Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
Add more filters










Publication year range
1.
J Neuroimmunol ; 229(1-2): 146-56, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20724006

ABSTRACT

There are currently four known isoforms of nitric oxide synthase (NOS). Of these, neuronal NOS (nNOS) is known to be present exclusively in neurons, endothelial NOS (eNOS) in vascular endothelium, while the inducible form of NOS (iNOS) is known to be activated in oligodendrocytes, astrocytes and microglia. The fourth isoform, mitochondrial NOS (mtNOS), represents a post-translational modification of nNOS. Using western blotting and real time-PCR, we show induction and activation of nNOS following culture of oligodendrocyte progenitor cells (OPC) with lipopolysaccharide (LPS). Activation of nNOS results in accumulation of peroxynitrite and tyrosine nitration of proteins in oligodendrocytes resulting in reduced cell viability. Injection of LPS in vivo into the corpus callosum of rats leads to the development of extensive demyelination of the white matter tracts. Immunostaining of regions close to the injection site shows the presence of nNOS, but not iNOS, in oligodendrocytes. Neither iNOS nor nNOS was seen in astrocytes in areas of demyelination. These studies suggest that activation of nNOS in oligodendrocytes leads to oligodendrocyte injury resulting in demyelination.


Subject(s)
Gene Expression Regulation, Enzymologic/drug effects , Lipopolysaccharides/pharmacology , Nitric Oxide Synthase Type I/metabolism , Oligodendroglia/drug effects , Stem Cells/drug effects , 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism , Analysis of Variance , Animals , Animals, Newborn , Arginine/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/cytology , Cell Survival/drug effects , Cells, Cultured , Citrulline/metabolism , Corpus Callosum/drug effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Indazoles/pharmacology , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type II/metabolism , O Antigens/metabolism , Peroxynitrous Acid/metabolism , Rats , Rats, Sprague-Dawley , Tritium/metabolism
2.
Exp Neurol ; 169(2): 340-50, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11358447

ABSTRACT

Mitochondrial defects, which occur in the brain of late-stage Huntington's disease (HD) patients, have been proposed to underlie the selective neuronal loss in the disease. To shed light on the possible role of mitochondrial energy impairment in the early phases of HD pathophysiology, we carried out Golgi impregnation and quantitative histochemical/biochemical studies in HD full-length cDNA transgenic mice that were symptomatic but had not developed to a stage in which neuronal loss could be documented. Golgi staining showed morphologic abnormalities that included a significant decrease in the number of dendritic spines and a thickening of proximal dendrites in striatal and cortical neurons. In contrast, measurements of mitochondrial electron transport Complexes I-IV did not reveal changes in the striatum and cerebral cortex in these mice. Examination of the neostriatum and cerebral cortex in human presymptomatic and pathological Grade 1 HD cases also showed no change in the activity of mitochondrial Complexes I-IV. These data suggest that dendritic alterations precede irreversible cell loss in HD, and that mitochondrial energy impairment is a consequence, rather than a cause, of early neuropathological changes.


Subject(s)
Corpus Striatum/pathology , Dendrites/pathology , Huntington Disease/genetics , Mitochondria/metabolism , Nerve Degeneration/genetics , Nerve Tissue Proteins/genetics , Neurons/pathology , Nuclear Proteins/genetics , Somatosensory Cortex/metabolism , Animals , Caudate Nucleus/pathology , Coloring Agents , DNA, Complementary , Dendrites/ultrastructure , Energy Metabolism , Functional Laterality , Golgi Apparatus/pathology , Golgi Apparatus/ultrastructure , Heterozygote , Humans , Huntingtin Protein , Mice , Mice, Inbred Strains , Mice, Transgenic , Mitochondria/pathology , Motor Activity , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Putamen/pathology , Somatosensory Cortex/pathology
3.
J Neurosci Methods ; 96(1): 35-45, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10704669

