Paraxial mesoderm organoids model development of human somites

  1. Christoph Budjan
  2. Shichen Liu
  3. Adrian Ranga
  4. Senjuti Gayen
  5. Olivier Pourquié
  6. Sahand Hormoz  Is a corresponding author
  1. Dana-Farber Cancer Institute, United States
  2. KU Leuven, Belgium
  3. Harvard Medical School, United States

Abstract

During the development of the vertebrate embryo, segmented structures called somites are periodically formed from the presomitic mesoderm (PSM), and give rise to the vertebral column. While somite formation has been studied in several animal models, it is less clear how well this process is conserved in humans. Recent progress has made it possible to study aspects of human paraxial mesoderm development such as the human segmentation clock in vitro using human pluripotent stem cells (hPSCs), however, somite formation has not been observed in these monolayer cultures. Here, we describe the generation of human paraxial mesoderm (PM) organoids from hPSCs (termed Somitoids), which recapitulate the molecular, morphological and functional features of paraxial mesoderm development, including formation of somite-like structures in vitro. Using a quantitative image-based screen, we identify critical parameters such as initial cell number and signaling modulations that reproducibly yielded formation of somite-like structures in our organoid system. In addition, using single-cell RNA sequencing and 3D imaging, we show that PM organoids both transcriptionally and morphologically resemble their in vivo counterparts and can be differentiated into somite derivatives. Our organoid system is reproducible and scalable, allowing for the systematic and quantitative analysis of human spinal cord development and disease in vitro.

Data availability

Sequencing data has been deposited in GEO under accession code GSE194214; All data used to generate the figures are included in the manuscript as Source Data files

The following data sets were generated

Article and author information

Author details

  1. Christoph Budjan

    Department of Data Sciences, Dana-Farber Cancer Institute, Boston, United States
    Competing interests
    The authors declare that no competing interests exist.
  2. Shichen Liu

    Department of Data Sciences, Dana-Farber Cancer Institute, Boston, United States
    Competing interests
    The authors declare that no competing interests exist.
  3. Adrian Ranga

    KU Leuven, KU Leuven, Department of Mechanical Engineering, Belgium
    Competing interests
    The authors declare that no competing interests exist.
  4. Senjuti Gayen

    Department of Data Sciences, Dana-Farber Cancer Institute, Boston, United States
    Competing interests
    The authors declare that no competing interests exist.
  5. Olivier Pourquié

    Department of Genetics, Harvard Medical School, Boston, United States
    Competing interests
    The authors declare that no competing interests exist.
  6. Sahand Hormoz

    Department of Genetics, Dana-Farber Cancer Institute, Boston, United States
    For correspondence
    sahand_hormoz@hms.harvard.edu
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-4384-4428

Funding

National Institutes of Health (P01GM099117)

  • Christoph Budjan
  • Sahand Hormoz

National Heart, Lung, and Blood Institute (R01HL158269)

  • Christoph Budjan
  • Sahand Hormoz

Chan Zuckerberg Initiative (2018-183143)

  • Christoph Budjan
  • Sahand Hormoz

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

Reviewing Editor

  1. Marianne E Bronner, California Institute of Technology, United States

Version history

  1. Preprint posted: March 22, 2021 (view preprint)
  2. Received: March 30, 2021
  3. Accepted: January 27, 2022
  4. Accepted Manuscript published: January 28, 2022 (version 1)
  5. Version of Record published: March 9, 2022 (version 2)

Copyright

© 2022, Budjan 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

  • 4,351
    views
  • 558
    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. Christoph Budjan
  2. Shichen Liu
  3. Adrian Ranga
  4. Senjuti Gayen
  5. Olivier Pourquié
  6. Sahand Hormoz
(2022)
Paraxial mesoderm organoids model development of human somites
eLife 11:e68925.
https://doi.org/10.7554/eLife.68925

Share this article

https://doi.org/10.7554/eLife.68925

Further reading

    1. Chromosomes and Gene Expression
    2. Developmental Biology
    F Javier DeHaro-Arbona, Charalambos Roussos ... Sarah Bray
    Research Article

    Developmental programming involves the accurate conversion of signalling levels and dynamics to transcriptional outputs. The transcriptional relay in the Notch pathway relies on nuclear complexes containing the co-activator Mastermind (Mam). By tracking these complexes in real time, we reveal that they promote the formation of a dynamic transcription hub in Notch ON nuclei which concentrates key factors including the Mediator CDK module. The composition of the hub is labile and persists after Notch withdrawal conferring a memory that enables rapid reformation. Surprisingly, only a third of Notch ON hubs progress to a state with nascent transcription, which correlates with polymerase II and core Mediator recruitment. This probability is increased by a second signal. The discovery that target-gene transcription is probabilistic has far-reaching implications because it implies that stochastic differences in Notch pathway output can arise downstream of receptor activation.

    1. Developmental Biology
    Rieko Asai, Vivek N Prakash ... Takashi Mikawa
    Research Article

    Large-scale cell flow characterizes gastrulation in animal development. In amniote gastrulation, particularly in avian gastrula, a bilateral vortex-like counter-rotating cell flow, called ‘polonaise movements’, appears along the midline. Here, through experimental manipulations, we addressed relationships between the polonaise movements and morphogenesis of the primitive streak, the earliest midline structure in amniotes. Suppression of the Wnt/planar cell polarity (PCP) signaling pathway maintains the polonaise movements along a deformed primitive streak. Mitotic arrest leads to diminished extension and development of the primitive streak and maintains the early phase of the polonaise movements. Ectopically induced Vg1, an axis-inducing morphogen, generates the polonaise movements, aligned to the induced midline, but disturbs the stereotypical cell flow pattern at the authentic midline. Despite the altered cell flow, induction and extension of the primitive streak are preserved along both authentic and induced midlines. Finally, we show that ectopic axis-inducing morphogen, Vg1, is capable of initiating the polonaise movements without concomitant PS extension under mitotic arrest conditions. These results are consistent with a model wherein primitive streak morphogenesis is required for the maintenance of the polonaise movements, but the polonaise movements are not necessarily responsible for primitive streak morphogenesis. Our data describe a previously undefined relationship between the large-scale cell flow and midline morphogenesis in gastrulation.