Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 166
Filter
1.
Drug Deliv ; 29(1): 3256-3269, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36321805

ABSTRACT

Acute liver injury is a common clinical disease, which easily leads to liver failure and endangers life, seriously threatening human health. Naringenin is a natural flavonoid that holds therapeutic potential against various liver injuries; however it has poor water solubility and bioavailability. In this study, we aimed to develop naringenin-loaded bovine serum albumin nanoparticles (NGNPs) and to evaluate their hepatoprotective effect and underlying mechanisms against acetaminophen overdose toxicity. In vitro data indicated that NGNPs significantly increased the drug solubility and also more effectively protected the hepatocyte cells from oxidative damage during hydrogen peroxide exposure or lipopolysaccharide (LPS) stimulation. In vivo results confirmed that NGNPs showed an enhanced accumulation in the liver tissue. In the murine model of acetaminophen-induced hepatotoxicity, NGNPs could effectively alleviate the progression of acute liver injury by reducing drug overdose-induced levels of oxidative stress, inflammation and apoptosis in hepatocytes. In conclusion, NGNPs has strong hepatoprotective effects against acetaminophen induced acute liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury , Drug Overdose , Nanoparticles , Mice , Humans , Animals , Acetaminophen/toxicity , Acetaminophen/metabolism , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/metabolism , Protective Agents/metabolism , Oxidative Stress , Liver , Drug Overdose/drug therapy , Drug Overdose/metabolism
2.
Sci Rep ; 12(1): 6429, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440607

ABSTRACT

The continuous rise in opioid overdoses in the United States is predominantly driven by very potent synthetic opioids, mostly fentanyl and its derivatives (fentanyls). Although naloxone (NLX) has been shown to effectively reverse overdoses by conventional opioids, there may be a need for higher or repeated doses of NLX to revert overdoses from highly potent fentanyls. Here, we used positron emission tomography (PET) to assess NLX's dose-dependence on both its rate of displacement of [11C]carfentanil ([11C]CFN) binding and its duration of mu opioid receptor (MOR) occupancy in the male rat brain. We showed that clinically relevant doses of intravenously (IV) administered NLX (0.035 mg/kg, Human Equivalent Dose (HED) 0.4 mg; 0.17 mg/kg, HED 2 mg) rapidly displaced the specific binding of [11C]CFN in the thalamus in a dose-dependent manner. Brain MOR occupancy by IV NLX was greater than 90% at 5 min after NLX administration for both doses, but at 27.3 min after 0.035 mg/kg dose and at 85 min after 0.17 mg/kg NLX, only 50% occupancy remained. This indicates that the duration of NLX occupancy at MORs is short-lived. Overall, these results show that clinically relevant doses of IV NLX can promptly displace fentanyls at brain MORs, but repeated or higher NLX doses may be required to prevent re-narcotization following overdoses with long-acting fentanyls.


Subject(s)
Analgesics, Opioid , Drug Overdose , Analgesics, Opioid/metabolism , Analgesics, Opioid/pharmacology , Animals , Brain/diagnostic imaging , Brain/metabolism , Drug Overdose/metabolism , Fentanyl/analogs & derivatives , Male , Naloxone , Rats , Receptors, Opioid, mu/metabolism , Tomography, X-Ray Computed
3.
Pharmacol Res ; 175: 106021, 2022 01.
Article in English | MEDLINE | ID: mdl-34883214

ABSTRACT

Acetaminophen (APAP) overdose is a major cause of acute liver failure, while the underlying mechanisms of APAP hepatotoxicity are not fully understood. Recently, emerging evidence suggests that epigenetic enzymes play roles in APAP-induced liver injury. Here, we found that Utx (ubiquitously transcribed tetratricopeptide repeat, X chromosome, also known as KDM6A), a X-linked histone demethylase which removes the di- and tri-methyl groups from histone H3K27, was markedly induced in the liver of APAP-overdosed female mice. Hepatic deletion of Utx suppressed APAP overdose-induced hepatotoxicity in female but not male mice. RNA-sequencing analysis suggested that Utx deficiency in female mice upregulated antitoxic phase II conjugating enzymes, including sulfotransferase family 2 A member 1 (Sult2a1), thus reduces the amount of toxic APAP metabolites in injured liver; while Utx deficiency also alleviated ER stress through downregulating transcription of ER stress genes including Atf4, Atf3, and Chop. Mechanistically, Utx promoted transcription of ER stress related genes in a demethylase activity-dependent manner, while repressed Sult2a1 expression through mediating H3K27ac levels independent of its demethylase activity. Moreover, overexpression of Sult2a1 in the liver of female mice rescued APAP-overdose induced liver injury. Together, our results indicated a novel UTX-Sult2a1 axis for the prevention or treatment of APAP-induced liver injury.


Subject(s)
Acetaminophen , Analgesics, Non-Narcotic , Chemical and Drug Induced Liver Injury , Histone Demethylases , Animals , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Drug Overdose/metabolism , Endoplasmic Reticulum Stress , Female , Histone Demethylases/genetics , Histone Demethylases/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Sex Characteristics , Sulfotransferases/genetics
4.
BMC Pharmacol Toxicol ; 22(1): 59, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34666816

ABSTRACT

BACKGROUND: Tramadol is a widely used synthetic opioid for moderate to severe pain. Some studies have shown that tramadol can increase oxidative stress in different tissues of the body. Quercetin is also a substance with various biological effects, including antioxidant, anti-inflammatory, hepatoprotective, nephroprotective, and cardioprotective activities. The current investigation aimed at determining the effects of quercetin, with or without naloxone, on tramadol intoxication. METHODS: This study was performed on 30 male Wistar rats divided into five groups: Group I) control group: intraperitoneal injections of normal saline 0.9% for 14 days; Group II) tramadol: 25 mg/kg for 14 days, and then a 50 mg/kg acute dose injection on the last day; Group III) acute quercetin (single dose): tramadol injection as with the second group plus 100 mg/kg of quercetin on the last day; Group IV) chronic quercetin: tramadol injection similar to the second group plus quercetin 100 mg/kg for 14 days; Group V) quercetin plus naloxone: tramadol injection similar to the second group plus injection of quercetin 100 mg/kg + intravenous naloxone 2 mg/kg on the last day, followed by a 4 mg/kg/h injection of naloxone for six hours. The rats were monitored for six hours on the last day, relating to the number and severity of seizures. Finally, the samples were prepared for biochemical investigation of the serum level of oxidative stress markers (MDA, SOD, NOx), inflammatory factors (IL-6, TNF-α), biochemical parameters (ALT, AST, creatinine, glucose) and hematological assay. The liver, heart, kidney, cortex, cerebellum, and adrenal tissues were collected to investigate the redox state. RESULTS: None of the treatments had positive effects on the number and severity of seizures. Chronic administration of quercetin led to alteration of some blood parameters, including reduced hemoglobin level and elevated platelet counts. Acute on chronic tramadol administration resulted in a significant rise in AST, where different treatments failed to reduce their levels down to the control group. CONCLUSION: chronic administration of quercetin showed decreased oxidative/nitrosative stress in the liver, kidney, adrenal, and heart tissues. Quercetin plus naloxone decreased oxidative stress in the heart and adrenal tissues, but adverse effects on the brain cortex and hepatic function. Single-dose quercetin reduced cardiac oxidative stress.


Subject(s)
Analgesics, Opioid/toxicity , Drug Overdose/drug therapy , Quercetin/therapeutic use , Seizures/drug therapy , Tramadol/toxicity , Adrenal Glands/drug effects , Adrenal Glands/metabolism , Animals , Brain/drug effects , Brain/metabolism , Drug Overdose/metabolism , Heart/drug effects , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Kidney/drug effects , Liver/drug effects , Liver/metabolism , Male , Malondialdehyde/metabolism , Myocardium/metabolism , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Quercetin/adverse effects , Rats, Wistar , Seizures/chemically induced , Seizures/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
5.
Mol Nutr Food Res ; 65(8): e2001176, 2021 04.
Article in English | MEDLINE | ID: mdl-33641237

ABSTRACT

SCOPE: Manganese (Mn) and zinc (Zn) are not only essential trace elements, but also potential exogenous risk factors for various diseases. Since the disturbed homeostasis of single metals can result in detrimental health effects, concerns have emerged regarding the consequences of excessive exposures to multiple metals, either via nutritional supplementation or parenteral nutrition. This study focuses on Mn-Zn-interactions in the nematode Caenorhabditis elegans (C. elegans) model, taking into account aspects related to aging and age-dependent neurodegeneration. METHODS AND RESULTS: Chronic co-exposure of C. elegans to Mn and Zn increases metal uptake, exceeding levels of single metal exposures. Supplementation with Mn and/or Zn also leads to an age-dependent increase in metal content, a decline in overall mRNA expression, and metal co-supplementation induced expression of target genes involved in Mn and Zn homeostasis, in particular metallothionein 1 (mtl-1). Studies in transgenic worms reveal that mtl-1 played a prominent role in mediating age- and diet-dependent alterations in metal homeostasis. Metal dyshomeostasis is further induced in parkin-deficient nematodes (Parkinson's disease (PD) model), but this did not accelerate the age-dependent dopaminergic neurodegeneration. CONCLUSIONS: A nutritive overdose of Mn and Zn can alter interactions between essential metals in an aging organism, and metallothionein 1 acts as a potential protective modulator in regulating homeostasis.


