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 ; 21(12)2020 Jun 14.
Article in English | MEDLINE | ID: mdl-32545914

ABSTRACT

Tendinopathy is a rare but serious complication of quinolone therapy. Risk factors associated with quinolone-induced tendon disorders include chronic kidney disease accompanied by the accumulation of uremic toxins. Hence, the present study explored the effects of the representative uremic toxins phenylacetic acid (PAA) and quinolinic acid (QA), both alone and in combination with ciprofloxacin (CPX), on human tenocytes in vitro. Tenocytes incubated with uremic toxins +/- CPX were investigated for metabolic activity, vitality, expression of the dominant extracellular tendon matrix (ECM) protein type I collagen, cell-matrix receptor ß1-integrin, proinflammatory interleukin (IL)-1ß, and the ECM-degrading enzyme matrix metalloproteinase (MMP)-1. CPX, when administered at high concentrations (100 mM), suppressed tenocyte metabolism after 8 h exposure and at therapeutic concentrations after 72 h exposure. PAA reduced tenocyte metabolism only after 72 h exposure to very high doses and when combined with CPX. QA, when administered alone, led to scarcely any cytotoxic effect. Combinations of CPX with PAA or QA did not cause greater cytotoxicity than incubation with CPX alone. Gene expression of the pro-inflammatory cytokine IL-1ß was reduced by CPX but up-regulated by PAA and QA. Protein levels of type I collagen decreased in response to high CPX doses, whereas PAA and QA did not affect its synthesis significantly. MMP-1 mRNA levels were increased by CPX. This effect became more pronounced in the form of a synergism following exposure to a combination of CPX and PAA. CPX was more tenotoxic than the uremic toxins PAA and QA, which showed only distinct suppressive effects.


Subject(s)
Ciprofloxacin/adverse effects , Interleukin-1beta/genetics , Phenylacetates/adverse effects , Quinolinic Acid/adverse effects , Tenocytes/cytology , Adult , Cell Survival/drug effects , Cells, Cultured , Collagen Type I/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Male , Matrix Metalloproteinase 1/genetics , Tenocytes/drug effects , Tenocytes/metabolism
2.
Eur J Pharmacol ; 934: 175316, 2022 Nov 05.
Article in English | MEDLINE | ID: mdl-36209926

ABSTRACT

Huntington disease (HD), an autosomal dominant neurodegenerative disorder characterized by involuntary choreatic movements with cognitive and behavioral disturbances. HD striatum has increased conversion of kynurenine to quinolinic acid (QA) which activates NMDA receptors leading to activation of microglia and increased levels of nuclear factor kappa B (NF-κB) leading to elevated transcription of inducible nitric oxide synthase (iNOS) and various cytokines causing neuronal death via neuroinflammation, oxidative stress, mitochondrial dysfunction and apoptosis. Therefore, inhibiting IKK-NF-κB pathway induced excitotoxicity, oxidative stress and neuroinflammation could be a potential intervention in slowing down the disease progression. QA injection intrastriatally (IS-QA) produce damage mimicking HD where neuroinflammation, oxidative stress and mitochondrial dysfunction play crucial role. Ellagic acid (EA) and vanillic acid (VA) are well reported to possess antioxidant and NF-κB inhibiting effect. Hence, in present study, rats administered IS-QA were treated with EA and VA for 21 days to explore their neuroprotective effects. Behavioral studies, biochemical estimations for oxidative stress and acetylcholinesterase assay were performed. Mitochondrial function was determined by estimating mitochondrial enzyme complexes; inflammatory markers like TNF-α, IL-6, NF-κB by ELISA and apoptosis by caspase-3 levels. Brain damage was determined by histopathology which revealed their neuroprotective effects. Various doses of EA and VA produced improved motor and cognitive functions, oxidative stress and neuroinflammation were also reduced and mitochondrial functioning was improved. In a nutshell, these results signify improved motor and cognitive functions by EA and VA in QA model of HD, along with declined oxidative stress, mitochondrial dysfunction and neuroinflammation.


Subject(s)
Huntington Disease , Neuroprotective Agents , Animals , Rats , Quinolinic Acid/adverse effects , Huntington Disease/chemically induced , Huntington Disease/drug therapy , Huntington Disease/metabolism , NF-kappa B/metabolism , Caspase 3/metabolism , Ellagic Acid/pharmacology , Ellagic Acid/therapeutic use , Vanillic Acid/pharmacology , Vanillic Acid/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nitric Oxide Synthase Type II/metabolism , Acetylcholinesterase/metabolism , Tumor Necrosis Factor-alpha/metabolism , Antioxidants/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Interleukin-6/metabolism , Kynurenine/metabolism , Oxidative Stress , Anti-Inflammatory Agents/pharmacology
3.
Chem Biol Interact ; 315: 108884, 2020 Jan 05.
Article in English | MEDLINE | ID: mdl-31678113

ABSTRACT

Quinolinic acid (QA) known as a neuro-active metabolite associated with the kynurenine pathway. At high concentrations, QA is often involved in the initiation and development of several human neurologic diseases, like Alzheimer's disease. Because of the QA action as the NMDA receptor, it is considered as a potent excitotoxin in vivo. Since it is probable that different mechanisms are employed by QA, activation of NMDA receptors cannot fully explain the revealed toxicity and it is even believed that there are multiple unknown mechanisms/targets leading to QA cytotoxicity. Herein we report accelerated amyloid oligomerization of 1N4R Tau under the effect of QA, in vitro, then the molecular structure, morphology and toxicity of the protein aggregate were documented by using various theoretical/experimental approaches. The possible mechanism of action of QA-induced Tau oligomerization has also been explored.


Subject(s)
Amyloid/metabolism , Neurotoxins/adverse effects , Protein Aggregates/drug effects , Pyridines/adverse effects , Quinolinic Acid/adverse effects , Alzheimer Disease/metabolism , Humans , Kynurenine/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
4.
PLoS One ; 14(6): e0218130, 2019.
Article in English | MEDLINE | ID: mdl-31181111

ABSTRACT

BACKGROUND: Unresponsiveness to dopaminergic therapies is a key feature in the diagnosis of multiple system atrophy (MSA) and a major unmet need in the treatment of MSA patients caused by combined striatonigral degeneration (SND). Transgenic, alpha-synuclein animal models do not recapitulate this lack of levodopa responsiveness. In order to preclinically study interventions including striatal cell grafts, models that feature SND are required. Most of the previous studies focused on extensive nigral and striatal lesions corresponding to advanced MSA-P/SND. The aim of the current study was to replicate mild stage MSA-P/SND with L-dopa failure. METHODS AND RESULTS: Two different striatal quinolinic acid (QA) lesions following a striatal 6-OHDA lesion replicating mild and severe MSA-P/SND, respectively, were investigated and compared to 6-OHDA lesioned animals. After the initial 6-OHDA lesion there was a significant improvement of motor performance after dopaminergic stimulation in the cylinder and stepping test (p<0.001). Response to L-dopa treatment declined in both MSA-P/SND groups reflecting striatal damage of lateral motor areas in contrast to the 6-OHDA only lesioned animals (p<0.01). The remaining striatal volume correlated strongly with contralateral apomorphine induced rotation behaviour and contralateral paw use during L-dopa treatment in cylinder and stepping test (p<0.001). CONCLUSION: Our novel L-dopa response data suggest that L-dopa failure can be induced by restricted lateral striatal lesions combined with dopaminergic denervation. We propose that this sequential striatal double-lesion model replicates a mild stage of MSA-P/SND and is suitable to address neuro-regenerative therapies aimed at restoring dopaminergic responsiveness.


Subject(s)
Levodopa/pharmacology , Multiple System Atrophy/etiology , Striatonigral Degeneration/drug therapy , Animals , Behavior, Animal/drug effects , Corpus Striatum/pathology , Denervation , Disease Models, Animal , Motor Activity/drug effects , Quinolinic Acid/adverse effects , Rats
5.
Eur J Pharmacol ; 828: 31-41, 2018 Jun 05.
Article in English | MEDLINE | ID: mdl-29577894

ABSTRACT

Huntington's disease (HD) is an inherited complex progressive neurodegenerative disorder with an established etiopathology linked to neuronal oxidative stress and corticostriatal excitotoxicity. Present study explores the effects of glucose-dependent insulinotropic polypeptide (GIP) receptor agonist on the neurobehavioral sequelae of quinolinic acid-induced phenotype of Huntington's disease in rats. Bilateral administration of quinolinic acid (300 nmol/4 µl) to the rat striatum led to characteristic deficits in, locomotor activity, motor coordination, neuromuscular coordination and short-term episodic memory. Therapeutic treatment for 14 days with a stable and brain penetrating GIP receptor agonist, D-Ala2GIP (100 nmol/kg, i.p.), attenuated the neurobehavioral deficits due to quinolinic acid (QA) administration. Protective actions of D-Ala2GIP were sensitive to blockade with a GIP receptor antagonist, (Pro3)GIP (50 nmol/kg, i.p.), indicating specific involvement of GIP receptor signaling pathway. Stimulation of GIP receptor with D-Ala2GIP attenuated lipid peroxidation, evidenced by reduced levels of brain malondialdehyde (MDA), and restoration of reduced glutathione (GSH) levels in brain. Quinolinic acid administration led to significant loss of striatal monoamines, e.g., norepinephrine, epinephrine, serotonin, dopamine, and metabolites, 3,4-Dihydroxyphenylacetic acid (DOPAC), homovanillic acid (HVA) and 5-Hydroxyindoleacetic acid (5-HIAA). D-Ala2GIP attenuated the QA-induced depletion of striatal monoamines, without affecting the monoamine degradation pathways. Thus, observed effects with D-Ala2GIP in the QA-induced Huntington's disease model could be attributable to reduction in lipid peroxidation, restoration of endogenous antioxidants and decreased striatal monoamine levels. These findings together suggest that stimulation of GIP receptor signaling pathway in brain could be a potential therapeutic strategy in the symptomatic management of Huntington's disease.


Subject(s)
Behavior, Animal/drug effects , Biogenic Monoamines/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Huntington Disease/metabolism , Lipid Peroxidation/drug effects , Neostriatum/metabolism , Receptors, Gastrointestinal Hormone/agonists , Animals , Body Weight/drug effects , Disease Models, Animal , Drug Interactions , Gait/drug effects , Gastric Inhibitory Polypeptide/chemistry , Glutathione/metabolism , Hand Strength , Huntington Disease/chemically induced , Huntington Disease/physiopathology , Locomotion/drug effects , Male , Malondialdehyde/metabolism , Memory, Episodic , Neostriatum/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Quinolinic Acid/adverse effects , Rats , Rats, Wistar
6.
FASEB J ; 20(7): 1021-3, 2006 May.
Article in English | MEDLINE | ID: mdl-16571773

ABSTRACT

According to the "indirect" excitotoxicity hypothesis, mitochondrial defects increase Ca2+ entry into neurons by rendering NMDA-R hypersensitive to glutamate. We tested this hypothesis by investigating in the rat striatum and cultured striatal cells how partial mitochondrial complex II inhibition produced by 3-nitropropionic acid (3NP) modifies the toxicity of the NMDA-R agonist quinolinate (QA). We showed that nontoxic 3NP treatment, leading to partial inhibition of complex II activity, greatly exacerbated striatal degeneration produced by slightly toxic QA treatment through an "all-or-nothing" process. The potentiation of QA-induced cell death by 3NP was associated with increased calpain activity and massive calpain-mediated cleavage of several postsynaptic proteins, suggesting major neuronal Ca2+ deregulation in the striatum. However, Ca2+ anomalies probably do not result from NMDA-R hypersensitivity. Indeed, brain imaging experiments using [(18)F]fluorodeoxyglucose indirectly showed that 3NP did not increase QA-induced ionic perturbations at the striatal glutamatergic synapses in vivo. Consistent with this, the exacerbation of QA toxicity by 3NP was not related to an increase in the QA-induced entry of 45Ca2+ into striatal neurons. The present results demonstrate that the potentiation of NMDA-R-mediated excitotoxicity by mitochondrial defects involves primarily intracellular Ca2+ deregulation, in the absence of NMDA-R hypersensitivity.


Subject(s)
Calcium Signaling/physiology , Corpus Striatum/metabolism , Mitochondria/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Calpain/metabolism , Cells, Cultured , Corpus Striatum/drug effects , Corpus Striatum/pathology , Male , Mitochondria/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Nitro Compounds/pharmacology , Propionates/pharmacology , Quinolinic Acid/adverse effects , Rats , Rats, Inbred Lew
7.
Sci Rep ; 7: 43642, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28276430

ABSTRACT

Quinolinic acid, a macrophage/microglia-derived excitotoxin fulfills a plethora of functions such as neurotoxin, gliotoxin, and proinflammatory mediator, and it alters the integrity and cohesion of the blood-brain barrier in several pathophysiological states. Beta-trace protein (BTP), a monomeric glycoprotein, is known to indicate cerebrospinal fluid leakage. Thus, the prior aim of this study was to investigate whether BTP might non-invasively indicate quinolinic acid-induced impaired blood-brain barrier integrity. The research hypotheses were tested in three subsamples with different states of immune activation (patients with HCV-infection and interferon-α, patients with major depression, and healthy controls). BTP has also been described as a sensitive marker in detecting impaired renal function. Thus, the renal function has been considered. Our study results revealed highest quinolinic acid and highest BTP- levels in the subsample of patients with HCV in comparison with the other subsamples with lower or no immune activation (quinolinic acid: F = 21.027, p < 0.001 [ANOVA]; BTP: F = 6.792, p < 0.01 [ANOVA]). In addition, a two-step hierarchical linear regression model showed that significant predictors of BTP levels are quinolinic acid, glomerular filtration rate and age. The neurotoxin quinolinic acid may impair blood-brain barrier integrity. BTP might be a new non-invasive biomarker to indicate quinolinic acid-induced impaired blood-brain barrier integrity.


Subject(s)
Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Intramolecular Oxidoreductases/metabolism , Lipocalins/metabolism , Quinolinic Acid/adverse effects , Adult , Biomarkers , Blood-Brain Barrier/immunology , Female , Glomerular Filtration Rate , Humans , Intramolecular Oxidoreductases/blood , Intramolecular Oxidoreductases/immunology , Lipocalins/blood , Lipocalins/immunology , Male , Middle Aged , Neurotoxins/adverse effects , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/metabolism
8.
Neurosci Res ; 55(4): 397-402, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16766071

ABSTRACT

The excitotoxicity induced by QA has been related to its ability to increase free radical content and oxidative stress. In order to investigate the time course of toxicity and oxidative profile in the mice hippocampus following seizures induced by QA infusion (36.8 nM, i.c.v.), we evaluated the cellular damage (PI uptake assay), content of ROS formation (DCF assay) and the total radical antioxidant potential (TRAP) and reactivity (TAR) levels. The present results showed that a cellular damage occurred as early as 4 h after QA infusion coincident with an increase in the ROS contents, which returned to control levels after 24 h, while the cellular damage persisted for 72 h. There was a marked increased in the total antioxidant capacity at 8 h after QA infusion in both reactivity and potential levels. By 72 h post-treatment, the TRAP levels decreased, but the TAR levels remained augmented. Therefore, the delayed and persistent increase in the antioxidant capacity after QA insult may be a cellular adaptative response, probably contributing to decrease the ROS levels in order to prevent the spreading of the cellular damage. Therefore, the increase in the QA level in the brain ventricle may induce oxidative stress, which is followed by a persistent response in the antioxidant system in the hippocampus. The present study may, therefore, contribute to elucidate the mechanism of the brain dysfunction in patients with several neurological disorders involving elevation of QA in the CSF.


Subject(s)
Encephalitis/metabolism , Hippocampus/metabolism , Neurodegenerative Diseases/metabolism , Oxidative Stress/physiology , Quinolinic Acid/adverse effects , AIDS Dementia Complex/complications , AIDS Dementia Complex/metabolism , AIDS Dementia Complex/physiopathology , Animals , Antioxidants/metabolism , Cells, Cultured , Disease Models, Animal , Encephalitis/chemically induced , Encephalitis/physiopathology , Free Radicals/metabolism , Hippocampus/drug effects , Hippocampus/physiopathology , Injections, Intraventricular , Male , Mice , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/physiopathology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Seizures/etiology , Seizures/metabolism , Seizures/physiopathology , Time Factors , Up-Regulation/physiology
9.
Neurosci Lett ; 202(1-2): 109-12, 1995 Dec 29.
Article in English | MEDLINE | ID: mdl-8787843

ABSTRACT

Striatal quinolinate lesions mimic many of the neuropathological characteristics of Huntington's disease. This excitotoxicity is mediated by combined activity of N-methyl-D-aspartate and metabotropic glutamate receptors (mGluRs). Using recently developed phenylglycine derivatives, (S)-4-carboxy-3-hydroxyphenylglycine ((S)-4C3HPG) and (+)-alpha-methyl-4-carboxyphenylglycine ((+)-MCPG), we investigated the role of the different sub-classes of mGluRs in the in vivo excitotoxic process. (S)-4C3HPG (500 and 1000 nmol), co-injected with quinolinic acid, significantly reduced lesion volumes by 52 and 89%, respectively, whereas the same doses of (+)-MCPG had no effect on lesion size. The differential actions of these two drugs at Group 1 and Group 2 metabotropic receptors may explain their differential effects. These observations confirm the important role of mGluRs in excitotoxicity and identify them as promising targets for intervention.


Subject(s)
Glycine/analogs & derivatives , Neostriatum/drug effects , Receptors, Metabotropic Glutamate/physiology , Animals , Benzoates/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Glycine/pharmacology , Male , Neostriatum/pathology , Neostriatum/ultrastructure , Neurons/drug effects , Neurons/ultrastructure , Neuroprotective Agents/pharmacology , Neurotoxins/pharmacology , Quinolinic Acid/adverse effects , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors
10.
J Neurosurg ; 89(2): 267-74, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9688122

ABSTRACT

OBJECT: The goal of this study was to investigate the effect of the severity of host neural damage on the morphological development of intrastriatal transplants in a rodent model of Huntington's disease. METHODS: Sprague-Dawley rats were subjected to unilateral striatal lesioning induced by administration of quinolinic acid (20 nM, 40 nM, or 90 nM). Seven days postlesioning, intrastriatal cell suspension grafts were placed in the right striatum in some of these animals. Grafts were also placed in the right striatum of additional animals that had not been subjected to lesioning. The rats were killed and processed for morphological analysis 8 weeks after grafting. The results indicate that striatal grafts survive and grow much better when implanted into a lesioned striatum rather than into an intact striatum, as measured both by the volume and the numbers of medium-sized spiny neurons within the graft. Only a small or modest lesion is necessary to produce this effect. By some measures (such as graft volume) grafts survive less well when the lesion is more extensive. The presence of a graft reduced the extent of striatal atrophy induced by the lesions, but this effect was not caused by differences in the numbers of surviving neurons per se. CONCLUSIONS: These results have significant implications for the timing of surgical intervention and patient selection with respect to current and future clinical trials of striatal transplantation in the treatment of Huntington's disease.


Subject(s)
Corpus Striatum/pathology , Huntington Disease/pathology , Acetylcholinesterase/analysis , Animals , Atrophy , Cell Count , Cell Survival , Cell Transplantation , Corpus Striatum/cytology , Corpus Striatum/transplantation , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32 , Enzyme Inhibitors/analysis , Female , Follow-Up Studies , Graft Survival , Huntington Disease/surgery , Nerve Degeneration/pathology , Nerve Degeneration/surgery , Nerve Tissue Proteins/analysis , Neurons/pathology , Phosphoproteins/analysis , Quinolinic Acid/adverse effects , Rats , Rats, Sprague-Dawley , Time Factors , Tyrosine 3-Monooxygenase/analysis
11.
Laryngoscope ; 104(2): 176-81, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8302120

ABSTRACT

Quinolinic acid (QUIN) is an endogenous metabolite that exerts a neurotoxic effect by binding to specific neuronal receptors. Studies involving a broad spectrum of infectious and inflammatory central nervous system diseases have suggested a role for QUIN in causing neuronal injury. Since there is evidence for presence of the QUIN receptor in mammalian cochleas, QUIN was measured in middle ear effusions (MEEs). Gas chromatography/mass spectrometry detected QUIN in each of 65 diluted human MEEs, with a mean of 482 +/- 75 (SEM) nmol/L and a range from 15 to 2667 nmol/L. QUIN was also detected in each of 197 chinchilla MEEs from five different models of otitis media, with a mean of 10.6 +/- 1.3 (SEM) mumol/L and a range from 0.23 to 146.0 mumol/L (corrected for dilution). To determine whether QUIN causes sensorineural hearing loss (SNHL), QUIN solutions were placed on round window membranes (RWM) for 20 to 240 minutes, in 20 chinchillas. SNHL was detected by electrocochleography in QUIN-exposed animals, but not in saline controls. We conclude that QUIN is present in MEEs and that QUIN in the middle ear has the potential to cross the RWM and cause sensorineural hearing loss, possibly by binding to specific neuronal receptors in mammalian cochleas.


Subject(s)
Cochlea/innervation , Hearing Loss, Sensorineural/chemically induced , Neurons/drug effects , Otitis Media with Effusion/metabolism , Quinolinic Acid/adverse effects , Animals , Audiometry, Evoked Response , Child , Chinchilla , Gas Chromatography-Mass Spectrometry , Hearing Loss, Sensorineural/diagnosis , Humans , In Vitro Techniques , Otitis Media with Effusion/complications , Quinolinic Acid/analysis , Round Window, Ear/drug effects
12.
Rev Neurol ; 30(11): 1016-21, 2000.
Article in Spanish | MEDLINE | ID: mdl-10904945

ABSTRACT

INTRODUCTION: Huntington's disease (HD) is a progressive neurodegenerative disorder, characterized by severe degeneration of basal ganglia neurons. Behavioral symptoms of HD include abnormal, uncontrollable and constant choreiform movements, impaired cognitive function and emotional disturbance. OBJECTIVE: In order to explore the changes of cognitive and motor functions induced by quinolinate lesion we realized this experiment. MATERIALS AND METHODS: We studied the behavior of rats with unilateral quinolinate induced lesions of the medial striatum. Intact 3 months old male rats (n = 23) were trained in the Morris Water Maze during three consecutive days, eight trials/day (acquisition), and before surgery they were randomly assigned either to intact or lesion groups. Fifteen days after the lesion the rats were tested using retention test (one day/four trials, with the escape platform in the same position as in acquisition test), on the next three days the rats were tested in the transfer test (three days/eight trials-day, with the platform in the new position). The Paw reaching test and the asymmetrical rotational behavior test in respond to amphetamine were also tested in these rats. RESULTS: Lesioned animals exhibited deficient retrieval of stored memories of visuospatial skills and impaired transfer of learning. In relation with motor activity the lesioned rats showed a profound impairment in the skill of the left forelimb for reaching food compared with its right forelimb as well as with the forelimb abilities of intact rats. The lesioned animals showed significant rotational behavior induced by amphetamine agonist, ipsilateral to the lesioned striatum. CONCLUSIONS: These results are consistent with the notion that the striatal degeneration could sufficiently account for the cognitive abnormalities associated with HD, and with the key role played by basal ganglia in enabling voluntary and postural adjustment of the movements.


Subject(s)
Behavior, Animal/drug effects , Disease Models, Animal , Excitatory Amino Acid Agonists/adverse effects , Huntington Disease/chemically induced , Quinolinic Acid/adverse effects , Animals , Cognition/drug effects , Corpus Striatum/metabolism , Excitatory Amino Acid Agonists/pharmacokinetics , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Locomotion/drug effects , Male , Maze Learning/drug effects , Quinolinic Acid/pharmacokinetics , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/drug effects , Retention, Psychology/drug effects , Spatial Behavior/drug effects , Visual Perception/drug effects
13.
J Pharm Pharmacol ; 66(9): 1294-302, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24707860

ABSTRACT

OBJECTIVES: Aloysia gratissima aqueous extract (AE) was investigated as a putative protective agent against quinolinic acid (QA)-induced seizures in mice and hippocampal cell damage. Additionally, AE and ferulic acid (FA), the major compound of AE, were tested against neurotoxicity evoked by glutamate or its N-methyl-D-aspartate receptor (NMDAR) agonist, QA on hippocampal slices, in vitro. METHODS: Mice were treated with AE before QA infusion (36.8 nmol/site) and seizures were analysed. Cellular viability and modulation of excitatory amino acid transport were verified in hippocampal slices. In-vitro AE or FA was tested against neurotoxicity induced by glutamate or QA. KEY FINDINGS: AE did not prevent QA-induced seizures; however, it prevented cellular death and disruption of excitatory amino acid transport. In-vitro AE (0.1 or 1.0 mg/ml) or FA (1 or 10 µm), improved cell viability against citotoxicity exerted by glutamate or QA, respectively. Both AE and FA have protective effects depending on activation of the phosphatidylinositol-3 kinase (PI3K) signalling pathway. CONCLUSIONS: AE attenuated QA-induced cell damage possibly involving the glutamate transport modulation through NMDAR interaction. FA shows a similar profile of neuroprotection promoted by AE. Therefore, AE treatment might be a useful strategy in preventing brain damage caused by exacerbation of glutamatergic toxicity in nervous system disorders.


Subject(s)
Glutamic Acid/adverse effects , Hippocampus/drug effects , Neurotoxicity Syndromes/drug therapy , Phytotherapy , Plant Extracts/therapeutic use , Quinolinic Acid/adverse effects , Verbenaceae/chemistry , Animals , Biological Transport , Cell Death/drug effects , Cell Survival/drug effects , Coumaric Acids/pharmacology , Coumaric Acids/therapeutic use , Excitatory Amino Acid Agonists/adverse effects , Excitatory Amino Acids/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Male , Mice, Inbred Strains , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Phosphatidylinositol 3-Kinase/metabolism , Plant Extracts/pharmacology , Receptors, N-Methyl-D-Aspartate/agonists , Seizures/chemically induced , Seizures/metabolism
14.
FEBS J ; 279(8): 1355, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22251552

ABSTRACT

This minireview series reviews some of the most recent findings about quinolinic acid's cellular toxicity and its implications in diseases such as HIV associated neurocognitive disorders, depressive disorders and schizophrenia, and finally therapeutic strategies with drugs able to interfere with quinolinic acid production and/or effects.


Subject(s)
Neurotoxicity Syndromes/etiology , Quinolinic Acid/adverse effects , Animals , Humans
15.
FEBS J ; 279(8): 1356-65, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22248144

ABSTRACT

Over the last two decades, evidence for the involvement of quinolinic acid (QUIN) in neuroinflammatory diseases has been exponentially increasing. Within the brain, QUIN is produced and released by infiltrating macrophages and activated microglia, the very cells that are prominent during neuroinflammation. QUIN acts as an agonist of the N-methyl-D-aspartate receptor and as such is considered to be a brain endogenous excitotoxin. Since the discovery of the excitotoxic activity of QUIN in the early 1980s, several other cytotoxic mechanisms have been identified. We know today that QUIN acts as a neurotoxin, gliotoxin, proinflammatory mediator, pro-oxidant molecule and can alter the integrity and cohesion of the blood-brain barrier. This paper aims to review some of the most recent findings about the effects of QUIN and its mode of action.


Subject(s)
Neurotoxicity Syndromes/etiology , Neurotoxins/adverse effects , Quinolinic Acid/adverse effects , Animals , Humans
16.
São Paulo; s.n; s.n; 2022. 112 p. graf.
Thesis in Portuguese | LILACS | ID: biblio-1397184

ABSTRACT

A Doença de Huntington (Huntington's disease - HD) trata-se de uma patologia neurodegenerativa hereditária caracteriza por meio da expressão das proteínas huntingtinas mutantes (mHtt), das mortes dos neurônios espinhais médios (medium spiny neurons MSNs) GABAérgicos D2-positivos do striatum e da hipercinesia. Uma hipótese se refere à função das mHtts de potencializarem os efeitos excitotóxicos das estimulações dos receptores de NMDA (NMDAR) por meio da inibição da succinato desidrogenase, resultando em desequilibrio das [Ca2+]i, estresse oxidativo e apoptose. A adenosina agonista dos receptores purinérgicos P1 tem sido descrita por conta das suas funções neuroprotetoras e neuromodulatórias. Assim, estabelecemos dois modelos in vitro da HD fundamentados nas neurodiferenciações das linhagens murinas de célula-tronco embrionárias E14-TG2a e progenitoras neurais do hipocampo HT-22; seguidas pelos tratamentos com ácido quinolínico (QA) agonista seletivo dos NMDARs , na ausência e na presença do ácido 3-nitropropiônico (3-NP) inibidor irreversível da succinato desidrogenase. Estes modelos foram utilizados nas avaliações das funções neuroprotetoras da adenosina. Os neurônios pós-mitóticos das culturas de E14-TG2a diferenciadas foram caracterizados conforme os MSNs GABAérgicos do striatum; enquanto os neurônios HT-22 diferenciados foram caracterizados de modo inespecífico. Metodologia: imunofluorescência (microscopia e citometria); PCR em tempo real; análise das variações dos potenciais das membranas plasmáticas e das variações transientes das [Ca2+]i por microfluorimetria; e quantificações das reduções do AlamarBlue® (% de sobrevida celular) e das atividades extracelulares de LDH (U/L) (necrose) por espectrometria. Avaliamos a capacidade do 3-NP de potencializar os efeitos excitotóxicos do QA comparando dois grupos de neurônios HT-22 diferenciados: QA 8mM (EC50) (controle); e 3-NP 5mM/QA 8mM. Avaliarmos o potencial neuroprotetor da adenosina comparando quatro grupos de neurônios HT-22 diferenciados: QA 8mM; adenosina 250µM/QA 8mM; 3-NP 5mM/QA 8mM; 3-NP 5mM/adenosina 250µM/QA 8mM. Os neurônios pós-mitóticos derivados das E14TG2a foram classificados como MSNsGABAérgicos do striatum integrantes de uma cultura neuronal heterogênea semelhante às conexões nigroestriatais, corticoestriatais, striatonigral e striatopallidal. Os neurônios HT-22 diferenciados perfaziam uma cultura neuronal heterogênea, não totalmente madura, composta por neurônios glutamatérgicos, dopaminérgicos, colinérgicos e GABAérgicos. Os neurônios HT-22 diferenciados 3-NP 5mM apresentaram menores % de sobrevida celular após os tratamentos com QA 8mM por 24h (p<0.05); e maiores amplitudes das variações das [Ca2+]i dependentes do QA 8mM (p<0.05) (cinética 6 minutos). Por outro lado, os neurônios HT-22 diferenciados pré- tratados com 3-NP 5mM apresentaram menores atividades extracelulares de LDH após o tratamento com QA 8mM por 24h menor proporção de necrose. Os pré-tratamentos com adenosina 250µM indicaram uma tendência dos efeitos neuroprotetores (p>0.05) maiores % de sobrevida celular; menores atividades extracelulares de LDH; e menores amplitudes das variações transientes das [Ca2+]i. Em conjunto, nossos resultados indicam que a inibição da succinato desidrogenase potencializa os efeitos excitotóxicos dos NMDARs por meio da alteração das [Ca2+]i e, provavelmente, dos mecanismos de morte celular; enquanto a adenosina apenas tendeu à neuroproteção


Huntington's disease (HD) is a hereditary neurodegenerative pathology characterized by mutant huntingtin proteins (mHtt) expression, striatum D2-positive GABAergic medium spiny neurons (MSNs) cell death and hyperkinetic motor symptoms development. One hypothesis refers to the principle that mHtt potentiates the excitotoxic effects of NMDA receptor (NMDAR) stimulation by the inhibition of mitochondrial succinate dehydrogenase, resulting in [Ca2+]i imbalance, oxidative stress and apoptosis. Adenosine P1 purinergic receptor agonist is related to neuroprotective and neuromodulatory functions. Thus, we established two in vitro HD models based on the neurodifferentiation of murine embryonic stem cell lines E14-TG2a and hippocampal neuroprogenitor cell line HT-22 followed by treatment with quinolinic acid (QA) selective agonist of NMDARs , in the absence and in the presence of 3-nitropropionic acid (3-NP) irreversible inhibitor of succinate dehydrogenase. These models were used to assess the neuroprotective functions of adenosine. Post-mitotic neurons from differentiated E14-TG2a cultures were characterized according to striatum's GABAergic MSNs; while the differentiated HT-22 neurons were characterized in a non-specific way. Methodology included immunofluorescence (microscopy and cytometry); real-time PCR; analysis of variations in the plasma membrane potentials and of transient variations in the [Ca2+]i by microfluorimetry; and quantification of AlamarBlue® reductions (% cell survival) and of extracellular LDH activity (U/L) (necrosis) by spectrometry. We evaluated the ability of 3-NP to potentiate the excitotoxic effects of QA by comparing two groups of differentiated HT-22 neurons: 8mM QA (control); and 5mM 3-NP/8mM QA. We evaluated the neuroprotective potential of adenosine comparing four groups of differentiated HT-22 neurons: QA 8mM; 250µM adenosine/8mM QA; 5mM 3-NP/8mM QA; 5mM 3-NP/250µM adenosine/8mM QA. Postmitotic neurons derived from E14TG2a were classified as striatums GABAergic MSNs that are part of a heterogeneous neuronal culture similar to nigrostriatal, corticostriatal, striatonigral, and striatopallidal connections. Differentiated HT-22 neurons consisted of a heterogeneous neuronal culture and not fully mature glutamatergic,dopaminergic, cholinergic and GABAergic neurons. Differentiated HT-22 neurons following 5mM 3-NP treatment showed lower % cell survival after treatments with 8mM QA for 24h (p<0.05); and higher amplitudes of the variations of [Ca2+]i induced by 8mM QA (p<0.05) (kinetics 6 minutes). On the other hand, differentiated HT-22 neurons 5mM 3-NP showed lower extracellular LDH activities after treatment with 8mM QA for 24h indicating a lower proportion of necrotic cells. Pretreatments with 250µM adenosine indicated a trend towards neuroprotective effects, such as higher percentages of cell survival; lower extracellular LDH activities; and lower amplitudes of transient variations of [Ca2+]i. Taken together, our results indicate that succinate dehydrogenase inhibition potentiated the excitotoxic effects of NMDARs by altering [Ca2+]i and, probably, cell death mechanisms, while adenosine only to neuroprotection


Subject(s)
In Vitro Techniques/methods , Quinolinic Acid/adverse effects , Huntington Disease/pathology , Models, Anatomic , Spectrum Analysis/methods , Adenosine/agonists , Receptors, N-Methyl-D-Aspartate , Neuroprotective Agents/administration & dosage , Absenteeism , Purinergic Agonists/adverse effects
17.
FEBS J ; 279(8): 1366-74, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22260426

ABSTRACT

This brief review will first consider HIV associated neurocognitive disorder followed by the current understanding of its neuropathogenesis. Against this background the role of the kynurenine pathway will be detailed. Evidence both direct and indirect will be discussed for involvement of the kynurenine pathway at each step in the neuropathogenesis of HIV associated neurocognitive disorder.


Subject(s)
Central Nervous System Diseases/etiology , Cognition Disorders/etiology , HIV Infections/complications , HIV/pathogenicity , Kynurenine/metabolism , Quinolinic Acid/adverse effects , Central Nervous System Diseases/metabolism , Cognition Disorders/metabolism , HIV Infections/drug therapy , HIV Infections/metabolism , Humans
18.
Acta Neuropathol ; 90(5): 504-10, 1995.
Article in English | MEDLINE | ID: mdl-8560984

ABSTRACT

To learn about the mechanisms of excitotoxic cell death in vivo, three different excitatory amino acid receptor agonists (kainic acid, quinolinic acid or quisqualic acid) were injected in the left striatum of adult rats. Brains were examined at 24 and 48 h after injection. Morphological and biochemical studies were performed using conventional stains, histochemistry, in situ labelling of nuclear DNA fragmentation, and agarose gel electrophoresis of extracted DNA. Large numbers of cells with cytoplasmic shrinkage and nuclear condensation or granular degeneration of the chromatin, and fewer cells with apoptotic morphology were distributed at random in the injured areas of the three groups of treated animals but not in rats injected with vehicle alone. A ladder pattern, typical of internucleosomal DNA fragmentation, was observed 24 h after treatment. This was replaced by a smear pattern, consistent with random DNA breakdown, at 48 h. These morphological and biochemical results suggest that prevailing necrosis together with apoptosis occur following intrastriatal injection of different excitotoxins.


Subject(s)
Apoptosis , Necrosis/chemically induced , Neurotoxins/adverse effects , Animals , Cell Death , Corpus Striatum/chemistry , Immunohistochemistry , Kainic Acid/adverse effects , Male , Quinolinic Acid/adverse effects , Rats , Rats, Sprague-Dawley
19.
Neurobiol Learn Mem ; 76(1): 106-16, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11525249

ABSTRACT

Rats with quinolinic acid lesions of the medial or lateral caudate-putamen (CPu) and controls were tested for performance of a previously learned auditory conditional response association task. The task involved the selection of two possible responses when presented with one of two different tones. Results indicated that lesions of either the medial or the lateral CPu produced a sustained deficit in the auditory conditional response association task. Only the lateral CPu lesioned rats exhibited transient motor problems immediately following surgery, but these problems did not interfere with the execution of the appropriate responses. It is suggested that both the medial and the lateral CPu are involved in response selection and response separation within egocentric space.


Subject(s)
Association Learning/physiology , Auditory Perception/physiology , Caudate Nucleus/physiopathology , Conditioning, Psychological , Putamen/physiopathology , Animals , Behavior, Animal/physiology , Caudate Nucleus/drug effects , Male , Psychomotor Disorders/chemically induced , Putamen/drug effects , Quinolinic Acid/adverse effects , Rats , Rats, Long-Evans
20.
Psychiatry Clin Neurosci ; 54(1): 83-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-15558884

ABSTRACT

To examine the effects of lesions of the entorhinal cortex on limbic dopamine (DA) metabolism, DA and its metabolites were assayed in five brain regions (the medial prefrontal cortex, anterior cingulate cortex, caudate-putamen, accumbens nucleus, and lateral amygdala), 14 and 28 days after quinolinic acid or sham lesions of the left entorhinal cortex in rats. Concentrations of 3,4-dihydroxyphenylacetic acid (DOPAC) on day 14 in the medial prefrontal cortex, accumbens nucleus, and lateral amygdala of the entorhinal cortex lesioned animals were significantly decreased compared with the controls, but they returned to control levels on day 28. The concentration of DA in the lateral amygdala and spontaneous locomotion to a novel environment were significantly increased on day 28 after the lesion. These results suggest that entorhinal cortex lesions alter mesolimbic dopamine metabolism, particularly in the amygdala.


Subject(s)
Amygdala/chemistry , Dopamine/metabolism , Entorhinal Cortex/pathology , Amygdala/physiology , Animals , Dose-Response Relationship, Drug , Exploratory Behavior , Locomotion , Male , Quinolinic Acid/administration & dosage , Quinolinic Acid/adverse effects , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL