1
|
Huang Y, Chen Z, Chen J, Liu J, Qiu C, Liu Q, Zhang L, Zhu G, Ma X, Sun S, Shi YS, Wan G. Direct reprogramming of fibroblasts into spiral ganglion neurons by defined transcription factors. Cell Prolif 2025; 58:e13775. [PMID: 39551613 PMCID: PMC11969255 DOI: 10.1111/cpr.13775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/19/2024] Open
Abstract
Degeneration of the cochlear spiral ganglion neurons (SGNs) is one of the major causes of sensorineural hearing loss and significantly impacts the outcomes of cochlear implantation. Functional regeneration of SGNs holds great promise for treating sensorineural hearing loss. In this study, we systematically screened 33 transcriptional regulators implicated in neuronal and SGN fate. Using gene expression array and principal component analyses, we identified a sequential combination of Ascl1, Pou4f1 and Myt1l (APM) in promoting functional reprogramming of SGNs. The neurons induced by APM expressed mature neuronal and SGN lineage-specific markers, displayed mature SGN-like electrophysiological characteristics and exhibited single-cell transcriptomes resembling the endogenous SGNs. Thus, transcription factors APM may serve as novel candidates for direct reprogramming of SGNs and hearing recovery due to SGN damages.
Collapse
Affiliation(s)
- Yuhang Huang
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Zhen Chen
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Jiang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Department of Neurology, The Affiliated Drum Tower Hospital of Medical School and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingChina
| | - Jingyue Liu
- National Institute of Biological SciencesBeijingChina
- Tsinghua Institute of Multidisciplinary Biomedical ResearchTsinghua UniversityBeijingChina
| | - Cui Qiu
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Qing Liu
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Linqing Zhang
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Guang‐Jie Zhu
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Xiaofeng Ma
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Shuohao Sun
- National Institute of Biological SciencesBeijingChina
- Tsinghua Institute of Multidisciplinary Biomedical ResearchTsinghua UniversityBeijingChina
| | - Yun Stone Shi
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Guangdong Institute of Intelligence Science and TechnologyZhuhaiChina
| | - Guoqiang Wan
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| |
Collapse
|
2
|
Singh N, Kaushik R, Prakash A, Singh Saini S, Garg S, Adhikary A, Ladher RK. Mosaic Atoh1 deletion in the chick auditory epithelium reveals a homeostatic mechanism to restore hair cell number. Dev Biol 2024; 516:35-46. [PMID: 39074652 DOI: 10.1016/j.ydbio.2024.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/16/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024]
Abstract
The mechanosensory hair cell of the vertebrate inner ear responds to the mechanical deflections that result from hearing or change in the acceleration due to gravity, to allow us to perceive and interpret sounds, maintain balance and spatial orientation. In mammals, ototoxic compounds, disease, and acoustic trauma can result in damage and extrusion of hair cells, without replacement, resulting in hearing loss. In contrast, non-mammalian vertebrates can regenerate sensory hair cells. Upon damage, hair cells are extruded and an associated cell type, the supporting cell is transformed into a hair cell. The mechanisms that can trigger regeneration are not known. Using mosaic deletion of the hair cell master gene, Atoh1, in the embryonic avian inner ear, we find that despite hair cells depletion at E9, by E12, hair cell number is restored in sensory epithelium. Our study suggests a homeostatic mechanism can restores hair cell number in the basilar papilla, that is activated when juxtracrine signalling is disrupted. Restoration of hair cell numbers during development may mirror regenerative processes, and our work provides insights into the mechanisms that trigger regeneration.
Collapse
Affiliation(s)
- Nishant Singh
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, GKVK PO, Bellary Road, Bangalore, 560065, India; The University of Trans-Disciplinary Health Sciences and Technology, Yelahanka, Bangalore, 560064, India
| | - Raman Kaushik
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, GKVK PO, Bellary Road, Bangalore, 560065, India
| | - Anubhav Prakash
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, GKVK PO, Bellary Road, Bangalore, 560065, India; Ashoka University, Sonipat, Haryana, 131029, India
| | - Surjit Singh Saini
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, GKVK PO, Bellary Road, Bangalore, 560065, India
| | - Sonal Garg
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, GKVK PO, Bellary Road, Bangalore, 560065, India
| | - Adrija Adhikary
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, GKVK PO, Bellary Road, Bangalore, 560065, India
| | - Raj K Ladher
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, GKVK PO, Bellary Road, Bangalore, 560065, India.
| |
Collapse
|
3
|
Żak M, Støle TP, Plagnol V, Daudet N. Regulation of otic neurosensory specification by Notch and Wnt signalling: insights from RNA-seq screenings in the embryonic chicken inner ear. Front Cell Dev Biol 2023; 11:1245330. [PMID: 37900277 PMCID: PMC10600479 DOI: 10.3389/fcell.2023.1245330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
The Notch and Wnt signalling pathways play key roles in the formation of inner ear sensory organs, but little is known about their transcriptional effectors and targets in this context. Here, we perturbed Notch and Wnt activities in the embryonic chicken otic vesicle using pharmacological treatment or in ovo electroporation of plasmid DNA, and used RNA-Seq to analyse the resulting changes in gene expression. Compared to pharmacological treatments, in ovo electroporation changed the expression of fewer genes, a likely consequence of the variability and mosaicism of transfection. The pharmacological inhibition of Notch activity induced a rapid change in the expression of known effectors of this pathway and genes associated with neurogenesis, consistent with a switch towards an otic neurosensory fate. The Wnt datasets contained many genes associated with a neurosensory biological function, confirming the importance of this pathway for neurosensory specification in the otocyst. Finally, the results of a preliminary gain-of-function screening of selected transcription factors and Wnt signalling components suggest that the endogenous programs of otic neurosensory specification are very robust, and in general unaffected by the overexpression of a single factor. Altogether this work provides new insights into the effectors and candidate targets of the Notch and Wnt pathways in the early developing inner ear and could serve as a useful reference for future functional genomics experiments in the embryonic avian inner ear.
Collapse
Affiliation(s)
- Magdalena Żak
- UCL Ear Institute, University College London, London, United Kingdom
| | - Thea P. Støle
- UCL Ear Institute, University College London, London, United Kingdom
| | - Vincent Plagnol
- Genetics Institute, University College London, London, United Kingdom
| | - Nicolas Daudet
- UCL Ear Institute, University College London, London, United Kingdom
| |
Collapse
|
4
|
Promotion of In Vitro Hair Cell-like Cell Differentiation from Human Embryonic Stem Cells through the Regulation of Notch Signaling. Metabolites 2021; 11:metabo11120873. [PMID: 34940631 PMCID: PMC8709284 DOI: 10.3390/metabo11120873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 11/25/2022] Open
Abstract
The Notch signaling pathway plays an important role in otic neurogenesis by regulating the differentiation of inner ear hair cells and supporting cells. Notch-regulated differentiation is required for the regeneration of hair cells in the inner ear. The temporal expression pattern of Notch ligands and receptors during in vitro hair cell-like cell differentiation from human embryonic stem cells (hESCs) was detected by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Subsequently, pAJ-U6-shRNA-CMV-Puro/GFP recombinant lentiviral vectors encoding short hairpin RNAs were used to silence JAG-1, JAG-2, and DLL-1, according to the temporal expression pattern of Notch ligands. Then, the effect of each ligand on the in vitro differentiation of hair cells was examined by RT-PCR, immunofluorescence, and scanning electron microscopy (SEM). The results showed that the individual deletion of JAG-2 or DLL-1 had no significant effect on the differentiation of hair cell-like cells. However, the simultaneous inhibition of both DLL-1 and JAG-2 increased the number of hair cell-like cells and decreased the number of supporting cells. JAG-2 and DLL-1 may have a synergistic role in in vitro hair cell differentiation.
Collapse
|
5
|
Riley BB. Comparative assessment of Fgf's diverse roles in inner ear development: A zebrafish perspective. Dev Dyn 2021; 250:1524-1551. [PMID: 33830554 DOI: 10.1002/dvdy.343] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 01/21/2023] Open
Abstract
Progress in understanding mechanisms of inner ear development has been remarkably rapid in recent years. The research community has benefited from the availability of several diverse model organisms, including zebrafish, chick, and mouse. The complexity of the inner ear has proven to be a challenge, and the complexity of the mammalian cochlea in particular has been the subject of intense scrutiny. Zebrafish lack a cochlea and exhibit a number of other differences from amniote species, hence they are sometimes seen as less relevant for inner ear studies. However, accumulating evidence shows that underlying cellular and molecular mechanisms are often highly conserved. As a case in point, consideration of the diverse functions of Fgf and its downstream effectors reveals many similarities between vertebrate species, allowing meaningful comparisons the can benefit the entire research community. In this review, I will discuss mechanisms by which Fgf controls key events in early otic development in zebrafish and provide direct comparisons with chick and mouse.
Collapse
Affiliation(s)
- Bruce B Riley
- Biology Department, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
6
|
Thulabandu V, Nehila T, Ferguson JW, Atit RP. Dermal EZH2 orchestrates dermal differentiation and epidermal proliferation during murine skin development. Dev Biol 2021; 478:25-40. [PMID: 34166654 PMCID: PMC8384472 DOI: 10.1016/j.ydbio.2021.06.008] [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: 03/08/2021] [Revised: 05/28/2021] [Accepted: 06/18/2021] [Indexed: 10/21/2022]
Abstract
Skin development and patterning is dependent on factors that regulate the stepwise differentiation of dermal fibroblasts concomitant with dermal-epidermal reciprocal signaling, two processes that are poorly understood. Here we show that dermal EZH2, the methyltransferase enzyme of the epigenetic Polycomb Repressive Complex 2 (PRC2), is a new coordinator of both these processes. Dermal EZH2 activity is present during dermal fibroblast differentiation and is required for spatially restricting Wnt/β-catenin signaling to reinforce dermal fibroblast cell fate. Later in development, dermal EZH2 regulates the expression of reticular dermal markers and initiation of secondary hair follicles. Embryos lacking dermal Ezh2 have elevated epidermal proliferation and differentiation that can be rescued by small molecule inhibition of retinoic acid (RA) signaling. Together, our study reveals that dermal EZH2 is acting like a rheostat to control the levels of Wnt/β-catenin and RA signaling to impact fibroblast differentiation cell autonomously and epidermal keratinocyte development non-cell autonomously, respectively.
Collapse
Affiliation(s)
| | - Timothy Nehila
- Dept. of Biology, Case Western Reserve University, Cleveland, OH, USA
| | - James W Ferguson
- Dept. of Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Radhika P Atit
- Dept. of Biology, Case Western Reserve University, Cleveland, OH, USA; Dept. of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA; Dept. of Dermatology, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
7
|
Kaiser M, Wojahn I, Rudat C, Lüdtke TH, Christoffels VM, Moon A, Kispert A, Trowe MO. Regulation of otocyst patterning by Tbx2 and Tbx3 is required for inner ear morphogenesis in the mouse. Development 2021; 148:dev.195651. [PMID: 33795231 DOI: 10.1242/dev.195651] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 03/23/2021] [Indexed: 12/21/2022]
Abstract
All epithelial components of the inner ear, including sensory hair cells and innervating afferent neurons, arise by patterning and differentiation of epithelial progenitors residing in a simple sphere, the otocyst. Here, we identify the transcriptional repressors TBX2 and TBX3 as novel regulators of these processes in the mouse. Ablation of Tbx2 from the otocyst led to cochlear hypoplasia, whereas loss of Tbx3 was associated with vestibular malformations. The loss of function of both genes (Tbx2/3cDKO) prevented inner ear morphogenesis at midgestation, resulting in indiscernible cochlear and vestibular structures at birth. Morphogenetic impairment occurred concomitantly with increased apoptosis in ventral and lateral regions of Tbx2/3cDKO otocysts around E10.5. Expression analyses revealed partly disturbed regionalisation, and a posterior-ventral expansion of the neurogenic domain in Tbx2/3cDKO otocysts at this stage. We provide evidence that repression of FGF signalling by TBX2 is important to restrict neurogenesis to the anterior-ventral otocyst and implicate another T-box factor, TBX1, as a crucial mediator in this regulatory network.
Collapse
Affiliation(s)
- Marina Kaiser
- Institute for Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Irina Wojahn
- Institute for Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Carsten Rudat
- Institute for Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Timo H Lüdtke
- Institute for Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Vincent M Christoffels
- Department of Anatomy, Embryology and Physiology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Anne Moon
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Clinic, Danville, PA 17822, USA.,Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Andreas Kispert
- Institute for Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Mark-Oliver Trowe
- Institute for Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| |
Collapse
|
8
|
Hearing Loss Caused by HCMV Infection through Regulating the Wnt and Notch Signaling Pathways. Viruses 2021; 13:v13040623. [PMID: 33917368 PMCID: PMC8067389 DOI: 10.3390/v13040623] [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: 02/16/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 01/27/2023] Open
Abstract
Hearing loss is one of the most prevalent sensory disabilities worldwide with huge social and economic burdens. The leading cause of sensorineural hearing loss (SNHL) in children is congenital cytomegalovirus (CMV) infection. Though the implementation of universal screening and early intervention such as antiviral or anti-inflammatory ameliorate the severity of CMV-associated diseases, direct and targeted therapeutics is still seriously lacking. The major hurdle for it is that the mechanism of CMV induced SNHL has not yet been well understood. In this review, we focus on the impact of CMV infection on the key players in inner ear development including the Wnt and Notch signaling pathways. Investigations on these interactions may gain new insights into viral pathogenesis and reveal novel targets for therapy.
Collapse
|
9
|
Żak M, Daudet N. A gradient of Wnt activity positions the neurosensory domains of the inner ear. eLife 2021; 10:59540. [PMID: 33704062 PMCID: PMC7993990 DOI: 10.7554/elife.59540] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 03/09/2021] [Indexed: 12/25/2022] Open
Abstract
The auditory and vestibular organs of the inner ear and the neurons that innervate them originate from Sox2-positive and Notch-active neurosensory domains specified at early stages of otic development. Sox2 is initially present throughout the otic placode and otocyst, and then it becomes progressively restricted to a ventro-medial domain. Using gain- and loss-of-function approaches in the chicken otocyst, we show that these early changes in Sox2 expression are regulated in a dose-dependent manner by Wnt/beta-catenin signalling. Both high and very low levels of Wnt activity repress Sox2 and neurosensory competence. However, intermediate levels allow the maintenance of Sox2 expression and sensory organ formation. We propose that a dorso-ventral (high-to-low) gradient and wave of Wnt activity initiated at the dorsal rim of the otic placode progressively restricts Sox2 and Notch activity to the ventral half of the otocyst, thereby positioning the neurosensory competent domains in the inner ear.
Collapse
Affiliation(s)
- Magdalena Żak
- UCL Ear Institute, University College London, London, United Kingdom
| | - Nicolas Daudet
- UCL Ear Institute, University College London, London, United Kingdom
| |
Collapse
|
10
|
Matsunaga M, Kita T, Yamamoto R, Yamamoto N, Okano T, Omori K, Sakamoto S, Nakagawa T. Initiation of Supporting Cell Activation for Hair Cell Regeneration in the Avian Auditory Epithelium: An Explant Culture Model. Front Cell Neurosci 2020; 14:583994. [PMID: 33281558 PMCID: PMC7688741 DOI: 10.3389/fncel.2020.583994] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/14/2020] [Indexed: 01/08/2023] Open
Abstract
Sensorineural hearing loss is a common disability often caused by the loss of sensory hair cells in the cochlea. Hair cell (HCs) regeneration has long been the main target for the development of novel therapeutics for sensorineural hearing loss. In the mammalian cochlea, hair cell regeneration is limited, but the auditory epithelia of non-mammalian organisms retain the capacity for hair cell regeneration. In the avian basilar papilla (BP), supporting cells (SCs), which give rise to regenerated hair cells, are usually quiescent. Hair cell loss induces both direct transdifferentiation and mitotic division of supporting cells. Here, we established an explant culture model for hair cell regeneration in chick basilar papillae and validated it for investigating the initial phase of hair cell regeneration. The histological assessment demonstrated hair cell regeneration via direct transdifferentiation of supporting cells. Labeling with 5-ethynyl-2′-deoxyuridine (EdU) revealed the occurrence of mitotic division in the supporting cells at specific locations in the basilar papillae, while no EdU labeling was observed in newly generated hair cells. RNA sequencing indicated alterations in known signaling pathways associated with hair cell regeneration, consistent with previous findings. Also, unbiased analyses of RNA sequencing data revealed novel genes and signaling pathways that may be related to the induction of supporting cell activation in the chick basilar papillae. These results indicate the advantages of our explant culture model of the chick basilar papillae for exploring the molecular mechanisms of hair cell regeneration.
Collapse
Affiliation(s)
- Mami Matsunaga
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoko Kita
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryosuke Yamamoto
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norio Yamamoto
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takayuki Okano
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koichi Omori
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Takayuki Nakagawa
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
11
|
Bocci F, Onuchic JN, Jolly MK. Understanding the Principles of Pattern Formation Driven by Notch Signaling by Integrating Experiments and Theoretical Models. Front Physiol 2020; 11:929. [PMID: 32848867 PMCID: PMC7411240 DOI: 10.3389/fphys.2020.00929] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
Notch signaling is an evolutionary conserved cell-cell communication pathway. Besides regulating cell-fate decisions at an individual cell level, Notch signaling coordinates the emergent spatiotemporal patterning in a tissue through ligand-receptor interactions among transmembrane molecules of neighboring cells, as seen in embryonic development, angiogenesis, or wound healing. Due to its ubiquitous nature, Notch signaling is also implicated in several aspects of cancer progression, including tumor angiogenesis, stemness of cancer cells and cellular invasion. Here, we review experimental and computational models that help understand the operating principles of cell patterning driven by Notch signaling. First, we discuss the basic mechanisms of spatial patterning via canonical lateral inhibition and lateral induction mechanisms, including examples from angiogenesis, inner ear development and cancer metastasis. Next, we analyze additional layers of complexity in the Notch pathway, including the effect of varying cell sizes and shapes, ligand-receptor binding within the same cell, variable binding affinity of different ligand/receptor subtypes, and filopodia. Finally, we discuss some recent evidence of mechanosensitivity in the Notch pathway in driving collective epithelial cell migration and cardiovascular morphogenesis.
Collapse
Affiliation(s)
- Federico Bocci
- Center for Theoretical Biological Physics, Rice University, Houston, TX, United States
| | - José Nelson Onuchic
- Center for Theoretical Biological Physics, Rice University, Houston, TX, United States
- Department of Physics and Astronomy, Rice University, Houston, TX, United States
- Department of Chemistry, Rice University, Houston, TX, United States
- Department of Biosciences, Rice University, Houston, TX, United States
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
12
|
Nichols DH, Bouma JE, Kopecky BJ, Jahan I, Beisel KW, He DZZ, Liu H, Fritzsch B. Interaction with ectopic cochlear crista sensory epithelium disrupts basal cochlear sensory epithelium development in Lmx1a mutant mice. Cell Tissue Res 2020; 380:435-448. [PMID: 31932950 PMCID: PMC7393901 DOI: 10.1007/s00441-019-03163-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/18/2019] [Indexed: 12/14/2022]
Abstract
The LIM homeodomain transcription factor Lmx1a shows a dynamic expression in the developing mouse ear that stabilizes in the non-sensory epithelium. Previous work showed that Lmx1a functional null mutants have an additional sensory hair cell patch in the posterior wall of a cochlear duct and have a mix of vestibular and cochlear hair cells in the basal cochlear sensory epithelium. In E13.5 mutants, Sox2-expressing posterior canal crista is continuous with an ectopic "crista sensory epithelium" located in the outer spiral sulcus of the basal cochlear duct. The medial margin of cochlear crista is in contact with the adjacent Sox2-expressing basal cochlear sensory epithelium. By E17.5, this contact has been interrupted by the formation of an intervening non-sensory epithelium, and Atoh1 is expressed in the hair cells of both the cochlear crista and the basal cochlear sensory epithelium. Where cochlear crista was formerly associated with the basal cochlear sensory epithelium, the basal cochlear sensory epithelium lacks an outer hair cell band, and gaps are present in its associated Bmp4 expression. Further apically, where cochlear crista was never present, the cochlear sensory epithelium forms a poorly ordered but complete organ of Corti. We propose that the core prosensory posterior crista is enlarged in the mutant when the absence of Lmx1a expression allows JAG1-NOTCH signaling to propagate into the adjacent epithelium and down the posterior wall of the cochlear duct. We suggest that the cochlear crista propagates in the mutant outer spiral sulcus because it expresses Lmo4 in the absence of Lmx1a.
Collapse
Affiliation(s)
- David H Nichols
- Department of Biomedical Sciences, Creighton University, Omaha, NE, USA
| | - Judith E Bouma
- Department of Biomedical Sciences, Creighton University, Omaha, NE, USA
| | - Benjamin J Kopecky
- Department of Biology, University of Iowa, Iowa City, IA, 52242-1324, USA
| | - Israt Jahan
- Department of Biology, University of Iowa, Iowa City, IA, 52242-1324, USA
| | - Kirk W Beisel
- Department of Biomedical Sciences, Creighton University, Omaha, NE, USA
| | - David Z Z He
- Department of Biomedical Sciences, Creighton University, Omaha, NE, USA
| | - Huizhan Liu
- Department of Biomedical Sciences, Creighton University, Omaha, NE, USA
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA, 52242-1324, USA.
| |
Collapse
|
13
|
Notch-mediated lateral induction is necessary to maintain vestibular prosensory identity during inner ear development. Dev Biol 2020; 462:74-84. [PMID: 32147304 DOI: 10.1016/j.ydbio.2020.02.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 01/24/2023]
Abstract
The five vestibular organs of the inner ear derive from patches of prosensory cells that express the transcription factor SOX2 and the Notch ligand JAG1. Previous work suggests that JAG1-mediated Notch signaling is both necessary and sufficient for prosensory formation and that the separation of developing prosensory patches is regulated by LMX1a, which antagonizes Notch signaling. We used an inner ear-specific deletion of the Rbpjκ gene in which Notch signaling is progressively lost from the inner ear to show that Notch signaling, is continuously required for the maintenance of prosensory fate. Loss of Notch signaling in prosensory patches causes them to shrink and ultimately disappear. We show this loss of prosensory fate is not due to cell death, but rather to the conversion of prosensory tissue into non-sensory tissue that expresses LMX1a. Notch signaling is therefore likely to stabilize, rather than induce prosensory fate.
Collapse
|
14
|
Brown R, Groves AK. Hear, Hear for Notch: Control of Cell Fates in the Inner Ear by Notch Signaling. Biomolecules 2020; 10:biom10030370. [PMID: 32121147 PMCID: PMC7175228 DOI: 10.3390/biom10030370] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 02/08/2023] Open
Abstract
The vertebrate inner ear is responsible for detecting sound, gravity, and head motion. These mechanical forces are detected by mechanosensitive hair cells, arranged in a series of sensory patches in the vestibular and cochlear regions of the ear. Hair cells form synapses with neurons of the VIIIth cranial ganglion, which convey sound and balance information to the brain. They are surrounded by supporting cells, which nourish and protect the hair cells, and which can serve as a source of stem cells to regenerate hair cells after damage in non-mammalian vertebrates. The Notch signaling pathway plays many roles in the development of the inner ear, from the earliest formation of future inner ear ectoderm on the side of the embryonic head, to regulating the production of supporting cells, hair cells, and the neurons that innervate them. Notch signaling is re-deployed in non-mammalian vertebrates during hair cell regeneration, and attempts have been made to manipulate the Notch pathway to promote hair cell regeneration in mammals. In this review, we summarize the different modes of Notch signaling in inner ear development and regeneration, and describe how they interact with other signaling pathways to orchestrate the fine-grained cellular patterns of the ear.
Collapse
Affiliation(s)
- Rogers Brown
- Program in Developmental Biology; Baylor College of Medicine, Houston, TX 77030, USA;
| | - Andrew K. Groves
- Program in Developmental Biology; Baylor College of Medicine, Houston, TX 77030, USA;
- Department of Neuroscience; Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-713-798-8743
| |
Collapse
|
15
|
Notch Signalling: The Multitask Manager of Inner Ear Development and Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1218:129-157. [DOI: 10.1007/978-3-030-34436-8_8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
16
|
Han X, Wei Y, Wu X, Gao J, Yang Z, Zhao C. PDK1 Regulates Transition Period of Apical Progenitors to Basal Progenitors by Controlling Asymmetric Cell Division. Cereb Cortex 2019; 30:406-420. [DOI: 10.1093/cercor/bhz146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/09/2019] [Accepted: 06/09/2019] [Indexed: 12/18/2022] Open
Abstract
Abstract
The six-layered neocortex consists of diverse neuron subtypes. Deeper-layer neurons originate from apical progenitors (APs), while upper-layer neurons are mainly produced by basal progenitors (BPs), which are derivatives of APs. As development proceeds, an AP generates two daughter cells that comprise an AP and a deeper-layer neuron or a BP. How the transition of APs to BPs is spatiotemporally regulated is a fundamental question. Here, we report that conditional deletion of phoshpoinositide-dependent protein kinase 1 (PDK1) in mouse developing cortex achieved by crossing Emx1Cre line with Pdk1fl/fl leads to a delayed transition of APs to BPs and subsequently causes an increased output of deeper-layer neurons. We demonstrate that PDK1 is involved in the modulation of the aPKC-Par3 complex and further regulates the asymmetric cell division (ACD). We also find Hes1, a downstream effecter of Notch signal pathway is obviously upregulated. Knockdown of Hes1 or treatment with Notch signal inhibitor DAPT recovers the ACD defect in the Pdk1 cKO. Thus, we have identified a novel function of PDK1 in controlling the transition of APs to BPs.
Collapse
Affiliation(s)
- Xiaoning Han
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yongjie Wei
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiaojing Wu
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jun Gao
- Department of Neurobiology
- Key Laboratory of Human Functional Genomics of Jiangsu, Nanjing Medical University, Nanjing 211166, China
| | - Zhongzhou Yang
- State Key Laboratory of Pharmaceutical Biotechnology
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210061, China
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
17
|
Hwang CH, Keller J, Renner C, Ohta S, Wu DK. Genetic interactions support an inhibitory relationship between bone morphogenetic protein 2 and netrin 1 during semicircular canal formation. Development 2019; 146:dev.174748. [PMID: 30770380 PMCID: PMC6398446 DOI: 10.1242/dev.174748] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 01/18/2019] [Indexed: 12/16/2022]
Abstract
The semicircular canals of the mammalian inner ear are derived from epithelial pouches in which epithelial cells in the central region of each pouch undergo resorption, leaving behind the region at the rim to form a tube-shaped canal. Lack of proliferation at the rim and/or over-clearing of epithelial cells in the center of the pouch can obliterate canal formation. Otic-specific knockout of bone morphogenetic protein 2 (Bmp2) results in absence of all three semicircular canals; however, the common crus and ampullae housing the sensory tissue (crista) are intact. The lack of Bmp2 causes Ntn1 (which encodes netrin 1), which is required for canal resorption, to be ectopically expressed at the canal rim. Ectopic Ntn1 results in reduction of Dlx5 and Lmo4, which are required for rim formation. These phenotypes can be partially rescued by removing one allele of Ntn1 in the Bmp2 mutants, indicating that Bmp2 normally negatively regulates Ntn1 for canal formation. Additionally, non-resorption of the canal pouch in Ntn1−/− mutants is partially rescued by removing one allele of Bmp2. Thus, reciprocal inhibition between Bmp2 and netrin 1 is involved in canal formation of the vestibule. Summary:Bmp2-conditional mutant analyses support the hypothesis that presumptive crista induces canal genesis zone in the canal pouch to express Bmp2, which promotes canal formation by restricting Ntn1 expression to the resorption domain.
Collapse
Affiliation(s)
- Chan Ho Hwang
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, MD 20892, USA
| | - James Keller
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, MD 20892, USA
| | - Charles Renner
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, MD 20892, USA
| | - Sho Ohta
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, MD 20892, USA
| | - Doris K Wu
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Lahlou H, Lopez-Juarez A, Fontbonne A, Nivet E, Zine A. Modeling human early otic sensory cell development with induced pluripotent stem cells. PLoS One 2018; 13:e0198954. [PMID: 29902227 PMCID: PMC6002076 DOI: 10.1371/journal.pone.0198954] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 05/24/2018] [Indexed: 11/18/2022] Open
Abstract
The inner ear represents a promising system to develop cell-based therapies from human induced pluripotent stem cells (hiPSCs). In the developing ear, Notch signaling plays multiple roles in otic region specification and for cell fate determination. Optimizing hiPSC induction for the generation of appropriate numbers of otic progenitors and derivatives, such as hair cells, may provide an unlimited supply of cells for research and cell-based therapy. In this study, we used monolayer cultures, otic-inducing agents, Notch modulation, and marker expression to track early and otic sensory lineages during hiPSC differentiation. Otic/placodal progenitors were derived from hiPSC cultures in medium supplemented with FGF3/FGF10 for 13 days. These progenitor cells were then treated for 7 days with retinoic acid (RA) and epidermal growth factor (EGF) or a Notch inhibitor. The differentiated cultures were analyzed in parallel by qPCR and immunocytochemistry. After the 13 day induction, hiPSC-derived cells displayed an upregulated expression of a panel of otic/placodal markers. Strikingly, a subset of these induced progenitor cells displayed key-otic sensory markers, the percentage of which was increased in cultures under Notch inhibition as compared to RA/EGF-treated cultures. Our results show that modulating Notch pathway during in vitro differentiation of hiPSC-derived otic/placodal progenitors is a valuable strategy to promote the expression of human otic sensory lineage genes.
Collapse
Affiliation(s)
- Hanae Lahlou
- Aix Marseille Université, CNRS, LNIA UMR 7260, Marseille, France
| | | | - Arnaud Fontbonne
- Aix Marseille Université, CNRS, LNIA UMR 7260, Marseille, France
| | - Emmanuel Nivet
- Aix Marseille Université, CNRS, NICN UMR 7259, Marseille, France
| | - Azel Zine
- Aix Marseille Université, CNRS, LNIA UMR 7260, Marseille, France
- Université de Montpellier, Faculté de Pharmacie, Montpellier, France
- * E-mail: ,
| |
Collapse
|
19
|
Weston MD, Tarang S, Pierce ML, Pyakurel U, Rocha-Sanchez SM, McGee J, Walsh EJ, Soukup GA. A mouse model of miR-96, miR-182 and miR-183 misexpression implicates miRNAs in cochlear cell fate and homeostasis. Sci Rep 2018; 8:3569. [PMID: 29476110 PMCID: PMC5824881 DOI: 10.1038/s41598-018-21811-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 02/12/2018] [Indexed: 11/21/2022] Open
Abstract
Germline mutations in Mir96, one of three co-expressed polycistronic miRNA genes (Mir96, Mir182, Mir183), cause hereditary hearing loss in humans and mice. Transgenic FVB/NCrl- Tg(GFAP-Mir183,Mir96,Mir182)MDW1 mice (Tg1MDW), which overexpress this neurosensory-specific miRNA cluster in the inner ear, were developed as a model system to identify, in the aggregate, target genes and biologic processes regulated by the miR-183 cluster. Histological assessments demonstrate Tg1MDW/1MDW homozygotes have a modest increase in cochlear inner hair cells (IHCs). Affymetrix mRNA microarray data analysis revealed that downregulated genes in P5 Tg1MDW/1MDW cochlea are statistically enriched for evolutionarily conserved predicted miR-96, miR-182 or miR-183 target sites. ABR and DPOAE tests from 18 days to 3 months of age revealed that Tg1MDW/1MDW homozygotes develop progressive neurosensory hearing loss that correlates with histologic assessments showing massive losses of both IHCs and outer hair cells (OHCs). This mammalian miRNA misexpression model demonstrates a potency and specificity of cochlear homeostasis for one of the dozens of endogenously co-expressed, evolutionally conserved, small non-protein coding miRNA families. It should be a valuable tool to predict and elucidate miRNA-regulated genes and integrated functional gene expression networks that significantly influence neurosensory cell differentiation, maturation and homeostasis.
Collapse
MESH Headings
- Animals
- Cell Differentiation/genetics
- Disease Models, Animal
- Ear, Inner/metabolism
- Ear, Inner/pathology
- Gene Expression Regulation
- Hair Cells, Auditory, Inner/metabolism
- Hair Cells, Auditory, Inner/pathology
- Hearing Loss, Sensorineural/genetics
- Hearing Loss, Sensorineural/pathology
- Homeostasis/genetics
- Humans
- Mice
- Mice, Transgenic
- MicroRNAs/genetics
- Microarray Analysis
- RNA, Messenger/genetics
Collapse
Affiliation(s)
- Michael D Weston
- Department of Oral Biology, School of Dentistry, Creighton University, 780729 California Plaza, Omaha, NE 68178-0729, USA.
| | - Shikha Tarang
- Department of Oral Biology, School of Dentistry, Creighton University, 780729 California Plaza, Omaha, NE 68178-0729, USA
| | - Marsha L Pierce
- Department of Pharmacology, School of Medicine, Creighton University, 2500 California Plaza, Omaha, NE 68178, USA
| | - Umesh Pyakurel
- Department of Oral Biology, School of Dentistry, Creighton University, 780729 California Plaza, Omaha, NE 68178-0729, USA
| | - Sonia M Rocha-Sanchez
- Department of Oral Biology, School of Dentistry, Creighton University, 780729 California Plaza, Omaha, NE 68178-0729, USA
| | - JoAnn McGee
- Developmental Auditory Physiology Laboratory, Boys Town National Research Hospital, 555 North 30th Street, Omaha, NE 68131, USA
| | - Edward J Walsh
- Developmental Auditory Physiology Laboratory, Boys Town National Research Hospital, 555 North 30th Street, Omaha, NE 68131, USA
| | - Garrett A Soukup
- Department of Biomedical Sciences, School of Medicine, Creighton University, 2500 California Plaza, Omaha, NE 68178, USA
| |
Collapse
|
20
|
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
|
21
|
Prasasya RD, Mayo KE. Notch Signaling Regulates Differentiation and Steroidogenesis in Female Mouse Ovarian Granulosa Cells. Endocrinology 2018; 159:184-198. [PMID: 29126263 PMCID: PMC5761600 DOI: 10.1210/en.2017-00677] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/02/2017] [Indexed: 01/04/2023]
Abstract
The Notch pathway is a highly conserved juxtacrine signaling mechanism that is important for many cellular processes during development, including differentiation and proliferation. Although Notch is important during ovarian follicle formation and early development, its functions during the gonadotropin-dependent stages of follicle development are largely unexplored. We observed positive regulation of Notch activity and expression of Notch ligands and receptors following activation of the luteinizing hormone-receptor in prepubertal mouse ovary. JAG1, the most abundantly expressed Notch ligand in mouse ovary, revealed a striking shift in localization from oocytes to somatic cells following hormone stimulation. Using primary cultures of granulosa cells, we investigated the functions of Jag1 using small interfering RNA knockdown. The loss of JAG1 led to suppression of granulosa cell differentiation as marked by reduced expression of enzymes and factors involved in steroid biosynthesis, and in steroid secretion. Jag1 knockdown also resulted in enhanced cell proliferation. These phenotypes were replicated, although less robustly, following knockdown of the obligate canonical Notch transcription factor RBPJ. Intracellular signaling analysis revealed increased activation of the mitogenic phosphatidylinositol 3-kinase/protein kinase B and mitogen-activated protein kinase/extracellular signal-regulated kinase pathways following Notch knockdown, with a mitogen-activated protein kinase kinase inhibitor blocking the enhanced proliferation observed in Jag1 knockdown granulosa cells. Activation of YB-1, a known regulator of granulosa cell differentiation genes, was suppressed by Jag1 knockdown. Overall, this study reveals a role of Notch signaling in promoting the differentiation of preovulatory granulosa cells, adding to the diverse functions of Notch in the mammalian ovary.
Collapse
MESH Headings
- Animals
- Cell Differentiation/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Chorionic Gonadotropin/pharmacology
- Estradiol/metabolism
- Female
- Gene Expression Regulation, Developmental/drug effects
- Genes, Reporter/drug effects
- Gonadotropins, Equine/pharmacology
- Granulosa Cells/cytology
- Granulosa Cells/drug effects
- Granulosa Cells/metabolism
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/antagonists & inhibitors
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism
- Jagged-1 Protein/antagonists & inhibitors
- Jagged-1 Protein/genetics
- Jagged-1 Protein/metabolism
- MAP Kinase Signaling System/drug effects
- Mice, Inbred Strains
- Mice, Transgenic
- Progesterone/metabolism
- RNA Interference
- Receptor, Notch2/agonists
- Receptor, Notch2/genetics
- Receptor, Notch2/metabolism
- Receptor, Notch3/agonists
- Receptor, Notch3/genetics
- Receptor, Notch3/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
Collapse
Affiliation(s)
- Rexxi D. Prasasya
- Department of Molecular Biosciences and Center for Reproductive Science, Northwestern University, Evanston, Illinois 60208
| | - Kelly E. Mayo
- Department of Molecular Biosciences and Center for Reproductive Science, Northwestern University, Evanston, Illinois 60208
| |
Collapse
|
22
|
Williams E, Villar-Prados A, Bowser J, Broaddus R, Gladden AB. Loss of polarity alters proliferation and differentiation in low-grade endometrial cancers by disrupting Notch signaling. PLoS One 2017; 12:e0189081. [PMID: 29206870 PMCID: PMC5716545 DOI: 10.1371/journal.pone.0189081] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 11/18/2017] [Indexed: 01/03/2023] Open
Abstract
Cell adhesion and apicobasal polarity together maintain epithelial tissue organization and homeostasis. Loss of adhesion has been described as a prerequisite for the epithelial to mesenchymal transition. However, what role misregulation of apicobasal polarity promotes tumor initiation and/or early progression remains unclear. We find that human low-grade endometrial cancers are associated with disrupted localization of the apical polarity protein Par3 and Ezrin while, the adhesion molecule E-cadherin remains unchanged, accompanied by decreased Notch signaling, and altered Notch receptor localization. Depletion of Par3 or Ezrin, in a cell-based model, results in loss of epithelial architecture, differentiation, increased proliferation, migration and decreased Notch signaling. Re-expression of Par3 in endometrial cancer cell lines with disrupted Par3 protein levels blocks proliferation and reduces migration in a Notch dependent manner. These data uncover a function for apicobasal polarity independent of cell adhesion in regulating Notch-mediated differentiation signals in endometrial epithelial cells.
Collapse
Affiliation(s)
- Erin Williams
- Department of Genetics, University of Texas, MD Anderson Cancer Center, Houston, TX, United States of America
- Program of Genes and Development, Graduate School of Biomedical Sciences, University of Texas Health Sciences Center, Houston, TX, United States of America
| | - Alejandro Villar-Prados
- Department of Genetics, University of Texas, MD Anderson Cancer Center, Houston, TX, United States of America
- Program of Genes and Development, Graduate School of Biomedical Sciences, University of Texas Health Sciences Center, Houston, TX, United States of America
| | - Jessica Bowser
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, United States of America
| | - Russell Broaddus
- Program of Genes and Development, Graduate School of Biomedical Sciences, University of Texas Health Sciences Center, Houston, TX, United States of America
- Department of Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, United States of America
| | - Andrew B. Gladden
- Department of Genetics, University of Texas, MD Anderson Cancer Center, Houston, TX, United States of America
- Program of Genes and Development, Graduate School of Biomedical Sciences, University of Texas Health Sciences Center, Houston, TX, United States of America
| |
Collapse
|
23
|
Chou CW, Lin J, Jiang YJ, Liu YW. Aberrant Global and Jagged-Mediated Notch Signaling Disrupts Segregation Between wt1-Expressing and Steroidogenic Tissues in Zebrafish. Endocrinology 2017; 158:4206-4217. [PMID: 29029162 DOI: 10.1210/en.2017-00548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 09/26/2017] [Indexed: 11/19/2022]
Abstract
Although the zebrafish interrenal tissue has been used as a model for steroidogenesis and genesis of the adrenal gland, its specification and morphogenesis remains largely unclear. In the present study, we explored how the Wilms tumor 1 (WT1)-expressing cells are segregated from the SF-1-expressing steroidogenic cells in the zebrafish model. The interrenal tissue precursors expressing ff1b, the equivalent of mammalian SF-1, were derived from wt1-expressing pronephric primordia in the zebrafish embryo. Through histochemistry and in situ hybridization, we demonstrated that the size of functionally differentiated interrenal tissue was substantially increased on global inhibition of the Notch signaling pathway and was accompanied by a disrupted segregation between the wt1- and ff1b-expressing cells. As the Notch pathway was conditionally activated during interrenal specification, differentiation, but not ff1b expression, of interrenal tissue was drastically compromised. In embryos deficient for Notch ligands jagged 1b and 2b, transgenic reporter activity of wt1b promoter was detected within the steroidogenic interrenal tissue. In conclusion, our results indicate that Jagged-Notch signaling is required (1) for segregation between wt1-expressing cells and differentiated steroidogenic tissue; and (2) to modulate the extent of functional differentiation in the steroidogenic interrenal tissue.
Collapse
Affiliation(s)
- Chih-Wei Chou
- Department of Life Science, Tunghai University, Taiwan
| | - Jamie Lin
- Department of Life Science, Tunghai University, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Taiwan
| | - Yun-Jin Jiang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Taiwan
| | - Yi-Wen Liu
- Department of Life Science, Tunghai University, Taiwan
| |
Collapse
|
24
|
Macchiarulo S, Morrow BE. Tbx1 and Jag1 act in concert to modulate the fate of neurosensory cells of the mouse otic vesicle. Biol Open 2017; 6:1472-1482. [PMID: 28838968 PMCID: PMC5665468 DOI: 10.1242/bio.027359] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The domain within the otic vesicle (OV) known as the neurosensory domain (NSD), contains cells that will give rise to the hair and support cells of the otic sensory organs, as well as the neurons that form the cochleovestibular ganglion (CVG). The molecular dynamics that occur at the NSD boundary relative to adjacent OV cells is not well defined. The Tbx1 transcription factor gene expression pattern is complementary to the NSD, and inactivation results in expansion of the NSD and expression of the Notch ligand, Jag1 mapping, in part of the NSD. To shed light on the role of Jag1 in NSD development, as well as to test whether Tbx1 and Jag1 might genetically interact to regulate this process, we inactivated Jag1 within the Tbx1 expression domain using a knock-in Tbx1Cre allele. We observed an enlarged neurogenic domain marked by a synergistic increase in expression of NeuroD and other proneural transcription factor genes in double Tbx1 and Jag1 conditional loss-of-function embryos. We noted that neuroblasts preferentially expanded across the medial-lateral axis and that an increase in cell proliferation could not account for this expansion, suggesting that there was a change in cell fate. We also found that inactivation of Jag1 with Tbx1Cre resulted in failed development of the cristae and semicircular canals, as well as notably fewer hair cells in the ventral epithelium of the inner ear rudiment when inactivated on a Tbx1 null background, compared to Tbx1Cre/− mutant embryos. We propose that loss of expression of Tbx1 and Jag1 within the Tbx1 expression domain tips the balance of cell fates in the NSD, resulting in an overproduction of neuroblasts at the expense of non-neural cells within the OV. Summary: Normal dosages of Tbx1 and Jag1 are required to maintain a proper balance of cell types within the neurosensory domain of the otic vesicle to form the inner ear.
Collapse
Affiliation(s)
- Stephania Macchiarulo
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461, USA
| | - Bernice E Morrow
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461, USA .,Departments of Obstetrics and Gynecology and Pediatrics, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461, USA
| |
Collapse
|
25
|
Siebel C, Lendahl U. Notch Signaling in Development, Tissue Homeostasis, and Disease. Physiol Rev 2017; 97:1235-1294. [PMID: 28794168 DOI: 10.1152/physrev.00005.2017] [Citation(s) in RCA: 686] [Impact Index Per Article: 85.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/19/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023] Open
Abstract
Notch signaling is an evolutionarily highly conserved signaling mechanism, but in contrast to signaling pathways such as Wnt, Sonic Hedgehog, and BMP/TGF-β, Notch signaling occurs via cell-cell communication, where transmembrane ligands on one cell activate transmembrane receptors on a juxtaposed cell. Originally discovered through mutations in Drosophila more than 100 yr ago, and with the first Notch gene cloned more than 30 yr ago, we are still gaining new insights into the broad effects of Notch signaling in organisms across the metazoan spectrum and its requirement for normal development of most organs in the body. In this review, we provide an overview of the Notch signaling mechanism at the molecular level and discuss how the pathway, which is architecturally quite simple, is able to engage in the control of cell fates in a broad variety of cell types. We discuss the current understanding of how Notch signaling can become derailed, either by direct mutations or by aberrant regulation, and the expanding spectrum of diseases and cancers that is a consequence of Notch dysregulation. Finally, we explore the emerging field of Notch in the control of tissue homeostasis, with examples from skin, liver, lung, intestine, and the vasculature.
Collapse
Affiliation(s)
- Chris Siebel
- Department of Discovery Oncology, Genentech Inc., DNA Way, South San Francisco, California; and Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Urban Lendahl
- Department of Discovery Oncology, Genentech Inc., DNA Way, South San Francisco, California; and Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
26
|
Fritzsch B, Elliott KL. Gene, cell, and organ multiplication drives inner ear evolution. Dev Biol 2017; 431:3-15. [PMID: 28866362 DOI: 10.1016/j.ydbio.2017.08.034] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 04/27/2017] [Accepted: 08/25/2017] [Indexed: 12/14/2022]
Abstract
We review the development and evolution of the ear neurosensory cells, the aggregation of neurosensory cells into an otic placode, the evolution of novel neurosensory structures dedicated to hearing and the evolution of novel nuclei in the brain and their input dedicated to processing those novel auditory stimuli. The evolution of the apparently novel auditory system lies in duplication and diversification of cell fate transcription regulation that allows variation at the cellular level [transforming a single neurosensory cell into a sensory cell connected to its targets by a sensory neuron as well as diversifying hair cells], organ level [duplication of organ development followed by diversification and novel stimulus acquisition] and brain nuclear level [multiplication of transcription factors to regulate various neuron and neuron aggregate fate to transform the spinal cord into the unique hindbrain organization]. Tying cell fate changes driven by bHLH and other transcription factors into cell and organ changes is at the moment tentative as not all relevant factors are known and their gene regulatory network is only rudimentary understood. Future research can use the blueprint proposed here to provide both the deeper molecular evolutionary understanding as well as a more detailed appreciation of developmental networks. This understanding can reveal how an auditory system evolved through transformation of existing cell fate determining networks and thus how neurosensory evolution occurred through molecular changes affecting cell fate decision processes. Appreciating the evolutionary cascade of developmental program changes could allow identifying essential steps needed to restore cells and organs in the future.
Collapse
Affiliation(s)
- Bernd Fritzsch
- University of Iowa, Department of Biology, Iowa City, IA 52242, United States.
| | - Karen L Elliott
- University of Iowa, Department of Biology, Iowa City, IA 52242, United States
| |
Collapse
|
27
|
Abstract
Neurons of the cochleovestibular ganglion (CVG) transmit hearing and balance information to the brain. During development, a select population of early otic progenitors express NEUROG1, delaminate from the otocyst, and coalesce to form the neurons that innervate all inner ear sensory regions. At present, the selection process that determines which otic progenitors activate NEUROG1 and adopt a neuroblast fate is incompletely understood. The transcription factor SOX2 has been implicated in otic neurogenesis, but its requirement in the specification of the CVG neurons has not been established. Here we tested SOX2's requirement during inner ear neuronal specification using a conditional deletion paradigm in the mouse. SOX2 deficiency at otocyst stages caused a near-absence of NEUROG1-expressing neuroblasts, increased cell death in the neurosensory epithelium, and significantly reduced the CVG volume. Interestingly, a milder decrease in neurogenesis was observed in heterozygotes, indicating SOX2 levels are important. Moreover, fate-mapping experiments revealed that the timing of SOX2 expression did not parallel the established vestibular-then-auditory sequence. These results demonstrate that SOX2 is required for the initial events in otic neuronal specification including expression of NEUROG1, although fate-mapping results suggest SOX2 may be required as a competence factor rather than a direct initiator of the neural fate.
Collapse
Affiliation(s)
- Aleta R Steevens
- Flaum Eye Institute, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Jenna C Glatzer
- Flaum Eye Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Amy E Kiernan
- Flaum Eye Institute, University of Rochester Medical Center, Rochester, NY, USA. .,Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
28
|
Role of Wnt and Notch signaling in regulating hair cell regeneration in the cochlea. Front Med 2016; 10:237-49. [PMID: 27527363 DOI: 10.1007/s11684-016-0464-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/12/2016] [Indexed: 01/22/2023]
Abstract
Sensory hair cells in the inner ear are responsible for sound recognition. Damage to hair cells in adult mammals causes permanent hearing impairment because these cells cannot regenerate. By contrast, newborn mammals possess limited regenerative capacity because of the active participation of various signaling pathways, including Wnt and Notch signaling. The Wnt and Notch pathways are highly sophisticated and conserved signaling pathways that control multiple cellular events necessary for the formation of sensory hair cells. Both signaling pathways allow resident supporting cells to regenerate hair cells in the neonatal cochlea. In this regard, Wnt and Notch signaling has gained increased research attention in hair cell regeneration. This review presents the current understanding of the Wnt and Notch signaling pathways in the auditory portion of the inner ear and discusses the possibilities of controlling these pathways with the hair cell fate determiner Atoh1 to regulate hair cell regeneration in the mammalian cochlea.
Collapse
|
29
|
Abstract
The highly conserved Notch signalling pathway functions in many different developmental and homeostatic processes, which raises the question of how this pathway can achieve such diverse outcomes. With a direct route from the membrane to the nucleus, the Notch pathway has fewer opportunities for regulation than do many other signalling pathways, yet it generates exquisitely patterned structures, including sensory hair cells and branched arterial networks. More confusingly, its activity promotes tissue growth and cancers in some circumstances but cell death and tumour suppression in others. Many different regulatory mechanisms help to shape the activity of the Notch pathway, generating functional outputs that are appropriate for each context. These mechanisms include the receptor-ligand landscape, the tissue topology, the nuclear environment and the connectivity of the regulatory networks.
Collapse
Affiliation(s)
- Sarah J Bray
- Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| |
Collapse
|
30
|
Lineage tracing of Sox2-expressing progenitor cells in the mouse inner ear reveals a broad contribution to non-sensory tissues and insights into the origin of the organ of Corti. Dev Biol 2016; 414:72-84. [PMID: 27090805 DOI: 10.1016/j.ydbio.2016.03.027] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 03/09/2016] [Accepted: 03/26/2016] [Indexed: 11/22/2022]
Abstract
The transcription factor Sox2 is both necessary and sufficient for the generation of sensory regions of the inner ear. It regulates expression of the Notch ligand Jag1 in prosensory progenitors, which signal to neighboring cells to up-regulate Sox2 and sustain prosensory identity. However, the expression pattern of Sox2 in the early inner ear is very broad, suggesting that Sox2-expressing progenitors form a wide variety of cell types in addition to generating the sensory regions of the ear. We used Sox2-CreER mice to follow the fates of Sox2-expressing cells at different stages in ear development. We find that Sox2-expressing cells in the early otocyst give rise to large numbers of non-sensory structures throughout the inner ear, and that Sox2 only becomes a truly prosensory marker at embryonic day (E)11.5. Our fate map reveals the organ of Corti derives from a central domain on the medial side of the otocyst and shows that a significant amount of the organ of Corti derives from a Sox2-negative population in this region.
Collapse
|
31
|
Basch ML, Brown RM, Jen H, Groves AK. Where hearing starts: the development of the mammalian cochlea. J Anat 2016; 228:233-54. [PMID: 26052920 PMCID: PMC4718162 DOI: 10.1111/joa.12314] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2015] [Indexed: 12/11/2022] Open
Abstract
The mammalian cochlea is a remarkable sensory organ, capable of perceiving sound over a range of 10(12) in pressure, and discriminating both infrasonic and ultrasonic frequencies in different species. The sensory hair cells of the mammalian cochlea are exquisitely sensitive, responding to atomic-level deflections at speeds on the order of tens of microseconds. The number and placement of hair cells are precisely determined during inner ear development, and a large number of developmental processes sculpt the shape, size and morphology of these cells along the length of the cochlear duct to make them optimally responsive to different sound frequencies. In this review, we briefly discuss the evolutionary origins of the mammalian cochlea, and then describe the successive developmental processes that lead to its induction, cell cycle exit, cellular patterning and the establishment of topologically distinct frequency responses along its length.
Collapse
Affiliation(s)
- Martin L. Basch
- Department of NeuroscienceBaylor College of MedicineHoustonTXUSA
| | - Rogers M. Brown
- Program in Developmental BiologyBaylor College of MedicineHoustonTXUSA
| | - Hsin‐I Jen
- Program in Developmental BiologyBaylor College of MedicineHoustonTXUSA
| | - Andrew K. Groves
- Department of NeuroscienceBaylor College of MedicineHoustonTXUSA
- Program in Developmental BiologyBaylor College of MedicineHoustonTXUSA
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTXUSA
| |
Collapse
|
32
|
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]
|
33
|
Żak M, Klis SFL, Grolman W. The Wnt and Notch signalling pathways in the developing cochlea: Formation of hair cells and induction of regenerative potential. Int J Dev Neurosci 2015; 47:247-58. [PMID: 26471908 DOI: 10.1016/j.ijdevneu.2015.09.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 09/25/2015] [Accepted: 09/26/2015] [Indexed: 12/21/2022] Open
Abstract
The Wnt and Notch signalling pathways control proliferation, specification, and cell fate choices during embryonic development and in adult life. Hence, there is much interest in both signalling pathways in the context of stem cell biology and tissue regeneration. In the developing ear, the Wnt and Notch signalling pathways specify otic cells and refine the ventral boundary of the otic placode. Since both signalling pathways control events essential for the formation of sensory cells, such as proliferation and hair cell differentiation, these pathways could hold promise for the regeneration of hair cells in adult mammalian cochlea. Indeed, modulating either the Wnt or Notch pathways can trigger the regenerative potential of supporting cells. In the neonatal mouse cochlea, Notch-mediated regeneration of hair cells partially depends on Wnt signalling, which implies an interaction between the pathways. This review presents how the Wnt and Notch signalling pathways regulate the formation of sensory hair cells and how modulating their activity induces regenerative potential in the mammalian cochlea.
Collapse
Affiliation(s)
- Magdalena Żak
- Department of Otorhinolaryngology and Head & Neck Surgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Room G.02.531, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands.
| | - Sjaak F L Klis
- Department of Otorhinolaryngology and Head & Neck Surgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Room G.02.531, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
| | - Wilko Grolman
- Department of Otorhinolaryngology and Head & Neck Surgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Room G.02.531, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
| |
Collapse
|
34
|
She ZY, Yang WX. SOX family transcription factors involved in diverse cellular events during development. Eur J Cell Biol 2015; 94:547-63. [PMID: 26340821 DOI: 10.1016/j.ejcb.2015.08.002] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 08/11/2015] [Accepted: 08/11/2015] [Indexed: 12/22/2022] Open
Abstract
In metazoa, SOX family transcription factors play many diverse roles. In vertebrate, they are well-known regulators of numerous developmental processes. Wide-ranging studies have demonstrated the co-expression of SOX proteins in various developing tissues and that they occur in an overlapping manner and show functional redundancy. In particular, studies focusing on the HMG box of SOX proteins have revealed that the HMG box regulates DNA-binding properties, and mediates both the nucleocytoplasmic shuttling of SOX proteins and their physical interactions with partner proteins. Posttranslational modifications are further implicated in the regulation of the transcriptional activities of SOX proteins. In this review, we discuss the underlying molecular mechanisms involved in the SOX-partner factor interactions and the functional modes of SOX-partner complexes during development. We particularly emphasize the representative roles of the SOX group proteins in major tissues during developmental and physiological processes.
Collapse
Affiliation(s)
- Zhen-Yu She
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou 310058, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou 310058, China.
| |
Collapse
|
35
|
Atkinson PJ, Huarcaya Najarro E, Sayyid ZN, Cheng AG. Sensory hair cell development and regeneration: similarities and differences. Development 2015; 142:1561-71. [PMID: 25922522 DOI: 10.1242/dev.114926] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sensory hair cells are mechanoreceptors of the auditory and vestibular systems and are crucial for hearing and balance. In adult mammals, auditory hair cells are unable to regenerate, and damage to these cells results in permanent hearing loss. By contrast, hair cells in the chick cochlea and the zebrafish lateral line are able to regenerate, prompting studies into the signaling pathways, morphogen gradients and transcription factors that regulate hair cell development and regeneration in various species. Here, we review these findings and discuss how various signaling pathways and factors function to modulate sensory hair cell development and regeneration. By comparing and contrasting development and regeneration, we also highlight the utility and limitations of using defined developmental cues to drive mammalian hair cell regeneration.
Collapse
Affiliation(s)
- Patrick J Atkinson
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elvis Huarcaya Najarro
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Zahra N Sayyid
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alan G Cheng
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
36
|
Shida H, Mende M, Takano-Yamamoto T, Osumi N, Streit A, Wakamatsu Y. Otic placode cell specification and proliferation are regulated by Notch signaling in avian development. Dev Dyn 2015; 244:839-51. [PMID: 25970828 DOI: 10.1002/dvdy.24291] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 04/26/2015] [Accepted: 05/01/2015] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND The entire inner ear including the cochlear-vestibular ganglion arises from a simple epithelium, the otic placode. Precursors for the placode originate from a pool of progenitors located in ectoderm next to the future hindbrain, the pre-otic field, where they are intermingled with future epibranchial and epidermal cells. While the importance of secreted proteins, such as FGFs and Wnts, in imparting otic identity has been well studied, how precursors for these different fates segregate locally is less well understood. RESULTS (1) The Notch ligand Delta1 and the Notch target Hes5-2 are expressed in a part of pre-otic field before otic commitment, indicative of active Notch signaling, and this is confirmed using a Notch reporter. (2) Loss and gain-of-function approaches reveal that Notch signaling regulates both proliferation and specification of pre-otic progenitors. CONCLUSIONS Our results identify a novel function of Notch signaling in cell fate determination in the pre-otic field of avian embryos.
Collapse
Affiliation(s)
- Hiroko Shida
- Department of Developmental Neuroscience, Center for Translational and Advanced Animal Research, Tohoku University Graduate School of Medicine, Miyagi, Japan.,Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Miyagi, Japan
| | - Michael Mende
- Department of Craniofacial Development and Stem Cell Biology, King's College London
| | - Teruko Takano-Yamamoto
- Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Miyagi, Japan
| | - Noriko Osumi
- Department of Developmental Neuroscience, Center for Translational and Advanced Animal Research, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Andrea Streit
- Department of Craniofacial Development and Stem Cell Biology, King's College London
| | - Yoshio Wakamatsu
- Department of Developmental Neuroscience, Center for Translational and Advanced Animal Research, Tohoku University Graduate School of Medicine, Miyagi, Japan
| |
Collapse
|
37
|
Romero-Guevara R, Cencetti F, Donati C, Bruni P. Sphingosine 1-phosphate signaling pathway in inner ear biology. New therapeutic strategies for hearing loss? Front Aging Neurosci 2015; 7:60. [PMID: 25954197 PMCID: PMC4407579 DOI: 10.3389/fnagi.2015.00060] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 04/08/2015] [Indexed: 12/13/2022] Open
Abstract
Hearing loss is one of the most prevalent conditions around the world, in particular among people over 60 years old. Thus, an increase of this affection is predicted as result of the aging process in our population. In this context, it is important to further explore the function of molecular targets involved in the biology of inner ear sensory cells to better individuate new candidates for therapeutic application. One of the main causes of deafness resides into the premature death of hair cells and auditory neurons. In this regard, neurotrophins and growth factors such as insulin like growth factor are known to be beneficial by favoring the survival of these cells. An elevated number of published data in the last 20 years have individuated sphingolipids not only as structural components of biological membranes but also as critical regulators of key biological processes, including cell survival. Ceramide, formed by catabolism of sphingomyelin (SM) and other complex sphingolipids, is a strong inducer of apoptotic pathway, whereas sphingosine 1-phosphate (S1P), generated by cleavage of ceramide to sphingosine and phosphorylation catalyzed by two distinct sphingosine kinase (SK) enzymes, stimulates cell survival. Interestingly S1P, by acting as intracellular mediator or as ligand of a family of five distinct S1P receptors (S1P1–S1P5), is a very powerful bioactive sphingolipid, capable of triggering also other diverse cellular responses such as cell migration, proliferation and differentiation, and is critically involved in the development and homeostasis of several organs and tissues. Although new interesting data have become available, the information on S1P pathway and other sphingolipids in the biology of the inner ear is limited. Nonetheless, there are several lines of evidence implicating these signaling molecules during neurogenesis in other cell populations. In this review, we discuss the role of S1P during inner ear development, also as guidance for future studies.
Collapse
Affiliation(s)
- Ricardo Romero-Guevara
- Department Scienze Biomediche Sperimentali e Cliniche "Mario Serio", University of Florence Firenze, Italy
| | - Francesca Cencetti
- Department Scienze Biomediche Sperimentali e Cliniche "Mario Serio", University of Florence Firenze, Italy
| | - Chiara Donati
- Department Scienze Biomediche Sperimentali e Cliniche "Mario Serio", University of Florence Firenze, Italy
| | - Paola Bruni
- Department Scienze Biomediche Sperimentali e Cliniche "Mario Serio", University of Florence Firenze, Italy
| |
Collapse
|
38
|
Eddison M, Weber SJ, Ariza-McNaughton L, Lewis J, Daudet N. Numb is not a critical regulator of Notch-mediated cell fate decisions in the developing chick inner ear. Front Cell Neurosci 2015; 9:74. [PMID: 25814931 PMCID: PMC4357303 DOI: 10.3389/fncel.2015.00074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 02/19/2015] [Indexed: 11/27/2022] Open
Abstract
The Notch signaling pathway controls differentiation of hair cells and supporting cells in the vertebrate inner ear. Here, we have investigated whether Numb, a known regulator of Notch activity in Drosophila, is involved in this process in the embryonic chick. The chicken homolog of Numb is expressed throughout the otocyst at early stages of development and is concentrated at the basal pole of the cells. It is asymmetrically allocated at some cell divisions, as in Drosophila, suggesting that it could act as a determinant inherited by one of the two daughter cells and favoring adoption of a hair-cell fate. To test the implication of Numb in hair cell fate decisions and the regulation of Notch signaling, we used different methods to overexpress Numb at different stages of inner ear development. We found that sustained or late Numb overexpression does not promote hair cell differentiation, and Numb does not prevent the reception of Notch signaling. Surprisingly, none of the Numb-overexpressing cells differentiated into hair cells, suggesting that high levels of Numb protein could interfere with intracellular processes essential for hair cell survival. However, when Numb was overexpressed early and more transiently during ear development, no effect on hair cell formation was seen. These results suggest that in the inner ear at least, Numb does not significantly repress Notch activity and that its asymmetric distribution in dividing precursor cells does not govern the choice between hair cell and supporting cell fates.
Collapse
Affiliation(s)
- Mark Eddison
- Howard Hughes Medical Institute, Janelia Research Campus Ashburn, VA, USA
| | - Sara J Weber
- Ear Institute, University College London London, UK
| | - Linda Ariza-McNaughton
- Haematopoietic Stem cell Laboratory, Cancer Research UK, London Research Institute London, UK
| | - Julian Lewis
- Formerly of Vertebrate Development Laboratory, Cancer Research UK London, UK
| | | |
Collapse
|
39
|
Maass JC, Gu R, Basch ML, Waldhaus J, Lopez EM, Xia A, Oghalai JS, Heller S, Groves AK. Changes in the regulation of the Notch signaling pathway are temporally correlated with regenerative failure in the mouse cochlea. Front Cell Neurosci 2015; 9:110. [PMID: 25873862 PMCID: PMC4379755 DOI: 10.3389/fncel.2015.00110] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/10/2015] [Indexed: 12/20/2022] Open
Abstract
Sensorineural hearing loss is most commonly caused by the death of hair cells in the organ of Corti, and once lost, mammalian hair cells do not regenerate. In contrast, other vertebrates such as birds can regenerate hair cells by stimulating division and differentiation of neighboring supporting cells. We currently know little of the genetic networks which become active in supporting cells when hair cells die and that are activated in experimental models of hair cell regeneration. Several studies have shown that neonatal mammalian cochlear supporting cells are able to trans-differentiate into hair cells when cultured in conditions in which the Notch signaling pathway is blocked. We now show that the ability of cochlear supporting cells to trans-differentiate declines precipitously after birth, such that supporting cells from six-day-old mouse cochlea are entirely unresponsive to a blockade of the Notch pathway. We show that this trend is seen regardless of whether the Notch pathway is blocked with gamma secretase inhibitors, or by antibodies against the Notch1 receptor, suggesting that the action of gamma secretase inhibitors on neonatal supporting cells is likely to be by inhibiting Notch receptor cleavage. The loss of responsiveness to inhibition of the Notch pathway in the first postnatal week is due in part to a down-regulation of Notch receptors and ligands, and we show that this down-regulation persists in the adult animal, even under conditions of noise damage. Our data suggest that the Notch pathway is used to establish the repeating pattern of hair cells and supporting cells in the organ of Corti, but is not required to maintain this cellular mosaic once the production of hair cells and supporting cells is completed. Our results have implications for the proposed used of Notch pathway inhibitors in hearing restoration therapies.
Collapse
Affiliation(s)
- Juan C Maass
- Department of Neuroscience, Baylor College of Medicine Houston, TX, USA ; Department of Otolaryngology, Hospital Clínico Universidad de Chile Santiago, Chile ; Interdisciplinary Program of Physiology and Biophysics, ICBM Universidad de Chile Santiago, Chile ; Department of Otolaryngology, Clínica Alemana de Santiago, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo Santiago, Chile
| | - Rende Gu
- Department of Neuroscience, Baylor College of Medicine Houston, TX, USA
| | - Martin L Basch
- Department of Neuroscience, Baylor College of Medicine Houston, TX, USA
| | - Joerg Waldhaus
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine Palo Alto, CA, USA
| | | | - Anping Xia
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine Palo Alto, CA, USA
| | - John S Oghalai
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine Palo Alto, CA, USA
| | - Stefan Heller
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine Palo Alto, CA, USA
| | - Andrew K Groves
- Department of Neuroscience, Baylor College of Medicine Houston, TX, USA ; Department of Molecular and Human Genetics, Baylor College of Medicine Houston, TX, USA ; Program in Developmental Biology, Baylor College of Medicine Houston, TX, USA
| |
Collapse
|
40
|
Brown AS, Rakowiecki SM, Li JYH, Epstein DJ. The cochlear sensory epithelium derives from Wnt responsive cells in the dorsomedial otic cup. Dev Biol 2015; 399:177-187. [PMID: 25592224 DOI: 10.1016/j.ydbio.2015.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/11/2014] [Accepted: 01/02/2015] [Indexed: 02/07/2023]
Abstract
Wnt1 and Wnt3a secreted from the dorsal neural tube were previously shown to regulate a gene expression program in the dorsal otic vesicle that is necessary for vestibular morphogenesis (Riccomagno et al., 2005. Genes Dev. 19, 1612-1623). Unexpectedly, Wnt1(-/-); Wnt3a(-/-) embryos also displayed a pronounced defect in the outgrowth of the ventrally derived cochlear duct. To determine how Wnt signaling in the dorsal otocyst contributes to cochlear development we performed a series of genetic fate mapping experiments using two independent Wnt responsive driver strains (TopCreER and Gbx2(CreER)) that when crossed to inducible responder lines (Rosa(lacZ) or Rosa(zsGreen)) permanently labeled dorsomedial otic progenitors and their derivatives. Tamoxifen time course experiments revealed that most vestibular structures showed some degree of labeling when recombination was induced between E7.75 and E12.5, consistent with continuous Wnt signaling activity in this tissue. Remarkably, a population of Wnt responsive cells in the dorsal otocyst was also found to contribute to the sensory epithelium of the cochlear duct, including auditory hair and support cells. Similar results were observed with both TopCreER and Gbx2(CreER) strains. The ventral displacement of Wnt responsive cells followed a spatiotemporal sequence that initiated in the anterior otic cup at, or immediately prior to, the 17-somite stage (E9) and then spread progressively to the posterior pole of the otic vesicle by the 25-somite stage (E9.5). These lineage-tracing experiments identify the earliest known origin of auditory sensory progenitors within a population of Wnt responsive cells in the dorsomedial otic cup.
Collapse
Affiliation(s)
- Alexander S Brown
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, 415 Curie Blvd, Philadelphia, PA 19104, USA
| | - Staci M Rakowiecki
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, 415 Curie Blvd, Philadelphia, PA 19104, USA
| | - James Y H Li
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, 400 Farmington Avenue, Farmington, CT 06030-6403, USA
| | - Douglas J Epstein
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, 415 Curie Blvd, Philadelphia, PA 19104, USA.
| |
Collapse
|
41
|
Faure S, McKey J, Sagnol S, de Santa Barbara P. Enteric neural crest cells regulate vertebrate stomach patterning and differentiation. Development 2015; 142:331-42. [DOI: 10.1242/dev.118422] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
In vertebrates, the digestive tract develops from a uniform structure where reciprocal epithelial-mesenchymal interactions pattern this complex organ into regions with specific morphologies and functions. Concomitant with these early patterning events, the primitive GI tract is colonized by the vagal enteric neural crest cells (vENCCs), a population of cells that will give rise to the enteric nervous system (ENS), the intrinsic innervation of the GI tract. The influence of vENCCs on early patterning and differentiation of the GI tract has never been evaluated. In this study, we report that a crucial number of vENCCs is required for proper chick stomach development, patterning and differentiation. We show that reducing the number of vENCCs by performing vENCC ablations induces sustained activation of the BMP and Notch pathways in the stomach mesenchyme and impairs smooth muscle development. A reduction in vENCCs also leads to the transdifferentiation of the stomach into a stomach-intestinal mixed phenotype. In addition, sustained Notch signaling activity in the stomach mesenchyme phenocopies the defects observed in vENCC-ablated stomachs, indicating that inhibition of the Notch signaling pathway is essential for stomach patterning and differentiation. Finally, we report that a crucial number of vENCCs is also required for maintenance of stomach identity and differentiation through inhibition of the Notch signaling pathway. Altogether, our data reveal that, through the regulation of mesenchyme identity, vENCCs act as a new mediator in the mesenchymal-epithelial interactions that control stomach development.
Collapse
Affiliation(s)
- Sandrine Faure
- INSERM U1046, Université Montpellier 1, Université Montpellier 2, Montpellier 34295, France
| | - Jennifer McKey
- INSERM U1046, Université Montpellier 1, Université Montpellier 2, Montpellier 34295, France
| | - Sébastien Sagnol
- INSERM U1046, Université Montpellier 1, Université Montpellier 2, Montpellier 34295, France
| | | |
Collapse
|
42
|
Freeman SD, Keino-Masu K, Masu M, Ladher RK. Expression of the heparan sulfate 6-O-endosulfatases, Sulf1 and Sulf2, in the avian and mammalian inner ear suggests a role for sulfation during inner ear development. Dev Dyn 2014; 244:168-80. [PMID: 25370455 DOI: 10.1002/dvdy.24223] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 10/21/2014] [Accepted: 10/21/2014] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Inner ear morphogenesis is tightly regulated by the temporally and spatially coordinated action of signaling ligands and their receptors. Ligand-receptor interactions are influenced by heparan sulfate proteoglycans (HSPGs), cell surface molecules that consist of glycosaminoglycan chains bound to a protein core. Diversity in the sulfation pattern within glycosaminoglycan chains creates binding sites for numerous cell signaling factors, whose activities and distribution are modified by their association with HSPGs. RESULTS Here we describe the expression patterns of two extracellular 6-O-endosulfatases, Sulf1 and Sulf2, whose activity modifies the 6-O-sulfation pattern of HSPGs. We use in situ hybridization to determine the temporal and spatial distribution of transcripts during the development of the chick and mouse inner ear. We also use immunocytochemistry to determine the cellular localization of Sulf1 and Sulf2 within the sensory epithelia. Furthermore, we analyze the organ of Corti in Sulf1/Sulf2 double knockout mice and describe an increase in the number of mechanosensory hair cells. CONCLUSIONS Our results suggest that the tuning of intracellular signaling, mediated by Sulf activity, plays an important role in the development of the inner ear.
Collapse
Affiliation(s)
- Stephen D Freeman
- Laboratory for Sensory Development, RIKEN Center for Developmental Biology, Chuo-ku, Kobe-shi, Japan
| | | | | | | |
Collapse
|
43
|
Petrovic J, Gálvez H, Neves J, Abelló G, Giraldez F. Differential regulation of Hes/Hey genes during inner ear development. Dev Neurobiol 2014; 75:703-20. [PMID: 25363712 DOI: 10.1002/dneu.22243] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 10/28/2014] [Indexed: 11/09/2022]
Abstract
Notch signaling plays a crucial role during inner ear development and regeneration. Hes/Hey genes encode for bHLH transcription factors identified as Notch targets. We have studied the expression and regulation of Hes/Hey genes during inner ear development in the chicken embryo. Among several Hes/Hey genes examined, only Hey1 and Hes5 map to the sensory regions, although with salient differences. Hey1 expression follows Jag1 expression except at early prosensory stages while Hes5 expression corresponds well to Dl1 expression throughout otic development. Although Hey1 and Hes5 are direct Notch downstream targets, they differ in the level of Notch required for activation. Moreover, they also differ in mRNA stability, showing different temporal decays after Notch blockade. In addition, Bmp, Wnt and Fgf pathways also modify Hey1 and Hes5 expression in the inner ear. Particularly, the Wnt pathway modulates Hey1 and Jag1 expression. Finally, gain of function experiments show that Hey1 and Hes5 cross-regulate each other in a complex manner. Both Hey1 and Hes5 repress Dl1 and Hes5 expression, suggesting that they prevent the transition to differentiation stages, probably by preventing Atoh1 expression. In spite of its association with Jag1, Hey1 does not seem to be instrumental for lateral induction as it does not promote Jag1 expression. We suggest that, besides being both targets of Notch, Hey1 and Hes5 are subject to a rather complex regulation that includes the stability of their transcripts, cross regulation and other signaling pathways.
Collapse
Affiliation(s)
- Jelena Petrovic
- Developmental Biology Unit, CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| | - Hector Gálvez
- Developmental Biology Unit, CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| | - Joana Neves
- Developmental Biology Unit, CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| | - Gina Abelló
- Developmental Biology Unit, CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| | - Fernando Giraldez
- Developmental Biology Unit, CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| |
Collapse
|
44
|
Voelkel JE, Harvey JA, Adams JS, Lassiter RN, Stark MR. FGF and Notch signaling in sensory neuron formation: A multifactorial approach to understanding signaling pathway hierarchy. Mech Dev 2014; 134:55-66. [DOI: 10.1016/j.mod.2014.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 08/08/2014] [Accepted: 09/21/2014] [Indexed: 10/24/2022]
|
45
|
Radosevic M, Fargas L, Alsina B. The role of her4 in inner ear development and its relationship with proneural genes and Notch signalling. PLoS One 2014; 9:e109860. [PMID: 25299450 PMCID: PMC4192589 DOI: 10.1371/journal.pone.0109860] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/08/2014] [Indexed: 01/13/2023] Open
Abstract
The generation of sensory neurons and hair cells of the inner ear is under tight control. Different members of the Hairy and Enhancer of Split genes (HES) are expressed in the inner ear, their full array of functions still not being disclosed. We have previously shown that zebrafish her9 acts as a patterning gene to restrict otic neurogenesis to an anterior domain. Here, we disclose the role of another her gene, her4, a zebrafish ortholog of Hes5 that is expressed in the neurogenic and sensory domains of the inner ear. The expression of her4 is highly dynamic and spatiotemporally regulated. We demonstrate by loss of function experiments that in the neurogenic domain her4 expression is under the regulation of neurogenin1 (neurog1) and the Notch pathway. Moreover, her4 participates in lateral inhibition during otic neurogenesis since her4 knockdown results in overproduction of the number of neurog1 and deltaB-positive otic neurons. In contrast, during sensorigenesis her4 is initially Notch-independent and induced by atoh1b in a broad prosensory domain. At later stages her4 expression becomes Notch-dependent in the future sensory domains but loss of her4 does not result in hair cell overproduction, suggesting that there other her genes can compensate its function.
Collapse
Affiliation(s)
- Marija Radosevic
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, Barcelona, Spain
| | - Laura Fargas
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, Barcelona, Spain
| | - Berta Alsina
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, Barcelona, Spain
- * E-mail:
| |
Collapse
|
46
|
Raft S, Groves AK. Segregating neural and mechanosensory fates in the developing ear: patterning, signaling, and transcriptional control. Cell Tissue Res 2014; 359:315-32. [PMID: 24902666 DOI: 10.1007/s00441-014-1917-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Accepted: 05/08/2014] [Indexed: 12/21/2022]
Abstract
The vertebrate inner ear is composed of multiple sensory receptor epithelia, each of which is specialized for detection of sound, gravity, or angular acceleration. Each receptor epithelium contains mechanosensitive hair cells, which are connected to the brainstem by bipolar sensory neurons. Hair cells and their associated neurons are derived from the embryonic rudiment of the inner ear epithelium, but the precise spatial and temporal patterns of their generation, as well as the signals that coordinate these events, have only recently begun to be understood. Gene expression, lineage tracing, and mutant analyses suggest that both neurons and hair cells are generated from a common domain of neural and sensory competence in the embryonic inner ear rudiment. Members of the Shh, Wnt, and FGF families, together with retinoic acid signals, regulate transcription factor genes within the inner ear rudiment to establish the axial identity of the ear and regionalize neurogenic activity. Close-range signaling, such as that of the Notch pathway, specifies the fate of sensory regions and individual cell types. We also describe positive and negative interactions between basic helix-loop-helix and SoxB family transcription factors that specify either neuronal or sensory fates in a context-dependent manner. Finally, we review recent work on inner ear development in zebrafish, which demonstrates that the relative timing of neurogenesis and sensory epithelial formation is not phylogenetically constrained.
Collapse
Affiliation(s)
- Steven Raft
- Section on Sensory Cell Regeneration and Development, National Institute on Deafness and Other Communication Disorders National Institutes of Health, 35 Convent Drive, Bethesda, MD, 20892, USA,
| | | |
Collapse
|
47
|
Petrovic J, Formosa-Jordan P, Luna-Escalante JC, Abelló G, Ibañes M, Neves J, Giraldez F. Ligand-dependent Notch signaling strength orchestrates lateral induction and lateral inhibition in the developing inner ear. Development 2014; 141:2313-24. [PMID: 24821984 DOI: 10.1242/dev.108100] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
During inner ear development, Notch exhibits two modes of operation: lateral induction, which is associated with prosensory specification, and lateral inhibition, which is involved in hair cell determination. These mechanisms depend respectively on two different ligands, jagged 1 (Jag1) and delta 1 (Dl1), that rely on a common signaling cascade initiated after Notch activation. In the chicken otocyst, expression of Jag1 and the Notch target Hey1 correlates well with lateral induction, whereas both Jag1 and Dl1 are expressed during lateral inhibition, as are Notch targets Hey1 and Hes5. Here, we show that Jag1 drives lower levels of Notch activity than Dl1, which results in the differential expression of Hey1 and Hes5. In addition, Jag1 interferes with the ability of Dl1 to elicit high levels of Notch activity. Modeling the sensory epithelium when the two ligands are expressed together shows that ligand regulation, differential signaling strength and ligand competition are crucial to allow the two modes of operation and for establishing the alternate pattern of hair cells and supporting cells. Jag1, while driving lateral induction on its own, facilitates patterning by lateral inhibition in the presence of Dl1. This novel behavior emerges from Jag1 acting as a competitive inhibitor of Dl1 for Notch signaling. Both modeling and experiments show that hair cell patterning is very robust. The model suggests that autoactivation of proneural factor Atoh1, upstream of Dl1, is a fundamental component for robustness. The results stress the importance of the levels of Notch signaling and ligand competition for Notch function.
Collapse
Affiliation(s)
- Jelena Petrovic
- Developmental Biology Unit, CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain
| | - Pau Formosa-Jordan
- Departament d'Estructura i Constituents de la Matèria, Facultat de Física, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Juan C Luna-Escalante
- Departament d'Estructura i Constituents de la Matèria, Facultat de Física, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Gina Abelló
- Developmental Biology Unit, CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain
| | - Marta Ibañes
- Departament d'Estructura i Constituents de la Matèria, Facultat de Física, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Joana Neves
- Developmental Biology Unit, CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain
| | - Fernando Giraldez
- Developmental Biology Unit, CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain
| |
Collapse
|
48
|
Durruthy-Durruthy R, Gottlieb A, Hartman BH, Waldhaus J, Laske RD, Altman R, Heller S. Reconstruction of the mouse otocyst and early neuroblast lineage at single-cell resolution. Cell 2014; 157:964-78. [PMID: 24768691 PMCID: PMC4051200 DOI: 10.1016/j.cell.2014.03.036] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 02/03/2014] [Accepted: 03/12/2014] [Indexed: 01/24/2023]
Abstract
The otocyst harbors progenitors for most cell types of the mature inner ear. Developmental lineage analyses and gene expression studies suggest that distinct progenitor populations are compartmentalized to discrete axial domains in the early otocyst. Here, we conducted highly parallel quantitative RT-PCR measurements on 382 individual cells from the developing otocyst and neuroblast lineages to assay 96 genes representing established otic markers, signaling-pathway-associated transcripts, and novel otic-specific genes. By applying multivariate cluster, principal component, and network analyses to the data matrix, we were able to readily distinguish the delaminating neuroblasts and to describe progressive states of gene expression in this population at single-cell resolution. It further established a three-dimensional model of the otocyst in which each individual cell can be precisely mapped into spatial expression domains. Our bioinformatic modeling revealed spatial dynamics of different signaling pathways active during early neuroblast development and prosensory domain specification.
Collapse
Affiliation(s)
- Robert Durruthy-Durruthy
- Department of Otolaryngology, Head & Neck Surgery and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Assaf Gottlieb
- Departments of Bioengineering and Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Byron H Hartman
- Department of Otolaryngology, Head & Neck Surgery and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jörg Waldhaus
- Department of Otolaryngology, Head & Neck Surgery and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Roman D Laske
- Department of Otolaryngology, Head & Neck Surgery and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Russ Altman
- Departments of Bioengineering and Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stefan Heller
- Department of Otolaryngology, Head & Neck Surgery and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
49
|
Sensational placodes: neurogenesis in the otic and olfactory systems. Dev Biol 2014; 389:50-67. [PMID: 24508480 PMCID: PMC3988839 DOI: 10.1016/j.ydbio.2014.01.023] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 01/27/2014] [Accepted: 01/28/2014] [Indexed: 11/22/2022]
Abstract
For both the intricate morphogenetic layout of the sensory cells in the ear and the elegantly radial arrangement of the sensory neurons in the nose, numerous signaling molecules and genetic determinants are required in concert to generate these specialized neuronal populations that help connect us to our environment. In this review, we outline many of the proteins and pathways that play essential roles in the differentiation of otic and olfactory neurons and their integration into their non-neuronal support structures. In both cases, well-known signaling pathways together with region-specific factors transform thickened ectodermal placodes into complex sense organs containing numerous, diverse neuronal subtypes. Olfactory and otic placodes, in combination with migratory neural crest stem cells, generate highly specialized subtypes of neuronal cells that sense sound, position and movement in space, odors and pheromones throughout our lives.
Collapse
|
50
|
Ono K, Kita T, Sato S, O'Neill P, Mak SS, Paschaki M, Ito M, Gotoh N, Kawakami K, Sasai Y, Ladher RK. FGFR1-Frs2/3 signalling maintains sensory progenitors during inner ear hair cell formation. PLoS Genet 2014; 10:e1004118. [PMID: 24465223 PMCID: PMC3900395 DOI: 10.1371/journal.pgen.1004118] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 12/02/2013] [Indexed: 12/11/2022] Open
Abstract
Inner ear mechanosensory hair cells transduce sound and balance information. Auditory hair cells emerge from a Sox2-positive sensory patch in the inner ear epithelium, which is progressively restricted during development. This restriction depends on the action of signaling molecules. Fibroblast growth factor (FGF) signalling is important during sensory specification: attenuation of Fgfr1 disrupts cochlear hair cell formation; however, the underlying mechanisms remain unknown. Here we report that in the absence of FGFR1 signaling, the expression of Sox2 within the sensory patch is not maintained. Despite the down-regulation of the prosensory domain markers, p27Kip1, Hey2, and Hes5, progenitors can still exit the cell cycle to form the zone of non-proliferating cells (ZNPC), however the number of cells that form sensory cells is reduced. Analysis of a mutant Fgfr1 allele, unable to bind to the adaptor protein, Frs2/3, indicates that Sox2 maintenance can be regulated by MAP kinase. We suggest that FGF signaling, through the activation of MAP kinase, is necessary for the maintenance of sensory progenitors and commits precursors to sensory cell differentiation in the mammalian cochlea. The ability of our brain to perceive sound depends on its conversion into electrical impulses within the cochlea of the inner ear. The cochlea has dedicated specialized cells, called inner ear hair cells, which register sound energy. Environmental effects, genetic disorders or just the passage of time can damage these cells, and the damage impairs our ability to hear. If we could understand how these cells develop, we might be able to exploit this knowledge to generate new hair cells. In this study we address an old problem: how do signals from the fibroblast growth factor (FGF) family control hair cell number? We used mice in which one of the receptors for FGF (Fgfr1) is mutated and found that the expression of a stem cell protein, Sox2 is not maintained. Sox2 generally acts to keep precursors in the cochlea in a pre-hair cell state. However, in mutant mice Sox2 expression is transient, diminishing the ability of precursors to commit to a hair cell fate. These findings suggest that it may be possible to amplify the number of hair cell progenitors in culture by tuning FGF activity, providing a route to replace damaged inner ear hair cells.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Cell Cycle
- Cell Differentiation/genetics
- Cochlea/growth & development
- Cochlea/metabolism
- Ear, Inner/cytology
- Ear, Inner/growth & development
- Epithelium/growth & development
- Epithelium/metabolism
- Gene Expression Regulation, Developmental
- Hair Cells, Auditory, Inner/cytology
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Protein Binding
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- SOXB1 Transcription Factors/genetics
- Signal Transduction
Collapse
Affiliation(s)
- Kazuya Ono
- Sensory Development, RIKEN Center for Developmental Biology, Chuo-ku, Kobe, Japan
- Neurogenesis and Organogenesis, RIKEN Center for Developmental Biology, Chuo-ku, Kobe, Japan
| | - Tomoko Kita
- Sensory Development, RIKEN Center for Developmental Biology, Chuo-ku, Kobe, Japan
| | - Shigeru Sato
- Division of Biology, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Paul O'Neill
- Sensory Development, RIKEN Center for Developmental Biology, Chuo-ku, Kobe, Japan
| | - Siu-Shan Mak
- Sensory Development, RIKEN Center for Developmental Biology, Chuo-ku, Kobe, Japan
| | - Marie Paschaki
- Sensory Development, RIKEN Center for Developmental Biology, Chuo-ku, Kobe, Japan
| | - Masataka Ito
- Department of Anatomy, National Defense Medical College, Tokorozawa, Japan
| | - Noriko Gotoh
- Division of Genetics, Institute of Medical Science, University of Tokyo, Minato-ku, Japan
| | - Kiyoshi Kawakami
- Division of Biology, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Yoshiki Sasai
- Neurogenesis and Organogenesis, RIKEN Center for Developmental Biology, Chuo-ku, Kobe, Japan
| | - Raj K. Ladher
- Sensory Development, RIKEN Center for Developmental Biology, Chuo-ku, Kobe, Japan
- * E-mail:
| |
Collapse
|