Subject(s)
Aging/drug effects , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/drug effects , Manganese/adverse effects , Metallothionein/metabolism , Zinc/adverse effects , Aging/physiology , Animals , Animals, Genetically Modified , Biological Availability , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Drug Overdose/metabolism , Homeostasis/drug effects , Homeostasis/genetics , Manganese/administration & dosage , Manganese/pharmacokinetics , Metallothionein/genetics , Mutation , Toxicity Tests, Chronic , Ubiquitin-Protein Ligases/genetics , Zinc/administration & dosage , Zinc/pharmacokinetics
6.
Cell Mol Neurobiol ; 41(8): 1635-1649, 2021 Nov.
Article in English | MEDLINE | ID: mdl-32712727

ABSTRACT

Acute methadone toxicity is a major public health concern which has adverse effects on brain tissue and results in recurrent or delayed respiratory arrest. Our study aimed to investigate the time-dependent changes in several serum biochemical markers of brain damage, spatial working memory, and the brain tissue following acute methadone overdose. Adolescent male rats underwent an intraperitoneal (i.p.) injection of 15 mg/kg methadone. In case of apnea occurrence, resuscitation was performed by a ventilatory pump and administrating naloxone (2 mg/kg; i.p.). The animals were classified into groups of treated rats; methadone and naloxone-Apnea (M/N-Apnea), M/N-Sedate, Methadone, Naloxone, and control (saline) groups. The serum levels of S100B, neuron-specific enolase (NSE), myelin basic protein factors, and (Lactate/Pyruvate) L/P ratio were evaluated at the time-points of 6, 24, and 48 h (h). We found that the alterations of S100B and L/P ratio were considerable in the M/N-Apnea and Methadone groups from the early hours post-methadone overdose, while NSE serum levels elevation was observed only in M/N-Apnea group with a delay at 48 h. Further, we assessed the spatial working memory (Y-maze test), morphological changes, and neuronal loss. The impaired spontaneous alternation behavior was detected in the M/N-Apnea groups on days 5 and 10 post-methadone overdose. The morphological changes of neurons and the neuronal loss were detectable in the CA1, striatum, and cerebellum regions, which were pronounced in both M/N-Apnea and Methadone groups. Together, our findings suggest that alterations in the serum levels of S100B and NSE factors as well as L/P ratio could be induced by methadone overdose with the presence or absence of apnea before the memory impairment and tissue injury in adolescent male rats.


Subject(s)
Analgesics, Opioid/toxicity , Drug Overdose/blood , Inflammation Mediators/blood , Methadone/toxicity , Age Factors , Animals , Biomarkers/blood , Brain/drug effects , Brain/metabolism , Brain/pathology , Drug Overdose/metabolism , Drug Overdose/pathology , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory, Short-Term/drug effects , Memory, Short-Term/physiology , Phosphopyruvate Hydratase/blood , Rats , Rats, Wistar , S100 Calcium Binding Protein beta Subunit/blood , Time Factors
7.
Clin Toxicol (Phila) ; 59(3): 231-234, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32734785

ABSTRACT

INTRODUCTION: Severe baclofen toxicity can result in respiratory failure, hemodynamic instability, bradycardia, hypothermia, seizures, coma, and death. While hemodialysis (HD) is well-described in treating acute baclofen toxicity in patients with end-stage kidney disease or acute kidney injury, the utility of HD for patients with normal kidney function is uncertain. Implementing HD to speed recovery after a large acute baclofen ingestion is appealing, considering: (a) potential for prolonged coma and ventilator-associated morbidity, and (b) baclofen's low protein-binding, low molecular-weight, and moderate volume of distribution. METHODS: We report a 51 kg, 14-year-old girl who presented to the emergency department (ED) with hypotension, obtundation, and status epilepticus after an intentional ingestion of 1200 mg baclofen. Her post-intubation neurologic examination was concerning for coma. A 14-hour post-ingestion baclofen concentration was 882 ng/mL (therapeutic range 80-400 ng/mL). Three urgent-HD sessions were performed to reduce her time on the ventilator. RESULTS: The total baclofen removed in the first three-hour HD session was 3.05 mg. The total urinary elimination of baclofen 42 mg over 24-hours on day one. She was discharged without neurologic deficits to psychiatry on day-14. CONCLUSION: In this case, the amount of baclofen recovered during HD is negligible in comparison to the amount cleared by kidney elimination in this patient with normal kidney function.


Subject(s)
Baclofen/toxicity , Drug Overdose/therapy , Renal Dialysis , Adolescent , Baclofen/blood , Baclofen/pharmacokinetics , Baclofen/urine , Drug Overdose/metabolism , Emergency Service, Hospital , Female , Humans , Kidney/metabolism , Suicide, Attempted
8.
Neurochem Int ; 143: 104936, 2021 02.
Article in English | MEDLINE | ID: mdl-33309980

ABSTRACT

Long term consequence of non-fatal overdose in people who use opioids are not well understood. The intermittent exposure to non-fatal overdose leads to a tauopathy that is often accompanied by abrogated neuroprotective response, abnormal amyloid processing and other pathologies. The scope and limitations of available literature are discussed including neuropathologies associated with opioid and overdose exposures, contributing comorbidities and proteinopathies. Contrasting postmortem data of overdose victims with animal models of opioid neuropathologies and hypoxic injury paints a picture distinct from other proteinopathies as well as effects of moderate opioid exposure. Furthermore the reported biochemical changes and potential targets for therapeutic intervention were mapped pointing to underlying imbalance between tau kinases and phosphatases that is characteristic of Alzheimer Disease.


Subject(s)
Alzheimer Disease/metabolism , Analgesics, Opioid/adverse effects , Drug Overdose/metabolism , Opioid-Related Disorders/metabolism , Alzheimer Disease/pathology , Animals , Drug Overdose/pathology , Humans , Opioid-Related Disorders/pathology , tau Proteins/metabolism
9.
PLoS One ; 15(4): e0231173, 2020.
Article in English | MEDLINE | ID: mdl-32251487

ABSTRACT

Acetaminophen is one of the most common over-the-counter pain medications used worldwide and is considered safe at therapeutic dose. However, intentional and unintentional overdose accounts for up to 70% of acute liver failure cases in the western world. Extensive research has demonstrated that the induction of oxidative stress and mitochondrial dysfunction are central to the development of acetaminophen-induced liver injury. Despite the insight gained on the mechanism of acetaminophen toxicity, there still is only one clinically approved pharmacological treatment option, N-acetylcysteine. N-acetylcysteine increases the cell's antioxidant defense and protects liver cells from further acetaminophen-induced oxidative damage. Because it primarily protects healthy liver cells rather than rescuing the already injured cells alternative treatment strategies that target the latter cell population are warranted. In this study, we investigated mitochondria as therapeutic target for the development of novel treatment strategies for acetaminophen-induced liver injury. Characterization of the mitochondrial toxicity due to acute acetaminophen overdose in vitro in human cells using detailed respirometric analysis revealed that complex I-linked (NADH-dependent) but not complex II-linked (succinate-dependent) mitochondrial respiration is inhibited by acetaminophen. Treatment with a novel cell-permeable succinate prodrug rescues acetaminophen-induced impaired mitochondrial respiration. This suggests cell-permeable succinate prodrugs as a potential alternative treatment strategy to counteract acetaminophen-induced liver injury.


Subject(s)
Acetaminophen/adverse effects , Analgesics, Non-Narcotic/adverse effects , Cell Membrane Permeability/drug effects , Cell Respiration/drug effects , Drug Overdose/drug therapy , Mitochondria/metabolism , Prodrugs/pharmacokinetics , Succinic Acid/pharmacokinetics , Acetaminophen/administration & dosage , Acetaminophen/pharmacology , Aged , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/pharmacology , Blood Platelets/metabolism , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/metabolism , Drug Overdose/metabolism , Electron Transport Complex I/antagonists & inhibitors , Hep G2 Cells , Hepatocytes/metabolism , Humans , Male , Mitochondria/drug effects
10.
Biomarkers ; 25(1): 9-19, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31735069

ABSTRACT

Acute intoxications account for a significant proportion of the patient population in intensive care units and sedative medications, ethanol, illicit drugs, inhalable poisons and mixed intoxications are the most common causes. The aim of this article is to describe biomarkers for screening and diagnosis of acute intoxications in critically ill patients. For this purpose, a survey of the relevant literature was conducted, and guidelines, case reports, expert assessments, and scientific publications were reviewed. In critical care, it should always be attempted to identify and quantify the poison or toxin with the assistance of enzyme immunoassay (EIA), chromatography, and mass spectrometry techniques and this section is critically appraised in this publication. The principles for anion gap, osmol gap and lactate gap and their usage in intoxications is shown. Basic rules in test methodology and pre-analytics are reviewed. Biomarkers in general are presented in part one and biomarkers for specific intoxications including ethanol, paracetamol, cardiovascular drugs and many others are presented in part two of these publications.


Subject(s)
Biomarkers/metabolism , Drug Overdose/diagnosis , Drug-Related Side Effects and Adverse Reactions/diagnosis , Poisoning/diagnosis , Acute Disease , Critical Illness , Drug Overdose/epidemiology , Drug Overdose/metabolism , Drug Overdose/therapy , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/metabolism , Drug-Related Side Effects and Adverse Reactions/therapy , Humans , Poisoning/epidemiology , Poisoning/metabolism , Poisoning/therapy , Predictive Value of Tests , Prognosis
11.
J Neuropathol Exp Neurol ; 78(11): 1059-1065, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31559425

ABSTRACT

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


Subject(s)
Brain/drug effects , Brain/pathology , Cocaine-Related Disorders/pathology , Encephalitis/pathology , Heroin Dependence/pathology , Adolescent , Adult , Autopsy , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/metabolism , Cocaine-Related Disorders/complications , Cocaine-Related Disorders/metabolism , Drug Overdose/metabolism , Drug Overdose/pathology , Encephalitis/etiology , Encephalitis/metabolism , Female , Heroin Dependence/complications , Heroin Dependence/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Young Adult
12.
J Pharmacol Exp Ther ; 371(2): 453-475, 2019 11.
Article in English | MEDLINE | ID: mdl-31492824

ABSTRACT

In December 2018, the Centers for Disease Control declared fentanyl the deadliest drug in America. Opioid overdose is the single greatest cause of death in the United States adult population (ages 18-50), and fentanyl and its analogs [fentanyl/fentanyl analogs (F/FAs)] are currently involved in >50% of these deaths. Anesthesiologists in the United States were introduced to fentanyl in the early 1970s when it revolutionized surgical anesthesia by combining profound analgesia with hemodynamic stability. However, they quickly had to master its unique side effect. F/FAs can produce profound rigidity in the diaphragm, chest wall and upper airway within an extremely narrow dosing range. This clinical effect was called wooden chest syndrome (WCS) by anesthesiologists and is not commonly known outside of anesthesiology or to clinicians or researchers in addiction research/medicine. WCS is almost routinely fatal without expert airway management. This review provides relevant clinical human pharmacology and animal data demonstrating that the significant increase in the number of F/FA-induced deaths may involve α-adrenergic and cholinergic receptor-mediated mechanical failure of the respiratory and cardiovascular systems with rapid development of rigidity and airway closure. Although morphine and its prodrug, heroin, can cause mild rigidity in abdominal muscles at high doses, neither presents with the distinct and rapid respiratory failure seen with F/FA-induced WCS, separating F/FA overdose from the slower onset of respiratory depression caused by morphine-derived alkaloids. This distinction has significant consequences for the design and implementation of new pharmacologic strategies to effectively prevent F/FA-induced death. SIGNIFICANCE STATEMENT: Deaths from fentanyl and F/FAs are increasing in spite of availability and awareness of the opioid reversal drug naloxone. This article reviews literature suggesting that naloxone may be ineffective against centrally mediated noradrenergic and cholinergic effects of F/FAs, which clinically manifest as severe muscle rigidity and airway compromise (e.g., wooden chest syndrome) that is rapid and distinct from respiratory depression seen with morphine-derived alkaloids. A physiologic model is proposed and implications for new drug development and treatment are discussed.


Subject(s)
Adrenergic Neurons/drug effects , Analgesics, Opioid/adverse effects , Fentanyl/adverse effects , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Opioid Epidemic/prevention & control , Adrenergic Neurons/metabolism , Analgesics, Opioid/metabolism , Drug Overdose/metabolism , Drug Overdose/prevention & control , Fentanyl/metabolism , Humans , Muscle Rigidity/chemically induced , Muscle Rigidity/drug therapy , Muscle Rigidity/metabolism , Naloxone/metabolism , Narcotic Antagonists/metabolism , Opioid Epidemic/trends , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/drug therapy , Respiratory Insufficiency/metabolism , Time-to-Treatment/trends
13.
Neth J Med ; 77(5): 189-192, 2019 06.
Article in English | MEDLINE | ID: mdl-31264585
14.
ACS Chem Neurosci ; 10(8): 3486-3499, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31257858

ABSTRACT

The United States of America is fighting against one of its worst-ever drug crises. Over 900 people a week die from opioid- or heroin-related overdoses, while millions more suffer from opioid prescription addiction. Recently, drug overdoses caused by fentanyl-laced cocaine specifically are on the rise. Due to drug synergy and an increase in side effects, polydrug addiction can cause more risk than addiction to a single drug. In the present work, we systematically analyzed the overdose and addiction mechanism of cocaine and fentanyl. First, we applied our established chemogenomics knowledgebase and machine-learning-based methods to map out the potential and known proteins, transporters, and metabolic enzymes and the potential therapeutic target(s) for cocaine and fentanyl. Sequentially, we looked into the detail of (1) the addiction to cocaine and fentanyl by binding to the dopamine transporter and the µ opioid receptor (DAT and µOR, respectively), (2) the potential drug-drug interaction of cocaine and fentanyl via p-glycoprotein (P-gp) efflux, (3) the metabolism of cocaine and fentanyl in CYP3A4, and (4) the physiologically based pharmacokinetic (PBPK) model for two drugs and their drug-drug interaction at the absorption, distribution, metabolism, and excretion (ADME) level. Finally, we looked into the detail of JWH133, an agonist of cannabinoid 2-receptor (CB2) with potential as a therapy for cocaine and fentanyl overdose. All these results provide a better understanding of fentanyl and cocaine polydrug addiction and future drug abuse prevention.


Subject(s)
Cocaine , Drug Overdose , Fentanyl , Machine Learning , Opioid-Related Disorders , Analgesics, Opioid , Cocaine/adverse effects , Cocaine/metabolism , Cocaine/pharmacology , Cocaine-Related Disorders/metabolism , Computer Simulation , Drug Overdose/metabolism , Fentanyl/adverse effects , Fentanyl/metabolism , Fentanyl/pharmacology , Humans
15.
J Clin Pharmacol ; 59(10): 1291-1299, 2019 10.
Article in English | MEDLINE | ID: mdl-31099052

ABSTRACT

Previous reports have questioned the safety of multiple doses of acetaminophen administered to ill children. Acetaminophen protein adducts (adducts) are a biomarker of acetaminophen-induced liver injury and reflect the oxidative metabolism of acetaminophen, a known mechanism in acetaminophen toxicity. In this prospective observational study, we analyzed adduct concentrations in 1034 blood samples obtained from 181 hospitalized children (1 to 18 years inclusive) who received 2 or more doses of acetaminophen. Linear regression analysis showed that serum adduct concentrations increased as a function of the cumulative acetaminophen dose, which could be attributed, in part, to a long half-life of adducts (2.17 ± 1.04 days [mean ± standard deviation]) in children. However, few patients (2%) were found to have adduct concentrations higher than 1.0 nmol/mL, a previously identified toxicity cut point for the diagnosis of acetaminophen-induced liver injury in patients with alanine aminotransferase values exceeding 1000 IU/L. A small cohort of patients with suspected infection was noted to show higher adduct concentrations. In addition, adduct concentrations showed a stronger correlation with cumulative acetaminophen doses in adolescents compared with children (R2 = 0.41 vs 0.26). No other covariates (body weight, body mass index z score, sex, race, or surgery) remarkably correlated with adduct elevation. In summary, low levels of adducts can be detected in hospitalized children receiving multiple doses of acetaminophen, and adduct levels correlate with cumulative acetaminophen dose.


Subject(s)
Acetaminophen/adverse effects , Acetaminophen/blood , Analgesics, Non-Narcotic/adverse effects , Analgesics, Non-Narcotic/blood , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/etiology , Drug Overdose/blood , Acetaminophen/administration & dosage , Adolescent , Alanine Transaminase/metabolism , Analgesics, Non-Narcotic/administration & dosage , Biomarkers/blood , Child , Child, Hospitalized , Child, Preschool , Drug Overdose/etiology , Drug Overdose/metabolism , Female , Half-Life , Humans , Infant , Liver/metabolism , Male , Prospective Studies
16.
Sci Rep ; 9(1): 5971, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30979951

ABSTRACT

Acetaminophen (APAP) overdose (APAPo) is predominant in the NIH Pediatric Acute Liver Failure (PALF) Study. We assayed multiple inflammatory mediators in serial serum samples from 13 PALF survivors with APAPo + N-acetylcysteine (NAC, the frontline therapy for APAPo), 8 non-APAPo + NAC, 40 non-APAPo non-NAC, and 12 non-survivors. High Mobility Group Box 1 (HMGB1) was a dominant mediator in dynamic inflammation networks in all sub-groups, associated with a threshold network complexity event at d1-2 following enrollment that was exceeded in non-survivors vs. survivors. We thus hypothesized that differential HMGB1 network connectivity after day 2 is related to the putative threshold event in non-survivors. DyNA showed that HMGB1 is most connected in non-survivors on day 2-3, while no connections were observed in APAPo + NAC and non-APAPo + NAC survivors. Inflammatory dynamic networks, and in particular HMGB1 connectivity, were associated with the use of NAC in the context of APAPo. To recapitulate hepatocyte (HC) damage in vitro, primary C57BL/6 HC and HC-specific HMGB1-null HC were treated with APAP + NAC. Network phenotypes of survivors were recapitulated in C57BL/6 mouse HC and were greatly altered in HMGB1-null HC. HC HMGB1 may thus coordinate a pro-inflammatory program in PALF non-survivors (which is antagonized by NAC), while driving an anti-inflammatory/repair program in survivors.


Subject(s)
Acetaminophen/adverse effects , Analgesics, Non-Narcotic/adverse effects , Chemical and Drug Induced Liver Injury/metabolism , HMGB1 Protein/metabolism , Liver Failure, Acute/chemically induced , Liver Failure, Acute/metabolism , Adolescent , Animals , Biomarkers/blood , Child , Child, Preschool , Cohort Studies , Drug Overdose/metabolism , HMGB1 Protein/genetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Infant , Inflammation/chemically induced , Inflammation/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Primary Cell Culture
17.
J Pharmacol Exp Ther ; 370(1): 84-91, 2019 07.
Article in English | MEDLINE | ID: mdl-31010842

ABSTRACT

The illicit use of γ-hydroxybutyric acid (GHB), and its prodrug, γ-butyrolactone (GBL), results in severe adverse effects including sedation, coma, respiratory depression, and death. Current treatment of GHB/GBL overdose is limited to supportive care. Recent reports indicate that GHB-related deaths are on the rise; a specific treatment may reduce lethality associated with GHB/GBL. Pretreatment with inhibitors of monocarboxylate transporter 1 (MCT1), a transporter that mediates many of the processes involved in the absorption, distribution (including brain uptake), and elimination of GHB/GBL, has been shown to prevent GHB-induced respiratory depression by increasing the renal clearance of GHB. To identify whether MCT1 inhibition is an effective treatment of GHB overdose, the impact of two MCT1 inhibitors, (S)-5-(4-hydroxy-4-methylisoxazolidine-2-carbonyl)-1-isopropyl-3-methyl-6-((3-methyl-5-(trifluoromethyl)-1H-pyrazol-4-yl)methyl)thieno[2,3-day]pyrimidine-2,4(1H,3H)-dione (AZD3965) and 6-[(3,5-dimethyl-1H-pyrazol-4-yl)methyl]-5-[[(4S)-4-hydroxy-2-isoxazolidinyl]carbonyl]-3-methyl-1-(2-methylpropyl)thieno[2,3-day]pyrimidine2,4(1H,3H)-dione (AR-C155858), on the toxicokinetics and toxicodynamics of GHB/GBL was assessed when the administration of the inhibitor was delayed 60 and 120 minutes (post-treatment) after administration of GHB/GBL. AR-C155858 and AZD3965 reduced the toxicodynamic effects of GHB when GHB was administered intravenously, orally, or orally as the prodrug GBL. The impact of these inhibitors on GHB toxicokinetics was dependent on the route of GHB administration and the delay between GHB/GBL administration and administration of the MCT1 inhibitor. The reduction in GHB plasma exposure did not explain the observed effect of MCT1 inhibition on GHB-induced respiratory depression. The efficacy of MCT1 inhibition on GHB toxicodynamics is likely driven by the pronounced reduction in GHB brain concentrations. Overall, this study indicates that inhibition of MCT1 is an effective treatment of GHB/GBL overdose.


Subject(s)
4-Butyrolactone/toxicity , Drug Overdose/drug therapy , Hydroxybutyrates/toxicity , Monocarboxylic Acid Transporters/antagonists & inhibitors , Prodrugs/pharmacology , Pyrimidinones/pharmacology , Symporters/antagonists & inhibitors , Thiophenes/pharmacology , Uracil/analogs & derivatives , 4-Butyrolactone/administration & dosage , Administration, Intravenous , Administration, Oral , Animals , Brain/drug effects , Brain/metabolism , Drug Overdose/blood , Drug Overdose/metabolism , Hydroxybutyrates/administration & dosage , Hydroxybutyrates/blood , Hydroxybutyrates/pharmacokinetics , Male , Pyrimidinones/therapeutic use , Rats , Rats, Sprague-Dawley , Thiophenes/therapeutic use , Uracil/pharmacology , Uracil/therapeutic use
18.
Mol Pharmacol ; 95(6): 597-605, 2019 06.
Article in English | MEDLINE | ID: mdl-30944208

ABSTRACT

Overdose of acetaminophen (APAP) is the leading cause of acute liver failure (ALF) in the United States. The sulfotransferase-mediated sulfation of APAP is widely believed to be a protective mechanism to attenuate the hepatotoxicity of APAP. The cholesterol sulfotransferase SULT2B1b is best known for its activity in catalyzing the sulfoconjugation of cholesterol to synthesize cholesterol sulfate. SULT2B1b can be transcriptionally and positively regulated by the hepatic nuclear factor 4α (HNF4α). In this study, we uncovered an unexpected role for SULT2B1b in APAP toxicity. Hepatic overexpression of SULT2B1b sensitized mice to APAP-induced liver injury, whereas ablation of the Sult2B1b gene in mice conferred resistance to the APAP hepatotoxicity. Consistent with the notion that Sult2B1b is a transcriptional target of HNF4α, overexpression of HNF4α sensitized mice or primary hepatocytes to APAP-induced hepatotoxicity in a Sult2B1b-dependent manner. We conclude that the HNF4α-SULT2B1b axis has a unique role in APAP-induced acute liver injury, and SULT2B1b induction might be a risk factor for APAP hepatotoxicity.


Subject(s)
Acetaminophen/adverse effects , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Drug Overdose/complications , Hepatocyte Nuclear Factor 4/metabolism , Sulfotransferases/genetics , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury, Chronic/genetics , Disease Models, Animal , Drug Overdose/etiology , Drug Overdose/metabolism , Female , Hepatocytes/cytology , Hepatocytes/metabolism , Mice , Sulfotransferases/metabolism
19.
Medisan ; 23(2)mar.-abr. 2019. ilus
Article in Spanish | LILACS | ID: biblio-1002636

ABSTRACT

Los digitálicos son fármacos con capacidad de aumentar la contractilidad miocárdica (inotrópico positivo), que han desempeñado un rol primordial en el tratamiento de pacientes con insuficiencia cardiaca; su uso inapropiado puede traer complicaciones serias a estos pacientes, incluso, hasta la muerte. La más importante de estas complicaciones es la intoxicación digitálica, originada por la sobredosis de dichos fármacos, a causa de la combinación del efecto inhibitorio en la conducción nodal y la estimulación sobre las fibras individuales auriculares y ventriculares. Debido al uso frecuente de estos medicamentos en todos los niveles de atención de salud y lo difícil que resulta diagnosticar dicha complicación por la complejidad de su cuadro clínico y de su expresión electrocardiográfica, se realizó una revisión bibliográfica exhaustiva sobre el tema para brindar amplia información, que permita una atención adecuada a los pacientes con este diagnóstico


Digitalis are drugs with the capacity of increasing myocardial contractility (inotropic positive agents) which have carried out an important role in the treatment of heart failure; their inappropriate use can bring severe complications to the patient, even, to death. The most important in these complications is the digitalis toxicity, originated by the overdose of these drugs, caused by the combination of the inhibitory effect in the nodal conduction and stimulation on the individual atrial and ventricular fibers. Due to the frequent use of these medications at all levels of medical care and to the difficulty in diagnosing this complication caused by the complexity of their clinical pattern and of their electrocardiographic expression, an exhaustive literature review was carried out on the topic to give a wide information that allows an appropriate care to the patients with this diagnosis


Subject(s)
Humans , Male , Female , Digitalis Glycosides/poisoning , Digitalis Glycosides/pharmacology , Drug-Related Side Effects and Adverse Reactions , Myocardial Contraction/drug effects , Poisoning , Drug Overdose/metabolism
20.
Article in English | MEDLINE | ID: mdl-30760194

ABSTRACT

OBJECTIVE: The aim of the study was to examine the impact of neonatal acetaminophen (APAP; paracetamol) administrations on the thyroid-liver axis in male Wistar rats. METHODS: APAP (100 or 350mg/kg) was orally administered to neonates from Postnatal Day (PND) 20 to 40. RESULTS: Both APAP doses elicited a substantial increase in serum TSH, albumin, AST, ALT, and ALP values, and a profound decrease in serum FT4 and FT3 values at PND 40 relative to those in the control group. Additionally, the hypothyroid state in both APAP-treated groups may increase the histopathological variations in the neonatal liver, such as destructive degeneration, fibrosis, fatty degeneration, fibroblast proliferation, haemorrhage, oedema, and vacuolar degeneration, at PND 40. Moreover, in the APAP groups, a marked depression was recorded in the t-SH and GSH levels and GPx and CAT activities at PND 40 in the neonatal liver compared to those in the control group. However, the levels of hepatic LPO, H2O2, and NO were increased in both APAP-treated groups at PND 40. All previous alterations were dose- dependent. CONCLUSION: Neonatal APAP caused a hypothyroidism and disturbed hepatic cellular components by increasing prooxidant markers and decreasing antioxidant markers, causing hepatotoxicity. Thus, neonatal administrations of APAP may act as a neonatal thyroid-liver disruptor.


Subject(s)
Acetaminophen/adverse effects , Drug Overdose , Liver/drug effects , Thyroid Gland/drug effects , Acetaminophen/administration & dosage , Acetaminophen/poisoning , Animals , Animals, Newborn , Biomarkers/analysis , Biomarkers/metabolism , Drug Overdose/complications , Drug Overdose/metabolism , Growth and Development/drug effects , Liver/growth & development , Liver/physiology , Male , Metabolic Networks and Pathways/drug effects , Rats , Rats, Wistar , Thyroid Gland/growth & development , Thyroid Gland/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...