Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
J Neurosci ; 44(8)2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38228367

ABSTRACT

Subconcussive head impacts are associated with the development of acute and chronic cognitive deficits. We recently reported that high-frequency head impact (HFHI) causes chronic cognitive deficits in mice through synaptic changes. To better understand the mechanisms underlying HFHI-induced memory decline, we used TRAP2/Ai32 transgenic mice to enable visualization and manipulation of memory engrams. We labeled the fear memory engram in male and female mice exposed to an aversive experience and subjected them to sham or HFHI. Upon subsequent exposure to natural memory recall cues, sham, but not HFHI, mice successfully retrieved fearful memories. In sham mice the hippocampal engram neurons exhibited synaptic plasticity, evident in amplified AMPA:NMDA ratio, enhanced AMPA-weighted tau, and increased dendritic spine volume compared with nonengram neurons. In contrast, although HFHI mice retained a comparable number of hippocampal engram neurons, these neurons did not undergo synaptic plasticity. This lack of plasticity coincided with impaired activation of the engram network, leading to retrograde amnesia in HFHI mice. We validated that the memory deficits induced by HFHI stem from synaptic plasticity impairments by artificially activating the engram using optogenetics and found that stimulated memory recall was identical in both sham and HFHI mice. Our work shows that chronic cognitive impairment after HFHI is a result of deficiencies in synaptic plasticity instead of a loss in neuronal infrastructure, and we can reinstate a forgotten memory in the amnestic brain by stimulating the memory engram. Targeting synaptic plasticity may have therapeutic potential for treating memory impairments caused by repeated head impacts.


Subject(s)
Amnesia , Memory , Male , Mice , Female , Animals , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid , Memory/physiology , Neuronal Plasticity/physiology , Hippocampus/physiology , Mice, Transgenic
2.
J Neuroinflammation ; 21(1): 135, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802931

ABSTRACT

Traumatic brain injury (TBI) is a major cause of disability and mortality worldwide, particularly among the elderly, yet our mechanistic understanding of what renders the post-traumatic brain vulnerable to poor outcomes, and susceptible to neurological disease, is incomplete. It is well established that dysregulated and sustained immune responses elicit negative consequences after TBI; however, our understanding of the neuroimmune interface that facilitates crosstalk between central and peripheral immune reservoirs is in its infancy. The meninges serve as the interface between the brain and the immune system, facilitating important bi-directional roles in both healthy and disease settings. It has been previously shown that disruption of this system exacerbates neuroinflammation in age-related neurodegenerative disorders such as Alzheimer's disease; however, we have an incomplete understanding of how the meningeal compartment influences immune responses after TBI. In this manuscript, we will offer a detailed overview of the holistic nature of neuroinflammatory responses in TBI, including hallmark features observed across clinical and animal models. We will highlight the structure and function of the meningeal lymphatic system, including its role in immuno-surveillance and immune responses within the meninges and the brain. We will provide a comprehensive update on our current knowledge of meningeal-derived responses across the spectrum of TBI, and identify new avenues for neuroimmune modulation within the neurotrauma field.


Subject(s)
Brain Injuries, Traumatic , Meninges , Neuroinflammatory Diseases , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/pathology , Humans , Animals , Meninges/immunology , Meninges/pathology , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/pathology , Neuroimmunomodulation/physiology , Neuroimmunomodulation/immunology
3.
Glia ; 65(9): 1423-1438, 2017 09.
Article in English | MEDLINE | ID: mdl-28608978

ABSTRACT

The activation of resident microglial cells, alongside the infiltration of peripheral macrophages, are key neuroinflammatory responses to traumatic brain injury (TBI) that are directly associated with neuronal death. Sexual disparities in response to TBI have been previously reported; however it is unclear whether a sex difference exists in neuroinflammatory progression after TBI. We exposed male and female mice to moderate-to-severe controlled cortical impact injury and studied glial cell activation in the acute and chronic stages of TBI using immunofluorescence and in situ hybridization analysis. We found that the sex response was completely divergent up to 7 days postinjury. TBI caused a rapid and pronounced cortical microglia/macrophage activation in male mice with a prominent activated phenotype that produced both pro- (IL-1ß and TNFα) and anti-inflammatory (Arg1 and TGFß) cytokines with a single-phase, sustained peak from 1 to 7 days. In contrast, TBI caused a less robust microglia/macrophage phenotype in females with biphasic pro-inflammatory response peaks at 4 h and 7 days, and a delayed anti-inflammatory mRNA peak at 30 days. We further report that female mice were protected against acute cell loss after TBI, with male mice demonstrating enhanced astrogliosis, neuronal death, and increased lesion volume through 7 days post-TBI. Collectively, these findings indicate that TBI leads to a more aggressive neuroinflammatory profile in male compared with female mice during the acute and subacute phases postinjury. Understanding how sex affects the course of neuroinflammation following brain injury is a vital step toward developing personalized and effective treatments for TBI.


Subject(s)
Brain Injuries, Traumatic/immunology , Inflammation/physiopathology , Sex Characteristics , Animals , Arginase/metabolism , Astrocytes/immunology , Astrocytes/pathology , Brain Injuries, Traumatic/pathology , Cell Death , Disease Models, Animal , Female , Gliosis/immunology , Gliosis/pathology , Inflammation/pathology , Macrophage Activation/immunology , Macrophage Activation/physiology , Male , Mice, Inbred C57BL , Microglia/immunology , Microglia/pathology , Neuroimmunomodulation/physiology , Transforming Growth Factor beta/metabolism
4.
J Neuroinflammation ; 14(1): 65, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28340575

ABSTRACT

BACKGROUND: NADPH oxidase (NOX2) is an enzyme system that generates reactive oxygen species (ROS) in microglia and macrophages. Excessive ROS production is linked with neuroinflammation and chronic neurodegeneration following traumatic brain injury (TBI). Redox signaling regulates macrophage/microglial phenotypic responses (pro-inflammatory versus anti-inflammatory), and NOX2 inhibition following moderate-to-severe TBI markedly reduces pro-inflammatory activation of macrophages/microglia resulting in concomitant increases in anti-inflammatory responses. Here, we report the signaling pathways that regulate NOX2-dependent macrophage/microglial phenotype switching in the TBI brain. METHODS: Bone marrow-derived macrophages (BMDMs) prepared from wildtype (C57Bl/6) and NOX2 deficient (NOX2-/-) mice were treated with lipopolysaccharide (LPS; 10 ng/ml), interleukin-4 (IL-4; 10 ng/ml), or combined LPS/IL-4 to investigate signal transduction pathways associated with macrophage activation using western immunoblotting and qPCR analyses. Signaling pathways and activation markers were evaluated in ipsilateral cortical tissue obtained from adult male wildtype and NOX2-/- mice that received moderate-level controlled cortical impact (CCI). A neutralizing anti-IL-10 approach was used to determine the effects of IL-10 on NOX2-dependent transitions from pro- to anti-inflammatory activation states. RESULTS: Using an LPS/IL-4-stimulated BMDM model that mimics the mixed pro- and anti-inflammatory responses observed in the injured cortex, we show that NOX2-/- significantly reduces STAT1 signaling and markers of pro-inflammatory activation. In addition, NOX2-/- BMDMs significantly increase anti-inflammatory marker expression; IL-10-mediated STAT3 signaling, but not STAT6 signaling, appears to be critical in regulating this anti-inflammatory response. Following moderate-level CCI, IL-10 is significantly increased in microglia/macrophages in the injured cortex of NOX2-/- mice. These changes are associated with increased STAT3 activation, but not STAT6 activation, and a robust anti-inflammatory response. Neutralization of IL-10 in NOX2-/- BMDMs or CCI mice blocks STAT3 activation and the anti-inflammatory response, thereby demonstrating a critical role for IL-10 in regulating NOX2-dependent transitions between pro- and anti-inflammatory activation states. CONCLUSIONS: These studies indicate that following TBI NOX2 inhibition promotes a robust anti-inflammatory response in macrophages/microglia that is mediated by the IL-10/STAT3 signaling pathway. Thus, therapeutic interventions that inhibit macrophage/microglial NOX2 activity may improve TBI outcomes by not only limiting pro-inflammatory neurotoxic responses, but also enhancing IL-10-mediated anti-inflammatory responses that are neuroprotective.


Subject(s)
Brain Injuries, Traumatic/immunology , Inflammation/pathology , Macrophage Activation/immunology , Macrophages/immunology , NADPH Oxidase 2/deficiency , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Inflammation/immunology , Inflammation/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism
5.
Am J Pathol ; 186(3): 552-67, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26857506

