Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 502
Filter
1.
Addict Biol ; 29(5): e13400, 2024 May.
Article in English | MEDLINE | ID: mdl-38706091

ABSTRACT

Substance use disorders are characterized by inhibition deficits related to disrupted connectivity in white matter pathways, leading via interaction to difficulties in resisting substance use. By combining neuroimaging with smartphone-based ecological momentary assessment (EMA), we questioned how biomarkers moderate inhibition deficits to predict use. Thus, we aimed to assess white matter integrity interaction with everyday inhibition deficits and related resting-state network connectivity to identify multi-dimensional predictors of substance use. Thirty-eight patients treated for alcohol, cannabis or tobacco use disorder completed 1 week of EMA to report substance use five times and complete Stroop inhibition testing twice daily. Before EMA tracking, participants underwent resting state functional MRI and diffusion tensor imaging (DTI) scanning. Regression analyses were conducted between mean Stroop performances and whole-brain fractional anisotropy (FA) in white matter. Moderation testing was conducted between mean FA within significant clusters as moderator and the link between momentary Stroop performance and use as outcome. Predictions between FA and resting-state connectivity strength in known inhibition-related networks were assessed using mixed modelling. Higher FA values in the anterior corpus callosum and bilateral anterior corona radiata predicted higher mean Stroop performance during the EMA week and stronger functional connectivity in occipital-frontal-cerebellar regions. Integrity in these regions moderated the link between inhibitory control and substance use, whereby stronger inhibition was predictive of the lowest probability of use for the highest FA values. In conclusion, compromised white matter structural integrity in anterior brain systems appears to underlie impairment in inhibitory control functional networks and compromised ability to refrain from substance use.


Subject(s)
Diffusion Tensor Imaging , Inhibition, Psychological , Magnetic Resonance Imaging , White Matter , Humans , White Matter/diagnostic imaging , White Matter/pathology , Male , Female , Adult , Ecological Momentary Assessment , Substance-Related Disorders/physiopathology , Substance-Related Disorders/diagnostic imaging , Stroop Test , Alcoholism/physiopathology , Alcoholism/diagnostic imaging , Brain/diagnostic imaging , Brain/physiopathology , Middle Aged , Tobacco Use Disorder/physiopathology , Tobacco Use Disorder/diagnostic imaging , Marijuana Abuse/physiopathology , Marijuana Abuse/diagnostic imaging , Corpus Callosum/diagnostic imaging , Corpus Callosum/pathology , Smartphone , Neural Pathways/diagnostic imaging , Neural Pathways/physiopathology , Anisotropy , Young Adult
2.
Addict Biol ; 29(5): e13395, 2024 May.
Article in English | MEDLINE | ID: mdl-38709211

ABSTRACT

The brain mechanisms underlying the risk of cannabis use disorder (CUD) are poorly understood. Several studies have reported changes in functional connectivity (FC) in CUD, although none have focused on the study of time-varying patterns of FC. To fill this important gap of knowledge, 39 individuals at risk for CUD and 55 controls, stratified by their score on a self-screening questionnaire for cannabis-related problems (CUDIT-R), underwent resting-state functional magnetic resonance imaging. Dynamic functional connectivity (dFNC) was estimated using independent component analysis, sliding-time window correlations, cluster states and meta-state indices of global dynamics and were compared among groups. At-risk individuals stayed longer in a cluster state with higher within and reduced between network dFNC for the subcortical, sensory-motor, visual, cognitive-control and default-mode networks, relative to controls. More globally, at-risk individuals had a greater number of meta-states and transitions between them and a longer state span and total distance between meta-states in the state space. Our findings suggest that the risk of CUD is associated with an increased dynamic fluidity and dynamic range of FC. This may result in altered stability and engagement of the brain networks, which can ultimately translate into altered cortical and subcortical function conveying CUD risk. Identifying these changes in brain function can pave the way for early pharmacological and neurostimulation treatment of CUD, as much as they could facilitate the stratification of high-risk individuals.


Subject(s)
Brain , Connectome , Magnetic Resonance Imaging , Marijuana Abuse , Humans , Male , Female , Marijuana Abuse/physiopathology , Marijuana Abuse/diagnostic imaging , Brain/physiopathology , Brain/diagnostic imaging , Young Adult , Adult , Case-Control Studies , Nerve Net/physiopathology , Nerve Net/diagnostic imaging , Default Mode Network/physiopathology , Default Mode Network/diagnostic imaging , Adolescent
3.
J Psychiatr Res ; 174: 121-128, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38626562

ABSTRACT

Biological sex differences in Cannabis Use Disorder (CUD) progression, cannabis withdrawal severity, and pharmacotherapy response have been reported, suggesting that CUD mechanisms may differ by sex. Drug cue reactivity is an established predictor of drug use behavior, but the literature on sex differences in drug cue reactivity is mixed, including in CUD. One possible moderator of sex differences in drug cue reactivity is hormonal contraceptive (HC) use. The aim of the present study was to test whether sex differences in neural cannabis cue reactivity and craving varied by female HC use in a CUD sample. As part of a larger study, 152 adults reporting frequent cannabis use completed a drug cue reactivity task during electrocenphalogram recording. Late positive potential (LPP) amplitude modulation by cannabis cues was used to measure neural cue reactivity. Craving after the cue reactivity task was also assessed. Males (n = 74) and naturally-cycling females (n = 26), who did not differ from each other, showed significantly greater LPP enhancement to cannabis vs. neutral cues compared to HC-using females (n = 52), an effect mostly driven by neutral cues. Craving was significantly higher in naturally-cycling but not HC-using females compared to males, but only in covariate-unadjusted analyses. Exploratory analyses of HC and menstrual phase characteristics indicate a progesterone-related mechanism may underlie HC effects on cannabis cue reactivity. The present study's results suggest that mixed findings on drug cue reactivity sex differences may be due to variability in HC use, which has implications for sex-specific models of CUD progression and treatment.


Subject(s)
Cues , Marijuana Abuse , Sex Characteristics , Humans , Female , Male , Adult , Marijuana Abuse/physiopathology , Young Adult , Craving/physiology , Craving/drug effects , Adolescent , Electroencephalography
4.
J Neural Transm (Vienna) ; 131(5): 563-580, 2024 05.
Article in English | MEDLINE | ID: mdl-38570361

ABSTRACT

Over 50 million Americans endure chronic pain where many do not receive adequate treatment and self-medicate to manage their pain by taking substances like opioids and cannabis. Research has shown high comorbidity between chronic pain and substance use disorders (SUD) and these disorders share many common neurobiological underpinnings, including hypodopaminergic transmission. Drugs commonly used for self-medication such as opioids and cannabis relieve emotional, bothersome components of pain as well as negative emotional affect that perpetuates misuse and increases the risk of progressing towards drug abuse. However, the causal effect between chronic pain and the development of SUDs has not been clearly established. In this review, we discuss evidence that affirms the proposition that chronic pain is a risk factor for the development of opioid and cannabis use disorders by outlining the clinical evidence and detailing neurobiological mechanisms that link pain and drug misuse. Central to the link between chronic pain and opioid and cannabis misuse is hypodopaminergic transmission and the modulation of dopamine signaling in the mesolimbic pathway by opioids and cannabis. Moreover, we discuss the role of kappa opioid receptor activation and neuroinflammation in the context of dopamine transmission, their contribution to opioid and cannabis withdrawal, along with potential new treatments.


Subject(s)
Analgesics, Opioid , Chronic Pain , Opioid-Related Disorders , Humans , Chronic Pain/drug therapy , Chronic Pain/physiopathology , Analgesics, Opioid/adverse effects , Animals , Marijuana Abuse/complications , Marijuana Abuse/physiopathology
5.
J Psychopharmacol ; 38(5): 471-480, 2024 May.
Article in English | MEDLINE | ID: mdl-38418434

ABSTRACT

BACKGROUND: Regular cannabis is known to impact higher-order cognitive processes such as attention, but far less is known regarding cognitive flexibility, a component of executive function. Moreover, whether such changes are related to aberrations in the neural oscillatory dynamics serving flexibility remains poorly understood. AIMS: Quantify the neural oscillatory dynamics serving cognitive flexibility by having participants complete a task-switching paradigm during magnetoencephalography (MEG). Probe whole-brain maps to identify alterations in chronic cannabis users relative to nonusers and determine how these alterations relate to the degree of cannabis use involvement. METHODS: In all, 25 chronic cannabis users and 30 demographically matched nonuser controls completed neuropsychological testing, an interview regarding their substance use, a urinalysis, and a task switch paradigm during MEG. Time-frequency windows of interest were identified using a data-driven statistical approach and these were imaged using a beamformer. Whole-brain neural switch cost maps were computed by subtracting the oscillatory maps of the no-switch condition from the switch condition per participant. These were examined for group differences. RESULTS: Cannabis users had weaker theta switch cost responses in the dorsolateral and dorsomedial prefrontal cortices, while nonusers showed the typical pattern of greater recruitment during switch relative to no switch trials. In addition, theta activity in the dorsomedial prefrontal cortex was significantly correlated with cannabis use involvement. CONCLUSIONS: Cannabis users exhibited altered theta switch cost activity compared to nonusers in prefrontal cortical regions, which are critical for cognitive flexibility. This activity scaled with cannabis use involvement, indicating a link between cannabis use and aberrant oscillatory activity underlying cognitive flexibility.


Subject(s)
Executive Function , Magnetoencephalography , Humans , Male , Female , Adult , Young Adult , Executive Function/physiology , Executive Function/drug effects , Cognition/drug effects , Cognition/physiology , Neuropsychological Tests , Attention/drug effects , Attention/physiology , Marijuana Abuse/physiopathology , Prefrontal Cortex/physiopathology , Prefrontal Cortex/drug effects , Brain/physiopathology , Brain/drug effects , Case-Control Studies
6.
Adicciones (Palma de Mallorca) ; 35(1): 33-46, 2023. tab
Article in Spanish | IBECS | ID: ibc-215863

ABSTRACT

El sistema cannabinoide se ha asociado con varios trastornos psiquiátricos como la esquizofrenia y las adicciones. Diversos estudios hanobservado que algunos polimorfismos del receptor cannabinoide tipo2 (CNR2), del receptor cannabinoide tipo 1 (CNR1) y del gen de la enzima amido hidrolasa de ácidos grasos (FAAH) pueden ser factores deriesgo de estos trastornos. Hemos analizado diversos polimorfismos delsistema cannabinoide en pacientes diagnosticados de esquizofrenia sintrastorno por uso de sustancias (n = 379), esquizofrenia con trastornopor uso de cannabis (n = 124), dependientes de cannabis sin psicosisasociada (n = 71) y un grupo de control (316) procedentes de diversoshospitales y centros de asistencia sanitaria españoles. Hemos encontrado una asociación entre los polimorfismos rs35761398 y rs12744386 delCNR2 con la presencia de esquizofrenia y trastorno por uso de cannabis comórbido y una pérdida de heterocigosidad en el polimorfismors324420 del gen FAAH con la dependencia de cannabis en poblaciónespañola. Los polimorfismos rs35761398 y rs12744386 en CNR2 son factores de riesgo para esquizofrenia en sujetos dependientes de cannabis.La pérdida de heterocigosidad en el polimorfismo rs324420 en el genFAAH es un factor de riesgo para la dependencia de cannabis. (AU)


The endocannabinoid system has been associated with various psychiatric disorders, such as schizophrenia or addictive disorders. Recent studies have found that some polymorphisms in the cannabinoid receptortype 2 (CNR2), cannabinoid receptor type 1 (CNR1) and fatty acid amide hydrolase (FAAH) genes could play an important role as risk factorsin the etiology of these diseases. We analysed different cannabinoidgene polimorphisms from non-substance using patients diagnosed withschizophrenia (n = 379), schizophrenic patients with cannabis use disorders (n = 124), cannabis users who did not have psychoses (n = 71),and 316 controls from various Spanish hospitals and health centres.We found a statistical association between polymorphisms rs35761398and rs12744386 in the CNR2 gene and comorbidity of schizophreniaand cannabis dependence, as well as an association between loss ofheterozygosity (overdominance) for polymorphism rs324420 in theFAAH gene and cannabis dependence in a Spanish population sample.The rs35761398 and rs12744386 polymorphisms in the CNR2 gene aregenetic risk factors for schizophrenia in cannabis-dependent subjects.Loss of heterozygosity for polymorphism rs324420 in the FAAH gene isa genetic risk factor for cannabis dependence in this population. (AU)


Subject(s)
Humans , Marijuana Abuse/physiopathology , Schizophrenia/genetics , Schizophrenia/physiopathology , Schizophrenia/therapy , Cannabinoid Receptor Agonists , Spain , Polymorphism, Genetic , Genes, MDR , Amidohydrolases/genetics
7.
J Clin Psychopharmacol ; 41(6): 623-628, 2021.
Article in English | MEDLINE | ID: mdl-34735097

ABSTRACT

PURPOSE/BACKGROUND: The link between substances of abuse, impulsivity, and violence in psychotic patients remains unclear. This study aims at unraveling whether cannabis use disorder is associated with violent and/or psychotic behavior in patients who are hospitalized in a high-security hospital. METHODS/PROCEDURES: We conducted a cross-sectional retrospective study in 124 patients with schizophrenia spectrum disorders admitted to a high-security hospital. Lifetime violent behavior was assessed using the History of Aggressive Behavior Form-Subject of the MacArthur Violence Risk Assessment Study and impulsivity using the Psychopathy Checklist-Revised (considering items: "proneness to boredom," "lack of self-control," and "impulsive thoughtless gestures"). Substance use disorder was diagnosed according to Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition criteria. Positive and Negative Syndrome Scale was also administered. FINDINGS/RESULTS: Violent and nonviolent psychotic patients showed similar prevalence of cannabis use disorder. Alcohol and cocaine use disorders were more prevalent among violent psychotic patients. Cannabis use disorder was not associated with any dimension of impulsivity, whereas alcohol use disorder was positively correlated to impulsive thoughtless gestures (standardized ß = 0.213, P = 0.027) and cocaine use disorder with proneness to boredom (standardized ß = 0.290, P = 0.002). Finally, logistic regression analysis revealed that, unlike cannabis and cocaine use disorders, alcohol use disorder (odds ratio, 3.964; 95% confidence interval, 1.729-9.087; P = 0.001) was a factor associated with violence. IMPLICATIONS/CONCLUSIONS: These findings show that cannabis and alcohol are largely abused and coabused by psychotic patients with a propsensity for violence, but only alcohol is associated with impulsive and violent behavior. Therefore, especially alcohol abuse should be seriously considered by practitioners when evaluating the dangerousness of patients with schizophrenia spectrum disorders.


Subject(s)
Alcoholism , Impulsive Behavior , Marijuana Abuse , Schizophrenia , Violence , Adult , Alcoholism/complications , Alcoholism/epidemiology , Alcoholism/physiopathology , Comorbidity , Cross-Sectional Studies , Hospitals, Psychiatric , Humans , Impulsive Behavior/drug effects , Impulsive Behavior/physiology , Male , Marijuana Abuse/complications , Marijuana Abuse/epidemiology , Marijuana Abuse/physiopathology , Middle Aged , Retrospective Studies , Schizophrenia/complications , Schizophrenia/epidemiology , Schizophrenia/physiopathology
8.
J Psychopharmacol ; 35(11): 1349-1355, 2021 11.
Article in English | MEDLINE | ID: mdl-34694178

ABSTRACT

BACKGROUND AND AIMS: Cannabis is a commonly used recreational drug in young adults. The worldwide prevalence in 18- to 25-year-olds is approximately 35%. Significant differences in cognitive performance have been reported previously for groups of cannabis users. However, the groups are often heterogeneous in terms of cannabis use. Here, we study daily cannabis users with a confirmed diagnosis of cannabis use disorder (CUD) to examine cognitive performance on measures of memory, executive function and risky decision-making. METHODS: Forty young adult daily cannabis users with diagnosed CUD and 20 healthy controls matched for sex and premorbid intelligence quotient (IQ) were included. The neuropsychological battery implemented was designed to measure multiple modes of memory (visual, episodic and working memory), risky decision-making and other domains of executive function using subtests from the Cambridge Neuropsychological Test Automated Battery (CANTAB). RESULTS: Our results showed that young adult daily cannabis users with CUD perform significantly poorer on tasks of visual and episodic memory compared with healthy controls. In addition, executive functioning was associated with the age of onset. CONCLUSIONS: Further research is required to determine whether worse performance in cognition results in cannabis use or is a consequence of cannabis use. Chronic heavy cannabis use during a critical period of brain development may have a particularly negative impact on cognition. Research into the persistence of cognitive differences and how they relate to functional outcomes such as academic/career performance is required.


Subject(s)
Cognitive Dysfunction/physiopathology , Marijuana Abuse/physiopathology , Psychomotor Performance/physiology , Adult , California , Cognitive Dysfunction/etiology , Female , Humans , Male , Marijuana Abuse/complications , Neuropsychological Tests , Young Adult
9.
Addict Biol ; 26(5): e13032, 2021 09.
Article in English | MEDLINE | ID: mdl-33951262

ABSTRACT

Sensorimotor dysfunction has been previously reported in persons with cannabis dependence. Such individuals can exhibit increased levels of neurological soft signs (NSS), particularly involving motor coordination and sensorimotor integration. Whether such abnormalities may also apply to non-dependent individuals with heavy cannabis use (HCU) is unknown, as much as the neural correlates underlying such deficits. In this study, we investigated associations between NSS and gray matter volume (GMV) in males with HCU and male controls. Twenty-four persons with HCU and 17 controls were examined using standardized assessment of NSS and structural magnetic resonance imaging (MRI) at 3 T. GMV was calculated using voxel-based morphometry algorithms provided by the Computational Anatomy Toolbox (CAT12). Individuals with HCU showed higher NSS total scores compared to controls. In particular, significant NSS-subdomain effects were found for "motor coordination" (MoCo), "complex motor tasks" (CoMT), and "hard signs" (HS) expression in HCU (p < 0.05, Bonferroni-corrected). Compared to controls, persons with HCU showed significant NSS/GMV interactions in putamen and inferior frontal cortex (MoCo), right cerebellum (CoMT) and middle and superior frontal cortices, and bilateral precentral cortex and thalamus (HS). In between-group analyses, individuals with HCU showed lower GMV in the right anterior orbital and precentral gyrus, as well as higher GMV in the right superior frontal gyrus and left supplementary motor cortex compared to controls. The data support the notion of abnormal sensorimotor performance associated with HCU. The data also provide a neuromechanistic understanding of such deficits, particularly with respect to aberrant cortical-thalamic-cerebellar-cortical circuit.


Subject(s)
Marijuana Abuse/physiopathology , Psychomotor Performance/drug effects , Adolescent , Adult , Brain/physiopathology , Cannabis , Cerebellum/pathology , Gray Matter/pathology , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Motor Cortex , Prefrontal Cortex/pathology , Young Adult
10.
J Neurochem ; 157(5): 1674-1696, 2021 06.
Article in English | MEDLINE | ID: mdl-33891706

ABSTRACT

Cannabis sativa is the most widely used illicit drug in the world. Its main psychoactive component is delta-9-tetrahydrocannabinol (THC), one of over 100 phytocannabinoid compounds produced by the cannabis plant. THC is the primary compound that drives cannabis abuse potential and is also used and prescribed medically for therapeutic qualities. Despite its therapeutic potential, a significant subpopulation of frequent cannabis or THC users will develop a drug use syndrome termed cannabis use disorder. Individuals suffering from cannabis use disorder exhibit many of the hallmarks of classical addictions including cravings, tolerance, and withdrawal symptoms. Currently, there are no efficacious treatments for cannabis use disorder or withdrawal symptoms. This makes both clinical and preclinical research on the neurobiological mechanisms of these syndromes ever more pertinent. Indeed, basic research using animal models has provided valuable evidence of the neural molecular and cellular actions of cannabis that mediate its behavioral effects. One of the main components being central action on the cannabinoid type-one receptor and downstream intracellular signaling related to the endogenous cannabinoid system. Back-translational studies have provided insight linking preclinical basic and behavioral biology research to better understand symptoms observed at the clinical level. This narrative review aims to summarize major research elucidating the molecular, cellular, and behavioral manifestations of cannabis/THC use that play a role in cannabis use disorder and withdrawal.


Subject(s)
Endocannabinoids , Marijuana Abuse/physiopathology , Marijuana Smoking/physiopathology , Receptors, Cannabinoid , Substance Withdrawal Syndrome/physiopathology , Animals , Dronabinol/pharmacology , Drug Tolerance , Humans , Marijuana Abuse/psychology , Marijuana Smoking/psychology , Substance Withdrawal Syndrome/psychology
11.
Article in English | MEDLINE | ID: mdl-33677045

ABSTRACT

The expanding legalization of cannabis across the United States is associated with increases in cannabis use, and accordingly, an increase in the number and severity of individuals with cannabis use disorder (CUD). The lack of FDA-approved pharmacotherapies and modest efficacy of psychotherapeutic interventions means that many of those who seek treatment for CUD relapse within the first few months. Consequently, there is a pressing need for innovative, evidence-based treatment development for CUD. Preliminary evidence suggests that repetitive transcranial magnetic stimulation (rTMS) may be a novel, non-invasive therapeutic neuromodulation tool for the treatment of a variety of substance use disorders (SUDs), including recently receiving FDA clearance (August 2020) for use as a smoking cessation aid in tobacco cigarette smokers. However, the potential of rTMS for CUD has not yet been reviewed. This paper provides a primer on therapeutic neuromodulation techniques for SUDs, with a particular focus on reviewing the current status of rTMS research in people who use cannabis. Lastly, future directions are proposed for rTMS treatment development in CUD, with suggestions for study design parameters and clinical endpoints based on current gold-standard practices for therapeutic neuromodulation research.


Subject(s)
Brain/physiopathology , Marijuana Abuse/therapy , Transcranial Magnetic Stimulation/methods , Brain/diagnostic imaging , Functional Neuroimaging , Humans , Marijuana Abuse/diagnostic imaging , Marijuana Abuse/physiopathology , Treatment Outcome
12.
Int Rev Neurobiol ; 157: 143-193, 2021.
Article in English | MEDLINE | ID: mdl-33648669

ABSTRACT

Cannabis use is increasing among some demographics in the United States and is tightly linked to anxiety, trauma, and stress reactivity at the epidemiological and biological level. Stress-coping motives are highly cited reasons for cannabis use. However, with increased cannabis use comes the increased susceptibility for cannabis use disorder (CUD). Indeed, CUD is highly comorbid with posttraumatic stress disorder (PTSD). Importantly, endogenous cannabinoid signaling systems play a key role in the regulation of stress reactivity and anxiety regulation, and preclinical data suggest deficiencies in this signaling system could contribute to the development of stress-related psychopathology. Furthermore, endocannabinoid deficiency states, either pre-existing or induced by trauma exposure, could provide explanatory insights into the high rates of comorbid cannabis use in patients with PTSD. Here we review clinical and preclinical literature related to the cannabis use-PTSD comorbidity, the role of endocannabinoids in the regulation of stress reactivity, and potential therapeutic implications of recent work in this area.


Subject(s)
Marijuana Abuse , Stress Disorders, Post-Traumatic , Comorbidity , Humans , Marijuana Abuse/epidemiology , Marijuana Abuse/physiopathology , Marijuana Abuse/therapy , Stress Disorders, Post-Traumatic/epidemiology , Stress Disorders, Post-Traumatic/physiopathology , Stress Disorders, Post-Traumatic/therapy , United States/epidemiology
13.
Toxins (Basel) ; 13(2)2021 02 05.
Article in English | MEDLINE | ID: mdl-33562446

ABSTRACT

For thousands of years, Cannabis sativa has been utilized as a medicine and for recreational and spiritual purposes. Phytocannabinoids are a family of compounds that are found in the cannabis plant, which is known for its psychotogenic and euphoric effects; the main psychotropic constituent of cannabis is Δ9-tetrahydrocannabinol (Δ9-THC). The pharmacological effects of cannabinoids are a result of interactions between those compounds and cannabinoid receptors, CB1 and CB2, located in many parts of the human body. Cannabis is used as a therapeutic agent for treating pain and emesis. Some cannabinoids are clinically applied for treating chronic pain, particularly cancer and multiple sclerosis-associated pain, for appetite stimulation and anti-emesis in HIV/AIDS and cancer patients, and for spasticity treatment in multiple sclerosis and epilepsy patients. Medical cannabis varies from recreational cannabis in the chemical content of THC and cannabidiol (CBD), modes of administration, and safety. Despite the therapeutic effects of cannabis, exposure to high concentrations of THC, the main compound that is responsible for most of the intoxicating effects experienced by users, could lead to psychological events and adverse effects that affect almost all body systems, such as neurological (dizziness, drowsiness, seizures, coma, and others), ophthalmological (mydriasis and conjunctival hyperemia), cardiovascular (tachycardia and arterial hypertension), and gastrointestinal (nausea, vomiting, and thirst), mainly associated with recreational use. Cannabis toxicity in children is more concerning and can cause serious adverse effects such as acute neurological symptoms (stupor), lethargy, seizures, and even coma. More countries are legalizing the commercial production and sale of cannabis for medicinal use, and some for recreational use as well. Liberalization of cannabis laws has led to increased incidence of toxicity, hyperemesis syndrome, lung disease cardiovascular disease, reduced fertility, tolerance, and dependence with chronic prolonged use. This review focuses on the potential therapeutic effects of cannabis and cannabinoids, as well as the acute and chronic toxic effects of cannabis use on various body systems.


Subject(s)
Cannabinoids/therapeutic use , Cannabis , Medical Marijuana/therapeutic use , Nervous System/drug effects , Plants, Toxic , Animals , Cannabinoids/adverse effects , Cannabinoids/isolation & purification , Cannabis/adverse effects , Humans , Marijuana Abuse/metabolism , Marijuana Abuse/physiopathology , Marijuana Abuse/psychology , Medical Marijuana/adverse effects , Nervous System/metabolism , Nervous System/physiopathology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/physiopathology , Neurotoxicity Syndromes/psychology , Plants, Toxic/adverse effects , Receptors, Cannabinoid/drug effects , Receptors, Cannabinoid/metabolism , Signal Transduction
14.
Article in English | MEDLINE | ID: mdl-31964646

ABSTRACT

Cannabis sativa, like all known drugs of abuse, leads to increased dopamine activation within the mesolimbic pathway. Consequent dopamine release within terminal regions of the striatum is a powerful mediator of reward and reinforcement and patterned dopamine release is critical for associative learning processes that are fundamentally involved in addiction. The endocannabinoid system modulates dopamine release at multiple sites, and the receptors, endogenous ligands, and synthetic and metabolic enzymes of the endocannabinoid system may provide key targets for pharmacotherapies to treat disorders of motivation including addiction. Disrupting endocannabinoid signaling decreases drug-induced increases in dopamine release as well those dopamine events evoked by conditioned stimuli during reward seeking. Advances in recording techniques for dopamine are allowing unprecedented examinations of these two interacting systems and elucidating the mechanisms of endocannabinoid modulation of dopamine release in reward and addiction.


Subject(s)
Brain/drug effects , Cannabinoid Receptor Agonists/pharmacology , Dopamine/metabolism , Marijuana Abuse/metabolism , Reward , Animals , Brain/physiology , Dronabinol/pharmacology , Endocannabinoids/physiology , Humans , Marijuana Abuse/physiopathology , Marijuana Abuse/psychology , Mice , Signal Transduction/drug effects , Signal Transduction/physiology , Substance Withdrawal Syndrome/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
15.
Addict Biol ; 26(1): e12870, 2021 01.
Article in English | MEDLINE | ID: mdl-31865628

ABSTRACT

Cannabis is the most commonly used illicit drug in the world. However, because of a changing legal landscape and rising interest in therapeutic utility, there is an increasing trend in (long-term) use and possibly cannabis impairment. Importantly, a growing body of evidence suggests that regular cannabis users develop tolerance to the impairing, as well as the rewarding, effects of the drug. However, the neuroadaptations that may underlie cannabis tolerance remain unclear. Therefore, this double-blind, randomized, placebo-controlled, cross-over study assessed the acute influence of cannabis on the brain and behavioral outcomes in two distinct cannabis user groups. Twelve occasional and 12 chronic cannabis users received acute doses of cannabis (300-µg/kg delta-9-tetrahydrocannabinol) and placebo and underwent ultrahigh field functional magnetic resonance imaging and magnetic resonance spectroscopy. In occasional users, cannabis induced significant neurometabolic alterations in reward circuitry, namely, decrements in functional connectivity and increments in striatal glutamate concentrations, which were associated with increases in subjective high and decreases in performance on a sustained attention task. Such changes were absent in chronic users. The finding that cannabis altered circuitry and distorted behavior in occasional, but not chronic users, suggests reduced responsiveness of the reward circuitry to cannabis intoxication in chronic users. Taken together, the results suggest a pharmacodynamic mechanism for the development of tolerance to cannabis impairment, of which is important to understand in the context of the long-term therapeutic use of cannabis-based medications, as well as in the context of public health and safety of cannabis use when performing day-to-day operations.


Subject(s)
Drug Tolerance , Marijuana Abuse/physiopathology , Reward , Attention , Brain/physiopathology , Cannabis , Cognition/drug effects , Cross-Over Studies , Double-Blind Method , Dronabinol/pharmacology , Female , Humans , Magnetic Resonance Imaging , Male , Neuropsychological Tests , Psychomotor Performance/drug effects
16.
Addict Biol ; 26(1): e12885, 2021 01.
Article in English | MEDLINE | ID: mdl-32135572

ABSTRACT

Two of the most commonly used substances by adolescents in the United States are cannabis and alcohol. Cannabis use disorder (CUD) and alcohol use disorder (AUD) are associated with impairments in decision-making processes. One mechanism for impaired decision-making in these individuals is thought to be an inability to adequately represent future events during decision-making. In the current study involving 112 adolescents, we used a comparative optimism task to examine the relationship between relative severity of CUD/AUD (as indexed by the CUD/AUD Identification Tests [CUDIT/AUDIT]) and atypical function within neural systems underlying affect-based neural represenation future events. Greater CUDIT scores were negatively related to responses within subgenual anterior and posterior cingulate cortex when processing high-intensity potential future positive and negative events. There was also a particularly marked negative relationship between CUD symptoms and blood oxygen level-dependent (BOLD) responses within visual and premotor cortices to high-intensity, negatively valenced potential future events. However, AUD symptom severity was not associated with dysfunction within these brain regions. These data indicate that relative risk/severity of CUD is associated with reduced responsiveness to future high-intensity events. This may impair decision-making where future significant consequences should guide response choice.


Subject(s)
Alcoholism/physiopathology , Brain/physiopathology , Marijuana Abuse/physiopathology , Adolescent , Emotions , Female , Humans , Magnetic Resonance Imaging , Male , United States
17.
Addiction ; 116(1): 182-190, 2021 01.
Article in English | MEDLINE | ID: mdl-32285993

ABSTRACT

Cannabis is one of the world's most widely used recreational drugs and the second most commonly smoked substance. Research on cannabis and the lungs has been limited by its illegal status, the variability in strength and size of cannabis cigarettes (joints), and the fact that most cannabis users also smoke tobacco, making the effects difficult to separate. Despite these difficulties, the available evidence indicates that smoking cannabis causes bronchitis and is associated with changes in lung function. The pattern of effects is surprisingly different from that of tobacco. Whereas smoking cannabis appears to increase the risk of severe bronchitis at quite low exposure, there is no convincing evidence that this leads to chronic obstructive pulmonary disease. Instead, cannabis use is associated with increased central airway resistance, lung hyperinflation and higher vital capacity with little evidence of airflow obstruction or impairment of gas transfer. There are numerous reports of severe bullous lung disease and pneumothorax among heavy cannabis users, but convincing epidemiological data of an increased risk of emphysema or alveolar destruction are lacking. An association between cannabis and lung cancer remains unproven, with studies providing conflicting findings.


Subject(s)
Lung/physiopathology , Marijuana Abuse/physiopathology , Marijuana Smoking/physiopathology , Cannabis , Humans , Lung Diseases/physiopathology , Risk Factors
18.
Article in English | MEDLINE | ID: mdl-32791165

ABSTRACT

Cannabis is the most widely used illicit substance among adolescents, and adolescent cannabis use is associated with various neurocognitive deficits that can extend into adulthood. A growing body of evidence supports the hypothesis that adolescence encompasses a vulnerable period of development where exposure to exogenous cannabinoids can alter the normative trajectory of brain maturation. In this review, we present an overview of studies of human and rodent models that examine lasting effects of adolescent exposure. We include evidence from meta-analyses, longitudinal, or cross-sectional studies in humans that consider age of onset as a factor that contributes to the behavioral dysregulation and altered structural or functional development in cannabis users. We also discuss evidence from preclinical rodent models utilizing well-characterized or innovative routes of exposure, investigating the effects of dose and timing to produce behavioral deficits or alterations on a neuronal and behavioral level. Multiple studies from both humans and animals provide contrasting results regarding the magnitude of residual effects. Combined evidence suggests that exposure to psychoactive cannabinoids during adolescence has the potential to produce subtle, but lasting, alterations in neurobiology and behavior.


