Complex structures of Rsu1 and PINCH1 reveal a regulatory mechanism of the ILK/PINCH/Parvin complex for F-actin dynamics

  1. Haibin Yang
  2. Leishu Lin
  3. Kan Sun
  4. Ting Zhang
  5. Wan Chen
  6. Lianghui Li
  7. Yuchen Xie
  8. Chuanyue Wu  Is a corresponding author
  9. Zhiyi Wei  Is a corresponding author
  10. Cong Yu  Is a corresponding author
  1. Southern University of Science and Technology, China, China
  2. University of Pittsburgh, United States

Abstract

Communications between actin filaments and integrin-mediated focal adhesion (FA) are crucial for cell adhesion and migration. As a core platform to organize FA proteins, the tripartite ILK/PINCH/Parvin (IPP) complex interacts with actin filaments to regulate the cytoskeleton-FA crosstalk. Rsu1, a Ras suppressor, is enriched in FA through PINCH1 and plays important roles in regulating F-actin structures. Here, we solved crystal structures of the Rsu1/PINCH1 complex, in which the Leucine-Rich-Repeats of Rsu1 form a solenoid structure to tightly associate with the C-terminal region of PINCH1. Further structural analysis uncovered that the interaction between Rsu1 and PINCH1 blocks the IPP-mediated F-actin bundling by disrupting the binding of PINCH1 to actin. Consistently, overexpressing Rsu1 in HeLa cells impairs stress fiber formation and cell spreading. Together, our findings demonstrated that Rsu1 is critical for tuning the communication between F-actin and FA by interacting with the IPP complex and negatively modulating the F-actin bundling.

Data availability

Diffraction data have been deposited in PDB under the accession code 7D2S, 7D2T and 7D2U.

The following data sets were generated

Article and author information

Author details

  1. Haibin Yang

    Department of Biology, Southern University of Science and Technology, China, Shenzhen, China
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-6902-6941
  2. Leishu Lin

    Department of Biology, Southern University of Science and Technology, China, Shenzhen, China
    Competing interests
    The authors declare that no competing interests exist.
  3. Kan Sun

    Department of Biology, Southern University of Science and Technology, China, Shenzhen, China
    Competing interests
    The authors declare that no competing interests exist.
  4. Ting Zhang

    Department of Biology, Southern University of Science and Technology, China, Shenzhen, China
    Competing interests
    The authors declare that no competing interests exist.
  5. Wan Chen

    Department of Biology, Southern University of Science and Technology, China, Shenzhen, China
    Competing interests
    The authors declare that no competing interests exist.
  6. Lianghui Li

    Department of Biology, Southern University of Science and Technology, China, Shenzhen, China
    Competing interests
    The authors declare that no competing interests exist.
  7. Yuchen Xie

    Department of Biology, Southern University of Science and Technology, China, Shenzhen, China
    Competing interests
    The authors declare that no competing interests exist.
  8. Chuanyue Wu

    Department of Pathology, University of Pittsburgh, Pittsburgh, United States
    For correspondence
    carywu@pitt.edu
    Competing interests
    The authors declare that no competing interests exist.
  9. Zhiyi Wei

    Department of Biology, Southern University of Science and Technology, China, Shenzhen, China
    For correspondence
    weizy@sustech.edu.cn
    Competing interests
    The authors declare that no competing interests exist.
  10. Cong Yu

    Department of Biology, Southern University of Science and Technology, China, Shenzhen, China
    For correspondence
    yuc@sustech.edu.cn
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-2912-6347

Funding

National Natural Science Foundation of China (31870757)

  • Cong Yu

National Natural Science Foundation of China (31970741)

  • Zhiyi Wei

National Natural Science Foundation of China (31770791)

  • Zhiyi Wei

Science and Technology Planning Project of Guangdong Province (2017B030301018)

  • Cong Yu

Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Fundamental Research Institutions (2019SHIBS0002)

  • Zhiyi Wei

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

Reviewing Editor

  1. Reinhard Fässler, Max Planck Institute of Biochemistry, Germany

Version history

  1. Received: October 27, 2020
  2. Accepted: February 12, 2021
  3. Accepted Manuscript published: February 15, 2021 (version 1)
  4. Version of Record published: February 26, 2021 (version 2)
  5. Version of Record updated: March 8, 2021 (version 3)

Copyright

© 2021, Yang 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

  • 1,249
    views
  • 204
    downloads
  • 8
    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. Haibin Yang
  2. Leishu Lin
  3. Kan Sun
  4. Ting Zhang
  5. Wan Chen
  6. Lianghui Li
  7. Yuchen Xie
  8. Chuanyue Wu
  9. Zhiyi Wei
  10. Cong Yu
(2021)
Complex structures of Rsu1 and PINCH1 reveal a regulatory mechanism of the ILK/PINCH/Parvin complex for F-actin dynamics
eLife 10:e64395.
https://doi.org/10.7554/eLife.64395

Share this article

https://doi.org/10.7554/eLife.64395

Further reading

    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.

    1. Biochemistry and Chemical Biology
    2. Plant Biology
    Dietmar Funck, Malte Sinn ... Jörg S Hartig
    Research Article

    Metabolism and biological functions of the nitrogen-rich compound guanidine have long been neglected. The discovery of four classes of guanidine-sensing riboswitches and two pathways for guanidine degradation in bacteria hint at widespread sources of unconjugated guanidine in nature. So far, only three enzymes from a narrow range of bacteria and fungi have been shown to produce guanidine, with the ethylene-forming enzyme (EFE) as the most prominent example. Here, we show that a related class of Fe2+- and 2-oxoglutarate-dependent dioxygenases (2-ODD-C23) highly conserved among plants and algae catalyze the hydroxylation of homoarginine at the C6-position. Spontaneous decay of 6-hydroxyhomoarginine yields guanidine and 2-aminoadipate-6-semialdehyde. The latter can be reduced to pipecolate by pyrroline-5-carboxylate reductase but more likely is oxidized to aminoadipate by aldehyde dehydrogenase ALDH7B in vivo. Arabidopsis has three 2-ODD-C23 isoforms, among which Din11 is unusual because it also accepted arginine as substrate, which was not the case for the other 2-ODD-C23 isoforms from Arabidopsis or other plants. In contrast to EFE, none of the three Arabidopsis enzymes produced ethylene. Guanidine contents were typically between 10 and 20 nmol*(g fresh weight)-1 in Arabidopsis but increased to 100 or 300 nmol*(g fresh weight)-1 after homoarginine feeding or treatment with Din11-inducing methyljasmonate, respectively. In 2-ODD-C23 triple mutants, the guanidine content was strongly reduced, whereas it increased in overexpression plants. We discuss the implications of the finding of widespread guanidine-producing enzymes in photosynthetic eukaryotes as a so far underestimated branch of the bio-geochemical nitrogen cycle and propose possible functions of natural guanidine production.