cell, vol. 85, 549–561, may 17, 1996, copyright 1996 by ...sethfield.ucsd.edu/papers/field..cell...

13
Cell, Vol. 85, 549–561, May 17, 1996, Copyright 1996 by Cell Press E2F-1 Functions in Mice to Promote Apoptosis and Suppress Proliferation Seth J. Field, 1,5 Fong-Ying Tsai, 2,6 Frank Kuo, 3 including Rb (Bagchi et al., 1991; Chellappan et al., 1991; Chittenden et al., 1991), Rb’s relatives p107 (Cao et al., Ana M. Zubiaga, 1,5,7 William G. Kaelin, Jr., 6 1992; Devoto et al., 1992; Lees et al., 1992; Shirodkar David M. Livingston, 6 Stuart H. Orkin, 2,4,6 et al., 1992) and p130 (Cobrinik et al., 1993), and cyclin- and Michael E. Greenberg 1,5 dependent kinases (Devoto et al., 1992; Lees et al., 1992; 1 Division of Neuroscience Pagano et al., 1992a, 1992b; Shirodkar et al., 1992; Dyn- 2 Division of Hematology/Oncology lacht et al., 1994; Krek et al., 1994; Xu et al., 1994). Children’s Hospital The best characterized member of the E2F family is 3 Department of Pathology E2F-1 (Helin et al., 1992; Kaelin et al., 1992; Shan et al., Brigham and Women’s Hospital 1992). During the S phase of the cell cycle, E2F-1 has 4 Howard Hughes Medical Institute been hypothesized to play a role in activating the expres- 5 Department of Neurobiology sion of genes important for the execution of S phase. Harvard Medical School The positive influence of E2F-1 on transcription of S Boston, Massachusetts 02115 phase genes may be restrained during G0 and G1 by 6 Dana Farber Cancer Institute E2F-1’s interaction with the dephosphorylated form of Boston, Massachusetts 02115 Rb. Prior to entry into S phase, Rb becomes highly phos- phorylated by cyclin-dependent kinases. This renders Rb incapable of binding to E2F-1 and may allow released Summary E2F-1 to activate gene expression (Weinberg, 1995). Further evidence that E2Fs promote entry into S phase Members of the E2F transcription factor family (E2F- derives from study of Drosophila lacking E2F function 1–E2F-5) are believed to be critical positive regulators in which cell proliferation was arrested in the G1 phase of cell cycle progression in eukaryotes although the of the cell cycle after the seventeenth embryonic division in vivo functions of the individual E2Fs have not been (Duronio et al., 1995). In addition, overexpression of elucidated. Mice were generated that lack E2F-1 and, E2F-1 in mammalian cells can cause oncogenesis (John- surprisingly, these mice develop and reproduce nor- son et al., 1994; Xu et al., 1995; Yang and Sladek, 1995) mally. However, E2F-1 2/2 mice exhibit a defect in T and was found to be sufficient to drive serum-starved, lymphocyte development leading to an excess of ma- growth-arrested fibroblasts into the cell cycle and ture T cells due to a maturation stage-specific defect through S phase (Johnson et al., 1993; Qin et al., 1994). in thymocyte apoptosis. As E2F-1 2/2 mice age they Together, these findings suggested that the primary exhibit a second phenotype marked by aberrant cell function of E2F-1 might be to activate genes that pro- proliferation. These findings suggest that while certain mote the G1 to S phase transition during cell prolifer- members of the E2F family may positively regulate cell ation. cycle progression, E2F-1 functions to regulate apop- However, several recent studies indicate that E2F-1 tosis and to suppress cell proliferation. may have functions in addition to promoting entry into S phase. One of these may involve regulation of apoptosis. Introduction The overexpression of E2F-1 or mutated forms of E2F-1 in serum-starved or actively growing fibroblasts Normal growth and development involve competing pro- can trigger apoptosis (Qin et al., 1994; Shan and Lee, cesses of cell proliferation and apoptotic cell death. 1994; Wu and Levine, 1994; Kowalik et al., 1995; Krek et Transcription factors of the E2F family, comprised of al., 1995) raising the possibility that E2F-1 may regulate E2F-1–E2F-5, have been suggested to play a key role genes important for cell death. Another possible func- in the regulation of cell proliferation (La Thangue, 1994; tion is suggested by the observation that Rb-bound Adams and Kaelin, 1995; Weinberg, 1995). First, E2F E2F-1 can still interact with its cognate DNA sequence binding sites are crucial for transcriptional activation element and thus repress the transcription of certain of genes that regulate S phase entry—such as c-myc cell cycle regulatory genes (Bandara et al., 1991; Chel- (Hiebert et al., 1989; Huang and Hearing, 1989; Thalmeier lappan et al., 1991; Chittenden et al., 1991; Weintraub et al., 1989), cyclin D (Motokura and Arnold, 1993; Sala et al., 1992, 1995; Neuman et al., 1995; Sellers et al., et al., 1994), and cyclin E (DeGregori et al., 1995; Duronio 1995). This finding has prompted the hypothesis that, and O’Farrell, 1995)—and genes that function to engage by repressing transcription, the Rb/E2F-1 complex may DNA synthesis—such as dihydrofolate reductase (Blake play an active role in suppressing progression through and Azizkhan, 1989; Mudryj et al., 1990), thymidine ki- G1 until an appropriate signal triggers Rb phosphoryla- nase (Dou et al., 1992; Ogris et al., 1993), and DNA tion and its release from E2F (Sellers et al., 1995). polymerase a (Pearson et al., 1991; Sala et al., 1994; Despite considerable progress in elucidating the mul- DeGregori et al., 1995). Second, the activity of E2Fs as tiple potential functions of E2F-1 during cell prolifera- transcription factors is regulated by their interaction tion, a potential limitation of the studies so far is their with other components of the cell cycle machinery— use of overexpression of wild-type or mutant versions of E2F-1 under conditions where wild-type E2F-1 was present and functional. Thus, it is uncertain whether the 7 Present address: Department of Genetics, University of the Basque Country, Bilbao 48080, Spain. predominant function of E2F-1 is to promote entry into

Upload: vulien

Post on 06-Apr-2018

216 views

Category:

Documents


2 download

TRANSCRIPT

Cell, Vol. 85, 549–561, May 17, 1996, Copyright 1996 by Cell Press

E2F-1 Functions in Mice to Promote Apoptosisand Suppress Proliferation

Seth J. Field,1,5 Fong-Ying Tsai,2,6 Frank Kuo,3 including Rb (Bagchi et al.,1991; Chellappan et al., 1991;Chittenden et al., 1991), Rb’s relatives p107 (Cao et al.,Ana M. Zubiaga,1,5,7 William G. Kaelin, Jr.,6

1992; Devoto et al., 1992; Lees et al., 1992; ShirodkarDavid M. Livingston,6 Stuart H. Orkin,2,4,6

et al., 1992) and p130 (Cobrinik et al., 1993), and cyclin-and Michael E. Greenberg1,5

dependent kinases (Devoto et al., 1992; Lees et al., 1992;1Division of NeurosciencePagano et al., 1992a, 1992b; Shirodkar et al., 1992; Dyn-2Division of Hematology/Oncologylacht et al., 1994; Krek et al., 1994; Xu et al., 1994).Children’s Hospital

The best characterized member of the E2F family is3Department of PathologyE2F-1 (Helin et al., 1992; Kaelin et al., 1992; Shan et al.,Brigham and Women’s Hospital1992). During the S phase of the cell cycle, E2F-1 has4Howard Hughes Medical Institutebeen hypothesized to play a role inactivating the expres-5Department of Neurobiologysion of genes important for the execution of S phase.Harvard Medical SchoolThe positive influence of E2F-1 on transcription of SBoston, Massachusetts 02115phase genes may be restrained during G0 and G1 by6Dana Farber Cancer InstituteE2F-1’s interaction with the dephosphorylated form ofBoston, Massachusetts 02115Rb. Prior to entry into S phase, Rb becomes highly phos-phorylated by cyclin-dependent kinases. This rendersRb incapable of binding to E2F-1 and may allowreleasedSummaryE2F-1 to activate gene expression (Weinberg, 1995).

Further evidence that E2Fs promote entry into SphaseMembers of the E2F transcription factor family (E2F-derives from study of Drosophila lacking E2F function1–E2F-5) are believed to be critical positive regulatorsin which cell proliferation was arrested in the G1 phaseof cell cycle progression in eukaryotes although theof thecell cycle after theseventeenth embryonic divisionin vivo functions of the individual E2Fs have not been(Duronio et al., 1995). In addition, overexpression ofelucidated. Mice were generated that lack E2F-1 and,E2F-1 in mammalian cellscan causeoncogenesis (John-surprisingly, these mice develop and reproduce nor-son et al., 1994; Xu et al., 1995; Yang and Sladek, 1995)mally. However, E2F-12/2 mice exhibit a defect in Tand was found to be sufficient to drive serum-starved,lymphocyte development leading to an excess of ma-growth-arrested fibroblasts into the cell cycle andture T cells due to a maturation stage-specific defectthrough S phase (Johnson et al., 1993; Qin et al., 1994).in thymocyte apoptosis. As E2F-12/2 mice age theyTogether, these findings suggested that the primary

exhibit a second phenotype marked by aberrant cellfunction of E2F-1 might be to activate genes that pro-

proliferation. These findings suggest that while certainmote the G1 to S phase transition during cell prolifer-

members of the E2F family may positively regulate cellation.

cycle progression, E2F-1 functions to regulate apop- However, several recent studies indicate that E2F-1tosis and to suppress cell proliferation. may have functions in addition to promoting entry into S

phase. One of these may involve regulation of apoptosis.Introduction The overexpression of E2F-1 or mutated forms of

E2F-1 in serum-starved or actively growing fibroblastsNormal growth and development involve competing pro- can trigger apoptosis (Qin et al., 1994; Shan and Lee,cesses of cell proliferation and apoptotic cell death. 1994; Wu and Levine, 1994; Kowalik et al., 1995; Krek etTranscription factors of the E2F family, comprised of al., 1995) raising the possibility that E2F-1 may regulateE2F-1–E2F-5, have been suggested to play a key role genes important for cell death. Another possible func-in the regulation of cell proliferation (La Thangue, 1994; tion is suggested by the observation that Rb-boundAdams and Kaelin, 1995; Weinberg, 1995). First, E2F E2F-1 can still interact with its cognate DNA sequencebinding sites are crucial for transcriptional activation element and thus repress the transcription of certainof genes that regulate S phase entry—such as c-myc cell cycle regulatory genes (Bandara et al., 1991; Chel-(Hiebertet al., 1989; Huang and Hearing, 1989; Thalmeier lappan et al., 1991; Chittenden et al., 1991; Weintraubet al., 1989), cyclin D (Motokura and Arnold, 1993; Sala et al., 1992, 1995; Neuman et al., 1995; Sellers et al.,et al., 1994), and cyclin E (DeGregori et al., 1995; Duronio 1995). This finding has prompted the hypothesis that,and O’Farrell, 1995)—and genes that function to engage by repressing transcription, the Rb/E2F-1 complex mayDNA synthesis—such as dihydrofolate reductase (Blake play an active role in suppressing progression throughand Azizkhan, 1989; Mudryj et al., 1990), thymidine ki- G1 until an appropriate signal triggers Rb phosphoryla-nase (Dou et al., 1992; Ogris et al., 1993), and DNA tion and its release from E2F (Sellers et al., 1995).polymerase a (Pearson et al., 1991; Sala et al., 1994; Despite considerable progress in elucidating the mul-DeGregori et al., 1995). Second, the activity of E2Fs as tiple potential functions of E2F-1 during cell prolifera-transcription factors is regulated by their interaction tion, a potential limitation of the studies so far is theirwith other components of the cell cycle machinery— use of overexpression of wild-type or mutant versions

of E2F-1 under conditions where wild-type E2F-1 waspresent and functional. Thus, it is uncertain whether the7 Present address: Department of Genetics, University of the Basque

Country, Bilbao 48080, Spain. predominant function of E2F-1 is to promote entry into

Cell550

Figure 1. Mutation of the E2F-1 Gene

(A) Partial structure of the E2F-1 gene deter-mined from restriction mapping, Southern hy-bridization, and DNA sequencing (data notshown). Exons are depicted as open boxes.The E2F-1 gene replacement construct is de-rived from a 7.4 kb fragment of the E2F-1gene disrupted in exon 3 by the PGK-neoselectable marker. In addition, exon 4 is de-leted. Adjacent to the E2F-1 DNA is the nega-tive selectable marker (PGK HSV-TK). Re-striction enzyme recognition sites areabbreviated as follows: E, EcoRV; S, SacI;Sm, SmaI; X, XcmI; H, HindIII; C, ClaI.

(B) Southern hybridization demonstrates thepresence of the targeted E2F-1 mutation inmice. Mouse genomic DNA was digested withthe restriction enzyme EcoRV, and analyzedby Southern blotting using an E2F-1 genomicprobe (as depicted in [A]). The wild-typeE2F-1 gene yields a band of 3.0 kb, while themutant gene yields a band of 2.3 kb. Southernhybridization using other restriction enzymesand both 59 and 39 E2F-1 genomic probesdemonstrates that the structure of the mutantgene is as expected in mice derived from allthree ES cell lines (data not shown). Further,these data and the use of a probe for neosequences indicate that additional copies ofthe gene targeting construct do not existelsewhere in the genome (data not shown).

(C) Northern hybridization demonstrates thatthe homozygous E2F-1 mutation abolishesE2F-1 mRNA expression. RNA was harvestedfrom cells arrested in S phase from two wild-type and two E2F-12/2 mice and used forNorthern hybridization with a probe derivedfrom the 39 coding region of the humanE2F-1 cDNA (see Experimental Procedures).As shown, wild-type fibroblasts express anz3.0 kb message corresponding to theE2F-1 mRNA. This mRNA is not present in

the E2F-1 homozygous mutant fibroblasts. The integrity and amount of RNA in each lane was demonstrated by stripping and rehybridizingthe same blot with a probe specific for the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mRNA.

S phase, suppress G1 progression, and/or regulate mutations abolish the ability of E2F-1 to bind DNA, acti-vate transcription, drive cell cycle progression, or triggerapoptosis. To address this issue, we generated and

studied E2F-1 deficient mice. Although these mice were apoptosis (Helin et al., 1992; Kaelin et al., 1992; Cresset al., 1993; Johnson et al., 1993; Shan and Lee, 1994).found to develop and reproduce normally, they exhibited

a number of defects in T cell development indicating The E2F-1 targeting construct also deletes E2F-1 exon4, encoding the leucine zipper and part of a “markedthat E2F-1 functions to positively regulate apoptosis

and suppress cell proliferation. E2F-1 appears not to be box” protein–protein interaction domain (Kaelin et al.,1992; Lees et al., 1993). Therefore, any RNA transcriptscritical for entry into S phase. Rather, this function may

be served by other members of the E2F family. Taken expressed from the mutated E2F-1 locus should notencode a functional protein. Gene-targeted J1 embry-together with previous findings demonstrating that

E2F-1 can cause oncogenesis when overexpressed, the onic stem (ES) cell lines were used to produce miceheterozygous for the E2F-1 mutation. Heterozygotesobservations described here reveal that a single gene,

depending on its level of expression or cellular environ- (1/2) were intercrossed to generate homozygous mu-tant mice (2/2), as demonstrated by Southern hybrid-ment, can function as either an oncogene, or paradoxi-

cally, as a suppressor of cell proliferation. ization (Figure 1B). Homozygotes produced from threeindependent ES cell clones were bred into both 129/Svand 129/Sv 3 C57BL/6 backgrounds and gave similarResultsresults in all experiments described here.

RNA expression of primary embryonic fibroblasts iso-Targeted Mutation of the E2F-1 GeneA 7.4 kb fragment of the E2F-1 gene was used to gener- lated from wild-type and E2F-12/2 mouse embryos was

examined by Northern analysis. Fibroblasts were syn-ate a targeting vector in which the DNA binding anddimerization domains of the E2F-1 gene were disrupted chronized at the start of S phase by exposure to hy-

droxyurea. Under these conditions, E2F-1 mRNA ex-by the insertion of a PGK-neo cassette (Figure 1A). Thesite of insertion lies within a region of E2F-1 in which pression is maximal (Kaelin et al., 1992; Li et al., 1994),

Apoptosis and Proliferation in E2F-1 Deficient Mice551

and wild-type fibroblasts express a 3.0 kb E2F-1 tran- with progenitors from thebone marrow, increased prolif-eration of thymocytes within the thymus, failure of ma-script (Figure 1C). This transcript was not detected inture thymocytes to leave the thymus, or a decreasedthe E2F-12/2 fibroblasts indicating that the engineeredrate of thymocyte death.mutation successfully disrupted E2F-1 mRNA expres-

To investigate these possibilities, the developmentalsion and therefore E2F-1 function.profiles of thymocytes from E2F-12/2 and wild-type micewere compared. By monitoring the expression of the

E2F-1 is Not Essential for OverallCD4 and CD8 cell surface markers, the extent of thymo-Development or Survivalcyte maturation was assessed. As thymocytes mature

E2F-12/2 mice are born and appear overtly normal. Ofthey progress sequentially through double-negative

351 mice born to crosses of E2F-11/2 mice, 97 (28%)(CD42/CD82), double-positive (CD41/CD81), and single-

were of the 1/1, 172 (49%) of the 1/2, and 82 (23%)positive (CD41/CD82 or CD42/CD81) stages (Scollay et

of the 2/2 genotype, indicating that E2F-1 1/1, 1/2, al., 1988). Upon staining with anti-CD4 and anti-CD8and 2/2 mice were equally viable. Moreover, crosses antibodies, thymuses of four- to six-week-old E2F-12/2

between E2F-12/2 mice also produced overtly normal mice were found consistently tocontain a higher fractionoffspring. Although E2F-12/2 mice weighed 17% less of mature thymocytes (CD41 or CD81) than wild-typethan wild-type littermates for at least the first 8 months littermates (Figure 3C). To establish further the identityof life (p < 0.01, n 5 56 wild type, n 5 43 E2F-12/2), no of the population of thymocytes expanded in the E2F-obvious gross or microscopic abnormalities were noted 12/2 mice, thymocytes were stained with an antibody toin the liver, brain, gut, cardiac or skeletal muscle, bone, CD3e, a component of the T cell receptor expressedspleen, kidneys, or pancreas at four to six weeks of age. at high levels in mature cells. The percent of CD3ehi

Thus, E2F-1 is not required for overall development, thymocytes was significantly increased in E2F-12/2

survival, or reproduction of mice. compared with wild-type mice (44% 6 12% versus 18%Since the E2Fs are believed to play a crucial role in 6 3% ina typicalexperiment), supporting the conclusion

regulating the G1/S transition, we examined the require- that in the absence of E2F-1 the fraction of mature thy-ment for E2F-1 in cell proliferation and cell cycle pro- mocytes is increased. Taking into account the totalnum-gression. Surprisingly, in E2F-12/2 primary embryonic ber of thymocytes per thymus, the absolute number offibroblasts, the doubling time, cell cycle distribution, cells in all thymocyte populations is increased in E2F-and timing of reentry into the cell cycle from G0 were 12/2 mice (Figure 3D). The E2F-12/2 mice exhibit a smallindistinguishable from that of wild-type fibroblasts (Fig- increase in the absolute number of immature (CD4/ures 2A and 2B). Likewise, when lymph node cells from CD8 double-negative and double-positive) thymocytes.four- to six-week-old E2F-12/2 mice and wild-type lit- However, E2F-12/2 mice exhibit a larger, selective in-termates were compared, they were found to respond crease in the number of mature (CD4/CD8 single-posi-to mitogenic signals equally well (Figure 2C). Cell pro- tive) thymocytes. Although the smaller increase in theliferation in vivo was examined by injecting mice with number of immature thymocytes could reflect increased5-bromo-29deoxyuridine (BrdU) to label cells in S phase. seeding of the thymus with precursor cells from the

bone marrow, the larger selective increase in the numberAs shown in Figures 2D and 2E (also Figure 5B and dataof mature thymocytes could not. In subsequent experi-not shown), proliferation ingut epithelium,spleen, lymphments, we focused our attention on elucidating thenodes, and thymus was indistinguishable between wild-mechanism by which a mutation in E2F-1 leads to atype and E2F-12/2 mice at four to six weeks of age.selective increase in the number of mature thymocytes.By contrast, in older mice differences in proliferation

A failure of thymocytes toexit the thymus and colonizebetween wild-type and E2F-12/2 mice were detectedthe periphery does not account for the increase in the(see below). Nevertheless, on the basis of the findingsnumber of mature thymocytes in E2F-12/2 mice. First,in younger animals, we can conclude that under manyflow cytometric analysis of lymph node cells using anti-circumstances E2F-1 is not required for cell proliferationbodies to CD3e or Thy1.2 (markers for T cells) and B220or cell cycle progression. This may reflect the ability(a marker for B cells) revealed that, as in wild-type mice,of other E2F family members to compensate for thethe lymph nodes of E2F-12/2 mice arecomposed primar-absence of E2F-1.ily (70% to 80%) of T cells. In addition, E2F-12/2 micehave visibly enlarged, hypercellular lymph nodes (Figure

Thymus Abnormalities in E2F-12/2 Mice 4; p < 0.01; Student’s t test, n 5 6 wild-type, n 5 6E2F-1 was found to play a critical role in normal thymic E2F-12/2), indicating that E2F-12/2 mice also have andevelopment. Upon dissection, the thymuses of four- increase in the number of mature T peripheral cells.to six-week-old E2F-12/2 mice were noticeably enlarged Thus, mature T cells effectively exit the thymus of E2F-compared with those of 1/1 littermates, even though 12/2 mice.other organs were consistently smaller. The ratio of thy- An increase in the rate of cell proliferation within themus:body weight in E2F-12/2 mice is increased by 25% thymus of young E2F-12/2mice was also ruled out asrelative to wild-type mice (Figure 3A; p < 0.03, Student’s the explanation for the thymic hypertrophy observed int test, n 5 14 wild-type, n 5 12 E2F-12/2). The increased these animals. Thymocyte proliferation in vitro in re-thymus size reflects a consistent increase in thymic cel- sponse to stimulation with mitogens was found to belularity demonstrated by a 55% increase in the number indistinguishable when thymocytes isolated from four-of thymocytes per thymus (Figure 3B; p < 0.02, Student’s to six-week-old wild-type and E2F-12/2 mice were com-t test, n 5 10 wild-type, n 5 10 E2F-12/2). Several mecha- pared. When the cell cycle distribution of freshly har-nisms might explain the increased number of thymo- vested thymocytes was analyzed, thymocytes from

four- to six-week-old wild-type and E2F-12/2 mice werecytes. These include increased seeding of the thymus

Cell552

Figure 2. Cell Proliferation and Cell CycleDistribution Are Unaltered in Young E2F-12/2

Mice

(A) Fibroblasts were harvested from wild-typeand E2F-12/2 mouse embryos and culturedin vitro. Doubling time was measured by fol-lowing the increase in cell number over timeas described in Experimental Procedures.Wild-type and E2F-12/2 primary embryonicfibroblasts had similar doublingtimes of 36.5 6

3.2 hr and 34.7 6 2.9 hr, respectively (mean 6

SEM). Error bars in this and subsequent fig-ures indicate the SEM.

(B) The cell cycle distributions of exponen-tially growing wild-type and E2F-12/2 primaryembryonic fibroblasts were found to be simi-lar. To assay cell cycle distribution, 75% con-fluent dishesof fibroblasts werepulse labeledwith BrdU for 1 hr, harvested, and fixed. DNAcontent was assessed by staining with pro-pidium iodide, and DNA synthesis assessedby staining with an antibody to BrdU, as de-scribed in Experimental Procedures.

(C) Lymphocytes isolated from the lymphnodes of four- to six-week-old wild-type andE2F-12/2 mice were stimulated in vitro with1.5 mg/ml concanavalin A and 100 U/ml in-terleukin-2 to assess cell cycle progressionto S phase from G0. At various times afterstimulation, cell cycle distribution was ana-lyzed, as described in Experimental Proce-dures. As shown, lymphocytes from wild-typeand E2F-12/2 mice responded equally wellto concanavalin A and interleukin-2. In otherexperiments, other stimuli including phorbolmyristate acetate and ionomycin or staphy-loccocal enterotoxin B and interleukin-2 werealso shown to produce similar responsesfrom wild-type and E2F-12/2 lymphocytes.

(D and E) In vivo BrdU labeling (as describedin Experimental Procedures) identifies cellsin S phase (appearing red) in organs fromfour- to six-week-old mice. As shown, gutepithelium (D) from four- to six-week-oldwild-type (1/1, on the left) and E2F-12/2 (onthe right) mice have similar numbers of cellsin S phase. Similarly, the spleens (E) fromfour- to six-week-old wild-type (1/1, on theleft) andE2F-12/2 (on the right) mice also havesimilar numbers of cells in S phase.

found to have similar cell cycle distributions (Figure 5A). Apoptosis was initially examined by monitoring the per-cent viability of in vitro cultured wild-type and E2F-12/2To examine in vivo the fraction of cells in S phase in the

thymuses of wild-type and E2F-12/2 mice, four- to six- thymocytes. Only 40% of wild-type thymocytes werestill viable after one day in culture. Control experimentsweek-old mice were injected with BrdU. Wild-type thy-

muses, as expected, demonstrated labeling predomi- demonstrated this loss of viability to be via an apoptoticpathway as measured by DNA end labeling techniques.nantly of cells of the thymic cortex (Figure 5B), indicating

proliferation of immature thymocytes. E2F-12/2 thy- Bycontrast, thymocytes from E2F-12/2 littermatesmain-tained 60% viability over the same time period, an in-muses demonstrated a similar distribution and extent

of labeling. Together, these experiments indicate that crease of 50% over wild-type (Figure 6A). In seven inde-pendent experiments comparing a total of 13 wild-typethymocyte proliferation both in vitro and in vivo in four-

to six-week-old mice is not affected by the absence and 13 E2F-12/2 littermates four to six weeks of age,the E2F-12/2 thymocytes consistently show increasedof E2F-1.

On the other hand, our analysis revealed a clear differ- viability. This increase in the survival of thymocytes fromE2F-12/2 mice is statistically highly significant (p < 0.001,ence in the extent of apoptotic cell death when thymo-

cytes from wild-type and E2F-12/2 mice were compared. two-way [genotype and days in culture] analysis of vari-

Apoptosis and Proliferation in E2F-1 Deficient Mice553

Figure 3. E2F-12/2 Mice Have an ExcessNumber of Mature Thymocytes and T Cells

(A) The weights of the thymuses from 14 wild-type and 12 E2F-12/2 mice four to six weeksof age are expressed as fraction of total bodyweight. As shown, relative thymus size ofE2F-12/2 mice is 25% greater than their wild-type littermates. For comparison, liver weightas a fraction of total body weight is 7% lessfor E2F-12/2 mice than their wild-type lit-termates (data not shown). The increase insize of the thymuses in E2F-12/2 mice com-pared with wild-type is statistically signifi-cant, with p < 0.03 (Student’s t test).

(B) Thymocytes were harvested from the thy-muses of 10 wild-type and 10 E2F-12/2 mice.The number of thymocytes per thymus wascounted and is shown in the graph. E2F-12/2

mice show a 55% increase in the number ofthymocytes per thymus compared with wild-type littermates (mean 6 SEM for wildtype 5 99 6 11 3 106 cells per thymus, forE2F-12/2 5154 6 18 3 106 cells per thymus).The effect of genotype on number of thymo-cytes per thymus is statistically significant,with p < 0.02 (Student’s t test).

(C) Thymocyte expression of the CD4 andCD8 cell surface antigens was examined byflow cytometry. Thymocytes from wild-typeand E2F-12/2 mice were labeled with a fluo-rescein conjugated antibody to CD8 and anR-phycoerythrin conjugatedantibody to CD4.CD4 and CD8 expression per cell were ana-lyzed by flow cytometry. In the graphs shown,each dot indicates the CD4 and CD8 expres-sion levels for a single cell. The cell popula-tions are divided into quadrants representingCD4/CD8 double-negative (lower left), CD4/CD8 double-positive (upper right), CD4 sin-gle-positive (upper left), and CD8 single-posi-tive (lower right) populations. The numbers ineach quadrant represent the percentage ofcells in that quadrant (mean 6 SEM). Asshown, the more mature CD4 and CD8 single-positive populations are expanded in theE2F-12/2 mice. The experiment shown repre-sents duplicate measurements from one pairof mice and is representative of seven experi-ments comparing in total 13 wild-type to 13E2F-12/2 mice.

(D) Calculation of the absolute number of thy-mocytes in each subpopulation based on thepercentages measured in (C) and the totalnumber of cells per thymus for each animal.E2F-12/2 mice have a small increase in theabsolute number of CD4/CD8 double-nega-tive and double-positive thymocytes com-pared to wild-type. However, E2F-12/2 micehave a larger selective increase in the abso-lute number of CD4/CD8 single-positive thy-mocytes compared with wild-type.

ance [ANOVA] of thymocyte viability, n 5 13 wild-type, homogeneous subpopulations of wild-type and E2F-12/2

thymocytes. CD4/CD8 double-positive or single-posi-n 5 13 E2F-12/2). In an average thymus, an increase insurvival of this magnitude would represent the inappro- tive thymocytes from wild-type or E2F-12/2 mice were

purified by cell sorting, and their survival in culture waspriate survival of over 30 million cells per day.Since the thymocyte population in E2F-12/2 mice dif- assayed. Purified CD41/CD81 thymocytes from E2F-

12/2 mice displayed a clearly enhanced level of survivalfered from that of wild-type mice in that it includes ahigher proportion of mature cells, a clear demonstration in culture when compared with their wild-type counter-

parts (Figure 6B), confirming a defect in apoptosis inof a defect in apoptosis in the E2F-12/2 thymocytes re-quired a comparison of the survival of developmentally the E2F-1 deficient thymocytes. By contrast, the survival

Cell554

Figure 4. E2F-12/2 Mice Have Enlarged Hypercellular Lymph Nodes

Lymph nodes from E2F-12/2 mice compared with lymph nodes fromcorresponding anatomical locations from wild-type littermates arevisibly enlarged (data not shown). To count the number of cells perlymph node, 12 or 13 lymph nodes were harvested from each ofsix E2F-12/2 mice and six wild-type littermates from correspondinganatomical locations (generally, two from external abdominal fascia,six axillary, four submandibular, and one from the ilial bifurcationof the abdominal aorta). Cells were harvested from the lymph nodes,counted, and the average number of cells per lymph node calcu-lated. The E2F-12/2 mice had 2.3 6 0.2 3 106 cells per lymph node,

Figure 5. Thymocyte Proliferation Is Not Detectably Altered inwhile wild-type littermates had 1.5 6 0.2 3 106 cells per lymph nodeYoung E2F-12/2 Mice(mean 6 SEM). The increased number of cells per lymph node in

E2F-12/2 mice relative to wild-type littermates is statistically signifi- (A) Thymocytes were harvested from wild-type and E2F-12/2 micecant with p < 0.01 (Student’s t test). four to six weeks of age and immediately pulse labeled for 1 hr

with 10 mM BrdU. Cells were fixed and cell cycle distribution wasmeasured as described in Experimental Procedures. As shown,of the more mature single-positive thymocytes was un-freshly isolated thymocytes from young wild-type andE2F-12/2 miceaffected by the loss of E2F-1 (Figure 6C). Like the matureare similarly distributed through the cell cycle.

thymocytes, the lymph node cells from E2F-12/2 and(B) In vivo BrdU labeling (as described in Experimental Procedures)wild-type mice, which are primarily mature single-posi-identifies thymocytes in S phase (appearing red) in thymuses from

tive T cells, also displayed comparable viability. We con- four- to six-week-old wild-type and E2F-12/2 mice. As shown, youngclude that the less mature double-positive E2F-12/2 thy- wild-type and E2F-12/2 mice have similar numbers of cells in Smocytes exhibit a stage-specific defect in apoptosis. phase in the thymus in vivo.

The observation that CD41/CD81 thymocytes fromE2F-1 deficient mice are not as susceptible to apoptosisas wild-type thymocytes raises the possibility that the thymocyte apoptosis both in vitro and in vivo. Moreover,mutant thymocytes might be defective in the process E2F-12/2 thymocytes demonstrate a defect in T cell re-of negative selection. Negative selection, the mecha- ceptor–stimulated apoptosis in vivo, as might be ex-nism by which thymocytes that recognize self-antigen pected if there is a defect in negative selection in theseare eliminated, results from apoptotic loss of self-anti- mice. The defect inCD4/CD8 double-positive thymocytegen-recognizing thymocytes stimulated through the T apoptosis in E2F-12/2 mice is likely the cause of thecell receptor. To investigate the possibility that E2F-1 increased production of single-positive thymocytes andmight play a role in immunologic negative selection, T T cells in these animals and probably accounts for thecell receptor signalling was stimulated in E2F-12/2 and thymic and lymph node hypertrophy observed in thewild-type mice by the injection of anti-CD3e antibodies

E2F-12/2 mice. We conclude that a primary function ofand the effect on thymic apoptosis analyzed. This stimu-

E2F-1 invivo is to regulate apoptosis in immature thymo-lus triggers apoptosis primarily in CD41/CD81 thymo-

cytes.cytes by a mechanism that may mimicnegative selection(Owen et al., 1988; Smith et al., 1989; Shi et al., 1991).Mice were injected with anti-CD3e or control vehicle,

Effects of E2F-1 Loss in Older Miceand 16 hr later the fraction of cells undergoing apoptosisWe next considered the possibility that the decreasedwas determined using an in situ single cellDNA fragmen-apoptosis in the CD41/CD81 thymocytes of young E2F-tation assay. As expected the injection of anti-CD3e12/2 mice and/or the continued absence of E2F-1 inantibodies into wild-type mice led to a significant in-these animals might affect the function of the thymuscrease (3- to6-fold) in thymocyte apoptosis. By contrast,or other organs in older E2F-12/2 mice. To examine thethymocytes of E2F-12/2 mice exhibited significantly lesseffect of the E2F-1 mutation in older mice, wild-type and(50%) anti-CD3e-induced thymocyte apoptosis (FigureE2F-12/2 mice 6 to 12 months of age were compared7; p < 0.002, paired t test, n 5 5 wild-type, n 5 5 E2F-

12/2). Thus, E2F-12/2 mice display a defect in CD41/CD81 in a variety of ways. Both the gross and microscopic

Apoptosis and Proliferation in E2F-1 Deficient Mice555

morphology of liver, gut, brain, cardiac and skeletalmus-cle, spleen, bone, and kidney were found to be similarin wild-type and E2F-12/2 mice. However, older E2F-12/2 mice displayed moderate testicular atrophy (z60%reduction in weight, data not shown). The reduction intesticular weight seems to result from a decrease in thenumber of sperm found in many of the seminiferoustubules. Furthermore, these same tubules are observedto contain an overgrowth of interstitial Leydig cells.While the mechanism of testicular atrophy is uncertain,it is possible that an overgrowth of Leydig cells is thecause of decreased sperm production, and thus thedecrease in testicular weight.

The most dramatic finding in the older E2F-12/2 micewas again in the thymus. Examination of cellproliferationin 10 wild-type and 11 E2F-12/2 mice by in vivo labelingwith BrdU revealed a striking increase in proliferation inthe thymus of older E2F-12/2 mice compared with wild-type littermates (Figure 8A). The proliferating cells inmost of these E2F-12/2 mice maintained normal mor-phology and were localized to the thymic cortex butnot the medulla. The localization of these proliferatingthymocytes within the cortex suggests that they arelikely to be immature thymocytes. The large increase inthe number of proliferating cells in the thymic cortex ofolder E2F-12/2 mice was seen in all 11 E2F-12/2 thy-muses examined, indicating a role for E2F-1 in the con-trol of thymocyte proliferation in older mice. Other tis-sues, including gut epithelium, spleen, lymph nodes,and liver in the E2F-12/2 mice displayed BrdU labelingindices that were comparable to those seen in wild-typemice (Figure 8B). One of the 11 older E2F-12/2 animals,in addition to displaying increased thymic proliferation,

Figure 6. CD4/CD8 Double-Positive Thymocytes from E2F-12/2

Mice Exhibit a Maturation Stage–Specific Defect in Apoptosis InVitro

(A) Thymocytes harvested from wild-type and E2F-12/2 mice werecultured in vitro for various amounts of time. Cell viability was as-sayed by trypan blue exclusion. Loss of viability was throughapoptosis as demonstrated by early DNA fragmentation measuredusing in situ nick translation (data not shown). The data shownrepresent the mean 6 SEM for quadruplicate measurements of via-bility of cultures from two wild-type and two E2F-12/2 mice after 24hr in vitro and are graphed as viability relative to one of the wild-type controls. These data are representative of experiments com-paring, in total, thymocytes from 13 wild-type and 13 E2F-12/2 mice.The difference in survival of thymocytes from wild-type and E2F-12/2 mice is statistically significant with p < 0.001 (two-way ANOVAof thymocyte viability grouped by genotype and days in culture).

(B) and (C) CD4/CD8 double-positive (B) and single-positive (C) thy-mocytes were purified by fluorescence-activated cell sorting afterlabeling with fluorescein isothiocyanate–conjugated anti-CD8 andR-phycoerythrin-conjugated anti-CD4. Cell viability over time wasassayed by trypan blue exclusion. The data shown represent themean 6 SEM for duplicate measurements at each time point forthymocytes from each animal. All thymocyte cultures had >95%viability at the start of the experiment. These data are representativeof experiments comparing, in total, thymocytes from 4 wild-typeand 3 E2F-12/2 mice. The difference between the rates of apoptosisof wild-type and E2F-12/2 double-positive, but not single-positive,thymocytes was corroborated by comparing the survival of thymo-cytes (predominantly double-positive cells) and lymph node cells(predominantly single-positive T cells) from two mice of each geno-type (data not shown).

Cell556

E2F-1 are in many ways normal. E2F-12/2 mice undergonormal embryonic development, are fully viable at birth,and mature to become reproductively competent. Theability of E2F-12/2 mice to do so well may be indicativeof the ability of other E2F family members to providesufficient E2F function for most developmental path-ways. Nevertheless, further examination of the E2F-12/2

mice has led to the identification of important nonredun-dant functions of E2F-1.

Most notably, within one month of birth E2F-12/2 micedisplay visible enlargement of the thymus and lymphnodes despite a reduction in the size of other organs.These enlarged lymphoid organs reflect an expansionof the population of mature CD4/CD8 single-positivethymocytes and peripheral T cells. This expansion ap-pears to be due to a defect in a normal pathway ofapoptosis. The defect in CD41/CD81 thymocyte apop-tosis in E2F-12/2 mice is observed in every animal exam-ined, indicating a consistent and important role for

Figure 7. E2F-12/2 Mice Display a Defect in Thymocyte Apoptosis E2F-1 in thymocyte apoptosis.In Vivo Apoptosis has been previously shown to be an impor-T cell receptor–mediated thymocyte apoptosis was stimulated by tant event duringnormal thymocyte maturation. Notably,injecting four- to six-week-old mice with an activating antibody to the elimination of thymocytes bearing self-reactive TCD3e (2C11) and assaying apoptosis 16 hr later using a DNA end– cell receptors occurs during negative selection and islabeling method (TUNEL; Gavrieliet al., 1992) as described in Experi-

believed to be mediated by T cell receptor–stimulatedmental Procedures. The figure depicts pooled data from fourapoptosis of CD4/CD8 double-positive thymocytes justindependent experiments comparing in total five wild-type and fiveprior to their conversion into mature thymocytes (Fowl-E2F-12/2 mice injected with anti-CD3e. The reduction in anti-CD3e-

induced apoptosis seen in E2F-12/2 mice compared with wild-type kes et al., 1988; Kisielow and von Boehmer, 1991). Theseis statistically significant with p < 0.002 (paired t test). Negative observations, taken together with our finding that E2F-controls omitting terminal deoxynucleotidyl transferase showed 12/2 mice have a defect in T cell receptor–stimulatedbackground staining of 0.6% 6 0.1%. Positive controls treated with

apoptosis of CD4/CD8 double-positive thymocytes,DNaseI showed 41% 6 9% staining, and 33% 6 3% of thymocytesraise the possibility that E2F-1 may play an importanttreated with dexamethasone were apoptotic.role in the process of negative selection.

As E2F-12/2 mice age, a second phenotype, charac-also contained a region of the thymus with altered mor- terized by a dramatic increase in thymocyte prolifera-phology indicative of a lymphoblastic lymphoma (Figure tion, is observed. This aberrant hyperproliferation oc-8C). In vivo labeling of this animal with BrdU demon- curs in the thymic cortex, but not thymic medulla orstrated that cells within the tumor were rapidly proliferat- lymph nodes, suggesting that it is due to the presenceing. None of the 10 wild-type littermates that were exam- of proliferating immature thymocytes. The finding thatined contained detectable tumors. More extensive the disruption of E2F-1 function leads to increased thy-analysis of older mice by Yamasaki et al. (1996 [this mocyte proliferation implies that E2F-1 normally func-issue of Cell]) has revealed sporadic tumorigenesis in a tions as a suppressor of cell proliferation.wide variety of tissues in E2F-12/2 animals, suggesting The fact that the decreased apoptosis detected inthat our detection of a tumor in one of the 11 older young mice and the increased cell proliferation seen inanimals that we have sacrificed may reflect a real in- older mice both were observed in the thymus suggestscrease in the rate of tumorigenesis in these mice. Con- that the two phenotypes may be mechanistically related.sistent with this possibility, we have found that as our For example, inactivation of E2F-1’s apoptotic functionE2F-12/2 mice age they die at a significantly higher rate may lead indirectly to the hyperproliferation of immaturethan their wild-type littermates. Although for many the thymocytes. Thus, the role proposed here for E2F-1 maycause of death remains to be elucidated, preliminary be analogous to that of the p53 tumor suppressor gene.evidence suggests that a number of these mice had p53 is believed to play a critical role in suppressingtumors. What we can conclude from our experiments inappropriate cell proliferation. In cells that have under-is that as E2F-12/2 mice age they exhibit a consistent gone a mutation that leads to their aberrant proliferation,phenotype of thymic hyperproliferation that at least in p53 somehow triggers cell death. In the absence of p53,one case resulted in the formation of a lymphoblastic for example in the cells of p53-deficient animals, theselymphoma. Together, these findings indicate that inappropriately proliferating cells do not die. Rather,E2F-1 functions in vivo as a suppressor of cell prolifer- they expand in number and can give rise to tumors.ation. Similarly, the observed failure of immature thymocytes in

E2F-12/2 mice toundergo normallyscheduled apoptosisDiscussion may result in the survival of an excessive number of

proliferating thymocytes as is observed in older E2F-To investigate the functions of E2F-1, we generated 12/2 mice. Interestingly, this suggests that E2F-1 may

play a direct role in regulating apoptosis independentE2F-1 deficient mice. Surprisingly, mice deficient in

Apoptosis and Proliferation in E2F-1 Deficient Mice557

Figure 8. Older E2F-12/2 Mice Display Thy-mic Hyperproliferation and Tumorigenesis

Wild-type and E2F-12/2 mice 6 to 12 monthsof age were injected with BrdU to label cellsin S phase. Organs were harvested 4 hr laterand stained to identify BrdU-incorporatingcells (stained red) as described in Experimen-tal Procedures. Asshown in (A), wild-type thy-muses (1/1, right) had few cells in S phase,while E2F-12/2 thymuses (left) had many cellsin S phase. In contrast, other organs, includ-ing gut epithelium, as shown in (B), had simi-lar numbers of cells in S phase in wild-type(1/1, right) and E2F-12/2 (left) mice. In 1 of11 older E2F-12/2 mice a tumor was present,as shown in (C). Tumor tissue is present onthe left side of this photograph, while normalthymus is seen on the right. The tumor is iden-tified as a lymphoblastic lymphoma on thebasis of standard pathologic criteria includ-ing monotony of cell morphology, nucleussize, dispersed chromatin pattern, and scantcytoplasm. Cells of this tumor are rapidly pro-liferating as demonstrated by the high frac-tion of BrdU-labeled cells (labeling indexof 41%).

of its function as a regulator of cell proliferation. Several sites during G0 or G1. This might lead to aberrant ex-pression of particular cell cycle regulatory genes andrecent findings provide support for this possibility. First,

we find that the thymuses of young E2F-12/2 mice exhibit inappropriate cell proliferation. A role for E2F-1 in thesuppression of thymocyte cell cycle progression mighta defect in apoptosis in the absence of any measurable

change in cell cycle distribution. Second, in a recent explain the aberrant hyperproliferation seen in the thy-mus of older E2F-12/2 mice.mutagenesis study E2F-1’s roles as a regulator of

apoptosis and cell cycle progression were found to be If E2F-1 functions primarily as a suppressor of cellcycle progression, then the defect in apoptosis seen inseparable (Krek et al., 1995).

Nevertheless, it remains a possibility that E2F-1 func- young E2F-12/2 mice could be an indirect consequenceof a defect in cell cycle progression. Although we de-tions primarily as a suppressor of cell proliferation. Con-

sistent with this possibility is the observation that E2F tected no change in the cell cycle distribution of thymo-cytes in young E2F-12/2 mice, we cannot exclude theDNA binding sites in the promoters of several cell cycle

genes function primarily to repress transcriptionof these possibility that there is a subtle increase in proliferationor progression through the cell cycle in the E2F-12/2genes during G0 and G1 (Weintraub et al., 1992, 1995;

Neuman et al., 1995), presumably by binding E2F-Rb cells. If it is necessary that a thymocyte be in a particularphase of the cell cycle to engage apoptosis, a changecomplexes. Recent data suggest that by repressing

transcription E2F-1–Rb complexes suppress entry into in cell cycle distribution caused by the mutation ofE2F-1 might lead indirectly to a decrease in apoptosis.S phase (Sellers et al., 1995). Thus, an E2F-1 deficient

cell may be unable to tether Rb to E2F-1 DNA binding A direct role for E2F-1 in either apoptosis or as a

Cell558

hybridization probe was prepared from a 0.9 kb SalI–EcoRI fragmentsuppressor of cell cycle progression may explain addi-of the human E2F-1 cDNA plasmid pSP72-RBAP-1 (Kaelin et al.,tional phenotypes that we and Yamasaki et al. (1996)1992) and labeled by random priming. DNA was isolated from puri-observe in the E2F-12/2 mice such as testicular atrophy,fied positives and a 7.4 kb fragment spanning the bulk of the

increased mortality, and sporadic tumor formation. For E2F-1 gene was subcloned into pBlueScript KS(1) from two inde-example, the overgrowth of interstitial Leydig cells in pendent l clones. Further restriction mapping, Southern hybridiza-the testes may be due to a failure of these cells to tion, and DNA sequencing suggested that these two fragments were

identical and represented the bona fide E2F-1 gene. To generate aundergo apoptosis or growth arrest. Similarly, sporadicgene targeting vector, a fragment containing PGK-neo was insertedtumors in the E2F-12/2 mice might be the result of aby blunt-end ligation into the XcmI site in exon 3 of E2F-1 and adefect in apoptosis or growth suppression in these cells.1.7 kb SmaI/EcoRV fragment containing E2F-1 exon 4 was excised.Although in the thymus E2F-1 plays a consistent roleThe disrupted E2F-1 genomic fragment was then inserted into a

in preventing abnormal cell proliferation, the sporadic backbone containing a PGK-TK gene to provide a negative se-nature of the abnormal proliferation seen in other cell lectable marker (Tybulewicz et al., 1991).types suggests that in these cell types random muta-tions may be required to unmask E2F-1’s role in growth

Embryonic Stem Cell Culture and Embryo Manipulationsuppression. J1 ES cell culture, electroporation, and selection of subclones were

The phenotype of the E2F-12/2 mice is surprising performed essentially as described (Li et al., 1992). Injection of ESgiven the widely held view that E2Fs function primarily cells into host blastocysts and their surgical introduction into foster

mothers were performed essentially as described (Robertson, 1987).as positive regulators of cell cycle progression and thatE2F-1 overexpression can cause oncogenesis. Our datadoes not support a unique role for E2F-1 as a promoter Southern Hybridization and Northern Hybridizationof cell proliferation. Rather, we find that E2F-1 plays a DNA isolated (Laird et al., 1991) from the offspring of chimeras

and subsequent generations was digested with EcoRV (or othercritical role as a positive regulator of apoptosis and arestriction enzymes) and used in standard Southern blots (Churchsuppressor of cell proliferation. How can these roles beand Gilbert, 1984) using a probe prepared from E2F-1 genomicreconciled with the observed ability of E2F-1 to drivesequences to determine the genotypes of the mice.cell cycle progression and oncogenesis when overex-

For Northern hybridization, embryonic fibroblasts were isolatedpressed? It is likely that the level of E2F-1 expression by standard methods (Robertson, 1987), grown to 75% confluence,is important. Under normal circumstances, in G0 and and arrested in early S phase by treatment with 100 mM hydroxyureaG1 E2F-1 may be completely bound up by Rb so that for 16 hr. RNA was isolated (Chomczynski and Sacchi, 1987) and

used in standard Northern hybridization (Church and Gilbert, 1984)it functions primarily as a repressor of transcription andusing a probe prepared from a 0.9 kb SalI–EcoRI fragment of thea suppressor of cell proliferation. When E2F-1 is overex-human E2F-1 cDNA (Kaelin et al., 1992) or a 1.3 kb probe preparedpressed there may not be sufficient Rb present in thefrom a PstI restriction fragment of the rat glyceraldehyde-3-phos-cell to bind up all the E2F-1. Under such a circumstance,phate dehydrogenase cDNA (Fort et al., 1985).

free E2F would be present and might be capable ofpromoting inappropriate entry into S phase and ulti-

Isolation of Primary Embryonic Fibroblastsmately oncogenesis. Since under normal circumstancesand Measurement of GrowthE2F-1 may have a dual role as a supressorof cell prolifer-Primary embryonic fibroblasts were harvested from 13.5- or 14.5-

ation in G0/G1 and an activator of proliferation in G1/S, day-old embryos using the protocol of Robertson (1987) with minorthe effect of a mutation in E2F-1 might be difficult to modifications. Doubling times were measured by plating fibroblastspredict. The fact that a deletion of E2F-1 leads to a on multipledishes, trypsinizing, and countingcell numbers on paired

dishes after one, two, or three days of culture. Doubling time wasdecrease in apoptosis and enhanced proliferation sug-calculated from the slope of the best fit line of logarithm of cellgests that in vivo the negative effects of E2F-1 on cellnumber as a function of time. Data were pooled from multipleexperi-cycle progression may be more important than the posi-ments comparing in total fibroblasts independently isolated fromtive effects. Nevertheless, the fact that E2F-1 is a mem-four wild-type and four E2F-12/2 mouse embryos.

ber of a large family of related genes leaves open thepossibility that E2F-1 also plays an important role in

Harvest of Thymocytes and Lymph Node Cellspromoting S phase entry in many cell types but that inThymuses or lymph nodes were mechanically dissociated betweenits absence this role is compensated for by other familytwo pieces of ground glass. Debris was allowed to settle, the cells

members. washed in medium (Iscove’s modified Dulbecco’s medium), andA more thorough understanding of the multiple over- contaminating erythrocytes removed by hypotonic lysis (155 mM

lapping functions of different E2F family members must NH4Cl/10 mM KHCO3/0.1 mM EDTA [pH 7.3] on ice for 5 min). Thecells were washed again in medium and then used in subsequentawait the production of mice containing mutations ofexperiments.other E2F family members, either singly or in combina-

tion. Nevertheless, the generation of E2F-1 deficientmice is a start in this direction and has provided new Cell Cycle Analysis

Cells were pulse labeled with 10 mM BrdU for 1 hr, washed in ice-insight into the function of the E2F family. Somewhatcold phosphate-buffered saline (PBS), and fixed in ice-cold 70%unexpectedly the analysis of this animal, taken togetherethanol. One to ten days later, cells were processed for flow cytome-with previous studies, has revealed that a single gene,tric analysis of DNA content (staining with propidium iodide) anddepending on the circumstance, can function both asDNA synthesis (staining with fluorescein isothiocyanate–conjugated

an oncogene and as a growth suppressor gene. antibody to BrdU) according to a protocol from the supplier of theantibody (Becton-Dickinson).

Experimental Procedures

In Vivo S Phase LabelingCloningMice were injected intraperitoneally with BrdU in PBS at a dose ofA murine E2F-1 genomic clone was isolated from a 129/Sv genomic50 mg per gram of body weight. Mice were sacrificed 4 hr later, andlibrary in lFIXII (Stratagene) screened by hybridization using Gene-

Screen (NEN) according to the instructions of the manufacturer. The organs harvested and fixed in 10% formalin. Slides of tissue sections

Apoptosis and Proliferation in E2F-1 Deficient Mice559

were stained with antibody to BrdU and processed using the Vecta- Blake, M.C., and Azizkhan, J.C. (1989). Transcription factor E2F isrequired for efficient expression of the hamster dihydrofolate reduc-Stain ABC alkaline phosphatase immunostaining kit (Vector Labora-

tories) according to the instructions of the manufacturer. Sections tase gene in vitro and in vivo. Mol. Cell. Biol. 9, 4994–5002.were counterstained with hematoxylin. Cao, L., Faha, B., Dembski, M., Tsai, L.H., Harlow, E., and Dyson,

N. (1992). Independent binding of the retinoblastoma protein andFlow Cytometry and Fluorescence-Activated Cell Sorting p107 to the transcription factor E2F. Nature 355, 176–179.Cells were suspended in blocking buffer (3% fetal bovine serum, Chellappan, S.P., Hiebert, S., Mudryj, M.,Horowitz, J.M., and Nevins,1% bovine serum albumin in PBS) and mixed with a fluorescein J.R. (1991). The E2F transcription factor is a cellular target for theisothiocyanate–conjugated antibody (either anti-CD8 [clone 53-6.7], RB protein. Cell 65, 1053–1061.anti-CD3e [clone 145-2C11], or anti-Thy1.2 [clone 30-H12]) and an

Chittenden, T., Livingston, D.M., and Kaelin, W.G., Jr. (1991). TheR-phycoerythrin-conjugated antibody (either anti-CD4 [cloneT/E1A-binding domain of the retinoblastoma product can interactH129.19], anti-CD8 [clone 53–6.7], or anti-B220 [clone RA3–6B2], allselectively with a sequence-specific DNA-binding protein. Cell 65,purchased from Pharmingen). After 15 min, labeled cells were1073–1082.washed twice in blocking buffer, once in PBS, and fixed in freshChomczynski, C., and Sacchi, N. (1987). Single-step method of RNA1% paraformaldehyde in PBS for flow cytometry or resuspended inisolation by acid guanidinium thiocyanate-phenol-chloroform ex-medium for fluorescence-activated cell sorting. Appropriate singletraction. Anal. Biochem. 162, 156–159.color controls were used to set flow cytometer compensation and

quadrants. Church, G.M., and Gilbert, W. (1984). Genomic sequencing. Proc.Natl. Acad. Sci. USA 81, 1991–1995.

In Vitro Apoptosis Cobrinik, D., Whyte, P., Peeper, D.S., Jacks, T., and Weinberg, R.A.Thymocytes or lymph node cells were plated in 96-well plates at a (1993). Cell cycle-specific association of E2F with the p130 E1A-concentration of 5 3 105 cellsper 100 ml in Iscove’s modified Dulbec- binding protein. Genes Dev. 7, 2392–2404.co’s medium supplemented with 10% heat-inactivated fetal bovine Cress, W.D., Johnson,D.G., and Nevins, J.R. (1993). Agenetic analy-serum, 0.5 mM b-mercaptoethanol, and 1 mM HEPES (pH 7.4). Im- sis of the E2F1 gene distinguishes regulation by Rb, p107, andmediately after plating, or at various time intervals, aliquots from adenovirus E4. Mol. Cell. Biol. 13, 6314–6325.replicate wells were mixed with an equal volume of 0.4% trypan

DeGregori, J., Kowalik, T., and Nevins, J.R. (1995). Cellular targetsblue, and the concentrations of live and dead cells counted on afor activation by the E2F1 transcription factor include DNA synthe-hemacytometer. In situ nick translation performed by the methodsis- and G1/S-regulatory genes. Mol. Cell. Biol. 15, 4215–4224.of Gold et al. (1993) was used to verify that DNA fragmentation wasDevoto, S.H., Mudryj, M., Pines, J., Hunter, T., and Nevins, J.R.an early event in cell death.(1992). A cyclin A–protein kinase complex possesses sequence-specific DNA binding activity: p33cdk2 is a component of the E2F–In Vivo Apoptosiscyclin A complex. Cell 68, 167–176.Antibody to CD3e (clone 145–2C11, no azide, low endotoxin formula-

tion, purchased from Pharmingen) diluted in PBS was injected intra- Dou, Q.P., Markell, P.J., and Pardee, A.B. (1992). Thymidine kinaseperitoneally at a dose of 1.9 to 2.5 mg per gram body weight (in a transcription is regulated at G1/S phase by a complex that containsgiven experiment all mice were injected at the same dose). Control retinoblastoma-like protein and a cdc2 kinase. Proc. Natl. Acad. Sci.mice were injected with a similar volume (0.2 ml) of PBS. Thymocytes USA 89, 3256–3260.were isolated 16 hr later and processed for TUNEL (Gavrieli et al., Duronio, R.J., and O’Farrell, P.H. (1995). Developmental control of1992) according to the instructions for the fluorescein Apoptag kit the G1 to S transition in Drosophila: cyclin Eis a limiting downstream(Oncor). Compensation and gates for flow cytometry were set using target of E2F. Genes Dev. 9, 1456–1468.appropriate controls. For negative controls TUNEL was performed

Duronio, R.J., O’Farrell, P.H., Xie, J.E., Brook, A., and Dyson, N.omitting the terminal deoxynucleotidyl transferase. As positive con-

(1995). The transcription factor E2F is required for S phase duringtrols, thymocytes were treated with 1 mM dexamethasone for 4 hr

Drosophila embryogenesis. Genes Dev. 9, 1445–1455.in vitro, or fixed thymocytes were treated with DNaseI.

Dynlacht, B.D., Flores, O., Lees, J.A., and Harlow, E. (1994). Differen-tial regulation of E2F transactivation by cyclin/cdk2 complexes.AcknowledgmentsGenes Dev. 8, 1772–1786.

Fort, P., Marty, L., Piechaczyk,M., ElSabrouty, S., Dani, C., Jeanteur,Correspondence should be addressed to Michael E. Greenberg.P., and Blanchard, J.M. (1985). Various rat adult tissues express onlyWe note that Lili Yamasaki, Tyler Jacks, Nicholas Dyson, andone major mRNA species from the glyceraldehyde-3-phosphate-Edward Harlow have also generated E2F-1 mutant mice and havedehydrogenase multigenic family. Nucleic Acids Res. 13, 1431–kindly exchanged data with us during the preparation of our manu-1442.scripts. In particular, they alerted us to the presence of testicular

atrophy in older E2F-12/2 mice and shared with us their extensive Fowlkes, B.J., Schwartz, R.H., and Pardoll, D.M. (1988). Deletionstatistics on the occurrence of sporadic tumors in the E2F-12/2 mice. of self-reactive thymocytes occurs at a CD4181 precursor stage.We thank Sallie Smith, Barbara Osborne, and members of the Nature 334, 620–623.Greenberg laboratory for helpful discussions; Susan Kim and Hong Gavrieli, Y., Sherman, Y., and Ben-Sasson, S.A. (1992). IdentificationYe for excellent technical assistance; and David Zurakowski for of programmed cell death in situ via specific labeling of nuclearadvice on statistics. This research was supported by National Insti- DNA fragmentation. J. Cell Biol. 119, 493–501.tutes of Health grant DK49216 awarded to M.E. G. as part of a Center

Gold, R., Schmied, M., Rothe, G., Zischler, H., Breitschopf, H., Wek-of Excellence in Molecular Hematology. S. H. O. is an Investigator oferle, H., and Lassmann, H. (1993). Detection of DNA fragmentationthe Howard Hughes Medical Institute.in apoptosis: application of in situ nick translation to cell culturesystems and tissue sections. J. Histochem. Cytochem. 41, 1023–Received February 22, 1996; revised March 27, 1996.1030.

Helin, K., Lees, J.A., Vidal, M., Dyson, N., Harlow, E., and Fattaey,ReferencesA. (1992). A cDNA encoding a pRB-binding protein with properties

Adams, P.D., and Kaelin, W.G., Jr. (1995). Transcriptional control by of the transcription factor E2F. Cell 70, 337–350.E2F. Semin. Cancer Biol. 6, 99–108. Hiebert, S.W., Lipp, M., and Nevins, J.R. (1989). E1A-dependentBagchi, S., Weinmann, R., and Raychaudhuri, P. (1991). The retino- trans-activation of the human MYC promoter is mediated by theblastoma protein copurifies with E2F-I, an E1A-regulated inhibitor E2F factor. Proc. Natl. Acad. Sci. USA 86, 3594–3598.of the transcription factor E2F. Cell 65, 1063–1072. Huang, M.M., and Hearing, P. (1989). The adenovirus early region

4 open reading frame 6/7 protein regulates the DNA binding activityBandara, L.R., Adamczewski, J.P., Hunt, T., and La Thangue, N.B.(1991). Cyclin A and the retinoblastoma gene product complex with of the cellular transcription factor, E2F, through a direct complex.

Genes Dev. 3, 1699–1710.a common transcription factor. Nature 352, 249–251.

Cell560

Johnson, D.G., Schwarz, J.K., Cress, W.D., and Nevins, J.R. (1993). polymerase a gene: sequences controlling expression in cyclingExpression of transcription factor E2F1 induces quiescent cells to and serum-stimulated cells. Mol. Cell. Biol. 11, 2081–2095.enter S phase. Nature 365, 349–352. Qin, X.Q., Livingston, D.M., Kaelin, W.G., Jr., and Adams, P.D. (1994).Johnson, D.G., Cress, W.D., Jakoi, L., and Nevins, J.R. (1994). Onco- Deregulated transcription factor E2F-1 expression leads to S-phasegenic capacity of the E2F1 gene. Proc. Natl. Acad. Sci. USA 91, entry and p53-mediated apoptosis. Proc. Natl. Acad. Sci. USA 91,12823–12827. 10918–10922.Kaelin, W.G., Jr., Krek, W., Sellers, W.R., DeCaprio, J.A., Ajchen- Robertson,E.J. (1987). Embryo-derived stem cells. In Teratocarcino-baum, F., Fuchs, C.S., Chittenden, T., Li, Y., Farnham, P.J., Blanar, mas and Embryonic Stem Cells: a Practical Approach, E.J. Robert-M.A., et al. (1992). Expression cloning of a cDNA encoding a retino- son, ed. (Oxford: IRL Press), pp. 104–108.blastoma-binding protein with E2F-like properties. Cell 70, 351–364.

Sala, A., Nicolaides, N.C., Engelhard, A., Bellon, T., Lawe, D.C.,Kisielow, P., and von Boehmer, H. (1991). Kinetics of negative and

Arnold, A., Grana, X., Giordano, A., and Calabretta, B. (1994). Corre-positive selection in the thymus. Adv. Exp. Med. Biol. 292, 31–42.lation between E2F-1 requirement in the S phase and E2F-1 trans-

Kowalik, T.F., DeGregori, J., Schwarz, J.K., and Nevins, J.R. (1995). activation of cell cycle-related genes in human cells. Cancer Res.E2F1 overexpression in quiescent fibroblasts leads to induction of 54, 1402–1406.cellular DNA synthesis and apoptosis. J. Virol. 69, 2491–2500.

Scollay, R., Wilson, A., D’Amico, A., Kelly, K., Egerton, M., Pearse,Krek, W., Ewen, M.E., Shirodkar, S., Arany, Z., Kaelin, W.G., Jr., and M., Wu, L., and Shortman, K. (1988). Developmental status andLivingston, D.M. (1994). Negative regulation of the growth-promot-

reconstitution potential of subpopulations of murine thymocytes.ing transcription factor E2F-1 by a stably bound cyclin A-dependentImmunological Rev. 104, 81–120.protein kinase. Cell 78, 161–172.Sellers, W., Rodgers, J., and Kaelin, W. (1995). A potent transrepres-Krek, W., Xu, G., and Livingston,D. (1995). Cyclin A-kinase regulationsion domain in the retinoblastoma protein induces a cell cycle arrestof E2F-1 DNA binding function underlies suppression of an S phasewhen bound to E2F sites. Proc. Natl. Acad. Sci. USA 92, 11544–checkpoint. Cell 83, 1149–1158.11548.

Laird, P.W., Zijderveld, A., Linders, K., Rudnicki, M.A., Jaenisch, R.,Shan, B., and Lee, W.H. (1994). Deregulated expression of E2F-1and Berns, A. (1991). Simplified mammalian DNA isolation proce-induces S-phase entry and leads to apoptosis. Mol. Cell. Biol. 14,dure. Nucleic Acids Res. 19, 4293.8166–8173.La Thangue, N.B. (1994). DRTF1/E2F: an expanding family of hetero-

dimeric transcription factors implicated in cell-cycle control. Trends Shan, B., Zhu, X., Chen, P.L., Durfee, T., Yang, Y., Sharp, D., andBiochem. Sci. 19, 108–114. Lee, W.H. (1992). Molecular cloning of cellular genes encoding reti-

noblastoma-associated proteins: identification of a gene with prop-Lees, E., Faha, B., Dulic, V., Reed, S.I., and Harlow, E. (1992). Cyclinerties of the transcription factor E2F. Mol. Cell. Biol. 12, 5620–5631.E/cdk2 and cyclin A/cdk2 kinases associate with p107 and E2F in

a temporally distinct manner. Genes Dev. 6, 1874–1885. Shi, Y.F., Bissonnette, R.P., Parfrey, N., Szalay, M., Kubo, R.T., andLees, J.A., Saito, M., Vidal, M., Valentine, M., Look, T., Harlow, E., Green, D.R. (1991). In vivo administration of monoclonal antibodiesDyson, N., and Helin, K. (1993). The retinoblastoma protein binds to the CD3 T cell receptor complex induces cell death (apoptosis)to a family of E2F transcription factors. Mol. Cell. Biol. 13, 7813–7825. in immature thymocytes. J. Immunol. 146, 3340–3346.

Li, E., Bestor, T.H., and Jaenisch, R. (1992). Targeted mutation of Shirodkar, S., Ewen, M., DeCaprio, J.A.,Morgan, J., Livingston, D.M.,the DNA methyltransferase gene results in embryonic lethality. Cell and Chittenden, T. (1992). The transcription factor E2F interacts with69, 915–926. the retinoblastoma product and a p107–cyclin A complex in a cell

cycle–regulated manner. Cell 68, 157–166.Li, Y., Slansky, J.E., Myers, D.J., Drinkwater, N.R., Kaelin, W.G., andFarnham, P.J. (1994). Cloning, chromosomal location, and charac- Smith, C.A., Williams, G.T., Kingston, R., Jenkinson, E.J., and Owen,terization of mouse E2F1. Mol. Cell. Biol. 14, 1861–1869. J.J. (1989). Antibodies to CD3/T-cell receptor complex induce deathMotokura, T., and Arnold, A. (1993). PRAD1/cyclin D1 proto-onco- by apoptosis in immature T cells in thymic cultures. Nature 337,gene: genomic organization, 59 DNA sequence, and sequence of a 181–184.tumor-specific rearrangement breakpoint. Genes Chromosom. Can-

Thalmeier, K., Synovzik, H., Mertz, R., Winnacker, E.L., and Lipp,cer 7, 89–95.M. (1989). Nuclear factor E2F mediatesbasic transcription and trans-

Mudryj, M., Hiebert, S.W., and Nevins, J.R. (1990). A role for the activation by E1a of the human MYC promoter. Genes Dev. 3,adenovirus inducible E2F transcription factor in a proliferation de- 527–536.pendent signal transduction pathway. EMBO J. 9, 2179–2184.

Tybulewicz, V.L., Crawford, C.E., Jackson, P.K., Bronson, R.T., andNeuman, E., Flemington, E.K., Sellers, W.R., and Kaelin, W.G., Jr.Mulligan, R.C. (1991). Neonatal lethality and lymphopenia in mice(1995). Transcription of the E2F-1 gene is rendered cell cycle depen-with a homozygous disruption of the c-abl proto-oncogene. Cell 65,dent by E2F DNA-binding sites within its promoter. Mol. Cell. Biol.1153–1163.15, 4660.Weinberg, R.A. (1995). The retinoblastoma protein and cell cycleOgris, E., Rotheneder, H., Mudrak, I., Pichler, A., and Wintersberger,control. Cell 81, 323–330.E. (1993). A binding site for transcription factor E2F is a target for

trans activation of murine thymidine kinase by polyomavirus large Weintraub, S.J., Prater, C.A., and Dean, D.C. (1992). RetinoblastomaT antigen and plays an important role in growth regulation of the protein switches the E2F site from positive to negative element.gene. J. Virol. 67, 1765–1771. Nature 358, 259–261.Owen, J.J., Owen, M.J., Williams, G.T., Kingston, R., and Jenkinson, Weintraub, S.J., Chow, K.N., Luo, R.X., Zhang, S.H., He, S., andE.J. (1988). The effects of anti-CD3 antibodies on the development Dean, D.C. (1995). Mechanism of active transcriptional repressionof T-cell receptor ab1 lymphocytes in embryonic thymus organ cul-

by the retinoblastoma protein. Nature 375, 812–815.tures. Immunology 63, 639–642.

Wu, X., and Levine, A.J. (1994). p53 and E2F-1 cooperate to mediatePagano, M., Draetta, G., and Jansen-Durr, P. (1992a). Associationapoptosis. Proc. Natl. Acad. Sci. USA 91, 3602–3606.of cdk2 kinase with the transcription factor E2F during S phase.

Science 255, 1144–1147. Xu, M., Sheppard, K.A., Peng, C.Y., Yee, A.S., and Piwnica-Worms,H. (1994). Cyclin A/CDK2 binds directly to E2F-1 and inhibits thePagano, M., Pepperkok, R., Verde, F., Ansorge, W., and Draetta, G.DNA-binding activity of E2F-1/DP-1 by phosphorylation. Mol. Cell.(1992b). Cyclin A is required at two points in the human cell cycle.Biol. 14, 8420–8431.EMBO J. 11, 961–971.

Pearson, B.E., Nasheuer, H.P., and Wang, T.S. (1991). Human DNA Xu, G., Livingston, D.M., and Krek, W. (1995). Multiple members of

Apoptosis and Proliferation in E2F-1 Deficient Mice561

the E2F transcription factor family are the products of oncogenes.Proc. Natl. Acad. Sci. USA 92, 1357–1361.

Yamasaki, L., Tyler, J., Roderick, B., Goillot, E., Harlow, E., andDyson, N.J. (1996). Tumor induction and tissue atrophy in micelacking E2F-1. Cell 85, this issue.

Yang, X.H., and Sladek, T.L. (1995). Overexpression of the E2F-1transcription factor gene mediates cell transformation. Gene Expr.4, 195–204.