Maladaptive plasticity in tinnitus--triggers, mechanisms and treatment

Susan E Shore, Larry E Roberts, Berthold Langguth, Susan E Shore, Larry E Roberts, Berthold Langguth

Abstract

Tinnitus is a phantom auditory sensation that reduces quality of life for millions of people worldwide, and for which there is no medical cure. Most cases of tinnitus are associated with hearing loss caused by ageing or noise exposure. Exposure to loud recreational sound is common among the young, and this group are at increasing risk of developing tinnitus. Head or neck injuries can also trigger the development of tinnitus, as altered somatosensory input can affect auditory pathways and lead to tinnitus or modulate its intensity. Emotional and attentional state could be involved in the development and maintenance of tinnitus via top-down mechanisms. Thus, military personnel in combat are particularly at risk owing to combined risk factors (hearing loss, somatosensory system disturbances and emotional stress). Animal model studies have identified tinnitus-associated neural changes that commence at the cochlear nucleus and extend to the auditory cortex and other brain regions. Maladaptive neural plasticity seems to underlie these changes: it results in increased spontaneous firing rates and synchrony among neurons in central auditory structures, possibly generating the phantom percept. This Review highlights the links between animal and human studies, and discusses several therapeutic approaches that have been developed to target the neuroplastic changes underlying tinnitus.

Conflict of interest statement

Conflict of Interest Statements

Berthold Langguth received honoraria for speaking and consultancy from ANM, Astra Zeneca, Autifony, Gerson Lehrman Group, Lundbeck, McKinsey, Merz, Magventure, Novartis, Neuromod Devices, Pfizer and Servier, grants and support for research from the Tinnitus Research Initiative, the German Forschungsgemeinschaft, the German Bundesministerium für Bildung und Forschung, the American Tinnitus Association, Astra Zeneca, Cerbomed, Deymed, Magventure, Sivantos and Otonomy, and travel and accommodation payments from the European Union (COST), Lilly, Servier and Pfizer.

Berthold Langguth holds patents for the use of neuronavigation for transcranial magnetic stimulation for tinnitus treatment and for the use of cyclobenzaprine for tinnitus treatment

Figures

Figure 1
Figure 1
A. Schematic describing the GPIAS assay for tinnitus. A startle stimulus (black) is inserted into a background noise without a gap (no gap; top row) and with a gap (middle and lower rows; 50 msec. gap, 50 ms before the startle sound) are presented to the animal. Each trial consists of a continuous 60 dB background sound (grey bar) with a 10 msec., 115 dB startle pulse embedded (black bar). The guinea pig startles in response to the startle stimulus, with the amplitude of the response shown by the height of each arrow. In animals without tinnitus, the gap suppresses the startle response (middle row). In animals with tinnitus, the gap is filled by the tinnitus (pink) and the startle response shows less reduction relative to the no gap startle response (white arrow). B–F. Gaussian-mixture model analysis partitions the normalized startle distribution into normal and tinnitus distributions. B. Histogram of the normalized startle distribution (white line) partitioned into two distributions: no evidence for tinnitus (black bars) and evidence for tinnitus (red bars). C. The probability distributions of normalized startle values belonging to the tinnitus or non-tinnitus distributions. D. Histogram of the partitioned distribution of post-exposure normalized startle observations for sham animals. E. Histogram of the partitioned distribution of normalized startle observations for baseline (pre-exposure) observations from sham and exposed animals. F. Histogram of the partitioned distribution of post-exposure normalized startle observations from exposed animals. The percentage of observations placed into the tinnitus group is shown on each panel in D–F. Adapted from ([41]).
Figure 2
Figure 2
Simplified representation of auditory and non-auditory pathways discussed in this article and revealed by functional imaging studies to be involved in aspects of tinnitus. Blue identifies principal structures in the auditory pathway commencing with the cochlear nucleus (CN) and projecting through the inferior colliculus (IC) to the thalamus and auditory cortex (A1, primary auditory cortex A2; secondary auditory cortex; AAC, anterior auditory field). Return projections to the thalamus (these projections more numerous than forward projections) and subcortical structures in the auditory pathway are represented by a thickened arrow. Output from auditory pathways distributes to several major nonauditory regions of the brain, here simplified by their putative functional roles in normal information processing as identified by neuroscience research (see color code). Prominent structures involved in memory and emotion include the parahippocampal gyrus, amygdala, and limbic region including the insula (yellow). Participating in attention and consciousness (green) are the anterior and posterior cingulate regions, orbitofrontal cortex, prefrontal cortex (dorsomedial and ventromedial divisions), the subcallosal region (nucleus accumbens), and posterior partietal cortex including the precuneus. Sensori-motor pathways (red) include somatosensory ganglia, brainstem somatosensory pathways, primary and secondary somatosensory cortex, and the cerebellum. In this summary diagram connections among these regions are portrayed by arrows, but the connections among them are densely parallel and reciprocal mediated by cortico-cortico projections directly or via the thalamus as well as by multisensory interactions that occur in subcortical auditory structures discussed in the article. A2, secondary auditory cortex; AAC, anterior auditory field; A1, primary auditory cortex; IC, inferior colliculus; CN, cochlear nucleus.
