Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 7(1): 11372, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28900205

ABSTRACT

The apolipoprotein E4 (ApoE4) genotype combines with traumatic brain injury (TBI) to increase the risk of developing Alzheimer's Disease (AD). However, the underlying mechanism(s) is not well-understood. We found that after exposure to repetitive blast-induced TBI, phosphoinositol biphosphate (PIP2) levels in hippocampal regions of young ApoE3 mice were elevated and associated with reduction in expression of a PIP2 degrading enzyme, synaptojanin 1 (synj1). In contrast, hippocampal PIP2 levels in ApoE4 mice did not increase after blast TBI. Following blast TBI, phospho-Tau (pTau) levels were unchanged in ApoE3 mice, whereas in ApoE4 mice, levels of pTau were significantly increased. To determine the causal relationship between changes in pTau and PIP2/synj1 levels after TBI, we tested if down-regulation of synj1 prevented blast-induced Tau hyper-phosphorylation. Knockdown of synj1 decreased pTau levels in vitro, and abolished blast-induced elevation of pTau in vivo. Blast TBI increased glycogen synthase kinase (GSK)-3ß activities in ApoE4 mice, and synj1 knockdown inhibited GSK3ß phosphorylation of Tau. Together, these data suggest that ApoE proteins regulate brain phospholipid homeostasis in response to TBI and that the ApoE4 isoform is dysfunctional in this process. Down-regulation of synj1 rescues blast-induced phospholipid dysregulation and prevents development of Tau hyper-phosphorylation in ApoE4 carriers.


Subject(s)
Apolipoprotein E4/genetics , Brain Injuries, Traumatic/metabolism , Phospholipids/metabolism , tau Proteins/metabolism , Animals , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Apolipoprotein E4/metabolism , Brain/metabolism , Brain/pathology , Brain Injuries, Traumatic/etiology , Brain Injuries, Traumatic/pathology , Cell Line , Hippocampus/metabolism , Mice , Mice, Transgenic , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation
2.
Sci Transl Med ; 8(321): 321ra6, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26764157

ABSTRACT

Blast exposure can cause mild traumatic brain injury (TBI) in mice and other mammals. However, there are important gaps in our understanding of the neuropathology underlying repetitive blast exposure in animal models compared to the neuroimaging abnormalities observed in blast-exposed veterans. Moreover, how an increase in the number of blast exposures affects neuroimaging endpoints in blast-exposed humans is not well understood. We asked whether there is a dose-response relationship between the number of blast-related mild TBIs and uptake of (18)F-fluorodeoxyglucose (FDG), a commonly used indicator of neuronal activity, in the brains of blast-exposed veterans with mild TBI. We found that the number of blast exposures correlated with FDG uptake in the cerebellum of veterans. In mice, blast exposure produced microlesions in the blood-brain barrier (BBB) predominantly in the ventral cerebellum. Purkinje cells associated with these BBB microlesions displayed plasma membrane disruptions and aberrant expression of phosphorylated tau protein. Purkinje cell loss was most pronounced in the ventral cerebellar lobules, suggesting that early-stage breakdown of BBB integrity may be an important factor driving long-term brain changes. Blast exposure caused reactive gliosis in mouse cerebellum, particularly in the deep cerebellar nuclei. Diffusion tensor imaging tractography of the cerebellum of blast-exposed veterans revealed that mean diffusivity correlated negatively with the number of blast-related mild TBIs. Together, these results argue that the cerebellum is vulnerable to repetitive mild TBI in both mice and humans.


Subject(s)
Blast Injuries/complications , Blast Injuries/physiopathology , Cerebellar Diseases/etiology , Veterans , Animals , Axons/pathology , Brain Concussion/etiology , Cerebellar Diseases/pathology , Cerebellum/pathology , Cerebellum/physiopathology , Disks Large Homolog 4 Protein , Fluorodeoxyglucose F18/metabolism , Gliosis/complications , Gliosis/pathology , Glucose/metabolism , Guanylate Kinases/metabolism , Humans , Male , Membrane Proteins/metabolism , Mice, Inbred C57BL , Motor Activity , Neuroglia/pathology , Neurons/pathology , Purkinje Cells/pathology , Synapses/pathology
3.
J Alzheimers Dis ; 45(2): 509-20, 2015.
Article in English | MEDLINE | ID: mdl-25589729