Subject(s)
Brain/drug effects , Cannabinoids/administration & dosage , Cannabis/adverse effects , Marijuana Abuse/physiopathology , Adolescent , Brain/physiopathology , Humans
19.
Article in English | MEDLINE | ID: mdl-32860840

ABSTRACT

BACKGROUND: Cannabis is a neuromodulating substance that acts on central synaptic transmission. Regular cannabis use induces a decreased capacity for dopamine synthesis in the brain. The retina is considered an easy means of investigating dysfunctions of synaptic transmission in the brain. We have previously studied the impact of regular cannabis use on retinal function. Using the N95 wave of the pattern electroretinogram, we found a 6 ms-delayed ganglion cells response. Using the b-wave of the photopic flash electroretinogram, we found a 1 ms-delayed bipolar cells response. Here, we investigated amacrine cells function because these cells are located between the bipolar cells and the ganglion cells and contribute to amplifying the signal between these two layers of the retina. We tested the effect of regular cannabis use on these retinal dopaminergic cells. We assessed the role of these cells in amplifying the delay observed previously. METHODS: We recorded dark-adapted 3.0 flash ERG oscillatory potentials in 56 regular cannabis users and 29 healthy controls. The amplitude and implicit time of OP1, OP2, OP3 and OP4 were evaluated. RESULTS: Cannabis users showed a significant decrease in OP2 amplitude (p = 0.029, Mann-Whitney test) and OP3 amplitude (p = 0.024, Mann-Whitney test). No significant difference was found between the groups for OP1 and OP4 amplitude or for the implicit time of oscillatory potentials. CONCLUSIONS: These results reflect the impact of regular cannabis use on amacrine cells function. They highlight abnormalities in dopaminergic transmission and are similar to those found in Parkinson's disease. Oscillatory potentials could be used as markers of central dopaminergic modulation.


Subject(s)
Amacrine Cells/physiology , Dopaminergic Neurons/physiology , Electroretinography/methods , Marijuana Abuse/diagnostic imaging , Marijuana Abuse/physiopathology , Adult , Female , Humans , Male , Retina/diagnostic imaging , Retina/physiopathology , Young Adult
20.
Hum Brain Mapp ; 42(6): 1727-1741, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33340172

ABSTRACT

Although previous studies have highlighted associations of cannabis use with cognition and brain morphometry, critical questions remain with regard to the association between cannabis use and brain structural and functional connectivity. In a cross-sectional community sample of 205 African Americans (age 18-70) we tested for associations of cannabis use disorder (CUD, n = 57) with multi-domain cognitive measures and structural, diffusion, and resting state brain-imaging phenotypes. Post hoc model evidence was computed with Bayes factors (BF) and posterior probabilities of association (PPA) to account for multiple testing. General cognitive functioning, verbal intelligence, verbal memory, working memory, and motor speed were lower in the CUD group compared with non-users (p < .011; 1.9 < BF < 3,217). CUD was associated with altered functional connectivity in a network comprising the motor-hand region in the superior parietal gyri and the anterior insula (p < .04). These differences were not explained by alcohol, other drug use, or education. No associations with CUD were observed in cortical thickness, cortical surface area, subcortical or cerebellar volumes (0.12 < BF < 1.5), or graph-theoretical metrics of resting state connectivity (PPA < 0.01). In a large sample collected irrespective of cannabis used to minimize recruitment bias, we confirm the literature on poorer cognitive functioning in CUD, and an absence of volumetric brain differences between CUD and non-CUD. We did not find evidence for or against a disruption of structural connectivity, whereas we did find localized resting state functional dysconnectivity in CUD. There was sufficient proof, however, that organization of functional connectivity as determined via graph metrics does not differ between CUD and non-user group.


Subject(s)
Cerebral Cortex , Cognitive Dysfunction , Marijuana Abuse , Nerve Net , Adult , Black or African American , Aged , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Cognitive Dysfunction/physiopathology , Connectome , Cross-Sectional Studies , Female , Humans , Magnetic Resonance Imaging , Male , Marijuana Abuse/complications , Marijuana Abuse/diagnostic imaging , Marijuana Abuse/pathology , Marijuana Abuse/physiopathology , Middle Aged , Nerve Net/diagnostic imaging , Nerve Net/pathology , Nerve Net/physiopathology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...