1
|
Sato R, Adachi R, Yokoi N, Tsujimura K, Egawa R, Hara Y, Fukata Y, Fukata M, Ogi T, Sone M, Kuba H. Loss of neuronal activity facilitates surface accumulation of p75NTR and cell death in avian cochlear nucleus. Neurosci Res 2025; 213:23-34. [PMID: 39848466 DOI: 10.1016/j.neures.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 01/25/2025]
Abstract
Sensorineural hearing loss causes cell death in central auditory neurons, but molecular mechanisms of triggering this process are not fully understood. We report here that loss of afferent activity promotes cell death by facilitating proBDNF-p75NTR signals in cochlear nucleus of chicks around hatch. RNA-seq analyses revealed up-regulation of genes related to proBDNF-p75NTR-JNK signals as well as apoptosis at the nucleus within 24 h after unilateral cochlea deprivation. Western blotting confirmed a high level of proBDNF protein at the nucleus. Moreover, FLAG-tagged p75NTR accumulated at the plasma membrane of the neurons within 6 h after the deprivation, well before the upregulation of apoptotic genes. Cell viability assay using propidium iodide in organ culture showed that proBDNF increased the fraction of dying neurons in a dose-dependent manner. In addition, pharmacological blockades of synaptic and spike activities in the culture reproduced the surface accumulation of p75NTR in vivo and increased the fraction of dying neurons, while genetic inhibition of p75NTR signals occluded the cell death during the activity blockades. These results indicate that afferent activity is crucial for suppressing surface accumulation of p75NTR and hence proBDNF-p75NTR signals and that the loss of this suppression would contribute to triggering cell death after deafferentation in the developing brainstem auditory circuit.
Collapse
Affiliation(s)
- Ryosuke Sato
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan; Department of Otorhinolaryngology Head and Neck Surgery, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Ryota Adachi
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Norihiko Yokoi
- Division of Neuropharmacology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Keita Tsujimura
- Group of Brain Function and Development, Nagoya University Neuroscience Institute of the Graduate School of Science, Nagoya 464-8602, Japan; Research Unit for Developmental Disorders, Institute for Advanced Research, Nagoya University, Nagoya 464-8602, Japan
| | - Ryo Egawa
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Yuichiro Hara
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan; Department of Human Genetics and Molecular Biology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Yuko Fukata
- Division of Molecular and Cellular Pharmacology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Masaki Fukata
- Division of Neuropharmacology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Tomoo Ogi
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan; Department of Human Genetics and Molecular Biology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Michihiko Sone
- Department of Otorhinolaryngology Head and Neck Surgery, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Hiroshi Kuba
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan.
| |
Collapse
|
2
|
Huang L, Hardyman F, Edwards M, Galliano E. Deprivation-Induced Plasticity in the Early Central Circuits of the Rodent Visual, Auditory, and Olfactory Systems. eNeuro 2024; 11:ENEURO.0435-23.2023. [PMID: 38195533 PMCID: PMC11059429 DOI: 10.1523/eneuro.0435-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 01/11/2024] Open
Abstract
Activity-dependent neuronal plasticity is crucial for animals to adapt to dynamic sensory environments. Traditionally, it has been investigated using deprivation approaches in animal models primarily in sensory cortices. Nevertheless, emerging evidence emphasizes its significance in sensory organs and in subcortical regions where cranial nerves relay information to the brain. Additionally, critical questions started to arise. Do different sensory modalities share common cellular mechanisms for deprivation-induced plasticity at these central entry points? Does the deprivation duration correlate with specific plasticity mechanisms? This study systematically reviews and meta-analyzes research papers that investigated visual, auditory, or olfactory deprivation in rodents of both sexes. It examines the consequences of sensory deprivation in homologous regions at the first central synapse following cranial nerve transmission (vision - lateral geniculate nucleus and superior colliculus; audition - ventral and dorsal cochlear nucleus; olfaction - olfactory bulb). The systematic search yielded 91 papers (39 vision, 22 audition, 30 olfaction), revealing substantial heterogeneity in publication trends, experimental methods, measures of plasticity, and reporting across the sensory modalities. Despite these differences, commonalities emerged when correlating plasticity mechanisms with the duration of sensory deprivation. Short-term deprivation (up to 1 d) reduced activity and increased disinhibition, medium-term deprivation (1 d to a week) involved glial changes and synaptic remodeling, and long-term deprivation (over a week) primarily led to structural alterations. These findings underscore the importance of standardizing methodologies and reporting practices. Additionally, they highlight the value of cross-modal synthesis for understanding how the nervous system, including peripheral, precortical, and cortical areas, respond to and compensate for sensory inputs loss.
Collapse
Affiliation(s)
- Li Huang
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB23EL Cambridge, United Kingdom
| | - Francesca Hardyman
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB23EL Cambridge, United Kingdom
| | - Megan Edwards
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB23EL Cambridge, United Kingdom
| | - Elisa Galliano
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB23EL Cambridge, United Kingdom
| |
Collapse
|
3
|
Singh S, Maheshwari A, Boppana S. CMV-induced Hearing Loss. NEWBORN (CLARKSVILLE, MD.) 2023; 2:249-262. [PMID: 38348106 PMCID: PMC10860330 DOI: 10.5005/jp-journals-11002-0081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
Congenital cytomegalovirus (cCMV) infection is the most common fetal viral infection and contributes to about 25% of childhood hearing loss by the age of 4 years. It is the leading nongenetic cause of sensorineural hearing loss (SNHL). Infants born to seroimmune mothers are not completely protected from SNHL, although the severity of their hearing loss may be milder than that seen in those whose mothers had a primary infection. Both direct cytopathic effects and localized inflammatory responses contribute to the pathogenesis of cytomegalovirus (CMV)-induced hearing loss. Hearing loss may be delayed onset, progressive or fluctuating in nature, and therefore, a significant proportion will be missed by universal newborn hearing screening (NHS) and warrants close monitoring of hearing function at least until 5-6 years of age. A multidisciplinary approach is required for the management of hearing loss. These children may need assistive hearing devices or cochlear implantation depending on the severity of their hearing loss. In addition, early intervention services such as speech or occupational therapy could help better communication, language, and social skill outcomes. Preventive measures to decrease intrauterine CMV transmission that have been evaluated include personal protective measures, passive immunoprophylaxis and valacyclovir treatment during pregnancy in mothers with primary CMV infection. Several vaccine candidates are currently in testing and one candidate vaccine in phase 3 trials. Until a CMV vaccine becomes available, behavioral and educational interventions may be the most effective strategy to prevent maternal CMV infection.
Collapse
Affiliation(s)
- Srijan Singh
- Department of Neonatology, Kailash Hospital, Noida, Uttar Pradesh, India
- Global Newborn Society (https://www.globalnewbornsociety.org/), Clarksville, Maryland, United States of America
| | - Akhil Maheshwari
- Global Newborn Society (https://www.globalnewbornsociety.org/), Clarksville, Maryland, United States of America
- Department of Pediatrics, Louisiana State University, Shreveport, Louisiana, United States of America
| | - Suresh Boppana
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
4
|
Yu X, Wang Y. Peripheral Fragile X messenger ribonucleoprotein is required for the timely closure of a critical period for neuronal susceptibility in the ventral cochlear nucleus. Front Cell Neurosci 2023; 17:1186630. [PMID: 37305436 PMCID: PMC10248243 DOI: 10.3389/fncel.2023.1186630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/08/2023] [Indexed: 06/13/2023] Open
Abstract
Alterations in neuronal plasticity and critical periods are common across neurodevelopmental diseases, including Fragile X syndrome (FXS), the leading single-gene cause of autism. Characterized with sensory dysfunction, FXS is the result of gene silencing of Fragile X messenger ribonucleoprotein 1 (FMR1) and loss of its product, Fragile X messenger ribonucleoprotein (FMRP). The mechanisms underlying altered critical period and sensory dysfunction in FXS are obscure. Here, we performed genetic and surgical deprivation of peripheral auditory inputs in wildtype and Fmr1 knockout (KO) mice across ages and investigated the effects of global FMRP loss on deafferentation-induced neuronal changes in the ventral cochlear nucleus (VCN) and auditory brainstem responses. The degree of neuronal cell loss during the critical period was unchanged in Fmr1 KO mice. However, the closure of the critical period was delayed. Importantly, this delay was temporally coincidental with reduced hearing sensitivity, implying an association with sensory inputs. Functional analyses further identified early-onset and long-lasting alterations in signal transmission from the spiral ganglion to the VCN, suggesting a peripheral site of FMRP action. Finally, we generated conditional Fmr1 KO (cKO) mice with selective deletion of FMRP in spiral ganglion but not VCN neurons. cKO mice recapitulated the delay in the VCN critical period closure in Fmr1 KO mice, confirming an involvement of cochlear FMRP in shaping the temporal features of neuronal critical periods in the brain. Together, these results identify a novel peripheral mechanism of neurodevelopmental pathogenesis.
Collapse
Affiliation(s)
| | - Yuan Wang
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| |
Collapse
|
5
|
Ebbers L, Runge K, Nothwang HG. Differential patterns of histone methylase EHMT2 and its catalyzed histone modifications H3K9me1 and H3K9me2 during maturation of central auditory system. Cell Tissue Res 2016; 365:247-64. [PMID: 27083448 DOI: 10.1007/s00441-016-2401-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 03/24/2016] [Indexed: 12/31/2022]
Abstract
Histone methylation is an important epigenetic mark leading to changes in DNA accessibility and transcription. Here, we investigate immunoreactivity against the euchromatic histone-lysine N-methyltransferase EHMT2 and its catalyzed mono- and dimethylation marks at histone 3 lysine 9 (H3K9me1 and H3K9me2) during postnatal differentiation of the mouse central auditory system. In the brainstem, expression of EHMT2 was high in the first postnatal week and down-regulated thereafter. In contrast, immunoreactivity in the auditory cortex (AC) remained high during the first year of life. This difference might be related to distinct demands for adult plasticity. Analyses of two deaf mouse models, namely Cldn14 (-/-) and Cacna1d (-/-), demonstrated that sound-driven or spontaneous activity had no influence on EHMT2 immunoreactivity. The methylation marks H3K9me1 and H3K9me2 were high throughout the auditory system up to 1 year. Young auditory neurons showed immunoreactivity against both methylations at similar intensities, whereas many mature neurons showed stronger labeling for either H3K9me1 or H3K9me2. These differences were only poorly correlated with cell types. To identify methyltransferases contributing to the persistent H3K9me1 and H3K9me2 marks in the adult brainstem, EHMT1 and the retinoblastoma-interacting zinc-finger protein RIZ1 were analyzed. Both were down-regulated during brainstem development, similar to EHMT2. Contrary to EHMT2, EHMT1 was also down-regulated in adult cortical areas. Together, our data reveal a marked difference in EHMT2 levels between mature brainstem and cortical areas and a decoupling between EHMT2 abundance and histone 3 lysine 9 methylations during brainstem differentiation. Furthermore, EHMT1 and EHMT2 are differentially expressed in cortical areas.
Collapse
Affiliation(s)
- Lena Ebbers
- Neurogenetics Group, Center of Excellence Hearing4All, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26111, Oldenburg, Germany
| | - Karen Runge
- Neurogenetics Group, Center of Excellence Hearing4All, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26111, Oldenburg, Germany
| | - Hans Gerd Nothwang
- Neurogenetics Group, Center of Excellence Hearing4All, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26111, Oldenburg, Germany. .,Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, 26111, Oldenburg, Germany.
| |
Collapse
|
6
|
Bradford RD, Yoo YG, Golemac M, Pugel EP, Jonjic S, Britt WJ. Murine CMV-induced hearing loss is associated with inner ear inflammation and loss of spiral ganglia neurons. PLoS Pathog 2015; 11:e1004774. [PMID: 25875183 PMCID: PMC4395355 DOI: 10.1371/journal.ppat.1004774] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 03/02/2015] [Indexed: 01/22/2023] Open
Abstract
Congenital human cytomegalovirus (HCMV) occurs in 0.5–1% of live births and approximately 10% of infected infants develop hearing loss. The mechanism(s) of hearing loss remain unknown. We developed a murine model of CMV induced hearing loss in which murine cytomegalovirus (MCMV) infection of newborn mice leads to hematogenous spread of virus to the inner ear, induction of inflammatory responses, and hearing loss. Characteristics of the hearing loss described in infants with congenital HCMV infection were observed including, delayed onset, progressive hearing loss, and unilateral hearing loss in this model and, these characteristics were viral inoculum dependent. Viral antigens were present in the inner ear as were CD3+ mononuclear cells in the spiral ganglion and stria vascularis. Spiral ganglion neuron density was decreased after infection, thus providing a mechanism for hearing loss. The lack of significant inner ear histopathology and persistence of inflammation in cochlea of mice with hearing loss raised the possibility that inflammation was a major component of the mechanism(s) of hearing loss in MCMV infected mice. Congenital infection with human cytomegalovirus (HCMV) is the most common viral infection of the fetus and occurs in 0.5–2.0% of all live births in most regions in the world. Infection of the fetus can result in a spectrum of end-organ disease, including long term damage to the central nervous system (CNS). Although less than 10% of infected infants exhibit clinical evidence of end-organ disease, up to 10% of the total number of infected infants develop hearing loss. Mechanisms of disease leading to hearing loss are poorly understood because of the limited availability of pathological specimens and accessibility of the inner ear. Existing small animal models fail to recapitulate many features of this infection of the inner ear. In this report we describe a mouse model in which newborn animals infected peripherally with murine CMV develop hearing loss following hematogenous spread of virus to the inner ear. Hearing loss occurs in 30–50% of animals and characteristics of hearing loss in infants with congenital HCMV infection, including delayed onset of hearing loss, progressive hearing loss, and unilateral hearing loss were present in infected mice. Our findings suggest that host derived inflammatory responses and not direct virus-mediated cytopathology are responsible for hearing loss. Findings from this study provide insight into potential mechanisms of hearing loss in infants with congenital HCMV infection.
Collapse
Affiliation(s)
- Russell D. Bradford
- Department of Pediatrics, University of Alabama School of Medicine, Birmingham, Alabama, United States of America
| | - Young-Gun Yoo
- Department of Pediatrics, University of Alabama School of Medicine, Birmingham, Alabama, United States of America
| | - Mijo Golemac
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Ester Pernjak Pugel
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Stipan Jonjic
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - William J. Britt
- Department of Pediatrics, University of Alabama School of Medicine, Birmingham, Alabama, United States of America
- Department of Microbiology, University of Alabama School of Medicine, Birmingham, Alabama, United States of America
- Department of Neurobiology, University of Alabama School of Medicine, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
7
|
Caminos E, Garcia-Pino E, Juiz JM. Loss of auditory activity modifies the location of potassium channel KCNQ5 in auditory brainstem neurons. J Neurosci Res 2014; 93:604-14. [PMID: 25421809 PMCID: PMC4359677 DOI: 10.1002/jnr.23516] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 09/19/2014] [Accepted: 10/17/2014] [Indexed: 12/19/2022]
Abstract
KCNQ5/Kv7.5, a low-threshold noninactivating voltage-gated potassium channel, is preferentially targeted to excitatory endings of auditory neurons in the adult rat brainstem. Endbulds of Held from auditory nerve axons on the bushy cells of the ventral cochlear nucleus (VCN) and calyces of Held around the principal neurons in the medial nucleus of the trapezoid body (MNTB) are rich in KCNQ5 immunoreactivity. We have previously shown that this synaptic distribution occurs at about the time of hearing onset. The current study tests whether this localization in excitatory endings depends on the peripheral activity carried by the auditory nerve. Auditory nerve activity was abolished by cochlear removal or intracochlear injection of tetrodotoxin (TTX). Presence of KCNQ5 was analyzed by immunocytochemistry, Western blotting, and quantitative reverse transcription polymerase chain reaction. After cochlear removal, KCNQ5 immunoreactivity was virtually undetectable at its usual location in endbulbs and calyces of Held in the anteroventral CN and in the MNTB, respectively, although it was found in cell bodies in the VCN. The results were comparable after intracochlear TTX injection, which drastically reduced KCNQ5 immunostaining in MNTB calyces and increased immunolabeling in VCN cell bodies. Endbulbs of Held in the VCN also showed diminished KCNQ5 labeling after intracochlear TTX injection. These results show that peripheral activity from auditory nerve afferents is necessary to maintain the subcellular distribution of KCNQ5 in synaptic endings of the auditory brainstem. This may contribute to adaptations in the excitability and neurotransmitter release properties of these presynaptic endings under altered input conditions. © 2014 The Authors. Journal of Neuroscience Research Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Elena Caminos
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | | | | |
Collapse
|
8
|
Ooi Y, Inui-Yamamoto C, Suzuki T, Nakadate H, Nagase Y, Seiyama A, Yoshioka Y, Seki J. In vivo magnetic resonance imaging at 11.7 Tesla visualized the effects of neonatal transection of infraorbital nerve upon primary and secondary trigeminal pathways in rats. Brain Res 2014; 1579:84-92. [PMID: 25038563 DOI: 10.1016/j.brainres.2014.07.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 06/20/2014] [Accepted: 07/08/2014] [Indexed: 01/03/2023]
Abstract
Using 11.7T ultra high-field T2-weighted MRI, the present study aimed to investigate pathological changes of primary and secondary trigeminal pathways following neonatal transection of infraorbital nerve in rats. The trigeminal pathways consist of spinal trigeminal tract, trigeminal sensory nuclear complex, medial lemniscus, ventromedial portion of external medullary lamina and ventral posterior nucleus of thalamus. By selecting optimum parameters of MRI such as repetition time, echo time, and slice orientation, this study visualized the trigeminal pathways in rats without any contrast agents. Pathological changes due to the nerve transection were found at 8 weeks of age as a marked reduction of the areas of the trigeminal pathways connecting from the injured nerve. In addition, T2-weighted MR images of the trigeminal nerve trunk and the spinal trigeminal tract suggest a communication of CSF through the trigeminal nerve between the inside and outside of the brain stem. These results support the utility of ultra high-field MRI system for noninvasive assessment of effects of trigeminal nerve injury upon the trigeminal pathways.
Collapse
Affiliation(s)
- Yasuhiro Ooi
- Division of Pathogenesis and Control of Oral Disease, Graduate School of Dentistry, Osaka University, Suita, Osaka 565-0871, Japan; CREST, Japan Science and Technology Agency (JST), Saitama 332-0012, Japan.
| | - Chizuko Inui-Yamamoto
- CREST, Japan Science and Technology Agency (JST), Saitama 332-0012, Japan; High Performance Bioimaging Research Facility, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Takashi Suzuki
- CREST, Japan Science and Technology Agency (JST), Saitama 332-0012, Japan; Department of Biomedical Engineering, National Cerebral & Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | - Hiromichi Nakadate
- CREST, Japan Science and Technology Agency (JST), Saitama 332-0012, Japan; Department of Biomedical Engineering, National Cerebral & Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | - Yoshitaka Nagase
- Department of Oral Anatomy and Neurobiology, Graduate School of Dentistry, Osaka University, Suita, Osaka 565-0871, Japan
| | - Akitoshi Seiyama
- CREST, Japan Science and Technology Agency (JST), Saitama 332-0012, Japan; Division of Medical Devices for Diagnoses, Faculty of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Yoshichika Yoshioka
- CREST, Japan Science and Technology Agency (JST), Saitama 332-0012, Japan; Biofunctional Imaging Laboratory, Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Junji Seki
- CREST, Japan Science and Technology Agency (JST), Saitama 332-0012, Japan; Department of Biomedical Engineering, National Cerebral & Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| |
Collapse
|
9
|
McBride EG, Rubel EW, Wang Y. Afferent regulation of chicken auditory brainstem neurons: rapid changes in phosphorylation of elongation factor 2. J Comp Neurol 2013; 521:1165-83. [PMID: 22987813 DOI: 10.1002/cne.23227] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 08/27/2012] [Accepted: 09/07/2012] [Indexed: 12/21/2022]
Abstract
The relationships between protein synthesis and neuronal survival are poorly understood. In chicken nucleus magnocellularis (NM), significant alterations in overall protein synthesis precede neuronal death induced by deprivation of excitatory afferent activity. Previously we demonstrated an initial reduction in the overall rate of protein synthesis in all deprived NM neurons, followed by quick recovery (starting at 6 hours) in some, but not all, neurons. Neurons with recovered protein synthesis ultimately survive, whereas others become "ghost" cells (no detectable Nissl substance) at 12-24 hours and die within 48 hours. To explore the mechanisms underlying this differential influence of afferent input on protein synthesis and cell survival, the current study investigates the involvement of eukaryotic translation elongation factor 2 (eEF2), the phosphorylation of which reduces overall protein synthesis. Using immunocytochemistry for either total or phosphorylated eEF2 (p-eEF2), we found significant reductions in the level of phosphorylated, but not total, eEF2 in NM neurons as early as 0.5-1 hour following cochlea removal. Unexpectedly, neurons with low levels of p-eEF2 show reduced protein synthesis at 6 hours, indicated by a marker for active ribosomes. At 12 hours, all "ghost" cells exhibited little or no p-eEF2 staining, although not every neuron with a comparable low level of p-eEF2 was a "ghost" cell. These observations demonstrate that a reduced level of p-eEF2 is not responsible for immediate responses (including reduced overall protein synthesis) of a neuron to compromised afferent input but may impair the neuron's ability to initiate recovery signaling for survival and make the neuron more vulnerable to death.
Collapse
Affiliation(s)
- Ethan G McBride
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, Washington 98195, USA
| | | | | |
Collapse
|
10
|
Transient, afferent input-dependent, postnatal niche for neural progenitor cells in the cochlear nucleus. Proc Natl Acad Sci U S A 2013; 110:14456-61. [PMID: 23940359 DOI: 10.1073/pnas.1307376110] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In the cochlear nucleus (CN), the first central relay of the auditory pathway, the survival of neurons during the first weeks after birth depends on afferent innervation from the cochlea. Although input-dependent neuron survival has been extensively studied in the CN, neurogenesis has not been evaluated as a possible mechanism of postnatal plasticity. Here we show that new neurons are born in the CN during the critical period of postnatal plasticity. Coincidently, we found a population of neural progenitor cells that are controlled by a complex interplay of Wnt, Notch, and TGFβ/BMP signaling, in which low levels of TGFβ/BMP signaling are permissive for progenitor proliferation that is promoted by Wnt and Notch activation. We further show that cells with activated Wnt signaling reside in the CN and that these cells have high propensity for neurosphere formation. Cochlear ablation resulted in diminishment of progenitors and Wnt/β-catenin-active cells, suggesting that the neonatal CN maintains an afferent innervation-dependent population of progenitor cells that display active canonical Wnt signaling.
Collapse
|
11
|
Nakamura PA, Cramer KS. EphB2 signaling regulates lesion-induced axon sprouting but not critical period length in the postnatal auditory brainstem. Neural Dev 2013; 8:2. [PMID: 23379484 PMCID: PMC3575227 DOI: 10.1186/1749-8104-8-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 01/14/2013] [Indexed: 11/25/2022] Open
Abstract
Background Studies of developmental plasticity may provide insight into plasticity during adulthood, when neural circuitry is less responsive to losses or changes in input. In the mammalian auditory brainstem, globular bushy cell axons of the ventral cochlear nucleus (VCN) innervate the contralateral medial nucleus of the trapezoid body (MNTB) principal neurons. VCN axonal terminations in MNTB, known as calyces of Held, are very large and specialized for high-fidelity transmission of auditory information. Following unilateral deafferentation during postnatal development, VCN axons from the intact side form connections with novel targets, including the ipsilateral MNTB. EphB signaling has been shown to play a role in this process during the first postnatal week, but mechanisms involved in this reorganization during later developmental periods remain unknown. Results We found that EphB2 signaling reduces the number of induced ipsilateral projections to the MNTB after unilateral VCN removal at postnatal day seven (P7), but not after removal of the VCN on one side at P10, after the closure of the critical period for lesion-induced innervation of the ipsilateral MNTB. Conclusions Results from this study indicate that molecular mechanisms involved in the development of circuitry may also play a part in rewiring after deafferentation during development, but do not appear to regulate the length of critical periods for plasticity.
Collapse
Affiliation(s)
- Paul A Nakamura
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA 92697, USA
| | | |
Collapse
|
12
|
Benoit ME, Hernandez MX, Dinh ML, Benavente F, Vasquez O, Tenner AJ. C1q-induced LRP1B and GPR6 proteins expressed early in Alzheimer disease mouse models, are essential for the C1q-mediated protection against amyloid-β neurotoxicity. J Biol Chem 2012; 288:654-65. [PMID: 23150673 DOI: 10.1074/jbc.m112.400168] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Complement protein C1q is induced in the brain in response to a variety of neuronal injuries, including Alzheimer disease (AD), and blocks fibrillar amyloid-β (fAβ) neurotoxicity in vitro. Here, we show that C1q protects immature and mature primary neurons against fAβ toxicity, and we report for the first time that C1q prevents toxicity induced by oligomeric forms of amyloid-β (Aβ). Gene expression analysis reveals C1q-activated phosphorylated cAMP-response element-binding protein and AP-1, two transcription factors associated with neuronal survival and neurite outgrowth, and increased LRP1B and G protein-coupled receptor 6(GPR6) expression in fAβ-injured neurons. Silencing of cAMP-response element-binding protein, LRP1B or GPR6 expression inhibited C1q-mediated neuroprotection from fAβ-induced injury. In addition, C1q altered the association of oligomeric Aβ and fAβ with neurons. In vivo, increased hippocampal expression of C1q, LRP1B, and GPR6 is observed as early as 2 months of age in the 3 × Tg mouse model of AD, whereas no such induction of LRP1B and GPR6 was seen in C1q-deficient AD mice. In contrast, expression of C1r and C1s, proteases required to activate the classical complement pathway, and C3 showed a significant age-dependent increase only after 10-13 months of age when Aβ plaques start to accumulate in this AD model. Thus, our results identify pathways by which C1q, up-regulated in vivo early in response to injury without the coordinate induction of other complement components, can induce a program of gene expression that promotes neuroprotection and thus may provide protection against Aβ in preclinical stages of AD and other neurodegenerative processes.
Collapse
Affiliation(s)
- Marie E Benoit
- Department of Molecular Biology and Biochemistry, of California at Irvine, Irvine, California 92697, USA
| | | | | | | | | | | |
Collapse
|
13
|
Egr2::cre mediated conditional ablation of dicer disrupts histogenesis of mammalian central auditory nuclei. PLoS One 2012; 7:e49503. [PMID: 23152916 PMCID: PMC3495878 DOI: 10.1371/journal.pone.0049503] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 10/09/2012] [Indexed: 11/19/2022] Open
Abstract
Histogenesis of the auditory system requires extensive molecular orchestration. Recently, Dicer1, an essential gene for generation of microRNAs, and miR-96 were shown to be important for development of the peripheral auditory system. Here, we investigated their role for the formation of the auditory brainstem. Egr2::Cre-mediated early embryonic ablation of Dicer1 caused severe disruption of auditory brainstem structures. In adult animals, the volume of the cochlear nucleus complex (CNC) was reduced by 73.5%. This decrease is in part attributed to the lack of the microneuronal shell. In contrast, fusiform cells, which similar to the granular cells of the microneural shell are derived from Egr2 positive cells, were still present. The volume reduction of the CNC was already present at birth (67.2% decrease). The superior olivary complex was also drastically affected in these mice. Nissl staining as well as Vglut1 and Calbindin 1 immunolabeling revealed that principal SOC nuclei such as the medial nucleus of the trapezoid body and the lateral superior olive were absent. Only choline acetyltransferase positive neurons of the olivocochlear bundle were observed as a densely packed cell group in the ventrolateral area of the SOC. Mid-embryonic ablation of Dicer1 in the ventral cochlear nucleus by Atoh7::Cre-mediated recombination resulted in normal formation of the cochlear nucleus complex, indicating an early embryonic requirement of Dicer1. Quantitative RT-PCR analysis of miR-96 demonstrated low expression in the embryonic brainstem and up-regulation thereafter, suggesting that other microRNAs are required for proper histogenesis of the auditory brainstem. Together our data identify a critical role of Dicer activity during embryonic development of the auditory brainstem.
Collapse
|
14
|
Seasonal changes in patterns of gene expression in avian song control brain regions. PLoS One 2012; 7:e35119. [PMID: 22529977 PMCID: PMC3329558 DOI: 10.1371/journal.pone.0035119] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Accepted: 03/13/2012] [Indexed: 01/23/2023] Open
Abstract
Photoperiod and hormonal cues drive dramatic seasonal changes in structure and function of the avian song control system. Little is known, however, about the patterns of gene expression associated with seasonal changes. Here we address this issue by altering the hormonal and photoperiodic conditions in seasonally-breeding Gambel's white-crowned sparrows and extracting RNA from the telencephalic song control nuclei HVC and RA across multiple time points that capture different stages of growth and regression. We chose HVC and RA because while both nuclei change in volume across seasons, the cellular mechanisms underlying these changes differ. We thus hypothesized that different genes would be expressed between HVC and RA. We tested this by using the extracted RNA to perform a cDNA microarray hybridization developed by the SoNG initiative. We then validated these results using qRT-PCR. We found that 363 genes varied by more than 1.5 fold (>log2 0.585) in expression in HVC and/or RA. Supporting our hypothesis, only 59 of these 363 genes were found to vary in both nuclei, while 132 gene expression changes were HVC specific and 172 were RA specific. We then assigned many of these genes to functional categories relevant to the different mechanisms underlying seasonal change in HVC and RA, including neurogenesis, apoptosis, cell growth, dendrite arborization and axonal growth, angiogenesis, endocrinology, growth factors, and electrophysiology. This revealed categorical differences in the kinds of genes regulated in HVC and RA. These results show that different molecular programs underlie seasonal changes in HVC and RA, and that gene expression is time specific across different reproductive conditions. Our results provide insights into the complex molecular pathways that underlie adult neural plasticity.
Collapse
|
15
|
Moore DR. Stroke recovery and sensory plasticity: common mechanisms? Dev Psychobiol 2012; 54:326-31. [PMID: 22415919 DOI: 10.1002/dev.20627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Presentations in this symposium are considered in the context of mechanisms of sensory plasticity, particularly in the auditory system. The auditory nervous system has two discrete end organs that are separately vulnerable to clinical and experimental injury, and brainstem processing mechanisms that are highly specialized for temporal, spectral, and spatial coding. These include giant axo-somatic synapses and neurons with spatially segregated bipolar dendrites, each innervated exclusively from one ear. This architecture allows exquisite control of afferent-target interactions, including known excitatory and inhibitory couplings, and consequently enhanced interpretation of data from other brain systems. For example, deafening can silence the auditory nerve, but has surprisingly little impact on normal brain function, as evidenced by the success of cochlear implants. The observed amplification from one brain region to another of degeneration following stroke or discrete brain lesions may thus be due to secondary rather than primary afferent consequences of the lesion.
Collapse
Affiliation(s)
- David R Moore
- MRC Institute of Hearing Research, Nottingham NG7 2RD, UK.
| |
Collapse
|
16
|
New criteria of indication and selection of patients to cochlear implant. Int J Otolaryngol 2011; 2011:573968. [PMID: 22013448 PMCID: PMC3195958 DOI: 10.1155/2011/573968] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 08/12/2011] [Indexed: 12/01/2022] Open
Abstract
Numerous changes continue to occur in cochlear implant candidacy. In general, these have been accompanied by concomitant and satisfactory changes in surgical techniques. Together, this has advanced the utility and safety of cochlear implantation. Most devices are now approved for use in patients with severe to profound unilateral hearing loss rather then the prior requirement of a bilateral profound loss. Furthermore, studies have begun utilizing short electrode arrays for shallow insertion in patients with considerable low-frequency residual hearing. This technique will allow the recipient to continue to use acoustically amplified hearing for the low frequencies simultaneously with a cochlear implant for the high frequencies. The advances in design of, and indications for, cochlear implants have been matched by improvements in surgical techniques and decrease in complications. The resulting improvements in safety and efficacy have further encouraged the use of these devices. This paper will review the new concepts in the candidacy of cochlear implant. Medline data base was used to search articles dealing with the following topics: cochlear implant in younger children, cochlear implant and hearing preservation, cochlear implant for unilateral deafness and tinnitus, genetic hearing loss and cochlear implant, bilateral cochlear implant, neuropathy and cochlear implant and neural plasticity, and the selection of patients for cochlear implant.
Collapse
|
17
|
Kazama H, Yaksi E, Wilson RI. Cell death triggers olfactory circuit plasticity via glial signaling in Drosophila. J Neurosci 2011; 31:7619-30. [PMID: 21613475 PMCID: PMC3119471 DOI: 10.1523/jneurosci.5984-10.2011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Revised: 03/29/2011] [Accepted: 04/11/2011] [Indexed: 12/13/2022] Open
Abstract
The Drosophila antennal lobe is organized into glomerular compartments, where olfactory receptor neurons synapse onto projection neurons. Projection neuron dendrites also receive input from local neurons, which interconnect glomeruli. In this study, we investigated how activity in this circuit changes over time when sensory afferents are chronically removed in vivo. In the normal circuit, excitatory connections between glomeruli are weak. However, after we chronically severed receptor neuron axons projecting to a subset of glomeruli, we found that odor-evoked lateral excitatory input to deafferented projection neurons was potentiated severalfold. This was caused, at least in part, by strengthened electrical coupling from excitatory local neurons onto projection neurons, as well as increased activity in excitatory local neurons. Merely silencing receptor neurons was not sufficient to elicit these changes, implying that severing receptor neuron axons is the relevant signal. When we expressed the neuroprotective gene Wallerian degeneration slow (Wld(S)) in receptor neurons before severing their axons, this blocked the induction of plasticity. Because expressing Wld(S) prevents severed axons from recruiting glia, this result suggests a role for glia. Consistent with this, we found that blocking endocytosis in ensheathing glia blocked the induction of plasticity. In sum, these results reveal a novel injury response whereby severed sensory axons recruit glia, which in turn signal to central neurons to upregulate their activity. By strengthening excitatory interactions between neurons in a deafferented brain region, this mechanism might help boost activity to compensate for lost sensory input.
Collapse
Affiliation(s)
- Hokto Kazama
- Department of Neurobiology, Harvard Medical School, Boston Massachusetts 02115
| | - Emre Yaksi
- Department of Neurobiology, Harvard Medical School, Boston Massachusetts 02115
| | - Rachel I. Wilson
- Department of Neurobiology, Harvard Medical School, Boston Massachusetts 02115
| |
Collapse
|
18
|
Abstract
Cochlear implants have provided hearing to more than 120,000 deaf people. Recent surgical developments include direct electrical stimulation of the brain, bilateral implants and implantation in children less than 1 year old. However, research is beginning to refocus on the role of the brain in providing benefits to implant users. The auditory system is able to use the highly impoverished input provided by implants to interpret speech, but this only works well in those who have developed language before their deafness or in those who receive their implant at a very young age. We discuss recent evidence suggesting that developing the ability of the brain to learn how to use an implant may be as important as further improvements of the implant technology.
Collapse
Affiliation(s)
- David R Moore
- Medical Research Council, Institute of Hearing Research, Nottingham, UK.
| | | |
Collapse
|