1
|
Thulasiram MR, Yamamoto R, Olszewski RT, Gu S, Morell RJ, Hoa M, Dabdoub A. Molecular differences between young and mature stria vascularis from organotypic explants and transcriptomics. iScience 2025; 28:111832. [PMID: 40028281 PMCID: PMC11869990 DOI: 10.1016/j.isci.2025.111832] [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: 04/05/2024] [Revised: 07/31/2024] [Accepted: 01/15/2025] [Indexed: 03/05/2025] Open
Abstract
The stria vascularis (SV) is an essential component of the inner ear that regulates the ionic environment required for hearing. SV degeneration disrupts cochlear homeostasis, leading to irreversible hearing loss, yet a comprehensive understanding of the SV, and consequently therapeutic availability for SV degeneration, is lacking. We developed a whole-tissue explant model from neonatal and mature mice to create a platform for advancing SV research. We validated our model by demonstrating that the proliferative behavior of the SV in vitro mimics SV in vivo. We also provided evidence for pharmacological experimentation by investigating the role of Wnt/β-catenin signaling in SV proliferation. Finally, we performed single-cell RNA sequencing from in vivo neonatal and mature mouse SV and surrounding tissue and revealed key genes and pathways that may play a role in SV proliferation and maintenance. Together, our results contribute new insights into investigating biological solutions for SV-associated hearing loss.
Collapse
Affiliation(s)
- Matsya Ruppari Thulasiram
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ryosuke Yamamoto
- Biological Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre Toronto, ON M4N 3M5, Canada
| | - Rafal T. Olszewski
- Auditory Development and Restoration Program, NIDCD Otolaryngology-Surgeon-Scientist Program, NIDCD Neurotology Branch, Division of Intramural Research, National Institutes on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shoujun Gu
- Auditory Development and Restoration Program, NIDCD Otolaryngology-Surgeon-Scientist Program, NIDCD Neurotology Branch, Division of Intramural Research, National Institutes on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert J. Morell
- NIDCD/NIDCR Genomics and Computational Biology Core, National Institutes of Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael Hoa
- Auditory Development and Restoration Program, NIDCD Otolaryngology-Surgeon-Scientist Program, NIDCD Neurotology Branch, Division of Intramural Research, National Institutes on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Otolaryngology-Head and Neck Surgery, Georgetown University School of Medicine, Washington, DC 20007, USA
| | - Alain Dabdoub
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Biological Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre Toronto, ON M4N 3M5, Canada
- Department of Otolaryngology–Head and Neck Surgery, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
2
|
Thulasiram MR, Yamamoto R, Olszewski RT, Gu S, Morell RJ, Hoa M, Dabdoub A. Molecular differences between neonatal and adult stria vascularis from organotypic explants and transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590986. [PMID: 38712156 PMCID: PMC11071502 DOI: 10.1101/2024.04.24.590986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Summary The stria vascularis (SV), part of the blood-labyrinth barrier, is an essential component of the inner ear that regulates the ionic environment required for hearing. SV degeneration disrupts cochlear homeostasis, leading to irreversible hearing loss, yet a comprehensive understanding of the SV, and consequently therapeutic availability for SV degeneration, is lacking. We developed a whole-tissue explant model from neonatal and adult mice to create a robust platform for SV research. We validated our model by demonstrating that the proliferative behaviour of the SV in vitro mimics SV in vivo, providing a representative model and advancing high-throughput SV research. We also provided evidence for pharmacological intervention in our system by investigating the role of Wnt/β-catenin signaling in SV proliferation. Finally, we performed single-cell RNA sequencing from in vivo neonatal and adult mouse SV and revealed key genes and pathways that may play a role in SV proliferation and maintenance. Together, our results contribute new insights into investigating biological solutions for SV-associated hearing loss. Significance Hearing loss impairs our ability to communicate with people and interact with our environment. This can lead to social isolation, depression, cognitive deficits, and dementia. Inner ear degeneration is a primary cause of hearing loss, and our study provides an in depth look at one of the major sites of inner ear degeneration: the stria vascularis. The stria vascularis and associated blood-labyrinth barrier maintain the functional integrity of the auditory system, yet it is relatively understudied. By developing a new in vitro model for the young and adult stria vascularis and using single cell RNA sequencing, our study provides a novel approach to studying this tissue, contributing new insights and widespread implications for auditory neuroscience and regenerative medicine. Highlights - We established an organotypic explant system of the neonatal and adult stria vascularis with an intact blood-labyrinth barrier. - Proliferation of the stria vascularis decreases with age in vitro , modelling its proliferative behaviour in vivo . - Pharmacological studies using our in vitro SV model open possibilities for testing injury paradigms and therapeutic interventions. - Inhibition of Wnt signalling decreases proliferation in neonatal stria vascularis.- We identified key genes and transcription factors unique to developing and mature SV cell types using single cell RNA sequencing.
Collapse
|
3
|
Hasegawa M, Yamamoto Y, Fujisawa A, Kashiba M. Prosaposin is a novel coenzyme Q10-binding protein. J Clin Biochem Nutr 2024; 74:108-112. [PMID: 38510690 PMCID: PMC10948348 DOI: 10.3164/jcbn.23-57] [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: 07/14/2023] [Accepted: 10/28/2023] [Indexed: 03/22/2024] Open
Abstract
Coenzyme Q10 (CoQ10) is essential for mitochondrial ATP production and functions as an important antioxidant in every biomembrane and lipoprotein. Due to its hydrophobicity, a binding and transfer protein for CoQ10 is plausible, and we previously described saposin B as a CoQ10-binding and transfer protein. Here, we report that prosaposin, the precursor of saposin B, also binds CoQ10. As prosaposin is both a secretory protein and integral membrane protein, it is ubiquitous in the body. Prosaposin was isolated from human seminal plasma, and CoQ10 was extracted from hexane solution into the water phase. It was additionally found that immunoprecipitates of mouse brain cytosol generated using two different anti-prosaposin antibodies contained coenzyme Q9. Furthermore, mouse liver cytosol and mouse kidney cytosol also contained prosaposin-coenzyme Q9 complex. These results suggest that prosaposin binds CoQ10 in human cells and body fluids. The significance and role of the Psap-CoQ10 complex in vivo is also discussed.
Collapse
Affiliation(s)
- Makoto Hasegawa
- School of Bionics, Tokyo University of Technology, 1404-1 Katakura-machi, Hachioji, Tokyo 192-0982, Japan
| | - Yorihiro Yamamoto
- School of Bionics, Tokyo University of Technology, 1404-1 Katakura-machi, Hachioji, Tokyo 192-0982, Japan
| | - Akio Fujisawa
- School of Bionics, Tokyo University of Technology, 1404-1 Katakura-machi, Hachioji, Tokyo 192-0982, Japan
| | - Misato Kashiba
- School of Bionics, Tokyo University of Technology, 1404-1 Katakura-machi, Hachioji, Tokyo 192-0982, Japan
| |
Collapse
|
4
|
Sohel MSH, Atoji Y, Onouchi S, Saito S. Expression patterns of prosaposin and neurotransmitter-related molecules in the chick paratympanic organ. Tissue Cell 2023; 83:102130. [PMID: 37320868 DOI: 10.1016/j.tice.2023.102130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/01/2023] [Accepted: 06/08/2023] [Indexed: 06/17/2023]
Abstract
The paratympanic organ (PTO) is a small sense organ in the middle ear of birds that contains hair cells similar to those found in vestibuloauditory organs and receives afferent fibers from the geniculate ganglion. To consider the histochemical similarities between the PTO and vestibular hair cells, we examined the expression patterns of representative molecules in vestibular hair cells, including prosaposin, G protein-coupled receptor (GPR) 37 and GPR37L1 as prosaposin receptors, vesicular glutamate transporter (vGluT) 2 and vGluT3, nicotinic acetylcholine receptor subunit α9 (nAChRα9), and glutamic acid decarboxylase (GAD) 65 and GAD67, in the postnatal day 0 chick PTO and geniculate ganglion by in situ hybridization. Prosaposin mRNA was observed in PTO hair cells, supporting cells, and geniculate ganglion cells. vGluT3 mRNA was observed in PTO hair cells, whereas vGluT2 was observed in a small number of ganglion cells. nAChRα9 mRNA was observed in a small number of PTO hair cells. The results suggest that the histochemical character of PTO hair cells is more similar to that of vestibular hair cells than that of auditory hair cells in chicks.
Collapse
Affiliation(s)
- Md Shahriar Hasan Sohel
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Yasuro Atoji
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Sawa Onouchi
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Shouichiro Saito
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan.
| |
Collapse
|
5
|
Kitamura K, Homma T, Sohel MSH, Fuyuki A, Miyawaki S, Onouchi S, Saito S. Expression patterns of prosaposin and its receptors, G protein-coupled receptor (GPR) 37 and GPR37L1, in the mouse olfactory organ. Tissue Cell 2023; 82:102093. [PMID: 37075680 DOI: 10.1016/j.tice.2023.102093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/24/2023] [Accepted: 04/14/2023] [Indexed: 04/21/2023]
Abstract
Prosaposin is a glycoprotein conserved widely in vertebrates, because it is a precursor for saposins that are required for normal lysosomal function and thus for autophagy, and acts as a neurotrophic factor. Most tetrapods possess two kinds of olfactory neuroepithelia, namely, the olfactory epithelium (OE) and the vomeronasal epithelium (VNE). This study examined the expression patterns of prosaposin and its candidate receptors, G protein-coupled receptor (GPR) 37 and GPR37L1, in mouse OE and VNE by immunofluorescence and in situ hybridization. Prosaposin immunoreactivity was observed in the olfactory receptor neurons, vomeronasal receptor neurons, Bowman's gland (BG), and Jacobson's gland (JG). Prosaposin expression was mainly observed in mature neurons. Prosaposin mRNA expression was observed not only in these cells but also in the apical region of the VNE. GPR37 and GPR37L1 immunoreactivities were found only in the BG and/or the JG. Prosaposin was suggested to secrete and facilitate the autophagic activities of the neurons and modulate the mucus secretion in mouse olfactory organ.
Collapse
Affiliation(s)
- Kai Kitamura
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Takeshi Homma
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, Japan
| | - Md Shahriar Hasan Sohel
- Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, Japan
| | - Aimi Fuyuki
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Shingo Miyawaki
- Laboratory of Veterinary Surgery, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Sawa Onouchi
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan; Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, Japan
| | - Shouichiro Saito
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan; Laboratory of Veterinary Anatomy, Joint Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu, Japan.
| |
Collapse
|
6
|
Fuyuki A, Yamamoto S, Sohel MSH, Homma T, Kitamura K, Onouchi S, Saito S. Expression of prosaposin and its G protein-coupled receptor (GPR) 37 in mouse cochlear and vestibular nuclei. J Vet Med Sci 2023; 85:266-270. [PMID: 36696997 PMCID: PMC10076190 DOI: 10.1292/jvms.22-0552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Prosaposin is a precursor of lysosomal hydrolases activator proteins, saposins, and also acts as a secretory protein that is not processed into saposins. Prosaposin elicits neurotrophic function via G protein-coupled receptor (GPR) 37, and prosaposin deficiency causes abnormal vestibuloauditory end-organ development. In this study, immunohistochemistry was used to examine prosaposin and GPR37 expression patterns in the mouse cochlear and vestibular nuclei. Prosaposin immunoreactivity was observed in neurons and glial cells in both nuclei. GPR37 immunoreactivity was observed in only some neurons, and its immunoreactivity in the vestibular nucleus was weaker than that in the cochlear nucleus. This study suggests a possibility that prosaposin deficiency affects not only the end-organs but also the first center of the vestibuloauditory system.
Collapse
Affiliation(s)
- Aimi Fuyuki
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Saki Yamamoto
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Md Shahriar Hasan Sohel
- Laboratory of Veterinary Anatomy, The Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| | - Takeshi Homma
- Laboratory of Veterinary Anatomy, The Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| | - Kai Kitamura
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Sawa Onouchi
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan.,Laboratory of Veterinary Anatomy, The Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| | - Shouichiro Saito
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan.,Laboratory of Veterinary Anatomy, The Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| |
Collapse
|
7
|
Bhatt IS, Wilson N, Dias R, Torkamani A. A genome-wide association study of tinnitus reveals shared genetic links to neuropsychiatric disorders. Sci Rep 2022; 12:22511. [PMID: 36581688 PMCID: PMC9800371 DOI: 10.1038/s41598-022-26413-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/14/2022] [Indexed: 12/30/2022] Open
Abstract
Tinnitus, a phantom perception of sound in the absence of any external sound source, is a prevalent health condition often accompanied by psychiatric comorbidities. Recent genome-wide association studies (GWAS) highlighted a polygenic nature of tinnitus susceptibility. A shared genetic component between tinnitus and psychiatric conditions remains elusive. Here we present a GWAS using the UK Biobank to investigate the genetic processes linked to tinnitus and tinnitus-related distress, followed by gene-set enrichment analyses. The UK Biobank sample comprised 132,438 individuals with tinnitus and genotype data. Among the study sample, 38,525 individuals reported tinnitus, and 26,889 participants mentioned they experienced tinnitus-related distress in daily living. The genome-wide association analyses were conducted on tinnitus and tinnitus-related distress. We conducted enrichment analyses using FUMA to further understand the genetic processes linked to tinnitus and tinnitus-related distress. A genome-wide significant locus (lead SNP: rs71595470) for tinnitus was obtained in the vicinity of GPM6A. Nineteen independent loci reached suggestive association with tinnitus. Fifteen independent loci reached suggestive association with tinnitus-related distress. The enrichment analysis revealed a shared genetic component between tinnitus and psychiatric traits, such as bipolar disorder, feeling worried, cognitive ability, fast beta electroencephalogram, and sensation seeking. Metabolic, cardiovascular, hematological, and pharmacological gene sets revealed a significant association with tinnitus. Anxiety and stress-related gene sets revealed a significant association with tinnitus-related distress. The GWAS signals for tinnitus were enriched in the hippocampus and cortex, and for tinnitus-related distress were enriched in the brain and spinal cord. This study provides novel insights into genetic processes associated with tinnitus and tinnitus-related distress and demonstrates a shared genetic component underlying tinnitus and psychiatric conditions. Further collaborative attempts are necessary to identify genetic components underlying the phenotypic heterogeneity in tinnitus and provide biological insight into the etiology.
Collapse
Affiliation(s)
- Ishan Sunilkumar Bhatt
- grid.214572.70000 0004 1936 8294Department of Communication Sciences & Disorders, University of Iowa, 250 Hawkins Dr, Iowa City, IA 52242 USA
| | - Nicholas Wilson
- Department of Integrative Structural and Computational Biology Scripps Science Institute, La Jolla, CA 92037 USA
| | - Raquel Dias
- grid.15276.370000 0004 1936 8091Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32608 USA
| | - Ali Torkamani
- Department of Integrative Structural and Computational Biology Scripps Science Institute, La Jolla, CA 92037 USA
| |
Collapse
|
8
|
Outer Hair Cell Glutamate Signaling through Type II Spiral Ganglion Afferents Activates Neurons in the Cochlear Nucleus in Response to Nondamaging Sounds. J Neurosci 2021; 41:2930-2943. [PMID: 33574178 DOI: 10.1523/jneurosci.0619-20.2021] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 01/24/2021] [Accepted: 02/04/2021] [Indexed: 11/21/2022] Open
Abstract
Cochlear outer hair cells (OHCs) are known to uniquely participate in auditory processing through their electromotility, and like inner hair cells, are also capable of releasing vesicular glutamate onto spiral ganglion (SG) neurons: in this case, onto the sparse Type II SG neurons. However, unlike glutamate signaling at the inner hair cell-Type I SG neuron synapse, which is robust across a wide spectrum of sound intensities, glutamate signaling at the OHC-Type II SG neuron synapse is weaker and has been hypothesized to occur only at intense, possibly damaging sound levels. Here, we tested the ability of the OHC-Type II SG pathway to signal to the brain in response to moderate, nondamaging sound (80 dB SPL) as well as to intense sound (115 dB SPL). First, we determined the VGluTs associated with OHC signaling and then confirmed the loss of glutamatergic synaptic transmission from OHCs to Type II SG neurons in KO mice using dendritic patch-clamp recordings. Next, we generated genetic mouse lines in which vesicular glutamate release occurs selectively from OHCs, and then assessed c-Fos expression in the cochlear nucleus in response to sound. From these analyses, we show, for the first time, that glutamatergic signaling at the OHC-Type II SG neuron synapse is capable of activating cochlear nucleus neurons, even at moderate sound levels.SIGNIFICANCE STATEMENT Evidence suggests that cochlear outer hair cells (OHCs) release glutamate onto Type II spiral ganglion neurons only when exposed to loud sound, and that Type II neurons are activated by tissue damage. Knowing whether moderate level sound, without tissue damage, activates this pathway has functional implications for this fundamental auditory pathway. We first determined that OHCs rely largely on VGluT3 for synaptic glutamate release. We then used a genetically modified mouse line in which OHCs, but not inner hair cells, release vesicular glutamate to demonstrate that moderate sound exposure activates cochlear nucleus neurons via the OHC-Type II spiral ganglion pathway. Together, these data indicate that glutamate signaling at the OHC-Type II afferent synapse participates in auditory function at moderate sound levels.
Collapse
|
9
|
Abstract
Cochlear gene therapy has made tremendous strides over the past 5 years. The first study documenting successful restoration of congenital hearing loss using AAV-mediated gene therapy occurred in a mouse model of deafness lacking vesicular glutamate transporter 3 (VGLUT 3). This study utilized a trans-bulla round window membrane (RWM) delivery approach. Since this study, these methodologies have been applied to a number of other mouse models of genetic deafness with varying degrees of success, lending promise for future clinical application of this burgeoning technology. Here we describe a method of virally mediated gene delivery into the cochlear scala tympani through the RWM. This method involves negligible damage to essential structures of the middle and inner ear while preserving hearing. The efficacy of this surgical technique will be demonstrated by the restoration of hearing to the VGLUT3 knockout mice (a mouse model of congenital deafness) after delivery of VGLUT3 gene to the inner ear using an adeno-associated virus as a vector.
Collapse
|
10
|
Akil O, Blits B, Lustig LR, Leake PA. Virally Mediated Overexpression of Glial-Derived Neurotrophic Factor Elicits Age- and Dose-Dependent Neuronal Toxicity and Hearing Loss. Hum Gene Ther 2018; 30:88-105. [PMID: 30183384 DOI: 10.1089/hum.2018.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Contemporary cochlear implants (CI) are generally very effective for remediation of severe to profound sensorineural hearing loss, but outcomes are still highly variable. Auditory nerve survival is likely one of the major factors underlying this variability. Neurotrophin therapy therefore has been proposed for CI recipients, with the goal of improving outcomes by promoting improved survival of cochlear spiral ganglion neurons (SGN) and/or residual hair cells. Previous studies have shown that glial-derived neurotrophic factor (GDNF), brain-derived neurotrophic factor, and neurotrophin-3 can rescue SGNs following insult. The current study was designed to determine whether adeno-associated virus vector serotype 5 (AAV-5) encoding either green fluorescent protein or GDNF can transduce cells in the mouse cochlea to express useful levels of neurotrophin and to approximate the optimum therapeutic dose(s) for transducing hair cells and SGN. The findings demonstrate that AAV-5 is a potentially useful gene therapy vector for the cochlea, resulting in extremely high levels of transgene expression in the cochlear inner hair cells and SGN. However, overexpression of human GDNF in newborn mice caused severe neurological symptoms and hearing loss, likely due to Purkinje cell loss and cochlear nucleus pathology. Thus, extremely high levels of transgene protein expression should be avoided, particularly for proteins that have neurological function in neonatal subjects.
Collapse
Affiliation(s)
- Omar Akil
- 1 Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, California
| | - Bas Blits
- 2 Department of Research and Development, UniQure Biopharma B.V., Amsterdam, The Netherlands
| | - Lawrence R Lustig
- 3 Department of Otolaryngology-Head and Neck Surgery, Columbia University Medical Center, New York, New York
| | - Patricia A Leake
- 1 Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, California
| |
Collapse
|
11
|
Gangliosides and hearing. Biochim Biophys Acta Gen Subj 2017; 1861:2485-2493. [PMID: 28571946 DOI: 10.1016/j.bbagen.2017.05.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/25/2017] [Accepted: 05/25/2017] [Indexed: 01/12/2023]
Abstract
Severe auditory impairment observed in GM3 synthase-deficient mice and humans indicates that glycosphingolipids, especially sialic-acid containing gangliosides, are indispensable for hearing. Gangliosides associate with glycoproteins to form membrane microdomains, the composition of which plays a special role in maintaining the structural and functional integrity of hair cells. These microdomains, also called lipid rafts, connect with intracellular signaling and cytoskeletal systems to link cellular responses to environmental cues. During development, ganglioside species are expressed in distinctive spatial and temporal patterns throughout the cochlea. In both mice and humans, blocking particular steps of ganglioside metabolism produces distinctive neurological and auditory phenotypes. Thus each ganglioside species may have specific, non-overlapping functions within the cochlea, central auditory network, and brain.
Collapse
|
12
|
Abstract
The auditory brainstem response (ABR) test provides information about the inner ear (cochlea) and the central pathways for hearing. The ABR reflects the electrical responses of both the cochlear ganglion neurons and the nuclei of the central auditory pathway to sound stimulation (Zhou et al., 2006; Burkard et al., 2007). The ABR contains 5 identifiable wave forms, labeled as I-V. Wave I represents the summated response from the spiral ganglion and auditory nerve while waves II-V represent responses from the ascending auditory pathway. The ABR is recorded via electrodes placed on the scalp of an anesthetized animal. ABR thresholds refer to the lowest sound pressure level (SPL) that can generate identifiable electrical response waves. This protocol describes the process of measuring the ABR of small rodents (mouse, rat, guinea pig, etc.), including anesthetizing the mouse, placing the electrodes on the scalp, recording click and tone burst stimuli and reading the obtained waveforms for ABR threshold values. As technology continues to evolve, ABR will likely provide more qualitative and quantitative information regarding the function of the auditory nerve and brainstem pathways involved in hearing.
Collapse
Affiliation(s)
- Omar Akil
- Department of Otolaryngology-Head & Neck Surgery, University of California San Francisco, San Francisco, USA
| | - A E Oursler
- Department of Otolaryngology-Head & Neck Surgery, University of California San Francisco, San Francisco, USA
| | - Kevin Fan
- Department of Otolaryngology-Head & Neck Surgery, University of California San Francisco, San Francisco, USA
| | - Lawrence R Lustig
- Department of Otolaryngology-Head & Neck Surgery, University of California San Francisco, San Francisco, USA
| |
Collapse
|
13
|
Lustig LR, Alemi S, Sun Y, Grabowski G, Akil O. Role of saposin C and D in auditory and vestibular function. Laryngoscope 2015. [PMID: 26198053 DOI: 10.1002/lary.25479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVES/HYPOTHESIS Saposins are small proteins derived from a precursor protein, prosaposin. Each of the four saposins (A-D) is necessary for the activity of lysosomal glycosphingolipid hydrolases. Individual saposin mutations lead to lysosomal storage diseases, some of which are associated with hearing loss. Here we evaluate the effects of the loss of saposins C and D on auditory and vestibular function in transgenic mice. METHODS Transgenic mice with either loss of saposin C function or a combined loss of saposin C + D function were studied. Light microscopy and immunofluorescence were used to evaluate histologic and morphologic changes in the auditory and vestibular organs. Acoustic brainstem response thresholds and distortion product otoacoustic emissions were used to study the auditory phenotype. RESULTS A null mutation of saposin C did not result in any identifiable histologic changes or loss of hearing through postnatal day 55. Combined losses of saposins C and D similarly did not result in any changes in organ of Corti histology or loss of hearing. However, inclusions within the vestibular end organs was noted, consistent with afferent and efferent neuronal sprouting, although to a much milder degree than seen in the previously studied prosaposin knockout mouse. CONCLUSIONS Loss of saposin C and D function, although causing mild phenotypic changes in the vestibular end organs, otherwise results in minimal functional impairment and no changes in the auditory system. It is more likely that the auditory and vestibular effects of the loss of prosaposin are mediated through the actions of saposin A and/or B. LEVEL OF EVIDENCE NA.
Collapse
Affiliation(s)
- Lawrence R Lustig
- Department of Otolaryngology-Head & Neck Surgery, Columbia University Medical Center, New York, New York
| | - Sean Alemi
- Department of Otolaryngology-Head & Neck Surgery, University of California, San Francisco, San Francisco, California
| | - Ying Sun
- Department of Human Genetics, University of Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, U.S.A
| | - Gregory Grabowski
- Department of Human Genetics, University of Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, U.S.A
| | - Omar Akil
- Department of Otolaryngology-Head & Neck Surgery, University of California, San Francisco, San Francisco, California
| |
Collapse
|
14
|
Spiral ganglion degeneration and hearing loss as a consequence of satellite cell death in saposin B-deficient mice. J Neurosci 2015; 35:3263-75. [PMID: 25698761 DOI: 10.1523/jneurosci.3920-13.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Saposin B (Sap B) is an essential activator protein for arylsulfatase A in the hydrolysis of sulfatide, a lipid component of myelin. To study Sap B's role in hearing and balance, a Sap B-deficient (B(-/-)) mouse was evaluated. At both light and electron microscopy (EM) levels, inclusion body accumulation was seen in satellite cells surrounding spiral ganglion (SG) neurons from postnatal month 1 onward, progressing into large vacuoles preceding satellite cell degeneration, and followed by SG degeneration. EM also revealed reduced or absent myelin sheaths in SG neurons from postnatal month 8 onwards. Hearing loss was initially seen at postnatal month 6 and progressed thereafter for frequency-specific stimuli, whereas click responses became abnormal from postnatal month 13 onward. The progressive hearing loss correlated with the accumulation of inclusion bodies in the satellite cells and their subsequent degeneration. Outer hair cell numbers and efferent function measures (distortion product otoacoustic emissions and contralateral suppression) were normal in the B(-/-) mice throughout this period. Alcian blue staining of SGs demonstrated that these inclusion bodies corresponded to sulfatide accumulation. In contrast, changes in the vestibular system were much milder, but caused severe physiologic deficits. These results demonstrate that loss of Sap B function leads to progressive sulfatide accumulation in satellite cells surrounding the SG neurons, leading to satellite cell degeneration and subsequent SG degeneration with a resultant loss of hearing. Relative sparing of the efferent auditory and vestibular neurons suggests that alternate glycosphingolipid metabolic pathways predominate in these other systems.
Collapse
|
15
|
Akil O, Rouse SL, Chan DK, Lustig LR. Surgical method for virally mediated gene delivery to the mouse inner ear through the round window membrane. J Vis Exp 2015:52187. [PMID: 25867531 PMCID: PMC4401361 DOI: 10.3791/52187] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Gene therapy, used to achieve functional recovery from sensorineural deafness, promises to grant better understanding of the underlying molecular and genetic mechanisms that contribute to hearing loss. Introduction of vectors into the inner ear must be done in a way that widely distributes the agent throughout the cochlea while minimizing injury to the existing structures. This manuscript describes a post-auricular surgical approach that can be used for mouse cochlear therapy using molecular, pharmacologic, and viral delivery to mice postnatal day 10 and older via the round window membrane (RWM). This surgical approach enables rapid and direct delivery into the scala tympani while minimizing blood loss and avoiding animal mortality. This technique involves negligible or no damage to essential structures of the inner and middle ear as well as neck muscles while wholly preserving hearing. To demonstrate the efficacy of this surgical technique, the vesicular glutamate transporter 3 knockout (VGLUT3 KO) mice will be used as an example of a mouse model of congenital deafness that recovers hearing after delivery of VGLUT3 to the inner ear using an adeno-associated virus (AAV-1).
Collapse
Affiliation(s)
- Omar Akil
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco;
| | - Stephanie L Rouse
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco
| | - Dylan K Chan
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco
| | - Lawrence R Lustig
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco
| |
Collapse
|
16
|
Disrupted bone remodeling leads to cochlear overgrowth and hearing loss in a mouse model of fibrous dysplasia. PLoS One 2014; 9:e94989. [PMID: 24788917 PMCID: PMC4006800 DOI: 10.1371/journal.pone.0094989] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/20/2014] [Indexed: 02/05/2023] Open
Abstract
Normal hearing requires exquisite cooperation between bony and sensorineural structures within the cochlea. For example, the inner ear secretes proteins such as osteoprotegrin (OPG) that can prevent cochlear bone remodeling. Accordingly, diseases that affect bone regulation can also result in hearing loss. Patients with fibrous dysplasia develop trabecular bone overgrowth resulting in hearing loss if the lesions affect the temporal bones. Unfortunately, the mechanisms responsible for this hearing loss, which could be sensorineural and/or conductive, remain unclear. In this study, we used a unique transgenic mouse model of increased Gs G-protein coupled receptor (GPCR) signaling induced by expression of an engineered receptor, Rs1, in osteoblastic cells. These ColI(2.3)+/Rs1+ mice showed dramatic bone lesions that histologically and radiologically resembled fibrous dysplasia. We found that ColI(2.3)+/Rs1+ mice showed progressive and severe conductive hearing loss. Ossicular chain impingement increased with the size and number of dysplastic lesions. While sensorineural structures were unaffected, ColI(2.3)+/Rs1+ cochleae had abnormally high osteoclast activity, together with elevated tartrate resistant acid phosphatase (TRAP) activity and receptor activator of nuclear factor kappa-B ligand (Rankl) mRNA expression. ColI(2.3)+/Rs1+ cochleae also showed decreased expression of Sclerostin (Sost), an antagonist of the Wnt signaling pathway that normally increases bone formation. The osteocyte canalicular networks of ColI(2.3)+/Rs1+ cochleae were disrupted and showed abnormal osteocyte morphology. The osteocytes in the ColI(2.3)+/Rs1+ cochleae showed increased expression of matrix metalloproteinase 13 (MMP-13) and TRAP, both of which can support osteocyte-mediated peri-lacunar remodeling. Thus, while the ossicular chain impingement is sufficient to account for the progressive hearing loss in fibrous dysplasia, the deregulation of bone remodeling extends to the cochlea as well. Our findings suggest that factors regulating bone remodeling, including peri-lacunar remodeling by osteocytes, may be useful targets for treating the bony overgrowths and hearing changes of fibrous dysplasia and other bony pathologies.
Collapse
|
17
|
Akil O, Seal RP, Burke K, Wang C, Alemi A, During M, Edwards RH, Lustig LR. Restoration of hearing in the VGLUT3 knockout mouse using virally mediated gene therapy. Neuron 2012; 75:283-93. [PMID: 22841313 PMCID: PMC3408581 DOI: 10.1016/j.neuron.2012.05.019] [Citation(s) in RCA: 269] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2012] [Indexed: 01/05/2023]
Abstract
Mice lacking the vesicular glutamate transporter-3 (VGLUT3) are congenitally deaf due to loss of glutamate release at the inner hair cell afferent synapse. Cochlear delivery of VGLUT3 using adeno-associated virus type 1 (AAV1) leads to transgene expression in only inner hair cells (IHCs), despite broader viral uptake. Within 2 weeks of AAV1-VGLUT3 delivery, auditory brainstem response (ABR) thresholds normalize, along with partial rescue of the startle response. Lastly, we demonstrate partial reversal of the morphologic changes seen within the afferent IHC ribbon synapse. These findings represent a successful restoration of hearing by gene replacement in mice, which is a significant advance toward gene therapy of human deafness.
Collapse
Affiliation(s)
- Omar Akil
- Department of Otolaryngology- Head & Neck Surgery, University of California San Francisco, San Francisco, CA, 94143-0449. Phone: 415-476-0728.
| | - Rebecca P. Seal
- Department of Neurology- University of Pittsburgh, Pittsburgh, PA 15213-3301. Phone: 412-624-5183.
| | - Kevin Burke
- Department of Otolaryngology- Head & Neck Surgery, University of California San Francisco, San Francisco, CA, 94143-0449. Phone: 415-476-0728.
| | - Chuansong Wang
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio. Phone: 614-247-4351
| | - Aurash Alemi
- Department of Otolaryngology- Head & Neck Surgery, University of California San Francisco, San Francisco, CA, 94143-0449. Phone: 415-476-0728.
| | - Matthew During
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio. Phone: 614-247-4351.
| | - Robert H. Edwards
- Department of Neurology, University of California San Francisco, San Francisco, CA, 94143-2140. Phone: 415-502-5687.
| | | |
Collapse
|
18
|
Jurkovičová-Tarabová B, Griesemer D, Pirone A, Sinnegger-Brauns MJ, Striessnig J, Friauf E. Repertoire of high voltage-activated Ca2+ channels in the lateral superior olive: functional analysis in wild-type, Cav1.3−/−, and Cav1.2DHP−/− mice. J Neurophysiol 2012; 108:365-79. [DOI: 10.1152/jn.00948.2011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Voltage-gated Ca2+ (Cav)1.3 α-subunits of high voltage-activated Ca2+ channels (HVACCs) are essential for Ca2+ influx and transmitter release in cochlear inner hair cells and therefore for signal transmission into the central auditory pathway. Their absence leads to deafness and to striking structural changes in the auditory brain stem, particularly in the lateral superior olive (LSO). Here, we analyzed the contribution of various types of HVACCs to the total Ca2+ current ( ICa) in developing mouse LSO neurons to address several questions: do LSO neurons express functional Cav1.3 channels? What other types of HVACCs are expressed? Are there developmental changes? Do LSO neurons of Cav1.3−/− mice show any compensatory responses, namely, upregulation of other HVACCs? Our electrophysiological and pharmacological results showed the presence of functional Cav1.3 and Cav1.2 channels at both postnatal days 4 and 12. Aside from these L-type channels, LSO neurons also expressed functional P/Q-type, N-type, and, most likely, R-type channels. The relative contribution of the four different subtypes to ICa appeared to be 45%, 29%, 22%, and 4% at postnatal day 12, respectively. The physiological results were flanked and extended by quantitative RT-PCR data. Altogether, LSO neurons displayed a broad repertoire of HVACC subtypes. Genetic ablation of Cav1.3 resulted in functional reorganization of some other HVACCs but did not restore normal ICa properties. Together, our results suggest that several types of HVACCs are of functional relevance for the developing LSO. Whether on-site loss of Cav1.3, i.e., in LSO neurons, contributes to the recently described malformation of the LSO needs to be determined by using tissue-specific Cav1.3−/− animals.
Collapse
Affiliation(s)
| | - Désirée Griesemer
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Antonella Pirone
- Institute of Physiology II and Department of Otolaryngology, Tübingen Hearing Research Centre, University of Tübingen, Tübingen, Germany; and
| | - Martina J. Sinnegger-Brauns
- Institute of Pharmacy, Pharmacology and Toxicology, Center of Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Jörg Striessnig
- Institute of Pharmacy, Pharmacology and Toxicology, Center of Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Eckhard Friauf
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| |
Collapse
|
19
|
Akil O, Lustig LR. Severe vestibular dysfunction and altered vestibular innervation in mice lacking prosaposin. Neurosci Res 2012; 72:296-305. [PMID: 22326583 DOI: 10.1016/j.neures.2012.01.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 01/06/2012] [Accepted: 01/25/2012] [Indexed: 11/25/2022]
Abstract
Prosaposin, a precursor of four glycoprotein activators (saposin A, B, C and D) for lysosomal hydrolases, has previously been shown to be important for normal adult cochlear innervation and the maintenance of normal hearing. In these studies, we now investigate prosaposin in normal vestibular epithelium and the functional impairment of balance caused by prosaposin ablation. In normal mice, prosaposin is localized to all 3 vestibular end-organs (ampullae, saccule, and utricle) and Scarpa's ganglion by RT-PCR, Western blot analysis and immunofluorescence. Ablation of prosaposin function caused severe vestibular dysfunction on a battery of behavioral tasks. Histologically, the KO mice demonstrated an exuberant cellular proliferation below the vestibular hair cells with disruption of the supporting cells. Electron microscopy further demonstrated inclusion bodies and cellular proliferation disturbing the normal neuroepithelial structure of the vestibular end-organs. Lastly, immunofluorescence (neurofilament 200 and synaptophysin) staining suggests that this cellular proliferation corresponds to afferent and efferent neuronal overgrowth. These data suggest that prosaposin plays a role not only in the maintenance of normal hearing but also an important role in the neuronal maturation processes of the vestibular sensory epithelium and the maintenance of normal vestibular system function.
Collapse
Affiliation(s)
- Omar Akil
- Department of Otolaryngology-Head & Neck Surgery, University of California San Francisco, San Francisco, CA 94143-0449, United States.
| | | |
Collapse
|
20
|
Park MH, Park HJ, Kim KJ, Woo HM, Kim HJ, Lee JY, Park HY, Koo SK. Genome-wide SNP-based linkage analysis for ADNSHL families identifies novel susceptibility loci with positive evidence for linkage. Genes Genet Syst 2011; 86:117-21. [PMID: 21670551 DOI: 10.1266/ggs.86.117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The linkage search for susceptibility loci using SNP markers in hereditary hearing loss has proven challenging due to genetic heterogeneity. We conducted a genome-wide linkage analysis using high-density SNP markers in two Korean families (families coded SD-J and SR-167) with autosomal dominant non-syndromic hearing loss (ADNSHL). Evidence was found of linkage at 8q24.13~q24.3 and 10p11.21~q22.2 (LOD 3.01) in the SD-J family. In the case of family SR-167, which had the most affected members, the parametric LOD score was low owing to the lack of power for linkage analysis. However, using non-parametric linkage analysis, it was possible to obtain significant evidence for linkage at 10q22.1~q23.31 (LOD 1.79; NPL 6.47, P<0.00001). There is an overlapping region with a significant LOD score between the SD-J and SR-167 families, which encompasses 4 cM at 10q22.1~22.2. Interestingly, the characteristics of hearing loss in both families were similar, and the haplotype within overlapping region was shared in the affected individuals of the two families. We performed direct sequencing of the candidate genes that are thought to be causing the condition, but no disease-causing mutations were identified.
Collapse
Affiliation(s)
- Mi-Hyun Park
- Division of Cardiovascular and Rare Diseases, Center for Biomedical Sciences, National Institute of Health, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Hirtz JJ, Boesen M, Braun N, Deitmer JW, Kramer F, Lohr C, Müller B, Nothwang HG, Striessnig J, Löhrke S, Friauf E. Cav1.3 calcium channels are required for normal development of the auditory brainstem. J Neurosci 2011; 31:8280-94. [PMID: 21632949 PMCID: PMC6622878 DOI: 10.1523/jneurosci.5098-10.2011] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 03/14/2011] [Accepted: 04/10/2011] [Indexed: 12/18/2022] Open
Abstract
Within the Ca(v)1 family of voltage-gated calcium channels, Ca(v)1.2 and Ca(v)1.3 channels are the predominant subtypes in the brain. Whereas specific functions for each subtype were described in the adult brain, their role in brain development is poorly understood. Here we assess the role of Ca(v)1.3 subunits in the activity-dependent development of the auditory brainstem. We used Ca(v)1.3-deficient (Ca(v)1.3(-/-)) mice because these mice lack cochlea-driven activity that deprives the auditory centers from peripheral input. We found a drastically reduced volume in all auditory brainstem centers (range 25-59%, total 35%), which was manifest before hearing onset. A reduction was not obvious outside the auditory system. The lateral superior olive (LSO) was strikingly malformed in Ca(v)1.3(-/-) mice and had fewer neurons (1/3 less). The remaining LSO neurons displayed normal dendritic trees and received functional glutamatergic input, yet they fired action potentials predominantly with a multiple pattern upon depolarization, in contrast to the single firing pattern prevalent in controls. The latter finding appears to be due to a reduction of dendrototoxin-sensitive potassium conductances, presumably mediated through the K(v)1.2 subtype. Fura2 imaging provided evidence for functional Ca(v)1.3 channels in the LSO of wild-type mice. Our results imply that Ca(v)1.3 channels are indispensable for the development of the central auditory system. We propose that the unique LSO phenotype in Ca(v)1.3(-/-) mice, which hitherto was not described in other hereditary deafness models, is caused by the synergistic contribution of two factors: on-site loss of Ca(v)1.3 channels in the neurons plus lack of peripheral input.
Collapse
Affiliation(s)
| | | | | | - Joachim W. Deitmer
- General Zoology, Department of Biology, University of Kaiserslautern, D-67653 Kaiserslautern, Germany
| | | | - Christian Lohr
- General Zoology, Department of Biology, University of Kaiserslautern, D-67653 Kaiserslautern, Germany
| | | | - Hans Gerd Nothwang
- Animal Physiology Group and
- Department of Neurogenetics, Institute for Biology and Environmental Sciences, University of Oldenburg, D-26129 Oldenburg, Germany, and
| | - Jörg Striessnig
- Institute of Pharmacy, Pharmacology and Toxicology, Center of Molecular Biosciences, University of Innsbruck, A-6020 Innsbruck, Austria
| | | | | |
Collapse
|
22
|
Chang JL, Brauer DS, Johnson J, Chen CG, Akil O, Balooch G, Humphrey MB, Chin EN, Porter AE, Butcher K, Ritchie RO, Schneider RA, Lalwani A, Derynck R, Marshall GW, Marshall SJ, Lustig L, Alliston T. Tissue-specific calibration of extracellular matrix material properties by transforming growth factor-β and Runx2 in bone is required for hearing. EMBO Rep 2010; 11:765-71. [PMID: 20847738 PMCID: PMC2948188 DOI: 10.1038/embor.2010.135] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Revised: 08/09/2010] [Accepted: 08/11/2010] [Indexed: 01/02/2023] Open
Abstract
Physical cues, such as extracellular matrix stiffness, direct cell differentiation and support tissue-specific function. Perturbation of these cues underlies diverse pathologies, including osteoarthritis, cardiovascular disease and cancer. However, the molecular mechanisms that establish tissue-specific material properties and link them to healthy tissue function are unknown. We show that Runx2, a key lineage-specific transcription factor, regulates the material properties of bone matrix through the same transforming growth factor-β (TGFβ)-responsive pathway that controls osteoblast differentiation. Deregulated TGFβ or Runx2 function compromises the distinctly hard cochlear bone matrix and causes hearing loss, as seen in human cleidocranial dysplasia. In Runx2+/⁻ mice, inhibition of TGFβ signalling rescues both the material properties of the defective matrix, and hearing. This study elucidates the unknown cause of hearing loss in cleidocranial dysplasia, and demonstrates that a molecular pathway controlling cell differentiation also defines material properties of extracellular matrix. Furthermore, our results suggest that the careful regulation of these properties is essential for healthy tissue function.
Collapse
Affiliation(s)
- Jolie L Chang
- Department of Otolaryngology, Head and Neck Surgery, University of California at San Francisco, San Francisco, California 94143, USA
| | - Delia S Brauer
- Department of Preventive and Restorative Dental Sciences, University of California at San Francisco, San Francisco, California 94143, USA
| | - Jacob Johnson
- Department of Otolaryngology, Head and Neck Surgery, University of California at San Francisco, San Francisco, California 94143, USA
| | - Carol G Chen
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California 94143, USA
| | - Omar Akil
- Department of Otolaryngology, Head and Neck Surgery, University of California at San Francisco, San Francisco, California 94143, USA
| | - Guive Balooch
- Department of Materials Science and Engineering, University of California, Berkeley, California 94720, USA
- Materials Science Division, Lawrence Berkeley National Laboratories, Berkeley, California 94720, USA
| | - Mary Beth Humphrey
- Department of Medicine and Microbiology/Immunology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73190, USA
| | - Emily N Chin
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California 94143, USA
| | - Alexandra E Porter
- Department of Materials, Imperial College, Exhibition Road, London SW7 2AZ, UK
| | - Kristin Butcher
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California 94143, USA
| | - Robert O Ritchie
- Department of Materials Science and Engineering, University of California, Berkeley, California 94720, USA
- Materials Science Division, Lawrence Berkeley National Laboratories, Berkeley, California 94720, USA
| | - Richard A Schneider
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California 94143, USA
| | - Anil Lalwani
- Department of Otolaryngology, New York University, New York, New York 10016, USA
| | - Rik Derynck
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California 94143, USA
- Department of Cell and Tissue Biology, University of California at San Francisco, San Francisco, California 94143, USA
| | - Grayson W Marshall
- Department of Preventive and Restorative Dental Sciences, University of California at San Francisco, San Francisco, California 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California 94143, USA
| | - Sally J Marshall
- Department of Preventive and Restorative Dental Sciences, University of California at San Francisco, San Francisco, California 94143, USA
| | - Lawrence Lustig
- Department of Otolaryngology, Head and Neck Surgery, University of California at San Francisco, San Francisco, California 94143, USA
| | - Tamara Alliston
- Department of Otolaryngology, Head and Neck Surgery, University of California at San Francisco, San Francisco, California 94143, USA
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California 94143, USA
- Department of Bioengineering and Therapeutic Sciences, University of California at San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
23
|
More SS, Akil O, Ianculescu AG, Geier EG, Lustig LR, Giacomini KM. Role of the copper transporter, CTR1, in platinum-induced ototoxicity. J Neurosci 2010; 30:9500-9. [PMID: 20631178 PMCID: PMC2949060 DOI: 10.1523/jneurosci.1544-10.2010] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 05/17/2010] [Accepted: 05/24/2010] [Indexed: 12/31/2022] Open
Abstract
The goal of this study was to determine the role of an influx copper transporter, CTR1, in the ototoxicity induced by cisplatin, a potent anticancer platinum analog used in the treatment of a variety of solid tumors. As determined through reverse transcriptase-PCR (RT-PCR), quantitative RT-PCR, Western blot, and immunohistochemistry, mouse CTR1 (Ctr1) was found to be abundantly expressed and highly localized at the primary sites of cisplatin toxicity in the inner ear, mainly outer hair cells (OHCs), inner hair cells, stria vascularis, spiral ganglia, and surrounding nerves in the mouse cochlea. A CTR1 substrate, copper sulfate, decreased the uptake and cytotoxicity of cisplatin in HEI-OC1, a cell line that expresses many molecular markers reminiscent of OHCs. Small interfering RNA-mediated knockdown of Ctr1 in this cell line caused a corresponding decrease in cisplatin uptake. In mice, intratympanic administration of copper sulfate 30 min before intraperitoneal administration of cisplatin was found to prevent hearing loss at click stimulus and 8, 16, and 32 kHz frequencies. To date, the utility of cisplatin remains severely limited because of its ototoxic effects. The studies described in this report suggest that cisplatin-induced ototoxicity and cochlear uptake can be modulated by administration of a CTR1 inhibitor, copper sulfate. The possibility of local administration of CTR1 inhibitors during cisplatin therapy as a means of otoprotection is thereby raised.
Collapse
Affiliation(s)
- Swati S. More
- Departments of Bioengineering and Therapeutic Sciences and
| | - Omar Akil
- Otolaryngology–Head and Neck Surgery, University of California, San Francisco, San Francisco, California 94143-0449
| | | | - Ethan G. Geier
- Departments of Bioengineering and Therapeutic Sciences and
| | - Lawrence R. Lustig
- Otolaryngology–Head and Neck Surgery, University of California, San Francisco, San Francisco, California 94143-0449
| | | |
Collapse
|
24
|
Akil O, Weber CM, Park SN, Ninkina N, Buchman V, Lustig LR. Localization of synucleins in the mammalian cochlea. J Assoc Res Otolaryngol 2008; 9:452-63. [PMID: 18665422 PMCID: PMC2580813 DOI: 10.1007/s10162-008-0134-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2008] [Accepted: 07/11/2008] [Indexed: 10/21/2022] Open
Abstract
Synucleins are widely expressed synaptic proteins within the central nervous system that have been implicated in such neurodegenerative disorders as Parkinson's disease. In this study, an initial characterization of all three synucleins, alpha-, beta-, and gamma-synuclein, within the cochlea was undertaken. Reverse transcriptase-polymerase chain reaction (PCR) demonstrated all three synuclein mRNA species within microdissected cochlear tissue. Quantitative PCR suggests that beta-synuclein is the most abundantly expressed form, followed by gamma- and then alpha-synuclein. Western blot analysis similarly demonstrates all three synuclein proteins within microdissected cochlear tissue. Immunofluorescence localizes the three synucleins predominantly to the efferent neuronal system at the efferent outer hair cell synapse, with some additional localization within the efferent tunnel-crossing fibers (alpha- and gamma-synuclein), spiral ganglion (beta-synuclein), inner spiral bundle (gamma-synuclein), and stria vascularis (alpha- > beta-synuclein). Developmentally, gamma-synuclein can be seen in the region of the outer hair cells by E19, while alpha- and beta-synuclein do not clearly appear there until approximately P10. Additional studies in a null-mutant gamma-synuclein mouse show no histological changes in the organ of Corti with normal hair cell and spiral ganglion cell counts, and normal ABR and DPOAE thresholds in wild-type vs mutant littermates. Together, these results localize synucleins to the efferent cholinergic neuronal auditory system, pointing to a role in normal auditory function, and raising the potential implications for their role in auditory neurodegenerative disorders. However, gamma-synuclein alone is not required for the development and maintenance of normal hearing through P21. Whether overlapping roles of the other synucleins help compensate for the loss of gamma-synuclein remains to be determined.
Collapse
MESH Headings
- Animals
- Auditory Pathways/physiology
- Blotting, Western
- Cochlea/cytology
- Cochlea/embryology
- Cochlea/growth & development
- Evoked Potentials, Auditory, Brain Stem/physiology
- Fluorescent Antibody Technique
- Gene Expression Regulation, Developmental
- Hair Cells, Auditory, Inner/physiology
- Hair Cells, Auditory, Outer/physiology
- Mammals
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Otoacoustic Emissions, Spontaneous/physiology
- Phenotype
- Rats
- Rats, Sprague-Dawley
- Receptors, Nicotinic/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Synucleins/genetics
- Synucleins/metabolism
- alpha-Synuclein/genetics
- alpha-Synuclein/metabolism
- beta-Synuclein/genetics
- beta-Synuclein/metabolism
- gamma-Synuclein/genetics
- gamma-Synuclein/metabolism
Collapse
Affiliation(s)
- O. Akil
- Department of Otolaryngology—Head and Neck Surgery, University of California San Francisco, UC Hall, Room U401, 533 Parnassus Avenue, San Francisco, CA 94143-0449 USA
| | - C. M. Weber
- Department of Otolaryngology—Head and Neck Surgery, University of California San Francisco, UC Hall, Room U401, 533 Parnassus Avenue, San Francisco, CA 94143-0449 USA
| | - S. N. Park
- Department of Otolaryngology—Head and Neck Surgery, University of California San Francisco, UC Hall, Room U401, 533 Parnassus Avenue, San Francisco, CA 94143-0449 USA
- Department of Otolaryngology—Head and Neck Surgery, The Catholic University of Korea, College of Medicine, Seoul, South Korea
| | - N. Ninkina
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3US UK
| | - V. Buchman
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3US UK
| | - L. R. Lustig
- Department of Otolaryngology—Head and Neck Surgery, University of California San Francisco, UC Hall, Room U401, 533 Parnassus Avenue, San Francisco, CA 94143-0449 USA
| |
Collapse
|
25
|
Seal RP, Akil O, Yi E, Weber CM, Grant L, Yoo J, Clause A, Kandler K, Noebels JL, Glowatzki E, Lustig LR, Edwards RH. Sensorineural deafness and seizures in mice lacking vesicular glutamate transporter 3. Neuron 2008; 57:263-75. [PMID: 18215623 PMCID: PMC2293283 DOI: 10.1016/j.neuron.2007.11.032] [Citation(s) in RCA: 298] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2007] [Revised: 10/26/2007] [Accepted: 11/19/2007] [Indexed: 12/22/2022]
Abstract
The expression of unconventional vesicular glutamate transporter VGLUT3 by neurons known to release a different classical transmitter has suggested novel roles for signaling by glutamate, but this distribution has raised questions about whether the protein actually contributes to glutamate release. We now report that mice lacking VGLUT3 are profoundly deaf due to the absence of glutamate release from hair cells at the first synapse in the auditory pathway. The early degeneration of some cochlear ganglion neurons in knockout mice also indicates an important developmental role for the glutamate released by hair cells before the onset of hearing. In addition, the mice exhibit primary, generalized epilepsy that is accompanied by remarkably little change in ongoing motor behavior. The glutamate release conferred by expression of VGLUT3 thus has an essential role in both function and development of the auditory pathway, as well as in the control of cortical excitability.
Collapse
MESH Headings
- Acoustic Stimulation/methods
- Amino Acid Transport Systems, Acidic/deficiency
- Animals
- Animals, Newborn
- Calcium/metabolism
- Disease Models, Animal
- Electric Stimulation/methods
- Electroencephalography/methods
- Excitatory Amino Acid Antagonists/pharmacology
- Glutamic Acid/metabolism
- Hair Cells, Auditory/metabolism
- Hearing Loss, Sensorineural/etiology
- Hearing Loss, Sensorineural/genetics
- Hearing Loss, Sensorineural/pathology
- Membrane Potentials/drug effects
- Membrane Potentials/physiology
- Membrane Potentials/radiation effects
- Membrane Proteins/metabolism
- Mice
- Mice, Knockout
- Microscopy, Electron, Transmission/methods
- Neurons/pathology
- Neurons/ultrastructure
- Quinoxalines/pharmacology
- Reflex, Startle/physiology
- Seizures/etiology
- Seizures/genetics
- Spiral Ganglion/pathology
Collapse
Affiliation(s)
- Rebecca P. Seal
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, CA 94143
| | - Omar Akil
- Department of Otolaryngology, University of California, San Francisco School of Medicine, CA 94143
| | - Eunyoung Yi
- Department of Otolaryngology, Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Christopher M. Weber
- Department of Otolaryngology, University of California, San Francisco School of Medicine, CA 94143
| | - Lisa Grant
- Department of Otolaryngology, Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Jong Yoo
- Department of Neurology, Baylor College of Medicine, Houston, TX. 77030
| | - Amanda Clause
- Department of Otolaryngology and Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261
| | - Karl Kandler
- Department of Otolaryngology and Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261
| | - Jeffrey L Noebels
- Department of Neurology, Baylor College of Medicine, Houston, TX. 77030
| | - Elisabeth Glowatzki
- Department of Otolaryngology, Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Lawrence R. Lustig
- Department of Otolaryngology, University of California, San Francisco School of Medicine, CA 94143
| | - Robert H. Edwards
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, CA 94143
| |
Collapse
|