Elsevier

Cellular Signalling

Volume 18, Issue 9, September 2006, Pages 1473-1481
Cellular Signalling

The very C-terminus of PRK1/PKN is essential for its activation by RhoA and downstream signaling

https://doi.org/10.1016/j.cellsig.2005.11.009Get rights and content

Abstract

PRK1 is a lipid- and Rho GTPase-activated serine/threonine protein kinase implicated in the regulation of receptor trafficking, cytoskeletal dynamics and tumorigenesis. Although Rho binding has been mapped to the HR1 region in the regulatory domain of PRK1, the mechanism involved in the control of PRK1 activation following Rho binding is poorly understood. We now provide the first evidence that the very C-terminus beyond the hydrophobic motif in PRK1 is essential for the activation of this kinase by RhoA. Deletion of the HR1 region did not completely abolish the binding of PRK1-ΔHR1 to GTPγS-RhoA nor the activation of this mutant by GTPγS-RhoA in vitro. In contrast, removing of the last six amino acid residues from the C-terminus of PRK1 or truncating of a single C-terminal residue from PRK1-ΔHR1 completely abrogated the activation of these mutants by RhoA both in vitro and in vivo. The critical dependence of the very C-terminus of PRK1 on the signaling downstream of RhoA was further demonstrated by the failure of the PRK1 mutant lacking its six C-terminal residues to augment lisophosphatidic acid-elicited neurite retraction in neuronal cells. Thus, we show that the HR1 region is necessary but not sufficient in eliciting a full activation of PRK1 upon binding of RhoA. Instead, such activation is controlled by the very C-terminus of PRK1. Our results also suggest that the very C-terminus of PRK1, which is the least conserved among members of the protein kinase C superfamily, is a potential drug target for pharmacological intervention of RhoA-mediated signaling pathways.

Introduction

Protein kinase C-related kinase 1 (PRK1, also known as PKN or PKNα) is a lipid-activated serine/threonine protein kinase and a member of the protein kinase C (PKC) superfamily [1], [2]. PRK1 was originally identified in the liver as a protease-activated serine/threonine kinase via conventional protein chemistry [3], [4], [5]. The cDNAs encoding PRK1 were subsequently cloned by independent groups using homology-based PCR cloning and low-stringency screening of cDNA library [1], [2], [6].

PRK1 consists of a single polypeptide chain of approximately 116 kDa with a regulatory domain at the N-terminal half of the protein and a catalytic domain in the C-terminal half of the kinase. The catalytic domain of PRK1 has 50% homology to that of the PKC, while its regulatory domain is less conserved as it lacks a C1 domain and has a C2-related domain that does not bind Ca2+. In the N-terminal, there is a region (residues 37–281) that consists of three repeats of approximately 80 amino acid residues with each containing a leucine zipper-like motif [7]. Due to its high homology with the corresponding region in its sister enzyme PRK2 [8], this region is known as homology region-1 (HR1), encompassing HR1a, HR1b and HR1c (Fig. 1A). Interestingly, the HR1 domain is not found in PKCs [1]. Therefore, PRK1 is not activated by classical PKC activators such as diacylglycerol, phorbol ester or Ca2+. Instead, it is activated by phospholipids and unsaturated fatty acids, notably by cardiolipin [1], [2], [5].

PRK1 is found in all eukaryotic organisms and its amino acid sequence is highly conserved during evolution [9]. For example, the Xenopus PRK1 possesses 75% amino acid sequence homology to that of human and rat PRK1 [10]. Although PRK1 is a late addition to the PKC superfamily, it might represent the precursor of the mammalian PKC during evolution. In the yeast, the PKC1p from Saccharomyces cerevisiae and the Pck1P and Pck2P from Saccharomyces pombe resemble PRKs structurally as they possess two copies of the HR1 domain at the N-terminus but lack the functional C1 or C2 domains. Furthermore, yeast PKCs are biochemically distinct from mammalian PKCs in that they are activated by Rho GTPase but not by diacylglycerol or calcium [11]. In fact, human PRK1 was one of the first RhoA effectors discovered in 1996 by the seminal work at two laboratories showing that PRK1 binds to and is activated by GTP-bound RhoA in mammalian cells [12], [13]. The importance of PRK1 in development is underscored by the finding that a loss-of-function mutation in the Drosophila Prk1 gene results in a dorsal closure defect during embryogenesis that is lethal to the developing fly embryo [14]. In mammalian cells, PRK1 has been shown to act downstream of Rho GTPase to regulate the intracellular trafficking of the epidermal growth factor receptor in HeLa carcinoma cells [15]; to modulate stress-induced gene expression in NIH3T3 and HEK293T cells [16] and to mediate the formation of cortical actin in mouse embryonic fibroblasts [17].

Elegant biochemical analyses revealed that a segment in the regulatory domain spanning amino acid residues 33–111 constitutes the minimal region in PRK1 that is required for the interaction with RhoA [18]. Further studies showed that HR1a and HR1b subdomains can each bind with RhoA in a GTP-dependent manner [25]. Detailed molecular interactions between PRK1 and RhoA were mapped by solving the crystal structure of PRK1 residues 13–98 (HR1a) complexed to guanosine 5′-3-O-thio-triphosphate (GTPγS)-RhoA at 2.2 Å resolution [19]. It was found that the HR1a domain forms an antiparallel coiled-coil (ACC) and binds to two distinct surface sites of RhoA, one within the switch II and the other with the switch I region of RhoA via hydrophobic and electrostatic interactions. The ACC-like structure was also found in HR1b of PRK1 [20]. The equilibrium dissociation constant between RhoA and the PRK1 HR1a or HR1b domain is estimated to be in the range of 150–300 nM and 180 μM, respectively [20], [21]. However, it is not clear whether the HR1 region of PRK1 is necessary and sufficient for the interaction with RhoA and for transducing signals from RhoA to downstream cellular machinery. Given the recently identified roles of PRK1 in transducing signals of RhoA to regulate cytoskeletal dynamics in neuronal cells and in initiating transcriptional superactivation of the androgen receptor during the progression of prostate cancer [22], [23], we carried out studies to elucidate molecular mechanisms underlying the control of the activation of PRK1 by RhoA. In particular, we wished to determine whether the HR1 region is the sole determinant for the activation of PRK1 by RhoA both in vitro and in cells. By employing a strategy that combines mutagenesis with biochemical analysis, we find unexpectedly that the very C-terminus of PRK1 is critical for the full activation of PRK1 by RhoA and subsequent downstream signaling in mammalian cells.

Section snippets

Materials

The monoclonal antibodies to Myc-tag (9E10) and the Complete protease inhibitor cocktail were purchased from Roche Applied Science. Horseradish peroxidase-conjugated secondary antibodies were obtained from Pierce. Monoclonal antibodies against FLAG-tag (M2) and β-actin (AC-15), a polyclonal antibody anti-GST as well as all other reagents were from Sigma-Aldrich.

cDNA constructs

An N-terminally Myc-epitope-tagged rat PRK1 cDNA cloned in a pcDNA3 vector was a gift from R.E.H. Wettenhall (University of Melbourne).

Generation and characterization of deletion mutants of PRK1

Previous work has revealed that GST fusion HR1a or HR1b can bind RhoA but GST-HR1c does not interact with RhoA [18], [25]. It has also been shown that HR1abc binds RhoA more tightly than HR1a or HR1b alone [20], [25]. In order to determine whether the HR1 region in the regulatory domain is the sole determinant for the interaction between PRK1 and RhoA, we generated an HR1 domain-deletion mutant of PRK1 by truncating residues 30 to 280 from an N-terminally Myc-tagged rat PRK1 (Fig. 1A). This HR1

Discussion

In this study, we have demonstrated that a segment distant from the RhoA binding sites in a Rho effector serine/threonine protein kinase critically controls the activation of the kinase upon binding of RhoA and the subsequent regulation of cytoskeleton dynamics in cells. We also show that the binding of RhoA to one of its effector kinases and the subsequent activation of the kinase are controlled by more than one domain in the effector kinase.

Given the importance of the Rho family of small

Acknowledgments

We thank A. Newton (University of California at San Diego) and G.M. Bokoch (Scripps Research Institute) for providing us with the P500 antibody and GST-RhoA plasmid, respectively. We also thank Dr. Y. Yuan for editing.

This work was supported by grants from the National Medical Research Council and ASTAR (BMRC), Singapore (to W.D.).

References (42)

  • B. Gabrielli et al.

    FEBS Lett.

    (1984)
  • N.A. Morrice et al.

    J. Biol. Chem.

    (1994)
  • L.V. Dekker et al.

    Curr. Opin. Struct. Biol.

    (1995)
  • H. Mukai et al.

    Biochem. Biophys. Res. Commun.

    (1994)
  • N. Ueno et al.

    Biochem. Biophys. Res. Commun.

    (1997)
  • H. Mukai et al.

    Biochim. Biophys. Acta

    (1995)
  • S. Jayadev et al.

    J. Biol. Chem.

    (1994)
  • A. Gampel et al.

    Curr. Biol.

    (1999)
  • H. Shibata et al.

    FEBS Lett.

    (1996)
  • R. Maesaki et al.

    Mol. Cell

    (1999)
  • D. Owen et al.

    J. Biol. Chem.

    (2003)
  • L. Blumenstein et al.

    J. Biol. Chem.

    (2004)
  • W. Duan et al.

    Gene

    (2000)
  • P. Flynn et al.

    J. Biol. Chem.

    (1998)
  • W.G. Lim et al.

    Cell. Signal.

    (2005)
  • B. Anliker et al.

    J. Biol. Chem.

    (2004)
  • X. Li et al.

    J. Biol. Chem.

    (2002)
  • P. Flynn et al.

    J. Biol. Chem.

    (2000)
  • J. Yang et al.

    Mol. Cell

    (2002)
  • S.S. Taylor et al.

    Pharmacol. Ther.

    (1999)
  • J.M. Sowadski et al.

    Pharmacol. Ther.

    (1999)
  • Cited by (11)

    • The interaction of PKN3 with RhoC promotes malignant growth

      2012, Molecular Oncology
      Citation Excerpt :

      Furthermore, PKN3 catalytic activity is significantly less responsive to fatty acid-mediated stimulation compared to PKN1 and PKN2, thus underscoring the diversity of PKN isoform function and regulation. The PKN family of PKC-related protein kinases constitutes some of the major Rho GTPase-associated protein kinase activities detected in mammalian tissues (Flynn et al., 1998; Lim et al., 2006, 2008; Vincent and Settleman, 1997). Although the interaction of PKN1/PKN2 with RhoA and RhoB has been reported, the biological significance of these interactions remains largely unknown.

    • Regulation of the interaction between protein kinase C-related protein kinase 2 (PRK2) and its upstream kinase, 3-phosphoinositide-dependent protein kinase 1 (PDK1)

      2009, Journal of Biological Chemistry
      Citation Excerpt :

      Besides the regulation by phosphorylation, other mechanisms exist to modulate the activity of PRKs, namely interaction with the small GTPase Rho and lipid binding (31). Recently, Lim et al. (32–34) described that the PRK C terminus is required for the activation by Rho and that it is additionally involved in the regulation of PRKs by lipids. However, it is not yet clear how this is related to the regulatory mechanisms we have observed in this work.

    • The C-terminus of PRK2/PKNγ is required for optimal activation by RhoA in a GTP-dependent manner

      2008, Archives of Biochemistry and Biophysics
      Citation Excerpt :

      Given the recent report that PRK2 regulates hepatitis C virus RNA polymerase function and that overexpression of PRK2 stimulates hepatitis C virus replication in cell cultures [47], the extreme C-terminus of PRK2 may serve as a promising target for specific pharmaceutical interventions in a number of human diseases.

    View all citing articles on Scopus
    View full text