<<

REVIEWS

The sensory and motor roles of auditory hair cells

Robert Fettiplace* and Carole M. Hackney‡ Abstract | Cochlear hair cells respond with phenomenal speed and sensitivity to sound vibrations that cause submicron deflections of their hair bundle. Outer hair cells are not only detectors, but also generate force to augment auditory sensitivity and frequency selectivity. Two mechanisms of force production have been proposed: contractions of the cell body or active motion of the hair bundle. Here, we describe recently identified proteins involved in the sensory and motor functions of auditory hair cells and present evidence for each force generator. Both motor mechanisms are probably needed to provide the high sensitivity and frequency discrimination of the mammalian .

Electromechanical feedback Auditory sensing in occurs in a subdivision OHC contribution is known as the ‘cochlear amplifier’, The electrical change in an of the inner known as the cochlea, a fluid-filled a mechanism that increases both the amplitude and OHC caused by basilar tube coiled up like a snail’s shell to fit into the temporal frequency selectivity of vibration for membrane vibration generates at the base of the . Sound pressure fluctua- low-level sounds. The unique features of mammalian a movement of the OHC that, tions are transmitted to the cochlea in several steps: — the , the extended basilar in turn, feeds back to or alters basilar membrane vibration. vibrations of each , conduction through three membrane and the separation of function between IHCs small middle ear bones that act in a lever-like fashion, and OHCs — have all evolved because of selective pres- Reverse transduction and production of pressure waves within the cochlea sure to extend the upper frequency limit of hearing5. In normal forward transduction, to displace the basilar membrane (FIG. 1). This com- These changes enabled the first nocturnal mammals with hair cells convert mechanical stimuli into electrical plex mechanical coup ling ensures that sound energy small heads to spatially localize the sounds made by other responses. In reverse is efficiently transferred from air to cochlear fluid animals, especially calls of their young, on the basis of transduction, the electrical over a wide frequency range1,2. Detection of the sound interaural intensity differences. signals of hair cells evoke a stimulus and its conversion to an equivalent electrical Here, we summarize recent discoveries of hair cell mechanical output. waveform, termed mechanoelectrical transduction, mechanisms underlying mechanoelectrical transduction occurs in hair cells riding on the elongated basilar and frequency tuning. We describe the newly discov- membrane. Sound-induced motion of the basilar ered proteins responsible for the precise organization membrane excites the hair cells by deflecting their of the hair bundle, many of which have been revealed hair bundles to activate mechanoelectrical transduc- by studying the genetics of hereditary deafness6 (BOX 3). tion (MET) ion channels3. Owing to gradients in the We present recent evidence about the performance and size and stiffness of the cochlear partition (the basilar molecular identity of the MET channel. Finally, we membrane and associated hair cells and supporting describe the basis of reverse transduction, which enables *Department of Physiology, University of Wisconsin cells), the place of maximal vibration varies systematic- OHCs to act as force generators and contribute to coch- Medical School, 185 Medical ally with the sound frequency like resonances in a musical lear frequency selectivity. A fuller understanding of hair Sciences Building, 1300 instrument1. As a consequence, each hair cell produces cell mechanisms may settle the long-standing question University Avenue, Madison, responses that, near the threshold of hearing, are tuned of how the mammalian cochlea achieves its remarkable Wisconsin 53706, USA. to a characteristic frequency (CF) (BOX 1). sensitivity and frequency discrimination. ‡MacKay Institute of Communication and The mammalian cochlea contains two classes of Neuroscience, School of Life hair cell, inner and outer, with distinct functions (FIG. 2). The hair bundle Sciences, Keele University, Information about the acoustic environment — speech, Actin filaments. The site of hair cell transduction is the Staffordshire ST5 5BG, UK. music or other sounds in the outside world — is relayed hair bundle, an array of modified microvilli or Correspondence to R.F. e-mail: fettiplace@physiology. primarily by the electrical signals of inner hair cells (IHCs) arranged like a staircase in ranks of increasing height wisc.edu (BOX 2), whereas the main task of outer hair cells (OHCs) (FIG. 3). Deflections of the bundle towards its taller edge doi:10.1038/nrn1828 is to boost the stimulus by electromechanical feedback4. The bend the stereocilia at their tapered base and open the

NATURE REVIEWS | NEUROSCIENCE VOLUME 7 | JANUARY 2006 | 19 © 2006 Nature Publishing Group

REVIEWS

Outer earMiddle 2 days, the newly incorporated actin monomers make ear Cochlea their way down each filament and dissociate at its bottom 2 end in a process known as treadmilling (note that 0.5 0.1 turnover rates were measured in developing neonatal 0.2 5 animals and might be different in adults). Remarkably, 20 1 the speed of treadmilling is proportional to the length 10 of the stereocilium, so that in a bundle with three rows of m i different heights, actin filaments in all rows are recycled 15 s together . The steepness of the staircase is therefore pre- Cochlea served by the relative rates of actin polymerization in fluid the stereocilia of each row. The same mechanism might determine the sixfold increase in maximum stereociliary 2 0.2 rw 20 height from the base to the apex of the cochlea. t A central agent in the control of treadmilling is myosin Basilar membrane fluid XVa, a cousin of the myosin that powers skeletal muscle contractions. Antibody labelling in high-resolution microscopy has shown that myosin XVa is restricted Figure 1 | The sound conduction pathway. A section through the to the tips of the stereocilia, the site of actin incorpora- showing the sound conduction pathway in the mammalian ear. Vibrations of the eardrum tion15,16. Furthermore, its concentration is greatest in the or tympanum (t) are relayed via three middle ear bones, the (m), (i) and tallest stereociliary rank. Evidence that myosin XVa is (s), and initiate pressure waves in the cochlear fluids, the pressure being relieved at the (rw). The pressure waves set in motion the basilar membrane, on necessary to determine stereociliary length comes from which the and hair cells ride. The cochlea is divided into compartments studies of shaker 2 mice, which have a mutation in this filled with fluids of distinct ionic composition. Perilymph is similar to extracellular fluid, non-muscle myosin. Cochlear hair cells of shaker 2 mice but endolymph, which is found in the central compartment above the tops of the hair lack myosin XVa expression and have abnormally short cells, contains a high K+ concentration and a small amount of (20 µM) Ca2+. The cochlea is stereocilia16. Other molecules might assist myosin XVa shown as straight to illustrate its internal structure, but it is normally coiled as in the inset. in regulating actin polymerization in the stereocilia: for Different sound frequencies differentially excite different regions of the cochlea, the example, whirlin, which contains multiple PDZ domains, specific locations being given in kHz from 0.1 to 20 kHz in humans. Note that the usually implicated in protein–protein interactions. Defects frequency map is logarithmic, so that each decade occupies an equivalent distance on in whirlin also cause abnormally short stereocilia in whirler the basilar membrane. The components are drawn roughly to scale for the human ear, in mice, which have hair cell pathology like that of shaker 2 which the cochlea is 35 mm in length. mice16,17. The similar phenotype suggests that myosin XVa and whirlin interact functionally. Moreover, myosin XVa is required to transport whirlin to the tips of the stereocilia16. MET channels, whereas displacements in the opposite Whirlin might behave like a scaffolding protein and, direction close these channels7. Owing to this directional through its PDZ domains, organize a group of proteins sensitivity, sinusoidal motion of the bundle about its into a functional complex at the tip of the stereocilium to resting position, as elicited by a pure tone, modulates the determine actin turnover and stereociliary length. likelihood of channel opening to generate an electrical Osmiophilic rootlet representation of the sound stimulus. Several of the Interciliary links. Stereocilia within a hair bundle are The stem of a stereocilium that molecular pathways that endow the hair bundle with its interconnected by a series of fine extracellular filaments in electron micrographs is seen (FIG. 3) to be heavily stained by precise architecture revolve around filamentous actin, . In mature mammalian hair bundles, adjacent osmium, which probably polymers of β- and γ-actin, both of which are present stereocilia are connected along their shafts by side links, indicates a dense in the stereocilia8. Stereocilia are larger versions of the whereas tip links extend from the vertex of each stereo- concentration of protein. microvilli on many epithelial cells and have a core of cilium to the side of its taller neighbour18–20. Together, actin filaments9, crosslinked with espin and fimbrin10, the links form an extracellular matrix coupling the Shaker 2 mouse A congenitally deaf mouse with spanning their length from tip to base. Most of the fila- stereocilia, so that when force is applied at the tip of the a mutation in myosin XV that ments terminate in the lower shaft of the stereocilium, bundle all stereocilia rotate together, ensuring uniform results in abnormally short where they are probably anchored to the membrane by excitation of the MET channels21. Clues to the molecular stereocilia. actin-binding proteins such as radixin11. The distribu- basis of the interciliary connections have been provided Usher type I syndrome PDZ domain tion of actin makes the stereociliary body rigid but by studies of human and equivalent A peptide-binding domain enables it to pivot at the base by flexure of ~10% of the mouse deaf mutants. These mutations result in hair bun- that is important for the filaments that traverse the ankle region. These filaments dle disorganization and splaying of the stereocilia. Three organization of membrane are reinforced by tropomyosin and other, unidentified classes of protein affected in Usher’s syndrome have been proteins, particularly at proteins, which appear as an osmiophilic rootlet in electron isolated using molecular genetics22. First, the cadherins cell–cell junctions. It can bind to the carboxyl termini of micrographs of OHCs (FIG. 3a; REFS 12,13). (cadherin 23 and protocadherin 15), which are mem- proteins or can form dimers The precise structure of the hair bundle is dynami- brane proteins with extracellular domains that interlink, with other PDZ domains. PDZ cally maintained by regulating the turnover of the actin known to be important in cell adhesion. Second, inter- domains are named after the filaments. Transfecting cultured hair cells with β-actin mediate coupling proteins (harmonin and SANS (Usher proteins in which these sequence motifs were made visible by coupling to green fluorescent protein has syndrome 1G homologue)), which, like whirlin, have originally identified (PSD-95, shown that actin monomers are constantly being added PDZ domains to facilitate protein–protein crossbridging. Discs large, zona occludens 1). at the tips of the stereocilia14. Over the course of about Third, a second non-muscle myosin, myosin VIIa, which

20 | JANUARY 2006 | VOLUME 7 www.nature.com/reviews/neuro © 2006 Nature Publishing Group

REVIEWS

Box 1 | Auditory frequency selectivity Frequency selectivity as applied to the 100 describes its ability to distinguish different frequency OHC loss components of a complex sound stimulus such as speech or music. As an example, when two strings on a guitar are 80 plucked simultaneously, two distinct notes are heard corresponding to each of the vibrating strings. Each component reflects a sinusoidal vibration of the sound 60 source, in this case the guitar, with a given frequency. Most sounds, even those emanating from a musical instrument, are composite, but can be represented as the sum of a set 40 of sinusoidal components of different frequency and (dB) Threshold amplitude. To represent a sound, the auditory system must encode the relative amplitudes of all the constituents. Frequency 20 decomposition by the auditory system occurs largely in the cochlea, which separates the different components along its length, with high frequencies at one end and low 0 frequencies at the other (FIG. 1). The different components 0.1 1.0 10 then excite different hair cells, giving rise to action potentials in different auditory nerve fibres. The frequency Frequency (kHz) selectivity of an auditory nerve fibre (or hair cell) can be mapped by determining its ‘tuning curve’ — the intensity of any sound frequency that will make just this fibre respond. This intensity is known as the threshold. The frequency selectivity of each nerve fibre is not perfect: a small range of frequencies is encoded at low sound levels, with the selectivity broadening as the sound gets louder. The tuning curves are therefore ‘V’ shaped, but each fibre has a particular frequency, its characteristic frequency (CF), at which the threshold reaches a minimum (at the tip of the ‘V’). The cochlea contains hair cells and afferent nerve fibres with CFs that span the entire auditory range of the animal. Examples of frequency tuning curves for three auditory nerve fibres are shown in the figure. The destruction of outer hair cells (OHCs) results in greatly elevated sound threshold and a deterioration of frequency selectivity, as shown for the neuron with the highest CF.

is thought to anchor the membrane complex to the actin that they are confined to the tips of the stereocilia in cytoskeleton. Together, these three types of protein might close proximity to the tip-link insertion26,27. Recent work form a functional unit for the side links, promoting has suggested that a Ca2+-stabilized protein, cadherin 23, cohesion of the hair bundle22. An additional role of the which is one of the proteins defective in Usher’s syn- protein complex might be to keep the plasma membrane drome, is also a component of the tip links based on taut over the submembraneous cytoskeleton like a rubber its biochemical properties and on antibody labelling at glove stretched over the fingers of a . This is espe- the tips of the stereocilia28,29. However, there have been cially relevant for MET channel activation because it reports that cadherin 23 disappears in adult cochlear hair ensures that bundle deformation is rapidly transmitted to cells30,31, so other proteins might be involved or able to the channel without loss of mechanical energy in tissue substitute for cadherin 23. But the disparity might also distortion. Transduction would be impaired by loosen- reflect differences in expression levels or sensitivity of ing of the membrane, as occurs in mutations of myosin antibody detection. The molecular structure of the tip VIIa in shaker 1 mice. The hair bundles in shaker 1 links and their connection to the MET channels has yet mice need greater positive displacements before the to be fully elucidated, but it is central to understanding MET channels start to open23. how the channel is mechanically activated. The tip links are thought to have a special role in Whirler mouse transduction — one of transmitting force due to hair The mechanoelectrical transduction channel A mouse mutant that is bundle deformation to MET channels. Because the tip MET channel activation and adaptation. The hair bundle unresponsive to sound and links run roughly parallel to the hair bundle’s axis of sym- may be regarded as an accessory structure for transform- shows circling and head-tossing behaviour, indicative of both metry from the shorter to taller stereociliary rows, they ing stimuli that occur during inner ear stimulation to auditory and vestibular are oriented to apply force during rotation of the bundle the molecular scale suitable for gating the MET channel. dysfunction. The mutated towards its taller edge, and to unload with deflection Because of the bundle’s geometry, there is a roughly tenfold protein is known as whirlin and towards its shorter edge18. Two lines of evidence support reduction in displacement at the tip of the bundle to is involved in stereocilia the role of the tip links in transduction. First, exposing that at the end of the . The probability of channel elongation. hair cells to submicromolar Ca2+, buffered with a calcium opening is modulated between fully closed and fully Usher type I syndrome chelator such as BAPTA, destroys the tip links24 (probably open by submicron displacements of the tip of the hair One of three subtypes (I,II and because their structural integrity depends on association bundle (FIG. 4; REF. 3). Even with the loudest sound press- III) of hereditary disorder with Ca2+), along with other interciliary links, and also ures, the hair bundles rock to and fro with amplitude no characterized by sensorineural 25 deafness of cochlear origin abolishes transduction . Second, experiments to localize greater than the diameter of a single stereocilium, and 2+ combined with loss of vision MET channels (for example, using intracellular Ca dyes at auditory threshold the bundles may move of the order due to retinitis pigmentosa. to visualize Ca2+ influx through open channels) show of 1 nm — a stimulus approaching atomic dimensions.

NATURE REVIEWS | NEUROSCIENCE VOLUME 7 | JANUARY 2006 | 21 © 2006 Nature Publishing Group

