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.
Sci Rep ; 12(1): 8982, 2022 05 28.
Article in English | MEDLINE | ID: mdl-35643779

ABSTRACT

Glutamate receptors, including mGluR5, are involved in learning and memory impairments triggered by aging and neurological diseases. However, each condition involves distinct molecular mechanisms. It is still unclear whether the mGluR5 cell signaling pathways involved in normal brain aging differ from those altered due to neurodegenerative disorders. Here, we employed wild type (WT), mGluR5-/-, BACHD, which is a mouse model of Huntington's Disease (HD), and mGluR5-/-/BACHD mice, at the ages of 2, 6 and 12 months, to distinguish the mGluR5-dependent cell signaling pathways involved in aging and neurodegenerative diseases. We demonstrated that the memory impairment exhibited by mGluR5-/- mice is accompanied by massive neuronal loss and decreased dendritic spine density in the hippocampus, similarly to BACHD and BACHD/mGluR5-/- mice. Moreover, mGluR5 ablation worsens some of the HD-related alterations. We also show that mGluR5-/- and BACHD/mGluR5-/- mice have decreased levels of PSD95, BDNF, and Arc/Arg3.1, whereas BACHD mice are mostly spared. PSD95 expression was affected exclusively by mGluR5 ablation in the aging context, making it a potential target to treat age-related alterations. Taken together, we reaffirm the relevance of mGluR5 for memory and distinguish the mGluR5 cell signaling pathways involved in normal brain aging from those implicated in HD.


Subject(s)
Huntington Disease , Neurodegenerative Diseases , Animals , Brain/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Memory Disorders/genetics , Memory Disorders/metabolism , Mice , Neurodegenerative Diseases/metabolism , Neuronal Plasticity , Phenotype
2.
Neurochem Int ; 126: 218-228, 2019 06.
Article in English | MEDLINE | ID: mdl-30930274

ABSTRACT

The growing elderly population world widely prompts the need for studies regarding aged brain and its susceptibility to neurodegenerative diseases. It has been shown that aged brain exhibits several alterations, including neuroinflammation, which prone this organ to neurodegenerative processes. Metabotropic glutamate receptor 5 (mGlu5 receptor) has a role in neuronal cell loss and inflammation. Although the relevance of mGlu5 receptor in different diseases has been investigated, its involvement in normal brain aging remains unclear. In the present study, we used the mGlu5 receptor knockout (mGluR5-/-) mice, a model of Huntington's Disease (BACHD), and the double mutant mice (mGluR5-/-/BACHD), at the ages of 2, 6 and 12 months, to investigate whether mGlu5 receptor has a role in brain aging. We demonstrated that mGluR5-/- mice exhibit diminished number of neurons at 12 months of age in the cortex and striatum, similarly to what was observed in the case of BACHD and mGluR5-/-/BACHD mice. In addition, ablation of mGlu5 receptor increased the number of astrocytes and microglia in BACHD and wild type (WT) mice in an age-dependent manner in the cortical region, but not in the striatum. Interestingly, 12-month-old mGluR5-/- mice induced microglia activation, evidenced by increased CD68 expression and diminished number of microglia ramifications in skeleton analyses. Importantly, the presence of mutant huntingtin and the absence of mGlu5 receptor promoted decreased levels of fractalkine expression in aged mice, which could account for the decreased levels of microglia activation in these mice. Together, our data provide evidence that mGlu5 receptor plays a role in brain aging by modulating different cell types in the central nervous system (CNS).


Subject(s)
Aging/metabolism , Brain/metabolism , Inflammation Mediators/metabolism , Neurodegenerative Diseases/metabolism , Receptor, Metabotropic Glutamate 5/deficiency , Aging/genetics , Aging/pathology , Animals , Brain/pathology , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, 129 Strain , Mice, Knockout , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Receptor, Metabotropic Glutamate 5/genetics
3.
J Neurochem ; 147(2): 222-239, 2018 10.
Article in English | MEDLINE | ID: mdl-30028018

ABSTRACT

Huntington's Disease (HD) is an autosomal-dominant neurodegenerative disorder, characterized by involuntary body movements, cognitive impairment, and psychiatric disorder. The metabotropic glutamate receptor 5 (mGluR5) plays an important role in HD and we have recently demonstrated that mGluR5-positive allosteric modulators (PAMs) can ameliorate pathology and the phenotypic signs of a mouse model of HD. In this study, we investigated the molecular mechanisms involved in mGluR5 PAMs effect on memory. Our results demonstrate that subchronic treatment with the mGluR5 PAM VU0409551 was effective in reversing the memory deficits exhibited by BACHD mice, a mouse model for HD. Moreover, VU0409551 treatment stabilized mGluR5 at the cellular plasma membrane of BACHD mice, increasing the expression of several genes important for synaptic plasticity, including c-Fos, brain-derived neurotrophic factor, Arc/Arg3.1, syntaxin 1A, and post-synaptic density-95. In addition, VU0409551 treatment also increased dendritic spine density and maturation and augmented the number of pre-synaptic sites. In conclusion, our results demonstrate that VU0409551 triggered the activation of cell signaling pathways important for synaptic plasticity, enhancing the level of dendritic spine maturation and rescuing BACHD memory impairment. OPEN PRACTICES: Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/.


Subject(s)
Huntington Disease/drug therapy , Huntington Disease/psychology , Memory Disorders/drug therapy , Memory Disorders/psychology , Neuronal Plasticity/drug effects , Oxazoles/pharmacology , Pyridines/pharmacology , Receptor, Metabotropic Glutamate 5/drug effects , Synapses/drug effects , Animals , Conditioning, Classical/drug effects , Dendritic Spines/drug effects , Gene Expression Regulation/drug effects , Huntington Disease/complications , Memory Disorders/etiology , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neuronal Plasticity/genetics , Receptor, Metabotropic Glutamate 5/metabolism , Recognition, Psychology/drug effects , Signal Transduction/drug effects
4.
mBio ; 8(2)2017 04 25.
Article in English | MEDLINE | ID: mdl-28442607

ABSTRACT

Zika virus (ZIKV) infection is a global health emergency that causes significant neurodegeneration. Neurodegenerative processes may be exacerbated by N-methyl-d-aspartate receptor (NMDAR)-dependent neuronal excitoxicity. Here, we have exploited the hypothesis that ZIKV-induced neurodegeneration can be rescued by blocking NMDA overstimulation with memantine. Our results show that ZIKV actively replicates in primary neurons and that virus replication is directly associated with massive neuronal cell death. Interestingly, treatment with memantine or other NMDAR blockers, including dizocilpine (MK-801), agmatine sulfate, or ifenprodil, prevents neuronal death without interfering with the ability of ZIKV to replicate in these cells. Moreover, in vivo experiments demonstrate that therapeutic memantine treatment prevents the increase of intraocular pressure (IOP) induced by infection and massively reduces neurodegeneration and microgliosis in the brain of infected mice. Our results indicate that the blockade of NMDARs by memantine provides potent neuroprotective effects against ZIKV-induced neuronal damage, suggesting it could be a viable treatment for patients at risk for ZIKV infection-induced neurodegeneration.IMPORTANCE Zika virus (ZIKV) infection is a global health emergency associated with serious neurological complications, including microcephaly and Guillain-Barré syndrome. Infection of experimental animals with ZIKV causes significant neuronal damage and microgliosis. Treatment with drugs that block NMDARs prevented neuronal damage both in vitro and in vivo These results suggest that overactivation of NMDARs contributes significantly to the neuronal damage induced by ZIKV infection, and this is amenable to inhibition by drug treatment.


Subject(s)
Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/pathology , Neuroprotective Agents/administration & dosage , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Zika Virus Infection/complications , Zika Virus Infection/pathology , Zika Virus/growth & development , Animals , Disease Models, Animal , Mice , Treatment Outcome
5.
Oncotarget ; 8(3): 3768-3769, 2017 Jan 17.
Article in English | MEDLINE | ID: mdl-28030853
6.
Mol Brain ; 9(1): 80, 2016 08 20.
Article in English | MEDLINE | ID: mdl-27543109

ABSTRACT

The metabotropic glutamate receptor 5 (mGluR5) and the cannabinoid receptor 1 (CB1) exhibit a functional interaction, as CB1 regulates pre-synaptic glutamate release and mGluR5 activation increases endocannabinoid synthesis at the post-synaptic site. Since both mGluR5 and CB1 promote neuroprotection, we delineated experiments to investigate a possible link between CB1 and mGluR5 activation in the induction of neuroprotection using primary cultured corticostriatal neurons. We find that either the pharmacological blockade or the genetic ablation of either mGluR5 or CB1 can abrogate both CB1- and mGluR5-mediated neuroprotection against glutamate insult. Interestingly, decreased glutamate release and diminished intracellular Ca(2+) do not appear to play a role in CB1 and mGluR5-mediated neuroprotection. Rather, these two receptors work cooperatively to trigger the activation of cell signaling pathways to promote neuronal survival, which involves MEK/ERK1/2 and PI3K/AKT activation. Interestingly, although mGluR5 activation protects postsynaptic terminals and CB1 the presynaptic site, intact signaling of both receptors is required to effectively promote neuronal survival. In conclusion, mGluR5 and CB1 act in concert to activate neuroprotective cell signaling pathways and promote neuronal survival.


Subject(s)
Neuroprotection , Receptor, Cannabinoid, CB1/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Animals , Benzamides/pharmacology , Benzodioxoles/pharmacology , Calcium/metabolism , Carbamates/pharmacology , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , Intracellular Space/metabolism , Mice, Inbred C57BL , Models, Biological , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , Piperidines/pharmacology , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Synapses/drug effects , Synapses/metabolism
7.
Toxicol Appl Pharmacol ; 286(3): 178-87, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25933444

ABSTRACT

Cocaine is an addictive substance with a potential to cause deleterious effects in the brain. The strategies for treating its neurotoxicity, however, are limited. Evidence suggests that the endocannabinoid system exerts neuroprotective functions against various stimuli. Thus, we hypothesized that inhibition of fatty acid amide hydrolase (FAAH), the main enzyme responsible for terminating the actions of the endocannabinoid anandamide, reduces seizures and cell death in the hippocampus in a model of cocaine intoxication. Male Swiss mice received injections of endocannabinoid-related compounds followed by the lowest dose of cocaine that induces seizures, electroencephalographic activity and cell death in the hippocampus. The molecular mechanisms were studied in primary cell culture of this structure. The FAAH inhibitor, URB597, reduced cocaine-induced seizures and epileptiform electroencephalographic activity. The cannabinoid CB1 receptor selective agonist, ACEA, mimicked these effects, whereas the antagonist, AM251, prevented them. URB597 also inhibited cocaine-induced activation and death of hippocampal neurons, both in animals and in primary cell culture. Finally, we investigated if the PI3K/Akt/ERK intracellular pathway, a cell surviving mechanism coupled to CB1 receptor, mediated these neuroprotective effects. Accordingly, URB597 injection increased ERK and Akt phosphorylation in the hippocampus. Moreover, the neuroprotective effect of this compound was reversed by the PI3K inhibitor, LY294002. In conclusion, the pharmacological facilitation of the anandamide/CB1/PI3K signaling protects the brain against cocaine intoxication in experimental models. This strategy may be further explored in the development of treatments for drug-induced neurotoxicity.


Subject(s)
Cocaine/toxicity , Endocannabinoids/metabolism , Receptor, Cannabinoid, CB1/metabolism , Signal Transduction/drug effects , Animals , Benzamides/pharmacology , Carbamates/pharmacology , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Endocannabinoids/agonists , Hippocampus/drug effects , Hippocampus/pathology , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Receptor, Cannabinoid, CB1/agonists , Signal Transduction/physiology
8.
Expert Opin Ther Targets ; 18(11): 1293-304, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25118797

ABSTRACT

INTRODUCTION: Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a polyglutamine expansion in the amino-terminal region of the huntingtin (htt) protein, which underlies the loss of striatal and cortical neurons. Glutamate has been implicated in a number of neurodegenerative diseases, and several studies suggest that the metabotropic glutamate receptor 5 (mGluR5) may represent a target for the treatment of HD. AREAS COVERED: The main goal of this review is to discuss the current data in the literature regarding the role of mGluR5 in HD and evaluate the potential of mGluR5 as a therapeutic target for the treatment of HD. mGluR5 is highly expressed in the brain regions affected in HD and is involved in movement control. Moreover, mGluR5 interacts with htt and mutated htt profoundly affects mGluR5 signaling. However, mGluR5 stimulation can activate both neuroprotective and neurotoxic signaling pathways, depending on the context of activation. EXPERT OPINION: Although the data published so far strongly indicate that mGluR5 plays a major role in HD-associated neurodegeneration, htt aggregation and motor symptoms, it is not clear whether mGluR5 stimulation can diminish or intensify neuronal cell loss and HD progression. Thus, future experiments will be necessary to further investigate the outcome of drugs acting on mGluR5 for the treatment of neurodegenerative diseases.


Subject(s)
Huntington Disease/drug therapy , Molecular Targeted Therapy , Receptor, Metabotropic Glutamate 5/drug effects , Animals , Brain/physiopathology , Drug Design , Glutamic Acid/metabolism , Humans , Huntingtin Protein , Huntington Disease/physiopathology , Nerve Tissue Proteins/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL