mras gtpase is a novel stemness marker that impacts mouse ... · differentiate toward the...

12
STEM CELLS AND REGENERATION RESEARCH ARTICLE 3311 Development 140, 3311-3322 (2013) doi:10.1242/dev.091082 © 2013. Published by The Company of Biologists Ltd INTRODUCTION Studies of stemness status in mouse embryonic stem cells (mESCs) through transcriptomic, proteomic and drug screenings have enabled the identification of many gene families potentially involved in the maintenance of cell pluripotency, among which are the regulators of Ras family (Chen et al., 2006; Ivanova et al., 2006; Wray et al., 2010). Understanding the function of newly discovered genes involved in the control of the pluripotency/differentiation balance in stem cells requires the use of complementary models to address cell biology and developmental questions in fast and efficient ways. In this report, we have characterised the effects of a novel regulated GTPase, muscle ras oncogene homologue (MRAS; also known as muscle and microspikes RAS), in mESCs and in Xenopus laevis. The convenient and efficient manipulation of gene expression in Xenopus embryos, along with studies performed in the mESC model, allow information concerning the basic conserved function of genes during evolution to be examined. mESCs are maintained in a pluripotent state in the presence of the leukemia inhibitory factor (LIF) cytokine, which induces the JAK1/STAT3/SOCS3 pathway that forms part of the pluripotency circuitry along with Wnt and bone morphogenetic protein 4 (BMP4) secreted factors, and the master genes such as Oct4 (Pou5f1), Nanog and Sox2 (Boeuf et al., 1997; Duval et al., 2000; Niwa, 2007; Chambers and Tomlinson, 2009; Trouillas et al., 2009a). Extensive microarray studies, performed in mESCs grown under pluripotent conditions (with LIF) or induced to differentiate (24 hours or 48 hours without LIF) and restimulated for a short period of time by LIF, have led to the identification of three LIF signatures (Pluri, Spe-Lifind and Pleio-Lifind) (Mathieu et al., 2012; Trouillas et al., 2009a; Trouillas et al., 2009b). An interesting feature of genes from the Pluri cluster is their early downregulation upon LIF withdrawal as compared with the master genes (Schulz et al., 2009; Trouillas et al., 2009b). Mras is a member of the R-ras subfamily of the large Ras family and encodes a small GTPase (Díez et al., 2011). In mESCs, Mras is in the Pluri gene cluster, with an expression profile similar to that of Esrrb and Tcl1 (Ivanova et al., 2006; Trouillas et al., 2009b). Mras expression is under the control of the KLF5 transcription factor (Parisi and Russo, 2011; Parisi et al., 2010), with elevated transcripts in specific brain areas, heart and the developing bones. The protein is concentrated on plasma membrane-associated structures (Kimmelman et al., 1997; Kimmelman et al., 2002; Matsumoto et al., 1997; Watanabe-Takano et al., 2010). MRAS expression and its GTPase activity are stimulated by nerve growth factor (NGF) in pheochromocytoma PC12 cells, in which it triggers ERK-dependent neuronal differentiation (Kimmelman et al., 2002; Sun et al., 2006). MRAS expression is also stimulated by the BMP2 signalling pathway in myoblast C2C12 cells, in which it induces osteoblast differentiation under the control of the p38 and JNK kinases (Watanabe-Takano et al., 2010). Activated forms of MRAS, such as the MRAS-G22V mutant which is constitutively in the GTP-bound form, mimic inducers (NGF or BMP2) and trigger neuronal or osteoblast differentiation in PC12 or C2C12 models, respectively (Kimmelman et al., 2002; Sun et al., 2006; Watanabe- Takano et al., 2010), induce formation of peripheral microspikes in fibroblasts and participate in the reorganisation of the actin 1 University of Bordeaux, CIRID, UMR 5164, F-33000 Bordeaux, France. 2 CNRS, CIRID, UMR 5164, F-33000 Bordeaux, France. *Present address: IRHT, F-68100 Mulhouse, France Present address: INSERM U1029, F-33405 Talence, France § Present address: CTSA, F-92141 Clamart, France Authors for correspondence ([email protected]; [email protected]) Accepted 4 June 2013 SUMMARY Pluripotent mouse embryonic stem cells (mESCs), maintained in the presence of the leukemia inhibitory factor (LIF) cytokine, provide a powerful model with which to study pluripotency and differentiation programs. Extensive microarray studies on cultured cells have led to the identification of three LIF signatures. Here we focus on muscle ras oncogene homolog (MRAS), which is a small GTPase of the Ras family encoded within the Pluri gene cluster. To characterise the effects of Mras on cell pluripotency and differentiation, we used gain- and loss-of-function strategies in mESCs and in the Xenopus laevis embryo, in which Mras gene structure and protein sequence are conserved. We show that persistent knockdown of Mras in mESCs reduces expression of specific master genes and that MRAS plays a crucial role in the downregulation of OCT4 and NANOG protein levels upon differentiation. In Xenopus, we demonstrate the potential of Mras to modulate cell fate at early steps of development and during neurogenesis. Overexpression of Mras allows gastrula cells to retain responsiveness to fibroblast growth factor (FGF) and activin. Collectively, these results highlight novel conserved and pleiotropic effects of MRAS in stem cells and early steps of development. KEY WORDS: LIF, Mras, mES cells, Neurogenesis, Pluripotency, Mouse, Xenopus MRAS GTPase is a novel stemness marker that impacts mouse embryonic stem cell plasticity and Xenopus embryonic cell fate Marie-Emmanuelle Mathieu 1,2 , Corinne Faucheux 1,2 , Claire Saucourt 1,2, *, Fabienne Soulet 1,2,‡ , Xavier Gauthereau 1,2 , Sandrine Fédou 1,2 , Marina Trouillas 1,§ , Nadine Thézé 1,2 , Pierre Thiébaud 1,2,¶ and Hélène Bœuf 1,2,¶ DEVELOPMENT

Upload: others

Post on 24-Nov-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: MRAS GTPase is a novel stemness marker that impacts mouse ... · differentiate toward the lineage-specific cell types of the three germ layers (Lan et al., 2009; Okabayashi and Asashima,

STEM CELLS AND REGENERATION RESEARCH ARTICLE 3311

Development 140, 3311-3322 (2013) doi:10.1242/dev.091082© 2013. Published by The Company of Biologists Ltd

INTRODUCTIONStudies of stemness status in mouse embryonic stem cells (mESCs)through transcriptomic, proteomic and drug screenings haveenabled the identification of many gene families potentiallyinvolved in the maintenance of cell pluripotency, among which arethe regulators of Ras family (Chen et al., 2006; Ivanova et al., 2006;Wray et al., 2010). Understanding the function of newly discoveredgenes involved in the control of the pluripotency/differentiationbalance in stem cells requires the use of complementary models toaddress cell biology and developmental questions in fast andefficient ways. In this report, we have characterised the effects of anovel regulated GTPase, muscle ras oncogene homologue (MRAS;also known as muscle and microspikes RAS), in mESCs and inXenopus laevis. The convenient and efficient manipulation of geneexpression in Xenopus embryos, along with studies performed inthe mESC model, allow information concerning the basic conservedfunction of genes during evolution to be examined.

mESCs are maintained in a pluripotent state in the presence ofthe leukemia inhibitory factor (LIF) cytokine, which induces theJAK1/STAT3/SOCS3 pathway that forms part of the pluripotencycircuitry along with Wnt and bone morphogenetic protein 4 (BMP4)secreted factors, and the master genes such as Oct4 (Pou5f1), Nanogand Sox2 (Boeuf et al., 1997; Duval et al., 2000; Niwa, 2007;

Chambers and Tomlinson, 2009; Trouillas et al., 2009a). Extensivemicroarray studies, performed in mESCs grown under pluripotentconditions (with LIF) or induced to differentiate (24 hours or48 hours without LIF) and restimulated for a short period of time byLIF, have led to the identification of three LIF signatures (Pluri,Spe-Lifind and Pleio-Lifind) (Mathieu et al., 2012; Trouillas et al.,2009a; Trouillas et al., 2009b). An interesting feature of genes fromthe Pluri cluster is their early downregulation upon LIF withdrawalas compared with the master genes (Schulz et al., 2009; Trouillas etal., 2009b).

Mras is a member of the R-ras subfamily of the large Ras familyand encodes a small GTPase (Díez et al., 2011). In mESCs, Mras isin the Pluri gene cluster, with an expression profile similar to thatof Esrrb and Tcl1 (Ivanova et al., 2006; Trouillas et al., 2009b).Mras expression is under the control of the KLF5 transcriptionfactor (Parisi and Russo, 2011; Parisi et al., 2010), with elevatedtranscripts in specific brain areas, heart and the developing bones.The protein is concentrated on plasma membrane-associatedstructures (Kimmelman et al., 1997; Kimmelman et al., 2002;Matsumoto et al., 1997; Watanabe-Takano et al., 2010). MRASexpression and its GTPase activity are stimulated by nerve growthfactor (NGF) in pheochromocytoma PC12 cells, in which it triggersERK-dependent neuronal differentiation (Kimmelman et al., 2002;Sun et al., 2006). MRAS expression is also stimulated by the BMP2signalling pathway in myoblast C2C12 cells, in which it inducesosteoblast differentiation under the control of the p38 and JNKkinases (Watanabe-Takano et al., 2010). Activated forms of MRAS,such as the MRAS-G22V mutant which is constitutively in theGTP-bound form, mimic inducers (NGF or BMP2) and triggerneuronal or osteoblast differentiation in PC12 or C2C12 models,respectively (Kimmelman et al., 2002; Sun et al., 2006; Watanabe-Takano et al., 2010), induce formation of peripheral microspikes infibroblasts and participate in the reorganisation of the actin

1University of Bordeaux, CIRID, UMR 5164, F-33000 Bordeaux, France. 2CNRS,CIRID, UMR 5164, F-33000 Bordeaux, France.

*Present address: IRHT, F-68100 Mulhouse, France‡Present address: INSERM U1029, F-33405 Talence, France§Present address: CTSA, F-92141 Clamart, France¶Authors for correspondence ([email protected]; [email protected])

Accepted 4 June 2013

SUMMARYPluripotent mouse embryonic stem cells (mESCs), maintained in the presence of the leukemia inhibitory factor (LIF) cytokine, providea powerful model with which to study pluripotency and differentiation programs. Extensive microarray studies on cultured cells haveled to the identification of three LIF signatures. Here we focus on muscle ras oncogene homolog (MRAS), which is a small GTPase ofthe Ras family encoded within the Pluri gene cluster. To characterise the effects of Mras on cell pluripotency and differentiation, weused gain- and loss-of-function strategies in mESCs and in the Xenopus laevis embryo, in which Mras gene structure and proteinsequence are conserved. We show that persistent knockdown of Mras in mESCs reduces expression of specific master genes and thatMRAS plays a crucial role in the downregulation of OCT4 and NANOG protein levels upon differentiation. In Xenopus, we demonstratethe potential of Mras to modulate cell fate at early steps of development and during neurogenesis. Overexpression of Mras allowsgastrula cells to retain responsiveness to fibroblast growth factor (FGF) and activin. Collectively, these results highlight novel conservedand pleiotropic effects of MRAS in stem cells and early steps of development.

KEY WORDS: LIF, Mras, mES cells, Neurogenesis, Pluripotency, Mouse, Xenopus

MRAS GTPase is a novel stemness marker that impacts mouse embryonic stem cell plasticity and Xenopus embryoniccell fateMarie-Emmanuelle Mathieu1,2, Corinne Faucheux1,2, Claire Saucourt1,2,*, Fabienne Soulet1,2,‡, Xavier Gauthereau1,2, Sandrine Fédou1,2, Marina Trouillas1,§, Nadine Thézé1,2, Pierre Thiébaud1,2,¶ and Hélène Bœuf1,2,¶

DEVELO

PMENT

Page 2: MRAS GTPase is a novel stemness marker that impacts mouse ... · differentiate toward the lineage-specific cell types of the three germ layers (Lan et al., 2009; Okabayashi and Asashima,

3312

cytoskeleton (Matsumoto et al., 1997). In addition, these activatedforms are oncogenic in murine mammary epithelial cell lines (Wardet al., 2004). MRAS function is potentially regulated by plexin B1,a dual-function GTPase-activating protein (GAP) for RRAS andMRAS, which remodels axon and dendrite morphology,respectively (Saito et al., 2009).

There is a single Mras gene from nematodes to mammals andin the ascidian embryo the orthologue has a role in neuraldifferentiation, suggesting an evolutionarily conserved function(Keduka et al., 2009). The Xenopus embryo has contributedsubstantially to the deciphering of the molecular mechanisms thatcontrol embryo patterning, which relies on signalling pathwaysthat are highly conserved between mammals and amphibians(Harland and Grainger, 2011; Heasman, 2006). LIF and itsreceptors (gp130 and gp190), along with the Pluri genes (such asEsrrb, Sod2, Cd9, Fzd5, Susd2 and Smarcd3), are conserved inXenopus laevis (our unpublished data). In addition, cells from theanimal part of the Xenopus blastula embryo (animal cap cells) aresimilar to mammalian ESCs. They are pluripotent and able todifferentiate toward the lineage-specific cell types of the threegerm layers (Lan et al., 2009; Okabayashi and Asashima, 2003).Moreover, the molecular mechanisms that govern self-renewalmediated by Oct4 are conserved between mammals and Xenopus(Morrison and Brickman, 2006; Abu-Remaileh et al., 2010). Thismakes Xenopus a suitable model in which the basic conservedfunctions of these genes can be studied during earlyembryogenesis and organogenesis, two processes not easilyamenable in the mouse embryo. For instance, our recent workusing a combined approach of mESCs and Xenopus embryoniccells has demonstrated a conserved function for a histonemethyltransferase adaptor protein in smooth muscle celldifferentiation (Gan et al., 2011).

In this study, we have carried out a functional analysis of Mras inmESC and Xenopus models, leading to the characterisation of Mrasas a novel stemness marker that impacts on pluripotent cells and onneurogenesis.

MATERIALS AND METHODSEthics statementThis study was carried out in accordance with the European CommunityGuide for Care and Use of Laboratory Animals and approved by the Comitéd’éthique en expérimentation de Bordeaux, No. 33011005-A.

Cell cultureCGR8, E14Tg2aWT and E14Tg2a GeneTrap Mras (feeder-free) ESC lineswere grown as described (Trouillas et al., 2009b). The E14Tg2a GeneTrapMras cell line (E14Dmras) was purchased from the Mutant Mouse RegionalResource Center (MMRRC) under the strain name BayGenomics ESC lineCSH312, stock number 014915-UCD. This cell line contains in exon 2 ofMras an insertion of the β-geo fusion cDNA in one allele. A half dose ofMras expression was validated with two sets of primers (sequences arelisted in supplementary material Table S1). The E14Tg2aWT (E14WT)control cell line was purchased from ATCC.

Heat map analysisHeat map analysis was performed using the FunGenES website interface(Schulz et al., 2009; Trouillas et al., 2009b).

Cell lysates and western blotsPlated ESCs were rinsed twice at room temperature with PBS and lyseddirectly in mild RIPA buffer (Trouillas et al., 2009b). Total protein (100 µg)was loaded on 10% SDS-PAGE gels, transferred to nitrocellulosemembranes and incubated with anti-OCT4 (Abcam, 18976; 1:500), anti-NANOG (Abcam, 21603; 1:1000) and anti-ERK2 (Santa Cruz Biotech, sc-154; 1:5000) antibodies.

Flow cytometryPlated cells were washed once at room temperature with PBS, trypsinisedand counted. Cells were fixed for 20 minutes in 4% paraformaldehyde,permeabilised with Perm/Wash Buffer (BD Biosciences) for 10 minutes andlabelled with 20 µl antibody for 1 hour with the Mouse Pluripotent StemCell Transcription Factor Analysis Kit (BD Biosciences). After two washeswith the Wash Buffer for 10 minutes, cells were processed on a FACSCantoII flow cytometer (BD Biosciences). The percentage of positive cells and themean fluorescence intensity (MFI) were obtained with DIVA software (BDBiosciences).

RT-PCR and RT-qPCRTotal RNA from adherent ESCs was prepared with Trizol reagent as described(Trouillas et al., 2009b). The reverse transcription reaction products were usedfor qPCR with specific primer sets and relative expression was calculatedusing Hprt (CGR8 cell line) or Erk2 (Mapk1) (E14 cell lines) as referencegenes. RNA from Xenopus embryo and explants was prepared and analysedaccording to standard protocols (Sive et al., 2000; Naye et al., 2007). Primersare listed in supplementary material Table S1.

Construction of expression vectors and establishment of stableclonesExpression vector constructionRT-PCR-amplified fragments of Mus musculus Mras (NM_008624) wereinserted in fusion with EGFP cDNA in the NheI/XhoI site of the pCXN2vector (Niwa et al., 1991). A 627 bp Xenopus laevis Mras cDNA was clonedby RT-PCR from stage 20 embryo RNA into the EcoRI/XhoI sites of thepCS2 vector (a gift from R. Rupp) using gene-specific primers: forward,5�-cgcggaattcATGGCAACGAGTGCTGTTCTGAG-3�; reverse, 5�-cgcgct -cgagTCACAATACGACACACTGCAGCCTG-3�.

Mouse Mras was cloned from pCAG-EGFP-MRAS into pCS2 bydigestion with NheI (end filled)/XhoI and ligation into the EcoRI (endfilled)/XhoI sites of pCS2. MRAS G22V and MRAS S27N encodingsequences were cloned from the pEF-BOS-MycMras constructs (Sun et al.,2006) into the BamHI and XbaI sites of pCS2. Plasmid constructs werechecked by DNA sequencing.

Derivation of mESC stable clonesTo establish ESC clones expressing EGFP or EGFP-MRAS, CGR8 cellsmaintained in LIF-containing medium were transfected usingLipofectamine (Invitrogen) with 0.5-2 µg pCXN2-derived construct per 6-cm Petri dish. After 2 weeks (for EGFP) and up to 4 weeks (for EGFP-MRAS) of selection in the presence of 300 µg/ml G418, double-positiveclones (NeoR and EGFP+) were picked and grown in the presence of G418.Images were captured on a Zeiss Axiovert 200 microscope with anAxioCam HR camera and AxioVision 3.0 and Axioviewer 3.0 software.

Embryological methodsXenopus laevis embryos were obtained by standard procedures (Sive et al.,2000) and staged according to Nieuwkoop and Faber (Nieuwkoop andFaber, 1967). Animal cap explants were dissected from stage 8 or stage 11embryos, treated with FGF2 or activin (R&D Systems) and cultured untilthe control embryos reached the appropriate stage before RT-PCR analysis.For microinjection experiments, capped mRNAs were synthesised usingthe mMessenger mMachine SP6 Kit (Ambion). Injections were performedinto one blastomere at the 2-cell stage, with 250 pg lacZ mRNA as lineagetracer. For knockdown experiments, 20-40 ng morpholinos (MOs) (GeneTools; supplementary material Table S2) were injected into 2-cell stageembryos. In the animal cap assay, 500 pg Mras mRNA and 50 pg nogginmRNA were injected into the animal pole of the 2-cell stage embryo in bothblastomeres.

Whole-mount in situ hybridisation and immunohistochemistryIn situ hybridisation and immunohistochemistry were performed accordingto standard protocols (Sive et al., 2000). For serial sections, embryos post-fixed in MEMFA (0.1 M MOPS, 2 mM EGTA, 1 mM MgSO4 and 3.7%formaldehyde) were embedded in Paraplast (Sigma) and cut on amicrotome. The 12/101 antibody (Developmental Studies Hybridoma Bank)was used at 1:2 dilution.

RESEARCH ARTICLE Development 140 (16)

DEVELO

PMENT

Page 3: MRAS GTPase is a novel stemness marker that impacts mouse ... · differentiate toward the lineage-specific cell types of the three germ layers (Lan et al., 2009; Okabayashi and Asashima,

In vitro translationIn vitro transcribed mRNAs (500 ng) were translated in rabbit reticulocytelysate (Promega) and the reaction products were analysed by 12% SDS-PAGE followed by autoradiography.

Sequence analysisThe Mus musculus MRAS protein sequence (NM_008624) was BLASTedin the SWISS-PROT database and the sequences of various organisms wereretrieved and aligned using the MultiAlin tool (Corpet, 1988). Phylogeneticanalyses were performed with the following sequences: Xenopus laevis (Xl),NP_001085506; Homo sapiens (Hs), NM_012219; Canis familiaris (Cf),XP_534277; Mus musculus (Mm), NM_008624; Rattus norvegicus (Rn),NM_012981; Gallus gallus (Gg), NM_204489; Danio rerio (Dr), 003201047.Genes structure and intron size comparisons were retrieved from Ensembl.

RESULTSMras, a new marker of stemnessExtensive microarray analyses performed with mESCs grown underdifferent culture conditions (pluripotency or differentiationinduction) have enabled the identification of novel markers ofpluripotent mESCs and the characterisation of a set of Pluri genesthat are highly expressed in undifferentiated mESCs (Schulz et al.,2009; Trouillas et al., 2009a; Trouillas et al., 2009b). Here, we focuson the expression profiles of Ras family members. Probe setnumbers of this gene family and related members (including theRas-Gap and Ras-Gef genes, 273 probe sets) were retrieved fromthe Affymetrix 420.2 microarray and their expression profilesanalysed in pluripotent cells and after LIF withdrawal from 1 to 10days. A heat map representation of a selection of genes (46 probesets) is presented (Fig. 1A).

This analysis showed heterogeneous expression profiles of Rasmembers, and a particular profile for Mras. Indeed it is the onlymember that is restricted in its expression to undifferentiated ESCs,with a drastic reduction of expression as early as 1 day after LIFwithdrawal. We confirmed, using RT-qPCR analysis, thatexpression of Mras, but not Hras, Kras, Rras or Eras, is regulatedby LIF (Fig. 1B). We also showed that Mras expression is underLIF control only in undifferentiated cells. Indeed, Mras expressionis not induced by short LIF treatment in committed (24 hours minusLIF) or differentiated (48 hours minus LIF) cells, in contrast to theLifind genes, which respond to LIF in ESC-derived committed anddifferentiated cells and in other mature LIF-responsive cell types(Trouillas et al., 2009a; Trouillas et al., 2009b; Mathieu et al., 2012).These results revealed that Mras constitutes a new marker ofstemness.

Persistent downregulation of Mras leads to adecrease in the expression of core pluripotencymarkersTo determine whether Mras controls the fate of pluripotent cells,the expression of the core pluripotent genes (Oct4, Sox2 and Nanog)and a selection of Pluri or differentiation markers was examined incells in which Mras expression was knocked down. We used aclonal heterozygous GeneTrap Mras ESC line (E14Dmras), whichharbours an insertion of a β-geo cassette in exon 2 (see Materialsand methods). Mras expression was decreased by 50% in this cellline when compared with the parental E14WT mESC line (Fig. 2A).This mutated cell line, despite being alkaline phosphatase positivein the presence of LIF, displays a reduced self-renewal efficiency(7%, versus 35% for E14WT; supplementary material Fig. S1A).

To further characterise the deficit caused by decreasedexpression of Mras, E14WT and E14Dmras were grown in thepresence or absence of LIF for 4 days. Whereas there was no

3313RESEARCH ARTICLEMras in mESCs and Xenopus embryo

Fig. 1. Mras has a unique expression profile among Ras and effector-of-Ras proteins. (A) Heat map analysis of expression profiles of Rasmembers and regulators (Gef in red and Gap in blue) on a timescale of LIFdepletion from days 1 to 10 in mESCs. (B) RT-qPCR analysis of Ras familymembers in cells grown with or without LIF for 24 hours or 48 hours andreinduced for 25 minutes with foetal calf serum (FCS) or FCS + LIF. Meanof expression with s.d. of triplicates is shown. D

EVELO

PMENT

Page 4: MRAS GTPase is a novel stemness marker that impacts mouse ... · differentiate toward the lineage-specific cell types of the three germ layers (Lan et al., 2009; Okabayashi and Asashima,

3314

change in the expression of Ras family members (Fig. 2A),significant downregulation in the expression of the Pluri genesCeacam1 and Esrrb and the master genes Oct4 and to a lesserextent Nanog was observed in the mutated cell line, with aconcomitant decrease in OCT4 and to a lesser extent NANOGprotein (Fig. 2B,D). Under these conditions, no significantdifferences in the expression of the differentiation markers testedwere detected between WT and Dmras cells (Fig. 2C). However,we have reproducibly observed that the formation of embryoidbodies (EBs) is slower and that they are smaller in Dmras versusWT lines. Furthermore, analysis of differentiation markers at twotime points in differentiation (8-day EBs and 13 days after EBplating) shows that there is a decrease in endodermal (Sox17) andmesodermal [brachyury (T)] but not neuronal [Tuj1 (Tubb3) andMap2] expression markers in Dmras versus E14WT only in plateddifferentiated cells (data not shown).

These results suggest that the decrease in Mras expression, whichaffects the proper balance in the expression of pluripotent genes,leads to an unpoised state of the cells, which have lost, in part, theirpotential to respond to differentiation cues.

Overexpression of MRAS leads to differentcellular phenotypesMRAS overexpression or its activation was previously shown toinduce differentiation in various cell models (Kimmelman et al.,2002; Sun et al., 2006; Watanabe-Takano et al., 2010). We testedthe effect of overexpression of MRAS in mESCs grown with orwithout LIF. MRAS protein expression was detected in 30-50% ofthe transfected cells (data not shown) the day after transfection, withan average of 10-fold overexpression of the transfected gene in

transient transfection versus endogenous Mras expression(Fig. 3A,B, Fig. 4A).

Cells transfected with the EGFP-MRAS fusion protein displayspecific morphological changes compared with EGFP controlcells. Formation of membrane microspikes was observed 3 daysafter transfection and an increase in the proportion ofdifferentiated cells was observed after 8 days of selection(Fig. 3A). Although differentiated cells could be maintained formore than a month without apparent proliferation, they died afterthe first passage following trypsin treatment. In addition, after 4weeks in selective medium with LIF, stable EGFP-MRAS clones,with a slow growth rate compared with the EGFP clones, wererecovered and amplified (Fig. 3B, Fig. 4A; supplementarymaterial Fig. S1B,C).

Our data suggest that Mras, like Oct4 and Sox2, behaves as arheostat gene that displays different morphological effectsdepending upon its level of expression, which is heterogeneousfollowing transfection (Kopp et al., 2008; Niwa et al., 2000).

Stable clones overexpressing MRAS havesustained expression of OCT4 and NANOG whengrown without LIFTo determine whether overexpression of MRAS impacts the corepluripotent complex, clones expressing EGFP or EGFP-MRASwere grown with or without LIF for 7 days. Sustained expression ofOCT4 was observed in Mras-overexpressing clones grown withoutLIF (Fig. 4B; supplementary material Fig. S2). In addition, proteinexpression was analysed by flow cytometry on the EGFP-positivecell fractions. A representative analysis of four clones (EGFP-MRAS-A to -D) displays sustained expression not only of OCT4

RESEARCH ARTICLE Development 140 (16)

Fig. 2. Sustained knockdown of Mrasleads to decreased expression of Oct4and Pluri genes. (A-C) RT-qPCR expressionanalysis of (A) Mras and a selection of Rasfamily members, (B) Pluri and master genesand (C) differentiation markers in E14WT orE14Dmras mESC lines grown with or withoutLIF for 4 days. The average of threeindependent experiments is shown with s.d.*P<0.05, WT versus Dmras in the presence ofLIF; **P<0.01, WT or Dmras with LIF versuswithout LIF. (D) Immunoblot of E14WT andE14Dmras cell lysates cultured with orwithout LIF for 4 days.

DEVELO

PMENT

Page 5: MRAS GTPase is a novel stemness marker that impacts mouse ... · differentiate toward the lineage-specific cell types of the three germ layers (Lan et al., 2009; Okabayashi and Asashima,

but also of NANOG upon LIF withdrawal (Fig. 4C). This findingcould indicate that cell differentiation has been delayed upon MRASoverexpression. The ERK and PI3K signalling pathways, which areinvolved in cell differentiation and self-renewal in mESCs,respectively, have been shown to be activated by MRAS in differentsituations (Kimmelman et al., 2000; Kimmelman et al., 2002).However, we did not observe a constitutive induction of thesepathways when Mras is overexpressed (supplementary material Fig. S3).

Since MRAS-overexpressing clones grew slowly and weredifficult to maintain in culture, it was not possible to use them for amore in-depth analysis. Therefore, to explore the functions of Mrasin cell fate decision we turned to the Xenopus model, which hasbeen fruitful for establishing the functional conservation ofpluripotent genes.

Mras gene structure and protein sequence areconserved among vertebratesXenopus MRAS protein shows 84% and 92% identity with thezebrafish and human orthologues, respectively (Fig. 5A). Thisidentity is in agreement with the phylogenetic analysis of the Rasfamily, which indicates that a single Mras orthologue is present inthe genome from tunicates to mammals (Keduka et al., 2009). Mrasgene structure is remarkably conserved among vertebrates and iscomposed of five exons, interrupted by four introns with identicalsplice site junctions, having a conserved coding capacity except forthe first exon of the zebrafish gene (Fig. 5B). The intronic regionsof mammalian Mras genes have a very long first intron comparedwith those of the chicken, Xenopus and zebrafish orthologues(Fig. 5C).

Xenopus Mras expression is maternal and zygoticand restricted to the adult brain and ovarySemi-quantitative RT-PCR analysis indicates that Mras isexpressed in unfertilised Xenopus eggs and at all embryonic stagesanalysed, but that the maternal transcript level decreases afterblastula stage (stage 9) and zygotic expression starts at late neurulastage (stage 17) (Fig. 6A). In the blastula embryo, Mras mRNA isfound throughout the embryo, with no difference between animaland vegetal poles, whereas in the gastrula embryo (stage 10.5) itis mainly found at the animal pole (Fig. 6B). In adult tissues, Mrasis mainly expressed in the brain (as in mammals) and ovary(Fig. 6C).

Rohon-Beard cells and trigeminal neurons are themajor sites of expression of Mras in the XenopusembryoWe did not detect, through whole-mount in situ hybridisation, anylocalised Mras expression in the early Xenopus embryo before stage24, when a faint labelling was observed in a string of cells alongthe nerve cord corresponding to Rohon-Beard (RB) cells, which areprimary sensory neurons (Roberts, 2000) (Fig. 7A,B). Later indevelopment, labelling also appears in the trigeminal neurons andepiphysis and is clearly detected between somites on each side of theneural tube (Fig. 7C-H).

Mras regulates neuronal differentiationMras overexpression induces the neuronal differentiation of PC12mammalian cells (Kimmelman et al., 2002; Sun et al., 2006).Moreover, microarray data analysis indicates that Mras is re-expressed in ESC-derived neuronal cells (Abranches et al., 2009;

3315RESEARCH ARTICLEMras in mESCs and Xenopus embryo

Fig. 3. Overexpression of MRAS leads tomorphological changes in cells grownwith LIF. (A) mESCs transfected with EGFPor EGFP-MRAS in selective medium withLIF as indicated. (B) Selection of individualclones grown for 4 weeks. Phase contrast(left) and fluorescence (right) images areshown. Scale bar: 100 μm.

DEVELO

PMENT

Page 6: MRAS GTPase is a novel stemness marker that impacts mouse ... · differentiate toward the lineage-specific cell types of the three germ layers (Lan et al., 2009; Okabayashi and Asashima,

3316

Schulz et al., 2009) (supplementary material Fig. S4). BecauseMras is expressed in RB cells of the Xenopus embryo, weinvestigated the effects of its loss-of-function on neuronaldifferentiation during development through the use of an antisensemorpholino oligonucleotide strategy. A morpholino directed

against Mras sequence (MO Mras) and a mismatch morpholino(MO mismatch) were tested for their ability to block XenopusMras translation. Only MO Mras reduced translation of MrasmRNA (supplementary material Fig. S5). Morpholinos wereinjected into one cell of 2-cell stage embryos and morphant

RESEARCH ARTICLE Development 140 (16)

Fig. 4. Overexpression of MRAS in the absence ofLIF leads to sustained expression of OCT4 andNANOG. (A) Relative expression level of Mras asdetermined by RT-qPCR. The endogenous level ofMras in mESCs + LIF or without LIF for 24 hours ortransfected with 1 μg pCAG-EGFP (EGFP) (lanes 1-4)is compared with the level of overexpressed Mrasafter transfection with pCAG-EGFP-MRAS (lanes 5, 6)the day after transfection. The expression of Mras instable clones selected for a month in G418 incomplete medium with LIF is shown for EGFP clones(lanes 7-9) and EGFP-MRAS clones (lanes 10-12). Themean fluorescence intensity (MFI) of transfected cellsis, on average, 15,000 the day after transfection. (B) OCT4 and ERK2 western blot analysis of celllysates from stable clones expressing EGFP or EGFP-MRAS with and without LIF. (C) FACS analysis of thepercentage of cells expressing OCT4, NANOG orSOX2 among WT cells (the mean of threeexperiments is shown with s.d.) or representativeindividual clones expressing EGFP or EGFP-MRAS inthe presence or absence of LIF for 7 days. The meanMFI is 2894 for clone A, 3000 for clone B, 3000 forclone C and 3822 for clone D, with a backgroundautofluorescence of 100.

Fig. 5. Conservation of vertebrate Mras at both protein and genomic levels. (A) MRAS protein sequence comparison between dog (Cf, Canisfamiliaris), human (Hs, Homo sapiens), mouse (Mm, Mus musculus), rat (Rn, Rattus norvegicus), chicken (Gg, Gallus gallus), Xenopus (Xl, Xenopus laevis) andzebrafish (Dr, Danio rerio). Identical amino acids are black, highly conserved in blue and neutral in red. (B) Vertebrate Mras gene structure with exonsrepresented by black boxes, introns (I-IV) by a thin line and untranslated regions by white boxes. The length of the coding sequence (in nucleotides) isindicated for each exon. The type of splice junction is indicated for each intron. (C) Intron size (in nucleotides) comparison between the different Mrasgenes. D

EVELO

PMENT

Page 7: MRAS GTPase is a novel stemness marker that impacts mouse ... · differentiate toward the lineage-specific cell types of the three germ layers (Lan et al., 2009; Okabayashi and Asashima,

embryos were analysed for the expression of the neuronal markerN-tubulin, which marks zones of primary neurogenesis in theembryo. In the stage 15 Xenopus embryo, N-tubulin is expressedin three stripes of cells that correspond to primary motor, inter-and sensory neurons, respectively, the latter of which will becomeRB sensory neurons (Chitnis et al., 1995). Mras knockdown byinjection of MO Mras resulted mostly in a reduction of the sensoryneuron stripe (75%, n=126) and of the trigeminal neurons, with a

milder effect on interneurons and almost no effect on motorneurons (Fig. 8).

The reduction in N-tubulin expression is specific as neither MOmismatch nor MO standard control (GeneTools) impaired itsexpression and, moreover, the injection of a Mras mRNA that isinsensitive to MO Mras can rescue N-tubulin expression (Fig. 8).Additional markers of neuronal differentiation were alsoinvestigated: Elrc, Runx1, Pak3 and Islet1 (Perron et al., 1999;Souopgui et al., 2002; Carruthers et al., 2003; Brade et al., 2007).The expression of Elrc (72%, n=77), Pak3 (71%, n=42) and Islet1(81%, n=32) but not of Runx1 (3%, n=86) is reduced in MO Mrasmorphants (Fig. 8). Together, these experiments indicate that Mrasmight function upstream of Elrc, Pak3 or Islet1 in the regulation ofprimary neuron formation but downstream or in parallel of Runx1in the generation of RB sensory neurons.

Mras acts at a late step of neurogenesis and isrequired for neuronal differentiationThe early pan-neural marker Sox2 is essential for neuroectodermformation, while Neurogenin is the earliest proneural geneexpressed within the neural plate (Kishi et al., 2000; Ma et al.,1996). Mras depletion in the Xenopus embryo did not affect theexpression of either marker (0%, n=65 for Sox2 and n=54 forNeurogenin), indicating a late role for Mras in neuronaldifferentiation (Fig. 9).

A constitutively active mutant of Mras (G22V) has been shownto induce neuronal differentiation of PC12 cells, whereas adominant-negative form of the protein (S27N) impairedneuritogenesis (Sun et al., 2006). We tested whether these two formsof Mras had an impact on neuronal differentiation. Whereas theconstitutively active form of MRAS induced increased expressionof N-tubulin (69%, n=86), the dominant-negative form decreasedN-tubulin expression (80%, n=25). Together, these data indicate thatMras is acting at a late phase of neuronal differentiation and that itsactivation is a prerequisite to its function during differentiation.

MRAS maintains the competence of gastrulaembryo cells for FGF and activin induction andstimulates neural differentiationEmbryonic cell patterning of the early Xenopus embryo relies on alimited number of signalling pathways that influence cell fatedecisions that can be reproduced in pluripotent animal cap cells ofthe blastula embryo (Heasman, 2006). We found that FGF2stimulates Mras expression in animal cap cells, whereas activin orBMP treatments have no significant effects (Fig. 10A). Knockdown

3317RESEARCH ARTICLEMras in mESCs and Xenopus embryo

Fig. 6. Mras expression is biphasic during Xenopus laevisdevelopment. RT-PCR analysis of Mras expression. (A) Egg andembryonic stages (st). (B) Stage 9 or 10.5 dissected embryos. AP, animalpart; VP, ventral part; DM, dorsal mesoderm; VM, ventral mesoderm; EN,endoderm. An1, Vg1, Chordin (Chd), Wnt8 and Sox17 are used as controls.Total embryo (E) was analysed in parallel. (C) Adult tissues. Sk.m, skeletalmuscle. Ornithine decarboxylase (Odc) and Ribosome protein like 8 (Rpl8)were used as controls, and a reaction was performed in the absence ofreverse transcriptase (–RT).

Fig. 7. Mras is expressed in Rohon-Beard cells and trigeminal neurons in the Xenopus embryo. (A-H) In situ hybridisation with Mras antisenseprobe (A,B,E,F) or combined with immunostaining with the muscle-specific antibody 12/101 (C,D,G,H) on stage 24 (A-D) or stage 33 (E-H) embryos.Lateral (A,C,E,G) and dorsal (B,F) views. The plane of the transverse sections shown in D and H are indicated in C and G, respectively. Arrowheads indicateMras expression in Rohon-Beard cells. Ep, epiphysis; No, notochord; Nt, neural tube; RB, Rohon-Beard cells; S, somites; Tn, trigeminal neurons. D

EVELO

PMENT

Page 8: MRAS GTPase is a novel stemness marker that impacts mouse ... · differentiate toward the lineage-specific cell types of the three germ layers (Lan et al., 2009; Okabayashi and Asashima,

3318

with MO Mras did not affect mesoderm induction mediated byFGF2 (Fig. 10B) or activin (data not shown), indicating that Mrasis not essential for mesoderm induction.

The Xenopus animal cap assay has been successfully used todemonstrate the involvement of OCT4 and BRG1 in themaintenance of pluripotency (Hansis et al., 2004; Snir et al., 2006).Animal cap cells from the blastula embryo can be induced to formmesoderm when treated with FGF2 or activin, but gastrula stageembryos lose this property (Grimm and Gurdon, 2002; Snape et al.,1987). Animal cap cells from the gastrula stage embryo injectedwith Mras mRNA still express Xbra in response to FGF2(Fig. 10C). Both Xenopus and mouse Mras showed this property(data not shown). Other marker genes (Cdx4, Pnp and Esr5)previously shown to be FGF targets (Branney et al., 2009) are alsore-expressed in late animal cap cells upon Mras expression(Fig. 10C). Similarly, we tested the effect of Mras on theresponsiveness of animal caps cells to activin. When Mras mRNAis overexpressed in the embryo, late animal cap cells can stillrespond to activin treatment and express Xbra, Chordin and Xnr1

(Fig. 10D). Together, these results indicate that Mras can maintainthe mesodermal competence of pluripotent animal cap cells to FGF2and activin. This property is not limited to mesoderm but can alsoapply to endoderm, as shown by the expression of the endoderm-specific gene Sox17 (Hudson et al., 1997) (Fig. 10D).

When considering the neurectoderm germ layer, the animal captest cannot be used because both early and late animal cap cellsrespond to neurogenesis induction by the neural inducer noggin(Lamb et al., 1993). When Mras is overexpressed in animal capswith noggin, we observed a strong induction of the N-tubulin andNcam genes, indicating a positive effect of Mras on neuronaldifferentiation (supplementary material Fig. S6). Together, theseresults support a conserved role of Mras in neurogenesis anddescribe a new function of this GTPase at the onset of gastrulation.

DISCUSSIONIn this study, we have focused on Mras, which encodes a GTPasethat is preferentially expressed in undifferentiated mESCs and re-expressed in ESC-derived neuronal cells. The impact of Mras was

RESEARCH ARTICLE Development 140 (16)

Fig. 8. Inhibition of Mras blocks neuronaldifferentiation. MO Mras, MO mismatch or MOstandard were injected into one blastomere of 2-cellstage Xenopus embryos and morphants were analysedby in situ hybridisation for N-tubulin, Elrc, Runx1, Pak3and Islet1 expression. In rescue experiments, 500 pgMras mRNA was co-injected with MO Mras (MO Mras +Mras). The injected side is on the right. Motor (m) inter-(i) and sensory (s) primary neurons and the trigeminalplacode (tg) are shown (arrowhead). Arrows indicatechange in expression induced by MO Mras. Dorsalviews, anterior to bottom.

Fig. 9. Mras acts at a late step of neurogenesis andis required for neuronal differentiation. MO Mras,MO standard, or 500 pg mRNA encoding aconstitutively active form (G22V) or a dominant-negative form (S27N) of MRAS was injected into oneblastomere of 2-cell stage Xenopus embryos. Embryoswere analysed by in situ hybridisation for Sox2,Neurogenin or N-tubulin expression. The injected side ison the right. Arrows indicate change in expression.Dorsal views, anterior to bottom.

DEVELO

PMENT

Page 9: MRAS GTPase is a novel stemness marker that impacts mouse ... · differentiate toward the lineage-specific cell types of the three germ layers (Lan et al., 2009; Okabayashi and Asashima,

evaluated in two models of pluripotent cells – mESCs and Xenopusblastula embryonic cells (animal cap) – and on the development ofthe Xenopus embryo.

The involvement of Ras GTPase members in the maintenance ofcell pluripotency in mESCs was suggested by the fact that achemical inhibitor of RasGAP, which maintains Ras members in aGTP-bound active conformation, allows the propagation of mESCsin an undifferentiated pluripotent state in the absence of LIF (Chenet al., 2006). In addition, a recent report has demonstrated theimportance of the nucleolar GTP-binding protein nucleostemin(Gnl3) for the self-renewal of mESCs by direct action on the G1phase of the cell cycle (Qu and Bishop, 2012).

Functional approaches have demonstrated various roles for Rasmembers in mESCs: ERAS (ES cell-expressed ras) promotes ESCproliferation and the formation of teratomas (Takahashi et al.,2003); the activated KRAS mutant (G12V) alters the expressionof pluripotent markers, leading to growth maintenance even witha low dose of LIF (Luo et al., 2007); and activated HRAS mutantsinduce differentiation toward trophectoderm or extra-embryonicendoderm lineages (Yoshida-Koide et al., 2004; Lu et al., 2008).However, knockout mice failed to reveal any early embryonicfunction as almost all individual Ras knockouts are viable and

fertile, except for Kras−/− mice, which display lethality at E12 (Iseet al., 2000; Koera et al., 1997; Nuñez Rodriguez et al., 2006;Umanoff et al., 1995). In this study, we have not observed acompensatory effect in Ras member expression in ESC linesexpressing a half dose of Mras in the presence or absence of LIF.Further studies will be required to determine which genes, if any,might compensate for the absence of individual Ras members andto reconcile the lack of an embryonic phenotype in knockout micewith the crucial role played by some Ras members in the mESCmodel.

Our results identify a novel role for MRAS, which was thus farknown for its involvement in differentiation processes (Kimmelmanet al., 2002; Sun et al., 2006; Watanabe-Takano et al., 2010). Weshowed that downregulation of Mras expression in mESCsdeconstructs the stemness program by decreasing the expression ofOct4 and of some Pluri genes, such as Ceacam1 and Esrrb, anddiminishes the self-renewal capacity of the cells. However, theDmras cell line and cells in which Mras has been repressed byshRNA (Parisi et al., 2010) maintain their undifferentiatedmorphology and are alkaline phosphatase positive, indicating thatcompensatory effects allow cells to grow and to be amplified in LIF-containing culture medium.

We also showed that overexpression of Mras leads to changes incell morphology, with more giant and spiky differentiated cellsobserved within the first week of cell culture. However, we did notdetect any MRAS-dependent induction of the ERK pathway in theseearly differentiated cells, which did not survive after the firstpassage in culture (data not shown; supplementary material Fig.S3). Stable clones overexpressing MRAS, which do eventuallygrow in selective medium, exhibit, in contrast to the wild-typeclones, sustained expression of OCT4 and NANOG when cells aredeprived of LIF for 7 days, with no effect on SOX2 expression. Thisindicates that MRAS activates a pathway that alters only theOCT4/NANOG subcomplex and probably not the SOX2/OCT4/NANOG complex. Oct4 is the paradigm of a rheostat effector gene,which modulates the stemness/differentiation balance depending onits level of expression (Niwa et al., 2000). In addition, an OCT4dose-dependent switch from the Sox2 to Sox17 promoter triggersmesodermal differentiation (Blin et al., 2010; Stefanovic et al.,2009). Our data suggest that Mras behaves as a rheostat gene andthat there is a link between MRAS and OCT4 expression. In cellsdeprived of LIF for a week, Oct4 mRNA expression diminisheswhether or not Mras is overexpressed (our unpublished data).Therefore, we favour an MRAS-dependent post-translationalstabilisation of OCT4 that could lead to sustained expression of theprotein in Mras-overexpressing cells grown without LIF. Such post-translational modifications (phosphorylation or ubiquitylation) alterOCT4 stabilisation (Xu et al., 2009; Saxe et al., 2009). Interestingly,Mras along with Tcl1, another member of the Pluri cluster (Trouillaset al., 2009b), are direct targets of the KLF5 transcription factor,which is involved in the maintenance of cell pluripotency (Parisi etal., 2008; Parisi et al., 2010). MRAS could be a signalling relay ofKLF5, acting both on gene transcription and the protein stabilisationmachinery.

Because stable clones grew slowly, it was not possible todetermine whether they remain pluripotent in the absence of LIFusing teratoma formation. In addition, we observed a poor capacityof the E14Dmras cell line to form EBs in comparison to WT cells,and found that Brachyury (T) and Sox17 expression was decreasedin comparison to the WT situation in late differentiated cells,indicating that a proper level of Mras is required for the cells tomaintain their pluripotency and to differentiate efficiently.

3319RESEARCH ARTICLEMras in mESCs and Xenopus embryo

Fig. 10. Mras can maintain the competence of Xenopus gastrulaembryo cells for FGF and activin induction but is not required formesoderm induction. RT-PCR analysis of gene expression in Xenopusanimal caps cultured until the control embryo (lane E) reached stage 12.5.(A) Activin, BMP or FGF2 treatment followed by Mras and Xbra expressionanalysis. (B) Animal caps from Mras morphants (MO) or control embryostreated with 50 or 100 ng/ml FGF2 (FGF50 or FGF100) and analysed forXbra expression. (C,D) Animal caps dissected at stage 8 (Early) or stage 11(Late) from embryos injected with Mras or Brg1 mRNAs or uninjectedembryo (Control) were treated with FGF2 (F) or activin (A) or leftuntreated (–) before expression analysis of Xbra, Cdx4, Pnp, Esr5, Xnr,Chordin (Chd) or Sox17 (Sox). Odc was used as control, and a reaction wasperformed in the absence of reverse transcriptase (–RT).

DEVELO

PMENT

Page 10: MRAS GTPase is a novel stemness marker that impacts mouse ... · differentiate toward the lineage-specific cell types of the three germ layers (Lan et al., 2009; Okabayashi and Asashima,

3320

Therefore, we took advantage of the complementary Xenopus modelto address Mras functions in pluripotency and early development.We used both the in vivo approach and the model of pluripotentanimal cap cells (Sasai et al., 2008). Mras is highly conservedamong vertebrates from zebrafish to human at both the gene andprotein levels, suggesting a selection pressure and conservedfunction. As in mESCs, there are clearly two phases of Mrasexpression in Xenopus, with a maternal expression that decreasesbetween the blastula and early gastrula stages followed by a zygoticexpression that increases in the early tadpole, a time when MrasmRNA is found in RB cells and trigeminal neurons. RB neuronsare a transient population of primary sensory neurons that isobserved in non-mammalian vertebrates, such as zebrafish andXenopus, and is replaced in the late embryo by dorsal root ganglia(Holland, 2009; Olesnicky et al., 2010; Roberts, 2000). Strikingly,in the mouse embryo, Mras is also expressed in dorsal root ganglia(Keduka et al., 2009). We conclude that the amphibian and murineMras genes share both strong sequence conservation and expressionpattern (Kimmelman et al., 2000).

Loss-of-function of Mras in morphant embryos results in aconsistent phenotype that is characterised by the suppression ofneuronal differentiation as assessed by N-tubulin, Pak3, Elrc orIslet1 expression. This is supported by experiments in which adominant-negative form results in fewer neurons. By contrast, aconstitutively active form of MRAS stimulates neuronaldifferentiation, expanding the N-tubulin expression domain (Fig. 9).Although Mras has been shown to induce neuronal differentiation,including neurite outgrowth in PC12 cells (Sun et al., 2006), ourdata constitute the first report of the involvement of Mras duringvertebrate neurogenesis in vivo. We hypothesise that this property isnot restricted to amphibians but is shared by mammalian speciesbecause the murine protein is able to rescue morphant embryos asefficiently as the amphibian protein. Mras acts at a late phase ofneurogenesis, downstream of Sox2 and Neurogenin. Surprisingly,Runx1 expression is unchanged in Mras morphant embryos.Because Runx1 is expressed in RB progenitors and has been shownto be required for their specification independently of Neurogenin(Park et al., 2012), we hypothesise that Mras can act eitherdownstream of Runx1 in a common pathway or independently ofRunx1 in a parallel pathway.

In mESCs, a half dose of Mras leads to downregulation ofCeacam1, Esrrb, Oct4 and, to a lesser extent, Nanog, whereas itsoverexpression in LIF-deprived cells leads to sustained expressionof OCT4 and NANOG, suggesting a role in pluripotencymaintenance. We have demonstrated that gastrula cells thatoverexpress Mras can be induced to form mesoderm when treatedwith FGF2, suggesting that Mras can prolong their pluripotency, asshown for Brg1 (Smarca4) (Hansis et al., 2004). Mras is also ableto maintain gastrula cell competence to form mesoderm, but alsoendoderm, under the control of activin. An evaluation of whetherOCT4 expression is modulated in Mras morphant embryos was notpossible owing to the lack of a Xenopus OCT4 antibody. However,we have not observed any variation in Oct4 expression at themRNA level in Mras morphants (data not shown).

In conclusion, we have observed in both models two phases ofMras expression that could support two distinct roles. Mrasexpression in LIF-treated ESCs declines after LIF removal and is re-induced during neural differentiation. In Xenopus, the elevated Mrasexpression that is observed throughout the embryo until blastulastage could be required for the maintenance of pluripotency,whereas its localised expression in the presumptive neuroectodermof the gastrula embryo supports its function in neurogenesis. Indeed,

in pluripotent cells neuralised by noggin, Mras expressionstimulates neuronal differentiation. Our work, along with the recentreport on the function of the GTP-binding protein nucleostemin inthe self-renewal of mESCs (Qu and Bishop, 2012), provide newtools and insights for characterising and understanding the linksbetween GTP-binding proteins and stemness and the neurogenicregulators in different models such as mESCs and the Xenopusembryo. In addition, our study reinforces the use of complementarymodels for a better understanding of gene functions in vertebratesand demonstrates the involvement of Mras in cell pluripotency andneurogenesis.

AcknowledgementsWe thank members of our group for discussions; Sophie Daburon forproviding human LIF; Vincent Pitard and Santiago Gonzales for advice on flowcytometry; Charline Maignan for experiments performed with the Dmras cellline; Drs Sachinidis and Pradier for microarray data generated during theFunGenES European program; Drs Kolde and Vilo for the FunGenES website;Austin Smith for the CGR8 cells; Drs Endo, Bellefroid, Grainger, Pandur, Pieler,Perron and Saint Jeannet for plasmids; Dr Goding for reading the manuscript;and Lionel Parra Iglesias for Xenopus colony care. We acknowledge NIHsponsorship of NHLBI-BayGenomics and the NCRR-MMRRC for providing theESC gene trap line.

FundingThis work was funded by the European consortium FunGenES [6th Framework,project no. LSHG-CT-2003-503494]; CNRS; University Bordeaux Segalen; LigueNational contre le Cancer; comité de Gironde; Association pour la Recherchesur le Cancer [no. A08/2/1017]; Region Aquitaine; and SFR TransBiomed. M.-E.M., C.S. and M.T. were funded by the Research Ministry, Region Aquitaineand a FunGenES Fellowship, respectively.

Competing interests statementThe authors declare no competing financial interests.

Author contributionsM.-E.M., C.F., C.S., F.S., X.G. and M.T. performed experiments and contributedto data analysis. S.F. performed experiments. N.T. contributed to data analysisand wrote the manuscript. P.T. and H.B. contributed to experimental design,performed experiments, contributed to data analysis and wrote themanuscript. All authors read and approved the final manuscript forpublication.

Supplementary materialSupplementary material available online athttp://dev.biologists.org/lookup/suppl/doi:10.1242/dev.091082/-/DC1

ReferencesAbranches, E., Silva, M., Pradier, L., Schulz, H., Hummel, O., Henrique, D. and

Bekman, E. (2009). Neural differentiation of embryonic stem cells in vitro: aroad map to neurogenesis in the embryo. PLoS ONE 4, e6286.

Abu-Remaileh, M., Gerson, A., Farago, M., Nathan, G., Alkalay, I., ZinsRousso, S., Gur, M., Fainsod, A. and Bergman, Y. (2010). Oct-3/4 regulatesstem cell identity and cell fate decisions by modulating Wnt/β-cateninsignalling. EMBO J. 29, 3236-3248.

Blin, G., Nury, D., Stefanovic, S., Neri, T., Guillevic, O., Brinon, B., Bellamy, V.,Rücker-Martin, C., Barbry, P., Bel, A. et al. (2010). A purified population ofmultipotent cardiovascular progenitors derived from primate pluripotent stemcells engrafts in postmyocardial infarcted nonhuman primates. J. Clin. Invest.120, 1125-1139.

Boeuf, H., Hauss, C., Graeve, F. D., Baran, N. and Kedinger, C. (1997).Leukemia inhibitory factor-dependent transcriptional activation in embryonicstem cells. J. Cell Biol. 138, 1207-1217.

Brade, T., Gessert, S., Kühl, M. and Pandur, P. (2007). The amphibian secondheart field: Xenopus islet-1 is required for cardiovascular development. Dev.Biol. 311, 297-310.

Branney, P. A., Faas, L., Steane, S. E., Pownall, M. E. and Isaacs, H. V. (2009).Characterisation of the fibroblast growth factor dependent transcriptome inearly development. PLoS ONE 4, e4951.

Carruthers, S., Mason, J. and Papalopulu, N. (2003). Depletion of the cell-cycleinhibitor p27(Xic1) impairs neuronal differentiation and increases the numberof ElrC(+) progenitor cells in Xenopus tropicalis. Mech. Dev. 120, 607-616.

Chambers, I. and Tomlinson, S. R. (2009). The transcriptional foundation ofpluripotency. Development 136, 2311-2322.

RESEARCH ARTICLE Development 140 (16)

DEVELO

PMENT

Page 11: MRAS GTPase is a novel stemness marker that impacts mouse ... · differentiate toward the lineage-specific cell types of the three germ layers (Lan et al., 2009; Okabayashi and Asashima,

Chen, S., Do, J. T., Zhang, Q., Yao, S., Yan, F., Peters, E. C., Schöler, H. R.,Schultz, P. G. and Ding, S. (2006). Self-renewal of embryonic stem cells by asmall molecule. Proc. Natl. Acad. Sci. USA 103, 17266-17271.

Chitnis, A., Henrique, D., Lewis, J., Ish-Horowicz, D. and Kintner, C. (1995).Primary neurogenesis in Xenopus embryos regulated by a homologue of theDrosophila neurogenic gene Delta. Nature 375, 761-766.

Corpet, F. (1988). Multiple sequence alignment with hierarchical clustering.Nucleic Acids Res. 16, 10881-10890.

Díez, D., Sánchez-Jiménez, F. and Ranea, J. A. G. (2011). Evolutionaryexpansion of the Ras switch regulatory module in eukaryotes. Nucleic Acids Res.39, 5526-5537.

Duval, D., Reinhardt, B., Kedinger, C. and Boeuf, H. (2000). Role of suppressorsof cytokine signaling (Socs) in leukemia inhibitory factor (LIF) -dependentembryonic stem cell survival. FASEB J. 14, 1577-1584.

Gan, Q., Thiébaud, P., Thézé, N., Jin, L., Xu, G., Grant, P. and Owens, G. K.(2011). WD repeat-containing protein 5, a ubiquitously expressed histonemethyltransferase adaptor protein, regulates smooth muscle cell-selectivegene activation through interaction with pituitary homeobox 2. J. Biol. Chem.286, 21853-21864.

Grimm, O. H. and Gurdon, J. B. (2002). Nuclear exclusion of Smad2 is amechanism leading to loss of competence. Nat. Cell Biol. 4, 519-522.

Hansis, C., Barreto, G., Maltry, N. and Niehrs, C. (2004). Nuclearreprogramming of human somatic cells by Xenopus egg extract requiresBRG1. Curr. Biol. 14, 1475-1480.

Harland, R. M. and Grainger, R. M. (2011). Xenopus research: metamorphosedby genetics and genomics. Trends Genet. 27, 507-515.

Heasman, J. (2006). Patterning the early Xenopus embryo. Development 133,1205-1217.

Holland, L. Z. (2009). Chordate roots of the vertebrate nervous system:expanding the molecular toolkit. Nat. Rev. Neurosci. 10, 736-746.

Hudson, C., Clements, D., Friday, R. V., Stott, D. and Woodland, H. R. (1997).Xsox17alpha and -beta mediate endoderm formation in Xenopus. Cell 91, 397-405.

Ise, K., Nakamura, K., Nakao, K., Shimizu, S., Harada, H., Ichise, T., Miyoshi,J., Gondo, Y., Ishikawa, T., Aiba, A. et al. (2000). Targeted deletion of the H-ras gene decreases tumor formation in mouse skin carcinogenesis. Oncogene19, 2951-2956.

Ivanova, N., Dobrin, R., Lu, R., Kotenko, I., Levorse, J., DeCoste, C., Schafer,X., Lun, Y. and Lemischka, I. R. (2006). Dissecting self-renewal in stem cellswith RNA interference. Nature 442, 533-538.

Keduka, E., Kaiho, A., Hamada, M., Watanabe-Takano, H., Takano, K.,Ogasawara, M., Satou, Y., Satoh, N. and Endo, T. (2009). M-Ras evolvedindependently of R-Ras and its neural function is conserved betweenmammalian and ascidian, which lacks classical Ras. Gene 429, 49-58.

Kimmelman, A., Tolkacheva, T., Lorenzi, M. V., Osada, M. and Chan, A. M.(1997). Identification and characterization of R-ras3: a novel member of theRAS gene family with a non-ubiquitous pattern of tissue distribution.Oncogene 15, 2675-2685.

Kimmelman, A. C., Osada, M. and Chan, A. M. (2000). R-Ras3, a brain-specificRas-related protein, activates Akt and promotes cell survival in PC12 cells.Oncogene 19, 2014-2022.

Kimmelman, A. C., Nuñez Rodriguez, N. and Chan, A. M. (2002). R-Ras3/M-Ras induces neuronal differentiation of PC12 cells through cell-type-specificactivation of the mitogen-activated protein kinase cascade. Mol. Cell. Biol. 22,5946-5961.

Kishi, M., Mizuseki, K., Sasai, N., Yamazaki, H., Shiota, K., Nakanishi, S. andSasai, Y. (2000). Requirement of Sox2-mediated signaling for differentiation ofearly Xenopus neuroectoderm. Development 127, 791-800.

Koera, K., Nakamura, K., Nakao, K., Miyoshi, J., Toyoshima, K., Hatta, T.,Otani, H., Aiba, A. and Katsuki, M. (1997). K-ras is essential for thedevelopment of the mouse embryo. Oncogene 15, 1151-1159.

Kopp, J. L., Ormsbee, B. D., Desler, M. and Rizzino, A. (2008). Small increases inthe level of Sox2 trigger the differentiation of mouse embryonic stem cells.Stem Cells 26, 903-911.

Lamb, T. M., Knecht, A. K., Smith, W. C., Stachel, S. E., Economides, A. N.,Stahl, N., Yancopolous, G. D. and Harland, R. M. (1993). Neural induction bythe secreted polypeptide noggin. Science 262, 713-718.

Lan, L., Vitobello, A., Bertacchi, M., Cremisi, F., Vignali, R., Andreazzoli, M.,Demontis, G. C., Barsacchi, G. and Casarosa, S. (2009). Noggin elicits retinalfate in Xenopus animal cap embryonic stem cells. Stem Cells 27, 2146-2152.

Lu, C.-W., Yabuuchi, A., Chen, L., Viswanathan, S., Kim, K. and Daley, G. Q.(2008). Ras-MAPK signaling promotes trophectoderm formation fromembryonic stem cells and mouse embryos. Nat. Genet. 40, 921-926.

Luo, F., Hamoudi, R., Brooks, D. G., Patek, C. E. and Arends, M. J. (2007). Stemcell gene expression changes induced specifically by mutated K-ras. Gene Expr.14, 101-115.

Ma, Q., Kintner, C. and Anderson, D. J. (1996). Identification of neurogenin, avertebrate neuronal determination gene. Cell 87, 43-52.

Mathieu, M.-E., Saucourt, C., Mournetas, V., Gauthereau, X., Thézé, N.,Praloran, V., Thiébaud, P. and Bœuf, H. (2012). LIF-dependent signaling: newpieces in the Lego. Stem Cell Rev. 8, 1-15.

Matsumoto, K., Asano, T. and Endo, T. (1997). Novel small GTPase M-Rasparticipates in reorganization of actin cytoskeleton. Oncogene 15, 2409-2417.

Morrison, G. M. and Brickman, J. M. (2006). Conserved roles for Oct4homologues in maintaining multipotency during early vertebratedevelopment. Development 133, 2011-2022.

Naye, F., Tréguer, K., Soulet, F., Faucheux, C., Fédou, S., Thézé, N. andThiébaud, P. (2007). Differential expression of two TEF-1 (TEAD) genes duringXenopus laevis development and in response to inducing factors. Int. J. Dev.Biol. 51, 745-752.

Nieuwkoop, P. D. and Faber, J. (1967). Normal Table of Xenopus laevis (Daudin).Amsterdam: North-Holland.

Niwa, H. (2007). How is pluripotency determined and maintained? Development134, 635-646.

Niwa, H., Yamamura, K. and Miyazaki, J. (1991). Efficient selection for high-expression transfectants with a novel eukaryotic vector. Gene 108, 193-199.

Niwa, H., Miyazaki, J. and Smith, A. G. (2000). Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat.Genet. 24, 372-376.

Nuñez Rodriguez, N., Lee, I. N. L., Banno, A., Qiao, H. F., Qiao, R. F., Yao, Z.,Hoang, T., Kimmelman, A. C. and Chan, A. M.-L. (2006). Characterization ofR-ras3/m-ras null mice reveals a potential role in trophic factor signaling. Mol.Cell. Biol. 26, 7145-7154.

Okabayashi, K. and Asashima, M. (2003). Tissue generation from amphibiananimal caps. Curr. Opin. Genet. Dev. 13, 502-507.

Olesnicky, E., Hernandez-Lagunas, L. and Artinger, K. B. (2010). prdm1aRegulates sox10 and islet1 in the development of neural crest and Rohon-Beard sensory neurons. Genesis 48, 656-666.

Parisi, S. and Russo, T. (2011). Regulatory role of Klf5 in early mousedevelopment and in embryonic stem cells. Vitam. Horm. 87, 381-397.

Parisi, S., Passaro, F., Aloia, L., Manabe, I., Nagai, R., Pastore, L. and Russo, T.(2008). Klf5 is involved in self-renewal of mouse embryonic stem cells. J. CellSci. 121, 2629-2634.

Parisi, S., Cozzuto, L., Tarantino, C., Passaro, F., Ciriello, S., Aloia, L., Antonini,D., De Simone, V., Pastore, L. and Russo, T. (2010). Direct targets of Klf5transcription factor contribute to the maintenance of mouse embryonic stemcell undifferentiated state. BMC Biol. 8, 128.

Park, B.-Y., Hong, C.-S., Weaver, J. R., Rosocha, E. M. and Saint-Jeannet, J.-P.(2012). Xaml1/Runx1 is required for the specification of Rohon-Beard sensoryneurons in Xenopus. Dev. Biol. 362, 65-75.

Perron, M., Furrer, M. P., Wegnez, M. and Théodore, L. (1999). Xenopus elav-like genes are differentially expressed during neurogenesis. Mech. Dev. 84, 139-142.

Qu, J. and Bishop, J. M. (2012). Nucleostemin maintains self-renewal ofembryonic stem cells and promotes reprogramming of somatic cells topluripotency. J. Cell Biol. 197, 731-745.

Roberts, A. (2000). Early functional organization of spinal neurons in developinglower vertebrates. Brain Res. Bull. 53, 585-593.

Saito, Y., Oinuma, I., Fujimoto, S. and Negishi, M. (2009). Plexin-B1 is a GTPaseactivating protein for M-Ras, remodelling dendrite morphology. EMBO Rep. 10,614-621.

Sasai, Y., Ogushi, M., Nagase, T. and Ando, S. (2008). Bridging the gap from frog research to human therapy: a tale of neural differentiation in Xenopus animalcaps and human pluripotent cells. Dev. Growth Differ. 50 Suppl. 1, S47-S55.

Saxe, J. P., Tomilin, A., Schöler, H. R., Plath, K. and Huang, J. (2009). Post-translational regulation of Oct4 transcriptional activity. PLoS ONE 4, e4467.

Schulz, H., Kolde, R., Adler, P., Aksoy, I., Anastassiadis, K., Bader, M., Billon,N., Boeuf, H., Bourillot, P.-Y., Buchholz, F. et al.; Functional Genomics inEmbryonic Stem Cells Consortium (2009). The FunGenES database: agenomics resource for mouse embryonic stem cell differentiation. PLoS ONE 4,e6804.

Sive, H. L., Grainger, R. M. and Harland, R. M. (2000). Early Development ofXenopus Laevis. A Laboratory Manual. Cold Spring Harbor, NY: Cold SpringHarbor Laboratory Press.

Snape, A., Wylie, C. C., Smith, J. C. and Heasman, J. (1987). Changes in statesof commitment of single animal pole blastomeres of Xenopus laevis. Dev. Biol.119, 503-510.

Snir, M., Ofir, R., Elias, S. and Frank, D. (2006). Xenopus laevis POU91 protein,an Oct3/4 homologue, regulates competence transitions from mesoderm toneural cell fates. EMBO J. 25, 3664-3674.

Souopgui, J., Sölter, M. and Pieler, T. (2002). XPak3 promotes cell cyclewithdrawal during primary neurogenesis in Xenopus laevis. EMBO J. 21, 6429-6439.

Stefanovic, S., Abboud, N., Désilets, S., Nury, D., Cowan, C. and Pucéat, M.(2009). Interplay of Oct4 with Sox2 and Sox17: a molecular switch from stemcell pluripotency to specifying a cardiac fate. J. Cell Biol. 186, 665-673.

3321RESEARCH ARTICLEMras in mESCs and Xenopus embryo

DEVELO

PMENT

Page 12: MRAS GTPase is a novel stemness marker that impacts mouse ... · differentiate toward the lineage-specific cell types of the three germ layers (Lan et al., 2009; Okabayashi and Asashima,

3322

Sun, P., Watanabe, H., Takano, K., Yokoyama, T., Fujisawa, J. and Endo, T.(2006). Sustained activation of M-Ras induced by nerve growth factor isessential for neuronal differentiation of PC12 cells. Genes Cells 11, 1097-1113.

Takahashi, K., Mitsui, K. and Yamanaka, S. (2003). Role of ERas in promotingtumour-like properties in mouse embryonic stem cells. Nature 423, 541-545.

Trouillas, M., Saucourt, C., Guillotin, B., Gauthereau, X., Taupin, J. L.,Moreau, J. F. and Boeuf, H. (2009a). The LIF cytokine: towards adulthood. Eur.Cytokine Netw. 20, 51-62.

Trouillas, M., Saucourt, C., Guillotin, B., Gauthereau, X., Ding, L., Buchholz,F., Doss, M. X., Sachinidis, A., Hescheler, J., Hummel, O. et al. (2009b).Three LIF-dependent signatures and gene clusters with atypical expressionprofiles, identified by transcriptome studies in mouse ES cells and earlyderivatives. BMC Genomics 10, 73.

Umanoff, H., Edelmann, W., Pellicer, A. and Kucherlapati, R. (1995). Themurine N-ras gene is not essential for growth and development. Proc. Natl.Acad. Sci. USA 92, 1709-1713.

Ward, K. R., Zhang, K.-X., Somasiri, A. M., Roskelley, C. D. and Schrader, J. W.(2004). Expression of activated M-Ras in a murine mammary epithelial cell lineinduces epithelial-mesenchymal transition and tumorigenesis. Oncogene 23,1187-1196.

Watanabe-Takano, H., Takano, K., Keduka, E. and Endo, T. (2010). M-Ras isactivated by bone morphogenetic protein-2 and participates in osteoblasticdetermination, differentiation, and transdifferentiation. Exp. Cell Res. 316, 477-490.

Wray, J., Kalkan, T. and Smith, A. G. (2010). The ground state of pluripotency.Biochem. Soc. Trans. 38, 1027-1032.

Xu, H., Wang, W., Li, C., Yu, H., Yang, A., Wang, B. and Jin, Y. (2009). WWP2promotes degradation of transcription factor OCT4 in human embryonic stemcells. Cell Res. 19, 561-573.

Yoshida-Koide, U., Matsuda, T., Saikawa, K., Nakanuma, Y., Yokota, T.,Asashima, M. and Koide, H. (2004). Involvement of Ras in extraembryonicendoderm differentiation of embryonic stem cells. Biochem. Biophys. Res.Commun. 313, 475-481.

RESEARCH ARTICLE Development 140 (16)

DEVELO

PMENT