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.
J Neurochem ; 160(2): 234-255, 2022 01.
Article in English | MEDLINE | ID: mdl-34816431

ABSTRACT

The nervous system monitors the environment to maintain homeostasis, which can be affected by stressful conditions. Using mammalian models of chronic stress, we previously observed altered brain levels of GPM6A, a protein involved in neuronal morphology. However, GPM6A's role in systemic stress responses remains unresolved. The nematode Caenorhabditis elegans expresses a GPM6A ortholog, the neuronal membrane glycoprotein 1 (NMGP-1). Because of the shared features between nematode and mammalian nervous systems and the vast genetic tools available in C. elegans, we used the worm to elucidate the role of GPM6A in the stress response. We first identified nmgp-1 expression in different amphid and phasmid neurons. To understand the nmgp-1 role, we characterized the behavior of nmgp-1(RNAi) animals and two nmgp-1 mutant alleles. Compared to control animals, mutant and RNAi-treated worms exhibited increased recovery time from the stress-resistant dauer stage, altered SDS chemosensation and reduced egg-laying rate resulting in egg retention (bag-of-worms phenotype). Silencing of nmgp-1 expression induced morphological abnormalities in the ASJ sensory neurons, partly responsible for dauer exit. These results indicate that nmgp-1 is required for neuronal morphology and for behaviors associated with chemosensation. Finally, we propose nmgp-1 mutants as a tool to screen drugs for human nervous system pathologies.


Subject(s)
Adaptation, Physiological/physiology , Behavior, Animal/physiology , Caenorhabditis elegans/physiology , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Animals , Caenorhabditis elegans Proteins/metabolism , Female
2.
J Mol Neurosci ; 65(3): 301-311, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29931501

ABSTRACT

Prenatal stress (PS) induces molecular changes that alter neural connectivity, increasing the risk for neuropsychiatric disorders. Here we analyzed -in the hippocampus of adult rats exposed to PS- the epigenetic signature mediating the PS-induced neuroplasticity changes. Furthermore, using cultured hippocampal neurons, we investigated the effects on neuroplasticity of an epigenetic modulator. PS induced significant modifications in the mRNA levels of stress-related transcription factor MEF2A, SUV39H1 histone methyltransferase, and TET1 hydroxylase, indicating that PS modifies gene expression through chromatin remodeling. In in vitro analysis, histone acetylation inhibition with apicidin increased filopodium density, suggesting that the external regulation of acetylation levels might modulate neuronal morphology. These results offer a way to enhance neural connectivity that could be considered to revert PS effects.


Subject(s)
Epigenesis, Genetic , Histone Code , Neuronal Plasticity , Prenatal Exposure Delayed Effects/genetics , Stress, Psychological/genetics , Animals , Cells, Cultured , Dioxygenases/genetics , Dioxygenases/metabolism , Female , Hippocampus/cytology , Hippocampus/growth & development , Hippocampus/metabolism , Histone Deacetylase Inhibitors/pharmacology , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Male , Methyltransferases/genetics , Methyltransferases/metabolism , Neurogenesis , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Peptides, Cyclic/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/physiopathology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Repressor Proteins/genetics , Repressor Proteins/metabolism , Stress, Psychological/metabolism , Stress, Psychological/physiopathology
3.
Sci Rep ; 7(1): 9788, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28851962

ABSTRACT

Membrane neuronal glycoprotein M6a is highly expressed in the brain and contributes to neural plasticity promoting neurite growth and spine and synapse formation. We have previously showed that chronic stressors alter hippocampal M6a mRNA levels in rodents and tree shrews. We now show that M6a glycoprotein can be detected in mouse blood. M6a is a transmembrane glycoprotein and, as such, unlikely to be free in blood. Here we demonstrate that, in blood, M6a is transported in extracellular vesicles (EVs). It is also shown that M6a-containing EVs are delivered from cultured primary neurons as well as from M6a-transfected COS-7 cells. Released EVs containing M6a can be incorporated into COS-7 cells changing its phenotype through formation of membrane protrusions. Thus, M6a-containing EVs might contribute to maintain cellular plasticity. M6a presence in blood was used to monitor stress effects. Chronic restraint stress modulated M6a protein level in a sex dependent manner. Analysis of individual animals indicated that M6a level variations depend on the stressor applied. The response to stressors in blood makes M6a amenable to further studies in the stress disorder field.


Subject(s)
Extracellular Vesicles/metabolism , Membrane Glycoproteins/blood , Nerve Tissue Proteins/blood , Stress, Physiological , Animals , Biological Transport , Biomarkers , COS Cells , Chlorocebus aethiops , Extracellular Vesicles/ultrastructure , Female , Fluorescent Antibody Technique , Hippocampus/metabolism , Male , Membrane Glycoproteins/cerebrospinal fluid , Membrane Glycoproteins/genetics , Mice , Nerve Tissue Proteins/cerebrospinal fluid , Nerve Tissue Proteins/genetics , Neurons/metabolism , Sex Factors
4.
Neurochem Int ; 88: 73-87, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26026592

ABSTRACT

Previous studies from our laboratory have shown that male adult offspring of stressed mothers exhibited higher levels of ionotropic and metabotropic glutamate receptors than control rats. These offspring also showed long-lasting astroglial hypertrophy and a reduced dendritic arborization with synaptic loss. Since metabolism of glutamate is dependent on interactions between neurons and surrounding astroglia, our results suggest that glutamate neurotransmitter pathways might be impaired in the brain of prenatally stressed rats. To study the effect of prenatal stress on the metabolism and neurotransmitter function of glutamate, pregnant rats were subjected to restrain stress during the last week of gestation. Brains of the adult offspring were used to assess glutamate metabolism, uptake and release as well as expression of glutamate receptors and transporters. While glutamate metabolism was not affected it was found that prenatal stress (PS) changed the expression of the transporters, thus, producing a higher level of vesicular vGluT-1 in the frontal cortex (FCx) and elevated levels of GLT1 protein and messenger RNA in the hippocampus (HPC) of adult male PS offspring. We also observed increased uptake capacity for glutamate in the FCx of PS male offspring while no such changes were observed in the HPC. The results show that changes mediated by PS on the adult glutamatergic system are brain region specific. Overall, PS produces long-term changes in the glutamatergic system modulating the expression of glutamate transporters and altering synaptic transmission of the adult brain.


Subject(s)
Glutamic Acid/metabolism , Prenatal Exposure Delayed Effects/metabolism , Stress, Psychological/metabolism , Synaptic Transmission/physiology , Animals , Female , Hippocampus/metabolism , Male , Organ Culture Techniques , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Rats , Rats, Wistar , Stress, Psychological/complications
5.
Stress ; 18(4): 435-45, 2015.
Article in English | MEDLINE | ID: mdl-25798813

ABSTRACT

Prenatal stress (PS) strongly impacts fetal brain development and function in adulthood. In normal aging and Alzheimer's disease, there is hypothalamic-pituitary-adrenal axis dysfunction and loss of cholinergic neurons and neuronal nicotinic acetylcholine receptors (nAChRs). This study investigated whether prenatal restraint stress affects nAChR expression in the brain of adult offspring. For PS, pregnant dams were placed in a plastic restrainer for 45 min, three times daily during the last week of pregnancy; controls were undisturbed. Male offspring were analyzed at postnatal day (PND) 60 (n = 4 rats per group). Western blot (WB) and fluorescence microscopy showed that PS decreased α7-AChR subunit expression (∼50%) in the frontal cortex in the adult offspring. PS decreased significantly the number of α7-AChR-expressing cells in the medial prefrontal cortex (by ∼25%) and in the sensory-motor cortex (by ∼20%) without affecting the total cell number in those areas. No alterations were found in the hippocampus by quantitative polymerase chain reaction (qPCR), or WB analysis, but a detailed fluorescence microscopy analysis showed that PS affected α7-AChR mainly in the CA3 and dentate gyrus subfields: PS decreased α7-AChR subunit expression by ∼25 and ∼30%, respectively. Importantly, PS decreased the number of α7-AChR-expressing cells and the total cell number (by ∼15 and 20%, respectively) in the dentate gyrus. PS differently affected α4-AChR: PS impaired its mRNA expression in the frontal cortex (by ∼50%), without affecting protein levels. These results demonstrate that disturbances during gestation produce long-term alterations in the expression pattern of α7-AChR in rat brain.


Subject(s)
Brain/metabolism , Pregnancy Complications/genetics , Prenatal Exposure Delayed Effects/genetics , RNA, Messenger/metabolism , Stress, Psychological/genetics , alpha7 Nicotinic Acetylcholine Receptor/genetics , Alzheimer Disease , Animals , Female , Hippocampus/metabolism , Hypothalamo-Hypophyseal System/metabolism , Male , Pituitary-Adrenal System/metabolism , Prefrontal Cortex/metabolism , Pregnancy , Pregnancy Complications/metabolism , Prenatal Exposure Delayed Effects/metabolism , Rats , Restraint, Physical , Reverse Transcriptase Polymerase Chain Reaction , Sensorimotor Cortex/metabolism , Stress, Psychological/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism
6.
Epigenetics ; 9(1): 152-60, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23959066

ABSTRACT

Prenatal stress (PS) exerts strong impact on fetal brain development and on adult offspring brain functions. Previous work demonstrated that chronic stress alters the mRNA expression of GPM6A, a neuronal glycoprotein involved in filopodium extension. In this work, we analyzed the effect of PS on gpm6a expression and the epigenetic mechanisms involved. Pregnant Wistar rats received restraint stress during the last week of gestation. Male offspring were sacrificed on postnatal days 28 and 60. Hippocampus and prefrontal cortex samples were analyzed for gene expression (qPCR for mRNAs and microRNAs), methylation status (bisulfite conversion) and protein levels. Hippocampal neurons in culture were used to analyze microRNA overexpression effects. Prenatal stress induced changes in gpm6a levels in both tissues and at both ages analyzed, indicating a persistent effect. Two CpG islands in the gpm6a gene were identified. Variations in the methylation pattern at three specific CpGs were found in hippocampus, but not in PFC samples from PS offspring. microRNAs predicted to target gpm6a were identified in silico. qPCR measurements showed that PS modified the expression of several microRNAs in both tissues, being microRNA-133b the most significantly altered. Further studies overexpressing this microRNA in neuronal cultures showed a reduction in gmp6a mRNA and protein level. Moreover filopodium density was also reduced, suggesting that GPM6A function was affected. Gestational stress affected gpm6a gene expression in offspring likely through changes in methylation status and in posttranscriptional regulation by microRNAs. Thus, our findings propose gpm6a as a novel target for epigenetic regulation during prenatal stress.


Subject(s)
Brain/metabolism , Epigenesis, Genetic , Glycoproteins/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Prenatal Exposure Delayed Effects/metabolism , Stress, Psychological/metabolism , Animals , Brain/embryology , Cells, Cultured , CpG Islands , Female , Glycoproteins/genetics , Hippocampus/embryology , Hippocampus/metabolism , Male , Membrane Glycoproteins/genetics , Methylation , MicroRNAs/genetics , MicroRNAs/metabolism , Nerve Tissue Proteins/genetics , Neurons/metabolism , Prefrontal Cortex/embryology , Prefrontal Cortex/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Rats, Wistar , Stress, Psychological/genetics
7.
Stress ; 16(4): 429-40, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23252714

ABSTRACT

Several studies have demonstrated that the presence of stressors during pregnancy induces adverse effects on the neuroendocrine system of the offspring later in life. In the present work, we investigated the effects of early programming on the male reproductive system, employing a prenatal stress (PS) paradigm. This study found that when pregnant dams were placed in a plastic restrainer three times a day during the last week of pregnancy, the offspring showed reduced anogenital distance and delayed testicular descent. Serum luteinising hormone (LH) and follicle-stimulating hormone (FSH) levels were decreased at postnatal day (PND) 28 and testosterone was decreased at PND 75. Increased testosterone plus dihydrotestosterone (T + DHT) concentrations correlated with increased testicular 5α Reductase-1 (5αR-1) mRNA expression at PND 28. Moreover, PS accelerated spermatogenesis at PND 35 and 60, and increased mean seminiferous tubule diameter in pubertal offspring and reduced Leydig cell number was observed at PND 35 and 60. PS offspring had increased androgen receptor (AR) mRNA level at PND 28, and at PND 35 had increased the numbers of Sertoli cells immunopositive for AR. Overall, the results confirm that stress during gestation can induce long-term effects on the male offspring reproductive system. Of particular interest is the pre-pubertal imbalance of circulating hormones that probably trigger accelerated testicular development, followed by an increase in total androgens and a decrease in testosterone concentration during adulthood. Exposure to an unfavourable intrauterine environment might prepare for harsh external conditions by triggering early puberty, increasing reproductive potential.


Subject(s)
Maternal Exposure , Prenatal Exposure Delayed Effects , Stress, Psychological , Testis/growth & development , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/biosynthesis , Animals , Dihydrotestosterone/blood , Female , Follicle Stimulating Hormone/blood , Leydig Cells/metabolism , Luteinizing Hormone/blood , Male , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Androgen/metabolism , Restraint, Physical , Spermatogenesis , Testosterone/blood
8.
PLoS One ; 7(6): e38948, 2012.
Article in English | MEDLINE | ID: mdl-22701734

ABSTRACT

PTP1B is an endoplasmic reticulum (ER) anchored enzyme whose access to substrates is partly dependent on the ER distribution and dynamics. One of these substrates, the protein tyrosine kinase Src, has been found in the cytosol, endosomes, and plasma membrane. Here we analyzed where PTP1B and Src physically interact in intact cells, by bimolecular fluorescence complementation (BiFC) in combination with temporal and high resolution microscopy. We also determined the structural basis of this interaction. We found that BiFC signal is displayed as puncta scattered throughout the ER network, a feature that was enhanced when the substrate trapping mutant PTP1B-D181A was used. Time-lapse and co-localization analyses revealed that BiFC puncta did not correspond to vesicular carriers; instead they localized at the tip of dynamic ER tubules. BiFC puncta were retained in ventral membrane preparations after cell unroofing and were also detected within the evanescent field of total internal reflection fluorescent microscopy (TIRFM) associated to the ventral membranes of whole cells. Furthermore, BiFC puncta often colocalized with dark spots seen by surface reflection interference contrast (SRIC). Removal of Src myristoylation and polybasic motifs abolished BiFC. In addition, PTP1B active site and negative regulatory tyrosine 529 on Src were primary determinants of BiFC occurrence, although the SH3 binding motif on PTP1B also played a role. Our results suggest that ER-bound PTP1B dynamically interacts with the negative regulatory site at the C-terminus of Src at random puncta in the plasma membrane/substrate interface, likely leading to Src activation and recruitment to adhesion complexes. We postulate that this functional ER/plasma membrane crosstalk could apply to a wide array of protein partners, opening an exciting field of research.


Subject(s)
Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , src-Family Kinases/metabolism , Animals , Antibodies, Monoclonal , Blotting, Western , CHO Cells , Cell Adhesion/physiology , Cricetinae , Cricetulus , Fluorescent Antibody Technique , Humans , Mice , Microscopy, Fluorescence , Polymerase Chain Reaction , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Time-Lapse Imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...