Modulation of the Ca2+ conductance of nicotinic acetylcholine receptors by Lypd6

https://doi.org/10.1016/j.euroneuro.2009.03.007Get rights and content

Abstract

The agonist binding sensitivity and desensitization kinetics of nicotinic acetylcholine receptors (nAChRs) can be modulated by snake venom neurotoxins and related endogenous small proteins of the uPAR-Ly6 family. Here we identify Lypd6, a distantly related member of the u-PAR/Ly-6 family expressed in neurons as a novel modulator of nAChRs. Lypd6 overexpressed in trigeminal ganglia neurons selectively enhanced the Ca2+-component of nicotine-evoked currents through nAChRs, as evidenced by comparative whole-cell patch clamp recordings and Ca2+-imaging in wildtype and transgenic mice overexpressing Lypd6. In contrast, a knockdown of Lypd6 expression using siRNAs selectively reduced nicotine-evoked Ca2+-currents. Pharmacological experiments revealed that the nAChRs involved in this process are heteromers. Transgenic mice displayed behaviors that were indicative of an enhanced cholinergic tone, such as a higher locomotor arousal, increased prepulse-inhibition and hypoalgesia. These mice overexpressing Lypd6 mice were also more sensitive to the analgesic effects of nicotine. Transgenic mice expressing siRNAs directed against Lypd6 were unable to procreate, thus indicating a vital role for this protein. Taken together, Lypd6 seems to constitute a novel modulator of nAChRs that affects receptor function by selectively increasing Ca2+-influx through this ion channels.

Introduction

Nicotinic acetylcholine receptors (nAChRs) belong to the “Cys-loop superfamily” of ligand-gated ion channels that are gated by the neurotransmitter acetylcholine, as well as nicotine, the major psychoactive component in tobacco. They have an important role in various functions of the central nervous system (CNS) including memory, locomotion, sensorimotor gating and pain sensation (Dani, 2001, Everitt and Robbins, 1997, Picciotto et al., 2000, Tassonyi et al., 2002). Based upon their expression and subunit composition, nAChRs can have quite different effects on neurotransmission. Presynaptic and preterminal nAChRs enhance neurotransmitter release (Lena et al., 1993, McGehee et al., 1995), postsynaptic nAChRs convey fast excitatory transmission (Hefft et al., 1999, Roerig et al., 1997) and nonsynaptic nAChRs have effects on many neurotransmitter systems by modulating neuronal excitability (Descarries et al., 1997, Vizi, 2000). In these processes influx of Ca2+ through nAChR-channels plays an important functional role (Dani, 2001, Vizi, 2000).

Nicotinic acetylcholine receptors represent a prototype of allosteric proteins. In addition to modulation through allosteric effectors such as divalent cations, natural steroid hormones (Curtis et al., 2002), drugs like 5-hydroxyindole (Zwart et al., 2002), PNU-120596 (Hurst et al., 2005) or ivermectin (Krause et al., 1998), several endogenous small proteins with homology to alpha-neurotoxins have been identified that bind nAChRs and modulate their activity (Hogg et al., 2005).

On neurons, the GPI-anchored protein Lynx1 alters current amplitudes and desensitization kinetics of α4/β2-nAChRs (Ibanez-Tallon et al., 2002, Miwa et al., 1999). Genetic deletion of Lynx1 in mice resulted in an increased sensitivity of nAChRs to nicotine and a prolonged nAChR receptor activation (Miwa et al., 2006). In the epidermis, the secreted proteins Slurp1 and Slurp2 regulate keratinocyte nAChRs (Adermann et al., 1999, Arredondo et al., 2006, Arredondo et al., 2005). Mutations in the human SLURP1 gene cause Mal de Meleda, an autosomal recessive inflammatory and palmoplantar skin disorder (Chimienti et al., 2003). Lynx1, Slurp1 and Slurp2 belong to the murine u-PAR/Ly-6 gene family, which is evolutionary related to the elapid snake venom neurotoxins (Fleming et al., 1993, Gumley et al., 1995). Most other members of the u-PAR/Ly-6 gene family code for GPI-linked cell accessory proteins of the immune system (Gumley et al., 1995, Hanninen et al., 1997, Mallya et al., 2006).

In this study we report the identification of a novel member of the u-PAR/Ly6 gene family, Lypd6 (LY6/PLAUR domain containing 6 (Ploug and Ellis, 1994)), which modulates nicotinic receptor signaling in vivo. Sequence analysis of Lypd6 and other members of the snake venom neurotoxin superfamily support a direct evolutionary relationship between Lypd6 and well-known endogenous modulators of cholinergic transmission like Lynx1, Slurp1 and Slurp2. Lypd6 is highly expressed in the brain and spinal cord. Lypd6-deficient and overexpressing transgenic mice reveal that Lypd6 acts as a modulator of nAChRs and is also essential for embryo development.

Section snippets

Generation of siRNA knockdown transgenic mice

To generate Lypd6 RNAi-inducing small hairpin RNA, we cloned 21 bp of the mouse Lypd6 coding region (5’-GGGAACAGCAUCUCUGUCAUU-3’) into the pLL3.7 vector (Rubinson et al., 2003) as an inverted repeat with a 9-bp spacer. The pLL3.7 vector contains a U6 promoter and a floxed, CMV promoter driven EGFP reporter gene. Six FVB/N transgenic founders were bred with FVB/N wild-type mice.

Generation of synapsin-Lypd6 transgenic mice

The Lypd6 ORF was fused to the amino-terminus of the HA epitope (YPYDVPDYA). The expression construct used the rat

Identification of Lypd6 as a member of the uPAR/Ly-6 gene family

We have recently screened the mouse transcriptome for secreted small proteins with a putative neuromodulatory function using a computational approach (Gustincich et al., 2003, Okazaki et al., 2002). This led to the identification of 90 novel hypothetical neuromodulators, of which we now selected 39 candidates that had (i) a predicted open reading frame of 100–200 amino acids, (ii) a gene structure with intron-exon boundaries and (iii) additional support from ESTs. When we established expression

Discussion

The experiments we have presented in this paper identify Lypd6 as a member of the uPAR/Ly-6 gene family that is highly enriched in the CNS and acts as a novel modulator of nAChRs in mice. As a consequence of this peak amplitudes of nicotine-induced calcium currents through nAChR-channels are significantly enhanced in trigeminal ganglial neurons (TrigGs) of Lypd6 overexpressing animals and much reduced in neurons with a siRNA-induced knock-down of Lypd6 expression.

Lypd6 contains a signaling

Role of the funding source

This work was supported by grants from the NIH (RO1, DA016768), the EU (Framework VI, PL005166) and the BMBF (NGFN2). The NIH and the EU had no further role in study design; in the collection, analysis and interpretation of data; in the writing of the report; and in the decision to submit the paper for publication.

Contributors

Authors MD,MM, SA, SD and AZ designed the study and performed the analyses. Authors MD, MM, IR and SA performed the experiments and undertook the statistical analysis. Author AZ wrote the first draft of the manuscript. All authors contributed to and have approved the final manuscript.

Conflict of interest

The authors declare no conflict of interest.

Acknowledgements

We thank Dr. David Otte for his advice and help, and Charlotte Schick und Jürgen Schmidt for the generation of transgenic mice.

References (53)

  • Ibanez-TallonI. et al.

    Tethering naturally occurring peptide toxins for cell-autonomous modulation of ion channels and receptors in vivo

    Neuron

    (2004)
  • LivakK.J. et al.

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

    Methods

    (2001)
  • MaelickeA. et al.

    Allosteric sensitization of nicotinic receptors by galantamine, a new treatment strategy for Alzheimer's disease

    Biol. Psychiatry

    (2001)
  • MiwaJ.M. et al.

    lynx1, an endogenous toxin-like modulator of nicotinic acetylcholine receptors in the mammalian CNS

    Neuron

    (1999)
  • MiwaJ.M. et al.

    The prototoxin lynx1 acts on nicotinic acetylcholine receptors to balance neuronal activity and survival in vivo

    Neuron

    (2006)
  • PicciottoM.R. et al.

    Nicotinic receptors in the brain. Links between molecular biology and behavior

    Neuropsychopharmacology

    (2000)
  • PlougM. et al.

    Structure-function relationships in the receptor for urokinase-type plasminogen activator. Comparison to other members of the Ly-6 family and snake venom alpha-neurotoxins

    FEBS Lett.

    (1994)
  • StevensK.E. et al.

    Normalizing effects of nicotine and a novel nicotinic agonist on hippocampal auditory gating in two animal models

    Pharmacol. Biochem. Behav.

    (1997)
  • TassonyiE. et al.

    The role of nicotinic acetylcholine receptors in the mechanisms of anesthesia

    Brain Res. Bull.

    (2002)
  • VinclerM.A. et al.

    Knock down of the alpha 5 nicotinic acetylcholine receptor in spinal nerve-ligated rats alleviates mechanical allodynia

    Pharmacol. Biochem. Behav.

    (2005)
  • ZwartR. et al.

    5-Hydroxyindole potentiates human alpha 7 nicotinic receptor-mediated responses and enhances acetylcholine-induced glutamate release in cerebellar slices

    Neuropharmacology

    (2002)
  • AdermannK. et al.

    Structural and phylogenetic characterization of human SLURP-1, the first secreted mammalian member of the Ly-6/uPAR protein superfamily

    Protein Sci.

    (1999)
  • ArredondoJ. et al.

    SLURP-2: A novel cholinergic signaling peptide in human mucocutaneous epithelium

    J. Cell. Physiol.

    (2006)
  • Bilkei-GorzoA. et al.

    Behavioral phenotype of pre-proenkephalin-deficient mice on diverse congenic backgrounds

    Psychopharmacology (Berl)

    (2004)
  • ChamptiauxN. et al.

    Knockout and knockin mice to investigate the role of nicotinic receptors in the central nervous system

    Prog. Brain Res.

    (2004)
  • ChimientiF. et al.

    Identification of SLURP-1 as an epidermal neuromodulator explains the clinical phenotype of Mal de Meleda

    Hum. Mol. Genet.

    (2003)
  • Cited by (42)

    • Loss of Lypd6 leads to reduced anxiety-like behaviour and enhanced responses to nicotine

      2018, Progress in Neuro-Psychopharmacology and Biological Psychiatry
      Citation Excerpt :

      Furthermore, Lynx2 is primarily expressed in the basolateral amygdala and in upper cortical layers (Miwa et al., 2012), and in the mouse visual cortex it is almost exclusively expressed in glutamatergic, excitatory neurons. Lypd6 on the contrary is primarily expressed in deep cortical layers, and in the visual cortex it is found mainly in GABAergic, inhibitory interneurons (Darvas et al., 2009; Demars and Morishita, 2014). Consequently, different Lynx proteins might influence distinct neural pathways related to anxiety, depending on their individual expression patterns.

    View all citing articles on Scopus
    1

    The first two authors contributed equally to this work.

    View full text