Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Neurotoxicology ; 81: 395-405, 2020 12.
Article in English | MEDLINE | ID: mdl-33080273

ABSTRACT

Lead (Pb) is an environmental contaminant that primarily affects the central nervous system, particularly the developing brain. Recently, increasing evidence indicates the important roles of histone deacetylases (HDACs) in Pb-induced neurotoxicity. However, the precise molecular mechanisms involving HDAC4 remains unknown. The purpose of this study was to investigate the role of HDAC4 in Pb-induced neurotoxicity both in vivo and in vitro. In vitro study, PC12 cells were exposed to Pb (10 µM) for 24 h, then the mRNA and protein levels of HDAC4 were analyzed. In vivo study, pregnant rats and their female offspring were treated with lead (50 ppm) until postnatal day 30. Then the pups were sacrificed and the mRNA and protein levels of HDAC4 in the hippocampus were analyzed. The results showed that HDAC4 was significantly increased in both PC12 cells and rat hippocampus upon Pb exposure. Blockade of HDAC4 with either LMK-235 (an inhibitor of HDAC4) or shHDAC4 (HDAC4-knocking down plasmid) ameliorated the Pb-induced neurite outgrowth deficits. Interestingly, HDAC4 was aberrantly accumulated in the nucleus upon Pb exposure. By contrast, blocking the HDAC4 shuffling from the cytosol to the nucleus with ΔNLS2-HDAC4 (the cytosol-localized HDAC4 mutant) was able to rescue the neuronal impairment. In addition, Pb increased PP1 (protein phosphatase 1) expression which in turn influenced the subcellular localization of HDAC4 by dephosphorylation of specific serine/threonine residues. What's more, blockade of PP1 with PP1-knocking down construct (shPP1) ameliorated Pb-induced neurite outgrowth deficits. Taken together, nuclear accumulation of HDAC4 by PP1-mediated dephosphorylation involved in Pb-induced neurotoxicity. This study might provide a promising molecular target for medical intervention with environmental cues.


Subject(s)
Hippocampus/drug effects , Histone Deacetylases/metabolism , Lead Poisoning, Nervous System/enzymology , Neuronal Outgrowth/drug effects , Neurons/drug effects , Organometallic Compounds/toxicity , Protein Phosphatase 1/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Cell Nucleus/pathology , Female , Hippocampus/enzymology , Hippocampus/pathology , Histone Deacetylases/genetics , Lead Poisoning, Nervous System/genetics , Lead Poisoning, Nervous System/pathology , Neurons/enzymology , Neurons/pathology , PC12 Cells , Phosphorylation , Protein Phosphatase 1/genetics , Rats , Rats, Sprague-Dawley
2.
Toxicology ; 416: 23-29, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30738087

ABSTRACT

Lead (Pb) is a widespread environmental heavy metal toxicant and chronic Pb exposure can have irreversible effects on memory and cognitive function, which is closely related to dendritic spines. Studies have shown that SNX6 and Homer1 can regulate the growth of dendritic spines. We aimed to investigate the effect of Pb exposure on the dendritic spines in hippocampus, the expression of SNX6 and Homer1 in rats and PC12 cells. The animals were randomly divided to three groups: control group, low lead group and high lead group. PC12 cells were divided into 3 groups: 0 µM, 1 µM and 100 µM Pb acetate. The results showed that the Pb levels in blood and hippocampus of all exposure groups were significantly higher than that of the control group. The morphology of dendritic spines in hippocampus after Pb treatment was changed and the density of dendritic spines was reduced. The expression of SNX6 and Homer1 was decreased in Pb exposed groups compared with the control group. Furthermore, up-regulation of SNX6 expression could reverse the down-regulation of Pb exposure on Homer1. These results indicate that Pb exposure can reduce the expression of SNX6 and lead to a decrease in Homer1 expression, which affects the changes in dendritic spines causing learning and memory impairment.


Subject(s)
Hippocampus/drug effects , Homer Scaffolding Proteins/metabolism , Lead Poisoning, Nervous System/etiology , Organometallic Compounds/toxicity , Sorting Nexins/metabolism , Animals , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Dendritic Spines/pathology , Down-Regulation , Hippocampus/metabolism , Hippocampus/pathology , Homer Scaffolding Proteins/genetics , Lead Poisoning, Nervous System/genetics , Lead Poisoning, Nervous System/metabolism , Lead Poisoning, Nervous System/pathology , Male , PC12 Cells , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Rats , Rats, Sprague-Dawley , Sorting Nexins/genetics
3.
Toxicol Sci ; 162(2): 688-701, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29301062

ABSTRACT

Lead (Pb) prevails among the environmental hazards against human health. Although increasing evidence highlights the epigenetic roles underlying the Pb-induced neurotoxicity, the exact mechanisms concerning histone acetylation and its causative agents are still at its infancy. In the present study, the roles of histone deacetylases 1 and 2 (HDAC1/2), as well as acetylation of Lys9 on histone H3 (Ac-H3K9), in Pb-induced neurotoxicity were investigated. Pb was administered to PC12 cells at 10 µM for 24 h. And Sprague Dawley rats were chronically exposed to Pb through drinking water containing 250 ppm Pb for 2 months. Owing to Pb exposure, it indicated that HDAC2 was up-regulated accompanied by Ac-H3K9 down-regulation. Meanwhile, chromatin immunoprecipitation assay revealed that the changes in HDAC2 were attributed to histone H3 Lys27 trimethylation occupancy on its promoter. Blockade of HDAC2 with either Trichostatin A or HDAC2-knocking down construct (shHDAC2) resulted in amelioration of neurite outgrowth deficits via increasing Ac-H3K9 levels. It implied that HDAC2 plays essential regulatory roles in Pb-induced neurotoxicity. And, coimmunoprecipitation trials revealed that HDAC2 colocalized with HDAC1, forming a so-called HDAC1/2 complex. Subsequently, it was shown that HDAC1/2 repression could markedly prevent neurite outgrowth impairment and rescue the spatial memory deficits caused by Pb exposure, unequivocally implicating this complex in the studied toxicological process. Furthermore, Notch2 maybe the functional target of the HDAC1/2 and Ac-H3K9 alterations. Our study provided insight into the precise roles of HDAC1/2 in Pb-induced neurotoxicity, and thereby provided a promising molecular target for medical intervention of neurological disorders with environmental etiology.


Subject(s)
Environmental Pollutants/toxicity , Gene Expression Regulation, Enzymologic/drug effects , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Lead Poisoning, Nervous System/enzymology , Lead/toxicity , Animals , Cell Culture Techniques , Gene Knockdown Techniques , Lead Poisoning, Nervous System/genetics , Lead Poisoning, Nervous System/pathology , Neuronal Outgrowth/drug effects , Neuronal Outgrowth/genetics , PC12 Cells , Rats , Rats, Sprague-Dawley , Up-Regulation
4.
J Appl Toxicol ; 37(4): 400-407, 2017 04.
Article in English | MEDLINE | ID: mdl-27535807

ABSTRACT

Developmental lead (Pb) exposure is suggested in laboratory studies to be a trigger for neurodegenerative diseases such as Alzheimer's disease (AD). Sortilin-related receptor, L (DLR class) A repeats-containing (SORL1) is a recently identified AD genetic risk factor. SORL1 has limited characterization in vertebrate models in comparison to other AD genetic risk factors. To characterize SORL1 further, protein sequence homology between humans, mice and zebrafish was analyzed and showed conservation of functional repeats and domain orientation. Next, spatial expression of sorl1 in zebrafish larvae was completed and diffuse expression in neural tissue that was not restricted to the brain was observed. Influences of sex and age on quantitative expression of sorl1 in the brain of adult zebrafish were then assessed. Sex-specific alteration of sorl1 expression transpired during the aging process in females. The zebrafish was then utilized to investigate the impacts of a 100 ppb embryonic Pb exposure on sorl1 expression and other known AD genetic risk factors. Sex-specific quantitative gene expression analysis was completed with adult zebrafish brain to compare those developmentally exposed to Pb or a control treatment, but no significant difference in sorl1 expression or other AD genetic risk factors was observed. Overall, this study provided characterization of sorl1 with changes in brain expression during aging being female-specific. This finding is in agreement with females being more prone to the onset of AD, but analysis of additional AD genetic risk factors is needed to facilitate our understanding of the impact of a 100 ppb embryonic Pb exposure. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Alzheimer Disease/chemically induced , Alzheimer Disease/genetics , Embryo, Nonmammalian/pathology , LDL-Receptor Related Proteins/genetics , Lead Poisoning, Nervous System/genetics , Zebrafish Proteins/genetics , Aging/pathology , Alzheimer Disease/pathology , Animals , Brain/pathology , Female , Lead Poisoning, Nervous System/pathology , Male , Protein Sorting Signals/genetics , Risk Factors , Sex Characteristics , Zebrafish
5.
Toxicol Ind Health ; 33(4): 332-339, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27230353

ABSTRACT

Lead acts as an antagonist of the N-methyl-d-aspartate receptor (NMDAR). GRIN2A encodes an important subunit of NMDARs and may be a critical factor in the mechanism of lead neurotoxicity. Changes in GRIN2A expression levels or gene variants may be mechanisms of lead-induced neurotoxicity. In this study, we hypothesized that GRIN2A might contribute to lead-induced neurotoxicity. A preliminary HEK293 cell experiment was performed to analyze the association between GRIN2A expression and lead exposure. In addition, in a population-based study, serum GRIN2A levels were measured in both lead-exposed and control populations. To detect further the influence of GRIN2A gene single nucleotide polymorphisms (SNPs) in lead-induced neurotoxicity, 3 tag SNPs (rs2650429, rs6497540, and rs9302415) were genotyped in a case-control study that included 399 lead-exposed subjects and 398 controls. Lead exposure decreased GRIN2A expression levels in HEK293 cells ( p < 0.001) compared with lead-free cells. Lead-exposed individuals had lower serum GRIN2A levels compared with controls ( p < 0.001), and we found a trend of decreasing GRIN2A level with an increase in blood lead level ( p < 0.001). In addition, we found a significant association between rs2650429 CT and TT genotypes and risk of lead poisoning compared with the rs2650429 CC genotype (adjusted odds ratio = 1.42, 95% confidence interval = 1.01-2.00]. Therefore, changes in GRIN2A expression levels and variants may be important mechanisms in the development of lead-induced neurotoxicity.


Subject(s)
Gene Expression Regulation/drug effects , Genetic Predisposition to Disease , Lead Poisoning, Nervous System/metabolism , Lead/toxicity , Occupational Diseases/metabolism , Polymorphism, Single Nucleotide , Receptors, N-Methyl-D-Aspartate/metabolism , Adult , Alleles , Case-Control Studies , China , Environmental Pollutants/toxicity , Female , Gene Frequency , Genetic Association Studies , HEK293 Cells , Humans , Lead/blood , Lead Poisoning, Nervous System/blood , Lead Poisoning, Nervous System/genetics , Male , Middle Aged , Occupational Diseases/blood , Occupational Diseases/genetics , Occupational Exposure/adverse effects , RNA, Messenger/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/blood , Receptors, N-Methyl-D-Aspartate/genetics , Young Adult
6.
Aging (Albany NY) ; 7(9): 629-47, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26363853

ABSTRACT

Due to its role in regulation of mitochondrial function, PGC1α is emerging as an important player in ageing and neurodegenerative disorders. PGC1α exerts its neuroprotective effects by promoting mitochondrial biogenesis (MB) and functioning. However, the precise regulatory role of PGC1α in the control of mitochondrial dynamics (MD) and neurotoxicity is still unknown. Here we elucidate the role of PGC1αin vitro and in vivo in the regulatory context of MB and MD in response to lead (II) acetate as a relevant model of neurotoxicity. We show that there is an adaptive response (AR) to lead, orchestrated by the BAP31-calcium signalling system operating between the ER and mitochondria. We find that this hormetic response is controlled by a cell-tolerated increase of PGC1α expression, which in turn induces a balanced expression of fusion/fission genes by binding to their promoters and implying its direct role in regulation of MD. However, dysregulation of PGC1α expression through either stable downregulation or overexpression, renders cells more susceptible to lead insult leading to mitochondrial fragmentation and cell death. Our data provide novel evidence that PGC1α expression is a key regulator of MD and the maintenance of tolerated PGC1α expression may offer a promising strategy for neuroprotective therapies.


Subject(s)
Lead Poisoning, Nervous System/genetics , Transcription Factors/genetics , Aging/genetics , Animals , Apoptosis/drug effects , Cell Death/drug effects , Cell Line , Dopaminergic Neurons/drug effects , Endoplasmic Reticulum/drug effects , Lead Poisoning, Nervous System/physiopathology , Membrane Proteins/genetics , Mitochondrial Dynamics/genetics , Neuroprotective Agents/pharmacology , Organelle Biogenesis , Organometallic Compounds/toxicity , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Signal Transduction/drug effects , Transcription Factors/biosynthesis
7.
Article in English | MEDLINE | ID: mdl-25293352

ABSTRACT

Our objective was to examine whether functional polymorphisms in hemochromatosis (HFE; H63D and C282Y), transferrin (TfC2), and glutathione-s-transferase Pi1 (GSTP1; Ile105Val) genes modify any lead-ALS association. We measured blood lead using atomic absorption spectroscopy and bone lead - a biomarker of cumulative lead exposure - using K-shell-X-ray fluorescence in 100 neurologist-confirmed ALS cases and 194 controls, the latter recruited as part of two separate studies; all subjects lived in New England. Participants were considered variant carriers or wild-type for each polymorphism. To assess effect modification, we included cross-product terms between lead biomarkers and each polymorphism in separate adjusted polytomous logistic regression models. Compared with wild-type, the odds ratio (OR) per 15.6 µg/g patella lead (interquartile range; IQR) was 8.24 (95% CI 0.94-72.19) times greater among C282Y variant carriers, and 0.34 (95% CI 0.15-0.78) times smaller among H63D variant carriers. Results were weaker for tibia lead. Compared with wild-type the OR per 2 µg/dl blood lead (IQR) was 0.36 (95% CI 0.19-0.68) times smaller among H63D variant carriers, and 1.96 (95% CI 0.98-3.92) times greater among GSTP1 variant carriers. In conclusion, we found that HFE and GSTP1 genotypes modified the association between lead biomarkers and ALS. Contrasting modification by the HFE polymorphisms H63D and C282Y may suggest that the modification is not simply the result of increased iron.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Iron , Lead Poisoning, Nervous System/genetics , Oxidative Stress/genetics , Polymorphism, Genetic/genetics , Adult , Aged , Amyotrophic Lateral Sclerosis/epidemiology , Bone and Bones/metabolism , Female , Genetic Association Studies , Genotype , Glutathione S-Transferase pi/genetics , Hemochromatosis/genetics , Humans , Lead/metabolism , Lead Poisoning, Nervous System/epidemiology , Male , Middle Aged , Spectrophotometry, Atomic , Transferrin/genetics
8.
Toxicol Mech Methods ; 25(2): 128-35, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25496477

ABSTRACT

In recent years, the use of stem cells as a new tool to create an in vitro model for toxicological studies has been considered. Adipose tissue-derived stem cells (ADSCs) are mesenchymal stem cells which have been extracted from adipose tissue by a less invasive method and rapidly propagated in culture medium compared with other sources. These cells have the capacity to differentiate into different cell lineage in vitro including neural cells. The aim of this study was to investigate the effect of lead exposure at various stages of differentiation on the neural differentiation of ADSCs. Third-passaged ADSCs were differentiated to neural cell in differentiation medium during 16 d. The ADSCs were exposed to lead (0.1-100 µg/ml) before differentiation and during differentiation on days 1, 7 and 14. The cell viability was assessed by MTT assay after 48 h. Also expression of ß-tubulin III protein and Nestin, NeuN, NF70, Synaptophysin genes were evaluated at the end of differentiation in all treated groups. The results showed that lead had no effect on viability of undifferentiated ADSCs but differentiating cells showed various sensitivities to lead exposure and cells were more vulnerable to lead exposure at early stage of differentiation. Also, lead exposure at different stages of differentiation had various effects on gene expressions. Our study indicated that neural cells differentiated from ADSCs in vitro are sensitive to neurotoxic effect of lead as well-known developmental neurotoxicant, and then ADSCs could be a candidate as an alternative method for assessing neurodevelopmental toxicity potential of chemicals.


Subject(s)
Adipose Tissue/cytology , Lead Poisoning, Nervous System , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Organometallic Compounds/toxicity , Animals , Cell Separation/methods , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Gene Expression Regulation, Developmental/drug effects , Lead Poisoning, Nervous System/genetics , Lead Poisoning, Nervous System/metabolism , Lead Poisoning, Nervous System/pathology , Male , Mice, Inbred BALB C , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Time Factors
9.
Toxicology ; 325: 67-73, 2014 Nov 05.
Article in English | MEDLINE | ID: mdl-25193092

ABSTRACT

Epidemiologic studies have provided solid evidence for the neurotoxic effect of lead for decades of years. In view of the fact that children are more vulnerable to the neurotoxicity of lead, lead exposure has been an urgent public health concern. The modes of action of lead neurotoxic effects include disturbance of neurotransmitter storage and release, damage of mitochondria, as well as induction of apoptosis in neurons, cerebrovascular endothelial cells, astroglia and oligodendroglia. Our studies here, from a novel point of view, demonstrates that lead specifically caused induction of COX-2, a well known inflammatory mediator in neurons and glia cells. Furthermore, we revealed that COX-2 was induced by lead in a transcription-dependent manner, which relayed on transcription factor NFAT, rather than AP-1 and NFκB, in glial cells. Considering the important functions of COX-2 in mediation of inflammation reaction and oxidative stress, our studies here provide a mechanistic insight into the understanding of lead-associated inflammatory neurotoxicity effect via activation of pro-inflammatory NFAT3/COX-2 axis.


Subject(s)
Cyclooxygenase 2/biosynthesis , Lead Poisoning, Nervous System/etiology , Lead/toxicity , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Neuroglia/drug effects , Transcription Factor AP-1/metabolism , Animals , Cyclooxygenase 2/genetics , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Enzyme Induction , Inflammation Mediators/metabolism , Lead Poisoning, Nervous System/enzymology , Lead Poisoning, Nervous System/genetics , Mice , NFATC Transcription Factors/genetics , Neural Stem Cells/drug effects , Neural Stem Cells/enzymology , Neuroglia/enzymology , Neurons/drug effects , Neurons/enzymology , PC12 Cells , RNA, Messenger/biosynthesis , Rats , Signal Transduction/drug effects , Time Factors , Transcription, Genetic/drug effects , Transfection , Up-Regulation
10.
Neurotoxicology ; 33(6): 1450-1453, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22743688

ABSTRACT

The progressive and latent nature of neurodegenerative diseases, such as Alzheimer's disease (AD) indicates the role of epigenetic modification in disease susceptibility. Previous studies from our lab show that developmental exposure to lead (Pb) perturbs the expression of AD-associated proteins. In order to better understand the role of DNA methylation as an epigenetic modifications mechanism in gene expression regulation, an integrative study of global gene expression and methylation profiles is essential. Given the different formats of gene expression and methylation data, combining these data for integrative analysis can be challenging. In this paper we describe a method to integrate and analyze gene expression and methylation arrays. Methylation array raw data contain the signal intensities of each probe of CpG sites, whereas gene expression data measure the signal intensity values of genes. In order to combine these data, methylation data of CpG sites have to be associated with genes.


Subject(s)
Aging/genetics , Alzheimer Disease/genetics , DNA Methylation , Lead Poisoning, Nervous System/genetics , Systems Biology , Systems Integration , Animals , CpG Islands , Disease Models, Animal , Gene Expression Profiling/methods , Gene Expression Regulation , Genetic Predisposition to Disease , Genome-Wide Association Study , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis
11.
Neurotoxicology ; 33(3): 280-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22342836

ABSTRACT

The role of green tea in protection against neurotoxicity induced by lead acetate was investigated in rats. Five equal groups, each of ten rats were used. The first group was served as control, the second, third, and fourth groups were given lead acetate, lead acetate and green tea, and green tea only, respectively, for one month, the fifth group was administered lead acetate for one month followed by green tea for 15 days. Lead acetate was given orally at a dose of 100 mg/kg b. wt, while green tea was given in drinking water at a concentration of 5 g/L. Lead acetate administration induced loss of body weight and decreased concentration of reduced glutathione and SOD activity in brain tissues as well as significantly high DNA fragmentation and pathological changes. Co-administration of green tea with lead acetate significantly alleviated these adverse effects.


Subject(s)
Antioxidants/pharmacology , Brain/drug effects , DNA Damage , Lead Poisoning, Nervous System/prevention & control , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Tea , Animals , Antioxidants/isolation & purification , Brain/metabolism , Brain/pathology , Cytoprotection , DNA Fragmentation , Disease Models, Animal , Glutathione/metabolism , Lead Poisoning, Nervous System/etiology , Lead Poisoning, Nervous System/genetics , Lead Poisoning, Nervous System/metabolism , Lead Poisoning, Nervous System/pathology , Male , Neuroprotective Agents/isolation & purification , Organ Size/drug effects , Organometallic Compounds , Plant Extracts/isolation & purification , Rats , Superoxide Dismutase/metabolism , Tea/chemistry , Time Factors , Weight Loss/drug effects
12.
J Mol Neurosci ; 47(1): 76-88, 2012 May.
Article in English | MEDLINE | ID: mdl-22160880

ABSTRACT

Although developmental lead exposure is known to have detrimental effects on a variety of cognitive functions that depend on the integrity of the hippocampus and frontal cortex, little is known about how low levels of lead exposure affect expression of key families of genes in these structures. The present study examined the effects of exposure to environmentally relevant levels of lead during the sensitive early post-weaning period in the rat on the expression profiles of a select number of neurobiologically relevant genes (i.e., genes for neurotrophic factors, NMDA receptors, metabotropic glutamate receptors, synaptic function/plasticity, cell signaling, and transcription/regulation) in the rat hippocampus and frontal cortex. Exposure to lead (180 and 375-ppm lead acetate in food for 30 days) significantly increased blood lead levels (5.8 to 10.3 µg/dl) and significantly affected expression of many of the genes examined. In many instances, lead exposure had different effects on the same gene depending on the brain region in which the expression of that gene was examined. Gene expression in the frontal cortex was often more sensitive to modification than gene expression in the hippocampus. These results suggest that even past infancy, exposures to low levels of lead can have significant effects on gene expression in the frontal cortex and the hippocampus with the potential to exert long-term effects on behavior and cognition.


Subject(s)
Aging/genetics , Frontal Lobe/drug effects , Gene Expression Regulation, Developmental/drug effects , Hippocampus/drug effects , Lead Poisoning, Nervous System/genetics , Lead/toxicity , Aging/drug effects , Aging/pathology , Animals , Cognition Disorders/chemically induced , Cognition Disorders/genetics , Cognition Disorders/pathology , Frontal Lobe/growth & development , Frontal Lobe/pathology , Hippocampus/growth & development , Hippocampus/pathology , Lead/blood , Lead Poisoning, Nervous System/pathology , Male , Rats , Rats, Long-Evans
13.
Neurotoxicology ; 33(3): 560-74, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22178136

ABSTRACT

Schizophrenia is a devastating neuropsychiatric disorder of unknown etiology. There is general agreement in the scientific community that schizophrenia is a disorder of neurodevelopmental origin in which both genes and environmental factors come together to produce a schizophrenia phenotype later in life. The challenging questions have been which genes and what environmental factors? Although there is evidence that different chromosome loci and several genes impart susceptibility for schizophrenia; and epidemiological studies point to broad aspects of the environment, only recently there has been an interest in studying gene × environment interactions. Recent evidence of a potential association between prenatal lead (Pb(2+)) exposure and schizophrenia precipitated the search for plausible neurobiological connections. The most promising connection is that in schizophrenia and in developmental Pb(2+) exposure there is strong evidence for hypoactivity of the N-methyl-d-aspartate (NMDA) subtype of excitatory amino acid receptors as an underlying neurobiological mechanism in both conditions. A hypofunction of the NMDA receptor (NMDAR) complex during critical periods of development may alter neurobiological processes that are essential for brain growth and wiring, synaptic plasticity and cognitive and behavioral outcomes associated with schizophrenia. We also describe on-going proof of concept gene-environment interaction studies of early life Pb(2+) exposure in mice expressing the human mutant form of the disrupted in schizophrenia 1 (DISC-1) gene, a gene that is strongly associated with schizophrenia and allied mental disorders.


Subject(s)
Environmental Exposure , Environmental Pollutants/adverse effects , Lead Poisoning, Nervous System/etiology , Lead/adverse effects , Neurons/drug effects , Schizophrenia/chemically induced , Age Factors , Animals , Apoptosis/drug effects , Dopamine/metabolism , Gene-Environment Interaction , Glutamine/metabolism , Humans , Lead Poisoning, Nervous System/genetics , Lead Poisoning, Nervous System/metabolism , Lead Poisoning, Nervous System/pathology , Lead Poisoning, Nervous System/psychology , Mutation , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/pathology , Receptors, N-Methyl-D-Aspartate/metabolism , Risk Assessment , Risk Factors , Schizophrenia/genetics , Schizophrenia/metabolism , Schizophrenia/pathology , Schizophrenic Psychology , gamma-Aminobutyric Acid/metabolism
14.
Neurotoxicol Teratol ; 33(6): 715-20, 2011.
Article in English | MEDLINE | ID: mdl-21839828

ABSTRACT

Previous studies have reported that environmental lead (Pb) exposure can result in neurological alterations in children leading to reduced IQ, attention deficit hyperactivity disorder, and diminished reading and learning abilities. However, the specific alterations in neurodevelopmental morphology and the underlying genetic mechanisms of these alterations have not yet been thoroughly defined. To investigate alterations in neurologic morphology and test the hypothesis that developmental Pb neurotoxicity is partially mediated through alterations in neuronal growth and transport function of axons, the changes of specific axon tracts in the embryonic zebrafish brain were observed with anti-acetylated α-tubulin staining at several developmental time points through 36hours post fertilization (hpf). In addition, the role of a subset of axonogenesis-related genes including shha, epha4b, netrin1b, netrin2, and noiwas investigated with real-time quantitative PCR (qPCR). Pb treatment resulted in decreased axonal density at 18, 20, and 24hpf for specific axon tracts in the midbrain and forebrain. These observations corresponded to an observed down-regulation of shha and epha4b at 14 and 16hpf, respectively. The axonal density in Pb exposed individuals at later stages (30 and 36hpf) was not significantly different from controls. An overexpression of netrin2 at these two developmental stages suggests a novel role for this gene in regulating axonal density specific to Pb neurotoxicity. Although no significant differences in axonal density was observed in the two later developmental stages, further studies are needed to determine if the morphologic alterations observed at the earlier stages will have lasting functional impacts.


Subject(s)
Axonal Transport/drug effects , Axons/drug effects , Embryo, Nonmammalian/drug effects , Gene Expression Regulation, Developmental/drug effects , Lead Poisoning, Nervous System/embryology , Zebrafish/embryology , Animals , Axonal Transport/genetics , Axons/pathology , Brain/drug effects , Brain/embryology , Disease Models, Animal , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Gene Expression Profiling , Lead Poisoning, Nervous System/genetics , Real-Time Polymerase Chain Reaction , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
15.
Toxicol Appl Pharmacol ; 256(2): 179-90, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21864555

ABSTRACT

The influence of sex as an effect modifier of childhood lead poisoning has received little systematic attention. Considering the paucity of information available concerning the interactive effects of lead and sex on the brain, the current study examined the interactive effects of lead and sex on gene expression patterns in the hippocampus, a structure involved in learning and memory. Male or female rats were fed either 1500 ppm lead-containing chow or control chow for 30 days beginning at weaning.Blood lead levels were 26.7±2.1 µg/dl and 27.1±1.7 µg/dl for females and males, respectively. The expression of 175 unique genes was differentially regulated between control male and female rats. A total of 167 unique genes were differentially expressed in response to lead in either males or females. Lead exposure had a significant effect without a significant difference between male and female responses in 77 of these genes. In another set of 71 genes, there were significant differences in male vs. female response. A third set of 30 genes was differentially expressed in opposite directions in males vs. females, with the majority of genes expressed at a lower level in females than in males. Highly differentially expressed genes in males and females following lead exposure were associated with diverse biological pathways and functions. These results show that a brief exposure to lead produced significant changes in expression of a variety of genes in the hippocampus and that the response of the brain to a given lead exposure may vary depending on sex.


Subject(s)
Gene Expression/drug effects , Hippocampus/drug effects , Lead Poisoning, Nervous System/genetics , Animals , Animals, Newborn/metabolism , Female , Gene Expression Profiling , Hippocampus/metabolism , Male , Oligonucleotide Array Sequence Analysis , Rats , Rats, Long-Evans , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors
16.
Neurotoxicol Teratol ; 32(2): 262-72, 2010.
Article in English | MEDLINE | ID: mdl-20006704

ABSTRACT

The relationship between the blood lead concentration and cognitive function in children and adults with different VDR genotypes who participated in the third National Health and Nutrition Examination Survey was investigated. The relationship between blood lead and serum homocysteine concentrations was also investigated. In children 12 to 16 years old, performance on the digit span and arithmetic tests as a function of the blood lead concentration varied by VDR rs2239185 and VDR rs731236 genotypes. Decreases in performance occurred in some genotypes, but not in others. In adults 20 to 59 years old, performance on the symbol-digit substitution test as a function of the blood lead concentration varied by VDR rs2239185-rs731236 haplotype. In the 12 to 16 year old children and adults 60 or more years old, the relationship between the serum homocysteine and blood lead concentrations varied by VDR genotype. The mean blood lead concentrations of the children and adults did not vary by VDR genotype.


Subject(s)
Cognition Disorders/chemically induced , Cognition Disorders/genetics , Genetic Predisposition to Disease/genetics , Lead Poisoning, Nervous System/genetics , Lead Poisoning, Nervous System/physiopathology , Receptors, Calcitriol/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Child , Cognition/drug effects , Cognition/physiology , Cognition Disorders/metabolism , DNA Mutational Analysis , Female , Genetic Testing , Genotype , Health Status , Health Surveys , Homocysteine/analysis , Homocysteine/blood , Humans , Lead/adverse effects , Lead/analysis , Lead/blood , Lead Poisoning, Nervous System/metabolism , Male , Middle Aged , Neuropsychological Tests , Polymorphism, Single Nucleotide/genetics , Young Adult
17.
Neurochem Res ; 35(3): 473-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19911273

ABSTRACT

Lead (Pb) exposure poses devastating effects on central nervous system development of children. To replicate aspects of this neurotoxicity, we examined the effect of lead on the expression of apoptosis and of apoptosis-related genes, XIAP (X chromosome-linked inhibitor of apoptosis protein) and Smac (second mitochondrial activator of caspase), in the hippocampus of developing rats. A total of 48 rats (30-day old) were randomly divided into four groups for intragastrical perfusion of lead acetate [Pb(Ac)2]: untreated, low (2 mg/kg/d), medium (20 mg/kg/d), and high (200 mg/kg/d) dose groups. Pb content was determined in blood, and the apoptosis indexes and XIAP and Smac gene expression were analyzed in the hippocampus. There was a significant difference in apoptosis indexes (AI) between the exposed and control groups (p < 0.01). AI was highest in the high exposure group. XIAP gene expression was reduced in the exposed groups and the expression was negatively correlated with blood lead levels (BLLs) (p < 0.05). But the four groups did not differ in the expression of Smac (p > 0.05). Our data indicate that exposure to Pb(Ac)2 caused a dose-dependent and significant increase of apoptosis in the hippocampus of developing rats through depressing the expression of the XIAP but not the Smac genes.


Subject(s)
Apoptosis/drug effects , Carrier Proteins/genetics , Hippocampus/metabolism , Hippocampus/pathology , Lead Poisoning, Nervous System/genetics , Lead Poisoning, Nervous System/pathology , Mitochondrial Proteins/genetics , Organometallic Compounds/toxicity , X-Linked Inhibitor of Apoptosis Protein/genetics , Animals , Apoptosis Regulatory Proteins , Dose-Response Relationship, Drug , Gene Expression/drug effects , Hippocampus/growth & development , In Situ Nick-End Labeling , Male , Organometallic Compounds/blood , Organometallic Compounds/pharmacokinetics , RNA/biosynthesis , RNA/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
18.
Rev Environ Health ; 24(1): 15-45, 2009.
Article in English | MEDLINE | ID: mdl-19476290

ABSTRACT

Lead, a systemic toxicant affecting virtually every organ system, primarily affects the central nervous system, particularly the developing brain. Consequently, children are at a greater risk than adults of suffering from the neurotoxic effects of lead. To date, no safe lead-exposure threshold has been identified. The ability of lead to pass through the blood-brain barrier is due in large part to its ability to substitute for calcium ions. Within the brain, lead-induced damage in the prefrontal cerebral cortex, hippocampus, and cerebellum can lead to a variety of neurologic disorders. At the molecular level, lead interferes with the regulatory action of calcium on cell functions and disrupts many intracellular biological activities. Experimental studies have also shown that lead exposure may have genotoxic effects, especially in the brain, bone marrow, liver, and lung cells. Knowledge of the neurotoxicology of lead has advanced in recent decades due to new information on its toxic mechanisms and cellular specificity. This paper presents an overview, updated to January 2009, of the neurotoxic effects of lead with regard to children, adults, and experimental animals at both cellular and molecular levels, and discusses the biomarkers of lead exposure that are useful for risk assessment in the field of environmental health.


Subject(s)
Lead Poisoning, Nervous System , Lead/adverse effects , Animals , Biomarkers , Humans , Lead/blood , Lead/toxicity , Lead Poisoning, Nervous System/blood , Lead Poisoning, Nervous System/epidemiology , Lead Poisoning, Nervous System/genetics , Lead Poisoning, Nervous System/physiopathology , Neurotoxicity Syndromes/blood , Neurotoxicity Syndromes/epidemiology , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/physiopathology , Sister Chromatid Exchange
19.
J Mol Neurosci ; 34(1): 1-7, 2008.
Article in English | MEDLINE | ID: mdl-18157652

ABSTRACT

Alzheimer's Disease (AD) is a progressive, irreversible neurodegenerative disease. Despite several genetic mutations (Haass et al., J. Biol. Chem. 269:17741-17748, 1994; Ancolio et al., Proc. Natl. Acad. Sci. USA 96:4119-4124, 1999; Munoz and Feldman, CMAJ 162:65-72, 2000; Gatz et al., Neurobiol. Aging 26:439-447, 2005) found in AD patients, more than 90% of AD cases are sporadic (Bertram and Tanzi, Hum. Mol. Genet. 13:R135-R141, 2004). Therefore, it is plausible that environmental exposure may be an etiologic factor in the pathogenesis of AD. The AD brain is characterized by extracellular beta-amyloid (Abeta) deposition and intracellular hyperphosphorylated tau protein. Our lab has demonstrated that developmental exposure of rodents to the heavy metal lead (Pb) increases APP (amyloid precursor protein) and Abeta production later in the aging brain (Basha et al., J. Neurosci. 25:823-829, 2005a). We also found elevations in the oxidative marker 8-oxo-dG in older animals that had been developmentally exposed to Pb (Bolin et al., FASEB J. 20:788-790, 2006) as well as promotion of amyloidogenic histopathology in primates. These findings indicate that early life experiences contribute to amyloidogenesis in old age perhaps through epigenetic pathways. Here we explore the role of epigenetics as the underlying mechanism that mediates this early exposure-latent pathogenesis with a special emphasis on alterations in the methylation profiles of CpG dinucleotides in the promoters of genes and their influence on both gene transcription and oxidative DNA damage.


Subject(s)
Alzheimer Disease/genetics , Environmental Exposure , Epigenesis, Genetic/genetics , Plaque, Amyloid/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/biosynthesis , Animals , DNA Damage/drug effects , DNA Damage/genetics , DNA Methylation/drug effects , Female , Humans , Lead Poisoning, Nervous System/complications , Lead Poisoning, Nervous System/genetics , Lead Poisoning, Nervous System/metabolism , Plaque, Amyloid/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...