Next Article in Journal
Adipose Tissue Inflammation and Pulmonary Dysfunction in Obesity
Next Article in Special Issue
Relationships between Inflammation and Age-Related Neurocognitive Changes
Previous Article in Journal
Impact of Double Covalent Binding of BV in NIR FPs on Their Spectral and Physicochemical Properties
Previous Article in Special Issue
Effects of Combined Gentamicin and Furosemide Treatment on Cochlear Macrophages
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Age-Related Hearing Loss: The Link between Inflammaging, Immunosenescence, and Gut Dysbiosis

by
Dagmara Kociszewska
and
Srdjan Vlajkovic
*
Department of Physiology and The Eisdell Moore Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, Auckland 1142, New Zealand
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(13), 7348; https://doi.org/10.3390/ijms23137348
Submission received: 27 May 2022 / Revised: 30 June 2022 / Accepted: 30 June 2022 / Published: 1 July 2022
(This article belongs to the Special Issue Effects of Chronic Inflammation on Brain Ageing and Sensory Decline)

Abstract

:
This article provides a theoretical overview of the association between age-related hearing loss (ARHL), immune system ageing (immunosenescence), and chronic inflammation. ARHL, or presbyacusis, is the most common sensory disability that significantly reduces the quality of life and has a high economic impact. This disorder is linked to genetic risk factors but is also influenced by a lifelong cumulative effect of environmental stressors, such as noise, otological diseases, or ototoxic drugs. Age-related hearing loss and other age-related disorders share common mechanisms which often converge on low-grade chronic inflammation known as “inflammaging”. Various stimuli can sustain inflammaging, including pathogens, cell debris, nutrients, and gut microbiota. As a result of ageing, the immune system can become defective, leading to the accumulation of unresolved inflammatory processes in the body. Gut microbiota plays a central role in inflammaging because it can release inflammatory mediators and crosstalk with other organ systems. A proinflammatory gut environment associated with ageing could result in a leaky gut and the translocation of bacterial metabolites and inflammatory mediators to distant organs via the systemic circulation. Here, we postulate that inflammaging, as a result of immunosenescence and gut dysbiosis, accelerates age-related cochlear degeneration, contributing to the development of ARHL. Age-dependent gut dysbiosis was included as a hypothetical link that should receive more attention in future studies.

1. Introduction

Age-related hearing loss (ARHL), or presbyacusis, is a type of sensorineural hearing loss that primarily affects the elderly [1]. However, the age of onset, rate of decline, and severity of hearing loss vary widely.
ARHL is the most common sensory disorder, with a high economic impact [2,3]. The World Health Organization (WHO) estimates that by 2050, 2.5 billion people, predominantly over 60, will be living with some degree of hearing loss [4,5]. Despite the high prevalence of this sensory disorder, there is a paucity of both preventative and treatment strategies other than prosthetic devices (hearing aids and cochlear implants).
Presbyacusis typically presents as bilateral, progressive, and irreversible [6,7]. The increasing prevalence of presbyacusis may be attributable to environmental factors, notably noise exposure and the rise in metabolic diseases [8,9,10].
This sensory disorder can be characterised by reduced hearing sensitivity and speech understanding in background noise, slowed central processing of acoustic information, and impaired localisation of sound sources [7]. Hearing loss affects high frequencies initially and eventually spreads to lower frequencies involved in speech understanding [7]. Untreated hearing impairment contributes to social isolation, loss of self-esteem, depression, and cognitive decline [11,12,13]. Even mild levels of hearing loss increase the long-term risk of cognitive decline and dementia [14].
ARHL has a complex pathophysiology linked to genetic risk factors that determine the rate and extent of cochlear degeneration. However, the severity of the hearing loss is also influenced by previous otological diseases, chronic illnesses, cumulative noise exposure, use of ototoxic drugs, and lifestyle [15]. Moreover, this condition has been associated with numerous comorbidities, including dementia, frailty, Alzheimer’s disease, and type II diabetes [16,17,18,19,20]. A common trait of these disorders is chronic inflammation in target organs [21]. More recently, changes in gut microbiota have been linked to systemic inflammation affecting multiple organ systems, including the brain and the inner ear [22,23,24]. This review focuses on cochlear inflammation, immunosenescence, and gut dysbiosis as putative underlying factors of ARHL.

2. Methodology

A literature search of related publications from 1995 to 2021 was conducted using PubMed, Google Scholar, and Embase medical databases. In addition, reference lists from the relevant papers were used. Following Boolean search logic, the main keywords included were: (age-related hearing loss OR presbyacusis OR cochlea) and (diet OR obesity OR diabetes OR diet-induced obesity OR inflammation) and (microbiota OR gut dysbiosis OR reactive oxygen species OR lipopolysaccharide OR high-fat diet OR intestinal hyperpermeability OR inflammatory bowel disorder OR inflammaging OR immunosenescence OR neurodegeneration or inflammation). The search results were consequently examined according to their relevance to this review. Only English language publications were included.

3. Pathophysiology of Age-Related Hearing Loss

Age-related hearing loss has mixed aetiology [7] and is likely a cumulative result of genetic and epigenetic factors [8,25,26,27] and environmental stressors [28]. Otological diseases, chronic exposure to noise, smoking, or exposure to ototoxic drugs can contribute to the development of ARHL [29,30]. Other factors include diet, gender, comorbidities, and lifestyle [20,31].
Reduced vascularisation in the cochlea, cumulative oxidative stress (OS), low-grade cochlear inflammation, impaired mitochondrial quality control, and mitochondrial DNA damage play a critical role in developing ARHL [8,32]. Age-related degenerative changes in the cochlea can lead to a loss of sensory hair cells and primary auditory neurons, damage to cochlear lateral wall tissues (stria vascularis and spiral ligament), and reduced vascularisation [29,33]. Post-mortem histological studies in the human cochlea have revealed sensory hair cell loss in the organ of Corti (OoC), degenerative changes in the auditory nerve, atrophy of the stria vascularis (SV), and loss of fibrocytes in the spiral ligament (SL) [20,34,35,36]. More recent studies have demonstrated a loss of auditory nerve afferent fibres in the cochlea and synapses between the inner hair cells and type I afferent fibres [37,38]. The ageing process also negatively affects the central auditory pathways [20]. Based on predominant histopathological findings and differences in pure-tone audiometric testing, Schuknetcht et al. proposed classifying ARHL into sensory, neural, strial, and cochlear conductive types [34,35,39]. Many people with ARHL likely have a mixed pathology, but in some cases, the cause of ARHL cannot be determined by histological evaluation of cochlear tissues [40].
At present, ARHL has not been fully reproduced in animal models. However, these models are often used to delineate human pathophysiology, as clinical studies are challenging due to cochlear localisation deep in the temporal bone, precluding histological and high-resolution imaging studies [20]. In animal studies, vascular changes include reduced capillary network and narrowing of the vascular lumen in the SV [41,42,43]. The secretory epithelium of the SV is responsible for maintaining the high potassium (K+) content of the endolymph and generation of the endocochlear potential (EP), which drives sensory transduction in the cochlea [44,45,46]. The SV typically deteriorates in the mid-cochlear to apical regions and is associated with reduced expression levels and activity of sodium-potassium pumps (Na-K-2Cl cotransporter NKCC1 and Na+, K+-ATPase), which leads to reduced EP [47,48,49,50,51,52,53,54]. Reduced activity of sodium-potassium pumps and decreased EP have been demonstrated in ageing gerbils raised in quiet and ageing mice [44,55].
The mouse is a robust and reliable mammalian model for ageing research, and the use of inbred mouse strains was instrumental in investigating the genetics of ARHL. For example, the commonly used C57BL/6 mouse strain develops progressive high-frequency hearing loss caused by a mutation of the cadherin 23 (Cdh23) gene, which encodes a component of the stereocilial tip-link required for gating of the mechanoelectrical transducer (MET) channel in sensory hair cells [56].
Elevated auditory thresholds in ARHL typically result from degeneration and loss of outer hair cells within the OoC. The loss of hair cells progresses from the basal turn of the cochlea (high-frequency region) to the apical turn (low-frequency region) [32]. Degenerative changes also affect synaptic networks between the inner hair cells and afferent auditory nerve fibres, which leads to reduced speech understanding in background noise [57,58,59].

4. Immunosenescence and Inflammaging

Immunosenescence is an age-dependent development of immune dysfunction that involves lymphoid organ remodelling, leading to reduced capacity to control inflammatory cytokines during and after the immune response. Immunosenescence can lead to chronic inflammation in ageing tissues, frequent infections, autoimmune diseases, and cancer due to impaired immune surveillance [21,60,61,62,63,64].
Inflammaging is a relatively new concept described as age-related, low-grade systemic inflammation that may not directly link to microbial infection [32,65]. Various stimuli, including cell debris, nutrients, and gut microbiota, can sustain inflammaging [66]. This sterile or pathogen-driven inflammation increases morbidity and mortality in the elderly [21,67,68]. As a result of ageing, the immune system becomes defective (immunosenescence), leading to the accumulation of unresolved inflammatory processes impacting otherwise healthy organ systems [69]. As a result, inflammaging can contribute to a spectrum of disorders such as Parkinson’s and Alzheimer’s disease, type II diabetes, and cardiovascular disease [70,71,72,73,74,75,76,77,78,79,80,81,82]. However, inflammaging is not a physiological or expected outcome of ageing; instead, a tell-tale of accelerated ageing [66].
Sensorineural hearing loss has also been linked with chronic inflammation [83,84,85]. Despite the historical belief that the cochlea is an immune-privileged organ [86], more recent studies have shown that the cochlea is vulnerable to systemic inflammation [22,87]. Cochlear microcirculation is controlled by tight junctions connecting vascular endothelial cells, forming the blood-labyrinth barrier (BLB) in the lateral wall [88]. The BLB plays a role in preventing pathogen infiltration, maintaining ion homeostasis, and transporting nutrients to the cochlea [89]. Pericytes and perivascular resident macrophage-like melanocytes (PVM/M) represent the second line of support for the BLB. Local inflammation activates PVM/M in the cochlea and thus increases the permeability of the BLB [90]. Furthermore, PVM/M can release proinflammatory cytokines through the tight-junction barrier [90] and increase the permeability of the BLB to the bacterial metabolite lipopolysaccharide (LPS) [88,91]. Similarly, acoustic trauma [88,92,93] and hypoxia [94] can also increase the permeability of the BLB, resulting in cochlear inflammation that predominantly affects the lateral wall tissues (SV and SL) [21,84]. It was shown that vascular cell senescence is a key factor in the breakdown of the blood-brain barrier (BBB) [95], which is physiologically and structurally equivalent to the BLB of the inner ear [22]. This suggests that vascular cell senescence may also affect the integrity and permeability of the BLB.
Inflammation has been identified in multiple preclinical and population health studies as a pathophysiological mechanism contributing to ARHL [85]. For example, in the “Hertfordshire Ageing Study”, Verschuur et al. described a progressive increase in the expression of markers associated with systemic inflammation (interleukin-6, C-reactive protein, white blood cell, and neutrophil counts) in subjects with ARHL, which correlated with the elevation in hearing thresholds [63,96]. That study concluded that low-grade inflammation is at the foundation of ARHL. In the English Longitudinal Study of Ageing, Lassale et al. also demonstrated an association between white blood cell counts and age-related hearing impairment [97]. Other studies revealed changes in the number and morphology of macrophages in the ageing cochlea [98,99]. Activated macrophages were present in the lateral wall and auditory nerve and were more abundant in the cochlear basal turn of the older donors [99]. Based on these studies, an ongoing ASPREE-HEARING study was designed to investigate the benefits of low dosages of the anti-inflammatory agent aspirin on the progression of ARHL [100]. The rationale for this study is that aspirin is an inflammation resolution mediator [101], as it decreases the levels of proinflammatory mediators, including TNF-α, IL-6, and thromboxane B2 (TXB2) [102,103,104]. The study is still recruiting participants at the time of this review.
Preclinical studies have shown that in ageing C57BL/6J mice, the resident macrophages in the basilar membrane of the OoC change morphologically in response to sensory cell degeneration, indicating their activation [105]. This finding is consistent with the up-regulation of genes linked with immune and inflammatory responses in older murine cochleae [106]. Using next-generation sequencing, Su and collaborators [106] revealed multiple immune and inflammatory transcriptomic changes during cochlear ageing. The TNF signalling pathway, toll-like receptor signalling pathway, Jak-STAT signalling pathway, and NF-kB signalling pathway featured prominently among up-regulated genes in aged mice [106].
A senescence-associated secretory phenotype (SASP) is one of the possible factors contributing to inflammaging and associated changes in the central nervous system (CNS). It has been established that cells change their phenotype to senescence as a preventative measure for malignancies; however, these cells accumulate within tissues as the body ages [107]. Even though these cells are growth-arrested, they are still metabolically active and change protein expression primarily due to DNA damage [108]. SASP promotes local inflammation via the secretion of cytokines, chemokines, reactive oxygen and nitrogen species, and growth factors [107]. Proliferative cells of the CNS, such as endothelial and glial cells, can adopt SASP, leading to low-grade chronic inflammation in the ageing brain [109]. It was proposed that the permeability of the BBB might be affected by the build-up of SASP cells [95,110,111].
Despite the similarities between the BBB and the BLB, this aspect of senescence has yet to be established for the BLB and ARHL.

5. Chronic Inflammation

Chronic inflammation is a complex process that includes various pathophysiological mechanisms, such as OS, ionic disbalance, mitochondrial damage, and epigenetic changes, to name a few [112,113,114,115,116,117]. Immunosenescence and gut dysbiosis can also result in chronic inflammation. The natural immune response (innate and adaptive) relies on the delicate balance between proinflammatory and anti-inflammatory mediators to protect us from pathogens. However, the inflammatory process cannot be resolved when this balance is disrupted, leading to unresolved chronic inflammation [70,118]. As the body ages, cellular debris accumulates because of inefficient waste management, thus perpetuating innate immune responses that contribute to chronic inflammation [119].
Interestingly, most age-associated inflammatory disorders share similar mechanisms, mediators, and intracellular signalling pathways. Widely used biomarkers of inflammation are interleukin 6 (IL-6) [74,83,120,121,122], tumour necrosis factor-α (TNF-α) [123,124] and transforming growth factor β (TGF-β) as a pro-resolution mediator [21]. The activation of other proinflammatory cytokines (e.g., IL-1α, IL-2, IL-18) and the NF-κB signalling pathway are consistent across various tissues, including the cochlea [125,126,127,128,129,130]. Inflammaging is associated with the release of multiple inflammatory mediators, such as interleukins IL-1, IL-2, IL-6, IL-8, IL-12, IL-13, IL-15, IL-18, IL-22, IL-23, TNF-α, and interferon-γ (IFN-γ). Anti-inflammatory cytokines IL-1Ra, IL-4, IL-10, and TGF-β, are also released, while lipoxin A4 and heat shock proteins act as cytokine mediators [65,131].

Oxidative Stress (OS) and Inflammation

OS is a well-established mechanism of sensorineural hearing loss, including ARHL [132,133]. OS is caused by the overproduction and accumulation of reactive oxygen species (ROS) [134]. ROS, produced as a by-product of cellular respiration in mitochondria, are physiologically relevant due to their role in cellular signalling and homeostasis [135]. However, multiple stressors can result in ROS overproduction and tissue accumulation [22,134]
Accumulation of ROS and other free radicals can impair DNA and break down cellular proteins and lipids, contributing to apoptotic cell death in the cochlea [85]. OS also contributes to the cellular senescence pattern in the ageing cochlea [1]. Rivas-Chacon and colleagues reported that OS and premature senescence significantly impaired cell viability in three auditory cell lines (HEI-OC1, OC-k3, and SV-k1) [1]. In addition, ROS overproduction can induce cochlear inflammation via inflammasomes [136] and thus enhance age-related sensory cell degeneration [137,138].
Inflammasomes are multiprotein oligomer components of the innate immune system, consisting of caspase 1, PYCARD (pyrin domain- or caspase activation and recruitment domain-only proteins), and pattern recognition receptors (PRRs), expressed on macrophages [139,140,141]. Inflammasome-forming PRRs include Nod-like receptor pyrin (NLRP) domain-containing proteins 1, 3, 6, 7 and 12 [142].
NLRP3 is associated with neurogenerative disorders [103,143], and genes encoding NLPR3 are also expressed in the cochlea [144,145]. NLRP3 is a ROS sensor, and it can bind to caspase 1 via apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain (ASC), a key adaptor molecule required for inflammatory processes [146]. The activation of caspase 1 allows for the maturation of downstream inflammatory cytokines such as interleukin-1 beta (IL-1β) and IL-18, inducing an early inflammatory response and lysis of cells by pyroptosis (a form of programmed cell death associated with inflammatory responses to microbes) [136,147,148]. NF-κB pathway activated by toll-like receptor 4 (TLR4) also up-regulates NLRP3 [149], leading to NLRP3 inflammasome priming and sterile inflammation [98]. Similarly, ROS overproduction can activate the NLRP3 inflammasome, which leads to the release of proinflammatory cytokines IL-1β and IL-18 and the activation of macrophages and microglia [98]. Based on this evidence, it was proposed that the sterile inflammation triggered by the activation of inflammasomes in the cochleae of ageing mice likely plays an important role in the pathogenesis of ARHL [136].

6. Association of the Gut Microbiome with Ageing and Immune System Dysfunction

Inflammaging may also result from gut dysbiosis [150]. The gut microbiome consists of the trillions of microorganisms and their genetic material in our intestinal tract. These microorganisms, mainly comprising bacteria, are critical to our health and wellbeing. However, the gut microbiome changes as we age, influencing the functions of other organs. In animal models, the permeability of intestinal barriers increases with age [150,151,152], suggesting that the leakage of gut metabolites in the elderly can potentially cause a systemic inflammatory response [22,150,153].
In young individuals, the microbiome landscape is dominated by Firmicutes and, to a slightly lesser extent, Bacteriodetes [154]. However, the elderly have a diminished population of Firmicutes and atypical Bacteriodetes/Firmicutes ratios [155] with a predominant population of Bacteriodetes [156]. Reduced microbiota diversity in aged individuals may result in the expansion of distinct groups of bacteria implicated in developing age-related type 1 diabetes mellitus, rheumatoid arthritis, and colitis [150]. This landscape can vary within certain groups from different geographical locations and is influenced by diet and genetic backgrounds [150,155,157]. For example, healthy centenarians and supercentenarians have abundant bacterial species that are considered beneficial, such as Gram-positive Bifidobacteria and non-spore-forming, anaerobic, and nonmotile Christensenella [155,157].
Bacterial metabolites and by-products can have different effects on the body. Some Firmicutes produce a short-chain fatty acid, butyrate [158]. Butyrate can mitigate age-related disorders via histone deacetylase inhibition [158] and suppression of inflammatory pathways [159,160,161], which also limits the permeability of the BBB [162]. In contrast, the microbiome in the elderly is rich in proinflammatory microbiota, such as LPS-producing Gram-negative bacteria [155,157,163], and low in butyrate producers [164]. This alteration of the gut microbiome is similar to that seen in the consumers of a high-fat diet [22].
The gut microbiota is considered a master regulator of immune homeostasis [165]. The absence of gut microbiota in germ-free mice impairs the development and maturation of the immune system, while its presence in the gut induces T regulatory cells, immunoglobulin A (IgA)-secreting B cells, Th17 cells, and type-2 lymphoid innate cells [165]. Recent studies strongly suggest that the gut microbiome can modulate the neuro-metabolic signalling pathways in the brain via a bidirectional communication with the endocrine and immune systems and thus contribute to the development of neuroinflammation and neurodegeneration [166]. It has been established that gut dysbiosis affects the permeability of the BBB by reducing the expression of tight-junction proteins, thus causing barrier dysfunction [162]. Previous studies on elderly mice with altered intestinal microbiota have shown increased levels of circulating proinflammatory cytokines compared to young mice, which was associated with anxiety-like behaviour, impaired object–place recognition memory, and social recognition [167]. In addition, gut dysbiosis associated with inflammatory bowel and celiac disease was linked to neurological manifestations [168].

7. Is Gut Dysbiosis Associated with Age-Related Hearing Loss?

In this review, we intended to capture the contribution of inflammatory processes to ARHL, and we have included age-dependent gut dysbiosis as a hypothetical link. In support, it was demonstrated that age-related dyslipidemia [169,170] and a high-fat diet [22] could lead to cochlear inflammation [171,172]. Obese mice display increased expression levels of hypoxia-induced factor 1 (HIF-1), TNF-α, NF-κB, caspase 3, poly (ADP-ribose) polymerase-1, and apoptosis-inducing factor in cochlear tissues (spiral ganglion and spiral ligament) [22,172]. Furthermore, mice fed a high-fat diet long-term showed increased immunoexpression of proinflammatory mediators in the cochlea, including intracellular adhesion molecule 1 (ICAM1), interleukin 6 receptor α (IL6Rα), and toll-like-receptor 2 (TLR2) [173]. These studies support the role of dyslipidemia and gut dysbiosis in cochlear inflammation and imply age-related changes in gut microbiota as underlying factors contributing to accelerated ARHL.
Population health studies have demonstrated that proinflammatory food with high sugar content and consumption of beer and spirits were associated with a higher prevalence of ARHL [174]. In contrast, meta-analyses and systematic reviews of observational studies have reported lower serum concentrations of C-Reactive Protein (CRP), IL-6, and TNF-α among vegetarian and Mediterranean diet eaters compared with omnivores [175]. Differences in the diet of elderly individuals may predict their vulnerability to age-associated disorders [163]. A healthy diet rich in antioxidants is associated with a decreased risk of developing ARHL [176], while a high-fat (Western) diet can cause the accumulation of ROS in the cochlea and chronic inflammation [22,171]. These studies suggest that diet could be a modifiable factor in preventing ARHL.
Therefore, we postulate that a proinflammatory gut environment associated with ageing could result in a leaky gut and the translocation of pathogens and their metabolites via the systemic circulation to distant organs, including the brain and the cochlea [22,168,173]. Systemic inflammation can affect the BBB’s and BLB’s permeability and thus spread to the brain and inner ear tissues, causing local and chronic inflammation [22,177]. The proposed relationship between ageing, gut dysbiosis, and hearing loss is presented as a conceptual model in Figure 1.

8. Conclusions

Presbyacusis has a complex pathophysiology that is not fully understood. Here, we focus on the role of chronic, unresolved inflammation (inflammaging) and immunosenescence in the pathogenesis of ARHL. We also postulate that gut dysbiosis contributes to chronic inflammation in distant organs, including the inner ear. The concept of the gut microbiota - inner ear axis is novel, and the experimental and clinical evidence for the link between gut dysbiosis and presbyacusis is still sparse. Another limitation of this review is a potential selection bias of articles and the lack of rigorous evaluation of selected papers for validity. However, the association between gut dysbiosis and inflammaging could represent a new link to accelerated ARHL that should be explored in future studies.

Author Contributions

Conceptualisation, D.K.; methodology, D.K.; validation, S.V.; investigation, D.K.; resources, D.K.; data curation, S.V.; writing—original draft preparation, D.K.; writing—review and editing, D.K. and S.V.; visualisation, D.K.; supervision, S.V.; project administration, S.V.; funding acquisition, S.V. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by Eisdell Moore Centre (Auckland, New Zealand), grant number 3721994.

