Gradients of glucose metabolism regulate morphogen signalling required for specifying tonotopic organization in the chicken cochlea

  1. James DB O'Sullivan
  2. Thomas S Blacker
  3. Claire Scott
  4. Weise Chang
  5. Mohi Ahmed
  6. Val Yianni
  7. Zoe F Mann  Is a corresponding author
  1. King's College London, United Kingdom
  2. University College London, United Kingdom
  3. National Institute on Deafness and Other Communication Disorders, United States

Abstract

In vertebrates with elongated auditory organs, mechanosensory hair cells (HCs) are organised such that complex sounds are broken down into their component frequencies along a proximal-to-distal long (tonotopic) axis. Acquisition of unique morphologies at the appropriate position along the chick cochlea, the basilar papilla (BP), requires that nascent HCs determine their tonotopic positions during development. The complex signalling within the auditory organ between a developing HC and its local niche along the cochlea is poorly understood. Using a combination of live imaging and NAD(P)H fluorescence lifetime imaging (FLIM) we reveal that there is a gradient in the cellular balance between glycolysis and the pentose phosphate pathway in developing HCs along the tonotopic axis. Perturbing this balance by inhibiting different branches of cytosolic glucose catabolism disrupts developmental morphogen signalling and abolishes the normal tonotopic gradient in hair cell morphology. These findings highlight a causal link between graded morphogen signalling and metabolic reprogramming in specifying the tonotopic identity of developing HCs.

Data availability

All data and source data are available in manuscript and supporting files.

Article and author information

Author details

  1. James DB O'Sullivan

    Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  2. Thomas S Blacker

    Research Department of Structural and Molecular Biology, University College London, London, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-8949-6238
  3. Claire Scott

    Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  4. Weise Chang

    National Institute on Deafness and Other Communication Disorders, Bethesda, United States
    Competing interests
    The authors declare that no competing interests exist.
  5. Mohi Ahmed

    Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  6. Val Yianni

    Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-9857-7577
  7. Zoe F Mann

    Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
    For correspondence
    zoe.mann@kcl.ac.uk
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-4916-9574

Funding

Biotechnology and Biological Sciences Research Council (BB/V006371/1)

  • Zoe F Mann

Physiological Society

  • Zoe F Mann

The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Copyright

This is an open-access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 public domain dedication.

Metrics

  • 1,043
    views
  • 184
    downloads
  • 5
    citations

Views, downloads and citations are aggregated across all versions of this paper published by eLife.

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. James DB O'Sullivan
  2. Thomas S Blacker
  3. Claire Scott
  4. Weise Chang
  5. Mohi Ahmed
  6. Val Yianni
  7. Zoe F Mann
(2023)
Gradients of glucose metabolism regulate morphogen signalling required for specifying tonotopic organization in the chicken cochlea
eLife 12:e86233.
https://doi.org/10.7554/eLife.86233

Share this article

https://doi.org/10.7554/eLife.86233

Further reading

    1. Cell Biology
    2. Developmental Biology
    Deepak Adhikari, John Carroll
    Insight

    The formation of large endolysosomal structures in unfertilized eggs ensures that lysosomes remain dormant before fertilization, and then shift into clean-up mode after the egg-to-embryo transition.

    1. Cell Biology
    2. Developmental Biology
    Filip Knop, Apolena Zounarová ... Marie Macůrková
    Research Article Updated

    During Caenorhabditis elegans development, multiple cells migrate long distances or extend processes to reach their final position and/or attain proper shape. The Wnt signalling pathway stands out as one of the major coordinators of cell migration or cell outgrowth along the anterior-posterior body axis. The outcome of Wnt signalling is fine-tuned by various mechanisms including endocytosis. In this study, we show that SEL-5, the C. elegans orthologue of mammalian AP2-associated kinase AAK1, acts together with the retromer complex as a positive regulator of EGL-20/Wnt signalling during the migration of QL neuroblast daughter cells. At the same time, SEL-5 in cooperation with the retromer complex is also required during excretory canal cell outgrowth. Importantly, SEL-5 kinase activity is not required for its role in neuronal migration or excretory cell outgrowth, and neither of these processes is dependent on DPY-23/AP2M1 phosphorylation. We further establish that the Wnt proteins CWN-1 and CWN-2, together with the Frizzled receptor CFZ-2, positively regulate excretory cell outgrowth, while LIN-44/Wnt and LIN-17/Frizzled together generate a stop signal inhibiting its extension.