Skip to main content
American Journal of Public Health logoLink to American Journal of Public Health
. 2011 Dec;101(Suppl 1):S94–S101. doi: 10.2105/AJPH.2011.300272

Exploring Potential Sources of Differential Vulnerability and Susceptibility in Risk From Environmental Hazards to Expand the Scope of Risk Assessment

Joel Schwartz 1,, David Bellinger 1, Thomas Glass 1
PMCID: PMC3222466  PMID: 22021315

Abstract

Genetic factors, other exposures, individual disease states and allostatic load, psychosocial stress, and socioeconomic position all have the potential to modify the response to environmental exposures. Moreover, many of these modifiers covary with the exposure, leading to much higher risks in some subgroups.

These are not theoretical concerns; rather, all these patterns have already been demonstrated in studies of the effects of lead and air pollution. However, recent regulatory impact assessments for these exposures have generally not incorporated these findings.

Therefore, differential risk and vulnerability is a critically important but neglected area within risk assessment, and should be incorporated in the future.


THIS ARTICLE EXPANDS ON the conceptual issues raised in our previous article in this issue,1 illustrates them with empirical examples, and suggests strategies for incorporating the concepts in risk assessments. The central issue is the importance of capturing variation in the distribution of risk within a population as well as differences between populations in overall risk. This variation exists because of differential susceptibility of people to a single agent, interactions among multiple exposures, transgenerational propagation of risk, and because of differential exposure to other agents that affect the distribution of cumulative risk in the population. Finally, the distribution of exposures to such other agents, or to agents that convey susceptibility to the agent under study, and even to individual level factors that are risk modifiers (e.g., genes, chronic disease states, stress), are usually not independent. Capturing this lack of independence is critical to risk assessment. Finally, incorporating dose–response curves, as opposed to reference doses, is critical to accomplishing these tasks, and to understanding the actual magnitude of risk.

GENETIC SOURCES OF SUSCEPTIBILITY IN RESPONSE TO EXPOSURE

Genetic susceptibility to environmental exposures is well established. As early as the 1970s, studies of people experimentally exposed to ozone in chambers demonstrated substantial variability in response. This variability was repeatable, and unexplained by phenotype.2 Animal studies identified genes with human homologs that might explain this result.3 Common polymorphisms affecting phase I and phase II detoxification pathways are likely sources of important variation in response to multiple toxicants. If genetic susceptibility affects response to an exposure, and several pathways contribute to that susceptibility, substantial differences could result in the distribution of risk, particularly if the prevalence of genetic variants varies by race/ethnicity.

Lead

The role of genetic factors in lead toxicity is unclear at this time. In some studies, carriers of the 2 allele of the amino levulinic acid dehydratase gene (ALAD-2 carriers) were at increased risk of lead-associated neurobehavioral deficits,4,5 whereas others of this subgroup were at reduced risk.6,7 Some of the inconsistency might be a result of age-dependence in this association. A study of lead and cognitive function using National Health and Nutrition Examination Survey (NHANES) data suggested modification by ALAD status in adults, but not in adolescents or adults older than 60 years.6 This highlights the importance of age at exposure, and not exposure alone. A similar age-dependence was reported for apolipoprotein-E (Apo-E). In adults, carriers of the Apo-E4 allele were at increased risk of lead-associated neurobehavioral deficits,7 but not children.8 The impact of Apo-E4 depends on other factors, including stress.9 In males, the adverse effects of lead on executive function were greatest among those lacking the dopamine receptor D4-7.10 Adult workers with the Vitamin D-B variant showed greater lead-associated impairment of renal function.11 Vitamin D polymorphisms modified the effect of lead on cognition in children.12

Lead may increase the risk of neurodegenerative processes by an epigenetic mechanism. In animals, early exposure caused developmental reprogramming, resulting in over expression in adulthood of the amyloid precursor protein (APP) gene, specifically APP mRNA, APP, and β-amyloid.13 In humans, prenatal lead exposure was inversely related to DNA methylation in cord blood,14 and bone lead levels were inversely related to DNA methylation in leukocytes in the elderly.15

Air pollution

Genetic polymorphisms modified the response to air pollution, particularly along the oxidative defense pathway. Such polymorphisms modified the effects of particles on heart rate variability,16,17 the effects of traffic particles on homocysteine levels,18 the effects of traffic pollutants on lengthening of the QT interval on electrocardiograms,19 the effects of air pollution on lung function,20,21 the risk of ozone-induced asthma22 and wheezing,23 and the risk of endothelial inflammation caused by traffic particles, etc. 24 Glutathione S transferase mu 1 (GSTM1) null variant was the most commonly reported modifier along this path, but the other genes along this pathway that matter varied among these studies. This might reflect differences in the outcomes studied, stochastic variability, or interactions with other risk modifiers. Other polymorphisms that might modify the effects of air pollution include those in the divalent metal metabolism pathway,25 the angiotensin pathway,26 the methyl metabolism pathway,27 and genes related to processing of micro RNAs, which are small noncoding RNAs that posttranscriptionally control gene expression.28

Epigenetic mechanisms might serve both as pathways for the effects of air pollution and modifiers of response. Metal-rich particles were associated with reduced methylation of the promoter region of the iNOS gene.29 Traffic officers showed changes in methylation of cancer suppressor and promotion genes similar to those seen in leukemia.30 Exposure to traffic particles31 and polycyclic aromatic hydrocarbons30 altered DNA methylation patterns.

PHENOTYPIC SOURCES OF VARIABLE RESPONSE

In the early 1800s, Scottish public health advocates argued that the (unknown) agents that caused infectious disease were ubiquitous, and what mattered was the person's susceptibility, principally driven by malnutrition. They recommended prescribing food.32 Others also argued that host factors that altered susceptibility were of paramount importance.33 The phenotype might be characterized by disturbances among one or more physiological pathways that were also important to the toxicity of the environmental agent. An environmental disturbance to those pathways might have greater effects if the reserve capacity for dealing with such disturbances was already impaired by the presence of disease or allostatic load (the notion that repeated successful short-term adaptation to stressors might have long-term consequences).34 For example, diabetes and obesity are phenotypes that are characterized by elevated baseline levels of oxidative stress. Coexposure to multiple agents that produce further oxidative stress might result in nonlinear increases in risk. Both lead and air pollution were shown to work, in part, by increasing oxidative stress. Interactions between lead exposure and air pollution were also reported.35 Dietary antioxidants, such as Vitamins C and E, or methyl-related substrates such as B-vitamins or methionine, or N-3 fatty acids36 modified responses to environmental agents.28,37,38 The potential for highly skewed risk distributions existed because these dietary intakes tended to be lower in more disadvantaged areas, where the prevalence of obesity and diabetes also tended to be higher, and where exposure to some environmental chemicals was higher.

