Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Langmuir ; 36(18): 5048-5057, 2020 May 12.
Article in English | MEDLINE | ID: mdl-32302141

ABSTRACT

Stimuli-responsive compartments are attracting more and more attention through the years motivated by their wide applications in different fields including encapsulation, manipulation, and triggering of chemical reactions on demand. Among others, magnetic responsive compartments are particularly attractive due to the numerous advantages of magnetic fields compared to other external stimuli. In this article, we used an oil-based ferrofluid where the magnetic nanoparticles have been coated with different polymers to increase their amphiphilic character and surface activity, consequently rendering the interface magnetically responsive. Microliter aqueous nonmagnetic droplets dispersed in the oil-based ferrofluid were used as a model of microreactors. A comprehensive experimental and theoretical study of the deformation, attraction, and coalescence processes of the nonmagnetic water droplets coated with the magnetic nanoparticles under an applied magnetic field in the continuous oil-based ferrofluid phase is provided. To manipulate the packing of the nanoparticles at the water/oil interface, the ionic strength of the aqueous droplets was varied using different NaCl concentrations, and its effect on modulating the coalescence of the droplets was probed. Our results show that the water droplets deform along the magnetic field depending on the magnetic properties of the ferrofluid itself and on the surface properties of the interface, attract in pairs under the action of the magnetic dipole force, and coalesce by the action of the same force with a stochastic behavior. We have studied all of these phenomena as a function of the magnetic field applied, evaluating in each case the forces and/or pressures acting on the droplets with particular attention to roles of magnetic attraction, interface properties, and viscosity in the system. This work offers an overall set of tools to understand and predict the behavior of multiple water droplets in an oil-based ferrofluid for lab-on-a-chip applications.

2.
Mol Psychiatry ; 22(4): 570-579, 2017 04.
Article in English | MEDLINE | ID: mdl-27595592

ABSTRACT

Reward-related memory is an important factor in cocaine seeking. One necessary signaling mechanism for long-term memory formation is the activation of poly(ADP-ribose) polymerase-1 (PARP-1), via poly(ADP-ribosyl)ation. We demonstrate herein that auto-poly(ADP-ribosyl)ation of activated PARP-1 was significantly pronounced during retrieval of cocaine-associated contextual memory, in the central amygdala (CeA) of rats expressing cocaine-conditioned place preference (CPP). Intra-CeA pharmacological and short hairpin RNA depletion of PARP-1 activity during cocaine-associated memory retrieval abolished CPP. In contrast, PARP-1 inhibition after memory retrieval did not affect CPP reconsolidation process and subsequent retrievals. Chromatin immunoprecipitation sequencing revealed that PARP-1 binding in the CeA is highly enriched in genes involved in neuronal signaling. We identified among PARP targets in CeA a single gene, yet uncharacterized and encoding a putative transposase inhibitor, at which PARP-1 enrichment markedly increases during cocaine-associated memory retrieval and positively correlates with CPP. Our findings have important implications for understanding drug-related behaviors, and suggest possible future therapeutic targets for drug abuse.


Subject(s)
Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerases/genetics , ADP-Ribosylation Factors/metabolism , Amygdala/metabolism , Animals , Cocaine/adverse effects , Cocaine/metabolism , Cocaine/pharmacology , Male , Memory/drug effects , Poly (ADP-Ribose) Polymerase-1/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Rats , Rats, Sprague-Dawley , Transposases/antagonists & inhibitors
3.
Transl Psychiatry ; 6(11): e943, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27824361

ABSTRACT

Exposure to early-life stress (ELS) may heighten the risk for psychopathology at adulthood. Here, in order to identify common genes that may keep the memory of ELS through changes in their methylation status, we intersected methylome analyses performed in different tissues and time points in rats, non-human primates and humans, all characterized by ELS. We identified Ankyrin-3 (Ank3), a scaffolding protein with a strong genetic association for psychiatric disorders, as a gene persistently affected by stress exposure. In rats, Ank3 methylation and mRNA changes displayed a specific temporal profile during the postnatal development. Moreover, exposure to prenatal stress altered the interaction of ankyrin-G, the protein encoded by Ank3 enriched in the post-synaptic compartment, with PSD95. Notably, to model in humans a gene by early stress interplay on brain phenotypes during cognitive performance, we demonstrated an interaction between functional variation in Ank3 gene and obstetric complications on working memory in healthy adult subjects. Our data suggest that alterations of Ank3 expression and function may contribute to the effects of ELS on the development of psychiatric disorders.


Subject(s)
Ankyrins/genetics , Disease Models, Animal , Genetic Markers/genetics , Genetic Predisposition to Disease/genetics , Life Change Events , Mental Disorders/genetics , Prenatal Exposure Delayed Effects/genetics , Animals , Bipolar Disorder/genetics , Cohort Studies , DNA Methylation , Female , Genome-Wide Association Study , Humans , Infant, Newborn , Macaca mulatta , Male , Memory, Short-Term , Phenotype , Pregnancy , Promoter Regions, Genetic/genetics , Rats , Schizophrenia/genetics
4.
Transl Psychiatry ; 5: e545, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25849984

ABSTRACT

Depression affects 10-15% of pregnant women and has been associated with preterm delivery and later developmental, behavioural and learning disabilities. We tested the hypothesis that maternal depression is associated with DNA methylation alterations in maternal T lymphocytes, neonatal cord blood T lymphocytes and adult offspring hippocampi. Genome-wide DNA methylation of CD3+ T lymphocytes isolated from 38 antepartum maternal and 44 neonatal cord blood samples were analyzed using Illumina Methylation 450 K microarrays. Previously obtained methylation data sets using methylated DNA immunoprecipitation and array-hybridization of 62 postmortem hippocampal samples of adult males were re-analyzed to test associations with history of maternal depression. We found 145 (false discovery rate (FDR) q<0.05) and 2520 (FDR q<0.1) differentially methylated CG-sites in cord blood T lymphocytes of neonates from the maternal depression group as compared with the control group. However, no significant DNA methylation differences were detected in the antepartum maternal T lymphocytes of our preliminary data set. We also detected 294 differentially methylated probes (FDR q<0.1) in hippocampal samples associated with history of maternal depression. We observed a significant overlap (P=0.002) of 33 genes with changes in DNA methylation in T lymphocytes of neonates and brains of adult offspring. Many of these genes are involved in immune system functions. Our results show that DNA methylation changes in offspring associated with maternal depression are detectable at birth in the immune system and persist to adulthood in the brain. This is consistent with the hypothesis that system-wide epigenetic changes are involved in life-long responses to maternal depression in the offspring.


Subject(s)
DNA Methylation/immunology , Depressive Disorder/immunology , Fetal Blood/immunology , Hippocampus/immunology , Mothers/psychology , T-Lymphocytes/immunology , Adult , Epigenesis, Genetic/immunology , Female , Humans , Pregnancy
5.
Transl Psychiatry ; 5: e515, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25710121

ABSTRACT

Prenatal maternal stress (PNMS) can impact a variety of outcomes in the offspring throughout childhood and persisting into adulthood as shown in human and animal studies. Many of the effects of PNMS on offspring outcomes likely reflect the effects of epigenetic changes, such as DNA methylation, to the fetal genome. However, no animal or human research can determine the extent to which the effects of PNMS on DNA methylation in human offspring is the result of the objective severity of the stressor to the pregnant mother, or her negative appraisal of the stressor or her resulting degree of negative stress. We examined the genome-wide DNA methylation profile in T cells from 34 adolescents whose mothers had rated the 1998 Québec ice storm's consequences as positive or negative (that is, cognitive appraisal). The methylation levels of 2872 CGs differed significantly between adolescents in the positive and negative maternal cognitive appraisal groups. These CGs are affiliated with 1564 different genes and with 408 different biological pathways, which are prominently featured in immune function. Importantly, there was a significant overlap in the differentially methylated CGs or genes and biological pathways that are associated with cognitive appraisal and those associated with objective PNMS as we reported previously. Our study suggests that pregnant women's cognitive appraisals of an independent stressor may have widespread effects on DNA methylation across the entire genome of their unborn children, detectable during adolescence. Therefore, cognitive appraisals could be an important predictor variable to explore in PNMS research.


Subject(s)
Cognition/physiology , DNA Methylation/physiology , Disasters , Pregnant Women , Prenatal Exposure Delayed Effects/physiopathology , Stress, Psychological/complications , Adolescent , Climatic Processes , Epigenesis, Genetic , Female , Humans , Ice , Male , Pregnancy , Quebec , Severity of Illness Index , Surveys and Questionnaires
6.
Mol Psychiatry ; 20(3): 405-12, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24776738

ABSTRACT

High ethanol intake is well known to induce both anxiolytic and anxiogenic effects, in correlation with chromatin remodeling in the amygdaloid brain region and deficits in cell proliferation and survival in the hippocampus of rodents. Whether only moderate but chronic ethanol intake in C57BL/6J mice could also have an impact on chromatin remodeling and neuroplasticity was addressed here. Chronic ethanol consumption in a free choice paradigm was found to induce marked changes in the expression of genes implicated in neural development and histone post-translational modifications in the mouse hippocampus. Transcripts encoding neural bHLH activators and those from Bdnf exons II, III and VI were upregulated, whereas those from Bdnf exon VIII and Hdacs were downregulated by ethanol compared with water consumption. These ethanol-induced changes were associated with enrichment in both acetylated H3 at Bdnf promoter PVI and trimethylated H3 at PII and PIII. Conversely, acetylated H3 at PIII and PVIII and trimethylated H3 at PVIII were decreased in ethanol-exposed mice. In parallel, hippocampal brain-derived neurotrophic factor (BDNF) levels and TrkB-mediated neurogenesis in the dentate gyrus were significantly enhanced by ethanol consumption. These results suggest that, in C57BL/6J mice, chronic and moderate ethanol intake produces marked epigenetic changes underlying BDNF overexpression and downstream hippocampal neurogenesis.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Central Nervous System Depressants/pharmacology , Epigenesis, Genetic/drug effects , Ethanol/pharmacology , Hippocampus/drug effects , Animals , Azepines/pharmacology , Benzamides/pharmacology , Brain-Derived Neurotrophic Factor/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Survival/drug effects , Choice Behavior/drug effects , Conditioning, Operant , Drinking/drug effects , Exons , Hippocampus/cytology , Hippocampus/metabolism , Histones/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/drug effects , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/metabolism
7.
Transl Psychiatry ; 4: e485, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25423137

ABSTRACT

Stress-induced alterations in neuronal plasticity and in hippocampal functions have been suggested to be involved in the development of mood disorders. In this context, we investigated in the hippocampus the activation of intracellular signaling cascades, the expression of epigenetic markers and plasticity-related genes in a mouse model of stress-induced hyperactivity and of mixed affective disorders. We also determined whether the antidepressant drug agomelatine, a MT1/MT2 melatonergic receptor agonist/5-HT2C receptor antagonist, could prevent some neurobiological and behavioral alterations produced by stress. C57BL/6J mice, exposed for 3 weeks to daily unpredictable socio-environmental stressors of mild intensity, were treated during the whole procedure with agomelatine (50 mg kg(-1) per day, intraperitoneal). Stressed mice displayed robust increases in emotional arousal, vigilance and motor activity, together with a reward deficit and a reduction in anxiety-like behavior. Neurobiological investigations showed an increased phosphorylation of intracellular signaling proteins, including Atf1, Creb and p38, in the hippocampus of stressed mice. Decreased hippocampal level of the repressive epigenetic marks HDAC2 and H3K9me2, as well as increased level of the permissive mark H3K9/14ac suggested that chronic mild stress was associated with increased gene transcription, and clear-cut evidence was further indicated by changes in neuroplasticity-related genes, including Arc, Bcl2, Bdnf, Gdnf, Igf1 and Neurod1. Together with other findings, the present data suggest that chronic ultra-mild stress can model the hyperactivity or psychomotor agitation, as well as the mixed affective behaviors often observed during the manic state of bipolar disorder patients. Interestingly, agomelatine could normalize both the behavioral and the molecular alterations induced by stress, providing further insights into the mechanism of action of this new generation antidepressant drug.


