1
|
Miranda Portillo LS, Huang AP, Hosamani IV, Sanchez CN, Heller S, Benkafadar N. Anatomical and Molecular Insights into Avian Inner Ear Sensory Hair Cell Regeneration. Dev Biol 2025:S0012-1606(25)00144-7. [PMID: 40414451 DOI: 10.1016/j.ydbio.2025.05.021] [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: 04/08/2025] [Revised: 05/19/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Inner ear sensory hair cells are essential for auditory and vestibular functions. In mammals, loss of these cells leads to permanent hearing loss due to the inability of supporting cells to regenerate hair cells. In contrast, avian species exhibit a remarkable capacity for hair cell regeneration, primarily through the activation and proliferation of supporting cells. This review provides a comprehensive examination of the anatomical and molecular mechanisms underlying sensory hair cell regeneration in two critical avian inner ear structures: the basilar papilla and the utricle. We describe the structural and functional differences between avian and mammalian inner ear epithelia and highlight how these distinctions correlate with regenerative capabilities. Specifically, we discuss two distinct regenerative mechanisms - mitotic regeneration and direct transdifferentiation - employed by avian supporting cells in response to hair cell loss. We also explore how epithelial organization influences regenerative responses, including cellular density, cytoskeletal dynamics such as circumferential filamentous actin bands, and mechanical properties like tissue jamming and unjamming states. Additionally, we examine molecular pathways such as Hippo signaling, which mediates mechanical cues critical for regulating supporting cell proliferation and differentiation during regeneration. Recent advancements in single-cell -omics technologies have further elucidated molecular signatures and signaling pathways involved in these processes, offering novel insights that may inform therapeutic strategies aimed at inducing hair cell regeneration in mammals. This review highlights key anatomical and molecular concepts derived from avian models that hold promise for overcoming regenerative limitations in mammalian inner ears, paving the way for innovative treatments for hearing loss.
Collapse
Affiliation(s)
- Lyn S Miranda Portillo
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Austin P Huang
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Ishwar V Hosamani
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Celeste N Sanchez
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Stefan Heller
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305.
| | - Nesrine Benkafadar
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305; Institute for Neurosciences of Montpellier (INM), University Montpellier, INSERM, UMR 1298, 80 Rue Augustin Fliche, 34295, Montpellier, France.
| |
Collapse
|
2
|
Li BZ, Lin CY, Xu WB, Zhang YM, Shao QJ, Dong WR, Shu MA. The first identification and functional analysis of two drosophila mothers against decapentaplegic protein genes (SpSmad1 and SpSmad2/3) and their involvement in the innate immune response in Scylla paramamosain. FISH & SHELLFISH IMMUNOLOGY 2023; 143:109183. [PMID: 37884105 DOI: 10.1016/j.fsi.2023.109183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
Smad,a member of the TGF-β superfamily,controls cell proliferation,growth and guiding cell differentiation, thus playing a crucial role in diseases. However, the presence as well as specific function of Smad in crabs is still unknown. In this study, two Smads (Smad1 and Smad2/3) were identified for the first time from the mud crab Scylla paramamosain. The complete open reading frames of SpSmad1 and SpSmad2/3 were 1,497bp and 1,338bp, encoding deduced proteins of 498 and 445 amino acids respectively. Moreover, under the administration of Vibrio alginolyticus and WSSV, the relative expression levels of SpSmad1 and SpSmad2/3 were significantly increased, indicating their involvement in the innate immune response of mud crabs. Knockdown of SpSmad1 and SpSmad2/3 in vivo not only led to the increasement of the expressions of NF-κB signaling genes and antimicrobial peptides genes, but also significantly affected the bacterial clearance process of mud crabs. Additionally, overexpression of SpSmad1 and SpSmad2/3 in HEK293T cells could markedly activate NF-κB signaling. These results indicated that Smad1 and Smad2/3 participated in the innate immunity of Scylla paramamosain, and might provide a better understanding of the presence and immune regulatory functions of Smad1 and Smad2/3 in crabs and even invertebrates.
Collapse
Affiliation(s)
- Bang-Ze Li
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Chen-Yang Lin
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wen-Bin Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yan-Mei Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qing-Jun Shao
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wei-Ren Dong
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Miao-An Shu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
3
|
Hansdah K, Singh N, Bouzid A, Priyadarshi S, Ray CS, Desai A, Panda KC, Choudhury JC, Biswal NC, Tekari A, Masmoudi S, Ramchander PV. Evaluation of the Genetic Association and mRNA Expression of the COL1A1, BMP2, and BMP4 Genes in the Development of Otosclerosis. Genet Test Mol Biomarkers 2020; 24:343-351. [PMID: 32379989 DOI: 10.1089/gtmb.2019.0235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: Otosclerosis (OTSC) is a genetically heterogeneous disorder, characterized by abnormal bone growth in the middle ear, affecting the stapes bone. Previous studies have shown that single nucleotide polymorphisms (SNPs) of the COL1A1, BMP2, and BMP4 genes are linked to susceptibility of OTSC, musculoskeletal degenerative diseases, and bone remodeling. Aims: To evaluate the genetic association and expression levels of COL1A1, BMP2, and BMP4 genes with OTSC in the Indian population. Methods: A total of 320 otosclerotic and 320 control samples were screened for four SNPs (rs1107946, rs11327935, rs2269336, and rs1800012) of the COL1A1 gene; rs3178250 of the BMP2 gene; and rs17563 of the BMP4 gene using single-strand conformation polymorphism analysis, and restriction fragment length polymorphism analyses. Genotypic, haplotypic, and linkage disequilibrium analyses were performed to assess the potential associations of these SNPs with OTSC. COL1A1, BMP2, and BMP4 mRNA expression levels were analyzed by semiquantitative RT-PCR and real-time PCR. Results: Genotypes of two SNPs, rs1800012 and rs17563, were found to be associated with OTSC (the rs1800012 GT genotype, p = 0.0022, OR = 0.481; and the rs17563 TC genotype, p = 0.0225, OR = 1.471). Haplotypic analyses revealed that the COL1A1 haplotype G-T-C-T (p = 0.021) was significantly increased among controls. Functional studies revealed an unexpected decrease in mRNA expression of COL1A1 but an increased expression of the BMP2 and BMP4 genes in otosclerotic stapes tissues. Conclusions: Our findings suggest that OTSC is a heterogeneous disorder, but that the GT genotype of the rs1800012 locus is protective and that the TC genotype at the rs17563 locus is a risk factor. In addition, our studies indicate that changes in the expression of the COL1A1, BMP2, and BMP4 genes may contribute to the genetic susceptibility of OTSC by regulating their mRNA levels.
Collapse
Affiliation(s)
- Kirtal Hansdah
- Institute of Life Sciences, Nalco Square, Bhubaneswar, India
| | - Neha Singh
- Institute of Life Sciences, Nalco Square, Bhubaneswar, India
| | - Amal Bouzid
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | | | - Chinmay Sundar Ray
- Department of Ear, Nose, and Throat (ENT), Shrirama Chandra Bhanja (SCB) Medical College & Hospital, Cuttack, India
| | - Ashim Desai
- Dr. ABR Desai Ear, Nose and Throat (ENT) Clinic and Research Centre, Mumbai, India
| | | | - Jyotish Chandra Choudhury
- Department of Forensic Medicine & Toxicology (FMT), Shrirama Chandra Bhanja (SCB) Medical College & Hospital, Cuttack, India
| | - Narayan Chandra Biswal
- Department of Ear, Nose, and Throat (ENT), Shrirama Chandra Bhanja (SCB) Medical College & Hospital, Cuttack, India
| | - Adel Tekari
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Saber Masmoudi
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | | |
Collapse
|
4
|
Lipopolysaccharide (LPS) inhibits ectopic bone formation induced by bone morphogenetic protein-2 and TGF-β1 through IL-1β production. J Oral Biosci 2020; 62:44-51. [PMID: 31987892 DOI: 10.1016/j.job.2020.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 01/06/2023]
Abstract
OBJECTIVES In order to gain new insight into bacterial infection during bone-regenerative treatment using bone morphogenetic proteins (BMPs), we examined the effects of lipopolysaccharide (LPS) on ectopic bone formation induced by BMP-2 and transforming growth factor (TGF)-β1 in mice. METHODS We implanted collagen sponges containing BMP-2, TGF-β1, and various amounts of LPS into mouse muscle tissues. Lump-like masses in which ectopic bones developed in mice were processed for microcomputed tomography, DNA microarray, reverse-transcription PCR, and histological analyses. RESULTS LPS treatment caused a dose-dependent reduction in the volume of ectopic bone. The total volume of ectopic bone induced by BMP-2 + TGF-β1 treatment was reduced by more than 75% in the presence of LPS. Histological analysis of the ectopic bone tissues revealed a significant reduction in total bone volume and bone volume/total volume in response to LPS. LPS treatment significantly increased the osteoblast number and osteoid volume, while the osteoclast number did not change. Since LPS induced production of TNF-α and IL-1β in lump-like masses, we implanted collagen sponges containing BMP-2 and TGF-β1 with or without LPS into TNF-α- or IL-1α/β-deficient mice. LPS treatment reduced the volume of ectopic bones in TNF-α-deficient mice but not in IL-1α/β-deficient mice. Furthermore, collagen sponges containing IL-1β reduced ectopic bone formation by BMP-2 and TGF-β1 in wild-type mice to the same extent as LPS treatment did. CONCLUSIONS LPS suppresses the ectopic bone formation induced by BMP-2 and TGF-β1 through IL-1β production.
Collapse
|
5
|
Hwang CH, Keller J, Renner C, Ohta S, Wu DK. Genetic interactions support an inhibitory relationship between bone morphogenetic protein 2 and netrin 1 during semicircular canal formation. Development 2019; 146:dev.174748. [PMID: 30770380 PMCID: PMC6398446 DOI: 10.1242/dev.174748] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 01/18/2019] [Indexed: 12/16/2022]
Abstract
The semicircular canals of the mammalian inner ear are derived from epithelial pouches in which epithelial cells in the central region of each pouch undergo resorption, leaving behind the region at the rim to form a tube-shaped canal. Lack of proliferation at the rim and/or over-clearing of epithelial cells in the center of the pouch can obliterate canal formation. Otic-specific knockout of bone morphogenetic protein 2 (Bmp2) results in absence of all three semicircular canals; however, the common crus and ampullae housing the sensory tissue (crista) are intact. The lack of Bmp2 causes Ntn1 (which encodes netrin 1), which is required for canal resorption, to be ectopically expressed at the canal rim. Ectopic Ntn1 results in reduction of Dlx5 and Lmo4, which are required for rim formation. These phenotypes can be partially rescued by removing one allele of Ntn1 in the Bmp2 mutants, indicating that Bmp2 normally negatively regulates Ntn1 for canal formation. Additionally, non-resorption of the canal pouch in Ntn1−/− mutants is partially rescued by removing one allele of Bmp2. Thus, reciprocal inhibition between Bmp2 and netrin 1 is involved in canal formation of the vestibule. Summary:Bmp2-conditional mutant analyses support the hypothesis that presumptive crista induces canal genesis zone in the canal pouch to express Bmp2, which promotes canal formation by restricting Ntn1 expression to the resorption domain.
