Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters










Publication year range
1.
J Mol Neurosci ; 17(3): 361-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11859932

ABSTRACT

THE RESEARCH OBJECTIVE: of this study was to test whether variation in mitochondrial composition is associated with "selective vulnerability" in Alzheimer brain. The term "selective vulnerability" refers to the loss of relatively vulnerable brain cells and the sparing of relatively resistant brain cells in disorders in which a genetic defect or environmental agent acts on both types of cells. The mechanisms underlying selective vulnerability are largely unknown, but mitochondria may be involved; the composition of mitochondria varies among different types of neurons, and mitochondria have an important role in cell death. Alzheimer's Disease (AD) is one of a number of neurodegenerative disorders in which both selective vulnerability and abnormalities of mitochondria occur. METHODS: We examined by immunohistochemistry the cellular distribution of a mitochondrial constituent (the alpha-ketoglutarate dehydrogenase complex, KGDHC) known to be deficient in AD, in relation to the known selective vulnerability of neurons in areas 21 and 22 of the temporal lobe in this neurodegenerative disorder. RESULTS: In normal human brain, cortical layers III and V contain neurons intensely immunoreactive for KGDHC, compared to other cells in these areas. The KGDHC-enriched cells are lost in AD (p < 0.001). In layer III, the loss of KGDHC-enriched cells is proportional to total loss of neurons, as determined by immunoreactivity to neuron specific enolase (NSE). In layer V, a higher proportion of the KGDHC-enriched neurons are lost than of other (NSE positive) neurons (p < 0.001). SIGNIFICANCE: Variations in mitochondrial composition may be one of the factors determining which cells die first when different types of cells are exposed to the same stress.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Ketoglutarate Dehydrogenase Complex/analysis , Mitochondria/enzymology , Temporal Lobe/enzymology , Temporal Lobe/pathology , Aged , Autopsy , Biopsy , Cell Count , Cell Death , Female , Glial Fibrillary Acidic Protein/analysis , Humans , Immunohistochemistry , Ketoglutarate Dehydrogenase Complex/immunology , Male , Middle Aged , Neurons/enzymology , Phosphopyruvate Hydratase/analysis
2.
J Biol Chem ; 275(18): 13441-7, 2000 May 05.
Article in English | MEDLINE | ID: mdl-10788456

ABSTRACT

Intracellular free Zn(2+) is elevated in a variety of pathological conditions, including ischemia-reperfusion injury and Alzheimer's disease. Impairment of mitochondrial respiration is also associated with these pathological conditions. To test whether elevated Zn(2+) and impaired respiration might be linked, respiration of isolated rat liver mitochondria was measured after addition of Zn(2+). Zn(2+) inhibition (K(i)(app) = approximately 1 micrometer) was observed for respiration stimulated by alpha-ketoglutarate at concentrations well within the range of intracellular Zn(2+) reported for cultured hepatocytes. The bc(1) complex is inhibited by Zn(2+) (Link, T. A., and von Jagow, G. (1995) J. Biol. Chem. 270, 25001-25006). However, respiration stimulated by succinate (K(i)(app) = approximately 6 micrometer) was less sensitive to Zn(2+), indicating the existence of a mitochondrial target for Zn(2+) upstream from bc(1) complex. Purified pig heart alpha-ketoglutarate dehydrogenase complex was strongly inhibited by Zn(2+) (K(i)(app) = 0.37 +/- 0.05 micrometer). Glutamate dehydrogenase was more resistant (K(i)(app) = 6 micrometer), malate dehydrogenase was unaffected, and succinate dehydrogenase was stimulated by Zn(2+). Zn(2+) inhibition of alpha-ketoglutarate dehydrogenase complex required enzyme cycling and was reversed by EDTA. Reversibility was inversely related to the duration of exposure and the concentration of Zn(2+). Physiological free Zn(2+) may modulate hepatic mitochondrial respiration by reversible inhibition of the alpha-ketoglutarate dehydrogenase complex. In contrast, extreme or chronic elevation of intracellular Zn(2+) could contribute to persistent reductions in mitochondrial respiration that have been observed in Zn(2+)-rich diseased tissues.


