Elsevier

Methods in Enzymology

Volume 406, 2006, Pages 425-437
Methods in Enzymology

Analysis of Activated GAPs and GEFs in Cell Lysates

https://doi.org/10.1016/S0076-6879(06)06031-9Get rights and content

Abstract

An assay was developed that allows the precipitation of the active pools of Rho‐GEFs, Rho‐GAPs, or effectors from cell or tissue lysates. This assay can be used to identify GEFs, GAPs, and effectors involved in specific cellular pathways to determine their GTPase specificity and to monitor the temporal activation of GEFs and GAPs in response to upstream signals.

Introduction

Rho GTPases control many aspects of cellular behavior, including the organization of the cytoskeleton, cell migration, cell adhesion, cell cycle, and gene expression (Burridge 2004, Hall 1998, Van Aelst 1997). Like all GTPases, Rho proteins act as molecular switches by cycling between an active (GTP bound) and an inactive (GDP bound) state. Exchange of GTP for GDP allows the GTPase to interact with downstream effectors to modulate their activity and localization. Cycling between the GDP‐bound and GTP‐bound state is regulated primarily by two distinct families of proteins: guanine‐nucleotide exchange factors (GEFs) activate Rho proteins by catalyzing the exchange of GDP for GTP, the GTPase activating proteins or GAPs negatively regulate GTPase function by stimulating GTP hydrolysis.

Since the development of the Rho pull‐down assay in 1999 (Ren et al., 1999), a lot of progress has been made in determining the identity of the Rho‐GTPases that are activated in response to certain stimuli or signals. However, much less has been done in terms of identifying the molecules involved in the activation and inactivation of each particular Rho‐GTPase. The issue becomes more complicated considering that the human genome contains more than 60 RhoGEFs and approximately 80 RhoGAPs (Moon 2003, Peck 2002, Rossman 2005, Schmidt 2002).

We have developed an affinity precipitation assay that allows us to specifically pull down RhoGEFs, RhoGAPs, or effectors that are being activated in the cell at any given time or condition. The assay takes advantage of constitutively active and dominant negative Rho‐family mutants that bind either to Rho‐GAPs and effectors or to RhoGEFs, respectively. Constitutively active Rho mutants were originally designed based on their analogous Ras mutants (G12V and Q61L) (Garrett 1989, Ridley 1992, Ridley 1992). These mutants have lost both their intrinsic capacity and their GAP‐mediated ability to hydrolyze GTP, and they bind with high affinity to GAPs and effectors (Barbacid 1987, Trahey 1987). Traditional dominant negative mutants like S17N‐Ras and the analogous Rho mutants have been shown to bind GDP with similar affinity to the corresponding wild‐type form. However, their affinity for GTP is extremely low, so virtually all of the protein is found in the GDP‐bound form (Ridley 1992, Feig 1999). Another dominant negative Ras mutant (RasG15A) has been previously shown to bind very poorly to both GDP and GTP, existing virtually in a nucleotide free state (Chen et al., 1994). A nucleotide‐free GTPase is one of the intermediates of the nucleotide exchange reaction and is able to form a high affinity binary complex with the GEF (Cherfils and Chardin, 1999). This intermediate is rapidly dissociated by GTP and does not accumulate in cells. We took advantage of the properties of these mutants and used them to pull down Rho effectors and GAPs from tissue and cell lysates (Arthur 2002, Noren 2003). We generated mutant versions of various representative GTPases of the Rho subfamily that harbor mutations equivalent to the Q61L and G15A mutations in Ras. We then used GST fusion proteins of these mutants to specifically pull down Rho family GEFs, GAPs, or effectors from cell or tissue lysates. This assay can be used to determine Rho protein specificity on GEFs, GAPs, or effectors (Arthur 2002, Ellerbroek 2004, Noren 2003, Wennerberg 2003). It can also be used as a simple way to find interactors such as GEFs, GAPs, and effectors to GTPases where little is known about upstream and downstream regulation. In addition, many GEFs and GAPs seem to be activated by making the binding site to their target GTPase available. This can be achieved either by unmasking an intramolecular inhibitory domain (Vav1, Dbl, Asef), by association with or dissociation from other proteins (p115‐RhoGEF, Sos, Dock180), or by release of the protein from a sequestering cellular compartment (GEF‐H1/Lfc, Net1, Ect2) (Schmidt 2002, Rossman 2005). Given this type of regulation, it is likely that the activated and nucleotide‐free mutants of Rho proteins will have highest affinity toward “activated” GAPs and GEFs, respectively, and that our assay can, indeed, be used to detect activation of GEFs and GAPs by specific stimuli.

Activation and inactivation of GEFs and GAPs can be studied either by analyzing the precipitation of known candidate components by immunoblot or by determining the identity of the bound proteins by mass spectrometry. In addition, GEF and GAP pull‐downs can be an extremely useful tool to follow the pattern of GAP or GEF activation over time or in response to different upstream signals.

Section snippets

DNA Constructs

Human cDNA for RhoA, Rac1, and Cdc42 were subcloned into pGEX 4T‐1 (Amersham) between the EcoRI and XhoI sites. Empty‐nucleotide mutants, G17A (RhoA), G15A (Rac1 and Cdc42), and constitutively active mutants, Q63L (RhoA), and Q61L (Rac1 and Cdc42) were generated by site‐directed mutagenesis using the Quick Change Site–directed mutagenesis kit (Stratagene) following the manufacturer's instructions.

Antibodies

Antibodies against p190RhoGAP, ROCK, Sos1, and GFP were from BD Biosciences; Lsc, LARG, and RhoGDI

Expression and Purification of GST‐Rho Constructs

  • 1

    Transform and grow up Escherichia coli with the appropriate pGEX construct. For RhoA, Rac1, and Cdc42 proteins, DH5α or any regular strain will work. For some less‐soluble Rho proteins (RhoB, RhoC, RhoG), it is good to use a codon optimized strain such as codon plus BL21 (Stratagene).

  • 2

    Grow up a 50‐ml culture overnight to full density in LB with 50 μg/ml ampicillin (LB‐Amp)(O.D. > 1.0).

  • 3

    Dilute the culture into 450 ml of LB‐Amp and let grow for 30 min at 37°.

  • 4

    Induce the protein by adding IPTG to a

GAP and GEF Pull‐Downs

Wild‐type RhoA, nucleotide‐free RhoA (G17A‐RhoA), and constitutively active Q63L‐RhoA were expressed as GST fusion proteins, and affinity precipitations were performed on lysates of CHO cells to assess the ability of these GST fusion proteins to bind to known regulators of Rho proteins. We immunoblotted the precipitated proteins with antibodies against a RhoA‐GAP (p190RhoGAP), a RhoA effector ROCK (ROK or Rho kinase), a RhoA‐GEF (Lsc/p115 RhoGEF), and RhoGDI. Our results revealed that

Stability of GST‐Rho Proteins

Some of the GST‐Rho mutants, in particular the empty nucleotide constructs, are not as stable as their wild‐type counterparts when expressed in bacteria. Protein stability decreases even more when proteins are eluted from beads. One way to deal with this problem is to prepare the GST proteins fresh when needed and to keep them at −20° with glycerol (see “Methods”) for no longer than 1 week.

Bacterial Contaminants

The method described here to purify the GST fusion proteins allows fast purification. However, depending

Acknowledgments

We thank Dr. Janiel Shields and Dr. Channing Der for the Ras‐transformed cell lines. We would also like to thank Adi Dubash for the critical reading of this manuscript. This work was supported by NIH grants GM29860 and HL45100. RGM was supported by a postdoctoral fellowship from the Susan G. Komen Breast Cancer Foundation.

References (27)

  • WennerbergK. et al.

    Rnd proteins function as RhoA antagonists by activating p190 RhoGAP

    Curr. Biol.

    (2003)
  • BarbacidM.

    Ras genes

    Annu. Rev. Biochem.

    (1987)
  • ChenS.Y. et al.

    Ras‐15A protein shares highly similar dominant‐negative biological properties with Ras‐17N and forms a stable, guanine‐nucleotide resistant complex with CDC25 exchange factor

    Oncogene

    (1994)
  • Cited by (163)

    • Gα<inf>s</inf>directly drives PDZ-RhoGEF signaling to Cdc42

      2020, Journal of Biological Chemistry
      Citation Excerpt :

      Activation of RhoA and Cdc42, using cells grown in 10-cm dishes, was assessed by pulldown using recombinant Rhotekin and PAK effector domains (13, 46). Active RhoGEFs were detected by pulldown with nucleotide-free GTPases (RhoA G17A and Cdc42 G15A) fused to GST (20, 45). Before lysis, the cells were washed with PBS containing 10 mm MgCl2 and lysed with 1 ml of ice-cold lysis buffer (50 mm Tris, 150 mm NaCl, pH 7.5, containing 1% Triton X-100, 5 mm EDTA, and protease and phosphatase inhibitors (13)).

    View all citing articles on Scopus
    View full text