Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Nov 1.
Published in final edited form as: Ann N Y Acad Sci. 2020 May 6;1479(1):108–121. doi: 10.1111/nyas.14353

Antidotal effects of methylene blue against cyanide neurological toxicity: in vivo and in vitro studies

Philippe Haouzi 1, Marissa McCann 1, JuFang Wang 2, Xue-Qian Zhang 2, Jianliang Song 2, Ilker Sariyer 3, Diane Langford 3, Maryline Santerre 4, Nicole Tubbs 1, Annick Haouzi-Judenherc 5, Joseph Y Cheung 2,6
PMCID: PMC7644644  NIHMSID: NIHMS1606217  PMID: 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.

Keywords: cyanide, antidote, neurons, methylene blue

Introduction

The current strategy for treating acute cyanide (CN) intoxication relies on symptomatic measures, comprising ventilatory and circulatory support1,2 and specific antidotes.35 The latter can act through two different mechanisms1: by “scavenging” the pool of free CN in the blood, using, for instance, cobalt-containing molecules2,6,7 or nitrite-induced methemoglobinemia811-taking the advantage of the affinity of CN for “metals”-or2 by increasing CN elimination as thiocyanate, using sodium thiosulfate9,1214 or other sulfur donors, such as sulfanegen.15,16 These antidotes can be combined.

Methylene blue (MB) is a dye that was synthesized at the end of the 19th century.17,18 We have recently revisited the effects of MB, which was briefly proposed in the 1930s1922 as a treatment of CN intoxication, and found that this molecule exerts potent antidotal effects against CN toxicity through a unique modality of action that results from its distinctive cyclic redox properties.2325 These properties are currently used to treat the excessive formation of ferric iron in hemoglobin (methemoglobin), restoring the ability of hemoglobin to carry O2 in patients with methemoglobinemia.17,18,26,27 After being administrated, MB is immediately reduced into its leucoform, leucomethylene blue (LMB),28 by reducing agents present in blood and cells, such as NAD(P)H.29 LMB, on the other hand, is reoxidized into MB by molecules with a higher redox potential (such as O2 or most metallocompounds), giving up to two electrons in the process (Fig. 1), and a new cycle of reduction can be initiated.29 Diffusing rapidly into the cytoplasm and in the mitochondria of any cells, including neurons,30 the MB/LMB couple can restore the TCA cycle and the glycolytic activity by oxidizing NADH and decreasing the NADH/NAD ratio,31 rescuing the TCA cycle when inhibited by the accumulation of NADH, like for instance during an impediment of the electron chain activity by CN or H2S intoxication.25,28 The reestablishment of the TCA cycle can, in turn, lead to the production of ATP via succinyl-CoA synthase-the mitochondrial substrate-level phosphorylation31-independently of the integrity of the mitochondrial ATPase activity. Also, MB/LMB redox properties appear to be able to restore the mitochondrial membrane potential, depressed by CN, through mechanisms that are not fully understood.25,28 Indeed, recent studies have shown a decrease, by MB, of postanoxic brain injury,3236 as well as in various conditions associated with altered mitochondrial functions.3741 It has been proposed that LMB could interact directly with the mitochondrial electron chain,42 providing electrons to the complexes I and III and allowing a partial resuscitation of the mitochondrial electron chain. Although such a mechanism would be difficult to reconcile with the effects of CN intoxication, which result, if anything, in a reduction of the electron chain complexes, MB does restore the mitochondrial membrane potential and decrease the production of reactive O2 species in cells intoxicated by CN.24,25

Figure 1.

Figure 1.

(A) Structure of MB. (B) Redox reactions illustrating the reaction of MB and NADH, as an electron donor. The reduction of MB into its leucoform results in oxidation of NADH. LeucoMB is reoxidized, in the presence of O2, into MB producing a molecule of H2O (via the formation of H2O2).

These effects translate into a drastic decrease in mortality following MB administration after lethal CN intoxication in the rat.24,25 This increased survival is mediated through the rescue of cardiac contractility,23,43 an effect also observed with other mitochondrial poisons.28,44,45 Whether MB is also able to counteract or rescue the neurological manifestations of CN toxicity, which precede CN-induced cardiac arrest, remains an outstanding question. The acute neurological manifestations of CN intoxication clearly mimic those observed in hypoxia/anoxia, consisting of rapid coma and seizures with an early depression in medullary neurons producing apnea and gasping.46 Neurological long-term sequelae are rare, ranging from minor cognitive dysfunctions to profound motor (including extrapyramidal syndrome), visual or memory deficits, akin to the effects of postanoxic injury.47,48 We have recently observed that rats with neurological deficits after acute CN intoxication presented lesions that affected areas of the brain similar to those found after ischemia: diffuse lesions of the hippocampus, cerebellum, cortex, and subcortical nuclei.49 Lesions were only present in rats with a clinical deficit.

In this study, we sought (1) to determine if MB could alter or prevent CN-induced coma and apnea/gasping in awake rats, (2) to establish the doses of MB required to produce such an effect, and (3) to ascertain whether such a protective mechanism, if present in vivo, could be ascribed to a direct rescue of neurons by MB, that is, independently of any improvement in circulatory status. This was achieved by studying the effects of MB in two neuronal models: a primary culture of human fetal neurons or an established human SH-SY5Y neuroblastoma cell line exposed to CN toxicity, using concentrations of CN compatible with those observed in human intoxication (100 micromolar).

Materials and methods

In vivo experiments

Animal preparation.

A total of 32 male Sprague-Dawley rats (Charles River Laboratories) weighing 0.392 ± 109 kg were studied (see power analysis below). All experiments were conducted in accordance with the Guide for the Care and Use of Laboratory Animals, 8th Edition (the National Research Council (United States) Institute for Laboratory Animal Research). The Penn State Hershey Medical Center Institutional Animal Care and Use Committee approved the study. The rats were housed at the Animal Resource Services at the Penn State College of Medicine, which conforms to the requirements of the U.S. Department of Agriculture and the Department of Health, Education and Welfare. Rats were provided with food and water ad libitum, on a standard 12-h (7 AM-7 PM) light/dark schedule, under the direct supervision of veterinarians.

Solutions.

Potassium cyanide (KCN; Sigma, ref. no. 60178) solutions were prepared at the concentration of 0.5 mg/mL (dilution in sterile saline) in keeping with our previous report.46 We have previously observed that KCN infused in rats at the dose of 0.375 mg/kg/min would produce a sequence of symptoms that leads to depression in medullary neurons and death by cardiac arrest (CA) within less than 15 minutes.46 In addition, an infusion of potassium chloride (KCl) at the same concentrations of KCN46 had no measurable effects. MB (10 mg/mL, Akron) was diluted in sterile saline (5–1 mg/mL), to keep the volume of saline constant (2 mL) regardless of the dose of MB that was used.

Protocol and data analysis.

On the day of the study, rats were briefly sedated (3.5% isoflurane) for about 15 min to place a heparinized, double lumen, venous catheter in the dorsal vein of the tail. Two to three hours were given for full recovery before doing the study.

KCN was infused intravenously using a high-precision infusion pump (Fusion 100; Chemyx Inc; Stafford, TX) at the rate of 0.375 mg/kg/min for 13 minutes. The duration of exposure was selected based on the fact that shorter exposures, using the same rate of CN infusion, were unable to lead to a lethal outcome. More specifically, when 0.375 mg/kg for 7 min (n = 3, total dose 2.6 mg/kg) was used, the coma was always reversible. Also, exposure up to 10 min at the same rate (total dose 3.7 mg/kg) was not always lethal (one out of three animals). The total dose of 4.8 mg/kg (infused over 13 min) was always lethal and was, therefore, chosen for our present study.

Intravenous CN infusion allows reproducible and stereotyped sequences of symptoms consisting of (1) a short phase of locomotor agitation and hyperventilation followed by (2) a period of general weakness leading to (3) a coma, which we define by the loss of righting reflex, often associated with (4) seizing; the coma is rapidly followed by (5) a period of apneas (cessation of visible breathing activity for more than 3 seconds) and gasping (large breathing activity associated with grunting sounds) leading to (6) a CA defined by the disappearance of the perception of any cardiac pulsation1,46 and complete whitening of the pupils.

MB was infused 3 min after the onset of CN infusion, that is, when the first neurological symptoms developed (agitation, weakness, and ataxia). Three different doses were used in keeping with doses we have previously used in rats, that is, 4, 10, or 20 mg/kg24,25,28,44,45,50,51 for 5 minutes. The rationale for the doses of MB is developed in the Discussion section, and these doses, which correspond to a range of doses, currently recommended for the treatment of methemoglobinemia in humans,17,18 in keeping with allometric considerations.52 In addition, in a separate group of animals, MB was infused at the dose of 20 mg/kg, but over 10 minutes. Animals were monitored every 15 seconds for 20 min, then every minute for the following hour in the surviving rats. All surviving animals were examined and weighed every day for 2 weeks.

Sample size estimation was established as follows: Mortality was our primary outcome; since the model is always lethal, 80% survival in the MB-treated group would require five animals per group to identify a significant effect (alpha 0.05, power of 80%), while three animals per group would be enough if the treated group allowed 90% survival. The study was initially designed with five animals per group. Our secondary outcomes were the onset of each of the following symptoms: 1-agitation, 2-general weakness or ataxia, 3-coma, 4-seizures, 5-apnea and gasping, and 6-asystole. The duration of coma of the animals that survived the intoxication was determined. Data are presented as median and range. Comparison between groups was done using nonparametric statistics (Mann-Whitney U test for two-group comparisons).

Comparisons between the effects of the doses were performed using the Kruskal-Wallis test for multiple comparison and the Mann-Whitney U test for two-group comparisons. Statistical analyses were done with GraphPad Prism® 6 (Graphpad Software, La Jolla, CA). P < 0.05 was regarded as significant for any of these comparisons.

In vitro experiments

Action potential measurement in primary human fetal neuron cultures.

Human fetal neuron cultures were obtained from the Basic Science Core I of the Comprehensive NeuroAIDS Center at Temple University Lewis Katz School of Medicine. Neurons were isolated and cultured as described previously.53 Briefly, primary neurons were prepared directly from fetal human brains by differentiation. Fetal human brain cells were plated in culture dishes coated with laminin and poly-l-ornithine. Cells were differentiated into neurons by neuronal differentiation media consisting of neurobasal media supplemented with B27, glutamax, recombinant human brain-derived neurotrophic factor, glial-derived neurotrophic factor, and penicillin/streptomycin for up to 4 weeks. Neurons were characterized by microtubule-associated protein 2 and synaptophysin immunostaining. Neuronal differentiation and function were monitored electrophysiologically by multielectrode arrays measuring local field potentials weekly in culture. The medium was changed every 3 days.

Action potential (AP) was measured in neurons after 2 weeks in culture with whole-cell patch clamp. Fire-polished pipettes (tip diameter 2 μm) with resistances of 5–6 MΩ when filled with pipette solutions were used. Pipette solution consisted of (in mM) 125 KCl, 4 MgCl2, 0.06 CaCl2, 10 HEPES, 5 K+-EGTA, 3 Na2ATP, and 5 Na2-creatine phosphate (pH 7.2). The external solution consisted of (in mM) 132 NaCl, 5.4 KCl, 1.8 CaCl2, 1.8 MgCl2, 0.6 NaH2PO4, 7.5 HEPES, 7.5 Na+-HEPES, and 5 glucose, pH 7.4. Phosphate-buffered saline (PBS) or NaCN (100 μM) was added at time zero, and AP was measured at 10 minutes. In another group of neurons, MB was added 3 min after NaCN and AP was measured at 10 minutes. APs were recorded using the current clamp configuration at 1.5× threshold stimulus, 0.5-ms duration, and at 30 °C.5457

[Ca2+]i measurement in differentiated SH-SY5Y neuroblastoma cells.

SH-SY5Y cells were plated at the low confluence in a 1:1 mixture of Ham’s F12 and Dulbecco Modified Eagle Medium (DMEM) supplemented with 10% heat-inactivated fetal bovine serum (FBS) and 2 mM glutamine, in a humidified atmosphere of 5% CO2 in the air at 37 °C. After 24 h of cell plating, differentiation into neuron-like phenotype was triggered by exposure to 10 μM retinoic acid (RA) in DMEM containing 1% FBS for 10 days. DMEM-containing RA was changed every 3 days to replenish RA in culture media. Under these conditions, SH-SY5Y cells expressed the neuronal marker neuron-specific enolase by 4 days and persisted to 10 days in culture58 at which time cells were used for [Ca2+]i measurements. Fura-2-loaded (0.67 μM fura-2 AM, 15 min) differentiated SH-SY5Y cells on glass coverslips were incubated in media containing (in mM) 87 NaCl, 40 KCl, 1.25 CaCl2, 1.2 MgCl2, 1.2 KH2PO4, 20 HEPES, and 5.6 glucose, pH 7.4. Differentiated SH-SY5Y cells were exposed to excitation light (360 and 380 nm) only during data acquisition, and epifluorescence (510 nm) was recorded. PBS (CTL) or NaCN (100 μM) was added at time zero, and [Ca2+]i was measured at 0, 2, 5, and 10 minutes. In another group of neurons, MB was added 3 min after NaCN and [Ca2+]i was measured at same time points. Daily calibration of fura-2 signals to convert fura-2 fluorescence intensity ratios (F360/F380) into [Ca2+]i values was performed as previously described.5963

Data analysis.

All results are expressed as the means ± standard error of the mean. For the analysis of [Ca2+]i as a function of group (control,NaCN, and MB+NaCN and time), a two-wayANOVAwas used. For the analysis of AP parameters, a one-way ANOVA was used. A commercially available software package (JMPR® version 14 Pro, SAS Institute, Cary, NC) was used. In primary analyses, P-≤ 0.05 was taken to be statistically significant. In subgroup analyses, the Bonferroni correction (P-≤ 0.05/3 or P-≤ 0.0167 was deemed significant since there were three subgroup comparisons) was applied.

Results

Time course of the effects of CN in unanaesthetized rats (n = 12)

As illustrated in Figure 2, the infusion of CN produced a rapid change in behavior consisting of an increased locomotor activity, typically associated with a phase of hyperventilation, starting 194 seconds (median) after the onset of infusion (range 150–212 seconds). More than 1 min later, 238 seconds (range 239–306 seconds) into CN infusion, all animals started to present some difficulty walking, which led to a coma ~6 min into the exposure (386 seconds, range 328–461 seconds). This coma was followed by seizures at 403 seconds (range 310–555 seconds), and then all the animals became apneic, with a typical gasping pattern 516 seconds (range 401–623 seconds) into CN exposure. Cardiac pulsations disappeared with a clear whitening of the pupils at 783 seconds (range 706–946 seconds).

Figure 2.

Figure 2.

(A) The protocol used in the present study. (B) Effects of KCN infusion (0.375 mg/kg/min for 13 min) on the occurrence of the various symptoms of CN toxicity (median and range of onset of symptoms are shown). The signs of toxicity developed in a very reproducible manner in keeping with the level of CN starting with a phase of agitation followed by a period of weakness, coma, and seizures than a depression in breathing leading to cardiac asystole (CA). The intoxication was always lethal within 15 min in all animals.

Effects of MB 20, 10, and 4 mg/kg

Responses were similar at 20 mg/kg (n = 5) and 10 mg/kg (n = 5), showing a dramatic change in the neurological outcomes and survival. As displayed in Figure 3, none of the treated animals presented an apnea or a gasping pattern reflecting the absence of depression of medullary respiratory neurons at either dose. The time to coma was significantly delayed and of briefer duration at the highest dose of MB with a median of 359 and 737 seconds for 20 and 10 mg/kg MB, respectively. Twenty milligrams per kilogram of MB, whether infused over 10 or 5 min, produced the same effects. Individual data are shown with P values in Figure 3. All the animals survived, and none of them presented any specific issue with feeding or behavior after recovery from the coma, except for a somnolence that lasted for less than 2 hours. MB, at 4 mg/kg, still had a significant impact on the development of the neurological toxic effects of CN, and the animals survived the exposure (Fig. 3); however, the time to coma and apnea was significantly shorter than at the dose of 10 or 20 mg/kg (Fig. 4). The duration of coma was four times longer than for the highest dose of MB as shown in Figure 4. Also, in the group that received the lowest doses of MB, one animal displayed difficulty walking after the exposure that did not resolve within 24 h following the end of CN exposure, requiring the euthanasia of the animal. Finally, a transient, but significant, loss of weight was observed for 1 day following CN exposure and treatment, and then all the animals regained weight at a rate ranging from 3 to 6 g per day.

Figure 3.

Figure 3.

Effects of the different doses of MB, infused 3 min into CN exposure, on the symptoms of CN toxicity. Open boxes show the median and range of the effects in untreated animals (displayed in Fig. 1). Individual data are shown for each dose of MB. The effects of MB were similar at 10 and 20 mg/kg, whether infused for 5 or 10 minutes (A-C). None of the animals presented apnea and they all survived. The duration of coma was significantly delayed at either dose, but was, however, significantly longer following 10 mg/kg than after 20 mg/kg. Following 4 mg/kg (D), the effects of MB were also significantly different from untreated intoxication, but (1) one of the rats that survived, presented with difficulty walking, had to be euthanized within 24 h (red dot) and (2) all animals presented a period of apnea and gasping, while the duration of coma was significantly longer than at the highest doses of MB (untreated versus MB *P < 0.001, and &P < 0.05). CA, cardiac arrest.

Figure 4.

Figure 4.

Comparison of the effects of three doses of MB (20 mg/kg, administered over 10 or 5 minutes; 10 mg/kg; and 4 mg/kg) at the onset of coma (A), apnea (B), and seizures (C) along with the duration of coma (D). (*P < 0.001, $P < 0.01, and &P < 0.05).

Effects of NaCN on AP in human embryonic neurons and effects of MB

Compared with control human embryonic neurons treated with PBS, exposure to NaCN (100 μM) for 10min resulted in Em depolarization from −70.5 ± 0.7 to −59.1 ± 0.5 mV (P < 0.0001), reduction in AP amplitude from 108.6 ± 1.7 to 90.8 ± 1.0 mV (P < 0.0001), and prolongation of AP duration at 100% repolarization (APD100) from 1.78 ± 0.06 to 2.69 ± 0.15 ms (P = 0.0007) (Fig. 5). Compared with neurons exposed to NaCN, MB (20 μg/mL) added 3 min after NaCN significantly repolarized Em (P < 0.0001), increased AP amplitude (P = 0.0005), and shortened APD100 (P = 0.0014) (Fig. 5) so that there were no longer any significant differences when compared with AP parameters measured in control neurons (Em, P = 0.1708; AP amplitude, P = 0.0623; and APD100, P = 0.0427).

Figure 5.

Figure 5.

NaCN depolarizes resting membrane potential (Em), reduces action potential (AP) amplitude, and prolongs action potential duration (APD). These changes were rescued bymethylene blue (MB).AP parameters weremeasured in cultured human embryonic neurons by whole-cell patch clamp (see the materials and methods section). PBS (CTL) or NaCN (100 μM) was added at time zero, and AP wasmeasured at 10minutes. In another group of neurons,MB (20 μg/mL) was added 3 min after NaCN and action potential was measured at 10 minutes. Top: Representative AP tracings from neurons treated with PBS (CTL) (A), NaCN (B), and NaCN+MB (C). Bottom: Themeans ±SE of resting Em, APD at 100% repolarization (APD100), and AP amplitude from five CTL (D), seven NaCN (E), and six NaCN + MB (F) neurons (two human fetal brain preparations). *P ≤ 0.0007, CTL versus NaCN; and #P < 0.0015, NaCN versus NaHS + MB.

NaCN increases [Ca2+]i in differentiated SH-SY5Y neuroblastoma cells; effects of MB

Depolarization of differentiated SH-SY5Y cells by incubation in high K+ (40 mM) media did not result in increases in [Ca2+]i over 10 min of observation (Fig. 6). Addition of NaCN (100 μM) in high K+ media caused progressive increases in [Ca2+]i (P = 0.0095; group × time interaction effect) (Fig. 6). When added 3 min after NaCN, MB (20 μg/mL) prevented the [Ca2+]i elevation observed in neurons exposed to NaCN alone (P = 0.0170; group × time interaction effect) such that there were no longer any differences in the time courses of [Ca2+]i between control and NaCN + MB neurons (P = 0.2644; group × time interaction effect) (Fig. 6).

Figure 6.

Figure 6.

Methylene blue reverses [Ca2+]i elevation in neurons exposed to NaCN. [Ca2+]i was measured in fura-2-loaded neurons in 40 mM K+ media (Methods). PBS (CTL) or NaCN (100 μM) was added at time zero, and fura-2 signals were measured at times indicated. In another group of neurons, MB (20μg/mL) was added 3 min after NaCN. Time courses of [Ca2+]i in neurons exposed to PBS (CTL, open circles), NaCN (closed circles), NaCN + MB (open square), (n = 3) are shown.

Discussion

This study has established for the first time that MB is able to mitigate the neurological toxicity of CN not only in vivo but also in vitro. In unanaesthetized rats, 20 or 10 mg/kg of MB was able to antagonize CN neurological toxicity by not only reducing the time and severity (duration) of coma but also by preventing the life-threatening depression in medullary respiratory neurons in an extremely lethal form of intravenous CN intoxication. All the treated animals survived with no sequelae in an otherwise fatal exposure. To determine whether the protection by MB against CN neurotoxicity in vivo was mediated by a direct protective effect on neurons, rather than or in addition, to a restoration of systemic circulation,23 two neuronal models were used: a primary culture of human fetal neurons or an established human SH-SY5Y neuroblastoma cell line. Since SH-SY5Y neuroblastoma cells resemble immature sympathetic neuroblasts in culture, SH-SY5Y cells were differentiated into more mature dopaminergic neuron-like cells58 before subjecting them to our experimental protocols: MB administered several minutes after CN exposure did restore normal neuronal activity of neurons intoxicated with CN.

Methodological considerations and the question of the doses

We used KCN, as in our previous studies,24,25,46 following many other studies that used KCN to produce lethal or sublethal CN intoxication.2,64 We have previously established that KCl infused at the same rate as KCN in the present study, with equimolar concentrations of potassium salt, had no effect on the concentration of potassium in the blood.46 Perhaps, more importantly, KCl infusion was unable to reproduce any of the effects of KCN on circulation or breathing.

The timing of the antidote injection was dictated by the time course of the development of the symptoms and the time to death. It is possible that a benefit could be observed even at the lowest dose (4 mg/kg) MB, which showed less efficacy in the rapidly lethal scenario created by our protocol.

The doses of MB that are currently used in humans for the treatment of methemoglobinemia are about 1–2 mg/kg to be repeated, if needed.17,18 According to the recommendations for the conversion of doses between animals and humans provided in the The Guidance for Industry: Estimating the Maximum Safe Starting Dose in Initial Clinical Trials for Therapeutics, published by the Department of Health and Human Services and the FDA,65 a dose of 4 mg/kg in humans, corresponding to about 150 mg/m2, would, therefore, be equivalent to a dose of about 20 mg/kg in a rat, while 10 mg/kg will correspond to the lowest dose currently used in the clinical setting in humans. In terms of toxicity, a deficit in G6PD is regarded as a classical contraindication to the utilization of MB;18 however, MB appears to be very safe in children presenting a deficit in G6PD,66 as well as in adults with nonsymptomatic G6PD deficit.67,68 Although MB has no serotonin “toxicity” by itself, MB can also interact with selective serotonin reuptake inhibitors (SSRIs).69 The incidence of “serotonin syndrome” in patients treated by SSRI agents receiving MB appears, however, to be relatively low.69,70 Based on the present data, it seems reasonable to assume that the doses currently used for the treatment of methemoglobinemia will be effective against life-threatening CN intoxication. Of note, MB, at the lowest doses of 4 mg/kg (corresponding to less than 1 mg/kg in humans), also shows efficacy, although less potent than at 10 or 20 milligrams per kilogram. This makes possible the use of small doses of MB in combination with other families of antidotes at levels wherein no toxicity related to potential oxidative stress is to be expected.

The in vivo model

The effects of MB significantly delayed and shortened, in a dose-dependent manner, the onset of coma and seizures, while preventing the potentially lethal depression of medullary neurons produced by CN. The latter, akin to the effects of hypoxia7174 or other mitochondrial poisons,75 has been shown to occur before a depression in cardiac contractility develops. Gasping and apnea were easy to clinically identify and distinguish from a regular breathing pattern. Of note while at 20 mg/kg, breathing was not affected, at 10 mg/kg, a very transient episode of apnea was observed in one animal, which was still delayed when compared with all untreated animals. At 4 mg/kg, all the animals had episodes of apneas that were not sustained preventing the animals to die. None of these animals presented a cardiac arrest, in striking contrast with the untreated group wherein all the animals died before or within a few minutes following the end of CN exposure (Fig. 2). All the treated animals recovered from their coma following 10 and 20 mg/kg with no sequelae. This was also true at the dose of 4 mg/kg, but for one animal that presented difficulty walking and had to be euthanized. This demonstrates a clear dose-effect of MB with maximal efficacy at 20 milligrams per kilogram. Whether higher doses, which may be associated with oxidative, and thus some toxic, effects of MB could be even more effective remains to be determined.

The in vitro model

The concentration (100 μM) of CN chosen for the present study was 1–2 orders of magnitude lower than that used in previous studies.76,78 The reason for selecting this CN dose was twofold. First, our CN concentration utilized corresponded to a realistic level of intoxication compatible with survival. For instance, Baud et al.79 have shown that clinically toxic effects were associated with CN concentrations between 40 and 100 μM in victims of smoke inhalation, while concentrations >100 μM were potentially lethal. In both mouse80 and rabbit7 models of CN intoxication, peak blood CN concentrations (80–120 μM) achieved with CN doses that produced 50% mortality (LD50) were similar to those observed in humans. Similarly, in a swine study in which 4% KCN was infused intravenously to effect a 50% reduction in baseline mean arterial pressure, the blood CN level achieved was 3.6 ± 0.7 μg/mL or ~140 μM,81 similar to the concentration of CN used in the present study. Second, at this CN dose, the depressed cardiac contractility could be rescued by antidotes: by hydroxocobalamin in vivo81 or MB in vitro.23

Our observation that NaCN (100 μM) depolarized human fetal neurons in primary culture is similar to that observed in rat hippocampal slices in which NaCN (1–2 mM) resulted in rapid depolarization.76,78 By contrast, in isolated rat hippocampal neurons in which the fast Na+ current was blocked by tetrodotoxin, NaCN (2 mM) induced hyperpolarization, rather than depolarization of the membrane potential.77 These considerations suggest that the responses to CN by our human fetal neurons in primary culture, in the absence of tetrodotoxin, were similar to those observed in freshly isolated neuronal tissues. One major finding is that NaCN exposure not only resulted in membrane depolarization but also reduction in AP amplitude and prolonged repolarization: all these adverse effects may conspire to render the neurons more excitable and impair nervous impulse conduction, accounting for the coma and depression in breathing as well as the early seizures consistently observed with CN. Another major finding is that MB was efficacious in reversing the deleterious effects of NaCN on neuron AP morphology; thereby providing the evidence for a direct cellular protective effect against NaCN neurotoxicity.

We used another neuronal model (established SH-SY5Y neuroblastoma cell line but differentiated into neuronal phenotype) to evaluate if MB exerted protection against CN neurotoxicity. Our baseline [Ca2+]i values of 30–60 nM in differentiated SH-SY5Y cells are similar to those reported for neonatal rat hippocampal neurons (50–130 nM).82 NaCN (100 μM) did not change [Ca2+]i in differentiated SH-SY5Y cells (data not shown), in agreement with the observations in neonatal rat hippocampus neurons82 and primary rat cerebellar granule cells.83 We, therefore, attempted to increase Ca2+ influx by incubating SH-SY5Y cells in high K+ media. Surprisingly, depolarizing the membrane with a high K+ solution did not increase [Ca2+]i in differentiated SH-SY5Y cells, in contrast to that observed in primary culture of rat cerebellar granule cells.83 Lack of [Ca2+]i increase may be due to more effective Ca2+ extrusion and Ca2+ uptake into intracellular organelles in differentiated SH-SY5Y cells. Interestingly, exposure to NaCN (100 μM) in high K+ medium resulted in significant increases in [Ca2+]i over 10 min; an effect that was reversed by MB added 3 min after NaCN. These data indicate that MB was efficacious in normalizing ion homeostasis in CN-intoxicated neurons.

Mechanisms of action of MB during CN intoxication

First, one should acknowledge that the mechanisms of CN toxicity are multifarious. Although signs of CN intoxication are usually attributed to the combination of ion CN with the mitochondrial complex IV (cytochrome c oxidase (CCO)), in turn preventing its reoxidation by O2,84 the mechanisms of CN toxicity are still poorly understood. As a consequence of the complex IV-CN interaction, the mitochondrial complexes upstream to CCO are all maintained in a reduced state and are unable to transfer protons across the inner mitochondrial membrane leading to a rapid depression of the membrane potential, preventing, in turn, mitochondrial ATP synthesis.85 The corollary reduction in the rate of O2 utilization and production of reactive O2 species, by complex I or III, are considered as the hallmarks of CN poisoning. In addition, since NADH is not oxidized anymore, by an already reduced mitochondrial complex I, the NADH/NAD ratio increases and virtually stops the TCA cycle.86,87 This, in turn, suppresses ATP synthesis via mitochondrial substrate-level phosphorylation (TCA cycle). The rise in NADH/NAD ratio also catalyzes the transformation of pyruvate into lactate in the cytoplasm.88 We have recently suggested that the manifestations of CN intoxication are not directly caused by a decrease in ATP concentration-at least in the forms where victims can survive-as a rapid decrease in ATP turn over and consumption occurs balancing any reduction in ATP production.23 This is well exemplified for CN cardiac toxicity, wherein CN toxicity8994 produces a reduction in cardiac contractility93 via reversible inhibition of LCa channel activity, reducing in turn ATP needed for contractions.23 A profound alteration of K+ channels95 is also present. We have recently found that they result from abnormal phosphorylation of L-type Ca2+ channel subunit α1c at T96, Na+-K+-ATPase subunit α2-peptide at S461 and S464, and voltage-gated K+ channel subfamily KQT member 2 at S429.43 We found that all these changes that could lead to a fatal outcome were rescued by MB.43

MB, a very potent redox compound, can oppose CN toxicity via different mechanisms.24,25 We have recently proposed that MB acts in vivo by increasing/restoring the NAD/NADH ratio and the activity of the TCA cycle, as well as by trapping CN on hemoglobin.23,24 The latter appears to rely on the cyclic oxidative and reducing effects of the couple MB/LMB on iron. A similar mechanism could take place at the level of the electron chain complexes upstream to the complex IV, which may account for the restoration of the mitochondrial function. We have suggested that molecules of hydrogen peroxide produced during the reoxidation of LMB by O2,96,97 before being “transformed” into H2O by a large amount of catalase present in cells, could oxidize various metalloproteins98,99 of the electron chain complexes “stuck” in a reduced state.24,25 Such reoxidation could restore the capacity of the electron chain to be reduced again (by LMB or NADH). Of note, MB appears to be effective not only against CN23,24 but also in intoxications affecting mitochondrial functions, including H2S,45,51 sodium azide,100 and rotenone.101

Conclusions and perspectives

In a lethal model of CN intoxication, we found that MB shows antidotal properties against CN intoxication-induced coma, seizure, and the lethal depression in breathing in a dose-dependent manner (from 4 to 20 mg/kg). The doses of MB used in this study are commensurate with those proposed in humans and currently used for the treatment of methemoglobinemia. Our in vitro data provide direct support for our premises; that is, MB directly counteracts the toxicity of CN-exposed neurons, restoring normal AP morphology, resting potential, and intracellular Ca2+ homeostasis. Efficacy studies must be confirmed in other species, and certainly in larger mammals, in keeping with the FDA animal rules and the issues related to the specificity of the response of rodents to hypoxic stress. Finally, due to the necessity of a rapid intervention and a high number of victims potentially involved, the efficacy of the intraosseous route for administering MB should be investigated.

Acknowledgments

This work was supported in part by the National Institutes of Health Grants RO1-HL123093, RO1-HL137426, UO1-NS097162, and 1R21NS110549-01. Core facilities were provided by Comprehensive NeuroAIDS Center P30-MH92177 at Temple University.

Footnotes

Competing interests

The authors declare no competing interests.

References

  • 1.Judenherc-Haouzi A, Sonobe T, Bebarta VS & Haouzi P. 2018. On the efficacy of cardio-pulmonary resuscitation and epinephrine following cyanide- and H2S intoxication-induced cardiac asystole. Cardiovasc. Toxicol 18: 436–449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Bebarta VS, Pitotti RL, Dixon PS, et al. 2012. Hydroxocobalamin and epinephrine both improve survival in a swine model of cyanide-induced cardiac arrest. Ann. Emerg. Med 60: 415–422. [DOI] [PubMed] [Google Scholar]
  • 3.Baud FJ 2007. Cyanide: critical issues in diagnosis and treatment. Hum. Exp. Toxicol 26: 191–201. [DOI] [PubMed] [Google Scholar]
  • 4.Borron SW & Baud FJ. 2012. Antidotes for acute cyanide poisoning. Curr. Pharm. Biotechnol 13: 1940–1948. [DOI] [PubMed] [Google Scholar]
  • 5.Bebarta VS 2013. Antidotes for cyanide poisoning. Eur. J. Emerg. Med 20: 65–66. [DOI] [PubMed] [Google Scholar]
  • 6.Borron SW, Baud FJ, Megarbane B & Bismuth C. 2007. Hydroxocobalamin for severe acute cyanide poisoning by ingestion or inhalation. Am. J. Emerg. Med 25: 551–558. [DOI] [PubMed] [Google Scholar]
  • 7.Lee J, Mahon SB, Mukai D, et al. 2016. The vitamin B12 analog cobinamide is an effective antidote for oral cyanide poisoning. J. Med. Toxicol 12: 370–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Baskin SI, Horowitz AM & Nealley EW. 1992. The antidotal action of sodium nitrite and sodium thiosulfate against cyanide poisoning. J. Clin. Pharmacol 32: 368–375. [DOI] [PubMed] [Google Scholar]
  • 9.Bebarta VS, Brittain M, Chan A, et al. 2017. Sodium nitrite and sodium thiosulfate are effective against acute cyanide poisoning when administered by intramuscular injection. Ann. Emerg. Med 69: 718–725.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Cambal LK, Swanson MR, Yuan Q, et al. 2011. Acute, sublethal cyanide poisoning in mice is ameliorated by nitrite alone: complications arising from concomitant administration of nitrite and thiosulfate as an antidotal combination. Chem. Res. Toxicol 24: 1104–1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Cambal LK, Weitz AC, Li HH, et al. 2013. Comparison of the relative propensities of isoamyl nitrite and sodium nitrite to ameliorate acute cyanide poisoning in mice and a novel antidotal effect arising from anesthetics. Chem. Res. Toxicol 26: 828–836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hall AH, Dart R & Bogdan G. 2007. Sodium thiosulfate or hydroxocobalamin for the empiric treatment of cyanide poisoning? Ann. Emerg. Med 49: 806–813. [DOI] [PubMed] [Google Scholar]
  • 13.Hall AH, Doutre WH, Ludden T, et al. 1987. Nitrite/thiosulfate treated acute cyanide poisoning: estimated kinetics after antidote. J. Toxicol. Clin. Toxicol 25: 121–133. [DOI] [PubMed] [Google Scholar]
  • 14.Hall AH & Rumack BH. 1987. Hydroxycobalamin/sodium thiosulfate as a cyanide antidote. J. Emerg. Med 5: 115–121. [DOI] [PubMed] [Google Scholar]
  • 15.Patterson SE, Moeller B, Nagasawa HT, et al. 2016. Development of sulfanegen for mass cyanide casualties. Ann. N.Y. Acad. Sci 1374: 202–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Patterson SE, Monteil AR, Cohen JF, et al. 2013. Cyanide antidotes for mass casualties: water-soluble salts of the dithiane (sulfanegen) from 3-mercaptopyruvate for intramuscular administration. J. Med. Chem 56: 1346–1349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ginimuge PR & Jyothi SD. 2010. Methylene blue: revisited. J. Anaesthesiol. Clin. Pharmacol 26: 517–520. [PMC free article] [PubMed] [Google Scholar]
  • 18.Clifton J 2nd & Leikin JB. 2003. Methylene blue. Am. J. Ther 10: 289–291. [DOI] [PubMed] [Google Scholar]
  • 19.Geiger JC 1933. Methylene blue solutions in potassium cyanide poisoning - report on cases 2 and 3. J. Am. Med. Assoc 101: 269. [Google Scholar]
  • 20.Geiger JC 1933. Methylene blue as antidote for cyanide and carbon monoxide poisoning. J. Am. Med. Assoc 100: 59. [Google Scholar]
  • 21.Hanzlik PJ 1933. Methylene blue as antidote for cyanide poisoning. J. Am. Med. Assoc 100: 357. [PMC free article] [PubMed] [Google Scholar]
  • 22.Moldenhauer M 1933. Methylene blue as antidote for cyanide and carbon monoxide poisoning. J. Am. Med. Assoc 100: 59. [Google Scholar]
  • 23.Cheung JY, Wang J, Zhang XQ, et al. 2018. Methylene blue counteracts cyanide cardiotoxicity: cellular mechanisms. J. Appl. Physiol 124: 1164–1176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Haouzi P, Gueguinou M, Sonobe T, et al. 2018. Revisiting the physiological effects of methylene blue as a treatment of cyanide intoxication. Clin. Toxicol. (Phila.) 56: 828–840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Haouzi P, McCann M, Tubbs N, et al. 2019. Antidotal effects of the phenothiazine chromophore methylene blue following cyanide intoxication. Toxicol. Sci 170: 82–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Wright RO, Lewander WJ & Woolf AD. 1999. Methemoglobinemia: etiology, pharmacology, and clinical management. Ann. Emerg. Med 34: 646–656. [DOI] [PubMed] [Google Scholar]
  • 27.Mayer B, Brunner F & Schmidt K. 1993. Novel actions of methylene blue. Eur. Heart J 14(Suppl. I): 22–26. [PubMed] [Google Scholar]
  • 28.Haouzi P, Tubbs N, Cheung J & Judenherc-Haouzi A. 2019. Methylene blue administration during and after life-threatening intoxication by hydrogen sulfide: efficacy studies in adult sheep and mechanisms of action. Toxicol. Sci 168: 443–459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Buchholz K, Schirmer RH, Eubel JK, et al. 2008. Interactions of methylene blue with human disulfide reductases and their orthologues from Plasmodium falciparum. Antimicrob. Agents Chemother 52: 183–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Peter C, Hongwan D, Kupfer A & Lauterburg BH. 2000. Pharmacokinetics and organ distribution of intravenous and oral methylene blue. Eur. J. Clin. Pharmacol 56: 247–250. [DOI] [PubMed] [Google Scholar]
  • 31.Komlodi T & Tretter L. 2017. Methylene blue stimulates substrate-level phosphorylation catalysed by succinyl-CoA ligase in the citric acid cycle. Neuropharmacology 123: 287–298. [DOI] [PubMed] [Google Scholar]
  • 32.Martijn C & Wiklund L. 2010. Effect of methylene blue on the genomic response to reperfusion injury induced by cardiac arrest and cardiopulmonary resuscitation in porcine brain. BMC Med. Genomics 3: 27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Miclescu A, Basu S & Wiklund L. 2006. Methylene blue added to a hypertonic-hyperoncotic solution increases short-term survival in experimental cardiac arrest. Crit. Care Med 34: 2806–2813. [DOI] [PubMed] [Google Scholar]
  • 34.Miclescu A, Basu S & Wiklund L. 2007. Cardio-cerebral and metabolic effects of methylene blue in hypertonic sodium lactate during experimental cardiopulmonary resuscitation. Resuscitation 75: 88–97. [DOI] [PubMed] [Google Scholar]
