Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Int J Legal Med ; 137(4): 1039-1049, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36786894

ABSTRACT

The most common pulmonary findings in opiate-related fatalities are congestion and oedema, as well as acute and/or chronic alveolar haemorrhage, the cause of which is thought to be a damage to the capillary endothelium related to ischemia. Human vascular mesenchymal stromal cells (vMSCs) play a fundamental role in tissue regeneration and repair after endothelial cell injury, and they express opioid receptors. The aim of this study was to assess the effect of in vitro morphine exposure on the physiological activity and maintenance of human vMSCs. vMSCs were obtained from abdominal aorta fragments collected during surgery repair and were exposed to incremental doses (0.1 mM, 0.4 mM, 0.8 mM and 1 mM) of morphine sulphate for 7 days. The effect was investigated through cell viability assessment, proliferation assay, reactive oxygen species (ROS) detection assay, senescence-associated ß-galactosidase assay, senescent-related markers (p21WAF1/CIP1 and p16INK4) and the apoptosis-related marker caspase 3. Moreover, an ultrastructural analysis by transmission electron microscopy and in vitro vascular differentiation were evaluated. Results showed a decrease of the cellular metabolic activity, a pro-oxidant and pro-senescence effect, an increase in intracellular ROS and the activation of the apoptosis signalling, as well as ultrastructural modifications and impairment of vascular differentiation after morphine treatment of vMSC. Although confirmation studies are required on real fatal opiate intoxications, the approach based on morphological and immunofluorescence methodologies may have a high potential also as a useful tool or as a complementary method in forensic pathology. The application of these techniques in the future may lead to the identification of new markers and morphological parameters useful as complementary investigations for drug-related deaths.


Subject(s)
Mesenchymal Stem Cells , Opiate Alkaloids , Humans , Cellular Senescence/physiology , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/pharmacology , Opiate Alkaloids/pharmacology , Mesenchymal Stem Cells/metabolism , Morphine Derivatives/pharmacology
2.
Surg Endosc ; 36(8): 6129-6137, 2022 08.
Article in English | MEDLINE | ID: mdl-35043232

ABSTRACT

BACKGROUND: Post-operative ileus and delayed return of gastrointestinal function are complications seen frequently in patients undergoing colorectal surgery. Many enhanced recovery after surgery protocols include alvimopan to inhibit the effects of opiates in the gastrointestinal tract and lidocaine to augment analgesics. Limited data exist regarding alvimopan's efficacy in opiate-sparing regimens. METHODS: This single-center, retrospective cohort analysis was conducted in a randomly selected population of adult patients undergoing colorectal resection between February 2018 and October 2019. Patients meeting inclusion criteria were divided into four groups dependent upon whether or not they received alvimopan (A or a) and/or lidocaine (L or l). The primary endpoint in this study was median time to first bowel movement or discharge, whichever came first. Our secondary endpoint was length of stay. RESULTS: Of the 430 patients evaluated, a total of 192 patients were included in the final evaluation in the following groups: AL (n = 93), Al (n = 34), aL (n = 44), and al (n = 21). A significant difference was found among the groups for the primary outcome of median time to bowel movement or discharge (p = 0.001). Three subsequent pair-wise comparisons resulted in a significant difference in the primary outcome: group AL 39.4 h vs. group aL 54.0 h (p = 0.003), group AL 39.4 h vs. group al 55.4 h (p = 0.001), and group Al 44.9 h vs. group al 55.4 h (p = 0.01). Length of stay was significantly reduced by 1.8 days in groups AL and Al compared to group aL (p < 0.001). CONCLUSION: Treatment with alvimopan resulted in a significant improvement in time to GI recovery and decreased length of stay in an established ERAS program. While lidocaine's reduction in opiates was minimal, the group receiving both alvimopan and lidocaine had the greatest reduction in time to GI recovery and length of stay.


Subject(s)
Colorectal Surgery , Ileus , Opiate Alkaloids , Adult , Gastrointestinal Agents/therapeutic use , Humans , Ileus/etiology , Ileus/prevention & control , Length of Stay , Lidocaine/pharmacology , Lidocaine/therapeutic use , Opiate Alkaloids/pharmacology , Piperidines , Postoperative Complications/chemically induced , Postoperative Complications/prevention & control , Recovery of Function , Retrospective Studies
3.
Article in English | MEDLINE | ID: mdl-34324921

ABSTRACT

The deleterious effects of the drug addiction epidemic are compounded by treatment strategies that are only marginally efficacious. Memantine is a unique glutamatergic medication with proven ability to attenuate drug addiction in preclinical models. However, clinical translational studies are inconsistent. In this review, we summarize preclinical evidences and clinical trials that investigated the efficacy of memantine in treating patients with alcohol, opiate, cocaine, and nicotine use disorders and discuss the results from a mechanistic point of view. Memantine has shown efficacy in reducing alcohol and opiate craving, consumption, and withdrawal severity. However, in cocaine and nicotine use disorders, memantine did not have significant effect on cravings or consumption. Additionally, memantine was associated with increased subjective effects of alcohol, cocaine, and nicotine. We discuss possible mechanisms behind this variability. Since memantine transiently blocks NMDA receptors and protects neurons from overstimulation by excessive synaptic glutamate, its efficacy should be observed in drug phases that cause hyperglutamatergic states, while hypoglutamatergic drug use states would not resolve with blocking NMDA receptors. Second, memantine pharmacokinetic studies have been done in rodents and healthy volunteers, but not in patients with substance use disorder. Memantine, opiates, cocaine, and nicotine share the same transporter family at the blood brain barrier. This shared transport mechanism could impact brain concentrations of memantine and its effects. In conclusion, memantine remains an intriguing compound in our pharmacopeia with controversial results in treating certain aspects of drug addiction. Further studies are needed to understand the clinical and biological correlates of its efficacy.


Subject(s)
Brain/drug effects , Excitatory Amino Acid Antagonists , Memantine , Substance-Related Disorders/drug therapy , Animals , Brain/metabolism , Cocaine/pharmacology , Ethanol/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Antagonists/therapeutic use , Glutamic Acid/metabolism , Humans , Memantine/pharmacology , Memantine/therapeutic use , Neurons/drug effects , Opiate Alkaloids/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism
4.
Platelets ; 32(4): 555-559, 2021 May 19.
Article in English | MEDLINE | ID: mdl-32543247

ABSTRACT

A novel enoxaparin regimen consisting of intra-arterial bolus (0.75 mg/kg) followed by intravenous infusion (0.75 mg/kg/6 hours) has been developed as a possible solution to the delayed absorption of oral P2Y12 inhibitors in opiate-treated ST-elevation myocardial infarction (STEMI) patients undergoing primary angioplasty. We aimed to study the feasibility of this regimen as an alternative to standard-of-care treatment (SOC) with unfractionated heparin ± glycoprotein IIb/IIIa antagonist (GPI). One hundred opiate-treated patients presenting with STEMI and accepted for primary angioplasty were randomized (1:1) to either enoxaparin or SOC. Fifty patients were allocated enoxaparin (median age 61, 40% females) and 49 allocated SOC (median age 62, 22% females). One developed stroke before angiography and was withdrawn. One SOC patient had a gastrointestinal bleed resulting in 1 g drop in hemoglobin and early cessation of GPI infusion. Two enoxaparin patients had transient minor bleeding: one transient gingival bleed and one episode of coffee ground vomit with no hemoglobin drop or hemodynamic instability. Two SOC and no enoxaparin group patients had acute stent thrombosis. These preliminary data support further study of this novel 6-hour enoxaparin regimen in opiate-treated PPCI patients.


Subject(s)
Enoxaparin/therapeutic use , Fibrinolytic Agents/therapeutic use , Opiate Alkaloids/therapeutic use , Percutaneous Coronary Intervention/methods , Enoxaparin/pharmacology , Feasibility Studies , Female , Fibrinolytic Agents/pharmacology , Humans , Male , Opiate Alkaloids/pharmacology
5.
Adicciones (Palma de Mallorca) ; 33(3): 235-244, 2021. graf, tab
Article in English, Spanish | IBECS | ID: ibc-208069