REVIEWS

a Hair bundle In response to a step deflection of the hair bundle, the MET channels open in ~100 ms, but then re-close in Outer hair cell sequential phases of adaptation (FIG. 4; REF. 41). One phase occurs rapidly in a millisecond or two, but others take tens or hundreds of milliseconds. In response to a sustained displacement of the bundle, adaptation shifts the working Inner limits of the MET channels in the direction of the imposed hair cell stimulus without reducing the maximum current. So, at least one function of adaptation is to keep the MET chan- nels within the most sensitive part of their operating range. The fast component of adaptation might have other roles Hinge Basilar because in auditory organs its rate varies systematically Afferents point membrane 37,42 Efferent with hair cell CF . Original conclusions about the MET Osseous spiral channel came from work on non-mammals, but most of lamina these have been corroborated for mammalian cochlear hair cells38,43–46. However, the kinetics of MET channel activation and adaptation are more than an order of magni- b tude faster in mammals, consistent with a need to detect higher sound frequencies than non-mammals46. Stereociliary Ca2+ is a crucial signal for MET channel performance. Ca2+ enters through these channels, is buffered in OHCs by millimolar concentrations of the calcium-binding protein parvalbumin-β (REF. 47), and is extruded by a high density of the PMCA2a isoform of the plasma membrane CaATPase pump48. Changes in stereociliary Ca2+ affect both fast and slow phases of adaptation, and channel activation41,46. So, larger Ca2+ influx speeds up both activation and fast adaptation. Conversely, reducing external Ca2+ or raising intra- cellular Ca2+ buffering both extend the time course of channel activation and fast adaptation42,46. Owing to the submillisecond speed of these events, the likely target for Ca2+ is the channel itself or some neighbouring protein. Figure 2 | Cellular structure of the sound-detecting organ of Corti. a | Transverse 2+ section through a middle turn of the cochlea, showing the organ of Corti, an assembly of Ca might act on the MET channel directly to re-close 25 intricately shaped supporting cells and inner and outer hair cells supported by a flexible it , or induce relaxation of an adjacent element in series 49 basilar membrane. The organ of Corti is ~150 µm wide. b | Upward displacement of the with the channel to counteract the stimulus . The time basilar membrane stimulates the hair cells by bending their stereociliary bundles against constant of fast adaptation varies inversely with hair cell the acellular tectorial membrane. Because of the point about which the basilar CF: cells tuned to higher frequencies have faster adapta- membrane hinges, the inner hair cells must be stimulated mainly by motion of the tion42,46. One way to achieve this variation is by larger tectorial membrane. Signals from each inner hair cell are relayed to the brain via 10 to 20 Ca2+ influx in cells with higher CF. This is implemented afferent fibres of the VIIIth cranial nerve. Outer hair cells have both sensory and motor in turtle cochlear hair cells37 by altering the unitary con- capabilities and possess electromotility that underlies the cochlear amplifier. They have a ductance of the MET channels from <100 pS at low CF sparse afferent innervation (not shown) and are contacted mainly by efferent nerves, to >300 pS at high CF: channels with larger conductance which regulate the electromotility and influence cochlear sensitivity. have greater Ca2+ currents and faster adaptation. Although the exact mechanism for fast adaptation is unknown, there is strong evidence that slow adaptation The minimum auditory threshold in mammals occurs is mediated by another non-muscle myosin, myosin 1c at a sound pressure of 20 µPa and, at this sound level, the (REF. 50). Myosin 1c has been proposed to control ten- displacement of the basilar membrane is ∼1 nm (REF. 32). sion in the tip links, and therefore force applied to the Little amplification is introduced by the geometry of channel, by driving the upper attachment of the link the organ of Corti, and the displacement amplitude at the along the stereociliary wall. A possible scenario is as tips of the stereocilia is thought to be similar to that of follows. First, a cluster of myosin 1c, connected to the the basilar membrane33. upper end of the tip link, ascends the actin backbone Adaptation Much has been learned about the biophysical proper- to sustain tension in the link. Following bundle deflec- The decline in response, and 2+ therefore sensitivity, to a ties of the MET channel from electrical recordings of hair tion, Ca binds to myosin 1c, causing it to dissociate sustained stimulus. Adaptation cells in in vitro preparations. It is a cation channel, most from actin, which allows the link’s attachment point to is a property of many sensory selective for Ca2+ but with measurable permeability to slip and so relieve tension on the channel. On removal receptors that enables them to small organic cations whose size implies a wide-diameter of the stimulus, the MET channels close, stereociliary adjust their sensitivity to 3,34–36 2+ prevailing conditions and pore . This might explain its unusually large single- Ca concentration drops, and myosin 1c climbs back 25,37,38 respond only to changes in channel conductance of 100 pS or more . It also up the stereocilium to re-tension the links. Support for stimulus intensity. displays ultrafast kinetics to hair bundle deformation39,40. the involvement of myosin 1c has come from mutating

22 | JANUARY 2006 | VOLUME 7 www.nature.com/reviews/neuro © 2006 Nature Publishing Group

REVIEWS

Box 2 | Inner hair cells versus outer hair cells Inner hair cells (IHCs) convert sound-induced motion of the cochlear partition into changes in membrane potential — receptor potentials — that immediately modulate neurotransmitter release at synapses on the auditory nerve afferents (FIG. 2). IHCs lack prestin and, therefore, any associated somatic contractility, and are thought to function solely as sensory elements. Recordings from IHCs have been made to determine their receptor potentials and frequency tuning curves in intact, anaesthetized animals90,101. However, compared with outer hair cells (OHCs), less is known about their mechanoelectrical transduction (MET) currents in response to direct manipulation of hair bundles. Available measurements indicate that IHCs have similar sensitivity and hair bundle mechanics102,62, although it is not known whether they show fast adaptation. The hair bundles of IHCs are of comparable size and stereociliary complement to those of OHCs, but in vivo they are not embedded in the tectorial membrane99 and are probably stimulated by motion of the surrounding fluid. As a consequence, they respond primarily to basilar membrane velocity rather than displacement90. This velocity coupling will behave as a high-pass filter similar to adaptation of the MET channels and eliminate the direct current (DC) response. The cut-off frequency is set by the mechanical compliance and viscous damping of the hair bundle. The large afferent supply to IHCs contrasts with that of OHCs, which have a sparse afferent innervation of unknown function. However, OHCs are contacted by efferent fibres with cell bodies in the brainstem whose axons make cholinergic inhibitory synapses103 on OHCs. They might be part of a feedback pathway to regulate cochlear sensitivity. Electrical stimulation of the efferent axons reduces IHC receptor potentials, particularly near the characteristic frequency, and blunts their frequency tuning curves104, a finding that is consistent with the idea that OHCs are mechanical effector cells.

its ATP-binding site to confer susceptibility to inhibi- There may be no more than 200 functional channels tion by certain ADP analogues. Expression of this per cell (roughly twice the number of stereocilia26,37) mutant myosin 1c rendered slow adaptation in ves- and 10,000 hair cells per cochlea. Only recently has a tibular hair cells susceptible to block by ADP analogues likely candidate for the MET channel been identified introduced through the recording pipette51. Although as a member of the TRP channel family. TRP channels, not seen in those experiments, more recent work, in so named for the consequence of their mutation in which greater expression of the mutant myosin 1c Drosophila melanogaster photoreceptors (transient was achieved, shows that fast adaptation can also be receptor potential), are distant relatives of voltage-gated suppressed in the mutant52. This surprising observa- K+ channels, and many have a high Ca2+ permeability tion implies a link between the two forms of adapta- and large unitary conductance comparable with that of tion, and that fast adaptation also involves myosin 1c. the MET channel53. A search of this family has revealed But it cannot require myosin 1c to progress through a TRPA1 isoform localized to mammalian hair cells54. its ATPase cycle because this would be too slow to TRPA1, also known as ANKTM1, was previously found account for time constants of fast adaptation of <100 ms in nociceptive neurons, where it is activated by cold seen in mammalian OHCs46. temperatures55. Evidence that it is a component of the MET channel was obtained by reducing its expression in The molecular identity of the MET channel. Compared cultured mammalian hair cells. This was done by trans- with other types of ion channel, such as voltage-gated fecting hair cell cultures with a small interfering RNA K+ channels, much less is known about the molecular that would block translation of the endogenous mes- Time constant makeup of the MET channel and how the stimulus sage, a procedure that, after a few days, reduced the size The time required for the energy is coupled to channel opening. One reason of the MET currents. Expression of TRPA1 in HEK293 response of a system to decline to 37% of its initial value. For for this paucity of information is the small number of cells produced channels with a unitary conductance the cell membrane, this is the channel protein molecules expected in a single cochlea. (50–100 pS) and a pharmacological profile similar, product of its capacitance and resistance, setting the timescale over which Box 3 | Hereditary hearing loss membrane currents change the voltage. A small time constant Hearing impairment is the most common disabling sensory defect in humans. Severe to profound hearing loss affects 1 in means that a system’s 1,000 newborns, and another 1 in 2,000 children before they reach adulthood, and 60% of people older than 70 years response can change rapidly. have hearing loss of at least 25 dB (REF. 105). Sensorineural hearing loss, in which there is damage to the inner ear, is the most prevalent form of hearing impairment. It has a range of causes, including noise-induced and drug-induced damage, Unitary conductance as well as being age-related, resulting in wide variations in age of onset and in the sites and severity of the lesions. At least The electrical conductance of a 60% of hearing loss may have a genetic basis, a significant proportion of which is non-syndromic (that is, no other single ion channel: the current pathology seems to be associated with the same gene), and most of these genes are inherited in an autosomal recessive flowing through the channel mode (see Hereditary Hearing Loss Homepage). divided by the voltage applied Genetic studies of families with hereditary hearing loss have led to the discovery of at least 40 genes associated with across the cell membrane. Unitary conductance is a non-syndromic hearing loss, and more are being found each year. Such studies are proving to be a rich source of 6 characteristic channel property information about which molecules are important in the inner ear , as well as giving hope for new therapeutic but varies according to the ion approaches. These include strategies to prevent hair cell loss by gene manipulation, through to investigations of the use species present. of stem cell technology as a treatment for cell loss. It has also been recognized that differences in susceptibility to ototoxic agents could be a result of the variety of genetic backgrounds displayed by the human genome. A case in point is Ototoxic agents streptomycin, which probably enters hair cells through the mechanoelectrical transduction (MET) channel106 and targets Procedures or drugs that the mitochondria, culminating in cell death. Hypersensitivity to the ototoxic effects of streptomycin is associated with a damage hearing, primarily by point mutation in a ribosomal subunit of the mitochondrial DNA107. acting on the hair cells.

