Impairment of dentate gyrus neuronal progenitor cell differentiation in a mouse model of temporal lobe epilepsy

https://doi.org/10.1016/j.expneurol.2006.02.010Get rights and content

Abstract

Unilateral intrahippocampal injection of kainic acid (KA) in adult mice induces an epileptic focus replicating major histopathological features of temporal lobe epilepsy (TLE). In this model, neurogenesis is impaired in the lesioned dentate gyrus, although cell proliferation transiently is increased bilaterally in the subgranular zone (SGZ). To investigate further the relationship between epileptogenesis and neurogenesis, we compared the differentiation of cells born shortly before and after KA injection. Immunohistochemical staining for doublecortin and PSA-NCAM, two markers of young neurons, revealed a rapid downregulation of both markers ipsilaterally, whereas they were increased transiently on the contralateral side. To determine whether KA treatment directly affects neural progenitors in the SGZ, dividing cells were prelabeled with 5′-bromo-2′deoxyuridine (BrdU) treatment before unilateral injection of KA. Double staining with the proliferation marker PCNA showed that prelabeled BrdU cells survived KA exposure and proliferated bilaterally. Unexpectedly, the neuronal differentiation of these cells, as assessed after 2 weeks with doublecortin and NeuN triple-staining, occurred to the same extent as on the contralateral side. Only 5% of pre-labeled BrdU cells were GFAP-positive within the lesion. Therefore, SGZ progenitor cells committed to a neuronal phenotype before KA treatment complete their differentiation despite the rapid down-regulation of doublecortin and PSA-NCAM. These findings suggest impaired fate commitment and/or early differentiation of proliferating cells in the lesioned dentate gyrus. Loss of neurogenesis in this TLE model likely reflects an irreversible alteration of the SGZ germinal niche during development of the epileptic focus and may therefore be relevant for human TLE.

Introduction

Temporal lobe epilepsy with hippocampal sclerosis (TLE-HS) is one of the most common forms of medically refractory epilepsies. Histopathologically, it is characterized by profound neuronal loss in CA1, CA3 and the hilus and a strong reactive astrogliosis (Babb and Najm, 2001). In almost 50% of the patients, the dentate gyrus (DG) is affected by dispersion and partial to severe loss of granule cells (Blumcke et al., 2002). These alterations are accompanied by profound reorganization of neuronal circuits underlying the generation of recurrent seizures. The demonstration that neurogenesis persists in the subgranular zone (SGZ) of human adult hippocampus (Eriksson et al., 1998) and the fact that multipotent progenitor cells can be isolated from resected tissue from epileptic patients (Moe et al., 2005) provide a strong stimulus to investigate whether neurogenesis is related to the pathophysiology of TLE.

Recent studies in patients with TLE-HS reported that cell proliferation (mitotic activity of progenitor cells) is markedly increased throughout the hippocampal formation, in particular in relation to granule cell dispersion (Thom et al., 2002, Crespel et al., 2005). Indirect evidence for neurogenesis (the process by which newborn cells differentiate into neurons) has also been suggested in a study of pediatric specimens with severe seizures (Blumcke et al., 2002). However, other studies observed decreased number of cells positive for polysialylated neural cell-adhesion molecule (PSA-NCAM), a marker of newborn neurons, in both adult and pediatric patients (Mathern et al., 2002, Pirttila et al., 2005). In animal models of TLE, it is well established that induction of acute seizures transiently increases the rate of neurogenesis in the SGZ (Bengzon et al., 1997, Parent et al., 1997, Gray and Sundstrom, 1998, Scott et al., 2000, Hattiangady et al., 2004, Jessberger et al., 2005). A major observation was the abnormal migration of some newly born granule cells into the CA3 area, where they form aberrant connections contributing to increased excitability of hippocampal networks (Scharfman et al., 2000). It remains unsettled, however, whether neurogenesis contributes to epileptogenesis and whether it is sustained in animals experiencing chronic recurrent seizures (Cha et al., 2004, Hattiangady et al., 2004, Mohapel et al., 2004, Jessberger et al., 2005).

Several factors have been identified that might interfere with neurogenesis in TLE. Stem and precursor cells in the SGZ rely on a neurogenic microenvironment and on specific cell–cell interactions that might be disrupted in epileptic tissue (Seki, 2003). Furthermore, inflammation, which often accompanies seizure-related brain injury, can be detrimental for neurogenesis (Ekdahl et al., 2003, Monje et al., 2003). Finally, the severity of the lesion induced upon prolonged experimental status epilepticus negatively influences long-term survival of newborn cells (Mohapel et al., 2004).

We have shown recently in a mouse model of TLE, characterized by a pronounced hypertrophy and dispersion of DG granule cells, that neurogenesis is suppressed selectively in the lesioned area, but not in the contralateral, unaffected hippocampus (Kralic et al., 2005). In this model, an epileptic focus develops unilaterally following intrahippocampal injection of kainic acid (KA) in adult mice, leading to chronic spontaneous recurrent seizures (SRS). KA treatment increased cell proliferation in the SGZ bilaterally. However, the vast majority of newly born cells in the injected DG were astrocytes when examined 4 weeks later. Thus, the neurogenic potential of the SGZ might be disrupted upon development of an epileptic focus. It is unclear, however, whether this effect is secondary to KA toxicity or whether it reflects an alteration of the germinal niche in the lesioned DG.

Here, we investigate further the relationship between epileptogenesis and neurogenesis in the KA mouse model of TLE. Cell proliferation and differentiation during 2 weeks following intrahippocampal KA injection were characterized immunohistochemically with proliferating cell nuclear antigen (PCNA), a marker of actively dividing cells, doublecortin (DCX) and PSA-NCAM, two markers of young neurons, and glial fibrillary acidic protein (GFAP), which labels astrocytes and radial glia/stem cells residing in the SGZ. Furthermore, cells proliferating in the SGZ shortly before KA administration were prelabeled with BrdU and their fate was monitored during the subsequent 2 weeks to determine how and when neurogenesis declines during development of the epileptic focus.

Section snippets

Animals

Experiments were performed on 8- to 10-week-old male Swiss mice (HanIbm:NMRI; Harlan) weighing 30–40 g that were housed in individual cages on a 12 h light/dark cycle (lights on at 7:00 a.m.) with food and water provided ad libitum. Animal procedures were approved by the Cantonal Veterinary Office of Zurich and were done in accordance with the European Community Council Directives of 24 November 1986 (86/609/EEC). All efforts were made to minimize animal suffering and to reduce the number of

KA treatment stimulates cell proliferation in the SGZ

Cell proliferation induced by unilateral intrahippocampal injection of KA was monitored by immunoperoxidase staining for PCNA (Fig. 1). This marker labels cells that are dividing at the time of sacrifice (Kurki et al., 1988). Baseline levels were assessed in control, saline-treated mice (Fig. 1A2). At day 3 post-KA injection, a rapid and marked increase in PCNA-positive cells was observed throughout the hippocampal formation, including the SGZ and the hilus of the DG (Fig. 1C2). The majority of

Discussion

The present results demonstrate that progenitor cells born in the SGZ shortly prior to unilateral intrahippocampal KA injection are not directly affected in their survival by this treatment. Some of these cells continue to proliferate while others differentiate into neurons to the same extent as on the contralateral side. We have shown previously that cells proliferating in the SGZ on the lesioned side mainly differentiate into reactive astrocytes (Kralic et al., 2005). Here, we confirm that

Acknowledgments

We are grateful to Corinne Sidler and Franziska Parpan for excellent technical assistance. This work was supported by Swiss National Science Foundation (National Center of Competence in Research-Neural Plasticity and Repair).

References (48)

  • G. Kempermann et al.

    Milestones of neuronal development in the adult hippocampus

    Trends Neurosci.

    (2004)
  • P. Kurki et al.

    Monoclonal antibodies to proliferating cell nuclear antigen (PCNA)/cyclin as probes for proliferating cells by immunofluorescence microscopy and flow cytometry

    J. Immunol. Methods

    (1988)
  • P. Mohapel et al.

    Status epilepticus severity influences the long-term outcome of neurogenesis in the adult dentate gyrus

    Neurobiol. Dis.

    (2004)
  • B.W. Scott et al.

    Neurogenesis in the dentate gyrus of the rat following electroconvulsive shock seizures

    Exp. Neurol.

    (2000)
  • W. Stockinger et al.

    The reelin receptor ApoER2 recruits JNK-interacting proteins-1 and -2

    J. Biol. Chem.

    (2000)
  • T. Tanaka et al.

    Cdk5 phosphorylation of doublecortin ser297 regulates its effect on neuronal migration

    Neuron

    (2004)
  • T.L. Babb et al.

    Hippocampal sclerosis: pathology, electrophysiology, and mechanisms of epileptogenesis

  • J. Bengzon et al.

    Apoptosis and proliferation of dentate gyrus neurons after single and intermittent limbic seizures

    Proc. Natl. Acad. Sci. U. S. A.

    (1997)
  • I. Blumcke et al.

    Ammon's horn sclerosis: a maldevelopmental disorder associated with temporal lobe epilepsy

    Brain Pathol.

    (2002)
  • V. Bouilleret et al.

    Early loss of interneurons and delayed subunit-specific changes in GABAA-receptor expression in a mouse model of mesial temporal lobe epilepsy

    Hippocampus

    (2000)
  • J.P. Brown et al.

    Transient expression of doublecortin during adult neurogenesis

    J. Comp. Neurol.

    (2003)
  • H.A. Cameron et al.

    Adult neurogenesis produces a large pool of new granule cells in the dentate gyrus

    J. Comp. Neurol.

    (2001)
  • V. des Portes et al.

    So-called ‘cryptogenic’ partial seizures resulting from a subtle cortical dysgenesis due to a doublecortin gene mutation

    Seizure

    (2002)
  • P.S. Eriksson et al.

    Neurogenesis in the adult human hippocampus

    Nat. Med.

    (1998)
  • Cited by (41)

    • Early Seizure Activity Accelerates Depletion of Hippocampal Neural Stem Cells and Impairs Spatial Discrimination in an Alzheimer's Disease Model

      2019, Cell Reports
      Citation Excerpt :

      After severe seizures such as status epilepticus (SE), there is also a rapid increase in proliferation of both NSCs and glia within the first hours to days, and then an increase in newborn neurons starting about 4 days after SE. However, in conditions in which seizures are recurrent, the numbers of newborn neurons are typically reduced (Hattiangady et al., 2004; Ledergerber et al., 2006). AD patients also exhibit epileptiform activity and spontaneous seizures that begin early in disease progression, often in the fifth or sixth decade of life.

    • Trimethyltin intoxication induces the migration of ventricular/subventricular zone cells to the injured murine hippocampus

      2016, NeuroToxicology
      Citation Excerpt :

      This model is useful for studying activation and proliferation of neural stem cells, as well as factors modulating migration of neuronal precursors (McPherson et al., 2011; Casalbore et al., 2010). While some injury models may irreversibly damage or alter the neurogenic microenvironment resulting in increased gliogenesis at the expense of neurogenesis (Monje et al., 2002; Rola et al., 2006; Ledergerber et al., 2006; Kralic et al., 2005), the TMT-injured hippocampus remains permissive for the formation of new neural cells. The primary goal of this study was to determine whether endogenous precursor cells derived from the ventricular/subventricular zone (SVZ) migrate into the hippocampus dentate gyrus following trimethyltin-induced injury.

    • Receptor for Advanced Glycation Endproducts is upregulated in temporal lobe epilepsy and contributes to experimental seizures

      2013, Neurobiology of Disease
      Citation Excerpt :

      When determining the spontaneous epileptic activity ensuing after SE, we found that although the time of onset of HPDs did not change in the absence of either TLR4 or RAGE, their recurrence over the time of monitoring was drastically reduced in both receptor KO vs Wt mice. Our data show that RAGE and TLR4 are also involved in some key neuropathologic changes progressively developing after SE, and before the onset of epilepsy, namely CA1 pyramidal cell loss and neurogenesis (Antonucci et al., 2008; Bouilleret et al., 1999; Duveau and Fritschy, 2011; Heinrich et al., 2006; Kralic et al., 2005; Ledergerber et al., 2006; Pernot et al., 2011). We found exacerbated CA1 neuronal loss in epileptic Rage−/− mice as compared to both Tlr4−/− and Wt mice.

    • Neurogenesis in the epileptic brain: A brief overview from temporal lobe epilepsy

      2011, Pharmacological Reports
      Citation Excerpt :

      Significant evidence for reduced neurogenesis in the KA model of mouse TLE was shown by Kralic and coworkers [39], who demonstrated a correlation between decreased neurogenesis and increased astrocyte production. Additional studies performed by Lederberger and colleagues [42] using the KA mouse model of TLE showed impaired fate commitment and/or early differentiation of proliferating cells in the lesioned DG. The number of spontaneous seizures has a strong impact on the extent of neurogenesis.

    View all citing articles on Scopus
    View full text