Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
J Neurosci Res ; 98(8): 1532-1548, 2020 08.
Article in English | MEDLINE | ID: mdl-32424971

ABSTRACT

Restless legs syndrome (RLS) is characterized by an irresistible need to move the legs while sitting or lying at night with insomnia as a frequent consequence. Human RLS has been associated with abnormalities in the endogenous opioid system, the dopaminergic system, the iron regulatory system, anemia, and inflammatory and auto-immune disorders. Our previous work indicates that mice lacking all three subtypes of opioid receptors have a phenotype similar to that of human RLS. To study the roles of each opioid receptor subtype in RLS, we first used mu opioid receptor knockout (MOR KO) mice based on our earlier studies using postmortem brain and cell culture. The KO mice showed decreased hemoglobin, hematocrit, and red blood cells (RBCs), with an appearance of microcytic RBCs indicating anemia. Together with decreased serum iron and transferrin, but increased ferritin levels, the anemia is similar to that seen with chronic inflammation in humans. A decreased serum iron level was also observed in the wildtype mice treated with an MOR antagonist. Iron was increased in the liver and spleen of the KO mice. Normal circadian variations in the dopaminergic and serotoninergic systems were absent in the KO mice. The KO mice showed hyperactivity and increased thermal sensitivity in wakefulness primarily during what would normally be the sleep phase similar to that seen in human RLS. Deficits in endogenous opioid system transmission could predispose to anemia of inflammation and loss of circadian variations in dopaminergic or serotonergic systems, thereby contributing to an RLS-like phenotype.


Subject(s)
Receptors, Opioid, mu/deficiency , Restless Legs Syndrome/blood , Restless Legs Syndrome/genetics , Anemia , Animals , Biogenic Monoamines/blood , Circadian Rhythm , Corpus Striatum , Dopamine/metabolism , Erythrocytes , Iron/blood , Mice , Mice, Knockout , Motor Activity , Pain , Psychomotor Agitation
2.
Sleep Med ; 71: 141-148, 2020 07.
Article in English | MEDLINE | ID: mdl-32094092

ABSTRACT

BACKGROUND: Brain iron deficiency (BID), especially for the substantia nigra (SN), without peripheral iron deficiency (ID) has been well documented as a ubiquitous finding for restless legs syndrome (RLS) patients. This close association suggests the biology of RLS BID can produce RLS symptoms. Association, however, cannot establish such a direct relationship. Instead, the BID of RLS could be experimentally produced to determine if it then produces significant RLS-like biological or behavioral features. Forward genetics approach led to identification from the BXD strains the BXD40 females (BXD40f) as a putative animal model for the RLS BID. The BXD40f on an iron-sufficient diet have a lower iron in the VMB (containing the SN) during the active but not inactive period. This was not found for the other BXD strains evaluated. The BXD40f on an ID diet uniquely have even greater reduced VMB but not peripheral iron, matching the RLS BID pathophysiology. A prior report found that the BXD40f on an iron-sufficient diet had an RLS-like behavior of increased activity occurring only in the last part of the active period that was not present in the other strains without the low VMB iron. This increased activity matches the circadian pattern of symptoms in RLS patients with increased urge or drive to move in the last part of the day. This study asks first: if you decrease the VMB iron by an iron deficient diet do the RLS-like behaviors worsen; and second will the dopaminergic treatments effective for RLS also reduce the worsened RLSlike behaviors. METHODS: In sum, 13 BXD40f mice post weaning were randomly assigned for 100 days to either a iron-sufficient diet (n = 6) or an ID diet (N = 7). They were then evaluated for 24-h activity in their home cage using implanted G2 EMitter telemetry device. At 3 h before the end of the active period IP doses were given every other day of either: saline (vehicle only), 12.5 mg levodopa, 25 mg levodopa, 0.5 mg quinpirole, or 1 0.0 mg quinpirole. RESULTS: The ID compared to irons-sufficient diet produced earlier onset of the RLS-like behavior matching the earlier onset of symptoms with increasing severity of RLS. The dopaminergic treatments significantly reduced the RLS-like behavior. Added analyses of the RLS-like behaviors as decreased resting times showed similar results to activity increases. CONCLUSIONS: These data demonstrate both that The BXD40f provide a useful animal model of RLS and also strongly support the hypothesis that the biology of RLS BID can produce RLS symptoms.


Subject(s)
Anemia, Iron-Deficiency , Restless Legs Syndrome , Anemia, Iron-Deficiency/drug therapy , Animals , Brain , Disease Models, Animal , Female , Humans , Iron , Mice , Restless Legs Syndrome/drug therapy
3.
Sleep Med ; 71: 135-140, 2020 07.
Article in English | MEDLINE | ID: mdl-32044226

ABSTRACT

BACKGROUND: The primary symptoms of Restless Legs Syndrome (RLS) are circadian-dependent, leading to increased activity or decreased rest, especially at night. The primary pathology in RLS is brain iron insufficiency despite normal systemic iron stores. Natural variances in brain and peripheral iron concentrations across recombinant inbred (RI) murine strains provide a biological model of RLS. The question is whether these RI mice strains show a behavioral analog to circadian-dependent clinical phenotype of RLS. METHODS: The home cage activity of eight female RI strains was measured over a 72-h period. The ratio of the average activity in the last 2 h of the active period relative to that in the total 12-h active period (late active period activity ratio, LAPAR) was the primary outcome variable. The relation of average LAPAR scores to measures of ventral midbrain (VMB) iron was evaluated across strains in this study. RESULTS: RI strain 40 (LAPAR = 1.28) and RI strain 21 (LAPAR = 1.02) were the only strains to show an increased activity in the last part of the active period. ANOVA showed the increased activity was significantly greater during the last 2 h compared to the preceding 10 h of the active phase only for the RI strain 40. Average LAPAR across the eight strains did not significantly correlate with the VMB iron content (r = -0.27, p < 0.10) but did correlate with changes in VMB iron with iron deficiency (r = 0.71, p < 0.05) and diurnal change in VMB iron (r = 0.65, p < 0.05). CONCLUSION: The female RI strain 40 mice exhibited a distinct end-of-active-period behavior analogous to circadian-dependent clinical phenotype of RLS.


Subject(s)
Restless Legs Syndrome , Animals , Brain , Female , Iron , Mesencephalon , Mice
4.
Physiol Behav ; 167: 282-288, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27640134

ABSTRACT

Iron deficiency (ID) is the most prevalent single-nutrient deficiency worldwide. There is evidence that ID early in development (preweaning in rat) causes irreversible neurologic, behavioral, and motor development deficits. Many of these effects have been attributed to damage to dopamine systems, including ID-induced changes in transporter and receptor numbers in the striatum and nucleus accumbens. These mesolimbic dopaminergic neurons are, in part, responsible for mediating reward and thus play a key role in addiction. However, there has been relatively little investigation into the behavioral effects of ID on drug addiction. In 2002, we found that rats made ID from weaning (postnatal day 21) and throughout the experiment acquired cocaine self-administration significantly more slowly than controls and failed to increase responding when the dose of the drug was decreased. In the present study, we assessed addiction for self-administered cocaine in rats with a history of preweaning ID only during postnatal days 4 through 21, and iron replete thereafter. The results showed that while ID did not affect the number of cocaine infusions or the overall addiction-like behavior score, ID rats scored higher on a measure of continued responding for drug than did iron replete controls. This increase in responding, however, was less goal-directed as ID rats also responded more quickly to the non-rewarded manipulandum than did control rats. Thus, while ID early in infancy did not significantly increase addiction-like behaviors for cocaine in this small study, the pattern of data suggests a possible underlying learning or performance impairment. Future studies will be needed to elucidate the exact neuro-behavioral deficits that lead to the increase in indiscriminate responding for drug in rats with a history of perinatal ID.


Subject(s)
Behavior, Addictive/physiopathology , Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Iron Deficiencies , Prenatal Exposure Delayed Effects/physiopathology , Analysis of Variance , Animals , Behavior, Addictive/chemically induced , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Dose-Response Relationship, Drug , Female , Hemoglobins/metabolism , Male , Pregnancy , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Reaction Time/physiology , Reinforcement Schedule , Self Administration
5.
J Am Assoc Lab Anim Sci ; 55(2): 204-12, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27025813

ABSTRACT

Blood collection is commonplace in biomedical research. Obtaining sufficient sample while minimizing animal stress requires significant skill and practice. Repeated needle punctures can cause discomfort and lead to variable release of stress hormones, potentially confounding analysis. We designed a handheld device to reduce the force necessary for needle insertion by using low-frequency, axial (forward and backward) micromotions (that is, vibration) delivered to the needle during venipuncture. Tests with cadaver rat-tail segments (n = 18) confirmed that peak insertion forces were reduced by 73% on average with needle vibration. A serial blood-sampling study was then conducted by using Sprague-Dawley rats divided into 2 groups based on needle condition used to cause bleeds: vibration on (n = 10) and vibration off (n = 9). On 3 days (1 wk apart), 3 tail-vein blood collections were performed in each subject at 1-h intervals. To evaluate associated stress levels, plasma corticosterone concentration was quantified by radioimmunoassay and behavior (that is, movement and vocalization) was scored by blinded review of blood-sampling videos. After the initial trial, average corticosterone was lower (46% difference), the mean intrasubject variance trended lower (72%), and behavioral indications of stress were rated lower for the vibration-on group compared with the vibration-off group. Adding controlled vibrations to needles during insertion may decrease the stress associated with blood sampling from rats--an important methodologic advance for investigators studying and assessing stress processes and a refinement over current blood sampling techniques.


Subject(s)
Blood Specimen Collection/veterinary , Corticosterone/blood , Rats, Sprague-Dawley , Animals , Blood Specimen Collection/methods , Female , Male , Needles , Phlebotomy/methods , Radioimmunoassay , Rats , Stress, Physiological , Vibration
6.
Exp Neurol ; 261: 462-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24999026

ABSTRACT

Iron deficiency (ID) in rodents leads to decreased ventral midbrain (VMB) iron concentrations and to changes in the dopamine (DA) system that mimic many of the dopaminergic changes seen in RLS patient where low substantia nigra iron is a known pathology of the disease. The ID-rodent model, therefore, has been used to explore the effects that low VMB iron can have on striatal DA dynamics with the hopes of better understanding the nature of iron-dopamine interaction in Restless Legs Syndrome (RLS). Using a post-weaning, diet-induced, ID condition in rats, the No-Net-Flux microdialysis technique was used to examine the effect of ID on striatal DA dynamics and it reversibility with acute infusion of physiological concentrations of iron into the VMB. This study replicated prior findings by showing that the ID condition is associated with increased extracellular striatal DA, reduced striatal DA uptake, and blunted DA-2-receptor-agonist feedback enhancement of striatal DA uptake. Despite the increase in extracellular striatal DA, intracellular striatal DA, as determined in tissue homogenates, was decrease in the ID rat. The study's key finding was that an infusion of physiological concentrations of iron into the VMB reversed the ID-induced increase in extracellular striatal DA and the ID-induced decrease in intracellular striatal DA but had no effect on the ID-induced changes in DA uptake or on the blunted DA-uptake response to quinpirole. In summary, the ID-rodent model provides highly reproducible changes in striatal DA dynamics that remarkably parallel dopaminergic changes seen in RLS patients. Some but not all of these ID-induced changes in striatal DA dynamics were reversible with physiological increases in VMB iron. The small changes in VMB iron induced by iron infusion likely represent biologically relevant changes in the non-transferrin-bound labile iron pool and may mimic circadian-dependent changes that have been found in VBM extracellular iron.


Subject(s)
Brain/metabolism , Corpus Striatum/metabolism , Dopamine/metabolism , Hydrodynamics , Iron Deficiencies , Iron Metabolism Disorders/pathology , Animals , Animals, Newborn , Brain/drug effects , Chromatography, High Pressure Liquid , Diet/adverse effects , Disease Models, Animal , Ferrous Compounds/administration & dosage , Iron Metabolism Disorders/drug therapy , Lipid Peroxidation , Male , Microdialysis , Rats , Rats, Sprague-Dawley , Thiobarbituric Acid Reactive Substances/metabolism
8.
Neurotoxicology ; 37: 26-34, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23558233

ABSTRACT

We analyzed genetic variation in severity of neuronal damage using the known dopaminergic neurotoxicant, MPTP, as a prototypical chemical denervation agent. Male mice from ten members of the BXD family of recombinant inbred strains received 12.5 mg/kg MPTP s.c. (vs. saline) and 48 h later brains were taken for multiple related biochemical analyses. Striatal dopamine (DA) and its metabolites, DOPAC and HVA, and serotonin and its metabolite, 5-HIAAA, were analyzed by HPLC. DA turnover was assessed using DOPAC/DA and HVA/DA ratios. Striatal tyrosine hydroxylase (TH), glial fibrilary acidic protein (GFAP), and iron content in ventral midbrain were quantified. All dopamine measures, as well as TH and GFAP, demonstrated wide, genotype-dependent differences in response to MPTP. Serotonin was largely unaffected. Principal components analysis (PC) on difference values, saline minus MPTP, for DA, DOPAC, HVA, and TH, yielded a dominant principal component. The PC trait residuals for each genotype were compared against complementary expression data for striatum of the same strains. Three transcripts representing Mtap2, Lancl 1, and Kansl1l were highly correlated with the PC, as was the difference score, MPTP minus saline for GFAP. This systems approach to the study of environmental neurotoxicants holds promise to define individual genetic differences that contribute to variability in susceptibility to risk factors for diseases such as Parkinson's disease.


Subject(s)
Brain/drug effects , Genetic Variation , MPTP Poisoning/genetics , Systems Biology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Biomarkers/metabolism , Brain/metabolism , Disease Models, Animal , Dopamine/metabolism , Gene Expression Regulation , Genetic Predisposition to Disease , Genomics , Glial Fibrillary Acidic Protein/metabolism , Homovanillic Acid/metabolism , Hydroxyindoleacetic Acid/metabolism , Iron/metabolism , MPTP Poisoning/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Multivariate Analysis , Phenotype , Principal Component Analysis , Risk Factors , Serotonin/metabolism , Species Specificity , Tyrosine 3-Monooxygenase/metabolism
9.
Biochim Biophys Acta ; 1832(6): 729-41, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23429074

ABSTRACT

Because of the increasing evidence that H63D HFE polymorphism appears in higher frequency in neurodegenerative diseases, we evaluated the neurological consequences of H63D HFE in vivo using mice that carry H67D HFE (homologous to human H63D). Although total brain iron concentration did not change significantly in the H67D mice, brain iron management proteins expressions were altered significantly. The 6-month-old H67D mice had increased HFE and H-ferritin expression. At 12 months, H67D mice had increased H- and L-ferritin but decreased transferrin expression suggesting increased iron storage and decreased iron mobilization. Increased L-ferritin positive microglia in H67D mice suggests that microglia increase iron storage to maintain brain iron homeostasis. The 6-month-old H67D mice had increased levels of GFAP, increased oxidatively modified protein levels, and increased cystine/glutamate antiporter (xCT) and hemeoxygenase-1 (HO-1) expression indicating increased metabolic and oxidative stress. By 12 months, there was no longer increased astrogliosis or oxidative stress. The decrease in oxidative stress at 12 months could be related to an adaptive response by nuclear factor E2-related factor 2 (Nrf2) that regulates antioxidant enzymes expression and is increased in the H67D mice. These findings demonstrate that the H63D HFE impacts brain iron homeostasis, and promotes an environment of oxidative stress and induction of adaptive mechanisms. These data, along with literature reports on humans with HFE mutations provide the evidence to overturn the traditional paradigm that the brain is protected from HFE mutations. The H67D knock-in mouse can be used as a model to evaluate how the H63D HFE mutation contributes to neurodegenerative diseases.


Subject(s)
Brain/metabolism , Heredodegenerative Disorders, Nervous System/metabolism , Histocompatibility Antigens Class I/metabolism , Iron/metabolism , Membrane Proteins/metabolism , Mutation, Missense , Nerve Tissue Proteins/metabolism , Oxidative Stress , Amino Acid Substitution , Amino Acid Transport System y+/biosynthesis , Amino Acid Transport System y+/genetics , Animals , Apoferritins/genetics , Apoferritins/metabolism , Brain/pathology , Glial Fibrillary Acidic Protein , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/genetics , Hemochromatosis Protein , Heredodegenerative Disorders, Nervous System/genetics , Histocompatibility Antigens Class I/genetics , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , NF-E2-Related Factor 2 , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics
10.
Pediatr Res ; 73(1): 31-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23095980

ABSTRACT

BACKGROUND: Long-term prefrontal cortex (PFC)- and hippocampus-based cognitive deficits are the sequelae of perinatal iron deficiency, despite iron supplementation starting in the newborn period. Whether high-dose iron supplementation prevents these deficits is yet to be determined. METHODS: Perinatal iron deficiency was induced in rat pups using a low-iron (3 mg/kg diet) diet during gestation until postnatal day (P)8. Iron was supplemented using a standard (40 mg/kg diet) or a 10-fold higher (400 mg/kg diet) iron-containing diet until P21. PFC and hippocampal neurochemistry was determined using in vivo (1)H nuclear magnetic resonance (NMR) spectroscopy at 9.4 Tesla on P90. RESULTS: Both standard and 10-fold higher iron supplementation doses corrected anemia and brain iron deficiency by P21. The neurochemical profile of the PFC in both supplementation groups was comparable with the control group. In the hippocampus, standard-dose iron supplementation resulted in lower concentrations of N-acetylaspartate (NAA) and phosphoethanolamine (PE) and higher concentrations of N-acetylaspartylglutamate (NAAG) and glycerophosphocholine + phosphocholine (GPC + PC). High-dose iron supplementation resulted in lower PE and higher GPC + PC concentrations. CONCLUSION: The iron supplementation dose for perinatal iron deficiency differentially alters the neurochemical profile of the PFC and hippocampus in adults. The neurochemical changes suggest altered glutamatergic neurotransmission, hypomyelination, and abnormal phospholipid metabolism in the formerly iron-deficient (FID) hippocampus.


Subject(s)
Anemia, Iron-Deficiency/complications , Cognition Disorders/prevention & control , Frontal Lobe/chemistry , Hippocampus/chemistry , Iron/pharmacology , Animals , Aspartic Acid/analogs & derivatives , Aspartic Acid/analysis , Cognition Disorders/etiology , Dietary Supplements , Dipeptides/analysis , Ethanolamines/analysis , Iron/metabolism , Magnetic Resonance Spectroscopy , Phosphorylcholine/analysis , Rats
11.
J Nutr ; 142(11): 2040-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22990465

ABSTRACT

Developmental iron deficiency anemia (IDA) causes brain and behavioral deficits in rodent models, which cannot be reversed when treated at periods equivalent to later infancy in humans. This study sought to determine whether earlier iron treatment can normalize deficits of IDA in rats and what iron dose is optimal. The offspring of dams with IDA during gestation were cross-fostered at postnatal d (P) 8 to dams receiving diets with 1 of 3 iron concentrations until weaning (P21): 0.003-0.01 g/kg [totally iron deficient (TID)]; 0.04 g/kg [formerly iron deficient (FID-40)]; or 0.4 g/kg (FID-400). Always iron-sufficient control dams (CN-40) received a 0.04-g/kg iron diet. At P21, TID pups received a 0.01 g iron/kg diet; all others received a 0.04 g iron/kg diet. Hematocrit and brain iron and monoamine concentrations were assessed at P21 and P100. Pup growth, development, activity, object recognition, hesitancy, and watermaze performance were evaluated. Regional brain iron was restored by iron treatment. Regional monoamine and metabolite concentrations were elevated in FID-40 rats and reduced in FID-400 and TID rats compared with CN-40 rats. FID-40 offspring had motor delays similar to TID during lactation and FID-400 rats had elevated thigmotaxis similar to TID rats at P25 and P100 in the spatial watermaze. In conclusion, iron treatment at P8 in rats did not normalize all monoamine or behavioral measures after early IDA. Moderate iron treatment improved adult behavior, but higher iron treatment caused brain and behavioral patterns similar to TID in the short and long term.


Subject(s)
Anemia, Iron-Deficiency/drug therapy , Behavior, Animal/drug effects , Biogenic Monoamines/metabolism , Iron/pharmacology , Animals , Brain/drug effects , Dietary Supplements , Dose-Response Relationship, Drug , Female , Iron/analysis , Male , Maternal Nutritional Physiological Phenomena , Milk/chemistry , Pregnancy , Pregnancy Complications, Hematologic , Rats , Rats, Sprague-Dawley
12.
Hum Mol Genet ; 21(18): 3984-92, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22678064

ABSTRACT

Restless legs syndrome (RLS), also known as Willis-Ekbom disease, is a sensory-motor neurological disorder with a circadian component. RLS is characterized by uncomfortable sensations in the extremities, generally at night or during sleep, which often leads to an uncontrollable urge to move them for relief. Recently, genomic studies identified single-nucleotide polymorphisms in BTBD9, along with three other genes, as being associated with a higher risk of RLS. Little is known about the function of BTBD9 or its potential role in the pathophysiology of RLS. We therefore examined a line of Btbd9 mutant mice we recently generated for phenotypes similar to symptoms found in RLS patients. We observed that the Btbd9 mutant mice had motor restlessness, sensory alterations likely limited to the rest phase, and decreased sleep and increased wake times during the rest phase. Additionally, the Btbd9 mutant mice had altered serum iron levels and monoamine neurotransmitter systems. Furthermore, the sensory alterations in the Btbd9 mutant mice were relieved using ropinirole, a dopaminergic agonist widely used for RLS treatment. These results, taken together, suggest that the Btbd9 mutant mice model several characteristics similar to RLS and would therefore be the first genotypic mouse model of RLS. Furthermore, our data provide further evidence that BTBD9 is involved in RLS, and future studies of the Btbd9 mutant mice will help shine light on its role in the pathophysiology of RLS. Finally, our data argue for the utility of Btbd9 mutant mice to discover and screen novel therapeutics for RLS.


Subject(s)
Carrier Proteins/genetics , Iron/blood , Nerve Tissue Proteins/genetics , Restless Legs Syndrome/genetics , Sleep Wake Disorders/genetics , Somatosensory Disorders/genetics , Animals , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/metabolism , Genetic Association Studies , Homozygote , Male , Mice , Mice, Transgenic , Motor Activity/genetics , Mutation , Photoperiod , Polysomnography , Restless Legs Syndrome/blood , Serotonergic Neurons/metabolism , Serotonin/metabolism , Sleep Wake Disorders/blood , Somatosensory Disorders/blood
13.
Am J Physiol Regul Integr Comp Physiol ; 302(11): R1282-96, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22461179

ABSTRACT

The aim of this study was to identify genes that influence iron regulation under varying dietary iron availability. Male and female mice from 20+ BXD recombinant inbred strains were fed iron-poor or iron-adequate diets from weaning until 4 mo of age. At death, the spleen, liver, and blood were harvested for the measurement of hemoglobin, hematocrit, total iron binding capacity, transferrin saturation, and liver, spleen and plasma iron concentration. For each measure and diet, we found large, strain-related variability. A principal-components analysis (PCA) was performed on the strain means for the seven parameters under each dietary condition for each sex, followed by quantitative trait loci (QTL) analysis on the factors. Compared with the iron-adequate diet, iron deficiency altered the factor structure of the principal components. QTL analysis, combined with PosMed (a candidate gene searching system) published gene expression data and literature citations, identified seven candidate genes, Ptprd, Mdm1, Picalm, lip1, Tcerg1, Skp2, and Frzb based on PCA factor, diet, and sex. Expression of each of these is cis-regulated, significantly correlated with the corresponding PCA factor, and previously reported to regulate iron, directly or indirectly. We propose that polymorphisms in multiple genes underlie individual differences in iron regulation, especially in response to dietary iron challenge. This research shows that iron management is a highly complex trait, influenced by multiple genes. Systems genetics analysis of iron homeostasis holds promise for developing new methods for prevention and treatment of iron deficiency anemia and related diseases.


Subject(s)
Anemia, Iron-Deficiency/genetics , Iron Deficiencies , Analysis of Variance , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/metabolism , Animals , Body Weight , Female , Gene Expression , Hemoglobins/analysis , Homeostasis/genetics , Iron/metabolism , Male , Mice , Mice, Inbred Strains , Multivariate Analysis , Polymorphism, Genetic , Quantitative Trait Loci
14.
Neurogenetics ; 13(2): 147-57, 2012 May.
Article in English | MEDLINE | ID: mdl-22457016

ABSTRACT

Iron regulation in the brain is both necessary and highly complex. Too little or too much iron can compromise neurological function, yet we still do not know all of the regulatory processes. In our research, we seek to identify genes and gene networks underlying individual differences in brain iron regulation. To this end, we fed mice from 20+ inbred strains a diet low in iron from weaning to 4 months of age. At sacrifice, we measured iron content in the ventral midbrain (VMB). The VMB contains the substantia nigra, a region particularly vulnerable to iron imbalance. The results showed high, inter-strain variability in dietary iron reduction, from almost no loss to more than 40 % vs. control. When we performed quantitative trait loci (QTL) analysis, we observed a significant area on chromosome 2. Within this QTL, we selected glial high-affinity glutamate transporter 1 (Glt1) as the leading candidate. Expression of this gene is both correlated with VMB iron and is also cis-modulated by local sequence variants that segregate in the BXD family. VMB expression differences of Glt1 in six strains covary with differential susceptibility to VMB iron loss.


Subject(s)
Brain/metabolism , Iron Deficiencies , Quantitative Trait Loci , Animals , Chromosome Mapping , Excitatory Amino Acid Transporter 2/metabolism , Female , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Phenotype
15.
J Neurochem ; 120(1): 125-34, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22035068

ABSTRACT

Ceruloplasmin (Cp) is a ferroxidase involved in iron metabolism by converting Fe(2+) to Fe(3+), and by regulating cellular iron efflux. In the ceruloplasmin knockout (CpKO) mouse, the deregulation of iron metabolism results in moderate liver and spleen hemosiderosis, but the impact of Cp deficiency on brain neurochemistry and behavior in this animal model is unknown. We found that in contrast to peripheral tissues, iron levels in the hippocampus are significantly reduced in CpKO mice. Although it does not cause any discernable deficits in motor function or learning and memory, Cp deficiency results in heightened anxiety-like behavior in the open field and elevated plus maze tests. This anxiety phenotype is associated with elevated levels of plasma corticosterone. Previous studies provided evidence that anxiety disorders and long-standing stress are associated with reductions in levels of serotonin (5HT) and brain-derived neurotrophic factor (BDNF) in the hippocampus. We found that levels of 5HT and norepinephrine (NE), and the expression of BDNF and its receptor trkB, are significantly reduced in the hippocampus of CpKO mice. Thus, Cp deficiency causes an anxiety phenotype by a mechanism that involves decreased levels of iron, 5HT, NE, and BDNF in the hippocampus.


Subject(s)
Anxiety/metabolism , Anxiety/psychology , Brain-Derived Neurotrophic Factor/deficiency , Ceruloplasmin/deficiency , Hippocampus/metabolism , Iron Deficiencies , Serotonin/deficiency , Animals , Brain Chemistry/genetics , Ceruloplasmin/genetics , Corticosterone/blood , Fear/physiology , Hindlimb Suspension , Learning/physiology , Male , Maze Learning/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Postural Balance/physiology , Psychomotor Performance/physiology , Real-Time Polymerase Chain Reaction , Recognition, Psychology/physiology , Transcription, Genetic
16.
PLoS One ; 6(9): e25077, 2011.
Article in English | MEDLINE | ID: mdl-21949858

ABSTRACT

Ceruloplasmin (Cp) is an essential ferroxidase that plays important roles in cellular iron trafficking. Previous findings suggest that the proper regulation and subcellular localization of iron are very important in brain cell function and viability. Brain iron dyshomeostasis is observed during normal aging, as well as in several neurodegenerative disorders such as Alzheimer's, Parkinson's and Huntington's diseases, coincident with areas more susceptible to insults. Because of their high metabolic demand and electrical excitability, neurons are particularly vulnerable to ischemic injury and death. We therefore set out to look for abnormalities in the brain of young adult mice that lack Cp. We found that iron levels in the striatum and cerebral cortex of these young animals are significantly lower than wild-type (WT) controls. Also mRNA levels of the neurotrophin brain derived neurotrophic factor (BDNF), known for its role in maintenance of cell viability, were decreased in these brain areas. Chelator-mediated depletion of iron in cultured neural cells resulted in reduced BDNF expression by a posttranscriptional mechanism, suggesting a causal link between low brain iron levels and reduced BDNF expression. When the mice were subjected to middle cerebral artery occlusion, a model of focal ischemic stroke, we found increased brain damage in Cp-deficient mice compared to WT controls. Our data indicate that lack of Cp increases neuronal susceptibility to ischemic injury by a mechanism that may involve reduced levels of iron and BDNF.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Ceruloplasmin/physiology , Corpus Striatum/metabolism , Iron/metabolism , Neostriatum/metabolism , Stroke/etiology , Animals , Blotting, Western , Brain/cytology , Brain/metabolism , Brain-Derived Neurotrophic Factor/genetics , Disease Models, Animal , Humans , Immunoenzyme Techniques , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Male , Mice , Mice, Knockout , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurons/cytology , Neurons/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Stroke/metabolism , Stroke/pathology , Tumor Cells, Cultured
17.
Mamm Genome ; 22(9-10): 556-62, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21732193

ABSTRACT

Iron homeostasis is crucial to many biological functions in nearly all organisms, with roles ranging from oxygen transport to immune function. Disruption of iron homeostasis may result in iron overload or iron deficiency. Iron deficiency may have severe consequences, including anemia or changes in immune or neurotransmitter systems. Here we report on the variability of phenotypic iron tissue loss and splenomegaly and the associated quantitative trait loci (QTLs), polymorphic areas in the mouse genome that may contain one or more genes that play a role in spleen iron concentration or spleen weight under each dietary treatment. Mice from 26 BXD/Ty recombinant inbred strains, including the parent C57BL/6 and DBA/2 strains, were randomly assigned to adequate iron or iron-deficient diets at weaning. After 120 days, splenomegaly was measured by spleen weight, and spleen iron was assessed using a modified spectrophotometry technique. QTL analyses and gene expression comparisons were then conducted using the WebQTL GeneNetwork. We observed wide, genetic-based variability in splenomegaly and spleen iron loss in BXD/Ty recombinant inbred strains fed an iron-deficient diet. Moreover, we identified several suggestive QTLs. Matching our QTLs with gene expression data from the spleen revealed candidate genes. Our work shows that individual differences in splenomegaly response to iron deficiency are influenced at least partly by genetic constitution. We propose mechanistic hypotheses by which splenomegaly may result from iron deficiency.


Subject(s)
Iron Deficiencies , Iron Metabolism Disorders/genetics , Quantitative Trait Loci , Spleen/metabolism , Animals , Female , Gene Expression Profiling , Iron/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Organ Size/genetics , Phenotype , Sex Factors
18.
Neurotoxicol Teratol ; 33(3): 415-21, 2011.
Article in English | MEDLINE | ID: mdl-21371552

ABSTRACT

Paraquat (PQ) is an herbicide used extensively in agriculture. This agent is also suspected to be a risk factor for Parkinson's disease (PD) by harming nigro-striatal dopamine neurons. There is likely, genetic-based, individual variability in susceptibility to PQ neurotoxicity related PD. In this study, we measured the delivery of PQ to the brain after three weekly injections of PQ at 5 mg kg(-1), PQ-related neural toxicity after three weekly injections of PQ at 1 mg kg(-1)or 5 mg kg(-1), PQ-related iron accumulation and PQ-related gene expression in midbrain of DBA/2J (D2) and C57BL/6J (B6) inbred mouse strains after a single injection of PQ at 15 mg kg(-1) and 10 mg kg(-1), respectively. Results showed that compared to controls, PQ-treated B6 mice lost greater numbers of dopaminergic neurons in the substantia nigra pars compacta than D2 mice; however, distribution of PQ to the midbrain was equal between the strains. PQ also significantly increased iron concentration in the midbrain of B6 but not D2 mice. Microarray analysis of the ventral midbrain showed greater PQ-induced changes in gene expression in B6 compared to D2 mice. This is the first study to report genetically-based differences in susceptibility to PQ neurotoxicity and to understanding individual differences in vulnerability to PQ neurotoxicity and its relation to PD in humans.


Subject(s)
Environmental Pollutants/toxicity , Gene Expression/drug effects , Genetic Predisposition to Disease , Neurotoxicity Syndromes/genetics , Paraquat/toxicity , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Count , Dopamine/metabolism , Dose-Response Relationship, Drug , Environmental Pollutants/pharmacokinetics , Female , Gene Expression Profiling , Immunohistochemistry , Iron/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Oligonucleotide Array Sequence Analysis , Paraquat/pharmacokinetics , Species Specificity , Tissue Distribution
19.
J Nutr ; 141(3): 386-90, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21209256

ABSTRACT

Iron deficiency is among the most prevalent of nutrient-related diseases worldwide, but the long-term consequences of maternal and neonatal iron deficiency on offspring are not well characterized. We investigated the effects of a postweaning iron-adequate diet following neonatal iron deficiency on the expression of genes involved in iron acquisition and homeostasis. Pregnant rats were fed an iron-adequate diet (0.08 g iron/kg diet) until gestational d 15, at which time they were divided into 2 groups: 1) a control group fed an iron-adequate diet, and 2) an iron-deficient group fed an iron-deficient diet (0.005 g iron/kg diet) through postnatal d (P) 23 (weaning). After weaning, pups from both dietary treatment groups were fed an iron-adequate diet until adulthood (P75). Rat pups that were iron deficient during the neonatal period (IDIA) had reduced weight gain and hemoglobin concentrations and decreased levels of serum, liver, and spleen iron on P75 compared with rats that were iron sufficient throughout early life (IA). IDIA rats developed erythrocytosis during postweaning development. Further, hepatic expression of hepcidin in IDIA rats was 1.4-fold greater than in IA rats, which paralleled an upregulation of IL-1 expression in the serum. Our data suggest that an iron-adequate diet following neonatal iron deficiency induced an inflammatory milieu that affected iron homeostasis and early growth and development.


Subject(s)
Anemia, Iron-Deficiency/physiopathology , Growth Disorders/etiology , Iron, Dietary/therapeutic use , Iron/metabolism , Maternal Nutritional Physiological Phenomena , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/diet therapy , Animals , Animals, Newborn , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Female , Gene Expression Regulation, Developmental , Hemoglobins/analysis , Hepcidins , Homeostasis , Interleukin-1/blood , Iron/blood , Liver/metabolism , Male , Polycythemia/etiology , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Spleen/metabolism , Weaning
20.
Am J Physiol Gastrointest Liver Physiol ; 300(3): G470-6, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21193529

ABSTRACT

Dietary iron is particularly critical during periods of rapid growth such as in neonatal development. Human and rodent studies have indicated that iron deficiency or excess during this critical stage of development can have significant long- and short-term consequences. Since the requirement for iron changes during development, the availability of adequate iron is critical for the differentiation and maturation of individual organs participating in iron homeostasis. We have examined in rats the effects of dietary iron supplement following neonatal iron deficiency on tissue iron status in relation to erythropoietic ability during 16 wk of postweaning development. This physiological model indicates that postweaning iron-adequate diet following neonatal iron deficiency adversely affects erythroid differentiation in the bone marrow and promotes splenic erythropoiesis leading to splenomegaly and erythrocytosis. This altered physiology of iron homeostasis during postweaning development is also reflected in the inability to maintain liver and spleen iron concentrations and the altered expression of iron regulatory proteins in the liver. These studies provide critical insights into the consequences of neonatal iron deficiency and the dietary iron-induced cellular signals affecting iron homeostasis during early development.


Subject(s)
Anemia, Iron-Deficiency/blood , Bone Marrow/metabolism , Erythroid Precursor Cells/metabolism , Erythropoiesis , Iron Deficiencies , Iron, Dietary/blood , Liver/metabolism , Spleen/metabolism , Age Factors , Anemia, Iron-Deficiency/diet therapy , Anemia, Iron-Deficiency/pathology , Animals , Animals, Newborn , Bone Marrow/pathology , Erythropoietin/blood , Female , Growth Differentiation Factor 15/blood , Hematocrit , Hemoglobins/metabolism , Homeostasis , Iron, Dietary/administration & dosage , Iron, Dietary/adverse effects , Iron-Regulatory Proteins/genetics , Iron-Regulatory Proteins/metabolism , Male , Maternal Nutritional Physiological Phenomena , Polycythemia/blood , Polycythemia/etiology , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Sprague-Dawley , Spleen/pathology , Splenomegaly , Transferrin/metabolism , Weaning
SELECTION OF CITATIONS
SEARCH DETAIL
...