Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.767
Filter
1.
Int J Mol Sci ; 25(10)2024 May 13.
Article in English | MEDLINE | ID: mdl-38791331

ABSTRACT

Dopamine is a key neurotransmitter involved in physiological processes such as motor control, motivation, reward, cognitive function, and maternal and reproductive behaviors. Therefore, dysfunctions of the dopaminergic system are related to a plethora of human diseases. Dopamine, via different circuitries implicated in compulsive behavior, reward, and habit formation, also represents a key player in substance use disorder and the formation and perpetuation of mechanisms leading to addiction. Here, we propose dopamine as a model not only of neurotransmission but also of neuromodulation capable of modifying neuronal architecture. Abuse of substances like methamphetamine, cocaine, and alcohol and their consumption over time can induce changes in neuronal activities. These modifications lead to synaptic plasticity and finally to morphological and functional changes, starting from maladaptive neuro-modulation and ending in neurodegeneration.


Subject(s)
Dopamine , Humans , Dopamine/metabolism , Animals , Substance-Related Disorders/metabolism , Neuronal Plasticity/drug effects , Synaptic Transmission/drug effects
2.
Neurosci Biobehav Rev ; 161: 105690, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38678736

ABSTRACT

Dopamine's role in addiction has been extensively studied, revealing disruptions in its functioning throughout all addiction stages. Neuromelanin in the substantia nigra (SN) may reflect dopamine auto-oxidation, and can be quantified using neuromelaninsensitive magnetic resonance imaging (neuromelanin-MRI) in a non-invasive manner.In this pre-registered systematic review, we assess the current body of evidence related to neuromelanin levels in substance use disorders, using both post-mortem and MRI examinations. The systematic search identified 10 relevant articles, primarily focusing on the substantia nigra. An early-stage meta-analysis (n = 6) revealed varied observations ranging from standardized mean differences of -3.55 to +0.62, with a pooled estimate of -0.44 (95 % CI = -1.52, 0.65), but there was insufficient power to detect differences in neuromelanin content among individuals with substance use disorders. Our gap analysis highlights the lack of sufficient replication studies, with existing studies lacking the power to detect a true difference, and a complete lack of neuromelanin studies on certain substances of clinical interest. We provide recommendations for future studies of dopaminergic neurobiology in addictions and related psychiatric comorbidities.


Subject(s)
Melanins , Substance-Related Disorders , Humans , Melanins/metabolism , Substance-Related Disorders/metabolism , Substance-Related Disorders/diagnostic imaging , Substantia Nigra/metabolism , Substantia Nigra/diagnostic imaging , Magnetic Resonance Imaging
3.
J Neural Transm (Vienna) ; 131(5): 509-523, 2024 05.
Article in English | MEDLINE | ID: mdl-38630190

ABSTRACT

Substance use disorders (SUDs) are the most costly and prevalent psychiatric conditions. Recent calls emphasize a need for biomarkers-measurable, stable indicators of normal and abnormal processes and response to treatment or environmental agents-and, in particular, brain-based neuromarkers that will advance understanding of the neurobiological basis of SUDs and clinical practice. To develop neuromarkers, researchers must be grounded in evidence that a putative marker (i) is sensitive and specific to the psychological phenomenon of interest, (ii) constitutes a predictive model, and (iii) generalizes to novel observations (e.g., through internal cross-validation and external application to novel data). These neuromarkers may be used to index risk of developing SUDs (susceptibility), classify individuals with SUDs (diagnostic), assess risk for progression to more severe pathology (prognostic) or index current severity of pathology (monitoring), detect response to treatment (response), and predict individualized treatment outcomes (predictive). Here, we outline guidelines for developing and assessing neuromarkers, we then review recent advances toward neuromarkers in addiction neuroscience centering our discussion around neuromarkers of craving-a core feature of SUDs. In doing so, we specifically focus on the Neurobiological Craving Signature (NCS), which show great promise for meeting the demand of neuromarkers.


Subject(s)
Biomarkers , Substance-Related Disorders , Humans , Biomarkers/metabolism , Substance-Related Disorders/diagnosis , Substance-Related Disorders/metabolism , Brain/metabolism , Behavior, Addictive/diagnosis , Behavior, Addictive/metabolism
4.
Pharmacol Res ; 202: 107143, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38499081

ABSTRACT

Substance use disorders (SUD) are chronic relapsing disorders governed by continually shifting cycles of positive drug reward experiences and drug withdrawal-induced negative experiences. A large body of research points to plasticity within systems regulating emotional, motivational, and cognitive processes as drivers of continued compulsive pursuit and consumption of substances despite negative consequences. This plasticity is observed at all levels of analysis from molecules to networks, providing multiple avenues for intervention in SUD. The cytoskeleton and its regulatory proteins within neurons and glia are fundamental to the structural and functional integrity of brain processes and are potentially the major drivers of the morphological and behavioral plasticity associated with substance use. In this review, we discuss preclinical studies that provide support for targeting the brain cytoskeleton as a therapeutic approach to SUD. We focus on the interplay between actin cytoskeleton dynamics and exposure to cocaine, methamphetamine, alcohol, opioids, and nicotine and highlight preclinical studies pointing to a wide range of potential therapeutic targets, such as nonmuscle myosin II, Rac1, cofilin, prosapip 1, and drebrin. These studies broaden our understanding of substance-induced plasticity driving behaviors associated with SUD and provide new research directions for the development of SUD therapeutics.


Subject(s)
Substance Withdrawal Syndrome , Substance-Related Disorders , Humans , Substance-Related Disorders/drug therapy , Substance-Related Disorders/metabolism , Cytoskeleton , Actin Cytoskeleton/metabolism , Brain , Substance Withdrawal Syndrome/metabolism
5.
Transl Psychiatry ; 14(1): 101, 2024 Feb 19.
Article in English | MEDLINE | ID: mdl-38374108

ABSTRACT

G protein-coupled receptor 55 (GPR55) has been thought to be a putative cannabinoid receptor. However, little is known about its functional role in cannabinoid action and substance use disorders. Here we report that GPR55 is predominantly found in glutamate neurons in the brain, and its activation reduces self-administration of cocaine and nicotine in rats and mice. Using RNAscope in situ hybridization, GPR55 mRNA was identified in cortical vesicular glutamate transporter 1 (VgluT1)-positive and subcortical VgluT2-positive glutamate neurons, with no detection in midbrain dopamine (DA) neurons. Immunohistochemistry detected a GPR55-like signal in both wildtype and GPR55-knockout mice, suggesting non-specific staining. However, analysis using a fluorescent CB1/GPR55 ligand (T1117) in CB1-knockout mice confirmed GPR55 binding in glutamate neurons, not in midbrain DA neurons. Systemic administration of the GPR55 agonist O-1602 didnt impact ∆9-THC-induced analgesia, hypothermia and catalepsy, but significantly mitigated cocaine-enhanced brain-stimulation reward caused by optogenetic activation of midbrain DA neurons. O-1602 alone failed to alter extracellar DA, but elevated extracellular glutamate, in the nucleus accumbens. In addition, O-1602 also demonstrated inhibitory effects on cocaine or nicotine self-administration under low fixed-ratio and/or progressive-ratio reinforcement schedules in rats and wildtype mice, with no such effects observed in GPR55-knockout mice. Together, these findings suggest that GPR55 activation may functionally modulate drug-taking and drug-seeking behavior possibly via a glutamate-dependent mechanism, and therefore, GPR55 deserves further study as a new therapeutic target for treating substance use disorders.


Subject(s)
Cannabidiol , Cocaine , Receptors, Cannabinoid , Substance-Related Disorders , Animals , Mice , Rats , Cannabidiol/analogs & derivatives , Cocaine/pharmacology , Dopaminergic Neurons/metabolism , Glutamic Acid/metabolism , Mice, Knockout , Nicotine/pharmacology , Pharmaceutical Preparations/metabolism , Receptors, Cannabinoid/metabolism , Receptors, G-Protein-Coupled/genetics , Substance-Related Disorders/genetics , Substance-Related Disorders/metabolism
6.
J Neural Transm (Vienna) ; 131(5): 495-503, 2024 05.
Article in English | MEDLINE | ID: mdl-38396082

ABSTRACT

Alternative splicing is a co-transcriptional process that significantly contributes to the molecular landscape of the cell. It plays a multifaceted role in shaping gene transcription, protein diversity, and functional adaptability in response to environmental cues. Recent studies demonstrate that drugs of abuse have a profound impact on alternative splicing patterns within different brain regions. Drugs like alcohol and cocaine modify the expression of genes responsible for encoding splicing factors, thereby influencing alternative splicing of crucial genes involved in neurotransmission, neurogenesis, and neuroinflammation. Notable examples of these alterations include alcohol-induced changes in splicing factors such as HSPA6 and PCBP1, as well as cocaine's impact on PTBP1 and SRSF11. Beyond the immediate effects of drug exposure, recent research has shed light on the role of alternative splicing in contributing to the risk of substance use disorders (SUDs). This is exemplified by exon skipping events in key genes like ELOVL7, which can elevate the risk of alcohol use disorder. Lastly, drugs of abuse can induce splicing alterations through epigenetic modifications. For example, cocaine exposure leads to alterations in levels of trimethylated lysine 36 of histone H3, which exhibits a robust association with alternative splicing and serves as a reliable predictor for exon exclusion. In summary, alternative splicing has emerged as a critical player in the complex interplay between drugs of abuse and the brain, offering insights into the molecular underpinnings of SUDs.


