Detailed analysis of chick optic fissure closure reveals Netrin-1 as an essential mediator of epithelial fusion

  1. Holly Hardy
  2. James GD Prendergast
  3. Aara Patel
  4. Sunit Dutta
  5. Violeta Trejo-Reveles
  6. Hannah Kroeger
  7. Andrea R Yung
  8. Lisa V Goodrich
  9. Brian Brooks
  10. Jane C Sowden
  11. Joe Rainger  Is a corresponding author
  1. University of Edinburgh, United Kingdom
  2. UCL Great Ormond Street Institute of Child Health, United Kingdom
  3. National Eye Institute, National Institutes of Health, United States
  4. Harvard Medical School, United States

Abstract

Epithelial fusion underlies many vital organogenic processes during embryogenesis. Disruptions to these cause a significant number of human birth defects, including ocular coloboma. We provide robust spatial-temporal staging and unique anatomical detail of optic fissure closure (OFC) in the embryonic chick, including evidence for roles of apoptosis and epithelial remodelling. We performed complementary transcriptomic profiling and show that Netrin-1 (NTN1) is precisely expressed in the chick fissure margin at the fusion plate but is immediately downregulated after fusion. We further provide a combination of protein localisation and phenotypic evidence in chick, humans, mice and zebrafish that Netrin-1 has an evolutionarily conserved and essential requirement for OFC, and is likely to have an important role in palate fusion. Our data suggest that NTN1 is a strong candidate locus for human coloboma and other multi-system developmental fusion defects, and show that chick OFC is a powerful model for epithelial fusion research.

Data availability

All RNAseq data files are submitted to the NCBI Gene Expression Ominibus database (http://www.ncbi.nlm.nih.gov/geo) with the accession number GSE84916.

The following data sets were generated

Article and author information

Author details

  1. Holly Hardy

    The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-4603-7784
  2. James GD Prendergast

    The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-8916-018X
  3. Aara Patel

    Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  4. Sunit Dutta

    Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, United States
    Competing interests
    The authors declare that no competing interests exist.
  5. Violeta Trejo-Reveles

    The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  6. Hannah Kroeger

    The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  7. Andrea R Yung

    Department of Neurobiology, Harvard Medical School, Boston, United States
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-4053-378X
  8. Lisa V Goodrich

    Department of Neurobiology, Harvard Medical School, Boston, United States
    Competing interests
    The authors declare that no competing interests exist.
  9. Brian Brooks

    Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, United States
    Competing interests
    The authors declare that no competing interests exist.
  10. Jane C Sowden

    Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  11. Joe Rainger

    The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
    For correspondence
    joe.rainger@roslin.ed.ac.uk
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-1091-5100

Funding

Fight for Sight UK (1590/1591)

  • Joe Rainger

Company of Biologists (DMMTF-180520)

  • Joe Rainger

Biotechnology and Biological Sciences Research Council (BB/P013732/1)

  • Joe Rainger

Wellcome (ISSF3)

  • Joe Rainger

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

Reviewing Editor

  1. Jeremy Nathans, Johns Hopkins University School of Medicine, United States

Ethics

Animal experimentation: All animal work was carried out in strict accordance with the United Kingdom Home Office Animal (Scientific Procedures) Act 1986. All chicken experiments, breeding and care procedures were approved and carried out under license from the UK Home Office (PPL 7008940 - Prof Helen Sang) and subject to local ethical review by the Roslin Institute AWERB. No regulated procedures were used in this study. Generation and maintenance of memGFP flock were in accordance with annex III of Directive 2010/63 EU and Home Office Codes of Practice. All mouse and zebrafish work was conducted in compliance with protocols approved by the Institutional Animal Care and Use Committee at Harvard Medical School, and at The NIH National Eye Institute. Mice were used from an existing study (Yung et al., Development. 2015). Ntn -/- (Ntn1tm1.1Good, MGI:5888900) and C57Bl/6J animals were maintained on a standard 12hr light-dark cycle. Mice received food and water ad lib and were provided with fresh bedding and nesting daily. All experiments were conducted in agreement with the Animals (Scientific Procedures) Act 1986 and the Association for Research in Vision and Ophthalmology Statement for the Use of Animals in Ophthalmic and Vision Research. Pregnant dams were anaesthetised by CO2 asphyxiation and euthanised by cervical dislocation. Embryos were collected at E11.5, E15.5 and E16.5. All embryos were immediately culled on ice by decapitation. All zebrafish embryos/larvae are taken at between 30 hpf-56 hpf and immediately anaesthetised with tricaine methane sulfonate (MS222, 168 mg/l) on ice. Embryos are then euthanised in bleach solution (sodium hypochlorite 6.15%) in water at 1 part bleach to 5 parts water. The larvae remain in this solution at least five minutes prior to disposal to ensure death.

Human subjects: Human foetal eyes were obtained from the Joint Medical Research Council UK (grant # G0700089)/Wellcome Trust (grant # GR082557) Human Developmental Biology Resource (http://www.hdbr.org/). The consent, use and disposal of HDBR samples is regulated by the UK Human Tissue Authority (HTA). The HDBR is a Research Ethics Committee (REC) approved and HTA licenced tissue bank. This means that most research projects based within the UK do not need to obtain their own REC approval.

Version history

  1. Received: November 23, 2018
  2. Accepted: June 3, 2019
  3. Accepted Manuscript published: June 4, 2019 (version 1)
  4. Version of Record published: July 2, 2019 (version 2)

Copyright

© 2019, Hardy et al.

This article is distributed under the terms of the Creative Commons Attribution License permitting unrestricted use and redistribution provided that the original author and source are credited.

Metrics

  • 2,395
    views
  • 313
    downloads
  • 28
    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. Holly Hardy
  2. James GD Prendergast
  3. Aara Patel
  4. Sunit Dutta
  5. Violeta Trejo-Reveles
  6. Hannah Kroeger
  7. Andrea R Yung
  8. Lisa V Goodrich
  9. Brian Brooks
  10. Jane C Sowden
  11. Joe Rainger
(2019)
Detailed analysis of chick optic fissure closure reveals Netrin-1 as an essential mediator of epithelial fusion
eLife 8:e43877.
https://doi.org/10.7554/eLife.43877

Share this article

https://doi.org/10.7554/eLife.43877

Further reading

    1. Developmental Biology
    2. Medicine
    Stephen E Flaherty III, Olivier Bezy ... Zhidan Wu
    Research Article

    From a forward mutagenetic screen to discover mutations associated with obesity, we identified mutations in the Spag7 gene linked to metabolic dysfunction in mice. Here, we show that SPAG7 KO mice are born smaller and develop obesity and glucose intolerance in adulthood. This obesity does not stem from hyperphagia, but a decrease in energy expenditure. The KO animals also display reduced exercise tolerance and muscle function due to impaired mitochondrial function. Furthermore, SPAG7-deficiency in developing embryos leads to intrauterine growth restriction, brought on by placental insufficiency, likely due to abnormal development of the placental junctional zone. This insufficiency leads to loss of SPAG7-deficient fetuses in utero and reduced birth weights of those that survive. We hypothesize that a ‘thrifty phenotype’ is ingrained in SPAG7 KO animals during development that leads to adult obesity. Collectively, these results indicate that SPAG7 is essential for embryonic development and energy homeostasis later in life.

    1. Developmental Biology
    2. Stem Cells and Regenerative Medicine
    Nikola Sekulovski, Jenna C Wettstein ... Kenichiro Taniguchi
    Research Article

    Amniogenesis, a process critical for continuation of healthy pregnancy, is triggered in a collection of pluripotent epiblast cells as the human embryo implants. Previous studies have established that bone morphogenetic protein (BMP) signaling is a major driver of this lineage specifying process, but the downstream BMP-dependent transcriptional networks that lead to successful amniogenesis remain to be identified. This is, in part, due to the current lack of a robust and reproducible model system that enables mechanistic investigations exclusively into amniogenesis. Here, we developed an improved model of early amnion specification, using a human pluripotent stem cell-based platform in which the activation of BMP signaling is controlled and synchronous. Uniform amniogenesis is seen within 48 hr after BMP activation, and the resulting cells share transcriptomic characteristics with amnion cells of a gastrulating human embryo. Using detailed time-course transcriptomic analyses, we established a previously uncharacterized BMP-dependent amniotic transcriptional cascade, and identified markers that represent five distinct stages of amnion fate specification; the expression of selected markers was validated in early post-implantation macaque embryos. Moreover, a cohort of factors that could potentially control specific stages of amniogenesis was identified, including the transcription factor TFAP2A. Functionally, we determined that, once amniogenesis is triggered by the BMP pathway, TFAP2A controls the progression of amniogenesis. This work presents a temporally resolved transcriptomic resource for several previously uncharacterized amniogenesis states and demonstrates a critical intermediate role for TFAP2A during amnion fate specification.