Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
J Gerontol A Biol Sci Med Sci ; 76(1): 23-31, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32154567

ABSTRACT

The accumulation of amyloid-ß (Aß) is a characteristic event in the pathogenesis of Alzheimer's disease (AD). Aquaporin 1 (AQP1) is a membrane water channel protein belonging to the AQP family. AQP1 levels are elevated in the cerebral cortex during the early stages of AD, but the role of AQP1 in AD pathogenesis is unclear. We first determined the expression and distribution of AQP1 in brain tissue samples of AD patients and two AD mouse models (3xTg-AD and 5xFAD). AQP1 accumulation was observed in vulnerable neurons in the cerebral cortex of AD patients, and in neurons affected by the Aß or tau pathology in the 3xTg-AD and 5xFAD mice. AQP1 levels increased in neurons as aging progressed in the AD mouse models. Stress stimuli increased AQP1 in primary cortical neurons. In response to cellular stress, AQP1 appeared to translocate to endocytic compartments of ß- and γ-secretase activities. Ectopic expression of AQP1 in human neuroblastoma cells overexpressing amyloid precussir protein (APP) with the Swedish mutations reduced ß-secretase (BACE1)-mediated cleavage of APP and reduced Aß production without altering the nonamyloidogenic pathway. Conversely, knockdown of AQP1 enhanced BACE1 activity and Aß production. Immunoprecipitation experiments showed that AQP1 decreased the association of BACE1 with APP. Analysis of a human database showed that the amount of Aß decreases as the expression of AQP1 increases. These results suggest that the upregulation of AQP1 is an adaptive response of neurons to stress that reduces Aß production by inhibiting the binding between BACE1 and APP.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Protein Precursor/physiology , Amyloid/biosynthesis , Aquaporin 1/physiology , Alzheimer Disease/metabolism , Animals , Aquaporin 1/metabolism , Disease Models, Animal , Humans , Mice , Neurons/metabolism
2.
Brain Behav Immun ; 48: 301-12, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26021559

ABSTRACT

Neuroinflammation in the central nervous system is detrimental for learning and memory, as evident form epidemiological studies linking developmental defects and maternal exposure to harmful pathogens. Postnatal infections can also induce neuroinflammatory responses with long-term consequences. These inflammatory responses can lead to motor deficits and/or behavioral disabilities. Toll like receptors (TLRs) are a family of innate immune receptors best known as sensors of microbial-associated molecular patterns, and are the first responders to infection. TLR2 forms heterodimers with either TLR1 or TLR6, is activated in response to gram-positive bacterial infections, and is expressed in the brain during embryonic development. We hypothesized that early postnatal TLR2-mediated neuroinflammation would adversely affect cognitive behavior in the adult. Our data indicate that postnatal TLR2 activation affects learning and memory in adult mice in a heterodimer-dependent manner. TLR2/6 activation improved motor function and fear learning, while TLR2/1 activation impaired spatial learning and enhanced fear learning. Moreover, developmental TLR2 deficiency significantly impairs spatial learning and enhances fear learning, stressing the involvement of the TLR2 pathway in learning and memory. Analysis of the transcriptional effects of TLR2 activation reveals both common and unique transcriptional programs following heterodimer-specific TLR2 activation. These results imply that adult cognitive behavior could be influenced in part, by activation or alterations in the TLR2 pathway at birth.


Subject(s)
Learning/physiology , Memory/physiology , Motor Skills/physiology , Neurons/metabolism , Toll-Like Receptor 2/metabolism , Animals , Conditioning, Psychological/physiology , Exploratory Behavior/physiology , Fear/physiology , Mice , Mice, Knockout , Rotarod Performance Test , Spatial Learning/physiology , Toll-Like Receptor 2/genetics
4.
Neuromolecular Med ; 16(2): 292-307, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24272426

ABSTRACT

Multiple sclerosis (MS) is an inflammatory autoimmune disease of the central nervous system (CNS) involving demyelinating and neurodegenerative processes. Several of the major pathological CNS alterations and behavioral deficits of MS are recapitulated in the experimental autoimmune encephalitis (EAE) mouse model in which the disease process is induced by administration of myelin peptides. Development of EAE requires infiltration of inflammatory cytokine-generating monocytes and macrophages, and auto-reactive T cells, into the CNS. Very late antigen-4 (VLA-4, α4ß1) is an integrin molecule that plays a role in inflammatory responses by facilitating the migration of leukocytes across the blood-brain barrier during inflammatory disease, and antibodies against VLA-4 exhibit therapeutic efficacy in mouse and monkey MS models. Here, we report that the tellurium compound AS101 (ammonium trichloro (dioxoethylene-o,o') tellurate) ameliorates EAE by inhibiting monocyte and T cell infiltration into the CNS. CD49d is an alpha subunit of the VLA-4 (α4ß1) integrin. During the peak stage of EAE, AS101 treatment effectively ameliorated the disease process by reducing the number of CD49d(+) inflammatory monocyte/macrophage cells in the spinal cord. AS101 treatment markedly reduced the pro-inflammatory cytokine levels, while increasing anti-inflammatory cytokine levels. In contrast, AS101 treatment did not affect the peripheral populations of CD11b(+) monocytes and macrophages. AS101 treatment reduced the infiltration of CD4(+) and CD49(+)/VLA4 T cells. In addition, treatment of T cells from MS patients with AS101 resulted in apoptosis, while such treatment did not affect T cells from healthy donors. These results suggest that AS101 reduces accumulation of leukocytes in the CNS by inhibiting the activity of the VLA-4 integrin and provide a rationale for the potential use of Tellurium IV compounds for the treatment of MS.


Subject(s)
Cell Movement/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Ethylenes/therapeutic use , Immunologic Factors/therapeutic use , Integrin alpha4beta1/antagonists & inhibitors , Monocytes/drug effects , Spinal Cord/immunology , T-Lymphocyte Subsets/drug effects , Animals , Apoptosis/drug effects , Blood-Brain Barrier/immunology , Brain/immunology , Brain/pathology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Chemotaxis, Leukocyte/drug effects , Cytokines/biosynthesis , Cytokines/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Ethylenes/pharmacology , Female , Humans , Immunologic Factors/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Monocytes/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Spinal Cord/metabolism , Spinal Cord/pathology , Spleen/metabolism , T-Lymphocyte Subsets/immunology
5.
Brain Res ; 1529: 200-8, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23856323

ABSTRACT

Age-associated dysregulation of sleep can be worsened by Alzheimer's disease (AD). AD and sleep restriction both impair cognition, yet it is unknown if mild chronic sleep restriction modifies the proteopathic processes involved in AD. The goal of this work was to test the hypothesis that sleep restriction worsens memory impairments, and amyloid ß-peptide (Aß) and pTau accumulations in the brain in a mouse model of AD, with a focus on a role for circulating glucocorticoids (GC). Male 3xTgAD mice were subjected to sleep restriction (SR) for 6h/day for 6 weeks using the modified multiple platform technique, and behavioral (Morris water maze, fear conditioning, open field) and biochemical (immunoblot) outcomes were compared to mice undergoing daily cage transfers (large cage control; LCC) as well as control mice that remained in their home cage (control; CTL). At one week, both LCC and SR mice displayed significant elevations in plasma corticosterone compared to CTL (p<0.002). By four weeks, SR mice displayed a two-fold increase in circulating corticosterone levels compared to CTL. Behavioral data indicated deficits in contextual and cued memory in SR mice that were not present for LCC or CTL (p<0.04). Both Aß and pTau levels increased in the cortex of SR mice compared to CTL and LCC; however these changes were not noted in the hippocampus. Significant positive correlations between cortical Aß and pTau levels and circulating corticosterone indicate a potential role for GCs in mediating behavioral and biochemical changes observed after sleep restriction in a mouse model of AD.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Cerebral Cortex/metabolism , Memory Disorders/etiology , Sleep Deprivation/physiopathology , tau Proteins/metabolism , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Conditioning, Psychological/physiology , Corticosterone/blood , Disease Models, Animal , Exploratory Behavior , Fear/psychology , Humans , Male , Maze Learning , Memory Disorders/blood , Mice , Mice, Transgenic , Mutation/genetics , Presenilin-1/genetics , tau Proteins/genetics
6.
Neuromolecular Med ; 15(2): 351-63, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23519441

ABSTRACT

Tomosyn, a syntaxin-binding protein, is known to inhibit vesicle priming and synaptic transmission via interference with the formation of SNARE complexes. Using a lentiviral vector, we specifically overexpressed tomosyn1 in hippocampal dentate gyrus neurons in adult mice. Mice were then subjected to spatial learning and memory tasks and electrophysiological measurements from hippocampal slices. Tomosyn1-overexpression significantly impaired hippocampus-dependent spatial memory while tested in the Morris water maze. Further, tomosyn1-overexpressing mice utilize swimming strategies of lesser cognitive ability in the Morris water maze compared with control mice. Electrophysiological measurements at mossy fiber-CA3 synapses revealed impaired paired-pulse facilitation in the mossy fiber of tomosyn1-overexpressing mice. This study provides evidence for novel roles for tomosyn1 in hippocampus-dependent spatial learning and memory, potentially via decreased synaptic transmission in mossy fiber-CA3 synapses. Moreover, it provides new insight regarding the role of the hippocampal dentate gyrus and mossy fiber-CA3 synapses in swimming strategy preference, and in learning and memory.


Subject(s)
CA3 Region, Hippocampal/physiopathology , Dentate Gyrus/physiopathology , Learning Disabilities/genetics , Memory Disorders/genetics , Nerve Tissue Proteins/physiology , R-SNARE Proteins/physiology , Animals , Bacterial Proteins/genetics , CA3 Region, Hippocampal/metabolism , Dentate Gyrus/metabolism , Exploratory Behavior/physiology , Genes, Reporter , Genetic Vectors , Learning Disabilities/physiopathology , Lentivirus , Luminescent Proteins/genetics , Male , Maze Learning , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Mossy Fibers, Hippocampal/physiopathology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuronal Plasticity/physiology , Psychomotor Performance/physiology , R-SNARE Proteins/biosynthesis , R-SNARE Proteins/genetics , Recombinant Fusion Proteins/metabolism , Swimming , Up-Regulation
7.
J Neurosci Res ; 91(5): 671-80, 2013 May.
Article in English | MEDLINE | ID: mdl-23404341

ABSTRACT

Tumor necrosis factor-α (TNF) plays a prominent role in the brain damage and functional deficits that result from ischemic stroke. It was recently reported that the thalidomide analog 3,6'-dithiothalidomide (3,6'-DT) can selectively inhibit the synthesis of TNF in cultured cells. We therefore tested the therapeutic potential of 3,6'-DT in a mouse model of focal ischemic stroke. Administration of 3,6'-DT immediately prior to a stroke or within 3 hr after the stroke reduced infarct volume, neuronal death, and neurological deficits, whereas thalidomide was effective only when administered prior to stroke. Neuroprotection was accompanied by decreased inflammation; 3,6'-DT-treated mice exhibited reduced expression of TNF, interleukin-1ß, and inducible nitric oxide synthase; reduced numbers of activated microglia/macrophages, astrocytes, and neutrophils; and reduced expression of intercellular adhesion molecule-1 in the ischemic brain tissue. 3,6'-DT treatment attenuated stroke-induced disruption of the blood-brain barrier by a mechanism that appears to involve suppression of matrix metalloproteinase-9 and preservation of occludin. Treatment with 3,6'-DT did not reduce ischemic brain damage in mice lacking TNF receptors, consistent with a critical role for suppression of TNF production and TNF signaling in the therapeutic action of 3,6'-DT. These findings suggest that anti-inflammatory mechanisms underlie the therapeutic actions of 3,6-DT in an animal model of stroke.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Encephalitis/drug therapy , Encephalitis/etiology , Stroke/complications , Stroke/drug therapy , Thalidomide/analogs & derivatives , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Brain Infarction/etiology , Brain Infarction/prevention & control , Cell Death/drug effects , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , In Situ Nick-End Labeling , Intercellular Adhesion Molecule-1/metabolism , Interleukin-3/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/genetics , Neutrophils/drug effects , Neutrophils/metabolism , Nitric Oxide Synthase Type II/metabolism , Recombinant Fusion Proteins/metabolism , Thalidomide/therapeutic use , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
Neurobiol Aging ; 34(6): 1530-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23276384

ABSTRACT

Alzheimer's disease (AD) involves progressive accumulation of amyloid ß-peptide (Aß) and neurofibrillary pathologies, and glucose hypometabolism in brain regions critical for memory. The 3xTgAD mouse model was used to test the hypothesis that a ketone ester-based diet can ameliorate AD pathogenesis. Beginning at a presymptomatic age, 2 groups of male 3xTgAD mice were fed a diet containing a physiological enantiomeric precursor of ketone bodies (KET) or an isocaloric carbohydrate diet. The results of behavioral tests performed at 4 and 7 months after diet initiation revealed that KET-fed mice exhibited significantly less anxiety in 2 different tests. 3xTgAD mice on the KET diet also exhibited significant, albeit relatively subtle, improvements in performance on learning and memory tests. Immunohistochemical analyses revealed that KET-fed mice exhibited decreased Aß deposition in the subiculum, CA1 and CA3 regions of the hippocampus, and the amygdala. KET-fed mice exhibited reduced levels of hyperphosphorylated tau deposition in the same regions of the hippocampus, amygdala, and cortex. Thus, a novel ketone ester can ameliorate proteopathic and behavioral deficits in a mouse AD model.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Anxiety/metabolism , Cognition Disorders/metabolism , Diet, Ketogenic/methods , tau Proteins/metabolism , Alzheimer Disease/diet therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/adverse effects , Animals , Anxiety/diet therapy , Anxiety/pathology , Cognition Disorders/diet therapy , Cognition Disorders/pathology , Disease Models, Animal , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Random Allocation , tau Proteins/adverse effects
9.
Biomaterials ; 34(9): 2194-201, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23266256

ABSTRACT

Rapid and effective wound healing requires a coordinated cellular response involving fibroblasts, keratinocytes and vascular endothelial cells (VECs). Impaired wound healing can result in multiple adverse health outcomes and, although antibiotics can forestall infection, treatments that accelerate wound healing are lacking. We now report that topical application of water soluble cerium oxide nanoparticles (Nanoceria) accelerates the healing of full-thickness dermal wounds in mice by a mechanism that involves enhancement of the proliferation and migration of fibroblasts, keratinocytes and VECs. The Nanoceria penetrated into the wound tissue and reduced oxidative damage to cellular membranes and proteins, suggesting a therapeutic potential for topical treatment of wounds with antioxidant nanoparticles.


Subject(s)
Antioxidants/pharmacology , Cerium/pharmacology , Endothelial Cells/drug effects , Keratinocytes/drug effects , Nanoparticles/chemistry , Wound Healing/drug effects , Animals , Antioxidants/chemistry , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cerium/chemistry , Endothelial Cells/cytology , Fibroblasts/cytology , Fibroblasts/drug effects , Fluorescent Antibody Technique , Humans , Keratinocytes/cytology , Mice , Mice, Inbred C57BL , Oxidative Stress , Skin/drug effects , Skin/injuries
10.
PLoS One ; 7(10): e47522, 2012.
Article in English | MEDLINE | ID: mdl-23071817

ABSTRACT

Toll-like receptors (TLRs) play essential roles in innate immunity and increasing evidence indicates that these receptors are expressed in neurons, astrocytes and microglia in the brain where they mediate responses to infection, stress and injury. Very little is known about the roles of TLRs in cognition. To test the hypothesis that TLR4 has a role in hippocampus-dependent spatial learning and memory, we used mice deficient for TLR4 and mice receiving chronic TLR4 antagonist infusion to the lateral ventricles in the brain. We found that developmental TLR4 deficiency enhances spatial reference memory acquisition and memory retention, impairs contextual fear-learning and enhances motor functions, traits that were correlated with CREB up-regulation in the hippocampus. TLR4 antagonist infusion into the cerebral ventricles of adult mice did not affect cognitive behavior, but instead affected anxiety responses. Our findings indicate a developmental role for TLR4 in shaping spatial reference memory, and fear learning and memory. Moreover, we show that central TLR4 inhibition using a TLR4 antagonist has no discernible physiological role in regulating spatial and contextual hippocampus-dependent cognitive behavior.


Subject(s)
Anxiety/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation/physiology , Hippocampus/physiology , Maze Learning/physiology , Memory/physiology , Toll-Like Receptor 4/physiology , Analysis of Variance , Animals , Conditioning, Psychological/physiology , Fear/physiology , Hippocampus/metabolism , Immunoblotting , Infusions, Intraventricular , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Knockout , Rotarod Performance Test , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics
11.
Neurobiol Aging ; 33(7): 1481.e1-5, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22209255

ABSTRACT

Huntington's disease (HD) is associated with profound autonomic dysfunction including dysregulation of cardiovascular control often preceding cognitive or motor symptoms. Brain-derived neurotrophic factor (BDNF) levels are decreased in the brains of HD patients and HD mouse models, and restoring BDNF levels prevents neuronal loss and extends survival in HD mice. We reasoned that heart rate changes in HD may be associated with altered BDNF signaling in cardiovascular control nuclei in the brainstem. Here we show that heart rate is elevated in HD (N171-82Q) mice at presymptomatic and early disease stages, and heart rate responses to restraint stress are attenuated. BDNF levels were significantly reduced in brainstem regions containing cardiovascular nuclei in HD mice and human HD patients. Central administration of BDNF restored the heart rate to control levels. Our findings establish a link between diminished BDNF expression in brainstem cardiovascular nuclei and abnormal heart rates in HD mice, and suggest a novel therapeutic target for correcting cardiovascular dysfunction in HD.


Subject(s)
Brain Stem/metabolism , Brain-Derived Neurotrophic Factor/physiology , Disease Models, Animal , Heart Rate/physiology , Huntington Disease/metabolism , Signal Transduction/physiology , Animals , Brain Stem/physiopathology , Humans , Huntington Disease/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
12.
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
13.
Hum Mol Genet ; 21(5): 963-77, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22045699

ABSTRACT

Endoplasmic reticulum (ER) stress has been implicated as an initiator or contributing factor in neurodegenerative diseases. The mechanisms that lead to ER stress and whereby ER stress contributes to the degenerative cascades remain unclear but their understanding is critical to devising effective therapies. Here we show that knockdown of Herp (Homocysteine-inducible ER stress protein), an ER stress-inducible protein with an ubiquitin-like (UBL) domain, aggravates ER stress-mediated cell death induced by mutant α-synuclein (αSyn) that causes an inherited form of Parkinson's disease (PD). Functionally, Herp plays a role in maintaining ER homeostasis by facilitating proteasome-mediated degradation of ER-resident Ca(2+) release channels. Deletion of the UBL domain or pharmacological inhibition of proteasomes abolishes the Herp-mediated stabilization of ER Ca(2+) homeostasis. Furthermore, knockdown or pharmacological inhibition of ER Ca(2+) release channels ameliorates ER stress, suggesting that impaired homeostatic regulation of Ca(2+) channels promotes a protracted ER stress with the consequent activation of ER stress-associated apoptotic pathways. Interestingly, sustained upregulation of ER stress markers and aberrant accumulation of ER Ca(2+) release channels were detected in transgenic mutant A53T-αSyn mice. Collectively, these data establish a causative link between impaired ER Ca(2+) homeostasis and chronic ER stress in the degenerative cascades induced by mutant αSyn and suggest that Herp is essential for the resolution of ER stress through maintenance of ER Ca(2+) homeostasis. Our findings suggest a therapeutic potential in PD for agents that increase Herp levels or its ER Ca(2+)-stabilizing action.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/physiology , Membrane Proteins/metabolism , Stress, Physiological , alpha-Synuclein/metabolism , Animals , Calcium Channels/metabolism , Cell Death , Endoplasmic Reticulum-Associated Degradation , HEK293 Cells , Homeostasis , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Membrane Proteins/genetics , Mice , Mice, Transgenic , Mutant Proteins/metabolism , PC12 Cells , RNA Interference , Rats , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , alpha-Synuclein/genetics
14.
Neurobiol Aging ; 33(1): 205.e1-18, 2012 Jan.
Article in English | MEDLINE | ID: mdl-20947216

ABSTRACT

Activity-dependent modulation of neuronal gene expression promotes neuronal survival and plasticity, and neuronal network activity is perturbed in aging and Alzheimer's disease (AD). Here we show that cerebral cortical neurons respond to chronic suppression of excitability by downregulating the expression of genes and their encoded proteins involved in inhibitory transmission (GABAergic and somatostatin) and Ca(2+) signaling; alterations in pathways involved in lipid metabolism and energy management are also features of silenced neuronal networks. A molecular fingerprint strikingly similar to that of diminished network activity occurs in the human brain during aging and in AD, and opposite changes occur in response to activation of N-methyl-D-aspartate (NMDA) and brain-derived neurotrophic factor (BDNF) receptors in cultured cortical neurons and in mice in response to an enriched environment or electroconvulsive shock. Our findings suggest that reduced inhibitory neurotransmission during aging and in AD may be the result of compensatory responses that, paradoxically, render the neurons vulnerable to Ca(2+)-mediated degeneration.


Subject(s)
Aging/genetics , Aging/physiology , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Cerebral Cortex/physiopathology , Gene Expression , Interneurons/physiology , Nerve Net/physiopathology , Animals , Calcium Signaling/genetics , Cell Survival/genetics , Cells, Cultured , Cerebral Cortex/cytology , Electroshock , Energy Metabolism/genetics , Environment , Humans , Lipid Metabolism/genetics , Mice , Mice, Inbred C57BL , N-Methylaspartate/physiology , Neuronal Plasticity/genetics , Receptor, trkB/physiology , Synaptic Transmission
15.
Neurobiol Aging ; 33(4): 830.e1-12, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21855175

ABSTRACT

Chronic stress may be a risk factor for developing Alzheimer's disease (AD), but most studies of the effects of stress in models of AD utilize acute adverse stressors of questionable clinical relevance. The goal of this work was to determine how chronic psychosocial stress affects behavioral and pathological outcomes in an animal model of AD, and to elucidate underlying mechanisms. A triple-transgenic mouse model of AD (3xTgAD mice) and nontransgenic control mice were used to test for an affect of chronic mild social stress on blood glucose, plasma glucocorticoids, plasma insulin, anxiety, and hippocampal amyloid ß-particle (Aß), phosphorylated tau (ptau), and brain-derived neurotrophic factor (BDNF) levels. Despite the fact that both control and 3xTgAD mice experienced rises in corticosterone during episodes of mild social stress, at the end of the 6-week stress period 3xTgAD mice displayed increased anxiety, elevated levels of Aß oligomers and intraneuronal Aß, and decreased brain-derived neurotrophic factor levels, whereas control mice did not. Findings suggest 3xTgAD mice are more vulnerable than control mice to chronic psychosocial stress, and that such chronic stress exacerbates Aß accumulation and impairs neurotrophic signaling.


Subject(s)
Alzheimer Disease/complications , Amyloid beta-Peptides/metabolism , Anxiety/etiology , Behavior, Animal/physiology , Social Behavior , Stress, Psychological/physiopathology , Alzheimer Disease/blood , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Blood Glucose/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Fasting , Glucocorticoids/blood , Hippocampus/pathology , Humans , Insulin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Presenilin-1/genetics , Time Factors , tau Proteins/genetics , tau Proteins/metabolism
16.
PLoS One ; 6(9): e24515, 2011.
Article in English | MEDLINE | ID: mdl-21931736

ABSTRACT

CHD5 is frequently deleted in neuroblastoma and is a tumor suppressor gene. However, little is known about the role of CHD5 other than it is homologous to chromatin remodeling ATPases. We found CHD5 mRNA was restricted to the brain; by contrast, most remodeling ATPases were broadly expressed. CHD5 protein isolated from mouse brain was associated with HDAC2, p66ß, MTA3 and RbAp46 in a megadalton complex. CHD5 protein was detected in several rat brain regions and appeared to be enriched in neurons. CHD5 protein was predominantly nuclear in primary rat neurons and brain sections. Microarray analysis revealed genes that were upregulated and downregulated when CHD5 was depleted from primary neurons. CHD5 depletion altered expression of neuronal genes, transcription factors, and brain-specific subunits of the SWI/SNF remodeling enzyme. Expression of gene sets linked to aging and Alzheimer's disease were strongly altered by CHD5 depletion from primary neurons. Chromatin immunoprecipitation revealed CHD5 bound to these genes, suggesting the regulation was direct. Together, these results indicate that CHD5 protein is found in a NuRD-like multi-protein complex. CHD5 expression is restricted to the brain, unlike the closely related family members CHD3 and CHD4. CHD5 regulates expression of neuronal genes, cell cycle genes and remodeling genes. CHD5 is linked to regulation of genes implicated in aging and Alzheimer's disease.


Subject(s)
Chromatin/chemistry , DNA Helicases/biosynthesis , Gene Expression Regulation, Enzymologic , Mi-2 Nucleosome Remodeling and Deacetylase Complex/biosynthesis , Neurons/metabolism , Trans-Activators/biosynthesis , Aging , Alzheimer Disease/metabolism , Animals , Brain/enzymology , Brain/physiology , Chromatin Immunoprecipitation , Gene Expression Profiling , Humans , Mice , Multiprotein Complexes , Oligonucleotide Array Sequence Analysis , Rats
17.
Neuromolecular Med ; 13(3): 175-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21660587

ABSTRACT

Preclinical evaluation of drugs for neurological disorders is usually performed on overfed rodents, without consideration of how metabolic state might affect drug efficacy. Using a widely employed mouse model of focal ischemic stroke, we found that that the NMDA receptor antagonist dizocilpine (MK-801) reduces brain damage and improves functional outcome in mice on the usual ad libitum diet, but exhibits little or no therapeutic efficacy in mice maintained on an energy-restricted diet. Thus, NMDA receptor activation plays a central role in the mechanism by which a high dietary energy intake exacerbates ischemic brain injury. These findings suggest that inclusion of subjects with a wide range of energy intakes in clinical trials for stroke may mask a drug benefit in the overfed/obese subpopulation of subjects.


Subject(s)
Brain Ischemia/drug therapy , Caloric Restriction , Energy Intake , Excitatory Amino Acid Antagonists/therapeutic use , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Stroke/drug therapy , Animals , Brain Ischemia/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Random Allocation , Stroke/pathology
18.
J Neurosci ; 31(9): 3500-7, 2011 Mar 02.
Article in English | MEDLINE | ID: mdl-21368062

ABSTRACT

Several mouse models of Alzheimer's disease (AD) with abundant ß-amyloid and/or aberrantly phosphorylated tau develop memory impairments. However, multiple non-mnemonic cognitive domains such as attention and executive control are also compromised early in AD individuals. Currently, it is unclear whether mutations in the ß-amyloid precursor protein (APP) and tau are sufficient to cause similar, AD-like attention deficits in mouse models of the disease. To address this question, we tested 3xTgAD mice (which express APPswe, PS1M146V, and tauP301L mutations) and wild-type control mice on a newly developed touchscreen-based 5-choice serial reaction time test of attention and response control. The 3xTgAD mice attended less accurately to short, spatially unpredictable stimuli when the attentional demand of the task was high, and also showed a general tendency to make more perseverative responses than wild-type mice. The attentional impairment of 3xTgAD mice was comparable to that of AD patients in two aspects: first, although 3xTgAD mice initially responded as accurately as wild-type mice, they subsequently failed to sustain their attention over the duration of the task; second, the ability to sustain attention was enhanced by the cholinesterase inhibitor donepezil (Aricept). These findings demonstrate that familial AD mutations not only affect memory, but also cause significant impairments in attention, a cognitive domain supported by the prefrontal cortex and its afferents. Because attention deficits are likely to affect memory encoding and other cognitive abilities, our findings have important consequences for the assessment of disease mechanisms and therapeutics in animal models of AD.


Subject(s)
Alzheimer Disease/drug therapy , Attention/drug effects , Conditioning, Operant/drug effects , Disease Models, Animal , Indans/therapeutic use , Piperidines/therapeutic use , Alzheimer Disease/genetics , Animals , Attention/physiology , Conditioning, Operant/physiology , Donepezil , Humans , Indans/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Transgenic , Piperidines/administration & dosage
19.
J Alzheimers Dis ; 24(2): 349-61, 2011.
Article in English | MEDLINE | ID: mdl-21258150

ABSTRACT

The cell fate determinant Numb exists in four alternatively spliced variants that differ in the length of their PTB (phosphotyrosine-binding domain, either lacking or containing an 11 amino acid insertion) and PRR (proline-rich region, either lacking or containing a 48 amino acid insertion). We previously reported that Numb switches from isoforms containing the PTB insertion to isoforms lacking this insertion in neural cultures subjected to stress induced by trophic factor withdrawal. The switch in Numb isoforms enhances the generation of amyloid-ß peptide (Aß), the principle component of senile plaques in Alzheimer's disease (AD). Here we examine the expression of the Numb isoforms in brains from AD patients and triple transgenic (3xTg) AD mice. We found that levels of the Numb isoforms lacking the PTB insertion are significantly elevated in the parietal cortex but not in the cerebellum of AD patients when compared to control subjects. Levels of Numb isoforms lacking the PTB insertion were also elevated in the cortex but not cerebellum of 12 month-old 3xTg AD mice with Aß deposits compared to younger 3xTg-AD mice and to non-transgenic mice. Exposure of cultured neurons to Aß resulted in an increase in the levels of Numb isoforms lacking the PTB domain, consistent with a role for Aß in the aberrant expression of Numb in vulnerable brain regions of AD patients and mice. Collectively, the data show that altered expression of Numb isoforms in vulnerable neurons occurs during AD pathogenesis and suggest a role for Numb in the disease process.


Subject(s)
Alzheimer Disease/metabolism , Gene Expression Regulation/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Isoforms/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/metabolism , Cerebral Cortex/cytology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Immunoprecipitation/methods , Intercellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Peptide Fragments/pharmacology , Phosphopyruvate Hydratase/metabolism , Presenilin-1/genetics , Protein Isoforms/genetics , Time Factors , Transfection/methods , rab5 GTP-Binding Proteins/metabolism , tau Proteins/genetics
20.
Hum Mol Genet ; 20(4): 659-69, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21106706

ABSTRACT

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by expanded polyglutamine repeats in the huntingtin (Htt) protein. Mutant Htt may damage and kill striatal neurons by a mechanism involving reduced production of brain-derived neurotrophic factor (BDNF) and increased oxidative and metabolic stress. Because electroconvulsive shock (ECS) can stimulate the production of BDNF and protect neurons against stress, we determined whether ECS treatment would modify the disease process and provide a therapeutic benefit in a mouse model of HD. ECS (50 mA for 0.2 s) or sham treatment was administered once weekly to male N171-82Q Htt mutant mice beginning at 2 months of age. Endpoints measured included motor function, striatal and cortical pathology, and levels of protein chaperones and BDNF. ECS treatment delayed the onset of motor symptoms and body weight loss and extended the survival of HD mice. Striatal neurodegeneration was attenuated and levels of protein chaperones (Hsp70 and Hsp40) and BDNF were elevated in striatal neurons of ECS-treated compared with sham-treated HD mice. Our findings demonstrate that ECS can increase the resistance of neurons to mutant Htt resulting in improved functional outcome and extended survival. The potential of ECS as an intervention in subjects that inherit the mutant Htt gene merits further consideration.


Subject(s)
Disease Progression , Electroshock , Huntington Disease/pathology , Huntington Disease/therapy , Mutation/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Gene Expression Regulation , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Huntington Disease/genetics , Male , Mice , Mice, Transgenic , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Proto-Oncogene Proteins c-akt/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Signal Transduction , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...