1
|
Messat Y, Martin-Fernandez M, Assou S, Chung K, Guérin F, Gergely C, Cuisinier F, Zine A. Differentiation of Spiral Ganglion Neurons from Human Dental Pulp Stem Cells: A Further Step towards Autologous Auditory Nerve Recovery. Int J Mol Sci 2024; 25:9115. [PMID: 39201803 PMCID: PMC11354632 DOI: 10.3390/ijms25169115] [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: 06/14/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 09/03/2024] Open
Abstract
The degeneration of spiral ganglion neurons (SGNs), which convey auditory signals from hair cells to the brain, can be a primary cause of sensorineural hearing loss (SNHL) or can occur secondary to hair cell loss. Emerging therapies for SNHL include the replacement of damaged SGNs using stem cell-derived otic neuronal progenitors (ONPs). However, the availability of renewable, accessible, and patient-matched sources of human stem cells is a prerequisite for successful replacement of the auditory nerve. In this study, we derived ONP and SGN-like cells by a reliable and reproducible stepwise guidance differentiation procedure of self-renewing human dental pulp stem cells (hDPSCs). This in vitro differentiation protocol relies on the modulation of BMP and TGFβ pathways using a free-floating 3D neurosphere method, followed by differentiation on a Geltrex-coated surface using two culture paradigms to modulate the major factors and pathways involved in early otic neurogenesis. Gene and protein expression analyses revealed efficient induction of a comprehensive panel of known ONP and SGN-like cell markers during the time course of hDPSCs differentiation. Atomic force microscopy revealed that hDPSC-derived SGN-like cells exhibit similar nanomechanical properties as their in vivo SGN counterparts. Furthermore, spiral ganglion neurons from newborn rats come in close contact with hDPSC-derived ONPs 5 days after co-culturing. Our data demonstrate the capability of hDPSCs to generate SGN-like neurons with specific lineage marker expression, bipolar morphology, and the nanomechanical characteristics of SGNs, suggesting that the neurons could be used for next-generation cochlear implants and/or inner ear cell-based strategies for SNHL.
Collapse
Affiliation(s)
- Yassine Messat
- LBN, Laboratory of Bioengineering and Nanoscience, University of Montpellier, 34193 Montpellier, France
| | - Marta Martin-Fernandez
- L2C, Laboratoire Charles Coulomb, University of Montpellier, CNRS, 34095 Montpellier, France
| | - Said Assou
- IRMB, Institute for Regenerative Medicine & Biotherapy, University of Montpellier, INSERM, CHU Montpellier, 34295 Montpellier, France;
| | - Keshi Chung
- LBN, Laboratory of Bioengineering and Nanoscience, University of Montpellier, 34193 Montpellier, France
| | - Frederic Guérin
- Faculté de Médecine, University of Montpellier, 34090 Montpellier, France
| | - Csilla Gergely
- L2C, Laboratoire Charles Coulomb, University of Montpellier, CNRS, 34095 Montpellier, France
| | - Frederic Cuisinier
- LBN, Laboratory of Bioengineering and Nanoscience, University of Montpellier, 34193 Montpellier, France
| | - Azel Zine
- LBN, Laboratory of Bioengineering and Nanoscience, University of Montpellier, 34193 Montpellier, France
| |
Collapse
|
2
|
Vecchi JT, Claussen AD, Hansen MR. Decreasing the physical gap in the neural-electrode interface and related concepts to improve cochlear implant performance. Front Neurosci 2024; 18:1425226. [PMID: 39114486 PMCID: PMC11303154 DOI: 10.3389/fnins.2024.1425226] [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: 04/29/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
Cochlear implants (CI) represent incredible devices that restore hearing perception for those with moderate to profound sensorineural hearing loss. However, the ability of a CI to restore complex auditory function is limited by the number of perceptually independent spectral channels provided. A major contributor to this limitation is the physical gap between the CI electrodes and the target spiral ganglion neurons (SGNs). In order for CI electrodes to stimulate SGNs more precisely, and thus better approximate natural hearing, new methodologies need to be developed to decrease this gap, (i.e., transitioning CIs from a far-field to near-field device). In this review, strategies aimed at improving the neural-electrode interface are discussed in terms of the magnitude of impact they could have and the work needed to implement them. Ongoing research suggests current clinical efforts to limit the CI-related immune response holds great potential for improving device performance. This could eradicate the dense, fibrous capsule surrounding the electrode and enhance preservation of natural cochlear architecture, including SGNs. In the long term, however, optimized future devices will likely need to induce and guide the outgrowth of the peripheral process of SGNs to be in closer proximity to the CI electrode in order to better approximate natural hearing. This research is in its infancy; it remains to be seen which strategies (surface patterning, small molecule release, hydrogel coating, etc.) will be enable this approach. Additionally, these efforts aimed at optimizing CI function will likely translate to other neural prostheses, which face similar issues.
Collapse
Affiliation(s)
- Joseph T. Vecchi
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, Iowa City, IA, United States
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, Iowa City, IA, United States
| | - Alexander D. Claussen
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, Iowa City, IA, United States
| | - Marlan R. Hansen
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, Iowa City, IA, United States
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, Iowa City, IA, United States
| |
Collapse
|
3
|
Wong EC, Lopez IA, Ishiyama A, Ishiyama G. Expression of Brain-Derived Neurotrophic Factor in Human Spiral Ganglia Neurons after Cochlear Implantation. Otol Neurotol 2024; 45:326-333. [PMID: 38238917 PMCID: PMC10922350 DOI: 10.1097/mao.0000000000004104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) is an important factor in the development and neuroprotection of afferent auditory pathways. In this study, we investigated the expression of BDNF in the afferent auditory pathway after cochlear implantation (CI), hypothesizing that electrical stimulation after CI stimulates BDNF expression in the afferent auditory pathway. METHODS Archival human temporal bones from eight patients with a history of CI and five patients with normal hearing (ages 65-93 years old) were studied. Temporal bone specimens were immunoreacted with rabbit polyclonal antibodies against BDNF and mouse monoclonal antibodies against pan-neurofilaments. In cases of unilateral CI, the BDNF expression was compared with the contralateral unimplanted ear and normal temporal bones without hearing loss. RESULTS BDNF immunoreactivity (IR) localized to the spiral ganglion neurons (SGNs) somata and the surrounding satellite cells. BDNF-IR in the spiral ganglia was similar in the apical, middle, and basal hook regions. Neurofilament IR localized to SGN nerve fibers in both implanted and unimplanted cochleae. BDNF-IR in the SGN and satellite cells was significantly increased in the implanted specimens compared with the unimplanted specimens ( p < 0.05) and the normal hearing specimens ( p < 0.05). BDNF-IR expression was similar in the unimplanted cochlea and in the normal cochlea. BDNF protein expression was increased despite complete loss of the organ of Corti hair cells and supporting cells. Even in the cases of CI with a 6-mm first-generation electrode, BDNF expression was upregulated throughout the cochlea. CONCLUSIONS BDNF expression in the SGN appears to be upregulated by the electrical stimulation from CI. This study provides evidence that the electrical stimulation from CI may stimulate the expression of BDNF, playing a neuroprotective role in the rehabilitation of hearing in the deafened ear.
Collapse
Affiliation(s)
| | | | | | - Gail Ishiyama
- UCLA Department of Head and Neck Surgery
- UCLA Department of Neurology
| |
Collapse
|
4
|
Lu J, Wang M, Wang X, Meng Y, Chen F, Zhuang J, Han Y, Wang H, Liu W. A basement membrane extract-based three-dimensional culture system promotes the neuronal differentiation of cochlear Sox10-positive glial cells in vitro. Mater Today Bio 2024; 24:100937. [PMID: 38269057 PMCID: PMC10805941 DOI: 10.1016/j.mtbio.2023.100937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/14/2023] [Accepted: 12/27/2023] [Indexed: 01/26/2024] Open
Abstract
Spiral ganglion neurons (SGNs) in the mammalian cochleae are essential for the delivery of acoustic information, and damage to SGNs can lead to permanent sensorineural hearing loss as SGNs are not capable of regeneration. Cochlear glial cells (GCs) might be a potential source for SGN regeneration, but the neuronal differentiation ability of GCs is limited and its properties are not clear yet. Here, we characterized the cochlear Sox10-positive (Sox10+) GCs as a neural progenitor population and developed a basement membrane extract-based three-dimensional (BME-3D) culture system to promote its neuronal generation capacity in vitro. Firstly, the purified Sox10+ GCs, isolated from Sox10-creER/tdTomato mice via flow cytometry, were able to form neurospheres after being cultured in the traditional suspension culture system, while significantly more neurospheres were found and the expression of stem cell-related genes was upregulated in the BME-3D culture group. Next, the BME-3D culture system promoted the neuronal differentiation ability of Sox10+ GCs, as evidenced by the increased number, neurite outgrowth, area of growth cones, and synapse density as well as the promoted excitability of newly induced neurons. Notably, the BME-3D culture system also intensified the reinnervation of newly generated neurons with HCs and protected the neurospheres and derived-neurons against cisplatin-induced damage. Finally, transcriptome sequencing analysis was performed to identify the characteristics of the differentiated neurons. These findings suggest that the BME-3D culture system considerably promotes the proliferation capacity and neuronal differentiation efficiency of Sox10+ GCs in vitro, thus providing a possible strategy for the SGN regeneration study.
Collapse
Affiliation(s)
- Junze Lu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Man Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Xue Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Yu Meng
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Fang Chen
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Jinzhu Zhuang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Yuechen Han
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Haibo Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Wenwen Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| |
Collapse
|
5
|
Mohamed T, Melfi V, Colciago A, Magnaghi V. Hearing loss and vestibular schwannoma: new insights into Schwann cells implication. Cell Death Dis 2023; 14:629. [PMID: 37741837 PMCID: PMC10517973 DOI: 10.1038/s41419-023-06141-z] [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: 04/12/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/25/2023]
Abstract
Hearing loss (HL) is the most common and heterogeneous disorder of the sensory system, with a large morbidity in the worldwide population. Among cells of the acoustic nerve (VIII cranial nerve), in the cochlea are present the hair cells, the spiral ganglion neurons, the glia-like supporting cells, and the Schwann cells (SCs), which alterations have been considered cause of HL. Notably, a benign SC-derived tumor of the acoustic nerve, named vestibular schwannoma (VS), has been indicated as cause of HL. Importantly, SCs are the main glial cells ensheathing axons and forming myelin in the peripheral nerves. Following an injury, the SCs reprogram, expressing some stemness features. Despite the mechanisms and factors controlling their biological processes (i.e., proliferation, migration, differentiation, and myelination) have been largely unveiled, their role in VS and HL was poorly investigated. In this review, we enlighten some of the mechanisms at the base of SCs transformation, VS development, and progression, likely leading to HL, and we pose great attention on the environmental factors that, in principle, could contribute to HL onset or progression. Combining the biomolecular bench-side approach to the clinical bedside practice may be helpful for the diagnosis, prediction, and therapeutic approach in otology.
Collapse
Affiliation(s)
- Tasnim Mohamed
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Valentina Melfi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133, Milan, Italy
| | - Alessandra Colciago
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Valerio Magnaghi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy.
| |
Collapse
|
6
|
Rahman MT, Bailey EM, Gansemer BM, Pieper AA, Manak JR, Green SH. Anti-inflammatory Therapy Protects Spiral Ganglion Neurons After Aminoglycoside Antibiotic-Induced Hair Cell Loss. Neurotherapeutics 2023; 20:578-601. [PMID: 36697994 PMCID: PMC10121993 DOI: 10.1007/s13311-022-01336-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2022] [Indexed: 01/26/2023] Open
Abstract
Destruction of cochlear hair cells by aminoglycoside antibiotics leads to gradual death of the spiral ganglion neurons (SGNs) that relay auditory information to the brain, potentially limiting the efficacy of cochlear implants. Because the reasons for this cochlear neurodegeneration are unknown, there are no neuroprotective strategies for patients. To investigate this problem, we assessed transcriptomic changes in the rat spiral ganglion following aminoglycoside antibiotic (kanamycin)-induced hair cell destruction. We observed selectively increased expression of immune and inflammatory response genes and increased abundance of activated macrophages in spiral ganglia by postnatal day 32 in kanamycin-deafened rats, preceding significant SGN degeneration. Treatment with the anti-inflammatory medications dexamethasone and ibuprofen diminished long-term SGN degeneration. Ibuprofen and dexamethasone also diminished macrophage activation. Efficacy of ibuprofen treatment was augmented by co-administration of the nicotinamide adenine dinucleotide-stabilizing agent P7C3-A20. Our results support a critical role of neuroinflammation in SGN degeneration after aminoglycoside antibiotic-mediated cochlear hair cell loss, as well as a neuroprotective strategy that could improve cochlear implant efficacy.
Collapse
Affiliation(s)
- Muhammad T Rahman
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA
| | - Erin M Bailey
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA
| | | | - Andrew A Pieper
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, 44106, USA
- Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, 44106, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - J Robert Manak
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA
- Department of Pediatrics, University of Iowa, Iowa City, IA, 52242, USA
| | - Steven H Green
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
7
|
Martins LC, Silva MDS, Pinheiro EF, da Penha LKRL, Passos ADCF, de Moraes SAS, Batista EDJO, Herculano AM, Oliveira KRHM. COCHLEAR GLIAL CELLS MEDIATES GLUTAMATE UPTAKE THROUGH A SODIUM-INDEPENDENT TRANSPORTER. Hear Res 2023; 432:108753. [PMID: 37054532 DOI: 10.1016/j.heares.2023.108753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023]
Abstract
Since glutamate is the primary excitatory neurotransmitter in the mammalian cochlea, the mechanisms for the removal of glutamate from the synaptic and extrasynaptic spaces are critical for maintaining normal function of this region. Glial cells of inner ear are crucial for regulation of synaptic transmission throughout since it closely interacts with neurons along the entire auditory pathway, however little is known about the activity and expression of glutamate transporters in the cochlea. In this study, using primary cochlear glial cells cultures obtained from newborn Balb/C mice, we determined the activity of a sodium-dependent and sodium-independent glutamate uptake mechanisms by means of High Performance Liquid Chromatography. The sodium-independent glutamate transport has a prominent contribution in cochlear glial cells which is similar to what has been demonstrated in other sensory organs, but it is not found in tissues less susceptible to continuous glutamate-mediated injuries. Our results showed that xCG- system is expressed in CGCs and is the main responsible for sodium-independent glutamate uptake. The identification and characterization of the xCG- transporter in the cochlea suggests a possible role of this transporter in the control of extracellular glutamate concentrations and regulation of redox state, that may aid in the preservation of auditory function.
Collapse
Affiliation(s)
- Luana Carvalho Martins
- Laboratory of Experimental Neuropharmacology, Biological Science Institute, UFPa. Belém, PA CEP:66075-110, Brazil
| | - Mateus Dos Santos Silva
- Laboratory of Experimental Neuropharmacology, Biological Science Institute, UFPa. Belém, PA CEP:66075-110, Brazil
| | - Emerson Feio Pinheiro
- Laboratory of Experimental Neuropharmacology, Biological Science Institute, UFPa. Belém, PA CEP:66075-110, Brazil
| | | | | | | | | | - Anderson Manoel Herculano
- Laboratory of Experimental Neuropharmacology, Biological Science Institute, UFPa. Belém, PA CEP:66075-110, Brazil
| | | |
Collapse
|
8
|
St. Peter M, Brough DE, Lawrence A, Nelson-Brantley J, Huang P, Harre J, Warnecke A, Staecker H. Improving Control of Gene Therapy-Based Neurotrophin Delivery for Inner Ear Applications. Front Bioeng Biotechnol 2022; 10:892969. [PMID: 35721868 PMCID: PMC9204055 DOI: 10.3389/fbioe.2022.892969] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Survival and integrity of the spiral ganglion is vital for hearing in background noise and for optimal functioning of cochlear implants. Numerous studies have demonstrated that supplementation of supraphysiologic levels of the neurotrophins BDNF and NT-3 by pumps or gene therapy strategies supports spiral ganglion survival. The endogenous physiological levels of growth factors within the inner ear, although difficult to determine, are likely extremely low within the normal inner ear. Thus, novel approaches for the long-term low-level delivery of neurotrophins may be advantageous. Objectives: This study aimed to evaluate the long-term effects of gene therapy-based low-level neurotrophin supplementation on spiral ganglion survival. Using an adenovirus serotype 28-derived adenovector delivery system, the herpes latency promoter, a weak, long expressing promoter system, has been used to deliver the BDNF or NTF3 genes to the inner ear after neomycin-induced ototoxic injury in mice. Results: Treatment of the adult mouse inner ear with neomycin resulted in acute and chronic changes in endogenous neurotrophic factor gene expression and led to a degeneration of spiral ganglion cells. Increased survival of spiral ganglion cells after adenoviral delivery of BDNF or NTF3 to the inner ear was observed. Expression of BDNF and NT-3 could be demonstrated in the damaged organ of Corti after gene delivery. Hearing loss due to overexpression of neurotrophins in the normal hearing ear was avoided when using this novel vector–promoter combination. Conclusion: Combining supporting cell-specific gene delivery via the adenovirus serotype 28 vector with a low-strength long expressing promoter potentially can provide long-term neurotrophin delivery to the damaged inner ear.
Collapse
Affiliation(s)
| | | | - Anna Lawrence
- Department of Otolaryngology, University of Kansas School of Medicine, Kansas City, KS, United States
| | | | - Peixin Huang
- Department of Otolaryngology, University of Kansas School of Medicine, Kansas City, KS, United States
| | - Jennifer Harre
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Athanasia Warnecke
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Hinrich Staecker
- Department of Otolaryngology, University of Kansas School of Medicine, Kansas City, KS, United States
- *Correspondence: Hinrich Staecker,
| |
Collapse
|
9
|
Wichova H, Shew M, Nelson-Brantley J, Warnecke A, Prentiss S, Staecker H. MicroRNA Profiling in the Perilymph of Cochlear Implant Patients: Identifying Markers that Correlate to Audiological Outcomes. J Am Acad Audiol 2022; 32:627-635. [PMID: 35609590 DOI: 10.1055/s-0041-1742234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
HYPOTHESIS MicroRNA (miRNA) expression profiles from human perilymph correlate to post cochlear implantation (CI) hearing outcomes. BACKGROUND The high inter-individual variability in speech perception among cochlear implant recipients is still poorly understood. MiRNA expression in perilymph can be used to characterize the molecular processes underlying inner ear disease and to predict performance with a cochlear implant. METHODS Perilymph collected during CI from 17 patients was analyzed using microarrays. MiRNAs were identified and multivariable analysis using consonant-nucleus-consonant testing at 6 and 18 months post implant activation was performed. Variables analyzed included age, gender, preoperative pure tone average (PTA), and preoperative speech discrimination (word recognition [WR]). Gene ontology analysis was performed to identify potential functional implications of changes in the identified miRNAs. RESULTS Distinct miRNA profiles correlated to preoperative PTA and WR. Patients classified as poor performers showed downregulation of six miRNAs that potentially regulate pathways related to neuronal function and cell survival. CONCLUSION Individual miRNA profiles can be identified in microvolumes of perilymph. Distinct non-coding RNA expression profiles correlate to preoperative hearing and postoperative cochlear implant outcomes.
Collapse
Affiliation(s)
| | - Matthew Shew
- Department of Otolaryngology Head and Neck Surgery, Washington University School of Medicine in St. Louis, Missouri
| | - Jennifer Nelson-Brantley
- Department of Anatomy and Cell Biology, School of Medicine, University of Kanas, Kansas City, Kansas
| | - Athanasia Warnecke
- Department of Otolaryngology Head and Neck Surgery, Hannover Medical School, Hannover, Germany
| | - Sandra Prentiss
- Department of Otolaryngology Head and Neck Surgery, University of Miami School of Medicine, Miami, Florida
| | - Hinrich Staecker
- Department of Otolaryngology Head and Neck Surgery, University of Kansas School of Medicine, Kansas City Kansas
| |
Collapse
|
10
|
Wang M, Xu L, Han Y, Wang X, Chen F, Lu J, Wang H, Liu W. Regulation of Spiral Ganglion Neuron Regeneration as a Therapeutic Strategy in Sensorineural Hearing Loss. Front Mol Neurosci 2022; 14:829564. [PMID: 35126054 PMCID: PMC8811300 DOI: 10.3389/fnmol.2021.829564] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/27/2021] [Indexed: 12/16/2022] Open
Abstract
In the mammalian cochlea, spiral ganglion neurons (SGNs) are the primary neurons on the auditory conduction pathway that relay sound signals from the inner ear to the brainstem. However, because the SGNs lack the regeneration ability, degeneration and loss of SGNs cause irreversible sensorineural hearing loss (SNHL). Besides, the effectiveness of cochlear implant therapy, which is the major treatment of SNHL currently, relies on healthy and adequate numbers of intact SGNs. Therefore, it is of great clinical significance to explore how to regenerate the SGNs. In recent years, a number of researches have been performed to improve the SGNs regeneration strategy, and some of them have shown promising results, including the progress of SGN regeneration from exogenous stem cells transplantation and endogenous glial cells’ reprogramming. Yet, there are challenges faced in the effectiveness of SGNs regeneration, the maturation and function of newly generated neurons as well as auditory function recovery. In this review, we describe recent advances in researches in SGNs regeneration. In the coming years, regenerating SGNs in the cochleae should become one of the leading biological strategies to recover hearing loss.
Collapse
|
11
|
Jeong M, Bojkovic K, Sagi V, Stankovic KM. Molecular and Clinical Significance of Fibroblast Growth Factor 2 in Development and Regeneration of the Auditory System. Front Mol Neurosci 2022; 14:757441. [PMID: 35002617 PMCID: PMC8733209 DOI: 10.3389/fnmol.2021.757441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/18/2021] [Indexed: 01/25/2023] Open
Abstract
The fibroblast growth factor 2 (FGF2) is a member of the FGF family which is involved in key biological processes including development, cellular proliferation, wound healing, and angiogenesis. Although the utility of the FGF family as therapeutic agents has attracted attention, and FGF2 has been studied in several clinical contexts, there remains an incomplete understanding of the molecular and clinical function of FGF2 in the auditory system. In this review, we highlight the role of FGF2 in inner ear development and hearing protection and present relevant clinical studies for tympanic membrane (TM) repair. We conclude by discussing the future implications of FGF2 as a potential therapeutic agent.
Collapse
Affiliation(s)
- Minjin Jeong
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States.,Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear and Harvard Medical School, Boston, MA, United States
| | - Katarina Bojkovic
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear and Harvard Medical School, Boston, MA, United States
| | - Varun Sagi
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States.,University of Minnesota Medical School, Minneapolis, MN, United States
| | - Konstantina M Stankovic
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States.,Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
12
|
Danielian A, Ishiyama G, Lopez IA, Ishiyama A. Predictors of Fibrotic and Bone Tissue Formation With 3-D Reconstructions of Post-implantation Human Temporal Bones. Otol Neurotol 2021; 42:e942-e948. [PMID: 33710156 PMCID: PMC8282738 DOI: 10.1097/mao.0000000000003106] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
HYPOTHESIS Years of implantation, surgical insertion approach, and electrode length will impact the volume of new tissue formation secondary to cochlear implantation. BACKGROUND New tissue formation, fibrosis, and osteoneogenesis after cochlear implantation have been implicated in increasing impedance and affecting performance of the cochlear implant. METHODS 3-D reconstructions of 15 archival human temporal bones from patients with a history of cochlear implantation (CI) were generated from H&E histopathologic slides to study factors which affect volume of tissue formation. RESULTS Years of implantation was a predictor of osteoneogenesis (r = 0.638, p-value = 0.011) and total new tissue formation (r = 0.588, p-value = 0.021), however not of fibrosis (r = 0.235, p-value = 0.399). Median total tissue formation differed between cochleostomy and round window insertions, 25.98 and 10.34%, respectively (Mann-Whitney U = 7, p = 0.018). No correlations were found between electrode length or angular insertion depth and total new tissue (p = 0.192, p = 0.35), osteoneogenesis (p = 0.193, p = 0.27), and fibrosis (p = 0.498, p = 0.83), respectively. However, the type II error for electrode length and angular insertion depth ranged from 0.73 to 0.90, largely due to small numbers of the shorter electrodes. CONCLUSIONS With numbers of cochlear implant recipients increasing worldwide, an understanding of how to minimize intracochlear changes from implantation is important. The present study demonstrates that increasing years of implantation and inserting electrodes via a cochleostomy compared with a round window approach are associated with significantly greater degree of new tissue volume formation. While previous studies have demonstrated increased intracochlear damage in the setting of translocation with longer electrodes, length, and angular insertion depth of CI electrodes were not associated with increased tissue formation.
Collapse
Affiliation(s)
- Arman Danielian
- UCLA David Geffen School of Medicine Department of Head and Neck Surgery, Los Angeles, CA, 90095, USA
| | - Gail Ishiyama
- UCLA David Geffen School of Medicine Department of Neurology, Los Angeles, CA, 90095, USA
| | - Ivan A Lopez
- UCLA David Geffen School of Medicine Department of Head and Neck Surgery, Los Angeles, CA, 90095, USA
| | - Akira Ishiyama
- UCLA David Geffen School of Medicine Department of Head and Neck Surgery, Los Angeles, CA, 90095, USA
| |
Collapse
|
13
|
Interaction of micropatterned topographical and biochemical cues to direct neurite growth from spiral ganglion neurons. Hear Res 2021; 409:108315. [PMID: 34343850 DOI: 10.1016/j.heares.2021.108315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/07/2021] [Accepted: 07/12/2021] [Indexed: 01/01/2023]
Abstract
Functional outcomes with neural prosthetic devices, such as cochlear implants, are limited in part due to physical separation between the stimulating elements and the neurons they stimulate. One strategy to close this gap aims to precisely guide neurite regeneration to position the neurites in closer proximity to electrode arrays. Here, we explore the ability of micropatterned biochemical and topographic guidance cues, singly and in combination, to direct the growth of spiral ganglion neuron (SGN) neurites, the neurons targeted by cochlear implants. Photopolymerization of methacrylate monomers was used to form unidirectional topographical features of ridges and grooves in addition to multidirectional patterns with 90o angle turns. Microcontact printing was also used to create similar uni- and multi-directional patterns of peptides on polymer surfaces. Biochemical cues included peptides that facilitate (laminin, LN) or repel (EphA4-Fc) neurite growth. On flat surfaces, SGN neurites preferentially grew on LN-coated stripes and avoided EphA4-Fc-coated stripes. LN or EphA4-Fc was selectively adsorbed onto the ridges or grooves to test the neurite response to a combination of topographical and biochemical cues. Coating the ridges with EphA4-Fc and grooves with LN lead to enhanced SGN alignment to topographical patterns. Conversely, EphA4-Fc coating on the grooves or LN coating on the ridges tended to disrupt alignment to topographical patterns. SGN neurites respond to combinations of topographical and biochemical cues and surface patterning that leverages both cues enhance guided neurite growth.
Collapse
|
14
|
Kohrman D, Borges BC, Cassinotti L, Ji L, Corfas G. Axon-glia interactions in the ascending auditory system. Dev Neurobiol 2021; 81:546-567. [PMID: 33561889 PMCID: PMC9004231 DOI: 10.1002/dneu.22813] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/25/2020] [Accepted: 02/05/2021] [Indexed: 11/09/2022]
Abstract
The auditory system detects and encodes sound information with high precision to provide a high-fidelity representation of the environment and communication. In mammals, detection occurs in the peripheral sensory organ (the cochlea) containing specialized mechanosensory cells (hair cells) that initiate the conversion of sound-generated vibrations into action potentials in the auditory nerve. Neural activity in the auditory nerve encodes information regarding the intensity and frequency of sound stimuli, which is transmitted to the auditory cortex through the ascending neural pathways. Glial cells are critical for precise control of neural conduction and synaptic transmission throughout the pathway, allowing for the precise detection of the timing, frequency, and intensity of sound signals, including the sub-millisecond temporal fidelity is necessary for tasks such as sound localization, and in humans, for processing complex sounds including speech and music. In this review, we focus on glia and glia-like cells that interact with hair cells and neurons in the ascending auditory pathway and contribute to the development, maintenance, and modulation of neural circuits and transmission in the auditory system. We also discuss the molecular mechanisms of these interactions, their impact on hearing and on auditory dysfunction associated with pathologies of each cell type.
Collapse
Affiliation(s)
- David Kohrman
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Beatriz C. Borges
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Luis Cassinotti
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Lingchao Ji
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Gabriel Corfas
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| |
Collapse
|
15
|
Dougherty MC, Shibata SB, Hansen MR. The biological underpinnings of radiation therapy for vestibular schwannomas: Review of the literature. Laryngoscope Investig Otolaryngol 2021; 6:458-468. [PMID: 34195368 PMCID: PMC8223465 DOI: 10.1002/lio2.553] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/05/2021] [Accepted: 03/12/2021] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Radiation therapy is a mainstay in the treatment of numerous neoplasms. Numerous publications have reported good clinical outcomes for primary radiation therapy for Vestibular Schwannomas (VS). However, there are relatively few pathologic specimens of VSs available to evaluate post-radiation, which has led to a relative dearth in research on the cellular mechanisms underlying the effects of radiation therapy on VSs. METHODS Here we review the latest literature on the complex biological effects of radiation therapy on these benign tumors-including resistance to oxidative stress, mechanisms of DNA damage repair, alterations in normal growth factor pathways, changes in surrounding vasculature, and alterations in immune responses following radiation. RESULTS Although VSs are highly radioresistant, radiotherapy is often successful in arresting their growth. CONCLUSION By better understanding the mechanisms underlying these effects, we could potentially harness such mechanisms in the future to potentiate the clinical effects of radiotherapy on VSs. LEVEL OF EVIDENCE N/A.
Collapse
Affiliation(s)
- Mark C. Dougherty
- Department of NeurosurgeryUniversity of Iowa Hospitals & ClinicsIowa CityIowaUSA
| | - Seiji B. Shibata
- Department of Otolaryngology, Keck School of Medicine of USCUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Marlan R. Hansen
- Department of Otolaryngology—Head & Neck SurgeryUniversity of Iowa Hospitals & ClinicsIowa CityIowaUSA
| |
Collapse
|
16
|
The Distribution and Prevalence of Macrophages in the Cochlea Following Cochlear Implantation in the Human: An Immunohistochemical Study Using Anti-Iba1 Antibody. Otol Neurotol 2021; 41:e304-e316. [PMID: 31821256 DOI: 10.1097/mao.0000000000002495] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
HYPOTHESIS Cochlear implantation may cause an increase in the number of macrophages in the human cochlea similar to previous findings in the vestibular endorgans. BACKGROUND Macrophages play a key role in both an inflammatory response and homeostatic maintenance. Recently, an increase in the prevalence of macrophages was demonstrated in the human vestibular endorgans after implantation. However, the prevalence of macrophages in the cochlea after implantation is unclear. The aim of this study was to compare the distribution and prevalence of macrophages in implanted human cochleae and the contralateral unimplanted ears. METHODS The prevalence of macrophages in the cochlea in 10 human subjects who had undergone unilateral cochlear implantation was studied by light microscopy using anti-Iba1 immunostaining. The densities of macrophages in the osseous spiral lamina (OSL) and Rosenthal's canal (RC) in implanted cochleae were compared with the contralateral unimplanted ears. The distribution of macrophage morphology (amoeboid, transitional, and ramified) was also compared. RESULTS There were activated and phagocytosing macrophages within the fibrotic sheath surrounding the electrode track and within fibrous tissue with lymphocytic infiltration in implanted ears. The densities of macrophages in OSL and RC in implanted ears were significantly greater than in unimplanted ears in some areas. There was also a difference in the prevalence of macrophage phenotype between the OSL and RC. CONCLUSION An increase in the density of macrophages in the cochlea after cochlear implantation was demonstrated. Both phagocytosis and anti-inflammatory activity of macrophages were suggested by the distribution and prevalence of macrophages in the implanted cochlea.
Collapse
|
17
|
Stojkovic M, Han D, Jeong M, Stojkovic P, Stankovic KM. Human induced pluripotent stem cells and CRISPR/Cas-mediated targeted genome editing: Platforms to tackle sensorineural hearing loss. STEM CELLS (DAYTON, OHIO) 2021; 39:673-696. [PMID: 33586253 DOI: 10.1002/stem.3353] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 12/13/2020] [Indexed: 11/09/2022]
Abstract
Hearing loss (HL) is a major global health problem of pandemic proportions. The most common type of HL is sensorineural hearing loss (SNHL) which typically occurs when cells within the inner ear are damaged. Human induced pluripotent stem cells (hiPSCs) can be generated from any individual including those who suffer from different types of HL. The development of new differentiation protocols to obtain cells of the inner ear including hair cells (HCs) and spiral ganglion neurons (SGNs) promises to expedite cell-based therapy and screening of potential pharmacologic and genetic therapies using human models. Considering age-related, acoustic, ototoxic, and genetic insults which are the most frequent causes of irreversible damage of HCs and SGNs, new methods of genome editing (GE), especially the CRISPR/Cas9 technology, could bring additional opportunities to understand the pathogenesis of human SNHL and identify novel therapies. However, important challenges associated with both hiPSCs and GE need to be overcome before scientific discoveries are correctly translated to effective and patient-safe applications. The purpose of the present review is (a) to summarize the findings from published reports utilizing hiPSCs for studies of SNHL, hence complementing recent reviews focused on animal studies, and (b) to outline promising future directions for deciphering SNHL using disruptive molecular and genomic technologies.
Collapse
Affiliation(s)
- Miodrag Stojkovic
- Eaton Peabody Laboratories, Department of Otolaryngology Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Dongjun Han
- Eaton Peabody Laboratories, Department of Otolaryngology Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Minjin Jeong
- Eaton Peabody Laboratories, Department of Otolaryngology Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Petra Stojkovic
- Eaton Peabody Laboratories, Department of Otolaryngology Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Konstantina M Stankovic
- Eaton Peabody Laboratories, Department of Otolaryngology Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA.,Program in Speech and Hearing Bioscience and Technology, Harvard University, Cambridge, Massachusetts, USA.,Harvard Program in Therapeutic Science, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
18
|
Affortit C, Casas F, Ladrech S, Ceccato JC, Bourien J, Coyat C, Puel JL, Lenoir M, Wang J. Exacerbated age-related hearing loss in mice lacking the p43 mitochondrial T3 receptor. BMC Biol 2021; 19:18. [PMID: 33526032 PMCID: PMC7852282 DOI: 10.1186/s12915-021-00953-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 01/08/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Age-related hearing loss (ARHL), also known as presbycusis, is the most common sensory impairment seen in elderly people. However, the cochlear aging process does not affect people uniformly, suggesting that both genetic and environmental (e.g., noise, ototoxic drugs) factors and their interaction may influence the onset and severity of ARHL. Considering the potential links between thyroid hormone, mitochondrial activity, and hearing, here, we probed the role of p43, a N-terminally truncated and ligand-binding form of the nuclear receptor TRα1, in hearing function and in the maintenance of hearing during aging in p43-/- mice through complementary approaches, including in vivo electrophysiological recording, ultrastructural assessments, biochemistry, and molecular biology. RESULTS We found that the p43-/- mice exhibit no obvious hearing loss in juvenile stages, but that these mice developed a premature, and more severe, ARHL resulting from the loss of cochlear sensory outer and inner hair cells and degeneration of spiral ganglion neurons. Exacerbated ARHL in p43-/- mice was associated with the early occurrence of a drastic fall of SIRT1 expression, together with an imbalance between pro-apoptotic Bax, p53 expression, and anti-apoptotic Bcl2 expression, as well as an increase in mitochondrial dysfunction, oxidative stress, and inflammatory process. Finally, p43-/- mice were also more vulnerable to noise-induced hearing loss. CONCLUSIONS These results demonstrate for the first time a requirement for p43 in the maintenance of hearing during aging and highlight the need to probe the potential link between human THRA gene polymorphisms and/or mutations and accelerated age-related deafness or some adult-onset syndromic deafness.
Collapse
Affiliation(s)
- Corentin Affortit
- INSERM - UMR 1051, Institut des Neurosciences de Montpellier, 80 rue Augustin Fliche, 34295, Montpellier, France
- Université de Montpellier, 34000, Montpellier, France
| | - François Casas
- INRA, UMR 866 Dynamique Musculaire et Métabolisme,, 34060, Montpellier, France
| | - Sabine Ladrech
- INSERM - UMR 1051, Institut des Neurosciences de Montpellier, 80 rue Augustin Fliche, 34295, Montpellier, France
- Université de Montpellier, 34000, Montpellier, France
| | - Jean-Charles Ceccato
- INSERM - UMR 1051, Institut des Neurosciences de Montpellier, 80 rue Augustin Fliche, 34295, Montpellier, France
- Université de Montpellier, 34000, Montpellier, France
| | - Jérôme Bourien
- INSERM - UMR 1051, Institut des Neurosciences de Montpellier, 80 rue Augustin Fliche, 34295, Montpellier, France
- Université de Montpellier, 34000, Montpellier, France
| | - Carolanne Coyat
- INSERM - UMR 1051, Institut des Neurosciences de Montpellier, 80 rue Augustin Fliche, 34295, Montpellier, France
- Université de Montpellier, 34000, Montpellier, France
| | - Jean-Luc Puel
- INSERM - UMR 1051, Institut des Neurosciences de Montpellier, 80 rue Augustin Fliche, 34295, Montpellier, France
- Université de Montpellier, 34000, Montpellier, France
| | - Marc Lenoir
- INSERM - UMR 1051, Institut des Neurosciences de Montpellier, 80 rue Augustin Fliche, 34295, Montpellier, France
- Université de Montpellier, 34000, Montpellier, France
| | - Jing Wang
- INSERM - UMR 1051, Institut des Neurosciences de Montpellier, 80 rue Augustin Fliche, 34295, Montpellier, France.
- Université de Montpellier, 34000, Montpellier, France.
- ENT Department, CHU Montpellier, 34295, Montpellier, France.
| |
Collapse
|
19
|
Kersigo J, Gu L, Xu L, Pan N, Vijayakuma S, Jones T, Shibata SB, Fritzsch B, Hansen MR. Effects of Neurod1 Expression on Mouse and Human Schwannoma Cells. Laryngoscope 2021; 131:E259-E270. [PMID: 32438526 PMCID: PMC7772964 DOI: 10.1002/lary.28671] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/11/2020] [Accepted: 03/18/2020] [Indexed: 12/22/2022]
Abstract
OBJECTIVES The objective was to explore the effect of the proneuronal transcription factor neurogenic differentiation 1 (Neurod1, ND1) on Schwann cells (SC) and schwannoma cell proliferation. METHODS Using a variety of transgenic mouse lines, we investigated how expression of Neurod1 effects medulloblastoma (MB) growth, schwannoma tumor progression, vestibular function, and SC cell proliferation. Primary human vestibular schwannoma (VS) cell cultures were transduced with adenoviral vectors expressing Neurod1. Cell proliferation was assessed by 5-ethynyl-2'-deoxyuridine (EdU) uptake. STUDY DESIGN Basic science investigation. RESULTS Expression of Neurod1 reduced the growth of slow-growing but not fast-growing MB models. Gene transfer of Neurod1 in human schwannoma cultures significantly reduced cell proliferation in dose-dependent way. Deletion of the neurofibromatosis type 2 (Nf2) tumor-suppressor gene via Cre expression in SCs led to increased intraganglionic SC proliferation and mildly reduced vestibular sensory-evoked potentials (VsEP) responses compared to age-matched wild-type littermates. The effect of Neurod1-induced expression on intraganglionic SC proliferation in animals lacking Nf2 was mild and highly variable. Sciatic nerve axotomy significantly increased SC proliferation in wild-type and Nf2-null animals, and expression of Neurod1 reduced the proliferative capacity of both wild-type and Nf2-null SCs following nerve injury. CONCLUSION Expression of Neurod1 reduces slow-growing MB progression and reduces human SC proliferation in primary VS cultures. In a genetic mouse model of schwannomas, we find some effects of Neurod1 expression; however, the high variability indicates that more tightly regulated Neurod1 expression levels that mimic our in vitro data are needed to fully validate Neurod1 effects on schwannoma progression. LEVEL OF EVIDENCE NA Laryngoscope, 131:E259-E270, 2021.
Collapse
Affiliation(s)
- Jennifer Kersigo
- Department of Biology, University of Lowa, Lowa City, Lowa, U.S.A
| | - Lintao Gu
- Department of Otolaryngology, University of Lowa, Lowa City, Lowa, U.S.A
- Decibel Pharmaceutical, Boston, Massachusetts, U.S.A
| | - Linjing Xu
- Department of Otolaryngology, University of Lowa, Lowa City, Lowa, U.S.A
| | - Ning Pan
- Department of Biology, University of Lowa, Lowa City, Lowa, U.S.A
- Department of Special Education & Communication Disorders, University of Nebraska, Lincoln, Nebraska, U.S.A
| | - Sarath Vijayakuma
- Department of Otolaryngology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Timothy Jones
- Department of Otolaryngology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Seiji B Shibata
- Department of Otolaryngology, University of Lowa, Lowa City, Lowa, U.S.A
| | - Bernd Fritzsch
- Department of Biology, University of Lowa, Lowa City, Lowa, U.S.A
- Department of Otolaryngology, University of Lowa, Lowa City, Lowa, U.S.A
| | - Marlan R Hansen
- Department of Otolaryngology, University of Lowa, Lowa City, Lowa, U.S.A
| |
Collapse
|
20
|
Ding D, Manohar S, Jiang H, Salvi R. Hydroxypropyl-β-cyclodextrin causes massive damage to the developing auditory and vestibular system. Hear Res 2020; 396:108073. [PMID: 32956992 DOI: 10.1016/j.heares.2020.108073] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/03/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022]
Abstract
2-hydroxypropyl-β-cyclodextrin (HPβCD), a cholesterol chelator used to treat Niemann-Pick C1 (NPC1) lysosomal storage disease, causes hearing loss in mammals by preferentially destroying outer hair cells. Because cholesterol plays an important role in early neural development, we hypothesized that HPβCD would cause more extensive damage to postnatal cochlear and vestibular structures in than adult rats. This hypothesis was tested by administering HPβCD to adult rats and postnatal day 3 (P3) cochlear and vestibular organ cultures. Adult rats treated with HPβCD developed hearing impairment and outer hair cell loss 3-day post-treatment; damage increased with dose from the high frequency base toward the low-frequency apex. The HPβCD-induced histopathologies were more severe and widespread in cochlear and vestibular cultures at P3 than in adults. HPβCD destroyed both outer and inner hair cells, auditory nerve fibers and spiral ganglion neurons as well as type I and type II vestibular hair cells and vestibular ganglion neurons. The early stage of HPβCD damage involved disruption of hair cell mechanotransduction and destruction of stereocilia. HPβCD-mediated apoptosis in P3 cultures was most-strongly initiated by activation of the extrinsic caspase-8 cell death pathway in cochlear and vestibular hair cells and neurons followed by activation of executioner caspase-3. Thus, HPβCD is toxic to all types of postnatal cochlear and vestibular hair cells and neurons in vitro whereas in vivo it only appears to destroy outer hair cells in adult cochleae. The more severe HPβCD-induced damage in postnatal cultures could be due to greater drug bioavailability in vitro and/or greater vulnerability of the developing inner ear.
Collapse
Affiliation(s)
- Dalian Ding
- Center for Hearing and Deafness, University at Buffalo, 137 Cary Hall, Buffalo, NY 14214, United States
| | - Senthilvelan Manohar
- Center for Hearing and Deafness, University at Buffalo, 137 Cary Hall, Buffalo, NY 14214, United States
| | - Haiyan Jiang
- Center for Hearing and Deafness, University at Buffalo, 137 Cary Hall, Buffalo, NY 14214, United States
| | - Richard Salvi
- Center for Hearing and Deafness, University at Buffalo, 137 Cary Hall, Buffalo, NY 14214, United States.
| |
Collapse
|
21
|
Seist R, Tong M, Landegger LD, Vasilijic S, Hyakusoku H, Katsumi S, McKenna CE, Edge ASB, Stankovic KM. Regeneration of Cochlear Synapses by Systemic Administration of a Bisphosphonate. Front Mol Neurosci 2020; 13:87. [PMID: 32765216 PMCID: PMC7381223 DOI: 10.3389/fnmol.2020.00087] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/28/2020] [Indexed: 12/12/2022] Open
Abstract
Sensorineural hearing loss (SNHL) caused by noise exposure and attendant loss of glutamatergic synapses between cochlear spiral ganglion neurons (SGNs) and hair cells is the most common sensory deficit worldwide. We show here that systemic administration of a bisphosphonate to mice 24 h after synaptopathic noise exposure regenerated synapses between inner hair cells and SGNs and restored cochlear function. We further demonstrate that this effect is mediated by inhibition of the mevalonate pathway. These results are highly significant because they suggest that bisphosphonates could reverse cochlear synaptopathy for the treatment of SNHL.
Collapse
Affiliation(s)
- Richard Seist
- Eaton-Peabody Laboratories, Department of Otolaryngology – Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
- Department of Otorhinolaryngology-Head and Neck Surgery, Paracelsus Medical University, Salzburg, Austria
| | - Mingjie Tong
- Eaton-Peabody Laboratories, Department of Otolaryngology – Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
| | - Lukas D. Landegger
- Eaton-Peabody Laboratories, Department of Otolaryngology – Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
- Department of Otorhinolaryngology-Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Sasa Vasilijic
- Eaton-Peabody Laboratories, Department of Otolaryngology – Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
| | - Hiroshi Hyakusoku
- Eaton-Peabody Laboratories, Department of Otolaryngology – Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
- Department of Otorhinolaryngology, Yokosuka Kyosai Hospital, Kanagawa, Japan
| | - Sachiyo Katsumi
- Eaton-Peabody Laboratories, Department of Otolaryngology – Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
| | - Charles E. McKenna
- Department of Chemistry, University of Southern California, Los Angeles, CA, United States
| | - Albert S. B. Edge
- Eaton-Peabody Laboratories, Department of Otolaryngology – Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
- Speech and Hearing Bioscience and Technology Program, Harvard Medical School, Boston, MA, United States
- Harvard Stem Cell Institute, Cambridge, MA, United States
| | - Konstantina M. Stankovic
- Eaton-Peabody Laboratories, Department of Otolaryngology – Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
- Speech and Hearing Bioscience and Technology Program, Harvard Medical School, Boston, MA, United States
- Harvard Stem Cell Institute, Cambridge, MA, United States
- Program in Therapeutic Science, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
22
|
β-Secretase BACE1 Is Required for Normal Cochlear Function. J Neurosci 2019; 39:9013-9027. [PMID: 31527119 DOI: 10.1523/jneurosci.0028-19.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 08/30/2019] [Accepted: 09/04/2019] [Indexed: 12/20/2022] Open
Abstract
Cleavage of amyloid precursor protein (APP) by β-secretase BACE1 initiates the production and accumulation of neurotoxic amyloid-β peptides, which is widely considered an essential pathogenic mechanism in Alzheimer's disease (AD). Here, we report that BACE1 is essential for normal auditory function. Compared with wild-type littermates, BACE1-/- mice of either sex exhibit significant hearing deficits, as indicated by increased thresholds and reduced amplitudes in auditory brainstem responses (ABRs) and decreased distortion product otoacoustic emissions (DPOAEs). Immunohistochemistry revealed aberrant synaptic organization in the cochlea and hypomyelination of auditory nerve fibers as predominant neuropathological substrates of hearing loss in BACE1-/- mice. In particular, we found that fibers of spiral ganglion neurons (SGN) close to the organ of Corti are disorganized and abnormally swollen. BACE1 deficiency also engenders organization defects in the postsynaptic compartment of SGN fibers with ectopic overexpression of PSD95 far outside the synaptic region. During postnatal development, auditory fiber myelination in BACE1-/- mice lags behind dramatically and remains incomplete into adulthood. We relate the marked hypomyelination to the impaired processing of Neuregulin-1 when BACE1 is absent. To determine whether the cochlea of adult wild-type mice is susceptible to AD treatment-like suppression of BACE1, we administered the established BACE1 inhibitor NB-360 for 6 weeks. The drug suppressed BACE1 activity in the brain, but did not impair hearing performance and, upon neuropathological examination, did not produce the characteristic cochlear abnormalities of BACE1-/- mice. Together, these data strongly suggest that the hearing loss of BACE1 knock-out mice represents a developmental phenotype.SIGNIFICANCE STATEMENT Given its crucial role in the pathogenesis of Alzheimer's disease (AD), BACE1 is a prime pharmacological target for AD prevention and therapy. However, the safe and long-term administration of BACE1-inhibitors as envisioned in AD requires a comprehensive understanding of the various physiological functions of BACE1. Here, we report that BACE1 is essential for the processing of auditory signals in the inner ear, as BACE1-deficient mice exhibit significant hearing loss. We relate this deficit to impaired myelination and aberrant synapse formation in the cochlea, which manifest during postnatal development. By contrast, prolonged pharmacological suppression of BACE1 activity in adult wild-type mice did not reproduce the hearing deficit or the cochlear abnormalities of BACE1 null mice.
Collapse
|
23
|
Bertram S, Roll L, Reinhard J, Groß K, Dazert S, Faissner A, Volkenstein S. Pleiotrophin increases neurite length and number of spiral ganglion neurons in vitro. Exp Brain Res 2019; 237:2983-2993. [PMID: 31515588 DOI: 10.1007/s00221-019-05644-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 09/03/2019] [Indexed: 11/28/2022]
Abstract
Acoustic trauma, aging, genetic defects or ototoxic drugs are causes for sensorineural hearing loss involving sensory hair cell death and secondary degeneration of spiral ganglion neurons. Auditory implants are the only available therapy for severe to profound sensorineural hearing loss when hearing aids do not provide a sufficient speech discrimination anymore. Neurotrophic factors represent potential therapeutic candidates to improve the performance of cochlear implants (CIs) by the support of spiral ganglion neurons (SGNs). Here, we investigated the effect of pleiotrophin (PTN), a well-described neurotrophic factor for different types of neurons that is expressed in the postnatal mouse cochlea. PTN knockout mice exhibit severe deficits in auditory brainstem responses, which indicates the importance of PTN in inner ear development and function and makes it a promising candidate to support SGNs. Using organotypic explants and dissociated SGN cultures, we investigated the influence of PTN on the number of neurons, neurite number and neurite length. PTN significantly increased the number and neurite length of dissociated SGNs. We further verified the expression of important PTN-associated receptors in the SG. mRNA of anaplastic lymphoma kinase, αv integrin, β3 integrin, receptor protein tyrosine phosphatase β/ζ, neuroglycan C, low-density lipoprotein receptor-related protein 1 and syndecan 3 was detected in the inner ear. These results suggest that PTN may be a novel candidate to improve sensorineural hearing loss treatment in the future.
Collapse
Affiliation(s)
- Sebastian Bertram
- Department of Otorhinolaryngology, Head and Neck Surgery, St. Elisabeth-Hospital, Ruhr-University Bochum, Bleichstr. 15, 44787, Bochum, Germany
| | - Lars Roll
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Katharina Groß
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Stefan Dazert
- Department of Otorhinolaryngology, Head and Neck Surgery, St. Elisabeth-Hospital, Ruhr-University Bochum, Bleichstr. 15, 44787, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Stefan Volkenstein
- Department of Otorhinolaryngology, Head and Neck Surgery, St. Elisabeth-Hospital, Ruhr-University Bochum, Bleichstr. 15, 44787, Bochum, Germany.
| |
Collapse
|
24
|
Zhang ZJ, Guan HX, Yang K, Xiao BK, Liao H, Jiang Y, Zhou T, Hua QQ. Estimation of the status of spiral ganglion neurons and Schwann cells in the auditory neural degeneration mouse using the auditory brainstem response. Acta Otolaryngol 2018; 138:603-609. [PMID: 29553844 DOI: 10.1080/00016489.2018.1436766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
CONCLUSION The auditory brainstem response (ABR) wave I threshold, latency and amplitude are insensitive to spiral ganglion neurons (SGNs) degeneration, but are sensitive to the degeneration of Schwann cells and can estimate the status of Schwann cells in a neural degeneration mouse model. The thorough pre-operative ABR assessment would be helpful in predicting cochlear implant performance. OBJECTIVES This study aimed in finding a non-invasive electrophysiological method to evaluate the status of the auditory nerve and the Schwann cells in sensorineural hearing loss (SNHL) and auditory neuropathy (AN) ears, and providing useful information for candidates screening and outcome prediction in cochlear implantation. METHODS The frequency-specific acoustic ABR was recorded in mice. The immunohistochemical staining was performed to detect the SGNs and Schwann cells in mice cochlea. The correlations between ABR wave I metrics and SGNs, Schwann cells were investigated. RESULTS In SNHL and AN mice cochlea, statistically significant correlations between ABR wave I thresholds, latencies and amplitudes at 8, 16, and 32 kHz and their corresponding SGNs densities were found only in wave I amplitude at 8 kHz. While the ABR wave I metrics at all three frequencies showed strong significant correlations with their corresponding Schwann cells densities.
Collapse
Affiliation(s)
- Zhi-Jian Zhang
- Department of Otolaryngology – Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hong-Xia Guan
- Department of Otolaryngology – Head and Neck Surgery, Hubei Maternal and Child Health Hospital, Wuhan, China
| | - Kun Yang
- Department of Otolaryngology – Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bo-Kui Xiao
- Department of Otolaryngology – Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hua Liao
- Department of Otolaryngology – Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Jiang
- Department of Otolaryngology – Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tao Zhou
- Department of Otolaryngology – Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing-Quan Hua
- Department of Otolaryngology – Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
25
|
Photopolymerized Microfeatures Guide Adult Spiral Ganglion and Dorsal Root Ganglion Neurite Growth. Otol Neurotol 2018; 39:119-126. [PMID: 29227456 DOI: 10.1097/mao.0000000000001622] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
HYPOTHESIS Microtopographical patterns generated by photopolymerization of methacrylate polymer systems will direct growth of neurites from adult neurons, including spiral ganglion neurons (SGNs). BACKGROUND Cochlear implants (CIs) provide hearing perception to patients with severe to profound hearing loss. However, their ability to encode complex auditory stimuli is limited due, in part, to poor spatial resolution caused by spread of the electrical currents in the inner ear. Directing the regrowth of SGN peripheral processes towards stimulating electrodes could help reduce current spread and improve spatial resolution provided by the CI. Previous work has demonstrated that micro- and nano-scale patterned surfaces precisely guide the growth of neurites from a variety of neonatal neurons including SGNs. Here, we sought to determine the extent to which adult neurons likewise respond to these topographical surface features. METHODS Photopolymerization was used to fabricate methacrylate polymer substrates with micropatterned surfaces of varying amplitudes and periodicities. Dissociated adult dorsal root ganglion neurons (DRGNs) and SGNs were cultured on these surfaces and the alignment of the neurite processes to the micropatterns was determined. RESULTS Neurites from both adult DRGNs and SGNs significantly aligned to the patterned surfaces similar to their neonatal counterparts. Further DRGN and SGN neurite alignment increased as the amplitude of the microfeatures increased. Decreased pattern periodicity also improved neurite alignment. CONCLUSION Microscale surface topographic features direct the growth of adult SGN neurites. Topographical features could prove useful for guiding growth of SGN peripheral axons towards a CI electrode array.
Collapse
|
26
|
Brown LN, Xing Y, Noble KV, Barth JL, Panganiban CH, Smythe NM, Bridges MC, Zhu J, Lang H. Macrophage-Mediated Glial Cell Elimination in the Postnatal Mouse Cochlea. Front Mol Neurosci 2017; 10:407. [PMID: 29375297 PMCID: PMC5770652 DOI: 10.3389/fnmol.2017.00407] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 11/23/2017] [Indexed: 12/20/2022] Open
Abstract
Hearing relies on the transmission of auditory information from sensory hair cells (HCs) to the brain through the auditory nerve. This relay of information requires HCs to be innervated by spiral ganglion neurons (SGNs) in an exclusive manner and SGNs to be ensheathed by myelinating and non-myelinating glial cells. In the developing auditory nerve, mistargeted SGN axons are retracted or pruned and excessive cells are cleared in a process referred to as nerve refinement. Whether auditory glial cells are eliminated during auditory nerve refinement is unknown. Using early postnatal mice of either sex, we show that glial cell numbers decrease after the first postnatal week, corresponding temporally with nerve refinement in the developing auditory nerve. Additionally, expression of immune-related genes was upregulated and macrophage numbers increase in a manner coinciding with the reduction of glial cell numbers. Transient depletion of macrophages during early auditory nerve development, using transgenic CD11bDTR/EGFP mice, resulted in the appearance of excessive glial cells. Macrophage depletion caused abnormalities in myelin formation and transient edema of the stria vascularis. Macrophage-depleted mice also showed auditory function impairment that partially recovered in adulthood. These findings demonstrate that macrophages contribute to the regulation of glial cell number during postnatal development of the cochlea and that glial cells play a critical role in hearing onset and auditory nerve maturation.
Collapse
Affiliation(s)
- LaShardai N. Brown
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Yazhi Xing
- Department of Otorhinolaryngology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai, China
| | - Kenyaria V. Noble
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Jeremy L. Barth
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Clarisse H. Panganiban
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Nancy M. Smythe
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Mary C. Bridges
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Juhong Zhu
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Hainan Lang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
27
|
Tuft BW, Xu L, Leigh B, Lee D, Guymon CA, Hansen MR. Photopolymerized micropatterns with high feature frequencies overcome chemorepulsive borders to direct neurite growth. J Tissue Eng Regen Med 2017; 12:e1392-e1403. [PMID: 28753740 DOI: 10.1002/term.2527] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 07/03/2017] [Accepted: 07/24/2017] [Indexed: 01/04/2023]
Abstract
Developing and regenerating neurites respond to a variety of biophysical and biochemical cues in their micro-environment to reach target cells and establish appropriate synapses. Defining the hierarchal relationship of both types of cues to direct neurite growth carries broad significance for neural development, regeneration, and, in particular, engineering of neural prostheses that improve tissue integration with native neural networks. In this work, chemorepulsive biochemical borders are established on substrates with a range of surface microfeatures to determine the potential of physical cues to overcome conflicting biochemical cues. Physical micropatterns are fabricated using photomasking techniques to spatially control photoinitiation events of the polymerization. Temporal control of the reaction allows for generation of microfeatures with the same amplitude across a range of feature frequencies or periodicities. The micropatterned substrates are then modified with repulsive chemical borders between laminin and either EphA4-Fc or tenascin C that compete with the surface microfeatures to direct neurite growth. Behaviour of neurites from spiral ganglion and trigeminal neurons is characterized at biochemical borders as cross, turn, stop, or repel events. Both the chemical borders and physical patterns significantly influence neurite pathfinding. On unpatterned surfaces, most neurites that originate on laminin are deterred by the border with tenascin C or EphA4-Fc. Importantly, substrates with frequent micropattern features overcome the influence of the chemorepulsive border to dominate neurite trajectory. Designing prosthesis interfaces with appropriate surface features may allow for spatially organized neurite outgrowth in vivo even in the presence of conflicting biochemical cues in native target tissues.
Collapse
Affiliation(s)
- Bradley W Tuft
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA, USA
| | - Linjing Xu
- Department of Otolaryngology - Head and Neck Surgery, University of Iowa, Iowa City, IA, USA
| | - Braden Leigh
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA, USA
| | - Daniel Lee
- Department of Otolaryngology - Head and Neck Surgery, University of Iowa, Iowa City, IA, USA
| | - C Allan Guymon
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA, USA
| | - Marlan R Hansen
- Department of Otolaryngology - Head and Neck Surgery, University of Iowa, Iowa City, IA, USA.,Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
28
|
Leigh BL, Truong K, Bartholomew R, Ramirez M, Hansen MR, Allan Guymon C. Tuning Surface and Topographical Features to Investigate Competitive Guidance of Spiral Ganglion Neurons. ACS APPLIED MATERIALS & INTERFACES 2017; 9:31488-31496. [PMID: 28841276 PMCID: PMC6341486 DOI: 10.1021/acsami.7b09258] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Cochlear Implants (CIs) suffer from limited tonal resolution due, in large part, to spatial separation between stimulating electrode arrays and primary neural receptors. In this work, a combination of physical and chemical micropatterns, formed on acrylate polymers, are used to direct the growth of primary spiral ganglion neurons (SGNs), the inner ear neurons. Utilizing the inherent temporal and spatial control of photopolymerization, physical microgrooves are fabricated using a photomask in a single step process. Biochemical patterns are generated by adsorbing laminin, a cell adhesion protein, to acrylate polymer surfaces followed by irradiation through a photomask with UV light to deactivate protein in exposed areas and generate parallel biochemical patterns. Laminin deactivation was shown increase as a function of UV light exposure while remaining adsorbed to the polymer surface. SGN neurites show alignment to both biochemical and physical patterns when evaluated individually. Competing biochemical and physical patterns were also examined. The relative guiding strength of physical cues was varied by independently changing both the amplitude and the band spacing of the microgrooves, with higher amplitudes and shorter band spacing providing cues that more effective guide neurite growth. SGN neurites aligned to laminin patterns with lower physical pattern amplitude and thus weaker physical cues. Alignment of SGNs shifted toward the physical pattern with higher amplitude and lower periodicity patterns which represent stronger cues. These results demonstrate the ability of photopolymerized microfeatures to modulate alignment of inner ear neurites even in the presence of conflicting physical and biochemical cues laying the groundwork for next generation cochlear implants and neural prosthetic devices.
Collapse
Affiliation(s)
- Braden L. Leigh
- Departments of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Kristy Truong
- Otolaryngology Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Reid Bartholomew
- Otolaryngology Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Mark Ramirez
- Otolaryngology Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Marlan R. Hansen
- Otolaryngology Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
- Neurosurgery, University of Iowa, Iowa City, IA 52242, USA
| | - C. Allan Guymon
- Departments of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
29
|
Leigh BL, Cheng E, Linjing X, Andresen C, Hansen MR, Guymon CA. Photopolymerizable Zwitterionic Polymer Patterns Control Cell Adhesion and Guide Neural Growth. Biomacromolecules 2017; 18:2389-2401. [PMID: 28671816 PMCID: PMC6372952 DOI: 10.1021/acs.biomac.7b00579] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Developing materials that reduce or eliminate fibrosis encapsulation of neural prosthetic implants could significantly enhance implant fidelity by improving the tissue/electrode array interface. Here, we report on the photografting and patterning of two zwitterionic materials, sulfobetaine methacrylate (SBMA) and carboxybetaine methacrylate (CBMA), for controlling the adhesion and directionality of cells relevant to neural prosthetics. CBMA and SBMA polymers were photopolymerized and grafted on glass surfaces then characterized by X-ray photoelectron spectroscopy, water contact angle, and protein adsorption. Micropatterned surfaces were fabricated with alternating zwitterionic and uncoated bands. Fibroblasts, cells prevalent in fibrotic tissue, almost exclusively migrate and grow on uncoated bands with little to no cells present on zwitterionic bands, especially for CBMA-coated surfaces. Astrocytes and Schwann cells showed similarly low levels of cell adhesion and morphology changes when cultured on zwitterionic surfaces. Additionally, Schwann cells and inner ear spiral ganglion neuron neurites aligned well to zwitterionic patterns.
Collapse
Affiliation(s)
- Braden L. Leigh
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Elise Cheng
- Department of Otolaryngology Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Xu Linjing
- Department of Otolaryngology Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Corinne Andresen
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Marlan R. Hansen
- Department of Otolaryngology Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
- Department of Neurosurgery, University of Iowa, Iowa City, IA 52242, USA
| | - C. Allan Guymon
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
30
|
Time-dependent activity of primary auditory neurons in the presence of neurotrophins and antibiotics. Hear Res 2017; 350:122-132. [DOI: 10.1016/j.heares.2017.04.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 03/16/2017] [Accepted: 04/23/2017] [Indexed: 12/19/2022]
|
31
|
Waqas M, Sun S, Xuan C, Fang Q, Zhang X, Islam IU, Qi J, Zhang S, Gao X, Tang M, Shi H, Li H, Chai R. Bone morphogenetic protein 4 promotes the survival and preserves the structure of flow-sorted Bhlhb5+ cochlear spiral ganglion neurons in vitro. Sci Rep 2017; 7:3506. [PMID: 28615657 PMCID: PMC5471210 DOI: 10.1038/s41598-017-03810-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/04/2017] [Indexed: 01/22/2023] Open
Abstract
SGNs are the primary auditory neurons, and damage or loss of SGNs leads to sensorineural hearing loss. BMP4 is a growth factor that belongs to the TGF-β superfamily and has been shown to play a key role during development, but little is known about its effect on postnatal cochlear SGNs in mice. In this study, we used the P3 Bhlhb5-cre/tdTomato transgenic mouse model and FACS to isolate a pure population of Bhlhb5+ SGNs. We found that BMP4 significantly promoted SGN survival after 7 days of culture. We observed fewer apoptotic cells and decreased expression of pro-apoptotic marker genes after BMP4 treatment. We also found that BMP4 promoted monopolar neurite outgrowth of isolated SGNs, and high concentrations of BMP4 preserved the number and the length of neurites in the explant culture of the modiolus harboring the SGNs. We showed that high concentration of BMP4 enhanced neurite growth as determined by the higher average number of filopodia and the larger area of the growth cone. Finally, we found that high concentrations of BMP4 significantly elevated the synapse density of SGNs in explant culture. Thus, our findings suggest that BMP4 has the potential to promote the survival and preserve the structure of SGNs.
Collapse
Affiliation(s)
- Muhammad Waqas
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Department of Biotechnology, Federal Urdu University of Arts, Science and Technology, Gulshan-e-Iqbal campus, Karachi, Pakistan
| | - Shan Sun
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China
| | - Chuanyin Xuan
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Qiaojun Fang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaoli Zhang
- Department of Otolaryngology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Irum-Us Islam
- Department of Biotechnology, Federal Urdu University of Arts, Science and Technology, Gulshan-e-Iqbal campus, Karachi, Pakistan
| | - Jieyu Qi
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Shasha Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xia Gao
- Department of Otolaryngology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Haibo Shi
- Department of Otorhinolaryngology Head & Neck Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200233, China.
| | - Huawei Li
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China. .,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China. .,Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China. .,Shanghai Engineering Research Centre of Cochlear Implants, Shanghai, 200031, China. .,The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China.
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China. .,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China. .,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
32
|
Schwieger J, Esser KH, Lenarz T, Scheper V. Establishment of a long-term spiral ganglion neuron culture with reduced glial cell number: Effects of AraC on cell composition and neurons. J Neurosci Methods 2016; 268:106-16. [DOI: 10.1016/j.jneumeth.2016.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/29/2016] [Accepted: 05/02/2016] [Indexed: 01/13/2023]
|
33
|
Charge-balanced biphasic electrical stimulation inhibits neurite extension of spiral ganglion neurons. Neurosci Lett 2016; 624:92-9. [DOI: 10.1016/j.neulet.2016.04.069] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/17/2016] [Accepted: 04/28/2016] [Indexed: 11/23/2022]
|
34
|
Cochlear afferent innervation development. Hear Res 2015; 330:157-69. [DOI: 10.1016/j.heares.2015.07.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 06/02/2015] [Accepted: 07/21/2015] [Indexed: 01/11/2023]
|
35
|
Bas E, Goncalves S, Adams M, Dinh CT, Bas JM, Van De Water TR, Eshraghi AA. Spiral ganglion cells and macrophages initiate neuro-inflammation and scarring following cochlear implantation. Front Cell Neurosci 2015; 9:303. [PMID: 26321909 PMCID: PMC4532929 DOI: 10.3389/fncel.2015.00303] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 07/23/2015] [Indexed: 12/03/2022] Open
Abstract
Conservation of a patient's residual hearing and prevention of fibrous tissue/new bone formation around an electrode array are some of the major challenges in cochlear implant (CI) surgery. Although it is well-known that fibrotic tissue formation around the electrode array can interfere with hearing performance in implanted patients, and that associated intracochlear inflammation can initiate loss of residual hearing, little is known about the molecular and cellular mechanisms that promote this response in the cochlea. In vitro studies in neonatal rats and in vivo studies in adult mice were performed to gain insight into the pro-inflammatory, proliferative, and remodeling phases of pathological wound healing that occur in the cochlea following an electrode analog insertion. Resident Schwann cells (SC), macrophages, and fibroblasts had a prominent role in the inflammatory process in the cochlea. Leukocytes were recruited to the cochlea following insertion of a nylon filament in adult mice, where contributed to the inflammatory response. The reparative stages in wound healing are characterized by persistent neuro-inflammation of spiral ganglion neurons (SGN) and expression of regenerative monocytes/macrophages in the cochlea. Accordingly, genes involved in extracellular matrix (ECM) deposition and remodeling were up-regulated in implanted cochleae. Maturation of scar tissue occurs in the remodeling phase of wound healing in the cochlea. Similar to other damaged peripheral nerves, M2 macrophages and de-differentiated SC were observed in damaged cochleae and may play a role in cell survival and axonal regeneration. In conclusion, the insertion of an electrode analog into the cochlea is associated with robust early and chronic inflammatory responses characterized by recruitment of leukocytes and expression of pro-inflammatory cytokines that promote intracochlear fibrosis and loss of the auditory hair cells (HC) and SGN important for hearing after CI surgery.
Collapse
Affiliation(s)
- Esperanza Bas
- Department of Otolaryngology, Miller School of Medicine, University of Miami Miami, FL, USA
| | - Stefania Goncalves
- Department of Otolaryngology, Miller School of Medicine, University of Miami Miami, FL, USA
| | - Michelle Adams
- Department of Otolaryngology, Miller School of Medicine, University of Miami Miami, FL, USA
| | - Christine T Dinh
- Department of Otolaryngology, Miller School of Medicine, University of Miami Miami, FL, USA
| | - Jose M Bas
- Department of Otolaryngology, Miller School of Medicine, University of Miami Miami, FL, USA
| | - Thomas R Van De Water
- Department of Otolaryngology, Miller School of Medicine, University of Miami Miami, FL, USA
| | - Adrien A Eshraghi
- Department of Otolaryngology, Miller School of Medicine, University of Miami Miami, FL, USA
| |
Collapse
|
36
|
Tang J, Qian Y, Li H, Kopecky BJ, Ding D, Ou HC, DeCook R, Chen X, Sun Z, Kobel M, Bao J. Canertinib induces ototoxicity in three preclinical models. Hear Res 2015; 328:59-66. [PMID: 26163095 DOI: 10.1016/j.heares.2015.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 06/23/2015] [Accepted: 07/03/2015] [Indexed: 12/16/2022]
Abstract
Neuregulin-1 (NRG1) ligand and its epidermal growth factor receptor (EGFR)/ERBB family regulate normal cellular proliferation and differentiation in many tissues including the cochlea. Aberrant NRG1 and ERBB signaling cause significant hearing impairment in mice. Dysregulation of the same signaling pathway in humans is involved in certain types of cancers such as breast cancer or non-small cell lung cancer (NSCLC). A new irreversible pan-ERBB inhibitor, canertinib, has been tested in clinical trials for the treatment of refractory NSCLC. Its possible ototoxicity was unknown. In this study, a significant dose-dependent canertinib ototoxicity was observed in a zebrafish model. Canertinib ototoxicity was further confirmed in two mouse models with different genetic backgrounds. The data strongly suggested an evolutionally preserved ERBB molecular mechanism underlying canertinib ototoxicity. Thus, these results imply that clinical monitoring of hearing loss should be considered for clinical testing of canertinib or other pan-ERBB inhibitors.
Collapse
Affiliation(s)
- Jian Tang
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Thoracic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yi Qian
- Department of Cardio-Thoracic Surgery, The Third Affiliated Hospital of Nantong University, Wuxi 214041, China
| | - Hui Li
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Benjamin J Kopecky
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dalian Ding
- Center for Hearing and Deafness, State University of New York at Buffalo, 137 Cary Hall, 3435 Main Street, Buffalo, NY 14214, USA
| | - Henry C Ou
- Department of Pediatrics, Seattle Children's Hospital, Seattle, WA, USA; Department of Otolaryngology, University of Washington, Seattle, WA, USA
| | - Rhonda DeCook
- Department of Statistics and Actuarial Science, University of Iowa, Iowa City, IA 52242, USA
| | - Xiaojie Chen
- Gateway Biotechnology Inc., St. Louis, MO 63108, USA
| | - Zhenyu Sun
- Department of Cardio-Thoracic Surgery, The Third Affiliated Hospital of Nantong University, Wuxi 214041, China
| | - Megan Kobel
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Jianxin Bao
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH 44272, USA.
| |
Collapse
|
37
|
Förthmann B, Grothe C, Claus P. A nuclear odyssey: fibroblast growth factor-2 (FGF-2) as a regulator of nuclear homeostasis in the nervous system. Cell Mol Life Sci 2015; 72:1651-62. [PMID: 25552245 PMCID: PMC11113852 DOI: 10.1007/s00018-014-1818-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/10/2014] [Accepted: 12/19/2014] [Indexed: 01/07/2023]
Abstract
Nuclear localization of classical growth factors is a well-known phenomenon but still remains a molecular and cellular conundrum. Fibroblast growth factor-2 (FGF-2) is an excellent example of a protein which functions as an extracellular molecule involved in canonical receptor tyrosine kinase signaling as well as displaying intracellular functions. Paracrine and nuclear functions are two important sides of the same protein. FGF-2 is expressed in isoforms with different molecular weights from one mRNA species. In rodents, all of these isoforms become imported to the nucleus. In this review, we discuss structural and functional aspects of FGF-2 isoforms in the nervous system. The nuclear odyssey of FGF-2 is reflected by nuclear dynamics, localization to nuclear bodies such as nucleoli, binding to chromatin and engagement in various protein interactions. Recently discovered molecular partnerships of the isoforms shed light on their nuclear functions, thereby greatly extending our knowledge of the multifaceted functions of FGF-2.
Collapse
Affiliation(s)
- Benjamin Förthmann
- Department of Neuroanatomy, Institute of Neuroanatomy, Hannover Medical School, OE 4140, Carl-Neuberg-Str.1, 30625 Hannover, Germany
| | - Claudia Grothe
- Department of Neuroanatomy, Institute of Neuroanatomy, Hannover Medical School, OE 4140, Carl-Neuberg-Str.1, 30625 Hannover, Germany
- Center for Systems Neuroscience, 30625 Hannover, Germany
| | - Peter Claus
- Department of Neuroanatomy, Institute of Neuroanatomy, Hannover Medical School, OE 4140, Carl-Neuberg-Str.1, 30625 Hannover, Germany
- Center for Systems Neuroscience, 30625 Hannover, Germany
| |
Collapse
|
38
|
Li S, Tuft BW, Xu L, Polacco MA, Clarke JC, Guymon CA, Hansen MR. Microtopographical features generated by photopolymerization recruit RhoA/ROCK through TRPV1 to direct cell and neurite growth. Biomaterials 2015; 53:95-106. [PMID: 25890710 DOI: 10.1016/j.biomaterials.2015.02.057] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 02/11/2015] [Accepted: 02/13/2015] [Indexed: 01/10/2023]
Abstract
Cell processes, including growth cones, respond to biophysical cues in their microenvironment to establish functional tissue architecture and intercellular networks. The mechanisms by which cells sense and translate biophysical cues into directed growth are unknown. We used photopolymerization to fabricate methacrylate platforms with patterned microtopographical features that precisely guide neurite growth and Schwann cell alignment. Pharmacologic inhibition of the transient receptor potential cation channel subfamily V member 1 (TRPV1) or reduced expression of TRPV1 by RNAi significantly disrupts neurite guidance by these microtopographical features. Exogenous expression of TRPV1 induces alignment of NIH3T3 fibroblasts that fail to align in the absence of TRPV1, further implicating TRPV1 channels as critical mediators of cellular responses to biophysical cues. Microtopographic features increase RhoA activity in growth cones and in TRPV1-expressing NIH3T3 cells. Further, Rho-associated kinase (ROCK) phosphorylation is elevated in growth cones and neurites on micropatterned surfaces. Inhibition of RhoA/ROCK by pharmacological compounds or reduced expression of either ROCKI or ROCKII isoforms by RNAi abolishes neurite and cell alignment, confirming that RhoA/ROCK signaling mediates neurite and cell alignment to microtopographic features. These studies demonstrate that microtopographical cues recruit TRPV1 channels and downstream signaling pathways, including RhoA and ROCK, to direct neurite and cell growth.
Collapse
Affiliation(s)
- Shufeng Li
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Otolaryngology, EYE & ENT Hospital of Fudan University, Shanghai 200031, China
| | - Bradley W Tuft
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Linjing Xu
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Marc A Polacco
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - Joseph C Clarke
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - C Allan Guymon
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Marlan R Hansen
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA; Department of Neurosurgery, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
39
|
Chow CL, Guo W, Trivedi P, Zhao X, Gubbels SP. Characterization of a unique cell population marked by transgene expression in the adult cochlea of nestin-CreER(T2)/tdTomato-reporter mice. J Comp Neurol 2015; 523:1474-87. [PMID: 25611038 DOI: 10.1002/cne.23747] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/18/2014] [Accepted: 01/13/2015] [Indexed: 02/06/2023]
Abstract
Hair cells in the adult mammalian cochlea cannot spontaneously regenerate after damage, resulting in the permanency of hearing loss. Stem cells have been found to be present in the cochlea of young rodents; however, there has been little evidence for their existence into adulthood. We used nestin-CreER(T2)/tdTomato-reporter mice to trace the lineage of putative nestin-expressing cells and their progeny in the cochleae of adult mice. Nestin, an intermediate filament found in neural progenitor cells during early development and adulthood, is regarded as a multipotent and neural stem cell marker. Other investigators have reported its presence in postnatal and young adult rodents; however, there are discrepancies among these reports. Using lineage tracing, we documented a robust population of tdTomato-expressing cells and evaluated these cells at a series of adult time points. Upon activation of the nestin promoter, tdTomato was observed just below and medial to the inner hair cell layer. All cells colocalized with the stem cell and cochlear-supporting-cell marker Sox2 as well as the supporting cell and Schwann cell marker Sox10; however, they did not colocalize with the Schwann cell marker Krox20, spiral ganglion marker NF200, nor glial fibrillary acidic acid (GFAP)-expressing supporting cell marker. The cellular identity of this unique population of tdTomato-expressing cells in the adult cochlea of nestin-CreER(T2)/tdTomato mice remains unclear; however, these cells may represent a type of supporting cell on the neural aspect of the inner hair cell layer.
Collapse
Affiliation(s)
- Cynthia L Chow
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, Wisconsin, 53706.,Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, 53705
| | - Weixiang Guo
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, 53705
| | - Parul Trivedi
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, 53705
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, 53705.,Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Samuel P Gubbels
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, 53705.,Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Wisconsin-Madison, Madison, Wisconsin, 53792
| |
Collapse
|
40
|
Klymov A, Rodrigues Neves CT, te Riet J, Agterberg MJ, Mylanus EA, Snik AF, Jansen JA, Walboomers XF. Nanogrooved Surface-Patterns induce cellular organization and axonal outgrowth in neuron-like PC12-Cells. Hear Res 2015; 320:11-7. [DOI: 10.1016/j.heares.2014.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 10/05/2014] [Accepted: 12/18/2014] [Indexed: 11/16/2022]
|
41
|
Postnatal expression of neurotrophic factors accessible to spiral ganglion neurons in the auditory system of adult hearing and deafened rats. J Neurosci 2014; 34:13110-26. [PMID: 25253857 DOI: 10.1523/jneurosci.1014-14.2014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Spiral ganglion neurons (SGNs) receive input from cochlear hair cells and project from the cochlea to the cochlear nucleus. After destruction of hair cells with aminoglycoside antibiotics or noise, SGNs gradually die. It has been assumed that SGN death is attributable to loss of neurotrophic factors (NTFs) derived from hair cells or supporting cells in the organ of Corti (OC). We used quantitative PCR (qPCR) to assay NTF expression-neurotrophin-3 (NT-3), BDNF, GDNF, neurturin, artemin, and CNTF-in the OC and cochlear nucleus at various ages from postnatal day 0 (P0) to P90 in control hearing and neonatally deafened rats. NT-3, neurturin, and CNTF were most abundant in the postnatal hearing OC; CNTF and neurturin most abundant in the cochlear nucleus. In the OC, NT-3 and CNTF showed a postnatal increase in expression approximately concomitant with hearing onset. In rats deafened by daily kanamycin injections (from P8 to P16), surviving inner hair cells were evident at P16 but absent by P19, with most postsynaptic boutons lost before P16. NT-3 and CNTF, which normally increase postnatally, had significantly reduced expression in the OC of deafened rats, although CNTF was expressed throughout the time that SGNs were dying. In contrast, neurturin expression was constant, unaffected by deafening or by age. CNTF and neurturin expression in the cochlear nucleus was unaffected by deafening or age. Thus, NTFs other than NT-3 are available to SGNs even as they are dying after deafening, apparently conflicting with the hypothesis that SGN death is attributable to lack of NTFs.
Collapse
|
42
|
Tuft BW, Zhang L, Xu L, Hangartner A, Leigh B, Hansen MR, Guymon CA. Material stiffness effects on neurite alignment to photopolymerized micropatterns. Biomacromolecules 2014; 15:3717-27. [PMID: 25211120 PMCID: PMC4195519 DOI: 10.1021/bm501019s] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The ability to direct neurite growth into a close proximity of stimulating elements of a neural prosthesis, such as a retinal or cochlear implant (CI), may enhance device performance and overcome current spatial signal resolution barriers. In this work, spiral ganglion neurons (SGNs), which are the target neurons to be stimulated by CIs, were cultured on photopolymerized micropatterns with varied matrix stiffnesses to determine the effect of rigidity on neurite alignment to physical cues. Micropatterns were generated on methacrylate thin film surfaces in a simple, rapid photopolymerization step by photomasking the prepolymer formulation with parallel line-space gratings. Two methacrylate series, a nonpolar HMA-co-HDDMA series and a polar PEGDMA-co-EGDMA series, with significantly different surface wetting properties were evaluated. Equivalent pattern periodicity was maintained across each methacrylate series based on photomask band spacing, and the feature amplitude was tuned to a depth of 2 μm amplitude for all compositions using the temporal control afforded by the UV curing methodology. The surface morphology was characterized by scanning electron microscopy and white light interferometry. All micropatterned films adsorb similar amounts of laminin from solution, and no significant difference in SGN survival was observed when the substrate compositions were compared. SGN neurite alignment significantly increases with increasing material modulus for both methacrylate series. Interestingly, SGN neurites respond to material stiffness cues that are orders of magnitude higher (GPa) than what is typically ascribed to neural environments (kPa). The ability to understand neurite response to engineered physical cues and mechanical properties such as matrix stiffness will allow the development of advanced biomaterials that direct de novo neurite growth to address the spatial signal resolution limitations of current neural prosthetics.
Collapse
Affiliation(s)
- Bradley W Tuft
- Department of Chemical and Biochemical Engineering, University of Iowa , Iowa City, Iowa 52242, United States
| | | | | | | | | | | | | |
Collapse
|
43
|
Tuft BW, Xu L, White SP, Seline AE, Erwood AM, Hansen MR, Guymon CA. Neural pathfinding on uni- and multidirectional photopolymerized micropatterns. ACS APPLIED MATERIALS & INTERFACES 2014; 6:11265-76. [PMID: 24911660 PMCID: PMC4215840 DOI: 10.1021/am501622a] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 06/09/2014] [Indexed: 05/22/2023]
Abstract
Overcoming signal resolution barriers of neural prostheses, such as the commercially available cochlear impant (CI) or the developing retinal implant, will likely require spatial control of regenerative neural elements. To rationally design materials that direct nerve growth, it is first necessary to determine pathfinding behavior of de novo neurite growth from prosthesis-relevant cells such as spiral ganglion neurons (SGNs) in the inner ear. Accordingly, in this work, repeating 90° turns were fabricated as multidirectional micropatterns to determine SGN neurite turning capability and pathfinding. Unidirectional micropatterns and unpatterned substrates are used as comparisons. Spiral ganglion Schwann cell alignment (SGSC) is also examined on each surface type. Micropatterns are fabricated using the spatial reaction control inherent to photopolymerization with photomasks that have either parallel line spacing gratings for unidirectional patterns or repeating 90° angle steps for multidirectional patterns. Feature depth is controlled by modulating UV exposure time by shuttering the light source at given time increments. Substrate topography is characterized by white light interferometry and scanning electron microscopy (SEM). Both pattern types exhibit features that are 25 μm in width and 7.4 ± 0.7 μm in depth. SGN neurites orient randomly on unpatterned photopolymer controls, align and consistently track unidirectional patterns, and are substantially influenced by, but do not consistently track, multidirectional turning cues. Neurite lengths are 20% shorter on multidirectional substrates compared to unidirectional patterns while neurite branching and microfeature crossing events are significantly higher. For both pattern types, the majority of the neurite length is located in depressed surface features. Developing methods to understand neural pathfinding and to guide de novo neurite growth to specific stimulatory elements will enable design of innovative biomaterials that improve functional outcomes of devices that interface with the nervous system.
Collapse
Affiliation(s)
- Bradley W. Tuft
- Department
of Chemical and Biochemical Engineering, University of Iowa, Iowa City, Iowa 52242,
United States, United States
| | - Linjing Xu
- Department
of Otolaryngology, University of Iowa Hospitals
and Clinics, Iowa City, Iowa 52242, United States, United States
| | - Scott P. White
- Department
of Chemical and Biochemical Engineering, University of Iowa, Iowa City, Iowa 52242,
United States, United States
| | - Alison E. Seline
- Department
of Otolaryngology, University of Iowa Hospitals
and Clinics, Iowa City, Iowa 52242, United States, United States
| | - Andrew M. Erwood
- Department
of Otolaryngology, University of Iowa Hospitals
and Clinics, Iowa City, Iowa 52242, United States, United States
| | - Marlan R. Hansen
- Department
of Otolaryngology, University of Iowa Hospitals
and Clinics, Iowa City, Iowa 52242, United States, United States
| | - C. Allan Guymon
- Department
of Chemical and Biochemical Engineering, University of Iowa, Iowa City, Iowa 52242,
United States, United States
- Tel.:(319)335-5015
| |
Collapse
|
44
|
Diensthuber M, Zecha V, Wagenblast J, Arnhold S, Edge ASB, Stöver T. Spiral ganglion stem cells can be propagated and differentiated into neurons and glia. Biores Open Access 2014; 3:88-97. [PMID: 24940560 PMCID: PMC4048968 DOI: 10.1089/biores.2014.0016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The spiral ganglion is an essential functional component of the peripheral auditory system. Most types of hearing loss are associated with spiral ganglion cell degeneration which is irreversible due to the inner ear's lack of regenerative capacity. Recent studies revealed the existence of stem cells in the postnatal spiral ganglion, which gives rise to the hope that these cells might be useful for regenerative inner ear therapies. Here, we provide an in-depth analysis of sphere-forming stem cells isolated from the spiral ganglion of postnatal mice. We show that spiral ganglion spheres have characteristics similar to neurospheres isolated from the brain. Importantly, spiral ganglion sphere cells maintain their major stem cell characteristics after repeated propagation, which enables the culture of spheres for an extended period of time. In this work, we also demonstrate that differentiated sphere-derived cell populations not only adopt the immunophenotype of mature spiral ganglion cells but also develop distinct ultrastructural features of neurons and glial cells. Thus, our work provides further evidence that self-renewing spiral ganglion stem cells might serve as a promising source for the regeneration of lost auditory neurons.
Collapse
Affiliation(s)
- Marc Diensthuber
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Frankfurt am Main , Goethe University, Frankfurt am Main, Germany . ; Department of Otology and Laryngology, Harvard Medical School , Boston, Massachusetts. ; Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary , Boston, Massachusetts
| | - Veronika Zecha
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Frankfurt am Main , Goethe University, Frankfurt am Main, Germany
| | - Jens Wagenblast
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Frankfurt am Main , Goethe University, Frankfurt am Main, Germany
| | - Stefan Arnhold
- Institute of Veterinary Anatomy, Histology, and Embryology, Justus-Liebig University Giessen , Giessen, Germany
| | - Albert S B Edge
- Department of Otology and Laryngology, Harvard Medical School , Boston, Massachusetts. ; Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary , Boston, Massachusetts. ; Program in Speech and Hearing Bioscience and Technology, Division of Health Sciences and Technology, Harvard and MIT , Cambridge, Massachusetts
| | - Timo Stöver
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Frankfurt am Main , Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
45
|
He Y, Zhang PZ, Sun D, Mi WJ, Zhang XY, Cui Y, Jiang XW, Mao XB, Qiu JH. Wnt1 from cochlear schwann cells enhances neuronal differentiation of transplanted neural stem cells in a rat spiral ganglion neuron degeneration model. Cell Transplant 2013; 23:747-60. [PMID: 23809337 DOI: 10.3727/096368913x669761] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Although neural stem cell (NSC) transplantation is widely expected to become a therapy for nervous system degenerative diseases and injuries, the low neuronal differentiation rate of NSCs transplanted into the inner ear is a major obstacle for the successful treatment of spiral ganglion neuron (SGN) degeneration. In this study, we validated whether the local microenvironment influences the neuronal differentiation of transplanted NSCs in the inner ear. Using a rat SGN degeneration model, we demonstrated that transplanted NSCs were more likely to differentiate into microtubule-associated protein 2 (MAP2)-positive neurons in SGN-degenerated cochleae than in control cochleae. Using real-time quantitative PCR and an immunofluorescence assay, we also proved that the expression of Wnt1 (a ligand of Wnt signaling) increases significantly in Schwann cells in the SGN-degenerated cochlea. We further verified that NSC cultures express receptors and signaling components for Wnts. Based on these expression patterns, we hypothesized that Schwann cell-derived Wnt1 and Wnt signaling might be involved in the regulation of the neuronal differentiation of transplanted NSCs. We verified our hypothesis in vitro using a coculture system. We transduced a lentiviral vector expressing Wnt1 into cochlear Schwann cell cultures and cocultured them with NSC cultures. The coculture with Wnt1-expressing Schwann cells resulted in a significant increase in the percentage of NSCs that differentiated into MAP2-positive neurons, whereas this differentiation-enhancing effect was prevented by Dkk1 (an inhibitor of the Wnt signaling pathway). These results suggested that Wnt1 derived from cochlear Schwann cells enhanced the neuronal differentiation of transplanted NSCs through Wnt signaling pathway activation. Alterations of the microenvironment deserve detailed investigation because they may help us to conceive effective strategies to overcome the barrier of the low differentiation rate of transplanted NSCs.
Collapse
Affiliation(s)
- Ya He
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Photopolymerized microfeatures for directed spiral ganglion neurite and Schwann cell growth. Biomaterials 2012; 34:42-54. [PMID: 23069708 DOI: 10.1016/j.biomaterials.2012.09.053] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 09/23/2012] [Indexed: 12/15/2022]
Abstract
Cochlear implants (CIs) provide auditory perception to individuals with severe hearing impairment. However, their ability to encode complex auditory stimuli is limited due, in part, to poor spatial resolution caused by electrical current spread in the inner ear. Directing nerve cell processes towards target electrodes may reduce the problematic current spread and improve stimulatory specificity. In this work, photopolymerization was used to fabricate micro- and nano-patterned methacrylate polymers to probe the extent of spiral ganglion neuron (SGN) neurite and Schwann cell (SGSC) contact guidance based on variations in substrate topographical cues. Micropatterned substrates are formed in a rapid, single-step reaction by selectively blocking light with photomasks which have parallel line-space gratings with periodicities of 10-100 μm. Channel amplitudes of 250 nm-10 μm are generated by modulating UV exposure time, light intensity, and photoinitiator concentration. Gradual transitions are observed between ridges and grooves using scanning electron and atomic force microscopy. The transitions stand in contrast to vertical features generated via etching lithographic techniques. Alignment of neural elements increases significantly with increasing feature amplitude and constant periodicity, as well as with decreasing periodicity and constant amplitude. SGN neurite alignment strongly correlates (r = 0.93) with maximum feature slope. Multiple neuronal and glial types orient to the patterns with varying degrees of alignment. This work presents a method to fabricate gradually-sloping micropatterns for cellular contact guidance studies and demonstrates spatial control of inner ear neural elements in response to micro- and nano-scale surface topography.
Collapse
|
47
|
Green SH, Bailey E, Wang Q, Davis RL. The Trk A, B, C's of Neurotrophins in the Cochlea. Anat Rec (Hoboken) 2012; 295:1877-95. [DOI: 10.1002/ar.22587] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 07/24/2012] [Indexed: 12/20/2022]
|
48
|
Needham K, Nayagam BA, Minter RL, O'Leary SJ. Combined application of brain-derived neurotrophic factor and neurotrophin-3 and its impact on spiral ganglion neuron firing properties and hyperpolarization-activated currents. Hear Res 2012; 291:1-14. [PMID: 22796476 DOI: 10.1016/j.heares.2012.07.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 06/29/2012] [Accepted: 07/03/2012] [Indexed: 01/11/2023]
Abstract
Neurotrophins provide an effective tool for the rescue and regeneration of spiral ganglion neurons (SGNs) following sensorineural hearing loss. However, these nerve growth factors are also potent modulators of ion channel activity and expression, and in the peripheral auditory system brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT3) have previously been shown to alter the firing properties of auditory neurons and differentially regulate the expression of some potassium channels in vitro. In this study we examined the activity of the hyperpolarization-mediated mixed-cation current (I(h)) in early post-natal cultured rat SGNs following exposure to combined BDNF and NT3. Whole-cell patch-clamp recordings made after 1 or 2 days in vitro revealed no change in the firing adaptation of neurons in the presence of BDNF and NT3. Resting membrane potentials were also maintained, but spike latency and firing threshold was subject to regulation by both neurotrophins and time in vitro. Current clamp recordings revealed an activity profile consistent with activation of the hyperpolarization-activated current. Rapid membrane hyperpolarization was followed by a voltage- and time-dependent depolarizing voltage sag. In voltage clamp, membrane hyperpolarization evoked a slowly-activating inward current that was reversibly blocked with cesium and inhibited by ZD7288. The amplitude and current density of I(h) was significantly larger in BDNF and NT3 supplemented cultures, but this did not translate to a significant alteration in voltage sag magnitude. Neurotrophins provided at 50 ng/ml produced a hyperpolarizing shift in the voltage-dependence and slower time course of I(h) activation compared to SGNs in control groups or cultured with 10 ng/ml BDNF and NT3. Our results indicate that combined BDNF and NT3 increase the activity of hyperpolarization-activated currents and that the voltage-dependence and activation kinetics of I(h) in SGNs are sensitive to changes in neurotrophin concentration. In addition, BDNF and NT3 applied together induce a decrease in firing threshold, but does not generate a shift in firing adaptation.
Collapse
Affiliation(s)
- Karina Needham
- Department of Otolaryngology, University of Melbourne, Royal Victorian Eye & Ear Hospital, Level 2, 32 Gisborne St., East Melbourne, Victoria 3002, Australia.
| | | | | | | |
Collapse
|
49
|
Palmgren B, Jiao Y, Novozhilova E, Stupp SI, Olivius P. Survival, migration and differentiation of mouse tau-GFP embryonic stem cells transplanted into the rat auditory nerve. Exp Neurol 2012; 235:599-609. [DOI: 10.1016/j.expneurol.2012.03.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Revised: 02/18/2012] [Accepted: 03/25/2012] [Indexed: 01/13/2023]
|
50
|
Xing Y, Samuvel DJ, Stevens SM, Dubno JR, Schulte BA, Lang H. Age-related changes of myelin basic protein in mouse and human auditory nerve. PLoS One 2012; 7:e34500. [PMID: 22496821 PMCID: PMC3320625 DOI: 10.1371/journal.pone.0034500] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 03/06/2012] [Indexed: 11/19/2022] Open
Abstract
Age-related hearing loss (presbyacusis) is the most common type of hearing impairment. One of the most consistent pathological changes seen in presbyacusis is the loss of spiral ganglion neurons (SGNs). Defining the cellular and molecular basis of SGN degeneration in the human inner ear is critical to gaining a better understanding of the pathophysiology of presbyacusis. However, information on age-related cellular and molecular alterations in the human spiral ganglion remains scant, owing to the very limited availably of human specimens suitable for high resolution morphological and molecular analysis. This study aimed at defining age-related alterations in the auditory nerve in human temporal bones and determining if immunostaining for myelin basic protein (MBP) can be used as an alternative approach to electron microscopy for evaluating myelin degeneration. For comparative purposes, we evaluated ultrastructural alternations and changes in MBP immunostaining in aging CBA/CaJ mice. We then examined 13 temporal bones from 10 human donors, including 4 adults aged 38-46 years (middle-aged group) and 6 adults aged 63-91 years (older group). Similar to the mouse, intense immunostaining of MBP was present throughout the auditory nerve of the middle-aged human donors. Significant declines in MBP immunoreactivity and losses of MBP(+) auditory nerve fibers were observed in the spiral ganglia of both the older human and aged mouse ears. This study demonstrates that immunostaining for MBP in combination with confocal microscopy provides a sensitive, reliable, and efficient method for assessing alterations of myelin sheaths in the auditory nerve. The results also suggest that myelin degeneration may play a critical role in the SGN loss and the subsequent decline of the auditory nerve function in presbyacusis.
Collapse
Affiliation(s)
- Yazhi Xing
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Devadoss J. Samuvel
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Shawn M. Stevens
- Department of Otolaryngology – Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Judy R. Dubno
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Department of Otolaryngology – Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Bradley A. Schulte
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Department of Otolaryngology – Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Hainan Lang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- * E-mail:
| |
Collapse
|