MCPH1 inhibits condensin II during interphase by regulating its SMC2-kleisin interface

  1. Martin Houlard
  2. Erin E Cutts
  3. Muhammad S Shamim
  4. Jonathan Godwin
  5. David Weisz
  6. Aviva Presser Aiden
  7. Erez Lieberman-Aiden
  8. Lothar Schermelleh
  9. Kim Nasmyth
  10. Alessandro Vannini  Is a corresponding author
  1. University of Oxford, United Kingdom
  2. The Institute of Cancer Research, United Kingdom
  3. Baylor College of Medicine, United States
  4. Human Technopole, Italy

Abstract

The dramatic change in morphology of chromosomal DNAs between interphase and mitosis is one of the defining features of the eukaryotic cell cycle. Two types of enzymes, namely cohesin and condensin confer the topology of chromosomal DNA by extruding DNA loops. While condensin normally configures chromosomes exclusively during mitosis, cohesin does so during interphase. The processivity of cohesin’s loop extrusion during interphase is limited by a regulatory factor called WAPL, which induces cohesin to dissociate from chromosomes via a mechanism that requires dissociation of its kleisin from the neck of SMC3. We show here that a related mechanism may be responsible for blocking condensin II from acting during interphase. Cells derived from patients affected by microcephaly caused by mutations in the MCPH1 gene undergo premature chromosome condensation but it has never been established for certain whether MCPH1 regulates condensin II directly. We show that deletion of Mcph1 in mouse embryonic stem cells unleashes an activity of condensin II that triggers formation of compact chromosomes in G1 and G2 phases, which is accompanied by enhanced mixing of A and B chromatin compartments, and that this occurs even in the absence of CDK1 activity. Crucially, inhibition of condensin II by MCPH1 depends on the binding of a short linear motif within MCPH1 to condensin II's NCAPG2 subunit. We show that the activities of both Cohesin and Condensin II may be restricted during interphase by similar types of mechanisms as MCPH1's ability to block condensin II's association with chromatin is abrogated by the fusion of SMC2 with NCAPH2. Remarkably, in the absence of both WAPL and MCPH1, cohesin and condensin II transform chromosomal DNAs of G2 cells into chromosomes with a solenoidal axis showing that both cohesin and condensin must be tightly regulated to adjust the structure of chromatids for their successful segregation.

Data availability

HiC sequencing data has been deposited in GEO. (accession number: GSE188988)

The following data sets were generated

Article and author information

Author details

  1. Martin Houlard

    Department of Biochemistry, University of Oxford, Oxford, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  2. Erin E Cutts

    Division of Structural Biology, The Institute of Cancer Research, London, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-3290-4293
  3. Muhammad S Shamim

    Baylor College of Medicine, Houston, United States
    Competing interests
    The authors declare that no competing interests exist.
  4. Jonathan Godwin

    Department of Biochemistry, University of Oxford, Oxford, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  5. David Weisz

    Baylor College of Medicine, Houston, United States
    Competing interests
    The authors declare that no competing interests exist.
  6. Aviva Presser Aiden

    Baylor College of Medicine, Houston, United States
    Competing interests
    The authors declare that no competing interests exist.
  7. Erez Lieberman-Aiden

    Baylor College of Medicine, Houston, United States
    Competing interests
    The authors declare that no competing interests exist.
  8. Lothar Schermelleh

    Department of Biochemistry, University of Oxford, Oxford, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-1612-9699
  9. Kim Nasmyth

    Department of Biochemistry, University of Oxford, Oxford, United Kingdom
    Competing interests
    The authors declare that no competing interests exist.
  10. Alessandro Vannini

    Structural Biology Research Centre, Human Technopole, Milan, Italy
    For correspondence
    alessandro.vannini@fht.org
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-7212-5425

Funding

Wellcome Trust (107935/Z/15/Z)

  • Martin Houlard
  • Jonathan Godwin
  • Kim Nasmyth

McNair Medical Institute Scholar Award

  • Muhammad S Shamim
  • David Weisz
  • Aviva Presser Aiden
  • Erez Lieberman-Aiden

NIH Encyclopedia of DNA Elements Mapping Center Award (UM1HG009375)

  • Muhammad S Shamim
  • David Weisz
  • Aviva Presser Aiden
  • Erez Lieberman-Aiden

US-Israel Binational Science Foundation Award (2019276)

  • Muhammad S Shamim
  • David Weisz
  • Aviva Presser Aiden
  • Erez Lieberman-Aiden

Behavioral Plasticity Research Institute (NSF DBI-2021795)

  • Muhammad S Shamim
  • David Weisz
  • Aviva Presser Aiden
  • Erez Lieberman-Aiden

NSF Physics Frontiers Center Award (NSF PHY-2019745)

  • Muhammad S Shamim
  • Aviva Presser Aiden
  • Erez Lieberman-Aiden
  • Lothar Schermelleh

NIH CEGS Award (RM1HG011016-01A1)

  • Muhammad S Shamim
  • David Weisz
  • Aviva Presser Aiden
  • Erez Lieberman-Aiden

European Research Council (294401)

  • Martin Houlard
  • Jonathan Godwin
  • Kim Nasmyth

Cancer Research UK (26747)

  • Martin Houlard
  • Jonathan Godwin
  • Kim Nasmyth

Wellcome Trust (107457/Z/15/Z)

  • Lothar Schermelleh

Wellcome Trust (091911)

  • Lothar Schermelleh

Paul and Daisy Soros Foundation

  • Muhammad S Shamim

Cancer Research UK (CR-UK C47547/A21536)

  • Erin E Cutts
  • Alessandro Vannini

Wellcome Trust (200818/Z/16/Z)

  • Erin E Cutts
  • Alessandro Vannini

Welch Foundation (Q-1866)

  • Muhammad S Shamim
  • David Weisz
  • Aviva Presser Aiden
  • Erez Lieberman-Aiden

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

Reviewing Editor

  1. Adèle L Marston, University of Edinburgh, United Kingdom

Ethics

Animal experimentation: This study was performed in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. All of the animals were handled according to approved institutional animal care and use committee (IACUC) protocols (#08-133) of the University of Arizona. The protocol was approved by the Committee on the Ethics of Animal Experiments of the University of Minnesota (Permit Number: 27-2956). All surgery was performed under sodium pentobarbital anesthesia, and every effort was made to minimize suffering.

Version history

  1. Preprint posted: July 20, 2021 (view preprint)
  2. Received: August 25, 2021
  3. Accepted: November 8, 2021
  4. Accepted Manuscript published: December 1, 2021 (version 1)
  5. Version of Record published: December 14, 2021 (version 2)

Copyright

© 2021, Houlard 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

  • 2,445
    views
  • 381
    downloads
  • 21
    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. Martin Houlard
  2. Erin E Cutts
  3. Muhammad S Shamim
  4. Jonathan Godwin
  5. David Weisz
  6. Aviva Presser Aiden
  7. Erez Lieberman-Aiden
  8. Lothar Schermelleh
  9. Kim Nasmyth
  10. Alessandro Vannini
(2021)
MCPH1 inhibits condensin II during interphase by regulating its SMC2-kleisin interface
eLife 10:e73348.
https://doi.org/10.7554/eLife.73348

Share this article

https://doi.org/10.7554/eLife.73348

Further reading

    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.

    1. Biochemistry and Chemical Biology
    2. Structural Biology and Molecular Biophysics
    Isabelle Petit-Hartlein, Annelise Vermot ... Franck Fieschi
    Research Article

    NADPH oxidases (NOX) are transmembrane proteins, widely spread in eukaryotes and prokaryotes, that produce reactive oxygen species (ROS). Eukaryotes use the ROS products for innate immune defense and signaling in critical (patho)physiological processes. Despite the recent structures of human NOX isoforms, the activation of electron transfer remains incompletely understood. SpNOX, a homolog from Streptococcus pneumoniae, can serves as a robust model for exploring electron transfers in the NOX family thanks to its constitutive activity. Crystal structures of SpNOX full-length and dehydrogenase (DH) domain constructs are revealed here. The isolated DH domain acts as a flavin reductase, and both constructs use either NADPH or NADH as substrate. Our findings suggest that hydride transfer from NAD(P)H to FAD is the rate-limiting step in electron transfer. We identify significance of F397 in nicotinamide access to flavin isoalloxazine and confirm flavin binding contributions from both DH and Transmembrane (TM) domains. Comparison with related enzymes suggests that distal access to heme may influence the final electron acceptor, while the relative position of DH and TM does not necessarily correlate with activity, contrary to previous suggestions. It rather suggests requirement of an internal rearrangement, within the DH domain, to switch from a resting to an active state. Thus, SpNOX appears to be a good model of active NOX2, which allows us to propose an explanation for NOX2’s requirement for activation.