Noise Induced Epigenetic Effects

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Review Article

Noise Induced Epigenetic Effects: A Systematic Review


Veruscka Leso, Luca Fontana, Ferdinando Finiello, Luigi De Cicco, Maria Luigia Ercolano, Ivo Iavicoli
Downloaded from http://journals.lww.com/nohe by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AW

Section of Occupational Medicine, Department of Public Health, University of Naples Federico II, Naples, Italy

Abstract
nYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 06/07/2024

Background: Noise-induced hearing loss (NIHL) is one of the leading causes of acquired sensorineural hearing loss. However, molecular
mechanisms responsible for its pathogenesis remain to be elucidated. Epigenetic changes, i.e. DNA methylation, histone and microRNA
expression modifications may function as a link between noise exposure and hearing loss. Therefore, the aim of the present review was to
assess whether epigenetic alterations may serve as biomarkers of noise exposure or early effect. Materials and Methods: A systematic review
of studies available in Pubmed, Scopus, and ISI Web of Science databases was performed. Results: Noise exposure was able to induce
alterations in DNA methylation levels in workers and animal models, resulting in expression changes of genes related to hearing loss and also
to extra-auditory effects. Differently expressed microRNAs were determined in NIHL workers compared to noise-exposed subjects with
normal hearing, supporting their possible role as biomarkers of effect. Acoustic trauma affected histon acethylation and methylation levels in
animals, suggesting their influence in the pathogenesis of acute noise-induced damage and their role as targets for potential therapeutic
treatments. Conclusions: Although preliminary data suggest a relationship between noise and epigenetic effects, the limited number of
studies, their different methodologies and the lack of adequate characterization of acoustic insults prevent definite conclusions. In this context,
further research aimed to define the epigenetic impact of workplace noise exposure and the role of such alterations in predicting hearing loss
may be important for the adoption of correct risk assessment and management strategies in occupational settings.

Keywords: DNA methylation, histone modification, hearing loss, occupational exposure, microRNA

BACKGROUND The pathogenesis of NIHL relies on the complex interplay


between genetic and environmental factors.[9] Susceptibility
Noise-induced hearing loss (NIHL) represents one of the
to the noise effects shows a relevant interindividual
most common worldwide types of acquired sensorineural
difference, meaning that an exposure to equal noise
hearing loss besides the age-related form.[1,2] Among
intensity levels may result in greatly different hearing
workplace hazards, occupational noise is one of the most
impairments.[10-12] Long or repeated exposure to sounds at
common, affecting the health of exposed workers and
or >85 dB can cause hearing loss inducing the irreversible
potentially responsible for the occupational NIHL
death or apoptosis of the specialized sensory hair epithelium
(ONIHL), which is a work-related medical condition
of the basilar membrane of the organ of Corti in the cochlea,
characterised by a permanent sensorineural hearing loss
the inner ear part responsible for hearing.[13] Additionally,
due to an excessive exposure to high levels of noise in
acute acoustic trauma, intended as a short exposure to intense
workplace settings.[3,4]
The rapid increase in the number of people exposed to the
hazardous effects of noise in both industrialized and
developing countries, the 7% to 21% prevalence of Address for correspondence: Luca Fontana, Professor, Section of
Occupational Medicine, Department of Public Health, Section of Occupational
ONIHL among exposed workers,[5-7] together with the
Medicine, University of Naples Federico II, Via S. Pansini 5, 80131, Naples,
disabling consequences deriving from such disorder,[8] Italy.
makes NIHL, and particularly ONIHL, a major public and E-mail: luca.fontana@unina.it
occupational health problem.
Received: 27 March 2020 Revised: 1 July 2020
Accepted: 14 July 2020 Published: 31 December 2020

This is an open access journal, and articles are distributed under the terms of the
Access this article online Creative Commons Attribution-NonCommercial-ShareAlike 4.0 License, which allows
Quick Response Code: others to remix, tweak, and build upon the work non-commercially, as long as
Website: appropriate credit is given and the7 new creations are licensed under the identical
www.noiseandhealth.org terms.

For reprints contact: reprints@medknow.com

DOI: How to cite this article: Leso V, Fontana L, Finiello F, De Cicco L,


10.4103/nah.NAH_17_20 Luigia Ercolano M, Iavicoli I. Noise Induced Epigenetic Effects: A
Systematic Review. Noise Health 2020;22:77-89.

© 2020 Noise & Health | Published by Wolters Kluwer - Medknow 77


Leso, et al.: Epigenetic alterations as biomarkers of noise damage

impulse sounds of 100–150 dB, can cause hearing loss or titles and abstracts were independently reviewed by two of the
impairment too.[14] authors with the aim to select papers suitable for the review
purposes. The following inclusion criteria were adopted: all
Although numerous studies revealed that multiple factors are
types of human peer-reviewed research articles (i.e., cross-
responsible for the development of NIHL, including
sectional, cohort, case-control studies) published in English,
increased oxidative stress, reduced blood flow, disruption
and exploring the noise exposure-epigenetic alterations
of calcium homeostasis, and mechanical trauma, the
relationship both in humans and in experimental animals.
Downloaded from http://journals.lww.com/nohe by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AW

mechanisms by which the cochlea is damaged following


Exclusion criteria regarded reviews, case reports, conference
noise exposure are not entirely clear.[15-18] The lack of
papers, publications not focusing on the association between
comprehensive understanding of the causal molecular
epigenetic effects and noise exposure, as well as all the papers
mechanisms underlying noise-induced damage, both in
published in languages other than English. The preliminary
nYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 06/07/2024

acute and chronic conditions of exposure, is responsible


search retrieved 96, 147 and 102 references through PubMed,
for the lack of established therapeutic strategies for
Scopus and ISI Web of Science databases, respectively, for a
prevention or amelioration of such disabling disorder and
total of 345 articles [Figure 1]. After duplicate removal, 177
for the poor prognosis of the NIHL.[18] Therefore, due to the
articles remained. Among those, studies that did not meet the
disease burden and disability associated with hearing
inclusion criteria were excluded according to the following
impairment, NIHL deserves a high priority of research to
reasons: 161 because considered out of the topic from the title
develop innovative screening and therapies.
and abstract analysis; 11 as review articles, 1 as a conference
In this scenario, epigenetics, referred to as changes that alter paper and 1 as published in a language other than English.
gene transcription in the absence of direct alterations to the Indeed, three publications could be identified in this
genetic sequence, may function as a link between noise preliminary phase. Therefore, we extended our search
exposure and hearing loss.[19] Alterations in DNA including the following terms “hearing or noise exposure”,
methylation, post-translational histone modifications, and individually combined with “DNA methylation”,
changes in microRNAs expression, as the most used “microRNA” and “histone”, that allowed to include six
mechanisms able to initiate and sustain epigenetic additional papers. All full texts of the articles considered
information, have been tought to serve as stable non- valuable for the aim of our review were obtained and a critical
invasive biomarkers of noise induced alterations.[20] evaluation was performed. Overall, our search retrieved a
Therefore, aim of the present review was to assess whether total of nine publications for review.
epigenetic changes may indicate noise exposure or
occurrence of NIHL. This was finalized to gain insight
into possible molecular mechanisms underlying noise EPIGENETIC EFFECTS INDUCED BY NOISE
exposure and to define possible early biological alterations EXPOSURE
that may act as suitable biomarkers of exposure, effect or Noise exposure was reported to induce epigenetic effects,
susceptibility in exposed workers. Overall, from an such as DNA methylation changes,[22,23] histone
occupational health perspective, this review may reveal a modifications,[24-27] as well as alterations in non-coding
useful means to understand the relationship between noise microRNAs (miRNAs),[28-30] both in occupational and
exposure, auditory and extra-auditory related injuries. This experimental settings [Tables 1–3]. The following
may be important to extrapolate information to lead future paragraphs will attempt to summarize such effects with the
reserch focused at the development of effective biological aim to point out currently available data and critical issues
monitoring strategies, diagnostic possibilities, and potential that require further research in order also to conceive
treatments as well as to point out biological mechanisms and epigenetic contribution to risk assessment and management
predictors of disease that may guide the adoption of the most strategies in noise exposure as well as NIHL.
effective preventive measures for exposed workers.

DNA methylation
PROCEDURE DNA methylation is recognized as an essential epigenetic
The Preferred Reporting Items for Systematic Reviews and mark that regulates chromatin structure and gene expression
Meta-Analyses Statement (PRISMA) criteria were followed throughout the genome.[31] Understanding how noise
to perform a systematic literature search.[21] Studies exposures may affect DNA methylation patterns may help
addressing epigenetic effects induced by noise exposure, to elucidate the relationship between noise and its possible
published until 15 March 2020, were identified by research auditory and extra-auditory effects [Table 1].[23] In this
on three principal scientific databases: PubMed, Scopus and context, Wang et al.[22] investigated DNA methylation and
ISIWeb of Science. A preliminary search was carried out for gene expression abnormalities induced by blast overpressure
the terms “hearing or noise exposure” to assess the context of exposures in male “breachers”, military and law enforcement
exposure and “epigenetic” as the investigation outcome, professionals subacutely and chronically exposed to repeated
which were combined with the Boolean operator “AND”. blasts as part of their job duties. Epigenetic effects were
The computerized search allowed to retrieve articles whose evaluated with respect to blast subacute exposures, intended

78 Noise & Health ¦ Volume 22 ¦ Issue 107 ¦ October-December 2020


Leso, et al.: Epigenetic alterations as biomarkers of noise damage
Downloaded from http://journals.lww.com/nohe by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AW
nYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 06/07/2024

Figure 1: Flow diagram of literature search

as controlled, low-level blast exposures experienced during a tinnitus. The authors identified genes with differential
10-day explosive breaching course, and to the number of DNA methylation changes associated with symptoms of
cumulative blast exposure events (< or >40) during the tinnitus in the high vs. low career breaching groups.
career in the military service. No different methylated Differently hypermethylated regions involved the
regions could be identified comparing pre- and post-blast potassium voltage-gated channel, Isk-related family,
exposure blood DNA methylation patterns in subjects member 1 (KCNE1), and the cytochrome P450 family 2,
attending the above-mentioned training courses. When subfamily E, member 1 (CYP2E1) genes, whose
DNA methylation analysis was conducted considering transcriptional expression resulted significantly reduced.
cumulative exposure, 10 differently hyper-methylated Interestingly, both KCNE1 and CYP2E1 genes have been
regions resulted positively associated to the highest reported to be implicated in noise-related hearing loss in
lifetime exposure with corresponding gene expression human genetics and animal investigations, respectively.[35-40]
changes. Interestingly, hypermethylation localized in the
promoter region of the antisense transcript within the DNA methylation changes were reported by Guo et al.[23]
paired box gene 8 (PAX8), resulted responsible for the when exploring the effects of environmental noise exposure
control of the expression of thyroid-specific genes in vivo. They analysed the global DNA methylation levels in
involved in the gland function as well as in sleep duration LINE-1 untranslated regions and the methylation of five
suggesting the possible epigenetic control of potential extra- selected genes expressed in different regions of the brain,
auditory effects of noise.[32-34] DNA methylation and gene i.e. the frontal lobe, hippocampus, inferior colliculus, and
expression changes were also investigated in relation to the medulla oblongata, of male Wistar rats exposed to moderate
more frequently self-reported symptoms, particularly intensity noise for 3 or 21 days, as short and long-term

Noise & Health ¦ Volume 22 ¦ Issue 107 ¦ October-December 2020 79


Leso, et al.: Epigenetic alterations as biomarkers of noise damage

Table 1: Studies addressing DNA-methylation changes induced by noise exposure


DNA methylation
Study Experimental settings Study design Results References
location
HUMAN INVESTIGATION
Downloaded from http://journals.lww.com/nohe by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AW

USA Participants at the US Army explosive Noise exposure ✓ Differently DNA methylated regions Wang
entry training sites (special operations (n. 10) with corresponding gene et al.[22]
and combat engineering 10-day course) expression changes (with the expected
(n. 34 healthy males; mean age: 30.79 anticorrelation between DNA
±4.57 years) methylation and gene expression) were
nYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 06/07/2024

demonstrated in relation to chronic


cumulative career blast exposures.
Subjects were divided in two groups ✓ A gain of methylation with a
according to the number of blast corresponding loss of gene expression
exposures reported (low: <40 vs high: was found in the promoter of the
>40) during their career in the military PAX8 gene (involved in thyroid
service. function control) in high cumulative
blast exposure group.
Exposure chronology during the 10-day ✓ No DNA methylation differences
training course: day 2: shotgun were detected between pre and post-
training; day 7: ∼12 psi blast exposure; acute blast exposures.
day 9: ∼4 psi blast exposure.
Biological analysis Blood sample for ✓ DNA methylation analyses
epigenetics and transcriptional studies conducted in conjunction with reported
were collected at the start (baseline) symptoms of tinnitus in the low versus
and at the end of the training course high blast incidents groups identified
(pre- vs post-blast exposure). differently DNA methylated regions in
KCNE1 and CYP2E1 genes (with loss
of gene expression) which have been
implicated in noise-related hearing loss.
IN VIVO STUDIES
China Male Wistar rats (n. 32) randomly Noise exposure ✓ Following short term noise exposure, Guo
divided into four groups. Comt gene DNA methylation was et al.[23]
significantly increased in medulla
oblongata. No additional significant
changes could be detected in other
genes and brain regions.
Rats were exposed to moderate ✓ Following long-term noise exposure,
intensity noise (70–75 dB with Comt methylation in the inferior
20–4000 Hz) at night (7.00 PM − 7.00 colliculus was significantly increased.
AM) for 3 days (short-term exposure The Mc2r gene displayed significantly
group:n. 8 rats) or for 21 days (long- decreased methylation in hippocampus.
term exposure group: n. 8 rats). Two Global DNA methylation significantly
control groups were exposed to 45 dB increased in the medulla oblongata.
sound intensity during daytime for 3
(n. 8 rats) and 21 (n. 8 rats) days.
Biological analysis DNA methylation ✓ No observed differences in DNA
in the Bdnf, Comt, Crhr1, Mc2r and methylation between short and long-
Snca genes was assessed in brain term exposure.
tissues: inferior colliculus, frontal lobe,
hippocampus, medulla oblongata.
Global DNA methylation was assessed
through LINE-1 (LINE-1 UTR and
ORF).
Bdnf, brain derived neurotrophic factor; Comt, catechol-O-methyltransferase; Crhr1, corticotropin-releasing hormone receptor 1; CYP2E1, cytochrome P450
family 2 subfamily E member 1; KCNE1, Isk-related family member 1; Mc2r, melanocortin 2 receptor; ORF, open reading frame; PAX8, pairedbox gene 8;
psi, pounds per square inch; Snca, synuclein Alpha; UTR, untranslated region.

exposures, respectively. Following short term noise exposure, colliculus, while the melanocortin 2 receptor (Mc2r) gene
only the DNA methylation of the catechol-O- demonstrated a significantly decreased methylation in
methyltransferase (Comt) gene was significantly increased hippocampus. As far as the Comt gene is concerned, it
in medulla oblongata. After long-term exposure, Comt codes for an enzyme involved in the dopaminergic circuit,
methylation was significantly increased in the inferior in turn linked to cognitiveness, working memory and

80 Noise & Health ¦ Volume 22 ¦ Issue 107 ¦ October-December 2020


Leso, et al.: Epigenetic alterations as biomarkers of noise damage

Table 2: Studies addressing micro-RNA expression changes induced by noise exposure


Micro-RNAs
Study Experimental settings Study design Results References
location
HUMAN INVESTIGATIONS
Downloaded from http://journals.lww.com/nohe by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AW

China NIHL male patients (n. 23); Noise- Biological analysis ✓ Compared with the noise-exposed Ding et al.
[28]
exposed male individuals with normal controls, 73 miRNAs demonstrated at
hearing (n. 23); Non-noise exposed least 1.5-fold differential expression
individuals with normal hearing (n. 23) levels in the NIHL patients, of which
Mean age of NIHL and noise-exposed 39 miRNAs were upregulated and 34
nYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 06/07/2024

subjects: 44.1 ± 5.1 years; Mean age of miRNAs were downregulated.


controls: 44.13±5.0 yearsNoise exposed
subjects were employed in textile
industry; non-noise exposed subjects
were teachers in elementary and
middle school.
MiRNAs were extracted and purified ✓ Five miRNAs (let-7d-5p, miR-16-
from plasma samples of enrolled 5p, miR-24-3p, miR-185-5p, miR-
subjects (20 NIHL and noise-exposed 451a) were significantly upregulated,
subjects for the preliminary validation; while miR-1915-3p was significantly
all 46 NIHL and noise-exposed + 23 downregulated in NIHL patients
controls in the expanded validation). compared to the noise-exposed
individuals with normal hearing.
RT-qPCR was performed to validate ✓ RT-qPCR demonstrated that
the expression levels of the candidate compared with the non-exposures, the
miRNAs screened from the microarray plasma levels of miR-24, miR-185-5p
assay. Noise exposure Occupational and miR-451a were all significantly
noise exposure meant levels of noise downregulated in the exposures while
exposure (Lex) are at least 85 dB (A) compared with the noise-exposed
for a nominal 8-h working day. NIHL controls, miR-185-5p and miR-451a
patients were exposed for 20.6 ± 6.5 were slightly upregulated in the NIHL
years; Noise exposed subjects with patients.
normal hearing were exposed for
21.3 ± 6.5 years. Acute injury: acoustic
trauma or extremely intense noise
(>130 dB) was excluded.
China Male workers (n.10) with ONIHL and Biological analysis ✓ Three miRNAs upregulated (hsa- Li et al.[29]
noise-exposed male individuals with miR-3162-5p, hsa-miR-4484, hsa-miR-
normal hearing as controls (n. 10)Age 1229-5p) and 1 downregulated (hsa-
range: 30–45 yearsThe industries of miR-4652-3p) in ONIHL subjects
ONIHL workers included machinery compared to controls.
(n. 8), food (n. 1) and chemical
industry (n. 1). The industries of
control subjects included machinery (n.
4), manufacturing (n. 4), energy (n. 1),
and printing (n. 1).
MiRNAs were extracted and purified ✓ Significantly increased serum levels
from blood serum of enrolled subjects. of miR-1229-5p in ONHIL group as
Microarray hybridization was compared to controls were confirmed
conducted for profiling differentially by RT-qPCR.
expressed miRNAs between the two
groups. RT-qPCR was performed to
validate the expression levels of the
candidate miRNAs screened from the
microarray assay. Noise exposure
Enrolled subjects had more than 1-year
work experience in a noisy
environment (no other details
provided).
IN VIVO STUDIES
USA Sprague Dawley male and female rats Biological analysis ✓ ABR thresholds were measured Patel et al.
(2013) with normal hearing sensitivity − were before and at 2 h and 1 d post-noise [30]

exposed to acute noise (2 h exposure)


(Continued )

Noise & Health ¦ Volume 22 ¦ Issue 107 ¦ October-December 2020 81


Leso, et al.: Epigenetic alterations as biomarkers of noise damage

Table 2 (Continued)
Micro-RNAs
Study Experimental settings Study design Results References
location
and sacrificed 2 h (n. 4) or 1 day post- exposure to determine the functional
noise exposure (n. 8).Control animals status of the cochlea.
Downloaded from http://journals.lww.com/nohe by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AW

(n. 12) received identical treatment


without noise.
MiRNA and mRNA gene array ✓ Relative to pre-noise thresholds
expression analysis was performed on ABR threshold shifts of 47.12±4.3 dB
the cochlear sensory epithelia sampled and 32.3±6.2 dB (mean ±SD) at 2 h and
nYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 06/07/2024

from euthanized animals. 1 d post-noise exposure were reported,


respectively.
Noise exposure ✓ At 2 h post-noise exposure, only 1
gene, miR-331-5p, was significantly
upregulated, but returned to a baseline
level at 1 d post-noise exposure.
Broadband continuous noise (1–7 kHz) ✓ At 1 d post-noise exposure, 20
at 120 dB SPL for 2 h. miRNAs (miRs 10a, 107, 124, 130b,
146b, 183, 186, 190b, 194, 200c, 30d,
30e, 325, 333, 339-3p, 381, 429, 532-
3p, 674 and 99b) were significantly
downregulated.
✓ At 1 d post-noise exposure Taok1
mRNA, a predicted target of miR-183,
was significantly upregulated (2.3 fold
compared to normal) as demonstrated
by the RT-qPCR.
ABR, auditory brainstem response; MiRNAs, microRNAs; NIHL, noise-induced hearing loss; ONIHL, occupational noise-induced hearing loss; RT-qPCR,
Real Time quantitative Polymerase Chain Reaction; SD, standard deviation; SPL, sound pressure level.

depression.[41] The hypermethylation of this gene in the potential effects on miRNAs expression induced by noise
inferior colliculus following a long-term noise exposure, exposure in humans [28,29] and animal models.[30]
which consequently leads to a reduced expression of the
A case-control study investigated the plasma miRNA profile
enzyme encoded by the same gene, could suggest a role of
of NIHL male patients, noise-exposed male individuals with
chronic noise exposure on certain brain functions, including
normal hearing, both employed in a textile industry, and of an
memory and cognition, and in the increased risk of
unexposed control group with normal hearing.[28] The authors
depression. Similarly, as regards Mc2r, receptor of the
found that the NIHL group showed 73 miRNAs with at least
adrenocorticotropic hormone (ACTH) involved in the
1.5-fold differential expression levels compared to noise-
response to stressors,[42] the finding of hypomethylation of
exposed controls at the microarray analysis, suggesting a
this gene in the hippocampus after chronic noise exposure,
possible specific mRNA expression pattern indicative of a
may be associated with memory deficit and cognition
hearing loss occurrence. Among the miRNAs differently
impairment. Global DNA methylation in LINE-1
expressed in NIHL patients and noise-exposed controls,
significantly increased in the medulla oblongata, but not in
miR-16-5p, miR-24-3p, miR-185-5p, miR-451a, all
the other brain regions examined at the same time point.
molecules implicated in the regulation of oxidative stress
However, the differences detected in the DNA methylation
responses, demonstrated a significant up-regulation in the
following long-term noise exposure, with respect to short-
first group. However, an additional validation analysis
term one, have not been fully explained and no relation could
demonstrated that miR-24-3p, miR-185-5p, miR-451a were
be detected for specific genes, brain regions and functions.
all significantly downregulated in both noise-exposed groups
compared to unexposed controls. Interestingly, the findings
Micro RNAs of a downregulation of key miRNAs in noise exposed vs.
MicroRNAs, small 20–22 nucleotide molecules, represent a unexposed subjects are in line with previous miRNA
new class of noncoding RNA genes able to regulate cellular expression research of oxidative stress-related changes in
functions by modulating mRNA expression levels.[13] cultured cells derived from the organ of Corti.[48]
Investigations on miRNA functions in the auditory system Excessive noise may generate, in the cochlea, an over-
have been mainly focused on their roles in inner ear production of reactive oxygen species which may be
development [Table 2].[43-47] However, the role of responsible for changes in the expression of miRNAs
miRNAs in noise-induced cochlear pathogenesis is yet to involved in the oxidative stress response. Such miRNA
be established, and only preliminary data are available on the expression pattern may therefore be involved in NIHL

82 Noise & Health ¦ Volume 22 ¦ Issue 107 ¦ October-December 2020


Leso, et al.: Epigenetic alterations as biomarkers of noise damage

Table 3: Studies addressing histone modifications induced by noise exposure


HISTONE MODIFICATIONS
Study Experimental settings Study design Results References
location
IN VIVO STUDIES
Downloaded from http://journals.lww.com/nohe by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AW

USA Male CBA/J mice assigned to 3 Noise exposure ✓ Noise exposure resulted in PTS at 4, Wen
experimental groups (n. 20 each 8, 12, 24, 32 and 48 kHz, 2 weeks et al.[24]
group): the control group; the DMSO following exposure to noise. Compared
group, that was exposed to noise and with the DMSO-injected group, pre-
received an intraperitoneal injection of treatment with the HDAC inhibitor,
nYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 06/07/2024

DMSO (10%) 3 days before the SAHA, significantly reduced PTS.


exposure; the SAHA group, that was
exposed to noise and received an
intraperitoneal injection of SAHA
(25 mg/kg) 3 days before noise
exposure.
Mice were exposed to broadband noise ✓ The expression of HDAC1 (2-fold)
with a frequency spectrum from 2 to and HDAC4 (3 fold) increased 1 h
20 kHz for 2 h/day for 2 weeks at following noise exposure compared to
110 dB SPL to induce a PTS with loss controls. The H3-AcK9 levels
of cochlear OHCs. Control animals decreased 1 h following noise exposure.
were not exposed to noise.
Biological analysis The expression ✓ The number of OHCs loss decreased
levels of H3K9ac and HDAC1 and following SAHA pre-treatment
HDAC4 were assessed by western blot compared to the DMSO pre-treatment.
analysis on cochlear tissue SAHA pre-treatment doubled the
homogenates 1 h after exposure. number of survival OHCs and
attenuated cilia damage.
USA Male CBA/J mice (max n.7 per group) Noise exposure ✓ Immunolabeling for H3K9ac Chen
were exposed to noise and cochlear decreased in the nuclei of OHCs (60%) et al.[25]
tissues were analysed 1 h after the and strial marginal cells of the cochlear
exposure completion basal turn 1 h after noise exposure
compared to control mice.
Control group: mice not exposed to Mice were exposed to broadband noise ✓ Noise exposure increased HDAC1,
noise with a frequency spectrum from 2 to 2, and 3 in cochlear tissues (organ of
20 kHz for 2 h at 98 dB SPL to induce Corti, spiral ganglion cells and stria
a PTS with loss of OHCs. vascularis) 1 h after noise exposure
compared to control mice.
Biological analysis ✓ Blockade of HDAC1, 2, or 3 alone
in cochlear tissues resulted not
sufficient to reduce PTS.
The expression levels of H3K9ac was ✓ Treatment with SAHA significantly
assessed by immunohistochemistry on decreased noise-induced OHCs loss
cryosection 1 h after the exposure. The (40% at 4 mm, 50% at 4.5 mm, and
levels of HDAC1, 2, and 3 using 20% at 5 mm), and significantly
paraffin-embedded cochlear sections attenuated PTS by about 20 dB at
from the mid-modiolar region were 16 kHz compared to vehicle-treated
assessed 1 h after the exposure. Two mice. After 1 h SAHA treatment there
week following noise exposure, was an enrichment of H3K9ac at the
treatment with SAHA (50 mg/kg nuclear periphery.
intraperitoneal injection) was applied to
the animals and noise-induced OHCs
loss, PTS and H3K0ac distribution in
the nuclei was assessed.
China Male albino guinea pigs (n. 120) Noise exposure ✓ Noise exposure induced PTS in both Yang
divided into 3 groups: control group the noise only group and the noise with et al.[26]
(no noise exposure, n. 40); noise only SB group. However, the ABR
group (n. 40); noise + SB threshold shifts in this latter group
(intraperitoneal injection at a dose of were significantly lower than those in
600 mg/kg once per day on the 3 days the noise only group.
before and after noise exposure, as
well as 30 min before and 2 h after the
noise exposure) group (n. 40).
(Continued )

Noise & Health ¦ Volume 22 ¦ Issue 107 ¦ October-December 2020 83


Leso, et al.: Epigenetic alterations as biomarkers of noise damage

Table 3 (Continued)
HISTONE MODIFICATIONS
Study Experimental settings Study design Results References
location
In the noise-exposed group: 10 animals Animals were exposed to 1/3-octave- ✓ Noise exposure significantly
underwent hearing test at 1 d before wide narrowband noise, centered at decreased H3? AcK9 expression, and
Downloaded from http://journals.lww.com/nohe by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AW

and 14 d after exposure and then were 4kHz at 122 dB SPL for 3 h. increased HDAC1 expression, in the
euthanized; 30 animals were nuclei of OHCs, IHCs, and Hensen’s
euthanized at 2 h after noise exposure cells. SB treatment partially reversed
these changes.
Biological analysis Cochlear tissues ✓ The 3-NT was significantly
nYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 06/07/2024

were collected for immunofluorescence increased in OHCs in the noise only


staining to assess H3AcK9, HDAC1 group compared to the control group.
and 3NT levels. Homogenized cochlear SB treatment significantly reduced the
tissues were used for protein total noise-induced increase of 3-NT
extraction and western blot analysis
USA Male CBA/J mice (max n.6 per group) Noise exposure ✓ Immunoreactivity of G9a protein Xiong et al.
[27]
were exposed to noise and cochlear increased in the nuclei of OHCs, IHCs,
tissues were analysed 1 h after the supporting cells of organ of Corti, the
exposure completion nuclei of SGNs and the nuclei of
marginal cells in lateral wall tissues in
the basal turn of the cochlea
Control group: mice not exposed to Mice were exposed to broadband noise ✓ Noise exposure increased
noise with a frequency spectrum from 2 to immunolabelling for H3K9me2 levels
20 kHz for 2 h at 101 dB SPL to induce in nuclei of OHCs, strial marginal
a PTS with loss of cochlear OHCs. cells, and SGNs from the basal turn of
the cochlea.
Biological analysis ✓ Noise exposure reduced the
expression of KCNQ4 in OHCs of the
basal turn of the cochlea (75%).
The levels of the lysine ✓ Treatment with BIX 01294 (a
dimethyltransferase G9a in cochlear specific inhibitor of G9a prevented the
cells were assessed by noise-induced decrease of KCNQ4
immunohistochemistry. Since a immunolabelling in OHCs, the loss of
primary function of G9a is to OHCs and reduces auditory threshold
demethylate lysine 9 of histone 3, shifts.
levels of H3K9me2 in the cochlea 1 h
after completion of noise exposure was
assessed through immunolabelling.
Expression levels of potassium channel
genes KCNQ4 was assessed 1 h after
noise exposure completion.
3NT, 3-nitrotyrosine; DMSO, Dimethyl Sulfoxide; H3K9me2, histone H3 lysine 9 dimethylation; HDAC, histone deacetylases; IHCs, inner hair cells;
KCNQ4, potassium voltage-gated channel subfamily Q member 4; OHCs, outer hair cells; PTS, permanent threshold shift; SAHA, Suberoylanilide
hydroxamic acid; SB, sodium butyrate; SGNs, spiral ganglion neurons; SPL, sound pressure level.

pathogenesis although the causal association and their role as 5p, hsa-miR-4484, hsa-miR- 1229-5p), and four were
possible early biomarkers of noise effects need further downregulated (hsa-miR-6752-3p, hsa-miR-6824-3p, hsa-
investigation. Importantly, the same additional verification miR-4769-3p, hsa-miR-4652-3p) in ONIHL subjects. The
confirmed that miR-185-5p and miR-451a were significantly upregulation of the above-mentioned miRNAs was
upregulated in the NIHL group compared to the noise- confirmed also using the real time quantitative polymerase
exposed subjects as previously demonstrated. This may chain reaction (RT-qPCR) as an additional analysis.
support the potential role of these two miRNAs as possible However, this technique found a significant increase of
biomarkers of noise exposure and hearing loss damage. serum expression only for miR-1229-5p in ONIHL group
suggesting that such molecule could be implicated in hearing
Comparably, a different miRNA expression was detected in
loss pathogenesis. The analyses of the predicted target genes
ONIHL workers compared to noise-exposed individuals with
of hsa-miR-1229-5p demonstrated that they were involved in
normal hearing enrolled as controls. In fact, seven
a great number of biological processes and molecular
differentially expressed miRNAs were determined between
functions, e.g. ion, nucleotide and ATP binding,
the two groups at the microarray analysis, with a >1.5 fold
transcription regulator and protein kinase activities. Among
changed expression: three were upregulated (hsa-miR-3162-
those involved genes, the mitogen-activated protein kinase

84 Noise & Health ¦ Volume 22 ¦ Issue 107 ¦ October-December 2020


Leso, et al.: Epigenetic alterations as biomarkers of noise damage

(MAPK) signaling pathway has been determined, reported to significant increase in the expression of HDAC1 and
function as a regulatory pathway in human genetic deafness HDAC4, and a decrease in the levels of acetyl-histone H3
and in the inner hair cell survival,[49] thus decreasing the (Lys9) (H3K9ac) protein compared to unexposed controls.
susceptibility to noise-induced hearing loss in animals.[50] Significant hearing impairment was evident 2 weeks
MiR-1229-5p may contribute to the pathogenesis of ONIHL following noise exposure in mice with a reduction in the
producing a post-transcriptional repression of MAPK1 thus number of outer hair cells (OHCs). Interestingly, a pre-
reducing a self-defensive mechanism against noise exposure. treatment with an HDAC inhibitor, the suberoylanilide
Downloaded from http://journals.lww.com/nohe by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AW

Overall, these findings may suggest serum miR-1229-5p level hydroxamic acid (SAHA) significantly prevented such
as a possible innovative biomarker for ONIHL prediction. molecular and morphological alterations. This suggests a
role for HDAC enzymes in NIHL. These results are in line
MiRNAs have been also implicated in responses to acoustic
with subsequent findings obtained by Chen et al.[25] who
nYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 06/07/2024

overstimulation. In this regard, Patel et al.[30] investigated the


determined increased levels of HDAC1, HDAC2 and
role of miRNAs in cochlear apoptotic pathogenesis after
HDAC3 prevalently in the nuclei of cochlear cells in
acoustic trauma in exposed Sprague Dawley rats. A
CBA/J mice following a traumatic noise exposure. As
significant auditory brainstem response (ABR) threshold
previously demonstrated, the levels of H3K9ac resulted
shift and apoptotic activity, characterized by an increased
significantly decreased in the nuclei of OHCs and
percentage of cells with condensated nuclei and increased
marginal cells of the stria vascularis, suggesting that
caspase-3 activity were demonstrated after the exposure to
histone H3 tail epigenetic modification could be involved
intense noise. Gene expression analysis of noise-traumatized
in noise-induced OHCs loss and NIHL. Inhibition of HDAC1,
cochleae revealed time-dependent transcriptional changes in
HDAC2, or HDAC3 with small-interfering RNAs decreased
the expression of miRNAs. Only miR-331-5p was
the expression of the target HDAC in OHCs, but did not
significantly upregulated at 2 h post-noise exposure, while
ameliorate the permanent threshold shift (PTS) induced by
returned to a baseline level at 1 d post-noise exposure. At this
the noise exposure. Conversely, the SAHA showed beneficial
time point, 20 miRNAs were significantly down-regulated
effects on OHCs loss, and reduced PTS. Comparably, Yang
with a fold change equal to or greater than 2.5, an alteration
et al.[26] confirmed such previous results demonstrating that
that was not evident at 2 h post-noise exposure. This may
traumatic noise exposure significantly increased HDAC1
suggest that the noise-induced miRNA expression changes
expression in the nuclei of OHCs and inner hair cells
are time-specific and involve different sets of miRNAs at
(IHCs) and decreased H3K9ac expression in guinea pig
different time points. When target prediction analysis was
cochlear tissues. The administration of the HDAC
performed to verify the possible involvement of potential
inhibitor, sodium butyrate, partially recovered these effects
regulators of the degenerative process of the cochlea
limiting the effects on OHCs and the PTS. Additionally, the
following acoustic overstimulation, a significant
sodium butyrate treatment reduced the noise-induced
upregulation of Taok1 mRNA, a target of miRNA-183,
oxidative stress, decreasing the 3-nytrotyrosine
was demonstrated. In non-cochlear tissues, Taok1 has been
upregulation induced by the traumatic exposure suggesting
associated with activation of mitogen-activated protein kinase
an oto-protective role of the HDAC inhibitors by limiting the
pathway in response to stress and DNA damage[51-53] and
overproduction of reactive oxygen and nitrogen species.
apoptosis induction.[54] This may suggest its possible role in
These findings suggest that histone acetylation is involved
the regulation of cochlear response to acoustic trauma
in the pathogenesis of noise-induced OHCs death and hearing
through the regulation of apoptotic pathways.
loss. Although the exact molecular mechanisms underlying
the protective effects of targeting histone deacetylases are still
Histone modifications not understood, this treatment may characterize a strategy for
Post-translational modifications of histones have emerged as protection against NIHL.
key regulators of genomic activity without affecting DNA
Apart from modifications in histone acetylation, also histone
sequence.[27] Histone acetylation catalyzed by the activity of
methylation has been investigated for a possible function in
histone acetyltransferases (HATs), and deacetylation
hearing loss development.[60,61] A traumatic noise exposure,
catalyzed by histone deacetylases (HDACs) are responsible
able to affect OHCs and PTS in mice, resulted in the
for the activation and repression of gene transcription,
activation of the G9a, a major histone lysine
respectively, influencing the interactions between histones
methyltransferase responsible for histone H3 lysine 9
and DNA.[55,56] Therefore, an imbalance in histone
dimethylation (H3K9me2) in both OHCs and IHCs, some
acetylation may be implicated in a wide range of
spiral ganglion neurons (SGNs) as well as marginal cells 1
transcriptional dysfunction, gene silencing and
hour after exposure.[27] Interestingly, the inhibition of G9a by
neurodegenerative disorders.[57-59]
treatment with BIX 01294 or G9a siRNA reduced noise-
Preliminary evidence in animal models is available induced losses of OHCs and attenuated NIHL. These findings
concerning the role of HDACs in hearing impairment suggest that epigenetic modifications of H3K9me2 may play
[Table 3].[24-27] Wen et al.[24], 1 h following an acute a role in NIHL and may function as a pharmacological target
traumatic noise in a CBA/J mouse model, demonstrated a to prevent NIHL.

