NKCC1 and KCC2 prevent hyperexcitability in the mouse hippocampus

https://doi.org/10.1016/j.eplepsyres.2008.02.005Get rights and content

Summary

During postnatal development of the central nervous system (CNS), the response of GABAA receptors to its agonist undergoes maturation from depolarizing to hyperpolarizing. This switch in polarity is due to the developmental decrease of the intracellular Cl concentration in neurons. Here we show that absence of NKCC1 in P9–P13 CA3 pyramidal neurons, through genetic manipulation or through bumetanide inhibition, results in a significant increase in cell excitability. Furthermore, the pro-convulsant agent 4-aminopyridine induces seizure-like events in NKCC1-null mice but not in wild-type mice. Measurements of muscimol responses in the presence and absence of NKCC1 shows that the Na–K–2Cl cotransporter only marginally affects intracellular Cl in P9–P13 CA3 principal neurons. However, large increases in intracellular Cl are observed in CA3 pyramidal neurons following increased hyperexcitability, indicating that P9–P13 CA3 pyramidal neurons lack robust mechanisms to regulate intracellular Cl during high synaptic activity. This increase in the Cl concentration is network-driven and activity-dependent, as it is blocked by the non-NMDA glutamate receptor antagonist DNQX. We also show that expression of the outward K–Cl cotransporter, KCC2, prevents the development of hyperexcitability, as a reduction of KCC2 expression by half results in increased susceptibility to seizure under control and 4-AP conditions.

Introduction

The hippocampus is an important CNS structure involved in temporal lobe epilepsy (Aronica et al., 2007, Liu et al., 2007). Inhibition provided by ionotropic GABAA receptors is essential in maintaining normal brain function and protecting against seizures. Indeed, the GABAergic tone, by hyperpolarizing postsynaptic membranes and increasing membrane conductances (shunting), dampens excessive glutamatergic excitation and prevents synchronization of neuronal networks into epileptiform activity. GABAA receptors mediate the hyperpolarization effect through the opening of an anion channel, allowing Cl to move into the cell (and HCO3 to move out under physiological conditions with much less permeability), and leading to plasma membrane hyperpolarization. The inward driving force for Cl ions is due to the low intracellular Cl concentration which is maintained by a secondary active transport mechanism, the neuronal-specific K–Cl cotransporter, KCC2. This overall process matures concomitant with the development of glutamatergic excitation (Stein et al., 2004, Ben-Ari et al., 2007, Ben-Ari, 2007, Represa and Ben-Ari, 2005), which in rodents translates to the first 2 weeks of postnatal life. At birth, however, neuronal Cl is elevated and the equilibrium potential of Cl (ECl) is above the resting membrane potential (Luhmann and Prince, 1991, Owens et al., 1996, LoTurco et al., 1995). This implies that the absence of KCC2 expression at birth, alone, cannot account for the high neuronal Cl.

The formation of neuronal connections during early brain development also depends on the delicate balance between inhibition and excitation (Ben-Ari, 2002). As the rodent brain switches from GABA excitation to GABA inhibition during the first 2 weeks of postnatal life, this period constitutes a critical moment as excessive GABAergic excitation might lead to increased seizure susceptibility (Dzhala and Staley, 2003, Khazipov et al., 2004) while excessive GABAergic inhibition might prevent growth or synapse formation (Ben-Ari, 2002). Based on the observation that NKCC1 expression is high in immature CNS neurons and down-regulated during postnatal development (Plotkin et al., 1997, Wang et al., 2002, Dzhala et al., 2005), several studies have examined the possibility that Cl accumulation in immature neurons is mediated by the inward Na–K–2Cl cotransporter (Fukuda et al., 1998, Sipila et al., 2006, Yamada et al., 2004, Chub et al., 2006, Achilles et al., 2007, Rohrbough and Spitzer, 1996, Ikeda et al., 2003). By using bumetanide to inhibit the Na–K–2Cl cotransporter, these studies show a role for NKCC1 in accumulation of intracellular Cl. Furthermore, a report indicates that NKCC1 may facilitate seizures in the developing brain (Dzhala et al., 2005) by accumulating intracellular Cl in the hippocampal pyramidal neurons and thus attenuating the GABAA receptor-mediated inhibition. However, high external [K+] was used in this study to increase brain hyperexcitability, which is a concern when studying the role of cation-chloride cotransporters, since they are K+-dependent transport pathways, and elevated external K+ increases inward Cl transport. Interestingly, in a recent study where l μM kainate was used as a model, inhibition of NKCC1 by bumetanide was shown to have opposite effect on seizure activities (Kilb et al., 2007). Thus, whether NKCC1 increases excitability by raising Cl in hippocampal pyramidal cells is still unresolved. In fact, several other studies have questioned the role of the cotransporter in Cl accumulation in developing neurons of the brainstem (Balakrishnan et al., 2003) and retina (Zhang et al., 2007). Furthermore, there is evidence that NKCC1 may not be ubiquitously down-regulated in the developing hippocampus, but instead undergoes a change of localization from the soma of interneurons and pyramidal neurons to dendritic compartments (Marty et al., 2002), or from neuronal layers to glial formations (Hubner et al., 2001). These observations are significant since NKCC1-mediated elevation of Cl in GABAergic interneurons may reduce their inhibition by GABA, resulting in increased inhibitory output to pyramidal neurons and higher network activity. Furthermore, glial cells also play an important role in epileptic activity by regulating the K+ ion concentration in the extracellular space (Lux et al., 1986, D’Ambrosio, 2004) and glial NKCC1 may be important for clearance of extracellular K+ (Chen and Sun, 2005). Thus, we wanted to re-address the role of the Na–K–2Cl cotransporter using control saline conditions versus 4-aminopyridine (4-AP) as a convulsive agent, control conditions versus bumetanide as an inhibitor of the cotransporter, and wild-type versus NKCC1 knockout (Delpire et al., 1999) hippocampal slices. To test the 4-AP seizure model, we also use brain slices from older animals, where KCC2 plays a key role in preventing hyperexcitability and where reduction of KCC2 expression by half leads to hyperexcitability in the slice model and increased seizure susceptibility in the whole animal (Woo et al., 2002). In summary, our data show that NKCC1, as KCC2, prevents hyperexcitability and the development of seizures in the hippocampus. However, as NKCC1 was shown to not accumulate Cl in young P9–P13 pyramidal neurons, the protective effect of NKCC1 is likely to occur through a mechanism or mechanisms independent of Cl regulation in the pyramidal cells, such as working as an extracellular K+ clearance pathway or increasing intracellular [Cl] of interneurons.

Section snippets

Animals

Mice used in these experiments were housed in microisolators in a standard animal care facility with a 12 h light/dark cycle, and free access to food and water. All procedures were approved by the Vanderbilt University Animal Care and Use Committee in agreement with the guidelines of the National Institutes of Health Guide for the Care and Use of Laboratory Animals.

Genotyping

Wild-type, heterozygous, and homozygous KCC2 and NKCC1 mice were generated from heterozygous matings. Genotyping was performed by

Results

NKCC1 expression is highest in immature CNS and decreases during postnatal development towards adulthood (Plotkin et al., 1997, Wang et al., 2002, Kanaka et al., 2001, Mikawa et al., 2002, Li et al., 2002). Due to the large inward Na+ and Cl driving force, NKCC1 is a good candidate for intracellular Cl accumulation in immature neurons and GABA excitatory effects. However, other evidences suggest that NKCC1 expression may not be ubiquitously down-regulated in all types of neurons during CNS

Discussion

Recent studies have uncovered multiple roles for the Na–K–2Cl cotransporter, NKCC1, in volume and ion homeostasis in the brain, and in diverse pathologies of the CNS. For instance, the endothelial NKCC1 is involved in the increased salt and fluid movement into the brain that is associated with ischemic brain injury (Pedersen et al., 2006, Chen and Sun, 2005). Furthermore, the cotransporter is also involved in glial cell swelling and glutamate release, leading to neuronal excitotoxicity (Chen

Acknowledgement

This work was supported by a grant from the National Institutes of Health (R01 NS36758) to E. Delpire.

References (62)

  • J.J. LoTurco et al.

    GABA and glutamate depolarize cortical progenitor cells and inhibit DNA synthesis

    Neuron

    (1995)
  • J. Louvel et al.

    Extracellular free potassium during synchronous activity induced by 4-aminopyridine in the juvenile rat hippocampus

    Neurosci. Lett.

    (1994)
  • M.J. Mason et al.

    The interpretation of current-clamp recordings in the cell-attached patch-clamp configuration

    Biophys. J.

    (2005)
  • S. Mikawa et al.

    Developmental changes in KCC1, KCC2 and NKCC1 mRNAs in the rat cerebellum

    Brain Res. Dev. Brain Res.

    (2002)
  • K.L. Perkins

    Cell-attached voltage-clamp and current-clamp recording and stimulation techniques in brain slices

    J. Neurosci. Methods

    (2006)
  • A. Represa et al.

    Trophic actions of GABA on neuronal development

    Trends Neurosci.

    (2005)
  • E.L. Roberts et al.

    Influence of age on the clearance of K+ from the extracellular space of rat hippocampal slices

    Brain Res.

    (1996)
  • P.W.L. Tas et al.

    Characterization of an Na+/K+/Cl co-transport in primary cultures of rat astrocytes

    Biochim. Biophys. Acta

    (1987)
  • C. Wang et al.

    Developmental changes in KCC1, KCC2, and NKCC1 mRNA expressions in the rat brain

    Brain Res. Dev. Brain Res.

    (2002)
  • K. Achilles et al.

    Kinetic properties of Cl uptake mediated by Na+-dependent K+–2Cl cotransport in immature rat neocortical neurons

    J. Neurosci.

    (2007)
  • F.J. Alvarez-Leefmans et al.

    Intracellular chloride regulation in amphibian dorsal root ganglion neurons studied with ion-selective microelectrodes

    J. Physiol. (Lond)

    (1988)
  • M. Avoli et al.

    On the synchronous activity induced by 4-aminopyridine in the CA3 subfield of juvenile rat hippocampus

    J. Neurophysiol.

    (1993)
  • V. Balakrishnan et al.

    Expression and function of chloride transporters during development of inhibitory neurotransmission in the auditory brainstem

    J. Neurosci.

    (2003)
  • T.G. Banke et al.

    GABAergic input onto CA3 hippocampal interneurons remains shunting throughout development

    J. Neurosci.

    (2006)
  • Y. Ben-Ari

    Excitatory actions of gaba during development: the nature of the nurture

    Nat. Rev. Neurosci.

    (2002)
  • Y. Ben-Ari

    GABA, a key transmitter for fetal brain maturation

    Med. Sci. (Paris)

    (2007)
  • Y. Ben-Ari et al.

    GABA: a pioneer transmitter that excites immature neurons and generates primitive oscillations

    Physiol. Rev.

    (2007)
  • H. Chen et al.

    The role of Na–K–Cl co-transporter in cerebral ischemia

    Neurol. Res.

    (2005)
  • H. Chen et al.

    Na(+)-dependent chloride transporter (NKCC1)-null mice exhibit less gray and white matter damage after focal cerebral ischemia

    J. Cereb. Blood Flow Metab.

    (2005)
  • N. Chub et al.

    Chloride-sensitive MEQ fluorescence in chick embryo motoneurons following manipulations of chloride and during spontaneous network activity

    J. Neurophysiol.

    (2006)
  • R.A. DeFazio et al.

    Potassium-coupled chloride cotransport controls intracellular chloride in rat neocortical pyramidal neurons

    J. Neurosci.

    (2000)
  • Cited by (86)

    • GABAergic circuits drive focal seizures

      2023, Neurobiology of Disease
    • Deletion of the Na-K-2Cl cotransporter NKCC1 results in a more severe epileptic phenotype in the intrahippocampal kainate mouse model of temporal lobe epilepsy

      2021, Neurobiology of Disease
      Citation Excerpt :

      Zhu et al. (2008) reported that absence of NKCC1 in neonatal CA3 pyramidal neurons, through genetic manipulation or through bumetanide inhibition, results in a significant increase in cell excitability. Furthermore, the proconvulsant agent 4-aminopyridine induced seizure-like events in NKCC1-null mice but not in wildtype (WT) mice (Zhu et al., 2008). Sipilä et al. (2009) found that hippocampal slices from Nkcc1−/− neonates of the mouse line used in the present study (Flagella et al., 1999) generate endogenous network events similar to giant depolarizing potential (GDPs), most likely based on homeostatic upregulation of intrinsic neuronal excitability as a response to the lack of NKCC1-mediated GABAergic excitation; furthermore, glutamatergic network activity was enhanced in the Nkcc1−/− hippocampus.

    • Disruptions in chloride transporter activity in autism spectrum disorders

      2020, Neuronal Chloride Transporters in Health and Disease
    • The GABA developmental shift in health and disease

      2020, Synapse Development and Maturation: Comprehensive Developmental Neuroscience
    • Postnatal Changes in K <sup>+</sup> /Cl <sup>−</sup> Cotransporter-2 Expression in the Forebrain of Mice Bearing a Mutant Nicotinic Subunit Linked to Sleep-Related Epilepsy

      2018, Neuroscience
      Citation Excerpt :

      Epilepsy, however, comprises a heterogeneous spectrum of disorders (Jensen, 2011). The role of NKCC1 and KCC2 has been extensively studied in parietal and temporal lobe epilepsy (Dzhala et al., 2005; Aronica et al., 2007; Pathak et al., 2007; Li et al., 2008; Zhu et al., 2008; Talos et al., 2012; Awad et al., 2016; Karlócai et al., 2016), but not in frontal epilepsy. The latter presents distinct pathophysiological features, particularly in relation to the sleep-waking cycle.

    View all citing articles on Scopus
    View full text