1
|
Prakash A, Weninger J, Singh N, Raman S, Rao M, Kruse K, Ladher RK. Junctional force patterning drives both positional order and planar polarity in the auditory epithelia. Nat Commun 2025; 16:3927. [PMID: 40280944 PMCID: PMC12032022 DOI: 10.1038/s41467-025-58557-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 03/19/2025] [Indexed: 04/29/2025] Open
Abstract
Tissue function depends on the precise organisation of the constituent cells. In the cochlea, the fidelity of hearing depends on mechanosensory hair cells being consistently surrounded by supporting cells. In addition to this positional order, auditory sensitivity depends crucially on planar cell polarity. This is characterised by the alignment of the orientation of eccentrically placed hair bundles on each hair cell. These two levels of order emerge simultaneously despite the cellular fluxes that occur during cochlear development. However, the link between tissue-scale cellular rearrangements and intrinsic cellular mechanisms remains unknown. By combining experimental and theoretical approaches, we find a precise force patterning underpinning positional order and planar cell polarity. This occurs through the modulation of the levels and phospho-type of the regulatory light chain of non-muscle myosin II at specific cell-cell junctions of the auditory epithelium. We propose that the control of junctional mechanics is vital for the organisation of multi-cell-type epithelia.
Collapse
Affiliation(s)
- Anubhav Prakash
- National Centre for Biological Sciences, Tata Institute for Fundamentals Research, GKVK PO, Bangalore, India
| | - Julian Weninger
- Departments of Biochemistry and Theoretical Physics, University of Geneva, Geneva, Switzerland
| | - Nishant Singh
- National Centre for Biological Sciences, Tata Institute for Fundamentals Research, GKVK PO, Bangalore, India
- The University of Trans-Disciplinary Health Sciences and Technology, Yelahanka, Bangalore, India
| | - Sukanya Raman
- National Centre for Biological Sciences, Tata Institute for Fundamentals Research, GKVK PO, Bangalore, India
| | - Madan Rao
- National Centre for Biological Sciences, Tata Institute for Fundamentals Research, GKVK PO, Bangalore, India
| | - Karsten Kruse
- Departments of Biochemistry and Theoretical Physics, University of Geneva, Geneva, Switzerland.
| | - Raj K Ladher
- National Centre for Biological Sciences, Tata Institute for Fundamentals Research, GKVK PO, Bangalore, India.
| |
Collapse
|
2
|
Song Y, Jian S, Teng J, Zheng P, Zhang Z. Structural basis of human VANGL-PRICKLE interaction. Nat Commun 2025; 16:132. [PMID: 39753555 PMCID: PMC11698917 DOI: 10.1038/s41467-024-55396-3] [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: 06/21/2024] [Accepted: 11/15/2024] [Indexed: 01/06/2025] Open
Abstract
Planar cell polarity (PCP) is an evolutionarily conserved process for development and morphogenesis in metazoans. The well-organized polarity pattern in cells is established by the asymmetric distribution of two core protein complexes on opposite sides of the cell membrane. The Van Gogh-like (VANGL)-PRICKLE (PK) pair is one of these two key regulators; however, their structural information and detailed functions have been unclear. Here, we present five cryo-electron microscopy structures of human VANGL1, VANGL2, and their complexes with PK1 at resolutions of 2.2-3.0 Å. Through biochemical and cell imaging experiments, we decipher the molecular details of the VANGL-PK interaction. Furthermore, we reveal that PK1 can target VANGL-containing intracellular vesicles to the peripheral cell membrane. These findings provide a solid foundation to understand the explicit interaction between VANGL and PK while opening new avenues for subsequent studies of the PCP pathway.
Collapse
Affiliation(s)
- Yanyi Song
- State Key Laboratory of Membrane Biology, Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Shuyi Jian
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Junlin Teng
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Pengli Zheng
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China.
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| | - Zhe Zhang
- State Key Laboratory of Membrane Biology, Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
- School of Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
3
|
Xie N, Landin Malt A, Adylkhan A, Rodeman N, Moraes Borges R, Hwang D, Liu A, Smith C, Hogan A, Lu X. Wnt7b acts in concert with Wnt5a to regulate tissue elongation and planar cell polarity via noncanonical Wnt signaling. Proc Natl Acad Sci U S A 2024; 121:e2405217121. [PMID: 39172791 PMCID: PMC11363310 DOI: 10.1073/pnas.2405217121] [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: 03/12/2024] [Accepted: 07/24/2024] [Indexed: 08/24/2024] Open
Abstract
Intercellular signaling mediated by evolutionarily conserved planar cell polarity (PCP) proteins aligns cell polarity along the tissue plane and drives polarized cell behaviors during tissue morphogenesis. Accumulating evidence indicates that the vertebrate PCP pathway is regulated by noncanonical, β-catenin-independent Wnt signaling; however, the signaling components and mechanisms are incompletely understood. In the mouse hearing organ, both PCP and noncanonical Wnt (ncWnt) signaling are required in the developing auditory sensory epithelium to control cochlear duct elongation and planar polarity of resident sensory hair cells (HCs), including the shape and orientation of the stereociliary hair bundle essential for sound detection. We have recently discovered a Wnt/G-protein/PI3K pathway that coordinates HC planar polarity and intercellular PCP signaling. Here, we identify Wnt7b as a ncWnt ligand acting in concert with Wnt5a to promote tissue elongation in diverse developmental processes. In the cochlea, Wnt5a and Wnt7b are redundantly required for cochlear duct coiling and elongation, HC planar polarity, and asymmetric localization of core PCP proteins Fzd6 and Dvl2. Mechanistically, Wnt5a/Wnt7b-mediated ncWnt signaling promotes membrane recruitment of Daple, a nonreceptor guanine nucleotide exchange factor for Gαi, and activates PI3K/AKT and ERK signaling, which promote asymmetric Fzd6 localization. Thus, ncWnt and PCP signaling pathways have distinct mutant phenotypes and signaling components, suggesting that they act as separate, parallel pathways with nonoverlapping functions in cochlear morphogenesis. NcWnt signaling drives tissue elongation and reinforces intercellular PCP signaling by regulating the trafficking of PCP-specific Frizzled receptors.
Collapse
Affiliation(s)
- Nicholas Xie
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Andre Landin Malt
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Aray Adylkhan
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Natalie Rodeman
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Ricardo Moraes Borges
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Diane Hwang
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Alice Liu
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Connor Smith
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Arielle Hogan
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Xiaowei Lu
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| |
Collapse
|
4
|
Kacker S, Parsad V, Singh N, Hordiichuk D, Alvarez S, Gohar M, Kacker A, Rai SK. Planar Cell Polarity Signaling: Coordinated Crosstalk for Cell Orientation. J Dev Biol 2024; 12:12. [PMID: 38804432 PMCID: PMC11130840 DOI: 10.3390/jdb12020012] [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: 01/28/2024] [Revised: 04/08/2024] [Accepted: 04/13/2024] [Indexed: 05/29/2024] Open
Abstract
The planar cell polarity (PCP) system is essential for positioning cells in 3D networks to establish the proper morphogenesis, structure, and function of organs during embryonic development. The PCP system uses inter- and intracellular feedback interactions between components of the core PCP, characterized by coordinated planar polarization and asymmetric distribution of cell populations inside the cells. PCP signaling connects the anterior-posterior to left-right embryonic plane polarity through the polarization of cilia in the Kupffer's vesicle/node in vertebrates. Experimental investigations on various genetic ablation-based models demonstrated the functions of PCP in planar polarization and associated genetic disorders. This review paper aims to provide a comprehensive overview of PCP signaling history, core components of the PCP signaling pathway, molecular mechanisms underlying PCP signaling, interactions with other signaling pathways, and the role of PCP in organ and embryonic development. Moreover, we will delve into the negative feedback regulation of PCP to maintain polarity, human genetic disorders associated with PCP defects, as well as challenges associated with PCP.
Collapse
Affiliation(s)
- Sandeep Kacker
- Department of Pharmacology, Medical University of the Americas, Charlestown KN 1102, Saint Kitts and Nevis;
| | - Varuneshwar Parsad
- Department of Human Body Structure and Function, Medical University of the Americas, Charlestown KN 1102, Saint Kitts and Nevis; (V.P.); (D.H.)
| | - Naveen Singh
- Department of Cerll and Molecular Biology, Medical University of the Americas, Charlestown KN 1102, Saint Kitts and Nevis; (N.S.); (S.A.); (M.G.)
| | - Daria Hordiichuk
- Department of Human Body Structure and Function, Medical University of the Americas, Charlestown KN 1102, Saint Kitts and Nevis; (V.P.); (D.H.)
| | - Stacy Alvarez
- Department of Cerll and Molecular Biology, Medical University of the Americas, Charlestown KN 1102, Saint Kitts and Nevis; (N.S.); (S.A.); (M.G.)
| | - Mahnoor Gohar
- Department of Cerll and Molecular Biology, Medical University of the Americas, Charlestown KN 1102, Saint Kitts and Nevis; (N.S.); (S.A.); (M.G.)
| | - Anshu Kacker
- Department of Histology and Human Physiology, Medical University of the Americas, Charlestown KN 1102, Saint Kitts and Nevis;
| | - Sunil Kumar Rai
- Department of Cerll and Molecular Biology, Medical University of the Americas, Charlestown KN 1102, Saint Kitts and Nevis; (N.S.); (S.A.); (M.G.)
| |
Collapse
|
5
|
Ebeid M, Kishimoto I, Roy P, Zaidi MAA, Cheng AG, Huh SH. β-Catenin transcriptional activity is required for establishment of inner pillar cell identity during cochlear development. PLoS Genet 2023; 19:e1010925. [PMID: 37639482 PMCID: PMC10491406 DOI: 10.1371/journal.pgen.1010925] [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: 10/14/2022] [Revised: 09/08/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023] Open
Abstract
The mammalian cochlea is composed of sensory hair cells as well as multiple different types of non-sensory supporting cells. Pillar cells are one type of supporting cell that form the tunnel of Corti and include two morphologically and functionally distinct subtypes: inner pillar cells (IPCs) and outer pillar cells (OPCs). The processes of specification and differentiation of inner versus outer pillar cells are still unclear. Here, we show that β-Catenin is required for establishing IPC identity in the mammalian cochlea. To differentiate the transcriptional and adhesion roles of β-Catenin in establishing IPC identity, we examined two different models of β-Catenin deletion; one that deletes both transcriptional and structural functions and one which retains cell adhesion function but lacks transcriptional function. Here, we show that cochleae lacking β-Catenin transcriptional function lost IPCs and displayed extranumerary OPCs, indicating its requirement for establishing IPC identity. Overexpression of β-Catenin induced proliferation within IPCs but not ectopic IPCs. Single-cell transcriptomes of supporting cells lacking β-Catenin transcriptional function show a loss of the IPC and gain of OPC signatures. Finally, targeted deletion of β-Catenin in IPCs also led to the loss of IPC identity, indicating a cell autonomous role of β-Catenin in establishing IPC identity. As IPCs have the capacity to regenerate sensory hair cells in the postnatal cochlea, our results will aid in future IPC-based hair cell regeneration strategies.
Collapse
Affiliation(s)
- Michael Ebeid
- Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Ippei Kishimoto
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Pooja Roy
- Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Mohd Ali Abbas Zaidi
- Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Alan G. Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Sung-Ho Huh
- Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Otolaryngology-Head and Neck Surgery, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| |
Collapse
|
6
|
Lee MP, Waldhaus J. In vitro and in vivo models: What have we learnt about inner ear regeneration and treatment for hearing loss? Mol Cell Neurosci 2022; 120:103736. [PMID: 35577314 PMCID: PMC9551661 DOI: 10.1016/j.mcn.2022.103736] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 04/29/2022] [Accepted: 05/10/2022] [Indexed: 01/07/2023] Open
Abstract
The sensory cells of the inner ear, called hair cells, do not regenerate spontaneously and therefore, hair cell loss and subsequent hearing loss are permanent in humans. Conversely, functional hair cell regeneration can be observed in non-mammalian vertebrate species like birds and fish. Also, during postnatal development in mice, limited regenerative capacity and the potential to isolate stem cells were reported. Together, these findings spurred the interest of current research aiming to investigate the endogenous regenerative potential in mammals. In this review, we summarize current in vitro based approaches and briefly introduce different in vivo model organisms utilized to study hair cell regeneration. Furthermore, we present an overview of the findings that were made synergistically using both, the in vitro and in vivo based tools.
Collapse
Affiliation(s)
- Mary P Lee
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Joerg Waldhaus
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
7
|
Signaling pathways and targeted therapy for myocardial infarction. Signal Transduct Target Ther 2022; 7:78. [PMID: 35273164 PMCID: PMC8913803 DOI: 10.1038/s41392-022-00925-z] [Citation(s) in RCA: 374] [Impact Index Per Article: 124.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/28/2022] [Accepted: 02/08/2022] [Indexed: 02/07/2023] Open
Abstract
Although the treatment of myocardial infarction (MI) has improved considerably, it is still a worldwide disease with high morbidity and high mortality. Whilst there is still a long way to go for discovering ideal treatments, therapeutic strategies committed to cardioprotection and cardiac repair following cardiac ischemia are emerging. Evidence of pathological characteristics in MI illustrates cell signaling pathways that participate in the survival, proliferation, apoptosis, autophagy of cardiomyocytes, endothelial cells, fibroblasts, monocytes, and stem cells. These signaling pathways include the key players in inflammation response, e.g., NLRP3/caspase-1 and TLR4/MyD88/NF-κB; the crucial mediators in oxidative stress and apoptosis, for instance, Notch, Hippo/YAP, RhoA/ROCK, Nrf2/HO-1, and Sonic hedgehog; the controller of myocardial fibrosis such as TGF-β/SMADs and Wnt/β-catenin; and the main regulator of angiogenesis, PI3K/Akt, MAPK, JAK/STAT, Sonic hedgehog, etc. Since signaling pathways play an important role in administering the process of MI, aiming at targeting these aberrant signaling pathways and improving the pathological manifestations in MI is indispensable and promising. Hence, drug therapy, gene therapy, protein therapy, cell therapy, and exosome therapy have been emerging and are known as novel therapies. In this review, we summarize the therapeutic strategies for MI by regulating these associated pathways, which contribute to inhibiting cardiomyocytes death, attenuating inflammation, enhancing angiogenesis, etc. so as to repair and re-functionalize damaged hearts.
Collapse
|
8
|
Deans MR. Conserved and Divergent Principles of Planar Polarity Revealed by Hair Cell Development and Function. Front Neurosci 2021; 15:742391. [PMID: 34733133 PMCID: PMC8558554 DOI: 10.3389/fnins.2021.742391] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Planar polarity describes the organization and orientation of polarized cells or cellular structures within the plane of an epithelium. The sensory receptor hair cells of the vertebrate inner ear have been recognized as a preeminent vertebrate model system for studying planar polarity and its development. This is principally because planar polarity in the inner ear is structurally and molecularly apparent and therefore easy to visualize. Inner ear planar polarity is also functionally significant because hair cells are mechanosensors stimulated by sound or motion and planar polarity underlies the mechanosensory mechanism, thereby facilitating the auditory and vestibular functions of the ear. Structurally, hair cell planar polarity is evident in the organization of a polarized bundle of actin-based protrusions from the apical surface called stereocilia that is necessary for mechanosensation and when stereociliary bundle is disrupted auditory and vestibular behavioral deficits emerge. Hair cells are distributed between six sensory epithelia within the inner ear that have evolved unique patterns of planar polarity that facilitate auditory or vestibular function. Thus, specialized adaptations of planar polarity have occurred that distinguish auditory and vestibular hair cells and will be described throughout this review. There are also three levels of planar polarity organization that can be visualized within the vertebrate inner ear. These are the intrinsic polarity of individual hair cells, the planar cell polarity or coordinated orientation of cells within the epithelia, and planar bipolarity; an organization unique to a subset of vestibular hair cells in which the stereociliary bundles are oriented in opposite directions but remain aligned along a common polarity axis. The inner ear with its complement of auditory and vestibular sensory epithelia allows these levels, and the inter-relationships between them, to be studied using a single model organism. The purpose of this review is to introduce the functional significance of planar polarity in the auditory and vestibular systems and our contemporary understanding of the developmental mechanisms associated with organizing planar polarity at these three cellular levels.
Collapse
Affiliation(s)
- Michael R Deans
- Department of Surgery, Division of Otolaryngology, University of Utah School of Medicine, Salt Lake City, UT, United States.,Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
9
|
Tarchini B. A Reversal in Hair Cell Orientation Organizes Both the Auditory and Vestibular Organs. Front Neurosci 2021; 15:695914. [PMID: 34646115 PMCID: PMC8502876 DOI: 10.3389/fnins.2021.695914] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 09/03/2021] [Indexed: 01/17/2023] Open
Abstract
Sensory hair cells detect mechanical stimuli with their hair bundle, an asymmetrical brush of actin-based membrane protrusions, or stereocilia. At the single cell level, stereocilia are organized in rows of graded heights that confer the hair bundle with intrinsic directional sensitivity. At the organ level, each hair cell is precisely oriented so that its intrinsic directional sensitivity matches the direction of mechanical stimuli reaching the sensory epithelium. Coordinated orientation among neighboring hair cells usually ensures the delivery of a coherent local group response. Accordingly, hair cell orientation is locally uniform in the auditory and vestibular cristae epithelia in birds and mammals. However, an exception to this rule is found in the vestibular macular organs, and in fish lateral line neuromasts, where two hair cell populations show opposing orientations. This mirror-image hair cell organization confers bidirectional sensitivity at the organ level. Here I review our current understanding of the molecular machinery that produces mirror-image organization through a regional reversal of hair cell orientation. Interestingly, recent evidence suggests that auditory hair cells adopt their normal uniform orientation through a global reversal mechanism similar to the one at work regionally in macular and neuromast organs. Macular and auditory organs thus appear to be patterned more similarly than previously appreciated during inner ear development.
Collapse
Affiliation(s)
- Basile Tarchini
- The Jackson Laboratory, Bar Harbor, ME, United States.,Department of Medicine, Tufts University, Boston, MA, United States.,Graduate School of Biomedical Science and Engineering (GSBSE), University of Maine, Orono, ME, United States
| |
Collapse
|
10
|
Oliver BL, Young CA, Munnamalai V. Spatial and temporal expression of PORCN is highly dynamic in the developing mouse cochlea. Gene Expr Patterns 2021; 42:119214. [PMID: 34547456 DOI: 10.1016/j.gep.2021.119214] [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: 08/04/2021] [Revised: 09/08/2021] [Accepted: 09/17/2021] [Indexed: 10/20/2022]
Abstract
The mammalian organ of Corti is a highly specialized sensory organ of the cochlea with a fine-grained pattern that is essential for auditory function. The sensory epithelium, the organ of Corti consists of a single row of inner hair cells and three rows of outer hair cells that are intercalated by support cells in a mosaic pattern. Previous studies show that the Wnt pathway regulates proliferation, promotes medial compartment formation in the cochlea, differentiation of the mechanosensory hair cells and axon guidance of Type II afferent neurons. WNT ligand expressions are highly dynamic throughout development but are insufficient to explain the roles of the Wnt pathway. We address a potential way for how WNTs specify the medial compartment by characterizing the expression of Porcupine (PORCN), an O-acyltransferase that is required for WNT secretion. We show PORCN expression across embryonic ages (E)12.5 - E14.5, E16.5, and postnatal day (P)1. Our results showed enriched PORCN in the medial domains during early stages of development, indicating that WNTs have a stronger influence on patterning of the medial compartment. PORCN was rapidly downregulated after E14.5, following the onset of sensory cell differentiation; residual expression remained in some hair cells and supporting cells. On E14.5 and E16.5, we also examined the spatial expression of Gsk3β, an inhibitor of canonical Wnt signaling to determine its potential role in radial patterning of the cochlea. Gsk3β was broadly expressed across the radial axis of the epithelium; therefore, unlikely to control WNT-mediated medial specification. In conclusion, the spatial expression of PORCN enriches WNT secretion from the medial domains of the cochlea to influence the specification of cell fates in the medial sensory domain.
Collapse
Affiliation(s)
| | - Caryl A Young
- The Jackson Laboratory, Bar Harbor, ME, 04609, United States; The University of Maine, Graduate School of Biomedical Sciences and Engineering, Orono, ME, 04469, United States
| | - Vidhya Munnamalai
- The Jackson Laboratory, Bar Harbor, ME, 04609, United States; The University of Maine, Graduate School of Biomedical Sciences and Engineering, Orono, ME, 04469, United States.
| |
Collapse
|
11
|
Canonical Wnt Signaling Pathway on Polarity Formation of Utricle Hair Cells. Neural Plast 2021; 2021:9950533. [PMID: 34122536 PMCID: PMC8166501 DOI: 10.1155/2021/9950533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/26/2021] [Accepted: 05/11/2021] [Indexed: 02/05/2023] Open
Abstract
As part of the inner ear, the vestibular system is responsible for sense of balance, which consists of three semicircular canals, the utricle, and the saccule. Increasing evidence has indicated that the noncanonical Wnt/PCP signaling pathway plays a significant role in the development of the polarity of the inner ear. However, the role of canonical Wnt signaling in the polarity of the vestibule is still not completely clear. In this study, we found that canonical Wnt pathway-related genes are expressed in the early stage of development of the utricle and change dynamically. We conditionally knocked out β-catenin, a canonical Wnt signaling core protein, and found that the cilia orientation of hair cells was disordered with reduced number of hair cells in the utricle. Moreover, regulating the canonical Wnt pathway (Licl and IWP2) in vitro also affected hair cell polarity and indicated that Axin2 may be important in this process. In conclusion, our results not only confirm that the regulation of canonical Wnt signaling affects the number of hair cells in the utricle but also provide evidence for its role in polarity development.
Collapse
|
12
|
Warnecke A, Giesemann A. Embryology, Malformations, and Rare Diseases of the Cochlea. Laryngorhinootologie 2021; 100:S1-S43. [PMID: 34352899 PMCID: PMC8354575 DOI: 10.1055/a-1349-3824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Despite the low overall prevalence of individual rare diseases, cochlear
dysfunction leading to hearing loss represents a symptom in a large
proportion. The aim of this work was to provide a clear overview of rare
cochlear diseases, taking into account the embryonic development of the
cochlea and the systematic presentation of the different disorders. Although
rapid biotechnological and bioinformatic advances may facilitate the
diagnosis of a rare disease, an interdisciplinary exchange is often required
to raise the suspicion of a rare disease. It is important to recognize that
the phenotype of rare inner ear diseases can vary greatly not only in
non-syndromic but also in syndromic hearing disorders. Finally, it becomes
clear that the phenotype of the individual rare diseases cannot be
determined exclusively by classical genetics even in monogenetic
disorders.
Collapse
Affiliation(s)
- Athanasia Warnecke
- Klinik für Hals-, Nasen- und Ohrenheilkunde, Medizinische Hochschule Hannover, Carl-Neuberg-Straße 1, 30625 Hannover.,Deutsche Forschungsgemeinschaft Exzellenzcluster"Hearing4all" - EXC 2177/1 - Project ID 390895286
| | - Anja Giesemann
- Institut für Neuroradiologie, Medizinische Hochschule Hannover, Carl-Neuberg-Straße 1, 30625 Hannover
| |
Collapse
|
13
|
Stabenau KA, Zimmermann MT, Mathison A, Zeighami A, Samuels TL, Chun RH, Papsin BC, McCormick ME, Johnston N, Kerschner JE. RNA Sequencing and Pathways Analyses of Middle Ear Epithelia From Patients With Otitis Media. Laryngoscope 2021; 131:2590-2597. [PMID: 33844317 DOI: 10.1002/lary.29551] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/04/2021] [Accepted: 03/23/2021] [Indexed: 01/24/2023]
Abstract
OBJECTIVES Otitis media (OM) is the most common pediatric diagnosis in the United States. However, our understanding of the molecular pathogenesis of OM remains relatively poor. Investigation of molecular pathways involved in OM may improve the understanding of this disease process and elucidate novel therapeutic targets. In this study, RNA sequencing (RNA-Seq) was used to discern cellular changes associated with OME compared to healthy middle ear epithelium (MEE). STUDY DESIGN Ex vivo case-control translational. METHODS Middle ear epithelia was collected from five pediatric patients diagnosed with OME undergoing tympanostomy tube placement and five otherwise healthy pediatric patients undergoing cochlear implantation. Specimens underwent RNA-Seq and pathways analyses. RESULTS A total of 1,292 genes exhibited differential expression in MEE from OME patients compared to controls including genes involved in inflammation, immune response to bacterial OM pathogens, mucociliary clearance, regulation of proliferation and transformation, and auditory cell differentiation. Top networks identified in OME were organismal injury and abnormalities, cell morphology, and auditory disease. Top Ingenuity canonical pathways identified were axonal guidance signaling, which contains genes associated with auditory development and disease and nicotine degradation II and III pathways. Associated upstream regulators included β-estradiol, dexamethasone, and G-protein-coupled estrogen receptor-1 (GPER1), which are associated with otoprotection or inflammation during insult. CONCLUSIONS RNA-Seq demonstrates differential gene expression in MEE from patients with OME compared to healthy controls with important implications for infection susceptibility, hearing loss, and a role for tobacco exposure in the development and/or severity of OME in pediatric patients. LEVEL OF EVIDENCE 4 Laryngoscope, 2021.
Collapse
Affiliation(s)
- Kaleigh A Stabenau
- Department of Otolaryngology and Communication Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, U.S.A
| | - Michael T Zimmermann
- Bioinformatics Research and Development Laboratory, Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin, U.S.A.,Clinical and Translational Science Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, U.S.A
| | - Angela Mathison
- Bioinformatics Research and Development Laboratory, Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin, U.S.A
| | - Atefeh Zeighami
- Bioinformatics Research and Development Laboratory, Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin, U.S.A
| | - Tina L Samuels
- Department of Otolaryngology and Communication Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, U.S.A
| | - Robert H Chun
- Department of Otolaryngology and Communication Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, U.S.A
| | - Blake C Papsin
- Archie's Cochlear Implant Laboratory, Department of Otolaryngology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael E McCormick
- Department of Otolaryngology and Communication Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, U.S.A
| | - Nikki Johnston
- Department of Otolaryngology and Communication Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, U.S.A.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, U.S.A
| | - Joseph E Kerschner
- Department of Otolaryngology and Communication Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, U.S.A
| |
Collapse
|
14
|
Reynolds K, Zhang S, Sun B, Garland M, Ji Y, Zhou CJ. Genetics and signaling mechanisms of orofacial clefts. Birth Defects Res 2020; 112:1588-1634. [PMID: 32666711 PMCID: PMC7883771 DOI: 10.1002/bdr2.1754] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/31/2022]
Abstract
Craniofacial development involves several complex tissue movements including several fusion processes to form the frontonasal and maxillary structures, including the upper lip and palate. Each of these movements are controlled by many different factors that are tightly regulated by several integral morphogenetic signaling pathways. Subject to both genetic and environmental influences, interruption at nearly any stage can disrupt lip, nasal, or palate fusion and result in a cleft. Here, we discuss many of the genetic risk factors that may contribute to the presentation of orofacial clefts in patients, and several of the key signaling pathways and underlying cellular mechanisms that control lip and palate formation, as identified primarily through investigating equivalent processes in animal models, are examined.
Collapse
Affiliation(s)
- Kurt Reynolds
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) graduate group, University of California, Davis, CA 95616
| | - Shuwen Zhang
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
| | - Bo Sun
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
| | - Michael Garland
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
| | - Yu Ji
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) graduate group, University of California, Davis, CA 95616
| | - Chengji J. Zhou
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) graduate group, University of California, Davis, CA 95616
| |
Collapse
|
15
|
Najarro EH, Huang J, Jacobo A, Quiruz LA, Grillet N, Cheng AG. Dual regulation of planar polarization by secreted Wnts and Vangl2 in the developing mouse cochlea. Development 2020; 147:dev.191981. [PMID: 32907846 DOI: 10.1242/dev.191981] [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: 04/22/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022]
Abstract
Planar cell polarity (PCP) proteins localize asymmetrically to instruct cell polarity within the tissue plane, with defects leading to deformities of the limbs, neural tube and inner ear. Wnt proteins are evolutionarily conserved polarity cues, yet Wnt mutants display variable PCP defects; thus, how Wnts regulate PCP remains unresolved. Here, we have used the developing cochlea as a model system to show that secreted Wnts regulate PCP through polarizing a specific subset of PCP proteins. Conditional deletion of Wntless or porcupine, both of which are essential for secretion of Wnts, caused misrotated sensory cells and shortened cochlea - both hallmarks of PCP defects. Wntless-deficient cochleae lacked the polarized PCP components dishevelled 1/2 and frizzled 3/6, while other PCP proteins (Vangl1/2, Celsr1 and dishevelled 3) remained localized. We identified seven Wnt paralogues, including the major PCP regulator Wnt5a, which was, surprisingly, dispensable for planar polarization in the cochlea. Finally, Vangl2 haploinsufficiency markedly accentuated sensory cell polarization defects in Wntless-deficient cochlea. Together, our study indicates that secreted Wnts and Vangl2 coordinate to ensure proper tissue polarization during development.
Collapse
Affiliation(s)
- Elvis Huarcaya Najarro
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jennifer Huang
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Adrian Jacobo
- Laboratory of Sensory Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | - Lee A Quiruz
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nicolas Grillet
- 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
|
16
|
Tona Y, Wu DK. Live imaging of hair bundle polarity acquisition demonstrates a critical timeline for transcription factor Emx2. eLife 2020; 9:e59282. [PMID: 32965215 PMCID: PMC7535933 DOI: 10.7554/elife.59282] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/17/2020] [Indexed: 12/22/2022] Open
Abstract
Directional sensitivity of hair cells (HCs) is conferred by the aymmetric apical hair bundle, comprised of a kinocilium and stereocilia staircase. The mother centriole (MC) forms the base of the kinocilium and the stereocilia develop adjacent to it. Previously, we showed that transcription factor Emx2 reverses hair bundle orientation and its expression in the mouse vestibular utricle is restricted, resulting in two regions of opposite bundle orientation (Jiang et al., 2017). Here, we investigated establishment of opposite bundle orientation in embryonic utricles by live-imaging GFP-labeled centrioles in HCs. The daughter centriole invariably migrated ahead of the MC from the center to their respective peripheral locations in HCs. Comparing HCs between utricular regions, centriole trajectories were similar but they migrated toward opposite directions, suggesting that Emx2 pre-patterned HCs prior to centriole migration. Ectopic Emx2, however, reversed centriole trajectory within hours during a critical time-window when centriole trajectory was responsive to Emx2.
Collapse
Affiliation(s)
- Yosuke Tona
- National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Doris K Wu
- National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| |
Collapse
|
17
|
Munnamalai V, Fekete DM. The acquisition of positional information across the radial axis of the cochlea. Dev Dyn 2019; 249:281-297. [PMID: 31566832 DOI: 10.1002/dvdy.118] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 09/10/2019] [Accepted: 09/10/2019] [Indexed: 12/11/2022] Open
Affiliation(s)
- Vidhya Munnamalai
- The Jackson Laboratory Bar Harbor Maine
- Graduate Program of Biomedical Sciences and EngineeringUniversity of Maine Orono Maine
- The Neuroscience ProgramSackler School of Biomedical Sciences, Tufts University Boston Massachusetts
| | - Donna M. Fekete
- Department of Biological SciencesPurdue University West Lafayette Indiana
- Purdue Institute for Integrative Neuroscience West Lafayette Indiana
- Purdue Center for Cancer Research West Lafayette Indiana
| |
Collapse
|
18
|
Tarchini B, Lu X. New insights into regulation and function of planar polarity in the inner ear. Neurosci Lett 2019; 709:134373. [PMID: 31295539 PMCID: PMC6732021 DOI: 10.1016/j.neulet.2019.134373] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/02/2019] [Accepted: 07/06/2019] [Indexed: 12/18/2022]
Abstract
Acquisition of cell polarity generates signaling and cytoskeletal asymmetry and thus underpins polarized cell behaviors during tissue morphogenesis. In epithelial tissues, both apical-basal polarity and planar polarity, which refers to cell polarization along an axis orthogonal to the apical-basal axis, are essential for epithelial morphogenesis and function. A prime example of epithelial planar polarity can be found in the auditory sensory epithelium (or organ of Corti, OC). Sensory hair cells, the sound receptors, acquire a planar polarized apical cytoskeleton which is uniformely oriented along an axis orthogonal to the longitudinal axis of the cochlear duct. Both cell-intrinsic and tissue-level planar polarity are necessary for proper perception of sound. Here we review recent insights into the novel roles and mechanisms of planar polarity signaling gained from genetic analysis in mice, focusing mainly on the OC but also with some discussions on the vestibular sensory epithelia.
Collapse
Affiliation(s)
- Basile Tarchini
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA; Department of Medicine, Tufts University, Boston, 02111, MA, USA; Graduate School of Biomedical Science and Engineering (GSBSE), University of Maine, Orono, 04469, ME, USA.
| | - Xiaowei Lu
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, 22908, USA.
| |
Collapse
|
19
|
PCP and Wnt pathway components act in parallel during zebrafish mechanosensory hair cell orientation. Nat Commun 2019; 10:3993. [PMID: 31488837 PMCID: PMC6728366 DOI: 10.1038/s41467-019-12005-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 08/05/2019] [Indexed: 12/03/2022] Open
Abstract
Planar cell polarity (PCP) plays crucial roles in developmental processes such as gastrulation, neural tube closure and hearing. Wnt pathway mutants are often classified as PCP mutants due to similarities between their phenotypes. Here, we show that in the zebrafish lateral line, disruptions of the PCP and Wnt pathways have differential effects on hair cell orientations. While mutations in the PCP genes vangl2 and scrib cause random orientations of hair cells, mutations in wnt11f1, gpc4 and fzd7a/b induce hair cells to adopt a concentric pattern. This concentric pattern is not caused by defects in PCP but is due to misaligned support cells. The molecular basis of the support cell defect is unknown but we demonstrate that the PCP and Wnt pathways work in parallel to establish proper hair cell orientation. Consequently, hair cell orientation defects are not solely explained by defects in PCP signaling, and some hair cell phenotypes warrant re-evaluation. Planar cell polarity (PCP) regulates hair cell orientation in the zebrafish lateral line. Here, the authors show that mutating Wnt pathway genes (wnt11f1, fzd7a/b, and gpc4) causes concentric hair cell patterns not regulated by PCP, thus showing PCP/Wnt pathway genes have different consequences on hair cell orientation.
Collapse
|
20
|
Frizzled3 and Frizzled6 Cooperate with Vangl2 to Direct Cochlear Innervation by Type II Spiral Ganglion Neurons. J Neurosci 2019; 39:8013-8023. [PMID: 31462532 DOI: 10.1523/jneurosci.1740-19.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 08/20/2019] [Accepted: 08/23/2019] [Indexed: 11/21/2022] Open
Abstract
Type II spiral ganglion neurons provide afferent innervation to outer hair cells of the cochlea and are proposed to have nociceptive functions important for auditory function and homeostasis. These neurons are anatomically distinct from other classes of spiral ganglion neurons because they extend a peripheral axon beyond the inner hair cells that subsequently makes a distinct 90 degree turn toward the cochlear base. As a result, patterns of outer hair cell innervation are coordinated with the tonotopic organization of the cochlea. Previously, it was shown that peripheral axon turning is directed by a nonautonomous function of the core planar cell polarity (PCP) protein VANGL2. We demonstrate using mice of either sex that Fzd3 and Fzd6 similarly regulate axon turning, are functionally redundant with each other, and that Fzd3 genetically interacts with Vangl2 to guide this process. FZD3 and FZD6 proteins are asymmetrically distributed along the basolateral wall of cochlear-supporting cells, and are required to promote or maintain the asymmetric distribution of VANGL2 and CELSR1. These data indicate that intact PCP complexes formed between cochlear-supporting cells are required for the nonautonomous regulation of axon pathfinding. Consistent with this, in the absence of PCP signaling, peripheral axons turn randomly and often project toward the cochlear apex. Additional analyses of Porcn mutants in which WNT secretion is reduced suggest that noncanonical WNT signaling establishes or maintains PCP signaling in this context. A deeper understanding of these mechanisms is necessary for repairing auditory circuits following acoustic trauma or promoting cochlear reinnervation during regeneration-based deafness therapies.SIGNIFICANCE STATEMENT Planar cell polarity (PCP) signaling has emerged as a complementary mechanism to classical axon guidance in regulating axon track formation, axon outgrowth, and neuronal polarization. The core PCP proteins are also required for auditory circuit assembly, and coordinate hair cell innervation with the tonotopic organization of the cochlea. This is a non-cell-autonomous mechanism that requires the formation of PCP protein complexes between cochlear-supporting cells located along the trajectory of growth cone navigation. These findings are significant because they demonstrate how the fidelity of auditory circuit formation is ensured during development, and provide a mechanism by which PCP proteins may regulate axon outgrowth and guidance in the CNS.
Collapse
|
21
|
Bademci G, Abad C, Incesulu A, Elian F, Reyahi A, Diaz-Horta O, Cengiz FB, Sineni CJ, Seyhan S, Atli EI, Basmak H, Demir S, Nik AM, Footz T, Guo S, Duman D, Fitoz S, Gurkan H, Blanton SH, Walter MA, Carlsson P, Walz K, Tekin M. FOXF2 is required for cochlear development in humans and mice. Hum Mol Genet 2019; 28:1286-1297. [PMID: 30561639 DOI: 10.1093/hmg/ddy431] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 11/14/2022] Open
Abstract
Molecular mechanisms governing the development of the human cochlea remain largely unknown. Through genome sequencing, we identified a homozygous FOXF2 variant c.325A>T (p.I109F) in a child with profound sensorineural hearing loss (SNHL) associated with incomplete partition type I anomaly of the cochlea. This variant is not found in public databases or in over 1000 ethnicity-matched control individuals. I109 is a highly conserved residue in the forkhead box (Fox) domain of FOXF2, a member of the Fox protein family of transcription factors that regulate the expression of genes involved in embryogenic development as well as adult life. Our in vitro studies show that the half-life of mutant FOXF2 is reduced compared to that of wild type. Foxf2 is expressed in the cochlea of developing and adult mice. The mouse knockout of Foxf2 shows shortened and malformed cochleae, in addition to altered shape of hair cells with innervation and planar cell polarity defects. Expressions of Eya1 and Pax3, genes essential for cochlear development, are reduced in the cochleae of Foxf2 knockout mice. We conclude that FOXF2 plays a major role in cochlear development and its dysfunction leads to SNHL and developmental anomalies of the cochlea in humans and mice.
Collapse
Affiliation(s)
- Guney Bademci
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Clemer Abad
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Armagan Incesulu
- Department of Otolaryngology-Head and Neck Surgery, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Fahed Elian
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Azadeh Reyahi
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Oscar Diaz-Horta
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Filiz B Cengiz
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Claire J Sineni
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Serhat Seyhan
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medical Genetics, Bakirkoy Dr Sadi Konuk Research and Training Hospital, Istanbul, Turkey
| | - Emine Ikbal Atli
- Department of Medical Genetics, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Hikmet Basmak
- Department of Ophthalmology, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Selma Demir
- Department of Medical Genetics, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Ali Moussavi Nik
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Tim Footz
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Shengru Guo
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Duygu Duman
- Division of Pediatric Genetics, Ankara University School of Medicine, Ankara, Turkey
| | - Suat Fitoz
- Department of Radiology, Ankara University School of Medicine, Ankara, Turkey
| | - Hakan Gurkan
- Department of Medical Genetics, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Susan H Blanton
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA.,Dr John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michael A Walter
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Peter Carlsson
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Katherina Walz
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA.,Dr John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mustafa Tekin
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA.,Dr John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
22
|
Čapek D, Smutny M, Tichy AM, Morri M, Janovjak H, Heisenberg CP. Light-activated Frizzled7 reveals a permissive role of non-canonical wnt signaling in mesendoderm cell migration. eLife 2019; 8:e42093. [PMID: 30648973 PMCID: PMC6365057 DOI: 10.7554/elife.42093] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/14/2019] [Indexed: 12/11/2022] Open
Abstract
Non-canonical Wnt signaling plays a central role for coordinated cell polarization and directed migration in metazoan development. While spatiotemporally restricted activation of non-canonical Wnt-signaling drives cell polarization in epithelial tissues, it remains unclear whether such instructive activity is also critical for directed mesenchymal cell migration. Here, we developed a light-activated version of the non-canonical Wnt receptor Frizzled 7 (Fz7) to analyze how restricted activation of non-canonical Wnt signaling affects directed anterior axial mesendoderm (prechordal plate, ppl) cell migration within the zebrafish gastrula. We found that Fz7 signaling is required for ppl cell protrusion formation and migration and that spatiotemporally restricted ectopic activation is capable of redirecting their migration. Finally, we show that uniform activation of Fz7 signaling in ppl cells fully rescues defective directed cell migration in fz7 mutant embryos. Together, our findings reveal that in contrast to the situation in epithelial cells, non-canonical Wnt signaling functions permissively rather than instructively in directed mesenchymal cell migration during gastrulation.
Collapse
Affiliation(s)
- Daniel Čapek
- Institute of Science and Technology AustriaKlosterneuburgAustria
| | - Michael Smutny
- Institute of Science and Technology AustriaKlosterneuburgAustria
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical SchoolUniversity of WarwickCoventryUnited Kingdom
| | - Alexandra-Madelaine Tichy
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health SciencesMonash UniversityClaytonAustralia
- European Molecular Biology Laboratory Australia (EMBL Australia)Monash UniversityClaytonAustralia
| | - Maurizio Morri
- Institute of Science and Technology AustriaKlosterneuburgAustria
| | - Harald Janovjak
- Institute of Science and Technology AustriaKlosterneuburgAustria
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health SciencesMonash UniversityClaytonAustralia
- European Molecular Biology Laboratory Australia (EMBL Australia)Monash UniversityClaytonAustralia
| | | |
Collapse
|
23
|
Diaz-Horta O, Abad C, Cengiz FB, Bademci G, Blackwelder P, Walz K, Tekin M. Ripor2 is involved in auditory hair cell stereociliary bundle structure and orientation. J Mol Med (Berl) 2018; 96:1227-1238. [PMID: 30280293 PMCID: PMC6238639 DOI: 10.1007/s00109-018-1694-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/20/2018] [Accepted: 09/07/2018] [Indexed: 11/30/2022]
Abstract
RIPOR2 (previously known as FAM65B) localizes to stereocilia of auditory hair cells and causes deafness when its function is disturbed by mutations. Here, we demonstrate that during the morphogenesis of the hair cell bundle, absence of Ripor2 affects the orientation of this key subcellular structure. We show that Ripor2 interacts with Myh9, a protein encoded by a known deafness gene. Absence of Ripor2 is associated with low Myh9 abundance in the mouse cochlea despite increased amount of Myh9 transcripts. While Myh9 is mainly expressed in stereocilia, a phosphorylated form of Myh9 is particularly enriched in the kinocilium. In Ripor2-deficient mice, kinocilium shows an aberrant localization which associates with a reduced content of phosphorylated Myh9. Acetylated alpha tubulin, another specific kinociliary protein which contributes to microtubule stabilization, is reduced in the absence of Ripor2 as well. We propose that Ripor2 deficiency influences abundance and/or post-translational modifications of proteins expressed in both stereocilia and kinocilia. This effect may have a negative impact on the structure and function of the auditory hair cell bundle.
Collapse
Affiliation(s)
- Oscar Diaz-Horta
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Clemer Abad
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Filiz Basak Cengiz
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Guney Bademci
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Pat Blackwelder
- MGS/RSMAS and UMCAM/Chemistry, University of Miami, Coral Gables, FL, 33146, USA
- NSU Oceanographic Center, Dania Beach, FL, 33004, USA
| | - Katherina Walz
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Mustafa Tekin
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
24
|
Haag N, Schüler S, Nietzsche S, Hübner CA, Strenzke N, Qualmann B, Kessels MM. The Actin Nucleator Cobl Is Critical for Centriolar Positioning, Postnatal Planar Cell Polarity Refinement, and Function of the Cochlea. Cell Rep 2018; 24:2418-2431.e6. [DOI: 10.1016/j.celrep.2018.07.087] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/18/2018] [Accepted: 07/26/2018] [Indexed: 11/26/2022] Open
|
25
|
Specific Influences of Early Acoustic Environments on Cochlear Hair Cells in Postnatal Mice. Neural Plast 2018; 2018:5616930. [PMID: 29849558 PMCID: PMC5926484 DOI: 10.1155/2018/5616930] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/05/2018] [Accepted: 03/21/2018] [Indexed: 01/24/2023] Open
Abstract
The auditory function develops and matures after birth in many mammalian species. After hearing onset, environmental sounds exert profound and long-term effects on auditory functions. However, the effects of the acoustic environment on the functional development of the peripheral auditory system, especially the cochlear sensory hair cells, are still unclear. In the present study, we exposed mouse pups to frequency-enriched acoustic environments in postnatal days 0–14. The results indicated that the acoustic environment significantly decreased the threshold of the auditory brainstem response in a frequency-specific manner. Compared with controls, no difference was found in the number and alignment of inner and outer hair cells or in the length of hair bundles after acoustic overstimulation. The expression and function of prestin, the motor protein of outer hair cells (OHCs), were specifically increased in OHCs activated by acoustic stimulation at postnatal days 7–11. We analyzed the postnatal maturation of ribbon synapses in the hair cell areas. After acoustic stimulation, the number of ribbon synapses was closer to the mature stage than to the controls. Taken together, these data indicate that early acoustic exposure could promote the functional maturation of cochlear hair cells and the development of hearing.
Collapse
|
26
|
Spatiotemporal coordination of cellular differentiation and tissue morphogenesis in organ of Corti development. Med Mol Morphol 2018. [PMID: 29536272 DOI: 10.1007/s00795-018-0185-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The organ of Corti, an acoustic sensory organ, is a specifically differentiated epithelium of the cochlear duct, which is a part of the membranous labyrinth in the inner ear. Cells in the organ of Corti are generally classified into two kinds; hair cells, which transduce the mechanical stimuli of sound to the cell membrane electrical potential differences, and supporting cells. These cells emerge from homogeneous prosensory epithelium through cell fate determination and differentiation. In the organ of Corti organogenesis, cell differentiation and the rearrangement of their position proceed in parallel, resulting in a characteristic alignment of mature hair cells and supporting cells. Recently, studies have focused on the signaling molecules and transcription factors that regulate cell fate determination and differentiation processes. In comparison, less is known about the mechanism of the formation of the tissue architecture; however, this is important in the morphogenesis of the organ of Corti. Thus, this review will introduce previous findings that focus on how cell fate determination, cell differentiation, and whole tissue morphogenesis proceed in a spatiotemporally and finely coordinated manner. This overview provides an insight into the regulatory mechanisms of the coordination in the developing organ of Corti.
Collapse
|
27
|
Chen Y, Lu X, Guo L, Ni W, Zhang Y, Zhao L, Wu L, Sun S, Zhang S, Tang M, Li W, Chai R, Li H. Hedgehog Signaling Promotes the Proliferation and Subsequent Hair Cell Formation of Progenitor Cells in the Neonatal Mouse Cochlea. Front Mol Neurosci 2017; 10:426. [PMID: 29311816 PMCID: PMC5742997 DOI: 10.3389/fnmol.2017.00426] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 12/06/2017] [Indexed: 12/11/2022] Open
Abstract
Hair cell (HC) loss is the major cause of permanent sensorineural hearing loss in mammals. Unlike lower vertebrates, mammalian cochlear HCs cannot regenerate spontaneously after damage, although the vestibular system does maintain limited HC regeneration capacity. Thus HC regeneration from the damaged sensory epithelium has been one of the main areas of research in the field of hearing restoration. Hedgehog signaling plays important roles during the embryonic development of the inner ear, and it is involved in progenitor cell proliferation and differentiation as well as the cell fate decision. In this study, we show that recombinant Sonic Hedgehog (Shh) protein effectively promotes sphere formation, proliferation, and differentiation of Lgr5+ progenitor cells isolated from the neonatal mouse cochlea. To further explore this, we determined the effect of Hedgehog signaling on cell proliferation and HC regeneration in cultured cochlear explant from transgenic R26-SmoM2 mice that constitutively activate Hedgehog signaling in the supporting cells of the cochlea. Without neomycin treatment, up-regulation of Hedgehog signaling did not significantly promote cell proliferation or new HC formation. However, after injury to the sensory epithelium by neomycin treatment, the over-activation of Hedgehog signaling led to significant supporting cell proliferation and HC regeneration in the cochlear epithelium explants. RNA sequencing and real-time PCR were used to compare the transcripts of the cochleae from control mice and R26-SmoM2 mice, and multiple genes involved in the proliferation and differentiation processes were identified. This study has important implications for the treatment of sensorineural hearing loss by manipulating the Hedgehog signaling pathway.
Collapse
Affiliation(s)
- Yan Chen
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission (NHFPC), Shanghai, China
| | - Xiaoling Lu
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission (NHFPC), Shanghai, China
| | - Luo Guo
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission (NHFPC), Shanghai, China
| | - Wenli Ni
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission (NHFPC), Shanghai, China
| | - Yanping Zhang
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission (NHFPC), Shanghai, China
| | - Liping Zhao
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission (NHFPC), Shanghai, China
| | - Lingjie Wu
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission (NHFPC), Shanghai, China
| | - Shan Sun
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission (NHFPC), Shanghai, China
| | - Shasha Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Wenyan Li
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission (NHFPC), Shanghai, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Huawei Li
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission (NHFPC), Shanghai, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China.,Shanghai Engineering Research Centre of Cochlear Implant, Shanghai, China
| |
Collapse
|
28
|
Abstract
The sensation of touch is mediated by mechanosensory neurons that are embedded in skin and relay signals from the periphery to the central nervous system. During embryogenesis, axons elongate from these neurons to make contact with the developing skin. Concurrently, the epithelium of skin transforms from a homogeneous tissue into a heterogeneous organ that is made up of distinct layers and microdomains. Throughout this process, each neuronal terminal must form connections with an appropriate skin region to serve its function. This Review presents current knowledge of the development of the sensory microdomains in mammalian skin and the mechanosensory neurons that innervate them.
Collapse
Affiliation(s)
- Blair A Jenkins
- Department of Physiology & Cellular Biophysics and Department of Dermatology, Columbia University in the City of New York, New York, NY 10032, USA
| | - Ellen A Lumpkin
- Department of Physiology & Cellular Biophysics and Department of Dermatology, Columbia University in the City of New York, New York, NY 10032, USA
| |
Collapse
|
29
|
Xia W, Hu J, Liu F, Ma J, Sun S, Zhang J, Jin K, Huang J, Jiang N, Wang X, Li W, Ma Z, Ma D. New role of LRP5, associated with nonsyndromic autosomal-recessive hereditary hearing loss. Hum Mutat 2017; 38:1421-1431. [PMID: 28677207 DOI: 10.1002/humu.23285] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 06/19/2017] [Accepted: 06/25/2017] [Indexed: 12/14/2022]
Abstract
Human hearing loss is a common neurosensory disorder about which many basic research and clinically relevant questions are unresolved. At least 50% of hearing loss are due to a genetic etiology. Although hundreds of genes have been reported, there are still hundreds of related deafness genes to be found. Clinical, genetic, and functional investigations were performed to identify the causative mutation in a distinctive Chinese family with postlingual nonsyndromic sensorineural hearing loss. Whole-exome sequencing (WES) identified lipoprotein receptor-related protein 5 (LRP5), a member of the low-density lipoprotein receptor family, as the causative gene in this family. In the zebrafish model, lrp5 downregulation using morpholinos led to significant abnormalities in zebrafish inner ear and lateral line neuromasts and contributed, to some extent, to disabilities in hearing and balance. Rescue experiments showed that LRP5 mutation is associated with hearing loss. Knocking down lrp5 in zebrafish results in reduced expression of several genes linked to Wnt signaling pathway and decreased cell proliferation when compared with those in wild-type zebrafish. In conclusion, the LRP5 mutation influences cell proliferation through the Wnt signaling pathway, thereby reducing the number of supporting cells and hair cells and leading to nonsyndromic hearing loss in this Chinese family.
Collapse
Affiliation(s)
- Wenjun Xia
- Institutes of Biomedical Science, Fudan University, Shanghai, China
| | - Jiongjiong Hu
- Department of Otorhinolaryngology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Fei Liu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jing Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Center Laboratory, Bao'an Maternal and Children Healthcare Hospital, Key Laboratory of Birth Defects Research, Shenzhen, China
| | - Shaoyang Sun
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jin Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kaiyue Jin
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jianbo Huang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Nan Jiang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xu Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wen Li
- Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Zhaoxin Ma
- Department of Otorhinolaryngology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Duan Ma
- Institutes of Biomedical Science, Fudan University, Shanghai, China.,Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Children's Hospital, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Cheng C, Guo L, Lu L, Xu X, Zhang S, Gao J, Waqas M, Zhu C, Chen Y, Zhang X, Xuan C, Gao X, Tang M, Chen F, Shi H, Li H, Chai R. Characterization of the Transcriptomes of Lgr5+ Hair Cell Progenitors and Lgr5- Supporting Cells in the Mouse Cochlea. Front Mol Neurosci 2017; 10:122. [PMID: 28491023 PMCID: PMC5405134 DOI: 10.3389/fnmol.2017.00122] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 04/11/2017] [Indexed: 12/27/2022] Open
Abstract
Cochlear supporting cells (SCs) have been shown to be a promising resource for hair cell (HC) regeneration in the neonatal mouse cochlea. Previous studies have reported that Lgr5+ SCs can regenerate HCs both in vitro and in vivo and thus are considered to be inner ear progenitor cells. Lgr5+ progenitors are able to regenerate more HCs than Lgr5- SCs, and it is important to understand the mechanism behind the proliferation and HC regeneration of these progenitors. Here, we isolated Lgr5+ progenitors and Lgr5- SCs from Lgr5-EGFP-CreERT2/Sox2-CreERT2/Rosa26-tdTomato mice via flow cytometry. As expected, we found that Lgr5+ progenitors had significantly higher proliferation and HC regeneration ability than Lgr5- SCs. Next, we performed RNA-Seq to determine the gene expression profiles of Lgr5+ progenitors and Lgr5- SCs. We analyzed the genes that were enriched and differentially expressed in Lgr5+ progenitors and Lgr5- SCs, and we found 8 cell cycle genes, 9 transcription factors, and 24 cell signaling pathway genes that were uniquely expressed in one population but not the other. Last, we made a protein–protein interaction network to further analyze the role of these differentially expressed genes. In conclusion, we present a set of genes that might regulate the proliferation and HC regeneration ability of Lgr5+ progenitors, and these might serve as potential new therapeutic targets for HC regeneration.
Collapse
Affiliation(s)
- Cheng Cheng
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China.,Research Institute of OtolaryngologyNanjing, China.,Co-innovation Center of Neuroregeneration, Nantong UniversityNantong, China
| | - Luo Guo
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning CommissionShanghai, China
| | - Ling Lu
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical SchoolNanjing, China.,Department of Otolaryngology-Head and Neck Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityNanjing, China
| | - Xiaochen Xu
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - ShaSha Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Junyan Gao
- Health Management and Policy, College of Public Health, Saint Louis University, St. LouisMO, USA
| | - Muhammad Waqas
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China.,Department of Biotechnology, Federal Urdu University of Arts, Science and TechnologyGulshan-e-Iqbal, Pakistan
| | - Chengwen Zhu
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical SchoolNanjing, China
| | - Yan Chen
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning CommissionShanghai, China
| | - Xiaoli Zhang
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical SchoolNanjing, China
| | - Chuanying Xuan
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Xia Gao
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical SchoolNanjing, China
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Fangyi Chen
- Department of Biomedical Engineering, Southern University of Science and TechnologyShenzhen, China
| | - Haibo Shi
- Department of Otorhinolaryngology Head and Neck Surgery, The Sixth People's Hospital, Shanghai Jiao Tong UniversityShanghai, China
| | - Huawei Li
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning CommissionShanghai, China.,Institutes of Biomedical Sciences, Fudan UniversityShanghai, China.,Shanghai Engineering Research Centre of Cochlear ImplantShanghai, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China.,Research Institute of OtolaryngologyNanjing, China.,Co-innovation Center of Neuroregeneration, Nantong UniversityNantong, China
| |
Collapse
|
31
|
Mohd-Zin SW, Marwan AI, Abou Chaar MK, Ahmad-Annuar A, Abdul-Aziz NM. Spina Bifida: Pathogenesis, Mechanisms, and Genes in Mice and Humans. SCIENTIFICA 2017; 2017:5364827. [PMID: 28286691 PMCID: PMC5327787 DOI: 10.1155/2017/5364827] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 11/14/2016] [Accepted: 12/01/2016] [Indexed: 05/26/2023]
Abstract
Spina bifida is among the phenotypes of the larger condition known as neural tube defects (NTDs). It is the most common central nervous system malformation compatible with life and the second leading cause of birth defects after congenital heart defects. In this review paper, we define spina bifida and discuss the phenotypes seen in humans as described by both surgeons and embryologists in order to compare and ultimately contrast it to the leading animal model, the mouse. Our understanding of spina bifida is currently limited to the observations we make in mouse models, which reflect complete or targeted knockouts of genes, which perturb the whole gene(s) without taking into account the issue of haploinsufficiency, which is most prominent in the human spina bifida condition. We thus conclude that the need to study spina bifida in all its forms, both aperta and occulta, is more indicative of the spina bifida in surviving humans and that the measure of deterioration arising from caudal neural tube defects, more commonly known as spina bifida, must be determined by the level of the lesion both in mouse and in man.
Collapse
Affiliation(s)
- Siti W. Mohd-Zin
- Department of Parasitology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Ahmed I. Marwan
- Laboratory for Fetal and Regenerative Biology, Colorado Fetal Care Center, Division of Pediatric Surgery, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, 12700 E 17th Ave, Aurora, CO 80045, USA
| | | | - Azlina Ahmad-Annuar
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Noraishah M. Abdul-Aziz
- Department of Parasitology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
32
|
Synchronized Progression of Prestin Expression and Auditory Brainstem Response during Postnatal Development in Rats. Neural Plast 2016; 2016:4545826. [PMID: 28097024 PMCID: PMC5210174 DOI: 10.1155/2016/4545826] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/08/2016] [Accepted: 11/30/2016] [Indexed: 12/23/2022] Open
Abstract
Prestin is the motor protein expressed in the cochlear outer hair cells (OHCs) of mammalian inner ear. The electromotility of OHCs driven by prestin is responsible for the cochlear amplification which is required for normal hearing in adult animals. Postnatal expression of prestin and activity of OHCs may contribute to the maturation of hearing in rodents. However, the temporal and spatial expression of prestin in cochlea during the development is not well characterized. In the present study, we examined the expression and function of prestin from the OHCs in apical, middle, and basal turns of the cochleae of postnatal rats. Prestin first appeared at postnatal day 6 (P6) for basal turn, P7 in middle turn, and P9 for apical turn of cochlea. The expression level increased progressively over the next few days and by P14 reached the mature level for all three segments. By comparison with the time course of the development of auditory brainstem response for different frequencies, our data reveal that prestin expression synchronized with the hearing development. The present study suggests that the onset time of hearing may require the expression of prestin and is determined by the mature function of OHCs.
Collapse
|
33
|
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
|
34
|
Mah AT, Yan KS, Kuo CJ. Wnt pathway regulation of intestinal stem cells. J Physiol 2016; 594:4837-47. [PMID: 27581568 DOI: 10.1113/jp271754] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 02/08/2016] [Indexed: 12/11/2022] Open
Abstract
Wnt signalling is involved in multiple aspects of embryonic development and adult tissue homeostasis, notably via controlling cellular proliferation and differentiation. Wnt signalling is subject to stringent positive and negative regulation to promote proper development and homeostasis yet avoid aberrant growth. Such multi-layer regulation includes post-translational modification and processing of Wnt proteins themselves, R-spondin (Rspo) amplification of Wnt signalling, diverse receptor families, and intracellular and extracellular antagonists and destruction and transcription complexes. In the gastrointestinal tract, Wnt signalling is crucial for development and renewal of the intestinal epithelium. Intestinal stem cells (ISCs) undergo symmetric division and neutral drift dynamics to renew the intestinal epithelium. Sources of Wnts and Wnt amplifers such as R-spondins are beginning to be elucidated as well as their functional contribution to intestinal homeostasis. In this review we focus on regulation of ISCs and intestinal homeostasis by the Wnt/Rspo pathway, the potential cellular sources of Wnt signalling regulators and highlight potential future areas of study.
Collapse
Affiliation(s)
- Amanda T Mah
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Kelley S Yan
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Calvin J Kuo
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
35
|
ROR1 is essential for proper innervation of auditory hair cells and hearing in humans and mice. Proc Natl Acad Sci U S A 2016; 113:5993-8. [PMID: 27162350 DOI: 10.1073/pnas.1522512113] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Hair cells of the inner ear, the mechanosensory receptors, convert sound waves into neural signals that are passed to the brain via the auditory nerve. Little is known about the molecular mechanisms that govern the development of hair cell-neuronal connections. We ascertained a family with autosomal recessive deafness associated with a common cavity inner ear malformation and auditory neuropathy. Via whole-exome sequencing, we identified a variant (c.2207G>C, p.R736T) in ROR1 (receptor tyrosine kinase-like orphan receptor 1), cosegregating with deafness in the family and absent in ethnicity-matched controls. ROR1 is a tyrosine kinase-like receptor localized at the plasma membrane. At the cellular level, the mutation prevents the protein from reaching the cellular membrane. In the presence of WNT5A, a known ROR1 ligand, the mutated ROR1 fails to activate NF-κB. Ror1 is expressed in the inner ear during development at embryonic and postnatal stages. We demonstrate that Ror1 mutant mice are severely deaf, with preserved otoacoustic emissions. Anatomically, mutant mice display malformed cochleae. Axons of spiral ganglion neurons show fasciculation defects. Type I neurons show impaired synapses with inner hair cells, and type II neurons display aberrant projections through the cochlear sensory epithelium. We conclude that Ror1 is crucial for spiral ganglion neurons to innervate auditory hair cells. Impairment of ROR1 function largely affects development of the inner ear and hearing in humans and mice.
Collapse
|
36
|
Tang ZH, Chen JR, Zheng J, Shi HS, Ding J, Qian XD, Zhang C, Chen JL, Wang CC, Li L, Chen JZ, Yin SK, Huang TS, Chen P, Guan MX, Wang JF. Genetic Correction of Induced Pluripotent Stem Cells From a Deaf Patient With MYO7A Mutation Results in Morphologic and Functional Recovery of the Derived Hair Cell-Like Cells. Stem Cells Transl Med 2016; 5:561-71. [PMID: 27013738 DOI: 10.5966/sctm.2015-0252] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 12/22/2015] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED The genetic correction of induced pluripotent stem cells (iPSCs) induced from somatic cells of patients with sensorineural hearing loss (caused by hereditary factors) is a promising method for its treatment. The correction of gene mutations in iPSCs could restore the normal function of cells and provide a rich source of cells for transplantation. In the present study, iPSCs were generated from a deaf patient with compound heterozygous MYO7A mutations (c.1184G>A and c.4118C>T; P-iPSCs), the asymptomatic father of the patient (MYO7A c.1184G>A mutation; CF-iPSCs), and a normal donor (MYO7A(WT/WT); C-iPSCs). One of MYO7A mutation sites (c.4118C>T) in the P-iPSCs was corrected using CRISPR/Cas9. The corrected iPSCs (CP-iPSCs) retained cell pluripotency and normal karyotypes. Hair cell-like cells induced from CP-iPSCs showed restored organization of stereocilia-like protrusions; moreover, the electrophysiological function of these cells was similar to that of cells induced from C-iPSCs and CF-iPSCs. These results might facilitate the development of iPSC-based gene therapy for genetic disorders. SIGNIFICANCE Induced pluripotent stem cells (iPSCs) were generated from a deaf patient with compound heterozygous MYO7A mutations (c.1184G>A and c.4118C>T). One of the MYO7A mutation sites (c.4118C>T) in the iPSCs was corrected using CRISPR/Cas9. The genetic correction of MYO7A mutation resulted in morphologic and functional recovery of hair cell-like cells derived from iPSCs. These findings confirm the hypothesis that MYO7A plays an important role in the assembly of stereocilia into stereociliary bundles. Thus, the present study might provide further insight into the pathogenesis of sensorineural hearing loss and facilitate the development of therapeutic strategies against monogenic disease through the genetic repair of patient-specific iPSCs.
Collapse
Affiliation(s)
- Zi-Hua Tang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jia-Rong Chen
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jing Zheng
- Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Hao-Song Shi
- Department of Otorhinolaryngology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai, People's Republic of China
| | - Jie Ding
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Xiao-Dan Qian
- The Affiliated Women's Hospital, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Cui Zhang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jian-Ling Chen
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Cui-Cui Wang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Liang Li
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jun-Zhen Chen
- Department of Otolaryngology, The Affiliated Wenling People's Hospital, Wenzhou Medical University, Wenling, Zhejiang, People's Republic of China
| | - Shan-Kai Yin
- Department of Otorhinolaryngology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital Shanghai, People's Republic of China
| | - Tao-Sheng Huang
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Ping Chen
- Departments of Cell Biology and Otolaryngology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Min-Xin Guan
- Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jin-Fu Wang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
37
|
Geng R, Noda T, Mulvaney JF, Lin VYW, Edge ASB, Dabdoub A. Comprehensive Expression of Wnt Signaling Pathway Genes during Development and Maturation of the Mouse Cochlea. PLoS One 2016; 11:e0148339. [PMID: 26859490 PMCID: PMC4747503 DOI: 10.1371/journal.pone.0148339] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 12/14/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND In the inner ear Wnt signaling is necessary for proliferation, cell fate determination, growth of the cochlear duct, polarized orientation of stereociliary bundles, differentiation of the periotic mesenchyme, and homeostasis of the stria vascularis. In neonatal tissue Wnt signaling can drive proliferation of cells in the sensory region, suggesting that Wnt signaling could be used to regenerate the sensory epithelium in the damaged adult inner ear. Manipulation of Wnt signaling for regeneration will require an understanding of the dynamics of Wnt pathway gene expression in the ear. We present a comprehensive screen for 84 Wnt signaling related genes across four developmental and postnatal time points. RESULTS We identified 72 Wnt related genes expressed in the inner ear on embryonic day (E) 12.5, postnatal day (P) 0, P6 and P30. These genes included secreted Wnts, Wnt antagonists, intracellular components of canonical signaling and components of non-canonical signaling/planar cell polarity. CONCLUSION A large number of Wnt signaling molecules were dynamically expressed during cochlear development and in the early postnatal period, suggesting complex regulation of Wnt transduction. The data revealed several potential key regulators for further study.
Collapse
Affiliation(s)
- Ruishuang Geng
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Teppei Noda
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Joanna F. Mulvaney
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Vincent Y. W. Lin
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Otolaryngology—Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Albert S. B. Edge
- Department of Otology and Laryngology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Alain Dabdoub
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Otolaryngology—Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
38
|
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: 33] [Impact Index Per Article: 3.3] [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
|
39
|
Abstract
UNLABELLED The mammalian cochlea exhibit minimal spontaneous regeneration, and loss of sensory hair cells (HCs) results in permanent hearing loss. In nonmammalian vertebrates, spontaneous HC regeneration occurs through both proliferation and differentiation of surrounding supporting cells (SCs). HC regeneration in postnatal mammalian cochleae in vivo remains limited by the small HC number and subsequent death of regenerated HCs. Here, we describe in vivo generation of 10-fold more new HCs in the mouse cochlea than previously reported, most of which survive to adulthood. We achieved this by combining the expression of a constitutively active form of β-catenin (a canonical Wnt activator) with ectopic expression of Atoh1 (a HC fate determination factor) in neonatal Lgr5+ cells (the presumed SC and HC progenitors of the postnatal mouse cochlea), and discovered synergistic increases in proliferation and differentiation. The new HCs were predominantly located near the endogenous inner HCs, expressed early HC differentiation markers, and were innervated despite incomplete alignment of presynaptic and postsynaptic markers. Surprisingly, genetic tracing revealed that only a subset of Lgr5+ cells that lie medial to the inner HCs respond to this combination, highlighting a previously unknown heterogeneity that exists among Lgr5+ cells. Together, our data indicate that β-catenin and Atoh1 mediate synergistic effects on both proliferation and differentiation of a subset of neonatal cochlear Lgr5+ cells, thus overcoming major limitations of HC regeneration in postnatal mouse cochleae in vivo. These results provide a basis for combinatorial therapeutics for hearing restoration. SIGNIFICANCE STATEMENT Hearing loss in humans from aging, noise exposure, or ototoxic drugs (i.e., cisplatin or some antibiotics) is permanent and affects every segments of the population, worldwide. However, birds, frog, and fish have the ability to recover hearing, and recent studies have focused on understanding and applying what we have learned from them for restoring hearing in humans. However, studies have been hampered by low efficiency, limited cell numbers, and subsequent death of these newly generated auditory cells. Here, we describe a combinatorial approach, which results in the generation of auditory cells in greater numbers than previously reported, with most of them surviving to adult ages in vivo. These results provide a basis for combinatorial therapeutics for hearing restoration efforts.
Collapse
|
40
|
Huyghe A, Van den Ackerveken P, Sacheli R, Prévot PP, Thelen N, Renauld J, Thiry M, Delacroix L, Nguyen L, Malgrange B. MicroRNA-124 Regulates Cell Specification in the Cochlea through Modulation of Sfrp4/5. Cell Rep 2015; 13:31-42. [PMID: 26387953 DOI: 10.1016/j.celrep.2015.08.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/01/2015] [Accepted: 08/18/2015] [Indexed: 02/08/2023] Open
Abstract
The organ of Corti, the auditory organ of the mammalian inner ear, contains sensory hair cells and supporting cells that arise from a common sensory progenitor. The molecular bases allowing the specification of these progenitors remain elusive. In the present study, by combining microarray analyses with conditional deletion of Dicer in the developing inner ear, we identified that miR-124 controls cell fate in the developing organ of Corti. By targeting secreted frizzled-related protein 4 (Sfrp4) and Sfrp5, two inhibitors of the Wnt pathway, we showed that miR-124 controls the β-catenin-dependent and also the PCP-related non-canonical Wnt pathways that contribute to HC differentiation and polarization in the organ of Corti. Thus, our work emphasizes the importance of miR-124 as an epigenetic safeguard that fine-tunes the expression of genes critical for cell patterning during cochlear differentiation.
Collapse
Affiliation(s)
- Aurélia Huyghe
- GIGA-Neurosciences, University of Liege, C.H.U. Sart Tilman, Liege 4000, Belgium
| | | | - Rosalie Sacheli
- GIGA-Neurosciences, University of Liege, C.H.U. Sart Tilman, Liege 4000, Belgium
| | - Pierre-Paul Prévot
- GIGA-Neurosciences, University of Liege, C.H.U. Sart Tilman, Liege 4000, Belgium
| | - Nicolas Thelen
- GIGA-Neurosciences, University of Liege, C.H.U. Sart Tilman, Liege 4000, Belgium
| | - Justine Renauld
- GIGA-Neurosciences, University of Liege, C.H.U. Sart Tilman, Liege 4000, Belgium
| | - Marc Thiry
- GIGA-Neurosciences, University of Liege, C.H.U. Sart Tilman, Liege 4000, Belgium
| | - Laurence Delacroix
- GIGA-Neurosciences, University of Liege, C.H.U. Sart Tilman, Liege 4000, Belgium
| | - Laurent Nguyen
- GIGA-Neurosciences, University of Liege, C.H.U. Sart Tilman, Liege 4000, Belgium
| | - Brigitte Malgrange
- GIGA-Neurosciences, University of Liege, C.H.U. Sart Tilman, Liege 4000, Belgium.
| |
Collapse
|
41
|
Chen SY, Han B, Zhu YT, Mahabole M, Huang J, Beebe DC, Tseng SCG. HC-HA/PTX3 Purified From Amniotic Membrane Promotes BMP Signaling in Limbal Niche Cells to Maintain Quiescence of Limbal Epithelial Progenitor/Stem Cells. Stem Cells 2015; 33:3341-55. [PMID: 26148958 DOI: 10.1002/stem.2091] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/08/2015] [Accepted: 05/23/2015] [Indexed: 12/13/2022]
Abstract
To explore how limbal niche cells (LNCs) may control quiescence, self-renewal, and corneal epithelial lineage commitment/differentiation of limbal epithelial progenitor/stem cells (LEPCs), we have established an in vitro sphere assay by reunion between the two cell types in three-dimensional Matrigel. The resultant sphere exhibits inhibition of corneal epithelial lineage commitment/differentiation and marked clonal growth of LEPCs, of which the latter is correlated with activation of canonical Wnt signaling. Herein, we have created a similar reunion assay in immobilized heavy chain-hyaluronic acid/pentraxin 3 (HC-HA/PTX3), which is purified from amniotic membrane (AM) and consists of a complex formed by hyaluronic covalently linked to heavy chain 1 of inter-α-inhibitor and noncovalently linked to pentraxin 3. The resultant spheres exhibited similar suppression of corneal epithelial lineage commitment/differentiation but upregulation of quiescence markers including nuclear translocation of Bmi-1, and negligible clonal growth of LEPCs. This outcome was correlated with the suppression of canonical Wnt but activation of noncanonical (Planar cell polarity) Wnt signaling as well as BMP signaling in both LEPCs and LNCs. The activation of BMP signaling in LNCs was pivotal because nuclear translocation of pSmad1/5/8 was prohibited in hLEPCs when reunioned with mLNCs of conditionally deleted Bmpr1a;Acvr1(DCKO) mice. Furthermore, ablation of BMP signaling in LEPCs led to upregulation of cell cycle genes, downregulation of Bmi-1, nuclear exclusion of phosphorylated Bmi-1, and marked promotion of the clonal growth of LEPCs. Hence, HC-HA/PTX3 uniquely upregulates BMP signaling in LNCs which leads to BMP signaling in LEPCs to achieve quiescence, helping explain how AM transplantation is clinically useful to be used as a matrix for ex vivo expansion of LEPCs and to treat corneal blindness caused by limbal stem cells deficiency.
Collapse
Affiliation(s)
- Szu-Yu Chen
- R&D Department, TissueTech, Inc., Miami, Florida, USA
| | - Bo Han
- Ocular Surface Research & Education Foundation, Miami, Florida, USA.,Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Ying-Ting Zhu
- R&D Department, TissueTech, Inc., Miami, Florida, USA
| | | | - Jie Huang
- Department of Ophthalmology and Visual Sciences, Washington University, St. Louis, Missouri, USA
| | - David C Beebe
- Department of Ophthalmology and Visual Sciences, Washington University, St. Louis, Missouri, USA
| | - Scheffer C G Tseng
- R&D Department, TissueTech, Inc., Miami, Florida, USA.,Ocular Surface Research & Education Foundation, Miami, Florida, USA
| |
Collapse
|
42
|
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: 111] [Impact Index Per Article: 11.1] [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
|
43
|
Paulus YM, Alcorn DM, Gaynon M, Moshfeghi DM. Peripheral Avascular Retina in a Term Male Neonate With Microvillus Inclusion Disease and Pancreatic Insufficiency. Ophthalmic Surg Lasers Imaging Retina 2015; 46:589-91. [PMID: 26057766 DOI: 10.3928/23258160-20150521-14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 03/17/2015] [Indexed: 11/20/2022]
Abstract
The authors present the first case of peripheral avascular retina in a term male neonate with pancreatic exocrine insufficiency, atypical microvillus inclusion disease, flat tympanograms, and recurrent urinary tract infections. Clinical examination showed avascular peripheral retina to posterior zone II temporally, with a flat stage 1-like demarcation line, and no plus disease. Genetic testing results were normal. The patient developed peripheral neovascularization and underwent panretinal photocoagulation. This case likely represents mild Norrie disease, familial exudative vitreoretinopathy, or incontinentia pigmenti due to a Wnt signaling abnormality. While these conditions are usually more severe, a variable spectrum of Wnt abnormalities exists throughout the body.
Collapse
|
44
|
He Y, Tang D, Cai C, Chai R, Li H. LSD1 is Required for Hair Cell Regeneration in Zebrafish. Mol Neurobiol 2015; 53:2421-34. [PMID: 26008620 DOI: 10.1007/s12035-015-9206-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 05/01/2015] [Indexed: 02/06/2023]
Abstract
Lysine-specific demethylase 1 (LSD1/KDM1A) plays an important role in complex cellular processes such as differentiation, proliferation, apoptosis, and cell cycle progression. It has recently been demonstrated that during development, downregulation of LSD1 inhibits cell proliferation, modulates the expression of cell cycle regulators, and reduces hair cell formation in the zebrafish lateral line, which suggests that LSD1-mediated epigenetic regulation plays a key role in the development of hair cells. However, the role of LSD1 in hair cell regeneration after hair cell loss remains poorly understood. Here, we demonstrate the effect of LSD1 on hair cell regeneration following neomycin-induced hair cell loss. We show that the LSD1 inhibitor trans-2-phenylcyclopropylamine (2-PCPA) significantly decreases the regeneration of hair cells in zebrafish after neomycin damage. In addition, immunofluorescent staining demonstrates that 2-PCPA administration suppresses supporting cell proliferation and alters cell cycle progression. Finally, in situ hybridization shows that 2-PCPA significantly downregulates the expression of genes related to Wnt/β-catenin and Fgf activation. Altogether, our data suggest that downregulation of LSD1 significantly decreases hair cell regeneration after neomycin-induced hair cell loss through inactivation of the Wnt/β-catenin and Fgf signaling pathways. Thus, LSD1 plays a critical role in hair cell regeneration and might represent a novel biomarker and potential therapeutic approach for the treatment of hearing loss.
Collapse
Affiliation(s)
- Yingzi He
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, 200031, People's Republic of China
| | - Dongmei Tang
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, 200031, People's Republic of China
| | - Chengfu Cai
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Renjie Chai
- Co-innovation Center of Neuroregeneration, Key Laboratory for Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing, Jiangsu, 210096, People's Republic of China
| | - Huawei Li
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, 200031, People's Republic of China. .,State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, People's Republic of China. .,Institute of Stem Cell and Regeneration Medicine, Institute of Biomedical Science, Fudan University, Shanghai, People's Republic of China. .,Key Laboratory of Hearing Science, Ministry of Health, EENT Hospital, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
45
|
Sienknecht UJ. Current concepts of hair cell differentiation and planar cell polarity in inner ear sensory organs. Cell Tissue Res 2015; 361:25-32. [PMID: 25959294 DOI: 10.1007/s00441-015-2200-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 04/15/2015] [Indexed: 11/25/2022]
Abstract
Phylogenetically and ontogenetically, vertebrate development led to the generation of several inner ear sensory organs. During embryogenesis, cell fate specification determines whether each progenitor cell differentiates into a sensory hair cell or a supporting cell within the common sensory primordium. Finally, all sensory epithelia of the inner ear consist of a hair cell/supporting cell mosaic, albeit with anatomical differences depending on the sensory organ type. Hair cells develop a polarized bundle of stereovilli that is of functional importance for mechanotransduction. After initiating stereovillar development, hair cells align their bundles in a coordinated fashion, generating a characteristic hair cell orientation pattern, a process referred to as planar cell polarity (PCP). The pathway that controls PCP in the inner ear needs both to establish the development of a polarized morphology of the stereovillar bundle of the hair cell and to organize a systematic hair cell alignment. Because the hair cell orientation patterns of the various inner ear organs and vertebrate species differ fundamentally, it becomes apparent that in vertebrates, different aspects of PCP need to be independently controlled. In spite of important progress recently gained in the field of PCP research, we still need to identify the mechanisms (1) that initiate molecular asymmetries in cells, (2) that guide the transmission of polarity information from cell to cell, and (3) that consistently translate such polarity information into morphological asymmetries of hair cells.
Collapse
Affiliation(s)
- Ulrike J Sienknecht
- Department of Neuroscience, School of Medicine and Health Science, Research Center Neurosensory Science, Cluster of Excellence "Hearing4all", Carl von Ossietzky University Oldenburg, Carl von Ossietzky Strasse 9-11, 26129, Oldenburg, Germany,
| |
Collapse
|
46
|
Iizuka-Kogo A, Senda T, Akiyama T, Shimomura A, Nomura R, Hasegawa Y, Yamamura KI, Kogo H, Sawai N, Matsuzaki T. Requirement of DLG1 for cardiovascular development and tissue elongation during cochlear, enteric, and skeletal development: possible role in convergent extension. PLoS One 2015; 10:e0123965. [PMID: 25860837 PMCID: PMC4393223 DOI: 10.1371/journal.pone.0123965] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 01/07/2015] [Indexed: 11/18/2022] Open
Abstract
The Dlg1 gene encodes a member of the MAGUK protein family involved in the polarization of epithelial cells. Null mutant mice for the Dlg1 gene (Dlg1-/- mice) exhibit respiratory failure and cyanosis, and die soon after birth. However, the cause of this neonatal lethality has not been determined. In the present study, we further examined Dlg1-/- mice and found severe defects in the cardiovascular system, including ventricular septal defect, persistent truncus arteriosus, and double outlet right ventricle, which would cause the neonatal lethality. These cardiovascular phenotypes resemble those of mutant mice lacking planar cell polarity (PCP) genes and support a recent notion that DLG1 is involved in the PCP pathway. We assessed the degree of involvement of DLG1 in the development of other organs, as the cochlea, intestine, and skeleton, in which PCP signaling has been suggested to play a role. In the organ of Corti, tissue elongation was inhibited accompanied by disorganized arrangement of the hair cell rows, while the orientation of the stereocilia bundle was normal. In the sternum, cleft sternum, abnormal calcification pattern of cartilage, and disorganization of chondrocytes were observed. Furthermore, shortening of the intestine, sternum, and long bones of the limbs was observed. These phenotypes of Dlg1-/- mice involving cellular disorganization and insufficient tissue elongation strongly suggest a defect in the convergent extension movements in these mice. Thus, our present results provide a possibility that DLG1 is particularly required for convergent extension among PCP signaling-dependent processes.
Collapse
Affiliation(s)
- Akiko Iizuka-Kogo
- Department of Anatomy I, Fujita Health University School of Medicine, Aichi, Japan
- Department of Anatomy and Cell Biology, Gunma University Graduate School of Medicine, Gunma, Japan
- * E-mail:
| | - Takao Senda
- Department of Anatomy I, Fujita Health University School of Medicine, Aichi, Japan
- Department of Anatomy, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tetsu Akiyama
- Laboratory of Molecular and Genetic Information, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Atsushi Shimomura
- Department of Anatomy I, Fujita Health University School of Medicine, Aichi, Japan
- The Department of Communication Disorders, School of Psychological Science, Health Sciences University of Hokkaido, Hokkaido, Japan
| | - Ryuji Nomura
- Department of Anatomy I, Fujita Health University School of Medicine, Aichi, Japan
| | - Yoshimi Hasegawa
- Department of Anatomy I, Fujita Health University School of Medicine, Aichi, Japan
| | - Ken-ichi Yamamura
- Division of Developmental Genetics, Institute of Resource Development Analysis, Kumamoto University, Kumamoto, Japan
| | - Hiroshi Kogo
- Department of Anatomy and Cell Biology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Nobuhiko Sawai
- Department of Anatomy and Cell Biology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Toshiyuki Matsuzaki
- Department of Anatomy and Cell Biology, Gunma University Graduate School of Medicine, Gunma, Japan
| |
Collapse
|
47
|
Jansson L, Kim GS, Cheng AG. Making sense of Wnt signaling-linking hair cell regeneration to development. Front Cell Neurosci 2015; 9:66. [PMID: 25814927 PMCID: PMC4356074 DOI: 10.3389/fncel.2015.00066] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 02/12/2015] [Indexed: 01/10/2023] Open
Abstract
Wnt signaling is a highly conserved pathway crucial for development and homeostasis of multicellular organisms. Secreted Wnt ligands bind Frizzled receptors to regulate diverse processes such as axis patterning, cell division, and cell fate specification. They also serve to govern self-renewal of somatic stem cells in several adult tissues. The complexity of the pathway can be attributed to the myriad of Wnt and Frizzled combinations as well as its diverse context-dependent functions. In the developing mouse inner ear, Wnt signaling plays diverse roles, including specification of the otic placode and patterning of the otic vesicle. At later stages, its activity governs sensory hair cell specification, cell cycle regulation, and hair cell orientation. In regenerating sensory organs from non-mammalian species, Wnt signaling can also regulate the extent of proliferative hair cell regeneration. This review describes the current knowledge of the roles of Wnt signaling and Wnt-responsive cells in hair cell development and regeneration. We also discuss possible future directions and the potential application and limitation of Wnt signaling in augmenting hair cell regeneration.
Collapse
Affiliation(s)
- Lina Jansson
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University Stanford, CA, USA
| | - Grace S Kim
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University Stanford, CA, USA
| | - Alan G Cheng
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University Stanford, CA, USA
| |
Collapse
|
48
|
Bokolia NP, Mishra M. Hearing molecules, mechanism and transportation: modeled in Drosophila melanogaster. Dev Neurobiol 2014; 75:109-30. [PMID: 25081222 DOI: 10.1002/dneu.22221] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 07/29/2014] [Accepted: 07/29/2014] [Indexed: 01/19/2023]
Abstract
Mechanosensory transduction underlies the perception of touch, sound and acceleration. The mechanical signals exist in the environment are resensed by the specialized mechanosensory cells, which convert the external forces into the electrical signals. Hearing is a magnificent example that relies on the mechanotransduction mediated by the auditory cells, for example the inner-ear hair cells in vertebrates and the Johnston's organ (JO) in fly. Previous studies have shown the fundamental physiological processes in the fly and vertebrate auditory organs are similar, suggesting that there might be a set of similar molecules underlying these processes. The molecular studies of the fly JO have been shown to be remarkably successful in discovering the developmental and functional genes that provided further implications in vertebrates. Several evolutionarily conserved molecules and signaling pathways have been shown to govern the development of the auditory organs in both vertebrates and invertebrates. The current review describes the similarities and differences between the vertebrate and fly auditory organs at developmental, structural, molecular, and transportation levels.
Collapse
Affiliation(s)
- Naveen Prakash Bokolia
- Department of Life Science, National Institute of Technology Rourkela, Rourkela, Orissa, India
| | | |
Collapse
|
49
|
Sienknecht UJ, Köppl C, Fritzsch B. Evolution and Development of Hair Cell Polarity and Efferent Function in the Inner Ear. BRAIN, BEHAVIOR AND EVOLUTION 2014; 83:150-61. [DOI: 10.1159/000357752] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 12/03/2013] [Indexed: 11/19/2022]
|
50
|
ACF7 is a hair-bundle antecedent, positioned to integrate cuticular plate actin and somatic tubulin. J Neurosci 2014; 34:305-12. [PMID: 24381291 DOI: 10.1523/jneurosci.1880-13.2014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The precise morphology of the mechanosensitive hair bundle requires seamless integration of actin and microtubule networks. Here, we identify Acf7a (actin crosslinking family protein 7a) as a protein positioned to bridge these distinct cytoskeletal networks in hair cells. By imaging Acf7a-Citrine fusion protein in zebrafish and immunolabeling of vestibular and cochlear mouse hair cells, we show that Acf7a and ACF7 circumscribe, underlie, and are interwoven into the cuticular plate (CP), and they also encircle the basal body of the kinocilium. In cochlear hair cells, ACF7 localization is graded, with the highest concentration near each fonticulus--an area free of F-actin in the region of the CP that contains the basal body. During hair-cell development and regeneration, Acf7a precedes formation of the hair bundle and CP. Finally, electron tomography demonstrates that the ends of microtubules insert into the CP and are decorated with filamentous linkers connecting microtubules to the CP. These observations are consistent with ACF7 being a linker protein, which may shape the cytoskeleton of the hair cell early during hair-bundle genesis.
Collapse
|