NATURE REVIEWS | NEUROSCIENCE VOLUME 7 | JANUARY 2006 | 23 © 2006 Nature Publishing Group

REVIEWS

abWhirlin

Myosin XVa

Actin PMCA2a Myosin 1c

Tip link (cadherin 23) 2+ Ca MET channel (TRPA1)

Ca2+

Top link Side link (myosin VIIa, Radixin Myosin harmonin, VIIa cadherin 23) Rootlet Cuticular plate

Figure 3 | Structure and protein composition of the stereociliary bundle. a | Transmission electron micrograph through the three rows of stereocilia on a guinea pig outer hair cell showing the osmiophilic regions at the stereociliary tips and side walls (the insertions of the tip link), and at the rootlets. The diameter of the tallest stereocilium is ~0.25 µm. The hair bundle of a mammalian outer hair cell contains 30 to 100 stereocilia and has a maximum height of between 1 and 6 µm according to its location99. b | Putative locations of some of the protein constituents of the stereocilia. Some of these (for example, myosin 1c) were first identified in vestibular hair cells and their location has not yet been confirmed in cochlear hair cells. MET, mechanoelectrical transduction; PMCA2a, plasma membrane CaATPase pump; TRPA1, transient receptor potential channel A1.

although not identical, to that of the MET channels56. The gating spring. Following provisional identification Definitive evidence that TRPA1 is an integral part of the of one subunit of the MET channel, an important step MET channel is still needed, and might be provided by is to elucidate how the channel connects to the tip link the generation of a TRPA1 knockout. on the external face of the membrane and is anchored to Even if TRPA1 is the MET channel, it is unlikely to the intracellular cytoskeleton. Knowledge of the protein be the only isoform in mammalian hair cells, because attachments will provide insight into how the force is to generate a range of MET channel properties (such as exerted across the channel, an important factor in deter- variation in unitary conductance or kinetics, as seen in mining the speed of activation. A key premise of existing the turtle) probably requires at least one other main (α) models of transduction is that the MET channels are subunit or an auxiliary (β) subunit. TRP channels are opened by force applied by elastic elements, the ‘gating tetrameric, with four α-subunits, like voltage-gated K+ springs’ , which are stretched when the hair bundle is channels, and mixing two α-subunit variants could theo- deflected. One end of the spring is pulled by displace- retically generate five species of heteromeric channel57 ment of the hair bundle, the other end is attached to with properties intermediate to channels formed from the channel’s hypothetical gate. It seems unlikely that each of the parent subunits alone. Another potential the gating springs correspond to the tip links, whose α-subunit is TRPML3, which, like TRPA1, localizes coiled double-helical structure suggests that they are to the hair cell cytoplasm and stereocilia58. This chan- inextensible and are probably rigid connections for force nel has not been fully characterized, but its mutation is transmission60. Alternative sites for the gating spring lie responsible for early-onset hearing loss in the varitint- in the channel itself or its coupling to the cytoskeleton. waddler mouse58. MET channel properties may also depend An important implication of the gating spring model Varitint-waddler mouse on the local membrane environment. The presence of is a reciprocal relationship between channel conforma- A mouse with deafness, vestibular and pigmentation the anionic lipid phosphatidylinositol-4,5-bisphosphate tional changes and force. A mechanical stimulus opens defects due to mutation in the (PtdIns(4,5)P2) in the apical membrane of the stereocilia the channel, but, conversely, opening the channel can mucolipin 3 gene, which is important for transduction, and depletion of this lipid itself generate force. A consequence of the model is that encodes the ion channel 59 slows the rate of adaptation . PtdIns(4,5)P2 might act by the force–displacement relationship of the hair bundle protein TRPML3. It is 2+ associated with progressive increasing the Ca affinity of the binding site on the MET is nonlinear over the range of bundle positions at which hair bundle disorganization channel or of another protein involved in the adaptation the channel is activated: it predicts that the bundle will and hair cell degeneration. mechanism. not behave like a simple spring for which extension is

24 | JANUARY 2006 | VOLUME 7 www.nature.com/reviews/neuro © 2006 Nature Publishing Group

REVIEWS

abeach hair cell conveys information about a narrow –1.5 0.8 frequency band. The origin of frequency selectivity has –1.0 0.4 fascinated both biological and physical scientists for more m) µ ( –0.5 than 100 years, but is still not fully understood. It relies x 0 ∆ partly on mechanical resonances of the basilar mem- 0 –0.4 brane. Sounds of different frequencies evoke localized –0.2 0 0.2 0.4 0.6 patterns of vibration at different positions along the ∆x (µm) membrane, high frequencies near the base of the coch- 0 c lea and low frequencies towards the apex1,32 (FIG. 1). One manifestation of mechanical tuning is the correlation τ = 100 µs –0.5 0 A between auditory frequency range and cochlear size. Elephants, which have low-frequency hearing extend- –0.5 ing to just 10 kHz, have a long, 60 mm cochlea (like a –1.0 tuba), whereas mice hear to 90 kHz and have a short,

Current (nA) Current –1.0 7 mm cochlea (like a piccolo). Humans are in between, Current (nA)Current (nA) Current 1 –1.5 –1.5 with a 35 mm cochlea and a 20 kHz auditory range . 0 5 10 15 20 0.6 ms Measurements of basilar membrane motion have been Time (ms) increasingly refined67 since von Békésy’s original obser- Figure 4 | Mechanoelectrical tranduction currents in outer hair cells. vations of basilar membrane tuning in human cadavers, a | Mechanoelectrical transduction (MET) ion channel currents measured at a –84 mV and now demonstrate a narrow mechanical tuning holding potential from a rat outer hair cell in response to displacements of the tip of closely resembling that of the electrical signals of IHCs the hair bundle (∆x). b | Sigmoidal relationship between peak current and bundle and auditory nerve fibres32. At least part of this tuning deflection. c | MET current onsets, showing the rapid activation of the response and is passive, underpinned by longitudinal gradients in the the adaptation, which has a time constant (τ ) of ~100 µs for small responses. A stiffness and dimensions of the cochlear partition1,68. However, there is substantial evidence that the passive tuning of the basilar membrane is highly augmented by

proportional to applied force. So, if a force (FB) is applied an active input from the hair cells that is contingent on to the tip of the bundle, the resulting displacement (X) their transducing ability. Both experimental and theoret- is given by61: ical arguments have led to a consensus that the cochlear amplifier is attributable to the generation of force by

FB = X∙Ks – A∙pO(X) + Fo (1) OHCs during excitation to oppose the viscous damping of the cochlear partition4,32,69,70. The most compelling

where Ks is the passive linear stiffness, pO is the prob- evidence for this idea is that damage to or loss of the

ability of opening of the MET channels, and Fo and A are OHCs or interference with their MET currents causes constants. In the gating spring model, A is the product an elevation in threshold and deterioration in cochlear of the single-channel gating force and the total number frequency selectivity. of channels per bundle. The negative term in equation 1 signifies an active component in which channel gating The somatic motor. The prevailing view is that OHCs generates a force in the same direction as the imposed produce force by longitudinal contractions of their cell displacement, causing hair bundle stiffness to decrease body, the somatic motor, driven by changes in membrane 61 (FIG. 5) with channel opening to reach a minimum when pO potential . Individual OHCs, dissociated from the is ~0.5. The existence of such a nonlinearity has been cochlea and stimulated electrically, shorten in response verified from measurements of bundle stiffness in vari- to depolarization and extend for hyperpolarization71,72. ous hair cell preparations61–64. These are performed by Because of the speed of the mechanical response, these applying force stimuli with a flexible glass fibre, and cells can theoretically change their shape in synchrony by monitoring the resulting movements using photo- with the sound stimulus for frequencies spanning the diode imaging of the bundle or the attached fibre. One entire auditory range. Although behaving like diminu- implication of the gating spring model is that effects of tive muscle fibres, OHCs use a quite different contractile adaptation are manifested not only in channel opening, mechanism from the ATP-driven actomyosin reaction but also in bundle motion. So, fast adaptation triggered in muscle. This involves a membrane protein, prestin73, by Ca2+ influx is accompanied by active motion of the which belongs to a solute carrier (SLC26) family of anion hair bundle65,66. Because the time constant of adaptation, transporters and is densely packed in the lateral mem- and therefore the speed of the hair bundle’s mechanical brane of OHCs74. Whether prestin transports anions Photodiode imaging response, is matched to hair cell CF, this process might is unclear75, but the presence of an anion-binding site is The stimulator casts a shadow 66 76 on a pair of photodiodes and, contribute to frequency tuning . believed to bestow voltage-sensitivity on the protein . as it moves, the light on one So, hyperpolarization promotes binding of intracellular photodiode increases while Cochlear frequency selectivity Cl– ions, inducing a conformational switch in prestin that on the other decreases. A remarkable feature of auditory sensation is its that expands its surface area and causes cell elongation. The difference in photocurrents (BOX 1) can be used to measure frequency selectivity — the ability of the cochlea In support of this mechanism, it has been shown that – displacements down to a few to separate the acoustic frequencies along its length like substituting other anions for intracellular Cl changes nanometres. an acoustic prism. As a consequence, at low sound levels the voltage dependence of the electromotile process. The

NATURE REVIEWS | NEUROSCIENCE VOLUME 7 | JANUARY 2006 | 25 © 2006 Nature Publishing Group

REVIEWS

abHair bundle Tectorial membrane Hair bundle frequency rather than decline. A way of circumventing motor Force the time constant limitation has been proposed, by Outer hair cell activating the prestin with extracellular potentials instead of the usual excursions in intracellular poten- tial83. Extracellular potentials are generated by flow of – – Cl– Cl –Cl + transduction currents summed over a group of hair – cells and are not attenuated by the OHC membrane time Cl– Cl– Cl Hinge point – + Basilar membrane constant. However, measurements of these voltages Somatic Cl– Cl– Cl– + motor adjacent to the OHC indicate that they are at most a – few millivolts even at high sound levels84, which might Cl– – – + Cl –Cl be insufficient to affect prestin. Although a small peri- Hair cell odic component of the receptor potential will remain contraction at high frequencies, unless a new mode of prestin gating is invoked75, the simplest hypothesis is that the Prestin somatic motor is restricted to operating on a cycle- by-cycle basis at frequencies less than a few kilohertz. Figure 5 | The putative motors of outer hair cells. Outer hair cells can generate force, This is not to say that somatic motility is useful only mechanically boosting sound-induced vibrations of the hair bundle and augmenting at low frequencies. At frequencies above that imposed frequency tuning. Two mechanisms have been advanced to explain this cochlear by the membrane time constant, prestin would be amplifier: the somatic motor and the hair bundle motor. a | In the resting state (–), controlled by sustained depolarizing receptor poten- Cl– ions are bound to prestin molecules in the lateral membrane of the hair cell. When tials, acting as another form of adaptation to keep the force is applied to the hair bundle, the cell is depolarized (+), the Cl– ions dissociate and the prestin changes conformation, reducing its area in the plane of the membrane and hair bundle in its most sensitive working range. Such shortening the hair cell body (the somatic motor). Adaptation of mechanoelectrical negative feedback by prestin activation (FIG. 5b) would transduction (MET) channels, which are activated by bending of the stereocilia at their decrease the proportion of MET conductance at rest tapered base, also causes the hair bundle to produce extra force in the direction of the and reduce the direct current (DC) component of the stimulus (the hair bundle motor). The amplitudes of the hair bundle movements have OHC receptor potential. In line with this suggestion is been exaggerated to illustrate the concept. b | Schematic diagram showing the effects the finding that changes in OHC membrane potential of the somatic motor (red arrows) on the organ of Corti mechanics100, which leads to produced by current injection can alter the operating downward motion of the reticular lamina (the upper surface of the organ of Corti) and range of the MET channels: depolarization, which a negative deflection of the hair bundle. This is a negative feedback pathway, as a would cause OHC contraction, decreased the fraction positive deflection of the hair bundle causes outer hair cell depolarization, cell of channels open at rest, equivalent to adaptation85. contraction and opposing motion of the bundle. The hair bundle motor. An alternative mode of ampli- fication arises from force generated by the hair bun- importance of prestin is underscored by construction of dle86,87. Active hair bundle movements have been well mutations that reduce its expression. Targeted deletion documented in non-mammalian hair cells, where they of prestin in mice causes loss of OHC electromotility have been linked to the two phases of MET channel and a 100-fold or more reduction in cochlear sensi- adaptation. The most prominent movement in turtle tivity77 without affecting forward mechano electrical auditory hair cells closely parallels fast adaptation66. A transduction78. positive deflection of the bundle opens MET channels There is a major unresolved problem with the and increases stereociliary Ca2+, which re-closes the somatic motor as the site of the cochlear amplifier. channels and causes the bundle to spring back, oppos- Because it is voltage-dependent, cell length is control- ing the stimulus. This has been referred to as a ‘twitch’ led by receptor potentials that result from hair bundle or recoil65,66, and it has sufficient speed to operate at motion. At high frequencies, the receptor potential is frequencies in the auditory range. The time constant low-pass filtered by the membrane time constant, the of the mechanical response, like that of fast adaptation, product of membrane resistance and capacitance. varies with hair cell CF and is smaller in cells tuned to Minimum time constants of a few tenths of a milli- higher frequencies. The mechanism follows from the second have been reported79,80, which implies that the gating spring model of transduction, in which channel Low-pass filter periodic component compliance 2+ A filter that suppresses all of the receptor potential will be opening increases hair bundle : Ca -evoked frequencies above a certain attenuated in proportion to frequency above ∼1 kHz. re-closure of the channel decreases compliance, caus- point known as the cut-off So, although somatic motor transitions are potentially ing the bundle to move back towards its original resting frequency. very fast81 and extend up to 80 kHz, their recruitment position. The hair bundle motor is probably controlled must be limited by the size of the driving voltage. The by conformational changes in the MET channel pro- Periodic component A repetitive waveform: for roll-off with increasing frequency might be partially tein, and so, unlike the somatic motor, is not limited 45,46 example, a sine wave. offset by a larger size of MET current as in turtles, by the membrane time constant, only by the kinetics and a greater density of prestin82 in OHCs with high of the channel activation and adaptation. However, Compliance CF. However, there is a more stringent demand on the compared with the somatic motor, the force developed The inverse of stiffness; the displacement of an elastic active process, because, in order to cancel a frictional may be small. According to the gating spring model element produced by a known force proportional to basilar membrane velocity, force (equation 1) the maximum force is A, the product of force. production by the OHCs might need to increase with the single-channel gating force (∼0.5 pN) and the total

26 | JANUARY 2006 | VOLUME 7 www.nature.com/reviews/neuro © 2006 Nature Publishing Group

