Potassium channels in the neuronal homeostasis and neurodegenerative pathways underlying Alzheimer’s disease: An update

https://doi.org/10.1016/j.mad.2019.111197Get rights and content

Highlights

  • Potassium (K+) channels are key regulators of cell excitability in several tissues.

  • A dysregulation of K+ channels is associated with Alzheimer’s disease (AD).

  • The role of K+ channels in AD is not completely elucidated.

  • Understanding their functions will be helpful for clarifying AD pathogenesis.

  • Modulating K+ channel expression should be useful for developing novel drugs.

Abstract

With more than 80 subunits, potassium (K+) channels represent a group of ion channels showing high degree of diversity and ubiquity. They play important role in the control of membrane depolarization and cell excitability in several tissues, including the brain. Controlling the intracellular and extracellular K+ flow in cells, they also modulate the hormone and neurotransmitter release, apoptosis and cell proliferation. It is therefore not surprising that an improper functioning of K+ channels in neurons has been associated with pathophysiology of a wide range of neurological disorders, especially Alzheimer’s disease (AD).

This review aims to give a comprehensive overview of the basic properties and pathophysiological functions of the main classes of K+ channels in the context of disease processes, also discussing the progress, challenges and opportunities to develop drugs targeting these channels as potential pharmacological approach for AD treatment.

Introduction

Alzheimer’s disease (AD) is an irreversible progressive neurodegenerative disorder with a complex etiology and represents the most prevalent form of dementia in elderly individuals (Crous-Bou et al., 2017). The major risk factor for AD is represented by aging, the inevitable degenerative physiological process which results in a progressive functional and structural decline, from the cellular level to the whole body, causing a reduced ability to adapt to environmental changes and stressors as well as the accumulation of misfolded proteins (Mecocci et al., 2018). AD has been characterized by the neuropathological hallmarks of extracellular senile plaques constituted by the amyloid-β (Aβ) peptide and intracellular paired helical filaments of hyper-phosphorylated microtubule-associated protein tau (Huang and Mucke, 2012). Aβ plaques are generated by sequential proteolytic cleavage of Aβ precursor protein (APP) by β-site APP-cleaving enzyme 1 (BACE1) and the γ-secretase complex. The extracellular deposition of Aβ in the brain could trigger a cascade of pathological events, including microglia-mediated inflammation, mitochondrial dysfunctions, excess in mitochondrial reactive oxygen species (ROS) generation and increased oxidative stress, which may represent causative factors of dementia and cell death (Tönnies and Trushina, 2017). Dysfunction of neurotransmission, brain networks and loss of synapses are also involved in AD pathogenesis (Reddy, 2017). In spite of its prevalence, the etiology of the disease remains to be fully elucidated and there are no effective therapeutic strategies to cure AD or inhibit the progression of its symptoms (Mangialasche et al., 2010).

Some evidence reported an emerging role of altered neuronal excitability, finely controlled by different ion channels and their associated proteins, occurring early during AD pathogenesis (Palop et al., 2007; Frazzini et al., 2016). Ion channels are membrane-spanning proteins forming selective pores for sodium (Na+), potassium (K+), calcium (Ca2+) or chloride (Cl) ions (Zaydman et al., 2012). Among them, K+ channels are the most numerous and diverse channels present in the mammalian brain. Controlling the intracellular and extracellular flow of K+ ions in different cells (e.g., neurons, glia and lymphocytes), they play crucial role as determinant of membrane excitability, setting resting membrane potentials and driving repolarization, becoming target drugs for the treatment of neurodegenerative disorders (Miller, 2000). K+ channels might be divided into five structural types depending on the activation modes and the number of transmembrane (TM) segments: 6-TM K+ channels (KV) with voltage gates, 2-TM K+ channel (Kir), 2-pore 4-TM K+ channels (K2P), Ca2+-activated 6-TM or 7-TM K+ channels (KCa) and the hyperpolarization-activated cyclic nucleotide-gated (HCN). It appears that K+ channels are naturally subjected to oxidation by ROS in both aging and neurodegenerative conditions characterized by high levels of ROS (Sesti, 2016). A dysfunction of K+ channels has been observed in fibroblasts (Etcheberrigaray et al., 1993) and platelets (de Silva et al., 1998) of AD patients. Post-mortem studies have also reported an alteration in K+ channels expression in AD brains (Ikeda et al., 1991). Moreover, it has been demonstrated that Aβ is not only involved in the AD pathogenesis, but it is also a modulator of K+ channel activity (Plant et al., 2005) and may have a physiological role in controlling neuronal excitability (Ramsden et al., 2002).

In this review, we report an updated discussion on the most prominent K+ channels involved in the pathophysiology of AD, that are summarized in Table 1.

Section snippets

Voltage-gated K+ (KV) channels

The KV channels represent the largest gene family of K+ channels, which are widely expressed in both the central and the peripheral nervous system where they participate in a wide range of biological processes (e.g., synaptic plasticity, regulation of apoptosis, modulation of the neurotransmitter release and the duration of action potentials). In humans, they are encoded by forty different genes and subgrouped into twelve subfamilies ranging from KV1 to KV12. KV channels have a tetrameric

Inwardly rectifying potassium (Kir) channels

The Kir channels are important for maintaining the resting membrane potential and are largely expressed in cells from both excitable and non-excitable tissues, playing a key role in controlling cell excitability. According to their sequence similarity and function properties, they are grouped into seven different families (Kir1-Kir7) and are a tetramer of subunits, each with two TM domains and a pore-lining loop. Under physiological conditions, these channels generate larger inward currents at

Calcium-activated (KCa) channels

The KCa channels comprises many members which are subdivided into three major classes according to their single channel conductance differing in biophysical and pharmacological properties: small (SK, also known as KCNN: KCa2.1, KCa2.2 and KCa2.3), intermediate (IK: KCa3.1) and large conductance (BK: KCa1.1, KCa1.2, and KCa1.3). In the human brain, these channels mediate the afterhyperpolarization that follows a neuronal action potential, playing therefore a critical role in circuit excitability

Cyclic nucleotide-gated (HCN) channels

The HCN channels belong to the super family of poor-loop cation channels. In mammals, the HCN channel family consists of 4 members (HCN1-4) which are expressed in the heart and nervous system. In neurons, they are found in axon terminals and dendrites with a heterogeneous distribution. The HCN channels are permeable to Na+ and K+ ions, activated by the membrane hyperpolarization and constitutively opened at voltages near the resting membrane potential (Benarroch, 2013).

A dysregulation of these

Conclusions

Among the ion channels regulating cell excitability, the K+ channels represent the most diverse and abundant group playing important physiological roles in both excitable and non-excitable cells (Miller, 2000). In the CNS, these channels do not only set the resting membrane potential and regulate nerve impulses, buy they are also crucial for preventing pathological hyperactivity, such as epileptic discharges (Villa and Combi, 2016), cell death following ischemic brain injury or the excessive Ca

Funding

This research did not receive any specific grant from funding agencies in the public, commercial or not-for-profit sectors.

Declaration of Competing Interest

None.

References (107)

  • J. Du et al.

    The K+ channel, Kv2.1, is apposed to astrocytic processes and is associated with inhibitory postsynaptic membranes in hippocampal and cortical principal neurons and inhibitory interneurons

    Neuroscience

    (1998)
  • J. Duran-Gonzalez et al.

    Amyloid beta peptides modify the expression of antioxidant repair enzymes and a potassium channel in the septohippocampal system

    Neurobiol. Aging

    (2013)
  • M.J. Eslamizade et al.

    Alterations in CA1 pyramidal neuronal intrinsic excitability mediated by Ih channel currents in a rat model of amyloid beta pathology

    Neuroscience

    (2015)
  • M.H. Esmaeili et al.

    ATP-sensitive potassium-channel inhibitor glibenclamide attenuates HPA axis hyperactivity, depression- and anxiety-related symptoms in a rat model of Alzheimer’s disease

    Brain Res. Bull.

    (2018)
  • S. Frykman et al.

    Maturation and processing of the amyloid precursor protein is regulated by the potassium/sodium hyperpolarization-activated cyclic nucleotide-gated ion channel 2 (HCN2)

    Biochem. Biophys. Res. Commun.

    (2017)
  • C.M. Griffith et al.

    Aberrant expression of the pore-forming KATP channel subunit Kir6.2 in hippocampal reactive astrocytes in the 3xTg-AD mouse model and human Alzheimer’s disease

    Neuroscience

    (2016)
  • M. Guéguinou et al.

    KCa and Ca2+ channels: the complex thought

    Biochim. Biophys. Acta

    (2014)
  • Y. Huang et al.

    Alzheimer mechanisms and therapeutic strategies

    Cell

    (2012)
  • M. Ikeda et al.

    Selective reduction of [125I] apamin binding sites in Alzheimer hippocampus: a quantitative autoradiographic study

    Brain Res.

    (1991)
  • A. Jafari et al.

    Brain mitochondrial ATP-insensitive large conductance Ca+2 activated K+ channel properties are altered in a rat model of amyloid-beta neurotoxicity

    Exp. Neurol.

    (2015)
  • H. Li et al.

    The discovery of fused oxadiazepines as gamma secretase modulators for treatment of Alzheimer’s disease

    Bioorg. Med. Chem. Lett.

    (2013)
  • I. Maezawa et al.

    Amyloid-beta protein oligomer at low nanomolar concentrations activates microglia and induces microglial neurotoxicity

    J. Biol. Chem.

    (2011)
  • F. Mangialasche et al.

    Alzheimer’s disease: clinical trials and drug development

    Lancet Neurol.

    (2010)
  • J.L. Methot et al.

    Potent benzoazepinone γ-secretase modulators with improved bioavailability

    Bioorg. Med. Chem. Lett.

    (2015)
  • Y. Noam et al.

    Towards an integrated view of HCN channel role in epilepsy

    Curr. Opin. Neurobiol.

    (2011)
  • 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)
  • C. Poulopoulou et al.

    Aberrant modulation of a delayed rectifier potassium channel by glutamate in Alzheimer’s disease

    Neurobiol. Dis.

    (2010)
  • M. Ramsden et al.

    Modulation of Ca2+ channel currents in primary cultures of rat cortical neurones by amyloid β protein (1–40) is dependent on solubility status

    Brain Res.

    (2002)
  • P.H. Reddy

    Amyloid beta, mitochondrial structural and functional dynamics in Alzheimer’s disease

    Exp. Neurol.

    (2009)
  • F. Wang et al.

    Improvement of spatial learning by facilitating large-conductance calcium-activated potassium channel with transcranial magnetic stimulation in Alzheimer’s disease model mice

    Neuropharmacology

    (2015)
  • L. Wang et al.

    Cognitive recovery by chronic activation of the large-conductance calcium-activated potassium channel in a mouse model of Alzheimer’s disease

    Neuropharmacology

    (2015)
  • D.M. Wilcock et al.

    Vascular amyloid alters astrocytic water and potassium channels in mouse models and humans with Alzheimer’s disease

    Neuroscience

    (2009)
  • X. Wu et al.

    Molecular mechanisms underlying the apoptotic effect of KCNB1 K+ channel oxidation

    J. Biol. Chem.

    (2013)
  • X.H. Xu et al.

    mRNA expression alterations of inward rectifier potassium channels in rat brain with cholinergic impairment

    Neurosci. Lett.

    (2002)
  • Alzheimer’s Association Update

    Alzheimers Dement.

    (2016)
  • E. Angulo et al.

    Up-regulation of the Kv3.4 potassium channel subunit in early stages of Alzheimer’s disease

    J. Neurochem.

    (2004)
  • E.E. Benarroch

    HCN channels: function and clinical implications

    Neurology

    (2013)
  • G.A. Bezerra et al.

    Structure of human dipeptidyl peptidase 10 (DPPY): a modulator of neuronal Kv4 channels

    Sci. Rep.

    (2015)
  • A. Binda et al.

    A novel KCNJ2 mutation identified in an autistic proband affects the single channel properties of Kir2.1

    Front. Cell. Neurosci.

    (2018)
  • E. Boda et al.

    Brain expression of Kv3 subunits during development, adulthood and aging and in a murine model of Alzheimer’s disease

    J. Mol. Neurosci.

    (2012)
  • B.M. Brown et al.

    KCa3.1 channel modulators as potential therapeutic compounds for glioblastoma

    Curr. Neuropharmacol.

    (2018)
  • D.A. Brown et al.

    Neural KCNQ (Kv7) channels

    Br. J. Pharmacol.

    (2009)
  • P. Campolongo et al.

    Systemic administration of substance P recovers β amyloid-induced cognitive deficits in rat: involvement of Kv potassium channels

    PLoS One

    (2013)
  • S. Chakroborty et al.

    Early presynaptic and postsynaptic calcium signaling abnormalities mask underlying synaptic depression in presymptomatic alzheimer’s disease mice

    J. Neurosci.

    (2012)
  • T. Chen et al.

    Molecular characterisation of a novel dipeptidyl peptidase like protein: its pathological link to Alzheimers disease

    Clin. Chem. Lab. Med.

    (2008)
  • T. Chen et al.

    Dipeptidyl peptidase 10 (DPP10(789)): a voltage gated potassium channel associated protein is abnormally expressed in Alzheimer’s and other neurodegenerative diseases

    Biomed Res. Int.

    (2014)
  • D. Cotella et al.

    Toxic role of K+ channel oxidation in mammalian brain

    J. Neurosci.

    (2012)
  • M. Crous-Bou et al.

    Alzheimer’s disease prevention: from risk factors to early intervention

    Alzheimers Res. Ther.

    (2017)
  • J. Cummings et al.

    Clinical trials for disease-modifying therapies in alzheimer’s disease: a primer, lessons learned, and a blueprint for the future

    J. Alzheimers Dis.

    (2018)
  • A. Demuro et al.

    Single-channel Ca2+ imaging implicates Aβ1-42 amyloid pores in Alzheimer’s disease pathology

    J. Cell Biol.

    (2011)
  • Cited by (20)

    • Alzheimer's disease/dementia-associated brain pathology in aging DPP6-KO mice

      2022, Neurobiology of Disease
      Citation Excerpt :

      The key to the connection between DPP6 and its associated potassium channels and AD is likely in the synapse. There is an enormous body of literature linking potassium channels with AD (Villa et al., 2020). For example, amyloid beta can modulate various kinds of potassium channels in the hippocampus (Mayordomo-Cava et al., 2015).

    • Preliminary study on impacts of polystyrene microplastics on the hematological system and gene expression in bone marrow cells of mice

      2021, Ecotoxicology and Environmental Safety
      Citation Excerpt :

      When genes encoding ion channel subunits are mutated or expressed abnormally, their functions can be weakened or enhanced, leading to disorders of the physiological functions. In addition, certain diseases or drugs can also cause changes in the number, function, and even structure of one or several ion channels, such as Alzheimer's disease (AD) (Villa et al., 2020), and cerebral ischemia (Woo et al., 2020). In this study, Kcnt2/Kcns3, two channel complex genes, were significantly upregulated after exposure to a 0.1 mg dose of 5 µm PS-MPs.

    • A novel role of cardiac inwardly rectifying potassium channels explaining autonomic cardiovascular dysfunctions in a cuprizone-induced mouse model of multiple sclerosis

      2020, Autonomic Neuroscience: Basic and Clinical
      Citation Excerpt :

      Abnormal cardiac repolarization evaluated by electrocardiography recording (ECG) occurs during the disease course, but the underlying physiology still remains unknown (Mikkola et al., 2017). Recent studies reported that a dysfunction of ion channels has been associated with pathophysiology of a wide range of neurological disorders (Akyuz et al., 2018; Binda et al., 2018; Noebels, 2017; Villa and Combi, 2016; Villa et al., 2020). Among them, the inwardly rectifying potassium (K+) channels (Kir) play a key role in the maintenance of the resting membrane potential and subsequently in the regulation of cell excitability by the controlling of the intracellular and extracellular flow of K+ ions in different cells (Hibino et al., 2010).

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text