Добавил:
Опубликованный материал нарушает ваши авторские права? Сообщите нам.
Вуз: Предмет: Файл:

Учебники / Auditory Trauma, Protection, and Repair Fay 2008

.pdf
Скачиваний:
200
Добавлен:
07.06.2016
Размер:
2.35 Mб
Скачать

302 S.H. Green, R.A. Altschuler, and J.M. Miller

earlier findings of Leake and colleagues (Leake et al. 1999). Differences in experimental design and other technical factors might also account for this discrepancy (Miller 2001). An important difference may be the density of surviving SGNs at the time when ES is initiated; that is, higher SGN density is synergistic with ES in promotion of SGN survival, and a “critical mass” of cells is necessary for ES to be successful. Deafening procedures that cause a rapid initial loss of SGNs, e.g., a single dose of aminoglycoside in combination with a loop diuretic, result in a lower density of SGNs at the time when ES is initiated. In contrast, repeated daily administrations of aminoglycoside alone presumably do not cause an immediate loss of all hair cells. The resulting death of SGNs would then be more gradual, even in the initial period, and SGN density higher at the initiation of ES.

Although there is no direct evidence to support the hypothesis that increased cell density in the spiral ganglion synergizes with ES to increase SGN survival, it is consistent with the presence of neurotrophin expression in the spiral ganglion in vivo (Zha et al. 2001) and autocrine and/or paracrine neurotrophic support of SGNs in vitro (Hansen et al. 2001a). Glia may also contribute to paracrine neurotrophic support of SGNs: as noted in the preceding text, spiral ganglion glia produce neurotrophic factors (Hansen et al. 2001a).

7.3 Combining Electrical Stimulation and Peptide Neurotrophic Factors Enhances SGN Survival over Either Alone

Given that neurotrophins and depolarization are additive in promoting survival of cultured SGNs (Hegarty et al. 1997; Hansen et al. 2001b), therapy involving a combination of these factors should be more effective than either alone in enhancing SGN survival after hair cell loss in vivo. Indeed, ES potentiated BDNF-dependent survival of deafferented SGNs in vivo in cats even when ES alone had no effect on SGN survival (Shepherd et al. 2005). Also, combination of ES with GDNF, the latter delivered either by a viral vector (Kanzaki et al. 2002) or by intracochlear perfusion, resulted in enhancement of SGN survival in vivo over GDNF alone. An important remaining question is whether ES alone will maintain SGN survival following withdrawal of neurotrophic factors. If so, this will permit maintenance of SGNs after hair cell loss without a need for continued long-term application of neurotrophic factors.

7.4 Protection of SGNs In Vivo Using Small-Molecule

Therapeutics

To date little in vivo work has been done testing the protective capacity of small molecules in the cochlea. However, on the basis of work in other systems and the initial work in the auditory system, at least three strategies—enhancement of neurotrophic signaling, inhibition of apoptosis, and antioxidants—hold promise, along with many challenges.

10. Cell Death and Cochlear Protection

303

7.4.1 Enhancement of Neurotrophic Signaling with Small Molecules

 

Neurotrophin signaling is enhanced by gangliosides such as GM1

(Ferrari

et al. 1995). Treatment with GM1 moderately reduces SGN death in vivo after hair cell loss, although severe shrinkage of the surviving SGNs was noted after termination of the GM1 treatment (Leake et al. 2007). However, a promising result of this study is that the trophic effects of GM1 were additive with those of ES and were maintained by continued ES after termination of the GM1.

7.4.2 Inhibition of Apoptosis by Small Molecules

Because apoptosis depends on caspases, caspase inhibition is a potential approach for therapy. The endogenous caspase inhibitors, IAPs, are thought to be particularly promising therapeutic targets and have been investigated in this regard in CNS trauma and neurodegenerative disease (Robertson et al. 2000). Targeting of caspases via IAPs has not yet been used as a protective mechanism for spiral ganglion neurons but inhibition of caspases by this or by the use of cell membrane–permeable inhibitors may be of therapeutic value in SGN protection (Liu et al. 1998). While more associated with excitotoxic cell death, calpains may play a role in the death of SGNs caused by other traumata including loss of neurotrophic support, suggesting that calpain inhibition may have a therapeutic role in SGN protection (Ding et al. 2002).