ABSTRACT

The glutamate transporter GLT-1 (also called EAAT2 in humans) plays a critical role in regulating extracellular glutamate levels in the central nervous system (CNS). In Alzheimer's disease (AD), EAAT2 loss is associated with neuropathology and cognitive impairment. In keeping with this, we have reported that partial GLT-1 loss (GLT-1+/-) causes early-occurring cognitive deficits in mice harboring familial AD AßPPswe/PS1ΔE9 mutations. GLT-1 plays important roles in several molecular pathways that regulate brain metabolism, including Akt and insulin signaling in astrocytes. Significantly, AD pathogenesis also involves chronic Akt activation and reduced insulin signaling in the CNS. In this report we tested the hypothesis that GLT-1 heterozygosity (which reduces GLT-1 to levels that are comparable to losses in AD patients) in AßPPswe/PS1ΔE9 mice would induce sustained activation of Akt and disturb components of the CNS insulin signaling cascade. We found that partial GLT-1 loss chronically increased Akt activation (reflected by increased phosphorylation at serine 473), impaired insulin signaling (reflected by decreased IRß phosphorylation of tyrosines 1150/1151 and increased IRS-1 phosphorylation at serines 632/635 - denoted as 636/639 in humans), and reduced insulin degrading enzyme (IDE) activity in brains of mice expressing familial AßPPswe/PS1ΔE9 AD mutations. GLT-1 loss also caused an apparent compensatory increase in IDE activity in the liver, an organ that has been shown to regulate peripheral amyloid-ß levels and expresses GLT-1. Taken together, these findings demonstrate that partial GLT-1 loss can cause insulin/Akt signaling abnormalities that are in keeping with those observed in AD.


Subject(s)
Alzheimer Disease/pathology , Brain/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Insulin/metabolism , Oncogene Protein v-akt/metabolism , Signal Transduction/genetics , Age Factors , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Presenilin-1/genetics
4.
J Histochem Cytochem ; 60(2): 139-51, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22114258

ABSTRACT

The glutamate uptake transporter GLT-1 is best understood for its critical role in preventing brain seizures. Increasing evidence argues that GLT-1 also modulates, and is modulated by, metabolic processes that influence glucose homeostasis. To investigate further the potential role of GLT-1 in these regards, the authors examined GLT-1 expression in pancreas and found that mature multimeric GLT-1 protein is stably expressed in the pancreas of wild-type, but not GLT-1 knockout, mice. There are three primary functional carboxyl-terminus GLT-1 splice variants, called GLT-1a, b, and c. Brain and liver express all three variants; however, the pancreas expresses GLT-1a and GLT-1b but not GLT-1c. Quantitative real time-PCR further revealed that while GLT-1a is the predominant GLT-1 splice variant in brain and liver, GLT-1b is the most abundant splice variant expressed in pancreas. Confocal microscopy and immunohistochemistry showed that GLT-1a and GLT-1b are expressed in both islet ß- and α-cells. GLT-1b was also expressed in exocrine ductal domains. Finally, glutamine synthetase was coexpressed with GLT-1 in islets, which suggests that, as with liver and brain, one possible role of GLT-1 in the pancreas is to support glutamine synthesis.


Subject(s)
Excitatory Amino Acid Transporter 2/genetics , Pancreas/metabolism , Animals , Excitatory Amino Acid Transporter 2/deficiency , Excitatory Amino Acid Transporter 2/metabolism , Gene Expression Profiling , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Pancreas/cytology , Real-Time Polymerase Chain Reaction
5.
J Alzheimers Dis ; 26(3): 447-55, 2011.
Article in English | MEDLINE | ID: mdl-21677376

ABSTRACT

Glutamate transporters regulate normal synaptic network interactions and prevent neurotoxicity by rapidly clearing extracellular glutamate. GLT-1, the dominant glutamate transporter in the cerebral cortex and hippocampus, is significantly reduced in Alzheimer's disease (AD). However, the role GLT-1 loss plays in the cognitive dysfunction and pathology of AD is unknown. To determine the significance of GLT-1 dysfunction on AD-related pathological processes, mice lacking one allele for GLT-1(+/-) were crossed with transgenic mice expressing mutations of the amyloid-ß protein precursor and presenilin-1 (AßPPswe/PS1ΔE9) and investigated at 6 or 9 months of age. Partial loss of GLT-1 unmasked spatial memory deficits in 6-month-old mice expressing AßPPswe/PS1ΔE9, with these mice also exhibiting an increase in the ratio of detergent-insoluble Aß42/Aß40. At 9 months both behavioral performance and insoluble Aß42/Aß40 ratios among GLT-1(+/+)/AßPPswe/PS1ΔE9 and GLT-1(+/-)/AßPPswe/PS1ΔE9 mice were comparable. These results suggest that deficits in glutamate transporter function compound the effects of familial AD AßPP/PS1 mutant transgenes in younger animals and thus may contribute to early occurring pathogenic processes associated with AD.


Subject(s)
Alzheimer Disease/metabolism , Cognition Disorders/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Aging/psychology , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Behavior, Animal/physiology , Blotting, Western , Brain Chemistry , Cognition Disorders/genetics , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Excitatory Amino Acid Transporter 2/genetics , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Presenilin-1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...