The proliferative and apoptotic activities of E2F1 in the mouse retina

Page created by Leon Campos
 
CONTINUE READING
The proliferative and apoptotic activities of E2F1 in the mouse retina
Oncogene (2001) 20, 7073 ± 7084
                                                                 ã 2001 Nature Publishing Group All rights reserved 0950 ± 9232/01 $15.00
                                                                 www.nature.com/onc

The proliferative and apoptotic activities of E2F1 in the mouse retina

Suh-Chin J Lin1, Stephen X Skapek2, David S Papermaster3, Mark Hankin4 and Eva Y-HP Lee*,1
1
 Department of Molecular Medicine/Institute of Biotechnology, University of Texas Health Science Center at San Antonio, Texas,
TX 78245-3207, USA; 2Department of Hematology/Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee,
TN 38105, USA; 3Department of Neuroscience, University of Connecticut Health Science Center, Farmington, Connecticut,
CT 06030-3401, USA; 4Department of Anatomy and Neurobiology, Medical College of Ohio, Toledo, Ohio, OH 43614, USA

The E2F1 transcription factor controls cell proliferation        (Adams and Kaelin, 1995; Slansky and Farnham,
and apoptosis. E2F1 activity is negatively regulated by          1996). Genetic studies in both Drosophila (Duronio et
the retinoblastoma (RB) protein. To study how inactiva-          al., 1995) and mammalian cells (Ishizaki et al., 1996;
tion of Rb and dysregulated E2F1 a€ects the developing           Wu et al., 1996) demonstrate that E2F activity is
retina, we analysed wild-type and Rb7/7 embryonic                required for cell proliferation. In addition, overexpres-
retinas and retinal transplants and we established               sion of E2F1 has been shown to be sucient to drive
transgenic mice expressing human E2F1 in retinal                 quiescent cells into S phase in vitro (Johnson et al.,
photoreceptor cells under the regulation of the IRBP             1993). E2F1 cooperates with activated Ras to trans-
promoter (TgIRBPE2F1). A marked increase in cell                 form ®broblasts (Johnson et al., 1994; Singh et al.,
proliferation and apoptosis was observed in the retinas of       1994; Xu et al., 1995). In mice, expression of E2F1 in
Rb7/7 mice and TgIRBPE2F1 transgenic mice. In the                the presence of activated Ras results in skin tumor
transgenic mice, photoreceptor cells formed rosette-like         formation (Pierce et al., 1998a). Interestingly, in
arrangements at postnatal days 9 through 28. Complete            addition to its role in cell proliferation, overexpression
loss of photoreceptors followed in the TgIRBPE2F1 mice           of E2F1 but not other E2F proteins also triggers
but not in the Rb7/7 retinal transplants. Both RB-               apoptosis in cells. In the absence of p53, E2F1-induced
de®cient and E2F1-overexpressing photoreceptor cells             apoptosis is signi®cantly reduced (DeGregori et al.,
expressed rhodopsin, a marker of terminal di€erentia-            1997; Kowalik et al., 1995; Qin et al., 1994; Shan and
tion. Loss of p53 partially reduced the apoptosis and            Lee, 1994; Wu and Levine, 1994). This apoptosis-
resulted in transient hyperplasia of multiple cell types in      promoting property of E2F1 likely contributes to the
the TgIRBPE2F1 retinas at postnatal day 6. Our                   phenotype of E2F1 de®cient mice, which are defective
®ndings support the concept that cross-talk occurs               in thymocyte apoptosis and predisposed to tumors at
between di€erent retinal cell types and that multiple            an advanced age (Field et al., 1996; Yamasaki et al.,
genetic pathways must become dysregulated for the full           1996).
oncogenic transformation of neuronal retinal cells.                 An understanding of how E2F activity may be
Oncogene (2001) 20, 7073 ± 7084.                                 regulated in mammalian cells ®rst came from the
                                                                 ®ndings of a functional interaction between E2F1 and
Keywords: E2F1; p53; RB and retinoblastoma                       the protein product of the retinoblastoma susceptibility
                                                                 gene Rb (reviewed in Dyson, 1998). RB may block the
                                                                 transcriptional activity of E2F1 by direct binding to its
Introduction                                                     transactivation domain and preventing its interaction
                                                                 with other components of the transcriptional machin-
The E2F transcription factor was initially identi®ed as          ery (Ross et al., 1999). In addition, the presence of
a cellular DNA-binding activity required for the                 histone deacetylases in the E2F/RB complexes could
adenovirus E1A-mediated activation of the viral E2               result in transcription repression by a mechanism
gene (reviewed in Dyson, 1998). E2F functions as a               involving chromatin regulation (Brehm and Kouzar-
heterodimer consisting of an E2F family member,                  ides, 1999). Finally, other RB-interacting proteins have
E2F1 through 6, and a DP protein, DP1 and DP2                    been identi®ed that could mediate transcriptional
(reviewed in Dyson, 1998). E2F can bind to speci®c               repression of E2F-dependent genes (Skapek et al.,
regulatory elements in genes required for DNA                    2000). The physical interaction between RB and E2F is
synthesis including DNA polymerase a, thymidine                  detected during the G1 phase of the cell cycle. Upon
kinase and dihydrofolate reductase; and cell cycle               phosphorylation of RB by cyclin-dependent kinases,
regulators including cyclin A, cyclin E and cdc2                 RB, E2F, and other proteins found in this complex
                                                                 (like HDAC-1) dissociate to allow the activation of the
                                                                 E2F responsive genes (reviewed in Dyson, 1998; Zhang
*Correspondence: EY-HP Lee; E-mail: Leee@uthscsa.edu
                                                                 et al., 1999).
Received 11 August 2000; revised 23 August 2001; accepted 23        Two Rb-related genes p107 and p130 have also been
August 2001                                                      characterized (reviewed in Mulligan and Jacks, 1998).
The proliferative and apoptotic activities of E2F1 in the mouse retina
E2F1 and p53 inactivation in retinoblastoma
                                                                    S-CJ Lin et al
7074
           Protein products of both genes interact with E2F and                      regulation of E2F1 and loss of the p53 tumor
           can act as negative regulators of cell proliferation.                     suppressor contribute, individually or in combination,
           Unlike Rb, however, mutations of p107 and p130 are                        to photoreceptor development and tumor formation.
           rarely found in human tumors and mutation of either
           p107 or p130 in mice leads to di€erent phenotypes
           (reviewed in Lin et al., 1996; Mulligan and Jacks,
                                                                                     Results
           1998). The di€erent biological functions of di€erent
           RB family members may be due to their association
                                                                                     Proliferation and apoptosis in Rb-deficient retinas
           with speci®c E2Fs (reviewed in Dyson, 1998; Nevins,
           1998). RB interacts with E2F1, E2F2 and E2F3 during                       There is aberrant proliferation and extensive apoptosis
           late G1 to S phase. In contrast, p107 preferentially                      in the developing nervous system of RB-de®cient
           interacts with E2F4 in proliferating cells during                         embryos (Lee et al., 1994; Macleod et al., 1996).
           S phase whereas p130 binds to E2F4 and E2F5                               How RB de®ciency a€ects retinal development is not
           primarily in G0 phase. Despite these distinctions,                        known, in part due to embryonic lethality of Rb7/7
           recent studies of mice lacking more than one member                       mice around E12.5 ± E14.5 (Lee et al., 1992). To begin
           of the Rb gene family have provided evidence that Rb,                     to study the e€ects of loss of RB in the retina, we
           p107 and p130 have overlapping as well as distinct                        analysed retinal cell proliferation and apoptosis in
           function (see below).                                                     retinas from E13.5 wild-type and Rb7/7 embryos. In
              That the loss of Rb causes retinoblastoma in humans                    normal embryos, the inner layers of the retinal
           has been well established. On the other hand, how loss                    ventricular cells did not incorporate BrdU at E13.5
           of Rb a€ects the retina in mouse models is more                           (Figure 1Aa). In contrast, in Rb7/7 embryos, many
           confusing. Homozygous Rb7/7 mouse embryos show                            cells in the same region continued to enter the cell cycle
           ectopic proliferation and excess apoptosis of the                         (Figure 1Ab). In wild-type embryos, little TUNEL
           nervous system and die around embryonic day 14.5                          staining was observed in the normal E13.5 retina
           (E14.5) (Clarke et al., 1992; Jacks et al., 1992; Lee et                  (Figure 1Ac), but TUNEL-stained cells were readily
           al., 1992). Heterozygous Rb+/7 mice develop pituitary                     detected in Rb7/7 retina (Figure 1Ad). Examination of
           and thyroid tumors (Hu et al., 1994; Jacks et al., 1992;                  cell proliferation and apoptosis at di€erent embryonic
           Nikitin and Lee, 1996) and all tumors harbor                              stages (E11.5 ± E15.5) demonstrated that ectopic BrdU-
           mutations of the wild-type Rb allele (Hu et al., 1994;                    positive cells appeared in the retina before TUNEL-
           Nikitin and Lee, 1996); however, retinoblastoma does                      stained cells (data not shown), a ®nding that is similar
           not occur. In contrast, multiple dysplastic lesions                       to ®ndings in the spinal cord and dorsal root ganglia of
           develop in the photoreceptor cell layer of the retina                     Rb7/7 embryos (Lee et al., 1994). These data indicate
           of Rb+/7; p1077/7 mice but not in Rb+/7 or p1077/7                        that RB loss causes excessive ectopic cell proliferation
           mice (Lee et al., 1996). Retinoblastomas of amacrine                      and apoptosis in embryonic retinal progenitor layers of
           cell origin were observed in chimeric mice harboring                      the mouse retina.
           Rb7/7; p1077/7 cells (Robanus-Maandag et al., 1998).                         In addition to regulating cell proliferation and
           Thus, it seems that dysfunction of both RB and p107                       apoptosis, RB is known to facilitate the di€erentiation
           are required for retinal tumor formation in mice.                         of certain types of cells (reviewed in Lipinski and
              One interpretation of these collective data is that loss               Jacks, 1999). Therefore, we sought to determine how
           of RB and RB-related proteins releases certain E2Fs                       loss of RB a€ected the di€erentiation of murine
           from suppression to enhance cell proliferation and                        photoreceptors. In wild-type mice, retina development
           tumor formation. Consistent with this, inactivation of                    continues after birth. Because loss of RB causes
           E2F1 reduces tumorigenesis in Rb+/7 mice and extends                      embryonic lethality, to fully address the e€ect of RB-
           the survival of Rb+/7 mice and Rb7/7 embryos (Tsai                        de®ciency on retinal development, we analysed retinal
           et al., 1998; Yamasaki et al., 1998). Furthermore, E2F1                   tissue that had been transplanted into normal neonatal
           de®ciency rescues cell death in Rb7/7 embryos in                          mice. It has previously been shown that transplanted
           tissues undergoing p53-dependent apoptosis (Macleod                       embryonic retinal tissue continues to develop and form
           et al., 1996; Morgenbesser et al., 1994) including the                    laminar structures (Hankin et al., 1993). Following
           ocular lens and the central nervous system (Tsai et al.,                  transplantation of retinas from E11.5 ± E12.5 embryos
           1998). Taken together, these data provide biochemical                     into wild-type or Rb+/7 neonates, the transplanted
           and genetic support for the regulatory role of RB on                      retinal tissues formed either sheet-like structures or
           E2F1 activities.                                                          rosette structures characterized by inside-out, con-
              We undertook the present series of experiments to                      centric foldings of the donor tissue (Figure 1B). The
           study the e€ects of dysregulated E2F1 activity in                         multi-layered structures were evident in both wild-type
           photoreceptors. We accomplished this by studying RB-                      and Rb-de®cient retinal transplants (Figure 1Ba,b). At
           de®cient photoreceptors in Rb7/7 mouse embryos and                        the cellular level, both wild-type and Rb7/7 retino-
           following transplantation of Rb7/7 retinal cells into                     blasts matured and di€erentiated into photoreceptors
           normal recipient mice. In addition, we have established                   as evidenced by expression of opsin (Figure 1Be,f).
           a transgenic mouse model in which E2F1 is constitu-                       Interestingly, using this model of retina development,
           tively expressed in maturing photoreceptors. We have                      we con®rmed that RB loss causes excess apoptosis.
           used these studies to further understand how loss of                      Whereas only a few apoptotic cells were seen in wild-
Oncogene
E2F1 and p53 inactivation in retinoblastoma
                                                                         S-CJ Lin et al
                                                                                                                                               7075

    Figure 1 E€ects of RB de®ciency on proliferation, apoptosis and di€erentiation of retinal cells. (A) Enhanced cell proliferation and
    cell death in the retina of Rb7/7 embryos. Pregnant mice were injected with BrdU. BrdU uptake in the retinas of E13.5 embryos
    was compared (a and b). Apoptotic cells were shown using TUNEL assay (c and d). Arrows indicate ectopic BrdU-positive cells and
    arrowheads indicate apoptotic cells. Scale bar, 100 mm. (B) Cell death and di€erentiation in the photoreceptor cells of retinal
    transplants. Retinal tissues of E11.5 ± 12.5 embryos were transplanted intracranially into neonatal Rb+/7 or wild-type mice. Sections
    of retinal transplants were stained with H&E (a and b), analysed for apoptosis using the TUNEL assay (c and d), or immunostained
    with anti-opsin antibodies (e and f). Arrowheads indicate apoptotic nuclei; arrows indicate opsin expression in the inner/outer
    segments of the photoreceptor cells. ONL, outer nuclear layer. Scale bar, (a,b) 100 mm, (c,d) 40 mm, (e,f) 15.6 mm. Left panel, wild-
    type embryos; right panel, Rb7/7 embryos

type transplants, abundant apoptotic cells were found                    these results demonstrate that RB may be required for
in Rb7/7 transplants (Figure 1Bc,d). Taken together,                     ecient cell cycle arrest and prevention of apoptosis in
                                                                                                                                            Oncogene
E2F1 and p53 inactivation in retinoblastoma
                                                                   S-CJ Lin et al
7076
           cells in the developing mouse retina, but is not required                di€erentiating photoreceptors that normally would
           for the expression of opsin in photoreceptors.                           have excited the cell cycle.

           Generation of TgIRBPE2F1 transgenic mice                                 Apoptosis of the neuroretinal cells in TgIRBPE2F1
                                                                                    transgenic mice
           Because the loss of RB appears to alter retinal
           development in these mouse transplant experiments,                       Despite the increased proliferation observed in retinas
           and RB is known to regulate E2F1, we established a                       from TgIRBPE2F1 mice, there were fewer photorecep-
           transgenic mouse model to study the e€ect of enhanced                    tor cells in the retinas in adult transgenic mice (Figure
           expression of E2F1 in retinas in vivo. To do this, we                    5f, discussed below). The decreased number of
           generated transgenic mice expressing human E2F1                          photoreceptor cells in the retina of the transgenic mice
           (hE2F1) cDNA under the regulation of IRBP                                suggests that overexpression of E2F1 may also result in
           promoter (TgIRBPE2F1 mice). This promoter directs                        cell death. To better characterize the mechanism by
           the expression of the transgene in retinal photoreceptor                 which the photoreceptors were lost, the degree of
           cells and pinealocytes (Liou et al., 1990). Several                      apoptosis was determined during the early stages of
           founder mice were identi®ed using PCR and Southern                       retinal development. In retinas from wild-type mice,
           analyses (data not shown) and two hE2F1 transgenic                       apoptotic cells were only occasionally observed during
           lines (IRBP-E37 and -E45) were established.                              the major period of retinal di€erentiation (Figure
              To con®rm that the transgene led to ectopic                           3A,B; Young, 1984). By contrast, abundant apoptotic
           expression of E2F1, we performed immunohistochem-                        cells were found throughout the ventricular layer at all
           ical analyses. In normal mice, E2F1 was detectable in                    stages in the transgenic mice with greatest apoptosis at
           the retina of E18.5 embryos (data not shown), but was                    P4 and P6 (Figure 3A,B). At later stages, fewer
           not detectable after birth (Figure 2A). In the retina of                 apoptotic cells were seen perhaps in part due to the
           transgenic mice, hE2F1 was expressed in the outer                        previous loss of cells in the ONL in the transgenic
           ventricular cell layer, the developing rods, from P0                     retinas (Figure 3A). The location of most of the
           (day of birth) through P6 (Figure 2A). The hE2F1 was                     apoptotic cells in the ONL indicates that the dying cells
           also present in mature photoreceptor cells at later                      are, in fact, developing photoreceptors.
           stages (data not shown). All o€spring from both lines                       During normal development, the arrest of cellular
           had similar expression patterns and developed the same                   proliferation and the initiation of di€erentiation
           phenotype (see below).                                                   proceeds from the central to the peripheral retina
                                                                                    (Young, 1985). Therefore, we sought to correlate the
                                                                                    time course of proliferation and apoptosis with this
           Developing neuroretinal cells in TgIRBPE2F1 transgenic
                                                                                    developmental feature. Proliferation in the central
           mice continue to synthesize DNA
                                                                                    retina was most robust at P0, gradually decreased
           During retinal development, the ventricular cells cease                  after birth and ceased at P9 in transgenic mice (Figure
           proliferation at P6 in the central region and at P11 in                  2C). Interestingly, there were more apoptotic cells in
           the peripheral region of the retina (Young, 1985). The                   the central region compared to the peripheral region in
           development of post-mitotic cells parallels morpholo-                    transgenic retinas earlier in postnatal development
           gical changes in the developing outer nuclear layer                      (Figure 3B), whereas at P9 and P10 there were more
           (ONL) of photoreceptors. Active proliferation, as                        apoptotic cells in the periphery (Figure 3A,B). Hence,
           evidenced by BrdU incorporation, was seen throughout                     it appears that the excess cell proliferation caused by
           the retina of the wild-type mice from P0 to P4 (Figure                   E2F1 precedes excess apoptosis and that the apoptosis
           2B,C). During this period, there were more BrdU-                         appears to occur in a developmentally regulated
           positive cells in the retina of transgenic mice than in                  pattern.
           the control retina (Figure 2B,C). By P6, BrdU stained
           cells were restricted to the peripheral retina in wild-
                                                                                    Delayed differentiation in the neuroretinal cells in
           type mice, whereas BrdU-positive cells were detected in
                                                                                    TgIRBPE2F1 transgenic mice
           the central as well as the peripheral retina in the
           transgenic mice at P6 (Figure 2B). These data                            During retinal development, multiple cell types are
           demonstrate that forced expression of E2F1 in vivo is                    generated and arranged into morphologically distinct
           sucient to drive DNA synthesis in the developing                        layers. To assess the e€ect of E2F1 expression on
           retina that should have been ®lled with post-mitotic                     development of the multi-layer structure of retina,
           photoreceptors. Interestingly, the ectopic BrdU-positive                 retina sections of P4 ± P6 were compared (Figure 4). At
           cells in the transgenic mice were di€erent from BrdU-                    P6 the outer plexiform layer (OPL), which is composed
           positive cells in the normal mice in two regards. First,                 of synaptic processes between photoreceptor cells in the
           they were located near the pigment epithelium (PE) in                    ONL and cells in the inner nuclear layer (INL), was
           transgenic mice (Figure 2Bb,d) as opposed to the inner                   evident in the control mice (Figure 4a), but was not
           plexiform layer (IPL) in wild-type mice (Figure 2Ba,c).                  apparent in transgenic mice (Figure 4b). However, the
           Second, their nuclei were more rounded than the                          OPL was visible at P8 and later stages in transgenic
           BrdU-positive cells in wild-type mice (data not shown).                  retinas (Figure 3Af,h) when the cells of the ONL were
           Their location and nuclear shape suggest that they are                   better di€erentiated. This ®nding suggests that ectopic
Oncogene
E2F1 and p53 inactivation in retinoblastoma
                                                                         S-CJ Lin et al
                                                                                                                                                7077

    Figure 2 Ectopic E2F1 expression enhances retinal cell proliferation. (a) E2F1 expression in the retinas. E2F1 immunostaining was
    performed on retinal sections of wild-type (left) and TgIRBPE2F1 retinas (right) of 6-day-old (P6) neonates using anti-E2F1
    antibodies. Scale bar, 100 mm. (B) BrdU uptake in P4 (a,b) and P6 (c,d) retinas of wild-type (left panels) and TgIRBPE2F1 (right
    panels) mice. Cen, central retina; Per, peripheral retina; G, Ganglion cell layer; IPL, inner plexiform layer; PE, pigment epithelium.
    Scale bar, 200 mm. (C) Quantitative comparison of BrdU-positive cells in wild-type and TgIRBPE2F1 retinas of di€erent ages (P0 ±
    P9). BrdU-positive nuclei within a length of 250 mm to optical nerve (center) and to ciliary body (periphery) were counted. The
    mean+s.e. is based on data collected from at least three mice

expression of E2F1 may delay certain aspects of retinal                    We next sought to determine how ectopic expression
development.                                                             of E2F1 altered the di€erentiation of individual
                                                                                                                                             Oncogene
E2F1 and p53 inactivation in retinoblastoma
                                                                     S-CJ Lin et al
7078

               Figure 3 E€ects of E2F1 overexpression on retinal apoptosis. (A) Apoptotic cells detected by TUNEL assay in wild-type (left
               panels) and TgIRBPE2F1 (right panels) retinas of P4 ± P10 mice. ONL, outer nuclear layer. Scale bar, 200 mm. (B) Quantitative
               comparison of apoptotic cells in wild-type and TgIRBPE2F1 retinas. Apoptotic nuclei within a length of 250 mm to optical nerve
               (center) and to ciliary body (periphery) were counted. The mean+s.e. is based on data collected from at least three mice

           photoreceptors. To accomplish this, opsin protein,                         but not the peripheral retina in wild-type mice at P2
           which is deposited along the lateral cell membrane                         (data not shown). Cells in the peripheral retina were
           and synaptic terminals of the immature rods as shown                       observed to express opsin at later stages. At P4 and P6,
           previously in several other vertebrates (Nir et al.,                       levels of opsin expression appeared to be increased in
           1984), was detected by immunohistochemical staining.                       all rod photoreceptor cells (Figure 4c,e). In transgenic
           Opsin ± positive cells were ®rst detected in the central                   mice, opsin-expressing cells were not seen until P6
Oncogene
E2F1 and p53 inactivation in retinoblastoma
                                                                        S-CJ Lin et al
                                                                                                                                            7079

     Figure 4 E€ects of E2F1 overexpression on photoreceptors cell di€erentiation. (a,b) Formation of OPL in the P6 retinas of wild-
     type (a) and TgIRBPE2F1 (b) mice. (c ± f) Expression of opsin in the retinal sections of wild-type (left panel) and TgIRBPE2F1
     (right panel) mice. Arrowheads indicate opsin-expressing cells; arrow indicates clusters of apoptotic cells. OPL, outer plexiform
     layer. Scale bar, 100 mm

(Figure 4f). At this stage, the opsin expression pattern                mice, at which time most regions were depleted of
in the retina of transgenic mice resembles that of the                  ONL cells, no rosettes were detected (Figure 5f).
P2 retinas in wild-type mice (data not shown). At later                    Remarkably, there was a striking hemispheric
stages, the photoreceptor cells that are present showed                 asymmetry with respect to the dysplastic changes and
robust expression of opsin (see below). In summary,                     the apoptosis discussed above. Divided by the optic
these data indicate that the transgenic expression of                   nerve, multiple layers of photoreceptor cells were
E2F1 delays normal development in the retina and the                    arranged into rosette-like structures on one side of
expression of a photoreceptor-speci®c gene, but does                    the section (Figure 5a), whereas no photoreceptor cells
not prevent eventual photoreceptor di€erentiation.                      were present in the other side (Figure 5c). The loss of
                                                                        photoreceptor cells was con®rmed by the lack of opsin
                                                                        staining (Figure 5d). Such initial side-to-side di€erence
Dysplasia and degeneration in the neuroretinal cells in
                                                                        and the ®nal photoreceptor cell loss to the whole retina
TgIRBPE2F1 transgenic mice
                                                                        suggest that there might be a progressive manifestation
Although the photoreceptors eventually expressed                        of the E2F1 e€ects from one retinal hemisphere to the
opsin at apparently normal levels, their di€erentiation                 other; however, the molecular basis for this is not
was still altered in the retinas of transgenic mice.                    known.
Speci®cally, rosettes were frequently observed in the
outer nuclear layer in the retinas of TgIRBPE2F1 mice,
                                                                        Loss of p53 promotes both severe dysplasia and
(Figure 5a). These rosettes were composed of at least
                                                                        hyperplasia in the retina of TgIRBPE2F1 mice but does
partially di€erentiated photoreceptors as evidenced by
                                                                        not prevent photoreceptor degeneration
opsin expression (Figure 5b). The dysplastic lesions
were observed from P9 through P28 TgIRBPE2F1                            Because E2F1-mediated cell death has been associated
mice. However, in retinas of 3 month old transgenic                     with elevated expression of p53 (Hiebert et al., 1995;
                                                                                                                                         Oncogene
E2F1 and p53 inactivation in retinoblastoma
                                                                     S-CJ Lin et al
7080

               Figure 5 Asymmetric formation of rosette-like structures and loss of photoreceptor cells in a single TgIRBPE2F1 retina. Retinal
               sections from di€erent hemispheres of the same TgIRBPE2F1 retina were stained with H&E (a,c,e,f) or immunostained with anti-
               opsin antibodies (b,d). (a ± e), TgIRBPE2F1 retinas; (f), wild-type retinas. Ages of the mice are indicated. Arrows indicate the
               rosettes. G, Ganglion cell layer; IPL, inner plexiform layer; INL, inner nuclear layer; OPL, outer plexiform layer; ONL, outer
               nuclear layer; OS&IS, outer segment and inner segment; PE, pigment epithelium. Scale bar, (a ± d) 50 mm, (e,f) 100 mm

           Kowalik et al., 1998), we tested whether the loss of p53                   although it remained signi®cantly higher than in
           a€ects the phenotype of TgIRBPE2F1 mice. TgIRB-                            normal retinas (data not shown). In 3-week-old and
           PE2F1; p537/7 mice were generated after two rounds                         adult mice, a similar degree of retinal degeneration
           of crossings to p53 null mice (Donehower et al., 1992).                    occurred in TgIRBPE2F1 and TgIRBPE2F1; p537/7
           The loss of p53 in the TgIRBPE2F1 mice a€ected                             mice (Figure 6c,d). Thus, although the loss of p53
           several aspects of the retina phenotype. First, rosette                    modi®es the degree of apoptosis detectable at any
           formation in TgIRBPE2F1; p537/7 mice appeared                              speci®c time, the loss of photoreceptors in the mature
           earlier (from P6 through P14) and dysplastic lesions                       retina of TgIRBPE2F1 mice is p53-independent. These
           were often present in both hemispheres of the retina                       results indicate that loss of p53 a€ects multiple aspects
           (Figure 6a,b). Second, in some regions of the retina,                      of the phenotype induced by dysregulated E2F1
           multiple layers were not evident. For example, the                         expression, which may allow excessive hyperplasia
           distinct layered structure of ONL in both wild-type and                    and disorganization of multiple retinal cell types.
           TgIRBPE2F1 mice was not observed in TgIRBPE2F1;                            However, this is still not sucient for oncogenic
           p537/7 mice (Figure 6b). At P10, although the OPL                          transformation of photoreceptors because signi®cant
           was evident between the INL and the remaining ONL                          retinal degeneration still occurs eventually.
           in TgIRBPE2F1 mice, the OPL was almost undistin-
           guishable in TgIRBPE2F1; p537/7 mice (data not
           shown). The INL was also in disarray as evidenced                          Discussion
           by focal expansion to nearly 14-cells thick (Figure
           6b,c). The loss of layers in the outer two layers of the                   E2F transcription factors are important targets for RB-
           retina is likely due to focal hyperplasia/dysplasia of                     dependent control of the cell cycle and tumor
           multiple cell types, including photoreceptor cells,                        formation. We have generated a transgenic mouse
           pigment epithelial cells and cells of the INL. Third,                      model to investigate how the aberrant expression of
           the delayed expression of opsin in the TgIRBPE2F1                          E2F1 a€ects photoreceptor cell development and
           retina was lessened in the p53 null background.                            contributes to tumorigenesis. In TgIRBPE2F1 retinas,
           Speci®cally, at P6, approximately eight photoreceptors                     ectopic expression of E2F1 resulted in increased DNA
           cells in every row expressed opsin in wild-type retina                     synthesis and apoptosis in developing photoreceptor
           (Figure 4e), yet there was only one opsin-positive cell                    cells, a ®nding that is similar to what occurs in Rb7/7
           in every 5 ± 20 rows in TgIRBPE2F1 retina (Figure 4f).                     retinas during embryonic development and in retinal
           However, there were one or two opsin-positive cells in                     transplant experiments. The di€erentiation of photo-
           every three or four rows in TgIRBPE2F1; p537/7                             receptor cells, judging from the delayed appearance of
           retina (data not shown). Fourth, the level of cell death                   the OPL and opsin expression, was also impaired but
           in the TgIRBPE2F1; p537/7 retinas was much lower                           not completely blocked. Ultimately, E2F1 deregulation
           than that in the TgIRBPE2F1 retinas during P6 to P14,                      led to photoreceptor cell degeneration rather than
Oncogene
E2F1 and p53 inactivation in retinoblastoma
                                                                     S-CJ Lin et al
                                                                                                                                        7081

    Figure 6 Retinal dysplasia and degeneration in TgIRBPE2F1; p537/7 mice. (a ± d) H&E staining of retinal sections of
    TgIRBPE2F1; p537/7 mice. Note dysplastic photoreceptor cells showing rosette structures in the P6 (a) and P14 (b) retinas. PE,
    pigment epithelium; INL, inner nuclear layer. Scale bar, 100 mm

retinoblastoma formation. p53 de®ciency partially                    may lead to retinal dysplasia by releasing E2F1 from
rescued the cell death of TgIRBPE2F1 photoreceptor                   RB (and p107) mediated repression.
cells and promoted retinal transient hyperplasia and                    Both Rb-de®ciency and E2F1 overexpression alter
dysplasia but did not allow macroscopic tumor                        opsin expression in the retinal transplant experiments
formation. Together, these studies indicate that normal              and transgenic mouse retinas, respectively (Figures 1B
regulation of E2F1, likely by RB-dependent mechan-                   and 4). These ®ndings are consistent with previous
isms, is required for photoreceptor and retinal                      studies of how loss of RB alters cellular di€erentiation.
development. However, loss of E2F1 regulation is not                 For example, the trigeminal ganglia of Rb-de®cient
sucient for retinal tumor formation.                                mice show incomplete di€erentiation (Lee et al., 1994).
   Several recent studies in other mouse tissues support             In Rb heterozygous knockout mice, mutation of the
our conclusions that deregulated E2F1 contributes to                 remaining wild-type Rb allele alters the di€erentiation
the phenotypes associated with Rb inactivation. First,               of Rb-null melanotrophs in the pituitary gland. These
aberrant S phase entry and apoptosis in the develop-                 cells fail to become mature melanotrophs and are not
ing central nervous system and lens of Rb7/7 embryos                 innervated by growth inhibitory dopaminergic nerve
is suppressed in Rb/E2F1 double mutants (Tsai et al.,                terminals (Nikitin and Lee, 1996). Finally, in vitro and
1998). Second, mutation of E2F1 suppresses inap-                     in vivo studies have demonstrated that loss of Rb, or
propriate proliferation and cell death of the lens ®ber              ectopic expression of E2F1 inhibits the full expression
cell compartment induced by the HPV E7 oncopro-                      of genes induced during skeletal muscle and adipose
tein, which binds to and inactivates the RB family                   di€erentiation (Chen and Lee, 1999; Chen et al., 1996;
proteins (McCa€rey et al., 1999). In these respects, the             Classon et al., 2000; Novitch et al., 1996; Zacksenhaus
e€ects of aberrant expression of E2F1 in mouse                       et al., 1996). Taken together, these data indicate that
photoreceptors are similar to e€ects in the lens and                 loss of RB function and ectopic expression of E2F1 is
other areas of the central nervous system. Further-                  sucient to prevent normal di€erentiation of numerous
more, our transgenic mouse model adds further                        types of cells. Interestingly, it does not completely
insight into how RB and the RB-related protein                       prevent di€erentiation per se but rather it appears to
p107 a€ects retinal development. Previous studies have               delay the expression of a di€erentiation-speci®c gene in
shown that knock out mice and chimeric mice in                       photoreceptors. Although photoreceptors in our trans-
which all or some of the retina is derived from Rb+/                 genic mice eventually show robust opsin expression, we
7
  ; p1077/7 or Rb7/7; p1077/7 cells have dysplasia                   have not performed a quantitative assay to compare
(Lee et al., 1996; Robanus-Maandag et al., 1998) that                the level of expression with normal photoreceptors. At
is similar to what we observed in our studies of E2F1.               a molecular level, how loss of Rb alters di€erentiation
Thus, our data suggest that loss of the Rb and p107                  is not known. It has been shown to interact with a
                                                                                                                                     Oncogene
E2F1 and p53 inactivation in retinoblastoma
                                                                    S-CJ Lin et al
7082
           number of transcription factors that mediate cellular                     cell proliferation of other retinal cells in a p53-
           di€erentiation (Chen et al., 1996; Gu et al., 1993;                       dependent manner. However, the molecular mechan-
           Lipinski and Jacks, 1999; Schneider et al., 1994). On                     isms by which this may occur are not known. It is also
           the other hand, it is conceivable that loss of Rb                         plausible that there might be low levels of ectopic E2F1
           indirectly alters the activity of transcription factors that              expression in non-photoreceptor cells and the resultant
           drive cellular di€erentiation by releasing E2F activity                   growth promoting e€ect is detectable only in the
           (Wang et al., 1995). Finally, it is also conceivable that                 absence of p53. Further experiments will be required
           either loss of Rb or aberrant E2F1 expression could                       to clarify the mechanisms involved.
           in¯uence cellular di€erentiation by allowing for the                         One notable di€erence between our studies and
           accumulation of additional genetic changes. Further                       previous studies of the RB gene pathway in the retina
           experiments will be required to clarify the molecular                     is the development of macroscopic retinal tumors,
           basis by which loss of Rb or aberrant expression of                       which occurred either in Rb7/7; p1077/7 chimeric mice
           E2F1 blocks the normal di€erentiation of photorecep-                      (Robanus-Maandag et al., 1998) or in the TgIRBPE7;
           tor cells.                                                                p537/7 mice (Howes et al., 1994). The molecular basis
              Compared to other models in which E2F1 is                              for the transient dysplasia and hyperplasia that occurs
           ectopically expressed, our study of E2F1-expressing                       in our TgIRBPE2F1; p537/7 mice is not known. It is
           photoreceptor cells o€ers several new observations.                       certainly conceivable that pro-apoptotic mechanisms in
           First, hyperplasia was not observed in photoreceptors                     the retina are activated in the presence of ectopically
           in our mice whereas transgenic expression of E2F-1 in                     expressed E2F1 to prevent tumor formation in the
           squamous epithelial cells caused increased cell prolif-                   retina. In this regard, it is interesting to note that
           eration and hyperplasia despite increased apoptosis                       overexpression of SV40 T antigen under the control of
           (Pierce et al., 1998a). Second, the loss of p53 only                      the rhodopsin promoter also results in retinal degen-
           partially abrogates E2F1-mediated cell death and only                     eration in mice (al-Ubaidi et al., 1992). However, these
           transiently sustains dysplasia and hyperplasia in                         photoreceptor cells proliferate in tissue culture and are
           photoreceptors of our TgIRBPE2F1 mice (Figure 6),                         tumorigenic when implanted subcutaneously into nude
           while E2F1-induced apoptosis in the epidermal cells is                    mice (al-Ubaidi et al., 1992). Furthermore, in the retina
           largely p53-dependent and its loss allows for the                         of Rb7/7 chimeric mice, there was severe cellular
           formation of spontaneous skin tumors (Pierce et al.,                      degeneration during E16.5 ± E18.5 (Maandag et al.,
           1998b). Similarly, apoptosis in Rb-de®cient embryos                       1994). The contribution of Rb7/7 retinal cells was
           depends on p53 and E2F1 in the central nervous                            reduced to less than 15% in adult mice (Maandag et
           system (Macleod et al., 1996; Tsai et al., 1998). Finally,                al., 1994). Similarly, our intracranial Rb7/7 retinal
           we demonstrated that the forced expression of E2F1                        transplants exhibited signi®cant apoptosis 1 week after
           alters photoreceptor and retina development (Figures 4                    transplantation, a time point equivalent to E18.5 of the
           and 5); however, similar forced expression of E2F1 in                     donor tissue (Figure 1B). However, transplanted Rb7/7
           epidermal cells does not appear to block cellular                         retinal cells were able to survive for more than 2
           di€erentiation (Pierce et al., 1998a). In summary, these                  months (data not shown). Taken together, these results
           ®ndings indicate that the e€ects of deregulated RB/                       indicate the presence of apoptotic factors in situ in the
           E2F1 on cellular proliferation, di€erentiation, and                       eyes of the transgenic mice. It is conceivable that these
           apoptosis di€er in speci®c types of cell and tissue.                      pro-apoptotic factors are disrupted in the TgIRB-
              Although no macroscopic tumors form in TgIRB-                          PE7; p537/7 mice but not in our TgIRBPE2F1; p537/7
           PE2F1; p537/7 mice, there is transient hyperplasia and                    mice. Delineating mechanisms underlying phenotypic
           dysplasia of multiple retinal cell types (Figure 6a,b).                   di€erences between genetic loss of Rb and p107 (Lee et
           This unexpected ®nding suggests that aberrant expres-                     al., 1996; Robanus-Maandag et al., 1998) or by
           sion of E2F1 in photoreceptors may promote pro-                           expression of IRBP-E7 in the p537/7 background
           liferation of nearby p53-de®cient inner retinal cells and                 (Howes et al., 1994), versus the non-tumorigenic
           pigment epithelial cells through a paracrine mechan-                      properties of IRBP ± E2F1 (even in the absence of
           ism. There is a precedent for paracrine regulation of                     p53) would likely lead to better understanding of the
           cells in vertebrate retinas. For example, during                          functions of these gene products and the biology of
           development, the seven major types of cells in the                        human retinoblastoma.
           retina are derived from a pool of multipotent
           progenitor cells (Cepko et al., 1996). Numerous studies
           have shown that cell ± cell interactions are involved in
           cell fate determination during this time (Belliveau and                   Materials and methods
           Cepko, 1999; Belliveau et al., 2000; Cepko et al., 1996).
           Similarly, studies using intact frog retina indicate that                 Construction of TgIRBPE2F1 transgene and generation of
           the phosphorylation status of rhodopsin is in part                        transgenic mice
           regulated by dopamine released from the inner cell                        A plasmid pBSpKCR3 modi®ed from pKCR3 that consists of a
           layers (Udovichenko et al., 1998). Clearly this is minor                  1.9 kb fragment of the murine IRBP promoter, a 1.2 kb part of
           compared to the phosphorylation induced by light.                         the rabbit b globin gene including partial exon 2, intron 2, and
           Thus, it seems reasonable to speculate that the aberrant                  exon 3, and a polyadenylation site, was provided by Dr Jolene
           expression of E2F1 in photoreceptors could alter the                      Windle (Howes et al., 1994). A 1.5 kb fragment containing the
Oncogene
E2F1 and p53 inactivation in retinoblastoma
                                                                   S-CJ Lin et al
                                                                                                                                        7083
full-length human E2F1 cDNA was inserted into exon 3 of the        in formalin, and embedded in paran. Tissue sections were
rabbit b globin gene of pBSpKCR3. The TgIRBPE2F1                   treated with 4N HCl/0.2% Triton X-100, and processed for
transgene was cut with KpnI and XbaI, gel-puri®ed, and             immunostaining as described above using anti-BrdU anti-
microinjected into the male pronucleus of fertilized eggs          bodies (1 : 200, Becton Dickinson). The terminal deoxyribo-
derived from CB6 F16C57BL/6 intercrosses. Transgenic               nucleotidyltransferase-mediated dUTP-biotin nick end
founder mice were identi®ed by Southern blot analysis of tail-     labeling (TUNEL) assay was performed as described (Howes
derived genomic DNA. Transgenic mice of subsequent                 et al., 1994).
generations were identi®ed by PCR using primers derived from
the human E2F1 cDNA sequence, 5' primer (5'-GGAAACT-
                                                                   Quantification of cells in the neuroretina
GACCATCAGTACCTG-3'), and 3' primer (5'-CCAGCCAC-
TGGATGTGGTTCTT-3'). Transgenic lines were established              Quanti®cation of the number of cells positive for BrdU and
by breeding founders to C57BL/6 mice. Subsequent genera-           TUNEL was performed using light microscopy. The numbers
tions of transgenic mice were maintained on a mixed CB6            of nuclei in the neuroretina within a length of 250 mm to the
F16C57BL/6 genetic background.                                     optical nerve (the central region) and to the ciliary body (the
                                                                   peripheral region) were counted. At least three mice were
                                                                   examined to obtain the mean and the standard error.
Histological analysis
Samples were ®xed in 10% bu€ered formalin and processed
                                                                   Retinal transplantation methods
through paran embedding follow standard procedures.
Sections (4 mm thick) were cut parallel to the optic nerve         Embryos were obtained from anesthetized, timed-pregnant
and stained with hematoxylin and eosin (H&E) for light             heterozygous Rb-1D20/+ mice (Lee et al., 1992). Retinas were
microscopy.                                                        removed from E11.5 ± E12.5 embryos and transplanted into
                                                                   the superior colliculus or cerebral cortex of 1-day-old (P1)
                                                                   wild-type or Rb-1D20/+ neonates as described (Hankin and
E2F1 and opsin immunostaining
                                                                   Lund, 1987). Brie¯y, a small glass pipet containing the donor
Immunostaining was performed using the Vectastain Elite            tissue and sterile F10 culture medium (GIBCO BRL) was
ABC Kit (Vector Laboratories). Brie¯y, paran-embedded             inserted through a small hole in the cartilagenous skull
sections were rehydrated, ®xed in methanol containing 3%           overlying the recipient midbrain or cortex. The donor tissue
hydrogen peroxide for 5 min, and blocked in 1% normal              was ejected from the pipet with 1 ml of F10.
horse serum (NHS) in PBS for 20 min. The sections were
incubated with anti-E2F1 monoclonal antibodies (1 : 200,
Pharmingen) or mouse anti-opsin antibodies (1 : 2000) diluted
in 2% NHS/PBS overnight at 48C. After washes in PBS,
sections were incubated with biotinylated anti-mouse IgG for
30 min. Slides were washed in PBS, incubated with                  Acknowledgments
streptavidin/biotinylated peroxidase conjugate, developed by       We thank Dr Jolene Windle for the plasmid pBSKCR3,
Vector VIP or DAB substrates (Vector Laboratories), and            Nancy Ransom for technical advice and Dr CL Cepko for
counterstained with methyl green or hematoxylin.                   helpful comment. We appreciate Drs NP Hu's, Y Min's,
                                                                   and E Gerbino's contribution in the early phase of the
                                                                   project. We also wish to thank K Bushnell for proof-
BrdU incorporation and TUNEL assays
                                                                   reading the manuscript. This work was supported by NIH
Mice received bromodeoxyuridine (BrdU) at a dose of                grants CA49649 to E Lee. Histology was performed in part
100 mg/g of body weight by intraperitoneal and subcutaneous        by a core facility of the San Antonio Cancer Institute
injection. After 2 h, retina tissue samples were collected, ®xed   (grant #P30 CA 54174).

References

Adams PD and Kaelin Jr WG. (1995). Semin. Cancer Biol., 6,         Classon M, Kennedy BK, Mulloy R and Harlow E. (2000).
   99 ± 108.                                                         Proc. Natl. Acad. Sci. USA, 97, 10826 ± 10831.
al-Ubaidi MR, Holly®eld JG, Overbeek PA and Baehr W.               DeGregori J, Leone G, Miron A, Jakoi L and Nevins JR.
   (1992). Proc. Natl. Acad. Sci. USA, 89, 1194 ± 1198.              (1997). Proc. Natl. Acad. Sci. USA, 94, 7245 ± 7250.
Belliveau MJ and Cepko CL. (1999). Development, 126,               Donehower LA, Harvey M, Slagle BL, McArthur MJ,
   555 ± 566.                                                        Montgomery Jr CA, Butel JS and Bradley A. (1992).
Belliveau MJ, Young TL and Cepko CL. (2000). J. Neurosci.,           Nature, 356, 215 ± 221.
   20, 2247 ± 2254.                                                Duronio RJ, O'Farrell PH, Xie JE, Brook A and Dyson N.
Brehm A and Kouzarides T. (1999). Trends Biochem. Sci., 24,          (1995). Genes Dev., 9, 1445 ± 1455.
   142 ± 145.                                                      Dyson N. (1998). Genes Dev., 12, 2245 ± 2262.
Cepko CL, Austin CP, Yang X, Alexiades M and Ezzeddine             Field SJ, Tsai FY, Kuo F, Zubiaga AM, Kaelin Jr WG,
   D. (1996). Proc. Natl. Acad. Sci. USA, 93, 589 ± 595.             Livingston DM, Orkin SH and Greenberg ME. (1996).
Chen G and Lee EY. (1999). DNA Cell Biol., 18, 305 ± 314.            Cell, 85, 549 ± 561.
Chen PL, Riley DJ, Chen Y and Lee WH. (1996). Genes Dev.,          Gu W, Schneider JW, Condorelli G, Kaushal S, Mahdavi V
   10, 2794 ± 2804.                                                  and Nadal-Ginard B. (1993). Cell, 72, 309 ± 324.
Clarke AR, Maandag ER, van Roon M, van der Lugt NM,                Hankin MH and Lund RD. (1987). J. Comp. Neurol., 263,
   van der Valk M, Hooper ML, Berns A and te Riele H.                455 ± 466.
   (1992). Nature, 359, 328 ± 330.

                                                                                                                                     Oncogene
E2F1 and p53 inactivation in retinoblastoma
                                                                    S-CJ Lin et al
7084
           Hankin M, Sefton AJ and Lund RD. (1993). Brain Res. Dev.                  Nir I, Cohen D and Papermaster DS. (1984). J. Cell Biol., 98,
              Brain Res., 75, 146 ± 150.                                               1788 ± 1795.
           Hiebert SW, Packham G, Strom DK, Ha€ner R, Oren M,                        Novitch BG, Mulligan GJ, Jacks T and Lassar AB. (1996).
              Zambetti G and Cleveland JL. (1995). Mol. Cell Biol., 15,                J. Cell Biol., 135, 441 ± 456.
              6864 ± 6874.                                                           Pierce AM, Fisher SM, Conti CJ and Johnson DG. (1998a).
           Howes KA, Ransom N, Papermaster DS, Lasudry JG,                             Oncogene, 16, 1267 ± 1276.
              Albert DM and Windle JJ. (1994). Genes Dev., 8, 1300 ±                 Pierce AM, Gimenez-Conti IB, Schneider-Broussard R,
              1310.                                                                    Martinez LA, Conti CJ and Johnson DG. (1998b). Proc.
           Hu N, Gutsmann A, Herbert DC, Bradley A, Lee WH and                         Natl. Acad. Sci. USA, 95, 8858 ± 8863.
              Lee EY. (1994). Oncogene, 9, 1021 ± 1027.                              Qin XQ, Livingston DM, Kaelin Jr WG and Adams PD.
           Ishizaki J, Nevins JR and Sullenger BA. (1996). Nat. Med., 2,               (1994). Proc. Natl. Acad. Sci. USA, 91, 10918 ± 10922.
              1386 ± 1389.                                                           Robanus-Maandag E, Dekker M, van der Valk M, Carrozza
           Jacks T, Fazeli A, Schmitt EM, Bronson RT, Goodell MA                       ML, Jeanny JC, Dannenberg JH, Berns A and te Riele H.
              and Weinberg RA. (1992). Nature, 359, 295 ± 300.                         (1998). Genes Dev., 12, 1599 ± 1609.
           Johnson DG, Cress WD, Jakoi L and Nevins JR. (1994).                      Ross JF, Liu X and Dynlacht BD. (1999). Mol. Cell, 3, 195 ±
              Proc. Natl. Acad. Sci. USA, 91, 12823 ± 12827.                           205.
           Johnson DG, Schwarz JK, Cress WD and Nevins JR. (1993).                   Schneider JW, Gu W, Zhu L, Mahdavi V and Nadal-Ginard
              Nature, 365, 349 ± 352.                                                  B. (1994). Science, 264, 1467 ± 1471.
           Kowalik TF, DeGregori J, Leone G, Jakoi L and Nevins JR.                  Shan B and Lee WH. (1994). Mol. Cell Biol., 14, 8166 ± 8173.
              (1998). Cell Growth Di€er., 359, 113 ± 118.                            Singh P, Wong SH and Hong W. (1994). EMBO J., 13,
           Kowalik TF, DeGregori J, Schwarz JK and Nevins JR.                          3329 ± 3338.
              (1995). J. Virol., 69, 2491 ± 2500.                                    Skapek SX, Jansen D, Wei TF, McDermott T, Huang W,
           Lee EY, Chang CY, Hu N, Wang YC, Lai CC, Herrup K,                          Olson EN and Lee EY. (2000). J. Biol. Chem., 275, 7212 ±
              Lee WH and Bradley A. (1992). Nature, 359, 288 ± 294.                    7223.
           Lee EY, Hu N, Yuan SS, Cox LA, Braley A, Lee WH and                       Slansky JE and Farnham PJ. (1996). Curr. Top Microbiol.
              Herrup K. (1994). Genes Dev., 8, 2008 ± 2021.                            Immunol., 208, 1 ± 30.
           Lee MH, Williams BO, Mulligan G, Mukai S, Bronson RT,                     Tsai KY, Hu Y, Macleod KF, Crowley D, Yamasaki L and
              Dyson N, Harlow E and Jacks T. (1996). Genes Dev., 10,                   Jacks T. (1998). Mol. Cell., 2, 293 ± 304.
              1621 ± 1632.                                                           Udovichenko IP, Newton AC and Williams DS. (1998). J.
           Lin SC, Skapek SX and Lee EY. (1996). Semin. Cancer Biol.,                  Biol. Chem., 273, 7181 ± 7184.
              7, 279 ± 289.                                                          Wang J, Helin K, Jin P and Nadal-Ginard B. (1995). Cell
           Liou GI, Geng L, al-Ubaidi MR, Matragoon S, Hanten G,                       Growth Di€er., 6, 1299 ± 1306.
              Baehr W and Overbeek PA. (1990). J. Biol. Chem., 265,                  Wu CL, Classon M, Dyson N and Harlow E. (1996). Mol.
              8373 ± 8376.                                                             Cell Biol., 16, 3698 ± 3706.
           Lipinski MM and Jacks T. (1999). Oncogene, 18, 7873 ± 7882.               Wu X and Levine AJ. (1994). Proc. Natl. Acad. Sci. USA, 91,
           Maandag EC, van der Valk M, Vlaar M, Feltkamp C,                            3602 ± 3606.
              O'Brien J, van Roon M, van der Lugt N, Berns A and te                  Xu G, Livingston DM and Krek W. (1995). Proc. Natl. Acad.
              Riele H. (1994). EMBO J., 13, 4260 ± 4268.                               Sci. USA, 92, 1357 ± 1361.
           Macleod KF, Hu Y and Jacks T. (1996). EMBO J., 15,                        Yamasaki L, Bronson R, Williams BO, Dyson NJ, Harlow E
              6178 ± 6188.                                                             and Jacks T. (1998). Nat. Genet., 18, 360 ± 364.
           McCa€rey J, Yamasaki L, Dyson NJ, Harlow E and Griep                      Yamasaki L, Jacks T, Bronson R, Goillot E, Harlow E and
              AE. (1999). Mol. Cell. Biol., 19, 6458 ± 6468.                           Dyson NJ. (1996). Cell, 85, 537 ± 548.
           Morgenbesser SD, Williams BO, Jacks T and DePinho RA.                     Young RW. (1984). Cell, 85, 537 ± 548.
              (1994). Nature, 371, 72 ± 74.                                          Young RW. (1985). Anat. Rec., 212, 199 ± 205.
           Mulligan G and Jacks T. (1998). Trends Genet., 14, 223 ± 229.             Zacksenhaus E, Jiang Z, Chung D, Marth JD, Phillips RA
           Nevins JR. (1998). Cell Growth Di€er., 9, 585 ± 593.                        and Gallie BL. (1996). Genes Dev., 10, 3051 ± 3064.
           Nikitin A and Lee WH. (1996). Genes Dev., 10, 1870 ± 1879.                Zhang HS, Postigo AA and Dean DC. (1999). Cell, 97, 53 ± 61.

Oncogene
You can also read