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.
Exp Biol Med (Maywood) ; 248(7): 588-595, 2023 04.
Article in English | MEDLINE | ID: mdl-37158084

ABSTRACT

Major depressive disorder (MDD) is a complex illness that is arising as a growing public health concern. Although several brain areas are related to this type of disorders, at the cellular level, the parvalbumin-positive cells of the hippocampus interplay a very relevant role. They control pyramidal cell bursts, neuronal networks, basic microcircuit functions, and other complex neuronal tasks involved in mood disorders. In resistant depressions, the efficacy of current antidepressant treatments drops dramatically, so the new rapid-acting antidepressants (RAADs) are being postulated as novel treatments. Ketamine at subanesthetic doses and its derivative metabolites have been proposed as RAADs due to their rapid and sustained action by blocking N-methyl-d-aspartate (NMDA) receptors, which in turn lead to the release of brain-derived neurotrophic factor (BDNF). This mechanism produces a rapid plasticity activation mediated by neurotransmitter homeostasis, synapse recovery, and increased dendritic spines and therefore, it is a promising therapeutic approach to improve cognitive symptoms in MDD.


Subject(s)
Depressive Disorder, Major , Ketamine , Humans , Ketamine/pharmacology , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/metabolism , Parvalbumins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/therapeutic use , Antidepressive Agents/pharmacology , Antidepressive Agents/metabolism , Antidepressive Agents/therapeutic use , Interneurons/metabolism , Hippocampus/metabolism , Brain-Derived Neurotrophic Factor/metabolism
2.
Mol Neurobiol ; 55(5): 4511, 2018 May.
Article in English | MEDLINE | ID: mdl-29504058

ABSTRACT

The authors found a terrible mistake in the manuscript. The legends from the Fig. 5 and 6 are interchanged. The Fig. 5 should be appeared with the legend from the Fig. 6 and Fig. 6 should be appeared with the legend from the Fig. 5.

3.
Mol Neurobiol ; 55(1): 201-212, 2018 01.
Article in English | MEDLINE | ID: mdl-28840516

ABSTRACT

Neurotrophic factors (NTFs) are a promising therapeutic option for Parkinson's disease (PD). They exert their function through tyrosine kinase receptors. Our goal was to assess the effects of administering a selective tyrosine kinase inhibitor (vandetanib) that blocks VEGFR2 and RET receptors in a preclinical model of PD. Rats underwent intrastriatal injections of 6-hydroxydopamine (6-OHDA). Two weeks later, the rats received 30 mg/kg vandetanib or saline orally. The effects were assessed using the rotational behavioral test, tyrosine hydroxylase (TH) immunohistochemistry, and western blot. In 6-OHDA-lesioned rats, motor symptoms were almost undetectable, but morphological and biochemical changes were significant. Vandetanib treatment, combined with the presence of 6-OHDA lesions, significantly increased behavioral impairment and morphological and biochemical changes. Therefore, after vandetanib treatment, the TH-immunopositive striatal volume, the percentage of TH+ neurons, and the extent of the axodendritic network in the substantia nigra decreased. Glial fibrillary acidic protein-positivity significantly decreased in the striatum and substantia nigra in the vandetanib-treated group. In addition, p-Akt and p-ERK 1/2 levels were significantly lower and caspase-3 expression significantly increased after vandetanib administration. In conclusion, we demonstrate for the first time the deleterious effect of a tyrosine kinase inhibitor on the dopaminergic system, supporting the beneficial and synergistic effect of NTFs reported in previous papers.


Subject(s)
Parkinsonian Disorders/metabolism , Piperidines/toxicity , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Proto-Oncogene Proteins c-ret/metabolism , Quinazolines/toxicity , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Male , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/pathology , Rats , Rats, Sprague-Dawley , Treatment Outcome
4.
Mol Neurobiol ; 54(10): 7722-7735, 2017 Dec.
Article in English | MEDLINE | ID: mdl-27844282

ABSTRACT

The unilateral 6-hydroxydopamine (6-OHDA) lesion of medial forebrain bundle (MFB) in rats affords us to study the advanced stages of Parkinson's disease (PD). Numerous evidences suggest synergic effects when various neurotrophic factors are administered in experimental models of PD. The aim of the present work was to assess the morphological changes along the rostro-caudal axis of caudo-putamen complex and substantia nigra (SN) in the referred model in order to test the suitability of a severe model to evaluate new neurorestorative therapies. Administration of 6-OHDA into MFB in addition to a remarkable depletion of dopamine in the nigrostriatal system induced an increase of glial fibrillary acidic protein (GFAP)-positive cells in SN and an intense immunoreactivity for OX-42, vascular endothelial growth factor (VEGF), and Lycopersycum esculentum agglutinin (LEA) in striatum and SN. Tyrosine hydroxylase (TH) immunostaining revealed a significant decrease of the TH-immunopositive striatal volume in 6-OHDA group from rostral to caudal one. The loss of TH-immunoreactive (TH-ir) neurons and axodendritic network (ADN) was higher in caudal sections. Morphological recovery after the implantation of microspheres loaded with VEGF and glial cell line-derived neurotrophic factor (GDNF) in parkinsonized rats was related to the preservation of the TH-ir cell number and ADN in the caudal region of the SN. In addition, these findings support the neurorestorative role of VEGF+GDNF in the dopaminergic system and the synergistic effect between both factors. On the other hand, a topological distribution of the dopaminergic system was noticeable in the severe model, showing a selective vulnerability to 6-OHDA and recovering after treatment.


Subject(s)
Drug Compounding , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/pathology , Severity of Illness Index , Vascular Endothelial Growth Factor A/administration & dosage , Animals , Drug Compounding/methods , Female , Neuroprotective Agents/administration & dosage , Rats , Rats, Sprague-Dawley , Treatment Outcome
5.
Mol Neurobiol ; 52(2): 846-58, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26041662

ABSTRACT

Administration of various neurotrophic factors is a promising strategy against Parkinson's disease (PD). An intrastriatal infusion of 6-hydroxidopamine (6-OHDA) in rats is a suitable model to study PD. This work aims to describe stereological parameters regarding rostro-caudal gradient, in order to characterize the model and verify its suitability for elucidating the benefits of therapeutic strategies. Administration of 6-OHDA induced a reduction in tyrosine hidroxylase (TH) reactivity in the dorsolateral part of the striatum, being higher in the caudal section than in the rostral one. Loss of TH-positive neurons and axodendritic network was highly significant in the external third of substantia nigra (e-SN) in the 6-OHDA group versus the saline one. After the administration of nanospheres loaded with neurotrophic factors (NTF: vascular endothelial growth factor (VEGF) + glial cell line-derived neurotrophic factor (GDNF)), parkinsonized rats showed more TH-positive fibers than those of control groups; this recovery taking place chiefly in the rostral sections. Neuronal density and axodendritic network in e-SN was more significant than in the entire SN; the topographical analysis showed that the highest difference between NTF versus control group was attained in the middle section. A high number of bromodeoxyuridine (BrdU)-positive cells were found in sub- and periventricular areas in the group receiving NTF, where most of them co-expressed doublecortin. Measurements on the e-SN achieved more specific and significant results than in the entire SN. This difference in rostro-caudal gradients underpins the usefulness of a topological approach to the assessment of the lesion and therapeutic strategies. Findings confirmed the neurorestorative, neurogenic, and synergistic effects of VEGF+GDNF administration.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Parkinsonian Disorders/drug therapy , Vascular Endothelial Growth Factor A/therapeutic use , Animals , Corpus Striatum/drug effects , Corpus Striatum/enzymology , DNA Replication , Doublecortin Domain Proteins , Doublecortin Protein , Drug Synergism , Drug Therapy, Combination , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/pharmacokinetics , Glial Fibrillary Acidic Protein/analysis , Injections, Intralesional , Male , Microtubule-Associated Proteins/analysis , Nanocapsules , Nerve Tissue Proteins/analysis , Neuroglia/chemistry , Neuroglia/drug effects , Neuroglia/ultrastructure , Neurons/chemistry , Neurons/drug effects , Neurons/ultrastructure , Neuropeptides/analysis , Oxidopamine/toxicity , Parkinsonian Disorders/pathology , Rats , Rats, Sprague-Dawley , Tissue Distribution , Tyrosine 3-Monooxygenase/analysis , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/pharmacokinetics
6.
Behav Brain Res ; 246: 36-46, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23470903

ABSTRACT

The role of VEGF in the nervous system is extensive; apart from its angiogenic effect, VEGF has been described as a neuroprotective, neurotrophic and neurogenic molecule. Similar effects have been described for enriched environment (EE). Moreover, both VEGF and EE have been related to improved spatial memory. Our aim was to investigate the neurovascular and cognitive effects of intracerebrally-administered VEGF and enriched environment during the critical period of the rat visual cortex development. Results showed that VEGF infusion as well as enriched environment induced neurovascular and cognitive effects in developing rats. VEGF administration produced an enhancement during the learning process of enriched animals and acted as an angiogenic factor both in primary visual cortex (V1) and dentate gyrus (DG) in order to counteract minipump implantation-induced damage. This fact revealed that DG vascularization is critical for normal learning. In contrast to this enriched environment acted on the neuronal density of the DG and V1 cortex, and results showed learning enhancement only in non-operated rats. In conclusion, VEGF administration only has effects if damage is observed due to injury. Once control values were reached, no further effects appeared, showing a ceiling effect. Our results strongly support that in addition to neurogenesis, vascularization plays a pivotal role for learning and memory.


Subject(s)
Brain Injuries/complications , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Environment , Vascular Endothelial Growth Factor A/therapeutic use , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Cholinesterases/metabolism , Cognition Disorders/pathology , Dentate Gyrus/growth & development , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Drug Delivery Systems , Female , Glial Fibrillary Acidic Protein/metabolism , Lectins/metabolism , Male , Maze Learning/drug effects , Rats , Rats, Long-Evans , Vascular Endothelial Growth Factor A/metabolism , Visual Cortex/growth & development , Visual Cortex/metabolism , Visual Cortex/pathology
7.
Acta Neurochir Suppl ; 106: 55-9, 2010.
Article in English | MEDLINE | ID: mdl-19812921

ABSTRACT

We investigated the effects of exogenous Vascular Endothelial Growth Factor VEGF combined with an enriched environment on BBB integrity after a minimal trauma induced during the first days of the critical visual period in rats, when peak levels of endogenous VEGF secretion are reached. VEGF was administered using osmotic mini-pumps placed in middle cortical layers of P18 Long-Evansrats. Tissue changes were evaluated using conventional histology. BBB integrity was shown by immunohistochemistry techniques for EBA and GluT-1. Mini-pump implantation produced a wider cavity in anti-VEGF infused rats. In VEGF-infused rats there was a damaged region around the cannula that was smaller in rats raised in an enriched environment (EE). The administration of VEGF induced a high concentration of plasma proteins in the neuropil around the point of cannula placement and a high inflammatory reaction. VEGF-infused rats raised in an EE showed a lower degree of extravasation and better tissue preservation. Anti-VEGF administration produced a lower protein expression profile and more widespread deterioration of tissue. Double immunofluorescence for EBA and GluT-1 showed that the administration of VEGF preserves the tissue, which remains present but not fully functional. In contrast, a combination of VEGF administration and an EE partially protects the functionally damaged tissue with a higher preservation of BBB integrity.


Subject(s)
Antibodies/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/growth & development , Brain/growth & development , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/pharmacology , Animals , Animals, Newborn , Autoantigens/metabolism , Brain/anatomy & histology , Environment , Excitatory Amino Acid Transporter 2/metabolism , Gene Expression Regulation/drug effects , Rats , Rats, Long-Evans
8.
Acta Neurochir Suppl ; 106: 277-82, 2010.
Article in English | MEDLINE | ID: mdl-19812964

ABSTRACT

Brain edema in gliomas is an epiphenomenon related to blood-brain-barrier (BBB) breakdown in which endothelial nitric oxide synthase (eNOS) plays a key role. When induced by vascular endothelial growth factor (VEGF), eNOS synthesizes nitric oxide that increases vascular permeability. We investigated the relationship between eNOS, VEGF and BBB dysfunction in experimental gliomas.Tumors were produced in Sprague-Dawley rats by transplacentary administration of Ethylnitrosourea (ENU). Immunoexpression of eNOS and VEGF(165) was studied to identify locations of vascular permeability. BBB permeability was evaluated using gadolinium and intravital dyes and BBB integrity by endothelial barrier antigen (EBA), glucose transporter-1 (GluT-1) and occludin immunostaining. Low grade gliomas displayed constitutive eNOS expression in endothelial cells and in VEGF-positive astrocytes surrounding vessels. Malignant gliomas overexpressed eNOS in aberrant vessels and displayed numerous adjacent reactive astrocytes positive for VEGF. Huge dilated vessels inside tumors and glomeruloid vessels on the periphery of the tumor showed strong immunopositivity for eNOS and a lack of occludin and EBA staining in several vascular sections. BBB dysfunction on these aberrant vessels caused increased permeability as shown by Gadolinium contrast enhancement and intravital dye extravasation.These findings support the central role of eNOS in intra- and peritumoral edema in ENU-induced gliomas.


Subject(s)
Brain Neoplasms , Capillary Permeability/drug effects , Ethylnitrosourea , Glioma , Nitric Oxide Synthase Type III/metabolism , Animals , Autoantigens/metabolism , Brain Neoplasms/chemically induced , Brain Neoplasms/enzymology , Brain Neoplasms/physiopathology , Capillary Permeability/physiology , Disease Models, Animal , Gadolinium , Glioma/chemically induced , Glioma/enzymology , Glioma/physiopathology , Glucose Transporter Type 1/metabolism , Pentetic Acid , Plant Lectins , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...