You are on page 1of 11

Physiology & Behavior 227 (2020) 113136

Contents lists available at ScienceDirect

Physiology & Behavior


journal homepage: www.elsevier.com/locate/physbeh

Noise and brain T


Archana Arjunan (Research Scholar), Ravindran Rajan (Associate Professor & Head)

Department of Physiology, Dr. Arcot Lakshmanaswami Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai –
600 113, India

ARTICLE INFO ABSTRACT

Keywords: Noise has become inexorable stress due to the increase in urbanization, automobile usage, Noise based occu­
Antioxidants pation, and lifestyle modifications such as recreation in day to day life of an individual. Noise not only affects the
Cognition auditory system but can also debilitate other non-auditory systems as evidenced in animal and human models.
Epigenetics Various studies have reported that noise has the potential to tarnish the Central Nervous System in hetero­
Neurotransmitter
geneous ways. This article reviews the researches on Noise and Brain with the spotlight on the effect of noise on
Noise
Stress
health that includes a) noise-induced impaired cognition on neurobehaviour, b) brain areas which are pre­
dominantly affected by the noise-induced oxidative stress, c) alteration of the neurotransmitter level in various
brain regions, d) alteration of the molecular mechanism induced by noise stress on brain cellular level, e) Noise-
induced deterioration of the morphological structures of brain and f) epigenetic modification by noise on brain.

1. Introduction 2. Noise and antioxidants

Noise is considered as a common environmental pollutant that is 2.1. Relationship between oxidative stress and antioxidants in noise stress
more than just a nuisance. It constitutes a danger that is real to people's
health by producing both physical and psychological stress [29]. Ac­ Oxidative stress refers to the cytopathological consequences of the
cording to the World Health Organization (WHO), noise is considered mismatch between the production of free radicals and the ability of the
as the second highest environmental stressor from the public health cells to defend against them. Free radicals are normal products of cel­
perspective [15]. In modern lifestyle, application of various auto­ lular aerobic metabolism. Superoxide (o-) and hydroxyl (OH'),
mobiles, urban traffic, household appliances, media [23], in­ Hydrogen peroxide (H2O2), and peroxynitrite (ONOO-) have an im­
dustrialization, using weapons in Military and Army forces have in­ portant contribution to the cellular redox state. These molecules are
creased the noise stress/ pollution to the individuals who are working referred to as reactive oxygen species (ROS). The major sources of ROS
in such environment [27]. Noise is described as an unwanted sound, are mitochondrial oxidative metabolism, enzymatic reactions involving
that is either chronic or intermittent, and can be described in terms mixed-function oxidation, and autoxidation of small molecules [63].
including its frequency, intensity, frequency spectrum, and shape of Oxidative stress originating from outside the body is a feature of life in
sound pressure through time [7]. If the level of noise exceeds 90 decibel the modern world. Negative lifestyle patterns can be ominous and can
dBA it becomes a source of stress [67] and affects the health in both also converge with environmental stressors to attack the health of the
physical and psychological perspectives. Various studies suggest that living organisms through related oxidative pathways [30].
noise can delimitate the cellular homeostasis in Hearing [27], immune To reduce the burden of oxidative stress, the body produces anti­
system [66], Nervous system [5,7,67], sleep disturbances [5] and oxidants that are either generated by endogenous (enzymatic and non-
Cardiovascular System [5,23]. enzymatic antioxidants) or exogenous mechanisms. The major anti­
In this review, the focus has been emphasized on the effects of noise oxidant enzymes that are directly involved in the neutralization of ROS
especially in the impairment of the CNS via increased oxidative stress, and RNS are superoxide dismutase (SOD), catalase (CAT), glutathione
imbalance in neurotransmitter level, altered cellular cytoarchitecture peroxidase (GPx) and glutathione reductase (GRx). The non-enzymatic
by the detoriatiation of the molecular functions, impaired cognition, antioxidants are also divided into metabolic antioxidants and nutrient
and genetic modification. antioxidants. Metabolic antioxidants are endogenous antioxidants,
produced by metabolism in the body, such as lipoic acid, glutathione, L-


Corresponding author.
E-mail address: ravindran89@gmail.com (R. Rajan).

https://doi.org/10.1016/j.physbeh.2020.113136
Received 25 December 2019; Received in revised form 13 July 2020; Accepted 11 August 2020
Available online 14 August 2020
0031-9384/ © 2020 Elsevier Inc. All rights reserved.
A. Arjunan and R. Rajan Physiology & Behavior 227 (2020) 113136

arginine, coenzyme Q10, melatonin, uric acid, bilirubin, metal-che­ reported that the presence of increased LPO in Hippocampus and
lating proteins, transferrin, etc. Nutrient antioxidants are exogenous medial Prefrontal Cortex in chronic stress-induced animals [39].
antioxidant natured compounds which cannot be produced in the body
and must be provided through food or supplements, such as vitamin E, 2.3. Nitric oxide (NO) and hydrogen peroxide (H2O2)
vitamin C, carotenoids, trace metals (selenium, manganese, zinc), fla­
vonoids, omega-3, and omega-6 fatty acids, etc. [52]. NO is a highly reactive signal molecule in the CNS. It is a unique
Overproduction of free radicals and ROS can cause oxidative da­ messenger molecule that serves various physiological functions
mage to biomolecules, leading to various chronic diseases such as throughout the body [67]. Table 1 showed that during sub-acute noise,
atherosclerosis, cancer, diabetes, rheumatoid arthritis, post-ischemic the NO and H2O2 levels were increased in various discrete brain regions
perfusion injury, myocardial infarction, cardiovascular diseases, in rats [67]. Elevated NO indicates increased nitrosative stress, leading
chronic inflammation, stroke and septic shock, aging and other de­ to cell degeneration and necrosis by causing inhibition of mitochondrial
generative diseases in humans [4]. In comparison to all the other sys­ cytochrome oxidase in neurons [76].
tems, the brain is considered to be the most vulnerable organ that can H2O2 is capable of serving as a messenger to carry a redox signal
be subjected to free radical damage due to peroxidation, and this fea­ from the site of its generation to a target site. Among the various oxygen
ture is due to its highest percentage of unsaturated fats which is en­ metabolites, H2O2 is considered as the most suitable messenger for
ormously supplied with molecular oxygen [39] and lower antioxidant redox signaling. Hydrogen peroxide modulates the activity of tran­
activity when compared to other tissues [4,31,70]. scription factors: in mammalian cells (AP-1, NRF2, CREB, HSF1, HIF-1,
Various studies have evidenced that noise stress instigates increased TP53, NF-κB, NOTCH, SP1, and SCREB-1) [62].
ROS and RNS generation which are capable of breaching lipid and
protein molecules and damaging DNA thereby triggering the loss of
2.4. Superoxide dismutase (SOD)
function and leading to cell death following the noise exposure
[39,48,67].
SOD, the first line of defense against free radicals, catalyzes the
dismutation of superoxide anion radical (O2•ˉ) into hydrogen peroxide
2.2. Lipid peroxidation (LPO) (H2O2) by reduction [52]. There are three distinct isoforms of SOD
(1,2,3) that have been identified. SOD1 and SOD2 are mainly involved
Lipid peroxides are an important class of biomolecules generated by in the elimination of O2 - in the cytosol and mitochondria, respectively
oxidative stress in cells [24]. The complex process of lipid peroxidation [31]. Table 1 shows a remarkable increase in SOD in discrete brain
involves the interaction of oxygen-derived free radicals with poly­ regions of noise stress group animals. SOD is an important enzyme fa­
unsaturated fatty acids and finally results in a variety of highly reactive mily in living cells for maintaining normal physiological conditions and
electrophilic aldehydes that are capable of easily attaching covalently for coping with stress by protecting the biological integrity of the cells
to proteins by forming adducts with cysteine, lysine, or histidine re­ and tissues against harmful effects of superoxide free radical [49].
sidues. Among the aldehydes formed, malondialdehyde (MDA), 4-hy­
droxynonenal (HNE), and acrolein represent the major products of lipid 2.5. Glutathione peroxidases (GPX)
peroxidation which affect the glutamate level in brain [70] Numerous
studies have confirmed that acute stress and sub-acute stress have in­ Table 1 shows that acute subacute and chronic noise stress increased
creased the LPO level in discrete regions of the brain (Table 1). No the GPx in discrete regions of the brain. GPX contains a family of
deviations in the LPO level were reported between animals exposed to multiple isoenzymes (GPX 1–8) which catalyze the reduction of H2O2
chronic noise stress and naïve animals [57] and also Manikandan et al., and lipid peroxides utilizing GSH as an electron donor. GPX is located in

Table 1.
Noise stress and antioxidant level in Brain.
S.No Model Noise Stress Antioxidant Brain Regions References

1 Wistar albino/ Acute (100 dB-4 h / 1 Day) SOD, CAT, GPx, GSSH, LPO Increased in CC, CB, Midbrain, Pons, Medulla, HY, HI [57]
Male GSH, GSH/GSSH Ratio,Protein thiols Decreased inCC, CB, Midbrain, Pons, Medulla, HY, HI
Sub acute Stress (100 dB-4 h/ 15 Days) SOD Increased in Midbrain, Medulla, Pons, Hippocampus
CAT Increased in CC, CB, Midbrain, HY
GPx, GSSH Increased in CC, CB, Midbrain, Pons, Medulla, HY, HI
GSH,GSH/GSSH Ratio, Protein Thiols Decreased in CC, CB, Midbrain, Pons, Medulla, HY, HI
LPO Increased in CB, Pons, Medulla, HY, HI
Chronic Stress (100 dB- 4 h/30 Days) SOD Increased in Midbrain, Pons, Medulla
CAT Increased in CC, Midbrain
GPx Increased in CB, Pons, HY
GSH, GSH/GSSH, Protein thiols Decreased in CC, CB, Midbrain, Pons, Medulla, HY, HI
GSSH Increased in CC, CB, Midbrain, Pons, Medulla, HY, HI
LPO Not deviate from the control in any regions of brain
2 Wistar albino/ Sub acute Stress (100 dB-4 h/ 15 Days NO, LPO, Vit- C, GST, GPx, SOD Increased in CC, CB,CS,BS,HY,HI [67]
Male CAT Increased in CC, CB,CS,HY,HI
Protein Thiols, GSH, GR, Decreased in CC, CB,CS,BS,HY,HI
3 Wistar albino/ Sub acute Stress (100 dB-4 h/ 15 Days H2O2,NO,LPO,SOD,CAT,GPx,GST Icreased in CC, CB,CS,BS,HY,HI [38]
Male Protein Thiols, GSH, GR, Vit-C, Vit-E Decreased in CC, CB,CS,BS,HY,HI
4 Wistar albino/ Chronic stress 100 dB-4 h/1,15,30 Days SOD, GPx, CAT, LPO Increased in HI, mPFC [40]
Male GSH Decreased in HI, mPFC
5 Mice 80 dB-2 h/Day 1wk / 3 wk MDA 1 Wk – Increased MDA in HI [10]
3 Wk – Increased MDA in HI & AC
SOD 1 Wk – Decreased in HI
3 Wk – Decreased in HI & AC

CC- Cerebral Cortex, CB – Cerebellum, CS – Corpus Striatum, BS – Brain Stem, HY – Hypothalamus, HI – Hippocampus, AC- Auditory Cortex, mPFC- medio Pre Frontal
Cortex.

2
A. Arjunan and R. Rajan Physiology & Behavior 227 (2020) 113136

both cytosol and mitochondria. GPX1 has been regarded as one of the concentrations which when exceeded can be lethal to neurons [11,34].
major antioxidant enzymes in the brain, which is expressed pre­ Table 2 shows that chronic noise increases the Glutamate level in
dominantly in microglia but not in neurons, and up-regulation of GPX1 hemicerebrum. Kumar et al. stated that glutamate induces LTP by
could be one of the protective responses against neuronal injury [31]. NMDAR activation and Ca2+ influx. When Glutamate concentration
becomes excess Ca2+ reversibly increases ROS, which leads to in­
2.6. Catalase (CAT) hibition of LTP [34].

Table 1 shows that acute, subacute, and chronic noise stress in­
creased the catalase in discrete regions of the brain. Catalase is re­ 3.2. Acetylcholine
sponsible for the conversion of H2O2 to water and oxygen using either
iron or manganese as a cofactor. It is located in peroxisomes and is also Acetylcholine (ACh) is a fast-acting, point-to-point neurotransmitter
found in the cytoplasm and mitochondria. The role of catalase is minor at the neuromuscular junction and autonomic ganglia. ACh appears to
at low levels of H2O2 but becomes increasingly important at higher act as the primary excitatory neurotransmitter in the periphery [53]. It
levels of H2O2 [31]. modulates various physiological functions and is also involved in
mammalian stress reactions. ACh is hydrolyzed by the enzyme known
2.7. GSH as Acetylcholinesterase [64]. ChE is involved in cell proliferation, dif­
ferentiation, responses to stress, and amyloid formation. Activation of
Glutathione reductase, a flavoprotein enzyme, regenerates GSH ACh in the brain depends on the nature of stress and its duration [16]
from GSSG, with NADPH as a source of reducing power [52]. Glu­ and also if the stress is predictable type then I can also increase the
tathione (γ-glutamylcysteinyl glycine, GSH) is a sulfhydryl (-SH) anti­ AChE activity in the brain.
oxidant, antitoxin, and enzyme cofactor. It exists in two forms: 1. The
antioxidant “reduced glutathione” (GSH), 2. The oxidized form “glu­
tathione disulfide (GSSH). The GSH/GSSH ratio can serve as a sensitive 3.3. GABA
indicator of oxidative stress. GSH plays a major role in several biolo­
gical functions such as protein synthesis, enzyme catalysis, transmem­ GABA is a primary inhibitory Neurotransmitter in CNS and it acts as
brane transport, and cell maturation [30]. Several studies have reported modulation of anxiety-like behavior [47] and is protective against
that GSH is involved in inhibiting apoptotic cell death and DNA damage oxidative stress [34,55]. During chronic noise exposure, the GABA level
in cells [31]. GSH level and GSH/GSSH ratio were found to be de­ in hemicerebrum was found to be decreased in Sprague-Dawley rats
creased in various brain regions when rats were exposed to acute, [14]. Noise increases H2O2 and reduces the GABA level by a) a sus­
subacute, and chronic noise stress (Table 1). tained increase in [Cl-]i, b)Desensitization of GABA receptors c) H2O2
directly or indirectly acts on GABAA [55] and also that Increased NO
2.8. Vitamin E and vitamin C inhibits the tonic and phasic GABAergic response in the hippocampus
[34].
When animals are exposed to subacute noise, the stress decreases
the vitamin E and vitamin C in various brain regions [76]. Vitamin E is
a lipid-soluble antioxidant that can attenuate the effects of peroxide and 3.4. Catecholamines
protect against lipid peroxidation in cell membranes [31]. Vitamin C
also known as ascorbic acid is a water-soluble vitamin. It is essential for The relationship between catecholamine and stress is intertwined.
collagen, carnitine, and neurotransmitters biosynthesis [52] and is in­ There are three distinct peripheral catecholamine systems, each with
volved in the removal of free radicals by electron transfer and can also different effectors, regulation, and roles. The three systems are the
act as a cofactor for antioxidant enzymes [31]. sympathetic nervous system, adrenomedullary hormonal system, and
DOPA-dopamine autocrine/paracrine system. Both the peripheral
3. Noise and neurotransmitters sympatho-adrenomedullary and central catecholaminergic systems are
activated by various psycho-social and physical stressors [18]. During
Efficient communication among a large number of brain cells (i.e., the stress activation of sympathoadreno medullary and Hypothalamic
neurons) is necessary for the normal functioning of the nervous system. –Pituitary – adrenal axis system [28,75,76], increased the production of
It involves the release of neurotransmitters that bind to specialized Corticosterone and Catecholamines [28,73]. Adrenal steroids bind with
receptors on the target cell, changing its activity and it can be divided the neurons by Type I and Type II glucocorticoid receptors [6]. Chronic
into three classes: (1) amino acids (e.g., γ¬aminobutyric acid [GABA], exposure of Glucocorticoid receptors in the brain results in the in­
glycine, and glutamate); (2) biogenic amines (e.g., serotonin, dopa­ creased release of serotonin and glutamate which may interact pre-or-
mine, norepinephrine, epinephrine, and histamine); and (3) other (e.g., post-synaptically and can thereby facilitate the destructive effects in the
acetylcholine, adenosine triphosphate, and adenosine) [72]. brain [51]. Enormous studies revealed that noise stress increased the
This review emphasizes that noise alters the neurotransmitter level corticosterone and catecholamines [5,7,14,32,48,75] and also corti­
in the experimental rat brain. Various studies have reported that costeroids were found to increase catecholaminergic and cholinergic
Oxidative stress can alter the neurotransmitter synthesis in neurons, activity in the brain [19,26]. Norepinephrine is a catecholamine. Nor­
and also numerous neurotransmitter systems are sensitive to ROS, in­ adrenergic neurons project bilaterally from the locus ceruleus along
cluding adrenergic, dopaminergic, serotonergic and GABAergicsystems distinct pathways to many locations, including the cerebral cortex,
[55]. The effects of ROS on neurotransmission can occur via various limbic system, and spinal cord, forming a neurotransmitter system.
mechanisms. Norepinephrine is released when a host of physiological changes are
activated by a stressful event. The effects of Norepinephrine are alert­
3.1. Glutamate ness and arousal, and influences on the reward system [46]. Noise stress
also exhibited significant elevation of CA in various regions (Table 2). It
Glutamate is the excitatory neurotransmitter that is present in the may be due to the following factors: 1) Noradrenergic pathways and
majority of synapses throughout the brain and spinal cord and acts on adrenergic receptors located from various regions of brain and 2) Ge­
metabotropic and inotropic receptors including the NMDA receptors. It netic manipulation of NE system that increases NE neurotransmission
excites all CNS neurons and is generally present at millimolar and protects animals from stress [46].

3
A. Arjunan and R. Rajan

Table 2.
Noise and Molecular mechanism in brain.
S.No Model Noise Stress Methods Molecules Regions Remarks Reference

1 Wistar / Male Sub acute stress 100 dB- ELISA TNF-α,IL-2, IF-ɣ Increased in CC, CB, HI neuronal damage [75]
4 h/15 Days
NF-κB Increased in CC, CB, HI
Hsp 70 Increased in CC, CB, HI
IL-4 Decreased in CC, CB, HI
DNA Fragmentation Increased in CC, CB, HI
2 Wistar /Male Chronic stress 100 dB- ELISA Aβ, Aβ1–40, Aβ1–42, APP, β- Increased in HI Aβ accumulation & AD [12]
4 h/28 Days secretase, ɣ- secretase
Western Blot APP, GFAP, IBA1
RT-PCR APP mRNA
ELISA TNF-α & RAGE Increased in HI
Activin A & PDGF-AA Decreased in HI

4
3 Wistar /Male 100 dB- 4 h/14Days Western Blot PS202, PS396,PS404, and PS422 Increased at 0,3,and 7 days after exposure of noise Decreased at 14th day of noise [14]
in HI
4 Sprague/ Male Acute Noise (100 dB – PCR CRH, CRH-1, CRH-2 Acute – Downregulate CRH-R1 & CRH-R2 Increased CRH mRNA expression [21]
4 h/ 1 day)
Chronic Noise (100 dB- Chroni– Upregualte $pCRH-R1 & downregulate CRH-R2
4 h/30 days)
5 Mice 80 dB-2 h/Day$p1 wk / 3 WB Tau Phosphorylation 1 wk – Increased non-phosphorylated tau at Ser195/198/199/202 & decreased Hyperphosphorylation in HI [10]
wk phophorylated tau at Ser396 epitope in HI$pNo change in AC
3 wk - Increased non-phosphorylated tau at Ser195/198/199/202 & decreased
phophorylated tau at Ser396 epitope in HI & AC
6 Mice/Male 80 dB – 2 h/ six weeks WB SYN, PSD - 95 Reduced expression in CC, HI Impaired Cognition [73]
PCR BDNF mRNA Reduced epression in HI & PFC

dB- decibel, TNF-α – Tumour Necrosis Factor – α, IL −2 – Interleukins 2, IF-ɣ - Interferon- ɣ, NF-κB- nuclear factor kappa-light-chain-enhancer of activated B cells, Hsp 70 – Heat Shock Protein 70, nNOS-, Aβ- Amyloid β,
APP- Amyloid Precursor Protein, GFAP - Glial fibrillary acidic protein, BDNF- Brain-derived neurotrophic factor, PSD-Post Synaptic Density, IBA1- Ionized calcium Binding Adaptor molecule 1,PDGF-AA – Platelet Derived
Growth Factor, CRH – Corticotrophin Releasing Hormone, ELISA –Enzyme Linked Immunosorbent Assay, WB-Western Blot, RT-PCR – Real Time polymerized Chain Reaction.
Physiology & Behavior 227 (2020) 113136
A. Arjunan and R. Rajan Physiology & Behavior 227 (2020) 113136

Fig. 1. When noise reaches the brain via the auditory pathway initiates the secretion of Cotricotrophin Releasing Hormone (CRH) by the Paraventricular Nucleus of
the hypothalamus (PVN). CRH act on corticotrophs cells of the anterior pituitary and its synthesis andrenocorticotrophic normone (ACTH) which enter into cir­
culation, reaches adrenal gland. Adrental cortex synthesizes corticosteroids (CORT). Ehen the CORT level increses it binds to the Type I & Type II glucocorticoid
receptors present in neurons, follows various inflammatory pathways such as TNF-α, NF-κB. Activation of Proinflammatory cytokins, oxidative stress which alerts the
neutrotransmitter synthesis, secretion, and neurotoxicity. Excess free redicals and inflammatory cytokines cause Neurofibrillary tangels formation (NFT), shrinkage
of dendrites in soma, decreased mRNA procduction, DNA methylation, and DNA fragmentation in nucleus leads to impairs Long term potentiation (LTP), cognition
and neurodegeneration.

4. Noise on molecular level in brain HSPs are a family of functionally-related proteins, these are in­
volved in the cellular repair by re-folding the proteins to act as mole­
Various studies have enumerated that stress can delimitate the cular chaperones in physiological processes [75]. The stress activation
molecular level in a living organism. This review explains how noise of the Hypothalamo pituitary adrenal (HPA) axis and sympathoadreno
can alter the molecular mechanism in the brain. Limited studies denote medullary system leads to increased Catecholamine levels which ele­
that noise disrupts the neuronal cell function by molecular level vate the immunosuppressive hormones glucocorticoids and activation
(Fig. 1). of inflammatory cytokines [75]. Altered cytokine secretion increases
Wankhar et al. reported that sub-acute noise significantly increased the inflammatory cytokine on exposure to noise which can result in the
pro-inflammatory cytokines such as Interleukin-2 (IL-2), Interferon- γ neuroinflammation with reactive oxygen/nitrogen species (RONS)
(IF-γ), Tumor necrosis factor- α (TNF- α), decreased anti-inflammatory playing a major role in eliciting neuronal DNA smear pattern in the
IL-4 cytokine, and increased Nuclear factor - κB (NF-KB), Heat shock brain [76]. Gai et al. reported that the long-term noise exposure causes
protein 70 (HSP 70) secretion in the cerebral cortex, cerebellum and significant and persistent abnormalities in the corticotropin-releasing
hippocampus [75]. TNFα is considered to be a “master regulator” of factor (CRF) signaling system and the hyperphosphorylation of tau in
pro-inflammatory cytokine production and can be synthesized in the the hippocampus [23]. The CRF system plays a major role in the co­
microglia, astrocytes, and by some populations of neurons [41,50]. ordination of neuroendocrine and neuropsychiatric responses to stress.
Activation of TNF signaling through TNFR1 and TNFR2 can activate the It consists of CRF and its receptors, CRFR1 and CRFR2. CRF and its
number of intracellular signaling pathways including nuclear factor receptors are found in the hippocampus and prefrontal cortex, and the
kappa –B (NF-κB), p38, C-jun N-terminal kinase (JNK) and the cer­ dysregulation of the CRF system in these regions cause alterations in
amide/sphingomyelinase signaling pathway, resulting in various re­ Alzheimer's disease (AD) -like pathology in animals [9]. Chronic noise
sponses including inflammation, proliferation, cell migration, apop­ exposure causes significant and persistent hyperphosphorylation of tau
tosis, necrosis [41], inhibition of long term potentiation, accumulation and formation of pathological Neurofibrillary tangles (NFT) tau (PS202,
of Aβ and decreased two neuroprotective cytokines such as activin A PS396, PS404, and PS422) in the hippocampus and the prefrontal
and Platelet derived growth factor (PDGF-AA) during chronic noise in cortex, which may lead to cognitive impairment and neurodegenerative
the hippocampus [12]. diseases like AD [14]. Glutamate and the proinflammatory cytokine,
Activin βA, a member of the TGF-β superfamily regulates the late- TNFα, have been suggested to contribute to increasing β -amyloid
long term potentiation (L-LTP) induction, morphology of dendritic peptide (Aβ) generation and also elevates the level of Receptor for
spines, and neurogenesis. It also has a neuroprotective function [1]. Advanced Glycation Endproducts (RAGE) in the hippocampus. Accu­
Activation of PDGF indirectly inhibits N-methyl-D-aspartate (NMDA) mulation of Aβ, neuroinflammation, and prominent tau pathology in
[12]. During Noise, a low level of PDGF expression can cause Glutamate the hippocampus are important pathological features of AD. Aβ pep­
excitotoxicity and neurodegeneration. Glial fibrillary acidic protein tides are generated as a product of APP sequential degradation by the
(GFAP), an astrocyte marker, can help astrocytes maintain their me­ APP-cleaving enzyme. An imbalance between the production, clear­
chanical strength and shape and participates in processes linked to cell ance, and aggregation of these peptides causes Aβ to accumulate and is
movement and structure and plays a vital role in astrocyte-neuron thought to be the initiating factor in AD. These processes can ultimately
communication [17]. Ionized calcium-binding adapter molecule 1 (Iba- lead to neuronal metabolic failure and synaptic dysfunction [12].
1) is involved in the membrane ruffling and phagocytosis and are ex­ Chronic exposure to corticosterone in rats reduces the mRNA expres­
pressed centrally by microglia and infiltrating macrophage [8]. sion of BDNF and NT-3 in the hippocampus [6].

5
A. Arjunan and R. Rajan

Table 3.
Noise on morphology of brain.
S.No Model Noise Stress Methods Brain regions Expression Remarks Reference

1 Wistar/Male Sub acute stress (100dB- Immunohistochemistry CB Increased number of nNOS positive cell in Purkinji Increased free radical [76]
4 Hrs / 15 D cells generation
Decreased number of Purkinji cells/50,000µm2per
area
2 Wistar/Male Chronic stress camera Lucida drawings mPFC Dendrite retraction and disbranching Impairment of spatial memory [40]
100 dB-4 h/1,15,30
D
HI Dentritic count (3rd& 4th order Neurons)
reduced.Atrophy of pyramidal neurons
3 Wistar/Male Chronic stress Protein array Imaging HI Increased expression of GFAP, IBA1 neuroinflammation [12]

6
100 dB- 4 h/28 D
4 Sprague–Dawley /Male Chronic stress Nissel staining HI Reduced MOD in CA1 and DG regions. Excitotoxicity in HI [13]
100 dB – 4 h / 30 Days No significant change in CA3 region
Immunohistochemistry Decreased NR2B expression in CA1, CA3, and DG Internalization of NR2B
regions
5 C57BL/6NJ < 85 dB – 8 h/1 D Coronal section from each hemisphere – measured Cortical thickness and Reduced cortical thickness and shrunken brain Impairs Cognition [29]
Mice by NanoZoomer Brain volume volume
Densitometry mPFC area and Neuronal No difference in right/left hemisphere mPFC area
density Reduced neuronal density
Volumetric analysis HI, Reduced HI volume and Neuroal density
Densitometry DG Density
PMC section - NanoZoomer Amygdala Cortical area Reduced
6 Wistar /Female 198 dB −1.7 ± Immunohistochemistry c-Fos,c-Myc,β-APP Increased expression of c-Fos,c-Myc,β-APP cerebral Loss of metabolic and growth [56]
Cortex, Thalamus, Hippocampus.
9 Mice/Male 80 dB – 2 h/ 6 Wks Nissl's staining HI, CX shrunken, irregularly arranged, and weakly stained Impaired Cognition [10]
in CA1 & PFC
Physiology & Behavior 227 (2020) 113136
Table 4.
A. Arjunan and R. Rajan

Noise on neurobehaviour.
S.No Model Noise Stress assessment Behavior Remarks Reference

1 Wistar albino/Male Sub acute stress (100 dB- 4 h / Rota Rod Decreased time spent in Rota Rod Decreased Motor coordination [67,76]
15 Days)
Narrow Beam Increased in Time taken to cross in seconds & number of slips Decreased Balance & Motor
coordination
Elevated Plus Maze Time spent and entry in open arm decreased Increased Anxiety Behaviour [67]
2 Rheus Mild Noise (100–110 dB – Delayed Response Task Delayed working memory Impaired Cognition [3]
Monkey/ Female & Male 30 min)
Visual Discrimination Task Decreased Visual Discrmination
3 C57BL/6NJ < 85 dB – 8 h/ 1 Days Novel Object Recognition Task Increased time spent with old object Impaired PFC cognitive function [29]
Mice
Elevated Plus Maze Increased Time spent in the closed arms
Rota Rod Increased shorter time
Morris Water Maze Increased swim latency and lower swim distance
Balance Beam Test Increasedtime (sec) to pass across the beam & number of falls and turns
4 Wistar albino Sub acute stress Open Field Test Day 1- Decreased ambulation peripheral square, central square, Increased Anxiety Behaviour [74]
/Male (100 dB- 4 h / 15 Days) grooming, increased rearing, fecal bolus &immobilization
Day 15 –Decreased ambulation in central squares, Increased
immobilization & fecal bolus

7
Elevated Plus Maze Decreased number of open arm entry & Percentage of open arm entry
5 Sprague–Dawley /Male 55–95 dB – 20 min/1 to 15 Days Eating Behaviuor Less food intake in beginning of Stress period and becone normalize Alters eating behavior [33]
course of further stress sessions.
Longer latency
Decreased duration, speed of food intake, defecation & Exploration
6 Sprague– 90 dB- 15 min/ 6 Session Eating Behaviuor by Tailpinch Increased Eating Stress induced eating behavior [77]
Dawley /Male experience
7 Holtzman male rats 90,100,110 dB - 20 min / 5 Days Feeding Beaviour No significance change in three groups Inibitory effect of feeding is lesser and [54]
shorter
8 Wistar albino /Male 15 min - 30 Days Food Intake Decreased food intake Reduced Body Weight [2]
9 Sprague–/Female (Virigin& 114 dB- 10 min – on 5th day of Endocrine Virigin-Increase total activity,grooming and rearing Lactaing rats showed normal behavior [78]
Lactating) Surgery Behavioural Lactating - higher than the virgin but directed towards to the pups. reactivity due to prolactin
Response
10 Wistar albino Sub acute stress Open Field Test Decreased ambulation in central squares, peripheral squares, rearing, Increased anxiety, fear and altered [67]
/Male (100 dB- 4 h / 15 Days grooming and fecal bolus &Increased immobilization locomotor activity.
11 Wistar albino/ Male Chronic stress Radial Arm Maze Increased number of reference memory error, working memeory error Spatial memory impairment [40]
100 dB-4 h/1,15,30 Days and time taken to visit all baited arms.
12 Sprague–Dawley /Male Chronic stress Morris Water Maze Increased escape latency, Decreased time spent in target quardarnt Spatial learning and memory [13]
100 dB – 4 h / 30 Days
13 Sprague–Dawley /Male Acute (100 dB – 4 h/ 1 Days) Defensive withdrawal test Decreased time spent in open field Anxiety behavior [21]
Chronic -(100 dB- 4 h/30 Days) No signifacant Decreased Anxiety
14 Mice 80 dB- Open Field Test Decreased number of crossing & Rearing impaired spatial learning [10]
/Male 2 h/ 6 Wks.
Morris Water maze increased escape latency
Physiology & Behavior 227 (2020) 113136
A. Arjunan and R. Rajan Physiology & Behavior 227 (2020) 113136

5. Noise on morphology of brain all the mental processes that are collectively known as cognition, in­
cluding the ability to learn new things, intuition, judgment, language,
Many studies have evidenced that the vulnerability may emerge and remembering.” Impaired cognition is often associated with con­
from stress effects on higher cortical and limbic structures [3,42]. siderable socioeconomic burden, adding to the public health imperative
Table 4 describes various types of noise stress which causes neuroa­ [22]. Table 5 shows that animals exposed to noise have impaired cog­
natomical changes in experimental animals. The cumulative exposure nition. Memory is defined as the process of “information acquisition,
to glucocorticoid results in the disruption of the hippocampus by the storage, and retrieval”. There are several dichotomies of memory (i.e.,
following mechanism: 1) inhibition of energy metabolism, 2) involution explicit and implicit, short and long-term, semantic, and episodic.
of the dendrite process 3) Inhibition of long term potentiating and Neurotransmitter systems and BDNF are involved in learning and
paradigms 4) inhibition of primed burst potentiation 5) Presence of memory processes, synaptic plasticity and a substantial part of learning
NMDA receptors of glucocorticoid [3, 79] 6) alteration in synaptic and memory impairment are due to changes in neurotransmission and
terminal structures and 7) Inhibition of neuronal regeneration in CA3 BDNF [79]. The brain is a major target organ for corticosteroids
regions [65]. [42]. Increased oxidative stress, neurotransmitter alteration may induce
Table 3 reports that noise stress significantly increased Dopamine changes in Long-term Potential and the synaptic plasticity leads to
levels in the brain [57,75,76]. Excess dopamine in the cytosol is easily learning and memory impairment in noise-induced animals.
metabolized by monoamine oxidase (MAO) to produce hydrogen per­
oxide or by autoxidation to synthesize quinine. Autoxidation of dopa­ 6.2. Motor coordination
mine or L-dopa via quinone formation generates free radicals such as
superoxide radicals and hydrogen peroxide. This increases free radicals CNS controls motor coordination by the activation of the motor
generation which could have initiated a cascade of free radical-medi­ cortex, cerebellum, and basal ganglia. The dopaminergic system is a
ated changes in the cerebellar Purkinje cells morphology [76]. Stress major neurotransmitter involved in movement and locomotion via the
elicits excessive DA release which can be normalized by DA receptor basal ganglia. The cerebellum is known to contribute to motor control
stimulation impairing the PFC function, high levels of Norepinephrine and coordination activity by integrating the motor output with ongoing
release impairs the PFC working memory abilities via α1-receptor sensory feedback. Noise increases dopamine which facilitates free ra­
mechanism and catecholamines may interact to take the PFC “Off-line” dical damage in Cerebellum leading to impaired motor coordination.
[3,79]. And also Cerebellar Purkinje cells release inhibitory neurotransmitter
Saljo et al. reported that 198 dB of short lasting impulse noise in­ GABA which reduces the transmission of impulses. Due to the reduction
creases the expression of c-Fos,c-Myc,β-APP cerebral Cortex, Thalamus, of glutamatergic and GABAergic neurotransmission systems which
Hippocampus [56]. The proto-oncogene c-Myc activates caspases, differently activate altering the motor coordination [76].
which mediate the apoptotic process and regulate genes, for iron
homeostasis. Increased expression of c-Myc indicates a loss of metabolic 6.3. Feeding behaviour
and growth control associated with the transformation of a normal cell
[37]. Feeding is essential. Table 5 described that various levels of noise
alter feeding behaviors’. Acute stress activates sympathetic arousal and
6. Noise and behavior GC release supports behavioral, automatic, and endocrinological
changes that promote energy mobilization such as increased cardiac
There is increasing evidence that noise stress impairs cognition output, blood pressure, gluconeogenesis, triglyceride levels, and re­
[73], motor coordination, eating, and lactational behavior in experi­ direction of blood flow to fuel the muscles, heart and the brain. Chronic
mental animals (Table 5). The adverse effect of noise includes meta­ exposures of stress can dysregulate the HPA axis, which affects energy
bolic and anatomical changes in neurons, reduced dendritic count, metabolism and feeding behavior [80].
impaired memory, cognition, and locomotor activity [75].
6.4. Fear and anxiety
6.1. Cognition
The bed nucleus of the stria terminalis and amygdala receive pro­
Fink defined cognition as “a healthy brain is one that can perform jections from the basolateral amygdala and project, in turn, to

Table 5.
Noise stress and neurotransmitter modulation.
S.No Model Noise Stress Methods Neurotransmitters Brain Regions Reference

1 Wistar albino/Male Acute (100dB- 30 mins / 1 Day) Spectrophotometer Acetylcholine Decreased in CC, CS, HY, HI [61]
Acetylcholinesterase Increased in CC, CS, HY, HI
2 Wistar albino/Male Sub acute stress HPLC Norepinephrine Increased in HY [75]
(100 dB- 4 h / 15 Days Dopamine
Sertonin
3 Wistar albino/Male Sub acute stress HPLC Norepinephrine Increased in CB, CS [76]
(100 dB- 4 h / 15 Days Dopamine
Sertonin
4 Sprague–Dawley /Male Chronic stress HPLC-EC System Glutamate Increased in Hemicerebrum [13]
100 dB – 4 h / 30 Days
GABA Decreased in Hemicerebrum
Asp, Gly No significant channge
5 Sprague–Dawley /Male 100 dB – 15, 30, 90 and 240 mins Lowry's Method [3H]5-HT Decreased in HY, HI, CC [43]
6 Wistar albino/Male Sub acute stress HPLC Norepinephrine Increased in CC, CB, CS, HY, HI, P [57]
(100 dB- 4 h / 15 Days Epinephrine
Dopamine Increased in CC, CB, HY, HI
5-HT Increased in CC, HY, P, CS

HPLC - High Performance Liquid Chromatography, HPLC-EC - High Performance Liquid Chromatography- Electrochemical Detection, P-Pons.

8
A. Arjunan and R. Rajan Physiology & Behavior 227 (2020) 113136

downstream target areas that mediate many of the behavioral, auto­ neuroanatomical and neurophysiological functions in Brain [45].
nomic, and electrophysiological consequences of fear and anxiety. Table 2 shows the increased acetylcholine and glutamate level in
Enormous studies denoted that noise increases anxiety-like behavior. hemicerebrum during the noise. Due to altered neurotransmitter levels
High level of glucocorticoids increases the excitation of amygdala in PFC, it undergoes neurochemical changes and morphological
neurons by decreasing the GABA level and also increases the cytosolic changes including reduced dendrite density and branches.
calcium which triggers cytoarchitectural changes in BLA neurons [44]. Studies in various animals have reported that noise stress results in
the damage to the hippocampus, a brain area involved in cognition
7. Epigenetics and noise [73]. The Hippocampus is well known as the “cognitive arm” of the
limbic system [20] and also for declarative or explicit memory espe­
Epigenetics is considered to be a link between environmental ex­ cially in the conscious or voluntary recollection of information [6].
posure and human diseases. Epigenetics refers to changes that alter Chronic exposure to a high level of CORT down-regulates the GLUCORT
gene transcription in the absence of direct alterations to the genetic receptors and increases the sensitivity of HPA axis – temporal, spatial
sequence. Limited studies explained the several environmental pollu­ aspects of skill movement, posture, balance coordination, affect neu­
tants such as toxic metals, pesticides, noise can induce aberrant DNA ronal metabolism, cell survival, physiology, neuronal morphology. The
methylation and histone modification in gene expression regulation hippocampus, mPFC, the amygdala is an essential component of the
[68]. Chronic exposure to stress leads to alteration of the expression of neuronal circuit mediating stress. The amygdale provides a primary link
longevity genes such as sirtuin-1, p53, thioredoxin-interacting protein, between subcortical areas that responsible for fear and cerebral cortical
HSP 70 in the rat hippocampus [58]. Guo et al. report that exposure of receiving sensory information about the external environment [29].
noise stress (70–75 dB) for short term and long term in Wistar rats is Joseph et al. reported that chronic exposure of the limbic system in the
associated with aberrant DNA methylation of Comt (Inferior colliculus), brain to the adrenal steroids has been correlated with neuropsychiatric,
Mc2r (hippocampus), BDNF and LINE-1 (Medulla oblongata) [25]. neurophysiologic, neurotransmitter, and neuroanatomical changes [6].

8. Discussion 9. Conclusion

Stress is a fundamental perception that discerns both life and evo­ Stress is an unavoidable circumstance in our life. Due to urbaniza­
lution. In life, stress affects social, psychological, and physiological tion and modern lifestyle noise pollution/ stress has increased tre­
environments which can simultaneously impinge on wellbeing [35]. mendously in our day to day life. Noise becomes a hazard to the phy­
Especially social factors may threaten the homeostasis. Selye also sup­ sical, mental, and social wellbeing of an individual who works in a
ported and proposed the theory that various external stimuli might noisy environment or to those who are vulnerable to exposure to noise.
generate undifferentiated physiological as well as psychological well­ Various studies have reported that chronic exposure to noise deterio­
being [60]. All creatures face threats to homeostasis, which are met rates brain function and may lead to neurodegenerative disease.
with adaptive responses. And also when an individual is exposed to Reduction of the impact of noise on health can be done by using various
acute stress adaptive strategies can be adopted which typically do not preventive measures such as earplugs, planting trees, regular main­
impose a health burden. In contrast, if stressors are chronic, it may lead tenance of vehicles, and development of a therapeutic intervention for
to diseases [59]. noise-induced neurodegenerative diseases
Exposure of stress is thought to precipitate or exacerbate several
psychiatric disorders, including schizophrenia, depression, and post- References
traumatic stress disorders [3,75]. In this review, we have documented
the associations between Noise stress and brain and have described how [1] H. Ageta, S. Ikegami, M. Miura, M. Masuda, R. Migishima, T. Hino, N. Takashima,
noise deteriorates the brain functions. Exposure to continuous noise of A. Murayama, H. Sugino, M. Setou, S. Kida, M. Yokoyama, Y. Hasegawa,
K. Tsuchida, T. Aosaki, K. Inokuchi, Activin plays a key role in the maintenance of
85–90 dBA, chronic in industrial settings, can lead to loss of hearing, long-term memory and late-LTP, Learn. Memory (2010), 10.1101/lm.16659010.
with an increase in the threshold of hearing sensitivity, and can also [2] P. Alario, A. Gamallo, M.J. Beato, G. Trancho, Body weight gain, food intake and
cause non-auditory health effects [69]. Oxidation-reduction (redox) adrenal development in chronic noise stressed rats, Physiol. Behav. (1987), 10.
1016/0031-9384(87)90181-8.
homeostasis, like pH control, is central to life. Redox processes pervade [3] A. AT, G.-R. PS, Noise stress impairs prefrontal cortical cognitive function in
practically all fundamental processes; from bioenergetics to metabolism monkeys: evidence for a hyperdopaminergic mechanism, Arch. Gen. Psychiatry
and life functions [62]. A balance between free radicals and anti­ (1998).
[4] U. B, S. A.V., Z. P., M. R.T., Oxidative stress and neurodegenerative diseases: a
oxidants is necessary for proper physiological function. If free radicals
review of upstream and downstream antioxidant therapeutic options, Curr.
exceed in range it can adversely alter lipids, proteins, and DNA and Neuropharmacol. (2009).
trigger several human diseases such as cardiovascular, inflammatory [5] M. Basner, W. Babisch, A. Davis, M. Brink, C. Clark, S. Janssen, S. Stansfeld,
Auditory and non-auditory effects of noise on health, Lancet (2014), 10.1016/
disease, cataract, cancer, and neurodegenerative diseases. Antioxidants
S0140-6736(13)61613-X.
prevent free radical-induced tissue damage by preventing the formation [6] J.K. Belanoff, K. Gross, A. Yager, A.F. Schatzberg, Corticosteroids and cognition, J.
of radicals, scavenging them, or by promoting their decomposition Psychiatr. Res. (2001), 10.1016/S0022-3956(01)00018-8.
[36]. [7] J. Brouček, Effect of noise on performance, stress, and behaviour of animals, Slovak
J. Anim. Sci. (2014).
From Table 1, studies denote that noise above 100 dB (acute, sub­ [8] M.A. Calcia, D.R. Bonsall, P.S. Bloomfield, S. Selvaraj, T. Barichello, O.D. Howes,
acute, and chronic) increased the oxidative stress and decreased the Stress and neuroinflammation: a systematic review of the effects of stress on mi­
antioxidant level in various regions of the brain. The increased oxida­ croglia and the implications for mental illness, Psychopharmacology (Berl.) (2016),
10.1007/s00213-016-4218-9.
tive stress is due to peroxidation of membrane lipids which affects a [9] S.N. Campbell, C. Zhang, L. Monte, A.D. Roe, K.C. Rice, Y. Taché, E. Masliah,
variety of functions resulting in increased membrane rigidity, decreased R.A. Rissman, Increased tau phosphorylation and aggregation in the hippocampus
activity of membrane-bound enzymes (e.g., sodium pump), impairment of mice overexpressing corticotropin-releasing factor, J. Alzheimer's Dis. (2015),
10.3233/JAD-141281.
of membrane receptors and altered permeability damage to phospho­ [10] L. Cheng, S.H. Wang, Y. Huang, X.M. Liao, The hippocampus may be more sus­
lipids, radicals also can directly attack membrane proteins and induce ceptible to environmental noise than the auditory cortex, Hear. Res. (2016), 10.
lipid-protein and protein-protein cross-linking, all of which contribute 1016/j.heares.2016.01.001.
[11] D.W. Choi, Excitotoxic cell death, J. Neurobiol. (1992), 10.1002/neu.480230915.
to altered membrane integrity [71]. [12] B. Cui, K. Li, Z. Gai, X. She, N. Zhang, C. Xu, X. Chen, G. An, Q. Ma, R. Wang,
Stress activates the hypothalamic-pituitary-adrenal axis, secretion Chronic noise exposure acts cumulatively to exacerbate Alzheimer's disease-like
of corticosterone in adrenal glands which crosses the blood-brain bar­ amyloid-β pathology and neuroinflammation in the rat hippocampus, Sci. Rep.
(2015), 10.1038/srep12943.
rier and interacts with the neurons and neuroglial cells which alters the

9
A. Arjunan and R. Rajan Physiology & Behavior 227 (2020) 113136

[13] B. Cui, M. Wu, X. She, Effects of chronic noise exposure on spatial learning and effects of buspirone and tianeptine, Eur. J. Pharmacol. (1993), 10.1016/0014-
memory of rats in relation to neurotransmitters and NMDAR2B alteration in the 2999(93)90211-Y.
hippocampus, J. Occup. Health (2009), 10.1539/joh.L8084. [44] R. Mitra, R.M. Sapolsky, Acute corticosterone treatment is sufficient to induce an­
[14] B. Cui, L. Zhu, X. She, M. Wu, Q. Ma, T. Wang, N. Zhang, C. Xu, X. Chen, G. An, xiety and amygdaloid dendritic hypertrophy, Proc. Natl. Acad. Sci. (2008), 10.
H. Liu, Chronic noise exposure causes persistence of tau hyperphosphorylation and 1073/pnas.0705615105.
formation of NFT tau in the rat hippocampus and prefrontal cortex, Exp. Neurol. [45] F. Mora, G. Segovia, A. Del Arco, M. De Blas, P. Garrido, Stress, neurotransmitters,
(2012), 10.1016/j.expneurol.2012.08.028. corticosterone and body-brain integration, Brain Res. (2012), 10.1016/j.brainres.
[15] E. Daniel, Noise and hearing loss: a review, J. Sch. Health (2007), 10.1111/j.1746- 2011.12.049.
1561.2007.00197.x. [46] C. Moret, M. Briley, The importance of norepinephrine in depression,
[16] A. Das, D. Rai, M. Dikshit, G. Palit, C. Nath, Nature of stress: differential effects on Neuropsychiatr. Dis. Treat. (2011), 10.2147/NDT.S19619.
brain acetylcholinesterase activity and memory in rats, Life Sci. (2005), 10.1016/j. [47] P. Nuss, Anxiety disorders and GABA neurotransmission: a disturbance of mod­
lfs.2005.02.020. ulation, Neuropsychiatr. Dis. Treat. (2015), 10.2147/NDT.S58841.
[17] X.-F. Ding, Y. Liu, Z.-Y. Yan, X.-J. Li, Q.-Y. Ma, Z.-Y. Jin, Y. Liu, Y.-H. Li, Y.-Y. Liu, [48] I. OA, J. v, V. OS, Mitigative effects of antioxidants in noise stress, J. Clin. Nutr.
Z. Xue, J.-X. Chen, Z.-P. Lv, Involvement of normalized glial fibrillary acidic protein Diet. (2017), 10.4172/2472-1921.100056.
expression in the hippocampi in antidepressant-like effects of Xiaoyaosan on [49] O. Otitoju, I.N.E. Onwurah, G.T.O. Otitoju, C.E. Ugwu, E. State, Oxidative stress and
chronically stressed mice, Evid.-Based Complement. Altern. Med. (2017), 10.1155/ superoxide dismutase activity in brain of rats fed with diet containing permethrin
2017/1960584. Olawale, Biokemistri (2008).
[18] S. Dronjak, L. Gavrilovic, Effects of stress on catecholamine stores in central and [50] N. Parameswaran, S. Patial, Tumor necrosis factor-α signaling in macrophages, Crit.
peripheral tissues of long-term socially isolated rats, Braz. J. Med. Biol. Res. (2006), Rev. Eukaryot. Gene Expr. (2010).
10.1590/S0100-879x2006000600011. [51] C. Pavlides, Y. Watanabe, B.S. McEwen, Effects of glucocorticoids on hippocampal
[19] A.J. Dunn, N.R. Kramarcy, Neurochemical responses in stress: relationships be­ long-term potentiation, Hippocampus (1993), 10.1002/hipo.450030210.
tween the hypothalamic-pituitary-adrenal and catecholamine systems, Drugs [52] L.A. Pham-Huy, H. He, C. Pham-Huy, Free radicals, antioxidants in disease and
Neurotransm. Behav. (2013), 10.1007/978-1-4615-7178-0_8. health, Int. J. Biomed. Sci. (2008).
[20] H. Eichenbaum, T. Otto, N.J. Cohen, The hippocampus—What does it do? Behav. [53] M.R. Picciotto, M.J. Higley, Y.S. Mineur, Acetylcholine as a Neuromodulator:
Neural Biol. (2004), 10.1016/0163-1047(92)90724-i. cholinergic Signaling Shapes Nervous System Function and Behavior, Neuron
[21] E. Eraslan, I. Akyazi, E. Ergül-Ekiz, E. Matur, Noise stress changes mRNA expres­ (2012), 10.1016/j.neuron.2012.08.036.
sions of corticotropin-releasing hormone, its receptors in amygdala, and anxiety- [54] W. Rasbury, K. Shemberg, The effects of aversive levels of white noise on con­
related behaviors, Noise Health (2015), 10.4103/1463-1741.155838. summatory behavior, Psychon. Sci. (1971), 10.3758/BF03332550.
[22] D.J. Fink, What is a safe noise level for the public? Am. J. Public Health (2017), 10. [55] R. Sah, F. Galeffi, R. Ahrens, G. Jordan, R.D. Schwartz-Bloom, Modulation of the
2105/AJPH.2016.303527. GABAA-gated chloride channel by reactive oxygen species, J. Neurochem. (2002),
[23] Z. Gai, K. Li, H. Sun, X. She, B. Cui, R. Wang, Effects of chronic noise on mRNA and 10.1046/j.0022-3042.2001.00706.x.
protein expression of CRF family molecules and its relationship with p-tau in the rat [56] A. Säljö, F. Bao, J. Shi, A. Hamberger, H.-A. Hansson, K.G. Haglid, Expression of c-
prefrontal cortex, J. Neurol. Sci. (2016), 10.1016/j.jns.2016.07.049. Fos and c-Myc and deposition of β -APP in neurons in the adult rat brain as a result
[24] M.M. Gaschler, B.R. Stockwell, Lipid peroxidation in cell death, Biochem. Biophys. of exposure to short-lasting impulse noise, J. Neurotrauma (2002), 10.1089/
Res. Commun. (2017), 10.1016/j.bbrc.2016.10.086. 089771502753594945.
[25] L. Guo, P.hui Li, H. Li, E. Colicino, S. Colicino, Y. Wen, R. Zhang, X. Feng, [57] J. Samson, R.S. Devi, R. Ravindran, M. Senthilvelan, Effect of noise stress on free
T.M. Barrow, A. Cayir, A.A. Baccarelli, H.M. Byun, Effects of environmental noise radical scavenging enzymes in brain, Environ. Toxicol. Pharmacol. (2005), 10.
exposure on DNA methylation in the brain and metabolic health, Environ. Res. 1016/j.etap.2004.12.059.
(2017), 10.1016/j.envres.2016.11.017. [58] A.C. Sánchez-Hidalgo, M.F. Muñoz, A.J. Herrera, A.M. Espinosa-Oliva, R. Stowell,
[26] E.D. Hall, Glucocorticoid effects on central nervous excitability and synaptic A. Ayala, A. Machado, J.L. Venero, R.M. De Pablos, Chronic stress alters the ex­
transmission, Int. Rev. Neurobiol. (1982), 10.1016/S0074-7742(08)60625-X. pression levels of longevity-related genes in the rat hippocampus, Neurochem. Int.
[27] D. Henderson, E.C. Bielefeld, K.C. Harris, B.H. Hu, The role of oxidative stress in (2016), 10.1016/j.neuint.2016.04.009.
noise-induced hearing loss, Ear Hear (2006), 10.1097/01.aud.0000191942. [59] N. Schneiderman, G. Ironson, S.D. Siegel, Stress and Health: psychological,
36672.f3. Behavioral, and Biological Determinants, Annu. Rev. Clin. Psychol. (2004), 10.
[28] A. Imperato, S. Puglisi-Allegra, P. Casolini, L. Angelucci, Changes in brain dopamine 1146/annurev.clinpsy.1.102803.144141.
and acetylcholine release during and following stress are independent of the pi­ [60] H. Selye, The Stress of the Life, McGraw-Hill, 1956, 10.1177/0098628316662768.
tuitary-adrenocortical axis, Brain Res. (1991), 10.1016/0006-8993(91)90384-8. [61] K. Sembulingam, P. Sembulingam, A. Namasivayam, Effect of Ocimum sanctum
[29] Z. Jafari, B.E. Kolb, M.H. Mohajerani, Chronic traffic noise stress accelerates brain Linn on the changes in central cholinergic system induced by acute noise stress, J.
impairment and cognitive decline in mice, Exp. Neurol. (2018), 10.1016/j. Ethnopharmacol. (2005), 10.1016/j.jep.2004.09.047.
expneurol.2018.06.011. [62] H. Sies, Hydrogen peroxide as a central redox signaling molecule in physiological
[30] P.M. Kidd, Glutathione: systemic protectant against oxidative and free radical da­ oxidative stress: oxidative eustress, Redox Biol. (2017), 10.1016/j.redox.2016.12.
mage, Alternat. Med. Rev. (1997). 035.
[31] G.H. Kim, J.E. Kim, S.J. Rhie, S. Yoon, The role of oxidative stress in neurode­ [63] N.A. Simonian, J.T. Coyle, Oxidative stress in neurodegenerative diseases [Review],
generative diseases, Exp. Neurobiol. (2015), 10.5607/en.2015.24.4.325. Ann. Rev. Pharmacol. Toxicol. (1996).
[32] E. Koc, S. Sahin, A. Ersoy, U. Duzgun, B. Acar, A. Ilhan, Possible effects of rosu­ [64] H. Soreq, R. Yirmiya, O. Cohen, D. Glick, Chapter 4.10 Acetylcholinesterase as a
vastatin on noise-induced oxidative stress in rat brain, Noise Health (2014), 10. window onto stress responses, Techn. Behav. Neural Sci. (2005), 10.1016/S0921-
4103/1463-1741.127849. 0709(05)80032-X.
[33] H. Krebs, M. Macht, P. Weyers, H.G. Weijers, W. Janke, Effects of stressful noise on [65] L.T. Spencer, J.D Bancroft, Tissue processing, Bancroft's Theory and Practice of
eating and non-eating behavior in rats, Appetite (1996), 10.1006/appe.1996.0015. Histological Techniques, (2013).
[34] A. Kumar, B. Yegla, T.C. Foster, Redox signaling in neurotransmission and cognition [66] R. Srikumar, N.J. Parthasarathy, S. Manikandan, G.S. Narayanan, R. Sheeladevi,
during aging, Antioxid. Redox Signal. (2017), 10.1089/ars.2017.7111. Effect of Triphala on oxidative stress and on cell-mediated immune response against
[35] N. Lin, W.M. Ensel, Life stress and health: stressors and resources, Am. Sociol. Rev. noise stress in rats, Mol. Cell. Biochem. (2006), 10.1007/s11010-006-2271-0.
(2006), 10.2307/2095612. [67] S. Srinivasan, W. Wankhar, S. Rathinasamy, R. Rajan, Neuroprotective effects of
[36] V. Lobo, A. Patil, A. Phatak, N. Chandra, Free radicals, antioxidants and functional Indigofera tinctoria on noise stress affected Wistar albino rat brain, J. Appl. Pharm.
foods: impact on human health, Pharmacogn. Rev. (2010), 10.4103/0973-7847. Sci. (2015), 10.7324/JAPS.2015.50609.
70902. [68] A.M. Stankiewicz, A.H. Swiergiel, P. Lisowski, Epigenetics of stress adaptations in
[37] H. Lodish, A. Berk, S.L. Zipursky, et al., Molecular Cell Biology, W.H. Freeman, the brain, Brain Res. Bull. (2013), 10.1016/j.brainresbull.2013.07.003.
2000 5th Ed 10.1016/S1470-8175(01)00023-6. [69] S.A. Stansfeld, M.P. Matheson, Noise pollution: non-auditory effects on health, Br.
[38] S. Loganathan, S. Srinivasan, W. Wankhar, A.K. Chodhary, S. Rathinasamy, Noise Med. Bull. (2003), 10.1093/bmb/ldg033.
exposure effect on selective inflammatory markers of Wistar Albino rats immune [70] R. Sultana, M. Perluigi, D.A. Butterfield, Protein oxidation and lipid peroxidation in
organs and role of Scoparia dulcis, J. Biol. Active Prod. Nat. (2017), 10.1080/ brain of subjects with Alzheimer's disease: insights into mechanism of neurode­
22311866.2017.1334587. generation from redox proteomics, Antioxid. Redox Signal. (2006), 10.1089/ars.
[39] S. Manikandan, R.S. Devi, Antioxidant property of α-asarone against noise-stress- 2006.8.2021.
induced changes in different regions of rat brain, Pharmacol. Res. (2005), 10.1016/ [71] R. Sultana, M. Perluigi, D.A. Butterfield, Lipid peroxidation triggers neurodegen­
j.phrs.2005.07.007. eration: a redox proteomics view into the Alzheimer disease brain, Free Radic. Biol.
[40] S. Manikandan, M.K. Padma, R. Srikumar, N. Jeya Parthasarathy, A. Muthuvel, Med. (2013), 10.1016/j.freeradbiomed.2012.09.027.
R.S. Devi, Effects of chronic noise stress on spatial memory of rats in relation to [72] C.F. Valenzuela, M.P. Puglia, S. Zucca, Focus on: neurotransmitter systems, Alcohol
neuronal dendritic alteration and free radical-imbalance in hippocampus and Res. Health (2011).
medial prefrontal cortex, Neurosci. Lett. (2006), 10.1016/j.neulet.2006.01.037. [73] S. Wang, Y. Yu, Y. Feng, F. Zou, X. Zhang, J. Huang, Y. Zhang, X. Zheng, X.F. Huang,
[41] M.K. McCoy, M.G. Tansey, TNF signaling inhibition in the CNS: implications for Y. Zhu, Y. Liu, Protective effect of the orientin on noise-induced cognitive impair­
normal brain function and neurodegenerative disease, J. Neuroinflammation ments in mice, Behav. Brain Res. (2016), 10.1016/j.bbr.2015.09.024.
(2008), 10.1186/1742-2094-5-45. [74] D. Wankhar, R. Sheela Devi, I. Ashok, Emblica officinalis outcome on noise stress
[42] B.S. McEwen, J.M. Weiss, L.S. Schwartz, Selective retention of corticosterone by induced behavioural changes in wistar albino rats, Biomed. Prev. Nutr. (2014), 10.
limbic structures in rat brain [18], Nature (1968), 10.1038/220911a0. 1016/j.bionut.2013.12.011.
[43] T. Mennini, C. Taddei, A. Codegoni, M. Gobbi, S. Garattini, Acute noise stress re­ [75] W. Wankhar, S. Srinivasan, R. Rajan, S. Rathinasamy, Efficacy of Scoparia dulcis on
duces [3H]5-hydroxytryptamine uptake in rat brain synaptosomes: protective noise-evoked neural cytokines and neurotransmitter level in experimental rat

10
A. Arjunan and R. Rajan Physiology & Behavior 227 (2020) 113136

model, J. Biol. Active Prod. Nat. (2016), 10.1080/22311866.2016.1164623. C.D. Ingram, S.L. Lightman, Endocrine and behavioural responses to noise stress:­
[76] W. Wankhar, S. Srinivasan, L. Sundareswaran, D. Wankhar, R. Rajan, R. Sheeladevi, comparison of virgin and lactating female ratsduring non-disrupted maternal ac­
Role of Scoparia dulcis linn on noise-induced nitric oxide synthase (NOS) expression tivity, J. Neuroendocrinol. (2003), 10.1046/j.1365-2826.1997.00587.x.
and neurotransmitter assessment on motor function in Wistar albino rats, Biomed. [79] B. Wright, E. Peters, U. Ettinger, E. Kuipers, V. Kumari, Understanding noise stress-
Pharmacother. (2017), 10.1016/j.biopha.2016.12.054. induced cognitive impairment in healthy adults and its implications for schizo­
[77] J.F. Wilson, M.B. Cantor, Noise-induced eating in rats facilitated by prior tail pinch phrenia, Noise Health (2014), 10.4103/1463-1741.134917.
experience, Physiol. Behav. (1986), 10.1016/0031-9384(86)90278-7. [80] Y.H.C. Yau, M.N. Potenza, Stress and eating behaviors, Minerva Endocrinologica,
[78] R.J. Windle, S. Wood, N. Shanks, P. Perks, G.L. Conde, A.P.C. Da Costa, 2013.

11

You might also like