+ All Categories
Home > Documents > From Ras to Rap and Back, a Journey of 35 Years

From Ras to Rap and Back, a Journey of 35 Years

Date post: 01-Oct-2021
Category:
Upload: others
View: 8 times
Download: 0 times
Share this document with a friend
13
From Ras to Rap and Back, a Journey of 35 Years Johannes L. Bos Molecular Cancer Research, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands Correspondence: [email protected] Our laboratory has studied Ras and Ras-like proteins since the discovery of the Ras oncogene 35 years ago. In this review, I will give an account of what we have done in these 35 years and indicate the main papers that have guided our research. Our efforts started with the early analysis of mutant Ras in human tumors followed by deciphering of the role of Ras in signal transduction pathways. In an attempt to interfere in Ras signaling we turned to Rap proteins. These proteins are the closest relatives of Ras and were initially identified as Ras antagonists. However, our studies revealed that the Rap signaling network primarily is involved in spa- tiotemporal control of cell adhesion, in part through regulation of the actin cytoskeleton. More recently we returned to Ras, trying to interfere in Ras signaling by combinatorial drug testing using the organoid technology. THE BEGINNING I n 1982, the stunning result that a single-point mutation converted a normal H-Ras gene into an oncogene was published by the Weinberg laboratory (Parada et al. 1982). The experimen- tal setup was based on the assumption that cancer is caused by mutations in genes and that these “oncogenes,” when transfected into normal cells, should be able to convert these cells in tumor cells. For this assay, Weinberg and colleagues transferred genomic DNA from a human kidney tumor cell line into NIH-3T3 mouse fibroblasts. This resulted in morpholog- ical transformation of NIH-3T3 cells, suggest- ing the presence of an oncogene in the genomic DNA of these tumor cells. However, the identi- fication of such an oncogene was a painstaking process that requested multiple rounds of selec- tion, to reduce the amount of human DNA present in the transformed mouse cell. The next step was the separation of the human DNA from the mouse DNA based on human- specific repetitive sequences, followed by clon- ing and sequence analysis. Many groups fol- lowed a similar path, resulting in the discovery of the H-ras-related genes K-ras and N-ras (Hall et al. 1983; Shimizu et al. 1983). Many tumor cell lines and tumor tissues were subsequently screened for oncogenes. But the assay remained extremely labor-intensive and the success rate was low. At that time, I was working in the lab- oratory of Alex van der Eb on cell transforma- tion by the adenovirus E1 region. To switch to human oncogenes would have been an obvious choice, but jumping on a very labor-intensive bandwagon was not very attractive. I was, how- ever, struck by a paper of Bruce Wallace showing a method to identify specific mutations in sickle cell anemia using synthetic oligonucleotides Editors: Linda VanAelst, Julian Downward, and Frank McCormick Additional Perspectives on Ras and Cancer in the 21st Centuryavailable at www.perspectivesinmedicine.org Copyright # 2018 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a031468 Cite this article as Cold Spring Harb Perspect Med 2018;8:a031468 1 www.perspectivesinmedicine.org Press on September 30, 2021 - Published by Cold Spring Harbor Laboratory http://perspectivesinmedicine.cshlp.org/ Downloaded from
Transcript
Page 1: From Ras to Rap and Back, a Journey of 35 Years

From Ras to Rap and Back, a Journey of 35 Years

Johannes L. Bos

Molecular Cancer Research, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht,The Netherlands

Correspondence: [email protected]

Our laboratory has studied Ras and Ras-like proteins since the discovery of the Ras oncogene35 years ago. In this review, I will give an account of what we have done in these 35 years andindicate the main papers that have guided our research. Our efforts started with the earlyanalysis of mutant Ras in human tumors followed by deciphering of the role of Ras in signaltransduction pathways. In an attempt to interfere in Ras signaling we turned to Rap proteins.These proteins are the closest relatives of Ras and were initially identified as Ras antagonists.However, our studies revealed that the Rap signaling network primarily is involved in spa-tiotemporal control of cell adhesion, in part through regulation of the actin cytoskeleton.More recently we returned to Ras, trying to interfere in Ras signaling by combinatorial drugtesting using the organoid technology.

THE BEGINNING

In 1982, the stunning result that a single-pointmutation converted a normal H-Ras gene into

an oncogene was published by the Weinberglaboratory (Parada et al. 1982). The experimen-tal setup was based on the assumption thatcancer is caused by mutations in genes andthat these “oncogenes,” when transfected intonormal cells, should be able to convert thesecells in tumor cells. For this assay, Weinbergand colleagues transferred genomic DNA froma human kidney tumor cell line into NIH-3T3mouse fibroblasts. This resulted in morpholog-ical transformation of NIH-3T3 cells, suggest-ing the presence of an oncogene in the genomicDNA of these tumor cells. However, the identi-fication of such an oncogene was a painstakingprocess that requested multiple rounds of selec-tion, to reduce the amount of human DNA

present in the transformed mouse cell. Thenext step was the separation of the humanDNA from the mouse DNA based on human-specific repetitive sequences, followed by clon-ing and sequence analysis. Many groups fol-lowed a similar path, resulting in the discoveryof the H-ras-related genes K-ras and N-ras (Hallet al. 1983; Shimizu et al. 1983). Many tumorcell lines and tumor tissues were subsequentlyscreened for oncogenes. But the assay remainedextremely labor-intensive and the success ratewas low. At that time, I was working in the lab-oratory of Alex van der Eb on cell transforma-tion by the adenovirus E1 region. To switch tohuman oncogenes would have been an obviouschoice, but jumping on a very labor-intensivebandwagon was not very attractive. I was, how-ever, struck by a paper of Bruce Wallace showinga method to identify specific mutations in sicklecell anemia using synthetic oligonucleotides

Editors: Linda VanAelst, Julian Downward, and Frank McCormick

Additional Perspectives on Ras and Cancer in the 21st Century available at www.perspectivesinmedicine.org

Copyright # 2018 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a031468

Cite this article as Cold Spring Harb Perspect Med 2018;8:a031468

1

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on September 30, 2021 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 2: From Ras to Rap and Back, a Journey of 35 Years

(Conner et al. 1983). The basic principle is thatthe thermal stability of a 20-mer oligonucleo-tide annealed to genomic DNA is reduced by acouple of degrees when one base pair is mis-matched. As mutations were only found in co-don 12 and 61 in H-, K-, and N-Ras, we neededa limited number of oligonucleotides represent-ing all possible single base pair changes. At thattime oligonucleotide synthesis was possible inonly a few laboratories, one of which was thelaboratory of Jacques van Boom next door.With some help from my thesis supervisorPiet Borst, I convinced him to make the re-quired 72 different oligonucleotides, coveringall possible codon 12 and 61 mutations. Detect-ing mutant Ras with this approach turned outto work quite well for cell lines and for reallypure tumor samples, but it also had its limits ofdetection (Bos et al. 1984). The discovery of thepolymerase chain reaction (PCR) method(Saiki et al. 1985) resolved this limitation andwe were able to identify Ras mutations in a largevariety of tumors by simple dot-blot assays(Verlaan-de Vries et al. 1986). As a PCR ma-chine, we used a robot arm moving betweenwater baths of different temperatures as our firstPCR machine, with homemade Klenow poly-merase to be added after each cycle. One ofthe highlights was the colon study with BertVogelstein showing K-ras mutations to occurin 50% of the colon tumors (Bos et al. 1987),an observation independently made by ManualPerucho and coworkers (Forrester et al. 1987).Interestingly, Ras mutations were also present in50% of benign adenomas. Together with othergenetic alterations detected by Vogelstein, thisled to the postulation of the sequential activa-tion of oncogenes in colorectal cancer (Vogel-stein et al. 1988). Another highlight was thestudy of lung adenocarcinoma together withSjoerd Rodenhuis, revealing 30% K-ras muta-tions (Rodenhuis et al. 1987). However, theclinical relevance/diagnostic value of Ras mu-tations became only apparent with the develop-ment of epidermal growth factor receptor(EGFR) inhibitors like cetuximab and erlotinib.The presence of a Ras mutation is currently anexclusion criterion for these drugs (Eberhardet al. 2005; Karapetis et al. 2008). Ras mutations

were most frequently found in pancreatic can-cer, as first described by Perucho and coworkers(Almoguera et al. 1988).

In these early studies, we already observedgenetic heterogeneity in tumors. For instance,in melanoma we detected two different Ras mu-tations in one primary tumor that separated inthe metastatic lesions (van ’t Veer et al. 1989). Inseminomas, a morphologically very homoge-neous tumor, Ras mutations were found insome parts of the primary tumor, but not inother parts (Mulder et al. 1989). In acutemyeloid leukemias (AMLs), 30% of which carryN-Ras mutations, we observed that in somecases the Ras mutation remained detectable af-ter complete remission. Interestingly, in thesecases, Ras mutations were also present in theapparently unaffected lymphoid lineage, sug-gesting that the Ras mutation is present in anearly hematopoietic stem cell not affected by thetreatment. In other cases of AML, the Ras mu-tation disappeared after treatment, and did notrecur after relapse. We concluded from thesestudies, first, that Ras mutations can contributeto the oncogenic process at different stages,second, that mutant Ras apparently has noeffect on the lymphoid lineage, and third, thattreatment kills all the aggressive blast cells of theleukemia, but not the underlying premalignantcell clone (Yunis et al. 1989). These and otherearly studies have been summarized in Bos(1989).

Ras REGULATION

After identification of mutated Ras as onco-gene, much effort was put in understandingthe function of Ras, a small GTPase thatnormally cycles between an active GTP-boundand an inactive GDP-bound state, but whenoncogenically mutated, is stalled in the activeGTP-bound state. The obvious rationale beingthat understanding the normal function of Raswould provide insight into how to treat tumorsharboring oncogenic Ras. The kinetics of Rascycling between a GTP- and a GDP-bound statein vitro indicated the existence of accessoryproteins to regulate Ras loading. The first regu-latory protein identified was the CDC25 pro-

J.L. Bos

2 Cite this article as Cold Spring Harb Perspect Med 2018;8:a031468

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on September 30, 2021 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 3: From Ras to Rap and Back, a Journey of 35 Years

tein, a guanine nucleotide exchange protein forRas in yeast (Broek et al. 1987; Robinson et al.1987). Soon after, Frank McCormick discovereda p120 GTPase-activating protein (p120GAP)guided by the observation that the in vivoGTPase activity of Ras was much higher thanthe in vitro activity (Trahey and McCormick1987; Wong et al. 1992).

At that time, GDP/GTP cycling of Ras wasstudied in vitro and a method measuring Rasactivation in vivo was initially lacking. JulianDownward developed a method based on label-ing of cells with radioactive orthophosphatefollowed by immunoprecipitation of Ras pro-teins. GDP and GTP were eluted and separatedby thin layer chromatography. Using this proce-dure, it was first found that T-cell receptor en-gagement was a strong inducer of Ras activity(Downward et al. 1990). We followed by show-ing that insulin could activate Ras, connectingRas to receptor tyrosine kinases (Burgering et al.1991). Interestingly, many other extracellularstimuli were shown to activate Ras, includingstimuli that use G-protein-coupled receptors(van Corven et al. 1993). This showed that Rasactivation was not part of a signaling pathwayinduced by one specific ligand, but contributesto signaling induced by many factors control-ling cellular responses. Subsequent geneticstudies by others revealed several mammaliancounterparts to the yeast CDC25 protein thatmediate Ras activation, such as the Sos proteins,all having a characteristic CDC25 homology do-main as catalytic domain. The various Ras gua-nine nucleotide exchange factors (GEFs) andGTPase activating proteins (GAPs) and theirmode of action have been reviewed extensively(Bos et al. 2007).

Ras EFFECTORS

In the early 1990s, we learned from Chris Mar-shall that extracellular regulated kinase (ERK),also known as microtubule-associated proteinkinase (MAPK), was activated in cells transfect-ed with mutant Ras (Leevers and Marshall1992). At that time, we were developing a systempioneered by Marino Zerial to use vaccinia virusto introduce dominant negative Ras (RasN17)

into cells. Indeed, in cells expressing RasN17insulin-induced ERK activation was completelyblocked: This made ERK the first “proven”downstream biochemical effect of Ras inmammalian cells (de Vries-Smits et al. 1992).However, Ras and ERK did not directly interactand subsequently Raf was found to be the effec-tor of Ras that, through MAPK/ERK kinase(MEK), activates ERK (Moodie et al. 1993).

Soon hereafter, multiple Ras effectorswere identified, including phosphatidylinositol3-kinase (PI3K) (Kodaki et al. 1994) and Ralguanine nucleotide dissociation stimulator(RalGDS) (Hofer et al. 1994). The discoveryby Boudewijn Burgering and Paul Coffer inour laboratory that the product of PI3K,phosphatidylinositol 3,4,5, triphosphate, acti-vates protein kinase B (Burgering and Coffer1995) opened a completely new line of research,resulting in the identification of FoxO tran-scription factors as main targets for proteinkinase B–mediated phosphorylation (Kopset al. 1999).

The importance of multiple effectors forRas function was beautifully shown by MichaelWhite using effector domain mutants that se-lectively bind to the various effectors: whentransfected separately they had hardly anytransforming activity, but in combination theydid (White et al. 1995).

In the years thereafter, many aspects ofthe signaling networks in which Ras plays acentral role and the role of Ras in cancer wereelucidated (Cox et al. 2014; Papke and Der2017). The most updated version of the Rassignaling network can be found at the NationalCancer Institute (www.cancer.gov/research/key-initiatives/ras/ras-central/blog/2017/mccormick-ras-pathway-v3).

HOW TO INTERFERE IN MUTANT RasSIGNALING

Knowledge on the regulation of Ras rapidly ac-cumulated but did not yet result in treatmentoptions for cancer patients. Initially, it wasthought that Ras action could be inhibited bydisplacing the GTP moiety by a small molecule,but this idea was rapidly abandoned when it be-

From Ras to Rap and Back, A Journey of 35 Years

Cite this article as Cold Spring Harb Perspect Med 2018;8:a031468 3

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on September 30, 2021 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 4: From Ras to Rap and Back, a Journey of 35 Years

came clear that the affinityof GTP (and GDP)forRas is within the picomolar range. Instead, mostattention was focused on interfering in the lipidmodification of Ras, particularly the carboxy-terminal farnesylation and later geranyl-gerany-lation. When it became clear that Ras belongsto a superfamily of small GTPases with over ahundred members almost all similarly modified,the enthusiasm for this approach diminished.These studies have recently been discussed byCox and coworkers (Cox et al. 2014). Nowadaysmost attention is focused on interfering in sig-naling downstream of Ras (see below).

Rap PROTEINS

Rap proteins were first identified by VeroniquePizon and Pierre Chardin and, based on theirsimilarity to Ras, the authors suggested thatthey may interact with the same effectors asRas (Pizon et al. 1988). Subsequently, Nodaand coworkers identified K-ras revertant 1 (K-rev1), whichwas identical to Rap1, in a screen forgenes that can revert morphological transfor-mation of mutant K-ras cells (Kitayama et al.1989). This ledto the hypothesisthatRap1 mightbe a decoy that traps Ras effectors in an inactivecomplex and could be a possible way to interferein mutant Ras tumors? We became interested inRap after our finding that in certain cell typescyclic adenosine monophosphate (cAMP) caninhibit growth-factor-induced (and Ras-medi-ated) activation of ERK (Burgering et al. 1991).Combined with the observation of DanielAltschuler that cAMP could activate Rap1(Altschuler et al. 1995), this suggested a role forRap1 in inhibiting Ras following cAMP stimu-lation of cells. However, despite the fact that invitro Rap1 can bind to the Ras-binding domainof Raf1, we were unable to show a direct involve-ment of Ras effector signaling (Zwartkruis et al.1998).

A function of Rap proteins distinct from Rasis further supported by the notion that Ras andRap were already present as separate proteins inthe hypothetical last eukaryotic common ances-tor (van Dam et al. 2011). Later in evolution atthe opisthokonta stage the Rap branch sepa-rated in Rap1 and Rap2 and at the vertebrata

stage in Rap1A and B and Rap2A, B, and C(Fig. 1). Research on Rap orthologs in manyspecies has provided pivotal knowledge abouthuman Rap proteins (Frische and Zwartkruis2010), but especially the analysis of the singleRap protein in budding yeast, Rsr1, has led to aframework for the regulation and function ofthese small GTPases. Budding always occurs ad-jacent to a scar of a previous bud. This bud siteselection is however abolished in cells mutatedfor Rsr1 (Bud1), for a GEF (Bud5) or for a GAP(Bud2) for Rsr1. It was found that these pro-teins form a complex that recognizes speciallandmark proteins in the rim of the bud scar.The signal is further propagated to the CDC42complex, which facilitates the local recruitmentof the actin cytoskeleton (Bi and Park 2012). Inaddition, in Drosophila, Rap proteins play a rolein cell migration and the proper localization ofadherence junctions (Asha et al. 1999; Knox andBrown 2002). This led to the hypothesis thatalso mammalian Rap proteins are involvedin spatial control of cell adhesion, most likelythrough actin-driven processes.

REGULATION OF Rap

Our initial studies were devoted to find stimulithat can activate Rap1 and we used platelets asthey abundantly express Rap1. To measure Rapactivity, we developed a nonradioactive pull-down assay, in which active GTP-bound Rap1is precipitated with a GST-coupled Ras-bindingdomain (Franke et al. 1997). Such pull-downassays have subsequently been developed forRas and many other GTPases and are now themost widely used assay to measure activity of avariety of small GTPases, including those fromthe Rho family. We found that all stimuli thatresulted in a rapid activation of platelets alsoactivated Rap1. As one of the hallmarks of plate-let activation is increased integrin-mediatedadhesion and spreading, we postulated thatRap1 may control integrin activation (Frankeet al. 1997). Subsequent studies revealed that alarge variety of stimuli are able to activate Rapproteins in all cell types tested.

The observation that, similar to Ras, Rap1 isactivated by many stimuli opened the quest for

J.L. Bos

4 Cite this article as Cold Spring Harb Perspect Med 2018;8:a031468

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on September 30, 2021 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 5: From Ras to Rap and Back, a Journey of 35 Years

guanine nucleotide exchange factors (GEFs)and GAPs that could regulate this activity. Thefirst Rap-specific GEF, C3G, was identified byMichiyuki Matsuda and coworkers (Gotoh et al.1995). Among others, it mediates signals fromreceptor tyrosine kinases through the adaptorprotein Crk. Currently at least nine Rap-selec-tive GEFs have been identified (Fig. 2), whichhave, similar to Ras GEFs, a CDC25 homologydomain. However, both in vivo and in vitrothere is a strong specificity for Rap, with Cal-DAGGEF3 (RasGrp3) as an exception as itacts both on Ras and Rap. In addition, almostall these proteins have protein–protein orprotein–lipid interaction domains, suggestingmultiple interaction partners. RapGAPs have adifferent structure and a different mechanism of

catalysis compared to RasGAPs; whereas Ras-GAPs provide a critical arginine (argininefinger) for hydrolysis of GTP, RapGAPs providean aspartate (aspartate thumb). Again, despitesimilarities between Ras and Rap, the GAPs arespecific for their cognate GTPase (Bos et al.2007). Exceptions are the GAP1(IP4BP) familymembers a Rap-like GAP, which upon mem-brane binding adopts activity toward Ras (Sotet al. 2013).

EXCHANGE PROTEIN DIRECTLY ACTIVATEDBY cAMP (Epac)

At one time, it was a dogma that all mammaliancAMP effects were mediated by protein kinaseA. However, we noted that inhibition of protein

Rap1ARap1BRap2ARap2BRap2CN-RasH-RasK-RasE-RasM-Ras

Ras1 and 2Ras

Rap

RSR1

Unikonta Metazoa

Myears

Mam

mal

ia

Verte

brat

a

Opi

stho

kont

a

LECA

~200

0

~100

0

~500

~200

Ral

REMRhebRGRRasD

Zygomycotal Ral

Rit

RhoRabRanSrβArfSar1

R-RasRaIARaIB

TC21

Figure 1. Evolutionary timeline of the Ras family, Ras, Rap, and Ral proteins. The last eukaryotic commonancestor (LECA) likely contained already representatives of these proteins. Later duplications are responsible forthe different members in mammalian cells. (From van Dam et al. 2011; adapted, with permission, from theauthors.)

From Ras to Rap and Back, A Journey of 35 Years

Cite this article as Cold Spring Harb Perspect Med 2018;8:a031468 5

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on September 30, 2021 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 6: From Ras to Rap and Back, a Journey of 35 Years

kinase A did not affect cAMP-induced Rap1activity. In a subsequent search in publicly avail-able databases of the human genome project, wefound cAMP-binding domains in close vicinityof CDC25 homology domains. Using purifiedprotein encoded by the corresponding comple-mentary DNA (cDNA), we could show a Rap-specific GEF with activity that was completelydependent on the presence of cAMP. Wecalled this protein exchange protein directly ac-tivated by cAMP (Epac) (de Rooij et al. 1998).Two human Epac proteins were identified: onewith a single cAMP-binding domain (Epac1)and one with two cAMP-binding domains(Epac2). Subsequent structural analysis, in col-laboration with Fred Wittinghofer, revealed adetailed mechanism on how Epac is regulated(Fig. 3A). In the absence of cAMP, Epac is in aclosed conformation with the cAMP-bindingdomain occluding the Rap-binding site. WhencAMP binds, Epac adopts an open conforma-tion, where Rap can bind and to become acti-vated (Rehmann et al. 2006, 2008). In addition,our studies for the first time revealed how in-genuously cAMP acts: the cyclic phosphategroup of cAMP releases a brake that allows amajor conformational change that is furtherstabilized by interactions with the adenosinegroup (Rehmann et al. 2003). Simultaneously,

cAMP also induces the translocation of Epac1(but not Epac2) from the cytosol to the plasmamembrane (Ponsioen et al. 2009). This isfacilitated by a cAMP-induced conformationalchange in the Dishevelled, EGL-10, and pleck-strin (DEP) domain (Li et al. 2011), resultingin an increased affinity for membrane-boundphosphatidic acid (Consonni et al. 2012).Several additional membrane anchors havebeen identified for Epac1, including activatedEzrin, showing that, dependent on the condi-tions, Epac1 can localize differently (Gloerichet al. 2010).

DEVELOPMENT OF 007

Further studies on the function of Epac1 werehampered by the fact that cAMP activates bothprotein kinase A (PKA) and Epac. However,Stein Døskeland pointed out to us a peculiarityin the cAMP-binding domain, where a highlyconserved glutamate that forms hydrogenbounds with the 20-hydroxyl of the ribose ofcAMP was absent in the cAMP-binding do-main of Epac1 and the high-affinity, secondbinding site of Epac2. This suggested that thesehydrogen bonds, and thus the 20OH of the ri-bose may be required specifically for cAMP tointeract with PKA, but not with Epac. Together

C3G

CNB-L CNB-L REM

REM CDC25-HD

CDC25-HD

CDC25-HD

CDC25-HD

CDC25-HD

CDC25-HD

CDC25-HD EF EF C1

C2 RA

GEFsGAPs

PLCCDC25-HD

CDC25-HD

PDZ

PDZREM

REM

REM

REM

REM

REM

REM

PDZ

PDZ

PH BTKGAP

GAPC2

C2C2

PH

GAP

GAP

GAPGoLoco

RA

RA

RA

RA

RA

CNB

CNB

CNB-LCNB-L

CNB

DEP

DEP

PDZ-GEFl

PDZ-GEF2

Epac1

Epac2

Repac

PLCε1

RasGEF1A,B,C

Rap1GAP1,2

Spa-1

SPAR1,2,3

SynGAP

GAP1IP4BP, RASAL, CAPRI

CalDAG-GEF1,3

Figure 2. Rap guanine nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs). Indicated arethe domain structures of Rap-specific GEFs and GAPs. (From Bos et al. 2007; adapted, with permission, fromthe authors.)

J.L. Bos

6 Cite this article as Cold Spring Harb Perspect Med 2018;8:a031468

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on September 30, 2021 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 7: From Ras to Rap and Back, a Journey of 35 Years

with Frank Schwede and Hans-GottfriedGenieser, we started to modify cAMP, resultingin a highly Epac-selective compound 8-CPT-20OMe-cAMP (Fig. 3B), which we called 007(Enserink et al. 2002). Using this compound,we demonstrated that cAMP through Epac isable to modify integrin-mediated cell-substra-tum adhesion and E-cadherin-mediated cell–cell adhesion (Rangarajan et al. 2003; Priceet al. 2004). Subsequently, many studies havebeen published using this compound therebyshowing the versatile role Epac proteins play incAMP signaling, including insulin secretion,endothelial barrier function, memory, andheart rhythm (for further details, see Gloerichand Bos 2010).

DOWNSTREAM TARGETS OF Rap1PROTEINS

007 did not induce activation of ERK, indicatingthat B-raf is not a downstream target of Rap1(Enserink et al. 2002). In this respect, a claimthat B-raf is a downstream target of Rap1 isincorrect. The first genuine effector for a Rapprotein identified was the Drosophila Canoeprotein (Boettner et al. 2003). This protein issimilar to the mammalian junctional proteinAF6 with ubiquitin-like Ras association (RA)domains to bind active Rap1. Subsequently,AF6 was found to be one of the effectors ofRap1 that mediate some, but not all, responsesof Rap1 activation. Indeed, of the many RA-

Inactive EpacA

B

cAMP

Rap

Active Epac • Rap complex

NH2

S

O

OCH3O

OO

O

O

P

CIN

NN

N

Figure 3. (A) Crystal structure of Epac 2 in the closed (left) and open (right) conformation. The gray area (right)represents the first cyclic adenosine monophosphate (cAMP) domain and Dishevelled, EGL-10, and pleckstrin(DEP) domain visualized by cryoelectron microscopy (Rehmann et al. 2006, 2008). (B) Structure of 8CPT-20OMe-cAMP (007).

From Ras to Rap and Back, A Journey of 35 Years

Cite this article as Cold Spring Harb Perspect Med 2018;8:a031468 7

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on September 30, 2021 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 8: From Ras to Rap and Back, a Journey of 35 Years

domain-containing proteins multiple have beenimplicated to transduce Rap1 signaling. Theseeffectors, together with the existence of multipleRap1-GEFs and GAPs, prompted us to extendthe hypothesis based on the yeast studies. Weproposed that a cell can have multiple indepen-dent Rap modules consisting of a Rap protein, aGEF, and probably a GAP, each recognizingdifferent landmarks (spatial cues) to regulatelocalized recruitment of the actin cytoskeletonin time and in space using a variety of effectorsystems. The presence of additional regulatoryand interaction domains, particularly in thevarious GEFs, provide ample opportunities forspecific landmark recognition and regulation.The consequence of this hypothesis was thatwe had to search for a specific Rap signalingmodule in a defined model system. Althoughwe explored Rap1-induced integrin-mediatedcell adhesion and Rap1-induced cell spreadingin epithelial cells, we particularly focused ourattention on endothelial barrier function. En-dothelial cells are highly responsive to 007 asmeasured by, for instance, an increased electri-cal resistance of the monolayer. This increasedbarrier function is accompanied by a shift that isobserved from radial stress fibers to circumfer-ential actin, which could explain the increasedtightness of endothelial cell–cell junctions.

Using small-interfering RNA (siRNA) screens,we found that Rap1 interacted with a complexof two related RA-domain-containing proteins,Rasip1 and Radil. Interestingly, both Radiland Rasip1 are homologous to Canoe, the firsteffector found for Rap1 in Drosophila. Impor-tantly, Radil directly interacts with ArhGAP29,a Rho-specific GAP, and we could show thatArhGAP29 mediates 007-induced decrease inradial stress fibers and increased barrier func-tion (Fig. 4, left) (Post et al. 2013, 2015). In-triguingly, multiple Rap1 modules may controlbarrier function simultaneously. For instance,Mochizuki and coworkers discovered that cir-cumferential actin formation is mediated by theCDC42-GEF FGD5 (Ando et al. 2013). HowFGD5 is activated is still unclear, but, in over-expression studies, we observe that FGD5 alsoforms a complex with Radil, suggesting that theRadil/Rasip complex regulates both pathways.Also, the Rap1 effector AF6 is involved in thisprocess (Birukova et al. 2013), but how thesedifferent effectors cooperate is still unclear.Surprisingly, Krit1, previously identified as aRap1 effector in a similar assay in Huvec cells(Glading et al. 2007), did not affect barrier func-tion in our experiments (Pannekoek et al. 2011).

Several other effectors for Rap1 have beenidentified, such as the RA-domain-containing

Brush borders

Mst4

TNIK

PDZ-GEF1/2

Rap2A

TNIK

Mst4

Ezrin

Api

cal m

embr

ane

PDZ-GEF1/2

PI4,5P2

A B

cAMP

Epac1

Rasip1

ArhGAP29Radil

Epac1

Rap1

Rasip1

RadilArhGAP29

Rho

Radial stress fibers

PLD

PA

PLD

Figure 4. Graphical summary of (A) Epac1-mediated regulation of radial stress fibers through the Radil-Rasip1-ArgGAP29 complex, and of (B) Rap2A-mediated intestinal brush borders formation (for explanation see text).

J.L. Bos

8 Cite this article as Cold Spring Harb Perspect Med 2018;8:a031468

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on September 30, 2021 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 9: From Ras to Rap and Back, a Journey of 35 Years

protein Riam, which plays an important role inintegrin-mediated cell adhesion (Lafuente et al.2004).

Rap2

The early deviation of Rap1 and Rap2 in evolu-tion suggests that they may have different func-tions and indeed, although Rap2 is regulated bythe same GEFs and GAPs as Rap1, it is clearlydifferent as Rap2(-GTP) binds to a differentset of effector proteins lacking an RA domain:the Traf2 and Nck-interacting kinases (TNIKs)(Taira et al. 2004). We stumbled on Rap2A inthe analysis of brush-border formation in W4intestinal epithelial cells. In these cells, brush-border formation is induced by doxycycline-in-duced activation of the LKB–Strd1a complex(Baas et al. 2004). When we tested the involve-ment of Rap proteins in this process, we foundthat siRNA mediated depletion of Rap2A, butthe other Rap proteins abolished this process.Subsequent studies revealed a pathway, whichis induced by PIP2 enriched apical membraneformation. PIP2 serves as anchor for phospho-lipase D, which produces phosphatidic acid.Phosphatidic acid is then the anchor for theRapGEF PDZ-GEF. As a consequence, PDZ-GEF is recruited to the apical membrane,activates Rap2A, which in turn recruits andactivates the serine-threonine kinase TNIK.TNIK activates the serine-threonine kinaseMst4, which induces the phosphorylation andactivation of Ezrin. This activation of Ezrin is acritical step as active Ezrin can rescue Rap2depletion. Ezrin recruits the actin cytoskeletonfor brush-border formation (Fig. 4, right)(Gloerich et al. 2012). The similarity with theyeast model is evident: A landmark is recog-nized in two steps by a GEF for Rap followedby effector binding, which in several steps leadsto the recruitment of actin.

ARE WE FINISHED WITH Ras?

Although our analysis and those of many otherresearch groups have provided much insightinto the role Rap proteins play in cellular re-sponses, the initial idea that Rap1 counteracts

mutant Ras signaling and could be used asa route to interfere in mutant Ras signalingin cancers turned out to be too naı̈ve. However,despite huge efforts by many groups and manypharmaceutical industries, the alternative ap-proach to interfere in the Ras pathway by selec-tive inhibitors has not worked either as we arestill unable to treat Ras-mutant cancers withtargeted drugs (Stephen et al. 2014). Ras itselfis still largely “undruggable,” and the alternativeto inhibit downstream targets of mutant Ras,most notably Raf, MEK, ERK, and PI3K, havethus far failed in the clinic, especially whensingle drugs are used. For such an approach tobe successful, it is essential that mutant Ras-containing tumors cells do have a specificsensitivity for these pathway drugs. Indeed, ge-netic and drug screens in mutant cell lines dididentify several possible targets for mutant Rastumor cell lines, but these have not been trans-lated into clinical targets (Downward 2015).Perhaps the commonly used mutant Ras-con-taining cell lines identify sensitivities that do notoccur in tumors. We therefore shifted to thetumor organoid model system developed byHans Clevers (Sato et al. 2009; van de Weteringet al. 2015). The advantage of this model systemis that tumor organoids can be rapidly estab-lished from almost all individual (epithelial)tumors and thus can capture the genetic diver-sity of these tumors. Moreover, drug responsescan be compared with organoids from normaltissue to determine the therapeutic window.Initial studies using a number of colon tumororganoids already indicated the power of thistechnology (van de Wetering et al. 2015). As afirst proof-of-concept, we tested various target-ed drugs that are currently being used in clinicaltrials for inhibiting mutant Ras-containingcolorectal cancer, most notably pan-EGFR in-hibitors in combination with MEK inhibitors(Sun et al. 2014). We found that these combi-nations gave a clear cell-cycle arrest of mutantK-ras-containing organoids, supporting theobservation that this combination has an effecton mutant Ras tumor cells. However, organoidswith nonmutated K-Ras remain far more sensi-tive for this drug combination. Furthermore, wenoted that organoids from normal tissue were

From Ras to Rap and Back, A Journey of 35 Years

Cite this article as Cold Spring Harb Perspect Med 2018;8:a031468 9

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on September 30, 2021 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 10: From Ras to Rap and Back, a Journey of 35 Years

similarly sensitive for this combination thantumor organoids with nonmutated K-ras, andthus much more sensitive than mutant K-rasorganoids, indicating that for this combinationthe therapeutic window is limited (Verissimoet al. 2016).

It is now more than 35 years ago that Raswas identified as an oncogene in human cancersand we still have not solved the problem ofhow to target mutant Ras in cancer. As a con-sequence, patients carrying mutant Ras in theirtumors are excluded from therapies targetingthe EGFR signaling cascade. Moreover, whenpatients carrying normal Ras in their tumorsare treated, the appearance of mutant Ras is afrequent resistance mechanism. To make per-sonalized medicine a success, we have to comeup with a solution. New model systems liketumor organoids may be helpful in the identi-fication of new drug combinations, but also infinding those patients that may uniquely re-spond to such combinations.

ACKNOWLEDGMENTS

I thank all members of my laboratory in pastyears for the tremendous contributions theyhave made and are still making to understandRas-like small GTPases and hopefully to find adrug combination that works for mutant Ras-containing tumors. I also thank my colleaguesFried Zwartkruis, Willem Jan Pannekoek, andBoudewijn Burgering for critically reading thismanuscript.

REFERENCES

Almoguera C, Shibata D, Forrester K, Martin J, Arnheim N,Perucho M. 1988. Most human carcinomas of the exo-crine pancreas contain mutant c-K-ras genes. Cell 53:549–554.

Altschuler DL, Peterson SN, Ostrowski MC, Lapetina EG.1995. Cyclic AMP-dependent activation of Rap1b. J BiolChem 270: 10373–10376.

Ando K, Fukuhara S, Moriya T, Obara Y, Nakahata N, Mo-chizuki N. 2013. Rap1 potentiates endothelial cell junc-tions by spatially controlling myosin II activity and actinorganization. J Cell Biol 202: 901–916.

Asha H, de Ruiter ND, Wang MG, Hariharan IK. 1999. TheRap1 GTPase functions as a regulator of morphogenesisin vivo. EMBO J 18: 605–615.

Baas AF, Kuipers J, van der Wel NN, Batlle E, Koerten HK,Peters PJ, Clevers HC. 2004. Complete polarization ofsingle intestinal epithelial cells upon activation of LKB1by STRAD. Cell 116: 457–466.

Bi E, Park HO. 2012. Cell polarization and cytokinesis inbudding yeast. Genetics 191: 347–387.

Birukova AA, Tian X, Tian Y, Higginbotham K, Birukov KG.2013. Rap-afadin axis in control of r signaling and en-dothelial barrier recovery. Mol Biol Cell 24: 2678–2688.

Boettner B, Harjes P, Ishimaru S, Heke M, Fan HQ, Qin Y,Van Aelst L, Gaul U. 2003. The AF-6 homolog canoe actsas a Rap1 effector during dorsal closure of the Drosophilaembryo. Genetics 165: 159–169.

Bos JL. 1989. ras oncogenes in human cancer: A review.Cancer Res 49: 4682–4689.

Bos JL, Verlaan-de Vries M, Jansen AM, Veeneman GH, vanBoom JH, van der Eb AJ. 1984. Three different mutationsin codon 61 of the human N-ras gene detected by syn-thetic oligonucleotide hybridization. Nucleic Acids Res12: 9155–9163.

Bos JL, Fearon ER, Hamilton SR, Verlaan-de Vries M, vanBoom JH, van der Eb AJ, Vogelstein B. 1987. Prevalenceof ras gene mutations in human colorectal cancers.Nature 327: 293–297.

Bos JL, Rehmann H, Wittinghofer A. 2007. GEFs and GAPs:critical elements in the control of small G proteins. Cell129: 865–877.

Broek D, Toda T, Michaeli T, Levin L, Birchmeier C, ZollerM, Powers S, Wigler M. 1987. The S. cerevisiae CDC25gene product regulates the RAS/adenylate cyclase path-way. Cell 48: 789–799.

Burgering BM, Coffer PJ. 1995. Protein kinase B (c-Akt) inphosphatidylinositol-3-OH kinase signal transduction.Nature 376: 599–602.

Burgering BM, Medema RH, Maassen JA, van de WeteringML, van der Eb AJ, McCormick F, Bos JL. 1991. Insulinstimulation of gene expression mediated by p21ras acti-vation. EMBO J 10: 1103–1109.

Conner BJ, Reyes AA, Morin C, Itakura K, Teplitz RL, Wal-lace RB. 1983. Detection of sickle cell b S-globin allele byhybridization with synthetic oligonucleotides. Proc NatlAcad Sci 80: 278–282.

Consonni SV, Gloerich M, Spanjaard E, Bos JL. 2012. cAMPregulates DEP domain-mediated binding of the guaninenucleotide exchange factor Epac1 to phosphatidic acidat the plasma membrane. Proc Natl Acad Sci 109:3814–3819.

Cox AD, Fesik SW, Kimmelman AC, Luo J, Der CJ. 2014.Drugging the undruggable RAS: Mission possible? NatRev Drug Discov 13: 828–851.

de Rooij J, Zwartkruis FJ, Verheijen MH, Cool RH, NijmanSM, Wittinghofer A, Bos JL. 1998. Epac is a Rap1 gua-nine-nucleotide-exchange factor directly activated bycyclic AMP. Nature 396: 474–477.

de Vries-Smits AM, Burgering BM, Leevers SJ, Marshall CJ,Bos JL. 1992. Involvement of p21ras in activation of ex-tracellular signal-regulated kinase 2. Nature 357: 602–604.

Downward J. 2015. RAS synthetic lethal screens revisited:Still seeking the elusive prize? Clin Cancer Res 21: 1802–1809.

J.L. Bos

10 Cite this article as Cold Spring Harb Perspect Med 2018;8:a031468

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on September 30, 2021 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 11: From Ras to Rap and Back, a Journey of 35 Years

Downward J, Graves JD, Warne PH, Rayter S, Cantrell DA.1990. Stimulation of p21ras upon T-cell activation.Nature 346: 719–723.

Eberhard DA, Johnson BE, Amler LC, Goddard AD, Hel-dens SL, Herbst RS, Ince WL, Janne PA, Januario T, John-son DH, et al. 2005. Mutations in the epidermal growthfactor receptor and in KRAS are predictive and prognos-tic indicators in patients with non-small-cell lung cancertreated with chemotherapy alone and in combinationwith erlotinib. J Clin Oncol 23: 5900–5909.

Enserink JM, Christensen AE, de Rooij J, van Triest M,Schwede F, Genieser HG, Doskeland SO, Blank JL, BosJL. 2002. A novel Epac-specific cAMP analogue demon-strates independent regulation of Rap1 and ERK. Nat CellBiol 4: 901–906.

Forrester K, Almoguera C, Han K, Grizzle WE, Perucho M.1987. Detection of high incidence of K-ras oncogenesduring human colon tumorigenesis. Nature 327: 298–303.

Franke B, Akkerman JW, Bos JL. 1997. Rapid Ca2þ-mediat-ed activation of Rap1 in human platelets. EMBO J 16:252–259.

Frische EW, Zwartkruis FJ. 2010. Rap1, a mercenary amongthe Ras-like GTPases. Dev Biol 340: 1–9.

Glading A, Han J, Stockton RA, Ginsberg MH. 2007. KRIT-1/CCM1 is a Rap1 effector that regulates endothelialcell–cell junctions. J Cell Biol 179: 247–254.

Gloerich M, Bos JL. 2010. Epac: Defining a new mechanismfor cAMP action. Annu Rev Pharmacol Toxicol 50: 355–375.

Gloerich M, Ponsioen B, Vliem MJ, Zhang Z, Zhao J, Koois-tra MR, Price LS, Ritsma L, Zwartkruis FJ, Rehmann H,et al. 2010. Spatial regulation of cyclic AMP-Epac1 sig-naling in cell adhesion by ERM proteins. Mol Cell Biol 30:5421–5431.

Gloerich M, ten Klooster JP, Vliem MJ, Koorman T, Zwart-kruis FJ, Clevers H, Bos JL. 2012. Rap2A links intestinalcell polarity to brush border formation. Nat Cell Biol 14:793–801.

Gotoh T, Hattori S, Nakamura S, Kitayama H, Noda M,Takai Y, Kaibuchi K, Matsui H, Hatase O, Takahashi H,et al. 1995. Identification of Rap1 as a target for the CrkSH3 domain-binding guanine nucleotide-releasing fac-tor C3G. Mol Cell Biol 15: 6746–6753.

Hall A, Marshall CJ, Spurr NK, Weiss RA. 1983. Identifica-tion of transforming gene in two human sarcoma celllines as a new member of the ras gene family located onchromosome 1. Nature 303: 396–400.

Hofer F, Fields S, Schneider C, Martin GS. 1994. ActivatedRas interacts with the Ral guanine nucleotide dissocia-tion stimulator. Proc Natl Acad Sci 91: 11089–11093.

Karapetis CS, Khambata-Ford S, Jonker DJ, O’Callaghan CJ,Tu D, Tebbutt NC, Simes RJ, Chalchal H, Shapiro JD,Robitaille S, et al. 2008. K-ras mutations and benefitfrom cetuximab in advanced colorectal cancer. N Engl JMed 359: 1757–1765.

Kitayama H, Sugimoto Y, Matsuzaki T, Ikawa Y, Noda M.1989. A ras-related gene with transformation suppressoractivity. Cell 56: 77–84.

Knox AL, Brown NH. 2002. Rap1 GTPase regulation ofadherens junction positioning and cell adhesion. Science295: 1285–1288.

Kodaki T, Woscholski R, Hallberg B, Rodriguez-Viciana P,Downward J, Parker PJ. 1994. The activation of phospha-tidylinositol 3-kinase by Ras. Curr Biol 4: 798–806.

Kops GJ, de Ruiter ND, De Vries-Smits AM, Powell DR, BosJL, Burgering BM. 1999. Direct control of the Forkheadtranscription factor AFX by protein kinase B. Nature 398:630–634.

Lafuente EM, van Puijenbroek AA, Krause M, Carman CV,Freeman GJ, Berezovskaya A, Constantine E, Springer TA,Gertler FB, Boussiotis VA. 2004. RIAM, an Ena/VASP andProfilin ligand, interacts with Rap1-GTP and mediatesRap1-induced adhesion. Dev Cell 7: 585–595.

Leevers SJ, Marshall CJ. 1992. Activation of extracellularsignal-regulated kinase, ERK2, by p21ras oncoprotein.EMBO J 11: 569–574.

Li S, Tsalkova T, White MA, Mei FC, Liu T, Wang D, WoodsVL Jr, Cheng X. 2011. Mechanism of intracellular cAMPsensor Epac2 activation: cAMP-induced conformationalchanges identified by amide hydrogen/deuterium ex-change mass spectrometry (DXMS). J Biol Chem 286:17889–17897.

Moodie SA, Willumsen BM, Weber MJ, Wolfman A. 1993.Complexes of Ras.GTP with Raf-1 and mitogen-activatedprotein kinase kinase. Science 260: 1658–1661.

Mulder MP, Keijzer W, Verkerk A, Boot AJ, Prins ME, Splin-ter TA, Bos JL. 1989. Activated ras genes in human semi-noma: Evidence for tumor heterogeneity. Oncogene 4:1345–1351.

Pannekoek WJ, van Dijk JJ, Chan OY, Huveneers S, Linne-mann JR, Spanjaard E, Brouwer PM, van der Meer AJ,Zwartkruis FJ, Rehmann H, et al. 2011. Epac1 and PDZ-GEF cooperate in Rap1 mediated endothelial junctioncontrol. Cell Signal 23: 2056–2064.

Papke B, Der CJ. 2017. Drugging RAS: Know the enemy.Science 355: 1158–1163.

Parada LF, Tabin CJ, Shih C, Weinberg RA. 1982. Human EJbladder carcinoma oncogene is homologue of Harveysarcoma virus ras gene. Nature 297: 474–478.

Pizon V, Lerosey I, Chardin P, Tavitian A. 1988. Nucleotidesequence of a human cDNA encoding a ras-related pro-tein (rap1B). Nucleic Acids Res 16: 7719.

Ponsioen B, Gloerich M, Ritsma L, Rehmann H, Bos JL,Jalink K. 2009. Direct spatial control of Epac1 by cyclicAMP. Mol Cell Biol 29: 2521–2531.

Post A, Pannekoek WJ, Ross SH, Verlaan I, Brouwer PM, BosJL. 2013. Rasip1 mediates Rap1 regulation of Rhor inendothelial barrier function through ArhGAP29. ProcNatl Acad Sci 110: 11427–11432.

Post A, Pannekoek WJ, Ponsioen B, Vliem MJ, Bos JL. 2015.Rap1 spatially controls ArhGAP29 to inhibit Rho signal-ing during endothelial barrier regulation. Mol Cell Biol35: 2495–2502.

Price LS, Hajdo-Milasinovic A, Zhao J, Zwartkruis FJ, Col-lard JG, Bos JL. 2004. Rap1 regulates E-cadherin-medi-ated cell–cell adhesion. J Biol Chem 279: 35127–35132.

Rangarajan S, Enserink JM, Kuiperij HB, de Rooij J, Price LS,Schwede F, Bos JL. 2003. Cyclic AMP induces integrin-mediated cell adhesion through Epac and Rap1 upon

From Ras to Rap and Back, A Journey of 35 Years

Cite this article as Cold Spring Harb Perspect Med 2018;8:a031468 11

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on September 30, 2021 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 12: From Ras to Rap and Back, a Journey of 35 Years

stimulation of the b2-adrenergic receptor. J Cell Biol 160:487–493.

Rehmann H, Prakash B, Wolf E, Rueppel A, de Rooij J, BosJL, Wittinghofer A. 2003. Structure and regulation of thecAMP-binding domains of Epac2. Nat Struct Biol 10: 26–32.

Rehmann H, Das J, Knipscheer P, Wittinghofer A, Bos JL.2006. Structure of the cyclic-AMP-responsive exchangefactor Epac2 in its auto-inhibited state. Nature 439: 625–628.

Rehmann H, Arias-Palomo E, Hadders MA, Schwede F,Llorca O, Bos JL. 2008. Structure of Epac2 in complexwith a cyclic AMP analogue and RAP1B. Nature 455:124–127.

Robinson LC, Gibbs JB, Marshall MS, Sigal IS, Tatchell K.1987. CDC25: A component of the RAS-adenylate cyclasepathway in Saccharomyces cerevisiae. Science 235: 1218–1221.

Rodenhuis S, van de Wetering ML, Mooi WJ, Evers SG, vanZandwijk N, Bos JL. 1987. Mutational activation of theK-ras oncogene. A possible pathogenetic factor in ade-nocarcinoma of the lung. N Engl J Med 317: 929–935.

Saiki RK, Scharf S, Faloona F, Mullis KB, Horn GT, ErlichHA, Arnheim N. 1985. Enzymatic amplification of b-globin genomic sequences and restriction site analysisfor diagnosis of sickle cell anemia. Science 230: 1350–1354.

Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N,Stange DE, van Es JH, Abo A, Kujala P, Peters PJ, et al.2009. Single Lgr5 stem cells build crypt-villus structuresin vitro without a mesenchymal niche. Nature 459: 262–265.

Shimizu K, Birnbaum D, Ruley MA, Fasano O, Suard Y,Edlund L, Taparowsky E, Goldfarb M, Wigler M. 1983.Structure of the Ki-ras gene of the human lung carcinomacell line Calu-1. Nature 304: 497–500.

Sot B, Behrmann E, Raunser S, Wittinghofer A. 2013. RasGTPase activating (RasGAP) activity of the dual specific-ity GAP protein Rasal requires colocalization and C2domain binding to lipid membranes. Proc Natl Acad Sci110: 111–116.

Stephen AG, Esposito D, Bagni RK, McCormick F. 2014.Dragging ras back in the ring. Cancer Cell 25: 272–281.

Sun C, Hobor S, Bertotti A, Zecchin D, Huang S, Galimi F,Cottino F, Prahallad A, Grernrum W, Tzani A, et al. 2014.Intrinsic resistance to MEK inhibition in KRAS mutantlung and colon cancer through transcriptional inductionof ERBB3. Cell Rep 7: 86–93.

Taira K, Umikawa M, Takei K, Myagmar BE, Shinzato M,Machida N, Uezato H, Nonaka S, Kariya K. 2004. The

Traf2- and Nck-interacting kinase as a putative effector ofRap2 to regulate actin cytoskeleton. J Biol Chem 279:49488–49496.

Trahey M, McCormick F. 1987. A cytoplasmic protein stim-ulates normal N-ras p21 GTPase, but does not affectoncogenic mutants. Science 238: 542–545.

van Corven EJ, Hordijk PL, Medema RH, Bos JL, MoolenaarWH. 1993. Pertussis toxin-sensitive activation of p21rasby G protein-coupled receptor agonists in fibroblasts.Proc Natl Acad Sci 90: 1257–1261.

van Dam TJ, Bos JL, Snel B. 2011. Evolution of the Ras-likesmall GTPases and their regulators. Small GTPases 2: 4–16.

van de Wetering M, Francies HE, Francis JM, Bounova G,Iorio F, Pronk A, van Houdt W, van Gorp J, Taylor-WeinerA, Kester L, et al. 2015. Prospective derivation of a livingorganoid biobank of colorectal cancer patients. Cell 161:933–945.

van ’t Veer LJ, Burgering BM, Versteeg R, Boot AJ, Ruiter DJ,Osanto S, Schrier PI, Bos JL. 1989. N-ras mutations inhuman cutaneous melanoma from sun-exposed bodysites. Mol Cell Biol 9: 3114–3116.

Verissimo CS, Overmeer RM, Ponsioen B, Drost J, MertensS, Verlaan-Klink I, Gerwen BV, van der Ven M, WeteringMV, Egan DA, et al. 2016. Targeting mutant RAS in pa-tient-derived colorectal cancer organoids by combinato-rial drug screening. eLife 5: e18489.

Verlaan-de Vries M, Bogaard ME, van den Elst H, van BoomJH, van der Eb AJ, Bos JL. 1986. A dot-blot screeningprocedure for mutated ras oncogenes using synthetic oli-godeoxynucleotides. Gene 50: 313–320.

Vogelstein B, Fearon ER, Hamilton SR, Kern SE, PreisingerAC, Leppert M, Nakamura Y, White R, Smits AM, Bos JL.1988. Genetic alterations during colorectal-tumor devel-opment. N Engl J Med 319: 525–532.

White MA, Nicolette C, Minden A, Polverino A, Van Aelst L,Karin M, Wigler MH. 1995. Multiple Ras functions cancontribute to mammalian cell transformation. Cell 80:533–541.

Wong G, Muller O, Clark R, Conroy L, Moran MF, Polakis P,McCormick F. 1992. Molecular cloning and nucleic acidbinding properties of the GAP-associated tyrosine phos-phoprotein p62. Cell 69: 551–558.

Yunis JJ, Boot AJ, Mayer MG, Bos JL. 1989. Mechanisms ofras mutation in myelodysplastic syndrome. Oncogene 4:609–614.

Zwartkruis FJ, Wolthuis RM, Nabben NM, Franke B, Bos JL.1998. Extracellular signal-regulated activation of Rap1fails to interfere in Ras effector signalling. EMBO J 17:5905–5912.

J.L. Bos

12 Cite this article as Cold Spring Harb Perspect Med 2018;8:a031468

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on September 30, 2021 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Page 13: From Ras to Rap and Back, a Journey of 35 Years

August 4, 20172018; doi: 10.1101/cshperspect.a031468 originally published onlineCold Spring Harb Perspect Med 

 Johannes L. Bos From Ras to Rap and Back, a Journey of 35 Years

Subject Collection Ras and Cancer in the 21st Century

Targeting Ras with MacromoleculesDehua Pei, Kuangyu Chen and Hui Liao RAS Family

MRAS: A Close but Understudied Member of the

Lucy C. Young and Pablo Rodriguez-Viciana

Structures, Mechanisms, and Interactions−−Ras-Specific GTPase-Activating Proteins

Klaus Scheffzek and Giridhar ShivalingaiahSon-of-Sevenless and RasThe Interdependent Activation of

KuriyanPradeep Bandaru, Yasushi Kondo and John

KinasesRas-Mediated Activation of the Raf Family

Elizabeth M. Terrell and Deborah K. MorrisonCancersTargeting the MAPK Pathway in RAS Mutant

Sarah G. Hymowitz and Shiva MalekPosttranslational Modifications of RAS Proteins

Ian Ahearn, Mo Zhou and Mark R. Philips RelationshipRas and the Plasma Membrane: A Complicated

Gorfe, et al.Yong Zhou, Priyanka Prakash, Alemayehu A.

Kras in OrganoidsDerek Cheng and David Tuveson

Kras and Tumor Immunity: Friend or Foe?Jane Cullis, Shipra Das and Dafna Bar-Sagi

for Pancreatic CancerKRAS: The Critical Driver and Therapeutic Target

Andrew M. Waters and Channing J. DerCancers

-MutantKRASSynthetic Lethal Vulnerabilities in

Andrew J. Aguirre and William C. Hahn

Targeting Oncogenic MutantsConformational Preferences and Implications for The K-Ras, N-Ras, and H-Ras Isoforms: Unique

Jillian A. Parker and Carla Mattos

Efforts to Develop KRAS InhibitorsMatthew Holderfield

PI3K: A Crucial Piece in the RAS Signaling PuzzleAgata Adelajda Krygowska and Esther Castellano

Validation of Therapeutic TargetsK-Ras-Driven Lung and Pancreatic Tumors: Genetically Engineered Mouse Models of

BarbacidMatthias Drosten, Carmen Guerra and Mariano

http://perspectivesinmedicine.cshlp.org/cgi/collection/ For additional articles in this collection, see

Copyright © 2018 Cold Spring Harbor Laboratory Press; all rights reserved

Press on September 30, 2021 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from


Recommended