Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Lupus ; 21(14): 1575-81, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23161578

ABSTRACT

Catatonia is a rare manifestation in patients with systemic lupus erythematosus (SLE). As catatonia can be associated with both psychiatric and organic conditions, this could create a diagnostic dilemma once this occurs in SLE patients. The report describes a 15-year-old female with SLE who developed catatonia three days after the diagnosis of SLE was made. Her catatonia was refractory to the treatment with immunosuppressive therapy, which included pulse methylprednisolone, intravenous cyclophosphamide, rituximab, intravenous immunoglobulin (IVIG) and plasmapheresis. Given her persistent catatonia, electroconvulsive therapy (ECT) was initiated three months after the onset of her symptoms. After the third ECT treatment, her mental status dramatically improved and returned nearly to baseline while she was continued on the immunosuppression. This is the first report of a successful ECT therapy in catatonic lupus in children.


Subject(s)
Catatonia/therapy , Electroconvulsive Therapy/methods , Lupus Erythematosus, Systemic/complications , Lupus Vasculitis, Central Nervous System/therapy , Adolescent , Age of Onset , Catatonia/etiology , Female , Humans , Immunosuppressive Agents/therapeutic use , Treatment Outcome
2.
Cell Death Differ ; 15(10): 1582-92, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18600230

ABSTRACT

There has been a growing controversy regarding the continued use of glucocorticoid therapy to treat respiratory dysfunction associated with prematurity, as mounting clinical evidence has shown neonatal exposure produces permanent neuromotor and cognitive deficits. Here we report that, during a selective neonatal window of vulnerability, a single glucocorticoid injection in the mouse produces rapid and selective apoptotic cell death of the proliferating neural progenitor cells in the cerebellar external granule layer and permanent reductions in neuronal cell counts of their progeny, the cerebellar internal granule layer neurons. Our estimates suggest that this mouse window of vulnerability would correspond in the human to a period extending from approximately 20 weeks gestation to 6.5 weeks after birth. This death pathway is critically regulated by the proapoptotic Bcl-2 family member Puma and is independent of p53 expression. These rodent data indicate that there exists a previously unknown window of vulnerability during which a single glucocorticoid exposure at clinically relevant doses can produce neural progenitor cell apoptosis and permanent cerebellar pathology that may be responsible for some of the iatrogenically induced neurodevelopmental abnormalities seen in children exposed to this drug. This vulnerability may be related to the physiological role of glucocorticoids in regulating programmed cell death in the mammalian cerebellum.


Subject(s)
Apoptosis/drug effects , Cerebellum/growth & development , Glucocorticoids/pharmacology , Neurons/drug effects , Neurons/physiology , Stem Cells/drug effects , Stem Cells/physiology , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins , Behavior, Animal/physiology , Cerebellum/cytology , Child , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Neurons/cytology , Signal Transduction/physiology , Stem Cells/cytology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
3.
Mol Psychiatry ; 7(7): 726-33, 2002.
Article in English | MEDLINE | ID: mdl-12192617

ABSTRACT

N-methyl-D-aspartate (NMDA) glutamate receptor antagonists are used in clinical anesthesia and are being developed as therapeutic agents for preventing neurodegeneration in stroke, epilepsy, and brain trauma. However, the ability of these agents to produce neurotoxicity in adult rats and psychosis in adult humans compromises their clinical usefulness. In addition, an NMDA receptor hypofunction (NRHypo) state might play a role in neurodegenerative and psychotic disorders, like Alzheimer's disease, bipolar disorder and schizophrenia. Thus, developing pharmacological means of preventing these NRHypo-induced effects could have significant clinically relevant benefits. NRHypo neurotoxicity appears to be mediated by a complex disinhibition mechanism that results in the excessive stimulation of certain vulnerable neurons. Here we report our findings that five agents (phenytoin, carbamazepine, valproic acid, lamotrigine, and riluzole), thought to possess anticonvulsant activity because they inhibit voltage-gated sodium channels, prevent NRHypo neurotoxicity. The ability of tetrodotoxin, a highly selective inhibitor of voltage-gated sodium channels, to prevent the same neurotoxicity suggests that inhibition of this ion channel is the likely mechanism of action of these five agents. We also found that three other anticonvulsants (felbamate, gabapentin and ethosuximide), whose mechanism is less clear, also prevent NRHypo neurotoxicity, suggesting that inhibition of voltage-gated sodium channels is not the only mechanism via which anticonvulsants can act to prevent NRHypo neurotoxicity. Several of these agents have been found to be of clinical use in bipolar disorder. It would be of interest to determine whether these agents might have therapeutic benefits for conditions in which a NRHypo state may exist.


