Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Behav Brain Res ; 320: 457-463, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27789343

ABSTRACT

The brain could be exposed to irradiation as part of a nuclear accident, radiological terrorism (dirty bomb scenario) or a medical radiological procedure. In the context of accidents or terrorism, there is considerable interest in compounds that can mitigate radiation-induced injury when treatment is initiated a day or more after the radiation exposure. As it will be challenging to determine the radiation exposure an individual has received within a relatively short time frame, it is also critical that the mitigating agent does not negatively affect individuals, including emergency workers, who might be treated, but who were not exposed. Alterations in hippocampus-dependent cognition often characterize radiation-induced cognitive injury. The catalytic ROS scavenger EUK-207 is a member of the class of metal-containing salen manganese (Mn) complexes that suppress oxidative stress, including in the mitochondria, and have been shown to mitigate radiation dermatitis, promote wound healing in irradiated skin, and mitigate vascular injuries in irradiated lungs. As the effects of EUK-207 against radiation injury in the brain are not known, we assessed the effects of EUK-207 on sham-irradiated animals and the ability of EUK-207 to mitigate radiation-induced cognitive injury. The day following irradiation or sham-irradiation, the mice started to receive EUK-207 and were cognitively tested 3 months following exposure. Mice irradiated at a dose of 15Gy showed cognitive impairments in the water maze probe trial. EUK-207 mitigated these impairments while not affecting cognitive performance of sham-irradiated mice in the water maze probe trial. Thus, EUK-207 has attractive properties and should be considered an ideal candidate to mitigate radiation-induced cognitive injury.


Subject(s)
Cognition Disorders/drug therapy , Cognition Disorders/etiology , Organometallic Compounds/therapeutic use , Radiation Injuries, Experimental/complications , Analysis of Variance , Animals , Conditioning, Psychological/drug effects , Dose-Response Relationship, Radiation , Fear/drug effects , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Superoxide Dismutase/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
2.
Int J Obes (Lond) ; 40(10): 1574-1581, 2016 10.
Article in English | MEDLINE | ID: mdl-27163745

ABSTRACT

BACKGROUND: The Apolipoprotein E (APOE) gene encodes for three isoforms in the human population (APOE2, APOE3 and APOE4). Whereas the role of APOE in lipid metabolism is well characterized, the specific metabolic signatures of the APOE isoforms during metabolic disorders, remain unclear. OBJECTIVE: To elucidate the molecular underpinnings of APOE-directed metabolic alterations, we tested the hypothesis that APOE4 drives a whole-body metabolic shift toward increased lipid oxidation. METHODS: We employed humanized mice in which the Apoe gene has been replaced by the human APOE*3 or APOE*4 allele to produce human APOE3 or APOE4 proteins and characterized several mechanisms of fatty-acid oxidation, lipid storage, substrate utilization and thermogenesis in those mice. RESULTS: We show that, whereas APOE4 mice gained less body weight and mass than their APOE3 counterparts on a Western-type diet (P<0.001), they displayed elevated insulin and homeostatic model assessment, markers of insulin resistance (P=0.004 and P=0.025, respectively). APOE4 mice also demonstrated a reduced respiratory quotient during the postprandial period (0.95±0.03 versus 1.06±0.03, P<0.001), indicating increased usage of lipids as opposed to carbohydrates as a fuel source. Finally, APOE4 mice showed increased body temperature (37.30±0.68 versus 36.9±0.58 °C, P=0.039), augmented cold tolerance and more metabolically active brown adipose tissue compared with APOE3 mice. CONCLUSION: These data suggest that APOE4 mice may resist weight gain via an APOE4-directed global metabolic shift toward lipid oxidation and enhanced thermogenesis, and may represent a critical first step in the development of APOE-directed therapies for a large percentage of the population affected by disorders with established links to APOE and metabolism.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis , Adipose Tissue/metabolism , Apolipoprotein E4/metabolism , Fatty Acids/metabolism , Thermogenesis , Adipose Tissue/cytology , Animals , Apolipoprotein E4/genetics , Body Weight , Disease Models, Animal , Gene Transfer Techniques , Humans , Lipid Metabolism/physiology , Male , Mice , Mice, Transgenic
3.
Neurotoxicology ; 53: 132-140, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26797589

ABSTRACT

Domoic acid (DA) is an algal neurotoxin that accumulates in marine fish and shellfish. DA can move across the placenta and concentrate in amniotic fluid, which can be swallowed during late gestation. DA also transfers to infants via milk. Preclinical studies to determine effects of developmental DA expose have primarily involved DA exposure during the postnatal period and little is known about late CNS effects following prenatal DA. In the present study, we tested the hypothesis that prenatal exposure of FVB mice to low levels of DA would result in diminished social interaction and sensory motor gating associated with alterations in parvalbumin immunoreactivity in relevant brain regions undergoing development during and following DA exposure. In addition to parvalbumin, we stained with NeuN for a neuronal specific nuclear protein to determine if neuronal loss followed prenatal DA exposure. A single moderate dose of DA administered during gestation produces diminishes social investigation and alters sensorimotor gating, behavioral effects more pronounced in males than females. These behavioral changes were associated with discrete alterations in the parvalbumin-positive subtype of GABAergic neurons in the dentate gyrus and lateral amygdala.


Subject(s)
Amygdala/pathology , Interpersonal Relations , Kainic Acid/analogs & derivatives , Neurons/pathology , Neurotoxins/toxicity , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/physiopathology , Sensory Gating/drug effects , Acoustic Stimulation , Age Factors , Amygdala/drug effects , Analysis of Variance , Animals , Animals, Newborn , Disease Models, Animal , Female , Kainic Acid/toxicity , Male , Mice , Neurons/drug effects , Parvalbumins/metabolism , Phosphopyruvate Hydratase/metabolism , Pregnancy , Prepulse Inhibition/drug effects , Psychoacoustics , Sex Factors , Vocalization, Animal/drug effects
4.
Genes Brain Behav ; 11(7): 806-12, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22883220

ABSTRACT

The three major human apoE isoforms (apoE2, apoE3 and apoE4) are encoded by distinct alleles (ϵ2, ϵ3 and ϵ4). Compared with ϵ3, ϵ4 is associated with increased risk to develop Alzheimer's disease (AD), cognitive impairments in Parkinson's disease (PD), and other conditions. In contrast, a recent study indicated an increased susceptibility to the recurring and re-experiencing symptom cluster of Post-Traumatic Stress Disorder (PTSD), as well as related memory impairments, in patients carrying at least one ϵ2 allele. Contextual fear conditioning and extinction are used in human and animal models to study this symptom cluster. In this study, acquisition (day 1, training), consolidation (day 2, first day of re-exposure) and extinction (days 2-5) of conditioned contextual fear in human apoE2, apoE3 and apoE4 targeted replacement and C57BL/6J wild-type (WT) mice was investigated. Male and female apoE2 showed acquisition and retrieval of conditioned fear, but failed to exhibit extinction. In contrast, WT, apoE3 and apoE4 mice showed extinction. While apoE2 mice exhibited lower freezing in response to the context on day 2 than apoE3 and apoE4 mice, this cannot explain their extinction deficit as WT mice exhibited similar freezing levels as apoE2 mice on day 2 but still exhibited extinction. Elevating freezing through extended training preserved extinction in controls, but failed to ameliorate extinction deficits in apoE2 animals. These data along with clinical data showing an association of apoE2 with susceptibility to specific symptom clusters in PTSD supports an important role for apoE isoform in the extinction of conditioned fear.


Subject(s)
Apolipoproteins E/genetics , Extinction, Psychological , Fear , Animals , Anxiety/genetics , Conditioning, Psychological , Female , Freezing Reaction, Cataleptic , Humans , Male , Mice , Mice, Inbred C57BL , Protein Isoforms/genetics
5.
Phytomedicine ; 17(12): 940-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20382514

ABSTRACT

Potential mechanisms of Passiflora incarnata extracts and the effect of extraction methods on ingredients and biological effects were explored. Using the same batch of plant material, total flavonoid yields as measured by high-performance liquid chromatography coupled to diode array detection (HPLC-DAD) increased substantially with hot versus cold extraction methods. Whole Passiflora extract induced prominent, dose-dependent direct GABA(A) currents in hippocampal slices, but the expected modulation of synaptic GABA(A) currents was not seen. GABA was found to be a prominent ingredient of Passiflora extract, and GABA currents were absent when amino acids were removed from the extract. Five different extracts, prepared from a single batch of Passiflora incarnata, were administered to CF-1 mice for 1 week in their drinking water prior to evaluation of their behavioral effects. Anticonvulsant effects against PTZ-induced seizures were seen in mice that received 2 of the 5 Passiflora extracts. Instead of the anxiolytic effects described by others, anxiogenic effects in the elevated plus maze were seen in mice receiving any of the 5 Passiflora extracts.


Subject(s)
Anticonvulsants/therapeutic use , Anxiety/drug therapy , GABA Agonists/therapeutic use , Hippocampus/drug effects , Passiflora/chemistry , Plant Extracts/therapeutic use , Seizures/drug therapy , Animals , Anticonvulsants/pharmacology , Anxiety/chemically induced , Dose-Response Relationship, Drug , Flavonoids/adverse effects , Flavonoids/pharmacology , Flavonoids/therapeutic use , GABA Agonists/pharmacology , Hippocampus/physiology , Male , Maze Learning , Mice , Mice, Inbred Strains , Pentylenetetrazole , Phytotherapy , Plant Extracts/adverse effects , Plant Extracts/pharmacology , Rats , Rats, Sprague-Dawley , Temperature , gamma-Aminobutyric Acid/pharmacology , gamma-Aminobutyric Acid/therapeutic use
6.
Exp Neurol ; 217(1): 55-62, 2009 May.
Article in English | MEDLINE | ID: mdl-19416674

ABSTRACT

In rodents, spatial learning and memory tests require navigation, whereas in nonhuman primates these tests generally do not involve a navigational component, thus assessing nonhomologous neural systems. To allow closer parallels between rodent and primate studies, we developed a navigational spatial learning and memory task for nonhuman primates and assessed the performance of elderly (19-25 years) female rhesus monkeys (Macaca mulatta). The animals were allowed to navigate in a room containing a series of food ports. After they learned to retrieve food from the ports, a single port was repeatedly baited and the animals were tested until they learned the correct location. The location of the baited port was then changed (shift position). We also determined whether test performance was associated with circadian activity measured with accelerometers. Performance measures included trials to criterion, search strategies, and several indices of circadian activity. Animals learned the task as reflected in their search strategies. Correlations were found between the number of initial or shift trials and circadian activity parameters including day activity, dark:light activity ratio, sleep latency, and wake bouts. Thus, disruptions in circadian rhythms in nonhuman primates are associated with poorer performance on this novel test. These data support the usefulness of this spatial navigational test to assess spatial learning and memory in rhesus monkeys and the importance of circadian activity in performance.


Subject(s)
Aging/physiology , Association Learning/physiology , Circadian Rhythm/physiology , Macaca mulatta/physiology , Memory/physiology , Spatial Behavior/physiology , Animals , Discrimination, Psychological , Female , Statistics as Topic
7.
Radiats Biol Radioecol ; 48(2): 167-70, 2008.
Article in English | MEDLINE | ID: mdl-18666648

ABSTRACT

Cranial irradiation is associated with long-term cognitive impairments, including deficits in hippocampus-dependent learning and memory. Not all people exposed to cranial radiation develop cognitive injury, suggesting the involvement of genetic risk factors. There may also be sex differences in susceptibility to develop radiation-induced cognitive injury. The three major human apolipoprotein E (apoE) isoforms are encoded by distinct alleles (epsilon2, epsilon3, and epsilon4). Compared with epsilon3, epsilon4 increases the risk of cognitive impairments following various challenges while epsilon2 provides relative protection. Women are at higher risk to develop Alzheimer's disease (AD) than men, particularly those carrying epsilon4. In previous experiments using male and female mice expressing human apoE-isoforms E2, E3 or E4 under the mouse apoE promoter, we showed that cranial irradiation with 137Cs (10 Gy) results in hippocampus-dependent cognitive impairments that are sex- and apoE-isoform dependent. 137Cs is a form of irradiation often used in the clinical setting. To investigate whether 56Fe irradiation also has sex- and apoE-isoform dependent effects on hippocampus-dependent cognitive function in human apoE mice, we sham-irradiated and irradiated 2-month old male and female human apoE mice at 3 Gy and assessed their performance in a passive avoidance learning and memory test three to five months later.


Subject(s)
Apolipoproteins E/metabolism , Avoidance Learning/radiation effects , Behavior, Animal/radiation effects , Iron Radioisotopes , Memory/radiation effects , Radiation Injuries, Experimental/psychology , Animals , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Isoforms/metabolism , Whole-Body Irradiation
8.
Neuroscience ; 147(1): 6-17, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17509769

ABSTRACT

To determine effects of APOE epsilon4 (epsilon4) on cognitive performance of healthy elderly, 116 nondemented elders (mean age 81 years) were cognitive tested. The established tests Faces, Family Pictures, Spatial Span Forward and Backward, and the object recognition and spatial navigation tests developed in our laboratory were used as cognitive tests. Salivary samples were collected to determine APOE genotype and salivary testosterone and cortisol levels. Non-epsilon4- and epsilon4-carrying men and women did not differ in age, Mini-Mental State Examination, Wide Range Achievement Test-Reading, Beck Anxiety Inventory, or reaction time scores. There was an effect of epsilon4 on the object recognition and spatial navigation tests, with non-epsilon4 carriers outperforming epsilon4 carriers, but not in the other cognitive tests. No relationship was found for sex and epsilon4 status or sex and performance during the hidden session of Memory Island. In men, salivary cortisol levels correlated with object recognition. These results show that object recognition and spatial navigation tests are useful to assess cognitive function in the elderly.


Subject(s)
Apolipoprotein E4/genetics , Cognition/physiology , Pattern Recognition, Visual/physiology , Recognition, Psychology/physiology , Spatial Behavior/physiology , Aged, 80 and over , Analysis of Variance , Apolipoprotein E2/metabolism , Female , Humans , Hydrocortisone/metabolism , Male , Matched-Pair Analysis , Photic Stimulation , Reaction Time/physiology , Reference Values , Saliva/metabolism , Sex Factors , Single-Blind Method , Space Perception/physiology , Statistics, Nonparametric , Testosterone/metabolism
9.
Cell Mol Life Sci ; 64(6): 735-41, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17310279

ABSTRACT

Histamine might have an important role in brain development. However, most studies have focused on short-term effects of histamine receptor-mediated signaling on brain function in adulthood. Little is known about the potential long-term effects of histamine receptor-mediated signaling during development on brain function in adulthood. We hypothesize that increased postsynaptic histamine receptor-mediated signaling during development has detrimental effects on brain function in adulthood. Our data support this hypothesis. In the developing mouse brain, histamine H3 receptor blockade, which increases histamine release, has detrimental sex-dependent effects on object recognition, spatial learning in the water maze, and pre-pulse inhibition in adulthood. Our data also support the hypothesis that histamine mediates the detrimental long-term sex-dependent effects of methamphetamine exposure early in life on these brain functions in adulthood. Therefore, increased efforts are warranted to carefully evaluate the effects of drugs that directly or indirectly affect histamine receptor-mediated signaling during development on cognitive function later in life.


Subject(s)
Aging/physiology , Brain/embryology , Brain/metabolism , Receptors, Histamine/metabolism , Signal Transduction , Animals , Brain/drug effects , Histamine/metabolism , Humans , Methamphetamine/adverse effects , Signal Transduction/drug effects
10.
Neuroscience ; 137(2): 413-23, 2006.
Article in English | MEDLINE | ID: mdl-16330151

ABSTRACT

Understanding cognitive aging is becoming more important as the elderly population grows. Here, the effects of age and sex on learning and memory performance were compared in female and male young (3-4 months old) middle-aged (10-12 months old) and old (18-20 months old) wild-type C57BL/6J mice. Old males and females performed worse than young or middle-aged mice in novel location, but not novel object recognition tasks. Old mice, of both sexes, also showed impaired spatial water maze performance during training compared with young or middle-aged mice, however only old females failed to show robust spatial bias during probe trials. While there was no age-difference in passive avoidance performance for males, females showed an age-related decline. There was no difference in cognitive performance between young and middle-age mice of either sex on any task. Cognitive performance was associated with alterations in immunoreactivity of microtubule-associated protein 2-positive dendrites and synaptophysin-positive pre-synaptic terminals in hippocampal CA1, CA3, and dentate, entorhinal cortex, and central nucleus of amygdala. Overall, microtubule-associated protein 2 immunoreactivity was increased in old females compared with both young and middle-age females with no significant difference in males. In contrast, synaptophysin immunoreactivity increased from young to middle-age in females, and from middle-age to old in males; females had higher levels of synaptophysin immunoreactivity than males in middle-age only. Elevated levels of microtubule-associated protein 2 and synaptophysin may constitute a compensatory response to age-related functional decline in mice.


Subject(s)
Aging/metabolism , Brain/metabolism , Cognition Disorders/metabolism , Memory Disorders/metabolism , Microtubule-Associated Proteins/metabolism , Synaptophysin/metabolism , Adaptation, Physiological/physiology , Aging/pathology , Amygdala/metabolism , Amygdala/pathology , Amygdala/physiopathology , Animals , Brain/pathology , Brain/physiopathology , Cognition Disorders/physiopathology , Dendrites/metabolism , Dendrites/ultrastructure , Female , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Immunohistochemistry , Male , Maze Learning/physiology , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Sex Characteristics , Up-Regulation/physiology
12.
Amino Acids ; 26(1): 37-43, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14752614

ABSTRACT

Neurons of the hypothalamo-neurohypophyseal system (HNS) are known to contain high amounts of neuronal nitric oxide (NO) synthase (nNOS). NO produced by those neurons is commonly supposed to be involved as modulator in the release of the two nonapeptides vasopressin (AVP) and oxytocin into the blood stream. Previous studies showed that forced swimming fails to increase the release of AVP into the blood stream while its secretion into the hypothalamus is triggered. We investigated here whether hypothalamically acting NO contributes to the control of the AVP release into blood under forced swimming conditions. Intracerebral microdialysis and in situ hybridization were employed to analyze the activity of the nitrergic system within the supraoptic nucleus (SON), the hypothalamic origin of the HNS. A 10-min forced swimming session failed to significantly alter the local NO release as indicated both by nitrite and, the main by-product of NO synthesis, citrulline levels in microdialysis samples collected from the SON. Microdialysis administration of NO directly into the SON increased the concentration of AVP in plasma samples collected during simultaneous forced swimming. In an additional experiment the effect of the defined stressor exposure on the concentration of mRNA coding for nNOS within the SON was investigated by in situ hybridization. Forced swimming increased the expression of nNOS mRNA at two and four hours after onset of the stressor compared to untreated controls. Taken together, our results imply that NO within the SON does not contribute to the regulation of the secretory activity of HNS neurons during acute forced swimming. Increased nNOS mRNA in the SON after forced swimming and the increase in AVP release in the presence of exogenous NO under forced swimming points to a possible role of NO in the regulation of the HNS under repeated stressor exposure.


Subject(s)
Nitrergic Neurons/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/blood , Supraoptic Nucleus/metabolism , Vasopressins/blood , Animals , Citrulline/blood , Citrulline/chemistry , Hypothalamo-Hypophyseal System/metabolism , Male , Oxytocin/blood , Oxytocin/metabolism , Physical Conditioning, Animal , Physical Exertion , Rats , Rats, Wistar , Supraoptic Nucleus/chemistry , Vasopressins/chemistry
13.
Nat Genet ; 25(1): 87-90, 2000 May.
Article in English | MEDLINE | ID: mdl-10802663

ABSTRACT

Triglycerides (or triacylglycerols) represent the major form of stored energy in eukaryotes. Triglyceride synthesis has been assumed to occur primarily through acyl CoA:diacylglycerol transferase (Dgat), a microsomal enzyme that catalyses the final and only committed step in the glycerol phosphate pathway. Therefore, Dgat has been considered necessary for adipose tissue formation and essential for survival. Here we show that Dgat-deficient (Dgat-/-) mice are viable and can still synthesize triglycerides. Moreover, these mice are lean and resistant to diet-induced obesity. The obesity resistance involves increased energy expenditure and increased activity. Dgat deficiency also alters triglyceride metabolism in other tissues, including the mammary gland, where lactation is defective in Dgat-/- females. Our findings indicate that multiple mechanisms exist for triglyceride synthesis and suggest that the selective inhibition of Dgat-mediated triglyceride synthesis may be useful for treating obesity.


Subject(s)
Acyltransferases/deficiency , Acyltransferases/genetics , Obesity/metabolism , Triglycerides/biosynthesis , Absorption , Animals , Body Temperature Regulation/genetics , Calorimetry , Diacylglycerol O-Acyltransferase , Dietary Fats/administration & dosage , Energy Metabolism/genetics , Female , Insulin Resistance/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/enzymology , Obesity/genetics , Triglycerides/genetics
15.
J Neurosci ; 20(5): 2064-71, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10684907

ABSTRACT

Several neurological diseases are frequently accompanied by dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis. The HPA axis regulates the secretion of glucocorticoids (GCs), which play important roles in diverse brain functions, including cognition, emotion, and feeding. Under physiological conditions, GCs are adaptive and beneficial; however, prolonged elevations in GC levels may contribute to neurodegeneration and brain dysfunction. In the current study, we demonstrate that apolipoprotein E (apoE) deficiency results in age-dependent dysregulation of the HPA axis through a mechanism affecting primarily the adrenal gland. Apoe(-/-) mice, which develop neurodegenerative alterations as they age, had an age-dependent increase in basal adrenal corticosterone content and abnormally increased plasma corticosterone levels after restraint stress, whereas their plasma and pituitary adrenocorticotropin levels were either unchanged or lower than those in controls. HPA axis dysregulation was associated with behavioral and metabolic alterations. When anxiety levels were assessed in the elevated plus maze, Apoe(-/-) mice showed more anxiety than wild-type controls. Apoe(-/-) mice also showed reduced activity in the open field. Finally, Apoe(-/-) mice showed age-dependent increases in food and water intake, stomach and body weights, and decreases in brown and white adipose tissues. These results support a key role for apoE in the tonic inhibition of steroidogenesis and HPA axis activity and have important implications for the behavioral analysis of Apoe(-/-) mice.


Subject(s)
Apolipoproteins E/genetics , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiopathology , Adrenal Glands/chemistry , Adrenal Glands/drug effects , Adrenal Glands/metabolism , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/chemistry , Adrenocorticotropic Hormone/pharmacology , Animals , Anxiety/metabolism , Anxiety/physiopathology , Behavior, Animal/physiology , Corticosterone/blood , Drinking , Eating , Energy Metabolism/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Stress, Physiological/metabolism , Stress, Physiological/physiopathology
16.
Mol Neurobiol ; 18(1): 1-22, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9824846

ABSTRACT

Increasing evidence suggests that the detrimental effects of glucocorticoid (GC) hypersecretion occur by activation of the hypothalamic-pituitary-adrenal (HPA) axis in several human pathologies, including obesity, Alzheimer's disease, AIDS dementia, and depression. The different patterns of response by the HPA axis during chronic activation are an important consideration in selecting an animal model to assess HPA axis function in a particular disorder. This article will discuss how chronic HPA axis activation and GC hypersecretion affect hippocampal function and contribute to the development of obesity. In the brain, the hippocampus has the highest concentration of GC receptors. Chronic stress or corticosterone treatment induces neuropathological alterations, such as dendritic atrophy in hippocampal neurons, which are paralleled by cognitive deficits. Excitatory amino acid (EAA) neurotransmission has been implicated in chronic HPA axis activation. EAAs play a major role in neuroendocrine regulation. Hippocampal dendritic atrophy may involve alterations in EAA transporter function, and decreased EAA transporter function may also contribute to chronic HPA axis activation. Understanding the molecular mechanisms of HPA axis activation will likely advance the development of therapeutic interventions for conditions in which GC levels are chronically elevated.


Subject(s)
Glucocorticoids/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Pituitary-Adrenal System/physiopathology , Acquired Immunodeficiency Syndrome/physiopathology , Adrenocorticotropic Hormone/physiology , Amygdala/physiology , Animals , Arginine Vasopressin/physiology , Aspartic Acid/physiology , Chronic Disease , Cognition Disorders/etiology , Corticotropin-Releasing Hormone/physiology , Feedback , Glutamic Acid/physiology , Hippocampus/physiopathology , Humans , Hydrocortisone/metabolism , Memory Disorders/etiology , Mice , Models, Biological , Obesity/etiology , Rats , Stress, Physiological/physiopathology , Synaptic Transmission
17.
Proc Natl Acad Sci U S A ; 95(18): 10914-9, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9724804

ABSTRACT

Apolipoprotein E (apoE) mediates the redistribution of lipids among cells and is expressed at highest levels in brain and liver. Human apoE exists in three major isoforms encoded by distinct alleles (epsilon2, epsilon3, and epsilon4). Compared with APOE epsilon2 and epsilon3, APOE epsilon4 increases the risk of cognitive impairments, lowers the age of onset of Alzheimer's disease (AD), and decreases the response to AD treatments. Besides age, inheritance of the APOE epsilon4 allele is the most important known risk factor for the development of sporadic AD, the most common form of this illness. Although numerous hypotheses have been advanced, it remains unclear how APOE epsilon4 might affect cognition and increase AD risk. To assess the effects of distinct human apoE isoforms on the brain, we have used the neuron-specific enolase (NSE) promoter to express human apoE3 or apoE4 at similar levels in neurons of transgenic mice lacking endogenous mouse apoE. Compared with NSE-apoE3 mice and wild-type controls, NSE-apoE4 mice showed impairments in learning a water maze task and in vertical exploratory behavior that increased with age and were seen primarily in females. These findings demonstrate that human apoE isoforms have differential effects on brain function in vivo and that the susceptibility to apoE4-induced deficits is critically influenced by age and gender. These results could be pertinent to cognitive impairments observed in human APOE epsilon4 carriers. NSE-apoE mice and similar models may facilitate the preclinical assessment of treatments for apoE-related cognitive deficits.


Subject(s)
Apolipoproteins E/physiology , Neurons/metabolism , Age Factors , Alleles , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Avoidance Learning , Exploratory Behavior , Female , Humans , Immunohistochemistry , Maze Learning , Mice , Mice, Knockout , Mice, Transgenic , Neurons/enzymology , Phosphopyruvate Hydratase/genetics , Sex Factors
19.
Brain Res Brain Res Rev ; 26(2-3): 320-6, 1998 May.
Article in English | MEDLINE | ID: mdl-9651548

ABSTRACT

The cytokines are a large and diverse family of polypeptide regulators with multiple regulatory functions that have been comprehensively evaluated in the immune system under strictly controlled experimental conditions. These peptide signals exhibit often unpredictable interactions when evaluated for their pathophysiological involvement in specific inflammatory conditions in vivo. In our joint efforts to understand the basis for early pathophysiological changes in the brains of HIV-infected subjects, we have developed animal models for lentivirus infections, and assessed the actions of various cytokines acutely on transmitter release properties in vitro, and in an in vivo transgenic mouse model. IL1beta, IL2, IL6, and IFNalpha will each enhance the release of AVP in slices of rat hypothalamus and amygdala. TGFbeta selectively blocks the ability of ACh to release AVP from hypothalamus or amygdala, but has no effects on the release stimulated by other cytokines. IFNalpha, but not TGFbeta will also activate CRH release; as with AVP, TGF selectively blocks the ACh-stimulated CRH release in both amygdala and hypothalamus. The IFNalpha-stimulated release of AVP and CRH appears to be mediated by cyclic GMP production, and this release by IFNalpha and IL-2 may be mediated in part by activation of constitutive nitric oxide synthase. These combined in vitro actions would suggest that cns cytokine actions should upregulate the hypothalamic pituitary adrenal axis. In a transgenic mouse model with increased astrocytic expression and release of the cytokine IL6, the HPA axis is upregulated, but the effect seems attributable to adrenocortical hypersensitization to ACTH. Lastly, in studies of cytokine mediated effects on astrocytic uptake of the excitatory transmitter glutamate, the reactive oxygen species hydrogen peroxide and peroxynitrite, but not nitric oxide, inhibited glutamate uptake in a concentration-dependent manner. Although superoxide and nitric oxide had no effect by themselves on the rate of glutamate uptake by astrocytes, the same cultures did respond to nitric oxide with a sustained increase in cytoplasmic free calcium. Thus while reactive oxygen species do provide a potential path to neurotoxicity but one apparently not involving nitric oxide. These various data provide important opportunities for early therapeutic interventions in neuro-inflammatory states such as Neuro-AIDS.


Subject(s)
Brain/physiopathology , Cytokines/physiology , Inflammation/physiopathology , Neurons/physiology , Neurosecretory Systems/physiology , Animals , Brain/immunology , Brain/physiology , Disease Models, Animal , HIV Infections/immunology , HIV Infections/physiopathology , Humans , Hypothalamo-Hypophyseal System/immunology , Hypothalamo-Hypophyseal System/physiology , Mice , Neurons/immunology , Neurosecretory Systems/immunology , Neurosecretory Systems/physiopathology , Neurotransmitter Agents/metabolism , Rats
20.
J Biol Chem ; 273(20): 12548-54, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9575214

ABSTRACT

Excitatory neurotransmitters such as glutamate are required for the normal functioning of the central nervous system but can trigger excitotoxic neuronal injury if allowed to accumulate to abnormally high levels. Their extracellular levels are controlled primarily by transmitter uptake into astrocytes. Here, we demonstrate that the amyloid protein precursor may participate in the regulation of this important process. The amyloid protein precursor has been well conserved through evolution, and a number of studies indicate that it may function as an endogenous excitoprotectant. However, the mechanisms underlying this neuroprotective capacity remain largely unknown. At moderate levels of expression, human amyloid protein precursors increased glutamate/aspartate uptake in brains of transgenic mice, with the 751-amino acid isoform showing greater potency than the 695-amino acid isoform. Cerebral glutamate/aspartate transporter protein levels were higher in transgenic mice than in non-transgenic controls, whereas transporter mRNA levels were unchanged. Amyloid protein precursor-dependent stimulation of aspartate uptake by cultured primary astrocytes was associated with increases in protein kinase A and C activity and could be blocked by inhibitors of these kinases. The stimulation of astroglial excitatory amino acid transport by amyloid protein precursors could protect the brain against excitotoxicity and may play an important role in neurotransmission.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Aspartic Acid/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Astrocytes/metabolism , Biological Transport , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Humans , Male , Mice , Mice, Transgenic , Neocortex/metabolism , Protein Kinase C/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Synaptic Transmission
SELECTION OF CITATIONS
SEARCH DETAIL
...