Vous êtes sur la page 1sur 10

HUMAN MUTATION 21:182^191 (2003)

p53 REVIEW ARTICLE

TP53 Family Members and Human Cancers


Jean Bénard,1,2n Setha Douc-Rasy,2 and Jean-Charles Ahomadegbe1,3,4
1
Unité de Génétique Tumorale, Service de Génétique, Département de Biologie Clinique, Institut Gustave Roussy, Villejuif, France; 2Centre
National de la Recherche Scientifique UMR8126, Institut Gustave Roussy, Villejuif, France; 3UPRESS Pharmacologie et Nouveaux
Traitements des Cancers, Institut Gustave Roussy, Villejuif, France; 4Faculté de Pharmacie, UPJV, Amiens, France

For the p53 Special Issue


Based on gene sequence homologies, a p53 (TP53) gene family become apparent with the addition of the
most recently identified p63 (TP73L; formerly TP63) and p73 (TP73) genes to the already known p53.
The p53 gene encodes for a unique protein eliciting well-known tumor suppressor gene (TSG) properties
that mediate cellular response to DNA damage, e.g., cell cycle arrest or apoptosis. In contrast, both
homologues specify an array of isoforms different in their N- and C-terminal domains. Transactivating
isoforms, such as TAp63/p73, show TSG properties similar to p53, while isoforms lacking N-terminal
transactivating domain such as DNp63/p73, induce a functional block against p53 as well as TAp63/p73
activities. Both p63/p73 types of isoforms are involved in development: p63 is critical for epithelial stem
cell renewal and epithelial homeostasis, and p73 is involved in neurogenesis and natural immune
response. These facts support interdependent functions for the p53 family members, which appear linked
together in a complex and tight regulation network to fulfill cellular functions related to DNA damage
and tissue homeostasis maintenance. The lack of p63/p73 mutations in human cancers rule out a typical
TSG role for either of the p53 homologues. Nonetheless, p63 and p73 genes seem strongly involved in
malignancy acquisition and maintenance process because of: 1) their tissue identities, and 2) their close
interplay activities within the p53 family members, and primarily through the negative regulatory role
played by DNp63/p73 isoforms for cell death control and differentiation. Hum Mutat 21:182–191,
2003. r 2003 Wiley-Liss, Inc.

KEY WORDS: p53; TP53; p63; TP73L; p73; TP73; tumor suppressor; splicing; transactivation; apoptosis;
differentiation; DNA damage; development; tumorigenesis; cancers
DATABASES:
TP53 – OMIM: 191170; GenBank: NM_000546 (mRNA)
http://p53.curie.fr/ (p53 Web Site at Institut Curie)
www.iarc.fr/P53/index.html (IARC p53 Mutation Database)
TP73L – OMIM: 603273; GenBank: NM_003722 (mRNA)
TP73 – OMIM: 601990; GenBank: NM_005427 (mRNA)

INTRODUCTION role for p53 homologues in tumorigenesis. Moreover,


in stark contrast to p53, which is mutated in about
The discovery in 1997 of the p73 gene (TP73; 50% of human cancers, it was soon revealed that p73
MIM# 601990) at the 1p36 locus, a location known and p63 genes were not inactivated by mutations in a
to be subject to recurrent loss of heterozygosity in large number of cancers from various tissues.
various human cancers [Kaghad et al., 1997], led to All together, these findings raised a hot question:
the critical issue of whether this first-identified p53 Do p53 homologues elicit functions similar to p53?
homologue functions as the archetypal tumor sup- These issues have been in part elucidated for each p53
pressor gene (TSG) p53 (TP53; MIM# 191170). One homologue from the evidence of an array of isoforms
year later, the p53 gene family expanded with the
cloning of a second homologue, the p63 gene (TP73L;
n
MIM# 603273) [Yang et al., 1998], located at 3q27- Correspondence to: Jean Be¤nard, UniteŁ de GeŁneŁtique Tumor-
ter. This chromosomal region is not subject to loss but ale, Service de GeŁneŁ tique, Departement de Biologie Clinique,
Institut Gustave Roussy, Villejuif, France.
to gain in cancers, and therefore suggests some
oncogenic functions for p63. These apparently para- DOI 10.1002/humu.10172
doxical findings caused head scratching and skepti- Published online in Wiley InterScience (www.interscience.wiley.
cism in an oncology research community in search of a com).

r2003 WILEY-LISS, INC.


TP53 FAMILY MEMBERS 183

1 59 142 321 353 397 641

p63α TAD PXXP DBD OD SAM Basic

22% 60% 37%

1 45 113 300 325 363 393


p53 TAD PXXP DBD NLS OD Basic
29% 63% 38%

1 54 131 310 345 390 636


p73α TAD PXXP DBD OD SAM Basic
(A)

P1 TAD DBD OD
1 2 3 4 5 6 7 8 9 10 11 12 13 14
p73α

∆exon2
P2
3bis
∆Np73α
(B)
δ
γ β
ζ
1 2 3 4 5 6 7 8 9 10 11 12 13 14

ε
(C)

FIGURE 1. Comparative gene structures and functional organization of the p53 family members. A: For each gene, the transac-
tivation (TAD), proline-rich (PXXP), DNA binding (DBD), oligomerization (OD), sterile a motif (SAM), and post SAM basic
domains are represented. B: Genomic organization of the p73 gene.The proximal promoter (P1) yields b,c,d,e,f TAp73 isoforms
while the distal promoter (P2) located in intron 3 gives rise to DN isoforms. C: In addition to the a isoform, COOH terminal
splicing leads to b,c,d,e,f p73 isoforms for bothTA and DNp73 molecules species. [Color ¢gure can be viewed in the online issue
at www.interscience.wiley.com.]

expressed in tissues, some harboring transactivation oligomerization domain (OD) [Kaghad et al., 1997].
domain (TAD), a hallmark for transcription factors Both p53 ‘‘siblings’’ exert p53-like activities: They can
with TSG activity, others lacking TAD, e.g., DN- bind to p53 DNA consensus target sites, transactivate
isoforms, with antagonistic biological properties. There- p53 responsive genes, and induce cell growth arrest or
fore, unlike their namesake, which stands as a canonical apoptosis [Kaghad et al., 1997; Jost et al., 1997; Yang
TSG, each p53 homologue can play either a TSG or an and McKeon, 2000]. The 20kb p53 is organized in 11
oncogenic role with respect to its specific isoforms. exons, whereas p63 and p73 genes are both over 60 kb
In this review we will first present what is currently and comprise 15 and 14 exons respectively. Unlike the
known about the two genes and their role during unique and alternative, but infrequently used, splicing
developmental and adult states, both at the cell and of the p53 gene, both p63 and p73 consistently give
tissue levels. Thereafter, we will examine their possible rise to an array of multiple protein isoforms due to
roles in pre-malignancies and cancers. differential mRNA splicing [Kaghad et al., 1997; De
Laurenzi et al., 1998; Fillippovich et al., 2001] and to
COMPARATIVE ORGANIZATION AND
alternative promoter usage [Yang and McKeon, 2000;
Pozniak et al., 2000]. Another specific feature of the
TRANSCRIPTION REGULATION IN THE TP53
p63 and p73 C-termini is the occurrence of a sterile
FAMILY
alpha motif domain (SAM), which is involved in
Akin to their p53 leader, p63 and p73 exhibit the protein–protein interactions.
three typical domains of a transcription factor across Figure 1 depicts a comparison of the domain
various species: namely an acidic, amino-terminal structure of p53 protein and major isoforms encoded
transactivation (TA) domain; a central core DNA- by human p63 and p73. Historically, the C-terminal
binding domain (DBD); and a carboxy-terminal complexity was decrypted before the N-terminal
184 BEŁNARD ET AL.

complexity of the 2 gene loci. Most of alternative ubiquitination has not been demonstrated, but a p73a
splicing occurs at the 30 end and involves more modified by a covalent linking with SUMO-1 (small
specifically exons 10 to 13, hence yielding transcripts ubiquitin-related modifier) was found to be more
that encode protein isoforms with various C-terminal rapidly degraded by the proteasome than the un-
structures. So far, among the high number of spliced modified p73 [Minty et al., 2000]. Unlike p53 and
p63 and p73 isoforms found at the RNA level, three of p73, p63 does not bind to MDM2 [Dohn et al., 2001],
them are consistently found at the protein level, revealing yet another difference between p53 family
namely a (full structure of both genes), b (splicing of members.
exon 13), and g (splicing of exons 10 to 12 for p73 and Therefore one can distinguish two negative feed-
15 for p63) [Yang et al., 2002]. In contrast, the p53 back loops controlling p53 and TAp73, namely the
gene encodes one major transcript yielding a unique mdm2 and DNp73 loops, keeping the cell death
protein with transcriptional activity. trigger under tight control.
Another striking finding regarding p63 and p73
gene expression regulation has been the occurrence of A RANGE OF P63 AND P73 ISOFORMS WITH
two different promoters, P1 and P2, that yield two
DISTINCT BIOLOGICAL ACTIVITIES
distinct classes of proteins [Yang et al., 1998, 2000].
The P1 promoter leads isoforms showing TA domain The combination of C-terminal diversity and acidic
with p53 homology. The P2 promoter, located within N-terminal regulation, including two distinct promo-
intron 3 and over 30-kb downstream, gives rise to N- ters, produces at least six major transcripts and
terminal-truncated (DN) isoforms with biological subsequent protein isoforms (a,b,g) for each gene
properties opposite to those of p63/p73 TA isoforms, [De Laurenzi et al., 1998; Schmale and Bamberger,
and lacking TA domain. The (DN) isoforms were 1997]: TAp63/p73 p53-like isoforms and DNp63/p73
identified first in mouse, then in human [Ishimoto p53-antagonist isoforms (Fig. 1). This unusual gene
et al., 2001]. Specific sequences of P1 and P2 bring regulation suggests a ‘‘two genes in one’’ concept for
about distinct biological properties. E2F-1 binding p63/p73 since TA and DN-isoforms act respectively as
sites are present in both promoters, but p53-binding tumor suppressors and oncogenes [Stiewe et al., 2002;
sites are present only in the P2 promoter [Yang et al., Stiewe and Pützer, 2002; Grob et al., 2002].
2000]. Very recently, two crucial studies have It is well known that p53 TSG properties arise from
indicated that: 1) TAp73 directly activates the its binding to specific DNA sequences and from
transcription of endogenous DNp73 by binding to transactivation of target genes that specify cell cycle
the two p73-specific target elements located on P2 control proteins (p21, MDM2, GADD 45, 14-3-3-s,
[Nakagawa et al., 2002]; and 2) p53 induces DNp73 BTG2) and apoptosis (PIG3, Bax). Active p53 was
both at the mRNA and protein levels, as a result of a also shown to play a key role in B lymphocyte, muscle,
p53 direct activation of the P2 promoter [Kartasheva spermatogenesis, and neuronal differentiation [Rotter
et al., 2002]. DNp73a-isoform readily associates with et al., 1994; Sidell and Koeffler, 1998] as well as in
TAp73a/b and p53, as assessed by immunoprecipita- retinoic acid-mediated terminal differentiation of
tion assay, and inhibits their transactivation activities. embryonic carcinoma cells [Curtin et al., 2001]. p53
On the contrary, TAp63 shows only a marginal DN is also involved in keratinocytic differentiation,
p73 transcription. Importantly, the negative feedback although it does not determine this process because,
regulation of TAp73 and p53 by their DNp73 target in p53 knock-out mice, keratinocytic differentiation
provides for a novel autoregulatory system modulating may be activated by Ca++ treatment.
cell survival and death. At the post-translational level, Another important difference between p53 family
DN-isoforms are consistently identified in various members concerns their interactions with viral onco-
tissues expressing p53-homologues, suggesting that proteins. Indeed if E6 protein, SV40 large T antigen,
DN-isoform elicits a protein of higher stability than and AdE1B 55 kd protein bind to and inactivate p53
the TA isoforms transiently expressed and subject to during cell transformation, they do not interact with
rapid degradation. either p63 or p73 [Irwin and Kaelin, 2001].
Yet another remarkable distinctive property of the When over-expressed in human cells, TA p63/p73
p53 homologues concerns their degradation process by proteins also bind to p53 DNA target sequences,
MDM2. The turnover of p53, a short-lived protein, is transactivate p53-responsive genes, and thereby
regulated by ubiquitination through MDM2 binding, induce cell cycle arrest, differentiation, and apoptosis
leading to p53 degradation by proteasome, and in a p53-like manner [Kaghad et al., 1997; Jost et al.,
thereby limiting p53 accumulation [Haupt et al., 1997; Yang et al., 1998] running through p53 most
1997]. Similar to p53, the p73a and b proteins bind to characteristic biological effects. However, transcrip-
MDM2 through their N-terminal but this interaction tional activity fluctuates with p63/p73 splice variants
leads to transcription and apoptosis inactivation, and and must be considered. Indeed, TAp63/p73a iso-
does not result in p73 degradation by proteasome forms display a less active transcriptional activity than
[Balint et al., 1999; Zeng et al., 1999]. So far, p73 TAp63/p73b isoforms in p53 assays [Yang et al., 1998;
TP53 FAMILY MEMBERS 185

Morphogenesis,
Stress
? Epithelial
UV ? p63 development/
differentiation

∆Np63

DNA ATM Growth


damage ATR, Casein KII arrest
p53
UV DNA-PK
Apoptosis
P300 p53 mutant
PCAF Adenovirus ∆Np73
IR E4orfE6
ATM Neurogenesis,
Mismatch Neuronal
c-Abl p73
repair development/
Cisplatin DNA-PK differentiation

FIGURE 2. Schematic p53 family members pathways. Besides speci¢c developmental and physiological functions, p63 and
p73 participate to p53 genomic guardian function. Upon genotoxic stresses induced by ultraviolet (UV), g irradiation (IR) or
cisplatin, the two homologues interplay with p53 to achieve growth arrest and apoptosis functions. It is now established that
p53 as well asTAp73 induce a direct activation of DN-p73 creating thus a feedback loop to control negatively these functions.
[Color ¢gure can be viewed in the online issue at www.interscience.wiley.com.]

Schmale and Bamberger, 1997] indicative of a phosphorylated by the ATM (ataxia telangiectasia-
negative regulatory effect of the a isoforms from the mutated) protein [Gong et al., 1999; Shaul, 2000].
sterile a motif (SAM) (Fig. 1) [Thanos and Bowie, Therefore, a p73 repair pathway for DNA damage
1999]. Similarly the COOH-terminal region located exists, which is p53-independent (Fig. 2).
in the post SAM domain can have an auto-inhibitory So far regarding p63 response to DNA damage,
effect on the TAp63/p73a transactivation [Ozaki important information has been gained from UV-
et al., 1999]. treated human keratinocytes: TAp63 isoforms are up-
Noteworthy, the above-described biological activa- regulated while DNp63 isoforms are dramatically
tion has been demonstrated using forced expression of down-regulated in these p63 expressing cells [Liefer
TA isoforms of the p53 homologues, likely to mask et al., 2000]. As a matter of fact, the DNp63 down
differences between the various p53 -family members. regulation parallels p53 stabilization, which, in turn,
Not to mention that, so far, no binding of endogenous acts as the apoptotic executor of UV-damaged
p63/p73 to their putative target gene promoters has keratinocytes. These findings provide evidence for a
been demonstrated in vivo by chromatin precipitation. full interplay between the two main antagonistic p63
Nonetheless, differences may exist between human isoforms and p53 for DNA-damage response (Fig. 2).
p53 family members regarding their potency to induce In p53-deficient SaoS-2 cells, oncogenes such as
activation of p53 target genes. Indeed, if p53 leads to E2F1, c-myc, and E1A can induce and activate
strong p21 and MDM2 induction, p73 was shown to endogenous p73 protein expression and drive cells to
induce a major and strong 14-3-3 s up-regulation apoptosis, pointing to a p73 upstream pathway that is
[Zhu et al., 1998]. p53-independent [Zaika et al., 1999]. Importantly, a
In response to various stresses resulting in DNA very recent work shows that, in response to various
damage, p53 is activated by phosphorylation to stresses (drug, g-irradiation), p53 requires TAp73, as
perform its cell-cycle arrest and apoptotic tasks. well as p63, to activate promoters of apoptotic genes
Similarly, in response to DNA-damaging agents such such as NOXA, PERP, and BAX [Flores et al., 2002],
as cis-platin and g-irradiation, p73 up-regulation supporting a crucial role for the two homologues in
results in apoptotic response albeit using a pathway p53-mediated apoptotic activity.
distinct from that of p53. p73 is phosphorylated by c- To sum up, a large body of evidence favors specific
abl, the non-tyrosine kinase receptor, which is itself upstream and downstream target gene regulation for
186 BEŁNARD ET AL.

each p53 family member, and for their respective DNp73 isoform is predominantly expressed in murine
isoforms, playing a role in non-redundant functions. developing brain and sympathetic neurons, hence
Undoubtedly, RNA interference strategies specifically explaining neuronal defects from lack of DNp73. As a
designed against these isoforms will provide definitive matter of fact, DNp73 isoform was shown to inhibit
clues with regard to biological activities of each p53- neuronal apoptosis from NGF withdrawal by blocking
family member. the p53 pro-apoptotic function [Pozniak et al., 2000].
These seminal findings designate p73 and DNp73
THE TP53 HOMOLOGUES AS DEVELOPMENT
isoforms as determinants of cellular differentiation and
apoptosis in neuronal tissues. Moreover, in developing
PROTAGONISTS
Cajal-Retzius cells of the human neocortex, very
The respective deficient mice have provided further recently, a close association was shown between p73
insights concerning the physiological role of p53, p73, expression and reelin, a glycoprotein involved in
and p63. p53 null-mice develop spontaneous tumors neuronal migration [Meyer et al., 2002]. Also
at high frequency without exhibiting a specific interesting in this respect, a recent study indicated
phenotype, in particular during their development that human cytomegalovirus CMV, which causes
[Donehower et al., 1992]. If active p53 does not severe neuronal defects in utero, induces dramatic
appear to be involved in physiological apoptosis apoptosis inhibition and thus abnormal nerve cell
occurring throughout embryogenesis, it becomes survival through accumulation of DNp73a [Allart
crucial for DNA-damage-induced apoptosis [Lowe et al., 2002]. Besides these neurological and cognitive
et al., 1993]. In stark contrast, both p73 and p63 null- impairments, p73 null-mice display severe defects in
mice show specific developmental defects but no pheromone-based social behavior, reproduction, and
spontaneous tumors at all [Yang et al., 1999, 2000]. infection control. Noteworthy, the complex range of
Studies with p63-null mice have revealed the physiological deficiencies correlates with high p73
occurrence of TA and DN isoforms in the stem cell levels expressed in corresponding tissues of p73-
compartment of stratified epithelia, thereby also proficient mice. These phenotypic traits suggest that
offering a very useful guide for our understanding of the p73 functions sum up a very basic signaling system
p53-homologues regulation and functions [Yang et al., in vertebrates, i.e., both sensor and controller
1998, 1999]. p63 knockout-mice are born alive but functions of intra and extra cellular signals, which
display severe deformations of limbs as well as of are integrated in cell homeostasis [Yang et al., 2002].
epithelia including skin, breast, urothelia, and prostate
[Mills et al., 1999; Yang et al., 1999]. The p63 –/–
P63 AND P73 GENES AND TUMORIGENESIS
mouse skin does not undergo normal development
because it is lacking stratification and differentiation Although, in contrast to p53 null-mice, p63/p73
markers. These mice also lack mammary glands, hair null-mice do not develop any spontaneous tumors, the
follicles, and teeth. Such murine phenotypic disorders complex network and regulation system formed by the
were confirmed by p63 mutations in various human main p53-homologue isoform proteins and p53 itself
syndromes involving limb and ectodermal develop- must be considered in the context of tumorigenesis.
ment. Indeed p63 mutations have been related to Two levels of interaction between p53 members can be
EEC syndrome, a set of autosomal dominant disorders distinguished: physical interactions and pathway
(ectrodactyly, ectodermal dysplasia, and cleft lip/ regulations.
palate), as well as to sporadic split, hand-split foot Human tumor-derived p53 mutants can engage
malformations [Yang et al., 1999; Celli et al., 1999; with different p73 and p63 isoforms in a physical
van Bokhoven et al., 2001]. In fact, epithelial loss in association. In fact, interaction with mutant p53
p63-deficient mice reflects the inability of the multi- impairs, in vitro and in vivo, p73 and p63 sequence-
layered regenerative epithelia stem cells to undergo specific DNA binding, consequently inhibiting their
asymmetric division. Indeed, in contrast to the transcriptional activity [Strano et al., 2002]. More-
physiological asymmetrical division (one stem cell over, in cells carrying endogenous p53 mutants, p53
providing one stem cell to repopulate the stem homologues are unable to recruit some of their target
compartment, and one basal cell for differentiation), gene promoters [Strano et al., 2002]. Therefore, p63
all p63 –/– basal cells undergo terminal differentiation, and p73 may contribute to the biological properties of
leading to a complete depletion of the stem cell specific mutants by promoting tumorigenesis and by
compartment. In mice [Yang et al., 1998] and human conferring selective survival advantage to cancer cells.
[Faridoni-Laurens et al., 2001] epithelia, p63 is highly The p53 homologues are also integrated in various
expressed in stem cells. p53-independent pathways conducting the comple-
Neuronal defects have been the first phenotypic tion of various functions, in particular apoptosis as
traits observed in p73-deficient mice, including high previously stressed. However, and strikingly, in
intracranial pressure and hippocampus dysgenesis response to DNA damage, p53-dependent apoptosis
[Yang et al., 2000]. Importantly, it was shown that relies upon the functionality of both p63 and p73:
TP53 FAMILY MEMBERS 187

TABLE 1. Meta-Analysis of Mutation, Loss of Heterozygozity, Imprinting and Methylation of p73 in Human Cancers (after
Stiewe and Pˇtzer [2002])
Tumor Mutation LOH Imprinting/methylation
Bladder cancer 0/23 B: 35/46
Breast cancer 1/145 29/194 B: 8/24
CNS-tumors 1/77
Colorectal cancer 0/125 8/46
Esophageal cancer 0/48 11/39 B: 4/9
S:1/9
Gastric cancer 0/82 12/32 B: 37/53
Head and neck cancers 3/58 16/108 B: 8/8
Hematological malignancies 0/91 B: 4/10
methylated:11/35
Hepatocellular carcinoma 1/70 21/71 B: 7/8
Lung cancer 1/114 32/107 B: 30/36
Melanoma 0/68 3/27
Neuroblastoma 2/302 46/246 B:16/25
Ovarian cancer 0/63 33/141
Prostate cancer 0/133 2/38 B: 27/27
Renal cancer 0/27 B: 8/12
S: 2/12

Total 9/1426 213/1049 B:184/258


S:3/21

LOH, loss of heterozygozity; B, biallelic; S, switch.

When using E1A p53-dependent apoptotic fibroblast (P405R and P425L) had no significant effect on
system, the lack of p63 and p73 clearly results in the p73a/b transcriptional activity and growth suppressing
cell’s loss of functional p53 necessary to undergo ability [Naka et al., 2001]. Given that p73 was located
apoptosis [Flores et al., 2002]. Another line of in a region so far described as imprinted, the lack of
evidence favoring the interplay between p53 and its mutation in the remaining allele raised the possibility
homologues for tumorigenesis is the fact that DNp73 that the loss of transcribed allele would render tumor
proteins are effective inhibitors of p53 and TAp73 cells totally p73-deficient. Although some studies
functions [Stiewe et al., 2002]. Consistent with this pointed out monoallelic expression, particularly in
fact, endogenous DNp73 protects cells from p53- inflammatory breast cancers [Ahomadegbe et al.,
dependent apoptosis. The same phenomenon may be 2000], most showed p73 biallelic expression or allele
proposed for DNp63 in squamous cells. These switching, which invalidates the latter working
functions provide a plausible explanation for high hypothesis. p73 expression seems very low and
p73 and p63 levels in human cancer cells even in the variable from tissue to tissue: This fact and the poor
absence of p63/p73 mutations. quality of the first antibodies raised against p73
Although completed soon after the p73 and p63 prevented western-blot use and favored RT-PCR
clonings and without any regulation knowledge of analyses. p73 transcript levels were regularly measured
these novel genes, translational studies in cell lines higher in a significant percentage of cancers than in
and fresh tumors of various cancers support the above normal tissues [Zaika et al., 1999; Stiewe and Pützer,
proposed mechanisms. Now let’s consider the devel- 2002]. However, when polymerase reactions were
opments of recent research saga. Since p73 gene was carried out using cDNA binding domain as substrate,
shown to reside at 1p36-3, a locus found to be subject the semi-quantitative data obtained did not permit an
to recurrent loss of heterozygosity in many cancers answer to the question of whether specific TAp73 or
(neuroblastoma, breast, colorectal cancers, melano- DNp73 isoform was overexpressed and which one
ma) and to belong to the smallest overlapping region could play a role in tumorigenesis. p73 up-regulation
[Kaghad et al., 1997], p73 has been proposed as the in some tumors can result from mitogenic oncogenes
long sought after TSG candidate at 1p36 [Versteeg as shown in experimental models [Zaika et al., 2001]
et al., 1995]. A thorough search of intragenic that lead to massive apoptosis. Attempts have been
sequence variations in the p73 gene resulted in an made to relate cancer’s p73 status and disease
unswerving lack of mutations. In a sampling of more prognosis: Studies of large-size patient groups with
than 1,400 human tumors including solid (breast, hepatocellular carcinomas [Tannapfel et al., 1999],
lung, colorectal, esophageal, hepatocellular, renal, colorectal carcinoma [Sun, 2002], and breast cancer
prostate, melanoma, neuroblastoma, and brain tu- [Dominguez et al., 2001] produced a poor prognosis
mors) as well as hematological malignancies, only nine statistical trend for tumors with high p73 level.
cases (0.6%) showed missense mutations (Table 1) However, again, no evidence pointed to either isoform
[Stiewe and Pützer, 2002]. The rare mutations found species whose transcript level increased: Was it TA or
188 BEŁNARD ET AL.

DNp73? Besides the physiological transcript DNp73, found to be significantly increased [Hibi et al., 2000;
an N-terminal splice variant lacking exon 2 could be Crook et al., 2000; Park et al., 2000]. In agreement
also found in tissues. Its role in malignancy of various with the DNp63 oncogenic role, premalignant and
tissues is under investigation (Fig. 1) [O’Nions et al., malignant lesions of the cervix and endometrium
2001; Douc-Rasy et al., 2002] (Ahomadegbe and show the highest p63 level in differentiated squamous
Bénard, in preparation). cells [Park et al., 2000]. Additional data regarding p73
So far, two tumor models highlight a direct role for involvement in cancers came recently from the biopsy
p73 and p63 in tumorigenesis, neuroblastoma (NB) analysis from 50 patients with head and neck
and squamous carcinoma cells. Remarkably NB squamous cell carcinoma (HNSCC). It showed a
constitutes a first tumor model in which p73 status down regulation of p73 expression in 30% of the
has been thoroughly studied and it provides a HNSCC associated with high p63 expression in basal
significant clinical impact [Melino et al., 2002]. In layers as assessed by immunohistochemistry [Faridoni-
NB, p53 is not mutated but inactivated by cytoplasmic Laurens et al., 2001]. Microdissection of HNS
retention [Moll et al., 1995], making p73 a good epithelium and HNSCC, currently in progress, is
candidate for NB tumorigenesis, given the recurrent likely to reveal a DNp63 overexpression concomittent
loss of heterozygosity at the p73 locus in these tumors. to this p73 down regulation.
Early research provided evidence for lack of p73 Another finding of interest is DNp63 isoform
mutations in fresh NB, ruling out this gene for a overexpression resulting from p63 amplification in
typical TSG role [Nakagawara et al., 1999; Zaika et al., these tumors. This overexpression mediates a decrease
1999; Barrois et al., 2001]. In fact, neuroblastic in phosphorylation levels of b-catenin, which in turn
tumors (NTs), occurring in early childhood, display a induces b-catenin nuclear accumulation and activates
wide spectrum of differentiation. Recurrent deletions b-catenin signaling pathway. So, DNp63 isoforms act
involving the p73 locus are frequently observed in as positive regulators of the b-catenin signaling
undifferentiated NTs. To address the question of pathway, hence providing a basis for their oncogenic
possible p73 implication in neuroblastic differentia- properties [Patturajan et al., 2002]. From all these
tion, this gene-expression status was investigated in a studies it emerges that p63, p73, and p53 play a role in
panel of differentiated and undifferentiated tumors. concert in the differentiation and carcinogenesis of
Although no mutations were found, p73 transcript HNS epithelium.
profiles differed between undifferentiated and differ- In conclusion, the discovery of the p53 homologues
entiated tumors. The frequency of the transcripts has defined the p53 family. Clearly, p53 is not a lonely
lacking exon 2 (species 1–3) appeared to be higher in genome guardian counteracting genotoxic insults: It
undifferentiated than in differentiating and differen- operates with the mutual assistance of p73 and p63
tiated NTs [Douc-Rasy et al., 2002]. In contrast, within a complex network including distinct but
products from the use of an alternate promoter complementary pathways. Lack of p63/73 mutations
(DNp73) were present in all NTs. In addition, only in human cancers definitively rule out a typical TSG
DNp73, but no full-length proteins, were detected by role for either p53 homologues. Nonetheless, it is
immunoblot, suggesting a greater stability of N- strongly suggested that p63 and p73 genes may be
truncated isoforms [Douc-Rasy et al., 2002]. To involved in acquisition and maintenance of malig-
determine the role of DNp73 in NB, the pattern of nancy given: 1) their tissues identities, as well as 2)
this gene’s in vivo expression has been analyzed and the close interplay activities within the p53-family
the prognostic significance of its expression evaluated. members, primarily through the negative regulatory
Recent results clearly indicate that DNp73 expression role played by DNp63/p73 isoforms for cell death
is linked to a reduced apoptosis in NB tumor tissue. control and differentiation. The state of the p53 field
Expression of this variant in NB patients significantly has increased in complexity tremendously over the
correlates with reduced survival and progression-free last 5 years. New clues may permit researchers to
survival: It is a predictor of poor outcome and accurately typify tumors according to alterations of
constitutes a prognostic factor independent of age, their respective p53 and p53 homologue pathways.
primary tumor site, stage, and MYCN amplification Undoubtedly, this p53 family genomic signature in
[Casciano et al., 2002a]. At a mechanistic level, it is every tumor will become a prerequisite for designing
noteworthy that P2 promoter functionality in NB cells new therapeutic approaches for cancers.
and tumor tissues is, at least in part, regulated by
epigenetic mechanisms [Casciano et al., 2002b].
ACKNOWLEDGMENTS
In direct correlation with its role in epithelia
development and homeostatic maintenance, and The authors thank Dr. Daniel CAPUT (Sanofi
consistent with its locus at 3q27-29, a chromosomal Recherches, Labèges, France) for invaluable discus-
region submitted to gain, p63 levels significantly sions about their research and Sanofi Recherches for
increased in squamous cell carcinoma [Hibi et al., financial support. The manuscript was edited by
2000]. Clearly, it is the DNp63 isoform level that was English Booster Ltd, www.englishbooster.com.
TP53 FAMILY MEMBERS 189

REFERENCES 2001. Wild type p73 overexpression and high-grade malig-


Ahomadegbe JC, Tourpin S, Kaghad M, Zelek L, Vayssade M, nancy in breast cancer. Breast Cancer Res 66:183–190.
Mathieu MC, Rochard F, Spielmann M, Tursz T, Caput D, Donehower LA, Harvey M, Slagle BL, McArthur MJ,
Riou G, Bénard J. 2000. Loss of heterozygosity, allele Montgomery CA, Jr., Butel JS, Bradley A. 1992. Mice
silencing and decreased expression of p73 gene in breast deficient for p53 are developmentally normal but susceptible
cancers: prevalence of alterations in inflammatory breast to spontaneous tumor. Nature 356:215–221.
cancers. Oncogene 19:5413–5418. Douc-Rasy S, Barrois M, Echeynne M, Kaghad M, Blanc E,
Allart S, Martin H, Detraves C, Terrasson J, Caput D, Davrinche Raguenez G, Goldschneider D, Terrier-Lacombe MJ, Hart-
C. 2002. Human cytomegalovirus induces drug resistance and mann O, Moll U, Caput D, Bénard J. 2002. DN-p73a
alteration of programmed cell death by accumulation of DN- accumulates in human neuroblastic tumors; Am J Pathol
p73 alpha. J Bioe Chem 277: 29063–29068. 160:631–639.
Balint E, Bates S, Vousden KH. 1999. Mdm-2 p73a without Faridoni-Laurens L, Bosq J, Janot F, Vayssade M, Le Bihan ML,
targeting degradation. Oncogene 18:3923–3929. Kaghad M, Caput D, Bénard J, Ahomadegbe JC. 2001.
Barrois M, Eychenne MK, Terrier-Lacombe MJ, Duarte N, p73 expression in basal layers of head and neck squamous
Dubourg C, Douc-Rasy S, Chompret A, Khagad M, epithelium: a role in differentiation and carcinogenesis
Hartmann O, Caput D, Bénard J. 2001. Genomic and allelic in concert with p53 and p63? Oncogene 20:5302–
expression status of the p73 gene in human neuroblastoma. 5312.
Med Pediatr Oncol 36:45–48. Fillippovich I, Sorokina N, Gatei M, Haupt Y, Hobson K,
Casciano I, Mazzocco K, Boni L, Pagnan G, Banelli B, Moallem E, Spring K, Mould M, McGuckin MA, Lavin MF,
Allemanni G, Ponzoni M, Tonini GP, Romani M. 2002a. Khanna KK. 2001. Transactivation deficient p73alpha (p73
Expression of DNp73 is a molecular marker for adverse Deltaexon2) inhibits apoptosis and competes with p53.
outcome in neuroblastoma patients. Cell Death Differ Oncogene 20:514–522.
9:246–251. Flores ER, Tsai KY, Crowley D, Sengupta S, Yang A, McKeon F,
Casciano I, Banelli B, Croce M, Allemanni G, Ferrini S, Jacks T. 2002. p63 and p73 are required for p53-dependent
Tonini GP, Ponzoni M, Romani M. 2002b. Epigenetic apoptosis in response to DNA damage. Nature 416:560–564.
control of transcription of the anti-apoptotic DN variant Gong J, Costanzo A, Yang H, Melino G, Kaelin WG, Levero M,
of the p73 gene in neuroblastoma. Cell Death Differ 9:343– Wang J. 1999. The tyrosine kinase c-Abl regulates p73 in
345. apoptotic response to cisplatin-induced DNA damage.
Celli J, Duijf P, Hamel BC, Bamshad M, Kramer B, Smits AP, Nature 399:806–808.
Newbury-Ecob R, Hennekam RC, Van Buggenhout G, van Grob TJ, Fey MF, Tobler A. 2002. The two faces of p73. Cell
Haeringen A, Woods CG, van Essen AJ, de Waal R, Vriend Death Differ 9:237–245.
G, Haber DA, Yang A, McKeon F, Brunner HG, van Haupt Y, Maya R, Kazaz A, Oren M. 1997. MDM2 promotes
Bokhoven H. 1999. Heteozygous germlin mutations in the the rapid degradation of p53. Nature 387:296–299.
p53 homolog p63 are the cause of EEC syndrome. Cell Hibi K, Trink BMP, Westra WH, Caballero OL, Hill DE,
99:143–153. Ratovitski EA, Jen J, Sidransky D. 2000. AIS is an oncogene
Crook T, Nicholls JM, Brooks L, O’Nions J, Allday MJ. 2000. amplified in squamous cell carcinoma. Proc Natl Acad Sci
High level expression of D-N-p63: a mechanism for the 97:5462–5467.
inactivation of p53 in undifferentiated nasopharyngeal Ishimoto O, Kawahara C, Enjo K, Obinata M, Nukiwa T, Ikawa
carcinoma (NPC). Oncogene 19:3439–3444. S. 2001. Possible oncogenic potential of its antagonist
Curtin JC, Dragnev KH, Sekul D, Christie AJ, Dmitrovsky E, DNp73: a newly identtified isoform of human p73. Cancer
Spinella MJ. 2001. Retinoic acid activates p53 in human Res 62:636–641.
embryonal carcinoma through retinoid receptor-dependent Irwin MS, Kaelin WG. 2001. P53 family update: p73 and p63
stimulation of p53 transactivation function. Oncogene develop their own identities. Cell Growth Differ 12:337–349.
20:2559–2569. Jost CA, Marin MC, Kaelin WG. 1997. P73 is a simian
De Laurenzi V, Costanzo A, Barcaroli D, Rerrinoni A, (correction of human) p53-related protein that can induce
Falco M, Annicchiarico-Petruzzelli M, Levrero M, Melino apoptosis. Nature 389:191–194.
G. 1998. Two new p73 splice variants gamma and delta, Kaghad M, Bonnet H, Yang A, Creancier L, Biscan JC, Valent
with different transcriptional activity. J Exp Med 188:1763– A, Minty A, Chalon P, Lelias JM, Dumont X, Ferrara P,
1768. McKeon F, Caput D. 1997. Monoallelically expressed
De Laurenzi V, Raschella G, Barcaroli D, Annicchiarico- gene related to p53 at 1p36, a region frequently
Petruzelli M, Ranalli M, Catani M, Tanno B, Costanzo A, deleted in neuroblastoma and other human cancers. Cell
Levrero M, Melino G. 2000. Induction of neuronal 90:809–819.
differentiation by p73 in a neuroblastoma cell line. J Biop Kartasheva N, Contente A, Lenz-Stöppler C, Roth J,
Chem 275:15226–15231. Dobelstein M. 2002. P53 induces the expression p73DN,
Dohn M, Zhang S, Chen X. 2001. p63alpha and DeltaNp63al- establishing an autoregulatory feedback loop. Oncogene
pha can induce cell cycle arrest and apoptosis and 21:4715–4727.
differentially regulate p53 target genes. Oncogene 20:3193– Kovalev S, Marchenko N, Swendeman S, La Quaglia M, Moll
3205. U. 1998. Expression level, allelic origin and mutation analysis
Dominguez G, Silva JM, Silva J, Garcia JM, Sanchez A, of the p73 gene in neuroblastoma tumors and cell lines. Cell
Navarro A, Gallego I, Provencio M, Espana P, Bonilla F. Growth Differ 9:897–903.
190 BEŁNARD ET AL.

Liefer KM, Koster MI, Wang XJ, Yang A, McKeon F, Roop DR. in normal cell differentiation? Semin Cancer Biol 5:
2000. Down-regulation of p63 is required for epidermal UV- 229–236.
B-induced apoptosis. Cancer Res 60:4016–4020. Schmale H, Bamberger C. 1997. A novel protein with strong
Lowe S, Ryley H, Jacks T, Houssman D. 1993. p53-dependent homology to the tumor suppressor p53. Oncogene 15:1363–
apoptosis modulates the cytotoxicity of anticancer agents. 1367.
Cell 74:957–967. Shaul Y. 2000. c-abl: activation and nuclear targets. Cell Death
Melino G, De Laurenzi V, Vousden K. 2002. p73: friend or foe Differ 7:10–16.
in tumorigenesis? Nature Rev 2:605–615. Sidell N, Koeffler HP. 1998. Modulation of Mr 53,000 protein
Meyer G, Perez-Garcia CG, Abraham H, Caput D. 2002. with induction of differentiation of human neuroblastoma
Expression of p73 and reelin in the developing human cells. Cancer Res 48:2226–2230.
cortex. J Neurosci 22:4973–4986. Stiewe T, Theseling CC, Pützer BM. 2002. Transactivation-
Mills AA, Zheng B, Wang XJ, Vogel H, Roop DR, Bradley A. deficient Delta TAp73 inhibits p53 by direct competition for
1999. p63 is a p53 homologue required for limb and DNA binding : implications for tumorigenesis. J Biol Chem
epidermal morphogenesis. Nature 398:708–713. 277:14177–14185.
Minty A, Dumont X, Kaghad M, Caput D. 2000. Covalent Stiewe T, Pützer BM. 2002. Role of p73 in malignancy : tumor
modification of p73a by SUMO-1. Two-hybrid screening suppressor or oncogene ? Cell Death Differ 9:237–245.
with p73 identifies novel SUMO-1 interacting proteins and a Strano S, Fontemaggi G, Costanzo A, Rizzo MG, Monti O,
SUMO-1 interaction motif. J Biol Chem 275:36316–36323. Baccarini A, Del Sal G, Levrero M, Sacchi A, Oren M,
Moll U, LaQuaglia M, Bénard J, Riou G. 1995. Wild-type p53 Blandino G. 2002. Physical interaction with human tumor-
protein undergoes cytoplasmic sequestration in undifferen- derived p53 mutants inhibits p63 activities. J Biol Chem
tiated neuroblastomas but not in differentiated tumors. Proc 277:18817–18826.
Natl Acad Sci USA 92:4407–4411. Sun XF. 2002. p73 overexpression is a prognostic factor in
Naka M, Ozaki T, Takada N, Takahashi M, Shishikura T, patients with colorectal adenocarcinoma; Clin Cancer Res
Sakiyama S, Tada M, Todo S, Nakagawara A. 2001. 8:165–170.
Functional characterization of naturally occurring Tannapfel A, Wasner M, Krause K, Geissler F, Katalinic A,
mutants (P405R and P425L) of p73 alpha and p73beta Hauss J, Mossner J, Engeland K, Witteking C. 1999.
found in neuroblastoma and lung cancer. Oncogene 20: Expression of p73 and its relation of histopathology and
3568–3572. prognosis in hepatocellular carcinoma. J Natl Cancer Inst
Nakagawa T, Takahashi M, Ozaki T, Watanabe KI, Todo S, 91:1154–1158.
Mizuguchi H, Hayakawa T, Nakagawara A. 2002. Thanos CD, Bowie JU. 1999. p53 family members p63 and p73
Autoinhibitory regulation of p63 by DNp73 to modulate are SAM domain-containing proteins. Protein Sci 8:1708–
cell survival and death through a p73-specific target 1710.
element within the DNp73 promoter. Mol Cell Biol 22: van Bokhoven H, Hamel BC, Bamshad M, Sangiorgi E,
2575–2585. Gurrieri F, Duijf PH, Vanmolkot KR, van Beusekom E, van
O’Nions J, Brooks LA, Sullivan A, Bell A, Dunne B, Rozycka Beersum SE, Celli J, Merkx GF, Tenconi R, Fryns JP, Verloes
M, Reddy A, Tidy JA, Evans D, Farrell PJ, Evans A, Gasco A, Newbury-Ecob RA, Raas-Rotschild A, Majewski F,
M, Gusterson B, Crook T. 2001. p73 is over-expressed in Beemer FA, Janecke A, Chitayat D, Crisponi G,
vulval cancer principally as the Delta 2 isoform. Br J Cancer Kayserili H, Yates JR, Neri G, Brunner HG. 2001. p63
85:1551–1556. gene mutations in EEC syndrome, limb-mammary syndrome,
Ozaki T, Naka M, Takda N, Tada M, Sakiyama S, Nakagawara and isolated split hand-split foot malformation suggest a
A. 1999. Deletion of the COOH-terminal region of p73alpha genotype–phenotype correlation. Am J Hum Genet 69:481–
enhances both its transactivation function and DNA-binding 492.
activity but inhibits induction of apoptosis in mammalian Versteeg R, Caron H, Cheng N, Van der Drift P, Slater R,
cells. Cancer Res 58:5902–5907. Westerveld A, Voûte P, Delattre O, Laureys G, Van Roy N,
Park JJ, Sun D, Quade BJ, Flynn C, Sheets EE, Tang A, Speleman F. 1995. 1p36 : every subband a suppressor? Eur J
McKeon F, Crum CP. 2000. Stratified mucin-producing Cancer 31A:538–541.
intraepithelial lesions of the cervix: adenosquamous Wang X, Arooz T, Siu WY, Chiu CH, Lau A, Yamashita K,
or columnar cell neoplasia? Am J Surg Pathol 24:1414– Poon RY. 2001. MDM2 and MDMX can interact differently
1419. with ARF and members of the p53 family. FEBS Lett
Patturajan M, Nomoto S, Sommer M, Hibi K, Zangen R, Poliak 490:202–208.
N, Califano J, Trink B, Ratoviski E, Sidransky D. 2002. Yang A, Kaghad M, Wang Y, Gillett E, Fleming MD, Dotsch V,
DNp63 induces b-catenin nuclear accumulation and signal- Andrews NC, Caput D, McKeon F. 1998. p63, a p53
ing. Cancer Cell 1:369–379. homolog at 3q27-29, encodes multiple products with
Pozniak CD, Radinovic S, Yang A, McKeon F, Kaplan DR, transactivating death-inducing and dominant negative
Miller FD. 2000. An anti-apoptotic role for the p53 family activities. Mol Cell 2:305–316.
member, p73, during developmental neuron death. Science Yang A, Schweitzer R, Sun D, Kaghad M, Walker N, Bronson
289:304–306. RT, Tabin C, Sharpe A, Caput D, Crum C, McKeon F. 1999.
Rotter V, Aloni-Grinstein R, Schwartz D, Elkind NB, Simons p63 is essential for regenerative proliferation in limb,
A, Wolkowicz R, Lavigne M, Beserman P, Kapon A, craniofacial and epithelial development. Nature 398:
Goldfinger N. 1994. Does wild-type p53 play a role 714–718.
TP53 FAMILY MEMBERS 191

Yang A, Walker N, Bronson R, Kaghad M, Oosterwegel M, Zaika A, Irwin M, Sansome C, Moll UM. 2001. Oncogenes
Bonin J, Vagner C, Bonnet H, Dikkes P, Sharpe A, McKeon F, induce and activate endogenous p73 protein. J Biol Chem
Caput D. 2000. P73-deficient mice have neurobiological, 276:11310–11316.
pheromonal and inflammatory defects but lack spontaneous Zeng X, Chen L, Jost CA, Maya R, Kelle D, Wang X, Kaelin
tumors. Nature 404:99–103. WG, Jr., Oren M, Chen J, Lu H. 1999. MDM2 suppresses
Yang A, Kaghad M, Caput D, McKeon F. 2002. On the shoulders p73 function without promoting p73 degradation. Mol Cell
of giants: p63, p73 and the rise of p53. Trends Genet 18:90–95. Biol 19:3257–3266.
Zaika AL, Kovalev S, Marchenko ND, Moll UM. 1999. Zhu J, Jiang J, Zhou W, Chen X. 1998. The potential tumor
Overexpression of the wild type p73 gene in breast cancer suppressor p73 differentially regulates cellular p53 target
tissues and cell lines. Cancer Res 59:3257–3263. genes. Cancer Res 58:5061–5065.

Vous aimerez peut-être aussi