Skip to main content
The Journal of Biological Chemistry logoLink to The Journal of Biological Chemistry
. 2020 Aug 14;295(40):13698–13710. doi: 10.1074/jbc.RA120.013919

Evaluation of active Rac1 levels in cancer cells: A case of misleading conclusions from immunofluorescence analysis

Martin J Baker 1,*, Mariana Cooke 1,2, Gabriel Kreider-Letterman 3, Rafael Garcia-Mata 3, Paul A Janmey 4, Marcelo G Kazanietz 1,*
PMCID: PMC7535912  PMID: 32817335

Abstract

A large number of aggressive cancer cell lines display elevated levels of activated Rac1, a small GTPase widely implicated in cytoskeleton reorganization, cell motility, and metastatic dissemination. A commonly accepted methodological approach for detecting Rac1 activation in cancer cells involves the use of a conformation-sensitive antibody that detects the active (GTP-bound) Rac1 without interacting with the GDP-bound inactive form. This antibody has been extensively used in fixed cell immunofluorescence and immunohistochemistry. Taking advantage of prostate and pancreatic cancer cell models known to have high basal Rac1-GTP levels, here we have established that this antibody does not recognize Rac1 but rather detects the intermediate filament protein vimentin. Indeed, Rac1-null PC3 prostate cancer cells or cancer models with low levels of Rac1 activation still show a high signal with the anti-Rac1-GTP antibody, which is lost upon silencing of vimentin expression. Moreover, this antibody was unable to detect activated Rac1 in membrane ruffles induced by epidermal growth factor stimulation. These results have profound implications for the study of this key GTPase in cancer, particularly because a large number of cancer cell lines with characteristic mesenchymal features show simultaneous up-regulation of vimentin and high basal Rac1-GTP levels when measured biochemically. This misleading correlation can lead to assumptions about the validity of this antibody and inaccurate conclusions that may affect the development of appropriate therapeutic approaches for targeting the Rac1 pathway.

Keywords: Rac1, small GTPase, Rac1-GTP, immunofluorescence, cancer cells, Rac (Rac GTPase), epithelial-to-mesenchymal transition (EMT), imaging, cancer


The GTPase Rac1, a member of the Rho family of small G-proteins, plays key roles in the regulation of cellular functions, including cell migration, proliferation, differentiation, and gene expression. This GTPase is ubiquitously expressed and has been established as a master regulator of actin cytoskeletal reorganization, particularly in the formation of protrusive membrane structures implicated in cell migration and invasion (1, 2). The importance of this protein has also been observed in human disease, including its misregulation or mutation in a number of cancer types (24).

Rac1 is a molecular switch and cycles between active GTP-bound and inactive GDP-bound states. The GTP-bound Rac1 adopts a conformation that is capable of binding effector proteins that mediate downstream signaling responses, most prominently the Pak kinases (5). Indeed, the biochemical assessment of Rac1 activation, determined from cell lysates, relies on the binding of the GTP-bound form to a Pak1-binding domain (PBD), allowing the “pulldown” of the active form of the GTPase and therefore separation from the inactive GDP-bound form. A drawback of this assay, however, is that it only provides information about global levels of active Rac1 in a cell population. Visualization of the Rac1-GTP state in single cells and the assessment of subcellular localization is difficult due to the transient nature of the Rac1 activation and the noncovalent interaction with GTP. The use of Rac-FRET reporters has, to a certain extent, overcome these obstacles by allowing for the spatiotemporal analysis of Rac1 activation in living cells (6, 7) or in transgenic organisms (8, 9). However, this approach requires ectopic expression of the reporter, therefore limiting its physiological relevance and making it an unsuitable tool for studies with fixed tissues, such as patient specimens. Furthermore, the low resolution of this technique can make studying the subcellular distribution of Rac1-GTP difficult.

One of the promising solutions to visualize the activation of endogenous Rac1 in cells or patient samples has been the development of a conformation-sensitive antibody capable of specifically detecting Rac1-GTP. This antibody purportedly provides a unique resource for visualization and quantification of the endogenous active Rac1 in fixed samples, circumventing the need presented by FRET approaches to overexpress probes, while also achieving greater resolution. Indeed, the anti-Rac1-GTP antibody has become a widely used tool for immunofluorescence (1041), proximity ligation (41), immunohistochemistry (19, 35, 38, 4252), flow cytometry (53), and immunoprecipitation assays (10, 11, 22, 33, 54, 55).

The extensive interest of our laboratory in the role of Rac1 signaling in tumorigenesis and metastasis (3, 56, 57) prompted us to use the anti-Rac1-GTP antibody in a number of cancer cell models. In the course of these studies, we stumbled upon a number of inconsistencies that persuaded us to pursue a deeper characterization of this antibody. We took advantage of our recent characterization of models displaying diverse levels of Rac1 activation (56), which we complemented with the use of Rac1 knockout cells. This analysis led us to question the validity of this widely used antibody, unambiguously demonstrating that it is not specific for Rac1-GTP but rather reacts with vimentin, a marker of the mesenchymal cancer cell phenotype. Furthermore, here we show two examples in which misleading information could be derived from its use.

Results

The Rac1-GTP antibody staining colocalizes with vimentin

In a recent study (56), we characterized prostate cancer cell lines with low and high Rac1-GTP levels. Fig. 1A shows that aggressive PC3 cells display markedly higher Rac1-GTP levels relative to less aggressive LNCaP cells. To determine the intracellular localization of Rac1-GTP, we carried out immunofluorescence studies with a widely used conformation-sensitive anti-Rac1-GTP antibody. Our initial experiment revealed that, as expected, prominent staining from the anti-Rac1-GTP antibody could be observed in PC3 cells but not in LNCaP cells (Fig. 1B). Analysis of confocal images from PC3 cells revealed that the antibody recognized a prominent filamentous network localized primarily in the perinuclear region. This staining did not colocalize with filamentous actin visualized by phalloidin (Fig. 1C). These results were unexpected, because active Rac1 would normally localize at the periphery of the cell, as is consistent with its main role in the regulation of actin cytoskeleton dynamics (1, 7, 8). Further co-staining immunofluorescence analysis revealed a lack of colocalization with microtubules (as determined by α-tubulin staining), endoplasmic reticulum (as determined by PDI staining), Golgi (golgin-97 staining), or mitochondria (Mitotracker Deep Red staining). Surprisingly, we did observe a remarkable colocalization with the intermediate filament vimentin, with prominent staining on vimentin filaments close to the nucleus (Fig. 1C). It should be noted that the signal from the vimentin or Rac1-GTP staining could only be observed in the appropriate channel, confirming the absence of any bleed-through of fluorescence. Moreover, no signal could be observed from the secondary antibodies when used alone, confirming the authenticity of the observed signal being from the primary antibody staining (Fig. S1). The colocalization of Rac1-GTP staining with vimentin filaments can be better appreciated with Airyscan super-resolution microscopy (Fig. 1D).

Figure 1.

Figure 1.

Characterization of the anti-Rac1-GTP antibody in PC3 prostate cancer cells. A, LNCaP and PC3 cells were serum-starved, and active Rac1 was isolated using a PBD pulldown assay. Left, representative experiment. Right, densitometric analysis of Rac1-GTP levels normalized to total Rac1. Results, relative to LNCaP cells, are expressed as mean ± S.E. (error bars) (n = 6). B, representative images of PC3 and LNCaP cells fixed and stained for DNA with Hoechst (blue) and anti-Rac1-GTP antibody (red). C, representative images of PC3 cells fixed and stained for DNA with Hoechst (blue) and anti-Rac1-GTP antibody (red) as well as co-stained with phalloidin, Mitotracker Deep Red, anti-α-tubulin, anti-PDI, anti-golgin-97, or anti-vimentin antibodies (green) and subjected to confocal microscopy. D, PC3 cells were fixed and stained for vimentin (green), for DNA (blue), and with the anti-Rac1-GTP antibody (red). Cells were subjected to Airyscan super-resolution confocal microscopy. Scale bar, 10 μm. Similar results were observed in three independent experiments. **, p ≤ 0.01.

Rac1-GTP antibody staining is not specific

To determine the specificity of the staining with the anti-Rac1-GTP antibody, we generated a Rac1 knockout (KO) PC3 cell line using a CRISPR/Cas9 approach. The knockout of Rac1 in these cells was confirmed with Western blotting and Sanger sequencing (Fig. 2, A and B). To our surprise, Rac1 KO PC3 cells display a prominent staining with the anti-Rac1-GTP antibody that has similar intensity as that observed in control PC3 cells and also colocalizes with vimentin (Fig. 2C). These results unambiguously establish that the anti-Rac1-GTP antibody is nonspecific for Rac1.

Figure 2.

Figure 2.

The Rac1-GTP antibody does not detect Rac1 in prostate cancer cells. A, CRISPR was used to generate a Rac1 KO PC3 cell line. Representative Western blotting of Rac1 in Rac1 KO and nontarget CRISPR cell lines is shown. B, sequencing trace of Rac1 KO PC3 cells compared with the CRISPR control showing the 16-base pair deletion. The guide sequence–binding site is indicated by a horizontal black line, and the PAM site is shown by a dashed horizontal red line. The cut site is indicated by a dashed vertical black line. C, representative images of serum-starved control and Rac1 KO cell lines stained with the anti-Rac1-GTP antibody. Cells were fixed and stained for vimentin (green), for DNA (blue), and with the anti-Rac1-GTP antibody (red). Image capture and processing procedures were identical for both images. Scale bar, 10 μm. D, representative confocal microscopy image of the ectopic expression of EGFP-Rac1Q61L in PC3 cells. Cells were transfected with a plasmid expressing EGFP-Rac1Q61L (green) and after 24 h were fixed and stained for DNA (blue) and with the anti-Rac1-GTP antibody (red).

Next, we ectopically expressed in PC3 cells the EGFP-tagged Rac1Q61L mutant, which has impaired GTPase activity and therefore remains in a GTP-bound state. As expected, EGFP-Rac1Q61L displays significant expression in peripheral structures. However, we were unable to find colocalization of this active Rac1 mutant with the staining from the anti-Rac1-GTP antibody (Fig. 2D). Although we cannot rule out that the EGFP tag or Q61L mutation may interfere with the antibody binding, this result is in line with the nonspecific staining in Rac1 KO PC3 cells and supports the idea that the anti-Rac1-GTP antibody is recognizing an alternative epitope to Rac1.

In an effort to analyze in greater detail the colocalization of the anti-Rac1-GTP antibody staining with vimentin, we generated vimentin-deficient PC3 cells. PC3 cells were infected with two different vimentin shRNA lentiviruses, which caused ∼80% depletion of vimentin as determined by Western blotting. The loss of vimentin had no effect on the levels of Rac1-GTP determined biochemically with a PBD pulldown assay (Fig. 3A). Interestingly, and despite the lack of changes in cellular active Rac1 levels, silencing vimentin expression essentially abolished the anti-Rac1-GTP signal detected by immunofluorescence (Fig. 3B). Therefore, the observed filamentous staining produced by this antibody depends on vimentin expression and is not representative of the levels of active, GTP-bound Rac1.

Figure 3.

Figure 3.

Loss of anti-Rac1-GTP antibody staining upon vimentin RNAi depletion. A stable depletion of vimentin from PC3 cells was generated with two different shRNA lentiviruses (#1 and #2). Active Rac1 levels were determined using a PBD pulldown assay. Left, representative experiment. Middle, densitometric analysis of Rac1-GTP levels normalized to total Rac1. Right, densitometric analysis of vimentin levels, normalized to vinculin. Results, relative to nontarget control (NTC), are expressed as mean ± S.E. (error bars) (n = 3). B, representative confocal microscopy images of anti-Rac1-GTP staining in vimentin-depleted PC3 cells. Cells were fixed and stained for vimentin (green), for DNA (blue), and with the anti-Rac1-GTP antibody (red). Capture and processing procedures were identical for all images. Scale bar, 10 μm. Similar results were observed in three independent experiments. **, p ≤ 0.01; ns, not significant.

To further investigate the specificity of this antibody, we stimulated cells to induce Rac1 activation. LNCaP C4-2, a cell line that does not express vimentin, was stimulated with epidermal growth factor (EGF), which caused a dramatic increase in active Rac1 (>20-fold), as determined biochemically using the pulldown assay (Fig. 4A). Despite the prominent elevation in Rac1-GTP levels, no noticeable corresponding increase in staining with the anti-Rac1-GTP antibody could be observed in response to EGF (Fig. 4B).

Figure 4.

Figure 4.

Lack of staining with the anti-Rac1-GTP antibody in EGF-stimulated prostate cancer cells. A, LNCaP C4-2 cells were stimulated with EGF (100 ng/ml, 1 min), and Rac1-GTP levels were determined using a PBD pulldown assay. Top, representative Western blotting. Bottom, densitometric analysis of Rac1-GTP levels normalized to total Rac1 showing mean ± S.E. (error bars). B, representative confocal microscopy images of control and EGF-stimulated C4-2 cells (100 ng/ml, 1 min) stained for DNA (blue), for phalloidin (green), and with the anti-Rac1-GTP antibody (red). Scale bar, 10 μm. Similar results were observed in three independent experiments. *, p ≤ 0.05.

To further substantiate our conclusions, we used A549 cells, a non-small-cell lung cancer model in which EGF causes significant actin cytoskeleton rearrangement into peripheral ruffles and triggers a Rac1-dependent motility response (58). EGF stimulates the formation of large actin structures in A549 cells in a Rac1-dependent manner, as judged by the impaired response in Rac1 KO A549 cells (Fig. 5A). We then sought to determine whether Rac1 is activated at sites of EGF-induced membrane ruffling by using a Rac1-FRET reporter (59). As shown in Fig. 5B and Video S1, stimulation of A549 cells with EGF caused significant activation of Rac1 detected by the FRET reporter at the site of peripheral protrusions. However, probing stimulated A549 cells with the anti-Rac1-GTP antibody resulted in no visible staining in or around peripheral actin structures and instead stained vimentin filaments (Fig. 5C). Taken together, these results support the evidence that this antibody does not recognize active Rac1.

Figure 5.

Figure 5.

The anti-Rac1-GTP antibody does not stain activated Rac1 in peripheral actin-rich protrusions. A, control A549 cells (scrambled sgRNA) or Rac1 KO A549 cells were treated with EGF (200 ng/ml, 5 min) and subjected to Alexa Fluor 488–conjugated phalloidin staining. Left, Rac1 levels determined by Western blotting. Right, representative images of phalloidin-stained cells. B, A549 cells co-expressing a Rac-FRET biosensor and mCherry-cortactin were stimulated with EGF (added at 1:15 min) and imaged every 15 s. The top panels show the mCherry-Cortactin, and the bottom panels show the Rac1 activation reported by the FRET biosensor. Color scale bar, dynamic range of the biosensor response (1, no significant response; 1.96, strongest response throughout the time-lapse sequence). Scale bar, 5 μm. C, representative confocal microscopy image of an actin-rich protrusion in parental EGF-stimulated A549 cells stained for DNA (blue), for phalloidin (green), for vimentin (gray), and with the anti-Rac1-GTP antibody (red).

Examples of misleading results with the anti-Rac1-GTP antibody in cancer models

Despite this lack of specificity, the anti-Rac1-GTP antibody has been widely used to support results obtained with other tools and techniques, particularly in cancer cell models. Although results obtained with this antibody often fit with the expected research paradigm, it should be noted that a large number of cancer cells display elevated levels of vimentin, a marker of the mesenchymal cancer cell phenotype (60). In the mesenchymal state, cancer cells are more migratory and may therefore have enhanced active Rac1 signaling. Moreover, it has been recently shown that cancer cells undergoing epithelial-to-mesenchymal transition increase their global Rac1-GTP levels as determined biochemically (61). Therefore, it is conceivable that the observed staining often reported in cancer cells with the anti-Rac1-GTP antibody reflects an elevated vimentin expression status and is being erroneously reported as Rac1-GTP.

To test this hypothesis, we first used prostate cancer cell lines with different levels of basal Rac1-GTP. We have previously shown that androgen-independent aggressive cell lines DU145, PC3, and PC3-ML have high levels of basal active Rac1 when compared with a normal prostate epithelial cell line (RWPE1), prostate hyperplasic cell line (BPH-1), and less aggressive prostate cancer cell lines (LNCaP and LNCaP variants), which all have low levels of Rac1-GTP (56) (Fig. 6A). Interestingly, only the aggressive prostate cancer cells display high levels of vimentin expression, with the notable exception of RWPE1 cells (Fig. 6B). Based on this observation, we carried out immunofluorescence analysis of three cell lines: RWPE1 (Rac1-GTP low, vimentin high), LNCaP (Rac1-GTP low, vimentin low), and PC3 (Rac1-GTP high, vimentin high). As shown in Fig. 6C, prominent staining with the anti-Rac1-GTP antibody could be observed only in those cell lines with high vimentin (RWPE1 and PC3). Again, these results indicate that this antibody does not recognize active Rac1; rather, the staining fits with the pattern of vimentin expression.

Figure 6.

Figure 6.

Vimentin expression and anti-Rac1-GTP staining in prostate cancer cell lines. A, representative Western blotting of a PBD assay showing Rac1-GTP and total Rac1 levels in prostate cancer cell lines. B, representative Western blotting of vimentin levels in prostate cancer cell lines. C, representative confocal images of anti-Rac1-GTP (red), vimentin (green), and DNA (blue) staining in the indicated prostate cancer cell lines. Capture and processing procedures were identical for all images. Scale bar, 10 μm. Similar results were observed in three independent experiments.

Last, we decided to evaluate this paradigm in cellular models of pancreatic cancer. Using the PBD pulldown assay, we identified pancreatic cancer cell lines with low Rac1-GTP (Capan-1 and BxPC-3) and cell lines with high Rac1-GTP levels (AsPC-1 and PANC-1). The expression of vimentin in these cell lines also correlates with the levels of Rac1-GTP (Fig. 7A), similar to the observation in prostate cancer models. When we examined the staining of these cells with the anti-Rac-1-GTP antibody, we found that it can only be detected in AsPC-1 and PANC-1, the cell lines that display high vimentin expression and visible vimentin staining (Fig. 7B). To establish a causal relationship between staining with the anti-Rac1-GTP antibody and vimentin expression, we subsequently depleted vimentin in PANC-1 cells using two different shRNA lentiviruses (Fig. 7C). Notably, there was a marked loss in anti-Rac1-GTP staining in the vimentin-knocked down cells concomitant with a loss of vimentin staining (Fig. 7D), thus recapitulating the results obtained in PC3 cells.

Figure 7.

Figure 7.

An example of misleading conclusions with the anti-Rac1-GTP antibody in pancreatic cancer cells. A, pancreatic cell lines Capan-1, BxPC-3, AsPC-1, and PANC-1 were serum-starved for 24 h, and Rac1-GTP levels were determined using a PBD pulldown assay. Left, representative experiment. Right, densitometric analysis of Rac1-GTP levels normalized to total Rac1. Results, relative to Capan-1, are expressed as mean ± S.E. (error bars) (n = 4). B, representative confocal microscopy images of anti-Rac1-GTP (red), vimentin (green), and DNA (blue) staining in the indicated pancreatic cell lines. Capture and processing procedures were identical for all images. Scale bar, 10 μm. C, representative Western blotting showing the expression of vimentin and Rac1 in PANC-1 cells subjected to vimentin shRNA lentiviral depletion or infected with NTC lentivirus. D, nontarget control (NTC) and stable vimentin-depleted PANC-1 cell lines were stained for anti-Rac1-GTP (red), vimentin (green), and DNA (blue). Image capture and processing procedures were identical for all images. Scale bar, 10 μm. B and D, confocal microscopy was used to capture images. Scale bar, 10 μm.

Discussion

Our detailed characterization of the anti-Rac1-GTP antibody clearly demonstrates that it is not specific for its stated target. Results presented in this study revealed that the staining from this antibody colocalizes with vimentin and depends on vimentin expression. The staining is not lost in PC3 cells subjected to CRISPR/Cas9-mediated KO of Rac1, therefore conclusively indicating that it does not recognize Rac1.

Previous users of this antibody have demonstrated its specificity through the use of dominant-negative Rac1 overexpression (12, 15, 22) or immunoprecipitation of endogenous Rac1 in response to stimuli (10, 11, 22, 33, 54). It may be possible that the overexpression of dominant-negative Rac1 inadvertently affected the expression of the Rac1-GTP antibody's true target. Indeed, it has previously been reported that treatment with the Rac inhibitor NCS23766 causes disassembly of the vimentin network (62). On the other hand, the data demonstrated by others showing that the antibody successfully precipitates Rac1 does raise the possibility that the antibody can recognize Rac1-GTP in addition to its primary vimentin-dependent epitope, presumably with lower affinity (10, 11, 22, 33, 54). However, it is clear from the present work that the primary epitope is not active Rac1. Furthermore, we were unable to immunoprecipitate Rac1 from PC3 cells using the anti-Rac1-GTP antibody (data not shown).

The primary epitope of this antibody being vimentin (or alternatively a vimentin-associated protein) may explain why the antibody has been extensively used without negative reports concerning its specificity. Vimentin is an intermediate filament often used as a marker to identify mesenchymal cells (63), which are characterized by increased migratory and invasive properties, functions known to be regulated by Rac1 (64). Moreover, it has recently been reported that active Rac1 levels are elevated in cancer cells that have undergone epithelial-to-mesenchymal transition (61). Therefore, it is unsurprising that this antibody can produce false positive results by primarily staining the highly motile and invasive vimentin-rich mesenchymal cells. We demonstrated the validity of this hypothesis using cancer models displaying high and low levels of Rac1-GTP and vimentin expression. The characterization of these two scenarios both in prostate and pancreatic cancer cells represents a compelling example of how vimentin expression can cause misleading conclusions where vimentin-dependent staining by this antibody is attributed to Rac1-GTP. In conclusion, we have demonstrated that without the appropriate controls, such as the use of Rac1 KO cells, the anti-Rac1-GTP antibody could be perceived as providing legitimate information on the level and localization of active Rac1 within cells. Yet, in reality, although the use of this antibody may be producing an expected result for the researcher, it is instead contributing deceptive information to the study of Rac1 activation in a variety of settings, cell types and diseases. Current assays such as the PBD pulldown assay and FRET reporter probes do not enable the visualization of endogenous active Rac1 in patient samples via immunohistochemistry or the high-resolution imaging of subcellular Rac1-GTP localization. Therefore, the nonspecific nature of this antibody creates a major gap in the toolbox for studying this GTPase, illustrating the importance of developing alternative robust methods for studying Rac activation.

Experimental procedures

Cell lines, cell culture, and reagents

Human prostate cancer cell lines (RWPE1, BPH-1, LNCaP, LNCaP-C4, LNCaP-C4-2, DU145, and PC3), pancreatic cancer cell lines (Capan-1, BxPC-3, AsPC-1, and PANC-1), and the non-small-cell lung cancer cell line A549 were obtained from ATCC (Manassas, VA). PC3-ML cells were obtained from Dr. Alessandro Fatatis (Drexel University), as described previously (65). EGF was purchased from R&D Systems (Minneapolis, MN; catalog no. AFL236).

Plasmid transfection

To express EGFP-Rac1Q61L, cells were transfected with a pcDNA3-EGFP-Rac1Q61L plasmid (Addgene, Watertown, MA) using Jet Optimus (Polyplus Transfection, Illkirch, France) following the manufacturer's protocol. Experiments were carried out 24 h after transfection.

Immunofluorescence

Cells were grown on glass coverslips for 24 h in complete medium followed by a further 24 h in starvation medium. After fixation in 3.6% formaldehyde/PBS and blocking with 10% goat serum/PBS, cells were incubated with the anti-Rac1-GTP antibody (catalog no. 26903, New East Biosciences, King of Prussia, PA) in 10% goat serum/PBS for 1 h. After washing with PBS (three quick washes followed by three additional 10-min washes), cells were incubated with Alexa Fluor 594–conjugated goat anti-mouse IgM secondary antibody (catalog no. A-21044, Thermo Fisher Scientific, Waltham, MA), used at a 1:500 dilution in 10% goat serum/PBS. For co-staining of vimentin, an anti-vimentin antibody (catalog no. NB300-223, Novus Biologicals, Centennial, CO) was used with an Alexa Fluor 488–conjugated goat anti-chicken secondary antibody (catalog no. A-11039, Thermo Fisher Scientific). Co-staining of filamentous actin was achieved with Alexa Fluor 488–conjugated phalloidin (catalog no. A12379, Thermo Fisher Scientific). Other co-staining experiments were as follows: for microtubules, we used an anti-α-tubulin antibody (catalog no. 6199, Millipore Sigma), and for Golgi, we used an anti-golgin-97 antibody (catalog no. A-21270, Thermo Fisher Scientific), both used with an Alexa Fluor 488–conjugated anti-mouse IgG secondary antibody (catalog no. A21121, Thermo Fisher Scientific). The endoplasmic reticulum was stained with anti-PDI (catalog no. 3501, Cell Signaling Technologies, Danvers, MA) and an Alexa Fluor 488–conjugated anti-rabbit secondary antibody (catalog no. A-11034, Thermo Fisher Scientific). For co-staining of mitochondria, we used Mitotracker Deep Red (catalog no. M22426, Thermo Fisher Scientific) and anti-Rac1-GTP with an Alexa Fluor 488–conjugated anti-mouse IgM secondary antibody to avoid signal bleed-through into the Mitotracker channel (catalog no. A21042, Thermo Fisher Scientific). Coverslips were mounted onto slides with Fluoromount-G (catalog no. 0100-01, SouthernBiotech, Birmingham, AL) and imaged using a Zeiss LSM 880 confocal with Airyscan module. EGF-stimulated A549 cells were stained with Hoechst, Alexa Fluor 488–conjugated phalloidin, anti-Rac1-GTP antibody with an Alexa Fluor 555–conjugated goat anti-mouse IgM secondary antibody (catalog no. A21426, Thermo Fisher Scientific), and anti-vimentin with an Alexa Fluor 647–conjugated goat anti-chicken IgM secondary antibody (catalog no. A32933, Thermo Fisher Scientific). Similar results for anti-Rac1-GTP staining were also obtained with other fixation/permeabilization methods, including methanol fixation and permeabilization, as well as when alternative blocking solutions were used (data not shown).

Vimentin shRNA silencing

Lentiviral particles targeting vimentin or pLKO nontargeting control particles were obtained from Sigma–Aldrich. pLKO.1 shRNAs for human vimentin (TRCN0000029119, TRCN0000029121) were used to generate PC3 and PANC-1 cell lines stably depleted of vimentin. Puromycin selection (1 μg/ml) was used to select for stably depleted vimentin cells.

Generation of Rac1 KO cell lines using CRISPR/Cas9

To generate the Rac1 KO PC3 and A549 cell lines the Gene KO kit 1 was purchased form Synthego (Menlo Park, CA) and used following the manufacturer's protocol. Briefly, an sgRNA targeted to exon 3 of Rac1 was complexed with a recombinant Cas9 protein and transfected into the cells using Lipofectamine CRISPRMAX (CMAX00001, Thermo Fisher Scientific). 2 days post-transfection, cells were seeded for single-cell clonal expansion. Rac1 KO was confirmed by Western blotting and Sanger sequencing.

PBD pulldown assays

Biochemical determination of active Rac1 was carried out using a PBD pulldown assay, as described previously (66, 67). Briefly, cells were seeded 48 h prior to the pulldown with a 24-h serum starvation immediately prior to assay. Cells were lysed in the presence of a recombinant Pak1-binding domain tagged to GST, and lysates were clarified and then incubated for 45 min in the presence of reduced GSH beads. Beads were washed twice with lysis buffer, and precipitated Rac1-GTP was assessed by Western blotting.

Western blotting

Western blotting was performed as described previously (68). The following antibodies were used at 1:1000 dilution: anti-Rac1 clone 23A8 (catalog no. 05-389, EMD Millipore, Burlington, MA) and anti-vimentin (catalog no. 3390, Cell Signaling Technologies, Danvers, MA). The following antibodies were used at 1:5000 dilution: anti-actin (catalog no. A544, Sigma–Aldrich) and anti-vinculin (catalog no. V9131, Sigma–Aldrich). Goat anti-mouse and goat anti-rabbit (catalog nos. 172-1017 and 170-6515, respectively, Bio-Rad) were used as secondary antibodies (1:3000 dilution). Bands were visualized using enhanced chemiluminescence with the LI-COR Odyssey Fc dual mode imaging system.

FRET acquisition and processing

Rac1 activation status was measured using a previously characterized dimerization-optimized reporter for activation (DORA) single-chain Rac1 biosensor (a generous gift from Yi Wu, UConn Health, Farmington, CT) (59). A549 cells were infected with an mCherry-Cortactin–encoding adenovirus (69) and a Rac1 biosensor–encoding lentivirus to visualize membrane ruffles and Rac1 activation status, respectively. Starved cells were treated with 50 ng/ml EGF to induce ruffle formation. We used 1:100 Oxyfluor reagent (Oxyrase Inc.) and 10 mm dl-lactate (Sigma–Aldrich) to reduce oxygen free radicals. Cells were imaged every 15 s with 2 × 2 binning and 16-bit depth using a dichroic splitter to facilitate simultaneous acquisition of Cerulean3 and Venus emissions. Raw images were processed using ImageJ. Images were corrected to account for background and bleaching, and a median filter with a 2-pixel radius was applied to reduce noise. The FRET ratio was calculated, and a custom LUT was applied to allow for the visualization of Rac1 activation.

Statistical analysis

Statistical significance was tested using paired Student's t test or, alternatively, when comparing more than two conditions, a one-way analysis of variance with Dunnett's multiple-comparison test was performed.

Data availability

All data are contained in the article.

Supplementary Material

Supporting Information

This article contains supporting information.

Author contributions—M. J. B., M. C., G. K.-L., R. G.-M., P. A. J., and M. G. K. conceptualization; M. J. B., R. G.-M., P. A. J., and M. G. K. formal analysis; M. J. B., R. G.-M., and M. G. K. funding acquisition; M. J. B., M. C., G. K.-L., and M. G. K. investigation; M. J. B. and M. G. K. visualization; M. J. B., M. C., G. K.-L., R. G.-M., P. A. J., and M. G. K. methodology; M. J. B., G. K.-L., and R. G.-M. writing-original draft; M. J. B. and M. G. K. project administration; M. J. B., P. A. J., and M. G. K. writing-review and editing; R. G.-M. resources; M. G. K. supervision.

Funding and additional information—This work was supported by National Institutes of Health Grants R01-CA189765, R01-CA196232, and R01-ES026023 (to M. G. K.) and 1R03CA234693-01 (to R. G.-M.) and United States Department of Defense Grant W81XWH1810274 (to M. J. B.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health or the Department of Defense.

Conflict of interestThe authors declare that they have no conflicts of interest with the contents of this article.

Abbreviations—The abbreviations used are:
PBD
Pak-binding domain
EGFP
enhanced green fluorescent protein
KO
knockout
PDI
protein-disulfide isomerase
Rac1
Ras-related C3 botulinum toxin substrate 1
EGF
epidermal growth factor
sgRNA
single guide RNA.

References

  • 1. Schaks M., Giannone G., and Rottner K. (2019) Actin dynamics in cell migration. Essays Biochem. 10.1042/EBC20190015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Svensmark J. H., and Brakebusch C. (2019) Rho GTPases in cancer: friend or foe? Oncogene 38, 7447–7456 10.1038/s41388-019-0963-7 [DOI] [PubMed] [Google Scholar]
  • 3. Casado-Medrano V., Baker M. J., Lopez-Haber C., Cooke M., Wang S., Caloca M. J., and Kazanietz M. G. (2018) The role of Rac in tumor susceptibility and disease progression: from biochemistry to the clinic. Biochem. Soc. Trans. 46, 1003–1012 10.1042/BST20170519 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. De P., Aske J. C., and Dey N. (2019) RAC1 takes the lead in solid tumors. Cells 8, 382 10.3390/cells8050382 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Bokoch G. M. (2003) Biology of the p21-activated kinases. Annu. Rev. Biochem. 72, 743–781 10.1146/annurev.biochem.72.121801.161742 [DOI] [PubMed] [Google Scholar]
  • 6. Itoh R. E., Kurokawa K., Ohba Y., Yoshizaki H., Mochizuki N., and Matsuda M. (2002) Activation of Rac and Cdc42 video imaged by fluorescent resonance energy transfer-based single-molecule probes in the membrane of living cells. Mol. Cell Biol. 22, 6582–6591 10.1128/mcb.22.18.6582-6591.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Welch C. M., Elliott H., Danuser G., and Hahn K. M. (2011) Imaging the coordination of multiple signalling activities in living cells. Nat. Rev. Mol. Cell Biol. 12, 749–756 10.1038/nrm3212 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Johnsson A. K. E., Dai Y., Nobis M., Baker M. J., McGhee E. J., Walker S., Schwarz J. P., Kadir S., Morton J. P., Myant K. B., Huels D. J., Segonds-Pichon A., Sansom O. J., Anderson K. I., Timpson P., et al. (2014) The Rac-FRET mouse reveals tight spatiotemporal control of Rac activity in primary cells and tissues. Cell Rep. 6, 1153–1164 10.1016/j.celrep.2014.02.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Matthews H. K., Marchant L., Carmona-Fontaine C., Kuriyama S., Larraín J., Holt M. R., Parsons M., and Mayor R. (2008) Directional migration of neural crest cells in vivo is regulated by Syndecan-4/Rac1 and non-canonical Wnt signaling/RhoA. Development 135, 1771–1780 10.1242/dev.017350 [DOI] [PubMed] [Google Scholar]
  • 10. Ward J. D., Ha J. H., Jayaraman M., and Dhanasekaran D. N. (2015) LPA-mediated migration of ovarian cancer cells involves translocalization of Gαi2 to invadopodia and association with Src and β-pix. Cancer Lett. 356, 382–391 10.1016/j.canlet.2014.09.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Jiang L., Mao R., Zhou Q., Yang Y., Cao J., Ding Y., Yang Y., Zhang X., Li L., and Xu L. (2016) Inhibition of Rac1 activity in the hippocampus impairs the forgetting of contextual fear memory. Mol. Neurobiol. 53, 1247–1253 10.1007/s12035-015-9093-6 [DOI] [PubMed] [Google Scholar]
  • 12. Biswas K., Yoshioka K., Asanuma K., Okamoto Y., Takuwa N., Sasaki T., and Takuwa Y. (2013) Essential role of class II phosphatidylinositol-3-kinase-C2α in sphingosine 1-phosphate receptor-1-mediated signaling and migration in endothelial cells. J. Biol. Chem. 288, 2325–2339 10.1074/jbc.M112.409656 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Higgins A. M., Banik B. L., and Brown J. L. (2015) Geometry sensing through POR1 regulates Rac1 activity controlling early osteoblast differentiation in response to nanofiber diameter. Integr. Biol. 7, 229–236 10.1039/c4ib00225c [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. De S., Tsimounis A., Chen X., and Rotenberg S. A. (2014) Phosphorylation of α-tubulin by protein kinase C stimulates microtubule dynamics in human breast cells. Cytoskeleton 71, 257–272 10.1002/cm.21167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Dai Y. K., Ma Y., Chen K., Mi Y. J., Fu H. L., Cui D. X., and Jin W. L. (2014) A link between the nuclear-localized srGAP3 and the SWI/SNF chromatin remodeler Brg1. Mol. Cell. Neurosci. 60, 10–25 10.1016/j.mcn.2014.02.005 [DOI] [PubMed] [Google Scholar]
  • 16. Parray A., Siddique H. R., Kuriger J. K., Mishra S. K., Rhim J. S., Nelson H. H., Aburatani H., Konety B. R., Koochekpour S., and Saleem M. (2014) ROBO1, a tumor suppressor and critical molecular barrier for localized tumor cells to acquire invasive phenotype: study in African-American and Caucasian prostate cancer models. Int. J. Cancer 135, 2493–2506 10.1002/ijc.28919 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Sharma N., Everingham S., Ramdas B., Kapur R., and Craig A. W. B. (2014) SHP2 phosphatase promotes mast cell chemotaxis toward stem cell factor via enhancing activation of the Lyn/Vav/Rac signaling axis. J. Immunol. 192, 4859–4866 10.4049/jimmunol.1301155 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Payne L. J., Eves R. L., Jia L., and Mak A. S. (2014) p53 Down regulates PDGF-induced formation of circular dorsal ruffles in rat aortic smooth muscle cells. PLoS ONE 9, e108257 10.1371/journal.pone.0108257 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Guilluy C., Zhang Z., Bhende P. M., Sharek L., Wang L., Burridge K., and Damania B. (2011) Latent KSHV infection increases the vascular permeability of human endothelial cells. Blood 118, 5344–5354 10.1182/blood-2011-03-341552 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Shoval I., and Kalcheim C. (2012) Antagonistic activities of Rho and Rac GTPases underlie the transition from neural crest delamination to migration. Dev. Dyn. 241, 1155–1168 10.1002/dvdy.23799 [DOI] [PubMed] [Google Scholar]
  • 21. Aslam M., Tanislav C., Troidl C., Schulz R., Hamm C., and Gündüz D. (2014) cAMP controls the restoration of endothelial barrier function after thrombin-induced hyperpermeability via Rac1 activation. Physiol. Rep. 2, e12175 10.14814/phy2.12175 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Frank S. R., Köllmann C. P., van Lidth de Jeude J. F., Thiagarajah J. R., Engelholm L. H., Frödin M., and Hansen S. H. (2017) The focal adhesion-associated proteins DOCK5 and GIT2 comprise a rheostat in control of epithelial invasion. Oncogene 36, 1816–1828 10.1038/onc.2016.345 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Long D. L., Willey J. S., and Loeser R. F. (2013) Rac1 is required for matrix metalloproteinase 13 production by chondrocytes in response to fibronectin fragments. Arthritis Rheum. 65, 1561–1568 10.1002/art.37922 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Hatanaka K., Simons M., and Murakami M. (2011) Phosphorylation of VE-cadherin controls endothelial phenotypes via p120-catenin coupling and Rac1 activation. Am. J. Physiol. Circ. Physiol. 300, H162–H172 10.1152/ajpheart.00650.2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Zhu S., Dai J., Liu H., Cong X., Chen Y., Wu Y., Hu H., Heng B. C., Ouyang H. W., and Zhou Y. (2015) Down-regulation of Rac GTPase-activating protein OCRL1 causes aberrant activation of Rac1 in osteoarthritis development. Arthritis Rheumatol. 67, 2154–2163 10.1002/art.39174 [DOI] [PubMed] [Google Scholar]
  • 26. Wang H., Fotheringham L., Wittchen E. S., and Hartnett M. E. (2015) Rap1 GTPase inhibits tumor necrosis factor-α–induced choroidal endothelial migration via NADPH oxidase– and NF-κB–dependent activation of Rac1. Am. J. Pathol. 185, 3316–3325 10.1016/j.ajpath.2015.08.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Haralalka S., Shelton C., Cartwright H. N., Katzfey E., Janzen E., and Abmayr S. M. (2011) Asymmetric Mbc, active Rac1 and F-actin foci in the fusion-competent myoblasts during myoblast fusion in Drosophila. Development 138, 1551–1562 10.1242/dev.057653 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. André-Grégoire G., Dilasser F., Chesné J., Braza F., Magnan A., Loirand G., and Sauzeau V. (2018) Targeting of Rac1 prevents bronchoconstriction and airway hyperresponsiveness. J. Allergy Clin. Immunol. 142, 824–833.e3 10.1016/j.jaci.2017.09.049 [DOI] [PubMed] [Google Scholar]
  • 29. Yang M., James A. D., Suman R., Kasprowicz R., Nelson M., O'Toole P. J., and Brackenbury W. J. (2020) Voltage-dependent activation of Rac1 by Nav1.5 channels promotes cell migration. J. Cell. Physiol. 235, 3950–3972 10.1002/jcp.29290 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Healy A., Berus J. M., Christensen J. L., Lee C., Mantsounga C., Dong W., Watts J. P., Assali M., Ceneri N., Nilson R., Neverson J., Wu W.-C., Choudhary G., and Morrison A. R. (2020) Statins disrupt macrophage Rac1 regulation leading to increased atherosclerotic plaque calcification. Arterioscler. Thromb. Vasc. Biol. 40, 713–732 10.1161/atvbaha.119.313832 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Stankiewicz T. R., Pena C., Bouchard R. J., and Linseman D. A. (2020) Dysregulation of Rac or Rho elicits death of motor neurons and activation of these GTPases is altered in the G93A mutant hSOD1 mouse model of amyotrophic lateral sclerosis. Neurobiol. Dis. 136, 104743 10.1016/j.nbd.2020.104743 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. McGuire S., Kara B., Hart P. C., Montag A., Wroblewski K., Fazal S., Huang X. Y., Lengyel E., and Kenny H. A. (2019) Inhibition of fascin in cancer and stromal cells blocks ovarian cancer metastasis. Gynecol. Oncol. 153, 405–415 10.1016/j.ygyno.2019.01.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Samuel M. S., Lourenço F. C., and Olson M. F. (2011) K-Ras mediated murine epidermal tumorigenesis is dependent upon and associated with elevated Rac1 activity. PLoS ONE 6, e17143 10.1371/journal.pone.0017143 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Ramadass M., Johnson J. L., Marki A., Zhang J., Wolf D., Kiosses W. B., Pestonjamasp K., Ley K., and Catz S. D. (2019) The trafficking protein JFC1 regulates Rac1-GTP localization at the uropod controlling neutrophil chemotaxis and in vivo migration. J. Leukoc. Biol. 105, 1209–1224 10.1002/JLB.1VMA0818-320R [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Winge M. C. G., Ohyama B., Dey C. N., Boxer L. M., Li W., Ehsani-Chimeh N., Truong A. K., Wu D., Armstrong A. W., Makino T., Davidson M., Starcevic D., Kislat A., Nguyen N. T., Hashimoto T., et al. (2016) RAC1 activation drives pathologic interactions between the epidermis and immune cells. J. Clin. Invest. 126, 2661–2677 10.1172/JCI85738 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Cao X., Kaneko T., Li J. S., Liu A.-D., Voss C., and Li S. S. C. (2015) A phosphorylation switch controls the spatiotemporal activation of Rho GTPases in directional cell migration. Nat. Commun. 6, 7721 10.1038/ncomms8721 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Logan C. M., Rajakaruna S., Bowen C., Radice G. L., Robinson M. L., and Menko A. S. (2017) N-cadherin regulates signaling mechanisms required for lens fiber cell elongation and lens morphogenesis. Dev. Biol. 428, 118–134 10.1016/j.ydbio.2017.05.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Taguchi K., Yamagishi S.-I., Yokoro M., Ito S., Kodama G., Kaida Y., Nakayama Y., Ando R., Yamada-Obara N., Asanuma K., Matsui T., Higashimoto Y., Brooks C. R., Ueda S., Okuda S., et al. (2018) RAGE-aptamer attenuates deoxycorticosterone acetate/salt-induced renal injury in mice. Sci. Rep. 8, 2686 10.1038/s41598-018-21176-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Davila J., Laws M. J., Kannan A., Li Q., Taylor R. N., Bagchi M. K., and Bagchi I. C. (2015) Rac1 regulates endometrial secretory function to control placental development. PLoS Genet. 11, e1005458 10.1371/journal.pgen.1005458 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Borin M., Saraceno C., Catania M., Lorenzetto E., Pontelli V., Paterlini A., Fostinelli S., Avesani A., Di Fede G., Zanusso G., Benussi L., Binetti G., Zorzan S., Ghidoni R., Buffelli M., et al. (2018) Rac1 activation links tau hyperphosphorylation and Aβ dysmetabolism in Alzheimer's disease. Acta Neuropathol. Commun. 6, 61 10.1186/s40478-018-0567-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Jamieson C., Lui C., Brocardo M. G., Martino-Echarri E., and Henderson B. R. (2015) Rac1 augments Wnt signaling by stimulating β-catenin-lymphoid enhancer factor-1 complex assembly independent of β-catenin nuclear import. J. Cell Sci. 128, 3933–3946 10.1242/jcs.167742 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Gonzalez-Villasana V., Fuentes-Mattei E., Ivan C., Dalton H. J., Rodriguez-Aguayo C., Fernandez-de Thomas R. J., Aslan B., del C., Monroig P., Velazquez-Torres G., Previs R. A., Pradeep S., Kahraman N., Wang H., Kanlikilicer P., Ozpolat. B., et al. (2015) Rac1/Pak1/p38/MMP-2 axis regulates angiogenesis in ovarian cancer. Clin. Cancer Res. 21, 2127–2137 10.1158/1078-0432.CCR-14-2279 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Jensen H. H., Login F. H., Park J.-Y., Kwon T.-H., and Nejsum L. N. (2017) Immunohistochemical evalulation of activated Ras and Rac1 as potential downstream effectors of aquaporin-5 in breast cancer in vivo. Biochem. Biophys. Res. Commun. 493, 1210–1216 10.1016/j.bbrc.2017.09.125 [DOI] [PubMed] [Google Scholar]
  • 44. Elnakish M. T., Moldovan L., Khan M., Hassanain H. H., and Janssen P. M. L. (2013) Myocardial Rac1 exhibits partial involvement in thyroxin-induced cardiomyocyte hypertrophy and its inhibition is not sufficient to improve cardiac dysfunction or contractile abnormalities in mouse papillary muscles. J. Cardiovasc. Pharmacol. 61, 536–544 10.1097/FJC.0b013e31828d4b9d [DOI] [PubMed] [Google Scholar]
  • 45. Liu W., Han J., Shi S., Dai Y., and He J. (2019) TUFT1 promotes metastasis and chemoresistance in triple negative breast cancer through the TUFT1/Rab5/Rac1 pathway. Cancer Cell Int. 19, 1–14 10.1186/s12935-018-0719-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Rao J., Zhou Z., Yang J., Shi Y., Xu S., Wang B., Ping Y., Chen L., Cui Y., Zhang X., Wu F., and Bian X. (2015) Semaphorin-3F suppresses the stemness of colorectal cancer cells by inactivating Rac1. Cancer Lett. 358, 76–84 10.1016/j.canlet.2014.12.040 [DOI] [PubMed] [Google Scholar]
  • 47. Reviriego-Mendoza M. M., and Santy L. C. (2015) The cytohesin guanosine exchange factors (GEFs) are required to promote HGF-mediated renal recovery after acute kidney injury (AKI) in mice. Physiol. Rep. 3, e12442 10.14814/phy2.12442 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Tamasas B., and Cox T. C. (2017) Massively increased caries susceptibility in an Irf6 cleft lip/palate model. J. Dent. Res. 96, 315–322 10.1177/0022034516679376 [DOI] [PubMed] [Google Scholar]
  • 49. Zhou K., Rao J., Zhou Z., Yao X., Wu F., Yang J., Yang L., Zhang X., Cui Y., Bian X.-W., Shi Y., and Ping Y. (2018) RAC1-GTP promotes epithelial-mesenchymal transition and invasion of colorectal cancer by activation of STAT3. Lab. Investig. 98, 989–998 10.1038/s41374-018-0071-2 [DOI] [PubMed] [Google Scholar]
  • 50. Gould R. A., Yalcin H. C., MacKay J. L., Sauls K., Norris R., Kumar S., and Butcher J. T. (2016) Cyclic mechanical loading is essential for Rac1-mediated elongation and remodeling of the embryonic mitral valve. Curr. Biol. 26, 27–37 10.1016/j.cub.2015.11.033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Danopoulos S., Krainock M., Toubat O., Thornton M., Grubbs B., and Al Alam D. (2016) Rac1 modulates mammalian lung branching morphogenesis in part through canonical Wnt signaling. Am. J. Physiol. Lung Cell. Mol. Physiol. 311, L1036–L1049 10.1152/ajplung.00274.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Göppner D., Müller J., Krüger S., Franke I., Gollnick H., and Quist S. (2014) High incidence of naevi-associated BRAF wild-type melanoma and dysplastic naevi under treatment with the class I BRAF inhibitor vemurafenib. Acta Derm. Venereol. 94, 517–520 10.2340/00015555-1813 [DOI] [PubMed] [Google Scholar]
  • 53. Laufer J. M., Hauser M. A., Kindinger I., Purvanov V., Pauli A., and Legler D. F. (2019) Chemokine receptor CCR7 triggers an endomembrane signaling complex for spatial Rac activation. Cell Rep. 29, 995–1009.e6 10.1016/j.celrep.2019.09.031 [DOI] [PubMed] [Google Scholar]
  • 54. He S., Zhao Z., Yang Y., O'Connell D., Zhang X., Oh S., Ma B., Lee J.-H., Zhang T., Varghese B., Yip J., Dolatshahi Pirooz S., Li M., Zhang Y., Li G.-M., et al. (2015) Truncating mutation in the autophagy gene UVRAG confers oncogenic properties and chemosensitivity in colorectal cancers. Nat. Commun. 6, 7839 10.1038/ncomms8839 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Fan M., Xu Y., Hong F., Gao X., Xin G., Hong H., Dong L., and Zhao X. (2016) Rac1/β-catenin signalling pathway contributes to trophoblast cell invasion by targeting Snail and MMP9. Cell. Physiol. Biochem. 38, 1319–1332 10.1159/000443076 [DOI] [PubMed] [Google Scholar]
  • 56. Baker M. J., Abba M. C., Garcia-Mata R., and Kazanietz M. G. (2020) P-REX1-independent, calcium-dependent RAC1 hyperactivation in prostate cancer. Cancers (Basel) 12, 480 10.3390/cancers12020480 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Cooke M., Baker M. J., and Kazanietz M. G. (2020) Rac-GEF/Rac signaling and metastatic dissemination in lung cancer. Front. Cell Dev. Biol. 8, 118 10.3389/fcell.2020.00118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Caino M. C., Lopez-Haber C., Kissil J. L., and Kazanietz M. G. (2012) Non-small cell lung carcinoma cell motility, Rac activation and metastatic dissemination are mediated by protein kinase C ε. PLoS ONE 7, e31714 10.1371/journal.pone.0031714 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Timmerman I., Heemskerk N., Kroon J., Schaefer A., van Rijssel J., Hoogenboezem M., van Unen J., Goedhart J., Gadella T. W. J., Yin T., Wu Y., Huveneers S., and van Buul J. D. (2015) A local VE-cadherin and Trio-based signaling complex stabilizes endothelial junctions through Rac1. J. Cell Sci. 128, 3041–3054 10.1242/jcs.168674 [DOI] [PubMed] [Google Scholar]
  • 60. Zeisberg M., and Neilson E. G. (2009) Biomarkers for epithelial-mesenchymal transitions. J. Clin. Invest. 119, 1429–1437 10.1172/JCI36183 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Casado-Medrano V., Barrio-Real L., Wang A., Cooke M., Lopez-Haber C., and Kazanietz M. G. (2019) Distinctive requirement of PKCε in the control of Rho GTPases in epithelial and mesenchymally transformed lung cancer cells. Oncogene 38, 5396–5412 10.1038/s41388-019-0796-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Murray M. E., Mendez M. G., and Janmey P. A. (2014) Substrate stiffness regulates solubility of cellular vimentin. Mol. Biol. Cell 25, 87–94 10.1091/mbc.E13-06-0326 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Odero-Marah V., Hawsawi O., Henderson V., and Sweeney J. (2018) Epithelial-Mesenchymal Transition (EMT) and Prostate Cancer, pp. 101–110, Springer, Cham, Switzerland: [DOI] [PubMed] [Google Scholar]
  • 64. Kazanietz M. G., and Caloca M. J. (2017) The Rac GTPase in cancer: from old concepts to new paradigms. Cancer Res. 77, 5445–5451 10.1158/0008-5472.CAN-17-1456 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Gutierrez-Uzquiza A., Lopez-Haber C., Jernigan D. L., Fatatis A., and Kazanietz M. G. (2015) PKC is an essential mediator of prostate cancer bone metastasis. Mol. Cancer Res. 13, 1336–1346 10.1158/1541-7786.MCR-15-0111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Benard V., Bohl B. P., and Bokoch G. M. (1999) Characterization of Rac and Cdc42 activation in chemoattractant-stimulated human neutrophils using a novel assay for active GTPases. J. Biol. Chem. 274, 13198–13204 10.1074/jbc.274.19.13198 [DOI] [PubMed] [Google Scholar]
  • 67. Caloca M. J., Wang H., and Kazanietz M. G. (2003) Characterization of the Rac-GAP (Rac-GTPase-activating protein) activity of β2-chimaerin, a “non-protein kinase C” phorbol ester receptor. Biochem. J. 375, 313–321 10.1042/BJ20030727 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Lopez-Haber C., and Kazanietz M. G. (2013) Cucurbitacin I inhibits Rac1 activation in breast cancer cells by a reactive oxygen species-mediated mechanism and independently of Janus tyrosine kinase 2 and P-Rex1. Mol. Pharmacol. 83, 1141–1154 10.1124/mol.112.084293 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Goicoechea S. M., Zinn A., Awadia S. S., Snyder K., and Garcia-Mata R. (2017) A RhoG-mediated signaling pathway that modulates invadopodia dynamics in breast cancer cells. J. Cell. Sci. 130, 1064–1077 10.1242/jcs.195552 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supporting Information
Download video file (6.1MB, avi)

Data Availability Statement

All data are contained in the article.


Articles from The Journal of Biological Chemistry are provided here courtesy of American Society for Biochemistry and Molecular Biology

RESOURCES