Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Neuromolecular Med ; 16(4): 727-41, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25063076

ABSTRACT

The nonselective inhibitors of class I/II histone deacetylases (HDACs) including trichostatin A and the clinically used suberoylanilide hydroxamic acid (SAHA, vorinostat) are neuroprotective in several models of neuronal injury. Here, we report that in cultured cortical neurons from newborn rats and in the cerebral cortex of whole neonate rats, these HDAC inhibitors exacerbated cytotoxicity of the DNA double-strand break (DSB)-inducing anticancer drug etoposide by enhancing apoptosis. Similar neurotoxic interactions were also observed in neurons that were treated with other DNA damaging drugs including cisplatin and camptothecin. In addition, in rat neonates, SAHA increased cortical neuron apoptosis that was induced by a single injection of the NMDA receptor antagonist dizocilpine (MK801). In etoposide-treated neurons, the nonselective HDAC inhibition resulted in more DSBs. It also potentiated etoposide-induced accumulation and phosphorylation of the pro-apoptotic transcription factor p53. Moreover, nonselective HDAC inhibition exacerbated neuronal apoptosis that was induced by the overexpressed p53. Importantly, such effects cannot be fully explained by inhibition of HDAC1, which is known to play a role in DSB repair and regulation of p53. The specific HDAC1 inhibitor MS275 only moderately enhanced etoposide-induced neuronal death. Although in etoposide-treated neurons MS275 increased DSBs, it did not affect activation of p53. Our findings suggest that besides HDAC1, there are other class I/II HDACs that participate in neuronal DNA damage response attenuating neurotoxic consequences of genotoxic insults to the developing brain.


Subject(s)
DNA Breaks, Double-Stranded/drug effects , Histone Deacetylase Inhibitors/pharmacology , Animals , Animals, Newborn , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Benzamides/pharmacology , Camptothecin/toxicity , Cells, Cultured , Cerebral Cortex/cytology , Cisplatin/toxicity , Dizocilpine Maleate/toxicity , Etoposide/administration & dosage , Etoposide/toxicity , Genes, p53 , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/physiology , Hydroxamic Acids/pharmacology , Injections, Intraventricular , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , Point Mutation , Protein Processing, Post-Translational/drug effects , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/metabolism , Vorinostat
2.
Neuroscience ; 163(4): 985-90, 2009 Nov 10.
Article in English | MEDLINE | ID: mdl-19635528

ABSTRACT

Traumatic human spinal cord injury (SCI) causes devastating and long-term hardships. These are due to the irreparable primary mechanical injury and secondary injury cascade. In particular, oligodendrocyte cell death, white matter axon damage, spared axon demyelination, and the ensuing dysfunction in action potential conduction lead to the initial deficits and impair functional recovery. For these reasons, and that oligodendrocyte and axon survival may be related, various neuroprotective strategies after spinal cord injury are being investigated. We previously demonstrated that oligodendrocytes in the adult rat epicenter ventrolateral funiculus (VLF) express 3'-5'-cyclic adenosine monophosphate-dependent phosphodiesterase 4 (PDE4) subtypes and that their death was attenuated up to 3 days after contusive cervical SCI when rolipram, a specific inhibitor of PDE4, was administered. Here, we report that (1) there are more oligodendrocyte somata in the adult rat epicenter VLF, (2) descending and ascending axonal conductivity in the VLF improves, and that (3) there are fewer hindlimb footfall errors during grid-walking at 5 weeks after contusive cervical SCI when rolipram is delivered for 2 weeks. This is the first demonstration of improved descending and ascending long-tract axonal conductivity across a SCI with this pharmacological approach. Since descending long-tract axonal conductivity did not return to normal, further evaluations of the pharmacokinetics and therapeutic window of rolipram as well as optimal combinations are necessary before consideration for neuroprotection in humans with SCI.


Subject(s)
Cervical Vertebrae , Neuroprotective Agents/pharmacology , Oligodendroglia/drug effects , Recovery of Function/drug effects , Rolipram/pharmacology , Spinal Cord Injuries/drug therapy , Animals , Axons/drug effects , Axons/pathology , Cell Count , Female , Nerve Fibers, Myelinated/drug effects , Neural Pathways/drug effects , Neural Pathways/pathology , Oligodendroglia/pathology , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/pathology , Time Factors , Walking
3.
J Biol Chem ; 276(36): 34206-12, 2001 Sep 07.
Article in English | MEDLINE | ID: mdl-11432866

ABSTRACT

Arterial smooth muscle cell (SMC) proliferation contributes to a number of vascular pathologies. Prostaglandin E(2) (PGE(2)), produced by the endothelium and by SMCs themselves, acts as a potent SMC growth inhibitor. The growth-inhibitory effects of PGE(2) are mediated through activation of G-protein-coupled membrane receptors, activation of adenylyl cyclases (ACs), formation of cAMP, and subsequent inhibition of mitogenic signal transduction pathways in SMCs. Of the 10 different mammalian AC isoforms known today, seven isoforms (AC2-7 and AC9) are expressed in SMCs from various species. We show that, despite the presence of several different AC isoforms, the principal AC isoform activated by PGE(2) in human arterial SMCs is a calmodulin kinase II-inhibited AC with characteristics similar to those of AC3. AC3 is expressed in isolated human arterial SMCs and in intact aorta. We further show that arterial SMCs isolated from AC3-deficient mice are resistant to PGE(2)-induced growth inhibition. In summary, AC3 is the principal AC isoform activated by PGE(2) in arterial SMCs, and AC3 mediates the growth-inhibitory effects of PGE(2). Because AC3 activity is inhibited by intracellular calcium through calmodulin kinase II, AC3 may serve as an important integrator of growth-inhibitory signals that stimulate cAMP formation and growth factors that increase intracellular calcium.


Subject(s)
Adenylyl Cyclases/physiology , Arteries/enzymology , Dinoprostone/metabolism , Isoenzymes/physiology , Muscle, Smooth, Vascular/enzymology , Animals , Aorta/embryology , Aorta, Thoracic/metabolism , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/pharmacology , Cell Division , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Immunohistochemistry , Infant, Newborn , Mice , Models, Biological , Muscle, Smooth/cytology , Precipitin Tests , Protein Isoforms
4.
J Neurosci ; 21(13): 4657-67, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11425893

ABSTRACT

Bcl-2, an antiapoptotic protein, protects cells against many but not all forms of apoptosis. For example, Bcl-2 does not protect non-neuronal cells against taxol, a microtubule-stabilizing agent. The underlying mechanism for the ineffectiveness of Bcl-2 against taxol has been the subject of intense interest. Data from non-neuronal cells indicate that taxol-induced apoptosis requires activation of N-terminal c-Jun protein kinase (JNK) that phosphorylates and inactivates Bcl-2. This suggests the interesting possibility that the apoptotic activity of JNK may be caused by phosphorylation of Bcl-2 and inhibition of the antiapoptotic activity of Bcl-2. Here we report that taxol induces apoptosis in cortical neurons but by a mechanism significantly different from that in non-neuronal cells. In contrast to human embryonic kidney 293 cells, expression of wild-type Bcl-2 in cortical neurons protected against taxol-induced apoptosis, and taxol did not induce Bcl-2 phosphorylation. Furthermore, cortical neurons express high basal JNK activity, and taxol did not stimulate total JNK activity. However, taxol activated a subpool of JNK in the nucleus and stimulated c-Jun phosphorylation. JNK inhibition or expression of a dominant-negative c-Jun abrogated taxol-induced apoptosis in cortical neurons, suggesting a role for JNK and JNK-mediated transcription in taxol-stimulated apoptosis. Furthermore, taxol-induced apoptosis in cortical neurons required inhibition of phosphatidylinositol 3-kinase signaling. These data suggest that taxol induces apoptosis in neurons by a mechanism quite distinct from that of non-neuronal cell lines and emphasize the importance of elucidating apoptotic mechanisms specific for neurons in the CNS.


Subject(s)
Apoptosis , Cerebral Cortex/drug effects , Neurons/drug effects , Paclitaxel/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression , Humans , JNK Mitogen-Activated Protein Kinases , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Neurons/cytology , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transcription, Genetic , Transfection
5.
J Neurosci ; 21(2): 434-43, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11160424

ABSTRACT

Activation of the extracellular signal-regulated kinase 1 (ERK1) and ERK2 by neurotrophins, neuronal activity, or cAMP has been strongly implicated in differentiation, survival, and adaptive responses of neurons during development and in the adult brain. Recently, a new member of the mitogen-activated protein (MAP) kinase family, ERK5, was discovered. Like ERK1 and ERK2, ERK5 is expressed in neurons, and ERK5 stimulation by epidermal growth factor is blocked by the MAP kinase/ERK kinase 1 (MEK1) inhibitors PD98059 and U0126. This suggests the interesting possibility that some of the functions attributed to ERK1/2 may be mediated by ERK5. However, the regulatory properties of ERK5 in primary cultured neurons have not been reported. Here we examined the regulation of ERK5 signaling in primary cultured cortical neurons. Our data demonstrate that, similar to ERK1/2, ERK5 is activated by neurotrophins including brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and NT-4. BDNF stimulation of ERK5 required the activity of MEK5. Surprisingly, ERK5 was not stimulated by cAMP or neuronal activity induced by glutamate or membrane depolarization. In contrast to ERK1/2, ERK5 strongly activated the transcriptional activity of myocyte enhancer factor 2C (MEF2C) in pheochromocytoma 12 (PC12) cells and was required for neurotrophin stimulation of MEF2C transcription in both PC12 cells and cortical neurons. Furthermore, ERK1/2, but not ERK5, induced transcription from Elk1 and the cAMP/ Ca(2+) response element in PC12 cells. Our data suggest that mechanisms for regulation of ERK5 and downstream transcriptional pathways regulated by ERK5 are distinct from those of ERK1/2 in neurons. Furthermore, ERK5 is the first MAP kinase identified whose activity is stimulated by neurotrophins but not by neuronal activity.


Subject(s)
Cyclic AMP/metabolism , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factors/metabolism , Neurons/metabolism , Signal Transduction/physiology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cyclic AMP/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Inhibitors/pharmacology , Genes, Reporter , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , MAP Kinase Kinase 5 , MEF2 Transcription Factors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase 7 , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Myogenic Regulatory Factors/biosynthesis , Neurons/cytology , PC12 Cells , Phosphorylation , Potassium Chloride/metabolism , Potassium Chloride/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
6.
J Cell Biol ; 150(2): 335-47, 2000 Jul 24.
Article in English | MEDLINE | ID: mdl-10908576

ABSTRACT

Nitric oxide is a chemical messenger implicated in neuronal damage associated with ischemia, neurodegenerative disease, and excitotoxicity. Excitotoxic injury leads to increased NO formation, as well as stimulation of the p38 mitogen-activated protein (MAP) kinase in neurons. In the present study, we determined if NO-induced cell death in neurons was dependent on p38 MAP kinase activity. Sodium nitroprusside (SNP), an NO donor, elevated caspase activity and induced death in human SH-SY5Y neuroblastoma cells and primary cultures of cortical neurons. Concomitant treatment with SB203580, a p38 MAP kinase inhibitor, diminished caspase induction and protected SH-SY5Y cells and primary cultures of cortical neurons from NO-induced cell death, whereas the caspase inhibitor zVAD-fmk did not provide significant protection. A role for p38 MAP kinase was further substantiated by the observation that SB203580 blocked translocation of the cell death activator, Bax, from the cytosol to the mitochondria after treatment with SNP. Moreover, expressing a constitutively active form of MKK3, a direct activator of p38 MAP kinase promoted Bax translocation and cell death in the absence of SNP. Bax-deficient cortical neurons were resistant to SNP, further demonstrating the necessity of Bax in this mode of cell death. These results demonstrate that p38 MAP kinase activity plays a critical role in NO-mediated cell death in neurons by stimulating Bax translocation to the mitochondria, thereby activating the cell death pathway.


Subject(s)
Apoptosis/physiology , Mitogen-Activated Protein Kinases/metabolism , Neurons/metabolism , Nitric Oxide/metabolism , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/metabolism , Translocation, Genetic/physiology , Caspases/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Humans , Neuroblastoma , Nitroprusside/pharmacology , Tumor Cells, Cultured , bcl-2-Associated X Protein , p38 Mitogen-Activated Protein Kinases
7.
J Neurosci ; 20(7): 2567-74, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10729337

ABSTRACT

Glycogen synthase kinase-3beta (GSK3beta) activity is negatively regulated by several signal transduction cascades that protect neurons against apoptosis, including the phosphatidylinositol-3 kinase (PI-3 kinase) pathway. This suggests the interesting possibility that activation of GSK3beta may contribute to neuronal apoptosis. Consequently, we evaluated the role of GSK3beta in apoptosis in cultured cortical neurons induced by trophic factor withdrawal or by PI-3 kinase inhibition. Neurons were subjected to several apoptotic paradigms, including serum deprivation, serum deprivation combined with exposure to NMDA receptor antagonists, or treatment with PI-3 kinase inhibitors. These treatments all led to stimulation of GSK3beta activity in cortical neurons, which preceded the induction of apoptosis. Expression of an inhibitory GSK3beta binding protein or a dominant interfering form of GSK3beta reduced neuronal apoptosis, suggesting that GSK3beta contributes to trophic factor withdrawal-induced apoptosis. Furthermore, overexpression of GSK3beta in neurons increased apoptosis, indicating that activation of this enzyme is sufficient to trigger programmed cell death. Although destabilization of beta-catenin is an important physiological effect of GSK3beta activation, expression of a mutant beta-catenin that is not destabilized by GSK3beta did not protect against apoptosis. We conclude that inhibition of GSK3beta is one of the mechanisms by which PI-3 kinase activation protects neurons from programmed cell death.


Subject(s)
Apoptosis/physiology , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Microtubule-Associated Proteins/physiology , Neurons/physiology , Trans-Activators , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/pharmacology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Chromones/pharmacology , Cytoskeletal Proteins/metabolism , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/drug effects , beta Catenin
8.
Acta Neurobiol Exp (Wars) ; 60(4): 531-45, 2000.
Article in English | MEDLINE | ID: mdl-11200182

ABSTRACT

Neurotrophins promote survival and suppress apoptosis in many populations of neurons. Currently, phosphatidylinositol-3 kinase (PI-3K) is recognized as the main mediator of this protective effect. However, most of the data collected so far on the anti-apoptotic signaling of neurotrophins were obtained using trophic withdrawal paradigms. Recent data from our and other groups indicate that extracellular-signal-regulated kinase 1/2 (Erk 1/2) may play a critical role in suppressing neuronal apoptosis triggered by cellular damage. Thus, it appears that either Erk1/2 or PI-3K, depending on the nature of the death-inducing stimulus, can mediate anti-apoptotic signaling of neurotrophins. In this review, we discuss the contribution of Erk1/2 and PI-3K to neuroprotection by neurotrophins. We also present data suggesting possible mechanisms by which these pathways might suppress neuronal death.


Subject(s)
Apoptosis/physiology , Nerve Growth Factor/physiology , Neurons/physiology , Signal Transduction/physiology , Animals , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Neurons/cytology , Phosphatidylinositol 3-Kinases/metabolism
9.
Neurotox Res ; 1(4): 299-310, 2000 Apr.
Article in English | MEDLINE | ID: mdl-12835097

ABSTRACT

High doses of NMDA antagonists e.g. (+)MK-801 evoke neurodegeneration in retrosplenial cortex in rodents. To assess functional consequences of such treatment, three paradigms of two-way active avoidance learning (with visual or auditory conditioned stimuli) and additionally a spatial learning paradigm - radial maze - were used. Female rats were treated i.p. with 5 mg/kg of (+)MK-801. Recumbence, severe hypothermia and loss of body weight were observed for 3-7 days. Despite that, there were no statistically significant differences in performance of avoidance reaction between saline and (+)MK-801 treated animals trained 10-40 days after the drug administration. However, in the radial maze test (+)MK-801 impaired reference (but not working) memory in the experiment that started 8 days after the treatment. Similar effect was observed on reversal learning. The clinically used NMDA receptor antagonist memantine at the doses of 20 and 40 mg/kg had also no such long term negative effect on working memory during training (even positive effect was seen at 20 mg/kg) but at 40 mg/kg impaired learning on the first day of reversal. This indicates that (+)MK-801 neurotoxicity in the retrosplenial cortex is connected with subtle alterations in the learning performance that may be seen in some tests only. Moreover, memantine doses greatly exceeding therapeutically relevant range produce minimal functional alteration. An additional experiment revealed that the same dose of memantine results in two fold higher serum levels of the antagonist in female than male rats. Hence, considering that profiling studies are done in male rats, a safety factor of over 16 fold can be calculated for memantine.

10.
J Biol Chem ; 274(40): 28106-12, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10497161

ABSTRACT

Understanding of biological function of AP-1 transcription factor in central nervous system may greatly benefit from identifying its target genes. In this study, we present several lines of evidence implying AP-1 in regulating expression of tissue inhibitor of metalloproteinases-1 (timp-1) gene in rodent hippocampus in response to increased neuronal excitation. Such a notion is supported by the findings that timp-1 mRNA accumulation occurs in the rat hippocampus after either kainate- or pentylenetetrazole-evoked seizures with a delayed, in comparison with AP-1 components, time course, as well as with spatial overlap with c-Fos protein (major inducible AP-1 component) expression. Furthermore, AP-1 sequence derived from timp-1 promoter is specifically bound by hippocampal AP-1 proteins after treating the rats with either pro-convulsive agent. Finally, timp-1 promoter responds to excitatory activation both in vivo, in transgenic mice harboring the timp-LacZ gene construct, and in vitro in neurons of the hippocampal dentate gyrus cultures. These findings suggest that the AP-1 transcription factor may exert its role in the brain through affecting extracellular matrix remodeling.


Subject(s)
Dentate Gyrus/metabolism , Gene Expression Regulation/physiology , Neurons/physiology , Tissue Inhibitor of Metalloproteinase-1/genetics , Transcription Factor AP-1/physiology , Animals , Cells, Cultured , Dentate Gyrus/cytology , Lac Operon , Male , Mice , Mice, Transgenic , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Regulatory Sequences, Nucleic Acid , Transcription Factor AP-1/metabolism
11.
J Biol Chem ; 274(32): 22569-80, 1999 Aug 06.
Article in English | MEDLINE | ID: mdl-10428835

ABSTRACT

Apoptosis is a form of programmed cell death that plays a pivotal role during development and in the homeostasis of the adult nervous systems. However, mechanisms that regulate neuronal apoptosis are not well defined. Here, we report that brain-derived neurotrophic factor (BDNF) protects cortical neurons against apoptosis induced by camptothecin or serum deprivation and activates the extracellular-signal-regulated kinase (ERK) and the phosphatidylinositol 3-kinase (PI 3-kinase) pathways. Using pharmacological agents and transient transfection with dominant interfering or constitutive active components of the ERK or the PI 3-kinase pathway, we demonstrate that the ERK pathway plays a major role in BDNF neuroprotection against camptothecin. Furthermore, ERK is activated in cortical neurons during camptothecin-induced apoptosis, and inhibition of ERK increases apoptosis. In contrast, the PI 3-kinase pathway is the dominant survival mechanism for serum-dependent survival under normal culture conditions and for BDNF protection against serum withdrawal. These results suggest that the ERK pathway is one of several neuroprotective mechanisms that are activated by stress to counteract death signals in central nervous system neurons. Furthermore, the relative contribution of the ERK and PI 3-kinase pathways to neuronal survival may depend on the type of cellular injury.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cerebral Cortex/metabolism , Neurons/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Animals , Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Camptothecin/pharmacology , Cells, Cultured , Cerebral Cortex/cytology , Chromones/pharmacology , Culture Media, Serum-Free/pharmacology , Flavonoids/pharmacology , Morpholines/pharmacology , Neuroprotective Agents/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Rats , Rats, Sprague-Dawley , Signal Transduction , Topoisomerase I Inhibitors
12.
Neuroscience ; 86(2): 499-510, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9881864

ABSTRACT

In order to explore the role of CREM (cyclic-AMP responsive element modulator) gene expression in the function of the central nervous system, the gene transcripts were investigated in the rat brain in several conditions linked to increased neuronal activity. Up-regulation of CREM messenger RNA levels in the hippocampus was found to follow intraperitoneal administration of kainate (10 mg/kg). This increase was observed in both the dentate gyrus and hippocampus proper (CA subfields) and reached its maximum at 6 h after the treatment. Intrahippocampal injection of N-methyl-D-aspartate (200 nmol) resulted in elevated CREM messenger RNA expression as well. A similar increase of the messenger RNA abundance was also observed in the retrosplenial cortex after treating the female rats with a high dose (5 mg/kg) of dizocilpine maleate, an N-methyl-D-aspartate receptor antagonist. All these conditions are linked to neuronal excitation and neurodegeneration. However, an increase in CREM messenger RNA accumulation was also observed in the visual cortex after exposure of dark-adapted animals to the light, a procedure linked to neuronal plasticity. In the latter condition, it was found that CREM messenger RNA reached its highest levels at 6 h, i.e. later than the maximal increase of expression of immediate early genes such as c-fos, jun B and zif268, observed 45 min following the onset of visual stimulation. The ICER (inducible cyclic-AMP early repressor) form of CREM messenger RNA was identified to be induced by the light exposure. Finally, it was also found that cycloheximide, an inhibitor of protein synthesis, overinduces CREM/ICER gene expression. Together, these data suggest that CREM/ICER may be responsive to neuronal activation. Furthermore, given that CREM products have been shown previously to down-regulate expression of immediate early genes in vitro, they suggest that ICER may function as a molecular switch involved in down-regulation of immediate early gene expression in the rat brain.


Subject(s)
Brain/physiology , DNA-Binding Proteins/genetics , Gene Expression Regulation/drug effects , Immediate-Early Proteins , Kainic Acid/pharmacology , N-Methylaspartate/pharmacology , Neuronal Plasticity/physiology , Transcription, Genetic , Animals , Brain/drug effects , Cyclic AMP/physiology , Cyclic AMP Response Element Modulator , Darkness , Dizocilpine Maleate/pharmacology , Early Growth Response Protein 1 , Female , Functional Laterality , Genes, fos , Genes, jun , Hippocampus/drug effects , Hippocampus/physiology , In Situ Hybridization , Light , Male , Photic Stimulation , RNA, Messenger/genetics , Rats , Rats, Wistar , Repressor Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic/drug effects
13.
Exp Neurol ; 147(2): 229-37, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9344548

ABSTRACT

Single administration of a high dose of an uncompetitive NMDA receptor antagonist-dizocilpine maleate (MK-801)-results in transient neuronal vacuolization and cell death in retrosplenial cortex in rodents. In this study expression of cathepsin D (CatD), a major lysosomal aspartic protease, was investigated in brains of female rats treated with 1, 5, or 10 mg/kg of MK-801. Northern blot analysis demonstrated that the CatD mRNA level was moderately increased in retrosplenial cortex 24 h-7 days after the treatment. Concomitantly, increased CatD immunoreactivity was observed, predominantly in the degenerating neurons in layer III of retrosplenial cortex. Neuronal response was spatially distinguished from glial reactivation marked by increased mRNA and protein levels of glial fibrillary acidic protein, as demonstrated by Northern blot and immunohistochemistry in retrosplenial cortex 24 h-7 days after MK-801 treatment. These data suggest that activation of the lysosomal proteolytic system of neurons may play a role in MK-801-evoked neurodegeneration.


Subject(s)
Cathepsin D/biosynthesis , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Nerve Degeneration/chemically induced , Nerve Tissue Proteins/biosynthesis , Neurons/drug effects , Parietal Lobe/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cathepsin D/genetics , Dizocilpine Maleate/toxicity , Enzyme Induction/drug effects , Excitatory Amino Acid Antagonists/toxicity , Female , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Nerve Tissue Proteins/genetics , Neurons/enzymology , Neurons/ultrastructure , Parietal Lobe/enzymology , Parietal Lobe/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/physiology , Vacuoles/drug effects
14.
J Biol Chem ; 272(30): 18628-35, 1997 Jul 25.
Article in English | MEDLINE | ID: mdl-9228031

ABSTRACT

Lysosomal acid phosphatase (LAP) is a tartrate-sensitive enzyme with ubiquitous expression. Neither the physiological substrates nor the functional significance is known. Mice with a deficiency of LAP generated by targeted disruption of the LAP gene are fertile and develop normally. Microscopic examination of various peripheral organs revealed progredient lysosomal storage in podocytes and tubular epithelial cells of the kidney, with regionally different ultrastructural appearance of the stored material. Within the central nervous system, lysosomal storage was detected to a regionally different extent in microglia, ependymal cells, and astroglia concomitant with the development of a progressive astrogliosis and microglial activation. Whereas behavioral and neuromotor analyses were unable to distinguish between control and deficient mice, approximately 7% of the deficient animals developed generalized seizures. From the age of 6 months onward, conspicuous alterations of bone structure became apparent, resulting in a kyphoscoliotic malformation of the lower thoracic vertebral column. We conclude from these findings that LAP has a unique function in only a subset of cells, where its deficiency causes the storage of a heterogeneously appearing material in lysosomes. The causal relationship of the enzyme defect to the clinical manifestations remains to be determined.


Subject(s)
Acid Phosphatase/deficiency , Central Nervous System Diseases/enzymology , Kidney Diseases/enzymology , Lysosomal Storage Diseases/enzymology , Lysosomes/enzymology , Acid Phosphatase/genetics , Animals , Antigens, CD/metabolism , Bone and Bones/abnormalities , Cathepsin D/metabolism , Central Nervous System Diseases/pathology , Fibroblasts/enzymology , Kidney Diseases/pathology , Lysosomal Membrane Proteins , Membrane Glycoproteins/metabolism , Mice , Microglia/enzymology , Microglia/pathology , Phenotype , Seizures/enzymology , Tartrates/pharmacology
15.
Acta Biochim Pol ; 44(4): 781-9, 1997.
Article in English | MEDLINE | ID: mdl-9584860

ABSTRACT

Kainate is a glutamate analog that produces neuronal excitation resulting in seizures within hours following its intraperitoneal injection into adult rats. Then, at 2-3 days after the treatment, neurodegeneration of apoptotic character can be observed in limbic system. As a consequence, plastic reorganization and glial reactivation phenomena occur. These physiological and pathological responses are reflected by specific changes in gene expression, that can be dissected according to their spatio-temporal patterns. The early phase of gene expression observed in all hippocampal subfields appears to reflect a sudden burst of spiking activity. Changes in mRNA levels restricted to dentate gyrus are suggestive of a link to neuronal plasticity. The late gene expression response implies its correlation either to neuronal cell death or glial reactivation, depending on cellular localization of gene products. Thus analysis of the temporal and spatial gene expression pattern in the hippocampus after kainate treatment may provide clues revealing specific phenomena to which gene expression could be attributed.


Subject(s)
Brain/drug effects , Brain/metabolism , Gene Expression/drug effects , Kainic Acid/pharmacology , Animals , Dystrophin/genetics , Excitatory Amino Acids/pharmacology , Models, Neurological , Nerve Degeneration/chemically induced , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Glutamate/drug effects , Receptors, Glutamate/metabolism , Seizures/chemically induced , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Exp Neurol ; 136(1): 53-63, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7589334

ABSTRACT

Expression patterns of cathepsin D (lysosomal aspartic protease) and glial fibrillary acidic protein (GFAP, a marker of reactive astroglia) were determined by Northern blot analysis and immunohistochemistry in the rat brain during neurodegeneration accompanying kainate-evoked seizures. The level of cathepsin D mRNA in the hippocampus, limbic cortex, and temporo-parieto-occipital neocortex was shown to increase, starting at 6 h after kainate treatment, and reaching peak values at 3-7 days after the neurotoxin administration. A similar time course of elevated accumulation was noted for GFAP mRNA in these structures. Immunohistochemical analysis performed 3 days after kainate treatment showed that the increased cathepsin D levels were confined mainly to the degenerating neurons in the susceptible brain areas, while the elevated GFAP immunoreactivity was observed in reactive astrocytes. Although cathepsin D and GFAP expression levels were elevated by kainate administration, their expression patterns revealed significant differences with regard to both intensity and site of induction.


Subject(s)
Brain/metabolism , Cathepsin D/genetics , Nerve Degeneration , Animals , Blotting, Northern , Cathepsin D/metabolism , Cerebral Cortex/immunology , Glial Fibrillary Acidic Protein/genetics , Immunohistochemistry , Kainic Acid/pharmacology , Male , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Seizures/chemically induced , Time Factors
17.
EMBO J ; 14(15): 3599-608, 1995 Aug 01.
Article in English | MEDLINE | ID: mdl-7641679

ABSTRACT

Mice deficient for the major lysosomal aspartic proteinase cathepsin D, generated by gene targeting, develop normally during the first 2 weeks, stop thriving in the third week and die in a state of anorexia at day 26 +/- 1. An atrophy of the ileal mucosa first observed in the third week progresses towards widespread intestinal necroses accompanied by thromboemboli. Thymus and spleen undergo massive destruction with fulminant loss of T and B cells. Lysosomal bulk proteolysis is maintained. These results suggest, that vital functions of cathepsin D are exerted by limited proteolysis of proteins regulating cell growth and/or tissue homeostasis, while its contribution to bulk proteolysis in lysosomes appears to be non-critical.


Subject(s)
Cathepsin D/deficiency , Intestinal Mucosa/pathology , Lysosomes/enzymology , Spleen/pathology , Thymus Gland/pathology , Animals , Atrophy , B-Lymphocytes/pathology , Cathepsin D/antagonists & inhibitors , Cathepsin D/biosynthesis , Cathepsin D/genetics , Chimera , Fibroblasts/metabolism , Gene Targeting , Ileum/pathology , Leupeptins/pharmacology , Male , Mice , Mice, Inbred C57BL , Pepstatins/pharmacology , RNA, Messenger/biosynthesis , Specific Pathogen-Free Organisms , T-Lymphocytes/pathology
18.
Neuroreport ; 5(12): 1538-40, 1994 Jul 21.
Article in English | MEDLINE | ID: mdl-7948857

ABSTRACT

Cell death occurs in many neuropathological conditions. However, the mechanisms governing this process(es) remain generally unknown. In this report we studied whether excitotoxic neuronal death evoked by kainic acid (KA) in rat brain is associated with ladder-like DNA fragmentation. DNA was isolated from hippocampi, entorhinal and sensory cortices at various times following intraperitoneal KA (10 mg kg-1) injections. Typical oligonucleosome-sized DNA fragmentation was observed in all three structures at 18 h and 72 h following KA administration. These findings were further confirmed by in situ nick-translation. DNA fragmentation is believed to be diagnostic for apoptosis. The clear ladders of DNA fragmentation appeared after 18 h, although slight degradation was observed as early as 12 h after KA administration.


Subject(s)
Brain/drug effects , DNA Damage , Kainic Acid/toxicity , Neurotoxins/toxicity , Animals , Brain/metabolism , Brain/pathology , Cell Death/drug effects , DNA/drug effects , DNA/isolation & purification , Genetic Techniques , Injections, Intraperitoneal , Kainic Acid/administration & dosage , Male , Neurotoxins/administration & dosage , Rats , Rats, Wistar , Time Factors
19.
DNA Cell Biol ; 13(4): 419-27, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8011168

ABSTRACT

A cloned mouse genomic DNA fragment containing the gene encoding cathepsin D (Catd) encompasses 11 kb of genomic DNA and is composed of 9 exons. Using recombinant inbred strains, we localized the Catd gene on chromosome 4, tightly linked to the loci Mtv-13, Cyp4a, Ms15-1, and Pmv-19. The exon-intron organization of the Catd gene was shown to be very similar to that of its human counterpart. Presence of a CpG island, absence of a TATA box, and initiation of transcription at more than one site indicate that the Catd gene is a "housekeeping" gene. A 1.2-kb fragment containing the 5'-flanking region of the gene displayed promoter activity in BHK-21 cells. Comparison of the nucleotide sequences of mouse and human cathepsin D promoter regions revealed conservation of three potential regulatory elements: an E box, a GC box and a potential cAMP-responsive element. In contrast to the 5' region of human cathepsin D, the murine gene contains three CCAAT boxes but lacks any of the four AP2 binding sites found in the human gene.


Subject(s)
Cathepsin D/genetics , Promoter Regions, Genetic , Restriction Mapping , Animals , Base Sequence , Cell Line , DNA , Exons , Humans , Introns , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Molecular Sequence Data , Regulatory Sequences, Nucleic Acid , Sequence Homology, Nucleic Acid , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...