Pre-Clinical Models of Acquired Neonatal Seizures: Differential Effects of Injury on Function of Chloride Co-Transporters

Review Article

Austin J Cerebrovasc Dis & Stroke. 2014;1(6): 1026.

Pre-Clinical Models of Acquired Neonatal Seizures: Differential Effects of Injury on Function of Chloride Co-Transporters

Kang SK1 and Kadam SD1,2*

1Neuroscience Laboratory, Hugo Moser Research Institute, USA

2Department of Neurology, Johns Hopkins University, USA

*Corresponding author: Kadam SD, Department of Neurology, Johns Hopkins University School of Medicine, 716 North Broadway, # 426, Baltimore, MD 21205, USA

Received: August 11, 2014; Accepted: October 07, 2014; Published: October 09, 2014

Abstract

Hypoxic-Ischemic Encephalopathy [HIE] represents the most common acquired pathology associated with neonatal seizures. HIE-associated neonatal seizures are often difficult to control, due to their refractoriness to traditional anti-seizure agents. Developmentally regulated chloride gradients during early development make the neonatal brain more seizure-susceptible by depolarizing GABAAR-mediated currents, and therefore hindering inhibition by conventional anti-seizure drugs such as Phenobarbital [PB] and benzodiazepines. Pharmaco-modulation of chloride co-transporters has become a current field of research in treating refractory neonatal seizures, and the basis of two clinical trials [NCT01434225; NCT00380531]. However, the recent termination of NEMO study [NCT01434225] on bumetanide, an NKCC1 antagonist, suggests that clinical utilization of bumetanide as an adjunct to treat neonatal seizures with PB may not be a viable option. Hence, re-evaluation of bumetanide as an adjunct through pre-clinical studies is warranted. Additionally, the model-specific variability in the efficacy of bumetanide in the pre-clinical models of neonatal seizures highlights the differential consequences of insults used to induce seizures in each pre-clinical model as worth exploration. Injury itself can significantly alter the function of chloride co-transporters, and therefore the efficacy of anti-seizure agents that follow.

Keywords: Neonatal seizures; Hypoxic Ischemic

Introduction

Seizures are detected much more frequently during the neonatal period than at any other age [1,2]. Untreated or poorly-controlled seizures in neonates can lead to adverse neuro developmental morbidity and lethality [3-5], and the causative acquired pathologies include intracranial hemorrhage, intracranial infection and Hypoxic- Ischemic Encephalopathy [HIE] [6,7]. HIE has remained the most common underlying pathology, constituting 50-60% of total seizures reported in neonates [7,8]. HIE-associated seizures in neonates display pharmaco-resistance to conventional and first-line antiseizure drugs such as Phenobarbital [PB]and benzodiazepines, which act as agonists to enhance GABAAR-mediated inhibition [2,9,10]. Therapeutic hypothermia has been reported to benefit treatment protocols of anti-seizure pharmacotherapies in neonates [11,12], and is being widely investigated. However, efficacious alternative pharmacotherapies to treat seizures in neonates and bypass the pharmaco-resistance are lacking [9,13]. An attempt to remedy this lack has been made using the NKCC1 blocker, bumetanide, based on the promising data from several pre-clinical models, and two clinical trials [NCT01434225; NCT00380531]were initiated in 2009 to investigate the anti-seizure efficacy of this drug as an adjuvant therapy towards PB-resistant neonatal seizures associated with HIE [14]. However, recent termination of NEMO clinical trial [NCT01434225] that reported non-efficacy of bumetanide and its ototoxicity suggests a need for re-evaluation of potential therapeutic benefit of bumetanide as an anti-seizure adjunct [15].

Intrinsic neuronal hyperexcitability of the immature brain results in a higher seizure susceptibility

The imbalance of excitation and inhibition in a seizing immature brain has been attributed to many factors such as developmentally regulated protein expression and pathology-induced inflammatory response, which ultimately results in neuronal hyperexcitability that leads to the higher seizure susceptibility in neonates [16-18]. During the critical period of synaptogenesis, the developmental regulation of GluR subunits in neurons and glia lowers the threshold for seizures and excitotoxic hypoxic-ischemic injury [16]. Additionally, alteration of proteins such as gephyrin and GABAAR that are crucial for GABA ergic signaling has been proposed as a possible mechanism that results in a diminished inhibition shown in a model with pilocarpine-induced status epilepticus [20].

Another potential factor that has recently emerged is an altered function of chloride co-transporters in immature brain that leads to a reversed movement of chloride ions [Cl-] [21,22]. The neuronal chemical gradient of Cl- is largely driven by membrane-localized chloride co-transporters, specifically Na+K+2Cl- Co-transporter 1 [NKCC1] [23] and K+Cl- Co-transporter 2 [KCC2] [24,25]. NKCC1, present in multiple cell types [26], pumps in Cl-and leads to its intracellular accumulation, whileKCC2, known to be expressed specifically in the CNS [27], extrudes Cl- to maintain a lowintracellular Cl- concentration, which allows for the inhibitory action of GABAAR agonists. In the mature brain, dominant expression of KCC2 over NKCC1 establishes a Cl-gradient that drives Cl-influx upon GABAAR-mediated activation of chloride channels, rendering GABA hyperpolarizing. In contrast, in immature neurons where KCC2 expression is lacking, insufficient Cl- extrusion leads to a high Cl-concentration, resulting in less inhibition or even depolarization in response to GABA. Therefore, pharmaco-modulation of chloride co-transporters to lower Cl- concentrations in immature brains to allow GABAAR-mediated inhibition has been investigated as a promising therapeutic intervention [28,29].

Bumetanideas an adjuvant therapy to PB in neonatal seizures

Clinical trials have investigated bumetanide as an adjunct, administered at a dose of 0.1-0.2mg/kgcon currently with PB to improve the efficacy of PB in neonates with PB-resistant seizures [30]. However, some concerns about bumetanide have been raised [31]. First, bumetanide functions as a potent loop diuretic with a short half-life in neonates [32]. In humans, NKCC2, another target of bumetanide, is robustly expressed in the kidney to extract and reabsorb ions from urine. Repeated doses of bumetanide can non-selectively block NKCC2 as well as NKCC1.Second, bumetanide has poor blood-brain barrier permeability that results in a less than 1% of bumetanide penetrating into the brain one hour after an intra peritoneal injection [33]. Although pathology-induced disruption of blood-brain barrier is expected in acquired neonatal seizures, the limited capability of bumetanide to reach the brain remains a major challenge [29]. Third, a few pre-clinical studies have reported that bumetanide is non-efficacious as an adjunct for anti-seizure pharmacotherapy [34,35]. One explanation for these findings is the possible KCC2 blockage by bumetanide in the seizing immature brain, which may result in an impaired extrusion of neuronal Cl-, further preventing GABAAR-targeted action of conventional anti-seizure agents. Finally, the NKCC1 knockout mouse is deaf, due to the loss of NKCC1 function in inner hair cells [36]. NKCC1 expression in the marginal cells of the inner ear is crucial for normal development of auditory function [36,37]. Therefore, systemic blockage of NKCC1 during the neonatal period could cause significant auditory deficits similar to those now evident in the NEMO study. The same isoform of NKCC1 is expressed in brain, hair cells, and kidney in humans [38], prohibiting alternative strategies like developing a CNS-specific NKCC1 blocker. This last concern became evident in the termination of the NEMO European clinical trial [NCT01434225], due to lack of anti-seizure efficacy of bumetanide associated with ototoxicity [15].

Model-specific variability of efficacy of bumetanide in preclinical studies of neonatal seizures

Understanding the injury-specific alterations of chloride co-transporters is a critical prerequisite for investigating pharmaco-modulation of chloride co-transporters [39]. Neuronal chloride gradients are not only affected by developmental shifts in co-transporter expression but also by the nature of the seizure-inducing injury. Altered function and expression of chloride co-transporters present one of the significant mechanisms for consequential seizure control, since chloride-co-transporters are key regulators of the neuronal Cl-gradient, which determine the efficacy of conventional anti-seizure agents. A few pre-clinical studies have reported the acute functional up regulation of NKCC1upon excitotoxic injury [40,41], which implicates a significant injury-induced alteration in the neuronal Cl-gradient even before the onset of any therapy. This acute post-injury up regulation of NKCC1 function may contribute to an increased Cl-concentration that may further prevent neuronal hyper polarization in response to GABAAR agonists. These pre-clinical observations originally led to the support and initiation of clinical trials on bumetanide as a promising adjuvant for treating refractory seizures in neonates.

Bumetanide has been studied in numerous pre-clinical models of neonatal seizures using chemoconvulsants [42,43], hypoxia [44], and ischemia [45], which all recapitulate the well-established neuronal hyperexcitability and higher seizure susceptibility during early development. However, the potential model-specificity in chloride co-transporter alteration following injury and its effect on the drug efficacy has been largely overlooked in recent reviews [31,46]. Chemoconvulsants, as shown in a kainic acid model, generate a substantial seizure-load that represent the status-like phenotypes frequently associated with HIE. However, chemoconvulsant-induced seizures may not be similar to ischemia- and hypoxia-induced seizures [Table 1]. Chemoconvulsant-related injury has been shown to up regulate KCC2 expression in contrast to an ischemia-related down regulation of KCC2, suggesting a differential modulation of KCC2 and NKCC1 by injury. Comparatively, hypoxic models, utilizing a hypoxia-only insult, consistent induction of transient seizures are reported; however the seizure loads are of a much lower severity compared to the chemoconvulsant and ischemia models. This low seizure-loads which consist of short duration seizures, may not result in excitotoxic injury similar to the other pre-clinical models nor represent the status-like seizure phenotypes associated with long-term morbidity and mortality in HIE [47].

Citation: Kang SK and Kadam SD. Pre-Clinical Models of Acquired Neonatal Seizures: Differential Effects of Injury on Function of Chloride Co-Transporters. Austin J Cerebrovasc Dis & Stroke. 2014;1(6): 1026. ISSN: 2381-9103.