A single-cell atlas of the mouse and human prostate reveals heterogeneity and conservation of epithelial progenitors

  1. Laura Crowley
  2. Francesco Cambuli
  3. Luis Aparicio
  4. Maho Shibata
  5. Brian D Robinson
  6. Shouhong Xuan
  7. Weiping Li
  8. Hanina Hibshoosh
  9. Massimo Loda
  10. Raul Rabadan  Is a corresponding author
  11. Michael M Shen  Is a corresponding author
  1. Columbia University Medical Center, United States
  2. Weill Cornell Medical Center, United States

Abstract

Understanding the cellular constituents of the prostate is essential for identifying the cell of origin for prostate adenocarcinoma. Here we describe a comprehensive single-cell atlas of the adult mouse prostate epithelium, which displays extensive heterogeneity. We observe distal lobe-specific luminal epithelial populations (LumA, LumD, LumL, and LumV), a proximally-enriched luminal population (LumP) that is not lobe-specific, and a periurethral population (PrU) that shares both basal and luminal features. Functional analyses suggest that LumP and PrU cells have multipotent progenitor activity in organoid formation and tissue reconstitution assays. Furthermore, we show that mouse distal and proximal luminal cells are most similar to human acinar and ductal populations, that a PrU-like population is conserved between species, and that the mouse lateral prostate is most similar to the human peripheral zone. Our findings elucidate new prostate epithelial progenitors, and help resolve long-standing questions about anatomical relationships between the mouse and human prostate.

Data availability

Single-cell RNA-sequencing data from this study have been deposited in the Gene Expression Omnibus (GEO) under the accession number GSE150692, and can also be accessed through the Broad Institute Single-Cell Portal (www.singlecell.broadinstitute.org).

The following data sets were generated

Article and author information

Author details

  1. Laura Crowley

    Medicine, Genetics and Development, Urology, and Systems Biology, Columbia University Medical Center, New York, United States
    Competing interests
    The authors declare that no competing interests exist.
  2. Francesco Cambuli

    Medicine, Genetics and Development, Urology, and Systems Biology, Columbia University Medical Center, New York, United States
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-8237-7121
  3. Luis Aparicio

    Systems Biology and Biomedical Informatics, Columbia University Medical Center, New York, United States
    Competing interests
    The authors declare that no competing interests exist.
  4. Maho Shibata

    Medicine, Genetics and Development, Urology, and Systems Biology, Columbia University Medical Center, New York, United States
    Competing interests
    The authors declare that no competing interests exist.
  5. Brian D Robinson

    Pathology and Laboratory Medicine, Weill Cornell Medical Center, New York, United States
    Competing interests
    The authors declare that no competing interests exist.
  6. Shouhong Xuan

    Department of Medicine, Columbia University Medical Center, New York, United States
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-0571-7855
  7. Weiping Li

    Medicine, Genetics and Development, Urology, and Systems Biology, Columbia University Medical Center, New York, United States
    Competing interests
    The authors declare that no competing interests exist.
  8. Hanina Hibshoosh

    Pathology and Cell Biology, Columbia University Medical Center, New York, United States
    Competing interests
    The authors declare that no competing interests exist.
  9. Massimo Loda

    Department of Pathology and Laboratory Medicin, Weill Cornell Medical Center, New York, United States
    Competing interests
    The authors declare that no competing interests exist.
  10. Raul Rabadan

    Department of Biomedical Informatics and Department of Systems Biology, College of Physicians & Surgeons, Columbia University Medical Center, New York, United States
    For correspondence
    rr2579@cumc.columbia.edu
    Competing interests
    The authors declare that no competing interests exist.
  11. Michael M Shen

    Medicine, Genetics and Development, Urology, and Systems Biology, Columbia University Medical Center, New York, United States
    For correspondence
    mshen@columbia.edu
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-4042-1657

Funding

National Cancer Institute (R01CA238005)

  • Michael M Shen

National Cancer Institute (U54CA193313)

  • Raul Rabadan
  • Michael M Shen

National Cancer Institute (P50CA211024)

  • Massimo Loda
  • Michael M Shen

National Cancer Institute (K99CA194287)

  • Maho Shibata

T.J. Martell Foundation

  • Michael M Shen

Department of Defense Prostate Cancer Research Program (W81XWH-18-1-0424)

  • Francesco Cambuli

National Science Foundation

  • Laura Crowley

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

Reviewing Editor

  1. Nima Sharifi, Cleveland Clinic, United States

Ethics

Animal experimentation: Animal studies were conducted according to protocols (AC-AABE0556, AC-AABG0564, AC-AABE5557) approved by the Columbia University Irving Medical Center (CUIMC) Institutional Animal Care and Use Committee (IACUC).

Human subjects: Human prostate tissue specimens were obtained from patients undergoing cystoprostatectomy for bladder cancer or radical prostatectomy at Columbia University Irving Medical Center or at Weill Cornell Medicine. Patients gave informed consent under an Institutional Review Board-approved protocol (AAAN8850).

Version history

  1. Received: May 29, 2020
  2. Accepted: September 10, 2020
  3. Accepted Manuscript published: September 11, 2020 (version 1)
  4. Version of Record published: October 1, 2020 (version 2)

Copyright

© 2020, Crowley 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

  • 8,978
    views
  • 1,149
    downloads
  • 71
    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. Laura Crowley
  2. Francesco Cambuli
  3. Luis Aparicio
  4. Maho Shibata
  5. Brian D Robinson
  6. Shouhong Xuan
  7. Weiping Li
  8. Hanina Hibshoosh
  9. Massimo Loda
  10. Raul Rabadan
  11. Michael M Shen
(2020)
A single-cell atlas of the mouse and human prostate reveals heterogeneity and conservation of epithelial progenitors
eLife 9:e59465.
https://doi.org/10.7554/eLife.59465

Share this article

https://doi.org/10.7554/eLife.59465

Further reading

    1. Cancer Biology
    2. Cell Biology
    Ian Lorimer
    Insight

    Establishing a zebrafish model of a deadly type of brain tumor highlights the role of the immune system in the early stages of the disease.

    1. Cancer Biology
    Xia Shen, Xiang Peng ... Chen-Ying Liu
    Research Article

    The role of processing bodies (P-bodies) in tumorigenesis and tumor progression is not well understood. Here, we showed that the oncogenes YAP/TAZ promote P-body formation in a series of cancer cell lines. Mechanistically, both transcriptional activation of the P-body-related genes SAMD4A, AJUBA, and WTIP and transcriptional suppression of the tumor suppressor gene PNRC1 are involved in enhancing the effects of YAP/TAZ on P-body formation in colorectal cancer (CRC) cells. By reexpression of PNRC1 or knockdown of P-body core genes (DDX6, DCP1A, and LSM14A), we determined that disruption of P-bodies attenuates cell proliferation, cell migration, and tumor growth induced by overexpression of YAP5SA in CRC. Analysis of a pancancer CRISPR screen database (DepMap) revealed co-dependencies between YAP/TEAD and the P-body core genes and correlations between the mRNA levels of SAMD4A, AJUBA, WTIP, PNRC1, and YAP target genes. Our study suggests that the P-body is a new downstream effector of YAP/TAZ, which implies that reexpression of PNRC1 or disruption of P-bodies is a potential therapeutic strategy for tumors with active YAP.