Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(15)2021 Jul 27.
Article in English | MEDLINE | ID: mdl-34360766

ABSTRACT

Age-related hearing loss (ARHL) is the most common sensory disorder among older people, and yet, the treatment options are limited to medical devices such as hearing aids and cochlear implants. The high prevalence of ARHL mandates the development of treatment strategies that can prevent or rescue age-related cochlear degeneration. In this study, we investigated a novel pharmacological strategy based on inhibition of the adenosine A2A receptor (A2AR) in middle aged C57BL/6 mice prone to early onset ARHL. C57BL/6J mice were treated with weekly istradefylline (A2AR antagonist; 1 mg/kg) injections from 6 to 12 months of age. Auditory function was assessed using auditory brainstem responses (ABR) to tone pips (4-32 kHz). ABR thresholds and suprathreshold responses (wave I amplitudes and latencies) were evaluated at 6, 9, and 12 months of age. Functional outcomes were correlated with quantitative histological assessments of sensory hair cells. Cognitive function was assessed using the Morris water maze and the novel object recognition test, and the zero maze test was used to assess anxiety-like behaviour. Weekly injections of istradefylline attenuated ABR threshold shifts by approximately 20 dB at mid to high frequencies (16-32 kHz) but did not improve ABR suprathreshold responses. Istradefylline treatment improved hair cell survival in a turn-dependent manner, whilst the cognitive function was unaffected by istradefylline treatment. This study presents the first evidence for the rescue potential of istradefylline in ARHL and highlights the role of A2AR in development of age-related cochlear degeneration.


Subject(s)
Aging , Auditory Threshold/drug effects , Evoked Potentials, Auditory, Brain Stem/drug effects , Presbycusis , Purines/pharmacology , Animals , Male , Mice , Presbycusis/drug therapy , Presbycusis/pathology , Presbycusis/physiopathology
2.
Biomolecules ; 11(2)2021 01 25.
Article in English | MEDLINE | ID: mdl-33503847

ABSTRACT

BYL719 (alpelisib) is a small molecule inhibitor of PI3K p110α developed for cancer therapy. Targeted suppression of PI3K has led to lifespan extension in rodents and model organisms. If PI3K inhibitors are to be considered as an aging therapeutic, it is important to understand the potential consequences of long-term exposure, and the most practical way to achieve this is through diet administration. Here, we investigated the pharmacokinetics of BYL719 delivered in diet and the efficacy of BYL719 to suppress insulin signaling when administered in the diet of 8-month-old male and female mice. Compared to oral gavage, diet incorporation resulted in a lower peak plasma BYL719 (3.6 vs. 9.2 µM) concentration but similar half-life (~1.5 h). Consuming BYL719 resulted in decreased insulin signaling in liver and muscle within 72 h, and mice still showed impaired glucose tolerance and insulin sensitivity following 6 weeks of access to a diet containing 0.3 g/kg BYL719. However, consuming BYL719 did not affect food intake, body mass, muscle function (rotarod and hang time performance) or cognitive behaviors. This provides evidence that BYL719 has long-term efficacy without major toxicity or side effects, and suggests that administering BYL719 in diet is suitable for studying the effect of pharmacological suppression of PI3K p110α on aging and metabolic function.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Thiazoles/pharmacology , Aging , Animals , Behavior, Animal , Female , Glucose/metabolism , Glucose Tolerance Test , Homeostasis , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscles/metabolism , Receptor, Insulin/metabolism
3.
Front Mol Neurosci ; 13: 522073, 2020.
Article in English | MEDLINE | ID: mdl-33224025

ABSTRACT

Alzheimer's disease (AD), the most common chronic neurodegenerative disorder, has complex neuropathology. The principal neuropathological hallmarks of the disease are the deposition of extracellular ß-amyloid (Aß) plaques and neurofibrillary tangles (NFTs) comprised of hyperphosphorylated tau (p-tau) protein. These changes occur with neuroinflammation, a compromised blood-brain barrier (BBB) integrity, and neuronal synaptic dysfunction, all of which ultimately lead to neuronal cell loss and cognitive deficits in AD. Aß1-42 was stereotaxically administered bilaterally into the CA1 region of the hippocampi of 18-month-old male C57BL/6 mice. This study aimed to characterize, utilizing immunohistochemistry and behavioral testing, the spatial and temporal effects of Aß1-42 on a broad set of parameters characteristic of AD: p-tau, neuroinflammation, vascular pathology, pyramidal cell survival, and behavior. Three days after Aß1-42 injection and before significant neuronal cell loss was detected, acute neuroinflammatory and vascular responses were observed. These responses included the up-regulation of glial fibrillary acidic protein (GFAP), cell adhesion molecule-1 (PECAM-1, also known as CD31), fibrinogen labeling, and an increased number of activated astrocytes and microglia in the CA1 region of the hippocampus. From day 7, there was significant pyramidal cell loss in the CA1 region of the hippocampus, and by 30 days, significant localized up-regulation of p-tau, GFAP, Iba-1, CD31, and alpha-smooth muscle actin (α-SMA) in the Aß1-42-injected mice compared with controls. These molecular changes in Aß1-42-injected mice were accompanied by cognitive deterioration, as demonstrated by long-term spatial memory impairment. This study is reporting a comprehensive examination of a complex set of parameters associated with intrahippocampal administration of Aß1-42 in mice, their spatiotemporal interactions and combined contribution to the disease progression. We show that a single Aß injection can reproduce aspects of the inflammatory, vascular, and p-tau induced pathology occurring in the AD human brain that lead to cognitive deficits.

4.
Front Behav Neurosci ; 12: 39, 2018.
Article in English | MEDLINE | ID: mdl-29559901

ABSTRACT

Computer-supported gait analysis has proven to be effective for the comprehensive assessment of gait changes in rodent models of neurodegenerative and neurological disorders. However, full characterization of individual gait parameters is required for specific neurological or neurodegenerative disorders such as Parkinson's disease (PD). Gait disturbances in particular present as the most constraining set of symptoms in PD, finally depriving patients from most activities of normal daily living. In this study, we have characterized the gait pattern abnormalities observed in two rat models of PD: the medial forebrain bundle (MFB) 6-OHDA lesion model and the striatal 6-OHDA lesion model. Our data indicates significant changes in 21 different gait parameters in the MFB lesion cohort. We observed a steady decline in the overall walking speed and cadence, as well as significant alterations in the gait parameters stride length, initial dual stance, paw print position, step cycle, swing phase of the step cycle, stand index, phase dispersion, print length, and print area in at least one of the paws. These alterations correlated with the extent of tyrosine hydroxylase (TH) neuronal loss observed in this group. These alterations were detected as early as 1 week post lesion. In contrast, limited gait dysfunction was detected in the striatal lesion cohort related to the low level of TH neuronal loss detected in this group. In this study we have demonstrated that gait analysis is a reliable method for the detection of motor deficiencies in a MFB 6-OHDA lesion model of PD and may prove a clinically relevant, low impact method of testing functional impairment as early as 1 week post lesion.

5.
Neurobiol Dis ; 94: 95-105, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27288154

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease where the degeneration of the nigrostriatal pathway leads to specific motor deficits. There is an unmet medical need for regenerative treatments that stop or reverse disease progression. Several growth factors have been investigated in clinical trials to restore the dopaminergic nigrostriatal pathway damaged in PD. Platelet-derived growth factor-BB (PDGF-BB), a molecule that recruits pericytes to stabilize microvessels, was recently investigated in a phase-1 clinical trial, showing a dose-dependent increase in dopamine transporter binding in the putamen of PD patients. Interestingly, evidence is accumulating that PD is paralleled by microvascular changes, however, whether PDGF-BB modifies pericytes in PD is not known. Using a pericyte reporter mouse strain, we investigate the functional and restorative effect of PDGF-BB in a partial 6-hydroxydopamine medial forebrain bundle lesion mouse model of PD, and whether this restorative effect is accompanied by changes in pericyte features. We demonstrate that a 2-week treatment with PDGF-BB leads to behavioural recovery using several behavioural tests, and partially restores the nigrostriatal pathway. Interestingly, we find that pericytes are activated in the striatum of PD lesioned mice and that these changes are reversed by PDGF-BB treatment. The modulation of brain pericytes may contribute to the PDGF-BB-induced neurorestorative effects, PDGF-BB allowing for vascular stabilization in PD. Pericytes might be a new cell target of interest for future regenerative therapies.


Subject(s)
Motor Activity/drug effects , Parkinson Disease/metabolism , Pericytes/drug effects , Proto-Oncogene Proteins c-sis/pharmacology , Animals , Becaplermin , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/metabolism , Male , Medial Forebrain Bundle/drug effects , Medial Forebrain Bundle/metabolism , Mice, Transgenic , Motor Activity/physiology , Oxidopamine/pharmacology , Parkinson Disease/pathology , Pericytes/metabolism , Proto-Oncogene Proteins c-sis/metabolism
6.
Behav Brain Res ; 284: 196-206, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25698603

