Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 187
Filter
1.
Child Dev ; 94(6): e308-e327, 2023.
Article in English | MEDLINE | ID: mdl-37307305

ABSTRACT

Developmental science has increasingly scrutinized how environmental hazards influence child outcomes, but few studies examine how contaminants affect disparities in early skill formation. Linking research on environmental inequality and early childhood development, this study assessed whether differences in exposure to neurotoxic lead explain sociodemographic gaps in school readiness. Using panel data tracking a representative sample of 1266 Chicago children (50% female, 16% White, 30% Black, 49% Hispanic, µage = 5.2 months at baseline, collected 1994-2002), analyses quantified the contribution of lead contamination to class and racial disparities in vocabulary skills and attention problems at ages 4 and 5. Results suggested that lead contamination explains 15%-25% and 33%-66% of the disparities in each outcome, respectively, although imprecise estimates preclude drawing firm inferences about attention problems.


Subject(s)
Child Development , Environmental Exposure , Hispanic or Latino , Lead Poisoning, Nervous System , Racial Groups , Child , Child, Preschool , Female , Humans , Infant , Male , Hispanic or Latino/psychology , Hispanic or Latino/statistics & numerical data , Racial Groups/ethnology , Racial Groups/psychology , Racial Groups/statistics & numerical data , Schools , Environmental Exposure/adverse effects , Environmental Exposure/statistics & numerical data , Lead Poisoning, Nervous System/complications , Lead Poisoning, Nervous System/epidemiology , Lead Poisoning, Nervous System/ethnology , Lead Poisoning, Nervous System/psychology
2.
Toxicol Ind Health ; 38(10): 655-664, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35838060

ABSTRACT

The developing brain is susceptible to the neurotoxic effects of lead. Exposure to lead has main effects on the cholinergic system and causes reduction of cholinergic neuron function during brain development. Disruption of the cholinergic system by chemicals, which play important roles during brain development, causes of neurodevelopmental toxicity. Differentiation of stem cells to neural cells is recently considered a promising tool for neurodevelopmental toxicity studies. This study evaluated the toxicity of lead acetate exposure during the differentiation of bone marrow-derived mesenchyme stem cells (bone marrow stem cells, BMSCs) to CCholinergic neurons. Following institutional animal care review board approval, BMSCs were obtained from adult rats. The differentiating protocol included two stages that were pre-induction with ß-mercaptoethanol (BME) for 24 h and differentiation to cholinergic neurons with nerve growth factor (NGF) over 5 days. The cells were exposed to different lead acetate concentrations (0.1-100 µm) during three stages, including undifferentiated, pre-induction, and neuronal differentiation stages; cell viability was measured by MTT assay. Lead exposure (0.01-100 µg/ml) had no cytotoxic effect on BMSCs but could significantly reduce cell viability at 50 and 100 µm concentrations during pre-induction and neuronal differentiation stages. MAP2 and choline acetyltransferase (ChAT) protein expression were investigated by immunocytochemistry. Although cells treated with 100 µm lead concentration expressed MAP2 protein in the differentiation stages, they had no neuronal cell morphology. The ChAT expression was negative in cells treated with lead. The present study showed that differentiated neuronal BMSCs are sensitive to lead toxicity during differentiation, and it is suggested that these cells be used to study neurodevelopmental toxicity.


Subject(s)
Lead Poisoning, Nervous System , Mesenchymal Stem Cells , Animals , Bone Marrow , Bone Marrow Cells , Cells, Cultured , Choline O-Acetyltransferase/metabolism , Choline O-Acetyltransferase/pharmacology , Cholinergic Agents/metabolism , Cholinergic Agents/pharmacology , Lead/metabolism , Lead Poisoning, Nervous System/metabolism , Mercaptoethanol/metabolism , Mercaptoethanol/pharmacology , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , Organometallic Compounds , Rats
3.
Toxicology ; 472: 153179, 2022 04 30.
Article in English | MEDLINE | ID: mdl-35429622

ABSTRACT

Toxic effects of lead (Pb) are principally manifested in the central nervous system (CNS) and a mounting body of evidence indicates that excessive chronic exposure to Pb participates in the pathological processes of numerous neurodegenerative disorders in humans.In this study we evaluated whether the prolonged pre- and postnatal exposure of rat pups to lead, administrated through ingestion in drinking water, as a typical environmental exposure, can determine alterations of the protein pattern of CNS myelin and the induction of myelin-associated proteinases. Pregnant dams were given distilled water or 0.3 mg/mL lead acetate in drinking water during gestation and lactation. At postnatal day (PND) 21, pups born from mothers poisoned with Pb continued the treatment with the metal. On PND 35 and 56, pups were sacrificed, and brains were subjected to myelin purification and extraction of myelin-associated proteinases. The SDS-PAGE analysis of protein pattern of myelin incubated in vitro with an oxidative system indicated that myelin proteins from Pb-treated pups were more sensitive to the toxicity of reactive oxygen species in comparison with those from untreated pups. The zymografic analysis of NaCl-extracts from myelin of Pb-treated pups showed a band of digestion of 54 kDa that increased in pups sacrificed at PND 56 in comparison with those sacrificed at PND 35 and correlated with the concentration of Pb, detected in purified myelin. The incubation of the NaCl-extract from Pb-treated pups with purified myelin basic protein (MBP) evidenced the presence of different MBP-degrading activities. These results suggest that Pb may influence the integrity of the myelin sheath, probably through the induction of anti-myelin proteinases.


Subject(s)
Drinking Water , Lead Poisoning, Nervous System , Prenatal Exposure Delayed Effects , Animals , Animals, Newborn , Female , Humans , Lead/toxicity , Myelin Sheath , Peptide Hydrolases , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Rats , Sodium Chloride/toxicity
4.
Molecules ; 27(3)2022 Feb 01.
Article in English | MEDLINE | ID: mdl-35164259

ABSTRACT

The use of natural products as therapeutic agents is rapidly growing recently. In the current study, we investigated the protective effects of green tea supplementation on lead-induced toxicity in mice. Forty albino mice were divided into four groups as follows: A: control group; B: green tea receiving group; C: lead-intoxicated group; and D: lead-intoxicated group supplemented with green tea. At the end of the experiment, the animals were tested for neurobehavioral and biochemical alterations. Green tea was analyzed through Gas Chromatography-Mass Spectrometry (GC/MS) analysis. We found that supplementation with green tea ameliorated the lead-associated increase in body weight and blood glucose. Green tea supplementation also changed the blood picture that was affected due to lead toxicity and ameliorated lead-induced dyslipidemia. The group of mice that were supplemented with green tea has shown positive alterations in locomotory, anxiety, memory, and learning behaviors. The GC/MS analysis revealed many active ingredients among which the two most abundant were caffeine and 1,2-benzenedicarboxylic acid, mono(2-ethylhexyl) ester. We concluded that green tea supplementation has several positive effects on the lead-induced neurotoxicity in mice and that these effects may be attributed to its main two active ingredients.


Subject(s)
Lead Poisoning, Nervous System/prevention & control , Lead/toxicity , Tea , Animals , Behavior, Animal/drug effects , Blood Glucose/metabolism , Body Weight/drug effects , Brain/metabolism , Dyslipidemias/chemically induced , Dyslipidemias/prevention & control , Gas Chromatography-Mass Spectrometry/methods , Lead/blood , Lead/metabolism , Mice
5.
Neurotoxicology ; 88: 106-115, 2022 01.
Article in English | MEDLINE | ID: mdl-34793780

ABSTRACT

Lead (Pb) is a well-known neurotoxicant and environmental hazard. Recent experimental evidence has linked Pb exposure with neurological deterioration leading to neurodegenerative diseases, such as Alzheimer's disease. To understand brain regional distribution of Pb and its interaction with other metal ions, we used synchrotron micro-x-ray fluorescence technique (µ-XRF) to map the metal distribution pattern and to quantify metal concentrations in mouse brains. Lead-exposed mice received oral gavage of Pb acetate once daily for 4 weeks; the control mice received sodium acetate. Brain tissues were cut into slices and subjected for analysis. Synchrotron µ-XRF scans were run on the PETRA III P06 beamline (DESY). Coarse scans of the entire brain were performed to locate the cortex and hippocampus, after which scans with higher resolution were run in these areas. The results showed that: a) the total Pb intensity in Pb-exposed brain slices was significantly higher than in control brain; b) Pb typically deposited in localized particles of <10 um2 in both the Pb-exposed and control brain slices, with more of these particles in Pb-exposed samples; c) selenium (Se) was significantly correlated with Pb in these particles in the cortex and hippocampus/corpus callosum regions in the Pb-exposed samples, and the molar ratio of the Se and Pb in these particles is close to 1:1. These results indicated that Se may play a crucial role in Pb-induced neurotoxicity. Our findings call for further studies to investigate the relationship between Pb exposure and possible Se detoxification responses, and the implication in the etiology of Alzheimer's disease.


Subject(s)
Brain Chemistry/drug effects , Lead Poisoning, Nervous System/metabolism , Lead/analysis , Selenium/analysis , Animals , Lead/administration & dosage , Male , Mice , Spectrometry, X-Ray Emission , Synchrotrons
6.
Int J Mol Sci ; 22(4)2021 Feb 06.
Article in English | MEDLINE | ID: mdl-33561959

ABSTRACT

Autism Spectrum Disorder (ASD) remains one of the most detrimental neurodevelopmental conditions in society today. Common symptoms include diminished social and communication ability. Investigations on autism etiology remain largely ambiguous. Previous studies have highlighted exposure to lead (Pb) may play a role in ASD. In addition, lead has been shown to be one of the most prevalent metal exposures associated with neurological deficits. A semi-systematic review was conducted using public databases in order to evaluate the extent of lead's role in the etiology of autism. This review examines the relationship between autistic comorbid symptoms-such as deterioration in intelligence scores, memory, language ability, and social interaction-and lead exposure. Specifically, the mechanisms of action of lead exposure, including changes within the cholinergic, dopaminergic, glutamatergic, gamma aminobutyric acid (GABA)ergic systems, are discussed. The goal of this review is to help illustrate the connections between lead's mechanistic interference and the possible furthering of the comorbidities of ASD. Considerations of the current data and trends suggest a potential strong role for lead in ASD.


Subject(s)
Autistic Disorder/chemically induced , Autistic Disorder/pathology , Environmental Exposure/adverse effects , Lead Poisoning, Nervous System/pathology , Lead/toxicity , Autistic Disorder/etiology , Humans , Intelligence/drug effects , Intelligence Tests , Language Development , Memory/drug effects , Social Interaction/drug effects
7.
Neurotoxicol Teratol ; 84: 106948, 2021.
Article in English | MEDLINE | ID: mdl-33418068

ABSTRACT

The morphogenesis of the head of vertebrates is a process that involves rapid growth and dynamic movements of various cell populations, including the neural crest cells (NCC). These pluripotent cells generated during neurulation have high proliferative and migratory capacity but xenobiotic agents can affect these migratory periods and cause congenital malformations. Lead (Pb) is the most common toxic metal in the environment and a potent teratogen that can affect growth and induce malformations. Despite the known toxic effects of Pb, there is a gap in knowledge about the impact of realistic concentrations of Pb at critical periods of early development. Here, we evaluated mortality, embryonic morphology, NCC migration, and the amount of Pb deposition in chicken embryos after 3 to 4 days of exposure. One of the most interesting observations in this study is that only about 34% of the injected Pb was present in the embryos after 4 days. We observed that exposure to Pb, even under low concentrations, increased mortality and the occurrence of malformations during embryonic development, especially in the cephalic region (CR). Although Pb was found widely distributed in the CR, no relation between its presence and the migration routes of cephalic NCC was observed. But the number of NCC and their migratory distance were reduced. These changes are consistent and explain the morphological anomalies described in this study, which also correlates with the morphofunctional abnormalities reported in the literature. Therefore, this study highlights the concern of exposure to low concentrations of this metal.


Subject(s)
Lead Poisoning, Nervous System/pathology , Neural Crest/pathology , Abnormalities, Drug-Induced/pathology , Animals , Biological Availability , Brain/abnormalities , Brain/pathology , Cell Movement , Chick Embryo , Embryonic Development/drug effects , Lead/metabolism , Lead/pharmacokinetics , Lead/toxicity , Lead Poisoning, Nervous System/mortality , Morphogenesis , Nitrates/toxicity
8.
Neurotoxicology ; 82: 119-129, 2021 01.
Article in English | MEDLINE | ID: mdl-33248188

ABSTRACT

Childhood lead (Pb2+) intoxication is a global public health problem best known for producing deficits in learning and poor school performance. Human and preclinical studies have suggested an association between childhood Pb2+ intoxication and proclivity to substance abuse and delinquent behavior. While environmental factors have been implicated in opioid addiction, less is known about the role of exposure to environmental pollutants on the brain opioid system. Opioid receptors are involved in the biological effects of opioids and other drugs of abuse. In this study, we examine the effect of chronic developmental Pb2+ exposure (1500 ppm in the diet) on µ-opioid receptor (MOR) levels in the rat brain using [3H]-d-Ala2-MePhe4-Gly-ol5 enkephalin ([3H]-DAMGO) quantitative receptor autoradiography at different developmental stages (juvenile, early-adolescent, late adolescent and adult) in male and female rats. Our results indicate that chronic developmental Pb2+ exposure increases the levels of [3H]-DAMGO specific binding to MOR in juvenile and early adolescent Pb2+-exposed male and female rat brain with no changes in late-adolescent (PN50) and minor changes in Pb2+-exposed adult male rats (PN120). Specifically, at PN14, Pb2+-exposed males had an increase in MOR binding in the lateral posthalamic nuclei (LPTN), and Pb2+-exposed females had increased MOR binding in LPTN, medial thalamus, and hypothalamus. At PN28, Pb2+-exposed males had increased MOR levels in the striatum, stria medullaris of the thalamus, LPTN, medial thalamus, and basolateral amygdala, while Pb2+-exposed females showed an increase in nucleus accumbens core, LPTN, and medial thalamus. No changes were detected in any brain region of male and female rats at PN50, and at PN120 there was a decrease in MOR binding of Pb2+-exposed males in the medial thalamus. Our findings demonstrate age and gender specific effects of MOR levels in the rat brain as a result of chronic developmental Pb2+ exposure. These results indicate that the major changes in brain MOR levels were during pre-adolescence and early adolescence, a developmental period in which there is higher engagement in reward and drug-seeking behaviors in humans. In summary, we show that chronic exposure to Pb2+, an ubiquitous and well-known environmental contaminant and neurotoxicant, alters MOR levels in brain regions associated with addiction circuits in the adolescent period, these findings have important implications for opioid drug use and abuse.


Subject(s)
Brain Chemistry/drug effects , Lead Poisoning, Nervous System/metabolism , Receptors, Opioid, mu/analysis , Animals , Brain/drug effects , Brain/growth & development , Brain/metabolism , Female , Lead/blood , Lead Poisoning, Nervous System/complications , Male , Rats/growth & development , Rats, Long-Evans , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/metabolism
9.
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
10.
Neurotoxicology ; 81: 80-88, 2020 12.
Article in English | MEDLINE | ID: mdl-32941938

ABSTRACT

Heavy metals are considered to be among the leading environmental factors that trigger amyotrophic lateral sclerosis (ALS). However, no convincing biopathological mechanism and therapeutic clinical implication of such metals in ALS pathogenesis have been established. This is partly attributable to the technical and scientific difficulties in demonstrating a direct and causative role of heavy metals in the onset of ALS in patients. However, a body of epidemiological, clinical and experimental evidences suggest that lead (Pb), more than other metals, could actually play a major role in the onset and progression of ALS. Here, to clarify the nature of the association and the causative role of Pb in ALS, we comprehensively reviewed the scientific literature of the last decade with objective database searches and the methods typically adopted in systematic reviews, critically analysing and summarising the various scientifically sound evidence on the relationship between ALS and Pb. From these tasks, we noted a number of multidisciplinary associations between ALS and Pb, and specifically the importance of occupational exposure to Pb in ALS development and/or progression. We also report the possible involvement of TAR DNA binding protein (TDP-43)-based molecular mechanism in Pb-mediated ALS, although these data rely on a single study, which included both in vitro experiments and an animal model, and are therefore still preliminary. Finally, we briefly examined whether this knowledge could inspire new targeted therapies and policies in the fight against ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Central Nervous System/drug effects , Environmental Pollutants/adverse effects , Lead Poisoning, Nervous System , Lead/adverse effects , Amyotrophic Lateral Sclerosis/diagnosis , Amyotrophic Lateral Sclerosis/epidemiology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Central Nervous System/metabolism , Central Nervous System/pathology , Central Nervous System/physiopathology , DNA-Binding Proteins/metabolism , Disease Progression , Environmental Exposure/adverse effects , Gene-Environment Interaction , Humans , Lead Poisoning, Nervous System/diagnosis , Lead Poisoning, Nervous System/epidemiology , Lead Poisoning, Nervous System/metabolism , Lead Poisoning, Nervous System/physiopathology , Protein Aggregates , Protein Aggregation, Pathological , Risk Assessment , Risk Factors
11.
Toxicology ; 440: 152492, 2020 07.
Article in English | MEDLINE | ID: mdl-32407874

ABSTRACT

Neurotoxicity induced by exposure to heavy metal lead (Pb) is a concern of utmost importance particularly for countries with industrial-based economies. The developing brain is especially sensitive to exposure to even minute quantities of Pb which can alter neurodevelopmental trajectory with irreversible effects on motor, emotive-social and cognitive attributes even into later adulthood. Chemical synapses form the major pathway of inter-neuronal communications and are prime candidates for higher order brain (motor, memory and behavior) functions and determine the resistance/susceptibility for neurological disorders, including neuropsychopathologies. The synaptic pathways and mechanisms underlying Pb-mediated alterations in neuronal signaling and plasticity are not completely understood. Employing a biochemically isolated synaptosomal fraction which is enriched in synaptic terminals and synaptic mitochondria, this study aimed to analyze the alterations in bioenergetic and redox/antioxidant status of cerebellar synapses induced by developmental exposure to Pb (0.2 %). Moreover, we test the efficacy of vitamin C (ascorbate; 500 mg/kg body weight), a neuroprotective and neuromodulatory antioxidant, in mitigation of Pb-induced neuronal deficits. Our results implicate redox and bioenergetic disruptions as an underlying feature of the synaptic dysfunction observed in developmental Pb neurotoxicity, potentially contributing to consequent deficits in motor, behavioral and psychological attributes of the organisms. In addition, we establish ascorbate as a key ingredient for therapeutic approach against Pb induced neurotoxicity, particularly for early-life exposures.


Subject(s)
Antioxidants/therapeutic use , Ascorbic Acid/therapeutic use , Cerebellum/metabolism , Energy Metabolism/drug effects , Lead Poisoning, Nervous System/pathology , Synapses/metabolism , Animals , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Cerebellum/drug effects , Female , Glutathione/metabolism , Lead/blood , Lead Poisoning, Nervous System/psychology , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Organ Size/drug effects , Oxidative Stress/drug effects , Pregnancy , Rats , Rats, Wistar , Synapses/drug effects , Synaptosomes/drug effects , Synaptosomes/metabolism
12.
Article in English | MEDLINE | ID: mdl-32340109

ABSTRACT

Chronic exposure to lead is neurotoxic to the human peripheral sensory system. Variant vitamin D receptor (VDR) genes and polymorphisms of metallothioneins (MTs) are associated with different outcomes following lead toxicity. However, no evidence of a relationship between lead neurotoxicity and polymorphisms has previously been presented. In this study, we investigated the relationship between the polymorphisms of VDR, MT1A, and MT2A genes and lead toxicity following chronic occupational lead exposure. We measured vibration perception thresholds (VPT) and current perception thresholds (CPT) in 181 workers annually for five years. The outcome variables were correlated to the subject's index of long-term lead exposure. Polymorphisms of VDR, MT1A, and MT2A were defined. The potential confounders, including age, sex, height, smoking, alcohol consumption, and working life span, were also collected and analyzed using linear regression. The regression coefficients of some gene polymorphisms were at least 20 times larger than regression coefficients of time-weighted index of cumulative blood lead (TWICL) measures. All regression coefficients of TWICL increased slightly. MT1A rs11640851 (AA/CC) was associated with a statistically significant difference in all neurological outcomes except hand and foot VPT. MT1A rs8052394 was associated with statistically significant differences in hand and foot CPT 2000 Hz. In MT2A rs10636, those with the C allele showed a greater effect on hand CPT than those with the G allele. Among the VDR gene polymorphisms, the Apa rs7975232 (CC/AA) single nucleotide polymorphism was associated with the greatest difference in hand CPT. MT2A rs28366003 appeared to have a neural protective effect, whereas Apa (rs7975232) of VDR and MT2A rs10636 increased the neurotoxicity as measured by CPT in the hands. MT1A rs8052394 had a protective effect on large myelinated nerves. MT1A rs11640851 was associated with susceptibility to neurotoxicity.


Subject(s)
Lead Poisoning, Nervous System/blood , Lead/toxicity , Metallothionein/genetics , Occupational Exposure/statistics & numerical data , Receptors, Calcitriol/genetics , Biomarkers/metabolism , Chemical Industry , Environmental Exposure , Female , Genotype , Humans , Lead/blood , Lead Poisoning, Nervous System/diagnosis , Male , Occupational Diseases/chemically induced , Occupational Diseases/genetics , Occupational Exposure/adverse effects , Occupational Exposure/prevention & control , Polymorphism, Single Nucleotide
13.
Ecotoxicol Environ Saf ; 192: 110297, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32061979

ABSTRACT

The present study was conducted in order to assess the chemical composition of Laurus, its antioxidant activities, and benefit from the Laurus extract effect on neurotoxicity caused by lead acetate (Pb). Chemical profile was assayed by using liquid chromatography coupled with high-resolution mass spectrometry (LC-HR-MS). In this study, 40 male rats were divided into four groups (10 rats per each group): (1) control group, (2) Laurus group: rats treated with 250 mg/kg b. wt. of Laurus leaves extract, (3) Pb group: rats treated with 100 mg/kg b. wt. of lead acetate, (4) Pb + Laurus group: rats treated with 250 mg/kg b. wt. of Laurus leaves extract in addition to lead acetate for 30 days. At the end of experiment, some estimates were calculated from blood samples, brain tissue, and histological examination. The results showed that the extract is highly affluent in total flavonoids, total phenolic, and also has antioxidant activity. The LC-MS appeared a wide range of compounds in the extract. The oxidative stress resulted from exposure to lead acetate has been reported to cause reduction in body and brain weights, levels of RBCs, acetylcholinesterase (AChE), GSH, SOD, and CAT in addition to increase in levels of WBCs and MAD. Moreover, Laurus leaves extract notably lessened the biochemical changes caused by lead acetate in the blood, homogenate, and brain tissue (P < 0.05). The current study indicates the antioxidant activity of Laurus leaves extract and assumes that it has a defensive role against the oxidative damage caused by lead in a rat's brain.


Subject(s)
Antioxidants/therapeutic use , Laurus/chemistry , Lead Poisoning, Nervous System/prevention & control , Plant Extracts/therapeutic use , Animals , Antioxidants/chemistry , Brain/drug effects , Brain/metabolism , Brain/pathology , Flavonoids/chemistry , Lead Poisoning, Nervous System/metabolism , Lead Poisoning, Nervous System/pathology , Male , Oxidative Stress/drug effects , Phenols/chemistry , Plant Extracts/chemistry , Plant Leaves/chemistry , Rats
14.
J Alzheimers Dis ; 73(2): 455-459, 2020.
Article in English | MEDLINE | ID: mdl-31815698

ABSTRACT

Limbic-predominant Age-related TDP-43 Encephalopathy (LATE) is a disease in which the clinical presentation mimics that of Alzheimer's disease. TDP-43 proteinopathy associated with LATE has been identified in more than 20% of autopsies of community-dwelling adults over the age of 80. It is believed to contribute significantly toward tau-negative dementia. Heavy metals such as lead has also been linked to TDP-43 proteinopathy. In particular, lead triggers TDP-43 accumulation and disrupts TDP-43 homeostasis. However, the specific relationship between LATE and lead remains unknown. Before leaded gasoline was phased out during the 1970s and 1980s, average blood lead levels were 15 times what they are today. Thus, each successive birth cohort entering old age has had less cumulative lifeime exposure to lead. Lifetime exposure can be tracked in the tibia bone, where the half-life of lead is many decades. We hypothesize that lead plays a role in the development of LATE. There are two ways to explore the validity of this hypothesis. Generational differences in lead exposure should result in a steady decline in the prevalence of LATE among older adults. We propose the use of tibia bone lead levels be examined in conjunction with brain autopsies from different birth cohorts to examine the link between lead exposure and LATE prevalence, holding age constant. Furthermore, individuals with genetic polymorphisms that confer a greater lead absorption phenotype should display a higher degree of TDP-43 accumulation in autopsies. The results of such studies could provide insight into gene by environment interactions relevant to the development of LATE.


Subject(s)
Dementia/pathology , Lead Poisoning, Nervous System/pathology , Limbic System/pathology , TDP-43 Proteinopathies/chemically induced , TDP-43 Proteinopathies/pathology , Adult , Aged , Aged, 80 and over , Autopsy , DNA-Binding Proteins , Dementia/chemically induced , Environmental Exposure , Female , Half-Life , Homeostasis , Humans , Lead/blood , Male , Middle Aged , TDP-43 Proteinopathies/epidemiology , Tetraethyl Lead/adverse effects , Tetraethyl Lead/blood , Tibia/metabolism
15.
Rev. toxicol ; 37(1): 17-18, 2020. ilus
Article in Spanish | IBECS | ID: ibc-194440

ABSTRACT

La exposición ocupacional al plomo continúa siendo un problema de salud pública que afecta mayoritariamente a trabajadores de países en vías de desarrollo. La exposición crónica produce síntomas similares a otras patologías clínicas motivo por el cual es importante poseer alto grado de sospecha. De acuerdo con los síntomas, signos y valor de plumbemia inicial, será necesario el alejamiento de la fuente y posterior tratamiento quelante con edetato cálcico disódico y/o dimercaprol. Presentamos un caso clínico de paresia radial bilateral por exposición crónica a plomo


Occupational exposure to lead continues to be a public health problem, affecting mainly workers of developing countries. Chronic exposure produces symptoms that can be confused with other clinical pathologies, which is why it is important to have a high degree of suspicion. According to the symptoms, signs and value of initial blood lead concentration, it will be necessary to remove from the source and posterior chelation with edetate calcium disodium and/or dimercaprol. We present a clinical case of bilateral radial paresis due to chronic lead exposure


Subject(s)
Humans , Male , Adult , Lead Poisoning, Nervous System/diagnosis , Occupational Diseases/chemically induced , Occupational Exposure/adverse effects , Paresis/chemically induced
16.
C R Biol ; 342(5-6): 192-198, 2019.
Article in English | MEDLINE | ID: mdl-31474522

ABSTRACT

Exposure to lead is a threat factor for neurodegenerative disorders progress as it could trigger dopaminergic deficiency. We aimed herein to assess the effect of acute lead exposure (25mg/kg B.W i.p.) during three continuous days on the dopaminergic and noradrenergic systems together with locomotor performance in Meriones shawi (M. shawi), then the neuroprotective potential of curcumin-III (30mg/kg B.W) by oral gavage. Pb-exposed M. shawi exhibited increased tyrosine hydroxylase (TH) immunoreactivity in substantia nigra compacta (SNc), ventral tegmental area (VTA), locus coeruleus (LC), and dorsal striatum (DS), unlike the controls. This was correlated with decreased locomotor performance. A noticeable protective effect by co-treatment with curcumin-III was observed; in consequence, TH-immunoreactivity and locomotor disturbance were restored in Pb-treated Meriones. Our data results proved, on the one hand, an evident neurotoxic effect of acute Pb exposure and, on the other hand, a potent therapeutic effect of curcumin-III. Thereby, this compound may be recommended as a neuroprotective molecule for neurodegenerative disorders involving catecholaminergic impairment initiated by metallic elements.


Subject(s)
Corpus Striatum/pathology , Curcumin/analogs & derivatives , Dopaminergic Neurons/drug effects , Lead Poisoning, Nervous System/drug therapy , Lead Poisoning, Nervous System/pathology , Neuroprotective Agents/therapeutic use , Parasympathetic Nervous System/pathology , Substantia Nigra/pathology , Administration, Oral , Animals , Curcumin/therapeutic use , Gerbillinae , Locus Coeruleus/pathology , Male , Movement Disorders/psychology , Ventral Tegmental Area/pathology
17.
Neurotoxicology ; 75: 174-185, 2019 12.
Article in English | MEDLINE | ID: mdl-31550440

ABSTRACT

Developmentally-lead (Pb)-exposed rats showed an enhanced vulnerability to the stimulating and motivational effects of ethanol (EtOH). This is accompanied by differential activity of the brain EtOH-metabolizing enzymes catalase (CAT) and mitochondrial aldehyde dehydrogenase (ALDH2). Based on the theory that brain acetaldehyde accumulation is associated with the reinforcing properties of EtOH, this study sought to determine brain CAT and ALDH2 expression in limbic areas of control and Pb-exposed animals after voluntary EtOH intake. Thirty-five-day-old rats perinatally exposed to 220 ppm Pb were offered with water or increasing EtOH solutions (2-10% v/v) during 28 days until postnatal day (PND) 63. Once intake was stable, the animals were administered: 1) saline (SAL; test days 21-24 or 21-28, as corresponds), or 2) a CAT inhibitor: 3-amine 1, 2, 4-triazole (AT; 250 mg/kg intraperitoneally [i.p.], 5 h before the last eight EtOH intake sessions -test days 21-24 and 25-28), or 3) a CAT booster: 3-nitropropionic acid (3NPA; 20 mg/kg subcutaneously [s.c.], 45 min before the last four EtOH intake sessions -test days 25-28). Two additional groups were centrally-administered cyanamide (CY, an ALDH2 inhibitor, 0.3 mg i.c.v. immediately before the last four EtOH sessions, test days 25-28) or its corresponding vehicle (VEH). Lead exposure increased EtOH intake, an effect potentiated in both groups by 3NPA or CY pretreatments and reduced by AT, albeit selectivity in the Pb group. Catalase abundance in limbic areas parallels these observations in the Pb group, showing higher CAT expression in all areas after EtOH consumption respect to the controls, an effect prevented by AT administration. In contrast, ALDH2 expression was reduced in the Pb animals after EtOH intake, with CY potentiating this effect in all brain areas under study. Based on these results and on previous evidences, we suggest that Pb exposure promotes acetaldehyde accumulation in limbic regions, providing some insights into the mechanism of action that underlies the vulnerability to the excessive EtOH consumption reported in these animals.


Subject(s)
Brain/drug effects , Ethanol/pharmacology , Lead Poisoning, Nervous System/metabolism , Alcohol Drinking/metabolism , Alcohol Drinking/psychology , Aldehyde Dehydrogenase, Mitochondrial/antagonists & inhibitors , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Animals , Brain/enzymology , Brain/metabolism , Catalase/metabolism , Cyanamide/pharmacology , Female , Male , Nitro Compounds/pharmacology , Propionates/pharmacology , Rats , Rats, Wistar
18.
Chemosphere ; 235: 1050-1058, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31561294

ABSTRACT

Lead (Pb) is one of the most toxic heavy metals and has aroused widespread concern as it can cause severe impairments in the developing nervous system. Autophagy has been proposed as an injury factor in Pb-induced neurotoxicity. In this study, we used zebrafish embryo as a model, measured the general toxic effects of Pb, and investigated the effect of Pb exposure on autophagy, and its role in Pb-induced developmental neurotoxicity. Zebrafish embryos were exposed to Pb at concentrations of 0, 0.1, 1 or 10 µM until 4 days post-fertilization. Our data showed that exposure to 10 µM Pb significantly reduced survival rates and impaired locomotor activity. Uptake of Pb was enhanced as the concentration and duration of exposure increased. Inhibition of lysosomal degradation with bafilomycin A1 treatment abolished the suppression of Lc3-II protein expression by Pb. Furthermore, autophagosome formation was inhibited by Pb in the brain. In addition, mRNA expression of beclin1, one of the critical genes in autophagy, were decreased in Pb exposure groups at 72 h post-fertilization. Whole-mount in situ hybridization assay showed that beclin1 gene expression in the brain was reduced by Pb. Rapamycin, an autophagy inducer, partly resolved developmental neurotoxicity induced by Pb exposure. Our results suggest that autophagy plays a protective role in the developmental neurotoxicity of Pb in zebrafish embryos and larvae.


Subject(s)
Autophagy/drug effects , Lead Poisoning, Nervous System/prevention & control , Lead/toxicity , Zebrafish/embryology , Animals , Gene Expression , Larva/drug effects , Water Pollutants, Chemical/toxicity , Zebrafish/metabolism
19.
Article in English | MEDLINE | ID: mdl-31412628

ABSTRACT

Heavy metal exposure, in lead (Pb) particularly, is associated with severe neuronal impairment though oxidative stress mediated by reactive oxygen species, and antioxidants may be used to abolish these adverse effects. This study investigated the potential neuroprotective role of coenzyme Q10 (CoQ10) against lead acetate (PbAc)-induced neurotoxicity. Twenty-eight male Wistar albino rats were divided into four equal groups (n = 7) and treated as follows: the control group was injected with physiological saline (0.9% NaCl); the CoQ10 group was injected with CoQ10 (10 mg/kg); PbAc group was injected with PbAc (20 mg/kg); PbAc + CoQ10 group was injected first with PbAc, and after 1 h with CoQ10. All groups were injected intraperitoneally for seven days. PbAc significantly increased cortical lipid peroxidation, nitrate/nitrite levels, and inducible nitric oxide synthase expression, and decreased glutathione content, superoxide dismutase, catalase, glutathione peroxidase, glutathione reductase activity and mRNA expression, as well as nuclear factor erythroid 2-related factor 2 (Nrf2) and homoxygenase-1 (HO-1) expression. PbAc also promoted the secretion of interleukin-1ß and tumor necrosis factor-α, inhibited interleukin-10 production, triggered the activation of pro-apoptotic proteins, and suppressed anti-apoptotic proteins. Additionally, PbAc increased the cortical levels of serotonin, dopamine, norepinephrine, GABA, and glutamate, and decreased the level of ATP. However, treatment with CoQ10 rescued cortical neurons from PbAc-induced neurotoxicity by restoring the balance between oxidants and antioxidants, activating the Nrf2/HO-1 pathway, suppressing inflammation, inhibiting the apoptotic cascade, and modulating cortical neurotransmission and energy metabolism. Altogether, our findings indicate that CoQ10 has beneficial effects against PbAc-induced neuronal damage through its antioxidant, anti-inflammatory, anti-apoptotic, and neuromodulatory activities.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Lead Poisoning, Nervous System/drug therapy , Organometallic Compounds/adverse effects , Ubiquinone/analogs & derivatives , Animals , Disease Models, Animal , Lead Poisoning, Nervous System/pathology , Male , Organometallic Compounds/administration & dosage , Rats , Rats, Wistar , Ubiquinone/pharmacology
20.
Int J Mol Sci ; 20(4)2019 Feb 25.
Article in English | MEDLINE | ID: mdl-30823541

ABSTRACT

The therapeutic value of inhibiting translation of the amyloid precursor protein (APP) offers the possibility to reduce neurotoxic amyloid formation, particularly in cases of familial Alzheimer's disease (AD) caused by APP gene duplications (Dup⁻APP) and in aging Down syndrome individuals. APP mRNA translation inhibitors such as the anticholinesterase phenserine, and high throughput screened molecules, selectively inhibited the uniquely folded iron-response element (IRE) sequences in the 5'untranslated region (5'UTR) of APP mRNA and this class of drug continues to be tested in a clinical trial as an anti-amyloid treatment for AD. By contrast, in younger age groups, APP expression is not associated with amyloidosis, instead it acts solely as a neuroprotectant while facilitating cellular ferroportin-dependent iron efflux. We have reported that the environmental metallotoxins Lead (Pb) and manganese (Mn) cause neuronal death by interfering with IRE dependent translation of APP and ferritin. The loss of these iron homeostatic neuroprotectants thereby caused an embargo of iron (Fe) export from neurons as associated with excess unstored intracellular iron and the formation of toxic reactive oxidative species (ROS). We propose that APP 5'UTR directed translation activators can be employed therapeutically to protect neurons exposed to high acute Pb and/or Mn exposure. Certainly, high potency APP translation activators, exemplified by the Food and Drug Administration (FDA) pre-approved M1 muscarinic agonist AF102B and high throughput-screened APP 5'UTR translation activators, are available for drug development to treat acute toxicity caused by Pb/Mn exposure to neurons. We conclude that APP translation activators can be predicted to prevent acute metal toxicity to neurons by a mechanism related to the 5'UTR specific yohimbine which binds and targets the canonical IRE RNA stem loop as an H-ferritin translation activator.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Ferritins/genetics , Iron-Regulatory Proteins/genetics , Lead Poisoning, Nervous System/drug therapy , Manganese Poisoning/drug therapy , Muscarinic Agonists/therapeutic use , Quinuclidines/therapeutic use , Response Elements/physiology , Thiophenes/therapeutic use , 5' Untranslated Regions/drug effects , Acute Disease , Alzheimer Disease/metabolism , Animals , Down Syndrome/metabolism , Humans , Iron/metabolism , Mice , Muscarinic Agonists/pharmacology , Neurons/metabolism , Protein Biosynthesis/drug effects , Quinuclidines/pharmacology , RNA, Messenger/genetics , Rats , Thiophenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL