Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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.

7.
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
8.
J Child Adolesc Psychopharmacol ; 20(6): 489-93, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21186967

ABSTRACT

OBJECTIVE: Atypical antipsychotic drugs are prescribed to young children for a number of symptoms, some with no Food and Drug Administration approval for children. Effects on growth of children have received little experimental study. We assessed the effects of two atypicals, risperidone and quetiapine, on growth, prolactin, and thyroid hormones of young pigtail macaques (macaca nemestrina), modeling potential effects on 4-8-year-old children. METHODS: Subjects were studied blindly after random assignment to risperidone (N = 10), quetiapine (N = 10), or placebo (N = 20). Four phases were studied: (1) predrug, 9-12 months of age; (2) low dose (risperidone 0.025 mg/kg, quetiapine 2 mg/kg), 13-16 months; (3) high dose (risperidone 0.05 mg/kg, quetiapine 4 mg/kg), 17-20 months; (4) postdrug, 21-24 months. Body weight was measured daily, skeletal dimension monthly, and bone mineralization and hormones bimonthly. RESULTS: Our primary result showed that, compared with placebo, neither drug had detrimental effects on body weight, skeletal dimensions, or thyroid hormones. However, in a transient effect, bone density was lower following low-dose risperidone than either quetiapine or placebo. In both drug phases, risperidone prolactin was higher than the other groups, which did not differ. The higher prolactin of the risperidone group may partially explain the bone density effect. CONCLUSION: This 16-month study of young, developing pigtail macaques given risperidone at doses from 0.025 to 0.05 mg/kg or quetiapine at doses from 2 to 4 mg/kg suggests that these drugs are safe for normal body weight and skeletal growth in young pigtail macaques given an adequate diet, although these drugs are known to cause significant weight gain and other metabolic changes in some children, adolescents, and adult humans. In addition, the results, although transient in our study, also suggest that research in children on bone mineralization effects of risperidone, and possibly other antipsychotic drugs, may be warranted.


Subject(s)
Antipsychotic Agents/adverse effects , Bone and Bones/drug effects , Dibenzothiazepines/adverse effects , Risperidone/adverse effects , Age Factors , Animals , Antipsychotic Agents/administration & dosage , Body Weight/drug effects , Bone Density/drug effects , Bone and Bones/metabolism , Child , Child, Preschool , Dibenzothiazepines/administration & dosage , Dose-Response Relationship, Drug , Humans , Macaca nemestrina , Male , Models, Animal , Prolactin/blood , Prolactin/drug effects , Quetiapine Fumarate , Random Allocation , Risperidone/administration & dosage , Thyroid Hormones/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...