Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Neurotoxicol Teratol ; 103: 107356, 2024.
Article in English | MEDLINE | ID: mdl-38719082

ABSTRACT

Developmental stress, including low socioeconomic status (SES), can induce dysregulation of the hypothalamic-pituitary-adrenal axis and result in long-term changes in stress reactivity. Children in lower SES conditions often experience more stress than those in other SES groups. There are multiple model systems of early environmental stress (EES), one of which is reduced cage bedding. Here we tested the effects of both prenatal and lactational EES in rats on a range of long-term behavioral and cognitive outcomes. There were persistent reductions in body weight in the EES rats in both sexes. The behavioral results showed no effects on learning and memory using tests of spatial learning or cognitive flexibility in the Morris water maze, egocentric learning in the Cincinnati water maze, or working memory in the radial-arm maze. There were no effects on basic open-field activity, elevated zero-maze, or forced swim test, but EES rats had reduced time in the dark side of the light/dark test. When rats were drug challenged in the open-field with d-amphetamine or MK-801, there were no differential responses to d-amphetamine, but the EES group under responded compared with the drug-induced hyperactivity in the control group in both males and females. The objective was to establish a developmental stress model that induced cognitive deficits and to the extent that this method did not cause such effects it was not the model we sought. However, the data showed several long-term effects of EES, including the reduced response to the irreversible NMDA antagonist MK-801. This effect merits further investigation.


Subject(s)
Rats, Sprague-Dawley , Stress, Psychological , Animals , Female , Male , Rats , Stress, Psychological/psychology , Pregnancy , Maze Learning/drug effects , Behavior, Animal/drug effects , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/physiopathology , Cognition/drug effects , Dizocilpine Maleate/pharmacology , Disease Models, Animal
2.
Int J Dev Neurosci ; 80(3): 163-174, 2020 May.
Article in English | MEDLINE | ID: mdl-32043612

ABSTRACT

Children exposed to methamphetamine (MA) in utero have cognitive deficits. MA administration in rats for 5-10 days between postnatal days (P)6 and 20 produces cognitive deficits. The purpose of this study was to determine if extending MA administration by 5 days within P6-20 would exacerbate allocentric (Morris water maze) and egocentric (Cincinnati water maze) learning deficits. Sprague Dawley female and male offspring (split-litter design) were administered saline (SAL) or MA (10 mg/kg) four times daily from P6 to 20 to create four groups: (a) SAL from P6 to 20, (b) MA from P6 to 20 (MA6-20), (c) MA from P6 to 15 (MA6-15), or (d) MA from P11 to 20 (MA11-20); the latter groups received saline on days they did not receive MA. Egocentric, allocentric, and conditioned freezing tests began on P60. The MA6-15 and MA6-20 groups showed egocentric deficits, all MA groups had allocentric deficits but no differences in conditioned freezing compared with SAL controls. The MA6-15 and MA6-20 groups had similar deficits in learning and memory that were larger than in the MA11-20 group. Learning in both mazes was sex dependent, but no interactions with MA were found. The data demonstrate that extending the exposure period of MA beyond the sensitive periods (P6-15 and P11-20) did not exacerbate the cognitive deficits.


Subject(s)
Central Nervous System Stimulants/pharmacology , Maze Learning/drug effects , Memory/drug effects , Methamphetamine/pharmacology , Spatial Navigation/drug effects , Animals , Animals, Newborn , Critical Period, Psychological , Female , Male , Rats , Rats, Sprague-Dawley
3.
J Toxicol Environ Health A ; 82(2): 99-112, 2019.
Article in English | MEDLINE | ID: mdl-30652531

ABSTRACT

Iron (Fe) deficiency (FeD) and manganese (Mn) overexposure (MnOE) may result in several neurological alterations in the nervous system. Iron deficiency produces unique neurological deficits due to its elemental role in central nervous system (CNS) development and myelination, which might persist after normalization of Fe in the diet. Conversely, MnOE is associated with diverse neurocognitive deficits. Despite these well-known neurotoxic effects on the CNS, the influence of FeD and MnOE on the peripheral nervous system (PNS) remains poorly understood. The aim of the present investigation was to examine the effects of developmental FeD and MnOE or their combination on the sciatic nerve of young and adult rats. The parameters measured included divalent metal transporter 1 (DMT1), transferrin receptor (TfR), myelin basic protein (MBP) and peripheral myelin protein 22 (PMP22) expression, as well as Fe levels in the nerve. Our results showed that FeD produced a significant reduction in MBP and PMP22 content at P29, which persisted at P60 after Fe-sufficient diet replenishment regardless of Mn exposure levels. At P60 MnOE significantly increased sciatic nerve Fe content and DMT1 expression. However, the combination of FeD and MnOE produced no marked motor skill impairment. Evidence indicates that FeD appears to hinder developmental peripheral myelination, while MnOE may directly alter Fe homeostasis. Further studies are required to elucidate the interplay between these pathological conditions.


Subject(s)
Gene Expression/drug effects , Iron Deficiencies , Manganese/adverse effects , Motor Activity/drug effects , Peripheral Nerves/drug effects , Age Factors , Animals , Male , Peripheral Nerves/chemistry , Rats , Rats, Sprague-Dawley
4.
PLoS One ; 13(6): e0199213, 2018.
Article in English | MEDLINE | ID: mdl-29953483

ABSTRACT

Although prenatal alcohol exposure (PAE) reduces offspring growth, it may increase obesity risk at adolescence. Animal models of PAE display glucose intolerance and increased adiposity, suggesting that PAE causes metabolic reprogramming. We tested this hypothesis in a mouse model of binge PAE, wherein pregnant C57Bl/6J females received 3 g/kg alcohol (ETOH) daily from gestational day 12.5 to 17.5; maltodextrin (MD) and medium chain triglycerides (MCT) served as isocaloric nutritional controls, and sham (H2O) treatment controlled for gavage stress. Our comprehensive assessment quantified body composition, energy expenditure, glucose tolerance, and cardiovascular function in offspring at age 17 weeks. Although ETOH pups were initially lighter than all other groups, they did not have a unique obesogenic phenotype. Instead, a similar obesogenic phenotype emerged in all three caloric groups (MCT, MD, ETOH), such that caloric groups had greater post-weaning weight gain (both sexes), reduced gonadal fat weight (males), and reduced glucose clearance (males) compared against H2O offspring. PAE did not affect body composition, respiratory exchange ratio, metabolic adaption to high-fat or low-fat diet, eating behavior, and blood pressure, and ETOH values did not differ from those obtained from isocaloric controls. Exposure to a higher alcohol dose (4.5 g/kg) or a high-fat (60%) diet did not exacerbate differences in body composition or glucose tolerance. "PAE-specific" effects on postnatal growth, glucose tolerance, adiposity, or hypertension only emerged when PAE offspring were compared just against H2O controls, or against MD controls. We conclude that prior reports of obesity and glucose intolerance in adult PAE offspring reflect the contribution of added gestational calories, and not alcohol's pharmacologic action. Results suggest that the increased adiposity risk in FASD is not caused by metabolic reprogramming, and instead originates from behavioral, medication, and/or dietary practices. This study highlights the importance of appropriate dietary controls in nutritional studies of PAE.


Subject(s)
Alcohol Drinking/adverse effects , Fetal Alcohol Spectrum Disorders/etiology , Maternal Exposure , Metabolic Syndrome/etiology , Metabolic Syndrome/metabolism , Prenatal Exposure Delayed Effects , Adiposity , Animals , Biomarkers , Body Weight , Diet, High-Fat/adverse effects , Disease Models, Animal , Energy Metabolism , Feeding Behavior , Female , Fetal Development/drug effects , Glucose Intolerance , Hemodynamics , Male , Mice , Pregnancy
5.
Neurotoxicol Teratol ; 67: 50-64, 2018.
Article in English | MEDLINE | ID: mdl-29631003

ABSTRACT

Developmental stress, including low socioeconomic status (SES), can induce dysregulation of the hypothalamic-pituitary-adrenal axis and result in long-term changes in stress reactivity. Children in lower SES households experience more stress and are more likely to be exposed to environmental neurotoxins such as lead (Pb) and manganese (Mn) than children in higher SES households. Co-exposure to stress, Pb, and Mn during early development may increase the risk of central nervous system dysfunction compared with unexposed children. To investigate the potential interaction of these factors, Sprague-Dawley rats were bred, and litters born in-house were culled on postnatal day (P)1 to 6 males and 6 females. One male and female within each litter were assigned to one of the following groups: 0 (vehicle), 10 mg/kg Pb, 100 mg/kg Mn, or 10 mg/kg Pb + 100 mg/kg Mn (PbMn), water gavage, and handled only from P4-28 with half the litters reared in cages with standard bedding (29 litters) and half with no bedding (Barren; 27 litters). Mn and PbMn groups had decreased anxiety, reduced acoustic startle, initial open-field hypoactivity, increased activity following (+)-methamphetamine, deficits in egocentric learning in the Cincinnati water maze (CWM), and deficits in latent inhibition conditioning. Pb increased anxiety and reduced open-field activity. Barren-reared rats had decreased anxiety, CWM deficits, increased startle, and initial open-field hyperactivity. Mn, PbMn, Pb Barren-reared groups had impaired Morris water maze performance. Pb altered neostriatal serotonin and norepinephrine, Mn increased hippocampal serotonin in males, Mn + Barren-rearing increased neostriatal serotonin, and Barren-rearing decreased neostriatal dopamine in males. At the doses used here, most effects were in the Mn and PbMn groups. Few interactions between Mn, Pb, and rearing stress were found, indicating that the interaction of these three variables is not as impactful as hypothesized.


Subject(s)
Behavior, Animal/drug effects , Dopamine/metabolism , Housing, Animal , Lead/toxicity , Manganese/toxicity , Norepinephrine/metabolism , Serotonin/metabolism , Animals , Drug Synergism , Female , Male , Neostriatum/metabolism , Rats
6.
ACS Chem Neurosci ; 8(7): 1479-1489, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28287691

ABSTRACT

Severe stress potentiates methamphetamine (MA) neurotoxicity. However, whether moderate stress increases or decreases the neurotoxic effects of MA is unknown. We assessed the effects of MA (4 × 10 mg/kg at 2 h intervals) in combination with prior barren-cage housing in adult male Sprague-Dawley rats on monoamines and glial fibrillary acid protein (GFAP) in one cohort and spatial learning and memory in the Morris water maze in another cohort. MA reduced dopamine (DA) and serotonin (5-HT) in the neostriatum and nucleus accumbens, 5-HT in the hippocampus, and increased GFAP in neostriatum and nucleus accumbens compared with saline controls. In neostriatum, barren-cage housing protected against MA-induced increases in GFAP, but it did not prevent DA and 5-HT reductions, although it did increase hippocampal norepinephrine. MA impaired spatial learning during acquisition, reversal, and shift phases and impaired reference memory on reversal and shift probe trials. Barren-cage housing enhanced performance during acquisition but not during reversal or shift or on probe trials. The data indicate that prior barren-cage housing moderates MA-induced neostriatal astrogliosis and initial spatial learning, but has no protective effect when the platform is smaller and relocated and therefore requires cognitive flexibility in relearning.


Subject(s)
Housing, Animal , Maze Learning/drug effects , Methamphetamine/toxicity , Spatial Memory/drug effects , Stress, Psychological/metabolism , Animals , Body Temperature/drug effects , Corticosterone/metabolism , Dopamine/metabolism , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Maze Learning/physiology , Motor Activity/drug effects , Neostriatum/drug effects , Neostriatum/metabolism , Norepinephrine/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Rats, Sprague-Dawley , Reversal Learning/drug effects , Reversal Learning/physiology , Serotonin/metabolism , Spatial Memory/physiology , Swimming
7.
Neurotoxicol Teratol ; 59: 16-26, 2017.
Article in English | MEDLINE | ID: mdl-27756629

ABSTRACT

Manganese (Mn) is an essential element but neurotoxic at higher exposure levels. The effects of Mn overexposure (MnOE) on hippocampal and striatal-dependent learning and memory in rats were tested in combination with iron deficiency (FeD) and developmental stress that often co-occur with MnOE. Moderate FeD affects up to 15% of U.S. children and developmental stress is common in lower socio-economic areas where MnOE occurs. Pregnant Sprague-Dawley rats and their litters were housed in cages with or without (barren cage (BAR)) standard bedding from embryonic day (E)7 to postnatal day (P)28. Dams were fed a 90% FeD or iron sufficient (FeS) diet from E15-P28. Within each litter, separate offspring were treated with 100mg/kg Mn (MnOE) or vehicle (VEH) by gavage on alternate days from P4-28. Offspring were tested as adults in the Morris and Cincinnati water mazes. FeD and developmental stress interactively impaired spatial learning in the Morris water maze. Developmental stress and MnOE impaired learning and memory in both mazes. MnOE resulted in reduced CA1 hippocampal long-term potentiation (LTP) and increased levels of α-synuclein. Preweaning MnOE resulted in cognitive deficits on multiple domains of learning and memory accompanied by impaired LTP and α-synuclein changes, effects worsened by developmental stress.


Subject(s)
Iron Deficiencies , Manganese/toxicity , Maze Learning/drug effects , Memory/drug effects , Stress, Psychological/psychology , Animals , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiopathology , Female , Long-Term Potentiation/physiology , Male , Pregnancy , Rats , alpha-Synuclein/metabolism
8.
Neurotoxicol Teratol ; 56: 55-67, 2016.
Article in English | MEDLINE | ID: mdl-27302314

ABSTRACT

Manganese (Mn) is an essential element but neurotoxic at higher exposures, however, Mn exposure seldom occurs in isolation. It often co-occurs in populations with inadequate dietary iron (Fe) and limited resources that result in stress. Subclinical FeD affects up to 15% of U.S. children and exacerbates Mn toxicity by increasing Mn bioavailability. Therefore, we investigated Mn overexposure (MnOE) in rats in combination with Fe deficiency (FeD) and developmental stress, for which we used barren cage rearing. For barren cage rearing (BAR), rats were housed in cages with a wire grid floor or standard bedding material (STD) from embryonic day (E)7 through postnatal day (P)28. For FeD, dams were fed a 90% Fe-deficient NIH-07 diet from E15 through P28. Within each litter, different offspring were treated with 100mg/kg Mn (MnOE) or vehicle (VEH) by gavage every other day from P4-28. Behavior was assessed at two ages and consisted of: open-field, anxiety tests, acoustic startle response (ASR) with prepulse inhibition (PPI), sociability, sucrose preference, tapered beam crossing, and the Porsolt's forced swim test. MnOE had main effects of decreasing activity, ASR, social preference, and social novelty. BAR and FeD transiently modified MnOE effects. BAR groups weighed less and showed decreased anxiety in the elevated zero maze, had increased ASR and decreased PPI, and exhibited reduced sucrose preference compared with the STD groups. FeD animals also weighed less and had increased slips on the tapered beam. Most of the monoamine effects were dopaminergic and occurred in the MnOE groups. The results showed that Mn is a pervasive developmental neurotoxin, the effects of which are modulated by FeD and/or BAR cage rearing.


Subject(s)
Behavior, Animal , Biogenic Monoamines/metabolism , Hippocampus/metabolism , Iron Deficiencies , Manganese/toxicity , Neostriatum/metabolism , Stress, Psychological , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Anxiety/chemically induced , Dopamine/metabolism , Female , Hematocrit , Male , Motor Activity , Norepinephrine/metabolism , Prepulse Inhibition , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Social Behavior
9.
Neurotox Res ; 30(2): 199-212, 2016 08.
Article in English | MEDLINE | ID: mdl-27003940

ABSTRACT

The nucleus accumbens (Nacc) and medial prefrontal cortex (mPFC) receive dopaminergic innervation from the ventral tegmental area and are involved in learning. Male rats with 6-hydroxydopamine (6-OHDA)-induced dopaminergic and noradrenergic reductions in the Nacc or mPFC were tested for allocentric and egocentric learning to determine their role in these forms of neuroplasticity. mPFC dopaminergic and noradrenergic reductions did not result in changes to either type of learning or memory. Nacc dopaminergic and noradrenergic reductions resulted in allocentric learning and memory deficits in the Morris water maze (MWM) on acquisition, reversal, and probe trials. MWM cued performance was also affected, but straight-channel swim times and swim speed during hidden platform trials in the MWM were not affected. Nacc dopaminergic and noradrenergic reductions also impaired egocentric learning in the Cincinnati water maze (CWM). Nacc-lesioned animals tested in the CWM in an alternate path through the maze were not significantly affected. 6-OHDA injections in the Nacc resulted in 63 % dopamine and 62 % norepinephrine reductions in the Nacc and 23 % reductions in adjacent dorsal striatum. 6-OHDA injections in the mPFC resulted in 88 % reductions in dopamine and 59 % reductions in norepinephrine. Hence, Nacc dopamine and/or norepinephrine play a role in egocentric and allocentric learning and memory, while mPFC dopamine and norepinephrine do not.


Subject(s)
Dopamine/deficiency , Maze Learning/physiology , Nucleus Accumbens/metabolism , Oxidopamine/toxicity , Prefrontal Cortex/metabolism , Spatial Navigation/physiology , Animals , Cohort Studies , Learning Disabilities/metabolism , Male , Memory Disorders/metabolism , Models, Animal , Random Allocation , Rats, Sprague-Dawley , Spatial Memory/physiology
10.
Neurotoxicology ; 54: 22-33, 2016 05.
Article in English | MEDLINE | ID: mdl-26943976

ABSTRACT

The level of lead (Pb) exposure in children has decreased dramatically since restrictions on its use were implemented. However, even with restrictions, children are exposed to Pb and still present with cognitive and behavioral deficits. One prominent aspect of the exposome of these children is that many come from low social economic status (SES) conditions, and low SES is associated with stress. In order to compare the combined effects of early stress and Pb, Sprague-Dawley rats were exposed to vehicle or Pb either alone or in combination with maternal separation stress during brain development (i.e., postnatal day (P)4-P11, P19, or P28). Maternally separated/isolated pups had lower body and thymus weights during exposure and had increased levels of blood Pb compared with vehicle controls. Isolation, but not Pb, affected the response to an acute stressor (standing in shallow water) when assessed on P19 and P29, but not earlier on P11. Interactions of Pb and isolation were found on monoamines in the neostriatum, hippocampus, and hypothalamus on turnover but not on levels, and most changes were on dopamine turnover. Isolation had greater short-term effects than Pb. Interactions were dependent on age, sex, and acute stress.


Subject(s)
Biogenic Monoamines/blood , Corticosterone/blood , Lead/adverse effects , Lead/blood , Maternal Deprivation , Prenatal Exposure Delayed Effects/blood , Stress, Psychological/physiopathology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Body Weight/drug effects , Female , Male , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Rats , Rats, Sprague-Dawley , Stress, Psychological/blood , Thymus Gland/drug effects , Thymus Gland/pathology
11.
J Pediatr ; 169: 194-200.e1, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26608087

ABSTRACT

OBJECTIVE: To compare the eating behaviors and nutrition-related concerns in children with fetal alcohol spectrum disorder (FASD) with those in typically developing children. STUDY DESIGN: A survey that assessed eating behaviors was completed between October 2013 and May 2014 by the caregivers of children screened for FASD at the University of Minnesota's Fetal Alcohol Spectrum Disorders Program, and typically developing children recruited from that clinic or from the Research Participation Core of the Waisman Center, University of Wisconsin. RESULTS: Compared with controls (N = 81), children with FASD (N = 74) had delayed acquisition of self-feeding behavior (P < .001) and solid food introduction (P < .001). Impaired satiety was common and independent of medication use: 23.0% were never full/satisfied, 31.1% snacked constantly, and 27.0% concealed food (all P ≤ .002). They consumed the equivalent of an additional meal/snack daily (P < .01). Children with FASD were more likely to have a past diagnosis of underweight (P < .001). Mean body mass index was significantly reduced for males (P = .009) but not females (P = .775) with FASD, and only 2 children with FASD were currently underweight. Children with FASD were more physically active (P < .01). CONCLUSIONS: Abnormal eating patterns are common in children with FASD and may contribute to their delayed growth and nutritional inadequacies. Their poor satiety may reflect poor impulse control. Children with FASD may benefit from diet counseling. Conversely, some children with hyperphagia may warrant referral for FASD screening.


Subject(s)
Feeding Behavior , Fetal Alcohol Spectrum Disorders , Child , Cross-Sectional Studies , Female , Fetal Alcohol Spectrum Disorders/physiopathology , Humans , Male
12.
Toxicol Rep ; 2: 1046-1056, 2015.
Article in English | MEDLINE | ID: mdl-26295019

ABSTRACT

Manganese overexposure (MnOE) can be neurotoxic. In humans this can occur through occupational exposure, air or water contamination, well water, soy milk, and some baby formulas. In children MnOE has been associated with cognitive and behavioral deficits. The effects of MnOE may be modified by factors such as iron status. We hypothesized that developmental MnOE would be exacerbated by iron deficiency. A diet with a 90% decrease in iron (FeD) was given to gravid female rats starting on embryonic day 15 and continued through postnatal day (P)28. Mn (100 mg/kg) or vehicle (VEH) was administered by gavage every other day from P4-28. Metal transporters and receptors (divalent metal transporter-1 (DMT1), transferrin (Tf), transferrin receptor (TfR), and zip8 (zrt8)) were quantified in brain at P28. These markers were increased but the changes were specific: MnOE increased TfR and decreased Tf in hippocampus, whereas FeD increased TfR in neostriatum and increased TfR and DMT1 in the hippocampus, and the combination increased TfR in neostriatum (zip8 was unaffected). Identically treated animals were tested behaviorally at P29 or P60. The combination of FeD+MnOE increased head dips in an elevated zero-maze, reversed deficits in sucrose preference induced by MnOE alone, and increased spontaneous locomotion in an open-field. Rats were also evaluated for changes in locomotor activity after challenge with (±)-fenfluramine (FEN, a 5-HT agonist: 5 mg/kg), MK-801 (MK801, an NMDA antagonist: 0.2 mg/kg), or (+)amphetamine (AMPH, a dopamine agonist: 1 mg/kg). Compared with VEH animals, MnOE animals were more hyperactive after fenfluramine, amphetamine, or MK-801, regardless of FeD exposure. The results indicate persistent effects of developmental MnOE on brain and behavior but few interactions with dietary iron deficiency.

13.
Neurobiol Learn Mem ; 118: 55-63, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25451306

ABSTRACT

Both egocentric route-based learning and spatial learning, as assessed by the Cincinnati water maze (CWM) and Morris water maze (MWM), respectively, are impaired following an 80% dopamine (DA) loss in the neostriatum after 6-hydroxydopamine (6-OHDA) administration in rats. The dorsolateral striatum (DLS) and the dorsomedial striatum (DMS) are implicated in different navigational learning types, namely the DLS is implicated in egocentric learning while the DMS is implicated in spatial learning. This experiment tested whether selective DA loss through 6-OHDA lesions in the DMS or DLS would impair one or both types of navigation. Both DLS and DMS DA loss significantly impaired route-based CWM learning, without affecting spatial or cued MWM performance. DLS 6-OHDA lesions produced a 75% DA loss in this region, with no changes in other monoamine levels in the DLS or DMS. DMS 6-OHDA lesions produced a 62% DA loss in this region, without affecting other monoamine levels in the DMS or DLS. The results indicate a role for DA in DLS and DMS regions in route-based egocentric but not spatial learning and memory. Spatial learning deficits may require more pervasive monoamine reductions within each region before deficits are exhibited. This is the first study to implicate DLS and DMS DA in route-based egocentric navigation.


Subject(s)
Dopamine/physiology , Maze Learning/physiology , Neostriatum/physiology , Spatial Navigation/physiology , Animals , Biogenic Monoamines/analysis , Biogenic Monoamines/chemical synthesis , Dopamine/chemical synthesis , Male , Maze Learning/drug effects , Neostriatum/chemistry , Oxidopamine/administration & dosage , Rats , Rats, Sprague-Dawley , Spatial Navigation/drug effects
14.
Int J Dev Neurosci ; 35: 7-15, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24594360

ABSTRACT

Ventriculomegaly occurs when there is imbalance between creation and absorption of cerebrospinal fluid (CSF); even when treated, long-term behavioral changes occur. Kaolin injection in the cisterna magna of rats produces an obstruction of CSF outflow and models one type of hydrocephalus. Previous research with this model shows that neonatal onset has mixed effects on Morris water maze (MWM) and motoric performance; we hypothesized that this might be because the severity of ventricular enlargement was not taken into consideration. In the present experiment, rats were injected with kaolin or saline on postnatal day (P)21 and analyzed in subgroups based on Evan's ratios (ERs) of the severity of ventricular enlargement at the end of testing to create 4 subgroups from least to most severe: ER0.4-0.5, ER0.51-0.6, ER0.61-0.7, and ER0.71-0.82, respectively. Locomotor activity (dry land and swimming), acoustic startle with prepulse inhibition (PPI), and MWM performance were tested starting on P28 (122cm maze) and again on P42 (244cm maze). Kaolin-treated animals weighed significantly less than controls at all times. Differences in locomotor activity were seen at P42 but not P28. On P28 there was an increase in PPI for all but the least severe kaolin-treated group, but no difference at P42 compared with controls. In the MWM at P28, all kaolin-treated groups had longer path lengths than controls, but comparable swim speeds. With the exception of the least severe group, probe trial performance was worse in the kaolin-treated animals. On P42, only the most severely affected kaolin-treated group showed deficits compared with control animals. This group showed no MWM learning and no memory for the platform position during probe trial testing. Swim speed was unaffected, indicating motor deficits were not responsible for impaired learning and memory. These findings indicate that kaolin-induced ventriculomegaly in rats interferes with cognition regardless of the final enlargement of the cerebral ventricles, but final size critically determines whether lasting locomotor, learning, and memory impairments occur.


Subject(s)
Disease Models, Animal , Hydrocephalus/physiopathology , Learning Disabilities/physiopathology , Memory Disorders/physiopathology , Mental Disorders/physiopathology , Movement Disorders/physiopathology , Animals , Chronic Disease , Hydrocephalus/chemically induced , Kaolin , Learning Disabilities/chemically induced , Male , Maze Learning , Memory Disorders/chemically induced , Mental Disorders/chemically induced , Rats , Rats, Sprague-Dawley , Weaning
15.
Toxicol Rep ; 1: 1046-1061, 2014.
Article in English | MEDLINE | ID: mdl-25574457

ABSTRACT

Developmental exposure to manganese (Mn) or stress can each be detrimental to brain development. Here, Sprague-Dawley rats were exposed to two housing conditions and Mn from postnatal day (P)4-28. Within each litter two males and 2 females were assigned to the following groups: 0 (vehicle), 50, or 100 mg/kg Mn by oral gavage every other day. Half the litters were reared in cages with standard bedding and half with no bedding. One pair/group in each litter had an acute shallow water stressor before tissue collection (i.e., standing in shallow water). Separate litters were assessed at P11, 19, or 29. Mn-treated rats raised in standard cages showed no change in baseline corticosterone but following acute stress increased more than controls on P19; no Mn effects were seen on P11 or P29. Mn increased neostriatal dopamine in females at P19 and norepinephrine at P11 and P29. Mn increased hippocampal dopamine at P11 and P29 and 5-HT at P29 regardless of housing or sex. Mn had no effect on hypothalamic dopamine, but increased norepinephrine in males at P29 and 5-HT in males at all ages irrespective of rearing condition. Barren reared rats showed no or opposite effects of Mn, i.e., barren rearing + Mn attenuated corticosterone increases to acute stress. Barren rearing also altered the Mn-induced changes in dopamine and norepinephrine in the neostriatum, but not in the hippocampus. Barren rearing caused a Mn-associated increase in hypothalamic dopamine at P19 and P29 not seen in standard reared Mn-treated groups. Developmental Mn alters monoamines and corticosterone as a function of age, stress (acute and chronic), and sex.

16.
Int J Neuropsychopharmacol ; 16(6): 1383-94, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23308402

ABSTRACT

We previously showed that developmental 3,4-methylenedioxymethamphetamine (MDMA) treatment induces long-term spatial and egocentric learning and memory deficits and serotonin (5-HT) reductions. During brain development, 5-HT is a neurotrophic factor influencing neurogenesis, synaptogenesis, migration, and target field organization. MDMA (10 mg/kg × 4/d at 2 h intervals) given on post-natal day (PD) 11-20 in rats (a period of limbic system development that approximates human third trimester brain development) induces 50% reductions in 5-HT during treatment and 20% reductions when assessed as adults. To determine whether the 5-HT reduction is responsible for the cognitive deficits, we used citalopram (Cit) pretreatment to inhibit the effects of MDMA on 5-HT reuptake in a companion study. Cit attenuated MDMA-induced 5-HT reductions by 50% (Schaefer et al., 2012). Here we tested whether Cit (5 or 7.5 mg/kg × 2/d) pretreatment attenuates the cognitive effects of MDMA. Within each litter, different offspring were treated on PD11-20 with saline (Sal) + MDMA, Cit + MDMA, Cit + Sal or Sal + Sal. Neither spatial nor egocentric learning/memory was improved by Cit pretreatment. Unexpectedly, Cit + Sal (at both doses) produced spatial and egocentric learning deficits as severe as those caused by Sal + MDMA. These are the first data showing cognitive deficits resulting from developmental exposure to a selective serotonin reuptake inhibitor. These data indicate the need for further research on the long-term safety of antidepressants during pregnancy.


Subject(s)
Citalopram/toxicity , Cognition Disorders/chemically induced , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Serotonin Agents/toxicity , Animals , Animals, Newborn , Body Weight/drug effects , Cognition Disorders/physiopathology , Disease Models, Animal , Female , Locomotion/drug effects , Male , Maze Learning/drug effects , Rats , Rats, Sprague-Dawley , Swimming/psychology
17.
Neurotoxicol Teratol ; 35: 34-45, 2013.
Article in English | MEDLINE | ID: mdl-23314114

ABSTRACT

Although maternal cigarette smoking during pregnancy is a well-documented risk factor for a variety of adverse pregnancy outcomes, how prenatal cigarette smoke exposure affects postnatal neurobehavioral/cognitive development remains poorly defined. In order to investigate the cause of an altered behavioral phenotype, mice developmentally exposed to a paradigm of 'active' maternal cigarette smoke is needed. Accordingly, cigarette smoke exposed (CSE) and air-exposed C57BL/6J mice were treated for 6h per day in paired inhalation chambers throughout gestation and lactation and were tested for neurobehavioral effects while controlling for litter effects. CSE mice exhibited less than normal anxiety in the elevated zero maze, transient hypoactivity during a 1h locomotor activity test, had longer latencies on the last day of cued Morris water maze testing, impaired hidden platform learning in the Morris water maze during acquisition, reversal, and shift trials, and impaired retention for platform location on probe trials after reversal but not after acquisition or shift. CSE mice also showed a sexually dimorphic response in central zone locomotion to a methamphetamine challenge (males under-responded and females over-responded), and showed reduced anxiety in the light-dark test by spending more time on the light side. No differences on tests of marble burying, acoustic startle response with prepulse inhibition, Cincinnati water maze, matching-to-sample Morris water maze, conditioned fear, forced swim, or MK-801-induced locomotor activation were found. Collectively, the data indicate that developmental cigarette smoke exposure induces subnormal anxiety in a novel environment, impairs spatial learning and reference memory while sparing other behaviors (route-based learning, fear conditioning, and forced swim immobility). The findings add support to mounting evidence that developmental cigarette smoke exposure has long-term adverse effects on brain function.


Subject(s)
Behavior, Animal/physiology , Developmental Disabilities/chemically induced , Developmental Disabilities/physiopathology , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/physiopathology , Tobacco Products/toxicity , Age Factors , Animals , Animals, Newborn , Behavior, Animal/drug effects , Body Weight/drug effects , Conditioning, Psychological/radiation effects , Fear/drug effects , Fear/psychology , Female , Inhibition, Psychological , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Pregnancy , Reflex, Acoustic/drug effects , Reflex, Acoustic/physiology , Time Factors
18.
Int J Neuropsychopharmacol ; 16(2): 377-91, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22391043

ABSTRACT

Neonatal exposure to (+)-methamphetamine (Meth) results in long-term behavioural abnormalities but its developmental mechanisms are unknown. In a series of experiments, rats were treated from post-natal days (PD) 11-20 (stage that approximates human development from the second to third trimester) with Meth or saline and assessed using locomotor activity as the readout following pharmacological challenge doses with dopamine, serotonin and glutamate agonists or antagonists during adulthood. Exposure to Meth early in life resulted in an exaggerated adult locomotor hyperactivity response to the dopamine D1 agonist SKF-82958 at multiple doses, a high dose only under-response activating effect of the D2 agonist quinpirole, and an exaggerated under-response to the activating effect of the N-methyl-d-aspartic acid (NMDA) receptor antagonist, MK-801. No change in locomotor response was seen following challenge with the 5-HT releaser p-chloroamphetamine or the 5-HT2/3 receptor agonist, quipazine. These are the first data to show that PD 11-20 Meth exposure induces long-lasting alterations to dopamine D1, D2 and glutamate NMDA receptor function and may suggest how developmental Meth exposure leads to many of its long-term adverse effects.


Subject(s)
Dopamine Agents/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/pharmacology , Methamphetamine/pharmacology , Motor Activity/drug effects , Analysis of Variance , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Drug Interactions , Female , Male , Quipazine/pharmacology , Rats , Rats, Sprague-Dawley , Serotonin Receptor Agonists/pharmacology , Time Factors
19.
Int J Neuropsychopharmacol ; 15(6): 811-24, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21733225

ABSTRACT

Previous findings showed allocentric and egocentric learning deficits in rats after MDMA treatment from postnatal days (PD) 11-20 but not after treatment from PD 1-10. Shorter treatment periods (PD 1-5, 6-10, 11-15, or 16-20) resulted in allocentric learning deficits averaged across intervals but not for any interval individually and no egocentric learning deficits individually or collectively. Whether this difference was attributable to treatment length or age at the start of treatment was unclear. In the present experiment rat litters were treated on PD 1-10, 6-15, or 11-20 with 0, 10, or 15 mg/kg MDMA q.i.d. at 2-h intervals. Two male/female pairs/litter received each treatment. One pair/litter received acoustic startle with prepulse inhibition, straight channel swimming, Cincinnati water maze (CWM), and conditioned fear in a latent inhibition paradigm. The other pair/litter received locomotor activity, straight channel swimming, Morris water maze (MWM), and locomotor activity retest with MK-801 challenge. MDMA impaired CWM learning following PD 6-15 or 11-20 exposure. In MWM acquisition, all MDMA-treated groups showed impairment. During reversal and shift, the PD 6-15 and PD 11-20 MDMA-treated groups were significantly impaired. Reductions in locomotor activity were most evident after PD 6-15 treatment while increases in acoustic startle were most evident after PD 1-10 treatment. After MK-801 challenge, MDMA-treated offspring showed less locomotion compared to controls. Region-specific changes in brain monoamines were also observed but were not significantly correlated with behavioural changes. The results show that PD 11-20 exposure to MDMA caused the largest long-term cognitive deficits followed by PD 6-15 exposure with PD 1-10 exposure least affected. Other effects, such as those upon MK-801-stimulated locomotion showed greatest effects after PD 1-10 MDMA exposure. Hence, each effect has a different window of developmental susceptibility.


Subject(s)
Aging/drug effects , Aging/physiology , Behavior, Animal/drug effects , Biogenic Monoamines/metabolism , Hallucinogens/pharmacology , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Acoustic Stimulation , Animals , Animals, Newborn , Body Weight/drug effects , Conditioning, Psychological/drug effects , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Fear/drug effects , Female , Inhibition, Psychological , Male , Maze Learning/drug effects , Mortality , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Reflex, Startle/drug effects , Swimming/psychology
20.
Neurosci Lett ; 502(3): 152-6, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21798318

ABSTRACT

Methylphenidate (MPD) is the most prescribed drug for attention deficit hyperactivity disorder. Licit and illicit use also occurs during pregnancy, however the effects from this use on offspring development are unknown. To model late gestational exposure, Sprague-Dawley litters were treated with 0, 5, 10, 20, or 30mg/kg×4/day every 2h with MPD on postnatal days 11-20 (within-litter design; days chosen to be comparable to human third trimester brain development). During treatment, body weights were decreased in MPD-treated groups; weight recovery occurred in all but the MPD-30 group by start of testing. MPD-treated rats showed no changes in anxiety (elevated zero maze), swimming ability (straight channel swimming), or spatial learning/reference memory (Morris water maze). MPD does not appear to pose a risk to these CNS functions after exposure during a stage of rat development analogous to third trimester human brain development.


Subject(s)
Central Nervous System Stimulants/pharmacology , Learning Disabilities/chemically induced , Maze Learning/physiology , Memory Disorders/chemically induced , Methylphenidate/pharmacology , Space Perception/drug effects , Aging/drug effects , Aging/physiology , Animals , Animals, Newborn , Disease Models, Animal , Female , Learning Disabilities/physiopathology , Male , Maze Learning/drug effects , Memory Disorders/physiopathology , Pregnancy , Rats , Rats, Sprague-Dawley , Space Perception/physiology
SELECTION OF CITATIONS
SEARCH DETAIL