1
|
Alvarado JC, Fuentes-Santamaría V, Benítez-Maicán Z, Díaz García CM, Gabaldón Ull MC, Juiz JM. An electrophysiological early marker of age-related hearing loss in the Wistar rat model. Heliyon 2024; 10:e40314. [PMID: 39584077 PMCID: PMC11585878 DOI: 10.1016/j.heliyon.2024.e40314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/26/2024] Open
Abstract
The goal of the present study was to determine, through a detailed study of the auditory brainstem response (ABR) waves, the possible existence of an early functional marker for the onset of presbycusis in an animal model. Toward this goal, Wistar rats were divided into four age groups: 3-month-old (3M, n = 6, control), 9-month-old (9M, n = 6), 14-month-old (14M, n = 6), and 20-month-old (20M, n = 6). ABR recordings were performed at 0.5, 1, 2, 4, 8, 16, and 32 kHz. The novel result reported here is that wave amplitudes, particularly wave II, were significantly diminished in the 9M group, even though there was no evidence of significant age-related threshold shift at that age. A significant increase in auditory thresholds with age was first detected at 14M, which further progressed at 20M, confirming our previous findings. These findings suggest that measurable alterations in ABR waves may precede age-related threshold shift and could serve as early markers to detect the onset of age-related hearing loss. Upon translation to humans, they could be used to implement early objective diagnosis, crucial to prevent or mitigate the negative consequences of presbycusis, a common, progressive, and irreversible neurodegenerative age-related disorder. This may allow, for instance, a better preservation of residual hearing, thus delaying the progression of the disease and minimizing the impact of hearing loss, ultimately improving the quality of life for those who suffer from this neurodegenerative condition.
Collapse
Affiliation(s)
- Juan Carlos Alvarado
- Facultad de Medicina e Instituto de Biomedicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | | | - Zaskya Benítez-Maicán
- Facultad de Medicina e Instituto de Biomedicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | | | - María Cruz Gabaldón Ull
- Facultad de Medicina e Instituto de Biomedicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - José M. Juiz
- Facultad de Medicina e Instituto de Biomedicina, Universidad de Castilla-La Mancha, Albacete, Spain
| |
Collapse
|
2
|
Udagawa T, Takahashi E, Tatsumi N, Mutai H, Saijo H, Kondo Y, Atkinson PJ, Matsunaga T, Yoshikawa M, Kojima H, Okabe M, Cheng AG. Loss of Pax3 causes reduction of melanocytes in the developing mouse cochlea. Sci Rep 2024; 14:2210. [PMID: 38278860 PMCID: PMC10817906 DOI: 10.1038/s41598-024-52629-9] [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: 05/28/2023] [Accepted: 01/22/2024] [Indexed: 01/28/2024] Open
Abstract
Cochlear melanocytes are intermediate cells in the stria vascularis that generate endocochlear potentials required for auditory function. Human PAX3 mutations cause Waardenburg syndrome and abnormalities of skin and retinal melanocytes, manifested as congenital hearing loss (~ 70%) and hypopigmentation of skin, hair and eyes. However, the underlying mechanism of hearing loss remains unclear. Cochlear melanocytes in the stria vascularis originated from Pax3-traced melanoblasts and Plp1-traced Schwann cell precursors, both of which derive from neural crest cells. Here, using a Pax3-Cre knock-in mouse that allows lineage tracing of Pax3-expressing cells and disruption of Pax3, we found that Pax3 deficiency causes foreshortened cochlea, malformed vestibular apparatus, and neural tube defects. Lineage tracing and in situ hybridization show that Pax3+ derivatives contribute to S100+, Kir4.1+ and Dct+ melanocytes (intermediate cells) in the developing stria vascularis, all of which are significantly diminished in Pax3 mutant animals. Taken together, these results suggest that Pax3 is required for the development of neural crest cell-derived cochlear melanocytes, whose absence may contribute to congenital hearing loss of Waardenburg syndrome in humans.
Collapse
Affiliation(s)
- Tomokatsu Udagawa
- Department of Otorhinolaryngology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan.
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan.
- Department of Otorhinolaryngology, Toho University School of Medicine, Tokyo, Japan.
| | - Erisa Takahashi
- Department of Otorhinolaryngology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| | - Norifumi Tatsumi
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| | - Hideki Mutai
- Division Hearing and Balance Research, National Institute of Sensory Organs, NHO Tokyo Medical Center, Tokyo, Japan
| | - Hiroki Saijo
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| | - Yuko Kondo
- Department of Otorhinolaryngology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Patrick J Atkinson
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Tatsuo Matsunaga
- Division Hearing and Balance Research, National Institute of Sensory Organs, NHO Tokyo Medical Center, Tokyo, Japan
| | - Mamoru Yoshikawa
- Department of Otorhinolaryngology, Toho University School of Medicine, Tokyo, Japan
| | - Hiromi Kojima
- Department of Otorhinolaryngology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Masataka Okabe
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| | - Alan G Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
3
|
Pressé MT, Malgrange B, Delacroix L. The cochlear matrisome: Importance in hearing and deafness. Matrix Biol 2024; 125:40-58. [PMID: 38070832 DOI: 10.1016/j.matbio.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/20/2023] [Accepted: 12/06/2023] [Indexed: 02/12/2024]
Abstract
The extracellular matrix (ECM) consists in a complex meshwork of collagens, glycoproteins, and proteoglycans, which serves a scaffolding function and provides viscoelastic properties to the tissues. ECM acts as a biomechanical support, and actively participates in cell signaling to induce tissular changes in response to environmental forces and soluble cues. Given the remarkable complexity of the inner ear architecture, its exquisite structure-function relationship, and the importance of vibration-induced stimulation of its sensory cells, ECM is instrumental to hearing. Many factors of the matrisome are involved in cochlea development, function and maintenance, as evidenced by the variety of ECM proteins associated with hereditary deafness. This review describes the structural and functional ECM components in the auditory organ and how they are modulated over time and following injury.
Collapse
Affiliation(s)
- Mary T Pressé
- Developmental Neurobiology Unit, GIGA-Neurosciences, University of Liège, 15 avenue Hippocrate - CHU - B36 (1st floor), Liège B-4000, Belgium
| | - Brigitte Malgrange
- Developmental Neurobiology Unit, GIGA-Neurosciences, University of Liège, 15 avenue Hippocrate - CHU - B36 (1st floor), Liège B-4000, Belgium
| | - Laurence Delacroix
- Developmental Neurobiology Unit, GIGA-Neurosciences, University of Liège, 15 avenue Hippocrate - CHU - B36 (1st floor), Liège B-4000, Belgium.
| |
Collapse
|
4
|
Rose KP, Manilla G, Milon B, Zalzman O, Song Y, Coate TM, Hertzano R. Spatially distinct otic mesenchyme cells show molecular and functional heterogeneity patterns before hearing onset. iScience 2023; 26:107769. [PMID: 37720106 PMCID: PMC10502415 DOI: 10.1016/j.isci.2023.107769] [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: 12/16/2022] [Revised: 06/29/2023] [Accepted: 08/25/2023] [Indexed: 09/19/2023] Open
Abstract
The cochlea consists of diverse cellular populations working in harmony to convert mechanical stimuli into electrical signals for the perception of sound. Otic mesenchyme cells (OMCs), often considered a homogeneous cell type, are essential for normal cochlear development and hearing. Despite being the most numerous cell type in the developing cochlea, OMCs are poorly understood. OMCs are known to differentiate into spatially and functionally distinct cell types, including fibrocytes of the lateral wall and spiral limbus, modiolar osteoblasts, and specialized tympanic border cells of the basilar membrane. Here, we show that OMCs are transcriptionally and functionally heterogeneous and can be divided into four distinct populations that spatially correspond to OMC-derived cochlear structures. We also show that this heterogeneity and complexity of OMCs commences during early phases of cochlear development. Finally, we describe the cell-cell communication network of the developing cochlea, inferring a major role for OMC in outgoing signaling.
Collapse
Affiliation(s)
- Kevin P. Rose
- Neurotology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gabriella Manilla
- Neurotology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Beatrice Milon
- Neurotology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ori Zalzman
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yang Song
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Thomas M. Coate
- Department of Biology, Georgetown University, Washington, DC 20007, USA
| | - Ronna Hertzano
- Neurotology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Otorhinolaryngology Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
5
|
Udagawa T, Takahashi E, Tatsumi N, Mutai H, Kondo Y, Atkinson PJ, Matsunaga T, Yoshikawa M, Kojima H, Okabe M, Cheng AG. Pax3 deficiency diminishes melanocytes in the developing mouse cochlea. RESEARCH SQUARE 2023:rs.3.rs-2990436. [PMID: 37333245 PMCID: PMC10274955 DOI: 10.21203/rs.3.rs-2990436/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Cochlear melanocytes are intermediate cells in the stria vascularis that generate endocochlear potentials required for auditory function. Human PAX3 mutations cause Waardenburg syndrome and abnormalities of melanocytes, manifested as congenital hearing loss and hypopigmentation of skin, hair and eyes. However, the underlying mechanism of hearing loss remains unclear. During development, cochlear melanocytes in the stria vascularis are dually derived from Pax3-Cre+ melanoblasts migrating from neuroepithelial cells including neural crest cells and Plp1+ Schwann cell precursors originated from also neural crest cells, differentiating in a basal-apical manner. Here, using a Pax3-Cre mouse line, we found that Pax3 deficiency causes foreshortened cochlea, malformed vestibular apparatus, and neural tube defects. Lineage tracing and in situ hybridization show that Pax3-Cre derivatives contribute to S100+ , Kir4.1+ and Dct+ melanocytes (intermediate cells) in the developing stria vascularis, all significantly diminished in Pax3 mutant animals. Taken together, these results suggest that Pax3 is required for the development of neural crest cell-derived cochlear melanocytes, whose absence may contribute to congenital hearing loss of Waardenburg syndrome in human.
Collapse
|
6
|
Ebeid M, Barnas K, Zhang H, Yaghmour A, Noreikaite G, Bjork BC. PRDM16 expression and function in mammalian cochlear development. Dev Dyn 2022; 251:1666-1683. [PMID: 35451126 PMCID: PMC9790675 DOI: 10.1002/dvdy.480] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/08/2022] [Accepted: 04/09/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND PR domain containing 16 (PRDM16) is a key transcriptional regulator in the development of craniofacial, adipose, and neural tissues. Our lab identified PRDM16 expression in the epithelial cells of the Kölliker's organ (KO) that starts at ~E13.5 and is maintained until KO disappearance. A transgenic mouse model that carries a gene trap null allele of Prdm16 (Prdm16cGT ) was used to characterize the impact of Prdm16 loss on cochlear development. RESULTS At P0 Prdm16cGT null cochlea exhibited hypoplastic KO, shortened cochlear duct, increased density of hair cells (HCs) and supporting cells (SCs) in the apical turn as well as multiple isolated ectopic HCs within the KO domain. KO epithelial cells proliferation rate was reduced in the apical turn of the developing Prdm16cGT null cochlea vs controls. Bulk RNA sequencing of cochlear duct cells at E14.5 followed by quantitative real time PCR and mRNA Fluorescence in-situ hybridization (FISH) validation identified differentially expressed genes in Prdm16cGT null vs littermate control cochleae. Upregulated genes at E14.5 included Fgf20, as well as several Notch pathway genes (Lfng, Hes1, and Jag1). CONCLUSIONS This study characterizes Prdm16 expression during cochlear development and establishes its requirement for KO development.
Collapse
Affiliation(s)
- Michael Ebeid
- College of Graduate Studies, Midwestern UniversityDowners GroveIllinoisUSA,Department of AnatomyMidwestern UniversityDowners GroveIllinoisUSA,Chicago College of Osteopathic MedicineMidwestern UniversityDowners GroveIllinoisUSA
| | - Kathy Barnas
- Biomedical Sciences ProgramMidwestern UniversityDowners GroveIllinoisUSA
| | - Hongji Zhang
- Department of AnatomyMidwestern UniversityDowners GroveIllinoisUSA
| | - Amal Yaghmour
- Biomedical Sciences ProgramMidwestern UniversityDowners GroveIllinoisUSA
| | - Gabriele Noreikaite
- Chicago College of Osteopathic MedicineMidwestern UniversityDowners GroveIllinoisUSA
| | - Bryan C. Bjork
- College of Graduate Studies, Midwestern UniversityDowners GroveIllinoisUSA
| |
Collapse
|
7
|
Petitpré C, Faure L, Uhl P, Fontanet P, Filova I, Pavlinkova G, Adameyko I, Hadjab S, Lallemend F. Single-cell RNA-sequencing analysis of the developing mouse inner ear identifies molecular logic of auditory neuron diversification. Nat Commun 2022; 13:3878. [PMID: 35790771 PMCID: PMC9256748 DOI: 10.1038/s41467-022-31580-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 06/21/2022] [Indexed: 11/08/2022] Open
Abstract
Different types of spiral ganglion neurons (SGNs) are essential for auditory perception by transmitting complex auditory information from hair cells (HCs) to the brain. Here, we use deep, single cell transcriptomics to study the molecular mechanisms that govern their identity and organization in mice. We identify a core set of temporally patterned genes and gene regulatory networks that may contribute to the diversification of SGNs through sequential binary decisions and demonstrate a role for NEUROD1 in driving specification of a Ic-SGN phenotype. We also find that each trajectory of the decision tree is defined by initial co-expression of alternative subtype molecular controls followed by gradual shifts toward cell fate resolution. Finally, analysis of both developing SGN and HC types reveals cell-cell signaling potentially playing a role in the differentiation of SGNs. Our results indicate that SGN identities are drafted prior to birth and reveal molecular principles that shape their differentiation and will facilitate studies of their development, physiology, and dysfunction.
