New insights into the function of Fascin in actin bundling: A combined theoretical and experimental study

https://doi.org/10.1016/j.biocel.2021.106056Get rights and content

Abstract

Fascin, one of actin bundling proteins, plays an important role in the cross-linking of actin filaments (F-actin). Phosphorylation of Fascin is an important posttranslational modification to affect its structure and function. For example, a phosphomimetic mutation of Fascin-S39D decrease its bundling ability with F-actin significantly. In this paper, we studied the actin-bundling activity of Fascin by using molecular dynamics (MD) simulations and biochemical methods. All single-site mutations from serine/threonine to aspartic acid were mimicked by MD simulations. For five mutants (S146D, S156D, S218D, T239D and S259D), the mutated residues in domain 2 of Fascin were found to form salt-bridge interactions with an adjacent residue, indicating that mutations of these residues could potentially reduce actin-bundling activity. Further, F-actin-bundling assays and immunofluorescence technique showed S146D and T239D to have a strong effect on Fascin bundling with F-actin. Finally, we show that single-site mutations do not change the general shape of Fascin, but local structures near the mutated residues in Fascin-S146D and T239D become unstable, thereby affecting the ability of Fascin to bind with F-actin. These findings suggest that targeting domain 2 of Fascin would be very useful for the drug design. In addition, our study indicates that MD simulation is a useful method to screening which residues on Fascin are important.

Introduction

According to the report of global cancer statistics 2018 (Bray et al., 2018), cancer has been among the top killer diseases in our society, owing to its rapidly growing incidence and high mortality. For cancer patients, tumor metastasis is the primary reason for cause of death (Hall, 2009). Thus, it is important to understand cancer cell migration and the mechanism of tumor metastasis to improve cancer treatment.

On the mechanism exploration of tumor metastasis, the formation of filopodia, as cell surface protrusions, is found to be very important to navigate its surroundings and determine the direction of migration. The filopodia is supported by actin filament network, which are rigid tightly bundling with actin-binding proteins, resulting in the filament bundles (Mattila and Lappalainen, 2008). Notably, Fascin, one of the actin cross-linking proteins, is detected in this process, indicating that Fascin interacts with F-actin (Otto et al., 1979). Also, expression of Fascin is up-regulated in various types of cancers to its overexpression correlates with increased tumor metastasis (Du et al., 2015; Hashimoto et al., 2005; Xie et al., 2010). In our previous studies, it also was found that expression of FSCN1 (gene of Fascin) is up-regulated in esophageal cancer (Wu et al., 2013, 2011). As a result, Fascin is used as a marker in cancer diagnosis and prognosis (Cao et al., 2014; Chen et al., 2017; Machesky and Li, 2010; Tan et al., 2014).

Fascin has a molecular mass of ∼55 kDa and functions as a monomer with fourβ-trefoil domains (Fig. 1a) (Yamashiro-Matsumura and Matsumura, 1985). All the charged groups located on the surface of Fascin are shown as balls in Fig. 1b. Fascin is required to facilitate the cross-linking of actin filaments into straight, compact, rigid bundles (Tilney et al., 1998). Biochemical characterization demonstrates that Fascin functions to generate the well-ordered parallel filaments (DeRosier and Edds, 1980) (Mattila and Lappalainen, 2008).

To understand how Fascin functions, many studies have been carried out to probe the actin-binding sites on Fascin (Chen et al., 2010; Sedeh et al., 2010). Early study by Ono and coworkers proposed that Fascin has two actin-binding sites to enable its bundling activity: one is located at the C terminus (domain 4, Fig. 1a, green) and the other one is at the N terminus (domain 1, Fig. 1a, blue), in which the latter binding site was confirmed by mutation of Ser39 (Ono et al., 1997). Later, Chen and coworkers found that both the N and C termini of Fascin contribute to the same binding site of actin based on their crystal structure (Chen et al., 2010). Importantly, they further confirmed this actin-binding site by experiments of binding macroketone (an inhibitor of actin-bundling) to Fascin and mutagenesis studies of residues His392, Lys471 and Ala488 of Fascin (Chen et al., 2010). In the study by Jansen et al., two multi-site mutants K22E/K43E/R100E/R109E and R271E/K353E/K358E of Fascin decreased the actin bundling activity, indicating that there exist two major actin-binding sites located in domains 1 and 3, respectively (Jansen et al., 2011). Furthermore, Yang and coworkers determined two major actin-binding sites and one possible site based on 100 mutants of Fascin (Yang et al., 2013). One of the two confirmed binding sites are locate between domains 1 and 4, and the other one is between domains 1 and 2 that is opposite to site 1. Additionally, another possible binding site is domain 3.

Further, the relation between structure and function of Fascin has been investigated by mutagenesis studies. The actin-bundling activity of the phosphomimetic S39D fascin mutant is significantly decreased within the phosphomimetic mutant of S39D (Chen et al., 2010; Ono et al., 1997). In a study by Yang and cowokers’ study, comparing the S39D mutant to the crystal structure of wild-type, several apparent conformational changes were found in the S39D mutant, namely inactive conformation (Yang et al., 2013). In addition, our recent study showed that other phosphomimetic mutations of Y23D, S38D and S274D of Fascin could also inhibit the filopodia formation, indicating that phosphorylation might play an important role in actin bundling by Fascin (Zeng et al., 2017).

On the other hand, the cryo-EM structure of actin filaments (F-actin) was also solved with some actin-bundling proteins (ABPs), e.g. tropomyosin, myosin, tropomodulin, cofilin (Tanaka et al., 2018) and filamin (Gurel et al., 2017; Iwamoto et al., 2018; Rao et al., 2014; von der Ecken et al., 2015). It has been found that, for F-actin, subdomains 2 and 4 (SD2 and SD4) of actin 3 (Ac3) interact with the SD3 of Ac5, and the SD3 of Ac3 interact with SD4 of Ac1 in the axial direction (Fig. 2). In the horizontal direction, SD1 and SD2 of Ac3 interact with the SD2 of Ac2 and SD2 of Ac4, respectively. In this way, each G-actin contacts with another 4 actins, and the contact area on each G-actin is ∼3500 Å2, which is ∼1/5 of total surface area of one G-actin (∼16,700 Å2). In addition, the formation of salt bridge on the interfaces between the F-actin and ABPs was found to be important for binding.

Although experimental studies have found some residues on Fascin are essential for the actin bundling, how these residues influence the structure is still not fully understood. Based on our previous studies, phosphorylation plays an important role in decreasing the actin-bundling function of Fascin (Zeng et al., 2017). In this paper, we further examine all the mutations that mimic phosphorylation with MD simulations, i.e. S → D and T → D. We focus mainly on the structural changes following single-site mutation. Finally, five mutants were selected to perform biochemical experiments for validation.

Section snippets

Results

We have studied the relation between single-site mutations of Fascin and their structure/function. First, we performed MD simulations for all the mutants that mimicked phosphorylation, i.e. S → D and T → D. Then, we analyzed how the mutations affect the structures. Finally, F-actin-bundling assays and immunofluorescence technique were carried out to verify our predictions.

Discussion

MD simulation is widely used to predict the important residues in biological study (Feng et al., 2014; Gur et al., 2018; Irani et al., 2013b; Wille et al., 2019; Zhao et al., 2015). It gives important insights into the dynamic information of proteins and the changes caused by mutation. Therefore, MD simulation offers a guidance for experimental study to design the biological assay. On the other hand, it is much faster than experimental study. For example, for a single-site mutation in

Molecular dynamics simulations

Calculations were based on the 1.8-Å crystal structure of Fascin from humans (PDB: 3LLP) (Chen et al., 2010). The Fascin-S39D uses another crystal structure of human Fascin (PDB: 4 GOV) (Yang et al., 2013). Because the biological unit of Fascin is a monomer, chain B of the crystal structure was chosen for our present MD simulations. All missing atoms in the experimental structure were automatically added by EasyModeller 4.0 (Bhusan et al., 2010).

All of the MD simulations were performed using

Data availability statement

The datasets generated for this study are available on request to the corresponding author.

Author contributions

All authors listed have made a substantial, direct, and intellectual contribution to the work and approved it for publication.

CRediT authorship contribution statement

Xiaodong Wu: Performing all MD simulations, most data collection and analysis, participate in writing original draft. Bing Wen: Performing experimental study, most data collection and analysis, participate in writing original draft. Lirui Lin: data analysis, participate in writing original draft. Wenqi Shi: Participate in the experiment, data collection. Dajia Li: Participate in the experiment, data collection. Yinwei Cheng: data analysis and participate in writing original draft. Li-Yan Xu:

Declaration of Competing Interest

The authors report no declarations of interest.

Acknowledgements

This investigation has been supported by grants from the National Natural Science Foundation of China (81872372, 81902469 and 21907063), Li Ka Shing Foundation(project LD0101) and 2020 Li Ka Shing Foundation Cross-Disciplinary Research Grant(2020LKSFG07B). We thank Dr. Stanley Li Lin, Department of Cell Biology and Genetics, Shantou University Medical College, for assistance in revising the manuscript.

References (56)

  • J.P. Ryckaert et al.

    Numerical integration of the Cartesian equations of motion of a system with constraints: molecular dynamics of n-alkanes

    J. Comput. Phys.

    (1977)
  • R.S. Sedeh et al.

    Structure, evolutionary conservation, and conformational dynamics of Homo sapiens fascin-1, an F-actin crosslinking protein

    J. Mol. Biol.

    (2010)
  • H. Wille et al.

    Combining molecular dynamics simulations and experimental analyses in protein misfolding

    Adv. Protein Chem. Struct. Biol.

    (2019)
  • X. Wu et al.

    Self-guided Langevin dynamics simulation method

    Chem. Phys. Lett.

    (2003)
  • J.J. Xie et al.

    Involvement of CYR61 and CTGF in the fascin-mediated proliferation and invasiveness of esophageal squamous cell carcinomas cells

    Am. J. Pathol.

    (2010)
  • Y. Yamakita et al.

    Phosphorylation of human Fascin inhibits its actin binding and bundling activities

    J. Biol. Chem.

    (1996)
  • S. Yamashiro-Matsumura et al.

    Purification and characterization of an F-actin-bundling 55-kilodalton protein from HeLa cells

    J. Biol. Chem.

    (1985)
  • S. Yang et al.

    Molecular mechanism of fascin function in filopodial formation

    J. Biol. Chem.

    (2013)
  • D.C. Bas et al.

    Very fast prediction and rationalization of pKa values for protein-ligand complexes

    Proteins

    (2008)
  • H. Berendsen et al.

    Molecular dynamics with coupling to an external bath

    J. Chem. Phys.

    (1984)
  • K.K. Bhusan et al.

    EasyModeller: a graphical interface to MODELLER

    BMC Res. Notes

    (2010)
  • F. Bray et al.

    Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries

    CA Cancer J. Clin.

    (2018)
  • H.H. Cao et al.

    A molecular prognostic model predicts esophageal squamous cell carcinoma prognosis

    PLoS One

    (2014)
  • D.A. Case et al.

    The FF14SB Force Field, AMBER 14 Reference Manual

    (2014)
  • D.A. Case et al.

    Amber 2018

    (2018)
  • L. Chen et al.

    Migrastatin analogues target fascin to block tumour metastasis

    Nature

    (2010)
  • T. Darden et al.

    Particle mesh Ewald: AnN⋅log(N) method for Ewald sums in large systems

    J. Chem. Phys.

    (1993)
  • Z.P. Du et al.

    Network analyses of gene expression following fascin knockdown in esophageal squamous cell carcinoma cells

    Asian Pac. J. Cancer Prev.

    (2015)
  • Cited by (7)

    • AKT serine/threonine kinase 2-mediated phosphorylation of fascin threonine 403 regulates esophageal cancer progression

      2022, International Journal of Biochemistry and Cell Biology
      Citation Excerpt :

      The mutant structure of the mimic Fascin threonine 403 phosphorylation (Fascin-T403D) was also obtained using Chimera. The experimental protocol has been described previously (Wu et al., 2021). For wild-type Fascin (Fascin-WT) and mimic Fascin threonine 403 phosphorylation (Fascin-T403D) simulation, we used the same simulation steps and conditions and ran triplicates.

    View all citing articles on Scopus
    1

    These authors have contributed equally to the investigation.

    View full text