Elsevier

Cellular Signalling

Volume 28, Issue 5, May 2016, Pages 448-459
Cellular Signalling

Shoc2-tranduced ERK1/2 motility signals — Novel insights from functional genomics

https://doi.org/10.1016/j.cellsig.2016.02.005Get rights and content

Highlights

  • Depletion of the Shoc2 scaffold attenuates cell motility and adhesion.

  • Shoc2-mediated ERK1/2 signals control expression of multiple proteins, including LGALS3BP.

  • Ectopic expression of LGALS3BP rescues adhesion deficiency of the Shoc2-depleted cells.

Abstract

The extracellular signal-regulated kinase 1 and 2 (ERK1/2) pathway plays a central role in defining various cellular fates. Scaffold proteins modulating ERK1/2 activity control growth factor signals transduced by the pathway. Here, we analyzed signals transduced by Shoc2, a critical positive modulator of ERK1/2 activity. We found that loss of Shoc2 results in impaired cell motility and delays cell attachment. As ERKs control cellular fates by stimulating transcriptional response, we hypothesized that the mechanisms underlying changes in cell adhesion could be revealed by assessing the changes in transcription of Shoc2-depleted cells. Using quantitative RNA-seq analysis, we identified 853 differentially expressed transcripts. Characterization of the differentially expressed genes showed that Shoc2 regulates the pathway at several levels, including expression of genes controlling cell motility, adhesion, crosstalk with the transforming growth factor beta (TGFβ) pathway, and expression of transcription factors. To understand the mechanisms underlying delayed attachment of cells depleted of Shoc2, changes in expression of the protein of extracellular matrix (lectin galactoside-binding soluble 3-binding protein; LGALS3BP) were functionally analyzed. We demonstrated that delayed adhesion of the Shoc2-depleted cells is a result of attenuated expression and secretion of LGALS3BP. Together our results suggest that Shoc2 regulates cell motility by modulating ERK1/2 signals to cell adhesion.

Introduction

The mechanisms leading to activation of RAF, MEK and ERK kinases in the extracellular signal-regulated kinase 1 and 2 (ERK1/2) pathway have been studied extensively [1], [2]. However, what determines the activity of the pathway in the context of a specific set of downstream targets and how ERK signals result in distinct biological outcomes are not yet clear. Several studies suggest that divergent cell fates induced by the ERK pathway are the result of a tight spatio-temporal control of ERK1/2 targeting, sequestration and activation of the kinases and phosphatases, as well as modulation of the strength and duration of the ERK signaling [3], [4]. Scaffold proteins have been proposed to fulfill some of these requirements. Scaffolds have been implicated in controlling the spatial organization of signaling enzymes, insulation of active modules and prevention of a spurious cross-talk of signaling networks [5], [6]. Yet, detailed mechanisms allowing scaffolds to elicit specific cellular responses at the molecular level remain to be elucidated.

Scaffold proteins of the ERK1/2 pathway represent a diverse group of proteins [2]. A well-studied scaffold kinase suppressor of Ras 1 (KSR1) [7], [8], [9] is the multifunctional protein that binds to and accelerates activity of MEK and RAF kinases thereby stimulating expression of genes that drive cell proliferation and differentiation [10], [11]. Other ERK1/2 scaffolds, mitogen-activated protein 1 (MP1) and p14 (also called the LAMTOR2/3 complex) are believed to regulate cytoskeletal dynamics [12], [13], [14], [15], and the MP1/p14 complex is involved in remodeling of focal adhesion and actin structures during cell spreading [16].

Ras–RAF-1–ERK1/2 signaling is accelerated by the scaffolding protein Shoc2 [17], [18]. This evolutionarily well-conserved protein is essential for normal development [19], [20], [21]. Loss of Shoc2 in mammalian cultured cells and Caenorhabditis elegans leads to a dramatic decrease in ERK1/2 activity [17], [22], [23]. As a scaffold protein, Shoc2 provides a molecular platform for multi-protein assemblies that modulate ERK1/2 activity [24], [25]. In addition to its signaling partners Ras and RAF-1, Shoc2 tethers the catalytic subunit of protein phosphatase 1c (PP1c) as well as proteins of the ubiquitin machinery HUWE1 and PSMC5 [23], [26], [27]. The ability of this non-catalytic scaffold to mediate ERK1/2 signaling is controlled through allosteric ubiquitination [24]. Alterations in the mechanisms controlling ubiquitination of the scaffold affect Shoc2-mediated ERK1/2 signals and cell motility [27].

Activation of the ERK1/2 pathway in response to epidermal growth factor (EGF) stimulation of the EGF receptor falls into three major regulatory loops: immediate, delayed, and late (secondary) [28], [29], [30]. The immediate regulatory loop induces phosphorylation of transcription factors such as FOS, Jun and EGR1 and does not require new protein synthesis for their transcription [30]. Expression of the genes of the immediate response induces transcription of delayed genes, such as the RNA-binding protein ZFP36 or dual specific phosphatases, which dephosphorylate ERK1/2 kinases that terminate the activity of the immediate loop [30]. Late (secondary) transcriptional response leads to expression of genes such as actin-binding proteins or genes encoding proteins that are involved in cell metabolism and biogenesis of membranes and appear to define cellular outcomes [31].

In the current study, we aimed to determine the specific ERK1/2 response elicited through the Shoc2 scaffolding module. Results of this study provide evidence that Shoc2-mediated ERK1/2 activity contributes to maintenance of the ERK1/2 feedback loop that regulates expression of genes of the TGFβ pathway. We also found that Shoc2–ERK1/2 signals control cell motility and adhesion, in part, through mechanisms that monitor expression of the protein of extracellular matrix — lectin galactoside-binding soluble 3-binding protein or LGALS3BP (also called Mac-2 binding protein) [32]. Deficient expression and secretion of this heavily glycosylated protein led to attenuated attachment of Shoc2-depleted cells. These results indicate that Shoc2 transduces signals to unique cellular responses and identifies novel molecular targets of the Shoc2–ERK1/2 signaling axis.

Section snippets

Reagents and antibodies

EGF was obtained from BD Bioscience. U0126 and PD98059 were obtained from LC Laboratories. Respective proteins were detected using specific primary antibodies, including: GAPDH, phospho-ERK1/2, ERK1/2, MEK1/2, COL1A1 and EGFR (Santa Cruz Biotechnology); His, Shoc2 and LGALS3BP (Proteintech); phospho-AKT, KSR1, and phospho-MEK1/2 (Cell Signaling).

Constructs

Shoc2-tRFP was described previously [25], [33]. The plasmid carrying full-length His-tagged LGAL3SBP was obtained from Dr. Enza Picollo (Chieti,

Cells depleted of Shoc2 scaffold exhibit reduced cell motility

We and others have suggested that Shoc2 is involved in regulating cell polarity and movement [26], [27], [41]. To understand the cellular functions controlled by the Shoc2-mediated ERK1/2 signals, we utilized Cos1 cells stably depleted of Shoc2 (Cos-LV1) as well as cells stably depleted of Shoc2 and then rescued with Shoc2-tRFP (Cos-SR) [25], [42]. We also generated T47D and MCF7 cells constitutively depleted of Shoc2 (T47D-LV1, MCF7-LV1) and T47D and MCF7 cells depleted of Shoc2 and then

Discussion

Diverse cellular outcomes triggered by the ERK1/2 signaling cascade include modulation of cell cycle, proliferation, transcription, migration, and senescence, to name a few. Scaffold proteins have been suggested to offer conduits that convey a multipotent general ERK signal into a specific cellular outcome [6], [56]. However, our understanding of how signals are directed by the individual ERK1/2 scaffolds is still limited. The findings presented in this study provide sufficient evidence for the

Author contributions

EG and MKJ planned the experiments, analyzed and interpreted data. MKJ and ERJ performed all biochemical assays. ER, JL, CW and WL performed biostatistical analysis. EG and MKJ drafted the manuscript.

Conflicts of interest

None.

Acknowledgments

We thank Drs. Matthew Gentry, Tianyan Gao, Charles Waechter and Stacy Smith for providing reagents and critical reading of the manuscript; the Genetic Technologies Core at the Department of Molecular and Cellular Biochemistry (University of Kentucky) for assistance with the production of lentiviruses; and the UK Flow Cytometry & Cell Sorting core facility for assistance in cell sorting.

The Genetic Technologies and Protein cores mentioned above are supported in part by a grant from the National

References (71)

  • M.F. Favata

    Identification of a novel inhibitor of mitogen-activated protein kinase kinase

    J. Biol. Chem.

    (1998)
  • J.W. Ramos

    The regulation of extracellular signal-regulated kinase (ERK) in mammalian cells

    Int. J. Biochem. Cell Biol.

    (2008)
  • C.J. Marshall

    Specificity of receptor tyrosine kinase signaling: transient versus sustained extracellular signal-regulated kinase activation

    Cell

    (1995)
  • T. Yamamoto

    Continuous ERK activation downregulates antiproliferative genes throughout G1 phase to allow cell-cycle progression

    Curr. Biol.

    (2006)
  • C. Brakebusch

    Expression of the 90K immunostimulator gene is controlled by a promoter with unique features

    J. Biol. Chem.

    (1997)
  • C. Brakebusch

    Isolation and functional characterization of the human 90K promoter

    Genomics

    (1999)
  • M. Nonaka

    Dendritic cell-specific intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN) recognizes a novel ligand, Mac-2-binding protein, characteristically expressed on human colorectal carcinomas

    J. Biol. Chem.

    (2011)
  • T.A. Ulmer

    The tumor-associated antigen 90K/Mac-2-binding protein secreted by human colon carcinoma cells enhances extracellular levels of promatrilysin and is a novel substrate of matrix metalloproteinases-2, -7 (matrilysin) and -9: implications of proteolytic cleavage

    Biochim. Biophys. Acta

    (2010)
  • M. Artini

    Elevated serum levels of 90K/MAC-2 BP predict unresponsiveness to alpha-interferon therapy in chronic HCV hepatitis patients

    J. Hepatol.

    (1996)
  • J.M. Mataraza

    Multiple proteins mediate IQGAP1-stimulated cell migration

    Cell. Signal.

    (2007)
  • W. Kolch

    Meaningful relationships: the regulation of the Ras/Raf/MEK/ERK pathway by protein interactions

    Biochem. J.

    (2000)
  • G. Pearson

    Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions

    Endocr. Rev.

    (2001)
  • M.D. Brown et al.

    Compartmentalised MAPK pathways

    Handb. Exp. Pharmacol.

    (2008)
  • W. Kolch

    Coordinating ERK/MAPK signalling through scaffolds and inhibitors

    Nat. Rev. Mol. Cell Biol.

    (2005)
  • L.K. Langeberg et al.

    Signalling scaffolds and local organization of cellular behaviour

    Nat. Rev. Mol. Cell Biol.

    (2015)
  • H. Zhang

    The dual function of KSR1: a pseudokinase and beyond

    Biochem. Soc. Trans.

    (2013)
  • L.G. Pérez-Rivas et al.

    Modulation of Ras Signaling by the KSR Family of Molecular Scaffolds

    (2010)
  • M.M. McKay et al.

    Signaling dynamics of the KSR1 scaffold complex

    Proc. Natl. Acad. Sci. U. S. A.

    (2009)
  • J. Lin

    KSR1 modulates the sensitivity of mitogen-activated protein kinase pathway activation in T cells without altering fundamental system outputs

    Mol. Cell. Biol.

    (2009)
  • A. Claperon et al.

    KSR and CNK: two scaffolds regulating RAS-mediated RAF activation

    Oncogene

    (2007)
  • N. Schiefermeier

    The late endosomal p14-MP1 (LAMTOR2/3) complex regulates focal adhesion dynamics during cell migration

    J. Cell Biol.

    (2014)
  • B. Thauerer

    LAMTOR2-mediated modulation of NGF/MAPK activation kinetics during differentiation of PC12 cells

    PLoS One

    (2014)
  • A. Pullikuth

    The MEK1 scaffolding protein MP1 regulates cell spreading by integrating PAK1 and Rho signals

    Mol. Cell. Biol.

    (2005)
  • L.M. Selfors

    soc-2 encodes a leucine-rich repeat protein implicated in fibroblast growth factor receptor signaling

    Proc. Natl. Acad. Sci. U. S. A.

    (1998)
  • J. Yi

    Endothelial SUR-8 acts in an ERK-independent pathway during atrioventricular cushion development

    Dev. Dyn.

    (2010)
  • Cited by (12)

    • Shoc2 controls ERK1/2-driven neural crest development by balancing components of the extracellular matrix

      2022, Developmental Biology
      Citation Excerpt :

      Our data suggest that changes in the expression of the central players regulating the specification of NC (e.g. foxd3, sox9a, and sox10) likely trigger changes in transcriptional programs responsible for cytoskeletal rearrangement, a gain of motility by NCC, and the downstream epithelial-to-mesenchymal transition (EMT) (e.g. expression of twist, chd1, chd2). These findings are well-aligned with the results of earlier studies demonstrating that signals of the Shoc2-ERK1/2 axis coordinate cell adhesion and movement of cultured cells via controlling the expression of the various proteins regulating these processes (Jeoung et al., 2016; Kaduwal et al., 2015; Young et al., 2013). Others suggested that M-Ras/Shoc2 signals regulate cell motility by coordinating the turnover of E-cadherin at cell-cell junctions and modulating its interaction with p120-catenin (Kota et al., 2019).

    • M-Ras is Muscle-Ras, Moderate-Ras, Mineral-Ras, Migration-Ras, and Many More-Ras

      2020, Experimental Cell Research
      Citation Excerpt :

      Cultured human cell lines overexpressing either wild-type or a constitutively active M-Ras show elevated collective cell migration ability in wound-healing and cell-scattering assays, whereas M-Ras or Shoc2 knockdown cells exhibit reduced migration ability [52,106]. In addition, M-Ras or Shoc2 knockdown tumor cells display decreased directed migration ability in a transwell migration assay [52,107]. In normal epithelial cells, active M-Ras recruits Shoc2 from the cytosol to cell–cell junctions.

    • Single-domain antibodies for functional targeting of the signaling scaffold Shoc2

      2020, Molecular Immunology
      Citation Excerpt :

      Together, these results strongly suggest that hs2dAbB99 and hs2dAbB120 facilitate Shoc2-mediated ERK1/2 activation. The role of the Shoc2 scaffold protein in regulating specificity of ERK1/2 signals has been well-recognized by a number of studies (Boned Del Rio et al., 2019; Jang et al., 2019b; Jeoung et al., 2016; Li et al., 2000; Rauen, 2013; Rodriguez-Viciana et al., 2006). Shoc2 assembles intricate multi-protein complex machinery that accelerates and fine-tunes the amplitude of ERK1/2 signals (Jang and Galperin, 2016).

    • The FBXW7-SHOC2-Raptor Axis Controls the Cross-Talks between the RAS-ERK and mTORC1 Signaling Pathways

      2019, Cell Reports
      Citation Excerpt :

      SHOC2 is an evolutionarily conserved protein, composed of an unstructured N-terminal domain and a long stretch of leucine-rich repeats (LRRs) (Jeoung et al., 2013). The N-terminal domain binds to RAS and RAF to activate ERK1 and ERK2 (Dai et al., 2006; Jeoung et al., 2013; Jeoung et al., 2016). In addition, SHOC2 is upregulated in the majority of human cancers (Young et al., 2013).

    View all citing articles on Scopus
    View full text