ABSTRACT

El consumo de analgésicos opiáceos ha provocado una situación deemergencia sanitaria y social en Estados Unidos. En España, segúndatos oficiales, la prescripción de estos fármacos ha experimentadoun espectacular ascenso en la última década. Este estudio explora laprevalencia del uso de estos fármacos y las características sanitarias ysociodemográficas de sus consumidores en la ciudad de Madrid. Serealizó una encuesta telefónica aplicando un muestreo estratificadoy aleatorizado, en la que se preguntó por el uso de estos fármacos ysi fueron médicamente prescritos o no. La muestra estuvo compuestapor n= 8.845 sujetos de edades entre 15 y 98 años. Un 16,0% declara haber usado estos fármacos en el último año y un 9,1 los tomaen las dos últimas semanas. El consumo es más frecuente en mujeres, clase social baja y menor nivel de estudios. El grupo más joven(15-29 años) ya lo usa en el 12,5%. Quienes usan opioides refierenpeor salud percibida, menor calidad de vida, más problemas de saludmental, más soledad no deseada, más uso de otros psicofármacos, másfrecuente uso diario de tabaco y menos consumo problemático dealcohol. Un 10% de quienes los usan lo hacen sin prescripción médica. Combinando estos datos con los de prescripción ofrecidos por elMinisterio de Sanidad, resulta necesario prestar atención a un problema que puede hacerse patente en los próximos años, aconsejando laadopción de medidas urgentes para atajarlo antes de que aproxime lasituación española a la ya bien conocida en otros países. (AU)


The use of opiate analgesics has led to a health and social emergencyin the United States. In Spain, according to official data, the prescription of these drugs has risen dramatically in the last decade. This studyexplores the prevalence of the use of these drugs and the health andsocio-demographic characteristics of their consumers in the city ofMadrid. A telephone survey was carried on a stratified, randomisedsample, asking about the use of these drugs and whether or not theywere medically prescribed. The sample consisted of n=8,845 subjectsaged between 15 and 98 years. Sixteen percent stated that they hadused these drugs in the last year and 9.1% had taken them in the lasttwo weeks. Consumption was more frequent among women, lower social class and lower level of education. Among the youngest group (15-29 years old) 12.5% had already used it. Those who use opioids reportworse perceived health, lower quality of life, more mental health problems, more loneliness, more use of other psychoactive drugs, morefrequent daily use of tobacco and less problematic consumption ofalcohol. Ten percent of those who use them do so without a doctor’sprescription. Combining these data with the prescription data offeredby the Ministry of Health, it is necessary to pay attention to a problem that may become apparent in the coming years, and the adoptionof urgent measures to tackle it before it brings the Spanish situationcloser to that already well known in countries of our socio-politicalenvironment is advised (AU)


Subject(s)
Humans , Opiate Alkaloids/administration & dosage , Opiate Alkaloids/pharmacology , Opioid-Related Disorders/prevention & control , Surveys and Questionnaires , Spain , Mental Health , Psychopharmacology/trends
7.
J Psychiatr Pract ; 25(4): 290-297, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31291209

ABSTRACT

This column is the sixth in a series exploring drug-drug interactions (DDIs) with a special emphasis on psychiatric medications. The first 3 columns in this DDI series discussed why patients being treated with psychiatric medications are at increased risk for taking multiple medications and thus experiencing DDIs, how to recognize such DDIs, strategies for avoiding and/or minimizing adverse outcomes from such DDIs, and pharmacokinetic considerations concerning DDIs in psychiatric practice. The fourth and fifth columns in this series presented a pair of parallel tables, one of which outlined the primary, known mechanism(s) of action of all commonly used psychiatric medications and one of which summarized major types of pharmacodynamic DDIs based on mechanism of action. Clinicians can use these 2 tables together to predict pharmacodynamically mediated DDIs. This sixth column in the series discusses some key issues related to pharmacodynamic interactions involving commonly used psychiatric medications. The column first discusses 3 types of pharmacological agents that deserve special mention because of the widespread types of pharmacodynamic DDIs they can have with psychiatric and other medications: ethanol, opioids, and monoamine oxidase inhibitors, with a special focus on hypertensive crises and serotonin syndrome with monoamine oxidase inhibitors. The column also discusses DDIs in terms of effects on the cardiovascular system, including QTc prolongation, blood pressure and heart rate regulation, increased risk of bleeding and abnormal bleeding, and valvular heart disease, and on the central nervous system, including increased sedation, respiratory depression, body temperature regulation, and tardive dyskinesia. The overall goal of this series of columns is to present a simple way of conceptualizing neuropsychiatric medications in terms of their pharmacodynamics and pharmacokinetics to allow prescribers to take these facts into consideration when they need to use more than 1 drug in combination to optimally treat a patient.


Subject(s)
Mental Disorders/drug therapy , Psychotropic Drugs/pharmacology , Drug Interactions , Ethanol/adverse effects , Ethanol/pharmacology , Humans , Hypertension/chemically induced , Long QT Syndrome/chemically induced , Monoamine Oxidase Inhibitors/adverse effects , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/therapeutic use , Opiate Alkaloids/adverse effects , Opiate Alkaloids/pharmacology , Opiate Alkaloids/therapeutic use , Psychotropic Drugs/therapeutic use , Serotonin Syndrome/chemically induced , Tardive Dyskinesia/chemically induced
9.
J Neuroinflammation ; 15(1): 285, 2018 Oct 10.
Article in English | MEDLINE | ID: mdl-30305110

ABSTRACT

BACKGROUND: The collective cognitive and motor deficits known as HIV-associated neurocognitive disorders (HAND) remain high even among HIV+ individuals whose antiretroviral therapy is optimized. HAND is worsened in the context of opiate abuse. The mechanism of exacerbation remains unclear but likely involves chronic immune activation of glial cells resulting from persistent, low-level exposure to the virus and viral proteins. We tested whether signaling through C-C chemokine receptor type 5 (CCR5) contributes to neurotoxic interactions between HIV-1 transactivator of transcription (Tat) and opiates and explored potential mechanisms. METHODS: Neuronal survival was tracked in neuronal and glial co-cultures over 72 h of treatment with HIV-1 Tat ± morphine using cells from CCR5-deficient and wild-type mice exposed to the CCR5 antagonist maraviroc or exogenously-added BDNF (analyzed by repeated measures ANOVA). Intracellular calcium changes in response to Tat ± morphine ± maraviroc were assessed by ratiometric Fura-2 imaging (analyzed by repeated measures ANOVA). Release of brain-derived neurotrophic factor (BDNF) and its precursor proBDNF from CCR5-deficient and wild-type glia was measured by ELISA (analyzed by two-way ANOVA). Levels of CCR5 and µ-opioid receptor (MOR) were measured by immunoblotting (analyzed by Student's t test). RESULTS: HIV-1 Tat induces neurotoxicity, which is greatly exacerbated by morphine in wild-type cultures expressing CCR5. Loss of CCR5 from glia (but not neurons) eliminated neurotoxicity due to Tat and morphine interactions. Unexpectedly, when CCR5 was lost from glia, morphine appeared to entirely protect neurons from Tat-induced toxicity. Maraviroc pre-treatment similarly eliminated neurotoxicity and attenuated neuronal increases in [Ca2+]i caused by Tat ± morphine. proBDNF/BDNF ratios were increased in conditioned media from Tat ± morphine-treated wild-type glia compared to CCR5-deficient glia. Exogenous BDNF treatments mimicked the pro-survival effect of glial CCR5 deficiency against Tat ± morphine. CONCLUSIONS: Our results suggest a critical role for glial CCR5 in mediating neurotoxic effects of HIV-1 Tat and morphine interactions on neurons. A shift in the proBDNF/BDNF ratio that favors neurotrophic support may occur when glial CCR5 signaling is blocked. Some neuroprotection occurred only in the presence of morphine, suggesting that loss of CCR5 may fundamentally change signaling through the MOR in glia.


Subject(s)
Analgesics, Opioid/pharmacology , Gene Expression Regulation/genetics , Neuroglia/metabolism , Opiate Alkaloids/pharmacology , Receptors, CCR5/deficiency , tat Gene Products, Human Immunodeficiency Virus/metabolism , AIDS Dementia Complex , Animals , CCR5 Receptor Antagonists/pharmacology , Corpus Striatum/cytology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Maraviroc/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Neuroglia/drug effects , Neurons/physiology , Opiate Alkaloids/metabolism , Receptors, CCR5/genetics , Receptors, Opioid, mu/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics
10.
Int Rev Neurobiol ; 140: 249-270, 2018.
Article in English | MEDLINE | ID: mdl-30193706

ABSTRACT

Many factors, including social elements, influence drug addiction in humans and can be modeled in laboratory rodents. In general, the presence of social reward is protective against drug abuse and the absence or removal of social reward in both humans and rodents increases vulnerability to drug addiction. The current review chapter is focused on studies from our lab that have examined the effects of sociosexual behavior in male rats on drug-induced behaviors, including changes in both psychostimulant and opiate behavior. Furthermore, we review the underlying neural mechanisms by which these effects occur. Together, these results may help elucidate the neural mechanisms underlying the interaction between social and drug rewards and the mechanisms by which a loss of social rewards increase the vulnerability to drug addiction development.


