­­­Kidney organoids recapitulate human basement membrane assembly in health and disease

  1. Mychel RPT Morais
  2. Pinyuan Tian
  3. Craig Lawless
  4. Syed Murtuza-Baker
  5. Louise Hopkinson
  6. Steven Woods
  7. Aleksandr Mironov
  8. David A Long
  9. Daniel P Gale
  10. Telma MT Zorn
  11. Susan J Kimber
  12. Roy Zent
  13. Rachel Lennon  Is a corresponding author
  1. University of Manchester, United Kingdom
  2. University College London, United Kingdom
  3. University of São Paulo, Brazil
  4. Vanderbilt University Medical Center, United States

Abstract

Basement membranes (BMs) are complex macromolecular networks underlying all continuous layers of cells. Essential components include collagen IV and laminins, which are affected by human genetic variants leading to a range of debilitating conditions including kidney, muscle, and cerebrovascular phenotypes. We investigated the dynamics of BM assembly in human pluripotent stem cell-derived kidney organoids. We resolved their global BM composition and discovered a conserved temporal sequence in BM assembly that paralleled mammalian fetal kidneys. We identified the emergence of key BM isoforms, which were altered by a pathogenic variant in COL4A5. Integrating organoid, fetal and adult kidney proteomes we found dynamic regulation of BM composition through development to adulthood, and with single-cell transcriptomic analysis we mapped the cellular origins of BM components. Overall, we define the complex and dynamic nature of kidney BM assembly and provide a platform for understanding its wider relevance in human development and disease.

Data availability

The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository (Perez-Riverol et al., 2019) with the dataset identifiers: PXD025838, PXD025874, PXD025911 and PXD026002.This project also contains the following source data hosted at:https://doi.org/10.6084/m9.figshare.c.5429628Figure 1 Original IF Images: B Whole-mount immunofluorescence for kidney cell types; F Representative whole mount immunofluorescence images of wild-type and Alport kidney organoids; G Immunofluorescence for LAMB2.Figure 1 Original light microscope Images: C Representative photomicrographs of day 18 kidney organoids (left) and human and mouse fetal kidneys (right).Figure 1 Original TEM Images: D Transmission electron micrographs of tubular BM in day 25 kidney organoid and E19 mouse fetal kidney.Figure 1 Original western blotting image: H Immunoblotting for LAMB2 using total lysates from wild-type and Alport organoids.Figure 2 Original IF Images: A Confocal immunofluorescence microscopy of wild-type kidney organoids; B perlecan and nidogen on days 11, 18 and 25 of differentiation.Figure 4 Original IF Images: A Immunofluorescence for key type IV collagen and laminin isoforms showing their emergence and distribution in kidney organoid BM; D Immunofluorescence for specific collagen IV isoforms in maturing glomeruli in E19 mouse kidney and in glomerular structures (indicated by dashed lines) in day 25 organoids.Figure 1-figure supplement 2A Original TEM photomicrographs: A Transmission electron microscopy of day 25 kidney organoids shows advanced differentiation of glomerular structures.Figure 1-figure supplement 2B Original TEM photomicrographs: B Transmission electron microscopy of day 25 kidney organoids shows advanced differentiation of glomerular structures.Figure 1-figure supplement 1C Original IF images: C Immunofluorescence for integrin beta-1 (ITGB1) in day 25 kidney organoid (wild-type). Anti-panlaminin or anti-collagen IV antibodies were used to label basement membranes.

The following data sets were generated
The following previously published data sets were used

Article and author information

Author details

  1. Mychel RPT Morais

    Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  2. Pinyuan Tian

    Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-6080-5378
  3. Craig Lawless

    Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  4. Syed Murtuza-Baker

    Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  5. Louise Hopkinson

    Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-1758-4201
  6. Steven Woods

    Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  7. Aleksandr Mironov

    Electron Microscopy Core Facility, University of Manchester, Manchester, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  8. David A Long

    Developmental Biology and Cancer Programme, 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:" 0000-0001-6580-3435
  9. Daniel P Gale

    Department of Renal Medicine, 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:" 0000-0002-9170-1579
  10. Telma MT Zorn

    Department of Cell and Developmental Biology, University of São Paulo, Sao Paulo, Brazil
    Competing interests
    The authors declare that no competing interests exist.
  11. Susan J Kimber

    Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  12. Roy Zent

    Department of Medicine, Vanderbilt University Medical Center, Nashville, United States
    Competing interests
    The authors declare that no competing interests exist.
  13. Rachel Lennon

    Division of Cell-Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, United Kingdom
    For correspondence
    Rachel.Lennon@manchester.ac.uk
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-6400-0227

Funding

Wellcome Trust (Wellcome Senior Fellowship award,202860/Z/16/Z)

  • Rachel Lennon

Kidney Research UK (Kidney Research UK grant (RP52/2014)

  • Pinyuan Tian
  • Rachel Lennon

São Paulo Research Foundation (Fellowship grants 2015/02535-2 and 2017/26785-5)

  • Mychel RPT Morais

Global Challenges Research Fund

  • Mychel RPT Morais

Veterans Affairs (Veterans Affairs Merit Awards 1I01BX002196-01 and DK069221)

  • Roy Zent

NIHR Biomedical Research Centre, Royal Marsden NHS Foundation Trust/Institute of Cancer Research

  • David A Long

Wellcome Trust (Investigator Award,220895/Z/20/Z)

  • David A Long

Medical Research Council (Project grants MR/P018629/1 and MR/J003638/1)

  • David A Long

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

Ethics

Animal experimentation: All mouse handling and experimental procedures were approved by the Animal EthicsCommittee of the Institute of Biomedical Sciences (University of São Paulo, Brazil; reference 019/2015). This was performed in accordance with recommendations from the current Brazilian legislation. All surgery was performed under avertin anaesthesia.

Human subjects: Human fetal kidney sections were provided by the Joint MRC/Wellcome Trust HumanDevelopmental Biology Resource (HDBR) (http://hdbr.org). The HDBR obtains written consent from the donors and has ethics approval (REC reference: 08/H0712/34+5) to collect, store and distribute human material sampled between 4 and 21 weeks post conception. All experimental protocols were approved by the Institute's Ethical Committee (reference 010/H0713/6) and were performed in accordance with institutional ethical and regulatory guidelines.

Copyright

© 2022, Morais 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

  • 4,063
    views
  • 747
    downloads
  • 25
    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. Mychel RPT Morais
  2. Pinyuan Tian
  3. Craig Lawless
  4. Syed Murtuza-Baker
  5. Louise Hopkinson
  6. Steven Woods
  7. Aleksandr Mironov
  8. David A Long
  9. Daniel P Gale
  10. Telma MT Zorn
  11. Susan J Kimber
  12. Roy Zent
  13. Rachel Lennon
(2022)
­­­Kidney organoids recapitulate human basement membrane assembly in health and disease
eLife 11:e73486.
https://doi.org/10.7554/eLife.73486

Share this article

https://doi.org/10.7554/eLife.73486

Further reading

    1. Neuroscience
    2. Stem Cells and Regenerative Medicine
    Amber R Philp, Carolina R Reyes ... Francisco J Rivera
    Short Report

    Revealing unknown cues that regulate oligodendrocyte progenitor cell (OPC) function in remyelination is important to optimise the development of regenerative therapies for multiple sclerosis (MS). Platelets are present in chronic non-remyelinated lesions of MS and an increase in circulating platelets has been described in experimental autoimmune encephalomyelitis (EAE) mice, an animal model for MS. However, the contribution of platelets to remyelination remains unexplored. Here we show platelet aggregation in proximity to OPCs in areas of experimental demyelination. Partial depletion of circulating platelets impaired OPC differentiation and remyelination, without altering blood-brain barrier stability and neuroinflammation. Transient exposure to platelets enhanced OPC differentiation in vitro, whereas sustained exposure suppressed this effect. In a mouse model of thrombocytosis (Calr+/-), there was a sustained increase in platelet aggregation together with a reduction of newly-generated oligodendrocytes following toxin-induced demyelination. These findings reveal a complex bimodal contribution of platelet to remyelination and provide insights into remyelination failure in MS.

    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.