ABSTRACT

The most frequently used animal models for Parkinson's disease (PD) utilize unilateral injection of 6-hydroxydopamine (6-OHDA) in the medial forebrain bundle (MFB), which results in total denervation of the dopaminergic nigrostriatal pathway. However, neuroprotective interventions in PD require models resembling earlier stages of PD, where some dopaminergic cells and fibres remain. The aim of the present study was therefore to establish a MFB partial lesion model in mice. We tested four different 6-OHDA doses, and our results show a dose-dependent loss of nigral dopaminergic cells and striatal fibres that correlated with behavioural impairment in several behavioural tests. Specifically, doses of 0.7 µg and 1 µg of 6-OHDA induced a partial denervation of the nigrostriatal pathway, associated with a mild but quantifiable behavioural impairment. We identified the amphetamine-induced rotation, stepping, corridor and cylinder test to be sensitive enough to select partial lesion animals. Based on our data, we proposed a range of cut-off values for these different behavioural tests to select partial lesion mice. Using a statistical prediction model we identified two behavioural tests (the stepping test and amphetamine-induced rotation test) that with a high sensitivity and specificity predict the extent of nigral dopaminergic cell loss and select mice with a partial nigrostriatal lesion prior to further interventions. This model can serve as an important tool to study neuroprotective therapies for PD in mouse models, especially when the treatment targets the substantia nigra and/or the striatum.


Subject(s)
Medial Forebrain Bundle/pathology , Oxidopamine , Parkinsonian Disorders/pathology , Parkinsonian Disorders/physiopathology , Amphetamine/pharmacology , Animals , Area Under Curve , Dopamine Agents/pharmacology , Dopaminergic Neurons/pathology , Dopaminergic Neurons/physiology , Dose-Response Relationship, Drug , Immunohistochemistry , Membrane Glycoproteins , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , ROC Curve , Receptors, Interleukin-1 , Rotation , Severity of Illness Index
7.
Chem Res Toxicol ; 26(8): 1159-67, 2013 Aug 19.
Article in English | MEDLINE | ID: mdl-23829299

ABSTRACT

It is known from controlled animal experiments and human epidemiologic studies that early life exposure to mixtures of polychlorinated biphenyls (PCBs) is a risk factor for developmental neurotoxicity. The importance of non-dioxin-like PCBs in the context of the observed effect is uncertain because of the blending with the more potent dioxin-like PCBs. Previously, a controlled rat perinatal exposure study with individual, purity-controlled, non-dioxin-like congeners (PCB52, PCB138, or PCB180) was set up. Impaired motor coordination, motor activity, and learning has been reported for the offspring at an age of approximately 4 months. Here, we report on the gene expression responses that have been observed in the blood of the same animals. ANOVA analysis called 1412 genes differentially expressed 4 months after the PCB treatment was stopped. Subsequently, each PCB exposure condition was compared to the corresponding vehicle control using a fold change analysis. The gene lists contained between 82 and 348 differentially expressed genes. Expression patterns were complex with sets of differentially expressed genes being specific for a particular PCB exposure and other sets in common between several exposure conditions. Thirty-two genes were differentially expressed under all conditions. Bioinformatic overrepresentation analysis identified enriched biological terms such as lipid metabolism, molecular transport, small molecule biochemistry, and cell signaling and proliferation. Gene lists were particularly enriched for nervous system development and function ontology. In conclusion, we have documented for the first time differential gene expression in a well-controlled animal study that reported behavioral effects of purity-controlled individual non-dioxin-like PCBs.


Subject(s)
Maternal Exposure , Polychlorinated Biphenyls/toxicity , Transcriptome/drug effects , Animals , Down-Regulation/drug effects , Female , Male , Motor Activity/drug effects , Nervous System/growth & development , Nervous System/metabolism , Polychlorinated Biphenyls/chemistry , Rats , Rats, Wistar , Signal Transduction/drug effects , Time Factors , Up-Regulation/drug effects
8.
Neurochem Int ; 61(1): 63-71, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22521775

ABSTRACT

Previous studies show that chronic hyperammonemia impairs learning ability of rats by impairing the glutamate-nitric oxide (NO)-cyclic guanosine mono-phosphate (cGMP) pathway in cerebellum. Three types of glutamate receptors cooperate in modulating the NO-cGMP pathway: metabotropic glutamate receptor 5 (mGluR5), (RS)-α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-d-aspartic acid (NMDA) receptors. The aim of this work was to assess whether hyperammonemia alters the modulation of this pathway by mGluR5 and AMPA receptors in cerebellum in vivo. The results support that in control rats: (1) low AMPA concentrations (0.1mM) activate nearly completely Ca(2+)-permeable (glutamate receptor subunit 2 (GluR2)-lacking) AMPA receptors and the NO-cGMP pathway; (2) higher AMPA concentrations (0.3 mM) also activate Ca(2+)-impermeable (GluR2-containing) AMPA receptors, leading to activation of NMDA receptors and of NO-cGMP pathway. Moreover, the data support that chronic hyperammonemia: (1) reduces glutamate release and activation of the glutamate-NO-cGMP pathway by activation of mGluR5; (2) strongly reduces the direct activation by AMPA receptors of the NO-cGMP pathway, likely due to reduced entry of Ca(2+) through GluR2-lacking, high affinity AMPA receptors; (3) strongly increases the indirect activation of the NO-cGMP pathway by high affinity AMPA receptors, likely due to increased entry of Na(+) through GluR2-lacking AMPA receptors and NMDA receptors activation; (4) reduces the indirect activation of the NO-cGMP pathway by low affinity AMPA receptors, likely due to reduced activation of NMDA receptors.


Subject(s)
Cyclic GMP/metabolism , Glutamic Acid/metabolism , Hyperammonemia/metabolism , Nitric Oxide/metabolism , Receptors, AMPA/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Blotting, Western , Chronic Disease , Citrulline/metabolism , Male , Rats , Rats, Wistar , Receptor, Metabotropic Glutamate 5 , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
9.
Neurochem Int ; 60(8): 809-16, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22426201

ABSTRACT

Although commercial production of polychlorinated biphenyls (PCBs) was banned in 1979, PCBs continue to be an environmental and health concern due to their high bioaccumulation and slow degradation rates. In fact, PCBs are still present in our food supply (fish, meat, and dairy products). In laboratory animals, exposure to single PCB congener or to mixtures of different congeners induces a variety of physiological alterations. PCBs cross the placenta and even exposure at low level is harmful for the foetus by leading to neurodevelopment alterations. Serotonin system which regulates many physiological functions from platelet activation to high cerebral processes and neurodevelopment is one of the targets of PCBs toxicity. The effects of PCBs exposure on serotonin system have been investigated although to a lesser extent compared to its effect in other neurotransmitter systems. This review provides a summary of the results concerning the impact of PCBs exposure (in vitro and in vivo) on serotonin system. Further research is needed to correlate specific deficits with PCB-induced changes in the serotonin system.


Subject(s)
Environmental Exposure , Polychlorinated Biphenyls/toxicity , Serotonin/metabolism , Animals , Brain/metabolism , Humans
10.
Metab Brain Dis ; 27(1): 37-49, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22083566

ABSTRACT

Hepatic encephalopathy (HE) is a neurological disease associated with hepatic dysfunction. Current knowledge suggests that hyperammonemia, related to liver failure, is a main factor contributing to the cerebral alterations in HE and that hyperammonemia might impair signal transduction associated with post-translational modification of proteins such as tyrosine-nitration and phosphorylation. However, the molecular bases of the HE remain unclear and very little is known about the occurrence of post-translational modification on in vivo proteins. In this exploratory study we look for evidence of post-translation modifications of proteins in the cerebellum of experimental HE rat models using a proteomic approach. For the first time we showed that hyperammonemia without liver failure (HA rats) and experimental HE with liver failure due to portacaval shunt (PCS rats) lead to a reduced protein nitration in rat cerebellum, where the undernitrated proteins were involved in energy metabolism and cytoskeleton remodelling. Moreover we showed that tyrosine nitration loss of these proteins was not necessarily associated to a change in their phosphorylation state as result of the disease. Interestingly the rat cerebellum phosphoproteome was mainly perturbed in PCS rats, whereas HA rats did not shown appreciable changes in their phosphoprotein profile. Since the protein nitration level decreased similarly in the cerebellum of both HA and PCS rats, this implies that the two disease models share common effects but also present some differential signalling effects in the cerebellum of the same animals. This study highlights the interest for studying the concerted action of multiple signalling pathways in HE development.


Subject(s)
Ammonia/metabolism , Cerebellum/metabolism , Hepatic Encephalopathy/metabolism , Hyperammonemia/metabolism , Proteomics/methods , Ammonia/adverse effects , Animals , Cerebellum/pathology , Cognition/drug effects , Disease Models, Animal , Hepatic Encephalopathy/etiology , Humans , Hyperammonemia/complications , Male , Models, Animal , Nitro Compounds/metabolism , Phosphorylation , Portacaval Shunt, Surgical/adverse effects , Protein Processing, Post-Translational , Rats , Rats, Wistar , Signal Transduction , Tyrosine/metabolism
11.
Clin Transl Med ; 1(1): 30, 2012 Nov 23.
Article in English | MEDLINE | ID: mdl-23369339

ABSTRACT

Perivascular adult stem cells have been isolated from several tissues, including the adult human brain. They have unique signatures resembling both pericytes and mesenchymal stem cells. Understanding the nature of these cells in their specific vascular niches is important to determine their clinical potential as a new adult stem cell source. Indeed, they have promising features in vitro in terms of multipotency, immunomodulation and secretion of growth factors and cytokines. However, their in vivo function is less known as yet. Recent emerging data show a crucial role of perivascular mesenchymal stem cells in tissue homeostasis and repair. Furthermore, these cells may play an important role in adult stem cell niche regulation and in neurodegeneration. Here we review the recent literature on perivascular mesenchymal stem cells, discuss their different in vitro functions and highlight especially the specific properties of brain-derived perivascular mesenchymal stem cells. We summarize current evidence that suggests an important in vivo function of these cells in terms of their regenerative potential that may indicate a new target cell for endogenous tissue regeneration and repair.

12.
Toxicol Sci ; 123(1): 170-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21673325

ABSTRACT

Developmental exposure to polychlorinated biphenyls (PCBs) has been associated with cognitive deficits in humans and laboratory animals by mechanisms that remain unknown. Recently, it has been shown that developmental exposure to 2,2',3,4,4',5'-hexachlorobiphenyl (PCB138), a food-relevant PCB congener, decreases the learning ability of young rats. The aim of this study was to characterize the effect of perinatal exposure to PCB138 on the brain proteome profile in young rats in order to gain insight into the mechanisms underlying PCB138 neurotoxicity. Comparison of the cerebellum proteome from 3-month-old unexposed and PCB138-exposed male offspring was performed using state-of-the-art label-free semiquantitative mass spectrometry method. Biological pathways associated with Ca(2+) homeostasis and androgen receptor signaling pathways were primarily disrupted. These perturbations may contribute toward a premature ageing-like proteome profile of the cerebellum that is triggered by PCB138 exposure in males. Our proteomic data provide insights into the phenomena that may be contributing to the PCB138 neurotoxicity effects observed in laboratory rodents and correlate with PCB exposure and decreased cognitive functions in humans. As such, this study highlights the importance of PCB138 as a risk factor in developmental neurotoxicity in laboratory rodents and humans.


Subject(s)
Cerebellum/drug effects , Cognition Disorders/chemically induced , Environmental Pollutants/toxicity , Polychlorinated Biphenyls/toxicity , Proteins/metabolism , Proteomics/methods , Animals , Cerebellum/chemistry , Cerebellum/metabolism , Cognition Disorders/physiopathology , Female , Food Contamination , Male , Maternal Exposure , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Wistar , Tandem Mass Spectrometry
13.
Neurochem Int ; 58(5): 599-604, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21300123

ABSTRACT

Metabotropic glutamate receptors (mGluRs) modulate important processes in cerebellum including long-term depression, which also requires formation of nitric oxide (NO) and cGMP. Some reports suggest that mGluRs could modulate the NO-cGMP pathway in cerebellum. However this modulation has not been studied in detail. The aim of this work was to assess by microdialysis in freely moving rats whether activation of mGluR5 modulates the NO-cGMP pathway in cerebellum in vivo and to analyze the underlying mechanisms. We show that mGluR5 activation increases extracellular glutamate, citrulline and cGMP in cerebellum. Blocking NMDA receptors with MK-801 does not prevent any of these effects, indicating that NMDA receptors activation is not required. However in the presence of MK-801 the effects are more transient, returning faster to basal levels. Blocking AMPA receptors prevents the increase in citrulline and cGMP induced by mGluR5 activation, but not the increase in glutamate. The release of glutamate is prevented by tetrodotoxin but not by fluoroacetate, indicating that glutamate is released from neurons and not from astrocytes. Activation of AMPA receptors increases citrulline and cGMP. These data indicate that activation of mGluR5 induces an increase of extracellular glutamate which activates AMPA receptors, leading to activation of nitric oxide synthase and increased NO, which activates guanylate cyclase, increasing cGMP. The response mediated by AMPA receptors desensitize rapidly. Activation of AMPA receptors also induces a mild depolarization, allowing activation of NMDA receptors which prolongs the duration of the effect initiated by activation of AMPA receptors. These data support that the three types of glutamate receptors: mGluR5, AMPA and NMDA cooperate in the modulation of the grade and duration of activation of the NO-cGMP pathway in cerebellum in vivo. This pathway would modulate cerebellar processes such as long-term depression.


Subject(s)
Cerebellum/metabolism , Cyclic GMP/metabolism , Nitric Oxide/metabolism , Receptors, AMPA/metabolism , Receptors, Metabotropic Glutamate/physiology , Animals , Cerebellum/drug effects , Dizocilpine Maleate/pharmacology , Male , Nitric Oxide/physiology , Rats , Rats, Wistar , Receptor, Metabotropic Glutamate 5 , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Time Factors
14.
Gastroenterology ; 140(2): 638-45, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20977905

ABSTRACT

BACKGROUND & AIMS: Patients with acute liver failure (ALF) often die of intracranial pressure (IP) and cerebral herniation. Main contributors to increased IP are ammonia, glutamine, edema, and blood flow. The sequence of events and underlying mechanisms, as well as the temporal pattern, regional distribution, and contribution of each parameter to the progression of neurologic deterioration and IP, are unclear. We studied rats with ALF to follow the progression of changes in ammonia, glutamine, grade and type (vasogenic or cytotoxic) of edema, blood-brain barrier permeability, cerebral blood flow, and IP. We assessed whether the changes in these parameters were similar between frontal cortex and cerebellum and evaluated the presence, type, and progression of edema in 12 brain areas. METHODS: ALF was induced by injection of galactosamine. The grade and type of edema was assessed by measuring the apparent diffusion coefficient by magnetic resonance imaging. Cerebral blood flow was measured by magnetic resonance and blood-brain barrier permeability by Evans blue-albumin extravasation. RESULTS: Increased IP arises from an early increase of blood-brain barrier permeability in certain areas (including cerebellum but not frontal cortex) followed by vasogenic edema. Ammonia and glutamine then increase progressively, leading to cytotoxic edema in many areas. Alterations in lactate and cerebral blood flow are later events that further increase IP. CONCLUSIONS: Different mechanisms in specific regions of the brain contribute, with different temporal patterns, to the progression of cerebral alterations and IP in ALF.


Subject(s)
Brain Edema/etiology , Cerebrum/physiopathology , Encephalocele/etiology , Intracranial Hypertension/etiology , Liver Failure, Acute/complications , Ammonia/blood , Animals , Blood-Brain Barrier/physiopathology , Brain Edema/physiopathology , Capillary Permeability , Cerebellum/blood supply , Cerebellum/physiopathology , Cerebrum/blood supply , Galactosamine/adverse effects , Glutamine/blood , Intracranial Hypertension/physiopathology , Lactic Acid/blood , Lactic Acid/metabolism , Liver Failure, Acute/chemically induced , Male , Rats , Rats, Wistar
15.
Neurochem Int ; 58(1): 69-77, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21056608

ABSTRACT

Developmental exposure to polychlorinated biphenyls (PCBs) induces motor alterations in humans by unknown mechanisms. It remains unclear whether: (a) all non-dioxin-like (NDL) PCBs are neurotoxic or it depends on the grade of chlorination; (b) they have different neurotoxicity mechanisms; (c) they affect differently males and females. The aims of this work were to assess: (1) whether perinatal exposure to 3 NDL-PCBs with different grades of chlorination, (PCBs 52, 138 or 180) affects differentially motor activity in adult rats; (2) whether the effects are different in males or females and (3) the mechanisms involved in impaired motor activity. Rats were exposed to PCBs from gestational day 7 to post-natal day 21. Experiments were performed when the rats were 4 months-old. PCB52 did not affect motor activity, PCB180 reduced it in males but not in females and PCB138 reduced activity both in males and females. PCB52 or 138 did not affect extracellular dopamine in nucleus accumbens (NAcc). PCB180 increased it both in males and females. Extracellular glutamate in NAcc was reduced by the three PCBs. Activation of metabotropic glutamate receptors (mGluRs) in NAcc increased extracellular dopamine in control rats and in those exposed to PCB52 and reduced dopamine in rats exposed to PCB180. In rats exposed to PCB138 activation of mGluRs increases dopamine in females and reduces it in males. The opposite changes were observed for glutamate. mGluRs activation reduced extracellular glutamate in control rats and in those exposed to PCB52 and increased glutamate in rats exposed to PCB180. In rats exposed to PCB138 activation of mGluRs reduces glutamate in females and increases it in males. The data support that different NDL-PCBs affect differently motor activity. Increased glutamate release in NAcc following activation of mGluRs would be involved in reduced dopamine release and reduced motor activity in rats exposed to PCB138 or 180.


Subject(s)
Motor Activity/drug effects , Polychlorinated Biphenyls/pharmacology , Synaptic Transmission/drug effects , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Brain Chemistry/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Extracellular Space/drug effects , Extracellular Space/metabolism , Female , Glutamic Acid/metabolism , Male , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Microdialysis , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Polychlorinated Biphenyls/chemistry , Quantitative Structure-Activity Relationship , Rats , Receptors, Glutamate/drug effects , Receptors, Glutamate/metabolism , Sex Characteristics
16.
Metab Brain Dis ; 25(1): 39-48, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20195723

ABSTRACT

Cyclic GMP (cGMP) modulates important cerebral processes including some forms of learning and memory. cGMP pathways are strongly altered in hyperammonemia and hepatic encephalopathy (HE). Patients with liver cirrhosis show reduced intracellular cGMP in lymphocytes, increased cGMP in plasma and increased activation of soluble guanylate cyclase by nitric oxide (NO) in lymphocytes, which correlates with minimal HE assessed by psychometric tests. Activation of soluble guanylate cyclase by NO is also increased in cerebral cortex, but reduced in cerebellum, from patients who died with HE. This opposite alteration is reproduced in vivo in rats with chronic hyperammonemia or HE. A main pathway modulating cGMP levels in brain is the glutamate-NO-cGMP pathway. The function of this pathway is impaired both in cerebellum and cortex of rats with hyperammonemia or HE. Impairment of this pathway is responsible for reduced ability to learn some types of tasks. Restoring the pathway and cGMP levels in brain restores learning ability. This may be achieved by administering phosphodiesterase inhibitors (zaprinast, sildenafil), cGMP, anti-inflammatories (ibuprofen) or antagonists of GABAA receptors (bicuculline). These data support that increasing cGMP by safe pharmacological means may be a new therapeutic approach to improve cognitive function in patients with minimal or clinical HE.


Subject(s)
Brain/metabolism , Cyclic GMP/metabolism , Hepatic Encephalopathy/drug therapy , Hepatic Encephalopathy/metabolism , Hyperammonemia/drug therapy , Hyperammonemia/metabolism , Animals , Brain/physiopathology , Glutamic Acid/metabolism , Hepatic Encephalopathy/physiopathology , Humans , Hyperammonemia/physiopathology , Learning Disabilities/drug therapy , Learning Disabilities/metabolism , Learning Disabilities/physiopathology , Nitric Oxide/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , Rats , Signal Transduction/drug effects , Signal Transduction/physiology
17.
Chem Res Toxicol ; 23(4): 813-20, 2010 Apr 19.
Article in English | MEDLINE | ID: mdl-20297801

ABSTRACT

Polychlorinated biphenyls (PCBs) are persistent organic pollutants that accumulate in the food chain and are present in human blood and milk. Children born to mothers exposed to PCBs show cognitive deficits, which are reproduced in rats perinatally exposed to PCBs. It has been proposed that PCB-induced cognitive impairment is due to impairment of the glutamate-nitric oxide (NO)-cGMP pathway. The aim of the present work was to assess whether chronic exposure to the nondioxin-like PCB52, PCB138, or PCB180 alters the function of this pathway in primary cultures of rat cerebellar neurons and to assess whether different PCBs have similar or different mechanisms of action. PCB180 and PCB138 impair the function of the glutamate-NO-cGMP pathway at nanomolar concentrations, and PCB52 impairs the function of the glutamate-NO-cGMP pathway at micromolar concentrations. The mechanisms by which different PCBs impair the function of the glutamate-NO-cGMP pathway are different. Each PCB affects the pathway at more than one step but with different potency and, for some steps, in opposite ways. Exposure to the PCBs alters the basal concentrations of intracellular calcium, NO, and cGMP. The three PCBs increase NO; however, PCB52 and PCB138 increase basal cGMP, while PCB180 decreases it. PCB52 and PCB138 decrease the activation of soluble guanylate cyclase by NO, and PCB180 increases it. Long-term exposure to PCB52, PCB180, or PCB138 reduces the activation of NO synthase and the whole glutamate-NO-cGMP pathway in response to activation of N-methyl-d-aspartate receptors. The EC(50) was 300 nM for PCB52 and 2 nM for PCB138 or PCB180. These results show that chronic exposure to nondioxin like PCBs impairs the function of the glutamate-NO-cGMP pathway in cerebellar neurons by different mechanisms and with different potencies. Impaired function of this pathway would contribute to the cognitive alterations induced by perinatal exposure to PCBs in humans.


Subject(s)
Cerebellum/cytology , Cyclic GMP/metabolism , Environmental Pollutants/toxicity , Glutamic Acid/metabolism , Neurons/metabolism , Nitric Oxide/metabolism , Polychlorinated Biphenyls/toxicity , Animals , Cells, Cultured , Environmental Exposure , Environmental Pollutants/chemistry , Guanylate Cyclase/metabolism , Nitric Oxide Synthase/metabolism , Polychlorinated Biphenyls/chemistry , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Soluble Guanylyl Cyclase
18.
J Neurochem ; 112(4): 1005-14, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20002515

ABSTRACT

Reduced function of the glutamate--nitric oxide (NO)--cGMP pathway is responsible for some cognitive alterations in rats with hyperammonemia and hepatic encephalopathy. Hyperammonemia impairs the pathway in cerebellum by increasing neuronal nitric oxide synthase (nNOS) phosphorylation in Ser847 by calcium-calmodulin-dependent protein kinase II (CaMKII), reducing nNOS activity, and by reducing nNOS amount in synaptic membranes, which reduces its activation following NMDA receptors activation. The reason for increased CaMKII activity in hyperammonemia remains unknown. We hypothesized that it would be as a result of increased tonic activation of NMDA receptors. The aims of this work were to assess: (i) whether tonic NMDA activation receptors is increased in cerebellum in chronic hyperammonemia in vivo; and (ii) whether this tonic activation is responsible for increased CaMKII activity and reduced activity of nNOS and of the glutamate--NO--cGMP pathway. Blocking NMDA receptors with MK-801 increases cGMP and NO metabolites in cerebellum in vivo and in slices from hyperammonemic rats. This is because of reduced phosphorylation and activity of CaMKII, leading to normalization of nNOS phosphorylation and activity. MK-801 also increases nNOS in synaptic membranes and reduces it in cytosol. This indicates that hyperammonemia increases tonic activation of NMDA receptors leading to reduced activity of nNOS and of the glutamate--NO--cGMP pathway.


Subject(s)
Cerebellum/metabolism , Hyperammonemia/pathology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cerebellum/drug effects , Cyclic GMP/metabolism , Disease Models, Animal , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/metabolism , In Vitro Techniques , Male , Microdialysis/methods , Nitrates/metabolism , Nitric Oxide Synthase Type I/metabolism , Nitrites/metabolism , Phosphorylation/physiology , Rats , Rats, Wistar , Serine/metabolism , Statistics, Nonparametric , Subcellular Fractions/enzymology , Threonine/metabolism
19.
Neurochem Int ; 55(1-3): 106-12, 2009.
Article in English | MEDLINE | ID: mdl-19428813

ABSTRACT

Patients with liver diseases (e.g. cirrhosis) may present hepatic encephalopathy (HE), an alteration in cerebral function which is a consequence of previous failure of liver function. Patients with minimal or clinical HE present different levels of cognitive impairment. Hyperammonemia is considered a main contributor to the neurological alterations in HE. Animal models of chronic HE (e.g. rats with portacaval shunts) or of "pure" hyperammonemia also show impaired cognitive function. The studies summarized here show that the impairment of some types of cognitive function in chronic HE is due to the impaired function of the glutamate-nitric oxide-cGMP pathway in brain. Both hyperammonemia and neuroinflammation contribute to the impairment of the pathway and of cognitive function. Treatment of rats with chronic HE or hyperammonemia with inhibitors of phosphodiesterase 5 restores the function of the glutamate-nitric oxide-cGMP pathway and cGMP levels in brain as well as the ability to learn a Y maze conditional discrimination task. The same beneficial effects may be obtained by treating the rats chronically with an anti-inflammatory, ibuprofen. As the function of this pathway is also altered in brain of patients died in HE, this alteration would also contribute to cognitive impairment in patients with HE. Increasing cGMP by using inhibitors of phosphodiesterase 5 (PDE-5) or anti-inflammatories (under safe conditions) would be therefore a new therapeutic approach to improve learning and memory performance in individuals with minimal or clinical HE.


Subject(s)
Cognition/physiology , Hepatic Encephalopathy/drug therapy , Hepatic Encephalopathy/psychology , Hyperammonemia/drug therapy , Hyperammonemia/psychology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cerebellum/physiology , Cyclic GMP/physiology , Hippocampus/drug effects , Hippocampus/physiology , Humans , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Memory/physiology , Motor Skills , Nitric Oxide/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Phosphodiesterase Inhibitors/therapeutic use , Receptors, N-Methyl-D-Aspartate/drug effects
20.
Neurochem Int ; 55(1-3): 113-8, 2009.
Article in English | MEDLINE | ID: mdl-19428814

ABSTRACT

Acute liver failure (ALF) may lead to rapid death unless the patients receive a liver for transplantation. However, the number of livers available is not enough and a number of patients die before a suitable liver is available for transplantation. The liver has a high capacity for regeneration which may allow complete recovery even in patients with severe liver failure. It would be therefore very useful to have procedures to prevent or delay the mechanisms by which ALF leads to death. These mechanisms are no well understood. Progression of ALF leads to multi-organ failure, systemic inflammatory response, hepatic encephalopathy, cerebral oedema and increased intracranial pressure, which seem the most important immediate causes of mortality in patients with ALF. A main contributor to these events is hyperammonemia, due to impaired ammonia detoxification in the liver. Acute hyperammonemia per se leads to death, which is mediated by activation of the NMDA type of glutamate receptors in brain and may be prevented by antagonists blocking these receptors. Acute liver failure also leads to hyperammonemia and excessive activation of NMDA receptors in brain which contributes to ALF-induced death. Sustained blocking of NMDA receptors by continuous administration of the antagonists MK-801 or memantine increases about twice the survival time of rats with severe ALF due to injection of 2.5g/kg of galactosamine. In rats with milder ALF due to injection of 1.5g/kg of galactosamine, blocking NMDA receptors increases the percentage of surviving rats from 23% to 62% and increases about twice the survival time of the rats which die. These data strongly support that blocking NMDA receptors would improve survival of patients with ALF, either by allowing more time for liver regeneration or to get a liver suitable for transplantation.


Subject(s)
Hyperammonemia/pathology , Liver Failure, Acute/pathology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Brain Chemistry/physiology , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Humans , Hyperammonemia/mortality , Liver Failure, Acute/mortality , Rats , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...