12
RasIns: Genetically Encoded Intrabodies of Activated Ras Proteins Mehmet Cetin 1 , William E. Evenson 2 , Garrett G. Gross 1 , Farzad Jalali-Yazdi 3 , Daniel Krieger 2 , Don Arnold 1 , Terry T. Takahashi 2 and Richard W. Roberts 1,2,3,4 1 - Department of Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA 2 - Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA 3 - Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA 4 - USC Norris Comprehensive Cancer Center, Los Angeles, CA 90089, USA Correspondence to Richard W. Roberts: Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA. [email protected] http://dx.doi.org/10.1016/j.jmb.2016.11.008 Edited by Sachdev Sidhu Abstract K- and H-Ras are the most commonly mutated genes in human tumors and are critical for conferring and maintaining the oncogenic phenotype in tumors with poor prognoses. Here, we design genetically encoded antibody-like ligands (intrabodies) that recognize active, GTP-bound K- and H-Ras. These ligands, which use the 10th domain of human fibronectin as their scaffold, are stable inside the cells and when fused with a fluorescent protein label, the constitutively active G12V mutant H-Ras. Primary selection of ligands against Ras with mRNA display resulted in an intrabody (termed RasIn1) that binds with a K D of 2.1 μM to H-Ras(G12V) (GTP), excellent state selectivity, and remarkable specificity for K- and H-Ras. RasIn1 recognizes residues in the Switch I region of Ras, similar to Raf-RBD, and competes with Raf-RBD for binding. An affinity maturation selection based on RasIn1 resulted in RasIn2, which binds with a K D of 120 nM and also retains excellent state selectivity. Both of these intrabodies colocalize with H-Ras, K-Ras, and G12V mutants inside the cells, providing new potential tools to monitor and modulate Ras-mediated signaling. Finally, RasIn1 and Rasin2 both display selectivity for the G12V mutants as compared with wild-type Ras providing a potential route for mutant selective recognition of Ras. © 2016 Published by Elsevier Ltd. Introduction K- and H-Ras have been identified as mediators of malignant characteristics [1,2], and oncogenic mutations in these proteins are commonly observed [3,4]. Missense mutations in human Ras were some of the first oncogenes ever discovered, with two groups reporting that the G12V mutation in H-Ras transforms cell lines [5,6]. Ras overactivation is associated with 71% of pancreatic cancers, 35% of colon cancers, and 19% of lung cancers [4]. Thus, molecules that could regulate oncogenic Ras func- tion would be of immense value as targeted ther- apeutics. However, despite decades of extensive research, there are currently no therapies directly targeting Ras. As a result, Ras continues to be con- sidered as an undruggable target[7]. Recent attempts to inhibit Ras signaling have focused on blocking the Switch regions of Ras to prevent Ras from interacting with downstream ef- fectors. Antibodies [8], compounds that covalently modify mutant Ras [9], and cyclic peptides [10,11] that bind to Ras have been generated, and many of these molecules can disrupt Ras signaling. However, while some these molecules are able to modulate Ras activity under certain conditions, each has drawbacks that limit their potential use, including a lack of state specificity, off-target effects, large size, and/or an inability to be effective inside the cells. We have previously used mRNA display to target G-proteins in a state-specific fashion [1214]. We have also developed mRNA display methods for generating fibronectins with high affinity to specific 0022-2836/© 2016 Published by Elsevier Ltd. J Mol Biol (2017) 429, 562573 Article

RasIns: Genetically Encoded Intrabodies of Activated Ras ......RasIns: Genetically Encoded Intrabodies of Activated Ras Proteins Mehmet Cetin1, William E. Evenson2, Garrett G. Gross1,

  • Upload
    others

  • View
    12

  • Download
    0

Embed Size (px)

Citation preview

Page 1: RasIns: Genetically Encoded Intrabodies of Activated Ras ......RasIns: Genetically Encoded Intrabodies of Activated Ras Proteins Mehmet Cetin1, William E. Evenson2, Garrett G. Gross1,

RasIns: Genetically Encoded Intrabodies ofActivated Ras Proteins

Mehmet Cetin 1, William E. Evenson 2, Garrett G. Gross 1, Farzad Jalali-Yazdi 3,Daniel Krieger 2, Don Arnold 1, Terry T. Takahashi 2 and Richard W. Roberts 1, 2, 3, 4

1 - Department of Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA

2 - Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA

3 - Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA

90089, USA

4 - USC Norris Comprehensive Cancer Center, Los Angeles, CA 90089, USA

Correspondence to Richard W. Roberts: Mork Family Department of Chemical Engineering and Materials Science,

University of Southern California, Los Angeles, CA 90089, USA. [email protected]

http://dx.doi.org/10.1016/j.jmb.2016.11.008

Edited by Sachdev Sidhu

Abstract

K- and H-Ras are the most commonly mutated genes in human tumors and are critical for conferring andmaintaining the oncogenic phenotype in tumors with poor prognoses. Here, we design genetically encodedantibody-like ligands (intrabodies) that recognize active, GTP-bound K- and H-Ras. These ligands, whichuse the 10th domain of human fibronectin as their scaffold, are stable inside the cells and when fused witha fluorescent protein label, the constitutively active G12V mutant H-Ras. Primary selection of ligands againstRas with mRNA display resulted in an intrabody (termed RasIn1) that binds with a KD of 2.1 μM toH-Ras(G12V) (GTP), excellent state selectivity, and remarkable specificity for K- and H-Ras. RasIn1recognizes residues in the Switch I region of Ras, similar to Raf-RBD, and competes with Raf-RBD for binding.An affinity maturation selection based on RasIn1 resulted in RasIn2, which binds with a KD of 120 nM and alsoretains excellent state selectivity. Both of these intrabodies colocalize with H-Ras, K-Ras, and G12V mutantsinside the cells, providing new potential tools to monitor and modulate Ras-mediated signaling. Finally, RasIn1and Rasin2 both display selectivity for the G12V mutants as compared with wild-type Ras providing a potentialroute for mutant selective recognition of Ras.

© 2016 Published by Elsevier Ltd.

Introduction

K- and H-Ras have been identified as mediatorsof malignant characteristics [1,2], and oncogenicmutations in these proteins are commonly observed[3,4]. Missense mutations in human Ras were someof the first oncogenes ever discovered, with twogroups reporting that the G12V mutation in H-Rastransforms cell lines [5,6]. Ras overactivation isassociated with 71% of pancreatic cancers, 35% ofcolon cancers, and 19% of lung cancers [4]. Thus,molecules that could regulate oncogenic Ras func-tion would be of immense value as targeted ther-apeutics. However, despite decades of extensiveresearch, there are currently no therapies directlytargeting Ras. As a result, Ras continues to be con-sidered as an “undruggable target” [7].

Recent attempts to inhibit Ras signaling havefocused on blocking the Switch regions of Ras toprevent Ras from interacting with downstream ef-fectors. Antibodies [8], compounds that covalentlymodify mutant Ras [9], and cyclic peptides [10,11]that bind to Ras have been generated, and manyof these molecules can disrupt Ras signaling.However, while some these molecules are ableto modulate Ras activity under certain conditions,each has drawbacks that limit their potential use,including a lack of state specificity, off-target effects,large size, and/or an inability to be effective insidethe cells.We have previously used mRNA display to target

G-proteins in a state-specific fashion [12–14]. Wehave also developed mRNA display methods forgenerating fibronectins with high affinity to specific

0022-2836/© 2016 Published by Elsevier Ltd. J Mol Biol (2017) 429, 562–573

Article

Page 2: RasIns: Genetically Encoded Intrabodies of Activated Ras ......RasIns: Genetically Encoded Intrabodies of Activated Ras Proteins Mehmet Cetin1, William E. Evenson2, Garrett G. Gross1,

targets [15,16]. The fibronectin scaffold (10FnIII)is based on the 10th fibronectin type III domain ofhuman fibronectin [17] and has an immunoglobulin-like fold with loops that are structurally similar toantibody CDRH1 and CDRH3 regions. The 10FnIIIscaffold is small (10 kDa), lacks disulfides, can beexpressed in Escherichia coli, and is an alternativeto antibodies for generating affinity reagents andintrabodies [16–23]. In addition, we evolved aversion of 10FnIII, termed e10FnIII, which improvessolubility and expression in E. coli and in thereticulocyte lysate translation system [15,24].Here, we describe the generation of two “RasIns”,

e10FnIII-based and disulfide-free intrabodies againstactivated (GTP-bound) Ras proteins. These fibro-nectins are selective for the active state of Rasand are specific for Ras over homologous proteins.At the same time, these fibronectins bind both H- andK-Ras-GTP selectively for mutants. Furthermore,Rasin1 and Rasin2 are functional when expressedinside the cells, and RasIn2 and Raf-kinase RBDdomain (Raf-RBD) (the canonical binding partnerof Ras [25]) bind Ras-GTP with comparable affinity.

Results

An mRNA display selection againstK-Ras(G12V)-GTP

A major goal of this work was to develop state-specific Ras ligands. In order to enhance the target-ing of the active state of Ras, our fibronectin librarywas constructed with a mutagenized sequence

(CDRH1) derived from the known K-Ras binder,iDab#6 [26], in the BC loop and a totally random FGloop (Fig. 1a). The initial diversity of the library was~1012 independent sequences (~5 copies each).mRNA display selection was performed againsthuman K-Ras(G12V) that had been exchangedwith GTPγS to create bias for binders to the activestate of the target (Fig. 1b). Target specific bindingwas first detected in round 4 (Fig. 1b). To increasestate-selective binding for active Ras, we addeda 15-fold molar excess of K-Ras(G12V)-GDP insolution that would compete and remove moleculesthat were not specific for active Ras during theaffinity enrichment step in rounds 5 and 6. Theresulting pool 6 has 23% binding to K-Ras(G12V)-GTP, 16% binding to K-Ras(G12V)-GDP, and no de-tectable binding to the selection matrix (neutravidin-agarose or streptavidin–agarose beads; Fig. 1b).Thus, pool 6 shows a measurable preference forthe GTP-bound form of K-Ras(G12V). We then se-quenced pool 6 using high-throughput sequencingtechniques and ranked the selected sequences bytheir abundance to determine the sequences thatwere enriched during selection (data not shown).

Isolation of RasIn1 via a cell-based screen

Ten high abundance sequences were tested forfunction using a cell-based screen we had previouslydeveloped to monitor in vivo protein binding andcolocalization [21]. In this assay, the target proteinis localized to the Golgi as a fusion protein bearing(1) a Golgi-targeting sequence (GTS), (2) streptavi-din (SA), and (3) the target [here, H-Ras(G12V)]. We

Fig. 1. An mRNA display selection against K-Ras(G12V)-GTPγS using the e10FnIII scaffold. (a) e10FnIII scaffold andlibrary design (PDB ID:1FNF). The BC loop is shown in blue, while the FG loop is shown in red. Backbone mutationsrelative to wild-type 10FnIII are shown in yellow [15,24]. In order to direct the library to recognize active Ras, weconstructed variants of e10FnIII where the BC loop was a doped sequence from a previous Ras ligand (iDab#6) [8]. BCloop doping was ~40% wild-type at each residue. The FG loop was a naïve random sequence. (b) In vitro selection forK-Ras(G12V)-GTPγS. Pool binding was measured for matrix without target (neutravidin or streptavidin agarose) or forimmobilized K-Ras exchanged with GTPγS or GDP. No binding is observed in matrix without target, while preferentialbinding to K-Ras(GTPγS) over K-Ras(GDP) is observed in Round 6. (c) Individual clones were screened for functioninside the cells using a cellular colocalization assay [21]. Scale bar represents 5 μm. (d) Screening resulted in theidentification of a Ras-specific clone termed RasIn1.

563Intrabodies of Activated Ras Proteins

Page 3: RasIns: Genetically Encoded Intrabodies of Activated Ras ......RasIns: Genetically Encoded Intrabodies of Activated Ras Proteins Mehmet Cetin1, William E. Evenson2, Garrett G. Gross1,

chose H-Ras for this screen to find those moleculesthat were Ras specific and would be able to bind toboth K- and H-Ras. We also utilized the constitu-tively active H-Ras(G12V) mutant in order to findstate-specific Ras binders that would not be down-regulated by endogenous RasGAP that might bepresent in the COS cells (CV-1 in Origin, and carryingSV40 genetic material). Each candidate wasscreened as an eGFP fusion that was co-transfectedwith the target. Candidates were scored for colocali-zation with the target by immunocytochemistry. Thebest-performing candidate targeting H-Ras(G12V)was termed RasIn1 (Fig. 1c and d). Images of theCOS cells demonstrate that eGFP-labeled RasIn1(green) and Golgi-targeted SA-H-Ras(G12V) (visual-ized with rhodamine-biotin; red) accurately colocalize(merged, yellow) with little excess staining elsewherein the cell. This result indicates that RasIn1 (1)expresses stably and functionally in mammaliancells (COS cells) and (2) has low background andlittle, nonspecific localization in the rest of the cell.Importantly, this low background binding demon-strates that RasIn1 accurately recognizes H-Ras inthe complex environment inside the cytosol.

RasIn1 is selective for the GTP-bound state,competes with Raf-RBD, and is sensitive tomutation of Switch I

RasIn1 shows efficient pull-down with the active(GTP-bound) form of K-Ras and less pull-down withK-Ras-GDP or beads that lack target (Fig. 2a).To determine where RasIn1 binds K-Ras, we per-formed competition binding experimentswith a naturalRas binding partner, the Raf-RBD. Coincubation ofRasIn1 with a molar excess of Raf-RBD decreasesthe binding 12-fold (Fig. 2a), consistent with the twoproteins competing for the same binding interfaceon Ras.Co-crystal structures of H-Ras with Raf-RBD

(PDBID: 3KUD [27]) make it clear that Raf-RBD

makes direct contacts with the Switch I region ofRas, proximal to the nucleotide-binding pocket. TheY32R mutation in the Switch I region of Ras haspreviously been shown to decrease the bindingbetween Ras and Raf-RBD [25]. This mutation alsodecreases the binding of Ras with Rasin1 (Fig. 2b).Taken together, our data argue that RasIn1 bindsRas in a state-specific fashion via recognition ofthe Switch I region of Ras in a functionally similarfashion to Raf-RBD.

RasIn1 is specific for K- and H-Ras anddiscriminates between highly homologousmembers of the Ras superfamily

K- and H-Ras are members of the Ras subfamilywith high sequence homology and identical Switch Isequences [28,29]. To further characterize thespecificity of RasIn1, we tested RasIn1 binding towild-type K-Ras, K-Ras(G12V), wild-type H-Ras, andH-Ras(G12V) (Fig. 3a). We observed mutant-selec-tive binding for both K- and H-Ras (GTP). We alsoobserve similar binding to both H- and K- Ras(G12V)and to wild-type H- and K-Ras, indicating that RasIn1is not selective for a particular isoform of Ras.The Ras superfamily is composed of five subfam-

ilies with related sequence and structure [28,29].To demonstrate that RasIn1 specifically recognizesRas, we tested RasIn1 against two other membersof the Ras superfamily using a radioactive in vitropull-down assay (Fig. 3b). RasIn1 shows little to nobinding to Rap1B(G12V)-GTP and Arf1-GTP. Theability to discriminate betweenRas andRap1B(G12V)bears comment, as these proteins are 57% sequenceidentical and the Ras Switch I region (YPDTIED)differs by switching 2 aa from the Rap1B Switch Iregion (YDPTIED). The Ras Switch I sequence is alsosimilar to the Switch I sequence of Arf1 (TIPTIGF).RasIn1 is thus mutant specific (G12V versus wild-type) and state specific (GTP versus GDP) and dif-ferentiates between close Ras homologs.

Fig. 2. Assessing the bindingcharacteristics of RasIn1 in vitro.(a) RasIn1 preferentially bindsactive (GTP) over inactive (GDP)forms of H-Ras(G12V) (p = 0.007)and preferentially binds unblockedactive H-Ras(G12V) as activeH-Ras(G12V) blocked with thec -Ra f -k inase RBD doma in(Raf-RBD; p = 0.002). (b) Bindingof RasIn1 to K-Ras is disrupted bythe Y32R mutation in the Switch Iregion (p = 0.03). Error bars indi-cate the standard deviation of themean.

564 Intrabodies of Activated Ras Proteins

Page 4: RasIns: Genetically Encoded Intrabodies of Activated Ras ......RasIns: Genetically Encoded Intrabodies of Activated Ras Proteins Mehmet Cetin1, William E. Evenson2, Garrett G. Gross1,

RasIn1 colocalizes with active Ras in vivo

Our in vitro data indicate that RasIn1 binds toboth wild-type and mutant activated Ras proteins(Fig. 3a). Using the COS cell assay (Fig. 1c), wetested if RasIn1 could also recognize wild-typeH-Ras and H-Ras(G12V) inside the cells (Fig. 4).Colocalization of RasIn1 is not seen when Golgi-bound target is not expressed (Fig. 4a–c).While RasIn1 binds to both H-Ras-GTP and

H-Ras(G12V)-GTP in vitro (Fig. 3a), we onlyobserve colocalization in the COS cell assay withH-Ras(G12V). In the cell-based assay, wild-typeH-Ras shows poor binding and colocalization, whilethe H-Ras(G12V) mutant shows good colocalizationand binding (Fig. 4). One possible explanation forthis observation is that RasIn1 has higher binding forH-Ras(G12V) than wild-type H-Ras. Alternatively,wild-type H-Ras may exist in the cell in a predom-inantly inactive, GDP-bound state due to downreg-ulation by endogenous Ras-GAP. This hypothesisis consistent with the wild-type protein being asubstrate for hydrolysis via endogenous Ras-GAP,whereas the mutant is not [30].

Mutational analysis of RasIn1

We next constructed several mutants of theRasIn1 loops to determine functionally importantresidues (Fig. 5a). In the BC loop, mutation of Ser-21to Ala (S21A) results in little decrease in pull-downefficiency, suggesting that this residue is not criticalfor the function of the intrabody. On the other hand,the S24A mutation results in a significant decreasein binding, while the S24R mutation that increasessteric bulk at this position results in complete loss ofbinding. These data suggest a specific role for serineat this position. In the FG loop, the R72A and R77Amutations also abolish binding completely, suggest-ing that these two residues are critical for the function

of RasIn1. Likewise, R80A also shows a significantdecrease in binding.High-throughput sequencing also affords new in-

sights into positional scanning. In the high-throughputsequencing data, we observed both the primaryclone, RasIn1, and a number of sequences that dif-fer from this clone by a single mutation (Fig. 5b).These mutations likely occurred during the courseof the selection, due to inherent error rates in PCR,transcription, and reverse transcription within eachmRNAdisplay selection round. Analysis of these pointmutants reveals 100% conservation at positions 22,72, 74, 76, and 77. This point mutational analysisagrees with and complements the mutational analysisabove, as both analyses identify R72 and R77 ashighly important residues for binding. In addition, pointmutational analysis also revealed potentially impor-tant contact sites thatwere not explored in the directedmutational studies, such as A22 and P74. Takentogether, the positional scanning data and the high-throughput sequencing mutagenesis data argue thata significant fraction of the residues on the BC andFG loops are involved and important for binding andrecognition of active Ras.

Affinity maturation results in a high affinitybinder, RasIn2

An affinity maturation selection was done to im-prove the affinity of RasIn1. A doped mRNA displaylibrary was constructed based on the loop se-quences of RasIn1 such that there was an averageof 33% wild-type amino acid at each position in thestarting pool. We performed five rounds of affinityenrichment against H-Ras(G12V) in the presence ofGppNHp, which, like GTPγS, allowed us to introducebias for the active state of Ras (Fig. 6a). Additionally,during rounds 4 and 5, the binding temperatureduring selection was raised to 37 °C to increaseselective pressure and to select for sequences with

Fig. 3. Binding of RasIn1 to different Ras homologs. (a) Radiolabeled RasIn1 shows excellent binding to differentversions of Ras (wild-type K- or H-Ras, and G12Vmutant K- or H-Ras), showing that the recognition of the active Ras stateis robust. (b) Binding specificity of RasIn1 against homologous members of the Ras superfamily. RasIn1 binds specificallyto active K-Ras, but less to homologous Ras family members Rap1B(G12V) (p = 0.01) or Arf1 (0.02) (percentage ofsequence identity to the K-Ras G domain is shown in parentheses). Error bars indicate the standard deviation of the mean.

565Intrabodies of Activated Ras Proteins

Page 5: RasIns: Genetically Encoded Intrabodies of Activated Ras ......RasIns: Genetically Encoded Intrabodies of Activated Ras Proteins Mehmet Cetin1, William E. Evenson2, Garrett G. Gross1,

Fig. 5. Mutational analysis of RasIn1 reveals functionally important residues. (a) In vitro pull-down efficiency of RasIn1point mutants. Radiolabeled point mutants of RasIn1 were tested for binding to immobilized K-Ras-GTP. The percentageof radioactive counts bound to beads is shown. S24, R72, and R77 are critical positions for binding. (b) BC and FG loopmutations observed in the top 20 single point mutants of RasIn1 in high-throughput sequencing of pool 6 (Fig. S2). Residuenumbers are shown above the sequence.

Fig. 4. Colocalization of RasIn1 (green) with various Ras forms (red) in COS-7 cells. RasIn1-EGFP was co-transfectedwith Golgi-targeting sequence-streptavidin (GTS-SA) (a–c), GTS-SA-wild-type H-Ras (d–f), or GTS-SA-H-Ras(G12V)(g–i). Cells were fixed and stained for EGFP (green) and streptavidin (red). Colocalization (yellow) is indicated in themerged image. Images are representative of at least 15 samples. Scale bar represents 5 μm.

566 Intrabodies of Activated Ras Proteins

Page 6: RasIns: Genetically Encoded Intrabodies of Activated Ras ......RasIns: Genetically Encoded Intrabodies of Activated Ras Proteins Mehmet Cetin1, William E. Evenson2, Garrett G. Gross1,

higher affinity and stability. Pool 5 correspondinglyshowed a pull-down efficiency of 65%, indicating thepresence of high affinity molecules in the pool.Pool 5 of the maturation selection was analyzed by

high-throughput sequencing. The four highest abun-dance clones (Clones 1–4) were selected for furthercharacterization. All clones contain V19 and F20in the BC loops and highly conserved FG loops,with the only variation in FG occurring at position 79(V and L). The high sequence conservation suggeststhat all the clones bind to the same epitope of Ras asRasIn1. These four clones can be further groupedinto two pairs (1 and 4 versus 2 and 3) based onthe similarity of each clone's BC loop (Fig. 6c). Fiveout of seven positions in the BC loop are identicalbetween the two respective clones in both groups.We then tested the binding of all four clones in

a radioactive pull-down assay (Fig. 6b). All clonesgive high levels of binding compared to the parentalRasIn1 clone (N70% versus ~5%, respectively) andalso show very good selectivity for Ras-GTP versusRas-GDP (Fig. 6). We note that RasIn1 shows lowerpull-down in this experiment (~5%; Fig. 6b) thatuses a relatively small amount of target, whereasin Figs. 2–5, RasIn1 gave a much higher pull-downefficiency due to the much higher target concentrationon the beads that were used in those experiments.From these experiments, we chose the highest affinitysequence (Clone 3), termed it RasIn2, and furtheranalyzed the clone.

Characterization of RasIn2

RasIn2 maintained many of the binding charac-teristics of its parent clone, RasIn1. RasIn2 wasmatured on H-Ras(G12V) and shows a preferencefor H-Ras and H-Ras(G12V) over wild-type K-Rasand K-Ras(G12V) (Fig. 7a). RasIn2 is state selectiveand preferentially binds active Ras (Fig. 7b). RasIn2competes with Raf-RBD (Fig. 7b) and selectivelyrecognizes K- and H-Ras over Ras family proteinsArf1 and Rap1B (Fig. 7c). RasIn2 also has reducedbinding to the Y32R Switch I mutant (Fig. 7d).

Comparison of the affinity-matured clone, RasIn2,to its parent, RasIn1, allows the identification ofresidues that are important for binding. Affinity matu-ration resulted in the conservation of 8 of the 10residues in the FG loop, demonstrating that theseresidues are highly optimized for binding. Theseconserved residues agree with the mutational anal-ysis from alanine scanning and from point mutationalanalysis from high-throughput sequencing (Fig. 5b).However, only two positions are retained in theBC loop after affinity maturation, arguing that the BCloop interactions have been optimized in the contextof the new FG loop in the affinity-matured clone andpotentially yielded the isoform selectivity of RasIn2for H-Ras.We then determined the binding affinity of the two

intrabodies by surface plasmon resonance (SPR;Figs. 7e, S3, and S4). These experiments indicatethat RasIn1 binds H-Ras(G12V) with a KD of 2.1 μM,while RasIn2 binds with a KD of 120 nM. Thus,affinity maturation provided a nearly 20-fold increasein affinity, an increase of about −1.7 kcal-mol -1,versus the parent clone. This affinity is notablebecause it is similar to the reported KD valuefor the Raf-RBD:wild-type H-Ras-GTP complex(KD = 80 nM) [31]. Furthermore, the fact thatRaf-RBD binds H-Ras(G12V) with lower affinitythan wild-type H-Ras indicates that RasIn2 may havea similar or greater affinity for mutant Ras comparedto Raf-RBD [25].To directly compare the affinities of Raf-RBD and

RasIn2 to mutant Ras, we used an ELISA-basedactivity assay (Fig. 8). In this assay, biotin-labeledH-Ras(G12V) was immobilized on a streptavidin-coated ELISA plate and then incubated with eitherFLAG-tagged RasIn2 or FLAG-tagged Raf-RBD.After the detection of the FLAG tag with anti-FLAGantibody, followed by a secondary anti-mouse anti-body conjugated to horseradish peroxidase (HRP),we observed that RasIn2 gave ~10-fold highersignal as compared to RBD. These data indicatethat RasIn2 has a higher affinity to activeH-Ras(G12V)than Raf-RBD.

Fig. 6. Selection of RasIn2. (a) Enrichment of Ras-binding sequences through affinity maturation. Pool 5 shows highlevels of binding to active H-Ras(G12V). (b) In vitro pull-down efficiency of high abundance sequences from pool 5 incomparison with RasIn1. (c) Sequence comparison of high abundance sequences from pool 5.

567Intrabodies of Activated Ras Proteins

Page 7: RasIns: Genetically Encoded Intrabodies of Activated Ras ......RasIns: Genetically Encoded Intrabodies of Activated Ras Proteins Mehmet Cetin1, William E. Evenson2, Garrett G. Gross1,

Lastly, we tested RasIn2 in the cell-based coloca-lization assay against H-Ras(G12V) (Fig. 9) todemonstrate its specific binding inside the cells.Similar to RasIn1, RasIn2 colocalizes well withH-Ras(G12V), arguing that the protein is stableand functional in mammalian cells and recognizesactivated Ras in that context. However, unlikeRasIn1, RasIn2 also colocalized with wild-typeH-Ras inside the cells (Fig. 9d–f). This colocalizationcould be due to competition between RasIn2 andRasGAP (due to the high affinity of RasIn2), thuspreserving or trapping substantial amounts of H-Rasin the GTP-bound state. Alternatively, some coloca-lization could be from the binding of RasIn2 to theGDP-bound state of H-Ras, possibly due to the highexpression levels of transfected RasIn2 and H-Raspresent in the cell. We note that colocalization withactive Ras can be observed with RasIn1 insidethe cells even though the KD is ~2.1 uM (Fig. 4), andin vitro RasIn2 has similar affinity for the GDP form

of Ras as RasIn1 has for active H-Ras(G12V)-GTP(Fig. 6b).

Discussion

The goal of this work was to develop reagentsaimed at detecting and modulating activated Rassignaling. The rationale for this effort is that the chronicsignaling of K- and H-Ras, by mutation or upstreamactivation, occurs in more than 50% of human tumors[1]. The result of our work here is the development oftwo new intrabodies, RasIn1 and RasIn2, which bindto mutant and wild-type K- and H-RAS in a GTP-sta-te-dependent fashion. Furthermore, both RasIn1 andRasIn2 show selectivity for G12V mutants.In order for these reagents to be considered useful,

they should have an array of properties, including (1)selectivity for the GTP-bound state of Ras with little orno binding to related homologs and (2) functionality in

Fig. 7. Characterization of RasIn2 binding. (a) RasIn2 shows binding to different versions of active Ras (wild-type K- orH-Ras, and G12V mutant K- or H-Ras), showing that the recognition of the active Ras state is robust. (b) RasIn2preferentially binds active (GTP) over inactive (GDP) forms of H-Ras(G12V) (p = 0.003) and preferentially bindsunblocked active H-Ras(G12V) over active H-Ras(G12V) blocked with the c-Raf-kinase RBD domain (Raf-RBD; p =0.002). (c) Binding specificity of RasIn2 against homologous members of the Ras superfamily. RasIn1 binds specifically toactive wild-type H-Ras, but less to homologous Ras family members Rap1B(G12V) (p = 0.0001) or Arf1 (p = 0.0002).Percentage of sequence identity to the H-Ras G domain is shown in parentheses. (d) Binding of RasIn2 to K-Ras isdisrupted by the Y32R mutation in the Switch I region (p = 0.002). (e) Dissociation constants of RasIn1 and RasIn2 forH-Ras(G12V)-GTP, determined by SPR (Figs. S3 and S4). Error bars indicate the standard deviation of the mean.

568 Intrabodies of Activated Ras Proteins

Page 8: RasIns: Genetically Encoded Intrabodies of Activated Ras ......RasIns: Genetically Encoded Intrabodies of Activated Ras Proteins Mehmet Cetin1, William E. Evenson2, Garrett G. Gross1,

Fig. 9. RasIn2 colocalizes with various Ras forms in COS-7 cells. COS cells were co-transfected with RasIn2-GFP andGTS-SA-H-Ras(G12V) (a–c), GTS-SA-wt H-Ras (d–f), or SA-GTS (no target; g–i). Cells were fixed and stained for EGFP(green) and streptavidin (red). Colocalization (yellow) is visible in the merged image. RasIn2 colocalizes with both active(GTP) and inactive (GDP) H-Ras. Images are representative of at least 15 samples. Scale bar represents 5 μm.

Fig. 8. ELISA assay indicates that RasIn2 has higher affinity for H-Ras(G12V)-GppNHp than Raf1-RBD. (a) Schematicof the ELISA assay. Biotinylated H-Ras(G12V) was captured on the ELISA plate via streptavidin–biotin link. FLAG-taggedRasIn2 or Raf-RBD was then incubated with the ELISA plate, followed by an anti-FLAG antibody, and then a secondaryantibody conjugated to horseradish peroxidase (HRP). The plate was then washed and incubated with tetramethylbenzi-dine (TMB). (b) The OD450 of the ELISA readout plotted over different concentrations of ligand (RasIn2 or Raf-RBD).RasIn2 shows ~10-fold higher specific signal level than Raf-RBD.

569Intrabodies of Activated Ras Proteins

Page 9: RasIns: Genetically Encoded Intrabodies of Activated Ras ......RasIns: Genetically Encoded Intrabodies of Activated Ras Proteins Mehmet Cetin1, William E. Evenson2, Garrett G. Gross1,

the cytosolic environment. Unlike previously generat-ed ligands that bind to Ras, both RasIn1 and RasIn2satisfy these criteria. RasIn1 has a KD of 2.1 μM, areasonable outcome from a primary selection with 17random/randomized residues. It is notable that thisaffinity is sufficient to provide very good colocalizationwith the intended target in the COS cells, even thoughthis affinity is less than the downstream partnerRaf-RBD for the active state of Ras (KD = 80 nM).RasIn1 also has excellent nucleotide, mutant, andhomolog selectivity, showing little or no binding toRas-GDP (Fig. 2a) or Rap1B-GTP (Fig. 3b). Thisresult is remarkable because Ras and Rap1B are57% sequence identical, and the Switch I se-quences of Ras (YPDTIED) and Rap1B (YDPTIED)differ by only 2 aa (bold). Mutation and competitionanalysis of the RasIn1 indicates that the bindingoverlaps the known Ras binding site for Raf-RBD,although the precise sequence interactions mustbe different—the recognition sequences overallshow basic character (R/K rich) and have nosequence homology [32].Affinity maturation of RasIn1 resulted in the high

affinity binderRasIn2,with aKDof 120nManda20-foldimprovement gained through sequence optimization ofthe recognition site. Interestingly, there are 8 con-served positions in the FG loop, even though the loopsequences are mutagenized at N65% per position,such that the 10-residue loop is expected to havebetween 3 and 4 conserved positions. RasIn2 givessubstantially improved pull-down with Ras comparedto RasIn1 (Fig. 6b), competes with Raf-RBD (Fig. 7b),shows broad recognit ion biochemically ofK-Ras(G12V)-GTP, wi ld- type K-Ras-GTP,H-Ras(G12V)-GTP, and wild-type H-Ras-GTP (Fig.7a), outperforms RBD in an ELISA assay (Fig. 8), andalso shows excellent colocalization with H-Ras(G12V)and wild-type H-Ras inside COS cells (Fig. 9). The factthat RasIn1 and RasIn2 show similar intensity in theCOS cell assay is likely a result of the high transientexpression levels of both the binder (RasIn) and thetarget (Ras) in the COS cells.Overall, the two proteins demonstrate that it is

possible to use mRNA display to engineer individual,Ras-directed reagents with broad specificity for boththe mutant and active state of K- and H-Ras. Fur-thermore, the selectivity of these binders argues thatit should be possible to develop similar reagents forother G-proteins and study other G-protein-mediatedpathways.Left open for future work are the cellular effects

that can be probed using these binders. The mutantselectivity of RasIn1 and the high affinity of RasIn2imply that these proteins may be able to modulateor block downstream signaling via Raf kinaseactivation, a long-sought goal in cancer therapeutics.RasIn1 or RasIn2 could also be developed intoactive Ras biosensors for in vitro or in vivo studies,histology, or cancer diagnostics. Their small size,

high affinity, lack of disulfide bonds, state selectiv-ity, Ras specificity, and ability to be transfected intocells lend RasIn1 and RasIn2 to a wide range ofpossible future uses that often restrict the applica-tion of antibodies or non-genetically encodedmolecules.

Materials and Methods

Protein expression and purification

pCDNA 3.1 vector (Invitrogen)-encoding Rasfamily genes were obtained from Missouri S&TcDNA Resource Center. K-Ras(G12V) and H-Ras(G12V) (1-166) with a 5′ avitag and His tag werecloned into pET16b or pET24a vector. Proteins wereexpressed in E. coli BL21 (DE3) containing plasmidpBirAcm (Avidity) that overexpresses the birA biotinligase for in vivo biotinylation. Cells were grown at30 °C for 4–5 h after induction with 1 mM IPTGat an OD of 0.4, and cells were pelleted and frozenat −80 °C. Cells were lysed with French Pressor BPER (Thermo Scientific), and the lysate wascleared by centrifugation and applied to a Ni(II)-NTAcolumn (Qiagen) pre-equilibrated with binding buffer[50 mM Tris–HCl or Hepes (pH 7.5), 100mM NaCl,0.05% (vol/vol) Tween-20, 1 mMMgCl2, 10 μMGDP,1 mM DTT, and 20 mM imidazole]. The columnwas washed with wash buffer (binding buffer with500 mM NaCl and 40 mM imidazole), and proteinswere eluted with a linear gradient of wash buffer andelution buffer (binding buffer with 20 mM NaCl and400 mM imidazole). The molecular weight and purityof the protein were confirmed by denaturing SDS-PAGE. Purified protein was buffer exchanged intoselection buffer [50 mM Tris–HCl (pH 7.5), 150 mMNaCl, 0.05% (vol/vol) Tween-20, 5 mM MgCl2, and1 mM DTT] using Amicon Ultra spin columns, sup-plemented with 2 mMGDP and 10% (vol/vol) glycerol,and then frozen in liquid nitrogen. The activity ofRas proteins was confirmed by an in vitro pull-downassay with Raf-RBDDNA-encoding c-Raf-RBD (c-Raf residues 51–

131) was obtained from Integrated DNA Technolo-gies. c-Raf-RBD, RasIn1, and RasIn2 were clonedinto the pAO9 vector, which fuses each protein toa C-terminal maltose binding protein [15]. Proteinswere expressed in E. coli BL21 (DE3) and purified ona Ni(II)-NTA column as described above. RasIn1and RasIn2 were subjected to a secondary purifica-tion step on an amylose column (New EnglandBiolabs). Proteins were stored at −80 °C in storagebuffer [50 mM Tris–HCl (pH 7.5), 150 mM NaCl,0.05% (vol/vol) Tween-20, and 1 mM DTT].Rap1B(G12V) (1-166), RhoA(1-181), and Arf1

(18-179) were cloned in pDW363 and expressed inE. coli BL21 (DE3) pBirAcm for 5 h after induction

570 Intrabodies of Activated Ras Proteins

Page 10: RasIns: Genetically Encoded Intrabodies of Activated Ras ......RasIns: Genetically Encoded Intrabodies of Activated Ras Proteins Mehmet Cetin1, William E. Evenson2, Garrett G. Gross1,

with 0.5 mM IPTG [33]. Cells were pelleted andfrozen at −80 °C. Pellets were lysed with BPER,lysate was cleared by centrifugation, and the bio-tinylated proteins immobilized on neutravidin agarosebeads (Thermo Scientific). Rap1B(G12V) functional-ity was validated with a radioactive pull-down of itscanonical binding partner, Ral-GDS (Fig. S1).K-Ras(G12V, Y32R) mutants were generated by

PCR-based site-directedmutagenesis using pDW363K-Ras(G12V) as template. Proteins were expressedand purified as described for Ras family memberproteins.

Nucleotide exchange

For data in Figs. 1, 5, and 6, Ras proteins wereimmobilized on neutravidin agarose (Thermo Fisher)or Dynabeads® M-280 streptavidin magnetic beads(Life Technologies) and washed three times withexchange buffer [50 mM Tris–HCl (pH 7.5), 5 mMEDTA, 150 mM NaCl, and 1 mM nucleotide]. Beadswere incubated at 30 °C for 30 min, transferred onice, and washed three times with ice-cold selectionbuffer to stop exchange.For the binding assays in Figs. 2, 3, and 7,

nucleotide exchange was facilitated by incubatingthe Ras beads in selection buffer (20 mM Hepes,200 mMNaCl, 200μMnucleotide, and0.05%TWEEN)for 3 h.

Library preparation

Libraries were built as described [16], except thatthe Arg-24 position in e10FnIII was randomizedto create a random region totaling eight residues inthe BC loop. Oligos encoding the random regionswere synthesized by the Yale Keck OligonucleotideSynthesis facility.

mRNA display selection

RasIn1. The selection protocol was carried out asdescribed by Xiao et al. [22]. The FG loop contained10 random residues (X10; where X = all 20 naturalamino acids, encoded by the NNS codon), andthe BC loop of the library was a doped sequenceof the CDRH1 loop of iDab#6, an antibody with state-specific affinity for Ras, with approximately 40%frequency at each residue for the initial selection[26]. For the first round, the library was translatedin a total volume of 2.5 mL, purified, and reversetranscribed. K-Ras(G12V) was immobilized onneutravidin agarose beads and exchanged withGTPγS as described above. The library was thenadded to immobilized K-Ras(G12V)GTPγS in selec-tion buffer [50 mM Tris–HCl (pH 7.5), 150 mM NaCl,0.05% (vol/vol) Tween-20, 5 mM MgCl2, 1 mM DTT,and 10 μM GTPγS] at 4 °C. Following the first round,a FLAG purification step was included after transla-

tion to purify mRNA–fibronectin fusions away fromunfused mRNA, and the target binding step wasperformed at 25 °C. At round 3, streptavidin ultralinkbeads (Thermo Fisher) were used to prevent theenrichment of neurtravidin agarose binding se-quences. At round 5, a 15-fold molar excess ofK-Ras(G12V)-GDP was added to the selectionbuffer to enhance selective binding for active Ras.

Affinity maturation. A new library was synthesizedcontaining BC and FG loops that were doped onthe RasIn1 sequence (BC = GPVFSAYS and FG =FRWPMPRLVR) for the affinity maturation selection.The frequency of wild-type RasIn1 in the startingpool was estimated to be approximately 1.8 in 108

sequences. For the first round of selection, 200 μLof library was translated. H-Ras(G12V) was immo-bilized on 40 μL of Dynabeads® M-280 streptavidinmagnetic beads and was exchanged with GppNHp.Immobilized H-Ras(G12V) and purified library wereincubated as above. Binding for the first round wasperformed at 4 °C, rounds 2 and 3 were performed at25 °C, and rounds 4 and 5 were performed at 37 °Cto increase binding stringency.

Radioactive in vitro pull-down assays. Ras proteinswere immobilized and exchanged with nucleotideas described above. Individual ligands or selectionpools were radiolabeled by translation in vitro inrabbit reticulocyte lysate in the presence of [35S]Met. After oligo dT or FLAG purification, radiolabeledligands were added to Ras beads, incubated forthe indicated duration, and washed three times withselection buffer. The percentage of total radioactivecounts remaining on the beads was determined byscintillation counting.

Cell-based colocalization assay andimmunocytochemistry

The cell-based colocalization assay and immunocy-tochemistry were done as previously described [21].Briefly, COS-7 cells (ATCC) were grown toa confluency of ~40–60% on poly-D-lysine-coated 22× 22 mm glass coverslips in Dulbecco's modifiedeagle's medium (ATCC) supplemented with 10% fetalbovine serum in 5%CO2. pGWmammalian expressionp lasmids assembled wi th Golg i - ta rgetedRas-Streptavidin or 10FnIII-EGFP fusion genesinserted into the MCS were then co-transfected intocells using Effectene transfection reagent with trans-fection rates of around 10–30% (Qiagen). Following 24h of plasmid expression, COS cells were washed oncewith Phosphate Buffered Saline (PBS) [137 mM NaCl,2.7 mM KCl, 10 mM Na2HPO4, 1.8 mM KH2PO4,pH = 7.4], fixed with 4% paraformaldehyde for 5 min,washed three times with PBS, blocked for 30 min inblocking buffer [1% bovine serum albumin, 5% normal

571Intrabodies of Activated Ras Proteins

Page 11: RasIns: Genetically Encoded Intrabodies of Activated Ras ......RasIns: Genetically Encoded Intrabodies of Activated Ras Proteins Mehmet Cetin1, William E. Evenson2, Garrett G. Gross1,

goat serum, 0.1% (vol/vol) Triton X-100 in PBS], andstained for 1 h with chicken anti-GFP antibody (Aves),diluted 1:1000 in blocking buffer. After incubation withprimary antibody, cells were washed three times withPBS and stained for 30 min with anti-chicken second-ary antibody conjugated toAlexaFluor 488 (Invitrogen),1:1000, and Biotin-Rhodamine conjugate (VWR),1:000, in blocking buffer. Cells were then washedthree timeswithPBSandmounted on 75× 25mmglassmicroscope slides in Fluoromount-G (Electron Micros-copy Sciences) for imaging. All steps were carried out atroom temperature. Images of cells were taken with a60× water objective at 1.0 zoom on an Olympus IX81invertedmicroscope equippedwith aGFP/mCherry filtercube (Chroma Technology), an X-cite exacte mercurylamp (Excelitas Technologies), an EM-CCD digitalcamera (Hamamatsu), and Metamorph software (Mo-lecularDevices). Cellswere scored for the colocalizationof Rhodamine and Alexa Fluor 488 fluorescence. Ineach sample, at least 15 cells were imaged, andcolocalization was observed either every time or never.

High-throughput sequencing

Selection pools were sequenced at the USCEpigenome Center and the USC Genome & Cytom-etry Core using MiSeq™ and HiSeq™ and Systems(Illumina). DNA was prepped and sequenced aspreviously described [34]. Data analysis was doneusing computer scripts developed in-house.

SPR measurements

Measurements were done in USC NanoBiophy-sics Core Facility on a Biacore T100 instrument(Biacore). H-Ras(G12V) was immobilized on strep-tavidin sensor chips at the indicated surface density.Ras protein was exchanged with GppNHp on a chipwhere indicated by injecting SPR exchange buffer[50 mM Tris–HCl (pH 7.5), 1 mM EDTA, and 0.5 mMGppNHp] at 35 μL/min for 10 min. A concentrationseries of maltose binding protein–intrabody fusionwas injected at a flow rate of 100 μL/min at 25 °C inSPR run buffer [50 mM Tris–HCl (pH 7.5), 150 mMNaCl, 0.01% (vol/vol) Tween-20, 5 mM MgCl2, and10 μM GppNHp]. Data were analyzed by BiacoreT100 Evaluation Software.

ELISA assay

The polystyrene platewas incubatedwith 50 μLof 30nM streptavidin in 1X PBS per well overnight at 4 °C.The plate was washed with 1X Tris Buffered Saline(TBS) [50 mMTris, 150 mMNaCl, pH = 7.5]with 0.1%(vol/vol) Tween-20, filled completely with 5% BSA (wt/vol) in 1X PBS, incubated for 3 h, and washed.Biotin-labeled H-Ras(G12V)-GppNHp was diluted insample buffer [1X TBS, 5mMMgCl2, 1mMDTT, 20 μMGppNHp, 0.1% (vol/vol) Tween-20, and 1 mg/mL

BSA]. We added 100 uL of 30 nM biotin-labeledH-Ras(G12V)-GppNHp to each well, and it wasincubated for 90 min. After the incubation, 100 μL ofexchange buffer (50 mM Tris, 5mM EDTA, 1mM DTT,600 μMGppNHp, and 1 μM biotin) was added to eachwell and incubated for 30 min, and the plate was thenwashed in wash buffer [1X TBS, 5 mM MgCl2, 50 μMGTP, and 0.1% (vol/vol) Tween-20]. RasIn2 proteinand RBD were diluted serially in sample buffer andadded to the plate. The plate was incubated for 1 h andwashed. We added 100 μL of 20 nM Anti-FLAGantibody (Sigma) to the plate. The plate was washedand incubated for 1 hwith 100μL/well of 1:1000 dilutionof HRP-conjugated anti-mouse antibody. The platewas washed and incubated with tetramethylbenzidinesubstrate for approximately 5 min. The reaction wasstopped with equal volume of 2M sulfuric acid, and theOD450 was measured using a plate reader.

Acknowledgments

This work was supported by grants from the NIH(R01CA170820 and R01GM083898).This work ac-knowledges the instrumentation provided by theUSC Nanobiophysics Core Facility.Author contributions:M.C. performed selections

and in vitro pull-down assays and wrote the draft ofthe manuscript. W.E.E. performed in vitro pull-downassays and assembled the manuscript. G.G.G.performed the COS cell assays. F.J.-Y. performedthe ELISA assay. D.K. performed in vitro pull-downassays. T.T.T. advised M.C. and assisted with theBiacore. D.A. advised G.G.G. R.W.R. advised M.C.,W.E.E., F.J.-Y., and D.K.Competing financial interests: The authors

declare no competing financial interests.

Appendix A. Supplementary Data

Supplementary data to this article can be foundonline at doi:10.1016/j.jmb.2016.11.008.

Received 2 June 2016;

Received in revised form 11 November 2016;

Accepted 12 November 2016

Available online 16 November 2016

Keywords:

mRNA display;

E10FnIII;

fibronectin;

Ras;

intrabody

Abbreviations used:

Raf-RBD, Raf-kinase Ras Binding Domain; SPR, surface

plasmon resonance; HRP, horseradish peroxidase; PBS,

572 Intrabodies of Activated Ras Proteins

Page 12: RasIns: Genetically Encoded Intrabodies of Activated Ras ......RasIns: Genetically Encoded Intrabodies of Activated Ras Proteins Mehmet Cetin1, William E. Evenson2, Garrett G. Gross1,

Phosphate Buffered Saline; TBS, Tris Buffered Saline;

COS cells, a cell line that is CV-1 in Origin, and carrys

SV40 genetic material.

References

[1] J. Downward, Targeting RAS signalling pathways in cancer

therapy, Nat. Rev. Cancer 3 (2003) 11–22.

[2] A.E. Karnoub, R.A. Weinberg, Ras oncogenes: split person-

alities, Nat. Rev. Mol. Cell Biol. 9 (2008) 517–531.

[3] Y. Pylayeva-Gupta, E. Grabocka, D. Bar-Sagi, RAS

oncogenes: weaving a tumorigenic web, Nat. Rev. Cancer

11 (2011) 761–774.

[4] A.G. Stephen, D. Esposito, R.K. Bagni, F. McCormick,

Dragging ras back in the ring, Cancer Cell 25 (2014) 272–281.

[5] E.P. Reddy, R.K. Reynolds, E. Santos, M. Barbacid, A point

mutation is responsible for the acquisition of transforming

properties by the T24 human bladder carcinoma oncogene,

Nature 300 (1982) 149–152.

[6] C.J. Tabin, S.M. Bradley, C.I. Bargmann, R.A. Weinberg,

A.G. Papageorge, E.M. Scolnick, R. Dhar, D.R. Lowy, E.H.

Chang, Mechanism of activation of a human oncogene,

Nature 300 (1982) 143–149.

[7] F. McCormick, KRAS as a therapeutic target, Clin. Cancer

Res. 21 (2015) 1797–1801.

[8] T. Tanaka, M.N. Lobato, T.H. Rabbitts, Single domain

intracellular antibodies: a minimal fragment for direct in vivo

selection of antigen-specific intrabodies, J. Mol. Biol. 331

(2003) 1109–1120.

[9] J.M. Ostrem, U. Peters, M.L. Sos, J.A. Wells, K.M. Shokat,

K-Ras(G12C) inhibitors allosterically control GTP affinity and

effector interactions, Nature 503 (2013) 548–551.

[10] P.C. Gareiss, A.R. Schneekloth, M.J. Salcius, S.Y. Seo, C.M.

Crews, Identification and characterization of a peptidic ligand

for Ras, Chembiochem 11 (2010) 517–522.

[11] P. Upadhyaya, Z. Qian, N.G. Selner, S.R. Clippinger, Z. Wu,

R. Briesewitz, D. Pei, Inhibition of Ras signaling by blocking

Ras–effector interactions with cyclic peptides, Angew. Chem.

54 (2015) 7602–7606.

[12] W.W. Ja, O. Wiser, R.J. Austin, L.Y. Jan, R.W. Roberts,

Turning G proteins on and off using peptide ligands, ACS

Chem. Biol. 1 (2006) 570–574.

[13] W.W. Ja, R.W. Roberts, In vitro selection of state-specific

peptide modulators of G protein signaling using mRNA

display, Biochemistry 43 (2004) 9265–9275.

[14] R.J. Austin, W.W. Ja, R.W. Roberts, Evolution of class-

specific peptides targeting a hot spot of the G alpha s subunit,

J. Mol. Biol. 377 (2008) 1406–1418.

[15] C.A. Olson, H.-I. Liao, R. Sun, R.W. Roberts, mRNA display

selection of a high-affinity,modification-specific phospho-I kappa

B alpha-binding fibronectin, ACSChem. Biol. 3 (2008) 480–485.

[16] C.A. Olson, R.W. Roberts, Design, expression, and stability

of a diverse protein library based on the human fibronectin

type III domain, Protein Sci. 16 (2007) 476–484.

[17] A. Koide, C.W. Bailey, X. Huang, S. Koide, The fibronectin

type III domain as a scaffold for novel binding proteins, J. Mol.

Biol. 284 (1998) 1141–1151.

[18] A. Koide, S. Abbatiello, L. Rothgery, S. Koide, Probing protein

conformational changes in living cells by using designer

binding proteins: application to the estrogen receptor, Proc.

Natl. Acad. Sci. 99 (2002) 1253–1258.

[19] H.-I. Liao, C.A. Olson, S. Hwang, H. Deng, E. Wong, R.S.

Baric, R.W. Roberts, R. Sun, mRNA display design of

fibronectin-based intrabodies that detect and inhibit severe

acute respiratory syndrome coronavirus nucleocapsid pro-

tein, J. Biol. Chem. 284 (2009) 17,512–17,520.

[20] D. Lipovšek, Adnectins: engineered target-binding protein

therapeutics, Protein Eng. Des. Sel. 24 (2011) 3–9.

[21] G.G. Gross, J.A. Junge, R.J. Mora, H.-B. Kwon, C.A. Olson,

T.T. Takahashi, E.R. Liman, G.C.R. Ellis-Davies, A.W.

McGee, B.L. Sabatini, R.W. Roberts, D.B. Arnold, Recombi-

nant probes for visualizing endogenous synaptic proteins in

living neurons, Neuron 78 (2013) 971–985.

[22] L. Xiao, K.-C. Hung, T.T. Takahashi, K.-I. Joo, M. Lim,

R.W. Roberts, P. Wang, Antibody-mimetic ligand selected

by mRNA display targets DC-SIGN for dendritic cell-

directed antigen delivery, ACS Chem. Biol. 8 (2013)

967–977.

[23] H.K. Binz, P. Amstutz, A. Plückthun, Engineering novel

binding proteins from nonimmunoglobulin domains, Nat.

Biotechnol. 23 (2005) 1257–1268.

[24] C.A. Olson, J.D. Adams, T.T. Takahashi, H. Qi, S.M. Howell,

T.T. Wu, R.W. Roberts, R. Sun, H.T. Soh, Rapid mRNA-

display selection of an IL-6 inhibitor using continuous-flow

magnetic separation, Angew. Chem. Int. Ed. 50 (2011)

8295–8298.

[25] C. Herrmann, G.A. Martin, A. Wittinghofer, Quantitative

analysis of the complex between p21 and the ras-binding

domain of the human raf-1 protein kinase, J. Biol. Chem. 270

(1995) 2901–2905.

[26] T. Tanaka, R.L. Williams, T.H. Rabbitts, Tumour prevention

by a single antibody domain targeting the interaction of signal

transduction proteins with RAS, EMBO J. 26 (2007)

3250–3259.

[27] D. Filchtinski, O. Sharabi, A. Rüppel, I.R. Vetter, C.

Herrmann, J.M. Shifman, What makes Ras an efficient

molecular switch: a computational, biophysical, and struc-

tural study of Ras–GDP interactions with mutants of Raf,

J. Mol. Biol. 399 (2010) 422–435.

[28] J. Colicelli, Human RAS superfamily proteins and related

GTPases, Sci. STKE 2004 (2004) RE13.

[29] S.R. Sprang, G protein mechanisms: insights from structural

analysis, Annu. Rev. Biochem. 66 (1997) 639–678.

[30] P. Gideon, J. John, M. Frech, A. Lautwein, R. Clark, J.E.

Scheffler, A. Wittinghofer, Mutational and kinetic analyses

of the GTPase-activating protein (GAP)-p21 interaction: the

C-terminal domain of GAP is not sufficient for full activity, Mol.

Cell. Biol. 12 (1992) 2050–2056.

[31] S. Wohlgemuth, C. Kiel, A. Krämer, L. Serrano, F.

Wittinghofer, C. Herrmann, Recognizing and defining true

Ras binding domains I: biochemical analysis, J. Mol. Biol.

348 (2005) 741–758.

[32] G.J. Clark, J.K. Drugan, R.S. Terrell, C. Bradham, C.J. Der,

R.M. Bell, S. Campbell, Peptides containing a consensus

Ras binding sequence from Raf-1 and theGTPase activating

protein NF1 inhibit Ras function, Proc. Natl. Acad. Sci. 93

(1996) 1577–1581.

[33] S. Duffy, K.-L. Tsao, D.S. Waugh, Site-specific, enzymatic

biotinylation of recombinant proteins in Spodoptera frugiperda

cells using biotin acceptor peptides, Anal. Biochem. 262 (1998)

122–128.

[34] C.A. Olson, J. Nie, J. Diep, I. Al-Shyoukh, T.T. Takahashi, L.Q.

Al-Mawsawi, J.M. Bolin, A.L. Elwell, S. Swanson, R. Stewart,

J.A. Thomson, H.T. Soh, R.W. Roberts, R. Sun, Single-round,

multiplexed antibody mimetic design through mRNA display,

Angew. Chem. Int. Ed. 51 (2012) 12,449–12,453.

573Intrabodies of Activated Ras Proteins