Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Exp Neurol ; 365: 114427, 2023 07.
Article in English | MEDLINE | ID: mdl-37116638

ABSTRACT

The retinotectal topography of rats develops within the first three postnatal weeks during the critical period. Previous studies have shown that monocular enucleation results in plasticity of the intact retinotectal pathway in a time-dependent manner. Glial fibrillary acidic protein (GFAP), an astrocyte marker, is up-regulated after central nervous system injury. Adenosine is a neuromodulator involved in the development and plasticity of the visual system acting through the inhibitory A1 and excitatory A2a receptor activities. Herein, we examined whether adenosine receptors and astrocytes are crucial for monocular enucleation (ME)-induced plasticity. We also investigate whether A2a blockade alters retinotectal plasticity in an astrocyte-dependent manner. Lister Hooded rats were submitted to monocular enucleation at postnatal day 10 (PND10) or PND21 and, after different survival times, were processed for immunohistochemistry or western blotting assays. Another group underwent subpial implantation of ELVAX containing vehicle (DMSO) or SCH58261 (1 µM - an A2a receptor antagonist), simultaneously with ME at PND10. After a 72 h survival, GFAP content and the retinotectal plasticity were evaluated. Our data show that monocular enucleation leads to an upregulation in GFAP expression in the contralateral superior colliculus. At PND10, a slight increase in GFAP labeling was observed at 72 h post-enucleation, while at PND21 GFAP increase was detected in the deafferented superior colliculus after 1 to 3 weeks of survival. The content of adenosine receptors also varies in the contralateral target after ME. A transient increase in A1 receptors is observed in the early periods of plasticity, while A2a receptors are upregulated later. Interestingly, the local blockade of A2a receptors abolished the increase in GFAP and the retinotectal reorganization induced by monocular enucleation during the critical period. Taken together these results suggest a correlation between astrocytes and A2a adenosine receptors in the subcortical visual plasticity.


Subject(s)
Astrocytes , Superior Colliculi , Animals , Rats , Astrocytes/metabolism , Eye Enucleation , Superior Colliculi/metabolism , Receptors, Purinergic P1/metabolism , Immunohistochemistry , Receptor, Adenosine A2A/metabolism
3.
Glia ; 69(6): 1429-1443, 2021 06.
Article in English | MEDLINE | ID: mdl-33497496

ABSTRACT

Central nervous system (CNS) function depends on precise synaptogenesis, which is shaped by environmental cues and cellular interactions. Astrocytes are outstanding regulators of synapse development and plasticity through contact-dependent signals and through the release of pro- and antisynaptogenic factors. Conversely, myelin and its associated proteins, including Nogo-A, affect synapses in a inhibitory fashion and contribute to neural circuitry stabilization. However, the roles of Nogo-A-astrocyte interactions and their implications in synapse development and plasticity have not been characterized. Therefore, we aimed to investigate whether Nogo-A affects the capacity of astrocytes to induce synaptogenesis. Additionally, we assessed whether downregulation of Nogo-A signaling in an in vivo demyelination model impacts the synaptogenic potential of astrocytes. Our in vitro data show that cortical astrocytes respond to Nogo-A through RhoA pathway activation, exhibiting stress fiber formation and decreased ramified morphology. This phenotype was associated with reduced levels of GLAST protein and aspartate uptake, decreased mRNA levels of the synaptogenesis-associated genes Hevin, glypican-4, TGF-ß1 and BDNF, and decreased and increased protein levels of Hevin and SPARC, respectively. Corroborating these findings, conditioned medium from Nogo-A-treated astrocytes suppressed the formation of structurally and functionally mature synapses in cortical neuronal cultures. After cuprizone-induced acute demyelination, we observed reduced immunostaining for Nogo-A in the visual cortex accompanied by higher levels of Hevin expression in astrocytes and an increase in excitatory synapse density. Hence, we suggest that interactions between Nogo-A and astrocytes might represent an important pathway of plasticity regulation and could be a target for therapeutic intervention in demyelinating diseases in the future.


Subject(s)
Astrocytes , Demyelinating Diseases , Humans , Neurogenesis , Nogo Proteins , Synapses
4.
Neuroimmunomodulation ; 25(2): 96-102, 2018.
Article in English | MEDLINE | ID: mdl-30110698

ABSTRACT

OBJECTIVE: Interleukin 4 (IL-4) is an anti-inflammatory cytokine related to different aspects of central nervous system development such as survival, proliferation, and differentiation, among others. Our goals were to investigate the effect of intravitreous treatment with IL-4 on the activation of downstream signaling pathways in the retina and the distribution of retinal axons within the superior colliculus (SC). MATERIAL AND METHODS: Lister hooded rats were submitted to an intravitreous injection of either IL-4 (5 U/µL) or PBS (vehicle) at postnatal day 10 (PND10). At PND11 or PND14, retinas were processed for Western blot or immunohistochemistry. At PND13, a group of animals received an intraocular injection of an anterograde tracer in the left (untreated) eye in order to label the uncrossed retinotectal axons. RESULTS: Our data revealed that intravitreous treatment with IL-4 at PND10 leads to a decrease in GFAP content and a sustained increase in the phosphorylation of STAT6 and ERK levels in the retina. IL-4 also increases retinal axonal arbors within the SC, compared to control groups. CONCLUSIONS: These data suggest that a single in vivo treatment with IL-4 during the early stages of development modulates signaling pathways in the retina, resulting in altered binocular subcortical visual connectivity.


Subject(s)
Interleukin-4/administration & dosage , MAP Kinase Signaling System/physiology , Nerve Net/metabolism , Retina/metabolism , STAT6 Transcription Factor/metabolism , Visual Pathways/metabolism , Animals , Intravitreal Injections , MAP Kinase Signaling System/drug effects , Nerve Net/drug effects , Phosphorylation/drug effects , Phosphorylation/physiology , Rats , Retina/drug effects , Rodentia , Visual Pathways/drug effects
5.
Neuroimmunomodulation ; 23(2): 81-7, 2016.
Article in English | MEDLINE | ID: mdl-27031648

ABSTRACT

OBJECTIVE: The development of retinotectal pathways form precise topographical maps is usually completed by the third postnatal week. Cytokines participate in the development and plasticity of the nervous system. We have previously shown that in vivo treatment with interleukin 2 disrupts the retinocollicular topographical order in early stages of development. Therefore, we decided to study the effect of a single intravitreous injection of IL-6 upon retinotectal circuitry in neonates and juvenile rats. MATERIALS AND METHODS: Lister Hooded rats received an intravitreous injection of IL-6 (50 ng/ml) or vehicle (PBS) at either postnatal day (PND)10 or PND30 and the ipsilateral retinotectal pathway was evaluated 4 or 8 days later, respectively. RESULTS: Our data showed that, at different stages of development, a single IL-6 intravitreous treatment did not produce an inflammatory response and increased retinal axon innervation throughout the visual layers of the superior colliculus. CONCLUSIONS: Taken together, our data provide the first evidence that a single intravitreous injection with IL-6 leads to sprouting in the subcortical visual connections and suggest that small changes in IL-6 levels might be sufficient to impair the correct neuronal circuitry fine-tuning during brain development.


Subject(s)
Interleukin-6/administration & dosage , Retina/growth & development , Superior Colliculi/growth & development , Visual Pathways/growth & development , Animals , Animals, Newborn , Intravitreal Injections , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Rats , Retina/cytology , Retina/drug effects , Superior Colliculi/cytology , Superior Colliculi/drug effects , Visual Pathways/cytology , Visual Pathways/drug effects
6.
Cell Metab ; 18(6): 831-43, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24315369

ABSTRACT

Alzheimer's disease (AD) and type 2 diabetes appear to share similar pathogenic mechanisms. dsRNA-dependent protein kinase (PKR) underlies peripheral insulin resistance in metabolic disorders. PKR phosphorylates eukaryotic translation initiation factor 2α (eIF2α-P), and AD brains exhibit elevated phospho-PKR and eIF2α-P levels. Whether and how PKR and eIF2α-P participate in defective brain insulin signaling and cognitive impairment in AD are unknown. We report that ß-amyloid oligomers, AD-associated toxins, activate PKR in a tumor necrosis factor α (TNF-α)-dependent manner, resulting in eIF2α-P, neuronal insulin receptor substrate (IRS-1) inhibition, synapse loss, and memory impairment. Brain phospho-PKR and eIF2α-P were elevated in AD animal models, including monkeys given intracerebroventricular oligomer infusions. Oligomers failed to trigger eIF2α-P and cognitive impairment in PKR(-/-) and TNFR1(-/-) mice. Bolstering insulin signaling rescued phospho-PKR and eIF2α-P. Results reveal pathogenic mechanisms shared by AD and diabetes and establish that proinflammatory signaling mediates oligomer-induced IRS-1 inhibition and PKR-dependent synapse and memory loss.


Subject(s)
Amyloid beta-Peptides/toxicity , Brain/drug effects , Insulin Receptor Substrate Proteins/metabolism , Polymers/toxicity , Tumor Necrosis Factor-alpha/metabolism , eIF-2 Kinase/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Animals , Brain/metabolism , Disease Models, Animal , Haplorhini/metabolism , Hypoglycemic Agents/pharmacology , Insulin Receptor Substrate Proteins/antagonists & inhibitors , Memory Disorders/metabolism , Memory Disorders/pathology , Mice , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , Polymers/chemistry , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/drug effects , Synapses/drug effects , Synapses/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , eIF-2 Kinase/deficiency , eIF-2 Kinase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...