Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Stress ; 18(1): 96-106, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25472821

ABSTRACT

Chronic stress is considered to be a major risk factor in the development of psychopathological syndromes in humans. Cognitive impairments and long-term potentiation (LTP) impairments are increasingly recognized as major components of depression, anxiety disorders and other stress-related chronic psychological illnesses. It seems timely to systematically study the potentially underlying neurobiological mechanisms of altered cognitive and synaptic plasticity in the course of chronic stress. In the present study, a rat model of chronic unpredictable stress (CUS) induced a cognitive impairment in spatial memory in the Morris water maze (MWM) test and a hippocampal LTP impairment. CUS also induced hippocampal microglial activation and attenuated phosphorylation of glutamate receptor 1 (GluR1 or GluA1). Moreover, chronic treatment with the selective microglial activation blocker, minocycline (120 mg/kg per day), beginning 3 d before CUS treatment and continuing through the behavioral testing period, prevented the CUS-induced impairments of spatial memory and LTP induction. Additional studies showed that minocycline-induced inhibition of microglia activation was associated with increased phosphorylation of GluR1. These results suggest that hippocampal microglial activation modulates the level of GluR1 phosphorylation and might play a causal role in CUS-induced cognitive and LTP disturbances.


Subject(s)
Behavior, Animal , Cognition Disorders/etiology , Cognition , Hippocampus/metabolism , Microglia/metabolism , Receptors, AMPA/metabolism , Stress, Psychological/complications , Animals , Behavior, Animal/drug effects , Chronic Disease , Cognition/drug effects , Cognition Disorders/metabolism , Cognition Disorders/physiopathology , Cognition Disorders/psychology , Cytokines/metabolism , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/physiopathology , In Vitro Techniques , Inflammation Mediators/metabolism , Long-Term Potentiation , Male , Maze Learning , Microglia/drug effects , Minocycline/pharmacology , Phosphorylation , Rats, Sprague-Dawley , Restraint, Physical/psychology , Spatial Learning , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Stress, Psychological/psychology , Time Factors
2.
Toxicol Lett ; 232(1): 301-9, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25445583

ABSTRACT

Arsenic is well established as a human carcinogen, but the molecular mechanisms leading to arsenic-induced carcinogenesis are complex and elusive. It is not been determined if the epithelial-mesenchymal transition (EMT) contributes to carcinogen-induced malignant transformation and subsequent tumor formation. We have found that, during the neoplastic transformation induced in human bronchial epithelial (HBE) cells by a low concentration (1.0µM) of arsenite, the cells undergo an EMT and show enhanced invasion and migration. With longer times for transformation of HBE cells, there was increased miR-21 expression. Further, during the transformation of HBE cells, inhibition of miR-21 with an miR-21 inhibitor increased levels of PDCD4, an inhibitor of neoplastic transformation; reduced Twist1, a transcription factor involved in cell differentiation; and inhibited cell invasion and migration. In addition, PDCD4 interacted with Twist1 and inhibited its expression function, which is involved in arsenite-induced EMT. Thus, miR-21, acting on PDCD4, which interacts with Twist1 and represses the expression of Twist1, contributes to the EMT induced by arsenite. These observations add to an understanding of the processes involved in arsenite-induced carcinogenesis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Arsenites/toxicity , Bronchi/drug effects , Cell Movement/drug effects , Cell Transformation, Neoplastic/chemically induced , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Lung Neoplasms/chemically induced , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , Sodium Compounds/toxicity , Apoptosis Regulatory Proteins/genetics , Bronchi/metabolism , Bronchi/pathology , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MicroRNAs/genetics , Nuclear Proteins/metabolism , RNA Interference , RNA-Binding Proteins/genetics , Signal Transduction/drug effects , Time Factors , Transfection , Twist-Related Protein 1/metabolism , Up-Regulation
3.
Toxicol Lett ; 229(1): 158-66, 2014 Aug 17.
Article in English | MEDLINE | ID: mdl-24960059

ABSTRACT

miRNAs have been found to contribute to normal brain functions, nervous system diseases, as well as neurotoxicities induced by external agents. However, whether they are involved in lead-induced neurotoxicities is still not clear. To identify that, a lead-induced chronic neurotoxicity model of rats was built. Both miRNA microarray analysis and qRT-PCR were performed to determine the change of miRNA expression in hippocampus. Then 3 bioinformatics databases were used to analyze the relative target genes of these miRNA, which were further confirmed by qRT-PCR and Western blot. In the present study, lead exposure resulted in the changed expression of 7 miRNAs: miR-204, miR-211, miR-448, miR-449a, miR-34b, and miR-34c were greatly up-regulated while miR-494 was greatly down-regulated. Bioinformatics analysis results showed that the target genes of 6 up-regulated miRNAs were related to neural injury and neurodegeration, axon and synapse function, neural development and regeneration. Correspondingly, the expression levels of mature mRNAs and proteins of three target genes (Bcl-2, Itpr1, and Map2k1) were greatly repressed, verifying the results of bioinformatics analysis. Taken together, our results showed that the expression of several miRNAs reported to be associated with neurophysiological pathways and neurodegenerative diseases changed in rat hippocampus following chronic lead exposure. These miRNAs may play important roles in lead-induced neurotoxicity.


Subject(s)
Hippocampus/drug effects , Hippocampus/metabolism , Lead/toxicity , MicroRNAs/biosynthesis , Animals , Blotting, Western , Body Weight/drug effects , Computational Biology , Drinking/drug effects , Lead/blood , Male , Maze Learning/drug effects , Microarray Analysis , Nerve Regeneration/drug effects , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/genetics , Rats , Rats, Sprague-Dawley , Synapses/drug effects
4.
Toxicol Lett ; 225(1): 110-8, 2014 Feb 10.
Article in English | MEDLINE | ID: mdl-24316150

ABSTRACT

The blood-cerebrospinal fluid barrier (BCB) plays a key role in maintaining copper (Cu) homeostasis in the brain. Cumulative evidences indicate that lead (Pb) exposure alters cerebral Cu homeostasis, which may underlie the development of neurodegenerative diseases. This study investigated the roles of Cu transporter 1 (CTR1) and ATP7A, two Cu transporters, in Pb-induced Cu accumulation in the choroidal epithelial cells. Pb exposure resulted in increased intracellular (64)Cu retention, accompanying with up-regulated CTR1 level. Knockdown of CTR1 using siRNA before Pb exposure diminished the Pb-induced increase of (64)Cu uptake. The expression level of ATP7A was down-regulated following the Pb exposure. ATP7A siRNA knockdown, or PCMB treatment, inhibited the (64)Cu efflux from the cells, while the following additional incubation with Pb failed to further increase the intracellular (64)Cu retention. Cu exposure, or intracellular Cu accumulation following the tetracycline (Tet)-induced overexpression of CTR1, did not result in significant change in ATP7A expression. Taken together, these data indicate that CTR1 and ATP7A play important roles in Cu transport in choroidal epithelial cells, and the Pb-induced intracellular Cu accumulation appears to be mediated, at least in part, via the alteration of CTR1 and ATP7A expression levels following Pb exposure.


Subject(s)
Adenosine Triphosphatases/metabolism , Cation Transport Proteins/drug effects , Cation Transport Proteins/metabolism , Choroid Plexus/drug effects , Copper/metabolism , Epithelial Cells/drug effects , Organometallic Compounds/toxicity , Adenosine Triphosphatases/genetics , Animals , Biological Transport , Cation Transport Proteins/genetics , Cell Line , Choroid Plexus/metabolism , Copper Transporter 1 , Copper-Transporting ATPases , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Gene Expression Regulation , Homeostasis , RNA Interference , RNA, Messenger/metabolism , Rats , Transfection
5.
Neurotoxicology ; 39: 173-81, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24144481

ABSTRACT

This study was designed to investigate the impact of lead (Pb(2+)) on the auditory system and its molecular mechanisms. Pb(AC)2 was administrated to male SD rats aged 21-22 d for 8 weeks at a dose of 300ppm. Male guinea pigs were also administrated with 50mg/kg Pb(AC)2 two times a week for 8 weeks. The auditory nerve-brainstem evoked responses (ABR) was recorded and the morphological changes of the outer hair cells (OHCs) were observed with Phallodin-FITC staining. In addition, the integrity of the blood-labyrinth barrier was observed by TEM and the expression of tight junction proteins (TJPs) in the cochlear stria vascularis was determined by immunofluorescence. Our results showed that Pb(2+) exposure resulted in increased ABR threshold in both rats and guinea pigs. Abnormal shapes and loss of OHCs were found in the cochlear basilar membrane following the Pb(2+) exposure. TEM study showed that the tight junctions between the endothelial cells and the border cells were lost and disrupted. Down-regulation of the occludin, ZO-1 and claudin-5 in the stria vascularis suggested that the increased permeability of the blood-labyrinth barrier may attribute to the Pb(2+)-induced decrease of TJPs' expression. Additionally, Fe(2+) supplement partly reversed the Pb(2+)-induced hearing loss and down-regulation of TJPs. Taken together, these data indicate that the disruption of blood-labyrinth barrier by down-regulating the expression of TJPs plays a role in the Pb(2+)-induced hearing loss, and Fe(2+) supplement protects the auditory system against Pb(2+)-induced toxicity and may have significant clinical implications.


Subject(s)
Cochlea/drug effects , Hearing Loss/chemically induced , Organometallic Compounds/toxicity , Animals , Animals, Newborn , Claudins/metabolism , Cochlea/blood supply , Dietary Supplements , Disease Models, Animal , Down-Regulation/drug effects , Electroencephalography , Evoked Potentials, Auditory, Brain Stem/drug effects , Guinea Pigs , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/ultrastructure , Male , Microscopy, Electron, Transmission , Occludin/metabolism , Organometallic Compounds/blood , Rats , Rats, Sprague-Dawley , Zonula Occludens-1 Protein/metabolism
6.
Neurotoxicology ; 36: 82-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23529067

ABSTRACT

The effect of lead (Pb) on spatial memory and hippocampal long-term potentiation (LTP) as a key risk factor has been widely recognized and the oxidative damage has been proposed as a possible mechanism of lead neurotoxicity. Selenium (Se) is a nutritionally essential trace element with known antioxidant potential. In this study we investigated the effect and the underlying mechanisms of Se supplementary on Pb induced cognition and synaptic plasticity impairment. Lactating Sprague-Dawley rats (SD rats) were randomly divided to four groups: 0ppm lead acetate (Pb); 0ppm Pb and 0.2ppm sodium selenite (Se); 100ppm Pb; 100ppm Pb and 0.2ppm Se. Lactating rats were treated with or without Pb and/or Se throughout lactation until weaning. The levels of hippocampal LTP, the spatial memory, the apoptosis of hippocampal neurons, the levels of lactate dehydrogenase (LDH) release, and the serum level of superoxide dismutase (SOD) and malondialdehyde (MDA) were assayed. It had been observed that in Pb group the spatial memory, the induce level of LTP, the serum SOD level decreased, the LDH release level, the neurons apoptosis level, the serum MDA level increased, while in the Se supplements groups, the spatial memory, the induce level of LTP increased significantly. Compared with the Pb group, Se supplements shown down regulated the level of LDH, the neurons apoptosis and the serum MDA, and up regulated the level of serum SOD. We could draw the conclusion that Se supplements could alleviate toxic effect of lead on hippocampal LTP and spatial memory. The treated with selenium around 0.2ppm may protect against spatial memory dysfunction induced by lead exposure.


Subject(s)
Antioxidants/therapeutic use , Cognition Disorders/chemically induced , Cognition Disorders/drug therapy , Lead/toxicity , Selenic Acid/therapeutic use , Analysis of Variance , Animals , Cognition Disorders/blood , Dose-Response Relationship, Drug , Electric Stimulation , Escape Reaction/drug effects , Excitatory Postsynaptic Potentials/drug effects , Female , In Situ Nick-End Labeling , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Lead/blood , Long-Term Potentiation/drug effects , Male , Malondialdehyde/metabolism , Maze Learning/drug effects , Patch-Clamp Techniques , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/physiopathology , Rats, Sprague-Dawley , Reaction Time/drug effects , Superoxide Dismutase/metabolism
7.
Neurotoxicology ; 33(5): 1230-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22824511

ABSTRACT

Many epidemiological studies and in vitro experiments have found that chronic arsenic exposure may influence memory formation. The goal of this study was to create an animal model of memory impairment induced by chronic arsenite exposure and to study the underlying mechanisms. Sixty male Sprague-Dawley (SD) male rats were randomly divided into a control group, a low-dose sodium arsenite exposure group and a high-dose sodium arsenite exposure group. Sodium arsenite was administered by adding it to drinking water for 3 months. Then, the spatial memory of the rats was examined with Morris water maze and Y maze. The concentration of arsenic in the blood and the brain was determined by an atomic fluorescence absorption spectrometer. The ultra-structure of hippocampal neurons was observed by an electron microscope. Timm staining was used for observing mossy fibers. We found that the concentration of arsenic in the blood and the brain increased in a dose-response manner (P<0.05). The performance of rats in the arsenite exposed group (15 mg/kg) was significantly impaired in the Morris water maze and Y maze tasks than those in the control group (P<0.05). Sodium arsenite exposure resulted in abnormal structural changes in the myelin sheaths of nerve fibers and decreases in the terminals of mossy fibers. Together, chronic sodium arsenite exposure through drinking water results in detrimental changes in the neuronal synapses, which may contribute to the arsenite-induced impairment of spatial memory.


Subject(s)
Arsenic Poisoning/complications , Hippocampus/pathology , Memory Disorders/etiology , Memory Disorders/pathology , Neurons/pathology , Synapses/pathology , Analysis of Variance , Animals , Arsenic Poisoning/etiology , Arsenites/metabolism , Arsenites/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Maze Learning/drug effects , Microscopy, Electron, Transmission , Mossy Fibers, Hippocampal/pathology , Mossy Fibers, Hippocampal/ultrastructure , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Spatial Behavior/drug effects , Synapses/drug effects , Synapses/ultrastructure , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...