Inhibitors of JNK or its upstream activators appear promising as therapeutics for neurodegeneration (Bogoyevitch et al. 2004), and JNK inhibitors have been used to protect SGNs (Section 5.4). Since neurotrophic factor deprivation leads to a change in the oxidative state of deafferented neurons, antioxidant treatment may provide another approach to protect SGN from degeneration after hair cell loss.

7.4.3 Antioxidants

Antioxidants (Trolox and acorbic acid) administered either locally (intrascalar) or systemically after ototoxic deafening may reduce degeneration of SGNs and maintain the sensitivity of the auditory nerve to electrical excitation (Miller et al. 2002). Moreover, this efficacy was observed to continue for at least 2 weeks after cessation of systemic administration.

7.5 Regrowth of the Peripheral Process

Studies of humans and experimental animal models have shown that the SGN peripheral process that normally projects to the organ of Corti degenerates quickly after loss of hair cells and is absent even in surviving SGNs (Leake and Hradek 1988; Fayad et al. 1991; Nadol 1997). As a consequence, stimulation by a cochlear implant requires superthreshold depolarization at the SGN cell body or axon within the modiolus, necessitating high stimulating currents that broadly stimulate SGNs. To achieve maximal benefit, therapy to promote SGN survival

304 S.H. Green, R.A. Altschuler, and J.M. Miller

should also promote regrowth of the peripheral process to allow more focal stimulation of SGNs near an electrode. Neurotrophins promote neurite growth from SGNs in vitro (Lefebvre et al. 1994; Hegarty et al. 1997), and some studies suggest that neurotrophic factor therapy can promote in vivo growth of the SGN peripheral process in guinea pigs: neurotrophins or GDNF induce a regrowth of peripheral processes lost within days following hair cell death (Altschuler et al. 1999; Wise et al. 2005), with BDNF-induced regrowth of peripheral processes enhanced by FGF or electrical stimulation (Altschuler et al. 1999).

Depolarization reduces axon growth in cultured SGNs (Hegarty et al. 1997), suggesting that electrical stimulation might have adverse effects on peripheral process growth, maintenance, of physiology in vivo. The particular depolarization-initiated intracellular signals responsible for inhibiting SGN neurite growth are being identified (e.g., Hansen et al. 2003) and may be able to be specifically inhibited as part of therapy. However, patterned electrical activity, as opposed to chronic depolarization, appears to promote rather than inhibit neurite growth in other neurons (Goldberg et al. 2002). This may account for recent observations that have demonstrated positive effects of chronic ES on SGN peripheral processes (Altschuler et al. 1999; Altschuler et al., unpublished observations): ES shortly following hair cell death, and before complete peripheral process degeneration is capable of maintaining these processes; ES initiated weeks after hair cell death, after complete process degeneration, is able to induce a regrowth of processes through the habenula perforata and into the scarred remnants of organ of Corti.

8. Cochlear Blood Flow and Protection

Inner ear blood flow is reduced by intense noise as shown by laser-Doppler measurements (Thorne and Nuttall 1987). This may be a consequence of a noise-induced increase in levels of the vasoconstrictor 8-isoprostane , a lipid proxidation product, in the cochlea (Ohinata et al. 2000a). Direct administration of 8-isoprostane to the anterior inferior cerebellar artery (main blood supply to the inner ear) resulted in a reduction in blood flow. This was blocked by a specific 8-isoprostane antagonist, SQ29548, which also blocked the noiseinduced reduction in CBF (Miller et al. 2003). ROS are presumably involved because glutathione-mono-ethyl ester, a scavenger of oxidative free radicals (Miller et al. 2003) also blocked the noise-induced reduction in CBF. Antioxidants reduce 8-isoprostane formation in the lateral wall and provide protection from noise-induced hearing loss (Ohinata et al. 2003). These data implicate ROS and one of its products, the vasoconstrictor 8-isoprostane , in noise-induced reduction in CBF.

Magnesium, which increases cochlear blood flow, reduces NIHL in guinea pigs (Scheibe et al. 2002) and humans (Attias et al. 2004). This not only indicates that noise-induced reduction in CBF can exacerbate NIHL but also suggests that interventions including vasodilators can be protective against acoustic trauma.

10. Cell Death and Cochlear Protection

305

Combination of antioxidants (vitamins A, C, E) with the vasodilator magnesium provides greater attenuation of NIHL than either alone (Le Prell et al. 2007).

9. Protection of Other Cochlear Elements

Damage from noise and ototoxins is not limited to the organ of Corti or spiral ganglion. Other cochlear elements, including the stria vascularis and fibrocytes of the lateral wall, are also affected (Hirose and Liberman 2003; Imamura and Adams 2003; see also Henderson and Hu, Chapter 7; Rybak, Talaska, and Schacht, Chapter 8). These latter effects can contribute directly to hearing loss, for example, by altering the endocochlear potential (Hirose and Liberman 2003), or can have indirect effects on hair cells and SGNs by affecting cochlear homeostasis. Therefore, protection of lateral wall structures might reduce hearing loss caused by noise or other stresses.

10. Summary

The increasing understanding of the basic biology of cell death, protection, and neurotrophic mechanisms has suggested means to reduce the effects of trauma to the inner ear. Generally, traumata that cause permanent hearing loss—e.g., noise, ototoxins, aging—directly affect the hair cells, with spiral ganglion neuron (SGN) death being secondary to the loss of hair cells. However, SGNs are directly susceptible to excitotoxic damage. Known intrinsic protective systems in hair cells include heat shock proteins, antioxidants, and Ca2+ homeostasis; known exogenous systems include the olivocochlear efferents. If these protective systems are overwhelmed by the trauma, hair cells die, typically via known apoptotic pathways but not necessarily in the same fashion in all types of stress. Current strategies for protection of hair cells target enhancing intrinsic protective mechanisms—e.g., treatment with antioxidants—or blocking apoptotic pathways—e.g., inhibition of JNK. In the case of SGNs, protective strategies are based on current understanding of the neurotrophic support SGNs receive from hair cells, the prosurvival intracellular signaling pathways that the neurotrophic stimuli activate and the proapoptotic pathways that they inhibit. Current strategies for protecting SGNs involve enhancing neurotrophic support, e.g., intracochlear application of peptide neurotrophic factors or electrical stimulation by an implanted electrode. In vitro and in vivo studies emphasize the value of combinatorial approaches. One challenge is the identification of the most effective approach(es). A possibly more daunting challenge is the identification of effective means to deliver protective molecules to their intended cellular or subcellular targets, particularly means that will allow translation from animal models to clinical application.

306 S.H. Green, R.A. Altschuler, and J.M. Miller

Acknowledgments. Richard A. Altschuler and Josef M. Miller would like to acknowledge the contributions of Drs. Margaret Lomax, Amy Miller, Annieliese Shrott Fischer, and Mats Ulfendahl and SHG, and the contributions of all the members of the Green lab and the University of Iowa Auditory Neuroscience Group, especially Dr. Marlan Hansen. We thank Drs. Richard Bobbin and, especially, Jochen Schacht for their considerable and helpful contributions to the text. Studies of Drs. Miller and Altschuler were supported by NIH/NIDCD grants R01 DC003820 and P30 DC005188. Studies in the Green lab were supported by NIH/NIDCD grant R01 DC002961 and by the American Hearing Research Foundation.

References

Adamec E, Beermann ML, Nixon RA (1998) Calpain I activation in rat hippocampal neurons in culture is NMDA receptor selective and not essential for excitotoxic cell death. Brain Res Mol Brain Res 54:35–48.

Adamson CL, Reid MA, Davis RL (2002) Opposite actions of brain-derived neurotrophic factor and neurotrophin-3 on firing features and ion channel composition of murine spiral ganglion neurons. J Neurosci 22:1385–1396.

Affaitati A,Cardone L, de Cristofaro T, Carlucci A, Ginsberg MD, Varrone S, Gottesman ME, Avvedimento EV, Feliciello A (2003) Essential role of A-kinase anchor protein 121 for cAMP signaling to mitochondria. J Biol Chem 278:4286–4294.

Alam S, Robinson BK, Huang J, Green SH (2007) Prosurvival and proapoptotic intracellular signaling in rat spiral ganglion neurons in vivo after the loss of hair cells. J Comp Neurol 503:832–852.

Altschuler RA, Lim HH, Ditto J, Dolan D, Raphael Y (1996) Protective mechanisms in the cochlea: heat shock proteins. In:Salvi RJ, Henderson D, Fiorino F, Colletti V (eds) Auditory Plasticity and Regeneration. New York: Tieman Med Publishers, pp. 202–212.

Altschuler RA, Cho Y, Ylikoski J, Pirvola U, Magal E, Miller JM (1999) Rescue and regrowth of sensory nerves following deafferentation by neurotrophic factors. Ann NY Acad Sci 884:305–311.

Araki S, Kawano A, Seldon L, Shepherd RK, Funasaka S, Clark GM (1998) Effects of chronic electrical stimulation on spiral ganglion neuron survival and size in deafened kittens. Laryngoscope 108:687–695.

Araki S, Kawano A, Seldon HL, Shepherd RK, Funasaka S, Clark GM (2000) Effects of intracochlear factors on spiral ganglion cells and auditory brain stem response after long-term electrical stimulation in deafened kittens. Otolaryngol Head Neck Surg 122:425–433.

Araujo IM, Carvalho CM (2005) Role of nitric oxide and calpain activation in neuronal death and survival. Curr Drug Targets CNS Neurol Disord 4:319–324.

Attias J, Sapir S, Bresloff I, Reshef-Haran I, Ising H (2004) Reduction in noise-induced temporary threshold shift in humans following oral magnesium intake. Clin Otolaryngol Allied Sci 29:635–641.

Baloh RH, Enomoto H, Johnson EM, Jr., Milbrandt J (2000) The GDNF family ligands and receptors—implications for neural development. Curr Opin Neurobiol 10:103–110.

Barthwal MK, Sathyanarayana P, Kundu CN, Rana B, Pradeep A, Sharma C, Woodgett JR, Rana A (2003) Negative regulation of mixed lineage kinase 3 by protein kinase B/AKT leads to cell survival. J Biol Chem 278:3897–3902.

10. Cell Death and Cochlear Protection

307

Berridge MJ (1998) Neuronal calcium signaling. Neuron 21:13–26.

Bhakar AL, Tannis LL, Zeindler C, Russo MP, Jobin C, Park DS, MacPherson S, Barker PA (2002) Constitutive NF- B activity is required for central neuron survival. J Neurosci 22:8466–8475.

Bianchi LM, Raz Y (2004) Methods for providing therapeutic agents to treat damaged spiral ganglion neurons. Curr Drug Targets CNS Neurol Disord 3:195–199.

Bichler E, Spoendlin H, Rauchegger H (1983) Degeneration of cochlear neurons after amikacin intoxication in the rat. Arch Otorhinolaryngol 237:201–208.

Biswas SC, Liu DX, Greene LA (2005) Bim is a direct target of a neuronal E2F-dependent apoptotic pathway. J Neurosci 25:8349–8358.

Blondeau N, Widmann C, Lazdunski M, Heurteaux C (2001) Activation of the nuclear factor- B is a key event in brain tolerance. J Neurosci 21:4668–4677.

Bobbin RP (2001) PPADS, an ATP antagonist, attenuates the effects of a moderately intense sound on cochlear mechanics. Hear Res 156:10–16.

Bobbin RP (2002) Caffeine and ryanodine demonstrate a role for the ryanodine receptor in the organ of Corti. Hear Res 174:172–182.

Bobbin RP, Blesoe SJ (2005) Asphyxia and depolarization increase adenosine levels in perilymph. Hear Res 205:110–114.

Bodmer D, Gloddek B, Ryan AF, Huverstuhl J, Brors D (2002) Inhibition of the c-Jun N-terminal kinase signaling pathway influences neurite outgrowth of spiral ganglion neurons in vitro. Laryngoscope 112:2057–2061.

Bogoyevitch MA, Boehm I, Oakley A, Ketterman AJ, Barr RK (2004) Targeting the JNK MAPK cascade for inhibition: basic science and therapeutic potential. Biochim Biophys Acta 1697:89–101.

Bohne BA (1976) Safe level for noise exposure? Ann Otol Rhinol Laryngol 85:711–724. Bok J, Huang J, Wang Q, Green SH (2007) CaMKII and CaMKIV mediate divergent prosurvival signaling pathways in response to depolarization in neurons. Mol Cell

Neurosci 36:13–26.

Bok J, Zha XM, Cho YS, Green SH (2003) An extranuclear locus of cAMP-dependent protein kinase action is necessary and sufficient for promotion of spiral ganglion neuronal survival by cAMP. J Neurosci 23:777–787.

Brosenitsch TA, Katz DM (2001) Physiological patterns of electrical stimulation can induce neuronal gene expression by activating N-type calcium channels. J Neurosci 21:2571–2579.

Brown MC, deV Venecia RK, Guinan JJ (2003) Responses of medial olivocochlear neurons. Specifying the central pathways of the medial olivocochlear reflex. Exp Brain Res 153:491–498.

Brunet A, Bonni A, Zigmond MJ, Lin MZ, Juo P, Hu LS, Anderson MJ, Arden KC, Blenis J, Greenberg ME (1999) Akt promotes cell survival by phosphorylating and inhibiting a forkhead transcription factor. Cell 96:857–868.

Chen C, Nenov A, Bobbin RP (1995) Noise exposure induced change in outer hair cell response to ATP. Hear Res 88:215–221.

Cheng AG, Cunningham LL, Rubel EW (2005) Mechanisms of hair cell death and protection. Curr Opin Otolaryngol Head Neck Surg 13:343–348.

Culmsee C, Siewe J, Junker V, Retiounskaia M, Schwarz S, Camandola S, El-Metainy S, Behnke H, Mattson MP, Krieglstein J (2003) Reciprocal inhibition of p53 and nuclear factor-kB transcriptional activities determines cell survival or death in neurons. J Neurosci 23:8586–8595.

308 S.H. Green, R.A. Altschuler, and J.M. Miller

d’Aldin C, Puel JL, Leducq R, Crambes O, Eybalin M, Pujol R (1995) Effects of a dopaminergic agonist in the guinea pig cochlea. Hear Res 90:202–211.

Dallos P, He DZ, Lin X, Sziklai I, Mehta S, Evans BN (1997) Acetylcholine, outer hair cell electromotility, and the cochlear amplifier. J Neurosci 17:2212–2226.

Datta SR, Brunet A, Greenberg ME (1999) Cellular survival: a play in three Akts. Genes Dev 13:2905–2927.

Davis RJ (2000) Signal transduction by the JNK group of MAP kinases. Cell 103:239–252. Dawson TM, Ginty DD (2002) CREB family transcription factors inhibit neuronal suicide.

Nat Med 8:450–451.

De Cesare D, Sassone-Corsi P (2000) Transcriptional regulation by cyclic AMPresponsive factors. Prog Nucleic Acid Res Mol Biol 64:343–369.

Deshmukh M, Johnson EM Jr (1998) Evidence of a novel event during neuronal death: development of competence-to-die in response to cytoplasmic cytochrome C. Neuron 21:695–705.

Ding L, McFadden SL, Salvi RJ (2002) Calpain immunoreactivity and morphological damage in chinchilla inner ears after carboplatin. J Assoc Res Otolaryngol 3:68–79.

Donovan N, Becker EB, Konishi Y, Bonni A (2002) JNK phosphorylation and activation of BAD couples the stress-activated signaling pathway to the cell death machinery. J Biol Chem 277:40944–40949.

Downward J (1999) How BAD phosphorylation is good for survival. Nat Cell Biol 1:E33–35.

Eilers A, Whitfield J, Shah B, Spadoni C, Desmond H, Ham J (2001) Direct inhibition of c-Jun N-terminal kinase in sympathetic neurons prevents c-jun promoter activation and NGF withdrawal-induced death. J Neurochem 76:1439–1454.

Encinas M, Tansey MG, Tsui-Pierchala BA, Comella JX, Milbrandt J, Johnson EM (2001) c-Src is required for glial cell line-derived neurotrophic factor (GDNF) family ligandmediated neuronal survival via a phosphatidylinositol-3 kinase (PI-3K)-dependent pathway. J Neurosci 21:1464–1472.

Ernfors P, Duan ML, ElShamy WM, Canlon B (1996) Protection of auditory neurons from aminoglycoside toxicity by neurotrophin-3. Nature Med 2:463–467.

Fairfield DA, Kanicki AC, Lomax MI, Altchuler RA (2002) Expression and localization of heat shock factor (HSF) 1 in the rodent cochlea. Hear Res 173:109–118.

Fairfield DA, Kanicki AC, Lomax MI, Altchuler RA (2004) Induction of heat shock protein 32 (Hsp32) in the rat cochlea following hyperthermia. Hear Res 188:1–11.

Fairfield DA, Lomax MI, Dootz GA, Chen S, Galecki TA, Benjamin IJ, Dolan DF, Altchuler RA (2005) Heat shock factor 1 (Hsf1) deficient mice exhibit decreased recovery of hearing following noise overstimulation. J Neurosci Res 81:589–596.

Fayad J, Linthicum FH, Jr., Otto SR, Galey FR, House WF (1991) Cochlear implants: histopathologic findings related to performance in 16 human temporal bones. Ann Otol Rhinol Laryngol 100:807–811.

Ferrari G, Anderson BL, Stephens RM, Kaplan DR, Green LA (1995) Prevention of apoptotic neuronal death by GM1 ganglioside. Involvement of Trk neurotrophin receptors. J Biol Chem 270:3074–3080.

Fesik SW, Shi Y (2001) Controlling the caspases. Science 294:1477–1478.

Ford MS, Maggirwar SB, Rybak LP, Whitworth C, Ramkumar V (1997) Expression and function of adenosine receptors in the chinchilla cochlea. Hear Res 105:130–140.

Forge A, Schacht J (2000) Aminoglycoside antibiotics. Audiol Neurootol 5:3–22. Franklin JL, Johnson EM Jr (1994) Block of neuronal apoptosis by a sustained increase of

steady-state free Ca2+ concentration. Philos Trans R Soc Lond B Biol Sci 345:251–256.

10. Cell Death and Cochlear Protection

309

Franklin JL, Sanz-Rodriguez C, Juhasz A, Deckwerth TL, Johnson EM Jr (1995) Chronic depolarization prevents programmed death of sympathetic neurons in vitro but does not support growth: requirement for Ca2+ influx but not Trk activation. J Neurosci 15:643–664.

Fridberger A, Flóck A, Ulfendahl M, Flóck B (1998) Acoustic overstimulation increases outer hair cell Ca2+ concentrations and causes dynamic contractions of the hearing organ. Proc Natl Acad Sci USA 95:7127–7132.

Fridmacher V, Kaltschmidt B, Goudeau B, Ndiaye D, Rossi FM, Pfeiffer J, Kaltschmidt C, Israel A, Memet S (2003) Forebrain-specific neuronal inhibition of nuclear factor- B activity leads to loss of neuroprotection. J Neurosci 23:9403–9408.

Fritzsch B, Tessarollo L, Coppola E, Reichardt LF (2004) Neurotrophins in the ear: their roles in sensory neuron survival and fiber guidance. Prog Brain Res 146:265–278.

Fritzsch B, Pauley S, Matei V, Katz DM, Xiang M, Tessarollo L (2005) Mutant mice reveal the molecular and cellular basis for specific sensory connections to inner ear epithelia and primary nuclei of the brain. Hear Res 206:52–63.

Galli C, Meucci O, Scorziello A, Werge TM, Calissano P, Schettini G (1995) Apoptosis in cerebellar granule cells is blocked by high KCl, forskolin, and IGF-1 through distinct mechanisms of action: the involvement of intracellular calcium and RNA synthesis. J Neurosci 15:1172–1179.

Garnier P, Ying W, Swanson RA (2003) Ischemic preconditioning by caspase cleavage of poly(ADP-ribose) polymerase-1. J Neurosci 23:7967–7973.

Gates GA (2006) The effect of noise on cochlear aging. Ear Hear 27:91.

Gentry JJ, Barker PA, Carter BD (2004) The p75 neurotrophin receptor: multiple interactors and numerous functions. Prog Brain Res 146:25–39.

Gestwa G, Wiechers B, Zimmermann U, Praetorius M, Rohbock K, Kopschall I, Zenner HP, Knipper M (1999) Differential expression of trkB.T1 and trkB.T2, truncated trkC, and p75NGFR in the cochlea prior to hearing function. J Comp Neurol 414: 33–49.

Ghahremani MH, Keramaris E, Shree T, Xia Z, Davis RJ, Flavell R, Slack RS, Park DS (2002) Interaction of the c-Jun/JNK pathway and cyclin-dependent kinases in death of embryonic cortical neurons evoked by DNA damage. J Biol Chem 277:35586–35596.

Ghoumari AM, Wehrle R, De Zeeuw CI, Sotelo C, Dusart I (2002) Inhibition of protein kinase C prevents Purkinje cell death but does not affect axonal regeneration. J Neurosci 22:3531–3542.

Gidday JM (2006) Cerebral preconditioning and ischaemic tolerance. Nat Rev Neurosci 7:437–448.

Gillespie LN, Clark GM, Bartlett PF, Marzella PL (2003) BDNF-induced survival of auditory neurons in vivo: cessation of treatment leads to accelerated loss of survival effects. J Neurosci Res 71:785–790.

Goldberg JL, Espinosa JS, Xu Y, Davidson N, Kovacs GT, Barres BA (2002) Retinal ganglion cells do not extend axons by default. Promotion by neurotrophic signaling and electrical activity. Neuron 33:689–702.

Green SH (2000) Neurotrophic signaling by membrane electrical activity in spiral ganglion neurons. In: Lim DJ (ed) Cell and Molecular Biology of the Ear. New York: Kluwer Academic/Plenum, pp. 165–182.

Greenlund LJS, Deckwerth TL, Johnson EM Jr (1995) Superoxide dismutase delays neuronal apoptosis: a role for reactive oxygen species in programmed neuronal death. Neuron 14:303–315.

310 S.H. Green, R.A. Altschuler, and J.M. Miller

Groff JA, Liberman MC (2003) Modulation of cochlear afferent response by the lateral olivocochlear system: activation via electrical stimulation of the inferior colliculus. J Neurophysiol 90:3178–3200.

Ham J, Eilers A, Whitfield J, Neame SJ, Shah B (2000) c-Jun and the transcriptional control of neuronal apoptosis. Biochem Pharmacol 60:1015–1021.

Hamernik RP, Henderson D (1974) Impulse noise trauma. A study of histological susceptibility. Arch Otorhinolaryngol 99:118–121.

Hansen MR, Vijapurkar U, Koland JG, Green SH (2001a) Reciprocal signaling between spiral ganglion neurons and Schwann cells involves neuregulin and neurotrophins. Hear Res 161:87–98.

Hansen MR, Zha X-M, Bok J, Green SH (2001b) Multiple distinct signal pathways, including an autocrine neurotrophic mechanism, contribute to the survival-promoting effect of depolarization on spiral ganglion neurons. J Neurosci 21:2256–2267.

Hansen MR, Devaiah AK, Bok J, Zha X, Green SH (2003) Ca2+/calmodulin-dependent protein kinases II and IV function similarly in neurotrophic signaling but differ in their effects on neurite growth in spiral ganglion neurons. J, Neurosci Res 72:169–184.

Harada H, Becknell B, Wilm M, Mann M, Huang LJ, Taylor SS, Scott JD, Korsmeyer SJ (1999) Phosphorylation and inactivation of BAD by mitochondria-anchored protein kinase A. Mol Cell 3:413–422.

Harden TK, Boyer JL, Nicholas RA (1995) P2-purinergic receptors: subtype-associated signaling responses and structure. Annu Rev Pharmacol Toxicol 35:541–579.

Harris KC, Bielefeld E, Hu BH, Henderson D (2006) Increased resistance to free radical damage induced by low-level sound conditioning. Hear Res 213:118–129.

Harris MH, Thompson CB (2000) The role of the Bcl-2 family in the regulation of outer mitochondrial membrane permeability. Cell Death Differ 7:1182–1191.

Hartnick C, Staecker H, Malgrange B, Lefebvre P, Liu W, Moonen G, van de Water TR (1996) Neurotrophic effects of BDNF and CNTF, alone and in combination, on postnatal day 5 rat acoustic ganglion neurons. J Neurobiol 30:246–254.

Hegarty JL, Kay AR, Green SH (1997) Trophic support of cultured spiral ganglion neurons by depolarization exceeds and is additive with that by neurotrophins or cyclic AMP, and requires elevation of [Ca2+]i within a set range. J Neurosci 17:1959–1970.

Heinrich UR, Maurer J, Mann W (1999) Ultrastructural evidence for protection of the outer hair cells of the inner ear during intense noise exposure by application of the organic calcium channel blocker diltiazem. ORL J Otolaryngolg Relat Spec 61:321–327.

Henderson D, Bielefeld EC, Harris KC, Hu BH (2006) The role of oxidative stress in noise-induced hearing loss. Ear Hear 27:1–19.

Hengartner MO (2000) The biochemistry of apoptosis. Nature 407:770 –776.

Hiebert SW, Packham G, Strom DK, Haffner R, Oren M, Zambetti G, Cleveland JL (1995) E2F-1:DP-1 induces p53 and overrides survival factors to trigger apoptosis. Mol Cell Biol 15:6864–6874.

Higuchi M, Tomioka M, Takano J, Shirotani K, Iwata N, Masumoto H, Maki M, Itohara S, Saido TC (2005) Distinct mechanistic roles of calpain and caspase activation in neurodegeneration as revealed in mice overexpressing their specific inhibitors. J Biol Chem 280:15229–15237.

Hirose K, Liberman MC (2003) Lateral wall histopathology and endocochlear potential in the noise-damaged mouse cochlea. J Assoc Res Otolaryngol 4:339–352.

Housley GD, Thorne PR (2000) Purinergic signalling: an experimental perspective. J Auton Nerv Syst 81:139–145.

10. Cell Death and Cochlear Protection

311

Hu BH, Zheng XY, McFadden SL, Kopke RD, Henderson D (1997) R-phenylisopro- pyladenosine attenuates noise-induced hearing loss in the chinchilla. Hear Res 113: 198–206.

Huang DC, Strasser A (2000) BH3-only proteins—essential initiators of apoptotic cell death. Cell 103:839–842.

Huang EJ, Reichardt LF (2001) Neurotrophins: roles in neuronal development and function. Annu Rev Neurosci 24:677–736.

Huang EJ, Reichardt LF (2003) Trk receptors: roles in neuronal signal transduction. Annu Rev Biochem 72:609–642.

Hunter-Duvar IM, Bredberg G (1974) Effects of intense auditory stimulation: hearing losses and inner ear changes in the chinchilla. J Acoust Soc Am 55:795–801.

Imamura S, Adams JC (2003) Changes in cytochemistry of sensory and nonsensory cells in gentamicin-treated cochleas. J Assoc Res Otolaryngol 4:196–218.

Ishii N, Wanaka A, Ohno K, Matsumoto K, Eguchi Y, Mori T, Tsujimoto Y, Tohyama M (1996) Localization of bcl-2, bax, and bcl-x mRNAs in the developing inner ear of the mouse. Brain Res 726:123–128.

Jacono AA, Hu B, Kopke RD, Henderson D, Van de Water TR, Steinman HM (1998) Changes in cochlear antioxidant enzyme activity after sound conditioning and noise exposure in the chinchilla. Hear Res 117:31–38.

Jiang H, Schacht J (2005) NF-kappaB pathway protects cochlear hair cells from aminoglycoside-induced ototoxicity. J Neurosci Res 79:644–651.

Jiang H, Sha S-H, Forge A, Schacht J (2006) Caspase-independent pathways of hair cell death induced by kanamycin in vivo. Cell Death Differ 13:20–30.

Kanzaki S, Stover T, Kawamoto K, Prieskorn DM, Altschuler RA, Miller JM, Raphael Y (2002) Glial cell line-derived neurotrophic factor and chronic electrical stimulation prevent VIII cranial nerve degeneration following denervation. J Comp Neurol 454:350–360.

Kanzaki S, Beyer L, Karolyi IJD, D.F., Fang Q, Probst FJ, Camper SA, Raphael Y (2006) Transgene correction maintains normal cochlear structure and function in 6–month-old Myo15a mutant mice. Hear Res 214:37–44.

Keithley EM, Ma CL, Ryan AF, Louis JC, Magal E (1998) GDNF protects the cochlea against noise damage. NeuroReport 9:2183–2187.

Keithley EM, Canto C, Zheng QY, Wang X, Fischel-Ghodsian N, Johnson KR (2005) Cu/Zn superoxide dismutase and age-related hearing loss. Hear Res 209:76–85.

Kelly TC, Whitworth CA, Husain K, Rybak LP (2003) Aminoguanidine reduces cisplatin ototoxicity. Hear Res 186:10–16.

Koike T, Tanaka S (1991) Evidence that nerve growth factor dependence of sympathetic neurons for survival in vitro may be determined by levels of cytoplasmic free Ca2+. Proc Natl Acad Sci USA 88:3892–3896.

Koitchev K, Guilhaume A, Cazals Y, Aran J-M (1982) Spiral ganglion changes after massive aminoglycoside treatment in the guinea pig. Counts and ultrastructure. Acta Otolaryngol 94:431–438.

Konishi Y, Bonni A (2003) The E2F-Cdc2 cell-cycle pathway specifically mediates activity deprivation-induced apoptosis of postmitotic neurons. J Neurosci 23: 1649–1658.

Konishi Y, Lehtinen M, Donovan N, Bonni A (2002) Cdc2 phosphorylation of BAD links the cell cycle to the cell death machinery. Mol Cell 9:1005–1016.

Kroemer G, Reed JC (2000) Mitochondrial control of cell death. Nat Med 6:513–519.