17 research outputs found

    Reduced Expression of the PP2A Methylesterase, PME-1, or the PP2A Methyltransferase, LCMT-1, Alters Sensitivity to Beta-Amyloid-Induced Cognitive and Electrophysiological Impairments in Mice

    Get PDF
    Beta-amyloid (Ab) is thought to play a critical role in Alzheimer’s disease (AD), and application of soluble oligomeric forms of Ab produces AD-like impairments in cognition and synaptic plasticity in experimental systems. We found previously that transgenic overexpression of the PP2A methylesterase, PME-1, or the PP2A methyltransferase, LCMT-1, altered the sensitivity of mice to Ab-induced impairments, suggesting that PME-1 inhibition may be an effective approach for preventing or treating these impairments. To explore this possibility, we examined the behavioral and electrophysiological effects of acutely applied synthetic Ab oligomers in male and female mice heterozygous for either a PME-1 KO or an LCMT-1 gene-trap mutation. We found that heterozygous PME-1 KO mice were resistant to Ab-induced impairments in cognition and synaptic plasticity, whereas LCMT-1 gene-trap mice showed increased sensitivity to Ab-induced impairments. The heterozygous PME-1 KO mice produced normal levels of endogenous Ab and exhibited normal electrophysiological responses to picomolar concentrations of Ab, suggesting that reduced PME-1 expression in these animals protects against Ab-induced impairments without impacting normal physiological Ab functions. Together, these data provide additional support for roles for PME-1 and LCMT-1 in regulating sensitivity to Ab-induced impairments, and suggest that inhibition of PME-1 may constitute a viable therapeutic approach for selectively protecting against the pathologic actions of Ab in AD

    Co‑expressed microRNAs, target genes and pathways related to metabolism, inflammation and endocrine function in individuals at risk for type 2 diabetes.

    No full text
    MicroRNAs (miRNAs) may be considered important regulators of risk for type 2 diabetes (T2D). The aim of the present study was to identify novel sets of miRNAs associated with T2D risk, as well as their gene and pathway targets. Circulating miRNAs (n=59) were measured in plasma from participants in a previously completed clinical trial (n=82). An agnostic statistical approach was applied to identify novel sets of miRNAs with optimal co‑expression patterns. In silico analyses were used to identify the messenger RNA and biological pathway targets of the miRNAs within each factor. A total of three factors of miRNAs were identified, containing 18, seven and two miRNAs each. Eight biological pathways were revealed to contain genes targeted by the miRNAs in all three factors, 38 pathways contained genes targeted by the miRNAs in two factors, and 55, 18 and two pathways were targeted by the miRNAs in a single factor, respectively (all q<0.05). The pathways containing genes targeted by miRNAs in the largest factor shared a common theme of biological processes related to metabolism and inflammation. By contrast, the pathways containing genes targeted by miRNAs in the second largest factor were related to endocrine function and hormone activity. The present study focused on the pathways uniquely targeted by each factor of miRNAs in order to identify unique mechanisms that may be associated with a subset of individuals. Further exploration of the genes and pathways related to these biological themes may provide insights about the subtypes of T2D and lead to the identification of novel therapeutic targets

    Chronic EHT treatment prevents Aβ-induced impairment of long-term potentiation.

    No full text
    <p><b>(</b>A-B) Time course of averaged Schaffer collateral fEPSP responses (± SEM) in hippocampal slices prepared from animals fed control or EHT-containing diets and treated with either vehicle or 100 nM Aβ (horizontal bar) 20 min prior to delivery of theta-burst stimulation (arrow). (A) Aβ treatment significantly reduces potentiated responses following TBS in slices prepared from animals on control diets (2-way RM-ANOVA for treatment with time and treatment as factors: F(1,29) = 8.913, P = 0.0057). (B) Mice fed diets containing 0.01% EHT are resistant to Aβ-induced LTP impairment (2-way RM-ANOVA for treatment with time and treatment as factors: F(1,26) = 0.0943, P = 0.7612). C) Input/output (2-way RM-ANOVA for treatment with stimulus and treatment as factors: F(1,57) = 0.5466, P = 0.4628) (N = 31 control, 28 0.01% EHT).</p

    EHT treatment reduces tau phosphorylation.

    No full text
    <p><b>(</b>A) Representative western blots for the indicated proteins and methylated PP2A/C performed on hippocampal homogenates prepared from animals fed control diet, or diets containing 0.01 or 0.1% EHT. (B) Histogram showing average ± SEM for tubulin-normalized band intensities expressed as average percent of control band intensity from replicate western blots in A show no significant differences in expression levels for any of the indicated proteins or for methylated PP2A/C (ANOVA for: PP2A/C: F(2,22) = 0.2533, P = 0.7784; PP2A/A: F(2,22) = 0.2588, P = 0.7743; B55α: F(2,22) = 0.06221, P = 0.9399; PME-1: F(2,22) = 0.4942, P = 0.6167; LCMT-1: F(2,22) = 0.2498, P = 0.7812; methyl-PP2A/C: F(2,22) = 0.1666, P = 0.8476). (C) Representative western blots for demethylated PP2A, and total PP2A/C performed on the homogenates described in A either treated (+) or mock treated (-) with 0.5 M sodium hydroxide. (D) Histogram of average demethylated PP2A/C (± SEM) in hippocampal homogenates prepared from animals fed control, or 0.01 or 0.1% EHT containing-diets show no significant differences in demethylated PP2A/C levels (ANOVA: F(2,22) = 0.1436, P = 0.8670). Values were calculated as ratios of demethyl-PP2A/C to total PP2A/C band intensities for -NaOH treated samples from replicate western blots shown in C and expressed as percent of the average of control. (E) Representative western blots performed on hippocampal homogenates prepared from animals fed control diet, or diets containing 0.01 or 0.1% EHT for phospho-Ser396/404 (PHF1), phospho-Ser202 (CP13) together with their corresponding total tau loading controls, as well as and total tau together with its corresponding β-actin loading control. (F) Histogram showing average band intensities ± SEM for phospho-Ser396/404-tau (PHF1), phospho-Ser202-tau (CP13) normalized to corresponding total tau loading control, and total tau normalized to corresponding β-actin loading control for replicate western blots shown in H show a trend for reduced phosphorylation at these sites in EHT-treated animals (ANOVA for: PHF1: F(2,22) = 5.147, P = 0.147, Bonferroni post-hoc for PHF1 0.1% EHT vs. Control: t = 3.154, P = 0.0092; CP13: F(2,22) = 1.433, P = 0.2599; total tau: F(2,22) = 0.1268, P = 0.8815). (G and H) Representative western blots for phospho-Ser9 and total GSK3B and phospho-Ser133 and total Creb performed on hippocampal homogenates prepared from animals fed control diet, or diets containing 0.01 or 0.1% EHT. (I) Histogram showing average ± SEM of phospho-GSK3β and phospho-Creb band intensities normalized to corresponding total GSK3β and Creb respectively for replicate western blots shown in G&H show no significant effect of EHT treatment on phosphorylation at these sites (ANOVA for: P-GSK3β F(2,22) = 1.761, P = 0.1952; P-Creb: F(2,22) = 0.1776, P = 0.8385). (N = 8 control, 8 0.01% EHT and 9 0.1% EHT treated animals for each measure).</p
    corecore