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










Publication year range
1.
Rev Neurosci ; 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38841811

ABSTRACT

Glioblastoma multiforme (GBM) exhibits genetic alterations that induce the deregulation of oncogenic pathways, thus promoting metabolic adaptation. The modulation of metabolic enzyme activities is necessary to generate nucleotides, amino acids, and fatty acids, which provide energy and metabolic intermediates essential for fulfilling the biosynthetic needs of glioma cells. Moreover, the TCA cycle produces intermediates that play important roles in the metabolism of glucose, fatty acids, or non-essential amino acids, and act as signaling molecules associated with the activation of oncogenic pathways, transcriptional changes, and epigenetic modifications. In this review, we aim to explore how dysregulated metabolic enzymes from the TCA cycle and oxidative phosphorylation, along with their metabolites, modulate both catabolic and anabolic metabolic pathways, as well as pro-oncogenic signaling pathways, transcriptional changes, and epigenetic modifications in GBM cells, contributing to the formation, survival, growth, and invasion of glioma cells. Additionally, we discuss promising therapeutic strategies targeting key players in metabolic regulation. Therefore, understanding metabolic reprogramming is necessary to fully comprehend the biology of malignant gliomas and significantly improve patient survival.

2.
Metabolites ; 14(5)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38786726

ABSTRACT

The metabolic reprogramming that promotes tumorigenesis in glioblastoma is induced by dynamic alterations in the hypoxic tumor microenvironment, as well as in transcriptional and signaling networks, which result in changes in global genetic expression. The signaling pathways PI3K/AKT/mTOR and RAS/RAF/MEK/ERK stimulate cell metabolism, either directly or indirectly, by modulating the transcriptional factors p53, HIF1, and c-Myc. The overexpression of HIF1 and c-Myc, master regulators of cellular metabolism, is a key contributor to the synthesis of bioenergetic molecules that mediate glioma cell transformation, proliferation, survival, migration, and invasion by modifying the transcription levels of key gene groups involved in metabolism. Meanwhile, the tumor-suppressing protein p53, which negatively regulates HIF1 and c-Myc, is often lost in glioblastoma. Alterations in this triad of transcriptional factors induce a metabolic shift in glioma cells that allows them to adapt and survive changes such as mutations, hypoxia, acidosis, the presence of reactive oxygen species, and nutrient deprivation, by modulating the activity and expression of signaling molecules, enzymes, metabolites, transporters, and regulators involved in glycolysis and glutamine metabolism, the pentose phosphate cycle, the tricarboxylic acid cycle, and oxidative phosphorylation, as well as the synthesis and degradation of fatty acids and nucleic acids. This review summarizes our current knowledge on the role of HIF1, c-Myc, and p53 in the genic regulatory network for metabolism in glioma cells, as well as potential therapeutic inhibitors of these factors.

3.
Int J Mol Sci ; 24(24)2023 Dec 18.
Article in English | MEDLINE | ID: mdl-38139462

ABSTRACT

Glioma cells exhibit genetic and metabolic alterations that affect the deregulation of several cellular signal transduction pathways, including those related to glucose metabolism. Moreover, oncogenic signaling pathways induce the expression of metabolic genes, increasing the metabolic enzyme activities and thus the critical biosynthetic pathways to generate nucleotides, amino acids, and fatty acids, which provide energy and metabolic intermediates that are essential to accomplish the biosynthetic needs of glioma cells. In this review, we aim to explore how dysregulated metabolic enzymes and their metabolites from primary metabolism pathways in glioblastoma (GBM) such as glycolysis and glutaminolysis modulate anabolic and catabolic metabolic pathways as well as pro-oncogenic signaling and contribute to the formation, survival, growth, and malignancy of glioma cells. Also, we discuss promising therapeutic strategies by targeting the key players in metabolic regulation. Therefore, the knowledge of metabolic reprogramming is necessary to fully understand the biology of malignant gliomas to improve patient survival significantly.


Subject(s)
Glioblastoma , Glioma , Humans , Glioblastoma/genetics , Glioblastoma/metabolism , Glutamine/metabolism , Metabolic Reprogramming , Glycolysis/physiology , Glioma/pathology , Signal Transduction , Apoptosis , Cell Proliferation/physiology
4.
Pharmaceutics ; 15(12)2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38140038

ABSTRACT

Progesterone (P4) is a neuroactive hormone having pleiotropic effects, supporting its pharmacological potential to treat global (cardiac-arrest-related) cerebral ischemia, a condition associated with an elevated risk of dementia. This review examines the current biochemical, morphological, and functional evidence showing the neuroprotective/neurorestorative effects of P4 against global cerebral ischemia (GCI). Experimental findings show that P4 may counteract pathophysiological mechanisms and/or regulate endogenous mechanisms of plasticity induced by GCI. According to this, P4 treatment consistently improves the performance of cognitive functions, such as learning and memory, impaired by GCI. This functional recovery is related to the significant morphological preservation of brain structures vulnerable to ischemia when the hormone is administered before and/or after a moderate ischemic episode; and with long-term adaptive plastic restoration processes of altered brain morphology when treatment is given after an episode of severe ischemia. The insights presented here may be a guide for future basic research, including the study of P4 administration schemes that focus on promoting its post-ischemia neurorestorative effect. Furthermore, considering that functional recovery is a desired endpoint of pharmacological strategies in the clinic, they could support the study of P4 treatment for decreasing dementia in patients who have suffered an episode of GCI.

5.
Rev Neurosci ; 34(8): 915-932, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37409540

ABSTRACT

The transsulfuration pathway (TSP) is a metabolic pathway involving sulfur transfer from homocysteine to cysteine. Transsulfuration pathway leads to many sulfur metabolites, principally glutathione, H2S, taurine, and cysteine. Key enzymes of the TSP, such as cystathionine ß-synthase and cystathionine γ-lyase, are essential regulators at multiple levels in this pathway. TSP metabolites are implicated in many physiological processes in the central nervous system and other tissues. TSP is important in controlling sulfur balance and optimal cellular functions such as glutathione synthesis. Alterations in the TSP and related pathways (transmethylation and remethylation) are altered in several neurodegenerative diseases, including Parkinson's disease, suggesting their participation in the pathophysiology and progression of these diseases. In Parkinson's disease many cellular processes are comprised mainly those that regulate redox homeostasis, inflammation, reticulum endoplasmic stress, mitochondrial function, oxidative stress, and sulfur content metabolites of TSP are involved in these damage processes. Current research on the transsulfuration pathway in Parkinson's disease has primarily focused on the synthesis and function of certain metabolites, particularly glutathione. However, our understanding of the regulation of other metabolites of the transsulfuration pathway, as well as their relationships with other metabolites, and their synthesis regulation in Parkinson´s disease remain limited. Thus, this paper highlights the importance of studying the molecular dynamics in different metabolites and enzymes that affect the transsulfuration in Parkinson's disease.


Subject(s)
Cysteine , Parkinson Disease , Humans , Cysteine/metabolism , Sulfur/metabolism , Cystathionine beta-Synthase/metabolism , Glutathione/metabolism
6.
Life (Basel) ; 13(2)2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36836894

ABSTRACT

Lithium is a therapeutic cation used to treat bipolar disorders but also has some important features as an anti-cancer agent. In this review, we provide a general overview of lithium, from its transport into cells, to its innovative administration forms, and based on genomic, transcriptomic, and proteomic data. Lithium formulations such as lithium acetoacetate (LiAcAc), lithium chloride (LiCl), lithium citrate (Li3C6H5O7), and lithium carbonate (Li2CO3) induce apoptosis, autophagy, and inhibition of tumor growth and also participate in the regulation of tumor proliferation, tumor invasion, and metastasis and cell cycle arrest. Moreover, lithium is synergistic with standard cancer therapies, enhancing their anti-tumor effects. In addition, lithium has a neuroprotective role in cancer patients, by improving their quality of life. Interestingly, nano-sized lithium enhances its anti-tumor activities and protects vital organs from the damage caused by lipid peroxidation during tumor development. However, these potential therapeutic activities of lithium depend on various factors, such as the nature and aggressiveness of the tumor, the type of lithium salt, and its form of administration and dosage. Since lithium has been used to treat bipolar disorder, the current study provides an overview of its role in medicine and how this has changed. This review also highlights the importance of this repurposed drug, which appears to have therapeutic cancer potential, and underlines its molecular mechanisms.

7.
Int J Mol Sci ; 23(20)2022 Oct 11.
Article in English | MEDLINE | ID: mdl-36292947

ABSTRACT

The presence of insoluble aggregates of amyloid ß (Aß) in the form of neuritic plaques (NPs) is one of the main features that define Alzheimer's disease. Studies have suggested that the accumulation of these peptides in the brain significantly contributes to extensive neuronal loss. Furthermore, the content and distribution of cholesterol in the membrane have been shown to have an important effect on the production and subsequent accumulation of Aß peptides in the plasma membrane, contributing to dysfunction and neuronal death. The monomeric forms of these membrane-bound peptides undergo several conformational changes, ranging from oligomeric forms to beta-sheet structures, each presenting different levels of toxicity. Aß peptides can be internalized by particular receptors and trigger changes from Tau phosphorylation to alterations in cognitive function, through dysfunction of the cholinergic system. The goal of this review is to summarize the current knowledge on the role of lipids in Alzheimer's disease and their relationship with the basal cholinergic system, as well as potential disease-modifying therapies.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Humans , Amyloid beta-Peptides/metabolism , Alzheimer Disease/metabolism , Lipid Metabolism , Basal Metabolism , Peptide Fragments/metabolism , Cholinergic Agents , Lipids
8.
Genes (Basel) ; 13(4)2022 03 25.
Article in English | MEDLINE | ID: mdl-35456392

ABSTRACT

The present study evaluated the risk effect of 12 Single Nucleotide Polymorphisms in the SORL1 gene in the Mexican population using Late-Onset Alzheimer's Disease (LOAD) and control subjects. Considering APOE as the strongest genetic risk factor for LOAD, we conducted interaction analyses between single nucleotide polymorphisms (SNPs) and the APOE genotype. METHODS: Patients were interviewed during their scheduled visits at neurologic and geriatric clinics from different institutions. The LOAD diagnosis included neurological, geriatric, and psychiatric examinations, as well as the medical history and neuroimaging. Polymorphisms in SORL1 were genotyped by real-time PCR in 156 subjects with LOAD and 221 controls. APOE genotype was determined in each study subject. Allelic, genotypic, and haplotypic frequencies were analyzed; an ancestry analysis was also performed. RESULTS: The A/A genotype in rs1784933 might be associated with an increased LOAD risk. Two blocks with high degree linkage disequilibrium (LD) were identified. The first block composed by the genetic variants rs668387, rs689021 and rs641120 showed a positive interaction (mainly the rs689021) with rs1784933 polymorphism. Moreover, we found a significant association between the APOE ε4 allele carriers and the variant rs2070045 located in the second LD block. CONCLUSION: The rs1784933 polymorphism is associated with LOAD in Mexican patients. In addition, the presence of APOE ε4 allele and SORL1 variants could represent a genetic interaction effect that favors LOAD risk in the Mexican population. SNPs have been proposed as genetic markers associated with the development of LOAD that can support the clinical diagnosis. Future molecular studies could help understand sporadic Alzheimer's Disease (AD) among the Mexican population, where currently there is a sub-estimate number in terms of disease frequency and incidence.


Subject(s)
Alzheimer Disease , Aged , Alzheimer Disease/epidemiology , Alzheimer Disease/genetics , Apolipoprotein E4/genetics , Humans , LDL-Receptor Related Proteins/genetics , Membrane Transport Proteins/genetics , Mexico , Polymorphism, Single Nucleotide
9.
J Toxicol ; 2021: 9983201, 2021.
Article in English | MEDLINE | ID: mdl-34858496

ABSTRACT

Neonicotinoids are pesticides that act as agonists of nicotinic receptors for acetylcholine in insects' central nervous system (CNS). Chronic exposure to neonicotinoids in humans is related to autism, memory loss, and finger tremor. In this article, we evaluate the effect of subchronic oral administration of two neonicotinoids in the same mixture: clothianidin and thiacloprid. Decreasing doses of both pesticides were administered to rats starting from the lethal dose 50 (LD50) reported by the manufacturer. Our results indicate that the administration of three doses of decreasing amounts of LD50 (5/10, 4/10, and 3/10 LD50) resulted in 100% death in all cases. Ten administration times of 2/10 LD50 of the mixture caused only 20% of death cases after twenty-seven days, which was determined as a subchronic administration scheme. The animals administered 2/10 LD50 showed behavioral alterations after the first and second administration. Electrographic studies showed abnormal discharge patterns in the CNS. 72 h after the tenth dose, learning and memory tests were performed in the Morris water maze. Our results revealed significant decreases in permanence at the quadrant and the number of crosses (P=0.0447, P=0.0193, respectively), which represent alterations in the short-term memory test, but there were no significant changes in a long-term memory test. Likewise, the brains of these animals showed tissue architecture loss, nucleosomal retraction, and a significant increase in the pycnosis of the granular neurons of the dentate gyrus analyzed at 72 h after the last dose (P=0.0125). Toxic effects and cognitive deterioration that have been found in communities living near contaminated areas are probably related to the agricultural use of neonicotinoids.

10.
Neurotoxicology ; 86: 125-138, 2021 09.
Article in English | MEDLINE | ID: mdl-34371026

ABSTRACT

Organisms have metabolic pathways responsible for eliminating endogenous and exogenous toxicants. Generally, we associate the liver par excellence as the organ in charge of detoxifying the body; however, this process occurs in all tissues, including the brain. Due to the presence of the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB), the Central Nervous System (CNS) is considered a partially isolated organ, but similar to other organs, the CNS possess xenobiotic transporters and metabolic pathways associated with the elimination of xenobiotic agents. In this review, we describe the different systems related to the detoxification of xenobiotics in the CNS, providing examples in which their association with neurodegenerative processes is suspected. The CNS detoxifying systems include carrier-mediated, active efflux and receptor-mediated transport, and detoxifying systems that include phase I and phase II enzymes, as well as those enzymes in charge of neutralizing compounds such as electrophilic agents, reactive oxygen species (ROS), and free radicals, which are products of the bioactivation of xenobiotics. Moreover, we discuss the differential expression of these systems in different regions of the CNS, showing the different detoxifying needs and the composition of each region in terms of the cell type, neurotransmitter content, and the accumulation of xenobiotics and/or reactive compounds.


Subject(s)
Brain/drug effects , Brain/metabolism , Metabolic Networks and Pathways/drug effects , Xenobiotics/metabolism , Xenobiotics/toxicity , Biological Transport/drug effects , Biological Transport/physiology , Biotransformation/drug effects , Biotransformation/physiology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Humans , Metabolic Networks and Pathways/physiology
11.
Front Cell Neurosci ; 14: 17, 2020.
Article in English | MEDLINE | ID: mdl-32194376

ABSTRACT

Exposure to toxic metals and metalloids is an important cause of preventable diseases worldwide. Inorganic arsenic (iAs) affects several organs and tissues, causing neurobehavioral alterations in the central nervous system (CNS) that might lead to neurodegeneration. In this work, we wanted to explore the time- and dose-related changes on glutathione (GSH) levels in several regions of the CNS, such as the cortex, striatum, hippocampus, and cerebellum, to identify the initial cellular changes associated to GSH depletion due to iAs exposure. Mice received a single intraperitoneal injection containing 5 or 14 mg/kg sodium arsenite. Animals were killed at 2, 6, and 24 h. Significant depletion of GSH levels was observed in the cortex at 2 and 6 h, while on the striatum, hippocampus, or cerebellum regions, no significant changes were observed. GSH depletion in the cortex was associated with the activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) and nuclear factor kappa B (NFκB) pathways, which led to the upregulation of xCT, excitatory amino acid carrier 1 (EAAC1), glutamate/aspartate transporter (GLAST), and glial glutamate transporter 1 (GLT-1), and the activation of the transsulfuration pathways, which led to the overproduction of H2S in the cortex and increased levels of GSH in the cortex and cerebellum at 24 h. In the cortex, the N-methyl-D-aspartate (NMDA) receptor subunits NR2A and NR2B were also altered at 24 h. These early effects were not homogeneous among different brain regions and indicate early neurotoxic alterations in the cortex and cerebellum.

12.
J Biomater Appl ; 31(7): 953-984, 2017 02.
Article in English | MEDLINE | ID: mdl-28178902

ABSTRACT

The blood-brain barrier is the interface between the blood and brain, impeding the passage of most circulating cells and molecules, protecting the latter from foreign substances, and maintaining central nervous system homeostasis. However, its restrictive nature constitutes an obstacle, preventing therapeutic drugs from entering the brain. Usually, a large systemic dose is required to achieve pharmacological therapeutic levels in the brain, leading to adverse effects in the body. As a consequence, various strategies are being developed to enhance the amount and concentration of therapeutic compounds in the brain. One such tool is nanotechnology, in which nanostructures that are 1-100 nm are designed to deliver drugs to the brain. In this review, we examine many nanotechnology-based approaches to the treatment of neurodegenerative diseases. The review begins with a brief history of nanotechnology, followed by a discussion of its definition, the properties of most reported nanomaterials, their biocompatibility, the mechanisms of cell-material interactions, and the current status of nanotechnology in treating Alzheimer's, Parkinson's diseases, and amyotrophic lateral sclerosis. Of all strategies to deliver drug to the brain that are used in nanotechnology, drug release systems are the most frequently reported.


Subject(s)
Blood-Brain Barrier/chemistry , Nanocapsules/administration & dosage , Nanocapsules/chemistry , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Animals , Biocompatible Materials/administration & dosage , Biocompatible Materials/chemical synthesis , Diffusion , Drug Compounding/methods , Evidence-Based Medicine , Humans , Nanocapsules/ultrastructure , Particle Size , Tissue Distribution
13.
Front Cell Neurosci ; 9: 21, 2015.
Article in English | MEDLINE | ID: mdl-25709567

ABSTRACT

Inorganic arsenic (iAs) is an important natural pollutant. Millions of individuals worldwide drink water with high levels of iAs. Chronic exposure to iAs has been associated with lower IQ and learning disabilities as well as memory impairment. iAs is methylated in tissues such as the brain generating mono and dimethylated species. iAs methylation requires cellular glutathione (GSH), which is the main antioxidant in the central nervous system (CNS). In humans, As species cross the placenta and are found in cord blood. A CD1 mouse model was used to investigate effects of gestational iAs exposure which can lead to oxidative damage, disrupted cysteine/glutamate transport and its putative impact in learning and memory. On postnatal days (PNDs) 1, 15 and 90, the expression of membrane transporters related to GSH synthesis and glutamate transport and toxicity, such as xCT, EAAC1, GLAST and GLT1, as well as LAT1, were analyzed. Also, the expression of the glutamate receptor N-methyl-D-aspartate (NMDAR) subunits NR2A and B as well as the presence of As species in cortex and hippocampus were investigated. On PND 90, an object location task was performed to associate exposure with memory impairment. Gestational exposure to iAs affected the expression of cysteine/glutamate transporters in cortex and hippocampus and induced a negative modulation of NMDAR NR2B subunit in the hippocampus. Behavioral tasks showed significant spatial memory impairment in males while the effect was marginal in females.

14.
Oxid Med Cell Longev ; 2014: 590808, 2014.
Article in English | MEDLINE | ID: mdl-24723994

ABSTRACT

The reactive oxygen species produced continuously during oxidative metabolism are generated at very high rates in the brain. Therefore, defending against oxidative stress is an essential task within the brain. An important cellular system against oxidative stress is the thioredoxin system (TS). TS is composed of thioredoxin, thioredoxin reductase, and NADPH. This review focuses on the evidence gathered in recent investigations into the central nervous system, specifically the different brain regions in which the TS is expressed. Furthermore, we address the conditions that modulate the thioredoxin system in both, animal models and the postmortem brains of human patients associated with the most common neurodegenerative disorders, in which the thioredoxin system could play an important part.


Subject(s)
Central Nervous System/metabolism , Disease Models, Animal , Thioredoxins/metabolism , Animals , Central Nervous System/pathology , Humans , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Stress, Physiological , Thioredoxin-Disulfide Reductase
15.
BMC Complement Altern Med ; 13: 262, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-24119308

ABSTRACT

BACKGROUND: Reactive oxygen species (ROS) are important mediators in a number of degenerative diseases. Oxidative stress refers to the imbalance between the production of ROS and the ability to scavenge these species through endogenous antioxidant systems. Since antioxidants can inhibit oxidative processes, it becomes relevant to describe natural compounds with antioxidant properties which may be designed as therapies to decrease oxidative damage and stimulate endogenous cytoprotective systems. The present study tested the protective effect of two xanthones isolated from the heartwood of Calophyllum brasilienses against FeSO4-induced toxicity. METHODS: Through combinatory chemistry assays, we evaluated the superoxide (O2·â»), hydroxyl radical (OH·), hydrogen peroxide (H2O2) and peroxynitrite (ONO⁻) scavenging capacity of jacareubin (xanthone III) and 2-(3,3-dimethylallyl)-1,3,5,6-tetrahydroxyxanthone (xanthone V). The effect of these xanthones on murine DNA and bovine serum albumin degradation induced by an OH· generator system was also evaluated. Additionally, we investigated the effect of these xanthones on ROS production, lipid peroxidation and glutathione reductase (GR) activity in FeSO4-exposed brain, liver and lung rat homogenates. RESULTS: Xanthone V exhibited a better scavenging capacity for O2·â», ONOO⁻ and OH· than xanthone III, although both xanthones were unable to trap H2O2. Additionally, xanthones III and V prevented the albumin and DNA degradation induced by the OH· generator system. Lipid peroxidation and ROS production evoked by FeSO4 were decreased by both xanthones in all tissues tested. Xanthones III and V also prevented the GR activity depletion induced by pro-oxidant activity only in the brain. CONCLUSIONS: Altogether, the collected evidence suggests that xanthones can play a role as potential agents to attenuate the oxidative damage produced by different pro-oxidants.


Subject(s)
Antioxidants/pharmacology , Calophyllum/chemistry , Ferrous Compounds/toxicity , Oxidative Stress/drug effects , Xanthones/pharmacology , Animals , Brain Chemistry/drug effects , DNA Damage/drug effects , Glutathione Reductase/metabolism , Kidney/chemistry , Kidney/drug effects , Lipid Peroxidation/drug effects , Liver/chemistry , Liver/drug effects , Male , Oxidation-Reduction/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
16.
Neurotoxicol Teratol ; 33(2): 303-12, 2011.
Article in English | MEDLINE | ID: mdl-20933078

ABSTRACT

The neuroactive metabolite at the kynunerine pathway, kynurenic acid (KYNA), is a well-known competitive antagonist at the co-agonist glycine site of the N-methyl-D-aspartate receptor (NMDAr), and also decreases the extracellular levels of glutamate by blocking α7-nicotinic acetylcholine receptor (α7-nAchr) located on glutamatergic terminals. KYNA has been often reported to be neuroprotective in different neurotoxic models. The systemic administration of L-kynurenine (L-KYN)--the precursor of KYNA--together with probenecid (PROB)--an inhibitor of organic acids transport--to rodents increases KYNA levels in the brain in a dose-dependent manner. The striatal infusion of the toxin 6-hydroxydopamine (6-OHDA) to rodents is one of the common models used to simulate Parkinson's disease (PD). Different studies have linked PD alterations with excessive glutamatergic transmission in the striatum since NMDAr antagonists exert beneficial effects in PD models. In this work we investigated the effect that a systemic administration of L-KYN+PROB exerted on the toxic model induced by 6-OHDA in rats. PROB (50 mg/kg, i.p.) + L-KYN (75 mg/kg, i.p.) were given to rats for seven consecutive days. On day two of treatment, the animals were infused with a single injection of 6-OHDA (20 µg/2 µl) into the right striatum. Fourteen days post-lesion, rotation behavior was assessed as a marker of motor impairment. The total levels of dopamine (DA) were also estimated in striatal tissue samples of 6-OHDA-treated animals as a neurochemical marker of damage. In addition, twenty eight days post-lesion, the striatal damage was assessed by hematoxylin/eosin staining and immunohistochemistry against glial fibrillary acidic protein (GFAP) in the same animals. Neurodegeneration was also assessed by Fluoro Jade staining. 6-OHDA infusion increased rotation behavior, striatal reactive gliosis and neurodegeneration, while DA levels were decreased. For all markers evaluated, we observed protective effects of L-KYN+PROB on the dopaminergic damage induced by 6-OHDA. Our results suggest that this strategy was useful to mitigate dopaminergic toxicity in the hemiparkinsonian model. The combined use of L-KYN and PROB is a valuable tool to modulate glutamatergic and cholinergic activities, presumably by means of increased levels of endogenous KYNA.


Subject(s)
Corpus Striatum/drug effects , Kynurenic Acid/metabolism , Kynurenine/therapeutic use , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/prevention & control , Oxidopamine/toxicity , Probenecid/therapeutic use , Animals , Behavior, Animal/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Therapy, Combination , Kynurenine/administration & dosage , Male , Neuroprotective Agents/administration & dosage , Neurotoxicity Syndromes/metabolism , Probenecid/administration & dosage , Rats , Rats, Wistar
17.
Neurotoxicology ; 31(6): 715-23, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20643160

ABSTRACT

The early effects of 6-OHDA as a Parkinsonian model in rodents are relevant since pharmacological and toxicological points of view, as they can explain the acute and chronic deleterious events occurring in the striatum. In this study, we focused our attention on the neurochemical and motor dysfunction produced after a pulse infusion of 6-OHDA, paying special attention to the capacity of this molecule to induce neurotransmitter release and behavioural alterations. Extracellular levels of dopamine, serotonin, norepinephrine, glutamate, glutamine, aspartate, glycine and GABA were all assessed in striatal dialysates in freely moving rats immediately after exposed to a single pulse of 6-OHDA in dorsal striatum, and major behavioural markers of motor alterations were simultaneously explored. Enhanced release of dopamine, serotonin and norepinephrine was found immediately after 6-OHDA pulse. Delayed glutamate and glycine release were detected and a biphasic effect on GABA was observed. Mostly serotonin and dopamine outflow, followed by glutamate, correlated with wet dog shakes and other behavioural qualitative alterations. Early dopamine release, accompanied by other neurotransmitters, can generate an excitatory environment affecting the striatal neurons with immediate consequences for behavioural performance. In turn, these changes might be accounting for later features of toxicity described in this model.


Subject(s)
Corpus Striatum/drug effects , Corpus Striatum/metabolism , Extracellular Space/drug effects , Extracellular Space/metabolism , Microdialysis/methods , Neurotransmitter Agents/metabolism , Oxidopamine/toxicity , Animals , Male , Rats , Rats, Wistar , Time Factors
18.
Neurochem Int ; 56(6-7): 834-42, 2010.
Article in English | MEDLINE | ID: mdl-20332007

ABSTRACT

Metabolic alterations in the nervous system can be produced at early stages of toxicity and are linked with oxidative stress, energy depletion and death signaling. Proteases activation is responsible for triggering deadly cascades during cell damage in toxic models. In this study we evaluated the early time-course of toxic events (oxidative damage to lipids, mitochondrial dysfunction and LDH leakage, all at 1, 3 and 6h) in rat striatal slices exposed to quinolinic acid (QUIN, 100 microM) as an excitotoxic/pro-oxidant model, 3-nitropropionic acid (3-NP, 1mM) as an inhibitor of mitochondrial succinate dehydrogenase, and a combined model produced by the co-administration of these two toxins at subtoxic concentrations (21 and 166 microM for QUIN and 3-NP, respectively). In order to further characterize a possible causality of caspases or calpains on the toxic mechanisms produced in these models, the broad calpain inhibitor IC1 (50 microM), and the pan-caspase inhibitor Z-VAD (100 microM) were tested. Lipid peroxidation (LP) was increased at all times and in all models evaluated. Both IC1 and Z-VAD exerted significant protection against LP in all models and at all times evaluated. Mitochondrial dysfunction (MD) was consistently affected by all toxic models at 3 and 6h, but was mostly affected by 3-NP and QUIN at 1h. IC1 differentially protected the slices against 3-NP and QUIN at 1h and against QUIN at 3h, while Z-VAD exhibited positive actions against QUIN and 3-NP at all times tested, and against their combination at 3 and 6h. LDH leakage was enhanced at 1 and 3h in all toxic models, but this effect was evident only for 3-NP + QUIN and 3-NP at 6h. IC1 protected against LDH leakage at 1h in 3-NP + QUIN and 3-NP models, at 3h in all toxic models, and at 6h in 3-NP + QUIN and 3-NP models. In turn, Z-VAD protected at 1 and 6h in all models tested, and at 3h in the combined and QUIN models. Our results suggest differential chronologic and mechanistic patterns, depending on the toxic insult. Although LP, MD and membrane cell rupture are shared by the three models, the occurrence of each event seems to obey to a selective recruitment of damaging signals, including a differential activation of proteases in time. Proteases activation is likely to be an up-stream event influencing oxidative stress and mitochondrial dysfunction in these toxic models.


Subject(s)
Corpus Striatum/drug effects , Nitro Compounds/toxicity , Oxidants/toxicity , Propionates/toxicity , Protease Inhibitors/pharmacology , Quinolinic Acid/toxicity , Animals , Calpain/antagonists & inhibitors , Calpain/metabolism , Caspase Inhibitors , Caspases/metabolism , Cell Death/drug effects , Corpus Striatum/ultrastructure , Enzyme Activation/drug effects , Enzyme Inhibitors , In Vitro Techniques , Kinetics , L-Lactate Dehydrogenase/metabolism , Leupeptins/pharmacology , Lipid Peroxidation/drug effects , Male , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/physiology , Models, Animal , Oligopeptides/pharmacology , Rats , Rats, Wistar , Succinate Dehydrogenase/antagonists & inhibitors
19.
J Neurochem ; 105(3): 677-89, 2008 May.
Article in English | MEDLINE | ID: mdl-18194214

ABSTRACT

Excitotoxicity and disrupted energy metabolism are major events leading to nerve cell death in neurodegenerative disorders. These cooperative pathways share one common aspect: triggering of oxidative stress by free radical formation. In this work, we evaluated the effects of the antioxidant and energy precursor, levocarnitine (L-CAR), on the oxidative damage and the behavioral, morphological, and neurochemical alterations produced in nerve tissue by the excitotoxin and free radical precursor, quinolinic acid (2,3-pyrindin dicarboxylic acid; QUIN), and the mitochondrial toxin, 3-nitropropionic acid (3-NP). Oxidative damage was assessed by the estimation of reactive oxygen species formation, lipid peroxidation, and mitochondrial dysfunction in synaptosomal fractions. Behavioral, morphological, and neurochemical alterations were evaluated as markers of neurotoxicity in animals systemically administered with L-CAR, chronically injected with 3-NP and/or intrastriatally infused with QUIN. At micromolar concentrations, L-CAR reduced the three markers of oxidative stress stimulated by both toxins alone or in combination. L-CAR also prevented the rotation behavior evoked by QUIN and the hypokinetic pattern induced by 3-NP in rats. Morphological alterations produced by both toxins (increased striatal glial fibrillary acidic protein-immunoreactivity for QUIN and enhanced neuronal damage in different brain regions for 3-NP) were reduced by L-CAR. In addition, L-CAR prevented the synergistic action of 3-NP and QUIN to increase motor asymmetry and depleted striatal GABA levels. Our results suggest that the protective properties of L-CAR in the neurotoxic models tested are mostly mediated by its characteristics as an antioxidant agent.


Subject(s)
Brain/metabolism , Carnitine/pharmacology , Energy Metabolism/drug effects , Neurodegenerative Diseases/drug therapy , Neurotoxins/antagonists & inhibitors , Oxidative Stress/drug effects , Animals , Antioxidants/pharmacology , Brain/drug effects , Convulsants/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Energy Metabolism/physiology , Free Radicals/metabolism , Gliosis/drug therapy , Gliosis/physiopathology , Gliosis/prevention & control , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Movement Disorders/drug therapy , Movement Disorders/physiopathology , Movement Disorders/prevention & control , Nerve Degeneration/drug therapy , Nerve Degeneration/physiopathology , Nerve Degeneration/prevention & control , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Neuroprotective Agents/pharmacology , Neurotoxins/metabolism , Nitro Compounds/toxicity , Oxidative Stress/physiology , Propionates/toxicity , Quinolinic Acid/metabolism , Quinolinic Acid/toxicity , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Treatment Outcome
20.
Neurosci Res ; 56(1): 39-44, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16806549

ABSTRACT

We investigated the effects of S-allylcysteine (SAC) on early behavioral alterations, striatal changes in superoxide dismutase (SOD) activity, lipid peroxidation (LP) and mitochondrial dysfunction induced by the systemic infusion of 3-nitropropionic acid (3-NPA) to rats. SAC (300 mg/kg, i.p.), given to animals 30 min before 3-NPA (30 mg/kg, i.p.), prevented the hyperkinetic pattern evoked by the toxin. In addition, 3-NPA alone produced decreased activities of manganese- (Mn-SOD) and copper/zinc-dependent superoxide dismutase (Cu,Zn-SOD), increased LP (evaluated as the formation of lipid fluorescent products) and produced mitochondrial dysfunction in the striatum (measured as decreased 3-(3,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide reduction). In contrast, pretreatment of 3-NPA-injected rats with SAC resulted in a significant prevention of all these markers. Our findings suggest that the protective actions of SAC are related with its antioxidant properties, which in turn may be accounting for the preservation of SOD activity and primary mitochondrial tasks.


Subject(s)
Akathisia, Drug-Induced , Biomarkers/metabolism , Convulsants/pharmacology , Cysteine/analogs & derivatives , Mitochondria/metabolism , Nitro Compounds/pharmacology , Oxidative Stress , Propionates/pharmacology , Animals , Antineoplastic Agents/metabolism , Behavior, Animal/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Cysteine/metabolism , Isoenzymes/metabolism , Lipid Peroxidation , Male , Psychomotor Agitation , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...