system and particularly the nucleus accumbens (NAcc) play a pivotal role in the pathophysiology of drug addiction. The aim of this study was to investigate whether there was a volume difference of the NAcc in heroin addiction in comparison to healthy controls. A further aim was to correlate subcortical volumes with clinical measurements on negative affects in addiction. Thirty heroin-dependent patients under maintenance treatment with diacetylmorphine and twenty healthy controls underwent structural MRI scanning at 3T. Subcortical segmentation analysis was performed using FMRIB's Integrated Registration and Segmentation Tool function of FSL. The State-Trait Anxiety Inventory and the Beck Depression Inventory were used to assess trait anxiety and depressive symptoms, respectively. A decreased volume of the left NAcc was observed in heroin-dependent patients compared to healthy controls. Depression score was negatively correlated with left NAcc volume in patients, whereas a positive correlation was found between the daily opioid dose and the volume of the right amygdala. This study indicates that there might be structural differences of the NAcc in heroin-dependent patients in comparison with healthy controls. Furthermore, correlations of subcortical structures with negative emotions and opioid doses might be of future relevance for the investigation of heroin addiction.


Subject(s)
Heroin Dependence/pathology , Nucleus Accumbens/pathology , Adult , Analysis of Variance , Cross-Over Studies , Female , Humans , Imaging, Three-Dimensional , Magnetic Resonance Imaging , Male , Middle Aged , Psychiatric Status Rating Scales , Statistics as Topic , Switzerland
16.
Addict Biol ; 20(3): 570-9, 2015 May.
Article in English | MEDLINE | ID: mdl-24720731

ABSTRACT

The compulsion to seek and use heroin is frequently driven by stress and craving during drug-cue exposure. Although previous neuroimaging studies have indicated that craving is mediated by increased prefrontal cortex activity, it remains unknown how heroin administration modulates the prefrontal cortex response. This study examines the acute effects of heroin on brain function in heroin-maintained patients. Using a crossover, double-blind, placebo-controlled design, 27 heroin-maintained patients performed functional magnetic resonance imaging 20 minutes after the administration of heroin or placebo (saline) while drug-related and neutral stimuli were presented. Images were processed and analysed with statistical parametric mapping. Plasma concentrations of heroin and its main metabolites were assessed using high-performance liquid chromatography. Region of interest analyses showed a drug-related cue-associated blood-oxygen-level-dependent activation in the orbitofrontal cortex (OFC) in heroin-dependent patients during both treatment conditions (heroin and placebo). This activation of the OFC was significantly higher after heroin than after placebo administration. These findings may indicate the importance of OFC activity for impulse control and decision-making after regular heroin administration and may emphasize the benefit of the heroin-assisted treatment in heroin dependence.


Subject(s)
Heroin Dependence/physiopathology , Heroin/pharmacology , Narcotics/pharmacology , Prefrontal Cortex/drug effects , Adult , Brain Diseases/pathology , Brain Diseases/physiopathology , Craving/drug effects , Cues , Female , Gray Matter/drug effects , Gray Matter/pathology , Heroin/administration & dosage , Heroin/pharmacokinetics , Heroin Dependence/pathology , Humans , Magnetic Resonance Imaging , Male , Narcotics/administration & dosage , Narcotics/pharmacokinetics , Neuropsychological Tests , Organ Size , Prefrontal Cortex/pathology
17.
Neuroimage ; 88: 295-307, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24140937

ABSTRACT

Accumulating evidence from brain structural imaging studies on heroin dependence has supported links between brain morphological alterations and heroin exposure, particularly in gray matter volume or gray matter density. However, the effects of heroin exposure on cortical thickness and the relationship between cortical thickness and heroin addiction are not yet known. In this study, we acquired 3D high-resolution brain structural magnetic resonance imaging (MRI) data from 18 heroin-dependent individuals (HDIs) and 15 healthy controls (HCs). Using FreeSurfer, we detected abnormalities in cortical thickness in the HDIs. Based on a vertex-wise analysis, the HDIs showed significantly decreased cortical thickness in the bilateral superior frontal, left caudal middle frontal, right superior temporal, and right insular regions compared to the HCs but significantly increased cortical thickness in the left superior parietal, bilateral lingual, left temporal pole, right inferior parietal, right lateral occipital, and right cuneus regions. To supplement these results, a subsequent ROI-wise analysis was performed and showed decreased cortical thickness in the left superior frontal sulcus, left precuneus gyrus, left calcarine sulcus, left anterior transverse collateral sulcus, and the right medial occipital-temporal and lingual sulcus. These regions partially overlapped with the areas identified using the vertex-wise analysis. In addition, we found that the thickness in the right superior frontal and right insular regions was negatively correlated with the duration of heroin use. These results provide compelling evidence for cortical abnormality in HDIs and also suggest that the duration of heroin use may be a critical factor associated with the brain alteration.


Subject(s)
Cerebral Cortex/pathology , Heroin Dependence/pathology , Adult , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged
18.
Int J Neuropsychopharmacol ; 17(9): 1375-85, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24641978

ABSTRACT

Neuroimaging studies have reported reduced activity in a broad network of brain regions during response inhibition in heroin-dependent patients. However, how heroin in an acute dose modulates the neural correlates of response inhibition and the underlying brain connectivity has not yet been investigated. In this double-blind placebo-controlled study, we used functional magnetic resonance imaging to examine whether acute heroin administration changed whole brain activity during response inhibition in 26 heroin-dependent patients. We then applied dynamic causal modelling to investigate the effect of an acute dose of heroin on the functional interactions between the dorsal anterior cingulate cortex (dACC) and the bilateral inferior frontal gyri (IFG). Heroin acutely reduced dACC activity, as well as the inhibition-induced modulation of connectivity from the dACC to the right IFG compared with placebo. Furthermore, dACC activity was positively related to false alarm rates after placebo but not heroin administration. These results suggest that acute heroin administration impairs cognitive control in dependent patients by reducing the activity in the dACC activity and the functional connectivity from the dACC to the right IFG.


Subject(s)
Analgesics, Opioid/administration & dosage , Cognition/drug effects , Heroin Dependence/drug therapy , Heroin Dependence/pathology , Heroin/administration & dosage , Prefrontal Cortex/drug effects , Adult , Bayes Theorem , Cross-Sectional Studies , Double-Blind Method , Female , Humans , Image Processing, Computer-Assisted , Inhibition, Psychological , Magnetic Resonance Imaging , Male , Middle Aged , Models, Neurological , Neural Pathways/blood supply , Neural Pathways/drug effects , Neuropsychological Tests , Oxygen/blood , Prefrontal Cortex/blood supply
19.
J Neurosci ; 32(34): 11600-9, 2012 Aug 22.
Article in English | MEDLINE | ID: mdl-22915104

ABSTRACT

In humans, exposure to cues previously associated with heroin use often provokes relapse after prolonged withdrawal periods. In rats, cue-induced heroin seeking progressively increases after withdrawal (incubation of heroin craving). Here, we examined the role of orbitofrontal cortex (OFC) neuronal ensembles in the enhanced response to heroin cues after prolonged withdrawal or the expression of incubation of heroin craving. We trained rats to self-administer heroin (6 h/d for 10 d) and assessed cue-induced heroin seeking in extinction tests after 1 or 14 withdrawal days. Cue-induced heroin seeking increased from 1 to 14 d and was accompanied by increased Fos expression in ∼12% of OFC neurons. Nonselective inactivation of OFC neurons with the GABA agonists baclofen + muscimol decreased cue-induced heroin seeking on withdrawal day 14 but not day 1. We then used the Daun02 inactivation procedure to assess a causal role of the minority of selectively activated Fos-expressing OFC neurons (that presumably form cue-encoding neuronal ensembles) in cue-induced heroin seeking after 14 withdrawal days. We trained c-fos-lacZ transgenic rats to self-administer heroin and 11 d later reexposed them to heroin-associated cues or novel cues for 15 min (induction day), followed by OFC Daun02 or vehicle injections 90 min later; we then tested the rats in extinction tests 3 d later. Daun02 selectively decreased cue-induced heroin seeking in rats previously reexposed to the heroin-associated cues on induction day but not in rats exposed previously to novel cues. Results suggest that heroin-cue-activated OFC neuronal ensembles contribute to the expression of incubation of heroin craving.


Subject(s)
Conditioning, Operant/drug effects , Cues , Heroin Dependence , Heroin/administration & dosage , Neurons/drug effects , Prefrontal Cortex/cytology , Animals , Baclofen/pharmacology , Behavior, Animal , Buprenorphine/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Daunorubicin/analogs & derivatives , Daunorubicin/pharmacology , Extinction, Psychological/drug effects , GABA-B Receptor Agonists/pharmacology , Gene Expression Regulation/drug effects , Glutamate Decarboxylase/metabolism , Heroin Dependence/pathology , Heroin Dependence/physiopathology , Heroin Dependence/psychology , In Vitro Techniques , Male , Muscimol/pharmacology , Narcotic Antagonists/pharmacology , Neurons/physiology , Oncogene Proteins v-fos/metabolism , Phosphopyruvate Hydratase/metabolism , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley , Self Administration
20.
Eur Arch Psychiatry Clin Neurosci ; 263(3): 197-203, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22569752

ABSTRACT

Experimental social neuroscience has shown that being socially excluded is processed in the anterior cingulate cortex (ACC). We hypothesize that a chronic form of social exclusion resembling one aspect of social stigmatization is associated with altered neural plasticity reflected by neurometabolic alterations in the ACC. To test this hypothesis, a highly stigmatized patient group of heroin addicts (N = 15) during opiate maintenance therapy rated a questionnaire about being stigmatized, and neurometabolic markers in the ACC were determined using (1)H MR spectroscopy. We found a negative correlation between discrimination experience and N-acetylaspartate (NAA), indicating attenuated neuron functioning in the anterior cingulate cortex in those patients reporting high discrimination experience. Furthermore, perceived stigmatization showed an association with anxiety that was mediated by NAA. Although the correlative analysis cannot give evidence for a causal relationship, the relation of NAA in the ACC and discrimination experience indicates a malfunction of the neural system involved in cognitive control over emotionally relevant social stimuli in discrimination reporting heroin addicts. Further research is needed to elucidate factors associated with chronic stigmatization.


Subject(s)
Gyrus Cinguli/metabolism , Heroin Dependence , Social Discrimination/psychology , Social Stigma , Adult , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Female , Gyrus Cinguli/pathology , Heroin Dependence/complications , Heroin Dependence/pathology , Heroin Dependence/psychology , Humans , Linear Models , Magnetic Resonance Spectroscopy , Male , Middle Aged , Pilot Projects , Surveys and Questionnaires , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL