1
|
Erjavec E, Angée C, Hadjadj D, Passet B, David P, Kostic C, Dodé E, Zanlonghi X, Cagnard N, Nedelec B, Crippa SV, Bole-Feysot C, Zarhrate M, Creuzet S, Castille J, Vilotte JL, Calvas P, Plaisancié J, Chassaing N, Kaplan J, Rozet JM, Fares Taie L. Congenital microcoria deletion in mouse links Sox21 dysregulation to disease and suggests a role for TGFB2 in glaucoma and myopia. Am J Hum Genet 2024; 111:2265-2282. [PMID: 39293448 PMCID: PMC11480854 DOI: 10.1016/j.ajhg.2024.08.019] [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: 01/12/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/20/2024] Open
Abstract
Congenital microcoria (MCOR) is a rare hereditary developmental defect of the iris dilator muscle frequently associated with high axial myopia and high intraocular pressure (IOP) glaucoma. The condition is caused by submicroscopic rearrangements of chromosome 13q32.1. However, the mechanisms underlying the failure of iris development and the origin of associated features remain elusive. Here, we present a 3D architecture model of the 13q32.1 region, demonstrating that MCOR-related deletions consistently disrupt the boundary between two topologically associating domains (TADs). Deleting the critical MCOR-causing region in mice reveals ectopic Sox21 expression precisely aligning with Dct, each located in one of the two neighbor TADs. This observation is consistent with the TADs' boundary alteration and adoption of Dct regulatory elements by the Sox21 promoter. Additionally, we identify Tgfb2 as a target gene of SOX21 and show TGFΒ2 accumulation in the aqueous humor of an MCOR-affected subject. Accumulation of TGFB2 is recognized for its role in glaucoma and potential impact on axial myopia. Our results highlight the importance of SOX21-TGFB2 signaling in iris development and control of eye growth and IOP. Insights from MCOR studies may provide therapeutic avenues for this condition but also for glaucoma and high myopia conditions, affecting millions of people.
Collapse
Affiliation(s)
- Elisa Erjavec
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetic Diseases, Imagine and Université Paris Cité, Paris, France
| | - Clémentine Angée
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetic Diseases, Imagine and Université Paris Cité, Paris, France
| | - Djihad Hadjadj
- Institut Cochin, Inserm U1016, CNRS UMR8104, UFR de Pharmacie de Paris, Université Paris Cité, CARPEM, Paris, France
| | - Bruno Passet
- University of Paris-Saclay, INRAE, AgroParisTech, UMR1313 GABI, Jouy-en-Josas, France
| | - Pierre David
- Transgenesis Platform, Laboratoire d'Expérimentation Animale et Transgenèse (LEAT), Imagine Institute, Structure Fédérative de Recherche Necker INSERM US24/CNRS UMS3633, Paris, France
| | - Corinne Kostic
- Group for Retinal Disorder Research, Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Emmanuel Dodé
- Institut Ophtalmologique de L'Ouest-Clinique Jules VERNE, Nantes, France
| | - Xavier Zanlonghi
- Institut Ophtalmologique de L'Ouest-Clinique Jules VERNE, Nantes, France
| | - Nicolas Cagnard
- Université Paris Cité, Bioinformatics Core Facility, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Brigitte Nedelec
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetic Diseases, Imagine and Université Paris Cité, Paris, France
| | - Sylvain V Crippa
- Group for Retinal Disorder Research, Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Christine Bole-Feysot
- Université Paris Cité, Genomics Platform, Imagine Institute, INSERM UMR 1163, INSERM US24/CNRS UAR3633, Paris, France
| | - Mohammed Zarhrate
- Université Paris Cité, Genomics Platform, Imagine Institute, INSERM UMR 1163, INSERM US24/CNRS UAR3633, Paris, France
| | - Sophie Creuzet
- Paris-Saclay Institute of Neuroscience, NeuroPSI, CNRS, Paris-Saclay University, Campus CEA Saclay, Bât 151, 151 Route de la Rotonde, 91400 Saclay, France
| | - Johan Castille
- University of Paris-Saclay, INRAE, AgroParisTech, UMR1313 GABI, Jouy-en-Josas, France
| | - Jean-Luc Vilotte
- University of Paris-Saclay, INRAE, AgroParisTech, UMR1313 GABI, Jouy-en-Josas, France
| | - Patrick Calvas
- Centre de Référence pour les Affections Rares en Génétique Ophtalmologique (CARGO), CHU Toulouse, Toulouse, France
| | - Julie Plaisancié
- Centre de Référence pour les Affections Rares en Génétique Ophtalmologique (CARGO), CHU Toulouse, Toulouse, France
| | - Nicolas Chassaing
- Centre de Référence pour les Affections Rares en Génétique Ophtalmologique (CARGO), CHU Toulouse, Toulouse, France
| | - Josseline Kaplan
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetic Diseases, Imagine and Université Paris Cité, Paris, France
| | - Jean-Michel Rozet
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetic Diseases, Imagine and Université Paris Cité, Paris, France.
| | - L Fares Taie
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetic Diseases, Imagine and Université Paris Cité, Paris, France.
| |
Collapse
|
2
|
Mercurio S. SOX2-Sensing: Insights into the Role of SOX2 in the Generation of Sensory Cell Types in Vertebrates. Int J Mol Sci 2023; 24:ijms24087637. [PMID: 37108798 PMCID: PMC10141063 DOI: 10.3390/ijms24087637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/17/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
The SOX2 transcription factor is a key regulator of nervous system development, and its mutation in humans leads to a rare disease characterized by severe eye defects, cognitive defects, hearing defects, abnormalities of the CNS and motor control problems. SOX2 has an essential role in neural stem cell maintenance in specific regions of the brain, and it is one of the master genes required for the generation of induced pluripotent stem cells. Sox2 is expressed in sensory organs, and this review will illustrate how it regulates the differentiation of sensory cell types required for hearing, touching, tasting and smelling in vertebrates and, in particular, in mice.
Collapse
Affiliation(s)
- Sara Mercurio
- Department of Biotechnologies and Biosciences, University of Milan-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| |
Collapse
|
3
|
Reagor CC, Velez-Angel N, Hudspeth AJ. Depicting pseudotime-lagged causality across single-cell trajectories for accurate gene-regulatory inference. PNAS NEXUS 2023; 2:pgad113. [PMID: 37113980 PMCID: PMC10129065 DOI: 10.1093/pnasnexus/pgad113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 04/29/2023]
Abstract
Identifying the causal interactions in gene-regulatory networks requires an accurate understanding of the time-lagged relationships between transcription factors and their target genes. Here we describe DELAY (short for Depicting Lagged Causality), a convolutional neural network for the inference of gene-regulatory relationships across pseudotime-ordered single-cell trajectories. We show that combining supervised deep learning with joint probability matrices of pseudotime-lagged trajectories allows the network to overcome important limitations of ordinary Granger causality-based methods, for example, the inability to infer cyclic relationships such as feedback loops. Our network outperforms several common methods for inferring gene regulation and, when given partial ground-truth labels, predicts novel regulatory networks from single-cell RNA sequencing (scRNA-seq) and single-cell ATAC sequencing (scATAC-seq) data sets. To validate this approach, we used DELAY to identify important genes and modules in the regulatory network of auditory hair cells, as well as likely DNA-binding partners for two hair cell cofactors (Hist1h1c and Ccnd1) and a novel binding sequence for the hair cell-specific transcription factor Fiz1. We provide an easy-to-use implementation of DELAY under an open-source license at https://github.com/calebclayreagor/DELAY.
Collapse
Affiliation(s)
| | - Nicolas Velez-Angel
- Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | | |
Collapse
|
4
|
Eenjes E, Tibboel D, Wijnen RMH, Schnater JM, Rottier RJ. SOX2 and SOX21 in Lung Epithelial Differentiation and Repair. Int J Mol Sci 2022; 23:13064. [PMID: 36361852 PMCID: PMC9657681 DOI: 10.3390/ijms232113064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 12/05/2022] Open
Abstract
The lung originates from the ventral foregut and develops into an intricate branched structure of airways, alveoli, vessels and support tissue. As the lung develops, cells become specified and differentiate into the various cell lineages. This process is controlled by specific transcription factors, such as the SRY-related HMG-box genes SOX2 and SOX21, that are activated or repressed through intrinsic and extrinsic signals. Disturbances in any of these processes during the development of the lung may lead to various pediatric lung disorders, such as Congenital Diaphragmatic Hernia (CDH), Congenital Pulmonary Airway Malformation (CPAM) and Broncho-Pulmonary Dysplasia (BPD). Changes in the composition of the airways and the alveoli may result in reduced respiratory function and eventually lead to chronic lung disorders. In this concise review, we describe different intrinsic and extrinsic cellular processes required for proper differentiation of the epithelium during development and regeneration, and the influence of the microenvironment on this process with special focus on SOX2 and SOX21.
Collapse
Affiliation(s)
- Evelien Eenjes
- Department of Pediatric Surgery, Erasmus MC-Sophia Children’s Hospital, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Dick Tibboel
- Department of Pediatric Surgery, Erasmus MC-Sophia Children’s Hospital, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Rene M. H. Wijnen
- Department of Pediatric Surgery, Erasmus MC-Sophia Children’s Hospital, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Johannes Marco Schnater
- Department of Pediatric Surgery, Erasmus MC-Sophia Children’s Hospital, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Robbert J. Rottier
- Department of Pediatric Surgery, Erasmus MC-Sophia Children’s Hospital, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
- Department of Cell Biology, Erasmus MC, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
5
|
Eenjes E, Buscop-van Kempen M, Boerema-de Munck A, Edel GG, Benthem F, de Kreij-de Bruin L, Schnater M, Tibboel D, Collins J, Rottier RJ. SOX21 modulates SOX2-initiated differentiation of epithelial cells in the extrapulmonary airways. eLife 2021; 10:57325. [PMID: 34286693 PMCID: PMC8331192 DOI: 10.7554/elife.57325] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/20/2021] [Indexed: 12/23/2022] Open
Abstract
SOX2 expression levels are crucial for the balance between maintenance and differentiation of airway progenitor cells during development and regeneration. Here, we describe patterning of the mouse proximal airway epithelium by SOX21, which coincides with high levels of SOX2 during development. Airway progenitor cells in this SOX2+/SOX21+ zone show differentiation to basal cells, specifying cells for the extrapulmonary airways. Loss of SOX21 showed an increased differentiation of SOX2+ progenitor cells to basal and ciliated cells during mouse lung development. We propose a mechanism where SOX21 inhibits differentiation of airway progenitors by antagonizing SOX2-induced expression of specific genes involved in airway differentiation. Additionally, in the adult tracheal epithelium, SOX21 inhibits basal to ciliated cell differentiation. This suppressing function of SOX21 on differentiation contrasts SOX2, which mainly drives differentiation of epithelial cells during development and regeneration after injury. Furthermore, using human fetal lung organoids and adult bronchial epithelial cells, we show that SOX2+/SOX21+ regionalization is conserved. Lastly, we show that the interplay between SOX2 and SOX21 is context and concentration dependent leading to regulation of differentiation of the airway epithelium.
Collapse
Affiliation(s)
- Evelien Eenjes
- Department of Pediatric Surgery, Erasmus Medical Center - Sophia Children's Hospital, Rotterdam, Netherlands
| | - Marjon Buscop-van Kempen
- Department of Pediatric Surgery, Erasmus Medical Center - Sophia Children's Hospital, Rotterdam, Netherlands
| | - Anne Boerema-de Munck
- Department of Pediatric Surgery, Erasmus Medical Center - Sophia Children's Hospital, Rotterdam, Netherlands
| | - Gabriela G Edel
- Department of Pediatric Surgery, Erasmus Medical Center - Sophia Children's Hospital, Rotterdam, Netherlands
| | - Floor Benthem
- Department of Pediatric Surgery, Erasmus Medical Center - Sophia Children's Hospital, Rotterdam, Netherlands
| | - Lisette de Kreij-de Bruin
- Department of Pediatric Surgery, Erasmus Medical Center - Sophia Children's Hospital, Rotterdam, Netherlands
| | - Marco Schnater
- Department of Pediatric Surgery, Erasmus Medical Center - Sophia Children's Hospital, Rotterdam, Netherlands
| | - Dick Tibboel
- Department of Pediatric Surgery, Erasmus Medical Center - Sophia Children's Hospital, Rotterdam, Netherlands
| | - Jennifer Collins
- Department of Pediatric Surgery, Erasmus Medical Center - Sophia Children's Hospital, Rotterdam, Netherlands
| | - Robbert J Rottier
- Department of Pediatric Surgery, Erasmus Medical Center - Sophia Children's Hospital, Rotterdam, Netherlands.,Department of Cell biology, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
6
|
Lin J, Yuan Y, Shi X, Fang S, Zhang Y, Guan M, Xie Z, Ma H, Lin F. Molecular cloning, characterization and expression profiles of a SoxB2 gene related to gonadal development in mud crab (Scylla paramamosain). INVERTEBR REPROD DEV 2020. [DOI: 10.1080/07924259.2020.1726515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Jiali Lin
- Institute of Marine Sciences, Shantou University, Shantou, China
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
| | - Yuying Yuan
- Institute of Marine Sciences, Shantou University, Shantou, China
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
| | - Xi Shi
- Institute of Marine Sciences, Shantou University, Shantou, China
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
| | - Shaobin Fang
- Institute of Marine Sciences, Shantou University, Shantou, China
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
| | - Yin Zhang
- Institute of Marine Sciences, Shantou University, Shantou, China
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
| | - Mengyun Guan
- Institute of Marine Sciences, Shantou University, Shantou, China
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
| | - Zhuofang Xie
- Institute of Marine Sciences, Shantou University, Shantou, China
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
| | - Hongyu Ma
- Institute of Marine Sciences, Shantou University, Shantou, China
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
| | - Fan Lin
- Institute of Marine Sciences, Shantou University, Shantou, China
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
| |
Collapse
|
7
|
You D, Guo L, Li W, Sun S, Chen Y, Chai R, Li H. Characterization of Wnt and Notch-Responsive Lgr5+ Hair Cell Progenitors in the Striolar Region of the Neonatal Mouse Utricle. Front Mol Neurosci 2018; 11:137. [PMID: 29760650 PMCID: PMC5937014 DOI: 10.3389/fnmol.2018.00137] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/04/2018] [Indexed: 12/22/2022] Open
Abstract
Dysfunctions in hearing and balance are largely connected with hair cell (HC) loss. Although regeneration of HCs in the adult cochlea does not occur, there is still limited capacity for HC regeneration in the mammalian utricle from a distinct population of supporting cells (SCs). In response to HC damage, these Lgr5+ SCs, especially those in the striolar region, can regenerate HCs. In this study, we isolated Lgr5+ SCs and Plp1+ SCs (which originate from the striolar and extrastriolar regions, respectively) from transgenic mice by flow cytometry so as to compare the properties of these two subsets of SCs. We found that the Lgr5+ progenitors had greater proliferation and HC regeneration ability than the Plp1+ SCs and that the Lgr5+ progenitors responded more strongly to Wnt and Notch signaling than Plp1+ SCs. We then compared the gene expression profiles of the two populations by RNA-Seq and identified several genes that were significantly differentially expressed between the two populations, including genes involved in the cell cycle, transcription and cell signaling pathways. Targeting these genes and pathways might be a potential way to activate HC regeneration.
Collapse
Affiliation(s)
- Dan You
- ENT Institute and Otorhinolaryngology, Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Luo Guo
- ENT Institute and Otorhinolaryngology, Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Wenyan Li
- ENT Institute and Otorhinolaryngology, Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Shan Sun
- ENT Institute and Otorhinolaryngology, Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Yan Chen
- ENT Institute and Otorhinolaryngology, Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Institute of Life Sciences, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Huawei Li
- ENT Institute and Otorhinolaryngology, Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,Institutes of Biomedical Sciences and The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Anishchenko E, Arnone MI, D'Aniello S. SoxB2 in sea urchin development: implications in neurogenesis, ciliogenesis and skeletal patterning. EvoDevo 2018; 9:5. [PMID: 29479411 PMCID: PMC5817722 DOI: 10.1186/s13227-018-0094-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/01/2018] [Indexed: 11/21/2022] Open
Abstract
Background Current studies in evolutionary developmental biology are focused on the reconstruction of gene regulatory networks in target animal species. From decades, the scientific interest on genetic mechanisms orchestrating embryos development has been increasing in consequence to the fact that common features shared by evolutionarily distant phyla are being clarified. In 2011, a study across eumetazoan species showed for the first time the existence of a highly conserved non-coding element controlling the SoxB2 gene, which is involved in the early specification of the nervous system. This discovery raised several questions about SoxB2 function and regulation in deuterostomes from an evolutionary point of view. Results Due to the relevant phylogenetic position within deuterostomes, the sea urchin Strongylocentrotus purpuratus represents an advantageous animal model in the field of evolutionary developmental biology. Herein, we show a comprehensive study of SoxB2 functions in sea urchins, in particular its expression pattern in a wide range of developmental stages, and its co-localization with other neurogenic markers, as SoxB1, SoxC and Elav. Moreover, this work provides a detailed description of the phenotype of sea urchin SoxB2 knocked-down embryos, confirming its key function in neurogenesis and revealing, for the first time, its additional roles in oral and aboral ectoderm cilia and skeletal rod morphology. Conclusions We concluded that SoxB2 in sea urchins has a neurogenic function; however, this gene could have multiple roles in sea urchin embryogenesis, expanding its expression in non-neurogenic cells. We showed that SoxB2 is functionally conserved among deuterostomes and suggested that in S. purpuratus this gene acquired additional functions, being involved in ciliogenesis and skeletal patterning. Electronic supplementary material The online version of this article (10.1186/s13227-018-0094-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Evgeniya Anishchenko
- Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn Napoli, Villa Comunale, 80121 Naples, Italy
| | - Maria Ina Arnone
- Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn Napoli, Villa Comunale, 80121 Naples, Italy
| | - Salvatore D'Aniello
- Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn Napoli, Villa Comunale, 80121 Naples, Italy
| |
Collapse
|
9
|
Gálvez H, Abelló G, Giraldez F. Signaling and Transcription Factors during Inner Ear Development: The Generation of Hair Cells and Otic Neurons. Front Cell Dev Biol 2017; 5:21. [PMID: 28393066 PMCID: PMC5364141 DOI: 10.3389/fcell.2017.00021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 03/02/2017] [Indexed: 12/21/2022] Open
Abstract
Integration between cell signals and bHLH transcription factors plays a prominent role during the development of hair cells of the inner ear. Hair cells are the sensory receptors of the inner ear, responsible for the mechano-transduction of sound waves into electrical signals. They derive from multipotent progenitors that reside in the otic placode. Progenitor commitment is the result of cell signaling from the surrounding tissues that result in the restricted expression of SoxB1 transcription factors, Sox2 and Sox3. In turn, they induce the expression of Neurog1 and Atoh1, two bHLH factors that specify neuronal and hair cell fates, respectively. Neuronal and hair cell development, however, do not occur simultaneously. Hair cell development is prevented during neurogenesis and prosensory stages, resulting in the delay of hair cell development with respect to neuron production. Negative interactions between Neurog1 and Atoh1, and of Atoh1 with other bHLH factors driven by Notch signaling, like Hey1 and Hes5, account for this delay. In summary, the regulation of Atoh1 and hair cell development relies on interactions between cell signaling and bHLH transcription factors that dictate cell fate and timing decisions during development. Interestingly, these mechanisms operate as well during hair cell regeneration after damage and during stem cell directed differentiation, making developmental studies instrumental for improving therapies for hearing impairment.
Collapse
Affiliation(s)
- Héctor Gálvez
- Developmental Biology, CEXS, Parc de Recerca Biomèdica de Barcelona, Universitat Pompeu Fabra Barcelona, Spain
| | - Gina Abelló
- Developmental Biology, CEXS, Parc de Recerca Biomèdica de Barcelona, Universitat Pompeu Fabra Barcelona, Spain
| | - Fernando Giraldez
- Developmental Biology, CEXS, Parc de Recerca Biomèdica de Barcelona, Universitat Pompeu Fabra Barcelona, Spain
| |
Collapse
|
10
|
Liu ZQ, Jiang XH, Qi HY, Xiong LW, Qiu GF. A novel SoxB2 gene is required for maturation of sperm nucleus during spermiogenesis in the Chinese mitten crab, Eriocheir sinensis. Sci Rep 2016; 6:32139. [PMID: 27561408 PMCID: PMC4999818 DOI: 10.1038/srep32139] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/03/2016] [Indexed: 01/10/2023] Open
Abstract
SRY-related HMG box (Sox) genes are characterized by the presence of a DNA-binding HMG domain and involved in a diverse range of developmental processes. In this study, we identified a novel Sox gene, designated as EsSoxB2-1, from the Chinese mitten crab Eriocheir sinensis. The EsSoxB2-1 encodes a protein of 259 amino acids, sharing the highest identity with the beetle Tribolium castaneum SOX21b. Unlike insect Sox21b, however, EsSoxB2-1 is intronless and exhibits a gonad-specific expression pattern at both mRNA and protein level. Two core promoters in 5′ flanking region were demonstrated to be essential for inducing transcriptional regulatory activity. The transcription of EsSoxB2-1 mRNA begins in spermatogonia stage, while the translation of EsSOXB2-1 protein initiates at spermiogenesis stage. Interestingly, EsSOXB2-1 protein was exclusively localized in the nucleus of spermatid and spermatozoa even at the end of acrosome reaction, and was bound to the uncondensed chromatin in nucleoplasm of mature spermatozoa. Knockdown of EsSoxB2-1 by RNAi leads to abnormal transformation of the nucleus during spermiogenesis. Together, these findings demonstrated the requirement of EsSoxB2-1 for the spermatozoa nucleus maturation and also suggested that EsSoxB2-1 would be delivered into fertilized eggs along with chromatins as a paternal transcription factor for regulating early embryonic development.
Collapse
Affiliation(s)
- Zhi-Qiang Liu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources Certificated by Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, 999 Hucheng Huan Road, Shanghai, 201306, P. R. China
| | - Xue-Hui Jiang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources Certificated by Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, 999 Hucheng Huan Road, Shanghai, 201306, P. R. China
| | - Hai-Yan Qi
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources Certificated by Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, 999 Hucheng Huan Road, Shanghai, 201306, P. R. China
| | - Liang-Wei Xiong
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources Certificated by Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, 999 Hucheng Huan Road, Shanghai, 201306, P. R. China
| | - Gao-Feng Qiu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources Certificated by Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, 999 Hucheng Huan Road, Shanghai, 201306, P. R. China
| |
Collapse
|
11
|
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
|
12
|
Mulvaney JF, Amemiya Y, Freeman SD, Ladher RK, Dabdoub A. Molecular cloning and functional characterisation of chicken Atonal homologue 1: a comparison with human Atoh1. Biol Cell 2015; 107:41-60. [PMID: 25412697 DOI: 10.1111/boc.201400078] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/20/2014] [Indexed: 12/19/2022]
Abstract
BACKGROUND INFORMATION The vertebrate basic helix-loop-helix transcription factor Atoh1 is essential for maturation and survival of mechanosensory hair cells of the inner ear, neurogenesis, differentiation of the intestine, homeostasis of the colon and is implicated in cancer progression. Given that mutations in Atoh1 are detected in malignant tumours, study of functionally different Atoh1 alleles and homologues might yield useful avenues for investigation. The predicted sequence of chicken Atoh1 (cAtoh1) has large regions of dissimilarity to that of mammalian Atoh1 homologues. We hypothesise that cAtoh1 might have intrinsic functional differences to mammalian Atoh1. RESULTS In this study, we cloned and sequenced the full open reading frame of cAtoh1. In overexpression experiments, we show that this sequence is sufficient to generate a cAtoh1 protein capable of inducing hair cell markers when expressed in nonsensory regions of the developing inner ear, and that morpholino-mediated knock-down using a section of the sequence 5' to the start codon inhibits differentiation of hair cells in the chicken basilar papilla. Furthermore, we compare the behaviour of cAtoh1 and human Atoh1 (hAtoh1) in embryonic mouse cochlear explants, showing that cAtoh1 is a potent inducer of hair cell differentiation and that it can overcome Sox2-mediated repression of hair cell differentiation more effectively than hAtoh1. CONCLUSIONS cAtoh1 is both necessary and sufficient for avian mechanosensory hair cell differentiation. The non-conserved regions of the cAtoh1 coding region have functional consequences on its behaviour.
Collapse
Affiliation(s)
- Joanna F Mulvaney
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, M4N 3M5, Canada
| | | | | | | | | |
Collapse
|
13
|
Can the ‘neuron theory’ be complemented by a universal mechanism for generic neuronal differentiation. Cell Tissue Res 2014; 359:343-84. [DOI: 10.1007/s00441-014-2049-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 10/23/2014] [Indexed: 12/19/2022]
|
14
|
Savoy-Burke G, Gilels FA, Pan W, Pratt D, Que J, Gan L, White PM, Kiernan AE. Activated notch causes deafness by promoting a supporting cell phenotype in developing auditory hair cells. PLoS One 2014; 9:e108160. [PMID: 25264928 PMCID: PMC4180070 DOI: 10.1371/journal.pone.0108160] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 08/25/2014] [Indexed: 01/10/2023] Open
Abstract
Purpose To determine whether activated Notch can promote a supporting cell fate during sensory cell differentiation in the inner ear. Methods An activated form of the Notch1 receptor (NICD) was expressed in early differentiating hair cells using a Gfi1-Cre mouse allele. To determine the effects of activated Notch on developing hair cells, Gfi1-NICD animals and their littermate controls were assessed at 5 weeks for hearing by measuring auditory brainstem responses (ABRs) and distortion product otoacoustic emissions (DPOAEs). The differentiation of NICD-expressing hair cells was assessed at postnatal day (P) 6, 11 and 20, using histological and molecular markers for hair cells, as well as supporting cells/progenitor cells. We also examined whether the effects of Notch were mediated by SOX2, a gene expressed in supporting cells and a likely downstream target of Notch, by crossing an inducible form of SOX2 to the Gfi1-Cre. Results Activation of Notch1 in developing auditory hair cells causes profound deafness. The NICD-expressing hair cells switch off a number of hair cell markers and lose their characteristic morphology. Instead, NICD-expressing hair cells adopt a morphology resembling supporting cells and upregulate a number of supporting cell markers. These effects do not appear to be mediated by SOX2, because although expression of SOX2 caused some hearing impairment, the SOX2-expressing hair cells did not downregulate hair cell markers nor exhibit a supporting cell-like phenotype. Conclusions Our data show that Notch signaling inhibits hair cell differentiation and promotes a supporting cell-like phenotype, and that these effects are unlikely to be mediated by SOX2.
Collapse
Affiliation(s)
- Grace Savoy-Burke
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Felicia A. Gilels
- Department of Neurobiology and Anatomy, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Wei Pan
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York, United States of America
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Diana Pratt
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Jianwen Que
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Lin Gan
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York, United States of America
- Department of Neurobiology and Anatomy, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Patricia M. White
- Department of Neurobiology and Anatomy, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Amy E. Kiernan
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York, United States of America
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
15
|
Neves J, Vachkov I, Giraldez F. Sox2 regulation of hair cell development: incoherence makes sense. Hear Res 2013; 297:20-9. [DOI: 10.1016/j.heares.2012.11.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 10/17/2012] [Accepted: 11/05/2012] [Indexed: 01/09/2023]
|