ABSTRACT

Mild traumatic brain injury (mTBI) is an emerging risk for chronic behavioral, cognitive, and neurodegenerative conditions. Athletes absorb several hundred mTBIs each year; however, rodent models of repeat mTBI (rmTBI) are often limited to impacts in the single digits. Herein, we describe the effects of 30 rmTBIs, examining structural and pathological changes in mice up to 365 days after injury. We found that single mTBI causes a brief loss of consciousness and a transient reduction in dendritic spines, reflecting a loss of excitatory synapses. Single mTBI does not cause axonal injury, neuroinflammation, or cell death in the gray or white matter. Thirty rmTBIs with a 1-day interval between each mTBI do not cause dendritic spine loss; however, when the interinjury interval is increased to 7 days, dendritic spine loss is reinstated. Thirty rmTBIs cause white matter pathology characterized by positive silver and Fluoro-Jade B staining, and microglial proliferation and activation. This pathology continues to develop through 60 days, and is still apparent at 365 days, after injury. However, rmTBIs did not increase ß-amyloid levels or tau phosphorylation in the 3xTg-AD mouse model of Alzheimer disease. Our data reveal that single mTBI causes a transient loss of synapses, but that rmTBIs habituate to repetitive injury within a short time period. rmTBI causes the development of progressive white matter pathology that continues for months after the final impact.


Subject(s)
Brain Injuries/pathology , Dendritic Spines/pathology , White Matter/pathology , Amyloid/metabolism , Animals , Behavior, Animal , Brain Concussion/pathology , Brain Injuries/metabolism , Craniocerebral Trauma/metabolism , Craniocerebral Trauma/pathology , Dendritic Spines/metabolism , Disease Models, Animal , Fluoresceins , Golgi Apparatus , Humans , Inflammation , Male , Maze Learning , Mice , Mice, Inbred C57BL , Recurrence , Unconsciousness , White Matter/metabolism , tau Proteins/metabolism
6.
J Neurochem ; 139(4): 610-623, 2016 11.
Article in English | MEDLINE | ID: mdl-27507246

ABSTRACT

The trans-activating response of DNA/RNA-binding protein (TDP)-43 pathology is associated with many neurodegenerative diseases via unknown mechanisms. Here, we use a transgenic mouse model over-expressing human wild-type neuronal TDP-43 to study the effects of TDP-43 pathology on glutamate metabolism and synaptic function. We found that neuronal TDP-43 over-expression affects synaptic protein expression, including Synapsin I, and alters surrounding astrocytic function. TDP-43 over-expression is associated with an increase in glutamate and γ-amino butyric acid and reduction of glutamine and aspartate levels, indicating impairment of presynaptic terminal. TDP-43 also decreases tricarboxylic acid cycle metabolism and induces oxidative stress via lactate accumulation. Neuronal TDP-43 does not alter microglia activity or significantly changes systemic and brain inflammatory markers compared to control. We previously demonstrated that brain-penetrant tyrosine kinase inhibitors (TKIs), nilotinib and bosutinib, reduce TDP-43-induced cell death in transgenic mice. Here, we show that TKIs reverse the effects of TDP-43 on synaptic proteins, increase astrocytic function and restore glutamate and neurotransmitter balance in TDP-43 mice. Nilotinib, but not bosutinib, reverses mitochondrial impairment and oxidative metabolism. Taken together, these data suggest that TKIs can attenuate TDP-43 toxicity and improve synaptic and astrocytic function, independent of microglial or other inflammatory effects. In conclusion, our data demonstrate novel mechanisms of the effects of neuronal TDP-43 over-expression on synaptic protein expression and alteration of astrocytic function.


Subject(s)
Astrocytes/physiology , DNA-Binding Proteins/biosynthesis , Homeostasis/physiology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/metabolism , Synapsins/biosynthesis , Animals , Astrocytes/drug effects , Cell Line, Tumor , Female , Gene Expression , Homeostasis/drug effects , Humans , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Synapses/drug effects , Synapses/metabolism , Synapsins/genetics
7.
Learn Mem ; 20(5): 256-66, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23592036

ABSTRACT

The apolipoprotein E4 (APOE-ε4) allele is the strongest genetic risk factor for developing late-onset Alzheimer's disease, and may predispose individuals to Alzheimer's-related cognitive decline by affecting normal brain function early in life. To investigate the impact of human APOE alleles on cognitive performance in mice, we trained 3-mo-old APOE targeted replacement mice (E2, E3, and E4) in the Barnes maze to locate and enter a target hole along the perimeter of the maze. Long-term spatial memory was probed 24 h and 72 h after training. We found that young E4 mice exhibited significantly impaired spatial learning and memory in the Barnes maze compared to E3 mice. Deficits in spatial cognition were also present in a second independent cohort of E4 mice tested at 18 mo of age. In contrast, cognitive performance in the hidden platform water maze was not as strongly affected by APOE genotype. We also examined the dendritic morphology of neurons in the medial entorhinal cortex of 3-mo-old TR mice, neurons important to spatial learning functions. We found significantly shorter dendrites and lower spine densities in basal shaft dendrites of E4 mice compared to E3 mice, consistent with spatial learning and memory deficits in E4 animals. These findings suggest that human APOE-ε4 may affect cognitive function and neuronal morphology early in life.