Subject(s)
Brain , Substance-Related Disorders , Humans , Substance-Related Disorders/genetics , Substance-Related Disorders/metabolism , Brain/metabolism , Brain/drug effects , Animals , Alternative Splicing , RNA Splicing/drug effects
7.
Pharmacol Ther ; 253: 108580, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38142862

ABSTRACT

Trace amines, a group of amines expressed at the nanomolar level in the mammalian brain, can modulate monoamine transmission. The discovery of and the functional research on the trace amine-associated receptors (TAARs), especially the most well-characterized TAAR1, have largely facilitated our understanding of the function of the trace amine system in the brain. TAAR1 is expressed in the mammalian brain at a low level and widely distributed in the monoaminergic system, including the ventral tegmental area and substantial nigra, where the dopamine neurons reside in the mammalian brain. Growing in vitro and in vivo evidence has demonstrated that TAAR1 could negatively modulate monoamine transmission and play a crucial role in many psychiatric disorders, including schizophrenia, substance use disorders, sleep disorders, depression, and anxiety. Notably, in the last two decades, many studies have repeatedly confirmed the pharmacological effects of the selective TAAR1 ligands in various preclinical models of psychiatric disorders. Recent clinical trials of the dual TAAR1 and serotonin receptor agonist ulotaront also revealed a potential efficacy for treating schizophrenia. Here, we review the current understanding of the TAAR1 system and the recent advances in the elucidation of behavioral and physiological properties of TAAR1 agonists evaluated both in preclinical animal models and clinical trials. We also discuss the potential TAAR1-dependent signaling pathways and the cellular mechanisms underlying the inhibitory effects of TAAR1 activation on drug addiction. We conclude that TAAR1 is an emerging target for the treatment of psychiatric disorders.


Subject(s)
Mental Disorders , Substance-Related Disorders , Animals , Humans , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Mental Disorders/drug therapy , Mental Disorders/metabolism , Brain/metabolism , Substance-Related Disorders/metabolism , Amines/metabolism , Mammals/metabolism
8.
Int J Mol Sci ; 24(23)2023 Dec 04.
Article in English | MEDLINE | ID: mdl-38069445

ABSTRACT

Substance addiction is a chronic and relapsing brain disorder characterized by compulsive seeking and continued substance use, despite adverse consequences. The high prevalence and social burden of addiction are indisputable; however, the available intervention is insufficient. The modulation of gene expression and aberrant adaptation of neural networks are attributed to the changes in brain functions under repeated exposure to addictive substances. Considerable studies have demonstrated that miRNAs are strong modulators of post-transcriptional gene expression in substance addiction. The emerging role of microRNA (miRNA) provides new insights into many biological and pathological processes in the central nervous system: their variable expression in different regions of the brain and tissues may play a key role in regulating the pathophysiological events of addiction. This work provides an overview of the current literature on miRNAs involved in addiction, evaluating their impaired expression and regulatory role in neuroadaptation and synaptic plasticity. Clinical implications of such modulatory capacities will be estimated. Specifically, it will evaluate the potential diagnostic role of miRNAs in the various stages of drug and substance addiction. Future perspectives about miRNAs as potential novel therapeutic targets for substance addiction and abuse will also be provided.


Subject(s)
Behavior, Addictive , MicroRNAs , Substance-Related Disorders , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Substance-Related Disorders/metabolism , Behavior, Addictive/metabolism , Brain/metabolism
9.
Nature ; 624(7992): 663-671, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37935377

ABSTRACT

Trace amine-associated receptor 1 (TAAR1), the founding member of a nine-member family of trace amine receptors, is responsible for recognizing a range of biogenic amines in the brain, including the endogenous ß-phenylethylamine (ß-PEA)1 as well as methamphetamine2, an abused substance that has posed a severe threat to human health and society3. Given its unique physiological role in the brain, TAAR1 is also an emerging target for a range of neurological disorders including schizophrenia, depression and drug addiction2,4,5. Here we report structures of human TAAR1-G-protein complexes bound to methamphetamine and ß-PEA as well as complexes bound to RO5256390, a TAAR1-selective agonist, and SEP-363856, a clinical-stage dual agonist for TAAR1 and serotonin receptor 5-HT1AR (refs. 6,7). Together with systematic mutagenesis and functional studies, the structures reveal the molecular basis of methamphetamine recognition and underlying mechanisms of ligand selectivity and polypharmacology between TAAR1 and other monoamine receptors. We identify a lid-like extracellular loop 2 helix/loop structure and a hydrogen-bonding network in the ligand-binding pockets, which may contribute to the ligand recognition in TAAR1. These findings shed light on the ligand recognition mode and activation mechanism for TAAR1 and should guide the development of next-generation therapeutics for drug addiction and various neurological disorders.


Subject(s)
Methamphetamine , Phenethylamines , Receptors, G-Protein-Coupled , Humans , Ligands , Methamphetamine/metabolism , Nervous System Diseases/metabolism , Phenethylamines/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Substance-Related Disorders/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Polypharmacology , Hydrogen Bonding
10.
Neurochem Int ; 169: 105572, 2023 10.
Article in English | MEDLINE | ID: mdl-37423274

ABSTRACT

Drug abuse changes neurophysiological functions at multiple cellular and molecular levels in the addicted brain. Well-supported scientific evidence suggests that drugs negatively affect memory formation, decision-making and inhibition, and emotional and cognitive behaviors. The mesocorticolimbic brain regions are involved in reward-related learning and habitual drug-seeking/taking behaviors to develop physiological and psychological dependence on the drugs. This review highlights the importance of specific drug-induced chemical imbalances resulting in memory impairment through various neurotransmitter receptor-mediated signaling pathways. The mesocorticolimbic modifications in the expression levels of brain-derived neurotrophic factor (BDNF) and the cAMP-response element binding protein (CREB) impair reward-related memory formation following drug abuse. The contributions of protein kinases and microRNAs (miRNAs), along with the transcriptional and epigenetic regulation have also been considered in memory impairment underlying drug addiction. Overall, we integrate the research on various types of drug-induced memory impairment in distinguished brain regions and provide a comprehensive review with clinical implications addressing the upcoming studies.


Subject(s)
Epigenesis, Genetic , Substance-Related Disorders , Humans , Signal Transduction/physiology , Substance-Related Disorders/metabolism , Memory Disorders/chemically induced , Memory Disorders/metabolism , Neuronal Plasticity/physiology , Neurogenesis , Brain-Derived Neurotrophic Factor/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Hippocampus/metabolism
11.
Biomolecules ; 13(6)2023 05 31.
Article in English | MEDLINE | ID: mdl-37371502

ABSTRACT

During the last decade, substance use disorders (SUDs) have been increasingly recognized as neuroinflammation-related brain diseases. Various types of abused drugs (cocaine, methamphetamine, alcohol, opiate-like drugs, marijuana, etc.) can modulate the activation status of microglia and neuroinflammation levels which are involved in the pathogenesis of SUDs. Several neuroimmune signaling pathways, including TLR/NF-кB, reactive oxygen species, mitochondria dysfunction, as well as autophagy defection, etc., have been implicated in promoting SUDs. Recently, inflammasome-mediated signaling has been identified as playing critical roles in the microglia activation induced by abused drugs. Among the family of inflammasomes, NOD-, LRR-, and pyrin-domain-containing protein 3 (NLRP3) serves the primary research target due to its abundant expression in microglia. NLRP3 has the capability of integrating multiple external and internal inputs and coordinately determining the intensity of microglia activation under various pathological conditions. Here, we summarize the effects of abused drugs on NLRP3 inflammasomes, as well as others, if any. The research on this topic is still at an infant stage; however, the readily available findings suggest that NLRP3 inflammasome could be a common downstream effector stimulated by various types of abused drugs and play critical roles in determining abused-drug-mediated biological effects through enhancing glia-neuron communications. NLRP3 inflammasome might serve as a novel target for ameliorating the development of SUDs.


Subject(s)
Inflammasomes , Substance-Related Disorders , Humans , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Microglia/metabolism , Neuroinflammatory Diseases , Substance-Related Disorders/metabolism
12.
Drug Alcohol Depend ; 248: 109900, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37148676

ABSTRACT

BACKGROUND: Although proton magnetic resonance spectroscopy (MRS) has been used to study metabolite alterations in stimulant (methamphetamine and cocaine) substance use disorders (SUDs) for over 25 years, data-driven consensus regarding the nature and magnitude of these alterations is lacking. METHOD: In this meta-analysis, we examined associations between SUD and regional metabolites (N-acetyl aspartate (NAA), choline, myo-inositol, creatine, glutamate, and glutamate+glutamine (glx)) in the medial prefrontal cortex (mPFC), frontal white matter (FWM), occipital cortex, and basal ganglia as measured by 1 H-MRS. We also examined moderating effects of MRS acquisition parameters (echo time (TE), field strength), data quality (coefficient of variation (COV)), and demographic/clinical variables. RESULTS: A MEDLINE search revealed 28 articles that met meta-analytic criteria. Significant effects included lower mPFC NAA, higher mPFC myo-inositol, and lower mPFC creatine in SUD relative to people without SUD. mPFC NAA effects were moderated by TE, with larger effects at longer TEs. For choline, although no group effects were observed, effect sizes in the mPFC were related to MRS technical indicators (field strength, COV). No effects of age, sex, primary drug of use (methamphetamine vs. cocaine), duration of use, or duration of abstinence were observed. Evidence for moderating effects of TE and COV may have implications for future MRS studies in SUDs. CONCLUSIONS: The observed metabolite profile in methamphetamine and cocaine SUD (lower NAA and creatine with higher myo-inositol) parallels that observed in Alzheimer's disease and mild cognitive impairment, suggesting these drugs are associated with neurometabolic differences similar to those characterizing these neurodegenerative conditions.


Subject(s)
Methamphetamine , Substance-Related Disorders , Humans , Magnetic Resonance Spectroscopy/methods , Creatine/metabolism , Substance-Related Disorders/metabolism , Glutamic Acid/metabolism , Choline/metabolism , Inositol/metabolism , Brain/metabolism
13.
Front Public Health ; 11: 1122455, 2023.
Article in English | MEDLINE | ID: mdl-37006591

ABSTRACT

Introduction: Patients with diabetes and comorbid substance use disorders (SUD) experience poor diabetes management, increased medical complications and mortality. However, research has documented that patients engaged in substance abuse treatment have better management of their comorbid conditions. The current study examines diabetes management among patients with type 2 diabetes, with and without comorbid SUD, receiving care at Florida-based Federally Qualified Health Centers (FQHC) of Health Choice Network (HCN). Methods: A retrospective analysis was conducted using deidentified electronic health records of 37,452 patients with type 2 diabetes who received care at a HCN site in Florida between 2016 and 2019. A longitudinal logistic regression analysis examined the impact of SUD diagnosis on achievement of diabetes management [HbA1c < 7.0% (53 mmol/mol)] over time. A secondary analysis evaluated, within those with an SUD diagnosis, the likelihood of HbA1c control between those with and without SUD treatment. Results: The longitudinal assessment of the relationship between SUD status and HbA1c control revealed that those with SUD (N = 6,878, 18.4%) were less likely to control HbA1c over time (OR = 0.56; 95% CI = 0.49-0.63). Among those with SUD, patients engaged in SUD treatment were more likely to control HbA1c (OR = 5.91; 95% CI = 5.05-6.91). Discussion: Findings highlight that untreated SUD could adversely affect diabetes control and sheds light on the opportunity to enhance care delivery for patients with diabetes and co-occurring SUD.


Subject(s)
Blood Glucose , Community Health Centers , Diabetes Mellitus, Type 2 , Substance-Related Disorders , Substance-Related Disorders/complications , Substance-Related Disorders/metabolism , Substance-Related Disorders/therapy , Blood Glucose/metabolism , Florida , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Humans , Male , Female , Adult , Middle Aged , Electronic Health Records , Retrospective Studies , Logistic Models , Longitudinal Studies
14.
Mol Cell Neurosci ; 125: 103838, 2023 06.
Article in English | MEDLINE | ID: mdl-36893849

ABSTRACT

Microglia are widely known for their role in immune surveillance and for their ability to refine neurocircuitry during development, but a growing body of evidence suggests that microglia may also play a complementary role to neurons in regulating the behavioral aspects of substance use disorders. While many of these efforts have focused on changes in microglial gene expression associated with drug-taking, epigenetic regulation of these changes has yet to be fully understood. This review provides recent evidence supporting the role of microglia in various aspects of substance use disorder, with particular focus on changes to the microglial transcriptome and the potential epigenetic mechanisms driving these changes. Further, this review discusses the latest technical advances in low-input chromatin profiling and highlights the current challenges for studying these novel molecular mechanisms in microglia.


Subject(s)
Microglia , Substance-Related Disorders , Humans , Microglia/metabolism , Epigenesis, Genetic , Chromatin/metabolism , Substance-Related Disorders/genetics , Substance-Related Disorders/metabolism , Transcriptome
15.
Drug Test Anal ; 15(7): 711-729, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36756728

ABSTRACT

Acetylbenzylfentanyl, benzoylbenzylfentanyl, 3-fluoro-methoxyacetylfentanyl, and 3-phenylpropanoylfentanyl are fentanyl analogs that have been reported to the European Monitoring Centre for Drugs and Drug Addiction in recent years. The aim of this study was to identify metabolic pathways and potential biomarker metabolites of these fentanyl analogs. The compounds were incubated (5 µM) with cryopreserved hepatocytes for up to 5 h in vitro. Metabolites were analyzed with liquid chromatography-quadrupole time of flight-high-resolution mass spectrometry (LC-QTOF-HRMS). The experiments showed that acetylbenzylfentanyl, benzoylbenzylfentanyl, and 3-phenylpropanoylfentanyl were mainly metabolized through N-dealkylation (forming nor-metabolites) and 3-fluoro-methoxyacetylfentanyl mainly through demethylation. Other observed metabolites were formed by mono-/dihydroxylation, dihydrodiol formation, demethylation, dehydrogenation, amide hydrolysis, and/or glucuronidation. The experiments showed that a large number of metabolites of 3-phenylpropanoylfentanyl were formed. The exact position of hydroxy groups in formed monohydroxy metabolites could not be established solely based upon recorded MSMS spectra of hepatocyte samples. Therefore, potential monohydroxy metabolites of 3-phenylpropanoylfentanyl, with the hydroxy group in different positions, were synthesized and analyzed together with the hepatocyte samples. This approach could reveal that the ß position of the phenylpropanoyl moiety was highly favored; ß-OH-phenylpropanoylfentanyl was the most abundant metabolite after the nor-metabolite. Both metabolites have the potential to serve as biomarkers for 3-phenylpropanoylfentanyl. The nor-metabolites of acetylbenzylfentanyl, benzoylbenzylfentanyl, and 3-fluoro-methoxyacetylfentanyl do also seem to be suitable biomarker metabolites, as do the demethylated metabolite of 3-fluoro-methoxyacetylfentanyl. Identified metabolic pathways and formed metabolites were in agreement with findings in previous studies of similar fentanyl analogs.


Subject(s)
Fentanyl , Substance-Related Disorders , Humans , Chromatography, Liquid , Mass Spectrometry , Substance-Related Disorders/metabolism , Microsomes, Liver/metabolism , Biomarkers/metabolism
16.
Nat Commun ; 14(1): 71, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36604429

ABSTRACT

The lateral hypothalamus (LH) is physiologically critical in brain functions. The LH also plays an important role in drug addiction. However, neural circuits underlying LH involvement of drug addiction remain obscure. In the present study,our results showed that in male mice, during context-induced expression of morphine withdrawal memory, LH glutamatergic neurons played an important role; dopamine D1 receptor-expressing medium spiny neurons (D1-MSNs) projecting from the core of nucleus accumbens (NAcC) to the LH were an important upstream circuit to activate LH glutamatergic neurons; D1-MSNs projecting from the NAcC to the LH activated LH glutamatergic neurons through inhibiting LH local gamma-aminobutyric acid (GABA) neurons. These results suggest that disinhibited LH glutamatergic neurons by neural circuits from the NAcC importantly contribute to context-induced the expression of morphine withdrawal memory.


Subject(s)
Morphine , Substance-Related Disorders , Mice , Male , Animals , Morphine/adverse effects , Nucleus Accumbens/metabolism , Hypothalamic Area, Lateral/metabolism , Neurons/metabolism , Receptors, Dopamine D1/metabolism , Substance-Related Disorders/metabolism
17.
Neuropharmacology ; 224: 109336, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36414149

ABSTRACT

L-type calcium channels (LTCCs), including the Cav1.2 and Cav1.3 LTCC subtypes, are important regulators of calcium entry into neurons, which mediates neurotransmitter release and synaptic plasticity. Cav1.2 and Cav1.3 are encoded by the CACNA1C and CACNA1D genes, respectively. These genes are implicated in substance use disorders and depression in humans, as demonstrated by genetic-wide association studies (GWAS). Pre-clinical models have also revealed a critical role of LTCCs on drug and mood related behavior, including the co-morbidity of substance use and mood disorders. Moreover, LTCCs have been shown to regulate the neuronal firing of dopamine (DA) neurons as well as drug and stress-induced plasticity within the ventral tegmental area (VTA) to nucleus accumbens (NAc) pathway. Thus, LTCCs are interesting targets for the treatment of neuropsychiatric diseases. In this review, we provide a brief introduction to voltage-gated calcium channels, specifically focusing on the LTCCs. We place particular emphasis on the ability of LTCCs to regulate DA neuronal activity and downstream signaling in the VTA to NAc pathway, and how such processes mediate substance use and mood disorder-related behavioral responses. We also discuss the bi-directional control of VTA LTCCs on drug and mood-related behaviors in pre-clinical models, with implications for co-morbid psychiatric diagnosis. We conclude with a section on the clinical implications of LTCC blockers, many which are already FDA approved as cardiac medications. Thus, pre-clinical and clinical work should examine the potential of LTCC blockers to be repurposed for neuropsychiatric illness. This article is part of the Special Issue on 'L-type calcium channel mechanisms in neuropsychiatric disorders'.


Subject(s)
Nucleus Accumbens , Substance-Related Disorders , Humans , Nucleus Accumbens/metabolism , Ventral Tegmental Area/metabolism , Calcium Channels, L-Type/metabolism , Dopamine/metabolism , Mood Disorders/metabolism , Substance-Related Disorders/metabolism , Morbidity , Calcium Channel Blockers/pharmacology
18.
Behav Brain Res ; 439: 114244, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36470419

ABSTRACT

Substance abuse is a worldwide problem with serious repercussions for patients and the communities where they live. Pregabalin (Lyrica), is a medication commonly used to treat neuropathic pain. Like other analgesic medications there has been concern about pregabalin abuse and misuse. Although it was initially suggested that pregabalin, like other gabapentinoids, has limited abuse liability, questions still remain concerning this inquiry. Changes in glutamate system homeostasis are a hallmark of adaptations underlying drug dependence, including down-regulation of the glutamate transporter 1 (GLT-1; SLC1A2) and the cystine/glutamate antiporter (xCT; SLC7A11). In this study, it was found that pregabalin (90 mg/kg) produces a conditioned place preference (CPP), indicative of reinforcing effects that suggest a potential for abuse liability. Moreover, like other drugs of abuse, pregabalin also produced alterations in glutamate homeostasis, reducing the mRNA expression of Slc1a2 and Slc7a11 in the nucleus accumbens (NAc) and medial prefrontal cortex (mPFC). Amoxicillin clavulanic acid, a ß-lactam antibiotic, blocked the reinforcing effects of pregabalin and normalized glutamate homeostasis. These results suggest that pregabalin has abuse potential that should be examined more critically, and that, moreover, the mechanisms underlying these effects are similar to those of other drugs of abuse, such as heroin and cocaine. Additionally, these results support previous findings showing normalization of glutamate homeostasis by ß-lactam drugs that provides a novel potential therapeutic approach for the treatment of drug abuse and dependence.


Subject(s)
Amoxicillin-Potassium Clavulanate Combination , Substance-Related Disorders , Humans , Amoxicillin-Potassium Clavulanate Combination/metabolism , Amoxicillin-Potassium Clavulanate Combination/pharmacology , Pregabalin/pharmacology , Nucleus Accumbens , Substance-Related Disorders/metabolism , Excitatory Amino Acid Transporter 2/metabolism , beta-Lactams/pharmacology , Glutamates/metabolism , Glutamates/pharmacology , Glutamic Acid/metabolism
19.
Eur Arch Psychiatry Clin Neurosci ; 273(2): 411-425, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36094569

ABSTRACT

Antisocial behavior (ASB) is characterized by frequent violations of the rights and properties of others, as well as aggressive conduct. While ample evidence points to a critical role of serotonin in the emotional modulation of social responses, the implication of this neurotransmitter in ASB is unclear. Here, we performed the first-ever postmortem analysis of serotonergic markers in the orbitofrontal cortex (OFC) of male subjects with ASB (n = 9). We focused on this brain region, given its well-recognized role in social response and ASB pathophysiology. Given that all individuals also had a substance use disorder (SUD) diagnosis, two age-matched control groups were used: SUD only and unaffected controls. Tissues were processed for immunoblotting analyses on eight key serotonergic targets: tryptophan hydroxylase 2 (TPH2), the rate-limiting enzyme of brain serotonin synthesis; serotonin transporter (SERT), the primary carrier for serotonin uptake; monoamine oxidase A (MAOA), the primary enzyme for serotonin catabolism; and five serotonin receptors previously shown to influence social behavior: 5-HT1A, 5-HT1B, 5-HT2A, 5-HT2C, and 5-HT4. Our analyses documented a significant increase in 5-HT2A receptor levels in the ASB + SUD group compared to SUD-only controls. Furthermore, TPH2 levels were significantly reduced in the SUD group (including SUD only and ASB + SUD) compared to unaffected controls. No difference was detected in the expression of any other serotonergic target. These results are in keeping with previous evidence showing high 5-HT2A receptor binding in the OFC of pathologically aggressive individuals and point to this molecule as a potential target for ASB treatment.


Subject(s)
Antisocial Personality Disorder , Prefrontal Cortex , Receptor, Serotonin, 5-HT2A , Adult , Humans , Male , Middle Aged , Young Adult , Antisocial Personality Disorder/complications , Antisocial Personality Disorder/enzymology , Antisocial Personality Disorder/metabolism , Autopsy , Monoamine Oxidase/metabolism , Prefrontal Cortex/enzymology , Prefrontal Cortex/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Substance-Related Disorders/complications , Substance-Related Disorders/enzymology , Substance-Related Disorders/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Aggression , Case-Control Studies
20.
West Afr J Med ; 39(8): 874-884, 2022 Aug 31.
Article in English | MEDLINE | ID: mdl-36063103

ABSTRACT

BACKGROUND: Drug addiction is a chronic biochemical drug use disorder that affects the human brain and behaviour, and leads to an uncontrollable use of a licit or illicit drug. Drug addiction can commence, usually in the young, with the use of a non-medical or a recreational drug in social gatherings, which becomes more frequent over a period of time. It is associated with incremental doses of the drug in order to achieve a state of euphoria. Addiction to drugs has been identified as a relevant social and health problem presenting a risk to public health, especially with regards to communicable diseases (e.g., HIV and AIDS, hepatitis B or C, tuberculosis, and sexually transmitted infections). OBJECTIVE: The objectives of the study were to discuss the neural mechanisms and circuitry responsible for the development and maintenance of addiction. It also examined the cycle of drug addiction and the associated encephalic regions and pathways. METHOD: The search strategy used for the review employed electronic databases in the search for relevant research articles, and they included Scopus, PubMed, Science Direct, Google Scholar, Springer, and the Directory of Open Access Journals. Articles on drug addiction were identified and reviewed for selection. The keywords used in the search were: Neurobiology and [Drug Abuse], Neurobiology and [Drug Addiction], Neurobiology and [Drug Misuse], Neurobiology and [Substance Abuse], Neurobiology and [Substance Misuse], Neural Mechanisms and [Drug Abuse], Neural Mechanisms and [Drug Addiction], and Neural Mechanisms and [Drug Use Disorders]. The search was also aided by scanning the references of identified journal articles. Works identified (86 in number) were those written in English and published between 1996 and 2020. RESULT: One hundred and fifty journal articles and other materials were identified. Eighty-six (86) articles and other works were extracted and reviewed after screening of the titles. abstracts and keywords, and in tandem with the selection criteria. Findings show that addiction is a complex neurobiochemical disorder that is learnt and stored in the brain as memory. The disorder alters the cyto-architecture of the brain and its functions. Relapse from drug addiction is a common occurrence. It is preventable and can be treated, although no single modality of treatment fits all forms of drug addiction. CONCLUSION: Addiction to drugs is the most severe form of drug use disorder. The abuse of drugs and psychoactive substances can harm the security of all societies, including the rule-of-law. It inflicts pain and suffering to individuals and families alike, and may eventuate in deaths. Repetitive use of an addictive drug alters the way and manner the brain perceives pleasure. Drugs of abuse induce structural changes in the neurones in the brain, and in turn, alter the neurotransmitter function, and thereby, create moods and other sensations. These anatomical and physiological changes in the brain may progress even after the stoppage of drugs.