Collapse
Affiliation(s)
- Charles Petitpré
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Louis Faure
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, 1090, Vienna, Austria
| | - Phoebe Uhl
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Paula Fontanet
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Iva Filova
- Institute of Biotechnology CAS, 25250, Vestec, Czech Republic
| | | | - Igor Adameyko
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, 1090, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Saida Hadjab
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Francois Lallemend
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
- Ming-Wai Lau Centre for Reparative Medicine, Stockholm Node, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
8
|
Fuentes-Santamaría V, Alvarado JC, Mellado S, Melgar-Rojas P, Gabaldón-Ull MC, Cabanes-Sanchis JJ, Juiz JM. Age-Related Inflammation and Oxidative Stress in the Cochlea Are Exacerbated by Long-Term, Short-Duration Noise Stimulation. Front Aging Neurosci 2022; 14:853320. [PMID: 35450058 PMCID: PMC9016828 DOI: 10.3389/fnagi.2022.853320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/14/2022] [Indexed: 12/29/2022] Open
Abstract
We have previously reported that young adult rats exposed to daily, short-duration noise for extended time periods, develop accelerated presbycusis starting at 6 months of age. Auditory aging is associated with progressive hearing loss, cell deterioration, dysregulation of the antioxidant defense system, and chronic inflammation, among others. To further characterize cellular and molecular mechanisms at the crossroads between noise and age-related hearing loss (ARHL), 3-month-old rats were exposed to a noise-accelerated presbycusis (NAP) protocol and tested at 6 and 16 months of age, using auditory brainstem responses, Real-Time Reverse Transcription-Quantitative PCR (RT-qPCR) and immunocytochemistry. Chronic noise-exposure leading to permanent auditory threshold shifts in 6-month-old rats, resulted in impaired sodium/potassium activity, degenerative changes in the lateral wall and spiral ganglion, increased lipid peroxidation, and sustained cochlear inflammation with advancing age. Additionally, at 6 months, noise-exposed rats showed significant increases in the gene expression of antioxidant enzymes (superoxide dismutase 1/2, glutathione peroxidase 1, and catalase) and inflammation-associated molecules [ionized calcium binding adaptor molecule 1, interleukin-1 beta (IL-1β), and tumor necrosis factor-alpha]. The levels of IL-1β were upregulated in the spiral ganglion and spiral ligament, particularly in type IV fibrocytes; these cells showed decreased levels of connective tissue growth factor and increased levels of 4-hydroxynonenal. These data provide functional, structural and molecular evidence that age-noise interaction contributes to exacerbating presbycusis in young rats by leading to progressive dysfunction and early degeneration of cochlear cells and structures. These findings contribute to a better understanding of NAP etiopathogenesis, which is essential as it affects the life quality of young adults worldwide.
Collapse
Affiliation(s)
- Verónica Fuentes-Santamaría
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Albacete, Spain
- Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Juan Carlos Alvarado
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Albacete, Spain
- Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Susana Mellado
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Albacete, Spain
- Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Pedro Melgar-Rojas
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Albacete, Spain
- Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - María Cruz Gabaldón-Ull
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Albacete, Spain
- Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - José J. Cabanes-Sanchis
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Albacete, Spain
- Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - José M. Juiz
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Albacete, Spain
- Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
- Department of Otolaryngology, Hannover Medical School, NIFE-VIANNA, Cluster of Excellence Hearing4all-German Research Foundation, Hanover, Germany
| |
Collapse
|
9
|
The effects of substrate composition and topography on the characteristics and growth of cell cultures of cochlear fibrocytes. Hear Res 2021; 415:108427. [PMID: 34999290 DOI: 10.1016/j.heares.2021.108427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 12/02/2021] [Accepted: 12/23/2021] [Indexed: 11/22/2022]
Abstract
Spiral ligament fibrocytes of the cochlea play homoeostatic roles in hearing and their degeneration contributes to hearing loss. Culturing fibrocytes in vitro provides a way to evaluate their functional characteristics and study possible therapies for hearing loss. We investigated whether in vivo characteristics of fibrocytes could be recapitulated in vitro by modifying the culture substrates and carried out proof of concept studies for potential transplantation of culture cells into the inner ear. Fibrocytes cultured from 4 to 5-week old CD/1 mice were grown on 2D substrates coated with collagen I, II, V or IX and, after harvesting, onto or into 3D substrates (hydrogels) of collagen I alone or mixed collagen I and II at a 1:1 ratio. We also assessed magnetic nanoparticle (MNP) uptake. Cell counts, immunohistochemical and ultrastructural studies showed that fibrocytes grown on 2D substrates proliferated, formed both small spindle-shaped and large flat cells that avidly took up MNPs. Of the different collagen coatings, only collagen II had an effect, causing a reduced size of the larger cells. On hydrogels, the cells were plump/rounded with extended processes, resembling native cells. They formed networks over the surface and became incorporated into the gel. In all culture formats, the majority co-expressed caldesmon, aquaporin 1, S-100 and sodium potassium ATPase, indicating a mixed or uncharacterised phenotype. Time-course experiments showed a decrease to ∼50% of the starting population by 4d after seeding on collagen I hydrogels, but better survival (∼60%) was found on collagen I + II gels, whilst TEM revealed the presence of apoptotic cells. Cells grown within gels additionally showed necrosis. These results demonstrate that fibrocytes grown in 3D recapitulate in vivo morphology of native fibrocytes, but have poorer survival, compared with 2D. Therefore hydrogel cultures could be used to study fibrocyte function and might also offer avenues for cell-replacement therapies, but need more optimization for therapeutic use. Fibrocyte function could be modified using MNPs in combination, for example, with gene transfection.
Collapse
|
10
|
Proteomic analysis identifies ZMYM2 as endogenous binding partner of TBX18 protein in 293 and A549 cells. Biochem J 2021; 479:91-109. [PMID: 34935912 DOI: 10.1042/bcj20210642] [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: 09/02/2021] [Revised: 12/15/2021] [Accepted: 12/21/2021] [Indexed: 11/17/2022]
Abstract
The TBX18 transcription factor regulates patterning and differentiation programs in the primordia of many organs yet the molecular complexes in which TBX18 resides to exert its crucial transcriptional function in these embryonic contexts have remained elusive. Here, we used 293 and A549 cells as an accessible cell source to search for endogenous protein interaction partners of TBX18 by an unbiased proteomic approach. We tagged endogenous TBX18 by CRISPR/Cas9 targeted genome editing with a triple FLAG peptide, and identified by anti-FLAG affinity purification and subsequent LC-MS analysis the ZMYM2 protein to be statistically enriched together with TBX18 in both 293 and A549 nuclear extracts. Using a variety of assays, we confirmed binding of TBX18 to ZMYM2, a component of the CoREST transcriptional corepressor complex. Tbx18 is coexpressed with Zmym2 in the mesenchymal compartment of the developing ureter of the mouse, and mutations in TBX18and in ZMYM2 were recently linked to congenital anomalies in the kidney and urinary tract (CAKUT) in line with a possible in vivo relevance of TBX18-ZMYM2 protein interaction in ureter development.
Collapse
|
11
|
Ng L, Liu Y, Liu H, Forrest D. Cochlear Fibrocyte and Osteoblast Lineages Expressing Type 2 Deiodinase Identified with a Dio2CreERt2 Allele. Endocrinology 2021; 162:bqab179. [PMID: 34436572 PMCID: PMC8475715 DOI: 10.1210/endocr/bqab179] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Indexed: 12/16/2022]
Abstract
Type 2 deiodinase (Dio2) amplifies levels of 3,5,3'-L-triiodothyronine (T3), the active form of thyroid hormone, and is essential for cochlear maturation and auditory development. However, cellular routes for endocrine signaling in the compartmentalized, anatomically complex cochlea are little understood. Dio2 generates T3 from thyroxine (T4), a more abundant thyroid hormone precursor in the circulation, and is dramatically induced in the cochlea before the onset of hearing. The evidence implies that specific Dio2-expressing cell types critically mediate T3 signaling but these cell types are poorly defined because Dio2 is expressed transiently at low levels. Here, using a Dio2CreERt2 knockin that activates a fluorescent reporter, we define Dio2-expressing cochlear cell types at high resolution in male or female mice. Dio2-positive cells were detected in vascularized supporting tissues but not in avascular internal epithelia, indicating segregation of T3-generating and T3-responding tissues. In the spiral ligament and spiral limbus, Dio2-positive fibrocytes clustered around vascular networks that convey T4 into cochlear tissues. In the otic capsule, Dio2-positive osteoblasts localized at cartilage surfaces as the bony labyrinth matures. We corroborated the identities of Dio2-positive lineages by RNA-sequencing of individual cells. The results suggest a previously unrecognized role for fibrocytes in mediating hormonal signaling. We discuss a model whereby fibrocytes mediate paracrine-like control of T3 signaling to the organ of Corti and epithelial target tissues.
Collapse
Affiliation(s)
- Lily Ng
- Laboratory of Endocrinology and Receptor Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ye Liu
- Laboratory of Endocrinology and Receptor Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hong Liu
- Laboratory of Endocrinology and Receptor Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Douglas Forrest
- Laboratory of Endocrinology and Receptor Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
12
|
Jan TA, Eltawil Y, Ling AH, Chen L, Ellwanger DC, Heller S, Cheng AG. Spatiotemporal dynamics of inner ear sensory and non-sensory cells revealed by single-cell transcriptomics. Cell Rep 2021; 36:109358. [PMID: 34260939 PMCID: PMC8378666 DOI: 10.1016/j.celrep.2021.109358] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 12/25/2020] [Accepted: 06/17/2021] [Indexed: 11/28/2022] Open
Abstract
The utricle is a vestibular sensory organ that requires mechanosensitive hair cells to detect linear acceleration. In neonatal mice, new hair cells are derived from non-sensory supporting cells, yet cell type diversity and mechanisms of cell addition remain poorly characterized. Here, we perform computational analyses on single-cell transcriptomes to categorize cell types and resolve 14 individual sensory and non-sensory subtypes. Along the periphery of the sensory epithelium, we uncover distinct groups of transitional epithelial cells, marked by Islr, Cnmd, and Enpep expression. By reconstructing de novo trajectories and gene dynamics, we show that as the utricle expands, Islr+ transitional epithelial cells exhibit a dynamic and proliferative phase to generate new supporting cells, followed by coordinated differentiation into hair cells. Taken together, our study reveals a sequential and coordinated process by which non-sensory epithelial cells contribute to growth of the postnatal mouse sensory epithelium. The postnatal mouse utricle expands by more than 35% and doubles its number of hair cells during the first 8 days. Using single-cell transcriptomics, Jan et al. show that the surrounding transitional epithelial cells proliferate and contribute to the expansion of the sensory epithelium through a stepwise differentiation mechanism.
Collapse
Affiliation(s)
- Taha A Jan
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco, San Francisco, CA 94115, USA
| | - Yasmin Eltawil
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Angela H Ling
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco, San Francisco, CA 94115, USA
| | - Leon Chen
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Daniel C Ellwanger
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Genome Analysis Unit, Amgen Research, Amgen Inc., South San Francisco, CA 94080, USA
| | - Stefan Heller
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA.
| | - Alan G Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA.
| |
Collapse
|
13
|
Wang S, Lee MP, Jones S, Liu J, Waldhaus J. Mapping the regulatory landscape of auditory hair cells from single-cell multi-omics data. Genome Res 2021; 31:1885-1899. [PMID: 33837132 DOI: 10.1101/gr.271080.120] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 03/23/2021] [Indexed: 11/25/2022]
Abstract
Auditory hair cells transduce sound to the brain and in mammals these cells reside together with supporting cells in the sensory epithelium of the cochlea, called the organ of Corti. To establish the organ's delicate function during development and differentiation, spatiotemporal gene expression is strictly controlled by chromatin accessibility and cell type-specific transcription factors, jointly representing the regulatory landscape. Bulk-sequencing technology and cellular heterogeneity obscured investigations on the interplay between transcription factors and chromatin accessibility in inner ear development. To study the formation of the regulatory landscape in hair cells, we collected single-cell chromatin accessibility profiles accompanied by single-cell RNA data from genetically labeled murine hair cells and supporting cells after birth. Using an integrative approach, we predicted cell type-specific activating and repressing functions of developmental transcription factors. Furthermore, by integrating gene expression and chromatin accessibility datasets, we reconstructed gene regulatory networks. Then, using a comparative approach, 20 hair cell-specific activators and repressors, including putative downstream target genes, were identified. Clustering of target genes resolved groups of related transcription factors and was utilized to infer their developmental functions. Finally, the heterogeneity in the single-cell data allowed us to spatially reconstruct transcriptional as well as chromatin accessibility trajectories, indicating that gradual changes in the chromatin accessibility landscape were lagging behind the transcriptional identity of hair cells along the organ's longitudinal axis. Overall, this study provides a strategy to spatially reconstruct the formation of a lineage specific regulatory landscape using a single-cell multi-omics approach.
Collapse
Affiliation(s)
- Shuze Wang
- University of Michigan, Kresge Hearing Research Institute
| | - Mary P Lee
- University of Michigan, Kresge Hearing Research Institute
| | - Scott Jones
- University of Michigan, Kresge Hearing Research Institute
| | | | - Joerg Waldhaus
- University of Michigan, Kresge Hearing Research Institute;
| |
Collapse
|
14
|
van der Valk WH, Steinhart MR, Zhang J, Koehler KR. Building inner ears: recent advances and future challenges for in vitro organoid systems. Cell Death Differ 2020; 28:24-34. [PMID: 33318601 PMCID: PMC7853146 DOI: 10.1038/s41418-020-00678-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
While inner ear disorders are common, our ability to intervene and recover their sensory function is limited. In vitro models of the inner ear, like the organoid system, could aid in identifying new regenerative drugs and gene therapies. Here, we provide a perspective on the status of in vitro inner ear models and guidance on how to improve their applicability in translational research. We highlight the generation of inner ear cell types from pluripotent stem cells as a particularly promising focus of research. Several exciting recent studies have shown how the developmental signaling cues of embryonic and fetal development can be mimicked to differentiate stem cells into “inner ear organoids” containing otic progenitor cells, hair cells, and neurons. However, current differentiation protocols and our knowledge of embryonic and fetal inner ear development in general, have a bias toward the sensory epithelia of the inner ear. We propose that a more holistic view is needed to better model the inner ear in vitro. Moving forward, attention should be made to the broader diversity of neuroglial and mesenchymal cell types of the inner ear, and how they interact in space or time during development. With improved control of epithelial, neuroglial, and mesenchymal cell fate specification, inner ear organoids would have the ability to truly recapitulate neurosensory function and dysfunction. We conclude by discussing how single-cell atlases of the developing inner ear and technical innovations will be critical tools to advance inner ear organoid platforms for future pre-clinical applications.
Collapse
Affiliation(s)
- Wouter H van der Valk
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, Netherlands.,Department of Otolaryngology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Matthew R Steinhart
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jingyuan Zhang
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA, 02115, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA.,Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Karl R Koehler
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA, 02115, USA. .,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA, 02115, USA. .,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA. .,Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
15
|
Wang J, Lu C, Zhao Y, Tang Z, Song J, Fan C. Transcriptome profiles of sturgeon lateral line electroreceptor and mechanoreceptor during regeneration. BMC Genomics 2020; 21:875. [PMID: 33287707 PMCID: PMC7720607 DOI: 10.1186/s12864-020-07293-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/28/2020] [Indexed: 11/10/2022] Open
Abstract
Background The electrosensory ampullary organs (AOs) and mechanosensory neuromasts (NMs) found in sturgeon and some other non-neopterygian fish or amphibians are both originated from lateral line placodes. However, these two sensory organs have characteristic morphological and physiological differences. The molecular mechanisms for the specification of AOs and NMs are not clearly understood. Results We sequenced the transcriptome for neomycin treated sturgeon AOs and NMs in the early regeneration stages, and de novo assembled a sturgeon transcriptome. By comparing the gene expression differences among untreated AOs, NMs and general epithelia (EPs), we located some specific genes for these two sensory organs. In sturgeon lateral line, the voltage-gated calcium channels and voltage-gated potassium channels were predominant calcium and potassium channel subtypes, respectively. And by correlating gene expression with the regeneration process, we predicated several candidate key transcriptional regulation related genes might be involved in AOs and NMs regeneration. Conclusions Genes with specific expression in the two lateral line sensory organs suggests their important roles in mechanoreceptor and electroreceptor formation. The candidate transcriptional regulation related genes may be important for mechano- and electro- receptor specification, in a “dosage-related” manner. These results suggested the molecular basis for specification of these two sensory organs in sturgeon.
Collapse
Affiliation(s)
- Jian Wang
- International Joint Center for Marine Biological Sciences Research, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
| | - Chengcheng Lu
- International Joint Center for Marine Biological Sciences Research, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China.,Institute for Marine Biosystem and Neuroscience, International Center for Marine Studies, Shanghai Ocean University, Shanghai, China
| | - Yifan Zhao
- International Joint Center for Marine Biological Sciences Research, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China.,Institute for Marine Biosystem and Neuroscience, International Center for Marine Studies, Shanghai Ocean University, Shanghai, China
| | - Zhijiao Tang
- Institute for Marine Biosystem and Neuroscience, International Center for Marine Studies, Shanghai Ocean University, Shanghai, China
| | - Jiakun Song
- Institute for Marine Biosystem and Neuroscience, International Center for Marine Studies, Shanghai Ocean University, Shanghai, China
| | - Chunxin Fan
- International Joint Center for Marine Biological Sciences Research, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China. .,Institute for Marine Biosystem and Neuroscience, International Center for Marine Studies, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|
16
|
Alvarado JC, Fuentes-Santamaría V, Melgar-Rojas P, Gabaldón-Ull MC, Cabanes-Sanchis JJ, Juiz JM. Oral Antioxidant Vitamins and Magnesium Limit Noise-Induced Hearing Loss by Promoting Sensory Hair Cell Survival: Role of Antioxidant Enzymes and Apoptosis Genes. Antioxidants (Basel) 2020; 9:E1177. [PMID: 33255728 PMCID: PMC7761130 DOI: 10.3390/antiox9121177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 01/01/2023] Open
Abstract
Noise induces oxidative stress in the cochlea followed by sensory cell death and hearing loss. The proof of principle that injections of antioxidant vitamins and Mg2+ prevent noise-induced hearing loss (NIHL) has been established. However, effectiveness of oral administration remains controversial and otoprotection mechanisms are unclear. Using auditory evoked potentials, quantitative PCR, and immunocytochemistry, we explored effects of oral administration of vitamins A, C, E, and Mg2+ (ACEMg) on auditory function and sensory cell survival following NIHL in rats. Oral ACEMg reduced auditory thresholds shifts after NIHL. Improved auditory function correlated with increased survival of sensory outer hair cells. In parallel, oral ACEMg modulated the expression timeline of antioxidant enzymes in the cochlea after NIHL. There was increased expression of glutathione peroxidase-1 and catalase at 1 and 10 days, respectively. Also, pro-apoptotic caspase-3 and Bax levels were diminished in ACEMg-treated rats, at 10 and 30 days, respectively, following noise overstimulation, whereas, at day 10 after noise exposure, the levels of anti-apoptotic Bcl-2, were significantly increased. Therefore, oral ACEMg improves auditory function by limiting sensory hair cell death in the auditory receptor following NIHL. Regulation of the expression of antioxidant enzymes and apoptosis-related proteins in cochlear structures is involved in such an otoprotective mechanism.
Collapse
Affiliation(s)
- Juan C. Alvarado
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), School of Medicine, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (V.F.-S.); (P.M.-R.); (M.C.G.-U.); (J.J.C.-S.)
| | - Verónica Fuentes-Santamaría
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), School of Medicine, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (V.F.-S.); (P.M.-R.); (M.C.G.-U.); (J.J.C.-S.)
| | - Pedro Melgar-Rojas
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), School of Medicine, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (V.F.-S.); (P.M.-R.); (M.C.G.-U.); (J.J.C.-S.)
| | - María C. Gabaldón-Ull
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), School of Medicine, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (V.F.-S.); (P.M.-R.); (M.C.G.-U.); (J.J.C.-S.)
| | - José J. Cabanes-Sanchis
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), School of Medicine, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (V.F.-S.); (P.M.-R.); (M.C.G.-U.); (J.J.C.-S.)
| | - José M. Juiz
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), School of Medicine, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (V.F.-S.); (P.M.-R.); (M.C.G.-U.); (J.J.C.-S.)
- Department of Otolaryngology, Hannover Medical School, NIFE-VIANNA, Cluster of Excellence Hearing4all-German Research Foundation, 30625 Hannover, Germany
| |
Collapse
|
17
|
Cochlear Implantation in a Patient with a Novel POU3F4 Mutation and Incomplete Partition Type-III Malformation. Neural Plast 2020; 2020:8829587. [PMID: 32952548 PMCID: PMC7481964 DOI: 10.1155/2020/8829587] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 08/05/2020] [Accepted: 08/24/2020] [Indexed: 01/01/2023] Open
Abstract
Aims This study is aimed at (1) analyzing the clinical manifestations and genetic features of a novel POU3F4 mutation in a nonsyndromic X-linked recessive hearing loss family and (2) reporting the outcomes of cochlear implantation in a patient with this mutation. Methods A patient who was diagnosed as the IP-III malformation underwent cochlear implantation in our hospital. The genetic analysis was conducted in his family, including the whole-exome sequencing combined with Sanger sequencing and bioinformatic analysis. Clinical features, preoperative auditory and speech performances, and postoperative outcomes of cochlear implant (CI) were assessed on the proband and his family. Results A novel variant c.400_401insACTC (p.Q136LfsX58) in the POU3F4 gene was detected in the family, which was cosegregated with the hearing loss. This variant was absent in 200 normal-hearing persons. The phylogenetic analysis and structure modeling of Pou3f4 protein further confirmed that the novel mutation was pathogenic. The proband underwent cochlear implantation on the right ear at four years old and gained greatly auditory and speech improvement. However, the benefits of the CI declined about three and a half years postoperation. Though the right ear had been reimplanted, the outcomes were still worse than before. Conclusion A novel frame shift variant c.400_401insACTC (p.Q136LfsX58) in the POU3F4 gene was identified in a Chinese family with X-linked inheritance hearing loss. A patient with this mutation and IP-III malformation could get good benefits from CI. However, the outcomes of the cochlear implantation might decline as the patient grows old.
Collapse
|
18
|
Hosoya M, Fujioka M, Murayama AY, Okano H, Ogawa K. The common marmoset as suitable nonhuman alternative for the analysis of primate cochlear development. FEBS J 2020; 288:325-353. [PMID: 32323465 PMCID: PMC7818239 DOI: 10.1111/febs.15341] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/30/2020] [Accepted: 04/20/2020] [Indexed: 12/13/2022]
Abstract
Cochlear development is a complex process with precise spatiotemporal patterns. A detailed understanding of this process is important for studies of congenital hearing loss and regenerative medicine. However, much of our understanding of cochlear development is based on rodent models. Animal models that bridge the gap between humans and rodents are needed. In this study, we investigated the development of hearing organs in a small New World monkey species, the common marmoset (Callithrix jacchus). We describe the general stages of cochlear development in comparison with those of humans and mice. Moreover, we examined more than 25 proteins involved in cochlear development and found that expression patterns were generally conserved between rodents and primates. However, several proteins involved in supporting cell processes and neuronal development exhibited interspecific expression differences. Human fetal samples for studies of primate‐specific cochlear development are extremely rare, especially for late developmental stages. Our results support the use of the common marmoset as an effective alternative for analyses of primate cochlear development.
Collapse
Affiliation(s)
- Makoto Hosoya
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masato Fujioka
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Ayako Y Murayama
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.,Laboratory for Marmoset Neural Architecture, Center for Brain Science, RIKEN, Wako, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.,Laboratory for Marmoset Neural Architecture, Center for Brain Science, RIKEN, Wako, Japan
| | - Kaoru Ogawa
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
19
|
Cheng C, Wang Y, Guo L, Lu X, Zhu W, Muhammad W, Zhang L, Lu L, Gao J, Tang M, Chen F, Gao X, Li H, Chai R. Age-related transcriptome changes in Sox2+ supporting cells in the mouse cochlea. Stem Cell Res Ther 2019; 10:365. [PMID: 31791390 PMCID: PMC6889721 DOI: 10.1186/s13287-019-1437-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/29/2019] [Accepted: 10/01/2019] [Indexed: 12/17/2022] Open
Abstract
Background Inner ear supporting cells (SCs) in the neonatal mouse cochlea are a potential source for hair cell (HC) regeneration, but several studies have shown that the regeneration ability of SCs decreases dramatically as mice age and that lost HCs cannot be regenerated in adult mice. To better understand how SCs might be better used to regenerate HCs, it is important to understand how the gene expression profile changes in SCs at different ages. Methods Here, we used Sox2GFP/+ mice to isolate the Sox2+ SCs at postnatal day (P)3, P7, P14, and P30 via flow cytometry. Next, we used RNA-seq to determine the transcriptome expression profiles of P3, P7, P14, and P30 SCs. To further analyze the relationships between these age-related and differentially expressed genes in Sox2+ SCs, we performed gene ontology (GO) analysis. Results Consistent with previous reports, we also found that the proliferation and HC regeneration ability of isolated Sox2+ SCs significantly decreased as mice aged. We identified numerous genes that are enriched and differentially expressed in Sox2+ SCs at four different postnatal ages, including cell cycle genes, signaling pathway genes, and transcription factors that might be involved in regulating the proliferation and HC differentiation ability of SCs. We thus present a set of genes that might regulate the proliferation and HC regeneration ability of SCs, and these might serve as potential new therapeutic targets for HC regeneration. Conclusions In our research, we found several genes that might play an important role in regulating the proliferation and HC regeneration ability of SCs. These datasets are expected to serve as a resource to provide potential new therapeutic targets for regulating the ability of SCs to regenerate HCs in postnatal mammals.
Collapse
Affiliation(s)
- Cheng Cheng
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, China.,Research Institute of Otolaryngology, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Yunfeng Wang
- Shanghai Fenyang Vision & Audition Center, Shanghai, China.,ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Room 611, Building 9, No. 83, Fenyang Road, Xuhui District, Shanghai, 200031, China
| | - Luo Guo
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Room 611, Building 9, No. 83, Fenyang Road, Xuhui District, Shanghai, 200031, China
| | - Xiaoling Lu
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Room 611, Building 9, No. 83, Fenyang Road, Xuhui District, Shanghai, 200031, China
| | - Weijie Zhu
- MOE Key Laboratory for Developmental Genes and Human Disease, State Key Laboratory of Bioelectronics, Co-Innovation Center of Neuroregeneration, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Waqas Muhammad
- MOE Key Laboratory for Developmental Genes and Human Disease, State Key Laboratory of Bioelectronics, Co-Innovation Center of Neuroregeneration, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.,Department of Biotechnology, Federal Urdu University of Arts, Science and Technology, Gulshan-e-Iqbal Campus, Karachi, Pakistan
| | - Liyan Zhang
- MOE Key Laboratory for Developmental Genes and Human Disease, State Key Laboratory of Bioelectronics, Co-Innovation Center of Neuroregeneration, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Ling Lu
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Junyan Gao
- Jiangsu Rehabilitation Research Center for Hearing and Speech Impairment, Nanjing, 210004, Jiangsu, China
| | - Mingliang Tang
- MOE Key Laboratory for Developmental Genes and Human Disease, State Key Laboratory of Bioelectronics, Co-Innovation Center of Neuroregeneration, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Fangyi Chen
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Xia Gao
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, China.
| | - Huawei Li
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Room 611, Building 9, No. 83, Fenyang Road, Xuhui District, Shanghai, 200031, China.
| | - Renjie Chai
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Room 611, Building 9, No. 83, Fenyang Road, Xuhui District, Shanghai, 200031, China. .,MOE Key Laboratory for Developmental Genes and Human Disease, State Key Laboratory of Bioelectronics, Co-Innovation Center of Neuroregeneration, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China. .,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, China. .,Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
20
|
Miwa T, Minoda R, Ishikawa Y, Kajii T, Orita Y, Ohyama T. Role of Dach1 revealed using a novel inner ear-specific Dach1-knockdown mouse model. Biol Open 2019; 8:bio.043612. [PMID: 31405829 PMCID: PMC6737983 DOI: 10.1242/bio.043612] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Dach1 gene is expressed in the inner ear of normal mouse embryos in the area that differentiates into the cochlear stria vascularis (SV). We hypothesised that Dach1 downregulation in the inner ear would lead to SV dysplasia. However, because Dach1 knockout is embryonic lethal in mice, the role of Dach1 in the inner ear is unclear. Here, we established inner ear-specific Dach1-knockdown mice and showed that Dach1 downregulation resulted in hearing loss, reduced endocochlear potential and secondary outer hair cell loss. There were no abnormalities in marginal cells and basal cells in the SV or spiral ligament in inner ear-specific Dach1-knockdown mature mice. However, intermediate cell dysplasia and thinning of the SV were observed. Moreover, dynamic changes in the expression of key genes related to the epithelial-mesenchymal transition were observed in the lateral wall of the cochlear epithelium, which differentiated into the SV in inner ear-specific Dach1-knockdown mice at embryonic stages. In summary, suppression of Dach1 expression in the inner ear caused the epithelial-mesenchymal transition in the lateral wall of cochlear epithelium, resulting in loss of intermediate cells in the SV and SV dysplasia. This article has an associated First Person interview with the first author of the paper. Summary: Inner ear-specific downregulation of Dach1 reveals that the epithelial–mesenchymal transition is crucial for the generation of the stria vascularis.
Collapse
Affiliation(s)
- Toru Miwa
- Department of Otolaryngology and Head and Neck Surgery, Graduate of School of Medicine, Kumamoto University, Kumamoto 8608556, Japan .,Otolaryngology-Head and Neck Surgery, JCHO Kumamoto General Hospital, Kumamoto 8668660, Japan
| | - Ryosei Minoda
- Otolaryngology-Head and Neck Surgery, JCHO Kumamoto General Hospital, Kumamoto 8668660, Japan
| | - Yoshihide Ishikawa
- Department of Otolaryngology and Head and Neck Surgery, Graduate of School of Medicine, Kumamoto University, Kumamoto 8608556, Japan
| | - Tomohito Kajii
- Department of Otolaryngology and Head and Neck Surgery, Graduate of School of Medicine, Kumamoto University, Kumamoto 8608556, Japan
| | - Yorihisa Orita
- Department of Otolaryngology and Head and Neck Surgery, Graduate of School of Medicine, Kumamoto University, Kumamoto 8608556, Japan
| | - Takahiro Ohyama
- USC-Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Zilkha Neurogenetic Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
21
|
Open chromatin dynamics in prosensory cells of the embryonic mouse cochlea. Sci Rep 2019; 9:9060. [PMID: 31227770 PMCID: PMC6588700 DOI: 10.1038/s41598-019-45515-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 06/10/2019] [Indexed: 12/13/2022] Open
Abstract
Hearing loss is often due to the absence or the degeneration of hair cells in the cochlea. Understanding the mechanisms regulating the generation of hair cells may therefore lead to better treatments for hearing disorders. To elucidate the transcriptional control mechanisms specifying the progenitor cells (i.e. prosensory cells) that generate the hair cells and support cells critical for hearing function, we compared chromatin accessibility using ATAC-seq in sorted prosensory cells (Sox2-EGFP+) and surrounding cells (Sox2-EGFP−) from E12, E14.5 and E16 cochlear ducts. In Sox2-EGFP+, we find greater accessibility in and near genes restricted in expression to the prosensory region of the cochlear duct including Sox2, Isl1, Eya1 and Pou4f3. Furthermore, we find significant enrichment for the consensus binding sites of Sox2, Six1 and Gata3—transcription factors required for prosensory development—in the open chromatin regions. Over 2,200 regions displayed differential accessibility with developmental time in Sox2-EGFP+ cells, with most changes in the E12-14.5 window. Open chromatin regions detected in Sox2-EGFP+ cells map to over 48,000 orthologous regions in the human genome that include regions in genes linked to deafness. Our results reveal a dynamic landscape of open chromatin in prosensory cells with potential implications for cochlear development and disease.
Collapse
|
22
|
Alvarado JC, Fuentes-Santamaría V, Gabaldón-Ull MC, Juiz JM. Age-Related Hearing Loss Is Accelerated by Repeated Short-Duration Loud Sound Stimulation. Front Neurosci 2019; 13:77. [PMID: 30872984 PMCID: PMC6402475 DOI: 10.3389/fnins.2019.00077] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 01/24/2019] [Indexed: 12/18/2022] Open
Abstract
Both age-related hearing loss (ARHL) and noise-induced hearing loss (NIHL) may share pathophysiological mechanisms in that they are associated with excess free radical formation and cochlear blood flow reduction, leading to cochlear damage. Therefore, it is possible that short, but repeated exposures to relatively loud noise during extended time periods, like in leisure (i.e., musical devices and concerts) or occupational noise exposures, may add to cochlear aging mechanisms, having an impact on the onset and/or progression of ARHL. Consequently, the aim of the present study was to determine if repeated short-duration overexposure to a long-term noise could accelerate permanent auditory threshold shifts associated with auditory aging in an animal model of ARHL. Toward this goal, young adult, 3-month-old Wistar rats were divided into two groups: one exposed (E) and the other non-exposed (NE) to noise overstimulation. The stimulation protocol consisted of 1 h continuous white noise at 110 dB sound pressure level (SPL), 5 days a week, allowing 2 days for threshold recovery before initiating another stimulation round, until the animals reached an age of 18 months. Auditory brainstem response (ABR) recordings at 0.5, 1, 2, 4, 8, 16, and 32 kHz were performed at 3, 6, 12, and 18 months of age. The results demonstrate that in the E group there were significant increases in auditory thresholds at all tested frequencies starting already at 6 months of age, which extended at 12 and 18 months. However, in NE animals threshold shifts were not evident until 12 months, extending to 18 months of age. Threshold shifts observed in the E animals at 6 and 12 months were significantly larger than those observed in the NE group at the same ages. Threshold shifts at 6 and 12 months in E animals resembled those at 12 and 18 months in NE animals, respectively. This suggests that repeated noise overstimulation in short-duration episodes accelerates the time-course of hearing loss in this animal model of ARHL.
Collapse
Affiliation(s)
- Juan Carlos Alvarado
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Albacete, Spain Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Verónica Fuentes-Santamaría
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Albacete, Spain Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - María Cruz Gabaldón-Ull
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Albacete, Spain Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - José M Juiz
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Albacete, Spain Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| |
Collapse
|
23
|
Sonntag M, Blosa M, Schmidt S, Reimann K, Blum K, Eckrich T, Seeger G, Hecker D, Schick B, Arendt T, Engel J, Morawski M. Synaptic coupling of inner ear sensory cells is controlled by brevican-based extracellular matrix baskets resembling perineuronal nets. BMC Biol 2018; 16:99. [PMID: 30253762 PMCID: PMC6156866 DOI: 10.1186/s12915-018-0566-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/15/2018] [Indexed: 02/08/2023] Open
Abstract
Background Perineuronal nets (PNNs) are specialized aggregations of extracellular matrix (ECM) molecules surrounding specific neurons in the central nervous system (CNS). PNNs are supposed to control synaptic transmission and are frequently associated with neurons firing at high rates, including principal neurons of auditory brainstem nuclei. The origin of high-frequency activity of auditory brainstem neurons is the indefatigable sound-driven transmitter release of inner hair cells (IHCs) in the cochlea. Results Here, we show that synaptic poles of IHCs are ensheathed by basket-like ECM complexes formed by the same molecules that constitute PNNs of neurons in the CNS, including brevican, aggreccan, neurocan, hyaluronan, and proteoglycan link proteins 1 and 4 and tenascin-R. Genetic deletion of brevican, one of the main components, resulted in a massive degradation of ECM baskets at IHCs, a significant impairment in spatial coupling of pre- and postsynaptic elements and mild impairment of hearing. Conclusions These ECM baskets potentially contribute to control of synaptic transmission at IHCs and might be functionally related to PNNs of neurons in the CNS. Electronic supplementary material The online version of this article (10.1186/s12915-018-0566-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mandy Sonntag
- Paul-Flechsig-Institute of Brain Research, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Maren Blosa
- Paul-Flechsig-Institute of Brain Research, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Sophie Schmidt
- Paul-Flechsig-Institute of Brain Research, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Katja Reimann
- Paul-Flechsig-Institute of Brain Research, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Kerstin Blum
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Tobias Eckrich
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Gudrun Seeger
- Paul-Flechsig-Institute of Brain Research, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Dietmar Hecker
- Department of Otorhinolaryngology, School of Medicine, Saarland University, Homburg, Germany
| | - Bernhard Schick
- Department of Otorhinolaryngology, School of Medicine, Saarland University, Homburg, Germany
| | - Thomas Arendt
- Paul-Flechsig-Institute of Brain Research, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Jutta Engel
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Markus Morawski
- Paul-Flechsig-Institute of Brain Research, Medical Faculty, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
24
|
Rivera-Reyes R, Kleppa MJ, Kispert A. Proteomic analysis identifies transcriptional cofactors and homeobox transcription factors as TBX18 binding proteins. PLoS One 2018; 13:e0200964. [PMID: 30071041 PMCID: PMC6071992 DOI: 10.1371/journal.pone.0200964] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 05/30/2018] [Indexed: 01/04/2023] Open
Abstract
The TBX18 transcription factor is a crucial developmental regulator of several organ systems in mice, and loss of its transcriptional repression activity causes dilative nephropathies in humans. The molecular complexes with which TBX18 regulates transcription are poorly understood prompting us to use an unbiased proteomic approach to search for protein interaction partners. Using overexpressed dual tagged TBX18 as bait, we identified by tandem purification and subsequent LC-MS analysis TBX18 binding proteins in 293 cells. Clustering of functional annotations of the identified proteins revealed a highly significant enrichment of transcriptional cofactors and homeobox transcription factors. Using nuclear recruitment assays as well as GST pull-downs, we validated CBFB, GAR1, IKZF2, NCOA5, SBNO2 and CHD7 binding to the T-box of TBX18 in vitro. From these transcriptional cofactors, CBFB, CHD7 and IKZF2 enhanced the transcriptional repression of TBX18, while NCOA5 and SBNO2 dose-dependently relieved it. All tested homeobox transcription factors interacted with the T-box of TBX18 in pull-down assays, with members of the Pbx and Prrx subfamilies showing coexpression with Tbx18 in the developing ureter of the mouse. In summary, we identified and characterized new TBX18 binding partners that may influence the transcriptional activity of TBX18 in vivo.
Collapse
Affiliation(s)
| | - Marc-Jens Kleppa
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Andreas Kispert
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
- * E-mail:
| |
Collapse
|
25
|
Alvarado JC, Fuentes-Santamaría V, Gabaldón-Ull MC, Juiz JM. An Oral Combination of Vitamins A, C, E, and Mg ++ Improves Auditory Thresholds in Age-Related Hearing Loss. Front Neurosci 2018; 12:527. [PMID: 30108480 PMCID: PMC6079267 DOI: 10.3389/fnins.2018.00527] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/13/2018] [Indexed: 12/02/2022] Open
Abstract
The increasing rate of age-related hearing loss (ARHL), with its subsequent reduction in quality of life and increase in health care costs, requires new therapeutic strategies to reduce and delay its impact. The goal of this study was to determine if ARHL could be reduced in a rat model by administering a combination of antioxidant vitamins A, C, and E acting as free radical scavengers along with Mg++, a known powerful cochlear vasodilator (ACEMg). Toward this goal, young adult, 3 month-old Wistar rats were divided into two groups: one was fed with a diet composed of regular chow (“normal diet,” ND); the other received a diet based on chow enriched in ACEMg (“enhanced diet,” ED). The ED feeding began 10 days before the noise stimulation. Auditory brainstem recordings (ABR) were performed at 0.5, 1, 2, 4, 8, 16, and 32 kHz at 3, 6–8, and 12–14 months of age. No differences were observed at 3 months of age, in both ND and ED animals. At 6–8 and 12–14 months of age there were significant increases in auditory thresholds and a reduction in the wave amplitudes at all frequencies tested, compatible with progressive development of ARHL. However, at 6–8 months threshold shifts in ED rats were significantly lower in low and medium frequencies, and wave amplitudes were significantly larger at all frequencies when compared to ND rats. In the oldest animals, differences in the threshold shift persisted, as well as in the amplitude of the wave II, suggesting a protective effect of ACEMg on auditory function during aging. These findings indicate that oral ACEMg may provide an effective adjuvant therapeutic intervention for the treatment of ARHL, delaying the progression of hearing impairment associated with age.
Collapse
Affiliation(s)
- Juan C Alvarado
- Instituto de Investigación en Discapacidades Neurológicas, Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Verónica Fuentes-Santamaría
- Instituto de Investigación en Discapacidades Neurológicas, Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - María C Gabaldón-Ull
- Instituto de Investigación en Discapacidades Neurológicas, Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - José M Juiz
- Instituto de Investigación en Discapacidades Neurológicas, Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| |
Collapse
|
26
|
Espino Guarch M, Font-Llitjós M, Murillo-Cuesta S, Errasti-Murugarren E, Celaya AM, Girotto G, Vuckovic D, Mezzavilla M, Vilches C, Bodoy S, Sahún I, González L, Prat E, Zorzano A, Dierssen M, Varela-Nieto I, Gasparini P, Palacín M, Nunes V. Mutations in L-type amino acid transporter-2 support SLC7A8 as a novel gene involved in age-related hearing loss. eLife 2018; 7:31511. [PMID: 29355479 PMCID: PMC5811215 DOI: 10.7554/elife.31511] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 01/18/2018] [Indexed: 12/19/2022] Open
Abstract
Age-related hearing loss (ARHL) is the most common sensory deficit in the elderly. The disease has a multifactorial etiology with both environmental and genetic factors involved being largely unknown. SLC7A8/SLC3A2 heterodimer is a neutral amino acid exchanger. Here, we demonstrated that SLC7A8 is expressed in the mouse inner ear and that its ablation resulted in ARHL, due to the damage of different cochlear structures. These findings make SLC7A8 transporter a strong candidate for ARHL in humans. Thus, a screening of a cohort of ARHL patients and controls was carried out revealing several variants in SLC7A8, whose role was further investigated by in vitro functional studies. Significant decreases in SLC7A8 transport activity was detected for patient’s variants (p.Val302Ile, p.Arg418His, p.Thr402Met and p.Val460Glu) further supporting a causative role for SLC7A8 in ARHL. Moreover, our preliminary data suggest that a relevant proportion of ARHL cases could be explained by SLC7A8 mutations. Age-related hearing loss affects about one in three individuals between the ages of 65 and 74. The first symptom is difficulty hearing high-pitched sounds like children’s voices. The disease starts gradually and worsens over time. Changes in the ear, the nerve that connects it to the brain, or the brain itself can cause hearing loss. Sometimes all three play a role. Genetics, exposure to noise, disease, and aging may all contribute. The condition is so complex it is difficult for scientists to pinpoint a primary suspect or develop treatments. Now, Guarch, Font-Llitjós et al. show that errors in a protein called SLC7A8 cause age-related hearing loss in mice and humans. The SLC7A8 protein acts like a door that allows amino acids – the building blocks of proteins – to enter or leave a cell. This door is blocked in mice lacking SLC7A8 and damage occurs in the part of their inner ear responsible for hearing. As a result, the animals lose their hearing. Next, Guarch, Font-Llitjós et al. scanned the genomes of 147 people from isolated villages in Italy for mutations in the gene for SLC7A8. The people also underwent hearing tests. Mutations in the gene for SLC7A8 that partially block the door and prevent the flow of amino acids were found in people with hearing loss. Some mutations in SLC7A8 that allow the door to stay open where found in people who could hear. The experiments suggest that certain mutations in the gene for SLC7A8 are likely an inherited cause of age-related hearing loss. It is possible that other proteins that control the flow of amino acids into or out of cells also may play a role in hearing. More studies are needed to see if it is possible to fix errors in the SLC7A8 protein to delay or restore the hearing loss.
Collapse
Affiliation(s)
- Meritxell Espino Guarch
- Experimental Genetics, Sidra Medical and Research Center, Doha, Qatar.,Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mariona Font-Llitjós
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain
| | - Silvia Murillo-Cuesta
- Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Alberto Sols Biomedical Research Institute (CSIC/UAM), Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Ekaitz Errasti-Murugarren
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain
| | - Adelaida M Celaya
- Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Alberto Sols Biomedical Research Institute (CSIC/UAM), Madrid, Spain
| | - Giorgia Girotto
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy.,Medical Genetics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Dragana Vuckovic
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy.,Medical Genetics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | | | - Clara Vilches
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain
| | - Susanna Bodoy
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain
| | - Ignasi Sahún
- Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Laura González
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain
| | - Esther Prat
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Genetics Section, Physiological Sciences Department, Health Sciences and Medicine Faculty, University of Barcelona, Barcelona, Spain
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Biochemistry and Molecular Biomedicine Department, Faculty of Biology, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre on Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - Mara Dierssen
- Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Isabel Varela-Nieto
- Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Alberto Sols Biomedical Research Institute (CSIC/UAM), Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Paolo Gasparini
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy.,Medical Genetics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Manuel Palacín
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Biochemistry and Molecular Biomedicine Department, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Virginia Nunes
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Genetics Section, Physiological Sciences Department, Health Sciences and Medicine Faculty, University of Barcelona, Barcelona, Spain
| |
Collapse
|
27
|
Regenerative medicine in hearing recovery. Cytotherapy 2017; 19:909-915. [DOI: 10.1016/j.jcyt.2017.04.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/24/2017] [Accepted: 04/21/2017] [Indexed: 12/20/2022]
|
28
|
Hepatocyte Growth Factor-c-MET Signaling Mediates the Development of Nonsensory Structures of the Mammalian Cochlea and Hearing. J Neurosci 2017; 36:8200-9. [PMID: 27488639 DOI: 10.1523/jneurosci.4410-15.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 06/28/2016] [Indexed: 12/28/2022] Open
Abstract
UNLABELLED The stria vascularis is a nonsensory structure that is essential for auditory hair cell function by maintaining potassium concentration of the scala media. During mouse embryonic development, a subpopulation of neural crest cell-derived melanocytes migrates and incorporates into a subregion of the cochlear epithelium, forming the intermediate cell layer of the stria vascularis. The relation of this developmental process to stria vascularis function is currently unknown. In characterizing the molecular differentiation of developing peripheral auditory structures, we discovered that hepatocyte growth factor (Hgf) is expressed in the future stria vascularis of the cochlear epithelium. Its receptor tyrosine kinase, c-Met, is expressed in the cochlear epithelium and melanocyte-derived intermediate cells in the stria vascularis. Genetic dissection of HGF signaling via c-MET reveals that the incorporation of the melanocytes into the future stria vascularis of the cochlear duct requires c-MET signaling. In addition, inactivation of either the ligand or receptor developmentally resulted in a profound hearing loss at young adult stages. These results suggest a novel connection between HGF signaling and deafness via melanocyte deficiencies. SIGNIFICANCE STATEMENT We found the roles of hepatocyte growth factor (HGF) signaling in stria vascularis development for the first time and that lack of HGF signaling in the inner ear leads to profound hearing loss in the mouse. Our findings reveal a novel mechanism that may underlie human deafness DFNB39 and DFNB97. Our findings reveal an additional example of context-dependent c-MET signaling diversity, required here for proper cellular invasion developmentally that is essential for specific aspects of auditory-related organogenesis.
Collapse
|
29
|
Fuentes-Santamaría V, Alvarado JC, Melgar-Rojas P, Gabaldón-Ull MC, Miller JM, Juiz JM. The Role of Glia in the Peripheral and Central Auditory System Following Noise Overexposure: Contribution of TNF-α and IL-1β to the Pathogenesis of Hearing Loss. Front Neuroanat 2017; 11:9. [PMID: 28280462 PMCID: PMC5322242 DOI: 10.3389/fnana.2017.00009] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/09/2017] [Indexed: 12/12/2022] Open
Abstract
Repeated noise exposure induces inflammation and cellular adaptations in the peripheral and central auditory system resulting in pathophysiology of hearing loss. In this study, we analyzed the mechanisms by which noise-induced inflammatory-related events in the cochlea activate glial-mediated cellular responses in the cochlear nucleus (CN), the first relay station of the auditory pathway. The auditory function, glial activation, modifications in gene expression and protein levels of inflammatory mediators and ultrastructural changes in glial-neuronal interactions were assessed in rats exposed to broadband noise (0.5-32 kHz, 118 dB SPL) for 4 h/day during 4 consecutive days to induce long-lasting hearing damage. Noise-exposed rats developed a permanent threshold shift which was associated with hair cell loss and reactive glia. Noise-induced microglial activation peaked in the cochlea between 1 and 10D post-lesion; their activation in the CN was more prolonged reaching maximum levels at 30D post-exposure. RT-PCR analyses of inflammatory-related genes expression in the cochlea demonstrated significant increases in the mRNA expression levels of pro- and anti-inflammatory cytokines, inducible nitric oxide synthase, intercellular adhesion molecule and tissue inhibitor of metalloproteinase-1 at 1 and 10D post-exposure. In noise-exposed cochleae, interleukin-1β (IL-1β), and tumor necrosis factor α (TNF-α) were upregulated by reactive microglia, fibrocytes, and neurons at all time points examined. In the CN, however, neurons were the sole source of these cytokines. These observations suggest that noise exposure causes peripheral and central inflammatory reactions in which TNF-α and IL-1β are implicated in regulating the initiation and progression of noise-induced hearing loss.
Collapse
Affiliation(s)
- Verónica Fuentes-Santamaría
- Instituto de Investigación en Discapacidades NeurológicasAlbacete, Spain; Facultad de Medicina, Universidad de Castilla-La ManchaAlbacete, Spain
| | - Juan Carlos Alvarado
- Instituto de Investigación en Discapacidades NeurológicasAlbacete, Spain; Facultad de Medicina, Universidad de Castilla-La ManchaAlbacete, Spain
| | - Pedro Melgar-Rojas
- Instituto de Investigación en Discapacidades NeurológicasAlbacete, Spain; Facultad de Medicina, Universidad de Castilla-La ManchaAlbacete, Spain
| | - María C Gabaldón-Ull
- Instituto de Investigación en Discapacidades NeurológicasAlbacete, Spain; Facultad de Medicina, Universidad de Castilla-La ManchaAlbacete, Spain
| | - Josef M Miller
- Center for Hearing and Communication Research and Department of Clinical Neuroscience, Karolinska InstitutetStockholm, Sweden; Kresge Hearing Research Institute, University of MichiganAnn Arbor, MI, USA
| | - José M Juiz
- Instituto de Investigación en Discapacidades NeurológicasAlbacete, Spain; Facultad de Medicina, Universidad de Castilla-La ManchaAlbacete, Spain
| |
Collapse
|
30
|
Abstract
T-box transcription factors play key roles in the regulation of developmental processes such as cell differentiation and migration. Mammals have 17 T-box genes, of which several regulate brain development. The Tbr1 subfamily of T-box genes is particularly important in development of the cerebral cortex, olfactory bulbs (OBs), and cerebellum. This subfamily is comprised of Tbr1, Tbr2 (also known as Eomes), and Tbx21. In developing cerebral cortex, Tbr2 and Tbr1 are expressed during successive stages of differentiation in the pyramidal neuron lineage, from Tbr2+ intermediate progenitors to Tbr1+ postmitotic glutamatergic neurons. At each stage, Tbr2 and Tbr1 regulate laminar and regional identity of cortical projection neurons, cell migration, and axon guidance. In the OB, Tbr1 subfamily genes regulate neurogenesis of mitral and tufted cells, and glutamatergic juxtaglomerular interneurons. Tbr2 is also prominent in the development of retinal ganglion cells in nonimage-forming pathways. Other regions that require Tbr2 or Tbr1 in development or adulthood include the cerebellum and adult dentate gyrus. In humans, de novo mutations in TBR1 are important causes of sporadic autism and intellectual disability. Further studies of T-box transcription factors will enhance our understanding of neurodevelopmental disorders and inform approaches to new therapies.
Collapse
|
31
|
Abstract
T-box (Tbx) genes encode an ancient group of transcription factors that play important roles in patterning, specification, proliferation, and differentiation programs in vertebrate organogenesis. This is testified by severe organ malformation syndromes in mice homozygous for engineered null alleles of specific T-box genes and by the large number of human inherited organ-specific diseases that have been linked to mutations in these genes. One of the organ systems that has not been associated with loss of specific T-box gene function in human disease for long is the excretory system. However, this has changed with the finding that mutations in TBX18, a member of a vertebrate-specific subgroup within the Tbx1-subfamily of T-box transcription factor genes, cause congenital anomalies of the kidney and urinary tract, predominantly hydroureter and ureteropelvic junction obstruction. Gene expression analyses, loss-of-function studies, and lineage tracing in the mouse suggest a primary role for this transcription factor in specifying the ureteric mesenchyme in the common anlage of the kidney, the ureter, and the bladder. We review the function of Tbx18 in ureterogenesis and discuss the body of evidence that Tbx18 and other members of the T-box gene family, namely, Tbx1, Tbx2, Tbx3, and Tbx20, play additional roles in development and homeostasis of other components of the excretory system in vertebrates.
Collapse
|
32
|
Alvarado JC, Fuentes-Santamaría V, Gabaldón-Ull MC, Jareño-Flores T, Miller JM, Juiz JM. Noise-Induced "Toughening" Effect in Wistar Rats: Enhanced Auditory Brainstem Responses Are Related to Calretinin and Nitric Oxide Synthase Upregulation. Front Neuroanat 2016; 10:19. [PMID: 27065815 PMCID: PMC4815363 DOI: 10.3389/fnana.2016.00019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/15/2016] [Indexed: 01/07/2023] Open
Abstract
An appropriate conditioning noise exposure may reduce a subsequent noise-induced threshold shift. Although this "toughening" effect helps to protect the auditory system from a subsequent traumatic noise exposure, the mechanisms that regulate this protective process are not fully understood yet. Accordingly, the goal of the present study was to characterize physiological processes associated with "toughening" and to determine their relationship to metabolic changes in the cochlea and cochlear nucleus (CN). Auditory brainstem responses (ABR) were evaluated in Wistar rats before and after exposures to a sound conditioning protocol consisting of a broad-band white noise of 118 dB SPL for 1 h every 72 h, four times. After the last ABR evaluation, animals were perfused and their cochleae and brains removed and processed for the activity markers calretinin (CR) and neuronal nitric oxide synthase (nNOS). Toughening was demonstrated by a progressively faster recovery of the threshold shift, as well as wave amplitudes and latencies over time. Immunostaining revealed an increase in CR and nNOS levels in the spiral ganglion, spiral ligament, and CN in noise-conditioned rats. Overall, these results suggest that the protective mechanisms of the auditory toughening effect initiate in the cochlea and extend to the central auditory system. Such phenomenon might be in part related to an interplay between CR and nitric oxide signaling pathways, and involve an increased cytosolic calcium buffering capacity induced by the noise conditioning protocol.
Collapse
Affiliation(s)
- Juan C Alvarado
- Instituto de Investigación en Discapacidades NeurológicasAlbacete, Spain; Facultad de Medicina, Universidad de Castilla-La ManchaAlbacete, Spain
| | - Verónica Fuentes-Santamaría
- Instituto de Investigación en Discapacidades NeurológicasAlbacete, Spain; Facultad de Medicina, Universidad de Castilla-La ManchaAlbacete, Spain
| | - María C Gabaldón-Ull
- Instituto de Investigación en Discapacidades NeurológicasAlbacete, Spain; Facultad de Medicina, Universidad de Castilla-La ManchaAlbacete, Spain
| | - Tania Jareño-Flores
- Grupo de Neurobiología de la Audición, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid Madrid, Spain
| | - Josef M Miller
- Center for Hearing and Communication Research and Department of Clinical Neuroscience, Karolinska InstitutetStockholm, Sweden; Kresge Hearing Research Institute, University of MichiganAnn Arbor, MI, USA
| | - José M Juiz
- Instituto de Investigación en Discapacidades NeurológicasAlbacete, Spain; Facultad de Medicina, Universidad de Castilla-La ManchaAlbacete, Spain
| |
Collapse
|
33
|
Yuan X, Du J, Qin Q, Li X, Deng S, Chen Y, Zhang L, She Q. The impact of sperm-expressed transcription factors on fate-mapping models. Reproduction 2015; 150:323-30. [DOI: 10.1530/rep-15-0014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 07/07/2015] [Indexed: 11/08/2022]
Abstract
Genetic lineage tracing has been used extensively in developmental biology. Many transcription factors expressed in sperm may induce Cre-mediated loxP recombination during early zygote development. In this study, we investigated the effect of sperm-expressed Cre on cell type-specific Cre-mediated loxP recombination in fate-mapping models of Tbx18+ progenitor cells. We found the recombination frequency in a reverse mating (RM) lineage was inconsistent with a normal Mendelian distribution. However, the recombination frequency in a positive mating (PM) lineage agreed with a Mendelian distribution. In the PM lineage, LacZ and EYFP were expressed in specific locations, such as the limb buds, heart, and hair follicles. Therefore, the reporter genes accurately and reliably traced cell differentiation in the PM lineage. In contrast, EYFP and LacZ were expressed throughout the embryo in the RM lineage. Thus, the reporter genes did not trace cell differentiation specifically in the RM lineage. Furthermore, Tbx18 mRNA and protein were expressed in the testicles of male mice, but almost no Tbx18 expression was detected in the ovaries of female mice. Similarly, reporter genes and Tbx18 were coexpressed in the seminiferous tubules and sperm cells of testicles. These results revealed that Cre-loxP-mediated pre-recombination in zygotes is due to Tbx18 expressed in testicle sperm cells when Cre is transmitted paternally. Our results indicate that Cre-mediated specific recombination in fate-mapping models of sperm-expressed genes may be influenced by the paternal origin of Cre. Therefore, a careful experimental design is critical when using the Cre-loxP system to trace spatial, temporal or tissue-specific fates.
Collapse
|
34
|
Melgar–Rojas P, Alvarado JC, Fuentes–Santamaría V, Gabaldón–Ull MC, Juiz JM. Validation of Reference Genes for RT-qPCR Analysis in Noise-Induced Hearing Loss: A Study in Wistar Rat. PLoS One 2015; 10:e0138027. [PMID: 26366995 PMCID: PMC4569353 DOI: 10.1371/journal.pone.0138027] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 08/24/2015] [Indexed: 12/26/2022] Open
Abstract
The reverse transcriptase–quantitative polymerase chain reaction (RT–qPCR) requires adequate normalization in order to ensure accurate results. The use of reference genes is the most common method to normalize RT–qPCR assays; however, many studies have reported that the expression of frequently used reference genes is more variable than expected, depending on experimental conditions. Consequently, proper validation of the stability of reference genes is an essential step when performing new gene expression studies. Despite the fact that RT–qPCR has been widely used to elucidate molecular correlates of noise–induced hearing loss (NIHL), up to date there are no reports demonstrating validation of reference genes for the evaluation of changes in gene expression after NIHL. Therefore, in this study we evaluated the expression of some commonly used reference genes (Arbp, b–Act, b2m, CyA, Gapdh, Hprt1, Tbp, Tfrc and UbC) and examined their suitability as endogenous control genes for RT–qPCR analysis in the adult Wistar rat in response to NIHL. Four groups of rats were noise–exposed to generate permanent cochlear damage. Cochleae were collected at different time points after noise exposure and the expression level of candidate reference genes was evaluated by RT–qPCR using geNorm, NormFinder and BestKeeper software to determine expression stability. The three independent applications revealed Tbp as the most stably expressed reference gene. We also suggest a group of top–ranked reference genes that can be combined to obtain suitable reference gene pairs for the evaluation of the effects of noise on gene expression in the cochlea. These findings provide essential basis for further RT–qPCR analysis in studies of NIHL using Wistar rats as animal model.
Collapse
Affiliation(s)
- Pedro Melgar–Rojas
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), School of Medicine, University of Castilla–La Mancha, Campus in Albacete, Albacete, Spain
- * E-mail:
| | - Juan Carlos Alvarado
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), School of Medicine, University of Castilla–La Mancha, Campus in Albacete, Albacete, Spain
| | - Verónica Fuentes–Santamaría
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), School of Medicine, University of Castilla–La Mancha, Campus in Albacete, Albacete, Spain
| | - María Cruz Gabaldón–Ull
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), School of Medicine, University of Castilla–La Mancha, Campus in Albacete, Albacete, Spain
| | - José M. Juiz
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), School of Medicine, University of Castilla–La Mancha, Campus in Albacete, Albacete, Spain
| |
Collapse
|
35
|
Papaioannou VE. The T-box gene family: emerging roles in development, stem cells and cancer. Development 2014; 141:3819-33. [PMID: 25294936 DOI: 10.1242/dev.104471] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The T-box family of transcription factors exhibits widespread involvement throughout development in all metazoans. T-box proteins are characterized by a DNA-binding motif known as the T-domain that binds DNA in a sequence-specific manner. In humans, mutations in many of the genes within the T-box family result in developmental syndromes, and there is increasing evidence to support a role for these factors in certain cancers. In addition, although early studies focused on the role of T-box factors in early embryogenesis, recent studies in mice have uncovered additional roles in unsuspected places, for example in adult stem cell populations. Here, I provide an overview of the key features of T-box transcription factors and highlight their roles and mechanisms of action during various stages of development and in stem/progenitor cell populations.
Collapse
Affiliation(s)
- Virginia E Papaioannou
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
36
|
Deficiency of transcription factor Brn4 disrupts cochlear gap junction plaques in a model of DFN3 non-syndromic deafness. PLoS One 2014; 9:e108216. [PMID: 25259580 PMCID: PMC4178122 DOI: 10.1371/journal.pone.0108216] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 08/26/2014] [Indexed: 11/19/2022] Open
Abstract
Brn4, which encodes a POU transcription factor, is the gene responsible for DFN3, an X chromosome-linked, non-syndromic type of hearing loss. Brn4-deficient mice have a low endocochlear potential (EP), hearing loss, and ultrastructural alterations in spiral ligament fibrocytes, however the molecular pathology through which Brn4 deficiency causes low EP is still unclear. Mutations in the Gjb2 and Gjb6 genes encoding the gap junction proteins connexin26 (Cx26) and connexin30 (Cx30) genes, respectively, which encode gap junction proteins and are expressed in cochlear fibrocytes and non-sensory epithelial cells (i.e., cochlear supporting cells) to maintain the proper EP, are responsible for hereditary sensorineural deafness. It has been hypothesized that the gap junction in the cochlea provides an intercellular passage by which K+ is transported to maintain the EP at the high level necessary for sensory hair cell excitation. Here we analyzed the formation of gap junction plaques in cochlear supporting cells of Brn4-deficient mice at different stages by confocal microscopy and three-dimensional graphic reconstructions. Gap junctions from control mice, which are composed mainly of Cx26 and Cx30, formed linear plaques along the cell-cell junction sites with adjacent cells. These plaques formed pentagonal or hexagonal outlines of the normal inner sulcus cells and border cells. Gap junction plaques in Brn4-deficient mice did not, however, show the normal linear structure but instead formed small spots around the cell-cell junction sites. Gap junction lengths were significantly shorter, and the level of Cx26 and Cx30 was significantly reduced in Brn4-deficient mice compared with littermate controls. Thus the Brn4 mutation affected the assembly and localization of gap junction proteins at the cell borders of cochlear supporting cells, suggesting that Brn4 substantially contributes to cochlear gap junction properties to maintain the proper EP in cochleae, similar to connexin-related deafness.
Collapse
|
37
|
A distant downstream enhancer directs essential expression of Tbx18 in urogenital tissues. Dev Biol 2014; 392:483-93. [PMID: 24854998 DOI: 10.1016/j.ydbio.2014.05.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 05/09/2014] [Accepted: 05/12/2014] [Indexed: 11/22/2022]
Abstract
The vertebrate T-box transcription factor gene Tbx18 performs a vital role in development of multiple organ systems. Tbx18 insufficiency manifests as recessive phenotypes in the upper urinary system, cardiac venous pole, inner ear, and axial skeleton; homozygous null mutant animals die perinatally. Here, we report a new regulatory mutation of Tbx18, a reciprocal translocation breaking 78kbp downstream of the gene. 12Gso homozygotes present urinary and vertebral defects very similar to those associated with Tbx18-null mutations, but 12Gso is clearly not a global null allele since homozygotes survive into adulthood. We show that 12Gso down-regulates Tbx18 expression in a manner that is both spatially- and temporally-specific; combined with other data, the mutation points particularly to the presence of an essential urogenital enhancer located near the translocation breakpoint site. In support of this hypothesis, we identify a distal enhancer element, ECR1, which is active in developing urogenital and other tissues; we propose that disruption of this element leads to premature loss of Tbx18 function in 12Gso mutant mice. These data reveal a long-range regulatory architecture extending far downstream of Tbx18, identify a novel and likely essential urogenital enhancer, and introduce a new tool for dissecting postnatal phenotypes associated with dysregulation of Tbx18.
Collapse
|
38
|
Bohnenpoll T, Trowe MO, Wojahn I, Taketo MM, Petry M, Kispert A. Canonical Wnt signaling regulates the proliferative expansion and differentiation of fibrocytes in the murine inner ear. Dev Biol 2014; 391:54-65. [PMID: 24727668 DOI: 10.1016/j.ydbio.2014.03.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 03/27/2014] [Accepted: 03/29/2014] [Indexed: 01/18/2023]
Abstract
Otic fibrocytes tether the cochlear duct to the surrounding otic capsule but are also critically involved in maintenance of ion homeostasis in the cochlea, thus, perception of sound. The molecular pathways that regulate the development of this heterogenous group of cells from mesenchymal precursors are poorly understood. Here, we identified epithelial Wnt7a and Wnt7b as possible ligands of Fzd-mediated β-catenin (Ctnnb1)-dependent (canonical) Wnt signaling in the adjacent undifferentiated periotic mesenchyme (POM). Mice with a conditional deletion of Ctnnb1 in the POM exhibited a complete failure of fibrocyte differentiation, a severe reduction of mesenchymal cells surrounding the cochlear duct, loss of pericochlear spaces, a thickening and partial loss of the bony capsule and a secondary disturbance of cochlear duct coiling shortly before birth. Analysis at earlier stages revealed that radial patterning of the POM in two domains with highly condensed cartilaginous precursors and more loosely arranged inner mesenchymal cells occurred normally but that proliferation in the inner domain was reduced and cytodifferentiation failed. Cells with mis/overexpression of a stabilized form of Ctnnb1 in the entire POM mesenchyme sorted to the inner mesenchymal compartment and exhibited increased proliferation. Our analysis suggests that Wnt signals from the cochlear duct epithelium are crucial to induce differentiation and expansion of fibrocyte precursor cells. Our findings emphasize the importance of epithelial-mesenchymal signaling in inner ear development.
Collapse
Affiliation(s)
- Tobias Bohnenpoll
- Institut für Molekularbiologie, OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Mark-Oliver Trowe
- Institut für Molekularbiologie, OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Irina Wojahn
- Institut für Molekularbiologie, OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | | | - Marianne Petry
- Institut für Molekularbiologie, OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Andreas Kispert
- Institut für Molekularbiologie, OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany.
| |
Collapse
|
39
|
Abstract
Regulated transcription controls the diversity, developmental pathways and spatial organization of the hundreds of cell types that make up a mammal. Using single-molecule cDNA sequencing, we mapped transcription start sites (TSSs) and their usage in human and mouse primary cells, cell lines and tissues to produce a comprehensive overview of mammalian gene expression across the human body. We find that few genes are truly 'housekeeping', whereas many mammalian promoters are composite entities composed of several closely separated TSSs, with independent cell-type-specific expression profiles. TSSs specific to different cell types evolve at different rates, whereas promoters of broadly expressed genes are the most conserved. Promoter-based expression analysis reveals key transcription factors defining cell states and links them to binding-site motifs. The functions of identified novel transcripts can be predicted by coexpression and sample ontology enrichment analyses. The functional annotation of the mammalian genome 5 (FANTOM5) project provides comprehensive expression profiles and functional annotation of mammalian cell-type-specific transcriptomes with wide applications in biomedical research.
Collapse
|
40
|
Takeichi M, Nimura K, Mori M, Nakagami H, Kaneda Y. The transcription factors Tbx18 and Wt1 control the epicardial epithelial-mesenchymal transition through bi-directional regulation of Slug in murine primary epicardial cells. PLoS One 2013; 8:e57829. [PMID: 23469079 PMCID: PMC3585213 DOI: 10.1371/journal.pone.0057829] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 01/26/2013] [Indexed: 01/05/2023] Open
Abstract
During cardiac development, a subpopulation of epicardial cells migrates into the heart as part of the epicardial epithelial-mesenchymal transition (EMT) and differentiates into smooth muscle cells and fibroblasts. However, the roles of transcription factors in the epicardial EMT are poorly understood. Here, we show that two transcription factors expressed in the developing epicardium, T-box18 (Tbx18) and Wilms’ tumor 1 homolog (Wt1), bi-directionally control the epicardial EMT through their effects on Slug expression in murine primary epicardial cells. Knockdown of Wt1 induced the epicardial EMT, which was accompanied by an increase in the migration and expression of N-cadherin and a decrease in the expression of ZO-1 as an epithelial marker. By contrast, knockdown of Tbx18 inhibited the mesenchymal transition induced by TGFβ1 treatment and Wt1 knockdown. The expression of Slug but not Snail decreased as a result of Tbx18 knockdown, but Slug expression increased following knockdown of Wt1. Knockdown of Slug also attenuated the epicardial EMT induced by TGFβ1 treatment and Wt1 knockdown. Furthermore, in normal murine mammary gland-C7 (NMuMG-C7) cells, Tbx18 acted to increase Slug expression, while Wt1 acted to decrease Slug expression. Chromatin immunoprecipitation and promoter assay revealed that Tbx18 and Wt1 directly bound to the Slug promoter region and regulated Slug expression. These results provide new insights into the regulatory mechanisms that control the epicardial EMT.
Collapse
Affiliation(s)
- Makiko Takeichi
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Keisuke Nimura
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Masaki Mori
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Stem Cell Program, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Hironori Nakagami
- Division of Vascular Medicine and Epigenetics, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan
- * E-mail: (HN); (YK)
| | - Yasufumi Kaneda
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- * E-mail: (HN); (YK)
| |
Collapse
|
41
|
Buniello A, Hardisty-Hughes RE, Pass JC, Bober E, Smith RJ, Steel KP. Headbobber: a combined morphogenetic and cochleosaccular mouse model to study 10qter deletions in human deafness. PLoS One 2013; 8:e56274. [PMID: 23457544 PMCID: PMC3572983 DOI: 10.1371/journal.pone.0056274] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 01/08/2013] [Indexed: 02/07/2023] Open
Abstract
The recessive mouse mutant headbobber (hb) displays the characteristic behavioural traits associated with vestibular defects including headbobbing, circling and deafness. This mutation was caused by the insertion of a transgene into distal chromosome 7 affecting expression of native genes. We show that the inner ear of hb/hb mutants lacks semicircular canals and cristae, and the saccule and utricle are fused together in a single utriculosaccular sac. Moreover, we detect severe abnormalities of the cochlear sensory hair cells, the stria vascularis looks severely disorganised, Reissner's membrane is collapsed and no endocochlear potential is detected. Myo7a and Kcnj10 expression analysis show a lack of the melanocyte-like intermediate cells in hb/hb stria vascularis, which can explain the absence of endocochlear potential. We use Trp2 as a marker of melanoblasts migrating from the neural crest at E12.5 and show that they do not interdigitate into the developing strial epithelium, associated with abnormal persistence of the basal lamina in the hb/hb cochlea. We perform array CGH, deep sequencing as well as an extensive expression analysis of candidate genes in the headbobber region of hb/hb and littermate controls, and conclude that the headbobber phenotype is caused by: 1) effect of a 648 kb deletion on distal Chr7, resulting in the loss of three protein coding genes (Gpr26, Cpmx2 and Chst15) with expression in the inner ear but unknown function; and 2) indirect, long range effect of the deletion on the expression of neighboring genes on Chr7, associated with downregulation of Hmx3, Hmx2 and Nkx1.2 homeobox transcription factors. Interestingly, deletions of the orthologous region in humans, affecting the same genes, have been reported in nineteen patients with common features including sensorineural hearing loss and vestibular problems. Therefore, we propose that headbobber is a useful model to gain insight into the mechanisms underlying deafness in human 10qter deletion syndrome.
Collapse
Affiliation(s)
- Annalisa Buniello
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | | | - Johanna C. Pass
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Eva Bober
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Karen P. Steel
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
- MRC Institute of Hearing Research, Nottingham, United Kingdom
- * E-mail:
| |
Collapse
|
42
|
Greulich F, Farin HF, Schuster-Gossler K, Kispert A. Tbx18 function in epicardial development. Cardiovasc Res 2012; 96:476-83. [PMID: 22926762 DOI: 10.1093/cvr/cvs277] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS The embryonic epicardium is a source of smooth muscle cells and fibroblasts of the coronary vasculature and of the myocardium, but the molecular circuits that direct the temporal and spatial generation of these cell types from epicardium-derived cells are only partly known. We aimed to elucidate the functional significance of the conserved epicardial expression of the T-box transcription factor gene Tbx18 using transgenic technology in the mouse. METHODS AND RESULTS We show by cellular and molecular analyses that in Tbx18-deficient mice the epicardium is formed normally and that epicardial cells undergo an epithelial-mesenchymal transition, differentiate into smooth muscle cells and fibroblasts, and form a normal coronary vasculature and fibrous skeleton. Prolonged expression of Tbx18 in epicardium-derived cells by a transgenic approach in vivo does not affect the differentiation and migratory behaviour of these cells. In contrast, epicardial misexpression of a transcriptional activator version of Tbx18, Tbx18VP16, results in premature smooth muscle differentiation of epicardial cells. Inhibition of Notch and transforming growth factor beta receptor signalling in Tbx18VP16 expressing epicardial cells in explant cultures reverts this phenotype. CONCLUSION Tbx18 is dispensable for epicardial development, yet a repressive T-box function may be required to prevent premature smooth muscle cell differentiation by repressing transforming growth factor beta receptor and Notch signalling in the embryonic epicardium.
Collapse
Affiliation(s)
- Franziska Greulich
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, OE5250, Carl-Neuberg-Str.1, Hannover D-30625, Germany
| | | | | | | |
Collapse
|
43
|
Zhang Y, Zhang W, Johnston AH, Newman TA, Pyykkö I, Zou J. Targeted delivery of Tet1 peptide functionalized polymersomes to the rat cochlear nerve. Int J Nanomedicine 2012. [PMID: 22403485 DOI: 10.2147/ijn.s28185ijn-7-1015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Polymersomes are nanosized vesicles formed from amphiphilic block copolymers, and have been identified as potential drug delivery vehicles to the inner ear. The aim of this study was to provide targeting to specific cells within the inner ear by functionalizing the polymersome surface with Tet1 peptide sequence. Tet1 peptide specifically binds to the trisialoganglioside clostridial toxin receptor on neurons and was expected to target the polymersomes toward the cochlear nerve. The Tet1 functionalized PEG-b-PCL polymersomes were administered using routine drug delivery routes: transtympanic injection and cochleostomy. Delivery via cochleostomy of Tet1 functionalized polymersomes resulted in cochlear nerve targeting; in contrast this was not seen after transtympanic injection.
Collapse
Affiliation(s)
- Ya Zhang
- Department of Otolaryngology, University of Tampere, Medical School, Tampere, Finland
| | | | | | | | | | | |
Collapse
|
44
|
Zhang Y, Zhang W, Johnston AH, Newman TA, Pyykkö I, Zou J. Targeted delivery of Tet1 peptide functionalized polymersomes to the rat cochlear nerve. Int J Nanomedicine 2012; 7:1015-22. [PMID: 22403485 PMCID: PMC3292415 DOI: 10.2147/ijn.s28185] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Polymersomes are nanosized vesicles formed from amphiphilic block copolymers, and have been identified as potential drug delivery vehicles to the inner ear. The aim of this study was to provide targeting to specific cells within the inner ear by functionalizing the polymersome surface with Tet1 peptide sequence. Tet1 peptide specifically binds to the trisialoganglioside clostridial toxin receptor on neurons and was expected to target the polymersomes toward the cochlear nerve. The Tet1 functionalized PEG-b-PCL polymersomes were administered using routine drug delivery routes: transtympanic injection and cochleostomy. Delivery via cochleostomy of Tet1 functionalized polymersomes resulted in cochlear nerve targeting; in contrast this was not seen after transtympanic injection.
Collapse
Affiliation(s)
- Ya Zhang
- Department of Otolaryngology, University of Tampere, Medical School, Tampere, Finland
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
Normal blood supply to the cochlea is critically important for establishing the endocochlear potential and sustaining production of endolymph. Abnormal cochlear microcirculation has long been considered an etiologic factor in noise-induced hearing loss, age-related hearing loss (presbycusis), sudden hearing loss or vestibular function, and Meniere's disease. Knowledge of the mechanisms underlying the pathophysiology of cochlear microcirculation is of fundamental clinical importance. A better understanding of cochlear blood flow (CoBF) will enable more effective management of hearing disorders resulting from aberrant blood flow. This review focuses on recent discoveries and findings related to the physiopathology of the cochlear microvasculature.
Collapse
Affiliation(s)
- Xiaorui Shi
- Oregon Hearing Research Center (NRC04), Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
46
|
Impaired stria vascularis integrity upon loss of E-cadherin in basal cells. Dev Biol 2011; 359:95-107. [PMID: 21925491 DOI: 10.1016/j.ydbio.2011.08.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 08/11/2011] [Accepted: 08/31/2011] [Indexed: 12/28/2022]
Abstract
In the cochlea, sensory transduction depends on the endocochlear potential (EP) and the unique composition of the endolymph, both of which are maintained by a highly specialized epithelium at the cochlear lateral wall, the stria vascularis. The generation of the EP by the stria vascularis, in turn, relies on the insulation of an intrastrial extracellular compartment by epithelial basal cells. Despite the physiological importance of basal cells, their cellular origin and the molecular pathways that lead to their differentiation are unclear. Here, we show by genetic lineage tracing in the mouse that basal cells exclusively derive from the otic mesenchyme. Conditional deletion of E-cadherin in the otic mesenchyme and its descendants does not abrogate the transition from mesenchymal precursors to epithelial basal cells. Rather, dedifferentiation of intermediate cells, altered morphology of basal and marginal cells and hearing impairment due to decreased EP in E-cadherin mutant mice demonstrate an essential role of E-cadherin in terminal basal cell differentiation and their interaction with other strial cell types to establish and maintain the functional architecture of the stria vascularis.
Collapse
|
47
|
Dai M, Shi X. Fibro-vascular coupling in the control of cochlear blood flow. PLoS One 2011; 6:e20652. [PMID: 21673815 PMCID: PMC3106013 DOI: 10.1371/journal.pone.0020652] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Accepted: 05/06/2011] [Indexed: 12/20/2022] Open
Abstract
Background Transduction of sound in the cochlea is metabolically demanding. The lateral
wall and hair cells are critically vulnerable to hypoxia, especially at high
sound levels, and tight control over cochlear blood flow (CBF) is a
physiological necessity. Yet despite the importance of CBF for hearing,
consensus on what mechanisms are involved has not been obtained. Methodology/Principal Findings We report on a local control mechanism for regulating inner ear blood flow
involving fibrocyte signaling. Fibrocytes in the super-strial region are
spatially distributed near pre-capillaries of the spiral ligament of the
albino guinea pig cochlear lateral wall, as demonstrably shown in
transmission electron microscope and confocal images. Immunohistochemical
techniques reveal the inter-connected fibrocytes to be positive for
Na+/K+ ATPase β1 and S100. The connected fibrocytes display
more Ca2+ signaling than other cells in the cochlear lateral
wall as indicated by fluorescence of a Ca2+ sensor, fluo-4.
Elevation of Ca2+ in fibrocytes, induced by photolytic
uncaging of the divalent ion chelator o-nitrophenyl EGTA,
results in propagation of a Ca2+ signal to neighboring
vascular cells and vasodilation in capillaries. Of more physiological
significance, fibrocyte to vascular cell coupled signaling was found to
mediate the sound stimulated increase in cochlear blood flow (CBF).
Cyclooxygenase-1 (COX-1) was required for capillary dilation. Conclusions/Significance The findings provide the first evidence that signaling between fibrocytes and
vascular cells modulates CBF and is a key mechanism for meeting the cellular
metabolic demand of increased sound activity.
Collapse
Affiliation(s)
- Min Dai
- Oregon Hearing Research Center, Department of Otolaryngology/Head and
Neck Surgery, Oregon Health & Science University, Portland, Oregon, United
States of America
| | - Xiaorui Shi
- Oregon Hearing Research Center, Department of Otolaryngology/Head and
Neck Surgery, Oregon Health & Science University, Portland, Oregon, United
States of America
- The Institute of Microcirculation, Chinese Academy of Medical Sciences
and Peking Union Medical College, Beijing, China
- Department of Otolaryngology, Renji Hospital, Shanghai Jiao Tong
University, Shanghai, China
- * E-mail:
| |
Collapse
|
48
|
Kelly JJ, Forge A, Jagger DJ. Development of gap junctional intercellular communication within the lateral wall of the rat cochlea. Neuroscience 2011; 180:360-9. [PMID: 21320575 DOI: 10.1016/j.neuroscience.2011.02.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 02/03/2011] [Accepted: 02/05/2011] [Indexed: 01/27/2023]
Abstract
Auditory function depends on gap junctional intercellular communication (GJIC) between fibrocytes within the cochlear spiral ligament, and basal cells and intermediate cells within stria vascularis. This communication within the lateral wall is hypothesized to support recirculation of K+ from perilymph to the intra-strial space, and thus is essential for the high [K+] measured within endolymph, and the generation of the endocochlear potential. In rats, the [K+] within endolymph reaches adult levels by postnatal day 7 (P7), several days before hearing onset, suggesting that GJIC matures before auditory responses are detectable. In this study we have mapped the postnatal development of GJIC within the cochlear lateral wall, to determine the stage at which direct communication first exists between the spiral ligament and stria vascularis. Connexin 30 immunofluorescence revealed a progressive increase of gap junction plaque numbers from P0 onwards, initially in the condensing mesenchyme behind strial marginal cells, and spreading throughout the lateral wall by P7-P8. Whole-cell patch clamp experiments revealed compartmentalized intercellular dye-coupling in the lateral wall between P2 and P5. There was extensive dye-coupling throughout the fibrocyte syncytium by P7. Also, by P7 dye introduced to fibrocytes could also be detected within strial basal cells and intermediate cells. These data suggest that lateral wall function matures several days in advance of hearing onset, and provide anatomical evidence of the existence of a putative K+ recirculation pathway within the cochlear lateral wall.
Collapse
Affiliation(s)
- J J Kelly
- UCL Ear Institute, University College London, 332 Gray's Inn Road, London WC1X8EE, UK
| | | | | |
Collapse
|
49
|
Haugas M, Lilleväli K, Hakanen J, Salminen M. Gata2 is required for the development of inner ear semicircular ducts and the surrounding perilymphatic space. Dev Dyn 2011; 239:2452-69. [PMID: 20652952 DOI: 10.1002/dvdy.22373] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Gata2 has essential roles in the development of many organs. During mouse inner ear morphogenesis, it is expressed in otic vesicle and the surrounding periotic mesenchyme from early on, but no defects in the ear development of Gata2 null mice have been observed before lethality at embryonic day (E) 10.5. Here, we used conditional gene targeting to reveal the role of Gata2 at later stages of inner ear development. We show that Gata2 is critically required from E14.5-E15.5 onward for vestibular morphogenesis. Without Gata2 the semicircular ducts fail to grow to their normal size and the surrounding mesenchymal cells are not removed properly to generate the perilymphatic space. Gata2 is the first factor known to control the clearing of the vestibular perilymphatic mesenchyme, but interestingly, it is not required for the formation of the cochlear perilymphatic areas, suggesting distinct molecular control for these processes.
Collapse
Affiliation(s)
- Maarja Haugas
- Department of Veterinary Biosciences, University of Helsinki, Finland
| | | | | | | |
Collapse
|
50
|
Song MH, Choi SY, Wu L, Oh SK, Lee HK, Lee DJ, Shim DB, Choi JY, Kim UK, Bok J. Pou3f4 deficiency causes defects in otic fibrocytes and stria vascularis by different mechanisms. Biochem Biophys Res Commun 2010; 404:528-33. [PMID: 21144821 DOI: 10.1016/j.bbrc.2010.12.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Accepted: 12/02/2010] [Indexed: 02/06/2023]
Abstract
DFN3, the most prevalent X-linked hearing loss, is caused by mutations in the POU3F4 gene. Previous studies in Pou3f4 knockout mice suggest that defective otic fibrocytes in the spiral ligament of the cochlear lateral wall may underlie the hearing loss in DFN3. To better understand the pathological mechanisms of the DFN3 hearing loss, we analyzed inner ears of Pou3f4-deficient mice during development. Our results indicate that compartmentalization of the spiral ligament mesenchyme setting up boundaries for specific otic fibrocytes occurs normally in Pou3f4-deficient cochlea. However, differentiation of the compartmentalized mesenchyme into specific otic fibrocytes was blocked in the absence of Pou3f4 function. In addition, we found that stria vascularis in the cochlear lateral wall was also affected in Pou3f4-deficient cochlea. Unlike the otic fibrocytes, differentiation of stria vascularis was completed in the absence of Pou3f4 function, yet expression of Kir4.1 channels in the strial intermediate cells, essential for the sound transduction, was lost afterwards. These results suggest that Pou3f4 deficiency causes defects in both otic fibrocytes and stria vascularis at different developmental stages and by different pathological mechanisms, which may account for the progressive nature of DFN3 hearing loss.
Collapse
Affiliation(s)
- Mee Hyun Song
- Department of Otorhinolaryngology, Kwandong University College of Medicine, Goyang 412-270, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|