ABSTRACT

We demonstrate here that electrical stimulation of organotypic cultures of rat hippocampus results in the prompt release of significant amounts of Zn(II) by a fluorescence microscopic method. The fluorescence imaging of free Zn(II) is achieved using a highly selective biosensing indicator system consisting of human apo-carbonic anhydrase II (apoCAII) and a fluorescent aryl sulfonamide inhibitor of the enzyme, ABD-N. The apoenzyme and ABD-N in the absence of Zn(II) exhibit weak, reddish fluorescence typical of the ABD-N alone; when Zn(II) is added it binds to the apoenzyme (K(D) = 4 pM), which strongly promotes binding of ABD-N to the holoenzyme (K(D) = 0.9 microM). Binding of ABD-N to the holoenzyme results in a 9-fold increase in apparent quantum yield, significant blue shifts in excitation and emission, an increase in average fluorescence lifetime, a 4-fold increase in the ratio of intensities at 560 and 680 nm, and a large increase in anisotropy. Prior to stimulation, cultures immersed in phosphate-buffered saline with glucose and apoCAII with ABD-N emitted negligible fluorescence, but within 20 s after electrical stimulation a diffuse cloud of greenish fluorescence emerged and subsequently covered most of the culture, indicating release of zinc into the extracellular medium.


Subject(s)
Biosensing Techniques/methods , Carbonic Anhydrases/pharmacology , Hippocampus/metabolism , Microscopy, Fluorescence/methods , Zinc/metabolism , Animals , Animals, Newborn , Anisotropy , Electric Stimulation , Humans , Mammals , Organ Culture Techniques , Oxadiazoles/chemical synthesis , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Sulfonamides/chemical synthesis
4.
Philos Trans R Soc Lond B Biol Sci ; 354(1386): 1035-45, 1999 Jun 29.
Article in English | MEDLINE | ID: mdl-10434303

ABSTRACT

Huntington's disease (HD) is a progressive neurodegenerative disorder characterized clinically by motor and psychiatric disturbances and pathologically by neuronal loss and gliosis (reactive astrocytosis) particularly in the striatum and cerebral cortex. We have recently created HD full-length cDNA transgenic mouse models that may serve as a paradigm for HD. A more detailed characterization of these models is presented here. The transgene encoding normal huntingtin consists of 9417 bp of the huntingtin coding sequences including 16 tandem CAGs coding for polyglutamines as part of exon 1. The transgene is driven by a heterologous cytomegalovirus promoter. Five independent transgenic mouse lines were obtained using this construct. An additional six transgenic lines were obtained using full-length HD constructs that have been modified to include either 48 or 89 CAG repeat expansions. Southern blot and densitometric analyses indicated unique integration sites for the transgene in each of the lines with a copy number ranging from two to 22 copies. Widespread expression of the transgene in brain, heart, spleen, kidney, lung, liver and gonads from each line was determined by Western blot analyses. In the brain, transgene expression was found in cerebral cortex, striatum, hippocampus and cerebellum. Expression of the transgene was as much as five times the endogenous mouse huntingtin level. Phenotypically, only mice expressing 48 or 89 CAG repeats manifested progressive behavioural and motor dysfunction. Early behavioural abnormalities were characterized by trunk curling and clasping of both fore- and hindlimbs when the animals were suspended by their tails. Subsequently, these mice exhibited hyperkinetic movements, including heightened exploratory activities, unidirectional rotational behaviour, backflipping and excessive grooming that lasted for several weeks. Eventually, the animals progressed to a hypokinetic phase consisting of slowed movements and lack of response to sensory stimuli. Urine retention or incontinence was also a prominent feature of the hypokinetic phase. At the end stage of the disease process, HD48(B,D) and HD89(A-C) mice became akinetic just prior to death. Neuropathological examination of mice at various stages indicated that it was only during the hypokinetic phase and thereafter when selective neuronal loss was most apparent. Regions of neurodegeneration and loss included the striatum, cerebral cortex, thalamus and hippocampus. TUNEL staining indicated an apoptotic mode of cell death in these brain regions. Comparative neuronal counts after Nissl staining showed as much as 20% loss of small and medium neurons in the striatum in mice at the hypokinetic and akinetic stages. Reactive astrocytosis accompanied the areas of neurodegeneration and loss. Polyglutamine aggregates in the form of neuronal intranuclear inclusions and diffuse nuclear and perinuclear aggregations were found in a small percentage of neurons, including those in brain regions that are typically spared in HD. This observation suggests that polyglutamine aggregates may not be sufficient to cause neuronal loss in HD. In both behavioural and neuropathological analyses, wild-type and transgenic animals with 16 CAG repeats were indistinguishable from each other and do not exhibit the changes observed for mice carrying the 48 and 89 CAG repeat mutations. Thus, animals expressing the CAG repeat expansions appear to represent clinically analogous models for HD pathogenesis, and may also provide insights into the underlying pathophysiological mechanisms of other triplet repeat disorders.


Subject(s)
Brain/pathology , Huntington Disease/genetics , Motor Activity , Nerve Tissue Proteins/genetics , Neurons/pathology , Nuclear Proteins/genetics , Animals , DNA, Complementary , Exons , Gliosis , Humans , Huntingtin Protein , Huntington Disease/pathology , Huntington Disease/physiopathology , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Peptides/genetics , Phenotype , Trinucleotide Repeat Expansion/genetics
5.
Nat Genet ; 20(2): 198-202, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9771716

ABSTRACT

Huntington disease (HD) is an adult-onset, autosomal dominant inherited human neurodegenerative disorder characterized by hyperkinetic involuntary movements, including motor restlessness and chorea, slowing of voluntary movements and cognitive impairment. Selective regional neuron loss and gliosis in striatum, cerebral cortex, thalamus, subthalamus and hippocampus are well recognized as neuropathological correlates for the clinical manifestations of HD. The underlying genetic mutation is the expansion of CAG trinucleotide repeats (coding for polyglutamines) to 36-121 copies in exon 1 of the HD gene. The HD mRNA and protein product (huntingtin) show widespread distribution, and thus much remains to be understood about the selective and progressive neurodegeneration in HD. To create an experimental animal model for HD, transgenic mice were generated showing widespread expression of full-length human HD cDNA with either 16, 48 or 89 CAG repeats. Only mice with 48 or 89 CAG repeats manifested progressive behavioural and motor dysfunction with neuron loss and gliosis in striatum, cerebral cortex, thalamus and hippocampus. These animals represent clinically relevant models for HD pathogenesis, and may provide insights into the underlying pathophysiological mechanisms of other triplet repeat disorders.


Subject(s)
Behavior, Animal , Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Neurons/pathology , Nuclear Proteins/genetics , Animals , Apoptosis , Corpus Striatum/chemistry , DNA, Complementary/biosynthesis , DNA, Complementary/chemistry , Humans , Huntingtin Protein , Huntington Disease/pathology , Mice , Mice, Transgenic , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/biosynthesis , Nuclear Proteins/analysis , Nuclear Proteins/biosynthesis , Restriction Mapping , Trinucleotide Repeats/genetics , Ubiquitins/analysis
6.
Exp Neurol ; 148(1): 26-33, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9398447

ABSTRACT

Progression of Parkinson's disease has been associated with several biochemical changes in the substantia nigra including increased oxidative challenge, catechol oxidation, and inhibition of mitochondrial complex I activity. Cysteinylcatechols, formed by nucleophilic addition of cysteine to oxidized catechols, have been identified as markers of catechol oxidation in brain tissue. We have examined the neurotoxicity of a series of cysteinylcatechols. Of the compounds examined, only 5-S-cysteinyl-3,4-dihydroxyphenylacetate (cysdopac) was specifically cytotoxic to differentiated P19 neuroglial cultures. Cysdopac also was neurotoxic to pyramidal neurons in organotypic cultures of hippocampus, and this effect was ablated by selective N-methyl-D-aspartate (NMDA) receptor antagonists. In vitro, cysdopac was a potent inhibitor of mitochondrial complex I activity. However, electrophysiologic experiments failed to demonstrate NMDA receptor agonist activity for cysdopac, nor did cysdopac inhibit glutamate uptake. These results showed that cysdopac was the most potent neurotoxin of this series of cysteinylcatechols and suggest that cysdopac may function as an indirect excitotoxin, potentially via inhibition of mitochondrial respiration.


Subject(s)
Catechols/toxicity , Cysteine/analogs & derivatives , Neuroglia/drug effects , Parkinson Disease/metabolism , Pyramidal Cells/drug effects , Animals , Brain Chemistry , Carcinoma, Embryonal/pathology , Catechols/metabolism , Cells, Cultured , Culture Media , Cysteine/metabolism , Cysteine/toxicity , Dopamine/metabolism , Electron Transport/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/drug effects , Magnetic Resonance Spectroscopy , N-Methylaspartate/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , Receptors, Glutamate/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Tumor Cells, Cultured
7.
Am J Pathol ; 150(2): 437-43, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9033259

ABSTRACT

Cumulative oxidative damage, including lipid peroxidation, is a central component of cellular aging and is thought to play a role in the pathogenesis of late-onset Alzheimer's disease (AD). Lipid peroxidation produces several cytotoxic aldehydes, one of the most potent being 4-hydroxy-2-nonenal (HNE). We have shown previously that HNE is a potent neurotoxin that covalently modifies and cross-links neuronal cytoskeletal protein in neuroglial cultures, suggesting that HNE may contribute to the pathogenesis of AD. In addition to aging, inheritance of the epsilon 4 allele of APOE is the other major risk factor for development of late-onset AD; however, the mechanisms through which aging and apolipoprotein E isoforms may collaborate in the onset or progression of AD are not known. We tested the hypothesis that HNE may yield a particular type of protein modification, pyrrole adduction, and that this may contribute to the pathogenesis of AD. Our data demonstrated that HNE formed pyrrole adducts with protein. Polyclonal antiserum was raised that specifically recognized HNE pyrrole adducts, and immunohistochemical analysis was performed on hippocampus and temporal cortex of 10 patients with histologically verified AD. Pyramidal neuron cytoplasm was immunoreactive in 4 of 4 APOE4 homozygotes, 2 of 3 APOE3/4 heterozygotes, and none of 3 APOE3 homozygotes (P < 0.05). The pattern of staining was highly suggestive of neurofibrillary tangles as the primary immunoreactive structure. These data suggest that differences in neuronal protein modification by HNE may account in part for the APOE-associated stratification of risk for late-onset AD.


Subject(s)
Aldehydes/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Apolipoproteins E/genetics , Aged , Aged, 80 and over , Apolipoprotein E4 , Brain/metabolism , Female , Genotype , Heterozygote , Homozygote , Humans , Immunohistochemistry , Male , Pyrroles/metabolism , Pyrrolidines/metabolism , Tissue Distribution
8.
J Neural Transm (Vienna) ; 104(8-9): 875-85, 1997.
Article in English | MEDLINE | ID: mdl-9451719

ABSTRACT

Systemic administration of MPTP to experimental animals induces neurodegeneration of dopaminergic neurons in the central nervous system. MPTP crosses the blood-brain barrier where it is taken up by astrocytes and converted to MPP+ by monamine oxidase-B (MAO-B). Subsequently, MPP+ is selectively taken up by dopaminergic neurons upon which it exerts intracellular neurotoxic effects. Systemic administration of the selective MAO-B inhibitor deprenyl prevents the conversion of MPTP to MPP+ and by this mechanism is able to protect against MPTP neurotoxicity. Deprenyl has also been reported to exert neuroprotective effects that are independent of its MAO-B inhibitory properties, but since MPP+ itself does not cross the blood-brain barrier it is difficult to directly study the MAO-B independent in vivo effects of MPP+ itself. One approach is to use organotypic tissue cultures of the canine substantia nigra (CSN) which permit administration of precise concentrations of pharmacological agents directly to mature, well-developed and metabolically active dopaminergic neurons. These neurons as well as other components of the cultures exhibit morphological and biochemical characteristics identical to their in vivo counterparts. This study was undertaken to evaluate the neuroprotective effects of deprenyl in MPP(+)-treated cultures by measuring changes in the levels of HVA as an indicator of dopamine release and metabolism by dopaminergic neurons and to correlate this indication of dopaminergic function with morphological evidence of survival or loss of dopaminergic neurons in mature CSN cultures. Mature CSN cultures, at 44 days in vitro (DIV), were exposed to either MPP+ alone, deprenyl alone or simultaneously to both deprenyl and MPP+ or to MPP+ following 4 day pretreatment with deprenyl. Exposure to MPP+ alone caused significant reduction in HVA levels, evidence of widespread injury and ultimate disappearance of large neurons in the cultures. These effects were attenuated by simultaneous exposure to MPP+ and deprenyl and the destructive effects of MPP+ appeared to be prevented by pretreatment with deprenyl. Thus the neuroprotective effects of deprenyl on MPP(+)-induced reduction of HVA levels in living cultures appears similar to the effects of deprenyl on dopamine levels and tyrosine hydroxylase activity reported by others in cultures previously exposed to deprenyl and MPP+. These studies also confirm that the neuroprotective effects of deprenyl against MPP+ in dopaminergic neurons are, at least in part, independent of deprenyl's inhibition of MAO-B.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Dopamine Agents/toxicity , MPTP Poisoning , Neuroprotective Agents/pharmacology , Selegiline/pharmacology , Substantia Nigra/drug effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenylpyridinium/metabolism , Animals , Animals, Newborn , Dogs , Dopamine/metabolism , Dopamine Agents/metabolism , Homovanillic Acid/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Organ Culture Techniques , Substantia Nigra/metabolism , Substantia Nigra/pathology
9.
Neurosci Res ; 29(4): 303-9, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9527621

ABSTRACT

Quinolinic acid (QUIN) is an endogenous excitatory amino acid, which is elevated in brain tissues or cerebrospinal fluid (CSF) in several acute and chronic inflammatory central nervous system (CNS) diseases. The functional significance of this elevation is unknown but speculations of excitotoxicity have been raised. We have begun to address the pathologic consequences of elevated CSF QUIN by studying the effects of intracerebroventricular (i.cv) administration of QUIN on regional choline acetyltransferase (ChAT) activity, somatostatin content and glucose metabolism in the rat brain. QUIN (12 and 60 nmol) i.cv administration once a day for 7 days (total dose; 84 and 420 nmol, respectively) had minimal effect on somatostatin content and no effect on ChAT activity. In contrast, following continuous i.cv infusion of QUIN for 14 days using an osmotic minipump (480 nmol), ChAT activity dropped in the hippocampus and the striatum and somatostatin content was reduced in the frontal cortex, hippocampus, striatum and amygdala. Moreover, following the QUIN infusion, glucose utilization decreased in the basal nucleus of Meynert, frontal cortex, and portions of the basal ganglia and the limbic system. These results indicate that subchronic i.cv infusion of QUIN to rats results in selective regional neurochemical and metabolic changes distributed throughout the CNS. These results suggest target brain areas and transmitter systems which may be associated with neurologic syndromes characterized by elevated CSF QUIN levels.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Quinolinic Acid/pharmacology , Animals , Brain/metabolism , Brain/pathology , Choline O-Acetyltransferase/metabolism , Glucose/metabolism , Injections, Intraventricular , Male , Quinolinic Acid/cerebrospinal fluid , Rats , Rats, Sprague-Dawley , Somatostatin/metabolism
10.
Pediatr Neurosurg ; 27(4): 190-3, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9577972

ABSTRACT

It has been proposed that the myelodysplastic components of a myelomeningocele are secondarily damaged as the result of exposure to amniotic fluid, the so-called 'two-hit' hypothesis. The critical time at which this secondary insult might occur has not been clearly defined. The present study addresses this issue by quantitatively assessing the toxic effects of human amniotic fluid of various gestational ages upon organotypic cultures of rat spinal cord. Using an assay for lactate dehydrogenase efflux to evaluate toxicity in such spinal cord cultures, we found that the amniotic fluid became toxic at approximately 34 weeks' gestation. This toxic effect of amniotic fluid appears to emerge rather suddenly. Accordingly, it seems reasonable to suggest that prevention of exposure of vulnerable spinal cord tissue to this toxicity by surgical closure of a myelomeningocele defect prior to the emergence of toxicity in amniotic fluid may prevent injury to vulnerable myelodysplastic spinal cord tissue.


Subject(s)
Amniotic Fluid/chemistry , L-Lactate Dehydrogenase/analysis , Meningomyelocele/embryology , Spinal Cord/embryology , Spinal Cord/enzymology , Animals , Biomarkers/analysis , Culture Techniques , Fetus , Gestational Age , Humans , Rats
12.
J Rheumatol ; 22(9): 1769-70, 1995 Sep.
Article in English | MEDLINE | ID: mdl-8523360

ABSTRACT

Malignant angioendotheliomatosis is characterized by the plugging of central nervous system blood vessels with large mononuclear cells, thus mimicking primary angiitis of the central nervous system. When autoantibodies are present, it can mimic connective tissue disease. Treatment of malignant angioendotheliomatosis with cytoxan may prolong survival.


Subject(s)
Central Nervous System Diseases/pathology , Lymphoma, Non-Hodgkin/pathology , Vasculitis/pathology , Aged , Central Nervous System Diseases/complications , Diagnosis, Differential , Fatal Outcome , Female , Humans , Vasculitis/complications
13.
J Neurol Sci ; 130(1): 39-47, 1995 May.
Article in English | MEDLINE | ID: mdl-7650530

ABSTRACT

The levels of the neuroprotective excitatory amino acid receptor antagonist kynurenic acid (KYNA) have been previously shown to be reduced in several regions of the brain of Huntington's disease (HD) patients. Thus, KYNA has been speculatively linked to the pathogenesis of HD. We have examined KYNA levels and the activity of its two biosynthetic enzymes (kynurenine aminotransferases (KAT) I and II) in 12 regions of brains from late-stage HD patients and control donors (n = 17 each). KYNA levels were measured in the original tissue homogenate. Using [3H]kynurenine as the substrate, enzyme activities were determined in dialyzed tissue homogenates. KYNA levels in the caudate nucleus decreased from 733 +/- 95 in controls to 401 +/- 62 fmol/mg tissue in HD (p < 0.01). The activity of both enzymes was highest in cortical areas (e.g. control frontal cortex: KAT I: 148 +/- 18 fmol/mg tissue/h; KAT II: 25 +/- 2 fmol/mg tissue/h). The activities of both KAT I and KAT II, when expressed per mg original weight, showed significant decreases (48-55%) in the HD putamen (p < 0.01). Trends toward lower enzyme activities and KYNA concentrations were detected in other brain areas as well. Kinetic analyses, performed in putamen and cerebellum, showed an approximately 3-fold increase in Km values for both KAT I and KAT II in the putamen only. Vmax values remained unchanged in the HD brain. These findings indicate a selective impairment in KYNA biosynthesis in the neostriatum of HD patients, possibly due to the loss of (an) endogenous KAT activator(s).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Brain Chemistry/physiology , Huntington Disease/metabolism , Kynurenic Acid/metabolism , Lyases , Transaminases/metabolism , Aged , Brain/enzymology , Brain/pathology , Female , Humans , Huntington Disease/enzymology , Huntington Disease/pathology , Kinetics , Male , Middle Aged , Nerve Tissue Proteins/metabolism
14.
Epilepsia ; 35(5): 1079-84, 1994.
Article in English | MEDLINE | ID: mdl-7925155

ABSTRACT

A 33-year-old right-handed woman had intractable simple and complex partial seizures (SPS, CPS) that began with global aphasia. EEG closed-circuit TV (EEG-CCTV) monitoring with sphenoidal electrodes showed left inferomesial temporal ictal onset of CPS. Subdural electrodes were implanted over the left frontotemporal convexity, subtemporally and subfrontally. Stimulation of the basotemporal cortex produced global aphasia. A posterolaterotemporal language area was also identified. Spontaneous SPS had focal onset in the basal temporal language area (BTLA). Ictal discharges did not involve the posterotemporal region. This case shows that aphasic speech arrest at seizure onset may be due to seizure discharge in the basotemporal region and that the BTLA is clinically relevant in seizure semiology.


Subject(s)
Aphasia/diagnosis , Electroencephalography , Epilepsy, Temporal Lobe/diagnosis , Functional Laterality/physiology , Temporal Lobe/physiopathology , Adult , Aphasia/physiopathology , Electric Stimulation , Epilepsy, Complex Partial/diagnosis , Epilepsy, Complex Partial/physiopathology , Epilepsy, Temporal Lobe/physiopathology , Female , Frontal Lobe/pathology , Frontal Lobe/physiopathology , Humans , Magnetic Resonance Imaging , Temporal Lobe/pathology , Tomography, Emission-Computed , Videotape Recording
15.
Hypertension ; 23(4): 491-5, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8144218

ABSTRACT

Animal studies have shown the importance of the nucleus tractus solitarii, a collection of neurons in the brain stem, in the acute regulation of blood pressure. Impulses arising from the carotid and aortic baroreceptors converge in this center, where the first synapse of the baroreflex is located. Stimulation of the nucleus tractus solitarii provides an inhibitory signal to other brain stem structures, particularly the rostral ventrolateral medulla, resulting in a reduction in sympathetic outflow and a decrease in blood pressure. Conversely, experimental lesions of the nucleus tractus solitarii lead to loss of baroreflex control of blood pressure, sympathetic activation, and severe hypertension in animals. In humans, baroreflex failure due to deafferentation of baroreceptors has been previously reported and is characterized by episodes of severe hypertension and tachycardia. We present a patient with an undetermined process of the central nervous system characterized pathologically by ubiquitous infarctions that were particularly prominent in the nucleus tractus solitarii bilaterally but spared the rostral ventrolateral medulla. Absence of a functioning baroreflex was evidenced by the lack of reflex tachycardia to the hypotensive effects of sodium nitroprusside, exaggerated pressor responses to handgrip and cold pressor test, and exaggerated depressor responses to meals and centrally acting alpha 2-agonists. This clinicopathological correlate suggests that the patient's baroreflex failure can be explained by the unique combination of the destruction of sympathetic inhibitory centers (ie, the nucleus tractus solitarii) and preservation of centers that exert a positive modulation on sympathetic tone (ie, the rostral ventrolateral medulla).


Subject(s)
Baroreflex/physiology , Cerebral Infarction/physiopathology , Hypertension/etiology , Solitary Nucleus/blood supply , Adult , Blood Pressure , Cerebral Infarction/complications , Eating/physiology , Humans , Male , Pressoreceptors/physiopathology , Smoking/physiopathology , Solitary Nucleus/physiopathology
16.
Alcohol Alcohol Suppl ; 2: 273-9, 1994.
Article in English | MEDLINE | ID: mdl-8974347

ABSTRACT

There is increasing evidence for the role of thiamine deficiency in ethanol neurotoxicity and in development of alcoholic organic brain disorders other than Wernicke-Korsakoff syndrome [WKS] and cerebellar degeneration. Investigations in humans and in animal models have implicated a reduction in the activities of thiamine-utilizing enzymes as the metabolic basis of tissue injury due to thiamine deficiency. We have investigated the interactions of the thiamine-utilizing enzyme transketolase [Tk], derived from human fibroblasts, lymphoblasts, and various brain regions, with its cofactor, thiamine pyrophosphate [TPP], in an attempt to elucidate the molecular basis of selective brain damage in alcoholism-associated thiamine deficiency. There were no significant differences in the isoelectric pattern of Tk among the nine brain regions (white matter and grey matter) examined. However, Tk activity/mg protein, increase in Tk activity with addition of excess TPP (TPP effect), and TPP-dependent rate of formation of active Tk holoenzyme (tau) varied 2.5-, 6-, and 4-fold, respectively, among these brain regions. These differences in tissue requirements for TPP may contribute to the selective vulnerability of certain brain regions to alcoholism-associated thiamine deficiency, and may influence the pattern of clinical impairment in the individual patient.


Subject(s)
Alcoholism/complications , Alcoholism/metabolism , Brain Injuries/etiology , Brain Injuries/metabolism , Thiamine/metabolism , Alcohol Amnestic Disorder/etiology , Alcohol Amnestic Disorder/metabolism , Animals , Humans , Thiamine Deficiency/etiology , Thiamine Deficiency/metabolism , Thiamine Pyrophosphate/metabolism , Tissue Distribution , Transketolase/metabolism , Wernicke Encephalopathy/etiology , Wernicke Encephalopathy/metabolism
17.
Epilepsy Res ; 16(3): 223-33, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8119273

ABSTRACT

We report a characteristic pattern of neuropathological change in the entorhinal cortex (EC) from four patients with temporal lobe epilepsy. Specimens of the EC were obtained during the surgical treatment of intractable partial seizures and were studied by light microscopy in Nissl-stained sections. A distinct loss of neurons was observed in the anterior portion of the medial EC in the absence of apparent damage to temporal neocortical gyri. Cell loss was most pronounced in layer III, but also noticed in layer II, particularly in the rostral field. A similar pattern of neurodegeneration in the EC was found in all specimens examined though the degree of neuronal loss varied between cases. These observations provide neuropathological evidence for an involvement of the EC in temporal lobe epilepsy. Since the EC occupies a pivotal position in gating hippocampal input and output, our results further support previous suggestions that dysfunction of this region may contribute, either independently or in concert with Ammon's horn sclerosis, to epileptogenesis in humans.


Subject(s)
Epilepsy, Temporal Lobe/pathology , Hippocampus/pathology , Neurons/pathology , Adult , Cell Death/physiology , Female , Humans , Male
18.
Brain Res ; 609(1-2): 237-43, 1993 Apr 23.
Article in English | MEDLINE | ID: mdl-8099521

ABSTRACT

The regional distribution of striatal and extrastriatal dopamine D2 receptors in human brain was studied in vitro with (S)-N-[(1-ethyl-2- pyrrolidinyl)methyl]-5-[125I]iodo-2,3-dimethoxybenzamide, [125I]epidepride, using post mortem brain specimens from six subjects. Scatchard analysis of the saturation equilibrium binding in twenty-three regions of post mortem brain revealed highest levels of binding in the caudate (16.5 pmol/g tissue) and putamen (16.6 pmol/g tissue) with lower levels seen in the globus pallidus (7.0 pmol/g tissue), nucleus accumbens (7.2 pmol/g tissue), hypothalamus (1.8 pmol/g tissue), pituitary (1.3 pmol/g tissue), substantia innominata (1.0 pmol/g tissue), and amygdala (0.87 pmol/g tissue). Of note was the presence of dopamine D2 receptors in the four thalamic nuclei studied, i.e. anterior nucleus (1.0 pmol/g tissue), dorsomedial nucleus (0.96 pmol/g tissue), ventral nuclei (0.72 pmol/g tissue), and pulvinar (0.86 pmol/g tissue), at levels comparable to the amygdala (0.87 pmol/g tissue) and considerably higher than levels seen in anterior cingulate (0.26 pmol/g tissue) or anterior hippocampus (0.36 pmol/g tissue). The frontal cortex had very low levels of dopamine D2 receptors (0.17-0.20 pmol/g tissue) while the inferior and medial temporal cortex had relatively higher levels (0.31-0.46 pmol/g tissue). Inhibition of [125I]epidepride binding by a variety of neurotransmitter ligands to striatal, ventral thalamic and inferior temporal cortical homogenates demonstrated that [125I]epidepride binding was potently inhibited only by dopamine D2 ligands. The present study demonstrates that dopamine D2 receptors are present in basal ganglia, many limbic regions, cortex and in the thalamus. The density of thalamic D2 receptors is comparable to many limbic regions and is considerably higher than in cortex.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Benzamides , Brain Chemistry/drug effects , Pyrrolidines , Receptors, Dopamine D2/metabolism , Adrenergic alpha-Antagonists/pharmacology , Aged , Aged, 80 and over , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Dioxanes/pharmacology , Humans , Idazoxan , In Vitro Techniques , Iodine Radioisotopes , Kinetics , Limbic System/drug effects , Limbic System/metabolism , Male , Receptors, Dopamine D2/analysis , Thalamus/drug effects , Thalamus/metabolism
20.
Laryngoscope ; 102(9): 1030-6, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1518348

ABSTRACT

The present study was undertaken to systematically examine and characterize pathological changes in vestibular nerve specimens obtained at surgery in patients with symptomatic cochleovestibular nerve compression syndrome (CNCS). Vestibular nerves were obtained in six cases of CNCS and were intermingled with vestibular nerves obtained in cases of Meniere's disease. All of the nerve specimens were coded and reviewed microscopically in a blind-study fashion by the neuropathologist. The vestibular nerves obtained from CNCS cases showed significant endoneurial fibrosis, compared to controls (specimens from patients with Meniere's disease). Based on observations in this study, as well as the clinical symptoms and audiovestibular test findings in these patients, a theory of pathophysiology in CNCS of the cochleovestibular nerve is proposed. The implications of this theory are discussed with respect to the diagnosis of CNCS.


Subject(s)
Cochlear Nerve/pathology , Cochlear Nerve/physiopathology , Nerve Compression Syndromes/pathology , Nerve Compression Syndromes/physiopathology , Vestibular Nerve/pathology , Vestibular Nerve/physiopathology , Axons/ultrastructure , Chronic Disease , Fibrosis , Hearing Loss, Sensorineural/pathology , Hearing Loss, Sensorineural/physiopathology , Humans , Meniere Disease/pathology , Meniere Disease/physiopathology , Myelin Sheath/ultrastructure , Nerve Compression Syndromes/diagnosis , Otitis Media/pathology , Otitis Media/physiopathology , Vertigo/pathology , Vertigo/physiopathology , Vestibular Nerve/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...