Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Mar 1.
Published in final edited form as: Hear Res. 2013 Jan 28;297:106–112. doi: 10.1016/j.heares.2013.01.017

Prospects for Replacement of Auditory Neurons by Stem Cells

Fuxin Shi 1,2, Albert SB Edge 1,2,3
PMCID: PMC3594553  NIHMSID: NIHMS440005  PMID: 23370457

Abstract

Sensorineural hearing loss is caused by degeneration of hair cells or auditory neurons. Spiral ganglion cells, the primary afferent neurons of the auditory system, are patterned during development and send out projections to hair cells and to the brainstem under the control of largely unknown guidance molecules. The neurons do not regenerate after loss and even damage to their projections tends to be permanent. The genesis of spiral ganglion neurons and their synapses forms a basis for regenerative approaches. In this review we critically present the current experimental findings on auditory neuron replacement. We discuss the latest advances with a focus on (a) exogenous stem cell transplantation into the cochlea for neural replacement, (b) expression of local guidance signals in the cochlea after loss of auditory neurons, (c) the possibility of neural replacement from an endogenous cell source, and (d) functional changes from cell engraftment.

1.1 Lack of spontaneous regeneration of the auditory nerve

The auditory nerve does not regenerate once damaged. There have been some reports that peripheral fibers could be repaired when the synapses had been destroyed by a mechanism identified as excess glutamate at the afferent endings in adult animals (Lerner-Natoli et al., 1997; Puel et al., 1997), but other mechanisms such as partial repair of damaged hair cells could account for recovery. There was no evidence of regenerated fibers or synapses in careful studies that quantified synaptic contacts after the fibers had retracted in models of noise damage (Kujawa and Liberman, 2009; Lin et al., 2011), and thus synaptogenesis with hair cells does not appear to be a mechanism for recovery, and after loss of fibers or of the neurons themselves, spontaneous regenerative mechanisms are lacking. Only in an in vitro model of newborn rat cochlear explants has there been any evidence that peripheral fibers could spontaneously regrow to hair cells (Wang and Green, 2011).

In our work using embryonic stem (ES) cells and spiral ganglion neurons (SGNs) from newborn mice we have conclusively established growth of fibers that make synapses with hair cells and contact neurons in the brainstem (Corrales et al., 2006; Martinez-Monedero et al., 2006; Shi et al., 2007; Martinez-Monedero et al., 2008; Brugeaud and Edge, 2009; Tong et al., 2011).

1.2. Characteristics of the auditory nerve

The auditory nerve conveys sound information rapidly to the brain and covers a wide range of sound levels and frequencies. Spontaneous firing of afferent neurons is thought to set a baseline that can then be perturbed to obtain an accurate estimate of onset of a sound (Liberman, 1982; Kawase and Liberman, 1992). Firing in response to single quanta from hair cells is thought to make the nerve particularly sensitive (Rutherford et al., 2012). A wide dynamic range is achieved by the sensitivity of hair cells combined with the frequency coding of single fibers in the auditory nerve (Liberman, 1982).

The timing, frequency and intensity of sounds are coded and transmitted from hair cells to SGNs via the ribbon synapse which promotes rapid, precise and sustained neurotransmitter release and signal transmission through multivesicular release and calcium channel positioning (Moser et al., 2006). These unique features are critical for transmitting the full complexity of sound. The inner hair cells transmit information to the brain through the primary afferent synapse while outer hair cells increase sensitivity of sound detection by amplifying the signal.

1.3. Development of afferent innervation

Understanding the genesis of SGNs and their afferent synapses is crucial for the development of regenerative approaches. SGNs are among the first neurons to be specified during development. As early as mouse embryonic day 9 (E9) neuroblasts delaminate from the otocyst to form the cochleovestibular ganglion (Carney and Silver, 1983). Initially, the delaminated neuroblasts consist entirely of proliferating progenitor cells, and the onset of neurogenesis is marked by the appearance of the first post-mitotic vestibular and spiral ganglion cells. Both the spiral ganglion, which coils along the length of the cochlear duct, and the vestibular ganglion are formed from the cochleovestibular ganglion. The proliferation phase ends at approximately E9.5 to E13.5 in the mouse and is followed by differentiation in a direction from base at E9.5 to E10.5 to apex at E12.5 to E13.5 (Matei et al., 2005; Koundakjian et al., 2007). SGNs are born 4 days earlier than hair cells at the base of the cochlea. Most SGNs generate action potentials by E14, 2 days after completion of neurogenesis (Marrs and Spirou, 2012). The SGNs form synapses throughout development continuing up to postnatal day 12 (P12).

The molecular cues that control the specification of auditory neurons have been largely attributed to a network of basic helix-loop-helix (bHLH) transcription factors. Neurogenin 1 (Ngn1) has a governing role in defining the early neural precursors within the Sox2 expressing sensory domain in the otocyst (Ma et al., 1998; Ma et al., 2000). Subsequently, NeuroD is activated in post-mitotic progenitors to promote neuronal differentiation by inhibiting non- neuronal fate (Liu et al., 2000; Kim et al., 2001; Lawoko-Kerali et al., 2004). Intriguingly, spiral ganglion cell fate is dependent as well on GATA3 which acts upstream of NeuroD (Karis et al., 2001; Lawoko-Kerali et al., 2004). The choice between sensory hair cell and neuronal fate by inner ear stem cells is also dependent on Notch signaling, and Sox2 acts upstream of Ngn1 (Jeon et al., 2011).

1.4. Potential for regeneration by manipulation of axonal guidance

After their genesis, SGNs project to hair cells and to cells in the cochlear nucleus. Their growth is guided by molecules such as netrins, semaphorins, slits and ephrins that exert repulsive and attractive forces on growth cones at the tips of axons and dendrites and are important for the development of the auditory circuitry. Axonal growth can also be guided by permissive substrates in the matrix and by soluble trophic factors. Soon after SGNs are born, they become bipolar and are guided to their targets through the mesenchyme and Kolliker’s organ. Thus, guidance information likely comes from surrounding tissues, such as Kolliker’s organ, mesenchyme, and glia. Indeed, the repulsive slit and ephrin ligands are expressed in non-sensory tissues of the ear and function to keep axonal growth within the spiral lamina (Bianchi and Liu, 1999; Pickles et al., 2002; Battisti and Fekete, 2008). Extracellular matrix components within the mesenchyme provide permissive substrates, such as, integrins, laminin, and tenascin-C, to guide axons through the spiral lamina (Whitlon et al., 1999; Rodgers et al., 2001; Davies, 2007) before reaching hair cells. Moreover, netrin1 promotes neurite outgrowth of cultured SGNs, while ephrinB1 inhibits outgrowth (Bianchi and Gray, 2002; Lee and Warchol, 2008). Despite the presence of many host molecules in the inner ear, specific functions remain unclear. BDNF and NT3, neurotrophins expressed in the ear, may have some guidance activity, but that role is hard to dissect from the overall role in neuronal growth, survival and innervation (Fritzsch et al., 2004). Neurotrophins may control pathfinding by regulating the expression of other guidance cues.

We have initiated studies to evaluate the role of axonal guidance molecules in regeneration, and have found a negative influence of some axonal guidance molecules on SGN regeneration (Brugeaud and Edge, 2009). Manipulation of developmentally relevant guidance molecules will be important for rewiring of auditory circuitry with in vitro-derived neurons. There is some precedent for the inhibition of neuronal regeneration in the nervous system of adults by axonal guidance molecules that guide correct innervation in the embryo (Pasterkamp et al., 1998; Harel and Strittmatter, 2006). Repulsive guidance molecules do not recapitulate their role in embryonic guidance but, rather, may act aberrantly to inhibit regeneration at glial scars (Pasterkamp et al., 1998; Hata et al., 2006; Pasterkamp and Verhaagen, 2006). The neuropilin 1/semaphorin 3a and neogenin 1/RGMa receptors and ligands have been shown to prevent regeneration of axons in peripheral neurites (Kyoto et al., 2007; Tang et al., 2007; Tannemaat et al., 2007). Expression of these signaling pathways in the tissue of the adult after damage may inhibit SGN regeneration.

1.5. Regeneration of afferent synapses

The formation of a synapse requires initial contact followed by stabilization and acquisition of appropriate pre-and postsynaptic elements. The onset of synaptogenesis occurs prior to birth in mice, and is not dependent on synaptic activity. Action potentials can be triggered in the most immature SGNs by activation of hair cells at the apex of the cochlea in newborn rats (Tritsch and Bergles, 2010). Non-sensory cells play an essential role in the development of central auditory pathways. Supporting cells in organ of Corti initiate electrical activity in auditory nerves before hearing through spontaneous ATP release. This experience-independent activity subsides after the onset of hearing, preventing interference with accurate encoding of sound (Tritsch et al., 2007). Afferent synaptic-like contacts were described at E18 in rats (Huang et al., 2005) and ribbon synapses with apposed postsynaptic densities were present at birth in mice (Shnerson et al., 1981; Sobkowicz et al., 1982). Maturation is not complete until P12 at the onset of hearing (Grant et al., 2010). At the morphological level, ribbon synapses are made in two steps, formation of the presynaptic complex and assembly of the postsynaptic density (Sobkowicz et al., 1986). This sequence of events is typical for establishment of a synapse, where postsynaptic specializations are later to develop (Goda and Davis, 2003). Although there is a prepatterning of rudimentary postsynaptic organization prior to the arrival of the nerve, differentiation of the postsynaptic specialization is induced upon contact with the presynaptic ending. The postsynaptic specialization then influences assembly of the presynaptic active zone and its alignment with the postsynaptic specialization. Similarly, after afferent nerve fibers reach the hair cells, synaptogenesis begins with accumulation of dense-core and sometimes clear vesicles at the hair cell membrane apposed to the nerve fiber. These vesicles are probably active zone precursors, which are found in other systems and deliver presynaptic proteins to the nascent synapses. Neurotransmitters, secreted signaling proteins such as Wnts, neurotrophins, and adhesion molecules provide the anterograde signals in the CNS (Goda and Davis, 2003). We have developed a system for studying the mechanisms of synaptogenesis in the postnatal animal (Tong et al., 2011), and the elucidation of factors affecting postnatal synaptogenesis can be informed by developmental mechanisms.

Type I SGNs in the mouse comprise 90% of the neurons in the ganglion and convey most of the sound information. Each type I neuron contacts only one hair cell, while each inner hair cell is connected with 10–20 type I neurons (Liberman, 1980; Meyer et al., 2009). The smaller type II neurons, which make up less that 10% of SGNs (Romand and Romand, 1987), contact multiple outer hair cells after passing the inner hair cells and turning towards the base of the cochlea. SGNs are pruned, resulting in type I contacts with inner, and type II contacts with outer hair cells (Echteler, 1992). At E12.5, peripheral projections of SGNs are branched and tangled, and pruning is not apparent until E15.5 when single unbranched fibers are seen in the base of the cochlea (Koundakjian et al., 2007). The mechanisms for pruning are not clear. The first contact of a projection with a hair cell may inhibit the progress of other branches and, in turn, induce their retraction. Our in vitro models of neural replacement provide systems for investigating mechanisms that determine synaptogenesis and pruning of type I and II SGNs.

1.6. Central connections: brainstem

Information is conveyed to the cochlear nucleus by axons of the SGNs, which bundle together to form the eighth nerve. The central projections of SGNs first reach the hindbrain around E12.5 in the mouse, as the SGNs are born, and precede their peripheral projections (Karis et al., 2001; Maklad et al., 2010; Lu et al., 2011). This process does not appear to depend on the presence of target neurons in the cochlear nucleus, as SGNs project into the hindbrain and bifurcate even in Atoh1 knockout animals, in which the majority of target neurons are never formed (Maricich et al., 2009). SGNs exhibit spontaneous activity by E14 and are able to drive third-order auditory brainstem neurons by E17 (Marrs and Spirou, 2012). SGNs acquire action potential-generating capacity at E14 prior to neurons of the ventral and medial nucleus of the trapezoid body (VCN and MNTB). Accordingly, auditory nerve synapses in the VCN are functional at E15, prior to VCN connectivity with the MNTB, which occurs at least 1 day later. This activity precedes cochlear-generated wave activity by 4 days and ear canal opening by at least 2 weeks (Marrs and Spirou, 2012).

Upon entering the brainstem, individual axons bifurcate and form two branches, one to the VCN and the other to the dorsal cochlear nucleus (DCN) (Fekete et al., 1984; Ryugo, 2008). Each branch independently seeks tonotopically appropriate postsynaptic targets and elaborates synapses, forming the large endbulbs of Held with spherical bushy cells located in the most anterior portion of the VCN and smaller, modified endbulbs with globular bushy cells (Ryugo and Parks, 2003; Ryugo et al., 2006) and boutons in the DCN. Type II neurons project to the small cell cap surrounding the cochlear nucleus. Endbulbs have a calyx-like appearance in which the main axon elaborates several branches that arborize repeatedly to enclose the postsynaptic cell in a nest of en passant swellings and terminals (Ryugo and Fekete, 1982). High frequency information from the base of the cochlea is sent dorsally while low frequency information is processed ventrally. Although each of these stages has been documented at the cellular level, our knowledge of the underlying mechanisms remains limited. The unusual size and shape of the endbulb of Held is essential for rapid and secure synaptic transmission that allows detection of timing differences in auditory stimuli. The central auditory synapses display greater plasticity than the periphery, as shown by rescue of endbulbs in deaf cats given cochlear implants (Ryugo et al., 2005).

The progress from homogeneous neuroblasts to fully functional bipolar adult SGNs involves cell fate determination, pathfinding and synaptogenesis. Regeneration of these neurons and synapses will require that this complex series of steps be recapitulated, including the choice of type I or II; synapse formation with IHC or OHC; bifurcation in the cochlear nucleus; and synaptogenesis with target neurons in the brainstem. A detailed understanding of the molecular cues for differentiation and pathfinding will help to improve the efficiency of transplantation in vivo.

2.1. Loss of neurons (auditory neuropathy)

Neural degeneration has been thought to occur as a secondary consequence of hair cell loss but can also result from primary damage (Spoendlin, 1971; Liberman and M.J., 1982; Robertson, 1983; Puel et al., 1998; Hakuba et al., 2000; Wang et al., 2003). Damage to SGNs is an important clinical issue that has received increased attention because of recent work in which noise exposure caused primary afferent degeneration (Kujawa and Liberman, 2009). Loss of afferent synapses and retraction of fibers was seen after a 105 dB exposure for 2 hours and ultimately resulted in loss of neurons. No loss of hair cells was seen in this model. The low spontaneous rate, high-threshold neurons were more susceptible to degeneration; their loss resulted in a profound reduction in the threshold and wave 1-amplitude of the ABR. Accelerated loss of primary afferents was seen with age despite the recovery of threshold. Slow primary loss of neurons after recovery from the initial damage overturns a long-held view that recovery from a temporary threshold shift signals a restoration of normal cochlear function after noise-induced hearing loss. Temporary threshold shifts can be a harbinger of future problems due to neural degeneration, and neural loss without hair cell loss has been observed in human autopsy specimens (Makary et al., 2011).

A second model of auditory neuropathy is the ouabain-treated gerbil, in which the type I afferents are removed by round window administration of ouabain, a Na-K ATPase binding toxin (Schmiedt et al., 2002; Lang et al., 2005). The model is largely devoid of type I neurons in our hands, although there are occasional neurons surviving (Corrales et al., 2006). The ABR reflects this loss with thresholds above system output at most frequencies and hair cells are intact as measured by DPOAE (Lang et al., 2005).

2.2. Replacement of afferent neurons from resident cells

One route towards functional recovery is the replacement of SGNs from endogenous cell sources. Replacement of the neurons by regeneration as opposed to replacement by cell transplantation has the advantage that it would use normal mechanisms of peripheral regeneration and avoids the risk of teratomas, but it will require a better knowledge of the cells and signaling pathways that induce synaptogenesis. Unlike evolutionarily older animals, such as fish and birds, mammalian hair cells and SGNs do not exhibit significant regenerative capacity. However, several groups have demonstrated that proliferative cells, which display some of the hallmarks associated with neural stem cells, can be isolated from the organ of Corti. Most recently, we identified cochlear stem cells as Lgr5-expressing supporting cells (Shi et al., 2012). Cells isolated from Rosenthal’s canal also showed stem cell properties and gave rise to neurons (Martinez-Monedero et al., 2008; Kempfle et al., 2010) with electrophysiological properties of SGNs. BMP signaling increased sensory neuronal differentiation from ES cell-derived neural progenitors (Shi et al., 2007) and also influenced neural differentiation from neurospheres obtained from spiral ganglion (Kempfle et al., 2010).

Regrowth of peripheral fibers and contacts with hair cells after glutamate damage to primary afferent neurons have been demonstrated in vitro (Wang and Green, 2011). Hair cells in organ of Corti explants were de-afferented over the course of several hours but fibers regrew and contacted hair cells. Neurotrophins had a marked effect on fiber regrowth.

One approach would be to stimulate expansion and differentiation of endogenous progenitor cells with a drug or paracrine factors. Neural differentiation might also be achieved by factors that induce neural fates from terminally differentiated cells (Vierbuchen et al., 2010; Caiazzo et al., 2011). Among the factors that have been used to induce a neural phenotype are bHLH transcription factors, including Ascl1 (Vierbuchen et al., 2010; Caiazzo et al., 2011), and conversion to neurons has been demonstrated for fibroblasts as well as glial cells (Heinrich et al., 2011).

The spiral ganglion contains myelinating Schwann cells and satellite cells as well as fibroblasts that might be a source of new neurons for repair (Eybalin et al., 1996). The peripheral nervous system utilizes Schwann cells, myelinating glial cells and macrophages for regenerative support and clearance of extracellular matrix debris, creating an environment conducive to axonal regrowth at a rate of 1.5–2.0 mm/day (Buchthal and Kuhl, 1979; Rotshenker, 2011). Schwann cells proliferate after loss of SGNs, but don’t spontaneously differentiate into neurons (Lang et al., 2011).

2.3. Replacement of afferent neurons from exogenous stem cells

Exogenous replacement of SGNs is a second approach to cell replacement after SGN death. This approach has been widely investigated in CNS diseases, such as Huntington’s and Parkinson’s and spinal cord injury (Lunn et al., 2011). Cell transplantation in the CNS is made easier by a low rate of immunological rejection. The inner ear has a degree of immune privilege that may help to reduce the rejection of transplanted cells (Harris and Ryan, 1995; Bodmer et al., 2002). Another advantage of the auditory system for the study of neuronal replacement is that a single primary afferent neuron connects the sensory receptor cell to the brain and that we have physiological metrics that can be applied in conjunction with histological tools for determination of cochlear structure and function.

The environment influences the function of transplanted cells, and one issue to be overcome in cell transplantation is the difference in environment compared to that during development. Newborn neurons in adult hippocampus and olfactory bulb exhibit hyperexcitability and enhanced synaptic plasticity at glutamatergic inputs during a critical period of maturation (Tashiro et al., 2006). One of the best-studied mediators of stem cell tropism is the G-protein coupled receptor CXCR4 and its only known ligand SDF-1, which promote chain migration, the principal mode of stem cell movement in the mature brain (Imitola et al., 2004). Stem cells may undergo tropism specific for pathology in several animal models of neurologic disease. Extrinsic factors drive transplanted neural stem cells to differentiate into neurons or glia (Gage, 2000). Both endogenous cells in the dentate gyrus and subventricular zone and cells engrafted into the brain have been observed to preferentially undergo neurogenesis when subjected to ischemia (Zhang et al., 2001; Chu et al., 2003), thus replacing the cells most susceptible to ischemic insult. Previous experiments have shown that the environment in the host cochlea is critical for transplanted stem cell survival, with greater engraftment seen at 1-3 days than at 7 days post-injury (Lang et al., 2008).

There are several sources available for exogenous replacement of SGNs. The adult inner ear stem cells are potentially a source of cells for transplantation. The identity of stem cells within the ganglion would be valuable information because these cells could then be enriched prior to use in cell replacement strategies. In addition, ES cells can be differentiated into neurons. ES cell-derived neural progenitors were induced to adopt a sensory fate by culture in BMP4 (Shi et al., 2007). ES cells expressing Ngn1 adopted an SGN-like fate after culture in BDNF and GDNF (Reyes et al., 2008). Induced pluripotent stem (iPS) cells (Takahashi and Yamanaka, 2006) have been used to generate auditory neural lineage by co-culturing with mouse cochlear explants (Nishimura et al., 2009). Mesenchymal stem cells (MSCs), adult stem cells of mesodermal derivation, can home to the deafened cochlea, although no spontaneous transdifferentiation to cochlear cell types occurred after acoustic trauma or bone marrow mobilization (Lang et al., 2006; Tan et al., 2008). MSCs are reported to elicit protective effects via secretion of neurotrophic factors (Crigler et al., 2006) and inflammatory modulators (Pluchino et al., 2005). Adipose-derived stem cells have similar traits as MSCs (Zhou et al., 2011).

ES cells could theoretically be transplanted at any stage of differentiation to neurons. In our work we have found that neural progenitors derived from ES cells could be obtained in vitro, and comparison to neural transplantation revealed that this was likely to be the optimal stage of transplantation to avoid apoptosis that is prevalent when differentiated neurons are transplanted in a suspension (Shi et al., 2007; Yuan et al., 2012). The same phenomenon has been observed in previous CNS work (Bjorklund et al., 2003). Although these neurons were at an intermediate stage at which they could become any type of neuron they were beyond differentiation stages at which endoderm or mesoderm would be generated, thus minimizing teratoma formation. A combination of in vitro methods to stimulate ectoderm and neural fate commitment combined with the local cues provided by the inner ear environment were found to be sufficient to convert the cells to differentiated neurons that responded to cues for neurite growth and synaptogenesis and were functionally active (Tokano et al., 2012; Yuan et al., 2012).

We have shown a regenerative capacity in neurons placed in proximity to hair cells in culture. Isolated SGNs from neonatal mice formed synaptic contacts with hair cells in organotypic cultures of organ of Corti (Martinez-Monedero et al., 2006). This was also seen with neurons differentiated from mouse (Corrales et al., 2006; Nayagam et al., 2012) or human (Shi et al., 2007) ES cells. SGNs contacted more than one hair cell in the initial cultures and then underwent pruning of branches (Martinez-Monedero et al., 2006; Martinez-Monedero and Edge, 2007). We also found that postsynaptic densities were induced in the fibers that contacted hair cells and were closely apposed to the points of contact with presynaptic ribbons (Tong et al., 2011). Since the efficiency of reinnervation in these cultures was low, we used the system as an in vitro synaptogenesis assay to assess the importance of various molecules for regeneration. The low efficiency could be overcome in part by inhibition of one of the molecules expressed in sensory epithelium, repulsive guidance molecule, RGMa (Brugeaud and Edge, 2009). RGMa inhibition increased sprouting and axonal growth and accelerated synaptic pruning (Brugeaud and Edge, 2009). This suggested that axonal growth and sprouting were intrinsic but could be influenced by guidance molecules from the surrounding cells. Neurotrophins, moreover, increased synaptogenesis of afferent neurons with hair cells in organ of Corti explant cultures, and glutamate played an important role in establishing the connections (Tong et al., 2012).

A number of groups have tested ES cells for SGN replacement in vivo (Okano et al., 2005; Coleman et al., 2006; Corrales et al., 2006; Sekiya et al., 2006; Shi et al., 2007; Lang et al., 2008; Praetorius et al., 2008; Reyes et al., 2008). In our studies ES cell-derived mouse neural progenitor cells transplanted into the cochlear nerve trunk sent out abundant processes that occupied a significant portion of the space formerly occupied by the cochlear nerve. A portion of the neurites grew in fasciculating bundles projecting through Rosenthal’s canal into the osseous spiral lamina and ultimately into the organ of Corti, where they contacted hair cells (Corrales et al., 2006). Reinnervation in the base of the cochlea led to partial restoration of ABR amplitude in gerbils (Tokano et al., 2012) and improved thresholds in the mouse (Yuan et al., 2012). The areas of synaptogenesis as assessed by histology correlated with the frequencies where improved function was apparent by measurement of ABR, confirming functional synaptogenesis.

To restore auditory circuitry, functional connections to hair cells and cochlear nucleus must be made. Reporters for tracking the donor cells are required to visualize the new synapses. Optimizing the surgical approach increases the chance of measuring an improvement in ABR. The placement of the transplanted cells is important for their projection (Coleman et al., 2006; Martinez-Monedero and Edge, 2007). Cells transplanted into Rosenthal’s canal and the auditory nerve trunk had more abundant projections to hair cells and cochlear nucleus than those placed in the scala tympani or scala media (Corrales et al., 2006; Shi et al., 2007; Yuan et al., 2012). The bony wall of the osseous spiral lamina can be a barrier to engraftment. The initial site for cell placement may also be critical, because neurites can be blocked when they encounter the Schwann-glial transitional zone (Sekiya et al., 2007). Growing axons from the peripheral nerve towards the spinal cord or brainstem abruptly stop when they encounter astrocytes in the CNS. Thus, neurites growing from cells transplanted to the cochlea may be inhibited when they reach the Schwann-glial transitional zone (Aldskogius and Kozlova, 2002).

ES cell-derived neural progenitors made contacts with vestibular hair cells (Kim et al., 2005), and ES cell transplantation into guinea pig cochlea resulted in growth of processes, although the processes did not appear to grow toward the organ of Corti (Okano et al., 2005). ES cells transplanted into the cochlear modiolus of animals with damaged SGNs grew fibers along the remaining neurons and to the organ of Corti (Sekiya et al., 2006). Neural stem cells have been used for transplantation into the cochlea of a mouse that had been treated with cisplatin to remove endogenous neurons, and the cells were present in the cochlea two weeks after grafting (Tamura et al., 2004).

The implanted cells would not have much clinical significance unless central connections could be established. In a co-culture system with the brainstem, ES cells survived, underwent neuronal differentiation, and migrated towards the cochlear nucleus (Thonabulsombat et al., 2007). We have found the neural progenitors derived from ES cells transplanted into the auditory nerve trunk sent extensive process towards the cochlear nucleus (Shi et al., 2007; Yuan et al., 2012).

The differentiation of ES cell-derived progenitors to neurons with the properties of SGNs, and our ability to transplant cells into the deafened cochlea followed by restoration of the neural circuitry is an exciting and significant step toward the ultimate goal of improved hearing. Whether the transplanted cells integrate is dependent upon the cell source, the site of engraftment, and the differentiation to physiologically functional neurons. Testing of molecules in the in vitro system provides a route to new methods to increase synaptogenesis.

Acknowledgements

This work was supported by grants RO1 DC007174 and P30 DC05209 from the National Institute on Deafness and other Communicative Disorders.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Aldskogius H, Kozlova EN. Strategies for repair of the deafferented spinal cord. Brain Res Brain Res Rev. 2002;40:301–308. doi: 10.1016/s0165-0173(02)00212-6. [DOI] [PubMed] [Google Scholar]
  2. Battisti AC, Fekete DM. Slits and Robos in the developing chicken inner ear. Dev Dyn. 2008;237:476–484. doi: 10.1002/dvdy.21429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Bianchi LM, Liu H. Comparison of ephrin-A ligand and EphA receptor distribution in the developing inner ear. Anat Rec. 1999;254:127–134. doi: 10.1002/(SICI)1097-0185(19990101)254:1<127::AID-AR16>3.0.CO;2-Q. [DOI] [PubMed] [Google Scholar]
  4. Bianchi LM, Gray NA. EphB receptors influence growth of ephrin-B1-positive statoacoustic nerve fibers. Eur J Neurosci. 2002;16:1499–1506. doi: 10.1046/j.1460-9568.2002.02248.x. [DOI] [PubMed] [Google Scholar]
  5. Bjorklund A, Dunnett SB, Brundin P, Stoessl AJ, Freed CR, Breeze RE, Levivier M, Peschanski M, Studer L, Barker R. Neural transplantation for the treatment of Parkinson’s disease. Lancet Neurol. 2003;2:437–445. doi: 10.1016/s1474-4422(03)00442-3. [DOI] [PubMed] [Google Scholar]
  6. Bodmer D, Brors D, Pak K, Keithley EM, Mullen L, Ryan AF, Gloddek B. Inflammatory signals increase Fas ligand expression by inner ear cells. J Neuroimmunol. 2002;129:10–17. doi: 10.1016/s0165-5728(02)00143-1. [DOI] [PubMed] [Google Scholar]
  7. Brugeaud A, Edge ASB. Characterization of in vitro regenerated synapses between auditory neurons and hair cells. ARO 32nd Annual Meeting Abstract #786. 2009 [Google Scholar]
  8. Buchthal F, Kuhl V. Nerve conduction, tactile sensibility, and the electromyogram after suture or compression of peripheral nerve: a longitudinal study in man. J Neurol Neurosurg Psychiatry. 1979;42:436–451. doi: 10.1136/jnnp.42.5.436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Caiazzo M, Dell’Anno MT, Dvoretskova E, Lazarevic D, Taverna S, Leo D, Sotnikova TD, Menegon A, Roncaglia P, Colciago G, Russo G, Carninci P, Pezzoli G, Gainetdinov RR, Gustincich S, Dityatev A, Broccoli V. Direct generation of functional dopaminergic neurons from mouse and human fibroblasts. Nature. 2011;476:224–227. doi: 10.1038/nature10284. [DOI] [PubMed] [Google Scholar]
  10. Carney PR, Silver J. Studies on cell migration and axon guidance in the developing distal auditory system of the mouse. J Comp Neurol. 1983;215:359–369. doi: 10.1002/cne.902150402. [DOI] [PubMed] [Google Scholar]
  11. Chu K, Kim M, Jeong SW, Kim SU, Yoon BW. Human neural stem cells can migrate, differentiate, and integrate after intravenous transplantation in adult rats with transient forebrain ischemia. Neurosci Lett. 2003;343:129–133. doi: 10.1016/s0304-3940(03)00174-5. [DOI] [PubMed] [Google Scholar]
  12. Coleman B, Hardman J, Coco A, Epp S, de Silva M, Crook J, Shepherd R. Fate of embryonic stem cells transplanted into the deafened mammalian cochlea. Cell Transplant. 2006;15:369–380. doi: 10.3727/000000006783981819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Corrales CE, Pan L, Li H, Liberman MC, Heller S, Edge AS. Engraftment and differentiation of embryonic stem cell-derived neural progenitor cells in the cochlear nerve trunk: Growth of processes into the organ of corti. J Neurobiol. 2006;66:1489–1500. doi: 10.1002/neu.20310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Crigler L, Robey RC, Asawachaicharn A, Gaupp D, Phinney DG. Human mesenchymal stem cell subpopulations express a variety of neuro-regulatory molecules and promote neuronal cell survival and neuritogenesis. Exp Neurol. 2006;198:54–64. doi: 10.1016/j.expneurol.2005.10.029. [DOI] [PubMed] [Google Scholar]
  15. Davies D. Temporal and spatial regulation of alpha6 integrin expression during the development of the cochlear-vestibular ganglion. J Comp Neurol. 2007;502:673–682. doi: 10.1002/cne.21302. [DOI] [PubMed] [Google Scholar]
  16. Echteler SM. Developmental segregation in the afferent projections to mammalian auditory hair cells. Proc Natl Acad Sci U S A. 1992;89:6324–6327. doi: 10.1073/pnas.89.14.6324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Eybalin M, Norenberg MD, Renard N. Glutamine synthetase and glutamate metabolism in the guinea pig cochlea. Hear Res. 1996;101:93–101. doi: 10.1016/s0378-5955(96)00136-0. [DOI] [PubMed] [Google Scholar]
  18. Fekete DM, Rouiller EM, Liberman MC, Ryugo DK. The central projections of intracellularly labeled auditory nerve fibers in cats. J Comp Neurol. 1984;229:432–450. doi: 10.1002/cne.902290311. [DOI] [PubMed] [Google Scholar]
  19. Fritzsch B, Tessarollo L, Coppola E, Reichardt LF. Neurotrophins in the ear: their roles in sensory neuron survival and fiber guidance. Prog Brain Res. 2004;146:265–278. doi: 10.1016/S0079-6123(03)46017-2. [DOI] [PubMed] [Google Scholar]
  20. Gage FH. Mammalian neural stem cells. Science. 2000;287:1433–1438. doi: 10.1126/science.287.5457.1433. [DOI] [PubMed] [Google Scholar]
  21. Goda Y, Davis GW. Mechanisms of synapse assembly and disassembly. Neuron. 2003;40:243–264. doi: 10.1016/s0896-6273(03)00608-1. [DOI] [PubMed] [Google Scholar]
  22. Grant L, Yi E, Glowatzki E. Two modes of release shape the postsynaptic response at the inner hair cell ribbon synapse. J Neurosci. 2010;30:4210–4220. doi: 10.1523/JNEUROSCI.4439-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Hakuba N, Koga K, Gyo K, Usami SI, Tanaka K. Exacerbation of noise-induced hearing loss in mice lacking the glutamate transporter GLAST. J Neurosci. 2000;20:8750–8753. doi: 10.1523/JNEUROSCI.20-23-08750.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Harel NY, Strittmatter SM. Can regenerating axons recapitulate developmental guidance during recovery from spinal cord injury? Nat Rev Neurosci. 2006;7:603–616. doi: 10.1038/nrn1957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Harris JP, Ryan AF. Fundamental immune mechanisms of the brain and inner ear. Otolaryngol Head Neck Surg. 1995;112:639–653. doi: 10.1016/s0194-5998(95)70170-2. [DOI] [PubMed] [Google Scholar]
  26. Hata K, Fujitani M, Yasuda Y, Doya H, Saito T, Yamagishi S, Mueller BK, Yamashita T. RGMa inhibition promotes axonal growth and recovery after spinal cord injury. J Cell Biol. 2006;173:47–58. doi: 10.1083/jcb.200508143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Heinrich C, Gascon S, Masserdotti G, Lepier A, Sanchez R, Simon-Ebert T, Schroeder T, Gotz M, Berninger B. Generation of subtype-specific neurons from postnatal astroglia of the mouse cerebral cortex. Nat Protoc. 2011;6:214–228. doi: 10.1038/nprot.2010.188. [DOI] [PubMed] [Google Scholar]
  28. Huang LC, Greenwood D, Thorne PR, Housley GD. Developmental regulation of neuron-specific P2X3 receptor expression in the rat cochlea. J Comp Neurol. 2005;484:133–143. doi: 10.1002/cne.20442. [DOI] [PubMed] [Google Scholar]
  29. Imitola J, Comabella M, Chandraker AK, Dangond F, Sayegh MH, Snyder EY, Khoury SJ. Neural stem/progenitor cells express costimulatory molecules that are differentially regulated by inflammatory and apoptotic stimuli. Am J Pathol. 2004;164:1615–1625. doi: 10.1016/S0002-9440(10)63720-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Jeon SJ, Fujioka M, Kim SC, Edge ASB. Notch signaling alters sensory or neuronal cell fate specification of inner ear stem cells. J Neurosci. 2011;31:8351–8358. doi: 10.1523/JNEUROSCI.6366-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Karis A, Pata I, van Doorninck JH, Grosveld F, de Zeeuw CI, de Caprona D, Fritzsch B. Transcription factor GATA-3 alters pathway selection of olivocochlear neurons and affects morphogenesis of the ear. J Comp Neurol. 2001;429:615–630. doi: 10.1002/1096-9861(20010122)429:4<615::aid-cne8>3.0.co;2-f. [DOI] [PubMed] [Google Scholar]
  32. Kawase T, Liberman MC. Spatial organization of the auditory nerve according to spontaneous discharge rate. J Comp Neurol. 1992;319:312–318. doi: 10.1002/cne.903190210. [DOI] [PubMed] [Google Scholar]
  33. Kempfle J, Shi F, Edge A. Reciprocal Effect of BMP4 on Differentiation of Hair Cells and Neurons from Inner Ear Stem Cells. ARO 33rd Annual Meeting Abstract #18. 2010 [Google Scholar]
  34. Kim TS, Nakagawa T, Kita T, Higashi T, Takebayashi S, Matsumoto M, Kojima K, Sakamoto T, Ito J. Neural connections between embryonic stem cell-derived neurons and vestibular hair cells in vitro. Brain Res. 2005;1057:127–133. doi: 10.1016/j.brainres.2005.07.059. [DOI] [PubMed] [Google Scholar]
  35. Kim WY, Fritzsch B, Serls A, Bakel LA, Huang EJ, Reichardt LF, Barth DS, Lee JE. NeuroD-null mice are deaf due to a severe loss of the inner ear sensory neurons during development. Development. 2001;128:417–426. doi: 10.1242/dev.128.3.417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Koundakjian EJ, Appler JL, Goodrich LV. Auditory neurons make stereotyped wiring decisions before maturation of their targets. J Neurosci. 2007;27:14078–14088. doi: 10.1523/JNEUROSCI.3765-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Kujawa SG, Liberman MC. Adding insult to injury: cochlear nerve degeneration after “temporary” noise-induced hearing loss. J Neurosci. 2009;29:14077–14085. doi: 10.1523/JNEUROSCI.2845-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Kyoto A, Hata K, Yamashita T. Synapse formation of the cortico-spinal axons is enhanced by RGMa inhibition after spinal cord injury. Brain Res. 2007;1186:74–86. doi: 10.1016/j.brainres.2007.10.038. [DOI] [PubMed] [Google Scholar]
  39. Lang H, Schulte BA, Schmiedt RA. Ouabain induces apoptotic cell death in type I spiral ganglion neurons, but not type II neurons. J Assoc Res Otolaryngol. 2005;6:63–74. doi: 10.1007/s10162-004-5021-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Lang H, Schulte BA, Goddard JC, Hedrick M, Schulte JB, Wei L, Schmiedt RA. Transplantation of mouse embryonic stem cells into the cochlea of an auditory-neuropathy animal model: effects of timing after injury. J Assoc Res Otolaryngol. 2008;9:225–240. doi: 10.1007/s10162-008-0119-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Lang H, Li M, Kilpatrick LA, Zhu J, Samuvel DJ, Krug EL, Goddard JC. Sox2 up-regulation and glial cell proliferation following degeneration of spiral ganglion neurons in the adult mouse inner ear. J Assoc Res Otolaryngol. 2011;12:151–171. doi: 10.1007/s10162-010-0244-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Lang H, Ebihara Y, Schmiedt RA, Minamiguchi H, Zhou D, Smythe N, Liu L, Ogawa M, Schulte BA. Contribution of bone marrow hematopoietic stem cells to adult mouse inner ear: mesenchymal cells and fibrocytes. J Comp Neurol. 2006;496:187–201. doi: 10.1002/cne.20929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Lawoko-Kerali G, Rivolta MN, Lawlor P, Cacciabue-Rivolta DI, Langton-Hewer C, van Doorninck JH, Holley MC. GATA3 and NeuroD distinguish auditory and vestibular neurons during development of the mammalian inner ear. Mech Dev. 2004;121:287–299. doi: 10.1016/j.mod.2003.12.006. [DOI] [PubMed] [Google Scholar]
  44. Lee KH, Warchol ME. Promotion of neurite outgrowth and axon guidance in spiral ganglion cells by netrin-1. Arch Otolaryngol Head Neck Surg. 2008;134:146–151. doi: 10.1001/archoto.2007.6. [DOI] [PubMed] [Google Scholar]
  45. Lerner-Natoli M, Ladrech S, Renard N, Puel JL, Eybalin M, Pujol R. Protein kinase C may be involved in synaptic repair of auditory neuron dendrites after AMPA injury in the cochlea. Brain Res. 1997;749:109–119. doi: 10.1016/s0006-8993(96)01306-6. [DOI] [PubMed] [Google Scholar]
  46. Liberman MC. Morphological differences among radial afferent fibers in the cat cochlea: an electron-microscopic study of serial sections. Hear Res. 1980;3:45–63. doi: 10.1016/0378-5955(80)90007-6. [DOI] [PubMed] [Google Scholar]
  47. Liberman MC. Single-neuron labeling in the cat auditory nerve. Science. 1982;216:1239–1241. doi: 10.1126/science.7079757. [DOI] [PubMed] [Google Scholar]
  48. Liberman MC, M MJ. Hamernik RP, Henderson D, Salvi R, editors. Acute and chronic effects of acoustic trauma: cochlear pathology and auditory nerve pathophysiology. New perspectives on noise-induced hearing loss. 1982:105–151. [Google Scholar]
  49. Lin HW, Furman AC, Kujawa SG, Liberman MC. Primary neural degeneration in the Guinea pig cochlea after reversible noise-induced threshold shift. J Assoc Res Otolaryngol. 2011;12:605–616. doi: 10.1007/s10162-011-0277-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Liu M, Pereira FA, Price SD, Chu MJ, Shope C, Himes D, Eatock RA, Brownell WE, Lysakowski A, Tsai MJ. Essential role of BETA2/NeuroD1 in development of the vestibular and auditory systems. Genes Dev. 2000;14:2839–2854. doi: 10.1101/gad.840500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Lu CC, Appler JM, Houseman EA, Goodrich LV. Developmental profiling of spiral ganglion neurons reveals insights into auditory circuit assembly. J Neurosci. 2011;31:10903–10918. doi: 10.1523/JNEUROSCI.2358-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Lunn JS, Sakowski SA, Hur J, Feldman EL. Stem cell technology for neurodegenerative diseases. Ann Neurol. 2011;70:353–361. doi: 10.1002/ana.22487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Ma Q, Anderson DJ, Fritzsch B. Neurogenin 1 null mutant ears develop fewer, morphologically normal hair cells in smaller sensory epithelia devoid of innervation. J Assoc Res Otolaryngol. 2000;1:129–143. doi: 10.1007/s101620010017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Ma Q, Chen Z, del Barco Barrantes I, de la Pompa JL, Anderson DJ. neurogenin1 is essential for the determination of neuronal precursors for proximal cranial sensory ganglia. Neuron. 1998;20:469–482. doi: 10.1016/s0896-6273(00)80988-5. [DOI] [PubMed] [Google Scholar]
  55. Makary CA, Shin J, Kujawa SG, Liberman MC, Merchant SN. Age-related primary cochlear neuronal degeneration in human temporal bones. J Assoc Res Otolaryngol. 2011;12:711–717. doi: 10.1007/s10162-011-0283-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Maklad A, Kamel S, Wong E, Fritzsch B. Development and organization of polarity-specific segregation of primary vestibular afferent fibers in mice. Cell Tissue Res. 2010;340:303–321. doi: 10.1007/s00441-010-0944-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Maricich SM, Xia A, Mathes EL, Wang VY, Oghalai JS, Fritzsch B, Zoghbi HY. Atoh1-lineal neurons are required for hearing and for the survival of neurons in the spiral ganglion and brainstem accessory auditory nuclei. J Neurosci. 2009;29:11123–11133. doi: 10.1523/JNEUROSCI.2232-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Marrs GS, Spirou GA. Embryonic assembly of auditory circuits: spiral ganglion and brainstem. J Physiol. 2012;590:2391–2408. doi: 10.1113/jphysiol.2011.226886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Martinez-Monedero R, Edge AS. Stem cells for the replacement of inner ear neurons and hair cells. Int J Dev Biol. 2007;51:655–661. doi: 10.1387/ijdb.072372rm. [DOI] [PubMed] [Google Scholar]
  60. Martinez-Monedero R, Corrales CE, Cuajungco MP, Heller S, Edge AS. Reinnervation of hair cells by auditory neurons after selective removal of spiral ganglion neurons. J Neurobiol. 2006;66:319–331. doi: 10.1002/neu.20232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Martinez-Monedero R, Yi E, Oshima K, Glowatzki E, Edge AS. Differentiation of inner ear stem cells to functional sensory neurons. Dev Neurobiol. 2008;68:669–684. doi: 10.1002/dneu.20616. [DOI] [PubMed] [Google Scholar]
  62. Matei V, Pauley S, Kaing S, Rowitch D, Beisel KW, Morris K, Feng F, Jones K, Lee J, Fritzsch B. Smaller inner ear sensory epithelia in Neurog 1 null mice are related to earlier hair cell cycle exit. Dev Dyn. 2005;234:633–650. doi: 10.1002/dvdy.20551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Meyer AC, Frank T, Khimich D, Hoch G, Riedel D, Chapochnikov NM, Yarin YM, Harke B, Hell SW, Egner A, Moser T. Tuning of synapse number, structure and function in the cochlea. Nat Neurosci. 2009;12:444–453. doi: 10.1038/nn.2293. [DOI] [PubMed] [Google Scholar]
  64. Moser T, Neef A, Khimich D. Mechanisms underlying the temporal precision of sound coding at the inner hair cell ribbon synapse. J Physiol. 2006;576:55–62. doi: 10.1113/jphysiol.2006.114835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Nayagam BA, Edge AS, Needham K, Hyakumura T, Leung J, Nayagam DA, Dottori M. An In Vitro Model of Developmental Synaptogenesis Using Cocultures of Human Neural Progenitors and Cochlear Explants. Stem Cells Dev. 2012 doi: 10.1089/scd.2012.0082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Nishimura K, Nakagawa T, Ono K, Ogita H, Sakamoto T, Yamamoto N, Okita K, Yamanaka S, Ito J. Transplantation of mouse induced pluripotent stem cells into the cochlea. Neuroreport. 2009;20:1250–1254. doi: 10.1097/WNR.0b013e32832ff287. [DOI] [PubMed] [Google Scholar]
  67. Okano T, Nakagawa T, Endo T, Kim TS, Kita T, Tamura T, Matsumoto M, Ohno T, Sakamoto T, Iguchi F, Ito J. Engraftment of embryonic stem cell-derived neurons into the cochlear modiolus. Neuroreport. 2005;16:1919–1922. doi: 10.1097/01.wnr.0000187628.38010.5b. [DOI] [PubMed] [Google Scholar]
  68. Pasterkamp RJ, Verhaagen J. Semaphorins in axon regeneration: developmental guidance molecules gone wrong? Philos Trans R Soc Lond B Biol Sci. 2006;361:1499–1511. doi: 10.1098/rstb.2006.1892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Pasterkamp RJ, De Winter F, Holtmaat AJ, Verhaagen J. Evidence for a role of the chemorepellent semaphorin III and its receptor neuropilin-1 in the regeneration of primary olfactory axons. J Neurosci. 1998;18:9962–9976. doi: 10.1523/JNEUROSCI.18-23-09962.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Pickles JO, Claxton C, Van Heumen WR. Complementary and layered expression of Ephs and ephrins in developing mouse inner ear. J Comp Neurol. 2002;449:207–216. doi: 10.1002/cne.10231. [DOI] [PubMed] [Google Scholar]
  71. Pluchino S, Zanotti L, Rossi B, Brambilla E, Ottoboni L, Salani G, Martinello M, Cattalini A, Bergami A, Furlan R, Comi G, Constantin G, Martino G. Neurosphere-derived multipotent precursors promote neuroprotection by an immunomodulatory mechanism. Nature. 2005;436:266–271. doi: 10.1038/nature03889. [DOI] [PubMed] [Google Scholar]
  72. Praetorius M, Vicario I, Schimmang T. Efficient transfer of embryonic stem cells into the cochlea via a non-invasive vestibular route. Acta Otolaryngol. 2008;128:720–723. doi: 10.1080/00016480701714236. [DOI] [PubMed] [Google Scholar]
  73. Puel JL, Ruel J, Gervais d’Aldin C, Pujol R. Excitotoxicity and repair of cochlear synapses after noise-trauma induced hearing loss. Neuroreport. 1998;9:2109–2114. doi: 10.1097/00001756-199806220-00037. [DOI] [PubMed] [Google Scholar]
  74. Puel JL, d’Aldin C, Ruel J, Ladrech S, Pujol R. Synaptic repair mechanisms responsible for functional recovery in various cochlear pathologies. Acta Otolaryngol. 1997;117:214–218. doi: 10.3109/00016489709117773. [DOI] [PubMed] [Google Scholar]
  75. Reyes JH, O’Shea KS, Wys NL, Velkey JM, Prieskorn DM, Wesolowski K, Miller JM, Altschuler RA. Glutamatergic neuronal differentiation of mouse embryonic stem cells after transient expression of neurogenin 1 and treatment with BDNF and GDNF: in vitro and in vivo studies. J Neurosci. 2008;28:12622–12631. doi: 10.1523/JNEUROSCI.0563-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Robertson D. Functional significance of dendritic swelling after loud sounds in the guinea pig cochlea. Hear Res. 1983;9:263–278. doi: 10.1016/0378-5955(83)90031-x. [DOI] [PubMed] [Google Scholar]
  77. Rodgers KD, Barritt L, Miner JH, Cosgrove D. The laminins in the murine inner ear: developmental transitions and expression in cochlear basement membranes. Hear Res. 2001;158:39–50. doi: 10.1016/s0378-5955(01)00283-0. [DOI] [PubMed] [Google Scholar]
  78. Romand MR, Romand R. The ultrastructure of spiral ganglion cells in the mouse. Acta Otolaryngol. 1987;104:29–39. doi: 10.3109/00016488709109044. [DOI] [PubMed] [Google Scholar]
  79. Rotshenker S. Wallerian degeneration: the innate-immune response to traumatic nerve injury. J Neuroinflammation. 2011;8:109. doi: 10.1186/1742-2094-8-109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Rutherford MA, Chapochnikov NM, Moser T. Spike encoding of neurotransmitter release timing by spiral ganglion neurons of the cochlea. J Neurosci. 2012;32:4773–4789. doi: 10.1523/JNEUROSCI.4511-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Ryugo DK. Projections of low spontaneous rate, high threshold auditory nerve fibers to the small cell cap of the cochlear nucleus in cats. Neuroscience. 2008;154:114–126. doi: 10.1016/j.neuroscience.2007.10.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Ryugo DK, Fekete DM. Morphology of primary axosomatic endings in the anteroventral cochlear nucleus of the cat: a study of the endbulbs of Held. J Comp Neurol. 1982;210:239–257. doi: 10.1002/cne.902100304. [DOI] [PubMed] [Google Scholar]
  83. Ryugo DK, Parks TN. Primary innervation of the avian and mammalian cochlear nucleus. Brain Res Bull. 2003;60:435–456. doi: 10.1016/s0361-9230(03)00049-2. [DOI] [PubMed] [Google Scholar]
  84. Ryugo DK, Kretzmer EA, Niparko JK. Restoration of auditory nerve synapses in cats by cochlear implants. Science. 2005;310:1490–1492. doi: 10.1126/science.1119419. [DOI] [PubMed] [Google Scholar]
  85. Ryugo DK, Montey KL, Wright AL, Bennett ML, Pongstaporn T. Postnatal development of a large auditory nerve terminal: the endbulb of Held in cats. Hear Res. 2006;216-217:100–115. doi: 10.1016/j.heares.2006.01.007. [DOI] [PubMed] [Google Scholar]
  86. Schmiedt RA, Okamura HO, Lang H, Schulte BA. Ouabain application to the round window of the gerbil cochlea: a model of auditory neuropathy and apoptosis. J Assoc Res Otolaryngol. 2002;3:223–233. doi: 10.1007/s1016200220017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Sekiya T, Kojima K, Matsumoto M, Holley MC, Ito J. Rebuilding lost hearing using cell transplantation. Neurosurgery. 2007;60:417–433. doi: 10.1227/01.NEU.0000249189.46033.42. [DOI] [PubMed] [Google Scholar]
  88. Sekiya T, Kojima K, Matsumoto M, Kim TS, Tamura T, Ito J. Cell transplantation to the auditory nerve and cochlear duct. Exp Neurol. 2006;198:12–24. doi: 10.1016/j.expneurol.2005.11.006. [DOI] [PubMed] [Google Scholar]
  89. Shi F, Kempfle JS, Edge AS. Wnt-responsive lgr5-expressing stem cells are hair cell progenitors in the cochlea. J Neurosci. 2012;32:9639–9648. doi: 10.1523/JNEUROSCI.1064-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Shi F, Corrales CE, Liberman MC, Edge AS. BMP4 induction of sensory neurons from human embryonic stem cells and reinnervation of sensory epithelium. Eur J Neurosci. 2007;26:3016–3023. doi: 10.1111/j.1460-9568.2007.05909.x. [DOI] [PubMed] [Google Scholar]
  91. Shnerson A, Devigne C, Pujol R. Age-related changes in the C57BL/6J mouse cochlea. II. Ultrastructural findings. Brain Res. 1981;254:77–88. doi: 10.1016/0165-3806(81)90060-2. [DOI] [PubMed] [Google Scholar]
  92. Sobkowicz HM, Rose JE, Scott GE, Slapnick SM. Ribbon synapses in the developing intact and cultured organ of Corti in the mouse. J Neurosci. 1982;2:942–957. doi: 10.1523/JNEUROSCI.02-07-00942.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Sobkowicz HM, Rose JE, Scott GL, Levenick CV. Distribution of synaptic ribbons in the developing organ of Corti. J Neurocytol. 1986;15:693–714. doi: 10.1007/BF01625188. [DOI] [PubMed] [Google Scholar]
  94. Spoendlin H. Primary structural changes in the organ of Corti after acoustic overstimulation. Acta Otolaryngol. 1971;71:166–176. doi: 10.3109/00016487109125346. [DOI] [PubMed] [Google Scholar]
  95. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–676. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
  96. Tamura T, Nakagawa T, Iguchi F, Tateya I, Endo T, Kim TS, Dong Y, Kita T, Kojima K, Naito Y, Omori K, Ito J. Transplantation of neural stem cells into the modiolus of mouse cochleae injured by cisplatin. Acta Otolaryngol. 2004;(Suppl):65–68. doi: 10.1080/03655230310016780. [DOI] [PubMed] [Google Scholar]
  97. Tan BT, Lee MM, Ruan R. Bone-marrow-derived cells that home to acoustic deafened cochlea preserved their hematopoietic identity. J Comp Neurol. 2008;509:167–179. doi: 10.1002/cne.21729. [DOI] [PubMed] [Google Scholar]
  98. Tang XQ, Heron P, Mashburn C, Smith GM. Targeting sensory axon regeneration in adult spinal cord. J Neurosci. 2007;27:6068–6078. doi: 10.1523/JNEUROSCI.1442-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Tannemaat MR, Korecka J, Ehlert EM, Mason MR, van Duinen SG, Boer GJ, Malessy MJ, Verhaagen J. Human neuroma contains increased levels of semaphorin 3A, which surrounds nerve fibers and reduces neurite extension in vitro. J Neurosci. 2007;27:14260–14264. doi: 10.1523/JNEUROSCI.4571-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Tashiro A, Sandler VM, Toni N, Zhao C, Gage FH. NMDA-receptor-mediated, cell-specific integration of new neurons in adult dentate gyrus. Nature. 2006;442:929–933. doi: 10.1038/nature05028. [DOI] [PubMed] [Google Scholar]
  101. Thonabulsombat C, Johansson S, Spenger C, Ulfendahl M, Olivius P. Implanted embryonic sensory neurons project axons toward adult auditory brainstem neurons in roller drum and Stoppini co-cultures. Brain Res. 2007;1170:48–58. doi: 10.1016/j.brainres.2007.06.085. [DOI] [PubMed] [Google Scholar]
  102. Tokano H, Shi F, Liberman MC, Edge ASB. Improvement in function after transplantation of ES cell-derived neural progenitors into the de-afferented cochlea. Submitted. 2012 [Google Scholar]
  103. Tong M, Brugeaud A, Edge A. Generation of new synapses between mammalian afferent neurons and hair cells in vitro. ARO 34th Annual Meeting Abstract #846. 2011 [Google Scholar]
  104. Tong M, Brugeaud A, Edge ASB. Regenerated Synapses between Postnatal Hair Cells and Auditory Neurons. J Assoc Res Otolaryngol in revision. 2012 doi: 10.1007/s10162-013-0374-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Tritsch NX, Bergles DE. Developmental regulation of spontaneous activity in the Mammalian cochlea. J Neurosci. 2010;30:1539–1550. doi: 10.1523/JNEUROSCI.3875-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Tritsch NX, Yi E, Gale JE, Glowatzki E, Bergles DE. The origin of spontaneous activity in the developing auditory system. Nature. 2007;450:50–55. doi: 10.1038/nature06233. [DOI] [PubMed] [Google Scholar]
  107. Vierbuchen T, Ostermeier A, Pang ZP, Kokubu Y, Sudhof TC, Wernig M. Direct conversion of fibroblasts to functional neurons by defined factors. Nature. 2010;463:1035–1041. doi: 10.1038/nature08797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Wang J, Ding D, Salvi RJ. Carboplatin-induced early cochlear lesion in chinchillas. Hear Res. 2003;181:65–72. doi: 10.1016/s0378-5955(03)00176-x. [DOI] [PubMed] [Google Scholar]
  109. Wang Q, Green SH. Functional role of neurotrophin-3 in synapse regeneration by spiral ganglion neurons on inner hair cells after excitotoxic trauma in vitro. J Neurosci. 2011;31:7938–7949. doi: 10.1523/JNEUROSCI.1434-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Whitlon DS, Zhang X, Kusakabe M. Tenascin-C in the cochlea of the developing mouse. J Comp Neurol. 1999;406:361–374. doi: 10.1002/(sici)1096-9861(19990412)406:3<361::aid-cne5>3.0.co;2-o. [DOI] [PubMed] [Google Scholar]
  111. Yuan Y, Mizutari K, Cheng Y, Lang H, Liberman C, Shi F, Edge A. Reinnervation of Hair Cells and Cochlear Nucleus by Engrafted Neurons Derived from StemCells. ARO 35th Annual Meeting Abstract #355. 2012 [Google Scholar]
  112. Zhang R, Zhang L, Zhang Z, Wang Y, Lu M, Lapointe M, Chopp M. A nitric oxide donor induces neurogenesis and reduces functional deficits after stroke in rats. Ann Neurol. 2001;50:602–611. doi: 10.1002/ana.1249. [DOI] [PubMed] [Google Scholar]
  113. Zhou Y, Yuan J, Zhou B, Lee AJ, Ghawji M, Jr., Yoo TJ. The therapeutic efficacy of human adipose tissue-derived mesenchymal stem cells on experimental autoimmune hearing loss in mice. Immunology. 2011;133:133–140. doi: 10.1111/j.1365-2567.2011.03421.x. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES