The neuronal calcium sensor NCS-1 regulates the phosphorylation state and activity of the Ga chaperone and GEF Ric-8A

  1. Daniel Muñoz-Reyes
  2. Levi J McClelland
  3. Sandra Arroyo-Urea
  4. Sonia Sánchez-Yepes
  5. Juan Sabín
  6. Sara Pérez-Suárez
  7. Margarita Menendez
  8. Alicia Mansilla
  9. Javier García-Nafría
  10. Stephen Sprang
  11. Maria Jose Sanchez-Barrena  Is a corresponding author
  1. Institute of Physical-Chemistry Blas Cabrera, Spain
  2. University of Montana, United States
  3. University of Zaragoza, Spain
  4. Hospital Universitario Ramón y Cajal, Spain
  5. Software 4 Science Developments, Spain
  6. Institute of Physical-Chemistry, Spain
  7. Institute of Physical Chemistry Blas Cabrera, Spain

Abstract

The Neuronal Calcium Sensor 1, an EF-hand Ca2+ binding protein, and Ric-8A coregulate synapse number and probability of neurotransmitter release. Recently, the structures of Ric-8A bound to Ga have revealed how Ric-8A phosphorylation promotes Ga recognition and activity as a chaperone and guanine nucleotide exchange factor. However, the molecular mechanism by which NCS-1 regulates Ric-8A activity and its interaction with Ga subunits is not well understood. Given the interest in the NCS-1/Ric-8A complex as a therapeutic target in nervous system disorders, it is necessary to shed light on this molecular mechanism of action at atomic level. We have reconstituted NCS-1/Ric-8A complexes to conduct a multimodal approach and determine the sequence of Ca2+ signals and phosphorylation events that promote the interaction of Ric-8A with Ga. Our data show that the binding of NCS-1 and Ga to Ric-8A are mutually exclusive. Importantly, NCS-1 induces a structural rearrangement in Ric-8A that traps the protein in a conformational state that is inaccessible to Casein Kinase II-mediated phosphorylation, demonstrating one aspect of its negative regulation of Ric-8A-mediated G-protein signaling. Functional experiments indicate a loss of Ric-8A GEF activity towards Ga when complexed with NCS-1, and restoration of nucleotide exchange activity upon increasing Ca2+ concentration. Finally, the high-resolution crystallographic data reported here define the NCS-1/Ric-8A interface and will allow the development of therapeutic synapse function regulators with improved activity and selectivity.

Data availability

The atomic coordinates and structure factors have been deposited in the Protein Data Bank, https://www.pdb.org/. PDB with codes: Structure 1 (8ALH), Structure 2 (8AHY), Structure 3 (8ALM).All data generated or analysed during this study are included in the manuscript and supporting file; Source Data files have been provided for Figure 2, Figure 3, Figure 4, Figure 5

Article and author information

Author details

  1. Daniel Muñoz-Reyes

    Department of Crystallography and Structural Biology, Institute of Physical-Chemistry Blas Cabrera, Madrid, Spain
    Competing interests
    The authors declare that no competing interests exist.
  2. Levi J McClelland

    Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, United States
    Competing interests
    The authors declare that no competing interests exist.
  3. Sandra Arroyo-Urea

    Institute for Biocomputation and Physics of Complex Systems, University of Zaragoza, Zaragoza, Spain
    Competing interests
    The authors declare that no competing interests exist.
  4. Sonia Sánchez-Yepes

    Department of Neurobiology, Hospital Universitario Ramón y Cajal, Madrid, Spain
    Competing interests
    The authors declare that no competing interests exist.
  5. Juan Sabín

    AFFINImeter Scientific & Development, Software 4 Science Developments, Santiago de Compostela, Spain
    Competing interests
    The authors declare that no competing interests exist.
  6. Sara Pérez-Suárez

    Department of Crystallography and Structural Biology, Institute of Physical-Chemistry Blas Cabrera, Madrid, Spain
    Competing interests
    The authors declare that no competing interests exist.
  7. Margarita Menendez

    Department of Biological Physical-Chemisty, Institute of Physical-Chemistry, Madrid, Spain
    Competing interests
    The authors declare that no competing interests exist.
  8. Alicia Mansilla

    Department of Neurobiology, Hospital Universitario Ramón y Cajal, Madrid, Spain
    Competing interests
    The authors declare that no competing interests exist.
  9. Javier García-Nafría

    Institute for Biocomputation and Physics of Complex Systems, University of Zaragoza, Zaragoza, Spain
    Competing interests
    The authors declare that no competing interests exist.
  10. Stephen Sprang

    Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, United States
    Competing interests
    The authors declare that no competing interests exist.
  11. Maria Jose Sanchez-Barrena

    Department of Crystallography and Structural Biology, Institute of Physical Chemistry Blas Cabrera, Madrid, Spain
    For correspondence
    xmjose@iqfr.csic.es
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-5986-1804

Funding

Spanish National Plan for Scientific and Technical Research and Innovation (PID2019-111737RB-I00)

  • Maria Jose Sanchez-Barrena

Spanish National Plan for Scientific and Technical Research and Innovation (PID2019-106608RB-I00)

