37 research outputs found

    A γ-secretase inhibitor, but not a γ-secretase modulator, induced defects in BDNF axonal trafficking and signaling: evidence for a role for APP.

    Get PDF
    Clues to Alzheimer disease (AD) pathogenesis come from a variety of different sources including studies of clinical and neuropathological features, biomarkers, genomics and animal and cellular models. An important role for amyloid precursor protein (APP) and its processing has emerged and considerable interest has been directed at the hypothesis that Aβ peptides induce changes central to pathogenesis. Accordingly, molecules that reduce the levels of Aβ peptides have been discovered such as γ-secretase inhibitors (GSIs) and modulators (GSMs). GSIs and GSMs reduce Aβ levels through very different mechanisms. However, GSIs, but not GSMs, markedly increase the levels of APP CTFs that are increasingly viewed as disrupting neuronal function. Here, we evaluated the effects of GSIs and GSMs on a number of neuronal phenotypes possibly relevant to their use in treatment of AD. We report that GSI disrupted retrograde axonal trafficking of brain-derived neurotrophic factor (BDNF), suppressed BDNF-induced downstream signaling pathways and induced changes in the distribution within neuronal processes of mitochondria and synaptic vesicles. In contrast, treatment with a novel class of GSMs had no significant effect on these measures. Since knockdown of APP by specific siRNA prevented GSI-induced changes in BDNF axonal trafficking and signaling, we concluded that GSI effects on APP processing were responsible, at least in part, for BDNF trafficking and signaling deficits. Our findings argue that with respect to anti-amyloid treatments, even an APP-specific GSI may have deleterious effects and GSMs may serve as a better alternative

    Loss of Caveolin-1 Accelerates Neurodegeneration and Aging

    Get PDF
    The aged brain exhibits a loss in gray matter and a decrease in spines and synaptic densities that may represent a sequela for neurodegenerative diseases such as Alzheimer's. Membrane/lipid rafts (MLR), discrete regions of the plasmalemma enriched in cholesterol, glycosphingolipids, and sphingomyelin, are essential for the development and stabilization of synapses. Caveolin-1 (Cav-1), a cholesterol binding protein organizes synaptic signaling components within MLR. It is unknown whether loss of synapses is dependent on an age-related loss of Cav-1 expression and whether this has implications for neurodegenerative diseases such as Alzheimer's disease.We analyzed brains from young (Yg, 3-6 months), middle age (Md, 12 months), aged (Ag, >18 months), and young Cav-1 KO mice and show that localization of PSD-95, NR2A, NR2B, TrkBR, AMPAR, and Cav-1 to MLR is decreased in aged hippocampi. Young Cav-1 KO mice showed signs of premature neuronal aging and degeneration. Hippocampi synaptosomes from Cav-1 KO mice showed reduced PSD-95, NR2A, NR2B, and Cav-1, an inability to be protected against cerebral ischemia-reperfusion injury compared to young WT mice, increased Aβ, P-Tau, and astrogliosis, decreased cerebrovascular volume compared to young WT mice. As with aged hippocampi, Cav-1 KO brains showed significantly reduced synapses. Neuron-targeted re-expression of Cav-1 in Cav-1 KO neurons in vitro decreased Aβ expression.Therefore, Cav-1 represents a novel control point for healthy neuronal aging and loss of Cav-1 represents a non-mutational model for Alzheimer's disease

    Real-time Imaging of Axonal Transport of Quantum Dot-labeled BDNF in Primary Neurons

    No full text
    BDNF plays an important role in several facets of neuronal survival, differentiation, and function. Structural and functional deficits in axons are increasingly viewed as an early feature of neurodegenerative diseases, including Alzheimer’s disease (AD) and Huntington’s disease (HD). As yet unclear is the mechanism(s) by which axonal injury is induced. We reported the development of a novel technique to produce biologically active, monobiotinylated BDNF (mBtBDNF) that can be used to trace axonal transport of BDNF. Quantum dot-labeled BDNF (QD-BDNF) was produced by conjugating quantum dot 655 to mBtBDNF. A microfluidic device was used to isolate axons from neuron cell bodies. Addition of QD-BDNF to the axonal compartment allowed live imaging of BDNF transport in axons. We demonstrated that QD-BDNF moved essentially exclusively retrogradely, with very few pauses, at a moving velocity of around 1.06 μm/sec. This system can be used to investigate mechanisms of disrupted axonal function in AD or HD, as well as other degenerative disorders

    Swedish Nerve Growth Factor Mutation (NGF R100W

    No full text
    Nerve growth factor (NGF) exerts multiple functions on target neurons throughout development. The recent discovery of a point mutation leading to a change from arginine to tryptophan at residue 100 in the mature NGFβ sequence (NGFR100W) in patients with hereditary sensory and autonomic neuropathy type V (HSAN V) made it possible to distinguish the signaling mechanisms that lead to two functionally different outcomes of NGF: trophic versus nociceptive. We performed extensive biochemical, cellular, and live-imaging experiments to examine the binding and signaling properties of NGFR100W Our results show that, similar to the wild-type NGF (wtNGF), the naturally occurring NGFR100W mutant was capable of binding to and activating the TrkA receptor and its downstream signaling pathways to support neuronal survival and differentiation. However, NGFR100W failed to bind and stimulate the 75 kDa neurotrophic factor receptor (p75NTR)-mediated signaling cascades (i.e., the RhoA-Cofilin pathway). Intraplantar injection of NGFR100W into adult rats induced neither TrkA-mediated thermal nor mechanical acute hyperalgesia, but retained the ability to induce chronic hyperalgesia based on agonism for TrkA signaling. Together, our studies provide evidence that NGFR100W retains trophic support capability through TrkA and one aspect of its nociceptive signaling, but fails to engage p75NTR signaling pathways. Our findings suggest that wtNGF acts via TrkA to regulate the delayed priming of nociceptive responses. The integration of both TrkA and p75NTR signaling thus appears to regulate neuroplastic effects of NGF in peripheral nociception.SIGNIFICANCE STATEMENT In the present study, we characterized the naturally occurring nerve growth factor NGFR100W mutant that is associated with hereditary sensory and autonomic neuropathy type V. We have demonstrated for the first time that NGFR100W retains trophic support capability through TrkA, but fails to engage p75NTR signaling pathways. Furthermore, after intraplantar injection into adult rats, NGFR100W induced neither thermal nor mechanical acute hyperalgesia, but retained the ability to induce chronic hyperalgesia. We have also provided evidence that the integration of both TrkA- and p75NTR-mediated signaling appears to regulate neuroplastic effects of NGF in peripheral nociception. Our study with NGFR100W suggests that it is possible to uncouple trophic effect from nociceptive function, both induced by wild-type NGF

    Differential effects of BMS-299897 and sGSM41 on APP processing.

    No full text
    <p><b>A</b>: A diagram depicts APP processing and the pathways that GSI or GSM treatment differentially affects Aβ peptide formation and the production of APP C-terminal fragments (APP CTFs). First, β-secretase or α-secretase cleaves APP, leading to the production of either β-CTF or α-CTF. Cleavage of β-CTF by γ-secretase at multiple sites yields several Aβ peptides and the APP intracellular domain (AICD). Cleavage of α-CTF by γ-secretase gives rise to and AICD and the P3 fragment. <b>B</b>: Differential effects of GSI and GSM on the production of Aβ species and APP β-CTF [<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0118379#pone.0118379.ref034" target="_blank">34</a>–<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0118379#pone.0118379.ref036" target="_blank">36</a>]. Rat E18 cortical neurons (DIV7) were treated with GSI BMS-299897 (<b>C</b>) or sGSM41 (<b>D</b>) for 24 hrs. The media were harvested and levels of Aβ species (Aβ38, 40, 42) in the media were measured as described in Materials and Methods (n = 3, *P < 0.05, **P < 0.01 using student’s <i>t</i>-test). Treatment with 1μM BMS-299897 or 2.5μM sGSM41 showed the most robust effect and these conditions were used in all other experiments herein. <b>E</b>: Western blotting analyses showing the processing of two substrates of γ-secretase, APP and N-cadherin, in cortical neurons treated with vehicle (0.1% DMSO), 1μM BMS-299897, 2.5μM sGSM41 for 24 hrs. <b>F</b>: Quantitative measurement of mRNA levels by real-time PCR in cortical neurons treated as in <b>E</b>. The mRNA levels are normalized to mRNA levels of β-actin (n = 3, mean±S.E., p values represent results of Student’s <i>t</i>-test).</p

    BMS-299897, not sGSM41, alters the distribution of mitochondria and synaptophysin-positive vesicles within processes of rat E18 hippocampal neurons.

    No full text
    <p><b>A</b>: Experimental design for examining distribution of mitochondria and synaptic vesicle precursors within neuronal processes. Rat E18 hippocampal neurons at DIV4 were treated every 24 hrs with vehicle, BMS-299897 or sGSM41. Neurons at DIV7 were either labeled with MitoTracker for analysis of mitochondria (<b>B-D</b>) or fixed and stained for synaptophysin with a specific antibody (<b>E-G</b>). <b>B</b>: Representative images of DIC and MitoTracker under each treatment conditions. The density of mitochondria (<b>C</b>) and the measurement of mitochondrial profile (<b>D</b>) are quantitated and presented (n = 15–20 neurites for vehicle, sGSM41 samples. For BMS-299897-treated samples, n = 95–150 puncta, mean±S.E., ***P<0.001). <b>E</b>: Representative images of DIC and synaptophysin immmunostaining for each treatment condition. The density of mitochondria (<b>F</b>) and the measurement of mitochondrial profile (<b>G</b>) are quantitated and presented (For density analysis, n = 29–40 neurites, mean±S.E., *P<0.05. For profile measurement, n = 26–27 puncta). Scale bar = 20μm.</p
    corecore