Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 318
Filter
1.
BMC Vet Res ; 16(1): 368, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32998730

ABSTRACT

BACKGROUND: Meperidine is a synthetic opioid that belongs to the phenylpiperidine class and is a weak mu receptor agonist. In horses there are a limited number of published studies describing the analgesic effects of systemically administered meperidine in horses. The objective of this study was to describe the pharmacokinetics, behavioral and physiologic effects and effect on thermal threshold of three doses of intravenously administered meperidine to horses. Eight University owned horses (four mares and four geldings, aged 3-8 years were studied using a randomized balanced 4-way cross-over design. Horses received a single intravenous dose of saline, 0.25, 0.5 and 1.0 mg/kg meperidine. Blood was collected before administration and at various time points until 96 hours post administration. Plasma and urine samples were analyzed for meperidine and normeperidine by liquid chromatography-mass spectrometry and plasma pharmacokinetics determined. Behavioral and physiologic data (continuous heart rate, step counts, packed cell volume, total plasma protein and gastrointestinal sounds) were collected at baseline through 6 hours post administration. The effect of meperidine administration on thermal nociception was determined and thermal excursion calculated. RESULTS: Meperidine was rapidly converted to the metabolite normeperidine. The volume of distribution at steady state and systemic clearance (mean ± SD) ranged from 0.829 ± 0.138-1.58 ± 0.280 L/kg and 18.0 ± 1.4-22.8 ± 3.60 mL/min/kg, respectively for 0.5-1.0 mg/kg doses. Adverse effects included increased dose-dependent central nervous excitation, heart rate and cutaneous reactions. Significant effects on thermal nociception were short lived (up to 45 minutes at 0.5 mg/kg and 15 minutes at 1.0 mg/kg). CONCLUSIONS: Results of the current study do not support routine clinical use of IV meperidine at a dose of 1 mg/kg to horses. Administration of 0.5 mg/kg may provide short-term analgesia, however, the associated inconsistent and/or short-term adverse effects suggest that its use as a sole agent at this dose, at best, must be cautiously considered.


Subject(s)
Analgesics, Opioid/pharmacology , Analgesics, Opioid/pharmacokinetics , Meperidine/pharmacology , Meperidine/pharmacokinetics , Administration, Intravenous/veterinary , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Animals , Central Nervous System/drug effects , Female , Heart Rate/drug effects , Horses , Hot Temperature , Male , Meperidine/administration & dosage , Meperidine/adverse effects , Meperidine/analogs & derivatives , Meperidine/blood , Meperidine/urine , Nociception/drug effects , Urticaria
2.
J Pharm Biomed Anal ; 186: 113320, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32387748

ABSTRACT

Pethidine is an opiate agonist used orally and parenterally. Pethidine-containing drugs abuse is frequently encountered on health workers and patients. The analysis methods used to determine the abuse of pethidine are important for forensic toxicology. Pethidine is metabolized to norpethidine by the liver. Therefore, the determination of pethidine and norpethidine in urine is one of the methods to determine the abuse of pethidine. In this study, we have developed a precise, simple and rapid ultra-performance liquid chromatography-tandem mass spectrometer method for the determination of pethidine and norpethidine simultaneously. The developed method was validated in terms of selectivity and linearity which was in the range of 9-1800 ng/mL for both pethidine and norpethidine. The intra-assay and inter-assay accuracy and precision were found within acceptable limits of the EMA guideline. Lower limits of quantitation were 9 ng/mL for both pethidine and norpethidine. The developed method was successfully applied for the determination of both analytes in the real samples.


Subject(s)
Analgesics, Opioid/urine , Chromatography, High Pressure Liquid/methods , Meperidine/analogs & derivatives , Tandem Mass Spectrometry/methods , Adult , Analgesics, Opioid/analysis , Female , Humans , Limit of Detection , Male , Meperidine/analysis , Meperidine/urine , Reproducibility of Results , Substance Abuse Detection/methods
3.
Xenobiotica ; 50(2): 209-222, 2020 Feb.
Article in English | MEDLINE | ID: mdl-30902024

ABSTRACT

1. Meperidine is an opioid analgesic that undergoes N-demethylation to form the neurotoxic metabolite normeperidine. Previous studies indicate that meperidine N-demethylation is catalyzed by cytochrome P450 2B6 (CYP2B6), CYP3A4, and CYP2C19.2. The purpose of this study was to examine the relative P450 contributions to meperidine N-demethylation and to evaluate the effect of CYP2C19 polymorphism on normeperidine generation. Experiments were performed using recombinant P450 enzymes, selective chemical inhibitors, enzyme kinetic assays, and correlation analysis with individual CYP2C19-genotyped human liver microsomes.3. The catalytic efficiency (kcat/Km) for meperidine N-demethylation was similar between recombinant CYP2B6 and CYP2C19, but markedly lower by CYP3A4.4. In CYP2C19-genotyped human liver microsomes, normeperidine formation was significantly correlated with CYP2C19 activity (S-mephenytoin 4´-hydroxylation).5. CYP2C19 inhibitor (+)-N-3-benzylnirvanol and CYP3A inhibitor ketoconazole significantly reduced microsomal normeperidine generation by an individual donor with high CYP2C19 activity, whereas donors with lower CYP2C19 activity were sensitive to inhibition by ketoconazole but not benzylnirvanol.6. These findings demonstrate that the relative CYP3A4, CYP2B6, and CYP2C19 involvement in meperidine N-demethylation depends on the enzyme activities in individual human liver microsomal samples. CYP2C19 is likely an important contributor to normeperidine generation in individuals with high CYP2C19 activity, but additional factors influence inter-individual metabolite accumulation.


Subject(s)
Cholinesterase Inhibitors/metabolism , Cytochrome P-450 Enzyme System/metabolism , Meperidine/analogs & derivatives , Cytochrome P-450 CYP2C19/metabolism , Demethylation , Humans , Meperidine/metabolism , Mephenytoin
4.
Future Med Chem ; 11(22): 2905-2917, 2019 11.
Article in English | MEDLINE | ID: mdl-31713454

ABSTRACT

Aim: Glycosphingolipids are conserved lipids displaying a variety of functions in fungal cells, such as determination of cell polarity and virulence. They have been considered as potent targets for new antifungal drugs. The present work aimed to test two inhibitors, myriocin and DL-threo-1-Phenyl-2-palmitoylamino-3-morpholino-1-propanol, in Scedosporium boydii, a pathogenic fungus which causes a wide range of disease. Materials & methods: Mass spectrometry, microscopy and cell biology approaches showed that treatment with both inhibitors led to defects in fungal growth and membrane integrity, and caused an increased susceptibility to the current antifungal agents. Conclusion: These data demonstrate the antifungal potential of drugs inhibiting sphingolipid biosynthesis, as well as the usefulness of sphingolipids as promising targets for the development of new therapeutic options.


Subject(s)
Biofilms/growth & development , Scedosporium/metabolism , Sphingolipids/biosynthesis , Cell Membrane/metabolism , Fatty Acids, Monounsaturated/metabolism , Meperidine/analogs & derivatives , Meperidine/metabolism
5.
Commun Biol ; 2: 59, 2019.
Article in English | MEDLINE | ID: mdl-30775460

ABSTRACT

Bacterial pore-forming toxin aerolysin-like proteins (ALPs) are widely distributed in animals and plants. However, functional studies on these ALPs remain in their infancy. ßγ-CAT is the first example of a secreted pore-forming protein that functions to modulate the endolysosome pathway via endocytosis and pore formation on endolysosomes. However, the specific cell surface molecules mediating the action of ßγ-CAT remain elusive. Here, the actions of ßγ-CAT were largely attenuated by either addition or elimination of acidic glycosphingolipids (AGSLs). Further study revealed that the ALP and trefoil factor (TFF) subunits of ßγ-CAT bind to gangliosides and sulfatides, respectively. Additionally, disruption of lipid rafts largely impaired the actions of ßγ-CAT. Finally, the ability of ßγ-CAT to clear pathogens was attenuated in AGSL-eliminated frogs. These findings revealed a previously unknown double binding pattern of an animal-secreted ALP in complex with TFF that initiates ALP-induced endolysosomal pathway regulation, ultimately leading to effective antimicrobial responses.


Subject(s)
Acidic Glycosphingolipids/chemistry , Amphibian Proteins/immunology , Bacterial Toxins/immunology , Gram-Negative Bacterial Infections/immunology , Lysosomes/immunology , Multiprotein Complexes/immunology , Pore Forming Cytotoxic Proteins/immunology , Trefoil Factor-3/immunology , Acidic Glycosphingolipids/antagonists & inhibitors , Acidic Glycosphingolipids/biosynthesis , Aeromonas hydrophila/growth & development , Aeromonas hydrophila/pathogenicity , Amphibian Proteins/genetics , Amphibian Proteins/metabolism , Animals , Anura , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Ceramides/antagonists & inhibitors , Ceramides/biosynthesis , Ceramides/chemistry , Cerebrosides/antagonists & inhibitors , Cerebrosides/biosynthesis , Cerebrosides/chemistry , Gangliosides/antagonists & inhibitors , Gangliosides/biosynthesis , Gangliosides/chemistry , Gene Expression , Gram-Negative Bacterial Infections/genetics , Gram-Negative Bacterial Infections/microbiology , Humans , Interleukin-1beta/biosynthesis , Lysosomes/drug effects , Lysosomes/microbiology , Membrane Microdomains/drug effects , Membrane Microdomains/immunology , Membrane Microdomains/microbiology , Meperidine/analogs & derivatives , Meperidine/pharmacology , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Sphingosine/antagonists & inhibitors , Sphingosine/biosynthesis , Sphingosine/chemistry , THP-1 Cells , Trefoil Factor-3/genetics , Trefoil Factor-3/metabolism
6.
Pharmazie ; 73(6): 324-328, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29880084

ABSTRACT

Oral fluid assays for quantifying drugs are useful in forensic toxicology and drug monitoring. Compared with blood and urine specimens, oral fluid collection is simple, non-invasive, and more difficult to adulterate. Therefore, we investigated whether meperidine and its metabolites could be detected in oral fluid and whether there was a predictable relationship between oral fluid and plasma concentrations. Male New Zealand white rabbits (n = 10) were administered meperidine hydrochloride (20 mg/kg, intravenous). Then, plasma and oral fluid were collected at various time points up to 10 h after administration. We developed a simple and sensitive gas chromatography-mass spectrometry method for the determination of meperidine and normeperidine in oral fluid and plasma. We estimated the apparent pharmacokinetic parameters for meperidine in oral fluid and plasma and determined the ratio and correlation between oral fluid and plasma concentrations. The results demonstrate that this method has excellent specificity, linearity, precision, and recovery. Meperidine and normeperidine were detected in both body fluids; meperidine was the most abundant analyte in oral fluid. The oral fluid-to-plasma drug concentration ratios did not differ significantly over time (p > 0.05). In addition, oral fluid and plasma levels of meperidine and normeperidine were significantly correlated over time (r = 0.713 and 0.725, respectively; p < 0.05). These results provide context for interpreting meperidine and metabolite concentrations in oral fluid and support the utility of oral fluid as an alternative matrix in clinical and forensic testing.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Gas Chromatography-Mass Spectrometry/methods , Meperidine/analogs & derivatives , Meperidine/pharmacokinetics , Administration, Intravenous , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/analysis , Animals , Drug Monitoring/methods , Male , Meperidine/administration & dosage , Meperidine/analysis , Rabbits , Reproducibility of Results , Time Factors
8.
Rapid Commun Mass Spectrom ; 31(18): 1519-1533, 2017 Sep 30.
Article in English | MEDLINE | ID: mdl-28686794

ABSTRACT

RATIONALE: Several opioid analgesics have been related to the prolongation of cardiac repolarization, a condition which can be fatal. In order to establish a correct estimation of the risk/benefit balance of therapeutic doses of meperidine, normeperidine, tramadol, propoxyphene and norpropoxyphene, it was necessary to develop an analytical method to determinate plasma concentrations of these opioids. METHODS: Here we describe a method which incorporates strong alkaline treatment to obtain norpropoxyphene amide followed by a one-elution step solid-phase extraction, and without further derivatization. Separation and quantification were achieved by gas chromatography/electron ionization mass spectrometry (GC/EI-MS) in selected-ion monitoring mode. Quantification was performed with 500 µL of plasma by the addition of deuterated analogues as internal standards. RESULTS: The proposed method has been validated in the linearity range of 25-1000 ng/mL for all the analytes, with correlation coefficients higher than 0.990. The lower limit of quantification was 25 ng/mL. The intra- and inter-day precision, calculated in terms of relative standard deviation, were 2.0-12.0% and 6.0-15.0%, respectively. The accuracy, in terms of relative error, was within a ± 10% interval. The absolute recovery and extraction efficiency ranged from 81.0 to 111.0% and 81.0 to 105.0%, respectively. CONCLUSIONS: A GC/MS method for the rapid and simultaneous determination of meperidine, normeperidine, tramadol, propoxyphene and norpropoxyphene in human plasma was developed, optimized and validated. This procedure was shown to be sensitive and specific using small specimen amounts, suitable for application in routine analysis for forensic purposes and therapeutic monitoring. To our knowledge, this is the first full validation of the simultaneous determination of these opioids and their metabolites in plasma samples.


Subject(s)
Analgesics, Opioid/blood , Dextropropoxyphene/analogs & derivatives , Dextropropoxyphene/blood , Gas Chromatography-Mass Spectrometry/methods , Meperidine/analogs & derivatives , Meperidine/blood , Solid Phase Extraction/methods , Tramadol/blood , Analgesics, Opioid/adverse effects , Analgesics, Opioid/isolation & purification , Dextropropoxyphene/adverse effects , Dextropropoxyphene/isolation & purification , Drug Monitoring , Heart/drug effects , Humans , Meperidine/adverse effects , Meperidine/isolation & purification , Tramadol/adverse effects , Tramadol/isolation & purification
9.
Biochem Biophys Res Commun ; 486(2): 551-557, 2017 04 29.
Article in English | MEDLINE | ID: mdl-28322796

ABSTRACT

Taxoids are anti-cancer drugs frequently used to treat solid tumors, but they are sometimes ineffective and tumors may become resistant to their action. Here, we examined the involvement of sphingolipid metabolic enzymes in paclitaxel (PTX) resistance using a human prostate cancer cell line, PC3, and its PTX-resistant subline, PC3-PR. PTX (20 nM) suppressed cell proliferation and increased various ceramide species in PC3, but not PC3-PR, cells. PC3-PR contained higher S1P levels than did PC3, regardless of PTX treatment. Western blotting revealed that PC3-PR cells expressed higher levels of sphingosine kinase 1 (SPHK1) and glucosylceramide synthase (GCS) but lower levels of acid sphingomyelinase (ASMase) and neutral sphingomyelinase 2 than did PC3 cells. Inhibition of SPHK1 using siRNA or a pharmacological inhibitor decreased S1P levels in PC3-PR cells and inhibited proliferation in the presence or absence of PTX, suggesting that SPHK1 is at least partially responsible for PTX resistance. Similarly, GCS inhibitors (PDMP and PPMP) increased cellular ceramides and suppressed the proliferation of PC3-PR. However, inhibition of proteasome function or histone deacetylase activity increased SMase and ceramide levels and suppressed PC3-PR proliferation. These results suggest that modulation of metabolic enzyme expression and alteration of the sphingolipid rheostat protects cancer cells against PTX.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Resistance, Neoplasm/genetics , Epithelial Cells/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Paclitaxel/pharmacology , Sphingolipids/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , K562 Cells , Male , Meperidine/analogs & derivatives , Meperidine/pharmacology , Morpholines/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/metabolism
10.
Int J Clin Pharmacol Ther ; 55(3): 275-285, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27509828

ABSTRACT

A prolongation of the QTc-interval has been described for several opioids, including pethidine (meperidine). OBJECTIVE: To evaluate in the clinical setting the frequency and risk factors associated with the QT-interval prolongation induced by meperidine. RESEARCH DESIGN AND METHODS: We recruited patients requiring meperidine administration and recorded their medical history and comorbidities predisposing to QT-interval prolongation. Ionograms and electrocardiograms (ECGs) were performed at baseline and during treatment; QT was corrected using the Bazzet, Fridericia, Framinghan, and Hogdes formulas. We measured meperidine and normeperidine by gas chromatography. Values are expressed as mean ± SD (range). RESULTS: 58 patients were studied (43.1% males). All patients received meperidine at a dose of 304 ± 133 (120 - 480) mg/day. Meperidine and normeperidine concentrations were 369 ± 60 (265 - 519) and 49 ± 17 (15 - 78) ng/mL, respectively. Intratreatment control found QTcB 370 ± 30 (305 - 433), QTcFri 353 ± 35 (281 - 429), QTcFra 360 ± 30 (299 - 429), QTcH 359 ± 27 (304 - 427), ΔQTcB +9 ± 42 (-90 to +136), ΔQTcFri +4 ± 45 (-86 to +137), ΔQTcFra +5 ± 40 (-77 to +129), and ΔQTcH +7 ± 40 (-76 to +129) ms. Meperidine concentration correlated with QTc-interval (R > 0.36) and ΔQTc (R > 0.69) but the correlation was even better for normeperidine concentration, QTc (R > 0.52) and ΔQTc (R > 0.81). Depending on the QTc correction formula used, 13 - 15 patients (22.41 - 25.86%) presented ΔQTc values > 30 ms, and 7 - 8 patients (12.07- 13.79%) showed ΔQTc values > 60 ms. Renal failure was associated with risk for ΔQTc > 30 ms of 3.74 (IC95% 1.73 - 8.10) and for ΔQTc > 60 ms of 4.27 (IC 95% 1.26 - 14.48). No patient developed arrhythmias during the study. CONCLUSIONS: Meperidine treatment causes ECG changes (QTc-interval prolongation) in high correlation with normeperidine plasma concentration. Renal failure increases the risk.
.


Subject(s)
Analgesics, Opioid/adverse effects , Heart Conduction System/drug effects , Long QT Syndrome/chemically induced , Meperidine/adverse effects , Action Potentials/drug effects , Aged , Aged, 80 and over , Analgesics, Opioid/blood , Analgesics, Opioid/pharmacokinetics , Argentina/epidemiology , Biotransformation , Electrocardiography , Female , Heart Conduction System/physiopathology , Heart Rate/drug effects , Humans , Long QT Syndrome/diagnosis , Long QT Syndrome/epidemiology , Long QT Syndrome/physiopathology , Longitudinal Studies , Male , Meperidine/analogs & derivatives , Meperidine/blood , Meperidine/pharmacokinetics , Middle Aged , Prevalence , Prospective Studies , Renal Insufficiency/epidemiology , Risk Assessment , Risk Factors
12.
J Anal Toxicol ; 40(8): 694-699, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27562966

ABSTRACT

Continuing our previous studies analyzing drugs of abuse in municipal wastewater, a method was developed for the analysis of miscellaneous drugs of abuse in wastewater samples using liquid chromatography coupled with tandem mass spectrometry (LC-MS-MS). Eight drugs and metabolites were analyzed including 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrolidine (EDDP), fentanyl, norfentanyl, meperidine, normeperidine, methadone, phencyclidine and tramadol. These drugs were chosen because of their widespread abuse. Wastewater samples were collected at both the Oxford Waste Water Treatment Plant in Oxford, Mississippi (MS) and the University Wastewater Treatment Plant in University, MS. These wastewater samples were collected on weekends in which the University of Mississippi football team (colloquially the "Ole Miss Rebels" football team) held home games (Vaught-Hemingway Stadium, University, MS 38677). The collected samples were analyzed using a validated method and found to contain tramadol in 25 samples at quantifiable levels. EDDP, meperidine, normeperidine and methadone were also detected but were under the limit of quantitation.


Subject(s)
Chromatography, Liquid , Drug Residues/analysis , Substance Abuse Detection/methods , Tandem Mass Spectrometry , Wastewater/analysis , Chromatography, High Pressure Liquid , Fentanyl/analogs & derivatives , Fentanyl/analysis , Football , Humans , Illicit Drugs/analysis , Limit of Detection , Meperidine/analogs & derivatives , Meperidine/analysis , Methadone/analysis , Mississippi , Phencyclidine/analysis , Pyrrolidines/analysis , Reproducibility of Results , Tramadol/analysis
13.
Oncotarget ; 7(21): 30977-89, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27129160

ABSTRACT

Esophageal cancer is one of the least studied and deadliest cancers worldwide with a poor prognosis due to limited options for treatment. Chemotherapy agents such as the microtubule-targeting compounds are the mainstay of palliation for advanced esophageal cancer treatment. However, the toxicity and side effects of tubulin-binding agents (TBAs) have promoted the development of novel, more potent but less toxic TBAs. Herein, we identified 2-[4-(3,4-dimethoxyphenyl)-3-methyl-1H-pyrazol-5-yl]-5-[(2-methylprop-2-en-1-yl)oxy] phenol (PPMP) as a novel TBA for esophageal cancer treatment. PPMP markedly inhibited tubulin polymerization, and decreased viability and anchorage-independent growth of esophageal cancer cell lines, effects that were accompanied by G2/M arrest and apoptosis. Importantly, we produced patient-derived esophageal cancer xenografts to evaluate the therapeutic effect of PPMP in a setting that best mimics the clinical context in patients with esophageal cancer. Overall, we identified PPMP as a novel microtubule-destabilizing compound and as a new therapeutic agent against esophageal carcinoma.


Subject(s)
Esophageal Neoplasms/drug therapy , Meperidine/analogs & derivatives , Tubulin Modulators/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , M Phase Cell Cycle Checkpoints/drug effects , Male , Meperidine/pharmacology , Mice , Middle Aged , Models, Molecular , Tubulin/metabolism , Xenograft Model Antitumor Assays
14.
Bioorg Med Chem Lett ; 25(22): 5032-5, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26494260

ABSTRACT

A series of pethidine analogs were synthesized and their affinities for the [(3)H]N-methyl-scopolamine (NMS) binding site on muscarinic acetylcholine receptors (mAChRs) were determined using M1, M3 or M5 human mAChRs expressed by Chinese hamster ovary (CHO) cell membranes. Compound 6b showed the highest binding affinities at M1, M3 and M5 mAChRs (Ki=0.67, 0.37, and 0.38 µM, respectively).


Subject(s)
Meperidine/analogs & derivatives , Meperidine/chemical synthesis , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M3/metabolism , Receptor, Muscarinic M5/metabolism , Animals , CHO Cells , Cricetulus , Female , Humans , Ligands , Meperidine/metabolism , Structure-Activity Relationship
15.
Ugeskr Laeger ; 177(22): V12140740, 2015 May 25.
Article in Danish | MEDLINE | ID: mdl-26027667

ABSTRACT

Calcinosis cutis is a rare disease entity characterized by deposits of calcium in the skin and subcutaneous tissue causing hard-to-heal ulcers. This is a case report on a patient with femoral ulcers in connection with densely mineralized skin caused by ketobemidon injections. Next to surgical excision of calcified tissue the patient received extracorporeal shockwave therapy (ESWT). On the basis of excellent healing, partial skin transplant was feasible. We advocate for randomized trials on ESWT as an adjunctive therapy for complex non-healing wounds.


Subject(s)
Calcinosis/therapy , High-Energy Shock Waves/therapeutic use , Skin Ulcer/therapy , Aged , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Calcinosis/chemically induced , Calcinosis/pathology , Female , Humans , Meperidine/administration & dosage , Meperidine/adverse effects , Meperidine/analogs & derivatives , Skin Ulcer/chemically induced , Skin Ulcer/pathology , Thigh/pathology
16.
Exp Cell Res ; 336(1): 23-32, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26004871

ABSTRACT

BACKGROUND: Acquired resistance to cisplatin treatment is a caveat when treating patients with non-small cell lung cancer (NSCLC) and malignant pleural mesothelioma (MPM). Ceramide increases in response to chemotherapy, leading to proliferation arrest and apoptosis. However, a tumour stress activation of glucosylceramide synthase (GCS) follows to eliminate ceramide by formation of glycosphingolipids (GSLs) such as globotriaosylceramide (Gb3), the functional receptor of verotoxin-1. Ceramide elimination enhances cell proliferation and apoptosis blockade, thus stimulating tumor progression. GSLs transactivate multidrug resistance 1/P-glycoprotein (MDR1) and multidrug resistance-associated protein 1 (MRP1) expression which further prevents ceramide accumulation and stimulates drug efflux. We investigated the expression of Gb3, MDR1 and MRP1 in NSCLC and MPM cells with acquired cisplatin resistance, and if GCS activity or MDR1 pump inhibitors would reduce their expression and reverse cisplatin-resistance. METHODS: Cell surface expression of Gb3, MDR1 and MRP1 and intracellular expression of MDR1 and MRP1 was analyzed by flow cytometry and confocal microscopy on P31 MPM and H1299 NSCLC cells and subline cells with acquired cisplatin resistance. The effect of GCS inhibitor PPMP and MDR1 pump inhibitor cyclosporin A for 72h on expression and cisplatin cytotoxicity was tested. RESULTS: The cisplatin-resistant cells expressed increased cell surface Gb3. Cell surface Gb3 expression of resistant cells was annihilated by PPMP whereas cyclosporin A decreased Gb3 and MDR1 expression in H1299 cells. No decrease of MDR1 by PPMP was noted in using flow cytometry, whereas a decrease of MDR1 in H1299 and H1299res was indicated with confocal microscopy. No certain co-localization of Gb3 and MDR1 was noted. PPMP, but not cyclosporin A, potentiated cisplatin cytotoxicity in all cells. CONCLUSIONS: Cell surface Gb3 expression is a likely tumour biomarker for acquired cisplatin resistance of NSCLC and MPM cells. Tumour cell resistance to MDR1 inhibitors of cell surface MDR1 and Gb3 could explain the aggressiveness of NSCLC and MPM. Therapy with GCS activity inhibitors or toxin targeting of the Gb3 receptor may substantially reduce acquired cisplatin drug resistance of NSCLC and MPM cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Cyclosporine/pharmacology , Drug Resistance, Neoplasm/drug effects , Glucosyltransferases/antagonists & inhibitors , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Trihexosylceramides/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Apoptosis/drug effects , Blotting, Western , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Ceramides/metabolism , Drug Resistance, Multiple/drug effects , Flow Cytometry , Humans , Immunosuppressive Agents/pharmacology , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Meperidine/analogs & derivatives , Meperidine/pharmacology , Mesothelioma/enzymology , Mesothelioma/pathology , Mesothelioma, Malignant , Microscopy, Confocal , Multidrug Resistance-Associated Proteins/metabolism , Tumor Cells, Cultured
17.
Glycobiology ; 25(4): 351-6, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25715344

ABSTRACT

Glucosylceramide synthase (GCS) catalyzes the first committed step in the biosynthesis of glucosylceramide (GlcCer)-related glycosphingolipids (GSLs). Although inhibitors of GCS, PPMP and PDMP have been widely used to elucidate their biological function and relevance, our comprehensive literature review revealed that the available data are ambiguous. We therefore investigated whether and to what extent GCS inhibitors affect the expression of lactosylceramide (LacCer), neolacto (nLc4 and P1), ganglio (GM1 and GD3) and globo (Gb3 and SSEA3) series GSLs in a panel of human cancer cell lines using flow cytometry, a commonly applied method investigating cell-surface GSLs after GCS inhibition. Their cell-surface GSL expression considerably varied among cell lines and more importantly, sublethal concentrations (IC10) of both inhibitors preferentially and significantly reduced the expression of Gb3 in the cancer cell lines IGROV1, BG1, HT29 and T47D, whereas SSEA3 was only reduced in BG1. Unexpectedly, the neolacto and ganglio series was not affected. LacCer, the precursor of all GlcCer-related GSL, was significantly reduced only in BG1 cells treated with PPMP. Future research questions addressing particular GSLs require careful consideration; our results indicate that the extent to which there is a decrease in the expression of one or more particular GSLs is dependent on the cell line under investigation, the type of GCS inhibitor and exposure duration.


Subject(s)
Enzyme Inhibitors/pharmacology , Glucosyltransferases/antagonists & inhibitors , Glycosphingolipids/biosynthesis , Meperidine/analogs & derivatives , Morpholines/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Glucosyltransferases/metabolism , Humans , Meperidine/pharmacology
18.
J Neurochem ; 129(5): 884-94, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24494600

ABSTRACT

Aggregate-prone mutant proteins, such as α-synuclein and huntingtin, play a prominent role in the pathogenesis of various neurodegenerative disorders; thus, it has been hypothesized that reducing the aggregate-prone proteins may be a beneficial therapeutic strategy for these neurodegenerative disorders. Here, we identified two previously described glucosylceramide (GlcCer) synthase inhibitors, DL-threo-1-Phenyl-2-palmitoylamino-3-morpholino-1-propanol and Genz-123346(Genz), as enhancers of autophagy flux. We also demonstrate that GlcCer synthase inhibitors exert their effects on autophagy by inhibiting AKT-mammalian target of rapamycin (mTOR) signaling. More importantly, siRNA knock down of GlcCer synthase had the similar effect as pharmacological inhibition, confirming the on-target effect. In addition, we discovered that inhibition of GlcCer synthase increased the number and size of lysosomal/late endosomal structures. Although inhibition of GlcCer synthase decreases levels of mutant α-synuclein in neurons, it does so, according to our data, through autophagy-independent mechanisms. Our findings demonstrate a direct link between glycosphingolipid biosynthesis and autophagy in primary neurons, which may represent a novel pathway with potential therapeutic value for the treatment of Parkinson's disease. Inhibition of GlcCer synthase enhances autophagy by inhibiting AKT-mTOR signaling, and increases the number and size of lysosomal/late endosomal structures. Furthermore, inhibition of GlcCer synthase decreased levels of mutant α-synuclein in neurons, which may represent a potential therapeutic target for Parkinson's disease.


Subject(s)
Autophagy/physiology , Enzyme Inhibitors/pharmacology , Glucosyltransferases/antagonists & inhibitors , Neurons/physiology , Animals , Blotting, Western , Cells, Cultured , Dioxanes/pharmacology , Female , Glycosphingolipids/biosynthesis , HEK293 Cells , Humans , Male , Meperidine/analogs & derivatives , Meperidine/pharmacology , Mice , Mice, Knockout , Mice, Transgenic , Oncogene Protein v-akt/metabolism , Parkinson Disease/genetics , Phosphorylation , Primary Cell Culture , Pyrrolidines/pharmacology , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction
19.
Br J Anaesth ; 112(2): 328-36, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24185607

ABSTRACT

BACKGROUND: Local anaesthetics (LA) injected intraperitoneally have been found to decrease postoperative pain. This double-blind randomized study was performed comparing continuous infusion or patient-controlled intraperitoneal (i.p.) bolus injection of LA. The primary endpoint was supplemental opioid consumption during the first 24 postoperative hours. METHODS: Two multi-hole catheters were placed intraperitoneally at the end of the surgery in 40 patients undergoing elective abdominal hysterectomy. The patients were randomized into two groups: Group P: patients self-injected 10 ml of levobupivacaine 1.25 mg ml(-1) via the i.p. catheter as needed, maximum once per hour, and had continuous saline infusion 10 ml h(-1) into the second catheter. Group C: patients received a continuous infusion of 10 ml h(-1) of levobupivacaine 1.25 mg ml(-1) intraperitoneally through one catheter and 10 ml saline as bolus as needed via the other. Ketobemidone was administered intravenously as rescue medication. RESULTS: Total ketobemidone consumption during 0-24 h was lower in Group P compared with Group C (mean 23.1 vs 35.7 mg, P=0.04). No differences in the median pain scores were found between the groups. Earlier return of gastrointestinal (GI) function was found in Group P vs Group C (mean 1.5 vs 2.2 days, P<0.01), which also resulted in earlier home-readiness (mean 1.9 vs 2.7 days, P=0.04). CONCLUSIONS: A statistically significant opioid-sparing effect was found when patient-controlled levobupivacaine was administered intraperitoneally as needed compared with continuous infusion. This was associated with a faster return of GI function and home-readiness. There was, however, a wide confidence interval in the primary endpoint, opioid consumption.


Subject(s)
Analgesia, Patient-Controlled/methods , Anesthetics, Local/therapeutic use , Bupivacaine/analogs & derivatives , Hysterectomy , Pain, Postoperative/drug therapy , Adult , Aged , Analgesics, Opioid/administration & dosage , Anesthetics, Local/administration & dosage , Bupivacaine/administration & dosage , Bupivacaine/therapeutic use , Double-Blind Method , Female , Humans , Injections, Intraperitoneal , Levobupivacaine , Meperidine/administration & dosage , Meperidine/analogs & derivatives , Middle Aged , Pain Measurement/methods , Treatment Outcome
20.
J Pain Symptom Manage ; 47(4): 801-5, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23870842

ABSTRACT

CONTEXT: Normeperidine accumulates in patients with impaired renal function and may cause central neurotoxicity. However, some uremic patients still undergo meperidine treatment for chronic pain. OBJECTIVES: To prevent normeperidine side effects and complications, we investigated the clearance rate and extraction ratio of meperidine and normeperidine in hemodialysis patients with chronic pain. METHODS: Three hemodialysis patients, with diagnoses of chronic pancreatitis, chronic back pain, and intractable intra-abdominal pain, received long-term (more than six months) administration of meperidine for chronic noncancer pain. During regular hemodialysis, 72 blood samples in total were collected from the afferent port, efferent port, and ultradiafiltrate port at eight time points. The plasma concentrations of meperidine and normeperidine were determined by high-performance liquid chromatography. RESULTS: The prehemodialysis plasma concentrations of meperidine and normeperidine were 2963 ± 315 and 2369 ± 1974 ng/mL, which declined to 591 ± 109 and 853 ± 765 ng/mL, with 80% and 65% reduction, respectively. The plasma clearance and extraction ratios of meperidine were 22.7 ± 9.8 mL/minute and 10.1 ± 5.6% and for normeperidine 26.0 ± 11.4 mL/minute and 10.8 ± 2.5%, respectively. CONCLUSION: Hemodialysis can efficiently remove meperidine and its active metabolite, normeperidine, in uremic patients receiving long-term meperidine therapy for chronic noncancer pain.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Chronic Pain/blood , Meperidine/analogs & derivatives , Meperidine/pharmacokinetics , Renal Dialysis , Abdominal Pain/blood , Abdominal Pain/drug therapy , Adult , Back Pain/blood , Back Pain/drug therapy , Blood Chemical Analysis , Chromatography, High Pressure Liquid , Chronic Pain/drug therapy , Female , Humans , Male , Metabolic Clearance Rate , Middle Aged , Pancreatitis, Chronic/therapy , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL