Exploring the Underlying Causes and Sustaining Factors of Subjective Tinnitus: An Insight into its Pathophysiology

This article explores the underlying causes and sustaining factors of subjective tinnitus, providing insight into its pathophysiology. It aims to raise awareness about the mechanisms that contribute to the development and maintenance of tinnitus and provide a deeper understanding of this condition

Exploring the Underlying Causes and Sustaining Factors of Subjective Tinnitus

Tinnitus is a condition where people hear a sound in their ears, but there is no actual external noise causing it. This phantom sound is a common problem, and researchers are trying to figure out what causes it. This study looked at published research to understand what we know about tinnitus and what still needs to be studied. The researchers found that tinnitus is a complex issue that affects both the hearing system and other parts of the brain. They concluded that it is important to continue to study tinnitus in order to better treat it.


This is the  more indepth version of Does Tinnitus Affect the Brain?

Researched Fact Checked: Ncbi and Frontiers

Finding Relevant Literature for Tinnitus Research: A Guide to Identifying and Choosing Appropriate Sources

Tinnitus is a subjective phantom sound perceived in the absence of external or internal stimuli and affects a large number of people worldwide. Its classification system is based on various criteria such as causes, comorbidities, symptom characteristics, and psychological burden. The most common form of tinnitus is associated with a hearing loss and is further classified into primary (associated with sensorineural hearing loss or idiopathic) and secondary (related to other causes such as organic). Tinnitus duration (acute or chronic) and the description of the tinnitus sound (continuous or intermittent, pulsatile or non-pulsatile) are also used in classifying tinnitus.

The functional and psychological impact of tinnitus are also important factors, and various questionnaires have been designed to assess this, such as the Tinnitus Handicap Inventory, Tinnitus Questionnaire, Tinnitus Functional Index, and Tinnitus Primary Function Questionnaire.

The origin of tinnitus is related to aberrant neural activity at certain levels of the auditory system, and tinnitus can be classified as peripheral or central depending on the level of aberrant neural activity. The distinction between peripheral and central tinnitus is not completely independent, and the latest research suggests that tinnitus may result from a combination of both peripheral and central mechanisms. The exact pathophysiology of tinnitus remains unclear and is the subject of ongoing research.



In 2017, a literature search was done to gather information about the pathophysiology of subjective chronic tinnitus. Three literature search platforms were used - PubMed, Medline and Web of Science - and the search terms pathophysiology and subjective chronic tinnitus were used. The search resulted in 373 records, after removing duplicates, 168 records were left for further review. After screening, 121 full texts were eligible for the review, and 50 additional records were found through a manual search. The information was then extracted and assessed by two team members, and in case of disagreement, a third member was consulted.


Demographic Traits Reflecting the Pathophysiology of Tinnitus

Studies have indicated that in about 30% of tinnitus cases, the cause is undetermined. Tinnitus is commonly associated with noise-induced hearing loss and presbycusis, and about 90% of people with tinnitus in the UK have some form of hearing loss. Other risk factors for tinnitus have been identified, such as vascular disease, hypertension, diabetes, autoimmune disorders, head injury, and degenerative neural disorders.

Contrasting Neurophysiological Findings in Animals and Humans

This section is discussing the comparison between animal and human studies on the pathophysiology of tinnitus. The advantages of using animal models in tinnitus research include the ability to control the etiology and to apply a wide range of experimental tools, but there are also disadvantages such as the lack of a standardized animal model and the difficulty in translating findings from animal models to humans. Animal studies typically focus on tinnitus as a consequence of acute peripheral lesion associated with severe hearing loss, while human neuroimaging studies tend to emphasize the role of auditory thalamus and auditory cortex in the chronification and maintenance of tinnitus. The limitations of measuring techniques in human studies also bring important caveats for drawing analogies between animal and human findings.



The current understanding of tinnitus suggests that it is a complex interaction between peripheral and central mechanisms within the auditory pathway. It is widely believed that tinnitus reflects a pathology of neural plasticity with both a molecular and systemic component. The molecular component has a cochlear origin and is related to the initiation phase of tinnitus, while the systemic component has a central aspect associated with the long-term maintenance of tinnitus.

It has been suggested that peripheral tinnitus may originate from the dysfunction of cochlear outer hair cells (OHCs) leading to increased spontaneous cochlear activity. Meanwhile, central tinnitus is mediated by the neuronal activity in the auditory centers. This central activity is often triggered by a reduction of cochlear activity, but damage to cochlear tissues is not necessary to produce central changes related to tinnitus.

Locations of Tinnitus Origin: A Study of its Generation

Based on these observations, tinnitus is classified into three distinct subtypes: cochlear tinnitus, peripheral-dependent central tinnitus, and peripheral-independent central tinnitus. Cochlear tinnitus is generated by aberrant activity in the inner ear, which is propagated through the cochlear nerve and the central auditory pathway. Peripheral-dependent central tinnitus is associated with cochlear spontaneous activity, while peripheral independent central tinnitus is independent from cochlear spontaneous activity.

In conclusion, the generation of tinnitus is a complex process that involves both peripheral and central mechanisms within the auditory pathway. Further research is needed to fully understand the mechanisms behind tinnitus and to develop effective treatments.

Exploring the Cellular Processes Behind Tinnitus

There are several cellular mechanisms that are believed to contribute to the development of tinnitus. Some of these mechanisms are the loss of outer hair cell (OHC) electromotility, damage to the stereocilia bundle, death of OHCs or inner hair cells (IHCs), or rupture of the basilar membrane. This can lead to a decrease in neuronal output from the cochlea to the brain and the potential for compensation mechanisms in the brain.

Examining the Role of Tectorial Membrane in Tinnitus

Change in the position of the tectorial membrane can also be a pathophysiological trigger for tinnitus. For example, intense noise exposure can alter the rootlets of stereocilia, leading to stiffness and an increase in cochlear spontaneous activity.

Investigating the Function of Outer Hair Cells (OHCs) in Tinnitus

Damage to the stereocilia of OHCs is another potential trigger for tinnitus, often following an intense noise exposure. The initiation of the pathological process begins at the stereocilia of OHCs, with two fundamental processes being damaged by the noise: intracellular calcium levels and biochemical changes of their structural proteins.

Exploring the Interaction between Inner Hair Cells (IHCs) and Cochlear NMDA Receptors in Tinnitus

The N-methyl-D-aspartate (NMDA) receptor has also been found to play a role in noise-induced tinnitus. NMDA receptors predominate on the modiolar side of IHCs, and an increase in glutamate levels derived from IHCs can activate these receptors and lead to excessive calcium influx in the dendrites of the spiral ganglion neurons, resulting in an over-excitation of NMDA-receptors and tinnitus.

Understanding the Impact of Endocochlear Potential Increase on Tinnitus

An increase in the endocochlear potential can also depolarize IHCs, leading to a sequence of events that includes the opening of voltage-gated calcium channels, an intracellular influx of calcium, and the fusion of the synaptic ribbon to the plasmatic membrane, resulting in glutamate release and depolarization of cochlear fibers. This process can be induced by acute noise trauma that reduces the opening probability of OHC's mechano-electrical transduction channels, leading to an increase in the endocochlear potential.

Finally, biochemical changes, such as the heat-shock protein group (stress proteins) that interacts with structural proteins of hair cells, can also contribute to the development of tinnitus. Any disturbance that causes a deficient heat-shock protein system response can lead to the onset of tinnitus.

Investigating the Role of Cochlear Synaptopathy in Tinnitus

The contribution of cochlear synaptopathy to tinnitus is still unclear and the subject of ongoing research. Cochlear synaptopathy refers to the loss of synapses between the inner hair cells (IHCs) and the cochlear nerve fibers, which can occur due to external factors such as noise exposure or aging. This condition is sometimes referred to as hidden hearing loss as it cannot be diagnosed through conventional tonal audiometry. Studies have shown a reduced amplitude of wave 1 in the auditory brainstem response (ABR) in people with tinnitus and normal hearing, which has been interpreted as evidence for reduced cochlear nerve output as a result of cochlear synaptopathy. However, this finding has not been consistently replicated, and other factors such as noise exposure may also play a role in tinnitus. Further research is needed to better understand the relationship between cochlear synaptopathy and tinnitus, and to develop improved methods for detecting this condition in humans.

Unraveling the Mechanisms Behind the Persistence of Tinnitus

The maintenance of tinnitus is thought to involve several mechanisms, many of which are associated with hearing loss. Research has shown that there is a strong association between high-pitched tinnitus and high-frequency sensorineural hearing loss (SNHL), suggesting that hearing loss is a main cause of tinnitus (Norena et al., 2002; Martines et al., 2010a,b,c; Sereda et al., 2011).

Studies have shown that the underlying cause of tinnitus may be associated with damage to the sensory cochlear epithelium (Henry et al., 2005), and this damage can result from various kinds of peripheral insults to the auditory system such as cochlear ablation, selective loss of inner hair cells (IHCs) or outer hair cells (OHCs), or mixed or incomplete IHC and OHC injuries. These insults can reduce cochlear output, which may then trigger a cascade of neuromodulatory events ultimately causing hyperactivity in central auditory circuits (central gain).

Reduced cochlear output through hearing loss may modify the gain of central neurons, resulting in increased spontaneous activity, and this increased activity has been proposed to contribute to tinnitus (Parra and Pearlmutter, 2007). The functional aberrations resulting from either the tonotopic over-representation, enhanced synchronicity, or elevated spontaneous firing rates may underlie the induction of tinnitus (Adjamian et al., 2014).

