U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Noebels JL, Avoli M, Rogawski MA, et al., editors. Jasper's Basic Mechanisms of the Epilepsies [Internet]. 4th edition. Bethesda (MD): National Center for Biotechnology Information (US); 2012.

  • This title is an author manuscript version first made accessible on the NCBI Bookshelf website July 2, 2012.

This title is an author manuscript version first made accessible on the NCBI Bookshelf website July 2, 2012.

Cover of Jasper's Basic Mechanisms of the Epilepsies

Jasper's Basic Mechanisms of the Epilepsies [Internet]. 4th edition.

Show details

Programmed Necrosis After Status Epilepticus

, PhD, , PhD, and , MD, LSc.

Author Information and Affiliations

We review recent progress in the field of seizure-induced neuronal injury. Clinical data suggest that seizures trigger neuronal death, but its mechanism is not well understood. We are beginning to understand the role of neuronal necrosis, the dominant form of death in the adult brain. In a postnatal model of status epilepticus (SE), apoptosis is mainly found in immature neurons, while necrosis is the main form of death in mature neurons, suggesting that the degree of maturity of individual neurons is more important that the level of maturity of the whole organism in determining the mode of neuronal death induced by seizures. It is traditionally thought that necrosis is a passive process which does not require the activation of orderly cell death program(s). Experimental models of SE helped elucidating the contribution of cell death pathways in some active forms of necrosis. SE-induced neuronal necrosis can be an active mechanism requiring the execution of a mitochondrial death program and/or the activation of a caspase cascade. Altogether, these data suggest that the neuronal death landscape is complex and redundant, and that successfully targeting cell death pathways may require polytherapy.

INTRODUCTION

In this review, we will discuss the most recent advances in the field of neuronal injury following epileptic seizures, with an emphasis on the mechanism of neuronal necrosis. The ultrastructure of cell death and the ubiquity of endogenous cell death programs were first described in the 1970’s, and the two main categories of cell death, apoptosis and necrosis, were originally defined according to morphological criteria1. The role of glutamate and its analogues in excitotoxic cell death, and the concept of excitotoxicity -- including its role in seizures-- derived from the pioneering studies of John Olney and his disciples2–5. Multiple cell death factors and cell death programs have been identified in developmental and disease-induced neuronal apoptosis6–8, and there is mounting clinical and experimental evidence of their contribution to seizure-induced neuronal injury9–16. Because these death factors were originally identified in classic apoptosis, any form of cell death in which they are expressed is often called apoptotic, even if its morphology suggests necrosis. This has caused considerable confusion in the literature, and should be discouraged17–20. In this chapter, we will discuss the involvement of cell death factors in morphologically-defined necrosis, the main mode of SE-induced cell death in the adult and even in the developing brain. We find that necrosis is frequently an active form of neuronal death, requiring the expression or activation of some of the same cell death factors usually identified with apoptosis. While this finding raises the hope that targeting common cell death pathways might have therapeutic benefits for both necrosis and apoptosis, the multiplicity and redundancy of cell death pathways for both modes of neuronal death also raises formidable problems when we consider the potential therapeutic applications of these mechanisms.

SEIZURE-INDUCED NEURONAL INJURY

Neuronal Injury and Status Epilepticus

Neuronal injury is widespread in the hippocampus and other brain regions of children or adults who died from SE21–24, or of patients with childhood-onset epilepsy who come to surgery for intractable seizures25, although epidemiologic evidence of the deleterious effects of SE is lacking26. DeGiorgio et al. found decreased hippocampal neuronal densities in five patients who died after SE, compared to epileptic patients without SE and to controls27. Neuron-specific enolase, a marker of neuronal injury, is increased in the serum of patients with SE even when the seizures are non-convulsive28–29. A number of anecdotal studies using MRI and other imaging techniques found cerebral edema acutely, and atrophy chronically, after non-convulsive SE30–32, but other studies failed to find lesions33. Hippocampal atrophy has also been reported after complex or prolonged febrile seizures 34–35. The presence of a normal brain MRI before non-convulsive SE, evolving into brain atrophy after SE, has been documented36, and neuronal loss was found at autopsy in areas that became atrophic after non-convulsive SE37. Atrophy (as seen on MRI) is strongly associated with areas of intense seizure activity in anecdotal cases38–40, and in one instance atrophy was shown at autopsy to correspond to neuronal necrosis and neuronal loss39, suggesting that neuronal loss and cerebral atrophy can result from SE in humans, although the incidence and severity of these complications after SE are not known.

Is Seizure-Associated Neuronal Injury Due To The Seizures Themselves Or To Seizure-Associated Systemic Factors?

Complex Partial SE

As mentioned above, the limited evidence available shows elevation of cell death markers in the serum of patients after non-convulsive complex partial SE, in the absence of systemic complications. However, there is also anecdotal evidence that complex partial SE is not always followed by cerebral atrophy or behavioral deficits41, and there is little evidence of any type of brain damage or neurological deficit after petit mal absence SE42. This suggests that seizures per se can cause neuronal injury in humans, but that seizure type is a key determinant of injury.

Evidence From Animal Models

Meldrum and collaborators proved that seizures in paralyzed, ventilated monkeys caused neuronal loss43–45. The occurrence of neuronal injury in remote areas synaptically connected to the epileptic focus suggested an excitotoxic mechanism46, and Sloviter and Damiona showed that excessive neuronal firing by itself induced neuronal death47. In the immature brain, Thompson et al.10–11 and Sankar et al.12 demonstrated that severe, prolonged seizures cause neuronal death, although the extent of neuronal injury was highly model-dependent48. Recent evidence suggests that under circumstances approximating the clinical situation seen in patients with uncontrolled seizures, the mixture of the death-inducing action of severe seizures with the neuroprotective effect of seizure-associated preconditioning, in which mild seizure-associated injury mitigates the neuronal loss resulting from subsequent severe seizures, results in a neuropathological picture resembling hippocampal sclerosis49. Thus, the experimental evidence seems clear: systemic complications associated with seizures greatly aggravate mortality and morbidity from experimental seizures45;50, but in their absence, seizures per se are quite capable of causing neuronal injury and death, even in the immature brain10–11, and appears to do so by a variety of excitotoxic mechanisms16.

NECROSIS AND APOPTOSIS IN EXPERIMENTAL MODELS OF EPILEPSY

Necrosis And Apoptosis: A Morphological Definition

Necrosis and apoptosis have been originally defined by morphological criteria1. In necrosis, early cytoplasmic changes, including severe mitochondrial and organelle swelling and rupture of the plasma membrane, precede late tigroid condensation of the nucleus with scattered, irregular small chromatin clumps (Figure 1). By contrast, in the early stage of apoptosis, chromatin condensation with loss of the nuclear membrane occurs in the presence of relatively preserved organelles and plasma/mitochondrial membranes. The advanced stages of apoptosis are characterized by the presence of multiple large rounded chromatin masses. The picture can be complicated by the occurrence of both processes in the same cell. Following excitotoxic exposure, for example, apoptotic cells may undergo secondary necrosis, with severe swelling of the organelles and breakdown of the plasma membrane.

Figure 1. Example of necrotic and apoptotic morphologies in rat pup brain.

Figure 1

Example of necrotic and apoptotic morphologies in rat pup brain. Images A–E show the ultrastructure of CA1 neurons in control (A) and experimental animals 6 hours (B), 24 hours (C) and 72 hours (D–E) following SE in postnatal day 14. Control (more...)

Following Status Epilepticus, Necrosis Is The Main Form Of Neuronal Death In The Adult Brain…

In the adult rat brain, several models of SE show that necrosis is the dominant form of neuronal death. In models of SE induced by lithium-pilocarpine or systemic injection of kainic acid in adult rats, Fujikawa et al17–18 and Tokuhara et al51 found necrosis in diverse areas of the brain, including the hippocampus, amygdala, and entorhinal, piriform and frontal cortices 24h–7 days following SE. The apoptotic morphology could not be found even when SE duration is reduced to 60 min, suggesting that seizure severity is not a determining factor in the mode of death.20 While necrosis is certainly the main form of seizure-associated death in the adult brain, apoptotic morphologies have been described in some models of SE, and their role cannot be cannot be disregarded.52–54

… And In The Developing Brain Too

It is now well-established that experimental seizures can induce neuronal injury in the developing brain.12;55–58 Both apoptotic and necrotic morphologies have been clearly reported by light and electron microscopy in the developing brain following SE. In postnatal day 14 (P14) rat pups, apoptosis is notably found in the inner granule cell layer of the dentate gyrus.12;16 In the same rats, SE-induced neuronal injury is extensive in the CA1 pyramidal cell layer. Twenty-four hours after SE, electron microscopy observations showed that 47 of 50 injured CA1 neurons had a necrotic morphology (Figure 1C). Seventy-two hours after SE, 50 of 50 injured neurons were necrotic (Niquet et al, 2007) (Figure 1D–E). In a model of lithium-pilocarpine SE in P12 rat pups, EM observations revealed that necrosis is the mode of death of injured neurons in the mediodorsal nucleus of the thalamus56. Altogether, these results show that necrosis is the main form of SE-induced neuronal death even in the immature brain.

Mechanisms Of Seizure-Induced Neuronal Necrosis

The Traditional View on Necrosis and Apoptosis

Traditionally, necrosis is viewed as a passive mechanism that does not require the activation of orderly cell death programs and/or synthesis of new proteins. However, many histological studies dispute this view. Following experimental SE, morphologically necrotic neurons display TUNEL staining12;59–60, Bax immuoreactivity61, caspase expression16;60, or caspase-independent cell death programs.62 Interestingly, nuclear expression of caspase-3, and nuclear translocation of Apoptosis-Inducing Factor (AIF) were found in injured neurons from patients with temporal lobe epilepsy.63

Are these “apoptotic” factors actively involved in the execution of necrosis? Alternatively, does their expression represent an epiphenomenon? The reduction of neuronal necrosis by caspase inhibitors in several models16;64–66 supports the view that “apoptotic” factors play an important role in neuronal necrosis. We will describe below the experimental data suggesting the existence of active forms of necrosis.

By contrast, apoptotic neuronal death following brain insults is conceptualized as an active form of death which results from the execution of cellular programs that resemble those involved in developmentally programmed cell death. The best characterized programs are the extrinsic and intrinsic pathways, offering many alternative routes of activation of caspases, a family of cysteine proteases.67 When the “extrinsic” pathway of programmed cell death is induced, the first step involves the activation of extracellular cell death receptors of the TNF superfamily, which recruit other proteins to form a complex that activates caspase-8, which in turn activates caspase-3. This “executioner” caspase kills the cell through its widespread proteolytic effects, activating DNA breakdown, inactivating DNA repair enzymes, and attacking the cytoskeleton, among other activities. In the “intrinsic” pathway of programmed cell death, the mitochondrion plays a critical role by releasing cytochrome c from its intermembrane space to the cytosol, where, in association with apoptotic protease-activating factor-1 and dATP, it forms the apoptosome complex, activating caspase-9, which in turn activates caspase-3.68 We show below evidence suggesting that these processes are not exclusively involved in apoptotic death, but can also participate in necrotic forms of seizure-induced neuronal death.

Evidence for Caspase-Dependent Forms of Neuronal Necrosis In Culture

In primary neuronal cultures, chemical hypoxia or glutamate excitotoxicity can activate initiator caspase-9 and cell executioner caspase-3 in necrotic neurons. In the minutes-to-hours following the insult, cytochrome c is released from swollen mitochondria at a time when the nucleus is still intact (a sign of early stage of necrosis) and later co-localizes with active caspase-3, suggesting the activation of the intrinsic pathway. Caspase inhibitors reduce neuronal necrosis, showing that this caspase cascade is not an epiphenomenon but does contribute to neuronal necrosis. We called this active form of necrosis “programmed necrosis”.64–66

Seizures and hypoxia/ischemia profoundly inhibit neuronal protein synthesis69–72 and therefore inhibit processes requiring macromolecular synthesis (such as some forms of apoptosis). In opposition to “classical apoptosis”, “programmed necrosis” does not require protein synthesis. In primary neuronal cultures, the protein synthesis inhibitor cycloheximide reduced caspase-3 activation in staurosporine-induced apoptosis but had no effect on hypoxic neuronal necrosis.64 Since the caspase-3 precursor is already expressed in control cultures, the execution of “programmed necrosis” does not require the energy-intensive process of expressing new genes, which is often necessary in classical apoptosis.

Evidence for a Caspase-Dependent Form of Necrosis Induced By SE

In a lithium-pilocarpine model of SE in rat pups16, upregulation of initiator caspase-8 precedes caspase-3 activation in CA1 (Figure 2). Post-embedding immunohistochemical studies show that caspase-3-immunoreactive CA1 neurons have a necrotic morphology. Pretreatment with a pan-caspase inhibitor reduces neuronal necrosis in CA1, showing that caspase activation is not an epiphenomenon but does contribute to SE-induced neuronal necrosis.16 These results suggest that the caspase cascade of the extrinsic pathway (upregulation of initiator caspase-8 followed by caspase-3 activation) can also contribute to necrotic neuronal death.

Figure 2. Putative neuronal death pathways induced by SE in the immature hippocampus.

Figure 2

Putative neuronal death pathways induced by SE in the immature hippocampus. Fluorescent images show caspase-3a (green), caspase-8 (red), caspase-9a (green) and DCX (red) immunoreactivity and Hoechst staining (chromatin dye, white) in CA1 and the dentate (more...)

There is indirect evidence suggesting that “programmed necrosis” can be found in other models of brain injury. In adult rats, kainic acid-induced SE caused two types of pyramidal cell death in CA1: early necrosis (1 day after SE) and delayed TUNEL-positive and caspase-3-dependent programmed cell death (3–7 days following SE). Despite evidence of caspase-3 activation, apoptotic morphology could not be found at 1, 3 and 7 days following seizures, suggesting that necrosis is the main form of CA1 neuronal death.51

Influence of Cell Maturity and Inflammation on “Programmed Necrosis”

Following brain injury, apoptosis has been considered to be more susceptible to occur in the developing brain than in the adult one,73 maybe because of an age-dependent downregulation of apoptotic death factors, such as caspases.74–75 In the lithium-pilocarpine model of SE in rat pups described above, the same seizures trigger two distinct caspase pathways leading to two different modes of death: the extrinsic pathway leading to necrosis in CA1 (as described above), and the intrinsic pathway (caspase-9 and -3 activation) leading to apoptosis in the dentate gyrus (Figure 2). One possible explanation for these results is the degree of cell maturity of these two neuronal populations. In the dentate gyrus, active caspase-3-immunoreactive cells had features of immature neurons: they expressed doublecortin (a marker of immature neurons76) and failed to express calbindin immunoreactivity (a marker of mature granule cells). Immature dentate gyrus cells may be more susceptible to die by apoptosis than CA1 pyramids, whose maturation occurs much earlier.77–79 From these studies, we may speculate that the degree of cell maturity is more important that the level of maturation of the whole organism. Alternative explanations might involve environmental factors, i.e. independent of the intrinsic properties of the neurons, such as inflammation or metabolic changes.

Caspase-Independent Forms Of Active Necrosis

Caspases are not the only “apoptotic” factors that may be involved in neuronal necrosis (Figure 3). There is evidence that some form of necrosis depend on the release of mitochondrial death factors, such as AIF and endonuclease G. Originally characterized for their role in apoptosis80–82, their release from mitochondria to cytosol and translocation to the nucleus can induce DNA fragmentation in a caspase-independent manner. AIF translocation is involved in many models of neuronal injury often associated with a necrotic morphology. In cultured neurons, neurons with a reduced AIF expression are more resistant to glutamate excitotoxicity or oxygen/glucose deprivation. Harlequin mice (expressing over 80% AIF expression reduction) exhibit less neuronal injury following ischemia or KA-induced seizures.83–85 However, in the latter study84, seizure severity was not properly monitored and it is unclear whether Harlequin and control mice display the same susceptibility to seizures. In a model of lithium-pilocarpine SE in adult rats62, nuclear translocation of mitochondrial factors AIF and endonuclease G occur within 60 min of seizure onset (i.e before sign of irreversible neuronal death), suggesting their involvement in nuclear pyknosis and DNA fragmentation. In the same study, nuclear translocation of other factors (cytochrome c, DNAse II) has been reported, suggesting the activation of various pathways leading to SE-neuronal necrosis.

Figure 3. Putative mechanisms of ‘programmed necrosis’.

Figure 3

Putative mechanisms of ‘programmed necrosis’. Following hypoxia, excitotoxicity and/or seizures, calcium entry into the mitochondria and/or energy failure contribute to mitochondrial swelling, outer membrane rupture, release of mitochondrial (more...)

The term necroptosis has been proposed for a kind of necrotic cell death that can be avoided by inhibiting the activity of the serine/threonine kinase RIP1, either through genetic or pharmacological methods. Necrostatins, specific and potent inhibitors of RIP1, have been used as an operational definition of necroptosis.86–88 By contrast with “programmed necrosis”, necroptosis involves caspase-independent pathways and other cell death factors have been implicated, including cyclophilin D, poly(ADP-ribose) polymerase 1 (PARP-1), and AIF (for review see Galluzzi and Kroemer89). Necroptosis has not been reported following SE, but may contribute to NMDA-induced excitotoxicity in cultured cortical neurons.90

Therapeutic Implication Of Programmed Necrosis

Blocking neuronal necrosis is an important therapeutic goal. Plasma membrane rupture in late stage of necrosis leads to release of cytoplasmic and/or nuclear material in the extracellular space, which may trigger an inflammatory response followed by secondary necrosis in neighboring neurons. The evidence of active forms of necrosis suggests the possibility of therapeutic interventions. An experimental model of SE showed that treatment with the cell permeable, irreversible pan-caspase inhibitor Q-VD-OPH can block active necrosis.16 Intraperitoneal delivery of fusion proteins composed of an anti-apoptotic element and a transduction domain which enables the molecule to cross the blood-brain barrier has shown neuroprective potential in models of SE.91–92 Recent literature has also shown that cytokines, T-cells and macrophages can add to the damage induced by seizures, and that targeting the innate and adaptive immune mechanisms may be a promising therapeutic approach.93 However, the multiplicity of cell death pathways (caspase-dependent, caspase-independent) suggests that no magic bullet will be able to block simultaneously all forms of neuronal death.94

REFERENCES

1.
Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer. 1972;26:239–257. [PMC free article: PMC2008650] [PubMed: 4561027]
2.
Olney JW, Rhee V, Ho OL. Kainic acid: a powerful neurotoxic analogue of glutamate. Brain Res. 1974;7:507–512. [PubMed: 4152936]
3.
Olney JW. The toxic effects of glutamate and related compounds in the retina and the brain. Retina. 1982;2:341–359. [PubMed: 6152914]
4.
Rothman S. Synaptic release of excitatory amino acid neurotransmitter mediates anoxic neuronal death. J Neurosci. 1984;4:1884–1891. [PMC free article: PMC6564878] [PubMed: 6737044]
5.
Choi DW. Glutamate neurotoxicity in cortical cell culture is calcium dependent. Neurosci Lett. 1985;58:293–297. [PubMed: 2413399]
6.
Niquet J, Seo DW, Wasterlain CG. Mitochondrial pathways of neuronal necrosis. Biochem Soc Trans. 2006;34:1347–1351. [PubMed: 17073816]
7.
Ribe EM, Serrano-Saiz E, Akpan N, Troy CM. Mechanisms of neuronal death in disease: defining the models and the players. Biochem J. 2008;415:165–82. [PMC free article: PMC9334905] [PubMed: 18800967]
8.
Lorz C, Mehmet H. The role of death receptors in neural injury. Front Biosci. 2009;14:583–95. [PubMed: 19273087]
9.
Penix LP, Thompson KW, Wasterlain CG. Selective vulnerability to perforant path stimulation: role of NMDA and non-NMDA receptors. Epilepsy Res Suppl. 1996;12:63–73. [PubMed: 9302504]
10.
Thompson KW, Wasterlain CG. Partial protection of hippocampal neurons by MK-801 during perforant path stimulation in the immature brain. Brain Res. 1997;751:96–101. [PubMed: 9098572]
11.
Thompson K, Holm AM, Schousboe A, Popper P, Micevych P, Wasterlain C. Hippocampal stimulation produces neuronal death in the immature brain. Neuroscience. 1998;82:337–348. [PubMed: 9466446]
12.
Sankar R, Shin DH, Liu H, Mazarati A, Pereira de Vasconcelos A, Wasterlain CG. Patterns of status epilepticus-induced neuronal injury during development and long-term consequences. J Neurosci. 1998;18:8382–8393. [PMC free article: PMC6792849] [PubMed: 9763481]
13.
Fujikawa DG. Prolonged seizures and cellular injury: understanding the connection. Epilepsy Behav. 2005;7:S3–11. [PubMed: 16278099]
14.
Niquet J, Liu H, Wasterlain CG. Programmed neuronal necrosis and status epilepticus. Epilepsia. 2005;46:43–48. [PubMed: 15987252]
15.
Henshall DC, Murphy BM. Modulators of neuronal cell death in epilepsy. Curr Opin Pharmacol. 2008;8:75–81. [PubMed: 17827063]
16.
Lopez-Meraz ML, Wasterlain CG, Rocha L, Allen S, Niquet J. Vulnerability of postnatal hippocampal neurons to seizures varies regionally with their maturational stage. Neurobiol Dis. 2010;37:394–402. [PMC free article: PMC2818381] [PubMed: 19879360]
17.
Fujikawa DG, Shinmei SS, Cai B. Lithium-pilocarpine-induced status epilepticus produces necrotic neurons with internucleosomal DNA fragmentation in adult rats. Eur J Neurosci. 1999;11:1605–1614. [PubMed: 10215913]
18.
Fujikawa DG, Shinmei SS, Cai B. Kainic acid-induced seizures produce necrotic, not apoptotic, neurons with internucleosomal DNA cleavage: implications for programmed cell death mechanisms. Neuroscience. 2000;98:41–53. [PubMed: 10858610]
19.
Fujikawa DG. Neuronal death in mesial temporal sclerosis: separating morphology from mechanism. Epilepsia. 2003;44:1607. [PubMed: 14636340]
20.
Fujikawa DG, Zhao S, Ke X, Shinmei SS, Allen SG. Mild as well as severe insults produce necrotic, not apoptotic, cells: evidence from 60-min seizures. Neurosci Lett. 2010;469:333–337. [PubMed: 20026247]
21.
Margerison JH, Corsellis JA. Epilepsy and the temporal lobes. A clinical, electroencephalographic and neuropathological study of the brain in epilepsy, with particular reference to the temporal lobes. Brain. 1966;89:499–530. [PubMed: 5922048]
22.
Corsellis JA, Bruton CJ. Neuropathology of status epilepticus in humans. Adv Neurol. 1983;34:129–39. [PubMed: 6829328]
23.
Sagar HJ, Oxbury JM. Hippocampal neuron loss in temporal lobe epilepsy: correlation with early childhood convulsions. Ann Neurol. 1987;22:334–340. [PubMed: 3674798]
24.
Sinha S, Satishchandra P, Mahadevan A, Bhimani BC, Kovur JM, Shankar SK. Fatal status epilepticus: A clinico-pathological analysis among 100 patients: From a developing country perspective. Epilepsy Res. 2010. in press. [PubMed: 20708383]
25.
Babb TL, Brown WJ. Neuronal, dendritic, and vascular profiles of human temporal lobe epilepsy correlated with cellular physiology in vivo. Adv Neurol. 1986;44:949–966. [PubMed: 3706028]
26.
Camfield PR. Recurrent seizures in the developing brain are not harmful. Epilepsia. 1997;38:735–7. [PubMed: 9186258]
27.
DeGiorgio CM, Tomyasu U, Gott PS, Treiman DM. Hippocampal pyramidal loss in human status epilepticus. Epilepsia. 1992;33:27–29. [PubMed: 1733757]
28.
Rabinowicz AL, Correale JD, Bracht KA, Smith TD, DeGiorgio CM. Neuron-specific enolase is increased after nonconvulsive status epilepticus. Epilepsia. 1995;36:475–479. [PubMed: 7614925]
29.
O’Regan ME, Brown JK. Serum neuron specific enolase: a marker for neuronal dysfunction in children with continuous EEG epileptiform activity. Eur J Paediatr Neurol. 1998:193–197. [PubMed: 10726591]
30.
Chu K, Kang DW, Kim JY, Chang KH, Lee SK. Diffusion-weighted magnetic resonance imaging in nonconvulsive status epilepticus. Arch Neurol. 2001;58:993–998. [PubMed: 11405815]
31.
Lansberg MG, O’Brien MW, Norbash AM, Moseley ME, Morrell M, Albers GW. MRI abnormalities associated with partial status epilepticus. Neurology. 1999;52:1021–1027. [PubMed: 10102423]
32.
Lazeyras F, Blanke O, Zimine I, Delavelle J, Perrig SH, Seeck M. MRI, (1)H-MRS, and functional MRI during and after prolonged nonconvulsive seizure activity. Neurology. 2000;55:1677–1682. [PubMed: 11113222]
33.
Salmenperä T, Kälviäinen R, Partanen K, Mervaala E, Pitkänen A. MRI volumetry of the hippocampus, amygdala, entorhinal cortex, and perirhinal cortex after status epilepticus. Epilepsy Res. 2000;40:155–170. [PubMed: 10863143]
34.
Wieshmann UC, Woermann FG, Lemieux L, Free SL, Bartlett PA, Smith SJ, Duncan JS, Stevens JM, Shorvon SD. Development of hippocampal atrophy: a serial magnetic resonance imaging study in a patient who developed epilepsy after generalized status epilepticus. Epilepsia. 1997;38:1238–1241. [PubMed: 9579926]
35.
VanLandingham KE, Heinz ER, Cavazos JE, Lewis DV. Magnetic resonance imaging evidence of hippocampal injury after prolonged focal febrile convulsions. Ann Neurol. 1998;43:413–426. [PubMed: 9546321]
36.
Pascual-Castroviejo I, Pascual-Pascual SI, Pena W, Talavera M. Status epilepticus-induced brain damage and opercular syndrome in childhood. Dev Med Child Neurol. 1999;41:420–423. [PubMed: 10400177]
37.
Nixon J, Bateman D, Moss T. An MRI and neuropathological study of a case of fatal status epilepticus. Seizure. 2001;10:588–591. [PubMed: 11792162]
38.
Freeman JL, Coleman LT, Smith LJ, Shield LK. Hemiconvulsion-hemiplegia-epilepsy syndrome: characteristic early magnetic resonance imaging findings. J Child Neurol. 2002;17:10–16. [PubMed: 11913562]
39.
Men S, Lee DH, Barron JR, Munoz DG. Selective neuronal necrosis associated with status epilepticus: MR findings. AJNR Am J Neuroradiol. 2000;21:1837–1840. [PMC free article: PMC7974287] [PubMed: 11110535]
40.
Morimoto T, Fukuda M, Suzuki Y, Kusu M, Kida K. Sequential changes of brain CT and MRI after febrile status epilepticus in a 6-year-old girl. Brain Dev. 2002;24:190–193. [PubMed: 11934519]
41.
Neligan A, Shorvon SD. Frequency and prognosis of convulsive status epilepticus of different causes: a systematic review. Arch Neurol. 2010;67:931–940. [PubMed: 20697043]
42.
Thomas P, Valton L, Genton P. Absence and myoclonic status epilepticus precipitated by antiepileptic drugs in idiopathic generalized epilepsy. Brain. 2006;129:1281–1292. [PubMed: 16513683]
43.
Meldrum BS, Brierley JB. Prolonged epileptic seizures in primates. Ischemic cell change and its relation to ictal physiological events. Arch Neurol. 1973;28:10–17. [PubMed: 4629379]
44.
Meldrum BS, Vigouroux RA, Rage P, Brierley JB. Hippocampal lesions produced by prolonged seizures in paralyzed artificially ventilated baboons. Experientia. 1973;29:561–563. [PubMed: 4199751]
45.
Meldrum BS, Vigouroux RA, Brierley JB. Systemic factors and epileptic brain damage. Prolonged seizures in paralyzed, artificially ventilated baboons. Arch Neurol. 1973;29:82–87. [PubMed: 4197956]
46.
Collins RC, Olney JW. Focal cortical seizures cause distant thalamic lesions. Science. 1982;218:177–179. [PubMed: 7123229]
47.
Sloviter RS, Damiano BP. On the relationship between kainic acid-induced epileptiform activity and hippocampal neuronal damage. Neuropharmacology. 1981;20:1003–1011. [PubMed: 7322276]
48.
Sankar R, Shin D, Liu H, Wasterlain C, Mazarati A. Epileptogenesis during development: injury, circuit recruitment, and plasticity. Epilepsia. 2002;43(S5):47–53. [PubMed: 12121295]
49.
Norwood BA, Bumanglag AV, Osculati F, Sbarbati A, Marzola P, Nicolato E, Fabene PF, Sloviter RS. Classic hippocampal sclerosis and hippocampal-onset epilepsy produced by a single “cryptic” episode of focal hippocampal excitation in awake rats. J Comp Neurol. 2010;518:3381–3407. [PMC free article: PMC2894278] [PubMed: 20575073]
50.
Wasterlain CG. Mortality and morbidity from serial seizures: An experimental study. Epilepsia. 1974;15:155–176. [PubMed: 4525503]
51.
Tokuhara D, Sakuma S, Hattori H, Matsuoka O, Yamano T. Kainic acid dose affects delayed cell death mechanism after status epilepticus. Brain Dev. 2007;29:2–8. [PubMed: 16790331]
52.
Sloviter RS, Dean E, Sollas AL, Goodman JH. Apoptosis and necrosis induced in different hippocampal neuron populations by repetitive perforant path stimulation in the rat. J Comp Neurol. 1996 Mar 11;366(3):516–33. [PubMed: 8907362]
53.
Liu H, Cao Y, Basbaum AI, Mazarati AM, Sankar R, Wasterlain CG. Resistance to excitotoxin-induced seizures and neuronal death in mice lacking the preprotachykinin A gene. Proc Natl Acad Sci. 1999;96:12096–12101. [PMC free article: PMC18418] [PubMed: 10518582]
54.
Baille V, Clarke PG, Brochier G, Dorandeu F, Verna JM, Four E, Lallement G, Carpentier P. Soman-induced convulsions: the neuropathology revisited. Toxicology. 2005 Nov 5;215(1–2):1–24. [PubMed: 16054742]
55.
Thompson K, Wasterlain C. Lithium-pilocarpine status epilepticus in the immature rabbit. Brain Res Dev Brain Res. 1997;100:1–4. [PubMed: 9174239]
56.
Kubová H, Druga R, Lukasiuk K, Suchomelová L, Haugvicová R, Jirmanová I, Pitkänen A. Status epilepticus causes necrotic damage in the mediodorsal nucleus of the thalamus in immature rats. J Neurosci. 2001;21:3593–3599. [PMC free article: PMC6762492] [PubMed: 11331388]
57.
Silva AV, Regondi MC, Cipelletti B, Frassoni C, Cavalheiro EA, Spreafico R. Neocortical and hippocampal changes after multiple pilocarpine-induced status epilepticus in rats. Epilepsia. 2005;46:636–642. [PubMed: 15857427]
58.
Nairismagi J, Pitkanen A, Kettunen MI, Kauppinen RA, Kubova H. Status epilepticus in 12-day-old rats leads to temporal lobe neurodegeneration and volume reduction: a histologic and MRI study. Epilepsia. 2006;47:479–488. [PubMed: 16529609]
59.
Charriaut-Marlangue C, Ben-Ari Y. A cautionary note on the use of the TUNEL stain to determine apoptosis. Neuroreport. 1995;7:61–64. [PubMed: 8742417]
60.
Niquet J, Auvin S, Archie M, Seo DW, Allen S, Sankar R, Wasterlain CW. Status epilepticus triggers caspase-3 activation and necrosis in the immature rat brain. Epilepsia. 2007;48:1203–1206. [PubMed: 17441993]
61.
Liu H, Cao Y, Basbaum AI, Mazarati AM, Sankar R, Wasterlain CG. Resistance to excitotoxin-induced seizures and neuronal death in mice lacking the preprotachykinin A gene. Proc Natl Acad Sci U S A. 1999;96:12096–12101. [PMC free article: PMC18418] [PubMed: 10518582]
62.
Zhao S, Aviles ER Jr, Fujikawa DG. Nuclear translocation of mitochondrial cytochrome c, lysosomal cathepsins B and D, and three other death-promoting proteins within the first 60 minutes of generalized seizures. J Neurosci Res. 2010;88:1727–1737. [PubMed: 20077427]
63.
Schindler CK, Pearson EG, Bonner HP, So NK, Simon RP, Prehn JH, Henshall DC. Caspase-3 cleavage and nuclear localization of caspase-activated DNase in human temporal lobe epilepsy. J Cereb Blood Flow Metab. 2006;26:583–589. [PubMed: 16121124]
64.
Niquet J, Baldwin RA, Allen SG, Fujikawa DG, Wasterlain CG. Hypoxic neuronal necrosis: protein synthesis-independent activation of a cell death program. Proc Natl Acad Sci U S A. 2003;100:2825–30. [PMC free article: PMC151425] [PubMed: 12606726]
65.
Niquet J, Seo DW, Allen SG, Wasterlain CG. Hypoxia in presence of blockers of excitotoxicity induces a caspase-dependent neuronal necrosis. Neuroscience. 2006;14:77–86. [PubMed: 16697116]
66.
Seo DW, Lopez-Meraz ML, Allen S, Wasterlain CG, Niquet J. Contribution of a mitochondrial pathway to excitotoxic neuronal necrosis. J Neurosci Res. 2009;87:2087–2094. [PMC free article: PMC2713718] [PubMed: 19235896]
67.
Liou AK, Clark RS, Henshall DC, Yin XM, Chen J. To die or not to die for neurons in ischemia, traumatic brain injury and epilepsy: a review on the stress-activated signaling pathways and apoptotic pathways. Prog Neurobiol. 2003 Feb;69(2):103–142. [PubMed: 12684068]
68.
Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, Wang X. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell. 1997;91:479–489. [PubMed: 9390557]
69.
Wasterlain CG. Inhibition of cerebral protein synthesis by epileptic seizures without motor manifestations. Neurology. 1974;24:175–180. [PubMed: 4855687]
70.
Fando JL, Conn M, Wasterlain CG. Brain protein synthesis during neonatal seizures: An experimental study. Exper Neurol. 1979;63:220–228. [PubMed: 437000]
71.
Dwyer BE, Wasterlain CG. Regulation of the first step of the initiation of brain protein synthesis by guanosine diphosphate. J Neurochem. 1980;34:1639–1647. [PubMed: 7381490]
72.
Dwyer BE, Wasterlain CG. Selective focal inhibition of brain protein synthesis during generalized bicuculline seizures in newborn marmoset monkeys. Brain Res. 1984;308:109–121. [PubMed: 6478197]
73.
Martin LJ, Al-Abdulla NA, Brambrink AM, Kirsch JR, Sieber FE, Portera-Cailliau C. Neurodegeneration in excitotoxicity, global cerebral ischemia, and target deprivation: A perspective on the contributions of apoptosis and necrosis. Brain Res Bull. 1998;46:281–309. [PubMed: 9671259]
74.
Shimohama S, Tanino H, Fujimoto S. Differential expression of rat brain caspase family proteins during development and aging. Biochem Biophys Res Commun. 2001;289:1063–1066. [PubMed: 11741299]
75.
Yakovlev AG, Ota K, Wang G, Movsesyan V, Bao WL, Yoshihara K, Faden AI. Differential expression of apoptotic protease-activating factor-1 and caspase-3 genes and susceptibility to apoptosis during brain development and after traumatic brain injury. J Neurosci. 2001;21:7439–46. [PMC free article: PMC6762901] [PubMed: 11567033]
76.
Francis F, Koulakoff A, Boucher D, Chafey P, Schaar B, Vinet MC, Friocourt G, McDonnell N, Reiner O, Kahn A, McConnell SK, Berwald-Netter Y, Denoulet P, Chelly J. Doublecortin is a developmentally regulated, microtubule-associated protein expressed in migrating and differentiating neurons. Neuron. 1999;23:247–256. [PubMed: 10399932]
77.
Harris KM, Teyler TJ. Developmental onset of long-term potentiation in area CA1 of the rat hippocampus. J Physiol. 1984;346:27–48. [PMC free article: PMC1199482] [PubMed: 6699775]
78.
Bekenstein JW, Lothman EW. An in vivo study of the ontogeny of long-term potentiation (LTP) in the CA1 region and in the dentate gyrus of the rat hippocampal formation. Brain Res Dev. 1991;63:245–251. [PubMed: 1790593]
79.
Bekenstein JW, Lothman EW. A comparison of the ontogeny of excitatory and inhibitory neurotransmission in the CA1 region and dentate gyrus of the rat hippocampal formation. Dev Brain Res. 1991;63:237–243. [PubMed: 1665107]
80.
Parrish J, Li L, Klotz K, Ledwich D, Wang X, Xue D. Mitochondrial endonuclease G is important for apoptosis in C. elegans. Nature. 2001;412:90–94. [PubMed: 11452313]
81.
Li LY, Luo X, Wang X. Endonuclease G is an apoptotic DNase when released from mitochondria. Nature. 2001;412:95–99. [PubMed: 11452314]
82.
Candé C, Cecconi F, Dessen P, Kroemer G. Apoptosis-inducing factor (AIF): key to the conserved caspase-independent pathways of cell death. J Cell Sci. 2002;115:4727–4734. [PubMed: 12432061]
83.
Yu SW, Wang H, Dawson TM, Dawson VL. Poly(ADP-ribose) polymerase-1 and apoptosis inducing factor in neurotoxicity. Neurobiol Dis. 2003;14:303–317. [PubMed: 14678748]
84.
Cheung EC, Melanson-Drapeau L, Cregan SP, Vanderluit JL, Ferguson KL, McIntosh WC, Park DS, Bennett SA, Slack RS. Apoptosis-inducing factor is a key factor in neuronal cell death propagated by BAX-dependent and BAX-independent mechanisms. J Neurosci. 2005;25:1324–1334. [PMC free article: PMC6726004] [PubMed: 15703386]
85.
Culmsee C, Zhu C, Landshamer S, Becattini B, Wagner E, Pellechia M, Blomgren K, Plesnila N. Apoptosis-inducing factor triggered by poly(ADP-ribose) polymerase and Bid mediates neuronal cell death after oxygen-glucose deprivation and focal cerebral ischemia. J Neurosci. 2005;25:10262–10272. [PMC free article: PMC6725791] [PubMed: 16267234]
86.
Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, Blagosklonny MV, El-Deiry WS, Golstein P, Green DR, Hengartner M, Knight RA, Kumar S, Lipton SA, Malorni W, Nunez G, Peter ME, Tschopp J, Yuan J, Piacentini M, Zhivotovsky B, Melino G. Nomenclature Committee on Cell Death 2009. Classification of cell death: recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ. 2009;16(1):3–11. [PMC free article: PMC2744427] [PubMed: 18846107]
87.
Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, Mizushima N, Cuny GD, Mitchison TJ, Moskowitz MA, Yuan J. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol. 2005;1(2):112–9. [PubMed: 16408008]
88.
Degterev A, Hitomi J, Germscheid M, Ch’en IL, Korkina O, Teng X, Abbott D, Cuny GD, Yuan C, Wagner G, Hedrick SM, Gerber SA, Lugovskoy A, Yuan J. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat Chem Biol. 2008 May;4(5):313–21. [PMC free article: PMC5434866] [PubMed: 18408713]
89.
Galluzzi L, Kroemer G. Necroptosis: a specialized pathway of programmed necrosis. Cell. 2008;26;135(7):1161–3. [PubMed: 19109884]
90.
Li Y, Yang X, Ma C, Qiao J, Zhang C. Necroptosis contributes to the NMDA-induced excitotoxicity in rat’s cultured cortical neurons. Neurosci Lett. 2008;447(2–3):120–3. [PubMed: 18723075]
91.
Li T, Fan Y, Luo Y, Xiao B, Lu C. In vivo delivery of a XIAP (BIR3-RING) fusion protein containing the protein transduction domain protects against neuronal death induced by seizures. Exp Neurol. 2006;197(2):301–8. [PubMed: 16336964]
92.
Ju KL, Manley NC, Sapolsky RM. Anti-apoptotic therapy with a Tat fusion protein protects against excitotoxic insults in vitro and in vivo. Exp Neurol. 2008;210(2):602–7. [PMC free article: PMC4782922] [PubMed: 18207142]
93.
Vezzani A, French J, Bartfai T, Baram TZ. The role of inflammation in epilepsy. Nat Rev Neurol. 2011 Jan;7(1):31–40. [PMC free article: PMC3378051] [PubMed: 21135885]
94.
Golstein P, Kroemer G. A multiplicity of cell death pathways. Symposium on Apoptotic and Non-Apoptotic Cell Death Pathways. EMBO reports. 2007:829–833. [PMC free article: PMC1973949] [PubMed: 17721445]
Copyright © 2012, Michael A Rogawski, Antonio V Delgado-Escueta, Jeffrey L Noebels, Massimo Avoli and Richard W Olsen.

All Jasper's Basic Mechanisms of the Epilepsies content, except where otherwise noted, is licensed under a Creative Commons Attribution-NonCommercial-NoDerivs 3.0 Unported license, which permits copying, distribution and transmission of the work, provided the original work is properly cited, not used for commercial purposes, nor is altered or transformed.

Bookshelf ID: NBK98212PMID: 22787596

Views

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...