Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters

Publication year range
1.
Ther Drug Monit ; 46(1): 1-5, 2024 02 01.
Article in English | MEDLINE | ID: mdl-37941108

ABSTRACT

ABSTRACT: Pharmacobezoars develop after an acute overdose or during routine drug administration. Here, the authors present a case of fatal multidrug overdose involving a 62-year-old woman. Her usual treatment included tramadol extended-release, citalopram, and mirtazapine. Furthermore, she self-medicated and misused her husband's medications. The autopsy revealed the presence of a voluminous medication bezoar in the stomach. No mechanical complication was noted. Toxicologic analyses were performed using gas chromatography with flame ionization detection, liquid chromatography with diode array detection, gas chromatography with mass spectrometry detection, and liquid chromatography coupled to tandem mass spectrometry. Tramadol (34,000 mcg/L), O-desmethyltramadol (2200 mcg/L), propranolol (6000 mcg/L), bromazepam (2500 mcg/L), zopiclone (1200 mcg/L), and citalopram (700 mcg/L) were identified in femoral blood at toxic concentrations. Interestingly, the femoral blood and vitreous humor concentration ratio was approximately 0.7. Furthermore, an English exhaustive literature search was performed using several different electronic databases without any limiting period to identify published pharmacobezoar-related fatalities. Seventeen publications were identified reporting a total of 19 cases. Decedents' mean age was 47.6 years [0.8-79] and a clear female predominance emerged. Several drugs were involved in pharmacobezoar formation. Death was attributed to drug toxicity in 13 cases, and to mechanical complications and/or sepsis in 4 cases. A mixed cause of death was reported in 2 cases. Although rare, pharmacobezoars remain potentially lethal and raise challenges in therapeutic management.


Subject(s)
Citalopram , Drug Overdose , Tramadol , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , Infant , Male , Middle Aged , Young Adult , Citalopram/toxicity , Drug Overdose/mortality , Gas Chromatography-Mass Spectrometry , Stomach , Tramadol/toxicity
2.
Neurochem Res ; 48(5): 1320-1333, 2023 May.
Article in English | MEDLINE | ID: mdl-36449200

ABSTRACT

Boldenone and tramadol are abused among large sectors of adolescents. Therefore, the behavioral changes concerned with memory and cognitive functions and neurochemical variations were investigated in the cortex of rats treated with boldenone and/or tramadol. Rats were divided into control and rats treated with boldenone, tramadol, or both drugs. At the end of the treatment period, the memory and cognitive functions were evaluated by the Y-maze test (YMT) and elevated plus maze test (EPMT) and the motor activity was determined by the open field test (OFT). The cortex was dissected to carry out the neurochemical analyses. Rats treated with boldenone and/or tramadol showed impaired memory and cognitive functions and reduced motor activity. A significant increase in lipid peroxidation (MDA), nitric oxide (NO), and a significant decrease in reduced glutathione (GSH) were observed in the cortex of rats treated with boldenone and/or tramadol. The levels of acetylcholinesterase (AChE) and monoamine oxidase (MAO) decreased significantly. Western blot data showed a significant decrease in Bcl2 and a significant increase in caspase-3 and inducible nitric oxide synthase (iNOS) in rats treated with boldenone and/or tramadol. These changes were associated with neuronal death as indicated from the histopathological examination.The present findings indicate that boldenone and/or tramadol induced impairment in memory and cognitive functions. These changes could be mediated by the increase in oxidative stress, neuroinflammation, reduced AChE level, and reduced number of survived neurons in the cortex as indicated from the decreased Bcl2 level and the histological examination.


Subject(s)
Tramadol , Rats , Male , Animals , Tramadol/toxicity , Acetylcholinesterase/metabolism , Testosterone , Proto-Oncogene Proteins c-bcl-2 , Oxidative Stress
3.
Biomarkers ; 28(4): 401-408, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36932833

ABSTRACT

INTRODUCTION: The risk of cardiotoxicity is associated with the use of anabolic-androgenic steroids and analgesics, several deaths were attributed to such medications. OBJECTIVES: This study investigates the effects of boldenone (BOLD) and tramadol (TRAM) alone or in combination on the heart. MATERIAL AND METHODS: Forty adult male rats were divided into four groups. Normal control group, BOLD (5 mg/kg, i.m.) per week, tramadol Hcl (TRAM) (20 mg/kg, i.p.) daily and a combination of BOLD (5 mg/kg) and TRAM (20 mg/kg), respectively for two months. Serum and cardiac tissue were extracted for determination of serum, aspartate aminotransferase (AST), creatine phosphokinase (CPK) and lipid profiles, tissue malondialdehyde (MDA), reduced glutathione (GSH), superoxide dismutase (SOD), nitric oxide (NO), tumour necrosis factor alpha (TNF-α), interleukin-6 (IL-6) and histopathological examination. Troponin I gene expression was quantified in cardiac tissue using real-time polymerase chain reaction technique. RESULTS: Groups received BOLD and TRAM alone and in combination showed elevated serum biochemical parameters (AST, CPK) and deviations in lipid profiles, elevation in oxidative and inflammatory parameters (MDA, NO, TNF-α and IL-6), and decrease in GSH and SOD, up-regulated cardiac troponin I as well as distorted cardiac histopathological pictures. CONCLUSION: The current study elucidated the risk of administration of these drugs for sustained periods as well as the marked detrimental effects of using these drugs in combination.


Subject(s)
Myocardium , Tramadol , Rats , Male , Animals , Myocardium/metabolism , Troponin I/genetics , Troponin I/metabolism , Tramadol/toxicity , Tramadol/metabolism , Cytokines/genetics , Cytokines/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , Doxorubicin , Oxidative Stress , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
4.
Toxicol Mech Methods ; 33(7): 607-623, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37051630

ABSTRACT

Tramadol (TRA) causes neurotoxicity whereas trimetazidine (TMZ) is neuroprotective. The potential involvement of the PI3K/Akt/mTOR signaling pathway in the neuroprotection of TMZ against TRA-induced neurotoxicity was evaluated. Seventy male Wistar rats were divided into groups. Groups 1 and 2 received saline or TRA (50 mg/kg). Groups 3, 4, and 5 received TRA (50 mg/kg) and TMZ (40, 80, or 160 mg/kg) for 14 days. Group 6 received TMZ (160 mg/kg). Hippocampal neurodegenerative, mitochondrial quadruple complex enzymes, phosphatidylinositol-3-kinases (PI3Ks)/protein kinase B levels, oxidative stress, inflammatory, apoptosis, autophagy, and histopathology were evaluated. TMZ decreased anxiety and depressive-like behavior induced by TRA. TMZ in tramadol-treated animals inhibited lipid peroxidation, GSSG, TNF-α, and IL-1ß while increasing GSH, SOD, GPx, GR, and mitochondrial quadruple complex enzymes in the hippocampus. TRA inhibited Glial fibrillary acidic protein expression and increased pyruvate dehydrogenase levels. TMZ reduced these changes. TRA decreased the level of JNK and increased Beclin-1 and Bax. TMZ decreased phosphorylated Bcl-2 while increasing the unphosphorylated form in tramadol-treated rats. TMZ activated phosphorylated PI3Ks, Akt, and mTOR proteins. TMZ inhibited tramadol-induced neurotoxicity by modulating the PI3K/Akt/mTOR signaling pathways and its downstream inflammatory, apoptosis, and autophagy-related cascades.


Subject(s)
Neuroprotective Agents , Neurotoxicity Syndromes , Tramadol , Trimetazidine , Male , Rats , Animals , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/pharmacology , Trimetazidine/pharmacology , Tramadol/toxicity , Neuroprotection , Rats, Wistar , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/pharmacology , Apoptosis , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Autophagy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
5.
BMC Endocr Disord ; 21(1): 180, 2021 Sep 06.
Article in English | MEDLINE | ID: mdl-34488743

ABSTRACT

BACKGROUND: Tramadol is a synthetic opioid and poisoning is increasing around the world day by day. Various treatments are applied for tramadol poisoning. Due to the unknown effects of tramadol poisoning and some of its treatments on blood glucose levels, this study was conducted to investigate the overdose of tramadol and its common treatments (naloxone, diazepam), and their combination on blood glucose levels in male rats. METHODS: This study was conducted in 45 male Wistar rats. The animals were randomly divided into five groups of 9. They received a 75 mg/kg dose of tramadol alone with naloxone, diazepam, and a combination of both of these two drugs. On the last day, animals' tail vein blood glucose levels (BGL) were measured using a glucometer at different times, including before the tramadol injection (baseline) and 1 hour, 3 hours, and 6 hours after wards. The rats were anesthetized and sacrificed 24 h after the last injection. Blood samples were then taken, and the serum obtained was used to verify the fasting glucose concentration. Data were analyzed using SPSS software at a significance level of 0.05 using a one-way analysis of variance (ANOVA) and a generalized estimating equation (GEE). RESULTS: According to the GEE model results, the diazepam-tramadol and naloxone-diazepam-tramadol groups showed blood glucose levels five units higher than the tramadol group (p < 0.05). The diazepam-tramadol group had significantly higher blood glucose levels than the naloxone-tramadol group (p < 0.05). The mean blood glucose levels before the intervention, 3 hours and 6 hours after the injection of tramadol did not differ between the groups, but the blood glucose levels 1 hour after the injection of tramadol in the group of naloxone-tramadol were significantly lower than in the control group (p < 0.05). Blood glucose levels did not differ between the groups 24 h after injection of tramadol. CONCLUSION: The results of the present study showed tramadol overdose does not affect blood glucose levels. The diazepam-tramadol combination and the diazepam-naloxone-tramadol combination caused an increase in blood glucose levels.


Subject(s)
Blood Glucose/metabolism , Diazepam/pharmacology , Drug Overdose/complications , Hyperglycemia/pathology , Naloxone/pharmacology , Tramadol/toxicity , Analgesics, Opioid/toxicity , Animals , Blood Glucose/drug effects , Hyperglycemia/chemically induced , Hyperglycemia/metabolism , Hypnotics and Sedatives/pharmacology , Male , Narcotic Antagonists/pharmacology , Rats , Rats, Wistar , Tramadol/administration & dosage
6.
Xenobiotica ; 51(9): 1029-1037, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34319855

ABSTRACT

1. The aim of this study was to compare the in vitro cytotoxic effect of tramadol and M1 metabolite in HepG2 cell line, the underlying mechanism, and PI3K/AKT/mTOR as potential target.2. Concentrations representing therapeutic level for tramadol (2 µM) and M1 metabolite (0.5 µM) were used. In addition, other increasing concentrations representing higher toxic levels were used (6, 10 µM for tramadol and 1.5, 2.5 µM for M1 metabolites). Cytotoxicity was assessed at 24, 48 and 72 h.3. Both tramadol and M1 metabolites were able to produce cytotoxicity in a dose and time dependent manner. Insignificant difference was detected between cells exposed to tramadol and M1 metabolite at therapeutic concentrations. Tramadol-induced apoptotic and autophagic cell death while M1 metabolite-induced apoptosis only. For PI3K/AKT/mTOR pathway, the therapeutic concentration of tramadol was only able to increase phosphorylation of AKT while higher toxic concentrations were able to increase phosphorylation of whole pathway; Meanwhile, M1 metabolite was able to increase the phosphorylation of the whole pathway significantly in therapeutic and toxic concentrations.4. In conclusion, both tramadol and M1 are equally cytotoxic. Apoptosis and autophagy both mediate hepatic cell death. PI3K/AKT pathway is involved in apoptosis induction while autophagy is regulated through mTOR independent pathway.


Subject(s)
Proto-Oncogene Proteins c-akt , Tramadol , Hep G2 Cells , Humans , Phosphatidylinositol 3-Kinases , TOR Serine-Threonine Kinases , Tramadol/analogs & derivatives , Tramadol/toxicity
7.
Arch Toxicol ; 95(12): 3681-3693, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34604914

ABSTRACT

New synthetic opioids (NSOs) pose a public health concern since their emergence on the illicit drug market and are gaining increasing importance in forensic toxicology. Like many other new psychoactive substances, NSOs are consumed without any preclinical safety data or any knowledge on toxicokinetic (TK) data. Due to ethical reasons, controlled human TK studies cannot be performed for the assessment of these relevant data. As an alternative animal experimental approach, six pigs per drug received a single intravenous dose of 100 µg/kg body weight (BW) of U-47700 or 1000 µg/kg BW of tramadol to evaluate whether this species is suitable to assess the TK of NSOs. The drugs were determined in serum and whole blood using a fully validated method based on solid-phase extraction and LC-MS/MS. The concentration-time profiles and a population (pop) TK analysis revealed that a three-compartment model best described the TK data of both opioids. Central volumes of distribution were 0.94 L/kg for U-47700 and 1.25 L/kg for tramadol and central (metabolic) clearances were estimated at 1.57 L/h/kg and 1.85 L/h/kg for U-47700 and tramadol, respectively. The final popTK model parameters for pigs were upscaled via allometric scaling techniques. In comparison to published human data, concentration-time profiles for tramadol could successfully be predicted with single species allometric scaling. Furthermore, possible profiles for U-47700 in humans were simulated. The findings of this study indicate that unlike a multiple species scaling approach, pigs in conjunction with TK modeling are a suitable tool for the assessment of TK data of NSOs and the prediction of human TK data.


Subject(s)
Benzamides/pharmacokinetics , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Tramadol/pharmacokinetics , Administration, Intravenous , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/toxicity , Animals , Benzamides/toxicity , Humans , Illicit Drugs/pharmacokinetics , Illicit Drugs/toxicity , Male , Models, Biological , Species Specificity , Swine , Tissue Distribution , Toxicokinetics , Tramadol/toxicity
8.
Ecotoxicol Environ Saf ; 212: 111999, 2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33550078

ABSTRACT

Tramadol is a widely used analgesic with additional antidepressant and anxiolytic effects. This compound has been reported in continental waters reaching concentrations of µg/L as a consequence of its inefficient removal in sewage treatment plants and increasing use over time. In this study, European chubs (Squalius cephalus) were exposed to 1 µg/L of tramadol in water for 42 days with a subsequent 14 days of depuration. Our results revealed that chubs exposed to this analgesic underwent changes in their behaviour as compared to the control group. The behavioural outcome was also influenced by the individual concentration of tramadol in brain tissue. In particular, experimental fish presented anxiolytic-like effects, characterized by less bold and less social individuals. Exposed animals were less frequently out of the shelter and moved a shorter distance, indicating that they explored the new environment less during the boldness test. In the novel object recognition experiment, although they distinguished the new item, they examined it less and displayed a reduced activity. Shoal cohesion was disrupted as observed in an increased distance between individuals. After the depuration phase, this alteration remained whereas the boldness effect disappeared. Moreover, the degree of behavioural changes was correlated with the concentration of the substance in brain. According to our findings, chronic presence of tramadol in the environment can impact the fitness of exposed aquatic fauna by altering evolutionary crucial behaviours.


Subject(s)
Behavior, Animal/drug effects , Cyprinidae/physiology , Tramadol/toxicity , Water Pollutants, Chemical/toxicity , Animals , Brain/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Fresh Water/chemistry , Tramadol/metabolism , Water Pollutants, Chemical/metabolism
9.
Drug Chem Toxicol ; 44(6): 613-619, 2021 Nov.
Article in English | MEDLINE | ID: mdl-31368376

ABSTRACT

Tramadol (TR) is an analgesic drug used to treat moderate-to-severe pain but it induces seizure even at therapeutic doses. The exact mechanism of TR-inducing seizure is not clear but inhibition of the serotonin, GABA, and nitrous oxide (NOS) pathways are the commonly proposed mechanisms. Adenosinergic system has a crucial function in the modulation of seizure. Also, oxidative damage is an unavoidable effect of the seizure. This study was conducted to evaluate the role of the adenosinergic system on the seizure and oxidative stress biomarkers induced by TR using antagonist of the adenosinergic receptors in the Albino mice. For that purpose, generated clonic seizure, as seizure threshold, was evaluated by TR. Caffeine (CAF; 8 mg/kg, i.p.), a nonselective antagonist of adenosine receptors, was administered 1 hour before the seizure induction. The seizure threshold significantly increased by CAF-treated group when compared to TR group (p < 0.001). Oxidative stress biomarkers such as reactive oxygen species, protein carbonyl content, and lipid peroxidation significantly decreased and glutathione content significantly increased by CAF in brain mitochondria compared to the TR group, whereas oxidative biomarkers significantly increased in the TR group compared to the control group. The results of the present study suggested that the adenosinergic system is involved in seizure induced by TR and meanwhile, inhibition of adenosine receptors can decrease the TR seizure threshold and also decrease the induced oxidative damage in the brain mitochondria.


Subject(s)
Caffeine , Tramadol , Animals , Brain/metabolism , Caffeine/toxicity , Disease Models, Animal , Mice , Mitochondria , Protein Carbonylation , Seizures/chemically induced , Tramadol/metabolism , Tramadol/toxicity
10.
Int J Mol Sci ; 22(14)2021 Jul 17.
Article in English | MEDLINE | ID: mdl-34299276

ABSTRACT

1-cyclohexyl-x-methoxybenzene is a novel psychoactive substance (NPS), first discovered in Europe in 2012 as unknown racemic mixture of its three stereoisomers: ortho, meta and para. Each of these has structural similarities with the analgesic tramadol and the dissociative anesthetic phencyclidine. In light of these structural analogies, and based on the fact that both tramadol and phencyclidine are substances that cause toxic effects in humans, the aim of this study was to investigate the in vitro and in vivo pharmacodynamic profile of these molecules, and to compare them with those caused by tramadol and phencyclidine. In vitro studies demonstrated that tramadol, ortho, meta and para were inactive at mu, kappa and delta opioid receptors. Systemic administration of the three stereoisomers impairs sensorimotor responses, modulates spontaneous motor activity, induces modest analgesia, and alters thermoregulation and cardiorespiratory responses in the mouse in some cases, with a similar profile to that of tramadol and phencyclidine. Naloxone partially prevents only the visual sensorimotor impairments caused by three stereoisomers, without preventing other effects. The present data show that 1-cyclohexyl-x-methoxybenzene derivatives cause pharmaco-toxicological effects by activating both opioid and non-opioid mechanisms and suggest that their use could potentially lead to abuse and bodily harm.


Subject(s)
Analgesics, Opioid/toxicity , Anisoles/toxicity , Benzene Derivatives/toxicity , Hallucinogens/toxicity , Phencyclidine/toxicity , Psychotropic Drugs/toxicity , Receptors, Opioid/metabolism , Tramadol/toxicity , Analgesics, Opioid/chemistry , Animals , Anisoles/chemistry , Benzene Derivatives/chemistry , Cells, Cultured , Cricetinae , Hallucinogens/chemistry , In Vitro Techniques , Male , Mice , Mice, Inbred ICR , Models, Animal , Phencyclidine/chemistry , Psychotropic Drugs/chemistry , Tramadol/chemistry
11.
Blood Purif ; 49(4): 509-512, 2020.
Article in English | MEDLINE | ID: mdl-32018266

ABSTRACT

We describe the case of a 49-year-old woman with a Tramadol intoxication associated with multiorgan failure. Veno-arterial femoro-femoral extracorporeal life support (VA-ECLS) and hemoperfusion (HP) were used as rescue treatments. The emergency medical service found a woman at home unconscious. Once in the hospital, she was intubated and catecholamines support was immediately started for a severe shock. Brain CT was normal, whereas EEG revealed a metabolic encephalopathy pattern. Toxic levels of Tramadol and Quetiapine were detected. VA-ECLS was implanted due to persistent multiorgan failure, and HP with a charcoal cartridge was set to increase the Tramadol clearance. To quantify the charcoal cartridge's removal efficiency of Tramadol, Tramadol concentration was measured before and after the cartridge and before and after the treatment in the patient's blood. The charcoal cartridge showed good extraction ratio during the treatment and no significant rebound effect. VA-ECLS and HP allowed the patient to be weaned from vasoconstrictors and the resolution of the organ failures. These treatments might be lifesaving in the Tramadol intoxication.


Subject(s)
Analgesics, Opioid/toxicity , Drug Overdose/therapy , Extracorporeal Membrane Oxygenation , Hemoperfusion , Tramadol/toxicity , Drug Overdose/complications , Extracorporeal Membrane Oxygenation/methods , Female , Hemoperfusion/methods , Humans , Middle Aged , Multiple Organ Failure/chemically induced , Multiple Organ Failure/complications
12.
Ultrastruct Pathol ; 44(1): 81-102, 2020 Jan 02.
Article in English | MEDLINE | ID: mdl-31924115

ABSTRACT

Tramadol is a common analgesic, frequently used for relieving moderate or severe pain and widely used to delay ejaculation. However, repeated large doses have several adverse effects, especially on the brain tissue. So, this study was designed to assess the potentially deleterious effects of chronic administration of tramadol on principal fields of the hippocampus in adult and juvenile male albino mice. Thirty swiss male albino mice were divided equally into three groups: Group Ia (control adult) 3 months old, Group Ib (control juvenile) 3-week postnatal mice, Group II (tramadol treated adult mice) and Group III (tramadol treated juvenile mice). Both treated groups received tramadol tablets dissolved in water in a dose of 40mg/kg for 1 month by gastric tube. Tramadol treated groups showed degenerative changes in dentate gyrus (DG) granule cells, pyramidal neurons of CA1and CA3 fields in the form of electron-dense or rarified cytoplasm, dilated rER and mitochondrial changes. Additionally, immunohistochemical results revealed significantly increased in caspase 3 positive cells in different hippocampal principal fields. Astrogliosis and microgliosis were proved by the increased immunoreactivity of astrocytes to glial fibrillary acidic protein (GFAP) and microglia to CD68. Morphometric findings showed a significant reduction of both surface area of granule and pyramidal cells, and in thickness of DG, CA1, CA3 layers. Moreover, most of these morphological changes were aggravated in the juvenile-treated group. So, it can be concluded that tramadol abuse can induce an altered morphological change on the principal fields of the hippocampus in adult and juvenile mice.


Subject(s)
Analgesics, Opioid/toxicity , Gliosis/chemically induced , Hippocampus/drug effects , Pyramidal Cells/drug effects , Tramadol/toxicity , Animals , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/pathology , Gliosis/pathology , Hippocampus/pathology , Male , Mice , Microglia/drug effects , Microglia/pathology , Pyramidal Cells/pathology
13.
Toxicol Mech Methods ; 30(3): 177-188, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31618080

ABSTRACT

Several deleterious effects of Tramadol including deaths were reported especially when used in large doses. Being metabolized mainly in the liver, Tramadol have serious hepatotoxic effects. This study investigates the effect of vitamin E on Tramadol-induced hepatotoxicity in rats by evaluating the antioxidant biochemical markers, the histopathological and immunohistochemical changes.Thirty adult mature male albino rats were divided into five groups (Gs); G1: negative control; G2: received Tramadol 150 mg/kg, G 3-5: received Tramadol plus vitamin E in concentrations of 50 mg/kg, 100 mg/kg and 200 mg/kg respectively. Liver function parameters and oxidative markers in liver tissue (CAT, SOD, GSH, and MDA) were estimated. Liver samples were processed for histopathological and immunohistochemical (Caspase 3 and TNF[Formula: see text]) examinations. The results indicated that Sub-chronic administration of Tramadol resulted in impaired liver functions, increased oxidative stress parameters with decreased antioxidant capacity of liver tissues, severe hepatocellular damage (hydropic degeneration, steatosis and apoptosis) and strong immunoexpression to TNF[Formula: see text] and Caspase 3. All these effects were ameliorated with concomitant administration of vitamin E especially with high doses. The co-treatment of Tramadol-intoxicated rats with Vitamin E, especially in high doses, protects against hepatic toxicity.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Tramadol/toxicity , Vitamin E/administration & dosage , Animals , Body Weight/drug effects , Caspase 3/metabolism , Dietary Supplements , Immunohistochemistry , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Organ Size/drug effects , Rats , Tumor Necrosis Factor-alpha/analysis
14.
Bratisl Lek Listy ; 121(6): 400-410, 2020.
Article in English | MEDLINE | ID: mdl-32484703

ABSTRACT

AIM: The present study investigated the role of redox balance, inflammation, mitochondrial dysfunction, and apoptosis in Tramadol (Tra)-induced testicular toxicity. METHOD: Twenty-four male Wistar rats were randomly divided into either the control group or the groups receiving different doses of Tra (25, 50, and 75 mg/kg/day, i.p.) for 21 successive days. Testicular tissues were collected for oxidative stress, mitochondrial function, sperm assays and histopathological evaluation. Real-time polymerase chain reaction was performed to evaluate the markers of inflammation and apoptosis. RESULTS: Tra caused a significant reduction in the sperm count, motility and morphology, while it caused a marked increase in oxidative stress parameters. In addition, Tra induced testicular mitochondrial dysfunction due to the collapse of mitochondrial membrane potential and mitochondrial swelling. It also led to the significant inhibition of anti-apoptotic Bcl-2 expression, besides a significant increase in pro-apoptotic Bax expression. There was a significant increase in the level of tumour necrosis factor-α, interlukin-1ß and nuclear factor kappa B. Histopathological degenerative changes were observed in the testis after Tra exposure. CONCLUSIONS: The present results suggest that Tra exposure may lead to reproductive toxicity due to the loss of the antioxidant defence system, mitochondrial dysfunction, and activation of inflammatory and apoptotic pathways (Tab. 4, Fig. 5, Ref. 63).


Subject(s)
Apoptosis , Narcotics , Oxidative Stress , Tramadol , Animals , Antioxidants , Apoptosis/drug effects , Humans , Male , Mitochondria , NF-kappa B , Narcotics/toxicity , Oxidative Stress/drug effects , Rats , Rats, Wistar , Spermatozoa , Testis/drug effects , Tramadol/toxicity
15.
Molecules ; 24(4)2019 Feb 15.
Article in English | MEDLINE | ID: mdl-30769936

ABSTRACT

In recent years, many studies have highlighted the consistent finding of tramadol (TRA) in the effluents from wastewater treatment plants (WTPs) and also in some rivers and lakes in both Europe and North America, suggesting that TRA is removed by no more than 36% by specific disinfection treatments. The extensive use of this drug has led to environmental pollution of both water and soil, up to its detection in growing plants. In order to expand the knowledge about TRA toxicity as well as the nature of its disinfection by-products (DBPs), a simulation of the waste treatment chlorination step has been reported herein. In particular, we found seven new by-products, that together with TRA, have been assayed on different living organisms (Aliivibrio fischeri, Raphidocelis subcapitata and Daphnia magna), to test their acute and chronic toxicity. The results reported that TRA may be classified as a harmful compound to some aquatic organisms whereas its chlorinated product mixture showed no effects on any of the organisms tested. All data suggest however that TRA chlorination treatment produces a variety of DBPs which can be more harmful than TRA and a risk for the aquatic environment and human health.


Subject(s)
Disinfection , Hypochlorous Acid/analysis , Hypochlorous Acid/toxicity , Tramadol/analysis , Tramadol/toxicity , Disinfection/methods , Hypochlorous Acid/chemistry , Molecular Structure , Spectrum Analysis , Toxicity Tests , Tramadol/chemistry
16.
Ultrastruct Pathol ; 42(3): 295-303, 2018.
Article in English | MEDLINE | ID: mdl-29668338

ABSTRACT

The effect of tramadol addiction on epididymal structure was not investigated before. Therefore, this experimental study was carried out to investigate the effect of chronic tramadol use on the epididymal structure using light and electron microscopies. Thirty adult Wister Albino male rats were divided into two groups: control group (five rats) and tramadol-treated group (25 rats), which was further subdivided into five subgroups that received tramadol orally at 4.5, 9, 45, 90, and 135 mg/kg/day, respectively, for 18 weeks. Epididymal tissues were dissected and processed for histopathological examination. Morphometric analysis showed significantly reduced mean values of epididymal ducts' diameters and epithelial height in the tramadol-treated group compared with the control group. Light microscopic examination revealed degeneration and necrosis of epididymal cells in the tramadol-treated group. Electron microscopic (EM) examination showed ultrastructure alterations in a dose-dependent manner. In conclusion, tramadol can adversely affect all epididymal cells, which subsequently deteriorate epididymal function and may affect sperm maturation, leading to subfertility.


Subject(s)
Analgesics, Opioid/toxicity , Epididymis/drug effects , Epididymis/ultrastructure , Tramadol/toxicity , Animals , Male , Microscopy, Electron, Transmission , Rats , Rats, Wistar
17.
Toxicol Appl Pharmacol ; 310: 108-119, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27641627

ABSTRACT

Poisoning with opioid analgesics including tramadol represents a challenge. Tramadol may induce respiratory depression, seizures and serotonin syndrome, possibly worsened when in combination to benzodiazepines. Our objectives were to investigate tramadol-related neurotoxicity, consequences of diazepam/tramadol combination, and mechanisms of drug-drug interactions in rats. Median lethal-doses were determined using Dixon-Bruce's up-and-down method. Sedation, seizures, electroencephalography and plethysmography parameters were studied. Concentrations of tramadol and its metabolites were measured using liquid-chromatography-high-resolution-mass-spectrometry. Plasma, platelet and brain monoamines were measured using liquid-chromatography coupled to fluorimetry. Median lethal-doses of tramadol and diazepam/tramadol combination did not significantly differ, although time-to-death was longer with combination (P=0.04). Tramadol induced dose-dependent sedation (P<0.05), early-onset seizures (P<0.001) and increase in inspiratory (P<0.01) and expiratory times (P<0.05). The diazepam/tramadol combination abolished seizures but significantly enhanced sedation (P<0.01) and respiratory depression (P<0.05) by reducing tidal volume (P<0.05) in addition to tramadol-related increase in respiratory times, suggesting a pharmacodynamic mechanism of interaction. Plasma M1 and M5 metabolites were mildly increased, contributing additionally to tramadol-related respiratory depression. Tramadol-induced early-onset increase in brain concentrations of serotonin and norepinephrine was not significantly altered by the diazepam/tramadol combination. Interestingly neither pretreatment with cyproheptadine (a serotonin-receptor antagonist) nor a benserazide/5-hydroxytryptophane combination (enhancing brain serotonin) reduced tramadol-induced seizures. Our study shows that diazepam/tramadol combination does not worsen tramadol-induced fatality risk but alters its toxicity pattern with enhanced respiratory depression but abolished seizures. Drug-drug interaction is mainly pharmacodynamic but increased plasma M1 and M5 metabolites may also contribute to enhancing respiratory depression. Tramadol-induced seizures are independent of brain serotonin.


Subject(s)
Analgesics, Opioid/toxicity , Diazepam/toxicity , Drug Overdose , Nervous System/drug effects , Tramadol/toxicity , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Animals , Brain/drug effects , Brain/metabolism , Diazepam/administration & dosage , Dose-Response Relationship, Drug , Male , Norepinephrine/metabolism , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Tramadol/administration & dosage , Tramadol/pharmacokinetics
18.
Epilepsy Behav ; 36: 90-6, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24892755

ABSTRACT

Tramadol is frequently used as a pain reliever. However, it has been sometimes noted to have the potential to cause seizures. Because of its dual mechanism of action (both opioid and nonopioid), the adverse effect profile of tramadol can be different in comparison with single-mechanism opioid analgesics, such as morphine. In the present study, the facilitatory effects of tramadol and morphine on pentylenetetrazol-induced seizures using different routes of administration were compared in rats. Adult female rats were divided into six groups and continuously received saline, morphine, or tramadol on a daily basis for 15 days [gavage (PO) or intraperitoneal (IP)]. An increasing dose of morphine and tramadol was used to prevent resistance to repetitive dose (20-125 mg/kg). Following one week of withdrawal period and 30 min before the seizure induction (PTZ=80 mg/kg, IP), each group of rats was further divided into subgroups that received saline, morphine, or tramadol for the second time on the 22nd day of the experiment. Results showed that, while morphine, tramadol, and their administration had different effects on seizure behaviors, both acute and chronic administrations of morphine and tramadol potentiated PTZ-induced seizures. However, there was no significant difference between morphine and tramadol in terms of seizure severity. Effects of morphine and tramadol on PTZ-induced seizures were also stable following one week of withdrawal. In conclusion, this study indicated similar severity in the proconvulsant effect of morphine and tramadol on PTZ-induced seizures, which might depend on their similar effects on GABAergic pathways.


Subject(s)
Analgesics/toxicity , Convulsants/administration & dosage , Morphine/toxicity , Pentylenetetrazole/administration & dosage , Seizures/chemically induced , Tramadol/toxicity , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Routes , Drug Synergism , Female , Rats , Rats, Wistar , Time Factors
19.
Naunyn Schmiedebergs Arch Pharmacol ; 397(2): 889-897, 2024 02.
Article in English | MEDLINE | ID: mdl-37526689

ABSTRACT

It is reported that tramadol can induce neurotoxic effects with the production of DNA damage, mitochondrial dysfunction, and oxidative stress. The current study aimed to evaluate the potential role of mitochondrial impairment in the pathogenesis of tramadol-induced neurotoxicity, and protective effect of sinapic acid (SA) against it in isolated mitochondria from rat brain. Mitochondria were isolated and were incubated with toxic concentrations (100 µM) of tramadol and then cotreated with tramadol + SA (10, 50, and 100 µM). Biomarkers of mitochondrial toxicity including succinate dehydrogenases (SDH) activity, reactive oxygen species (ROS), lipid peroxidation (LPO), mitochondrial membrane potential (MMP), GSH depletion, and mitochondrial swelling were assessed. Our results showed a significant decrease in SDH activity, and a significant increase in ROS, LPO, GSH depletion, MMP collapse, and mitochondrial swelling was detected in tramadol group. We observed that 50 and 100 µM SA cotreatment for 1 h efficiently ameliorated tramadol-caused damage in mitochondrial dysfunction, accumulation of ROS, LPO, GSH depletion, depolarization of mitochondrial membrane potential, and mitochondrial swelling. These data suggest that mitochondrial impairment and oxidative stress are mechanisms involved in the pathogenesis of tramadol-induced neurotoxicity. Also, results indicate that SA antagonizes against tramadol-induced mitochondrial toxicity and suggest SA may be a preventive/therapeutic agent for tramadol-induced neurotoxicity complications.


Subject(s)
Coumaric Acids , Mitochondrial Diseases , Tramadol , Rats , Animals , Reactive Oxygen Species/metabolism , Tramadol/toxicity , Mitochondria , Oxidative Stress , Lipid Peroxidation , Brain , Membrane Potential, Mitochondrial
20.
Neurotoxicology ; 99: 1-9, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37652187

ABSTRACT

Most teenagers mix up various psychoactive cocktail substances in combinations to get intoxicated. The role of the mixture combination of codeine (CDE), tramadol (TMD), and Cannabis sativa (CNB) on brain cognition, purinergic, cholinergic, and antioxidant enzyme activities remains unknown. This study sought to assess the mechanism of action of combinations of CDE+ TMD+ CNB on the function and activities of the brain of male Wistar rats. Forty-eight male Wistar rats were divided into 8 groups, n = 6. Group 1 served as a control, groups 2, 3, and 4 were exposed to CDE (2 mg/kg bw), TMD (10 mg/kg bw), and CNB (200 mg/kg bw), while groups 5, 6, 7, and 8 were co-administered with CDE+TMD, CNB+ TMD, CNB+CDE, and CNB+TMD+CDE orally for 28 days. This study revealed the effect of prolonged administration of CNB, TMD, and CDE on the suppression of cognitive function, acetyl-cholinesterase (AChE), butyl-cholinesterase (BChE), monoamine oxidase (MAO) enzyme activities, and antioxidant enzyme activities in rats' brains when compared against control rats (P < 0.05). However, the activities of ectonucleosides (NTPdase), adenosine deaminase (ADA), and malondialdehyde levels produced in the brain of rats were significantly elevated (P < 0.05). This study reported the mechanism behind the neurotoxicity of CNB, TMD, and CDE on rats' cognitive, cholinergic, purinergic, and antioxidant enzymes as a consequence of the drastic reduction in cholinesterase enzyme activities leading to neurotransmitter poisoning.


Subject(s)
Cannabis , Tramadol , Rats , Male , Animals , Rats, Wistar , Antioxidants , Tramadol/toxicity , Codeine , Cholinesterases , Cholinergic Agents , Cognition
SELECTION OF CITATIONS
SEARCH DETAIL