Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Front Neurosci ; 9: 23, 2015.
Article in English | MEDLINE | ID: mdl-25709564

ABSTRACT

Glucose is the main energy substrate for the brain. There is now extensive evidence indicating that the metabolic profile of neural cells with regard to glucose utilization and glycolysis rate is not homogenous, with a marked propensity for glycolytic glucose processing in astrocytes compared to neurons. Methylglyoxal, a highly reactive dicarbonyl compound, is inevitably formed as a by-product of glycolysis. Methylglyoxal is a major cell-permeant precursor of advanced glycation end-products (AGEs), which are associated with several pathologies including diabetes, aging and neurodegenerative diseases. In normal situations, cells are protected against methylglyoxal toxicity by different mechanisms and in particular the glyoxalase system, which represents the most important pathway for the detoxification of methylglyoxal. While the neurotoxic effects of methylglyoxal and AGEs are well characterized, our understanding the glyoxalase system in the brain is more scattered. Considering the high energy requirements (i.e., glucose) of the brain, one should expect that the cerebral glyoxalase system is adequately fitted to handle methylglyoxal toxicity. This review focuses on our actual knowledge on the cellular aspects of the glyoxalase system in brain cells, in particular with regard to its activity in astrocytes and neurons. A main emerging concept is that these two neural cell types have different and energetically adapted glyoxalase defense mechanisms which may serve as protective mechanism against methylglyoxal-induced cellular damage.

2.
Liver Int ; 34(3): 353-61, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23910048

ABSTRACT

BACKGROUND & AIMS: Acute liver failure (ALF) is frequently complicated by infection leading to precipitation of central nervous system complications such as hepatic encephalopathy (HE) and increased mortality. There is evidence to suggest that when infection occurs in ALF patients, the resulting pro-inflammatory mechanisms may be amplified that could, in turn, have a major impact on blood-brain barrier (BBB) function. The aim of this study was to investigate the role of endotoxemia on the progression of encephalopathy in relation to BBB permeability during ALF. METHODS: Adult male C57-BL6 mice with ALF resulting from azoxymethane-induced toxic liver injury were administered trace amounts of the endotoxin component lipopolysaccharide (LPS). Effects on the magnitude of the systemic inflammatory response, liver pathology and BBB integrity were measured as a function of progression of HE, defined as time to loss of corneal reflex (coma). RESULTS: Lipopolysaccharide caused additional two- to seven-fold (P < 0.001) increases in circulating pro-inflammatory cytokines (TNF-α, IL-1ß, IL-6), worsening liver pathology and associated increases of circulating transaminases as well as increased hyperammonaemia consistent with a further loss of viable hepatocytes. LPS treatment of ALF mice led to a rapid precipitation of hepatic coma and the BBB became permeable to the 25-kDa protein immunoglobulin G (IgG). This extravasation of IgG was accompanied by ignificant up-regulation of matrix metalloproteinase-9 (MMP-9), an endopeptidase known to modulate opening of the BBB in a wide range of neurological disorders. CONCLUSIONS: These findings represent the first direct evidence of inflammation-related BBB permeability changes in ALF.


Subject(s)
Blood-Brain Barrier/drug effects , Hepatic Encephalopathy/physiopathology , Lipopolysaccharides/pharmacology , Liver Failure, Acute/pathology , Liver/pathology , Ammonia/blood , Animals , Azoxymethane/toxicity , Cytokines/metabolism , Disease Models, Animal , Inflammation/pathology , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Permeability , Transaminases/blood
3.
PLoS One ; 7(11): e49670, 2012.
Article in English | MEDLINE | ID: mdl-23166746

ABSTRACT

BACKGROUND/AIMS: Acute liver failure (ALF) due to ischemic or toxic liver injury is a clinical condition that results from massive loss of hepatocytes and may lead to hepatic encephalopathy (HE), a serious neuropsychiatric complication. Although increased expression of tumor necrosis factor-alpha (TNF-α) in liver, plasma and brain has been observed, conflicting results exist concerning its roles in drug-induced liver injury and on the progression of HE. The present study aimed to investigate the therapeutic value of etanercept, a TNF-α neutralizing molecule, on the progression of liver injury and HE in mice with ALF resulting from azoxymethane (AOM) hepatotoxicity. METHODS/PRINCIPAL FINDINGS: Mice were administered saline or etanercept (10 mg/kg; i.p.) 30 minutes prior to, or up to 6 h after AOM. Etanercept-treated ALF mice were sacrificed in parallel with vehicle-treated comatose ALF mice and controls. AOM induced severe hepatic necrosis, leading to HE, and etanercept administered prior or up to 3 h after AOM significantly delayed the onset of coma stages of HE. Etanercept pretreatment attenuated AOM-induced liver injury, as assessed by histological examination, plasma ammonia and transaminase levels, and by hepatic glutathione content. Peripheral inflammation was significantly reduced by etanercept as shown by decreased plasma IL-6 (4.1-fold; p<0.001) and CD40L levels (3.7-fold; p<0.001) compared to saline-treated ALF mice. Etanercept also decreased IL-6 levels in brain (1.2-fold; p<0.05), attenuated microglial activation (assessed by OX-42 immunoreactivity), and increased brain glutathione concentrations. CONCLUSIONS: These results indicate that systemic sequestration of TNF-α attenuates both peripheral and cerebral inflammation leading to delayed progression of liver disease and HE in mice with ALF due to toxic liver injury. These results suggest that etanercept may provide a novel therapeutic approach for the management of ALF patients awaiting liver transplantation.


Subject(s)
Hepatic Encephalopathy/etiology , Hepatic Encephalopathy/metabolism , Liver Failure, Acute/complications , Liver Failure, Acute/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ammonia/blood , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Azoxymethane/adverse effects , CD40 Ligand/blood , Cerebrum/drug effects , Cerebrum/metabolism , Disease Progression , Etanercept , Hepatic Encephalopathy/drug therapy , Immunoglobulin G/administration & dosage , Immunoglobulin G/pharmacology , Interleukin-6/blood , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Failure, Acute/chemically induced , Liver Failure, Acute/drug therapy , Male , Mice , Microglia/drug effects , Microglia/metabolism , Oxidative Stress/drug effects , Receptors, Tumor Necrosis Factor/administration & dosage , Tumor Necrosis Factor-alpha/blood
4.
Cell Metab ; 14(6): 724-38, 2011 Dec 07.
Article in English | MEDLINE | ID: mdl-22152301

ABSTRACT

The energy requirements of the brain are very high, and tight regulatory mechanisms operate to ensure adequate spatial and temporal delivery of energy substrates in register with neuronal activity. Astrocytes-a type of glial cell-have emerged as active players in brain energy delivery, production, utilization, and storage. Our understanding of neuroenergetics is rapidly evolving from a "neurocentric" view to a more integrated picture involving an intense cooperativity between astrocytes and neurons. This review focuses on the cellular aspects of brain energy metabolism, with a particular emphasis on the metabolic interactions between neurons and astrocytes.


Subject(s)
Astrocytes/metabolism , Brain/blood supply , Brain/physiology , Energy Metabolism/physiology , Models, Biological , Neurons/metabolism , Brain/cytology , Glycogen/metabolism , Lactic Acid/metabolism , Oxidative Stress/physiology , Regional Blood Flow/physiology
5.
J Neurosci ; 31(50): 18338-52, 2011 Dec 14.
Article in English | MEDLINE | ID: mdl-22171037