Subject(s)
Acetamides/pharmacology , Antidepressive Agents/pharmacology , Behavior, Animal/physiology , Depression/drug therapy , Epigenesis, Genetic/physiology , Hippocampus/metabolism , Neuronal Plasticity/physiology , Signal Transduction/physiology , Stress, Psychological/complications , Acetamides/administration & dosage , Affective Symptoms/drug therapy , Affective Symptoms/etiology , Animals , Antidepressive Agents/administration & dosage , Behavior, Animal/drug effects , Depression/etiology , Disease Models, Animal , Epigenesis, Genetic/drug effects , Hippocampus/drug effects , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Psychomotor Agitation/drug therapy , Psychomotor Agitation/etiology , Receptors, Melatonin/agonists , Serotonin 5-HT2 Receptor Antagonists/administration & dosage , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Signal Transduction/drug effects , Stress, Psychological/metabolism
8.
Transl Psychiatry ; 4: e429, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25158004

ABSTRACT

Early life stress (ELS) is associated with increased vulnerability for diseases in later life, including psychiatric disorders. Animal models and human studies suggest that this effect is mediated by epigenetic mechanisms. In humans, epigenetic studies to investigate the influence of ELS on psychiatric phenotypes are limited by the inaccessibility of living brain tissue. Due to the tissue-specific nature of epigenetic signatures, it is impossible to determine whether ELS induced epigenetic changes in accessible peripheral cells, for example, blood lymphocytes, reflect epigenetic changes in the brain. To overcome these limitations, we applied a cross-species approach involving: (i) the analysis of CD34+ cells from human cord blood; (ii) the examination of blood-derived CD3+ T cells of newborn and adolescent nonhuman primates (Macaca mulatta); and (iii) the investigation of the prefrontal cortex of adult rats. Several regions in MORC1 (MORC family CW-type zinc finger 1; previously known as: microrchidia (mouse) homolog) were differentially methylated in response to ELS in CD34+ cells and CD3+ T cells derived from the blood of human and monkey neonates, as well as in CD3+ T cells derived from the blood of adolescent monkeys and in the prefrontal cortex of adult rats. MORC1 is thus the first identified epigenetic marker of ELS to be present in blood cell progenitors at birth and in the brain in adulthood. Interestingly, a gene-set-based analysis of data from a genome-wide association study of major depressive disorder (MDD) revealed an association of MORC1 with MDD.


Subject(s)
DNA Methylation/genetics , Depressive Disorder, Major/genetics , Epigenesis, Genetic/genetics , Genome-Wide Association Study , Stress, Psychological/complications , Animals , Animals, Newborn , Cohort Studies , Female , Fetal Blood/cytology , Genetic Predisposition to Disease/genetics , Humans , Infant, Newborn , Macaca mulatta , Prefrontal Cortex/metabolism , Pregnancy , Species Specificity , Stem Cells , T-Lymphocytes/metabolism
9.
Neuroscience ; 268: 139-48, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24657458

ABSTRACT

5-Hydroxymethylcytosine (5hmC) is abundant in the brain, suggesting an important role in epigenetic control of neuronal functions. In this paper, we show that 5hmC and 5-methylcytosine (5mC) levels are coordinately distributed in gene promoters of the rhesus macaque prefrontal cortex. Although promoter hydroxymethylation and methylation are overall negatively correlated with expression, a subset of highly expressed genes involved in specific cerebral functions is associated with high levels of 5mC and 5hmC. These relationships were also observed in the mouse cortex. Furthermore, we found that early-life maternal deprivation is associated, in the adult monkey cortex, with DNA hydroxymethylation changes of promoters of genes related to neurological functions and psychological disorders. These results reveal that early social adversity triggers variations in brain DNA hydroxymethylation that could be detected in adulthood.


Subject(s)
5-Methylcytosine/metabolism , Cytosine/analogs & derivatives , DNA Methylation , Epigenesis, Genetic , Maternal Deprivation , Prefrontal Cortex/metabolism , Animals , Cytosine/metabolism , Databases, Genetic , Frontal Lobe/metabolism , Gene Expression , Macaca mulatta , Methylation , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic
10.
Transl Psychiatry ; 4: e347, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24448209

ABSTRACT

Sleep is critical for normal brain function and mental health. However, the molecular mechanisms mediating the impact of sleep loss on both cognition and the sleep electroencephalogram remain mostly unknown. Acute sleep loss impacts brain gene expression broadly. These data contributed to current hypotheses regarding the role for sleep in metabolism, synaptic plasticity and neuroprotection. These changes in gene expression likely underlie increased sleep intensity following sleep deprivation (SD). Here we tested the hypothesis that epigenetic mechanisms coordinate the gene expression response driven by SD. We found that SD altered the cortical genome-wide distribution of two major epigenetic marks: DNA methylation and hydroxymethylation. DNA methylation differences were enriched in gene pathways involved in neuritogenesis and synaptic plasticity, whereas large changes (>4000 sites) in hydroxymethylation where observed in genes linked to cytoskeleton, signaling and neurotransmission, which closely matches SD-dependent changes in the transcriptome. Moreover, this epigenetic remodeling applied to elements previously linked to sleep need (for example, Arc and Egr1) and synaptic partners of Neuroligin-1 (Nlgn1; for example, Dlg4, Nrxn1 and Nlgn3), which we recently identified as a regulator of sleep intensity following SD. We show here that Nlgn1 mutant mice display an enhanced slow-wave slope during non-rapid eye movement sleep following SD but this mutation does not affect SD-dependent changes in gene expression, suggesting that the Nlgn pathway acts downstream to mechanisms triggering gene expression changes in SD. These data reveal that acute SD reprograms the epigenetic landscape, providing a unique molecular route by which sleep can impact brain function and health.


Subject(s)
Cerebral Cortex/metabolism , DNA Methylation/physiology , Genome/genetics , Neuronal Plasticity/genetics , Sleep Deprivation/metabolism , Transcriptome/genetics , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cerebral Cortex/physiopathology , DNA Methylation/genetics , Electroencephalography , Epigenesis, Genetic/genetics , Epigenesis, Genetic/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Sleep Deprivation/physiopathology , Sleep Stages/genetics , Sleep Stages/physiology
11.
J Phys Condens Matter ; 20(20): 204104, 2008 May 21.
Article in English | MEDLINE | ID: mdl-21694234

ABSTRACT

This paper describes an electrochemical method which permits us to transform solid metals (cobalt, iron or nickel) into nanoparticles. An electrolysis cell is made, the anode being a metal bar and the cathode a mercury layer. Magnetic nanoparticles are obtained in one step by electroreduction of mercury. Electrolysis is performed in an aqueous medium at pH above 6 in order to avoid the reduction of protons. The magnetic nanoparticles obtained are kept in mercury and can be recovered in an organic solvent.

12.
Phys Rev Lett ; 64(5): 539-542, 1990 Jan 29.
Article in English | MEDLINE | ID: mdl-10042010
SELECTION OF CITATIONS
SEARCH DETAIL
...