Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nutr Neurosci ; : 1-11, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38193451

ABSTRACT

BACKGROUND: Calyxes of Hibiscus sabdariffa (Hs) contain anthocyanins, that normalize blood glucose levels (BGL) in diabetic patients. Diabetes also causes memory alterations, which could hypothetically decrease with the consumption of Hs. OBJECTIVES: To investigate the effect of dietary supplementation with a Hs extract on working memory and BGL in rats. METHODS: Diabetic hyperglycemia (DHG) was induced with streptozotocin (STZ, 55 mg/kg i.p.) in Wistar rats. After 72 h DHG was confirmed, and the consumption of Hs extract began (50 mg/Kg/day). BGL and body weight (BW) were measured at 10, 20 and 30 days after DHG induction in controls and treated animals. Learning and short-term memory were evaluated after 30 days with Novel Object Recognition Test (NOR) and Barnes Maze (BM). The gross hippocampal structure was histologically analyzed. RESULTS: STZ-treated animals presented low BW and persistent DHG (BGL <300 mg/dL). Diabetic animals consuming the Hs extract had a dual response: some showed BGL comparable to controls, while others had levels comparable to diabetic animals not consuming extract. Diabetic animals that consumed the Hs extract had a better performance in NOR and BM than the diabetic animals not consuming the extract. At the histological level, hippocampal morphological differences were observed between diabetic animals that consumed the extract and those that did not. DISCUSSION: The Hs extract used here could be a good co-adjuvant in the treatment of DHG, aimed at mitigating memory deficits and high BGL. These beneficial effects could be attributed to the anthocyanin content in the extract.

2.
J Biochem Mol Toxicol ; 37(5): e23315, 2023 May.
Article in English | MEDLINE | ID: mdl-36732937

ABSTRACT

Vascular endothelial growth factor (VEGF) exerts neuroprotective or proinflammatory effects, depending on what VEGF forms (A-E), receptor types (VEGFR1-3), and intracellular signaling pathways are involved. Neonatal monosodium glutamate (MSG) treatment triggers neuronal death by excitotoxicity, which is commonly involved in different neurological disorders, including neurodegenerative diseases. This study was designed to evaluate the effects of VEGFR-2 inhibition on neuronal damage triggered by excitotoxicity in the cerebral motor cortex (CMC) and hippocampus (Hp) after neonatal MSG treatment. MSG was administered at a dose of 4 g/kg of body weight (b.w.) subcutaneously on postnatal days (PD) 1, 3, 5, and 7, whereas the VEGFR-2 inhibitor SU5416 was administered at a dose of 10 mg/kg b.w. subcutaneously on PD 5 and 7, 30 min before the MSG treatment. Neuronal damage was assessed using hematoxylin and eosin staining, fluoro-Jade staining, and TUNEL assay. Additionally, western blot assays for some proteins of the VEGF-A/VEGFR-2 signaling pathway (VEGF-A, VEGFR-2, PI3K, Akt, and iNOS) were carried out. All assays were performed on PD 6, 8, 10, and 14. Inhibition of VEGFR-2 signaling by SU5416 increases the neuronal damage induced by neonatal MSG treatment in both the CMC and Hp. Moreover, neonatal MSG treatment increased the expression levels of the studied VEGF-A/VEGFR-2 signaling pathway proteins, particularly in the CMC. We conclude that VEGF-A/VEGFR-2 signaling pathway activation could be part of the neuroprotective mechanisms that attempt to compensate for neuronal damage induced by neonatal MSG treatment and possibly also in other conditions involving excitotoxicity.


Subject(s)
Hippocampus , Motor Cortex , Vascular Endothelial Growth Factor Receptor-2 , Hippocampus/drug effects , Motor Cortex/drug effects , Sodium Glutamate/toxicity , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals
3.
Front Biosci (Landmark Ed) ; 27(5): 146, 2022 05 06.
Article in English | MEDLINE | ID: mdl-35638413

ABSTRACT

The increases in population ageing and growth are leading to a boosting in the number of people living with dementia, Alzheimer's disease (AD) being the most common cause. In spite of decades of intensive research, no cure for AD has been found yet. However, some treatments that may change disease progression and help control symptoms have been proposed. Beyond the classical hypotheses of AD etiopathogenesis, i.e., amyloid beta peptide (Aß) accumulation and tau hyperphosphorylation, a trend in attributing a key role to other molecular mechanisms is prompting the study of different therapeutic targets. Hence, drugs designed to modulate inflammation, insulin resistance, synapses, neurogenesis, cardiovascular factors and dysbiosis are shaping a new horizon in AD treatment. Within this frame, an increase in the number of candidate drugs for disease modification treatments is expected, as well as a focus on potential combinatory multidrug strategies.The present review summarizes the latest advances in drugs targeting Aß and tau as major contributors to AD pathophysiology. In addition, it introduces the most important drugs in clinical studies targeting alternative mechanisms thought to be involved in AD's neurodegenerative process.


Subject(s)
Alzheimer Disease , Alzheimer Disease/drug therapy , Amyloid beta-Peptides , Disease Progression , Humans
4.
Microvasc Res ; 132: 104059, 2020 11.
Article in English | MEDLINE | ID: mdl-32798551

ABSTRACT

The blood-brain barrier (BBB) maintains the optimal microenvironment for brain function. Tight junctions (TJs) allow endothelial cells to adhere to each other, leading to the formation of a barrier that prevents the penetration of most molecules via transcellular routes. Evidence has indicated that seizure-induced vascular endothelial growth factor (VEGF) type 2 receptor (VEGFR-2) pathway activation weakens TJs, inducing vasodilatation and increasing vascular permeability and subsequent brain injury. The present study focused on investigating the expression levels of VEGF-related (VEGF-A and VEGFR-2) and TJ-related proteins (claudin-5, occludin and ZO-1) in the neocortical microvasculature of patients with drug-resistant temporal lobe epilepsy (TLE). The results obtained from hippocampal sclerosis TLE (HS-TLE) patients were compared with those obtained from patients with TLE secondary to lesions (lesion-TLE) and autopsy samples. The Western blotting and immunofluorescence results showed that VEGF-A and VEGFR-2 protein expression levels were increased in HS-TLE and lesion-TLE patients compared to autopsy group. On the other hand, claudin-5 expression was higher in HS-TLE patients and lesion-TLE patients than autopsies. The expression level of occludin and ZO-1 was decreased in HS-TLE patients. Our study described modifications to the integrity of the BBB that may contribute to the pathogenesis of TLE, in which the VEGF system may play an important role. We demonstrated that the same modifications were present in both HS-TLE and lesion-TLE patients, which suggests that seizures modify these systems and that they are not associated with the establishment of epilepsy.


Subject(s)
Blood-Brain Barrier/metabolism , Drug Resistant Epilepsy/metabolism , Epilepsy, Temporal Lobe/metabolism , Microvessels/metabolism , Neocortex/blood supply , Tight Junction Proteins/metabolism , Tight Junctions/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adolescent , Adult , Blood-Brain Barrier/pathology , Claudin-5/metabolism , Drug Resistant Epilepsy/drug therapy , Drug Resistant Epilepsy/pathology , Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/pathology , Female , Humans , Male , Microvessels/pathology , Middle Aged , Occludin/metabolism , Signal Transduction , Tight Junctions/pathology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Young Adult , Zonula Occludens-1 Protein/metabolism
5.
Neurosci Lett ; 735: 135237, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32645399

