Elsevier

Neurobiology of Aging

Volume 50, February 2017, Pages 13-24
Neurobiology of Aging

Regular article
PGE2-EP3 signaling pathway impairs hippocampal presynaptic long-term plasticity in a mouse model of Alzheimer's disease

https://doi.org/10.1016/j.neurobiolaging.2016.10.012Get rights and content

Abstract

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by early cognitive deficits linked to synaptic dysfunction and loss. Considerable evidence suggests that neuroinflammation contributes to AD. Prostaglandin E2 (PGE2), a key neuroinflammatory molecule, modulates hippocampal synaptic transmission and plasticity. We investigated the effect of PGE2 on synaptic transmission and presynaptic plasticity at synapses between mossy fibers from the dentate gyrus and CA3 pyramidal cells (Mf-CA3 synapse). These synapses are involved in mnemonic processes and consequently may be of relevance for AD. We provide evidence that although PGE2 had no effect both on either basal transmission or short-term plasticity, it strongly impaired presynaptic Mf-CA3 long-term potentiation (LTP) by acting on PGE2 receptor 3 (EP3) receptors. During aging, hippocampal levels of PGE2 markedly increased in the APP/PS1 mouse model of AD and impaired specifically presynaptic LTP via a PGE2-EP3 signaling pathway. In summary, the building up of PGE2 during the progression of AD leads to specific impairment of hippocampal presynaptic plasticity and highlights EP3 receptors as a potential target to alleviate cognitive deficits in AD.

Introduction

Alzheimer's disease (AD) is a progressive and devastating neurodegenerative disease characterized by deficits in learning and memory processes. There is considerable evidence that neuroinflammation involving the activation of glial cells contributes to the disease progression and pathology (Akiyama et al., 2000, Heneka and O'Banion, 2007). As such, epidemiological studies have revealed that chronic intake of nonsteroidal anti-inflammatory drugs (NSAIDs) reduced the prevalence of AD (McGeer and McGeer, 2007). Furthermore, clinical trials have revealed that NSAIDs, when given to asymptomatic patients, reduce AD incidence, whereas they have adverse effect on AD pathogenesis in its later stages (Breitner et al., 2011).

When activated in neuroinflammatory conditions, glia release a plethora of neuroinflammatory molecules such as inflammatory cytokines, chemokines, and prostanoids (Eikelenboom et al., 1994). The prostanoid prostaglandin E2 (PGE2) plays pivotal functions in inflammation (Bos et al., 2004). PGE2 is produced from arachidonic acid by the microsomal prostaglandin-E2 synthase (mPGES) and by 2 rate-limiting enzymes, Cox-1 and Cox-2 (Smith et al., 1991), which are expressed by hippocampal neurons (Yasojima et al., 1999), astrocytes and microglia (Font-Nieves et al., 2012). Although Cox-1 is constitutively expressed, Cox-2 and mPGES are strongly activated by and during neuroinflammation (Font-Nieves et al., 2012). PGE2 can bind to 4 different subtypes of G-protein–coupled receptors (EP1–4), which regulate adenylyl cyclase (AC) activity and/or phosphoinositol turnover and intracellular calcium mobilization (Breyer et al., 2001).

PGE2 plays an important role in the pathophysiology of AD. It is a primary target of NSAIDs, and its level is elevated together with the expression of Cox-2 in the brain of AD patients (Kitamura et al., 1999, Montine et al., 1999, Yasojima et al., 1999). Both in vitro and in vivo, PGE2 stimulates amyloid beta (Aß) production by microglial cells, astrocytes, and neurons (Hoshino et al., 2007). Selective inhibition of Cox-2 by NS398 acutely prevents the Aß-induced impairment of long-term potentiation (LTP) of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors–mediated synaptic transmission at CA3–CA1 synapses (Kotilinek et al., 2008). Exogenous addition of PGE2 did not impair by itself LTP in wild-type mice but prevented the ability of NS398 to restore LTP impairment induced by synthetic Aß suggesting complex interactions between PGE2, Aβ, and postsynaptic forms of LTP (Kotilinek et al., 2008).

AD, at least in its early stage, is thought to involve synaptic dysfunction and loss (Jacobsen et al., 2006, Selkoe, 2002, Sheng et al., 2012). Connections between the dentate gyrus (DG) and the hippocampal CA3 region through mossy fiber (Mf) synapses have been proposed to participate in the rapid encoding of novel memory (Kesner, 2007), a mnemonic process particularly affected in AD conditions. Mf synapses onto CA3 pyramidal cells display a wide dynamic range of presynaptic plasticity, including prominent short-term plasticity and a form of LTP that is independent of N-methyl-D-aspartate (NMDA) receptors (Henze et al., 2002, Salin et al., 1996). Alterations in Mf synaptic function in the context of AD have only been studied in senescent (24- to 25-month old) Tg2576 mice that is long after a massive load of amyloid plaques (Witton et al., 2010).

Here we thus studied Mf-CA3 synaptic function in the APPswe/PS1ΔE9 (APP/PS1) mouse model of AD in relation with PGE2. APP/PS1 mice are characterized by an over-production of Aß protein leading to a noticeable load of amyloid plaques around 12 months of age (Jankowsky et al., 2004) and display synaptic dysfunction (Volianskis et al., 2010) and learning deficits (Lagadec et al., 2012, Reiserer et al., 2007). We report that acute application of PGE2 impairs presynaptic Mf-LTP in young mice through activation of EP3 receptors. In APP/PS1 mice, presynaptic Mf-LTP was noticeably impaired at 12 months of age in parallel with a building up of endogeneous levels of PGE2 in the hippocampus. Mf-LTP could be fully rescued by blockade of EP3 receptors. Considering the diverse physiological roles of PGE2, directly targeting the EP3 receptor may prove to be a more specific therapeutical strategy in AD than the global inhibition of prostaglandins by NSAIDs.

Section snippets

Ethical approval

Animal anesthesia and euthanasia procedures were carried out in accordance with the Animal Protection Association of ethical standards and the French legislation concerning animal experimentation and were approved by the University of Bordeaux/CNRS Animal Care and Use Committee (#55).

Animals

The animals used in this study were male APP/PS1 mice obtained from Jackson Laboratory (Bar Harbor, ME, USA) and their wild-type (WT) littermates (C57BL6/J). The APP/PS1 mice express a chimeric mouse/human amyloid

PGE2 impairs presynaptic long-term potentiation at Mf–CA3 synapses

We tested the effects of PGE2 on Mf-EPSCs recorded from CA3 pyramidal cells of P19–P21 wild-type mice using the whole-cell voltage clamp mode of the patch clamp technique (Fig. 1A). Mf-EPSCs evoked by minimal intensity stimulation at low frequency (0.1 Hz) were not affected by bath application of 1–10 μM PGE2 (Fig. 1B). Mf-CA3 synapses display prominent forms of presynaptic short-term plasticity (Nicoll and Schmitz, 2005), including paired-pulse facilitation (PPF: 2 stimulations separated by

Discussion

Our findings demonstrate that PGE2, acting through the EP3 receptor subtype, inhibits presynaptic long-term plasticity at Mf-CA3 synapses. We further provide evidence that in the APP/PS1 mouse model of AD, the PGE2-EP3 receptor signaling pathway is responsible for the impairment of presynaptic LTP at Mf-CA3 synapses by showing that the blockade of EP3 receptors fully rescues the LTP at 12-month-old, an age characterized by a neuroinflammatory reaction and chronically elevated levels of the

Disclosure statement

The authors have no conflicts of interest to disclose.

Acknowledgements

The authors thank Sílvia Viana da Silva for help with electrophysiological recordings and fruitful discussions and Noelle Grosjean for genotyping APPswe/PS1ΔE9 mice. The authors thank Ashley L. Kees for careful reading of the manuscript and Benoit Silvestre de Ferron for the cartoon shown in Fig. 1A. The authors also thank Marlène Maitre (Microdissection Laser plateform, Neurocentre Magendie, Inserm U862), Thierry Lesté-Lasserre (Genotyping plateform, Neurocentre Magendie, Inserm U862) for

References (63)

  • A. Irie et al.

    Mouse prostaglandin E receptor EP3 subtype mediates calcium signals via Gi in cDNA-transfected Chinese hamster ovary cells

    Biochem. Biophys. Res. Commun.

    (1994)
  • Y. Kitamura et al.

    Increased expression of cyclooxygenases and peroxisome proliferator-activated receptor-gamma in Alzheimer's disease brains

    Biochem. Biophys. Res. Commun.

    (1999)
  • K.J. Livak et al.

    Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T))

    Methods

    (2001)
  • P.L. McGeer et al.

    NSAIDs and Alzheimer disease: epidemiological, animal model and clinical studies

    Neurobiol. Aging

    (2007)
  • A. Mori et al.

    The prostanoid EP(2) receptor agonist ONO-AE1-259-01 protects against glutamate-induced neurotoxicity in rat retina

    Eur. J. Pharmacol.

    (2009)
  • M.D. Nielsen et al.

    Differential regulation of type I and type VIII Ca2+-stimulated adenylyl cyclases by Gi-coupled receptors in vivo

    J. Biol. Chem.

    (1996)
  • J.M. Rall et al.

    Intrahippocampal infusion of a cyclooxygenase-2 inhibitor attenuates memory acquisition in rats

    Brain Res.

    (2003)
  • M. Sharifzadeh et al.

    A time course analysis of cyclooxygenase-2 suggests a role in spatial memory retrieval in rats

    Neurosci. Res.

    (2006)
  • H. Slawik

    Microglial expression of prostaglandin EP3 receptor in excitotoxic lesions in the rat striatum

    Neurochem. Int.

    (2004)
  • W.L. Smith et al.

    Prostaglandin and thromboxane biosynthesis

    Pharmacol. Ther.

    (1991)
  • A. Volianskis et al.

    Episodic memory deficits are not related to altered glutamatergic synaptic transmission and plasticity in the CA1 hippocampus of the APPswe/PS1δE9-deleted transgenic mice model of ß-amyloidosis

    Neurobiol. Aging

    (2010)
  • Z.G. Xia et al.

    Distribution of mRNA for the calmodulin-sensitive adenylate cyclase in rat brain: expression in areas associated with learning and memory

    Neuron

    (1991)
  • K. Yasojima et al.

    Distribution of cyclooxygenase-1 and cyclooxygenase-2 mRNAs and proteins in human brain and peripheral organs

    Brain Res.

    (1999)
  • Y. Akitake et al.

    Microsomal prostaglandin E synthase-1 is induced in Alzheimer's disease and its deletion mitigates Alzheimer's disease-like pathology in a mouse model

    J. Neurosci. Res.

    (2013)
  • K.I. Andreasson et al.

    Age-dependent cognitive deficits and neuronal apoptosis in cyclooxygenase-2 transgenic mice

    J. Neurosci.

    (2001)
  • R.M. Breyer et al.

    Prostanoid receptors: subtypes and signaling

    Annu. Rev. Pharmacol. Toxicol.

    (2001)
  • S.A. Bustin et al.

    The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments

    Clin. Chem.

    (2009)
  • C. Chen et al.

    Endogenous PGE2 regulates membrane excitability and synaptic transmission in hippocampal CA1 pyramidal neurons

    J. Neurophysiol.

    (2005)
  • C. Chen et al.

    Cyclooxygenase-2 regulates prostaglandin E2 signaling in hippocampal long-term synaptic plasticity

    J. Neurophysiol.

    (2002)
  • R.A. Coleman et al.

    New evidence with selective agonists and antagonists for the subclassification of PGE2-sensitive (EP) receptors

    Adv. Prostaglandin Thromboxane Leukot. Res.

    (1987)
  • T.R. Cowley et al.

    COX-2, but not COX-1, activity is necessary for the induction of perforant path long-term potentiation and spatial learning in vivo

    Eur. J. Neurosci.

    (2008)
  • Cited by (21)

    • Aβ oligomers from human brain impair mossy fiber LTP in CA3 of hippocampus, but activating cAMP-PKA and cGMP-PKG prevents this

      2022, Neurobiology of Disease
      Citation Excerpt :

      oAβ is already known to impair LTP in the CA1 area of hippocampus through the cAMP/PKA/CREB or cGMP/PKG/CREB pathways (see e.g., Vitolo et al., 2002; Puzzo et al., 2005). Several studies have observed an age-dependent impairment of mossy fiber LTP in APP transgenic mice, including for example 12 mo transgenic AAP/PS1 mice and 24 mo Tg2576 mice (Maingret et al., 2017; Witton et al., 2010). However, it is not possible to definitively attribute these changes in mice to oAβ per se, as heterogenous Aβ species and many secondary effectors are present in vivo.

    • Bifenthrin insecticide promotes oxidative stress and increases inflammatory mediators in human neuroblastoma cells through NF-kappaB pathway

      2020, Toxicology in Vitro
      Citation Excerpt :

      In contrast, BF reduced the expression of Nrf-2, an antioxidant transcription factor. These inflammatory molecules are known to be important mediators of neuroinflammation and neurodegeneration associated with several neurological conditions, including AD and Parkinson's disease (PD) (Kang et al., 2017; King et al., 2019; Maingret et al., 2017; Miron et al., 2019; Souchet et al., 2019). Neurons are mainly susceptible to oxidative damage due to the presence of high levels of polyunsaturated fatty acids, which are sensitive to peroxidation and oxidative changes (Butterfield et al., 2002; Floyd and Carney, 1992; Uttara et al., 2009).

    • Interplay between inflammation and neural plasticity: Both immune activation and suppression impair LTP and BDNF expression

      2019, Brain, Behavior, and Immunity
      Citation Excerpt :

      As an example, endogenous basal levels of prostaglandin E2 (PGE2) regulate membrane excitability, synaptic transmission, and plasticity in hippocampal CA1 pyramidal neurons (Chen and Bazan, 2005; Chen et al., 2002) and are necessary for memory acquisition (Rall et al., 2003), consolidation (Teather et al., 2002), and retention (Cowley et al., 2008; Sharifzadeh et al., 2006; Shaw et al., 2003). Similarly, cytokines such as Interleukin (IL)-1β, IL-1 receptor antagonist (IL-1RA) and transforming growth factor (TGF)-β play functional roles in the mechanisms of synaptic plasticity and cognitive functions (Levin and Godukhin, 2017; Maingret et al., 2017; Ross et al., 2003). According to this new emerging framework, inflammatory mediators and brain immune cells would be critical in regulating neural plasticity, including processes such as growing and pruning of dendrites and axons, shaping of synapses and associated structures, apoptosis and neurogenesis of neurons (Yirmiya and Goshen, 2011).

    • Ethanol-induced PGE<inf>2</inf> up-regulates Aβ production through PKA/CREB signaling pathway

      2017, Biochimica et Biophysica Acta - Molecular Basis of Disease
      Citation Excerpt :

      In an AD animal model, deletion of the EP-2 receptor counteracts the aggravation of AD [60]. Another recent study showed the essential role of EP-3 downstream signals which intensify the cognitive deficits in AD [61]. Concerning EP receptors, our results showed that ethanol significantly up-regulates EP-2 receptor compared to the EP-3 receptor.

    • Closing the gap: long-term presynaptic plasticity in brain function and disease

      2017, Current Opinion in Neurobiology
      Citation Excerpt :

      Fmr1 knockout mice also show impaired presynaptic LTP of excitatory transmission in the anterior cingulate cortex [65]. Lastly, altered presynaptic plasticity has been reported in animal models of neurodegenerative diseases, including Alzheimer’s disease [66,67] and Parkinson’s disease [68]. Whether and how presynaptic plasticity alterations contribute to neurodegenerative diseases in humans remains to be determined.

    View all citing articles on Scopus
    View full text