Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 772
1.
Neuropsychopharmacology ; 49(6): 915-923, 2024 May.
Article En | MEDLINE | ID: mdl-38374364

Opioid use disorder is a chronic relapsing disorder encompassing misuse, dependence, and addiction to opioid drugs. Long term maintenance of associations between the reinforcing effects of the drug and the cues associated with its intake are a leading cause of relapse. Indeed, exposure to the salient drug-associated cues can lead to drug cravings and drug seeking behavior. The dorsal hippocampus (dHPC) and locus coeruleus (LC) have emerged as important structures for linking the subjective rewarding effects of opioids with environmental cues. However, their role in cue-induced reinstatement of opioid use remains to be further elucidated. In this study, we showed that chemogenetic inhibition of excitatory dHPC neurons during re-exposure to drug-associated cues significantly attenuates cue-induced reinstatement of morphine-seeking behavior. In addition, the same manipulation reduced reinstatement of sucrose-seeking behavior but failed to alter memory recall in the object location task. Finally, intact activity of tyrosine hydroxylase (TH) LC-dHPCTh afferents is necessary to drive cue induced reinstatement of morphine-seeking as inhibition of this pathway blunts cue-induced drug-seeking behavior. Altogether, these studies show an important role of the dHPC and LC-dHPCTh pathway in mediating cue-induced reinstatement of opioid seeking.


Cues , Drug-Seeking Behavior , Hippocampus , Locus Coeruleus , Self Administration , Animals , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , Male , Hippocampus/drug effects , Hippocampus/metabolism , Rats , Female , Drug-Seeking Behavior/drug effects , Drug-Seeking Behavior/physiology , Morphine/pharmacology , Morphine/administration & dosage , Rats, Sprague-Dawley , Neural Pathways/drug effects , Neural Pathways/physiology , Analgesics, Opioid/pharmacology , Analgesics, Opioid/administration & dosage , Opioid-Related Disorders/physiopathology , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Conditioning, Operant/drug effects , Conditioning, Operant/physiology
2.
Am J Drug Alcohol Abuse ; 50(2): 207-217, 2024 Mar 03.
Article En | MEDLINE | ID: mdl-38386811

Background: Numerous studies have highlighted the pivotal role of alterations in the monetary reward system in the development and maintenance of substance use disorder (SUD). Although these alterations have been well documented in various forms of SUD, the electrophysiological mechanisms specific to opioid use disorder (OUD) remain underexplored. Understanding these mechanisms is critical for developing targeted interventions and advancing theories of addiction specific to opioid use.Objectives: To explore abnormalities in monetary reward outcome processing in males with OUD. We hypothesized that control individuals would show higher feedback-related negativity (FRN) to losses, unlike those in the OUD group, where FRN to losses and gains would not differ significantly.Methods: Fifty-seven participants (29 male individuals with OUD [heroin] and 28 male controls) were evaluated. A combination of the monetary incentive delay task (MIDT) and event-related potential (ERP) technology was used to investigate electrophysiological differences in monetary reward feedback processing between the OUD and healthy control groups.Results: We observed a significant interaction between group (control vs. OUD) and monetary outcome (loss vs. gain), indicated by p < .05 and η2p = 0.116. Specifically, control participants showed stronger negative FRN to losses than gains (p < .05), unlike the OUD group (p > .05).Conclusion: This study's FRN data indicate that males with OUD show altered processing of monetary rewards, marked by reduced sensitivity to loss. These findings offer electrophysiological insights into why males with OUD may pursue drugs despite potential economic downsides.


Evoked Potentials , Opioid-Related Disorders , Reward , Humans , Male , Adult , Opioid-Related Disorders/physiopathology , Evoked Potentials/physiology , Case-Control Studies , Electroencephalography , Young Adult , Motivation , Feedback, Psychological/physiology
3.
J Psychosoc Nurs Ment Health Serv ; 62(1): 19-26, 2024 Jan.
Article En | MEDLINE | ID: mdl-37379124

The current systematic review synthesized available original research on objective and self-reported sleep health dimensions among adults aged 18 to 50 years in outpatient treatment for opioid use disorder (OUD). A comprehensive search was conducted using multiple electronic databases followed by screening 2,738 records published in English from the inception of each database to September 14, 2021. Quality was assessed with the Mixed Methods Appraisal Tool (version 2001). Fifty nine studies-50 descriptive (21 longitudinal, 18 cross-sectional, and 11 case control), seven interventional (five non-randomized), and two mixed/multi method designs-were included, comprising 18,195 adults with mean ages ranging from 23 to 49 years (mean age = 37.5 [SD = 5.9] years; 54.4% female) with OUD and 604 comparison participants without OUD. Studies were predominantly observational with various designs with self-report and objective measures with participants at various points in treatment. More work is needed to understand the multidimensional depth of sleep health in adults with OUD. Optimizing sleep health in adults with OUD may improve their addiction trajectory and should be a priority in practice and research. [Journal of Psychosocial Nursing and Mental Health Services, 62(1), 19-26.].


Mental Health Services , Opioid-Related Disorders , Sleep , Adult , Female , Humans , Male , Middle Aged , Young Adult , Analgesics, Opioid , Cross-Sectional Studies , Opioid-Related Disorders/physiopathology , Opioid-Related Disorders/therapy , Outpatients , Sleep/physiology
4.
Respir Physiol Neurobiol ; 296: 103800, 2022 02.
Article En | MEDLINE | ID: mdl-34626831

Infants born with neonatal opioid withdrawal syndrome (NOWS) can display abnormal cardiorespiratory patterns including tachypnea, tachycardia, and impaired ventilatory responses to hypoxia (HVR) and hypercapnia (HCVR). Chronic morphine exposure is associated with increased midbrain microglial expression. Using a rat model of pre- and post-natal morphine exposure, we assessed cardiorespiratory features of NOWS (resting tachycardia and tachypnea) including the attenuated HVR and HCVR and whether they are associated with increased brainstem microglia expression. Pregnant rats (dams) received twice-daily subcutaneous injections of morphine (5 mg/kg) during the third (last) week of pregnancy to simulate 3rd trimester in utero opioid exposure. Offspring then received once-daily subcutaneous injections of morphine (0.5 mg/kg) until postnatal (P) day P10 days of age to simulate postnatal morphine therapy. Cardiorespiratory responses were assessed 24 h later (P11 days) following spontaneous withdrawal. Compared to saline-treated pups, morphine-exposed offspring exhibited tachycardia and tachypnea as well as an attenuated HVR and HCVR. Microglial cell counts were increased in the nucleus tractus solitarius (nTS), dorsal motor nucleus of the vagus (DMNV) and nucleus ambiguous (NAamb), but not the retrapezoid nucleus (RTN) or the non-cardiorespriatory region, the cuneate nucleus (CN). These data suggest that the cardiorespiratory features and autonomic dysregulation in NOWS infants may be associated with altered microglial function in specific brainstem cardiorespiratory control regions.


Brain Stem , Infant, Newborn, Diseases , Microglia , Opioid-Related Disorders , Substance Withdrawal Syndrome , Tachycardia , Tachypnea , Animals , Animals, Newborn , Brain Stem/immunology , Brain Stem/physiopathology , Disease Models, Animal , Female , Humans , Hypercapnia/immunology , Hypercapnia/physiopathology , Hypoxia/immunology , Hypoxia/physiopathology , Infant, Newborn , Infant, Newborn, Diseases/etiology , Infant, Newborn, Diseases/immunology , Infant, Newborn, Diseases/physiopathology , Microglia/immunology , Opioid-Related Disorders/complications , Opioid-Related Disorders/immunology , Opioid-Related Disorders/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/physiopathology , Rats , Substance Withdrawal Syndrome/complications , Substance Withdrawal Syndrome/immunology , Substance Withdrawal Syndrome/physiopathology , Tachycardia/etiology , Tachycardia/immunology , Tachycardia/physiopathology , Tachypnea/etiology , Tachypnea/immunology , Tachypnea/physiopathology
5.
Nurs Res ; 71(1): 54-65, 2022.
Article En | MEDLINE | ID: mdl-34596065

BACKGROUND: Neonatal abstinence syndrome (NAS) rates have dramatically increased. Breastfeeding is a nonpharmacological intervention that may be beneficial, reducing NAS symptom severity and thus the need for and duration of pharmacological treatment and length of hospital stay. OBJECTIVES: Conduct meta-analysis to determine whether breastfeeding results in better outcomes for NAS infants. Variables included symptom severity, need for and duration of pharmacological treatment, and length of hospital stay. METHODS: PubMed, Scopus, Embase, and Cochrane Library were searched from 2000 to 2020, and comparative studies examining breastfeeding for NAS infants were extracted. Randomized trials and cohort studies were included. Data were extracted and evaluated with Review Manager Version 5.3. A random-effects model was used to pool discontinuous outcomes using risk ratio and 95% confidence intervals. Continuous outcomes were evaluated by mean differences and 95% confidence intervals. RESULTS: Across 11 studies, 6,375 neonates were included in the meta-analysis. Using a random-effects analysis, breastfeeding reduced initiation of pharmacological treatment, reduced duration of pharmacological treatment, and reduced length of stay. No differences were detected for severity of NAS symptoms. Most studies only reported one to two variables of interest. For most studies, these variables were not the primary study outcomes. All studies were found to be of low risk and good quality based on the Cochrane Risk Assessment Tools. Varying breastfeeding definitions limit generalizability. DISCUSSION: Breastfeeding is associated with decreased initiation and duration of pharmacological treatment and length of stay.


Breast Feeding/adverse effects , Neonatal Abstinence Syndrome/complications , Female , Humans , Infant , Infant, Newborn , Male , Neonatal Abstinence Syndrome/physiopathology , Opioid-Related Disorders/complications , Opioid-Related Disorders/physiopathology
6.
Lancet Psychiatry ; 9(1): 84-96, 2022 01.
Article En | MEDLINE | ID: mdl-34774203

Understanding the interface between opioid use disorder (OUD) and post-traumatic stress disorder (PTSD) is challenging. By use of a dimensional framework, such as research domain criteria, convergent and targetable neurobiological processes in OUD-PTSD comorbidity can be identified. We hypothesise that, in OUD-PTSD, circuitry that is implicated in two research domain criteria systems (ie, negative valence and cognitive control) underpins dysregulation of incentive salience, negative emotionality, and executive function. We also propose that the OUD-PTSD state might be systematically investigated with approaches outlined within a neuroclinical assessment framework for addictions and PTSD. Our dimensional analysis of the OUD-PTSD state shows how first-line therapeutic approaches (ie, partial µ-type opioid receptor [MOR1] agonism) modulate overlapping neurobiological and clinical features and also provides mechanistic rationale for evaluating polytherapeutic strategies (ie, partial MOR1 agonism, κ-type opioid receptor [KOR1] antagonism, and α-2A adrenergic receptor [ADRA2A] agonism). A combination of these therapeutic mechanisms is projected to facilitate recovery in patients with OUD-PTSD by mitigating negative valence states and enhancing executive control.


Brain , Cognitive Dysfunction/physiopathology , Executive Function , Nerve Net , Opioid-Related Disorders/physiopathology , Reward , Stress Disorders, Post-Traumatic/physiopathology , Brain/metabolism , Brain/physiopathology , Cognitive Dysfunction/epidemiology , Cognitive Dysfunction/metabolism , Comorbidity , Executive Function/physiology , Humans , Nerve Net/metabolism , Nerve Net/physiopathology , Opioid-Related Disorders/epidemiology , Opioid-Related Disorders/metabolism , Opioid-Related Disorders/therapy , Stress Disorders, Post-Traumatic/epidemiology , Stress Disorders, Post-Traumatic/metabolism , Stress Disorders, Post-Traumatic/therapy
7.
Neuropharmacology ; 205: 108925, 2022 03 01.
Article En | MEDLINE | ID: mdl-34921830

The abuse of oral formulations of prescription opioids has precipitated the current opioid epidemic. We developed an oral oxycodone consumption model consisting of a limited access (4 h) two-bottle choice drinking in the dark (TBC-DID) paradigm and quantified dependence with naloxone challenge using mice of both sexes. We also assessed neurobiological correlates of withdrawal and dependence elicited via oral oxycodone consumption using immunohistochemistry for DeltaFosB (ΔFosB), a transcription factor described as a molecular marker for drug addiction. Neither sex developed a preference for the oxycodone bottle, irrespective of oxycodone concentration, bottle position or prior water restriction. Mice that volitionally consumed oxycodone exhibited hyperlocomotion in an open field test and supraspinal but not spinally-mediated antinociception. Both sexes also developed robust, dose-dependent levels of opioid withdrawal that was precipitated by the opioid antagonist naloxone. Oral oxycodone consumption followed by naloxone challenge led to mesocorticolimbic region-dependent increases in the number of ΔFosB expressing cells. Naloxone-precipitated withdrawal jumps, but not the oxycodone bottle % preference, was positively correlated with the number of ΔFosB expressing cells specifically in the nucleus accumbens shell. Thus, limited access oral consumption of oxycodone produced physical dependence and increased ΔFosB expression despite the absence of opioid preference. Our TBC-DID paradigm allows for the study of oral opioid consumption in a simple, high-throughput manner and elucidates the underlying neurobiological substrates that accompany opioid-induced physical dependence.


Analgesics, Opioid/pharmacology , Nucleus Accumbens/drug effects , Opioid-Related Disorders , Oxycodone/pharmacology , Proto-Oncogene Proteins c-fos/drug effects , Substance Withdrawal Syndrome , Analgesics, Opioid/administration & dosage , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Locomotion/drug effects , Mice , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nociception/drug effects , Nucleus Accumbens/metabolism , Opioid-Related Disorders/metabolism , Opioid-Related Disorders/physiopathology , Oxycodone/administration & dosage , Proto-Oncogene Proteins c-fos/metabolism , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/physiopathology
8.
Int Rev Neurobiol ; 161: 147-165, 2022.
Article En | MEDLINE | ID: mdl-34801168

Opioids are widely prescribed for pain management, and prescription opioid misuse in adolescents has become a major epidemic in the United States and worldwide. Emerging data indicate that adolescence represents a critical period of brain development, and exposure to opioids during adolescence may increase the risk of addiction in adulthood. There is growing evidence that disruptions in brain glial function may be implicated in numerous chronic neuropathologies. Evidence suggests that glial mechanisms have an important role in the development and maintenance of opioid abuse and the risk for addiction. This review will describe glial and neuroimmune mechanisms involved in opioid use disorders during adolescence, which may increase substance use disorder liability later in life. Moreover, this review will identify some important neuro-glial targets, involved in opioid abuse and addiction, to develop future preventions and treatment strategies.


Neuroglia , Neuroimmunomodulation , Opioid-Related Disorders , Adolescent , Adult , Humans , Neuroglia/physiology , Neuroimmunomodulation/physiology , Opioid-Related Disorders/epidemiology , Opioid-Related Disorders/physiopathology , United States/epidemiology
9.
Article En | MEDLINE | ID: mdl-34509531

Opioid withdrawal can be associated to environmental cues through classical conditioning. Exposure to these cues can precipitate a state of conditioned withdrawal in abstinent subjects, and there are suggestions that conditioned withdrawal can perpetuate the addiction cycle in part by promoting the storage of memories. This review discusses evidence supporting the hypothesis that conditioned withdrawal facilitates memory consolidation by activating a neurocircuitry that involves the extended amygdala. Specifically, the central amygdala, the bed nucleus of the stria terminalis, and the nucleus accumbens shell interact functionally during withdrawal, mediate expression of conditioned responses, and are implicated in memory consolidation. From this perspective, the extended amygdala could be a neural pathway by which drug-seeking behaviour performed during a state of conditioned withdrawal is more likely to become habitual and persistent.


Amygdala/physiopathology , Conditioning, Classical/drug effects , Cues , Memory Consolidation/physiology , Substance Withdrawal Syndrome/physiopathology , Animals , Behavior, Addictive/physiopathology , Drug-Seeking Behavior , Humans , Neural Pathways , Nucleus Accumbens/physiopathology , Opioid-Related Disorders/physiopathology , Rats
10.
PLoS One ; 16(9): e0248794, 2021.
Article En | MEDLINE | ID: mdl-34506492

Electroencephalography (EEG) likely reflects activity of cortical neurocircuits, making it an insightful estimation for mental health in patients with substance use disorder (SUD). EEG signals are recorded as sinusoidal waves, containing spectral amplitudes across several frequency bands with high spatio-temporal resolution. Prior work on EEG signal analysis has been made mainly at individual electrodes. These signals can be evaluated from advanced aspects, including sub-regional and hemispheric analyses. Due to limitation of computational techniques, few studies in earlier work could conduct data analyses from these aspects. Therefore, EEG in patients with SUD is not fully understood. In the present retrospective study, spectral powers from a data house containing opioid (OUD), methamphetamine/stimulants (MUD), and alcohol use disorder (AUD) were extracted, and then converted into five distinct topographic data (i.e., electrode-based, cortical subregion-based, left-right hemispheric, anterior-posterior based, and total cortex-based analyses). We found that data conversion and reorganization in the topographic way had an impact on EEG spectral powers in patients with OUD significantly different from those with MUD or AUD. Differential changes were observed from multiple perspectives, including individual electrodes, subregions, hemispheres, anterior-posterior cortices, and across the cortex as a whole. Understanding the differential changes in EEG signals may be useful for future work with machine learning and artificial intelligence (AI), not only for diagnostic but also for prognostic purposes in patients with SUD.


Brain/diagnostic imaging , Electroencephalography/methods , Substance-Related Disorders/diagnostic imaging , Adult , Alcoholism/diagnostic imaging , Alcoholism/physiopathology , Female , Humans , Male , Methamphetamine , Middle Aged , Opioid-Related Disorders/diagnostic imaging , Opioid-Related Disorders/physiopathology , Retrospective Studies , Substance-Related Disorders/physiopathology
11.
Genes Brain Behav ; 20(8): e12770, 2021 11.
Article En | MEDLINE | ID: mdl-34459088

The steady rise in prescription opioids such as oxycodone has led to a virulent epidemic of widespread abuse and deaths in the United States; approximately 80% of affected individuals initiate the habitual use of oxycodone by using prescription oral oxycodone. Given the importance of drug pharmacokinetics in determining abuse potential, we designed an oral operant oxycodone self-administration (SA) procedure in rats to model drug intake by most human users/abusers of oxycodone. Key aspects of the model include limited initial drug intake followed by increasing drug concentrations during extended 4-h sessions on alternating days. Sex and genetic predisposition are major determinants of human opiate abuse. Therefore, we studied females in seven inbred strains (WLI, WMI, LEW, DSS, F344, BN and SHR) and both sexes in three of these strains. All strains increased intake across serially increasing doses (0.025-0.2 mg/ml; p < 0.001): the range of intakes at the final concentration of oxycodone was 0.72 ± 0.17-4.84 ± 1.42 mg/kg (mean ± SEM) - a 6.7-fold difference across strains. In LEW, WLI and WMI strains, oxycodone intake increased significantly across all sessions in both sexes. However, in LEW and WMI male rats but not WLI, daily oxycodone intake was significantly lower across all 4-h sessions than females (p < 0.005). The estimated heritability in oxycodone intake was in the range of 0.21-0.41. In summary, our novel operant oral oxycodone SA model captures the strong abuse potential of oral oxycodone and shows dose, sex and strain-specific drug intake that is significantly dependent on heredity.


Analgesics, Opioid/administration & dosage , Disease Models, Animal , Genetic Predisposition to Disease , Opioid-Related Disorders/genetics , Oxycodone/administration & dosage , Analgesics, Opioid/toxicity , Animals , Female , Male , Opioid-Related Disorders/physiopathology , Oxycodone/toxicity , Rats , Rats, Inbred Dahl , Rats, Inbred F344 , Rats, Inbred Lew , Rats, Wistar , Self Administration , Sex
12.
Eur J Pharmacol ; 909: 174407, 2021 Oct 15.
Article En | MEDLINE | ID: mdl-34363830

Opioid use disorder (OUD) has a variety of adverse effects on both the users and their offspring. In the current study, a random group of Sprague-Dawley rats (25 females and 15 males) were tested for intravenous self-administration of the opioid agonist remifentanil to determine the range of acquisition for opioid. One-month after the end of self-administration of remifentanil, rats with the highest intake were mated together and rats with lowest intake were mated together. Then, the offspring of the two groups were tested for anxiety-like behavior, locomotor activity, nociception and intravenous remifentanil self-administration. The parents showed a range of remifentanil self-administration, especially in the female rats. The offspring of the parents with low and high remifentanil self-administration showed significant differences in specific behavioral functions. On the hotplate test of nociception, the female offspring parents with high remifentanil self-administration had significantly longer hotplate latencies, indicating reduced nociception, than the female offspring of parents with low remifentanil-self-administration, whereas there was no difference in the male offspring of low and high responding parents. In the elevated plus maze test of anxiety-like behavior, the offspring of the parents with high remifentanil intake showed more anxiety-like behavior than the offspring of the parents with low remifentanil intake regardless of sex. Locomotor activity was not significantly different. Interestingly, no significant differences in remifentanil self-administration in the offspring of parents with low and high remifentanil self-administration were detected. Overall, our data suggest a considerable range in remifentanil self-administration in rats and the offspring of rats with high opioid self-administration exhibit different behaviors vs offspring of rats with low opioid self-administration.


Analgesics, Opioid/adverse effects , Behavior, Animal/drug effects , Opioid-Related Disorders/physiopathology , Remifentanil/adverse effects , Analgesics, Opioid/administration & dosage , Animals , Behavior, Animal/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Male , Maternal Exposure/adverse effects , Paternal Exposure/adverse effects , Rats , Remifentanil/administration & dosage , Self Administration
13.
Neuropharmacology ; 198: 108748, 2021 10 15.
Article En | MEDLINE | ID: mdl-34389397

The paraventricular nucleus of the thalamus (PVT) is a midline thalamic brain region that has emerged as a critical circuit node in the regulation of behaviors across domains of affect and motivation, stress responses, and alcohol- and drug-related behaviors. The influence of the PVT in this diverse array of behaviors is a function of its ability to integrate and convey information about salience and valence through its connections with cortical, hypothalamic, hindbrain, and limbic brain regions. While understudied to date, recent studies suggest that several PVT efferents play critical and complex roles in drug and alcohol-related phenotypes. The PVT is also the site of signaling for many neuropeptides released from the synaptic terminals of distal inputs and local neuropeptidergic neurons within. While there is some evidence that neuropeptides including orexin, neurotensin, substance P, and cocaine and amphetamine-related transcript (CART) signal in the PVT to regulate alcohol/drug intake and reinstatement, there remains an overall lack of understanding of the roles of neuropeptides in the PVT in addiction-related behaviors, especially in a circuit-specific context. In this review, we present the current status of preclinical research regarding PVT circuits and neuropeptide modulation of the PVT in three aspects of the addiction cycle: reward/acquisition, withdrawal, and relapse, with a focus on alcohol, opioids (particularly morphine), and psychostimulants (particularly cocaine). Given the PVT's unique position within the broader neural landscape, we further discuss the potential ways in which neuropeptides may regulate these behaviors through their actions upon PVT circuits. This article is part of the special Issue on 'Neurocircuitry Modulating Drug and Alcohol Abuse'.


Alcoholism/physiopathology , Nerve Net/physiopathology , Neuropeptides/metabolism , Paraventricular Hypothalamic Nucleus/physiopathology , Substance-Related Disorders/physiopathology , Animals , Humans , Opioid-Related Disorders/physiopathology , Reward
14.
Gut Microbes ; 13(1): 1946368, 2021.
Article En | MEDLINE | ID: mdl-34313547

Over the past three decades the United States has experienced a devastating opioid epidemic. One of the many debilitating side effects of chronic opioid use is opioid-induced bowel dysfunction. We investigated the impact of methadone maintenance treatment (MMT) on the gut microbiome, the gut bacterial metabolite profile, and intestinal barrier integrity. An imbalance in key bacterial communities required for production of short-chain fatty acids (SCFAs), mucus degradation, and maintenance of barrier integrity was identified. Consistent with dysbiosis, levels of fecal SCFAs were reduced in MMT. We demonstrated that metabolites synthesized by Akkermansia muciniphila modulate intestinal barrier integrity in vitro by strengthening the pore pathway and regulating tight junction protein expression. This study provides essential information about the therapeutic potential of A. muciniphila and warrants development of new clinical strategies that aim to normalize the gut microbiome in individuals affected by chronic opioid use.


Analgesics, Opioid/adverse effects , Dysbiosis/chemically induced , Dysbiosis/physiopathology , Gastrointestinal Microbiome/drug effects , Methadone/therapeutic use , Opioid-Related Disorders/drug therapy , Opioid-Related Disorders/physiopathology , Adult , Analgesics, Opioid/therapeutic use , Animals , Female , Healthy Volunteers , Humans , Male , Middle Aged , Opioid-Related Disorders/epidemiology , United States
15.
Am J Emerg Med ; 49: 104-109, 2021 Nov.
Article En | MEDLINE | ID: mdl-34098328

INTRODUCTION: The opioid epidemic has significantly evolved over the last three decades. The initiation and continuation of prescription opioids for pain control were one of the primary contributors, across different medical settings. The emergency department (ED) is typically the first place patients go to for management of acute pain, and often where opioid naïve patients first become exposed to opioids. In 2018, the ED of University Hospital located in Newark, NJ implemented a pain guideline to ensure that patients are not unnecessarily exposed to opioids. The goal of our study was to determine whether provider adherence was successful in reducing opioid administration. METHODS: We conducted a retrospective review of pharmacy records of patients treated for pain in the ED within the time frame January 1, 2017 and December 31, 2019. We analyzed the change in our practice by comparing the amount of opioid and non-opioid medications administered and the number of patients administered each type, as well as the change in our utilization of specific medications. The t-test or the χ2 test were used as applicable. RESULTS: There were decreases in the mean number of opioid doses administered in 2017 (1273) compared to 2019 (498; p = 0.027). There was an increase in non-opioid analgesics administered, (mean 2017: 1817, mean 2019: 2432.5, p = 0.018). There was also an increase in the proportion of patients given non-opioid analgesics (mean 2017: 22%, mean 2019: 28%, p < 0.0001). There were increases in administrations of acetaminophen (40% to 52%) and ibuprofen (30% to 35.1%), and decreases in administrations of hydromorphone (2.5% to 0.03%), morphine (11.5% to 5.6%), oxycodone (10.6% to 5.3%), and tramadol (5.7% to 1.9%) (all p < 0.0001). DISCUSSION: A guideline that emphasizes the use of non-opioid analgesics first line treatment for acute pain can be effective for reducing opioid administration in the ED. Through the use of our guideline, we reduced the number of patients who have received opioid analgesics and, at the same time, increased non-opioid analgesic administration. Future studies should explore readmission rates, duration of pain relief in patients managed with non-opioid versus opioid analgesics, and perception of relief through the use of satisfaction scores.


Guideline Adherence/standards , Opioid-Related Disorders/drug therapy , Pain Management/standards , Adult , Emergency Service, Hospital/organization & administration , Emergency Service, Hospital/statistics & numerical data , Female , Guideline Adherence/statistics & numerical data , Humans , Male , Middle Aged , Opioid-Related Disorders/physiopathology , Pain/drug therapy , Pain/physiopathology , Pain Management/methods , Pain Management/statistics & numerical data , Retrospective Studies
16.
Biochem Biophys Res Commun ; 558: 8-13, 2021 06 18.
Article En | MEDLINE | ID: mdl-33894675

Increasing evidences suggest the involvement of disrupted circadian clock in various pathologies including stroke and substance abuse. Here we took an attempt to do a comparative study on the regulation of circadian clock gene expression under two pathological circumstances - Opioid addiction and Ischemic stroke in the same cell line model (human neuroblastoma SH-SY5Y cells). To mimic in vivo ischemic stroke condition cells were placed in a hypoxia chamber and incubated for 10 h in balanced salt solution lacking glucose, aerated with an anaerobic gas mixture (95% N2 and 5% C02). For opioid addiction cells were treated with morphine sulphate at 10 µM dose for 48 h. We found that although circadian clock gets disturbed in both states, pattern of alteration of clock gene expressions were different and change was more severe in ischemic stroke than addiction. Interestingly, increase in expression of Cry1 showed as a common factor to both the diseases. This paper also emphasizes the interconnection between the severities of neuronal injury induced by ischemic stroke or opioid abuse to circadian system. Finally, this study will further enrich our knowledge towards the pattern of circadian rhythm disturbances under different pathological states.


Circadian Clocks/genetics , Circadian Clocks/physiology , Cryptochromes/genetics , Cryptochromes/physiology , Ischemic Stroke/genetics , Ischemic Stroke/physiopathology , Opioid-Related Disorders/genetics , Opioid-Related Disorders/physiopathology , Cell Hypoxia , Cell Line , Cell Survival , Glucose/deficiency , Humans , Ischemic Stroke/pathology , Models, Biological , Morphine/administration & dosage , Opioid-Related Disorders/pathology , Up-Regulation/drug effects
17.
J Clin Invest ; 131(10)2021 05 17.
Article En | MEDLINE | ID: mdl-33848264

Opioid use disorder (OUD) has become a leading cause of death in the United States, yet current therapeutic strategies remain highly inadequate. To identify potential treatments for OUD, we screened a targeted selection of over 100 drugs using a recently developed opioid self-administration assay in zebrafish. This paradigm showed that finasteride, a steroidogenesis inhibitor approved for the treatment of benign prostatic hyperplasia and androgenetic alopecia, reduced self-administration of multiple opioids without affecting locomotion or feeding behavior. These findings were confirmed in rats; furthermore, finasteride reduced the physical signs associated with opioid withdrawal. In rat models of neuropathic pain, finasteride did not alter the antinociceptive effect of opioids and reduced withdrawal-induced hyperalgesia. Steroidomic analyses of the brains of fish treated with finasteride revealed a significant increase in dehydroepiandrosterone sulfate (DHEAS). Treatment with precursors of DHEAS reduced opioid self-administration in zebrafish in a fashion akin to the effects of finasteride. These results highlight the importance of steroidogenic pathways as a rich source of therapeutic targets for OUD and point to the potential of finasteride as a new treatment option for this disorder.


5-alpha Reductase Inhibitors/pharmacology , Finasteride/pharmacology , Opioid-Related Disorders/drug therapy , Animals , Disease Models, Animal , Humans , Male , Opioid-Related Disorders/physiopathology , Rats , Rats, Sprague-Dawley , Zebrafish
18.
Anesth Analg ; 132(5): 1244-1253, 2021 05 01.
Article En | MEDLINE | ID: mdl-33857966

Opioids are commonly used for pain management, perioperative procedures, and addiction treatment. There is a current opioid epidemic in North America that is paralleled by a marked increase in related deaths. Since 2000, chronic opioid users have been recognized to have significant central sleep apnea (CSA). After heart failure-related Cheyne-Stokes breathing (CSB), opioid-induced CSA is now the second most commonly seen CSA. It occurs in around 24% of chronic opioid users, typically after opioids have been used for more than 2 months, and usually corresponds in magnitude to opioid dose/plasma concentration. Opioid-induced CSA events often mix with episodes of ataxic breathing. The pathophysiology of opioid-induced CSA is based on dysfunction in respiratory rhythm generation and ventilatory chemoreflexes. Opioids have a paradoxical effect on different brain regions, which result in irregular respiratory rhythm. Regarding ventilatory chemoreflexes, chronic opioid use induces hypoxia that appears to stimulate an augmented hypoxic ventilatory response (high loop gain) and cause a narrow CO2 reserve, a combination that promotes respiratory instability. To date, no direct evidence has shown any major clinical consequence from CSA in chronic opioid users. A line of evidence suggested increased morbidity and mortality in overall chronic opioid users. CSA in chronic opioid users is likely to be a compensatory mechanism to avoid opioid injury and is potentially beneficial. The current treatments of CSA in chronic opioid users mainly focus on continuous positive airway pressure (CPAP) and adaptive servo-ventilation (ASV) or adding oxygen. ASV is more effective in reducing CSA events than CPAP. However, a recent ASV trial suggested an increased all-cause and cardiovascular mortality with the removal of CSA/CSB in cardiac failure patients. A major reason could be counteracting of a compensatory mechanism. No similar trial has been conducted for chronic opioid-related CSA. Future studies should focus on (1) investigating the phenotypes and genotypes of opioid-induced CSA that may have different clinical outcomes; (2) determining if CSA in chronic opioid users is beneficial or detrimental; and (3) assessing clinical consequences on different treatment options on opioid-induced CSA.


Analgesics, Opioid/adverse effects , Brain/drug effects , Lung/innervation , Opioid-Related Disorders/complications , Respiration/drug effects , Sleep Apnea, Central/chemically induced , Brain/physiopathology , Humans , Opioid-Related Disorders/mortality , Opioid-Related Disorders/physiopathology , Prognosis , Respiratory Center/drug effects , Respiratory Center/physiopathology , Risk Assessment , Risk Factors , Sleep Apnea, Central/mortality , Sleep Apnea, Central/physiopathology
20.
Am J Drug Alcohol Abuse ; 47(3): 319-329, 2021 05 04.
Article En | MEDLINE | ID: mdl-33735587

Background: Negative emotion is associated with substance craving and use in individuals recovering from substance use disorders, including prescription opioid use disorder (POUD). Decisions to abandon or persist towards a goal after negative emotion-eliciting events, and neural responses that shape such decisions, may be important in maintaining recovery from POUD.Objectives: We examined differences in neural responses to negative events and subsequent persistence decisions in individuals recovering from POUD without a history of a substance use disorder. Methods: 20 individuals with POUD (POUD group: 4 females, abstinent 2-3 weeks after admission to an inpatient treatment facility post-detoxification, no other substance use disorder), and 20 individuals with no substance use history (control group: 6 females) completed a persistence-after-setbacks task during functional magnetic resonance imaging. Participants advanced along a path toward a reward; after encountering each negative event (i.e., progress-erasing setback), participants made decisions to persist or abandon the path. Persistence decision rates were compared between groups and blood-oxygen-level-dependent signal to negative events was analyzed within a striatum region of interest (ROI) as well as whole-brain.Results: The POUD group persisted less (t(38) = 2.293, p = .028, d = .725) and showed lower striatum (left ventral putamen) signal to negative events compared to the control group (p < .05, corrected for striatum ROI).Conclusions: In POUD, neural and behavioral responses to negative events differ from controls. These differences are a target for research to address whether POUD treatment increases persistence and striatum responses to negative events and improves recovery outcomes.


Affect/physiology , Opioid-Related Disorders/physiopathology , Adult , Brain/physiopathology , Brain Mapping , Case-Control Studies , Craving , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , New Jersey , Young Adult
...