Activation of a nucleotide-dependent RCK domain requires binding of a cation cofactor to a conserved site

  1. Celso M Teixeira-Duarte
  2. Fátima Fonseca
  3. João H Morais Cabral  Is a corresponding author
  1. i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal

Abstract

RCK domains regulate the activity of K+ channels and transporters in eukaryotic and prokaryotic organisms by responding to ions or nucleotides. The mechanisms of RCK activation by Ca2+ in the eukaryotic BK and bacterial MthK K+ channels are well understood. However, the molecular details of activation in nucleotide-dependent RCK domains are not clear. Through a functional and structural analysis of the mechanism of ATP activation in KtrA, a RCK domain from the B. subtilis KtrAB cation channel, we have found that activation by nucleotide requires binding of cations to an intra-dimer interface site in the RCK dimer. In particular, divalent cations are coordinated by the γ-phosphates of bound-ATP, tethering the two subunits and stabilizing the active state conformation. Strikingly, the binding site residues are highly conserved in many different nucleotide-dependent RCK domains, indicating that divalent cations are a general cofactor in the regulatory mechanism of many nucleotide-dependent RCK domains.

Data availability

Diffraction data have been deposited in PDB under the accession code 6S2J, 6S5B, 6S5D, 6S7R, 6S5N, 6S5O, 6S5E, 6S5G,6S5C.Source data files have been provided for Figures 6, 7, 8 and 9.

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

Article and author information

Author details

  1. Celso M Teixeira-Duarte

    i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-8036-7069
  2. Fátima Fonseca

    i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-7979-5907
  3. João H Morais Cabral

    i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
    For correspondence
    jcabral@ibmc.up.pt
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-4461-9716

Funding

Fundação Luso-Americana para o Desenvolvimento (FLAD Life Science 2020)

  • João H Morais Cabral

Fundação para a Ciência e a Tecnologia (POCI-01-0145-FEDER-029863 (PTDC/BIA-BQM/29863/2017))

  • João H Morais Cabral

Fundação para a Ciência e a Tecnologia (POCI-01-0145-FEDER-007274)

  • João H Morais Cabral

Fundação para a Ciência e a Tecnologia (SFRH/BD/123761/2016)

  • Celso M Teixeira-Duarte

Fundação para a Ciência e a Tecnologia (SFRH/BPD/102753/2014)

  • Fátima Fonseca

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

Reviewing Editor

  1. László Csanády, Semmelweis University, Hungary

Version history

  1. Received: July 29, 2019
  2. Accepted: December 22, 2019
  3. Accepted Manuscript published: December 23, 2019 (version 1)
  4. Version of Record published: January 13, 2020 (version 2)

Copyright

© 2019, Teixeira-Duarte 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

  • 1,039
    views
  • 177
    downloads
  • 11
    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. Celso M Teixeira-Duarte
  2. Fátima Fonseca
  3. João H Morais Cabral
(2019)
Activation of a nucleotide-dependent RCK domain requires binding of a cation cofactor to a conserved site
eLife 8:e50661.
https://doi.org/10.7554/eLife.50661

Share this article

https://doi.org/10.7554/eLife.50661

Further reading

    1. Biochemistry and Chemical Biology
    2. Structural Biology and Molecular Biophysics
    Amy H Andreotti, Volker Dötsch
    Editorial

    The articles in this special issue highlight how modern cellular, biochemical, biophysical and computational techniques are allowing deeper and more detailed studies of allosteric kinase regulation.

    1. Developmental Biology
    2. Structural Biology and Molecular Biophysics
    Samuel C Griffiths, Jia Tan ... Hsin-Yi Henry Ho
    Research Article Updated

    The receptor tyrosine kinase ROR2 mediates noncanonical WNT5A signaling to orchestrate tissue morphogenetic processes, and dysfunction of the pathway causes Robinow syndrome, brachydactyly B, and metastatic diseases. The domain(s) and mechanisms required for ROR2 function, however, remain unclear. We solved the crystal structure of the extracellular cysteine-rich (CRD) and Kringle (Kr) domains of ROR2 and found that, unlike other CRDs, the ROR2 CRD lacks the signature hydrophobic pocket that binds lipids/lipid-modified proteins, such as WNTs, suggesting a novel mechanism of ligand reception. Functionally, we showed that the ROR2 CRD, but not other domains, is required and minimally sufficient to promote WNT5A signaling, and Robinow mutations in the CRD and the adjacent Kr impair ROR2 secretion and function. Moreover, using function-activating and -perturbing antibodies against the Frizzled (FZ) family of WNT receptors, we demonstrate the involvement of FZ in WNT5A-ROR signaling. Thus, ROR2 acts via its CRD to potentiate the function of a receptor super-complex that includes FZ to transduce WNT5A signals.