Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 266
Filter
1.
Eur J Neurosci ; 59(10): 2502-2521, 2024 May.
Article in English | MEDLINE | ID: mdl-38650303

ABSTRACT

The emergence of compulsive drug-seeking habits, a hallmark feature of substance use disorder, has been shown to be predicated on the engagement of dorsolateral striatal control over behaviour. This process involves the dopamine-dependent functional coupling of the anterior dorsolateral striatum (aDLS) with the nucleus accumbens core, but the mechanisms by which this coupling occurs have not been fully elucidated. The striatum is tiled by a syncytium of astrocytes that express the dopamine transporter (DAT), the level of which is altered in individuals with heroin use disorder. Astrocytes are therefore uniquely placed functionally to bridge dopamine-dependent mechanisms across the striatum. Here we tested the hypothesis that exposure to heroin influences the expression of DAT in striatal astrocytes across the striatum before the development of DLS-dependent incentive heroin seeking habits. Using Western-blot, qPCR, and RNAscope™, we measured DAT protein and mRNA levels in whole tissue, culture and in situ astrocytes from striatal territories of rats with a well-established cue-controlled heroin seeking habit and rats trained to respond for heroin or food under continuous reinforcement. Incentive heroin seeking habits were associated with a reduction in DAT protein levels in the anterior aDLS that was preceded by a heroin-induced reduction in DAT mRNA and protein in astrocytes across the striatum. Striatal astrocytes were also shown to be susceptible to direct dopamine- and opioid-induced downregulation of DAT expression. These results suggest that astrocytes may critically regulate the striatal dopaminergic adaptations that lead to the development of incentive heroin seeking habits.


Subject(s)
Astrocytes , Corpus Striatum , Dopamine Plasma Membrane Transport Proteins , Dopamine , Drug-Seeking Behavior , Heroin , Animals , Dopamine Plasma Membrane Transport Proteins/metabolism , Astrocytes/metabolism , Astrocytes/drug effects , Corpus Striatum/metabolism , Corpus Striatum/drug effects , Male , Rats , Drug-Seeking Behavior/physiology , Drug-Seeking Behavior/drug effects , Heroin/pharmacology , Heroin/administration & dosage , Dopamine/metabolism , Motivation/drug effects , Motivation/physiology , Heroin Dependence/metabolism , Rats, Sprague-Dawley
2.
Neuroreport ; 35(7): 486-498, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38526939

ABSTRACT

This study aimed to investigate the effects of SIRT1 modulation on heroin addiction-like behavior and its possible biological mechanisms. Wild-type C57BL/6J and Sirt1loxp/loxp D1-Cre mice were used in this experiment, and Sirt1 loxp/loxp D1-Cre(-) mice were used as a control for conditional knockout mice. Mice were divided into saline control and heroin-dependent groups. Behavioral methods were used to record the withdrawal response, conditioned place preference (CPP) changes, and open field test results. Transmission electron microscopy (TEM) was used to observe the structure of autophagosomes in nucleus accumbens (NAc) neurons. The expression of SIRT1 and autophagy-related proteins and genes, such as LC3Ⅱ, ATG5 , and ATG7 , was detected in the NAc of each mouse group via western blot, real-time quantitative PCR (qPCR) analyzes, and immunofluorescence. The results of this experiment showed that compared with the saline group, mice in the wild-type heroin-dependent group showed marked withdrawal symptoms, with more autophagosomes observed in NAc via TEM. Compared with wild-type and Sirt1loxp/loxp D1-Cre(-) heroin-dependent groups, CPP formation was found to be reduced in the conditional knockout mouse group, with a significant decrease in spontaneous activity. Western blot, qPCR, and immunofluorescence results indicated that the expression of LC3Ⅱ, ATG-5, and ATG-7 was significantly reduced in the NAc of the Sirt1loxp/loxp D1-Cre(+) group. It was still, however, higher than that in the saline control group. These results suggest that inhibition of Sirt1 expression may prevent heroin-induced addiction-related behaviors via reducing D1 neuronal autophagy.


Subject(s)
Heroin Dependence , Nucleus Accumbens , Mice , Animals , Heroin Dependence/metabolism , Heroin , Sirtuin 1/metabolism , Mice, Inbred C57BL , Mice, Knockout , Autophagy
3.
Acta Pharmacol Sin ; 45(5): 945-958, 2024 May.
Article in English | MEDLINE | ID: mdl-38326624

ABSTRACT

Glutamatergic neurons in ventral pallidum (VPGlu) were recently reported to mediate motivational and emotional behavior, but its role in opioid addiction still remains to be elucidated. In this study we investigated the function of VPGlu in the context-dependent heroin taking and seeking behavior in male rats under the ABA renewal paradigm. By use of cell-type-specific fiber photometry, we showed that the calcium activity of VPGlu were inhibited during heroin self-administration and context-induced relapse, but activated after extinction in a new context. The drug seeking behavior was accompanied by the decreased calcium signal of VPGlu. Chemogenetic manipulation of VPGlu bidirectionally regulated heroin taking and seeking behavior. Anterograde tracing showed that the lateral habenula, one of the epithalamic structures, was the major output region of VPGlu, and its neuronal activity was consistent with VPGlu in different phases of heroin addiction and contributed to the motivation for heroin. VPGlu axon terminals in LHb exhibited dynamic activity in different phases of heroin addiction. Activation of VPGlu-LHb circuit reduced heroin seeking behavior during context-induced relapse. Furthermore, the balance of excitation/inhibition from VP to LHb was shifted to enhanced glutamate transmission after extinction of heroin seeking motivation. Overall, the present study demonstrated that the activity of VPGlu was involved in the regulation of heroin addiction and identified the VPGlu-LHb pathway as a potential intervention to reduce heroin seeking motivation.


Subject(s)
Basal Forebrain , Glutamic Acid , Heroin Dependence , Neurons , Rats, Sprague-Dawley , Animals , Male , Heroin Dependence/metabolism , Heroin Dependence/psychology , Basal Forebrain/metabolism , Glutamic Acid/metabolism , Neurons/metabolism , Drug-Seeking Behavior , Heroin , Rats , Self Administration , Habenula/metabolism
4.
Psychopharmacology (Berl) ; 241(6): 1265-1275, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38396195

ABSTRACT

There is a significant co-occurrence of opioid use disorder (OUD) and post-traumatic stress disorder (PTSD) in clinical populations. However, the neurobiological mechanisms linking chronic opioid use, withdrawal, and the development of PTSD are poorly understood. Our previous research has shown that proinflammatory cytokines, expressed primarily by astrocytes in the dorsal hippocampus (DH), play a role in the development of heroin withdrawal-enhanced fear learning (HW-EFL), an animal model of PTSD-OUD comorbidity. Given the role of astrocytes in memory, fear learning, and opioid use, our experiments aimed to investigate their involvement in HW-EFL. Experiment 1 examined the effect of withdrawal from chronic heroin administration on GFAP surface area and volume, and identified increased surface area and volume of GFAP immunoreactivity in the dentate gyrus (DG) following 24-hour heroin withdrawal. Experiment 2 examined astrocyte morphology and synaptic interactions at the 24-hour withdrawal timepoint using an astroglial membrane-bound GFP (AAV5-GfaABC1D-lck-GFP). Although we did not detect significant changes in surface area and volume of GfaABC1D-Lck-GFP labelled astrocytes, we did observe a significant increase in the colocalization of astrocyte membranes with PSD-95 (postsynaptic density protein 95) in the DG. Experiment 3 tested if stimulating astroglial Gi signaling in the DH alters HW-EFL, and our results demonstrate this manipulation attenuates HW-EFL. Collectively, these findings contribute to our current understanding of the effects of heroin withdrawal on astrocytes and support the involvement of astrocytes in the comorbid relationship between opioid use and anxiety disorders.


Subject(s)
Astrocytes , Fear , Heroin , Hippocampus , Substance Withdrawal Syndrome , Astrocytes/metabolism , Substance Withdrawal Syndrome/metabolism , Animals , Heroin/administration & dosage , Male , Hippocampus/metabolism , Fear/physiology , Stress Disorders, Post-Traumatic/metabolism , Learning/physiology , Disease Models, Animal , Heroin Dependence/metabolism , Glial Fibrillary Acidic Protein/metabolism , Mice
5.
Medicine (Baltimore) ; 101(33): e30093, 2022 Aug 19.
Article in English | MEDLINE | ID: mdl-35984180

ABSTRACT

Orexins regulate the reward-seeking pathway and also play a role in drug addiction. The aim of this study was an investigation of the changes in serum level of orexin-A as well as changes in the functional brain network in heroin use disorder (HUD) patients undergoing harm reduction therapy (HRT). Twenty-five HUD patients undergoing HRT that included methadone and buprenorphine, and 31 healthy control (HC) subjects, were enrolled for this study. Serum orexin-A levels and brain-derived neurotrophic factor were measured with assay kits. The functional brain network in HUD patients and HC was investigated and assessed using seed-based analysis and functional brain MRI scans. t Tested orexin-A levels were found to be significantly higher in HUD patients undergoing HRT than in HCs (P < .05). Analysis showed the functional activity of the right ventral anterior insula (RVAI) in HUD patients to be significantly lower than in HCs (P < .05, Family-Wise Error) corrected). In addition, the internetwork functional connectivity was significantly lower in the left nucleus accumbens and left dorsal anterior insula in the HUD subjects than in HCs (P < .05, Family-Wise Error corrected). In this study, no significant correlation between orexin-A levels and functional brain networks was found. However, the results suggest that HRT might increase orexin-A levels and decrease functional activity in RVAI in HUD patients.


Subject(s)
Harm Reduction , Heroin Dependence , Brain/diagnostic imaging , Buprenorphine/therapeutic use , Heroin , Heroin Dependence/drug therapy , Heroin Dependence/metabolism , Humans , Methadone/therapeutic use , Orexins
6.
Neurotox Res ; 40(4): 1070-1085, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35759084

ABSTRACT

Heroin is a highly addictive drug that causes axonal damage. Here, manganese-enhanced magnetic resonance imaging (MEMRI) was used to dynamically monitor axonal transport at different stages of heroin addiction. Rat models of heroin addiction (HA) and prolonged heroin addiction (PHA) were established by injecting rats with heroin at different stages. Heroin-induced learning and memory deficits were evaluated in the Morris water maze (MWM), and MEMRI was used to dynamically evaluate axonal transport in the olfactory pathway. The expression of proteins related to axonal structure and function was also assessed by Western blotting. Transmission electron microscopy (TEM) was used to observe ultrastructural changes, and protein levels of neurofilament heavy chain (NF-H) were analyzed by immunofluorescence staining. HA rats, especially PHA rats, exhibited worse spatial learning and memory than control rats. Compared with HA rats and control rats, PHA rats exhibited significantly longer escape latencies, significantly fewer platform-location crossings, and significantly more time in the target quadrant during the MWM test. Mn2+ transport was accelerated in HA rats. PHA rats exhibited severely reduced Mn2+ transport, and the axonal transport rate (ATR) was significantly lower in these rats than in control rats (P < 0.001). The levels of cytoplasmic dynein and kinesin-1 were significantly decreased in the PHA group than in the control group (P < 0.001); additionally, the levels of energy-related proteins, including cytochrome c oxidase (COX) IV and ATP synthase subunit beta (ATPB), were lower in the PHA group (P < 0.001). The brains of heroin-exposed rats displayed an abnormal ultrastructure, with neuronal apoptosis and mitochondrial dysfunction. Heroin exposure decreased the expression of NF-H, as indicated by significantly reduced staining intensities in tissues from HA and PHA rats (P < 0.05). MEMRI detected axonal transport dysfunction caused by long-term repeated exposure to heroin. The main causes of axonal transport impairment may be decreases in the levels of motor proteins and mitochondrial dysfunction. This study shows that MEMRI is a potential tool for visualizing axonal transport in individuals with drug addictions, providing a new way to evaluate addictive encephalopathy.


Subject(s)
Axonal Transport , Heroin Dependence , Animals , Axonal Transport/physiology , Brain/metabolism , Heroin/metabolism , Heroin/toxicity , Heroin Dependence/diagnostic imaging , Heroin Dependence/metabolism , Heroin Dependence/pathology , Kinesins , Magnetic Resonance Imaging/methods , Rats
7.
Life Sci ; 287: 120103, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34743944

ABSTRACT

BACKGROUND: Heroin is a semi-synthetic opioid that is commonly abused drugs in the world. It can cause hepatic injury and lead to multiple organs dysfunction to its addicts. Only a few reports exist on the metabolic changes and mechanisms in the liver of heroin-addicted mice with hepatic injury. METHODS: Twelve adult male Kunming mice (30-40 g) were divided into two groups randomly. The mice in the heroin-addicted group were injected subcutaneously in the first ten days with an increased dosage of heroin from 10 mg/kg to 55 mg/kg. The dosage was then stabilized at 55 mg/kg for three days. The control group was injected with the same amount of saline in the same manner. The hepatic injury was confirmed through the combination of histopathological observation and aminotransferase (AST) and alanine aminotransferase (ALT) determination. The withdrawal symptoms were recorded and used for assessment of heroin addiction. Eventually, liver metabolic biomarkers of heroin-addicted mice with hepatotoxicity were measured using UHPLC-MS/MS. RESULTS: Biochemical analysis and histopathological observation showed that heroin-addicted mice had a liver injury. The liver metabolites of heroin-addicted mice changed significantly. Metabonomics analysis revealed 41 metabolites in the liver of addicted heroin mice as biomarkers involving 34 metabolic pathways. Among them, glutathione metabolism, taurine and hypotaurine metabolism, vitamin B2 metabolism, riboflavin metabolism, and single-carbon metabolism pathways were markedly dispruted. CONCLUSIONS: Heroin damages the liver and disrupts the liver's metabolic pathways. Glutathione, taurine, riboflavin, 4-pyridoxate, folic acid, and methionine are important metabolic biomarkers, which may be key targets of heroin-induced liver damage. Thus, this study provides an in-depth understanding of the mechanisms of heroin-induced hepatotoxicity and potential biomarkers of liver damage.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Heroin Dependence/metabolism , Heroin/toxicity , Liver/metabolism , Metabolomics/methods , Phenotype , Animals , Animals, Outbred Strains , Biomarkers/blood , Biomarkers/metabolism , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/pathology , Heroin Dependence/blood , Heroin Dependence/pathology , Liver/drug effects , Liver/pathology , Male , Mice
8.
Addict Biol ; 26(5): e13013, 2021 09.
Article in English | MEDLINE | ID: mdl-33619816

ABSTRACT

Akt is initially identified as one of the downstream targets of phosphatidylinositol-3 kinase (PI3K) and is involved in morphine reward and tolerance. However, whether phospholyration of Akt (p-Akt) mediates heroin relapse remains unclear. Here, we aimed to explore the role of p-Akt in the nucleus accumbens (NAc) in cue-induced heroin-seeking behaviors after withdrawal. First, rats were trained to self-administer heroin for 14 days, after which we assessed heroin-seeking behaviors induced by a context cue (CC) or by discrete conditioned cues (CS) after 1 day or 14 days of withdrawal. We found that the active responses induced by CC or CS after 14 days of withdrawal were higher than those after 1 day of withdrawal. Meanwhile, the expression of p-Akt in the NAc was also greatest when rats were exposed to the CS after 14 days of withdrawal. Additionally, a microinjection of LY294002, an inhibitor of PI3K, into the NAc inhibited the CS-induced heroin-seeking behaviors after 14 days of withdrawal, paralleling the decreased levels of p-Akt in the NAc. Finally, Akt1 or ß-arrestin 2 was downregulated via a lentiviral injection to assess the effect on heroin seeking after 14 days of withdrawal. CS-induced heroin-seeking behavior was inhibited by downregulation of Akt1, but not ß-arrestin 2, in the NAc. These data demonstrate that Akt phosphorylation in the NAc may play an important role in the incubation of heroin-seeking behavior, suggesting that the PI3K/Akt pathways may be involved in the process of heroin relapse and addiction.


Subject(s)
Drug-Seeking Behavior/drug effects , Heroin/pharmacology , Nucleus Accumbens/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cues , Heroin Dependence/metabolism , Male , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Reward , Self Administration , Substance Withdrawal Syndrome/metabolism
9.
Drug Alcohol Depend ; 219: 108485, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33360853

ABSTRACT

INTRODUCTION: The striatum mediates reward processing in addiction, and previous fMRI (functional Magnetic Resonance Imaging) studies have revealed abnormal striatofrontal functional connectivity in heroin addiction. However, little is known about whether there is abnormal structural connectivity of the striatal circuit in heroin addiction. This study investigated the structural connectivity of striatal circuits in abstinent heroin-dependent individuals (HDIs) without methadone treatment. METHODS: Forty-three (age: 38.8 ± 7.1) male HDIs and twenty-one (age: 42.4 ± 7.9) matched healthy controls underwent high-resolution T1 and whole-brain diffusion tensor imaging (64 directions) magnetic resonance imaging. Connectivity-based seed classification probabilistic tractography was used to detect the tract strengths of striatal circuits with 10 a priori target masks. Tract strengths were compared between groups and correlated with impulsivity behavior, evaluated using the Barratt Impulsivity Scale (BIS), and craving, measured on visual analogue scale (VAS). RESULTS: HDIs showed significantly weaker tract strength of the left striatum-medial orbitofrontal cortex (mOFC) (Bonferroni corrected, p < 0.05/20 = 0.0025) and significantly higher BIS total, attention, motor, and non-planning scores (Bonferroni corrected, p < 0.05/4 = 0.0125) than controls. In HDIs, negative correlations were observed between the left striatum- mOFC tract strengths and the BIS total, attention and non-planning scores (r1=-0.410, p1 = 0.005; r2=-0.432, p2 = 0.003; r3=-0.506, p3<0.001) and between the right striatum-posterior cingulate cortex (PCC) tract strengths and craving scores (r=-0.433, p = 0.009) in HDIs. CONCLUSION: HDIs displayed decreased structural connectivity of the striatum-mOFC circuit and higher impulsivity. Higher impulsive behavior was associated with decreased left striatal circuit connectivity. These findings suggest that the striatal circuit tract strengths might be a novel potential biomarker in heroin and, potentially, general opioid addiction.


Subject(s)
Craving/physiology , Heroin Dependence/psychology , Heroin , Adult , Behavior, Addictive , Biomarkers , Brain/physiopathology , Brain Mapping , Case-Control Studies , Cerebral Cortex/physiopathology , Corpus Striatum/physiopathology , Diffusion Tensor Imaging , Gyrus Cinguli/physiopathology , Heroin Dependence/metabolism , Humans , Impulsive Behavior , Magnetic Resonance Imaging , Male , Middle Aged , Opioid-Related Disorders/physiopathology , Reward
10.
Urol J ; 17(6): 638-644, 2020 Jul 28.
Article in English | MEDLINE | ID: mdl-32748386

ABSTRACT

PURPOSE: Recreational use of illicit drugs is one of the main factors affecting male fertility. However, the mechanisms of heroin smoke-associated damage to mature spermatozoa are still completely unknown. The aim of this study was to concomitantly examine the levels of protamine-2 gene and protein concentrations, the amount of miRNA-122 in seminal plasma and semen analysis findings in heroin-addicted men. MATERIALS AND METHODS: In a case control study, twenty-four fertile men that lacked any recreational drug abuse were considered as the healthy group, and 24 addicted men who used only heroin for at least four months were selected as the addicted group. Semen samples were gathered by masturbation after 2 - 5 days of sexual abstinence. Following the preparation of a semen analysis by computer-assisted sperm analysis according to WHO (2010), the level of protamine-2 gene expression in sperm and miRNA-122 in seminal plasma was measured using real-time sqPCR. Also, protamine-2 protein concentrations were quantified by nuclear protein extraction, SDS-Page and western blotting. RESULTS: Among the studied variables, body mass index (27.75±0.88 vs. 22.30±0.36, p=0.001), seminal pH (7.79±0.06 vs. 7.58±0.06, p=0.003), white blood cell count in semen (1.69±0.41 vs. 8.61±1.73, p=0.001), motility (65.51±2.57 vs. 41.96±3.58, p=0.001) and survival rate (87.41±1.00 vs. 71.50±4.59, p=0.002) of sperm cells was significantly different between the healthy and addicted groups. In addition, the levels of protamine-2 gene and protein expression in the addicted group (0.05±0.02 and 0.10±0.02, respectively) were significantly lower than the healthy group (3.59±0.94 and 0.27±0.06, respectively) (p=0.002 and p=0.017, respectively). Seminal miRNA-122 levels in addicted men (3.51±0.73) were statistically higher than in healthy men (1.52±0.54) (p=0.034). However, there were some significant relationship between the studied parameters and addiction (p<0.05). CONCLUSION: This is one study on human infertility that evaluates the effects of heroin on protamine deficiency and seminal small RNAs expression levels. Heroin abuse may lead to male infertility by causing leukocytospermia, asthenozoospermia, protamine deficiency, and seminal plasma miRNA profile alteration.


Subject(s)
Heroin Dependence/metabolism , MicroRNAs/analysis , Protamines/analysis , Protamines/genetics , Semen Analysis , Semen/chemistry , Spermatozoa/chemistry , Adult , Case-Control Studies , Correlation of Data , Humans , Male
11.
J Clin Lab Anal ; 34(11): e23486, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32748469

ABSTRACT

BACKGROUND: Drug addiction is an uncontrolled, chronic, and recurrent encephalopathy that presently lacks specific and characteristic biomarkers for diagnosis and treatment. As regulators of gene expression, microRNAs (miRNAs) are increasingly used for diagnostic and prognostic purposes in various disease states. Previous studies indicated that miRNAs play important roles in the development and progression of drug addictions, including addiction to methamphetamine, cocaine, alcohol, and heroin. METHODS: We identified significant miRNAs using the microarray method and then validated the hsa-miR-181a expression levels in 53 heroin addiction patients and 49 normal controls using quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR). Finally, the potential associations between transcriptional levels in heroin addiction patients and their clinicopathological features were analyzed. RESULTS: A total of 2006 miRNAs were differentially expressed between heroin addiction patients and normal controls. The top 10 up-regulated miRNAs in patients were hsa-miR-21a, hsa-miR-181a, hsa-miR-4459, hsa-miR-4430, hsa-miR-4306, hsa-miR-22-3P, hsa-miR-486-5P, hsa-miR-371b-5P, hsa-miR-92a-3P, and hsa-miR-5001-5P. The top 10 down-regulated miRNAs in patients were hsa-miR-3195, hsa-miR-4767, hsa-miR-3135b, hsa-miR-6087, hsa-miR-1181, hsa-miR-4785, hsa-miR-718, hsa-miR-3141, hsa-miR-652-5P, and hsa-miR-6126. The expression level of hsa-miR-181a in heroin addiction patients was significantly increased compared with that in normal controls (P < .001). The area under the receiver operating characteristic curve of hsa-miR-181a was 0.783, the sensitivity was 0.867, and the specificity was 0.551. CONCLUSIONS: The increased expression of hsa-miR-181a in the plasma of heroin patients may be a consequence of the pathological process of heroin abuse. This study highlights the potential of hsa-miR-181a as a novel biomarker for the diagnosis of heroin addiction.


Subject(s)
Heroin Dependence , MicroRNAs , Adult , Biomarkers/blood , China , Heroin Dependence/blood , Heroin Dependence/epidemiology , Heroin Dependence/metabolism , Humans , Male , MicroRNAs/blood , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Transcriptome/genetics , Up-Regulation/genetics , Young Adult
12.
Article in English | MEDLINE | ID: mdl-32673834

ABSTRACT

A common phenomenon shows that ingestion of opium poppy shell-containing drugs can result in a "false-positive" urinalysis test result for mandatory or workplace heroin abuse screening. Owing to the short detection window (8 h in urine) of the characteristic heroin metabolite 6-monoacetylmorphine (6-MAM) confirmation or exclusion of heroin abusers still presents major challenges for toxicologists. In this work, we developed an ultra-performance liquid chromatography-time-of-flight mass spectrometry method (UPLC-TOF-MS) with online data acquisition and multiple post-data-mining technologies combined with a multivariate statistical and batch validation analysis workflow to assess the characteristic urine metabolites of heroin abusers. Based on the proposed methods, 28 characteristic metabolites were structurally identified, and their fragmentation patterns and metabolite pathways were also summarized. Correlation analysis was used to investigate the internal relationship and similarities among the identified metabolites, and seven representative metabolites were selected as "Target-metabolites". Multi-batch urine of samples of heroin abusers were certified based on the UPLC-MS/MS method for further validation of the practicability of using this method for routine analysis. Overall, the target-metabolites can be utilized as assistant "biomarkers" in workplace or mandatory drug screenings. This approach encourages further studies on the development of the "false-positive" identification system.


Subject(s)
Heroin Dependence/metabolism , Heroin Dependence/urine , Heroin/metabolism , Heroin/urine , Substance Abuse Detection/methods , Chromatography, High Pressure Liquid/methods , Data Mining/methods , Female , Humans , Male , Mass Spectrometry/methods , Morphine Derivatives/metabolism , Morphine Derivatives/urine , Reproducibility of Results
13.
Neurosci Lett ; 721: 134819, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32032749

ABSTRACT

It is widely reported that drug addiction involves the strengthening of specific reward circuits through N-methyl-d-aspartic acid receptor (NMDAR)-dependent synaptic potentiation, and several lines of evidence strongly implicate NMDA receptor 2 (NR2) subunits in drug abuse. To explore the potential mechanism of heroin dependence, this study examined changes in the expression levels of NR2 subunits NR2A-D in the prelimbic (PL) region of the medial prefrontal cortex (mPFC) after repeated heroin administration and subsequent abstinence. The conditioned place preference (CPP) test confirmed successful induction of heroin dependence and withdrawal. Western blotting and qRT-PCR revealed no differences in NR2A subunit expression among heroin-exposure, heroin-withdrawal, and control group rats; in contrast, expression of NR2B was significantly higher in the heroin-exposure group, whereas expression levels of NR2C and NR2D were significantly higher in the heroin-withdrawal group relative to the controls. Further studies are needed to identify the functional significance based on alterations of NR2 subunits.


Subject(s)
Heroin Dependence/metabolism , Heroin/adverse effects , Prefrontal Cortex/metabolism , Receptors, N-Methyl-D-Aspartate/biosynthesis , Substance Withdrawal Syndrome/metabolism , Animals , Gene Expression , Heroin Dependence/genetics , Male , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , Substance Withdrawal Syndrome/genetics
14.
Mol Med Rep ; 21(1): 405-412, 2020 01.
Article in English | MEDLINE | ID: mdl-31939625

ABSTRACT

Heroin addiction is a chronic relapsing brain disorder with negative social consequences. Histone acetylation serves a role in drug­induced behavior and neuroplasticity impairment. Brahma/SWI2­related gene­1 (BRG1) participates in cerebellar development, embryogenesis and transcriptional regulation of neuronal genes concurrent with histone modifications. However, little is known about the relationship between histone H3 lysine 9 acetylation (H3K9ac) and BRG1 in response to heroin. The present study aimed to assess the contribution of histone 3 lysine 9 acetylation of BRG1 to heroin self­administration. The present study established a Sprague­Dawley rat model of heroin self­administration under a fixed­ratio­1 paradigm. Chromatin immunoprecipitation followed by reverse transcription­quantitative PCR (RT­qPCR) was used to detect the accumulation of H3K9ac on BRG1 in the medial prefrontal cortex (mPFC) and nucleus accumbens (NAc) following heroin self­administration. The relative expression levels of BRG1 were analyzed by RT­qPCR. H3K9ac at the promoter region of BRG1 was significantly elevated (P=0.002), and the expression of BRG1 in the mPFC increased 1.47­fold in the heroin self­administration group compared with the control group. No significant difference in H3K9ac at the BRG1 locus was observed in the NAc (P=0.323), with the expression of BRG1 decreasing 1.38­fold in the heroin self­administering rats compared with the control group. H3K9ac is associated with transcriptional activation, and the increased BRG1 expression suggested an essential and novel role for BRG1 and its H3K9ac­mediated regulation in the mPFC after heroin self­administration; and this may function through epigenetically modulating the activation of neuroplasticity­associated genes. This association may provide a novel therapeutic target for the treatment of heroin addiction.


Subject(s)
DNA Helicases/metabolism , Heroin Dependence/metabolism , Heroin/administration & dosage , Histones/metabolism , Lysine/metabolism , Prefrontal Cortex/metabolism , Transcription Factors/metabolism , Acetylation , Animals , Chromatin Immunoprecipitation , DNA Helicases/genetics , Epigenesis, Genetic , Heroin Dependence/genetics , Histone Code , Male , Nucleus Accumbens/metabolism , Prefrontal Cortex/drug effects , Protein Processing, Post-Translational , Rats , Rats, Sprague-Dawley , Self Administration , Transcription Factors/genetics , Transcriptional Activation
15.
Eur Addict Res ; 26(2): 103-108, 2020.
Article in English | MEDLINE | ID: mdl-31940647

ABSTRACT

AIMS: Childhood trauma is of importance for the manifestation of substance-related disorders and maintenance of hypothalamic-pituitary-adrenal (HPA)-axis disorders. Since stress plays a crucial role in opioid compliance and craving, we investigated the immediate effects of diacetylmorphine application on the HPA axis. In particular, adrenocorticotropic hormone (ACTH) and cortisol secretion, as well as satiety regulating proopiomelanocortin peptides α-melanocyte-stimulating hormone (MSH) and ß-endorphin (END) in a cohort of opioid-dependent patients in diamorphine maintenance treatment concerning the clinical severity of their childhood trauma. METHODS: We compared the serum levels of ACTH, cortisol, MSH, and END in 15 opioid-dependent patients. All participants received treatment with diamorphine and were observed at 5 timepoints before and after injection. We split the cohort into 2 subgroups concerning childhood trauma measured by the Childhood Trauma Questionnaire. RESULTS: Splitting in 2 subgroups for mild (5) and severe trauma (10), we found that while both groups show a significant reduction of ACTH and cortisol levels over time, slopes display different progressions over time for cortisol (F[1.6] = 9.38, p = 0.02), while remaining identical for ACTH (F[1.6] = 1.69, p = 0.24). Also, levels of both MSH and END were significantly lower in severely traumatized patients. CONCLUSIONS: For the first time, we present a detailed representation of stress- and addiction-related proteins for the first 5 h after diamorphine application, demonstrating the interrelationship between stress hormones and childhood trauma as well as its potential effects on the progression of addictions such as opioid dependence.


Subject(s)
Adverse Childhood Experiences , Heroin Dependence , Heroin , Stress, Psychological/psychology , Wounds and Injuries/psychology , Adrenocorticotropic Hormone/blood , Adult , Child , Cohort Studies , Female , Heroin/pharmacology , Heroin/therapeutic use , Heroin Dependence/drug therapy , Heroin Dependence/metabolism , Humans , Hydrocortisone/blood , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Male , Middle Aged , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Surveys and Questionnaires/statistics & numerical data , beta-Endorphin/blood
16.
Addict Biol ; 25(4): e12793, 2020 07.
Article in English | MEDLINE | ID: mdl-31339209

ABSTRACT

Postretrieval extinction procedures are effective nonpharmacological interventions for disrupting drug-associated memories. Nonetheless, the conditioned stimulus (CS) memory retrieval-extinction procedure is ineffective in inhibiting drug craving and relapse after prolonged withdrawal, which significantly undermines its therapeutic potential. In the present study, we showed that, unlike the CS memory retrieval-extinction procedure, noncontingent heroin injections (unconditioned stimulus [UCS]) 1 hour before the extinction sessions decreased the heroin-priming-induced reinstatement, renewal, and spontaneous recovery of heroin seeking after 28 days of withdrawal (ie, remote heroin-associated memories) in rats. The UCS retrieval manipulation induced reactivation of the basolateral amygdala (BLA) after prolonged withdrawal, and this reactivation was absent with the CS retrieval manipulation. Chemogenetic inactivation of the BLA abolished the inhibitory effect of the UCS memory retrieval-extinction procedure on heroin-priming-induced reinstatement after prolonged withdrawal. Furthermore, the combination of chemogenetic reactivation of BLA and CS retrieval-extinction procedure resembled the inhibitory effect of UCS retrieval-extinction procedure on heroin seeking after prolonged withdrawal. We also observed that the inhibitory effect of the UCS retrieval-extinction procedure is mediated by regulation of AMPA receptor endocytosis in the BLA. Our results demonstrate critical engagement of the BLA in reconsolidation updating of heroin-associated memory after prolonged withdrawal, extending our knowledge of the boundary conditions of the reconsolidation of drug-associated memories.


Subject(s)
Basolateral Nuclear Complex/metabolism , Drug-Seeking Behavior/physiology , Extinction, Psychological/physiology , Heroin Dependence/metabolism , Heroin/pharmacology , Memory Consolidation/physiology , Narcotics/pharmacology , Animals , Basolateral Nuclear Complex/physiology , Central Amygdaloid Nucleus/metabolism , Central Amygdaloid Nucleus/physiology , Endocytosis , Heroin Dependence/physiopathology , Male , Rats , Receptors, AMPA/metabolism , Time Factors
17.
J Neuroimmune Pharmacol ; 15(3): 400-408, 2020 09.
Article in English | MEDLINE | ID: mdl-31828734

ABSTRACT

Exosomes play an important role in cell-to-cell communication as they can transfer functional molecules such as microRNAs (miRNAs) from one cell to another, exerting biological and immunological functions. Here, we investigated the impact of HIV infection and/or heroin use on the expression of the miRNAs in plasma exosomes. We found that HIV infection or heroin use upregulated the majority (98%) of a panel of plasma exosomal miRNAs associated with immune regulation and inflammation. We also observed the enhanced effect of HIV infection and heroin use on some of these upregulated miRNAs. Our further investigation showed that the levels of four of neuro-inflammation-related miRNAs (146a, 126, 21, and let-7a) were higher in HIV-infected heroin users as compared with the control subjects. These findings indicate that the dysregulations of the plasma exosomal miRNAs support further studies to determine the role of the miRNAs in HIV and/or heroin use-mediated immune modulation and neuro-inflammation. Graphical abstract.


Subject(s)
Exosomes/metabolism , HIV Infections/genetics , HIV Infections/metabolism , Heroin Dependence/genetics , Heroin Dependence/metabolism , MicroRNAs/blood , Adult , Cell Communication , Encephalitis/genetics , Encephalitis/metabolism , Female , HIV Infections/immunology , Heroin Dependence/immunology , Humans , Male , Middle Aged , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/metabolism , Up-Regulation , Young Adult
18.
J Neuropathol Exp Neurol ; 78(11): 1059-1065, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31559425

ABSTRACT

The aim of the study was to investigate blood-brain barrier alterations, neuroinflammation, and glial responses in drug abusers. Five immunohistochemical markers (CD3, zonula occludens-1 [ZO-1], intracellular adhesion molecule 1 [ICAM-1], vascular cell adhesion molecule [VCAM-1], and glial fibrillary acidic protein [GFAP]) were assessed on postmortem brain samples collected from drug abusers who died from acute intoxication of cocaine, heroin, or a combination of both, compared with controls. CD3 and ICAM-1 immunopositivity were significantly stronger in drug abusers than in controls. VCAM-1 immunopositivity was similar across drug abuser and control groups. In heroin abusers, significantly lower ZO-1 immunopositivity was observed relative to controls. GFAP positivity did not show significant differences between groups, but its distribution within the brain did differ. Both cocaine and heroin abuse promoted neuroinflammation, increasing expression of ICAM-1 and recruiting CD3+ lymphocytes. Heroin affected the molecular integrity of tight junctions, as reflected by reduced ZO-1 expression. The outcomes of the present study are, overall, consistent with prior available evidence, which is almost exclusively from studies conducted in vitro or in animal models. These findings provide important information about the downstream consequences of neuroinflammation in drug abusers and may help to inform the development of potential therapeutic targets.


Subject(s)
Brain/drug effects , Brain/pathology , Cocaine-Related Disorders/pathology , Encephalitis/pathology , Heroin Dependence/pathology , Adolescent , Adult , Autopsy , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/metabolism , Cocaine-Related Disorders/complications , Cocaine-Related Disorders/metabolism , Drug Overdose/metabolism , Drug Overdose/pathology , Encephalitis/etiology , Encephalitis/metabolism , Female , Heroin Dependence/complications , Heroin Dependence/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Young Adult
19.
Physiol Res ; 68(5): 835-844, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31424247

ABSTRACT

Opiate addiction has a high rate of relapse. The accumulating evidence shows that electroacupuncture (EA) may be effective for the treatment of opiate relapse. However, the change of expression of CB1-Rs and CB2-Rs involve in 2Hz EA anti-relapse pathway is still unclear. To explore the changes of expression of CB1-Rs and CB2-Rs, heroin self-administration (SA) model rats were adopted and treated using 2Hz EA. The expressions of CB1-Rs and CB2-Rs were observed using immunohistochemistry method. The results showed that, compared with the control group, active pokes in the heroin-addicted group increased, while the active pokes decreased significantly in 2Hz EA group compared with heroin-addicted group. Correspondingly, the expression of CB1-Rs in prefrontal cortex (PFC), hippocampus (Hip), nucleus accumbens (NAc) and ventral tegmental area (VTA) all increased significantly while the expression of CB2-Rs in those relapse-relevant brain regions decreased obviously in heroin-addicted group when compared with the control group. In addition, the expression of CB1-Rs obviously decreased in the 2Hz EA group while the expression of CB2-Rs in those relapse-relevant brain regions increased significantly when compared with the heroin-addicted group. It indicated that 2Hz EA could attenuate the heroin-evoked seeking behaviors effectively. The anti-relapse effects of 2Hz EA might be related to the decrease of CB1-Rs and increase of CB2-Rs expression in relapse-relevant brain regions of heroin SA rats.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Drug-Seeking Behavior/drug effects , Electroacupuncture , Heroin Dependence/therapy , Heroin/administration & dosage , Narcotic Antagonists/administration & dosage , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Animals , Brain/metabolism , Brain/physiopathology , Disease Models, Animal , Extinction, Psychological/drug effects , Heroin Dependence/metabolism , Heroin Dependence/physiopathology , Heroin Dependence/psychology , Locomotion/drug effects , Male , Rats, Sprague-Dawley , Recurrence , Self Administration , Signal Transduction
20.
Eur Psychiatry ; 59: 15-24, 2019 06.
Article in English | MEDLINE | ID: mdl-30981746

ABSTRACT

BACKGROUND: Stimulant drugs can cause persistent changes in the brain. Imaging studies show that these changes are most apparent in dopamine transporter (DAT) or receptor availability within the striatum. METHODS: This work focuses on influences of stimulant use on dopaminergic function assessed using nuclear-medicine imaging (PET/SPECT). Included are 39 studies on 655 cocaine, amphetamine, methamphetamine or nicotine users, as well as 690 healthy controls. Metaanalyses were conducted separately for D2/D3 receptors and dopamine transporters of the entire striatum, its subregions caudate and putamen respectively. RESULTS: Meta-analyses results regarding nicotine did not show significant effects between smokers and nonsmokers. In cocaine users there was a significant decrease in dopamine receptor availability in all regions. The striatal DAT availability was significantly increased in cocaine users. Methamphetamine users showed a significantly decreased dopamine receptor and transporter density in all regions. Significant results also indicate a lower transporter availability in all regions. Amphetamine users showed reduced DAT availability in the striatum, as well as in the sub regions. CONCLUSION: This meta-analysis provides evidence that there are ongoing changes in the dopaminergic system associated with the use of stimulants. Especially the results of cocaine, methamphetamine and amphetamine use mainly showed a downregulation. In addition, this meta-analysis is the first to include nicotine. This subset of studies showed evidence for a decreased receptor and DAT availability but no significant results were found in the metaanalyses.


Subject(s)
Amphetamine-Related Disorders/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Heroin Dependence/metabolism , Methamphetamine/metabolism , Receptors, Dopamine D2/metabolism , Brain/metabolism , Central Nervous System Stimulants/pharmacology , Dopamine , Dopamine Plasma Membrane Transport Proteins/drug effects , Humans , Neostriatum/metabolism , Tomography, Emission-Computed, Single-Photon
SELECTION OF CITATIONS
SEARCH DETAIL
...