Subject(s)
Anticonvulsants/pharmacology , Dizocilpine Maleate/toxicity , Excitatory Amino Acid Antagonists/toxicity , Phenytoin/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Sodium Channels/physiology , Animals , Carbamazepine/pharmacology , Drug Interactions , Female , Ion Channel Gating/physiology , Lamotrigine , Neurotransmitter Agents/metabolism , Rats , Rats, Sprague-Dawley , Triazines/pharmacology , Valproic Acid/pharmacology
4.
Mol Psychiatry ; 7(1): 32-43, 2002.
Article in English | MEDLINE | ID: mdl-11803444

ABSTRACT

NMDA glutamate receptor antagonists are used in clinical anesthesia, and are being developed as therapeutic agents for preventing neurodegeneration in stroke, epilepsy, and brain trauma. However, the ability of these agents to produce neurotoxicity in adult rats and psychosis in adult humans compromises their clinical usefulness. In addition, an NMDA receptor hypofunction (NRHypo) state might play a role in neurodegenerative and psychotic disorders, like Alzheimer's disease and schizophrenia. Thus, understanding the mechanism underlying NRHypo-induced neurotoxicity and psychosis could have significant clinically relevant benefits. NRHypo neurotoxicity can be prevented by several classes of agents (e.g. antimuscarinics, non-NMDA glutamate antagonists, and alpha(2) adrenergic agonists) suggesting that the mechanism of neurotoxicity is complex. In the present study a series of experiments was undertaken to more definitively define the receptors and complex neural circuitry underlying NRHypo neurotoxicity. Injection of either the muscarinic antagonist scopolamine or the non-NMDA antagonist NBQX directly into the cortex prevented NRHypo neurotoxicity. Clonidine, an alpha(2) adrenergic agonist, protected against the neurotoxicity when injected into the basal forebrain. The combined injection of muscarinic and non-NMDA Glu agonists reproduced the neurotoxic reaction. Based on these and other results, we conclude that the mechanism is indirect, and involves a complex network disturbance, whereby blockade of NMDA receptors on inhibitory neurons in multiple subcortical brain regions, disinhibits glutamatergic and cholinergic projections to the cerebral cortex. Simultaneous excitotoxic stimulation of muscarinic (m(3)) and glutamate (AMPA/kainate) receptors on cerebrocortical neurons appears to be the proximal mechanism by which the neurotoxic and psychotomimetic effects of NRHypo are mediated.


Subject(s)
Cerebral Cortex/drug effects , Excitatory Amino Acid Antagonists/toxicity , Nerve Tissue Proteins/antagonists & inhibitors , Phenazocine/analogs & derivatives , Quinoxalines/toxicity , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Adrenergic alpha-Agonists/administration & dosage , Adrenergic alpha-Agonists/therapeutic use , Animals , Carbachol/administration & dosage , Carbachol/toxicity , Carbazoles/pharmacology , Cerebral Cortex/ultrastructure , Clonidine/administration & dosage , Clonidine/therapeutic use , Dizocilpine Maleate/administration & dosage , Dizocilpine Maleate/pharmacology , Drug Interactions , Excitatory Amino Acid Antagonists/administration & dosage , Female , Kainic Acid/administration & dosage , Kainic Acid/toxicity , Models, Neurological , Muscarinic Antagonists/administration & dosage , Muscarinic Antagonists/toxicity , Nerve Tissue Proteins/physiology , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Phenazocine/administration & dosage , Phenazocine/toxicity , Prosencephalon/drug effects , Prosencephalon/physiology , Quinoxalines/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/drug effects , Receptors, Glutamate/physiology , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Receptors, sigma/drug effects , Receptors, sigma/physiology , Scopolamine/administration & dosage , Scopolamine/therapeutic use , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/administration & dosage , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/toxicity
5.
Arch Gen Psychiatry ; 57(12): 1165-73, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11115331