Collapse
Affiliation(s)
- Chan Ho Hwang
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, MD 20892, USA
| | - James Keller
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, MD 20892, USA
| | - Charles Renner
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, MD 20892, USA
| | - Sho Ohta
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, MD 20892, USA
| | - Doris K Wu
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, MD 20892, USA
| |
Collapse
|
6
|
Yang LM, Ornitz DM. Sculpting the skull through neurosensory epithelial-mesenchymal signaling. Dev Dyn 2018; 248:88-97. [PMID: 30117627 DOI: 10.1002/dvdy.24664] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/09/2018] [Accepted: 08/10/2018] [Indexed: 12/16/2022] Open
Abstract
The vertebrate skull is a complex structure housing the brain and specialized sensory organs, including the eye, the inner ear, and the olfactory system. The close association between bones of the skull and the sensory organs they encase has posed interesting developmental questions about how the tissues scale with one another. Mechanisms that regulate morphogenesis of the skull are hypothesized to originate in part from the encased neurosensory organs. Conversely, the developing skull is hypothesized to regulate the growth of neurosensory organs, through mechanical forces or molecular signaling. Here, we review studies of epithelial-mesenchymal interactions during inner ear and olfactory system development that may coordinate the growth of the two sensory organs with their surrounding bone. We highlight recent progress in the field and provide evidence that mechanical forces arising from bone growth may affect olfactory epithelium development. Developmental Dynamics 248:88-97, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lu M Yang
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri
| | - David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
7
|
Kumar A, Davies TG, Itasaki N. Developmental abnormalities of the otic capsule and inner ear following application of prolyl-hydroxylase inhibitors in chick embryos. Birth Defects Res 2018; 110:1194-1204. [PMID: 30079508 DOI: 10.1002/bdr2.1375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 06/30/2018] [Accepted: 07/08/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND Naturally hypoxic conditions in amniote embryos play important roles in normal development. We previously showed that a hypoxic condition is required to produce a sufficient amount of neural crest cells (NCCs) during embryogenesis and that promoting a hypoxic response by prolyl-hydroxylase (PHD) inhibitors increases NCCs. Given that PHD inhibitors are considered as a potential treatment for anemia and ischemic diseases, we investigated the phenotypic effect of PHD inhibitors on embryonic development. METHODS Chick embryos were administered with PHD inhibitors prior to the induction of NCCs on day 1.5. Three main events relating to hypoxia, NCCs induction, vasculogenesis and chondrogenesis, were examined. RESULTS PHD inhibitors caused an increase of Sox10-positive NCCs in vivo. Vasculogenesis was promoted temporarily, although rapid vasculogenesis diminished the effect by day 5 in cephalic and pharyngeal regions. Studies on chondrogenesis at day 7 showed advanced development of the otic capsule, a cartilaginous structure encapsulating the inner ear. Analysis by X-ray micro-computed-tomography (μCT) revealed smaller otic capsule, suggesting premature differentiation. This in turn, deformed the developing semicircular canals within it. Other skeletal structures such as the palate and jaw were unaffected. The localized effect on the otic capsule was considered a result of the multiple effects from the hypoxic responses, increased NCCs and promoted chondrogenesis. CONCLUSION Given the wide range of clinical applications being considered for PHD inhibitors, this study provides crucial information to caution and guide use of PHD inhibitors when treating women of childbearing age.
Collapse
Affiliation(s)
- Akshay Kumar
- Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Thomas G Davies
- School of Earth Sciences, University of Bristol, Bristol, United Kingdom
| | - Nobue Itasaki
- Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
8
|
Ma JY, You D, Li WY, Lu XL, Sun S, Li HW. Bone morphogenetic proteins and inner ear development. J Zhejiang Univ Sci B 2018; 20:131-145. [PMID: 30112880 DOI: 10.1631/jzus.b1800084] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bone morphogenetic proteins (BMPs) are the largest subfamily of the transforming growth factor-β superfamily, and they play important roles in the development of numerous organs, including the inner ear. The inner ear is a relatively small organ but has a highly complex structure and is involved in both hearing and balance. Here, we discuss BMPs and BMP signaling pathways and then focus on the role of BMP signal pathway regulation in the development of the inner ear and the implications this has for the treatment of human hearing loss and balance dysfunction.
Collapse
Affiliation(s)
- Jiao-Yao Ma
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Dan You
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Wen-Yan Li
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Xiao-Ling Lu
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Shan Sun
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Hua-Wei Li
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China.,Institutes of Biomedical Sciences and the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
9
|
Waqas M, Sun S, Xuan C, Fang Q, Zhang X, Islam IU, Qi J, Zhang S, Gao X, Tang M, Shi H, Li H, Chai R. Bone morphogenetic protein 4 promotes the survival and preserves the structure of flow-sorted Bhlhb5+ cochlear spiral ganglion neurons in vitro. Sci Rep 2017; 7:3506. [PMID: 28615657 PMCID: PMC5471210 DOI: 10.1038/s41598-017-03810-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/04/2017] [Indexed: 01/22/2023] Open
Abstract
SGNs are the primary auditory neurons, and damage or loss of SGNs leads to sensorineural hearing loss. BMP4 is a growth factor that belongs to the TGF-β superfamily and has been shown to play a key role during development, but little is known about its effect on postnatal cochlear SGNs in mice. In this study, we used the P3 Bhlhb5-cre/tdTomato transgenic mouse model and FACS to isolate a pure population of Bhlhb5+ SGNs. We found that BMP4 significantly promoted SGN survival after 7 days of culture. We observed fewer apoptotic cells and decreased expression of pro-apoptotic marker genes after BMP4 treatment. We also found that BMP4 promoted monopolar neurite outgrowth of isolated SGNs, and high concentrations of BMP4 preserved the number and the length of neurites in the explant culture of the modiolus harboring the SGNs. We showed that high concentration of BMP4 enhanced neurite growth as determined by the higher average number of filopodia and the larger area of the growth cone. Finally, we found that high concentrations of BMP4 significantly elevated the synapse density of SGNs in explant culture. Thus, our findings suggest that BMP4 has the potential to promote the survival and preserve the structure of SGNs.
Collapse
Affiliation(s)
- Muhammad Waqas
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Department of Biotechnology, Federal Urdu University of Arts, Science and Technology, Gulshan-e-Iqbal campus, Karachi, Pakistan
| | - Shan Sun
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China
| | - Chuanyin Xuan
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Qiaojun Fang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaoli Zhang
- Department of Otolaryngology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Irum-Us Islam
- Department of Biotechnology, Federal Urdu University of Arts, Science and Technology, Gulshan-e-Iqbal campus, Karachi, Pakistan
| | - Jieyu Qi
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Shasha Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xia Gao
- Department of Otolaryngology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Haibo Shi
- Department of Otorhinolaryngology Head & Neck Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200233, China.
| | - Huawei Li
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China. .,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China. .,Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China. .,Shanghai Engineering Research Centre of Cochlear Implants, Shanghai, 200031, China. .,The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China.
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China. .,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China. .,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
10
|
Magariños M, Pulido S, Aburto MR, de Iriarte Rodríguez R, Varela-Nieto I. Autophagy in the Vertebrate Inner Ear. Front Cell Dev Biol 2017; 5:56. [PMID: 28603711 PMCID: PMC5445191 DOI: 10.3389/fcell.2017.00056] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 05/09/2017] [Indexed: 12/13/2022] Open
Abstract
Autophagy is a conserved catabolic process that results in the lysosomal degradation of cell components. During development, autophagy is associated with tissue and organ remodeling, and under physiological conditions it is tightly regulated as it plays a housekeeping role in removing misfolded proteins and damaged organelles. The vertebrate inner ear is a complex sensory organ responsible for the perception of sound and for balance. Cell survival, death and proliferation, as well as cell fate specification and differentiation, are processes that are strictly coordinated during the development of the inner ear in order to generate the more than a dozen specialized cell types that constitute this structure. Here, we review the existing evidence that implicates autophagy in the generation of the vertebrate inner ear. At early stages of chicken otic development, inhibiting autophagy impairs neurogenesis and causes aberrant otocyst morphogenesis. Autophagy provides energy for the clearing of dying cells and it favors neuronal differentiation. Moreover, autophagy is required for proper vestibular development in the mouse inner ear. The autophagy-related genes Becn1, Atg4g, Atg5, and Atg9, are expressed in the inner ear from late developmental stages to adulthood, and Atg4b mutants show impaired vestibular behavior associated to defects in otoconial biogenesis that are also common to Atg5 mutants. Autophagic flux appears to be age-regulated, augmenting from perinatal stages to young adulthood in mice. This up-regulation is concomitant with the functional maturation of the hearing receptor. Hence, autophagy can be considered an intracellular pathway fundamental for in vertebrate inner ear development and maturation.
Collapse
Affiliation(s)
- Marta Magariños
- Department of Endocrine and Nervous Systems Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols," CSIC-UAMMadrid, Spain.,CIBERER, Unit 761, Instituto de Salud Carlos IIIMadrid, Spain.,Departamento de Biología, Universidad Autónoma de MadridMadrid, Spain
| | - Sara Pulido
- Department of Endocrine and Nervous Systems Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols," CSIC-UAMMadrid, Spain.,CIBERER, Unit 761, Instituto de Salud Carlos IIIMadrid, Spain
| | - María R Aburto
- Department of Endocrine and Nervous Systems Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols," CSIC-UAMMadrid, Spain
| | - Rocío de Iriarte Rodríguez
- Department of Endocrine and Nervous Systems Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols," CSIC-UAMMadrid, Spain
| | - Isabel Varela-Nieto
- Department of Endocrine and Nervous Systems Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols," CSIC-UAMMadrid, Spain.,CIBERER, Unit 761, Instituto de Salud Carlos IIIMadrid, Spain.,Instituto de Investigación Hospital Universitario La Paz (IdiPAZ)Madrid, Spain
| |
Collapse
|
11
|
Ohta S, Wang B, Mansour SL, Schoenwolf GC. BMP regulates regional gene expression in the dorsal otocyst through canonical and non-canonical intracellular pathways. Development 2016; 143:2228-37. [PMID: 27151948 DOI: 10.1242/dev.137133] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 04/27/2016] [Indexed: 12/13/2022]
Abstract
The inner ear consists of two otocyst-derived, structurally and functionally distinct components: the dorsal vestibular and ventral auditory compartments. BMP signaling is required to form the vestibular compartment, but how it complements other required signaling molecules and acts intracellularly is unknown. Using spatially and temporally controlled delivery of signaling pathway regulators to developing chick otocysts, we show that BMP signaling regulates the expression of Dlx5 and Hmx3, both of which encode transcription factors essential for vestibular formation. However, although BMP regulates Dlx5 through the canonical SMAD pathway, surprisingly, it regulates Hmx3 through a non-canonical pathway involving both an increase in cAMP-dependent protein kinase A activity and the GLI3R to GLI3A ratio. Thus, both canonical and non-canonical BMP signaling establish the precise spatiotemporal expression of Dlx5 and Hmx3 during dorsal vestibular development. The identification of the non-canonical pathway suggests an intersection point between BMP and SHH signaling, which is required for ventral auditory development.
Collapse
Affiliation(s)
- Sho Ohta
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84132-3401, USA
| | - Baolin Wang
- Department of Cell and Developmental Biology and Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Suzanne L Mansour
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84132-3401, USA Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112-5330, USA
| | - Gary C Schoenwolf
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84132-3401, USA
| |
Collapse
|
12
|
Kanaya E, Yamahara K, Okano T, Yoshida A, Katsuno T, Takebayashi H, Ito J, Yamamoto N. Expression of the Olig gene family in the developing mouse inner ear. Gene Expr Patterns 2015; 17:79-86. [PMID: 25778822 DOI: 10.1016/j.gep.2015.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 02/15/2015] [Accepted: 03/07/2015] [Indexed: 10/23/2022]
Abstract
Transcription factors are believed to play key roles in determining cell fate in inner ear development. Olig genes, which are basic helix-loop-helix transcription factors, have been reported to play important roles in the development of the central nervous system. However, members of this family have not previously been implicated in inner ear development, despite the similarity between otocyst and neural tube development. Olig1 begins to be expressed at the ventral domain of the otocyst at embryonic day (E) 9.5, and Olig1 expression in the epithelium of the developing inner ear persists to E15.5. Olig2 expression is localized to the cochleovestibular ganglia from E12.5 through E14.5. Olig3 has a diffuse expression pattern in the developing inner ear from E12.5 through the postnatal stage. Furthermore, at early stages of inner ear development, the Olig1 expression domain overlaps a region that is positive for Sox2 and Jagged1. This observation indicates that Olig1 may play an important role in the specification of the prosensory domain in the developing inner ear. As Olig genes are expressed in the mouse developing inner ear in a temporospatially distinct fashion, they may play substantial roles in the regulation of mammalian inner ear development.