Lead

Few data are available on disease states that modify the effects of lead. Lead-associated decrement in renal function might be more pronounced in patients with preexisting chronic kidney disease.39 In an elderly cohort, higher lead level was associated with impaired renal function, but only in diabetic individuals.37 In adult men, the association between higher patella lead and autonomic dysfunction was greater among those with metabolic syndrome.38

Air pollution

Although several conditions appear to modify the effects of air pollution on health, the strongest evidence is for obesity and diabetes. The increasing prevalence of both conditions make these susceptibility factors especially important for risk assessments, which need to take into account the changing proportion of the population that is susceptible. In a study of 4 US cities, patients with diabetes had double the risk of a particulate matter (PM) 10 associated cardiovascular admission compared with nondiabetics40 In Montreal, air pollution was associated with a much higher risk of death from diabetes than for all causes.41 A 2-fold higher mortality risk associated with PM10 exposure was found for patients with diabetes in a case-crossover study.42 Similar results were reported in 9 Italian cities.43 Diabetes also modified the effects of air pollution on endothelial function44 and on systemic inflammation.45,46

Obese individuals were found to have twice the PM2.5-induced reduction in heart rate variability than nonobese individuals and had more PM2.5-mediated heart rate increases.47 Obese individuals had twice the estimated decrease in forced expiratory volume in 1 second because of ozone compared with nonobese subjects.48 This was supported by animal data showing increased lung inflammation in response to ozone in obese animals.49,50 In addition, obesity worsened the PM2.5 effects on the high frequency component of heart rate variability,51 and there was a greater effect of traffic-related PM on inflammatory markers in obese individuals.24,45,52 In the NHANES III, metabolic syndrome modified the PM10 effect on inflammatory markers.53

Diet modifies the effects of air pollution. In a randomized trial, ω-3 fatty acid supplementation reduced the effect of particles on heart rate variability.54 A chamber study of asthmatics found that Vitamin C and E supplementation reduced the increase in bronchial responsiveness after ozone exposure.55

PSYCHOSOCIAL HAZARDS AND STRESS

Psychological stress is a physiological response to environmental stimuli. It can be positive and adaptive, but under certain conditions, including prolonged exposure, can become dysregulated, impairing health.56 Stress partly arises from exposure to a psychosocial hazard, defined as relatively stable, visible features of the social and built environment, that gives rise to a heightened state of vigilance, alarm, and fear.57,58 Dysregulation of the stress response was linked to cardiovascular and other diseases.59 Animal and epidemiological studies suggested that social context modified environmental neurotoxicants.60 Poverty causes stress, and in poor communities, social and chemical hazard exposures in childhood jointly altered the development of the central nervous system.61

Lead

The study of how stress exacerbates the influence of lead dates back to classic studies by Selye et al.,62 who investigated how stressors (both systemic and local) act as “conditioners,” whose individual effects are minor, but when combined with lead exposure, are powerful and complex. Although their work on these “pluricausal” syndromes focused on physiological stressors (skin clip), the concepts were applied to the study of psychological and social stresses.

Stress is consistently observed to exacerbate the deleterious effects of lead. Rodent pups who experienced both early lead exposure and maternal stress during pregnancy (novelty, restraint, cold) showed more impaired learning compared with controls on fixed-interval, schedule-controlled responding, and had increased basal and stress-induced corticosterone responses than did pups exposed to stress alone or to lead alone.63,64 Rats raised in social isolation were more affected by lead than rats raised in an enriched environment.65 Stress increased the hormonal mobilization of lead from bone to blood,66 and lead altered the response to environmental stress.65,67 Exposure to psychosocial hazards increased cortisol production, the primary hormonal mediator of the hypothalamic–pituitary–adrenal axis. Cortisol itself is associated with impaired memory and executive ability in older adults.6870 Further, both lead and cortisol are thought to alter common pathways in the mesocorticolimbic system, including calcium- and glutamate-mediated processes.64,67,71 Both cortisol and lead are associated with similar domains of cognitive function (especially memory and executive functioning). Glucocorticoid receptors are present in brain structures underlying these domains.

In 2 human studies, the inverse associations between bone lead level and cognition and blood pressure were more pronounced among men who self-reported greater stress.72,73

The role of the social or physical environment in modifying lead neurotoxicity was demonstrated by a study in which impaired spatial learning caused by lead exposure in utero through lactation was mitigated by rearing in an enriched environment.74 It also remediated deficits in gene expression in the hippocampus (i.e., N-methyl-d-asparate receptor subunit 1 mRNA and brain-derived neurotrophic factor mRNA).

Early life lead exposure also impairs the response to later brain insult, which may be more common in persons with lower socioeconomic position (SEP). For example, lead-exposed rats showed reduced behavioral recovery to an induced ischemic stroke in the hind limb parietal sensory-motor cortex in adulthood.75 Early lead exposure also impaired the topographic organization of the columnar processing units in the barrel field somatosensory cortex in rats76 and reorganization of the barrel field after whisker follicle ablation.77

This general finding was also shown in humans. In children, higher cord blood lead levels were associated with higher baseline systolic blood pressure, and higher early childhood lead levels were associated with greater total peripheral (vascular) resistance responses to acute stress.78 Few studies examined the environmental backdrop that gave rise to the spatial distribution of stress dysregulation. Among older adults living in neighborhoods with the most psychosocial hazards, tibia bone lead concentration had a more deleterious effect on certain domains of cognitive function.58 Despite these findings, most studies either did not systematically investigate how host characteristics (e.g., stress) altered the effect of lead, or were underpowered to do so.79

Air Pollution

Psychological stress might alter susceptibility to air pollution. Social stress and SEP modifed traffic-related air pollution effects on asthma etiology80 and exacerbation, and on birth weight.8183 Given that both psychosocial stress and air pollutants influence oxidative stress and cellular aging processes, future risk assessments must address whether social–environmental interactions contribute to cardiovascular disease.

SOCIOECONOMIC POSITION

SEP has a robust and complex association with many health states. Although the mechanisms underlying SEP gradients in health are not precisely known, different SEP groups clearly have markedly different health statuses as well as vulnerability to the impact of common exposures. The relationship between SEP and poor health is not confined to poor people because the dose–response is continuous.84 This potential risk modifier cannot be addressed simply by looking at the extremes.

SEP can be conceptualized and measured at both the individual and the area level (e.g., neighborhoods). Each level exerts an independent influence on an individual's chances of health. A wealthy person living in a poor area is exposed to the same excess of fast food, lack of nearby fresh produce, higher crime rate, greater distance to pharmacies, and lack of attractive green space as their neighbors, and this tends to impact their health and potentially their response to environmental pollutants.

Lead

Several studies demonstrated that the impact of lead on health was modified by SEP.85 Children from families of low SEP either expressed an exposure-associated deficit at lower levels of lead or failed to recover and compensate as quickly or completely as children of higher SEP.79 The effect of increased blood lead levels on children's performance on an end-of-grade reading test was more pronounced at the lower end rather than the upper end of the distribution of reading scores86 (i.e., the effect was greater among children facing other risk factors for lower achievement). This phenomenon was not restricted to lead. The adverse impact of prenatal exposure to secondhand smoke was significantly greater among children whose families faced greater material hardship.87 To understand such findings, it is necessary to deconstruct the complex construct of SEP into its component features, including nutrition, stress, other chemical exposures, and the social and physical environment. All might contribute to the apparent modification of lead toxicity by SEP.

Many studies treated SEP solely as a confounder of the lead–health association.88 Given the usual association between increased lead exposure and other risk factors, potential confounding by SEP must be considered, but careful consideration must also be given to the possibility that treating SEP solely as a confounder could lead to bias if lead exposure is on the causal pathway between SEP and health. This arises from the social patterning of lead exposure along socioeconomic lines.89,90 Methodological tools such as directed acyclic graphs or structural equation modeling might be useful in dissecting these complex relationships.

Air Pollution

Some evidence suggested that SEP modified the effect of air pollution. In a city where upper SEP individuals had higher exposures, the effects of PM10 on daily death varied by SEP,91 a result consistent with other findings.92,93 There was similar evidence on other outcomes, such as preterm delivery94 or birthweight.83

AGE

The elderly represent a particularly susceptible population, and by 2030, the proportion of the US population aged 65 and older will double. Cognitive decline in the elderly is a growing burden, and the number with dementia is expected to almost triple by 2040. Heterogeneity in cognition is especially pronounced in the elderly compared with younger adults,69 raising the question of whether environmental factors might be contributory. Air pollution was linked to increased inflammation in the brain,95 which was implicated in the development of Alzheimer's disease.96

The elderly are also at increased risk of cardiovascular disease, and air pollution has differential effects for a number of cardiovascular endpoints, including mortality.42,9799

Children are also considered to be a susceptible subgroup. Their greater vulnerability is attributed to metabolism (e.g., greater absorption of toxicants, reduced excretion, immaturity of detoxification pathways), developmental stage (e.g., ongoing development and organization of organs such as the central nervous system), and behavior (e.g., greater hand-to-mouth activity, greater relative dietary and respiratory intake of toxicants).100

Infants are born with only one-tenth the number of alveoli of adults and an underdeveloped epithelium. Alveolar development begins late in the third trimester.101 This postnatal development pattern is not merely a theoretical concern for air pollutants. Compared with controls, infant monkeys exposed to 5 months of episodic exposure to 0.5 parts per million of ozone had fewer airway generations, hyperplastic bronchiolar epithelium, and altered smooth muscle in terminal and respiratory bronchioles.102

OTHER ENVIRONMENTAL AGENTS

Evidence that coexposure to other neurotoxicants increases the likelihood of lead-associated impairments is limited and comes mostly from animal models.103106 In humans, 2 studies107,108 reported that the slope of the inverse relationship between blood lead level and neurodevelopment in infants was steeper among those with higher blood manganese levels. In NHANES (1999–2006), adults whose blood levels of cadmium and lead were both in the highest quartiles had greater odds of albuminuria and a reduced glomerular filtration rate.109

SOURCES OF SUSCEPTIBILITY

In addition to interacting with risk factors such as stress, SEP, genetics, and preexisting disease, lead and air pollution often covary with them, resulting in a further skewing of the risk distribution.

Lead

Since the 1980s, NHANES surveys have documented substantial socioeconomic and ethnic disparities in blood lead levels.110,111 Although blood lead levels have fallen, disparities remain. Among 1–5 year olds in NHANES 1999–2004, the frequencies of blood lead levels greater than 10 micrograms per deciliter, by race/ethnicity, were non-Hispanic Black 3.4%, Mexican-American 1.2%, and non-Hispanic White 1.2%. The frequencies were higher among poorer children and those on Medicaid. The strongest risk factors for higher blood lead levels were residence in older housing, poverty, age, and being non-Hispanic Black.112

Several genetic variants or polymorphisms that affect lead metabolism were examined in relation to their influence on internal dose or lead-related health effects. These included ALAD, the dopamine receptor D4, the HFE protein, Apo-E, and peptide transporter 2. Several studies compared lead biomarkers in individuals with the 2 codominant ALAD alleles (ALAD-1 and ALAD-2). There were substantial inconsistencies across studies, although some reported that ALAD-2 carriers had greater blood or cortical bone lead levels.5,113115 The plasma and whole blood lead ratio might be greater in ALAD-2 carriers.116 In a sample of Hispanic children, those homozygous for the peptide transporter 2 polymorphism had higher blood lead levels than those who were heterozygous or without this polymorphism.115 Children who were carriers of the variant HFE or transferrin gene had significantly higher blood lead levels than wild-type children, and children carrying both variants were more likely to have a blood lead level > 10 microgram per deciliter.117 Adult workers with the Vitamin D B allele had significantly higher patella lead levels.118

Systematic data are not available on the distribution of genetic variants of interest in relation to ethnicity and other demographic characteristics.119 However, the prevalence of some variants did vary by geographic region. The prevalence of ALAD-2 carrier status is 3% in Indian workers,113 8% in Chinese children,120 and 16% in US men.4 In another study, the frequencies of ALAD-2 allele were comparable in Asian and Caucasian samples but absent in African samples.121

Dietary factors such as iron and calcium might modify lead absorption or toxicity.122 In adult men, reduced dietary vitamin D was associated with increased bone lead levels, whereas decreased dietary Vitamin C and iron were associated with increased blood lead levels.123 Among children in the Philippines, higher folate and iron levels mitigated the inverse association between blood lead level and cognition.124

Air pollution

SEP is associated with exposure to a variety of air pollutants. In one study, Blacks and respondents of lower educational achievement and, to a lesser degree, lower income levels, were significantly more likely to live within a mile of a polluting facility.125 Exposure to air pollution from traffic was higher in persons of lower SEP.83,126,127 In addition, exposure to pollution from concentrated animal feed lots varied by SEP and race.128 Ambient air pollution concentrations were higher in neighborhoods where pregnant women were at higher risk for adverse pregnancy outcomes because of lower social conditions.129 People living in disadvantaged communities had higher exposure to both indoor and outdoor air pollution, and factors associated with poverty (e.g., cooking time, gas stove usage, occupant density, humidifiers) contributed to higher indoor concentrations of PM2.5 and nitrogen dioxide.130 Using such covariation of exposures with susceptibility factors, a recent risk assessment showed considerable disparity in the impact of air pollution on mortality in Mexico,131 and, relevant to cumulative risk assessment, showed the same disparity gradient for poor water quality and cooking fuel use.

CUMULATIVE EXPOSURE

The phrase cumulative exposure is used to describe 2 phenomena: (1) long-term sequelae of continuing exposure to a substance, and (2) the cumulative burden resulting from exposure to many stressors. Regarding the first, links were demonstrated between cumulative lead exposure (i.e., bone lead) and heart rate variability,38 hypertension,132,133 ischemic heart disease,134,135 and death.136 Examples of the second phenomenon were provided, namely, the enhancement of toxicity through interactions among different stressors. Several frameworks were established to conceptualize cumulative exposure to all stressors over the life course. These included Geronimus's concept of weathering137,138 and the concept of allostatic load.34,139 Both attempted to capture the cumulative wear and tear that occurs as a result of long-term exposure to multiple stressors and results in increased vulnerability and decreased reserve capacity. Zartarian and Schultz140 summarized various efforts undertaken by the Environmental Protection Agency to assess cumulative exposure in communities, including the “Cumulative Communities Research Program” within the National Exposure Research Laboratory. As Menzie et al.141 argued, a key requirement for thinking about cumulative exposures is the development of clear conceptual frameworks. Complications arise when there are interactions among multiple exposures, or when there are latencies in the onset of biological effect.

MARKERS OF CUMULATIVE EXPOSURE

Assessing cumulative burden from multiple stressors is difficult, and consideration of this issue could be advanced if a biomarker of cumulative burden were available. Telomeres are regions of noncoding DNA at the ends of chromosomes that protect against structural degradation, inappropriate recombination, and end-to-end fusion of chromosomes.142,143 Telomere length declines with each successive cell division and thus serves as a measure of biological aging.144 Shorter telomeres are also associated with chronic diseases, including diabetes,145 hypertension,145148 atherosclerosis,149 coronary artery disease,150,151 heart failure,152 and increased cardiovascular risk.145,153 Evidence from in vitro154156 and human studies145,155 suggested that oxidative stress and inflammation accelerate telomere shortening. Reduced blood DNA telomere length was also related to cumulative long-life exposure to tobacco smoking157,158 and to traffic pollution.159 Whether this is a useful biomarker either of susceptibility or cumulative burden is unclear, but the possibility deserves consideration.

CONCLUSIONS

Many risk assessments use uncertainty factors to address variability in susceptibility. However, that is usually taken as reflecting both a stochastic issue in extrapolating from observed data and an assumed random variability in toxicokinetics. Studies based on human epidemiology in the exposure range of interest, such as those done for criteria air pollutants, rarely incorporate uncertainty factors. The examples provided show that these approaches are not tenable because not only are there substantial variations in the effects of exposure, but the factors that produce such variation often covary with the exposure of interest. Hence, the distribution of risk is a key issue, which must be directly quantitated and provided to decision makers to use in conducting risk assessments.

Acknowledgments

This study was supported by the US Environmental Protection Agency.

Note. None of the statements in the article represent EPA policy.

Human Participant Protection

Institutional review board approval was not needed because no human subjects took part in this study.

References

  • 1.Schwartz J, Bellinger D, Glass T. Expanding the scope of environmental risk assessment to better include differential vulnerability and susceptibility. Am J Public Health 2011;101(suppl1):S88–S93 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Hackney JD, Linn WS, Buckley RD, et al. Experimental studies on human health effects of air pollutants: I. Design considerations. Arch Environ Health. 1975;30(8):373–378 [DOI] [PubMed] [Google Scholar]
  • 3.Kleeberger SR. Genetic aspects of susceptibility to air pollution. Eur Respir J. 2003;40:52s–56s [DOI] [PubMed] [Google Scholar]
  • 4.Weuve J, Kelsey KT, Schwartz J, et al. Delta-aminolevulinic acid dehydratase polymorphism and the relation between low level lead exposure and the Mini-Mental Status Examination in older men: the Normative Aging Study. Occup Environ Med. 2006;63(11):746–753 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Rajan P, Kelsey KT, Schwartz JD, et al. Interaction of the delta-aminolevulinic acid dehydratase polymorphism and lead burden on cognitive function: the VA Normative Aging Study. J Occup Environ Med. 2008;50(9):1053–1061 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Krieg EF, Jr, Butler MA, Chang MH, et al. Lead and cognitive function in ALAD genotypes in the third National Health and Nutrition Examination Survey. Neurotoxicol Teratol. 2009;31(6):364–371 [DOI] [PubMed] [Google Scholar]
  • 7.Stewart WF, Schwartz BS, Simon D, et al. ApoE genotype, past adult lead exposure, and neurobehavioral function. Environ Health Perspect. 2002;110(5):501–505 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Wright RO, Hu H, Silverman EK, et al. Apolipoprotein E genotype predicts 24-month bayley scales infant development score. Pediatr Res. 2003;54(6):819–825 [DOI] [PubMed] [Google Scholar]
  • 9.Lee BK, Glass TA, Wand GS, et al. Apolipoprotein E genotype, cortisol, and cognitive function in community-dwelling older adults. Am J Psychiatry. 2008;64(7):810–818 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Froehlich T, Lanphear B, Dietrich K, et al. Interactive effects of a DRD4 polymorphism, lead, and sex on executive functions in children. Biol Psychiatry. 2007;62:243–249 [DOI] [PubMed] [Google Scholar]
  • 11.Weaver V, Lee B, Todd A, et al. Effect modification by delta-aminolevulinic acid dehydratase, vitamin D receptor, and nitric oxide synthase gene polymorphisms on associations between patella lead and renal function in lead workers. Environ Res. 2006;102:61–69 [DOI] [PubMed] [Google Scholar]
  • 12.Krieg EF, Jr, Butler MA, Chang MH, et al. Lead and cognitive function in VDR genotypes in the third National Health and Nutrition Examination Survey. Neurotoxicol Teratol. 2010;32(2):262–272 [DOI] [PubMed] [Google Scholar]
  • 13.Wu J, Basha M, Zawia N. The environment, epigenetics and amyloidogenesis. J Mol Neurosci. 2008;34:1–7 [DOI] [PubMed] [Google Scholar]
  • 14.Pilsner JR, Hu H, Ettinger A, et al. Influence of prenatal lead exposure on genomic methylation of cord blood DNA. Environ Health Perspect. 2009;117(9):1466–1471 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Wright RO, Schwartz J, Wright RJ, et al. Biomarkers of lead exposure and DNA methylation within retrotransposons. Environ Health Perspect. 2010;118(6):790–795 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Chahine T, Baccarelli A, Litonjua A, et al. Particulate air pollution, oxidative stress genes, and heart rate variability in an elderly cohort. Environ Health Perspect. 2007;115(11):1617–1622 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Le Tertre A, Schwartz J, Touloumi G. Empirical Bayes and adjusted estimates approach to estimating the relation of mortality to exposure of PM(10). Risk Anal. 2005;25(3):711–718 [DOI] [PubMed] [Google Scholar]
  • 18.Ren C, Park SK, Vokonas P, et al. Air pollution and homocysteine: modification by oxidative stress related genes. Epidemiology. 2010;21(2):198–206 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Baja ES, Schwartz JD, Wellenius GA, et al. Traffic-related air pollution and QT interval: modification by diabetes, obesity, and oxidative stress gene polymorphisms in the Normative Aging Study. Environ Health Perspect. 2010;118:840–846 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Curjuric I, Imboden M, Schindler C, et al. HMOX1 and GST variants modify attenuation of FEF25-75% decline due to PM10 reduction. Euro Respir J. 2010;35:505–514 [DOI] [PubMed] [Google Scholar]
  • 21.Alexeeff SE, Litonjua AA, Wright RO, et al. Ozone exposure, antioxidant genes, and lung function in an elderly cohort: VA Normative Aging Study. Occup Environ Med. 2008;65(11):736–742 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Islam T, Berhane K, McConnell R, et al. Glutathione-S-transferase (GST) P1, GSTM1, exercise, ozone and asthma incidence in school children. Thorax. 2009;64(3):197–202 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Schroer KT, Biagini Myers JM, Ryan PH, et al. Associations between multiple environmental exposures and glutathione S-transferase P1 on persistent wheezing in a birth cohort. J Pediatr. 2009;154(3):401–408 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Madrigano J, Baccarelli A, Wright R, et al. Air pollution, obesity, genes, and cellular adhesion molecules. Occup Environ Med. 2010:67:312–317 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Park SK, O'Neill MS, Wright RO, et al. HFE genotype, particulate air pollution, and heart rate variability: a gene-environment interaction. Circulation. 2006;114(25):2798–2805 [DOI] [PubMed] [Google Scholar]
  • 26.Wilker E, Mittleman MA, Litonjua AA, et al. Postural changes in blood pressure associated with interactions between candidate genes for chronic respiratory diseases and exposure to particulate matter. Environ Health Perspect. 2009;117(6):935–940 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Baccarelli A, Cassano PA, Litonjua A, et al. Cardiac autonomic dysfunction: effects from particulate air pollution and protection by dietary methyl nutrients and metabolic polymorphisms. Circulation. 2008;117(14):1802–1809 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Wilker EH, Baccarelli A, Suh H, et al. Black carbon exposures, blood pressure and interactions with SNPs in microRNA processing genes. Environ Health Perspect. 2010;118(7):943–948 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Tarantini L, Bonzini M, Apostoli P, et al. Effects of particulate matter on genomic DNA methylation content and iNOS promoter methylation. Environ Health Perspect. 2009;117(2):217–222 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Perera F, Tang WY, Herbstman J, et al. Relation of DNA methylation of 5′-CpG island of ACSL3 to transplacental exposure to airborne polycyclic aromatic hydrocarbons and childhood asthma. PLoS ONE. 2009;4(2):e4488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Baccarelli A, Wright RO, Bollati V, et al. Rapid DNA methylation changes after exposure to traffic particles. Am J Respir Crit Care Med. 2009;179:572–578 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hamlin C. Predisposing causes and public health in early nineteenth century medical thought. Soc Hist Med. 1992;5:43–70 [DOI] [PubMed] [Google Scholar]
  • 33.Cassel J. The contribution of the social environment to host resistance: the Fourth Wade Hampton Frost Lecture. Am J Epidemiol. 1976;104(2):107–123 [DOI] [PubMed] [Google Scholar]
  • 34.McEwen BS, Stellar E. Stress and the individual. Mechanisms leading to disease. Arch Intern Med. 1993;153(18):2093–2101 [PubMed] [Google Scholar]
  • 35.Park SK, O'Neill MS, Vokonas PS, et al. Air pollution and heart rate variability: effect modification by chronic lead exposure. Epidemiology. 2008;19(1):111–120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Samet JM, Hatch GE, Horstman D, et al. Effect of antioxidant supplementation on ozone-induced lung injury in human subjects. Am J Respir Crit Care Med. 2001;164(5):819–825 [DOI] [PubMed] [Google Scholar]
  • 37.Tsaih SW, Korrick S, Schwartz J, et al. Lead, diabetes, hypertension, and renal function: the Normative Aging Study. Environ Health Perspect. 2004;112(11):1178–1182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Park SK, Schwartz J, Weisskopf M, et al. Low-level lead exposure, metabolic syndrome, and heart rate variability: the VA Normative Aging Study. Environ Health Perspect. 2006;114(11):1718–1724 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Ekong EB, Jaar BG, Weaver VM. Lead-related nephrotoxicity: a review of the epidemiologic evidence. Kidney Int. 2006;70(12):2074–2084 [DOI] [PubMed] [Google Scholar]
  • 40.Zanobetti A, Schwartz J. Cardiovascular damage by airborne particles: are diabetics more susceptible? Epidemiology. 2002;13(5):588–592 [DOI] [PubMed] [Google Scholar]
  • 41.Goldberg MS, Burnett RT, Bailar JC, 3rd, et al. The association between daily mortality and ambient air particle pollution in Montreal, Quebec. 2. Cause-specific mortality. Environ Res. 2001;86(1):26–36 [DOI] [PubMed] [Google Scholar]
  • 42.Bateson TF, Schwartz J. Who is sensitive to the effects of particulate air pollution on mortality? A case-crossover analysis of effect modifiers. Epidemiology. 2004;15(2):143–149 [DOI] [PubMed] [Google Scholar]
  • 43.Forastiere F, Stafoggia M, Berti G, et al. Particulate matter and daily mortality: a case-crossover analysis of individual effect modifiers. Epidemiology. 2008;19(4):571–580 [DOI] [PubMed] [Google Scholar]
  • 44.O'Neill MS, Veves A, Zanobetti A, et al. Diabetes enhances vulnerability to particulate air pollution-associated impairment in vascular reactivity and endothelial function. Circulation. 2005;111(22):2913–2920 [DOI] [PubMed] [Google Scholar]
  • 45.Dubowsky SD, Suh H, Schwartz J, et al. Diabetes, obesity, and hypertension may enhance associations between air pollution and markers of systemic inflammation. Environ Health Perspect. 2006;114(7):992–998 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.O'Neill MS, Veves A, Sarnat JA, et al. Air pollution and inflammation in type 2 diabetes: a mechanism for susceptibility. Occup Environ Med. 2007;64(6):373–379 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Chen JC, Cavallari JM, Stone PH, Christiani DC. Obesity is a modifier of autonomic cardiac responses to fine metal particulates. Environ Health Perspect. 2007;115(7):1002–1006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Alexeeff SE, Litonjua AA, Suh H, et al. Ozone exposure and lung function: effect modified by obesity and airways hyperresponsiveness in the VA Normative Aging Study. Chest. 2007;132(6):1890–1897 [DOI] [PubMed] [Google Scholar]
  • 49.Johnston RA, Theman TA, Lu FL, et al. Diet-induced obesity causes innate airway hyperresponsiveness to methacholine and enhances ozone-induced pulmonary inflammation. J Appl Physiol. 2008;104(6):1727–1735 [DOI] [PubMed] [Google Scholar]
  • 50.Lu FL, Johnston RA, Flynt L, et al. Increased pulmonary responses to acute ozone exposure in obese db/db mice. Am J Physiol Lung Cell Mol Physiol. 2006;290(5):L856–L865 [DOI] [PubMed] [Google Scholar]
  • 51.Schwartz J, Park SK, O'Neill MS, et al. Glutathione-S-transferase M1, obesity, statins, and autonomic effects of particles: gene-by-drug-by-environment interaction. Am J Respir Crit Care Med. 2005;172(12):1529–1533 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zeka A, Sullivan JR, Vokonas PS, et al. Inflammatory markers and particulate air pollution: characterizing the pathway to disease. Int J Epidemiol. 2006;35(5):1347–1354 [DOI] [PubMed] [Google Scholar]
  • 53.Chen JC, Schwartz J. Metabolic syndrome and inflammatory responses to long-term particulate air pollutants. Environ Health Perspect. 2008;116(5):612–617 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Romieu I, Tellez-Rojo MM, Lazo M, et al. Omega-3 fatty acid prevents heart rate variability reductions associated with particulate matter. Am J Respir Crit Care Med. 2005;172(12):1534–1540 [DOI] [PubMed] [Google Scholar]
  • 55.Trenga CA, Koenig JQ, Williams PV. Dietary antioxidants and ozone-induced bronchial hyperresponsiveness in adults with asthma. Arch Environ Health. 2001;56(3):242–249 [DOI] [PubMed] [Google Scholar]
  • 56.Chrousos GP, Gold PW. The concepts of stress and stress system disorders. Overview of physical and behavioral homeostasis. JAMA. 1992;267(9):1244–1252 [PubMed] [Google Scholar]
  • 57.Augustin T, Glass TA, James BD, Schwartz BS. Neighborhood psychosocial hazards and cardiovascular disease: the Baltimore Memory Study. Am J Public Health. 2008;98(9):1664–1670 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Glass TA, Bandeen-Roche K, McAtee M, et al. Neighborhood psychosocial hazards and the association of cumulative lead dose with cognitive function in older adults. Am J Epidemiol. 2009;169(6):683–692 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Kubzansky LD. Sick at heart: the pathophysiology of negative emotions. Cleve Clin J Med. 2007;74(Suppl 1):S67–S72 [DOI] [PubMed] [Google Scholar]
  • 60.Hubbs-Tait L, Nation JR, Krebs NF, Bellinger DC. Neurotoxicants, micronutrients, and social environments: individual and combined effects of children's development. Psychol Sci Public Interest. 2005;6(3):57–121 [DOI] [PubMed] [Google Scholar]
  • 61.Calderon J, Navarro ME, Jimenez-Capdeville ME, et al. Exposure to arsenic and lead and neuropsychological development in Mexican children. Environ Res. 2001;85(2):69–76 [DOI] [PubMed] [Google Scholar]
  • 62.Selye H, Somogyi A, Vegh P. Inflammation, topical stress and the concept of pluricausal diseases. Biochem Pharmacol. 1968;17(Suppl):107–122 [DOI] [PubMed] [Google Scholar]
  • 63.Virgolini MB, Chen K, Weston DD, et al. Interactions of chronic lead exposure and intermittent stress: consequences for brain catecholamine systems and associated behaviors and HPA axis function. Toxicol Sci. 2005;87(2):469–482 [DOI] [PubMed] [Google Scholar]
  • 64.Cory-Slechta DA, Virgolini MB, Thiruchelvam M, et al. Maternal stress modulates the effects of developmental lead exposure. Environ Health Perspect. 2004;112(6):717–730 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Schneider JS, Lee MH, Anderson DW, et al. Enriched environment during development is protective against lead-induced neurotoxicity. Brain Res. 2001;896(1-2):48–55 [DOI] [PubMed] [Google Scholar]
  • 66.Bushnell PJ, Shelton SE, Bowman RE. Elevation of blood lead concentration by confinement in the rhesus monkey. Bull Environ Contam Toxicol. 1979;22(6):819–826 [DOI] [PubMed] [Google Scholar]
  • 67.Virgolini MB, Bauter MR, Weston DD, Cory-Slechta DA. Permanent alterations in stress responsivity in female offspring subjected to combined maternal lead exposure and/or stress. Neurotoxicology. 2006;27(1):11–21 [DOI] [PubMed] [Google Scholar]
  • 68.Lupien SJ, de Leon M, de Santi S, et al. Cortisol levels during human aging predict hippocampal atrophy and memory deficits. Nat Neurosci. 1998;1(1):69–73 [DOI] [PubMed] [Google Scholar]
  • 69.Lupien SJ, Fiocco A, Wan N, et al. Stress hormones and human memory function across the lifespan. Psychoneuroendocrinology. 2005;30(3):225–242 [DOI] [PubMed] [Google Scholar]
  • 70.Lupien SJ, Lepage M. Stress, memory, and the hippocampus: can't live with it, can't live without it. Behav Brain Res. 2001;127(1-2):137–158 [DOI] [PubMed] [Google Scholar]
  • 71.Bowirrat A, Oscar-Berman M. Relationship between dopaminergic neurotransmission, alcoholism, and reward deficiency syndrome. Am J Med Genet B Neuropsychiatr Genet. 2005;132(1):29–37 [DOI] [PubMed] [Google Scholar]
  • 72.Peters JL, Kubzansky L, McNeely E, et al. Stress as a potential modifier of the impact of lead levels on blood pressure: the Normative Aging Study. Environ Health Perspect. 2007;115(8):1154–1159 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Peters J, Weisskopf M, Spiro A, 3rd, et al. Interaction of stress, lead burden and age on cognition in older men: the Normative Aging Study. Environ Health Perspect. 2010;118(4):505–510 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Guilarte T, Toscano C, McGlothan J, Weaver S. Environmental enrichment reverses cognitive and molecular deficits induced by developmental lead exposure. Ann Neurol. 2003;53:50–56 [DOI] [PubMed] [Google Scholar]
  • 75.Schneider JS, Decamp E. Postnatal lead poisoning impairs behavioral recovery following brain damage. Neurotoxicology. 2007;28(6):1153–1157 [DOI] [PubMed] [Google Scholar]
  • 76.Wilson M, Johnston M, Goldstein G, Blue M. Neonatal lead exposure impairs development of rodent barrel field cortex. Proc Natl Acad Sci USA. 2000;97:5540–5545 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Wilson M, Blue M, Patra R. Neonatal lead exposure impairs plasticity in developing rat somatosensory cortex. Paper presented at: 23rd International Neurotoxicology Conference; September 17–21, 2006; Little Rock, AR [Google Scholar]
  • 78.Gump BB, Stewart P, Reihman J, et al. Prenatal and early childhood blood lead levels and cardiovascular functioning in 9(1/2) year old children. Neurotoxicol Teratol. 2005;27(4):655–665 [DOI] [PubMed] [Google Scholar]
  • 79.Bellinger DC. Effect modification in epidemiologic studies of low-level neurotoxicant exposures and health outcomes. Neurotoxicol Teratol. 2000;22(1):133–140 [DOI] [PubMed] [Google Scholar]
  • 80.Clougherty JE, Levy JI, Kubzansky LD, et al. Synergistic effects of traffic-related air pollution and exposure to violence on urban asthma etiology. Environ Health Perspect. 2007;115(8):1140–1146 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Chen E, Schreier HM, Strunk RC, Brauer M. Chronic traffic-related air pollution and stress interact to predict biologic and clinical outcomes in asthma. Environ Health Perspect. 2008;116(7):970–975 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Shankardass K, McConnell R, Jerrett M, et al. Parental stress increases the effect of traffic-related air pollution on childhood asthma incidence. Proc Natl Acad Sci USA. 2009;106(30):12406–12411 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Zeka A, Melly SJ, Schwartz J. The effects of socioeconomic status and indices of physical environment on reduced birth weight and preterm births in Eastern Massachusetts. Environ Health. 2008;7:60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Marmot M. Health in an unequal world. Lancet. 2006;368(9552):2081–2094 [DOI] [PubMed] [Google Scholar]
  • 85.Hu H, Shih R, Rothenberg S, Schwartz BS. The epidemiology of lead toxicity in adults: measuring dose and consideration of other methodologic issues. Environ Health Perspect. 2007;115(3):455–462 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Miranda ML, Kim D, Reiter J, et al. Environmental contributors to the achievement gap. Neurotoxicology. 2009;30(6):1019–1024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Rauh VA, Whyatt RM, Garfinkel R, et al. Developmental effects of exposure to environmental tobacco smoke and material hardship among inner-city children. Neurotoxicol Teratol. 2004;26(3):373–385 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Bellinger DC. Lead neurotoxicity and socioeconomic status: conceptual and analytical issues. Neurotoxicology. 2008;29(5):828–832 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Elreedy S, Krieger N, Ryan PB, et al. Relations between individual and neighborhood-based measures of socioeconomic position and bone lead concentrations among community-exposed men: the Normative Aging Study. Am J Epidemiol. 1999;150(2):129–141 [DOI] [PubMed] [Google Scholar]
  • 90.Krieger N, Chen JT, Waterman PD, et al. Choosing area based socioeconomic measures to monitor social inequalities in low birth weight and childhood lead poisoning: The Public Health Disparities Geocoding Project (US). J Epidemiol Community Health. 2003;57(3):186–199 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Forastiere F, Stafoggia M, Tasco C, et al. Socioeconomic status, particulate air pollution, and daily mortality: differential exposure or differential susceptibility. Am J Ind Med. 2007;50(3):208–216 [DOI] [PubMed] [Google Scholar]
  • 92.Ou CQ, Hedley AJ, Chung RY, et al. Socioeconomic disparities in air pollution-associated mortality. Environ Res. 2008;107(2):237–244 [DOI] [PubMed] [Google Scholar]
  • 93.Zanobetti A, Schwartz J. Race, gender, and social status as modifiers of the effects of PM10 on mortality. J Occup Environ Med. 2000;42(5):469–474 [DOI] [PubMed] [Google Scholar]
  • 94.Yi O, Kim H, Ha E. Does area level socioeconomic status modify the effects of PM(10) on preterm delivery? Environ Res. 2010;110(1):55–61 [DOI] [PubMed] [Google Scholar]
  • 95.Campbell A, Oldham M, Becaria A, et al. Particulate matter in polluted air may increase biomarkers of inflammation in mouse brain. Neurotoxicology. 2005;26(1):133–140 [DOI] [PubMed] [Google Scholar]
  • 96.Akiyama H, Barger S, Barnum S, et al. Inflammation and Alzheimer's disease. Neurobiol Aging. 2000;21(3):383–421 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Fischer P, Hoek G, Brunekreef B, et al. Air pollution and mortality in The Netherlands: are the elderly more at risk? Eur Respir J Suppl. 2003;40:34s–38s [DOI] [PubMed] [Google Scholar]
  • 98.Aga E, Samoli E, Touloumi G, et al. Short-term effects of ambient particles on mortality in the elderly: results from 28 cities in the APHEA2 project. Eur Respir J Suppl. 2003;40:28s–33s [DOI] [PubMed] [Google Scholar]
  • 99.Devlin RB, Ghio AJ, Kehrl H, et al. Elderly humans exposed to concentrated air pollution particles have decreased heart rate variability. Eur Respir J Suppl. 2003;40:76s–80s [DOI] [PubMed] [Google Scholar]
  • 100.Moya J, Bearer C, Etzel R. Children's behavior and physiology and how it affects exposure to environmental contaminants. Pediatrics. 2004;113:996–1006 [PubMed] [Google Scholar]
  • 101.Langston C, Kida K, Reed M, Thurlbeck WM. Human lung growth in late gestation and in the neonate. Am Rev Respir Dis. 1984;129:607–613 [PubMed] [Google Scholar]
  • 102.Fanucchi MV, Plopper CG, Evans MJ, et al. Cyclic exposure to ozone alters distal airway development in infant rhesus monkeys. Am J Physiol Lung Cell Mol Physiol. 2006;291(4):L644–L650 [DOI] [PubMed] [Google Scholar]
  • 103.Chandra A, Ali M, Saxena D, Murthy R. Behavioral and neurochemical changes in rats simultaneously exposed to manganese and lead. Arch Toxicol. 1981;49:49–56 [DOI] [PubMed] [Google Scholar]
  • 104.Chandra A, Murthy R, Saxena D, Lal B. Effects of pre- and postnatal combined exposure to Pb and Mn on brain development in rats. Ind Health. 1983;21:273–279 [DOI] [PubMed] [Google Scholar]
  • 105.Rodriguez VM, Dufour L, Carrizales L, et al. Effects of oral exposure to mining waste on in vivo dopamine release from rat striatum. Environ Health Perspect. 1998;106(8):487–491 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Mejia JJ, Diaz-Barriga F, Calderon J, et al. Effects of lead-arsenic combined exposure on central monoaminergic systems. Neurotoxicol Teratol. 1997;19(6):489–497 [DOI] [PubMed] [Google Scholar]
  • 107.Claus Henn B, Ettinger A, Schwartz J, et al. Early postnatal blood manganese levels and children's neurodevelopment. Epidemiology. 2010;21(4):433–439 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Kim Y, Kim BN, Hong YC, et al. Co-exposure to environmental lead and manganese affects the intelligence of school-aged children. Neurotoxicology. 2009;30(4):564–571 [DOI] [PubMed] [Google Scholar]
  • 109.Navas-Acien A, Tellez-Plaza M, Guallar E, et al. Blood cadmium and lead and chronic kidney disease in US adults: a joint analysis. Am J Epidemiol. 2009;170(9):1156–1164 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Mahaffey KR, Annest JL, Roberts J, Murphy RS. National estimates of blood lead levels: United States, 1976-1980: association with selected demographic and socioeconomic factors. N Engl J Med. 1982;307(10):573–579 [DOI] [PubMed] [Google Scholar]
  • 111.Mahaffey KR, Gartside PS, Glueck CJ. Blood lead levels and dietary calcium intake in 1- to 11-year-old children: the Second National Health and Nutrition Examination Survey, 1976 to 1980. Pediatrics. 1986;78(2):257–262 [PubMed] [Google Scholar]
  • 112.Jones RL, Homa DM, Meyer PA, et al. Trends in blood lead levels and blood lead testing among US children aged 1 to 5 years, 1988-2004. Pediatrics. 2009;123(3):e376–e385 [DOI] [PubMed] [Google Scholar]
  • 113.Shaik AP, Jamil K. A study on the ALAD gene polymorphisms associated with lead exposure. Toxicol Ind Health. 2008;24(7):501–506 [DOI] [PubMed] [Google Scholar]
  • 114.Shen XM, Wu SH, Yan CH, et al. Delta-aminolevulinate dehydratase polymorphism and blood lead levels in Chinese children. Environ Res. 2001;85(3):185–190 [DOI] [PubMed] [Google Scholar]
  • 115.Sobin C, Gutierrez M, Alterio H. Polymorphisms of delta-aminolevulinic acid dehydratase (ALAD) and peptide transporter 2 (PEPT2) genes in children with low-level lead exposure. Neurotoxicology. 2009;30(6):881–887 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Montenegro MF, Barbosa F, Jr, Sandrim VC, et al. A polymorphism in the delta-aminolevulinic acid dehydratase gene modifies plasma/whole blood lead ratio. Arch Toxicol. 2006;80(7):394–398 [DOI] [PubMed] [Google Scholar]
  • 117.Hopkins MR, Ettinger AS, Hernandez-Avila M, et al. Variants in iron metabolism genes predict higher blood lead levels in young children. Environ Health Perspect. 2008;116(9):1261–1266 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Theppeang K, Schwartz BS, Lee BK, et al. Associations of patella lead with polymorphisms in the vitamin D receptor, delta-aminolevulinic acid dehydratase and endothelial nitric oxide synthase genes. J Occup Environ Med. 2004;46(6):528–537 [DOI] [PubMed] [Google Scholar]
  • 119.Pritchard JK. Are rare variants responsible for susceptibility to complex diseases? Am J Hum Genet. 2001;69(1):124–137 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Wu S, Yan C, Shen X. Molecular genetic susceptibility to lead poisoning. J Hygiene Res. 2004;33(2):226–228, 232 [PubMed] [Google Scholar]
  • 121.Fujihara J, Agusa T, Yasuda T, et al. Ethnic variation in genotype frequencies of delta-aminolevulinic acid dehydratase (ALAD). Toxicol Lett. 2009;191(2-3):236–239 [DOI] [PubMed] [Google Scholar]
  • 122.Elmarsafawy SF, Jain NB, Schwartz J, et al. Dietary calcium as a potential modifier of the relationship of lead burden to blood pressure. Epidemiology. 2006;17(5):531–537 [DOI] [PubMed] [Google Scholar]
  • 123.Cheng Y, Willett WC, Schwartz J, et al. Relation of nutrition to bone lead and blood lead levels in middle-aged to elderly men. The Normative Aging Study. Am J Epidemiol. 1998;147(12):1162–1174 [DOI] [PubMed] [Google Scholar]
  • 124.Solon O, Riddell TJ, Quimbo SA, et al. Associations between cognitive function, blood lead concentration, and nutrition among children in the central Philippines. J Pediatr. 2008;152(2):237–243 [DOI] [PubMed] [Google Scholar]
  • 125.Mohai P, Lantz PM, Morenoff J, et al. Racial and socioeconomic disparities in residential proximity to polluting industrial facilities: evidence from the Americans’ Changing Lives Study. Am J Public Health. 2009;99(Suppl 3):S649–S656 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Havard S, Deguen S, Zmirou-Navier D, et al. Traffic-related air pollution and socioeconomic status: a spatial autocorrelation study to assess environmental equity on a small-area scale. Epidemiology. 2009;20(2):223–230 [DOI] [PubMed] [Google Scholar]
  • 127.Yanosky JD, Schwartz J, Suh HH. Associations between measures of socioeconomic position and chronic nitrogen dioxide exposure in Worcester, Massachusetts. J Toxicol Environ Health A. 2008;71(24):1593–1602 [DOI] [PubMed] [Google Scholar]
  • 128.Mirabelli MC, Wing S, Marshall SW, Wilcosky TC. Race, poverty, and potential exposure of middle-school students to air emissions from confined swine feeding operations. Environ Health Perspect. 2006;114(4):591–596 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Woodruff TJ, Parker JD, Kyle AD, Schoendorf KC. Disparities in exposure to air pollution during pregnancy. Environ Health Perspect. 2003;111(7):942–946 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Baxter LK, Clougherty JE, Laden F, Levy JI. Predictors of concentrations of nitrogen dioxide, fine particulate matter, and particle constituents inside of lower socioeconomic status urban homes. J Expo Sci Environ Epidemiol. 2007;17(5):433–444 [DOI] [PubMed] [Google Scholar]
  • 131.Stevens GA, Dias RH, Ezzati M. The effects of 3 environmental risks on mortality disparities across Mexican communities. Proc Natl Acad Sci USA. 2008;105(44):16860–16865 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Cheng Y, Schwartz J, Sparrow D, et al. Bone lead and blood lead levels in relation to baseline blood pressure and the prospective development of hypertension: the Normative Aging Study. Am J Epidemiol. 2001;153(2):164–171 [DOI] [PubMed] [Google Scholar]
  • 133.Hu H, Aro A, Payton M, et al. The relationship of bone and blood lead to hypertension. The Normative Aging Study. JAMA. 1996;275(15):1171–1176 [PubMed] [Google Scholar]
  • 134.Jain NB, Potula V, Schwartz J, et al. Lead levels and ischemic heart disease in a prospective study of middle-aged and elderly men: the VA Normative Aging Study. Environ Health Perspect. 2007;115(6):871–875 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Schwartz J. Lead, blood pressure, and cardiovascular disease in men. Arch Environ Health. 1995;50(1):31–37 [DOI] [PubMed] [Google Scholar]
  • 136.Weisskopf MG, Jain N, Nie H, et al. A prospective study of bone lead concentration and death from all causes, cardiovascular diseases, and cancer in the Department of Veterans Affairs Normative Aging Study. Circulation. 2009; 120(12):1056–1064 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Geronimus AT. The weathering hypothesis and the health of African-American women and infants: evidence and speculations. Ethn Dis. 1992;2(3):207–221 [PubMed] [Google Scholar]
  • 138.Geronimus AT, Hicken M, Keene D, Bound J. “Weathering” and age patterns of allostatic load scores among blacks and whites in the United States. Am J Public Health. 2006;96(5):826–833 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.McEwen BS. Allostasis and allostatic load: implications for neuropsychopharmacology. Neuropsychopharmacology. 2000;22(2):108–124 [DOI] [PubMed] [Google Scholar]
  • 140.Zartarian VG, Schultz BD. The EPA's human exposure research program for assessing cumulative risk in communities. J Expo Sci Environ Epidemiol. 2010;20(4):351–358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Menzie CA, MacDonell MM, Mumtaz M. A phased approach for assessing combined effects from multiple stressors. Environ Health Perspect. 2007;115(5):807–816 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Blackburn EH. Switching and signaling at the telomere. Cell. 2001;106(6):661–673 [DOI] [PubMed] [Google Scholar]
  • 143.Wong JM, Collins K. Telomere maintenance and disease. Lancet. 2003;362(9388):983–988 [DOI] [PubMed] [Google Scholar]
  • 144.Aviv A. Chronology versus biology: telomeres, essential hypertension, and vascular aging. Hypertension. 2002;40(3):229–232 [DOI] [PubMed] [Google Scholar]
  • 145.Fitzpatrick AL, Kronmal RA, Gardner JP, et al. Leukocyte telomere length and cardiovascular disease in the Cardiovascular Health Study. Am J Epidemiol. 2007;165(1):14–21 [DOI] [PubMed] [Google Scholar]
  • 146.Yang Z, Huang X, Jiang H, et al. Short telomeres and prognosis of hypertension in a Chinese population. Hypertension. 2009;53(4):639–645 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Perez-Rivero G, Ruiz-Torres MP, Rivas-Elena JV, et al. Mice deficient in telomerase activity develop hypertension because of an excess of endothelin production. Circulation. 2006;114(4):309–317 [DOI] [PubMed] [Google Scholar]
  • 148.Vasan RS, Massaro JM, Wilson PW, et al. Antecedent blood pressure and risk of cardiovascular disease: the Framingham Heart Study. Circulation. 2002;105(1):48–53 [DOI] [PubMed] [Google Scholar]
  • 149.Benetos A, Gardner JP, Zureik M, et al. Short telomeres are associated with increased carotid atherosclerosis in hypertensive subjects. Hypertension. 2004;43(2):182–185 [DOI] [PubMed] [Google Scholar]
  • 150.Brouilette SW, Moore JS, McMahon AD, et al. Telomere length, risk of coronary heart disease, and statin treatment in the West of Scotland Primary Prevention Study: a nested case-control study. Lancet. 2007;369(9556):107–114 [DOI] [PubMed] [Google Scholar]
  • 151.Mukherjee M, Brouilette S, Stevens S, et al. Association of shorter telomeres with coronary artery disease in Indian subjects. Heart. 2009;95(8):669–673 [DOI] [PubMed] [Google Scholar]
  • 152.van der Harst P, van der Steege G, de Boer RA, et al. Telomere length of circulating leukocytes is decreased in patients with chronic heart failure. J Am Coll Cardiol. 2007;49(13):1459–1464 [DOI] [PubMed] [Google Scholar]
  • 153.Edo MD, Andres V. Aging, telomeres, and atherosclerosis. Cardiovasc Res. 2005;66(2):213–221 [DOI] [PubMed] [Google Scholar]
  • 154.Honda S, Hjelmeland LM, Handa JT. Oxidative stress–induced single-strand breaks in chromosomal telomeres of human retinal pigment epithelial cells in vitro. Invest Ophthalmol Vis Sci. 2001;42(9):2139–2144 [PubMed] [Google Scholar]
  • 155.Saretzki G, Von Zglinicki T. Replicative aging, telomeres, and oxidative stress. Ann N Y Acad Sci. 2002;959:24–29 [DOI] [PubMed] [Google Scholar]
  • 156.von Zglinicki T. Oxidative stress shortens telomeres. Trends Biochem Sci. 2002;27(7):339–344 [DOI] [PubMed] [Google Scholar]
  • 157.Nawrot TS, Staessen JA, Gardner JP, Aviv A. Telomere length and possible link to X chromosome. Lancet. 2004;363(9408):507–510 [DOI] [PubMed] [Google Scholar]
  • 158.Muller KC, Welker L, Paasch K, et al. Lung fibroblasts from patients with emphysema show markers of senescence in vitro. Respir Res. 2006;7:32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Hoxha M, Dioni L, Bonzini M, et al. Association between leukocyte telomere shortening and exposure to traffic pollution: a cross-sectional study on traffic officers and indoor office workers. Environ Health. 2009;8:41. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Public Health are provided here courtesy of American Public Health Association

RESOURCES