ABSTRACT

The glyoxalase system is the most important pathway for the detoxification of methylglyoxal (MG), a highly reactive dicarbonyl compound mainly formed as a by-product of glycolysis. MG is a major precursor of advanced glycation end products (AGEs), which are associated with several neurodegenerative disorders. Although the neurotoxic effects of MG and AGEs are well characterized, little is known about the glyoxalase system in the brain, in particular with regards to its activity in different neural cell types. Results of the present study reveal that both enzymes composing the glyoxalase system [glyoxalase-1 (Glo-1) and Glo-2] were highly expressed in primary mouse astrocytes compared with neurons, which translated into higher enzymatic activity rates in astrocytes (9.9- and 2.5-fold, respectively). The presence of a highly efficient glyoxalase system in astrocytes was associated with lower accumulation of AGEs compared with neurons (as assessed by Western blotting), a sixfold greater resistance to MG toxicity, and the capacity to protect neurons against MG in a coculture system. In addition, Glo-1 downregulation using RNA interference strategies resulted in a loss of viability in neurons, but not in astrocytes. Finally, stimulation of neuronal glycolysis via lentiviral-mediated overexpression of 6-phosphofructose-2-kinase/fructose-2,6-bisphosphatase-3 resulted in increased MG levels and MG-modified proteins. Since MG is largely produced through glycolysis, this suggests that the poor capacity of neurons to upregulate their glycolytic flux as compared with astrocytes may be related to weaker defense mechanisms against MG toxicity. Accordingly, the neuroenergetic specialization taking place between these two cell types may serve as a protective mechanism against MG-induced neurotoxicity.


Subject(s)
Astrocytes/enzymology , Cytoprotection/physiology , Lactoylglutathione Lyase/metabolism , Neurons/enzymology , Thiolester Hydrolases/metabolism , Animals , Astrocytes/cytology , CHO Cells , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/enzymology , Cricetinae , Lactoylglutathione Lyase/genetics , Mice , Neurons/cytology , Thiolester Hydrolases/genetics
6.
Trends Neurosci ; 34(2): 76-87, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21236501

ABSTRACT

In recent years, previously unsuspected roles of astrocytes have been revealed, largely owing to the development of new tools enabling their selective study in situ. These exciting findings add to the large body of evidence demonstrating that astrocytes play a central role in brain homeostasis, in particular via the numerous cooperative metabolic processes they establish with neurons, such as the supply of energy metabolites and neurotransmitter recycling functions. Furthermore, impairments in astrocytic function are increasingly being recognized as an important contributor to neuronal dysfunction and, in particular, neurodegenerative processes. In this review, we discuss recent evidence supporting important roles for astrocytes in neuropathological conditions such as neuroinflammation, amyotrophic lateral sclerosis and Alzheimer's disease. We also explore the potential for neuroprotective therapeutics based on the modulation of astrocytic functions.


Subject(s)
Astrocytes/metabolism , Neurons/metabolism , Animals , Encephalitis/pathology , Encephalitis/physiopathology , Energy Metabolism/physiology , Homeostasis/physiology , Humans , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology
7.
J Neurochem ; 116(4): 564-76, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21143598

ABSTRACT

We have previously reported that the pro-inflammatory cytokines tumor necrosis factor-α (TNFα) and interleukin-1ß (IL-1ß) induce profound modifications of the metabolic profile of astrocytes. The present study was undertaken to further characterize the effects of cytokines in astrocytes and to determine whether similar effects could also be observed in neurons. To do so, selected pro-inflammatory (IL-6 and interferon-γ, in addition to the above-mentioned TNFα and IL-1ß) and anti-inflammatory cytokines (IL-4, IL-10, transforming growth factor-ß1 and interferon-ß) were applied to primary neuronal and astrocytic cultures, and key metabolic parameters were assessed. As a general pattern, we observed that pro-inflammatory cytokines increased glucose utilization in astrocytes while the anti-inflammatory cytokines IL-4 and IL-10 decreased astrocytic glucose utilization. In contrast, no significant change could be observed in neurons. When pairs of pro-inflammatory cytokines were co-applied in astrocytes, several additive or synergistic modifications could be observed. In contrast, IL-10 partially attenuated the effects of pro-inflammatory cytokines. Finally, the modifications of the astrocytic metabolism induced by TNFα + IL-1ß and interferon-γ modulated neuronal susceptibility to an excitotoxic insult in neuron-astrocyte co-cultures. Together, these results suggest that pro- and anti-inflammatory cytokines differentially affect the metabolic profile of astrocytes, and that these changes have functional consequences for surrounding neurons.


Subject(s)
Astrocytes/metabolism , Cytokines/physiology , Inflammation Mediators/physiology , Metabolome/physiology , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/pathology , Cell Survival/physiology , Cells, Cultured , Coculture Techniques , Cytokines/classification , Down-Regulation/physiology , Energy Metabolism/physiology , Glucose/antagonists & inhibitors , Glucose/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Mice , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Up-Regulation/physiology
8.
J Neurosci ; 30(9): 3326-38, 2010 Mar 03.
Article in English | MEDLINE | ID: mdl-20203192

ABSTRACT

Amyloid-beta (Abeta) peptides play a key role in the pathogenesis of Alzheimer's disease and exert various toxic effects on neurons; however, relatively little is known about their influence on glial cells. Astrocytes play a pivotal role in brain homeostasis, contributing to the regulation of local energy metabolism and oxidative stress defense, two aspects of importance for neuronal viability and function. In the present study, we explored the effects of Abeta peptides on glucose metabolism in cultured astrocytes. Following Abeta(25-35) exposure, we observed an increase in glucose uptake and its various metabolic fates, i.e., glycolysis (coupled to lactate release), tricarboxylic acid cycle, pentose phosphate pathway, and incorporation into glycogen. Abeta increased hydrogen peroxide production as well as glutathione release into the extracellular space without affecting intracellular glutathione content. A causal link between the effects of Abeta on glucose metabolism and its aggregation and internalization into astrocytes through binding to members of the class A scavenger receptor family could be demonstrated. Using astrocyte-neuron cocultures, we observed that the overall modifications of astrocyte metabolism induced by Abeta impair neuronal viability. The effects of the Abeta(25-35) fragment were reproduced by Abeta(1-42) but not by Abeta(1-40). Finally, the phosphoinositide 3-kinase (PI3-kinase) pathway appears to be crucial in these events since both the changes in glucose utilization and the decrease in neuronal viability are prevented by LY294002, a PI3-kinase inhibitor. This set of observations indicates that Abeta aggregation and internalization into astrocytes profoundly alter their metabolic phenotype with deleterious consequences for neuronal viability.


Subject(s)
Amyloid beta-Peptides/metabolism , Astrocytes/metabolism , Brain/metabolism , Energy Metabolism/physiology , Nerve Degeneration/metabolism , Neurons/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/toxicity , Animals , Animals, Newborn , Astrocytes/drug effects , Brain/pathology , Brain/physiopathology , Cell Communication/drug effects , Cell Communication/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Energy Metabolism/drug effects , Free Radicals/metabolism , Glucose/metabolism , Glutathione/metabolism , Hydrogen Peroxide/metabolism , Mice , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Peptide Fragments/toxicity , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors
9.
Dialogues Clin Neurosci ; 11(3): 281-95, 2009.
Article in English | MEDLINE | ID: mdl-19877496

ABSTRACT

Astrocytes are the main neural cell type responsible for the maintenance of brain homeostasis. They form highly organized anatomical domains that are interconnected into extensive networks. These features, along with the expression of a wide array of receptors, transporters, and ion channels, ideally position them to sense and dynamically modulate neuronal activity. Astrocytes cooperate with neurons on several levels, including neurotransmitter trafficking and recycling, ion homeostasis, energy metabolism, and defense against oxidative stress. The critical dependence of neurons upon their constant support confers astrocytes with intrinsic neuroprotective properties which are discussed here. Conversely, pathogenic stimuli may disturb astrocytic function, thus compromising neuronal functionality and viability. Using neuroinflammation, Alzheimer's disease, and hepatic encephalopathy as examples, we discuss how astrocytic defense mechanisms may be overwhelmed in pathological conditions, contributing to disease progression.


Subject(s)
Astrocytes/physiology , Brain Diseases , Neurons/metabolism , Neuroprotective Agents/metabolism , Animals , Brain/cytology , Brain Diseases/metabolism , Brain Diseases/pathology , Brain Diseases/prevention & control , Homeostasis/physiology , Humans , Models, Biological
10.
Hepatology ; 46(6): 1883-92, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17705298

ABSTRACT

UNLABELLED: We previously demonstrated in pigs with acute liver failure (ALF) that albumin dialysis using the molecular adsorbents recirculating system (MARS) attenuated a rise in intracranial pressure (ICP). This was independent of changes in arterial ammonia, cerebral blood flow and inflammation, allowing alternative hypotheses to be tested. The aims of the present study were to determine whether changes in cerebral extracellular ammonia, lactate, glutamine, glutamate, and energy metabolites were associated with the beneficial effects of MARS on ICP. Three randomized groups [sham, ALF (induced by portacaval anastomosis and hepatic artery ligation), and ALF+MARS] were studied over a 6-hour period with a 4-hour MARS treatment given beginning 2 hours after devascularization. Using cerebral microdialysis, the ALF-induced increase in extracellular brain ammonia, lactate, and glutamate was significantly attenuated in the ALF+MARS group as well as the increases in extracellular lactate/pyruvate and lactate/glucose ratios. The percent change in extracellular brain ammonia correlated with the percent change in ICP (r(2) = 0.511). Increases in brain lactate dehydrogenase activity and mitochondrial complex activity for complex IV were found in ALF compared with those in the sham, which was unaffected by MARS treatment. Brain oxygen consumption did not differ among the study groups. CONCLUSION: The observation that brain oxygen consumption and mitochondrial complex enzyme activity changed in parallel in both ALF- and MARS-treated animals indicates that the attenuation of increased extracellular brain ammonia (and extracellular brain glutamate) in the MARS-treated animals reduces energy demand and increases supply, resulting in attenuation of increased extracellular brain lactate. The mechanism of how MARS reduces extracellular brain ammonia requires further investigation.


Subject(s)
Ammonia/analysis , Brain Chemistry , Intracranial Pressure , Lactic Acid/analysis , Liver Failure, Acute/therapy , Sorption Detoxification , Animals , Disease Models, Animal , Extracellular Space , Female , Glutamic Acid/analysis , Glutamine/analysis , Swine
11.
Gastroenterology ; 132(1): 372-83, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17241886

ABSTRACT

BACKGROUND & AIMS: Body temperature may critically affect mechanisms of liver injury in acetaminophen (APAP) hepatotoxicity. In addition, mild hypothermia is used to treat intracranial hypertension in human liver failure without detailed information on its effects on the injured liver itself. Therefore, we investigated the effects of body temperature on the progression of APAP-induced liver injury in mice. METHODS: Male C57BL6 mice treated with saline or APAP (300 mg/kg intraperitoneally) were maintained at normothermia (35.5-37.5 degrees C) by external warming or were allowed to develop mild hypothermia (32.0-35.0 degrees C) after 2 hours from APAP administration. RESULTS: Mild hypothermia resulted in improved survival after APAP intoxication. Liver damage was reduced, as assessed histologically and by plasma alanine aminotransferase levels. Early effects of hypothermia included a reduction of hepatic congestion and improved recovery of glycogen stores. At later time points (8-12 hours), APAP-treated mice that were maintained at normothermia manifested increased hepatocyte apoptosis, as assessed by terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling staining and cleavage of poly(adenosine diphosphate-ribose) polymerase. Mild hypothermia did not affect the formation of APAP-protein adducts or the depletion of glutathione, nor did it abrogate hepatocyte DNA synthesis. CONCLUSIONS: Mild hypothermia improved survival and attenuated liver injury and apoptosis in APAP-treated mice by reducing hepatic congestion and improving glycogen recovery without affecting hepatic regeneration. Results of the study underscore the need for a strict control of body temperature in animal models of liver failure and suggest that the benefits of mild hypothermia in liver failure may extend beyond those related to reduced cerebral complications.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Chemical and Drug Induced Liver Injury/therapy , Hypothermia, Induced , Animals , Apoptosis , Body Temperature , Cell Count , Cell Division , Chemical and Drug Induced Liver Injury/mortality , Chemical and Drug Induced Liver Injury/pathology , DNA Adducts/metabolism , Glutathione/metabolism , Glycogen/metabolism , Hemoglobins/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , In Situ Nick-End Labeling , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Necrosis , Severity of Illness Index , Survival Rate
12.
Neurochem Int ; 50(1): 95-101, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16956696

ABSTRACT

Ammonia is a key neurotoxin involved in the neurological complications of acute liver failure. The present study was undertaken to study the effects of exposure to pathophysiologically relevant concentrations of ammonium chloride on cultured brain capillary endothelial cells in order to identify mechanisms by which ammonia may alter blood-brain barrier function. Conditionally immortalized mouse brain capillary endothelial cells (TM-BBB) were used as an in vitro model of the blood-brain barrier. Gene expression of a series of blood-brain barrier transporters and tight junction proteins was assessed by quantitative real time PCR analysis. Exposure to ammonia (5mM for 72h) resulted in significant increases in mRNA levels of taurine transporter (TAUT; 2.0-fold increase) as well as creatine transporter (CRT; 1.9-fold increase) whereas claudin-12 mRNA expression was significantly reduced to 67.7% of control levels. Furthermore, [(3)H]taurine and [(14)C]creatine uptake were concomitantly increased following exposure to ammonia, suggesting that up-regulation of both TAUT and CRT under hyperammonemic conditions results in an increased function of these two transporters in TM-BBB cells. TAUT and CRT are respectively involved in osmoregulation and energy buffering in the brain, two systems that are thought to be affected in acute liver failure. Furthermore, claudin-12 down-regulation suggests that hyperammonemia may also affect tight junction integrity. Our results provide evidence that ammonia can alter brain capillary endothelial cell gene expression and transporter function. These findings may be relevant to pathological situations involving hyperammonemia, such as liver disease.


Subject(s)
Brain/blood supply , Capillaries/drug effects , Creatine/metabolism , Endothelium, Vascular/drug effects , Gene Expression/drug effects , Hyperammonemia/metabolism , Membrane Proteins/genetics , Taurine/metabolism , Animals , Base Sequence , Capillaries/cytology , Cell Line, Transformed , Claudins , DNA Primers , Endothelium, Vascular/cytology , Mice , Polymerase Chain Reaction
14.
Neurochem Int ; 48(6-7): 434-40, 2006.
Article in English | MEDLINE | ID: mdl-16563565

ABSTRACT

Molecular biological approaches continue to lead to the identification of alterations in expression of genes coding for key central nervous system proteins involved in water homeostasis, energy metabolism and neurotransmitter regulation in acute liver failure (ALF). However, studies aimed at elucidating the pathophysiological consequences of these changes in gene expression are impeded by the lack of a suitable mouse model of ALF. A previous report described hepatic pathology characteristic of ALF resulting from the administration of azoxymethane (AOM) in mice [Matkowskyj, K.A., Marrero, J.A., Carroll, R.E., Danilkovich, A.V., Green, R.M., Benya, R.V., 1999. Azoxymethane-induced fulminant hepatic failure in C57BL/6J mice: characterization of a new animal model. Am. J. Physiol. 277, G455-G462]. In a series of experiments to further assess this treatment as an effective model of ALF, the effects of administration of AOM to male C57BL mice on hepatic and cerebral function were studied. With maintenance of body temperature at 37 degrees C and control of hypoglycemia, mice developed signs of encephalopathy (decreased locomotor activity followed by loss of righting and corneal reflexes) within 16 h of AOM treatment. AOM-treated mice were hyperammonemic, developed spontaneous hypothermia and brain edema. Brain ammonia concentrations were increased to 0.98+/-0.12 mM at coma stages of encephalopathy. Brain amino acid profiles determined by HPLC were typical of ALF in other species including humans. Mild hypothermia (35 degrees C) led to significant attenuation of brain edema, ammonia, and amino acid changes. These findings demonstrate that AOM treatment affords a simple, reproducible mouse model of ALF which may be suitable for the study of the effects of gene manipulation on the cerebral complications of ALF.


Subject(s)
Azoxymethane , Liver Failure, Acute/chemically induced , Liver Failure, Acute/metabolism , Amino Acids/analysis , Ammonia/analysis , Ammonia/blood , Animals , Body Water , Brain Chemistry , Disease Models, Animal , Hypothermia, Induced , Liver/metabolism , Liver/pathology , Liver Failure, Acute/prevention & control , Male , Mice , Mice, Inbred C57BL
15.
Glia ; 53(5): 557-62, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16374780

ABSTRACT

Acute liver failure (ALF) is consistently accompanied by alterations in brain energy metabolites and recent nuclear magnetic resonance (NMR) studies suggest disturbances in brain oxidative metabolism in experimental ALF. Glucose transport across the blood-brain barrier is essential to sustain brain energy metabolism and is accomplished by the facilitative glucose transporter GLUT1. To investigate alterations in brain glucose uptake in acute liver failure further, GLUT1 expression and [14C]-2-deoxy-D-glucose uptake were measured in the brains of rats with hepatic devascularization. RT-PCR and Western blot analyses showed significant increases in steady-state levels of GLUT1 mRNA and protein in frontal cortex as early as 6 h following hepatic devascularization, (prior to the onset of brain edema and encephalopathy) which remained elevated at coma stages of encephalopathy. Expression of the astrocytic (45-kDa) and endothelial (55-kDa) forms of GLUT1 was increased as a result of hepatic devascularization. Exposure of cultured astrocytes to pathophysiologically relevant concentrations of ammonia resulted in increased GLUT1 expression, suggesting that elevated ammonia levels are responsible for GLUT1 upregulation in ALF. Increased GLUT1 expression in ALF was selective, since expression of the neuronal glucose transporter GLUT3 and other glucose-regulated proteins (GRP-78 and GRP-94) was unaltered. [14C]-2-deoxy-D-glucose autoradiography revealed increases in cerebral glucose uptake following the induction of GLUT1 in ALF. These results suggest that ammonia-induced increases of GLUT1 expression resulting in increased cerebral glucose uptake occur in ALF and could contribute to the pathophysiological mechanisms responsible for the neurological complications of this condition.


Subject(s)
Astrocytes/metabolism , Endothelium, Vascular/metabolism , Glucose Transporter Type 1/biosynthesis , Liver Failure, Acute/metabolism , Ammonia/metabolism , Animals , Antimetabolites/pharmacology , Antisense Elements (Genetics) , Blotting, Western , Brain Chemistry/drug effects , Brain Edema/metabolism , Brain Edema/pathology , Cells, Cultured , Deoxyglucose/pharmacology , Immunohistochemistry , Male , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
16.
Metab Brain Dis ; 20(4): 409-23, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16382351

ABSTRACT

The availability of adequate experimental models of acute liver failure (ALF) is of prime importance to provide a better understanding of this condition and allow the development and testing of new therapeutic approaches for patients with ALF. However, the numerous etiologies and complications of ALF contribute to the complexity of this condition and render the development of an ideal experimental model of ALF more difficult than expected. Instead, a number of different models that may be used for the study of specific aspects of ALF have been developed. The most common approaches used to induce ALFin experimental animals are surgical procedures, toxic liver injury,or a combination of both. Despite the high prevalence of viral hepatitis worldwide, very few satisfactory viral models of ALF are available. Established and newly developed models of ALF are reviewed.


Subject(s)
Acute Kidney Injury/pathology , Animals , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Hepatectomy , Hepatitis, Viral, Animal/pathology , Humans , Liver/surgery
17.
J Hepatol ; 42(5): 694-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15826719

ABSTRACT

BACKGROUND/AIMS: Mild hypothermia has proven useful in the clinical management of patients with acute liver failure. Acute liver failure in experimental animals results in alterations in the expression of genes coding for astrocytic proteins including the "peripheral-type" (astrocytic) benzodiazepine receptor (PTBR), a mitochondrial complex associated with neurosteroid synthesis. To gain further insight into the mechanisms whereby hypothermia attenuates the neurological complications of acute liver failure, we investigated PTBR expression in the brains of hepatic devascularized rats under normothermic (37 degrees C) and hypothermic (35 degrees C) conditions. METHODS: PTBR mRNA was measured using semi-quantitative RT-PCR in cerebral cortical extracts and densities of PTBR sites were measured by quantitative receptor autoradiagraphy. Brain pregnenolone content was measured by radioimmunoassay. RESULTS: At coma stages of encephalopathy, animals with acute liver failure manifested a significant increase of PTBR mRNA levels. Brain pregnenolone content and [(3)H]PK 11195 binding site densities were concomitantly increased. Mild hypothermia prevented brain edema and significantly attenuated the increased receptor expression and pregnenolone content. CONCLUSIONS: These findings suggest that an attenuation of PTBR up-regulation resulting in the prevention of increased brain neurosteroid content represents one of the mechanisms by which mild hypothermia exerts its protective effects in ALF.


Subject(s)
Astrocytes/physiology , Brain Edema/prevention & control , Hypothermia, Induced , Liver Failure, Acute/therapy , Receptors, GABA-A/genetics , Animals , Brain/cytology , Brain/metabolism , Brain Edema/etiology , Brain Edema/physiopathology , Hepatic Encephalopathy/etiology , Hepatic Encephalopathy/physiopathology , Hepatic Encephalopathy/prevention & control , Liver Failure, Acute/complications , Liver Failure, Acute/physiopathology , Male , Pregnenolone/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
18.
Neurochem Int ; 41(2-3): 155-60, 2002.
Article in English | MEDLINE | ID: mdl-12020615

ABSTRACT

Glial fibrillary acid protein (GFAP) is a major component of the glial filament network and alterations in expression of this protein in cultured astrocytes have been reported in response to acute ammonia exposure in vitro. In order to determine the effects of acute hyperammonemia in vivo on GFAP expression, brain extracts from rats with acute liver failure due to hepatic devascularization (portacaval anastomosis followed 24h later by hepatic artery ligation, HAL) were analyzed for GFAP mRNA using reverse transcription-polymerase chain reaction (RT-PCR) and appropriate oligonucleotide primers. GFAP protein was assayed by immunoblotting using a polyclonal antibody. Hepatic devascularization resulted in a significant 55-68% decrease (P<0.01) of GFAP mRNA and a concomitant loss of GFAP protein at precoma and coma stages of encephalopathy when brain water content was significantly increased and brain ammonia concentrations were in the millimolar range (1-5mM). Expression of a second glial filament protein S-100beta was unaffected by acute hyperammonemia. These findings suggest a role for GFAP in cell volume regulation and that loss of GFAP expression could contribute to the pathogenesis of brain edema in acute hyperammonemic syndromes.


Subject(s)
Glial Fibrillary Acidic Protein/metabolism , Hyperammonemia/metabolism , Acute Disease , Animals , Brain/metabolism , Glial Fibrillary Acidic Protein/genetics , Hepatic Encephalopathy/metabolism , Liver Failure/metabolism , Male , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...