Collapse
Affiliation(s)
- Eriko Kanaya
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo, Kyoto 606-8507, Japan
| | - Kohei Yamahara
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo, Kyoto 606-8507, Japan
| | - Takayuki Okano
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo, Kyoto 606-8507, Japan
| | - Atsuhiro Yoshida
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo, Kyoto 606-8507, Japan
| | - Tatsuya Katsuno
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo, Kyoto 606-8507, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Asahimachi Chuo-ku, Niigata 951-8510, Japan
| | - Juichi Ito
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo, Kyoto 606-8507, Japan
| | - Norio Yamamoto
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo, Kyoto 606-8507, Japan.
| |
Collapse
|
13
|
Nakajima Y. Signaling regulating inner ear development: cell fate determination, patterning, morphogenesis, and defects. Congenit Anom (Kyoto) 2015; 55:17-25. [PMID: 25040109 DOI: 10.1111/cga.12072] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 06/07/2014] [Indexed: 12/28/2022]
Abstract
The membranous labyrinth of the inner ear is a highly complex organ that detects sound and balance. Developmental defects in the inner ear cause congenital hearing loss and balance disorders. The membranous labyrinth consists of three semicircular ducts, the utricle, saccule, and endolymphatic ducts, and the cochlear duct. These complex structures develop from the simple otic placode, which is established in the cranial ectoderm adjacent to the neural crest at the level of the hindbrain at the early neurula stage. During development, the otic placode invaginates to form the otic vesicle, which subsequently gives rise to neurons for the vestibulocochlear ganglion, the non-sensory and sensory epithelia of the membranous labyrinth that includes three ampullary crests, two maculae, and the organ of Corti. Combined paracrine and autocrine signals including fibroblast growth factor, Wnt, retinoic acid, hedgehog, and bone morphogenetic protein regulate fate determination, axis formation, and morphogenesis in the developing inner ear. Juxtacrine signals mediated by Notch pathways play a role in establishing the sensory epithelium, which consists of mechanosensory hair cells and supporting cells. The highly differentiated organ of Corti, which consists of uniformly oriented inner/outer hair cells and specific supporting cells, develops during fetal development. Developmental alterations/arrest causes congenital malformations in the inner ear in a spatiotemporal-restricted manner. A clearer understanding of the mechanisms underlying inner ear development is important not only for the management of patients with congenital inner ear malformations, but also for the development of regenerative therapy for impaired function.
Collapse
Affiliation(s)
- Yuji Nakajima
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| |
Collapse
|
14
|
Kim HJ, Woo HM, Ryu J, Bok J, Kim JW, Choi SB, Park MH, Park HY, Koo SK. Conditional deletion of pten leads to defects in nerve innervation and neuronal survival in inner ear development. PLoS One 2013; 8:e55609. [PMID: 23393595 PMCID: PMC3564925 DOI: 10.1371/journal.pone.0055609] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 01/02/2013] [Indexed: 12/25/2022] Open
Abstract
All cellular phenomena and developmental events, including inner ear development, are modulated through harmonized signaling networks. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a tumor suppressor, is a major signaling component involved in cross talk with key regulators of development; i.e., Wnt, Notch, and bone morphogenetic proteins. Although Pten function has been studied in various systems, its role in inner ear development is poorly understood. Here, we used inner ear-specific Pten conditional knockout mice and examined the characteristics of the inner ear. In a detailed analysis of the phenotype, reduced cochlear turning and widened epithelia were observed. Phalloidin staining of sensory epithelium revealed that hair cell patterns were disturbed; i.e., additional rows of hair cells were discovered. The neural abnormality revealed a reduction in and disorganization of nerve fibers, including apoptosis at the neural precursor stage. Pten deficiency induced increased phosphorylation of Akt at Ser473. The elevation of inhibitory glycogen synthase kinase 3β Ser9 phosphorylation (pGSK3β) was sustained until the neuronal differentiation stage at embryonic day 14.5, instead of pGSK3β downregulation. This is the first report on the influence of Pten/Akt/GSK3β signaling on the development of spiral ganglia. These results suggest that Pten is required for the maintenance of neuroblast number, neural precursors, and differentiation in the inner ear.
Collapse
Affiliation(s)
- Hyung Jin Kim
- Center for Biomedical Sciences, National Institute of Health, Osong Health Technology Administration Complex 643, Yeonje-ri, Osong-eup, Cheongwon-gun, Chungcheongbuk-do, South Korea
| | - Hae-Mi Woo
- Center for Biomedical Sciences, National Institute of Health, Osong Health Technology Administration Complex 643, Yeonje-ri, Osong-eup, Cheongwon-gun, Chungcheongbuk-do, South Korea
| | - Jihee Ryu
- Center for Biomedical Sciences, National Institute of Health, Osong Health Technology Administration Complex 643, Yeonje-ri, Osong-eup, Cheongwon-gun, Chungcheongbuk-do, South Korea
| | - Jinwoong Bok
- Department of Anatomy, BK21 Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jin Woo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, South Korea
| | - Sang Back Choi
- Center for Biomedical Sciences, National Institute of Health, Osong Health Technology Administration Complex 643, Yeonje-ri, Osong-eup, Cheongwon-gun, Chungcheongbuk-do, South Korea
| | - Mi-Hyun Park
- Center for Biomedical Sciences, National Institute of Health, Osong Health Technology Administration Complex 643, Yeonje-ri, Osong-eup, Cheongwon-gun, Chungcheongbuk-do, South Korea
| | - Hyun-Young Park
- Center for Biomedical Sciences, National Institute of Health, Osong Health Technology Administration Complex 643, Yeonje-ri, Osong-eup, Cheongwon-gun, Chungcheongbuk-do, South Korea
| | - Soo Kyung Koo
- Center for Biomedical Sciences, National Institute of Health, Osong Health Technology Administration Complex 643, Yeonje-ri, Osong-eup, Cheongwon-gun, Chungcheongbuk-do, South Korea
- * E-mail:
| |
Collapse
|
15
|
Magariños M, Contreras J, Aburto MR, Varela-Nieto I. Early development of the vertebrate inner ear. Anat Rec (Hoboken) 2012; 295:1775-90. [PMID: 23044927 DOI: 10.1002/ar.22575] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 07/24/2012] [Indexed: 12/12/2022]
Abstract
This is a review of the biological processes and the main signaling pathways required to generate the different otic cell types, with particular emphasis on the actions of insulin-like growth factor I. The sensory organs responsible of hearing and balance have a common embryonic origin in the otic placode. Lineages of neural, sensory, and support cells are generated from common otic neuroepithelial progenitors. The sequential generation of the cell types that will form the adult inner ear requires the coordination of cell proliferation with cell differentiation programs, the strict regulation of cell survival, and the metabolic homeostasis of otic precursors. A network of intracellular signals operates to coordinate the transcriptional response to the extracellular input. Understanding the molecular clues that direct otic development is fundamental for the design of novel treatments for the protection and repair of hearing loss and balance disorders.
Collapse
Affiliation(s)
- Marta Magariños
- Instituto de Investigaciones Biomédicas, Alberto Sols, CSIC-UAM, Madrid, Spain
| | | | | | | |
Collapse
|
16
|
Delayed fusion and altered gene expression contribute to semicircular canal defects in Chd7 deficient mice. Mech Dev 2012; 129:308-23. [PMID: 22705977 DOI: 10.1016/j.mod.2012.06.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Revised: 05/30/2012] [Accepted: 06/07/2012] [Indexed: 11/20/2022]
Abstract
Proper morphogenesis of inner ear semicircular canals requires precise regulation of cellular proliferation, epithelial-to-mesenchymal transition, and fusion of epithelial plates. Epigenetic regulation of these processes is not well understood, but is likely to involve chromatin remodeling enzymes. CHD7 is a chromodomain-containing, ATP dependent helicase protein that is highly expressed in the developing ear and is required for semicircular canal development in both humans and mice. Here we report that mice with heterozygous loss of Chd7 function exhibit delayed semicircular canal genesis, delayed Netrin1 expression and disrupted expression of genes that are critical for semicircular canal formation (Bmp2, Bmp4, Msx1 and Fgf10). Complete loss of Chd7 results in aplasia of the semicircular canals and sensory vestibular organs, with reduced or absent expression of Otx1, Hmx3, Jagged1, Lmo4, Msx1 and Sox2. Our results suggest that Chd7 may have critical selector gene functions during inner ear morphogenesis. Detailed analysis of the epigenetic modifications underlying these gene expression changes should provide insights into semicircular canal development and help in the design of therapies for individuals with inner ear malformations.
Collapse
|
17
|
Sánchez-Guardado LÓ, Ferran JL, Rodríguez-Gallardo L, Puelles L, Hidalgo-Sánchez M. Meis gene expression patterns in the developing chicken inner ear. J Comp Neurol 2011; 519:125-47. [PMID: 21120931 DOI: 10.1002/cne.22508] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We are interested in stable gene network activities operating sequentially during inner ear specification. The implementation of this patterning process is a key event in the generation of functional subdivisions of the otic vesicle during early embryonic development. The vertebrate inner ear is a complex sensory structure that is a good model system for characterization of developmental mechanisms controlling patterning and specification. Meis genes, belonging to the TALE family, encode homodomain-containing transcription factors remarkably conserved during evolution, which play a role in normal and neoplastic development. To gain understanding of the possible role of homeobox Meis genes in the developing chick inner ear, we comprehensively analyzed their spatiotemporal expression patterns from early otic specification stages onwards. In the invaginating otic placode, Meis1/2 transcripts were observed in the borders of the otic cup, being absent in the portion of otic epithelium closest to the hindbrain. As development proceeds, Meis1 and Meis2 expressions became restricted to the dorsomedial otic epithelium. Both genes were strongly expressed in the entire presumptive domain of the semicircular canals, and more weakly in all associated cristae. The endolymphatic apparatus was labeled in part by Meis1/2. Meis1 was also expressed in the lateral wall of the growing cochlear duct, while Meis2 expression was detected in a few cells of the developing acoustic-vestibular ganglion. Our results suggest a possible role of Meis assigning regional identity in the morphogenesis, patterning, and specification of the developing inner ear.
Collapse
|
18
|
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: 3.6] [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
|
19
|
Robert-Moreno À, Naranjo S, de la Calle-Mustienes E, Gómez-Skarmeta JL, Alsina B. Characterization of new otic enhancers of the pou3f4 gene reveal distinct signaling pathway regulation and spatio-temporal patterns. PLoS One 2010; 5:e15907. [PMID: 21209840 PMCID: PMC3013142 DOI: 10.1371/journal.pone.0015907] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 11/26/2010] [Indexed: 02/01/2023] Open
Abstract
POU3F4 is a member of the POU-homedomain transcription factor family with a prominent role in inner ear development. Mutations in the human POU3F4 coding unit leads to X-linked deafness type 3 (DFN3), characterized by conductive hearing loss and progressive sensorineural deafness. Microdeletions found 1 Mb 5' upstream of the coding region also displayed the same phenotype, suggesting that cis-regulatory elements might be present in that region. Indeed, we and others have recently identified several enhancers at the 1 Mb 5' upstream interval of the pou3f4 locus. Here we characterize the spatio-temporal patterns of these regulatory elements in zebrafish transgenic lines. We show that the most distal enhancer (HCNR 81675) is activated earlier and drives GFP reporter expression initially to a broad ear domain to progressively restrict to the sensory patches. The proximal enhancer (HCNR 82478) is switched later during development and promotes expression, among in other tissues, in sensory patches from its onset. The third enhancer (HCNR 81728) is also active at later stages in the otic mesenchyme and in the otic epithelium. We also characterize the signaling pathways regulating these enhancers. While HCNR 81675 is regulated by very early signals of retinoic acid, HCNR 82478 is regulated by Fgf activity at a later stage and the HCNR 81728 enhancer is under the control of Hh signaling. Finally, we show that Sox2 and Pax2 transcription factors are bound to HCNR 81675 genomic region during otic development and specific mutations to these transcription factor binding sites abrogates HCNR 81675 enhancer activity. Altogether, our results suggest that pou3f4 expression in inner ear might be under the control of distinct regulatory elements that fine-tune the spatio-temporal activity of this gene and provides novel data on the signaling mechanisms controlling pou3f4 function.
Collapse
Affiliation(s)
- Àlex Robert-Moreno
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra/Parc de Recerca Biomèdica de Barcelona, Barcelona, Spain
| | - Silvia Naranjo
- Centro Andaluz de Biología del Desarrollo, CSIC/Universidad Pablo de Olavide, Sevilla, Spain
| | | | | | - Berta Alsina
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra/Parc de Recerca Biomèdica de Barcelona, Barcelona, Spain
| |
Collapse
|
20
|
Ohta S, Mansour SL, Schoenwolf GC. BMP/SMAD signaling regulates the cell behaviors that drive the initial dorsal-specific regional morphogenesis of the otocyst. Dev Biol 2010; 347:369-81. [PMID: 20837004 PMCID: PMC2962452 DOI: 10.1016/j.ydbio.2010.09.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Revised: 09/02/2010] [Accepted: 09/02/2010] [Indexed: 12/20/2022]
Abstract
During development of the otocyst, regional morphogenesis establishes a dorsal vestibular chamber and a ventral auditory chamber, which collectively constitute the membranous labyrinth of the inner ear. We identified the earliest morphogenetic event heralding the formation of the vestibular chamber, a rapid thinning and expansion of the dorsolateral wall of the otocyst, and showed that this process is generated by changes in otocyst cell shape from columnar to squamous, as opposed to changes in other cell behaviors, such as localized changes in cell proliferation or cell death. Moreover, we showed that thinning and expansion of the dorsolateral otocyst is regulated by BMP/SMAD signaling, which is both sufficient and necessary for localized thinning and expansion. Finally, we showed that BMP/SMAD signaling causes fragmentation of E-cadherin in the dorsolateral otocyst, occurring concomitantly with cell shape change, suggesting that BMP/SMAD signaling regulates cell-cell adhesion during the initial morphogenesis of the otocyst epithelium. Collectively, our results show that BMP signaling via SMADs regulates the cell behaviors that drive the initial dorsal-specific morphogenesis of the otocyst, providing new information about how regional morphogenesis of a complex organ rudiment, the developing membranous labyrinth, is initiated.
Collapse
Affiliation(s)
- Sho Ohta
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Suzanne L. Mansour
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Gary C. Schoenwolf
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
21
|
Ohyama T, Basch ML, Mishina Y, Lyons KM, Segil N, Groves AK. BMP signaling is necessary for patterning the sensory and nonsensory regions of the developing mammalian cochlea. J Neurosci 2010; 30:15044-51. [PMID: 21068310 PMCID: PMC3074492 DOI: 10.1523/jneurosci.3547-10.2010] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Revised: 08/27/2010] [Accepted: 08/28/2010] [Indexed: 01/04/2023] Open
Abstract
The mammalian inner ear detects sound with the organ of Corti, an intricately patterned region of the cochlea in which one row of inner hair cells and three rows of outer hair cells are surrounded by specialized supporting cells. The organ of Corti derives from a prosensory domain that runs the length of the cochlear duct and is bounded by two nonsensory domains, Kölliker's organ on the neural side and the outer sulcus on the abneural side. Although much progress has been made in identifying the signals regulating organ of Corti induction and differentiation, less is known about the mechanisms that establish sensory and nonsensory territories in the cochlear duct. Here, we show that a gradient of bone morphogenetic protein (BMP) signaling is established in the abneural-neural axis of the cochlea. Analysis of compound mutants of Alk3/6 type I BMP receptors shows that BMP signaling is necessary for specification of the prosensory domain destined to form the organ of Corti. Reduction of BMP signaling in Alk3/6 compound mutants eliminates both the future outer sulcus and the prosensory domain, with all cells expressing markers of Kölliker's organ. BMP4 upregulates markers of the future outer sulcus and downregulates marker genes of Kölliker's organ in cochlear organ cultures in a dose-dependent manner. Our results suggest BMP signaling is required for patterning sensory and nonsensory tissue in the mammalian cochlea.
Collapse
Affiliation(s)
- Takahiro Ohyama
- Division of Cell Biology and Genetics, House Ear Institute, Los Angeles, California 90057, USA.
| | | | | | | | | | | |
Collapse
|
22
|
Tachi K, Takami M, Sato H, Mochizuki A, Zhao B, Miyamoto Y, Tsukasaki H, Inoue T, Shintani S, Koike T, Honda Y, Suzuki O, Baba K, Kamijo R. Enhancement of bone morphogenetic protein-2-induced ectopic bone formation by transforming growth factor-β1. Tissue Eng Part A 2010; 17:597-606. [PMID: 20874259 DOI: 10.1089/ten.tea.2010.0094] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) possess osteoinductive activities and are useful for clinical treatments, including bone regeneration. We found that transforming growth factor (TGF)-β1 strongly enhances the osteoinductive activity of BMP-2. Collagen sponges containing 5 μg of BMP-2 were implanted into mouse muscle tissues, after which lump-like masses appeared and grew until day 7. Subsequently, calcification occurred in the lump-like masses by day 14. Addition of 50 ng of TGF-β1 to the BMP-2-containing sponges markedly accelerated the growth of the lump-like masses and resulted in a fivefold increase in total bone volume as compared with BMP-2 alone. The number of osteoblasts in ectopic bone tissues at 14 days after implantation induced by BMP-2+TGF-β1 was twofold greater than that with BMP-2 alone, whereas the number of osteoclasts was decreased by half. On the other hand, TGF-β1 accelerated the differentiation of both osteoblasts and osteoclasts in the early stage (2-7 days after implantation) of ectopic bone formation. We also implanted collagen sponges into bone defects surgically created in mouse calvaria. Sponges containing 2.5 μg of BMP-2 and 25 ng of TGF-β1 caused complete filling of the defects with orthotopic bone, whereas those containing 2.5 μg of BMP-2 alone caused only partial filling. These results suggest that TGF-β1 enhances BMP-2-induced ectopic bone formation by accelerating the growth of lump-like masses, and regulates osteoblast and osteoclast generation. Our findings may contribute to the development of a new treatment method for skeletal disorders.
Collapse
Affiliation(s)
- Keita Tachi
- Department of Biochemistry, School of Dentistry, Showa University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Id gene regulation and function in the prosensory domains of the chicken inner ear: a link between Bmp signaling and Atoh1. J Neurosci 2010; 30:11426-34. [PMID: 20739564 DOI: 10.1523/jneurosci.2570-10.2010] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Bone morphogenetic proteins (Bmps) regulate the expression of the proneural gene Atoh1 and the generation of hair cells in the developing inner ear. The present work explored the role of Inhibitor of Differentiation genes (Id1-3) in this process. The results show that Id genes are expressed in the prosensory domains of the otic vesicle, along with Bmp4 and Bmp7. Those domains exhibit high levels of the phosphorylated form of Bmp-responding R-Smads (P-Smad1,5,8), and of Bmp-dependent Smad transcriptional activity as shown by the BRE-tk-EGFP reporter. Increased Bmp signaling induces the expression of Id1-3 along with the inhibition of Atoh1. Conversely, the Bmp antagonist Noggin or the Bmp-receptor inhibitor Dorsomorphin elicit opposite effects, indicating that Bmp signaling is necessary for Id expression and Atoh1 regulation in the otocyst. The forced expression of Id3 is sufficient to reduce Atoh1 expression and to prevent the expression of hair cell differentiation markers. Together, these results suggest that Ids are part of the machinery that mediates the regulation of hair cell differentiation exerted by Bmps. In agreement with that, during hair cell differentiation Bmp4 expression, P-Smad1,5,8 levels and Id expression are downregulated from hair cells. However, Ids are also downregulated from the supporting cells which contrarily to hair cells exhibit high levels of Bmp4 expression, P-Smad1,5,8, and BRE-tk-EGFP activity, suggesting that in these cells Ids escape from Bmp/Smad signaling. The differential regulation of Ids in time and space may underlie the multiple functions of Bmp signaling during sensory organ development.
Collapse
|
24
|
Stankovic KM, Adachi O, Tsuji K, Kristiansen AG, Adams JC, Rosen V, McKenna MJ. Differences in gene expression between the otic capsule and other bones. Hear Res 2010; 265:83-9. [PMID: 20146935 PMCID: PMC3586807 DOI: 10.1016/j.heares.2010.02.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2009] [Revised: 01/20/2010] [Accepted: 02/04/2010] [Indexed: 10/19/2022]
Abstract
Our long term goal is to understand the molecular pathology of otosclerosis and to develop better forms of therapy. Toward this goal, the current study focused on characterizing the molecular factors responsible for the unique biological features of the otic capsule: its minimal rate of remodeling, and lack of healing capacity when fractured. We compared expression levels of 62 genes involved in bone metabolism between the adult murine otic capsule and the tibia and parietal bones; the latter exemplify bones formed by endochondral and intramembranous ossification, respectively. Gene expression levels were measured using real-time quantitative RT-PCR and analyzed using tools of bioinformatics. Expression patterns of key genes were verified with in situ hybridization. The molecular profile of the otic capsule was distinctly different from that of the tibia and parietal bone. Genes found to be most characteristic of the otic capsule were: osteoprotegerin (opg), bone morphogenetic protein receptor 1b (bmpr1b) and bone morphogenetic protein 3 (bmp3). Expression levels were high for opg and bmpr1b, and minimal for bmp3 within the otic capsule. We concluded that opg and bmpr1b likely play important roles in inhibition of remodeling within the otic capsule.
Collapse
Affiliation(s)
- Konstantina M. Stankovic
- Eaton Peabody Laboratory and Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
- Department of Otology and Laryngology, Harvard Medical School, Boston, Massachusetts, USA
| | - Osamu Adachi
- Eaton Peabody Laboratory and Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
- Department of Otology and Laryngology, Harvard Medical School, Boston, Massachusetts, USA
| | - Kunikazu Tsuji
- Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Arthur G. Kristiansen
- Eaton Peabody Laboratory and Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| | - Joe C. Adams
- Eaton Peabody Laboratory and Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
- Department of Otology and Laryngology, Harvard Medical School, Boston, Massachusetts, USA
| | - Vicki Rosen
- Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Michael J. McKenna
- Eaton Peabody Laboratory and Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
- Department of Otology and Laryngology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
25
|
|
26
|
Yang SM, Hou ZH, Yang G, Zhang JS, Hu YY, Sun JH, Guo WW, He DZZ, Han DY, Young WY, Yang X. Chondrocyte-specific Smad4 gene conditional knockout results in hearing loss and inner ear malformation in mice. Dev Dyn 2009; 238:1897-908. [PMID: 19582869 DOI: 10.1002/dvdy.22014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Smad4 is the central intracellular mediator of transforming growth factor-beta (TGF-beta) signaling, which plays crucial roles in tissue regeneration, cell differentiation, embryonic development, and regulation of the immune system. Conventional Smad4 gene knockout results in embryonic lethality, precluding its use in studies of the role of Smad4 in inner ear development. We used chondrocyte-specific Smad4 knockout mice (Smad4Co/Co) to investigate the function of Smad4 in inner ear development. Smad4Co/Co mice were characterized by a smaller cochlear volume, bone malformation, and abnormalities of the osseous spiral lamina and basilar membrane. The development of the hair cells was also abnormal, as evidenced by the disorganized stereocilia and reduced density of the neuronal processes beneath the hair cells. Auditory function tests revealed the homozygous Smad4Co/Co mice suffered from severe sensorineural hearing loss. Our results suggest that Smad4 is required for inner ear development and normal auditory function in mammals.
Collapse
Affiliation(s)
- Shi-ming Yang
- Department of Otolaryngology, Head and Neck Surgery, Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Soukup GA. Little but loud: small RNAs have a resounding affect on ear development. Brain Res 2009; 1277:104-14. [PMID: 19245798 PMCID: PMC2700218 DOI: 10.1016/j.brainres.2009.02.027] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Revised: 02/10/2009] [Accepted: 02/11/2009] [Indexed: 02/07/2023]
Abstract
The impact of small RNA function has resonated throughout nearly every aspect of eukaryotic biology and captured the varied interests of researchers, whether they are endeavoring to understand the basis of development and disease or seeking novel therapeutic targets and tools. The genetic regulatory roles of microRNAs (miRNAs) are particularly interesting given that these often highly conserved factors post-transcriptionally silence many complementary target genes by inhibiting messenger RNA translation. In this regard, miRNAs can be considered as counterparts to transcription factors, the ensemble of which establishes the set of expressed genes that define the characteristics of a specific cell type. In this review, evidence supporting a resounding role for small RNAs in development and maturation of sensory epithelia in the mouse inner ear will be considered with an emphasis on the contribution of one hair cell miRNA family (miR-183, miR-96, and miR-182). Although there is much yet to be explored in this fledgling aspect of ear biology, the breadth of miRNA expression and functional requirement for ear development are already sounding off.
Collapse
Affiliation(s)
- Garrett A Soukup
- Department of Biomedical Sciences, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE 68178, USA.
| |
Collapse
|
28
|
Braunstein EM, Monks DC, Aggarwal VS, Arnold JS, Morrow BE. Tbx1 and Brn4 regulate retinoic acid metabolic genes during cochlear morphogenesis. BMC DEVELOPMENTAL BIOLOGY 2009; 9:31. [PMID: 19476657 PMCID: PMC2700094 DOI: 10.1186/1471-213x-9-31] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Accepted: 05/29/2009] [Indexed: 11/10/2022]
Abstract
BACKGROUND In vertebrates, the inner ear is comprised of the cochlea and vestibular system, which develop from the otic vesicle. This process is regulated via inductive interactions from surrounding tissues. Tbx1, the gene responsible for velo-cardio-facial syndrome/DiGeorge syndrome in humans, is required for ear development in mice. Tbx1 is expressed in the otic epithelium and adjacent periotic mesenchyme (POM), and both of these domains are required for inner ear formation. To study the function of Tbx1 in the POM, we have conditionally inactivated Tbx1 in the mesoderm while keeping expression in the otic vesicle intact. RESULTS Conditional mutants (TCre-KO) displayed malformed inner ears, including a hypoplastic otic vesicle and a severely shortened cochlear duct, indicating that Tbx1 expression in the POM is necessary for proper inner ear formation. Expression of the mesenchyme marker Brn4 was also lost in the TCre-KO. Brn4-;Tbx1+/-embryos displayed defects in growth of the distal cochlea. To identify a potential signal from the POM to the otic epithelium, expression of retinoic acid (RA) catabolizing genes was examined in both mutants. Cyp26a1 expression was altered in the TCre-KO, while Cyp26c1 showed reduced expression in both TCre-KO and Brn4-;Tbx1+/- embryos. CONCLUSION These results indicate that Tbx1 expression in the POM regulates cochlear outgrowth potentially via control of local retinoic acid activity.
Collapse
Affiliation(s)
- Evan M Braunstein
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, USA.
| | | | | | | | | |
Collapse
|
29
|
Petko JA, Kabbani N, Frey C, Woll M, Hickey K, Craig M, Canfield VA, Levenson R. Proteomic and functional analysis of NCS-1 binding proteins reveals novel signaling pathways required for inner ear development in zebrafish. BMC Neurosci 2009; 10:27. [PMID: 19320994 PMCID: PMC2679751 DOI: 10.1186/1471-2202-10-27] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Accepted: 03/25/2009] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The semicircular canals, a subdivision of the vestibular system of the vertebrate inner ear, function as sensors of angular acceleration. Little is currently known, however, regarding the underlying molecular mechanisms that govern the development of this intricate structure. Zebrafish represent a particularly tractable model system for the study of inner ear development. This is because the ear can be easily visualized during early embryogenesis, and both forward and reverse genetic techniques are available that can be applied to the discovery of novel genes that contribute to proper ear development. We have previously shown that in zebrafish, the calcium sensing molecule neuronal calcium sensor-1 (NCS-1) is required for semicircular canal formation. The function of NCS-1 in regulating semicircular canal formation has not yet been elucidated. RESULTS We initiated a multistep functional proteomic strategy to identify neuronal calcium sensor-1 (NCS-1) binding partners (NBPs) that contribute to inner ear development in zebrafish. By performing a Y2H screen in combination with literature and database searches, we identified 10 human NBPs. BLAST searches of the zebrafish EST and genomic databases allowed us to clone zebrafish orthologs of each of the human NBPs. By investigating the expression profiles of zebrafish NBP mRNAs, we identified seven that were expressed in the developing inner ear and overlapped with the ncs-1a expression profile. GST pulldown experiments confirmed that selected NBPs interacted with NCS-1, while morpholino-mediated knockdown experiments demonstrated an essential role for arf1, pi4kbeta, dan, and pink1 in semicircular canal formation. CONCLUSION Based on their functional profiles, the hypothesis is presented that Ncs-1a/Pi4kbeta/Arf1 form a signaling pathway that regulates secretion of molecular components, including Dan and Bmp4, that are required for development of the vestibular apparatus. A second set of NBPs, consisting of Pink1, Hint2, and Slc25a25, are destined for localization in mitochondria. Our findings reveal a novel signalling pathway involved in development of the semicircular canal system, and suggest a previously unrecognized role for NCS-1 in mitochondrial function via its association with several mitochondrial proteins.
Collapse
Affiliation(s)
- Jessica A Petko
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey PA 17033, USA.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Volkenstein S, Brors D, Hansen S, Minovi A, Laub M, Jennissen HP, Dazert S, Neumann A. Influence of bone morphogenetic protein-2 on spiral ganglion neurite growth in vitro. Eur Arch Otorhinolaryngol 2009; 266:1381-9. [DOI: 10.1007/s00405-009-0930-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Accepted: 02/17/2009] [Indexed: 11/28/2022]
|
31
|
Hammond KL, Loynes HE, Mowbray C, Runke G, Hammerschmidt M, Mullins MC, Hildreth V, Chaudhry B, Whitfield TT. A late role for bmp2b in the morphogenesis of semicircular canal ducts in the zebrafish inner ear. PLoS One 2009; 4:e4368. [PMID: 19190757 PMCID: PMC2629815 DOI: 10.1371/journal.pone.0004368] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Accepted: 12/18/2008] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The Bone Morphogenetic Protein (BMP) genes bmp2 and bmp4 are expressed in highly conserved patterns in the developing vertebrate inner ear. It has, however, proved difficult to elucidate the function of BMPs during ear development as mutations in these genes cause early embryonic lethality. Previous studies using conditional approaches in mouse and chicken have shown that Bmp4 has a role in semicircular canal and crista development, but there is currently no direct evidence for the role of Bmp2 in the developing inner ear. METHODOLOGY/PRINCIPAL FINDINGS We have used an RNA rescue strategy to test the role of bmp2b in the zebrafish inner ear directly. Injection of bmp2b or smad5 mRNA into homozygous mutant swirl (bmp2b(-/-)) embryos rescues the early patterning defects in these mutants and the fish survive to adulthood. As injected RNA will only last, at most, for the first few days of embryogenesis, all later development occurs in the absence of bmp2b function. Although rescued swirl adult fish are viable, they have balance defects suggestive of vestibular dysfunction. Analysis of the inner ears of these fish reveals a total absence of semicircular canal ducts, structures involved in the detection of angular motion. All other regions of the ear, including the ampullae and cristae, are present and appear normal. Early stages of otic development in rescued swirl embryos are also normal. CONCLUSIONS/SIGNIFICANCE Our findings demonstrate a critical late role for bmp2b in the morphogenesis of semicircular canals in the zebrafish inner ear. This is the first demonstration of a developmental role for any gene during post-embryonic stages of otic morphogenesis in the zebrafish. Despite differences in the early stages of semicircular canal formation between zebrafish and amniotes, the role of Bmp2 in semicircular canal duct outgrowth is likely to be conserved between different vertebrate species.
Collapse
Affiliation(s)
- Katherine L. Hammond
- MRC Centre for Developmental and Biomedical Genetics and Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Helen E. Loynes
- MRC Centre for Developmental and Biomedical Genetics and Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Catriona Mowbray
- MRC Centre for Developmental and Biomedical Genetics and Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Greg Runke
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | | | - Mary C. Mullins
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Victoria Hildreth
- Institute of Human Genetics, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Bill Chaudhry
- Institute of Human Genetics, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Tanya T. Whitfield
- MRC Centre for Developmental and Biomedical Genetics and Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
32
|
Liu W, Li L, Li G, Garritano F, Shanske A, Frenz DA. Coordinated molecular control of otic capsule differentiation: functional role of Wnt5a signaling and opposition by sfrp3 activity. Growth Factors 2008; 26:343-54. [PMID: 18991062 DOI: 10.1080/08977190802442013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Wnt proteins constitute one of the major families of secreted ligands that function in developmental signaling, however, little is known of the role of Wnt5a during inner ear development. It is hypothesized that Wnt5a acts as a mediator of chondrogenesis in the developing otic capsule, a cartilaginous structure that surrounds the developing inner ear and presages the formation of the endochondral bony labyrinth. We report the pattern of expression of Wnt5a protein and mRNA in the developing mouse inner ear using immunohistochemistry, whole-mount in situ hybridization and RT-PCR, and the ability of exogenous Wnt5a to stimulate otic capsule chondrogenesis when added to high-density cultures of periotic mesenchyme containing otic epithelium (periotic mesenchyme + otic epithelium), a well-established model of otic capsule formation. We show that in the presence of secreted frizzled related protein 3 (sfrp3), a Wnt antagonist expressed in the developing inner ear, or Wnt5a-specific antisense oligonucleotide, which diminishes endogenous Wnt5a, otic capsule chondrogenesis is suppressed in culture. We determined by histological analysis and aggrecan immunoreactivity that chondrogenic differentiation is disturbed in Wnt5a null embryos, and provide evidence that the periotic mesenchyme + otic epithelium harvested from Wnt5a null mice is compromised in its ability to differentiate into cartilage when interacted in culture. We propose a model whereby sfrp3 and Wnt5a act antagonistically to ensure appropriate patterns of chondrogenesis and provide coordinated control of otic capsule formation. Our findings support Wnt5a and sfrp3 as regulators of otic capsule formation in the developing mouse inner ear.
Collapse
Affiliation(s)
- Wei Liu
- Department of Otorhinolaryngology-Head & Neck Surgery, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | | | |
Collapse
|
33
|
Shawi M, Serluca FC. Identification of a BMP7 homolog in zebrafish expressed in developing organ systems. Gene Expr Patterns 2008; 8:369-375. [PMID: 18602348 DOI: 10.1016/j.gep.2008.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2008] [Revised: 05/20/2008] [Accepted: 05/21/2008] [Indexed: 11/29/2022]
Abstract
The Bone morphogenetic proteins (BMPs) act in many key regulatory processes during development, including dorsoventral axis specification and organ development and are part of a conserved signal pathway. Specifically, BMP7 is a vital signaling molecule for normal development in the mammalian system. The zebrafish mutant snailhouse (snh) was originally isolated as being strongly dorsalized and the mutation was determined to lie within the bmp7 gene. We report here the cloning and expression of a second bmp7 homolog, which we term bmp7b. Sequence alignments show that bmp7b is more closely related to human, mouse and non-mammalian BMP7 than is snh. We further show that bmp7b is strongly expressed in developing organ systems such as the eyes, the ears, the pronephric kidney and the gastrointestinal system.
Collapse
Affiliation(s)
- May Shawi
- Developmental and Molecular Pathways, The Novartis Institutes for Biomedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | | |
Collapse
|
34
|
Chang W, Lin Z, Kulessa H, Hebert J, Hogan BLM, Wu DK. Bmp4 is essential for the formation of the vestibular apparatus that detects angular head movements. PLoS Genet 2008; 4:e1000050. [PMID: 18404215 PMCID: PMC2274953 DOI: 10.1371/journal.pgen.1000050] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Accepted: 03/11/2008] [Indexed: 12/30/2022] Open
Abstract
Angular head movements in vertebrates are detected by the three semicircular canals of the inner ear and their associated sensory tissues, the cristae. Bone morphogenetic protein 4 (Bmp4), a member of the Transforming growth factor family (TGF-β), is conservatively expressed in the developing cristae in several species, including zebrafish, frog, chicken, and mouse. Using mouse models in which Bmp4 is conditionally deleted within the inner ear, as well as chicken models in which Bmp signaling is knocked down specifically in the cristae, we show that Bmp4 is essential for the formation of all three cristae and their associated canals. Our results indicate that Bmp4 does not mediate the formation of sensory hair and supporting cells within the cristae by directly regulating genes required for prosensory development in the inner ear such as Serrate1 (Jagged1 in mouse), Fgf10, and Sox2. Instead, Bmp4 most likely mediates crista formation by regulating Lmo4 and Msx1 in the sensory region and Gata3, p75Ngfr, and Lmo4 in the non-sensory region of the crista, the septum cruciatum. In the canals, Bmp2 and Dlx5 are regulated by Bmp4, either directly or indirectly. Mechanisms involved in the formation of sensory organs of the vertebrate inner ear are thought to be analogous to those regulating sensory bristle formation in Drosophila. Our results suggest that, in comparison to sensory bristles, crista formation within the inner ear requires an additional step of sensory and non-sensory fate specification. Disruption of the sense of balance is highly debilitating, causing vertigo and nausea. Maintenance of proper balance requires sensory inputs from many body parts, including the inner ears and the eyes. Within the inner ear, the vestibular apparatus plays a key role in the sense of balance and is responsible for detecting head orientation and movements. The portion of the vestibular apparatus that detects angular head movements consists of three fluid-filled, semicircular canals oriented at right angles to each other. At one end of each canal is an enlargement that houses the sensory tissue, crista ampullaris, consisting of sensory hair cells and supporting cells. Bone morphogenetic protein 4 (Bmp4), a secreted signaling molecule, is expressed in these sensory regions during development. However, the lack of Bmp4 in mice affects the formation of not only the sensory regions but also their associated canals. These results demonstrate for the first time that a single gene, Bmp4, is required for the formation of the entire sensory apparatus for detecting angular head movements.
Collapse
Affiliation(s)
- Weise Chang
- National Institute on Deafness and Other Communication Disorders, NIH, Rockville, Maryland, United States of America
| | - Zhengshi Lin
- National Institute on Deafness and Other Communication Disorders, NIH, Rockville, Maryland, United States of America
| | - Holger Kulessa
- National Institute on Deafness and Other Communication Disorders, NIH, Rockville, Maryland, United States of America
- Department of Neuroscience, Albert Einstein College of Medicine, New York, New York, United States of America
- Department of Molecular Genetics, Albert Einstein College of Medicine, New York, New York, United States of America
- Department of Cell Biology, Duke University Medical Centre, Durham, North Carolina, United States of America
| | - Jean Hebert
- Department of Neuroscience, Albert Einstein College of Medicine, New York, New York, United States of America
- Department of Molecular Genetics, Albert Einstein College of Medicine, New York, New York, United States of America
| | - Brigid L. M. Hogan
- Department of Cell Biology, Duke University Medical Centre, Durham, North Carolina, United States of America
| | - Doris K. Wu
- National Institute on Deafness and Other Communication Disorders, NIH, Rockville, Maryland, United States of America
- * E-mail:
| |
Collapse
|
35
|
Cooperative function of Tbx1 and Brn4 in the periotic mesenchyme is necessary for cochlea formation. J Assoc Res Otolaryngol 2008; 9:33-43. [PMID: 18231833 DOI: 10.1007/s10162-008-0110-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Accepted: 01/03/2008] [Indexed: 10/22/2022] Open
Abstract
The T-box transcription factor TBX1 has been identified as the major gene responsible for the etiology of velocardiofacial syndrome/DiGeorge syndrome (VCFS/DGS). Conductive hearing loss occurs in a majority of patients with this syndrome, while sensorineural deafness has also been reported in some cases. Mutations in POU3F4/BRN4, a POU domain transcription factor, cause DFN3, an X-linked nonsyndromic form of deafness characterized by mixed conductive and sensorineural hearing loss. Inactivation of the murine orthologues of these genes causes similar defects to those seen in humans and has provided excellent models for the study of inner ear development. Tbx1 and Brn4 are expressed in the mesenchymal cells surrounding the otic vesicle and have been shown to play roles in cochlear outgrowth. Furthermore, expression of Brn4 is reduced in Tbx1 null mutants, suggesting a possible genetic interaction between these genes. To test whether Tbx1 and Brn4 function in a common pathway, mice mutant for both genes were generated and analyzed for inner ear defects. Brn4-;Tbx1+/- mutants displayed a significant reduction in the number of turns of the cochlea compared to Brn4- or Tbx1+/- mice. In addition, Brn4-;Tbx1+/- mice displayed structural defects in the apical cochlea indicative of Mondini dysplasia found in patients with either VCFS/DGS or DFN3. These data establish a genetic interaction between Tbx1 and Brn4 relevant to human disease and indicate a function of these genes in signaling from the periotic mesenchyme to the otic vesicle to direct proper coiling of the cochlear duct.
Collapse
|
36
|
Bok J, Brunet LJ, Howard O, Burton Q, Wu DK. Role of hindbrain in inner ear morphogenesis: analysis of Noggin knockout mice. Dev Biol 2007; 311:69-78. [PMID: 17900554 PMCID: PMC2215324 DOI: 10.1016/j.ydbio.2007.08.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2006] [Revised: 08/02/2007] [Accepted: 08/06/2007] [Indexed: 11/17/2022]
Abstract
Signaling from rhombomeres 5 and 6 of the hindbrain is thought to be important for inner ear patterning. In Noggin -/- embryos, the gross anatomy of the inner ear is distorted and malformed, with cochlear duct outgrowth and coiling most affected. We attributed these defects to a caudal shift of the rhombomeres caused by the shortened body axis and the kink in the neural tube. To test the hypothesis that a caudal shift of the rhombomeres affects inner ear development, we surgically generated chicken embryos in which rhombomeres 5 and 6 were similarly shifted relative to the position of the inner ears, as in Noggin mutants. All chicken embryos with shifted rhombomeres showed defects in cochlear duct formation indicating that signaling from rhombomeres 5 and 6 is important for cochlear duct patterning in both chicken and mice. In addition, the size of the otic capsule is increased in Noggin -/- mutants, which most likely is due to unopposed BMP signaling for chondrogenesis in the peri-otic mesenchyme.
Collapse
Affiliation(s)
- Jinwoong Bok
- National Institute on Deafness and Other Communication Disorders, Rockville, MD 20850
| | - Lisa J. Brunet
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Omar Howard
- National Institute on Deafness and Other Communication Disorders, Rockville, MD 20850
| | - Quianna Burton
- National Institute on Deafness and Other Communication Disorders, Rockville, MD 20850
| | - Doris K. Wu
- National Institute on Deafness and Other Communication Disorders, Rockville, MD 20850
| |
Collapse
|
37
|
Omata Y, Nojima Y, Nakayama S, Okamoto H, Nakamura H, Funahashi JI. Role of Bone morphogenetic protein 4 in zebrafish semicircular canal development. Dev Growth Differ 2007; 49:711-9. [PMID: 17908182 DOI: 10.1111/j.1440-169x.2007.00964.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Bone morphogenetic proteins (BMPs) are known to play roles in inner ear development of higher vertebrates. In zebrafish, there are several reports showing that members of the BMP family are expressed in the otic vesicle. We have isolated a novel zebrafish mutant gallery, which affects the development of the semicircular canal. Gallery merely forms the lateral and the immature anterior protrusion, and does not form posterior and ventral protrusions. We found that the expression of bmp2b and bmp4, both expressed in the normal optic vesicle at the protrusion stage, are extremely upregulated in the otic vesicle of gallery. To elucidate the role of BMPs in the development of the inner ear of zebrafish, we have applied excess BMP to the wild-type otic vesicle. The formation of protrusions was severely affected, and in some cases, they were completely lost in BMP4-treated embryos. Furthermore, the protrusions in gallery treated with Noggin were partially rescued. These data indicate that BMP4 plays an important role in the development of protrusions to form semicircular canals.
Collapse
Affiliation(s)
- Yasuhiro Omata
- Department of Molecular Neurobiology, Institute of Development, Aging and Cancer, Tohoku University, Seiryou-machi 4-1, Sendai 980-8575, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Stankovic KM, Kristiansen AG, Bizaki A, Lister M, Adams JC, McKenna MJ. Studies of Otic Capsule Morphology and Gene Expression in the Mov13 Mouse – An Animal Model of Type I Osteogenesis Imperfecta. Audiol Neurootol 2007; 12:334-43. [PMID: 17595534 DOI: 10.1159/000104789] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2006] [Accepted: 03/28/2007] [Indexed: 11/19/2022] Open
Abstract
Type I osteogenesis imperfecta (OI) is a disorder of skeletal bones characterized by bone fragility and blue sclera, which can result from mutations in genes encoding for type I collagen--the COL1A1 and COL1A2 genes. Fifty percent of patients with type I OI develop hearing loss and associated histopathological changes in the otic capsule that are indistinguishable from otosclerosis, a major cause of acquired hearing loss. In an attempt to elucidate molecular and cellular mechanisms of hearing loss in type I OI, we have studied the Mov13 mouse, which has served as an animal model of type I OI by virtue of exhibiting variable transcriptional block of the COL1A1 gene. We studied the morphometry of the Mov13 otic capsule and compared expression levels of 60 genes in the otic capsule with those in the tibia and parietal bone of the Mov13 and wild-type mice. The degree of transcriptional block of the COL1A1 gene and its downstream effects differed significantly between the bones examined. We found that expression levels of bone morphogenetic protein 3 and nuclear factor kappa-B1 best distinguished Mov13 otic capsule from wild-type otic capsule, and that osteoprotegerin, caspase recruitment domain containing protein 1, and partitioning defective protein 3 best distinguished Mov13 otic capsule from Mov13 tibia and parietal bone. Although the Mov13 mouse did not demonstrate evidence of active abnormal otic capsule remodeling as seen in type I OI and otosclerosis, studying gene expression in the Mov13 mouse has provided evidence that osteocytes of the otic capsule differ from osteocytes in other bones.
Collapse
Affiliation(s)
- Konstantina M Stankovic
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Mass. 02114-3096, USA
| | | | | | | | | | | |
Collapse
|
39
|
Lehnerdt G, Metz KA, Trellakis S, Jahnke K, Neumann A. Signaling by way of type IB and II bone morphogenetic protein receptors regulates bone formation in otospongiosis. Laryngoscope 2007; 117:812-6. [PMID: 17473674 DOI: 10.1097/mlg.0b013e31803300a2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
HYPOTHESIS The main goal of this study was to perform an immunohistologic analysis of bone morphogenetic protein receptors (BMPR) in otospongiosis. BACKGROUND BMP-2, -4, and -7 play an essential role in bone formation and repair. They do so as well in otosclerosis. It has been shown that these BMPs are traceable in osteocytes and osteoclasts in the active phase of otosclerosis (otospongiosis). The role of the different BMP receptors in otosclerotic bone transformation has not been previously analyzed. METHODS The posterior parts of the stapes footplates, collected during partial stapedectomies in 35 patients with clinical otosclerosis, were analyzed for histologic otosclerotic lesions after hematoxylin staining. Immunohistochemical analysis was performed using polyclonal immunoglobulin G antibodies for BMPR-IA, -IB, and -II, as well as biotinylated secondary antibodies, avidin-biotin-peroxidase complex reaction, and alkaline phosphatase staining with nitroblue-tetrazolium-chloride. RESULTS Seventeen of 35 (49%) specimens contained otosclerosis, but only 5 of these exhibited an otospongiotic phase. The abundant osteoblasts and osteoclasts in these cases showed distinct immunochemical staining for BMP-2, -4, and -7. In two cases, there could also be found an immense positive staining for BMPR-IB and modest staining for BMPR-II, whereas BMPR-1A always remained negative. CONCLUSION It was demonstrated for the first time that in otospongiosis, the actions of the BMPs are mediated through BMPR-IB and BMPR-II. To determine this role in detail, further investigations, especially for the phosphorylated Smad proteins within the BMP dependent mediator cascade, will be necessary.
Collapse
Affiliation(s)
- Goetz Lehnerdt
- Department of Otorhinolaryngology, University of Duisburg-Essen, Essen, Germany.
| | | | | | | | | |
Collapse
|
40
|
Yamanishi T, Katsu K, Funahashi JI, Yumoto E, Yokouchi Y. Dan is required for normal morphogenesis and patterning in the developing chick inner ear. Dev Growth Differ 2007; 49:13-26. [PMID: 17227341 DOI: 10.1111/j.1440-169x.2007.00900.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
During vertebrate inner ear development, compartmentalization of the auditory and vestibular apparatuses along two axes depends on the patterning of transcription factors expressed in a region-specific manner. Although most of the patterning is regulated by extrinsic signals, it is not known how Nkx5.1 and Msx1 are patterned. We focus on Dan, the founding member of the Cerberus/Dan gene family that encodes BMP antagonists, and describe its function in morphogenesis and patterning. First, we confirmed that Dan is expressed in the dorso-medial region of the otic vesicle that corresponds to the presumptive endolymphatic duct and sac (ed/es). Second, we used siRNA knockdown to demonstrate that depletion of Dan induced both a severe reduction in the size of the ed/es and moderate deformities of the semicircular canals and cochlear duct. Depletion of Dan also caused suppression of Nkx5.1 in the dorso-lateral region, suppression of Msx1 in the dorso-medial region, and ectopic induction of Nkx5.1 and Msx1 in the ventro-medial region. Most of these phenotypes also appeared following misexpression of the constitutively active form of BMP receptor type Ib. Thus, Dan is required for the normal morphogenesis of the inner ear and, by inhibiting BMP signaling, for the patterning of the transcription factors Nkx5.1 and Msx1.
Collapse
Affiliation(s)
- Takahiro Yamanishi
- Division of Pattern Formation, Department of Organogenesis, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto 860-0811, Japan
| | | | | | | | | |
Collapse
|
41
|
Kelly M, Chen P. Shaping the mammalian auditory sensory organ by the planar cell polarity pathway. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2007; 51:535-47. [PMID: 17891715 PMCID: PMC4158833 DOI: 10.1387/ijdb.072344mk] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The human ear is capable of processing sound with a remarkable resolution over a wide range of intensity and frequency. This ability depends largely on the extraordinary feats of the hearing organ, the organ of Corti and its sensory hair cells. The organ of Corti consists of precisely patterned rows of sensory hair cells and supporting cells along the length of the snail-shaped cochlear duct. On the apical surface of each hair cell, several rows of actin-containing protrusions, known as stereocilia, form a "V"-shaped staircase. The vertices of all the "V"-shaped stereocilia point away from the center of the cochlea. The uniform orientation of stereocilia in the organ of Corti manifests a distinctive form of polarity known as planar cell polarity (PCP). Functionally, the direction of stereociliary bundle deflection controls the mechanical channels located in the stereocilia for auditory transduction. In addition, hair cells are tonotopically organized along the length of the cochlea. Thus, the uniform orientation of stereociliary bundles along the length of the cochlea is critical for effective mechanotransduction and for frequency selection. Here we summarize the morphological and molecular events that bestow the structural characteristics of the mammalian hearing organ, the growth of the snail-shaped cochlear duct and the establishment of PCP in the organ of Corti. The PCP of the sensory organs in the vestibule of the inner ear will also be described briefly.
Collapse
Affiliation(s)
- Michael Kelly
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ping Chen
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
42
|
Blauwkamp MN, Beyer LA, Kabara L, Takemura K, Buck T, King W, Dolan DF, Barald KF, Raphael Y, Koenig RJ. The role of bone morphogenetic protein 4 in inner ear development and function. Hear Res 2006; 225:71-9. [PMID: 17275231 PMCID: PMC1868473 DOI: 10.1016/j.heares.2006.12.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2006] [Revised: 12/06/2006] [Accepted: 12/07/2006] [Indexed: 11/26/2022]
Abstract
Bone Morphogenetic Protein 4 (BMP4) is a member of the TGF-beta superfamily and is known to be important for the normal development of many tissues and organs, including the inner ear. Bmp4 homozygous null mice die as embryos, but Bmp4 heterozygous null (Bmp4(+/-)) mice are viable and some adults exhibit a circling phenotype, suggestive of an inner ear defect. To understand the role of BMP4 in inner ear development and function, we have begun to study C57BL/6 Bmp4(+/-) mice. Quantitative testing of the vestibulo-collic reflex, which helps maintain head stability, demonstrated that Bmp4(+/-) mice that exhibit circling behavior have a poor response in the yaw axis, consistent with semicircular canal dysfunction. Although the hair cells of the ampullae were grossly normal, the stereocilia were greatly reduced in number. Auditory brainstem responses showed that Bmp4(+/-) mice have elevated hearing thresholds and immunohistochemical staining demonstrated decreased numbers of neuronal processes in the organ of Corti. Thus Bmp4(+/-) mice have structural and functional deficits in the inner ear.
Collapse
Affiliation(s)
- Marsha N. Blauwkamp
- Cellular and Molecular Biology Program 2966 Taubman Medical Library, University of Michigan, Ann Arbor MI 48109-0619, USA
| | - Lisa A. Beyer
- Kresge Hearing Research Institute, Department of Otolaryngology 1301 East Ann Street, Ann Arbor, MI 48109-0506, USA
| | - Lisa Kabara
- Kresge Hearing Research Institute, Department of Otolaryngology 1301 East Ann Street, Ann Arbor, MI 48109-0506, USA
| | - Keiji Takemura
- Kresge Hearing Research Institute, Department of Otolaryngology 1301 East Ann Street, Ann Arbor, MI 48109-0506, USA
| | - Timothy Buck
- Kresge Hearing Research Institute, Department of Otolaryngology 1301 East Ann Street, Ann Arbor, MI 48109-0506, USA
| | - W.M. King
- Kresge Hearing Research Institute, Department of Otolaryngology 1301 East Ann Street, Ann Arbor, MI 48109-0506, USA
| | - David F. Dolan
- Kresge Hearing Research Institute, Department of Otolaryngology 1301 East Ann Street, Ann Arbor, MI 48109-0506, USA
| | - Kate F. Barald
- Cellular and Molecular Biology Program 2966 Taubman Medical Library, University of Michigan, Ann Arbor MI 48109-0619, USA
- Departments of Cell and Developmental Biology, and Biomedical Engineering 3053 BSRB, 109 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, 48109-2200, USA
| | - Yehoash Raphael
- Kresge Hearing Research Institute, Department of Otolaryngology 1301 East Ann Street, Ann Arbor, MI 48109-0506, USA
| | - Ronald J. Koenig
- Cellular and Molecular Biology Program 2966 Taubman Medical Library, University of Michigan, Ann Arbor MI 48109-0619, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes 5560 MSRB-II, 1150 W. Medical Center Drive, University of Michigan, Ann Arbor MI, 48109-0678, USA
| |
Collapse
|
43
|
Dudley BM, Runyan C, Takeuchi Y, Schaible K, Molyneaux K. BMP signaling regulates PGC numbers and motility in organ culture. Mech Dev 2006; 124:68-77. [PMID: 17112707 DOI: 10.1016/j.mod.2006.09.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2006] [Revised: 09/08/2006] [Accepted: 09/27/2006] [Indexed: 12/29/2022]
Abstract
Members of the bone morphogenetic protein (BMP) family play diverse roles in multiple developmental processes. However, in the mouse, mutations in many BMPs, BMP receptors and signaling components result in early embryonic lethality making it difficult to analyze the role of these factors during organogenesis or tissue homeostasis in the adult. To bypass this early lethality, we used an organ culture system to study the role of BMPs during primordial germ cell (PGC) migration. PGCs are the embryonic precursors of the sperm and eggs. BMPs induce formation of primordial germ cells within the proximal epiblast of embryonic day 7.5 (E7.5) mouse embryos. PGCs then migrate via the gut to arrive at the developing gonads by E10.5. Addition of BMP4 or the BMP-antagonist Noggin to transverse slices dissected from E9.5 embryos elevated PGC numbers or reduced PGC numbers, respectively. Noggin treatment also slowed and randomized PGC movements, resulting in a failure of PGCs to colonize the urogenital ridges (UGRs). Based on p-Smad1/5/8 staining, migratory PGCs do not respond to endogenous BMPs. Instead, the somatic cells of the urogenital ridges exhibit elevated p-Smad1/5/8 staining revealing active BMP signaling within the UGRs. Noggin treatment abrogated p-Smad staining within the UGRs and blocked localized expression of Kitl, a cytokine known to regulate the survival and motility of PGCs and Id1, a transcription factor expressed within the UGRs. We propose that BMP signaling regulates PGC migration by controlling gene expression within the somatic cells along the migration route and within the genital ridges.
Collapse
Affiliation(s)
- Brian M Dudley
- Department of Genetics, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | | | | | | | | |
Collapse
|
44
|
Wang Y, Shupenko CC, Melo LF, Strauss PR. DNA repair protein involved in heart and blood development. Mol Cell Biol 2006; 26:9083-93. [PMID: 16966376 PMCID: PMC1636828 DOI: 10.1128/mcb.01216-06] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Apurinic/apyrimidinic endonuclease 1, a key enzyme in repairing abasic sites in DNA, is an embryonic lethal in mice. We are examining its role in embryogenesis in zebra fish. Zebra fish contain two genomic copies (zfAPEX1a and zfAPEX1b) with identical coding sequences. zfAPEX1b lacks introns. Recombinant protein (ZAP1) is highly homologous with and has the same enzymatic properties as its human orthologue. ZAP1 is highly expressed throughout development. Embryos microinjected with morpholino oligonucleotide (MO) targeting the translation start site die at approximately the midblastula transition (MBT) without apoptosis. They are rescued with mRNA for human wild-type APEX1 but not for APEX1 encoding endonuclease-defective protein. Rescued embryos develop dysmorphic hearts, pericardial edema, few erythrocytes, small eyes, and abnormal notochords. Although the hearts in rescued embryos form defective loops ranging from no loop to one that is abnormally shaped, cardiac myosin (cmlc2) is present and contraction occurs. Embryos microinjected with MO targeting zfAPEX1a intron-exon junctions also pass the MBT with similar abnormalities. We conclude that AP endonuclease 1 is involved in both repairing DNA and regulating specific early stages of embryonic development.
Collapse
Affiliation(s)
- Yi Wang
- Department of Biology, Northeastern University, 360 Huntington Ave., Boston, MA 02115, USA
| | | | | | | |
Collapse
|
45
|
Romand R, Kondo T, Fraulob V, Petkovich M, Dollé P, Hashino E. Dynamic expression of retinoic acid-synthesizing and -metabolizing enzymes in the developing mouse inner ear. J Comp Neurol 2006; 496:643-54. [PMID: 16615129 PMCID: PMC2845518 DOI: 10.1002/cne.20936] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Retinoic acid signaling plays essential roles in morphogenesis and neural development through transcriptional regulation of downstream target genes. It is believed that the balance between the activities of synthesizing and metabolizing enzymes determines the amount of active retinoic acid to which a developing tissue is exposed. In this study, we investigated spatiotemporal expression patterns of four synthesizing enzymes, the retinaldehyde dehydrogenases 1, 2, 3, and 4 (Raldh1, Raldh2, Raldh3, and Raldh4) and two metabolizing enzymes (Cyp26A1 and Cyp26B1) in the embryonic and postnatal mouse inner ear by using quantitative reverse transcriptase polymerase chain reaction (RT-PCR), in situ hybridization, and Western blot analysis. Quantitative RT-PCR analysis and Western blot data revealed that the expression of CYP26s was much higher than that of Raldhs at early embryonic ages but that Cyp26 expression was downregulated during embryonic development. Conversely, the expression levels of Raldh2 and -3 increased during development and were significantly higher than the Cyp26 levels at postnatal day 20. At this age, Raldh3 was expressed predominantly in the cochlea, whereas Raldh2 was present in the vestibular end organ. At early embryonic stages, as observed by in situ hybridization, the synthesizing enzymes were expressed only in the dorsoventral epithelium of the otocyst, whereas the metabolizing enzymes were present mainly in mesenchymal cells surrounding the otic epithelium. At later stages, Raldh2, Raldh3, and Cyp26B1 were confined to the stria vascularis, spiral ganglion, and supporting cells in the cochlear and vestibular epithelia, respectively. The downregulation of Cyp26s and the upregulation of Raldhs after birth during inner ear maturation suggest tissue changes in the sensitivity to retinoic acid concentrations.
Collapse
|
46
|
Pujades C, Kamaid A, Alsina B, Giraldez F. BMP-signaling regulates the generation of hair-cells. Dev Biol 2006; 292:55-67. [PMID: 16458882 DOI: 10.1016/j.ydbio.2006.01.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2005] [Revised: 12/22/2005] [Accepted: 01/03/2006] [Indexed: 11/22/2022]
Abstract
Bone morphogenetic proteins (BMPs) are diffusible molecules involved in a variety of cellular interactions during development. Bmp4 expression accompanies the development of the ear sensory organs during patterning and specification of sensory cell fates, yet there is no understanding of the role of BMP4 in this process. The present work was aimed at exploring the effects of BMP-signaling on the development of hair-cells. For this purpose, we studied gene expression, cell proliferation and cell death in isolated chick otic vesicles that were grown in vitro in the presence of recombinant BMP4 or the BMP-inhibitor Noggin. Cath1 was used as a marker for hair-cell specification. BMP4 reduced the number of Cath1-cells and, conversely, Noggin increased the size of the sensory patches and the number of Cath1-positive cells. The effect of BMP4 was irreversible and occurred before hair-cell specification. Lfng and Fgf10 were expressed in the prosensory domain before Cath1, and their expression was expanded by Noggin. At these stages, modifications of BMP activity did not respecify non-sensory epithelium of the otic vesicle. The expression of Bmp4 at sensory patches was suppressed by BMP4 and induced by Noggin suggesting an autoregulatory loop. Analysis of BrdU incorporation during 6 and 18 h indicated that the effects of BMP4 were due to its ability to reduce the number of actively proliferating progenitors and inhibit cell fate specification. BMP4 induced cell death within the prosensory domain of the otic vesicle, along with the expression of Msx1, but not Msx2. On the contrary, BMP-inhibition with Noggin favored hair-cell specification without changes in the overall cell proliferation. We propose that about the stage of terminal division, the balance between BMP and BMP-inhibitory signals regulates survival and specification of hair-cell precursors, the final number of sensory hair-cells being limited by excess levels of BMPs. The final size of sensory patches would hence depend on the balance between BMP4 and opposing signals.
Collapse
Affiliation(s)
- Cristina Pujades
- Biologia del Desenvolupament, Departament de Ciències Experimentals i de la Salut (DCEXS), Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB) c/Dr. Aiguader 80, 08003-Barcelona, Spain
| | | | | | | |
Collapse
|
47
|
Pimanda JE, Chan WYI, Donaldson IJ, Bowen M, Green AR, Göttgens B. Endoglin expression in the endothelium is regulated by Fli-1, Erg, and Elf-1 acting on the promoter and a -8-kb enhancer. Blood 2006; 107:4737-45. [PMID: 16484587 DOI: 10.1182/blood-2005-12-4929] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis is critical to the growth and regeneration of tissue but is also a key component of tumor growth and chronic inflammatory disorders. Endoglin plays a key role in angiogenesis by modulating cellular responses to transforming growth factor-beta (TGF-beta) signaling and is upregulated in proliferating endothelial cells. To gain insights into the transcriptional hierarchies that govern endoglin expression, we used a combination of comparative genomic, biochemical, and transgenic approaches. Both the promoter and a region 8 kb upstream of exon 1 were active in transfection assays in endothelial cells. In transgenic mice, the promoter directed low-level expression to a subset of endothelial cells. By contrast, inclusion of the -8 enhancer resulted in robust endothelial activity with additional staining in developing ear mesenchyme. Subsequent molecular analysis demonstrated that both the -8 enhancer and the promoter depend on conserved Ets sites, which were bound in endothelial cells in vivo by Fli-1, Erg, and Elf-1. This study therefore establishes the transcriptional framework within which endoglin functions during angiogenesis.
Collapse
Affiliation(s)
- John E Pimanda
- Department of Hematology, Cambridge Institute of Medical Research, University of Cambridge, Cambridge CB2 2XY, UK
| | | | | | | | | | | |
Collapse
|
48
|
Kiernan AE, Xu J, Gridley T. The Notch ligand JAG1 is required for sensory progenitor development in the mammalian inner ear. PLoS Genet 2006; 2:e4. [PMID: 16410827 PMCID: PMC1326221 DOI: 10.1371/journal.pgen.0020004] [Citation(s) in RCA: 242] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2005] [Accepted: 11/30/2005] [Indexed: 02/07/2023] Open
Abstract
In mammals, six separate sensory regions in the inner ear are essential for hearing and balance function. Each sensory region is made up of hair cells, which are the sensory cells, and their associated supporting cells, both arising from a common progenitor. Little is known about the molecular mechanisms that govern the development of these sensory organs. Notch signaling plays a pivotal role in the differentiation of hair cells and supporting cells by mediating lateral inhibition via the ligands Delta-like 1 and Jagged (JAG) 2. However, another Notch ligand, JAG1, is expressed early in the sensory patches prior to cell differentiation, indicating that there may be an earlier role for Notch signaling in sensory development in the ear. Here, using conditional gene targeting, we show that the Jag1 gene is required for the normal development of all six sensory organs within the inner ear. Cristae are completely lacking in Jag1-conditional knockout (cko) mutant inner ears, whereas the cochlea and utricle show partial sensory development. The saccular macula is present but malformed. Using SOX2 and p27kip1 as molecular markers of the prosensory domain, we show that JAG1 is initially expressed in all the prosensory regions of the ear, but becomes down-regulated in the nascent organ of Corti by embryonic day 14.5, when the cells exit the cell cycle and differentiate. We also show that both SOX2 and p27kip1 are down-regulated in Jag1-cko inner ears. Taken together, these data demonstrate that JAG1 is expressed early in the prosensory domains of both the cochlear and vestibular regions, and is required to maintain the normal expression levels of both SOX2 and p27kip1. These data demonstrate that JAG1-mediated Notch signaling is essential during early development for establishing the prosensory regions of the inner ear. Deafness and adult-onset hearing loss are significant health problems. In most cases, deafness or vestibular dysfunction results when the sensory cells in the inner ear, known as hair cells, degenerate due to environmental or genetic causes. In the mammalian inner ear, the hair cells and their associated supporting cells can be found in six different patches that have particular functions related to hearing or balance. Unfortunately, unlike in birds or fish, mammalian hair cells show little ability to regenerate, resulting in a permanent hearing or balance disorder when damaged. Here, the authors show that a protein called JAG1, a ligand in the Notch signaling pathway, is required for the normal development of all six sensory regions in the mammalian inner ear. In ears that lacked JAG1, some of the sensory patches were missing completely, whereas others were small and lacked particular cell types. The authors showed that JAG1 is required by the sensory precursors, progenitor cells that give rise to both the hair cells and the supporting cells. By understanding how the sensory areas develop normally, it is hoped that molecular tools can be developed that will aid sensory regeneration in the mammalian inner ear.
Collapse
Affiliation(s)
- Amy E Kiernan
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Jingxia Xu
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Thomas Gridley
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
49
|
Abstract
The inner ear originates from an embryonic ectodermal placode and rapidly develops into a three-dimensional structure (the otocyst) through complex molecular and cellular interactions. Many genes and their products are involved in inner ear induction, organogenesis, and cell differentiation. Retinoic acid (RA) is an endogenous signaling molecule that may play a role during different phases of inner ear development, as shown from pathological observations. To gain insight into the function of RA during inner ear development, we have investigated the spatio-temporal expression patterns of major components of RA signaling pathway, including cellular retinoic acid binding proteins (CRABPs), cellular retinoid binding proteins (CRBPs), retinaldehyde dehydrogenases (RALDHs), catabolic enzymes (CYP26s), and nuclear receptors (RARs). Although the CrbpI, CrabpI, and -II genes are specifically expressed in the inner ear throughout development, loss-of-function studies have revealed that these proteins are dispensable for inner development and function. Several Raldh and Cyp26 gene transcripts are expressed at embryological day (E) 9.0-9.5 in the otocyst and show mainly complementary distributions in the otic epithelium and mesenchyme during following stages. From Western blot, RT-PCR, and in situ hybridization analysis, there is a low expression of Raldhs in the early otocyst at E9, while Cyp26s are strongly expressed. During the following days, there is an up-regulation of Raldhs and a down-regulation for Cyp26s. Specific RA receptor (Rar and Rxr) genes are expressed in the otocyst and during further development of the inner ear. At the otocyst stage, most of the components of the retinoid pathway are present, suggesting that the embryonic inner ear might act as an autocrine system, which is able to synthesize and metabolize RA necessary for its development. We propose a model in which two RA-dependent pathways may control inner ear ontogenesis: one indirect with RA from somitic mesoderm acting to regulate gene expression within the hindbrain neuroepithelium, and another with RA acting directly on the otocyst. Current evidence suggests that RA may regulate several genes involved in mesenchyme-epithelial interactions, thereby controlling inner ear morphogenesis. Our investigations suggest that RA signaling is a critical component not only of embryonic development, but also of postnatal maintenance of the inner ear.
Collapse
Affiliation(s)
- Raymond Romand
- Institut Clinique de la Souris and Institut de Génétique et de Biologie Moléculaire et cellulaire, B.P. 10142, 67404 Illkirch Cedex, France.
| | | | | |
Collapse
|
50
|
Abstract
Members of the Dlx gene family play essential roles in the development of the zebrafish and mouse inner ear, but little is known regarding Dlx genes and avian inner ear development. We have examined the inner ear expression patterns of Dlx1, Dlx2, Dlx3, Dlx5, and Dlx6 during the first 7 days of chicken embryonic development. Dlx1 and Dlx2 expression was seen only in nonneuronal cells of the cochleovestibular ganglion and nerves from stage 21 to stage 32. Dlx3 marks the otic placode beginning at stage 9 and becomes limited to epithelium adjacent to the hindbrain as invagination of the placode begins. Dlx3 expression then resolves to the dorsal otocyst and gradually becomes limited to the endolymphatic sac by stage 30. Dlx5 and Dlx6 expression in the developing inner ear is first seen at stages 12 and 13, respectively, in the rim of the otic pit, before spreading throughout the dorsal otocyst. As morphogenesis proceeds, Dlx5 and Dlx6 expression is seen throughout the forming semicircular canals and endolymphatic structures. During later stages, both genes are seen to mark the distal surface of the forming canals and display expression complementary to that of BMP4 in the vestibular sensory regions. Dlx5 expression is also seen in the lagena macula and the cochlear and vestibular nerves by stage 30. These findings suggest important roles for Dlx genes in the vestibular and neural development of the avian inner ear.
Collapse
Affiliation(s)
- Stephen T Brown
- Gonda Department of Cell and Molecular Biology, House Ear Institute, Los Angeles, California 90057-1922, USA
| | | | | |
Collapse
|