Elsevier

Cell Calcium

Volume 41, Issue 1, January 2007, Pages 77-86
Cell Calcium

Purinergic signalling and intercellular Ca2+ wave propagation in the organ of Corti

https://doi.org/10.1016/j.ceca.2006.05.005Get rights and content

Abstract

Extracellular ATP is a key neuromodulator of visual and auditory sensory epithelia. In the rat cochlea, pharmacological dissection indicates that ATP, acting through a highly sensitive purinergic/IP3-mediated signaling pathway with (little or) no involvement of ryanodine receptors, is the principal paracrine mediator implicated in the propagation of calcium waves through supporting and epithelial cells. Measurement of sensitivity to UTP and other purinergic agonists implicate P2Y2 and P2Y4 as the main P2Y receptor isoforms involved in these responses. Ca2+ waves, elicited under highly reproducible conditions by carefully controlling dose (1 μM) and timing of focal agonist application (0.2 s), extended over radial distance greater than 160 μm from the source, identical to those activated by damaging single outer hair cells. Altogether, these results indicate that intercellular calcium waves are a robust phenomenon that confers a significant ability for cell–cell communication in the mammalian cochlea. Further ongoing research will reveal the roles that such Ca2+ waves play in the inner ear.

Introduction

Extracellular signalling by purine nucleotides has long been associated with sensory systems, where ATP acts as a co-transmitter and/or neuromodulator [1]. Purinergic receptors of the G protein-coupled (P2YR) type, as well as of the ionotropic type (P2XR) [2] are widely distributed on inner ear structures [3], [4].

Currently, seven subtypes of the P2X family (P2X1–7) and eight subtypes of the P2Y family (P2Y1–2, P2Y4, P2Y6 and P2Y11–14) have been cloned and functionally characterised, albeit typically in expression systems rather than native tissue [5]. As non-selective cation channels, P2XRs provide direct routes for Ca2+ entry into the cell cytoplasm, whereas P2YRs either activate phosholipase C (PLC) and release intracellular Ca2+ or affect adenylyl cyclase and alter cAMP levels [6].

In the mammalian cochlea, the organ of Corti comprises a polarized epithelium that contains sensory receptors, the hair cells, in which ATP has been shown to affect Ca2+ homeostasis [7]. Sensory hair cells are surrounded by various types of supporting cells, including Deiters’ [8], [9], [10], Hensen's [11] and pillar cells [12], which all respond to focal application of ATP with a transient elevation of the intracellular free Ca2+ concentration ([Ca2+]i). Cochlear supporting cells and adjacent epithelial (Böttcher's and Claudius’) cells are considered essential constituents of an epithelial network that participates in the regulation of the unique ionic composition of cochlear extracellular fluids [13]. Hensen's and Deiters’ cells, the two main types of supporting cells in the organ of Corti [14], [15], are highly permeable to K+ [16], [17] which is found at unusually high concentration in endolymph, the fluid bathing the apical surface of the organ of Corti [18]. K+ ions flow into the organ of Corti as they are the major component of the hair cell mechanoelectrical transduction current [19], [20], [21], [22]. Purinergic signalling in the cochlea can affect several essential functions, from ion homeostasis to active mechanical amplification by the outer hair cells [23], [24]. Notably, the level of ATP in cochlear fluids increases in response to modest sound over-stimulation [25]. ATP release from marginal cells in the stria vascularis has been proposed to exert a homeostatic regulatory mechanism [26], promoting K+ efflux from scala media and a G protein-mediated reduction of K+ import into the endolymph from stria vascularis [27]. An expected outcome of both of these effects is a reduction in cochlear sensitivity in noisy environments.

Mechanical stimulation of many cell types induces transient rises in [Ca2+]i that can spread from cell to cell in a wave-like pattern by the release of an extracellular nucleotide intermediate [28]. Indeed, nucleotides are frequently released into the extracellular space as a consequence of cell damage and, once in the pericellular milieu, they can activate P2 nucleotide receptors or be rapidly hydrolyzed by ecto-ATPases and ectonucleotidases [29]. Using neonatal organotypic cultures from rat inner ear [30], [31], [32], we have recently shown, in response to sensory hair cell damage, activation of Ca2+ oscillations and ATP-dependent propagation of Ca2+ waves through cochlear supporting and epithelial cells that surround the damaged hair cells. ATP concentrations above about 15 nM, similar to that measured in noise damaged cochlea, reliably elicit Ca2+ oscillations in the same organ preparation [33].

Extracellular ATP possesses all the properties of a bona fide fast-acting intercellular messenger: (a) it is released in a controlled fashion, (b) ligates specific plasma membrane receptors coupled to signal transduction and (c) is quickly degraded to terminate its actions [29]. However, an alternative concept has been proposed for the mechanism of the Ca2+ wave propagation between glial [34] and epithelial cells [35] that, like cochlear supporting cells [36], [37], [38], are highly coupled by gap junctions. In this paradigm, Ca2+ waves are sustained by the intercellular diffusion of second messengers through gap junctions, with subsequent release of Ca2+ from intracellular stores [39]. As Ca2+ may permeate gap junctions [40] but is heavily buffered in the cytosol [41], IP3 has been proposed as a better intercellular messenger in epithelial cells [35], [42], hepatocytes [43] and glial cells [44], [45], [46], [47], [48]. In the retina, both mechanism seem active: Ca2+ wave propagation among astrocytes depends on gap junctional communication while propagation between astrocytes and Müller cells involves purinergic signalling [49].

The contribution of gap junction communication to the propagation of cochlear Ca2+ waves is not explored in this paper and will be dealt with in a separate publication. In this work, we have analyzed the responses to different types of stimuli capable of eliciting Ca2+ waves in the organ of Corti. Experiments were performed in low divalent cation solutions, thus direct contribution from Ca2+ entry through ionotropic P2X receptors can be excluded. Our results implicate P2Y2 and P2Y4 receptors acting via the PLC-IP3 signal transduction pathway with little or no contribution from ryanodine-sensitive Ca2+ stores.

Section snippets

Organ cultures

Cochleas were dissected from postnatal days 1–2 Sprague–Dawley rat pups in ice-cold Hepes-buffered (10 mM, pH 7.2) Hanks’ balanced salt solutions (HBSS, Sigma, Milan, Italy) and placed onto glass coverslips coated with 10 μg ml−1 of CellTak (Becton Dickinson, Milan, Italy), as described [33]. Cultures were incubated in Dulbecco's modified Eagle's medium DMEM/F12 (Invitrogen, Leek, The Netherlands), supplemented with FBS 5% and maintained at 37 °C for 1 day. During experiments, cultures were

Propagation of intercellular Ca2+ waves in the outer sulcus of the organ of Corti

To study Ca2+ wave propagation in the organ of Corti, perfusion was switched to DFM, a commonly used medium (e.g., see Ref. [51]) virtually devoid of divalent ions (see Section 2.1) 2 min prior to stimulation, and then stopped altogether. Cochlear cultures were kept in DFM for the duration of the recording (generally less than 1 min) and were subsequently returned to ECS, a Ca2+-containing Mg2+-free medium, in the interval between repeated stimulations. To exclude run-down effects, we elicited

Discussion

ATP levels in scala media increase during brief periods of acoustic overstimulation [25], reaching concentrations above the activation threshold (∼10 nM) for P2YRs in this tissue [33]. Signal transduction by P2YRs occurs via the classical pathways triggered by most seven-transmembrane-spanning receptors: (i) agonist-bound receptors determine the rate of G protein activation; (ii) heteromeric G proteins transmit the signal to PLC-β by transiting a controlled cycle of GTP binding and hydrolysis;

Acknowledgements

Control experiments in Fig. 3A and C were performed by Fabio Anselmi. This work has been supported by grants to FM from Telethon Italy (GGP05131) and the European commission FP6 Integrated Project EuroHear (LSHG-CT-20054-512063) under the Sixth Research Frame Program of The European Union and by a Royal Society University Research Fellowship to JEG. We thank Tullio Pozzan, Bruce Tempel and Luigia Santella for helpful comments and constructive criciticism.

References (82)

  • G.P. Richardson et al.

    Cochlear cultures as a model system for studying aminoglycoside induced ototoxicity

    Hear. Res.

    (1991)
  • J.E. Gale et al.

    A mechanism for sensing noise damage in the inner ear

    Curr. Biol.

    (2004)
  • T. Kikuchi et al.

    Gap junction systems in the mammalian cochlea

    Brain Res. Brain Res. Rev.

    (2000)
  • N.L. Allbritton et al.

    Localized calcium spikes and propagating calcium waves

    Cell Calcium

    (1993)
  • A. Verkhratsky et al.

    Calcium signalling in glial cells

    Trends Neurosci.

    (1996)
  • K. Chang et al.

    Molecular cloning and functional analysis of a novel P2 nucleotide receptor

    J. Biol. Chem.

    (1995)
  • D. Communi et al.

    Pharmacological characterization of the human P2Y4 receptor

    Eur. J. Pharmacol.

    (1996)
  • D. Communi et al.

    Cloning, functional expression and tissue distribution of the human P2Y6 receptor

    Biochem. Biophys. Res. Commun.

    (1996)
  • K. Ayyanathan et al.

    Cloning and chromosomal localization of the human P2Y1 purinoceptor

    Biochem. Biophys. Res. Commun.

    (1996)
  • D.J. Henderson et al.

    Cloning and characterisation of a bovine P2Y receptor

    Biochem. Biophys. Res. Commun.

    (1995)
  • D. Communi et al.

    Cloning of a human purinergic P2Y receptor coupled to phospholipase C and adenylyl cyclase

    J. Biol. Chem.

    (1997)
  • M.P. Abbracchio et al.

    Purinoceptors: are there families of P2X and P2Y purinoceptors?

    Pharmacol. Ther.

    (1994)
  • M. Marhl et al.

    Mitochondria as an important factor in the maintenance of constant amplitudes of cytosolic calcium oscillations

    Biophys. Chem.

    (1998)
  • U. Kummer et al.

    Switching from simple to complex oscillations in calcium signalling

    Biophys. J.

    (2000)
  • K. Prank et al.

    Coding efficiency and information rates in transmembrane signaling

    Biosystems

    (2000)
  • A. Atri et al.

    A single-pool model for intracellular calcium oscillations and waves in the Xenopus laevis oocyte

    Biophys. J.

    (1993)
  • G. Dupont et al.

    Signal-induced Ca2+ oscillations: properties of a model based on Ca2+-induced Ca2+ release

    Cell Calcium

    (1991)
  • G.H. Biddlecome et al.

    Regulation of phospholipase C-beta1 by Gq and m1 muscarinic cholinergic receptor. Steady-state balance of receptor-mediated activation and GTPase-activating protein-promoted deactivation

    J. Biol. Chem.

    (1996)
  • V. Ralevic et al.

    Receptors for purines and pyrimidines

    Pharmacol. Rev.

    (1998)
  • G.D. Housley et al.

    Expression of the P2X(2) receptor subunit of the ATP-gated ion channel in the cochlea: implications for sound transduction and auditory neurotransmission

    J. Neurosci.

    (1999)
  • G. Burnstock

    Introduction: P2 receptors

    Curr. Top. Med. Chem.

    (2004)
  • G. Burnstock

    Purinergic signalling

    Br. J. Pharmacol.

    (2006)
  • F. Mammano et al.

    ATP-Induced Ca2+ release in cochlear outer hair cells: localization of an inositol triphosphate-gated Ca2+ store to the base of the sensory hair bundle

    J. Neurosci.

    (1999)
  • C. Chen et al.

    P2X receptors in cochlear Deiters’ cells

    Br. J. Pharmacol.

    (1998)
  • L. Lagostena et al.

    Purinergic control of intercellular communication between Hensen's cells of the guinea pig cochlea

    J. Physiol.

    (2001)
  • J.W. Chung et al.

    ATP and nitric oxide modulate intracellular calcium in isolated pillar cells of the guinea pig cochlea

    J. Assoc. Res. Otolaryngol.

    (2001)
  • R. Glueckert et al.

    Ultrastructure of the normal human organ of Corti. New anatomical findings in surgical specimens

    Acta Otolaryngol.

    (2005)
  • F. Mammano et al.

    Electrophysiological properties of Hensen's cells investigated in situ

    Neuroreport

    (1996)
  • L. Lagostena et al.

    Frequency dependence of electrical coupling in Deiters’ cells of the guinea pig cochlea

    Cell Commun. Adhes.

    (2001)
  • D.P. Corey et al.

    Ionic basis of the receptor potential in a vertebrate hair cell

    Nature

    (1979)
  • T.J. Jentsch

    Neuronal KCNQ potassium channels: physiology and role in disease

    Nat. Rev. Neurosci.

    (2000)
  • Cited by (82)

    • Comparative Anatomy of Glial Cells in Mammals

      2020, Evolutionary Neuroscience
    • Purinergic signaling in the organ of Corti: Potential therapeutic targets of sensorineural hearing losses

      2019, Brain Research Bulletin
      Citation Excerpt :

      In acute preparations, as the hemicochlea, this motion was not observed (Horváth et al., 2016), probably because of the strong coupling between the cells which limits the detection of visible motion of the cellular compartments. In case of hair cell damage elevated intracellular Ca2+ levels was detected in the neighbouring supporting cells which spread as Ca2+ waves to several 10–100 μm from the damaged area (Gale et al., 2004; Lahne and Gale, 2010; Piazza et al., 2007; Wong and Ryan, 2015). These intracellular Ca2+ level elevations are supposed to be caused by ATP (Berekmöri et al., 2019; Horváth et al., 2016) liberated from the injured cells.

    • Targeted single-cell electroporation loading of Ca<sup>2+</sup> indicators in the mature hemicochlea preparation

      2019, Hearing Research
      Citation Excerpt :

      The preparation was first used by our group for real functional imaging of intracellular Ca2+ signaling, which is implicated in the aforementioned phenomena (Horváth et al., 2016). In that study, the indicator dye was bulk loaded in its AM form, as in the majority of Ca2+ imaging studies on cells in the cochlea (Chan and Rouse, 2016; Dulon et al., 1993; Matsunobu and Schacht, 2000; Piazza et al., 2007). Bulk loading is convenient, but the dye remains in the extracellular space resulting in significant background staining and low S/N. AM dyes can be taken up by every cell, contaminating the responses of the cell of interest by fluorescence from adjacent responding cells (Fridberger et al., 1998).

    • Ca<sup>2+</sup> signaling, apoptosis and autophagy in the developing cochlea: Milestones to hearing acquisition

      2018, Cell Calcium
      Citation Excerpt :

      Other pharmacological manipulations showed that Ca2+ signaling in non-sensory cells of the GER and the LER share the same canonical IP3-dependent signal transduction cascade [115] described above. In particular, ATP-activated Ca2+ oscillations in the LER and spontaneous Ca2+ transients in the GER were both eliminated by thapsigargin, a noncompetitive inhibitor of the Ca2+-ATPase (SERCA) that causes the complete and irreversible depletion of Ca2+ from the endoplasmic reticulum, or by U73122, an inhibitor of phospholipase C [99,109] (Video S5). In addition, both types of IP3R-dependent Ca2+ signals were hampered by genetic manipulation of phosphatidylinositol phosphate kinase type 1γ (PIPKIγ), a key enzyme involved in the generation of the IP3 precursor phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] by phosphorylation of PI(4)P [116].

    View all citing articles on Scopus
    1

    The authors contributed equally.

    View full text