REVIEWS ab Sylgard bead 1.0 current. This pheno menon differs from the process linked to adaptation in non-mammalian hair cells in two respects: it has opposite polarity (equivalent Piezo O Hair bundle p 0.5 to positive feedback) and it generates much larger forces. Maximum force generation, estimated as the difference between steady state and instantane- Flexible –0.2 0.2 0.4 0.6 0.8 ous force–displacement plots, is >500 pN. The time ∆ µ fibre x ( m) course of force production is similar to that of adapta- c 2 tion, and, like adaptation, is slowed by lowering the Instant extracellular Ca2+ concentration. The temporal limits 1 of this mechanical process are unknown because of the limited speed with which forces can be delivered Steady state Force (nN) Force ∆ force to the hair bundle using the flexible-fibre technique. However, fast adaptation time constants of <50 µs have –0.2 0.2 0.4 0.6 0.8 Sylgard bead been measur ed in rat OHCs, and when corrected for Outer hair cell ∆x (µm) –1 in vivo conditions they might be substantially faster46. So, even in the relatively low Ca2+ concentration of 20 Figure 6 | Mechanical properties of an outer hair cell bundle. a | A method of µM in cochlear endolymph, prominent fast adapta- measuring hair bundle mechanics. The outer hair cell bundle is deflected with a tion is present with a submillisecond time constant Sylgard bead on the tip of a flexible glass fibre whose fixed end is deflected by a 46 piezoelectric actuator (piezo). Hair bundle motion is inferred from the change in that varies with CF . When these time constants are photocurrent in a photodiode array (not shown) on which the image of the bead is extrapolated to the conditions prevailing in vivo (for projected. b | Mechanoelectrical transduction (MET) channels increase their example, temperature and endolymphatic potential), ∆ cut-off frequencies similar to the CF are predicted46. probability of opening (pO) with positive hair bundle displacement ( x). c | Relationship between applied force and bundle displacement. At early times At present, the mechanism of force generation is (<0.1 ms; instant) the bundle behaves like a simple spring in which displacement is unknown, but its speed and relationship to adaptation approximately proportional to force. At later times (1 ms; steady state), with the implicate the mechanotransduction apparatus as the adaptation of the MET channels, the force–displacement relationship becomes source. The OHC hair bundle motor might be sufficiently nonlinear. Both plots have been calculated from equation 1 (main text) using the powerful to complement forces generated by the somatic p – ∆x relationship shown in b, a passive stiffness, K, equal to 3 mN m–1 and values for O motor, especially at high frequencies at which the intra- the constant A of 100 pN (instant) and 2,000 pN (steady state). Note that in this description, A is a function of time, whereas in the original derivation of equation 1, cellular receptor potential is attenuated. This conclusion A was a constant. The deviation between the instantaneous and steady-state plots is bolstered by measurements of IHC bundle motion in represents the force developed by the bundle. The time course of force production an in vitro cochlear preparation, which show a nonlinear mirrors that of fast adaptation. mechanical amplification dependent on the Ca2+ influx through MET channels94. So, vibrations of the organ of Corti evoked by acoustic stimulation were augmented by a mechanism requiring only Ca2+ current to flow through number of channels (200)61,63. This sets an upper limit the MET channels, which would generate meagre receptor of 100 pN, less than a tenth of that generated by the potentials unable to activate prestin. somatic motor. The peak force produced by electrically stimulating isolated OHCs has been estimated as ∼100 Conclusions pN per millivolt change in membrane potential88,89. During the past decade, important advances have been As maximum receptor potentials above 20 mV have made in uncovering the molecular basis of auditory been recorded in OHCs45,90, the force produced by sensation, but crucial steps in the transduction chain the somatic motor is more than 2,000 pN. The active still need to be determined. One is the complete char- bundle movements characterized in lower acterization of the MET channel and its attachments to might be the primary source of mechanical amplifica- the cytoskeleton and the tip link. This may clarify how Sylgard bead tion in the avian cochlea5, where force generation is force is delivered to the channel to explain its sensitivity A drop of silicone rubber, a summed from up to 12 rows of short hair cells that are and speed, enabling the hair cell to detect bundle vibra- mixture of a base and a curing equivalent to mammalian OHCs but lack prestin or any tions with subnanometre amplitude and microsecond agent, that is deposited on the 91,92 tip of a glass fibre while liquid, sign of somatic motility . delay. How many different channel subunits are needed then polymerized and Although OHCs in mammals, similar to those in to produce a variation in channel conductance with CF? hardened by heating. frogs and turtles, show fast adaptation, until recently A second area is apportioning the relative contributions there has been no evidence that they could generate of the OHC bundle motor and the somatic motor in Piezoelectric actuator forces comparable to those seen in non-mammals. cochlear amplification. The simplest hypothesis is that A ceramic crystal that deforms 95 when a potential difference is However, new experiments on rat OHCs have revealed both mechanisms are important , but perhaps their 93 applied across it. Piezoelectric the presence of a strong force generator . The force– relative contributions vary with frequency. Such divi- devices require large voltages displacement relationship of the hair bundle was sion of labour might accord with the change in form of to induce movements of a found to be nonlinear, the bundle becoming increas- the neural tuning curves with increasing frequency96. micron. Such devices are (FIG. 6) 97 87,98 reversible and also generate a ingly comp liant with MET channel gating . How much does hair bundle or somatic stiffness voltage in response to Furthermore, the nonlinearity developed with a time contribute to the mechanical properties of the cochlear mechanical stress. course similar to that of fast adaptation of the MET partition and how does the organ of Corti deform during

NATURE REVIEWS | NEUROSCIENCE VOLUME 7 | JANUARY 2006 | 27 © 2006 Nature Publishing Group

REVIEWS

sound stimulation? Resolving these issues will require of proteins such as myosin VIIa to hair bundle develop- several lines of inquiry: defining the molecular basis ment from their role in the adult cochlea. Identifying the of force generation by the hair bundle; exploring the molecular constituents alone will not solve the problem, micromechanics of the cochlear partition; and devising which will require a broader approach that incorporates models that realistically incorporate OHC properties. It both physiological measurements and modelling to dis- will also be important to distinguish the contributions sect the cochlear machine.

1. Von Békésy, G. Experiments in Hearing (McGraw-Hill, Evidence that interactions between three Usher I 42. Ricci, A. J., Wu, Y.-C. & Fettiplace, R. The endogenous New York, 1960). gene products are important for hair bundle calcium buffer and the time course of transducer 2. Geisler, C. D. From Sound to Synapse (Oxford Univ. morphogenesis. adaptation in auditory hair cells. J. Neurosci. 18, Press, New York, 1998). 23. Kros, C. J. et al. Reduced climbing and increased 8261–8277 (1998). 3. Hudspeth, A. J. How the ear’s works work. Nature slipping adaptation in cochlear hair cells of mice with 43. Kros, C. J., Rüsch, A. & Richardson, G. P. Mechano- 341, 397–404 (1989). Myo7a mutations. Nature Neurosci. 5, 41–47 electrical transducer currents in hair cells of the 4. Dallos, P. The active cochlea. J. Neurosci. 12, (2002). cultured neonatal mouse cochlea. Proc. R. Soc. 4575–4585 (1992). 24. Assad, J. A., Shepherd, G. M. & Corey, D. P. Tip-link Lond. B 249, 185–193 (1992). 5. Manley, G. A. Cochlear mechanisms from a integrity and mechanical transduction in Reports the first use of an isolated mammalian phylogenetic viewpoint. Proc. Natl Acad. Sci. USA 97, hair cells. Neuron 7, 985–994 (1991). cochlear preparation to record large transducer 11736–11743 (2000). 25. Crawford, A. C., Evans, M. G. & Fettiplace, R. The currents. 6. Friedman, T. B. & Griffith, A. J. Human nonsyndromic actions of calcium on the mechano-electrical 44. Kennedy, H. J., Evans, M. G., Crawford, A. C. & sensorineural deafness. Annu. Rev. Genomics Hum. transducer current of turtle hair cells. J. Physiol. Fettiplace, R. Fast adaptation of mechanoelectrical Genet. 4, 341–402 (2003). (Lond.) 434, 369–398 (1991). transducer channels in mammalian cochlear hair cells. 7. Shotwell, S. L., Jacobs, R. & Hudspeth, A. J. 26. Denk, W., Holt, J. R., Shepherd, G. M. & Corey, D. P. Nature Neurosci. 6, 832–836 (2003). Directional sensitivity of individual vertebrate hair Calcium imaging of single stereocilia in hair cells: 45. He, D. Z. Z., Jia, S. & Dallos, P. Mechanoelectrical cells to controlled deflection of their hair bundles. Ann. localization of transduction channels at both ends of transduction of adult outer hair cells studied in a NY Acad. Sci. 374, 1–10 (1981). tip links. Neuron 15, 1311–1321 (1995). gerbil cochlea. Nature 429, 766–770 (2004). 8. Furness, D. N., Katori, Y., Mahendrasingam, S. & 27. Lumpkin, E. A. & Hudspeth, A. J. Detection of Ca2+ 46. Ricci, A. J., Kennedy, H. J., Crawford, A. C. & Hackney, C. M. Differential distribution of β- and entry through mechanosensitive channels localizes the Fettiplace, R. The transduction channel filter in γ-actin in guinea-pig cochlear sensory and supporting site of mechanoelectrical transduction in hair cells. auditory hair cells. J. Neurosci. 25, 7831–7839 cells. Hear. Res. 207, 22–34 (2005). Proc. Natl Acad. Sci. USA 92, 10297–10301 (1995). (2005). 9. Tilney, L. G. & Tilney, M. S. Functional organization of 28. Siemens, J. et al. Cadherin 23 is a component of the 47. Hackney, C. M., Mahendrasingam, S., Penn, A. & the cytoskeleton. Hear. Res. 22, 55–77 (1986). tip link in hair-cell stereocilia. Nature 428, 950–955 Fettiplace, R. The concentrations of calcium buffering 10. Bartles, J. R. Parallel actin bundles and their multiple (2004). proteins in mammalian cochlear hair cells. J. Neurosci. actin-binding proteins. Curr. Opin. Cell Biol. 12, 29. Söllner, C. et al. Mutations in cadherin 23 affect tip 25, 7867–7875 (2005). 72–78 (2000). links in zebrafish sensory hair cells. Nature 428, 48. Dumont, R. A. et al. Plasma membrane Ca2+-ATPase 11. Pataky, F., Pironkova, R. & Hudspeth, A. J. Radixin is a 955–959 (2004). isoform 2a is the PMCA of hair bundles. J. Neurosci. constituent of stereocilia in hair cells. Proc. Natl Acad. 30. Michel, V. et al. Cadherin 23 is a component of the 21, 5066–5078 (2001). Sci. USA 101, 2601–2606 (2004). transient lateral links in the developing hair bundles of 49. Martin, P., Bozovic, D., Choe, Y. & Hudspeth, A. J. 12. Slepecky, N. & Chamberlain, S. C. Immunoelectron cochlear sensory cells. Dev. Biol. 280, 281–294 Spontaneous oscillation by hair bundles of the microscopic and immunofluorescent localization of (2005). bullfrog’s sacculus. J. Neurosci. 23, 4533–4548 cytoskeletal and muscle-like contractile proteins in 31. Lagziel, A. et al. Spatiotemporal pattern and isoforms (2003). inner ear sensory hair cells. Hear. Res. 20, 245–260 of cadherin 23 in wild type and waltzer mice during 50. Gillespie, P. G. & Cyr, J. L. Myosin-1c, the hair cell’s (1985). inner ear hair cell development. Dev. Biol. 280, adaptation motor. Annu. Rev. Physiol. 66, 521–545 13. Hackney, C. M., Karkanevatos, A. & Furness, D. N. 295–306 (2005). (2004). Distribution of tropomyosin in guinea pig cochlear hair 32. Robles, L. & Ruggero, M. A. Mechanics of the 51. Holt, J. R. et al. A chemical-genetic strategy implicates cells. J. Physiol. (Lond.) 527, 92P (2000). mammalian cochlea. Physiol. Rev. 81, 1305–1352 myosin-1c in adaptation by hair cells. Cell 108, 14. Schneider, M. E., Belyantseva, I. A., Azevedo, R. B. & (2001). 371–381 (2002). Kachar, B. Rapid renewal of auditory hair bundles. Comprehensive review of the measurements and Mutation of myosin-1c to confer selectivity to block Nature 418, 837–838 (2002). mechanisms of the mechanical frequency tuning of by ADP analogues unequivocally shows the role of 15. Rzadzinska, A. K., Schneider, M. E., Davies, C., the basilar membrane. the motor protein in hair cell adaptation. Riordan, G. P. & Kachar, B. An actin molecular 33. Hu, X., Evans, B. N. & Dallos, P. Direct visualization of 52. Stauffer, E. A. et al. Fast adaptation in vestibular hair treadmill and myosins maintain stereocilia functional organ of Corti kinematics in a hemicochlea. cells requires myosin-1c activity. Neuron 47, 541–553 architecture and self-renewal. J. Cell Biol. 164, J. Neurophysiol. 82, 2798–2807 (1999). (2005). 887–897 (2004). 34. Ohmori, H. Mechano-electrical transduction currents 53. Pedersen, S. F., Owsianik, G. & Nilius, B. TRP Shows treadmilling of actin along the stereocilia at in isolated vestibular hair cells of the chick. J. Physiol. channels: an overview. Cell Calcium 38, 233–252 a rate proportional to their length and proposes (Lond.) 359, 189–217 (1985). (2005). that myosin XVa that is localized at stereociliary 35. Ricci, A. J. & Fettiplace, R. Calcium permeation of the 54. Corey, D. P. et al. TRPA1 is a candidate for the tips regulates treadmilling rate and stereociliary turtle hair cell mechanotransducer channel and its mechanosensitive transduction channel of vertebrate length. relation to the composition of endolymph. J. Physiol. hair cells. Nature 432, 723–730 (2004). 16. Belyantseva, I. A. et al. Myosin-XVa is required for tip (Lond.) 506, 159–173 (1998). Identification of the most plausible candidate for localization of whirlin and differential elongation of 36. Farris, H. E., LeBlanc, C. L., Goswami, J. & Ricci, A. J. the long sought-after MET channel protein. hair-cell stereocilia. Nature Cell Biol. 7, 148–156 Probing the pore of the auditory hair cell 55. Story, G. M. et al. ANKTM1, a TRP-like channel (2005). mechanotransducer channel in turtle. J. Physiol. expressed in nociceptive neurons is activated at cold 17. Mburu, P. et al. Defects in whirlin, a PDZ domain (Lond.) 558, 769–792 (2004). temperatures. Cell 112, 819–829 (2003). molecule involved in stereocilia elongation, cause 37. Ricci, A. J., Crawford, A. C. & Fettiplace, R. Tonotopic 56. Nagata, K., Duggan, A., Kumar, G. & Garcia-Anoveros, J. deafness in the whirler mouse and families with variation in the conductance of the hair cell Nociceptor and hair cell transducer properties of DFNB31. Nature Genet. 34, 421–428 (2003). mechanotransducer channel. Neuron 40, 983–990 TRPA1, a channel for pain and hearing. J. Neurosci. 18. Pickles, J. O., Comis, S. D. & Osborne, M. P. Cross- (2003). 25, 4052–4061 (2005). links between stereocilia in the guinea pig organ of Uses the destruction of tip links with BAPTA to 57. Schaefer, M. Homo- and heteromeric assembly of Corti, and their possible relation to sensory record single MET channels, showing their variation TRP channel subunits. Pflugers Arch. 451, 35–42 transduction. Hear. Res. 15, 103–112 (1984). in conductance along the cochlea. (2005). 19. Furness, D. N. & Hackney, C. M. Cross-links between 38. Géléoc, G. S., Lennan, G. W., Richardson, G. P. & 58. Di Palma, F. et al. Mutations in Mcoln3 associated stereocilia in the guinea pig cochlea. Hear. Res. 18, Kros, C. J. A quantitative comparison of with deafness and pigmentation defects in varitint- 177–188 (1985). mechanoelectrical transduction in vestibular and waddler (Va) mice. Proc. Natl Acad. Sci. USA 99, 20. Goodyear, R. J., Marcotti, W., Kros, C. J. & auditory hair cells of neonatal mice. Proc. R. Soc. 14994–14999 (2002). Richardson, G. P. Development and properties of Lond. B 264, 611–621 (1997). 59. Hirono, M., Denis, C. S., Richardson, G. P. & stereociliary link types in hair cells of the mouse 39. Corey, D. P. & Hudspeth, A. J. Kinetics of the receptor Gillespie, P. G. Hair cells require phosphatidylinositol cochlea. J. Comp. Neurol. 485, 75–85 (2005). current in bullfrog saccular hair cells. J. Neurosci. 3, 4,5-bisphosphate for mechanical transduction and 21. Flock, A., Flock, B. & Murray, E. Studies on the 962–976 (1983). adaptation. Neuron 44, 309–320 (2004). sensory hairs of receptor cells in the inner ear. Acta 40. Crawford, A. C., Evans, M. G. & Fettiplace, R. 60. Kachar, B., Parakka, l. M., Kurc, M., Zhao, Y. & Otolaryngol. 83, 85–91 (1977). Activation and adaptation of transducer currents in Gillespie, P. G. High-resolution structure of hair-cell tip 22. Boeda, B. et al. Myosin VIIa, harmonin and cadherin turtle hair cells. J. Physiol. (Lond.) 419, 405–434 links. Proc. Natl Acad. Sci. USA 97, 13336–13341 23, three Usher I gene products that cooperate to (1989). (2000). shape the sensory hair bundle. EMBO J. 21, 41. Eatock, R. A. Adaptation in hair cells. Annu. Rev. 61. Howard, J. & Hudspeth, A. J. Compliance of the hair 6689–6699 (2002). Neurosci. 23, 285–314 (2000). bundle associated with gating of mechanoelectrical

28 | JANUARY 2006 | VOLUME 7 www.nature.com/reviews/neuro © 2006 Nature Publishing Group

REVIEWS

transduction channels in the bullfrog’s saccular hair cochlear amplifier. Nature 419, 300–304 96. Kiang, N. Y., Liberman, M. C., Sewell, W. F. & cell. Neuron 1, 189–199 (1988). (2003). Guinan, J. J. Single unit clues to cochlear mechanisms. First demonstration of and elegant theory for the 78. Cheatham, M. A., Huynh, K. H., Gao, J., Zuo, J. & Hear. Res. 22, 171–182 (1986). contribution of MET channel gating to hair bundle Dallos, P. Cochlear function in Prestin knockout mice. 97. Chan, D. K. & Hudspeth, A. J. Mechanical responses mechanics. J. Physiol. (Lond.) 560, 821–830 (2004). of the organ of Corti to acoustic and electrical 62. Russell, I. J., Kössl, M. & Richardson, G. P. Nonlinear 79. Santos-Sacchi, J. On the frequency limit and phase of stimulation in vitro. Biophys. J. 16 Sep 2005 mechanical responses of mouse cochlear hair bundles. outer hair cell motility. J. Neurosci. 12, 1906–1916 (doi:10.1529/biophysj.105.070474). Proc. R. Soc. Lond. B 250, 217–227 (1992). (1992). 98. He, D. Z., Jia, S. & Dallos, P. Prestin and the dynamic 63. van Netten, S. M. & Kros, C. J. Gating energies and 80. Housley, G. D. & Ashmore, J. F. Ionic currents of outer stiffness of cochlear outer hair cells. J. Neurosci. 23, forces of the mammalian hair cell transducer channel hair cells isolated from the guinea-pig cochlea. 9089–9096 (2003). and related hair bundle mechanics. Proc. R. Soc. J. Physiol. (Lond.) 448, 73–98 (1992). 99. Lim, D. J. Cochlear anatomy related to cochlear Lond. B 267, 1915–1923 (2000). 81. Frank, G., Hemmert, W. & Gummer, A. W. Limiting micromechanics. A review. J. Acoust. Soc. Am. 67, 64. Ricci, A. J., Crawford, A. C. & Fettiplace, R. dynamics of high-frequency electromechanical 1686–1695 (1980). Mechanisms of active hair bundle motion in auditory transduction of outer hair cells. Proc. Natl Acad. Sci. 100. Mammano, F. & Ashmore, J. F. Reverse transduction hair cells. J. Neurosci. 22, 44–52 (2002). USA 96, 4420–4425 (1999). measured in the isolated cochlea by laser 65. Benser, M. E., Marquis, R. E. & Hudspeth, A. J. Rapid, 82. Santos-Sacchi, J., Kakehata, S., Kikuchi, T., Katori, Y. & Michelson interferometry. Nature 365, 838–841 active hair bundle movements in hair cells from the Takasaka, T. Density of motility-related charge in the (1993). bullfrog’s sacculus. J. Neurosci. 16, 5629–5643 outer hair cell of the guinea pig is inversely related to 101. Russell, I. J. & Sellick, P. M. Intracellular studies of (1996). best frequency. Neurosci. Lett. 256, 155–158 hair cells in the mammalian cochlea. J. Physiol. (Lond.) 66. Ricci, A. J., Crawford, A. C. & Fettiplace, R. Active hair (1998). 284, 261–290 (1978). bundle motion linked to fast transducer adaptation in 83. Dallos, P. & Evans, B. N. High frequency outer hair cell 102. Russell, I. J., Cody, A. R. & Richardson, G. P. The auditory hair cells. J. Neurosci. 20, 7131–7142 motility: corrections and corrigendum. Science 268, responses of inner and outer hair cells in the basal turn (2000). 1420–1421 (1995). of the guinea-pig cochlea and in the mouse cochlea 67. Rhode, W. S. Observations of the vibration of the 84. Fridberger, A. et al. Organ of Corti potentials and the grown in vitro. Hear. Res. 22, 199–216 (1986). basilar membrane in squirrel monkeys using the motion of the basilar membrane. J. Neurosci. 24, 103. Eybalin, M. Neurotransmitters and neuromodulators Mossbauer technique. J. Acoust. Soc. Am. 49, 10057–10063 (2004). of the mammalian cochlea. Physiol. Rev. 73, 1218–1231 (1971). 85. Russell, I. J. & Kössl, M. The voltage responses of hair 309–373 (1993). 68. Emadi, G., Richter, C. P. & Dallos, P. Stiffness of the cells in the basal turn of the guinea-pig cochlea. 104. Brown, M. C. & Nuttall, A. L. Efferent control of gerbil basilar membrane: radial and longitudinal J. Physiol. (Lond.) 435, 493–511 (1991). cochlear inner hair cell responses in the guinea-pig. variations. J. Neurophysiol. 91, 474–488 86. Crawford, A. C. & Fettiplace, R. The mechanical J. Physiol. (Lond.) 354, 625–646 (1984). (2004). properties of ciliary bundles of turtle cochlear hair 105. Atar, O. & Avraham, K. B. Therapeutics of hearing loss: 69. Neely, S. T. & Kim, D. O. An active cochlear model cells. J. Physiol. (Lond.) 364, 359–379 expectations vs reality. Drug Discov. Today 10, showing sharp tuning and high sensitivity. Hear. Res. (1985). 1323–1230 (2005). 9, 123–130 (1983). Used photodiode imaging to show both active and 106. Marcotti, W., van Netten, S. M. & Kros, C. J. The 70. Allen, J. B. & Neely, S. T. Micromechanical models of spontaneous motion of the hair bundle driven by aminoglycoside antibiotic dihydrostreptomycin the cochlea. Phys. Today July, 40–47 (1992). changes in membrane potential rapidly enters mouse outer hair cells through the 71. Brownell, W. E., Bader, C. R., Bertrand, D. & 87. Hudspeth, A. J. Mechanical amplification of stimuli by mechano-electrical transducer channels. J. Physiol. de Ribaupierre, Y. Evoked mechanical responses of hair cells. Curr. Opin. Neurobiol. 7, 480–486 (Lond.) 567, 505–521 (2005). isolated cochlear outer hair cells. Science 227, (1997). 107. Cortopassi, G. & Hutchin, T. A molecular and cellular 194–196 (1985). 88. Hallworth, R. Passive compliance and active force hypothesis for aminoglycoside-induced deafness. Hear. 72. Ashmore, J. F. A fast motile response in guinea-pig generation in the guinea pig outer hair cell. Res. 78, 27–30 (1994). outer hair cells: the cellular basis of the cochlear J. Neurophysiol. 74, 2319–2328 (1995). amplifier. J. Physiol. (Lond.) 388, 323–347 (1987). 89. Iwasa, K. H. & Adachi, M. Force generation in the outer Acknowledgements Detailed description of the voltage-dependent hair cell of the cochlea. Biophys. J. 73, 546–555 This work was supported by a grant from the National somatic contractility of isolated OHCs and (1997). Institute on Deafness and Other Communication Disorders, evidence for a novel mechanism resident in the cell 90. Dallos, P. Neurobiology of cochlear inner and outer National Institutes of Health, USA. We thank Y. Katori and cortex. hair cells: intracellular recordings. Hear. Res. 22, D. Furness for the electron micrograph in figure 3b, and 73. Zheng, J. et al. Prestin is the motor protein of 185–198 (1986). C. Dizack for help with the artwork. cochlear outer hair cells. Nature 405, 149–155 91. Adler, H. J. et al. Expression of prestin, a membrane (2000). motor protein, in the mammalian auditory and Competing interests statement Cloning of prestin by subtractive hybridization of vestibular periphery. Hear. Res. 184, 27–40 (2003). The authors declare no competing financial interests. cDNA pools from OHCs relative to IHCs. Showed 92. He, D. Z. et al. Chick hair cells do not exhibit voltage- that prestin bestows voltage-dependent motility dependent somatic motility. J. Physiol. (Lond.) 546, DATABASES when expressed heterologously. 511–520 (2003). The following terms in this article are linked online to: 74. Belyantseva, I. A., Adler, H. J., Curi, R., Frolenkov, G. I. 93. Kennedy, H. J., Crawford, A. C. & Fettiplace, R. Force Entrez Gene: http://www.ncbi.nlm.nih.gov/entrez/query. & Kachar, B. Expression and localization of prestin and generation by mammalian hair bundles supports a role fcgi?db=gene the sugar transporter GLUT-5 during development of in cochlear amplification. Nature 433, 880–883 cadherin 23 | harmonin | myosin 1c | myosin VIIa | myosin XVa | electromotility in cochlear outer hair cells. J. Neurosci. (2005). prestin | protocadherin 15 | SANS | TRPA1 20, RC116 (2000). 94. Chan, D. K. & Hudspeth, A. J. Ca2+-current driven 75. Rybalchenko, V. & Santos-Sacchi, J. Cl– flux through a non-linear amplification by the mammalian cochlea FURTHER INFORMATION non-selective, stretch-sensitive conductance influences in vitro. Nature Neurosci. 8, 149–155 (2005). Fettiplace’s homepage: http://www.physiology.wisc.edu/ the outer hair cell motor of the guinea pig. J. Physiol. Imaging of IHC bundles in an in vitro cochlear faculty/fettiplace.html (Lond.) 547, 873–891 (2003). preparation to show mechanical amplification Hackney’s homepage: http://www.keele.ac.uk/depts/co/ 76. Oliver, D. et al. Intracellular anions as the voltage dependent on MET channel function. staff/chackney.html sensor of prestin, the outer hair cell motor protein. 95. Kim, D. O. Active and nonlinear cochlear biomechanics Hereditary Hearing Loss Homepage: http://webhost.ua.ac. Science 292, 2340–2343 (2001). and the role of outer-hair-cell subsystem in the be/hhh/ 77. Liberman, M. C. et al. Prestin is required for mammalian auditory system. Hear. Res. 22, 105–114 Access to this interactive links box is free online. electromotility of the outer hair cell and for the (1986).

NATURE REVIEWS | NEUROSCIENCE VOLUME 7 | JANUARY 2006 | 29 © 2006 Nature Publishing Group