mammalian tafii30 is required for cell cycle progression

12
The EMBO Journal Vol.18 No.17 pp.4823–4834, 1999 Mammalian TAF II 30 is required for cell cycle progression and specific cellular differentiation programmes Daniel Metzger, Elisabeth Scheer, Aleksey Soldatov 1 and La ` szlo ` Tora 2 Institut de Ge ´ne ´tique et de Biologie Mole ´culaire et Cellulaire, CNRS/INSERM/ULP, Colle `ge de France, BP 163-67404 Illkirch Cedex, CU de Strasbourg, France and 1 Institute of Gene Biology, Russian Academy of Sciences, Vavilov st. 26, Moscow 117 334, Russia 2 Corresponding author e-mail: [email protected] The two alleles of the 30 kDa TATA-binding protein associated factor (TAF II 30) gene, have been targeted by homologous recombination in murine F9 embryonal carcinoma cells and subsequently disrupted using a Cre recombinase–loxP strategy. The TAF II 30-null cells are not viable, but are rescued by the expression of human TAF II 30. Cells lacking TAF II 30 are blocked in G 1 /G 0 phase of the cell cycle and undergo apoptosis. In agreement with the G 1 arrest phenotype, the expres- sion of cyclin E is impaired and the retinoblastoma protein is hypophosphorylated in the TAF II 30-null cells. Interestingly, retinoic acid (RA) treatment prevented TAF II 30-null cell death and induced primitive endo- dermal differentiation. In contrast, the RA- and cAMP- induced parietal endodermal differentiation was impaired in the TAF II 30-null cells. Thus, TAF II 30 is not indispensable for class II gene transcription in general, but seems to be required for the expression of a subset of genes. Keywords: apoptosis/conditional gene knock-out/Cre recombinase/proliferation/TFIID Introduction Correct initiation of transcription of protein-encoding genes by RNA polymerase II (Pol II) requires the transcrip- tion factor TFIID, which is comprised of TATA-binding protein (TBP) and a number of TBP-associated factors (TAF II s) (Tansey and Herr, 1997). The exact role of TAF II s in transcription initiation is unknown. Initial in vitro transcription studies suggested that TAF II s may work as general co-activators to mediate the transcriptional activation of the different activators (Goodrich et al., 1996). It was also shown that recombinant TBP can substitute in vitro for TFIID in reconstituted transcription systems, but does not support transcription from TATA- less promoters (Pugh and Tjian, 1991; Zhou et al., 1992). Evidence that TAF II s function as co-activators in mam- malian cells has been provided by studies showing that hTAF II 28 and hTAF II 135 enhance transcriptional activation by nuclear receptors (May et al., 1996; Mengus et al., 1997). Further evidence that TAF II s are required for transcriptional activation came from genetic studies of © European Molecular Biology Organization 4823 D.melanogaster which showed that mutations in dTAF II s lead to specific defects in transcriptional activation in the embryo (Zhou et al., 1998). In yeast (y) strains lacking functional yTAF II s, a variety of inducible genes are normally transcribed in vivo (Apone et al., 1996; Moqtaderi et al., 1996a; Walker et al., 1996). Nevertheless, TAF II s do play an essential role in yeast, as mutant strains are not viable (Moqtaderi et al., 1996b). Thus, TAF II s may not generally be required for activator- dependent transcription, but seem to be essential for the transcription of certain genes. Functionally distinct TFIID populations, sharing com- mon TAF II s but differing in their specific TAF II composi- tion, have been isolated from mammalian cells (Bell and Tora, 1999). TFIID complexes that either contain (TFIIDβ) or lack (TFIIDα) hTAF II 30 have been separated from mammalian cells extracts (Jacq et al., 1994; Mengus et al., 1995). Human TAF II 30 is present in only ~50% of TFIID complexes and is therefore considered a ‘specific’ TAF II (Jacq et al., 1994). Recently, a novel multiprotein complex containing neither TBP nor TBP-like factor (TLF), but comprising hTAF II 30 and several other TAF II s, has been characterized (Wieczorek et al., 1998). This complex, called TBP-free TAF II -containing complex (TFTC), can replace TFIID in in vitro transcription assays (Wieczorek et al., 1998) and contains histone acetyl transferase (HAT) activity (Brand et al., 1999). Moreover, TAF II 30 and its yeast homologue yTAF II 25 are present in several other TAF II –HAT-containing complexes, the yeast SAGA and the human PCAF/GCN5 complexes (Grant et al., 1998; Ogryzko et al., 1998). To understand the functions of mammalian TAF II s in the regulation of Pol II transcription in vivo, and more specifically that of TAF II 30, we have inactivated both alleles of the TAF II 30 gene in murine embryonal carcinoma F9 cells. The F9 cell line provides an interesting model system to analyse the role of a given transcription factor during either cell proliferation and/or cell differentiation. Treatment of F9 cells with all-trans retinoic acid (RA) induces primitive endodermal differentiation (Strickland and Mahdavi, 1978), whereas treatment with RA and dibutyryl c-AMP (bt 2 cAMP) induces a parietal endodermal phenotype (Strickland et al., 1980). Moreover, the RA- induced differentiation of F9 cells is accompanied by a dramatic decrease in the proliferation rate, an increase in the fraction of cells arrested in G 1 /G 0 phase of the cell cycle and an increase in the number of apoptotic cells (Clifford et al., 1996, and references therein). The RA- induced differentiation is mediated by the RA receptors (RARs) and the retinoic X receptors (RXRs) (Chiba et al., 1997a, and references therein). The two TAF II 30 alleles have been targeted here by homologous recombination (HR) and subsequently disrupted by using a Cre recombin- ase–loxP strategy (Sauer and Henderson, 1990). F9 cells

Upload: others

Post on 13-Nov-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Mammalian TAFII30 is required for cell cycle progression

The EMBO Journal Vol.18 No.17 pp.4823–4834, 1999

Mammalian TAFII30 is required for cell cycleprogression and specific cellular differentiationprogrammes

Daniel Metzger, Elisabeth Scheer,Aleksey Soldatov1 and Laszlo Tora2

Institut de Ge´netique et de Biologie Mole´culaire et Cellulaire,CNRS/INSERM/ULP, Colle`ge de France, BP 163-67404 IllkirchCedex, CU de Strasbourg, France and1Institute of Gene Biology,Russian Academy of Sciences, Vavilov st. 26, Moscow 117 334,Russia2Corresponding authore-mail: [email protected]

The two alleles of the 30 kDaTATA-binding proteinassociated factor (TAFII 30) gene, have been targetedby homologous recombination in murine F9 embryonalcarcinoma cells and subsequently disrupted using aCre recombinase–loxP strategy. The TAFII 30-null cellsare not viable, but are rescued by the expression ofhuman TAFII 30. Cells lacking TAFII 30 are blocked inG1/G0 phase of the cell cycle and undergo apoptosis.In agreement with the G1 arrest phenotype, the expres-sion of cyclin E is impaired and the retinoblastomaprotein is hypophosphorylated in the TAFII 30-null cells.Interestingly, retinoic acid (RA) treatment preventedTAFII 30-null cell death and induced primitive endo-dermal differentiation. In contrast, the RA- and cAMP-induced parietal endodermal differentiation wasimpaired in the TAF II 30-null cells. Thus, TAFII 30 isnot indispensable for class II gene transcription ingeneral, but seems to be required for the expressionof a subset of genes.Keywords: apoptosis/conditional gene knock-out/Crerecombinase/proliferation/TFIID

Introduction

Correct initiation of transcription of protein-encodinggenes by RNA polymerase II (Pol II) requires the transcrip-tion factor TFIID, which is comprised of TATA-bindingprotein (TBP) and a number of TBP-associated factors(TAFIIs) (Tansey and Herr, 1997). The exact role of TAFIIsin transcription initiation is unknown. Initialin vitrotranscription studies suggested that TAFIIs may workas general co-activators to mediate the transcriptionalactivation of the different activators (Goodrichet al.,1996). It was also shown that recombinant TBP cansubstitutein vitro for TFIID in reconstituted transcriptionsystems, but does not support transcription from TATA-less promoters (Pugh and Tjian, 1991; Zhouet al., 1992).

Evidence that TAFIIs function as co-activators in mam-malian cells has been provided by studies showing thathTAFII28 and hTAFII135 enhance transcriptional activationby nuclear receptors (Mayet al., 1996; Menguset al.,1997). Further evidence that TAFIIs are required fortranscriptional activation came from genetic studies of

© European Molecular Biology Organization 4823

D.melanogasterwhich showed that mutations in dTAFIIslead to specific defects in transcriptional activation in theembryo (Zhouet al., 1998).

In yeast (y) strains lacking functional yTAFIIs, a varietyof inducible genes are normally transcribedin vivo (Aponeet al., 1996; Moqtaderiet al., 1996a; Walkeret al., 1996).Nevertheless, TAFIIs do play an essential role in yeast, asmutant strains are not viable (Moqtaderiet al., 1996b).Thus, TAFIIs may not generally be required for activator-dependent transcription, but seem to be essential for thetranscription of certain genes.

Functionally distinct TFIID populations, sharing com-mon TAFIIs but differing in their specific TAFII composi-tion, have been isolated from mammalian cells (Bell andTora, 1999). TFIID complexes that either contain (TFIIDβ)or lack (TFIIDα) hTAFII30 have been separated frommammalian cells extracts (Jacqet al., 1994; Menguset al.,1995). Human TAFII30 is present in only ~50% of TFIIDcomplexes and is therefore considered a ‘specific’ TAFII(Jacqet al., 1994). Recently, a novel multiprotein complexcontaining neither TBP nor TBP-like factor (TLF), butcomprising hTAFII30 and several other TAFIIs, has beencharacterized (Wieczoreket al., 1998). This complex,called TBP-free TAFII-containing complex (TFTC), canreplace TFIID inin vitro transcription assays (Wieczoreket al., 1998) and contains histone acetyl transferase (HAT)activity (Brandet al., 1999). Moreover, TAFII30 and itsyeast homologue yTAFII25 are present in several otherTAFII–HAT-containing complexes, the yeast SAGA andthe human PCAF/GCN5 complexes (Grantet al., 1998;Ogryzkoet al., 1998).

To understand the functions of mammalian TAFIIs inthe regulation of Pol II transcriptionin vivo, and morespecifically that of TAFII30, we have inactivated bothalleles of theTAFII30gene in murine embryonal carcinomaF9 cells. The F9 cell line provides an interesting modelsystem to analyse the role of a given transcription factorduring either cell proliferation and/or cell differentiation.Treatment of F9 cells with all-trans retinoic acid (RA)induces primitive endodermal differentiation (Stricklandand Mahdavi, 1978), whereas treatment with RA anddibutyryl c-AMP (bt2cAMP) induces a parietal endodermalphenotype (Stricklandet al., 1980). Moreover, the RA-induced differentiation of F9 cells is accompanied by adramatic decrease in the proliferation rate, an increase inthe fraction of cells arrested in G1/G0 phase of the cellcycle and an increase in the number of apoptotic cells(Clifford et al., 1996, and references therein). The RA-induced differentiation is mediated by the RA receptors(RARs) and the retinoic X receptors (RXRs) (Chibaet al.,1997a, and references therein). The twoTAFII30 alleleshave been targeted here by homologous recombination(HR) and subsequently disrupted by using a Cre recombin-ase–loxP strategy (Sauer and Henderson, 1990). F9 cells

Page 2: Mammalian TAFII30 is required for cell cycle progression

D.Metzger et al.

with both TAFII30 alleles inactivated are not viable,but can be rescued by expression of human TAFII30.Furthermore, we present evidence that TAFII30 is requiredfor cell cycle progression through G1 phase. TAFII30-nullcells undergo massive apoptosis which is prevented byRA-induced differentiation. Interestingly, parietal, but notprimitive endodermal differentiation is strongly impairedin the TAFII30-null cells. Taken together, our resultsindicate that TAFII30 plays an important and selective rolein establishing gene expression patterns.

Results

Conditional targeting of the TAFII30 gene in F9cellsTo inactivate theTAFII30 gene by HR, we first clonedand characterized the mouse gene (Figure 1). The structureof the mouse (m) TAFII30 gene is very similar to thehuman TAFII30 gene (TAF2H; Scheeret al., 1995), i.e. itconsists of five exons (Figure 1). ThemTAFII30 codingsequence deduced from the genomic sequence is 759bases long (89% identical to the human cDNA) andencodes a 218 amino acid protein that is 90.3% identicalto the human TAFII30 protein.

To perform a conditional disruption of themTAFII30gene in F9 cells, we used a Cre–loxP strategy, similar tothat previously described (Sumi-Ichinoseet al., 1997). Weconstructed two targeting vectors (Figure 1B and C), inwhich a loxP site (hereafter termed L) was inserted intothe intron located upstream of exon 2, while a neomycin-resistance gene expression cassette (tk-neo, hereaftertermed N) flanked by two loxP sites (‘floxed’) was insertedinto the intron located downstream of exon 2. Thus,following targeting of the wild-type (WT) alleles by HR,the Cre recombinase will allow the excision of exon 2 aswell as the floxed selection marker cassette, resulting inthe deletion of the sequences encoding amino acid residues78 (alanine) to 130 (isoleucine) and the creation of aframe shift in the beginning of exon 3 with a stop codon(Figure 1 and data not shown). Thus, the putative truncatedprotein produced from the disrupted gene will lack theevolutionary highly conserved C-terminal domain ofTAFII30.

To target the first allele of theTAFII30 gene, F9cells were electroporated with the pTAFII30(L:LNL)a vector(Figure 1B). Southern blotting and genomic PCR analysesrevealed clones that had one targetedTAFII30 allele[TAFII30(L:LNL)a/1; see Figures 1B, 2A and C]. As thepresence of the marker gene in the targetedmTAFII30allele might affect its expression, we established a cellline in which the floxed neomycin-resistance gene wasexcised before targeting the second allele. To this end,TAFII30(L:LNL)a/1 cells were electroporated with the Crerecombinase expression vector. Clones were obtained inwhich either exon 2 and the selection marker cassette(TAFII30–La/1 cells) or only the selection marker cassette[TAFII30(L:L)a/1 cells] were deleted (see Figure 2A andC). Cells from one of the TAFII30(L:L)a/1 clones were thenelectroporated with the second targeting vector (Figure 1C)and cells containing the second targetedTAFII30 allelewere identified (Figures 1, 2B and C). Note that in thesecond targeting vector, the three loxP sites were in the

4824

opposite orientation when compared with the previouslyused vector.

To excise the sequences located between the loxP sites,the TAFII30(L:L)a/(L:LNL)b cells (Figure 2B) were electropor-ated with the Cre recombinase expression vector. Noclones with both alleles inactivated [TAFII30–La/–Lb orTAFII30–La/–( LNL)b] were obtained out of 560 analysedclones, but 30 clones had one inactivatedTAFII30 allele(Figure 2B). Amongst these clones, all the possible differ-ent genotypes for the twoTAFII30 alleles were present(Figure 2B and C and data not shown). Thus, it appearsthat either one of the two alleles of the mouseTAFII30gene can be independently inactivated, but that inactivationof both TAFII30 alleles may be lethal.

Inactivation of both TAFII30 alleles impairs F9 cellviabilityTo investigate whether the inactivation of bothTAFII30 alleles impairs the proliferation of F9 cells,TAFII30–La/( L:L)b cells were electroporated with the Crerecombinase expression vector and the cell populationwas genotyped by PCR using DNA extracted over an11 day period (Figure 3). Southern blotting was used forthe detection of the (L:L)b and –Lb alleles (Figure 3B).The intensity of the 706 bp signal corresponding to the(L:L)b allele was similar in all the samples analysed. Incontrast, although the 233 bp signal corresponding to the –Lb allele could be detected 1 day after Cre recombinaseelectroporation, it decreased at day four, and finallydisappeared totally between days five and seven (Figure 3Band data not shown). Thus, F9 cells with both TAFII30alleles inactivated were obtained, but were apparentlyseverely affected in their proliferation and could notbe cloned.

Conditional rescue of TAFII30–La/–Lb cells withhuman TAFII30To demonstrate unequivocally that the failure to obtain anF9 cell line in which bothmTAFII30alleles are inactivated,was due to an absolute requirement of the TAFII30 proteinfor cell proliferation, and to allow further studies ofTAFII30 function, we generated a Dox-inducible TAFII30expression system. We stably integrated an expressioncassette encoding the reverse tetracycline-controlled trans-activator (rtTA; Gossenet al., 1995) in the genome of theTAFII30–La/( L:L)b cells, as well as another cassette inwhich the humanTAFII30 cDNA is under the control oftetracycline-operators, resulting in the TAFII30–La/( L:L)b:Rcell line. The inducibility of hTAFII30 protein synthesisby the tetracycline analogue Dox was verified by Westernblotting analysis using the anti-hTAFII30 monoclonal anti-body 2F4 (Jacqet al., 1994), which is specific for thehuman protein. Whereas no hTAFII30 protein could bedetected in TAFII30–La/( L:L)b:R F9 cells grown in theabsence of Dox, the amount of hTAFII30 protein afterDox treatment of these cells was comparable to that ofmTAFII30 in WT F9 cells (Figure 3C). To inactivate bothmTAFII30alleles, TAFII30–La/( L:L)b:R cells were electropor-ated with the Cre recombinase expression vector andcultured in the presence of Dox. Under these conditionswe obtained several clones in which exon 2 was deletedon both alleles, as verified by Southern blotting andgenomic PCR analyses (TAFII30–La/Lb:R; Figure 3D). Inter-

Page 3: Mammalian TAFII30 is required for cell cycle progression

Conditional TAFII30 gene knock-out

Fig. 1. Strategy for targeting the mouseTAFII30 gene. (A) Structure of the mouseTAFII30 gene (mTAFII301 allele). Exons 1 to 5, and the 59 probecorresponding to a 0.8 kbEcoRV–KpnI fragment are indicated. (B) First allele targeting. The targeting vector pTAFII30(L:LNL)a is represented. TheloxP sites (open triangles) and the tk-Neo cassette (tk-Neo) are indicated. The expected genomic maps after HR [TAFII30(L:LNL)a allele] andCre-mediated excision of loxP-flanked DNA segments [TAFII30(L:L)a and TAFII30–La alleles] are shown. (C) Second allele targeting. The targetingvector pTAFII30(L:LNL)b is represented. The loxP sites (open triangles) and the tk-Neo cassette, in opposite orientations relative to those in thepTAFII30(L:LNL)a vector, are indicated. The expected genomic maps after HR [TAFII30(L:LNL)b allele] and Cre-mediated excision of loxP-flankedDNA segments [TAFII30(L:L)b and TAFII30–Lb alleles] are shown. The horizontal arrowheads indicate the location of the primers used for PCRs (seeFigures 2C and 3D). DNA fragments obtained afterSacI-digestion and detection with the 59 probe (see Figures 2C and 3D) are also indicated:B, BamHI; S, SacI; X, XhoI; and E,EcoRI (E is only indicated in the mTAFII301 allele). The solid line in the targeting vectors corresponds to theDNA fragment used for gene targeting. Dotted lines represent plasmid sequences.

estingly, in the TAFII30–La/–Lb:R cells neither a truncatedmTAFII30mRNA nor a truncated protein could be detectedby Northern and Western blotting analyses, respectively(Figure 3D). Therefore, the –La and –Lb alleles are truenull alleles.

To investigate the role of TAFII30 in cell proliferation,we cultured the TAFII30–La/–Lb:R cells in the absence andpresence of Dox. One day after Dox withdrawal, thehTAFII30 protein level decreased by ~50% and becameundetectable from the second day after Dox withdrawal(Figure 4A), whereas in the presence of Dox the hTAFII30levels were constant over several weeks (Figure 4A anddata not shown). The growth rates of TAFII30–La/–Lb:R

4825

cells grown either in the presence or absence of Dox weresimilar for the first 2 days, but in the absence of Dox thecells became arrested after 3 days, and almost all the cellswere dead after 6 days. Whereas cells grew to confluencyin the presence of Dox [Figures 4B and 7A (b) and (c)].Taken together, these results demonstrate that theTAFII30gene product is indispensable for the viability of un-differentiated F9 cells.

Interestingly, even 5 days after Dox withdrawal, theprotein levels of other TFIID and TFTC components (suchas mTBP and mTAFII135, mTAFII100, mTAFII80 andmTAFII55) were not affected in TAFII30-null cells(Figure 4A and data not shown).

Page 4: Mammalian TAFII30 is required for cell cycle progression

D.Metzger et al.

Fig. 2. Nomenclature and characterization of the genotypes of the different F9 cell lines targeted in theTAFII30 locus. Schematic representation ofthe different TAFII30 mutant cell lines obtained after targeting the first (A) and the second (B) alleles and after Cre-mediated recombination. Theexons are represented as boxes (1–3) and the loxP sites in orientation a (La) or b (Lb) as triangles. The genotypes of cells indicated by dotted arrowswere not obtained. (C) Southern blot ofSacI-digested genomic DNA from the different cell lines (as indicated) hybridized with the 59 probe (seeFigure 1). Lane 2,1/1 (14.8 kb); lane 3, (L:LNL)a/1 (4.9 and 14.8 kb); lane 4, (L:L)a/1 (4.9 and 14.8 kb); lane 5, –La/1 (14.4 and 14.8 kb);lane 6, (L:L)a/(L:LNL)b (4.9 and 6 kb); lane 7, –La/(L:LNL)b (14.4 and 6 kb); lane 8, (L:L)a/–Lb (4.9 and 4.5 kb); and lane 9, –La/(L:L)b (14.4 and4.5 kb). Genomic DNA extracted from the indicated cell lines was analysed by PCR using the primers indicated in Figure 1B. The SG12–SE89primer pair amplifies an 823 bp fragment from the WT and the (L:LNL)a/1 alleles (lanes 2 and 3), 945 and 823 bp fragments from the (L:L)a/1alleles (lane 4), 823 and 478 bp fragments from the –La/1 alleles (lane 5), 945 bp fragment from the (L:L)a/(L:LNL)b alleles (lane 6), 478 bpfragment from –La/(L:LNL)b alleles (lane 7), 472 and 945 bp fragments from the (L:L)a/–Lb alleles (lane 8), and 478 and 945 bp fragments fromthe –La/(L:L)b alleles (lane 9), respectively. The * indicates a non specific band. The 1 kb ladder DNA (M; Gibco-BRL) is shown in lane 1.

TAFII30 is required for G1–S phase cell cycleprogressionThe cell cycle profile of the TAFII30–La/–Lb:R cells grownin the absence or presence of Dox was investigated byflow cytometry. In agreement with the proliferation ratestudies (Figure 4B), no significant differences in the cellcycle profiles were seen between TAFII30–La/–Lb:R cellsgrown for 2 days in the absence or presence of Dox(Figure 4C). In contrast, 5 days after Dox withdrawalthe TAFII30-null cells accumulated in G1/G0 phase. Theproportion of cells in G1/G0 phase increased from ~39 to61% (Figure 4C).

We then investigated the mRNA level of different cellcycle-related cyclin genes in TAFII30-null cells. Reversetranscription–polymerase chain reaction (RT–PCR) ana-lysis was used to obtain a semi-quantitative estimation ofthe expression of different genes (see Materials andmethods). Cyclin E expression was progressively reducedfrom the third to the fifth day after Dox withdrawal (~6.6-fold reduction at the fifth day), when compared with eitherthe expression in cells grown in the presence of Dox or

4826

WT cells (Figure 5A and B). In contrast, the expressionof the other tested cyclins was either not, or was onlyweakly altered after the suppression of hTAFII30 synthesis(Figure 5A and B). Cyclin E, which is expressed atmaximal levels in the G1–S transition and interacts with itscatalytic partnercdk2, is a key regulator of G1 progressionduring the cell cycle (Sherr, 1994; Reed, 1997). The strongreduction of cyclin E expression in TAFII30-null cells isin good agreement with the results obtained by flowcytometry indicating that TAFII30-null cells accumulatein G1 phase.

Phosphorylation of retinoblastoma protein (pRb) in G1inactivates its growth inhibitory function, allowing cellcycle progression (Barteket al., 1997). As cyclin E–cdk2complex collaborates with the D-type cyclin–cdk4/6complexes for pRb phosphorylation (Hatakeyamaet al.,1994; Lundberg and Weinberg, 1998), we investigated thephosphorylation state of pRb in the TAFII30-null cells.In WT and TAFII30–La/–Lb:R cells expressing hTAFII30protein, ~70% of pRb was phosphorylated (Figure 5C,lanes 1 and 2). In contrast, the level of pRb phosphorylation

Page 5: Mammalian TAFII30 is required for cell cycle progression

Conditional TAFII30 gene knock-out

Fig. 3. TAFII30–La/–Lb cells are not viable, but can be rescued by human TAFII30. (A) The PCR strategy to identify the different alleles is outlined.Exons are indicated as numbered boxes. LoxP sites in orientation a (La) are indicated as black triangles. The oligonucleotides used either as PCRprimers (SG12 and WY60; see Materials and methods) or as a probe WY59 (59-GGAGGGCATCGTCGACATAA-39) are represented by arrows. TheloxP site in orientation b (Lb) with its surrounding sequences, and recognized by the PCR primer WY60, is shown as black triangles fused to anopen box. The length of the amplified fragments from the different alleles is given. (B) Detection of TAFII30–La/–Lb cells by PCR. TAFII30–La/(L:L)b

cells were electroporated with the pSG–Cre expression vector, plated at a density of 2.53103 cells/well in 6-well plates and collected from days zeroto 10 and stored at –80°C. Detection of the TAFII30(L/L)b and TAFII30–Lb alleles was performed according to the Materials and methods, as depictedin (A). (C) Dox-induced expression of hTAFII30 in TAFII30–La/(L:L)b:R cells. Cell extracts were prepared from cells grown in the presence or absenceof Dox, as well as from HeLa and F9 cells, as indicated. Twenty-five microgrammes of each protein extract was loaded per lane and analysed byWestern blotting using the anti-hTAFII30 monoclonal antibody 2F4 (Jacqet al., 1994), which is specific for the human protein. (D) Characterizationby Southern, Northern and Western blotting and PCR amplification of TAFII30 –La/–Lb:R cells. Southern blotting was performed onSacI-digestedgenomic DNA isolated from the different cell lines (as indicated), and hybridized with the 59 probe (see Figure 1). Lane 1, WT (14.8 kb); lane 2,–La/(L:L)b (14.4 and 4.5 kb); and lane 3, –La/–Lb (14.4 and 4.5 kb). Genomic DNA extracted from the indicated cell lines was analysed by PCRusing the primers indicated in Figure 1B. The SG12–SE89 primer pair amplifies an 823 bp fragment from the WT alleles (lane 2), 945 and 472 bpfragments from the –La/(L:L)b alleles (lane 3), and 472 and 478 bp fragments from the –La/–Lb alleles (lane 4), respectively. The asterisk indicates anon specific band. The 1 kb ladder DNA (M; Gibco-BRL) is shown in lane 1. The Northern blotting was carried out by standard methods usingcytoplasmic RNA isolated from the indicated cell lines and hybridized with a mTAFII30 cDNA probe. The Western blotting was carried out withprotein extracts isolated from the indicated cell lines using the 1H8 anti-TAFII30 mAb, directed against the N-terminal end of TAFII30 (Wieczoreket al., 1998). The arrowheads on the Northern and Western blots indicate the sizes of the putative truncated mTAFII30 RNA and protein,respectively.

decreased after stopping hTAFII30 expression, and after5 days of culture in the absence of Dox, pRb wasessentially unphosphorylated (Figure 5C, lane 6). Notethat the expression of pRb mRNA was not affected in theTAFII30-null cells (data not shown). Taken together, theabove results show that TAFII30 is required for G1–Sphase cell cycle progression and cyclin E expression.

TAFII30-null F9 cells undergo apoptosisWhen the TAFII30–La/–Lb:R cells were grown for 5 daysin the absence of Dox, ~82% of the cells had a sub-2Nsize DNA content, whereas only ~6% of such particleswere detected in the presence of Dox, suggesting that most

4827

of the TAFII30-lacking cells die by apoptosis (Figure 4C).During apoptosis endonucleases that preferentially cutDNA are activated. The characteristic DNA ‘ladder’ offragmented DNA appeared 4 days after growing theTAFII30–La/–Lb:R cells in the absence of Dox and itsamount increased with time (Figure 6C, lanes 17, 21 and25). The translocation of the membrane phospholipidphosphatidylserine (PS) from the inner to the outer leafletof the plasma membrane is another feature of apoptoticcells. Annexin V binds specifically to PS of cells andtherefore allows detection of cells early in apoptosis(Philippeet al., 1997). To distinguish early apoptotic fromlate apoptotic cells we used annexin V in combination with

Page 6: Mammalian TAFII30 is required for cell cycle progression

D.Metzger et al.

Fig. 4. TAFII30-null cells are impaired in their proliferation and accumulate in G1 phase of the cell cycle. (A) Analysis of TAFII30, TBP, TAFII55and TAFII100 protein levels in HeLa, F9 WT and TAFII30–La/–Lb:R cells grown in the presence (1) or absence (–) of Dox. Whole-cell extracts wereprepared from the various cell lines as indicated, and 25µg of each protein extract was loaded on to a 10% gel and analysed by Western blottingusing mAbs raised against TBP, hTAFII55 and TAFII100 (see Wieczoreket al., 1998). (B) 104 cells were plated on day zero in the presence (1Dox)or absence (–Dox) of Dox and counted for 6 days. The number of cells grown in the presence (black columns) and absence (white columns) of Doxare indicated. (C) TAFII30–La/–Lb:R cells were grown in the presence (1Dox) or absence (–Dox) of Dox for 2 and 5 days as indicated, assubconfluent cultures. Cells were fixed, stained with PI and analysed by FACS. Thex-axis indicates the DNA content and they-axis the cell number.For each histogram ~20 000 particles have been gated. For each1Dox and –Dox sample, the living cells (x-axis is a linear scale) and all theparticles (x-axis is a log scale) were gated in the right and left panels, respectively. The percentage of cells in G1/G0, S and G2/M are indicated(accurate to within6 5%). M1 represents the sub-2N size DNA-containing particles.

propidium iodide (PI). The proportion of pre-apoptoticparticles was similar when TAFII30–La/–Lb:R cells weregrown for 2 days in the absence or presence of Dox(Figure 6A, 4 and 10% annexin V1, PI– cells, respect-ively). However, 5 days after Dox withdrawal.60% ofthe TAFII30–La/–Lb:R cells were pre-apoptotic, whereaswhen the cells were grown in the presence of Dox for5 days, only 16% were pre-apoptotic (Figure 6A). Takentogether, these results demonstrate that F9 cells undergoapoptosis in the absence of TAFII30 expression.

4828

Parietal, but not primitive endodermaldifferentiation is impaired in TAFII30-null cellsTo analyse the function of TAFII30 during RA-induceddifferentiation, TAFII30–La/–Lb:R cells were grown for 60 has monolayers in the presence or absence of Dox, andthen grown further in the presence or absence of 1µMRA. As for WT cells, RA induced a severe decrease ofthe proliferation rate of TAFII30–La/–Lb:R cells expressinghTAFII30 (1Dox) (Figure 6D and data not shown) andinduced a primitive endodermal-like morphological

Page 7: Mammalian TAFII30 is required for cell cycle progression

Conditional TAFII30 gene knock-out

Fig. 5. Cyclin E expression and phosphorylation of pRb is impaired inTAFII30-null cells. (A) Total cytoplasmic RNA from WT orTAFII30–La/–Lb:R cells, grown either in the presence of Dox (1) or forthe indicated number of days without Dox (–), was subjected toRT–PCR analysis for the following cyclin genes (number of PCRcycles in parentheses): cyclin A (25), cyclin B1 (15), cyclin C (20),cyclin D1 (18), cyclin D3 (15), cyclin E (15), and cyclin F (18). Thehypoxanthine phosphoribonucleotide transferase (HPRT) gene (10 PCRcycles) was used as an internal control. (B) RT–PCR analysis of thecyclin genes was performed as in (A) for at least three independentexperiments [the number of experiments (n) is indicated]. The mRNAlevels of the different cyclin genes are expressed relative to theamount present in the WT cells, which was taken as 100%. The errorbars represent the minimal and maximal values obtained. The numberunder each bar corresponds to the same lane in (A). (C) Whole-cellextracts from WT or TAFII30–La/–Lb:R cells, grown either in thepresence of (1) or for the indicated days without (–) Dox, weresubjected to Western blot analysis using the anti-pRb G3-245 mAb(PharMingen). The hypophosphorylated (pRb) and thehyperphosphorylated (pRb-P) forms of pRb are indicated.

differentiation (Figure 7). Whereas undifferentiatedTAFII30-null cells died within 5–7 days of Dox withdrawal[Figures 4B, 6D and 7A (c); data not shown], surprisinglythe RA treatment of TAFII30-null cells prevented rapid celldeath and induced a primitive endodermal morphologicaldifferentiation, similar to that observed with TAFII30-containing cells [Figure 7A (e) and (f)]. Thus, the geneswhich are required for this differentiation pathway areexpressed in the TAFII30-null cells. Furthermore, severalRA-inducible genes, i.e.RARβ2, Hox-1, Stra4 and Stra8(Chibaet al., 1997b) were similarly induced in TAFII30-null and WT cells after 2 days of RA treatment (datanot shown). Moreover, the RA-treated TAFII30-null cellssurvived for.10 days (Figure 6D). Interestingly, in thepresence of RA the TAFII30–La/–Lb:R cells arrested with a

4829

similar time course when grown in the absence orpresence of Dox (Figure 6D). The RA treatment ofTAFII30–La/–Lb:R cells grown in the presence of Doxresulted in a moderate increase (from 16 to 22%) in pre-apoptotic cells (compare1Dox 5 days in Figure 6A withB), in agreement with previous studies performed on WTF9 cells (Clifford et al., 1996). Note that this low levelof apoptosis could not be visualized in the DNA ladderassay used in this study (Figure 6C and data not shown).In contrast, the RA treatment of the TAFII30–La/–Lb:R cellsin the absence of Dox resulted in a strong reduction inpre-apoptotic cells (from 61 to 20%) as detected byfluorescence-activated cell sorting (FACS) analysis (com-pare –Dox 5 days in Figure 6A with B) or by theabsence of fragmented apoptotic DNA ‘ladder’ (Figure 6C,compare lanes 17 with 18, 21 with 22, and 25 with 26).

To induce parietal endodermal differentiation, WT andTAFII30–La/–Lb:R cells were grown in monolayer culturein the absence or presence of Dox and treated with 1µMRA and 250µM bt2cAMP. The morphological changeswhich normally accompany RA1 cAMP-induced differ-entiation of WT cells were observed in TAFII30–La/–Lb:Rcells expressing hTAFII30 [Figure 7A (h)]. In contrast,TAFII30-null cells (–Dox), although they differentiatedinto primitive endoderm-like cells, were strongly impairedin parietal endodermal differentiation. The parietal differ-entiation of WT and mutant cells was also investigatedby determining the expression of the thrombomodulin(TM) gene, a marker of parietal endodermal differentiation(Niforas et al., 1996; C.Rochette-Egly, personal com-munication). As expected, after 10 days of RA1cAMPtreatment, TM was strongly stimulated in WT and mutantcells expressing hTAFII30 (1Dox) (.90-fold, Figure 7B).In contrast, in the TAFII30-null cells (–Dox) the inductionof TM was very low (,6-fold) and the levels of TM wereat least five times lower than in the WT or hTAFII30expressing (1Dox) cells (Figure 7B). Taken together,these results indicate that TAFII30-null cells arestrongly impaired in parietal, but not primitive endodermaldifferentiation.

Discussion

TAFII30-null F9 cells are not viableIn this study, we performed conditional disruption of theTAFII30 gene in F9 murine embryonal carcinoma cells,using a Cre/LoxP strategy. No F9 cell clone disruptedfor the two alleles could be isolated, indicating thatundifferentiated TAFII30-null F9 cells are impaired in theirproliferation. This was confirmed by a study of thegenotype of a TAFII301/– [TAFII30–La/(L/L)b] F9 cell popula-tion after Cre-mediated disruption of the second allele.Although TAFII30–/–(TAFII30–La/–Lb) cells could bedetected during the 5 days which followed Cre re-combinase transfection, their number did not increase withtime and they eventually disappeared after a week, whereasTAFII301/– cells proliferated normally. The Cre-mediateddisruption of the targeted alleles encodes a truncatedprotein lacking the C-terminal evolutionary conserveddomain. Importantly, neither the truncated mRNA nor theprotein could be detected in heterozygous cells(TAFII301/–) or in rescue cells in which both alleles are

Page 8: Mammalian TAFII30 is required for cell cycle progression

D.Metzger et al.

Fig. 6. TAFII30-null cells die by apoptosis but RA strongly reduces the rate of apoptosis of the TAFII30-null cells. TAFII30–La/–Lb:R cells, grown inthe presence (1Dox) or absence (–Dox) of Dox, and either in the absence (A) or presence (B) of 1 µM RA for the indicated number of days, werelabelled with annexin V–fluorescein isothiocyanate (annexin V–FITC) and PI, and subjected to FACS. The results in (A) and (B) are represented asdual parameter scatter plots. The values indicated for the percentage of living (L; annexin V–, PI–), early apoptotic (Ap; annexin V1, PI–) and dead(D; annexin V1, PI1) cells are accurate to within6 5%. (C) TAFII30–La/–Lb:R cells were grown in the presence (1Dox) or absence (–Dox) of Dox,and in the presence (1RA) or absence (–RA) of retinoic acid, as indicated. Low mol. wt DNA (see Materials and methods) was electrophoresed in a1.2% agarose gel, followed by ethidium bromide staining. The DNA marker (M) size is expressed in bp. (D) 104 TAFII30–La/–Lb:R cells were platedon day zero. Cells were grown in the presence (1Dox) or absence (–Dox) of Dox and 1µM RA (added 60 h after plating) as indicated. Cells werecounted each day for 10 days.

inactivated. Therefore, inactivation of bothTAFII30allelesresults in TAFII30-null F9 cells.

Human TAFII30 can functionally replace its mousecounterpartTo analyse further the function of TAFII30 in F9 cells, wehave generated an F9 cell line expressing the humanTAFII30 protein in a conditional manner, in which bothTAFII30 alleles have been inactivated (TAFII30–La/–Lb:Rcells). TAFII30–La/–Lb:R cells expressing hTAFII30 areviable, in contrast to TAFII30-null cells, showing thathTAFII30 can functionally replace mTAFII30 in F9 cells

4830

and suggesting that hTAFII30 can enter into the differentmurine TAFII30-containing complexes such as TFIID,TFTC and PCAF/GCN5 complexes. This is in goodagreement with the high homology between the mouseand human proteins. However, some differences betweenTAFII30–La/–Lb:R cells expressing hTAFII30 and WT cellswere observed. For example, their RA-induced morpho-logical primitive endodermal differentiation is similar,although not identical, and parietal endodermal differenti-ation of the rescue cells is delayed by 48–56 h (data notshown). This partial rescue might reflect the amino aciddifferences between the N-terminal region of the mouse

Page 9: Mammalian TAFII30 is required for cell cycle progression

Conditional TAFII30 gene knock-out

Fig. 7. Parietal but not primitive endodermal differentiation isimpaired in TAFII30-null cells. (A) WT [(a), (d) and (g)], andTAFII30–La/–Lb:R cultured in the presence [1Dox; (b), (e) and (h)] orabsence [–Dox; (c), (f) and (i)] of Dox were treated 60 h after platingwith either control vehicle [(a), (b) and (c)], 1µM RA [(d), (e) and(f)] or 1 µM RA 1 250µM bt2cAMP [(g), (h) and (i)]. Cells werephotographed [8, 10 and 12 days following the plating for (a–f), (g)and (h–i), respectively] under a phase-contrast microscope at 1253magnification. (B) Cytoplasmic RNA from WT or TAFII30–La/–Lb:Rcells (grown either in the presence or absence of Dox) and treatedwith RA alone or RA1 cAMP (as indicated) for 10 days, wassubjected to RT–PCR analysis to test the expression of the parietalmarker gene TM. The HPRT gene was used as an internal control.

and the human protein and/or the absence of regulationof the transgene expression.

TAFII30 is required for G1/S progression of the cellcycle and cyclin E expressionThe conditional expression of hTAFII30 in TAFII30–/– cellsallowed us to study its role in the cell cycle regulation.FACS analysis showed that TAFII30-null cells accumulatein G1/G0 phase. Molecular analyses revealed that cyclin Eexpression is down-regulated after suppression of TAFII30expression, while expression of cyclin-dependent kinaseinhibitor p21 is enhanced (data not shown). Consistentwith the decreased cyclin E and increased p21 expression,we found that phosphorylation of pRb is decreased inTAFII30 lacking cells. These changes are all possiblecauses of the G1 cell cycle arrest phenotype of the TAFII30lacking cells. Taken together, these results suggest thatTAFII30 may regulate cyclin E expression and that onlycertain genes are regulated (directly or indirectly) byTAFII30.

Interestingly, growth arrest ofyTAFII90 andyTAFII150(Tsm1) mutant yeast strains occurs at the G2/M boundaryof the cell cycle, whereasyTAFII145 mutant strains arrestin G1 phase (Aponeet al., 1996; Walkeret al., 1996).yTAFII145 has been shown to be required for transcriptionof G1 cyclins and certain B-type cyclin genes (Walker

4831

et al., 1997). Furthermore, a hamster cell line containinga temperature-sensitive mutation in TAFII250 undergoesG1 arrest upon shifting to the restrictive temperature(Sekiguchiet al., 1991), resulting in reduced transcriptionof G1 cyclin genes such as cyclin A and D1 (Wang andTjian, 1994; Suzuki-Yagawaet al., 1997; Wanget al.,1997). Thus, different TAFIIs may specifically regulatedistinct cyclin genes to control cell proliferation.

RA prevents cell death of TAFII30-null cells andinduces primitive, but not parietal endodermaldifferentiationOur results indicate that cells lacking TAFII30 are notsimply arrested in G1/G0 phase of the cell cycle, butundergo apoptosis (Figures 4C, 6A and C). The analysisof the expression of 80 genes (on a mouse cDNA expres-sion array) involved in the regulation of apoptosis, revealedthat the mRNA levels of 14 genes were up-regulated (i.e.encoding caspase 3, 6 and 11, BAD and BID) and fivegenes were down-regulated (encoding i.e. the p55 celldivision control protein and the glutathioneS-tranferaseMu 1 protein) in TAFII30-null cells when compared withcells expressing hTAFII30 (data not shown). WhetherTAFII30 is directly or indirectly involved in the regulationof these genes is unknown. The lack of TAFII30 mayinduce apoptosis indirectly by deregulating distinct cellcycle check-points, i.e. pRb and/or E2F activities. Arecent study has suggested that another ‘specific’ TAFII,hTAFII105, is involved in activation of anti-apoptoticgenes by serving as a co-activator for NF-κB, a factorknown to protect cells from apoptotic stimuli (Yamit-Heziand Dikstein, 1998). It is therefore conceivable thatTAFIIs may regulate anti-apoptotic events through differentpathways.

Interestingly, after RA-treatment, TAFII30-null cells didnot die, but differentiated into primitive endodermal-likecells, and arrested with a similar time course to the RA-treated WT F9 cells (Figure 6D). Therefore, in contrastto WT cells (where RA not only induces differentiation,but also antiproliferation and apoptosis), RA-induced dif-ferentiation prevents TAFII30-null cell death by stronglyreducing apoptosis. These data suggest that when the cellsare programmed to proliferate, a G1 check-point, wherethe TAFII30 function is indispensable, does not functionproperly and apoptosis is triggered. On the other hand,when cells are programmed to differentiate by RA, theG1 check-point is not used since the cells enter G0,and thus TAFII30-null cells differentiate into primitiveendodermal cells and survive. Strikingly however, thegenes that are needed to trigger the morphological changesleading to primitive endodermal differentiation, arecorrectly expressed in TAFII30-null cells. In contrast,parietal endodermal differentiation is strongly impaired inTAFII30-null cells. Parietal endodermal differentiation isapparently achieved in two steps, an initial RA-induceddifferentiation into primitive endoderm, followed by acAMP-induced differentiation switch from primitive toparietal endoderm (Stricklandet al., 1980). Thus, TAFII30may be involved in the regulation of certain cAMP-induced genes. In any event, our results indicate thatTAFII30 is not needed for the transcription of genesrequired for the differentiation pathways leading to theprimitive endodermal differentiation of F9 cells and for

Page 10: Mammalian TAFII30 is required for cell cycle progression

D.Metzger et al.

their survival in the G0 state. Interestingly, several RA-responsive genes can be activated in TAFII30-null cells tosimilar levels to those in WT cells (data not shown),indicating that TAFII30-containing complexes are notabsolutely required for transcriptional activation by RAR/RXR heterodimers. In conclusion, we have identified acellular context in which mammalian cells can survive inthe absence of TAFII30, indicating that all genes necessaryfor primitive differentiation and survival of F9 cells canbe activated without TAFII30. In contrast to the yeastTAFII knock-out experiments (Moqtaderiet al., 1996a;Walker et al., 1996), the expression of different mTAFIIsand mTBP was not affected in TAFII30-null cells(Figure 4B) further suggesting that the TFIID, TFTC andPCAF/GCN5 complexes may function without TAFII30.

Our results demonstrate that mammalian TAFIIs are notgenerally required for activator-dependent transcription,but are essential for the transcription of specific genes.Furtherin vitro experiments will be needed to understandthe exact molecular mechanisms by which TAFII30 in thedifferent TAFII30-containing complexes, such as TFIID,TFTC and PCAF/GCN5, modulates the transcription of asubset of genes.

Materials and methods

Cloning of the mouse TAFII30 gene and determination ofexon–intron boundariesA mouse embryonic stem (ES) cell genomic library constructed inλGEM12 phage was screened with a 300 bp human TAFII30 cDNAprobe (from position 355 to 654; Jacqet al., 1994). A 16 kb DNAfragment containing the entire mouseTAFII30 gene and its flankingsequences was subcloned in pBluescript II SK1 (pBSK, Stratagene) andmapped with restriction enzymes (Figure 1). The exon–intron boundariesof the mouse (m) TAFII30 gene were determined by DNA sequencing.

Targeting of the TAFII30 gene in F9 cellsThe targeting vectors pTAFII30(L:LNL)a and pTAFII30(L:LNL)b were con-structed as follows. The 3.2 kbBamHI–EcoRI restriction fragment,containing exons 1, 2 and 3 of themTAFII30 gene (Figure 1A), wassubcloned into the corresponding sites of pBEX, resulting in pBEX–TAF1-3. pBEX is a pBSK derived vector containing a new polylinker(BamHI, EcoRI and XhoI), obtained by cloning the oligonucleotides59-GGTACCGTGGAATTCAGCTCGAGAGTAC-39 and 59-TCTCGA-GCTGAATTCCACGGTACCAGCT-39 into the SacI and KpnI sites ofpBSK. A SalI site was introduced between exons 1 and 2, and aNotIsite between exons 2 and 3 by site directed mutagenesis of pBEX–TAF1-3 with the oligonucleotides 59-CCCCGGCACCTAGTCGACG-ATGCCCTCCTCCTCCCT-39 and 59-GGCAGAGGCAGGCGGCCGC-TGAGTTCAAAGCCAG-39, respectively, resulting in pBEX–TAFSN.The oligonucleotides 59-TCGACATAACTTCGTATAATGTATGCTA-TACGAAGTTATGGTACC-39 and 59-TCGAGGTACCATAACTTCGT-ATAGCATACATTATACGAAGTTATG-3 9, containing a loxP site(labelled asLa or Lb in the following constructions, depending on theirorientation), were cloned into theSalI site of pBEX–TAFSN, resultingin pBEX–TAFLaN and pBEX–TAFLbN. To clone the floxed tk-Neocassette (Metzgeret al., 1995) into theNotI site of pBEX–TAFLaN andpBEX–TAFLbN vectors, a polylinker containing the following restrictionsites (NotI, ClaI, BglII, XbaI, SacI andNotI; 59-GCGGCCGCTAAATCG-ATTCGAGATCTCAAGTCTAGAGACGAGCTCTAAGCGGCCGCG-TAC-39 ) was cloned in theSacI and KpnI sites of pBSK, resulting inpBSK–NBXN. The 3.2 kbBamHI–XbaI fragment containing the floxedtk-Neo cassette, isolated from pHR56 (Metzgeret al., 1995), was clonedinto the corresponding sites of pBSK–NBXN, resulting in pNLNL–XN.The 3 kbNotI fragment containing the floxed tk-Neo cassette excisedfrom this vector and introduced into theNotI sites of pBEX–TAFLaNand pBEX–TAFLbN vectors, resulted in pBEX–TAFII30(L:LNL)a andpBEX–TAFII30(L:LNL)b, respectively. In the pBEX–TAFII30(L:LNL)a vec-tor the three loxP sites are in the (a) orientation and in the pBEX–TAFII30(L:LNL)b vector the three loxP sites are in the opposite, (b)orientation. To obtain the final targeting vectors, the 6.7 kbEcoRI–XhoI

4832

DNA fragment containingmTAFII30 exons 4 and 5 and its 39 flankingsequences, was inserted into the pBEX–TAFII30(L:LN)La and pBEX–TAFII30(L:LNL)b, resulting in the targeting vectors pTAFII30(L:LNL)a andpTAFII30(L:LNL)b (Figure 1). TheCre expression vector pSG–Cre wasobtained by cloning a PCR-amplifiedCre gene (Metzgeret al., 1995)into theEcoRI–BglII sites of pSG5.

F9 cells were electroporated with 5µg of the 13.8 kbBamHI–XhoIfragment, isolated either from pTAFII30(L:LNL)a or pTAFII30(L:LNL)b

vectors (Figure 1), or with 5µg of the pSG–Cre expression vector(Clifford et al., 1996). The selection of neomycin-resistant clones,Cre recombinase mediated excision and Southern blot analysis wereperformed as described (Metzgeret al., 1995). The 59 probe correspondsto a 0.8 kbEcoRV–KpnI genomic fragment, located outside the genomicsequences present in the targeting vector (Figure 1A). The primers usedfor the identification of the genotypes (Figures 2C and 3C) and theirlocations were as follows: SG12 (59-CTGCCCACTAGCACGGCC-39)located in the first exon, SE89 (59-CAGTCTAACCTGCTCCGAG-39)located between exons 3 and 4, and WY60 (59-CGCTTAGAGCTCGT-CTCTAG-39) specific to the junction between the Loxb site and intron 2(Figures 1 and 3).

A Dox-inducible hTAFII30 expression system in F9TAFII30–La/(L:L)b cellsThe expression vector pDG1–rtTA, in which the expression of the rtTA(Gossenet al., 1995) is under the control of the PGK promoter, wasconstructed by inserting the 1044-bpEcoRI–BamHI fragment isolatedfrom pUHD172–1Neo (Gossenet al., 1995) into the corresponding sitesof the pDG1 expression vector. AnEcoRI–BglII humanTAFII30 cDNAfragment was excised from pXJ–TAF30 (Jacqet al., 1994) and clonedinto the EcoRI–BamHI sites of the pUHD10-3 vector (Gossen andBujard, 1992), resulting in the expression vector pUHD10–TAF30 inwhich the expression of hTAFII30 is under the control of the tet-operator.The TAFII30–La/( L:L)b F9 cells were co-electroporated with 5µg ofpDG1–rtTA and linearized byAatII and with 0.5 µg of XhoI-digestedpPGK-hygro vector (te Rieleet al., 1990). After hygromycin selection,rtTA positive clones were selected by genomic PCR using the primersQY74 (59-CACGCTTCAAAAGCGCACGT-39) and WK31 (59-CAAT-ACAGTGTAGGCTGCTGCTC-39), and by Western blotting analysisusing three anti-VP16 monoclonal antibodies (2GV4, 5GV2 and 5GV7;White et al., 1992). A rtTA positive TAFII30–La/( L:L)b cell line was co-electroporated with 5µg of the 2 kb AatII–HindIII DNA fragmentisolated from pUHD10–TAFII30 and with 0.5µg of the neomycin-resistance cassette (a 1.6 kbBglII DNA fragment isolated from thevector pKJ-1B; pKJ-1B was obtained by cloning the oligonucleotide59-AATTAGATCT-39 into the EcoRI site of pKJ1; McBurneyet al.,1991). After neomycin selection, cells were treated with 1µg/ml Doxfor 3 days, and hTAFII30 positive clones were selected by Westernblot analysis using the human TAFII30-specific antibody 2F4 (Jacqet al., 1994).

Western blot analysisCells resuspended in 20 mM Tris–HCl pH 7.5, 2 mM DTT, 20% glycerol,0.4 M KCl, 1 mM PMSF and protease inhibitor cocktail (2.5µg/ml ofleupeptin, pepstatin, chymostatin, antipain and aprotinin), were lysed bythree cycles of freeze–thaw and cell debris was eliminated by centrifug-ation. Proteins were loaded on a 10% SDS–polyacrylamide gel, separatedby electrophoresis and electroblotted. The blots were then treatedwith monoclonal antibodies followed by incubation with peroxidase-conjugated goat anti-mouse IgG antibody (Jackson Immuno Research).Chemiluminescence detection was performed according to the manufac-turer’s instructions (Amersham).

PCR amplification of genomic DNA and RT–PCRGenomic DNA was prepared as follows: cells grown in 6-well plateswere collected in 500µl of lysis buffer (50 mM Tris–HCl pH 8.0,200 mM NaCl, 5 mM EDTA, 1% SDS and 0.2 mg/ml proteinase K) andincubated overnight at 55°C. The lysate was extracted with phenol/chloroform and chloroform and precipitated with ethanol at roomtemperature. PCR amplifications were performed in 50µl reactionscontaining 67 mM Tris–HCl pH 8.8, 16 mM (NH4)2SO4, 10 mMβ-mercaptoethanol, 1.5 mM MgCl2, 10% DMSO, 0.2 mM dNTPs,primers (12.5 pmol each), 1µl of proteinase K-treated cell extract or20 ng of purified genomic DNA and 2.5 UTaq polymerase. Theamplification conditions were: 94°C for 5 min followed by 35 cycles at94°C for 30 s and 55°C for 30 s, and finally one cycle at 94°C for 30 s,50°C for 30 s and 72°C for 5 min.

Cytoplasmic RNA extraction following cell lysis with 0.5% NP-40

Page 11: Mammalian TAFII30 is required for cell cycle progression

Conditional TAFII30 gene knock-out

and semi-quantitative RT–PCRs were performed as described (Chibaet al., 1997b). The PCR primers used for amplification of the differenttranscripts are available upon request. To make sure that amplificationswere in the linear range, aliquots of PCR products were taken from eachreaction at three different cycles between cycles 15 and 25, dependingon the gene analysed (Freemanet al., 1994). Control experiments wereperformed to determine the range of PCR cycles over which amplificationefficiency remained linear and to verify that the amount of PCR productwas directly proportional to the amount of input RNA (data not shown).End-labelled oligonucleotides were generated for probing the PCRproducts on Southern blots. Transcript levels were quantified with aBAS 2000 bio-imaging analyser (Fuji Ltd) and were normalized to thecorresponding HPRT levels.

Analysis of F9 cell growth and differentiationCells were seeded in 10-cm culture dishes at a density of 104 cells/wellat day zero, in the presence or absence of 1µg/ml Dox. The mediumwas changed every second day, and 1µM RA or 1 µM RA 1 250µMbt2cAMP was added as indicated. Cells were counted with a particlecounter (Coulter Z2) and their differentiation status was examined underlight microscopy as described (Cliffordet al., 1996).

Flow cytometric quantitation of cell cycle and apoptosis,and extraction of low mol. wt DNAFlow cytometric quantitation of cell cycle was carried out as describedin Clifford et al. (1996), except that instead of ethidium bromide,propidum iodide (PI) was used at a final concentration of 25µg/ml.Flow cytometric quantitation of apoptosis using annexin V together withPI was as described (Lecoeur and Gougeon, 1996; Philippeet al., 1997).PI stains non-fixed cells in late-apoptotic and necrotic stages as thesecells have lost their membrane inegrity. Briefly, 0.23106 cells wereresuspended in 50µl of AV buffer (10 mM HEPES–NaOH pH 7.4,150 mM NaCl, 5 mM KCl, 1 mM MgCl2 and 1.8 mM CaCl2) andincubated with 1µl FITC-labelled AV (CALTAG Laboratories) for30 min at room temperature in the dark. Five hundred microlitres of AVbuffer containing 5µg/ml PI was added for 5 min (in the dark) and thesamples were analysed on a FACScan (Beckton Dickinson) equippedwith a single argon ion laser. A minimum of 10 000 cells per samplewere analysed with the CELLQuest software (Beckton Dickinson).

Extraction of low mol. wt DNA from apoptotic cells was done in0.2 M phosphate–citrate buffer pH 7.8, as described by Gonget al. (1994).

Acknowledgements

We thank P.Chambon for his continuous support. We are grateful toJ.M.Garnier for the ES cell library, to C.Waltzinger for help in theflow cytometry studies, to P.Bouillet, J.Brocard, H.Bujard, H.Chiba,S.Girardin, X.Jacq, M.Lieb, Y.Lutz, J.Mountford and C.Rochette-Eglyfor reagents and advice. We also thank the cell culture group forproviding cells, F.Ruffenach for oligonucleotide synthesis, and R.Buchert,B.Boulay and J.M.Lafontaine for preparing the figures. This work wassupported by funds from the Institut National de la Sante´ et de laRecherche Me´dicale, the Centre National de la Recherche Scientifique,the Hopital Universitaire de Strasbourg, the Association pour laRecherche sur le Cancer, the Fondation pour la Recherche Me´dicale, theLigue Nationale Contre le Cancer, the Comite´ Departemental du Haut-Rhin de la Ligue National Contre le Cancer and the Human FrontierScience Program.

References

Apone,L.M., Virbasius,C.M., Reese,J.C. and Green,M.R. (1996) YeastTAFII90 is required for cell-cycle progression through G2/M but notfor general transcription activation.Genes Dev., 10, 2368–2380.

Bartek,J., Bartkova,J. and Lukas,J. (1997) The retinoblastoma proteinpathway in cell cycle control and cancer.Exp. Cell Res., 237, 1–6.

Bell,B. and Tora,L. (1999) Regulation of gene expression by multipleforms of TFIID and other novel TAFII-containing complexes.Exp.Cell. Res., 246, 11–19.

Brand,M., Yamamoto,K., Staub,A. and Tora,L. (1999) Identificationof TBP-free TAFII-containing complex subunits suggests a role innucleosome acetylation and signal transduction.J. Biol. Chem., 274,18285–18289.

Chiba,H., Clifford,J., Metzger,D. and Chambon,P. (1997a) Specific andredundant functions of retinoid X receptor/retinoic acid receptor

4833

heterodimers in differentiation, proliferation and apoptosis of F9embryonal carcinoma cells.J. Cell Biol., 139, 735–747.

Chiba,H., Clifford,J., Metzger,D. and Chambon,P. (1997b) Distinctretinoid X receptor/retinoic acid receptor heterodimers aredifferentially involved in the control of expression of retinoid targetgenes in F9 embryonal carcinoma cells.Mol. Cell. Biol., 17, 3013–3020.

Clifford,J., Chiba,H., Sobieszczuk,D., Metzger,D. and Chambon,P. (1996)RXRα-null F9 embryonal carcinoma cells are resistant to thedifferentiation, anti-proliferative and apoptotic effects of retinoids.EMBO J., 15, 4142–4155.

Freeman,R.S., Estus,S. and Johnson,E.M.,Jr (1994) Analysis of cell cycle-related gene expression in postmitotic neurons: selective induction ofCyclin D1 during programmed cell death.Neuron, 12, 343–355.

Gong,J., Traganos,F. and Darzynkiewicz,Z. (1994) A selective procedurefor DNA extraction from apoptotic cells applicable for gelelectrophoresis and flow cytometry.Anal. Biochem., 218, 314–319.

Goodrich,J.A., Cutler,G. and Tjian,R. (1996) Contacts in context:promoter specificity and macromolecular interactions in transcription.Cell, 84, 825–830.

Gossen,M. and Bujard,H. (1992) Tight control of gene expression inmammalian cells by tetracycline-responsive promoters.Proc. NatlAcad. Sci. USA, 89, 5547–5551.

Gossen,M., Freundlieb,S., Bender,G., Muller,G., Hillen,W. and Bujard,H.(1995) Transcriptional activation by tetracyclines in mammalian cells.Science, 268, 1766–1769.

Grant,P.A., Schieltz,D., Pray-Grant,M.G., Steger,D.J., Reese,J.C.,Yates,J.R. and Workman,J.L. (1998) A subset of TAFIIs are integralcomponents of the SAGA complex required for nucleosome acetylationand transcriptional stimulation. Cell, 94, 45–53.

Hatakeyama,M., Brill,J.A., Fink,G.R. and Weinberg,R.A. (1994)Collaboration of G1 cyclins in the functional inactivation of theretinoblastoma protein.Genes Dev., 8, 1759–1771.

Jacq,X., Brou,C., Lutz,Y., Davidson,I., Chambon,P. and Tora,L. (1994)Human TAFII30 is present in a distinct TFIID complex and is requiredfor transcriptional activation by the estrogen receptor. Cell, 79,107–117.

Lecoeur,H. and Gougeon,M.L. (1996) Comparative analysis of flowcytometric methods for apoptosis quantitation in murine thymocytesand human peripheral lymphocytes from controls and HIV-infectedpersons. Evidence for interference by granulocytes and erythrocytes.J. Immunol. Methods, 198, 87–99.

Lundberg,A.S. and Weinberg,R.A. (1998) Functional inactivation of theretinoblastoma protein requires sequential modification by at least twodistinct cyclin–cdk complexes.Mol. Cell. Biol., 18, 753–761.

May,M., Mengus,G., Lavigne,A.C., Chambon,P. and Davidson,I. (1996)Human TAFII28 promotes transcriptional stimulation by activationfunction 2 of the retinoid X receptors.EMBO J., 15, 3093–3104.

McBurney,M.W., Sutherland,L.C., Adra,C.N., Leclair,B., Rudnicki,M.A.and Jardine,K. (1991) The mousePgk-1 gene promoter contains anupstream activator sequence.Nucleic Acids Res., 19, 5755–5761.

Mengus,G., May,M., Jacq,X., Staub,A., Tora,L., Chambon,P. andDavidson,I. (1995) Cloning and characterization of hTAFII18,hTAFII20 and hTAFII28: three subunits of the human transcriptionfactor TFIID. EMBO J., 14, 1520–1531.

Mengus,G., May,M., Carre,L., Chambon,P. and Davidson,I. (1997)Human TAFII135 potentiates transcriptional activation by the AF-2sof the retinoic acid, vitamin D3 and thyroid hormone receptors inmammalian cells.Genes Dev., 11, 1381–1395.

Metzger,D., Clifford,J., Chiba,H. and Chambon,P. (1995) Conditionalsite-specific recombination in mammalian cells using a ligand-dependent chimeric Cre recombinase.Proc. Natl Acad. Sci. USA, 92,6991–6995.

Moqtaderi,Z., Bai,Y., Poon,D., Weil,P.A. and Struhl,K. (1996a) TBP-associated factors are not generally required for transcriptionalactivation in yeast. Nature, 383, 188–191.

Moqtaderi,Z., Yale,J.D., Struhl,K. and Buratowski,S. (1996b) Yeasthomologues of higher eukaryotic TFIID subunits.Proc. Natl Acad.Sci. USA, 93, 14654–14658.

Niforas,P., Chu,M.D. and Bird,P. (1996) A retinoic acid/cAMP-responsive enhancer containing a cAMP responsive element is requiredfor the activation of the mouse thrombomodulin-encoding gene indifferentiating F9 cells.Gene, 176, 139–147.

Ogryzko,V.V., Kotani,T., Zhang,X., Schlitz,R.L., Howard,T., Yang,X.J.,Howard,B.H., Qin,J. and Nakatani,Y. (1998) Histone-like TAFs withinthe PCAF histone acetylase complex. Cell, 94, 35–44.

Page 12: Mammalian TAFII30 is required for cell cycle progression

D.Metzger et al.

Philippe,J., Louagie,H., Thierens,H., Vral,A., Cornelissen,M. andDe Ridder,L. (1997) Quantification of apoptosis in lymphocyte subsetsand effect of apoptosis on apparent expression of membrane antigens.Cytometry, 29, 242–249.

Pugh,B.F. and Tjian,R. (1991) Transcription from a TATA-less promoterrequires a multisubunit TFIID complex.Genes Dev., 5, 1935–1945.

Reed,S.I. (1997) Control of the G1–S transition.Cancer Surv., 29, 7–23.Sauer,B. and Henderson,N. (1990) Targeted insertion of exogenous DNA

into the eukaryotic genome by the Cre recombinase.New Biol., 2,441–449.

Scheer,E., Mattei,M.G., Jacq,X., Chambon,P. and Tora,L. (1995)Organization and chromosomal localization of the gene (TAF2H)encoding the human TBP-associated factor II 30 (TAFII30).Genomics,29, 269–272.

Sekiguchi,T., Nohiro,Y., Nakamura,Y., Hisamoto,N. and Nishimoto,T.(1991) The humanCCG1 gene, essential for progression of the G1phase, encodes a 210-kilodalton nuclear DNA-binding protein.Mol.Cell. Biol., 11, 3317–3325.

Sherr,C.J. (1994) G1 phase progression: cycling on cue. Cell, 79, 551–555.Strickland,S. and Mahdavi,V. (1978) The induction of differentiation in

teratocarcinoma stem cells by retinoic acid. Cell, 15, 393–403.Strickland,S., Smith,K.K. and Marotti,K.R. (1980) Hormonal induction

of differentiation in teratocarcinoma stem cells: generation of parietalendoderm by retinoic acid and dibutyryl cAMP. Cell, 21, 347–355.

Sumi-Ichinose,C., Ichinose,H., Metzger,D. and Chambon,P. (1997)SNF2β-BRG1 is essential for the viability of F9 murine embryonalcarcinoma cells.Mol. Cell. Biol., 17, 5976–5986.

Suzuki-Yagawa,Y., Guermah,M. and Roeder,R.G. (1997) The ts13mutation in the TAFII250 subunit (CCG1) of TFIID directly affectstranscription of D-type cyclin genes in cells arrested in G1 at thenonpermissive temperature.Mol. Cell. Biol., 17, 3284–3294.

Tansey,W.P. and Herr,W. (1997) TAFs: guilt by association?Cell, 88,729–732.

te Riele,H., Maandag,E.R., Clarke,A., Hooper,M. and Berns,A. (1990)Consecutive inactivation of both alleles of thepim-1 proto-oncogeneby homologous recombination in embryonic stem cells. Nature, 348,649–651.

Walker,S.S., Reese,J.C., Apone,L.M. and Green,M.R. (1996)Transcription activation in cells lacking TAFIIs. Nature, 383, 185–188.

Walker,S.S., Shen,W.C., Reese,J.C., Apone,L.M. and Green,M.R. (1997)Yeast TAFII145 is required for transcription of G1/S cyclin genes andregulated by the cellular growth state. Cell, 90, 607–614.

Wang,E.H. and Tjian,R. (1994) Promoter-selective transcriptional defectin cell cycle mutant ts13 rescued by hTAFII250. Science, 263, 811–814.

Wang,E.H., Zou,S. and Tjian,R. (1997) TAFII250-dependent transcriptionof cyclin A is directed by ATF activator proteins.Genes Dev., 11,2658–2669.

White,J., Brou,C., Wu,J., Lutz,Y., Moncollin,V. and Chambon,P. (1992)The acidic transcriptional activator GAL-VP16 acts on preformedtemplate-committed complexes.EMBO J., 11, 2229–2240.

Wieczorek,E., Brand,M., Jacq,X. and Tora,L. (1998) Function of TAFII-containing complex without TBP in transcription by RNApolymerase II. Nature, 393, 187–191.

Yamit-Hezi,A. and Dikstein,R. (1998) TAFII105 mediates activation ofanti-apoptotic genes by NF-κB. EMBO J., 17, 5161–5169.

Zhou,J., Zwicker,J., Szymanski,P., Levine,M. and Tjian,R. (1998) TAFIImutations disrupt dorsal activation in theDrosophila embryo.Proc.Natl Acad. Sci. USA, 95, 13483–13488.

Zhou,Q., Lieberman,P.M., Boyer,T.G. and Berk,A.J. (1992) Holo-TFIIDsupports transcriptional stimulation by diverse activators and from aTATA-less promoter.Genes Dev., 6, 1964–1974.

Received June 1, 1999; revised and accepted July 8, 1999

4834