Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Eur J Neurosci ; 59(10): 2596-2615, 2024 May.
Article in English | MEDLINE | ID: mdl-38441248

ABSTRACT

Auditory deprivation following congenital/pre-lingual deafness (C/PD) can drastically affect brain development and its functional organisation. This systematic review intends to extend current knowledge of the impact of C/PD and deafness duration on brain resting-state networks (RSNs), review changes in RSNs and spoken language outcomes post-cochlear implant (CI) and draw conclusions for future research. The systematic literature search followed the PRISMA guideline. Two independent reviewers searched four electronic databases using combined keywords: 'auditory deprivation', 'congenital/prelingual deafness', 'resting-state functional connectivity' (RSFC), 'resting-state fMRI' and 'cochlear implant'. Seventeen studies (16 cross-sectional and one longitudinal) met the inclusion criteria. Using the Crowe Critical Appraisal Tool, the publications' quality was rated between 65.0% and 92.5% (mean: 84.10%), ≥80% in 13 out of 17 studies. A few studies were deficient in sampling and/or ethical considerations. According to the findings, early auditory deprivation results in enhanced RSFC between the auditory network and brain networks involved in non-verbal communication, and high levels of spontaneous neural activity in the auditory cortex before CI are evidence of occupied auditory cortical areas with other sensory modalities (cross-modal plasticity) and sub-optimal CI outcomes. Overall, current evidence supports the idea that moreover intramodal and cross-modal plasticity, the entire brain adaptation following auditory deprivation contributes to spoken language development and compensatory behaviours.


Subject(s)
Cochlear Implantation , Deafness , Humans , Deafness/physiopathology , Cochlear Implantation/methods , Brain/physiopathology , Brain/diagnostic imaging , Brain/physiology , Nerve Net/physiopathology , Nerve Net/diagnostic imaging , Magnetic Resonance Imaging , Auditory Cortex/physiopathology , Auditory Cortex/diagnostic imaging , Cochlear Implants , Treatment Outcome
2.
Exp Neurol ; 368: 114498, 2023 10.
Article in English | MEDLINE | ID: mdl-37536439

ABSTRACT

Alzheimer's disease (AD) is associated with cerebral plaques and tangles, reduced synapse number, and shrinkage in several brain areas and these morphological effects are associated with the onset of compromised cognitive, motor, and anxiety-like behaviours. The appearance of both anatomical and behavioural symptoms is worsened by stress. The focus of this study was to examine the effect of neonatal tactile stimulation on AD-like behavioural and neurological symptoms on APP NL-G-F/NL-G-F mice, a mouse model of AD, who have been gestationally stressed. Our findings indicate that neonatal tactile stimulation improves cognition, motor skills, and anxiety-like symptoms in both gestationally stressed and non-stressed adult APP mice and that these alterations are associated with reduced Aß plaque formation. Thus, tactile stimulation appears to be a promising non-invasive preventative strategy for slowing the onset of dementia in aging animals.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Mice, Transgenic , Brain/metabolism , Disease Models, Animal
3.
Exp Neurol ; 365: 114413, 2023 07.
Article in English | MEDLINE | ID: mdl-37075972

ABSTRACT

Current evidence supports the link between hearing loss and Alzheimer's disease (AD). However, few studies report the hearing status of AD mice compared to wild-type mice. This study aimed to compare hearing thresholds and short-term memory (STM) performance of an AD (APPNL-G-F) mouse model of amyloid-beta (Aꞵ) pathology with C57BL/6 J and CBA/CaJ mice across age. The auditory brainstem response (ABR) test, using click and five tone-burst (TB) stimuli, was recorded at 2, 4, 6, 9, and 12 months. The novel object recognition (NOR) test, a measure of STM, was conducted at 6 and 12 months. While hearing thresholds were almost preserved in CBA/CaJ mice, they were not recorded at high frequencies with age in C57BL/6 J and AD mice, leading to island hearing (severe to profound hearing loss) at 9 and 12 months. AD mice showed increased hearing thresholds in TB8 and TB16 kHz at 6 and 9 months compared to C57BL/6 J mice. NOR findings were evidence of impaired STM in both C57BL/6 J and AD mice relative to CBA/CaJ mice, and a relationship was found between hearing thresholds and NOR measures in three groups. The findings were in support of the link between the degree of hearing loss and impaired STM.


Subject(s)
Alzheimer Disease , Deafness , Hearing Loss , Mice , Animals , Alzheimer Disease/complications , Memory, Short-Term , Mice, Inbred C57BL , Mice, Inbred CBA , Amyloid beta-Peptides , Hearing Loss/etiology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem
4.
Synapse ; 77(2): e22257, 2023 03.
Article in English | MEDLINE | ID: mdl-36255152

ABSTRACT

Alzheimer's disease (AD) is one of the largest health crises in the world. There are limited pharmaceutical interventions to treat AD, however, and most of the treatment options are not for cure or prevention, but rather to slow down the progression of the disease. The aim of this study was to examine the effect of tactile stimulation (TS) on AD-like symptoms and pathology in APPNL-G-F/NL-G-F mice, a mouse model of AD. The results show that TS reduces the AD-like symptoms on tests of cognition, motor, and anxiety-like behaviors and these improvements in behavior are associated with reduced AD pathology in APP mice. Thus, TS appears to be a promising noninvasive strategy for slowing the onset of dementia in aging animals.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Mice , Animals , Alzheimer Disease/therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides , Cognitive Dysfunction/pathology , Mice, Transgenic , Cognition , Anxiety/therapy , Disease Models, Animal , Amyloid beta-Protein Precursor
5.
Ear Hear ; 43(6): 1643-1652, 2022.
Article in English | MEDLINE | ID: mdl-35612517

ABSTRACT

OBJECTIVES: Current evidence supports the growing application of extended high-frequency (EHF: 9 to 20 kHz) audiometry in hearing research, which likely results from the high vulnerability of this frequency region to damage induced by known auditory risk factors. The present systematic review and meta-analysis were performed to investigate whether adults with a normal audiogram and tinnitus show increased EHF hearing thresholds relative to control peers. DESIGN: A comprehensive search was undertaken on electronic databases consisting of PubMed, ScienceDirect, Wiley, and Google Scholar using combined keywords: "tinnitus," "extended high frequency," "normal audiogram," and "hidden hearing loss." RESULTS: From 261 articles found by searching databases, nine studies met the inclusion criteria for the meta-analysis. A significant difference was observed between tinnitus and control groups in the effect size analysis of hearing thresholds at 10, 12.5, 14, 16, and 18 kHz ( p ≤ 0.001), and the I-square heterogeneity analysis was below 50% in all studies ( p ≥ 0.131). Visual inspection by the Funnel plot and Egger's regression test ( p ≥ 0.211) also exhibited no publication bias in the meta-analyses. CONCLUSIONS: Our findings are in support of the idea that in most cases, tinnitus is associated with some degree of cochlear mechanical dysfunction, which may not be detected by conventional audiometry alone. This finding underscores the significance of EHF audiometry in clinical practice, which may help both early identification of individuals susceptible to developing tinnitus and reduce the number of new cases through preventive counseling programs.


Subject(s)
Hearing Loss , Tinnitus , Adult , Humans , Tinnitus/psychology , Auditory Threshold , Hearing , Audiometry/methods , Audiometry, Pure-Tone
7.
Eur Arch Otorhinolaryngol ; 279(11): 5161-5170, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35359185

ABSTRACT

PURPOSE: Whereas chronic noise exposure (CNE) is a known risk factor for tinnitus, little is known about how a history of CNE impacts tinnitus characteristics and its comorbid symptoms. METHODS: Seventy-five participants with chronic tinnitus (59m/16f, 22-78 years, 48 with sensory-neural hearing loss, and 27 with a normal audiogram) including 43 individuals with (Tin-CNE group) and 32 without (Tin group) a history of long-term occupational noise exposure were studied. Tinnitus characteristics were rated by a visual analog scale, and tinnitus comorbid symptoms were scored using self-assessment questionnaires. RESULTS: The Tin-CNE group showed reduced uncomfortable loudness level (ULL), sound tolerance, and quality of life (QoL), and increased tinnitus loudness, tinnitus handicap, anxiety, depression, insomnia severity, and tinnitus annoyance scores compared to the Tin group. Higher tinnitus loudness and a lower anxiety score were observed in participants with hearing loss relative to those without. Using a stepwise regression model also showed that tinnitus-related characteristics, hyperacusis, and tinnitus comorbid symptoms enhance one another. CONCLUSIONS: The findings were in support of accumulative evidence indicating the adverse auditory and non-auditory effects of CNE, including exacerbated sound intolerance and tinnitus-related psychiatric symptoms. The results also showed that tinnitus alone can affect mental health regardless of hearing loss.


Subject(s)
Hearing Loss , Noise, Occupational , Tinnitus , Humans , Hyperacusis/epidemiology , Hyperacusis/etiology , Hyperacusis/psychology , Mental Health , Noise, Occupational/adverse effects , Quality of Life , Tinnitus/epidemiology , Tinnitus/etiology , Tinnitus/psychology
8.
Can J Neurol Sci ; 49(2): 184-195, 2022 03.
Article in English | MEDLINE | ID: mdl-33843530

ABSTRACT

OBJECTIVES: Extensive studies indicate that severe acute respiratory syndrome coronavirus (SARS-CoV-2) involves human sensory systems. A lack of discussion, however, exists given the auditory-vestibular system involvement in CoV disease 2019 (COVID-19). The present systematic review and meta-analysis were performed to determine the event rate (ER) of hearing loss, tinnitus, and dizziness caused by SARS-CoV-2. METHODS: Databases (PubMed, ScienceDirect, Wiley) and World Health Organization updates were searched using combined keywords: 'COVID-19,' 'SARS-CoV-2,' 'pandemic,' 'auditory dysfunction,' 'hearing loss,' 'tinnitus,' 'vestibular dysfunction,' 'dizziness,' 'vertigo,' and 'otologic symptoms.' RESULTS: Twelve papers met the eligibility criteria and were included in the study. These papers were single group prospective, cross-sectional, or retrospective studies on otolaryngologic, neurologic, or general clinical symptoms of COVID-19 and had used subjective assessments for data collection (case histories/medical records). The results of the meta-analysis demonstrate that the ER of hearing loss (3.1%, CIs: 0.01-0.09), tinnitus (4.5%, CIs: 0.012-0.153), and dizziness (12.2%, CIs: 0.070-0.204) is statistically significant in patients with COVID-19 (Z ≤ -4.469, p ≤ 0.001). CONCLUSIONS: COVID-19 can cause hearing loss, tinnitus, and dizziness. These findings, however, should be interpreted with caution given insufficient evidence and heterogeneity among studies. Well-designed studies and follow-up assessments on otologic symptoms of SARS-CoV-2 using standard objective tests are recommended.


Subject(s)
COVID-19 , Hearing Loss , Tinnitus , COVID-19/complications , Cross-Sectional Studies , Dizziness/epidemiology , Dizziness/etiology , Hearing Loss/complications , Humans , Prospective Studies , Retrospective Studies , SARS-CoV-2 , Tinnitus/epidemiology , Tinnitus/etiology , Vertigo/diagnosis
9.
Ann N Y Acad Sci ; 1505(1): 8-22, 2021 12.
Article in English | MEDLINE | ID: mdl-34309857

ABSTRACT

The past decade marked the beginning of the use of resting-state functional connectivity (RSFC) imaging in bilingualism studies. This paper intends to review the latest evidence of changes in RSFC in language and cognitive control networks in bilinguals during adulthood, aging, and early Alzheimer's disease, which can add to our understanding of brain functional reshaping in the context of second language (L2) acquisition. Because of high variability in bilingual experience, recent studies mostly focus on the role of the main aspects of bilingual experience (age of acquisition (AoA), language proficiency, and language usage) on intrinsic functional connectivity (FC). Existing evidence accounts for stronger FC in simultaneous rather than sequential bilinguals in language and control networks, and the modulation of the AoA impact by language proficiency and usage. Studies on older bilingual adults show stronger FC in language and frontoparietal networks and preserved FC in posterior brain regions, which can protect the brain against cognitive decline and neurodegenerative processes. Altered RSFC in language and control networks subsequent to L2 training programs also is associated with improved global cognition in older adults. This review ends with a brief discussion of potential confounding factors in bilingualism research and conclusions and suggestions for future research.


Subject(s)
Aging , Alzheimer Disease/diagnostic imaging , Brain/diagnostic imaging , Magnetic Resonance Imaging/methods , Multilingualism , Nerve Net/diagnostic imaging , Adult , Aging/physiology , Alzheimer Disease/physiopathology , Brain/physiology , Humans , Nerve Net/physiology
10.
Neurobiol Stress ; 15: 100345, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34124321

ABSTRACT

Prenatal stress (PS) can impact fetal brain structure and function and contribute to higher vulnerability to neurodevelopmental and neuropsychiatric disorders. To understand how PS alters evoked and spontaneous neocortical activity and intrinsic brain functional connectivity, mesoscale voltage imaging was performed in adult C57BL/6NJ mice that had been exposed to auditory stress on gestational days 12-16, the age at which neocortex is developing. PS mice had a four-fold higher basal corticosterone level and reduced amplitude of cortical sensory-evoked responses to visual, auditory, whisker, forelimb, and hindlimb stimuli. Relative to control animals, PS led to a general reduction of resting-state functional connectivity, as well as reduced inter-modular connectivity, enhanced intra-modular connectivity, and altered frequency of auditory and forelimb spontaneous sensory motifs. These resting-state changes resulted in a cortical connectivity pattern featuring disjoint but tight modules and a decline in network efficiency. The findings demonstrate that cortical connectivity is sensitive to PS and exposed offspring may be at risk for adult stress-related neuropsychiatric disorders.

11.
Synapse ; 75(9): e22217, 2021 09.
Article in English | MEDLINE | ID: mdl-34120374

ABSTRACT

The epileptogenic-prone (FAST) and epileptogenic-resistant (SLOW) rat strains have become a valuable tool for investigating neural plasticity. The strains were generated by breeding the rats that required the fewest amygdala stimulations to elicit a stage-5 convulsive seizure (FAST) and rats requiring the most stimulations (SLOW). Previous studies have shown differences in behavior and amygdala physiology in the two strains. This study examined the dendritic morphology of pyramidal neurons in the brains of adult male and female rats of the two strains. The brains were stained with the Golgi-Cox method and the length and branching from layer III pyramidal cells were measured in parietal cortex (Zilles Par1), medial frontal cortex (Zilles Cg3), and orbitofrontal cortex (Zilles AID) in these two strains of rats. We observed significantly longer dendrites in Cg3 in the FAST group but longer dendrites in the SLOW group in AID and Par1. There was also a sex difference (M > F) in Par1 in both strains. These morphological differences can provide insights into the neurobiological basis of the behavioral differences and suggest that localized changes in the amygdala do not occur independently of changes in other brain regions, and especially prefrontal cortex.


Subject(s)
Kindling, Neurologic , Amygdala/physiology , Animals , Dendrites/physiology , Female , Kindling, Neurologic/physiology , Male , Neuronal Plasticity , Neurons , Prefrontal Cortex , Pyramidal Cells , Rats
12.
Ann N Y Acad Sci ; 1500(1): 17-33, 2021 09.
Article in English | MEDLINE | ID: mdl-34114212

ABSTRACT

Extensive evidence supports the association between age-related hearing loss (ARHL) and cognitive decline. It is, however, unknown whether a causal relationship exists between these two, or whether they both result from shared mechanisms. This paper intends to study this relationship through a comprehensive review of MRI findings as well as evidence of cellular alterations. Our review of structural MRI studies demonstrates that ARHL is independently linked to accelerated atrophy of total and regional brain volumes and reduced white matter integrity. Resting-state and task-based fMRI studies on ARHL also show changes in spontaneous neural activity and brain functional connectivity; and alterations in brain areas supporting auditory, language, cognitive, and affective processing independent of age, respectively. Although MRI findings support a causal relationship between ARHL and cognitive decline, the contribution of potential shared mechanisms should also be considered. In this regard, the review of cellular evidence indicates their role as possible common mechanisms underlying both age-related changes in hearing and cognition. Considering existing evidence, no single hypothesis can explain the link between ARHL and cognitive decline, and the contribution of both causal (i.e., the sensory hypothesis) and shared (i.e., the common cause hypothesis) mechanisms is expected.


Subject(s)
Aging , Brain/metabolism , Brain/physiopathology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Hearing Loss/etiology , Hearing Loss/metabolism , Aging/psychology , Animals , Biomarkers , Brain/diagnostic imaging , Disease Susceptibility , Hearing Loss/diagnosis , Humans , Magnetic Resonance Imaging , Mitochondria/genetics , Mitochondria/metabolism
13.
Synapse ; 75(4): e22192, 2021 04.
Article in English | MEDLINE | ID: mdl-33096582

ABSTRACT

Concerns are growing that exposure to environmental pollutants, such as traffic noise, might cause cognitive impairments and predispose individuals toward the development of Alzheimer's disease (AD) dementia. In this study in a knock-in mouse model of AD, we investigated how chronic traffic noise exposure (CTNE) impacts cognitive performance and amyloid-beta (Aß) pathology. A group of APPNL-G-F/NL-G-F mice was exposed to CTNE (70 dBA , 8 hr/day for 1 month) and compared with nonexposed counterparts. Following CTNE, an increase in hypothalamic-pituitary-adrenal (HPA) axis responsivity was observed by corticosterone assay of the blood. One month after CTNE, the CTNE group demonstrated impairments in cognitive and motor functions, and indications of anxiety-like behavior, relative to the control animals. The noise-exposed group also showed elevated Aß aggregation, as inferred by a greater number of plaques and larger average plaque size in various regions of the brain, including regions involved in stress regulation. The results support that noise-associated dysregulation of the neuroendocrine system as a potential risk factor for developing cognitive impairment and Aß pathology, which should be further investigated in human studies.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Noise, Transportation , Alzheimer Disease/pathology , Amyloid beta-Peptides , Amyloid beta-Protein Precursor , Animals , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Disease Models, Animal , Mice , Mice, Transgenic
14.
Otol Neurotol ; 41(10): 1316-1326, 2020 12.
Article in English | MEDLINE | ID: mdl-32810017

ABSTRACT

OBJECTIVE: Age-related hearing loss (ARHL) is the third most challenging disability in older adults. Noise is a known modifiable risk factor of ARHL, which can drive adverse health effects. Few large-scale studies, however, have shown how chronic noise exposure (CNE) impacts the progression of ARHL and tinnitus. STUDY DESIGN: Retrospective large-scale study. SETTING: Audiology clinical practice. PATIENTS: In this study, 928 individuals aged 30-100 years without (n=497) or with the experience of CNE (n=431) were compared in their hearing assessments and tinnitus. In order to only investigate the impact of CNE on ARHL and tinnitus, people with other risk factors of hearing loss were excluded from the study. INTERVENTION: Diagnostic. MAIN OUTCOME MEASURES: Noise damage was associated with a greater ARHL per age decades (pure-tone average(PTA)0.5-4kHz alterations 19.6-70.8 dB vs. 8.0-63.2 dB, ≤0.001), an acceleration of developing a significant ARHL at least by two decades (PTA0.5-4kHz 33.4 dB at 50-59yr vs. 28.2 dB at 30-39yr, ≤0.001), and an increased loss of word recognition scores (total average 84.7% vs. 80.0%, ≤0.001). Significant noise-associated growth in the prevalence of tinnitus also was shown, including more than a triple prevalence for constant tinnitus (28.10% vs. 8.85%, ≤0.001) and near to a double prevalence for intermittent tinnitus (19.10% vs. 11.10%, ≤0.001). Noise also resulted in the elevation of the static compliance of the tympanic membrane throughout age (total average 0.61 vs. 0.85 mmho, ≤0.001). CONCLUSIONS: Our findings emphasize the significant contribution of CNE in auditory aging and the precipitation of both ARHL and tinnitus.


Subject(s)
Tinnitus , Adult , Aged , Aged, 80 and over , Audiometry, Pure-Tone , Auditory Threshold , Canada , Humans , Middle Aged , Noise/adverse effects , Retrospective Studies , Tinnitus/epidemiology , Tinnitus/etiology
16.
Prog Neurobiol ; 194: 101878, 2020 11.
Article in English | MEDLINE | ID: mdl-32615147

ABSTRACT

Aging is associated with alterations in cognitive processing and brain neurophysiology. Whereas the primary symptom of amnestic mild cognitive impairment (aMCI) is memory problems greater than normal for age and education, patients with Alzheimer's disease (AD) show impairments in other cognitive domains in addition to memory dysfunction. Resting-state electroencephalography (rsEEG) studies in physiological aging indicate a global increase in low-frequency oscillations' power and the reduction and slowing of alpha activity. The enhancement of slow and the reduction of fast oscillations, and the disruption of brain functional connectivity, however, are characterized as major rsEEG changes in AD. Recent rodent studies also support human evidence of age- and AD-related changes in resting-state brain oscillations, and the neuroprotective effect of brain stimulation techniques through gamma-band stimulations. Cumulatively, current evidence moves toward optimizing rsEEG features as reliable predictors of people with aMCI at risk for conversion to AD and mapping neural alterations subsequent to brain stimulation therapies. The present paper reviews the latest evidence of changes in rsEEG oscillations in physiological aging, aMCI, and AD, as well as findings of various brain stimulation therapies from both human and non-human studies.


Subject(s)
Aging/physiology , Alzheimer Disease/physiopathology , Alzheimer Disease/therapy , Brain Waves/physiology , Cognitive Dysfunction/physiopathology , Connectome , Deep Brain Stimulation , Music Therapy , Transcranial Magnetic Stimulation , Alzheimer Disease/diagnosis , Animals , Cognitive Dysfunction/diagnosis , Humans
17.
Ageing Res Rev ; 59: 101028, 2020 05.
Article in English | MEDLINE | ID: mdl-32092463

ABSTRACT

Inhibition plays a crucial role in many functional domains, such as cognition, emotion, and actions. Studies on cognitive aging demonstrate changes in inhibitory mechanisms are age- and pathology-related. Prepulse inhibition (PPI) is the suppression of an acoustic startle reflex (ASR) to an intense stimulus when a weak prepulse stimulus precedes the startle stimulus. A reduction of PPI is thought to reflect dysfunction of sensorimotor gating which normally suppresses excessive behavioral responses to disruptive stimuli. Both human and rodent studies show age-dependent alterations of PPI of the ASR that are further compromised in Alzheimer's disease (AD). The auditory P50 gating, an index of repetition suppression, also is characterized as a putative electrophysiological biomarker of prodromal AD. This review provides the latest evidence of age- and AD-associated impairment of sensorimotor gating based upon both human and rodent studies, as well as the AD-related disruption of P50 gating in humans. It begins with a concise review of neural networks underlying PPI regulation. Then, evidence of age- and AD-related dysfunction of both PPI and P50 gating is discussed. The attentional/ emotional aspects of sensorimotor gating and the neurotransmitter mechanisms underpinning PPI and P50 gating are also reviewed. The review ends with conclusions and research directions.


Subject(s)
Aging , Alzheimer Disease/physiopathology , Neural Inhibition/physiology , Reflex, Startle/physiology , Acoustic Stimulation/methods , Humans , Prepulse Inhibition , Reaction Time/physiology
18.
Mov Disord ; 35(4): 537-550, 2020 04.
Article in English | MEDLINE | ID: mdl-32052894

ABSTRACT

PD is a progressive and complex neurological disorder with heterogeneous symptomatology. PD is characterized by classical motor features of parkinsonism and nonmotor symptoms and involves extensive regions of the nervous system, various neurotransmitters, and protein aggregates. Extensive evidence supports auditory dysfunction as an additional nonmotor feature of PD. Studies indicate a broad range of auditory impairments in PD, from the peripheral hearing system to the auditory brainstem and cortical areas. For instance, research demonstrates a higher occurrence of hearing loss in early-onset PD and evidence of abnormal auditory evoked potentials, event-related potentials, and habituation to novel stimuli. Electrophysiological data, such as auditory P3a, also is suggested as a sensitive measure of illness duration and severity. Improvement in auditory responses following dopaminergic therapies also indicates the presence of similar neurotransmitters (i.e., glutamate and dopamine) in the auditory system and basal ganglia. Nonetheless, hearing impairments in PD have received little attention in clinical practice so far. This review summarizes evidence of peripheral and central auditory impairments in PD and provides conclusions and directions for future empirical and clinical research. © 2020 International Parkinson and Movement Disorder Society.


Subject(s)
Parkinson Disease , Dopamine , Evoked Potentials , Humans , Neurotransmitter Agents , Parkinson Disease/complications
19.
Cereb Cortex ; 30(1): 311-325, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31070710

ABSTRACT

The prepulse inhibition (PPI) of the acoustic startle reflex (ASR), as an index of sensorimotor gating, is one of the most extensively used paradigms in the field of neuropsychiatric disorders. Few studies have examined how prenatal stress (PS) regulates the sensorimotor gating during the lifespan and how PS modifies the development of amyloid-beta (Aß) pathology in brain areas underlying the PPI formation. We followed alternations in corticosterone levels, learning and memory, and the PPI of the ASR measures in APPNL-G-F/NL-G-F offspring of dams exposed to gestational noise stress. In-depth quantifications of the Aß plaque accumulation were also performed at 6 months. The results indicated an age-dependent deterioration of sensorimotor gating, long-lasting PS-induced abnormalities in PPI magnitudes, as well as deficits in spatial memory. The PS also resulted in a higher Aß aggregation predominantly in brain areas associated with the PPI modulation network. The findings suggest the contribution of a PS-induced hypothalamic-pituitary-adrenal (HPA) axis hyperactivity in regulating the PPI modulation substrates leading to the abnormal development of the neural protection system in response to disruptive stimuli. The long-lasting HPA axis dysregulation appears to be the major underlying mechanism in precipitating the Aß deposition, especially in brain areas contributed to the PPI modulation network.


Subject(s)
Alzheimer Disease/physiopathology , Brain/physiopathology , Prenatal Exposure Delayed Effects/physiopathology , Prepulse Inhibition/physiology , Reflex, Startle/physiology , Acoustic Stimulation , Alzheimer Disease/pathology , Animals , Brain/pathology , Disease Models, Animal , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/pathology , Neural Pathways/physiopathology , Plaque, Amyloid/pathology , Pregnancy , Prenatal Exposure Delayed Effects/pathology
20.
Neurosci Biobehav Rev ; 117: 110-128, 2020 10.
Article in English | MEDLINE | ID: mdl-30978359

ABSTRACT

This review examines the adverse impacts of different noise exposure paradigms on the neuroendocrine system, hippocampal and neocortical structures, cognitive performances, and the development of Alzheimer's disease (AD)-like neuropathological changes in the brain of laboratory animals. Studies were reviewed in three periods during the lifespan including: adult animals exposed to noise, female rodents exposed to noise during gestation, and offspring exposed to noise during the prenatal period. Findings imply that chronic noise exposure dysregulates the neuroendocrine system leading to hyperactivation of the sympathetic divisions of the autonomic nervous system (i.e., the hypothalamic-pituitary-adrenal (HPA)-axis), and increases stress hormones that affect brain and behaviour. Enduring dysregulation of the HPA-axis was the most discussed mechanism for the harmful effect of noise during the lifespan. Studies also suggest a causative association of noise with diverse indicators of the AD-like neuropathology in rodents and a hypersusceptibility in females. The results indicate the importance of future neuroimaging studies to quantify the potential contribution of noise in predisposing cognitive decline and preclinical signs of dementia in humans.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Prenatal Exposure Delayed Effects , Alzheimer Disease/etiology , Animals , Animals, Laboratory , Cognitive Dysfunction/etiology , Female , Hypothalamo-Hypophyseal System , Pituitary-Adrenal System , Pregnancy , Stress, Psychological
SELECTION OF CITATIONS
SEARCH DETAIL
...