Subject(s)
Behavior, Addictive/physiopathology , Behavior, Animal/physiology , Central Nervous System Stimulants/pharmacology , Dopaminergic Neurons/metabolism , Nucleus Accumbens/metabolism , Opiate Alkaloids/pharmacology , Reinforcement, Psychology , Sexual Behavior, Animal/physiology , Signal Transduction/physiology , Social Behavior , Ventral Tegmental Area/metabolism , Animals , Behavior, Animal/drug effects , Dopaminergic Neurons/cytology , Dopaminergic Neurons/drug effects , Male , Nucleus Accumbens/cytology , Nucleus Accumbens/drug effects , Rats , Signal Transduction/drug effects , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects
11.
J Cell Biochem ; 119(11): 8684-8693, 2018 11.
Article in English | MEDLINE | ID: mdl-30076655

ABSTRACT

Pain is a common experience for most patients in the intensive care unit (ICU). In the current study, the advantages and disadvantages of analgesic and sedative drugs used in the ICU are reviewed. An ideal sedative and analgesic agent should have features such as rapid onset of action, rapid recovery after discontinuation, predictability, minimal accumulation of the agent and metabolites in the body, and lack of toxicity. None of the sedative and analgesic agents have all of these desired characteristics; nevertheless, clinicians must be familiar with these classes of drugs to optimize pharmacotherapy and ensure as few side-effects as possible for ICU patients.


Subject(s)
Analgesics, Opioid/pharmacology , Hypnotics and Sedatives/pharmacology , Intensive Care Units , Opiate Alkaloids/pharmacology , Pain Management , Pain/drug therapy , Analgesics, Opioid/adverse effects , Analgesics, Opioid/classification , Anxiety/drug therapy , Humans , Hypnotics and Sedatives/adverse effects , Opiate Alkaloids/adverse effects , Opiate Alkaloids/classification , Pain/physiopathology , Pain/psychology , Treatment Outcome
12.
Curr Pharm Des ; 24(20): 2250-2254, 2018.
Article in English | MEDLINE | ID: mdl-30039753

ABSTRACT

Chronic use of opiates for control of chronic pain is complicated by the development of tolerance and hyperalgesia, and hence usually entails dose escalation and diminished efficacy. Our evolving understanding of the mechanisms mediating induction of morphine tolerance may enable discovery of adjunct measures which can prevent this tolerance; this essay proposes that certain nutraceuticals may have utility in this regard. Considerable evidence now points to an obligate role for production of peroxynitrite and other oxidants in the dorsal horn in development of morphine tolerance. Various isoforms of NADPH oxidase are the chief source of the superoxide which gives rise to these oxidants. Since heme oxygenase, via its products bilirubin and carbon monoxide, functions as a physiological inhibitor of various isoforms of NADPH oxidase, phase 2-inducing nutraceuticals with blood brain-barrier permeability such as lipoic acid, an effective inducer of heme oxygenase-1, may have potential for prevention of morphine tolerance; indeed, this has been demonstrated in a mouse study. The phycocyanobilin (PhyCB) chromophore of spirulina, a structural analog of biliverdin, shares bilirubin's ability to inhibit NAPDH oxidase complexes; hence, administration of spirulina or of PhyCB-enriched spirulina extracts merits evaluation in rodent models of morphine tolerance. Uric acid quenches peroxynitrite-derived radicals, and its plasma level can be boosting via supplementation with inosine; indeed, administration of inosine has been shown to counteract development of hyperalgesia in rodents. If practical doses of these agents can be shown to prevent morphine tolerance and hyperalgesia in rodents, their use as adjuvants to clinical opiate therapy should be assessed.


Subject(s)
Dietary Supplements , Drug Tolerance , Opiate Alkaloids/pharmacology , Phycobilins/pharmacology , Phycocyanin/pharmacology , Humans , Opiate Alkaloids/adverse effects , Oxidants/metabolism , Phycobilins/administration & dosage , Phycocyanin/administration & dosage
13.
Sci Transl Med ; 10(447)2018 06 27.
Article in English | MEDLINE | ID: mdl-29950444

ABSTRACT

The changes in brain function that perpetuate opiate addiction are unclear. In our studies of human narcolepsy, a disease caused by loss of immunohistochemically detected hypocretin (orexin) neurons, we encountered a control brain (from an apparently neurologically normal individual) with 50% more hypocretin neurons than other control human brains that we had studied. We discovered that this individual was a heroin addict. Studying five postmortem brains from heroin addicts, we report that the brain tissue had, on average, 54% more immunohistochemically detected neurons producing hypocretin than did control brains from neurologically normal subjects. Similar increases in hypocretin-producing cells could be induced in wild-type mice by long-term (but not short-term) administration of morphine. The increased number of detected hypocretin neurons was not due to neurogenesis and outlasted morphine administration by several weeks. The number of neurons containing melanin-concentrating hormone, which are in the same hypothalamic region as hypocretin-producing cells, did not change in response to morphine administration. Morphine administration restored the population of detected hypocretin cells to normal numbers in transgenic mice in which these neurons had been partially depleted. Morphine administration also decreased cataplexy in mice made narcoleptic by the depletion of hypocretin neurons. These findings suggest that opiate agonists may have a role in the treatment of narcolepsy, a disorder caused by hypocretin neuron loss, and that increased numbers of hypocretin-producing cells may play a role in maintaining opiate addiction.


Subject(s)
Brain/metabolism , Cataplexy/drug therapy , Narcolepsy/drug therapy , Opiate Alkaloids/therapeutic use , Orexins/biosynthesis , Animals , Brain/pathology , Cataplexy/complications , Cell Count , Disease Models, Animal , Dose-Response Relationship, Drug , Heroin , Humans , Male , Mice, Inbred C57BL , Morphine/administration & dosage , Morphine/pharmacology , Morphine/therapeutic use , Narcolepsy/complications , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Opiate Alkaloids/pharmacology , Rats, Sprague-Dawley , Substance-Related Disorders/metabolism , Substance-Related Disorders/pathology
14.
Clin Chem ; 64(8): 1221-1229, 2018 08.
Article in English | MEDLINE | ID: mdl-29776952

ABSTRACT

BACKGROUND: Detection of new highly potent synthetic opioids is challenging as new compounds enter the market. Here we present a novel screening method for the detection of opiates and (synthetic) opioids based on their activity. METHODS: A cell-based system was set up in which activation of the µ-opioid receptor (MOR) led to recruitment of ß-arrestin 2, resulting in functional complementation of a split NanoLuc luciferase and allowing readout via bioluminescence. Assay performance was evaluated on 107 postmortem blood samples. Blood (500 µL) was extracted via solid-phase extraction. Following evaporation and reconstitution in 100 µL of Opti-MEM® I, 20 µL was analyzed in the bioassay. RESULTS: In 8 samples containing synthetic opioids, in which no positive signal was obtained in the bioassay, quadrupole time-of-flight mass spectrometry revealed the MOR antagonist naloxone, which can prevent receptor activation. Hence, further evaluation did not include these samples. For U-47700 (74.5-547 ng/mL) and furanyl fentanyl (<1-38.8 ng/mL), detection was 100% (8/8) for U-47700 and 95% (21/22) for furanyl fentanyl. An analytical specificity of 93% (55/59) was obtained for the opioid negatives. From an additional 10 samples found to contain other opioids, 5 were correctly scored positive. Nondetection in 5 cases could be explained by very low concentrations (<1 ng/mL alfentanil/sufentanil) or presence of inactive enantiomers. CONCLUSIONS: The MOR reporter assay allows rapid identification of opioid activity in blood. Although the cooccurrence of opioid antagonists is currently a limitation, the bioassay's high detection capability, specificity, and untargeted nature may render it a useful first-line screening tool to investigate potential opioid intoxications.


Subject(s)
Analgesics, Opioid/analysis , Opiate Alkaloids/analysis , Analgesics, Opioid/blood , Analgesics, Opioid/pharmacology , Biological Assay , HEK293 Cells , Humans , Limit of Detection , Opiate Alkaloids/blood , Opiate Alkaloids/pharmacology , Receptors, G-Protein-Coupled/metabolism , Receptors, Opioid, mu/drug effects
15.
ACS Chem Neurosci ; 8(9): 1830-1838, 2017 09 20.
Article in English | MEDLINE | ID: mdl-28768409

ABSTRACT

Opioids are among the most effective pain relievers; however, their abuse has been on the rise worldwide evident from an alarming increase in accidental opioid overdoses. This demands for an urgent increase in scientific endeavors for better understanding of main cellular mechanisms and circuits involved in opiate addiction. Preclinical studies strongly suggest that memories associated with positive and negative opioid experiences are critical in promoting compulsive opiate-seeking and opiate-taking behaviors, and relapse. Particular focus on synaptic plasticity as the cellular correlate of learning and memory has rapidly evolved in drug addiction field over the past two decades. Several critical addiction-related brain areas are identified, one of which is the ventral tegmental area (VTA), an area intensively studied as the initial locus for drug reward. Here, we provide an update to our previous review on "Opiates and Plasticity" highlighting the most recent discoveries of synaptic plasticity associated with opiates in the VTA. Electrophysiological studies of plasticity of addiction to date have been invaluable in addressing learning processes and mechanisms that underlie motivated and addictive behaviors, and now with the availability of powerful technologies of transgenic approaches and optogenetics, circuit-based studies hold high promise in fostering synaptic studies of opiate addiction.


Subject(s)
Analgesics, Opioid/pharmacology , Neuronal Plasticity/drug effects , Opiate Alkaloids/pharmacology , Opioid-Related Disorders/metabolism , Ventral Tegmental Area/drug effects , Analgesics, Opioid/adverse effects , Animals , Epigenesis, Genetic , Humans , Neuronal Plasticity/physiology , Opiate Alkaloids/adverse effects , Synapses/drug effects , Synapses/metabolism , Ventral Tegmental Area/metabolism
16.
J Pharmacol Exp Ther ; 361(2): 280-291, 2017 05.
Article in English | MEDLINE | ID: mdl-28336575

ABSTRACT

Opioid-induced constipation (OIC) is a common side effect of opioid pharmacotherapy for the management of pain because opioid agonists bind to µ-opioid receptors in the enteric nervous system (ENS). Naloxegol, a polyethylene glycol derivative of naloxol, which is a derivative of naloxone and a peripherally acting µ-opioid receptor antagonist, targets the physiologic mechanisms that cause OIC. Pharmacologic measures of opioid activity and pharmacokinetic measures of central nervous system (CNS) penetration were employed to characterize the mechanism of action of naloxegol. At the human µ-opioid receptor in vitro, naloxegol was a potent inhibitor of binding (Ki = 7.42 nM) and a neutral competitive antagonist (pA2 - 7.95); agonist effects were <10% up to 30 µM and identical to those of naloxone. The oral doses achieving 50% of the maximal effect in the rat for antagonism of morphine-induced inhibition of gastrointestinal transit and morphine-induced antinociception in the hot plate assay were 23.1 and 55.4 mg/kg for naloxegol and 0.69 and 1.14 mg/kg by for naloxone, respectively. In the human colon adenocarcinoma cell transport assay, naloxegol was a substrate for the P-glycoprotein transporter, with low apparent permeability in the apical to basolateral direction, and penetrated the CNS 15-fold slower than naloxone in a rat brain perfusion model. Naloxegol-derived radioactivity was poorly distributed throughout the rat CNS and was eliminated from most tissues within 24 hours. These findings corroborate phase 3 clinical studies demonstrating that naloxegol relieves OIC-associated symptoms in patients with chronic noncancer pain by antagonizing the µ-opioid receptor in the ENS while preserving CNS-mediated analgesia.


Subject(s)
Constipation/drug therapy , Morphinans/pharmacology , Opiate Alkaloids/toxicity , Polyethylene Glycols/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , Analgesics, Opioid/pharmacology , Analgesics, Opioid/toxicity , Animals , Brain/metabolism , Caco-2 Cells , Cell Membrane Permeability , Constipation/chemically induced , Female , Gastrointestinal Transit/drug effects , HEK293 Cells , Humans , Male , Morphinans/pharmacokinetics , Morphine/pharmacology , Morphine/toxicity , Opiate Alkaloids/pharmacology , Polyethylene Glycols/pharmacokinetics , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/agonists , Tissue Distribution
17.
Expert Rev Clin Pharmacol ; 10(2): 167-177, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27835034

ABSTRACT

INTRODUCTION: A wide range of medications have been studied for posttraumatic stress disorder (PTSD) and a number are registered for this indication. Nevertheless, current pharmacotherapies are only partially effective in some patients, and are minimally effective in others. Thus novel treatment avenues need to be explored. Areas covered: In considering novel pharmacological agents for the treatment of PTSD, this paper takes a translational approach. We outline how advances in our understanding of the underlying neurobiology of PTSD may inform the identification of potential new treatment targets, including glutamatergic, noradrenergic and opioid pathways. Expert commentary: Continued investigation of the neural substrates and signalling pathways involved in responses to trauma may inform the development of novel treatment targets for future drug development for PTSD. However, the translation of preclinical findings to clinical practice is likely to be complex and gradual.


Subject(s)
Excitatory Amino Acid Agents/therapeutic use , Opiate Alkaloids/therapeutic use , Psychotropic Drugs/therapeutic use , Stress Disorders, Post-Traumatic/therapy , Sympathomimetics/therapeutic use , Excitatory Amino Acid Agents/pharmacology , Humans , Opiate Alkaloids/pharmacology , Psychotherapy , Sympathomimetics/pharmacology
19.
Med Sci (Paris) ; 32(6-7): 619-24, 2016.
Article in French | MEDLINE | ID: mdl-27406773

ABSTRACT

Since the work of Johnson and North, it is known that opiates increase the activity of dopaminergic neurons by a GABA neuron-mediated desinhibition. This model should however be updated based on recent advances. Thus, the neuroanatomical location of the GABA neurons responsible for this desinhibition has been recently detailed: they belong to a brain structure in continuity with the posterior part of the ventral tegmental area and discovered this past decade. Other data also highlighted the critical role played by glutamatergic transmission in the opioid regulation of dopaminergic neuron activity. During protracted opiate withdrawal, the inhibitory/excitatory balance exerted on dopaminergic neurons is altered. These results are now leading to propose an original hypothesis for explaining the impact of protracted opiate withdrawal on mood.


Subject(s)
Dopaminergic Neurons/drug effects , Opiate Alkaloids/pharmacology , Animals , Brain/drug effects , Brain/physiology , Dopaminergic Neurons/physiology , GABAergic Neurons/drug effects , GABAergic Neurons/physiology , Humans , Synaptic Transmission/drug effects , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/physiology
20.
J Neurosci ; 36(20): 5498-508, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27194330

ABSTRACT

UNLABELLED: The ability to detect noxious stimuli, process the nociceptive signal, and elicit an appropriate behavioral response is essential for survival. In Caenorhabditis elegans, opioid receptor agonists, such as morphine, mimic serotonin, and suppress the overall withdrawal from noxious stimuli through a pathway requiring the opioid-like receptor, NPR-17. This serotonin- or morphine-dependent modulation can be rescued in npr-17-null animals by the expression of npr-17 or a human κ opioid receptor in the two ASI sensory neurons, with ASI opioid signaling selectively inhibiting ASI neuropeptide release. Serotonergic modulation requires peptides encoded by both nlp-3 and nlp-24, and either nlp-3 or nlp-24 overexpression mimics morphine and suppresses withdrawal. Peptides encoded by nlp-3 act differentially, with only NLP-3.3 mimicking morphine, whereas other nlp-3 peptides antagonize NLP-3.3 modulation. Together, these results demonstrate that opiates modulate nociception in Caenorhabditis elegans through a complex monoaminergic/peptidergic cascade, and suggest that this model may be useful for dissecting opiate signaling in mammals. SIGNIFICANCE STATEMENT: Opiates are used extensively to treat chronic pain. In Caenorhabditis elegans, opioid receptor agonists suppress the overall withdrawal from noxious chemical stimuli through a pathway requiring an opioid-like receptor and two distinct neuropeptide-encoding genes, with individual peptides from the same gene functioning antagonistically to modulate nociception. Endogenous opioid signaling functions as part of a complex, monoaminergic/peptidergic signaling cascade and appears to selectively inhibit neuropeptide release, mediated by a α-adrenergic-like receptor, from two sensory neurons. Importantly, receptor null animals can be rescued by the expression of the human κ opioid receptor, and injection of human opioid receptor ligands mimics exogenous opiates, highlighting the utility of this model for dissecting opiate signaling in mammals.


Subject(s)
Biogenic Monoamines/metabolism , Caenorhabditis elegans/metabolism , Neuropeptides/metabolism , Nociception , Opiate Alkaloids/pharmacology , Receptors, Opioid/metabolism , Animals , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Receptors, Opioid/agonists , Receptors, Opioid/genetics , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...