Background

The antifibrinolytic drug tranexamic acid is structurally similar to the amino acid glycine and may cause seizures and myoclonus by acting as a competitive antagonist of glycine receptors. Glycine is an obligatory co-agonist of the N-methyl-d-aspartate (NMDA) subtype of glutamate receptors. Thus, it is plausible that tranexamic acid inhibits NMDA receptors by acting as a competitive antagonist at the glycine binding site. The aim of this study was to determine whether tranexamic acid inhibits NMDA receptors, as well as α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid and kainate subtypes of ionotropic glutamate receptors.

Methods

Tranexamic acid modulation of NMDA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, and kainate receptors was studied using whole cell voltage-clamp recordings of current from cultured mouse hippocampal neurons.

Results

Tranexamic acid rapidly and reversibly inhibited NMDA receptors (half maximal inhibitory concentration = 241 ± 45 mM, mean ± SD; 95% CI, 200 to 281; n = 5) and shifted the glycine concentration–response curve for NMDA-evoked current to the right. Tranexamic acid also inhibited α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (half maximal inhibitory concentration = 231 ± 91 mM; 95% CI, 148 to 314; n = 5 to 6) and kainate receptors (half maximal inhibitory concentration = 90 ± 24 mM; 95% CI, 68 to 112; n = 5).

Conclusions

Tranexamic acid inhibits NMDA receptors likely by reducing the binding of the co-agonist glycine and also inhibits α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid and kainate receptors. Receptor blockade occurs at high millimolar concentrations of tranexamic acid, similar to the concentrations that occur after topical application to peripheral tissues. Glutamate receptors in tissues including bone, heart, and nerves play various physiologic roles, and tranexamic acid inhibition of these receptors may contribute to adverse drug effects.

What We Already Know about This Topic
  • The antifibrinolytic drug tranexamic acid may cause seizures by acting as a competitive antagonist of glycine receptors.

  • Glycine is an obligatory co-agonist of the N-methyl-d-aspartate receptors found in the brain and peripheral tissues.

What This Article Tells Us That Is New
  • Tranexamic acid inhibits N-methyl-d-aspartate receptors likely by reducing the binding of the co-agonist glycine and also inhibits other ionotropic glutamate receptors. Receptor blockade only occurs at high concentrations, similar to those that occur after topical application to peripheral tissues.

  • Inhibition of glutamate receptors in peripheral tissues may contribute to adverse effects observed at high concentrations.

TRANEXAMIC ACID (TXA) is an antifibrinolytic drug that is used widely to reduce blood loss resulting from a variety of hemorrhagic causes, including trauma, postpartum hemorrhage, and surgical procedures.1–8  TXA is a synthetic analog of the endogenous amino acid lysine, which binds to the lysine binding site of plasminogen.9  TXA blocks the conversion of plasminogen to plasmin and the degradation of fibrin blood clots, thereby producing hemostatic effects.9 

TXA is administered either systemically or topically. Systemic administration to patients by intravenous injection produces concentrations in the cerebrospinal fluid of 30 to 200 μM and plasma of 0.6 to 2 mM.7,10–14  In contrast, topical application of TXA directly to peripheral tissues during some surgical procedures would produce high localized concentrations (0.7 to 100 mg/ml, equivalent to 5 to 600 mM).2  Topical application of TXA is becoming increasingly popular, as this method may reduce bleeding and produce fewer side effects than systemic administration.2  Plasma concentrations after topical application generally are less than one tenth of the level after intravenous administration.2,15–17 

TXA inhibition of receptors other than plasminogen may cause adverse effects including seizures and myoclonus.14,18,19  TXA is structurally similar to the inhibitory neurotransmitter glycine and acts as a competitive antagonist at glycine receptors.14  Concentrations of TXA in the low millimolar range reduce inhibitory neurotransmission (or disinhibition), which causes network hyperexcitability and seizure-like events in animal models.14,19–21 

The effects of TXA on the major subtypes of excitatory ionotropic glutamate receptors that also modify brain network excitability have not been elucidated fully. These receptor subtypes include N-methyl-d-aspartate (NMDA) receptors, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, and kainate receptors.22  TXA modulation of NMDA receptors is of particular interest because these receptors contain a high-affinity glycine binding site.23  Binding of both glutamate and glycine is required for full activation of the NMDA receptors, and drugs that inhibit glycine binding reduce NMDA receptor function.24  TXA could compete with glycine at the glycine binding of the NMDA receptor and thereby reduce receptor function. Others have shown that TXA, at low concentrations (1 to 5 mM), does not inhibit excitatory synaptic currents in amygdala slices.25  However, the effects of high concentrations of TXA on glutamate receptors, such as those that occur during topical application, have not been elucidated. Glutamate receptors are expressed widely in peripheral tissues, including bone, heart, pancreas, and nerves, where they serve diverse physiologic roles.26–32  Thus, it is of interest to determine whether TXA blocks ionotropic glutamate receptors.

For these proof-of-concept studies, currents generated by NMDA, AMPA, and kainate receptors were recorded from hippocampal neurons. The rationale is that these cells express high levels of ionotropic glutamate receptors,33  and receptors in hippocampal neurons and peripheral tissues have similar structural, physiologic, and pharmacologic properties.26,28,31  The results show that TXA reduces NMDA receptor function by acting as a competitive antagonist at the glycine binding site. Surprisingly, TXA also inhibits AMPA and kainate receptors. These results predict that TXA at high concentrations, applied topically to peripheral tissues, inhibits ionotropic glutamate receptors.