ABSTRACT

BACKGROUND: Psychosis is common in patients with Alzheimer disease. While the relationship between psychosis and clinical variables has been examined frequently, few studies have examined the relationship between psychosis and the 2 major neuropathological hallmarks of Alzheimer disease: neurofibrillary tangles and senile plaques. We characterized the occurrence of psychosis in relation to dementia severity and determined if subjects with Alzheimer disease and psychosis had a greater neurofibrillary tangle or senile plaque burden than subjects with Alzheimer disease and no psychosis. METHODS: One hundred nine subjects with Alzheimer disease were followed longitudinally with semistructured assessments in order to assign a Clinical Dementia Rating and determine whether psychosis was present. After the subjects died, their brains were obtained for histological examination. Analysis of variance was used to compare the densities of neurofibrillary tangles, total senile plaques, and cored senile plaques in subjects with psychosis vs subjects without psychosis, in several neocortical regions, the hippocampus, and the entorhinal cortex. RESULTS: Psychosis occurred commonly in Alzheimer disease, affecting 63% of subjects. The frequency of psychosis increased with increasing dementia severity. More importantly, we found that subjects with psychosis had a 2.3-fold (95% confidence interval, 1.2-3.9) greater density of neocortical neurofibrillary tangles than did subjects without psychosis. The increase was independent of dementia severity. No similar relationship with psychosis was seen for total senile plaques or cored senile plaques. CONCLUSIONS: The increase in psychosis frequency that occurs with the progression of dementia severity and the independent association between psychosis and neurofibrillary tangle density suggest the possibility that some common underlying process or processes specific to Alzheimer disease may regulate both phenomena. Arch Gen Psychiatry. 2000;57:1165-1173.


Subject(s)
Alzheimer Disease/epidemiology , Alzheimer Disease/pathology , Neocortex/pathology , Neurofibrillary Tangles/pathology , Psychotic Disorders/epidemiology , Psychotic Disorders/pathology , Aged , Alzheimer Disease/diagnosis , Comorbidity , Delusions/diagnosis , Delusions/epidemiology , Delusions/pathology , Entorhinal Cortex/pathology , Female , Hallucinations/diagnosis , Hallucinations/epidemiology , Hallucinations/pathology , Hippocampus/pathology , Humans , Longitudinal Studies , Male , Plaque, Amyloid/pathology , Psychiatric Status Rating Scales/statistics & numerical data , Psychotic Disorders/diagnosis , Severity of Illness Index
6.
Environ Health Perspect ; 108 Suppl 3: 383-8, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10852832

ABSTRACT

We review recent findings pertaining to several environmental agents (ethanol, phencyclidine, ketamine, nitrous oxide, barbiturates, benzodiazepines, halothane, isoflurane, and propofol) that have the potential to delete large numbers of neurons from the developing brain by a newly discovered mechanism involving interference in the action of neurotransmitters [glutamate and gamma-amino butyric acid (GABA) at (italic)N(/italic)-methyl-d-aspartate (NMDA)] and GABA(subscript)A(/subscript) receptors during the synaptogenesis period, also known as the brain growth-spurt period. Transient interference (lasting >= 4 hr) in the activity of these transmitters during the synaptogenesis period (the last trimester of pregnancy and the first several years after birth in humans) causes millions of developing neurons to commit suicide (die by apoptosis). Many of these agents are drugs of abuse (ethanol is a prime example) to which the human fetal brain may be exposed during the third trimester by drug-abusing mothers. Ethanol triggers massive apoptotic neurodegeneration in the developing brain by interfering with both the NMDA and GABA(subscript)A(/subscript) receptor systems, and this can explain the reduced brain mass and lifelong neurobehavioral disturbances associated with intrauterine exposure of the human fetus to ethanol (fetal alcohol syndrome). Exposure of the immature brain in a medical treatment context is also of concern because many of these agents are drugs used frequently as sedatives, tranquilizers, anticonvulsants, or anesthetics in pediatric and/or obstetrical medicine. Because this is a newly discovered mechanism, further research will be required to fully ascertain the nature and degree of risk posed by exposure of the developing human brain to environmental agents that act by this mechanism.


Subject(s)
Apoptosis , Brain/drug effects , Brain/embryology , Environmental Health , Xenobiotics/adverse effects , Embryonic and Fetal Development , Female , Humans , Neurons/drug effects , Neurons/pathology , Pregnancy , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology , Substance-Related Disorders
7.
Dialogues Clin Neurosci ; 2(3): 219-32, 2000 Sep.
Article in English | MEDLINE | ID: mdl-22034391

ABSTRACT

An increasing level of N-methyl-D-aspartate (NMDA) receptor hypofunction within the brain is associated with memory and learning impairments, with psychosis, and ultimately with excitotoxic brain injury. As the brain ages, the NMDA receptor system becomes progressively hypofunctional, contributing to decreases in memory and learning performance. In those individuals destined to develop Alzheimer's disease, other abnormalities (eg, amyloidopathy and oxidative stress) interact to increase the NMDA receptor hypofunction (NRHypo) burden. In these vulnerable individuals, the brain then enters into a severe and persistent NRHypo state, which can lead to widespread neurodegeneration with accompanying mental symptoms and further cognitive deterioration. If the hypotheses described herein prove correct, treatment implications may be considerable. Pharmacological methods for preventing the overstimulation of vulnerable corticolimbic pyramidal neurons developed in an animal model may be applicable to the prevention and treatment of Alzheimer's disease.

8.
Mol Psychiatry ; 4(4): 344-52, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10483051

ABSTRACT

N-methyl-D-aspartate (NMDA) glutamate (Glu) receptor antagonists (eg MK-801, ketamine, phencyclidine [PCP]) injure cerebrocortical neurons in the posterior cingulate and retrosplenial cortex (PC/RSC). We have proposed that the neurotoxic action of these agents is mediated in part by a complex polysynaptic mechanism involving an interference in GABAergic inhibition resulting in excessive release of acetylcholine (ACh). Previously we have found that the systemic injection of GABAergic agents and alpha2-adrenergic agonists can block this neurotoxicity. In the present study we tested the hypothesis that NMDA antagonists trigger release of ACh in PC/RSC and that this action of NMDA antagonists is suppressed by GABAergic agents or alpha2-adrenergic agonists. The effect of MK-801 and ketamine on PC/RSC ACh output (and the ability of pentobarbital, diazepam and clonidine to modify MK-801-induced ACh release) was studied in adult female rats using in vivo microdialysis. Both MK-801 and ketamine caused a significant rise in PC/RSC ACh output compared to basal levels. Pentobarbital, diazepam and clonidine suppressed MK-801's effect on ACh release. Exploratory studies indicated that the site of action of these agents was outside of the PC/RSC. The microdialysis results are consistent with several aspects of the circuitry proposed to mediate the neurotoxic action of NMDA antagonists.


