Elsevier

Neurobiology of Aging

Volume 36, Issue 2, February 2015, Pages 886-900
Neurobiology of Aging

Regular article
Intraneuronal Aβ accumulation induces hippocampal neuron hyperexcitability through A-type K+ current inhibition mediated by activation of caspases and GSK-3

https://doi.org/10.1016/j.neurobiolaging.2014.10.034Get rights and content

Abstract

Amyloid β-protein (Aβ) pathologies have been linked to dysfunction of excitability in neurons of the hippocampal circuit, but the molecular mechanisms underlying this process are still poorly understood. Here, we applied whole-cell patch-clamp electrophysiology to primary hippocampal neurons and show that intracellular Aβ42 delivery leads to increased spike discharge and action potential broadening through downregulation of A-type K+ currents. Pharmacologic studies showed that caspases and glycogen synthase kinase 3 (GSK-3) activation are required for these Aβ42-induced effects. Extracellular perfusion and subsequent internalization of Aβ42 increase spike discharge and promote GSK-3-dependent phosphorylation of the Kv4.2 α-subunit, a molecular determinant of A-type K+ currents, at Ser-616. In acute hippocampal slices derived from an adult triple-transgenic Alzheimer's mouse model, characterized by endogenous intracellular accumulation of Aβ42, CA1 pyramidal neurons exhibit hyperexcitability accompanied by increased phosphorylation of Kv4.2 at Ser-616. Collectively, these data suggest that intraneuronal Aβ42 accumulation leads to an intracellular cascade culminating into caspases activation and GSK-3-dependent phosphorylation of Kv4.2 channels. These findings provide new insights into the toxic mechanisms triggered by intracellular Aβ42 and offer potentially new therapeutic targets for Alzheimer's disease treatment.

Introduction

Alzheimer's disease (AD) is the most common form of dementia with a high prevalence among the aging population (Citron, 2010). Extracellular fibrillar amyloid β-protein (Aβ) accumulation and neurofibrillary tangles composed of hyperphosphorylated tau are considered the neuropathologic hallmarks of AD (Selkoe, 2001). Aβ originates from the amyloid β-precursor protein (APP) through sequential cleavage by the β- and the γ-secretase enzyme complex (Thinakaran and Koo, 2008). Most of the full-length Aβ peptide consists of the 40-amino-acid long Aβ (Aβ40), whereas a small proportion (10%) is the 42-residues variant (Aβ42), a species that because of its high hydrophobicity has been considered more prone to fibril formation and neurotoxicity than the Aβ40 (Jarrett et al., 1993).

A plethora of neurophysiological studies have reported toxic activity of Aβ42 oligomers on synaptic function and activity-dependent plasticity in the hippocampus and cerebral cortex building the notion that AD is a synaptopathy (Crimins et al., 2013, Klyubin et al., 2012, Selkoe, 2002, Shankar et al., 2008, Sheng et al., 2012). It is believed, indeed, that disruption in synaptic structure and function induced by Aβ42 is one of the underlying mechanisms leading to the aberrant neuronal processing and network dysfunction that characterize AD cognitive impairment and memory loss (Mucke and Selkoe, 2012). Surprisingly, although, much less knowledge has been gained on other Aβ toxic effects on neuronal activity that might precede synaptic dysfunction and as such could be targeted for early therapeutic interventions against AD.

Emerging evidence indicates that changes in neuronal excitability could play a fundamental role in initiating early Aβ42 pathology predisposing to and/or triggering subsequent synaptic dysfunction. In vitro and animal model studies have demonstrated that Aβ pathology associates with increased excitability of hippocampal neurons, leading to hypersynchronous network activity and higher risk for seizures (Born et al., 2014, Brown et al., 2011, Busche et al., 2012, Davis et al., 2014, Del Vecchio et al., 2004, Minkeviciene et al., 2009, Palop et al., 2007, Putcha et al., 2011). Epidemiologic studies have confirmed comorbidity between patients with AD and epilepsy, showing increased risk of seizures in AD patients (Amatniek et al., 2006, Hommet et al., 2008). This repertoire of cellular and clinical studies indicating potential commonalities in early AD and epilepsy opens new horizons in the understanding of early AD triggers and therapies. However, despite this evidence, the molecular mechanisms linking Aβ pathology to aberrant neuronal firing is still poorly understood. A missing link in associating Aβ effects to neuronal excitability is the identification of the source of toxic oligomers.