  • Alicia Mansilla

Spanish National Plan for Scientific and Technical Research and Innovation (PDC2022-133775-I00)

  • Alicia Mansilla

Spanish National Plan for Scientific and Technical Research and Innovation (RTI2018-099985-B-I00)

  • Margarita Menendez

Spanish National Plan for Scientific and Technical Research and Innovation (PID2020-113359GA-I00)

  • Javier García-Nafría

Spanish National Plan for Scientific and Technical Research and Innovation (RYC-2017-22392)

  • Alicia Mansilla

Spanish National Plan for Scientific and Technical Research and Innovation (RYC2018-025731-I)

  • Javier García-Nafría

National Institutes of Health (P30GM140963)

  • Stephen Sprang

National Institutes of Health (R01GM105993)

  • Stephen Sprang

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

Reviewing Editor

  1. Randy B Stockbridge, University of Michigan, United States

Version history

  1. Preprint posted: December 10, 2022 (view preprint)
  2. Received: January 12, 2023
  3. Accepted: November 24, 2023
  4. Accepted Manuscript published: November 29, 2023 (version 1)
  5. Version of Record published: December 20, 2023 (version 2)

Copyright

© 2023, Muñoz-Reyes 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

  • 828
    views
  • 162
    downloads
  • 1
    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. Daniel Muñoz-Reyes
  2. Levi J McClelland
  3. Sandra Arroyo-Urea
  4. Sonia Sánchez-Yepes
  5. Juan Sabín
  6. Sara Pérez-Suárez
  7. Margarita Menendez
  8. Alicia Mansilla
  9. Javier García-Nafría
  10. Stephen Sprang
  11. Maria Jose Sanchez-Barrena
(2023)
The neuronal calcium sensor NCS-1 regulates the phosphorylation state and activity of the Ga chaperone and GEF Ric-8A
eLife 12:e86151.
https://doi.org/10.7554/eLife.86151

Share this article

https://doi.org/10.7554/eLife.86151

Further reading

    1. Biochemistry and Chemical Biology
    2. Cell Biology
    Ya-Juan Wang, Xiao-Jing Di ... Ting-Wei Mu
    Research Article

    Protein homeostasis (proteostasis) deficiency is an important contributing factor to neurological and metabolic diseases. However, how the proteostasis network orchestrates the folding and assembly of multi-subunit membrane proteins is poorly understood. Previous proteomics studies identified Hsp47 (Gene: SERPINH1), a heat shock protein in the endoplasmic reticulum lumen, as the most enriched interacting chaperone for gamma-aminobutyric type A (GABAA) receptors. Here, we show that Hsp47 enhances the functional surface expression of GABAA receptors in rat neurons and human HEK293T cells. Furthermore, molecular mechanism study demonstrates that Hsp47 acts after BiP (Gene: HSPA5) and preferentially binds the folded conformation of GABAA receptors without inducing the unfolded protein response in HEK293T cells. Therefore, Hsp47 promotes the subunit-subunit interaction, the receptor assembly process, and the anterograde trafficking of GABAA receptors. Overexpressing Hsp47 is sufficient to correct the surface expression and function of epilepsy-associated GABAA receptor variants in HEK293T cells. Hsp47 also promotes the surface trafficking of other Cys-loop receptors, including nicotinic acetylcholine receptors and serotonin type 3 receptors in HEK293T cells. Therefore, in addition to its known function as a collagen chaperone, this work establishes that Hsp47 plays a critical and general role in the maturation of multi-subunit Cys-loop neuroreceptors.

    1. Biochemistry and Chemical Biology
    Hao Wang, Chen Ye ... Yan Li
    Research Article

    Bacterial exonuclease III (ExoIII), widely acknowledged for specifically targeting double-stranded DNA (dsDNA), has been documented as a DNA repair-associated nuclease with apurinic/apyrimidinic (AP)-endonuclease and 3′→5′ exonuclease activities. Due to these enzymatic properties, ExoIII has been broadly applied in molecular biosensors. Here, we demonstrate that ExoIII (Escherichia coli) possesses highly active enzymatic activities on ssDNA. By using a range of ssDNA fluorescence-quenching reporters and fluorophore-labeled probes coupled with mass spectrometry analysis, we found ExoIII cleaved the ssDNA at 5′-bond of phosphodiester from 3′ to 5′ end by both exonuclease and endonuclease activities. Additional point mutation analysis identified the critical residues for the ssDNase action of ExoIII and suggested the activity shared the same active center with the dsDNA-targeted activities of ExoIII. Notably, ExoIII could also digest the dsDNA structures containing 3′-end ssDNA. Considering most ExoIII-assisted molecular biosensors require the involvement of single-stranded DNA (ssDNA) or nucleic acid aptamer containing ssDNA, the activity will lead to low efficiency or false positive outcome. Our study revealed the multi-enzymatic activity and the underlying molecular mechanism of ExoIII on ssDNA, illuminating novel insights for understanding its biological roles in DNA repair and the rational design of ExoIII-ssDNA involved diagnostics.