ABSTRACT

Glutamate-mediated excitatory synaptic signalling is primarily controlled by excitatory amino acid transporters (EAATs), such as EAAT1 and EAAT2, which are located mostly on astrocytes and, together, uptake more than 95 % of extracellular glutamate. Alterations in the functional expression levels of EAATs can lead to excessive extracellular glutamate accumulation, potentially triggering excitotoxicity and seizures, among other neurological disorders. Excitotoxicity induced in early developmental stages can lead to lasting changes in several neurotransmission systems, including the glutamatergic system, which could make the brain more susceptible to a second insult. In this study, the expression levels of EAAT1 (GLAST) and EAAT2 (GLT-1) proteins were assessed in the cerebral motor cortex (CMC), striatum, hippocampus and entorhinal cortex (EC) of male adult rats following the neonatal excitotoxic process triggered by monosodium glutamate (MSG)-treatment (4 g/kg of body weight at postnatal days 1,3,5 and 7, subcutaneously). Western blot analysis showed that neonatal MSG-treatment decreased EAAT1 expression levels in the CMC, striatum and hippocampus, while EAAT2 levels were increased in the striatum and EC and decreased in the CMC. Immunofluorescence staining confirmed the changes in EAAT1 and EAAT2 expression induced by neonatal MSG-treatment, which were accompanied by an increase in the glial fibrillary acidic protein (GFAP) immunofluorescence signalthat was particularly significant in the hippocampus. Our results show that a neonatal excitotoxic processes can induce lasting changes in the expression levels of EAAT1 and EAAT2 proteins and suggest that although astrogliosis occurs, glutamate uptake could be deficient, particularly in the CMC and hippocampus.


Subject(s)
Brain/growth & development , Brain/metabolism , Excitatory Amino Acid Transporter 1/biosynthesis , Excitatory Amino Acid Transporter 2/biosynthesis , Sodium Glutamate/toxicity , Age Factors , Animals , Animals, Newborn , Brain/drug effects , Excitatory Amino Acid Transporter 1/antagonists & inhibitors , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/genetics , Gene Expression , Glutamic Acid/toxicity , Male , Rats , Rats, Wistar
7.
Int J Dev Neurosci ; 80(4): 335-346, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32198947

ABSTRACT

Early responses to a neurological excitotoxic process include blood-brain barrier (BBB) impairment and overexpression of vascular endothelial growth factor (VEGF), but the long-term effects of excitotoxicity on the BBB properties remain unknown. To assess this, we induced an excitotoxic process on male rats by neonatal monosodium glutamate (MSG) treatment. At postnatal day (PD) 60, we measured the expression level of structural proteins of the BBB and the VEGF type-2 receptor (VEGFR-2) protein in the cerebral motor cortex (CMC), striatum (STR), hippocampus (Hp), entorhinal cortex (Ent), and hypothalamus (Hyp). We also measured BBB permeability in the same cerebral regions. Neonatal MSG treatment significantly reduced the protein expression level of claudin-5 in the CMC, and of ZO-1 in the CMC and Hp, and increased the expression level of plasmalemmal vesicle-associated protein in the CMC, and of VEGFR-2 in all regions except for the Hyp. BBB permeability was significantly higher in all studied regions of MSG-treated animals after hypertonic shock (HS). The increased BBB permeability observed in the MSG-treated animals after HS was reversed by VEGFR-2 inhibition with SU5416. We conclude that neonatal excitotoxicity leads to lasting impairment on BBB properties in adulthood, increasing its susceptibility to HS that could be regulated by VEGFR-2 activity inhibition.


Subject(s)
Blood-Brain Barrier/drug effects , Sodium Glutamate/toxicity , Animals , Animals, Newborn , Brain/drug effects , Brain Chemistry/drug effects , Indoles/pharmacology , Male , Osmotic Pressure/drug effects , Pyrroles/pharmacology , Rats , Rats, Wistar , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/genetics
8.
J Mol Neurosci ; 70(6): 916-929, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32144725

ABSTRACT

In the epilepsy spectrum, temporal lobe epilepsy (TLE) is the most common and devastating focal and symptomatic epilepsy form in adults, where more than 30% of patients develop pharmacoresistance. It is not fully understood how the gene expression contributes to establishing an epileptic phenotype. Cerebrovascular remodeling directed by VEGF (vascular endothelial growth factor) signaling might modulate the synaptic neurotransmission in the epileptic brain. To address this question, the gene expression was profiled in biopsies of the temporal cortex from diagnosed patients with pharmacoresistant TLE that underwent surgical resection to seizure control. One hundred sixty-eight genes related to VEGF signaling and GABA and glutamate neurotransmissions were evaluated. Genes related to downstream signaling -phosphoinositide 3-kinase (PI3K), mitogen-activated protein kinases (MAPK), and Janus-activated kinase/signal transducer and activator of transcription (JAK/STAT) pathways- and neurotransmitters metabolism were evaluated too. Thirty-nine genes were upregulated. The genes encoding for G protein q polypeptide, serine racemase, gephyrin, and glutamate/cystine antiporter system xCT appeared as novel upregulated genes in the pharmacoresistant TLE. ClueGO, a Cytoscape plugin, was used to build a gene network associated using Gene Ontology (GO) terminology. Enrichment analysis by ClueGO retrieves that positive regulation of endothelial cell proliferation, nerve development, and neuronal apoptosis were over-represented categories. In conclusion, VEGF signaling is confirmed as a relevant mediator in the pharmacoresistant TLE. In addition, the enrichment analysis applied to differentially expressed genes suggests new pharmacological targets to be assessed in the treatment of pharmacoresistant TLE. Results make up an approximation to better understand the epileptic brain and complement the available data.


Subject(s)
Drug Resistant Epilepsy/metabolism , Epilepsy, Temporal Lobe/metabolism , Glutamic Acid/metabolism , Neocortex/metabolism , Receptors, GABA/metabolism , Transcriptome , Vascular Endothelial Growth Factor A/metabolism , Adolescent , Adult , Drug Resistant Epilepsy/genetics , Epilepsy, Temporal Lobe/genetics , Female , Humans , MAP Kinase Signaling System , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Middle Aged , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Racemases and Epimerases/genetics , Racemases and Epimerases/metabolism , Receptors, GABA/genetics , Vascular Endothelial Growth Factor A/genetics
9.
Curr Pharm Des ; 26(12): 1263-1276, 2020.
Article in English | MEDLINE | ID: mdl-31942853

ABSTRACT

BACKGROUND: Erythropoietin (Epo) and vascular endothelial growth factor (VEGF) are two vasoactive molecules with essential trophic effects for brain development. The expression and secretion of both molecules increase in response to neuronal damage and they exert protective and restorative effects, which may also be accompanied by adverse side effects. OBJECTIVE: We review the most relevant evidence on the neuroprotective and neurorestorative effects of Epo and VEGF in three of the most frequent neurological disorders, namely, stroke, epilepsy and Alzheimer's disease, to develop new therapeutic approaches. METHODS: Several original scientific manuscripts and reviews that have discussed the evidence in critical way, considering both the beneficial and adverse effects of Epo and VEGF in the selected neurological disorders, were analysed. In addition, throughout this review, we propose several considerations to take into account in the design of therapeutic approaches based on Epo and VEGF signalling. RESULTS: Although the three selected disorders are triggered by different mechanisms, they evolve through similar processes: excitotoxicity, oxidative stress, neuroinflammation, neuronal death, glial reactivity and vascular remodelling. Epo and VEGF exert neuroprotective and neurorestorative effects by acting on these processes due to their pleiotropism. In general, the evidence shows that both Epo and VEGF reduce neuronal death but that at the vascular level, their effects are contradictory. CONCLUSION: Because the Epo and VEGF signalling pathways are connected in several ways, we conclude that more experimental studies, primarily studies designed to thoroughly assess the functional interactions between Epo and VEGF in the brain under both physiological and pathophysiological conditions, are needed.


Subject(s)
Erythropoietin , Neuroprotective Agents , Neurons/physiology , Neuroprotection , Neuroprotective Agents/pharmacology , Vascular Endothelial Growth Factor A/metabolism
10.
J Neuroimmunol ; 328: 68-72, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30597392

ABSTRACT

The vascular endothelial growth factor (VEGF) system has been shown to play a crucial role in several neuropathological processes. Temporal lobe epilepsy (TLE) is the most common focal epilepsy type in adult humans. We assessed the protein expression levels of VEGF-A, VEGF-B, and VEGF-C, their specific receptors VEGFR-2 and -3, their accessory receptors neuropilins 1 and 2, and PI3 and Akt kinases, in temporal neocortex from pharmacoresistant TLE (PR-TLE) patients and control subjects by western blotting. All proteins were found to be significantly overexpressed in samples of PR-TLE patients, indicating that the VEGF system contributes to PR-TLE pathogenesis and should be further studied.


Subject(s)
Drug Resistant Epilepsy/metabolism , Epilepsy, Temporal Lobe/metabolism , Neocortex/metabolism , Receptors, Vascular Endothelial Growth Factor/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult
11.
Pharmaceuticals (Basel) ; 11(1)2018 Feb 07.
Article in English | MEDLINE | ID: mdl-29414852

ABSTRACT

Undoubtedly, one of the most interesting topics in the field of neuroscience is the ability of the central nervous system to respond to different stimuli (normal or pathological) by modifying its structure and function, either transiently or permanently, by generating neural cells and new connections in a process known as neuroplasticity. According to the large amount of evidence reported in the literature, many stimuli, such as environmental pressures, changes in the internal dynamic steady state of the organism and even injuries or illnesses (e.g., epilepsy) may induce neuroplasticity. Epilepsy and neuroplasticity seem to be closely related, as the two processes could positively affect one another. Thus, in this review, we analysed some neuroplastic changes triggered in the hippocampus in response to seizure-induced neuronal damage and how these changes could lead to the establishment of temporal lobe epilepsy, the most common type of focal human epilepsy.

12.
J Mol Neurosci ; 63(1): 17-27, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28755050

ABSTRACT

Vascular endothelial growth factor (VEGF) exerts both neuroprotective and proinflammatory effects in the brain, depending on the VEGF (A-E) and VEGF receptor (VEGFR1-3) types involved. Neonatal monosodium glutamate (MSG) treatment triggers an excitotoxic degenerative process associated with several neuropathological conditions, and VEGF messenger RNA (mRNA) expression is increased at postnatal day (PD) 14 in rat hippocampus (Hp) following the treatment. The aim of this work was to establish the changes in immunoreactivity to VEGF-A, VEGF-B, VEGFR-1 and VEGFR-2 proteins induced by neonatal MSG treatment (4 g/kg, subcutaneous, at PD1, 3, 5 and 7) in the cerebral motor cortex (CMC) and Hp. Samples collected from PD2 to PD60 from control and MSG-treated male Wistar rats were assessed by western blotting for each protein. Considering that immunoreactivity measured by western blotting is related to the protein expression level, we found that each protein in each cerebral region has a specific expression profile throughout the studied ages, and all profiles were differentially modified by MSG. Specifically, neonatal MSG treatment significantly increased the immunoreactivity to the following: (1) VEGF-A at PD8-PD10 in the CMC and at PD6-PD8 in the Hp; (2) VEGF-B at PD2, PD6 and PD10 in the CMC and at PD8-PD9 in the Hp; and (3) VEGFR-2 at PD6-PD8 in the CMC and at PD21-PD60 in the Hp. Also, MSG significantly reduced the immunoreactivity to the following: (1) VEGF-B at PD8-PD9 and PD45-PD60 in the CMC; and (2) VEGFR-1 at PD4-PD6 and PD14-PD21 in the CMC and at PD4, PD9-PD10 and PD60 in the Hp. Our results indicate that VEGF-mediated signalling is involved in the excitotoxic process triggered by neonatal MSG treatment and should be further characterized.


Subject(s)
Glutamic Acid/toxicity , Hippocampus/drug effects , Motor Cortex/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Hippocampus/growth & development , Hippocampus/metabolism , Male , Motor Cortex/growth & development , Motor Cortex/metabolism , Rats , Rats, Wistar , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor B/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
13.
J Biomed Sci ; 24(1): 27, 2017 May 09.
Article in English | MEDLINE | ID: mdl-28486943

ABSTRACT

BACKGROUND: Neonatal monosodium glutamate (MSG) treatment triggers excitotoxicity and induces a degenerative process that affects several brain regions in a way that could lead to epileptogenesis. Na+/Ca2+ exchangers (NCX1-3) are implicated in Ca2+ brain homeostasis; normally, they extrude Ca2+ to control cell inflammation, but after damage and in epilepsy, they introduce Ca2+ by acting in the reverse mode, amplifying the damage. Changes in NCX3 expression in the hippocampus have been reported immediately after neonatal MSG treatment. In this study, the expression level of NCX1-3 in the entorhinal cortex (EC) and hippocampus (Hp); and the effects of blockade of NCXs on the seizures induced by 4-Aminopyridine (4-AP) were analysed in adult rats after neonatal MSG treatment. KB-R7943 was applied as NCXs blocker, but is more selective to NCX3 in reverse mode. METHODS: Neonatal MSG treatment was applied to newborn male rats at postnatal days (PD) 1, 3, 5, and 7 (4 g/kg of body weight, s.c.). Western blot analysis was performed on total protein extracts from the EC and Hp to estimate the expression level of NCX1-3 proteins in relative way to the expression of ß-actin, as constitutive protein. Electrographic activity of the EC and Hp were acquired before and after intracerebroventricular (i.c.v.) infusion of 4-AP (3 nmol) and KB-R7943 (62.5 pmol), alone or in combination. All experiments were performed at PD60. Behavioural alterations were also recorder. RESULTS: Neonatal MSG treatment significantly increased the expression of NCX3 protein in both studied regions, and NCX1 protein only in the EC. The 4-AP-induced epileptiform activity was significantly higher in MSG-treated rats than in controls, and KB-R7943 co-administered with 4-AP reduced the epileptiform activity in more prominent way in MSG-treated rats than in controls. CONCLUSIONS: The long-term effects of neonatal MSG treatment include increases on functional expression of NCXs (mainly of NCX3) in the EC and Hp, which seems to contribute to improve the control that KB-R7943 exerted on the seizures induced by 4-AP in adulthood. The results obtained here suggest that the blockade of NCXs could improve seizure control after an excitotoxic process; however, this must be better studied.


Subject(s)
4-Aminopyridine/toxicity , Anticonvulsants/pharmacology , Sodium Glutamate/toxicity , Thiourea/analogs & derivatives , Animals , Entorhinal Cortex/drug effects , Entorhinal Cortex/metabolism , Entorhinal Cortex/physiopathology , Gene Expression/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Homeodomain Proteins/metabolism , Infusions, Intraventricular , Male , Rats , Rats, Wistar , Thiourea/pharmacology
14.
Arch Med Res ; 45(8): 653-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25431840

ABSTRACT

It is likely that monosodium glutamate (MSG) is the excitotoxin that has been most commonly employed to characterize the process of excitotoxicity and to improve understanding of the ways that this process is related to several pathological conditions of the central nervous system. Excitotoxicity triggered by neonatal MSG treatment produces a significant pathophysiological impact on adulthood, which could be due to modifications in the blood-brain barrier (BBB) permeability and vice versa. This mini-review analyzes this topic through brief descriptions about excitotoxicity, BBB structure and function, role of the BBB in the regulation of Glu extracellular levels, conditions that promote breakdown of the BBB, and modifications induced by neonatal MSG treatment that could alter the behavior of the BBB. In conclusion, additional studies to better characterize the effects of neonatal MSG treatment on excitatory amino acids transporters, ionic exchangers, and efflux transporters, as well as the role of the signaling pathways mediated by erythropoietin and vascular endothelial growth factor in the cellular elements of the BBB, should be performed to identify the mechanisms underlying the increase in neurovascular permeability associated with excitotoxicity observed in several diseases and studied using neonatal MSG treatment.


Subject(s)
Blood-Brain Barrier/drug effects , Neurotoxins/toxicity , Sodium Glutamate/toxicity , Blood-Brain Barrier/physiopathology , Capillary Permeability/drug effects , Erythropoietin/metabolism , Glutamate Plasma Membrane Transport Proteins/physiology , Humans , Infant, Newborn , Neurotoxins/metabolism , Neurotoxins/therapeutic use , Signal Transduction/drug effects , Sodium Glutamate/metabolism , Sodium Glutamate/therapeutic use , Vascular Endothelial Growth Factor A/metabolism
15.
Neurosci Lett ; 552: 52-7, 2013 Sep 27.
Article in English | MEDLINE | ID: mdl-23932891

ABSTRACT

Seizure susceptibility appears to be greater in males than females during the early developmental stages of the brain when the gamma-aminobutyric acid (GABA), acting through its GABA-A receptor, predominantly produces neuronal depolarization. GABA-mediated excitation has been observed when the NKCC1 (chloride importer) expression level is higher than KCC2 (chloride exporter). In this study, the relative protein expression of NKCC1 and KCC2 over ß-actin was evaluated in the hippocampus and entorhinal cortex of male and female rats during postnatal days (PND) 1, 3, 5, 7, 9, 11, 13 and 15 using Western blotting assays. For both cerebral regions in the females, the NKCC1/ß-actin expression ratio was constant during all evaluated ages, whereas the KCC2/ß-actin expression ratio increased gradually until reaching a maximal level at PND9 that was nearly three- and ten-fold higher in the hippocampus and entorhinal cortex, respectively, compared with the initial level. In males, the NKCC1/ß-actin expression ratio was constant during the first week, peaking almost three-fold higher than the initial level at PND9 in the hippocampus and at PND11 in the entorhinal cortex and then returning to the initial values at PND13, whereas the KCC2/ß-actin expression ratio increased gradually to reach a maximal and steady level at PND5, which were nearly two- and four-fold higher in the hippocampus and entorhinal cortex, respectively, compared with the intial level. In conclusion, the NKCC1/ß-actin and KCC2/ß-actin expression ratios displayed a specific expression profile for each gender and cerebral region, which could be related with the differences in seizure susceptibility observed between genders.


Subject(s)
Entorhinal Cortex/metabolism , Hippocampus/metabolism , Sex Characteristics , Solute Carrier Family 12, Member 2/biosynthesis , Symporters/biosynthesis , Actins/biosynthesis , Animals , Animals, Newborn/metabolism , Female , Gene Expression Regulation, Developmental , Male , Rats , Time Factors , K Cl- Cotransporters
16.
Int J Dev Neurosci ; 27(8): 845-55, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19733649

ABSTRACT

Monosodium glutamate (MSG) administered to neonatal rats during the first week of life induces a neurodegenerative process, which is represented by several neurochemical alterations of surviving neurons in the brain, where signalling mediated by GABA is essential for excitation threshold maintenance. GABA-positive cells, [(3)H]-GABA uptake, expression of mRNA for GABA transporters GAT-1 and GAT-3, and expression of mRNA and protein for two main GABA synthesizing enzymes, GAD(65) and GAD(67), were measured at postnatal day 60, after MSG neonatal treatment in two critical cerebral regions, cerebral cortex and hippocampus. GABA-positive cells, [(3)H]-GABA uptake, and mRNA for GAT-1, were significantly diminished in both cerebral regions. In the cerebral cortex, MSG neonatal treatment also decreased the mRNA for GAD(67) and protein for GAD(65) without significant changes in its corresponding protein and mRNA, respectively. Moreover in the hippocampus, mRNA and protein for GAD(65) were increased, whilst GAD(67) protein was elevated without significant changes in its mRNA. Clearly these results confirm the GABA cells loss after MSG neonatal treatment in both cerebral regions. As most of the GABAergic markers measured were reduced in the cerebral cortex, this region seems to be more sensitive than hippocampus, where interesting compensatory changes over GAD(65) and GAD(67) proteins were observed. However, it is possible that others neurotransmission systems are also compensating the GABA-positive cells loss in the cerebral cortex, and that elevations in two main forms of GAD in the hippocampus are not sufficient to maintain the neural excitation threshold for this region.


Subject(s)
Animals, Newborn , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Sodium Glutamate/pharmacology , gamma-Aminobutyric Acid/metabolism , Animals , Cerebral Cortex/cytology , Female , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Hippocampus/cytology , Male , Radioligand Assay , Rats , Rats, Wistar , Signal Transduction/physiology , gamma-Aminobutyric Acid/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...