Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Curr Protoc ; 3(3): e698, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36912610

ABSTRACT

Although noteworthy progress has been made in developing alternatives to animal testing, nonhuman primates still play a critical role in advancing biomedical research and will likely do so for many years. Core similarities between monkeys and humans in genetics, physiology, reproduction, development, and behavior make them excellent models for translational studies relevant to human health. This unit is designed to specifically address the role of nonhuman primates in neurotoxicology research and outlines the specialized assessments that can be used to measure exposure-related changes at the structural, chemical, cellular, molecular, and functional levels. © 2023 Wiley Periodicals LLC.


Subject(s)
Biomedical Research , Primates , Animals , Humans , Haplorhini , Research Design , Reproduction
2.
Environ Health Perspect ; 130(9): 97003, 2022 09.
Article in English | MEDLINE | ID: mdl-36102641

ABSTRACT

BACKGROUND: The excitotoxic molecule, domoic acid (DA), is a marine algal toxin known to induce overt hippocampal neurotoxicity. Recent experimental and epidemiological studies suggest adverse neurological effects at exposure levels near the current regulatory limit (20 ppm, ∼0.075-0.1mg/kg). At these levels, cognitive effects occur in the absence of acute symptoms or evidence of neuronal death. OBJECTIVES: This study aimed to identify adverse effects on the nervous system from prolonged, dietary DA exposure in adult, female Macaca fascicularis monkeys. METHODS: Monkeys were orally exposed to 0, 0.075, and 0.15mg/kg per day for an average of 14 months. Clinical blood counts, chemistry, and cytokine levels were analyzed in the blood. In-life magnetic resonance (MR) imaging assessed volumetric and tractography differences in and between the hippocampus and thalamus. Histology of neurons and glia in the fornix, fimbria, internal capsule, thalamus, and hippocampus was evaluated. Hippocampal RNA sequencing was used to identify differentially expressed genes. Enrichment of gene networks for neuronal health, excitotoxicity, inflammation/glia, and myelin were assessed with Gene Set Enrichment Analysis. RESULTS: Clinical blood counts, chemistry, and cytokine levels were not altered with DA exposure in nonhuman primates. Transcriptome analysis of the hippocampus yielded 748 differentially expressed genes (fold change≥1.5; p≤0.05), reflecting differences in a broad molecular profile of intermediate early genes (e.g., FOS, EGR) and genes related to myelin networks in DA animals. Between exposed and control animals, MR imaging showed comparable connectivity of the hippocampus and thalamus and histology showed no evidence of hypomyelination. Histological examination of the thalamus showed a larger microglia soma size and an extension of cell processes, but suggestions of a GFAP+astrocyte response showed no indication of astrocyte hypertrophy. DISCUSSION: In the absence of overt hippocampal excitotoxicity, chronic exposure of Macaca fascicularis monkeys to environmentally relevant levels of DA suggested a subtle shift in the molecular profile of the hippocampus and the microglia phenotype in the thalamus that was possibly reflective of an adaptive response due to prolonged DA exposure. https://doi.org/10.1289/EHP10923.


Subject(s)
Kainic Acid , Neurotoxicity Syndromes , Animals , Cytokines , Female , Kainic Acid/analogs & derivatives , Kainic Acid/toxicity , Macaca fascicularis , Marine Toxins/toxicity
3.
Pharmacol Ther ; 227: 107865, 2021 11.
Article in English | MEDLINE | ID: mdl-33930455

ABSTRACT

Domoic acid (DA), the causative agent for the human syndrome Amnesic Shellfish Poisoning (ASP), is a potent, naturally occurring neurotoxin produced by common marine algae. DA accumulates in seafood, and humans and wildlife alike can subsequently be exposed when consuming DA-contaminated shellfish or finfish. While strong regulatory limits protect people from the acute effects associated with ASP, DA is an increasingly significant public health concern, particularly for coastal dwelling populations, and there is a growing body of evidence suggesting that there are significant health consequences following repeated exposures to levels of the toxin below current safety guidelines. However, gaps in scientific knowledge make it difficult to precisely determine the risks of contemporary low-level exposure scenarios. The present review characterizes the toxicokinetics and neurotoxicology of DA, discussing results from clinical and preclinical studies after both adult and developmental DA exposure. The review also highlights crucial areas for future DA research and makes the case that DA safety limits need to be reassessed to best protect public health from deleterious effects of this widespread marine toxin.


Subject(s)
Environmental Exposure , Kainic Acid/analogs & derivatives , Public Health , Environmental Exposure/adverse effects , Environmental Exposure/statistics & numerical data , Humans , Kainic Acid/adverse effects , Risk Assessment
4.
Toxicol Appl Pharmacol ; 398: 115027, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32360744

ABSTRACT

Domoic acid (DA) is a marine algal toxin that causes acute and chronic neurotoxicity in animals and humans. Prenatal exposure to DA has been associated with neuronal damage and cognitive and behavioral deficits in juvenile California sea lions, cynomolgus monkeys and rodents. Yet, the toxicokinetics (TK) of DA during pregnancy and the maternal-fetal disposition of DA have not been fully elucidated. In this study, we investigated the TK before, during, and after pregnancy and the maternal-fetal disposition of DA in 22 cynomolgus monkeys following daily oral doses of 0.075 or 0.15 mg/kg/day of DA. The AUC0-τ of DA was not changed while the renal clearance of DA was increased by 30-90% during and after pregnancy when compared to the pre-pregnancy values. DA was detected in the infant plasma and in the amniotic fluid at delivery. The infant plasma concentrations correlated positively with both the maternal plasma and the amniotic fluid concentrations. The paired infant-to-maternal plasma DA concentration ratios ranged from 0.3 to 0.6 and increased as a function of time which suggests placental efflux and longer apparent fetal half-life than the maternal half-life. The paired amniotic fluid-to-infant plasma DA concentration ratios ranged from 4.5 to 7.5 which indicates significant accumulation of DA in the amniotic fluid. A maternal-fetal TK model was developed to explore the processes that give the observed maternal-fetal disposition of DA. The final model suggests that placental transport and recirculation of DA between the fetus and amniotic fluid are major determining factors of the maternal-fetal TK of DA.


Subject(s)
Kainic Acid/analogs & derivatives , Maternal-Fetal Exchange/physiology , Primates/metabolism , Amniotic Fluid/metabolism , Animals , Double-Blind Method , Female , Fetus/metabolism , Kainic Acid/metabolism , Macaca fascicularis/metabolism , Placenta/metabolism , Pregnancy
5.
Neurotoxicology ; 72: 114-124, 2019 05.
Article in English | MEDLINE | ID: mdl-30826346

ABSTRACT

Domoic acid (DA) is an excitatory neurotoxin produced by marine algae and responsible for Amnesiac Shellfish Poisoning in humans. Current regulatory limits (˜0.075-0.1 mg/kg/day) protect against acute toxicity, but recent studies suggest that the chronic consumption of DA below the regulatory limit may produce subtle neurotoxicity in adults, including decrements in memory. As DA-algal blooms are increasing in both severity and frequency, we sought to better understand the effects of chronic DA exposure on reproductive and neurobehavioral endpoints in a preclinical nonhuman primate model. To this end, we initiated a long-term study using adult, female Macaca fascicularis monkeys exposed to daily, oral doses of 0.075 or 0.15 mg/kg of DA for a range of 321-381, and 346-554 days, respectively. This time period included a pre-pregnancy, pregnancy, and postpartum period. Throughout these times, trained data collectors observed intentional tremors in some exposed animals during biweekly clinical examinations. The present study explores the basis of this neurobehavioral finding with in vivo imaging techniques, including diffusion tensor magnetic resonance imaging and spectroscopy. Diffusion tensor analyses revealed that, while DA exposed macaques did not significantly differ from controls, increases in DA-related tremors were negatively correlated with fractional anisotropy, a measure of structural integrity, in the internal capsule, fornix, pons, and corpus callosum. Brain concentrations of lactate, a neurochemical closely linked with astrocytes, were also weakly, but positively associated with tremors. These findings are the first documented results suggesting that chronic oral exposure to DA at concentrations near the current human regulatory limit are related to structural and chemical changes in the adult primate brain.


Subject(s)
Brain/drug effects , Brain/pathology , Kainic Acid/analogs & derivatives , Marine Toxins/toxicity , Neurotoxins/toxicity , Animals , Diffusion Tensor Imaging , Female , Kainic Acid/administration & dosage , Kainic Acid/toxicity , Macaca fascicularis , Marine Toxins/administration & dosage , Neurotoxins/administration & dosage , Postpartum Period , Pregnancy , Tremor/chemically induced
6.
Neurotoxicol Teratol ; 73: 1-8, 2019.
Article in English | MEDLINE | ID: mdl-30690118

ABSTRACT

Domoic Acid (DA) is a naturally-occurring marine neurotoxin that is increasingly recognized as an important public health issue. Prenatal DA exposure occurs through the maternal consumption of contaminated shellfish/finfish. To better understand the fetal risks associated with DA, we initiated a longitudinal, preclinical study focused on the reproductive and developmental effects of chronic, low-dose oral DA exposure. To this end, 32 adult female Macaca fascicularis monkeys were orally dosed with 0, 0.075 or 0.15 mg/kg/day DA on a daily basis prior to breeding and throughout breeding and pregnancy. The doses included the proposed human Tolerable Daily Intake (TDI) (0.075 mg/kg/day) for DA. Adult females were bred to nonexposed males. To evaluate development during early infancy, offspring were administered a Neonatal Assessment modeled after the human Neonatal Behavior Assessment Scale and a series of Visual Recognition Memory problems using the novelty paradigm. Results indicated that prenatal DA exposure did not impact early survival reflexes or responsivity to the environment. Findings from the recognition memory assessment, given between 1 and 2 months of age, showed that exposed and control infants demonstrated robust novelty scores when test problems were relatively easy to solve. Performance was not diminished by the introduction of delay periods. However, when more difficult recognition problems were introduced, the looking behavior of the 0.15 mg/kg DA group was random and infants failed to show differential visual attention to novel test stimuli. This finding suggests subtle but significant impairment in recognition memory and demonstrates that chronic fetal exposure to DA may impact developing cognitive processes.


Subject(s)
Animals, Newborn/psychology , Behavior, Animal/drug effects , Kainic Acid/analogs & derivatives , Marine Toxins/toxicity , Memory/drug effects , Neurotoxins/toxicity , Prenatal Exposure Delayed Effects/etiology , Animals , Dose-Response Relationship, Drug , Female , Kainic Acid/blood , Kainic Acid/toxicity , Macaca fascicularis , Male , Marine Toxins/blood , Neurotoxins/blood , Pregnancy , Prenatal Exposure Delayed Effects/blood , Prenatal Exposure Delayed Effects/psychology
7.
Neurotoxicol Teratol ; 72: 10-21, 2019.
Article in English | MEDLINE | ID: mdl-30615984

ABSTRACT

Domoic Acid (DA) is a naturally-occurring excitotoxin, produced by marine algae, which can bioaccumulate in shellfish and finfish. The consumption of seafood contaminated with DA is associated with gastrointestinal illness that, in the case of high DA exposure, can evolve into a spectrum of responses ranging from agitation to hallucinations, memory loss, seizures and coma. Because algal blooms that produce DA are becoming more widespread and very little is known about the dangers of chronic, low-dose exposure, we initiated a preclinical study focused on the reproductive and developmental effects of DA in a nonhuman primate model. To this end, 32 adult female Macaca fascicularis monkeys were orally exposed to 0, 0.075 or 0.15 mg/kg/day DA on a daily basis, prior to and during pregnancy. Females were bred to non-exposed males and infants were evaluated at birth. Results from this study provided no evidence of changes in DA plasma concentrations with chronic exposure. DA exposure was not associated with reproductive toxicity or adverse changes in the physical characteristics of newborns. However, in an unanticipated finding, our clinical observations revealed the presence of subtle neurological effects in the form of intentional tremors in the exposed adult females. While females in both dose groups displayed increased tremoring, the effect was dose-dependent and observed at a higher rate in females exposed to 0.15 mg/kg/day. These results demonstrate that chronic, low-level exposure to DA is associated with injury to the adult CNS and suggest that current regulatory guidelines designed to protect human health may not be adequate for high-frequency shellfish consumers.


Subject(s)
Kainic Acid/analogs & derivatives , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects/chemically induced , Reproduction/drug effects , Water Pollutants, Chemical/toxicity , Administration, Oral , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Female , Kainic Acid/administration & dosage , Kainic Acid/blood , Kainic Acid/toxicity , Macaca fascicularis , Male , Pregnancy , Prenatal Exposure Delayed Effects/blood , Water Pollutants, Chemical/administration & dosage , Water Pollutants, Chemical/blood
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.
ACS Omega ; 3(9): 12079-12088, 2018 Sep 30.
Article in English | MEDLINE | ID: mdl-30320288

ABSTRACT

Domoic acid (DA) is a marine neurotoxin produced by several species of Pseudo-nitzschia. DA causes severe neurological toxicity in humans and animals. To address the current analytical need to quantify low levels of DA in human and animal body fluids, a sensitive and selective high performance liquid chromatography-tandem mass spectrometry method was developed to measure DA in plasma and urine. This method was fully validated to accurately and precisely quantify DA between 0.31 and 16 ng/mL in plasma and between 7.8 and 1000 ng/mL in urine. Our group introduced the use of a novel internal standard, tetrahydrodomoic acid to control for matrix effects and other sources of variability. This validated method will be useful to assess DA concentrations in biological samples of human or animal origin after suspected DA exposure from contaminated food. It will also be applicable to sentinel programs and research studies to analyze body fluids with low levels of DA.

10.
Pharmacol Ther ; 182: 133-151, 2018 02.
Article in English | MEDLINE | ID: mdl-28847562

ABSTRACT

The broad-based legalization of cannabis use has created a strong need to understand its impact on human health and behavior. The risks that may be associated with cannabis use, particularly for sensitive subgroups such as pregnant women, are difficult to define because of a paucity of dose-response data and the recent increase in cannabis potency. Although there is a large body of evidence detailing the mode of action of Δ9-tetrahydrocannabinol (THC) in adults, little work has focused on understanding how cannabis use during pregnancy may impact the development of the fetal nervous system and whether additional plant-derived cannabinoids might participate. This manuscript presents an overview of the historical and contemporary literature focused on the mode of action of THC in the developing brain, comparative pharmacokinetics in both pregnant and nonpregnant model systems and neurodevelopmental outcomes in exposed offspring. Despite growing public health significance, pharmacokinetic studies of THC have focused on nonpregnant adult subjects and there are few published reports on disposition parameters during pregnancy. Data from preclinical species show that THC readily crosses the placenta although fetal exposures appear lower than maternal exposures. The neurodevelopmental data in humans and animals suggest that prenatal exposure to THC may lead to subtle, persistent changes in targeted aspects of higher-level cognition and psychological well-being. There is an urgent need for well-controlled studies in humans and preclinical models on THC as a developmental neurotoxicant. Until more information is available, pregnant women should not assume that using cannabis during pregnancy is safe.


Subject(s)
Brain/drug effects , Cognition/drug effects , Dronabinol/adverse effects , Dronabinol/pharmacokinetics , Marijuana Use/adverse effects , Prenatal Exposure Delayed Effects/chemically induced , Animals , Brain/growth & development , Female , Humans , Models, Neurological , Pregnancy
11.
Drug Metab Dispos ; 46(2): 155-165, 2018 02.
Article in English | MEDLINE | ID: mdl-29150543

ABSTRACT

Domoic acid (DA), a neurotoxin, is produced by marine algae and has caused toxications worldwide in animals and humans. However, the toxicokinetics of DA have not been fully evaluated, and information is missing on the disposition of DA following oral exposures at doses that are considered safe for human consumption. In this study, toxicokinetics of DA were investigated in cynomolgus monkeys following single doses of 5 µg/kg DA intravenously, 0.075 mg/kg DA orally, and 0.15 mg/kg DA orally. After intravenous dosing, DA had a systemic clearance of 124 ± 71 (ml/h)/kg, volume of distribution at steady state of 131 ± 71 ml/kg and elimination half-life of 1.2 ± 1.1 hours. However, following oral dosing, the average terminal half-life of DA was 11.3 ± 2.4 hours, indicating that DA disposition follows flip-flop kinetics with slow, rate-limiting absorption. The absorption of DA was low after oral dosing with absolute bioavailability of 6% ± 4%. The renal clearance of DA was variable [21-152 (ml/h)/kg] with 42% ± 11% of the intravenous DA dose recovered in urine. A physiologically based pharmacokinetic model was developed for DA in monkeys and humans that replicated the flip-flop kinetics observed after oral administration and allowed simulation of urinary excretion and brain and kidney distribution of DA following intravenous and oral dosing. This study is the first to characterize DA disposition at exposure levels close to the current estimated tolerable daily intake and to mechanistically model DA disposition in a model species, providing important information of the toxicokinetics of DA for human safety assessment.


Subject(s)
Kainic Acid/analogs & derivatives , Administration, Oral , Adolescent , Adult , Aged , Animals , Biological Availability , Female , Half-Life , Humans , Injections, Intravenous/methods , Kainic Acid/pharmacokinetics , Kinetics , Macaca fascicularis , Male , Middle Aged , Shellfish , Tissue Distribution , Toxicokinetics , Young Adult
12.
J Clin Transl Sci ; 1(3): 184-191, 2017 Jun.
Article in English | MEDLINE | ID: mdl-29082032

ABSTRACT

INTRODUCTION: We describe the effectiveness of community outreach and engagement in supporting recruitment for the US National Children's Vanguard Study between 2009 and 2012. METHODS: Thirty-seven study locations used 1 of 4 strategies to recruit 18-49-year-old pregnant or trying to conceive women: (1) Initial Vanguard Study used household-based recruitment; (2) Direct Outreach emphasized self-referral; (3) Enhanced Household-Based Recruitment enhanced Initial Vanguard Study strategies; and (4) Provider-Based Recruitment recruited through healthcare providers. Outreach and engagement included advance letters, interactions with healthcare providers, participation in community events, contacts with community organizations, and media outreach. RESULTS: After 1-2 years, 41%-74% of 9844 study-eligible women had heard about the National Children's Vanguard Study when first approached. Women who heard were 1.5-3 times more likely to consent. Hearing via word-of-mouth or the media most frequently predicted consent. The more sources women heard from the higher the odds of consent. CONCLUSIONS: We conclude that tailored outreach and engagement facilitate recruitment in cohort studies.

13.
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
14.
Am J Primatol ; 79(2): 1-9, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27643479

ABSTRACT

Cortisol is a well-known glucocorticoid that can be used as a biomarker of hypothalamic-pituitary-adrenocortical activity. To explore basal cortisol physiology during pregnancy and infancy in Macaca nemestrina monkeys, hair was collected from a convenience sample of 22 healthy mother-infant dyads. Adult females were housed in pairs as part of a small breeding colony at the Washington National Primate Research Center and infants were reared in a specialized nursery. Maternal samples were collected from females during a pregnancy-detection ultrasound and immediately following labor and delivery. Infant samples were collected at birth, 20 days, 4, 6, 8, and 10 months of age. Hair cortisol concentrations (HCCs) were determined using an enzyme immunoassay in washed and ground hair samples. Like human mothers, macaque HCCs rose during pregnancy (paired t = 5.8, df = 16, P < 0.001). Maternal HCCs at pregnancy-detection (114.2 ± 12.07 picogram/milligram [pg/mg]) were highly predictive of maternal HCCs at delivery (144.8 ± 13.60 pg/mg), suggesting a trait-like quality (r = 0.90, P < 0.001). When maternal HCCs were viewed on a continuum, the absolute rise in cortisol over the course of pregnancy was significantly related to newborn HCCs (r = 0.55, P = 0.02). Infant birth HCCs (1,027.43 ± 97.95 pg/mg) were seven times higher than maternal HCCs at delivery (paired t = 19.1, df = 16, P < 0.001). Higher birth HCCs were strongly associated with larger decreases in infant hair cortisol until 6 months of postnatal age when infant HCCs converged on values indistinguishable from adults. Overall, study results demonstrate a marked degree of fetal cortisol exposure during the latter part of gestation and suggest that the rise in maternal cortisol over pregnancy may play an influential role on HCCs in the newborn.


Subject(s)
Hair/chemistry , Hydrocortisone/analysis , Macaca nemestrina , Animals , Female , Longitudinal Studies , Mothers , Pregnancy
16.
Public Health Nutr ; 19(10): 1795-803, 2016 07.
Article in English | MEDLINE | ID: mdl-26626702

ABSTRACT

OBJECTIVE: To investigate associations of maternal periconceptional shellfish, lean fish and fatty fish intake with risk of pregnancy complications. DESIGN: In this prospective cohort study, we collected information on intake of seafood subtypes using FFQ. We categorized seafood intake into frequencies of 1 servings/week. We ascertained gestational hypertension, pre-eclampsia, gestational diabetes and preterm birth diagnoses from medical records. Using generalized linear models with a log link, the Poisson family and robust standard errors, we estimated risk ratios and 95 % confidence intervals across seafood intake categories. SETTING: The Omega study, a study of risk factors for pregnancy complications among women recruited from prenatal clinics in Washington State, USA, 1996-2008. SUBJECTS: The current study included 3279 participants from the Omega study. RESULTS: Median (interquartile range) shellfish, lean fish and fatty fish intake was 0·3 (0-0·9), 0·5 (0-1·0) and 0·5 (0·1-1·0) servings/week, respectively. Lean fish intake of >1 servings/week (v. <0·2 servings/month) was associated with a 1·55-fold higher risk of preterm birth (95 % CI 1·04, 2·30) and was not associated with the other pregnancy complications. Higher intake of seafood (total or other subtypes) was not associated with pregnancy complications (separately or combined). CONCLUSIONS: Higher intake of lean fish, but not fatty fish or shellfish, was associated with a higher risk of preterm birth; these findings may have significance for preterm birth prevention. Studies of mechanisms and potential contributing factors (including seafood preparation and nutrient/contaminant content) are warranted.


Subject(s)
Diet , Maternal Nutritional Physiological Phenomena , Pregnancy Complications/epidemiology , Seafood , Adult , Animals , Diabetes, Gestational/epidemiology , Fatty Acids, Omega-3 , Female , Humans , Hypertension, Pregnancy-Induced/epidemiology , Pre-Eclampsia/epidemiology , Pregnancy , Premature Birth/epidemiology , Prospective Studies , Washington
17.
Paediatr Perinat Epidemiol ; 29(5): 376-87, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26147526

ABSTRACT

BACKGROUND: Previous reports of associations of maternal seafood intake with fetal growth were inconsistent. Further, little is known whether associations differ across seafood subtypes or fetal growth indices. METHODS: Among 3141 participants of the Omega study, a pregnancy cohort study, we investigated associations of periconceptional shell, lean, and fatty fish intake with fetal growth indices. We categorised food frequency questionnaire reported seafood intake into frequencies of: <0.2 servings/month, 0.2 servings/month -<0.5 servings/week, 0.5-1 servings/week, and >1 servings/week. We abstracted birthweight, birth length, and head circumference from medical records. Using generalised linear models with a log link, the Poisson family, and robust standard errors, we estimated relative risks and 95% confidence intervals (CI) for low birthweight (LBW, <2500 g) and linear regression models to estimate mean differences for continuous fetal growth indices across seafood intake categories. RESULTS: Medians (interquartile range) of shell, lean, and fatty fish intake were 0.3 (0-0.9), 0.5 (0-1.0), and 0.5 (0.1-1.0) servings/week, respectively. Lean fish intake of >1 servings/week (vs. <0.2 servings/month) was associated with a 2.2-fold higher risk of LBW (95% CI 1.2, 4.1). Shellfish intake of >1 servings/week (vs. <0.2 servings/month) was associated with a 0.6 kg/m(3) higher mean ponderal index (95% CI 0.0, 1.2 kg/m(3) ). There was no evidence for associations of total seafood or seafood subtype intake with other fetal growth indices. CONCLUSIONS: Higher intakes of lean fish and shellfish were associated with a higher risk of LBW and higher mean ponderal index, respectively. Findings highlight the importance of considerations of seafood subtype in similar investigations.


Subject(s)
Fetal Growth Retardation/etiology , Maternal Exposure/adverse effects , Seafood , Shellfish , Adult , Diet Records , Fatty Acids, Omega-3 , Female , Fetal Development , Fetal Growth Retardation/prevention & control , Humans , Infant, Low Birth Weight , Infant, Newborn , Maternal Nutritional Physiological Phenomena , Pregnancy , Pregnant Women , Prospective Studies , Seafood/adverse effects , Shellfish/adverse effects
18.
Environ Res ; 138: 74-81, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25701811

ABSTRACT

BACKGROUND: Recent evidence suggests that maternal cadmium (Cd) burden and fetal growth associations may vary by fetal sex. However, mechanisms contributing to these differences are unknown. OBJECTIVES: Among 24 maternal-infant pairs, we investigated infant sex-specific associations between placental Cd and placental genome-wide DNA methylation. METHODS: We used ANOVA models to examine sex-stratified associations of placental Cd (dichotomized into high/low Cd using sex-specific Cd median cutoffs) with DNA methylation at each cytosine-phosphate-guanine site or region. Statistical significance was defined using a false discovery rate cutoff (<0.10). RESULTS: Medians of placental Cd among females and males were 5 and 2 ng/g, respectively. Among females, three sites (near ADP-ribosylation factor-like 9 (ARL9), siah E3 ubiquitin protein ligase family member 3 (SIAH3), and heparin sulfate (glucosamine) 3-O-sulfotransferase 4 (HS3ST4) and one region on chromosome 7 (including carnitine O-octanoyltransferase (CROT) and TP5S target 1 (TP53TG1)) were hypomethylated in high Cd placentas. Among males, high placental Cd was associated with methylation of three sites, two (hypomethylated) near MDS1 and EVI1 complex locus (MECOM) and one (hypermethylated) near spalt-like transcription factor 1 (SALL1), and two regions (both hypomethylated, one on chromosome 3 including MECOM and another on chromosome 8 including rho guanine nucleotide exchange factor (GEF) 10 (ARHGEF10). Differentially methylated sites were at or close to transcription start sites of genes involved in cell damage response (SIAH3, HS3ST4, TP53TG1) in females and cell differentiation, angiogenesis and organ development (MECOM, SALL1) in males. CONCLUSIONS: Our preliminary study supports infant sex-specific placental Cd-DNA methylation associations, possibly accounting for previously reported differences in Cd-fetal growth associations across fetal sex. Larger studies are needed to replicate and extend these findings. Such investigations may further our understanding of epigenetic mechanisms underlying maternal Cd burden with suboptimal fetal growth associations.


Subject(s)
Birth Weight/drug effects , Cadmium/toxicity , DNA Methylation/drug effects , Environmental Exposure , Environmental Pollutants/toxicity , Placenta/metabolism , Female , Humans , Infant, Newborn , Male , Placenta/drug effects , Pregnancy , Sex Factors
19.
Environ Health Perspect ; 123(6): 579-89, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25690930

ABSTRACT

BACKGROUND: In the 1990s, the mercury-based preservative thimerosal was used in most pediatric vaccines. Although there are currently only two thimerosal-containing vaccines (TCVs) recommended for pediatric use, parental perceptions that vaccines pose safety concerns are affecting vaccination rates, particularly in light of the much expanded and more complex schedule in place today. OBJECTIVES: The objective of this study was to examine the safety of pediatric vaccine schedules in a non-human primate model. METHODS: We administered vaccines to six groups of infant male rhesus macaques (n = 12-16/group) using a standardized thimerosal dose where appropriate. Study groups included the recommended 1990s Pediatric vaccine schedule, an accelerated 1990s Primate schedule with or without the measles-mumps-rubella (MMR) vaccine, the MMR vaccine only, and the expanded 2008 schedule. We administered saline injections to age-matched control animals (n = 16). Infant development was assessed from birth to 12 months of age by examining the acquisition of neonatal reflexes, the development of object concept permanence (OCP), computerized tests of discrimination learning, and infant social behavior. Data were analyzed using analysis of variance, multilevel modeling, and survival analyses, where appropriate. RESULTS: We observed no group differences in the acquisition of OCP. During discrimination learning, animals receiving TCVs had improved performance on reversal testing, although some of these same animals showed poorer performance in subsequent learning-set testing. Analysis of social and nonsocial behaviors identified few instances of negative behaviors across the entire infancy period. Although some group differences in specific behaviors were reported at 2 months of age, by 12 months all infants, irrespective of vaccination status, had developed the typical repertoire of macaque behaviors. CONCLUSIONS: This comprehensive 5-year case-control study, which closely examined the effects of pediatric vaccines on early primate development, provided no consistent evidence of neurodevelopmental deficits or aberrant behavior in vaccinated animals.


Subject(s)
Immunization Schedule , Learning/drug effects , Neurodevelopmental Disorders/chemically induced , Social Behavior , Thimerosal/adverse effects , Vaccines/adverse effects , Animals , Case-Control Studies , Macaca mulatta , Male , Models, Animal , Neurotoxins/adverse effects , Neurotoxins/toxicity , Preservatives, Pharmaceutical/adverse effects , Preservatives, Pharmaceutical/pharmacology , Thimerosal/pharmacology , Vaccines/pharmacology
20.
Dev Neurosci ; 35(6): 491-503, 2013.
Article in English | MEDLINE | ID: mdl-24192275

ABSTRACT

BACKGROUND: Up to 65% of untreated infants suffering from moderate to severe hypoxic-ischemic encephalopathy (HIE) are at risk of death or major disability. Therapeutic hypothermia (HT) reduces this risk to approximately 50% (number needed to treat: 7-9). Erythropoietin (Epo) is a neuroprotective treatment that is promising as an adjunctive therapy to decrease HIE-induced injury because Epo decreases apoptosis, inflammation, and oxidative injury and promotes glial cell survival and angiogenesis. We hypothesized that HT and concurrent Epo will be safe and effective, improve survival, and reduce moderate-severe cerebral palsy (CP) in a term nonhuman primate model of perinatal asphyxia. METHODOLOGY: Thirty-five Macaca nemestrina were delivered after 15-18 min of umbilical cord occlusion (UCO) and randomized to saline (n = 14), HT only (n = 9), or HT+Epo (n = 12). There were 12 unasphyxiated controls. Epo (3,500 U/kg × 1 dose followed by 3 doses of 2,500 U/kg, or Epo 1,000 U/kg/day × 4 doses) was given on days 1, 2, 3, and 7. Timed blood samples were collected to measure plasma Epo concentrations. Animals underwent MRI/MRS and diffusion tensor imaging (DTI) at <72 h of age and again at 9 months. A battery of weekly developmental assessments was performed. RESULTS: UCO resulted in death or moderate-severe CP in 43% of saline-, 44% of HT-, and 0% of HT+Epo-treated animals. Compared to non-UCO control animals, UCO animals exhibit poor weight gain, behavioral impairment, poor cerebellar growth, and abnormal brain DTI. Compared to UCO saline, UCO HT+Epo improved motor and cognitive responses, cerebellar growth, and DTI measures and produced a death/disability relative risk reduction of 0.911 (95% CI -0.429 to 0.994), an absolute risk reduction of 0.395 (95% CI 0.072-0.635), and a number needed to treat of 2 (95% CI 2-14). The effects of HT+Epo on DTI included an improved mode of anisotropy, fractional anisotropy, relative anisotropy, and volume ratio as compared to UCO saline-treated infants. No adverse drug reactions were noted in animals receiving Epo, and there were no hematology, liver, or kidney laboratory effects. CONCLUSIONS/SIGNIFICANCE: HT+Epo treatment improved outcomes in nonhuman primates exposed to UCO. Adjunctive use of Epo combined with HT may improve the outcomes of term human infants with HIE, and clinical trials are warranted.


Subject(s)
Asphyxia/drug therapy , Erythropoietin/therapeutic use , Hypothermia/metabolism , Hypoxia-Ischemia, Brain/drug therapy , Animals , Asphyxia/metabolism , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Epoetin Alfa , Humans , Hypoxia-Ischemia, Brain/metabolism , Infant , Macaca nemestrina , Recombinant Proteins/therapeutic use , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...