1
|
Gil Y, Ryu J, Yang H, Ma Y, Nam KH, Jang SW, Shim S. Molecular Characterization of Subdomain Specification of Cochlear Duct Based on Foxg1 and Gata3. Int J Mol Sci 2024; 25:12700. [PMID: 39684410 DOI: 10.3390/ijms252312700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
The inner ear is one of the sensory organs of vertebrates and is largely composed of the vestibule, which controls balance, and the cochlea, which is responsible for hearing. In particular, a problem in cochlear development can lead to hearing loss. Although numerous studies have been conducted on genes involved in the development of the cochlea, many areas still need to be discovered regarding factors that control the patterning of the early cochlear duct. Herein, based on the dynamic expression pattern of FOXG1 in the apical and basal regions of the E13.5 cochlear duct, we identified detailed expression regions through an open-source analysis of single-cell RNA analysis data and demonstrated a clinical correlation with hearing loss. The distinct expression patterns of FOXG1 and GATA3 during the patterning process of the cochlear duct provide important clues to understanding how the fates of the apical and basal regions are divided. These results are expected to be extremely important not only for understanding the molecular mechanisms involved in the early development of the cochlear duct, but also for identifying potential genes that cause hearing loss.
Collapse
Affiliation(s)
- Yongjin Gil
- Department of Biochemistry, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Jiho Ryu
- Department of Biochemistry, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Hayoung Yang
- Department of Biochemistry, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Yechan Ma
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 138-736, Republic of Korea
| | - Ki-Hoan Nam
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Sung-Wuk Jang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 138-736, Republic of Korea
| | - Sungbo Shim
- Department of Biochemistry, Chungbuk National University, Cheongju 28644, Republic of Korea
| |
Collapse
|
2
|
Edens BM, Bronner ME. Making developmental sense of the senses, their origin and function. Curr Top Dev Biol 2024; 159:132-167. [PMID: 38729675 DOI: 10.1016/bs.ctdb.2024.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
The primary senses-touch, taste, sight, smell, and hearing-connect animals with their environments and with one another. Aside from the eyes, the primary sense organs of vertebrates and the peripheral sensory pathways that relay their inputs arise from two transient stem cell populations: the neural crest and the cranial placodes. In this chapter we consider the senses from historical and cultural perspectives, and discuss the senses as biological faculties. We begin with the embryonic origin of the neural crest and cranial placodes from within the neural plate border of the ectodermal germ layer. Then, we describe the major chemical (i.e. olfactory and gustatory) and mechanical (i.e. vestibulo-auditory and somatosensory) senses, with an emphasis on the developmental interactions between neural crest and cranial placodes that shape their structures and functions.
Collapse
Affiliation(s)
- Brittany M Edens
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States.
| |
Collapse
|
3
|
Norton B, Quirk A, Matsuoka AJ. Unraveling the Mechanisms of Vestibular Neuron Formation from Human Induced Pluripotent Stem Cells. Tissue Eng Part A 2024; 30:131-143. [PMID: 37917115 DOI: 10.1089/ten.tea.2023.0166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023] Open
Abstract
The development of in vitro models that accurately recapitulate the complex cellular and molecular interactions of the inner ear is crucial for understanding inner ear development, function, and disease. In this study, we utilized a customized microfluidic platform to generate human induced pluripotent stem cell (hiPSC)-derived three-dimensional otic sensory neurons (OSNs). hiPSC-derived otic neuronal progenitors (ONPs) were cultured in hydrogel-embedded microfluidic channels over a 40-day period. Careful modulation of Wnt and Shh signaling pathways was used to influence dorsoventral patterning and direct differentiation toward a vestibular neuron lineage. After validating the microfluidic platform, OSN spheroid transcription factor and protein expression were assessed using real-time quantitative polymerase chain reaction (RT-qPCR), immunocytochemistry, and flow cytometry. The results demonstrated the successful differentiation of hiPSCs into ONPs and subsequent divergent differentiation into vestibular neuronal lineages, as evidenced by the expression of characteristic markers. Overall, our microfluidic platform provides a physiologically relevant environment for the culture and differentiation of hiPSCs, offering a valuable tool for studying inner ear development, disease and drug screening, and regenerative medicine applications.
Collapse
Affiliation(s)
- Benjamin Norton
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Analia Quirk
- Department of Otolaryngology and Head and Neck Surgery, University of California San Diego, La Jolla, California, USA
| | - Akihiro J Matsuoka
- Department of Otolaryngology and Head and Neck Surgery, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
4
|
Wang SX, Streit A. Shared features in ear and kidney development - implications for oto-renal syndromes. Dis Model Mech 2024; 17:dmm050447. [PMID: 38353121 PMCID: PMC10886756 DOI: 10.1242/dmm.050447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024] Open
Abstract
The association between ear and kidney anomalies has long been recognized. However, little is known about the underlying mechanisms. In the last two decades, embryonic development of the inner ear and kidney has been studied extensively. Here, we describe the developmental pathways shared between both organs with particular emphasis on the genes that regulate signalling cross talk and the specification of progenitor cells and specialised cell types. We relate this to the clinical features of oto-renal syndromes and explore links to developmental mechanisms.
Collapse
Affiliation(s)
- Scarlet Xiaoyan Wang
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Andrea Streit
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| |
Collapse
|
5
|
Blinkiewicz PV, Long MR, Stoner ZA, Ketchum EM, Sheltz-Kempf SN, Duncan JS. Gata3 is required in late proneurosensory development for proper sensory cell formation and organization. Sci Rep 2023; 13:12573. [PMID: 37537240 PMCID: PMC10400699 DOI: 10.1038/s41598-023-39707-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/29/2023] [Indexed: 08/05/2023] Open
Abstract
It has previously been shown that the zinc-finger transcription factor Gata3 has dynamic expression within the inner ear throughout embryonic development and is essential for cochlear neurosensory development. However, the temporal window for which Gata3 is required for proper formation of the cochlear neurosensory epithelia remains unclear. To investigate the role of Gata3 in cochlear neurosensory development in the late prosensory stages, we used the Sox2-creERT2 mouse line to target and conditionally delete Gata3 at E11.5, a timepoint before cells have fully committed to a neurosensory fate. While the inner ears of Sox2-creERT2: Gata3 f/f mice appear normal with no gross structural defects, the sensory cells in the organ of Corti are partially lost and disorganized in an increasing severity from base to apex. Additionally, spiral ganglion neurons display aberrant peripheral projections, including increased distances between radial bundles and disorganization upon reaching the organ of Corti. Furthermore, heterozygous Sox2-creERT2: Gata3 f/+ mice show a reduced aberrant phenotype in comparison to the homozygous mutant, supporting the hypothesis that Gata3 is not only required for proper formation at the later proneurosensory stage, but also that a specific expression level of Gata3 is required. Therefore, this study provides evidence that Gata3 plays a time-sensitive and dose-dependent role in the development of sensory and neuronal cells in late proneurosensory stages.
Collapse
Affiliation(s)
- Paige V Blinkiewicz
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, USA
| | - Makayla R Long
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, USA
| | - Zachary A Stoner
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, USA.
- Section On Sensory Cell Regeneration and Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Elizabeth M Ketchum
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, USA
| | | | - Jeremy S Duncan
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, USA.
- Department of Biomedical Sciences, Western Michigan School of Medicine, Kalamazoo, MI, USA.
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
6
|
Blinkiewicz PV, Long MR, Stoner ZA, Ketchum EM, Sheltz-Kempf SN, Duncan JS. Gata3 is Required in Late Proneurosensory Development for Proper Sensory Cell Formation and Organization. RESEARCH SQUARE 2023:rs.3.rs-2747944. [PMID: 37090645 PMCID: PMC10120746 DOI: 10.21203/rs.3.rs-2747944/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
It has been previously shown that zinc-finger transcription factor Gata3 has dynamic expression within the inner ear throughout embryonic development and is essential for cochlear neurosensory development. However, the temporal window to which Gata3 is required for the formation of the cochlear neurosensory epithelia remains unclear. To investigate the role of Gata3 on cochlear neurosensory development in the late prosensory stages, we used the Sox2-cre ERT2 mouse line to target and conditionally delete Gata3 at E11.5 before the cells have fully committed to a neurosensory fate. While the inner ears of Sox2-cre ERT2 : Gata3 f/f mice appear morphologically normal, the sensory cells in the organ of Corti are partially lost and disorganized in a basal to apical gradient with the apex demonstrating the more severe phenotype. Additionally, spiral ganglion neurons display aberrant peripheral projections, such as increased distances between radial bundles and disorganization upon reaching the organ of Corti. Furthermore, heterozygous Sox2-cre ERT2 : Gata3 f/+ mice show a reduced phenotype in comparison to the homozygous mutant, supporting the concept that Gata3 is not only required for proper formation at the later proneurosensory stage, but also that a specific level of Gata3 is required. Therefore, our studies confirm that Gata3 plays a time-sensitive and dose-dependent role in the development of sensory cells in the late proneurosensory stages.
Collapse
|
7
|
Origin of Neuroblasts in the Avian Otic Placode and Their Distributions in the Acoustic and Vestibular Ganglia. BIOLOGY 2023; 12:biology12030453. [PMID: 36979145 PMCID: PMC10045822 DOI: 10.3390/biology12030453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
The inner ear is a complex three-dimensional sensorial structure with auditory and vestibular functions. This intricate sensory organ originates from the otic placode, which generates the sensory elements of the membranous labyrinth, as well as all the ganglionic neuronal precursors. How auditory and vestibular neurons establish their fate identities remains to be determined. Their topological origin in the incipient otic placode could provide positional information before they migrate, to later segregate in specific portions of the acoustic and vestibular ganglia. To address this question, transplants of small portions of the avian otic placode were performed according to our previous fate map study, using the quail/chick chimeric graft model. All grafts taking small areas of the neurogenic placodal domain contributed neuroblasts to both acoustic and vestibular ganglia. A differential distribution of otic neurons in the anterior and posterior lobes of the vestibular ganglion, as well as in the proximal, intermediate, and distal portions of the acoustic ganglion, was found. Our results clearly show that, in birds, there does not seem to be a strict segregation of acoustic and vestibular neurons in the incipient otic placode.
Collapse
|
8
|
Tan AL, Mohanty S, Guo J, Lekven AC, Riley BB. Pax2a, Sp5a and Sp5l act downstream of Fgf and Wnt to coordinate sensory-neural patterning in the inner ear. Dev Biol 2022; 492:139-153. [PMID: 36244503 DOI: 10.1016/j.ydbio.2022.10.004] [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: 07/22/2022] [Revised: 09/25/2022] [Accepted: 10/10/2022] [Indexed: 01/21/2023]
Abstract
In zebrafish, sensory epithelia and neuroblasts of the inner ear form simultaneously in abutting medial and lateral domains, respectively, in the floor of the otic vesicle. Previous studies support regulatory roles for Fgf and Wnt, but how signaling is coordinated is poorly understood. We investigated this problem using pharmacological and transgenic methods to alter Fgf or Wnt signaling from early placodal stages to evaluate later changes in growth and patterning. Blocking Fgf at any stage reduces proliferation of otic tissue and terminates both sensory and neural specification. Wnt promotes proliferation in the otic vesicle but is not required for sensory or neural development. However, sustained overactivation of Wnt laterally expands sensory epithelia and blocks neurogenesis. pax2a, sp5a and sp5l are coregulated by Fgf and Wnt and show overlapping expression in the otic placode and vesicle. Gain- and loss-of-function studies show that these genes are together required for Wnt's suppression of neurogenesis, as well as some aspects of sensory development. Thus, pax2a, sp5a and sp5l are critical for mediating Fgf and Wnt signaling to promote spatially localized sensory and neural development.
Collapse
Affiliation(s)
- Amy L Tan
- Biology Department, Texas A&M University, College Station, TX, United States
| | - Saurav Mohanty
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| | - Jinbai Guo
- Biology Department, Texas A&M University, College Station, TX, United States
| | - Arne C Lekven
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| | - Bruce B Riley
- Biology Department, Texas A&M University, College Station, TX, United States.
| |
Collapse
|
9
|
Linker SB, Narvaiza I, Hsu JY, Wang M, Qiu F, Mendes APD, Oefner R, Kottilil K, Sharma A, Randolph-Moore L, Mejia E, Santos R, Marchetto MC, Gage FH. Human-specific regulation of neural maturation identified by cross-primate transcriptomics. Curr Biol 2022; 32:4797-4807.e5. [PMID: 36228612 DOI: 10.1016/j.cub.2022.09.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 07/08/2022] [Accepted: 09/14/2022] [Indexed: 11/06/2022]
Abstract
Unique aspects of human behavior are often attributed to differences in the relative size and organization of the human brain: these structural aspects originate during early development. Recent studies indicate that human neurodevelopment is considerably slower than that in other nonhuman primates, a finding that is termed neoteny. One aspect of neoteny is the slow onset of action potentials. However, which molecular mechanisms play a role in this process remain unclear. To examine the evolutionary constraints on the rate of neuronal maturation, we have generated transcriptional data tracking five time points, from the neural progenitor state to 8-week-old neurons, in primates spanning the catarrhine lineage, including Macaca mulatta, Gorilla gorilla, Pan paniscus, Pan troglodytes, and Homo sapiens. Despite finding an overall similarity of many transcriptional signatures, species-specific and clade-specific distinctions were observed. Among the genes that exhibited human-specific regulation, we identified a key pioneer transcription factor, GATA3, that was uniquely upregulated in humans during the neuronal maturation process. We further examined the regulatory nature of GATA3 in human cells and observed that downregulation quickened the speed of developing spontaneous action potentials, thereby modulating the human neotenic phenotype. These results provide evidence for the divergence of gene regulation as a key molecular mechanism underlying human neoteny.
Collapse
Affiliation(s)
- Sara B Linker
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Pines Road, La Jolla, CA 92037, USA
| | - Iñigo Narvaiza
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Pines Road, La Jolla, CA 92037, USA
| | - Jonathan Y Hsu
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Pines Road, La Jolla, CA 92037, USA
| | - Meiyan Wang
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Pines Road, La Jolla, CA 92037, USA
| | - Fan Qiu
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Pines Road, La Jolla, CA 92037, USA
| | - Ana P D Mendes
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Pines Road, La Jolla, CA 92037, USA
| | - Ruth Oefner
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Pines Road, La Jolla, CA 92037, USA
| | - Kalyani Kottilil
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Pines Road, La Jolla, CA 92037, USA
| | - Amandeep Sharma
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Pines Road, La Jolla, CA 92037, USA
| | - Lynne Randolph-Moore
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Pines Road, La Jolla, CA 92037, USA
| | - Eunice Mejia
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Pines Road, La Jolla, CA 92037, USA
| | - Renata Santos
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Pines Road, La Jolla, CA 92037, USA; Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Laboratory of Dynamics of Neuronal Structure in Health and Disease, 102 rue de la Santé, 75014 Paris, France; Institut des Sciences Biologiques, CNRS, 16 rue Pierre et Marie Curie, 75005 Paris, France
| | - Maria C Marchetto
- Department of Anthropology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Center for Academic Research and Training in Anthropogeny (CARTA), University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Fred H Gage
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
10
|
Shively SB, Edwards NA, MacDonald TJ, Johnson KR, Diaz-Rodriguez NM, Merrill MJ, Vortmeyer AO. Developmentally Arrested Basket/Stellate Cells in Postnatal Human Brain as Potential Tumor Cells of Origin for Cerebellar Hemangioblastoma in von Hippel-Lindau Patients. J Neuropathol Exp Neurol 2022; 81:885-899. [PMID: 35980299 PMCID: PMC9803908 DOI: 10.1093/jnen/nlac073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
von Hippel-Lindau (VHL) disease is an autosomal dominant hereditary cancer disorder caused by a germline mutation in the VHL tumor suppressor gene. Loss of the wild-type allele results in VHL deficiency and the potential formation of cerebellar hemangioblastomas, which resemble embryonic hemangioblast proliferation and differentiation processes. Multiple, microscopic, VHL-deficient precursors, termed developmentally arrested structural elements (DASEs), consistently involve the cerebellar molecular layer in VHL patients, indicating the tumor site of origin. Unlike hemangioblastomas, however, cerebellar DASEs do not express brachyury, a mesodermal marker for hemangioblasts. In this study, neuronal progenitors occupying the molecular layer were investigated as tumor cells of origin. By immunohistochemistry, cerebellar DASEs and hemangioblastomas lacked immunoreactivity with antibody ZIC1 (Zic family member 1), a granule cell progenitor marker with concordance from oligonucleotide RNA expression array analyses. Rather, cerebellar DASEs and hemangioblastomas were immunoreactive with antibody PAX2 (paired box 2), a marker of basket/stellate cell progenitors. VHL cerebellar cortices also revealed PAX2-positive cells in Purkinje and molecular layers, resembling the histological and molecular development of basket/stellate cells in postnatal non-VHL mouse and human cerebella. These data suggest that VHL deficiency can result in the developmental arrest of basket/stellate cells in the human cerebellum and that these PAX2-positive, initiated cells await another insult or signal to form DASEs and eventually, tumors.
Collapse
Affiliation(s)
- Sharon Baughman Shively
- From the Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA,Department of Molecular Medicine, Institute for Biomedical Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Nancy A Edwards
- From the Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Kory R Johnson
- Bioinformatics Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | - Alexander O Vortmeyer
- Send correspondence to: Alexander O. Vortmeyer, MD, PhD, Division of Neuropathology, Department of Pathology and Laboratory Medicine, Indiana University-Purdue University Indianapolis, 350 W. 11th Street, Suite 4034, Indianapolis, IN 46202, USA; E-mail:
| |
Collapse
|
11
|
Saeki T, Yoshimatsu S, Ishikawa M, Hon CC, Koya I, Shibata S, Hosoya M, Saegusa C, Ogawa K, Shin JW, Fujioka M, Okano H. Critical roles of FGF, RA, and WNT signalling in the development of the human otic placode and subsequent lineages in a dish. Regen Ther 2022; 20:165-186. [PMID: 35620640 PMCID: PMC9114627 DOI: 10.1016/j.reth.2022.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/19/2022] [Accepted: 04/26/2022] [Indexed: 11/30/2022] Open
Abstract
Introduction Efficient induction of the otic placode, the developmental origin of the inner ear from human pluripotent stem cells (hPSCs), provides a robust platform for otic development and sensorineural hearing loss modelling. Nevertheless, there remains a limited capacity of otic lineage specification from hPSCs by stepwise differentiation methods, since the critical factors for successful otic cell differentiation have not been thoroughly investigated. In this study, we developed a novel differentiation system involving the use of a three-dimensional (3D) floating culture with signalling factors for generating otic cell lineages via stepwise differentiation of hPSCs. Methods We differentiated hPSCs into preplacodal cells under a two-dimensional (2D) monolayer culture. Then, we transferred the induced preplacodal cells into a 3D floating culture under the control of the fibroblast growth factor (FGF), bone morphogenetic protein (BMP), retinoic acid (RA) and WNT signalling pathways. We evaluated the characteristics of the induced cells using immunocytochemistry, quantitative PCR (qPCR), population averaging, and single-cell RNA-seq (RNA-seq) analysis. We further investigated the methods for differentiating otic progenitors towards hair cells by overexpression of defined transcription factors. Results We demonstrated that hPSC-derived preplacodal cells acquired the potential to differentiate into posterior placodal cells in 3D floating culture with FGF2 and RA. Subsequent activation of WNT signalling induced otic placodal cell formation. By single-cell RNA-seq (scRNA-seq) analysis, we identified multiple clusters of otic placode- and otocyst marker-positive cells in the induced spheres. Moreover, the induced otic cells showed the potential to generate hair cell-like cells by overexpression of the transcription factors ATOH1, POU4F3 and GFI1. Conclusions We demonstrated the critical role of FGF2, RA and WNT signalling in a 3D environment for the in vitro differentiation of otic lineage cells from hPSCs. The induced otic cells had the capacity to differentiate into inner ear hair cells with stereociliary bundles and tip link-like structures. The protocol will be useful for in vitro disease modelling of sensorineural hearing loss and human inner ear development and thus contribute to drug screening and stem cell-based regenerative medicine. A 3D floating culture condition is critical for inducing otic placodal cells from hPSCs-derived preplacodal cells. Activation of FGF, RA, WNT signalling pathways is indispensable for differentiating otic lineage under the 3D condition. Overexpression of defined transcription factors facilitated the generation of hair cells from hPSCs-derived otic cells.
Collapse
|
12
|
Hosoya M, Fujioka M, Okahara J, Yoshimatsu S, Okano H, Ozawa H. Early development of the cochlea of the common marmoset, a non-human primate model. Neural Dev 2022; 17:6. [PMID: 35524278 PMCID: PMC9077934 DOI: 10.1186/s13064-022-00162-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/13/2022] [Indexed: 11/12/2022] Open
Abstract
Background Fine-tuned cochlear development is essential for hearing. Owing to the difficulty in using early human fetal samples, most of our knowledge regarding cochlear development has been obtained from rodents. However, several inter-species differences in cochlear development between rodents and humans have been reported. To bridge these differences, we investigated early otic development of a non-human primate model animal, the common marmoset (Callithrix jacchus). Methods We examined 20 genes involved in early cochlear development and described the critical developmental steps for morphogenesis, which have been reported to vary between rodents and marmosets. Results The results revealed that several critical genes involved in prosensory epithelium specifications showed higher inter-species differences, suggesting that the molecular process for hair cell lineage acquisition in primates differs considerably from that of rodents. We also observed that the tempo of cochlear development was three times slower in the primate than in rodents. Conclusions Our data provide new insights into early cochlear development in primates and humans and imply that the procedures used for manipulating rodent cochlear sensory cells cannot be directly used for the research of primate cells due to the intrinsic inter-species differences in the cell fate determination program.
Collapse
Affiliation(s)
- Makoto Hosoya
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masato Fujioka
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, 160-8582, Japan. .,Department of Molecular Genetics, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan.
| | - Junko Okahara
- Laboratory for Marmoset Neural Architecture, Center for Brain Science, RIKEN, 2-1 Hirosawa Wako, Saitama, 351-0193, Japan.,Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, 3-25-12 Tonomachi Kawasaki-ku Kawasaki, Kanagawa, 210-0821, Japan
| | - Sho Yoshimatsu
- Laboratory for Marmoset Neural Architecture, Center for Brain Science, RIKEN, 2-1 Hirosawa Wako, Saitama, 351-0193, Japan.,Department of Physiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hideyuki Okano
- Laboratory for Marmoset Neural Architecture, Center for Brain Science, RIKEN, 2-1 Hirosawa Wako, Saitama, 351-0193, Japan.,Department of Physiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroyuki Ozawa
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
13
|
Wang M, Guo X, Yang M, Zhang Y, Meng F, Chen Y, Chen M, Qiu T, Li J, Li Z, Zhang Q, Xu F, Zhang H, Wang W. Synergistic antitumor activity of 5-fluorouracil and atosiban against microsatellite stable colorectal cancer through restoring GATA3. Biochem Pharmacol 2022; 199:115025. [PMID: 35367196 DOI: 10.1016/j.bcp.2022.115025] [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: 11/26/2021] [Revised: 03/19/2022] [Accepted: 03/25/2022] [Indexed: 11/02/2022]
Abstract
Clinically, 5-fluorouracil (5-Fu) is a first-line drug for the treatment of patients with colorectal cancer (CRC). However, chemoresistance to 5-Fu-based chemotherapy is a leading obstacle in achieving effective treatment for CRC, especially microsatellite stable (MSS) CRC. Since the cytotoxicity of 5-Fu is negatively correlated with oxytocin receptor (OXTR) expression in MSS CRC cell lines, our current study aimed to investigate the synergistic antitumor activity of 5-Fu combined with atosiban, an antagonist of OXTR. Our results suggested that atosiban remarkably potentiated the inhibitory effect of 5-Fu on the growth of MSS-type CRC cells in vitro and in vivo. Moreover, 5-Fu induced GATA3 in MSS CRC cells and tumors, which were eradicated by atosiban. Further investigation showed that atosiban strengthened the antitumor activity of 5-Fu through eradiation of 5-Fu-induced GATA3 in MSS-type CRC cells. Taken together, our findings suggest that atosiban potentiates the antitumor effect of 5-Fu by abolishing 5-Fu-induced GATA3, which provides a novel therapeutic strategy for MSS-type CRC via the combination of atosiban and 5-Fu.
Collapse
Affiliation(s)
- Mengmeng Wang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Xuqin Guo
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Man Yang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yawen Zhang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Fanyi Meng
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yinshuang Chen
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Mengxi Chen
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Tian Qiu
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Jiawei Li
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Zhi Li
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Qi Zhang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Fang Xu
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Haiyang Zhang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China.
| | - Weipeng Wang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China.
| |
Collapse
|
14
|
Maul A, Huebner AK, Strenzke N, Moser T, Rübsamen R, Jovanovic S, Hübner CA. The Cl--channel TMEM16A is involved in the generation of cochlear Ca2+ waves and promotes the refinement of auditory brainstem networks in mice. eLife 2022; 11:72251. [PMID: 35129434 PMCID: PMC8871368 DOI: 10.7554/elife.72251] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 02/06/2022] [Indexed: 11/17/2022] Open
Abstract
Before hearing onset (postnatal day 12 in mice), inner hair cells (IHCs) spontaneously fire action potentials, thereby driving pre-sensory activity in the ascending auditory pathway. The rate of IHC action potential bursts is modulated by inner supporting cells (ISCs) of Kölliker’s organ through the activity of the Ca2+-activated Cl--channel TMEM16A (ANO1). Here, we show that conditional deletion of Ano1 (Tmem16a) in mice disrupts Ca2+ waves within Kölliker’s organ, reduces the burst-firing activity and the frequency selectivity of auditory brainstem neurons in the medial nucleus of the trapezoid body (MNTB), and also impairs the functional refinement of MNTB projections to the lateral superior olive. These results reveal the importance of the activity of Kölliker’s organ for the refinement of central auditory connectivity. In addition, our study suggests the involvement of TMEM16A in the propagation of Ca2+ waves, which may also apply to other tissues expressing TMEM16A.
Collapse
Affiliation(s)
- Alena Maul
- Neuroscience Department, Max Delbrück Center for Molecular Medicine
| | | | - Nicola Strenzke
- Institute for Auditory Neuroscience, Department of Otolaryngology, University of Göttingen
| | - Tobias Moser
- Institute for Auditory Neuroscience, Department of Otolaryngology, University of Göttingen
| | - Rudolf Rübsamen
- Faculty of Bioscience, Pharmacy and Psychology, University of Leipzig
| | - Saša Jovanovic
- Faculty of Bioscience, Pharmacy and Psychology, University of Leipzig
| | | |
Collapse
|
15
|
GATA3 improves the protective effects of bone marrow-derived mesenchymal stem cells against ischemic stroke induced injury by regulating autophagy through CREG. Brain Res Bull 2021; 176:151-160. [PMID: 34500038 DOI: 10.1016/j.brainresbull.2021.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/14/2021] [Accepted: 09/02/2021] [Indexed: 01/15/2023]
Abstract
BACKGROUND Bone marrow-derived mesenchymal stem cells (BMSCs) transplantation has been demonstrated to benefit functional recovery after ischemic stroke, however, the low survival rate of BMSCs in ischemic microenvironment largely limits its use. METHODS Rat BMSCs (rBMSCs) were isolated from SD rats and treated with oxygen glucose deprivation/reoxygenation (OGD) to mimic ischemic microenvironment in vitro. Expression of mRNAs and proteins were assessed by qRT-PCR and western blot, respectively. Cell viability was detected using MTT. ROS level was evaluated by DCFH-DA Assay Kit. TUNEL and flow cytometry analysis were adopted to detect cell apoptosis. Immunofluorescence analysis was used to examine LC3 expression. Dual-luciferase reporter and ChIP assays were employed to determine the interaction between CREG and GATA3. Middle cerebral artery occlusion (MCAO) model was established to mimic ischemic stroke in vivo. TTC staining was used to measure the infarcts area in the brain of MCAO rats. Nissl staining was used to examine the quantity of neurons, and mNSS test was applied to compare behavioral functions of animals. RESULTS The rBMSCs were successfully isolated from SD rats. OGD exposure decreased the expression of GATA3 in rBMSCs, GATA3 overexpression alleviated OGD-induced cell injury and enhanced autophagy. Treatment with autophagy inhibitor (3-MA) abolished the protective effects of GATA3 against OGD-induced cell injury. GATA3 targeted the promoter of CREG and positively regulated its expression. The protective effect of GATA3 overexpression on autophagy during OGD exposure was reversed by CREG knockdown. Moreover, GATA3 overexpression improved the therapeutic effects of BMSCs transplantation on ischemic stroke in vivo. CONCLUSION Our results indicated that GATA3 overexpression improved the therapeutic effects of rBMSCs transplantation against ischemic stroke induced injury by regulating autophagy through CREG.
Collapse
|
16
|
Wen J, Song J, Bai Y, Liu Y, Cai X, Mei L, Ma L, He C, Feng Y. A Model of Waardenburg Syndrome Using Patient-Derived iPSCs With a SOX10 Mutation Displays Compromised Maturation and Function of the Neural Crest That Involves Inner Ear Development. Front Cell Dev Biol 2021; 9:720858. [PMID: 34426786 PMCID: PMC8379019 DOI: 10.3389/fcell.2021.720858] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 07/22/2021] [Indexed: 12/20/2022] Open
Abstract
Waardenburg syndrome (WS) is an autosomal dominant inherited disorder that is characterized by sensorineural hearing loss and abnormal pigmentation. SOX10 is one of its main pathogenicity genes. The generation of patient-specific induced pluripotent stem cells (iPSCs) is an efficient means to investigate the mechanisms of inherited human disease. In our work, we set up an iPSC line derived from a WS patient with SOX10 mutation and differentiated into neural crest cells (NCCs), a key cell type involved in inner ear development. Compared with control-derived iPSCs, the SOX10 mutant iPSCs showed significantly decreased efficiency of development and differentiation potential at the stage of NCCs. After that, we carried out high-throughput RNA-seq and evaluated the transcriptional misregulation at every stage. Transcriptome analysis of differentiated NCCs showed widespread gene expression alterations, and the differentially expressed genes (DEGs) were enriched in gene ontology terms of neuron migration, skeletal system development, and multicellular organism development, indicating that SOX10 has a pivotal part in the differentiation of NCCs. It's worth noting that, a significant enrichment among the nominal DEGs for genes implicated in inner ear development was found, as well as several genes connected to the inner ear morphogenesis. Based on the protein-protein interaction network, we chose four candidate genes that could be regulated by SOX10 in inner ear development, namely, BMP2, LGR5, GBX2, and GATA3. In conclusion, SOX10 deficiency in this WS subject had a significant impact on the gene expression patterns throughout NCC development in the iPSC model. The DEGs most significantly enriched in inner ear development and morphogenesis may assist in identifying the underlying basis for the inner ear malformation in subjects with WS.
Collapse
Affiliation(s)
- Jie Wen
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China.,Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China.,Department of Geriatrics, National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Song
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China.,Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China.,Department of Geriatrics, National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yijiang Bai
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China.,Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China.,Department of Geriatrics, National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yalan Liu
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China.,Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China.,Department of Geriatrics, National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xinzhang Cai
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China.,Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China.,Department of Geriatrics, National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lingyun Mei
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China.,Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China.,Department of Geriatrics, National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Ma
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Chufeng He
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China.,Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China.,Department of Geriatrics, National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yong Feng
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China.,Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| |
Collapse
|
17
|
A Missense Mutation in the KLF7 Gene Is a Potential Candidate Variant for Congenital Deafness in Australian Stumpy Tail Cattle Dogs. Genes (Basel) 2021; 12:genes12040467. [PMID: 33805165 PMCID: PMC8064056 DOI: 10.3390/genes12040467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/21/2021] [Accepted: 03/23/2021] [Indexed: 12/20/2022] Open
Abstract
Congenital deafness is prevalent among modern dog breeds, including Australian Stumpy Tail Cattle Dogs (ASCD). However, in ASCD, no causative gene has been identified so far. Therefore, we performed a genome-wide association study (GWAS) and whole genome sequencing (WGS) of affected and normal individuals. For GWAS, 3 bilateral deaf ASCDs, 43 herding dogs, and one unaffected ASCD were used, resulting in 13 significantly associated loci on 6 chromosomes, i.e., CFA3, 8, 17, 23, 28, and 37. CFA37 harbored a region with the most significant association (−log10(9.54 × 10−21) = 20.02) as well as 7 of the 13 associated loci. For whole genome sequencing, the same three affected ASCDs and one unaffected ASCD were used. The WGS data were compared with 722 canine controls and filtered for protein coding and non-synonymous variants, resulting in four missense variants present only in the affected dogs. Using effect prediction tools, two variants remained with predicted deleterious effects within the Heart development protein with EGF like domains 1 (HEG1) gene (NC_006615.3: g.28028412G>C; XP_022269716.1: p.His531Asp) and Kruppel-like factor 7 (KLF7) gene (NC_006619.3: g.15562684G>A; XP_022270984.1: p.Leu173Phe). Due to its function as a regulator in heart and vessel formation and cardiovascular development, HEG1 was excluded as a candidate gene. On the other hand, KLF7 plays a crucial role in the nervous system, is expressed in the otic placode, and is reported to be involved in inner ear development. 55 additional ASCD samples (28 deaf and 27 normal hearing dogs) were genotyped for the KLF7 variant, and the variant remained significantly associated with deafness in ASCD (p = 0.014). Furthermore, 24 dogs with heterozygous or homozygous mutations were detected, including 18 deaf dogs. The penetrance was calculated to be 0.75, which is in agreement with previous reports. In conclusion, KLF7 is a promising candidate gene causative for ASCD deafness.
Collapse
|
18
|
Su Y, Yang LM, Ornitz DM. FGF20-FGFR1 signaling through MAPK and PI3K controls sensory progenitor differentiation in the organ of Corti. Dev Dyn 2021; 250:134-144. [PMID: 32735383 PMCID: PMC8415122 DOI: 10.1002/dvdy.231] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Fibroblast Growth Factor 20 (FGF20)-FGF receptor 1 (FGFR1) signaling is essential for cochlear hair cell (HC) and supporting cell (SC) differentiation. In other organ systems, FGFR1 signals through several intracellular pathways including MAPK (ERK), PI3K, phospholipase C ɣ (PLCɣ), and p38. Previous studies implicated MAPK and PI3K pathways in HC and SC development. We hypothesized that one or both would be important downstream mediators of FGF20-FGFR1 signaling for HC differentiation. RESULTS By inhibiting pathways downstream of FGFR1 in cochlea explant cultures, we established that both MAPK and PI3K pathways are required for HC differentiation while PLCɣ and p38 pathways are not. Examining the canonical PI3K pathway, we found that while AKT is necessary for HC differentiation, it is not sufficient to rescue the Fgf20-/- phenotype. To determine whether PI3K functions downstream of FGF20, we inhibited Phosphatase and Tensin Homolog (PTEN) in Fgf20-/- explants. Overactivation of PI3K resulted in a partial rescue of the Fgf20-/- phenotype, demonstrating a requirement for PI3K downstream of FGF20. Consistent with a requirement for the MAPK pathway for FGF20-regulated HC differentiation, we show that treating Fgf20-/- explants with FGF9 increased levels of dpERK. CONCLUSIONS Together, these data provide evidence that both MAPK and PI3K are important downstream mediators of FGF20-FGFR1 signaling during HC and SC differentiation.
Collapse
Affiliation(s)
- Yutao Su
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lu M Yang
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
19
|
Pouraghaei S, Moztarzadeh F, Chen C, Ansari S, Moshaverinia A. Microenvironment Can Induce Development of Auditory Progenitor Cells from Human Gingival Mesenchymal Stem Cells. ACS Biomater Sci Eng 2020; 6:2263-2273. [PMID: 33455314 DOI: 10.1021/acsbiomaterials.9b01795] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Sensorineural hearing loss in mammals occurs due to irreversible damage to the sensory epithelia of the inner ear and has very limited treatment options. The ability to regenerate the auditory progenitor cells is a promising approach for the treatment of sensorineural hearing loss; therefore, finding an appropriate and easily accessible stem cell source for restoring the sense of hearing would be of great interest. Here, we proposed a novel easy-to-access source of cells with the ability to recover auditory progenitor cells. In this study, gingival mesenchymal stem cells (GMSCs) were utilized, as these cells have high self-renewal and multipotent differentiation capacity and can be obtained easily from the oral cavity or discarded tissue samples at dental clinics. To manipulate the biophysical properties of the cellular microenvironment for promoting GMSC differentiation toward the target cells, we also tried to propose a candidate biomaterial. GMSCs in combination with an appropriate scaffold material can, therefore, present advantageous therapeutic options for a number of conditions. Here, we report the potential of GMSCs to differentiate into auditory progenitor cells while supporting them with an optimized three-dimensional scaffold and certain growth factors. A hybrid hydrogel scaffold based on peptide modified alginate and Matrigel was used here in addition to the presence of fibroblast growth factor-basic (bFGF), insulin-like growth factor (IGF), and epidermal growth factor (EGF). Our in vitro and in vivo studies confirmed the auditory differentiation potential of GMSCs within the engineered microenvironment.
Collapse
Affiliation(s)
- Sevda Pouraghaei
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, California, United States
| | - Fathollah Moztarzadeh
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Sahar Ansari
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Alireza Moshaverinia
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, California, United States
- California NanoSystems Institute, University of California, Los Angeles, California, United States
| |
Collapse
|
20
|
He D, Guo R, Zheng D, Xu M, Li P, Guo L, Gan L. Transcription factor Isl1 is dispensable for the development of the mouse prosensory region. Cytotechnology 2020; 72:407-414. [PMID: 32219582 DOI: 10.1007/s10616-020-00387-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 02/28/2020] [Indexed: 01/17/2023] Open
Abstract
In order to identify genes involved in the development of inner ear hair cells, we investigated the role of the transcription factor Islet-class LIM-homeodomain (LIM-HD) 1 (Isl1) in the development of the mouse prosensory region. Isl1 was deleted using the Pax2-Cre system, and deletion of both alleles was verified using cochlea sections. Changes in the number of prosensory region cells were measured to determine the effect of Isl1 on the development of the mouse prosensory region. In order to test whether Isl1 formed a protein complex with Ldb1 and Gata3, co-immunoprecipitation experiments were performed in HEK293 cells using the Flag-tagged LIM-domain of Isl1, HA-tagged LID of Ldb1 and Myc-tagged C-terminal domain of Gata3. The expression of Gata3, Sox2, Jag1 and P27 proteins in the prosensory region were not affected in Isl1-/- prosensory cells. Thus, Isl1 did not form a protein complex with Gata3 through Ldb1 in the Isl1-/- cells. Our results suggest that Isl1 may be dispensable for the development of the mouse prosensory region.
Collapse
Affiliation(s)
- Daqiang He
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Institute of Life Sciences, Hangzhou Normal University, Hangzhou, 310018, Zhejiang, China
- Department of Laboratory Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Rui Guo
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Dongwang Zheng
- Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences, HangZhou Medical College, Hangzhou, 310000, Zhejiang, China
| | - Mei Xu
- Institute of Life Sciences, Hangzhou Normal University, Hangzhou, 310018, Zhejiang, China
| | - Ping Li
- HangZhou CalyGene Bitechnology Limited Company, Hangzhou, 310013, Zhejiang, China
| | - Luming Guo
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Lin Gan
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
- Institute of Life Sciences, Hangzhou Normal University, Hangzhou, 310018, Zhejiang, China.
| |
Collapse
|
21
|
Hoshino T, Terunuma T, Takai J, Uemura S, Nakamura Y, Hamada M, Takahashi S, Yamamoto M, Engel JD, Moriguchi T. Spiral ganglion cell degeneration-induced deafness as a consequence of reduced GATA factor activity. Genes Cells 2019; 24:534-545. [PMID: 31141264 DOI: 10.1111/gtc.12705] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/18/2019] [Accepted: 05/21/2019] [Indexed: 12/19/2022]
Abstract
Zinc-finger transcription factors GATA2 and GATA3 are both expressed in the developing inner ear, although their overlapping versus distinct activities in adult definitive inner ear are not well understood. We show here that GATA2 and GATA3 are co-expressed in cochlear spiral ganglion cells and redundantly function in the maintenance of spiral ganglion cells and auditory neural circuitry. Notably, Gata2 and Gata3 compound heterozygous mutant mice had a diminished number of spiral ganglion cells due to enhanced apoptosis, which resulted in progressive hearing loss. The decrease in spiral ganglion cellularity was associated with lowered expression of neurotrophin receptor TrkC that is an essential factor for spiral ganglion cell survival. We further show that Gata2 null mutants that additionally bear a Gata2 YAC (yeast artificial chromosome) that counteracts the lethal hematopoietic deficiency due to complete Gata2 loss nonetheless failed to complement the deficiency in neonatal spiral ganglion neurons. Furthermore, cochlea-specific Gata2 deletion mice also had fewer spiral ganglion cells and resultant hearing impairment. These results show that GATA2 and GATA3 redundantly function to maintain spiral ganglion cells and hearing. We propose possible mechanisms underlying hearing loss in human GATA2- or GATA3-related genetic disorders.
Collapse
Affiliation(s)
- Tomofumi Hoshino
- Department of Otolaryngology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tsumoru Terunuma
- Department of Otolaryngology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Jun Takai
- Division of Medical Biochemistry, Tohoku Medical Pharmaceutical University, Sendai, Japan
| | - Satoshi Uemura
- Division of Medical Biochemistry, Tohoku Medical Pharmaceutical University, Sendai, Japan
| | - Yasuhiro Nakamura
- Division of Pathology, Tohoku Medical Pharmaceutical University, Sendai, Japan
| | - Michito Hamada
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Takashi Moriguchi
- Division of Medical Biochemistry, Tohoku Medical Pharmaceutical University, Sendai, Japan
| |
Collapse
|
22
|
Bardhan T, Jeng J, Waldmann M, Ceriani F, Johnson SL, Olt J, Rüttiger L, Marcotti W, Holley MC. Gata3 is required for the functional maturation of inner hair cells and their innervation in the mouse cochlea. J Physiol 2019; 597:3389-3406. [PMID: 31069810 PMCID: PMC6636704 DOI: 10.1113/jp277997] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/07/2019] [Indexed: 01/14/2023] Open
Abstract
KEY POINTS The physiological maturation of auditory hair cells and their innervation requires precise temporal and spatial control of cell differentiation. The transcription factor gata3 is essential for the earliest stages of auditory system development and for survival and synaptogenesis in auditory sensory afferent neurons. We show that during postnatal development in the mouse inner ear gata3 is required for the biophysical maturation, growth and innervation of inner hair cells; in contrast, it is required only for the survival of outer hair cells. Loss of gata3 in inner hair cells causes progressive hearing loss and accounts for at least some of the deafness associated with the human hypoparathyroidism, deafness and renal anomaly (HDR) syndrome. The results show that gata3 is critical for later stages of mammalian auditory system development where it plays distinct, complementary roles in the coordinated maturation of sensory hair cells and their innervation. ABSTRACT The zinc finger transcription factor gata3 regulates inner ear development from the formation of the embryonic otic placode. Throughout development, gata3 is expressed dynamically in all the major cochlear cell types. Its role in afferent formation is well established but its possible involvement in hair cell maturation remains unknown. Here, we find that in heterozygous gata3 null mice (gata3+/- ) outer hair cells (OHCs) differentiate normally but their numbers are significantly lower. In contrast, inner hair cells (IHCs) survive normally but they fail to acquire adult basolateral membrane currents, retain pre-hearing current and efferent innervation profiles and have fewer ribbon synapses. Targeted deletion of gata3 driven by otoferlin-cre recombinase (gata3fl/fl otof-cre+/- ) in IHCs does not affect OHCs or the number of IHC afferent synapses but it leads to a failure in IHC maturation comparable to that observed in gata3+/- mice. Auditory brainstem responses in gata3fl/fl otof-cre+/- mice reveal progressive hearing loss that becomes profound by 6-7 months, whilst distortion product otoacoustic emissions are no different to control animals up to this age. Our results, alongside existing data, indicate that gata3 has specific, complementary functions in different cell types during inner ear development and that its continued expression in the sensory epithelium orchestrates critical aspects of physiological development and neural connectivity. Furthermore, our work indicates that hearing loss in human hypoparathyroidism, deafness and renal anomaly (HDR) syndrome arises from functional deficits in IHCs as well as loss of function from OHCs and both afferent and efferent neurons.
Collapse
MESH Headings
- Animals
- Cell Differentiation/physiology
- Cochlea/metabolism
- Cochlea/physiology
- GATA3 Transcription Factor/metabolism
- Hair Cells, Auditory, Inner/metabolism
- Hair Cells, Auditory, Inner/physiology
- Hair Cells, Auditory, Outer/metabolism
- Hair Cells, Auditory, Outer/physiology
- Hair Cells, Vestibular/metabolism
- Hair Cells, Vestibular/physiology
- Hearing/physiology
- Hearing Loss/metabolism
- Hearing Loss/physiopathology
- Membrane Proteins/metabolism
- Mice, Knockout
- Mice, Transgenic
- Sensory Receptor Cells/metabolism
- Sensory Receptor Cells/physiology
- Synapses/metabolism
Collapse
Affiliation(s)
- Tanaya Bardhan
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | - Jing‐Yi Jeng
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | - Marco Waldmann
- Department of OtolaryngologyTübingen Hearing Research CenterSection of Physiological Acoustics and CommunicationUniversity of Tübingen72076TübingenGermany
| | - Federico Ceriani
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | | | - Jennifer Olt
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | - Lukas Rüttiger
- Department of OtolaryngologyTübingen Hearing Research CenterSection of Physiological Acoustics and CommunicationUniversity of Tübingen72076TübingenGermany
| | - Walter Marcotti
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | | |
Collapse
|
23
|
Eckrich S, Hecker D, Sorg K, Blum K, Fischer K, Münkner S, Wenzel G, Schick B, Engel J. Cochlea-Specific Deletion of Ca v1.3 Calcium Channels Arrests Inner Hair Cell Differentiation and Unravels Pitfalls of Conditional Mouse Models. Front Cell Neurosci 2019; 13:225. [PMID: 31178698 PMCID: PMC6538774 DOI: 10.3389/fncel.2019.00225] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 05/03/2019] [Indexed: 12/29/2022] Open
Abstract
Inner hair cell (IHC) Cav1.3 Ca2+ channels are multifunctional channels mediating Ca2+ influx for exocytosis at ribbon synapses, the generation of Ca2+ action potentials in pre-hearing IHCs and gene expression. IHCs of deaf systemic Cav1.3-deficient (Cav1.3-/-) mice stay immature because they fail to up-regulate voltage- and Ca2+-activated K+ (BK) channels but persistently express small conductance Ca2+-activated K+ (SK2) channels. In pre-hearing wildtype mice, cholinergic neurons from the superior olivary complex (SOC) exert efferent inhibition onto spontaneously active immature IHCs by activating their SK2 channels. Because Cav1.3 plays an important role for survival, health and function of SOC neurons, SK2 channel persistence and lack of BK channels in systemic Cav1.3-/- IHCs may result from malfunctioning neurons of the SOC. Here we analyze cochlea-specific Cav1.3 knockout mice with green fluorescent protein (GFP) switch reporter function, Pax2::cre;Cacna1d-eGFPflex/flexand Pax2::cre;Cacna1d-eGFPflex/-. Profound hearing loss, lack of BK channels and persistence of SK2 channels in Pax2::cre;Cacna1d-eGFPflex/- mice recapitulated the phenotype of systemic Cav1.3-/- mice, indicating that in wildtype mice, regulation of SK2 and BK channel expression is independent of Cav1.3 expression in SOC neurons. In addition, we noticed dose-dependent GFP toxicity leading to death of basal coil IHCs of Pax2::cre;Cacna1d-eGFPflex/flex mice, likely because of high GFP concentration and small repair capacity. This and the slower time course of Pax2-driven Cre recombinase in switching two rather than one Cacna1d-eGFPflex allele lead us to study Pax2::cre;Cacna1d-eGFPflex/- mice. Notably, control Cacna1d-eGFPflex/- IHCs showed a significant reduction in Cav1.3 channel cluster sizes and currents, suggesting that the intronic construct interfered with gene translation or splicing. These pitfalls are likely to be a frequent problem of many genetically modified mice with complex or multiple gene-targeting constructs or fluorescent proteins. Great caution and appropriate controls are therefore required.
Collapse
Affiliation(s)
- Stephanie Eckrich
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Dietmar Hecker
- Department of Otorhinolaryngology, Saarland University, Homburg, Germany
| | - Katharina Sorg
- Department of Otorhinolaryngology, Saarland University, Homburg, Germany
| | - Kerstin Blum
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Kerstin Fischer
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Stefan Münkner
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Gentiana Wenzel
- Department of Otorhinolaryngology, Saarland University, Homburg, Germany
| | - Bernhard Schick
- Department of Otorhinolaryngology, Saarland University, Homburg, Germany
| | - Jutta Engel
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| |
Collapse
|
24
|
Schlosser G. A Short History of Nearly Every Sense-The Evolutionary History of Vertebrate Sensory Cell Types. Integr Comp Biol 2019; 58:301-316. [PMID: 29741623 DOI: 10.1093/icb/icy024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Evolving from filter feeding chordate ancestors, vertebrates adopted a more active life style. These ecological and behavioral changes went along with an elaboration of the vertebrate head including novel complex paired sense organs such as the eyes, inner ears, and olfactory epithelia. However, the photoreceptors, mechanoreceptors, and chemoreceptors used in these sense organs have a long evolutionary history and homologous cell types can be recognized in many other bilaterians or even cnidarians. After briefly introducing some of the major sensory cell types found in vertebrates, this review summarizes the phylogenetic distribution of sensory cell types in metazoans and presents a scenario for the evolutionary history of various sensory cell types involving several cell type diversification and fusion events. It is proposed that the evolution of novel cranial sense organs in vertebrates involved the redeployment of evolutionarily ancient sensory cell types for building larger and more complex sense organs.
Collapse
Affiliation(s)
- Gerhard Schlosser
- School of Natural Sciences and Regenerative Medicine Institute (REMEDI), National University of Ireland, Biomedical Sciences Building, Newcastle Road, Galway H91 TK33, Ireland
| |
Collapse
|
25
|
Zaidan N, Ottersbach K. The multi-faceted role of Gata3 in developmental haematopoiesis. Open Biol 2018; 8:rsob.180152. [PMID: 30463912 PMCID: PMC6282070 DOI: 10.1098/rsob.180152] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 10/29/2018] [Indexed: 12/22/2022] Open
Abstract
The transcription factor Gata3 is crucial for the development of several tissues and cell lineages both during development as well as postnatally. This importance is apparent from the early embryonic lethality following germline Gata3 deletion, with embryos displaying a number of phenotypes, and from the fact that Gata3 has been implicated in several cancer types. It often acts at the level of stem and progenitor cells in which it controls the expression of key lineage-determining factors as well as cell cycle genes, thus being one of the main drivers of cell fate choice and tissue morphogenesis. Gata3 is involved at various stages of haematopoiesis both in the adult as well as during development. This review summarizes the various contributions of Gata3 to haematopoiesis with a particular focus on the emergence of the first haematopoietic stem cells in the embryo—a process that appears to be influenced by Gata3 at various levels, thus highlighting the complex nature of Gata3 action.
Collapse
Affiliation(s)
- Nada Zaidan
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK.,King Abdullah International Medical Research Centre, Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Katrin Ottersbach
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| |
Collapse
|
26
|
Martynova E, Bouchard M, Musil LS, Cvekl A. Identification of Novel Gata3 Distal Enhancers Active in Mouse Embryonic Lens. Dev Dyn 2018; 247:1186-1198. [PMID: 30295986 PMCID: PMC6246825 DOI: 10.1002/dvdy.24677] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/30/2018] [Accepted: 10/01/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The tissue-specific transcriptional programs during normal development require tight control by distal cis-regulatory elements, such as enhancers, with specific DNA sequences recognized by transcription factors, coactivators, and chromatin remodeling enzymes. Gata3 is a sequence-specific DNA-binding transcription factor that regulates formation of multiple tissues and organs, including inner ear, lens, mammary gland, T-cells, urogenital system, and thyroid gland. In the eye, Gata3 has a highly restricted expression domain in the posterior part of the lens vesicle; however, the underlying regulatory mechanisms are unknown. RESULTS Here we describe the identification of a novel bipartite Gata3 lens-specific enhancer located ∼18 kb upstream from its transcriptional start site. We also found that a 5-kb Gata3 promoter possesses low activity in the lens. The bipartite enhancer contains arrays of AP-1, Ets-, and Smad1/5-binding sites as well as binding sites for lens-associated DNA-binding factors. Transient transfection studies of the promoter with the bipartite enhancer showed enhanced activation by BMP4 and FGF2. CONCLUSIONS These studies identify a novel distal enhancer of Gata3 with high activity in lens and indicate that BMP and FGF signaling can up-regulate expression of Gata3 in differentiating lens fiber cells through the identified Gata3 enhancer and promoter elements. Developmental Dynamics 247:1186-1198, 2018. © 2018 The Authors. Developmental Dynamics published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.
Collapse
Affiliation(s)
- Elena Martynova
- Departments of Ophthalmology and Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Maxime Bouchard
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Linda S Musil
- Department of Biochemistry and Molecular Biology, Oregon Health Science University, Portland, Oregon
| | - Ales Cvekl
- Departments of Ophthalmology and Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
27
|
A Gata3 3' Distal Otic Vesicle Enhancer Directs Inner Ear-Specific Gata3 Expression. Mol Cell Biol 2018; 38:MCB.00302-18. [PMID: 30126893 DOI: 10.1128/mcb.00302-18] [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] [Received: 06/21/2018] [Accepted: 08/13/2018] [Indexed: 12/21/2022] Open
Abstract
Transcription factor GATA3 plays vital roles in inner ear development, while regulatory mechanisms controlling its inner ear-specific expression are undefined. We demonstrate that a cis-regulatory element lying 571 kb 3' to the Gata3 gene directs inner ear-specific Gata3 expression, which we refer to as the Gata3 otic vesicle enhancer (OVE). In transgenic murine embryos, a 1.5-kb OVE-directed lacZ reporter (TgOVE-LacZ) exhibited robust lacZ expression specifically in the otic vesicle (OV), an inner ear primordial tissue, and its derivative semicircular canal. To further define the regulatory activity of this OVE, we generated Cre transgenic mice in which Cre expression was directed by a 246-bp core sequence within the OVE element (TgcoreOVE-Cre). TgcoreOVE-Cre successfully marked the OV-derived inner ear tissues, including cochlea, semicircular canal and spiral ganglion, when crossed with ROSA26 lacZ reporter mice. Furthermore, Gata3 conditionally mutant mice, when crossed with the TgcoreOVE-Cre, showed hypoplasia throughout the inner ear tissues. These results demonstrate that OVE has a sufficient regulatory activity to direct Gata3 expression specifically in the otic vesicle and semicircular canal and that Gata3 expression driven by the OVE is crucial for normal inner ear development.
Collapse
|
28
|
The Key Transcription Factor Expression in the Developing Vestibular and Auditory Sensory Organs: A Comprehensive Comparison of Spatial and Temporal Patterns. Neural Plast 2018; 2018:7513258. [PMID: 30410537 PMCID: PMC6205106 DOI: 10.1155/2018/7513258] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/31/2018] [Accepted: 09/06/2018] [Indexed: 11/17/2022] Open
Abstract
Inner ear formation requires that a series of cell fate decisions and morphogenetic events occur in a precise temporal and spatial pattern. Previous studies have shown that transcription factors, including Pax2, Sox2, and Prox1, play important roles during the inner ear development. However, the temporospatial expression patterns among these transcription factors are poorly understood. In the current study, we present a comprehensive description of the temporal and spatial expression profiles of Pax2, Sox2, and Prox1 during auditory and vestibular sensory organ development in mice. Using immunohistochemical analyses, we show that Sox2 and Pax2 are both expressed in the prosensory cells (the developing hair cells), but Sox2 is later restricted to only the supporting cells of the organ of Corti. In the vestibular sensory organ, however, the Pax2 expression is localized in hair cells at postnatal day 7, while Sox2 is still expressed in both the hair cells and supporting cells at that time. Prox1 was transiently expressed in the presumptive hair cells and developing supporting cells, and lower Prox1 expression was observed in the vestibular sensory organ compared to the organ of Corti. The different expression patterns of these transcription factors in the developing auditory and vestibular sensory organs suggest that they play different roles in the development of the sensory epithelia and might help to shape the respective sensory structures.
Collapse
|
29
|
Li X, Jin J, Yang S, Xu W, Meng X, Deng H, Zhan J, Gao S, Zhang H. GATA3 acetylation at K119 by CBP inhibits cell migration and invasion in lung adenocarcinoma. Biochem Biophys Res Commun 2018; 497:633-638. [PMID: 29453984 DOI: 10.1016/j.bbrc.2018.02.120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 02/13/2018] [Indexed: 12/12/2022]
Abstract
GATA3 is a transcriptional factor involved in the development of multiple organs. Post translational modifications of GATA3 are critical to its function. Here, we report that GATA3 interacts with and is acetylated by the acetyltransferase CBP. Class I deacetylases HDAC1, HDAC2 and HDAC3 deacetylate GATA3. The major acetylated site of GATA3 in lung adenocarcinoma cells was determined at lysine 119 (AcK119). Functionally, GATA3-acetylation mimics K119Q mutant was found to inhibit lung adenocarcinoma cell migration and invasion with concomitant downregulation of EMT-controlling transcriptional factors Slug, Zeb1 and Zeb2. Taken together, we demonstrated that GATA3 acetylation at lysine 119 by CBP hinders the migration and invasion of lung adenocarcinoma cells.
Collapse
Affiliation(s)
- Xueying Li
- Department of Anatomy, Histology and Embryology, MOE Key Laboratory of Carcinogenesis and Translational Research, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191, China
| | - Jiaqi Jin
- Department of Anatomy, Histology and Embryology, MOE Key Laboratory of Carcinogenesis and Translational Research, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191, China
| | - Siyuan Yang
- Department of Anatomy, Histology and Embryology, MOE Key Laboratory of Carcinogenesis and Translational Research, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191, China
| | - Weizhi Xu
- Department of Anatomy, Histology and Embryology, MOE Key Laboratory of Carcinogenesis and Translational Research, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191, China
| | - Xianbin Meng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jun Zhan
- Department of Anatomy, Histology and Embryology, MOE Key Laboratory of Carcinogenesis and Translational Research, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191, China
| | - Shan Gao
- CAS Key Laboratory of Bio-medical Diagnostic, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China.
| | - Hongquan Zhang
- Department of Anatomy, Histology and Embryology, MOE Key Laboratory of Carcinogenesis and Translational Research, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
30
|
Gou Y, Vemaraju S, Sweet EM, Kwon HJ, Riley BB. sox2 and sox3 Play unique roles in development of hair cells and neurons in the zebrafish inner ear. Dev Biol 2018; 435:73-83. [PMID: 29355523 DOI: 10.1016/j.ydbio.2018.01.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/15/2018] [Accepted: 01/15/2018] [Indexed: 11/24/2022]
Abstract
Formation of neural and sensory progenitors in the inner ear requires Sox2 in mammals, and in other species is thought to rely on both Sox2 and Sox3. How Sox2 and/or Sox3 promote different fates is poorly understood. Our mutant analysis in zebrafish showed that sox2 is uniquely required for sensory development while sox3 is uniquely required for neurogenesis. Moderate misexpression of sox2 during placodal stages led to development of otic vesicles with expanded sensory and reduced neurogenic domains. However, high-level misexpression of sox2 or sox3 expanded both sensory and neurogenic domains to fill the medial and lateral halves of the otic vesicle, respectively. Disruption of medial factor pax2a eliminated the ability of sox2/3 misexpression to expand sensory but not neurogenic domains. Additionally, mild misexpression of fgf8 during placodal development was sufficient to specifically expand the zone of prosensory competence. Later, cross-repression between atoh1a and neurog1 helps maintain the sensory-neural boundary, but unlike mouse this does not require Notch activity. Together, these data show that sox2 and sox3 exhibit intrinsic differences in promoting sensory vs. neural competence, but at high levels these factors can mimic each other to enhance both states. Regional cofactors like pax2a and fgf8 also modify sox2/3 functions.
Collapse
Affiliation(s)
- Yunzi Gou
- Department of Biology, Texas A&M University, College Station, TX 77843-3258, USA
| | - Shruti Vemaraju
- Department of Biology, Texas A&M University, College Station, TX 77843-3258, USA
| | - Elly M Sweet
- Department of Biology, Texas A&M University, College Station, TX 77843-3258, USA
| | - Hye-Joo Kwon
- Department of Biology, Texas A&M University, College Station, TX 77843-3258, USA
| | - Bruce B Riley
- Department of Biology, Texas A&M University, College Station, TX 77843-3258, USA.
| |
Collapse
|
31
|
Schaefer SA, Higashi AY, Loomis B, Schrepfer T, Wan G, Corfas G, Dressler GR, Duncan RK. From Otic Induction to Hair Cell Production: Pax2 EGFP Cell Line Illuminates Key Stages of Development in Mouse Inner Ear Organoid Model. Stem Cells Dev 2018; 27:237-251. [PMID: 29272992 DOI: 10.1089/scd.2017.0142] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Producing hair cells of the inner ear is the major goal of ongoing research that combines advances in developmental and stem cell biology. The recent advent of an inner ear organoid protocol-resulting in three-dimensional stem cell-derived tissues resembling vestibular sensory epithelia-has sparked interest in applications such as regeneration, drug discovery, and disease modeling. In this study, we adapted this protocol for a novel mouse embryonic stem cell line with a fluorescent reporter for Pax2 expression. We used Pax2EGFP/+ organoid formation to model otic induction, the pivotal developmental event when preplacodal tissue adopts otic fate. We found upregulation of Pax2 and activation of ERK downstream of fibroblast growth factor signaling in organoid formation as in embryonic inner ear development. Pax2 expression was evident from the EGFP reporter beginning at the vesicle formation stage and persisting through generation of the sensory epithelium. The native ventralizing signal sonic hedgehog was largely absent from the cell aggregates as otic vesicles began to form, confirming the dorsal vestibular organoid fate. Nonetheless, cochlear- or vestibular-like neurons appeared to delaminate from the derived otic vesicles and formed synaptic contacts with hair cells in the organoids. Cell lines with transcriptional reporters such as Pax2EGFP/+ facilitate direct evaluation of morphological changes during organoid production, a major asset when establishing and validating the culture protocol.
Collapse
Affiliation(s)
- Stacy A Schaefer
- 1 Department of Otolaryngology, Kresge Hearing Research Institute, University of Michigan , Ann Arbor, Michigan
| | - Atsuko Y Higashi
- 2 Department of Pathology, University of Michigan , Ann Arbor, Michigan
| | - Benjamin Loomis
- 1 Department of Otolaryngology, Kresge Hearing Research Institute, University of Michigan , Ann Arbor, Michigan
| | - Thomas Schrepfer
- 1 Department of Otolaryngology, Kresge Hearing Research Institute, University of Michigan , Ann Arbor, Michigan
| | - Guoqiang Wan
- 1 Department of Otolaryngology, Kresge Hearing Research Institute, University of Michigan , Ann Arbor, Michigan
| | - Gabriel Corfas
- 1 Department of Otolaryngology, Kresge Hearing Research Institute, University of Michigan , Ann Arbor, Michigan
| | | | - Robert Keith Duncan
- 1 Department of Otolaryngology, Kresge Hearing Research Institute, University of Michigan , Ann Arbor, Michigan
| |
Collapse
|
32
|
The histone demethylase LSD1 regulates inner ear progenitor differentiation through interactions with Pax2 and the NuRD repressor complex. PLoS One 2018; 13:e0191689. [PMID: 29370269 PMCID: PMC5784988 DOI: 10.1371/journal.pone.0191689] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/09/2018] [Indexed: 01/07/2023] Open
Abstract
The histone demethylase LSD1 plays a pivotal role in cellular differentiation, particularly in silencing lineage-specific genes. However, little is known about how LSD1 regulates neurosensory differentiation in the inner ear. Here we show that LSD1 interacts directly with the transcription factor Pax2 to form the NuRD co-repressor complex at the Pax2 target gene loci in a mouse otic neuronal progenitor cell line (VOT-N33). VOT-N33 cells expressing a Pax2-response element reporter were GFP-negative when untreated, but became GFP positive after forced differentiation or treatment with a potent LSD inhibitor. Pharmacological inhibition of LSD1 activity resulted in the enrichment of mono- and di-methylation of H3K4, upregulation of sensory neuronal genes and an increase in the number of sensory neurons in mouse inner ear organoids. Together, these results identify the LSD1/NuRD complex as a previously unrecognized modulator for Pax2-mediated neuronal differentiation in the inner ear.
Collapse
|
33
|
Di Bonito M, Studer M. Cellular and Molecular Underpinnings of Neuronal Assembly in the Central Auditory System during Mouse Development. Front Neural Circuits 2017; 11:18. [PMID: 28469562 PMCID: PMC5395578 DOI: 10.3389/fncir.2017.00018] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/01/2017] [Indexed: 11/13/2022] Open
Abstract
During development, the organization of the auditory system into distinct functional subcircuits depends on the spatially and temporally ordered sequence of neuronal specification, differentiation, migration and connectivity. Regional patterning along the antero-posterior axis and neuronal subtype specification along the dorso-ventral axis intersect to determine proper neuronal fate and assembly of rhombomere-specific auditory subcircuits. By taking advantage of the increasing number of transgenic mouse lines, recent studies have expanded the knowledge of developmental mechanisms involved in the formation and refinement of the auditory system. Here, we summarize several findings dealing with the molecular and cellular mechanisms that underlie the assembly of central auditory subcircuits during mouse development, focusing primarily on the rhombomeric and dorso-ventral origin of auditory nuclei and their associated molecular genetic pathways.
Collapse
|
34
|
GATA3 interacts with and stabilizes HIF-1α to enhance cancer cell invasiveness. Oncogene 2017; 36:4243-4252. [PMID: 28263977 PMCID: PMC5537608 DOI: 10.1038/onc.2017.8] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 12/23/2016] [Accepted: 01/11/2017] [Indexed: 12/11/2022]
Abstract
GATA binding protein 3 (GATA3) is indispensable in development of human organs. However, the role of GATA3 in cancers remains elusive. Hypoxia inducible factor (HIF)-1 plays an important role in pathogenesis of human cancers. Regulation of HIF-1α degradation is orchestrated through collaboration of its interacting proteins. In this study, we discover that GATA3 is upregulated in head and neck squamous cell carcinoma (HNSCC) and is an independent predictor for poor disease-free survival. GATA3 promotes invasive behaviours of HNSCC and melanoma cells in vitro and in immunodeficient mice. Mechanistically, GATA3 physically associates with HIF-1α under hypoxia to inhibit ubiquitination and proteasomal degradation of HIF-1α, which is independent of HIF-1α prolyl hydroxylation. Chromatin immunoprecipitation assays show that the GATA3/HIF-1α complex binds to and regulates HIF-1 target genes, which is also supported by the microarray analysis. Notably, the GATA3-mediated invasiveness can be significantly reversed by HIF-1α knockdown, suggesting a critical role of HIF-1α in the underlying mechanism of GATA3-mediated effects. Our findings suggest that GATA3 stabilizes HIF-1α to enhance cancer invasiveness under hypoxia and support the GATA3/HIF-1α axis as a potential therapeutic target for cancer treatment.
Collapse
|
35
|
Nishimura K, Noda T, Dabdoub A. Dynamic Expression of Sox2, Gata3, and Prox1 during Primary Auditory Neuron Development in the Mammalian Cochlea. PLoS One 2017; 12:e0170568. [PMID: 28118374 PMCID: PMC5261741 DOI: 10.1371/journal.pone.0170568] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 01/06/2017] [Indexed: 12/15/2022] Open
Abstract
Primary auditory neurons (PANs) connect cochlear sensory hair cells in the mammalian inner ear to cochlear nucleus neurons in the brainstem. PANs develop from neuroblasts delaminated from the proneurosensory domain of the otocyst and keep maturing until the onset of hearing after birth. There are two types of PANs: type I, which innervate the inner hair cells (IHCs), and type II, which innervate the outer hair cells (OHCs). Glial cells surrounding these neurons originate from neural crest cells and migrate to the spiral ganglion. Several transcription factors are known to regulate the development and differentiation of PANs. Here we systematically examined the spatiotemporal expression of five transcription factors: Sox2, Sox10, Gata3, Mafb, and Prox1 from early delamination at embryonic day (E) 10.5 to adult. We found that Sox2 and Sox10 were initially expressed in the proneurosensory cells in the otocyst (E10.5). By E12.75 both Sox2 and Sox10 were downregulated in the developing PANs; however, Sox2 expression transiently increased in the neurons around birth. Furthermore, both Sox2 and Sox10 continued to be expressed in spiral ganglion glial cells. We also show that Gata3 and Prox1 were first expressed in all developing neurons, followed by a decrease in expression of Gata3 and Mafb in type I PANs and Prox1 in type II PANs as they matured. Moreover, we describe two subtypes of type II neurons based on Peripherin expression. These results suggest that Sox2, Gata3 and Prox1 play a role during neurogenesis as well as maturation of the PANs.
Collapse
Affiliation(s)
- Koji Nishimura
- Shiga Medical Center Research Institute, Moriyama, Shiga, Japan
| | - Teppei Noda
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Alain Dabdoub
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Otolaryngology – Head & Neck Surgery, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
36
|
Differential effects of pannexins on noise-induced hearing loss. Biochem J 2016; 473:4665-4680. [PMID: 27784763 DOI: 10.1042/bcj20160668] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/14/2016] [Accepted: 10/24/2016] [Indexed: 12/20/2022]
Abstract
Hearing loss, including noise-induced hearing loss, is highly prevalent and severely hinders an individual's quality of life, yet many of the mechanisms that cause hearing loss are unknown. The pannexin (Panx) channel proteins, Panx1 and Panx3, are regionally expressed in many cell types along the auditory pathway, and mice lacking Panx1 in specific cells of the inner ear exhibit hearing loss, suggesting a vital role for Panxs in hearing. We proposed that Panx1 and/or Panx3 null mice would exhibit severe hearing loss and increased susceptibility to noise-induced hearing loss. Using the auditory brainstem response, we surprisingly found that Panx1-/- and Panx3-/- mice did not harbor hearing or cochlear nerve deficits. Furthermore, while Panx1-/- mice displayed no protection against loud noise-induced hearing loss, Panx3-/- mice exhibited enhanced 16- and 24-kHz hearing recovery 7 days after a loud noise exposure (NE; 12 kHz tone, 115 dB sound pressure level, 1 h). Interestingly, Cx26, Cx30, Cx43, and Panx2 were up-regulated in Panx3-/- mice compared with wild-type and/or Panx1-/- mice, and assessment of the auditory tract revealed morphological changes in the middle ear bones of Panx3-/- mice. It is unclear if these changes alone are sufficient to provide protection against loud noise-induced hearing loss. Contrary to what we expected, these data suggest that Panx1 and Panx3 are not essential for baseline hearing in mice tested, but the therapeutic targeting of Panx3 may prove protective against mid-high-frequency hearing loss caused by loud NE.
Collapse
|
37
|
Smith ME, Rajadinakaran G. The Transcriptomics to Proteomics of Hair Cell Regeneration: Looking for a Hair Cell in a Haystack. MICROARRAYS 2016; 2. [PMID: 24416530 PMCID: PMC3886832 DOI: 10.3390/microarrays2030186] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mature mammals exhibit very limited capacity for regeneration of auditory hair cells, while all non-mammalian vertebrates examined can regenerate them. In an effort to find therapeutic targets for deafness and balance disorders, scientists have examined gene expression patterns in auditory tissues under different developmental and experimental conditions. Microarray technology has allowed the large-scale study of gene expression profiles (transcriptomics) at whole-genome levels, but since mRNA expression does not necessarily correlate with protein expression, other methods, such as microRNA analysis and proteomics, are needed to better understand the process of hair cell regeneration. These technologies and some of the results of them are discussed in this review. Although there is a considerable amount of variability found between studies owing to different species, tissues and treatments, there is some concordance between cellular pathways important for hair cell regeneration. Since gene expression and proteomics data is now commonly submitted to centralized online databases, meta-analyses of these data may provide a better picture of pathways that are common to the process of hair cell regeneration and lead to potential therapeutics. Indeed, some of the proteins found to be regulated in the inner ear of animal models (e.g., IGF-1) have now gone through human clinical trials.
Collapse
Affiliation(s)
- Michael E. Smith
- Bioinformatics and Information Science Center, Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-270-745-2405; Fax: +1-270-745-6856
| | - Gopinath Rajadinakaran
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA; E-Mail:
| |
Collapse
|
38
|
Kwan KY. Single-Cell Transcriptome Analysis of Developing and Regenerating Spiral Ganglion Neurons. ACTA ACUST UNITED AC 2016; 2:211-220. [PMID: 28758056 DOI: 10.1007/s40495-016-0064-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The spiral ganglion neurons (SGNs) of the cochlea are essential for our ability to hear. SGN loss after exposure to ototoxic drugs or loud noise results in hearing loss. Pluripotent stem cell-derived and endogenous progenitor cell types have the potential to become SGNs and are cellular foundations for replacement therapies. Repurposing transcriptional regulatory networks to promote SGN differentiation from progenitor cells is a strategy for regeneration. Advances in the Fludigm C1 workflow or Drop-seq allow sequencing of single cell transcriptomes to reveal variability between cells. During differentiation, the individual transcriptomes obtained from single-cell RNA-seq can be exploited to identify different cellular states. Pseudotemporal ordering of transcriptomes describes the differentiation trajectory, allows monitoring of transcriptional changes and determines molecular barriers that prevent the progression of progenitors into SGNs. Analysis of single cell transcriptomes will help develop novel strategies for guiding efficient SGN regeneration.
Collapse
Affiliation(s)
- Kelvin Y Kwan
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, NJ 08854, USA.,Stem Cell Research Center and Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
39
|
Tang ZH, Chen JR, Zheng J, Shi HS, Ding J, Qian XD, Zhang C, Chen JL, Wang CC, Li L, Chen JZ, Yin SK, Huang TS, Chen P, Guan MX, Wang JF. Genetic Correction of Induced Pluripotent Stem Cells From a Deaf Patient With MYO7A Mutation Results in Morphologic and Functional Recovery of the Derived Hair Cell-Like Cells. Stem Cells Transl Med 2016; 5:561-71. [PMID: 27013738 DOI: 10.5966/sctm.2015-0252] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 12/22/2015] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED The genetic correction of induced pluripotent stem cells (iPSCs) induced from somatic cells of patients with sensorineural hearing loss (caused by hereditary factors) is a promising method for its treatment. The correction of gene mutations in iPSCs could restore the normal function of cells and provide a rich source of cells for transplantation. In the present study, iPSCs were generated from a deaf patient with compound heterozygous MYO7A mutations (c.1184G>A and c.4118C>T; P-iPSCs), the asymptomatic father of the patient (MYO7A c.1184G>A mutation; CF-iPSCs), and a normal donor (MYO7A(WT/WT); C-iPSCs). One of MYO7A mutation sites (c.4118C>T) in the P-iPSCs was corrected using CRISPR/Cas9. The corrected iPSCs (CP-iPSCs) retained cell pluripotency and normal karyotypes. Hair cell-like cells induced from CP-iPSCs showed restored organization of stereocilia-like protrusions; moreover, the electrophysiological function of these cells was similar to that of cells induced from C-iPSCs and CF-iPSCs. These results might facilitate the development of iPSC-based gene therapy for genetic disorders. SIGNIFICANCE Induced pluripotent stem cells (iPSCs) were generated from a deaf patient with compound heterozygous MYO7A mutations (c.1184G>A and c.4118C>T). One of the MYO7A mutation sites (c.4118C>T) in the iPSCs was corrected using CRISPR/Cas9. The genetic correction of MYO7A mutation resulted in morphologic and functional recovery of hair cell-like cells derived from iPSCs. These findings confirm the hypothesis that MYO7A plays an important role in the assembly of stereocilia into stereociliary bundles. Thus, the present study might provide further insight into the pathogenesis of sensorineural hearing loss and facilitate the development of therapeutic strategies against monogenic disease through the genetic repair of patient-specific iPSCs.
Collapse
Affiliation(s)
- Zi-Hua Tang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jia-Rong Chen
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jing Zheng
- Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Hao-Song Shi
- Department of Otorhinolaryngology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai, People's Republic of China
| | - Jie Ding
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Xiao-Dan Qian
- The Affiliated Women's Hospital, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Cui Zhang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jian-Ling Chen
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Cui-Cui Wang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Liang Li
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jun-Zhen Chen
- Department of Otolaryngology, The Affiliated Wenling People's Hospital, Wenzhou Medical University, Wenling, Zhejiang, People's Republic of China
| | - Shan-Kai Yin
- Department of Otorhinolaryngology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai, People's Republic of China
| | - Tao-Sheng Huang
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Ping Chen
- Departments of Cell Biology and Otolaryngology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Min-Xin Guan
- Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jin-Fu Wang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
40
|
Kempfle JS, Turban JL, Edge ASB. Sox2 in the differentiation of cochlear progenitor cells. Sci Rep 2016; 6:23293. [PMID: 26988140 PMCID: PMC4796895 DOI: 10.1038/srep23293] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 03/02/2016] [Indexed: 12/19/2022] Open
Abstract
HMG domain transcription factor, Sox2, is a critical gene for the development of cochlear hair cells, the receptor cells for hearing, but this has been ascribed to expansion of the progenitors that become hair cells. Here, we show that Sox2 activated Atoh1, a transcription factor important for hair cell differentiation, through an interaction with the 3′ enhancer of Atoh1. Binding to consensus sequences in the Atoh1 enhancer was dependent on the level of Sox2, and the extent of enhancer binding correlated to the extent of activation. Atoh1 activation by Sox2 was required for embryonic hair cell development: deletion of Sox2 in an inducible mutant, even after progenitor cells were fully established, halted development of hair cells, and silencing also inhibited postnatal differentiation of hair cells induced by inhibition of γ-secretase. Sox2 is thus required in the cochlea to both expand the progenitor cells and initiate their differentiation to hair cells.
Collapse
Affiliation(s)
- Judith S Kempfle
- Department of Otology and Laryngology, Harvard Medical School, Boston, MA 02115, USA.,Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA
| | - Jack L Turban
- Department of Otology and Laryngology, Harvard Medical School, Boston, MA 02115, USA.,Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA
| | - Albert S B Edge
- Department of Otology and Laryngology, Harvard Medical School, Boston, MA 02115, USA.,Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA.,Program in Speech and Hearing Bioscience and Technology, Division of Health Science and Technology, Harvard &MIT, Cambridge, MA 02139, USA
| |
Collapse
|
41
|
Goodrich LV. Early Development of the Spiral Ganglion. THE PRIMARY AUDITORY NEURONS OF THE MAMMALIAN COCHLEA 2016. [DOI: 10.1007/978-1-4939-3031-9_2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
42
|
Lorenzen SM, Duggan A, Osipovich AB, Magnuson MA, García-Añoveros J. Insm1 promotes neurogenic proliferation in delaminated otic progenitors. Mech Dev 2015; 138 Pt 3:233-45. [PMID: 26545349 DOI: 10.1016/j.mod.2015.11.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 10/02/2015] [Accepted: 11/02/2015] [Indexed: 01/12/2023]
Abstract
INSM1 is a zinc-finger protein expressed throughout the developing nervous system in late neuronal progenitors and nascent neurons. In the embryonic cortex and olfactory epithelium, Insm1 may promote the transition of progenitors from apical, proliferative, and uncommitted to basal, terminally-dividing and neuron producing. In the otocyst, delaminating and delaminated progenitors express Insm1, whereas apically-dividing progenitors do not. This expression pattern is analogous to that in embryonic olfactory epithelium and cortex (basal/subventricular progenitors). Lineage analysis confirms that auditory and vestibular neurons originate from Insm1-expressing cells. In the absence of Insm1, otic ganglia are smaller, with 40% fewer neurons. Accounting for the decrease in neurons, delaminated progenitors undergo fewer mitoses, but there is no change in apoptosis. We conclude that in the embryonic inner ear, Insm1 promotes proliferation of delaminated neuronal progenitors and hence the production of neurons, a similar function to that in other embryonic neural epithelia. Unexpectedly, we also found that differentiating, but not mature, outer hair cells express Insm1, whereas inner hair cells do not. Insm1 is the earliest known gene expressed in outer versus inner hair cells, demonstrating that nascent outer hair cells initiate a unique differentiation program in the embryo, much earlier than previously believed.
Collapse
Affiliation(s)
- Sarah M Lorenzen
- Department of Anesthesiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Anne Duggan
- Department of Anesthesiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Anna B Osipovich
- Center for Stem Cell Biology, Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Mark A Magnuson
- Center for Stem Cell Biology, Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jaime García-Añoveros
- Department of Anesthesiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Departments of Neurology and Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Hugh Knowles Center for Clinical and Basic Science in Hearing and Its Disorders, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
43
|
Abstract
OBJECTIVE To report the auditory and vestibular phenotypes of patients with GATA3 mutation. STUDY DESIGN Case series of 6 patients. SETTING Tertiary referral center. PATIENTS All patients had the classic triad of GATA3 deficiency: hypoparathyroidism, hearing loss, and renal dysplasia. Patients (29-60 yr old; mean age, 42.5 yr; 3 male and 3 female subjects) were confirmed to have heterozygous mutations involving GATA3 by Sanger sequencing. INTERVENTIONS Behavioral audiometry, distortion product otoacoustic emissions (DPOAEs), and auditory brainstem responses (ABRs) were used to assess hearing. Rotational vestibular testing was used to assess vestibular function. RESULTS All patients with GATA3 mutation presented with hearing loss during childhood. The mean 3-frequency (0.5/1/2 kHz) pure tone average was 67 dB HL (range, 50-83 dB HL; SD, 9.3). The average speech discrimination score was 73% (range, 36%-100%; SD, 15.9). DPOAEs were absent in all patients. ABRs were remarkably robust and provided no evidence of retrocochlear dysfunction. Some patients complained of dizziness, but rotary chair testing was normal across participants for whom testing occurred. CONCLUSION Patients with GATA3 mutation present with early-onset sensorineural hearing loss (SNHL). DPOAEs were absent, supporting outer hair cell dysfunction, whereas ABRs were present and robust. Rotational vestibular testing revealed no evidence of abnormal horizontal semicircular canal function.
Collapse
|
44
|
Comparative expression analysis of POU4F1, POU4F2 and ISL1 in developing mouse cochleovestibular ganglion neurons. Gene Expr Patterns 2014; 15:31-7. [PMID: 24709358 DOI: 10.1016/j.gep.2014.03.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 03/10/2014] [Accepted: 03/13/2014] [Indexed: 12/26/2022]
Abstract
POU-homeodomain and LIM-homeodomain transcription factors are expressed in developing projection neurons within retina, inner ear, dorsal root ganglion, and trigeminal ganglion, and play synergistic roles in their differentiation and survival. Here, using immunohistochemistry, we present a comparative analysis of the spatiotemporal expression pattern of POU4F1, POU4F2, and ISL1 during the development of cochleovestibular ganglion (CVG) neurons in mouse inner ear. At early stages, when otic neurons are first detected in the otic epithelium (OE) and migrate into periotic mesenchyme to form the CVG, POU4F1 and ISL1 are co-expressed in a majority of the delaminated CVG neurons, which are marked by NEUROD1 expression, but POU4F1 is absent in the otic epithelium. The onset of POU4F2 expression starts after that of POU4F1 and ISL1, and is observed in the NEUROD1-negative, post-mitotic CVG neurons. When the CVG neurons innervate the vestibular and cochlear sensory organs, the expression of POU4F1, POU4F2, and ISL1 continues in both vestibular and spiral ganglion cells. Later in development, POU4F1 expression becomes down-regulated in a majority of spiral ganglion (SG) neurons and more neurons express POU4F2 expression while ISL1 expression is maintained. The differential as well as overlapping expression of POU4F1, POU4F2, and ISL1 combined with previous studies suggests possible functional interaction and regulatory relationship of these transcription factors in the development of inner ear neurons.
Collapse
|
45
|
van Beelen E, Leijendeckers JM, Admiraal RJC, Huygen PLM, Hoefsloot LH, Pennings RJE, Snik AFM, Kunst HPM. Audiometric characteristics of a dutch family with a new mutation in GATA3 causing HDR syndrome. Audiol Neurootol 2014; 19:106-14. [PMID: 24434941 DOI: 10.1159/000356303] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 10/09/2013] [Indexed: 11/19/2022] Open
Abstract
We present the case of a Dutch family with a new mutation (c523_528dup) in GATA3 causing HDR syndrome. HDR syndrome is characterised by hypoparathyroidism, deafness and renal defects. In this study, we describe the audiometric characteristics of 5 patients from this family. Their hearing impairment was congenital, bilateral and symmetric. Audiograms showed mild-to-moderate hearing impairment with a flat audiogram configuration. Higher frequencies tended to be affected more strongly. Cross-sectional analyses showed no progression, and a mean audiogram was established. Psychophysical measurements in 3 HDR patients - including speech reception in noise, loudness scaling, gap detection and difference limen for frequency - were obtained to assess hearing function in greater detail. Overall, the results of the psychophysical measurements indicated characteristics of outer hair cell loss. CT scanning showed no anomalies in 3 of the HDR patients. Although 2 patients displayed vestibular symptoms, no anomalies in the vestibular system were found by vestibulo-ocular examination. Our results are in agreement with the theory that outer hair cell malfunctioning can play a major role in HDR syndrome.
Collapse
Affiliation(s)
- E van Beelen
- Department of Otorhinolaryngology, Head and Neck Surgery, Radboud University, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Chen Y, Yu H, Zhang Y, Li W, Lu N, Ni W, He Y, Li J, Sun S, Wang Z, Li H. Cotransfection of Pax2 and Math1 promote in situ cochlear hair cell regeneration after neomycin insult. Sci Rep 2013; 3:2996. [PMID: 24141260 PMCID: PMC3801138 DOI: 10.1038/srep02996] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 10/02/2013] [Indexed: 01/08/2023] Open
Abstract
The ideal strategy for hair cell regeneration is promoting residual supporting cell proliferation followed by induction of hair cell differentiation. In this study, cultured neonatal mouse organs of Corti were treated with neomycin to eliminate hair cells followed by incubation with recombined adenovirus expressing Pax2 and/or Math1. Results showed that overexpression of Pax2 significantly promoted proliferation of supporting cells. The number of BrdU+/myosin VIIA+ cells increased significantly in hair cell pre-existing region two weeks after adenovirus infection in Ad-Pax2-IRES-Math1 group compared to Ad-Pax2 and Ad-Math1 groups. This indicated that cotransfection of Pax2 and Math1 induced supporting cells to proliferate and differentiate into hair cells in situ. Most new hair cells were labeled by FM1-43 suggesting they acquired certain function. The results also suggest that inducing proliferating cells rather than quiescent cells to differentiate into hair cells by forced expression of Math1 is feasible for mammalian hair cell regeneration.
Collapse
Affiliation(s)
- Yan Chen
- 1] Otology Skull base Surgery Department, Hearing Research Institute, Eye and ENT Hospital of Shanghai Medical School, Fudan University. Shanghai, 200031, P.R.China [2] Central laboratory, Eye and ENT Hospital of Shanghai Medical School, Fudan University. Shanghai, 200031, P.R.China [3]
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Chervenak AP, Hakim IS, Barald KF. Spatiotemporal expression of Zic genes during vertebrate inner ear development. Dev Dyn 2013; 242:897-908. [PMID: 23606270 DOI: 10.1002/dvdy.23978] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 04/09/2013] [Accepted: 04/10/2013] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Inner ear development involves signaling from surrounding tissues, including the adjacent hindbrain, periotic mesenchyme, and notochord. These signals include SHH, FGFs, BMPs, and WNTs from the hindbrain and SHH from the notochord. Zic genes, which are expressed in the dorsal neural tube and act during neural development, have been implicated as effectors of these pathways. This report examines whether Zic genes' involvement in inner ear development is a tenable hypothesis based on their expression patterns. RESULTS In the developing inner ear of both the chick and mouse, all of the Zic genes were expressed in the dorsal neural tube and variably in the periotic mesenchyme, but expression of the Zic genes in the otic epithelium was not found. The onset of expression differed among the Zic genes; within any given region surrounding the otic epithelium, multiple Zic genes were expressed in the same place at the same time. CONCLUSIONS Zic gene expression in the region of the developing inner ear is similar between mouse and chick. Zic expression domains overlap with sites of WNT and SHH signaling during otocyst patterning, suggesting a role for Zic genes in modulating signaling from these pathways.
Collapse
Affiliation(s)
- Andrew P Chervenak
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | |
Collapse
|
48
|
Luo XJ, Deng M, Xie X, Huang L, Wang H, Jiang L, Liang G, Hu F, Tieu R, Chen R, Gan L. GATA3 controls the specification of prosensory domain and neuronal survival in the mouse cochlea. Hum Mol Genet 2013; 22:3609-23. [PMID: 23666531 DOI: 10.1093/hmg/ddt212] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
HDR syndrome (also known as Barakat syndrome) is a developmental disorder characterized by hypoparathyroidism, sensorineural deafness and renal disease. Although genetic mapping and subsequent functional studies indicate that GATA3 haplo-insufficiency causes human HDR syndrome, the role of Gata3 in sensorineural deafness and auditory system development is largely unknown. In this study, we show that Gata3 is continuously expressed in the developing mouse inner ear. Conditional knockout of Gata3 in the developing inner ear disrupts the morphogenesis of mouse inner ear, resulting in a disorganized and shortened cochlear duct with significant fewer hair cells and supporting cells. Loss of Gata3 function leads to the failure in the specification of prosensory domain and subsequently, to increased cell death in the cochlear duct. Moreover, though the initial generation of cochleovestibular ganglion (CVG) cells is not affected in Gata3-null mice, spiral ganglion neurons (SGNs) are nearly depleted due to apoptosis. Our results demonstrate the essential role of Gata3 in specifying the prosensory domain in the cochlea and in regulating the survival of SGNs, thus identifying a molecular mechanism underlying human HDR syndrome.
Collapse
Affiliation(s)
- Xiong-jian Luo
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Continued expression of GATA3 is necessary for cochlear neurosensory development. PLoS One 2013; 8:e62046. [PMID: 23614009 PMCID: PMC3628701 DOI: 10.1371/journal.pone.0062046] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 03/18/2013] [Indexed: 01/19/2023] Open
Abstract
Hair cells of the developing mammalian inner ear are progressively defined through cell fate restriction. This process culminates in the expression of the bHLH transcription factor Atoh1, which is necessary for differentiation of hair cells, but not for their specification. Loss of several genes will disrupt ear morphogenesis or arrest of neurosensory epithelia development. We previously showed in null mutants that the loss of the transcription factor, Gata3, results specifically in the loss of all cochlear neurosensory development. Temporal expression of Gata3 is broad from the otic placode stage through the postnatal ear. It therefore remains unclear at which stage in development Gata3 exerts its effect. To better understand the stage specific effects of Gata3, we investigated the role of Gata3 in cochlear neurosensory specification and differentiation utilizing a LoxP targeted Gata3 line and two Cre lines. Foxg1Cre∶Gata3f/f mice show recombination of Gata3 around E8.5 but continue to develop a cochlear duct without differentiated hair cells and spiral ganglion neurons. qRT-PCR data show that Atoh1 was down-regulated but not absent in the duct whereas other hair cell specific genes such as Pou4f3 were completely absent. In addition, while Sox2 levels were lower in the Foxg1Cre:Gata3f/f cochlea, Eya1 levels remained normal. We conclude that Eya1 is unable to fully upregulate Atoh1 or Pou4f3, and drive differentiation of hair cells without Gata3. Pax2-Cre∶Gata3f/f mice show a delayed recombination of Gata3 in the ear relative to Foxg1Cre:Gata3f/f. These mice exhibited a cochlear duct containing patches of partially differentiated hair cells and developed only few and incorrectly projecting spiral ganglion neurons. Our conditional deletion studies reveal a major role of Gata3 in the signaling of prosensory genes and in the differentiation of cochlear neurosenory cells. We suggest that Gata3 may act in combination with Eya1, Six1, and Sox2 in cochlear prosensory gene signaling.
Collapse
|
50
|
Gata3 directly regulates early inner ear expression of Fgf10. Dev Biol 2013; 374:210-22. [DOI: 10.1016/j.ydbio.2012.11.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 11/23/2012] [Accepted: 11/26/2012] [Indexed: 01/19/2023]
|