27 research outputs found

    M-Current Inhibition in Hippocampal Excitatory Neurons Triggers Intrinsic and Synaptic Homeostatic Responses at Different Temporal Scales

    Get PDF
    Persistent alterations in neuronal activity elicit homeostatic plastic changes in synaptic transmission and/or intrinsic excitability. However, it is unknown whether these homeostatic processes operate in concert or at different temporal scales to maintain network activity around a set-point value. Here we show that chronic neuronal hyperactivity, induced by M-channel inhibition, triggered intrinsic and synaptic homeostatic plasticity at different timescales in cultured hippocampal pyramidal neurons from mice of either sex. Homeostatic changes of intrinsic excitability occurred at a fast timescale (1–4 h) and depended on ongoing spiking activity. This fast intrinsic adaptation included plastic changes in the threshold current and a distal relocation of FGF14, a protein physically bridging Nav1.6 and Kv7.2 channels along the axon initial segment. In contrast, synaptic adaptations occurred at a slower timescale (∼2 d) and involved decreases in miniature EPSC amplitude. To examine how these temporally distinct homeostatic responses influenced hippocampal network activity, we quantified the rate of spontaneous spiking measured by multielectrode arrays at extended timescales. M-Channel blockade triggered slow homeostatic renormalization of the mean firing rate (MFR), concomitantly accompanied by a slow synaptic adaptation. Thus, the fast intrinsic adaptation of excitatory neurons is not sufficient to account for the homeostatic normalization of the MFR. In striking contrast, homeostatic adaptations of intrinsic excitability and spontaneous MFR failed in hippocampal GABAergic inhibitory neurons, which remained hyperexcitable following chronic M-channel blockage. Our results indicate that a single perturbation such as M-channel inhibition triggers multiple homeostatic mechanisms that operate at different timescales to maintain network mean firing rate. SIGNIFICANCE STATEMENT: Persistent alterations in synaptic input elicit homeostatic plastic changes in neuronal activity. Here we show that chronic neuronal hyperexcitability, induced by M-type potassium channel inhibition, triggered intrinsic and synaptic homeostatic plasticity at different timescales in hippocampal excitatory neurons. The data indicate that the fast adaptation of intrinsic excitability depends on ongoing spiking activity but is not sufficient to provide homeostasis of the mean firing rate. Our results show that a single perturbation such as M-channel inhibition can trigger multiple homeostatic processes that operate at different timescales to maintain network mean firing rate

    Enhancement of Learning and Memory by Elevating Brain Magnesium

    Get PDF
    Learning and memory are fundamental brain functions affected by dietary and environmental factors. Here, we show that increasing brain magnesium using a newly developed magnesium compound (magnesium-L-threonate, MgT) leads to the enhancement of learning abilities, working memory, and short- and long-term memory in rats. The pattern completion ability was also improved in aged rats. MgT-treated rats had higher density of synaptophysin-/synaptobrevin-positive puncta in DG and CA1 subregions of hippocampus that were correlated with memory improvement. Functionally, magnesium increased the number of functional presynaptic release sites, while it reduced their release probability. The resultant synaptic reconfiguration enabled selective enhancement of synaptic transmission for burst inputs. Coupled with concurrent upregulation of NR2B-containing NMDA receptors and its downstream signaling, synaptic plasticity induced by correlated inputs was enhanced. Our findings suggest that an increase in brain magnesium enhances both short-term synaptic facilitation and long-term potentiation and improves learning and memory functions.National Institutes of Health (U.S.) (NS37342)National Basic Research Program of China (2006CB3031)National Basic Research Program of China (2009CB941303)National Natural Science Foundation (China) (30630026)National High Technology Research and Development Program of China (2007AA02Z443)Tsinghua-Yue-Yuen Medical Sciences Fun

    APP Homodimers Transduce an Amyloid-β-Mediated Increase in Release Probability at Excitatory Synapses

    Get PDF
    SummaryAccumulation of amyloid-β peptides (Aβ), the proteolytic products of the amyloid precursor protein (APP), induces a variety of synaptic dysfunctions ranging from hyperactivity to depression that are thought to cause cognitive decline in Alzheimer’s disease. While depression of synaptic transmission has been extensively studied, the mechanisms underlying synaptic hyperactivity remain unknown. Here, we show that Aβ40 monomers and dimers augment release probability through local fine-tuning of APP-APP interactions at excitatory hippocampal boutons. Aβ40 binds to the APP, increases the APP homodimer fraction at the plasma membrane, and promotes APP-APP interactions. The APP activation induces structural rearrangements in the APP/Gi/o-protein complex, boosting presynaptic calcium flux and vesicle release. The APP growth-factor-like domain (GFLD) mediates APP-APP conformational changes and presynaptic enhancement. Thus, the APP homodimer constitutes a presynaptic receptor that transduces signal from Aβ40 to glutamate release. Excessive APP activation may initiate a positive feedback loop, contributing to hippocampal hyperactivity in Alzheimer’s disease

    Early alterations in the MCH system link aberrant neuronal activity and sleep disturbances in a mouse model of Alzheimer's disease.

    Get PDF
    Early Alzheimer's disease (AD) is associated with hippocampal hyperactivity and decreased sleep quality. Here we show that homeostatic mechanisms transiently counteract the increased excitatory drive to CA1 neurons in AppNL-G-F mice, but that this mechanism fails in older mice. Spatial transcriptomics analysis identifies Pmch as part of the adaptive response in AppNL-G-F mice. Pmch encodes melanin-concentrating hormone (MCH), which is produced in sleep-active lateral hypothalamic neurons that project to CA1 and modulate memory. We show that MCH downregulates synaptic transmission, modulates firing rate homeostasis in hippocampal neurons and reverses the increased excitatory drive to CA1 neurons in AppNL-G-F mice. AppNL-G-F mice spend less time in rapid eye movement (REM) sleep. AppNL-G-F mice and individuals with AD show progressive changes in morphology of CA1-projecting MCH axons. Our findings identify the MCH system as vulnerable in early AD and suggest that impaired MCH-system function contributes to aberrant excitatory drive and sleep defects, which can compromise hippocampus-dependent functions

    Mutant SOD1 Increases APP Expression and Phosphorylation in Cellular and Animal Models of ALS.

    No full text
    Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease and it is the most common adult onset neurodegenerative disorder affecting motor neurons. There is currently no effective treatment for ALS and our understanding of the pathological mechanism is still far away from prevention and/or treatment of this devastating disease. Amyloid precursor protein (APP) is a transmembrane protein that undergoes processing either by β-secretase or α-secretase, followed by γ-secretase. In the present study, we show that APP levels, and aberrant phosphorylation, which is associated with enhanced β-secretase cleavage, are increased in SOD1G93A ALS mouse model. Fluorescence resonance energy transfer (FRET) analysis suggests a close interaction between SOD1 and APP at hippocampal synapses. Notably, SOD1G93A mutation induces APP-SOD1 conformational changes, indicating a crosstalk between these two signaling proteins. Inhibition of APP processing via monoclonal antibody called BBS that blocks APP β-secretase cleavage site, resulted in reduction of mutant SOD1G93A levels in animal and cellular models of ALS, significantly prolonged life span of SOD1G93A mice and diminished inflammation. Beyond its effect on toxic mutant SOD1G93A, BBS treatment resulted in a reduction in the levels of APP, its processing product soluble APPβ and pro-apoptotic p53. This study demonstrates that APP and its processing products contribute to ALS pathology through several different pathways; thus BBS antibody could be a promising neuroprotective strategy for treatment of this disease

    BBS treatment leads increase in survival of SOD1<sup>G93A</sup> transgenic mice.

    No full text
    <p>55-day-old female and male SODG93A mice received a weekly dose of 3mg/kg MAb BBS via i.p. injection. Controls were treated with PBS. A. Weight of each animal was recorded weekly. B. Motor functions of the i.p. treated mice were assessed by performing Rotarod test. Mice were trained to run on the 2-cm-diameter rod, which rotated at a fixed speed of 13 turns per minute. Mice were allowed to run for up to 1 min in each trial, or until they fell off. C. Plots of disease onset (Median BBS = 95; Median PBS = 91.5; *p <0.05; Average BBS = 97.2; Average = 91.9; *p <0.05), survival(Median BBS = 95; Median PBS = 91.5; *p <0.05; Average BBS = 97.2; Average = 91.9; *p <0.05) and the average survival in days. N (PBS) = 24, N (BBS) = 24.</p

    BBS treatment leads to decrease in p53 levels and astrogliosis marker GFAP.

    No full text
    <p>A. The levels of p53were quantified using immunoblot analysis in the soluble fraction of spinal cord homogenates of SOD<sup>G93A</sup> mice treated with BBS or non relevant MAb for 42 days. p53 levels were detected using PAb 240 antibody and normalized to β-Actin. All values are expressed as the mean ± SEM. Statistical comparisons were performed using Student's t-test. N(BBS) = 4, N(Non relevant) = 5. *<i>p</i> < 0.05; **<i>p</i> < 0.01 (v.s. Non relevant). B. Lumber spinal cords were subjected to immunohistochemical analysis. Spinal cord sections were stained with anti-GFAP antibody, and the intensity of the staining was analyzed using Image-J Software. Scale bars in panel B correspond to 1 mm, respectively. Statistical comparisons were performed using Student's t-test. N(BBS) = 5, N(Non relevant) = 5. **<i>p</i> < 0.01 (v.s. Non relevant).</p

    APP expression and phosphorylation in spinal cords of pre-symptomatic and symptomatic mice.

    No full text
    <p>Spinal cords from 30 -and 80-day-old and end stage SOD1<sup>G93A</sup> mice were homogenized and their membrane fractions were subjected to immunoblot analysis. Age matched NT littermates served as control. For APP detection 22C11 antibody was used. For detection of phosphorylated APP (pAPP) a specific polyclonal anti pAPP T688 was used. β-Actin was used as an internal loading control. Immunoblot blot and densitometric analysis of the relative expression and phosphorylation levels of APP in the spinal cord of (A) 30-day-old mice, (B) 80-day-old and end stage SOD1G93A mice. All values are expressed as the mean ± SEM. Statistical comparisons were performed using one-way ANOVA followed by Tukey–Kramer post hoc test. N(NT 30D) = 3, N(G93A 30D) = 3, N(NT = 4), N(G93A 80D) = 4, N(G93A end stage = 3).*p < 0.05; **p < 0.01 (v.s. NT littermates).</p
    corecore