Figure 2 is a representation of the pathophysiology of tinnitus and how it relates to pain and phantom limb perception. The figure explains that damage to the cochlea, such as hair cell loss or synaptic damages, leads to a frequency-specific decrease in output from the cochlear nerve. In response, the central auditory pathway compensates by increasing its activity, leading to increased gain and the perception of a non-existing sound, which is tinnitus.

In addition to the auditory pathway, tinnitus is thought to share non-auditory networks similar to those in chronic pain, such as perception, salience, distress, and memory. These networks can maintain tinnitus in the absence of the initial tinnitus-initiator.

The model also suggests that tinnitus and phantom pain share underlying mechanisms, such as sensory deafferentation resulting in activity in the primary and secondary auditory cortices. This activity becomes a conscious percept when connected to larger brain networks located in the frontal and parietal cortex, such as the self-awareness and salience network. The salience network intersects with the central autonomic control system and is essential for consciously maintaining the phantom perception.

The phantom perception may also be associated with distress and activate non-specific distress networks located in the anterior cingulate cortex, anterior insula, and amygdala. It is also proposed that memory mechanisms may reinforce and maintain awareness of the phantom percept.


Exploring the Central Processes in Tinnitus

Tinnitus has been linked to changes in the central nervous system, in particular, to the phenomenon of homeostatic plasticity. This occurs when auditory neurons in the brain attempt to maintain a neuronal network similar to the one that existed before peripheral damage occurred.

The posteroventral cochlear nucleus (PVCN), the inferior colliculus (IC), the dorsal cochlear nucleus (DCN), and the paraflocculus lobe of the cerebellum (PFL) have been suggested as possible neuronal correlates of tinnitus. There is evidence that elevated responses to sound are common in individuals with tinnitus, particularly in the IC.

Studies in animals have shown increased activity in fusiform neurons of the DCN during noise-induced tinnitus, and this area has been suggested as an important site of maladaptive auditory-somatosensory plasticity. However, a specific drug compound designed to modulate the Kv7.2/3 channel, which has been found to show decreased activity in the DCN after noise-induced tinnitus, has not been found to alleviate tinnitus in humans.

Exploring the Central Processes in Tinnitus

Tinnitus is complex and is still not fully understood. There are several theories on the potential causes of tinnitus, including neuronal hyperactivity in certain regions of the central auditory system and changes in the auditory cortex. One theory suggests that decreased neural input to the cochlear amplifier may lead to increased spontaneous activity and a chain reaction of neuroplastic changes in the afferent auditory relays, eventually leading to tinnitus. Another theory focuses on the brainstem as the place of integration of efferent neuronal drive and afferent tinnitus-related stimuli. Some studies have suggested that the medial olivocochlear bundle may play a role in tinnitus onset, while others have not confirmed this finding.

Electrophysiological measurements have revealed fundamental differences between people with tinnitus and healthy controls, including hyperactivity in the gamma frequency range within the temporal cortex. There have also been observations of aberrant neuronal oscillations in the alpha and gamma frequency range within the frontal cortex. The correlation between tinnitus perception and the frequency band power in EEG and MEG remains unclear, with some studies finding no clear relationship. Acoustic stimulation studies have shown that tinnitus is associated with changes in the delta/theta and gamma frequency bands, although the findings in this area are mixed and not yet fully understood. Overall, while progress has been made in understanding the potential causes of tinnitus, further research is needed to fully understand this complex phenomenon.

Tinnitus complex and multi-faceted involves a range of auditory and non-auditory structures within the brain. In terms of auditory structures, the auditory pathway, including the inner ear and central auditory nuclei, has been shown to be affected in tinnitus, with changes seen in central gain that are related to loss of GABAergic inhibition and decreased activity of specific potassium channels (Kv7.2/3).

Investigating the Non-Auditory Neuronal Networks in the Development of Tinnitus

In addition to the auditory structures, recent research supports the involvement of emotional and cognitive relays of the brain, such as the temporal, parietal, sensorimotor, and limbic cortex, in the pathophysiology of tinnitus. These regions include the medial prefrontal cortex and ventromedial parts of the basal ganglia, dorsal prefrontal regions, the insula, thalamus, anterior and posterior cingulate, amygdala, parahippocampus, hippocampus, and the subcallosal region, including the nucleus accumbens. The precise role of these extra-auditory structures in tinnitus is difficult to determine and may vary depending on the individual tinnitus profile.


There is evidence of a tight interaction between limbic non-auditory and auditory pathways, as well as anatomical and functional abnormalities, in chronic tinnitus, as seen through various neuroimaging techniques. However, not all studies have been able to find significant differences in the connectivity of the auditory network between control and tinnitus groups. It is important to note that the relationship between the psychoacoustic tinnitus characteristics, the degree of tinnitus distress, and underlying neural patterns of activity is not yet scientifically confirmed and requires further study.

The findings of the study by Daftary et al. (2004) suggest that the reduction in benzodiazepine receptor density in the frontal lobes and the cerebellum of patients with severe chronic tinnitus might play a role in the development and maintenance of tinnitus. The frontal lobes are part of the frontostriatal circuit, which appears to have a central role in the evaluation of sensory stimuli and the management of information flow, as discussed in previous research (Rauschecker et al., 2015). The cerebellum, on the other hand, is known for its role in motor control and coordination, as well as its involvement in cognitive and emotional processes (D'Angelo and De Zeeuw, 2009).

The role of the thalamus, particularly the medial geniculate body (MGB), has also been implicated in the pathology of tinnitus (Caspary and Llano, 2017). The MGB is thought to act as a gatekeeper, regulating the perception of sound on its way to the auditory cortex and the limbic system. Abnormalities in the thalamocortical circuits, as well as in the limbic and auditory cortices, have been suggested to contribute to the development and maintenance of tinnitus (Winer et al., 1999; Rauschecker et al., 2010; Leaver et al., 2011).

Moreover, the potential role of GABA, a neurotransmitter involved in fast synaptic inhibition and persistent tonic inhibition, has been explored in the context of tinnitus. Two opposing hypotheses have been proposed, one suggesting an up-regulation of GABAergic inhibition and the other a suppression of GABAergic inhibition. These observations highlight the complex interplay between different brain regions and their associated neurotransmitter systems in the development and maintenance of tinnitus.

Investigating the Non-Auditory Neuronal Networks in the Development of Tinnitus

These studies show that there is evidence of structural changes in the brain in individuals with tinnitus and/or hearing loss when compared to a control group. However, there is still some variability in the results obtained, which highlights the need for greater standardization in study design and analysis techniques, as well as more precise subtyping of the condition. The left primary auditory cortex and other non-auditory brain structures appear to play a role in the development of tinnitus, but the exact nature of these changes and their relationship to tinnitus and hearing loss is still not fully understood.

In summary, tinnitus is thought to be related to disruptions in neural synchrony between different cortical networks, including the thalamus. The edge effect theory suggests that a steep audiometric edge between normal and impaired hearing may lead to changes in neural synchrony and the development of tinnitus. However, some studies have found tinnitus-related changes in the magnitude of oscillatory power in different frequency bands beyond just the auditory cortex, suggesting a more diffuse network-based interaction between tinnitus perception and memory processes. Another theory is that changes in alpha power in tinnitus patients reflect an enhancement or reduction of the excitability of engaged neuronal networks. Further research is needed to fully understand the pathophysiology of tinnitus and its underlying mechanisms.


Examining the Theoretical Approaches and Models of Tinnitus Pathophysiology

This model suggests that tinnitus may be a result of the auditory system’s predictive coding process, where the auditory subcortex produces spontaneous activity (the tinnitus precursor) which is normally ignored against the prevailing percept of silence. However, in some individuals with tinnitus, the tinnitus precursor becomes more noticeable and the individual perceives a continuous sound even in silence. This model has some similarities with other theories that propose a role for increased neural synchrony in tinnitus development, but the sensory precision tinnitus model also accounts for the graded processing of spontaneous sensory input and the influence of cognitive factors in tinnitus perception.

While this model provides a comprehensive framework to explain tinnitus pathophysiology, it is important to note that this model may not be applicable to all forms of chronic pain, such as central post-stroke pain, and may need to be modified or refined to account for these conditions. The role of predictive coding in tinnitus and chronic pain is an active area of research, and further studies are needed to fully understand the underlying mechanisms of these conditions.



The cognitive-behavioral model of tinnitus proposes that the way a person thinks about and interprets their tinnitus has a significant impact on the level of distress they experience. This model suggests that negative thoughts, beliefs and attitudes about tinnitus can lead to increased anxiety and avoidance behaviors, which can exacerbate the distress associated with tinnitus. The model proposes that by changing the way a person thinks about their tinnitus and by teaching them coping skills, they can reduce the level of distress they experience.

The fear-avoidance model of tinnitus posits that tinnitus-related distress arises from the avoidance of activities and situations that trigger the perception of tinnitus. This model suggests that a person with tinnitus may experience increased anxiety when they encounter sounds that are similar to their tinnitus, leading them to avoid these sounds. Over time, this avoidance can lead to a decrease in the person's quality of life and an increase in their tinnitus-related distress.

Overall, the psychological models of tinnitus provide a framework for understanding the relationship between the cognitive and emotional aspects of tinnitus and their impact on the level of distress experienced by individuals with tinnitus. These models can inform the development of effective treatment strategies, such as cognitive behavioral therapy, that target the psychological components of tinnitus.

Conclusion

In conclusion, tinnitus is a complex and multi-faceted phenomenon that involves both peripheral and central auditory systems, as well as attentional, memory, and emotional systems. Despite the significant advances made in our understanding of tinnitus, much remains to be discovered about its underlying mechanisms. However, the current view of tinnitus is that it is a symptom encompassing a distributed network, and the restoration of cochlear output to the brain may abolish tinnitus. There is a growing body of evidence supporting the use of various therapeutic approaches, including molecular interventions, systemic approaches, and hybrid solutions, for the treatment of tinnitus. However, the therapy of tinnitus will have to be strictly individualized, taking into account the phase of tinnitus pathophysiology, the level of lesion, and the mechanism of tinnitus maintenance. The multidisciplinary approach will be the key to providing optimal care for tinnitus patients, with the goal of improving their subjective well-being.

Acknowledgements

https://www.news-medical.net/health/Does-Tinnitus-Affect-the-Brain.aspx#:~:text=Tinnitus%20causes%20changes%20in%20brain,can%20negatively%20impact%20the%20brain.  

https://www.frontiersin.org/articles/10.3389/fnins.2018.00866/full  

https://www.nidcd.nih.gov/health/tinnitus  


original artwork Fernando Vilhena de Mendonça MD,in Figures 2, 3.

Footnotes

^ https://clinicaltrials.gov/ct2/show/NCT02315508?term=QUIET-1&rank=1 

^ https://autifony.com/wp-content/uploads/2017/10/AUTIFONY-CLARITY-1-RESULTS-08-Aug-2016-FINAL.pdf 

References

Adamchic, I., Hauptmann, C., and Tass, P. A. (2012). Changes of oscillatory activity in pitch processing network and related tinnitus relief induced by acoustic CR neuromodulation. Front. Syst. Neurosci. 6:18. doi: 10.3389/fnsys.2012.00018


PubMed Abstract | CrossRef Full Text | Google Scholar


Adamchic, I., Toth, T., Hauptmann, C., and Tass, P. A. (2014). Reversing pathologically increased EEG power by acoustic coordinated reset neuromodulation. Hum. Brain Mapp. 35, 2099–2118. doi: 10.1002/hbm.22314


PubMed Abstract | CrossRef Full Text | Google Scholar


Adjamian, P., Hall, D. A., Palmer, A. R., Allan, T. W., and Langers, D. R. (2014). Neuroanatomical abnormalities in chronic tinnitus in the human brain. Neurosci. Biobehav. Rev. 45, 119–133. doi: 10.1016/j.neubiorev.2014.05.013


PubMed Abstract | CrossRef Full Text | Google Scholar


Adjamian, P., Sereda, M., and Hall, D. A. (2009). The mechanisms of tinnitus: perspectives from human functional neuroimaging. Hear. Res. 253, 15–31. doi: 10.1016/j.heares.2009.04.001


PubMed Abstract | CrossRef Full Text | Google Scholar


Adjamian, P., Sereda, M., Zobay, O., Hall, D. A., and Palmer, A. R. (2012). Neuromagnetic indicators of tinnitus and tinnitus masking in patients with and without hearing loss. J. Assoc. Res. Otolaryngol. 13, 715–731. doi: 10.1007/s10162-012-0340-5


PubMed Abstract | CrossRef Full Text | Google Scholar


Allan, T. W., Besle, J., Langers, D. R., Davies, J., Hall, D. A., Palmer, A. R., et al. (2016). Neuroanatomical alterations in tinnitus assessed with magnetic resonance imaging. Front. Aging Neurosci. 8:221. doi: 10.3389/fnagi.2016.00221


PubMed Abstract | CrossRef Full Text | Google Scholar


Auerbach, B. D., Rodrigues, P. V., and Salvi, R. J. (2014). Central gain control in tinnitus and hyperacusis. Front. Neurol. 5:206. doi: 10.3389/fneur.2014.00206


CrossRef Full Text | Google Scholar


Ayache, D., Earally, F., and Elbaz, P. (2003). Characteristics and postoperative course of tinnitus in otosclerosis. Otol. Neurotol. 24, 48–51. doi: 10.1097/00129492-200301000-00011


CrossRef Full Text | Google Scholar


Baguley, D., Mcferran, D., and Hall, D. (2013). Tinnitus. Lancet 382, 1600–1607. doi: 10.1016/S0140-6736(13)60142-7


CrossRef Full Text | Google Scholar


Baguley, D. M. (2002). Mechanisms of tinnitus. Br. Med. Bull. 63, 195–212. doi: 10.1093/bmb/63.1.195


CrossRef Full Text | Google Scholar


Bajo, V. M., Rouiller, E. M., Welker, E., Clarke, S., Villa, A. E., De Ribaupierre, Y., et al. (1995). Morphology and spatial distribution of corticothalamic terminals originating from the cat auditory cortex. Hear. Res. 83, 161–174. doi: 10.1016/0378-5955(94)00199-Z


PubMed Abstract | CrossRef Full Text | Google Scholar


Bauer, C. A., and Brozoski, T. J. (2001). Assessing tinnitus and prospective tinnitus therapeutics using a psychophysical animal model. J. Assoc. Res. Otolaryngol. 2, 54–64. doi: 10.1007/s101620010030


PubMed Abstract | CrossRef Full Text | Google Scholar


Bing, D., Lee, S. C., Campanelli, D., Xiong, H., Matsumoto, M., Panford-Walsh, R., et al. (2015). Cochlear NMDA receptors as a therapeutic target of noise-induced tinnitus. Cell Physiol. Biochem. 35, 1905–1923. doi: 10.1159/000374000


PubMed Abstract | CrossRef Full Text | Google Scholar


Bojic, T., Perovic, V. R., Sencanski, M., and Glisic, S. (2017). Identification of candidate allosteric modulators of the m1 muscarinic acetylcholine receptor which may improve vagus nerve stimulation in chronic tinnitus. Front. Neurosci. 11:636. doi: 10.3389/fnins.2017.00636


PubMed Abstract | CrossRef Full Text | Google Scholar


Bonham, B. H., Cheung, S. W., Godey, B., and Schreiner, C. E. (2004). Spatial organization of frequency response areas and rate/level functions in the developing AI. J. Neurophysiol. 91, 841–854. doi: 10.1152/jn.00017.2003


PubMed Abstract | CrossRef Full Text | Google Scholar


Boyen, K., Langers, D. R., De Kleine, E., and Van Dijk, P. (2013). Gray matter in the brain: differences associated with tinnitus and hearing loss. Hear. Res. 295, 67–78. doi: 10.1016/j.heares.2012.02.010


PubMed Abstract | CrossRef Full Text | Google Scholar


Brozoski, T. J., and Bauer, C. A. (2016). Animal models of tinnitus. Hear. Res. 338, 88–97. doi: 10.1016/j.heares.2015.10.011


PubMed Abstract | CrossRef Full Text | Google Scholar


Brozoski, T. J., Bauer, C. A., and Caspary, D. M. (2002). Elevated fusiform cell activity in the dorsal cochlear nucleus of chinchillas with psychophysical evidence of tinnitus. J. Neurosci. 22, 2383–2390. doi: 10.1523/JNEUROSCI.22-06-02383.2002


PubMed Abstract | CrossRef Full Text | Google Scholar


Brozoski, T. J., Wisner, K. W., Sybert, L. T., and Bauer, C. A. (2012). Bilateral dorsal cochlear nucleus lesions prevent acoustic-trauma induced tinnitus in an animal model. J. Assoc. Res. Otolaryngol. 13, 55–66. doi: 10.1007/s10162-011-0290-3


PubMed Abstract | CrossRef Full Text | Google Scholar


Cacace, A. T., Brozoski, T., Berkowitz, B., Bauer, C., Odintsov, B., Bergkvist, M., et al. (2014). Manganese enhanced magnetic resonance imaging (MEMRI): a powerful new imaging method to study tinnitus. Hear. Res. 311, 49–62. doi: 10.1016/j.heares.2014.02.003


PubMed Abstract | CrossRef Full Text | Google Scholar


Caspary, D. M., and Llano, D. A. (2017). Auditory thalamic circuits and GABAA receptor function: putative mechanisms in tinnitus pathology. Hear. Res. 349, 197–207. doi: 10.1016/j.heares.2016.08.009


PubMed Abstract | CrossRef Full Text | Google Scholar


Chen, G. D., and Fechter, L. D. (2003). The relationship between noise-induced hearing loss and hair cell loss in rats. Hear. Res. 177, 81–90. doi: 10.1016/S0378-5955(02)00802-X


CrossRef Full Text | Google Scholar


Cheung, S. W., and Larson, P. S. (2010). Tinnitus modulation by deep brain stimulation in locus of caudate neurons (area LC). Neuroscience 169,1768–1778. doi: 10.1016/j.neuroscience.2010.06.007


PubMed Abstract | CrossRef Full Text | Google Scholar


Cima, R. F. F. (2018). Bothersome tinnitus: cognitive behavioral perspectives. HNO 66, 369–374. doi: 10.1007/s00106-018-0502-9


PubMed Abstract | CrossRef Full Text | Google Scholar


Daftary, A., Shulman, A., Strashun, A. M., Gottschalk, C., Zoghbi, S. S., and Seibyl, J. P. (2004). Benzodiazepine receptor distribution in severe intractable tinnitus. Int. Tinnitus J. 10, 17–23.


PubMed Abstract | Google Scholar


Davies, J., Gander, P. E., Andrews, M., and Hall, D. A. (2014). Auditory network connectivity in tinnitus patients: a resting-state fMRI study. Int. J. Audiol. 53, 192–198. doi: 10.3109/14992027.2013.846482


PubMed Abstract | CrossRef Full Text | Google Scholar


Davis, A., and Rafaie, E. A. (2000). “Epidemiology of tinnitus,” in Tinnitus Handbook, ed. R. S. Tyler (San Diego, CA: Singular).


Google Scholar


De Ridder, D., Congedo, M., and Vanneste, S. (2015). The neural correlates of subjectively perceived and passively matched loudness perception in auditory phantom perception. Brain Behav. 5:e00331. doi: 10.1002/brb3.331


PubMed Abstract | CrossRef Full Text | Google Scholar


De Ridder, D., Elgoyhen, A. B., Romo, R., and Langguth, B. (2011a). Phantom percepts: tinnitus and pain as persisting aversive memory networks. Proc. Natl. Acad. Sci. U.S.A. 108, 8075–8080. doi: 10.1073/pnas.1018466108


PubMed Abstract | CrossRef Full Text | Google Scholar


De Ridder, D., Vanneste, S., and Congedo, M. (2011b). The distressed brain: a group blind source separation analysis on tinnitus. PLoS One 6:e24273. doi: 10.1371/journal.pone.0024273


PubMed Abstract | CrossRef Full Text | Google Scholar


De Ridder, D., Vanneste, S., Weisz, N., Londero, A., Schlee, W., Elgoyhen, A. B., et al. (2014). An integrative model of auditory phantom perception: tinnitus as a unified percept of interacting separable subnetworks. Neurosci. Biobehav. Rev. 44, 16–32. doi: 10.1016/j.neubiorev.2013.03.021


PubMed Abstract | CrossRef Full Text | Google Scholar


Dechesne, C. J., Kim, H. N., Nowak, T. S. Jr., and Wenthold, R. J. (1992). Expression of heat shock protein, HSP72, in the guinea pig and rat cochlea after hyperthermia: immunochemical and in situ hybridization analysis. Hear. Res. 59, 195–204. doi: 10.1016/0378-5955(92)90116-5


PubMed Abstract | CrossRef Full Text | Google Scholar


Dehmel, S., Pradhan, S., Koehler, S., Bledsoe, S., and Shore, S. (2012). Noise overexposure alters long-term somatosensory-auditory processing in the dorsal cochlear nucleus–possible basis for tinnitus-related hyperactivity? J. Neurosci. 32, 1660–1671. doi: 10.1523/JNEUROSCI.4608-11.2012


PubMed Abstract | CrossRef Full Text | Google Scholar


Don, M., and Eggermont, J. J. (1978). Analysis of the click-evoked brainstem potentials in man unsing high-pass noise masking. J. Acoust. Soc. Am. 63, 1084–1092. doi: 10.1121/1.381816


PubMed Abstract | CrossRef Full Text | Google Scholar


Dong, S., Mulders, W. H., Rodger, J., Woo, S., and Robertson, D. (2010). Acoustic trauma evokes hyperactivity and changes in gene expression in guinea-pig auditory brainstem. Eur. J. Neurosci. 31, 1616–1628. doi: 10.1111/j.1460-9568.2010.07183.x


PubMed Abstract | CrossRef Full Text | Google Scholar


Drevets, W. C., Price, J. L., and Furey, M. L. (2008). Brain structural and functional abnormalities in mood disorders: implications for neurocircuitry models of depression. Brain Struct. Funct. 213, 93–118. doi: 10.1007/s00429-008-0189-x


PubMed Abstract | CrossRef Full Text | Google Scholar


Eggermont, J. J. (2000). “Physiological mechanisms and neural models,” in Tinnitus handbook, ed. R. S. Tyler (San Diego, CA: Singular).


Google Scholar


Eggermont, J. J. (2005). Tinnitus: neurobiological substrates. Drug Discov. Today 10, 1283–1290. doi: 10.1016/S1359-6446(05)03542-7


PubMed Abstract | CrossRef Full Text | Google Scholar


Eggermont, J. J. (2007). Pathophysiology of tinnitus. Prog. Brain Res. 166, 19–35. doi: 10.1016/S0079-6123(07)66002-6


CrossRef Full Text | Google Scholar


Eggermont, J. J. (2013). Hearing loss, hyperacusis, or tinnitus: what is modeled in animal research? Hear. Res. 295, 140–149. doi: 10.1016/j.heares.2012.01.005


PubMed Abstract | CrossRef Full Text | Google Scholar


Eggermont, J. J. (2015). The auditory cortex and tinnitus - a review of animal and human studies. Eur. J. Neurosci. 41, 665–676. doi: 10.1111/ejn.12759


PubMed Abstract | CrossRef Full Text | Google Scholar


Eggermont, J. J., and Kral, A. (2016). Somatic memory and gain increase as preconditions for tinnitus: insights from congenital deafness. Hear. Res. 333, 37–48. doi: 10.1016/j.heares.2015.12.018


PubMed Abstract | CrossRef Full Text | Google Scholar


Eggermont, J. J., and Moore, J. K. (2012). “Morphological and functional development of the auditory nervous system,” in Human Auditory Development, eds L. Werner, R. R. Fay, and A. N. Popper (New York, NY: Springer New York), 61–105.


Google Scholar


Eggermont, J. J., and Roberts, L. E. (2012). The neuroscience of tinnitus: understanding abnormal and normal auditory perception. Front. Syst. Neurosci. 6:53. doi: 10.3389/fnsys.2012.00053


PubMed Abstract | CrossRef Full Text | Google Scholar


Eggermont, J. J., and Tass, P. A. (2015). Maladaptive neural synchrony in tinnitus: origin and restoration. Front. Neurol. 6:29. doi: 10.3389/fneur.2015.00029


PubMed Abstract | CrossRef Full Text | Google Scholar


Engineer, N. D., Riley, J. R., Seale, J. D., Vrana, W. A., Shetake, J. A., Sudanagunta, S. P., et al. (2011). Reversing pathological neural activity using targeted plasticity. Nature 470, 101–104. doi: 10.1038/nature09656


PubMed Abstract | CrossRef Full Text | Google Scholar


Frank, E., Schecklmann, M., Landgrebe, M., Burger, J., Kreuzer, P., Poeppl, T. B., et al. (2011). Treatment of chronic tinnitus with repeated sessions of prefrontal transcranial direct current stimulation: outcomes from an open-label pilot study. J. Neurol. 259, 327–333. doi: 10.1007/s00415-011-6189-4


PubMed Abstract | CrossRef Full Text | Google Scholar


Furman, A. C., Kujawa, S. G., and Liberman, M. C. (2013). Noise-induced cochlear neuropathy is selective for fibers with low spontaneous rates. J. Neurophysiol. 110, 577–586. doi: 10.1152/jn.00164.2013


PubMed Abstract | CrossRef Full Text | Google Scholar


Gersdorff, M., Nouwen, J., Gilain, C., Decat, M., and Betsch, C. (2000). Tinnitus and otosclerosis. Eur. Arch. Otorhinolaryngol. 257, 314–316. doi: 10.1007/s004059900138


CrossRef Full Text | Google Scholar


Geven, L. I., De Kleine, E., Willemsen, A. T., and Van Dijk, P. (2014). Asymmetry in primary auditory cortex activity in tinnitus patients and controls. Neuroscience 256, 117–125. doi: 10.1016/j.neuroscience.2013.10.015


PubMed Abstract | CrossRef Full Text | Google Scholar


Gilles, A., Schlee, W., Rabau, S., Wouters, K., Fransen, E., and Van De Heyning, P. (2016). Decreased speech-in-noise understanding in young adults with tinnitus. Front. Neurosci. 10:288. doi: 10.3389/fnins.2016.00288


PubMed Abstract | CrossRef Full Text | Google Scholar


Guest, H., Munro, K. J., and Plack, C. J. (2017). Tinnitus with a normal audiogram: role of high-frequency sensitivity and reanalysis of brainstem-response measures to avoid audiometric over-matching. Hear. Res. 356,116–117. doi: 10.1016/j.heares.2017.10.002


PubMed Abstract | CrossRef Full Text | Google Scholar


Guitton, M. J. (2012). Tinnitus: pathology of synaptic plasticity at the cellular and system levels. Front. Syst. Neurosci. 6:12. doi: 10.3389/fnsys.2012.00012


CrossRef Full Text | Google Scholar


Guitton, M. J., Caston, J., Ruel, J., Johnson, R. M., Pujol, R., and Puel, J. L. (2003). Salicylate induces tinnitus through activation of cochlear NMDA receptors. J. Neurosci. 23, 3944–3952. doi: 10.1523/JNEUROSCI.23-09-03944.2003


PubMed Abstract | CrossRef Full Text | Google Scholar


Hall, D. A., Lainez, M. J., Newman, C. W., Sanchez, T. G., Egler, M., Tennigkeit, F., et al. (2011). Treatment options for subjective tinnitus: self reports from a sample of general practitioners and ENT physicians within Europe and the USA. BMC Health Serv. Res. 11:302. doi: 10.1186/1472-6963-11-302


PubMed Abstract | CrossRef Full Text | Google Scholar


Hallam, R. S., Jakes, S. C., and Hinchcliffe, R. (1988). Cognitive variables in tinnitus annoyance. Br. J. Clin. Psychol. 27(Pt 3), 213–222. doi: 10.1111/j.2044-8260.1988.tb00778.x


CrossRef Full Text | Google Scholar


Heinz, M. G., and Young, E. D. (2004). Response growth with sound level in auditory-nerve fibers after noise-induced hearing loss. J. Neurophysiol. 91, 784–795. doi: 10.1152/jn.00776.2003


PubMed Abstract | CrossRef Full Text | Google Scholar


Heller, A. J. (2003). Classification and epidemiology of tinnitus. Otolaryngol. Clin. North Am. 36, 239–248. doi: 10.1016/S0030-6665(02)00160-3


CrossRef Full Text | Google Scholar


Heller, M. F., and Bergman, M. (1953). VII Tinnitus aurium in normally hearing persons. Ann. Otol. Rhinol. Laryngol. 62, 73–83. doi: 10.1177/000348945306200107


PubMed Abstract | CrossRef Full Text | Google Scholar


Henry, J. A., Dennis, K. C., and Schechter, M. A. (2005). General review of tinnitus: prevalence, mechanisms, effects, and management. J. Speech Lang. Hear. Res. 48, 1204–1235. doi: 10.1044/1092-4388(2005/084)


PubMed Abstract | CrossRef Full Text | Google Scholar


Henry, J. A., Roberts, L. E., Caspary, D. M., Theodoroff, S. M., and Salvi, R. J. (2014). Underlying mechanisms of tinnitus: review and clinical implications. J. Am. Acad. Audiol. 25, 5–22; quiz 126. doi: 10.3766/jaaa.25.1.2


PubMed Abstract | CrossRef Full Text | Google Scholar


Hiller, W., and Goebel, G. (1992). A psychometric study of complaints in chronic tinnitus. J. Psychosom. Res. 36, 337–348. doi: 10.1016/0022-3999(92)90070-I


CrossRef Full Text | Google Scholar


House, J. W., and Brackmann, D. E. (1981). Tinnitus: surgical treatment. Ciba Found. Symp. 85, 204–216. doi: 10.1002/9780470720677.ch12


CrossRef Full Text | Google Scholar


Hudspeth, A. J. (1985). The cellular basis of hearing: the biophysics of hair cells. Science 230, 745–752. doi: 10.1126/science.2414845


CrossRef Full Text | Google Scholar


Husain, F. T. (2016). Neural networks of tinnitus in humans: elucidating severity and habituation. Hear. Res. 334, 37–48. doi: 10.1016/j.heares.2015.09.010


PubMed Abstract | CrossRef Full Text | Google Scholar


Husain, F. T., Medina, R. E., Davis, C. W., Szymko-Bennett, Y., Simonyan, K., Pajor, N. M., et al. (2011). Neuroanatomical changes due to hearing loss and chronic tinnitus: a combined VBM and DTI study. Brain Res. 1369, 74–88. doi: 10.1016/j.brainres.2010.10.095


PubMed Abstract | CrossRef Full Text | Google Scholar


Husain, F. T., and Schmidt, S. A. (2014). Using resting state functional connectivity to unravel networks of tinnitus. Hear. Res. 307, 153–162. doi: 10.1016/j.heares.2013.07.010


PubMed Abstract | CrossRef Full Text | Google Scholar


Jastreboff, P. J. (1990). Phantom auditory perception (tinnitus): mechanisms of generation and perception. Neurosci. Res. 8, 221–254. doi: 10.1016/0168-0102(90)90031-9


CrossRef Full Text | Google Scholar


Jastreboff, P. J., Brennan, J. F., Coleman, J. K., and Sasaki, C. T. (1988). Phantom auditory sensation in rats: an animal model for tinnitus. Behav. Neurosci. 102, 811–822. doi: 10.1037/0735-7044.102.6.811


CrossRef Full Text | Google Scholar


Jastreboff, P. J., and Hazell, J. W. (1993). A neurophysiological approach to tinnitus: clinical implications. Br. J. Audiol. 27, 7–17. doi: 10.3109/03005369309077884


PubMed Abstract | CrossRef Full Text | Google Scholar


Jastreboff, P. J., and Hazell, J. W. (2004). Tinnitus Retraining Therapy: Implementing the Neurophysiological Model. Cambridge, MA: Cambridge University Press. doi: 10.1017/CBO9780511544989


CrossRef Full Text | Google Scholar


Kalappa, B. I., Brozoski, T. J., Turner, J. G., and Caspary, D. M. (2014). Single unit hyperactivity and bursting in the auditory thalamus of awake rats directly correlates with behavioural evidence of tinnitus. J. Physiol. 592, 5065–5078. doi: 10.1113/jphysiol.2014.278572


PubMed Abstract | CrossRef Full Text | Google Scholar


Kaltenbach, J. A. (2006). Summary of evidence pointing to a role of the dorsal cochlear nucleus in the etiology of tinnitus. Acta Otolaryngol. Suppl. 556, 20–26. doi: 10.1080/03655230600895309


PubMed Abstract | CrossRef Full Text | Google Scholar


Kaltenbach, J. A. (2007). The dorsal cochlear nucleus as a contributor to tinnitus: mechanisms underlying the induction of hyperactivity. Prog. Brain Res. 166, 89–106. doi: 10.1016/S0079-6123(07)66009-9


PubMed Abstract | CrossRef Full Text | Google Scholar


Kaltenbach, J. A. (2011). Tinnitus: models and mechanisms. Hear. Res. 276, 52–60. doi: 10.1016/j.heares.2010.12.003


PubMed Abstract | CrossRef Full Text | Google Scholar


Klit, H., Finnerup, N. B., and Jensen, T. S. (2009). Central post-stroke pain: clinical characteristics, pathophysiology, and management. Lancet Neurol. 8, 857–868. doi: 10.1016/S1474-4422(09)70176-0


CrossRef Full Text | Google Scholar


Knipper, M., Van Dijk, P., Nunes, I., Ruttiger, L., and Zimmermann, U. (2013). Advances in the neurobiology of hearing disorders: recent developments regarding the basis of tinnitus and hyperacusis. Prog. Neurobiol. 111, 17–33. doi: 10.1016/j.pneurobio.2013.08.002


PubMed Abstract | CrossRef Full Text | Google Scholar


Kujawa, S. G., and Liberman, M. C. (2009). Adding insult to injury: cochlear nerve degeneration after “temporary” noise-induced hearing loss. J. Neurosci. 29, 14077–14085. doi: 10.1523/JNEUROSCI.2845-09.2009


PubMed Abstract | CrossRef Full Text | Google Scholar


Kujawa, S. G., and Liberman, M. C. (2015). Synaptopathy in the noise-exposed and aging cochlea: primary neural degeneration in acquired sensorineural hearing loss. Hear. Res. 330, 191–199. doi: 10.1016/j.heares.2015.02.009


PubMed Abstract | CrossRef Full Text | Google Scholar


Landgrebe, M., Langguth, B., Rosengarth, K., Braun, S., Koch, A., Kleinjung, T., et al. (2009). Structural brain changes in tinnitus: grey matter decrease in auditory and non-auditory brain areas. Neuroimage 46, 213–218. doi: 10.1016/j.neuroimage.2009.01.069


PubMed Abstract | CrossRef Full Text | Google Scholar


Langers, D. R., De Kleine, E., and Van Dijk, P. (2012). Tinnitus does not require macroscopic tonotopic map reorganization. Front. Syst. Neurosci. 6:2. doi: 10.3389/fnsys.2012.00002


CrossRef Full Text | Google Scholar


Leaver, A. M., Renier, L., Chevillet, M. A., Morgan, S., Kim, H. J., and Rauschecker, J. P. (2011). Dysregulation of limbic and auditory networks in tinnitus. Neuron 69, 33–43. doi: 10.1016/j.neuron.2010.12.002


PubMed Abstract | CrossRef Full Text | Google Scholar


Leaver, A. M., Seydell-Greenwald, A., and Rauschecker, J. P. (2016a). Auditory-limbic interactions in chronic tinnitus: challenges for neuroimaging research. Hear. Res. 334, 49–57. doi: 10.1016/j.heares.2015.08.005


PubMed Abstract | CrossRef Full Text | Google Scholar


Leaver, A. M., Turesky, T. K., Seydell-Greenwald, A., Morgan, S., Kim, H. J., and Rauschecker, J. P. (2016b). Intrinsic network activity in tinnitus investigated using functional MRI. Hum. Brain Mapp. 37, 2717–2735. doi: 10.1002/hbm.23204


PubMed Abstract | CrossRef Full Text | Google Scholar


Lee, C. C., and Winer, J. A. (2008a). Connections of cat auditory cortex: I. Thalamocortical system. J. Comp. Neurol. 507, 1879–1900. doi: 10.1002/cne.21611


PubMed Abstract | CrossRef Full Text | Google Scholar


Lee, C. C., and Winer, J. A. (2008b). Connections of cat auditory cortex: II. Commissural system. J. Comp. Neurol. 507, 1901–1919. doi: 10.1002/cne.21614


PubMed Abstract | CrossRef Full Text | Google Scholar


Lee, C. C., and Winer, J. A. (2008c). Connections of cat auditory cortex: III. Corticocortical system. J. Comp. Neurol. 507, 1920–1943. doi: 10.1002/cne.21613


PubMed Abstract | CrossRef Full Text | Google Scholar


LePage, E. L. (1989). Functional role of the olivo-cochlear bundle: a motor unit control system in the mammalian cochlea. Hear. Res. 38, 177–198. doi: 10.1016/0378-5955(89)90064-6


PubMed Abstract | CrossRef Full Text | Google Scholar


Levine, R. A. (1999). Somatic (craniocervical) tinnitus and the dorsal cochlear nucleus hypothesis. Am. J. Otolaryngol. 20, 351–362. doi: 10.1016/S0196-0709(99)90074-1


CrossRef Full Text | Google Scholar


Li, S., Choi, V., and Tzounopoulos, T. (2013). Pathogenic plasticity of Kv7.2/3 channel activity is essential for the induction of tinnitus. Proc. Natl. Acad. Sci. U.S.A. 110, 9980–9985. doi: 10.1073/pnas.1302770110


PubMed Abstract | CrossRef Full Text | Google Scholar


Liberman, M. C. (1980). Efferent synapses in the inner hair cell area of the cat cochlea: an electron microscopic study of serial sections. Hear. Res. 3, 189–204. doi: 10.1016/0378-5955(80)90046-5


PubMed Abstract | CrossRef Full Text | Google Scholar


Liberman, M. C., and Dodds, L. W. (1984a). Single-neuron labeling and chronic cochlear pathology. II. Stereocilia damage and alterations of spontaneous discharge rates. Hear. Res. 16, 43–53.


PubMed Abstract | Google Scholar


Liberman, M. C., and Dodds, L. W. (1984b). Single-neuron labeling and chronic cochlear pathology. III. Stereocilia damage and alterations of threshold tuning curves. Hear. Res. 16, 55–74.


PubMed Abstract | Google Scholar


Liberman, M. C., and Dodds, L. W. (1987). Acute ultrastructural changes in acoustic trauma: serial-section reconstruction of stereocilia and cuticular plates. Hear. Res. 26, 45–64. doi: 10.1016/0378-5955(87)90035-9


PubMed Abstract | CrossRef Full Text | Google Scholar


Lobarinas, E., Hayes, S. H., and Allman, B. L. (2013). The gap-startle paradigm for tinnitus screening in animal models: limitations and optimization. Hear. Res. 295, 150–160. doi: 10.1016/j.heares.2012.06.001


PubMed Abstract | CrossRef Full Text | Google Scholar


Lockwood, A. H., Salvi, R. J., Coad, M. L., Towsley, M. L., Wack, D. S., and Murphy, B. W. (1998). The functional neuroanatomy of tinnitus: evidence for limbic system links and neural plasticity. Neurology 50, 114–120. doi: 10.1212/WNL.50.1.114


PubMed Abstract | CrossRef Full Text | Google Scholar


Lowry, L. D., Eisenman, L. M., and Saunders, J. C. (2004). An absence of tinnitus. Otol. Neurotol. 25, 474–478. doi: 10.1097/00129492-200407000-00013


CrossRef Full Text | Google Scholar


Manzoor, N. F., Gao, Y., Licari, F., and Kaltenbach, J. A. (2013). Comparison and contrast of noise-induced hyperactivity in the dorsal cochlear nucleus and inferior colliculus. Hear. Res. 295, 114–123. doi: 10.1016/j.heares.2012.04.003


PubMed Abstract | CrossRef Full Text | Google Scholar


Martines, F., Bentivegna, D., Di Piazza, F., Martines, E., Sciacca, V., and Martinciglio, G. (2010a). Investigation of tinnitus patients in Italy: clinical and audiological characteristics. Int. J. Otolaryngol. 2010:265861. doi: 10.1155/2010/265861


PubMed Abstract | CrossRef Full Text | Google Scholar


Martines, F., Bentivegna, D., Martines, E., Sciacca, V., and Martinciglio, G. (2010b). Assessing audiological, pathophysiological and psychological variables in tinnitus patients with or without hearing loss. Eur. Arch. Otorhinolaryngol. 267, 1685–1693. doi: 10.1007/s00405-010-1302-3


PubMed Abstract | CrossRef Full Text | Google Scholar


Martines, F., Bentivegna, D., Martines, E., Sciacca, V., and Martinciglio, G. (2010c). Characteristics of tinnitus with or without hearing loss: clinical observations in Sicilian tinnitus patients. Auris Nasus Larynx 37, 685–693. doi: 10.1016/j.anl.2010.03.008


PubMed Abstract | CrossRef Full Text | Google Scholar


Maudoux, A., Lefebvre, P., Cabay, J. E., Demertzi, A., Vanhaudenhuyse, A., Laureys, S., et al. (2012). Connectivity graph analysis of the auditory resting state network in tinnitus. Brain Res. 1485, 10–21. doi: 10.1016/j.brainres.2012.05.006


PubMed Abstract | CrossRef Full Text | Google Scholar


McKenna, L., Handscomb, L., Hoare, D. J., and Hall, D. A. (2014). A scientific cognitive-behavioral model of tinnitus: novel conceptualizations of tinnitus distress. Front. Neurol. 5:196. doi: 10.3389/fneur.2014.00196


PubMed Abstract | CrossRef Full Text | Google Scholar


Meikle, M. B., Henry, J. A., Griest, S. E., Stewart, B. J., Abrams, H. B., McArdle, R., et al. (2012). The tinnitus functional index: development of a new clinical measure for chronic, intrusive tinnitus. Ear Hear. 33, 153–176. doi: 10.1097/AUD.0b013e3822f67c0


PubMed Abstract | CrossRef Full Text | Google Scholar


Melcher, J. R., Levine, R. A., Bergevin, C., and Norris, B. (2009). The auditory midbrain of people with tinnitus: abnormal sound-evoked activity revisited. Hear. Res. 257, 63–74. doi: 10.1016/j.heares.2009.08.005


PubMed Abstract | CrossRef Full Text | Google Scholar


Michiels, S., Ganz Sanchez, T., Oron, Y., Gilles, A., Haider, H. F., Erlandsson, S., et al. (2018). Diagnostic criteria for somatosensory tinnitus: a delphi process and face-to-face meeting to establish consensus. Trends Hear. 22:2331216518796403. doi: 10.1177/2331216518796403


PubMed Abstract | CrossRef Full Text | Google Scholar


Midani, A., Carels, I., Marks, M., Wall, M., and Ear Wax Removal Solution Study Team (2006). Safety and efficacy of Sofenz ceruminolytic solution. Ear Nose Throat J. 85, 87–88, 90–92.


PubMed Abstract | Google Scholar


Middleton, J. W., Kiritani, T., Pedersen, C., Turner, J. G., Shepherd, G. M., and Tzounopoulos, T. (2011). Mice with behavioral evidence of tinnitus exhibit dorsal cochlear nucleus hyperactivity because of decreased GABAergic inhibition. Proc. Natl. Acad. Sci. U.S.A. 108, 7601–7606. doi: 10.1073/pnas.1100223108


PubMed Abstract | CrossRef Full Text | Google Scholar


Mirz, F., Gjedde, A., Sodkilde-Jrgensen, H., and Pedersen, C. B. (2000). Functional brain imaging of tinnitus-like perception induced by aversive auditory stimuli. Neuroreport 11, 633–637. doi: 10.1097/00001756-200002280-00039


PubMed Abstract | CrossRef Full Text | Google Scholar


Mittal, R., Aranke, M., Debs, L. H., Nguyen, D., Patel, A. P., Grati, M., et al. (2017). Indispensable role of ion channels and transporters in the auditory system. J. Cell. Physiol. 232, 743–758. doi: 10.1002/jcp.25631


PubMed Abstract | CrossRef Full Text | Google Scholar


Moazami-Goudarzi, M., Michels, L., Weisz, N., and Jeanmonod, D. (2010). Temporo-insular enhancement of EEG low and high frequencies in patients with chronic tinnitus. QEEG study of chronic tinnitus patients. BMC Neurosci. 11:40. doi: 10.1186/1471-2202-11-40


PubMed Abstract | CrossRef Full Text | Google Scholar


Moser, T., Neef, A., and Khimich, D. (2006). Mechanisms underlying the temporal precision of sound coding at the inner hair cell ribbon synapse. J. Physiol. 576, 55–62. doi: 10.1113/jphysiol.2006.114835


CrossRef Full Text | Google Scholar


Muhlau, M., Rauschecker, J. P., Oestreicher, E., Gaser, C., Rottinger, M., Wohlschlager, A. M., et al. (2006). Structural brain changes in tinnitus. Cereb. Cortex 16, 1283–1288. doi: 10.1093/cercor/bhj070


PubMed Abstract | CrossRef Full Text | Google Scholar


Muhlnickel, W., Elbert, T., Taub, E., and Flor, H. (1998). Reorganization of auditory cortex in tinnitus. Proc. Natl. Acad. Sci. U.S.A. 95, 10340–10343. doi: 10.1073/pnas.95.17.10340


CrossRef Full Text | Google Scholar


Mulders, W. H., and Robertson, D. (2009). Hyperactivity in the auditory midbrain after acoustic trauma: dependence on cochlear activity. Neuroscience 164,733–746. doi: 10.1016/j.neuroscience.2009.08.036


PubMed Abstract | CrossRef Full Text | Google Scholar


Muller, N., Lorenz, I., Langguth, B., and Weisz, N. (2013). rTMS induced tinnitus relief is related to an increase in auditory cortical alpha activity. PLoS One 8:e55557. doi: 10.1371/journal.pone.0055557


PubMed Abstract | CrossRef Full Text | Google Scholar


Newman, C. W., Jacobson, G. P., and Spitzer, J. B. (1996). Development of the tinnitus handicap inventory. Arch. Otolaryngol. Head Neck Surg. 122, 143–148. doi: 10.1001/archotol.1996.01890140029007


CrossRef Full Text | Google Scholar


Nicolas-Puel, C., Akbaraly, T., Lloyd, R., Berr, C., Uziel, A., Rebillard, G., et al. (2006). Characteristics of tinnitus in a population of 555 patients: specificities of tinnitus induced by noise trauma. Int. Tinnitus J. 12, 64–70.


PubMed Abstract | Google Scholar


Norena, A., Micheyl, C., Chery-Croze, S., and Collet, L. (2002). Psychoacoustic characterization of the tinnitus spectrum: implications for the underlying mechanisms of tinnitus. Audiol. Neurootol. 7, 358–369. doi: 10.1159/000066156


PubMed Abstract | CrossRef Full Text | Google Scholar


Norena, A. J. (2011). An integrative model of tinnitus based on a central gain controlling neural sensitivity. Neurosci. Biobehav. Rev. 35, 1089–1109. doi: 10.1016/j.neubiorev.2010.11.003


PubMed Abstract | CrossRef Full Text | Google Scholar


Norena, A. J. (2015). Revisiting the cochlear and central mechanisms of tinnitus and therapeutic approaches. Audiol. Neurootol. 20(Suppl. 1), 53–59. doi: 10.1159/000380749


PubMed Abstract | CrossRef Full Text | Google Scholar


Norena, A. J., and Farley, B. J. (2013). Tinnitus-related neural activity: theories of generation, propagation, and centralization. Hear. Res. 295, 161–171. doi: 10.1016/j.heares.2012.09.010


PubMed Abstract | CrossRef Full Text | Google Scholar


Nuttal, A. L., Meikle, M. B., and Trune, D. R. (2004). “Peripheral process involved in tinnitus,” in Tinnitus: Theory and Management, ed. J. Snow (Ontario: BC Decker), 52–68.


Google Scholar


Pace, E., Luo, H., Bobian, M., Panekkad, A., Zhang, X., Zhang, H., et al. (2016). A conditioned behavioral paradigm for assessing onset and lasting tinnitus in rats. PLoS One 11:e0166346. doi: 10.1371/journal.pone.0166346


PubMed Abstract | CrossRef Full Text | Google Scholar


Parra, L. C., and Pearlmutter, B. A. (2007). Illusory percepts from auditory adaptation. J. Acoust. Soc. Am. 121, 1632–1641. doi: 10.1121/1.2431346


CrossRef Full Text | Google Scholar


Parsons, M. P., and Raymond, L. A. (2014). Extrasynaptic NMDA receptor involvement in central nervous system disorders. Neuron 82, 279–293. doi: 10.1016/j.neuron.2014.03.030


PubMed Abstract | CrossRef Full Text | Google Scholar


Patuzzi, R. (2002). Non-linear aspects of outer hair cell transduction and the temporary threshold shifts after acoustic trauma. Audiol. Neurootol. 7, 17–20. doi: 10.1159/000046857


PubMed Abstract | CrossRef Full Text | Google Scholar


Pienkowski, M., and Harrison, R. V. (2005). Tone frequency maps and receptive fields in the developing chinchilla auditory cortex. J. Neurophysiol. 93, 454–466. doi: 10.1152/jn.00569.2004


PubMed Abstract | CrossRef Full Text | Google Scholar


Pierzycki, R. H., Mcnamara, A. J., Hoare, D. J., and Hall, D. A. (2016). 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. 331, 101–108. doi: 10.1016/j.heares.2015.11.003


PubMed Abstract | CrossRef Full Text | Google Scholar


Puel, J. L., and Guitton, M. J. (2007). Salicylate-induced tinnitus: molecular mechanisms and modulation by anxiety. Prog. Brain Res. 166, 141–146. doi: 10.1016/S0079-6123(07)66012-9


PubMed Abstract | CrossRef Full Text | Google Scholar


Pujol, R., and Puel, J. L. (1999). Excitotoxicity, synaptic repair, and functional recovery in the mammalian cochlea: a review of recent findings. Ann. N. Y. Acad. Sci. 884, 249–254. doi: 10.1111/j.1749-6632.1999.tb08646.x


PubMed Abstract | CrossRef Full Text | Google Scholar


Pujol, R., Puel, J. L., and Eybalin, M. (1992). Implication of non-NMDA and NMDA receptors in cochlear ischemia. Neuroreport 3, 299–302. doi: 10.1097/00001756-199204000-00002


PubMed Abstract | CrossRef Full Text | Google Scholar


Pujol, R., Puel, J. L., Gervais D’aldin, C., and Eybalin, M. (1993). Pathophysiology of the glutamatergic synapses in the cochlea. Acta Otolaryngol. 113, 330–334. doi: 10.3109/00016489309135819


CrossRef Full Text | Google Scholar


Rabau, S., Cox, T., Punte, A. K., Waelkens, B., Gilles, A., Wouters, K., et al. (2015). Changes over time of psychoacoustic outcome measurements are not a substitute for subjective outcome measurements in acute tinnitus. Eur. Arch. Otorhinolaryngol. 272, 573–581. doi: 10.1007/s00405-013-2876-3


PubMed Abstract | CrossRef Full Text | Google Scholar


Rauschecker, J. P., Leaver, A. M., and Muhlau, M. (2010). Tuning out the noise: limbic-auditory interactions in tinnitus. Neuron 66, 819–826. doi: 10.1016/j.neuron.2010.04.032


PubMed Abstract | CrossRef Full Text | Google Scholar


Rauschecker, J. P., May, E. S., Maudoux, A., and Ploner, M. (2015). Frontostriatal gating of tinnitus and chronic pain. Trends Cogn. Sci. 19, 567–578. doi: 10.1016/j.tics.2015.08.002


PubMed Abstract | CrossRef Full Text | Google Scholar


Riga, M., Katotomichelakis, M., and Danielides, V. (2015). The potential role of the medial olivocochlear bundle in the generation of tinnitus: controversies and weaknesses in the existing clinical studies. Otol. Neurotol. 36, 201–208. doi: 10.1097/MAO.0000000000000384


PubMed Abstract | CrossRef Full Text | Google Scholar


Rojas, R., Palacios, E., D’antonio, M., and Correa, G. (2003). Aberrant internal carotid artery as a cause of pulsatile tinnitus and an intratympanic mass. Ear Nose Throat J. 82, 173–174.


PubMed Abstract | Google Scholar


Ruttiger, L., Singer, W., Panford-Walsh, R., Matsumoto, M., Lee, S. C., Zuccotti, A., et al. (2013). The reduced cochlear output and the failure to adapt the central auditory response causes tinnitus in noise exposed rats. PLoS One 8:e57247. doi: 10.1371/journal.pone.0057247


PubMed Abstract | CrossRef Full Text | Google Scholar


Sahley, T. L., Hammonds, M. D., and Musiek, F. E. (2013). Endogenous dynorphins, glutamate and N-methyl-d-aspartate (NDMA) receptors may participate in a stress-mediated Type-I auditory neural exacerbation of tinnitus. Brain Res. 1499, 80–108. doi: 10.1016/j.brainres.2013.01.006


PubMed Abstract | CrossRef Full Text | Google Scholar


Sanchez, J. T., Ghelani, S., and Otto-Meyer, S. (2015). From development to disease: diverse functions of NMDA-type glutamate receptors in the lower auditory pathway. Neuroscience 285, 248–259. doi: 10.1016/j.neuroscience.2014.11.027


PubMed Abstract | CrossRef Full Text | Google Scholar


Satar, B., Kapkin, O., and Ozkaptan, Y. (2003). Evaluation of cochlear function in patients with normal hearing and tinnitus: a distortion product otoacoustic emission study. Kulak Burun Bogaz Ihtis Derg 10, 177–182.


Google Scholar


Schaette, R. (2013). Tinnitus in men, mice (as well as other rodents), and machines. Hear. Res. 311, 63–71. doi: 10.1016/j.heares.2013.12.004


PubMed Abstract | CrossRef Full Text | Google Scholar


Schaette, R., and McAlpine, D. (2011). Tinnitus with a normal audiogram: physiological evidence for hidden hearing loss and computational model. J. Neurosci. 31, 13452–13457. doi: 10.1523/JNEUROSCI.2156-11.2011


PubMed Abstract | CrossRef Full Text | Google Scholar


Schaette, R., Turtle, C., and Munro, K. J. (2012). Reversible induction of phantom auditory sensations through simulated unilateral hearing loss. PLoS One 7:e35238. doi: 10.1371/journal.pone.0035238


PubMed Abstract | CrossRef Full Text | Google Scholar


Schecklmann, M., Lehner, A., Schlee, W., Vielsmeier, V., Landgrebe, M., and Langguth, B. (2015). Validation of screening questions for hyperacusis in chronic tinnitus. Biomed. Res. Int. 2015:191479. doi: 10.1155/2015/191479


PubMed Abstract | CrossRef Full Text | Google Scholar


Schlee, W., Mueller, N., Hartmann, T., Keil, J., Lorenz, I., and Weisz, N. (2009). Mapping cortical hubs in tinnitus. BMC Biol. 7:80. doi: 10.1186/1741-7007-7-80


PubMed Abstract | CrossRef Full Text | Google Scholar


Schlee, W., Schecklmann, M., Lehner, A., Kreuzer, P. M., Vielsmeier, V., Poeppl, T. B., et al. (2014). Reduced variability of auditory alpha activity in chronic tinnitus. Neural Plast. 2014:436146. doi: 10.1155/2014/436146


PubMed Abstract | CrossRef Full Text | Google Scholar


Schlee, W., Weisz, N., Bertrand, O., Hartmann, T., and Elbert, T. (2008). Using auditory steady state responses to outline the functional connectivity in the tinnitus brain. PLoS One 3:e3720. doi: 10.1371/journal.pone.0003720


PubMed Abstract | CrossRef Full Text | Google Scholar


Sedley, W., Friston, K. J., Gander, P. E., Kumar, S., and Griffiths, T. D. (2016). An integrative tinnitus model based on sensory precision. Trends Neurosci. 39, 799–812. doi: 10.1016/j.tins.2016.10.004


PubMed Abstract | CrossRef Full Text | Google Scholar


Sedley, W., Gander, P. E., Kumar, S., Oya, H., Kovach, C. K., Nourski, K. V., et al. (2015). Intracranial mapping of a cortical tinnitus system using residual inhibition. Curr. Biol. 25, 1208–1214. doi: 10.1016/j.cub.2015.02.075


PubMed Abstract | CrossRef Full Text | Google Scholar


Sedley, W., Teki, S., Kumar, S., Barnes, G. R., Bamiou, D.-E., and Griffiths, T. D. (2012). Single-subject oscillatory gamma responses in tinnitus. Brain 135, 3089–3100. doi: 10.1093/brain/aws220


PubMed Abstract | CrossRef Full Text | Google Scholar


Sereda, M., Hall, D. A., Bosnyak, D. J., Edmondson-Jones, M., Roberts, L. E., Adjamian, P., et al. (2011). Re-examining the relationship between audiometric profile and tinnitus pitch. Int. J. Audiol. 50, 303–312. doi: 10.3109/14992027.2010.551221


PubMed Abstract | CrossRef Full Text | Google Scholar


Sergeyenko, Y., Lall, K., Liberman, M. C., and Kujawa, S. G. (2013). Age-related cochlear synaptopathy: an early-onset contributor to auditory functional decline. J. Neurosci. 33, 13686–13694. doi: 10.1523/JNEUROSCI.1783-13.2013


PubMed Abstract | CrossRef Full Text | Google Scholar


Sewell, W. F. (1984). The effects of furosemide on the endocochlear potential and auditory-nerve fiber tuning curves in cats. Hear. Res. 14, 305–314. doi: 10.1016/0378-5955(84)90057-1


PubMed Abstract | CrossRef Full Text | Google Scholar


Shinonaga, Y., Takada, M., and Mizuno, N. (1994). Direct projections from the non-laminated divisions of the medial geniculate nucleus to the temporal polar cortex and amygdala in the cat. J. Comp. Neurol. 340, 405–426. doi: 10.1002/cne.903400310


PubMed Abstract | CrossRef Full Text | Google Scholar


Shore, S., Zhou, J., and Koehler, S. (2007). Neural mechanisms underlying somatic tinnitus. Prog. Brain Res. 166, 107–123. doi: 10.1016/S0079-6123(07)66010-5


CrossRef Full Text | Google Scholar


Shulman, A. (1995). A final common pathway for tinnitus - the medial temporal lobe system. Int. Tinnitus J. 1, 115–126.


Google Scholar


Sindhusake, D., Golding, M., Wigney, D., Newall, P., Jakobsen, K., and Mitchell, P. (2004). Factors predicting severity of tinnitus: a population-based assessment. J. Am. Acad. Audiol. 15, 269–280. doi: 10.3766/jaaa.15.4.2


CrossRef Full Text | Google Scholar


Smith, P. F., and Zheng, Y. W. (2016). Cannabinoids, cannabinoid receptors and tinnitus. Hear. Res. 332, 210–216. doi: 10.1016/j.heares.2015.09.014


PubMed Abstract | CrossRef Full Text | Google Scholar


Sobrinho, P. G., Oliveira, C. A., and Venosa, A. R. (2004). Long-term follow-up of tinnitus in patients with otosclerosis after stapes surgery. Int. Tinnitus J. 10, 197–201.


Google Scholar


Sweetow, R. W. (1986). Cognitive aspects of tinnitus patient management. Ear Hear. 7, 390–396. doi: 10.1097/00003446-198612000-00008


CrossRef Full Text | Google Scholar


Tucker, D. A., Phillips, S. L., Ruth, R. A., Clayton, W. A., Royster, E., and Todd, A. D. (2005). The effect of silence on tinnitus perception. Otolaryngol. Head Neck Surg. 132, 20–24. doi: 10.1016/j.otohns.2005.08.016


PubMed Abstract | CrossRef Full Text | Google Scholar


Tunkel, D. E., Bauer, C. A., Sun, G. H., Rosenfeld, R. M., Chandrasekhar, S. S., Cunningham, E. R., et al. (2014). Clinical practice guideline: tinnitus. Otolaryngol. Head Neck Surg. 151, S1–S40. doi: 10.1177/0194599814545325


PubMed Abstract | CrossRef Full Text | Google Scholar


Turner, J. G., and Larsen, D. (2016). Effects of noise exposure on development of tinnitus and hyperacusis: prevalence rates 12 months after exposure in middle-aged rats. Hear. Res. 334, 30–36. doi: 10.1016/j.heares.2015.11.004


PubMed Abstract | CrossRef Full Text | Google Scholar


Tyler, R., Ji, H., Perreau, A., Witt, S., Noble, W., and Coelho, C. (2014). Development and validation of the tinnitus primary function questionnaire. Am. J. Audiol. 23, 260–272. doi: 10.1044/2014_AJA-13-0014


PubMed Abstract | CrossRef Full Text | Google Scholar


Vanneste, S., and De Ridder, D. (2011). Bifrontal transcranial direct current stimulation modulates tinnitus intensity and tinnitus-distress-related brain activity. Eur. J. Neurosci. 34, 605–614. doi: 10.1111/j.1460-9568.2011.07778.x


PubMed Abstract | CrossRef Full Text | Google Scholar


Vogler, D. P., Robertson, D., and Mulders, W. H. (2011). Hyperactivity in the ventral cochlear nucleus after cochlear trauma. J. Neurosci. 31, 6639–6645. doi: 10.1523/JNEUROSCI.6538-10.2011


CrossRef Full Text | Google Scholar


Wan, G., and Corfas, G. (2017). Transient auditory nerve demyelination as a new mechanism for hidden hearing loss. Nat. Commun. 8:14487. doi: 10.1038/ncomms14487


PubMed Abstract | CrossRef Full Text | Google Scholar


Weisz, N., Dohrmann, K., and Elbert, T. (2007). The relevance of spontaneous activity for the coding of the tinnitus sensation. Prog. Brain Res. 166, 61–70. doi: 10.1016/S0079-6123(07)66006-3


PubMed Abstract | CrossRef Full Text | Google Scholar


Weisz, N., Moratti, S., Meinzer, M., Dohrmann, K., and Elbert, T. (2005). Tinnitus perception and distress is related to abnormal spontaneous brain activity as measured by magnetoencephalography. PLoS Med. 2:e153. doi: 10.1371/journal.pmed.0020153


PubMed Abstract | CrossRef Full Text | Google Scholar


Winer, J. A., Sally, S. L., Larue, D. T., and Kelly, J. B. (1999). Origins of medial geniculate body projections to physiologically defined zones of rat primary auditory cortex. Hear. Res. 130, 42–61. doi: 10.1016/S0378-5955(98)00217-2


PubMed Abstract | CrossRef Full Text | Google Scholar


Wu, C., Stefanescu, R. A., Martel, D. T., and Shore, S. E. (2015). Tinnitus: maladaptive auditory-somatosensory plasticity. Hear. Res. 334, 20–29. doi: 10.1016/j.heares.2015.06.005


PubMed Abstract | CrossRef Full Text | Google Scholar


Yang, S., Weiner, B. D., Zhang, L. S., Cho, S. J., and Bao, S. (2011). Homeostatic plasticity drives tinnitus perception in an animal model. Proc. Natl. Acad. Sci. U.S.A. 108, 14974–14979. doi: 10.1073/pnas.1107998108


PubMed Abstract | CrossRef Full Text | Google Scholar


Zenner, H. P. (1998). A systematic classification of tinnitus generator mechanisms. Int. Tinnitus J. 4, 109–113.


PubMed Abstract | Google Scholar


Zhao, Y., Song, Q., Li, X. Y., and Li, C. Y. (2016). Neural hyperactivity of the central auditory system in response to peripheral damage. Neural Plast. 2016:2162105. doi: 10.1155/2016/2162105


PubMed Abstract | CrossRef Full Text | Google Scholar


Keywords: idiopathic, auditory system, pathophysiology, central tinnitus, peripheral tinnitus, causes, maintenance


Citation: Haider HF, Bojić T, Ribeiro SF, Paço J, Hall DA and Szczepek AJ (2018) Pathophysiology of Subjective Tinnitus: Triggers and Maintenance. Front. Neurosci. 12:866. doi: 10.3389/fnins.2018.00866


Received: 08 July 2018; Accepted: 06 November 2018;

Published: 27 November 2018.


Edited by:


Victoria M. Bajo Lorenzana, University of Oxford, United Kingdom

Reviewed by:


Daniel Llano, University of Illinois at Urbana–Champaign, United States

Carine Signoret, Linköping University, Sweden

Copyright © 2018 Haider, Bojić, Ribeiro, Paço, Hall and Szczepek. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.


*Correspondence: Haúla Faruk Haider, haula.f.haider@jmellosaude.pt; hfhaider@gmail.com orcid.org/0000-0002-3860-5895 Agnieszka J. Szczepek, Agnes.Szczepek@charite.de orcid.org/0000-0002-9292-6606


Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.

It's important to consult with a healthcare professional if you are experiencing any symptoms. They can help to determine the underlying cause and recommend the appropriate treatment options.