Noise & Health ¦ Volume 22 ¦ Issue 107 ¦ October-December 2020 85


Leso, et al.: Epigenetic alterations as biomarkers of noise damage

DISCUSSION AND CONCLUSION LINE-1, and decreased Mr2c methylation, compared to the
previous described subacute effects.[23]
This review represents the first attempt to comprehensively
assess the possible role of epigenetic alterations in Moreover, possible time-dependent trends have been
contributing to the pathogenesis of noise-induced injury, described in epigenetic effects. Patel et al.[30] demonstrated
functioning both as potential biomarkers of early damage, that a traumatic noise exposure caused a reversible up-
possible effective diagnostic tools as well as potential target regulation of a single miRNA and a significant down-
Downloaded from http://journals.lww.com/nohe by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AW

of treatments to prevent/ameliorate hearing loss. Although the regulation of a greater number of miRNAs, 2 hours and 1-
number of available investigations on the topic is limited, day post-exposure, respectively. This temporal pattern of
some interesting issues for the discussion emerged from our changes may be related to the progression of sensory cell
revision. degeneration post-noise insult.[66,67] The growth of the noise-
nYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 06/07/2024

induced lesion is expected to induce more cells to degenerate


Concerning acute to subacute noise exposure, no alterations and consequently, more miRNAs to be differently expressed.
in blood DNA methylation in military personnel could be Another possible factor influencing the temporal change in
detected following 10 day-blast exposure trainings (∼12 miRNA expression may be the differences in damaging
pounds per square inch.),[22] while an in vivo experiment triggers during the diverse phases of cochlear damage
demonstrated that moderate intensity noise (70–75 dB) for 3 pathogenesis. Mechanical stress may function as an initial
days was able to increase the DNA methylation of the Comt cause of cochlear effect, while other mechanisms of action,
gene in the medulla oblongata of exposed animals.[23] These i.e. energy exhaustion,[68] oxidative stress,[69] and ionic
latter results suggest that early epigenetic alterations may imbalance,[70-72] may contribute to longer-term damage.
occur at even not so high levels of noise in a gene that has
been reported to be involved with stress response, cognition, The gene expression analysis in relation to the epigenetic
anxiety, that may be all factors influenced by the noise modifications may also be important to understand the
insult.[62-65] However, the different species susceptibility to pathogenesis of noise-induced effects. DNA methylation
noise injury, the diverse type of noise exposure in terms of changes detected by Wang et al.[22] were associated with
duration and intensity, as well as the variable biological a decreased expression of KCNE1 and CYP2E1 genes,
matrices analysed may all play a role in affecting results directly involved in auditory processing and sound
and need further investigation. Acoustic trauma induced by perception. Such kind of analysis may also support the
broadband continuous noise exposure to 98-122 dB SPL for 2 understanding of possible noise-induced extra-auditory
to 3 hours was able to induce an increased expression of the effects as suggested by the reduced expression of the
enzymes HDAC 1–4[24-26] involved into the deacetylation of PAX8 gene in response to the increased DNA methylation
the lysine 9 residue on histone 3, with a consequent reduction in its promoter region, which may affect the control of the
of the H3K9ac levels 1 h post-exposure. Additionally, a expression of thyroid-specific genes and thyroid
comparable noise exposure induced the activation of the development, as well as sleep patterns. Also changes in
methyltransferase G9a, responsible for the Comt and Mc2r DNA methylation may influence certain
hypermethylation of the H3K9 site.[27] These preliminary brain functions, including memory and cognition, as a
data support the possible role of histone acetylation and possible outcome of chronic noise exposure.[23]
methylation levels in the pathogenesis of acute noise- Additionally, Patel et al.,[30] exploring the miRNA/mRNA
induced damage. This was further confirmed by the target pairs showed that the down-regulation of miR-183 was
capacity of HDAC inhibitors to reduce NIHL, suggesting associated with a Taok1 mRNA overexpression, responsible
their possible role as potential therapeutic strategies for the for the activation of the MAPK pathway potentially involved
prevention of hearing loss. in cochlear apoptosis as a mechanism of noise damage.
Overall, these data can assist the future development of
When the epigenetic impact of chronic, cumulative noise target molecular therapeutic perspectives in reducing
exposure along a professional career was assessed compared noise-induced cochlear damage.
to shorter periods of exposure, some DNA methylation
differences emerged.[22] In fact, several hypermethylated Importantly, as suggested for plasma miR-1229-5p[29] and
regions resulted associated with lifetime exposure miR-185-5p and miR-451[28] in occupationally noise-
compared to the absence of subacute alterations. It may be exposed workers, the differences in microRNA and
argued that DNA methylation effects, which represent highly downstream gene expression alterations between ONIHL
stable, long-lasting effect indicators, may be more likely patients and noise-exposed subjects with normal hearing,
representative of cumulative noise exposures, with respect may be important to understand the molecular responses to
to acute or proximal events. Interestingly, changes in DNA noise exposure and to define possible novel biomarkers to
methylation resulted also positively associated with noise- predict and eventually prevent its development and
related symptoms reported by military and veterans, such as progression. However, to achieve more sustained
tinnitus. In line with these results, sub-chronic conditions of conclusions in this regard, further research is necessary to
exposure have been demonstrated to induce different prove the causal association between miRNA changes and
epigenetic alterations in animals, i.e. increased Comt and noise exposure, and to determine whether these miRNAs may

86 Noise & Health ¦ Volume 22 ¦ Issue 107 ¦ October-December 2020


Leso, et al.: Epigenetic alterations as biomarkers of noise damage

effectively function as biomarkers of exposure and/or early 3. Lie A, Skogstad M, Johnsen TS, Engdahl B, Tambs K. The prevalence
effect. of notched audiograms in a cross-sectional study of 12,055 railway
workers. Ear Hear 2015;36:86-92.
Finally, some limitations emerged from our revision should 4. Nelson DI, Nelson RY, Concha-Barrientos M, Fingerhut M. The global
be stressed in order to plan methodologically adequate burden of occupational noise-induced hearing loss. Am J Ind Med
2005;48:446-58.
investigations able to provide more informative data. In 5. Metidieri MM, Rodrigues HF, Filho FJ, Ferraz DP, Neto AF, Torres
this context, additional studies should include a greater S. Noise-Induced Hearing Loss (NIHL): literature review with a
Downloaded from http://journals.lww.com/nohe by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AW

number of subjects/animals to avoid statistical power focus on occupational medicine. Int Arch Otorhinolaryngol
limitations due to the small sample size. Most of the 2013;17:208-12.
analysed investigations employed different kind of 6. Van der Molen HF, de Vries SC, Stocks SJ, Warning J, Frings-Dresen
MH. Incidence rates of occupational diseases in the Dutch construction
exposures. As the impact of noise depends upon the sector, 2010-2014. Occup Environ Med 2016;73:350-2.
nYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 06/07/2024

context and characteristics of the acoustic insult, including 7. Lie A, Skogstad M, Johannessen HA, Tynes T, Mehlum IS, Nordby
duration, intensity, intermittency, frequency, and KC, et al. Occupational noise exposure and hearing: a systematic
predictability,[73-76] a future standardized epigenetic review. Int Arch Occup Environ Health 2016;89:351-72.
research approach should provide a precise description of 8. WHO. Deafness and hearing loss. 2020. Available from: https://www.
who.int/health-topics/hearing-loss (Accessed March 19, 2020).
noise characteristics in order to achieve more comparable and 9. Sliwinska-Kowalska M, Pawelczyk M. Contribution of genetic factors
effective results. Additionally, most of the reviewed studies to noise-induced hearing loss: a human studies review. Mutat Res
could only applied a candidate gene approach analysis, 2013;752:61-5
uneffective to identify all genes that are potentially 10. Pawelczyk M, Van Laer L, Fransen E, Rajkowska E, Konings A,
affected by the exposure. Therefore, an extended analytical Carlsson PI, et al. Analysis of gene polymorphisms associated with K
ion circulation in the inner ear of patients susceptible and resistant to
strategy, on suitable and routinary applicable biological noise-induced hearing loss. Ann Hum Genet 2009;73:411-21.
matrices, should be pursued. Finally, a longitudinal 11. Konings A, Van Laer L, Van Camp G. Genetic studies on noise-
methodological design, for both genetic and symptom- induced hearing loss: a review. Ear Hear 2009;30:151-9.
based studies, appears necessary to understand molecular 12. Le Prell CG, Yamashita D, Minami SB, Yamasoba T, Miller JM.
alterations time-dependent trends and to verify their Mechanisms of noise-induced hearing loss indicate multiple methods
of prevention. Hear Res 2007;226:22-43.
possible employment as early biomarkers for hearing 13. Miguel V, Cui JY, Daimiel L, Espinosa-Díez C, Fernández-Hernando
damage, particularly in chronic occupational exposure C, Kavanagh TJ, et al. The role of MicroRNAs in environmental risk
settings. factors, noise-induced hearing loss, and mental stress. Antioxid Redox
Signal 2018;28:773-96.
In conclusion, additional studies are necessary to overcome 14. Wada T, Sano H, Nishio SY, Kitoh R, Ikezono T, Iwasaki S, et al.
the current gap in noise exposure-epigenetic changes Differences between acoustic trauma and other types of acute noise-
relationship. From an occupational health perspective, to induced hearing loss in terms of treatment and hearing prognosis. Acta
define the epigenetic impact of workplace noise exposure Otolaryngol 2017;137:48-52.
15. Hu BH, Henderson D, Yang WP. The impact of mitochondrial
and the role of such alterations in predicting hearing loss energetic dysfunction on apoptosis in outer hair cells of the cochlea
development may be absolutely important for a correct following exposure to intense noise. Hear Res 2008;236:11-21.
evaluation of risks, according also to an individually 16. Kamogashira T, Fujimoto C, Yamasoba T. Reactive oxygen species,
tailored-based approach. Additionally, epigenetic data may apoptosis, and mitochondrial dysfunction in hearing loss. Biomed Res
provide future support to the health surveillance plans of Int 2015;2015:617207.
17. Kurabi A, Keithley EM, Housley GD, Ryan AF, Wong AC. Cellular
chronically exposed workers, with the aim to detect early, mechanisms of noise-induced hearing loss. Hear Res 2017;349:129-37.
maybe preclinical alterations, and therefore guiding the 18. Sha SH, Schacht J. Emerging therapeutic interventions against noise-
adoption/implementation of the most effective preventive induced hearing loss. Expert Opin Investig Drugs 2017;26:85-96.
and protective measures to avoid disabling hearing loss. 19. Murgatroyd C, Wu Y, Bockmühl Y, Spengler D. The Janus face of
DNA methylation in aging. Aging (Albany NY) 2010;2:107-10.
20. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-
Financial support and sponsorship Agadjanyan EL, et al. Circulating microRNAs as stable blood-based
markers for cancer detection. Proc Natl Acad Sci USA
Nil.
2008;105:10513-8.
21. Moher D, Liberati A, Tetzlaff J, Altman DG; PRISMA Group.
Conflicts of interest Preferred reporting items for systematic reviews and meta-analyses:
the PRISMA statement. Int J Surg 2010;8:336-41.
There are no conflicts of interest. 22. Wang Z, Wilson CM, Mendelev N, Ge Y, Galfalvy H, Elder G, et al.
Acute and chronic molecular signatures and associated symptoms of
blast exposure in military breachers. J Neurotrauma 2019.
REFERENCES 23. Guo L, Li PH, Li H, Colicino E, Colicino S, Wen Y, et al. Effects of
1. Concha-Barrientos M, Campbell-Lendrum D, Steenland K. environmental noise exposure on DNA methylation in the brain and
Occupational noise: assessing the burden of disease from work- metabolic health. Environ Res 2017;153:73-82.
related hearing impairment at national and local levels. Geneva, 24. Wen LT, Wang J, Wang Y, Chen FQ. Association between histone
World Health Organization, 2004. (WHO Environmental Burden of deacetylases and the loss of cochlear hair cells: role of the former in
Disease Series, No. 9). noise-induced hearing loss. Int J Mol Med 2015;36:534-40.
2. Ding T, Yan A, Liu K. What is noise-induced hearing loss? Br J Hosp 25. Chen J, Hill K, Sha SH. Inhibitors of histone deacetylases attenuate
Med (Lond) 2019;80:525-9. noise-induced hearing loss. J Assoc Res Otolaryngol 2016;17:289-302.

Noise & Health ¦ Volume 22 ¦ Issue 107 ¦ October-December 2020 87


Leso, et al.: Epigenetic alterations as biomarkers of noise damage

26. Yang DH, Xie J, Liu K, Peng Z, Guo JY, Yu SK, et al. The histone 48. Wang Z, Liu Y, Han N, Chen X, Yu W, Zhang W, et al. Profiles of
deacetylase inhibitor sodium butyrate protects against noise-induced oxidative stress-related microRNA and mRNA expression in auditory
hearing loss in Guinea pigs. Neurosci Lett 2017;660:140-6. cells. Brain Res 2010;1346:14-25.
27. Xiong H, Long H, Pan S, Lai R, Wang X, Zhu Y, et al. Inhibition of 49. Stamatiou GA, Stankovic KM. A comprehensive network and pathway
histone methyltransferase g9a attenuates noise-induced cochlear analysis of human deafness genes. Otol Neurotol 2013;34:961-70.
synaptopathy and hearing loss. J Assoc Res Otolaryngol 50. Kurioka T, Matsunobu T, Satoh Y, Niwa K, Endo S, Fujioka M, et al.
2019;20:217-32. ERK2 mediates inner hair cell survival and decreases susceptibility to
28. Ding L, Liu J, Shen HX, Pan LP, Liu QD, Zhang HD, et al. Analysis of noise-induced hearing loss. Sci Rep 2015;5:16839.
Downloaded from http://journals.lww.com/nohe by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AW

plasma microRNA expression profiles in male textile workers with 51. Hutchison M, Berman KS, Cobb MH. Isolation of TAO1, a protein
noise-induced hearing loss. Hear Res 2016;333:275-82. kinase that activates MEKs in stress-activated protein kinase cascades.
29. Li YH, Yang Y, Yan YT, Xu LW, Ma HY, Shao YX, et al. Analysis of J Biol Chem 1998;273:28625-32.
serum microRNA expression in male workers with occupational noise- 52. Chen Z, Hutchison M, Cobb MH. Isolation of the protein kinase TAO2 and
induced hearing loss. Braz J Med Biol Res 2018;51:e6426. identification of its mitogen-activated protein kinase/extracellular signal-
nYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 06/07/2024

30. Patel M, Cai Q, Ding D, Salvi R, Hu Z, Hu BH. The miR-183/Taok1 regulated kinase kinase binding domain. J Biol Chem 1999;274:28803-7.
target pair is implicated in cochlear responses to acoustic trauma. PLoS 53. Raman M, Earnest S, Zhang K, Zhao Y, Cobb MH. TAO kinases mediate
One 2013;8:e58471. activation of p38 in response to DNA damage. EMBO J 2007;26:2005-14.
31. Moore LD, Le T, Fan G. DNA methylation and its basic function. 54. Wu MF, Wang SG. Human TAO kinase 1 induces apoptosis in SH-
Neuropsychopharmacology 2013;38:23-38. SY5Y cells. Cell Biol Int 2008;32:151-6.
32. Mansouri A, Chowdhury K, Gruss P. Follicular cells of the thyroid 55. Layman WS, Zuo J. Epigenetic regulation in the inner ear and its
gland require Pax8 gene function. Nat Genet 1998;19:87-90. potential roles in development, protection, and regeneration. Front Cell
33. Gottlieb DJ, Hek K, Chen TH, Watson NF, Eiriksdottir G, Byrne EM, Neurosci 2015;8:446.
et al. Novel loci associated with usual sleep duration: the CHARGE 56. Lu X, Wang L, Yu C, Yu D, Yu G. Histone acetylation modifiers in
Consortium Genome-Wide Association Study. Mol Psychiatry the pathogenesis of Alzheimer’s disease. Front Cell Neurosci
2015;20:1232-9. 2015;9:226.
34. Jones SE, Tyrrell J, Wood AR, Beaumont RN, Ruth KS, Tuke MA, 57. Song C, Kanthasamy A, Anantharam V, Sun F, Kanthasamy AG.
et al. Genome-wide association analyses in 128, 266 individuals Environmental neurotoxic pesticide increases histone acetylation to
identifies new morningness and sleep duration loci. PLoS Genet promote apoptosis in dopaminergic neuronal cells: relevance to
2016;12:e1006125. epigenetic mechanisms of neurodegeneration. Mol Pharmacol
35. Fechter LD, Gearhart C, Shirwany NA. Acrylonitrile potentiates noise- 2010;77:621-32.
induced hearing loss in rat. J Assoc Res Otolaryngol 2004;5:90-8. 58. Stilling RM, Fischer A. The role of histone acetylation in age-
36. Van Laer L, Carlsson PI, Ottschytsch N, Bondeson ML, Konings A, associated memory impairment and Alzheimer’s disease. Neurobiol
Vandevelde A, et al. The contribution of genes involved in potassium- Learn Mem 2011;96:19-26.
recycling in the inner ear to noise-induced hearing loss. Hum Mutat 59. Gräff J, Tsai LH. Histone acetylation: molecular mnemonics on the
2006;27:786-95. chromatin. Nat Rev Neurosci 2013;14:97-111.
37. Pawełczyk M, Rajkowska E, Kotyło P, Dudarewicz A, Van Camp G, 60. Tachibana M, Sugimoto K, Nozaki M, Ueda J, Ohta T, Ohki M, et al.

Sliwinska-Kowalska M. Analysis of inner ear potassium recycling G9a histone methyltransferase plays a dominant role in euchromatic
genes as potential factors associated with tinnitus. Int J Occup Med histone H3 lysine 9 methylation and is essential for early
Environ Health 2012;25:356-64. embryogenesis. Genes Dev 2002;16:1779-91.
38. Ramakrishnan NA, Drescher MJ, Khan KM, Hatfield JS, Drescher DG. 61. Yokochi T, Poduch K, Ryba T, Lu J, Hiratani I, Tachibana M, et al.
HCN1 and HCN2 proteins are expressed in cochlear hair cells: HCN1 G9a selectively represses a class of late-replicating genes at the nuclear
can form a ternary complex with protocadherin 15 CD3 and F-actin- periphery. Proc Natl Acad Sci USA 2009;10:19363-8.
binding filamin A or can interact with HCN2. J Biol Chem 62. Gogos JA, Morgan M, Luine V, Santha M, Ogawa S, Pfaff D, et al.
2012;287:3762846. Catechol-O-methyltransferase-deficient mice exhibit sexually
39. Kim YH, Holt JR. Functional contributions of HCN channels in the dimorphic changes in catecholamine levels and behavior. Proc Natl
primary auditory neurons of the mouse inner ear. J Gen Physiol Acad Sci USA 1998;95:9991-6.
2013;142:207-23. 63. Hains AB, Arnsten AF. Molecular mechanisms of stress-induced
40. Shen H, Liu W, Geng Q, Li H, Lu M, Liang P, et al. Age-dependent up- prefrontal cortical impairment: implications for mental illness. Learn
regulation of hcn channels in spiral ganglion neurons coincide with Mem 2008;15:551-64.
hearing loss in mice. Front Aging Neurosci 2018;10:353. 64. Papaleo F, Crawley JN, Song J, Lipska BK, Pickel J, Weinberger
41. Dickinson D, Elvevåg B. Genes, cognition and brain through a COMT DR, et al. Genetic dissection of the role of catechol-O-
lens. Neuroscience 2009;164:72-87. methyltransferase in cognition and stress reactivity in mice. J
42. Fridmanis D, Roga A, Klovins J. ACTH receptor (MC2R) specificity: Neurosci 2008;28:8709-23.
what do we know about underlying molecular mechanisms? Front 65. Ursini G, Bollati V, Fazio L, Porcelli A, Iacovelli L, Catalani A, et al.
Endocrinol (Lausanne) 2017;8:13. Stress-related methylation of the catechol-O-methyltransferase Val 158
43. Weston MD, Pierce ML, Rocha-Sanchez S, Beisel KW, Soukup GA. allele predicts human prefrontal cognition and activity. J Neurosci
MicroRNA gene expression in the mouse inner ear. Brain Res 2011;31:6692-8.
2006;1111:95-104. 66. Hu BH, Henderson D, Nicotera TM. Involvement of apoptosis in
44. Friedman LM, Dror AA, Mor E, Tenne T, Toren G, Satoh T, et al. progression of cochlear lesion following exposure to intense noise.
MicroRNAs are essential for development and function of inner ear Hear Res 2002;16:62-71.
hair cells in vertebrates. Proc Natl Acad Sci USA 2009;106:7915-20. 67. Yang WP, Henderson D, Hu BH, Nicotera TM. Quantitative analysis of
45. Lewis MA, Quint E, Glazier AM, Fuchs H, De Angelis MH, Langford apoptotic and necrotic outer hair cells after exposure to different levels
C, et al. An ENU-induced mutation of miR-96 associated with of continuous noise. Hear Res 200;196:69-76.
progressive hearing loss in mice. Nat Genet 2009;41:614-8. 68. Saunders JC, Dear SP, Schneider ME. The anatomical consequences of
46. Frucht CS, Santos-Sacchi J, Navaratnam DS. MicroRNA181a plays a acoustic injury: a review and tutorial. J Acoust Soc Am 1985;78:833-60.
key role in hair cell regeneration in the avian auditory epithelium. 69. Henderson D, Bielefeld EC, Harris KC, Hu BH. The role of oxidative
Neurosci Lett 2011;493:44-8. stress in noise-induced hearing loss. Ear Hear 2006;27:1-19.
47. Patel M, Hu BH. MicroRNAs in inner ear biology and pathogenesis. 70. Konishi T, Salt AN, Hamrick PE. Effects of exposure to noise on ion
Hear Res 2012;287:6-14. movement in guinea pig cochlea. Hear Res 1979;1:325-42.

88 Noise & Health ¦ Volume 22 ¦ Issue 107 ¦ October-December 2020


Leso, et al.: Epigenetic alterations as biomarkers of noise damage

71. Hsu CJ, Shau WY, Chen YS, Liu TC, Lin-Shiau SY. Activities of Na 74. Hong J, Kim J, Lim C, Kim K, Lee S. The effects of long-term exposure
(+),K(+)-ATPase and Ca(2+)-ATPase in cochlear lateral wall after to railway and road traffic noise on subjective sleep disturbance. J
acoustic trauma. Hear Res 2000;142:203-11. Acoust Soc Am 2010;12:2829-35.
72. Yamamoto H, Shi X, Nuttall AL. The influence of loud sound stress on 75. Kawada T. Noise and health−sleep disturbance in adults. J Occup
expression of osmotic stress protein 94 in the murine inner ear. Health 2011;53:413-6.
Neuroscience 2009;158:1691-8. 76. Salloum RH, Yurosko C, Santiago L, Sandridge SA, Kaltenbach JA.
73. Turner JG, Parrish JL, Hughes LF, Toth LA, Caspary DM. Hearing in Induction of enhanced acoustic startle response by noise exposure:
laboratory animals: strain differences and nonauditory effects of noise. dependence on exposure conditions and testing parameters and
Downloaded from http://journals.lww.com/nohe by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AW

Comp Med 2005;55:12-23. possible relevance to hyperacusis. PLoS One 2014;9:e111747.


nYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 06/07/2024

Noise & Health ¦ Volume 22 ¦ Issue 107 ¦ October-December 2020 89

You might also like