Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Type of study
Publication year range
1.
Environ Toxicol Pharmacol ; 96: 103998, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36228991

ABSTRACT

We sought to evaluate the efficacy of trapping free hydrogen sulfide (H2S) following severe H2S intoxication. Sodium hydrosulfide solution (NaHS, 20 mg/kg) was administered intraperitoneally in 69 freely moving rats. In a first group (protocol 1), 40 rats were randomly assigned to receive saline (n = 20) or the cobalt compound tetranitrocobinamide (TNCbi) (n = 20, 75 mg/kg iv), one minute into coma, when free H2S was still present in the blood. A second group of 27 rats received TNCbi or saline, following epinephrine, 5 min into coma, when the concentration of free H2S has drastically decreased in the blood. In protocol 1, TNCbi significantly increased immediate survival (65 vs 20 %, p < 0.01) while in protocol 2, administration of TNCbi led to the same outcome as untreated animals. We hypothesize that the decreased efficacy of TNCbi with time likely reflects the rapid spontaneous disappearance of the pool of free H2S in the blood following H2S exposure.


Subject(s)
Coma , Hydrogen Sulfide , Animals , Rats , Sulfides , Hydrogen Sulfide/toxicity , Epinephrine
2.
J Appl Physiol (1985) ; 132(6): 1407-1422, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35421320

ABSTRACT

The objective of our study was to establish in sedated rats the consequences of high-dose fentanyl-induced acute muscle rigidity on the mechanical properties of the respiratory system and on the metabolic rate. Doses of fentanyl that we have previously shown to produce persistent rigidity of the muscles of the limbs and trunk in the rat (150-300 µg/kg iv), were administered in 23 volume-controlled mechanically ventilated and sedated rats. The effects of a low dose of the FDA-approved central α-2 agonist, dexmedetomidine (3 µg/kg iv), which has been suggested to oppose fentanyl-induced muscle rigidity, were determined after fentanyl administration. Fentanyl produced a significant decrease in compliance of the respiratory system (Crs) in all the rats that were studied. In 13 rats, an abrupt response occurred within 90 s, consisting of rapid rhythmic contractions of most skeletal muscles that were replaced by persistent tonic/tetanic contractions leading to a significant decrease of Crs (from 0.51 ± 0.11 mL/cmH2O to 0.36 ± 0.08 mL/cmH2O, 3 min after fentanyl injection). In the other 10 animals, Crs progressively decreased to 0.26 ± 0.06 mL/cmH2O at 30 min. There was a significant rise in oxygen consumption (V̇o2) during these muscle contractions (from 8.48 ± 4.31 to 11.29 ± 2.57 mL/min), which led to a significant hypoxemia, despite ventilation being held constant. Dexmedetomidine provoked a significant and rapid increase in Crs toward baseline levels, whereas decreasing the metabolic rate and restoring normoxemia. We propose that the changes in respiratory mechanics and metabolism produced by opioid-induced muscle rigidity contribute to fentanyl lethality.NEW & NOTEWORTHY The decrease in respiratory compliance and increased metabolism-induced hypoxemia produced by an overdose of fentanyl, in and of themselves, contribute to fentanyl toxicity.


Subject(s)
Dexmedetomidine , Analgesics, Opioid , Animals , Dexmedetomidine/adverse effects , Fentanyl/pharmacology , Hypoxia/chemically induced , Muscle Rigidity , Pulmonary Gas Exchange , Rats , Respiratory Mechanics
3.
J Neurophysiol ; 125(4): 1396-1407, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33656934

ABSTRACT

We have investigated the potential acute desensitizing role of the ß arrestin 2 (ß-arr2) pathway on the ventilatory depression produced by levels of fentanyl ranging from analgesic to life-threatening (0.1 to 60 mg/kg ip) in control and ß-arr2-deficient nonsedated mice. Fentanyl at doses of 0.1, 0.5, and 1 mg/kg ip-corresponding to the doses previously used to study the role of ß-arr2 pathway-decreased ventilation, but along the V̇e/V̇co2 relationship established in baseline conditions. This reduction in ventilation was therefore indistinguishable from the decrease in breathing during the periods of spontaneous immobility. Above 1.5 mg/kg, however, ventilation was depressed out of proportion of the changes in metabolic rate, suggesting a specific depression of the drive to breathe. The ventilatory responses were similar between the two groups. At high doses of fentanyl (60 mg/kg ip) 1 out of 20 control mice died by apnea versus 8 out of 20 ß-arr2-deficient mice (P = 0.008). In the surviving mice, ventilation was however identical in both groups. The ventilatory effects of fentanyl in ß-arr2-deficient mice, reported in the literature, are primarily mediated by the "indirect" effects of sedation/hypometabolism on breathing control. There was an excess mortality at very high doses of fentanyl in the ß-arr2-deficient mice, mechanisms of which are still open to question, as the capacity of maintaining a rhythmic, although profoundly depressed, breathing activity remains similar in all of the surviving control and ß-arr2-deficient mice.NEW & NOTEWORTHY When life-threatening doses of fentanyl are used in mice, the ß-arrestin 2 pathway appears to play a critical role in the recovery from opioid overdose. This observation calls into question the use of G protein-biased µ-opioid receptor agonists, as a strategy for safer opioid analgesic drugs.


Subject(s)
Analgesics, Opioid/pharmacology , Apnea/chemically induced , Fentanyl/pharmacology , Opiate Overdose/metabolism , Respiration/drug effects , Signal Transduction/drug effects , beta-Arrestin 2/metabolism , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Female , Fentanyl/administration & dosage , Fentanyl/adverse effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , beta-Arrestin 2/deficiency
4.
Toxicol Rep ; 7: 1459-1464, 2020.
Article in English | MEDLINE | ID: mdl-33194557

ABSTRACT

We have determined the effects of azure B (AzB), the main demethylated metabolite of methylene blue (MB), on a model of lethal cyanide intoxication. Our rationale was the following: AzB 1- possesses redox properties very similar to those of MB, which is a potent cyanide antidote, 2- may present a higher intracellular diffusibility than MB, 3- is already present in commercially available solutions of MB, and 4- appears very quickly in the blood after MB administration. AzB could therefore be a member of the phenothiazium chromophore family of interest to treat cyanide intoxication. We found, in spontaneously breathing urethane sedated rats, that AzB mimicked the effects of MB by increasing metabolism, ventilation and cardiac contractility up to 30-40 mg/kg. AzB had a lethal toxicity when the dose of 60 mg/kg was reached. Doses of AzB were therefore chosen in keeping with these data and the doses of MB previously used against cyanide intoxication (4-20 mg/kg) in the rat - doses corresponding to those used in humans to treat methemoglobinemia. KCN, infused at the rate of 0.375 mg/kg/min iv for 13 min, was fatal within 15 min in 100 % of our un-anesthetized rats. AzB at the dose of 4 mg/kg (n = 5) or 10 mg/kg (n = 5) administered 3 min into cyanide infusion allowed 100 % of the animals to survive with no clinical sequelae. The onset of coma was also significantly delayed and no apnea or gasping occurred. At the dose of 20 mg/kg, AzB was much less effective. At 4 mg/kg, the antidotal effects of AzB were significantly better than those produced by MB at the same dose and were not different from the effects produced by 20 mg/kg MB. We conclude that AzB is a potent cyanide antidote at relatively low doses.

5.
Ann N Y Acad Sci ; 1479(1): 108-121, 2020 11.
Article in English | MEDLINE | ID: mdl-32374444

ABSTRACT

The aim of the present study was to determine whether methylene blue (MB) could directly oppose the neurological toxicity of a lethal cyanide (CN) intoxication. KCN, infused at the rate of 0.375 mg/kg/min intravenously, produced 100% lethality within 15 min in unanaesthetized rats (n = 12). MB at 10 (n = 5) or 20 mg/kg (n = 5), administered 3 min into CN infusion, allowed all animals to survive with no sequelae. No apnea and gasping were observed at 20 mg/kg MB (P < 0.001). The onset of coma was also significantly delayed and recovery from coma was shortened in a dose-dependent manner (median of 359 and 737 seconds, respectively, at 20 and 10 mg/kg). At 4 mg/kg MB (n = 5), all animals presented faster onset of coma and apnea and a longer period of recovery than at the highest doses (median 1344 seconds, P < 0.001). MB reversed NaCN-induced resting membrane potential depolarization and action potential depression in primary cultures of human fetal neurons intoxicated with CN. MB restored calcium homeostasis in the CN-intoxicated human SH-SY5Y neuroblastoma cell line. We conclude that MB mitigates the neuronal toxicity of CN in a dose-dependent manner, preventing the lethal depression of respiratory medullary neurons and fatal outcome.


Subject(s)
Antidotes/pharmacology , Methylene Blue/pharmacology , Neurons , Neurotoxicity Syndromes , Potassium Cyanide/toxicity , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Male , Neurons/metabolism , Neurons/pathology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/prevention & control , Rats , Rats, Sprague-Dawley
6.
Respir Physiol Neurobiol ; 277: 103428, 2020 06.
Article in English | MEDLINE | ID: mdl-32151709

ABSTRACT

Breathing resumes within one to two minutes following fentanyl overdose induced apnea in spontaneously breathing rats. As this regular rhythm is produced at a time wherein fentanyl concentrations and receptor occupancy are likely to be extremely high, the mechanisms initiating and sustaining such a respiratory activity remain unclear. Forty-four un-anesthetized adult rats were studied in an open-flow plethysmograph. Regardless of the dose of fentanyl that was used, i.e. 50 µg.kg-1 (n = 8), 100 µg.kg-1 (n = 8) or 300 µg.kg-1 (n = 7), all rats developed an immediate central apnea followed by a depressed regular rhythm that was produced 118, 97 and 81 s (median) later, respectively. Only one rat did not recover. This inspiratory and regular activity consisted of a low frequency and tidal volume pattern with a significant reduction in V̇E/V̇CO2 ratio, which persisted for at least 30 min and that was not different between 100 or 300 µg.kg-1. The time at which this respiratory rhythm emerged, following the highest dose of fentanyl, was not affected by 100 % O2 or 8% CO2/15 % O2. The absolute level of ventilation was however higher in hypercapnic and moderately hypoxic conditions than in hyperoxia. When a second injection of the highest dose of fentanyl (300 µg.kg-1) was performed at 10 min, ventilation was not significantly affected and no apnea was produced in major contrast to the first injection. When a similar injection was performed 30 min after the first injection, in a separate group of rats, an apnea and breathing depression was produced in 30 % of the animals, while in the other rats, ventilation was unaffected. We conclude that the depressed regular respiratory activity emerging during and following fentanyl overdose is uniquely resistant to fentanyl.


Subject(s)
Analgesics, Opioid/toxicity , Drug Overdose/physiopathology , Fentanyl/toxicity , Respiratory Mechanics/physiology , Animals , Apnea/chemically induced , Apnea/physiopathology , Male , Rats , Rats, Sprague-Dawley , Respiratory Mechanics/drug effects , Tidal Volume/drug effects , Tidal Volume/physiology
7.
Anesthesiology ; 132(5): 1138-1150, 2020 05.
Article in English | MEDLINE | ID: mdl-32044798

ABSTRACT

BACKGROUND: As severe acute hypoxemia produces a rapid inhibition of the respiratory neuronal activity through a nonopioid mechanism, we have investigated in adult rats the effects of hypoxemia after fentanyl overdose-induced apnea on (1) autoresuscitation and (2) the antidotal effects of naloxone. METHODS: In nonsedated rats, the breath-by-breath ventilatory and pulmonary gas exchange response to fentanyl overdose (300 µg · kg · min iv in 1 min) was determined in an open flow plethysmograph. The effects of inhaling air (nine rats) or a hypoxic mixture (fractional inspired oxygen tension between 7.3 and 11.3%, eight rats) on the ability to recover a spontaneous breathing rhythm and on the effects of naloxone (2 mg · kg) were investigated. In addition, arterial blood gases, arterial blood pressure, ventilation, and pulmonary gas exchange were determined in spontaneously breathing tracheostomized urethane-anesthetized rats in response to (1) fentanyl-induced hypoventilation (7 rats), (2) fentanyl-induced apnea (10 rats) in air and hyperoxia, and (3) isolated anoxic exposure (4 rats). Data are expressed as median and range. RESULTS: In air-breathing nonsedated rats, fentanyl produced an apnea within 14 s (12 to 29 s). A spontaneous rhythmic activity always resumed after 85.4 s (33 to 141 s) consisting of a persistent low tidal volume and slow frequency rhythmic activity that rescued all animals. Naloxone, 10 min later, immediately restored the baseline level of ventilation. At fractional inspired oxygen tension less than 10%, fentanyl-induced apnea was irreversible despite a transient gasping pattern; the administration of naloxone had no effects. In sedated rats, when PaO2 reached 16 mmHg during fentanyl-induced apnea, no spontaneous recovery of breathing occurred and naloxone had no rescuing effect, despite circulation being maintained. CONCLUSIONS: Hypoxia-induced ventilatory depression during fentanyl induced apnea (1) opposes the spontaneous emergence of a respiratory rhythm, which would have rescued the animals otherwise, and (2) prevents the effects of high dose naloxone.


Subject(s)
Analgesics, Opioid/toxicity , Fentanyl/toxicity , Hypoxia/physiopathology , Naloxone/therapeutic use , Narcotic Antagonists/therapeutic use , Wakefulness/drug effects , Animals , Hypnotics and Sedatives/toxicity , Hypoxia/chemically induced , Hypoxia/drug therapy , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Recovery of Function/physiology , Severity of Illness Index , Wakefulness/physiology
8.
Toxicol Sci ; 170(1): 82-94, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30907955

ABSTRACT

Our study was aimed at (1) determining the efficacy of the dye methylene blue (MB), following a rapidly lethal cyanide (CN) intoxication in un-sedated rats; (2) clarifying some of the mechanisms responsible for the antidotal properties produced by this potent cyclic redox dye. Sixty-nine awake rats acutely intoxicated by CN (IP, KCN 7 mg/kg) received saline, MB (20 mg/kg) or hydroxocobalamin (HyCo, 150 mg/kg) when in deep coma. Survival in this model was very low, reaching 9% at 60 min without any treatment. Methylene blue significantly increased survival (59%, p < .001) at 60 min, versus 37% with HyCo (p < .01). In addition, 8 urethane-anesthetized rats were exposed to a sublethal CN intoxication (KCN, 0.75 mg/kg/min IV for 4 min); they received MB (20 mg/kg, IV) or saline, 5 min after the end of CN exposure. All MB-treated rats displayed a significant reduction in hyperlactacidemia, a restoration of pyruvate/lactate ratio-a marker of NAD/NADH ratio-and an increase in CO2 production, a marker of the activity of the TCA cycle. These changes were also associated with a 2-fold increase in the pool of CN in red cells. Based on series of in vitro experiments, looking at the effects of MB on NADH, as well as the redox effects of MB on hemoglobin and cytochrome c, we hypothesize that the antidotal properties of MB can in large part be accounted for by its ability to readily restore NAD/NADH ratio and to cyclically re-oxidize then reduce the iron in hemoglobin and the electron chain complexes. All of these effects can account for the rapid antidotal properties of this dye following CN poisoning.


Subject(s)
Antidotes/pharmacology , Cyanides/poisoning , Methylene Blue/pharmacology , Animals , Coma/chemically induced , Coma/drug therapy , Coma/metabolism , Cytochromes c/metabolism , Hemoglobins/metabolism , Hydroxocobalamin/pharmacology , Male , Methemoglobin/metabolism , NAD/metabolism , Rats
9.
Toxicol Sci ; 168(2): 443-459, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30590764

ABSTRACT

Exposure to toxic levels of hydrogen sulfide (H2S) produces an acute cardiac depression that can be rapidly fatal. We sought to characterize the time course of the cardiac effects produced by the toxicity of H2S in sheep, a human sized mammal, and to describe the in vivo and in vitro antidotal properties of methylene blue (MB), which has shown efficacy in sulfide intoxicated rats. Infusing NaHS (720 mg) in anesthetized adult sheep produced a rapid dilation of the left ventricular with a decrease in contractility, which was lethal within about 10 min by pulseless electrical activity. MB (7 mg/kg), administered during sulfide exposure, maintained cardiac contractility and allowed all of the treated animals to recover. At a dose of 350 mg NaHS, we were able to produce an intoxication, which led to a persistent decrease in ventricular function for at least 1 h in nontreated animals. Administration of MB, 3 or 30 min after the end of exposure, whereas all free H2S had already vanished, restored cardiac contractility and the pyruvate/lactate (P/L) ratio. We found that MB exerts its antidotal effects through at least 4 different mechanisms: (1) a direct oxidation of free sulfide; (2) an increase in the pool of "trapped" H2S in red cells; (3) a restoration of the mitochondrial substrate-level phosphorylation; and (4) a rescue of the mitochondrial electron chain. In conclusion, H2S intoxication produces acute and long persisting alteration in cardiac function in large mammals even after all free H2S has vanished. MB exerts its antidotal effects against life-threatening sulfide intoxication via multifarious properties, some of them unrelated to any direct interaction with free H2S.


Subject(s)
Antidotes/pharmacology , Gas Poisoning/prevention & control , Hydrogen Sulfide/poisoning , Methylene Blue/pharmacology , Ventricular Dysfunction, Left/prevention & control , Animals , Antidotes/administration & dosage , Cytochromes c/blood , Echocardiography , Female , Gas Poisoning/blood , Gas Poisoning/etiology , Hemoglobins/analysis , Membrane Potential, Mitochondrial/drug effects , Methylene Blue/administration & dosage , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Sheep , Ventricular Dysfunction, Left/blood , Ventricular Dysfunction, Left/chemically induced
10.
Clin Toxicol (Phila) ; 56(9): 828-840, 2018 09.
Article in English | MEDLINE | ID: mdl-29451035

ABSTRACT

BACKGROUND: Although methylene blue (MB) had long been proposed to counteract the effects of cyanide (CN) intoxication, research on its mechanisms of action and efficacy has been abandoned for decades. Recent studies on the benefits of MB in post-anoxic injuries have prompted us to reexamine the relevance of this historical observation. METHODS: Our study was performed in adult male Sprague-Dawley rats and on HEK293T epithelial cells. First, the effects and toxicity of MB (0-80 mg/kg) on circulation and metabolism were established in four urethane-anesthetized rats. Then nine rats received a lethal infusion of a solution of KCN (0.75 mg/kg/min) and were treated by either saline or MB, at 20 mg/kg, a dose that we found to be innocuous in rat and to correspond to a dose of about 4 mg/kg in humans. MB was also administered 5 min after the end of a sub-lethal exposure to CN in a separate group of 10 rats. In addition, ATP/ADP ratio, ROS production, mitochondrial membrane potential (Δψm) and cellular O2 consumption rate (OCR) were determined in HEK293T cells exposed to toxic levels of CN (200 µM for 10 min) before and after applying a solution containing MB (1-100 µM for 10 min). RESULTS: Methylene blue was found to be innocuous up to 50 mg/kg. KCN infusion (0.75 mg/kg/min) killed all animals within 7-8 min. MB (20 mg/kg) administered at the same time restored blood pressure, cardiac contractility and limited O2 deficit, allowing all the animals to survive, without any significant methemoglobinemia. When administered 5 min after a non-lethal CN intoxication, MB sped up the recovery of lactate and O2 deficit. Finally, MB was able to decrease the production of ROS and restore the ATP/ADP ratio, Δψm as well as OCR of epithelial cells intoxicated by CN. CONCLUSIONS: The present observations should make us consider the potential interest of MB in the treatment of CN intoxication. The mechanisms of the antidotal properties of MB cannot be accounted for by the creation of a cyanomethemoglobinemia, rather its protective effects appears to be related to the unique properties of this redox dye, which, depending on the dose, could directly oppose some of the consequences of the metabolic depression produced by CN at the cellular level.


Subject(s)
Antidotes/pharmacology , Antidotes/therapeutic use , Cyanides/poisoning , HEK293 Cells/drug effects , Methylene Blue/pharmacology , Methylene Blue/therapeutic use , Poisoning/drug therapy , Animals , Dose-Response Relationship, Drug , Humans , Male , Rats , Rats, Sprague-Dawley
11.
Shock ; 47(3): 352-362, 2017 03.
Article in English | MEDLINE | ID: mdl-27513083

ABSTRACT

Our objective was to determine how circulatory failure develops following systemic administration of potassium cyanide (KCN). We used a noninhaled modality of intoxication, wherein the change in breathing pattern would not influence the diffusion of CN into the blood, akin to the effects of ingesting toxic levels of CN. In a group of 300 to 400 g rats, CN-induced coma (CN i.p., 7 mg/kg) produced a central apnea within 2 to 3 min along with a potent and prolonged gasping pattern leading to autoresuscitation in 38% of the animals. Motor deficits and neuronal necrosis were nevertheless observed in the surviving animals. To clarify the mechanisms leading to potential autoresuscitation versus asystole, 12 urethane-anesthetized rats were then exposed to the lowest possible levels of CN exposure that would lead to breathing depression within 7 to 8 min; this dose averaged 0.375 mg/kg/min i.v. At this level of intoxication, a cardiac depression developed several minutes only after the onset of the apnea, leading to cardiac asystole as PaO2 reached value approximately 15 Torr, unless breathing was maintained by mechanical ventilation or through spontaneous gasping. Higher levels of KCN exposure in 10 animals provoked a primary cardiac depression, which led to a rapid cardiac arrest by pulseless electrical activity (PEA) despite the maintenance of PaO2 by mechanical ventilation. These effects were totally unrelated to the potassium contained in KCN. It is concluded that circulatory failure can develop as a direct consequence of CN-induced apnea but in a narrow range of exposure. In this "low" range, maintaining pulmonary gas exchange after exposure, through mechanical ventilation (or spontaneous gasping), can reverse cardiac depression and restore spontaneous breathing. At higher level of intoxication, cardiac depression is to be treated as a specific and spontaneously irreversible consequence of CN exposure, leading to a PEA.


Subject(s)
Cyanides/toxicity , Shock/chemically induced , Animals , Apnea/chemically induced , Blood Gas Analysis , Male , Potassium Chloride , Pulmonary Gas Exchange , Rats , Rats, Sprague-Dawley
12.
Am J Physiol Regul Integr Comp Physiol ; 310(11): R1030-44, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26962024

ABSTRACT

We have previously reported that methylene blue (MB) can counteract hydrogen sulfide (H2S) intoxication-induced circulatory failure. Because of the multifarious effects of high concentrations of H2S on cardiac function, as well as the numerous properties of MB, the nature of this interaction, if any, remains uncertain. The aim of this study was to clarify 1) the effects of MB on H2S-induced cardiac toxicity and 2) whether L-type Ca(2+) channels, one of the targets of H2S, could transduce some of the counteracting effects of MB. In sedated rats, H2S infused at a rate that would be lethal within 5 min (24 µM·kg(-1)·min(-1)), produced a rapid fall in left ventricle ejection fraction, determined by echocardiography, leading to a pulseless electrical activity. Blood concentrations of gaseous H2S reached 7.09 ± 3.53 µM when cardiac contractility started to decrease. Two to three injections of MB (4 mg/kg) transiently restored cardiac contractility, blood pressure, and V̇o2, allowing the animals to stay alive until the end of H2S infusion. MB also delayed PEA by several minutes following H2S-induced coma and shock in unsedated rats. Applying a solution containing lethal levels of H2S (100 µM) on isolated mouse cardiomyocytes significantly reduced cell contractility, intracellular calcium concentration ([Ca(2+)]i) transient amplitudes, and L-type Ca(2+) currents (ICa) within 3 min of exposure. MB (20 mg/l) restored the cardiomyocyte function, ([Ca(2+)]i) transient, and ICa The present results offer a new approach for counteracting H2S toxicity and potentially other conditions associated with acute inhibition of L-type Ca(2+) channels.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium/metabolism , Hydrogen Sulfide/poisoning , Methylene Blue/administration & dosage , Myocardial Contraction/drug effects , Myocytes, Cardiac/metabolism , Animals , Antidotes/administration & dosage , Antioxidants/administration & dosage , Calcium Channels, L-Type/drug effects , Calcium Signaling/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Ion Channel Gating/drug effects , Male , Myocytes, Cardiac/drug effects , Rats , Rats, Sprague-Dawley , Treatment Outcome
13.
Toxicol Sci ; 141(2): 493-504, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25015662

ABSTRACT

Hydrogen sulphide (H2S), a chemical hazard in oil and gas production, has recently become a dreadful method of suicide, posing specific risks and challenges for the first responders. Currently, there is no proven effective treatment against H2S poisoning and its severe neurological, respiratory or cardiac after-effects. We have recently described that H2S is present in various compartments, or pools, in the body during sulphide exposure, which have different levels of toxicity. The general goals of our study were to (1) determine the concentrations and kinetics of the various pools of hydrogen sulphide in the blood, i.e., gaseous (CgH2S) versus total sulphide, i.e., reacting with monobromobimane (CMBBH2S), during and following H2S exposure in a small and large mammal and (2) establish the interaction between the pools of H2S and a methemoglobin (MetHb) solution or a high dose of hydroxocobalamin (HyCo). We found that CgH2S during and following H2S infusion was similar in sedated sheep and rats at any given rate of infusion/kg and provoked symptoms, i.e., hyperpnea and apnea, at the same CgH2S. After H2S administration was stopped, CgH2S disappeared within 1 min. CMBBH2S also dropped to 2-3µM, but remained above baseline levels for at least 30 min. Infusion of a MetHb solution during H2S infusion produced an immediate reduction in the free/soluble pool of H2S only, whereas CMBBH2S increased by severalfold. HyCo (70 mg/kg) also decreased the concentrations of free/soluble H2S to almost zero; CgH2S returned to pre-HyCo levels within a maximum of 20 min, if H2S infusion is maintained. These results are discussed in the context of a relevant scenario, wherein antidotes can only be administered after H2S exposure.


Subject(s)
Antidotes/administration & dosage , Hydrogen Sulfide/toxicity , Hydroxocobalamin/administration & dosage , Methemoglobin/administration & dosage , Poisoning/blood , Poisoning/drug therapy , Sulfides/toxicity , Animals , Female , Gases , Hydrogen Sulfide/blood , Hydrogen Sulfide/pharmacokinetics , Hydroxocobalamin/blood , Male , Methemoglobin/metabolism , Poisoning/etiology , Rats, Sprague-Dawley , Sheep , Sulfides/blood , Sulfides/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...