Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Pharmacol Biochem Behav ; 101(3): 479-86, 2012 May.
Article in English | MEDLINE | ID: mdl-22366219

ABSTRACT

Cholinergic deficit is one of the most remarkable symptoms and plays an important role in Alzheimer's disease. In the present study, the protective effects of Akebia saponin D (ASD) on learning and memory impairments induced by excitatory neurotoxin ibotenic acid injection were examined in vivo. Our findings suggest that ASD (90 mg/kg, p.o.) would exert a rescue effect on rats both in behavioral performances in Morris water maze and Y maze and cholinergic functions detected by chemical methods. We further investigated in the hippocampus and found ASD could regulate apoptosis-related proteins expression following ibotenic acid injection. Additionally, mitogen-activated protein kinase (MAPK) family phosphorylations were inhibited after ASD treatment, implicating that the MAPK signaling pathway could be involved in the mechanism underlying neuroprotection of ASD against ibotenic acid-induced excitotoxicity.


Subject(s)
Cognition Disorders/chemically induced , Cognition Disorders/prevention & control , Ibotenic Acid/toxicity , MAP Kinase Signaling System/drug effects , Neuroprotective Agents/pharmacology , Saponins/pharmacology , Acetylcholine/metabolism , Acetylcholinesterase/metabolism , Animals , Apoptosis/drug effects , Brain/drug effects , Brain/metabolism , Brain/pathology , Choline O-Acetyltransferase/metabolism , Cognition Disorders/physiopathology , Cognition Disorders/psychology , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Ibotenic Acid/antagonists & inhibitors , MAP Kinase Signaling System/physiology , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , Memory/physiology , Rats , Rats, Sprague-Dawley
2.
Neuroscience ; 167(3): 716-23, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20188153

ABSTRACT

Glutamate excitotoxicity is among the main cellular mechanisms leading to perinatal insults in human newborns. We used intracerebral injection of the glutamatergic glutamate N-methyl-D-aspartate-receptor agonist ibotenate to produce excitotoxic lesions mimicking the acquired white matter lesions seen in human preterm infants. We evaluated whether nonsteroidal antiinflammatory drugs (NSAIDs) protected against glutamate excitotoxicity. Aspirin (0.01-100 microg/d), indomethacin (0.1-10 microg/d), paracetamol (10-100 microg/d), or NS-398 (12.5 microg/d) was given daily before ibotenate (P1 to P5) or after ibotenate (P5 to P9). Lesion size was measured on Cresyl Violet-stained brain sections collected on P10. None of the drugs tested alone or in combination increased lesion size. Pretreatment with low- or high-dose aspirin and post-treatment with paracetamol or NS-398 protected against white matter lesions, whereas cortical lesions were decreased by pretreatment with low- or high-dose aspirin or post-treatment with NS-398. The corticosteroid betamethasone (0.18 microg/d) was neuroprotective when given before or after ibotenate and this effect was reversed by concomitant aspirin therapy (10 microg/d). In conclusion, perinatal NSAID administration may have beneficial effects on brain injury if appropriately timed.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Brain Damage, Chronic/drug therapy , Encephalitis/drug therapy , Neuroprotective Agents/pharmacology , Neurotoxins/antagonists & inhibitors , Pregnancy Complications/drug therapy , Acetaminophen/pharmacology , Acetaminophen/therapeutic use , Adrenal Cortex Hormones/antagonists & inhibitors , Adrenal Cortex Hormones/pharmacology , Animals , Animals, Newborn , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Aspirin/pharmacology , Aspirin/therapeutic use , Betamethasone/antagonists & inhibitors , Betamethasone/pharmacology , Brain/drug effects , Brain/growth & development , Brain/pathology , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Drug Therapy, Combination , Encephalitis/metabolism , Encephalitis/physiopathology , Female , Ibotenic Acid/antagonists & inhibitors , Ibotenic Acid/toxicity , Indomethacin/pharmacology , Indomethacin/therapeutic use , Mice , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Myelinated/pathology , Neuroprotective Agents/therapeutic use , Neurotoxins/toxicity , Pregnancy , Pregnancy Complications/physiopathology , Pregnancy Complications/prevention & control , Treatment Outcome
3.
Eur J Pharmacol ; 588(1): 58-63, 2008 Jun 24.
Article in English | MEDLINE | ID: mdl-18466899

ABSTRACT

Periventricular leukomalacia is a major cause of cerebral palsy. Perinatal white matter lesions associated with cerebral palsy appears to involve glutamate excitotoxicity. When injected intracerebrally into newborn mice, the glutamatergic analog, ibotenate, induces white matter cysts mimicking human periventricular leukomalacia. Intraperitoneal injection of melatonin was previously shown to be neuroprotective in this mouse model. The goal of the present study was to compare in this model the protective effects of agomelatine (S 20098), a melatonin derivative, with melatonin. Mice that received intraperitoneal S 20098 or melatonin had significant reductions in size of ibotenate-induced white matter cysts when compared with controls. Although agomelatine and melatonin did not prevent the initial appearance of white matter lesions, they did promote secondary lesion repair. Interestingly, while melatonin effects were only observed when given within the first two hours following the excitotoxic insult, agomelatine was still significantly neuroprotective when administered eight hours after the insult. The protective effects of agomelatine and melatonin were counter-acted by co-administration of luzindole or S 20928, two melatonin receptor antagonists. Agomelatine, acting through melatonin receptors, could represent a promising new drug for treating human periventricular leukomalacia and have beneficial effects on neuroplasticity.


Subject(s)
Acetamides/pharmacology , Brain Diseases/prevention & control , Excitatory Amino Acid Agonists/toxicity , Ibotenic Acid/toxicity , Neuroprotective Agents , Receptor, Serotonin, 5-HT2C/drug effects , Receptors, Melatonin/agonists , Serotonin Antagonists , Acetamides/antagonists & inhibitors , Acetamides/toxicity , Animals , Brain/pathology , Brain Diseases/chemically induced , Brain Diseases/pathology , Ibotenic Acid/antagonists & inhibitors , Melatonin/pharmacology , Mice , Receptors, Melatonin/antagonists & inhibitors , Tryptamines/antagonists & inhibitors , Tryptamines/toxicity
4.
Neurobiol Dis ; 24(2): 357-66, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16959492

ABSTRACT

Using an established mouse model of human periventricular leukomalacia, we investigated whether EPO could reduce excitotoxic damage. When administered 1 h following intracerebral injection of 10 microg ibotenic acid at day 5 of life, both a single injection of EPO (5000 IU/kg bw) and repetitive administrations of EPO reduced white and gray matter lesion size. The therapeutic window for protection was small as the protective effect of EPO was lost when EPO administration was delayed to 4 h post-insult. EPO-mediated upregulation of EPO-R, but not EPO, mRNA was observed within 4 h of the excitotoxic insult. The EPO effect was gender independent. Minor hematopoetic effects were observed following EPO treatment. We conclude that a single dose of EPO is sufficient to reduce excitotoxic brain injury and may therefore possess therapeutic relevance in the clinical setting.


Subject(s)
Erythropoietin/pharmacology , Leukomalacia, Periventricular/drug therapy , Nerve Degeneration/drug therapy , Neuroprotective Agents/pharmacology , Neurotoxins/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Animals, Newborn , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Cytoprotection/drug effects , Cytoprotection/physiology , Disease Models, Animal , Drug Administration Schedule , Erythropoietin/therapeutic use , Female , Glutamic Acid/metabolism , Humans , Ibotenic Acid/antagonists & inhibitors , Ibotenic Acid/metabolism , Infant, Newborn , Injections, Intraventricular , Leukomalacia, Periventricular/metabolism , Leukomalacia, Periventricular/physiopathology , Male , Mice , Nerve Degeneration/physiopathology , Nerve Degeneration/prevention & control , Neuroprotective Agents/therapeutic use , Neurotoxins/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, Erythropoietin/genetics , Receptors, N-Methyl-D-Aspartate/agonists , Time Factors
5.
Brain Res ; 1058(1-2): 183-8, 2005 Oct 05.
Article in English | MEDLINE | ID: mdl-16140286

ABSTRACT

Dietary supplementation with creatine has proven to be beneficial in models of acute and chronic neurodegeneration. We report here data on the neurochemical correlates of differential protection of long-term creatine supplementation in two models of excitotoxicity in rats, as well as in the mouse model for ALS (G93A mice). In rats, the fall in cholinergic and GABAergic markers due to the excitotoxic death of intrinsic neurons caused by intrastriatal infusion of the neurotoxin, ibotenic acid, was significantly prevented by long-term dietary supplementation with creatine. On the contrary, creatine was unable to recover a cholinergic marker in the cortex of rats subjected to the excitotoxic death of the cholinergic basal forebrain neurons. In G93A mice, long-term creatine supplementation marginally but significantly increased mean lifespan, as previously observed by others, and reverted the cholinergic deficit present in some forebrain areas at an intermediate stage of the disease. In both rats and mice, creatine supplementation increased the activity of the GABAergic enzyme, glutamate decarboxylase, in the striatum but not in other brain regions. The present data point at alterations of neurochemical parameters marking specific neuronal populations, as a useful way to evaluate neuroprotective effects of long-term creatine supplementation in animal models of neurodegeneration.


Subject(s)
Brain/drug effects , Creatine/pharmacology , Dietary Supplements , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Neurotoxins/antagonists & inhibitors , Acetylcholine/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Basal Nucleus of Meynert/drug effects , Basal Nucleus of Meynert/metabolism , Basal Nucleus of Meynert/physiopathology , Biomarkers/analysis , Biomarkers/metabolism , Brain/metabolism , Brain/physiopathology , Cell Death/drug effects , Cell Death/physiology , Cholinergic Fibers/drug effects , Cholinergic Fibers/metabolism , Cholinergic Fibers/pathology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Creatine/therapeutic use , Glutamate Decarboxylase/drug effects , Glutamate Decarboxylase/metabolism , Ibotenic Acid/antagonists & inhibitors , Male , Mice , Mice, Transgenic , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/therapeutic use , Neurotoxins/toxicity , Rats , Rats, Wistar , Survival Rate , Time , Treatment Outcome
6.
Neurobiol Dis ; 20(3): 837-48, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16009561

ABSTRACT

Brain lesions induced in newborn mice by the glutamatergic agonists ibotenate (acting on NMDA and metabotropic receptors) and S-bromowillardiine (acting on AMPA-kainate receptors) mimic some aspects of white matter cysts and transcortical necrosis observed in human perinatal brain damage. Topiramate (TPM), already used in children to manage newly diagnosed and refractory epilepsy, has potential neuroprotective effects that may be useful in human perinatal brain lesions. In the excitotoxic newborn mouse model, TPM provided dose-dependent and long-lasting protection of developing white matter and cortical plate against S-bromowillardiine. TPM had no significant effect on ibotenate-induced brain lesions. TPM-induced neuroprotection potentially involved increased survival of pre-oligodendrocytes, decreased neuronal apoptosis, inhibition of microglial activation and astrogliosis, and decreased seizure activity. Diazepam, phenytoin, and carbamazepine had no neuroprotective effect in this model. The present study provides experimental support for the consideration of TPM as a candidate therapy for excitotoxic perinatal brain lesions.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Fructose/analogs & derivatives , Hypoxia, Brain/drug therapy , Hypoxia, Brain/prevention & control , Neurotoxins/antagonists & inhibitors , Alanine/analogs & derivatives , Alanine/antagonists & inhibitors , Alanine/toxicity , Animals , Animals, Newborn , Brain/drug effects , Brain/growth & development , Brain/physiopathology , Cerebral Palsy/drug therapy , Cerebral Palsy/physiopathology , Cerebral Palsy/prevention & control , Disease Models, Animal , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/metabolism , Excitatory Amino Acid Agonists/toxicity , Excitatory Amino Acid Antagonists/therapeutic use , Female , Fructose/pharmacology , Fructose/therapeutic use , Glutamic Acid/metabolism , Humans , Hypoxia, Brain/physiopathology , Ibotenic Acid/antagonists & inhibitors , Ibotenic Acid/toxicity , Infant, Newborn , Leukomalacia, Periventricular/drug therapy , Leukomalacia, Periventricular/physiopathology , Leukomalacia, Periventricular/prevention & control , Male , Mice , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurotoxins/toxicity , Rats , Rats, Sprague-Dawley , Topiramate , Treatment Outcome
7.
Brain Res Dev Brain Res ; 141(1-2): 25-32, 2003 Mar 14.
Article in English | MEDLINE | ID: mdl-12644245

ABSTRACT

Interleukin-10 markedly reduces production of proinflammatory cytokines by activated microglia or macrophages and downregulates the expression of activating molecules on these cells. In studies performed in adults or in cell cultures, interleukin-10 protected against hypoxic-ischemic neuronal death and against lipopolysaccharide-mediated oligodendrocyte cell death. Furthermore, it was recently shown that interleukin-10 counteracts metabolic and microcirculatory effects of hypoxia-ischemia in the perinatal pig brain. Intracerebral injection of the glutamatergic analogue ibotenate to newborn mice induces cortical plate and white matter lesions mimicking the brain damage associated with cerebral palsy, and pretreatment with proinflammatory cytokines such as interleukin-1-beta or with interleukin-9 significantly exacerbates these lesions. The present study evaluated the influence of interleukin-10 on ibotenate-induced brain lesions in newborn mice under basal conditions or after exposure to cytokines. Intraperitoneal injection of interleukin-10 for 3 days following ibotenate significantly reduced the size of excitotoxic brain lesions. Intraperitoneal injection of neutralizing anti-interleukin-10 antibody for 3 days following ibotenate had no detectable effect and no difference in ibotenate-induced brain lesion size was found between wild type pups and pups deleted for the interleukin-10 gene, suggesting that endogenous interleukin-10 in newborn mice may have limited effects. Co-administration of intracerebral ibotenate and interleukin-10 had no detectable effect, arguing against a direct neuroprotective effect of interleukin-10 on neurons. While pretreatment with intraperitoneal interleukin-10 alone had no detectable effect on excitotoxic brain lesions, interleukin-10 given with interleukin-1-beta pretreatment blunted the toxic effects of interleukin-1-beta. On the other hand, combined pretreatment with IL-9 and anti-IL-10 antibody largely reversed the exacerbating effect of IL-9 on excitotoxic brain lesions. Altogether, these data suggest that, in newborn mice, exogenous interleukin-10 can be neuroprotective when acting in an inflammatory context.


Subject(s)
Cerebral Cortex/drug effects , Cerebral Palsy/drug therapy , Encephalitis/drug therapy , Interleukin-10/pharmacology , Neurotoxins/antagonists & inhibitors , Animals , Animals, Newborn , Cell Death/drug effects , Cell Death/genetics , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Cerebral Palsy/immunology , Cerebral Palsy/metabolism , Drug Interactions/physiology , Encephalitis/immunology , Encephalitis/metabolism , Female , Ibotenic Acid/antagonists & inhibitors , Interleukin-1/pharmacology , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-9/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , Nerve Degeneration/drug therapy , Nerve Degeneration/immunology , Nerve Degeneration/metabolism , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Myelinated/immunology , Nerve Fibers, Myelinated/metabolism , Survival Rate
8.
J Pharmacol Exp Ther ; 302(3): 881-8, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12183643

ABSTRACT

Glucagon-like peptide-1 (7-36)-amide (GLP-1) is an endogenous insulinotropic peptide that is secreted from the L cells of the gastrointestinal tract in response to food. It has potent effects on glucose-dependent insulin secretion, insulin gene expression, and pancreatic islet cell formation. In type 2 diabetes, GLP-1, by continuous infusion, can normalize blood glucose and is presently being tested in clinical trials as a therapy for this disease. More recently, GLP-1 has been found to have central nervous system (CNS) effects and to stimulate neurite outgrowth in cultured cells. We now report that GLP-1, and its longer-acting analog exendin-4, can completely protect cultured rat hippocampal neurons against glutamate-induced apoptosis. Extrapolating these effects to a well defined rodent model of neurodegeneration, GLP-1 and exendin-4 greatly reduced ibotenic acid-induced depletion of choline acetyltransferase immunoreactivity in basal forebrain cholinergic neurons. These findings identify a novel neuroprotective/neurotrophic function of GLP-1 and suggest that such peptides may have potential for halting or reversing neurodegenerative processes in CNS disorders, such as Alzheimer's disease, and in neuropathies associated with type 2 diabetes mellitus.


Subject(s)
Excitatory Amino Acid Antagonists/toxicity , Glucagon/pharmacology , Glutamic Acid/toxicity , Nerve Degeneration/chemically induced , Nerve Degeneration/prevention & control , Neurons/drug effects , Peptide Fragments/pharmacology , Peptides/pharmacology , Protein Precursors/pharmacology , Venoms , Animals , Basal Ganglia/pathology , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Choline O-Acetyltransferase/metabolism , Cyclic AMP/metabolism , Excitatory Amino Acid Agonists/toxicity , Exenatide , Glial Fibrillary Acidic Protein/metabolism , Glucagon/metabolism , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Hippocampus/cytology , Hippocampus/drug effects , Ibotenic Acid/antagonists & inhibitors , Ibotenic Acid/toxicity , Immunohistochemistry , Neurons/pathology , Parasympathetic Nervous System/drug effects , Peptide Fragments/metabolism , Protein Precursors/metabolism , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Receptors, Glucagon/drug effects , Receptors, Glucagon/metabolism
9.
FASEB J ; 16(3): 423-5, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11790724

ABSTRACT

The incidence of neurological disabilities ascribable to perinatal injury is rising in Western countries, raising ethical and financial problems. No curative treatments are available. The pathophysiology of brain lesions of hypoxic-ischemic or inflammatory origin involves various neurotransmitters or neuromodulators. Among these, glutamate plays a key role. By overactivating N-methyl-D-aspartate receptors, it triggers the excitotoxic cascade. Although addictive, nicotine prevents excitotoxic neuronal death in adult animals. Its potential neuroprotective effects have not been evaluated in neonates. We found that nicotine is neuroprotective in vivo, in a murine model of neonatal excitotoxic brain injury, and in vitro, in primary cultures of cortical neurons. We investigated the respective roles in nicotine-related neuroprotection of the two dominant nicotinic acetylcholine receptor (nAChR) isoforms, namely, alpha4beta2 (heteropentameric) and alpha7 (homopentameric). Inhibition of alpha4beta2, either pharmacological (i.e., an alpha4beta2 nAChR antagonist) or molecular (beta2-/- knockout mice), abolished the protective effect of nicotine in vivo and in vitro, suggesting the involvement of alpha4beta2 nAChR in neonatal nicotine-related neuroprotection. In contrast, activation of alpha7 nAChR, which is protective in adult animals, was deleterious in our neonatal model, whereas its blockade, either pharmacological or molecular (alpha7-/- knockout mice) provided neuroprotection. Neuroprotective strategies must consider these opposite properties of distinct nAChR isoforms in neonates.


Subject(s)
Brain Diseases/pathology , Receptors, Nicotinic/metabolism , Receptors, Nicotinic/physiology , Animals , Animals, Newborn , Autoradiography , Brain Diseases/chemically induced , Brain Diseases/metabolism , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/growth & development , Cerebral Cortex/pathology , Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Agonists/pharmacology , Ibotenic Acid/administration & dosage , Ibotenic Acid/antagonists & inhibitors , Injections , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Neurological , N-Methylaspartate/metabolism , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Nicotine/pharmacology , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/genetics , alpha7 Nicotinic Acetylcholine Receptor
10.
J Pharmacol Exp Ther ; 288(3): 1207-13, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10027860

ABSTRACT

Periventricular leukomalacia (PVL), a necrotic and often cystic lesion of the cerebral white matter occurring in very premature babies, is the leading cause of cerebral palsy in this population. Increased glutamate release and the excitotoxic cascade thus triggered may be critical factors in the development of PVL. The glutamatergic analog ibotenate injected intracerebrally into newborn mice produces white matter cysts that mimic human PVL. Concomitant injection of vasoactive intestinal peptide (VIP), a trophic factor, protects the white matter against excitotoxic lesions. The goal of the present study was to assess the protective properties of systemically injected VIP analogs against ibotenate-induced excitotoxic white matter lesions in newborn mice. VIP analogs were selected on the basis of their low susceptibility to endopeptidases and their potential ability to cross biological membranes. RO-25-1553, a long-lasting cyclic VIP analog, and stearyl-norleucine-VIP, a fatty derivative of VIP, reduced ibotenate-induced white matter cysts by up to 87% and 84%, respectively, when injected i.p. immediately after ibotenate. By comparison, i.p. coadministration of VIP and ibotenate was not protective against the excitotoxic insult. Furthermore, RO-25-1553 and stearyl-norleucine-VIP still induced significant neuroprotection of the developing white matter when injected systemically 8 and 12 h, respectively, after ibotenate, establishing these peptides as therapeutic agents in this murine model. VIP analogs may have therapeutic potential in human premature babies at high risk for PVL.


Subject(s)
Brain/drug effects , Neuroprotective Agents/pharmacology , Vasoactive Intestinal Peptide/analogs & derivatives , Amino Acid Sequence , Animals , Animals, Newborn , Brain/growth & development , Brain/pathology , Excitatory Amino Acid Agonists , Female , Humans , Ibotenic Acid/antagonists & inhibitors , Infant, Newborn , Injections, Intraperitoneal , Injections, Intraventricular , Leukomalacia, Periventricular/chemically induced , Leukomalacia, Periventricular/drug therapy , Male , Mice , Molecular Sequence Data , Peptides, Cyclic/pharmacology , Vasoactive Intestinal Peptide/administration & dosage , Vasoactive Intestinal Peptide/antagonists & inhibitors , Vasoactive Intestinal Peptide/pharmacology
11.
J Clin Invest ; 100(2): 390-7, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9218516

ABSTRACT

Excitotoxic damage may be a critical factor in the formation of brain lesions associated with cerebral palsy. When injected at birth, the glutamatergic analog ibotenate induces mouse brain lesions that strikingly mimic human microgyria. When ibotenate is injected at postnatal day 5, it produces transcortical necrosis and white matter cysts that mimic human perinatal hypoxic-like lesions. Vasoactive intestinal peptide (VIP) has potent growth-related actions and neuroprotective properties that influence mitosis and neuronal survival in culture. The goal of this study was to assess the protective role of VIP against excitotoxic lesions induced by ibotenate in developing mouse brain. VIP cotreatment reduced ibotenate-induced microgyric-like cortical lesions and white matter cysts by up to 77 and 85%, respectively. VIP protective effects were reproduced by a peptide derived from activity-dependent neurotrophic factor (ADNF), a trophic factor released by VIP-stimulated astrocytes, and by stearyl norleucine VIP, a specific VIP agonist that does not activate adenylate cyclase. Neither forskolin, an adenylate cyclase activator, nor pituitary adenylate cyclase-activating peptide, provided VIP-like protection. VIP and neurotrophic analogs, acting through a cAMP-independent mechanism and inducing ADNF release, could represent new avenues in the understanding and prevention of human cerebral palsy.


Subject(s)
Brain/drug effects , Excitatory Amino Acid Agonists/pharmacology , Ibotenic Acid/pharmacology , Neurons/drug effects , Vasoactive Intestinal Peptide/pharmacology , Animals , Binding Sites , Brain/cytology , Brain/growth & development , Brain/pathology , Cell Death , Cerebral Cortex/metabolism , Colforsin/pharmacology , Histocytochemistry , Ibotenic Acid/antagonists & inhibitors , Mice , Mice, Inbred Strains , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/pharmacology , Neurons/cytology , Neuropeptides/pharmacology , Neuroprotective Agents/pharmacology , Oligopeptides , Peptide Fragments/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide , Signal Transduction/physiology , Vasoactive Intestinal Peptide/metabolism
12.
Neurosci Lett ; 223(3): 161-4, 1997 Feb 28.
Article in English | MEDLINE | ID: mdl-9080457

ABSTRACT

The aim of this experiment was to test whether or not nerve growth factor (NGF) is involved in cholinergic processes in the avian brain, by injecting NGF into the higher vocal center (HVC) and examining its effects on adult male zebra finch song. Since NGF has been hypothesized to protect cells after injury, some birds received both NGF and ibotenic acid (IBO) lesions of HVC, while others received either NGF or IBO or neither (SHAM). Only the IBO-treated birds showed alterations in song. Although there was no evidence of cell preservation in the immunocytochemical and morphological analysis NGF appears to prevent the IBO induced impairment in song augmenting the activity of the remaining neurons and enhancing brain repair.


Subject(s)
Birds/physiology , Brain/drug effects , Brain/physiology , Nerve Growth Factors/pharmacology , Vocalization, Animal/drug effects , Animals , Brain/pathology , Choline O-Acetyltransferase/metabolism , Ibotenic Acid/antagonists & inhibitors , Ibotenic Acid/pharmacology , Immunohistochemistry , Male
13.
Neuroreport ; 7(1): 85-8, 1995 Dec 29.
Article in English | MEDLINE | ID: mdl-8742423

ABSTRACT

We investigated the effect of prolonged electroconvulsive shock (ECS) or imipramine treatment on cyclic AMP accumulation induced by ibotenate and glutamate in rat cerebral cortical slices. Prolonged imipramine or electroconvulsive shock treatment attenuated the ibotenate-induced increase in cyclic AMP accumulation and inhibited the synergistic interaction between ibotenate and noradrenaline; the glutamate-mediated inhibition of forskolin-stimulated cyclic AMP accumulation was not modified. Our results indicate that multiple effects of excitatory amino acids on cyclic AMP accumulation are modified differently by antidepressant treatment.


Subject(s)
Antidepressive Agents, Tricyclic/pharmacology , Cerebral Cortex/drug effects , Cyclic AMP/metabolism , Glutamic Acid/pharmacology , Ibotenic Acid/pharmacology , Imipramine/pharmacology , Analysis of Variance , Animals , Cerebral Cortex/metabolism , Colforsin/pharmacology , Drug Interactions , Electroshock , Ibotenic Acid/antagonists & inhibitors , In Vitro Techniques , Male , Norepinephrine/pharmacology , Rats , Rats, Wistar , Time Factors
14.
Brain Res ; 695(2): 289-92, 1995 Oct 16.
Article in English | MEDLINE | ID: mdl-8556348

ABSTRACT

Lithium was tested on an animal model of a brain cholinergic excitotoxic lesion. Male Wistar rats received unilaterally 50 nmol ibotenic acid in the nucleus basalis magnocellularis. Some were treated intraperitoneally with LiCl from two days before to six days after lesioning. Such treated rats showed less deficits than untreated lesioned animals on passive avoidance, ambulatory behaviour and choline acetyltransferase activity in the lesioned cortex. Lithium protection against excitatory amino acid neurotoxicity is suggested.


Subject(s)
Behavior, Animal/drug effects , Excitatory Amino Acid Agonists/toxicity , Ibotenic Acid/antagonists & inhibitors , Lithium/pharmacology , Parasympathetic Nervous System/physiology , Prosencephalon/physiology , Animals , Avoidance Learning/drug effects , Basal Ganglia/drug effects , Basal Ganglia/metabolism , Basal Ganglia/physiology , Choline O-Acetyltransferase/antagonists & inhibitors , Ibotenic Acid/toxicity , Male , Motor Activity/drug effects , Parasympathetic Nervous System/drug effects , Parasympathetic Nervous System/metabolism , Prosencephalon/drug effects , Prosencephalon/metabolism , Rats , Rats, Wistar
15.
Neuropharmacology ; 33(9): 1095-101, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7838322

ABSTRACT

The potential neuroprotective effects of phencyclidine (5 mg/kg i.p.) were assessed in rats which had been treated with the excitotoxin, ibotenic acid (IBO) (0.015 M) to lesion the nucleus basalis magnocellularis. IBO treated rats showed a significant impairment in 13 of the 25 test trials in the spatial navigation Morris water maze task and deficits in passive avoidance learning. Phencyclidine was found to prevent the IBO-induced impairment in 4 of the 13 test trials in which the IBO Morris maze deficit was observed and also successfully prevented the passive avoidance learning deficits. Neurochemically, IBO was shown to reduce the levels of gamma amino-n-butyric acid (GABA) in the cortex. This effect of IBO on the inhibitory GABAergic system may contribute to the direct toxic effects of IBO which is mediated through excitatory amino acid receptors. Phencyclidine had no effect on the changes in GABA produced by IBO. The effect of phencyclidine treatment on IBO behavioural toxicity observed in this study demonstrates that antagonism of the phencyclidine receptor site on the N-methyl-D-aspartate receptor complex may be partially protective against the excitotoxic damage induced by IBO.


Subject(s)
Avoidance Learning/drug effects , Ibotenic Acid/antagonists & inhibitors , Phencyclidine/pharmacology , Space Perception/drug effects , Animals , Basal Ganglia/drug effects , Basal Ganglia/physiology , Behavior, Animal/drug effects , Brain Chemistry/drug effects , Ibotenic Acid/toxicity , Male , Maze Learning/drug effects , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/metabolism
16.
Mol Cell Neurosci ; 5(3): 269-76, 1994 Jun.
Article in English | MEDLINE | ID: mdl-7522104

ABSTRACT

The metabotropic glutamate receptors (mGluRs) form a family of G-protein-coupled receptors which consists of at least seven members termed mGluR1-mGluR7. These members are classified into subfamilies according to their sequence similarities, signal transduction mechanisms and agonist selectivities. mGluR1 and mGluR5 are coupled to the phosphoinositide hydrolysis/Ca2+ signal transduction and efficiently respond to quisqualate. In this study, we have stably expressed mGluR1 in Chinese hamster ovary cells on which the activation of the phosphoinositide signal transduction pathway was evaluated by means of two methods and their degree of correspondence was analyzed. These two methods involve the Li(+)-dependent accumulation of [3H]inositol-labeled inositol phosphates or the [3H]cytidine-labeled phospholiponucleotide cytidine diphospho (CDP)- diacylglycerol (DAG). The correlation between the two measures was found to be generally uniform for the different agonists evaluated. However, the levels of CDP-DAG were found to be consistently higher. Furthermore, quisqualate showed a differential activity on the two methods behaving as a partial agonist and as a full agonist on the inositol phosphate and the CDP-DAG responses, respectively. On the same cells the activity of a series of carboxyphenylglycines recently described as possible new tools for investigating the role of mGluRs has been evaluated. Three phenylglycine derivatives were tested and found to be competitive antagonists at this mGluR subtype. They inhibited both the phosphoinositide signal transduction pathway and the release of intracellular Ca2+ induced by quisqualate the most potent agonist at mGluR1. The pharmacological nature of these compounds and their relative potencies in antagonizing mGluR1 activation are described.


Subject(s)
Benzoates/pharmacology , Glycine/analogs & derivatives , Neurotoxins/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Aminobutyrates/antagonists & inhibitors , Aminobutyrates/pharmacology , Animals , Binding, Competitive , CHO Cells , Calcium/metabolism , Cells, Cultured , Cricetinae , Cycloleucine/analogs & derivatives , Cycloleucine/antagonists & inhibitors , Cycloleucine/pharmacology , Cytidine Diphosphate Diglycerides/metabolism , Excitatory Amino Acid Antagonists , Glutamates/pharmacology , Glutamic Acid , Glycine/pharmacology , Ibotenic Acid/antagonists & inhibitors , Ibotenic Acid/pharmacology , Kainic Acid/antagonists & inhibitors , Kainic Acid/pharmacology , Neurons/drug effects , Neurons/metabolism , Phosphatidylinositol Diacylglycerol-Lyase , Phosphatidylinositols/metabolism , Phosphoric Diester Hydrolases/metabolism , Quisqualic Acid/antagonists & inhibitors , Quisqualic Acid/pharmacology , Receptors, Metabotropic Glutamate/classification , Recombinant Fusion Proteins/antagonists & inhibitors , Signal Transduction/drug effects , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/antagonists & inhibitors , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
17.
Br J Pharmacol ; 109(3): 779-87, 1993 Jul.
Article in English | MEDLINE | ID: mdl-7689403

ABSTRACT

1 Responses to kainate (KA), willardiine and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) were recorded from rat brain mRNA-injected Xenopus laevis oocytes by use of a two-electrode voltage clamp. 2 Thiocyanate (SCN-; 50 microM-4 mM) ions reversibly and selectively inhibited the membrane current responses to AMPA in a non-competitive manner without affecting KA or willardiine-induced responses. 3 The inhibition of AMPA-induced responses by SCN- was dependent on the SCN- concentration with an estimated IC50 of 1 mM. The antagonism was not dependent on the AMPA concentration. 4 The response to a high concentration of AMPA (100-200 microM) exhibited a peak inward current which declined to a steady-state. SCN- inhibited the steady-state current more than the peak response. The inhibition was unaffected by prior incubation with concanavalin-A (Con-A; 10 microM). 5 Responses to KA were antagonized by AMPA in a competitive manner, suggesting that both agonists may activate a common receptor-channel complex. This interaction between two non-NMDA agonists was not affected by the SCN(-)-induced inhibition of the AMPA response. 6 AMPA-induced responses recorded from large cultured cerebellar neurones by whole-cell recording were also inhibited by SCN- in a non-competitive manner. The AMPA-induced peak current was less affected than the steady-state response. 7 We conclude that SCN- can inhibit the response to AMPA in expressed non-NMDA receptors in Xenopus oocytes and also in native receptors in cultured cerebellar neurones. One possible mechanism of action for SCN- inhibition of responses to AMPA may involve a Con-A-insensitive, non-NMDA receptor-mediated desensitization.


Subject(s)
Brain Chemistry/physiology , Ibotenic Acid/analogs & derivatives , Oocytes/drug effects , RNA, Messenger/pharmacology , Thiocyanates/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cerebellum/cytology , Cerebellum/drug effects , Electrophysiology , Female , Ibotenic Acid/antagonists & inhibitors , Ibotenic Acid/pharmacology , In Vitro Techniques , Kainic Acid/antagonists & inhibitors , Kainic Acid/pharmacology , Microinjections , Neurons/drug effects , Pregnancy , Pyrimidinones , RNA, Messenger/administration & dosage , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/drug effects , Uracil , Xenopus laevis , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid
18.
Neuroreport ; 4(6): 795-8, 1993 Jun.
Article in English | MEDLINE | ID: mdl-7688593

ABSTRACT

We have postulated elsewhere (Shaw CA and Lanius RA. Dev Brain Res 70, 153-161 (1992)) that the kinase/phosphatase regulation of AMPA receptors is mediated by specific ions. Using an in vitro cortical slice preparation we have now examined the roles of calcium (Ca2+), chloride (Cl-), potassium (K+), and sodium (Na+) in the regulation of AMPA receptors. Ca2+ led to a concentration-dependent decrease in [3H]-CNQX binding which could be blocked by a general protein kinase inhibitor (H-7) and a protein kinase A inhibiting peptide. Tamoxifen, a relatively specific protein kinase C inhibitor, had no effect. In contrast, Cl- led to concentration-dependent increases in [3H]-CNQX binding which could be blocked by both sodium-ortho-vanadate, a tyrosine residue selective phosphatase inhibitor, and sodium-beta-D-glycerol phosphate, a serine residue selective phosphatase blocker. K+ and Na+ had no effect on [3H]-CNQX binding. These results suggest that Ca2+ and Cl- may be acting as signals which trigger kinase(s) and phosphatase(s) involved in the regulation of AMPA receptors.


Subject(s)
Phosphoric Monoester Hydrolases/metabolism , Protein Kinases/metabolism , Receptors, Glutamate/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine , 6-Cyano-7-nitroquinoxaline-2,3-dione , Animals , Calcium/metabolism , Chlorides/metabolism , Glycerophosphates/pharmacology , Ibotenic Acid/analogs & derivatives , Ibotenic Acid/antagonists & inhibitors , In Vitro Techniques , Isoquinolines/pharmacology , Peptides/pharmacology , Piperazines/pharmacology , Potassium/metabolism , Protein Kinase Inhibitors , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, AMPA , Sodium/metabolism , Tamoxifen/pharmacology , Vanadates/pharmacology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid
19.
Brain Res ; 614(1-2): 10-4, 1993 Jun 18.
Article in English | MEDLINE | ID: mdl-7688644

ABSTRACT

The effects of kainate (KA) on the recovery of neuronal function in rat hippocampal slices after hypoxia or glucose deprivation (GD) were investigated and compared to those of (R,S)-alpha-amino-3-hydroxy-5-methyl-4- isoxazoleproprionate (AMPA). KA and AMPA were found to be more toxic than either N-methyl-D-aspartate (NMDA), quinolinate, or glutamate, both under normal conditions and under states of energy deprivation. Doses as low as 1 microM KA or AMPA were sufficient to significantly reduce the recovery rate of neuronal function in slices after a standardized period of hypoxia or GD. The enhancement of hypoxic neuronal damage by both agonists could be partially blocked by the antagonist kynurenate, by the NMDA competitive antagonist AP5, and by elevating [Mg2+] in or by omitting Ca2+ from the perfusion medium. The AMPA antagonist glutamic acid diethyl ester was ineffective in preventing the enhanced hypoxic neuronal damage by either KA or AMPA. The antagonist of the glycine modulatory site on the NMDA receptor, 7-chlorokynurenate, did not block the KA toxicity but was able to block the toxicity of AMPA. 2,3-Dihydroxyquinoxaline completely blocked the KA- and AMPA-enhanced hypoxic neuronal damage. The KA-enhanced, GD-induced neuronal damage was prevented by Ca2+ depletion and partially antagonized by kynurenate but not by AP5 or elevated [Mg2+]. The results of the present study indicate that the KA receptor is involved in the mechanism of neuronal damage induced by hypoxia and GD, probably allowing Ca2+ influx and subsequent intracellular Ca2+ overload.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Energy Metabolism/physiology , Glucose/deficiency , Hippocampus/drug effects , Hypoxia, Brain/metabolism , Kainic Acid/toxicity , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Calcium/deficiency , Electrophysiology , Evoked Potentials/drug effects , Hippocampus/metabolism , Ibotenic Acid/analogs & derivatives , Ibotenic Acid/antagonists & inhibitors , Ibotenic Acid/toxicity , In Vitro Techniques , Kainic Acid/antagonists & inhibitors , Magnesium/pharmacology , Male , N-Methylaspartate/antagonists & inhibitors , N-Methylaspartate/physiology , N-Methylaspartate/toxicity , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/drug effects , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid
20.
Neurosci Lett ; 156(1-2): 78-82, 1993 Jun 25.
Article in English | MEDLINE | ID: mdl-8414194

ABSTRACT

We have previously demonstrated that ibotenate (IBO) injected into the pedunculopontine tegmental nucleus (PPTg) damages all neurones there while quinolinate (QUIN) makes relatively selective lesions of cholinergic neurones. We now compare the effects of two anaesthetics, sodium pentobarbitone and Avertin (tribromoethanol/tert-amylalcohol dissolved in ethanol, saline and phosphate buffer) on three doses of IBO and QUIN in the PPTg. Diaphorase-positive cell loss after QUIN was attenuated under barbiturate, the relative selectivity of QUIN for diaphorase-positive neurones was lost and lesion volumes were uniformly small compared with lesions made under Avertin anaesthesia. IBO toxicity was unaffected by anaesthesia. These data are discussed with reference to the actions of excitotoxins at glutamate receptor subtypes and interactions of barbiturates with the GABAA receptor.


Subject(s)
Anesthetics/pharmacology , Ethanol/analogs & derivatives , Ibotenic Acid/toxicity , Neurons/pathology , Neurotoxins/toxicity , Pentobarbital/pharmacology , Pons/pathology , Quinolinic Acid/toxicity , Anesthesia, General , Animals , Dose-Response Relationship, Drug , Ethanol/pharmacology , Ibotenic Acid/antagonists & inhibitors , Male , Neurons/drug effects , Neurotoxins/antagonists & inhibitors , Pons/drug effects , Quinolinic Acid/antagonists & inhibitors , Rats , Rats, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL
...