Subject(s)
Apolipoprotein E4/genetics , Dendritic Spines/metabolism , Entorhinal Cortex/metabolism , Maze Learning/physiology , Memory/physiology , Neurons/metabolism , Animals , Apolipoprotein E4/metabolism , Behavior, Animal , Dendrites/genetics , Dendrites/metabolism , Dendritic Spines/genetics , Mice , Mice, Transgenic , Spatial Behavior/physiology
8.
Front Neurosci ; 17: 1210175, 2023.
Article in English | MEDLINE | ID: mdl-37588516

ABSTRACT

Traumatic Brain Injury (TBI) is a major cause of disability and mortality, particularly among the elderly, yet our mechanistic understanding of how age renders the post-traumatic brain vulnerable to poor clinical outcomes and susceptible to neurological disease remains poorly understood. It is well established that dysregulated and sustained immune responses contribute to negative outcomes after TBI, however our understanding of the interactions between central and peripheral immune reservoirs is still unclear. The meninges serve as the interface between the brain and the immune system, facilitating important bi-directional roles in healthy and disease settings. It has been previously shown that disruption of this system exacerbates inflammation in age related neurodegenerative disorders such as Alzheimer's disease, however we have an incomplete understanding of how the meningeal compartment influences immune responses after TBI. Here, we examine the meningeal tissue and its response to brain injury in young (3-months) and aged (18-months) mice. Utilizing a bioinformatic approach, high-throughput RNA sequencing demonstrates alterations in the meningeal transcriptome at sub-acute (7-days) and chronic (1 month) timepoints after injury. We find that age alone chronically exacerbates immunoglobulin production and B cell responses. After TBI, adaptive immune response genes are up-regulated in a temporal manner, with genes involved in T cell responses elevated sub-acutely, followed by increases in B cell related genes at chronic time points after injury. Pro-inflammatory cytokines are also implicated as contributing to the immune response in the meninges, with ingenuity pathway analysis identifying interferons as master regulators in aged mice compared to young mice following TBI. Collectively these data demonstrate the temporal series of meningeal specific signatures, providing insights into how age leads to worse neuroinflammatory outcomes in TBI.

9.
Neuroinformatics ; 21(3): 501-516, 2023 07.
Article in English | MEDLINE | ID: mdl-37294503

ABSTRACT

Traumatic brain injury (TBI) and repetitive head impacts can result in a wide range of neurological symptoms. Despite being the most common neurological disorder in the world, repeat head impacts and TBI do not have any FDA-approved treatments. Single neuron modeling allows researchers to extrapolate cellular changes in individual neurons based on experimental data. We recently characterized a model of high frequency head impact (HFHI) with a phenotype of cognitive deficits associated with decreases in neuronal excitability of CA1 neurons and synaptic changes. While the synaptic changes have been interrogated in vivo, the cause and potential therapeutic targets of hypoexcitability following repetitive head impacts are unknown. Here, we generated in silico models of CA1 pyramidal neurons from current clamp data of control mice and mice that sustained HFHI. We use a directed evolution algorithm with a crowding penalty to generate a large and unbiased population of plausible models for each group that approximated the experimental features. The HFHI neuron model population showed decreased voltage gated sodium conductance and a general increase in potassium channel conductance. We used partial least squares regression analysis to identify combinations of channels that may account for CA1 hypoexcitability after HFHI. The hypoexcitability phenotype in models was linked to A- and M-type potassium channels in combination, but not by any single channel correlations. We provide an open access set of CA1 pyramidal neuron models for both control and HFHI conditions that can be used to predict the effects of pharmacological interventions in TBI models.


Subject(s)
Neurons , Potassium Channels , Mice , Animals , Potassium Channels/pharmacology , Action Potentials/physiology , Neurons/physiology , Pyramidal Cells/physiology
10.
PLoS One ; 18(7): e0288363, 2023.
Article in English | MEDLINE | ID: mdl-37440485

ABSTRACT

The pathophysiological changes that occur after traumatic brain injury (TBI) can lead to the development of post-traumatic epilepsy, a life-long complication of brain trauma. The etiology of post-traumatic epilepsy remains unknown, but TBI brains exhibit an abnormal excitatory / inhibitory balance. In this study, we examine how brain injury alters susceptibility to chemically-induced seizures in C57Bl/6J mice, and if pharmacological enhancement of glutamate transporters can reduce chronic post-traumatic seizures. We found that controlled cortical impact (CCI) mice display delayed susceptibility to pentylenetetrazol (PTZ)-induced seizures. While CCI mice have no change in seizure susceptibility at 7d post-injury (dpi), at 70dpi they have reduced latency to PTZ-induced seizure onset, higher seizure frequency and longer seizure duration. Quantification of glutamate transporter mRNA showed that levels of Scl1a2 and Scl1a3 mRNA were increased at 7dpi, but significantly decreased at 70dpi. To test if increased levels of glutamate transporters can ameliorate delayed-onset seizure susceptibility in TBI mice, we exposed a new cohort of mice to CCI and administered ceftriaxone (200mg/kg/day) for 14d from 55-70dpi. We found that ceftriaxone significantly increased Scl1a2 and Scl1a3 in CCI mouse brain at 70dpi, and prevented the susceptibility of CCI mice to PTZ-induced seizures. This study demonstrates cortical impact can induce a delayed-onset seizure phenotype in mice. Delayed (55dpi) ceftriaxone treatment enhances glutamate transporter mRNA in the CCI brain, and reduces PTZ-induced seizures in CCI mice.


Subject(s)
Brain Injuries, Traumatic , Epilepsy, Post-Traumatic , Humans , Mice , Animals , Ceftriaxone/pharmacology , Ceftriaxone/therapeutic use , Time-to-Treatment , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Seizures/chemically induced , Seizures/drug therapy , Seizures/complications , Pentylenetetrazole/toxicity , Mice, Inbred C57BL , Glutamates , Disease Models, Animal
11.
Methods Protoc ; 5(5)2022 Sep 27.
Article in English | MEDLINE | ID: mdl-36287049

ABSTRACT

In aging, the brain is more vulnerable to injury and neurodegenerative disease, but the mechanisms responsible are largely unknown. Evidence now suggests that neuroinflammation, mediated by resident brain astrocyte and microglia populations, are key players in the generation of inflammatory responses and may influence both age related processes and the initiation/progression of neurodegeneration. Consequently, targeting these cell types individually and collectively may aid in the development of novel disease-modifying therapies. We have optimized and characterized a protocol for the effective sequential isolation of both microglia and astrocytes from the adult mouse brain in young and aged mice. We demonstrate a technique for the sequential isolation of these immune cells by using magnetic beads technology, optimized to increase yield and limit potential artifacts in downstream transcriptomic applications, including RNA-sequencing pipelines. This technique is versatile, cost-effective, and reliable for the study of responses within the same biological context, simultaneously being advantageous in reducing mice numbers required to assess cellular responses in normal and age-related pathological conditions.

12.
Biology (Basel) ; 11(7)2022 Jul 08.
Article in English | MEDLINE | ID: mdl-36101412

ABSTRACT

Traumatic brain injury (TBI) is a significant risk factor for the development of sleep and circadian rhythm impairments. In this study we compare the circadian rhythms and sleep patterns in the high-frequency head impact (HFHI) and controlled cortical impact (CCI) mouse models of TBI. These mouse models have different injury mechanisms key differences of pathology in brain regions controlling circadian rhythms and EEG wave generation. We found that both HFHI and CCI caused dysregulation in the diurnal expression of core circadian genes (Bmal1, Clock, Per1,2, Cry1,2) at 24 h post-TBI. CCI mice had reduced locomotor activity on running wheels in the first 7 d post-TBI; however, both CCI and HFHI mice were able to maintain circadian behavior cycles even in the absence of light cues. We used implantable EEG to measure sleep cycles and brain activity and found that there were no differences in the time spent awake, in NREM or REM sleep in either TBI model. However, in the sleep states, CCI mice have reduced delta power in NREM sleep and reduced theta power in REM sleep at 7 d post-TBI. Our data reveal that different types of brain trauma can result in distinct patterns of circadian and sleep disruptions and can be used to better understand the etiology of sleep disorders after TBI.

13.
Biochim Biophys Acta ; 1801(8): 853-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20304094

ABSTRACT

Many preclinical and clinical studies have implied a role for cholesterol in the pathogenesis of Alzheimer's disease (AD). In this review we will discuss the movement of intracellular cholesterol and how normal distribution, transport, and export of cholesterol are vital for regulation of the AD related protein, Abeta. We focus on cholesterol distribution in the plasma membrane, transport through the endosomal/lysosomal system, control of cholesterol intracellular signaling at the endoplasmic reticulum and Golgi, the HMG-CoA reductase pathway and finally export of cholesterol from the cell.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cholesterol/metabolism , Cholesterol/physiology , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Animals , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/physiology , Endosomes/metabolism , Endosomes/physiology , Humans , Intracellular Space/metabolism , Lysosomes/metabolism , Lysosomes/physiology , Models, Biological , Protein Processing, Post-Translational/physiology
14.
Hum Mol Genet ; 18(17): 3206-16, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19483198

ABSTRACT

Alzheimer's disease and Parkinson's disease are common neurodegenerative diseases that may share some underlying mechanisms of pathogenesis. Abeta(1-42) fragments are found intracellularly, and extracellularly as amyloid plaques, in Alzheimer's disease and in dementia with Lewy Bodies. Parkin is an E3-ubiquitin ligase involved in proteasomal degradation of intracellular proteins. Mutations in parkin, which result in loss of parkin function, lead to early onset Parkinsonism. Here we tested whether the ubiquitin ligase activity of parkin could lead to reduction in intracellular human Abeta(1-42). Lentiviral constructs encoding either human parkin or human Abeta(1-42) were used to infect M17 neuroblastoma cells. Parkin expression resulted in reduction of intracellular human Abeta(1-42) levels and protected against its toxicity in M17 cells. Co-injection of lentiviral constructs into control rat primary motor cortex demonstrated that parkin co-expression reduced human Abeta(1-42) levels and Abeta(1-42)-induced neuronal degeneration in vivo. Parkin increased proteasomal activity, and proteasomal inhibition blocked the effects of parkin on reducing Abeta(1-42) levels. Incubation of Abeta(1-42) cell lysates with ubiquitin, in the presence of parkin, demonstrated the generation of Abeta-ubiquitin complexes. These data indicate that parkin promotes ubiquitination and proteasomal degradation of intracellular Abeta(1-42) and demonstrate a protective effect in neurodegenerative diseases with Abeta deposits.


Subject(s)
Amyloid beta-Peptides/metabolism , Neurodegenerative Diseases/metabolism , Peptide Fragments/metabolism , Ubiquitin-Protein Ligases/metabolism , Amyloid beta-Peptides/genetics , Cell Line , Humans , Mutation , Neurodegenerative Diseases/genetics , Peptide Fragments/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
15.
Dev Neurosci ; 33(5): 395-403, 2011.
Article in English | MEDLINE | ID: mdl-22067669

ABSTRACT

Hyperactivity, hypersensitivity to auditory stimuli, and exaggerated fear are common behavioral abnormalities observed in individuals with fragile X syndrome (FXS), a neurodevelopmental disorder that is the most common genetic cause of autism. Evidence from studies of the Fmr1 knockout (KO) mouse model of FXS supports the notion that impaired GABAergic transmission in different brain regions such as the amygdala, striatum or cerebral cortex is central to FXS behavioral abnormalities. This suggests that the GABAergic system might be an intriguing target to ameliorate some of the phenotypes in FXS. Our recent work revealed that THIP (gaboxadol), a GABA(A) receptor agonist, can restore principal neuron excitability deficits in the Fmr1 KO amygdala, suggesting that THIP may also restore some of the key behavioral abnormalities in Fmr1 KO mice. Here, we reveal that THIP significantly attenuated hyperactivity in Fmr1 KO mice, and reduced prepulse inhibition in a volume-dependent manner. In contrast, THIP did not reverse the deficits in cued fear or startle response. Thus, this study shows that enhancing GABAergic transmission can correct specific behavioral phenotypes of the Fmr1 KO mouse further supporting that targeting the GABAergic system, and specifically tonic inhibition, might be important for correcting or ameliorating some key behaviors in FXS.


Subject(s)
Behavior, Animal/drug effects , Fragile X Syndrome/drug therapy , Fragile X Syndrome/physiopathology , GABA-A Receptor Agonists/therapeutic use , Isoxazoles/therapeutic use , Receptors, GABA-A/metabolism , Animals , Conditioning, Psychological/drug effects , Disease Models, Animal , Fear/drug effects , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Humans , Male , Mice , Mice, Knockout , Motor Activity/drug effects , Neuropsychological Tests , gamma-Aminobutyric Acid/metabolism
16.
FASEB J ; 24(1): 58-69, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19720620

ABSTRACT

The goal of this study was to determine the effect of X11alpha on ApoE receptor 2 (ApoEr2) trafficking and the functional significance of this interaction on cell movement in MCF 10A epithelial cells. We found that X11alpha increased surface levels of ApoEr2 by 64% compared to vector control, as determined by surface protein biotinylation. To examine the functional significance of this effect, we tested whether ApoEr2 played a novel role in cell movement in a wound-healing assay. We found that overexpression of ApoEr2 in MCF 10A cells increased cell migration velocity by 87% (P<0.01, n=4) compared to GFP control. Cotransfection of X11alpha had an additive effect on average velocity compared to ApoEr2 alone (13%; P<0.05, n=4). In addition, we tested whether ApoEr2 ligands altered the effect of ApoEr2 on cell movement. We found that treatment with concentrated medium containing the extracellular matrix protein Reelin, but not control medium, further increased the velocity of ApoEr2- but not APP-transfected cells (20%; P<0.001, n=4). Similarly, Reelin treatment increased cell velocity in the presence of ApoEr2 and X11alpha (10%; P<0.05, n=4). In the present study, we are the first to demonstrate that ApoEr2 regulates cell movement, and both X11alpha and Reelin enhance this effect.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cell Movement/physiology , Extracellular Matrix Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Lipoprotein/metabolism , Serine Endopeptidases/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Binding Sites/genetics , Biological Transport, Active , COS Cells , Cadherins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Line , Cell Membrane/metabolism , Chlorocebus aethiops , Exons , Extracellular Matrix Proteins/genetics , Humans , LDL-Receptor Related Proteins , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neurons/metabolism , PDZ Domains , Phosphorylation , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , RNA, Small Interfering/genetics , Rats , Receptors, Lipoprotein/chemistry , Receptors, Lipoprotein/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reelin Protein , Serine Endopeptidases/genetics , Transfection , Two-Hybrid System Techniques , Wound Healing/physiology
17.
Front Cell Neurosci ; 15: 763423, 2021.
Article in English | MEDLINE | ID: mdl-35115908

ABSTRACT

We have recently shown that the cognitive impairments in a mouse model of high-frequency head impact (HFHI) are caused by chronic changes to synaptic physiology. To better understand these synaptic changes occurring after repeat head impact, we used Thy1-GcCAMP6f mice to study intracellular and intercellular calcium dynamics and neuronal ensembles in HFHI mice. We performed simultaneous calcium imaging and local field potential (LFP) recordings of the CA1 field during an early-LTP paradigm in acute hippocampal slice preparations 24 h post-impact. As previously reported, HFHI causes a decrease in early-LTP in the absence of any shift in the input-output curve. Calcium analytics revealed that HFHI hippocampal slices have similar numbers of active ROIs, however, the number of calcium transients per ROI was significantly increased in HFHI slices. Ensembles consist of coordinated activity between groups of active ROIs. We exposed the CA1 ensemble to Schaffer-collateral stimulation in an abbreviated LTP paradigm and observed novel coordinated patterns of post stimulus calcium ensemble activity. HFHI ensembles displayed qualitatively similar patterns of post-stimulus ensemble activity to shams but showed significant changes in quantitative ensemble inactivation and reactivation. Previous in vivo and in vitro reports have shown that ensemble activity frequently occurs through a similar set of ROIs firing in a repeating fashion. HFHI slices showed a decrease in such coordinated firing patterns during post stimulus ensemble activity. The present study shows that HFHI alters synaptic activity and disrupts neuronal organization of the ensemble, providing further evidence of physiological synaptic adaptation occurring in the brain after a high frequency of non-pathological head impacts.

18.
Nat Commun ; 12(1): 2613, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33972519

ABSTRACT

Repeated head impact exposure can cause memory and behavioral impairments. Here, we report that exposure to non-damaging, but high frequency, head impacts can alter brain function in mice through synaptic adaptation. High frequency head impact mice develop chronic cognitive impairments in the absence of traditional brain trauma pathology, and transcriptomic profiling of mouse and human chronic traumatic encephalopathy brain reveal that synapses are strongly affected by head impact. Electrophysiological analysis shows that high frequency head impacts cause chronic modification of the AMPA/NMDA ratio in neurons that underlie the changes to cognition. To demonstrate that synaptic adaptation is caused by head impact-induced glutamate release, we pretreated mice with memantine prior to head impact. Memantine prevents the development of the key transcriptomic and electrophysiological signatures of high frequency head impact, and averts cognitive dysfunction. These data reveal synapses as a target of high frequency head impact in human and mouse brain, and that this physiological adaptation in response to head impact is sufficient to induce chronic cognitive impairment in mice.


Subject(s)
Brain Injuries, Traumatic/metabolism , Cognition , Neurons/pathology , Synapses/metabolism , Synapses/pathology , Transcriptome/genetics , Amyloid beta-Peptides/metabolism , Animals , Behavior Rating Scale , Brain Injuries, Traumatic/genetics , Cognition/drug effects , Cognitive Dysfunction/pathology , Electrophysiology , Gene Ontology , Glutamic Acid/metabolism , Memantine/administration & dosage , Mice , Microglia/metabolism , Multigene Family , Neuronal Plasticity/genetics , Neurons/cytology , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/genetics , tau Proteins/metabolism
19.
J Neurotrauma ; 37(3): 517-527, 2020 02 01.
Article in English | MEDLINE | ID: mdl-30343623

ABSTRACT

As rodent locomotion becomes a more popular behavioral assay, proper rodent gait analysis becomes more and more important. Gait measures, such as stride length, cycle time, and duty factor, are not independent of one another, making statistical comparisons between groups a tricky endeavor. Instead of identifying the mathematical relationships between a group of locomotor measures, we simply tracked the steps of rodents in x,y,t space. By plotting with respect to the reference limb, we are able to quantify locomotor changes in space, time, and coordination simultaneously. With our technique, we show that the overall locomotion of 77 rats 1 week after a C4/5 right overhemisection injury was significantly different than pre-injury. This difference was maintained in untreated animals for the entire 7 weeks of the study, but how this difference arose changed. Initially, the right forelimb exhibited very abnormal stepping, but eventually reduced its difference from pre-injury levels. Conversely, the left forelimb was initially mildly different from pre-injury, but further deviated from normal stepping as the weeks went on. Our new gait analysis technique helps to show the trade-off between the restoration of function and the spontaneous development of compensatory techniques. When we applied this new analysis technique to 13 mice after a severe controlled cortical impact, we found that their locomotion was no different from 12 sham mice for the entire 4 weeks of the study. We believe that this gait analysis method succinctly addresses the confound of interdependency of gait measures and does so across multiple injury models.


Subject(s)
Adaptation, Physiological/physiology , Brain Injuries, Traumatic/physiopathology , Gait Analysis/methods , Gait/physiology , Recovery of Function/physiology , Spinal Cord Injuries/physiopathology , Animals , Brain Injuries, Traumatic/diagnosis , Cervical Vertebrae/injuries , Female , Gait Analysis/trends , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Rodentia , Spinal Cord Injuries/diagnosis
20.
Front Endocrinol (Lausanne) ; 11: 556380, 2020.
Article in English | MEDLINE | ID: mdl-33071972

ABSTRACT

Glucose is an essential cellular fuel for maintaining normal brain functions. Traumatic brain injury (TBI) decreases brain glucose utilization in both human and experimental animals during the acute or subacute phase of TBI. It remains unclear as to how the damages affect brain glucose utilization and its association with persistent neurobehavioral impairments in the chronic phase of mild TBI (mTBI). Accordingly, we compared expression of selected genes important to brain glucose utilization in different brain regions of mice during the chronic phase in mTBI vs. sham operated mice. These genes included hexokinase-1 (HK1), phosphofructokinase (PFK), pyruvate kinase (PK), pyruvate dehydrogenase (PDH), capillary glucose transporter (Glut-1), neuron glucose transporter (Glut-3), astrocyte lactate transpor1 (MCT-1), neuron lactate transporter (MCT-2), lactate receptor (GPR81), and Hexokinase isoform-2 (HK2). Young adult male C57BL/6J mice were brain injured with repetitive closed-head concussions. Morris water maze (MWM), elevated plus maze (EPM), and neurological severity score test (NSS) were performed for evaluation of mice neurobehavioral impairments at 2, 4, and 6 months post mTBI. Two days after completion of the last behavioral test, the frontal cortex, hippocampus, brainstem, hypothalamus, and cerebellum were collected for gene expression measurements. The expression of the mRNAs encoding PK, and PDH, two critical enzymes in glucose metabolism, was decreased at all-time points only in the hippocampus, but was unchanged in the brainstem, hypothalamus, and cortex in mTBI mice. mTBI mice also exhibited the following behavioral alterations: (1) decreased spatial learning and memory 2, 4, and 6 months after the injury, (2) increased proportion of time spent on open vs. closed arms determined by EPM, and (3) accelerated reduction in motor activity observed at 4 months, two months earlier than observed in the sham group, during the EPM testing. There were no significant differences in NSS between injury and sham groups at any of the three time points. Thus, mTBI in male mice led to persistent decreased hippocampal expression of mRNAs that encode critical glucose utilization related enzymes in association with long-term impairments in selected neurobehavioral outcomes.


Subject(s)
Brain Concussion/metabolism , Brain/metabolism , Glucose/metabolism , Hippocampus/metabolism , Mental Disorders/etiology , Animals , Brain Concussion/psychology , Chronic Disease , Disease Models, Animal , Gene Expression , Male , Mice , Mice, Inbred C57BL , Morris Water Maze Test
SELECTION OF CITATIONS
SEARCH DETAIL