1
|
Attanasio C, Palladino A, Giaquinto D, Scavizzi F, Raspa M, Peres C, Anastasio C, Scocco P, Lucini C, de Girolamo P, D'Angelo L, De Felice E. Morphological phenotyping of the aging cochlea in inbred C57BL/6N and outbred CD1 mouse strains. Aging Cell 2024:e14362. [PMID: 39482905 DOI: 10.1111/acel.14362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 11/03/2024] Open
Abstract
Morphological mouse phenotyping plays a pivotal role in the translational setting and even more in the area of auditory research, where mouse is a central model organism due to the evolutionary genetic relationship and morpho-functional analogies with the human auditory system. However, some results obtained in murine models cannot be translated to humans due to the inadequate description of experimental conditions underlying poor reproducibility. We approach the characterization of the aging process of the mouse cochlea in animals up to 18 months of age belonging to two of the most used outbred (CD1) and inbred (C57BL/6N) strains. Striving to reduce any environmental variable we performed our study compliantly to the ARRIVE guidelines. We integrated instrumental data (auditory brainstem response test), with morphological analyses to correlate functional discrepancies to morphological changes and track the differences in the evolution of sensorineural hearing loss in the two strains. We featured the localization of Gipc3, Myosin VIIa, and TMC1 in hair cells of the Corti organ as well as NF 200 and the density of type I neuron in the spiral ganglion. We outlined age-related hearing loss (ARHL) in both strains, and a clear drop in the selected marker localization. However, in CD1 we detected a different trend allowing the identification of potential strain-specific mechanisms, namely an increase in myosin VIIa in 6 months aging mice in comparison to 2 months old animals. Our findings represent an asset to investigate the strain-dependent physiological trigger of ARHL providing new insights in the translational area.
Collapse
Affiliation(s)
- Chiara Attanasio
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Antonio Palladino
- Department of Agricultural Sciences, University of Naples Federico II, Naples, Italy
| | - Daniela Giaquinto
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Ferdinando Scavizzi
- National Research Council, CNR-Institute of Cellular Biology and Neurobiology, Monterotondo, Italy
- National Research Council, CNR-Institute of Biochemistry and Cell Biology - International Campus EMMA-INFRAFRONTIER-IMPC, Monterotondo, Italy
| | - Marcello Raspa
- National Research Council, CNR-Institute of Cellular Biology and Neurobiology, Monterotondo, Italy
- National Research Council, CNR-Institute of Biochemistry and Cell Biology - International Campus EMMA-INFRAFRONTIER-IMPC, Monterotondo, Italy
| | - Chiara Peres
- National Research Council, CNR-Institute of Cellular Biology and Neurobiology, Monterotondo, Italy
| | - Camilla Anastasio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Paola Scocco
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Carla Lucini
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Paolo de Girolamo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Livia D'Angelo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Elena De Felice
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| |
Collapse
|
2
|
Sanzhaeva U, Boyd-Pratt H, Bender PTR, Saravanan T, Rhodes SB, Guan T, Billington N, Boye SE, Cunningham CL, Anderson CT, Ramamurthy V. TUBB4B is essential for the cytoskeletal architecture of cochlear supporting cells and motile cilia development. Commun Biol 2024; 7:1146. [PMID: 39277687 PMCID: PMC11401917 DOI: 10.1038/s42003-024-06867-2] [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: 04/15/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024] Open
Abstract
Microtubules are essential for various cellular processes. The functional diversity of microtubules is attributed to the incorporation of various α- and β-tubulin isotypes encoded by different genes. In this work, we investigated the functional role of β4B-tubulin isotype (TUBB4B) in hearing and vision as mutations in TUBB4B are associated with sensorineural disease. Using a Tubb4b knockout mouse model, our findings demonstrate that TUBB4B is essential for hearing. Mice lacking TUBB4B are profoundly deaf due to defects in the inner and middle ear. Specifically, in the inner ear, the absence of TUBB4B lead to disorganized and reduced densities of microtubules in pillar cells, suggesting a critical role for TUBB4B in providing mechanical support for auditory transmission. In the middle ear, Tubb4b-/- mice exhibit motile cilia defects in epithelial cells, leading to the development of otitis media. However, Tubb4b deletion does not affect photoreceptor function or cause retinal degeneration. Intriguingly, β6-tubulin levels increase in retinas lacking β4B-tubulin isotype, suggesting a functional compensation mechanism. Our findings illustrate the essential roles of TUBB4B in hearing but not in vision in mice, highlighting the distinct functions of tubulin isotypes in different sensory systems.
Collapse
Affiliation(s)
- Urikhan Sanzhaeva
- Department of Biochemistry and Molecular Medicine, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Helen Boyd-Pratt
- Clinical Translational Sciences Institute, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Philip T R Bender
- Rockefeller Neuroscience Institute and Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Thamaraiselvi Saravanan
- Department of Biochemistry and Molecular Medicine, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Scott B Rhodes
- Department of Biochemistry and Molecular Medicine, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Tongju Guan
- Department of Biochemistry and Molecular Medicine, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Neil Billington
- Department of Biochemistry and Molecular Medicine, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Shannon E Boye
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Christopher L Cunningham
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charles T Anderson
- Rockefeller Neuroscience Institute and Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Visvanathan Ramamurthy
- Department of Biochemistry and Molecular Medicine, West Virginia University School of Medicine, Morgantown, WV, USA.
- Department of Ophthalmology and Visual Sciences, West Virginia University School of Medicine, Morgantown, WV, USA.
| |
Collapse
|
3
|
Lee JH, Park S, Perez-Flores MC, Chen Y, Kang M, Choi J, Levine L, Gratton MA, Zhao J, Notterpek L, Yamoah EN. Demyelination and Na + Channel Redistribution Underlie Auditory and Vestibular Dysfunction in PMP22-Null Mice. eNeuro 2024; 11:ENEURO.0462-23.2023. [PMID: 38378628 PMCID: PMC11059428 DOI: 10.1523/eneuro.0462-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 02/22/2024] Open
Abstract
Altered expression of peripheral myelin protein 22 (PMP22) results in demyelinating peripheral neuropathy. PMP22 exhibits a highly restricted tissue distribution with marked expression in the myelinating Schwann cells of peripheral nerves. Auditory and vestibular Schwann cells and the afferent neurons also express PMP22, suggesting a unique role in hearing and balancing. Indeed, neuropathic patients diagnosed with PMP22-linked hereditary neuropathies often present with auditory and balance deficits, an understudied clinical complication. To investigate the mechanism by which abnormal expression of PMP22 may cause auditory and vestibular deficits, we studied gene-targeted PMP22-null mice. PMP22-null mice exhibit an unsteady gait, have difficulty maintaining balance, and live for only ∼3-5 weeks relative to unaffected littermates. Histological analysis of the inner ear revealed reduced auditory and vestibular afferent nerve myelination and profound Na+ channel redistribution without PMP22. Yet, Na+ current density was unaltered, in stark contrast to increased K+ current density. Atypical postsynaptic densities and a range of neuronal abnormalities in the organ of Corti were also identified. Analyses of auditory brainstem responses (ABRs) and vestibular sensory-evoked potential (VsEP) revealed that PMP22-null mice had auditory and vestibular hypofunction. These results demonstrate that PMP22 is required for hearing and balance, and the protein is indispensable for the formation and maintenance of myelin in the peripheral arm of the eighth nerve. Our findings indicate that myelin abnormalities and altered signal propagation in the peripheral arm of the auditory nerve are likely causes of auditory deficits in patients with PMP22-linked neuropathies.
Collapse
Affiliation(s)
- Jeong Han Lee
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
| | - Seojin Park
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
- Prestige BioPharma, Busan 67264, South Korea
| | - Maria C Perez-Flores
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
| | - Yingying Chen
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
| | - Mincheol Kang
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
- Prestige BioPharma, Busan 67264, South Korea
| | - Jinsil Choi
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
| | - Lauren Levine
- Program in Audiology and Communication Sciences, Washington University, St. Louis 63110, Missouri
| | | | - Jie Zhao
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
| | - Lucia Notterpek
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
| | - Ebenezer N Yamoah
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
| |
Collapse
|
4
|
Abou Assale T, Kuenzel T, Schink T, Shahraz A, Neumann H, Klaus C. 6'-sialyllactose ameliorates the ototoxic effects of the aminoglycoside antibiotic neomycin in susceptible mice. Front Immunol 2023; 14:1264060. [PMID: 38130726 PMCID: PMC10733791 DOI: 10.3389/fimmu.2023.1264060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
Sialic acids are terminal sugars of the cellular glycocalyx and are highly abundant in the nervous tissue. Sialylation is sensed by the innate immune system and acts as an inhibitory immune checkpoint. Aminoglycoside antibiotics such as neomycin have been shown to activate tissue macrophages and induce ototoxicity. In this study, we investigated the systemic subcutaneous application of the human milk oligosaccharide 6'-sialyllactose (6SL) as a potential therapy for neomycin-induced ototoxicity in postnatal mice. Repeated systemic treatment of mice with 6SL ameliorated neomycin-induced hearing loss and attenuated neomycin-triggered macrophage activation in the cochlear spiral ganglion. In addition, 6SL reversed the neomycin-mediated increase in gene transcription of the pro-inflammatory cytokine interleukin-1β (Il-1b) and the apoptotic/inflammatory kinase Pik3cd in the inner ear. Interestingly, neomycin application also increased the transcription of desialylating enzyme neuraminidase 3 (Neu3) in the inner ear. In vitro, we confirmed that treatment with 6SL had anti-inflammatory, anti-phagocytic, and neuroprotective effects on cultured lipopolysaccharide-challenged human THP1-macrophages. Thus, our data demonstrated that treatment with 6SL has anti-inflammatory and protective effects against neomycin-mediated macrophage activation and ototoxicity.
Collapse
Affiliation(s)
- Tawfik Abou Assale
- Neural Regeneration, Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital of Bonn, University of Bonn, Bonn, Germany
| | - Thomas Kuenzel
- Auditory Neurophysiology, Department of Chemosensation, Institute for Biology II, RWTH Aachen University, Aachen, Germany
| | - Tamara Schink
- Neural Regeneration, Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital of Bonn, University of Bonn, Bonn, Germany
| | - Anahita Shahraz
- Neural Regeneration, Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital of Bonn, University of Bonn, Bonn, Germany
| | - Harald Neumann
- Neural Regeneration, Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital of Bonn, University of Bonn, Bonn, Germany
| | - Christine Klaus
- Neural Regeneration, Institute of Reconstructive Neurobiology, Medical Faculty and University Hospital of Bonn, University of Bonn, Bonn, Germany
| |
Collapse
|
5
|
Chen Y, Lee JH, Li J, Park S, Flores MCP, Peguero B, Kersigo J, Kang M, Choi J, Levine L, Gratton MA, Fritzsch B, Yamoah EN. Genetic and pharmacologic alterations of claudin9 levels suffice to induce functional and mature inner hair cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.08.561387. [PMID: 37873357 PMCID: PMC10592694 DOI: 10.1101/2023.10.08.561387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Hearing loss is the most common form of sensory deficit. It occurs predominantly due to hair cell (HC) loss. Mammalian HCs are terminally differentiated by birth, making HC loss incurable. Here, we show the pharmacogenetic downregulation of Cldn9, a tight junction protein, generates robust supernumerary inner HCs (IHCs) in mice. The putative ectopic IHCs have functional and synaptic features akin to typical IHCs and were surprisingly and remarkably preserved for at least fifteen months >50% of the mouse's life cycle. In vivo, Cldn9 knockdown using shRNA on postnatal days (P) P1-7 yielded analogous functional putative ectopic IHCs that were equally durably conserved. The findings suggest that Cldn9 levels coordinate embryonic and postnatal HC differentiation, making it a viable target for altering IHC development pre- and post-terminal differentiation.
Collapse
Affiliation(s)
- Yingying Chen
- University of Nevada, Reno, School of Medicine, Department of Physiology and Cell Biology, Reno NV 89557
- Indiana University School of Medicine, Department of Pharmacology and Toxicology, Indianapolis, IN, 46202, USA
| | - Jeong Han Lee
- University of Nevada, Reno, School of Medicine, Department of Physiology and Cell Biology, Reno NV 89557
| | - Jin Li
- Department of Otolaryngology, University of Washington Seattle, WA, USA
| | - Seojin Park
- University of Nevada, Reno, School of Medicine, Department of Physiology and Cell Biology, Reno NV 89557
- Prestige Biopharma, 11-12F, 44, Myongjigukje7-ro, Gangseo-gu, Busan, South Korea 67264
| | - Maria C. Perez Flores
- University of Nevada, Reno, School of Medicine, Department of Physiology and Cell Biology, Reno NV 89557
| | - Braulio Peguero
- Otolaryngology-Head, Neck Surgery, St. Louis University, St. Louis, Missouri 63108
| | | | - Mincheol Kang
- University of Nevada, Reno, School of Medicine, Department of Physiology and Cell Biology, Reno NV 89557
- Prestige Biopharma, 11-12F, 44, Myongjigukje7-ro, Gangseo-gu, Busan, South Korea 67264
| | - Jinsil Choi
- University of Nevada, Reno, School of Medicine, Department of Physiology and Cell Biology, Reno NV 89557
| | | | | | | | - Ebenezer N. Yamoah
- University of Nevada, Reno, School of Medicine, Department of Physiology and Cell Biology, Reno NV 89557
| |
Collapse
|
6
|
Na D, Yang Y, Xie L, Piekna-Przybylska D, Bunn D, Shamambo M, White P. Neuroinflammation in a Mouse Model of Alzheimer's Disease versus Auditory Dysfunction: Machine Learning Interpretation and Analysis. RESEARCH SQUARE 2023:rs.3.rs-3370200. [PMID: 37841847 PMCID: PMC10571613 DOI: 10.21203/rs.3.rs-3370200/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Background Auditory dysfunction, including central auditory hyperactivity, hearing loss and hearing in noise deficits, has been reported in 5xFAD Alzheimer's disease (AD) mice, suggesting a causal relationship between amyloidosis and auditory dysfunction. Central auditory hyperactivity correlated in time with small amounts of plaque deposition in the inferior colliculus and medial geniculate body, which are the auditory midbrain and thalamus, respectively. Neuroinflammation has been associated with excitation to inhibition imbalance in the central nervous system, and therefore has been proposed as a link between central auditory hyperactivity and AD in our previous report. However, neuroinflammation in the auditory pathway has not been investigated in mouse amyloidosis models. Methods Machine learning was used to classify the previously obtained auditory brainstem responses (ABRs) from 5xFAD mice and their wild type (WT) littermates. Neuroinflammation was assessed in six auditory-related regions of the cortex, thalamus, and brainstem. Cochlear pathology was assessed in cryosection and whole mount. Behavioral changes were assessed with fear conditioning, open field testing and novel objection recognition. Results Reliable machine learning classification of 5xFAD and WT littermate ABRs were achieved for 6M and 12M, but not 3M. The top features for accurate classification at 6 months of age were characteristics of Waves IV and V. Microglial and astrocytic activation were pronounced in 5xFAD inferior colliculus and medial geniculate body at 6 months, two neural centers that are thought to contribute to these waves. Lower regions of the brainstem were unaffected, and cortical auditory centers also displayed inflammation beginning at 6 months. No losses were seen in numbers of spiral ganglion neurons (SGNs), auditory synapses, or efferent synapses in the cochlea. 5xFAD mice had reduced responses to tones in fear conditioning compared to WT littermates beginning at 6 months. Conclusions Serial use of ABR in early AD patients represents a promising approach for early and inexpensive detection of neuroinflammation in higher auditory brainstem processing centers. As changes in auditory processing are strongly linked to AD progression, central auditory hyperactivity may serve as a biomarker for AD progression and/or stratify AD patients into distinct populations.
Collapse
Affiliation(s)
| | | | - Li Xie
- University of Rochester Medical Center
| | | | | | | | | |
Collapse
|
7
|
Cortada M, Levano S, Hall MN, Bodmer D. mTORC2 regulates auditory hair cell structure and function. iScience 2023; 26:107687. [PMID: 37694145 PMCID: PMC10484995 DOI: 10.1016/j.isci.2023.107687] [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: 03/30/2023] [Revised: 07/14/2023] [Accepted: 08/17/2023] [Indexed: 09/12/2023] Open
Abstract
mTOR broadly controls cell growth, but little is known about the role of mTOR complex 2 (mTORC2) in the inner ear. To investigate the role of mTORC2 in sensory hair cells (HCs), we generated HC-specific Rictor knockout (HC-RicKO) mice. HC-RicKO mice exhibited early-onset, progressive, and profound hearing loss. Increased DPOAE thresholds indicated outer HC dysfunction. HCs are lost, but this occurs after hearing loss. Ultrastructural analysis revealed stunted and absent stereocilia in outer HCs. In inner HCs, the number of synapses was significantly decreased and the remaining synapses displayed a disrupted actin cytoskeleton and disorganized Ca2+ channels. Thus, the mTORC2 signaling pathway plays an important role in regulating auditory HC structure and function via regulation of the actin cytoskeleton. These results provide molecular insights on a central regulator of cochlear HCs and thus hearing.
Collapse
Affiliation(s)
- Maurizio Cortada
- Department of Biomedicine, University of Basel, CH-4031 Basel, Switzerland
| | - Soledad Levano
- Department of Biomedicine, University of Basel, CH-4031 Basel, Switzerland
| | | | - Daniel Bodmer
- Department of Biomedicine, University of Basel, CH-4031 Basel, Switzerland
- Clinic for Otorhinolaryngology, Head and Neck Surgery, University of Basel Hospital, CH-4031 Basel, Switzerland
| |
Collapse
|
8
|
Xie Z, Ma XH, Bai QF, Tang J, Sun JH, Jiang F, Guo W, Wang CM, Yang R, Wen YC, Wang FY, Chen YX, Zhang H, He DZ, Kelley MW, Yang S, Zhang WJ. ZBTB20 is essential for cochlear maturation and hearing in mice. Proc Natl Acad Sci U S A 2023; 120:e2220867120. [PMID: 37279265 PMCID: PMC10268240 DOI: 10.1073/pnas.2220867120] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 04/25/2023] [Indexed: 06/08/2023] Open
Abstract
The mammalian cochlear epithelium undergoes substantial remodeling and maturation before the onset of hearing. However, very little is known about the transcriptional network governing cochlear late-stage maturation and particularly the differentiation of its lateral nonsensory region. Here, we establish ZBTB20 as an essential transcription factor required for cochlear terminal differentiation and maturation and hearing. ZBTB20 is abundantly expressed in the developing and mature cochlear nonsensory epithelial cells, with transient expression in immature hair cells and spiral ganglion neurons. Otocyst-specific deletion of Zbtb20 causes profound deafness with reduced endolymph potential in mice. The subtypes of cochlear epithelial cells are normally generated, but their postnatal development is arrested in the absence of ZBTB20, as manifested by an immature appearance of the organ of Corti, malformation of tectorial membrane (TM), a flattened spiral prominence (SP), and a lack of identifiable Boettcher cells. Furthermore, these defects are related with a failure in the terminal differentiation of the nonsensory epithelium covering the outer border Claudius cells, outer sulcus root cells, and SP epithelial cells. Transcriptome analysis shows that ZBTB20 regulates genes encoding for TM proteins in the greater epithelial ridge, and those preferentially expressed in root cells and SP epithelium. Our results point to ZBTB20 as an essential regulator for postnatal cochlear maturation and particularly for the terminal differentiation of cochlear lateral nonsensory domain.
Collapse
Affiliation(s)
- Zhifang Xie
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Institute of Early Life Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai200092, China
- Department of Pathophysiology, Naval Medical University, Shanghai200433, China
| | - Xian-Hua Ma
- Department of Pathophysiology, Naval Medical University, Shanghai200433, China
| | - Qiu-Fang Bai
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin300134, China
| | - Jie Tang
- Department of Physiology, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Jian-He Sun
- Senior Department of Otolaryngology-Head and Neck Surgery, National Clinical Research Center for Otolaryngologic Diseases, the Sixth Medical Center of PLA General Hospital, Beijing100141, China
| | - Fei Jiang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Institute of Early Life Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai200092, China
| | - Wei Guo
- Senior Department of Otolaryngology-Head and Neck Surgery, National Clinical Research Center for Otolaryngologic Diseases, the Sixth Medical Center of PLA General Hospital, Beijing100141, China
| | - Chen-Ma Wang
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin300134, China
| | - Rui Yang
- Department of Pathophysiology, Naval Medical University, Shanghai200433, China
| | - Yin-Chuan Wen
- Department of Physiology, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Fang-Yuan Wang
- Senior Department of Otolaryngology-Head and Neck Surgery, National Clinical Research Center for Otolaryngologic Diseases, the Sixth Medical Center of PLA General Hospital, Beijing100141, China
| | - Yu-Xia Chen
- Department of Pathophysiology, Naval Medical University, Shanghai200433, China
| | - Hai Zhang
- Department of Pathophysiology, Naval Medical University, Shanghai200433, China
| | - David Z. He
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE68178
| | | | - Shiming Yang
- Senior Department of Otolaryngology-Head and Neck Surgery, National Clinical Research Center for Otolaryngologic Diseases, the Sixth Medical Center of PLA General Hospital, Beijing100141, China
| | - Weiping J. Zhang
- Department of Pathophysiology, Naval Medical University, Shanghai200433, China
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin300134, China
| |
Collapse
|
9
|
Strepay D, Olszewski R, Taukulis I, Johns JD, Gu S, Hoa M. Dissection of Adult Mouse Stria Vascularis for Single-Nucleus Sequencing or Immunostaining. J Vis Exp 2023:10.3791/65254. [PMID: 37154552 PMCID: PMC10443831 DOI: 10.3791/65254] [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: 05/10/2023] Open
Abstract
Endocochlear potential, which is generated by the stria vascularis, is essential to maintain an environment conducive to appropriate hair cell mechanotransduction and ultimately hearing. Pathologies of the stria vascularis can result in a decreased hearing. Dissection of the adult stria vascularis allows for focused single-nucleus capture and subsequent single-nucleus sequencing and immunostaining. These techniques are used to study stria vascularis pathophysiology at the single-cell level. Single-nucleus sequencing can be used in the setting of transcriptional analysis of the stria vascularis. Meanwhile, immunostaining continues to be useful in identifying specific populations of cells. Both methods require proper stria vascularis dissection as a prerequisite, which can prove to be technically challenging.
Collapse
Affiliation(s)
- Dillon Strepay
- Auditory Development and Restoration Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health
| | - Rafal Olszewski
- Auditory Development and Restoration Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health
| | - Ian Taukulis
- Auditory Development and Restoration Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health
| | - J Dixon Johns
- Auditory Development and Restoration Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health
| | - Shoujun Gu
- Auditory Development and Restoration Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health
| | - Michael Hoa
- Auditory Development and Restoration Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health;
| |
Collapse
|
10
|
De la Torre I MH, Mauricio Flores M J, Piazza V, Velazquez EDH, Hernandez VH. Concurrent optical inspection to boost characterization of plastic cortical bone under mechanical deformation. APPLIED OPTICS 2023; 62:1483-1491. [PMID: 36821308 DOI: 10.1364/ao.476551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/19/2023] [Indexed: 06/18/2023]
Abstract
The simultaneous surface and internal measurements from a chemically modified cortical bovine bone suffering a plastic range deformation are presented. Since the bone is an anisotropic structure, its mechanical response could be modified if its organic or inorganic phases change. The latter could result in high plastic deformations, where the interferometrical signal from an optical analysis is easily de-correlated. In this work, digital holography interferometry (DHI) and Fourier domain optical coherence tomography (FD-OCT) are used to analyze the plastic range deformation of the bone under compression. The simultaneous use of these two optical methods gives information even when one of them de-correlates. The surface results retrieved with DHI show the high anisotropy of the bone as a continuously increasing displacement field map. Meanwhile, the internal information obtained with FD-OCT records larger deformations at different depths. Due to the optical phase, it is possible to complement the measurements of these two methods during the plastic deformation.
Collapse
|
11
|
Zhang Y, Neng L, Sharma K, Hou Z, Johnson A, Song J, Dabdoub A, Shi X. Pericytes control vascular stability and auditory spiral ganglion neuron survival. eLife 2023; 12:e83486. [PMID: 36719173 PMCID: PMC9940910 DOI: 10.7554/elife.83486] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/26/2023] [Indexed: 02/01/2023] Open
Abstract
The inner ear has a rich population of pericytes, a multi-functional mural cell essential for sensory hair cell heath and normal hearing. However, the mechanics of how pericytes contribute to the homeostasis of the auditory vascular-neuronal complex in the spiral ganglion are not yet known. In this study, using an inducible and conditional pericyte depletion mouse (PDGFRB-CreERT2; ROSA26iDTR) model, we demonstrate, for the first time, that pericyte depletion causes loss of vascular volume and spiral ganglion neurons (SGNs) and adversely affects hearing sensitivity. Using an in vitro trans-well co-culture system, we show pericytes markedly promote neurite and vascular branch growth in neonatal SGN explants and adult SGNs. The pericyte-controlled neural growth is strongly mediated by pericyte-released exosomes containing vascular endothelial growth factor-A (VEGF-A). Treatment of neonatal SGN explants or adult SGNs with pericyte-derived exosomes significantly enhances angiogenesis, SGN survival, and neurite growth, all of which were inhibited by a selective blocker of VEGF receptor 2 (Flk1). Our study demonstrates that pericytes in the adult ear are critical for vascular stability and SGN health. Cross-talk between pericytes and SGNs via exosomes is essential for neuronal and vascular health and normal hearing.
Collapse
Affiliation(s)
- Yunpei Zhang
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science UniversityPortlandUnited States
| | - Lingling Neng
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science UniversityPortlandUnited States
| | - Kushal Sharma
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science UniversityPortlandUnited States
| | - Zhiqiang Hou
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science UniversityPortlandUnited States
| | - Anatasiya Johnson
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science UniversityPortlandUnited States
| | - Junha Song
- Life Sciences Division, Lawrence Berkeley National LaboratoryBerkeleyUnited States
| | - Alain Dabdoub
- Biological Sciences, Sunnybrook Research InstituteTorontoCanada
- Department of Otolaryngology-Head & Neck Surgery, University of TorontoTorontoCanada
- Department of Laboratory Medicine and Pathobiology, University of TorontoTorontoCanada
| | - Xiaorui Shi
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science UniversityPortlandUnited States
| |
Collapse
|
12
|
Generotti C, Cox BC, Singh J, Hamilton D, McKenzie E, O'Malley BW, Li D. Subclinical diagnosis of cisplatin-induced ototoxicity with biomarkers. Sci Rep 2022; 12:18032. [PMID: 36302835 PMCID: PMC9613680 DOI: 10.1038/s41598-022-23034-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/25/2022] [Indexed: 01/24/2023] Open
Abstract
A mouse model with cisplatin-induced ototoxicity was used in addition to human samples from the ITMAT Biobank at the University of Pennsylvania. Mouse auditory brainstem responses (ABR), inner ear histology, perilymph cisplatin sampling, and measurement of serum prestin via ELISA were performed. Human serum prestin level was measured via ELISA in patients with otological issues after cisplatin treatment and compared to matched controls. Serum prestin was significantly elevated before ABR threshold shifts in mice exposed to cisplatin compared to control mice. Prestin concentration also correlated with the severity of hearing threshold shifts in mice. After an extended rest post-cisplatin treatment, prestin returned to baseline levels in mice and humans. Prestin was significantly elevated in the serum before the onset of objective hearing loss and correlated with the severity of hearing damage indicating that prestin may function as an effective biomarker of cisplatin-induced ototoxicity. Human serum prestin levels responded similarly to mice > 3 weeks from ototoxic exposure with decreased levels of prestin in the serum.
Collapse
Affiliation(s)
- Charles Generotti
- Department of Otolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., BRB 1212, Philadelphia, PA, 19104, USA
| | - Brandon C Cox
- Department of Pharmacology, Southern Illinois University School of Medicine, 801 N. Rutledge St, Springfield, IL, 62702, USA
| | - Jarnail Singh
- Department of Pharmacology, Southern Illinois University School of Medicine, 801 N. Rutledge St, Springfield, IL, 62702, USA
| | - Deborah Hamilton
- Department of Pharmacology, Southern Illinois University School of Medicine, 801 N. Rutledge St, Springfield, IL, 62702, USA
| | - Erica McKenzie
- Department of Civil and Environmental Engineering, Temple University, Philadelphia, PA, 19122, USA
| | - Bert W O'Malley
- University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Daqing Li
- Department of Otolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., BRB 1212, Philadelphia, PA, 19104, USA.
| |
Collapse
|
13
|
Scheibinger M, Janesick A, Benkafadar N, Ellwanger DC, Jan TA, Heller S. Cell-type identity of the avian utricle. Cell Rep 2022; 40:111432. [PMID: 36170825 PMCID: PMC9588199 DOI: 10.1016/j.celrep.2022.111432] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/18/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022] Open
Abstract
The avian utricle, a vestibular organ of the inner ear, displays turnover of sensory hair cells throughout life. This is in sharp contrast to the mammalian utricle, which shows limited regenerative capacity. Here, we use single-cell RNA sequencing to identify distinct marker genes for the different sensory hair cell subtypes of the chicken utricle, which we validated in situ. We provide markers for spatially distinct supporting cell populations and identify two transitional cell populations of dedifferentiating supporting cells and developing hair cells. Trajectory reconstruction resulted in an inventory of gene expression dynamics of natural hair cell generation in the avian utricle. Scheibinger et al. provide a single-cell transcriptomic atlas of the chicken utricle, a vestibular organ. Hair cell and supporting cell subtypes are defined by marker genes, and trajectories of gene expression dynamics during hair cell turnover are shown. This resource provides a baseline to study inner ear damage and regeneration.
Collapse
Affiliation(s)
- Mirko Scheibinger
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Amanda Janesick
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nesrine Benkafadar
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Daniel C Ellwanger
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Taha A Jan
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stefan Heller
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
14
|
Effects of pyrroloquinoline quinone on noise-induced and age-related hearing loss in mice. Sci Rep 2022; 12:15911. [PMID: 36151123 PMCID: PMC9508078 DOI: 10.1038/s41598-022-19842-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 09/05/2022] [Indexed: 11/10/2022] Open
Abstract
We investigated whether the oxidoreductase cofactor pyrroloquinoline quinone (PQQ) prevents noise-induced and age-related hearing loss (NIHL and ARHL) in mice. To assess NIHL, 8 week-old mice with and without PQQ administration were exposed to noise for 4 h. PQQ was orally administered for one week before and after noise exposure and subcutaneously once before noise exposure. For ARHL evaluation, mice were given drinking water with or without PQQ starting at 2 months of age. In the NIHL model, PQQ-treated mice had auditory brainstem response (ABR) thresholds of significantly reduced elevation at 8 kHz, a significantly increased number of hair cells at the basal turn, and significantly better maintained synapses beneath the inner hair cells compared to controls. In the ARHL model, PQQ significantly attenuated the age-related increase in ABR thresholds at 8 and 32 kHz at 10 months of age compared to controls. In addition, the hair cells, spiral ganglion cells, ribbon synapses, stria vascularis and nerve fibers were all significantly better maintained in PQQ-treated animals compared to controls at 10 months of age. These physiological and histological results demonstrate that PQQ protects the auditory system from NIHL and ARHL in mice.
Collapse
|
15
|
Heuermann ML, Matos S, Hamilton D, Cox BC. Regenerated hair cells in the neonatal cochlea are innervated and the majority co-express markers of both inner and outer hair cells. Front Cell Neurosci 2022; 16:841864. [PMID: 36187289 PMCID: PMC9524252 DOI: 10.3389/fncel.2022.841864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 08/29/2022] [Indexed: 11/30/2022] Open
Abstract
After a damaging insult, hair cells can spontaneously regenerate from cochlear supporting cells within the first week of life. While the regenerated cells express several markers of immature hair cells and have stereocilia bundles, their capacity to differentiate into inner or outer hair cells, and ability to form new synaptic connections has not been well-described. In addition, while multiple supporting cell subtypes have been implicated as the source of the regenerated hair cells, it is unclear if certain subtypes have a greater propensity to form one hair cell type over another. To investigate this, we used two CreER mouse models to fate-map either the supporting cells located near the inner hair cells (inner phalangeal and border cells) or outer hair cells (Deiters’, inner pillar, and outer pillar cells) along with immunostaining for markers that specify the two hair cell types. We found that supporting cells fate-mapped by both CreER lines responded early to hair cell damage by expressing Atoh1, and are capable of producing regenerated hair cells that express terminal differentiation markers of both inner and outer hair cells. The majority of regenerated hair cells were innervated by neuronal fibers and contained synapses. Unexpectedly, we also found that the majority of the laterally positioned regenerated hair cells aberrantly expressed both the outer hair cell gene, oncomodulin, and the inner hair cell gene, vesicular glutamate transporter 3 (VGlut3). While this work demonstrates that regenerated cells can express markers of both inner and outer hair cells after damage, VGlut3 expression appears to lack the tight control present during embryogenesis, which leads to its inappropriate expression in regenerated cells.
Collapse
Affiliation(s)
- Mitchell L. Heuermann
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Sophia Matos
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Deborah Hamilton
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Brandon C. Cox
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
- *Correspondence: Brandon C. Cox,
| |
Collapse
|
16
|
Ontogeny of cellular organization and LGR5 expression in porcine cochlea revealed using tissue clearing and 3D imaging. iScience 2022; 25:104695. [PMID: 35865132 PMCID: PMC9294204 DOI: 10.1016/j.isci.2022.104695] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/20/2022] [Accepted: 06/27/2022] [Indexed: 11/23/2022] Open
Abstract
Over 11% of the world's population experience hearing loss. Although there are promising studies to restore hearing in rodent models, the size, ontogeny, genetics, and frequency range of hearing of most rodents' cochlea do not match that of humans. The porcine cochlea can bridge this gap as it shares many anatomical, physiological, and genetic similarities with its human counterpart. Here, we provide a detailed methodology to process and image the porcine cochlea in 3D using tissue clearing and light-sheet microscopy. The resulting 3D images can be employed to compare cochleae across different ages and conditions, investigate the ontogeny of cochlear cytoarchitecture, and produce quantitative expression maps of LGR5, a marker of cochlear progenitors in mice. These data reveal that hair cell organization, inner ear morphology, cellular cartography in the organ of Corti, and spatiotemporal expression of LGR5 are dynamic over developmental stages in a pattern not previously documented.
Collapse
|
17
|
Li J, Liu C, Zhao B. Collapsin Response Mediator Protein 1 (CRMP1) Is Required for High-Frequency Hearing. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:805-812. [PMID: 35181334 PMCID: PMC9088201 DOI: 10.1016/j.ajpath.2022.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/04/2022] [Accepted: 01/11/2022] [Indexed: 05/03/2023]
Abstract
Collapsin response mediator protein 1 (CRMP1), also known as dihydropyrimidinase-related protein 1, participates in cytoskeleton remodeling during axonal guidance and neuronal migration. In cochlear hair cells, the assembly and maintenance of the cytoskeleton is of great interest because it is crucial for the morphogenesis and maintenance of hair cells. Previous RNA sequencing analysis found that Crmp1 is highly expressed in cochlear hair cells. However, the expression profile and functions of CRMP1 in the inner ear remain unknown. In this study, the expression and localization of CRMP1 in hair cells was investigated using immunostaining, and was shown to be highly expressed in both outer and inner hair cells. Next, the stereocilia morphology of Crmp1-deficient mice was characterized. Abolishing CRMP1 did not affect the morphogenesis of hair cells. Interestingly, scanning electron microscopy detected hair cell loss at the basal cochlear region, an area responsible for high-frequency auditory perception, in Crmp1-deficient mice. Correspondingly, an auditory brainstem response test showed that mice lacking CRMP1 had progressive hearing loss at high frequencies. In summary, these data suggest that CRMP1 is required for high-frequency auditory perception.
Collapse
Affiliation(s)
- Jinan Li
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Chang Liu
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Bo Zhao
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
18
|
Pravastatin Administration Alleviates Kanamycin-Induced Cochlear Injury and Hearing Loss. Int J Mol Sci 2022; 23:ijms23094524. [PMID: 35562915 PMCID: PMC9105065 DOI: 10.3390/ijms23094524] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/13/2022] [Accepted: 04/18/2022] [Indexed: 11/23/2022] Open
Abstract
The effect of statins on aminoglycoside-induced ototoxicity is controversial. This study aimed to explore the role of pravastatin (PV) in kanamycin-induced hearing loss in rats. Adult rats were intraperitoneally treated with 20 mg/kg/day of kanamycin (KM) for 10 days. In the PV- and PV + KM-treated rats, 25 mg/kg/day of PV was intraperitoneally administered for 5 days. The auditory brainstem response (ABR) thresholds were measured before and after drug treatment using a smartEP system at 4, 8, 16, and 32 kHz. Cochlear changes in poly ADP-ribose (PAR) polymerase (PARP), PAR, and caspase 3 were estimated using Western blotting. PV administration did not increase the ABR thresholds. The KM-treated rats showed elevated ABR thresholds at 4, 8, 16, and 32 kHz. The PV + KM-treated rats demonstrated lower ABR thresholds than the KM-treated rats at 4, 8, and 16 kHz. The cochlear outer hair cells and spiral ganglion cells were relatively preserved in the PV + KM-treated rats when compared with that in the KM-treated rats. The cochlear expression levels of PARP, PAR, and caspase 3 were higher in the KM-treated rats. The PV + KM-treated rats showed lower levels of PARP, PAR, and caspase 3 than the KM-treated rats. PV protected cochleae from KM-induced hearing loss in rats. The regulation of autophagy and apoptosis mediated the otoprotective effects of PV.
Collapse
|
19
|
Shankhwar S, Schwarz K, Katiyar R, Jung M, Maxeiner S, Südhof TC, Schmitz F. RIBEYE B-Domain Is Essential for RIBEYE A-Domain Stability and Assembly of Synaptic Ribbons. Front Mol Neurosci 2022; 15:838311. [PMID: 35153673 PMCID: PMC8831697 DOI: 10.3389/fnmol.2022.838311] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/03/2022] [Indexed: 01/17/2023] Open
Abstract
Synaptic ribbons are presynaptic specializations that define eponymous ribbon synapses. Synaptic ribbons are largely composed of RIBEYE, a protein containing an N-terminal A-domain and a carboxyterminal B-domain that is identical with CtBP2, a NAD(H)-binding transcriptional co-repressor. Previously we showed that synaptic ribbons are completely absent in RIBEYE knockout mice in which the RIBEYE A-domain-encoding exon had been deleted, but CtBP2 is still made, demonstrating that the A-domain is required for synaptic ribbon assembly. In the present study, we asked whether the RIBEYE B-domain also has an essential role in the assembly of synaptic ribbons. For this purpose, we made use of RIBEYE knockin mice in which the RIBEYE B-domain was replaced by a fluorescent protein domain, whereas the RIBEYE A-domain was retained unchanged. We found that replacing the RIBEYE B-domain with a fluorescent protein module destabilizes the resulting hybrid protein and causes a complete loss of synaptic ribbons. Our results thus demonstrate an essential role of the RIBEYE B-domain in enabling RIBEYE assembly into synaptic ribbons, reinforcing the notion that RIBEYE is the central organizer of synaptic ribbons.
Collapse
Affiliation(s)
- Soni Shankhwar
- Institute of Anatomy and Cell Biology, Saarland University, Medical School, Homburg, Germany
- *Correspondence: Soni Shankhwar Frank Schmitz
| | - Karin Schwarz
- Institute of Anatomy and Cell Biology, Saarland University, Medical School, Homburg, Germany
| | - Rashmi Katiyar
- Institute of Anatomy and Cell Biology, Saarland University, Medical School, Homburg, Germany
| | - Martin Jung
- Institute of Medical Biochemistry and Molecular Biology, Saarland University, Medical School, Homburg, Germany
| | - Stephan Maxeiner
- Institute of Anatomy and Cell Biology, Saarland University, Medical School, Homburg, Germany
| | - Thomas C. Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Frank Schmitz
- Institute of Anatomy and Cell Biology, Saarland University, Medical School, Homburg, Germany
- *Correspondence: Soni Shankhwar Frank Schmitz
| |
Collapse
|
20
|
Lee CH, Lee SM, Kim SY. Telmisartan Attenuates Kanamycin-Induced Ototoxicity in Rats. Int J Mol Sci 2021; 22:ijms222312716. [PMID: 34884516 PMCID: PMC8657567 DOI: 10.3390/ijms222312716] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/23/2021] [Accepted: 11/23/2021] [Indexed: 12/31/2022] Open
Abstract
Telmisartan (TM) has been proposed to relieve inflammatory responses by modulating peroxisome proliferator activator receptor-γ (PPARγ) signaling. This study aimed to investigate the protective effects of TM on kanamycin(KM)-induced ototoxicity in rats. Forty-eight, 8-week-old female Sprague Dawley rats were divided into four groups: (1) control group, (2) TM group, (3) KM group, and (4) TM + KM group. Auditory brainstem response was measured. The histology of the cochlea was examined. The protein expression levels of angiotensin-converting enzyme 2 (ACE2), HO1, and PPARγ were measured by Western blotting. The auditory threshold shifts at 4, 8, 16, and 32 kHz were lower in the TM + KM group than in the KM group (all p < 0.05). The loss of cochlear outer hair cells and spiral ganglial cells was lower in the TM + KM group than in the KM group. The protein expression levels of ACE2, PPARγ, and HO1 were higher in the KM group than in the control group (all p < 0.05). The TM + KM group showed lower expression levels of PPARγ and HO1 than the KM group.TM protected the cochlea from KM-induced injuries in rats. TM preserved hearing levels and attenuated the increase in PPARγ and HO1 expression levels in KM-exposed rat cochleae.
Collapse
|
21
|
Badash I, Quiñones PM, Oghalai KJ, Wang J, Lui CG, Macias-Escriva F, Applegate BE, Oghalai JS. Endolymphatic Hydrops is a Marker of Synaptopathy Following Traumatic Noise Exposure. Front Cell Dev Biol 2021; 9:747870. [PMID: 34805158 PMCID: PMC8602199 DOI: 10.3389/fcell.2021.747870] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/20/2021] [Indexed: 12/28/2022] Open
Abstract
After acoustic trauma, there can be loss of synaptic connections between inner hair cells and auditory neurons in the cochlea, which may lead to hearing abnormalities including speech-in-noise difficulties, tinnitus, and hyperacusis. We have previously studied mice with blast-induced cochlear synaptopathy and found that they also developed a build-up of endolymph, termed endolymphatic hydrops. In this study, we used optical coherence tomography to measure endolymph volume in live CBA/CaJ mice exposed to various noise intensities. We quantified the number of synaptic ribbons and postsynaptic densities under the inner hair cells 1 week after noise exposure to determine if they correlated with acute changes in endolymph volume measured in the hours after the noise exposure. After 2 h of noise at an intensity of 95 dB SPL or below, both endolymph volume and synaptic counts remained normal. After exposure to 2 h of 100 dB SPL noise, mice developed endolymphatic hydrops and had reduced synaptic counts in the basal and middle regions of the cochlea. Furthermore, round-window application of hypertonic saline reduced the degree of endolymphatic hydrops that developed after 100 dB SPL noise exposure and partially prevented the reduction in synaptic counts in the cochlear base. Taken together, these results indicate that endolymphatic hydrops correlates with noise-induced cochlear synaptopathy, suggesting that these two pathologic findings have a common mechanistic basis.
Collapse
Affiliation(s)
- Ido Badash
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Patricia M Quiñones
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Kevin J Oghalai
- Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Juemei Wang
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Christopher G Lui
- Department of Otolaryngology-Head and Neck Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Frank Macias-Escriva
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Brian E Applegate
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States.,Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - John S Oghalai
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States.,Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
22
|
Diaz GH, Heller S. Fluorescent in situ mRNA detection in the adult mouse cochlea. STAR Protoc 2021; 2:100711. [PMID: 34401777 PMCID: PMC8358468 DOI: 10.1016/j.xpro.2021.100711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Ossification and the delicateness of the cochlear duct make histologic assessments of the mature cochlea a challenging endeavor. Treatments to soften the bone facilitate sectioning and dissection of the cochlear duct but limit in situ mRNA detection in such specimens. Here, we provide a protocol for in situ mRNA detection using hybridization chain reaction in whole-mount preparations of the adult mouse cochlea. We show examples for multi-probe detection of different mRNAs and describe combination of this method with conventional immunohistochemistry.
Collapse
Affiliation(s)
- Giovanni H. Diaz
- Department of Otolaryngology – Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Stefan Heller
- Department of Otolaryngology – Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
23
|
Toulemonde P, Risoud M, Lemesre PE, Beck C, Wattelet J, Tardivel M, Siepmann J, Vincent C. Evaluation of the Efficacy of Dexamethasone-Eluting Electrode Array on the Post-Implant Cochlear Fibrotic Reaction by Three-Dimensional Immunofluorescence Analysis in Mongolian Gerbil Cochlea. J Clin Med 2021; 10:jcm10153315. [PMID: 34362099 PMCID: PMC8347204 DOI: 10.3390/jcm10153315] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 11/16/2022] Open
Abstract
Cochlear implant is the method of choice for the rehabilitation of severe to profound sensorineural hearing loss. The study of the tissue response to cochlear implantation and the prevention of post-cochlear-implant damages are areas of interest in hearing protection research. The objective was to assess the efficacy of dexamethasone-eluting electrode array on endo canal fibrosis formation by three-dimensional immunofluorescence analysis in implanted Mongolian gerbil cochlea. Two trials were conducted after surgery using Mongolian gerbil implanted with dexamethasone-eluting or non-eluting intracochlear electrode arrays. The animals were then euthanised 10 weeks after implantation. The cochleae were prepared (electrode array in place) according to a 29-day protocol with immunofluorescent labelling and tissue clearing. The acquisition was carried out using light-sheet microscopy. Imaris software was then used for three-dimensional analysis of the cochleae and quantification of the fibrotic volume. The analysis of 12 cochleae showed a significantly different mean volume of fibrosis (2.16 × 108 μm3 ± 0.15 in the dexamethasone eluting group versus 3.17 × 108 μm3 ± 0.54 in the non-eluting group) (p = 0.004). The cochlear implant used as a corticosteroid delivery system appears to be an encouraging device for the protection of the inner ear against fibrosis induced by implantation. Three-dimensional analysis of the cochlea by light-sheet microscopy was suitable for studying post-implantation tissue damage.
Collapse
Affiliation(s)
- Philippine Toulemonde
- Department of Otology and Neurotology, CHU Lille, University of Lille 2 Henri Warembourg, F-59000 Lille, France; (M.R.); (P.E.L.); (C.B.); (J.W.); (J.S.); (C.V.)
- INSERM U1008—Controlled Drug Delivery Systems and Biomaterials, F-59000 Lille, France
- Correspondence: ; Tel.: +33-6851-91052
| | - Michaël Risoud
- Department of Otology and Neurotology, CHU Lille, University of Lille 2 Henri Warembourg, F-59000 Lille, France; (M.R.); (P.E.L.); (C.B.); (J.W.); (J.S.); (C.V.)
- INSERM U1008—Controlled Drug Delivery Systems and Biomaterials, F-59000 Lille, France
| | - Pierre Emmanuel Lemesre
- Department of Otology and Neurotology, CHU Lille, University of Lille 2 Henri Warembourg, F-59000 Lille, France; (M.R.); (P.E.L.); (C.B.); (J.W.); (J.S.); (C.V.)
- INSERM U1008—Controlled Drug Delivery Systems and Biomaterials, F-59000 Lille, France
| | - Cyril Beck
- Department of Otology and Neurotology, CHU Lille, University of Lille 2 Henri Warembourg, F-59000 Lille, France; (M.R.); (P.E.L.); (C.B.); (J.W.); (J.S.); (C.V.)
- INSERM U1008—Controlled Drug Delivery Systems and Biomaterials, F-59000 Lille, France
| | - Jean Wattelet
- Department of Otology and Neurotology, CHU Lille, University of Lille 2 Henri Warembourg, F-59000 Lille, France; (M.R.); (P.E.L.); (C.B.); (J.W.); (J.S.); (C.V.)
- INSERM U1008—Controlled Drug Delivery Systems and Biomaterials, F-59000 Lille, France
| | - Meryem Tardivel
- BioImaging Center Lille-Nord de France (BICeL), University of Lille 2 Henri Warembourg, F-59000 Lille, France;
| | - Juergen Siepmann
- Department of Otology and Neurotology, CHU Lille, University of Lille 2 Henri Warembourg, F-59000 Lille, France; (M.R.); (P.E.L.); (C.B.); (J.W.); (J.S.); (C.V.)
- INSERM U1008—Controlled Drug Delivery Systems and Biomaterials, F-59000 Lille, France
| | - Christophe Vincent
- Department of Otology and Neurotology, CHU Lille, University of Lille 2 Henri Warembourg, F-59000 Lille, France; (M.R.); (P.E.L.); (C.B.); (J.W.); (J.S.); (C.V.)
- INSERM U1008—Controlled Drug Delivery Systems and Biomaterials, F-59000 Lille, France
| |
Collapse
|
24
|
Bryant JP, Chandrashekhar V, Cappadona AJ, Lookian PP, Chandrashekhar V, Donahue DR, Munasinghe JB, Kim HJ, Vortmeyer AO, Heiss JD, Zhuang Z, Rosenblum JS. Multimodal Atlas of the Murine Inner Ear: From Embryo to Adult. Front Neurol 2021; 12:699674. [PMID: 34335453 PMCID: PMC8319626 DOI: 10.3389/fneur.2021.699674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/22/2021] [Indexed: 12/02/2022] Open
Abstract
The inner ear is a complex organ housed within the petrous bone of the skull. Its intimate relationship with the brain enables the transmission of auditory and vestibular signals via cranial nerves. Development of this structure from neural crest begins in utero and continues into early adulthood. However, the anatomy of the murine inner ear has only been well-characterized from early embryogenesis to post-natal day 6. Inner ear and skull base development continue into the post-natal period in mice and early adulthood in humans. Traditional methods used to evaluate the inner ear in animal models, such as histologic sectioning or paint-fill and corrosion, cannot visualize this complex anatomy in situ. Further, as the petrous bone ossifies in the postnatal period, these traditional techniques become increasingly difficult. Advances in modern imaging, including high resolution Micro-CT and MRI, now allow for 3D visualization of the in situ anatomy of organs such as the inner ear. Here, we present a longitudinal atlas of the murine inner ear using high resolution ex vivo Micro-CT and MRI.
Collapse
Affiliation(s)
- Jean-Paul Bryant
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Vikram Chandrashekhar
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.,Center for Imaging Science, Johns Hopkins University, Baltimore, MD, United States
| | - Anthony J Cappadona
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Pashayar P Lookian
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States.,Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | | | - Danielle R Donahue
- Mouse Imaging Facility, National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States
| | - Jeeva B Munasinghe
- Mouse Imaging Facility, National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States
| | - H Jeffrey Kim
- Department of Otolaryngology, Georgetown University School of Medicine, Washington, DC, United States.,Office of Clinical Director, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, United States
| | - Alexander O Vortmeyer
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - John D Heiss
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Zhengping Zhuang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jared S Rosenblum
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States.,Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
25
|
Beaulac HJ, Gilels F, Zhang J, Jeoung S, White PM. Primed to die: an investigation of the genetic mechanisms underlying noise-induced hearing loss and cochlear damage in homozygous Foxo3-knockout mice. Cell Death Dis 2021; 12:682. [PMID: 34234110 PMCID: PMC8263610 DOI: 10.1038/s41419-021-03972-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023]
Abstract
The prevalence of noise-induced hearing loss (NIHL) continues to increase, with limited therapies available for individuals with cochlear damage. We have previously established that the transcription factor FOXO3 is necessary to preserve outer hair cells (OHCs) and hearing thresholds up to two weeks following mild noise exposure in mice. The mechanisms by which FOXO3 preserves cochlear cells and function are unknown. In this study, we analyzed the immediate effects of mild noise exposure on wild-type, Foxo3 heterozygous (Foxo3+/-), and Foxo3 knock-out (Foxo3-/-) mice to better understand FOXO3's role(s) in the mammalian cochlea. We used confocal and multiphoton microscopy to examine well-characterized components of noise-induced damage including calcium regulators, oxidative stress, necrosis, and caspase-dependent and caspase-independent apoptosis. Lower immunoreactivity of the calcium buffer Oncomodulin in Foxo3-/- OHCs correlated with cell loss beginning 4 h post-noise exposure. Using immunohistochemistry, we identified parthanatos as the cell death pathway for OHCs. Oxidative stress response pathways were not significantly altered in FOXO3's absence. We used RNA sequencing to identify and RT-qPCR to confirm differentially expressed genes. We further investigated a gene downregulated in the unexposed Foxo3-/- mice that may contribute to OHC noise susceptibility. Glycerophosphodiester phosphodiesterase domain containing 3 (GDPD3), a possible endogenous source of lysophosphatidic acid (LPA), has not previously been described in the cochlea. As LPA reduces OHC loss after severe noise exposure, we treated noise-exposed Foxo3-/- mice with exogenous LPA. LPA treatment delayed immediate damage to OHCs but was insufficient to ultimately prevent their death or prevent hearing loss. These results suggest that FOXO3 acts prior to acoustic insult to maintain cochlear resilience, possibly through sustaining endogenous LPA levels.
Collapse
MESH Headings
- Animals
- Cell Death
- Disease Models, Animal
- Female
- Forkhead Box Protein O3/deficiency
- Forkhead Box Protein O3/genetics
- Gene Expression Regulation
- Hair Cells, Auditory, Outer/drug effects
- Hair Cells, Auditory, Outer/metabolism
- Hair Cells, Auditory, Outer/pathology
- Hearing
- Hearing Loss, Noise-Induced/drug therapy
- Hearing Loss, Noise-Induced/genetics
- Hearing Loss, Noise-Induced/metabolism
- Hearing Loss, Noise-Induced/pathology
- Homozygote
- Lysophospholipids/metabolism
- Lysophospholipids/pharmacology
- Male
- Mice, Knockout
- Noise
- Phosphoric Diester Hydrolases/genetics
- Phosphoric Diester Hydrolases/metabolism
- Time Factors
- Mice
Collapse
Affiliation(s)
- Holly J Beaulac
- Department of Neuroscience, Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- The Jackson Laboratory, Bar Harbor, ME, USA
| | - Felicia Gilels
- Department of Neuroscience, Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Department of Pathology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Jingyuan Zhang
- Department of Neuroscience, Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Department of Otolaryngology, Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston Children's Hospital Center for Life Science, Boston, MA, USA
| | - Sarah Jeoung
- University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Patricia M White
- Department of Neuroscience, Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
26
|
Yu Y, Yang J, Luan F, Gu G, Zhao R, Wang Q, Dong Z, Tang J, Wang W, Sun J, Lv P, Zhang H, Wang C. Sensorineural Hearing Loss and Mitochondrial Apoptosis of Cochlear Spiral Ganglion Neurons in Fibroblast Growth Factor 13 Knockout Mice. Front Cell Neurosci 2021; 15:658586. [PMID: 34220452 PMCID: PMC8242186 DOI: 10.3389/fncel.2021.658586] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022] Open
Abstract
Deafness is known to occur in more than 400 syndromes and accounts for almost 30% of hereditary hearing loss. The molecular mechanisms underlying such syndromic deafness remain unclear. Furthermore, deafness has been a common feature in patients with three main syndromes, the BÖrjeson-Forssman-Lehmann syndrome, Wildervanck syndrome, and Congenital Generalized Hirsutism, all of which are characterized by loss-of-function mutations in the Fgf13 gene. Whether the pathogenesis of deafness in these syndromes is associated with the Fgf13 mutation is not known. To elucidate its role in auditory function, we generated a mouse line with conditional knockout of the Fgf13 gene in the inner ear (Fgf13 cKO). FGF13 is expressed predominantly in the organ of Corti, spiral ganglion neurons (SGNs), stria vascularis, and the supporting cells. Conditional knockout of the gene in the inner ear led to sensorineural deafness with low amplitude and increased latency of wave I in the auditory brainstem response test but had a normal distortion product otoacoustic emission threshold. Fgf13 deficiency resulted in decreased SGN density from the apical to the basal region without significant morphological changes and those in the number of hair cells. TUNEL and caspase-3 immunocytochemistry assays showed that apoptotic cell death mediated the loss of SGNs. Further detection of apoptotic factors through qRT-PCR suggested the activation of the mitochondrial apoptotic pathway in SGNs. Together, this study reveals a novel role for Fgf13 in auditory function, and indicates that the gene could be a potential candidate for understanding deafness. These findings may provide new perspectives on the molecular mechanisms and novel therapeutic targets for treatment deafness.
Collapse
Affiliation(s)
- Yulou Yu
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Jing Yang
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Feng Luan
- Department of Otolaryngology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guoqiang Gu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ran Zhao
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Qiong Wang
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Zishan Dong
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Junming Tang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Wei Wang
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Jinpeng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ping Lv
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Hailin Zhang
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Chuan Wang
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
27
|
Yang S, Ma N, Wu X, Ni H, Gao S, Sun L, Zhou P, Tala, Ran J, Zhou J, Liu M, Li D. CYLD deficiency causes auditory neuropathy due to reduced neurite outgrowth. J Clin Lab Anal 2021; 35:e23783. [PMID: 33934395 PMCID: PMC8183908 DOI: 10.1002/jcla.23783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/19/2021] [Accepted: 03/28/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Auditory neuropathy is a cause of hearing loss that has been studied in a number of animal models. Signal transmission from hair cells to spiral ganglion neurons plays an important role in normal hearing. CYLD is a microtubule-binding protein, and deubiquitinase involved in the regulation of various cellular processes. In this study, we used Cyld knockout (KO) mice and nerve cell lines to examine whether CYLD is associated with auditory neuropathy. METHODS Hearing of Cyld KO mice was studied using the TDT RZ6 auditory physiology workstation. The expression and localization of CYLD in mouse cochlea and cell lines were examined by RT-PCR, immunoblotting, and immunofluorescence. CYLD expression was knocked down in SH-SY5Y cells by shRNAs and in PC12 and N2A cells by siRNAs. Nerve growth factor and retinoic acid were used to induce neurite outgrowth, and the occurrence and length of neurites were statistically analyzed between knockdown and control groups. RESULTS Cyld KO mice had mild hearing impairment. Moreover, CYLD was widely expressed in mouse cochlear tissues and different nerve cell lines. Knocking down CYLD significantly reduced the length and proportion of neurites growing from nerve cells. CONCLUSIONS The abnormal hearing of Cyld KO mice might be caused by a decrease in the length and number of neurites growing from auditory nerve cells in the cochlea, suggesting that CYLD is a key protein affecting hearing.
Collapse
Affiliation(s)
- Song Yang
- State Key Laboratory of Medicinal Chemical BiologyCollege of Life SciencesNankai UniversityTianjinChina
| | - Nan Ma
- State Key Laboratory of Medicinal Chemical BiologyCollege of Life SciencesNankai UniversityTianjinChina
| | - Xuemei Wu
- State Key Laboratory of Medicinal Chemical BiologyCollege of Life SciencesNankai UniversityTianjinChina
| | - Hua Ni
- State Key Laboratory of Medicinal Chemical BiologyCollege of Life SciencesNankai UniversityTianjinChina
| | - Siqi Gao
- State Key Laboratory of Medicinal Chemical BiologyCollege of Life SciencesNankai UniversityTianjinChina
| | - Lei Sun
- State Key Laboratory of Medicinal Chemical BiologyCollege of Life SciencesNankai UniversityTianjinChina
| | - Peng Zhou
- Shandong Provincial Key Laboratory of Animal Resistance BiologyCollaborative Innovation Center of Cell Biology in Universities of ShandongInstitute of Biomedical SciencesCollege of Life SciencesShandong Normal UniversityJinanChina
| | - Tala
- State Key Laboratory of Medicinal Chemical BiologyCollege of Life SciencesNankai UniversityTianjinChina
| | - Jie Ran
- Shandong Provincial Key Laboratory of Animal Resistance BiologyCollaborative Innovation Center of Cell Biology in Universities of ShandongInstitute of Biomedical SciencesCollege of Life SciencesShandong Normal UniversityJinanChina
| | - Jun Zhou
- State Key Laboratory of Medicinal Chemical BiologyCollege of Life SciencesNankai UniversityTianjinChina
- Shandong Provincial Key Laboratory of Animal Resistance BiologyCollaborative Innovation Center of Cell Biology in Universities of ShandongInstitute of Biomedical SciencesCollege of Life SciencesShandong Normal UniversityJinanChina
| | - Min Liu
- Shandong Provincial Key Laboratory of Animal Resistance BiologyCollaborative Innovation Center of Cell Biology in Universities of ShandongInstitute of Biomedical SciencesCollege of Life SciencesShandong Normal UniversityJinanChina
| | - Dengwen Li
- State Key Laboratory of Medicinal Chemical BiologyCollege of Life SciencesNankai UniversityTianjinChina
| |
Collapse
|
28
|
In Situ 3D-Imaging of the Inner Ear Synapses with a Cochlear Implant. Life (Basel) 2021; 11:life11040301. [PMID: 33915846 PMCID: PMC8066088 DOI: 10.3390/life11040301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/27/2021] [Accepted: 03/28/2021] [Indexed: 11/17/2022] Open
Abstract
In recent years sensorineural hearing loss was found to affect not exclusively, nor at first, the sensory cells of the inner ear. The sensory cells' synapses and subsequent neurites are initially damaged. Auditory synaptopathies also play an important role in cochlear implant (CI) care, as they can lead to a loss of physiological hearing in patients with residual hearing. These auditory synaptopathies and in general the cascades of hearing pathologies have been in the focus of research in recent years with the aim to develop more targeted and individually tailored therapeutics. In the current study, a method to examine implanted inner ears of guinea pigs was developed to examine the synapse level. For this purpose, the cochlea is made transparent and scanned with the implant in situ using confocal laser scanning microscopy. Three different preparation methods were compared to enable both an overview image of the cochlea for assessing the CI position and images of the synapses on the same specimen. The best results were achieved by dissection of the bony capsule of the cochlea.
Collapse
|
29
|
Juergens L, Bieniussa L, Voelker J, Hagen R, Rak K. Spatio-temporal distribution of tubulin-binding cofactors and posttranslational modifications of tubulin in the cochlea of mice. Histochem Cell Biol 2020; 154:671-681. [PMID: 32712744 PMCID: PMC7723944 DOI: 10.1007/s00418-020-01905-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2020] [Indexed: 02/06/2023]
Abstract
The five tubulin-binding cofactors (TBC) are involved in tubulin synthesis and the formation of microtubules. Their importance is highlighted by various diseases and syndromes caused by dysfunction or mutation of these proteins. Posttranslational modifications (PTMs) of tubulin promote different characteristics, including stability-creating subpopulations of tubulin. Cell- and time-specific distribution of PTMs has only been investigated in the organ of Corti in gerbils. The aim of the presented study was to investigate the cell type-specific and time-specific expression patterns of TBC proteins and PTMs for the first time in murine cochleae over several developmental stages. For this, murine cochleae were investigated at the postnatal (P) age P1, P7 and P14 by immunofluorescence analysis. The investigations revealed several profound interspecies differences in the distribution of PTMs between gerbil and mouse. Furthermore, this is the first study to describe the spatio-temporal distribution of TBCs in any tissue ever showing a volatile pattern of expression. The expression analysis of TBC proteins and PTMs of tubulin reveals that these proteins play a role in the physiological development of the cochlea and might be essential for hearing.
Collapse
Affiliation(s)
- Lukas Juergens
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, The Comprehensive Hearing Center, University of Wuerzburg, Josef-Schneider-Strasse 11, 97080, Wuerzburg, Germany
- Department of Ophthalmology, University of Duesseldorf, Duesseldorf, Germany
| | - Linda Bieniussa
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, The Comprehensive Hearing Center, University of Wuerzburg, Josef-Schneider-Strasse 11, 97080, Wuerzburg, Germany
| | - Johannes Voelker
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, The Comprehensive Hearing Center, University of Wuerzburg, Josef-Schneider-Strasse 11, 97080, Wuerzburg, Germany
| | - Rudolf Hagen
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, The Comprehensive Hearing Center, University of Wuerzburg, Josef-Schneider-Strasse 11, 97080, Wuerzburg, Germany
| | - Kristen Rak
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, The Comprehensive Hearing Center, University of Wuerzburg, Josef-Schneider-Strasse 11, 97080, Wuerzburg, Germany.
| |
Collapse
|
30
|
Qian ZJ, Ricci AJ. Effects of cochlear hair cell ablation on spatial learning/memory. Sci Rep 2020; 10:20687. [PMID: 33244175 PMCID: PMC7692547 DOI: 10.1038/s41598-020-77803-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022] Open
Abstract
Current clinical interest lies in the relationship between hearing loss and cognitive impairment. Previous work demonstrated that noise exposure, a common cause of sensorineural hearing loss (SNHL), leads to cognitive impairments in mice. However, in noise-induced models, it is difficult to distinguish the effects of noise trauma from subsequent SNHL on central processes. Here, we use cochlear hair cell ablation to isolate the effects of SNHL. Cochlear hair cells were conditionally and selectively ablated in mature, transgenic mice where the human diphtheria toxin (DT) receptor was expressed behind the hair-cell specific Pou4f3 promoter. Due to higher Pou4f3 expression in cochlear hair cells than vestibular hair cells, administration of a low dose of DT caused profound SNHL without vestibular dysfunction and had no effect on wild-type (WT) littermates. Spatial learning/memory was assayed using an automated radial 8-arm maze (RAM), where mice were trained to find food rewards over a 14-day period. The number of working memory errors (WME) and reference memory errors (RME) per training day were recorded. All animals were injected with DT during P30-60 and underwent the RAM assay during P90-120. SNHL animals committed more WME and RME than WT animals, demonstrating that isolated SNHL affected cognitive function. Duration of SNHL (60 versus 90 days post DT injection) had no effect on RAM performance. However, younger age of acquired SNHL (DT on P30 versus P60) was associated with fewer WME. This describes the previously undocumented effect of isolated SNHL on cognitive processes that do not directly rely on auditory sensory input.
Collapse
MESH Headings
- Animals
- Cognition/physiology
- Deafness/metabolism
- Deafness/physiopathology
- Evoked Potentials, Auditory, Brain Stem/physiology
- Hair Cells, Auditory/metabolism
- Hair Cells, Auditory/physiology
- Hair Cells, Vestibular/metabolism
- Hair Cells, Vestibular/physiology
- Hearing/physiology
- Hearing Loss, Sensorineural/metabolism
- Hearing Loss, Sensorineural/physiopathology
- Heparin-binding EGF-like Growth Factor/metabolism
- Memory/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Noise
- Spatial Learning/physiology
- Transcription Factor Brn-3C/metabolism
Collapse
Affiliation(s)
- Z Jason Qian
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 240 Pasteur Drive, Biomedical Innovations Building, R0551, Palo Alto, CA, 94304, USA
| | - Anthony J Ricci
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 240 Pasteur Drive, Biomedical Innovations Building, R0551, Palo Alto, CA, 94304, USA.
| |
Collapse
|
31
|
The Notch Ligand Jagged1 Is Required for the Formation, Maintenance, and Survival of Hensen's Cells in the Mouse Cochlea. J Neurosci 2020; 40:9401-9413. [PMID: 33127852 DOI: 10.1523/jneurosci.1192-20.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 10/16/2020] [Accepted: 10/23/2020] [Indexed: 01/09/2023] Open
Abstract
During cochlear development, the Notch ligand JAGGED 1 (JAG1) plays an important role in the specification of the prosensory region, which gives rise to sound-sensing hair cells and neighboring supporting cells (SCs). While JAG1's expression is maintained in SCs through adulthood, the function of JAG1 in SC development is unknown. Here, we demonstrate that JAG1 is essential for the formation and maintenance of Hensen's cells, a highly specialized SC subtype located at the edge of the auditory epithelium. Using Sox2 CreERT2/+::Jag1loxP/loxP mice of both genders, we show that Jag1 deletion at the onset of differentiation, at embryonic day 14.5, disrupted Hensen's cell formation. Similar loss of Hensen's cells was observed when Jag1 was deleted after Hensen's cell formation at postnatal day (P) 0/P1 and fate-mapping analysis revealed that in the absence of Jag1, some Hensen's cells die, but others convert into neighboring Claudius cells. In support of a role for JAG1 in cell survival, genes involved in mitochondrial function and protein synthesis were downregulated in the sensory epithelium of P0 cochlea lacking Jag1 Finally, using Fgfr3-iCreERT2 ::Jag1loxP/loxP mice to delete Jag1 at P0, we observed a similar loss of Hensen's cells and found that adult Jag1 mutant mice have hearing deficits at the low-frequency range.SIGNIFICANCE STATEMENT Hensen's cells play an essential role in the development and homeostasis of the cochlea. Defects in the biophysical or functional properties of Hensen's cells have been linked to auditory dysfunction and hearing loss. Despite their importance, surprisingly little is known about the molecular mechanisms that guide their development. Morphologic and fate-mapping analyses in our study revealed that, in the absence of the Notch ligand JAGGED1, Hensen's cells died or converted into Claudius cells, which are specialized epithelium-like cells outside the sensory epithelium. Confirming a link between JAGGED1 and cell survival, transcriptional profiling showed that JAGGED1 maintains genes critical for mitochondrial function and tissue homeostasis. Finally, auditory phenotyping revealed that JAGGED1's function in supporting cells is necessary for low-frequency hearing.
Collapse
|
32
|
Ghosh S, Lewis MB, Walters BJ. Comparison of ethylenediaminetetraacetic acid and rapid decalcificier solution for studying human temporal bones by immunofluorescence. Laryngoscope Investig Otolaryngol 2020; 5:919-927. [PMID: 33134540 PMCID: PMC7585256 DOI: 10.1002/lio2.449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/21/2020] [Accepted: 08/03/2020] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVES The pervasiveness of hearing loss and the development of new potential therapeutic approaches have led to increased animal studies of the inner ear. However, translational relevance of such studies depends upon verification of protein localization data in human samples. Cadavers used for anatomical education provide a potential research resource, but are limiting due to difficulties in accessing sensory tissues from the dense temporal bones. This study seeks to reduce the often months-long process of decalcification and improve immunofluorescent staining of human cadaveric temporal bones for research use. METHODS Temporal bones were decalcified in either (a) hydrochloric acid-containing RDO solution for 2 days followed by 0.5 M ethylenediaminetetraacetic acid (EDTA) for 3 to 5 additional days, or (b) 0.5 M EDTA alone for 2 to 4 weeks. Image-iT FX signal enhancer (ISE) was used to improve immunofluorescent signal-to-noise ratios. RESULTS The data indicate that both methods speed decalcification and allow for immunolabeling of the extranuclear proteins neurofilament (heavy chain), myosin VIIa, oncomodulin and prestin. However, RDO decalcification was more likely to alter structural morphology of sensory tissues and hindered effective labeling of the nuclear proteins SRY-box transcription factor 2 and GATA binding protein 3. CONCLUSIONS Although both approaches allow for rapid decalcification, EDTA appears superior to RDO for preserving cytoarchitecture and immunogenicity. LEVEL OF EVIDENCE NA.
Collapse
Affiliation(s)
- Sumana Ghosh
- Department of Neurobiology and Anatomical SciencesUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Mark B. Lewis
- Department of Neurobiology and Anatomical SciencesUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Bradley J. Walters
- Department of Neurobiology and Anatomical SciencesUniversity of Mississippi Medical CenterJacksonMississippiUSA
- Department Otolaryngology—Head and Neck SurgeryUniversity of Mississippi Medical CenterJacksonMississippiUSA
| |
Collapse
|
33
|
Intratympanic Diltiazem-Chitosan Hydrogel as an Otoprotectant Against Cisplatin-Induced Ototoxicity in a Mouse Model. Otol Neurotol 2020; 41:115-122. [PMID: 31746818 DOI: 10.1097/mao.0000000000002417] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
HYPOTHESIS Local administration of the calcium-channel blocker (CCB), diltiazem, via intratympanic (IT) chitosan-glycerophosphate (CGP) hydrogel will protect against cisplatin-induced ototoxicity. BACKGROUND Cisplatin induces calcium-mediated apoptosis of cochlear outer hair cells (OHCs). Previous work demonstrated otoprotection and reduced auditory brainstem response (ABR) threshold shifts in a cisplatin-induced ototoxicity mouse model treated with multiple doses of IT diltiazem given in solution. Here, we evaluated the role of a single dose of IT CGP-diltiazem as a novel otoprotectant against cisplatin-induced ototoxicity. METHODS Baseline pure-tone and click-evoked ABRs were performed in control (IT CGP-saline, n = 13) and treatment (IT CGP-diltiazem 2 mg/kg, n = 9) groups of female CBA/J mice. A single dose of IT CGP hydrogel was administered just before intraperitoneal injection of cisplatin (14 mg/kg). On Day 7 posttreatment, ABRs were performed and cochleae were harvested. Hair cells were quantified using anti-myosin VIIa immunostaining and inner hair cell ribbon synapses were quantified using Ctbp2 immunostaining. RESULTS There was a statistically significant effect of treatment on click- and tone-evoked ABRs between groups. The mean threshold shifts were significantly reduced in both click- and tone-evoked ABRs on Day 7 in IT CGP-diltiazem treated mice compared with CGP-saline control mice. There were no significant differences in OHC counting between groups, but there appears to be an otoprotection against loss of synapses in the apical turn from IT CGP-diltiazem treated mice (p < 0.05). CONCLUSIONS This preliminary work suggests that IT CGP-diltiazem reduces ABR threshold shifts with possible mechanisms of protecting ribbon synapses in the setting of cisplatin-induced ototoxicity. More work is necessary to determine the mechanism underlying this otoprotection.
Collapse
|
34
|
Patel S, Shah L, Dang N, Tan X, Almudevar A, White PM. SIRT3 promotes auditory function in young adult FVB/nJ mice but is dispensable for hearing recovery after noise exposure. PLoS One 2020; 15:e0235491. [PMID: 32658908 PMCID: PMC7357743 DOI: 10.1371/journal.pone.0235491] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 06/16/2020] [Indexed: 01/08/2023] Open
Abstract
Noise-induced hearing loss (NIHL) affects millions of people worldwide and presents a large social and personal burden. Pharmacological activation of SIRT3, a regulator of the mitochondrial oxidative stress response, has a protective effect on hearing thresholds after traumatic noise damage in mice. In contrast, the role of endogenously activated SIRT3 in hearing recovery has not been established. Here we tested the hypothesis that SIRT3 is required in mice for recovery of auditory thresholds after a noise exposure that confers a temporary threshold shift (TTS). SIRT3-specific immunoreactivity is present in outer hair cells, around the post-synaptic regions of inner hair cells, and faintly within inner hair cells. Prior to noise exposure, homozygous Sirt3-KO mice have slightly but significantly higher thresholds than their wild-type littermates measured by the auditory brainstem response (ABR), but not by distortion product otoacoustic emissions (DPOAE). Moreover, homozygous Sirt3-KO mice display a significant reduction in the progression of their peak 1 amplitude at higher frequencies prior to noise exposure. After exposure to a single sub-traumatic noise dose that does not permanently reduce cochlear function, compromise cell survival, or damage synaptic structures in wild-type mice, there was no difference in hearing function between the two genotypes, measured by ABR and DPOAE. The numbers of hair cells and auditory synapses were similar in both genotypes before and after noise exposure. These loss-of-function studies complement previously published gain-of-function studies and help refine our understanding of SIRT3’s role in cochlear homeostasis under different damage paradigms. They suggest that SIRT3 may promote spiral ganglion neuron function. They imply that cellular mechanisms of homeostasis, in addition to the mitochondrial oxidative stress response, act to restore hearing after TTS. Finally, we present a novel application of a biomedical statistical analysis for identifying changes between peak 1 amplitude progressions in ABR waveforms after damage.
Collapse
Affiliation(s)
- Sally Patel
- Department of Neuroscience, Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Lisa Shah
- Department of Neuroscience, Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Natalie Dang
- Department of Neuroscience, Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Xiaodong Tan
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Anthony Almudevar
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Patricia M. White
- Department of Neuroscience, Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
35
|
Ballesteros A, Swartz KJ. Dextran Labeling and Uptake in Live and Functional Murine Cochlear Hair Cells. J Vis Exp 2020. [PMID: 32090986 PMCID: PMC11384666 DOI: 10.3791/60769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The hair cell mechanotransduction (MET) channel plays an important role in hearing. However, the molecular identity and structural information of MET remain unknown. Electrophysiological studies of hair cells revealed that the MET channel has a large conductance and is permeable to relatively large fluorescent cationic molecules, including some styryl dyes and Texas Red-labeled aminoglycoside antibiotics. In this protocol, we describe a method to visualize and evaluate the uptake of fluorescent dextrans in hair cells of the organ of Corti explants that can be used to assay for functional MET channels. We found that 3 kDa Texas Red-labeled dextran specifically labels functional auditory hair cells after 1-2 h incubation. In particular, 3 kDa dextran labels the two shorter stereocilia rows and accumulates in the cell body in a diffuse pattern when functional MET channels are present. An additional vesicle-like pattern of labeling was observed in the cell body of hair cells and surrounding supporting cells. Our data suggest that 3 kDa Texas-Red dextran can be used to visualize and study two pathways for cellular dye uptake; a hair cell-specific entry route through functional MET channels and endocytosis, a pattern also available to larger dextran.
Collapse
Affiliation(s)
- Angela Ballesteros
- Molecular Physiology and Biophysics Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health;
| | - Kenton J Swartz
- Molecular Physiology and Biophysics Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health
| |
Collapse
|
36
|
Abstract
Auditory processing in the cochlea depends on the integrity of the mechanosensory hair cells. Over a lifetime, hearing loss can be acquired from numerous etiologies such as exposure to excessive noise, the use of ototoxic medications, bacterial or viral ear infections, head injuries, and the aging process. Loss of sensory hair cells is a common pathological feature of the varieties of acquired hearing loss. Additionally, the inner hair cell synapse can be damaged by mild insults. Therefore, surface preparations of cochlear epithelia, in combination with immunolabeling techniques and confocal imagery, are a very useful tool for the investigation of cochlear pathologies, including losses of ribbon synapses and sensory hair cells, changes in protein levels in hair cells and supporting cells, hair cell regeneration, and determination of report gene expression (i.e., GFP) for verification of successful transduction and identification of transduced cell types. The cochlea, a bony spiral-shaped structure in the inner ear, holds the auditory sensory end organ, the organ of Corti (OC). Sensory hair cells and surrounding supporting cells in the OC are contained in the cochlear duct and rest on the basilar membrane, organized in a tonotopic fashion with high-frequency detection occurring in the base and low-frequency in the apex. With the availability of molecular and genetic information and the ability to manipulate genes by knockout and knock-in techniques, mice have been widely used in biological research, including in hearing science. However, the adult mouse cochlea is miniscule, and the cochlear epithelium is encapsulated in a bony labyrinth, making microdissection difficult. Although dissection techniques have been developed and used in many laboratories, this modified microdissection method using cell and tissue adhesive is easier and more convenient. It can be used in all types of adult mouse cochleae following decalcification.
Collapse
Affiliation(s)
- Qiao-Jun Fang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina; MOE Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University
| | - Fan Wu
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina
| | - Renjie Chai
- MOE Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University
| | - Su-Hua Sha
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina;
| |
Collapse
|
37
|
Kwak SH, Nam GS, Bae SH, Jung J. Effect of Specific Retinoic Acid Receptor Agonists on Noise-Induced Hearing Loss. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16183428. [PMID: 31527426 PMCID: PMC6765908 DOI: 10.3390/ijerph16183428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 12/27/2022]
Abstract
Noise is one of the most common causes of hearing loss in industrial countries. There are many studies about chemical agents to prevent noise-induced hearing loss (NIHL). However, there is no commercially available drug yet. Retinoic acid is an active metabolite of Vitamin A; it has an anti-apoptic role in NIHL. This study aims to verify the differences among selective agonists of retinoic acid receptors (RARs) in NIHL. All-trans retinoic acid (ATRA), AM80 (selective retinoic acid receptor α agonist), AC261066 (Selective retinoic acid receptor β1 agonist), and CD1530 (Selective retinoic acid λ agonist) were injected to 6–7 weeks old CJ5BL/6 mice before noise (110 dB for 3 h) exposure. In the auditory brainstem response test pre-, post 1, 3, and 7 days after noise exposure, not only ATRA but all kinds of selective RAR agonists showed protective effects in hearing threshold and wave I amplitude. Though there was no significant difference in the level of protective effects between agonists, α agonist showed the most prominent effect in preserving hearing function as well as outer hair cells after noise exposure. In conclusion, selective agonists of RAR demonstrate comparable protective effects against NIHL to retinoic acid. Given that these selective RAR agonists have less side effects than retinoic acid, they may be promising potential drugs against NIHL.
Collapse
Affiliation(s)
- Sang Hyun Kwak
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Korea.
| | - Gi-Sung Nam
- Department of Otorhinolaryngology, Chonbuk National University College of Medicine, Jeonju 54907, Korea.
| | - Seong Hoon Bae
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Korea.
| | - Jinsei Jung
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Korea.
| |
Collapse
|
38
|
Carignano C, Barila EP, Rías EI, Dionisio L, Aztiria E, Spitzmaul G. Inner Hair Cell and Neuron Degeneration Contribute to Hearing Loss in a DFNA2-Like Mouse Model. Neuroscience 2019; 410:202-216. [PMID: 31102762 DOI: 10.1016/j.neuroscience.2019.05.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/03/2019] [Accepted: 05/07/2019] [Indexed: 01/19/2023]
Abstract
DFNA2 is a progressive deafness caused by mutations in the voltage-activated potassium channel KCNQ4. Hearing loss develops with age from a mild increase in the hearing threshold to profound deafness. Studies using transgenic mice for Kcnq4 expressed in a mixed background demonstrated the implication of outer hair cells at the initial phase. However, it could not explain the last phase mechanisms of the disease. Genetic backgrounds are known to influence disease expressivity. To unmask the cause of profound deafness phenotype, we backcrossed the Kcnq4 knock-out allele to the inbred strain C3H/HeJ and investigated inner and outer hair cell and spiral ganglion neuron degeneration across the lifespan. In addition to the already reported outer hair cell death, the C3H/HeJ strain also exhibited inner hair cell and spiral ganglion neuron death. We tracked the spatiotemporal survival of cochlear cells by plotting cytocochleograms and neuronal counts at different ages. Cell loss progressed from basal to apical turns with age. Interestingly, the time-course of cell degeneration was different for each cell-type. While for outer hair cells it was already present by week 3, inner hair cell and neuronal loss started 30 weeks later. We also established that outer hair cell loss kinetics slowed down from basal to apical regions correlating with KCNQ4 expression pattern determined in wild-type mice. Our findings indicate that KCNQ4 plays differential roles in each cochlear cell-type impacting in their survival ability. Inner hair cell and spiral ganglion neuron death generates severe hearing loss that could be associated with the last phase of DFNA2.
Collapse
Affiliation(s)
- Camila Carignano
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad Nacional del Sur (UNS), Camino La Carrindanga Km 7, B8000FWB, Bahía Blanca, Argentina
| | - Esteban Pablo Barila
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad Nacional del Sur (UNS), Camino La Carrindanga Km 7, B8000FWB, Bahía Blanca, Argentina
| | - Ezequiel Ignacio Rías
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad Nacional del Sur (UNS), Camino La Carrindanga Km 7, B8000FWB, Bahía Blanca, Argentina.; Departamento de Biología, Bioquímica y Farmacia (BByF)-UNS, San Juan 670, 8000 Bahía Blanca, Argentina
| | - Leonardo Dionisio
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad Nacional del Sur (UNS), Camino La Carrindanga Km 7, B8000FWB, Bahía Blanca, Argentina.; Departamento de Biología, Bioquímica y Farmacia (BByF)-UNS, San Juan 670, 8000 Bahía Blanca, Argentina
| | - Eugenio Aztiria
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad Nacional del Sur (UNS), Camino La Carrindanga Km 7, B8000FWB, Bahía Blanca, Argentina.; Departamento de Biología, Bioquímica y Farmacia (BByF)-UNS, San Juan 670, 8000 Bahía Blanca, Argentina
| | - Guillermo Spitzmaul
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad Nacional del Sur (UNS), Camino La Carrindanga Km 7, B8000FWB, Bahía Blanca, Argentina.; Departamento de Biología, Bioquímica y Farmacia (BByF)-UNS, San Juan 670, 8000 Bahía Blanca, Argentina..
| |
Collapse
|
39
|
Yang J, Pan H, Mishina Y. Tissue Preparation and Immunostaining of Mouse Craniofacial Tissues and Undecalcified Bone. J Vis Exp 2019. [PMID: 31132049 DOI: 10.3791/59113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Tissue immunostaining provides highly specific and reliable detection of proteins of interest within a given tissue. Here we describe a complete and simple protocol to detect protein expression during craniofacial morphogenesis/pathogenesis using mouse craniofacial tissues as examples. The protocol consists of preparation and cryosectioning of tissues, indirect immunofluorescence, image acquisition, and quantification. In addition, a method for preparation and cryosectioning of undecalcified hard tissues for immunostaining is described, using craniofacial tissues and long bones as examples. Those methods are key to determine the protein expression and morphological/anatomical changes in various tissues during craniofacial morphogenesis/pathogenesis. They are also applicable to other tissues with appropriate modifications. Knowledge of the histology and high quality of sections are critical to draw scientific conclusions from experimental outcomes. Potential limitations of this methodology include but are not limited to specificity of antibodies and difficulties of quantification, which are also discussed here.
Collapse
Affiliation(s)
- Jingwen Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University; Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan
| | - Haichun Pan
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan;
| |
Collapse
|
40
|
Neagu AN. Proteome Imaging: From Classic to Modern Mass Spectrometry-Based Molecular Histology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1140:55-98. [PMID: 31347042 DOI: 10.1007/978-3-030-15950-4_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In order to overcome the limitations of classic imaging in Histology during the actually era of multiomics, the multi-color "molecular microscope" by its emerging "molecular pictures" offers quantitative and spatial information about thousands of molecular profiles without labeling of potential targets. Healthy and diseased human tissues, as well as those of diverse invertebrate and vertebrate animal models, including genetically engineered species and cultured cells, can be easily analyzed by histology-directed MALDI imaging mass spectrometry. The aims of this review are to discuss a range of proteomic information emerging from MALDI mass spectrometry imaging comparative to classic histology, histochemistry and immunohistochemistry, with applications in biology and medicine, concerning the detection and distribution of structural proteins and biological active molecules, such as antimicrobial peptides and proteins, allergens, neurotransmitters and hormones, enzymes, growth factors, toxins and others. The molecular imaging is very well suited for discovery and validation of candidate protein biomarkers in neuroproteomics, oncoproteomics, aging and age-related diseases, parasitoproteomics, forensic, and ecotoxicology. Additionally, in situ proteome imaging may help to elucidate the physiological and pathological mechanisms involved in developmental biology, reproductive research, amyloidogenesis, tumorigenesis, wound healing, neural network regeneration, matrix mineralization, apoptosis and oxidative stress, pain tolerance, cell cycle and transformation under oncogenic stress, tumor heterogeneity, behavior and aggressiveness, drugs bioaccumulation and biotransformation, organism's reaction against environmental penetrating xenobiotics, immune signaling, assessment of integrity and functionality of tissue barriers, behavioral biology, and molecular origins of diseases. MALDI MSI is certainly a valuable tool for personalized medicine and "Eco-Evo-Devo" integrative biology in the current context of global environmental challenges.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, "Alexandru Ioan Cuza" University of Iasi, Iasi, Romania.
| |
Collapse
|
41
|
Zhang J, Wang Q, Abdul-Aziz D, Mattiacio J, Edge ASB, White PM. ERBB2 signaling drives supporting cell proliferation in vitro and apparent supernumerary hair cell formation in vivo in the neonatal mouse cochlea. Eur J Neurosci 2018; 48:3299-3316. [PMID: 30270571 DOI: 10.1111/ejn.14183] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/07/2018] [Accepted: 09/03/2018] [Indexed: 12/29/2022]
Abstract
In mammals, cochlear hair cells are not regenerated once they are lost, leading to permanent hearing deficits. In other vertebrates, the adjacent supporting cells act as a stem cell compartment, in that they both proliferate and differentiate into de novo auditory hair cells. Although there is evidence that mammalian cochlear supporting cells can differentiate into new hair cells, the signals that regulate this process are poorly characterized. We hypothesize that signaling from the epidermal growth factor receptor (EGFR) family may play a role in cochlear regeneration. We focus on one such member, ERBB2, and report the effects of expressing a constitutively active ERBB2 receptor in neonatal mouse cochlear supporting cells, using viruses and transgenic expression. Lineage tracing with fluorescent reporter proteins was used to determine the relationships between cells with active ERBB2 signaling and cells that divided or differentiated into hair cells. In vitro, individual supporting cells harbouring a constitutively active ERBB2 receptor appeared to signal to their neighbouring supporting cells, inducing them to down-regulate a supporting cell marker and to proliferate. In vivo, we found supernumerary hair cell-like cells near supporting cells that expressed ERBB2 receptors. Both supporting cell proliferation and hair cell differentiation were largely reproduced in vitro using small molecules that we show also activate ERBB2. Our data suggest that signaling from the receptor tyrosine kinase ERBB2 can drive the activation of secondary signaling pathways to regulate regeneration, suggesting a new model where an interplay of cell signaling regulates regeneration by endogenous stem-like cells.
Collapse
Affiliation(s)
- Jingyuan Zhang
- Department of Biology, School of Arts and Sciences, University of Rochester, Rochester, New York
| | - Quan Wang
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts
| | - Dunia Abdul-Aziz
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts
| | - Jonelle Mattiacio
- Department of Microbiology and Immunology, University of Rochester School of Medicine, Rochester, New York
| | - Albert S B Edge
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts.,Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts.,Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Patricia M White
- Department of Neuroscience, The Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine, Rochester, New York
| |
Collapse
|
42
|
Trans-differentiation of outer hair cells into inner hair cells in the absence of INSM1. Nature 2018; 563:691-695. [PMID: 30305733 PMCID: PMC6279423 DOI: 10.1038/s41586-018-0570-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/14/2018] [Indexed: 01/09/2023]
Abstract
The mammalian cochlea contains two types of mechanosensory hair cells (HCs) that play different and critical roles in hearing. Inner hair cells (IHCs), with an elaborate presynaptic apparatus, signal to cochlear neurons and communicate sound information to the brain. Outer hair cells (OHCs) mechanically amplify sound-induced vibrations, enabling enhanced sensitivity to sound and sharp tuning. Cochlear HCs are solely generated during development and their death, most often of OHCs, is the main cause of deafness. OHCs and IHCs, together with supporting cells, originate embryonically from the prosensory region of the otocyst, but how HCs differentiate into two different types is unknown1–3. Here we show that Insm1, which encodes a zinc finger protein transiently expressed in nascent OHCs, consolidates their fate by preventing trans-differentiation into IHCs. In the absence of INSM1 many HCs born embryonically as OHCs switch fates to become mature IHCs. In order to identify the genetic mechanisms by which Insm1 operates, we compared transcriptomes of immature IHCs vs OHCs, as well as OHCs with and without INSM1. OHCs lacking INSM1 upregulate a set of genes, most of which are normally preferentially expressed by IHCs. The homeotic cell transformation of OHCs without INSM1 into IHCs reveals for the first time a mechanism by which these neighboring mechanosensory cells begin to differ: INSM1 represses a core set of early IHC-enriched genes in embryonic OHCs and makes them unresponsive to an IHC-inducing gradient, so that they proceed to mature as OHCs. Without INSM1, some of the OHCs upregulating these few IHC-enriched transcripts trans-differentiate into IHCs, revealing the first candidate genes for IHC-specific differentiation.
Collapse
|
43
|
Chronic cigarette smoke exposure drives spiral ganglion neuron loss in mice. Sci Rep 2018; 8:5746. [PMID: 29636532 PMCID: PMC5893541 DOI: 10.1038/s41598-018-24166-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/28/2018] [Indexed: 02/07/2023] Open
Abstract
Tobacco use is associated with an increased risk of hearing loss in older individuals, suggesting cigarette smoke (CS) exposure may target the peripheral auditory organs. However, the effects of CS exposure on general cochlear anatomy have not previously been explored. Here we compare control and chronic CS exposed cochleae from adult mice to assess changes in structure and cell survival. Two-photon imaging techniques, including the imaging of second harmonic generation (SHG) and two-photon excitation fluorescence (TPEF) from native molecules, were used to probe the whole cochlear organ for changes. We found evidence for fibrillar collagen accumulation in the spiral ganglion and organ of Corti, consistent with fibrosis. Quantitative TPEF indicated that basal CS-exposed spiral ganglion neurons experienced greater oxidative stress than control neurons, which was confirmed by histological staining for lipid peroxidation products. Cell counts confirmed that the CS-exposed spiral ganglion also contained fewer basal neurons. Taken together, these data support the premise that CS exposure induces oxidative stress in cochlear cells. They also indicate that two-photon techniques may screen cochlear tissues for oxidative stress.
Collapse
|
44
|
Wiwatpanit T, Remis NN, Ahmad A, Zhou Y, Clancy JC, Cheatham MA, García-Añoveros J. Codeficiency of Lysosomal Mucolipins 3 and 1 in Cochlear Hair Cells Diminishes Outer Hair Cell Longevity and Accelerates Age-Related Hearing Loss. J Neurosci 2018; 38:3177-3189. [PMID: 29453205 PMCID: PMC5884457 DOI: 10.1523/jneurosci.3368-17.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/16/2018] [Accepted: 02/02/2018] [Indexed: 01/11/2023] Open
Abstract
Acquired hearing loss is the predominant neurodegenerative condition associated with aging in humans. Although mutations on several genes are known to cause congenital deafness in newborns, few genes have been implicated in age-related hearing loss (ARHL), perhaps because its cause is likely polygenic. Here, we generated mice lacking lysosomal calcium channel mucolipins 3 and 1 and discovered that both male and female mice suffered a polygenic form of hearing loss. Whereas mucolipin 1 is ubiquitously expressed in all cells, mucolipin 3 is expressed in a small subset of cochlear cells, hair cells (HCs) and marginal cells of the stria vascularis, and very few other cell types. Mice lacking both mucolipins 3 and 1, but not either one alone, experienced hearing loss as early as at 1 month of age. The severity of hearing impairment progressed from high to low frequencies and increased with age. Early onset of ARHL in these mice was accompanied by outer HC (OHC) loss. Adult mice conditionally lacking mucolipins in HCs exhibited comparable auditory phenotypes, thereby revealing that the reason for OHC loss is mucolipin codeficiency in the HCs and not in the stria vascularis. Furthermore, we observed that OHCs lacking mucolipins contained abnormally enlarged lysosomes aggregated at the apical region of the cell, whereas other organelles appeared normal. We also demonstrated that these aberrant lysosomes in OHCs lost their membrane integrity through lysosomal membrane permeabilization, a known cause of cellular toxicity that explains why and how OHCs die, leading to premature ARHL.SIGNIFICANCE STATEMENT Presbycusis, or age-related hearing loss (ARHL), is a common characteristic of aging in mammals. Although many genes have been identified to cause deafness from birth in both humans and mice, only a few are known to associate with progressive ARHL, the most prevalent form of deafness. We have found that mice lacking two lysosomal channels, mucolipins 3 and 1, suffer accelerated ARHL due to auditory outer hair cell degeneration, the most common cause of hearing loss and neurodegenerative condition in humans. Lysosomes lacking mucolipins undergo organelle membrane permeabilization and promote cytotoxicity with age, revealing a novel mechanism of outer hair cell degeneration and ARHL. These results underscore the importance of lysosomes in hair cell survival and the maintenance of hearing.
Collapse
Affiliation(s)
- Teerawat Wiwatpanit
- Driskill Graduate Program in Life Sciences, Northwestern University, Chicago, Illinois 60611
- Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611
| | - Natalie N Remis
- Driskill Graduate Program in Life Sciences, Northwestern University, Chicago, Illinois 60611
- Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611
| | - Aisha Ahmad
- Communication Sciences and Disorders Knowles Hearing Center, Northwestern University, Evanston, Illinois 60208
| | - Yingjie Zhou
- Communication Sciences and Disorders Knowles Hearing Center, Northwestern University, Evanston, Illinois 60208
| | - John C Clancy
- Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611
| | - Mary Ann Cheatham
- Communication Sciences and Disorders Knowles Hearing Center, Northwestern University, Evanston, Illinois 60208
- Hugh Knowles Center for Clinical and Basic Science in Hearing and Its Disorders, Northwestern University, Chicago, Illinois 60611, and
| | - Jaime García-Añoveros
- Driskill Graduate Program in Life Sciences, Northwestern University, Chicago, Illinois 60611,
- Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611
- Hugh Knowles Center for Clinical and Basic Science in Hearing and Its Disorders, Northwestern University, Chicago, Illinois 60611, and
- Departments of Neurology and Physiology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611
| |
Collapse
|
45
|
Sheehan K, Sheth S, Mukherjea D, Rybak LP, Ramkumar V. Trans-Tympanic Drug Delivery for the Treatment of Ototoxicity. J Vis Exp 2018. [PMID: 29608150 DOI: 10.3791/56564] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The systemic administration of protective agents to treat drug-induced ototoxicity is limited by the possibility that these protective agents could interfere with the chemotherapeutic efficacy of the primary drugs. This is especially true for the drug cisplatin, whose anticancer actions are attenuated by antioxidants which provide adequate protection against hearing loss. Other current or potential otoprotective agents could pose a similar problem, if administered systemically. The application of various biologicals or protective agents directly to the cochlea would allow for high levels of these agents locally with limited systemic side effects. In this report, we demonstrate a trans-tympanic method of delivery of various drugs or biological reagents to the cochlea, which should enhance basic science research on the cochlea and provide a simple way of directing the use of otoprotective agents in the clinics. This report details a method of trans-tympanic drug delivery and provides examples of how this technique has been used successfully in experimental animals to treat cisplatin ototoxicity.
Collapse
Affiliation(s)
- Kelly Sheehan
- Department of Surgery, Division of Otolaryngology, Southern Illinois University School of Medicine
| | - Sandeep Sheth
- Department of Pharmacology, Southern Illinois University School of Medicine
| | - Debashree Mukherjea
- Department of Surgery, Division of Otolaryngology, Southern Illinois University School of Medicine
| | - Leonard P Rybak
- Department of Surgery, Division of Otolaryngology, Southern Illinois University School of Medicine; Department of Pharmacology, Southern Illinois University School of Medicine
| | - Vickram Ramkumar
- Department of Pharmacology, Southern Illinois University School of Medicine;
| |
Collapse
|
46
|
Cai R, Montgomery SC, Graves KA, Caspary DM, Cox BC. The FBN rat model of aging: investigation of ABR waveforms and ribbon synapse changes. Neurobiol Aging 2018; 62:53-63. [PMID: 29107847 PMCID: PMC5743589 DOI: 10.1016/j.neurobiolaging.2017.09.034] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 09/22/2017] [Accepted: 09/30/2017] [Indexed: 01/25/2023]
Abstract
Age-related hearing loss is experienced by one-third of individuals aged 65 years and older and can be socially debilitating. Historically, there has been poor correlation between age-related threshold changes, loss of speech understanding, and loss of cochlear hair cells. We examined changes in ribbon synapse number at four different ages in Fisher Brown Norway rats, an extensively studied rat model of aging. In contrast to previous work in mice/Wistar rats, we found minimal ribbon synapse loss before 20 months, with significant differences in 24- and 28-month-old rats at 4 kHz. Significant outer HC loss was observed at 24 and 28 months in low- to mid-frequency regions. Age-related reductions in auditory brainstem response wave I amplitude and increases in threshold were strongly correlated with ribbon synapse loss. Wave V/I ratios increased across age for click, 2, 4, and 24 kHz. Together, we find that ribbon synapses in the Fisher Brown Norway rat cochlea show resistance to aging until ∼60% of their life span, suggesting species/strain differences may underpin decreased peripheral input into the aging central processor.
Collapse
Affiliation(s)
- Rui Cai
- Division of Otolaryngology, Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Scott C Montgomery
- Division of Otolaryngology, Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA; Division of Otolaryngology, Department of Surgery, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kaley A Graves
- Division of Otolaryngology, Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Donald M Caspary
- Division of Otolaryngology, Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA; Division of Otolaryngology, Department of Surgery, Southern Illinois University School of Medicine, Springfield, IL, USA.
| | - Brandon C Cox
- Division of Otolaryngology, Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA; Division of Otolaryngology, Department of Surgery, Southern Illinois University School of Medicine, Springfield, IL, USA.
| |
Collapse
|
47
|
Bardot E, Tzavaras N, Benson DL, Dubois NC. Quantitative Whole-mount Immunofluorescence Analysis of Cardiac Progenitor Populations in Mouse Embryos. J Vis Exp 2017:10.3791/56446. [PMID: 29053691 PMCID: PMC6475905 DOI: 10.3791/56446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The use of ever-advancing imaging techniques has contributed broadly to our increased understanding of embryonic development. Pre-implantation development and organogenesis are two areas of research that have benefitted greatly from these advances, due to the high quality of data that can be obtained directly from imaging pre-implantation embryos or ex vivo organs. While pre-implantation embryos have yielded data with especially high spatial resolution, later stages have been less amenable to three-dimensional reconstruction. Obtaining high-quality 3D or volumetric data for known embryonic structures in combination with fate mapping or genetic lineage tracing will allow for a more comprehensive analysis of the morphogenetic events taking place during embryogenesis. This protocol describes a whole-mount immunofluorescence approach that allows for the labeling, visualization, and quantification of progenitor cell populations within the developing cardiac crescent, a key structure formed during heart development. The approach is designed in such a way that both cell- and tissue-level information can be obtained. Using confocal microscopy and image processing, this protocol allows for three-dimensional spatial reconstruction of the cardiac crescent, thereby providing the ability to analyze the localization and organization of specific progenitor populations during this critical phase of heart development. Importantly, the use of reference antibodies allows for successive masking of the cardiac crescent and subsequent quantitative measurements of areas within the crescent. This protocol will not only enable a detailed examination of early heart development, but with adaptations should be applicable to most organ systems in the gastrula to early somite stage mouse embryo.
Collapse
Affiliation(s)
- Evan Bardot
- Cell, Developmental, and Regenerative Biology Department, Icahn School of Medicine at Mount Sinai; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai
| | - Nikos Tzavaras
- Microscopy Core, Icahn School of Medicine at Mount Sinai
| | - Deanna L Benson
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai
| | - Nicole C Dubois
- Cell, Developmental, and Regenerative Biology Department, Icahn School of Medicine at Mount Sinai; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai;
| |
Collapse
|
48
|
McGovern MM, Brancheck J, Grant AC, Graves KA, Cox BC. Quantitative Analysis of Supporting Cell Subtype Labeling Among CreER Lines in the Neonatal Mouse Cochlea. J Assoc Res Otolaryngol 2016; 18:227-245. [PMID: 27873085 DOI: 10.1007/s10162-016-0598-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 10/17/2016] [Indexed: 11/30/2022] Open
Abstract
Four CreER lines that are commonly used in the auditory field to label cochlear supporting cells (SCs) are expressed in multiple SC subtypes, with some lines also showing reporter expression in hair cells (HCs). We hypothesized that altering the tamoxifen dose would modify CreER expression and target subsets of SCs. We also used two different reporter lines, ROSA26 tdTomato and CAG-eGFP, to achieve the same goal. Our results confirm previous reports that Sox2 CreERT2 and Fgfr3-iCreER T2 are not only expressed in neonatal SCs but also in HCs. Decreasing the tamoxifen dose did not reduce HC expression for Sox2 CreERT2 , but changing to the CAG-eGFP reporter decreased reporter-positive HCs sevenfold. However, there was also a significant decrease in the number of reporter-positive SCs. In contrast, there was a large reduction in reporter-positive HCs in Fgfr3-iCreER T2 mice with the lowest tamoxifen dose tested yet only limited reduction in SC labeling. The targeting of reporter expression to inner phalangeal and border cells was increased when Plp-CreER T2 was paired with the CAG-eGFP reporter; however, the total number of labeled cells decreased. Changes to the tamoxifen dose or reporter line with Prox1 CreERT2 caused minimal changes. Our data demonstrate that modifications to the tamoxifen dose or the use of different reporter lines may be successful in narrowing the numbers and/or types of cells labeled, but each CreER line responded differently. When the ROSA26 tdTomato reporter was combined with any of the four CreER lines, there was no difference in the number of tdTomato-positive cells after one or two injections of tamoxifen given at birth. Thus, tamoxifen-mediated toxicity could be reduced by only giving one injection. While the CAG-eGFP reporter consistently labeled fewer cells, both reporter lines are valuable depending on the goal of the study.
Collapse
Affiliation(s)
- Melissa M McGovern
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, 62711, USA
| | - Joseph Brancheck
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, 62711, USA
| | - Auston C Grant
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, 62711, USA
| | - Kaley A Graves
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, 62711, USA
| | - Brandon C Cox
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, 62711, USA.
- Department of Surgery, Division of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, 62711, USA.
| |
Collapse
|