Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Environ Toxicol ; 38(4): 899-913, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36629036

ABSTRACT

Epidemiological studies support an association between air pollution exposure, specifically particulate matter (PM), and neurodegenerative disease. Diesel exhaust (DE) is a principal component of ambient air pollution and a major contributor of PM. Our study aimed to examine whether early-life perinatal DE exposure is sufficient to affect behavioral and biochemical endpoints related to Alzheimer's disease later in life. To achieve this, mice were perinatally exposed (embryonic day 0-postnatal day 21) to DE (250-300 µg/m3 ) or filtered air (FA), and allowed to reach aged status (>18 months). Mice underwent behavioral assessment at 6 and 20 months of age, with tissue collected at 22 months for biochemical endpoints. At 6 months, minimal changes were noted in home-cage behavior of DE treated animals. At 20 months, an alternation deficit was noted with the T-maze, although no difference was seen in the object location task or any home-cage metrics. DE exposure did not alter the expression of Aß42, phosphorylated tau S199, or total tau. However, IBA-1 protein, a microglial activation marker, was significantly higher in DE exposed animals. Further, lipid peroxidation levels were significantly higher in the DE exposed animals compared to FA controls. Cytokine levels were largely unchanged with DE exposure, suggesting a lack of inflammation despite persistent lipid peroxidation. Taken together, the findings of this study support that perinatal exposure alone is sufficient to cause lasting changes in the brain, although the effects appear to be less striking than those previously reported in younger animals, suggesting some effects do not persist over time. These findings are encouraging from a public health standpoint and support the aggressive reduction of DE emissions to reduce lifetime exposure and potentially reduce disease outcome.


Subject(s)
Air Pollutants , Neurodegenerative Diseases , Female , Pregnancy , Mice , Animals , Vehicle Emissions , Brain , Particulate Matter
2.
Curr Protoc ; 1(8): e220, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34370398

ABSTRACT

Motor deficits can significantly affect the completion of daily life activities and have a negative impact on quality of life. Consequently, motor function is an important behavioral endpoint to measure for in vivo pathophysiologic studies in a variety of research areas, such as toxicant exposure, drug development, disease characterization, and transgenic phenotyping. Evaluation of motor function is also critical to the interpretation of cognitive behavioral assays, as many rely on intact motor abilities to derive meaningful data. As such, gait analysis is an important component of behavioral research and can be achieved by manual or video-assisted methods. Manual gait analysis methods, however, are prone to observer bias and are unable to capture many critical parameters. In contrast, automated video-assisted gait analysis can quickly and reliably assess gait and locomotor abnormalities that were previously difficult to collect manually. Here, we describe the evaluation of gait and locomotion in rodents using the automated Noldus CatWalk XT system. We include a step-by-step guide for running an experiment using the CatWalk XT system and discuss theory and considerations when evaluating rodent gait. The protocol and discussion provided here act as a supplemental resource to the manual for this commercially available system and can assist CatWalk users in their experimental design and implementation. © 2021 Wiley Periodicals LLC.


Subject(s)
Quality of Life , Rodentia , Animals , Gait , Gait Analysis , Locomotion
3.
Neurotoxicol Teratol ; 87: 107010, 2021.
Article in English | MEDLINE | ID: mdl-34216730

ABSTRACT

Paraoxonase 2 (PON2) is an intracellular antioxidant enzyme shown to play an important role in mitigating oxidative stress in the brain. Oxidative stress is a common mechanism of toxicity for neurotoxicants and is increasingly implicated in the etiology of multiple neurological diseases. While PON2 deficiency increases oxidative stress in the brain in-vitro, little is known about its effects on behavior in-vivo and what global transcript changes occur from PON2 deficiency. We sought to characterize the effects of PON2 deficiency on behavior in mice, with an emphasis on locomotion, and evaluate transcriptional changes with RNA-Seq. Behavioral endpoints included home-cage behavior (Noldus PhenoTyper), motor coordination (Rotarod) and various gait metrics (Noldus CatWalk). Home-cage behavior analysis showed PON2 deficient mice had increased activity at night compared to wildtype controls and spent more time in the center of the cage, displaying a possible anxiolytic phenotype. PON2 deficient mice had significantly shorter latency to fall when tested on the rotarod, suggesting impaired motor coordination. Minimal gait alterations were observed, with decreased girdle support posture noted as the only significant change in gait with PON2 deficiency. Beyond one home-cage metric, no significant sex-based behavioral differences were found in this study. Finally, A subset of samples were utilized for RNA-Seq analysis, looking at three discrete brain regions: cerebral cortex, striatum, and cerebellum. Highly regional- and sex-specific changes in RNA expression were found when comparing PON2 deficient and wildtype mice, suggesting PON2 may play distinct regional roles in the brain in a sex-specific manner. Taken together, these findings demonstrates that PON2 deficiency significantly alters the brain on both a biochemical and phenotypic level, with a specific impact on motor function. These data have implications for future gene-environment toxicological studies and warrants further investigation of the role of PON2 in the brain.


Subject(s)
Aryldialkylphosphatase/deficiency , Behavior, Animal/physiology , Brain/metabolism , Motor Activity/physiology , Animals , Aryldialkylphosphatase/genetics , Corpus Striatum/metabolism , Female , Male , Mice , Motor Activity/genetics , Oxidative Stress/genetics , Oxidative Stress/physiology , Sex Characteristics
4.
J Neurodev Disord ; 12(1): 41, 2020 12 16.
Article in English | MEDLINE | ID: mdl-33327933

ABSTRACT

BACKGROUND: Exposure to traffic-related air pollution (TRAP) during development and/or in adulthood has been associated in many human studies with both neurodevelopmental and neurodegenerative diseases, such as autism spectrum disorder (ASD) and Alzheimer's disease (AD) or Parkinson's disease (PD). METHODS: In the present study, C57BL/6 J mice were exposed to environmentally relevant levels (250+/-50 µg/m3) of diesel exhaust (DE) or filtered air (FA) during development (E0 to PND21). The expression of several transcription factors relevant for CNS development was assessed on PND3. To address possible mechanistic underpinnings of previously observed behavioral effects of DE exposure, adult neurogenesis in the hippocampus and laminar organization of neurons in the somatosensory cortex were analyzed on PND60. Results were analyzed separately for male and female mice. RESULTS: Developmental DE exposure caused a male-specific upregulation of Pax6, Tbr1, Tbr2, Sp1, and Creb1 on PND3. In contrast, in both males and females, Tbr2+ intermediate progenitor cells in the PND60 hippocampal dentate gyrus were decreased, as an indication of reduced adult neurogenesis. In the somatosensory region of the cerebral cortex, laminar distribution of Trb1, calbindin, and parvalbumin (but not of Ctip2 or Cux1) was altered by developmental DE exposure. CONCLUSIONS: These results provide additional evidence to previous findings indicating the ability of developmental DE exposure to cause biochemical/molecular and behavioral alterations that may be involved in neurodevelopmental disorders such as ASD.


Subject(s)
Autism Spectrum Disorder , Hippocampus , Neurogenesis , Vehicle Emissions , Animals , Cerebral Cortex , Female , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Transcription Factors
5.
Pharmacol Ther ; 210: 107523, 2020 06.
Article in English | MEDLINE | ID: mdl-32165138

ABSTRACT

Recent extensive evidence indicates that air pollution, in addition to causing respiratory and cardiovascular diseases, may also negatively affect the brain and contribute to central nervous system diseases. Air pollution is comprised of ambient particulate matter (PM) of different sizes, gases, organic compounds, and metals. An important contributor to PM is represented by traffic-related air pollution, mostly ascribed to diesel exhaust (DE). Epidemiological and animal studies have shown that exposure to air pollution may be associated with multiple adverse effects on the central nervous system. In addition to a variety of behavioral abnormalities, the most prominent effects caused by air pollution are oxidative stress and neuro-inflammation, which are seen in both humans and animals, and are supported by in vitro studies. Among factors which can affect neurotoxic outcomes, age is considered most relevant. Human and animal studies suggest that air pollution may cause developmental neurotoxicity, and may contribute to the etiology of neurodevelopmental disorders, including autism spectrum disorder. In addition, air pollution exposure has been associated with increased expression of markers of neurodegenerative disease pathologies, such as alpha-synuclein or beta-amyloid, and may thus contribute to the etiopathogenesis of neurodegenerative diseases, particularly Alzheimer's disease and Parkinson's disease.


Subject(s)
Adolescent Development/drug effects , Air Pollutants/adverse effects , Air Pollution/adverse effects , Child Development/drug effects , Environmental Exposure/adverse effects , Nervous System/drug effects , Neurodegenerative Diseases/etiology , Neurotoxicity Syndromes/etiology , Adolescent , Adolescent Behavior/drug effects , Age Factors , Animals , Child , Child Behavior/drug effects , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Nervous System/growth & development , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/physiopathology , Neurodegenerative Diseases/psychology , Neurotoxicity Syndromes/diagnosis , Neurotoxicity Syndromes/physiopathology , Neurotoxicity Syndromes/psychology , Pregnancy , Prenatal Exposure Delayed Effects , Risk Assessment , Risk Factors
6.
Neurochem Int ; 131: 104580, 2019 12.
Article in English | MEDLINE | ID: mdl-31626830

ABSTRACT

Air pollution is an important contributor to the global burden of disease, particularly to respiratory and cardiovascular diseases. In recent years, evidence is accumulating that air pollution may adversely affect the nervous system as shown by human epidemiological studies and by animal models. Age appears to play a relevant role in air pollution-induced neurotoxicity, with growing evidence suggesting that air pollution may contribute to neurodevelopmental and neurodegenerative diseases. Traffic-related air pollution (e.g. diesel exhaust) is an important contributor to urban air pollution, and fine and ultrafine particulate matter (PM) may possibly be its more relevant component. Air pollution is associated with increased oxidative stress and inflammation both in the periphery and in the nervous system, and fine and ultrafine PM can directly access the central nervous system. This short review focuses on the adverse effects of air pollution on the developing brain; it discusses some characteristics that make the developing brain more susceptible to toxic effects, and summarizes the animal and human evidence suggesting that exposure to elevated air pollution is associated with a number of behavioral and biochemical adverse effects. It also discusses more in detail the emerging evidence of an association between perinatal exposure to air pollution and increased risk of autism spectrum disorder. Some of the common mechanisms that may underlie the neurotoxicity and developmental neurotoxicity of air pollution are also discussed. Considering the evidence presented in this review, any policy and legislative effort aimed at reducing air pollution would be protective of children's well-being.


Subject(s)
Air Pollutants/toxicity , Air Pollution/adverse effects , Brain/drug effects , Neurotoxicity Syndromes/psychology , Animals , Autism Spectrum Disorder , Child , Developmental Disabilities/diagnosis , Developmental Disabilities/psychology , Female , Humans , Male , Pregnancy
7.
Brain Behav Immun ; 78: 105-115, 2019 05.
Article in English | MEDLINE | ID: mdl-30668980

ABSTRACT

Several epidemiological studies have shown associations between developmental exposure to traffic-related air pollution and increased risk for autism spectrum disorders (ASD), a spectrum of neurodevelopmental disorders with increasing prevalence rate in the United States. Though animal studies have provided support for these associations, little is known regarding possible underlying mechanisms. In a previous study we found that exposure of C57BL/6J mice of both sexes to environmentally relevant levels (250-300 µg/m3) of diesel exhaust (DE) from embryonic day 0 to postnatal day 21 (E0 to PND21) caused significant changes in all three characteristic behavioral domains of ASD in the offspring. In the present study we investigated a potential mechanistic pathway that may be of relevance for ASD-like changes associated with developmental DE exposure. Using the same DE exposure protocol (250-300 µg/m3 DE from E0 to PND21) several molecular markers were examined in the brains of male and female mice at PND3, 21, and 60. Exposure to DE as above increased levels of interleukin-6 (IL-6) in placenta and in neonatal brain. The JAK2/STAT3 pathway, a target for IL-6, was activated by STAT3 phosphorylation, and the expression of DNA methyltransferase 1 (DNMT1), a STAT3 target gene, was increased in DE-exposed neonatal brain. DNMT1 has been reported to down-regulate expression of reelin (RELN), an extracellular matrix glycoprotein important in regulating the processes of neuronal migration. RELN is considered an important modulator for ASD, since there are several polymorphisms in this gene linked to the disease, and since lower levels of RELN have been reported in brains of ASD patients. We observed decreased RELN expression in brains of the DE-exposed mice at PND3. Since disorganized patches in the prefrontal cortex have been reported in ASD patients and disrupted cortical organization has been found in RELN-deficient mice, we also assessed cortical organization, by labeling cells expressing the lamina-specific-markers RELN and calretinin. In DE-exposed mice we found increased cell density in deeper cortex (lamina layers VI-IV) for cells expressing either RELN or calretinin. These findings demonstrate that developmental DE exposure is associated with subtle disorganization of the cerebral cortex at PND60, and suggest a pathway involving IL-6, STAT3, and DNMT1 leading to downregulation of RELN expression that could be contributing to this long-lasting disruption in cortical laminar organization.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cerebral Cortex/physiopathology , Extracellular Matrix Proteins/metabolism , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Vehicle Emissions/toxicity , Air Pollutants/toxicity , Animals , Brain/metabolism , Calbindin 2 , Cell Adhesion Molecules, Neuronal/genetics , Cerebral Cortex/metabolism , Extracellular Matrix Proteins/genetics , Female , Inhalation Exposure/adverse effects , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Prefrontal Cortex/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Reelin Protein , Serine Endopeptidases/genetics
8.
Neurobiol Learn Mem ; 165: 106780, 2019 11.
Article in English | MEDLINE | ID: mdl-29307548

ABSTRACT

Behavioral neuroscience research incorporates the identical high level of meticulous methodologies and exacting attention to detail as all other scientific disciplines. To achieve maximal rigor and reproducibility of findings, well-trained investigators employ a variety of established best practices. Here we explicate some of the requirements for rigorous experimental design and accurate data analysis in conducting mouse and rat behavioral tests. Novel object recognition is used as an example of a cognitive assay which has been conducted successfully with a range of methods, all based on common principles of appropriate procedures, controls, and statistics. Directors of Rodent Core facilities within Intellectual and Developmental Disabilities Research Centers contribute key aspects of their own novel object recognition protocols, offering insights into essential similarities and less-critical differences. Literature cited in this review article will lead the interested reader to source papers that provide step-by-step protocols which illustrate optimized methods for many standard rodent behavioral assays. Adhering to best practices in behavioral neuroscience will enhance the value of animal models for the multiple goals of understanding biological mechanisms, evaluating consequences of genetic mutations, and discovering efficacious therapeutics.


Subject(s)
Behavioral Research/methods , Mice/psychology , Rats/psychology , Animals , Behavioral Research/standards , Reproducibility of Results , Research Design
9.
Part Fibre Toxicol ; 15(1): 18, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29678176

ABSTRACT

BACKGROUND: Escalating prevalence of autism spectrum disorders (ASD) in recent decades has triggered increasing efforts in understanding roles played by environmental risk factors as a way to address this widespread public health concern. Several epidemiological studies show associations between developmental exposure to traffic-related air pollution and increased ASD risk. In rodent models, a limited number of studies have shown that developmental exposure to ambient ultrafine particulates or diesel exhaust (DE) can result in behavioral phenotypes consistent with mild ASD. We performed a series of experiments to determine whether developmental DE exposure induces ASD-related behaviors in mice. RESULTS: C57Bl/6J mice were exposed from embryonic day 0 to postnatal day 21 to 250-300 µg/m3 DE or filtered air (FA) as control. Mice exposed developmentally to DE exhibited deficits in all three of the hallmark categories of ASD behavior: reduced social interaction in the reciprocal interaction and social preference tests, increased repetitive behavior in the T-maze and marble-burying test, and reduced or altered communication as assessed by measuring isolation-induced ultrasonic vocalizations and responses to social odors. CONCLUSIONS: These findings demonstrate that exposure to traffic-related air pollution, in particular that associated with diesel-fuel combustion, can cause ASD-related behavioral changes in mice, and raise concern about air pollution as a contributor to the onset of ASD in humans.


Subject(s)
Air Pollutants/toxicity , Autistic Disorder/chemically induced , Behavior, Animal/drug effects , Inhalation Exposure/adverse effects , Prenatal Exposure Delayed Effects/chemically induced , Vehicle Emissions/toxicity , Animals , Animals, Newborn , Autistic Disorder/physiopathology , Disease Models, Animal , Female , Gestational Age , Maze Learning/drug effects , Mice, Inbred C57BL , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology
10.
Arch Toxicol ; 92(5): 1815-1829, 2018 05.
Article in English | MEDLINE | ID: mdl-29523932

ABSTRACT

Adult neurogenesis is the process by which neural stem cells give rise to new functional neurons in specific regions of the adult brain, a process that occurs throughout life. Significantly, neurodegenerative and psychiatric disorders present suppressed neurogenesis, activated microglia, and neuroinflammation. Traffic-related air pollution has been shown to adversely affect the central nervous system. As the cardinal effects of air pollution exposure are microglial activation, and ensuing oxidative stress and neuroinflammation, we investigated whether acute exposures to diesel exhaust (DE) would inhibit adult neurogenesis in mice. Mice were exposed for 6 h to DE at a PM2.5 concentration of 250-300 µg/m3, followed by assessment of adult neurogenesis in the hippocampal subgranular zone (SGZ), the subventricular zone (SVZ), and olfactory bulb (OB). DE impaired cellular proliferation in the SGZ and SVZ in males, but not females. DE reduced adult neurogenesis, with male mice showing fewer new neurons in the SGZ, SVZ, and OB, and females showing fewer new neurons only in the OB. To assess whether blocking microglial activation protected against DE-induced suppression of adult hippocampal neurogenesis, male mice were pre-treated with pioglitazone (PGZ) prior to DE exposure. The effects of DE exposure on microglia, as well as neuroinflammation and oxidative stress, were reduced by PGZ. PGZ also antagonized DE-induced suppression of neurogenesis in the SGZ. These results suggest that DE exposure impairs adult neurogenesis in a sex-dependent manner, by a mechanism likely to involve microglia activation and neuroinflammation.


Subject(s)
Brain/drug effects , Neurogenesis/drug effects , Pioglitazone/pharmacology , Vehicle Emissions/toxicity , Animals , Brain/pathology , Cell Proliferation/drug effects , Female , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , Protective Agents/pharmacology , Sex Factors , Toxicity Tests, Acute/methods
11.
Neurotoxicol Teratol ; 64: 8-19, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28916171

ABSTRACT

Domoic acid (DA) is an algal toxin which has been associated with significant neurotoxicity in humans, non-human primates, rodents, and marine mammals. Developmental exposure to DA is believed to result in neurotoxicity that may persist into adulthood. DA is produced by harmful algal blooms of Pseudo-nitzschia, raising concerns about the consumption of contaminated seafood. We evaluated oral exposures to DA during pregnancy in mice. Doses of 0 (vehicle), 1 or 3mg/kg/d of DA were administered by gavage to C57BL/6J mice on gestational days 10 to 17. The offspring were tested for persistent neurobehavioral consequences during early development, adolescence and adulthood. Neurobehavioral tests revealed both dose- and gender-related differences in several neurobehavioral measures, including motor coordination in the rotarod test, behavior in the elevated plus maze, circadian patterns of activity in Phenotyper cages, gait as assessed in the Catwalk, and exploratory activity in the Morris water maze. This study demonstrated significant gender-specific and persistent neurobehavioral effects of repeated prenatal oral exposures to DA at low-dose levels that did not induce toxicity in dams.


Subject(s)
Behavior, Animal/drug effects , Kainic Acid/analogs & derivatives , Neurotoxins/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Administration, Oral , Animals , Conditioning, Classical/drug effects , Fear , Female , Kainic Acid/administration & dosage , Kainic Acid/toxicity , Male , Maternal Exposure , Maze Learning , Mice, Inbred C57BL , Neurotoxins/administration & dosage , Pregnancy , Prenatal Exposure Delayed Effects/psychology , Prepulse Inhibition/drug effects , Reflex, Startle/drug effects , Rotarod Performance Test
12.
Adv Neurobiol ; 18: 85-111, 2017.
Article in English | MEDLINE | ID: mdl-28889264

ABSTRACT

The paraoxonases (PONs) are a three-gene family which includes PON1, PON2, and PON3. PON1 and PON3 are synthesized primarily in the liver and a portion is secreted in the plasma, where they are associated with high-density lipoproteins (HDLs), while PON2 is an intracellular enzyme, expressed in most tissues and organs, including the brain. PON1 received its name from its ability to hydrolyze paraoxon, the active metabolite of the organophosphorus (OP) insecticide parathion, and also more efficiently hydrolyzes the active metabolites of several other OPs. PON2 and PON3 do not have OP-esterase activity, but all PONs are lactonases and are capable of hydrolyzing a variety of lactones, including certain drugs, endogenous compounds, and quorum-sensing signals of pathogenic bacteria. In addition, all PONs exert potent antioxidant effects. PONs play important roles in cardiovascular diseases and other oxidative stress-related diseases, modulate susceptibility to infection, and may provide neuroprotection (PON2). Hence, significant attention has been devoted to their modulation by a variety of dietary, pharmacological, lifestyle, or environmental factors. A number of metals have been shown in in vitro, animal, and human studies to mostly negatively modulate expression of PONs, particularly PON1, the most studied in this regard. In addition, different levels of expression of PONs may affect susceptibility to toxicity and neurotoxicity of metals due to their aforementioned antioxidant properties.


Subject(s)
Aryldialkylphosphatase/drug effects , Heavy Metal Poisoning, Nervous System/metabolism , Metals/pharmacology , Animals , Antioxidants , Aryldialkylphosphatase/metabolism , Cadmium/pharmacology , Cadmium Poisoning/metabolism , Disease Susceptibility , Humans , Lead/pharmacology , Lead Poisoning, Nervous System/metabolism , Lipoproteins, HDL/metabolism , Liver/enzymology , Liver/metabolism , Manganese/pharmacology , Manganese Poisoning , Mercury/pharmacology , Mercury Poisoning, Nervous System/metabolism , Oxidative Stress/drug effects
13.
Curr Protoc Toxicol ; 72: 11.22.1-11.22.21, 2017 May 02.
Article in English | MEDLINE | ID: mdl-28463420

ABSTRACT

Autism spectrum disorder (ASD) represents a heterogeneous group of disorders characterized by alterations in three behavioral symptom domains: Social interactions, verbal and nonverbal communication, and repetitive behaviors. Increasing prevalence of ASD in recent years suggests that exposure to environmental toxicants may be critical in modulating etiology of this disease. As clinical diagnosis of autism still relies on behavioral evaluation, it is important to be able to assess similar behavioral traits in animal models, to provide biological plausibility of associations between environmental exposures and ASD. Rodents naturally exhibit a large number of behaviors that can be linked to similar behaviors in human. In this unit, behavioral tests are described that are relevant to the domains affected in ASD. For the repetitive domain, the T-maze spontaneous alternation test and marble burying test are described. For the communication domain, neonatal ultrasonic vocalization and olfactory habituation test toward social and non-social odor are described. Finally, for the sociability domain, the three-chambered social preference test and the reciprocal interaction test are presented. © 2017 by John Wiley & Sons, Inc.


Subject(s)
Autism Spectrum Disorder/psychology , Behavior, Animal , Animals , Animals, Newborn , Disease Models, Animal , Habituation, Psychophysiologic , Mice , Obsessive-Compulsive Disorder/psychology , Odorants , Phenotype , Smell , Social Behavior , Urine/chemistry , Vocalization, Animal
14.
Curr Environ Health Rep ; 4(2): 156-165, 2017 06.
Article in English | MEDLINE | ID: mdl-28417440

ABSTRACT

PURPOSE OF REVIEW: Epidemiological and animal studies suggest that air pollution may negatively affect the central nervous system (CNS) and contribute to CNS diseases. Traffic-related air pollution is a major contributor to global air pollution, and diesel exhaust (DE) is its most important component. RECENT FINDINGS: Several studies suggest that young individuals may be particularly susceptible to air pollution-induced neurotoxicity and that perinatal exposure may cause or contribute to developmental disabilities and behavioral abnormalities. In particular, a number of recent studies have found associations between exposures to traffic-related air pollution and autism spectrum disorders (ASD), which are characterized by impairment in socialization and in communication and by the presence of repetitive and unusual behaviors. The cause(s) of ASD are unknown, and while it may have a hereditary component, environmental factors are increasingly suspected as playing a pivotal role in its etiology, particularly in genetically susceptible individuals. Autistic children present higher levels of neuroinflammation and systemic inflammation, which are also hallmarks of exposure to traffic-related air pollution. Gene-environment interactions may play a relevant role in determining individual susceptibility to air pollution developmental neurotoxicity. Given the worldwide presence of elevated air pollution, studies on its effects and mechanisms on the developing brain, genetic susceptibility, role in neurodevelopmental disorders, and possible therapeutic interventions are certainly warranted.


Subject(s)
Air Pollution/analysis , Autism Spectrum Disorder/etiology , Neurotoxicity Syndromes , Vehicle Emissions/toxicity , Air Pollutants/analysis , Air Pollutants/toxicity , Air Pollution/adverse effects , Autism Spectrum Disorder/genetics , Central Nervous System , Environmental Exposure/adverse effects , Gene-Environment Interaction , Humans , Inflammation , Reelin Protein , Risk Factors
15.
Neurotoxicology ; 59: 133-139, 2017 03.
Article in English | MEDLINE | ID: mdl-26610921

ABSTRACT

The central nervous system is emerging as an important target for adverse health effects of air pollution, where it may contribute to neurodevelopmental and neurodegenerative disorders. Air pollution comprises several components, including particulate matter (PM) and ultrafine particulate matter (UFPM), gases, organic compounds, and metals. An important source of ambient PM and UFPM is represented by traffic-related air pollution, primarily diesel exhaust (DE). Human epidemiological studies and controlled animal studies have shown that exposure to air pollution, and to traffic-related air pollution or DE in particular, may lead to neurotoxicity. In particular, air pollution is emerging as a possible etiological factor in neurodevelopmental (e.g. autism spectrum disorders) and neurodegenerative (e.g. Alzheimer's disease) disorders. The most prominent effects caused by air pollution in both humans and animals are oxidative stress and neuro-inflammation. Studies in mice acutely exposed to DE (250-300µg/m3 for 6h) have shown microglia activation, increased lipid peroxidation, and neuro-inflammation in various brain regions, particularly the hippocampus and the olfactory bulb. An impairment of adult neurogenesis was also found. In most cases, the effects of DE were more pronounced in male mice, possibly because of lower antioxidant abilities due to lower expression of paraoxonase 2.


Subject(s)
Neurodegenerative Diseases/epidemiology , Neurotoxicity Syndromes/epidemiology , Neurotoxicity Syndromes/etiology , Oxidative Stress/drug effects , Vehicle Emissions/toxicity , Animals , Cytokines/metabolism , Female , Hippocampus/drug effects , Hippocampus/pathology , Humans , International Cooperation , Male , Malondialdehyde/metabolism , Mice , Neurodegenerative Diseases/etiology , Particulate Matter/toxicity
16.
Toxicology ; 374: 1-9, 2016 Dec 30.
Article in English | MEDLINE | ID: mdl-27865893

ABSTRACT

In addition to increased morbidity and mortality caused by respiratory and cardiovascular diseases, air pollution may also contribute to central nervous system (CNS) diseases. Traffic-related air pollution is a major contributor to global air pollution, and diesel exhaust (DE) is its most important component. DE contains more than 40 toxic air pollutants and is a major constituent of ambient particulate matter (PM), particularly of ultrafine-PM. Limited information suggests that exposure to DE may cause oxidative stress and neuroinflammation in the CNS. We hypothesized that males may be more susceptible than females to DE neurotoxicity, because of a lower level of expression of paraoxonase 2 (PON2), an intracellular anti-oxidant and anti-inflammatory enzyme. Acute exposure of C57BL/6 mice to DE (250-300µg/m3 for 6h) caused significant increases in lipid peroxidation and of pro-inflammatory cytokines (IL-1α, IL-1ß, IL-3, IL-6, TNF-α) in various brain regions (particularly olfactory bulb and hippocampus). In a number of cases the observed effects were more pronounced in male than in female mice. DE exposure also caused microglia activation, as measured by increased Iba1 (ionized calcium-binding adapter molecule 1) expression, and of TSPO (translocator protein) binding. Mice heterozygotes for the modifier subunit of glutamate cysteine ligase (the limiting enzyme in glutathione biosynthesis; Gclm+/- mice) appeared to be significantly more susceptible to DE-induced neuroinflammation than wild type mice. These findings indicate that acute exposure to DE causes neuroinflammation and oxidative stress in brain, and suggest that sex and genetic background may play important roles in modulating susceptibility to DE neurotoxicity.


Subject(s)
Air Pollutants/toxicity , Brain Chemistry/drug effects , Brain/pathology , Inflammation/chemically induced , Oxidative Stress/drug effects , Vehicle Emissions/toxicity , Animals , Aryldialkylphosphatase/biosynthesis , Aryldialkylphosphatase/genetics , Cytokines/biosynthesis , Female , Genetic Variation , Glutamate-Cysteine Ligase/biosynthesis , Glutamate-Cysteine Ligase/genetics , Inflammation/genetics , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurotoxicity Syndromes/pathology , Particle Size , Particulate Matter/toxicity , Sex Characteristics
17.
Chem Biol Interact ; 259(Pt B): 168-174, 2016 Nov 25.
Article in English | MEDLINE | ID: mdl-27062895

ABSTRACT

Paraoxonase 2 (PON2) is a member of the paraoxonase gene family also comprising PON1 and PON3. PON2 functions as a lactonase and exhibits anti-bacterial as well as antioxidant properties. At the cellular level, PON2 localizes to the mitochondrial and endoplasmic reticulum membranes where it scavenges reactive oxygen species. PON2 is of particular interest as it is the only paraoxonase expressed in brain tissue and appears to play a critical role in mitigating oxidative stress in the brain. The aim of this study was to investigate the expression of PON2 at the protein and mRNA level in the brain and liver of mice through development to identify potential age windows of susceptibility to oxidative stress, as well as to compare expression of hepatic PON2 to expression of PON1 and PON3. Overall, PON2 expression in the brain was lower in neonatal mice and increased with age up to postnatal day (PND) 21, with a significant decrease observed at PND 30 and 60. In contrast, the liver showed continuously increasing levels of PON2 with age, similar to the patterns of PON1 and PON3. PON2 protein levels were also investigated in brain samples from non-human primates, with PON2 increasing with age up to the infant stage and decreasing at the juvenile stage, mirroring the results observed in the mouse brain. These variable expression levels of PON2 suggest that neonatal and young adult animals may be more susceptible to neurological insult by oxidants due to lower levels of PON2 in the brain.


Subject(s)
Aryldialkylphosphatase/metabolism , Gene Expression Regulation, Developmental/physiology , Animals , Aryldialkylphosphatase/genetics , Brain/metabolism , Female , Haplorhini , Immunoblotting , Liver/metabolism , Male , Mice , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction
18.
Toxicol Sci ; 141(2): 409-22, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25070982

ABSTRACT

Chlorpyrifos oxon (CPO), the toxic metabolite of the organophosphorus (OP) insecticide chlorpyrifos, causes developmental neurotoxicity in humans and rodents. CPO is hydrolyzed by paraoxonase-1 (PON1), with protection determined by PON1 levels and the human Q192R polymorphism. To examine how the Q192R polymorphism influences fetal toxicity associated with gestational CPO exposure, we measured enzyme inhibition and fetal-brain gene expression in wild-type (PON1(+/+)), PON1-knockout (PON1(-/-)), and tgHuPON1R192 and tgHuPON1Q192 transgenic mice. Pregnant mice exposed dermally to 0, 0.50, 0.75, or 0.85 mg/kg/d CPO from gestational day (GD) 6 through 17 were sacrificed on GD18. Biomarkers of CPO exposure inhibited in maternal tissues included brain acetylcholinesterase (AChE), red blood cell acylpeptide hydrolase (APH), and plasma butyrylcholinesterase (BChE) and carboxylesterase (CES). Fetal plasma BChE was inhibited in PON1(-/-) and tgHuPON1Q192, but not PON1(+/+) or tgHuPON1R192 mice. Fetal brain AChE and plasma CES were inhibited in PON1(-/-) mice, but not in other genotypes. Weighted gene co-expression network analysis identified five gene modules based on clustering of the correlations among their fetal-brain expression values, allowing for correlation of module membership with the phenotypic data on enzyme inhibition. One module that correlated highly with maternal brain AChE activity had a large representation of homeobox genes. Gene set enrichment analysis revealed multiple gene sets affected by gestational CPO exposure in tgHuPON1Q192 but not tgHuPON1R192 mice, including gene sets involved in protein export, lipid metabolism, and neurotransmission. These data indicate that maternal PON1 status modulates the effects of repeated gestational CPO exposure on fetal-brain gene expression and on inhibition of both maternal and fetal biomarker enzymes.


Subject(s)
Aryldialkylphosphatase/metabolism , Brain/drug effects , Chlorpyrifos/analogs & derivatives , Insecticides/toxicity , Acetylcholinesterase/metabolism , Animals , Aryldialkylphosphatase/deficiency , Aryldialkylphosphatase/genetics , Brain/enzymology , Butyrylcholinesterase/blood , Carboxylesterase/blood , Chlorpyrifos/toxicity , Erythrocytes/drug effects , Erythrocytes/enzymology , Female , GPI-Linked Proteins/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental/drug effects , Genome-Wide Association Study , Genotype , Gestational Age , Humans , Maternal Exposure , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Peptide Hydrolases/blood , Phenotype , Polymorphism, Genetic , Pregnancy , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction
19.
Biomed Res Int ; 2014: 736385, 2014.
Article in English | MEDLINE | ID: mdl-24524086

ABSTRACT

In addition to increased morbidity and mortality caused by respiratory and cardiovascular diseases, air pollution may also negatively affect the brain and contribute to central nervous system diseases. Air pollution is a mixture comprised of several components, of which ultrafine particulate matter (UFPM; <100 nm) is of much concern, as these particles can enter the circulation and distribute to most organs, including the brain. A major constituent of ambient UFPM is represented by traffic-related air pollution, mostly ascribed to diesel exhaust (DE). Human epidemiological studies and controlled animal studies have shown that exposure to air pollution may lead to neurotoxicity. In addition to a variety of behavioral abnormalities, two prominent effects caused by air pollution are oxidative stress and neuroinflammation, which are seen in both humans and animals and are confirmed by in vitro studies. Among factors which can affect neurotoxic outcomes, age is considered the most relevant. Human and animal studies suggest that air pollution (and DE) may cause developmental neurotoxicity and may contribute to the etiology of neurodevelopmental disorders, including autistic spectrum disorders. In addition, air pollution exposure has been associated with increased expression of markers of neurodegenerative disease pathologies.


Subject(s)
Air Pollutants , Brain/drug effects , Neurotoxins , Air Pollutants/analysis , Air Pollutants/toxicity , Animals , Biomedical Research , Cell Line , Dogs , Humans , Mice , Neurotoxins/analysis , Neurotoxins/toxicity , Rats , Toxicity Tests
20.
Neurotoxicology ; 43: 3-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24012887

ABSTRACT

Paraoxonase 2 (PON2) is a member of a gene family which also includes the more studied PON1, as well as PON3. PON2 is unique among the three PONs, as it is expressed in brain tissue. PON2 is a lactonase and displays anti-oxidant and anti-inflammatory properties. PON2 levels are highest in dopaminergic regions (e.g. striatum), are higher in astrocytes than in neurons, and are higher in brain and peripheral tissues of female mice than male mice. At the sub-cellular level, PON2 localizes primarily in mitochondria, where it scavenges superoxides. Lack of PON2 (as in PON2(-/-) mice), or lower levels of PON2 (as in male mice compared to females) increases susceptibility to oxidative stress-induced toxicity. Estradiol increases PON2 expression in vitro and in vivo, and provides neuroprotection against oxidative stress. Such neuroprotection is not present in CNS cells from PON2(-/-) mice. Similar results are also found with the polyphenol quercetin. PON2, given its cellular localization and antioxidant and anti-inflammatory actions, may represent a relevant enzyme involved in neuroprotection, and may represent a novel target for neuroprotective strategies. Its differential expression in males and females may explain gender differences in the incidence of various diseases, including neurodevelopmental, neurological, and neurodegenerative diseases.


Subject(s)
Antipsychotic Agents/metabolism , Aryldialkylphosphatase/metabolism , Brain/metabolism , Neuroprotective Agents/metabolism , Animals , Antipsychotic Agents/therapeutic use , Aryldialkylphosphatase/genetics , Aryldialkylphosphatase/therapeutic use , Female , Humans , Male , Mice , Mitochondria/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...