Cell Culture

All experimental procedures were approved by the Animal Care Committee of the University of Toronto (Toronto, Ontario, Canada). Primary cultures of hippocampal neurons were prepared from Swiss White mice (Charles River, Canada) as previously described.34  In brief, fetal pups (embryonic day 18) were removed from maternal mice that had been euthanized by cervical dislocation. The hippocampi of each fetus were collected and placed on an ice-cooled culture dish. Neurons were then dissociated by mechanical trituration with a Pasteur pipette (tip diameter 150 to 200 μm) and plated on 35-mm culture dishes. The culture dishes were coated with collagen or poly-d-lysine (Sigma-Aldrich, Canada). The density of neurons per culture dish was approximately 1 × 106 cells. Two hours later, the medium was changed to a neurobasal medium supplemented with 2% B27 and 1% GlutaMAX (Life Technologies, USA). The medium was changed every 3 days. The low-density dissociated neurons were maintained in culture for 14 to 20 days before use. At this point in time, hippocampal neurons become appropriately polarized, develop extensive axonal and dendritic arbors, and form numerous, functional synaptic connections with one another, which resemble mature hippocampal neurons in vivo.35  Culture dishes were prepared from at least two different mice for each experiment, and a maximum of two cells was recorded from each dish.

Whole Cell Voltage-clamp Recordings

Whole cell currents were recorded under voltage-clamp (–60 mV) conditions with an Axopatch 1D amplifier (Molecular Devices, USA) controlled with pClamp 8.0 software (Molecular Devices) via a Digidata 1322A interface (Molecular Devices). Patch pipettes with open-tip resistances of 2 to 3 MΩ were pulled from thin-walled borosilicate glass capillary tubes. Patch electrodes were filled with an intracellular solution containing (in mM) 140 cesium chloride, 10 HEPES, 11 EGTA, 2 magnesium chloride, 1 calcium chloride, 4 magnesium adenosine triphosphate, and 2 triethanolamine (adjusted to pH 7.3 with cesium hydroxide and to 285 to 295 mOsm with water). To record NMDA-evoked current, magnesium-free extracellular solution was used and contained (in mM) 140 sodium chloride, 1.3 calcium chloride, 2 potassium chloride, 25 HEPES, and 28 glucose (adjusted to pH 7.4 with sodium hydroxide and to 320 to 330 mOsm with water). To record AMPA- and kainate-evoked current, magnesium chloride (1 mM) was added to the extracellular solution. Tetrodotoxin (300 nM) was added to the extracellular solution to block voltage-sensitive sodium channels. A computer-controlled, multibarrelled perfusion system (SF-77B; Warner Instruments, USA) was used to apply the extracellular solution to neurons. The time interval between applications of agonists, including NMDA, AMPA, and kainate, was 2 min. This interval was sufficient to allow receptors to recover from desensitization. No randomization methods were applied. Currents were recorded before and during the application of TXA, and the experimenters were not blinded to the drug application conditions.

Drugs

TXA, NMDA, AMPA, kainate, and glycine were obtained from Sigma-Aldrich. Tetrodotoxin was purchased from Alomone Labs (Israel). Stock solutions of these reagents were prepared with distilled water.

Data Analysis

Currents were analyzed with pClamp 10 software (Molecular Devices). The concentration–response plots were fitted to the modified Michaelis–Menten equation: I = Imax/[1 + (EC50/c)nH], where I is the current amplitude, EC50 is the concentration of agonist that produces currents with 50% of the maximal amplitude, c is the concentration of agonist, and nH is the estimated Hill coefficient. The concentration–response plots for TXA inhibition were fitted to the following equation: I = (IC50) nH/[cnH + (IC50)nH], where IC50 is the concentration of TXA that inhibited 50% of the current amplitude.

Statistical Analyses

No statistical power calculation was conducted before the study, and the sample size was based on our previous experience with this experimental design. There were no missing data from the results presented in this manuscript. Results are presented as mean ± SD together with 95% CI of the mean. Statistical analysis was performed with GraphPad Prism 5 software (GraphPad Software Inc., USA). Differences between groups were determined with the Student’s t test or a one-way ANOVA with a Dunnett multiple-comparison post hoc test. A two-tailed hypothesis test was used, and any P value less than 0.05 was considered significant.

High Concentrations of TXA Inhibit NMDA Receptors

We first identified the concentration of NMDA that evoked 50% of the maximal current (EC50) in hippocampal neurons as this information was used to design studies that examined TXA inhibition of NMDA receptors. Application of NMDA (3 to 3,000 μM) rapidly activated inward current, which increased in amplitude with increasing concentrations of NMDA in the presence of glycine (1 μM). The concentration–response plot for NMDA-evoked current revealed an EC50 value of 98 ± 44 μM (95% CI, 51 to 146; n = 4; fig. 1A).

Fig. 1.

TXA inhibits N-methyl-d-aspartate (NMDA)-evoked currents in hippocampal neurons. (A) Representative traces showing currents evoked by increasing concentrations of NMDA and the corresponding concentration–response plot (n = 4). (B) Representative traces and the corresponding concentration–response plot (n = 5) showing the inhibitory effects of tranexamic acid (TXA) on currents activated by NMDA (100 μM). Glycine (1 μM) was present continuously in the bath. All data are presented as mean ± SD.

Fig. 1.

TXA inhibits N-methyl-d-aspartate (NMDA)-evoked currents in hippocampal neurons. (A) Representative traces showing currents evoked by increasing concentrations of NMDA and the corresponding concentration–response plot (n = 4). (B) Representative traces and the corresponding concentration–response plot (n = 5) showing the inhibitory effects of tranexamic acid (TXA) on currents activated by NMDA (100 μM). Glycine (1 μM) was present continuously in the bath. All data are presented as mean ± SD.

Close modal

Next, to study the inhibitory effects of TXA on NMDA receptors, NMDA was applied at a concentration close to the EC50 value (100 μM) together with glycine (1 μM), in the absence or presence of TXA. Low concentrations of TXA (1 mM, 3 mM) had no effect on the amplitude of the peak current. However, higher TXA concentrations (10 mM or greater) rapidly and reversibly inhibited the NMDA-evoked current (fig. 1B). TXA at the threshold concentration (10 mM) inhibited the NMDA current by 4.2 ± 5.4% (95% CI, −0.5 to 8.9; n = 5), and the maximum inhibition at 3 M was 97.1 ± 1.8% (95% CI, 95.5 to 98.7; n = 5). The concentration–response plot showed that the concentration of TXA required to inhibit 50% of the maximal peak current (IC50) was 241 ± 45 mM (95% CI, 200 to 281; n = 5).

TXA Is a Competitive Antagonist at the Glycine Binding Site

To further probe the hypothesis that TXA decreases NMDA receptor function by competitively inhibiting glycine binding, we next examined the inhibitory effects of TXA (100 mM) on current evoked by NMDA (100 μM) with different concentrations of glycine in the extracellular solution. The extent of TXA-mediated inhibition decreased with increasing concentrations of glycine (fig. 2A), and a near-saturating concentration of glycine almost completely abolished TXA inhibition of NMDA-evoked current. Specifically, TXA inhibited NMDA currents by 59.1 ± 23.2% (95% CI, 43.0 to 75.2; n = 8) in the presence of 0.3 μM glycine but by only 2.2 ± 3.7% (95% CI, −0.4 to 4.8; n = 8) when the glycine concentration was increased to 30 μM. We next constructed glycine (0.03 to 100 μM) concentration–response plots for NMDA current, recorded in the absence and presence of TXA (fig. 2B). TXA shifted the glycine concentration–response curve to the right without changing the maximum response. The glycine EC50 increased from 0.48 ± 0.09 μM (95% CI, 0.43 to 0.53; n = 10) to 0.95 ± 0.19 μM (95% CI, 0.81 to 1.1; n = 8) in the presence of TXA (P = 0.02), whereas the Hill coefficient was unchanged (control 1.5 ± 0.6; 95% CI, 1.1 to 1.9; n = 10 vs. TXA 1.6 ± 0.3; 95% CI, 1.5 to 1.7; n = 8, P = 0.9). These results are consistent with the hypothesis that TXA inhibits NMDA receptors by acting as a competitive antagonist at the glycine binding site.

Fig. 2.

Tranexamic acid (TXA) inhibition of N-methyl-d-aspartate (NMDA)-evoked currents decreases with increased glycine concentration. (A) Representative traces demonstrating decreased TXA (100 mM) inhibition of currents evoked by NMDA (100 μM) with an increased glycine concentration. (B) Glycine concentration–response plots for currents evoked by NMDA (100 μM) in the absence and presence of TXA. Glycine half maximal effective concentration was increased from 0.48 ± 0.09 μM (95% CI, 0.43 to 0.53) without TXA to 0.95 ± 0.19 μM (95% CI, 0.81 to 1.1) with TXA (P = 0.02, unpaired one-tailed Student’s t test). All responses were normalized to the peak current evoked by NMDA (100 μM) and glycine (1 μM) without TXA. The sample sizes in the absence and presence of TXA were n = 10 and n = 8, respectively. All data are presented as mean ± SD.

Fig. 2.

Tranexamic acid (TXA) inhibition of N-methyl-d-aspartate (NMDA)-evoked currents decreases with increased glycine concentration. (A) Representative traces demonstrating decreased TXA (100 mM) inhibition of currents evoked by NMDA (100 μM) with an increased glycine concentration. (B) Glycine concentration–response plots for currents evoked by NMDA (100 μM) in the absence and presence of TXA. Glycine half maximal effective concentration was increased from 0.48 ± 0.09 μM (95% CI, 0.43 to 0.53) without TXA to 0.95 ± 0.19 μM (95% CI, 0.81 to 1.1) with TXA (P = 0.02, unpaired one-tailed Student’s t test). All responses were normalized to the peak current evoked by NMDA (100 μM) and glycine (1 μM) without TXA. The sample sizes in the absence and presence of TXA were n = 10 and n = 8, respectively. All data are presented as mean ± SD.

Close modal

The binding sites of NMDA and glycine interact allosterically such that glycine affinity increases with the binding of NMDA and, conversely, NMDA affinity increases with the binding of glycine.36,37  These allosteric interactions predict that TXA blockade should decrease with increasing concentrations of NMDA (because of increased glycine binding). To test this prediction, the inhibitory effects of TXA (100 mM) on NMDA currents were studied under conditions in which the concentration of NMDA was varied whereas the concentration of glycine (1 μM) was fixed (fig. 3). For NMDA 1, 10, and 100 μM, TXA inhibited the currents by 66.1 ± 10.3% (95% CI, 57.9 to 74.3), 48.3 ± 12.7% (95% CI, 38.1 to 58.5), and 26.4 ± 2.5% (95% CI, 24.6 to 28.6), respectively (n = 6). As predicted, TXA inhibition decreased with increasing concentrations of NMDA.

Fig. 3.

Tranexamic acid (TXA) inhibition of N-methyl-d-aspartate (NMDA)-evoked currents is reduced with increasing NMDA concentrations. (A) Representative traces showing decreased effects of TXA (100 mM) on currents evoked by an increased concentration of NMDA. (B) Summarized data for the responses shown in (A) (n = 6). Glycine (1 μM) was present continuously in the bath. *P < 0.05, **P < 0.01, ***P < 0.001, one-way ANOVA with Dunnett multiple-comparison post hoc test. All data are presented as mean ± SD.

Fig. 3.

Tranexamic acid (TXA) inhibition of N-methyl-d-aspartate (NMDA)-evoked currents is reduced with increasing NMDA concentrations. (A) Representative traces showing decreased effects of TXA (100 mM) on currents evoked by an increased concentration of NMDA. (B) Summarized data for the responses shown in (A) (n = 6). Glycine (1 μM) was present continuously in the bath. *P < 0.05, **P < 0.01, ***P < 0.001, one-way ANOVA with Dunnett multiple-comparison post hoc test. All data are presented as mean ± SD.

Close modal

Inhibition by TXA Is Not Use Dependent or Voltage Dependent

TXA inhibition should exhibit no use or voltage dependence if it acts as a competitive antagonist at the glycine binding site, rather than as a noncompetitive blocker of the open channel pore.38  The extent of TXA (100 mM) inhibition was similar after repeated application of NMDA and TXA (fig. 4A), suggesting the block is not use dependent. Next, the concentration of TXA in the extracellular solution was increased to 300 mM (close to IC50 value). Neurons were perfused continuously with this solution. Three sequential applications of NMDA (100 μM) showed that inhibition of three current pulses by TXA was similar (78.9 ± 10.3% with 95% CI, 69.9 to 87.9; 78.5 ± 9.6% with 95% CI, 70.1 to 86.9; and 79.8 ± 9.8% with 95% CI, 71.2 to 88.4; respectively, n = 5; P = 0.5, one-way ANOVA). These results further indicate that TXA inhibition of NMDA receptors was not use dependent. To determine whether TXA-mediated inhibition was voltage dependent, NMDA currents were recorded at different holding potentials in the absence or presence of TXA (100 mM) and current–voltage plots were constructed. TXA decreased the slope of the current–voltage plot but did not change the reversal potential (NMDA: –5.2 mV, +TXA: –5.5 mV, P = 0.9; fig. 4B). Moreover, the extent of TXA-induced inhibition of NMDA currents was equivalent at holding potentials between –60 and +60 mV (P = 0.8, fig. 4C). Thus, TXA inhibition of NMDA currents was not voltage dependent.

Fig. 4.

Tranexamic acid (TXA) inhibition of N-methyl-d-aspartate (NMDA) currents is not use and voltage dependent. (A) Repeated application of TXA (100 mM) did not cause a statistically significant increase in blockade of currents evoked by NMDA (100 μM). Currents recorded during the sequential application of TXA are identified as 1, 2, and 3. The bar graph shows mean peak amplitude of the current recorded with and without coapplication of TXA (n = 5). ***P < 0.001 versus NMDA, one-way ANOVA with Dunnett multiple-comparison post hoc test. (B and C) Representative traces and summarized data showing the inhibitory effects of TXA (100 mM) on currents evoked by NMDA (100 μM) at different holding potentials (VH). For current–voltage (I–V) plots in (B), the currents were normalized to the peak current induced by NMDA without TXA at a holding potential of –60 mV. The sample sizes for the I–V plot in the absence and presence of TXA were n = 9 and n = 7, respectively. In (C), note that TXA-induced inhibition of NMDA currents was similar for all holding potentials. P = 0.8, one-way ANOVA with Dunnett multiple-comparison post hoc test. For all recordings, glycine (1 μM) was continuously in the bath. All data are presented as mean ± SD.

Fig. 4.

Tranexamic acid (TXA) inhibition of N-methyl-d-aspartate (NMDA) currents is not use and voltage dependent. (A) Repeated application of TXA (100 mM) did not cause a statistically significant increase in blockade of currents evoked by NMDA (100 μM). Currents recorded during the sequential application of TXA are identified as 1, 2, and 3. The bar graph shows mean peak amplitude of the current recorded with and without coapplication of TXA (n = 5). ***P < 0.001 versus NMDA, one-way ANOVA with Dunnett multiple-comparison post hoc test. (B and C) Representative traces and summarized data showing the inhibitory effects of TXA (100 mM) on currents evoked by NMDA (100 μM) at different holding potentials (VH). For current–voltage (I–V) plots in (B), the currents were normalized to the peak current induced by NMDA without TXA at a holding potential of –60 mV. The sample sizes for the I–V plot in the absence and presence of TXA were n = 9 and n = 7, respectively. In (C), note that TXA-induced inhibition of NMDA currents was similar for all holding potentials. P = 0.8, one-way ANOVA with Dunnett multiple-comparison post hoc test. For all recordings, glycine (1 μM) was continuously in the bath. All data are presented as mean ± SD.

Close modal

High Concentrations of TXA Inhibit AMPA and Kainate Receptors

Unlike NMDA receptors, AMPA receptors and kainate receptors do not require the binding of the co-agonist glycine to promote channel opening. Thus, we hypothesized that TXA would not inhibit AMPA and kainate receptor function. To test this hypothesis, AMPA and kainate were first applied to the neurons to determine EC50 values (fig. 5, A and B). Next, the inhibitory effects of TXA on currents evoked by AMPA (10 μM) and kainate (20 μM), applied at concentrations close to the EC50 values, were studied. Contrary to what was predicted, TXA caused concentration-dependent inhibition of both AMPA- and kainate-evoked currents, with IC50 values of 231 ± 91 mM (95% CI, 148 to 314; n = 5 to 6; fig. 5C) and 90 ± 24 mM (95% CI, 68 to 112; n = 5; fig. 5D), respectively. TXA blockade was rapid and was completely reversed after drug washout.

Fig. 5.

Tranexamic acid (TXA) inhibits α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)- and kainate-evoked currents. (A and B) Representative traces and the concentration–response plots for currents evoked by (A) AMPA and (B) kainate. The half maximal effective concentration was 8.6 ± 4.0 μM (95% CI, 5.2 to 12.0) for AMPA responses and 21.8 ± 3.6 μM (95% CI, 18.3 to 25.3) for kainate responses. The sample sizes for 0.3 to 10, 30 to 100, and 300 μM AMPA were n = 6, n = 5, and n = 4, respectively. The sample size for kainate was n = 4. (C and D) Representative traces and the corresponding concentration–response plots demonstrating the inhibitory effects of TXA on currents induced by AMPA (10 μM, C) and kainate (20 μM, D). The sample sizes for 3 to 100 mM and 300 to 3,000 mM TXA on AMPA currents were n = 6 and n = 5, respectively. The sample sizes for TXA on kainate currents were n = 5. All data are presented as mean ± SD.

Fig. 5.

Tranexamic acid (TXA) inhibits α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)- and kainate-evoked currents. (A and B) Representative traces and the concentration–response plots for currents evoked by (A) AMPA and (B) kainate. The half maximal effective concentration was 8.6 ± 4.0 μM (95% CI, 5.2 to 12.0) for AMPA responses and 21.8 ± 3.6 μM (95% CI, 18.3 to 25.3) for kainate responses. The sample sizes for 0.3 to 10, 30 to 100, and 300 μM AMPA were n = 6, n = 5, and n = 4, respectively. The sample size for kainate was n = 4. (C and D) Representative traces and the corresponding concentration–response plots demonstrating the inhibitory effects of TXA on currents induced by AMPA (10 μM, C) and kainate (20 μM, D). The sample sizes for 3 to 100 mM and 300 to 3,000 mM TXA on AMPA currents were n = 6 and n = 5, respectively. The sample sizes for TXA on kainate currents were n = 5. All data are presented as mean ± SD.

Close modal

The results show that TXA inhibited ionotropic glutamate receptors, but only at high millimolar concentrations. The efficacy of TXA inhibition of NMDA receptors depended on the concentrations of glycine and NMDA, and the blockade was neither use dependent nor voltage dependent. TXA also inhibited AMPA and kainate receptors at concentrations that are similar to those that inhibited NMDA receptors.

These results build on two previous studies that examined the effects of TXA on glutamate receptor function.18,25  In one of these studies, electrophysiologic recordings showed that TXA did not inhibit excitatory currents in amygdala slices, but only low millimolar concentrations were studied.25  More specifically, TXA concentrations less than 10 mM did not modify synaptic currents generated by glutamate receptors.25  These findings are consistent with our results. In the second study, TXA, at concentrations up to 10 mM, did not bind to NMDA receptors in rat cortical tissue.18  However, the experimental conditions used in the study did not favor TXA binding as the extracellular solution contained high concentrations of NMDA and glycine. TXA blockade of NMDA receptors is reduced under these conditions, possibly because of a competitive interaction between glycine and TXA at the NMDA receptor.

The results from our electrophysiologic studies do not discern whether TXA binds directly to NMDA receptors or indirectly reduces receptor function. However, several lines of evidence suggest that TXA inhibits NMDA receptors directly by preventing glycine from acting as a full co-agonist at these receptors. First, high extracellular concentrations of glycine reduce TXA blockade. Second, TXA shifts the glycine concentration–response plot to the right without reducing the maximal NMDA-evoked current response. Third, TXA inhibition of NMDA receptors is not use dependent or voltage dependent, which suggests that TXA lacks the features of steric blockers, such as magnesium and ketamine, which occlude the open channel pore.39–41 

Interestingly, increasing the concentration of NMDA reduced TXA inhibition of NMDA receptors. This result is consistent with evidence indicating that glutamate and glycine binding to the NMDA receptors is allosterically coupled.36,37  Glutamate agonists increase [3H]glycine binding, whereas glutamate antagonists decrease [3H]glycine binding.37  Reciprocally, competitive antagonists at the glycine binding site allosterically reduce glutamate binding.36  Increasing the concentration of NMDA likely increases glycine affinity, thereby reducing TXA inhibition.

TXA also inhibits AMPA receptors and kainate receptors. These results were unexpected, given that the AMPA and kainate receptors lack a glycine binding site.22  However, TXA inhibits other transmitter-gated ion channels that lack a glycine binding site, including γ-aminobutyric acid type A receptors.14,18,25  It remains unknown how TXA inhibits AMPA and kainate receptors, and further studies are required to determine whether inhibition results from steric, allosteric, or other indirect mechanisms.

Although we examined the TXA sensitivity of glutamate receptors expressed in central neurons, the receptors expressed in peripheral tissues are more likely to be exposed to high millimolar concentrations of TXA. Nevertheless, ionotropic glutamate receptors in peripheral tissues and central neurons exhibit similar structural, kinetic, and pharmacologic properties, suggesting that the results are clinically relevant.26,28,31  For example, in both central neurons and peripheral tissues, GluN1 and GluN2 subunits form functional NMDA receptors.28,31  Also, current generated by glutamate receptors in bone and heart tissue exhibits kinetic and pharmacologic sensitivities that are similar to those of glutamate receptors in neurons.26,28  For example, NMDA receptors in bone cells and neurons are inhibited by both magnesium ions and the high-affinity antagonist MK-801,26  suggesting that receptors expressed in neurons and nonneuronal tissues likely exhibit similar sensitivity to TXA.

High concentrations of TXA that inhibit glutamate receptors could occur after topical application of the drug. Glutamate receptors expressed in peripheral tissues would be exposed to such high concentrations. For example, topical application of TXA is used commonly during orthopedic surgery.17,42–45  NMDA receptors composed of GluN1 and GluN2A-D are expressed in osteoblasts and osteoclasts in bone.28,31  As such, TXA inhibition of NMDA receptors could alter bone healing. Furthermore, receptor subunits of NMDA (GluN1 and GluN2), AMPA (GluA2 and GluA3), and kainate (GluK1 and GluK2) are expressed in the heart, pancreas, and gastrointestinal tissue.28–30,46  Cardiac glutamate receptors, which are localized preferentially within the conducting system, the nerve terminals, and the intramural ganglia cells,27  are involved in conducting impulses and rhythm control.27,47  It is plausible that topical application of TXA to the heart’s conducting system could cause dysrhythmias. Inhibition of NMDA receptors in human and mouse pancreatic islets increases secretion of insulin, so receptor inhibition by TXA could alter glucose levels.48  Finally, inhibition of NMDA receptors in peripheral nerves alters nociception in both rodents and humans.49 

Awareness of such potential “off-target” effects secondary to TXA effects on glutamate receptors will inform studies that seek to identify adverse effects of topically applied TXA in patients. Clinical studies first focused on the adverse consequences of antifibrinolytic effects of TXA, including deep vein thrombosis, stroke, and myocardial infarction.50–53  Side effects due to off-target receptor effects only have been recognized recently. For example, seizure and myoclonus likely due to TXA inhibition of glycine receptors and γ-aminobutyric acid type A receptors and mechanistically based treatments for those seizures only have been identified recently.19  By analogy, the physiologic roles of glutamate receptors expressed in peripheral tissues are only beginning to be understood. It will be worthwhile, in future studies, to determine whether TXA inhibition of such physiologic functions contributes to adverse effects of TXA.

In summary, high concentrations of TXA inhibit ionotropic glutamate receptors. These preclinical results will support future studies aiming to clarify the safety and consequences of applying TXA topically to nonneuronal tissues and peripheral nerves in patients.

The authors thank Ella Czerwinska, M.Sc., and Junhui Wang, M.D., Ph.D., Department of Physiology, University of Toronto, Toronto, Ontario, Canada, for their assistance with the cell cultures.

This work was supported by operating grants from the Canadian Institutes of Health Research (CIHR), Ottawa, Ontario, Canada, to Dr. Orser (grant Nos.: 416838, 480143). Dr. Lecker was supported by the Frederick Banting and Charles Best Doctoral Scholarship from CIHR. K. Kaneshwaran was supported by the Kirk Weber Award in Anesthesia from the Department of Anesthesia, Sunnybrook Health Services Centre, Toronto, Ontario, Canada; and the Ontario Graduate Scholarship, Ontario, Canada.

The authors declare no competing interests.

1.
Henry
DA
,
Carless
PA
,
Moxey
AJ
,
O’Connell
D
,
Stokes
BJ
,
Fergusson
DA
,
Ker
K
:
Anti-fibrinolytic use for minimising perioperative allogeneic blood transfusion.
Cochrane Database Syst Rev
2011
, pp
CD001886
2.
Ker
K
,
Beecher
D
,
Roberts
I
:
Topical application of tranexamic acid for the reduction of bleeding.
Cochrane Database Syst Rev
2013
, pp
CD010562
3.
Leminen
H
,
Hurskainen
R
:
Tranexamic acid for the treatment of heavy menstrual bleeding: Efficacy and safety.
Int J Womens Health
2012
;
4
:
413
21
4.
Morrison
JJ
,
Dubose
JJ
,
Rasmussen
TE
,
Midwinter
MJ
:
Military Application of Tranexamic Acid in Trauma Emergency Resuscitation (MATTERs) study.
Arch Surg
2012
;
147
:
113
9
5.
Park
KJ
,
Couch
CG
,
Edwards
PK
,
Siegel
ER
,
Mears
SC
,
Barnes
CL
:
Tranexamic acid reduces blood transfusions in revision total hip arthroplasty.
J Arthroplasty
2016
;
31
:
2850
2855.e1
6.
Roberts
I
,
Shakur
H
,
Afolabi
A
,
Brohi
K
,
Coats
T
,
Dewan
Y
,
Gando
S
,
Guyatt
G
,
Hunt
BJ
,
Morales
C
,
Perel
P
,
Prieto-Merino
D
,
Woolley
T
:
The importance of early treatment with tranexamic acid in bleeding trauma patients: An exploratory analysis of the CRASH-2 randomised controlled trial.
Lancet
2011
;
377
:
1096
101
7.
Sharma
V
,
Fan
J
,
Jerath
A
,
Pang
KS
,
Bojko
B
,
Pawliszyn
J
,
Karski
JM
,
Yau
T
,
McCluskey
S
,
Wąsowicz
M
:
Pharmacokinetics of tranexamic acid in patients undergoing cardiac surgery with use of cardiopulmonary bypass.
Anaesthesia
2012
;
67
:
1242
50
8.
Simonazzi
G
,
Bisulli
M
,
Saccone
G
,
Moro
E
,
Marshall
A
,
Berghella
V
:
Tranexamic acid for preventing postpartum blood loss after cesarean delivery: A systematic review and meta-analysis of randomized controlled trials.
Acta Obstet Gynecol Scand
2016
;
95
:
28
37
9.
Hoylaerts
M
,
Lijnen
HR
,
Collen
D
:
Studies on the mechanism of the antifibrinolytic action of tranexamic acid.
Biochim Biophys Acta
1981
;
673
:
75
85
10.
Abou-Diwan
C
,
Sniecinski
RM
,
Szlam
F
,
Ritchie
JC
,
Rhea
JM
,
Tanaka
KA
,
Molinaro
RJ
:
Plasma and cerebral spinal fluid tranexamic acid quantitation in cardiopulmonary bypass patients.
J Chromatogr B Analyt Technol Biomed Life Sci
2011
;
879
:
553
6
11.
Dowd
NP
,
Karski
JM
,
Cheng
DC
,
Carroll
JA
,
Lin
Y
,
James
RL
,
Butterworth
J
:
Pharmacokinetics of tranexamic acid during cardiopulmonary bypass.
Anesthesiology
2002
;
97
:
390
9
12.
Fiechtner
BK
,
Nuttall
GA
,
Johnson
ME
,
Dong
Y
,
Sujirattanawimol
N
,
Oliver
WC
Jr
,
Sarpal
RS
,
Oyen
LJ
,
Ereth
MH
:
Plasma tranexamic acid concentrations during cardiopulmonary bypass.
Anesth Analg
2001
;
92
:
1131
6
13.
Grassin-Delyle
S
,
Tremey
B
,
Abe
E
,
Fischler
M
,
Alvarez
JC
,
Devillier
P
,
Urien
S
:
Population pharmacokinetics of tranexamic acid in adults undergoing cardiac surgery with cardiopulmonary bypass.
Br J Anaesth
2013
;
111
:
916
24
14.
Lecker
I
,
Wang
DS
,
Romaschin
AD
,
Peterson
M
,
Mazer
CD
,
Orser
BA
:
Tranexamic acid concentrations associated with human seizures inhibit glycine receptors.
J Clin Invest
2012
;
122
:
4654
66
15.
Almer
S
,
Andersson
T
,
Ström
M
:
Pharmacokinetics of tranexamic acid in patients with ulcerative colitis and in healthy volunteers after the single instillation of 2 g rectally.
J Clin Pharmacol
1992
;
32
:
49
54
16.
Sindet-Pedersen
S
:
Distribution of tranexamic acid to plasma and saliva after oral administration and mouth rinsing: A pharmacokinetic study.
J Clin Pharmacol
1987
;
27
:
1005
8
17.
Wong
J
,
Abrishami
A
,
El Beheiry
H
,
Mahomed
NN
,
Roderick Davey
J
,
Gandhi
R
,
Syed
KA
,
Muhammad Ovais Hasan
S
,
De Silva
Y
,
Chung
F
:
Topical application of tranexamic acid reduces postoperative blood loss in total knee arthroplasty: A randomized, controlled trial.
J Bone Joint Surg Am
2010
;
92
:
2503
13
18.
Furtmüller
R
,
Schlag
MG
,
Berger
M
,
Hopf
R
,
Huck
S
,
Sieghart
W
,
Redl
H
:
Tranexamic acid, a widely used antifibrinolytic agent, causes convulsions by a γ-aminobutyric acidA receptor antagonistic effect.
J Pharmacol Exp Ther
2002
;
301
:
168
73
19.
Lecker
I
,
Wang
DS
,
Whissell
PD
,
Avramescu
S
,
Mazer
CD
,
Orser
BA
:
Tranexamic acid-associated seizures: Causes and treatment.
Ann Neurol
2016
;
79
:
18
26
20.
Pellegrini
A
,
Giaretta
D
,
Chemello
R
,
Zanotto
L
,
Testa
G
:
Feline generalized epilepsy induced by tranexamic acid (AMCA).
Epilepsia
1982
;
23
:
35
45
21.
Schlag
MG
,
Hopf
R
,
Redl
H
:
Convulsive seizures following subdural application of fibrin sealant containing tranexamic acid in a rat model.
Neurosurgery
2000
;
47
:
1463
7
22.
Traynelis
SF
,
Wollmuth
LP
,
McBain
CJ
,
Menniti
FS
,
Vance
KM
,
Ogden
KK
,
Hansen
KB
,
Yuan
H
,
Myers
SJ
,
Dingledine
R
:
Glutamate receptor ion channels: Structure, regulation, and function.
Pharmacol Rev
2010
;
62
:
405
96
23.
Johnson
JW
,
Ascher
P
:
Glycine potentiates the NMDA response in cultured mouse brain neurons.
Nature
1987
;
325
:
529
31
24.
Kauer
JA
,
Malenka
RC
,
Nicoll
RA
:
NMDA application potentiates synaptic transmission in the hippocampus.
Nature
1988
;
334
:
250
2
25.
Kratzer
S
,
Irl
H
,
Mattusch
C
,
Bürge
M
,
Kurz
J
,
Kochs
E
,
Eder
M
,
Rammes
G
,
Haseneder
R
:
Tranexamic acid impairs γ-aminobutyric acid receptor type A-mediated synaptic transmission in the murine amygdala: A potential mechanism for drug-induced seizures?
Anesthesiology
2014
;
120
:
639
49
26.
Espinosa
L
,
Itzstein
C
,
Cheynel
H
,
Delmas
PD
,
Chenu
C
:
Active NMDA glutamate receptors are expressed by mammalian osteoclasts.
J Physiol
1999
;
518
:
47
53
27.
Gill
S
,
Veinot
J
,
Kavanagh
M
,
Pulido
O
:
Human heart glutamate receptors—implications for toxicology, food safety, and drug discovery.
Toxicol Pathol
2007
;
35
:
411
7
28.
Gill
SS
,
Pulido
OM
:
Glutamate receptors in peripheral tissues: Current knowledge, future research, and implications for toxicology.
Toxicol Pathol
2001
;
29
:
208
23
29.
Gill
SS
,
Pulido
OM
,
Mueller
RW
,
McGuire
PF
:
Molecular and immunochemical characterization of the ionotropic glutamate receptors in the rat heart.
Brain Res Bull
1998
;
46
:
429
34
30.
Inagaki
N
,
Kuromi
H
,
Gonoi
T
,
Okamoto
Y
,
Ishida
H
,
Seino
Y
,
Kaneko
T
,
Iwanaga
T
,
Seino
S
:
Expression and role of ionotropic glutamate receptors in pancreatic islet cells.
FASEB J
1995
;
9
:
686
91
31.
Itzstein
C
,
Espinosa
L
,
Delmas
PD
,
Chenu
C
:
Specific antagonists of NMDA receptors prevent osteoclast sealing zone formation required for bone resorption.
Biochem Biophys Res Commun
2000
;
268
:
201
9
32.
Skerry
TM
,
Genever
PG
:
Glutamate signalling in non-neuronal tissues.
Trends Pharmacol Sci
2001
;
22
:
174
81
33.
Eshhar
N
,
Petralia
RS
,
Winters
CA
,
Niedzielski
AS
,
Wenthold
RJ
:
The segregation and expression of glutamate receptor subunits in cultured hippocampal neurons.
Neuroscience
1993
;
57
:
943
64
34.
MacDonald
JF
,
Mody
I
,
Salter
MW
,
Pennefather
P
,
Schneiderman
JH
:
The regulation of NMDA receptors in the central nervous system.
Prog Neuropsychopharmacol Biol Psychiatry
1989
;
13
:
481
8
35.
Kaech
S
,
Banker
G
:
Culturing hippocampal neurons.
Nat Protoc
2006
;
1
:
2406
15
36.
Grimwood
S
,
Kulagowski
JJ
,
Mawer
IM
,
Rowley
M
,
Leeson
PD
,
Foster
AC
:
Allosteric modulation of the glutamate site on the NMDA receptor by four novel glycine site antagonists.
Eur J Pharmacol
1995
;
290
:
221
6
37.
Lester
RA
,
Tong
G
,
Jahr
CE
:
Interactions between the glycine and glutamate binding sites of the NMDA receptor.
J Neurosci
1993
;
13
:
1088
96
38.
MacDonald
JF
,
Miljkovic
Z
,
Pennefather
P
:
Use-dependent block of excitatory amino acid currents in cultured neurons by ketamine.
J Neurophysiol
1987
;
58
:
251
66
39.
MacDonald
JF
,
Wojtowicz
JM
:
Two conductance mechanisms activated by applications of L-glutamic, L-aspartic, DL-homocysteic, N-methyl-D-aspartic, and DL-kainic acids to cultured mammalian central neurones.
Can J Physiol Pharmacol
1980
;
58
:
1393
7
40.
Mayer
ML
,
Westbrook
GL
,
Guthrie
PB
:
Voltage-dependent block by Mg2+ of NMDA responses in spinal cord neurones.
Nature
1984
;
309
:
261
3
41.
Nowak
L
,
Bregestovski
P
,
Ascher
P
,
Herbet
A
,
Prochiantz
A
:
Magnesium gates glutamate-activated channels in mouse central neurones.
Nature
1984
;
307
:
462
5
42.
Sun
X
,
Dong
Q
,
Zhang
YG
:
Intravenous versus topical tranexamic acid in primary total hip replacement: A systemic review and meta-analysis.
Int J Surg
2016
;
32
:
10
8
43.
Tzatzairis
TK
,
Drosos
GI
,
Kotsios
SE
,
Ververidis
AN
,
Vogiatzaki
TD
,
Kazakos
KI
:
Intravenous vs topical tranexamic acid in total knee arthroplasty without tourniquet application: A randomized controlled study.
J Arthroplasty
2016
;
31
:
2465
70
44.
Uğurlu
M
,
Aksekili
MA
,
Çağlar
C
,
Yüksel
K
,
Şahin
E
,
Akyol
M
:
Effect of topical and intravenously applied tranexamic acid compared to control group on bleeding in primary unilateral total knee arthroplasty.
J Knee Surg
2017
;
30
:
152
7
45.
Wang
H
,
Shen
B
,
Zeng
Y
:
Blood loss and transfusion after topical tranexamic acid administration in primary total knee arthroplasty.
Orthopedics
2015
;
38
:
e1007
16
46.
Tsai
LH
,
Tsai
W
,
Wu
JY
:
Effect of L-glutamic acid on acid secretion and immunohistochemical localization of glutamatergic neurons in the rat stomach.
J Neurosci Res
1994
;
38
:
188
95
47.
Huang
CF
,
Su
MJ
:
Positive inotropic action of NMDA receptor antagonist (+)-MK801 in rat heart.
J Biomed Sci
1999
;
6
:
387
98
48.
Marquard
J
,
Otter
S
,
Welters
A
,
Stirban
A
,
Fischer
A
,
Eglinger
J
,
Herebian
D
,
Kletke
O
,
Klemen
MS
,
Stožer
A
,
Wnendt
S
,
Piemonti
L
,
Köhler
M
,
Ferrer
J
,
Thorens
B
,
Schliess
F
,
Rupnik
MS
,
Heise
T
,
Berggren
PO
,
Klöcker
N
,
Meissner
T
,
Mayatepek
E
,
Eberhard
D
,
Kragl
M
,
Lammert
E
:
Characterization of pancreatic NMDA receptors as possible drug targets for diabetes treatment.
Nat Med
2015
;
21
:
363
72
49.
Cairns
BE
,
Svensson
P
,
Wang
K
,
Hupfeld
S
,
Graven-Nielsen
T
,
Sessle
BJ
,
Berde
CB
,
Arendt-Nielsen
L
:
Activation of peripheral NMDA receptors contributes to human pain and rat afferent discharges evoked by injection of glutamate into the masseter muscle.
J Neurophysiol
2003
;
90
:
2098
105
50.
Garg
J
,
Pinnamaneni
S
,
Aronow
WS
,
Ahmad
H
:
ST elevation myocardial infarction after tranexamic acid: First reported case in the United States.
Am J Ther
2014
;
21
:
e221
4
51.
Nardi
K
,
Pelone
G
,
Bartolo
M
,
Di Ruzza
MR
,
Storto
M
,
Notte
A
,
Grillea
G
,
Colonnese
C
,
Lembo
G
,
Vecchione
C
:
Ischaemic stroke following tranexamic acid in young patients carrying heterozygosity of MTHFR C677T.
Ann Clin Biochem
2011
;
48
(
Pt 6
):
575
8
52.
Sundström
A
,
Seaman
H
,
Kieler
H
,
Alfredsson
L
:
The risk of venous thromboembolism associated with the use of tranexamic acid and other drugs used to treat menorrhagia: A case-control study using the General Practice Research Database.
BJOG
2009
;
116
:
91
7
53.
Taparia
M
,
Cordingley
FT
,
Leahy
MF
:
Pulmonary embolism associated with tranexamic acid in severe acquired haemophilia.
Eur J Haematol
2002
;
68
:
307
9