Acknowledgments

We thank Corey Beran for proofreading this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rivas-Chacon, L.D.M.; Martinez-Rodriguez, S.; Madrid-Garcia, R.; Yanes-Diaz, J.; Riestra-Ayora, J.I.; Sanz-Fernandez, R.; Sanchez-Rodriguez, C. Role of oxidative stress in the senescence pattern of auditory cells in age-related hearing loss. Antioxidants 2021, 10, 1497. [Google Scholar] [CrossRef]
  2. Haile, L.M.; Kamenov, K.; Briant, P.S.; Orji, A.U.; Steinmetz, J.D.; Abdoli, A.; Abdollahi, M.; Abu-Gharbieh, E.; Afshin, A.; Ahmed, H.; et al. Hearing loss prevalence and years lived with disability, 1990–2019: Findings from the global burden of disease study 2019. Lancet 2021, 397, 996–1009. [Google Scholar] [CrossRef]
  3. Kiely, K.M.; Mitchell, P.; Gopinath, B.; Luszcz, M.A.; Jagger, C.; Anstey, K.J. Estimating the years lived with and without age-related sensory impairment. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2016, 71, 637–642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. World Health Organization. World Report on Hearing—Executive Summary; World Health Organization: Geneva, Switzerland, 2021. [Google Scholar]
  5. HCIA. The Social and Economic Cost of Hearing Loss in Australia; Deloitte Access Economics: Canberra, ACT, Australia, 2017. [Google Scholar]
  6. Kiely, K.M.; Gopinath, B.; Mitchell, P.; Luszcz, M.; Anstey, K.J. Cognitive, health, and sociodemographic predictors of longitudinal decline in hearing acuity among older adults. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2012, 67, 997–1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Gates, G.A.; Mills, J.H. Presbycusis. Lancet 2005, 366, 1111–1120. [Google Scholar] [CrossRef]
  8. Wang, J.; Puel, J.L. Presbycusis: An update on cochlear mechanisms and therapies. J. Clin. Med. 2020, 9, 218. [Google Scholar] [CrossRef] [Green Version]
  9. Samocha-Bonet, D.; Wu, B.; Ryugo, D.K. Diabetes mellitus and hearing loss: A review. Ageing Res. Rev. 2021, 71, 101423. [Google Scholar] [CrossRef]
  10. Horikawa, C.; Kodama, S.; Tanaka, S.; Fujihara, K.; Hirasawa, R.; Yachi, Y.; Shimano, H.; Yamada, N.; Saito, K.; Sone, H. Diabetes and risk of hearing impairment in adults: A meta-analysis. J. Clin. Endocrinol. Metab. 2013, 98, 51–58. [Google Scholar] [CrossRef] [Green Version]
  11. Besser, J.; Stropahl, M.; Urry, E.; Launer, S. Comorbidities of hearing loss and the implications of multimorbidity for audiological care. Hear. Res. 2018, 369, 3–14. [Google Scholar] [CrossRef]
  12. Dawes, P.; Emsley, R.; Cruickshanks, K.J.; Moore, D.R.; Fortnum, H.; Edmondson-Jones, M.; McCormack, A.; Munro, K.J. Hearing loss and cognition: The role of hearing AIDS, social isolation and depression. PLoS ONE 2015, 10, e0119616. [Google Scholar] [CrossRef] [Green Version]
  13. Yuan, J.; Sun, Y.; Sang, S.; Pham, J.H.; Kong, W.-J. The risk of cognitive impairment associated with hearing function in older adults: A pooled analysis of data from eleven studies. Sci. Rep. 2018, 8, 2137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Livingston, G.; Sommerlad, A.; Orgeta, V.; Costafreda, S.G.; Huntley, J.; Ames, D.; Ballard, C.; Banerjee, S.; Burns, A.; Cohen-Mansfield, J.; et al. Dementia prevention, intervention, and care. Lancet 2017, 390, 2673–2734. [Google Scholar] [CrossRef] [Green Version]
  15. Davis, A.; McMahon, C.M.; Pichora-Fuller, K.M.; Russ, S.; Lin, F.; Olusanya, B.O.; Chadha, S.; Tremblay, K.L. Aging and hearing health: The life-course approach. Gerontologist 2016, 56 (Suppl. S2), S256–S267. [Google Scholar] [CrossRef] [PubMed]
  16. Uchida, Y.; Sugiura, S.; Nishita, Y.; Saji, N.; Sone, M.; Ueda, H. Age-related hearing loss and cognitive decline—The potential mechanisms linking the two. Auris Nasus Larynx 2019, 46, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Shen, Y.; Ye, B.; Chen, P.; Wang, Q.; Fan, C.; Shu, Y.; Xiang, M. Cognitive decline, dementia, Alzheimer’s disease and presbycusis: Examination of the possible molecular mechanism. Front. Neurosci. 2018, 12, 394. [Google Scholar] [CrossRef] [PubMed]
  18. Fortunato, S.; Forli, F.; Guglielmi, V.; de Corso, E.; Paludetti, G.; Berrettini, S.; Fetoni, A.R. A review of new insights on the association between hearing loss and cognitive decline in ageing. Acta Otorhinolaryngol. Ital. 2016, 36, 155–166. [Google Scholar] [CrossRef]
  19. Mitchell, P.; Gopinath, B.; McMahon, C.M.; Rochtchina, E.; Wang, J.J.; Boyages, S.C.; Leeder, S.R. Relationship of type 2 diabetes to the prevalence, incidence and progression of age-related hearing loss. Diabet. Med. J. Br. Diabet. Assoc. 2009, 26, 483–488. [Google Scholar] [CrossRef]
  20. Bowl, M.R.; Dawson, S.J. Age-related hearing loss. Cold Spring Harb. Perspect. Med. 2019, 9, a033217. [Google Scholar] [CrossRef] [Green Version]
  21. Watson, N.; Ding, B.; Zhu, X.; Frisina, R.D. Chronic inflammation—Inflammaging—In the ageing cochlea: A novel target for future presbycusis therapy. Ageing Res. Rev. 2017, 40, 142–148. [Google Scholar] [CrossRef]
  22. Kociszewska, D.; Chan, J.; Thorne, P.R.; Vlajkovic, S.M. The link between gut dysbiosis caused by a high-fat diet and hearing loss. Int. J. Mol. Sci. 2021, 22, 13177. [Google Scholar] [CrossRef]
  23. Liu, S.; Gao, J.; Zhu, M.; Liu, K.; Zhang, H.L. Gut microbiota and dysbiosis in Alzheimer’s disease: Implications for pathogenesis and treatment. Mol. Neurobiol. 2020, 57, 5026–5043. [Google Scholar] [CrossRef] [PubMed]
  24. Cunningham, A.L.; Stephens, J.W.; Harris, D.A. Gut microbiota influence in type 2 diabetes mellitus (T2DM). Gut Pathog. 2021, 13, 50. [Google Scholar] [CrossRef] [PubMed]
  25. Vaiserman, A.; Lushchak, O. Developmental origins of type 2 diabetes: Focus on epigenetics. Ageing Res. Rev. 2019, 55, 100957. [Google Scholar] [CrossRef] [PubMed]
  26. Pal, S.; Tyler, J.K. Epigenetics and aging. Sci. Adv. 2016, 2, e1600584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Johnson, K.R.; Zheng, Q.Y. Ahl2, a second locus affecting age-related hearing loss in mice. Genomics 2002, 80, 461–464. [Google Scholar] [CrossRef] [PubMed]
  28. Wang, B.; Jenkins, J.R.; Trayhurn, P. Expression and secretion of inflammation-related adipokines by human adipocytes differentiated in culture: Integrated response to TNF-alpha. Am. J. Physiol. Endocrinol. Metab. 2005, 288, E731–E740. [Google Scholar] [CrossRef]
  29. Yang, C.H.; Schrepfer, T.; Schacht, J. Age-related hearing impairment and the triad of acquired hearing loss. Front. Cell. Neurosci. 2015, 9, 276. [Google Scholar] [CrossRef]
  30. Dawes, P.; Cruickshanks, K.J.; Moore, D.R.; Edmondson-Jones, M.; McCormack, A.; Fortnum, H.; Munro, K.J. Cigarette smoking, passive smoking, alcohol consumption, and hearing loss. J. Assoc. Res. Otolaryngol. 2014, 15, 663–674. [Google Scholar] [CrossRef] [Green Version]
  31. Yamasoba, T.; Lin, F.R.; Someya, S.; Kashio, A.; Sakamoto, T.; Kondo, K. Current concepts in age-related hearing loss: Epidemiology and mechanistic pathways. Hear. Res. 2013, 303, 30–38. [Google Scholar] [CrossRef] [Green Version]
  32. Bazard, P.; Pineros, J.; Frisina, R.D.; Bauer, M.A.; Acosta, A.A.; Paganella, L.R.; Borakiewicz, D.; Thivierge, M.; Mannering, F.L.; Zhu, X.; et al. Cochlear inflammaging in relation to ion channels and mitochondrial functions. Cells 2021, 10, 2761. [Google Scholar] [CrossRef]
  33. Keithley, E.M. Pathology and mechanisms of cochlear aging. J. Neurosci. Res. 2020, 98, 1674–1684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Schuknecht, H.F. Presbycusis. Laryngoscope 1955, 65, 402–419. [Google Scholar] [CrossRef]
  35. Schuknecht, H.F.; Gacek, M.R. Cochlear pathology in presbycusis. Ann. Otol. Rhinol. Laryngol. 1993, 102, 1–16. [Google Scholar] [CrossRef]
  36. Ohlemiller, K.K. Age-related hearing loss: The status of Schuknecht’s typology. Curr. Opin. Otolaryngol. Head Neck Surg. 2004, 12, 439–443. [Google Scholar] [CrossRef] [PubMed]
  37. Viana, L.M.; O’Malley, J.T.; Burgess, B.J.; Jones, D.D.; Oliveira, C.A.; Santos, F.; Merchant, S.N.; Liberman, L.D.; Liberman, M.C. Cochlear neuropathy in human presbycusis: Confocal analysis of hidden hearing loss in post-mortem tissue. Hear. Res. 2015, 327, 78–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Wu, P.Z.; Liberman, L.D.; Bennett, K.; de Gruttola, V.; O’Malley, J.T.; Liberman, M.C. Primary Neural degeneration in the human cochlea: Evidence for hidden hearing loss in the aging ear. Neuroscience 2019, 407, 8–20. [Google Scholar] [CrossRef]
  39. Coppell, K.J.; Galts, C.P.; Huizing, F.Y.; Norton, J.K.; Gray, A.R.; Schultz, K.; Hobbs, C.E.; Aluzaite, K.; Schultz, M. Annual incidence and phenotypic presentation of IBD in southern New Zealand: An 18-year epidemiological analysis. Inflamm. Intest. Dis. 2018, 3, 32–39. [Google Scholar] [CrossRef]
  40. Tu, N.C.; Friedman, R.A. Age-related hearing loss: Unraveling the pieces. Laryngoscope Investig. Otolaryngol. 2018, 3, 68–72. [Google Scholar] [CrossRef] [Green Version]
  41. Carraro, M.; Harrison, R.V. Degeneration of stria vascularis in age-related hearing loss; a corrosion cast study in a mouse model. Acta Oto-Laryngol. 2016, 136, 385–390. [Google Scholar] [CrossRef]
  42. Fetoni, A.R.; Picciotti, P.M.; Paludetti, G.; Troiani, D. Pathogenesis of presbycusis in animal models: A review. Exp. Gerontol. 2011, 46, 413–425. [Google Scholar] [CrossRef]
  43. Gratton, M.A.; Schmiedt, R.A.; Schulte, B.A. Age-related decreases in endocochlear potential are associated with vascular abnormalities in the stria vascularis. Hear. Res. 1996, 102, 181–190. [Google Scholar] [CrossRef]
  44. Bazard, P.; Frisina, R.D.; Acosta, A.A.; Dasgupta, S.; Bauer, M.A.; Zhu, X.; Ding, B. Roles of key ion channels and transport proteins in age-related hearing loss. Int. J. Mol. Sci. 2021, 22, 6158. [Google Scholar] [CrossRef] [PubMed]
  45. Liu, H.; Li, Y.; Chen, L.; Zhang, Q.; Pan, N.; Nichols, D.H.; Zhang, W.J.; Fritzsch, B.; He, D.Z. Organ of corti and stria vascularis: Is there an interdependence for survival? PLoS ONE 2016, 11, e0168953. [Google Scholar] [CrossRef] [Green Version]
  46. Takeuchi, S.; Ando, M.; Kakigi, A. Mechanism generating endocochlear potential: Role played by intermediate cells in stria vascularis. Biophys. J. 2000, 79, 2572–2582. [Google Scholar] [CrossRef] [Green Version]
  47. Dallos, P.; Oertel, D. The Senses a Comprehensive Reference; Elsevier: Amsterdam, The Netherlands, 2008. [Google Scholar]
  48. Chen, J.; Zhao, H.B. The role of an inwardly rectifying K(+) channel (Kir4.1) in the inner ear and hearing loss. Neuroscience 2014, 265, 137–146. [Google Scholar] [CrossRef] [Green Version]
  49. Mills, J.H.; Schmiedt, R.A.; Schulte, B.A.; Dubno, J.R. Age-related hearing loss: A loss of voltage, not hair cells. In Seminars in Hearing; Thieme Medical Publishers, Inc.: New York, NY, USA, 2006; Volume 27, pp. 228–236. [Google Scholar]
  50. Ohlemiller, K.; Frisina, R.; Schacht, J.; Popper, A.; Fay, R. Auditory Trauma, Protection, and Repair; Springer: Boston, MA, USA, 2008; pp. 145–194. [Google Scholar]
  51. Pan, C.C.; Chu, H.Q.; Lai, Y.B.; Sun, Y.B.; Du, Z.H.; Liu, Y.; Chen, J.; Tong, T.; Chen, Q.G.; Zhou, L.Q.; et al. Downregulation of inwardly rectifying potassium channel 5.1 expression in C57BL/6J cochlear lateral wall. J. Huazhong Univ. Sci. Technol. 2016, 36, 406–409. [Google Scholar] [CrossRef] [PubMed]
  52. Pauler, M.; Schuknecht, H.F.; White, J.A. Atrophy of the stria vascularis as a cause of sensorineural hearing loss. Laryngoscope 1988, 98, 754–759. [Google Scholar] [CrossRef]
  53. Schmiedt, R.A.; Lang, H.; Okamura, H.O.; Schulte, B.A. Effects of furosemide applied chronically to the round window: A model of metabolic presbyacusis. J. Neurosci. Off. J. Soc. Neurosci. 2002, 22, 9643–9650. [Google Scholar] [CrossRef] [Green Version]
  54. Schuknecht, H.F.; Montandon, P. Pathology of the ear in pneumococcal meningitis. Arch. Klin. Exp. Ohren Nasen Kehlkopfheilkd. 1970, 195, 207–225. [Google Scholar] [CrossRef]
  55. Ding, B.; Walton, J.P.; Zhu, X.; Frisina, R.D. Age-related changes in Na, K-ATPase expression, subunit isoform selection and assembly in the stria vascularis lateral wall of mouse cochlea. Hear. Res. 2018, 367, 59–73. [Google Scholar] [CrossRef]
  56. Noben-Trauth, K.; Zheng, Q.Y.; Johnson, K.R. Association of cadherin 23 with polygenic inheritance and genetic modification of sensorineural hearing loss. Nat. Genet. 2003, 35, 21–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Kujawa, S.G.; Liberman, M.C. Adding insult to injury: Cochlear nerve degeneration after “temporary” noise-induced hearing loss. J. Neurosci. Off. J. Soc. Neurosci. 2009, 29, 14077–14085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Parthasarathy, A.; Kujawa, S.G. Synaptopathy in the aging cochlea: Characterizing early-neural deficits in auditory temporal envelope processing. J. Neurosci. Off. J. Soc. Neurosci. 2018, 38, 7108–7119. [Google Scholar] [CrossRef] [Green Version]
  59. Sergeyenko, Y.; Lall, K.; Liberman, M.C.; Kujawa, S.G. Age-related cochlear synaptopathy: An early-onset contributor to auditory functional decline. J. Neurosci. Off. J. Soc. Neurosci. 2013, 33, 13686–13694. [Google Scholar] [CrossRef]
  60. Finger, C.E.; Moreno-Gonzalez, I.; Gutierrez, A.; Moruno-Manchon, J.F.; McCullough, L.D. Age-related immune alterations and cerebrovascular inflammation. Mol. Psychiatry 2022, 27, 803–818. [Google Scholar] [CrossRef] [PubMed]
  61. Gruver, A.L.; Hudson, L.L.; Sempowski, G.D. Immunosenescence of ageing. J. Pathol. 2007, 211, 144–156. [Google Scholar] [CrossRef] [PubMed]
  62. Capri, M.; Monti, D.; Salvioli, S.; Lescai, F.; Pierini, M.; Altilia, S.; Sevini, F.; Valensin, S.; Ostan, R.; Bucci, L.; et al. Complexity of anti-immunosenescence strategies in humans. Artif. Organs 2006, 30, 730–742. [Google Scholar] [CrossRef]
  63. Verschuur, C.; Agyemang-Prempeh, A.; Newman, T.A. Inflammation is associated with a worsening of presbycusis: Evidence from the MRC national study of hearing. Int. J. Audiol. 2014, 53, 469–475. [Google Scholar] [CrossRef]
  64. Fulop, T.; Dupuis, G.; Witkowski, J.M.; Larbi, A. The role of immunosenescence in the development of age-related diseases. Rev. Investig. Clin. Organo Hosp. Enferm. Nutr. 2016, 68, 84–91. [Google Scholar]
  65. Santoro, A.; Bientinesi, E.; Monti, D. Immunosenescence and inflammaging in the aging process: Age-related diseases or longevity? Ageing Res. Rev. 2021, 71, 101422. [Google Scholar] [CrossRef]
  66. Franceschi, C.; Garagnani, P.; Parini, P.; Giuliani, C.; Santoro, A. Inflammaging: A new immune-metabolic viewpoint for age-related diseases. Nat. Rev. Endocrinol. 2018, 14, 576–590. [Google Scholar] [CrossRef] [PubMed]
  67. Ferrucci, L.; Fabbri, E. Inflammageing: Chronic inflammation in ageing, cardiovascular disease, and frailty. Nat. Rev. Cardiol. 2018, 15, 505–522. [Google Scholar] [CrossRef] [PubMed]
  68. Franceschi, C.; Bonafe, M.; Valensin, S.; Olivieri, F.; de Luca, M.; Ottaviani, E.; de Benedictis, G. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann. N. Y. Acad. Sci. 2000, 908, 244–254. [Google Scholar] [CrossRef] [PubMed]
  69. Muller, L.; di Benedetto, S.; Pawelec, G. The immune system and its dysregulation with aging. Subcell. Biochem. 2019, 91, 21–43. [Google Scholar] [CrossRef]
  70. Chung, H.Y.; Kim, H.J.; Kim, K.W.; Choi, J.S.; Yu, B.P. Molecular inflammation hypothesis of aging based on the anti-aging mechanism of calorie restriction. Microsc. Res. Tech. 2002, 59, 264–272. [Google Scholar] [CrossRef]
  71. Murakami, M.; Hirano, T. The molecular mechanisms of chronic inflammation development. Front. Immunol. 2012, 3, 323. [Google Scholar] [CrossRef] [Green Version]
  72. Sanada, F.; Taniyama, Y.; Muratsu, J.; Otsu, R.; Shimizu, H.; Rakugi, H.; Morishita, R. Source of chronic inflammation in aging. Front. Cardiovasc. Med. 2018, 5, 12. [Google Scholar] [CrossRef] [Green Version]
  73. Blake, G.J.; Ridker, P.M. Inflammatory bio-markers and cardiovascular risk prediction. J. Intern. Med. 2002, 252, 283–294. [Google Scholar] [CrossRef]
  74. Dandona, P.; Aljada, A.; Bandyopadhyay, A. Inflammation: The link between insulin resistance, obesity and diabetes. Trends Immunol. 2004, 25, 4–7. [Google Scholar] [CrossRef]
  75. Dorfmuller, P.; Perros, F.; Balabanian, K.; Humbert, M. Inflammation in pulmonary arterial hypertension. Eur. Respir. J. 2003, 22, 358–363. [Google Scholar] [CrossRef] [Green Version]
  76. Harrison, D.G.; Guzik, T.J.; Lob, H.E.; Madhur, M.S.; Marvar, P.J.; Thabet, S.R.; Vinh, A.; Weyand, C.M. Inflammation, immunity, and hypertension. Hypertension 2011, 57, 132–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Heppner, F.L.; Ransohoff, R.M.; Becher, B. Immune attack: The role of inflammation in Alzheimer disease. Nat. Rev. Neurosci. 2015, 16, 358–372. [Google Scholar] [CrossRef] [PubMed]
  78. Koenig, W.; Sund, M.; Frohlich, M.; Fischer, H.G.; Lowel, H.; Doring, A.; Hutchinson, W.L.; Pepys, M.B. C-reactive protein, a sensitive marker of inflammation, predicts future risk of coronary heart disease in initially healthy middle-aged men: Results from the MONICA (monitoring trends and determinants in cardiovascular disease) augsburg cohort study, 1984 to 1992. Circulation 1999, 99, 237–242. [Google Scholar] [CrossRef] [Green Version]
  79. Koziorowski, D.; Tomasiuk, R.; Szlufik, S.; Friedman, A. Inflammatory cytokines and NT-proCNP in Parkinson’s disease patients. Cytokine 2012, 60, 762–766. [Google Scholar] [CrossRef]
  80. Luft, V.C.; Schmidt, M.I.; Pankow, J.S.; Couper, D.; Ballantyne, C.M.; Young, J.H.; Duncan, B.B. Chronic inflammation role in the obesity-diabetes association: A case-cohort study. Diabetol. Metab. Syndr. 2013, 5, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Stenvinkel, P.; Heimburger, O.; Paultre, F.; Diczfalusy, U.; Wang, T.; Berglund, L.; Jogestrand, T. Strong association between malnutrition, inflammation, and atherosclerosis in chronic renal failure. Kidney Int. 1999, 55, 1899–1911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Walker, D.G.; Dalsing-Hernandez, J.E.; Campbell, N.A.; Lue, L.F. Decreased expression of CD200 and CD200 receptor in Alzheimer’s disease: A potential mechanism leading to chronic inflammation. Exp. Neurol. 2009, 215, 5–19. [Google Scholar] [CrossRef] [Green Version]
  83. Fujioka, M.; Kanzaki, S.; Okano, H.J.; Masuda, M.; Ogawa, K.; Okano, H. Proinflammatory cytokines expression in noise-induced damaged cochlea. J. Neurosci. Res. 2006, 83, 575–583. [Google Scholar] [CrossRef] [Green Version]
  84. Fujioka, M.; Okano, H.; Ogawa, K. Inflammatory and immune responses in the cochlea: Potential therapeutic targets for sensorineural hearing loss. Front. Pharmacol. 2014, 5, 287. [Google Scholar] [CrossRef] [Green Version]
  85. Paplou, V.; Schubert, N.M.A.; Pyott, S.J. Age-related changes in the cochlea and vestibule: Shared patterns and processes. Front. Neurosci. 2021, 15, 680856. [Google Scholar] [CrossRef]
  86. Harris, J.P. Immunology of the inner ear: Response of the inner ear to antigen challenge. Otolaryngol.-Head Neck Surg. 1983, 91, 18–32. [Google Scholar] [CrossRef] [PubMed]
  87. Kampfe Nordstrom, C.; Danckwardt-Lilliestrom, N.; Laurell, G.; Liu, W.; Rask-Andersen, H. The human endolymphatic sac and inner ear immunity: Macrophage interaction and molecular expression. Front. Immunol. 2018, 9, 3181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Shi, X. Pathophysiology of the cochlear intrastrial fluid-blood barrier (review). Hear. Res. 2016, 338, 52–63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Nyberg, S.; Abbott, N.J.; Shi, X.; Steyger, P.S.; Dabdoub, A. Delivery of therapeutics to the inner ear: The challenge of the blood-labyrinth barrier. Sci. Transl. Med. 2019, 11, eaao0935. [Google Scholar] [CrossRef]
  90. Zhang, F.; Zhang, J.; Neng, L.; Shi, X. Characterization and inflammatory response of perivascular-resident macrophage-like melanocytes in the vestibular system. J. Assoc. Res. Otolaryngol. 2013, 14, 635–643. [Google Scholar] [CrossRef] [Green Version]
  91. Hirose, K.; Hartsock, J.J.; Johnson, S.; Santi, P.; Salt, A.N. Systemic lipopolysaccharide compromises the blood-labyrinth barrier and increases entry of serum fluorescein into the perilymph. J. Assoc. Res. Otolaryngol. 2014, 15, 707–719. [Google Scholar] [CrossRef] [Green Version]
  92. Taylor, R.R.; Nevill, G.; Forge, A. Rapid hair cell loss: A mouse model for cochlear lesions. J. Assoc. Res. Otolaryngol. 2008, 9, 44–64. [Google Scholar] [CrossRef] [Green Version]
  93. Suzuki, M.; Yamasoba, T.; Ishibashi, T.; Miller, J.M.; Kaga, K. Effect of noise exposure on blood-labyrinth barrier in guinea pigs. Hear. Res. 2002, 164, 12–18. [Google Scholar] [CrossRef]
  94. Del Zoppo, G.J.; Hallenbeck, J.M. Advances in the vascular pathophysiology of ischemic stroke. Thromb. Res. 2000, 98, 73–81. [Google Scholar] [CrossRef]
  95. Yamazaki, Y.; Baker, D.J.; Tachibana, M.; Liu, C.C.; van Deursen, J.M.; Brott, T.G.; Bu, G.; Kanekiyo, T. Vascular cell senescence contributes to blood-brain barrier breakdown. Stroke 2016, 47, 1068–1077. [Google Scholar] [CrossRef]
  96. Verschuur, C.A.; Dowell, A.; Syddall, H.E.; Ntani, G.; Simmonds, S.J.; Baylis, D.; Gale, C.R.; Walsh, B.; Cooper, C.; Lord, J.M.; et al. Markers of inflammatory status are associated with hearing threshold in older people: Findings from the hertfordshire ageing study. Age Ageing 2012, 41, 92–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Lassale, C.; Vullo, P.; Cadar, D.; Batty, G.D.; Steptoe, A.; Zaninotto, P. Association of inflammatory markers with hearing impairment: The English longitudinal study of ageing. Brain Behav. Immun. 2020, 83, 112–119. [Google Scholar] [CrossRef] [PubMed]
  98. Noble, K.; Brown, L.; Elvis, P.; Lang, H. Cochlear immune response in presbyacusis: A focus on dysregulation of macrophage activity. J. Assoc. Res. Otolaryngol. 2022, 23, 1–16. [Google Scholar] [CrossRef] [PubMed]
  99. Noble, K.V.; Liu, T.; Matthews, L.J.; Schulte, B.A.; Lang, H. Age-related changes in immune cells of the human cochlea. Front. Neurol. 2019, 10, 895. [Google Scholar] [CrossRef] [Green Version]
  100. Lowthian, J.A.; Britt, C.J.; Rance, G.; Lin, F.R.; Woods, R.L.; Wolfe, R.; Nelson, M.R.; Dillon, H.A.; Ward, S.; Reid, C.M.; et al. Slowing the progression of age-related hearing loss: Rationale and study design of the ASPIRIN in HEARING, retinal vessels imaging and neurocognition in older generations (ASPREE-HEARING) trial. Contemp. Clin. Trials 2016, 46, 60–66. [Google Scholar] [CrossRef]
  101. Kalinec, G.M.; Lomberk, G.; Urrutia, R.A.; Kalinec, F. Resolution of cochlear inflammation: Novel target for preventing or ameliorating drug-, noise- and age-related hearing loss. Front. Cell. Neurosci. 2017, 11, 192. [Google Scholar] [CrossRef] [Green Version]
  102. Coleman, J.; Huang, X.; Liu, J.; Kopke, R.; Jackson, R. Dosing study on the effectiveness of salicylate/N-acetylcysteine for prevention of noise-induced hearing loss. Noise Health 2010, 12, 159–165. [Google Scholar] [CrossRef]
  103. Gao, X.R.; Adhikari, C.M.; Peng, L.Y.; Guo, X.G.; Zhai, Y.S.; He, X.Y.; Zhang, L.Y.; Lin, J.; Zuo, Z.Y. Efficacy of different doses of aspirin in decreasing blood levels of inflammatory markers in patients with cardiovascular metabolic syndrome. J. Pharm. Pharmacol. 2009, 61, 1505–1510. [Google Scholar] [CrossRef]
  104. Yamashita, D.; Jiang, H.Y.; le Prell, C.G.; Schacht, J.; Miller, J.M. Post-exposure treatment attenuates noise-induced hearing loss. Neuroscience 2005, 134, 633–642. [Google Scholar] [CrossRef]
  105. Frye, M.D.; Yang, W.; Zhang, C.; Xiong, B.; Hu, B.H. Dynamic activation of basilar membrane macrophages in response to chronic sensory cell degeneration in aging mouse cochleae. Hear. Res. 2017, 344, 125–134. [Google Scholar] [CrossRef] [Green Version]
  106. Su, Z.; Xiong, H.; Liu, Y.; Pang, J.; Lin, H.; Zhang, W.; Zheng, Y. Transcriptomic analysis highlights cochlear inflammation associated with age-related hearing loss in C57BL/6 mice using next generation sequencing. PeerJ 2020, 8, e9737. [Google Scholar] [CrossRef] [PubMed]
  107. Coppe, J.P.; Desprez, P.Y.; Krtolica, A.; Campisi, J. The senescence-associated secretory phenotype: The dark side of tumor suppression. Annu. Rev. Pathol. 2010, 5, 99–118. [Google Scholar] [CrossRef] [Green Version]
  108. Tchkonia, T.; Zhu, Y.; van Deursen, J.; Campisi, J.; Kirkland, J.L. Cellular senescence and the senescent secretory phenotype: Therapeutic opportunities. J. Clin. Investig. 2013, 123, 966–972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Chinta, S.J.; Woods, G.; Rane, A.; Demaria, M.; Campisi, J.; Andersen, J.K. Cellular senescence and the aging brain. Exp. Gerontol. 2015, 68, 3–7. [Google Scholar] [CrossRef] [Green Version]
  110. Salminen, A.; Ojala, J.; Kaarniranta, K.; Haapasalo, A.; Hiltunen, M.; Soininen, H. Astrocytes in the aging brain express characteristics of senescence-associated secretory phenotype. Eur. J. Neurosci. 2011, 34, 3–11. [Google Scholar] [CrossRef] [PubMed]
  111. Salvador, E.; Burek, M.; Löhr, M.; Nagai, M.; Hagemann, C.; Förster, C.Y. Senescence and associated blood-brain barrier alterations in vitro. Histochem. Cell Biol. 2021, 156, 283–292. [Google Scholar] [CrossRef] [PubMed]
  112. Jenny, N.S. Inflammation in aging: Cause, effect, or both? Discov. Med. 2012, 13, 451–460. [Google Scholar]
  113. Singh, T.; Newman, A.B. Inflammatory markers in population studies of aging. Ageing Res. Rev. 2011, 10, 319–329. [Google Scholar] [CrossRef] [Green Version]
  114. Marzetti, E.; Hwang, J.C.; Lees, H.A.; Wohlgemuth, S.E.; Dupont-Versteegden, E.E.; Carter, C.S.; Bernabei, R.; Leeuwenburgh, C. Mitochondrial death effectors: Relevance to sarcopenia and disuse muscle atrophy. Biochim. Biophys. Acta 2010, 1800, 235–244. [Google Scholar] [CrossRef] [Green Version]
  115. Kundu, T.K. Epigenetics: Development and Disease; Springer Science & Business Media: Berlin/Heidelberg, Germany, 2012; Volume 61. [Google Scholar]
  116. Sohal, R.S.; Orr, W.C. The redox stress hypothesis of aging. Free Radic. Biol. Med. 2012, 52, 539–555. [Google Scholar] [CrossRef] [Green Version]
  117. Tarantino, G.; Savastano, S.; Colao, A. Hepatic steatosis, low-grade chronic inflammation and hormone/growth factor/adipokine imbalance. World J. Gastroenterol. 2010, 16, 4773–4783. [Google Scholar] [CrossRef] [PubMed]
  118. Woods, J.A.; Wilund, K.R.; Martin, S.A.; Kistler, B.M. Exercise, inflammation and aging. Aging Dis. 2012, 3, 130–140. [Google Scholar] [PubMed]
  119. Franceschi, C.; Campisi, J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2014, 69 (Suppl. S1), S4–S9. [Google Scholar] [CrossRef] [PubMed]
  120. Duncan, B.B.; Schmidt, M.I.; Pankow, J.S.; Ballantyne, C.M.; Couper, D.; Vigo, A.; Hoogeveen, R.; Folsom, A.R.; Heiss, G. Low-grade systemic inflammation and the development of type 2 diabetes: The atherosclerosis risk in communities study. Diabetes 2003, 52, 1799–1805. [Google Scholar] [CrossRef] [Green Version]
  121. Kubaszek, A.; Pihlajamaki, J.; Komarovski, V.; Lindi, V.; Lindstrom, J.; Eriksson, J.; Valle, T.T.; Hamalainen, H.; Ilanne-Parikka, P.; Keinanen-Kiukaanniemi, S.; et al. Promoter polymorphisms of the TNF-alpha (G-308A) and IL-6 (C-174G) genes predict the conversion from impaired glucose tolerance to type 2 diabetes: The finnish diabetes prevention study. Diabetes 2003, 52, 1872–1876. [Google Scholar] [CrossRef] [Green Version]
  122. Pradhan, A.D.; Manson, J.E.; Rifai, N.; Buring, J.E.; Ridker, P.M. C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA 2001, 286, 327–334. [Google Scholar] [CrossRef]
  123. Popa, C.; Netea, M.G.; van Riel, P.L.; van der Meer, J.W.; Stalenhoef, A.F. The role of TNF-alpha in chronic inflammatory conditions, intermediary metabolism, and cardiovascular risk. J. Lipid Res. 2007, 48, 751–762. [Google Scholar] [CrossRef] [Green Version]
  124. Scheller, J.; Chalaris, A.; Schmidt-Arras, D.; Rose-John, S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim. Biophys. Acta 2011, 1813, 878–888. [Google Scholar] [CrossRef] [Green Version]
  125. Satoh, H.; Billings, P.; Firestein, G.S.; Harris, J.P.; Keithley, E.M. Transforming growth factor beta expression during an inner ear immune response. Ann. Otol. Rhinol. Laryngol. 2006, 115, 81–88. [Google Scholar] [CrossRef]
  126. Satoh, H.; Firestein, G.S.; Billings, P.B.; Harris, J.P.; Keithley, E.M. Proinflammatory cytokine expression in the endolymphatic sac during inner ear inflammation. J. Assoc. Res. Otolaryngol. 2003, 4, 139–147. [Google Scholar] [CrossRef] [Green Version]
  127. Rea, I.M.; Gibson, D.S.; McGilligan, V.; McNerlan, S.E.; Alexander, H.D.; Ross, O.A. Age and age-related diseases: Role of inflammation triggers and cytokines. Front. Immunol. 2018, 9, 586. [Google Scholar] [CrossRef] [PubMed]
  128. Tilstra, J.S.; Clauson, C.L.; Niedernhofer, L.J.; Robbins, P.D. NF-kappaB in aging and disease. Aging Dis. 2011, 2, 449–465. [Google Scholar] [PubMed]
  129. Kaneko, N.; Kurata, M.; Yamamoto, T.; Morikawa, S.; Masumoto, J. The role of interleukin-1 in general pathology. Inflamm. Regen. 2019, 39, 12. [Google Scholar] [CrossRef] [Green Version]
  130. Meftahi, G.H.; Jangravi, Z.; Sahraei, H.; Bahari, Z. The possible pathophysiology mechanism of cytokine storm in elderly adults with COVID-19 infection: The contribution of “inflame-aging”. Inflamm. Res. 2020, 69, 825–839. [Google Scholar] [CrossRef] [PubMed]
  131. Minciullo, P.L.; Catalano, A.; Mandraffino, G.; Casciaro, M.; Crucitti, A.; Maltese, G.; Morabito, N.; Lasco, A.; Gangemi, S.; Basile, G. Inflammaging and anti-inflammaging: The role of cytokines in extreme longevity. Arch. Immunol. Ther. Exp. 2016, 64, 111–126. [Google Scholar] [CrossRef]
  132. Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D.; et al. Oxidative stress, aging, and diseases. Clin. Interv. Aging 2018, 13, 757–772. [Google Scholar] [CrossRef] [Green Version]
  133. Prasad, K.N.; Bondy, S.C. Increased oxidative stress, inflammation, and glutamate: Potential preventive and therapeutic targets for hearing disorders. Mech. Ageing Dev. 2020, 185, 111191. [Google Scholar] [CrossRef]
  134. Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative stress: Harms and benefits for human health. Oxid. Med. Cell. Longev. 2017, 2017, 8416763. [Google Scholar] [CrossRef]
  135. Ray, P.D.; Huang, B.W.; Tsuji, Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell. Signal. 2012, 24, 981–990. [Google Scholar] [CrossRef] [Green Version]
  136. Shi, X.; Qiu, S.; Zhuang, W.; Yuan, N.; Wang, C.; Zhang, S.; Sun, T.; Guo, W.; Gao, F.; Yang, S.; et al. NLRP3-inflammasomes are triggered by age-related hearing loss in the inner ear of mice. Am. J. Transl. Res. 2017, 9, 5611–5618. [Google Scholar]
  137. Fujimoto, C.; Yamasoba, T. Oxidative stresses and mitochondrial dysfunction in age-related hearing loss. Oxid. Med. Cell. Longev. 2014, 2014, 582849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Riva, C.; Donadieu, E.; Magnan, J.; Lavieille, J.-P. Age-related hearing loss in CD/1 mice is associated to ROS formation and HIF target proteins up-regulation in the cochlea. Exp. Gerontol. 2007, 42, 327–336. [Google Scholar] [CrossRef] [PubMed]
  139. Martinon, F. Signaling by ROS drives inflammasome activation. Eur. J. Immunol. 2010, 40, 616–619. [Google Scholar] [CrossRef] [PubMed]
  140. Sorbara, M.T.; Girardin, S.E. Mitochondrial ROS fuel the inflammasome. Cell Res. 2011, 21, 558–560. [Google Scholar] [CrossRef] [PubMed]
  141. Kolli, D.; Velayutham, T.S.; Casola, A. Host-viral interactions: Role of pattern recognition receptors (PRRs) in human pneumovirus infections. Pathogens 2013, 2, 232–263. [Google Scholar] [CrossRef] [Green Version]
  142. Latz, E.; Xiao, T.S.; Stutz, A. Activation and regulation of the inflammasomes. Nat. Rev. Immunol. 2013, 13, 397–411. [Google Scholar] [CrossRef]
  143. Halle, A.; Hornung, V.; Petzold, G.C.; Stewart, C.R.; Monks, B.G.; Reinheckel, T.; Fitzgerald, K.A.; Latz, E.; Moore, K.J.; Golenbock, D.T. The NALP3 inflammasome is involved in the innate immune response to amyloid-β. Nat. Immunol. 2008, 9, 857–865. [Google Scholar] [CrossRef] [Green Version]
  144. Nakanishi, H.; Kawashima, Y.; Kurima, K.; Chae, J.J.; Ross, A.M.; Pinto-Patarroyo, G.; Patel, S.K.; Muskett, J.A.; Ratay, J.S.; Chattaraj, P.; et al. NLRP3 mutation and cochlear autoinflammation cause syndromic and nonsyndromic hearing loss DFNA34 responsive to anakinra therapy. Proc. Natl. Acad. Sci. USA 2017, 114, E7766–E7775. [Google Scholar] [CrossRef]
  145. Nakanishi, H.; Kawashima, Y.; Kurima, K.; Muskett, J.A.; Kim, H.J.; Brewer, C.C.; Griffith, A.J. Gradual symmetric progression of DFNA34 hearing loss caused by an NLRP3 mutation and cochlear autoinflammation. Otol. Neurotol. 2018, 39, e181–e185. [Google Scholar] [CrossRef]
  146. Schroder, K.; Muruve, D.A.; Tschopp, J. Innate immunity: Cytoplasmic DNA sensing by the AIM2 inflammasome. Curr. Biol. 2009, 19, R262–R265. [Google Scholar] [CrossRef] [Green Version]
  147. LaRock, C.N.; Nizet, V. Inflammasome/IL-1beta responses to streptococcal pathogens. Front. Immunol. 2015, 6, 518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Sagulenko, V.; Vitak, N.; Vajjhala, P.R.; Vince, J.E.; Stacey, K.J. Caspase-1 is an apical caspase leading to caspase-3 cleavage in the AIM2 inflammasome response, independent of caspase-8. J. Mol. Biol. 2018, 430, 238–247. [Google Scholar] [CrossRef] [PubMed]
  149. Schroder, K.; Tschopp, J. The inflammasomes. Cell 2010, 140, 821–832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Kim, M.; Benayoun, B.A. The microbiome: An emerging key player in aging and longevity. Transl. Med. Aging 2020, 4, 103–116. [Google Scholar] [CrossRef]
  151. Clark, R.I.; Salazar, A.; Yamada, R.; Fitz-Gibbon, S.; Morselli, M.; Alcaraz, J.; Rana, A.; Rera, M.; Pellegrini, M.; William, W.J.; et al. Distinct shifts in microbiota composition during drosophila aging impair intestinal function and drive mortality. Cell Rep. 2015, 12, 1656–1667. [Google Scholar] [CrossRef] [Green Version]
  152. Rera, M.; Clark, R.I.; Walker, D.W. Intestinal barrier dysfunction links metabolic and inflammatory markers of aging to death in drosophila. Proc. Natl. Acad. Sci. USA 2012, 109, 21528–21533. [Google Scholar] [CrossRef] [Green Version]
  153. Thevaranjan, N.; Puchta, A.; Schulz, C.; Naidoo, A.; Szamosi, J.C.; Verschoor, C.P.; Loukov, D.; Schenck, L.P.; Jury, J.; Foley, K.P.; et al. Age-associated microbial dysbiosis promotes intestinal permeability, systemic inflammation, and macrophage dysfunction. Cell Host Microbe 2017, 21, 455–466.e454. [Google Scholar] [CrossRef] [Green Version]
  154. Eckburg, P.B.; Bik, E.M.; Bernstein, C.N.; Purdom, E.; Dethlefsen, L.; Sargent, M.; Gill, S.R.; Nelson, K.E.; Relman, D.A. Diversity of the human intestinal microbial flora. Science 2005, 308, 1635–1638. [Google Scholar] [CrossRef] [Green Version]
  155. Biagi, E.; Candela, M.; Turroni, S.; Garagnani, P.; Franceschi, C.; Brigidi, P. Ageing and gut microbes: Perspectives for health maintenance and longevity. Pharmacol. Res. 2013, 69, 11–20. [Google Scholar] [CrossRef]
  156. Claesson, M.J.; Cusack, S.; O’Sullivan, O.; Greene-Diniz, R.; de Weerd, H.; Flannery, E.; Marchesi, J.R.; Falush, D.; Dinan, T.; Fitzgerald, G.; et al. Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. S1), 4586–4591. [Google Scholar] [CrossRef] [Green Version]
  157. Biagi, E.; Nylund, L.; Candela, M.; Ostan, R.; Bucci, L.; Pini, E.; Nikkila, J.; Monti, D.; Satokari, R.; Franceschi, C.; et al. Through ageing, and beyond: Gut microbiota and inflammatory status in seniors and centenarians. PLoS ONE 2010, 5, e10667. [Google Scholar] [CrossRef]
  158. Pryde, S.E.; Duncan, S.H.; Hold, G.L.; Stewart, C.S.; Flint, H.J. The microbiology of butyrate formation in the human colon. FEMS Microbiol. Lett. 2002, 217, 133–139. [Google Scholar] [CrossRef] [PubMed]
  159. Klampfer, L.; Huang, J.; Sasazuki, T.; Shirasawa, S.; Augenlicht, L. Inhibition of interferon γ signaling by the short chain fatty acid butyrate11montefiore medical center new research initiative award to LK and the American Cancer Society institutional research grant to LK (ACS IRG# 98-274-01), UO1 CA88104 (to LA), and P30-13330 from NCI. Mol. Cancer Res. 2003, 1, 855–862. [Google Scholar]
  160. Alex, S.; Lange, K.; Amolo, T.; Grinstead, J.S.; Haakonsson, A.K.; Szalowska, E.; Koppen, A.; Mudde, K.; Haenen, D.; Al-Lahham, S.; et al. Short-chain fatty acids stimulate angiopoietin-like 4 synthesis in human colon adenocarcinoma cells by activating peroxisome proliferator-activated receptor gamma. Mol. Cell. Biol. 2013, 33, 1303–1316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Bach Knudsen, K.E.; Laerke, H.N.; Hedemann, M.S.; Nielsen, T.S.; Ingerslev, A.K.; Gundelund Nielsen, D.S.; Theil, P.K.; Purup, S.; Hald, S.; Schioldan, A.G.; et al. Impact of diet-modulated butyrate production on intestinal barrier function and inflammation. Nutrients 2018, 10, 1499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Toth, M.; Korecka, A.; Bakocevic, N.; Ng, L.G.; Kundu, P.; et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 2014, 6, 263ra158. [Google Scholar] [CrossRef] [Green Version]
  163. Park, S.H.; Kim, K.A.; Ahn, Y.T.; Jeong, J.J.; Huh, C.S.; Kim, D.H. Comparative analysis of gut microbiota in elderly people of urbanized towns and longevity villages. BMC Microbiol. 2015, 15, 49. [Google Scholar] [CrossRef] [Green Version]
  164. Hippe, B.; Zwielehner, J.; Liszt, K.; Lassl, C.; Unger, F.; Haslberger, A.G. Quantification of butyryl CoA: Acetate CoA-transferase genes reveals different butyrate production capacity in individuals according to diet and age. FEMS Microbiol. Lett. 2011, 316, 130–135. [Google Scholar] [CrossRef] [Green Version]
  165. Ragonnaud, E.; Biragyn, A. Gut microbiota as the key controllers of “healthy” aging of elderly people. Immun. Ageing 2021, 18, 2. [Google Scholar] [CrossRef]
  166. Askarova, S.; Umbayev, B.; Masoud, A.-R.; Kaiyrlykyzy, A.; Safarova, Y.; Tsoy, A.; Olzhayev, F.; Kushugulova, A. The links between the gut microbiome, aging, modern lifestyle and Alzheimer’s disease. Front. Cell. Infect. Microbiol. 2020, 10, 104. [Google Scholar] [CrossRef] [Green Version]
  167. Scott, K.A.; Ida, M.; Peterson, V.L.; Prenderville, J.A.; Moloney, G.M.; Izumo, T.; Murphy, K.; Murphy, A.; Ross, R.P.; Stanton, C.; et al. Revisiting metchnikoff: Age-related alterations in microbiota-gut-brain axis in the mouse. Brain Behav. Immun. 2017, 65, 20–32. [Google Scholar] [CrossRef] [PubMed]
  168. Kociszewska, D.; Vlajkovic, S.M. The association of inflammatory gut diseases with neuroinflammatory and auditory disorders. Front. Biosci.-Elite 2022, 14, 8. [Google Scholar] [CrossRef] [PubMed]
  169. Cho, S.M.J.; Lee, H.J.; Shim, J.S.; Song, B.M.; Kim, H.C. Associations between age and dyslipidemia are differed by education level: The cardiovascular and metabolic diseases etiology research center (CMERC) cohort. Lipids Health Dis. 2020, 19, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Liu, H.H.; Li, J.J. Aging and dyslipidemia: A review of potential mechanisms. Ageing Res. Rev. 2015, 19, 43–52. [Google Scholar] [CrossRef] [PubMed]
  171. Du, Z.; Yang, Y.; Hu, Y.; Sun, Y.; Zhang, S.; Peng, W.; Zhong, Y.; Huang, X.; Kong, W. A long-term high-fat diet increases oxidative stress, mitochondrial damage and apoptosis in the inner ear of D-galactose-induced aging rats. Hear. Res. 2012, 287, 15–24. [Google Scholar] [CrossRef]
  172. Hwang, J.H.; Hsu, C.J.; Yu, W.H.; Liu, T.C.; Yang, W.S. Diet-induced obesity exacerbates auditory degeneration via hypoxia, inflammation, and apoptosis signaling pathways in CD/1 mice. PLoS ONE 2013, 8, e60730. [Google Scholar] [CrossRef]
  173. Chan, J.; Telang, R.; Kociszewska, D.; Thorne, P.R.; Vlajkovic, S.M. A high-fat diet induces low-grade cochlear inflammation in CD-1 mice. Int. J. Mol. Sci. 2022, 23, 5179. [Google Scholar] [CrossRef]
  174. Sardone, R.; Lampignano, L.; Guerra, V.; Zupo, R.; Donghia, R.; Castellana, F.; Battista, P.; Bortone, I.; Procino, F.; Castellana, M.; et al. Relationship between inflammatory food consumption and age-related hearing loss in a prospective observational cohort: Results from the salus in apulia study. Nutrients 2020, 12, 426. [Google Scholar] [CrossRef] [Green Version]
  175. Hart, M.J.; Torres, S.J.; McNaughton, S.A.; Milte, C.M. Dietary patterns and associations with biomarkers of inflammation in adults: A systematic review of observational studies. Nutr. J. 2021, 20, 24. [Google Scholar] [CrossRef]
  176. Choi, J.E.; Ahn, J.; Moon, I.J. Associations between age-related hearing loss and dietaryassessment using data from Korean national health and nutrition examination survey. Nutrients 2021, 13, 1230. [Google Scholar] [CrossRef]
  177. Erickson, M.A.; Banks, W.A. Age-associated changes in the immune system and blood-brain barrier functions. Int. J. Mol. Sci. 2019, 20, 1632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Takiishi, T.; Fenero, C.I.M.; Camara, N.O.S. Intestinal barrier and gut microbiota: Shaping our immune responses throughout life. Tissue Barriers 2017, 5, e1373208. [Google Scholar] [CrossRef] [PubMed]
  179. Salminen, A.; Huuskonen, J.; Ojala, J.; Kauppinen, A.; Kaarniranta, K.; Suuronen, T. Activation of innate immunity system during aging: NF-kB signaling is the molecular culprit of inflamm-aging. Ageing Res. Rev. 2008, 7, 83–105. [Google Scholar] [CrossRef] [PubMed]
  180. Jurjus, A.; Eid, A.; Al Kattar, S.; Zeenny, M.N.; Gerges-Geagea, A.; Haydar, H.; Hilal, A.; Oueidat, D.; Matar, M.; Tawilah, J.; et al. Inflammatory bowel disease, colorectal cancer and type 2 diabetes mellitus: The links. BBA Clin. 2016, 5, 16–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Sansone, C.; Cohen, J.; Gold, B.; Zhu, W.; Misic, A.M.; Beiting, D.P.; Winter, S.E.; Cherry, S. Aging-associated dysbiosis increases susceptibility to enteric viral infection in Drosophila. bioRxiv 2017. [Google Scholar] [CrossRef] [Green Version]
  182. Jones, R.M.; Neish, A.S. Redox signaling mediated by the gut microbiota. Free Radic. Biol. Med. 2017, 105, 41–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Shoelson, S.E.; Lee, J.; Yuan, M. Inflammation and the IKK beta/I kappa B/NF-kappa B axis in obesity- and diet-induced insulin resistance. Int. J. Obes. Relat. Metab. Disord. 2003, 27 (Suppl. S3), S49–S52. [Google Scholar] [CrossRef] [Green Version]
  184. Zand, H.; Morshedzadeh, N.; Naghashian, F. Signaling pathways linking inflammation to insulin resistance. Diabetes Metab. Syndr. 2017, 11 (Suppl. S1), S307–S309. [Google Scholar] [CrossRef]
  185. Adams, J.C.; Seed, B.; Lu, N.; Landry, A.; Xavier, R.J. Selective activation of nuclear factor kappa B in the cochlea by sensory and inflammatory stress. Neuroscience 2009, 160, 530–539. [Google Scholar] [CrossRef] [Green Version]
  186. Jian-He, S.; Wei-Yan, Y.; Su-Hua, S.; Schacht, J. Nuclear factor kappa B p65 expression in mouse cochlea. J. Otol. 2007, 2, 30–35. [Google Scholar] [CrossRef] [Green Version]
  187. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 inflammasome: An overview of mechanisms of activation and regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [Green Version]
  188. Bauernfeind, F.G.; Horvath, G.; Stutz, A.; Alnemri, E.S.; MacDonald, K.; Speert, D.; Fernandes-Alnemri, T.; Wu, J.; Monks, B.G.; Fitzgerald, K.A.; et al. Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J. Immunol. 2009, 183, 787–791. [Google Scholar] [CrossRef] [PubMed]
  189. Su, Q.; Li, L.; Sun, Y.; Yang, H.; Ye, Z.; Zhao, J. Effects of the TLR4/Myd88/NF-kappaB signaling pathway on NLRP3 inflammasome in coronary microembolization-induced myocardial injury. Cell. Physiol. Biochem. 2018, 47, 1497–1508. [Google Scholar] [CrossRef] [PubMed]
  190. Feng, L.; Nian, S.; Tong, Z.; Zhu, Y.; Li, Y.; Zhang, C.; Bai, X.; Luo, X.; Wu, M.; Yan, Z. Age-related trends in lipid levels: A large-scale cross-sectional study of the general Chinese population. BMJ Open 2020, 10, e034226. [Google Scholar] [CrossRef] [PubMed]
  191. Chi, Y.; Lin, Y.; Lu, Y.; Huang, Q.; Ye, G.; Dong, S. Gut microbiota dysbiosis correlates with a low-dose PCB126-induced dyslipidemia and non-alcoholic fatty liver disease. Sci. Total Environ. 2019, 653, 274–282. [Google Scholar] [CrossRef] [PubMed]
  192. Horne, R.G.; Yu, Y.; Zhang, R.; Abdalqadir, N.; Rossi, L.; Surette, M.; Sherman, P.M.; Adeli, K. High fat-high fructose diet-induced changes in the gut microbiota associated with dyslipidemia in Syrian hamsters. Nutrients 2020, 12, 3557. [Google Scholar] [CrossRef]
  193. Rashnuodi, P.; Amiri, A.; Omidi, M.; Mohammadi, A. The effects of dyslipidemia on noise-induced hearing loss in petrochemical workers in the southwest of Iran. Work 2021, 70, 875–882. [Google Scholar] [CrossRef]
  194. Evans, M.B.; Tonini, R.; Shope, C.D.; Oghalai, J.S.; Jerger, J.F.; Insull, W., Jr.; Brownell, W.E. Dyslipidemia and auditory function. Otol. Neurotol. 2006, 27, 609–614. [Google Scholar] [CrossRef] [Green Version]
  195. Li, X.; Chen, B.; Zhou, X.; Ye, F.; Wang, Y.; Hu, W. Identification of dyslipidemia as a risk factor for sudden sensorineural hearing loss: A multicenter case-control study. J. Clin. Lab. Anal. 2021, 35, e24067. [Google Scholar] [CrossRef]
  196. Dolganiuc, A. Role of lipid rafts in liver health and disease. World J. Gastroenterol. 2011, 17, 2520–2535. [Google Scholar] [CrossRef]
  197. Amiya, E. Interaction of hyperlipidemia and reactive oxygen species: Insights from the lipid-raft platform. World J. Cardiol. 2016, 8, 689–694. [Google Scholar] [CrossRef] [PubMed]
  198. Shirao, S.; Yoneda, H.; Shinoyama, M.; Sugimoto, K.; Koizumi, H.; Ishihara, H.; Oka, F.; Sadahiro, H.; Nomura, S.; Fujii, M.; et al. A novel trigger for cholesterol-dependent smooth muscle contraction mediated by the sphingosylphosphorylcholine-rho-kinase pathway in the rat basilar artery: A mechanistic role for lipid rafts. J. Cereb. Blood Flow Metab. 2015, 35, 835–842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Schieber, M.; Chandel, N.S. ROS function in redox signaling and oxidative stress. Curr. Biol. 2014, 24, R453–R462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Nathan, C.; Cunningham-Bussel, A. Beyond oxidative stress: An immunologist’s guide to reactive oxygen species. Nat. Rev. Immunol. 2013, 13, 349–361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Lingappan, K. NF-kappaB in oxidative stress. Curr. Opin. Toxicol. 2018, 7, 81–86. [Google Scholar] [CrossRef]
  202. Staecker, H.; Zheng, Q.Y.; van de Water, T.R. Oxidative stress in aging in the C57B16/J mouse cochlea. Acta Oto-Laryngol. 2001, 121, 666–672. [Google Scholar] [CrossRef] [PubMed]
  203. Menardo, J.; Tang, Y.; Ladrech, S.; Lenoir, M.; Casas, F.; Michel, C.; Bourien, J.; Ruel, J.; Rebillard, G.; Maurice, T.; et al. Oxidative stress, inflammation, and autophagic stress as the key mechanisms of premature age-related hearing loss in SAMP8 mouse Cochlea. Antioxid. Redox Signal. 2012, 16, 263–274. [Google Scholar] [CrossRef]
  204. Samson, J.; Wiktorek-Smagur, A.; Politanski, P.; Rajkowska, E.; Pawlaczyk-Luszczynska, M.; Dudarewicz, A.; Sha, S.H.; Schacht, J.; Sliwinska-Kowalska, M. Noise-induced time-dependent changes in oxidative stress in the mouse cochlea and attenuation by D-methionine. Neuroscience 2008, 152, 146–150. [Google Scholar] [CrossRef]
  205. Poirrier, A.L.; Pincemail, J.; van den Ackerveken, P.; Lefebvre, P.P.; Malgrange, B. Oxidative stress in the cochlea: An update. Curr. Med. Chem. 2010, 17, 3591–3604. [Google Scholar] [CrossRef]
  206. Huang, T.; Cheng, A.G.; Stupak, H.; Liu, W.; Kim, A.; Staecker, H.; Lefebvre, P.P.; Malgrange, B.; Kopke, R.; Moonen, G.; et al. Oxidative stress-induced apoptosis of cochlear sensory cells: Otoprotective strategies. Int. J. Dev. Neurosci. 2000, 18, 259–270. [Google Scholar] [CrossRef]
  207. Mazurek, B.; Amarjargal, N.; Haupt, H.; Fuchs, J.; Olze, H.; Machulik, A.; Gross, J. Expression of genes implicated in oxidative stress in the cochlea of newborn rats. Hear. Res. 2011, 277, 54–60. [Google Scholar] [CrossRef] [PubMed]
  208. Fetoni, A.R.; Rolesi, R.; Paciello, F.; Eramo, S.L.M.; Grassi, C.; Troiani, D.; Paludetti, G. Styrene enhances the noise induced oxidative stress in the cochlea and affects differently mechanosensory and supporting cells. Free Radic. Biol. Med. 2016, 101, 211–225. [Google Scholar] [CrossRef] [PubMed]
  209. Satar, B.; Ozkaptan, Y.; Surucu, H.S.; Ozturk, H. Ultrastructural effects of hypercholesterolemia on the cochlea. Otol. Neurotol. 2001, 22, 786–789. [Google Scholar] [CrossRef]
  210. Maru, Y.; Jain, N. Lipid profile studies in inner ear dysfunction. Indian J. Otolaryngol. Head Neck Surg. 1994, 46, 17–20. [Google Scholar] [CrossRef]
  211. Odeh, O.; Kuti, M.; Fasunla, A.; Nwaorgu, O. Sensorineural hearing loss and dyslipidemia: Is there any relationship. West Afr. J. Med. 2015, 34, 27–31. [Google Scholar]
  212. Thakur, J.; Mohindroo, N.; Vasanthalakshmi, M.; Kashyap, N.; Azad, R.; Sharma, D. Auditory brainstem evoked responses in hyperlipidaemia: Effect of various lipid fractions on auditory function. J. Laryngol. Otol. 2012, 126, 249–256. [Google Scholar] [CrossRef] [PubMed]
  213. Aimoni, C.; Bianchini, C.; Borin, M.; Ciorba, A.; Fellin, R.; Martini, A.; Scanelli, G.; Volpato, S. Diabetes, cardiovascular risk factors and idiopathic sudden sensorineural hearing loss: A case-control study. Audiol. Neurootol. 2010, 15, 111–115. [Google Scholar] [CrossRef]
  214. Xipeng, L.; Ruiyu, L.; Meng, L.; Yanzhuo, Z.; Kaosan, G.; Liping, W. Effects of diabetes on hearing and cochlear structures. J. Otol. 2013, 8, 82–87. [Google Scholar] [CrossRef] [Green Version]
  215. Pålbrink, A.-K.; Kopietz, F.; Morén, B.; In’t Zandt, R.; Kalinec, F.; Stenkula, K.; Göransson, O.; Holm, C.; Magnusson, M.; Degerman, E. Inner ear is a target for insulin signaling and insulin resistance: Evidence from mice and auditory HEI-OC1 cells. BMJ Open Diabetes Res. Care 2020, 8, e000820. [Google Scholar] [CrossRef] [Green Version]
  216. Palbrink, A.K.; Moren, B.; Stenkula, K.G.; Magnusson, M.; Degerman, E. Insulin regulates Nedd4-2 via a PKB-dependent mechanism in HEI-OC1 auditory cells-crosstalks with sphingolipid and cAMP signaling. Acta Oto-Laryngol. 2022, 142, 6–12. [Google Scholar] [CrossRef]
  217. Wang, H.; Wang, A.X.; Aylor, K.; Barrett, E.J. Nitric oxide directly promotes vascular endothelial insulin transport. Diabetes 2013, 62, 4030–4042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Wu, G.; Meininger, C.J. Nitric oxide and vascular insulin resistance. Biofactors 2009, 35, 21–27. [Google Scholar] [CrossRef] [PubMed]
  219. Perreault, M.; Marette, A. Targeted disruption of inducible nitric oxide synthase protects against obesity-linked insulin resistance in muscle. Nat. Med. 2001, 7, 1138–1143. [Google Scholar] [CrossRef] [PubMed]
  220. Duplain, H.; Burcelin, R.; Sartori, C.; Cook, S.; Egli, M.; Lepori, M.; Vollenweider, P.; Pedrazzini, T.; Nicod, P.; Thorens, B.; et al. Insulin resistance, hyperlipidemia, and hypertension in mice lacking endothelial nitric oxide synthase. Circulation 2001, 104, 342–345. [Google Scholar] [CrossRef] [PubMed]
  221. Ruan, R.S. Possible roles of nitric oxide in the physiology and pathophysiology of the mammalian cochlea. Ann. N. Y. Acad. Sci. 2002, 962, 260–274. [Google Scholar] [CrossRef]
  222. Nakashima, T.; Naganawa, S.; Sone, M.; Tominaga, M.; Hayashi, H.; Yamamoto, H.; Liu, X.; Nuttall, A.L. Disorders of cochlear blood flow. Brain Res. Rev. 2003, 43, 17–28. [Google Scholar] [CrossRef]
  223. Xi, Y.; Yan, J.; Li, M.; Ying, S.; Shi, Z. Gut microbiota dysbiosis increases the risk of visceral gout in goslings through translocation of gut-derived lipopolysaccharide. Poult. Sci. 2019, 98, 5361–5373. [Google Scholar] [CrossRef]
  224. De Waal, G.M.; de Villiers, W.J.; Forgan, T.; Roberts, T.; Pretorius, E. Colorectal cancer is associated with increased circulating lipopolysaccharide, inflammation and hypercoagulability. Sci. Rep. 2020, 10, 8777. [Google Scholar] [CrossRef]
  225. Grosicki, G.J.; Fielding, R.A.; Lustgarten, M.S. Gut microbiota contribute to age-related changes in skeletal muscle size, composition, and function: Biological basis for a gut-muscle axis. Calcif. Tissue Int. 2018, 102, 433–442. [Google Scholar] [CrossRef] [Green Version]
  226. Utzschneider, K.M.; Kratz, M.; Damman, C.J.; Hullar, M. Mechanisms linking the gut microbiome and glucose metabolism. J. Clin. Endocrinol. Metab. 2016, 101, 1445–1454. [Google Scholar] [CrossRef] [Green Version]
  227. Violi, F.; Nocella, C.; Carnevale, R. Gut microbiota and myocardial infarction. Eur. Heart J. 2020, 41, 2221–2222. [Google Scholar] [CrossRef] [PubMed]
  228. Salguero, M.V.; Al-Obaide, M.A.; Singh, R.; Siepmann, T.; Vasylyeva, T.L. Dysbiosis of gram-negative gut microbiota and the associated serum lipopolysaccharide exacerbates inflammation in type 2 diabetic patients with chronic kidney disease. Exp. Ther. Med. 2019, 18, 3461–3469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  229. Andreakos, E.; Sacre, S.M.; Smith, C.; Lundberg, A.; Kiriakidis, S.; Stonehouse, T.; Monaco, C.; Feldmann, M.; Foxwell, B.M. Distinct pathways of LPS-induced NF-kappa B activation and cytokine production in human myeloid and nonmyeloid cells defined by selective utilization of MyD88 and Mal/TIRAP. Blood 2004, 103, 2229–2237. [Google Scholar] [CrossRef] [PubMed]
  230. Kuper, C.; Beck, F.X.; Neuhofer, W. Toll-like receptor 4 activates NF-kappaB and MAP kinase pathways to regulate expression of proinflammatory COX-2 in renal medullary collecting duct cells. Am. J. Physiol. Ren. Physiol. 2012, 302, F38–F46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  231. Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.; Chabo, C.; et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 1761–1772. [Google Scholar] [CrossRef] [Green Version]
  232. Sugita, H.; Kaneki, M.; Tokunaga, E.; Sugita, M.; Koike, C.; Yasuhara, S.; Tompkins, R.G.; Martyn, J.A. Inducible nitric oxide synthase plays a role in LPS-induced hyperglycemia and insulin resistance. Am. J. Physiol. Endocrinol. Metab. 2002, 282, E386–E394. [Google Scholar] [CrossRef] [Green Version]
  233. Baker, P.J.; Boucher, D.; Bierschenk, D.; Tebartz, C.; Whitney, P.G.; D’Silva, D.B.; Tanzer, M.C.; Monteleone, M.; Robertson, A.A.; Cooper, M.A. NLRP3 inflammasome activation downstream of cytoplasmic LPS recognition by both caspase-4 and caspase-5. Eur. J. Immunol. 2015, 45, 2918–2926. [Google Scholar] [CrossRef]
  234. Wang, S.; Yuan, Y.H.; Chen, N.H.; Wang, H.B. The mechanisms of NLRP3 inflammasome/pyroptosis activation and their role in Parkinson’s disease. Int. Immunopharmacol. 2019, 67, 458–464. [Google Scholar] [CrossRef]
  235. Jiang, Y.; Zhang, J.; Rao, Y.; Chen, J.; Chen, K.; Tang, Y. Lipopolysaccharide disrupts the cochlear blood-labyrinth barrier by activating perivascular resident macrophages and up-regulating MMP-9. Int. J. Pediatr. Otorhinolaryngol. 2019, 127, 109656. [Google Scholar] [CrossRef]
  236. Hirose, K.; Li, S.-Z. The role of monocytes and macrophages in the dynamic permeability of the blood-perilymph barrier. Hear. Res. 2019, 374, 49–57. [Google Scholar] [CrossRef]
Figure 1. The proposed relationship between ageing, gut dysbiosis, and hearing loss. Numbers in arrows indicate a pathway described below. Abbreviations: LPS—lipopolysaccharides; ROS—reactive oxygen species; NF-κB—nuclear factor kappa-light-chain-enhancer of activated B cells; TLR—toll-like receptor; NLRP3—Nod-Like Receptor Pyrin domain-containing protein 3. Ageing is associated with developing gut dysbiosis, and it was proposed that dysbiosis is an underlying cause of age-related morbidities (1) [165,178]. As humans age, inflammaging results from the activation of NF-κB signalling pathways (2) [179]. In addition, the leaky intestinal barrier causes the release of bacterial metabolites into the bloodstream, systemic inflammation, activation of NF-κB (4), and ROS overproduction (32) [180,181,182]. Activation of NF-κB may result in reduced insulin signalling (26) [183,184], which can lead to sterile cochlear inflammation (25) [185,186], and priming of NLRP3 (15) [187,188,189]. Ageing (8) and gut dysbiosis (3) are both associated with dyslipidemia [22,170,190,191,192]. Dyslipidemia can lead to the narrowing of cochlear blood vessels (13) [193,194,195] and lipid raft formation (9) [196,197,198]. Lipid rafts lead to the accumulation of ROS (10) [197] and oxidative stress (OS) (11) [197,199]. OS can activate the NF-κB pathway (24) [200,201] and cause cochlear damage and hearing loss (12) [202,203,204,205,206,207,208]. The cholesterol and lipids obstructing cochlear vessels lead to cochlear ischemia (14), which also contributes to cochlear damage (15) [209,210,211,212,213,214]. Insulin resistance can lead to an electrochemical imbalance in endolymph by decreasing the expression of the sodium-potassium pump NKCC1, reducing the endocochlear potential [215,216,217]. Insulin resistance also decreases nitric oxide production (27) [218,219] resulting in impaired blood flow regulation (28) [188,220], cochlear ischemia (29) and eventually hearing loss (15) [221,222]. Gut dysbiosis can lead to increased levels of circulating lipopolysaccharide (LPS) (5) [223,224,225,226,227,228]. LPS is a bacterial metabolite that can activate NF-κB via toll-like receptors (TLRs) (7) [229,230]. LPS also affects insulin signalling (23) [231,232] and contributes to NLRP3 inflammasome activation [233]. After priming via the NF-κB signalling pathway (15), NLRP3 is activated via ROS (30), resulting in NLRP3 inflammasomes (17). NLRP3 inflammasomes can cause pyroptosis (19) [234], which also produces cochlear damage [32] (21). NLRP3 can also initiate the activation of cochlear macrophages (18) [32], which can increase the BLB permeability and induce sterile cochlear inflammation (20) [22,235,236], leading to hearing loss (22) [22,236].
Figure 1. The proposed relationship between ageing, gut dysbiosis, and hearing loss. Numbers in arrows indicate a pathway described below. Abbreviations: LPS—lipopolysaccharides; ROS—reactive oxygen species; NF-κB—nuclear factor kappa-light-chain-enhancer of activated B cells; TLR—toll-like receptor; NLRP3—Nod-Like Receptor Pyrin domain-containing protein 3. Ageing is associated with developing gut dysbiosis, and it was proposed that dysbiosis is an underlying cause of age-related morbidities (1) [165,178]. As humans age, inflammaging results from the activation of NF-κB signalling pathways (2) [179]. In addition, the leaky intestinal barrier causes the release of bacterial metabolites into the bloodstream, systemic inflammation, activation of NF-κB (4), and ROS overproduction (32) [180,181,182]. Activation of NF-κB may result in reduced insulin signalling (26) [183,184], which can lead to sterile cochlear inflammation (25) [185,186], and priming of NLRP3 (15) [187,188,189]. Ageing (8) and gut dysbiosis (3) are both associated with dyslipidemia [22,170,190,191,192]. Dyslipidemia can lead to the narrowing of cochlear blood vessels (13) [193,194,195] and lipid raft formation (9) [196,197,198]. Lipid rafts lead to the accumulation of ROS (10) [197] and oxidative stress (OS) (11) [197,199]. OS can activate the NF-κB pathway (24) [200,201] and cause cochlear damage and hearing loss (12) [202,203,204,205,206,207,208]. The cholesterol and lipids obstructing cochlear vessels lead to cochlear ischemia (14), which also contributes to cochlear damage (15) [209,210,211,212,213,214]. Insulin resistance can lead to an electrochemical imbalance in endolymph by decreasing the expression of the sodium-potassium pump NKCC1, reducing the endocochlear potential [215,216,217]. Insulin resistance also decreases nitric oxide production (27) [218,219] resulting in impaired blood flow regulation (28) [188,220], cochlear ischemia (29) and eventually hearing loss (15) [221,222]. Gut dysbiosis can lead to increased levels of circulating lipopolysaccharide (LPS) (5) [223,224,225,226,227,228]. LPS is a bacterial metabolite that can activate NF-κB via toll-like receptors (TLRs) (7) [229,230]. LPS also affects insulin signalling (23) [231,232] and contributes to NLRP3 inflammasome activation [233]. After priming via the NF-κB signalling pathway (15), NLRP3 is activated via ROS (30), resulting in NLRP3 inflammasomes (17). NLRP3 inflammasomes can cause pyroptosis (19) [234], which also produces cochlear damage [32] (21). NLRP3 can also initiate the activation of cochlear macrophages (18) [32], which can increase the BLB permeability and induce sterile cochlear inflammation (20) [22,235,236], leading to hearing loss (22) [22,236].
Ijms 23 07348 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kociszewska, D.; Vlajkovic, S. Age-Related Hearing Loss: The Link between Inflammaging, Immunosenescence, and Gut Dysbiosis. Int. J. Mol. Sci. 2022, 23, 7348. https://doi.org/10.3390/ijms23137348

AMA Style

Kociszewska D, Vlajkovic S. Age-Related Hearing Loss: The Link between Inflammaging, Immunosenescence, and Gut Dysbiosis. International Journal of Molecular Sciences. 2022; 23(13):7348. https://doi.org/10.3390/ijms23137348

Chicago/Turabian Style

Kociszewska, Dagmara, and Srdjan Vlajkovic. 2022. "Age-Related Hearing Loss: The Link between Inflammaging, Immunosenescence, and Gut Dysbiosis" International Journal of Molecular Sciences 23, no. 13: 7348. https://doi.org/10.3390/ijms23137348

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop