201
|
Chuang JI, Chen ST, Chang YH, Jen LS. Alteration of Bcl-2 expression in the nigrostriatal system after kainate injection with or without melatonin co-treatment. J Chem Neuroanat 2001; 21:215-23. [PMID: 11382533 DOI: 10.1016/s0891-0618(01)00109-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In order to understand further the role of the anti-apoptotic Bcl-2 proto-oncogene protein in excitotoxin-induced brain injury and possible interaction between Bcl-2 and the antioxidant melatonin, the expression of Bcl-2 in various brain parts was studied after intrastriatal injection of kainate (KA, 2.5 nmol) with or without co-treatment of melatonin (10 mg/kg, intraperitoneally (i.p.)). Three days after unilateral injection of KA to the striatum in the rat, a dramatic direct cytotoxic effect was observed, as indicated an expression of Bcl-2 immunoreactivity in TUNEL- and OX-42-positive cells in the KA-injected striatum and traumatized cortical region. A less severe detrimental effect was also observed in the ipsilateral substantia nigra and peritraumatic cortex, as reflected by an upregulation of Bcl-2-immunostained neurons. Surprisingly, a reduction in Bcl-2-immunoreactive neurons that was accompanied by a less severe loss of tyrosine hydroxylase-immunoreactive neurons in the nigrostriatal pathway was observed after co-treatment with melatonin. Western blot analysis confirmed that Bcl-2 expression is elevated in striatum and cortex on the lesioned side, and that its expression was attenuated substantially after systemic administration of melatonin. The results showing an upregulation of Bcl-2 in nigral neurons and reactive microglia after KA lesion are consistent with the view that Bcl-2 is protective in function in the central nervous system.
Collapse
Affiliation(s)
- J I Chuang
- Department of Physiology, National Cheng Kung University, Taiwan 701, Taiwan, ROC
| | | | | | | |
Collapse
|
202
|
McCullers DL, Herman JP. Adrenocorticosteroid receptor blockade and excitotoxic challenge regulate adrenocorticosteroid receptor mRNA levels in hippocampus. J Neurosci Res 2001; 64:277-83. [PMID: 11319772 DOI: 10.1002/jnr.1076] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The mineralocorticoid receptor (MR) and glucocorticoid receptor (GR) are glucocorticoid-activated transcription factors essential for maintenance of cellular homeostasis. Differential activation of these adrenocorticosteroid receptors (ACR) is thought to influence neuronal viability, particularly under challenging cellular conditions. The present study is designed to determine the effects of receptor blockade and excitotoxic insult on MR and GR mRNA expression and neuronal viability in hippocampus. Male Sprague--Dawley rats were pretreated for 48 hr with vehicle, MR antagonist spironolactone (SPIRO) (50 mg/kg, twice daily, s.c.), or GR antagonist RU486 (25 mg/kg, twice daily, s.c.) and subsequently injected with saline or the glutamate analog kainic acid (KA) (12 mg/kg i.p.). Twenty-four hr post-insult, MR and GR mRNA levels were assessed by in situ hybridization analysis, and hippocampal neurons were counted to assess KA-induced cell loss. MR blockade with SPIRO increased basal MR mRNA levels in hippocampal subregions CA1, CA3, and dentate gyrus (DG) and increased basal GR mRNA levels in CA3. GR blockade with RU486 increased basal GR mRNA levels in CA3. The excitotoxin KA decreased MR mRNA levels in CA1 and CA3, decreased GR mRNA levels in DG, and negated all antagonist-induced increases of ACR mRNAs. Cell counts quantifying KA damage indicated increased CA3 vulnerability to KA insult after treatment with MR antagonist spironolactone but demonstrated no significant cell loss in any other group or region. These results demonstrate dynamic regulation of hippocampal MR and GR mRNAs after ACR antagonist treatment and kainate toxicity, underscoring the potential importance of MR and GR availability to neuronal viability after insult.
Collapse
Affiliation(s)
- D L McCullers
- Department of Anatomy and Neurobiology, University of Kentucky Medical Center, Lexington, Kentucky, USA.
| | | |
Collapse
|
203
|
Zagulska-Szymczak S, Filipkowski RK, Kaczmarek L. Kainate-induced genes in the hippocampus: lessons from expression patterns. Neurochem Int 2001; 38:485-501. [PMID: 11248397 DOI: 10.1016/s0197-0186(00)00101-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Kainate, the analog of the excitatory amino acid L-glutamate, upon binding to non-NMDA glutamate receptors, causes depolarization of neurons followed by severe status epilepticus, neurodegeneration, plasticity and gliosis. These events are best observed in hippocampus, the limbic structure implicated in learning and long-term memory formation. Neurons in all hippocampal structures undergo hyper-activation, however, whereas the cells in the CA subfields degenerate within 2--3 days following the application of kainate, the granule cells of the dentate gyrus are resistant to any form of neurodegeneration and even initiate new synaptic contacts. These physiological and histological changes are modulated by short-term and long-term alterations in gene expression. Perhaps close examination of the changing spatio-temporal patterns of mRNAs of various genes may help in generating a clearer picture of the molecular events leading to complex cognitive functions.
Collapse
Affiliation(s)
- S Zagulska-Szymczak
- Department of Molecular and Cellular Neurobiology, Nencki Institute, Pasteura 3, 02-093 Warsaw, Poland
| | | | | |
Collapse
|
204
|
Roth SU, Sommer C, Mundel P, Kiessling M. Expression of synaptopodin, an actin-associated protein, in the rat hippocampus after limbic epilepsy. Brain Pathol 2001; 11:169-81. [PMID: 11303792 PMCID: PMC8098178 DOI: 10.1111/j.1750-3639.2001.tb00389.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Synaptopodin, a 100 kD protein, associated with the actin cytoskeleton of the postsynaptic density and dendritic spines, is thought to play a role in modulating actin-based shape and motility of dendritic spines during formation or elimination of synaptic contacts. Temporal lobe epilepsy in humans and in rats shows neuronal damage, aberrant sprouting of hippocampal mossy fibers and subsequent synaptic remodeling processes. Using kainic acid (KA) induced epilepsy in rats, the postictal hippocampal expression of synaptopodin was analyzed by in situ hybridization (ISH) and immunohistochemistry. Sprouting of mossy fibers was visualized by a modified Timm's staining. ISH showed elevated levels of Synaptopodin mRNA in perikarya of CA3 principal neurons, dentate granule cells and in surviving hilar neurons these levels persisted up to 8 weeks after seizure induction. Synaptopodin immunoreactivity in the dendritic layers of CA3, in the hilus and in the inner molecular layer of the dentate gyrus (DG) was initially reduced. Eight weeks after KA treatment Synaptopodin protein expression returned to control levels in dendritic layers of CA3 and in the entire molecular layer of the DG. The recovery of protein expression was accompanied by simultaneous supra- and infragranular mossy fiber sprouting. Postictal upregulation of Synaptopodin mRNA levels in target cell populations of limbic epilepsy-elicited damage and subsequent Synaptopodin protein expression largely co-localized with remodeling processes as demonstrated by mossy fiber sprouting. It may thus represent a novel postsynaptic molecular correlate of hippocampal neuroplasticity.
Collapse
Affiliation(s)
- S U Roth
- Department of Neuropathology, University of Heidelberg, Germany.
| | | | | | | |
Collapse
|
205
|
Abstract
Clinical studies of the treatment of status epilepticus are extremely difficult to carry out, therefore a paucity of new clinical studies have been reported. Much of the progress regarding the therapy of status epilepticus has come from a better understanding of the epidemiology of status epilepticus and its consequences and from laboratory studies of experimental status. Status epilepticus has been used as an experimental tool to study epileptogenesis, but from such studies have come insights that can be applied to the therapy of status epilepticus itself. This review will focus on information from epidemiological, experimental, and clinical studies of status epilepticus, which may contribute to the improved treatment of this life-threatening disorder.
Collapse
Affiliation(s)
- D M Treiman
- University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901, USA.
| |
Collapse
|
206
|
Abstract
In human epilepsy, diurnal variation in seizure phenomena suggests the involvement of a time-dependent biological signal. Clinical evidence indicates that in some cases, temporal clustering of epileptic seizures is in phase with the nocturnal rise in circulating melatonin. Although this hormone has been reported to stabilize the brain against seizure-producing stimuli, these pharmacological doses are not representative of physiological conditions but would nonetheless facilitate widespread inhibitory neurotransmission characteristic of traditional anticonvulsants. Instead, it is proposed that endogenous melatonin contributes to epileptiform activity through inhibitory actions on dopaminergic activity. Dopamine is considered a natural downregulator of seizure activity in a number of species, including humans, and numerous lines of evidence suggest that melatonin is capable of stimulating a decrease in dopamine output within areas of the brain thought to participate in the control of epileptic seizures. Pharmacological manipulation of the endogenous melatonin rhythm may provide a useful therapeutic strategy against the occurrence of seizures during increased hormone production.
Collapse
Affiliation(s)
- L S Stewart
- Department of Physiology, The University of Western Ontario, London, Ontario Canada N6A 3C1.
| |
Collapse
|
207
|
Szabó G, Kartarova Z, Hoertnagl B, Somogyi R, Sperk G. Differential regulation of adult and embryonic glutamate decarboxylases in rat dentate granule cells after kainate-induced limbic seizures. Neuroscience 2001; 100:287-95. [PMID: 11008167 DOI: 10.1016/s0306-4522(00)00275-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
In adult brain, the inhibitory GABAergic neurons utilize two distinct molecular forms of the GABA-synthesizing enzyme glutamate decarboxylase (GAD), GAD65 and GAD67. During embryonic development, two truncated forms of GAD67 are also expressed (GAD25 and GAD44), which are translated from two embryonic-specific splice variants of GAD67 messenger RNA. It has recently been established that the excitatory dentate granule cells, in addition to the neurotransmitter glutamate, also contain low levels of GABA and GAD67, which are increased after limbic seizures. To study the seizure-induced activation of glutamate decarboxylase, we investigated the expression of both embryonic and adult glutamate decarboxylase messenger RNAs in the adult rat hippocampus after kainic acid administration by semi-quantitative reverse transcription-coupled polymerase chain reaction, in situ hybridization and immunoblotting. We observed a rapid induction of the embryonic glutamate decarboxylase messenger RNA in the granule cells of dentate gyrus. The expression of embryonic glutamate decarboxylase transcripts, identified here as the splice variant that contains exon 7/B, peaked at about 2h after kainic acid injection and gradually returned to nearly basal levels by 24h. Strikingly, this transient induction of embryonic glutamate decarboxylase messenger RNA was not accompanied by concomitant synthesis of its corresponding protein product GAD25. In contrast, the adult GAD67 messenger RNA and protein were both clearly up-regulated in granule cells, albeit with a certain delay, reaching a maximum around 4-6h after kainic acid injection and gradually returned to control levels by 24h. GAD65 remained unchanged at both messenger RNA and protein levels during the studied period. These characteristic and highly reproducible changes in the synthesis of glutamate decarboxylases indicate that GAD67 is the predominant form of glutamate decarboxylases involved in the elevated synthesis of GABA during seizures and suggest that the transient induction of the embryonic GAD67 messenger RNA that contains exon 7/B, but not GAD25 protein, may exert a role solely in the subsequent up-regulation of adult GAD67 transcription. Expression of the messenger RNA encoding for an alternatively spliced, truncated form of the GABA-synthesizing enzyme glutamate decarboxylase was detected in dentate granule cells briefly after kainic acid-induced seizures. Just as during embryonic development, expression of the alternatively spliced messenger RNA was transient and followed by transcription of its adult form, indicating a possible recapitulation of an embryonic program of gene expression in adult granule cells after epileptic seizures.
Collapse
Affiliation(s)
- G Szabó
- Laboratory of Molecular Neurogenetics BRC, Institute of Biochemistry, 6701, Szeged, Hungary
| | | | | | | | | |
Collapse
|
208
|
Blondeau N, Plamondon H, Richelme C, Heurteaux C, Lazdunski M. K(ATP) channel openers, adenosine agonists and epileptic preconditioning are stress signals inducing hippocampal neuroprotection. Neuroscience 2001; 100:465-74. [PMID: 11098109 DOI: 10.1016/s0306-4522(00)00304-3] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Many models of induced ischemic and epileptic tolerance have now been described in the brain. Although detailed mechanisms underlying such protections still remain largely unknown, induction of heat shock proteins is amongst the endogenous responses believed to play an important role in cellular defense mechanisms. This study reveals that the development of epileptic tolerance also coincides with the induction of the 70,000 mol. wt heat shock protein expression within the time window of protection. Adenosine agonists or ATP-sensitive potassium channel openers have also been shown to exert strong neuroprotective effects when injected shortly prior to a severe ischemic or epileptic insult. The present work shows that adenosine receptor activation and ATP-sensitive potassium channel opening induce 70,000 mol. wt heat shock protein expression in the rat hippocampus and are able to mimic neuroprotection driven by preconditioning. R-phenylisopropyladenosine, a purine agonist, or (-)cromakalim, an ATP-sensitive potassium channel opener, was administered three days prior to a lethal ischemic or epileptic episode to mimic preconditioning. Neurodegeneration was assessed using Cresyl Violet staining and cellular DNA fragmentation visualized by the terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'-triphosphate-biotin nick end labeling method. 70, 000 mol. wt heat shock protein expression was analysed by western blotting and immunohistochemistry. The results show a long-lasting neuroprotection induced by activation of adenosine receptors or ATP-sensitive K(+) channels as early as three days prior to induction of a severe ischemic or epileptic challenge. This protective effect is associated with enhanced 70,000 mol. wt heat shock protein expression also occurring three days following administration of R-phenylisopropyladenosine or (-)cromakalim. These findings support the idea that preconditioning doses of R-phenylisopropyladenosine and (-)cromakalim act as mild cellular stresses inducing neuroprotection in a manner similar to a mild kainate treatment prior to a lethal ischemic or severe epileptic insult three days later. They also suggest that a delayed 70,000 mol. wt heat shock protein expression induced by excitatory neuronal stresses such as short ischemia, mild kainic acid treatment or activation of adenosine receptors and ATP-sensitive potassium channels is predictive of neuronal survival against a subsequent lethal injury.
Collapse
Affiliation(s)
- N Blondeau
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, UPR 411 660 route des Lucioles, Sophia Antipolis, 06560, Valbonne, France
| | | | | | | | | |
Collapse
|
209
|
Bourne JA, Fosbraey P, Halliday J. Changes in striatal electroencephalography and neurochemistry induced by kainic acid seizures are modified by dopamine receptor antagonists. Eur J Pharmacol 2001; 413:189-98. [PMID: 11226392 DOI: 10.1016/s0014-2999(01)00747-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We investigated the involvement of striatal dopamine release in electrographic and motor seizure activity evoked by kainic acid in the guinea pig. The involvement of the dopamine receptor subtypes was studied by systemic administration of the dopamine D(1) receptor antagonist, R(+)-7-chloro-8-hydroxy-3-methyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine hydrochloride (SCH 23390; 0.5 mg kg(-1)), or the dopamine D(2) antagonist, (5-aminosulphonyl)-N-[(1-ethyl-2-pyrrolidinyl)-methyl]-2-methoxybenzamide (sulpiride, 30 mg kg(-1)). Microdialysis and high performance liquid chromatography were used to monitor changes in extracellular levels of striatal dopamine and its metabolites, glutamate, aspartate and gamma-amino-butyric acid (GABA). These data were correlated with changes in the striatal and cortical electroencephalographs and clinical signs. We found that, although neither dopamine receptor antagonist inhibited behavioural seizure activity, blockade of the dopamine D(1)-like receptor with SCH 23390 significantly reduced both the 'power' of the electrical seizure activity and the associated change in extracellular striatal concentration of glutamate, whilst increasing the extracellular striatal concentration of GABA. In contrast, blockade of the dopamine D(2)-like receptor with sulpiride significantly increased the extracellular, striatal content of glutamate and the dopamine metabolites. These results confirm previous evidence in other models of chemically-evoked seizures that antagonism of the dopamine D(1) receptor tends to reduce motor and electrographic seizure activity as well as excitatory amino-acid transmitter activity, while antagonism of the dopamine D(2) receptor has relatively less apparent effect.
Collapse
Affiliation(s)
- J A Bourne
- Biomedical Sciences Department, CBD Porton Down, Salisbury SP4 0JQ, UK.
| | | | | |
Collapse
|
210
|
Fujita M, Aihara N, Yamamoto M, Ueki T, Asai K, Tada T, Kato T, Yamada K. Regulation of rat hippocampal neural cadherin in the kainic acid induced seizures. Neurosci Lett 2001; 297:13-6. [PMID: 11114473 DOI: 10.1016/s0304-3940(00)01635-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Regulation of neural (N-) cadherin expression in the hippocampus was examined by in situ hybridization and immunohistochemistry methods in the rat model of kainic acid (KA) induced seizures. After 12 and 24 h of KA administration, mRNA expression level of N-cadherin decreased in the hippocampal CA1 and CA3 area in parallel with decrease of the number of neural cells. In contrast, after 48 h and 7 days, mRNA expression level recovered partially, although the number of neural cells remained small. In addition, immunohistochemical staining indicated that N-cadherin protein expression of survived neurons increased significantly after 48 h of KA administration. These results indicated that N-cadherin might be involved in neuronal reconstruction at the hippocampus.
Collapse
Affiliation(s)
- M Fujita
- Department of Neurosurgery, Nagoya City University Medical School, 1 Kawasumi, Mizuho-cho, Mizuho-ku, 467-8601, Nagoya, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
211
|
Dube C, Chen K, Eghbal-Ahmadi M, Brunson K, Soltesz I, Baram TZ. Prolonged febrile seizures in the immature rat model enhance hippocampal excitability long term. Ann Neurol 2001. [DOI: 10.1002/1531-8249(200003)47:3<336::aid-ana9>3.0.co;2-w] [Citation(s) in RCA: 233] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
212
|
Eriksson C, Zou LP, Ahlenius S, Winblad B, Schultzberg M. Inhibition of kainic acid induced expression of interleukin-1 beta and interleukin-1 receptor antagonist mRNA in the rat brain by NMDA receptor antagonists. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2000; 85:103-13. [PMID: 11146112 DOI: 10.1016/s0169-328x(00)00251-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The cytokines interleukin-1 beta (IL-1 beta) and IL-1 receptor antagonist (IL-1ra) are rapidly induced in response to excitotoxic and ischemic brain damage. The aim of the present study was to investigate the influence of a non-competitive (dizocilpine maleate, MK-801) and a competitive ((R)-CPP) NMDA receptor antagonist on the transient cytokine expression in the rat brain induced by systemic kainic acid administration. Peripheral administration of kainic acid (10 mg/kg, i.p.) results in a transient expression of IL-1 beta and IL-1ra mRNA, mainly in microglia, in regions showing neurodegeneration such as the hippocampus, thalamus, amygdala, and certain cortical regions. In addition, a few neurons expressing IL-1ra mRNA were observed in the piriform cortex and amygdala following kainic acid injection. Administration of MK-801 (i.p.) 1 h prior to kainic acid injection reduced cytokine expression in all of these regions. MK-801 at 3.0 mg/kg decreased the IL-1 beta mRNA expression, blocked or decreased the IL-1ra mRNA expression, depending on the brain region. MK-801 at 5.0 mg/kg abolished IL-1ra mRNA expression in all of the regions, whereas the IL-1 beta mRNA expression was decreased or blocked, depending on the brain region, or the time point investigated. Peripheral administration of (R)-CPP (15 mg/kg, i.p.) 15 min prior to the kainic acid injection abolished the IL-1 beta mRNA expression. The IL-1ra mRNA expression was abolished in all regions except for a few neurons in the piriform cortex. The finding that NMDA receptor antagonists inhibit the IL-1 beta and IL-1ra mRNA synthesis induced by kainic acid suggests that NMDA receptor activation may be involved in triggering cytokine synthesis following excitotoxic brain damage.
Collapse
Affiliation(s)
- C Eriksson
- Division of Geriatric Medicine, NEUROTEC, Karolinska Institute, Huddinge Hospital, Novum, S-141 86 Huddinge, Sweden
| | | | | | | | | |
Collapse
|
213
|
Schauwecker PE. Seizure-induced neuronal death is associated with induction of c-Jun N-terminal kinase and is dependent on genetic background. Brain Res 2000; 884:116-28. [PMID: 11082493 DOI: 10.1016/s0006-8993(00)02888-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Previous studies have shown that expression of c-Jun protein, as well as the c-Jun amino-terminal kinase (JNK) group of mitogen-activated protein kinases, may play a critical role in the pathogenesis of glutamate neurotoxicity. In order to define the molecular cascade that leads to c-Jun activation following excitotoxic injury and delineate whether induction of protein synthesis is related to cell death signaling cascades or those changes associated with increased seizure activity, we examined the expression of JNK-1, as well as its substrate, c-Jun and N-terminal phosphorylated c-Jun following kainic acid (KA) administration in two strains of mice. In the present study, we assessed the immunohistochemical expression of these proteins at time points between 2 h and 7 days, in excitotoxic cell death-resistant (C57BL/6) and -susceptible (FVB/N) mouse strains that were systemically injected with saline or kainic acid. No strain-related differences in the immunohistochemical expression of any of the proteins were observed in intact control mice. However, following KA administration, the magnitude and period of induction of JNK-1 protein was associated with impending cell death, while increased phosphorylation of c-Jun protein was associated with resistance to cell death. In contrast, expression of c-Jun protein does not appear to be a reliable indicator of impending cell death, as it was expressed in resistant and vulnerable subfields in mice susceptible to kainate injury. These results provide the first evidence that JNK-1 expression may be involved in producing the neuronal cell death response following excitotoxin-induced injury.
Collapse
Affiliation(s)
- P E Schauwecker
- Department of Cell and Neurobiology, University of Southern California, Keck School of Medicine, BMT 401, 1333 San Pablo Street, Los Angeles, CA 90033, USA.
| |
Collapse
|
214
|
Söhl G, Güldenagel M, Beck H, Teubner B, Traub O, Gutiérrez R, Heinemann U, Willecke K. Expression of connexin genes in hippocampus of kainate-treated and kindled rats under conditions of experimental epilepsy. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2000; 83:44-51. [PMID: 11072094 DOI: 10.1016/s0169-328x(00)00195-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We have analyzed whether the expression of connexin genes is altered in the hippocampus of kindled and kainate-treated rats, i.e., animal models of human temporal lobe epilepsy. We have tested this hypothesis by analyzing mRNA, protein abundance and cellular location of connexins (Cx) 43, 36, 32 and 30. The expression of glial fibrillary acid protein and mRNA was also monitored both in kainate-treated and kindled rats, in order to take into account reactive gliosis under these conditions. We found significantly increased expression of GFAP mRNA (100%) and protein (178%) in kainate-treated rats 4 weeks after kainate application, whereas in kindled rats only moderate increases of GFAP mRNA and protein were detected 2-3 weeks (group 2) or 4-6 weeks (group 1) after the last stage 5 induced seizure. Under gliotic conditions, connexins 43 and 30 mRNA or protein expression in astrocytes of kainate-treated rats were nearly unaffected. Cx36 mRNA expression (presumably in neurons) was significantly reduced (44%), whereas abundance of Cx36 protein was only slightly reduced. In both groups of kindled rats, Cx30 and Cx43 mRNA or protein expression were either slightly decreased or unchanged. Again, Cx36 mRNA and protein expression were reduced by about half in group 2. Immunofluorescence analysis of Cx43, Cx36 and Cx30 expression revealed that 4 weeks after the last kainate administration or kindling, cellular localization of these connexins was indistinguishable from control animals.
Collapse
Affiliation(s)
- G Söhl
- Abteilung für Molekulargenetik, Institut für Genetik, Universität Bonn, 53117, Bonn, Germany
| | | | | | | | | | | | | | | |
Collapse
|
215
|
McNamara RK, Lenox RH. Differential regulation of primary protein kinase C substrate (MARCKS, MLP, GAP-43, RC3) mRNAs in the hippocampus during kainic acid-induced seizures and synaptic reorganization. J Neurosci Res 2000; 62:416-26. [PMID: 11054811 DOI: 10.1002/1097-4547(20001101)62:3<416::aid-jnr12>3.0.co;2-v] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the mature hippocampus, kainic acid seizures lead to excitotoxic cell death and synaptic reorganization in which granule cell axons (mossy fibers) form ectopic synapses on granule cell dendrites. In the present study, we examined the expression of four major, developmentally regulated protein kinase C (PKC) substrates (MARCKS, MLP, GAP-43, RC3), which have different subcellular and regional localizations in the hippocampus at several time points (6 hr, 12 hr, 18 hr, 24 hr, 48 hr, 5 days, or 15 days) following kainic acid seizures using in situ hybridization. Consistent with previous reports, following kainate seizures, GAP-43 mRNA expression exhibited a delayed and protracted elevation in the granule cell layer, which peaked at 24 hr, whereas expression in fields CA1 and CA3 remained relatively unchanged. Conversely, RC3 mRNA expression exhibited a delayed reduction in the granule cell layer that was maximal at 18 hr, as well as a reduction CA1 at 48 hr, whereas CA3 levels did not change. MARCKS mRNA expression in the granule cell layer and CA1 remained stable following kainate, although an elevation was observed in subfield CA3c at 12 hr. Similarly, MLP mRNA expression did not change in the granule cell layer or CA1 following kainate but exhibited a protracted elevation in subfields CA3b,c beginning at 6 hr post-kainate. Collectively these data demonstrate that different PKC substrate mRNAs exhibit unique expression profiles and regulation in the different cell fields of the mature hippocampus following kainic acid seizures and during subsequent synaptic reorganization. The expression profiles following kainate seizures bear resemblance to those observed during postnatal hippocampal development, which may indicate the recruitment of common regulatory mechanisms.
Collapse
Affiliation(s)
- R K McNamara
- Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia 19104-6140, USA.
| | | |
Collapse
|
216
|
Fujikawa DG, Shinmei SS, Cai B. Seizure-induced neuronal necrosis: implications for programmed cell death mechanisms. Epilepsia 2000; 41 Suppl 6:S9-13. [PMID: 10999512 DOI: 10.1111/j.1528-1157.2000.tb01549.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
PURPOSE To determine definitively the morphology of neuronal death from lithium-pilocarpine (LPC)-and kainic acid (KA)-induced status epilepticus (SE), and to correlate this with markers of DNA fragmentation that have been associated with cellular apoptosis. Endogenous glutamate release is probably responsible for neuronal death in both seizure models, because neuronal death in both is N-methyl-D-aspartate receptor-mediated. METHODS SE was induced for 3 hours in adult male Wistar rats with either LPC or KA, and 24 or 72 hours later the rats were killed. One group of rats had brain sections, stained with hematoxylin and eosin and the terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) technique, examined by light microscopy and by electron microscopy. A separate group of rats had DNA extracted from the same brain regions examined by electron microscopy in the first group. The extracted DNA was electrophoresed on an agarose gel with ethidium bromide and was examined for the presence or absence of internucleosomal DNA cleavage (DNA "laddering"). RESULTS Twenty-four and 72 hours after 3 hours of LPC- or KA-induced SE, neuronal death in the hippocampus, amygdala, and piriform, entorhinal, and frontal cortices was morphologically necrotic, in spite of DNA laddering in these regions 24 and 72 hours after SE and positive TUNEL staining in some of the regions 72 hours after SE. Ultrastructurally, necrotic neurons were dark and shrunken, with cytoplasmic vacuoles and pyknotic nuclei with small, irregular, dispersed chromatin clumps. CONCLUSIONS Our results, together with those of other reports, suggest that programmed cell death-promoting mechanisms are activated by SE in neurons that become necrotic rather than apoptotic and point to the possibility that such mechanisms may contribute to SE-induced neuronal necrosis.
Collapse
Affiliation(s)
- D G Fujikawa
- Neurology Department, Sepulveda Ambulatory Care Center, VA Greater Los Angeles Healthcare System, California 91343, USA.
| | | | | |
Collapse
|
217
|
Rocha L, González-Trujano ME, Jiménez G, Gaona A, Ondarza R. Characterization of benzodiazepine receptor binding in immature rat brain after kainic acid administration. Epilepsia 2000; 41 Suppl 6:S44-7. [PMID: 10999518 DOI: 10.1111/j.1528-1157.2000.tb01555.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE To evaluate the effects of status epilepticus on benzodiazepine (BDZ) receptor binding in immature rat brain. METHODS Twenty-four immature (15 days old) and six adult (90 days old) rats were used in this study. Status epilepticus was induced in immature animals by administration of kainic acid (7 mg/kg intraperitoneal), whereas adults rats received saline. Animals were killed 72 hours or 35 days after treatment, and their brains were used for in vitro autoradiography experiments to determine BDZ binding. RESULTS In basal conditions and compared with the adult group, immature animals presented reduced BDZ binding in the entorhinal cortex, substantia nigra pars reticulata, and periaqueductal gray. Seventy-two hours after kainic acid-induced status epilepticus, immature rats showed significantly increased BDZ in the frontal (48%), cingulate (39%), sensorimotor (39%), piriform (57%), and entorhinal (59%) cortices, the medial (84%) and basolateral (27%) amygdaloid nuclei, the dentate gyrus (51%), and the substantia nigra pars reticulata (43%). Thirty-five days after status epilepticus, immature rats displayed decreased BDZ binding in the entorhinal cortex (48%), dentate gyrus (36%), and fields CA1, CA2, and CA3 of Ammon's horn (30%). CONCLUSIONS The present study demonstrates that status epilepticus and temporal lobe epilepsy produce a characteristic pattern of BDZ binding changes in the immature rat brain that differs from the one previously seen in adults.
Collapse
Affiliation(s)
- L Rocha
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México.
| | | | | | | | | |
Collapse
|
218
|
Zipfel GJ, Babcock DJ, Lee JM, Choi DW. Neuronal apoptosis after CNS injury: the roles of glutamate and calcium. J Neurotrauma 2000; 17:857-69. [PMID: 11063053 DOI: 10.1089/neu.2000.17.857] [Citation(s) in RCA: 201] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
While a role has been well established for excitotoxic necrosis in the pathogenesis of traumatic or ischemic damage to the CNS, accumulating evidence now suggests that apoptosis may also be a prominent contributor. In this review we focus on the role of glutamate and attendant intracellular calcium influx in triggering or modifying excitotoxic necrosis and apoptosis, raising the possibility that calcium influx may affect these two death pathways in opposite directions. Incorporating consideration of both pathways will probably be needed to develop the most effective neuroprotective treatments for CNS injury.
Collapse
Affiliation(s)
- G J Zipfel
- Center for the Study of Nervous System Injury and Department of Neurology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
219
|
Lee YK, Lee SR, Kim CY. Melatonin attenuates the changes in polyamine levels induced by systemic kainate administration in rat brains. J Neurol Sci 2000; 178:124-31. [PMID: 11018704 DOI: 10.1016/s0022-510x(00)00393-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Systemically administered kainate has been demonstrated to induce neuronal damage and changes of the levels of biochemical substances related to neurotoxicity. Polyamines are thought to be important in the generation of edema and neuronal cell loss associated with various type of excitotoxicity. Melatonin exerts potent free radical scavenging, antioxidant, and neuroprotective properties. This study was designed to estimate the effect of exogenous melatonin administration on the changes of polyamine levels in rat brains after systemic administration of kainate. Kainate [10 mg/kg, intraperitoneally (i.p.)] was injected into the rats to produce excitotoxicity. Melatonin (15 mg/kg, i.p.) was administered 1 h before, immediately after, and 1 h after kainate treatment. We examined the polyamine [putrescine (PU), spermidine (SD) and spermine (SM)] levels in the cerebral cortex and hippocampus and neuronal density in the hippocampal CA1 and CA3 subsectors in brain sections. PU levels were increased 8 and 24 h after kainate treatment and the administration of melatonin attenuated these changes. Only minor changes were noted in the levels of the polyamine SD and SM after the kainate treatment. In histology, neuronal injuries in the hippocampal CA1 and CA3 subsectors were examined 3 days after kainate treatment and melatonin reduced the kainate-induced neuronal injuries. Our results show that melatonin inhibits the polyamine responses in the cerebral cortex and hippocampus following kainate-induced excitotoxicity and PU may be responsible for the protective effect of melatonin against kainate-induced excitotoxicity.
Collapse
Affiliation(s)
- Y K Lee
- Department of Pharmacology, School of Medicine, Kyungpook National University, 2-101 Dongin dong, 700-422, Taegu, South Korea
| | | | | |
Collapse
|
220
|
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. [PMID: 10858610 DOI: 10.1016/s0306-4522(00)00085-3] [Citation(s) in RCA: 161] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Prolonged seizures (status epilepticus) induced by kainic acid activate programmed cell death mechanisms, and it is believed that kainic acid-induced status epilepticus induces neuronal apoptosis. In order to test this hypothesis, adult rats were subjected to 3-h kainic acid-induced seizures, with 24- or 72-h recovery periods. Neuronal death was assessed by light microscopy with the Hematoxylin and Eosin stain and with in situ terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL stain), by electron microscopy, and by agarose gel electrophoresis of DNA extracted from five vulnerable brain regions. Spontaneous and MK-801-induced apoptotic neurons from retrosplenial cortex of neonatal rats, evaluated by light and electron microscopy, were used as positive controls for apoptosis. Surprisingly, the large chromatin clumps of apoptotic neurons were TUNEL negative, whereas the cytoplasm showed light-to-moderate TUNEL staining, consistent with a lack of identifiable nuclear membranes ultrastructurally, and with intermingling of nuclear and cytoplasmic contents. Ultrastructurally, the acidophilic neurons produced by kainic acid-induced status epilepticus, identified with Hematoxylin and Eosin stain, were dark, shrunken and necrotic, with pyknotic nuclei containing small, dispersed chromatin clumps, and with cytoplasmic vacuoles, some of which were swollen, disrupted mitochondria. No apoptotic cells were seen. Acidophilic neurons were found in up to 20 of 23 brain regions examined and comprised 10-25% of the total number of neurons examined. A subset of these neurons (<10% of the total number of neurons in five of 23 regions) had TUNEL-positive nuclei 72h but not 24h after status epilepticus. Internucleosomal DNA cleavage (DNA "laddering") occurred in the four most damaged brain regions examined by electron microscopy 24h after SE and the three most damaged regions 72h after status epilepticus. Our results demonstrate that kainic acid-induced status epilepticus produces neuronal necrosis and not apoptosis in adult rats. The necrotic neurons show nuclear pyknosis, chromatin condensation and DNA laddering. Programmed cell death mechanisms activated by kainic acid-induced status epilepticus occur in neurons which become necrotic and could contribute to necrotic, as well as apoptotic, neuronal death.
Collapse
Affiliation(s)
- D G Fujikawa
- Experimental Neurology Laboratory, VA Greater Los Angeles Healthcare System, Sepulveda Ambulatory Care Center and Nursing Home Care Unit, Sepulveda CA 91343, USA.
| | | | | |
Collapse
|
221
|
Fujikawa DG, Itabashi HH, Wu A, Shinmei SS. Status epilepticus-induced neuronal loss in humans without systemic complications or epilepsy. Epilepsia 2000; 41:981-91. [PMID: 10961625 DOI: 10.1111/j.1528-1157.2000.tb00283.x] [Citation(s) in RCA: 126] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE To determine the regional distribution of neuronal damage caused strictly by status epilepticus (SE) without systemic complications, underlying brain pathology, or a history of preexisting epilepsy. METHODS The medical records and electroencephalograms (EEGs) of three deceased patients who developed SE in the hospital were reviewed. Their brains were formalin-fixed, and 17 brain regions were selected, embedded in paraffin, and sectioned. Alternate sections were stained with either hematoxylin and eosin and cresyl violet to determine the extent of neuronal loss and gliosis or glial fibrillary astrocytic protein to confirm the extent of astrocytic proliferation. RESULTS The three patients died 11 to 27 days after the onset of focal motor SE; none had hypotension, hypoxemia, hypoglycemia, or significant hyperthermia. Two patients had no prior seizures and no underlying brain pathology. The third patient, who had leptomeningeal carcinomatosis, had one seizure 2 months before the onset of SE. The duration of SE was 8.8 hours to 3 days. EEGs showed unilateral temporal lobe sharp-wave discharges in one patient and independent temporal lobe sharp-wave discharges bilaterally in the other two patients. In addition to widespread neuronal loss and reactive gliosis in the hippocampus, amygdala, dorsomedial thalamic nucleus, and Purkinje cell layer of the cerebellum, we report for the first time periamygdaloid (piriform) and entorhinal cortical damage occurring acutely after SE in humans. CONCLUSIONS In the absence of systemic complications or preexisting epilepsy, SE produces neuronal loss in a distribution similar to that from domoic acid-induced SE in humans and from kainic acid- and pilocarpine-induced SE in rats.
Collapse
Affiliation(s)
- D G Fujikawa
- Neurology Department, VA Greater Los Angeles Healthcare System, Sepulveda Ambulatory Care Center, California 91343, USA.
| | | | | | | |
Collapse
|
222
|
Baran H, Kepplinger B, Hörtnagl H. Clonidine modulates BAY K 8644-induced rat behavior and neurotransmitter changes in the brain. Eur J Pharmacol 2000; 401:31-7. [PMID: 10915834 DOI: 10.1016/s0014-2999(00)00404-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BAY K 8644 (methyl-1,4-dihydro-2, 6-dimethyl-3-nitro-4[2-trifluoromethyl-phenyl]-pyridine-5-carboxylate), an activator of dihydropyridine-sensitive Ca(2+) channels, injected in rats [2 mg/kg intraperitoneally (i.p.)], induces behavioral changes including ataxia, increased sensitivity to auditory stimulation, stiff tail, arched back, limb tonus and clonus, and rolling over. Neurochemical changes in the brain 45 min after application of 2 mg/kg were characterized by a significant decrease of noradrenaline in the amygdala (-27.8%, P<0.02) and piriform cortex (-16.3%, P<0.02). No significant changes of catecholamines were found in the hippocampal subregions CA1, CA3 and dentate gyrus or in the septum as compared to controls. The dopamine metabolites, 3,4-dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA), in the amygdala were elevated by 60% (P<0.02) and 66.7% (P<0.02), respectively. In the septum, a 52.6% (P<0.02) increase of HVA was observed. Analysis of amino acids revealed a marked increase of gamma-aminobutyric acid (GABA) content (+50.4%, P<0.001) in the septum. Pretreatment of the rats with the alpha(2)-adrenoceptor agonist, clonidine (0.1 mg/kg i.p.), 30 min before BAY K 8644 (2 mg/kg i.p.) injection completely abolished the behavioral and neurochemical changes. The data suggest that the Ca(2+)-dependent neurotransmitter release provoked by BAY K 8644 can be modulated by stimulation of presynaptic alpha(2)-adrenoceptors. The effect of clonidine on the GABAergic system may represent an important mechanism involved in the prevention of BAY K 8644-induced behavior.
Collapse
Affiliation(s)
- H Baran
- Institute of Pharmacology and Toxicology, Veterinary University Vienna, A-1210, Vienna, Austria.
| | | | | |
Collapse
|
223
|
Grootendorst J, Mulder M, Haasdijk E, de Kloet ER, Jaarsma D. Presence of apolipoprotein E immunoreactivity in degenerating neurones of mice is dependent on the severity of kainic acid-induced lesion. Brain Res 2000; 868:165-75. [PMID: 10854569 DOI: 10.1016/s0006-8993(00)02250-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Apolipoprotein E (apoE) is a major apolipoprotein in the central nervous system (CNS) that may play a role in various CNS disorders. ApoE is primarily localised in astrocytes, but neuronal apoE mRNA expression has been demonstrated in normal and diseased human brain, as well as in ischaemic rat brain. To obtain further insight into the role of apoE in neuronal degeneration in the CNS and conditions of neuronal apoE localisation, we have investigated in mice the distribution of apoE following neuronal injury induced by kainic acid (n=35, 25 or 35 mg kainic acid/kg BW). Consecutive series of brain sections were immunostained for apoE and markers for astroglia (GFAP) and microglia/macrophage cells (CR3). Degenerating neurones were identified with a silver-degeneration staining technique. The intensity and cellular distribution of apoE-immunoreactivity (apoE-ir) was dependent on the severity of neuronal injury. Mice that developed mild neuronal degeneration, restricted to a subset of neurones in the hippocampus, showed increased apoE-ir in astrocytes concomitant with increased GFAP-ir and mild microgliosis. In these mice, no neuronal apoE-ir was detected. In contrast, mice developing severe neuronal injury in the hippocampus - frequently also showing degeneration in other brain regions including cortex, thalamus, striatum and amygdala - showed intense apoE-ir in degenerating neurones. Surrounding the lesion, apoE-ir was increased in neuropil recurrently whereas GFAP-ir astrocytes disappeared. Thus, in mice apoE accumulates in degenerating neurones in conditions of severe neuronal injury putatively in association with disruption of the glial network.
Collapse
Affiliation(s)
- J Grootendorst
- Leiden/Amsterdam Center for Drug Research, Division of Medical Pharmacology, Leiden University, P.O. Box 9503, 2300 RA, Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
224
|
DePrato Primeaux S, Holmes PV, Martin RJ, Dean RG, Edwards GL. Experimentally induced attenuation of neuropeptide-Y gene expression in transgenic mice increases mortality rate following seizures. Neurosci Lett 2000; 287:61-4. [PMID: 10841991 DOI: 10.1016/s0304-3940(00)01137-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Previous experiments have reported increased seizure susceptibility in transgenic mice lacking normal neuropeptide-Y (NPY) gene expression (i.e. NPY 'knock-out' mice). A critical issue inherent in such experiments concerns the confounding of developmental influences of NPY and its neurotransmitter functions in the mature organism. The present experiments directly addressed this issue by studying seizure susceptibility in transgenic mice possessing an inducible antisense transcript that can be experimentally manipulated to attenuate NPY synthesis. NPY-deficient and control mice were injected with kainic acid (40 mg/kg, i.p.) and several seizure-related behaviors were measured. Consistent with previously reported effects in NPY knock-out mice, significantly more NPY-deficient mice died within 24 h than control mice. In situ hybridization analyses confirmed a decrease in prepro-NPY gene expression in transgenic mice. The experiments support the hypothesis that the control of neural excitability is a prominent function of NPY.
Collapse
Affiliation(s)
- S DePrato Primeaux
- Department of Psychology, The University of Georgia, Athens, GA 30602, USA
| | | | | | | | | |
Collapse
|
225
|
Schwarcz R, Eid T, Du F. Neurons in layer III of the entorhinal cortex. A role in epileptogenesis and epilepsy? Ann N Y Acad Sci 2000; 911:328-42. [PMID: 10911883 DOI: 10.1111/j.1749-6632.2000.tb06735.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A preferential lesion of neurons in layer III of the entorhinal cortex (EC) is often observed in patients suffering from temporal lobe epilepsy and in several animal models of the disease. This lesion is duplicated in rats by a focal, intra-entorhinal injection of the "indirect" excitotoxin aminooxyacetic acid (AOAA), providing a model that can be used to study the mechanisms underlying seizure-induced cell death and epilepsy. Doomed neurons in the EC and in several associated limbic structures show pathological changes within hours after the AOAA injection, but GABAergic neurons in layer III of the EC are quite resistant. This pattern of neuron loss eventually results in hippocampal and entorhinal hyperexcitability. Notably, the seizure-induced death of layer III neurons in the EC can be attenuated by eliminating the prominent excitatory input from the presubiculum. Taken together, these results suggest opportunities to target parahippocampal structures for the treatment of temporal lobe epilepsy.
Collapse
Affiliation(s)
- R Schwarcz
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore 21228, USA.
| | | | | |
Collapse
|
226
|
Benes FM, Berretta S. Amygdalo-entorhinal inputs to the hippocampal formation in relation to schizophrenia. Ann N Y Acad Sci 2000; 911:293-304. [PMID: 10911881 DOI: 10.1111/j.1749-6632.2000.tb06733.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This chapter reviews recent postmortem studies of schizophrenic brain and discusses the potential role of the amygdala in the induction of hippocampal abnormalities in this disorder. Based on available evidence, sectors CA4, CA3, and CA2, but not CA1, show preferential changes in schizophrenic subjects, although the most pronounced changes have been found in CA3 and CA2. It seems likely that the amygdala would contribute in some way to the induction of abnormalities along the trisynaptic pathway via its direct input to sectors CA3 and CA2, as well as an indirect one that involves the entorhinal cortex and its perforant path projection to the area dentata. The postmortem findings reported to date have been integrated into a working model in which decreases of inhibitory GABAergic modulation are invoked to explain the observation from a recent PET scan study (Heckers et al., 1999) that baseline metabolic activity in the hippocampus of schizophrenics is increased. In addition, however, the apparent inability of schizophrenics to increase metabolic activity in the hippocampus when challenged with a memory retrieval task may reflect a disturbance of disinhibitory modulation postulated herein to occur in sector CA3, a key relay point along the trisynaptic pathway. Overall, it seems plausible that an increase of excitatory activity entering the hippocampus from the basolateral complex via both direct and indirect pathways may make a significant contribution to the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- F M Benes
- Laboratory for Structural Neuroscience, McLean Hospital, Belmont, Massachusetts, USA.
| | | |
Collapse
|
227
|
Hopkins KJ, Wang G, Schmued LC. Temporal progression of kainic acid induced neuronal and myelin degeneration in the rat forebrain. Brain Res 2000; 864:69-80. [PMID: 10793188 DOI: 10.1016/s0006-8993(00)02137-5] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The excitatory amino acid glutamate has been implicated in the neurodegeneration associated with several different central nervous system diseases. Treatment with kainic acid (KA), a glutamate analog known to activate the AMPA/KA subtype of glutamate receptor, has been widely used as a model of epilepsy. Long term temporal studies of its neuropathological effects, however, are lacking. In this study, two techniques were used to directly visualize and characterize the neuropathology that occurred over a 2-month period following KA-induced status epilepticus in adult female Sprague-Dawley rats. Post-injection survival was 2, 4, 8 h, 2 days, 2 weeks, or 2 months. Labeling with Fluoro-Jade B (FJB), a fluorescent green dye that labels the cell body, dendrites, axons and axon terminals of degenerating neurons, was observed within the cortex, hippocampus, thalamus, basal ganglia, and amygdala by 4 h post-treatment. The highest level of labeling was seen in the piriform cortex, hippocampus, and thalamus. Myelin changes in the rat forebrain following KA treatment were also examined using the myelin-specific Black-Gold (BG) stain. Varicose myelinated fibers were observed in the same regions as FJB positive neurons, although these changes were evident by the 2-h survival time-point. Both stains showed a temporal progression of brain damage throughout the affected areas. By 2 months post-treatment, few degenerating neurons could be detected and abnormal myelin was absent in most regions. As myelin changes can be seen prior to neuronal degeneration, and oligodendrocytes express functional AMPA/kainate-type glutamate receptors, the neurodegeneration and myelin pathologies may occur as independent events. Thus, researchers should consider the temporal and multiple effects of kainic acid to optimize conditions for their endpoint of interest when designing experiments.
Collapse
Affiliation(s)
- K J Hopkins
- Division of Neurotoxicology, National Center for Toxicological Research, Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, USA.
| | | | | |
Collapse
|
228
|
Ransohoff RM, Bacon KB. Chemokine receptor antagonism as a new therapy for multiple sclerosis. Expert Opin Investig Drugs 2000; 9:1079-97. [PMID: 11060729 DOI: 10.1517/13543784.9.5.1079] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
New information about the role of tissue inflammation in the pathogenesis of multiple sclerosis (MS) has driven a search for effective and specific therapeutics that address leukocyte trafficking. These developments in understanding MS are complemented by advances in clarifying the molecular mechanisms of leukocyte extravasation and providing the knowledge base needed to modulate tissue inflammation. Of particular interest are the chemokines and their receptors. Chemokines constitute a large family of chemoattractant peptides that regulate the vast spectrum of leukocyte migration events. This review discusses MS and proposes that identifying the chemokines and receptors involved in the inflammation associated with this disorder may lead to therapeutic intervention.
Collapse
Affiliation(s)
- R M Ransohoff
- The Lerner Research Institute, Cleveland Clinic Foundation, NC30, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | | |
Collapse
|
229
|
Hashizume K, Kunimoto M, Maeda T, Tanaka T. Antiepileptic effect of nefiracetam on kainic acid-induced limbic seizure in rats. Epilepsy Res 2000; 39:221-8. [PMID: 10771248 DOI: 10.1016/s0920-1211(00)00099-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Nefiracetam is being studied as a novel cognition-enhancing agent; however, it has been suggested from studying its chemical structure that it has a potential anticonvulsive effect. We examined the antiepileptic effect of nefiracetam on kainic acid (KA)-induced seizures. KA was infused into the left basolateral amygdaloid nucleus and focal limbic seizures were induced in 43 male Wistar rats. During status epilepticus, 10, 50, 100 or 200 mg/kg of nefiracetam was intravenously injected. Nefiracetam inhibited KA-induced limbic seizures at doses over 100 mg/kg while it had a sedative effect on the animals. In (14C) deoxyglucose autoradiographic studies, the propagation of seizure-induced hypermetabolic areas was also suppressed dose-dependently. From the results, it was indicated that nefiracetam has an antiepileptic effect and that its application may suppress seizure propagation. Further study is required, whether this agent is available as a novel anticonvulsant.
Collapse
Affiliation(s)
- K Hashizume
- Department of Neurosurgery, Asahikawa Medical College, 4-5 Nishikagura, Asahikawa, Japan.
| | | | | | | |
Collapse
|
230
|
Ekonomou A, Sperk G, Kostopoulos G, Angelatou F. Reduction of A1 adenosine receptors in rat hippocampus after kainic acid-induced limbic seizures. Neurosci Lett 2000; 284:49-52. [PMID: 10771159 DOI: 10.1016/s0304-3940(00)00954-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In a temporal lobe epilepsy (TLE) model induced by kainic acid (KA), we examined the effect of limbic seizures on A1 adenosine receptor distribution in hippocampus and cortex. By using quantitative autoradiography, we determined a progressive decrease in A1 receptor density in CA1 and CA3 regions of hippocampus, which coincided in time with the degenerating process of hippocampal pyramidal cells. This result indicates that a great amount of A1 receptors are located postsynaptically on pyramidal cell dendrites. No difference in A1 receptor density was observed in the inner compared to the outer molecular layer of dentate gyrus, or in the infrapyramidal band compared to the outer layer of stratum oriens of CA3. This could indicate that the newly sprouted mossy fiber glutamatergic terminals do not contain A1 receptors, thus lacking a restrain in the release of glutamate.
Collapse
Affiliation(s)
- A Ekonomou
- Department of Physiology, School of Medicine, University of Patras, 265 00, Patras, Greece
| | | | | | | |
Collapse
|
231
|
Eriksson C, Tehranian R, Iverfeldt K, Winblad B, Schultzberg M. Increased expression of mRNA encoding interleukin-1beta and caspase-1, and the secreted isoform of interleukin-1 receptor antagonist in the rat brain following systemic kainic acid administration. J Neurosci Res 2000; 60:266-79. [PMID: 10740232 DOI: 10.1002/(sici)1097-4547(20000415)60:2<266::aid-jnr16>3.0.co;2-p] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Kainic acid, an analogue of glutamate, injected systemically to rats evokes seizures that are accompanied by nerve cell damage primarily in the limbic system. In the present study, we have analyzed the temporal profile of the expression of the cytokines interleukin-1beta (IL-1beta) and IL-1 receptor antagonist (IL-1ra), and the related IL-1beta-converting enzyme (ICE/caspase-1), in different regions of the rat brain in response to peripheral kainic acid administration (10 mg/kg, i.p.). In situ hybridization histochemistry experiments revealed that IL-1beta mRNA-expressing cells, morphologically identified as microglial cells, were mainly localized to regions showing pronounced neuronal degeneration; hippocampus, thalamus, amygdala, and certain cortical regions. The strongest expression of IL-1beta mRNA was observed after 12 hr in these regions. A weak induction of the IL-1beta mRNA expression was observed already at 2 hr. Similar results were obtained by RT-PCR analysis, showing a significantly increased expression of IL-1beta mRNA in the hippocampus and amygdala after 12 hr. In addition, RT-PCR analysis revealed that IL-1ra mRNA, and specifically mRNA encoding the secreted isoform of IL-1ra (sIL-1ra), was strongly induced in the hippocampus and amygdala at 12 and 24 hr post-injection. RT-PCR analysis of mRNA encoding caspase-1 showed a significantly increased expression in the amygdala after 12 hr. In conclusion, in response to systemic kainic acid injection IL-1beta mRNA is rapidly induced and followed by induction of IL-1ra mRNA and caspase-1 mRNA, supporting a role of the IL-1 system in the inflammatory response during excitotoxic damage.
Collapse
Affiliation(s)
- C Eriksson
- Division of Geriatric Medicine, NEUROTEC, Karolinska Institute, Huddinge Hospital, Novum, Huddinge, Sweden.
| | | | | | | | | |
Collapse
|
232
|
Ding M, Haglid KG, Hamberger A. Quantitative immunochemistry on neuronal loss, reactive gliosis and BBB damage in cortex/striatum and hippocampus/amygdala after systemic kainic acid administration. Neurochem Int 2000; 36:313-8. [PMID: 10732998 DOI: 10.1016/s0197-0186(99)00139-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cell specific markers were quantified in the hippocampus, the amygdala/pyriform cortex, the frontal cerebral cortex and the striatum of the rat brain after systemic administration of kainic acid. Neuron specific enolase (NSE) reflects loss of neurons, glial fibrillary acidic protein (GFAP) reflects reactive gliosis, and brain levels of serum proteins measures blood-brain-barrier permeability. While the concentration of NSE remained unaffected in the frontal cerebral cortex and the striatum, their GFAP content increased during the first three days. In the hippocampus and amygdala, NSE levels decreased significantly. GFAP levels in the hippocampus were unaffected after one day and decreased in the amygdala/pyriform cortex. After that, GFAP increased strikingly until day 9 or, in the case of amygdala/pyriform cortex, even longer. This biphasic time course for GFAP was accompanied by a decrease of S-100 during days 1-9 followed by a significant increase at day 27 above the initial level. The regional differences in GFAP and S-100 could result from the degree of neuronal degeneration, the astrocytic receptor set-up and/or effects on the blood-brain barrier.
Collapse
Affiliation(s)
- M Ding
- Department of Anatomy and Cell Biology, University of Göteborg, Sweden.
| | | | | |
Collapse
|
233
|
Abstract
The neurotoxic actions of kainic acid can be partly suppressed by antagonists acting at N-methyl-D-aspartate (NMDA) receptors. The present study examined the possible role of endogenous components of the kynurenine pathway to this phenomenon. Administration of kainate (2 nmols) into the hippocampus of anaesthetized rats produced damage in the CA1 and CA3 regions. The involvement of NMDA receptors was confirmed by the ability of dizocilpine (1 mg kg(-1)) to reduce cell loss in the CA1 region from 92 to 42%. The co-administration of m-nitrobenzoylalanine (20 nmols into the hippocampus), an inhibitor of kynurenine hydroxylase and kynureninase, together with a systemic injection of the compound (100 mg kg(-1), i.p.), afforded some protection against kainate, reducing cell loss from 91 to 48%. Protection was not exerted against damage by quinolinic acid or NMDA, excluding a direct interaction between m-nitrobenzoylalanine and NMDA receptors. The protective effect of m-nitrobenzoylalanine was not prevented by glycine, which would be expected to reverse protection caused by an elevation in the levels of endogenous kynurenic acid, arguing against a major role for increased levels of kynurenic acid. The results indicate that inhibition of the kynurenine pathway offers protection against kainate-induced damage. One possible mechanism for the protection is that an increased production of quinolinic acid in the brain, possibly from glial cells and macrophages activated by the initial kainate insult, normally contributes to the local activation of NMDA receptors and thus to kainate-induced cerebral insults. This generation of endogenous quinolinic acid would be suppressed by m-nitrobenzoylalanine.
Collapse
Affiliation(s)
- W M Behan
- Department of Pathology, University of Glasgow, Glasgow G12 8QQ. Institute of Biomedical & Life Sciences, West Medical Building, University of Glasgow, Glasgow G12 8QQ
| | | |
Collapse
|
234
|
Kondratyev A, Gale K. Intracerebral injection of caspase-3 inhibitor prevents neuronal apoptosis after kainic acid-evoked status epilepticus. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2000; 75:216-24. [PMID: 10686342 DOI: 10.1016/s0169-328x(99)00292-2] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In the aftermath of prolonged continuous seizure activity (status epilepticus, SE), neuronal cell death occurs in the brain regions through which the seizure propagates. Recent studies have implicated apoptotic processes in this seizure-related injury. Because activation of caspase-3-like cysteine proteases plays a crucial role in mammalian neuronal apoptosis, we explored the possibility that activation of caspase-3 is involved in the neuronal apoptotic cell death that occurs in rat brain following SE induced by systemic kainic acid. Caspase-3 activity was determined immunocytochemically using CM1 antibodies specific for catalytically active subunit (p17) of the enzyme. We found an induction of caspase-3 activity in rhinal cortex and amygdala at 24 h after SE. To determine whether activation of caspase-3-like proteases is a necessary component of the injury process, we delivered a caspase-3 inhibitor, z-DEVD-fmk, into the lateral ventricle prior to, and following SE. z-DEVD-fmk treatment substantially attenuated apoptotic cell death after SE, both in hippocampus and rhinal cortex, as evaluated by analysis of internucleosomal DNA fragmentation and neuronal nuclear morphology. Our findings implicate caspase-3 cysteine protease in the neurodegenerative response to SE and suggest that this degeneration can be attenuated by inhibition of caspase-3-like enzyme activity.
Collapse
Affiliation(s)
- A Kondratyev
- Department of Pharmacology, Georgetown University Medical Center, Washington, DC, USA.
| | | |
Collapse
|
235
|
Venero JL, Revuelta M, Machado A, Cano J. Delayed apoptotic pyramidal cell death in CA4 and CA1 hippocampal subfields after a single intraseptal injection of kainate. Neuroscience 2000; 94:1071-81. [PMID: 10625049 DOI: 10.1016/s0306-4522(99)00226-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We have performed a detailed time-course analysis of cell death in the hippocampal formation, basal forebrain and amygdala following a single intraseptal injection of kainate in adult rats. Acetylcholinesterase histochemistry revealed a profound loss of staining in the medial septum but not in the diagonal band, and cholinergic fiber density was highly reduced in the hippocampus and amygdala at 10 days postinjection. Terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphatebiotin nick end labeling (TUNEL) histochemistry was performed for precise location of apoptotic cells. Both the medial septum and amygdala exhibited numerous TUNEL-positive nuclei after the intraseptal injection of kainate, while the lateral septum exhibited a lower but significant incidence in terms of apoptotic cells. In the medial septum, the presence of apoptotic cells was at a location displaying acetylcholinesterase staining. TUNEL histochemistry revealed a time-dependent sequential apoptotic cell death in hippocampal pyramidal cells. During the first two days postinjection, apoptosis in the hippocampus was only evident in the CA3 region. At five days postinjection, the entire CA4 region became apoptotic. At 10 days postinjection, the whole extent of the CA1 pyramidal cell layer exhibited numerous TUNEL-positive nuclei. The time-course of kainate-induced apoptosis in Ammons's horn correlated with the disappearance of hippocampal pyramidal neurons as detected by Nissl staining, which is suggestive of a prominent apoptotic death for these cells. The temporal delayed distant damage to CA4 and CA1 hippocampal subfields after a single intraseptal kainate injection is not seen in other models employing kainate and may be a valuable tool for exploring the cellular mechanisms leading to cell death in conditions of status epilepticus.
Collapse
Affiliation(s)
- J L Venero
- Departamento de Bioquímica, Bromatología y Toxicología, Facultad de Farmacia, Universidad de Sevilla, Spain
| | | | | | | |
Collapse
|
236
|
Kim H, Bing G, Jhoo W, Ko KH, Kim WK, Suh JH, Kim SJ, Kato K, Hong JS. Changes of hippocampal Cu/Zn-superoxide dismutase after kainate treatment in the rat. Brain Res 2000; 853:215-26. [PMID: 10640619 DOI: 10.1016/s0006-8993(99)02254-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
In order to evaluate the putative role of Cu,Zn-superoxide dismutase (SOD-1) in the antioxidant defense mechanism during the neurodegenerative process, we examined the level of mRNA, the specific activity and immunocytochemical distribution for SOD-1 in the rat hippocampus after systemic injection of kainic acid (KA). Hippocampal SOD-1 mRNA levels were significantly increased by the seizure intensity 3 and 7 days after KA. These enhanced mRNA levels for SOD-1 were consistent with the increased specific activities for SOD-1, suggesting that the superoxide radical generated in neurotoxic lesion, induced SOD-1 mRNA. The CA1 and CA3 neurons lost their SOD-1-like immunoreactivity, whereas SOD-1-positive glia-like cells mainly proliferated throughout the CA1 sector and had an intense immunoreactivity at 3 and 7 days after KA. This immunocytochemical distribution for SOD-1-positive non-neuronal elements was similar to that for glial fibrillary acidic protein (GFAP)-positive cells. Each immunoreactivity for SOD-1-positive non-neuronal cell or GFAP in the layers of CA1 and CA3 disappeared 3 and 7 days after a maximal stage 5 seizure. On the other hand, activated microglial cells as selectively marked with the lectin occurred in the areas affected by KA-induced lesion. Double-labeling immunocytochemical analysis demonstrated the co-localization of SOD-1-positive glia-like cells and reactive astrocytes as labeled by GFAP or S-100 protein immunoreactivity. This finding suggested that the mobilization of astroglial cells for the synthesis of SOD-1 protein is a response to the KA insult designed to decrease the neurotoxicity induced by oxygen-derived free radicals. Therefore, these alterations might reflect the regulatory role of SOD-1 against oxygen-derived free radical-induced neuronal degeneration after systemic KA administration.
Collapse
Affiliation(s)
- H Kim
- Section of Pharmacology, Department of Pharmacy, College of Pharmacy, Kangwon National University, Chunchon, South Korea.
| | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Schauwecker PE, Ramirez JJ, Steward O. Genetic dissection of the signals that induce synaptic reorganization. Exp Neurol 2000; 161:139-52. [PMID: 10683280 DOI: 10.1006/exnr.1999.7251] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Synaptic reorganization of mossy fibers following kainic acid (KA) administration has been reported to contribute to the formation of recurrent excitatory circuits, resulting in an epileptogenic state. It is unclear, however, whether KA-induced mossy fiber sprouting results from neuronal cell loss or the seizure activity that KA induces. We have recently demonstrated that certain strains of mice are resistant to excitotoxic cell death, yet exhibit seizure activity similar to what has been observed in rodents susceptible to KA. The present study takes advantage of these strain differences to explore the roles of seizure activity vs cell loss in triggering mossy fiber sprouting. In order to understand the relationships between gene induction, cell death, and the sprouting response, we assessed the regulation of two molecules associated with the sprouting response, c-fos and GAP-43, in mice resistant (C57BL/6) and susceptible (FVB/N) to KA-induced cell death. Following administration of KA, increases in c-fos immunoreactivity were observed in both strains, although prolonged induction of c-fos was present only in the hippocampal neurons of FVB/N mice. Mossy fiber sprouting following KA administration was also only observed in FVB/N mice, while induction of GAP-43, a marker associated with mossy fiber sprouting, was not observed in either strain. These results indicate that: (i) KA-induced seizure activity alone is insufficient to induce mossy fiber sprouting; (ii) mossy fiber sprouting may be due to the loss of hilar neurons following kainate administration; and (iii) induction of GAP-43 is not a necessary component of the sprouting response that occurs following KA in mice.
Collapse
Affiliation(s)
- P E Schauwecker
- Departments of Neuroscience, University of Virginia Health Sciences Center, Charlottesville, Virginia, 22908, USA
| | | | | |
Collapse
|
238
|
Won SJ, Ko HW, Kim EY, Park EC, Huh K, Jung NP, Choi I, Oh YK, Shin HC, Gwag BJ. Nuclear factor kappa B-mediated kainate neurotoxicity in the rat and hamster hippocampus. Neuroscience 1999; 94:83-91. [PMID: 10613499 DOI: 10.1016/s0306-4522(99)00196-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Administration of the excitotoxin kainate produces seizure activity and selective neuronal death in various brain areas. We examined the degeneration pattern of hippocampal neurons following systemic injections of kainate in the hamster and the rat. As reported, treatment with kainate resulted in severe neuronal loss in the hilus and CA3 in the rat. While the hilar neurons were also highly vulnerable to kainate in the hamster, neurons in the CA1 area, but not CA3, were highly sensitive to kainate. In both animals, immunoreactivity to anti-p50 nuclear factor kappa B antibody was increased in nuclei of the hilar neurons within 4 h following administration of kainate. Kainate treatment also increased the nuclear factor kappa B immunoreactivity in hamster CA1 neurons and rat CA3 neurons 24 h later. Neurons showing intense nuclear factor kappa B signal were stained with acid fuchsin. Kainate also increased DNA binding activity of p50 and p65 nuclear factor kappa B in the nuclear extract of the hippocampal formation as analysed by electrophoretic mobility shift assay in the hamster, suggesting that activation of nuclear factor kappa B may contribute to kainate-induced hippocampal degeneration. Administration of 100 nmol dizocilpine maleate 3 h prior to kainate attenuated kainate-induced activation of nuclear factor kappa B and neuronal death in CA1 in the hamster. The present study provides evidence that the differential vulnerability of neurons in the rat and the hamster hippocampus to kainate is partly mediated by mechanisms involving N-methyl-D-aspartate-dependent activation of nuclear factor kappa B.
Collapse
Affiliation(s)
- S J Won
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Kyungkido, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
239
|
Plamondon H, Blondeau N, Heurteaux C, Lazdunski M. Mutually protective actions of kainic acid epileptic preconditioning and sublethal global ischemia on hippocampal neuronal death: involvement of adenosine A1 receptors and K(ATP) channels. J Cereb Blood Flow Metab 1999; 19:1296-308. [PMID: 10598933 DOI: 10.1097/00004647-199912000-00002] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Preconditioning with sublethal ischemia attenuates the detrimental effects of subsequent prolonged ischemic insults. This research elucidates potential in vivo cross-tolerance between different neuronal death-generating treatments such as kainate administration, which induces seizures and global ischemia. This study also investigates the effects of a mild epileptic insult on neuronal death in rat hippocampus after a subsequent, lethal epileptic stress using kainic acid (KA) as a model of epilepsy. Three preconditioning groups were as follows: group 1 was injected with 5 mg/kg KA before a 6-minute global ischemia; group 2 received a 3-minute global ischemia before 7.5 mg/kg KA; and group 3 was injected with a 5-mg/kg dose of KA before a 7.5-mg/kg KA injection. The interval between treatments was 3 days. Neuronal degeneration, revealed by the silver impregnation method and analysis of cresyl violet staining, was markedly reduced in rats preconditioned with a sublethal ischemia or a 5-mg/kg KA treatment. Labeling with terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'triphosphate-biotin nick-end labeling and DNA laddering confirmed the component of DNA fragmentation in the death of ischemic and epileptic neurons and its reduction in all preconditioned animals. The current study supports the existence of bidirectional cross-tolerance between KA excitotoxicity and global ischemia and suggests the involvement of adenosine A1 receptors and sulfonylurea- and ATP-sensitive K+ channels in this protective phenomenon.
Collapse
Affiliation(s)
- H Plamondon
- University of Ottawa, School of Psychology, Ontario, Canada
| | | | | | | |
Collapse
|
240
|
Eisch AJ, Lammers CH, Yajima S, Mouradian MM, Nestler EJ. In vivo regulation of glial cell line-derived neurotrophic factor-inducible transcription factor by kainic acid. Neuroscience 1999; 94:629-36. [PMID: 10579223 DOI: 10.1016/s0306-4522(99)00302-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A putative transcription factor induced in vitro by glial cell line-derived neurotrophic factor (GDNF) and transforming growth factor-beta was recently cloned and characterized [Yajima S. et al. (1997) J. Neurosci. 17, 8657-8666]. The messenger RNA of this protein, termed murine GDNF-inducible transcription factor (mGIF, hereafter referred to as GIF), is localized within cortical and hippocampal regions of brain, suggesting that GIF might be regulated by perturbations of these brain regions. In an effort to learn more about the role of GIF in vivo, we examined GIF messenger RNA in the brains of rats treated with the glutamatergic agonist kainic acid. This treatment is known to induce seizures and alter the messenger RNA expression of several growth factors, including GDNF, in several brain regions. Rats were given intraperitoneal saline (1 ml/kg) or kainic acid (15 mg/kg) and were killed at various time-points for in situ hybridization of brain sections with a GIF messenger RNA riboprobe. In saline-treated rats, GIF messenger RNA was present at low levels in cerebral cortex, hippocampus and hippocampal remnants such as the taenia tecta. Kainic acid treatment induced robust increases in GIF messenger RNA in several brain regions, including cerebral cortex, hippocampus, caudate-putamen, nucleus accumbens, and several nuclei of the amygdala and hypothalamus. Most brain regions showed the greatest increase in GIF messenger RNA 4-6 h after kainic acid administration and a return towards normal levels at 48 h. The CA3 region of hippocampus, however, showed a more rapid increase in GIF messenger RNA that was also evident 48 h after kainic acid administration. These results demonstrate that GIF messenger RNA can be regulated in vivo, and that this novel factor warrants further study as a central mediator of GDNF and perhaps other neurotrophic factors.
Collapse
Affiliation(s)
- A J Eisch
- Department of Psychiatry, Yale University School of Medicine, Connecticut Mental Health Center, New Haven 06508, USA
| | | | | | | | | |
Collapse
|
241
|
Boschert U, Merlo-Pich E, Higgins G, Roses AD, Catsicas S. Apolipoprotein E expression by neurons surviving excitotoxic stress. Neurobiol Dis 1999; 6:508-14. [PMID: 10600406 DOI: 10.1006/nbdi.1999.0251] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the adult brain, apolipoprotein E (apoE) mRNA is thought to be expressed by nonneuronal cells. Yet, when a brain damage has occurred, the protein is found in neurons. We have studied apoE expression following systemic kainic acid (KA), injected in rats to induce hippocampal neurodegeneration. We describe two effects. First, a moderate increase of apoE levels in astrocytes. Second, and unexpected, a very strong increase of apoE mRNA levels in clusters of CA1 and CA3 pyramidal neurons. Neuronal identity of these cells is supported by a series of observations. First, apoE hybridization signals were found in cells with morphological characteristics of pyramidal neurons. Second, the cells were positive for the neuronal marker MAP2. Third, the cells were negative for the astrocytic marker GFAP and for the microglia marker OX42. Fourth, the same distribution pattern was found with probes hybridizing to c-fos, a transcription factor transiently expressed in neurons under stress. At 48 and 72 h following KA, most of the excitotoxic cell death had already occurred. Since no morphological signs of programmed cell death were observed in apoE-positive pyramidal neurons, we suggest that expression of apoE by neurons may be part of a rescue program to counteract neurodegeneration.
Collapse
Affiliation(s)
- U Boschert
- Geneva Biomedical Research Institute, 14 Chemin des Aulx, Geneva, 1228, Switzerland
| | | | | | | | | |
Collapse
|
242
|
Hafidi A, Lanjun G, Sanes DH. Age-dependent failure of axon regeneration in organotypic culture of gerbil auditory midbrain. ACTA ACUST UNITED AC 1999. [DOI: 10.1002/(sici)1097-4695(19991105)41:2<267::aid-neu9>3.0.co;2-d] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
243
|
Eriksson C, Van Dam AM, Lucassen PJ, Bol JG, Winblad B, Schultzberg M. Immunohistochemical localization of interleukin-1beta, interleukin-1 receptor antagonist and interleukin-1beta converting enzyme/caspase-1 in the rat brain after peripheral administration of kainic acid. Neuroscience 1999; 93:915-30. [PMID: 10473257 DOI: 10.1016/s0306-4522(99)00178-5] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The temporal and anatomical distribution of members of the interleukin-1 system in the rat brain following intraperitoneal kainic acid administration was studied in relation to neurodegeneration as detected with in situ end labelling. Kainic acid administration (10 mg/kg, i.p.) resulted in the induced expression of interleukin-1beta, interleukin- receptor antagonist and caspase-1p10 immunoreactivity in areas known to display neuronal and tissue damage upon excitotoxic lesions. The induction of these proteins was transient. Interleukin-1 immunoreactivity appeared at 5 h, and the interleukin-1 receptor antagonist-immunoreactive cells were first detected at 12 h, whereas the induction of caspase- 1p10 expression was first detected 24 h after kainic acid injection. Double labelling with the microglial marker Ox42 confirmed that both interleukin-1beta and interleukin-1 receptor antagonist were mainly localized in microglial cells. The regional distribution of in situ end-labelled neurons was similar to the distribution of cells expressing interleukin-1beta and interleukin-1 receptor antagonist, whereas the distribution of caspase-1 was more limited. The in situ end-labelled neurons, were, similarly to the interleukin-1beta-positive cells, first detected at 5 h, which is earlier than the induction of caspase-1. Our results show that the induction of IL-1beta and IL-1 receptor antagonist proteins after kainic acid are closely associated with the temporal as well as the anatomical distribution of in situ end-labelled neurons, whereas the induction of caspase-1 protein exhibited a delayed temporal profile and limited distribution. Since cytokine production occurs in activated microglial cells, the inflammatory component seems to be a strong mediator of this type of excitotoxic damage. The late onset of the caspase-1 expression would seem to indicate that this enzyme has no fundamental role in directly causing neuronal cell death induced by systemic kainic acid.
Collapse
Affiliation(s)
- C Eriksson
- Department of Clinical Neuroscience and Family Medicine, Karolinska Institute, Huddinge Hospital, Novum, Sweden
| | | | | | | | | | | |
Collapse
|
244
|
Nonaka M, Kohmura E, Yamashita T, Yamauchi A, Fujinaka T, Yoshimine T, Tohyama M, Hayakawa T. Kainic acid-induced seizure upregulates Na(+)/myo-inositol cotransporter mRNA in rat brain. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1999; 70:179-86. [PMID: 10407166 DOI: 10.1016/s0169-328x(99)00127-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
A major organic osmolyte, myo-inositol protects cells from perturbing effects of high intracellular concentrations of electrolytes. Myo-inositol is accumulated into cells through Na(+)/myo-inositol cotransporter (SMIT). In order to investigate the regulation of SMIT in generalized seizure, we employed Northern blot analysis and in situ hybridization to study the changes in SMIT mRNA expression in kainic acid-injected rats. Northern blot analysis demonstrated that SMIT mRNA began to increase in the brain 2 h after onset of seizure, and peaked at 12 h. In situ hybridization revealed rapid increase of SMIT mRNA (2 h of seizure) in the CA3 hippocampal pyramidal cells and in the dentate granular cells. Then, at 4-6 h SMIT mRNA expression was observed in the other limbic structure such as amygdala and piriform cortex. Finally, in neocortex and in CA1 pyramidal cells, SMIT mRNA was slowly increased and peaked at 12 h. Microautoradiogram demonstrated that cells expressed SMIT mRNA were mainly neurons. These results suggest that SMIT mRNA is upregulated by kainic acid-induced seizure primarily in structures involved in seizure activity.
Collapse
Affiliation(s)
- M Nonaka
- Department of Neurosurgery, Osaka University School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
245
|
López E, Pozas E, Rivera R, Ferrer I. Bcl-2, Bax and Bcl-x expression following kainic acid administration at convulsant doses in the rat. Neuroscience 1999; 91:1461-70. [PMID: 10391451 DOI: 10.1016/s0306-4522(98)00704-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Neuronal death was produced in the CA1 and CA3 areas of the hippocampus, amygdala, and piriform and entorhinal cortices after intraperitioneal administration of kainic acid at convulsant doses to adult rats. To assess the involvement of members of the Bcl-2 family in cell death or survival, immunohistochemistry, western and northern blotting to Bcl-2, Bcl-x and Bax, and in situ hybridization to Bax were examined at different time-points after kainic acid treatment. Members of the Bcl-2 family were expressed in the cytoplasm of pyramidal neurons in the hippocampus, and in a subset of neurons of the piriform and the entorhinal cortices, amygdala and neocortex in the normal adult brain. Dying neurons in the pyramidal cell layer of CA1 and CA3 areas, entorhinal and piriform cortices, and amygdala also expressed Bcl-2, Bax and Bcl-x following excitotoxicity, although many dying cells did not. In addition, a number of cells in the affected areas showed Bax immunoreactivity in their nuclei at 24-48 h following kainic acid administration, thus indicating Bax nuclear translocation in a subset of dying cells. Western blots disclosed no modifications in the intensity of the bands corresponding to Bcl-2, Bcl-x and Bax, between control and kainic acid-treated rats. No modifications in the intensity of the bcl-2 messenger RNA band on northern blots was observed in kainic acid-treated rats. However, a progressive increase in the intensity of the bax messenger RNA band was found in kainic acid-treated rats at 6 h, 12 h and 24 h following kainic acid administration. Interestingly, a slight increase in Bax immunoreactivity was observed in the cytoplasm of neurons of the dentate gyrus at 24-48 h, a feature which matches the increase of bax messenger RNA in the same area, as shown by in situ hybridization at 12-24 h following kainic acid injection. The present results suggest that cell death or survival does not correlate with modifications of Bcl-2, Bax and Bcl-x protein, and messenger RNA expression, but rather that kainic acid excitotoxicity is associated with Bax translocation to the nucleus in a subset of dying cells.
Collapse
Affiliation(s)
- E López
- Hospital Prínceps d'Espanya, Departament de Biologia Cellular i Anatomia Patològica, Facultat de Medecina, Universitat de Barcelona, Hospitalet de Llobregat, Spain
| | | | | | | |
Collapse
|
246
|
Faherty CJ, Xanthoudakis S, Smeyne RJ. Caspase-3-dependent neuronal death in the hippocampus following kainic acid treatment. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1999; 70:159-63. [PMID: 10381555 DOI: 10.1016/s0169-328x(99)00143-6] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
In this study, we examined the levels of activated caspase-3 in the kainic acid (KA) model of hippocampal degeneration in both sensitive (FVB/N) and resistant (129/SvEMS) strains of mice. At 30 h, 2 and 4 days following KA administration, animals were sacrificed and brains examined for pyknosis, TUNEL labeling, and activated caspase-3 immunoreactivity. Catalytically active caspase-3 was first detected 30 h following KA treatment in the sensitive, FVB/N strain. This was 18 h before the appearance of pyknosis or TUNEL labeling. The expression of activated caspase-3 continues up to 4 days post-injection. No activated caspase-3 immunoreactivity was detected in the resistant, 129/SvEMS strain, neither was there evidence of pyknosis or TUNEL staining. This suggests that activation of caspase-3 is a necessary component of KA-induced cell death.
Collapse
Affiliation(s)
- C J Faherty
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 332 North Lauderdale, Memphis, TN 38105, USA
| | | | | |
Collapse
|
247
|
Martí E, Blasi J, Gomez De Aranda I, Ribera R, Blanco R, Ferrer I. Selective early induction of synaptosomal-associated protein (molecular weight 25,000) following systemic administration of kainate at convulsant doses in the rat. Neuroscience 1999; 90:1421-32. [PMID: 10338309 DOI: 10.1016/s0306-4522(98)00522-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
SNAP-25 (synaptosomal-associated protein of mol. wt 25,000) is an essential component for neurotransmitter release, and its expression has been related to the plastic responses that follow CNS injury. In the present study, transient induction of SNAP-25 in selected brain areas is shown by immunohistochemistry at short times after a single intraperitoneal injection of kainate at convulsant doses. Six hours after kainate injection, SNAP-25 immunoreactivity was noticed in the perikarya of certain neurons of the perirhinal and lateral cortices, polymorphic layer of the dentate gyrus, CA3 pyramidal area of the hippocampus, and thalamus. In the same areas, a strong increase in SNAP-25 immunorectivity was detected at 12 and 24 h after kainate injection in cell bodies and fibers. Four days after kainate administration, the immunostaining pattern was similar to that observed in control animals. Intraperitoneal injection of cycloheximide blocked the expression of SNAP-25, thus suggesting de novo SNAP-25 protein synthesis following kainate administration. Kainate-dependent induction of SNAP-25a messenger RNA synthesis was observed by in situ hybridization in the mentioned brain areas. Heat shock protein of mol. wt 72,000 (HSP70/72) is a chaperone whose expression is induced early under stress conditions. Its expression and distribution were compared to that of SNAP-25 after the excitotoxic insult. Brain areas overexpressing SNAP-25 and HSP70/72 overlapped. In addition, partial co-localization of both antigens was observed by double-labeling immunohistochemistry. These results provide evidence of an involvement of SNAP-25 in the reactive response that follows kainate administration, and support the role of this protein in the plastic events that take place after kainate excitotoxicity.
Collapse
Affiliation(s)
- E Martí
- Departament de Biologia Cel.lular i Anatomia Patològica, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain
| | | | | | | | | | | |
Collapse
|
248
|
Abstract
Kainic acid can induce a continuum of non-convulsive seizures characterised by epileptic automatisms and convulsive motor seizures depending on the dose. There are scarce data on the behavioural effects of low doses of kainate inducing only non-convulsive seizures. Therefore, we studied spontaneous behaviour of adult male rats using a method of positive habituation based on a detailed analysis of patterns and attention of animals to a stimulus object. Twenty-three animals were individually tested in the experimental arena on two consecutive days. Comparing the data from the first two exposures, a conspicuous habituation in all animals was observed. On experimental day 3, 12 rats received kainate (6 mg/kg intraperitoneally) and the remaining 11 animals received a physiological saline. After 1 h, animals were put into the arena with an object localised in the centre. It was found that both kainate and saline treated animals exhibited a significant increase in the total number of central area visits, and both the total and mean time spent in the vicinity of the object. However, the mean time spent was significantly shorter in kainate treated rats. Furthermore. kainate rats exhibited a significant decrease in rearing as compared with the controls. In addition, an epileptic automatism (wet dog shakes) was observed in seven out of 12 animals given kainate. The comparison of transition matrices between consecutive behavioural categories showed significant differences between the kainate and control groups. Our results demonstrate that a non-convulsive dose of kainate induced changes in the structure of spontaneous behaviour and impaired the processes related to maintenance of attention.
Collapse
Affiliation(s)
- A Mikulecká
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague.
| | | | | |
Collapse
|
249
|
Becker AJ, Gillardon F, Blümcke I, Langendörfer D, Beck H, Wiestler OD. Differential regulation of apoptosis-related genes in resistant and vulnerable subfields of the rat epileptic hippocampus. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1999; 67:172-6. [PMID: 10101244 DOI: 10.1016/s0169-328x(99)00060-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Animals exposed to kainic acid (KA) induced status epilepticus display a striking pattern of selective neuronal vulnerability in the hippocampus. Neurons in the hilus/CA3 and CA1 subfields appear particularly sensitive whereas dentate gyrus (DG) granule cells are resistant. The molecular basis for this differential susceptibility remains largely unknown. Recently, an involvement of nitric oxide, c-Jun amino-terminal kinases (JNK) and interleukin-1 beta converting enzyme (ICE)-related proteases has been proposed in KA induced neuronal cell death. In the present study, we have determined the regional expression of transcripts for two modulating genes operating in these pathways, i.e., the endogenous protein inhibitor of neuronal nitric oxide synthase (PIN), and a cytoplasmic inhibitor of the JNK signal transduction pathway, designated JNK interacting protein-1 (JIP-1) and of the gene for the apoptosis-executing protease Caspase-3 in KA-treated animals. The expression of PIN and JIP-1 was found significantly upregulated in granule cells of the resistant DG. In contrast, an induction of the ICE-related protease Caspase-3 was observed in vulnerable hippocampal regions, i.e. CA1, CA3 and hilus. These results point towards PIN and JIP-1 as antiapoptotic factors contributing to selective resistance of granule cells, whereas Caspase-3 may be involved in cell death of hippocampal CA1, CA3 and hilar neurons in the kainate epilepsy model.
Collapse
Affiliation(s)
- A J Becker
- Department of Neuropathology, University of Bonn Medical Center, Sigmund-Freud-Str. 25, 53105, Bonn, Germany
| | | | | | | | | | | |
Collapse
|
250
|
Vreugdenhil E, Datson N, Engels B, de Jong J, van Koningsbruggen S, Schaaf M, de Kloet ER. Kainate-elicited seizures induce mRNA encoding a CaMK-related peptide: A putative modulator of kinase activity in rat hippocampus. ACTA ACUST UNITED AC 1999. [DOI: 10.1002/(sici)1097-4695(199904)39:1<41::aid-neu4>3.0.co;2-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|