Subject(s)
Acetylcholine/metabolism , Adrenergic alpha-Agonists/pharmacology , Cerebral Cortex/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Receptors, Adrenergic, alpha-2/physiology , Receptors, GABA/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Cerebral Cortex/drug effects , Clonidine/pharmacology , Diazepam/pharmacology , Dizocilpine Maleate/pharmacology , Female , Ketamine/pharmacology , Microdialysis , Pentobarbital/pharmacology , Rats , Rats, Sprague-Dawley
9.
Neuropsychopharmacology ; 20(2): 106-18, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9885791

ABSTRACT

N-methyl-D-aspartate (NMDA) glutamate receptor antagonists are reported to induce schizophrenia-like symptoms in humans, including cognitive impairments. Shortcomings of most previous investigations include failure to maintain steady-state infusion conditions, test multiple doses and/or measure antagonist plasma concentrations. This double-blind, placebo-controlled, randomized, within-subjects comparison of three fixed subanesthetic, steady-state doses of intravenous ketamine in healthy males (n = 15) demonstrated dose-dependent increases in Brief Psychiatric Rating Scale positive (F[3,42] = 21.84; p < 0.0001) and negative symptoms (F[3,42] = 2.89; p = 0.047), and Scale for the Assessment of Negative Symptoms (SANS) total scores (F[3,42] = 10.55; p < 0.0001). Ketamine also produced a robust dose-dependent decrease in verbal declarative memory performance (F[3,41] = 5.11; p = 0.004), and preliminary evidence for a similar dose-dependent decrease in nonverbal declarative memory, occurring at or below plasma concentrations producing other symptoms. Increasing NMDA receptor hypofunction is associated with early occurring memory impairments followed by other schizophrenia-like symptoms.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Memory Disorders/chemically induced , Psychoses, Substance-Induced/psychology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Adult , Animals , Cognition/drug effects , Depression, Chemical , Dose-Response Relationship, Drug , Double-Blind Method , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/blood , Female , Humans , Injections, Intravenous , Ketamine/administration & dosage , Ketamine/blood , Male , Memory Disorders/psychology , Mental Recall/drug effects , Rats
11.
J Psychiatr Res ; 33(6): 523-33, 1999.
Article in English | MEDLINE | ID: mdl-10628529

ABSTRACT

Several decades of research attempting to explain schizophrenia in terms of the dopamine hyperactivity hypothesis have produced disappointing results. A new hypothesis focusing on hypofunction of the NMDA glutamate transmitter system is emerging as a potentially more promising concept. In this article, we present a version of the NMDA receptor hypofunction hypothesis that has evolved from our recent studies pertaining to the neurotoxic and psychotomimetic effects of PCP and related NMDA antagonist drugs. In this article, we examine this hypothesis in terms of its strengths and weaknesses, its therapeutic implications and ways in which it can be further tested.


Subject(s)
Models, Biological , Receptors, N-Methyl-D-Aspartate/physiology , Schizophrenia/etiology , Animals , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Humans , Models, Psychological , Phencyclidine/pharmacology , Rats , Schizophrenia/physiopathology , Schizophrenia/therapy
12.
Neuropsychopharmacology ; 18(1): 57-62, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9408919

ABSTRACT

Phencyclidine, ketamine, and other agents that block NMDA glutamate receptors trigger a schizophrenia-like psychosis in humans and induce pathomorphological changes in cerebrocortical neurons in rat brain. Accumulating evidence suggests that a complex network disturbance involving multiple transmitter receptor systems is responsible for the neuronal injury, and it is proposed that a similar network disturbance is responsible for the psychotomimetic effects of NMDA antagonists, and might also be involved in the pathophysiology of schizophrenia. In the present study we present evidence that serotonergic agents possessing 5HT2A agonist activity prevent NMDA antagonist neurotoxicity in rat brain. It is proposed that 5HT2A agonists may also prevent the psychotomimetic effects of NMDA antagonists. Among the 5HT2A agonists examined and found to be neuroprotective are LSD and related hallucinogens. The apparent contradiction in proposing that these agents might have antipsychotic properties is resolved by evidence linking their hallucinogenic activity to agonist action at 5HT2C receptors, whereas antipsychotic activity would be attributable to agonist action at 5HT2A receptors.


Subject(s)
Excitatory Amino Acid Antagonists/toxicity , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Serotonin Receptor Agonists/pharmacology , Amphetamines/antagonists & inhibitors , Amphetamines/toxicity , Animals , Dizocilpine Maleate/antagonists & inhibitors , Dizocilpine Maleate/toxicity , Dose-Response Relationship, Drug , Female , Rats , Rats, Sprague-Dawley , Ritanserin/pharmacology , Serotonin Antagonists/pharmacology
13.
Restor Neurol Neurosci ; 13(1-2): 75-83, 1998.
Article in English | MEDLINE | ID: mdl-12671289

ABSTRACT

In this article we review the hypothesis that impaired function of the N-methyl-Daspartate (NMDA) glutamate receptor system may be an important mechanism for understanding the pathophysiology of Alzheimer's disease (AD). We propose a two stage process, the first involving amyloidopathy, oxidative stress and/or energy metabolic disturbances promoting neuronal sensitivity to glutamate-induced excitotoxic injury to an extent that even normal amounts of Glu become excitotoxic. As a consequence, NMDA receptor-bearing neurons (and their NMDA receptors) are deleted from critical corticolimbic brain circuits, which leaves these circuits in an NMDA receptor hypofunctional (NRHypo) state. In the second stage this NRHypo state results in the disinhibition of a complicated neural circuitry that leads to widespread neurodegeneration in corticolimbic areas, consquent neurofibrillary tangle formation and cognitive decline. We propose that certain pharmacological methodes which have been found to protect against NRHypo-induced neurodegeneration in animal brain might be useful treatments for AD.

14.
Prog Brain Res ; 116: 421-37, 1998.
Article in English | MEDLINE | ID: mdl-9932393

ABSTRACT

Here we have described a novel excitotoxic process in which hypofunctional NMDA receptors cease driving GABA ergic neurons which cease inhibiting excitatory transmitters in the brain. These disinhibited excitatory transmitters then act in concert to slowly hyperstimulate neurons in corticolimbic brain regions. We have discussed how such an abnormality could exist in the brains of individuals with schizophrenia or AD and could account for the clinical stigmata of the two disorders. In addition, we have highlighted how other disorder-specific factors would account for the differences in the clinical presentation of AD and schizophrenia. In an animal model, pharmacological methods have been developed for preventing the overstimulation of these vulnerable corticolimbic pyramidal neurons and at least some of these methods may be applicable for treating AD and schizophrenia.


Subject(s)
Alzheimer Disease/physiopathology , Glutamic Acid/physiology , Mental Disorders/physiopathology , Schizophrenia/physiopathology , Synapses/physiology , Animals , Humans
15.
Arch Neurol ; 54(10): 1234-40, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9341569

ABSTRACT

Excessive activation of N-methyl D-aspartate (NMDA) receptors by endogenous glutamate (Glu) causes excitotoxic neuronal degeneration in acute central nervous system injury syndromes such as stroke and trauma. Early attempts to link NMDA receptor hyperactivity (NRHyper) to Alzheimer disease (AD) were stymied by evidence in 3 separate species (mice, rats, and monkeys) that, with advancing age, the NMDA receptor system becomes markedly hypoactive. While this would seem to argue against a role for NMDA receptors in AD, we have recently found in animal studies that, when the NMDA receptor system is rendered markedly hypoactive, a disinhibition syndrome is triggered in which low-grade chronic excitotoxic activity (fueled by acetylcholine and Glu) is unleashed that can cause a widespread pattern of neuronal degeneration resembling that seen in AD. Therefore, we postulate that NMDA receptor hypoactivity (NRHypo) associated with advancing age may have an important contributory role in AD and that the main difference between the aging AD brain and the aging "normal" brain is that a heavier burden of certain adjunctive risk factors may be present in the AD brain that promote the NRHypo state and increase the likelihood that widespread neurodegeneration will occur.