Early studies based on the observation of senile plaques in the extracellular space (Hardy and Selkoe, 2002) have pointed for extracellular Aβ42 (eAβ42) as the primary cause of AD. However, in studies on postmortem AD and mild cognitively impaired patients immunoreactivity of intracellular Aβ42 (iAβ42) has been identified in neurons of the hippocampus and entorhinal cortex (Gouras et al., 2000, Mori et al., 2002), the 2 main brain regions affected in early AD pathology. These findings have been corroborated by results from transgenic mouse models (Guzmán et al., 2014, Youmans et al., 2012; for review see; LaFerla et al., 2007), leading to the overall hypothesis of iAβ accumulation as an early triggering event in the AD progression preceding senile plaques (Kuo et al., 2001, LaFerla et al., 2007, Oakley et al., 2006). Intracellular Aβ-driven effects on neuronal firing might therefore be one of the earliest detectable triggers of the AD pathology preceding and predisposing to synaptic deficits.

Building on the growing interest in identifying early Aβ targets, we studied the effect of iAβ42 on neuronal excitability in primary hippocampal neurons and in CA1 hippocampal neurons of triple-transgenic AD (3xTg-AD) mouse model that harbor the mutant genes for amyloid precursor protein (APPSwe), presenilin 1PS1M146V and for tauP301L (Oddo et al., 2003) and overexpresses intracellular Aβ42.

We found that Aβ42 internalization is required for inducing increase in intrinsic excitability in hippocampal pyramidal neurons, an effect that is mediated by inhibition of A-type K+ channels (Kv), and requires caspases activation and glycogen synthase kinase 3- (GSK-3) dependent phosphorylation of Kv4.2. Parallel changes in intrinsic excitability, A-type K+ currents, and Kv4.2 phosphorylation are found in CA1 hippocampal neurons derived from brain slices of 3xTg-AD mice.

Section snippets

Animals

A colony of 3xTg-AD (harboring the PS1 M146V, APPSwe KM670/671NL, and tau P301L transgenes) and non-transgenic (B6129SF2/J, named non-Tg) mice were used for electrophysiological experiments in brain slices and for Western blot analysis. The colonies were established in-house from breeding pairs purchased by the Jackson Laboratory. All animal procedures were approved by the Ethics Committee of the Università Cattolica “S. Cuore” and were fully compliant with the Italian and European Union

Intracellular Aβ42 increases neuronal firing in primary mouse hippocampal neurons

To investigate whether iAβ42 affects intrinsic excitability, whole-cell patch-clamp recordings were performed on primary mouse hippocampal neurons in current-clamp mode. Action potential firing was examined by exposing cells to a series of 800 ms current pulses (1 every 5 minutes), whereas recombinant Aβ42 (200 nM) was intracellularly perfused through the patch pipette. The amplitude of current pulses ranged from −50 pA (hyperpolarizing, subthreshold) to 200 pA (depolarizing, suprathreshold) in

Discussion

The present study revealed that intracellularly injected Aβ42 downregulates A-type K+ currents and increases spike width of hippocampal pyramidal neurons, thereby increasing excitability. These effects require caspases and GSK-3 activation and correlate with an increase in phosphorylation of a molecular determinant of A-type K+ currents, the Kv4.2 subunit, at Ser-616, a predicted GSK-3 site. At an early AD stage (5 months old), the 3xTg-AD mouse model exhibits intracellular accumulation of Aβ

Disclosure statement

None of the authors has any actual or potential conflicts of interest to report. All animal procedures were approved by the Ethics Committee of the Catholic University and complied with the Italian Ministry of Health guidelines and with national laws (legislative decree 116/1992) and the European Union guidelines on animal research (No. 86/609/EEC).

Acknowledgements

This work was supported by grants from Università Cattolica (D1-2012 #70200969 to Claudio Grassi and Marcello D’Ascenzo), Alzheimer's Association (NIRG-14-321307; to Cristian Ripoli), the Institute for Translational Sciences at the University of Texas Medical Branch as part of a Clinical and Translational Science Award (UL1TR000071, Fernanda Laezza) from the National Center for Advancing Translational Sciences, National Institutes of Health (NIH NIMH R01 MH095995 to Fernanda Laezza).

References (109)

  • Y. Deng et al.

    β-Amyloid impairs the regulation of N-methyl-D-aspartate receptors by glycogen synthase kinase 3

    Neurobiol. Aging

    (2014)
  • G.K. Gouras et al.

    Intraneuronal Aβ42 accumulation in human brain

    Am. J. Pathol.

    (2000)
  • G.K. Gouras et al.

    Critical role of intraneuronal Aβ in Alzheimer's disease: technical challenges in studying intracellular Aβ

    Life Sci.

    (2012)
  • C. Grassi et al.

    Modulation of Ca(v)1 and Ca(v)2.2 channels induced by nitric oxide via cGMP-dependent protein kinase

    Neurochem. Int.

    (2004)
  • H.J. Hu et al.

    The Kv4.2 potassium channel subunit is required for pain plasticity

    Neuron

    (2006)
  • R.S. Jope et al.

    The glamour and gloom of glycogen synthase kinase-3

    Trends. Biochem. Sci.

    (2004)
  • J. Kim et al.

    The role of apolipoprotein E in Alzheimer's disease

    Neuron

    (2009)
  • M.A. Kuszczyk et al.

    Blocking the interaction between apolipoprotein E and Aβ reduces intraneuronal accumulation of Aβ and inhibits synaptic degeneration

    Am. J. Pathol.

    (2013)
  • N. Louneva et al.

    Caspase-3 is enriched in postsynaptic densities and increased in Alzheimer's

    Am. J. Pathol.

    (2008)
  • S. Oddo et al.

    Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Aβ and synaptic dysfunction

    Neuron

    (2003)
  • J.J. Palop et al.

    Aberrant excitatory neuronal activity and compensatory remodeling of inhibitory hippocampal circuits in mouse models of Alzheimer's disease

    Neuron

    (2007)
  • A.D. Randall et al.

    The functional neurophysiology of the amyloid precursor protein (APP) processing pathway

    Neuropharmacology

    (2010)
  • C. Ripoli et al.

    Effects of different amyloid β-protein analogues on synaptic function

    Neurobiol. Aging

    (2013)
  • D. Tampellini et al.

    Internalized antibodies to the Aβ domain of APP reduce neuronal Aβ and protect against synaptic alterations

    J. Biol. Chem.

    (2007)
  • G. Thinakaran et al.

    Amyloid precursor protein trafficking, processing, and function

    J. Biol. Chem.

    (2008)
  • J.P. Adams et al.

    The A-type potassium channel Kv4.2 is a substrate for the mitogen-activated protein kinase ERK

    J. Neurochem.

    (2000)
  • J.W. Allen et al.

    Multiple caspases are involved in β-amyloid-induced neuronal apoptosis

    J. Neurosci. Res.

    (2001)
  • J.C. Amatniek et al.

    Incidence and predictors of seizures in patients with Alzheimer's disease

    Epilepsia

    (2006)
  • A. Attar et al.

    Protection of primary neurons and mouse brain from Alzheimer's pathology by molecular tweezers

    Brain

    (2012)
  • R.P. Bonin et al.

    Alpha5GABAA receptors regulatethe intrinsic excitability of mouse hippocampal pyramidal neurons

    J. Neurophysiol.

    (2007)
  • H.A. Born et al.

    Genetic suppression of transgenic APP rescues hypersynchronous network activity in a mouse model of Alzheimer's disease

    J. Neurosci.

    (2014)
  • C.A. Bradley et al.

    A pivotal role of GSK-3 in synaptic plasticity

    Front. Mol. Neurosci.

    (2012)
  • J.T. Brown et al.

    Altered intrinsic neuronal excitability and reduced Na+ currents in a mouse model of Alzheimer's disease

    Neurobiol. Aging

    (2011)
  • M.A. Busche et al.

    Critical role of soluble amyloid-β for early hippocampal hyperactivity in a mouse model of Alzheimer's disease

    Proc. Natl. Acad. Sci. U. S. A.

    (2012)
  • Y. Carrasquillo et al.

    A-type K+ channels encoded by Kv4.2, Kv4.3 and Kv1.4 differentially regulate intrinsic excitability of cortical pyramidal neurons

    J. Physiol.

    (2012)
  • M. Citron

    Alzheimer's disease: strategies for disease modification

    Nat. Rev. Drug Discov.

    (2010)
  • W.A. Coetzee et al.

    Molecular diversity of K+ channels

    Ann. N. Y. Acad. Sci.

    (1999)
  • L. Curcio et al.

    Reduced D-serine levels in the nucleus accumbens of cocaine-treated rats hinder the induction of NMDA receptor-dependent synaptic plasticity

    Brain

    (2013)
  • M. D'Ascenzo et al.

    Activation of mGluR5 induces spike after depolarization and enhanced excitability in medium spiny neurons of the nucleus accumbens by modulating persistent Na+ currents

    J. Physiol.

    (2009)
  • K.E. Davis et al.

    Increased hippocampal excitability in the 3xTgAD mouse model for Alzheimer's disease in vivo

    PLoS One

    (2014)
  • E.S. Emamian et al.

    Convergent evidence for impaired AKT1-GSK-3β signaling in schizophrenia

    Nat. Genet.

    (2004)
  • S. Frame et al.

    GSK3 takes centre stage more than 20 years after its discovery

    Biochem. J.

    (2001)
  • T.A. Good et al.

    Effect of β-amyloid block of the fast-inactivating K+ channel on intracellular Ca2+ and excitability in a modeled neuron

    Proc. Natl. Acad. Sci. U. S. A.

    (1996)
  • G.A. Gutman et al.

    International Union of Pharmacology. XLI. Compendium of voltage-gated ion channels: potassium channels

    Pharmacol. Rev.

    (2003)
  • E.A. Guzmán et al.

    Abundance of Aβ5-x like immunoreactivity in transgenic 5XFAD, APP/PS1KI and 3xTG mice, sporadic and familial Alzheimer's disease

    Mol. Neurodegener.

    (2014)
  • K.H. Gylys et al.

    Apolipoprotein E enhances uptake of soluble but not aggregated amyloid-β protein into synaptic terminals

    J. Neurochem.

    (2003)
  • J. Hardy et al.

    The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics

    Science

    (2002)
  • M.T. Heneka et al.

    NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice

    Nature

    (2013)
  • D.A. Hoffman et al.

    Downregulation of transient K+ channels in dendrites of hippocampal CA1 pyramidal neurons by activation of PKA and PKC

    J. Neurosci.

    (1998)
  • C. Hommet et al.

    Epilepsy and dementia in the elderly

    Dement. Geriatr. Cogn. Disord.

    (2008)
  • Cited by (73)

    • Theta and gamma oscillatory dynamics in mouse models of Alzheimer's disease: A path to prospective therapeutic intervention

      2022, Neuroscience and Biobehavioral Reviews
      Citation Excerpt :

      Further, an increase in theta power is one of the earliest alterations to be noticed in AD pathology (Bennys et al., 2001; Coben et al., 1990; Czigler et al., 2008; Jeong, 2004; Moretti et al., 2010; Musaeus et al., 2018). Although the aggregation of Aβ and the resulting synaptic dysfunction have been reported to cause hippocampal hyperexcitability (Kazim et al., 2021; Scala et al., 2015), alterations to theta and gamma rhythm were observed prior to the detection of any robust Aβ plaque in several AD mouse models (Goutagny et al., 2013; Hamm et al., 2017; Kazim et al., 2017; Mahar et al., 2017; Mondragón-Rodríguez et al., 2018; Wang et al., 2020). Perturbed oscillations seen in association with tau pathology also show similar patterns of attenuated theta and gamma rhythm (Ahnaou et al., 2017, 2019, 2020; Booth et al., 2016; Tanninen et al., 2017).

    View all citing articles on Scopus
    View full text