Subject(s)
Ketoglutarate Dehydrogenase Complex/metabolism , Ketoglutaric Acids/metabolism , Mitochondria, Liver/metabolism , Zinc/metabolism , Animals , Electron Transport/drug effects , Enzyme Activation/drug effects , Ketoglutaric Acids/pharmacology , Rats , Zinc/pharmacology
3.
Exp Neurol ; 161(1): 115-26, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10683278

ABSTRACT

Chronic impairment of aerobic energy metabolism accompanies global cerebral ischemia and reperfusion and likely contributes to delayed neuronal cell death. Reperfusion-dependent inhibition of pyruvate dehydrogenase complex (PDHC) enzyme activity has been described and proposed to be at least partially responsible for this metabolic abnormality. This study tested the hypothesis that global cerebral ischemia and reperfusion results in the loss of pyruvate dehydrogenase immunoreactivity and that such loss is associated with selective neuronal vulnerability to transient ischemia. Following 10 min canine cardiac arrest, resuscitation, and 2 or 24 h of restoration of spontaneous circulation, brains were either perfusion fixed for immunohistochemical analyses or biopsy samples were removed for Western immunoblot analyses of PDHC immunoreactivity. A significant decrease in immunoreactivity was observed in frontal cortex homogenates from both 2 and 24 h reperfused animals compared to samples from nonischemic control animals. These results were supported by confocal microscopic immunohistochemical determinations of pyruvate dehydrogenase immunoreactivity in the neuronal cell bodies located within different layers of the frontal cortex. Loss of immunoreactivity was greatest for pyramidal neurons located in layer V compared to neurons in layers IIIc/IV, which correlates with a greater vulnerability of layer V neurons to delayed death caused by transient global cerebral ischemia.


Subject(s)
Brain Ischemia/metabolism , Heart Arrest/metabolism , Neurons/enzymology , Pyruvate Dehydrogenase Complex/analysis , Reperfusion Injury/metabolism , Animals , Antibodies , Cardiopulmonary Resuscitation , Dogs , Female , Frontal Lobe/blood supply , Frontal Lobe/cytology , Frontal Lobe/enzymology , Microscopy, Confocal , Microtubule-Associated Proteins/analysis , Mitochondria/enzymology , Neurons/chemistry , Pyruvate Dehydrogenase Complex/immunology
4.
Neurochem Int ; 36(2): 97-112, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10676873

ABSTRACT

Altered energy metabolism is characteristic of many neurodegenerative disorders. Reductions in the key mitochondrial enzyme complex, the alpha-ketoglutarate dehydrogenase complex (KGDHC), occur in a number of neurodegenerative disorders including Alzheimer's Disease (AD). The reductions in KGDHC activity may be responsible for the decreases in brain metabolism, which occur in these disorders. KGDHC can be inactivated by several mechanisms, including the actions of free radicals (Reactive Oxygen Species, ROS). Other studies have associated specific forms of one of the genes encoding KGDHC (namely the DLST gene) with AD, Parkinson's disease, as well as other neurodegenerative diseases. Reductions in KGDHC activity can be plausibly linked to several aspects of brain dysfunction and neuropathology in a number of neurodegenerative diseases. Further studies are needed to assess mechanisms underlying the sensitivity of KGDHC to oxidative stress and the relation of KGDHC deficiency to selective vulnerability in neurodegenerative diseases.


Subject(s)
Ketoglutarate Dehydrogenase Complex , Neurodegenerative Diseases/enzymology , Brain/enzymology , Brain/pathology , Brain/physiopathology , Gene Expression Regulation , Humans , Ketoglutarate Dehydrogenase Complex/analysis , Ketoglutarate Dehydrogenase Complex/genetics , Ketoglutarate Dehydrogenase Complex/metabolism , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology
5.
Anal Biochem ; 277(1): 86-93, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10610692

ABSTRACT

The activity of a key mitochondrial enzyme, the alpha-ketoglutarate dehydrogenase complex (KGDHC), declines in the brains of patients with neurodegenerative diseases such as Alzheimer's disease, as well as in thiamine-deficient (TD) animals. The decreased activity often occurs without a reduction in enzyme protein, which negates the use of immunocytochemistry to study cellular or regional changes in enzyme activity within the brain. To overcome this limitation, an activity staining method using nitroblue tetrazolium was developed. The histochemical activity staining was standardized in cultured cells. The assay was linear with time and was highly specific for KGDHC. The dark-blue reaction product (formazan) formed a pattern that was consistent with mitochondrial localization. Treatment of the cultured cells with both reversible and irreversible inhibitors decreased formazan production, whereas conventional enzyme assays on cell lysates only revealed loss of KGDHC activity with irreversible inhibitors. The activity staining was also linear with time and highly specific for KGDHC activity in mouse brain sections. Staining occurred throughout the brain, and discrete neuronal populations exhibited particularly intense staining. The pattern of staining differed markedly from the distribution of KGDHC protein by immunocytochemistry. Generalized decreases in the intensity of activity staining that occurred in the TD brains compared to controls were comparable with the loss of KGDHC activity by conventional enzyme assay. Thus, the present study introduces a new histochemical method to measure KGDHC activity at the cellular and regional level, which will be useful to determine changes of in situ enzyme activity.


Subject(s)
Brain/enzymology , Ketoglutarate Dehydrogenase Complex/metabolism , Neuroblastoma/enzymology , Neurons/enzymology , Animals , Brain/cytology , Coloring Agents , Histocytochemistry , Humans , Immunohistochemistry , Ketoglutarate Dehydrogenase Complex/analysis , Mice , Mice, Inbred C57BL , Neuroblastoma/pathology , Neurons/cytology , Organ Specificity , Tumor Cells, Cultured
6.
Neurology ; 52(7): 1505-7, 1999 Apr 22.
Article in English | MEDLINE | ID: mdl-10227647

ABSTRACT

Recent studies suggest that variants of the DLST gene alter the risk of AD. DLST encodes the core subunit of the mitochondrial alpha-ketoglutarate dehydrogenase complex, which is deficient in AD. The authors report that in 247 US white subjects, homozygosity for DLST A19,117, T19,183 was associated with a reduced risk of AD (odds ratio [OR] = 0.35, p = 0.018). The reduced risk was marked in subjects who did not carry the apolipoprotein (APOE)-4 allele (OR = 0.16, p = 0.014). Further study of DLST in AD appears warranted.


Subject(s)
Acyltransferases/genetics , Alzheimer Disease/genetics , Aged , Aged, 80 and over , Alleles , Apolipoprotein E4 , Apolipoproteins E/genetics , Female , Genotype , Humans , Male , Risk Factors
7.
J Neurochem ; 72(5): 1948-58, 1999 May.
Article in English | MEDLINE | ID: mdl-10217272

ABSTRACT

Microglial activation, oxidative stress, and dysfunctions in mitochondria, including the reduction of cytochrome oxidase activity, have been implicated in neurodegeneration. The current experiments tested the effects of reducing cytochrome oxidase activity on the ability of microglia to respond to inflammatory insults. Inhibition of cytochrome oxidase by azide reduced oxygen consumption and increased reactive oxygen species (ROS) production but did not affect cell viability. Azide also attenuated microglial activation, as measured by nitric oxide (NO.) production in response to lipopolysaccharide (LPS). It is surprising that the inhibition of cytochrome oxidase also diminished the activity of the alpha-ketoglutarate dehydrogenase complex (KGDHC), a Krebs cycle enzyme. This reduction was exaggerated when the azide-treated microglia were also treated with LPS. The combination of the azide-stimulated ROS and LPS-induced NO. would likely cause peroxynitrite formation in microglia. Thus, the possibility that KGDHC was inactivated by peroxynitrite was tested. Peroxynitrite inhibited the activity of isolated KGDHC, nitrated tyrosine residues of all three KGDHC subunits, and reduced immunoreactivity to antibodies against two KGDHC components. Thus, our data suggest that inhibition of the mitochondrial respiratory chain diminishes aerobic energy metabolism, interferes with microglial inflammatory responses, and compromises mitochondrial function, including KGDHC activity, which is vulnerable to NO. and peroxynitrite that result from microglial activation. Thus, activation of metabolically compromised microglia can further diminish their oxidative capacity, creating a deleterious spiral that may contribute to neurodegeneration.


Subject(s)
Microglia/physiology , Mitochondria/enzymology , Oxidative Stress/physiology , Animals , Azides/pharmacology , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation/physiology , Glutamate Dehydrogenase/metabolism , Inflammation/physiopathology , Ketoglutarate Dehydrogenase Complex/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Microglia/metabolism , Nitrates/pharmacology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Nitric Oxide/pharmacology , Oxidoreductases/metabolism , Oxygen Consumption/drug effects , Reactive Oxygen Species/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
8.
J Neurochem ; 72(3): 889-99, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10037459

