Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
1.
Clin Nutr ESPEN ; 25: 8-17, 2018 06.
Article in English | MEDLINE | ID: mdl-29779823

ABSTRACT

The WHO 2016 report indicates that worldwide obesity is rising, with over 600 million people in the obese range (BMI>30). The recommended daily calorie intake for adults is 2000 kcal and 2500 kcal for women and men respectively. The average American consumes 3770 kcal/day and the average person in the UK consumes 3400 kcal/day. With such increased caloric intake, there is an increased load on metabolic pathways, in particular glucose metabolism. Such metabolism requires micronutrients as enzyme co-factors. The recommended daily allowance (RDA) for thiamine is 1.3 mg/day and 0.5 mg thiamine is required to process 1000 kilocalories (kcal). Therefore, despite the appearance of being overfed, there is now increasing evidence that the obese population may nutritionally depleted of essential micronutrients. Thiamine deficiency has been reported to be in the region of 16-47% among patients undergoing bariatric surgery for obesity. Thiamine, in turn, requires magnesium to be in its active form thiamine diphosphate, (TDP). TDP also requires magnesium to achieve activation of TDP dependent enzymes, including transketolase (TK), pyruvate dehydrogenase (PDH) and alpha-keto glutaric acid dehydrogenase (AKGDH), during metabolism of glucose. Thiamine and magnesium therefore play a critical role in glucose metabolism and their deficiency may result in the accumulation of anaerobic metabolites including lactate due to a mismatch between caloric burden and function of thiamine dependent enzymes. It may therefore be postulated that thiamine and magnesium deficiency are under-recognized in obesity and may be important in the progress of obesity and obesity related chronic disease states. The aim of the present systematic review was to examine the role of thiamine dependent enzymes in obesity and obesity related chronic disease states.


Subject(s)
Nutritional Status , Obesity/enzymology , Recommended Dietary Allowances , Thiamine Deficiency/enzymology , Thiamine/administration & dosage , Body Mass Index , Chronic Disease , Energy Intake , Glucose/metabolism , Humans , Magnesium/administration & dosage , Magnesium Deficiency/enzymology , Magnesium Deficiency/epidemiology , Magnesium Deficiency/physiopathology , Obesity/epidemiology , Obesity/physiopathology , Prevalence , Prognosis , Risk Factors , Thiamine/adverse effects , Thiamine/metabolism , Thiamine Deficiency/epidemiology , Thiamine Deficiency/physiopathology
2.
Biomed Res Int ; 2014: 572915, 2014.
Article in English | MEDLINE | ID: mdl-25028661

ABSTRACT

In animal experiments, hippocampal neurogenesis and the activity of thiamine-dependent transketolase decrease markedly under conditions of thiamine deficiency. To further investigate the effect of thiamine deficiency on the proliferation of hippocampal progenitor cells (HPCs) and the potential mechanisms involved in this effect, we cultured HPCs in vitro in the absence of thiamine and found that proliferation and transketolase activity were both significantly repressed. Furthermore, specific inhibition of transketolase activity by oxythiamine strongly inhibited HPC proliferation in a dose-dependent manner. However, thiamine deficiency itself inhibited the proliferation to a greater degree than did oxythiamine. Taken together, our results suggest that modulation of transketolase activity might be one of the mechanisms by which thiamine regulates the proliferation of hippocampal progenitor cells.


Subject(s)
Cell Proliferation , Down-Regulation , Gene Expression Regulation, Enzymologic , Hippocampus/enzymology , Neural Stem Cells/enzymology , Thiamine Deficiency/enzymology , Transketolase/biosynthesis , Animals , Antimetabolites/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Hippocampus/pathology , Neural Stem Cells/pathology , Oxythiamine/pharmacology , Rats , Rats, Sprague-Dawley , Thiamine Deficiency/pathology
3.
Am J Alzheimers Dis Other Demen ; 26(8): 588-98, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22218733

