Abstract

Disruption of the sumoylation/desumoylation equilibrium is associated with several disease states such as cancer and infections, however the mechanisms regulating the global SUMO balance remain poorly defined. Here, we show that infection by Shigella flexneri, the causative agent of human bacillary dysentery, switches off host sumoylation during epithelial cell infection in vitro and in vivo and that this effect is mainly mediated by a calcium/calpain-induced cleavage of the SUMO E1 enzyme SAE2, thus leading to sumoylation inhibition. Furthermore, we describe a mechanism by which Shigella promotes its own invasion by altering the sumoylation state of RhoGDIa, a master negative regulator of RhoGTPase activity and actin polymerization. Together, our data suggest that SUMO modification is essential to restrain pathogenic bacterial entry by limiting cytoskeletal rearrangement induced by bacterial effectors. Moreover, these findings identify calcium-activated calpains as powerful modulators of cellular sumoylation levels with potentially broad implications in several physiological and pathological situations.

Article and author information

Author details

  1. Pierre Lapaquette

    Nuclear Organization and Oncogenesis, Institut Pasteur, Paris, France
    Competing interests
    The authors declare that no competing interests exist.
  2. Sabrina Fritah

    Nuclear Organization and Oncogenesis, Institut Pasteur, Paris, France
    Competing interests
    The authors declare that no competing interests exist.
  3. Nouara Lhocine

    Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France
    Competing interests
    The authors declare that no competing interests exist.
  4. Alexandra Andrieux

    Nuclear Organization and Oncogenesis, Institut Pasteur, Paris, France
    Competing interests
    The authors declare that no competing interests exist.
  5. Giulia Nigro

    Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France
    Competing interests
    The authors declare that no competing interests exist.
  6. Joëlle Mounier

    Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France
    Competing interests
    The authors declare that no competing interests exist.
  7. Philippe Sansonetti

    Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France
    Competing interests
    The authors declare that no competing interests exist.
  8. Anne Dejean

    Nuclear Organization and Oncogenesis, Institut Pasteur, Paris, France
    For correspondence
    anne.dejean@pasteur.fr
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-4778-6840

Funding

Ligue Contre le Cancer (Post-doc fellowship to P. Lapaquette and labelled team to A. Dejean)

  • Pierre Lapaquette
  • Anne Dejean

Institut Pasteur

  • Philippe Sansonetti
  • Anne Dejean

Institut National Du Cancer

  • Anne Dejean

European Research Council (SUMOSTRESS)

  • Anne Dejean

Sidaction

  • Sabrina Fritah

Odyssey RE

  • Anne Dejean

Institut National de la Santé et de la Recherche Médicale

  • Philippe Sansonetti
  • Anne Dejean

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

Reviewing Editor

  1. Dominique Soldati-Favre, University of Geneva, Switzerland

Ethics

Animal experimentation: Animal experiments were performed accordingly to the guidelines of the Institut Pasteur's ethical committee for animal use in research (CETEA number 2013-0028).

Version history

  1. Received: April 6, 2017
  2. Accepted: December 11, 2017
  3. Accepted Manuscript published: December 12, 2017 (version 1)
  4. Accepted Manuscript updated: December 13, 2017 (version 2)
  5. Accepted Manuscript updated: December 14, 2017 (version 3)
  6. Version of Record published: December 27, 2017 (version 4)

Copyright

© 2017, Lapaquette 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

  • 1,508
    views
  • 284
    downloads
  • 17
    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. Pierre Lapaquette
  2. Sabrina Fritah
  3. Nouara Lhocine
  4. Alexandra Andrieux
  5. Giulia Nigro
  6. Joëlle Mounier
  7. Philippe Sansonetti
  8. Anne Dejean
(2017)
Shigella entry unveils a calcium/calpain-dependent mechanism for inhibiting sumoylation
eLife 6:e27444.
https://doi.org/10.7554/eLife.27444

Share this article

https://doi.org/10.7554/eLife.27444

Further reading

    1. Cell Biology
    Mathieu C Husser, Nhat P Pham ... Alisa Piekny
    Tools and Resources

    Endogenous tags have become invaluable tools to visualize and study native proteins in live cells. However, generating human cell lines carrying endogenous tags is difficult due to the low efficiency of homology-directed repair. Recently, an engineered split mNeonGreen protein was used to generate a large-scale endogenous tag library in HEK293 cells. Using split mNeonGreen for large-scale endogenous tagging in human iPSCs would open the door to studying protein function in healthy cells and across differentiated cell types. We engineered an iPS cell line to express the large fragment of the split mNeonGreen protein (mNG21-10) and showed that it enables fast and efficient endogenous tagging of proteins with the short fragment (mNG211). We also demonstrate that neural network-based image restoration enables live imaging studies of highly dynamic cellular processes such as cytokinesis in iPSCs. This work represents the first step towards a genome-wide endogenous tag library in human stem cells.

    1. Biochemistry and Chemical Biology
    2. Cell Biology
    Natalia Dolgova, Eva-Maria E Uhlemann ... Oleg Y Dmitriev
    Research Article

    Mediator of ERBB2-driven Cell Motility 1 (MEMO1) is an evolutionary conserved protein implicated in many biological processes; however, its primary molecular function remains unknown. Importantly, MEMO1 is overexpressed in many types of cancer and was shown to modulate breast cancer metastasis through altered cell motility. To better understand the function of MEMO1 in cancer cells, we analyzed genetic interactions of MEMO1 using gene essentiality data from 1028 cancer cell lines and found multiple iron-related genes exhibiting genetic relationships with MEMO1. We experimentally confirmed several interactions between MEMO1 and iron-related proteins in living cells, most notably, transferrin receptor 2 (TFR2), mitoferrin-2 (SLC25A28), and the global iron response regulator IRP1 (ACO1). These interactions indicate that cells with high MEMO1 expression levels are hypersensitive to the disruptions in iron distribution. Our data also indicate that MEMO1 is involved in ferroptosis and is linked to iron supply to mitochondria. We have found that purified MEMO1 binds iron with high affinity under redox conditions mimicking intracellular environment and solved MEMO1 structures in complex with iron and copper. Our work reveals that the iron coordination mode in MEMO1 is very similar to that of iron-containing extradiol dioxygenases, which also display a similar structural fold. We conclude that MEMO1 is an iron-binding protein that modulates iron homeostasis in cancer cells.