ABSTRACT

At least eight neurodegenerative diseases, including Huntington disease, are caused by expansions in (CAG)n repeats in the affected gene and by an increase in the size of the corresponding polyglutamine domain in the expressed protein. A hallmark of several of these diseases is the presence of aberrant, proteinaceous aggregates in the nuclei and cytosol of affected neurons. Recent studies have shown that expanded polyglutamine (Qn) repeats are excellent glutaminyl-donor substrates of tissue transglutaminase, and that the substrate activity increases with increasing size of the polyglutamine domain. Tissue transglutaminase is present in the cytosol and nuclear fractions of brain tissue. Thus, the nuclear and cytosolic inclusions in Huntington disease may contain tissue transglutaminase-catalyzed covalent aggregates. The (CAG)n/Qn-expansion diseases are classic examples of selective vulnerability in the nervous system, in which certain cells/structures are particularly susceptible to toxic insults. Quantitative differences in the distribution of the brain transglutaminase(s) and its substrates, and in the activation mechanism of the brain transglutaminase(s), may explain in part selective vulnerability in a subset of neurons in (CAG)n-expansion diseases, and possibly in other neurodegenerative disease. If tissue transglutaminase is found to be essential for development of pathogenesis, then inhibitors of this enzyme may be of therapeutic benefit.


Subject(s)
Inclusion Bodies/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Transglutaminases/metabolism , Trinucleotide Repeat Expansion/genetics , Animals , Humans , Neurodegenerative Diseases/enzymology
9.
Ann Neurol ; 45(1): 48-53, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9894876

ABSTRACT

The mitochondrial alpha-ketoglutarate dehydrogenase complex (KGDHC) is deficient in Alzheimer's disease (AD). The DLST gene encodes the core, dihydrolipoyl succinyltransferase (DLST) component of KGDHC, and recent reports indicate an association between polymorphisms of DLST and AD in both white and Japanese patients. We therefore examined the relationship between AD and the DLST and apolipoprotein E (APOE) genes in elderly (89 +/- 7 years) AD patients, in whom the epsilon4 allele of APOE (APOE4) is a weak risk factor for AD. Polymorphisms of DLST (A19,117G and T19,183C), shown to be of interest in previous studies, were analyzed by restriction fragment length polymorphism analysis after polymerase chain reaction amplification. In a series of 429 white subjects from two Jewish nursing homes, an association of APOE4 with AD was found only in patients homozygous for the G,C allele of DLST. Similar relationships occurred in the "very elderly" (> or =85 years, n = 302) subgroup of this series, and also in an autopsy series (n = 225) that included white subjects from the Jewish nursing homes as well as other white subjects. These findings suggest a relationship between APOE4 and a DLST locus in the pathogenesis of AD in very elderly subjects.


Subject(s)
Acyltransferases/genetics , Alzheimer Disease/epidemiology , Alzheimer Disease/genetics , Age Factors , Aged , Aged, 80 and over , Alleles , Apolipoprotein E4 , Apolipoproteins E/genetics , Female , Genotype , Humans , Male , Polymorphism, Genetic , Restriction Mapping , Risk Factors
10.
Ann N Y Acad Sci ; 893: 61-78, 1999.
Article in English | MEDLINE | ID: mdl-10672230

ABSTRACT

The alpha-ketoglutarate dehydrogenase complex (KGDHC) is an important mitochondrial constituent, and deficiency of KGDHC is associated with a number of neurological disorders. KGDHC is composed of three proteins, each encoded on a different and well-characterized gene. The sequences of the human proteins are known. The organization of the proteins into a large, ordered multienzyme complex (a "metabolon") has been well studied in prokaryotic and eukaryotic species. KGDHC catalyzes a critical step in the Krebs tricarboxylic acid cycle, which is also a step in the metabolism of the potentially excitotoxic neurotransmitter glutamate. A number of metabolites modify the activity of KGDHC, including inactivation by 4-hydroxynonenal and other reactive oxygen species (ROS). In human brain, the activity of KGDHC is lower than that of any other enzyme of energy metabolism, including phosphofructokinase, aconitase, and the electron transport complexes. Deficiencies of KGDHC are likely to impair brain energy metabolism and therefore brain function, and lead to manifestations of brain disease. In general, the clinical manifestations of KGDHC deficiency relate to the severity of the deficiency. Several such disorders have been recognized: infantile lactic acidosis, psychomotor retardation in childhood, intermittent neuropsychiatric disease with ataxia and other motor manifestations, Friedreich's and other spinocerebellar ataxias, Parkinson's disease, and Alzheimer's disease (AD). A KGDHC gene has been associated with the first two and last two of these disorders. KGDHC is not uniformly distributed in human brain, and the neurons that appear selectively vulnerable in human temporal cortex in AD are enriched in KGDHC. We hypothesize that variations in KGDHC that are not deleterious during reproductive life become deleterious with aging, perhaps by predisposing this mitochondrial metabolon to oxidative damage.


Subject(s)
Brain/metabolism , Ketoglutarate Dehydrogenase Complex/genetics , Ketoglutarate Dehydrogenase Complex/metabolism , Neurodegenerative Diseases/enzymology , Animals , Chromosome Mapping , Energy Metabolism , Humans , Ketoglutarate Dehydrogenase Complex/deficiency , Neurodegenerative Diseases/genetics
11.
J Neural Transm (Vienna) ; 105(8-9): 855-70, 1998.
Article in English | MEDLINE | ID: mdl-9869323

ABSTRACT

Abundant evidence, including critical information gathered by Prof. Siegfried Hoyer and his colleagues, indicates that abnormalities of cerebral metabolism are common in neurodegenerative diseases, including Alzheimer's Disease (AD). Alterations in mitochondrial enzymes likely underlie these deficits. Replicable reductions in AD brain occur in the pyruvate dehydrogenase complex (the link of glycolysis to the Kreb's cycle), the alpha-ketoglutarate dehydrogenase complex (KGDHC; the link of Kreb's cycle to glutamate metabolism) and cytochrome oxidase (the link of the Kreb's cycle to oxygen utilization). Available evidence suggests that deficiencies in KGDHC may be genetic in some cases, whereas evidence that the other two enzyme systems have a genetic component is lacking. Additional results indicate that the reductions can also be secondary to other causes including oxidative stress. A variety of data suggest that the mitochondrial insufficiencies contribute significantly to the pathophysiology of AD.


Subject(s)
Alzheimer Disease/enzymology , Mitochondria/enzymology , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Electron Transport Complex IV/metabolism , Humans , Ketoglutarate Dehydrogenase Complex/metabolism , Mitochondria/metabolism , Oxidative Stress/physiology , Pyruvate Dehydrogenase Complex/metabolism
12.
Ann Neurol ; 44(4): 676-81, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9778267

ABSTRACT

Alzheimer's disease (AD) is associated with a striking reduction in the activity of the alpha-ketoglutarate dehydrogenase complex (KGDHC). The deficiency occurs in brains from AD patients of undefined etiology, and in fibroblasts from both sporadic and familial AD cases. To further assess the nature of the abnormality of KGDHC in AD, KGDHC activities and immunoreactivities were analyzed in brains from AD patients bearing the Swedish APP670/671 mutation. This gene defect causes overproduction of the amyloid beta peptide. KGDHC activities were reduced by 55 to 57% compared with control values in the mutation-bearing AD cases in the medial temporal and superior frontal cortices. The immunochemical levels of KGDHC subunits Elk (-51%) and E2k (-76%) declined, whereas E3 concentrations were unchanged. The results suggest that mitochondrial dysfunction is a part of the pathophysiological process in AD even when the primary pathogenic cause is nonmitochondrial.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Ketoglutarate Dehydrogenase Complex/metabolism , Acyltransferases/metabolism , Aged , Glutamate Dehydrogenase/metabolism , Humans , Middle Aged , Tissue Distribution
13.
Dev Neurosci ; 20(4-5): 462-8, 1998.
Article in English | MEDLINE | ID: mdl-9778585

ABSTRACT

Huntington disease (HD) fibroblasts subjected to stress exhibit an enzyme profile that is different from that exhibited by escapee (unaffected members of families with HD) or control fibroblasts. The specific activity of glyceraldehyde 3-phosphate dehydrogenase (GAPDH) in normally cultured HD fibroblasts was not different from that in control and escapee fibroblasts. However, in escapee and control fibroblasts subjected to stress by withholding fresh medium, the specific activity of GAPDH in cells harvested by trypsinization increased greatly 3 weeks after withholding medium ( approximately 8-fold), but the increase was significantly less pronounced ( approximately 3-fold) in the HD fibroblasts. In contrast, only small changes occurred in the specific activity of lipoamide dehydrogenase (LADH) over the same time period, and the values were not significantly different among the three groups at any time point. The specific activity of hexokinase (HK) was significantly higher in the HD fibroblasts at 1-3 weeks after withholding fresh medium than in the escapee/control fibroblasts. Finally, the total yield of fibroblasts per culture flask (as judged by protein content) was significantly greater for the stressed HD fibroblasts than for the escapee and control fibroblasts at 2 and 3 weeks after withholding medium. The present results are in accord with the hypothesis that HD is a disease associated with latent, generalized metabolic abnormalities.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Huntington Disease/enzymology , Huntington Disease/pathology , Peptide Fragments/metabolism , Stress, Physiological/metabolism , Cells, Cultured , Cytological Techniques , Dihydrolipoamide Dehydrogenase/metabolism , Fibroblasts/enzymology , Hexokinase/metabolism , Humans , Phosphofructokinase-1/metabolism , Reference Values , Transglutaminases/metabolism
14.
J Neurochem ; 70(3): 1143-50, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9489735

ABSTRACT

Mitochondrial dysfunction is a common feature of many neurodegenerative disorders. The metabolic encephalopathy caused by thiamine deficiency (TD) is a classic example in which an impairment of cerebral oxidative metabolism leads to selective cell death. In experimental TD in rodents, a reduction in the activity of the thiamine diphosphate-dependent, mitochondrial enzyme alpha-ketoglutarate dehydrogenase complex (KGDHC) occurs before the onset of pathologic lesions and is among the earliest biochemical deficits found. To understand the molecular basis and the significance of the deficiency of KGDHC in TD-induced brain damage, the enzyme activity and protein levels of KGDHC were analyzed. The effect of TD on the subregional/cellular distribution of KGDHC and the anatomic relation of KGDHC with selective cell death were also tested by immunocytochemistry. Consistent with several previous studies, TD dramatically reduced KGDHC activity in both anatomically damaged (thalamus and inferior colliculus) and spared (cerebral cortex) regions. Immunocytochemistry revealed no apparent correlation of regional KGDHC immunoreactivity or its response to TD with affected regions in TD. The basis of the enzymatic and immunocytochemical behavior of KGDHC was further assessed by quantitative immunoblots, using antibodies specific for each of the three KGDHC components. Despite the marked decrease of KGDHC activity in TD, no reduction of any of the three KGDHC protein levels was found. Thus, TD impairs the efficacy of the KGDHC catalytic machinery, whereas the concentration of protein molecules persists. The generalized decline of KGDHC activity with no apparent anatomic selectivity is consistent with the notion that the compromised mitochondrial oxidation sensitizes the brain cells to various other insults that precipitate the cell death. The current TD model provides a relevant experimental system to understand the molecular basis of many neurodegenerative conditions in which mitochondrial dysfunction and KGDHC deficiency are prominent features.


Subject(s)
Brain/enzymology , Ketoglutarate Dehydrogenase Complex/deficiency , Thiamine Deficiency/enzymology , Alcohol Amnestic Disorder/enzymology , Animals , Disease Models, Animal , Immunohistochemistry , Ketoglutarate Dehydrogenase Complex/metabolism , Male , Oxidative Stress/physiology , Rats , Rats, Inbred F344 , Transketolase/metabolism
15.
J Neurochem ; 70(1): 233-41, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9422367

ABSTRACT

The mechanisms of selective neuronal loss after short-term global ischemia remain undefined, but processes including increased proteolytic activity, impaired protein synthesis, and oxidative damage have been proposed to contribute. A decrease in activity of the pyruvate dehydrogenase complex in the dorsolateral striatum, an ischemia-susceptible region, is one change apparently differentiating this region from ischemia-resistant areas during early recirculation. To provide an insight into processes contributing to postischemic cell damage, the changes in the pyruvate dehydrogenase complex during early recirculation have been further characterized. These studies provide clear confirmation that the activity of the pyruvate dehydrogenase complex is reduced in mitochondria from the dorsolateral striatum by 3 h of recirculation. The decrease in activity was not accompanied by a loss of antigenic sites or by changes in electrophoretic mobility of the components of the complex. A reduction in activity of the E1 component of the complex (39-42% decrease), but not the E2 and E3 components, was observed that was apparently sufficient to explain the decrease in activity of the whole complex. These results indicate that the changes in activity of the pyruvate dehydrogenase complex in the dorsolateral striatum are not due to loss or gross disruption of the constituent proteins but rather most likely reflect a selective inactivation of a specific component of the complex.


Subject(s)
Ischemic Attack, Transient/enzymology , Pyruvate Dehydrogenase Complex/metabolism , Reperfusion , Animals , Cerebral Cortex/enzymology , Corpus Striatum/enzymology , Enzyme Activation , Isoenzymes/metabolism , Male , Mitochondria/enzymology , Rats , Rats, Inbred Strains , Time Factors
16.
Age (Omaha) ; 21(1): 25-30, 1998 Jan.
Article in English | MEDLINE | ID: mdl-23604331

ABSTRACT

At least seven adult-onset neurodegenerative diseases, including Huntington's disease (HD), are caused by genes containing expanded CAG triplets within their coding regions. The expanded CAG repeats give rise to extended stretches of polyglutamines (Qn) in the proteins expressed by the affected genes. Generally, n ≥40 in affected individuals and ≤36 in clinically unaffected individuals. The expansion has been proposed to confer a "toxic gain of function" to the mutated protein. Poly-Q domains have recently been shown to be excellent substrates of tissue transglutaminase. We investigated the effects of expression of glutathione S-transferase constructs containing poly-Q inserts of various lengths (GSTQn where n = 0, 10, 62 or 81) on the activity of some key metabolic enzymes in the host Escherischia coil-an organism not known to have transglutaminase activity. E. coil carrying the GSTQ62 construct exhibited statistically significant decreases in the specific activities of α-ketoglutarate dehydrogenase complex (KGDHC) and pyruvate dehydrogenase complex (PDHC). Previous work has shown that KGDHC and PDHC activities are reduced in the brains of Alzheimer's disease (AD) patients. Our results suggest that KGDHC and PDHC may be particularly susceptible to the effects of a number of disparate insults, including those associated with AD and HD.

17.
Ann N Y Acad Sci ; 826: 382-5, 1997 Sep 26.
Article in English | MEDLINE | ID: mdl-9329708

ABSTRACT

Extensive studies over the last 20 years have documented the existence of inherent abnormalities in oxidative/energy metabolism in Alzheimer's disease (AD). These abnormalities can be linked to characteristics of AD by plausible pathophysiological mechanisms for which there is abundant, robust evidence. The inherent abnormalities in cerebral metabolism of oxygen and glucose can reasonably be expected to interact synergistically with vascular compromise of cerebral oxygen and glucose metabolism in causing brain damage in AD.


Subject(s)
Alzheimer Disease/metabolism , Cerebrovascular Circulation , Oxygen/metabolism , Alzheimer Disease/pathology , Brain/metabolism , Energy Metabolism , Glucose/metabolism , Humans , In Vitro Techniques
18.
Ann N Y Acad Sci ; 826: 516-9, 1997 Sep 26.
Article in English | MEDLINE | ID: mdl-9329737

ABSTRACT

These results demonstrate that early alterations in the BBB may underlie selective vulnerability in this model of chronic reduced oxidative metabolism. Changes in the BBB (IgG extravasation) precede alterations in APP processing and cell death. Since thiamine-dependent enzymes are also reduced in the brain in Alzheimer's disease, similar processes may be important in the pathophysiology of the disease.


Subject(s)
Cerebrovascular Circulation , Nerve Degeneration/pathology , Thiamine Deficiency/pathology , Amyloid beta-Protein Precursor/metabolism , Blood-Brain Barrier , Humans , Pyrophosphatases/metabolism
19.
J Neurochem ; 69(1): 431-4, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9202340

ABSTRACT

Huntington's disease and six other neurodegenerative diseases are associated with abnormal gene products containing expanded polyglutamine (poly-Q; Qn) domains (n > or = 40). In the present work, we show that glutathione S-transferase (GST) fusion proteins containing a small, physiological-length poly-Q domain (GSTQ10) or a large, pathological-length poly-Q domain (GSTQ62) are excellent substrates of guinea pig liver (tissue) transglutaminase and that both GSTQ10 and GSTQ62 are activators of tissue transglutaminase-catalyzed hydroxaminolysis of N-alpha-carbobenzoxyglutaminylglycine. The present findings have implications for understanding the pathophysiological mechanisms of expanded CAG/poly-Q domain diseases.


Subject(s)
Nerve Degeneration/physiology , Peptides/chemistry , Repetitive Sequences, Nucleic Acid , Transglutaminases/metabolism , Amino Acid Sequence , Animals , Brain/cytology , Brain/enzymology , Guinea Pigs , Liver/enzymology , Peptides/genetics , Protein Structure, Tertiary , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...