  • 35.Wiklund L, Basu S, Miclescu A, et al. 2007. Neuro- and cardioprotective effects of blockade of nitric oxide action by administration of methylene blue. Ann. N.Y. Acad. Sci 1122: 231–244. [DOI] [PubMed] [Google Scholar]
  • 36.Wiklund L, Zoerner F, Semenas E, et al. 2013. Improved neuroprotective effect of methylene blue with hypothermia after porcine cardiac arrest. Acta Anaesthesiol. Scand 57: 1073–1082. [DOI] [PubMed] [Google Scholar]
  • 37.Lin AL, Poteet E, Du F, et al. 2012. Methylene blue as a cerebral metabolic and hemodynamic enhancer. PLoS One 7: e46585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Poteet E, Winters A, Yan LJ, et al. 2012. Neuroprotective actions of methylene blue and its derivatives. PLoS One 7: e48279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Atamna H & Kumar R. 2010. Protective role of methylene blue in Alzheimer’s disease via mitochondria and cytochrome c oxidase. J. Alzheimers Dis 20(Suppl. 2): S439–S452. [DOI] [PubMed] [Google Scholar]
  • 40.Atamna H, Nguyen A, Schultz C, et al. 2008. Methylene blue delays cellular senescence and enhances key mitochondrial biochemical pathways. FASEB J. 22: 703–712. [DOI] [PubMed] [Google Scholar]
  • 41.Daudt DR 3rd, Mueller B, Park YH, et al. 2012. Methylene blue protects primary rat retinal ganglion cells from cellular senescence. Invest. Ophthalmol. Vis. Sci 53: 4657–4667. [DOI] [PubMed] [Google Scholar]
  • 42.Rojas JC, Bruchey AK & Gonzalez-Lima F. 2012. Neurometabolic mechanisms for memory enhancement and neuroprotection of methylene blue. Prog. Neurobiol 96: 32–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Cheung JY, Merali S, Wang J, et al. 2019. The central role of protein kinase C epsilon in cyanide cardiotoxicity and its treatment. Toxicol. Sci 10.1093/toxsci/kfz137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Sonobe T & Haouzi P. 2015. H2S induced coma and cardiogenic shock in the rat: effects of phenothiazinium chromophores. Clin. Toxicol. (Phila.) 53: 525–539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Judenherc-Haouzi A, Zhang XQ, Sonobe T, et al. 2016. Methylene blue counteracts H2S toxicity-induced cardiac depression by restoring L-type Ca channel activity. Am. J. Physiol. Regul. Integr. Comp. Physiol 310: R1030–R1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Haouzi P, Tubbs N, Rannals MD, et al. 2017. Circulatory failure during noninhaled forms of cyanide intoxication. Shock 47: 352–362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Grandas F, Artieda J & Obeso JA. 1989. Clinical and CT scan findings in a case of cyanide intoxication. Mov. Disord 4: 188–193. [DOI] [PubMed] [Google Scholar]
  • 48.Borgohain R, Singh AK, Radhakrishna H, et al. 1995. Delayed onset generalised dystonia after cyanide poisoning. Clin. Neurol. Neurosurg 97: 213–215. [DOI] [PubMed] [Google Scholar]
  • 49.Rosenow F, Herholz K, Lanfermann H, et al. 1995. Neurological sequelae of cyanide intoxication-the patterns of clinical, magnetic resonance imaging, and positron emission tomography findings. Ann. Neurol 38: 825–828. [DOI] [PubMed] [Google Scholar]
  • 50.Haouzi P, Sonobe T & Judenherc-Haouzi A. 2020. Hydrogen sulfide intoxication induced brain injury and methylene blue. Neurobiol. Dis 133: 104474. [DOI] [PubMed] [Google Scholar]
  • 51.Sonobe T, Chenuel B, Cooper TK & Haouzi P. 2015. Immediate and long-term outcome of acute H2S intoxication induced coma in unanesthetized rats: effects of methylene blue. PLoS One 10: e0131340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Nair AB & Jacob S. 2016. A simple practice guide for dose conversion between animals and human. J. Basic Clin. Pharm 7: 27–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Donadoni M, Cicalese S, Sarkar DK, et al. 2019. Alcohol exposure alters pre-mRNA splicing of antiapoptotic Mcl-1L isoform and induces apoptosis in neural progenitors and immature neurons. Cell Death Dis. 10: 447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Tadros GM, Zhang XQ, Song J, et al. 2002. Effects of Na+/Ca2+ exchanger downregulation on contractility and [Ca2+]i transients in adult rat myocytes. Am. J. Physiol. Heart Circ. Physiol 283: H1616–H1626. [DOI] [PubMed] [Google Scholar]
  • 55.Tucker AL, Song J, Zhang XQ, et al. 2006. Altered contractility and [Ca2+]i homeostasis in phospholemman-deficient murine myocytes: role of Na+/Ca2+ exchange. Am. J. Physiol. Heart Circ. Physiol 291: H2199–H2209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Zhang XQ, Qureshi A, Song J, et al. 2003. Phospholemman modulates Na+/Ca2+ exchange in adult rat cardiac myocytes. Am. J. Physiol. Heart Circ. Physiol 284: H225–H233. [DOI] [PubMed] [Google Scholar]
  • 57.Zhang XQ, Zhang LQ, Palmer BM, et al. 2001. Sprint training shortens prolonged action potential duration in postinfarction rat myocyte: mechanisms. J. Appl. Physiol 90: 1720–1728. [DOI] [PubMed] [Google Scholar]
  • 58.Lopes FM, Schroder R, da Frota ML Jr., et al. 2010. Comparison between proliferative and neuron-like SH-SY5Y cells as an in vitro model for Parkinson disease studies. Brain Res. 1337: 85–94. [DOI] [PubMed] [Google Scholar]
  • 59.Song J, Zhang XQ, Wang J, et al. 2008. Regulation of cardiac myocyte contractility by phospholemman: Na+/Ca2+ exchange versus Na+-K+-ATPase. Am. J. Physiol. Heart Circ. Physiol 295: H1615–H1625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Song J, Gao E, Wang J, et al. 2012. Constitutive overexpression of phospholemman S68E mutant results in arrhythmias, early mortality and heart failure: potential involvement of Na+/Ca2+ exchanger. Am. J. Physiol. Heart Circ. Physiol 302: H770–H781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Wang J, Gao E, Song J, et al. 2010. Phospholemman and beta-adrenergic stimulation in the heart. Am. J. Physiol. Heart Circ. Physiol 298: H807–H815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Wang J, Chan TO, Zhang XQ, et al. 2009. Induced overexpression of Na+/Ca2+ exchanger transgene: altered myocyte contractility, [Ca2+]i transients, SR Ca2+ contents and action potential duration. Am. J. Physiol. Heart Circ. Physiol 297: H590–H601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Wang J, Gao E, Rabinowitz J, et al. 2011. Regulation of in vivo cardiac contractility by phospholemman: role of Na+/Ca2+ exchange. Am. J. Physiol. Heart Circ. Physiol 300: H859–H868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Bebarta VS, Pitotti RL, Dixon P, et al. 2012. Hydroxocobalamin versus sodium thiosulfate for the treatment of acute cyanide toxicity in a swine (Sus scrofa) model. Ann. Emerg. Med 59: 532–539. [DOI] [PubMed] [Google Scholar]
  • 65.U.S. Department of Health and Human Services FaDA. 2005. Guidance for industry estimating the maximum safe starting dose in initial clinical trials for therapeutics in adult healthy volunteers. https://www.federalregister.gov/d/05-14456. [Google Scholar]
  • 66.Meissner PE, Mandi G, Witte S, et al. 2005. Safety of the methylene blue plus chloroquine combination in the treatment of uncomplicated falciparum malaria in young children of Burkina Faso [ISRCTN27290841]. Malar. J 4: 45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Mandi G, Witte S, Meissner P, et al. 2005. Safety of the combination of chloroquine and methylene blue in healthy adult men with G6PD deficiency from rural Burkina Faso. Trop. Med. Int. Health 10: 32–38. [DOI] [PubMed] [Google Scholar]
  • 68.Muller O, Meissner P & Mansmann U. 2012. Glucose-6-phosphate dehydrogenase deficiency and safety of methylene blue. Drug Saf. 35: 85; author reply −6. [DOI] [PubMed] [Google Scholar]
  • 69.Ng BK & Cameron AJ. 2010. The role of methylene blue in serotonin syndrome: a systematic review. Psychosomatics 51: 194–200. [DOI] [PubMed] [Google Scholar]
  • 70.Grubb KJ, Kennedy JL, Bergin JD, et al. 2012. The role of methylene blue in serotonin syndrome following cardiac transplantation: a case report and review of the literature. J. Thorac. Cardiovasc. Surg 144: e113–e116. [DOI] [PubMed] [Google Scholar]
  • 71.Duffin J 2003. A commentary on eupnoea and gasping. Respir. Physiol. Neurobiol 139: 105–111. [DOI] [PubMed] [Google Scholar]
  • 72.Guntheroth WG & Kawabori I. 1975. Hypoxic apnea and gasping. J. Clin. Invest 56: 1371–1377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.St John WM 2009. Noeud vital for breathing in the brainstem: gasping-yes, eupnoea-doubtful. Philos. Trans. R. Soc. Lond. B Biol. Sci 364: 2625–2633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.St John WM & Bartlett D Jr. 1981. Comparison of phrenic motoneuron activity during eupnea and gasping. J. Appl. Physiol 50: 994–998. [DOI] [PubMed] [Google Scholar]
  • 75.Haouzi P 2012. Ventilatory and metabolic effects of exogenous hydrogen sulfide. Respir. Physiol. Neurobiol 184: 170–177. [DOI] [PubMed] [Google Scholar]
  • 76.Reiner PB, Laycock AG & Doll CJ. 1990. A pharmacological model of ischemia in the hippocampal slice. Neurosci. Lett 119: 175–178. [DOI] [PubMed] [Google Scholar]
  • 77.Nowicky AV & Duchen MR. 1998. Changes in [Ca2+]i and membrane currents during impaired mitochondrial metabolism in dissociated rat hippocampal neurons. J. Physiol 507: 131–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Englund M, Hyllienmark L & Brismar T. 2001. Chemical hypoxia in hippocampal pyramidal cells affects membrane potential differentially depending on resting potential. Neuroscience 106: 89–94. [DOI] [PubMed] [Google Scholar]
  • 79.Baud FJ, Barriot P, Toffis V, et al. 1991. Elevated blood cyanide concentrations in victims of smoke inhalation. N. Engl. J. Med 325: 1761–1766. [DOI] [PubMed] [Google Scholar]
  • 80.Ahmed AE & Patel K. 1981. Acrylonitrile: in vivo metabolism in rats and mice. Drug Metab. Dispos 9: 219–222. [PubMed] [Google Scholar]
  • 81.Bebarta VS, Pitotti RL, Boudreau S & Tanen DA. 2014. Intraosseous versus intravenous infusion of hydroxocobalamin for the treatment of acute severe cyanide toxicity in a Swine model. Acad. Emerg. Med 21: 1203–1211. [DOI] [PubMed] [Google Scholar]
  • 82.Dubinsky JM & Rothman SM. 1991. Intracellular calcium concentrations during “chemical hypoxia” and excitotoxic neuronal injury. J. Neurosci 11: 2545–2551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Sun P, Rane SG, Gunasekar PG, et al. 1997. Modulation of the NMDA receptor by cyanide: enhancement of receptor-mediated responses. J. Pharmacol. Exp. Ther 280: 1341–1348. [PubMed] [Google Scholar]
  • 84.Petersen LC 1977. The effect of inhibitors on the oxygen kinetics of cytochrome c oxidase. Biochim. Biophys. Acta 460: 299–307. [DOI] [PubMed] [Google Scholar]
  • 85.Kim HJ, Khalimonchuk O, Smith PM & Winge DR. 2012. Structure, function, and assembly of heme centers in mitochondrial respiratory complexes. Biochim. Biophys. Acta 1823: 1604–1616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.LaNoue KF, Bryla J & Williamson JR. 1972. Feedback interactions in the control of citric acid cycle activity in rat heart mitochondria. J. Biol. Chem 247: 667–679. [PubMed] [Google Scholar]
  • 87.Liu Y, Hu L, Ma T, et al. 2018. Insights into the inhibitory mechanisms of NADH on the alphagamma heterodimer of human NAD-dependent isocitrate dehydrogenase. Sci. Rep 8: 3146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Burgner JW 2nd & Ray WJ Jr. 1984. On the origin of the lactate dehydrogenase induced rate effect. Biochemistry 23: 3636–3648. [DOI] [PubMed] [Google Scholar]
  • 89.Allen DG & Orchard CH. 1983. Intracellular calcium concentration during hypoxia and metabolic inhibition in mammalian ventricular muscle. J. Physiol 339: 107–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Eisner DA, Nichols CG, O’Neill SC, et al. 1989. The effects of metabolic inhibition on intracellular calcium and pH in isolated rat ventricular cells. J. Physiol 411: 393–418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Jose AD & Stitt F. 1969. Effects of hypoxia and metabolic inhibitors on the intrinsic heart rate and myocardial contractility in dogs. Circ. Res 25: 53–66. [DOI] [PubMed] [Google Scholar]
  • 92.Suzuki T 1968. Ultrastructural changes of heart muscle in cyanide poisoning. Tohoku J. Exp. Med 95: 271–287. [DOI] [PubMed] [Google Scholar]
  • 93.Elliott AC, Smith GL, Eisner DA & Allen DG. 1992. Metabolic changes during ischaemia and their role in contractile failure in isolated ferret hearts. J. Physiol 454: 467–490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Pham JC, Huang DT, McGeorge FT & Rivers EP. 2007. Clarification of cyanide’s effect on oxygen transport characteristics in a canine model. Emerg. Med. J 24: 152–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Ito H, Nakajima T, Takikawa R, et al. 1991. Coenzyme Q10 attenuates cyanide-activation of the ATP-sensitive K+ channel current in single cardiac myocytes of the guineapig. Naunyn Schmiedebergs Arch. Pharmacol 344: 133–136. [DOI] [PubMed] [Google Scholar]
  • 96.Wainwright M & Amaral L. 2005. The phenothiazinium chromophore and the evolution of antimalarial drugs. Trop. Med. Int. Health 10: 501–511. [DOI] [PubMed] [Google Scholar]
  • 97.Schirmer RH, Adler H, Pickhardt M & Mandelkow E. 2011. “Lest we forget you-methylene blue…”. Neurobiol. Aging 32: 2325.e7–2325.e16. [DOI] [PubMed] [Google Scholar]
  • 98.Jancura D, Stanicova J, Palmer G & Fabian M. 2014. How hydrogen peroxide is metabolized by oxidized cytochrome c oxidase. Biochemistry 53: 3564–3575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Jünemann S, Heathcote P & Rich PR. 2000. The reactions of hydrogen peroxide with bovine cytochrome c oxidase. Biochim. Biophys. Acta Bioenerg 1456: 56–66. [DOI] [PubMed] [Google Scholar]
  • 100.Riha PD, Rojas JC & Gonzalez-Lima F. 2011. Beneficial network effects of methylene blue in an amnestic model. Neuroimage 54: 2623–2634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Zhang X, Rojas JC & Gonzalez-Lima F. 2006. Methylene blue prevents neurodegeneration caused by rotenone in the retina. Neurotox. Res 9: 47–57. [DOI] [PubMed] [Google Scholar]

RESOURCES