Subject(s)
Alzheimer Disease/physiopathology , Nerve Degeneration/physiopathology , Neurotoxins/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Animals , Humans , Medical Illustration , Parasympathetic Nervous System/physiopathology , Rats , Receptors, N-Methyl-D-Aspartate/physiology
16.
Brain Res ; 752(1-2): 1-14, 1997 Mar 28.
Article in English | MEDLINE | ID: mdl-9106435

ABSTRACT

Phencyclidine and other antagonists of the N-methyl-D-aspartate subtype of glutamate receptor cause psychosis in humans. In low doses these agents induce a reversible neurotoxic reaction in the rat brain that is limited to the retrosplenial granular cortex. Some investigators have reported that phencyclidine at higher doses or by more prolonged treatment causes a more disseminated pattern of damage. However, it has not been clearly demonstrated whether the disseminated damage is reversible or irreversible and whether it is consistently reproducible, nor is it known how many and which neurons are at risk. In the present study we addressed these questions using several histological approaches (plastic-embedded thin sections for light microscopy and ultrathin plastic sections for electron microscopy, paraffin-embedded haematoxylin and eosin sections, 72 kDa heat shock protein immunocytochemistry and de Olmos silver impregnation) to study the lesions induced in rat brain by phencyclidine (alone or when augmented with pilocarpine). We found that phencyclidine can kill a relatively large number of neurons distributed over many cerebrocortical and limbic brain regions, but the multifocal pattern of damage occurred in only a small percentage of treated rats. The addition of a low dose of pilocarpine to phencyclidine caused the widespread pattern of damage to manifest on a much more consistent basis. Available evidence suggests that disinhibition of multiple converging excitatory pathways is the mechanism by which phencyclidine triggers widespread neuronal degeneration; however, the specific combination of excitatory inputs that contributes to the pathological process may differ from region to region.


Subject(s)
Brain Damage, Chronic/chemically induced , Phencyclidine , Pilocarpine , Animals , Brain/pathology , Brain Damage, Chronic/pathology , Dose-Response Relationship, Drug , Drug Synergism , Female , Nerve Degeneration , Phencyclidine/administration & dosage , Pilocarpine/administration & dosage , Rats , Rats, Sprague-Dawley
17.
Schizophr Bull ; 23(3): 533-6, 1997.
Article in English | MEDLINE | ID: mdl-9327519

ABSTRACT

Olney and Farber present their work with N-methyl-D-aspartate (NMDA) antagonists, which are psychotogens, and propose that the structural changes described by Bogerts could be accounted for by a two-stage process. The first stage of the process would occur early in life and would culminate in the selective loss of NMDA-receptor bearing gamma-aminobutyric acid (GABA)ergic neurons and thus render the brain into a NMDA receptor hypofunctional (NRH) state. Such a loss would set the foundation for the second stage in which the neural circuits that have been altered by the loss of these GABAergic interneurons would become activated in late adolescence but would be dysfunctional. Dysfunction of this circuit would lead to the psychopathology of schizophrenia and potentially, if severe enough, to neuronal degeneration. Thus, the changes described by Bogerts could originate partially in early life and partially in adulthood. Based on their animal model, the authors suggest studies that should be carried out in humans.


Subject(s)
Limbic System/physiopathology , Schizophrenia, Paranoid/physiopathology , Temporal Lobe/physiopathology , Adolescent , Adult , Animals , Brain Mapping , Humans , Limbic System/pathology , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Nerve Net/pathology , Nerve Net/physiopathology , Receptors, N-Methyl-D-Aspartate/physiology , Schizophrenia, Paranoid/pathology , Schizophrenia, Paranoid/psychology , Synaptic Transmission/physiology , Temporal Lobe/pathology , gamma-Aminobutyric Acid/physiology
18.
J Neuropathol Exp Neurol ; 55(11): 1115-23, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8939194

ABSTRACT

In the past two decades brain tumor rates have risen in several industrialized countries, including the United States. During this time, brain tumor data have been gathered by the National Cancer Institute from catchment areas representing 10% of the United States population. In the present study, we analyzed these data from 1975 to 1992 and found that the brain tumor increases in the United States occurred in two distinct phases, an early modest increase that may primarily reflect improved diagnostic technology, and a more recent sustained increase in the incidence and shift toward greater malignancy that must be explained by some other factor(s). Compared to other environmental factors putatively linked to brain tumors, the artificial sweetener aspartame is a promising candidate to explain the recent increase in incidence and degree of malignancy of brain tumors. Evidence potentially implicating aspartame includes an early animal study revealing an exceedingly high incidence of brain tumors in aspartame-fed rats compared to no brain tumors in concurrent controls, the recent finding that the aspartame molecule has mutagenic potential, and the close temporal association (aspartame was introduced into US food and beverage markets several years prior to the sharp increase in brain tumor incidence and malignancy). We conclude that there is need for reassessing the carcinogenic potential of aspartame.


Subject(s)
Aspartame/adverse effects , Brain Neoplasms/chemically induced , Brain Neoplasms/epidemiology , Sweetening Agents/adverse effects , Animals , Brain Neoplasms/pathology , Humans , Incidence , Mortality , United States
19.
Schizophr Res ; 21(1): 33-7, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8998274

ABSTRACT

Antagonists of the N-methyl-D-Aspartate (NMDA) subtype of glutamate receptor (e.g., phencyclidine, ketamine, MK-801) cause a schizophrenia-like psychosis in humans and neurotoxicity in the adult rat brain. We report here that clozapine and structurally related agents (olanzapine, fluperlapine, loxapine, amoxapine) can prevent NMDA antagonist neurotoxicity in the rat with a rank order corresponding to their ability to mimic the antipsychotic properties of clozapine.


Subject(s)
Antipsychotic Agents/pharmacology , Dibenzazepines/pharmacology , Dizocilpine Maleate/antagonists & inhibitors , Pirenzepine/analogs & derivatives , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Benzodiazepines , Clozapine/pharmacology , Dibenzoxazepines/pharmacology , Dizocilpine Maleate/adverse effects , Female , N-Methylaspartate/antagonists & inhibitors , Nervous System/drug effects , Olanzapine , Pirenzepine/pharmacology , Rats , Rats, Sprague-Dawley
20.
Arch Gen Psychiatry ; 52(12): 998-1007, 1995 Dec.
Article in English | MEDLINE | ID: mdl-7492260

ABSTRACT

In this article, we advance a unified hypothesis pertaining to combined dysfunction of dopamine and N-methyl-D-aspartate glutamate receptors that highlights N-methyl-D-aspartate receptor hypofunction as a key mechanism that can help explain major clinical and pathophysiological aspects of schizophrenia. The following fundamental features of schizophrenia are accommodated by this hypothesis: (1) the occurrence of structural brain changes during early development that have the potential for producing subsequent clinical manifestations of schizophrenia, (2) a quiescent period in infancy and adolescence before clinical manifestations are expressed, (3) onset in early adulthood of psychotic symptoms, (4) involvement of dopamine (D2) receptors in some cases but not others that would explain why some but not all patients are responsive to typical neuroleptic therapy, and (5) ongoing neurodegenerative changes and cognitive deterioration in some patients. We propose that since N-methyl-D-aspartate receptor hypofunction can cause psychosis in humans and corticolimbic neurodegenerative changes in the rat brain, and since these changes are prevented by certain antipsychotic drugs, including atypical neuroleptic agents (clozapine, olanzapine, fluperlapine), a better understanding of the N-methyl-D-aspartate receptor hypofunction mechanism and ways of preventing its neurodegenerative consequences in the rat brain may lead to improved pharmacotherapy in schizophrenia.


Subject(s)
Brain/pathology , Gyrus Cinguli/ultrastructure , Receptors, Glutamate/metabolism , Schizophrenia/metabolism , Animals , Microscopy, Electron , Models, Neurological , Pyramidal Cells/ultrastructure , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia/pathology
SELECTION OF CITATIONS
SEARCH DETAIL