CONTEXTE: La toxicomanie est un trouble biochimique chronique de la consommation de drogues qui affecte le cerveau et le comportement humain et conduit à une consommation incontrôlable d'une drogue licite ou illicite. La toxicomanie peut commencer, généralement chez les jeunes, par la consommation d'une drogue à usage non médical ou récréatif lors de rencontres sociales, qui devient plus fréquente au fil du temps. Elle est associée à des doses progressives de la drogue afin d'atteindre un état d'euphorie. La dépendance aux drogues a été identifiée comme un problème social et sanitaire important, présentant un risque pour la santé publique, notamment en matière des maladies transmissibles (par exemple, le VIH et le sida, l'hépatite B ou C) la tuberculose et les infections sexuellement transmissibles). OBJECTIF: Les objectifs de l'étude étaient de discuter des mécanismes et circuits neuronaux responsables du développement du entretien de la dépendance. Elle a également examiné le cycle de la toxicomanie et les régions et voies encéphaliques associées. MÉTHODE: La stratégie de recherche utilisée pour l'examen a fait appel à des bases de données électroniques pour la recherche d'articles pertinent et ils consistent Scopus, PubMed, Science Direct, Google Scholar, Springer et le Directory of Open Access Journals. Les articles sur la toxicomanie ont été identifiés et examinés pour être sélectionnés. Les mots-clés utilisés dans la recherche étaient : Neurobiologie et [Abus de drogues], Neurobiologie et [Toxicomanie], neurobiologie et [abus de drogues], neurobiologie et [abus de substances], neurobiologie et [abus de substances], Mécanismes neuraux et [abus de drogues], Mécanismes neuraux et [toxicomanie]. Mécanismes neuraux et [troubles liés à la consommation de drogues]. La recherche a également été facilitée par l'analyse des références des articles de revues identifiés. Les ouvrages identifiés (au nombre de 86) étaient ceux rédigés en anglais et publiés entre 1996 et 2020. RÉSULTAT: Cent cinquante articles de journaux et autres matériaux ont été identifiés. Quatre-vingt-six (86) articles et autres travaux ont été extraits et examinés après avoir passé en revue les titres, les résumés et les mots-clés, et en tandem avec les critères de sélection.Les résultats montrent que l'addiction est un trouble neurobiochimique complexe qui est appris et stocké dans le cerveau en tant que mémoire. Ce trouble modifie la cyto-architecture du cerveau et ses fonctions. La rechute de la toxicomanie est un phénomène courant. Elle est évitable et peut être traitée, bien qu'il n'existe pas de modalité unique de traitement pour toutes les formes de toxicomanie. CONCLUSION: La toxicomanie est la forme la plus grave de trouble lié à l'usage de drogues. L'abus de drogues et de substances psychoactives peut nuire à la sécurité de toutes les sociétés, y compris à l'État de droit. Il inflige de la douleur et de la souffrance aux individus et aux familles, et peut entraîner la mort. L'usage répétitif d'une drogue addictive modifie la manière dont le cerveau perçoit le plaisir. Les drogues d'abus induisent des changements structurels dans les neurones du cerveau et modifient à leur tour la fonction des neurotransmetteurs et créent ainsi des humeurs et d'autres sensations. Ces changements anatomiques et physiologiques dans le cerveau peuvent progresser même après l'arrêt de la drogue. MOTS CLÉS: Neurone, Compulsivité, Impulsivité, Récompense, Renforcement, Dépendance, Tolerance.


Subject(s)
Neurobiology , Substance-Related Disorders , Brain/metabolism , Humans , Learning , Recurrence , Substance-Related Disorders/epidemiology , Substance-Related Disorders/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...