Elsevier

Brain Research Bulletin

Volume 160, July 2020, Pages 107-120
Brain Research Bulletin

Epilepsy and Alzheimer’s Disease: Potential mechanisms for an association

https://doi.org/10.1016/j.brainresbull.2020.04.009Get rights and content

Highlights

  • AD patients have increased risk for seizures, particularly in familial forms of AD.

  • Epilepsy may, in turn, lead to or exacerbate AD pathology and clinical symptoms.

  • Seizures are difficult to identity in AD: clinical studies have several limitations.

  • Subcortical structures (cholinergic nuclei and Locus Coeruleus) may be involved.

  • At cellular level, Glutamate/GABA imbalance, channels and inflammation play a role.

Abstract

Alzheimer’s Disease (AD) and epilepsy are common neurological diseases. The prevalence of epilepsy in AD patients is higher than in healthy subjects, but identifying the reasons for this association, the characteristics of seizures in AD, and the implications for prognosis and treatment is challenging. The present review provides first of all an overview of the main clinical aspects of AD and epilepsy, of their reciprocal relationship, and of the challenges that identifying seizures in AD patients presents. Limitations of clinical studies addressing this topic are discussed, including their mostly prospective nature and possible selection biases. A comprehensive, mechanistic discussion on the factors that are most likely to underlie the increased risk for seizures in AD follows. These include, for instance, GABAergic and glutamatergic alterations, Aβ and Tau protein, the role of the noradrenergic nucleus Locus Coeruleus, and neuroinflammation. Finally, evidence concerning the role that epilepsy may have in exacerbating or initiating AD is reviewed. A mechanistic insight on the relationship between epilepsy and AD might have relevant implications for improving the treatment of AD patients, as well as in elucidating pathophysiological mechanisms.

Introduction

There is evidence that epilepsy occurs more frequently in Alzheimer's Disease (AD) patients than in the general population. Surprisingly, the first account of senile plaques - which are considered a hallmark of AD (Braak et al., 2011; Braak and Braak, 1991) - dates to fifteen years before the first report of a case of AD, and such plaques were first described in epileptic patients (reviewed in (Buda et al., 2009)). Remarkably, Alois Alzheimer himself (who is considered the discoverer of AD) recognized an increased prevalence of epilepsy among his patients with AD (revised in (Vossel et al., 2017)), and ever since the connection between these two pathologies has been puzzling researchers and clinicians. Early clinical studies aimed at specifically assessing the relationship between AD and epilepsy date back to the ‘50s (Letemendia and Pampiglione, 1958; Sjogren et al., 1952). Up to the ‘90s, the occurrence of seizures were described in more advanced stages of AD (Raudino, 2016). In the last decades, the concept of Mild Cognitive Impairment (MCI) has been introduced to describe subtle cognitive alterations preceding frank dementia and, in the case of MCI due to AD, sharing with AD dementia the same pathological mechanisms (Dubois et al., 2014). Also among MCI, it is now known that seizures and subclinical epileptiform activity are more frequent than in age-matched cognitively healthy controls (Sherzai et al., 2014).

In developed countries, the prevalence of both epilepsy and AD increases with age (Banerjee et al., 2009; Cloyd et al., 2006). Also for this reason, one might argue that an increased incidence of seizures in AD might be just due to the advanced age of AD patients. However, several reports have shown a higher incidence of seizures among AD patients, even though with major discrepancies. In particular, the prevalence of seizures in AD ranges from 3.5% to 64 %–64 %, according to the population studied and the type of monitoring used (Friedman et al., 2012). In line with this, several experimental studies have confirmed a reduced threshold to seizures in AD models (Palop et al., 2007; Palop and Mucke, 2010). More intriguingly, recent experimental data even suggest that hyperexcitability itself may play a role in promoting cognitive decline and increase of AD neuropathological burden and cognitive decline, as will be extensively discussed below.

Among AD subtypes, a particularly seizure-prone population might be represented by patients affected by Early Onset Alzheimer Disease (EOAD). In fact, in EOAD patients, myoclonus and seizures rates have been found to be as high as 30 % (Raudino, 2016; Vossel et al., 2017). EOAD is typically caused by abnormalities in presenilin (PS) 1, 2 or APP (Amyloid Precursor Protein) genes, and this supports a key role of amyloid in seizure susceptibility.

Thus, one might hypothesize that a self-perpetuating cycle occurs, in which AD pathology predisposes to seizures (Horváth et al., 2016), which in turn might concur to and exacerbate AD pathology (Vossel et al., 2017).

Section snippets

Alzheimer’s Disease

Dementia, an acquired cognitive decline that hampers activities of daily life, is the most common neurologic disease in elderly people, among whom it is mainly caused by AD (Hauser, 2013). However, AD can occur also earlier in life, especially considering genetic forms such as EOAD (Hauser, 2013), see above). Importantly, AD prevalence is higher in women, who have an higher lifetime risk of developing the disease (Nebel et al., 2018; Viña and Lloret, 2010) and are more severely cognitively

Clinical evidence for co-existence of AD and seizures

As described in paragraph 1, the relationship between AD and epilepsy has been intriguing researchers and clinicians for a long time (Vossel et al., 2017). However, studies on this topic are mainly cross-sectional or retrospective, with only a few prospective ones. Already in one of the first retrospective studies (Hauser et al., 1986), a high prevalence not only of seizures but also of myoclonus was identified in AD patients. Recent evidence from retrospective (Beagle et al., 2017; Cheng et

Mechanisms underlying the increased risk of seizures in AD

Various mechanisms connecting AD and epilepsy have been proposed and many elements involved in AD pathogenesis have been found to regulate neuronal excitability (Fig. 1). However, it has to be remembered that AD experimental models are mainly transgenic mice, which, as such, reproduce a condition similar to familiar AD in humans. Further studies are necessary as far as sporadic AD is concerned (Cretin et al., 2017).

How epilepsy may cause AD/neurodegeneration

In the previous paragraphs, the mechanisms through which AD may lead to a pro-epileptogenic state have been discussed. The reverse hypothesis, albeit less studied, is fascinating: how might epilepsy cause or worsen neurodegeneration and AD? In line with this hypothesis, it has been shown in AD transgenic mice that AED-mediated suppression of IED (which occurs early in the course of the disease) strongly improves cognitive deterioration, synaptic dysfunction and hippocampal remodeling (Sanchez

Funding

This research was funded by Italian Ministry of Health Ricerca Corrente (F.F.); and by Italian Ministry of Health Ricerca Finalizzata 2013, project code:# PE2013-02359574 (“In vivo assessment of the role of Locus Coeruleus in the development of Alzheimer's Disease and other types of Dementia”) (F.S.G.).

References (247)

  • J. Cloyd et al.

    Epidemiological and medical aspects of epilepsy in the elderly

    Epilepsy Res.

    (2006)
  • C. Costa et al.

    Epilepsy, amyloid-β, and D1 dopamine receptors: a possible pathogenetic link?

    Neurobiol. Aging

    (2016)
  • C. Costa et al.

    Alzheimer’s disease and late-onset epilepsy of unknown origin: two faces of beta amyloid pathology

    Neurobiol. Aging

    (2019)
  • E. Cumbo et al.

    Levetiracetam, lamotrigine, and phenobarbital in patients with epileptic seizures and Alzheimer’s disease

    Epilepsy Behav.

    (2010)
  • A. Del Felice et al.

    Transient epileptic amnesia mistaken for mild cognitive impairment? A high-density EEG study

    Epilepsy Behav.

    (2014)
  • R.A. Del Vecchio et al.

    Increased seizure threshold and severity in young transgenic CRND8 mice

    Neurosci. Lett.

    (2004)
  • K.T. Dineley et al.

    Beta -Amyloid peptide activates alpha 7 nicotinic acetylcholine receptors expressed in Xenopus oocytes

    J. Biol. Chem.

    (2002)
  • C.M. Dube et al.

    Fever, febrile seizures and epilepsy

    Trends Neurosci.

    (2007)
  • B. Dubois et al.

    Advancing research diagnostic criteria for Alzheimer’s disease: the IWG-2 criteria

    Lancet Neurol.

    (2014)
  • M.J. During et al.

    Extracellular hippocampal glutamate and spontaneous seizure in the conscious human brain

    Lancet

    (1993)
  • G. Ferraro et al.

    Locus coeruleus noradrenaline system and focal penicillin hippocampal epilepsy: neurophysiological study

    Epilepsy Res.

    (1994)
  • F. Fornai et al.

    Region-and neurotransmitter-dependent species and strain differences in DSP–4–induced monoamine depletion in rodents

    Neurodegeneration

    (1996)
  • A.M. García-Cabrero et al.

    Hyperexcitability and epileptic seizures in a model of frontotemporal dementia

    Neurobiol. Dis.

    (2013)
  • F.S. Giorgi et al.

    The role of norepinephrine in epilepsy: from the bench to the bedside

    Neurosci. Biobehav. Rev.

    (2004)
  • F.S. Giorgi et al.

    Activation of brain metabolism and fos during limbic seizures: the role of locus coeruleus

    Neurobiol. Dis.

    (2008)
  • F.S. Giorgi et al.

    The role of Locus Coeruleus in neuroinflammation occurring in Alzheimer’s disease

    Brain Res. Bull.

    (2019)
  • H. Hampel et al.

    Precision medicine and drug development in Alzheimer’s disease: the importance of sexual dimorphism and patient stratification

    Front. Neuroendocrinol.

    (2018)
  • A. Hazra et al.

    Corticothalamic network dysfunction and behavioral deficits in a mouse model of Alzheimer’s disease HHS Public Access

    Neurobiol. Aging

    (2016)
  • C. Helmstaedter et al.

    Epilepsy and cognition–a bidirectional relationship?

    Seizure

    (2017)
  • M.T. Heneka et al.

    Neuroinflammation in Alzheimer’s disease

    Lancet Neurol.

    (2015)
  • H. Hsieh et al.

    AMPAR removal underlies aβ-induced synaptic depression and dendritic spine loss

    Neuron

    (2006)
  • L.M. Ittner et al.

    Dendritic function of tau mediates Amyloid-β toxicity in Alzheimer’s disease mouse models

    Cell

    (2010)
  • D. Jardanhazi-Kurutz et al.

    Distinct adrenergic system changes and neuroinflammation in response to induced locus ceruleus degeneration in APP/PS1 transgenic mice

    Neuroscience

    (2011)
  • A.K. Abbas et al.

    Cellular and Molecular Immunology

    (2017)
  • J.N. Acharya et al.

    Epilepsy in the elderly: special considerations and challenges

    Ann. Indian Acad. Neurol.

    (2014)
  • J. Albrecht et al.

    Mechanisms of excessive extracellular glutamate accumulation in temporal lobe epilepsy

    Neurochem. Res.

    (2017)
  • F. Alyu et al.

    Inflammatory aspects of epileptogenesis: contribution of molecular inflammatory mechanisms

    Acta Neuropsychiatr.

    (2017)
  • J.C. Amatniek et al.

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

    Epilepsia

    (2006)
  • Y. Andrews-Zwilling et al.

    Apolipoprotein E4 causes age- and tau-dependent impairment of GABAergic interneurons, leading to learning and memory deficits in mice

    J. Neurosci.

    (2010)
  • T. Babic et al.

    Convulsions induced by donepezil [7]

    J. Neurol. Neurosurg. Psychiatry

    (1999)
  • J. Baker et al.

    The prevalence and clinical features of epileptic seizures in a memory clinic population

    Seizure

    (2019)
  • J. Baker et al.

    A longitudinal study of epileptic seizures in Alzheimer’s disease

    Front. Neurol.

    (2019)
  • E.C. Ballinger et al.

    Basal forebrain cholinergic circuits and signaling in cognition and cognitive decline

    Neuron

    (2016)
  • S. Balosso et al.

    A novel non-transcriptional pathway mediates the proconvulsive effects of interleukin-1β

    Brain

    (2008)
  • A.J. Beagle et al.

    Relative incidence of seizures and myoclonus in Alzheimer’s disease, dementia with Lewy Bodies, and frontotemporal dementia

    J. Alzheimers Dis.

    (2017)
  • H. Braak et al.

    Neuropathological stageing of Alzheimer-related changes

    Acta Neuropathol.

    (1991)
  • H. Braak et al.

    Stages of the pathologic process in Alzheimer disease: age categories from 1 to 100 years

    J. Neuropathol. Exp. Neurol.

    (2011)
  • L. Broestl et al.

    Ovarian cycle stages modulate Alzheimer-related cognitive and brain network alterations in female mice

    eNeuro

    (2018)
  • O. Buda et al.

    Georges Marinesco and the early research in neuropathology

    Neurology

    (2009)
  • M.A. Busche et al.

    Clusters of hyperactive neurons near amyloid plaques in a mouse model of Alzheimer’s disease

    Science

    (2008)
  • Cited by (41)

    • The effect of valproic acid and furosemide on the regulation of the inflammasome complex (NLRP1 and NLRP3 mRNA) in the brain of epileptic animal model

      2022, Brain Research Bulletin
      Citation Excerpt :

      Epilepsy is a disease of the nervous system that can lead to seizures or psychological injuries. Approximately 1% of people suffer from epilepsy (Giorgi et al., 2020). Epilepsy is a neurological disorder (central nervous system disorder) in which the function of neural cells in the brain is disrupted.

    • On the relationships between epilepsy, sleep, and Alzheimer's disease: A narrative review

      2022, Epilepsy and Behavior
      Citation Excerpt :

      This allows deeper insights into a biomolecular level into the underlying – and potentially shared – pathogenesis, which could open up to potential new treatments. For example, besides network hyperexcitability, neuroinflammation is more and more revealed as playing an important role in the pathogenesis of AD [150] and also of epilepsy [151]. To conclude, detecting neurodegeneration at an earlier stage could not only allow the implementation of personalized preventive measures, but also – bearing in mind possible interactions and side effects – to begin treatment as early as possible, thus slowing disease progression and maintaining cognitive functions and independence for a longer time.

    View all citing articles on Scopus
    1

    FSG and LFS equally contributed to the article.

    View full text