Vous êtes sur la page 1sur 6

478 Review TRENDS in Cell Biology Vol.13 No.

9 September 2003

PTEN: from pathology to biology


Maria Luisa Sulis1,2 and Ramon Parsons1,3
1
Institute for Cancer Genetics, College of Physicians & Surgeons, Columbia University, New York, USA
2
Division of Pediatric Oncology, Childrens Hospital of New York, Herbert Irving Cancer Center, College of Physicians & Surgeons,
Columbia University, New York, USA
3
Departments of Pathology and Medicine, Herbert Irving Cancer Center, College of Physicians & Surgeons, Columbia University,
New York, USA

The PTEN tumour suppressor gene is mutated fre- Because this review is limited in length we recommend
quently in many malignancies and its importance in the several comprehensive reviews for historical perspective
development of cancer is probably underestimated. As [1 4]. Moreover, most of the proteins discussed here have
the primary phosphatase of phosphatidylinositol complex regulatory inputs from signalling partners other
(3,4,5)-trisphosphate, PTEN has a central role in reign- than PTEN that is not covered in depth for the sake of
ing in the phosphoinositide 3-kinase (PI 3-kinase) net- simplicity and brevity. We focus on the most recent
work to control cellular homeostasis. Cells that lack progress in understanding the role of PTEN in oncogenesis
PTEN are unable to regulate the PtdIns 3-kinase pro- as well as the regulation of normal cellular functions.
gramme, which stimulates a variety of cellular pheno- PTEN has a tyrosine phosphatase domain with dual-
types that favour oncogenesis. As well as the well- specificity protein-phosphatase and lipid-phosphatase
known role as tumour suppressor, recent studies show activity in vitro [5]. The lipid-phosphatase activity of
that PTEN is involved in the regulation of several basic PTEN is highly specific. PTEN recognizes the substrate
cellular functions, such as cell migration, cell size, con- phosphatidylinositol (3,4,5)-trisphosphate [PtdIns(3,4,5)P3]
tractility of cardiac myocytes and chemotaxis. Here, we and removes the D3 phosphate from the inositol ring.
review the roles of PTEN in normal cellular functions Work from many groups has established a strong
and disease development. foundation for the biological importance of the lipid
phosphatase activity of PTEN and here we focus on
The discovery of phosphatase and tensin homolog on this enzymatic function of PTEN (Fig. 1).
chromosome ten (PTEN, also known as MMAC1 and The level of PtdIns(3,4,5)P3 is very low in resting cells
TEP1) has been like the opening of Pandoras box. It has that contain wild type PTEN. Activation of cell-surface
provided clues for understanding tumourigenesis as well receptors recruits PI 3-kinase to the receptor where it
as other aspects of biology. In a relatively short time, a phosphorylates the PtdIns(4,5)P2 substrate to generate
highly complex signalling network has been refined and PtdIns(3,4,5)P3. The half-life of PtdIns(3,4,5)P3 is short
the pivotal role of PTEN in regulating many biological and it is metabolized by PTEN and the D5 phosphatases,
functions has been established. including SHIP1 and SHIP2 [6]. Cells that lack PTEN
PTEN is a tumour suppressor gene that is located on have elevated levels of PtdIns(3,4,5)P3, even in the resting
chromosome 10q23. Germline mutations of PTEN cause state. PtdIns(3,4,5)P3 is a potent second messenger that
four rare autosomal-dominant syndromes that have similar binds proteins that contain pleckstrin-homology (PH)
clinical features: Cowden Syndrome; Lhermitte-Duclos syn- domains to elicit several cellular behaviours that favour
drome; Bannayan-Zonana syndrome; and Proteus syndrome. oncogenesis. Among the most important PH-domain-
These diseases share the tendency to develop hamartomas containing proteins are the AKT family (AKT1, AKT2
and other benign tumours, and Cowden Syndrome is and AKT3) and their regulator phosphoinositide-depen-
associated with the development of malignant tumours [1]. dent kinase 1 (PDK1). Loss of PTEN creates a state in
Somatic mutations of PTEN are documented in an increasing which the PI 3-kinase pathway is constitutively active.
variety of malignant tumours at both advanced and early This stimulates cell division, increases cell size and
stages; these mutations lead to complete inactivation of the angiogenesis, and inhibits apoptosis. In addition, cell
phosphatase activity of PTEN as well as to either total or migration is often increased but can also be impaired.
partial loss of expression of either mRNA and/or protein [2].
The frequency of PTEN mutation rivals that of p53 [1]. Like PTEN: a small gene family lacking redundancy
p53, mutation of PTEN is associated with the inactivation of The gene that encodes PTEN is expressed in all eukaryotic
both alleles. Understanding the reason for the preferential cells. The crystal structure of human PTEN shows
selection of PTEN mutations in tumours is fundamental to adjacent phosphatase and C2-domain lobes [7] that is
cancer biology and it is now clear that a major selective predicted to be preserved in all metazoans. PTEN
pressure for inactivating PTEN is to stimulate the homologues n Saccharomyces cerevisiae and Schizosac-
phosphatidylinositol 3-kinase (PI 3-kinase) pathway. charomyces pombe fungi have the phosphatase domain,
but they lack the C2 domain [8] that is crucial for
Corresponding author: Ramon Parsons (rep15@columbia.edu). interaction with the plasma membrane. Although there
http://ticb.trends.com 0962-8924/$ - see front matter q 2003 Elsevier Ltd. All rights reserved. doi:10.1016/S0962-8924(03)00175-2
Review TRENDS in Cell Biology Vol.13 No.9 September 2003 479

tumour progression in humans is controversial. However,


Unstimulated Stimulated
Pten/2 mice develop colonic adenomas and lymph node
hyperplasia at high frequency because of haploinsuffi-
PI3K ciency [18]. In recent work with a mouse SV40 T antigen
PTEN prostate-tumour model, PTEN haploinsufficiency stimu-
lates tumour progression [19]. In addition, many tumours
PIP3
have lowered PTEN expression of poorly defined origin.
Stable loss of PTEN expression for any reason is a
significant factor in the development of a tumour because
it leads to the deregulation of PI 3-kinase signalling.

The PTEN2/2 cellular phenotypes


Initial experiments in mice showed that complete loss of
PTEN is lethal early in development. Heterozygous mice
are viable, however, and adults develop a variety of
tumours [1]. Recently, several groups have analysed the
effects of conditionally mutating both alleles of PTEN by
incorporating lox recombination sites that flank PTEN
TRENDS in Cell Biology exons and using tissue-specific promoters to express Cre
recombinase. Recombination elicited by Cre led to the
Fig. 1. Proposed model for phosphoinositide 3-kinase (PI 3-kinase)/PTEN mediated
mutational inactivation of PTEN. Surprisingly, loss of
chemotaxis. In resting cells PTEN is distributed uniformly at the membrane,
whereas PI 3-kinase is in the cytoplasm. Following chemoattractant stimulation PTEN by itself in these mice does not appear to be
with cAMP, activated PI 3-kinase and its product, phosphatidylinositol (3,4,5)-tri- sufficient for oncogenesis. Instead, a period of time is
sphosphate [PtdIns(3,4,5)P3], localize at the leading edge of the cell, whereas PTEN
localizes at the rear and sides. The gradient in PtdIns(3,4,5)P3 that results allows
required for a subpopulation of PTEN2/2 cells to trans-
pseudopods to form [48 50]. form into malignancies. As of now, PTEN has been
conditionally inactivated in mouse T cells, B cells, cardiac
is only one PTEN gene in fungi and lower metazoans myocytes, mammary epithelium, primordial germ cells,
(Caenorhabditis elegans and Drosophila melanogaster), neurons and keratinocytes [20 25]. In all cases, the loss of
additional orthologues, including TPTE, PTEN 2 and PTEN did not lead to tumour formation initially. Although
TPIP, have evolved in mammals [9 11]. In addition to the unexpected, these data are not unprecedented and
phosphatase and C2 domains these proteins have multiple indicate that other, unknown, genetic alterations that
transmembrane domains at their amino termini. Like favour oncogenesis cooperate with PTEN inactivation to
PTEN, PTEN 2 and TPIP are PtdIns(3,4,5)P3 phospha- produce malignancy. However, several signalling events
tases in vitro. However, unlike PTEN, which is expressed and phenotypes are observed consistently, which indicates
ubiquitously, expression of TPTE and PTEN 2 is restricted a common PTEN pathway.
to the testis, and TPIP is expressed only in the testis, brain Cells that lack PTEN have elevated levels of
and stomach. Analysis of the cellular localization of these PtdIns(3,4,5)P3, phosphorylated AKT and p70 S6 kinase
transmembrane homologs of PTEN demonstrates that the (S6K) [21,23]. These changes are associated with increased
proteins are limited to either the Golgi apparatus or proliferation and reduced apoptosis in T cells, B cells,
endoplasmic reticulum. Based on their defined tissue- immature neurons, keratinocytes and mammary epi-
expression patterns and lack of access to the plasma thelium [24,26]. Paradoxically however, in primordial
membrane, these orthologues are not expected to regulate germ cells increased proliferation is associated with
PI 3-kinase signalling at the plasma membrane. Overall, increased apoptosis [25]. Increased cell size is observed
the lack of redundancy of PTEN at the plasma membrane in neurons, cardiac myocytes and T cells. Altered
might explain the high frequency with which PTEN migration is a characteristic of immature neurons and
inactivation is selected during tumour development. reduced contractility occurs in cardiac myocytes. The
plethora of phenotypes that are associated with mutation
Reduced expression of PTEN in tumours of PTEN appear to be caused by accumulation of
Genetic alteration of both alleles of PTEN occurs in nearly PtdIns(3,4,5)P3. For example, in myocytes, a dominant-
all types of human cancers examined, with the highest negative PI 3-kinase-a reverses the increase in cell size,
frequency of inactivation in glioblastoma and endometrial whereas deletion of PI 3-kinase-g to restores myocyte
cancer [12 15]. The typical mechanism of inactivation is contractility.
mutation accompanied by loss of heterozygosity, the gold The effect of PTEN mutation has also been studied in
standard for gene inactivation. However, in some cases several model organisms. As with mice, mutation of PTEN
tumours appear to evolve mechanisms to reduce the in Drosophila alters cell size, proliferation, apoptosis and
concentration of PTEN without mutation of the gene migration [27 30]. Tissue-specific mutation of PTEN in
[16]. Methylation of the PTEN promoter region, which is mice reveals multiple phenotypes, but inactivation of
associated with reduced PTEN expression, appears to PTEN in this context is not sufficient to produce tumours.
occur in some cancers [17]. Loss of one allele of PTEN with These data demonstrate that inactivation of PTEN is
the preservation of the remaining wild-type allele is also insufficient to cause tumour development but that it
very common. Whether PTEN haploinsufficiency favours creates an environment that selects for tumour growth.
http://ticb.trends.com
480 Review TRENDS in Cell Biology Vol.13 No.9 September 2003

Cellular localization of PTEN membrane has a pivotal role in the chemotactic response of
Because PTEN regulates PtdIns(3,4,5)P3 at the plasma the cell to a cAMP chemoattractant stimulus by creating a
membrane, some portion of PTEN must reside at this gradient of PtdIns(3,4,5)P3 that enhances intracellular
location in the cell. Consistent with this, Das et al. found signalling and directional movement [43 45]. By contrast,
that a mutant version of PTEN that contains the cells with reduced PI 3-kinase at the membrane move
phosphatase and C2 domains fused with green fluorescent slowly, lose polarity and formed multiple pseudopodia
protein (GFP) is expressed at the plasma membrane [31]. [46 48]. Interestingly, Iijima et al. have showed that the
Thus, it is surprising that most endogenous PTEN is not effects of loss of PTEN in D. discoideum are similar to those
found at the plasma membrane. Immunohistochemical in PI 3-kinase-null cells; although PTEN-null cells can
studies show that the cellular distribution of PTEN varies sense chemoattractants, as shown by a change in shape,
between tissues. In most epithelial cells, such as skin, they are unable to move toward them and form multiple,
colon, breast and prostate, the bulk of PTEN is in the changing pseudopodia [49]. In addition, localization and
cytoplasm [32,33]. By contrast, in neurons, fibroblasts, and translocation of PTEN following a chemoattractant stimu-
cells of the adrenal medulla and thyroid most PTEN is in lus is reciprocal to PI 3-kinase localization [49,50]. The
the nucleus [34 36]. Examination of polarized MDCK above studies delineate a model in which a stimulus causes
cells indicates that PTEN localizes at cell cell tight PI 3-kinase to localize to the front edge of the membrane,
junctions [37]. The basis for the different tissue-localiz- which leads to PtdIns(3,4,5)P3 accumulation, AKT acti-
ation patterns is unknown but it might involve the 50 vation and the formation of a pseudopod. At the same time,
amino acids at the carboxy-terminal of PTEN. This region PTEN dissociates from the front edge of the membrane and
contains multiple phosphorylation sites for casein kinase localizes posteriorly, thus creating a steeper gradient of
II and a PDZ-binding domain that can bind to membrane- PtdIns(3,4,5)P3 (Fig. 2).
associated guanylate kinase inverted (MAGI) proteins
[31,37]. Recent studies by Ginn-Pease and Eng show that PTEN and PtdIns(3,4,5)P3 targets
the concentration of PTEN in the nucleus parallels the Many proteins that containing PH domains bind to
cytoplasmic levels during the cell cycle. The highest PtdIns(3,4,5)P3 with high affinity. In PTEN2/2 cells, a
concentration of nuclear as well as cytoplasmic PTEN large number of different signalling proteins are brought
are associated with the G0 G1 phase, which indicates a to the plasma membrane by elevated PtdIns(3,4,5)P3
possible role of nuclear PTEN in the well-known, PTEN- levels. Surprisingly, however, the primary phenotypic
related G1 arrest [38]. These data are intriguing. output of elevated PtdIns(3,4,5)P3 in metazoans streams
Localization of PTEN in the cytoplasm and nucleus through PDK1 to its substrate AKT. Although much
might function merely to separate it from the plasma experimental data substantiates the importance of the
membrane; alternatively, PTEN might participate actively link between PTEN and AKT, perhaps the best evidence is
in cellular processes in the cytoplasm and nucleus. the ability of a hypomorphic allele of AKT to rescue
Changes in the subcellular localization of PTEN have PTEN2/2 Drosophila embryos from lethality [51]. Other
been reported in several malignancies. PTEN is localized model systems also demonstrate the importance of PDK1
mostly in nucleus in normal pancreatic islets, but occurs and AKT. In C. elegans, PTEN2/2 phenotypes are
predominantly in the cytoplasm of 80% of endocrine ameliorated by double-strand RNA interference of AKT1
pancreatic tumours [39]. Similarly, in thyroid carcinoma and AKT2 [52]. Analysis of PDK1 mutants in Drosophila
and melanoma the decrease in nuclear staining of PTEN is demonstrates that PDK1 is essential for PI 3-kinase-
more marked than in the cytoplasm, particularly in mediated activation of AKT [53]. Similarly, in C. elegans,
undifferentiated and metastatic tumours [36,40]. The PDK1 is necessary and sufficient to transmit signals
reasons for alterations of PTEN in the nucleus are obscure. from PI 3-kinase to AKT1 and AKT2 [54]. Therefore, the
Whether and how a shift in the cellular localization of type I PI 3-kinase PtdIns(3,4,5)P3 PDK1 AKT axis is
PTEN might have a role in the pathogenesis of tumours the major developmental pathway that involves
remains to be elucidated. PtdIns(3,4,5)P3 and is modulated by PTEN in animals.

PTEN and cell migration AKT substrates


It has been shown that PTEN might exert effects on the Many AKT substrates are phosphorylated when PTEN is
cytoskeleton and have a role in controlling cell migration. inactive (Fig. 2) Alterations in the transcription profiles
Introducing PTEN into PTEN2/2 human tumour cells are caused, at least in part, by changes in activity of
alters actin fibres and inhibits cell migration [41]. nuclear factor kB, HIF1-a and forkhead transcription
Migration is increased in mouse embryo fibroblasts that factors [55 57]. Phosphorylation and inhibition of GSK3
lack PTEN. In these cells, elevated PtdIns(3,4,5)P3 leads to (which increases cyclin D and myc levels) [58,59], and
activation of Rac1 and Cdc42, both of which are small phosphorylation and cytoplasmic sequestration of p27
GTPase mediators of cellular migration [42]. alters the cell cycle. Apoptosis is inhibited by phosphoryl-
In the past 2 years, several studies in Dictyostelium ation of caspase 9 and [60,61]. Recent data indicates that
discoideum have started to define a sophisticated mechan- AKT might inhibit p53-dependent apoptosis: several
ism by which the concerted, opposing action of PI 3-kinase groups have shown that AKT interacts and phosphorylates
and PTEN finely regulate cell motility in response to a Mdm2. Phosphorylation induces the nuclear localization of
point source of extracellular cAMP. Initial studies showed Mdm2, which results in the subsequent export and
that PI 3-kinase localized at the leading edge of the degradation of p53 in the cytoplasm [62,63]. Inhibition of
http://ticb.trends.com
Review TRENDS in Cell Biology Vol.13 No.9 September 2003 481

PI3K
PI(4,5)P2 PI(3,4,5)P3
PTEN
PH PH
A P
K D
T K

FKHRLI GSK3
FKHR TSC2-TSC1
p21
p27
Caspase 9 mTOR S6K
Cyclin D myc BAD pMDM2
pNFKB

Cell Cell growth


Cell cycle survival translation
entry

TRENDS in Cell Biology

Fig. 2. Schematic representation of the role of PTEN in the phosphoinositide 3-kinase (PI 3-kinase)/Akt pathway. PTEN dephosphorylates phosphatidylinositol (3,4,5)-tri-
sphosphate [PtdIns(3,4,5)P3], at the D3 position. A lower amount of PtdIns(3,4,5)P3 at the membrane decreases the concentration of phosphorylated (active) Akt, thereby
downregulating downstream effectors. As a result, PTEN regulates cell growth, survival and entry into the cell cycle.

AKT, either by PTEN or by expression of a dominant- not the catalytically inactive mutant, inhibited phos-
negative form of AKT, sensitizes tumour cells to DNA- phorylation of TSC2. AKT-mediated phosphorylation of
damaging agents, perhaps by restoring p53 activity TSC2 results in the degradation of the TSC1 TSC2
[62,64]. The interaction between p53 and PTEN is likely complex, which regulates mTOR and thereby S6K [68].
to be complex and occur at different levels. Stambolic et al. Therefore, cells lacking either TSC1 or TSC2 have
demonstrated direct binding of p53 to the PTEN promoter elevated S6K activity. Both TSC1 and TSC2 are tumour-
and the levels of PTEN mRNA increase in several cell lines suppressor genes that are mutated in TSC, a syndrome
following induction of p53 [65]. In addition, PTEN is that is associated with benign hamartomatous tumours
required for p53-mediated apoptosis in PTEN-null mouse and CNS abnormalities. The ability of AKT to phosphor-
embryonic fibroblasts. More recently, Freeman et al. [66] ylate TSC2 is linked directly to its ability to stimulate cell
have performed GST-pull down experiments that show growth, because phosphorylation-site mutants of TSC2 are
that the C-terminus of p53 binds to the C2 domain of resistant to AKT-mediated growth signals [69]. Given that
PTEN. This report also provides evidence that wild-type PTEN and TSC tumour suppressors regulate S6K, it is
PTEN, as well as the catalytically inactive form, stabilize likely to play a role in tumour development. It is also
p53 through an MDM2-independent mechanism. It seems interesting that, although hamartomas develop because of
likely that a connection exists between these two major germline mutations in PTEN and TSC, PTEN mutation
tumour suppressors, although the precise mechanisms of are a much more potent stimulus for malignant trans-
interaction and their functional importance need to be formation. This might be due to the more upstream
defined completely. Important clinical implications might location of PTEN in the signalling pathway and, therefore,
result from subsequent studies. the larger number of targets affected by its mutation.

S6K and tuberous sclerosis (TSC) Concluding remarks


Although the importance of the PDK1 AKT connection is The discovery of PTEN represents a milestone in the
undisputed, other targets of PDK1 such as S6K might also understanding of tumourigenesis and solves many pieces
contribute to tumour development. S6K is a regulator of of a complicated puzzle. But, as always in science,
cell size that requires PtdIns(3,4,5)P3, PDK1 and the answering one question opens up new ones. In the past
mammalian target of rapamycin (mTOR) to be activated few years, many downstream targets of PTEN have been
(Fig. 1). Mammalian cells that lack PTEN have elevated identified and the fundamental role of the PI 3-kinase
S6K activity [67]. Recently, it has been shown that TSC1 PTEN AKT axis has been further elucidated, but there is
and TSC2 form a complex that inactivates mTOR and so still much to be explained. Little is known about the
inhibits the activity of S6K [68]. This finding led to further regulation of PTEN transcription and translation, and the
investigations on the possible interaction between half-life, cellular localization and enzymatic activity of
TSC1 TSC2 and AKT/PTEN. Experiments in vitro and PTEN protein. It is likely that mutations in the regulatory
in vivo show that coexpression of TSC1 TSC2 with wild- circuitry that lead to altered PTEN function will, in turn,
type AKT, but not a kinase-inactive AKT mutant, results cause tumour development. The rapid activation and
in phosphorylation of TSC2, but no effect on TSC1 was inactivation kinetics of PtdIns(3,4,5)P3 hints at the
noted [68]. As expected, expression of wild-type PTEN, but possibility that PTEN has equally rapid regulatory
http://ticb.trends.com
482 Review TRENDS in Cell Biology Vol.13 No.9 September 2003

mechanisms. The ability of PTEN to reside in the nucleus 17 Soria, J.C. et al. (2002) Lack of PTEN expression in non-small cell lung
indicates that PTEN might function to regulate a nuclear cancer could be related to promoter methylation. Clin. Cancer Res. 8,
1178 1184
PI 3-kinase pathway. 18 Podsypanina, K. et al. (1999) Mutation of Pten/Mmac1 in mice causes
Another fascinating area for study is why PTEN is such neoplasia in multiple organ systems. Proc. Natl. Acad. Sci. U. S. A. 96,
a potent tumour suppressor. In tumour development, 1563 1568
PTEN inactivation does not appear to be equivalent to 19 Kwabi-Addo, B. et al. (2001) Haploinsufficiency of the Pten tumour
AKT activation [70]. This indicates that, as yet unknown, suppressor gene promotes prostate cancer progression. Proc. Natl.
Acad. Sci. U. S. A. 98, 11563 11568
PI 3-kinase-dependent and independent outputs of PTEN 20 Suzuki, A. et al. (2001) T cell-specific loss of Pten leads to defects in
might influence cancer development. However, many central and peripheral tolerance. Immunity 14, 523 534
questions remain, such as what is the exact role of the 21 Crackower, M.A. et al. (2002) Regulation of myocardial contractility
protein phosphatase activity of PTEN, and is and cell size by distinct PI3K-PTEN signaling pathways. Cell 110,
PtdIns(3,4,5)P3 the only substrate for the lipid phospha- 737 749
22 Kwon, C.H. et al. (2001) Pten regulates neuronal soma size: a mouse
tase, how is PTEN inactivated in tumours, why does the model of Lhermitte-Duclos disease. Nat. Genet. 29, 404 411
localization of PTEN change in different tissues, and what 23 Li, G. et al. (2002) Conditional loss of PTEN leads to precocious
cooperating genetic events are favoured in PTEN-deficient development and neoplasia in the mammary gland. Development 129,
tumours? 4159 4170
The high prevalence of alterations in PTEN in a variety 24 Backman, S.A. et al. (2001) Deletion of Pten in mouse brain causes
seizures, ataxia and defects in soma size resembling Lhermitte-Duclos
of human tumours stresses the need for better under-
disease. Nat. Genet. 29, 396 403
standing of the normal and pathological regulatory 25 Kimura, T. et al. (2003) Conditional loss of PTEN leads to testicular
mechanisms of PTEN. Improved knowledge of PTEN teratoma and enhances embryonic germ cell production. Development
will lead to a fuller understanding of human oncogenesis. 130, 1691 1700
26 Ashcroft, M. et al. (2002) Phosphorylation of HDM2 by Akt. Oncogene
21, 1955 1962
References
27 Oldham, S. et al. (2002) The Drosophila insulin/IGF receptor controls
1 Cantley, L.C. and Neel, B.G. (1999) New insights into tumour
growth and size by modulating PtdInsP(3) levels. Development 129,
suppression: PTEN suppresses tumour formation by restraining the
4103 4109
phosphoinositide 3-kinase/AKT pathway. Proc. Natl. Acad. Sci. U. S. A.
28 Gao, X. et al. (2000) Drosophila PTEN regulates cell growth and
96, 4240 4245
proliferation through PI3K-dependent and -independent pathways.
2 Simpson, L. and Parsons, R. (2001) PTEN: life as a tumour suppressor.
Dev. Biol. 221, 404 418
Exp. Cell Res. 264, 29 41
29 Scanga, S.E. et al. (2000) The conserved PI30 K/PTEN/Akt signaling
3 Maehama, T. and Dixon, J.E. (1999) PTEN: a tumour suppressor that
pathway regulates both cell size and survival in Drosophila. Oncogene
functions as a phospholipid phosphatase. Trends Cell Biol. 9, 125 128
19, 3971 3977
4 Di Cristofano, A. and Pandolfi, P.P. (2000) The multiple roles of PTEN
30 Huang, H. et al. (1999) PTEN affects cell size, cell proliferation and
in tumour suppression. Cell 100, 387 390
5 Maehama, T. and Dixon, J.E. (1998) The tumour suppressor, PTEN/ apoptosis during Drosophila eye development. Development 126,
MMAC1, dephosphorylates the lipid second messenger, phosphatidyl- 5365 5372
inositol 3,4,5-trisphosphate. J. Biol. Chem. 273, 13375 13378 31 Das, S. et al. (2003) Membrane-binding and activation mechanism of
6 Lioubin, M.N. et al. (1996) p150Ship, a signal transduction molecule PTEN. Proc. Natl. Acad. Sci. U. S. A. 100, 7491 7496
with inositol polyphosphate-5-phosphatase activity. Genes Dev. 10, 32 Deichmann, M. et al. (2002) PTEN/MMAC1 expression in melanoma
1084 1095 resection specimens. Br. J. Cancer 87, 1431 1436
7 Lee, J.O. et al. (1999) Crystal structure of the PTEN tumour 33 McMenamin, M.E. et al. (1999) Loss of PTEN expression in paraffin-
suppressor: implications for its phosphoinositide phosphatase activity embedded primary prostate cancer correlates with high Gleason score
and membrane association. Cell 99, 323 334 and advanced stage. Cancer Res. 59, 4291 4296
8 Li, L. et al. (1997) A family of putative tumour suppressors is 34 Lachyankar, M.B. et al. (2000) A role for nuclear PTEN in neuronal
structurally and functionally conserved in humans and yeast. J. Biol. differentiation. J. Neurosci. 20, 1404 1413
Chem. 272, 29403 29406 35 Sano, T. et al. (1999) Differential expression of MMAC/PTEN in
9 Chen, H. et al. (1999) A testis-specific gene, TPTE, encodes a putative glioblastoma multiforme: relationship to localization and prognosis.
transmembrane tyrosine phosphatase and maps to the pericentro- Cancer Res. 59, 1820 1824
meric region of human chromosomes 21 and 13, and to chromosomes 36 Gimm, O. et al. (2000) Differential nuclear and cytoplasmic expression
15, 22, and Y. Hum. Genet. 105, 399 409 of PTEN in normal thyroid tissue, and benign and malignant epithelial
10 Wu, Y. et al. (2001) PTEN 2, a Golgi-associated testis-specific thyroid tumours. Am. J. Pathol. 156, 1693 1700
homologue of the PTEN tumour suppressor lipid phosphatase. 37 Wu, X. et al. (2000) Evidence for regulation of the PTEN tumour
J. Biol. Chem. 276, 21745 21753 suppressor by a membrane-localized multi-PDZ domain containing
11 Walker, S.M. et al. (2001) TPIP: a novel phosphoinositide 3-phosphatase. scaffold protein MAGI-2. Proc. Natl. Acad. Sci. U. S. A. 97, 4233 4238
Biochem. J. 360, 277283 38 Ginn-Pease, M.E. and Eng, C. (2003) Increased nuclear phosphatase
12 Li, J. et al. (1997) PTEN, a putative protein tyrosine phosphatase gene and tensin homologue deleted on chromosome 10 is associated with
mutated in human brain, breast, and prostate cancer. Science 275, G0-G1 in MCF-7 cells. Cancer Res. 63, 282 286
1943 1947 39 Perren, A. et al. (2000) Mutation and expression analyses reveal
13 Risinger, J.I. et al. (1997) PTEN/MMAC1 mutations in endometrial differential subcellular compartmentalization of PTEN in endocrine
cancers. Cancer Res. 57, 4736 4738 pancreatic tumours compared to normal islet cells. Am. J. Pathol. 157,
14 Tashiro, H. et al. (1997) Mutations in PTEN are frequent in 1097 1103
endometrial carcinoma but rare in other common gynecological 40 Whiteman, D.C. et al. (2002) Nuclear PTEN expression and clinico-
malignancies. Cancer Res. 57, 3935 3940 pathologic features in a population-based series of primary cutaneous
15 Steck, P.A. et al. (1997) Identification of a candidate tumour suppressor melanoma. Int. J. Cancer 99, 63 67
gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple 41 Tamura, M. et al. (1998) Inhibition of cell migration, spreading, and
advanced cancers. Nat. Genet. 15, 356 362 focal adhesions by tumour suppressor PTEN. Science 280, 1614 1617
16 Whang, Y.E. et al. (1998) Inactivation of the tumour suppressor PTEN/ 42 Liliental, J. et al. (2000) Genetic deletion of the Pten tumour
MMAC1 in advanced human prostate cancer through loss of suppressor gene promotes cell motility by activation of Rac1 and
expression. Proc. Natl. Acad. Sci. U. S. A. 95, 5246 5250 Cdc42 GTPases. Curr. Biol. 10, 401 404
http://ticb.trends.com
Review TRENDS in Cell Biology Vol.13 No.9 September 2003 483

43 Parent, C.A. et al. (1998) G protein signaling events are activated at 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells:
the leading edge of chemotactic cells. Cell 95, 81 91 implications for tumour angiogenesis and therapeutics. Cancer Res.
44 Servant, G. et al. (2000) Polarization of chemoattractant receptor 60, 1541 1545
signaling during neutrophil chemotaxis. Science 287, 1037 1040 57 Brunet, A. et al. (1999) Akt promotes cell survival by phosphorylating
45 Comer, F.I. and Parent, C.A. (2002) PI 3-kinases and PTEN: how and inhibiting a Forkhead transcription factor. Cell 96, 857 868
opposites chemoattract. Cell 109, 541 544 58 Sears, R. et al. (2000) Multiple Ras-dependent phosphorylation
46 Chung, C.Y. et al. (2001) Control of cell polarity and chemotaxis by Akt/ pathways regulate Myc protein stability. Genes Dev. 14, 2501 2514
PKB and PI3 kinase through the regulation of PAKa. Mol. Cell 7, 59 Cross, D.A. et al. (1995) Inhibition of glycogen synthase kinase-3 by
937 947 insulin mediated by protein kinase B. Nature 378, 785 789
47 Funamoto, S. et al. (2001) Role of phosphatidylinositol 30 kinase and a 60 Cardone, M.H. et al. (1998) Regulation of cell death protease caspase-9
downstream pleckstrin homology domain-containing protein in con- by phosphorylation. Science 282, 1318 1321
trolling chemotaxis in dictyostelium. J. Cell Biol. 153, 795 810 61 Datta, S.R. et al. (1997) Akt phosphorylation of BAD couples survival
48 Funamoto, S. et al. (2002) Spatial and temporal regulation of signals to the cell-intrinsic death machinery. Cell 91, 231 241
3-phosphoinositides by PI 3-kinase and PTEN mediates chemotaxis. 62 Zhou, B.P. et al. (2001) HER-2/neu induces p53 ubiquitination via Akt-
Cell 109, 611 623 mediated MDM2 phosphorylation. Nat. Cell Biol. 3, 973 982
49 Iijima, M. and Devreotes, P. (2002) Tumour suppressor PTEN mediates 63 Mayo, L.D. and Donner, D.B. (2001) A phosphatidylinositol 3-kinase/
sensing of chemoattractant gradients. Cell 109, 599 610 Akt pathway promotes translocation of Mdm2 from the cytoplasm to
50 Iijima, M. et al. (2002) Temporal and spatial regulation of chemotaxis. the nucleus. Proc. Natl. Acad. Sci. U. S. A. 98, 11598 11603
Dev. Cell 3, 469 478 64 Mayo, L.D. et al. (2002) PTEN protects p53 from Mdm2 and sensitizes
51 Stocker, H. et al. (2002) Living with lethal PIP3 levels: viability of flies cancer cells to chemotherapy. J. Biol. Chem. 277, 5484 5489
lacking PTEN restored by a PH domain mutation in Akt/PKB. Science 65 Stambolic, V. et al. (2001) Regulation of PTEN transcription by p53.
295, 2088 2091 Mol. Cell 8, 317 325
52 Ogg, S. and Ruvkun, G. (1998) The C. elegans PTEN homolog, DAF-18, 66 Freeman, D.J. et al. (2003) PTEN tumour suppressor regulates p53
acts in the insulin receptor-like metabolic signaling pathway. Mol. Cell protein levels and activity through phosphatase-dependent and -
2, 887 893 independent mechanisms. Cancer Cell 3, 117 130
53 Cho, K.S. et al. (2001) Drosophila phosphoinositide-dependent kinase- 67 Lu, Y. et al. (1999) The PTEN/MMAC1/TEP tumour suppressor gene
1 regulates apoptosis and growth via the phosphoinositide 3-kinase- decreases cell growth and induces apoptosis and anoikis in breast
dependent signaling pathway. Proc. Natl. Acad. Sci. U. S. A. 98, cancer cells. Oncogene 18, 7034 7045
6144 6149 68 Inoki, K. et al. (2002) TSC2 is phosphorylated and inhibited by Akt and
54 Paradis, S. et al. (1999) A PDK1 homolog is necessary and sufficient to suppresses mTOR signalling. Nat. Cell Biol. 4, 648 657
transduce AGE-1 PI3 kinase signals that regulate diapause in 69 Radimerski, T. et al. (2002) Lethality of Drosophila lacking TSC
Caenorhabditis elegans. Genes Dev. 13, 1438 1452 tumour suppressor function rescued by reducing dS6K signaling.
55 Romashkova, J.A. and Makarov, S.S. (1999) NF-kappaB is a target of Genes Dev. 16, 2627 2632
AKT in anti-apoptotic PDGF signalling. Nature 401, 86 90 70 Majumder, P.K. et al. (2003) Prostate intraepithelial neoplasia induced
56 Zhong, H. et al. (2000) Modulation of hypoxia-inducible factor 1alpha by prostate restricted Akt activation: The MPAKT model. Proc. Natl.
expression by the epidermal growth factor/phosphatidylinositol Acad. Sci. U. S. A. 100, 7841 7846

TCB: making the most of your personal subscription


High quality printouts (from PDF files)
Links to other articles, other journals and cited software and databases
All you have to do is:
Obtain your subscription key from the address label of your print subscription. Then go to
http://www.trends.com, click on the Claim online access button and select
Trends in Cell Biology
You will see a BioMedNet login screen. Enter your BioMedNet username and password. If you are not already a BioMedNet
member, please click on the Register button. Once registered, you will be asked to enter your subscription key. Following
confirmation, you will have full access to Trends in Cell Biology
If you obtain an error message please contact Customer Services (info@current-trends.com) stating your subscription key and
BioMedNet username and password. Please note that you only need to enter your subscription key once; BioMedNet remembers
your subscription. Institutional online access is available at a premium. If your institute is interested in subscribing online, please
ask them to contact reviews.subscribe@biomednet.com

http://ticb.trends.com

Vous aimerez peut-être aussi