Elsevier

Neuroscience

Volume 85, Issue 3, 24 April 1998, Pages 931-938
Neuroscience

Riluzole interacts with voltage-activated sodium and potassium currents in cultured rat cortical neurons

https://doi.org/10.1016/S0306-4522(97)00604-0Get rights and content

Abstract

The actions of the neuroprotective and anticonvulsant agent riluzole on voltage-activated currents were studied in primary cultures of rat cortical neurons by using whole-cell patch-clamp recording techniques. Isolated Na+, Ca2+ and K+ currents were generated in these cells by depolarizing commands from a holding potential of −80 mV. Riluzole (10–300 μM) reversibly reduced in a dose-dependent manner the inward Na+ currents with an IC50 of 51 μM in all the tested neurons (n=29). This drug also shifted the steady-state inactivation curve of the sodium current towards more negative values (about 20 mV, n=15) while it did not change significantly the decay phase of the Na+ current. Furthermore, riluzole (100 and 300 μM; n=5 and n=3, respectively) did not modulate the inward Ca2+ currents evoked by depolarizing steps on cortical cells. An additional concentration-dependent effect of riluzole was observed on the outward potassium currents. In fact, while the amplitude of the peak of the outward current (IA) was not changed significantly, the amplitude of the late component of the outward K+ current (Iss) was markedly decreased during the perfusion of riluzole (IC50=88 μM; n=16).

It is concluded that riluzole modulates the Na+- and the late K+-dependent currents in cortical neurons. Both phenomena may explain, at least in part, the anticonvulsant and neuroprotective properties of this compound.

Section snippets

Cell culture

Experiments were performed on neocortical neurons removed from 14 day Wistar rat embryos (Morini, Reggio Emilia, Italy) and grown in dissociated cell culture as described previously.6, 7Briefly, the animals were killed by decapitation under ether anaesthesia. Experiments were performed at 21–23°C, on neocortical neurons cultured for 12–16 days in Eagle's minimum essential medium (Gibco) with 5% rat serum, 6 mM of glucose, 2 mM of glutamine, and 100 μg/ml of gentamicine (Gibco). For more details,

Voltage-activated Na+ currents

In a first series of experiments, Ca2+ and K+ currents were pharmacologically blocked by extracellular application of Cd2+ and 4-AP as well as by the addition of Cs+ and TEA in the recording electrode. Under this experimental condition, depolarizing voltage commands from a holding potential of −80 mV elicited a voltage-dependent inward current (Fig. 1A) sensitive to TTX (complete block with 0.5 μM; data not illustrated). This inward Na+ current was maximally activated by a test pulse at −20 mV and

Discussion

Our findings have shown that riluzole induces multiple effects on voltage-activated currents in cultured rat cortical neurons. In fact. this drug causes: (i) a reversible and dose-dependent decrease of the Na+ current; (ii) a negative shift of the steady-state inactivation curve for the Na+ currents; (iii) a reversible and concentration-dependent reduction of the late K+ current. It is also worth mentioning that riluzole does not affect the fast transient outward currents, the voltage-dependent

Conclusions

The reduction of the Na+ current in cortical neurons and the shift of the steady-state inactivation Na+ curve might be related to the clinical effectiveness of riluzole as neuroprotectant. In fact, a depression of the voltage-activated Na+ current certainly reduces an excessive increase in cellular excitability and an enhanced release of excitatory amino acids.[23]These mechanisms might contribute to spare energy consumption and to preserve the ionic milieu during conditions of altered neuronal

Acknowledgements

The authors thank R. Sorge for the help in the statistical analysis and G. Gattoni and M. Federici for their excellent technical assistance.

References (29)

  • X.M. Xie et al.

    State-dependent inhibition of Na+ currents by the neuroprotective agent 619C89 in rat hippocampal neurons and in a mammalian cell line expressing rat brain type IIA Na+ channels

    Neuroscience

    (1996)
  • C. Zona et al.

    Delayed and fast transient potassium currents in rat neocortical neurons in cell culture

    Neurosci. Lett.

    (1988)
  • E. Benoit et al.

    Riluzole specifically blocks inactivated Na channels in myelinated nerve fibre

    Eur. J. Physiol.

    (1991)
  • G. Bensimon et al.

    A controlled trial of riluzole in amyotrophic lateral sclerosis. ALS/Riluzole Study Group

    N. Engl. J. Med.

    (1994)
  • Cited by (111)

    • Riluzole inhibits Kv4.2 channels acting on the closed and closed inactivated states

      2021, European Journal of Pharmacology
      Citation Excerpt :

      Moreover, the inhibition of Kv1.4 channels by riluzole is not voltage-dependent (Xu et al., 2001), and the effect of this compound on Na+ channels is also use-independent (Mestre et al., 2000). Consistent with the hyperpolarizing shift of the steady-state inactivation relationship produced by riluzole on Kv4.3 (Ahn et al., 2006), Kv1.5, Kv3.1 (Ahn et al., 2005), and Na+ channels (Zona et al., 1998), riluzole also provoked this effect on Kv4.2 channels. Furthermore, riluzole accelerated the inactivation rate, the transition of Kv4.2 channels to the closed-inactivated state, and increased the percentage of channels in the closed-inactivated state, indicating that riluzole facilitates the channel transition to the closed-state inactivation.

    • Synthesis and biological evaluation of a new class of benzothiazines as neuroprotective agents

      2017, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Our attention was firstly attracted by riluzole (1, 2-amino-6-(trifluoromethoxy)benzothiazole) (Fig. 1), which has been shown to possess neuroprotective effects in animal models of PD [7], HD [8] and cerebral ischemia [9]. Interestingly, 1 is a small molecule (MW = 234.2) that in vitro may elicit multiple molecular actions, among which those clinically relevant are inhibition of voltage-gated sodium channels [10–13], which can lead to reduced neurotransmitter release, non-competitive inhibition of NMDA receptors [14,15], inhibition of glutamate release [16], and enhanced astrocytic uptake of extracellular glutamate [17]. Currently, 1 is the only drug that has proved able to modify the progression of ALS [18–20] and the only one approved for the treatment of this disease.

    View all citing articles on Scopus
    View full text