Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 2134, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37185241

ABSTRACT

Severe psychological trauma triggers genetic, biochemical and morphological changes in amygdala neurons, which underpin the development of stress-induced behavioural abnormalities, such as high levels of anxiety. miRNAs are small, non-coding RNA fragments that orchestrate complex neuronal responses by simultaneous transcriptional/translational repression of multiple target genes. Here we show that miR-483-5p in the amygdala of male mice counterbalances the structural, functional and behavioural consequences of stress to promote a reduction in anxiety-like behaviour. Upon stress, miR-483-5p is upregulated in the synaptic compartment of amygdala neurons and directly represses three stress-associated genes: Pgap2, Gpx3 and Macf1. Upregulation of miR-483-5p leads to selective contraction of distal parts of the dendritic arbour and conversion of immature filopodia into mature, mushroom-like dendritic spines. Consistent with its role in reducing the stress response, upregulation of miR-483-5p in the basolateral amygdala produces a reduction in anxiety-like behaviour. Stress-induced neuromorphological and behavioural effects of miR-483-5p can be recapitulated by shRNA mediated suppression of Pgap2 and prevented by simultaneous overexpression of miR-483-5p-resistant Pgap2. Our results demonstrate that miR-483-5p is sufficient to confer a reduction in anxiety-like behaviour and point to miR-483-5p-mediated repression of Pgap2 as a critical cellular event offsetting the functional and behavioural consequences of psychological stress.


Subject(s)
Basolateral Nuclear Complex , MicroRNAs , Animals , Male , Mice , Amygdala/metabolism , Basolateral Nuclear Complex/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Neurons/metabolism , Synapses/metabolism
2.
Ann Med ; 53(1): 1710-1721, 2021 12.
Article in English | MEDLINE | ID: mdl-34601991

ABSTRACT

INTRODUCTION: Taking into account the possibility of myelin-associated proteins having a role in brain tumour development, the study aimed to evaluate the diagnostic usefulness of myelin-associated proteins (Nogo-A, MAG, OMgp) released into extracellular space in patients with brain tumours. PATIENTS AND METHODS: Protein concentration in primary brain tumour (n = 49) and non-tumoural subjects (n = 24) was measured in cerebrospinal fluid (CSF) and serum by means of ELISA. Immunohistochemistry for IDH1-R132H was done on 5-µm thick formalin-fixed, paraffin-embedded tumour sections with the use of an antibody specific for the mutant IDH1-R132H protein. RESULTS: The receiver operator characteristic curve analysis showed that CSF Nogo-A and serum MAG were useful in differentiating patients with primary brain tumour from non-tumoural individuals. This was also true in the case of the separate analysis of the astrocytic tumour versus non-tumoural groups and the meningeal tumour versus non-tumoural groups. Neither Nogo-A nor MAG or OMgp concentrations were significantly different, in serum or CSF, between IDH1 wild-type astrocytic brain tumour patients compared to IDH1 mutant patients. CONCLUSIONS: Our results indicated the potential usefulness of CSF Nogo-A and serum MAG evaluation as circulating biomarkers of primary brain tumours. Because blood is relatively easy to obtain, future research should be conducted to explicitly indicate the value of serum MAG concentration evaluation as a brain tumour biomarker.Key messagesMyelin-associated proteins may be circulating brain tumour biomarkers.Nogo-A and MAG proteins seem to be the most useful in brain tumour diagnosis.Decreased CSF Nogo-A concentration is an adverse prognostic factor for patients' survival.


Subject(s)
Brain Neoplasms/diagnosis , Myelin-Associated Glycoprotein/blood , Nogo Proteins/cerebrospinal fluid , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Brain Neoplasms/blood , Case-Control Studies , Female , GPI-Linked Proteins , Humans , Male , Middle Aged , Myelin Proteins/blood , Myelin Proteins/cerebrospinal fluid , Myelin Sheath , Receptors, Cell Surface
3.
Nat Commun ; 11(1): 78, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31911626

ABSTRACT

The SLC12A cation-Cl- cotransporters (CCC), including NKCC1 and the KCCs, are important determinants of brain ionic homeostasis. SPAK kinase (STK39) is the CCC master regulator, which stimulates NKCC1 ionic influx and inhibits KCC-mediated efflux via phosphorylation at conserved, shared motifs. Upregulation of SPAK-dependent CCC phosphorylation has been implicated in several neurological diseases. Using a scaffold-hybrid strategy, we develop a novel potent and selective SPAK inhibitor, 5-chloro-N-(5-chloro-4-((4-chlorophenyl)(cyano)methyl)-2-methylphenyl)-2-hydroxybenzamide ("ZT-1a"). ZT-1a inhibits NKCC1 and stimulates KCCs by decreasing their SPAK-dependent phosphorylation. Intracerebroventricular delivery of ZT-1a decreases inflammation-induced CCC phosphorylation in the choroid plexus and reduces cerebrospinal fluid (CSF) hypersecretion in a model of post-hemorrhagic hydrocephalus. Systemically administered ZT-1a reduces ischemia-induced CCC phosphorylation, attenuates cerebral edema, protects against brain damage, and improves outcomes in a model of stroke. These results suggest ZT-1a or related compounds may be effective CCC modulators with therapeutic potential for brain disorders associated with impaired ionic homeostasis.


Subject(s)
Brain/metabolism , Enzyme Inhibitors/administration & dosage , Hydrocarbons, Chlorinated/administration & dosage , Nitriles/administration & dosage , Protein Serine-Threonine Kinases/antagonists & inhibitors , Solute Carrier Family 12, Member 2/metabolism , Stroke/drug therapy , Animals , Brain/drug effects , Brain/enzymology , Humans , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Solute Carrier Family 12, Member 2/genetics , Stroke/genetics , Stroke/metabolism
5.
Front Cell Neurosci ; 12: 209, 2018.
Article in English | MEDLINE | ID: mdl-30072872

ABSTRACT

Genetically encoded calcium indicators (GECIs) have gained widespread use for measurement of neuronal activity but their low expression levels in transgenic mice tend to limit sensitivity. We have developed a transgenic mouse line (SyG37) that expresses a ratiometric calcium sensor, SyGCaMP2-mCherry, that is expressed throughout the brain but targeted to presynaptic terminals. Within the CA1 and CA3 regions of hippocampus of male and female mice, SyGaMP2 fluorescence responds linearly up to 10 electrical stimuli at frequencies up to 100 Hz and it can detect responses to a single stimulus. Responses in single boutons can be measured using multiphoton microscopy. The ensemble amplitude of SyGCaMP2 responses is a function of the number of stimuli applied and the number of contributing boutons. The peak responses and initial rates of calcium influx in single boutons in CA1 and CA3 were similar but the rate of calcium clearance from CA3 boutons after stimulation was significantly faster. In CA1, DNQX reduced SyGCaMP2 responses to Schaffer collateral stimulation to 86% of baseline indicating that 14% of the total response originated from presynaptic terminals of neurones synaptically driven via AMPA receptors. Theta burst stimulation induced long-term potentiation (LTP) of both SyGCaMP2 and fEPSP responses in both young and 18-month-old mice. The proportion of postsynaptically connected terminals increased significantly to 76% of the total after LTP induction. The SyG37 mouse allows stable optical detection of synaptic activation and connectivity at the single bouton level and can be used to characterize the contributions of presynaptic calcium to synaptic transmission and plasticity.

6.
Hum Mutat ; 37(11): 1157-1161, 2016 11.
Article in English | MEDLINE | ID: mdl-27492651

ABSTRACT

Hereditary spastic paraplegias (HSPs) are genetically and clinically heterogeneous axonopathies primarily affecting upper motor neurons and, in complex forms, additional neurons. Here, we report two families with distinct recessive mutations in TFG, previously suggested to cause HSP based on findings in a single small family with complex HSP. The first carried a homozygous c.317G>A (p.R106H) variant and presented with pure HSP. The second carried the same homozygous c.316C>T (p.R106C) variant previously reported and displayed a similarly complex phenotype including optic atrophy. Haplotyping and bisulfate sequencing revealed evidence for a c.316C>T founder allele, as well as for a c.316_317 mutation hotspot. Expression of mutant TFG proteins in cultured neurons revealed mitochondrial fragmentation, the extent of which correlated with clinical severity. Our findings confirm the causal nature of bi-allelic TFG mutations for HSP, broaden the clinical and mutational spectra, and suggest mitochondrial impairment to represent a pathomechanistic link to other neurodegenerative conditions.


Subject(s)
Mutation, Missense , Proteins/genetics , Proteins/metabolism , Spastic Paraplegia, Hereditary/pathology , Animals , Cells, Cultured , Female , Genetic Predisposition to Disease , Humans , Magnetic Resonance Imaging/methods , Male , Mice , Mitochondria/pathology , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Pedigree , Sequence Analysis, DNA , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism
7.
Prog Brain Res ; 214: 135-57, 2014.
Article in English | MEDLINE | ID: mdl-25410356

ABSTRACT

Recent studies implicate extracellular proteases in synaptic plasticity, learning, and memory. The data are especially strong for such serine proteases as thrombin, tissue plasminogen activator, neurotrypsin, and neuropsin as well as matrix metalloproteinases, MMP-9 in particular. The role of those enzymes in the aforementioned phenomena is supported by the experimental results on the expression patterns (at the gene expression and protein and enzymatic activity levels) and functional studies, including knockout mice, specific inhibitors, etc. Counterintuitively, the studies have shown that the extracellular proteolysis is not responsible mainly for an overall degradation of the extracellular matrix (ECM) and loosening perisynaptic structures, but rather allows for releasing signaling molecules from the ECM, transsynaptic proteins, and latent form of growth factors. Notably, there are also indications implying those enzymes in the major neuropsychiatric disorders, probably by contributing to synaptic aberrations underlying such diseases as schizophrenia, bipolar, autism spectrum disorders, and drug addiction.


Subject(s)
Brain/cytology , Extracellular Matrix/enzymology , Learning/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Animals , Brain Diseases/pathology , Brain Diseases/physiopathology , Humans , Peptide Hydrolases
8.
PLoS One ; 8(4): e61046, 2013.
Article in English | MEDLINE | ID: mdl-23593384

ABSTRACT

Behavioural adaptation to psychological stress is dependent on neuronal plasticity and dysfunction at this cellular level may underlie the pathogenesis of affective disorders such as depression and post-traumatic stress disorder. Taking advantage of genome-wide microarray assay, we performed detailed studies of stress-affected transcripts in the amygdala - an area which forms part of the innate fear circuit in mammals. Having previously demonstrated the role of lipocalin-2 (Lcn-2) in promoting stress-induced changes in dendritic spine morphology/function and neuronal excitability in the mouse hippocampus, we show here that the Lcn-2 gene is one of the most highly upregulated transcripts detected by microarray analysis in the amygdala after acute restraint-induced psychological stress. This is associated with increased Lcn-2 protein synthesis, which is found on immunohistochemistry to be predominantly localised to neurons. Stress-naïve Lcn-2(-/-) mice show a higher spine density in the basolateral amygdala and a 2-fold higher rate of neuronal firing rate compared to wild-type mice. Unlike their wild-type counterparts, Lcn-2(-/-) mice did not show an increase in dendritic spine density in response to stress but did show a distinct pattern of spine morphology. Thus, amygdala-specific neuronal responses to Lcn-2 may represent a mechanism for behavioural adaptation to psychological stress.


Subject(s)
Amygdala/cytology , Amygdala/metabolism , Dendritic Spines , Lipocalins/metabolism , Neurons/metabolism , Stress, Psychological , Action Potentials/genetics , Alternative Splicing , Animals , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation , Lipocalins/genetics , Male , Mice , Mice, Knockout , Stress, Psychological/genetics , Transcription, Genetic
9.
Article in English | MEDLINE | ID: mdl-23015780

ABSTRACT

CORTICAL GABAERGIC INTERNEURONS IN RODENTS ORIGINATE IN THREE SUBCORTICAL REGIONS: the medial ganglionic eminence (MGE), the lateral/caudal ganglionic eminence (LGE/CGE), and the preoptic area (POA). Each of these neuroepithelial precursor domains contributes different interneuron subtypes to the cortex. Neuronal NOS (nNOS)-expressing neurons represent a heterogenous population of cortical interneurons. We examined the development of these cells in the mouse embryonic cortex and their abundance and distribution in adult animals. Using genetic lineage tracing in transgenic mice we find that nNOS type I cells originate only in the MGE whereas type II cells have a triple origin in the MGE, LGE/CGE, and POA. The two populations are born at different times during development, occupy different layers in the adult cortex and have distinct neurochemical profiles. nNOS neurons are more numerous in the adult cortex than previously reported and constitute a significant proportion of the cortical interneuron population. Our data suggest that the heterogeneity of nNOS neurons in the cortex can be attributed to their multiple embryonic origins which likely impose distinct genetic specification programs.

10.
Proc Natl Acad Sci U S A ; 108(45): 18436-41, 2011 Nov 08.
Article in English | MEDLINE | ID: mdl-21969573

ABSTRACT

Psychological stress causes adaptive changes in the nervous system directed toward maintaining homoeostasis. These biochemical and structural mechanisms regulate animal behavior, and their malfunction may result in various forms of affective disorders. Here we found that the lipocalin-2 (Lcn2) gene, encoding a secreted protein of unknown neuronal function, was up-regulated in mouse hippocampus following psychological stress. Addition of lipocalin-2 to cultured hippocampal neurons reduced dendritic spine actin's mobility, caused retraction of mushroom spines, and inhibited spine maturation. These effects were further enhanced by inactivating iron-binding residues of Lcn-2, suggesting that they were facilitated by the iron-free form of Lcn-2. Concurrently, disruption of the Lcn2 gene in mice promoted stress-induced increase in spine density and caused an increase in the proportion of mushroom spines. The above changes correlated with higher excitability of CA1 principal neurons and with elevated stress-induced anxiety in Lcn-2(-/-) mice. Our study demonstrates that lipocalin-2 promotes stress-induced changes in spine morphology and function to regulate neuronal excitability and anxiety.


Subject(s)
Acute-Phase Proteins/physiology , Anxiety/physiopathology , Dendritic Spines/physiology , Lipocalins/physiology , Neurons/physiology , Oncogene Proteins/physiology , Acute-Phase Proteins/genetics , Animals , Base Sequence , Blotting, Western , DNA Primers , Immunohistochemistry , Lipocalin-2 , Lipocalins/genetics , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Oncogene Proteins/genetics , Real-Time Polymerase Chain Reaction
11.
Nature ; 473(7347): 372-5, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21508957

ABSTRACT

A minority of individuals experiencing traumatic events develop anxiety disorders. The reason for the lack of correspondence between the prevalence of exposure to psychological trauma and the development of anxiety is unknown. Extracellular proteolysis contributes to fear-associated responses by facilitating neuronal plasticity at the neuron-matrix interface. Here we show in mice that the serine protease neuropsin is critical for stress-related plasticity in the amygdala by regulating the dynamics of the EphB2-NMDA-receptor interaction, the expression of Fkbp5 and anxiety-like behaviour. Stress results in neuropsin-dependent cleavage of EphB2 in the amygdala causing dissociation of EphB2 from the NR1 subunit of the NMDA receptor and promoting membrane turnover of EphB2 receptors. Dynamic EphB2-NR1 interaction enhances NMDA receptor current, induces Fkbp5 gene expression and enhances behavioural signatures of anxiety. On stress, neuropsin-deficient mice do not show EphB2 cleavage and its dissociation from NR1 resulting in a static EphB2-NR1 interaction, attenuated induction of the Fkbp5 gene and low anxiety. The behavioural response to stress can be restored by intra-amygdala injection of neuropsin into neuropsin-deficient mice and disrupted by the injection of either anti-EphB2 antibodies or silencing the Fkbp5 gene in the amygdala of wild-type mice. Our findings establish a novel neuronal pathway linking stress-induced proteolysis of EphB2 in the amygdala to anxiety.


Subject(s)
Amygdala/metabolism , Anxiety/metabolism , Kallikreins/metabolism , Receptor, EphB2/metabolism , Amygdala/cytology , Animals , Anxiety/genetics , Anxiety Disorders/etiology , Anxiety Disorders/genetics , Anxiety Disorders/metabolism , Electric Conductivity , Fear , Gene Expression Regulation , Kallikreins/deficiency , Kallikreins/genetics , Long-Term Potentiation , Mice , Mice, Inbred C57BL , Neuronal Plasticity , Neurons/metabolism , Protein Binding , Receptor, EphB2/chemistry , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism , Stress, Psychological/metabolism , Tacrolimus Binding Proteins/genetics
12.
J Neurosci ; 30(40): 13235-45, 2010 Oct 06.
Article in English | MEDLINE | ID: mdl-20926649

ABSTRACT

Regulation of the resting membrane potential and the repolarization of neurons are important in regulating neuronal excitability. The potassium channel subunits Kv7.2 and Kv7.3 play a key role in stabilizing neuronal activity. Mutations in KCNQ2 and KCNQ3, the genes encoding Kv7.2 and Kv7.3, cause a neonatal form of epilepsy, and activators of these channels have been identified as novel antiepileptics and analgesics. Despite the observations that regulation of these subunits has profound effects on neuronal function, almost nothing is known about the mechanisms responsible for controlling appropriate expression levels. Here we identify two mechanisms responsible for regulating KCNQ2 and KCNQ3 mRNA levels. We show that the transcription factor Sp1 activates expression of both KCNQ2 and KCNQ3, whereas the transcriptional repressor REST (repressor element 1-silencing transcription factor) represses expression of both of these genes. Furthermore, we show that transcriptional regulation of KCNQ genes is mirrored by the correlated changes in M-current density and excitability of native sensory neurons. We propose that these mechanisms are important in the control of excitability of neurons and may have implications in seizure activity and pain.


Subject(s)
Gene Expression Regulation/physiology , KCNQ2 Potassium Channel/genetics , KCNQ3 Potassium Channel/genetics , Repressor Proteins/physiology , Sensory Receptor Cells/physiology , Sp1 Transcription Factor/physiology , Transcriptional Activation/genetics , Animals , Cell Line , Cell Line, Tumor , Chronic Disease , Epilepsy/genetics , Epilepsy/physiopathology , Humans , KCNQ2 Potassium Channel/antagonists & inhibitors , KCNQ2 Potassium Channel/biosynthesis , KCNQ3 Potassium Channel/antagonists & inhibitors , KCNQ3 Potassium Channel/biosynthesis , Neural Inhibition/genetics , Neural Pathways/physiopathology , Pain/genetics , Pain/physiopathology , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , Rats , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Sp1 Transcription Factor/genetics , Up-Regulation/physiology
13.
Gene ; 300(1-2): 63-8, 2002 Oct 30.
Article in English | MEDLINE | ID: mdl-12468087

ABSTRACT

Recent localization of cohesin association regions along the yeast chromatin fibre suggests that compositional variability of DNA in yeast is related to the function and organization of the chromosomal loops. The bases of the loops, where the chromatin fibre is attached to the chromosomal axis, are AT-rich, bind cohesin, and are flanked by genes transcribed convergently. The hotspots of meiotic recombination are mainly found in the GC-rich parts of the loops, 'external' with respect to the chromosomal axis, frequently in the vicinity of the promoters of divergently transcribed genes. There are two possible reasons why the regions of the hotspots of recombination were enriched in GC content during evolution. One is a biased repair of recombination intermediates, and the second is a selective advantage due to an increased chromatin accessibility, which may have the carriers of GC-enriched alleles over the carriers of AT-rich alleles.


Subject(s)
Chromatin/genetics , DNA, Fungal/genetics , Saccharomyces cerevisiae/genetics , Base Composition , Chromatin/physiology , Chromosomes, Fungal/genetics , Chromosomes, Fungal/physiology , DNA Repair , DNA, Fungal/chemistry , Mutation , Recombination, Genetic , Saccharomyces cerevisiae/physiology , Selection, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...