Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Glycoconj J ; 39(5): 685-699, 2022 10.
Article in English | MEDLINE | ID: mdl-35653015

ABSTRACT

Neurodegeneration is a pathological condition that is associated with the loss of neuronal function and structure. In neurodegenerative diseases, mounting evidence indicates that neuroinflammation is a common factor that contributes to neuronal damage and neurodegeneration. Neuroinflammation is characterized by the activation of microglia, the neuroimmune cells of the central nervous system (CNS), which have been implicated as active contributors to neuronal damage. Glycan structure modification is defining the outcome of neuroinflammation and neuronal regeneration; moreover, the expression of galectins, a group of lectins that specifically recognize ß-galactosides, has been proposed as a key factor in neuronal regeneration and modulation of the inflammatory response. Of the different galectins identified, galectin-1 stimulates the secretion of neurotrophic factors in astrocytes and promotes neuronal regeneration, whereas galectin-3 induces the proliferation of microglial cells and modulates cell apoptosis. Galectin-8 emerged as a neuroprotective factor, which, in addition to its immunosuppressive function, could generate a neuroprotective environment in the brain. This review describes the role of galectins in the activation and modulation of astrocytes and microglia and their anti- and proinflammatory functions within the context of neuroinflammation. Furthermore, it discusses the potential use of galectins as a therapeutic target for the inflammatory response and remodeling in damaged tissues in the central nervous system.


Subject(s)
Neurodegenerative Diseases , Astrocytes/metabolism , Astrocytes/pathology , Galectins/metabolism , Humans , Microglia/metabolism , Microglia/pathology , Neurodegenerative Diseases/pathology , Neuroinflammatory Diseases
2.
Neuropeptides ; 74: 11-23, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30795916

ABSTRACT

Galectins are animal lectins that bind to ß-galactosides, such as lactose and N-acetyllactosamine, contained in glycoproteins or glycolipids. Galectin-1 (Gal-1) and Galectin-3 (Gal-3) are involved in pathologies associated with the inflammatory process, cell proliferation, adhesion, migration, and apoptosis. Recent evidence has shown that the administration of Amyloid-ß 25-35 (Aß25-35) into the hippocampus of rats increases the inflammatory response that is associated with memory impairment and neurodegeneration. Galectins could participate in the modulation of the neuroinflammation induced by the Aß25-35. The aim of this study was to evaluate the presence of Gal-1 and Gal-3 in the neuroinflammation induced by administration of Aß25-35 into the hippocampus and to examine spatial memory in the Morris water maze. After the administration of Aß25-35, animals were tested for learning and spatial memory in the Morris water maze. Behavioral performance showed that Aß25-35 didn't affect spatial learning but did impair memory, with animals taking longer to find the platform. On the day 32, hippocampus was examined for astrocytes (GFAP), microglia (Iba1), Gal-1 and Gal-3 via immunohistochemical analysis, and the cytokines IL-1ß, TNF-α, IFN-γ by ELISA. This study's results showed a significant increase in the expression of Gal-3 in the microglia and astrocytes, while Gal-1 didn't increase in the dorsal hippocampus. The expression of galectins is associated with increased cytokines in the hippocampal formation of Aß25-35 treated rats. These findings suggest that Gal-3 could participate in the inflammation induced by administration of Aß25-35 and could be involved in the neurodegeneration progress and memory impairment.


Subject(s)
Amyloid beta-Peptides/toxicity , Astrocytes/metabolism , Encephalitis/metabolism , Galectin 3/metabolism , Microglia/metabolism , Peptide Fragments/toxicity , Spatial Memory/physiology , Animals , Astrocytes/drug effects , Encephalitis/chemically induced , Galectin 1/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Maze Learning/drug effects , Maze Learning/physiology , Microglia/drug effects , Rats, Wistar , Spatial Memory/drug effects
3.
Neurotox Res ; 35(3): 563-574, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30645726

ABSTRACT

Levodopa-induced dyskinesia (LID) is the main side effect associated with levodopa treatment and represents the biggest challenge for Parkinson's disease therapy. While the overexpression of ΔFosB transcription factor is related to the development of LID, few studies have been undertaken on fosB gene transcriptional regulation induced by levodopa in vivo. The aim of this study is to evaluate the expression of ΔFosB mRNA and FosB mRNA in the striatum after acute, chronic, and subchronic levodopa treatment in rats with unilateral 6-OHDA-lesion in the medial forebrain bundle. qRT-PCR was used to compare the levels of ΔFosB and FosB mRNA expression in the dopamine-denervated striatum following levodopa treatment. While the results obtained after a single levodopa dose indicate a significant increase of ∆FosB mRNA expression in the striatum 1 h post-injection, the levels returned to baseline values after 24 h. After subchronic levodopa treatment, the levels of ∆FosB and FosB mRNA expression were lower 1 h post-administration of levodopa in comparison with acute effect. However, after chronic levodopa treatment, ∆FosB mRNA expression in the striatum persisted in dyskinetic rats only, and positive correlation was found between the levels of ∆FosB mRNA expression 1 h after levodopa administration and the level of dyskinetic severity. In summary, acute levodopa treatment led to highly increased levels of ∆FosB mRNA expression in the striatum. While repeated administration induced a partial desensitization of the fosB gene in the striatum, it did not suppress its activity completely, which could explain why dyskinesia appears after chronic levodopa treatment.


Subject(s)
Antiparkinson Agents/pharmacology , Corpus Striatum/drug effects , Levodopa/pharmacology , Parkinsonian Disorders/drug therapy , Proto-Oncogene Proteins c-fos/metabolism , Animals , Corpus Striatum/metabolism , Functional Laterality , Gene Expression/drug effects , Male , Parkinsonian Disorders/metabolism , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Time Factors
4.
Neuropeptides ; 67: 9-19, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29129405

ABSTRACT

Memory impairment by the Amyloid-ß 25-35 (Aß25-35) peptide in animal models has provided an understanding of the causes behind the similar deterioration that occurs in Alzheimer's disease. However, it is uncertain if a decrease of dendritic spines and neurogenesis conduces to cognitive impairment by an impairment in the retrieval of stored memory. The aim of this study was to evaluate the consequences of impairment on spatial memory caused by the administration of the Aß25-35 peptide in the hippocampus, which is associated whit morphological changes and neurogenesis in the dentate gyrus (DG). The vehicle or Aß25-35 peptide (0.1µg/µL) were bilaterally administered in the CA1 subfield of the rat hippocampus. The animals were tested for spatial learning and memory in the Morris Water Maze. In the day's 11, 18 and 32 after administration of the Aß25-35 peptide were examined the morphological changes in the DG using a Golgi-Cox stain. In the day 32, the neurogenesis was evaluated by the immunoreactivity to 5-bromo-2'-deoxyuridine (BrdU; 100mg/kg, i.p.) that corresponding to cellular proliferation post damage, the neuronal specific nuclear protein (NeuN) and doublecortin (DCX). This study found a memory retrieval impairment occurring at day 17, a cognitive deficit which had increased significantly at day 31 after the administration of Aß25-35 peptide. These results are related to morphological changes in the granular cells of the DG, such as a shorter dendritic length and a decrease in the number of dendritic spines. In neurogenesis, the total number of cells positive to BrdU, NeuN and DCX in the hippocampal granule cell layer was found to have declined in animals treated with Aß25-35. The results suggest that the Aß25-35 peptide impairs memory retrieval by decreasing the number of dendritic spines and altering neurogenesis in the DG.


Subject(s)
Amyloid beta-Peptides/pharmacology , Memory Disorders/drug therapy , Neurogenesis/drug effects , Neurons/drug effects , Peptide Fragments/pharmacology , Amyloid beta-Peptides/metabolism , Animals , Dendritic Spines/drug effects , Dentate Gyrus/drug effects , Disease Models, Animal , Doublecortin Protein , Hippocampus/drug effects , Hippocampus/metabolism , Male , Memory Disorders/chemically induced , Neurogenesis/physiology , Neurons/metabolism , Rats, Wistar , Spatial Memory
5.
Neural Plast ; 2015: 375391, 2015.
Article in English | MEDLINE | ID: mdl-26355725

ABSTRACT

Prophylactic subacute administration of zinc decreases lipoperoxidation and cell death following a transient cerebral hypoxia-ischemia, thus suggesting neuroprotective and preconditioning effects. Chemokines and growth factors are also involved in the neuroprotective effect in hypoxia-ischemia. We explored whether zinc prevents the cerebral cortex-hippocampus injury through regulation of CCL2, CCR2, FGF2, and IGF-1 expression following a 10 min of common carotid artery occlusion (CCAO). Male rats were grouped as follows: (1) Zn96h, rats injected with ZnCl2 (one dose every 24 h during four days); (2) Zn96h + CCAO, rats treated with ZnCl2 before CCAO; (3) CCAO, rats with CCAO only; (4) Sham group, rats with mock CCAO; and (5) untreated rats. The cerebral cortex-hippocampus was dissected at different times before and after CCAO. CCL2/CCR2, FGF2, and IGF-1 expression was assessed by RT-PCR and ELISA. Learning in Morris Water Maze was achieved by daily training during 5 days. Long-term memory was evaluated on day 7 after learning. Subacute administration of zinc increased expression of CCL2, CCR2, FGF2, and IGF-1 in the early and late phases of postreperfusion and prevented the CCAO-induced memory loss in the rat. These results might be explained by the induction of neural plasticity because of the expression of CCL2 and growth factors.


Subject(s)
Chlorides/therapeutic use , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/psychology , Memory Disorders/etiology , Memory Disorders/prevention & control , Zinc Compounds/therapeutic use , Animals , Carotid Stenosis/psychology , Chemokine CCL2/biosynthesis , Fibroblast Growth Factor 2/biosynthesis , Insulin-Like Growth Factor I/biosynthesis , Male , Maze Learning/drug effects , Memory, Long-Term/drug effects , Rats , Rats, Wistar , Receptors, CCR2/biosynthesis
6.
Synapse ; 69(3): 103-14, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25522178

ABSTRACT

The cannabinoid CB1 (CB1R) and dopaminergic D2 (D2R) receptors modify GABAergic transmission in the globus pallidus. Although dopaminergic denervation produces changes in the expression and supersensitization of these receptors, the consequences of these changes on GABAergic neurotransmission are unknown. The aim of this study was to show the effects of CB1R and D2R activation and coactivation on the uptake and release of [(3) H]GABA in the globus pallidus of hemiparkinsonian rats as well as their effects on motor behavior. The activation of CB1R blocked GABA uptake and decreased GABA release in the globus pallidus in the dopamine denervated side, whereas the co-activation of CB1R-D2R increased GABA release and had no effect on GABA uptake. A microinjection of the CB1R agonist ACEA into the globus pallidus ipsilaterally to a 6-OHDA lesion potentiated turning behavior that was induced by methamphetamine. However, a microinjection of the D2R agonist quinpirole did not modify this behavior, and a microinjection of a mixture of CB1R and D2R agonists significantly potentiated turning behavior. The behavioral effects produced after the activation of the CB1R and the co-activation of CB1R and D2R can be explained by increased GABAergic neurotransmission produced by a block of GABA uptake and an increase in the release of GABA in the globus pallidus, respectively.


Subject(s)
GABAergic Neurons/metabolism , Globus Pallidus/metabolism , Receptor, Cannabinoid, CB1/metabolism , Receptors, Dopamine D2/metabolism , Synaptic Transmission , Animals , Arachidonic Acids/pharmacology , GABAergic Neurons/drug effects , Globus Pallidus/drug effects , Globus Pallidus/physiology , Male , Methamphetamine/pharmacology , Movement , Oxidopamine/toxicity , Quinpirole/pharmacology , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/agonists , Receptors, Dopamine D2/agonists , gamma-Aminobutyric Acid/metabolism
7.
J Neural Transm (Vienna) ; 120(8): 1179-89, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23430275

ABSTRACT

There is evidence to support that an impaired energy metabolism and the excessive generation of reactive oxygen species (ROS) contribute to brain injury in neurodegenerative disorders such as Parkinson's disease (PD), whereas diets enriched in foods with an antioxidant action may modulate its progression. Several studies have proved that the antioxidant components produced by Spirulina, a microscopic blue-green alga, might prevent cell death by decreasing free radicals, inhibiting lipoperoxidation and upregulating the antioxidant enzyme systems. In our study, we investigated the protective effect of the Spirulina maxima (S. maxima) against the 6-OHDA-caused toxicity in the rat striatum. The S. maxima (700 mg/kg/day, vo) was administered for 40 days before and 20 days after a single injection of 6-OHDA (16 µg/2 µL) into the dorsal striatum. At 20-day postsurgery, the brain was removed and the striatum was obtained to evaluate the indicators of toxicity, such as nitric oxide levels, ROS formation, lipoperoxidation, and mitochondrial activity. These variables were found significantly stimulated in 6-OHDA-treated rats and were accompanied by declines in dopamine levels and motor activity. In contrast, the animals that received the chronic treatment with S. maxima had a restored locomotor activity, which is associated with the decreased levels of nitric oxide, ROS, and lipoperoxidation in the striatum, although mitochondrial functions and dopamine levels remained preserved. These findings suggest that supplementation with antioxidant phytochemicals (such as contained in S. maxima) represents an effective neuroprotective strategy against 6-OHDA-caused neurotoxicity vía free radical production to preserve striatal dopaminergic neurotransmission in vivo.


Subject(s)
Antioxidants/pharmacology , Corpus Striatum/drug effects , Models, Animal , Oxidopamine/toxicity , Spirulina , Animals , Corpus Striatum/microbiology , Corpus Striatum/pathology , Male , Motor Activity/drug effects , Motor Activity/physiology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Treatment Outcome
8.
Neurosci Res ; 63(2): 129-37, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19084561

ABSTRACT

beta-Amyloid plays an important role in the neurodegeneration process of Alzheimer's disease (AD), but its neurotoxic mechanisms are not clear. It has been associated with the increase of oxidative stress and cognitive impairment because the beta-amyloid peptide 25-35 (Abeta((25-35))) has the critical neurotoxic properties of the full-length Abeta(1-42). Our present study shows the role of Abeta((25-35)) when injected into the temporal cortex on the nitric oxide pathways, 3-nitrotyrosine, neuronal death, and the spatial memory of rats 1 month after the injection. Our data showed that Abeta((25-35)) increases oxidative stress, causes neuronal damage, and decreases spatial memory in rats. Notably, the injection of the fraction Abeta((25-35)) caused an increase of nNOS and iNOS immunoreactivity in the temporal cortex and hippocampus. We demonstrated a significant increase of reactive astrocytosis, which was accompanied by neuronal damage in the temporal cortex and hippocampus of rats injected with Abeta((25-35)). These data suggest that the fraction Abeta((25-35)) injected into the temporal cortex might contribute to understanding the role of nitric oxide on the biological changes related to the neuropathological progression and the memory impairment in AD.


Subject(s)
Amyloid beta-Peptides , Memory Disorders/chemically induced , Memory Disorders/pathology , Nitric Oxide/metabolism , Peptide Fragments , Temporal Lobe/enzymology , Animals , Behavior, Animal/drug effects , Glial Fibrillary Acidic Protein/metabolism , Male , Maze Learning/drug effects , Nitric Oxide Synthase Type I/metabolism , Rats , Rats, Wistar , Silver Staining/methods , Space Perception/drug effects , Temporal Lobe/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...