Figure 3. Mechanisms of tinnitus initiation in…
Figure 3. Mechanisms of tinnitus initiation in the dorsal cochlear nucleus
A. Schematic of DCN circuitry showing the principal output neurons, fusiform cells and inhibitory interneurons, the cartwheel cells. Electrical stimulation and recording locations are shown in spinal trigeminal nucleus (Sp5) and DCN for measuring StDP. The thirty two-channel recording electrodes (black) spanned the tonotopic axis of the DCN. Short current pulses delivered via a bipolar stimulating electrode (brown) placed into Sp5 activated parallel-fiber inputs to DCN that activate fusiform cells subsequent to granule-cell activation. Tones were delivered through calibrated, hollow ear bars. Ca - cartwheel cell; Fu - fusiform cell; Gr - granule cell; St – Stellate cell; IC - inferior colliculus; Sp5 - spinal trigeminal nucleus; a.n.f - auditory nerve fiber; p.f - parallel fiber. ET- Exposed with tinnitus; ENT – exposed without tinnitus. B. Cartoon of the locations of the recording and stimulating electrodes relative to the animal’s head. C. Bimodal plasticity of spontaneous firing rates (SFR) shifts from predominantly Hebbian in shams (where somatosensory preceding auditory produces facilitation at 20 ms) to anti-Hebbian rules in guinea pigs with tinnitus (facilitation now seen when auditory precedes somatosensory stimulation at −20 ms) and suppressive at all pairing intervals in guinea pigs without tinnitus. Mean timing rules are shown for SFRs for units from Sham (gray), ENT (pink), and ET (red) guinea pigs. Cartoon inset at top represents the relative order of Sp5 and sound stimuli. The brown vertical line indicates the Sp5 stimulation, and the sinusoid represents the tone stimulus. Mean timing rules were computed for all measurements from DCN fusiform cell units. Error bars indicate SEM. Adapted from ([41]). D. Putative molecular mechanisms underlying changes in StDP, SFR and synchrony of DCN principal output neurons (fusiform cells) in initiating tinnitus. Noise over-exposure triggers auditory nerve fiber (ANF) deafferentation or neuropathy leading to alterations in the DCN circuitry via the following processes: Synaptic changes include decreased glycinergic (Gly), GABAergic, and increased glutamatergic (Glu) neurotransmission; intrinsic cellular changes include NMDA receptor (NMDA-R), voltage-gated potassium channel (Kv), and hyperpolarization-activated cyclic nucleotide-gated (HCN) channel properties. Each of these processes has been shown to affect one or more tinnitus phenotypes in DCN: increased SFR, increased neural synchrony, and inverted STDP.

References

    1. Axelsson A, Ringdahl A. Tinnitus–a study of its prevalence and characteristics. Br J Audiol. 1989;23(1):53–62.
    1. Park B, et al. Analysis of the prevalence of and risk factors for tinnitus in a young population. Otol Neurotol. 2014;35(7):1218–22.
    1. Gallus S, et al. Prevalence and Determinants of Tinnitus in the Italian Adult Population. Neuroepidemiology. 2015;45(1):12–9.
    1. Roberts LE, et al. Ringing ears: the neuroscience of tinnitus. J Neurosci. 2010;30(45):14972–9.
    1. Reed GF. An audiometric study of two hundred cases of subjective tinnitus. AMA Arch Otolaryngol. 1960;71:84–94.
    1. Vernon J. Attemps to relieve tinnitus. J Am Audiol Soc. 1977;2(4):124–31.
    1. Hallberg LR, Erlandsson SI. Tinnitus characteristics in tinnitus complainers and noncomplainers. Br J Audiol. 1993;27(1):19–27.
    1. Landgrebe M, et al. The Tinnitus Research Initiative (TRI) database: a new approach for delineation of tinnitus subtypes and generation of predictors for treatment outcome. BMC Med Inform Decis Mak. 2010;10:42.
    1. De Ridder D, et al. Phantom percepts: tinnitus and pain as persisting aversive memory networks. Proc Natl Acad Sci U S A. 2011;108(20):8075–80.
    1. Lockwood AH, Salvi RJ, Burkard RF. Tinnitus. N Engl J Med. 2002;347(12):904–10.
    1. Mardini MK. Ear-clicking “tinnitus” responding to carbamazepine. N Engl J Med. 1987;317(24):1542.
    1. House JW, Brackmann DE. Tinnitus: surgical treatment. Ciba Found Symp. 1981;85:204–216.
    1. Baguley DM, et al. The effect of vestibular nerve section upon tinnitus. Clin Otolaryngol. 2002;27(4):219–26.
    1. Soleymani T, et al. Surgical approaches to tinnitus treatment: A review and novel approaches. Surg Neurol Int. 2011;2:154.
    1. Blasco MA, Redleaf MI. Cochlear implantation in unilateral sudden deafness improves tinnitus and speech comprehension: meta-analysis and systematic review. Otol Neurotol. 2014;35(8):1426–32.
    1. Barnea G, et al. Tinnitus with normal hearing sensitivity: extended high-frequency audiometry and auditory-nerve brain-stem-evoked responses. Audiology. 1990;29(1):36–45.
    1. Schaette R, McAlpine D. Tinnitus with a normal audiogram: physiological evidence for hidden hearing loss and computational model. J Neurosci. 2011;31(38):13452–7.
    1. Roberts LE, et al. Residual Inhibition Functions Overlap Tinnitus Spectra and the Region of Auditory Threshold Shift. J Assoc Res Otolaryngol. 2008
    1. Kujawa SG, Liberman MC. Adding insult to injury: cochlear nerve degeneration after “temporary” noise-induced hearing loss. J Neurosci. 2009;29(45):14077–85.
    1. Furman AC, Kujawa SG, Liberman MC. Noise-induced cochlear neuropathy is selective for fibers with low spontaneous rates. J Neurophysiol. 2013;110(3):577–86.
    1. Sergeyenko Y, et al. Age-related cochlear synaptopathy: an early-onset contributor to auditory functional decline. J Neurosci. 2013;33(34):13686–94.
    1. Kujawa SG, Liberman MC. Synaptopathy in the noise-exposed and aging cochlea: Primary neural degeneration in acquired sensorineural hearing loss. Hear Res. 2015
    1. Lin HW, et al. Primary neural degeneration in the Guinea pig cochlea after reversible noise-induced threshold shift. J Assoc Res Otolaryngol. 2011;12(5):605–16.
    1. Gu JW, et al. Brainstem Auditory Evoked Potentials Suggest a Role for the Ventral Cochlear Nucleus in Tinnitus. J Assoc Res Otolaryngol. 2012
    1. Norena A, et al. Psychoacoustic characterization of the tinnitus spectrum: implications for the underlying mechanisms of tinnitus. Audiol Neurootol. 2002;7(6):358–69.
    1. Sereda M, et al. Re-examining the relationship between audiometric profile and tinnitus pitch. Int J Audiol. 2011;50(5):303–12.
    1. Zhou X, et al. Impaired cochlear function correlates with the presence of tinnitus and its estimated spectral profile. Hear Res. 2011;277(1–2):107–16.
    1. Heijneman KM, et al. Can the tinnitus spectrum identify tinnitus subgroups? Noise Health. 2013;15(63):101–6.
    1. Roberts LE. Residual inhibition. Prog Brain Res. 2007;166:487–95.
    1. Heffner HE, Heffner RS. Behavioral Tests for Tinnisus in Animals. In: Eggermont JJ, et al., editors. Tinnitus. Springer; New York: 2012.
    1. Turner J, et al. Time course of tinnitus development following noise exposure in mice. J Neurosci Res. 2012;90(7):1480–8.
    1. Turner JG, et al. Gap detection deficits in rats with tinnitus: a potential novel screening tool. Behav Neurosci. 2006;120(1):188–95.
    1. Dehmel S, Eisinger D, Shore SE. Gap prepulse inhibition and auditory brainstem-evoked potentials as objective measures for tinnitus in guinea pigs. Front Syst Neurosci. 2012;6:42.
    1. Berger JI, et al. A novel behavioural approach to detecting tinnitus in the guinea pig. J Neurosci Methods. 2013;213(2):188–95.
    1. Hayes SH, et al. Behavioral models of tinnitus and hyperacusis in animals. Front Neurol. 2014;5:179.
    1. von der Behrens W. Animal models of subjective tinnitus. Neural Plast. 2014;2014:741452.
    1. Galazyuk A, Hebert S. Gap-Prepulse Inhibition of the Acoustic Startle Reflex (GPIAS) for Tinnitus Assessment: Current Status and Future Directions. Front Neurol. 2015;6:88.
    1. Kaltenbach JA, et al. Activity in the dorsal cochlear nucleus of hamsters previously tested for tinnitus following intense tone exposure. Neurosci Lett. 2004;355(1–2):121–5.
    1. Brozoski TJ, Bauer CA, Caspary DM. Elevated fusiform cell activity in the dorsal cochlear nucleus of chinchillas with psychophysical evidence of tinnitus. J Neurosci. 2002;22(6):2383–90.
    1. Dehmel S, et al. Noise overexposure alters long-term somatosensory-auditory processing in the dorsal cochlear nucleus–possible basis for tinnitus-related hyperactivity? J Neurosci. 2012;32(5):1660–71.
    1. Koehler SD, Shore SE. Stimulus timing-dependent plasticity in dorsal cochlear nucleus is altered in tinnitus. J Neurosci. 2013;33(50):19647–56.
    1. Xiong H, et al. Hidden hearing loss in tinnitus patients with normal audiograms: implications for the origin of tinnitus. Lin Chung Er Bi Yan Hou Tou Jing Wai Ke Za Zhi. 2013;27(7):362–5.
    1. Vogler DP, Robertson D, Mulders WH. Hyperactivity following unilateral hearing loss in characterized cells in the inferior colliculus. Neuroscience. 2014;265C:28–36.
    1. Sumner CJ, Tucci DL, Shore SE. Responses of ventral cochlear nucleus neurons to contralateral sound after conductive hearing loss. J Neurophysiol. 2005;94(6):4234–43.
    1. Bledsoe SC, Jr, et al. Ventral cochlear nucleus responses to contralateral sound are mediated by commissural and olivocochlear pathways. J Neurophysiol. 2009;102(2):886–900.
    1. Kaltenbach JA. Summary of evidence pointing to a role of the dorsal cochlear nucleus in the etiology of tinnitus. Acta Otolaryngol Suppl. 2006;556:20–6.
    1. Wu C, et al. Tinnitus: Maladaptive auditory-somatosensory plasticity. Hear Res. 2015
    1. Robertson D, et al. Spontaneous hyperactivity in the auditory midbrain: relationship to afferent input. Hear Res. 2013;295:124–9.
    1. Mulders WH, Barry KM, Robertson D. Effects of furosemide on cochlear neural activity, central hyperactivity and behavioural tinnitus after cochlear trauma in guinea pig. PLoS One. 2014;9(5):e97948.
    1. Ropp TJ, et al. Effects of unilateral acoustic trauma on tinnitus-related spontaneous activity in the inferior colliculus. J Assoc Res Otolaryngol. 2014;15(6):1007–22.
    1. Coomber B, et al. Neural changes accompanying tinnitus following unilateral acoustic trauma in the guinea pig. Eur J Neurosci. 2014
    1. Brozoski TJ, et al. Bilateral Dorsal Cochlear Nucleus Lesions Prevent Acoustic-Trauma Induced Tinnitus in an Animal Model. J Assoc Res Otolaryngol. 2011
    1. Manzoor NF, et al. Noise-induced hyperactivity in the inferior colliculus: its relationship with hyperactivity in the dorsal cochlear nucleus. J Neurophysiol. 2012;108(4):976–88.
    1. Brozoski TJ, Bauer CA. The effect of dorsal cochlear nucleus ablation on tinnitus in rats. Hear Res. 2005;206(1–2):227–36.
    1. Mulders WH, et al. Relationship between auditory thresholds, central spontaneous activity, and hair cell loss after acoustic trauma. J Comp Neurol. 2011;519(13):2637–47.
    1. Zacharek MA, et al. Effects of cochlear ablation on noise induced hyperactivity in the hamster dorsal cochlear nucleus: implications for the origin of noise induced tinnitus. Hear Res. 2002;172(1–2):137–43.
    1. Zhang JS, et al. Origin of hyperactivity in the hamster dorsal cochlear nucleus following intense sound exposure. J Neurosci Res. 2006;84(4):819–31.
    1. Manzoor NF, et al. Comparison and contrast of noise-induced hyperactivity in the dorsal cochlear nucleus and inferior colliculus. Hear Res. 2013;295:114–23.
    1. Anderson LA, et al. Evidence for a direct, short latency projection from the dorsal cochlear nucleus to the auditory thalamus in the guinea pig. Eur J Neurosci. 2006;24(2):491–8.
    1. Malmierca MS, et al. Direct projections from cochlear nuclear complex to auditory thalamus in the rat. J Neurosci. 2002;22(24):10891–7.
    1. Schofield BR, Mellott JG, Motts SD. Subcollicular projections to the auditory thalamus and collateral projections to the inferior colliculus. Front Neuroanat. 2014;8:70.
    1. Kalappa BI, et al. Single unit hyperactivity and bursting in the auditory thalamus of awake rats directly correlates with behavioural evidence of tinnitus. J Physiol. 2014;592(Pt 22):5065–78.
    1. Eggermont JJ, Roberts LE. The neuroscience of tinnitus. Trends Neurosci. 2004;27(11):676–82.
    1. Engineer ND, et al. Reversing pathological neural activity using targeted plasticity. Nature. 2011;470(7332):101–4.
    1. Basura G, Koehler S, S SE. Stimulus-timing dependence of auditory-somatosensory plasticity in auditory cortex neurons after noise induced temporary threshold shifts and tinnitus. Journal of Neurophysiology. 2015 In Press.
    1. Ilin V, et al. Fast Computations in Cortical Ensembles Require Rapid Initiation of Action Potentials. Journal of Neuroscience. 2013;33(6):2281–2292.
    1. Kaltenbach JA. Tinnitus: Models and mechanisms. Hear Res. 2011;276(1–2):52–60.
    1. Wu c, Martel D, Shore S. Indreased synchrony and bursting of dorsal cochlear nucleus fusiform cells correlates with tinnitus. Journal of Neuroscience. 2015 In Press.
    1. Bauer CA, et al. Tinnitus and inferior colliculus activity in chinchillas related to three distinct patterns of cochlear trauma. J Neurosci Res. 2008;86(11):2564–78.
    1. Robertson D, Irvine DR. Plasticity of frequency organization in auditory cortex of guinea pigs with partial unilateral deafness. J Comp Neurol. 1989;282(3):456–71.
    1. Norena AJ, Tomita M, Eggermont JJ. Neural changes in cat auditory cortex after a transient pure-tone trauma. J Neurophysiol. 2003;90(4):2387–401.
    1. Basura GJ, Koehler SD, Shore SE. Bimodal Stimulus Timing Dependent Plasticity in Primary Auditory Cortex is Altered After Noise Exposure With and Without Tinnitus. J Neurophysiol. 2015 p. jn 00319 2015.
    1. Wang H, et al. Plasticity at glycinergic synapses in dorsal cochlear nucleus of rats with behavioral evidence of tinnitus. Neuroscience. 2009;164(2):747–59.
    1. Middleton JW, et al. Mice with behavioral evidence of tinnitus exhibit dorsal cochlear nucleus hyperactivity because of decreased GABAergic inhibition. Proc Natl Acad Sci U S A. 2011;108(18):7601–6.
    1. Zeng C, et al. Cochlear damage changes the distribution of vesicular glutamate transporters associated with auditory and nonauditory inputs to the cochlear nucleus. J Neurosci. 2009;29(13):4210–7.
    1. Barker M, et al. Acoustic overexposure increases the expression of VGLUT-2 mediated projections from the lateral vestibular nucleus to the dorsal cochlear nucleus. PLoS One. 2012;7(5):e35955.
    1. Zeng C, et al. Somatosensory projections to cochlear nucleus are upregulated after unilateral deafness. J Neurosci. 2012;32(45):15791–801.
    1. Schofield BR, et al. Projections from the dorsal and ventral cochlear nuclei to the medial geniculate body. Front Neuroanat. 2014;8:10.
    1. Sametsky EA, et al. Enhanced GABAA-Mediated Tonic Inhibition in Auditory Thalamus of Rats with Behavioral Evidence of Tinnitus. J Neurosci. 2015;35(25):9369–80.
    1. Llinas RR, et al. Thalamocortical dysrhythmia: A neurological and neuropsychiatric syndrome characterized by magnetoencephalography. Proc Natl Acad Sci USA. 1999;96:15222–15227.
    1. Tzounopoulos T, et al. Cell-specific, spike timing-dependent plasticities in the dorsal cochlear nucleus. Nat Neurosci. 2004;7(7):719–25.
    1. Koehler SD, Shore SE. Stimulus-timing dependent multisensory plasticity in the guinea pig dorsal cochlear nucleus. PLoS One. 2013;8(3):e59828.
    1. Roberts PD, Leen TK. Anti-hebbian spike-timing-dependent plasticity and adaptive sensory processing. Front Comput Neurosci. 2010;4:156.
    1. Stefanescu RA, Shore SE. NMDA receptors mediate stimulus timing dependent plasticity and neural synchrony in dorsal cochlear nucleus. Front Syst Neurosci. 2015 In Press.
    1. Wu C, Martel D, Shore S. Transcutaneous induction of stimulus timing dependent plasticity in dorsal cochlear nucleus. Front Syst Neurosci. 2015 doi: 10.3389/fnsys.2015.00116.
    1. Kaltenbach JA, Zhang J. Intense sound-induced plasticity in the dorsal cochlear nucleus of rats: evidence for cholinergic receptor upregulation. Hear Res. 2007;226(1–2):232–43.
    1. Jin YM, et al. Effects of intense tone exposure on choline acetyltransferase activity in the hamster cochlear nucleus. Hear Res. 2006;216–217:168–75.
    1. D’Amour JA, Froemke RC. Inhibitory and excitatory spike-timing-dependent plasticity in the auditory cortex. Neuron. 2015;86(2):514–28.
    1. Tass PA, Popovych OV. Unlearning tinnitus-related cerebral synchrony with acoustic coordinated reset stimulation: theoretical concept and modelling. Biol Cybern. 2012;106(1):27–36.
    1. Talathi SS, Hwang DU, Ditto WL. Spike timing dependent plasticity promotes synchrony of inhibitory networks in the presence of heterogeneity. J Comput Neurosci. 2008;25(2):262–81.
    1. Tziridis K, et al. Noise Trauma Induced Neural Plasticity Throughout the Auditory System of Mongolian Gerbils: Differences between Tinnitus Developing and Non-Developing Animals. Front Neurol. 2015;6:22.
    1. Singer W, et al. Noise-induced inner hair cell ribbon loss disturbs central arc mobilization: a novel molecular paradigm for understanding tinnitus. Mol Neurobiol. 2013;47(1):261–79.
    1. Li S, Choi V, Tzounopoulos T. Pathogenic plasticity of Kv7.2/3 channel activity is essential for the induction of tinnitus. Proc Natl Acad Sci U S A. 2013;110(24):9980–5.
    1. Li S, Kalappa BI, Tzounopoulos T. Noise-induced plasticity of KCNQ2/3 and HCN channels underlies vulnerability and resilience to tinnitus. Elife. 2015;4
    1. Dehmel S, Cui YL, Shore SE. Cross-modal interactions of auditory and somatic inputs in the brainstem and midbrain and their imbalance in tinnitus and deafness. Am J Audiol. 2008;17(2):S193–209.
    1. Wu C, et al. Listening to another sense: somatosensory integration in the auditory system. Cell Tissue Res. 2014
    1. Haenggeli CA, et al. Projections from the spinal trigeminal nucleus to the cochlear nucleus in the rat. J Comp Neurol. 2005;484(2):191–205.
    1. Wright DD, Ryugo DK. Mossy fiber projections from the cuneate nucleus to the cochlear nucleus in the rat. J Comp Neurol. 1996;365(1):159–72.
    1. Zhan X, Pongstaporn T, Ryugo DK. Projections of the second cervical dorsal root ganglion to the cochlear nucleus in rats. J Comp Neurol. 2006;496(3):335–48.
    1. Zeng C, Shroff H, Shore SE. Cuneate and spinal trigeminal nucleus projections to the cochlear nucleus are differentially associated with vesicular glutamate transporter-2. Neuroscience. 2011;176:142–51.
    1. Shore SE, et al. Dorsal cochlear nucleus responses to somatosensory stimulation are enhanced after noise-induced hearing loss. Eur J Neurosci. 2008;27(1):155–68.
    1. Zhou J, Nannapaneni N, Shore S. Vessicular glutamate transporters 1 and 2 are differentially associated with auditory nerve and spinal trigeminal inputs to the cochlear nucleus. J Comp Neurol. 2007;500(4):777–87.
    1. Heeringa A, et al. Altered vesicular glutamate transporter distributions in the mouse cochlear nucleus following cochlear insult. Neuroscience. 2015 In Press.
    1. Basura GJ, Koehler SD, Shore SE. Multi-sensory integration in brainstem and auditory cortex. Brain Res. 2012;1485:95–107.
    1. Sanchez TG, Rocha CB. Diagnosis and management of somatosensory tinnitus: review article. Clinics (Sao Paulo) 2011;66(6):1089–94.
    1. Levine RA, Abel M, Cheng H. CNS somatosensory-auditory interactions elicit or modulate tinnitus. Exp Brain Res. 2003;153(4):643–648.
    1. Zhou J, Shore S. Projections from the trigeminal nuclear complex to the cochlear nuclei: a retrograde and anterograde tracing study in the guinea pig. J Neurosci Res. 2004;78(6):901–7.
    1. Zhou J, Shore S. Convergence of spinal trigeminal and cochlear nucleus projections in the inferior colliculus of the guinea pig. J Comp Neurol. 2006;495(1):100–12.
    1. Wallhausser-Franke E, et al. Expression of c-fos in auditory and non-auditory brain regions of the gerbil after manipulations that induce tinnitus. Exp Brain Res. 2003;153:649–654.
    1. Adjamian P, et al. Neuroanatomical abnormalities in chronic tinnitus in the human brain. Neurosci Biobehav Rev. 2014;45C:119–133.
    1. Lockwood AH, et al. The functional neuroanatomy of tinnitus: evidence for limbic system links and neural plasticity. Neurology. 1998;50(1):114–20.
    1. Pinchoff RJ, et al. Modulation of tinnitus by voluntary jaw movements. Am J Otol. 1998;19(6):785–9.
    1. Vanneste S, De Ridder D. The auditory and non-auditory brain areas involved in tinnitus. An emergent property of multiple parallel overlapping subnetworks. Front Syst Neurosci. 2012;6:31.
    1. Melcher JR, Knudson IM, Levine RA. Subcallosal brain structure: correlation with hearing threshold at supra-clinical frequencies (>8 kHz), but not with tinnitus. Hear Res. 2013;295:79–86.
    1. Landgrebe M, et al. Structural brain changes in tinnitus: grey matter decrease in auditory and non-auditory brain areas. Neuroimage. 2009;46(1):213–218.
    1. Muhlau M, et al. Structural brain changes in tinnitus. Cereb Cortex. 2006;16(9):1283–1288.
    1. Gu JW, et al. Tinnitus, diminished sound-level tolerance, and elevated auditory activity in humans with clinically normal hearing sensitivity. J Neurophysiol. 2010;104(6):3361–3370.
    1. Maudoux A, et al. Auditory resting-state network connectivity in tinnitus: a functional MRI study. PLoS One. 2012;7(5):e36222.
    1. Schlee W, et al. Mapping cortical hubs in tinnitus. BMC Biol. 2009;7:80.
    1. De Ridder D, et al. An integrative model of auditory phantom perception: Tinnitus as a unified percept of interacting separable subnetworks. Neurosci Biobehav Rev. 2014;44C:16–32.
    1. de Lafuente V, Romo R. Neuronal correlates of subjective sensory experience. Nat Neurosci. 2005;8(12):1698–703.
    1. Golm D, et al. Neural correlates of tinnitus related distress: an fMRI-study. Hear Res. 2013;295:87–99.
    1. Vanneste S, et al. The neural correlates of tinnitus-related distress. Neuroimage. 2010;52(2):470–80.
    1. Schecklmann M, et al. Auditory cortex is implicated in tinnitus distress: a voxel-based morphometry study. Brain Struct Funct. 2013;218(4):1061–70.
    1. Vanneste S, et al. The difference between uni- and bilateral auditory phantom percept. Clin Neurophysiol. 2011;122(3):578–87.
    1. Carpenter-Thompson JR, et al. Alterations of the emotional processing system may underlie preserved rapid reaction time in tinnitus. Brain Res. 2014;1567:28–41.
    1. Vanneste S, Heyning PV, Ridder DD. Contralateral parahippocampal gamma-band activity determines noise-like tinnitus laterality: a region of interest analysis. Neuroscience. 2011;199:481–490.
    1. Schecklmann M, et al. Neural correlates of tinnitus duration and distress: a positron emission tomography study. Hum Brain Mapp. 2013;34(1):233–40.
    1. Maudoux A, et al. Connectivity graph analysis of the auditory resting state network in tinnitus. Brain Res. 2012;1485:10–21.
    1. Roberts LE, Husain FT, Eggermont JJ. Role of attention in the generation and modulation of tinnitus. Neurosci Biobehav Rev. 2013;37(8):1754–73.
    1. Menon V, Uddin LQ. Saliency, switching, attention and control: a network model of insula function. Brain Struct Funct. 2010;214(5–6):655–67.
    1. Martinez-Devesa P, et al. Cognitive behavioural therapy for tinnitus. Cochrane Database Syst Rev. 2010;(9):CD005233.
    1. Cima RF, et al. Specialised treatment based on cognitive behaviour therapy versus usual care for tinnitus: a randomised controlled trial. Lancet. 2012;379(9830):1951–1959.
    1. Davis PB, Paki B, Hanley PJ. Neuromonics Tinnitus Treatment: third clinical trial. Ear Hear. 2007;28(2):242–59.
    1. Vanneste S, et al. Does enriched acoustic environment in humans abolish chronic tinnitus clinically and electrophysiologically? A double blind placebo controlled study. Hear Res. 2013;296:141–8.
    1. Okamoto H, et al. Listening to tailor-made notched music reduces tinnitus loudness and tinnitus-related auditory cortex activity. Proc Natl Acad Sci USA. 2010;107(3):1207–1210.
    1. Tass PA, et al. Counteracting tinnitus by acoustic coordinated reset neuromodulation. Restor Neurol Neurosci. 2012;30(2):137–59.
    1. Hoare DJ, et al. Sound therapy for tinnitus management: practicable options. J Am Acad Audiol. 2014;25(1):62–75.
    1. De Ridder D, et al. Safety and efficacy of vagus nerve stimulation paired with tones for the treatment of tinnitus: a case series. Neuromodulation. 2014;17(2):170–9.
    1. Hoare DJ, et al. Amplification with hearing aids for patients with tinnitus and co-existing hearing loss. Cochrane Database Syst Rev. 2014;1:CD010151.
    1. Schaette R, et al. Acoustic stimulation treatments against tinnitus could be most effective when tinnitus pitch is within the stimulated frequency range. Hear Res. 2010;269(1–2):95–101.
    1. McNeill C, et al. Tinnitus pitch, masking, and the effectiveness of hearing aids for tinnitus therapy. Int J Audiol. 2012;51(12):914–9.
    1. Langguth B, Elgoyhen AB. Current pharmacological treatments for tinnitus. Expert Opin Pharmacother. 2012;13(17):2495–509.
    1. Zoger S, Svedlund J, Holgers KM. The effects of sertraline on severe tinnitus suffering–a randomized, double-blind, placebo-controlled study. J Clin Psychopharmacol. 2006;26(1):32–39.
    1. Baldo P, et al. Antidepressants for patients with tinnitus. Cochrane Database Syst Rev. 2012;9:CD003853.
    1. Hoekstra CE, et al. Anticonvulsants for tinnitus. Cochrane Database Syst Rev. 2011;(7):CD007960.
    1. Han SS, et al. Clonazepam Quiets tinnitus: a randomised crossover study with Ginkgo Biloba. J Neurol Neurosurg Psychiatry. 2012;83(8):821–827.
    1. Tunkel DE, et al. Clinical practice guideline: tinnitus. Otolaryngol Head Neck Surg. 2014;151(2 Suppl):S1–S40.
    1. Zenner HP, et al. On the interdisciplinary S3 guidelines for the treatment of chronic idiopathic tinnitus. HNO. 2015;63(6):419–27.
    1. van de Heyning P, et al. Efficacy and safety of AM-101 in the treatment of acute inner ear tinnitus–a double-blind, randomized, placebo-controlled phase II study. Otol Neurotol. 2014;35(4):589–97.
    1. Hebert S, Fournier P, Norena A. The Auditory Sensitivity is Increased in Tinnitus Ears. J Neurosci. 2013;33(6):2356–64.
    1. Diesch E, et al. Interaction among the components of multiple auditory steady-state responses: enhancement in tinnitus patients, inhibition in controls. Neuroscience. 2010;167(2):540–53.
    1. Wienbruch C, et al. Frequency organization of the 40-Hz auditory steady-state response in normal hearing and in tinnitus. Neuroimage. 2006;33(1):180–94.
    1. Langers DR, de Kleine E, van Dijk P. Tinnitus does not require macroscopic tonotopic map reorganization. Front Syst Neurosci. 2012;6:2.
    1. Roberts LE, et al. Evidence for differential modulation of primary and nonprimary auditory cortex by forward masking in tinnitus. Hear Res. 2015;327:9–27.
    1. Paul BT, et al. Modulation of electrocortical brain activity by attention in individuals with and without tinnitus. Neural Plast. 2014;2014:127824.
    1. Jastreboff PJ. Phantom auditory perception (tinnitus): mechanisms of generation and perception. Neurosci Res. 1990;8:221–254.
    1. Leaver AM, et al. Cortico-limbic morphology separates tinnitus from tinnitus distress. Front Syst Neurosci. 2012;6:21.
    1. Weisz N, et al. The neural code of auditory phantom perception. J Neurosci. 2007;27(6):1479–84.
    1. Weisz N, et al. Tinnitus perception and distress is related to abnormal spontaneous brain activity as measured by magnetoencephalography. PLoS Med. 2005;2(6):e153.
    1. Adjamian P, et al. Neuromagnetic indicators of tinnitus and tinnitus masking in patients with and without hearing loss. J Assoc Res Otolaryngol. 2012;13(5):715–31.
    1. Pierzycki RH, et al. Whole scalp resting state EEG of oscillatory brain activity shows no parametric relationship with psychoacoustic and psychosocial assessment of tinnitus: A repeated measures study. Hear Res. 2015
    1. Sedley W, et al. Intracranial Mapping of a Cortical Tinnitus System using Residual Inhibition. Curr Biol. 2015;25(9):1208–14.

Source: PubMed

3
Prenumerera