Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Nutr ; 148(10): 1521-1528, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30169712

RESUMEN

Background: Iron deficiency (ID) compromises the developing nervous system, including the hippocampus, resulting in later-life deficits despite iron repletion. The iron-dependent molecular changes driving these lasting deficits, and the effect of early iron repletion, are incompletely understood. Previous studies have utilized dietary models of maternal-fetal ID anemia (IDA) to address these questions; however, concurrent anemia prevents delineation of the specific role of iron. Objective: The aim of the study was to isolate the effects of developmental ID on adult hippocampal gene expression and to determine if iron repletion reverses these effects in a mouse model of nonanemic hippocampal neuronal ID. Methods: Nonanemic, hippocampus-specific neuronal ID was generated by using a Tet-OFF dominant negative transferrin receptor (DN-TFR1) mouse model that impairs cellular iron uptake. Hippocampal ID was reversed with doxycycline at postnatal day 21 (P21) in a subset of mice to create 2 experimental groups, chronically iron-deficient and formerly iron-deficient mice, which were compared with their respective doxycycline-treated and untreated iron-sufficient controls. RNA from adult male hippocampi was sequenced. Paired-end reads were analyzed for differential expression. Differentially expressed genes were analyzed in Ingenuity Pathway Analysis. Results: A total of 346 genes were differentially expressed in adult, chronically iron-deficient hippocampi compared with controls. ID dysregulated genes in critical neurodevelopmental pathways, including axonal guidance, CDK5, Ephrin receptor, Rac, and Neurotrophin/Trk signaling. Iron repletion at P21 normalized adult hippocampal expression of 198 genes; however, genes involved in cAMP response element-binding protein (CREB) signaling, neurocognition, and neurologic disease remained dysregulated in adulthood. Conclusions: Chronic ID during development, independent of anemia, alters the adult mouse hippocampal transcriptome. Restoring iron status during a known critical period of hippocampal neurodevelopment incompletely normalized these changes, suggesting a need for additional studies to identify the most effective timeline for iron therapy, and adjunctive treatments that can fully restore ID-induced molecular changes, particularly in human populations in whom chronic ID is endemic.


Asunto(s)
Anemia Ferropénica/complicaciones , Animales Recién Nacidos/metabolismo , Hipocampo/metabolismo , Deficiencias de Hierro , Sistema Nervioso/crecimiento & desarrollo , Transcriptoma , Anemia Ferropénica/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Hipocampo/citología , Hierro/metabolismo , Hierro/uso terapéutico , Masculino , Ratones , Ratones Transgénicos , Sistema Nervioso/metabolismo , Neurogénesis , Neuronas/metabolismo , Estado Nutricional , ARN/análisis , Receptores de Transferrina/metabolismo
2.
Hippocampus ; 23(10): 952-62, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23733517

RESUMEN

Iron deficiency (ID) during early life causes long-lasting detrimental cognitive sequelae, many of which are linked to alterations in hippocampus function, dopamine synthesis, and the modulation of dopaminergic circuitry by the hippocampus. These same features have been implicated in the origins of schizophrenia, a neuropsychiatric disorder with significant cognitive impairments. Deficits in sensorimotor gating represent a reliable endophenotype of schizophrenia that can be measured by prepulse inhibition (PPI) of the acoustic startle reflex. Using two rodent model systems, we investigated the influence of early-life ID on PPI in adulthood. To isolate the role of hippocampal iron in PPI, our mouse model utilized a timed (embryonic day 18.5), hippocampus-specific knockout of Slc11a2, a gene coding an important regulator of cellular iron uptake, the divalent metal transport type 1 protein (DMT-1). Our second model used a classic rat dietary-based global ID during gestation, a condition that closely mimics human gestational ID anemia (IDA). Both models exhibited impaired PPI in adulthood. Furthermore, our DMT-1 knockout model displayed reduced long-term potentiation (LTP) and elevated paired-pulse facilitation (PPF), electrophysiological results consistent with previous findings in the IDA rat model. These results, in combination with previous findings demonstrating impaired hippocampus functioning and altered dopaminergic and glutamatergic neurotransmission, suggest that iron availability within the hippocampus is critical for the neurodevelopmental processes underlying sensorimotor gating. Ultimately, evidence of reduced PPI in both of our models may offer insights into the roles of fetal ID and the hippocampus in the pathophysiology of schizophrenia.


Asunto(s)
Conducta Animal/fisiología , Enfermedades Carenciales/complicaciones , Hipocampo/metabolismo , Deficiencias de Hierro , Trastornos del Metabolismo del Hierro/complicaciones , Inhibición Prepulso/fisiología , Reflejo de Sobresalto/fisiología , Anemia Ferropénica/sangre , Anemia Ferropénica/complicaciones , Anemia Ferropénica/fisiopatología , Animales , Proteínas de Transporte de Catión/genética , Enfermedades Carenciales/fisiopatología , Modelos Animales de Enfermedad , Femenino , Trastornos Nutricionales en el Feto/sangre , Trastornos Nutricionales en el Feto/fisiopatología , Hipocampo/patología , Hipocampo/fisiopatología , Hierro/metabolismo , Trastornos del Metabolismo del Hierro/sangre , Trastornos del Metabolismo del Hierro/fisiopatología , Potenciación a Largo Plazo/fisiología , Ratones , Ratones Noqueados , Embarazo , Complicaciones del Embarazo/sangre , Ratas , Ratas Sprague-Dawley , Potenciales Sinápticos/fisiología
3.
J Nutr ; 143(3): 260-6, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23303869

RESUMEN

Iron deficiency (ID) is the most common nutrient deficiency worldwide, disproportionally affecting infants, children, and women of childbearing age. Although ID commonly occurs with anemia (IDA), nonanemic ID is 3 times more common than IDA in toddlers and also occurs in infants following gestational complications. Both conditions negatively affect motor, socio-emotional, and cognitive behaviors, suggesting that iron, apart from anemia, has a critical role in neurodevelopment. Here, the specific role of iron in regulation of mammalian target of rapamycin (mTOR) signaling (a kinase pathway that integrates metabolic supply and demand to regulate cell growth and morphology) was examined using 2 hippocampal, pyramidal cell-specific, nonanemic, genetic mouse models of ID: a CAMKIIα cre-loxP permanent knockout of divalent metal transporter-1 (DMT-1 CKO) and a CAMKIIα-tTA-driven reversible, overexpression of nonfunctional, dominant negative transferrin receptor-1 (DN TfR-1). In both models, mTOR activity, assessed by phosphorylation levels of key proteins, was upregulated during development by ID [S6K(Thr389) phosphorylation increased 87 and 57% in the DMT-1 CKO and DN TfR-1 models, respectively; P < 0.05]. This effect was shown to be iron-dependent, because iron repletion at postnatal d 21 normalized mTOR activity in the reversible DN TfR-1 model (62% reduction compared with unrepleted mice; P < 0.05). In the permanent DMT-1 CKO model, suppression of ID-induced mTOR hyperactivity by rapamycin administered during the sensitive period for iron improved Morris water maze performance despite ongoing ID (DMT-1 wild-type and DMT-1 CKO mice reached criterion in 3 d compared with 4 d necessary for vehicle-treated DMT-1 CKO mice; P < 0.05). Together, these findings implicate mTOR dysregulation as a cellular mechanism underlying the acute and persistent neurodevelopmental deficits that accompany early-life ID.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Hipocampo/crecimiento & desarrollo , Deficiencias de Hierro , Trastornos del Metabolismo del Hierro/metabolismo , Células Piramidales/metabolismo , Receptores de Transferrina/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Animales Recién Nacidos , Proteínas de Transporte de Catión/genética , Modelos Animales de Enfermedad , Hipocampo/citología , Hipocampo/metabolismo , Hierro/metabolismo , Trastornos del Metabolismo del Hierro/genética , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Noqueados , Mutación , Fosforilación , Receptores de Transferrina/genética , Transducción de Señal , Sirolimus/farmacología , Regulación hacia Arriba
4.
Am J Physiol Endocrinol Metab ; 302(3): E316-24, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22068601

RESUMEN

Gestational-neonatal iron deficiency, a common micronutrient deficiency affecting the offspring of more than 30% of pregnancies worldwide, leads to long-term cognitive and behavioral abnormalities. Preclinical models of gestational-neonatal iron deficiency result in reduced energy metabolism and expression of genes critical for neuronal plasticity and cognitive function, which are associated with a smaller hippocampal volume and abnormal neuronal dendrite growth. Because insulin-like growth factor (IGF) modulates early postnatal cellular growth, differentiation, and survival, we used a dietary-induced rat model to assess the effects of gestational iron deficiency on activity of the IGF system. We hypothesized that gestational iron deficiency attenuates postnatal hippocampal IGF signaling and results in downstream effects that contribute to hippocampal anatomic and functional deficits. At postnatal day (P) 15 untreated gestational-neonatal iron deficiency markedly suppressed hippocampal IGF activation and protein kinase B signaling, and reduced neurogenesis, while elevating extracellular signal-regulated kinase 1/2 signaling and hypoxia-inducible factor-1α expression. Iron treatment beginning at P7 restored IGF signaling, increased neurogenesis, and normalized all parameters by the end of rapid hippocampal differentiation (P30). Expression of the neuron-specific synaptogenesis marker, disc-large homolog 4 (PSD95), increased more rapidly than the glia-specific myelination marker, myelin basic protein, following iron treatment, suggesting a more robust response to iron therapy in IGF-I-dependent neurons than IGF-II-dependent glia. Collectively, our findings suggest that IGF dysfunction is in part responsible for hippocampal abnormalities in untreated iron deficiency. Early postnatal iron treatment of gestational iron deficiency reactivates the IGF system and promotes neurogenesis and differentiation in the hippocampus during a critical developmental period.


Asunto(s)
Anemia Ferropénica/dietoterapia , Anemia Ferropénica/metabolismo , Hipocampo/metabolismo , Hierro de la Dieta/uso terapéutico , Fenómenos Fisiologicos Nutricionales Maternos , Transducción de Señal , Somatomedinas/metabolismo , Anemia Ferropénica/patología , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Hipocampo/patología , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Hierro de la Dieta/administración & dosificación , Lactancia , Masculino , Neurogénesis , Neuronas/metabolismo , Embarazo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Somatomedinas/genética
5.
Pediatr Res ; 65(5): 493-8, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19190544

RESUMEN

Fetal-neonatal iron deficiency acutely alters hippocampal biochemistry, neural morphology, and electrophysiology accompanied by a downregulation of brain-derived neurotrophic factor (BDNF). These changes provide a cellular and molecular basis for observed short-term learning and memory impairments. However, the etiology of residual, long-term hippocampal neurotransmission abnormalities and learning impairments after treatment remain unclear. Because BDNF modulates learning and memory, we assessed its expression in 65-d-old formerly iron deficient (FID) male rats that had been iron deficient during the fetal-neonatal period and treated with iron since postnatal day 7. BDNF-III and -IV mRNAs and BDNF protein expression remained down-regulated in FID rats when compared with the always iron-sufficient rats. Expressions of BDNF activity-dependent downstream targets (3-hydroxy-3-methylglutaryl CoA reductase and immediate early genes c-fos, early growth response gene 1 and 2) were reduced in FID rats. In turn, hippocampal expressions of direct targets of early-growth response genes, including hypoxia-inducible factor 1, dual-specificity phosphatase 4, IGF 2, and myelin basic protein were also diminished in FID rats. Collectively, fetal-neonatal iron deficiency lowers hippocampal BDNF expression and function beyond the period of iron deficiency. These findings may underlie the persistence of learning deficits seen after fetal-neonatal iron deficiency.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hipocampo/metabolismo , Deficiencias de Hierro , Efectos Tardíos de la Exposición Prenatal , Factores de Edad , Animales , Animales Recién Nacidos , Factor Neurotrófico Derivado del Encéfalo/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Fosfatasas de Especificidad Dual/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Femenino , Edad Gestacional , Hidroximetilglutaril-CoA Reductasas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factor II del Crecimiento Similar a la Insulina/metabolismo , Hierro de la Dieta/administración & dosificación , Masculino , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Proteína Básica de Mielina/metabolismo , Embarazo , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor trkB/metabolismo
6.
J Nutr ; 138(12): 2495-501, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19022978

RESUMEN

Fetal-neonatal iron deficiency alters hippocampal neuronal morphology, reduces its volume, and is associated with acute and long-term learning impairments. However, neither the effects of early-life iron deficiency anemia on growth, differentiation, and survival of hippocampal neurons nor regulation of the neurotrophic factors that mediate these processes has been investigated. We compared hippocampal expression of neurotrophic factors in male rats made iron deficient (ID) from gestational d 2 to postnatal d (P) 7 to iron-sufficient controls at P7, 15, and 30 with quantitative RT-PCR, Western analysis, and immunohistology. Iron deficiency downregulated brain-derived neurotrophic factor (BDNF) expression in the hippocampus without compensatory upregulation of its specific receptor, tyrosine-receptor kinase B. Consistent with low overall BDNF activity, we found lower expression of early-growth response gene-1 and -2, transcriptional targets of BDNF signaling. Doublecortin expression, a marker of differentiating neurons, was reduced during peak iron deficiency, suggesting impaired neuronal differentiation in the ID hippocampus. In contrast, iron deficiency upregulated hippocampal nerve growth factor, epidermal growth factor, and glial-derived neurotrophic factor accompanied by an increase in neurotrophic receptor p75 expression. Our findings suggest that fetal-neonatal iron deficiency lowers BDNF function and impairs neuronal differentiation in the hippocampus.


Asunto(s)
Anemia Ferropénica/metabolismo , Anemia Ferropénica/patología , Hipocampo/metabolismo , Hipocampo/patología , Factores de Crecimiento Nervioso/metabolismo , Anemia Ferropénica/genética , Animales , Animales Recién Nacidos , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Diferenciación Celular , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 2 de la Respuesta de Crecimiento Precoz/genética , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Expresión Génica , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Masculino , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Factores de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso , Neuronas/patología , Neuropéptidos/genética , Neuropéptidos/metabolismo , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo
7.
Curr Protoc Toxicol ; 64: 11.19.1-11, 2015 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-26344221

RESUMEN

In this protocol, we combine two-photon excitation fluorescence with nonlinear optical measurements to reconstruct the three-dimensional architecture of the pharyngeal region and the muscular system of the anterior and mid-body region of Caenorhabditis elegans (C. elegans). Femto-second laser pulses excite second-harmonic generation (SHG) and third-harmonic generation (THG) signals, which show detailed structural information regarding the organization of myofibrils that are arranged around the central pharynx region. The combination of two-photon excitation with SHG and THG imaging is a very powerful tool to study cell morphology, microarchitecture, and tissue arrangement in C. elegans.


Asunto(s)
Caenorhabditis elegans/anatomía & histología , Imagenología Tridimensional/métodos , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Músculos/anatomía & histología , Neuronas/ultraestructura , Faringe/anatomía & histología , Animales , Caenorhabditis elegans/ultraestructura , Dinámicas no Lineales
8.
Metallomics ; 4(7): 593-605, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22337135

RESUMEN

Metals can have a number of detrimental or beneficial effects in the cell, but first they must get in. Organisms have evolved transport mechanisms to get metals that are required, or essential into the cell. Nonessential metals often enter the cell through use of the machinery provided for essential metals. Much work has been done to advance our understanding of how these metals are transported across plasma and organelle membranes. This review provides an overview of essential and nonessential metal transport and homeostatic processes.


Asunto(s)
Células/metabolismo , Homeostasis , Metales/metabolismo , Animales , Transporte Biológico , Humanos , Metales/toxicidad
9.
Toxicol Res (Camb) ; 1(1): 32-38, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27795823

RESUMEN

The neurotoxic consequences of methylmercury (MeHg) exposure have long been known, however a complete understanding of the mechanisms underlying this toxicity is elusive. Recent epidemiological and experimental studies have provided many mechanistic insights, particularly into the contribution of genetic and environmental factors that interact with MeHg to modify toxicity. This review will outline cellular processes directly and indirectly affected by MeHg, including oxidative stress, cellular signaling and gene expression, and discuss genetic, environmental and nutritional factors capable of modifying MeHg toxicity.

10.
Endocrinology ; 153(11): 5668-80, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23054056

RESUMEN

Copper (Cu), iron (Fe), and thyroid hormone (TH) deficiencies produce similar defects in late brain development including hypomyelination of axons and impaired synapse formation and function, suggesting that these micronutrient deficiencies share a common mechanism contributing to these derangements. We previously demonstrated that fetal/neonatal Cu and Fe deficiencies lower circulating TH concentrations in neonatal rats. Fe deficiency also reduces whole-brain T(3) content, suggesting impaired TH action in the developing Fe-deficient brain. We hypothesized that fetal/neonatal Cu and Fe deficiencies will produce mild or moderate TH deficiencies and will impair TH-responsive gene expression in the neonatal cerebral cortex and hippocampus. To test this hypothesis, we rendered pregnant Sprague Dawley rats Cu-, Fe-, or TH-deficient from early gestation through postnatal d 10 (P10). Mild and moderate TH deficiencies were induced by 1 and 3 ppm propylthiouracil treatment, respectively. Cu deficiency did not significantly alter serum or tissue TH concentrations or TH-responsive brain mRNA expression. Fe deficiency significantly lowered P10 serum total T(3) (45%), serum total T(4) (52%), whole brain T(3) (14%), and hippocampal T(3) (18%) concentrations, producing a mild TH deficiency similar to 1 ppm propylthiouracil treatment. Fe deficiency lowered Pvalb, Enpp6, and Mbp mRNA levels in the P10 hippocampus. Fe deficiency also altered Hairless, Dbm, and Dio2 mRNA levels in the P10 cerebral cortex. These results suggest that some of the brain defects associated with Fe deficiency may be mediated through altered thyroidal status and the concomitant alterations in TH-responsive gene transcription.


Asunto(s)
Corteza Cerebral/metabolismo , Hipocampo/metabolismo , Deficiencias de Hierro , Glándula Tiroides/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Animales Recién Nacidos , Cobre/deficiencia , Femenino , Expresión Génica/genética , Masculino , Embarazo , Ratas , Ratas Sprague-Dawley
11.
Front Genet ; 3: 52, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22514555

RESUMEN

Metals are major contaminants that influence human health. Many metals have physiologic roles, but excessive levels can be harmful. Advances in technology have made toxicogenomic analyses possible to characterize the effects of metal exposure on the entire genome. Much of what is known about cellular responses to metals has come from mammalian systems; however the use of non-mammalian species is gaining wider attention. Caenorhabditis elegans is a small round worm whose genome has been fully sequenced and its development from egg to adult is well characterized. It is an attractive model for high throughput screens due to its short lifespan, ease of genetic mutability, low cost, and high homology with humans. Research performed in C. elegans has led to insights in apoptosis, gene expression, and neurodegeneration, all of which can be altered by metal exposure. Additionally, by using worms one can potentially study mechanisms that underline differential responses to metals in nematodes and humans, allowing for identification of novel pathways and therapeutic targets. In this review, toxicogenomic studies performed in C. elegans exposed to various metals will be discussed, highlighting how this non-mammalian system can be utilized to study cellular processes and pathways induced by metals. Recent work focusing on neurodegeneration in Parkinson's disease will be discussed as an example of the usefulness of genetic screens in C. elegans and the novel findings that can be produced.

12.
Adv Nutr ; 2(2): 112-21, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22332040

RESUMEN

Iron deficiency (ID) is the most common nutrient deficiency, affecting 2 billion people and 30% of pregnant women and their offspring. Early life ID affects at least 3 major neurobehavioral domains, including speed of processing, affect, and learning and memory, the latter being particularly prominent. The learning and memory deficits occur while the infants are iron deficient and persist despite iron repletion. The neural mechanisms underlying the short- and long-term deficits are being elucidated. Early ID alters the transcriptome, metabolome, structure, intracellular signaling pathways, and electrophysiology of the developing hippocampus, the brain region responsible for recognition learning and memory. Until recently, it was unclear whether these effects are directly due to a lack of iron interacting with important transcriptional, translational, or post-translational processes or to indirect effects such as hypoxia due to anemia or stress. Nonanemic genetic mouse models generated by conditionally altering expression of iron transport proteins specifically in hippocampal neurons in late gestation have led to a greater understanding of iron's role in learning and memory. The learning deficits in adulthood likely result from interactions between direct and indirect effects that contribute to abnormal hippocampal structure and plasticity.


Asunto(s)
Deficiencias de Hierro , Hierro de la Dieta/metabolismo , Aprendizaje/fisiología , Memoria/fisiología , Oligoelementos/deficiencia , Adolescente , Adulto , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Niño , Preescolar , Femenino , Expresión Génica , Hipocampo/metabolismo , Hipocampo/fisiopatología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Lactante , Recién Nacido , Ratones , Neuronas/metabolismo , Embarazo , Ratas , Serina-Treonina Quinasas TOR/metabolismo
13.
J Neurodev Disord ; 2(3): 133-43, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20824191

RESUMEN

UNLABELLED: Iron deficiency (ID) is the most common gestational micronutrient deficiency in the world, targets the fetal hippocampus and striatum and results in long-term behavioral abnormalities. These structures primarily mediate spatial and procedural memory, respectively, in the rodent but have interconnections that result in competition or cooperation during cognitive tasks. We determined whether ID-induced impairment of one alters the function of the other by genetically inducing a 40% reduction of hippocampus iron content in late fetal life in mice and measuring dorsal striatal gene expression and metabolism and the behavioral balance between the two memory systems in adulthood. Slc11a2(hipp/hipp) mice had similar striatum iron content, but 18% lower glucose and 44% lower lactate levels, a 30% higher phosphocreatine:creatine ratio, and reduced iron transporter gene expression compared to wild type (WT) littermates, implying reduced striatal metabolic function. Slc11a2(hipp/hipp) mice had longer mean escape times on a cued task paradigm implying impaired procedural memory. Nevertheless, when hippocampal and striatal memory systems were placed in competition using a Morris Water Maze task that alternates spatial navigation and visual cued responses during training, and forces a choice between hippocampal and striatal strategies during probe trials, Slc11a2(hipp/hipp) mice used the hippocampus-dependent response less often (25%) and the visual cued response more often (75%) compared to WT littermates that used both strategies approximately equally. Hippocampal ID not only reduces spatial recognition memory performance but also affects systems that support procedural memory, suggesting an altered balance between memory systems. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1007/s11689-010-9049-0) contains supplementary material, which is available to authorized users.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA