Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
EMBO J ; 40(3): e103701, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33319920

ABSTRACT

SATB2 is a schizophrenia risk gene and is genetically associated with human intelligence. How it affects cognition at molecular level is currently unknown. Here, we show that interactions between SATB2, a chromosomal scaffolding protein, and the inner nuclear membrane protein LEMD2 orchestrate the response of pyramidal neurons to neuronal activation. Exposure to novel environment in vivo causes changes in nuclear shape of CA1 hippocampal neurons via a SATB2-dependent mechanism. The activity-driven plasticity of the nuclear envelope requires not only SATB2, but also its protein interactor LEMD2 and the ESCRT-III/VPS4 membrane-remodeling complex. Furthermore, LEMD2 depletion in cortical neurons, similar to SATB2 ablation, affects neuronal activity-dependent regulation of multiple rapid and delayed primary response genes. In human genetic data, LEMD2-regulated genes are enriched for de novo mutations reported in intellectual disability and schizophrenia and are, like SATB2-regulated genes, enriched for common variants associated with schizophrenia and cognitive function. Hence, interactions between SATB2 and the inner nuclear membrane protein LEMD2 influence gene expression programs in pyramidal neurons that are linked to cognitive ability and psychiatric disorder etiology.


Subject(s)
Gene Regulatory Networks , Hippocampus/cytology , Intellectual Disability/genetics , Matrix Attachment Region Binding Proteins/metabolism , Membrane Proteins/metabolism , Mutation , Nuclear Proteins/metabolism , Schizophrenia/genetics , Transcription Factors/metabolism , ATPases Associated with Diverse Cellular Activities/metabolism , Animals , Cell Nucleus/metabolism , Cell Plasticity , Cells, Cultured , Cognition , Endosomal Sorting Complexes Required for Transport/metabolism , HeLa Cells , Hippocampus/metabolism , Humans , Intellectual Disability/metabolism , Male , Matrix Attachment Region Binding Proteins/chemistry , Matrix Attachment Region Binding Proteins/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Neurons/cytology , Neurons/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Schizophrenia/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Vacuolar Proton-Translocating ATPases/metabolism
2.
Int J Mol Sci ; 22(1)2020 Dec 31.
Article in English | MEDLINE | ID: mdl-33396297

ABSTRACT

Evidence suggests that PKA activity in the nucleus accumbens (NAc) plays an essential role in reward-related learning. In this study, we investigated whether PKA is differentially involved in the expression of learning produced by either natural reinforcers or psychostimulants. For that purpose, we inhibited PKA through a bilateral infusion of Rp-cAMPS, a specific PKA inhibitor, directly into the NAc. The effects of PKA inhibition in the NAc on the expression of concurrent conditioned place preference (CPP) for cocaine (drug) and social interaction (natural reward) in rats were evaluated. We found that PKA inhibition increased the expression of cocaine preference. This effect was not due to altered stress levels or decreased social reward. PKA inhibition did not affect the expression of natural reward as intra-NAc Rp-cAMPS infusion did not affect expression of social preference. When rats were trained to express cocaine or social interaction CPP and tested for eventual persisting preference 7 and 14 days after CPP expression, cocaine preference was persistent, but social preference was abolished after the first test. These results suggest that PKA in the NAc is involved in drug reward learning that might lead to addiction and that only drug, but not natural, reward is persistent.


Subject(s)
Cocaine/pharmacology , Conditioning, Operant/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Nucleus Accumbens/drug effects , Reward , Social Interaction , Animals , Central Nervous System Stimulants/pharmacology , Cyclic AMP/metabolism , Male , Nucleus Accumbens/metabolism , Rats , Rats, Sprague-Dawley
3.
PLoS Genet ; 15(2): e1007890, 2019 02.
Article in English | MEDLINE | ID: mdl-30726206

ABSTRACT

During CNS development, the nuclear protein SATB2 is expressed in superficial cortical layers and determines projection neuron identity. In the adult CNS, SATB2 is expressed in pyramidal neurons of all cortical layers and is a regulator of synaptic plasticity and long-term memory. Common variation in SATB2 locus confers risk of schizophrenia, whereas rare, de novo structural and single nucleotide variants cause severe intellectual disability and absent or limited speech. To characterize differences in SATB2 molecular function in developing vs adult neocortex, we isolated SATB2 protein interactomes at the two ontogenetic stages and identified multiple novel SATB2 interactors. SATB2 interactomes are highly enriched for proteins that stabilize de novo chromatin loops. The comparison between the neonatal and adult SATB2 protein complexes indicates a developmental shift in SATB2 molecular function, from transcriptional repression towards organization of chromosomal superstructure. Accordingly, gene sets regulated by SATB2 in the neocortex of neonatal and adult mice show limited overlap. Genes encoding SATB2 protein interactors were grouped for gene set analysis of human GWAS data. Common variants associated with human cognitive ability are enriched within the genes encoding adult but not neonatal SATB2 interactors. Our data support a shift in the function of SATB2 in cortex over lifetime and indicate that regulation of spatial chromatin architecture by the SATB2 interactome contributes to cognitive function in the general population.


Subject(s)
Cognition/physiology , Matrix Attachment Region Binding Proteins/genetics , Neocortex/physiology , Transcription Factors/genetics , Adult , Animals , Humans , Memory, Long-Term/physiology , Mice , Mice, Inbred C57BL , Neurons/physiology , Polymorphism, Single Nucleotide/genetics , Transcription, Genetic/genetics
4.
Elife ; 52016 11 29.
Article in English | MEDLINE | ID: mdl-27897969

ABSTRACT

SATB2 is a risk locus for schizophrenia and encodes a DNA-binding protein that regulates higher-order chromatin configuration. In the adult brain Satb2 is almost exclusively expressed in pyramidal neurons of two brain regions important for memory formation, the cerebral cortex and the CA1-hippocampal field. Here we show that Satb2 is required for key hippocampal functions since deletion of Satb2 from the adult mouse forebrain prevents the stabilization of synaptic long-term potentiation and markedly impairs long-term fear and object discrimination memory. At the molecular level, we find that synaptic activity and BDNF up-regulate Satb2, which itself binds to the promoters of coding and non-coding genes. Satb2 controls the hippocampal levels of a large cohort of miRNAs, many of which are implicated in synaptic plasticity and memory formation. Together, our findings demonstrate that Satb2 is critically involved in long-term plasticity processes in the adult forebrain that underlie the consolidation and stabilization of context-linked memory.


Subject(s)
Gene Expression Regulation , Hippocampus/physiology , Matrix Attachment Region Binding Proteins/metabolism , Memory, Long-Term , MicroRNAs/biosynthesis , Transcription Factors/metabolism , Animals , Gene Knockout Techniques , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Knockout , Transcription Factors/genetics
5.
Mol Biosyst ; 11(6): 1584-93, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25728364

ABSTRACT

Microglia are resident macrophages in the central nervous system, whose participation against exogenous injuries and infections is mainly marked by an immediate release of inflammatory cytokines along with a toxic efflux of superoxide radicals. Indeed, many lines of evidence indicate that persistent activation of these cells turns their neuroprotective phenotype into a neurotoxic one, which contributes to destroy neuronal activity and induces neuronal loss in several neurodegeneration processes, such as Alzheimer's disease. In this study we attempted to fill-in the gap in our knowledge about redox regulation of amyloid activated microglia. With this aim, we carried out a robust and comprehensive characterization of the reversibly redox modified proteome both at the level of resting and amyloid-activated BV2 cells, an immortalised cell line of murine microglia. The approach we used combined the selective enrichment of reversible redox modified proteins through a biotin bait with nanoscale liquid chromatography tandem mass spectrometry of their proteolytic peptides. By this reliable approach, we identified 60 proteins changing the redox status of their selective cysteine residues upon treatment with the amyloidogenic Aß25-35 peptide. These results assessed that in microglia stimulated by amyloids, redox modifications of the proteome specifically target proteins involved in crucial cell processes, i.e. those involved in the protein synthesis. In particular, for peroxiredoxin-6 (Prdx6) and Ras-related C3 botulinum toxin substrate 1 (Rac1) we suggest mechanisms through which reversible redox modifications could affect the peculiar role of microglia in amyloidogenic injury, which at the same time reinforce the oxidative burst and resist toward it. Moreover, the redox modulation we observed on chloride intracellular channel protein 1 (CLIC1) strengthens the structural and functional relationship between the oxidative stress and the metamorphic transition of this protein from a soluble form to an integral membrane form. The redox signatures we determined might also provide neurologists with more specific and reliable biomarkers to distinguish the diverse microglia status in neurodegeneration and then to drive targeted drug design.


Subject(s)
Amyloid beta-Peptides/pharmacology , Microglia/drug effects , Peptide Fragments/pharmacology , Proteome/analysis , Proteome/drug effects , Animals , Cell Line , Mice , Microglia/metabolism , Oxidation-Reduction , Proteome/chemistry , Proteomics
SELECTION OF CITATIONS
SEARCH DETAIL
...