Elsevier

Neurobiology of Disease

Volume 83, November 2015, Pages 54-66
Neurobiology of Disease

Continuous neurodegeneration and death pathway activation in neurons and glia in an experimental model of severe chronic epilepsy

https://doi.org/10.1016/j.nbd.2015.08.002Get rights and content

Highlights

  • We exploited MP rats sacrificed at different time points after SE.

  • We addressed the relationship between seizure recurrence and cell death.

  • We demonstrated the existence of an ongoing neurodegenerative process.

  • We showed the activation of distinct cell death pathways in neurons and glia.

Abstract

Whether seizures might determine the activation of cell death pathways and what could be the relevance of seizure-induced cell death in epilepsy are still highly debated issues. We recently developed an experimental model of acquired focal cortical dysplasia (the MAM-pilocarpine or MP rat) in which the occurrence of status epilepticus – SE – and subsequent seizures induced progressive cellular/molecular abnormalities and neocortical/hippocampal atrophy. Here, we exploited the same model to verify when, where, and how cell death occurred in neurons and glia during epilepsy course. We analyzed Fluoro Jade (FJ) staining and the activation of c-Jun- and caspase-3-dependent pathways during epilepsy, from few hours post-SE up to six months of spontaneous recurrent seizures. FJ staining revealed that cell injury in MP rats was not temporally restricted to SE, but extended throughout the different epileptic stages. The region-specific pattern of FJ staining changed during epilepsy, and FJ+ neurons became more prominent in the dorsal and ventral hippocampal CA at chronic epilepsy stages. Phospho-c-Jun- and caspase-3-dependent pathways were selectively activated respectively in neurons and glia, at early but even more conspicuously at late chronic stages. Phospho-c-Jun activation was associated with increased cytochrome-c staining, particularly at chronic stages, and the staining pattern of cytochrome-c was suggestive of its release from the mitochondria. Taken together, these data support the content that at least in the MP rat model the recurrence of seizures can also sustain cell death mechanisms, thus continuously contributing to the pathologic process triggered by the occurrence of SE.

Introduction

Several studies in experimental models and patients with chronic epilepsy suggested that recurring seizures could produce long-term alterations in neuronal circuits including synaptic re-organization, hippocampal mossy fiber sprouting, and neurogenesis (Buckmaster and Dudek, 1997, Cha et al., 2004, Gorter et al., 2001, Pitkänen and Sutula, 2002). Whether seizures might eventually lead to the activation of cell death pathways and whether neuronal death could play a role in epileptogenesis is still highly debated (Dingledine et al., 2014). In human patients, diffuse cell injury and death were reported in children with intractable epilepsy (Choi et al., 2009), and longitudinal or cross-sectional MRI studies suggested slowly progressive neocortical and hippocampal atrophy in drug-resistant temporal lobe epilepsy (TLE) cases (Bernhardt et al., 2009, Fuerst et al., 2003). By contrast, in experimental epilepsy models, the prevailing view considered cell death the direct consequence of the initial insult (i.e., status epilepticus or SE) and not significantly related to subsequent seizures (Gorter et al., 2003, Pitkänen et al., 2002).

Among cell death pathways, caspase-3 is the main executioner protease of the apoptotic cascade (Kajta, 2004). Data in experimental models showed that SE triggered caspase-3 activation and cell death (Ferrer et al., 2000, Henshall et al., 2000a) for a limited time-period after the initial insult (Faherty et al., 1999, Narkilahti et al., 2003a) with no direct contribution of further recurring seizures (Pitkänen et al., 2002). The application of caspase-3 inhibitors reduced SE-induced neuronal loss in the rat model based on amygdala stimulation (Narkilahti et al., 2003b). In humans, elevated levels of cleaved caspase-3 were found in the temporal cortex of patients surgically treated for drug-refractory epilepsy (Henshall et al., 2000b). All these data underscored the potential relevance of active caspase-3 in human and experimental epilepsy.

Another cell mechanism possibly leading to neuronal loss is the activation of c-Jun N-terminal kinase (JNK), which in turn activates the transcription factor c-Jun. The JNK pathway is implicated in multiple physiological processes, in particular in the response to stress stimuli (Antoniou and Borsello, 2012). Regarding cell death, c-Jun activation was associated with resistance to death in C57BL/6 mice after kainic acid (Schauwecker, 2000). However, a number of data in different experimental models pointed out that phosphorylated c-Jun (phospho-c-Jun or p-c-Jun) mediated excitotoxic cell death (Ferrer et al., 1996a, Herdegen et al., 1998, Mielke et al., 1999, Yang et al., 1997). Accordingly, inhibiting the JNK pathway with the cell-permeable peptide D-JNKI-1 provided strong neuroprotection against excitotoxicity in cerebral ischemia in vitro and in vivo (Borsello et al., 2003) and reduced excitotoxic neuronal death in kainic acid-induced SE in vivo (Spigolon et al., 2010).

Previous data in experimental epilepsy models, therefore, demonstrated that the occurrence of SE could rapidly activate pathways able to produce cell death. However, whether such activation is only temporally related to the induction of SE, whether and what pathways are activated and what cell types are involved in the cell death processes during the course of chronic seizures remain to be clarified. This is of particular relevance for brain malformations associated with severe drug-resistant epilepsy, such as focal cortical dysplasia (FCD). Apoptotic cell death may occur in FCD patients (Choi et al., 2009, Iyer et al., 2014), and we recently demonstrated reduced neuronal density in dysplastic cortical areas of FCD patients with longer vs shorter epilepsy duration, thus suggesting an ongoing, seizure-related neuronal degeneration in the malformed epileptic human brain (Finardi et al., 2013).

To address the relationship between the recurrence of seizures and the occurrence of cell death, we exploited here an experimental model of acquired FCD, the methylazoxymethanol/pilocarpine rat (MAM-PILO or MP rat, Colciaghi et al., 2011, Colciaghi et al., 2014, Marchi et al., 2006) generated in our lab. In particular, by analyzing MP rats at different time points after SE induction (from few hours up to 6 months after epilepsy onset) we verified (through Fluoro-Jade, caspase-3, and c-Jun labeling) when, where, and how cell death occurred in neurons or glia. We demonstrated the existence of an ongoing neurodegenerative process and the activation of distinct cell pathways in both neuronal and glial cells.

Section snippets

Ethical statement

Procedures were carried out with care to minimize discomfort and pain to treated rats, in compliance with national (D.L. 116 Suppl 40/1992 and D.L. 26/2014) and international guidelines and laws (2010/63/EU Legislation for the protection of animals used for scientific purposes). The experimental protocol was approved by the Ministry of Health, Italy (protocol number: BR1/2012).

MAM and pilocarpine administration

Pregnant Sprague–Dawley rats (Charles River, Calco, Italy) received two intraperitoneal (ip) doses of MAM (15 mg/kg

Seizure onset and evaluation

Four MP rats were sacrificed 18 h after SE onset and referred to as MP-acute. All the remaining 25 MP rats experienced spontaneous seizures after SE. Mean seizure onset was 8.21 ± 4.63 days after pilocarpine-induced SE. Of the 25 rats, 8 rats (referred to as MP-EC) were sacrificed 3–5 days after the onset of the first clinical seizure, 9 rats were sacrificed after 3 and 8 rats after 6 months of chronic epileptic seizures and referred to as MP-3m and MP-6m, respectively. The video-monitoring

Discussion

A bulk of evidence indicated that in experimental models of epilepsy the occurrence of SE was associated with the activation of neurodegenerative processes culminating in cell death, particularly in neurons (do Nascimiento et al., 2012, Liu et al., 2010). Most studies, however, suggested that neuronal cell death was temporally related to the acute phase following SE (Gorter et al., 2003, Pitkänen et al., 2002). In addition, some studies demonstrated that neuronal cell death, particularly in the

Conclusions

In summary, our results clearly indicate that cell death is an ongoing process active throughout all epilepsy stages in the MP model of chronic epilepsy. As already long established by other studies, and confirmed by the present data, SE is the main trigger of the cell damage. However, as clearly demonstrated here, seizure recurrence could not only contribute but also modify the diffusion and progression of cell death mechanisms in neurons as well as in glial cells. Finally, our MP rats could

Conflicts of interest

None declared.

Acknowledgments

The authors wish to thank Prof. Alessandro Vercelli for critically reviewing the manuscript, Dr. Marina Boido for support in the stereological analysis, and Dr. Andrea Legati for support in the statistical analysis. This work was supported by research grants from the Ministry of Health, Italy and AICE-FIRE to GB.

References (69)

  • J. Liu et al.

    Oxidative stress mediates hippocampal neuron death in rats after lithium–pilocarpine-induced status epilepticus

    Seizure

    (2010)
  • N. Marchi et al.

    Determinants of drug brain uptake in a rat model of seizure-associated malformations of cortical development

    Neurobiol. Dis.

    (2006)
  • K. Mielke et al.

    Activity and expression of JNK1, p38 and ERK kinases, c-Jun N-terminal phosphorylation, and c-Jun promoter binding in the adult rat brain following kainate-induced seizures

    Neuroscience

    (1999)
  • S. Narkilahti et al.

    Caspase 6 expression in the rat hippocampus during epileptogenesis and epilepsy

    Neuroscience

    (2005)
  • S. Narkilahti et al.

    Administration of caspase 3 inhibitor during and after status epilepticus in rat: effect on neuronal damage and epileptogenesis

    Neuropharmacology

    (2003)
  • C.R. Pantoja-Jimenez et al.

    Anti-epileptogenic effect of high-frequency stimulation in the thalamic reticular nucleus on PTZ-induced seizures

    Brain Stimul.

    (2014)
  • A. Pitkänen et al.

    Is epilepsy a progressive disorder? Prospects for new therapeutic approaches in temporal-lobe epilepsy

    Lancet Neurol.

    (2002)
  • A. Pitkänen et al.

    Progression of neuronal damage after status epilepticus and during spontaneous seizures in a rat model of temporal lobe epilepsy

    Prog. Brain Res.

    (2002)
  • R.J. Racine

    Modification of seizure activity by electrical stimulation. II. Motor seizure

    Electroencephalogr. Clin. Neurophysiol.

    (1972)
  • P.E. Schauwecker

    Seizure-induced neuronal death is associated with induction of c-Jun N-terminal kinase and is dependent on genetic background

    Brain Res.

    (2000)
  • A. Vercelli et al.

    Human mesenchymal stem cell transplantation extends survival, improves motor performance and decreases neuroinflammation in mouse model of amyotrophic lateral sclerosis

    Neurobiol. Dis.

    (2008)
  • S. Villapol et al.

    Survivin and heat shock protein 25/27 colocalize with cleaved caspase-3 in surviving reactive astrocytes following excitotoxicity to the immature brain

    Neuroscience

    (2008)
  • D.C. Wagner et al.

    Cleaved caspase-3 expression after experimental stroke exhibits different phenotypes and is predominantly non-apoptotic

    Brain Res.

    (2011)
  • L. Wang et al.

    Time-course of neuronal death in the mouse pilocarpine model of chronic epilepsy using Fluoro-Jade C staining

    Brain Res.

    (2008)
  • J. Whitfield et al.

    Dominant-negative c-Jun promotes neuronal survival by reducing BIM expression and inhibiting mitochondrial cytochrome c release

    Neuron

    (2001)
  • Y. Zhao et al.

    The JNK inhibitor D-JNKI-1 blocks apoptotic JNK signaling in brain mitochondria

    Mol. Cell. Neurosci.

    (2012)
  • L. Acarin et al.

    Caspase-3 activation in astrocytes following postnatal excitotoxic damage correlates with cytoskeletal remodeling but not with cell death or proliferation

    Glia

    (2007)
  • K.J. Anderson et al.

    Fluoro-Jade B stains quiescent and reactive astrocytes in the rodent spinal cord

    J. Neurotrauma

    (2003)
  • X. Antoniou et al.

    The JNK signaling transduction pathway in the brain

    Front. Biosci.

    (2012)
  • B.C. Bernhardt et al.

    Longitudinal and cross-sectional analysis of atrophy in pharmacoresistant temporal lobe epilepsy

    Neurology

    (2009)
  • E.H. Bertam

    Temporal lobe epilepsy: where do the seizures really begin?

    Epilepsy Behav.

    (2009)
  • I. Blümcke et al.

    International consensus classification of hippocampal sclerosis in temporal lobe epilepsy: a task force report from the ILAE commission on diagnostic methods

    Epilepsia

    (2013)
  • T. Borsello et al.

    N-methyl-d-aspartate-triggered neuronal death in organotypic hippocampal cultures is endocytic, autophagic and mediated by the c-Jun N-terminal kinase pathway

    Eur. J. Neurosci.

    (2003)
  • P.S. Buckmaster et al.

    Neuron loss, granule cell axon reorganization, and functional changes in the dentate gyrus of epileptic kainate-treated rats

    J. Comp. Neurol.

    (1997)
  • Cited by (0)

    View full text