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia in elderly individuals and is associated with progressive neurodegeneration of the human neocortex. Thiamine levels and the activity of thiamine-dependent enzymes are reduced in the brains and peripheral tissues of patients with AD. Genetic studies have provided the opportunity to determine what proteins link thiamine to AD pathology (ie, transketolase, apolipoprotein E, α-1-antitrypsin, pyruvate dehydrogenase complex, p53, glycogen synthetase kinase-3ß, c-Fos gene, the Sp1 promoter gene, and the poly(ADP-ribosyl) polymerase-1 gene). We reviewed the association between histopathogenesis and neurotransmitters to understand the relationship between thiamine and AD pathology. Oral thiamine trials have been shown to improve the cognitive function of patients with AD; however, absorption of thiamine is poor in elderly individuals. In the early stage of thiamine-deficient encephalopathy (Wernicke's encephalopathy), however, parental thiamine has been used successfully. Therefore, further studies are needed to determine the benefits of using parental thiamine as a treatment for AD.


Subject(s)
Alzheimer Disease/etiology , Thiamine Deficiency/complications , Thiamine Pyrophosphate/metabolism , Thiamine/metabolism , Aged , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Brain/enzymology , Brain/metabolism , Humans , Pyruvate Dehydrogenase Complex/metabolism , Thiamine Deficiency/enzymology , Transketolase/metabolism
4.
Neurochem Int ; 57(7): 851-6, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20850489

ABSTRACT

Several pathologic conditions are known to cause thiamine deficiency, which induce energy shortages in all tissues, due to impairment of pyruvate decarboxylation. Brain is particularly susceptible to these conditions due to its high rate of glucose to pyruvate-driven energy metabolism. However, cellular compartmentalization of a key energy metabolite, acetyl-CoA, in this pathology remains unknown. Pyrithiamine-evoked thiamine deficiency caused no significant alteration in pyruvate dehydrogenase and 30% inhibition of α-ketoglutarate dehydrogenase activities in rat whole forebrain mitochondria. It also caused 50% reduction of the metabolic flux of pyruvate through pyruvate dehydrogenase, 78% inhibition of its flux through α-ketoglutarate dehydrogenase steps, and nearly 60% decrease of intramitochondrial acetyl-CoA content, irrespective of the metabolic state. State 3 caused a decrease in citrate and an increase in α-ketoglutarate accumulation. These alterations were more evident in thiamine-deficient mitochondria. Simultaneously thiamine deficiency caused no alteration of relative, state 3-induced increases in metabolic fluxes through pyruvate and α-ketoglutarate dehydrogenase steps. These data indicate that a shortage of acetyl-CoA in the mitochondrial compartment may be a primary signal inducing impairment of neuronal and glial cell functions and viability in the thiamine-deficient brain.


Subject(s)
Acetyl Coenzyme A/deficiency , Brain/enzymology , Mitochondria/enzymology , Thiamine Deficiency/enzymology , Wernicke Encephalopathy/enzymology , Animals , Brain/pathology , Disease Models, Animal , Enzyme Activation/physiology , Humans , Male , Rats , Rats, Wistar , Thiamine Deficiency/pathology , Wernicke Encephalopathy/pathology
6.
Neurochem Int ; 55(5): 275-81, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19576514

ABSTRACT

Wernicke's encephalopathy (WE) in humans is a metabolic disorder caused by thiamine deficiency (TD). In both humans and experimental animals, TD leads to selective neuronal cell death in diencephalic and brainstem structures. Neuropathologic features of WE include petechial hemorrhagic lesions, and blood-brain barrier (BBB) breakdown has been suggested to play an important role in the pathogenesis of TD. The goal of the present study was to examine expression of the tight junction (TJ) protein occludin, its associated scaffolding proteins zona occludens (ZO-1 and ZO-2), and to measure matrix metalloproteinase (MMP) levels as a function of regional BBB permeability changes in thiamine-deficient mice. TD was induced in 12-week-old male C57Bl/6 mice by feeding a thiamine-deficient diet and administration of the central thiamine antagonist pyrithiamine. BBB permeability was measured by IgG extravasation; expression of occludin, ZO-1 and ZO-2 was measured by Western blot analysis and RT-PCR, structural integrity of the BBB was assessed using occludin and ZO-1 immunostaining, and MMPs levels were measured by gelatin zymography and immunohistochemistry. Studies were performed in vulnerable (medial thalamus) versus spared (frontal cortex) regions of the brain. Hemorrhagic lesions, selective increases in brain IgG extravasation, a concomitant loss in protein expression of occludin, ZO-1 and ZO-2, as well as decreased and disrupted patterns of occludin and ZO-1 immunostaining were observed in the medial thalamus of thiamine-deficient mice. MMP-9 levels were also selectively increased in the medial thalamus of these animals, and were found to be localized in the vascular endothelium, as well as in cells with an apparent polymorphonuclear morphology. No changes of TJ gene expression were observed. These results indicate that alterations in TJ proteins occur in TD, and offer a plausible explanation for the selective increase in BBB permeability in thiamine-deficient animals. They also suggest a role for MMP-9 in the initiation of changes to BBB integrity in TD.


Subject(s)
Brain/metabolism , Matrix Metalloproteinases/metabolism , Membrane Proteins/metabolism , Thiamine Deficiency/metabolism , Tight Junctions/metabolism , Animals , Base Sequence , Blood-Brain Barrier , Brain/enzymology , DNA Primers , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Thiamine Deficiency/enzymology , Up-Regulation
7.
Int J Biochem Cell Biol ; 41(4): 717-20, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18490188

ABSTRACT

Thiamin, or vitamin B1, is crucial for brain function. In its active form, thiamin pyrophosphate (TPP), it is a co-enzyme for several enzymes, including transketolase. Transketolase is an important enzyme in the non-oxidative branch of the pentose phosphate pathway (PPP), a pathway responsible for generating reducing equivalents, which is essential for energy transduction and for generating ribose for nucleic acid synthesis. Transketolase also links the PPP to glycolysis, allowing a cell to adapt to a variety of energy needs, depending on its environment. Abnormal transketolase expression and/or activity have been implicated in a number of diseases where thiamin availability is low, including Wernicke-Korsakoff's Syndrome and alcoholism. Yet, the precise mechanism by which this enzyme is involved in the pathophysiology of these disorders remains controversial.


Subject(s)
Alcoholism/enzymology , Brain Damage, Chronic/enzymology , Transketolase/metabolism , Brain Damage, Chronic/chemically induced , Humans , Korsakoff Syndrome/enzymology , Thiamine Deficiency/enzymology , Thiamine Pyrophosphate/deficiency
8.
J Aquat Anim Health ; 20(1): 63-71, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18536504

ABSTRACT

No consistent explanation has been found for the variability in the thiaminase activity of alewives Alosa pseudoharengus despite the role of alewife thiaminase in large-scale salmonine mortality in the Laurentian Great Lakes. We conducted experiments to evaluate the effect of two stressors, reduced salt content in the water and food limitation, on alewife thiaminase activity. Alewives were subjected to treatments in replicated tanks in which conductivity was lowered (< 100 microS/cm) for 8 d and feeding was limited for 39 d. Circulating white blood cells, plasma cortisol, plasma glucose, and whole-body thiaminase were measured in individual alewives to assess their response to these experimental treatments. Alewives from the controls had significantly larger numbers of circulating white blood cells than those in the salt-reduced and food-limited treatments (24,000 and 19,000 cells/microL and 11,000 and 9,000 cells/microL for alewives from the two control and salt-reduced treatment tanks, respectively, and 34,000 and 30,000 cells/microL and 21,000 and 16,000 cells/microL for alewives from the two control and food-limited treatment tanks). No significant differences in alewife thiaminase activity were found between treatment fish and their controls. The mean thiaminase activity in the alewives studied increased from 6,900 to 16,000 pmol x g(-1) x min(-1) from the time of their collection in Cayuga Lake to the start of laboratory experiments 1.5-2.5 years later; the latter value was more than twice that of previously reported levels of thiaminase activity from alewives collected in the wild. These data suggest that the variability in alewife thiaminase is not related to stress from salt reduction or food limitation, but laboratory holding conditions significantly increased thiaminase through a mechanism not evaluated by our experimental treatments.


Subject(s)
Animal Husbandry , Fish Diseases/enzymology , Hydrolases/metabolism , Thiamine Deficiency/veterinary , Animals , Blood Glucose , Fish Diseases/blood , Fish Diseases/physiopathology , Fishes , Food Deprivation , Fresh Water , Hydrocortisone/blood , Hydrolases/blood , Leukocyte Count/veterinary , Stress, Physiological , Thiamine Deficiency/enzymology
9.
Metab Brain Dis ; 23(2): 175-87, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18481165

ABSTRACT

Thiamine deficiency (TD) in both humans and experimental animals results in severe mitochondrial dysfunction and leads to selective neuronal cell death in diencephalic and cerebellar structures. We have investigated cyclooxygenase-2 (COX-2) expression in vulnerable (medial thalamus, inferior colliculus) and spared (frontal cortex) regions of rats with thiamine deficiency. Expression of COX-2 mRNA was selectively increased (twofold, p < 0.001) in vulnerable regions at symptomatic stages of encephalopathy (14 days) of TD compared to pair-fed controls or presymptomatic (days 12) rats. Induction of COX-2 expression was accompanied by a significant increase (two- to threefold, p < 0.001) in prostanglandin E2 (PGE2) synthesis in vulnerable regions at symptomatic stages of TD. COX-2 immunolabeling revealed a neuronal localization and COX-2 immunoreactive neurons were significantly increased at symptomatic stages of encephalopathy. Administration of nimesulide, a highly specific COX-2 inhibitor, significantly reduced PGE-2 levels in vulnerable regions but, rather than being neuroprotective, precipitated encephalopathy and exacerbated neuronal cell death due to TD. These findings suggest that newly synthesized prostanoids exert a neuroprotective role in TD.


Subject(s)
Brain/enzymology , Cyclooxygenase 2/metabolism , Neurons/enzymology , Sulfonamides/adverse effects , Thiamine Deficiency/enzymology , Wernicke Encephalopathy/enzymology , Animals , Brain/drug effects , Brain/physiopathology , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/genetics , Cytoprotection/drug effects , Cytoprotection/physiology , Dinoprostone/biosynthesis , Disease Models, Animal , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/adverse effects , Immunohistochemistry , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/enzymology , Nerve Degeneration/physiopathology , Neurons/drug effects , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Thiamine Deficiency/physiopathology , Up-Regulation/drug effects , Up-Regulation/physiology , Wernicke Encephalopathy/chemically induced , Wernicke Encephalopathy/physiopathology
10.
Neurochem Int ; 50(7-8): 921-31, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17482317

ABSTRACT

Thiamine-dependent enzymes are diminished in multiple neurodegenerative diseases. Thiamine deficiency (TD) reduces the activity of thiamine dependent-enzymes [e.g., the alpha-ketoglutarate dehydrogenase complex (KGDHC)], induces regional selective neurodegeneration and serves as a model of a mild impairment of oxidative metabolism. The current experiments tested whether changes in KGDHC protein subunits (E1k, E2k and E3) or activity or message levels underlie the selective loss of neurons in particular brain regions. Thus, TD-induced changes in these variables in the brain region most vulnerable to TD [the sub-medial thalamic nucleus (SmTN)] were compared to those in a region that is relatively resistant to TD (cortex) at stages of TD when the neuron loss in SmTN is not present, minimal or severe. Impaired motor performance on rotarod was apparent by 8 days of TD (-32%) and was severe by 10 days of TD (-97%). At TD10, the overall KGDHC activity measured by an in situ histochemical staining method declined 52% in SmTN but only 20% in cortex. Reductions in the E2k and E3 mRNA in SmTN occurred as early as TD6 (-28 and -18%, respectively) and were more severe by TD10 (-61 and -66%, respectively). On the other hand, the level of E1k mRNA did not decline in SmTN until TD10 (-48%). In contrast, TD did not alter mRNA levels of the subunits in cortex at late stages. Western blots and immunocytochemistry revealed different aspects of the changes in protein levels. In SmTN, the immunoreactivity of E1k and E3 by Western blotting increased 34 and 40%, respectively, only at TD8. In cortex, the immunoreactivity of the three subunits was not altered. Immunocytochemical staining of brain sections from TD10 mice indicated a reduction in the immunoreactivity of all subunits in SmTN, but not in cortex. These findings demonstrate that the response of the KGDHC activity, mRNA and immunoreactivity of E1k, E2k and E3 to TD is region and time dependent. Loss of KGDHC activity in cortex is likely related to post-translational modification rather than a loss of protein, whereas in SmTN transcriptional and post-translational modifications may account for diminished KGDHC activity. Moreover, the earlier detection in TD induced-changes of the transcripts of KGDHC indicates that transcriptional modification of the two subunits (E2k and E3) of KGDHC may be one of the early events in the cascade leading to selective neuronal death.


Subject(s)
Brain/enzymology , Ketoglutarate Dehydrogenase Complex/metabolism , Mitochondria/enzymology , Psychomotor Performance/physiology , Thiamine Deficiency/enzymology , Animals , Disease Models, Animal , Ketoglutarate Dehydrogenase Complex/genetics , Male , Mice , Mice, Inbred C57BL , RNA/genetics , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
11.
Article in English | MEDLINE | ID: mdl-17120961

ABSTRACT

A study was carried out to determine whether thiamine deficiency in northeast Thailand is linked with parasitic infection. We assessed the thiamine status and tested for parasitic infections in 231 children between 6 and 12 years of age in Nam Phong District of Khon Kaen Province, Thailand. Thiamine deficiency [thiamine pyrophosphate effect (TPPE) > 20%] and parasitic infections (mostly Strongyloides stercoralis) were identified in 16 and 45% of the children, respectively. No association between thiamine deficiency and parasitic infection was found. The general health of the children, according to Thai standard anthropometric curves, was within the normal range. The suboptimal thiamine status and the high prevalence of parasitic infection require attention.


Subject(s)
Parasitic Diseases/complications , Rural Health , Thiamine Deficiency/complications , Child , Female , Humans , Male , Nutritional Status , Parasitic Diseases/epidemiology , Prevalence , Thailand/epidemiology , Thiamine Deficiency/enzymology
12.
FEBS Lett ; 579(25): 5596-602, 2005 Oct 24.
Article in English | MEDLINE | ID: mdl-16214141

ABSTRACT

We hypothesized that in marginal thiamin deficiency intracellular alpha-oxoaldehydes form macromolecular adducts that could possibly be genotoxic in colon cells; and that in the presence of oxidative stress these effects are augmented because of decreased detoxification of these aldehydes. We have demonstrated that reduced dietary thiamin in F344 rats decreased transketolase activity and increased alpha-oxoaldehyde adduct levels. The methylglyoxal protein adduct level was not affected by oral glyoxal or methylglyoxal in the animals receiving thiamin at the control levels but was markedly increased in the animals on a thiamin-reduced diet. These observations are consistent with our suggestion that the induction of aberrant crypt foci with marginally thiamin-deficient diets may be a consequence of the formation of methylglyoxal adducts.


Subject(s)
Aldehydes/blood , Blood Proteins/analysis , Oxidative Stress , Thiamine Deficiency/enzymology , Transketolase/metabolism , Aldehydes/chemistry , Animals , Blood Proteins/chemistry , Blood Proteins/drug effects , Down-Regulation , Glyoxal/blood , Glyoxal/pharmacology , Male , Pyruvaldehyde/blood , Pyruvaldehyde/pharmacology , Rats , Rats, Inbred F344 , Thiamine/pharmacology
13.
Neurochem Int ; 45(7): 1021-8, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15337301

ABSTRACT

Thiamine (Vitamin B1) deficiency (TD) leads to memory deficits and neurological disease in animals and humans. The thiamine-dependent enzymes of the tricarboxylic acid (TCA) cycle are reduced following TD and in the brains of patients that died from multiple neurodegenerative diseases. Whether reductions in thiamine or thiamine-dependent enzymes leads to changes in all TCA cycle enzymes has never been tested. In the current studies, the pyruvate dehydrogenase complex (PDHC) and all of enzymes of the TCA cycle were measured in the brains of TD mice. Non-thiamine-dependent enzymes such as succinate dehydrogenase (SDH), succinate thiokinase (STH) and malate dehydrogenase (MDH) were altered as much or more than thiamine-dependent enzymes such as the alpha-ketoglutarate dehydrogenase complex (KGDHC) (-21.5%) and PDHC (-10.5%). Succinate dehydrogenase (SDH) activity decreased by 27% and succinate thiokinase (STH) decreased by 24%. The reductions in these other enzymes may result from oxidative stress because of TD or because these other enzymes of the TCA cycle are part of a metabolon that respond as a group of enzymes. The results suggest that other TCA cycle enzymes should be measured in brains from patients that died from neurological disease in which thiamine-dependent enzymes are known to be reduced. The diminished activities of multiple TCA cycle enzymes may be important in our understanding of how metabolic lesions alter brain function in neurodegenerative disorders.


Subject(s)
Brain/enzymology , Citric Acid Cycle/physiology , Thiamine Deficiency/enzymology , Animals , Mice , Mice, Inbred C57BL
14.
Neurochem Int ; 45(1): 49-56, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15082221

ABSTRACT

Thiamine deficiency results in selective neuronal cell death in thalamic structures. Previous studies provide evidence for a role implicating nitric oxide (NO) in the pathogenesis of cell death due to thiamine deficiency. In order to ascertain the origin of increased NO in the thiamine deficient brain, expression of endothelial nitric oxide synthase isoform (eNOS), was measured in the medial thalamus and in the inferior colliculus and compared to the frontal cortex (a spared region) of rats in which thiamine deficiency was induced through a feeding protocol of thiamine-deficient diet concomitant with daily administration of pyrithiamine, a central thiamine antagonist. eNOS mRNA and protein expression were significantly increased as a function of the severity of neurological impairment and the degree of neuronal cell loss in the medial thalamus and in the inferior colliculus. These findings suggest that the vascular endothelium is a major site of NO production in the brain in thiamine deficiency and that eNOS-derived NO could account for the selective damage to the thalamic structures that are observed in this particular disorder.


Subject(s)
Brain/enzymology , Gene Expression Regulation, Enzymologic/physiology , Nitric Oxide Synthase/biosynthesis , Thiamine Deficiency/enzymology , Animals , Male , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type III , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Thiamine Deficiency/genetics
15.
J Neuropathol Exp Neurol ; 62(2): 195-207, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12578229

ABSTRACT

Neurodegenerative diseases are characterized by abnormalities in oxidative processes, region-selective neuron loss, and diminished thiamine-dependent enzymes. Thiamine deficiency (TD) diminishes thiamine dependent enzymes, alters mitochondrial function, impairs oxidative metabolism, and causes selective neuronal death. In mice, the time course of TD-induced changes in neurons and microglia were determined in the brain region most sensitive to TD. Significant neuron loss (29%) occurred after 8 or 9 days of TD (TD8-9) and increased to 90% neuron loss by TD10-11. The number of microglia increased 16% by TD8 and by nearly 400% on TD11. Hemeoxygenase-1 (HO-1)-positive microglia were not detectable at TD8, yet increased dramatically coincident with neuron loss. To test the duration of TD critical for irrevocable changes, mice received thiamine after various durations of TD. Thiamine administration on TD8 blocked further neuronal loss and induction of HO-1-positive microglia, whereas other microglial changes persisted. Thiamine only partially reversed effects on TD9, and was ineffective on TD10-11. These studies indicate that irreversible steps leading to neuronal death and induction of HO-1-positive microglia occur on TD9. The results indicate that TD induces alterations in neurons. endothelial cells, and microglia contemporaneously. This model provides a unique paradigm for elucidating the molecular mechanisms involved in neuronal commitment to neuronal death cascades and contributory microglial activity.


Subject(s)
Brain/enzymology , Cell Death/physiology , Neurodegenerative Diseases/enzymology , Recovery of Function/physiology , Thiamine Deficiency/enzymology , Thiamine/metabolism , Animals , Brain/pathology , Brain/physiopathology , Cell Death/drug effects , Disease Models, Animal , Fluoresceins , Fluorescent Dyes/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1 , Immunohistochemistry , Male , Membrane Proteins , Mice , Microglia/drug effects , Microglia/enzymology , Microglia/pathology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/physiopathology , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Organic Chemicals , Recovery of Function/drug effects , Thiamine/pharmacology , Thiamine Deficiency/drug therapy , Thiamine Deficiency/physiopathology
16.
J Neurochem ; 74(1): 114-24, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10617112

ABSTRACT

Abnormalities in oxidative metabolism and inflammation accompany many neurodegenerative diseases. Thiamine deficiency (TD) is an animal model in which chronic oxidative stress and inflammation lead to selective neuronal death, whereas other cell types show an inflammatory response. Therefore, the current studies determined the response of different brain cell types to TD and/or inflammation in vitro and tested whether their responses reflect inherent properties of the cells. The cells that have been implicated in TD-induced neurotoxicity, including neurons, microglia, astrocytes, and brain endothelial cells, as well as neuroblastoma and BV-2 microglial cell lines, were cultured in either thiamine-depleted media or in normal culture media with amprolium, a thiamine transport inhibitor. The activity levels of a key mitochondrial enzyme, alpha-ketoglutarate dehydrogenase complex (KGDHC), were uniquely distributed among different cell types: The highest activity was in the endothelial cells, and the lowest was in primary microglia and neurons. The unique distribution of the activity did not account for the selective response to TD. TD slightly inhibited general cellular dehydrogenases in all cell types, whereas it significantly reduced the activity of KGDHC exclusively in primary neurons and neuroblastoma cells. Among the cell types tested, only in neurons did TD induce apoptosis and cause the accumulation of 4-hydroxy-2-nonenal, a lipid peroxidation product. On the other hand, chronic lipopolysaccharide-induced inflammation significantly inhibited cellular dehydrogenase and KGDHC activities in microglia and astrocytes but not in neurons or endothelial cells. The results demonstrate that the selective cell changes during TD in vivo reflect inherent properties of the different brain cell types.


Subject(s)
Brain/metabolism , Neurons/physiology , Oxidative Stress/physiology , Aldehydes/metabolism , Amprolium , Animals , Apoptosis , Brain/cytology , Cell Death/physiology , Cells, Cultured , Coloring Agents , Ketoglutarate Dehydrogenase Complex/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Neurons/classification , Neurons/enzymology , Neurons/metabolism , Oxidoreductases/metabolism , Tetrazolium Salts/pharmacokinetics , Thiamine Deficiency/chemically induced , Thiamine Deficiency/enzymology , Thiazoles/pharmacokinetics
17.
Toxicology ; 133(2-3): 105-13, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10378477

ABSTRACT

Many neurological disorders that occur frequently in lead intoxicated animals, have also been observed in thiamine deficient animals. To test whether lead intoxication could decrease the thiamine status and thresholds of electroshock seizure in rats, 3-week-old Wistar rats were treated with lead or lead plus thiamine. For comparison, a thiamine deficient group was included. Thiamine contents and transketolase activity, one of the thiamine dependent enzymes in the brain regions were significantly lowered by lead intoxication and thiamine deficiency. In both cases, thresholds of the electroshock seizure were significantly decreased. Thiamine supplementation reversed these signs and decreased the brain lead concentration in the lead treated group. The results from the present study suggest that the increased seizure susceptibility induced by lead intoxication in rats may be mediated at least in part through the changes of thiamine status.


Subject(s)
Brain/drug effects , Brain/metabolism , Lead/toxicity , Seizures/etiology , Thiamine Deficiency/chemically induced , Thiamine Deficiency/physiopathology , Animals , Brain/enzymology , Dose-Response Relationship, Drug , Electroshock , Female , Lead/pharmacokinetics , Rats , Rats, Wistar , Seizures/chemically induced , Seizures/enzymology , Thiamine/pharmacology , Thiamine Deficiency/enzymology , Transketolase/metabolism
18.
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
19.
Alcohol Clin Exp Res ; 21(4): 576-80, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9194907

ABSTRACT

Thiamine deficiency, a frequent complication of alcoholism, contributes significantly to the development of damage in various organ systems, including the brain. The molecular mechanisms that underlie the differential vulnerabilities to thiamine deficiency of tissue and cell types and among individuals are not understood. Investigations into these mechanisms have examined potential variations in thiamine utilizing enzymes. Transketolase is a homodimeric enzyme containing two molecules of noncovalently bound thiamine pyrophosphate. In the present study, we examined a his-tagged human transketolase that was produced in and purified from Escherichia coli cells. Previous findings demonstrated that purified his-transketolase had a Km app for cofactor and a thiamine pyrophosphate-dependent lag period for attaining steady-state kinetics that was similar to transketolase purified from human tissues. Interestingly, the time of the lag period, which is normally independent of enzyme concentration, was found herein to be dependent on the concentration of the recombinant protein. This atypical behavior was due to production in E. coli. Generation of the normal, enzyme concentration-independent state required a cytosolic factor(s) derived from human cells. Importantly, the required factor(s) was found to be defective in a Wernicke-Korsakoff patient whose cells in culture show an enhanced sensitivity to thiamine deficiency.


Subject(s)
Alcohol Amnestic Disorder/enzymology , Thiamine Deficiency/enzymology , Transketolase/chemistry , Wernicke Encephalopathy/enzymology , Adult , Cytosol/physiology , Dimerization , Escherichia coli/enzymology , Humans , Protein Structure, Secondary , Structure-Activity Relationship , Thiamine Pyrophosphate/metabolism , Transketolase/isolation & purification , Transketolase/metabolism
20.
Clin Chim Acta ; 255(1): 13-25, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8930410

ABSTRACT

Pyrithiamine-induced thiamin deficiency has been used in rat as an experimental form of Wernicke-Korsakoff encephalopathy, a disease associated with chronic alcoholism. Although the main etiological factor is known to be the lack of thiamin, the biochemical mechanisms involved in the pathogenesis remain unclear. Thiamin-dependent enzymes were studied in brain mitochondria: alpha-ketoglutarate dehydrogenase activity exhibited 40% reduction, whereas pyruvate dehydrogenase did not change significantly. Polarographic recordings of mitochondrial respiration revealed a decreased State 3, when using pyruvate/malate, alpha-ketoglutarate or glutamine as a substrate, but the respiration rates remained unchanged with glutamate or succinate. This fall in pyruvate oxidation may be due to the impairment of alpha-ketoglutarate dehydrogenase, which follows pyruvate dehydrogenase in the metabolic pathway. A time course of lactate concentration showed dramatic increases in thalamus, mid brain, hypothalamus and colliculli, consistent with the anatomopathological findings. No increases were found before the onset of neurological symptoms.


Subject(s)
Alcohol Amnestic Disorder/etiology , Brain/enzymology , Ketoglutarate Dehydrogenase Complex/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Pyruvic Acid/metabolism , Thiamine Deficiency/enzymology , Wernicke Encephalopathy/etiology , Animals , Brain Chemistry , Female , Hypothalamus/chemistry , Lactic Acid/metabolism , Mesencephalon/chemistry , Mitochondria/enzymology , Oxygen Consumption , Pyrithiamine , Rats , Rats, Sprague-Dawley , Substrate Specificity , Thalamus/chemistry , Thiamine Deficiency/chemically induced , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...